1
|
Wu X, Ferguson N, Tang LWT. Efficient protein quantification in drug metabolism and pharmacokinetics with an accelerated proteomic workflow. Drug Metab Dispos 2025; 53:100048. [PMID: 40068543 DOI: 10.1016/j.dmd.2025.100048] [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: 12/16/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 03/30/2025] Open
Abstract
Quantifying proteins involved in the absorption, distribution, metabolism, and excretion (ADME) of drugs is essential to improve understanding of their disposition and pharmacokinetics. Proteomics, because of its great versatility, is a widely used approach for protein analysis. However, existing protocols face challenges, such as poor peptide identification in liquid chromatography with tandem mass spectrometry under multiple reaction monitoring mode as well as the time- and labor-intensive nature of detergent-engaged workflows. In this study, we compared and evaluated several targeted ADME proteomic methods, including a novel approach called Fast Surfactant-Treated (FAST). Using FAST in ADME proteome analysis of primary human hepatocytes revealed that most proteins, especially membrane proteins, were efficiently solubilized and digested, with the ionic detergent sodium deoxycholate and rapidly removed during preparation by the incorporation of a centrifugation step following acetonitrile precipitation. Compared with the traditional proteomic workflow involving dithiothreitol reduction and iodoacetamide alkylation, FAST achieved an approximately 4-fold increase in cytochrome P450 and UDP-glucuronosyltransferases quantification and 5-fold increase in transporters, based on endogenous tryptic peptide signals. For specific proteins such as CYP2J2, organic anion transporter, and organic anion transporting polypeptide 1B1, FAST generated peptide quantification peaks with significantly higher signal-to-noise ratios and in a shorter amount of sample processing time. We then further validated the FAST proteomics workflow using the pregnane X receptor agonist rifampicin in human hepatocytes, which revealed that CYP3A4 protein levels were induced to a similar extent as observed in the CYP3A midazolam-1'-hydroxylase activity assay. Altogether, these results suggest that FAST proteomics is a robust, efficient, and versatile method for ADME bioanalysis. SIGNIFICANCE STATEMENT: Quantifying absorption, distribution, metabolism, and excretion (ADME) proteins from in vitro matrices remains a challenge, particularly when speed and efficiency are critical. By incorporating sodium deoxycholate detergent into the ADME proteome sample preparation workflow, we developed a methodology called Fast Surfactant-Treated proteomics. This approach enabled more efficient quantification of drug-metabolizing enzymes and membrane transporters, offering a streamlined protocol with reduced bench time.
Collapse
Affiliation(s)
- Xiaofeng Wu
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc, Groton, Connecticut.
| | - Nicholas Ferguson
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc, Groton, Connecticut
| | - Lloyd Wei Tat Tang
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc, Groton, Connecticut
| |
Collapse
|
2
|
Suo Y, Fedor JG, Zhang H, Tsolova K, Shi X, Sharma K, Kumari S, Borgnia M, Zhan P, Im W, Lee SY. Molecular basis of the urate transporter URAT1 inhibition by gout drugs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612563. [PMID: 39314352 PMCID: PMC11419087 DOI: 10.1101/2024.09.11.612563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Hyperuricemia is a condition when uric acid, a waste product of purine metabolism, accumulates in the blood1. Untreated hyperuricemia can lead to crystal formation of monosodium urate in the joints, causing a painful inflammatory disease known as gout. These conditions are associated with many other diseases and affect a significant and increasing proportion of the population2-4. The human urate transporter 1 (URAT1) is responsible for the reabsorption of ~90% of uric acid in the kidneys back into the blood, making it a primary target for treating hyperuricemia and gout5. Despite decades of research and development, clinically available URAT1 inhibitors have limitations because the molecular basis of URAT1 inhibition by gout drugs remains unknown5. Here we present cryo-electron microscopy structures of URAT1 alone and in complex with three clinically relevant inhibitors: benzbromarone, lesinurad, and the novel compound TD-3. Together with functional experiments and molecular dynamics simulations, we reveal that these inhibitors bind selectively to URAT1 in inward-open states. Furthermore, we discover differences in the inhibitor dependent URAT1 conformations as well as interaction networks, which contribute to drug specificity. Our findings illuminate a general theme for URAT1 inhibition, paving the way for the design of next-generation URAT1 inhibitors in the treatment of gout and hyperuricemia.
Collapse
Affiliation(s)
- Yang Suo
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Justin G. Fedor
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Han Zhang
- Departments of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, 18015, USA
| | - Kalina Tsolova
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Xiaoyu Shi
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012 Shandong, P.R. China
| | - Kedar Sharma
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA
| | - Shweta Kumari
- Departments of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, 18015, USA
| | - Mario Borgnia
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012 Shandong, P.R. China
| | - Wonpil Im
- Departments of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, 18015, USA
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| |
Collapse
|
3
|
Wei W, Tang LWT, Verma RK, Fan H, Chan ECY. Probe Substrate Dependencies in CYP3A4 Allosteric Inhibition: A Novel Molecular Mechanism Involving F-F' Loop Perturbations. J Chem Inf Model 2024; 64:2058-2067. [PMID: 38457234 DOI: 10.1021/acs.jcim.3c01837] [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: 03/09/2024]
Abstract
The biochemical basis for substrate dependences in apparent inhibition constant values (Ki) remains unknown. Our study aims to elucidate plausible structural determinants underpinning these observations. In vitro steady-state inhibition assays conducted using human recombinant CYP3A4 enzyme and testosterone substrate revealed that fibroblast growth factor receptor (FGFR) inhibitors erdafitinib and pemigatinib noncompetitively inhibited CYP3A4 with apparent Ki values of 10.2 ± 1.1 and 3.3 ± 0.9 μM, respectively. However, when rivaroxaban was adopted as the probe substrate, there were 2.0- and 3.2-fold decreases in its apparent Ki values. To glean mechanistic insights into this phenomenon, erdafitinib and pemigatinib were docked to allosteric sites in CYP3A4. Subsequently, molecular dynamics (MD) simulations of apo- and holo-CYP3A4 were conducted to investigate the structural changes induced. Comparative structural analyses of representative MD frames extracted by hierarchical clustering revealed that the allosteric inhibition of CYP3A4 by erdafitinib and pemigatinib did not substantially modulate its active site characteristics. In contrast, we discovered that allosteric binding of the FGFR inhibitors reduces the structural flexibility of the F-F' loop region, an important gating mechanism to regulate access of the substrate to the catalytic heme. We surmised that the increased rigidity of the F-F' loop engenders a more constrained entrance to the CYP3A4 active site, which in turn impedes access to the larger rivaroxaban molecule to a greater extent than testosterone and culminates in more potent inhibition of its CYP3A4-mediated metabolism. Our findings suggest a potential mechanism to rationalize probe substrate dependencies in Ki arising from the allosteric noncompetitive inhibition of CYP3A4.
Collapse
Affiliation(s)
- Wan Wei
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, 138671 Singapore
| | - Lloyd Wei Tat Tang
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, 117543 Singapore
| | - Ravi Kumar Verma
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, 138671 Singapore
| | - Hao Fan
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, 138671 Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, 117543 Singapore
| |
Collapse
|
4
|
Metabolic activation of drugs by cytochrome P450 enzymes: Biochemical insights into mechanism-based inactivation by fibroblast growth factor receptor inhibitors and chemical approaches to attenuate reactive metabolite formation. Biochem Pharmacol 2022; 206:115336. [DOI: 10.1016/j.bcp.2022.115336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
|
5
|
Atypical kinetics of cytochrome P450 enzymes in pharmacology and toxicology. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 95:131-176. [PMID: 35953154 DOI: 10.1016/bs.apha.2022.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Atypical kinetics are observed in metabolic reactions catalyzed by cytochrome P450 enzymes (P450). Yet, this phenomenon is regarded as experimental artifacts in some instances despite increasing evidence challenging the assumptions of typical Michaelis-Menten kinetics. As P450 play a major role in the metabolism of a wide range of substrates including drugs and endogenous compounds, it becomes critical to consider the impact of atypical kinetics on the accuracy of estimated kinetic and inhibitory parameters which could affect extrapolation of pharmacological and toxicological implications. The first half of this book chapter will focus on atypical non-Michaelis-Menten kinetics (e.g. substrate inhibition, biphasic and sigmoidal kinetics) as well as proposed underlying mechanisms supported by recent insights in mechanistic enzymology. In particular, substrate inhibition kinetics in P450 as well as concurrent drug inhibition of P450 in the presence of substrate inhibition will be further discussed. Moreover, mounting evidence has revealed that despite the high degree of sequence homology between CYP3A isoforms (i.e. CYP3A4 and CYP3A5), they have the propensities to exhibit vastly different susceptibilities and potencies of mechanism-based inactivation (MBI) with a common drug inhibitor. These experimental observations pertaining to the presence of these atypical isoform- and probe substrate-specific complexities in CYP3A isoforms by several clinically-relevant drugs will therefore be expounded and elaborated upon in the second half of this book chapter.
Collapse
|
6
|
Tang LWT, Wu G, Chan ECY. Identification of Infigratinib as a Potent Reversible Inhibitor and Mechanism-Based Inactivator of CYP2J2: Nascent Evidence for a Potential In Vivo Metabolic Drug-Drug Interaction with Rivaroxaban. J Pharmacol Exp Ther 2022; 382:123-134. [PMID: 35640957 PMCID: PMC9639665 DOI: 10.1124/jpet.122.001222] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/04/2022] [Indexed: 11/22/2022] Open
Abstract
Infigratinib (INF) is a fibroblast growth factor receptor inhibitor that was recently United States Food and Drug Administration-approved for the treatment of advanced or metastatic cholangiocarcinoma. We previously established that INF inhibited and inactivated cytochrome P450 3A4 (CYP3A4). Here, in a follow up to our previous study, we identified for the first time that INF also elicited potent competitive inhibition and mechanism-based inactivation of CYP2J2 with kinetic parameters K i, K I, k inact, and a partition ratio of 1.94 µM, 0.10 µM, 0.026 minute-1, and ∼3, respectively, when rivaroxaban was harnessed as the probe substrate. Inactivation was revealed to exhibit cofactor-dependency and was attenuated by an alternative substrate (astemizole) and direct inhibitor (nilotinib) of CYP2J2. Additionally, the nature of inactivation was unlikely to be pseudo-irreversible and instead arose from covalent modification due to the lack of substantial enzyme activity recovery after dialysis and chemical oxidation, as well as the lack of a resolvable Soret band in spectral scans. Glutathione trapping confirmed that the identity of the putative reactive intermediate implicated in the covalent inactivation of both CYP2J2 and CYP3A4 was identical and likely attributable to an electrophilic p-benzoquinonediimine intermediate of INF. Finally, mechanistic static modeling revealed that by integrating the previously arcane inhibition and inactivation kinetic parameters of CYP2J2-mediated rivaroxaban hydroxylation by INF illuminated in this work, together with those previously documented for CYP3A4, a 49% increase in the systemic exposure of rivaroxaban was projected. Our modeling results predicted a potential risk of metabolic drug-drug interactions between the clinically relevant combination of rivaroxaban and INF in the setting of cancer. SIGNIFICANCE STATEMENT: This study reported that INF elicits potent reversible inhibition and mechanism-based inactivation of CYP2J2. Furthermore, static modelling predicted that its coadministration with the direct oral anticoagulant rivaroxaban may potentially culminate in a metabolic drug-drug interaction (DDI) leading to an increased risk of major bleeding. As rivaroxaban is steadily gaining prominence as the anticoagulant of choice in the treatment of cancer-associated venous thromboembolism, the DDI projections reported here are clinically relevant and warrant further investigation via physiologically based pharmacokinetic modelling and simulation.
Collapse
Affiliation(s)
- Lloyd Wei Tat Tang
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Guoyi Wu
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| |
Collapse
|
7
|
Tang LWT, Wei W, Verma RK, Koh SK, Zhou L, Fan H, Chan ECY. Direct and Sequential Bioactivation of Pemigatinib to Reactive Iminium Ion Intermediates Culminates in Mechanism-Based Inactivation of Cytochrome P450 3A. Drug Metab Dispos 2022; 50:529-540. [PMID: 35153194 DOI: 10.1124/dmd.121.000804] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/01/2022] [Indexed: 02/13/2025] Open
Abstract
We recently established the mechanism-based inactivation (MBI) of cytochrome P450 3A (CYP3A) by the fibroblast growth factor receptor (FGFR) inhibitors erdafitinib and infigratinib. Serendipitously, our preliminary data have also revealed that pemigatinib (PEM), another clinically approved FGFR1-3 inhibitor, similarly elicited time-dependent inhibition of CYP3A. This was rather unexpected, as it was previously purported that PEM did not pose any metabolism-dependent liabilities due to the absence of glutathione-related conjugates in metabolic profiling experiments conducted in human liver microsomes. Here, we confirmed that PEM inhibited both CYP3A isoforms in a time-, concentration-, and cofactor-dependent manner consistent with MBI, with inactivator concentration at half-maximum rate constant, maximum inactivation rate constant, and partition ratio of 8.69 and 11.95 μM, 0.108 and 0.042 min-1, and approximately 44 and approximately 47 for CYP3A4 and CYP3A5, respectively. Although the rate of inactivation was diminished by coincubation with an alternative substrate or direct inhibitor of CYP3A, the inclusion of nucleophilic trapping agents afforded no such protection. Furthermore, the lack of catalytic activity recovery following dialysis and oxidation with potassium ferricyanide coupled with the absence of a spectrally resolvable peak in the Soret region collectively implied that the underlying mechanism of inactivation was not elicited via the formation of pseudo-irreversible metabolite-intermediate complexes. Finally, utilizing cyanide trapping and high-resolution mass spectrometry, we illuminated the direct and sequential oxidative bioactivation of PEM and its major O-desmethylated metabolite at its distal morpholine moiety to reactive iminium ion hard electrophilic species that could covalently inactivate CYP3A via MBI. SIGNIFICANCE STATEMENT: This study reports for the first time the covalent MBI of CYP3A by PEM and deciphered its bioactivation pathway involving the metabolic activation of PEM and its major O-desmethylated metabolite to reactive iminium ion intermediates. Following which, a unique covalent docking methodology was harnessed to unravel the structural and molecular determinants underpinning its inactivation. Findings from this study lay the foundation for future investigation of clinically relevant drug-drug interactions between PEM and concomitant substrates of CYP3A.
Collapse
Affiliation(s)
- Lloyd Wei Tat Tang
- Department of Pharmacy, Faculty of Science, (L.W.T.T., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (W.W., R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| | - Wan Wei
- Department of Pharmacy, Faculty of Science, (L.W.T.T., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (W.W., R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| | - Ravi Kumar Verma
- Department of Pharmacy, Faculty of Science, (L.W.T.T., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (W.W., R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| | - Siew Kwan Koh
- Department of Pharmacy, Faculty of Science, (L.W.T.T., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (W.W., R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| | - Lei Zhou
- Department of Pharmacy, Faculty of Science, (L.W.T.T., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (W.W., R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| | - Hao Fan
- Department of Pharmacy, Faculty of Science, (L.W.T.T., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (W.W., R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, (L.W.T.T., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (W.W., R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| |
Collapse
|
8
|
Tang LWT, Lim RYR, Venkatesan G, Chan ECY. Rational deuteration of dronedarone attenuates its toxicity in human hepatic HepG2 cells. Toxicol Res (Camb) 2022; 11:311-324. [PMID: 35510231 PMCID: PMC9052316 DOI: 10.1093/toxres/tfac017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/19/2022] [Accepted: 03/11/2022] [Indexed: 11/14/2022] Open
Abstract
Deuteration is a chemical modification strategy that has recently gained traction in drug development. The replacement of one or more hydrogen atom(s) in a drug molecule with its heavier stable isotope deuterium can enhance its metabolic stability and pharmacokinetic properties. However, it remains uninterrogated if rational deuteration at bioactivation "hot-spots" could attenuate its associated toxicological consequences. Here, our preliminary screening with benzofuran antiarrhythmic agents first revealed that dronedarone and its major metabolite N-desbutyldronedarone elicited a greater loss of viability and cytotoxicity in human hepatoma G2 (HepG2) cells as compared with amiodarone and its corresponding metabolite N-desethylamiodarone. A comparison of dronedarone and its in-house synthesized deuterated analogue (termed poyendarone) demonstrated that deuteration could attenuate its in vitro toxicity in HepG2 cells by modulating the extent of mitochondrial dysfunction, reducing the dissipation of mitochondrial membrane potential, and evoking a distinct apoptotic kinetic signature. Furthermore, although pretreatment with the CYP3A inducer rifampicin or the substitution of glucose with galactose in the growth media significantly augmented the loss of cell viability elicited by dronedarone and poyendarone, a lower loss of cell viability was consistently observed in poyendarone across all concentrations. Taken together, our preliminary investigations suggested that the rational deuteration of dronedarone at its benzofuran ring reduces aberrant cytochrome P450 3A4/5-mediated bioactivation, which attenuated its mitochondrial toxicity in human hepatic HepG2 cells.
Collapse
Affiliation(s)
- Lloyd Wei Tat Tang
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, 117543, Singapore
| | - Royden Yu Ren Lim
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, 117543, Singapore
| | - Gopalakrishnan Venkatesan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, 117543, Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, 117543, Singapore
| |
Collapse
|
9
|
Tang LWT, Teng JW, Verma RK, Koh SK, Zhou L, Go ML, Fan H, Chan ECY. Infigratinib Is a Reversible Inhibitor and Mechanism-Based Inactivator of Cytochrome P450 3A4. Drug Metab Dispos 2021; 49:856-868. [PMID: 34326139 DOI: 10.1124/dmd.121.000508] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/08/2021] [Indexed: 11/22/2022] Open
Abstract
Infigratinib (INF) is a promising selective inhibitor of fibroblast growth factor receptors 1-3 that has recently been accorded both orphan drug designation and priority review status by the US Food and Drug Administration for the treatment of advanced cholangiocarcinoma. Its propensity to undergo bioactivation to electrophilic species was recently expounded upon. However, other than causing aberrant idiosyncratic toxicities, these reactive intermediates may elicit mechanism-based inactivation of cytochrome P450 enzymes. In this study, we investigated the interactions between INF and the most abundant hepatic CYP3A. Our findings revealed that, apart from being a potent noncompetitive reversible inhibitor of CYP3A4, INF inactivated CYP3A4 in a time-, concentration- and NADPH-dependent manner with inactivator concentration at half-maximum inactivation rate constant, maximum inactivation rate constant, and partition ratio of 4.17 µM, 0.068 minute-1, and 41, respectively, when rivaroxaban was employed as the probe substrate. Coincubation with testosterone (alternative CYP3A substrate) or ketoconazole (direct CYP3A inhibitor) attenuated the rate of inactivation, whereas the inclusion of glutathione and catalase did not confer such protection. The lack of enzyme activity recovery after dialysis for 4 hours and oxidation with potassium ferricyanide, coupled with the absence of the characteristic Soret peak signature collectively substantiated that inactivation of CYP3A4 by INF was not mediated by the formation of quasi-irreversible metabolite-intermediate complexes but rather through irreversible covalent adduction to the prosthetic heme and/or apoprotein. Finally, glutathione trapping and high-resolution mass spectrometry experimental results unraveled two plausible bioactivation mechanisms of INF arising from the generation of a p-benzoquinonediimine and epoxide reactive intermediate. SIGNIFICANCE STATEMENT: The potential of INF to cause MBI of CYP3A4 was unknown. This study reports the reversible noncompetitive inhibition and irreversible covalent MBI of CYP3A4 by INF and proposes two potential bioactivation pathways implicating p-benzoquinonediimine and epoxide reactive intermediates, following which a unique covalent docking methodology was harnessed to elucidate the structural and molecular determinants underscoring its inactivation. Findings from this study lay the groundwork for future investigation of clinically relevant drug-drug interactions between INF and concomitant substrates of CYP3A4.
Collapse
Affiliation(s)
- Lloyd Wei Tat Tang
- Department of Pharmacy, Faculty of Science (L.W.T.T., J.W.T, M.L.G., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| | - Jian Wei Teng
- Department of Pharmacy, Faculty of Science (L.W.T.T., J.W.T, M.L.G., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| | - Ravi Kumar Verma
- Department of Pharmacy, Faculty of Science (L.W.T.T., J.W.T, M.L.G., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| | - Siew Kwan Koh
- Department of Pharmacy, Faculty of Science (L.W.T.T., J.W.T, M.L.G., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| | - Lei Zhou
- Department of Pharmacy, Faculty of Science (L.W.T.T., J.W.T, M.L.G., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| | - Mei Lin Go
- Department of Pharmacy, Faculty of Science (L.W.T.T., J.W.T, M.L.G., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| | - Hao Fan
- Department of Pharmacy, Faculty of Science (L.W.T.T., J.W.T, M.L.G., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science (L.W.T.T., J.W.T, M.L.G., E.C.Y.C.) and Department of Ophthalmology, Yong Loo Lin School of Medicine (L.Z.), National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology and Research, Singapore (R.K.V., H.F.); Singapore Eye Research Institute, Singapore (S.K.K., L.Z.); and Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, Singapore (L.Z.)
| |
Collapse
|