1
|
Tanaudommongkon I, Rashidian A, Gufford BT, Lu JBL, Desta Z. The strong clinical interaction between bupropion and CYP2D6 is primarily mediated through bupropion metabolites and their stereoisomers: A paradigm for evaluating metabolites in drug-drug interaction risk. Drug Metab Dispos 2025; 53:100070. [PMID: 40245579 DOI: 10.1016/j.dmd.2025.100070] [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: 10/21/2024] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Bupropion (BUP) is a potent clinical inhibitor of CYP2D6, although the specific mechanisms underlying this interaction are not fully understood. We comprehensively evaluated the inhibition potencies of racemic BUP and its stereoisomers, as well as its major metabolites (4-hydroxybupropion [OHBUP], threohydrobupropion [THBUP], and erythrohydrobupropion [EHBUP]), on CYP2D6-mediated dextromethorphan O-demethylation in pooled human liver microsomes. The Ki value for racemic EHBUP was 5.5-, 11.4-, and 13-fold lower than those for THBUP, OHBUP, and BUP, respectively. THBUP demonstrated over 2-fold greater potency than OHBUP and BUP. Additionally, RR-THBUP had a 2.1-fold lower Ki value than SS-THBUP, while S-BUP and RR-OHBUP exhibited 3.0-fold and 1.5-fold lower Ki values than R-BUP and SS-OHBUP, respectively, indicating modest stereoselective inhibition. The Ki values of stereoisomers EHBUP were comparable. Using a mechanistic static interaction model that incorporated in vitro Ki value and unbound steady-state concentration in plasma (Cmax) or estimated liver concentrations of each inhibitor, we found significant underprediction of the observed clinical BUP-CYP2D6 interaction, indicating that no single inhibitor can predict observed in vivo BUP-CYP2D6 interaction. Accurate predictions of observed clinical interactions were achieved using all racemic (within 11.6%) or all stereoisomeric forms (within 5.4%) of BUP and its metabolites, along with their liver concentrations (but not plasma concentrations). Our findings highlight the crucial role of circulating BUP metabolites, particularly the summation of EHBUP and THBUP or their stereoisomers, in the in vivo inhibition of CYP2D6 by BUP. These data provide mechanistic and quantitative insight into the partially understood clinical CYP2D6-dependent interactions associated with BUP. SIGNIFICANCE STATEMENT: This article describes comprehensively the inhibition of CYP2D6 by racemic and stereoisomers of bupropion (BUP) and its main metabolites in pooled human liver microsomes. The strong interaction between BUP and CYP2D6 observed clinically is mainly due to BUP metabolites and inhibition is stereospecific. Accounting for inhibition constants and steady-state unbound estimated liver (but not plasma) concentrations of all racemic or all stereoisomers accurately predicted the clinically observed BUP-CYP2D6 interactions.
Collapse
Affiliation(s)
- Irin Tanaudommongkon
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Amir Rashidian
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Brandon T Gufford
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jessica Bo Li Lu
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Zeruesenay Desta
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
2
|
Nahid NA, Kanumuri SRR, Sharma A, Wang D, Johnson JA. In vitro comparative analysis of metabolic capabilities and inhibitory profiles of selected CYP2D6 alleles on tramadol metabolism. Clin Transl Sci 2025; 18:e70059. [PMID: 39870079 PMCID: PMC11772017 DOI: 10.1111/cts.70059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/18/2024] [Accepted: 10/10/2024] [Indexed: 01/30/2025] Open
Abstract
Tramadol, the 41st most prescribed drug in the United States in 2021 is a prodrug activated by CYP2D6, which is highly polymorphic. Previous studies showed enzyme-inhibitor affinity varied between different CYP2D6 allelic variants with dextromethorphan and atomoxetine metabolism. However, no study has compared tramadol metabolism in different CYP2D6 alleles with different CYP2D6 inhibitors. We hypothesize that the inhibitory effects of CYP2D6 inhibitors on CYP2D6-mediated tramadol metabolism are inhibitor- and CYP2D6-allele-specific. We performed comparative analyses of CYP2D6*1, CYP2D6*2, CYP2D6*10, and CYP2D6*17 using recombinant enzymes to metabolize tramadol to O-desmethyltramadol, measured via UPLC-MS/MS. The Michaelis constant (Km) and maximum velocity (Vmax) for each CYP2D6 allele, and IC50 values for different inhibitors were determined by nonlinear regression analysis. Intrinsic clearance was calculated as Vmax/Km. The intrinsic clearance of tramadol was almost double for CYP2D6*2 (180%) but was much lower for CYP2D6*10 and *17 (20% and 10%, respectively) compared to CYP2D6*1. The inhibitor potencies (defined by Ki) for the various inhibitors for the CYP2D6*1 allele were quinidine > terbinafine > paroxetine ≈ duloxetine >>bupropion. CYP2D6*2 showed the next greatest inhibition, with Ki ratios compared to CYP2D6*1 ranging from 0.96 to 3.87. For each inhibitor tested, CYP2D6*10 and CYP2D6*17 were more resistant to inhibition than CYP2D6*1 or CYP2D6*2, with most Ki ratios in the 3-9 range. Three common CYP2D6 allelic variants showed different metabolic capacities toward tramadol and genotype-dependent inhibition compared to CYP2D6*1. Further studies are warranted to understand the clinical consequences of inhibitor and CYP2D6 genotype-dependent drug-drug interactions on tramadol bioactivation.
Collapse
Affiliation(s)
- Noor Ahmed Nahid
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision MedicineUniversity of Florida College of PharmacyGainesvilleFloridaUSA
| | - Siva Rama Raju Kanumuri
- Department of PharmaceuticsUniversity of Florida College of PharmacyGainesvilleFloridaUSA
- Translational Drug Development CoreUniversity of Florida Clinical and Translational Science InstituteGainesvilleFloridaUSA
| | - Abhisheak Sharma
- Department of PharmaceuticsUniversity of Florida College of PharmacyGainesvilleFloridaUSA
- Translational Drug Development CoreUniversity of Florida Clinical and Translational Science InstituteGainesvilleFloridaUSA
| | - Danxin Wang
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision MedicineUniversity of Florida College of PharmacyGainesvilleFloridaUSA
| | - Julie A. Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision MedicineUniversity of Florida College of PharmacyGainesvilleFloridaUSA
- Clinical and Translational Science InstituteThe Ohio State UniversityColumbusOhioUSA
- Department of Internal Medicine and Department of Pharmaceutics & PharmacologyThe Ohio State University Colleges of Medicine and PharmacyColumbusOhioUSA
| |
Collapse
|
3
|
Raichura Z, Heck K, Choi J, Yang L, Brandes M, Marney L, Mangaña AA, Neff C, Maier CS, Soumyanath A, van Breemen RB, Arnold RD, Calderón AI. Evaluation of reversible cytochrome P450 inhibition by Withania somnifera leaf and root extracts. Drug Metab Dispos 2025; 53:100024. [PMID: 40023576 DOI: 10.1016/j.dmd.2024.100024] [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: 05/15/2024] [Accepted: 11/19/2024] [Indexed: 03/04/2025] Open
Abstract
It is important to understand the potential of botanical-drug interactions to ensure the safe use of botanical dietary supplements (BDS). Cytochrome P450 (P450) is one of the most abundant phase 1 drug-metabolizing enzymes and is accountable for a great deal of pharmacokinetic botanical-drug interactions. This problem is particularly acute for older adults who often consume BDS with multiple prescription medicines. The consequences of botanical-drug interactions can lead to lack of prodrug efficacy or drug toxicity from reduced drug clearance through inhibition of P450 metabolizing enzymes. In this study, a 7-in-1 cocktail P450 inhibition assay with 7 Food and Drug Administration-recommended P450s (CYP1A2, CYP2A6, CYP2C8, CYP2C9, CYP2D6, and CYP3A4/5) including CYP2B6 recombinant enzyme was performed, minimizing substrate interactions with respect to specificity while maximizing assay sensitivity. High-performance liquid chromatography-mass spectrometry was used for quantitative determination of probe substrate metabolism. Withania somnifera L. Dunal (ashwagandha), a popular BDS in the United States with sales of ∼$16 million in 2021, is used to promote sleep and relieve stress and anxiety, especially in older adults. However, comprehensive studies of pharmacokinetic drug interactions with ashwagandha, especially with leaf extracts, have not been reported. Four extracts from ashwagandha root or leaf were evaluated for P450 inhibition, and no reversible inhibition was detected at IC50 > 100 μg/mL extract. SIGNIFICANCE STATEMENT: Ashwagandha is often consumed by older adults, who also often use multiple prescribed medications concomitantly. Polypharmacy, combined with age-related decline of drug metabolism and other changes in drug disposition in this population, increases the risk of adverse events due to botanical inhibition of drug metabolism, indicating the significance of evaluating ashwagandha for potential pharmacokinetic drug interactions. This study will support our understanding for the safe use of ashwagandha.
Collapse
Affiliation(s)
- Zarna Raichura
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Kabre Heck
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Jaewoo Choi
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon; Botanicals Enhancing Neurological and Functional Resilience in Aging (BENFRA), Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, Oregon
| | - Liping Yang
- Department of Chemistry, Oregon State University, Corvallis, Oregon; Botanicals Enhancing Neurological and Functional Resilience in Aging (BENFRA), Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, Oregon
| | - Mikah Brandes
- Botanicals Enhancing Neurological and Functional Resilience in Aging (BENFRA), Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, Oregon; Department of Neurology, Oregon Health & Science University, Portland, Oregon
| | - Luke Marney
- Department of Chemistry, Oregon State University, Corvallis, Oregon; Botanicals Enhancing Neurological and Functional Resilience in Aging (BENFRA), Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, Oregon
| | - Armando Alcázar Mangaña
- Department of Chemistry, Oregon State University, Corvallis, Oregon; Botanicals Enhancing Neurological and Functional Resilience in Aging (BENFRA), Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, Oregon
| | - Cody Neff
- Botanicals Enhancing Neurological and Functional Resilience in Aging (BENFRA), Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, Oregon; Department of Neurology, Oregon Health & Science University, Portland, Oregon
| | - Claudia S Maier
- Department of Chemistry, Oregon State University, Corvallis, Oregon; Linus Pauling Institute, Oregon State University, Corvallis, Oregon; Botanicals Enhancing Neurological and Functional Resilience in Aging (BENFRA), Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, Oregon
| | - Amala Soumyanath
- Botanicals Enhancing Neurological and Functional Resilience in Aging (BENFRA), Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, Oregon; Department of Neurology, Oregon Health & Science University, Portland, Oregon
| | | | - Robert D Arnold
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Angela I Calderón
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama.
| |
Collapse
|
4
|
Vashisth C, Raghav N. Preferential inhibition of α-amylase by cinnamaldehyde-based hydrazones: A comparative study. Int J Biol Macromol 2024; 281:136654. [PMID: 39423966 DOI: 10.1016/j.ijbiomac.2024.136654] [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: 06/02/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
The escalating issue of obesity and related health conditions, including diabetes mellitus and coronary heart disease, necessitates the development of digestive enzyme inhibitors. Targeting pancreatic lipase to inhibit fatty acid generation to attain reduced lipid absorption is a promising approach for identifying effective agents. Orlistat, the only clinically approved pancreatic lipase inhibitor, is associated with gastrointestinal side effects like oily stools, spotting, and flatulence. Concurrently, α-amylase inhibitors can mitigate rapid spikes in blood sugar levels. With the growing significance of natural products in pharmaceutical research, cinnamaldehyde has been recognized as a potential enzyme inhibitor. This study explores the effects of cinnamaldehyde-derived hydrazone Schiff bases on lipase and α-amylase through in vitro experiments. The synthesized compounds exhibited IC50 value for lipase in the same range ((0.41-12.58) × 10-8 M) as exhibited by the commercial drug orlistat (5.76 × 10-8 M). However, for α-amylase inhibition, the compounds (IC50 value = 0.37-9.54 × 10-9 M) were found effective inhibitors of acarbose (IC50 value = 6.76 × 10-6 M) and curcumin (IC50 value = 467 × 10-9 M). In silico DFT studies, molecular docking, drug likeliness, bioactivity score, ADME, and toxicity are also investigated for these compounds.
Collapse
Affiliation(s)
- Chanchal Vashisth
- Department of Chemistry, Kurukshetra University, Kurukshetra, Haryana 136119, India
| | - Neera Raghav
- Department of Chemistry, Kurukshetra University, Kurukshetra, Haryana 136119, India.
| |
Collapse
|
5
|
Wang Z, Luk KHY, Cheong EJY, Tham SM, Periaswami R, Toh PC, Wang Z, Wu QH, Tsang WC, Kesavan A, Wong ASC, Wong PT, Lim F, Chiong E, Chan ECY. Characterization and Prediction of Organic Anion Transporting Polypeptide 1B Activity in Prostate Cancer Patients on Abiraterone Acetate Using Endogenous Biomarker Coproporphyrin I. Drug Metab Dispos 2024; 52:1356-1362. [PMID: 39187385 DOI: 10.1124/dmd.124.001878] [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: 07/12/2024] [Revised: 08/10/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024] Open
Abstract
Organic anion transporting polypeptide (OATP) 1B1 and OATP1B3 are important hepatic transporters. We previously identified OATP1B3 being critically implicated in the disposition of abiraterone. We aimed to further investigate the effects of abiraterone on the activities of OATP1B1 and OATP1B3 utilizing a validated endogenous biomarker coproporphyrin I (CP-I). We used OATP1B-transfected cells to characterize the inhibitory potential of abiraterone against OATP1B-mediated uptake of CP-I. Inhibition constant (K i) was incorporated into our physiologically based pharmacokinetic (PBPK) modeling to simulate the systemic exposures of CP-I among cancer populations receiving either our model-informed 500 mg or clinically approved 1000 mg abiraterone acetate (AA) dosage. Simulated data were compared with clinical CP-I concentrations determined among our nine metastatic prostate cancer patients receiving 500 mg AA treatment. Abiraterone inhibited OATP1B3-mediated, but not OATP1B1-mediated, uptake of CP-I in vitro, with an estimated K i of 3.93 μM. Baseline CP-I concentrations were simulated to be 0.81 ± 0.26 ng/ml and determined to be 0.72 ± 0.16 ng/ml among metastatic prostate cancer patients, both of which were higher than those observed for healthy subjects. PBPK simulations revealed an absence of OATP1B3-mediated interaction between abiraterone and CP-I. Our clinical observations confirmed that CP-I concentrations remained comparable to baseline levels up to 12 weeks post 500 mg AA treatment. Using CP-I as an endogenous biomarker, we identified the inhibition of abiraterone on OATP1B3 but not OATP1B1 in vitro, which was predicted and observed to be clinically insignificant. We concluded that the interaction risk between AA and substrates of OATP1Bs is low. SIGNIFICANCE STATEMENT: The authors used the endogenous biomarker coproporphyrin I (CP-I) and identified abiraterone as a moderate inhibitor of organic anion transporting polypeptide (OATP) 1B3 in vitro. Subsequent physiologically based pharmacokinetic (PBPK) simulations and clinical observations suggested an absence of OATP1B-mediated interaction between abiraterone and CP-I among prostate cancer patients. This multipronged study concluded that the interaction risk between abiraterone acetate and substrates of OATP1Bs is low, demonstrating the application of PBPK-CP-I modeling in predicting OATP1B-mediated interaction implicating abiraterone.
Collapse
Affiliation(s)
- Ziteng Wang
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| | - Kylie Hoi Yan Luk
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| | - Eleanor Jing Yi Cheong
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| | - Sin Mun Tham
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| | - Revathi Periaswami
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| | - Poh Choo Toh
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| | - Ziting Wang
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| | - Qing Hui Wu
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| | - Woon Chau Tsang
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| | - Arshvin Kesavan
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| | - Alvin Seng Cheong Wong
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| | - Patrick Thomas Wong
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| | - Felicia Lim
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| | - Edmund Chiong
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| | - Eric Chun Yong Chan
- Department of Pharmacy and Pharmaceutical Sciences (Zite.W., K.H.Y.L., E.J.Y.C., E.C.Y.C.) and Department of Surgery, Yong Loo Lin School of Medicine (S.M.T., R.P., E.C.), National University of Singapore, Singapore; Department of Urology (P.C.T., Ziti.W., Q.H.W., W.C.T., A.K., E.C.) and Department of Pharmacy (P.T.W., F.L.), National University Hospital, National University Health System, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore (A.S.C.W.); and National University Cancer Institute, Singapore (P.T.W.)
| |
Collapse
|
6
|
Wang E, Wang M, Gao M. Probe substrates assay estimates the effect of polyphyllin H on the activity of cytochrome P450 enzymes in human liver microsomes. Pharmacol Res Perspect 2024; 12:e70002. [PMID: 39210686 PMCID: PMC11362609 DOI: 10.1002/prp2.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/08/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024] Open
Abstract
Cytochrome P450 enzymes (CYPs) play a crucial role in phase I metabolic reactions. The activity of CYPs would affect therapeutic efficacy and may even induce toxicity. Given the complex components of traditional Chinese medicine, it is important to understand the effect of active ingredients on CYPs activity to guide their prescription. This study aimed to evaluate the effect of polyphyllin H on the activity of CYPs major isoforms providing a reference for the clinical prescription of polyphyllin H and its source herbs. The effects of polyphyllin H were evaluated in pooled human liver microsomes using probe substrates of CYP1A2, 2A6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A4 to determine their activities. The Lineweaver-Burk was used to model the inhibition, and a time-dependent inhibition experiment was performed to understand the characteristics of the inhibition. Polyphyllin H significantly suppressed the activity of CYP1A2, 2D6, and 3A4 with IC50 values of 6.44, 13.88, and 4.52 μM, respectively. The inhibition of CYP1A2 and 2D6 was best fitted with a competitive model, yielding the inhibition constant (Ki) values of 3.18 and 6.77 μM, respectively. The inhibition of CYP3A4 was fitted with the non-competitive model with the Ki value of 2.38 μM. Moreover, the inhibition of CYP3A4 was revealed to be time-dependent with the inhibition parameters inhibition constant (KI) and inactivation rate constant (Kinact) values of 2.26 μM-1 and 0.045 min-1. Polyphyllin H acted as a competitive inhibitor of CYP1A2 and 2D6 and a non-competitive and time-dependent inhibitor of CYP3A4.
Collapse
Affiliation(s)
- Erhao Wang
- Pharmacy Department, Hainan Women and Children's Medical Center, Haikou, Hainan, China
| | - Mengxi Wang
- Pharmacy Department, Seafarers General Hospital of Heilongjiang Province/Heilongjiang Sixth Hospital, Harbin, Heilongjiang, China
| | - Ming Gao
- Pharmacy Department, The Affiliated Hospital of Chengdu University of Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Yadav J, Maldonato BJ, Roesner JM, Vergara AG, Paragas EM, Aliwarga T, Humphreys S. Enzyme-mediated drug-drug interactions: a review of in vivo and in vitro methodologies, regulatory guidance, and translation to the clinic. Drug Metab Rev 2024:1-33. [PMID: 39057923 DOI: 10.1080/03602532.2024.2381021] [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: 02/23/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
Enzyme-mediated pharmacokinetic drug-drug interactions can be caused by altered activity of drug metabolizing enzymes in the presence of a perpetrator drug, mostly via inhibition or induction. We identified a gap in the literature for a state-of-the art detailed overview assessing this type of DDI risk in the context of drug development. This manuscript discusses in vitro and in vivo methodologies employed during the drug discovery and development process to predict clinical enzyme-mediated DDIs, including the determination of clearance pathways, metabolic enzyme contribution, and the mechanisms and kinetics of enzyme inhibition and induction. We discuss regulatory guidance and highlight the utility of in silico physiologically-based pharmacokinetic modeling, an approach that continues to gain application and traction in support of regulatory filings. Looking to the future, we consider DDI risk assessment for targeted protein degraders, an emerging small molecule modality, which does not have recommended guidelines for DDI evaluation. Our goal in writing this report was to provide early-career researchers with a comprehensive view of the enzyme-mediated pharmacokinetic DDI landscape to aid their drug development efforts.
Collapse
Affiliation(s)
- Jaydeep Yadav
- Department of Pharmacokinetics, Dynamics, Metabolism & Bioanalytics (PDMB), Merck & Co., Inc., Boston, MA, USA
| | - Benjamin J Maldonato
- Department of Nonclinical Development and Clinical Pharmacology, Revolution Medicines, Inc., Redwood City, CA, USA
| | - Joseph M Roesner
- Department of Pharmacokinetics, Dynamics, Metabolism & Bioanalytics (PDMB), Merck & Co., Inc., Boston, MA, USA
| | - Ana G Vergara
- Department of Pharmacokinetics, Dynamics, Metabolism & Bioanalytics (PDMB), Merck & Co., Inc., Rahway, NJ, USA
| | - Erickson M Paragas
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, South San Francisco, CA, USA
| | - Theresa Aliwarga
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, South San Francisco, CA, USA
| | - Sara Humphreys
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, South San Francisco, CA, USA
| |
Collapse
|
8
|
Asano S, Kurosaki C, Mori Y, Shigemi R. Quantitative prediction of transporter-mediated drug-drug interactions using the mechanistic static pharmacokinetic (MSPK) model. Drug Metab Pharmacokinet 2024; 54:100531. [PMID: 38064927 DOI: 10.1016/j.dmpk.2023.100531] [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: 05/29/2023] [Revised: 08/21/2023] [Accepted: 10/02/2023] [Indexed: 02/06/2024]
Abstract
Guidance/guidelines on drug-drug interactions (DDIs) have been issued in Japan, the United States, and Europe. These guidance/guidelines provide decision trees for conducting metabolizing enzyme-mediated clinical DDI studies; however, the decision trees for transporter-mediated DDIs lack quantitative prediction methods. In this study, the accuracy of a net-effect mechanistic static pharmacokinetics (MSPK) model containing the fraction transported (ft) of transporters was examined to predict transporter-mediated DDIs. This study collected information on 25 oral drugs with new active reagents that were used in clinical DDI studies as perpetrators (42 cases) from drugs approved in Japan between April 2016 and June 2020. The AUCRs (AUC ratios with and without perpetrators) of victim drugs were predicted using the net-effect MSPK model. As a result, 83 and 95% of the predicted AUCRs were within 1.5- and 2-fold error in the observed AUCRs, respectively. In cases where the victims were statins in which pharmacokinetics several transporters are involved, 70 and 91% of the predicted AUCRs were within 1.5- and 2-fold errors, respectively. Therefore, the net-effect MSPK model was applicable for predicting the AUCRs of victims, which are substrates for multiple transporters.
Collapse
Affiliation(s)
- Satoshi Asano
- Japan Pharmaceutical Manufacturers Association, Nihonbashi Life Science Bldg, 2-3-11 Nihonbashi-honcho, Chuo-Ku, Tokyo, Japan; Teijin Pharma Limited, Toxicology & DMPK Development Research Group, 4-3-2, Asahigaoka, Hino, Tokyo, 191-8512, Japan.
| | - Chie Kurosaki
- Japan Pharmaceutical Manufacturers Association, Nihonbashi Life Science Bldg, 2-3-11 Nihonbashi-honcho, Chuo-Ku, Tokyo, Japan; FUJIFILM Toyama Chemical Co., Ltd, ADME-Tox Group, Bioanalytical Sciences Research Department, Toyama Research and Development Center, 4-1, Shimo-Okui 2-chome, Toyama-shi, Toyama, Japan
| | - Yuko Mori
- Japan Pharmaceutical Manufacturers Association, Nihonbashi Life Science Bldg, 2-3-11 Nihonbashi-honcho, Chuo-Ku, Tokyo, Japan; Pfizer R&D Japan, Clinical Pharmacology and Bioanalytics, Shinjuku Bunka Quint Bldg., 3-22-7, Yoyogi, Shibuya-ku, Tokyo, Japan
| | - Ryota Shigemi
- Japan Pharmaceutical Manufacturers Association, Nihonbashi Life Science Bldg, 2-3-11 Nihonbashi-honcho, Chuo-Ku, Tokyo, Japan; Bayer Yakuhin, Ltd, Preclinical Development, Breeze Tower, 2-4-9, Umeda, Kita-ku, Osaka, Japan
| |
Collapse
|
9
|
Wang R, Zheng K, Liu Y, Ji S, Tang Y, Wang J, Jiang R. Effect of tubeimoside I on the activity of cytochrome P450 enzymes in human liver microsomes. Xenobiotica 2024; 54:57-63. [PMID: 38166553 DOI: 10.1080/00498254.2023.2301352] [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: 11/13/2023] [Accepted: 12/29/2023] [Indexed: 01/04/2024]
Abstract
This study assessed the effect of tubeimoside I on CYP1A2, 2A6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A4 to reveal the potential of tubeimoside I to induce drug-drug interaction.The evaluation of cytochromes P450 enzyme (CYP) activity was performed in pooled human liver microsomes with probing substrates of CYP1A2, 2A6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A4. Typical inhibitors were employed as positive controls and the effect of 0, 2.5, 5, 10, 25, 50, and 100 μM tubeimoside I was investigated.The activity of CYP2D6, 2E1, and 3A4 was significantly inhibited by tubeimoside I with the IC50 values of 10.34, 11.58, and 9.74 μM, respectively. The inhibition of CYP2D6 and 2E1 was competitive with the Ki value of 5.66 and 5.29 μM, respectively. While the inhibition of CYP3A4 was non-competitive with the Ki value of 4.87 μM. Moreover, the inhibition of CYP3A4 was time-dependent with the KI and Kinact values of 0.635 μM-1 and 0.0373 min-1, respectively.Tubeimoside I served as a competitive inhibitor of CYP2D6 and 2E1 exerting weak inhibition and a non-competitive inhibitor of CYP3A4 exerting moderate inhibition.
Collapse
Affiliation(s)
- Rui Wang
- Department of Pharmacy, Shanghai Zhongye Hospital, Shanghai, China
| | - Kai Zheng
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Yunjiao Liu
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Shuxia Ji
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Yaxin Tang
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Jie Wang
- Department of Bone and Joint Rehabilitation, The Second Rehabilitation Hospital of Shanghai, Shanghai, China
| | - Rong Jiang
- Department of Bone and Joint Rehabilitation, The Second Rehabilitation Hospital of Shanghai, Shanghai, China
| |
Collapse
|
10
|
Purnawita W, Rahayu WP, Lioe HN, Nurjanah S, Wahyudi ST. Potential molecular mechanism of reuterin on the inhibition of Aspergillus flavus conidial germination: An in silico study. J Food Sci 2024; 89:1167-1186. [PMID: 38193164 DOI: 10.1111/1750-3841.16904] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/28/2023] [Accepted: 12/08/2023] [Indexed: 01/10/2024]
Abstract
Reuterin is a natural antifungal agent derived from certain strains of Limosilactobacillus reuteri. Our previous study revealed that 6 mM reuterin inhibited completely the conidial germination of aflatoxigenic Aspergillus flavus. This study investigated the potential molecular mechanism of reuterin in inhibiting A. flavus conidial germination, which was pre-assumed that it correlated to the inhibition of some essential enzyme activity involved in conidial germination, specifically 1,3-β-glucan synthase, chitin synthase, and catalases (catalase, bifunctional catalase-peroxidase, and spore-specific catalase). The complex of 1,3-β-glucan synthase and chitin synthase with reuterin had a lower binding affinity than that with the substrate. Conversely, the complex of catalases with reuterin had a higher binding affinity than that with the substrate. It was suggested that 1,3-β-glucan synthase and chitin synthase tended to bind the substrate rather than bind reuterin. In contrast, catalases tended to bind reuterin rather than bind the substrate. Therefore, reuterin could be a potential inhibitor of catalases but may not be an inhibitor of 1,3-β-glucan synthase and chitin synthase. In this in silico study, we predicted that the potential molecular mechanism of reuterin in inhibiting A. flavus conidial germination was due to the inhibition of catalases activities by competitively binding to the enzymes active sites, thus resulting in the accumulation of reactive oxygen species in cells, leading to cells damage. PRACTICAL APPLICATION: This in silico study revealed that reuterin is a potential inhibitor of catalases in A. flavus, thereby interfering with the antioxidant system during conidial germination. This finding shows that reuterin can be used as an antifungal agent in food or agricultural products, inhibiting conidial germination completely.
Collapse
Affiliation(s)
- Widiati Purnawita
- Department of Food Science and Technology, Faculty of Agricultural Engineering and Technology, IPB University (Bogor Agricultural University), Bogor, Indonesia
| | - Winiati Pudji Rahayu
- Department of Food Science and Technology, Faculty of Agricultural Engineering and Technology, IPB University (Bogor Agricultural University), Bogor, Indonesia
- Southeast Asian Food and Agricultural Science and Technology (SEAFAST) Center, IPB University (Bogor Agricultural University), Bogor, Indonesia
| | - Hanifah Nuryani Lioe
- Department of Food Science and Technology, Faculty of Agricultural Engineering and Technology, IPB University (Bogor Agricultural University), Bogor, Indonesia
- Southeast Asian Food and Agricultural Science and Technology (SEAFAST) Center, IPB University (Bogor Agricultural University), Bogor, Indonesia
| | - Siti Nurjanah
- Department of Food Science and Technology, Faculty of Agricultural Engineering and Technology, IPB University (Bogor Agricultural University), Bogor, Indonesia
- Southeast Asian Food and Agricultural Science and Technology (SEAFAST) Center, IPB University (Bogor Agricultural University), Bogor, Indonesia
| | - Setyanto Tri Wahyudi
- Southeast Asian Food and Agricultural Science and Technology (SEAFAST) Center, IPB University (Bogor Agricultural University), Bogor, Indonesia
- Tropical Biopharmaca Research Center, IPB University (Bogor Agricultural University), Bogor, Indonesia
| |
Collapse
|
11
|
Kwain S, Dominy BN, Whitehead KJ, Miller BA, Whitehead DC. Exploring the interactive mechanism of acarbose with the amylase SusG in the starch utilization system of the human gut symbiont Bacteroides thetaiotaomicron through molecular modeling. Chem Biol Drug Des 2023; 102:486-499. [PMID: 37062591 DOI: 10.1111/cbdd.14251] [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: 01/24/2023] [Revised: 03/15/2023] [Accepted: 04/04/2023] [Indexed: 04/18/2023]
Abstract
The α-amylase, SusG, is a principal component of the Bacteroides thetaiotaomicron (Bt) starch utilization system (Sus) used to metabolize complex starch molecules in the human gastrointestinal (GI) tract. We previously reported the non-microbicidal growth inhibition of Bt by the acarbose-mediated arrest of the Sus as a potential therapeutic strategy. Herein, we report a computational approach using density functional theory (DFT), molecular docking, and molecular dynamics (MD) simulation to explore the interactive mechanism between acarbose and SusG at the atomic level in an effort to understand how acarbose shuts down the Bt Sus. The docking analysis reveals that acarbose binds orthosterically to SusG with a binding affinity of -8.3 kcal/mol. The MD simulation provides evidence of conformational variability of acarbose at the active site of SusG and also suggests that acarbose interacts with the main catalytic residues via a general acid-base double-displacement catalytic mechanism. These results suggest that small molecule competitive inhibition against the SusG protein could impact the entire Bt Sus and eliminate or reduce the system's ability to metabolize starch. This computational strategy could serve as a potential avenue for structure-based drug design to discover other small molecules capable of inhibiting the Sus of Bt with high potency, thus providing a holistic approach for selective modulation of the GI microbiota.
Collapse
Affiliation(s)
- Samuel Kwain
- Department of Chemistry, Clemson University, Clemson, South Carolina, USA
| | - Brian N Dominy
- Department of Chemistry, Clemson University, Clemson, South Carolina, USA
| | - Kristi J Whitehead
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
| | - Brock A Miller
- Department of Chemistry, Clemson University, Clemson, South Carolina, USA
| | - Daniel C Whitehead
- Department of Chemistry, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
12
|
Scaffa PMC, Kendall A, Icimoto MY, Fugolin APP, Logan MG, DeVito-Moraes AG, Lewis SH, Zhang H, Wu H, Pfeifer CS. The potential use of glycosyl-transferase inhibitors for targeted reduction of S. mutans biofilms in dental materials. Sci Rep 2023; 13:11889. [PMID: 37482546 PMCID: PMC10363545 DOI: 10.1038/s41598-023-39125-2] [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: 05/01/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023] Open
Abstract
Streptococcus mutans is the primary oral caries-forming bacteria, adept at producing "sticky" biofilms via the synthesis of insoluble extracellular polysaccharides (EPS), catalyzed by glucosyltransferases (GTFs). To circumvent the use of broad-spectrum antibiotics to combat these bacteria, this study sought to modify existing EPS-targeting small molecules with the ultimate goal of producing anti-biofilm polymer surfaces specifically targeting S. mutans. To achieve this, a known GTF inhibitor (G43) was modified with methoxy or tetraethyleneglycol substitutions in different positions (nine derivatives, tested at 50-µM) to pinpoint potential sites for future methacrylate functionalization, and then assessed against single-species S. mutans biofilms. As expected, the compounds did not diminish the bacterial viability. In general, the compounds with methoxy substitution were not effective in reducing EPS formation, whereas the tetraethyleneglycol substitution (G43-C3-TEG) led to a decrease in the concentration of insoluble EPS, although the effect is less pronounced than for the parent G43. This aligns with the reduced GTF-C activity observed at different concentrations of G43-C3-TEG, as well as the consequent decrease in EPS formation, and notable structural changes. In summary, this study determined that G43-C3-TEG is non-bactericidal and can selectively reduce the biofilm formation, by decreasing the production of EPS. This molecule will serve to functionalize surfaces of materials to be tested in future research.
Collapse
Affiliation(s)
- Polliana Mendes Candia Scaffa
- Division of Biomaterial and Biomedical Sciences, Department of Oral Rehabilitation and Biosciences, Oregon Health & Science University, OHSU, 2730 S Moody Ave., Portland, OR, 97201, USA
| | - Alexander Kendall
- Division of Biomaterial and Biomedical Sciences, Department of Oral Rehabilitation and Biosciences, Oregon Health & Science University, OHSU, 2730 S Moody Ave., Portland, OR, 97201, USA
| | - Marcelo Yudi Icimoto
- Division of Biomaterial and Biomedical Sciences, Department of Oral Rehabilitation and Biosciences, Oregon Health & Science University, OHSU, 2730 S Moody Ave., Portland, OR, 97201, USA
- Department of Biophysics, Federal University of Sao Paulo, UNIFESP-EPM, R. Sena Madureira, 1500, Sao Paulo, SP, 04021-001, Brazil
| | - Ana Paula Piovezan Fugolin
- Division of Biomaterial and Biomedical Sciences, Department of Oral Rehabilitation and Biosciences, Oregon Health & Science University, OHSU, 2730 S Moody Ave., Portland, OR, 97201, USA
| | - Matthew G Logan
- Division of Biomaterial and Biomedical Sciences, Department of Oral Rehabilitation and Biosciences, Oregon Health & Science University, OHSU, 2730 S Moody Ave., Portland, OR, 97201, USA
| | - Andre G DeVito-Moraes
- Division of Biomaterial and Biomedical Sciences, Department of Oral Rehabilitation and Biosciences, Oregon Health & Science University, OHSU, 2730 S Moody Ave., Portland, OR, 97201, USA
| | - Steven H Lewis
- Division of Biomaterial and Biomedical Sciences, Department of Oral Rehabilitation and Biosciences, Oregon Health & Science University, OHSU, 2730 S Moody Ave., Portland, OR, 97201, USA
| | - Hua Zhang
- Division of Biomaterial and Biomedical Sciences, Department of Oral Rehabilitation and Biosciences, Oregon Health & Science University, OHSU, 2730 S Moody Ave., Portland, OR, 97201, USA
| | - Hui Wu
- Division of Biomaterial and Biomedical Sciences, Department of Oral Rehabilitation and Biosciences, Oregon Health & Science University, OHSU, 2730 S Moody Ave., Portland, OR, 97201, USA
| | - Carmem S Pfeifer
- Division of Biomaterial and Biomedical Sciences, Department of Oral Rehabilitation and Biosciences, Oregon Health & Science University, OHSU, 2730 S Moody Ave., Portland, OR, 97201, USA.
| |
Collapse
|
13
|
Bouargalne Y, Guilbaud F, Macherel D, Delalande O, Deleu C, Le Cahérec F. Brassica napus Drought-Induced 22-kD Protein (BnD22) Acts Simultaneously as a Cysteine Protease Inhibitor and Chlorophyll-Binding Protein. PLANT & CELL PHYSIOLOGY 2023; 64:536-548. [PMID: 36905393 DOI: 10.1093/pcp/pcad016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/20/2023] [Accepted: 03/09/2023] [Indexed: 05/17/2023]
Abstract
Class II water-soluble chlorophyll proteins (WSCPs) from Brassicaceae are non-photosynthetic proteins that bind with chlorophyll (Chl) and its derivatives. The physiological function of WSCPs is still unclear, but it is assumed to be involved in stress responses, which is likely related to their Chl-binding and protease inhibition (PI) activities. Yet, the dual function and simultaneous functionality of WSCPs must still be better understood. Here, the biochemical functions of Brassica napus drought-induced 22-kDa protein (BnD22), a major WSCP expressed in B. napus leaves, were investigated using recombinant hexahistidine-tagged protein. We showed that BnD22 inhibited cysteine proteases, such as papain, but not serine proteases. BnD22 was able to bind with Chla or Chlb to form tetrameric complexes. Unexpectedly, BnD22-Chl tetramer displays higher inhibition toward cysteine proteases, indicating (i) simultaneous Chl-binding and PI activities and (ii) Chl-dependent activation of PI activity of BnD22. Moreover, the photostability of BnD22-Chl tetramer was reduced upon binding with the protease. Using three-dimensional structural modeling and molecular docking, we revealed that Chl binding favors interaction between BnD22 and proteases. Despite its Chl-binding ability, the BnD22 was not detected in chloroplasts but rather in the endoplasmic reticulum and vacuole. In addition, the C-terminal extension peptide of BnD22, which cleaved off post-translationally in vivo, was not implicated in subcellular localization. Instead, it drastically promoted the expression, solubility and stability of the recombinant protein.
Collapse
Affiliation(s)
| | - Florian Guilbaud
- IGEPP, INRAE, Institut Agro, Université Rennes, Rennes 35000, France
| | - David Macherel
- IRHS, INRAE, Institut Agro, Université Angers, Angers 49000, France
| | | | - Carole Deleu
- IGEPP, INRAE, Institut Agro, Université Rennes, Rennes 35000, France
| | | |
Collapse
|
14
|
Das A, Sharma HK, Lather V, Pandita D, Agarwal P. Structure-based virtual screening for identification of potential CDC20 inhibitors and their therapeutic evaluation in breast cancer. 3 Biotech 2023; 13:141. [PMID: 37124982 PMCID: PMC10133423 DOI: 10.1007/s13205-023-03554-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 04/06/2023] [Indexed: 05/02/2023] Open
Abstract
Cell division cycle 20 (CDC20), a critical partner of anaphase promoting complex (APC/C), is indispensably required for metaphase-to-anaphase transition. CDC20 overexpression in TNBC breast cancer patients has been found to be correlated with poor prognosis, hence, we aimed to target CDC20 for TNBC therapeutics. In silico molecular docking of large-scale chemical libraries (phytochemicals/synthetic drugs) against CDC20 protein structure identified five synthetic drugs and four phytochemicals as potential hits interacting with CDC20 active site. The molecular selection was done based on docking scores, binding interactions, binding energies and MM/GBSA scores. Further, we analysed ADME profiles for all the hits and identified lidocaine, an aminoamide anaesthetic group of synthetic drug, with high drug-likeness properties. We explored the anti-tumorigenic effects of lidocaine on MDA-MB-231 TNBC breast cancer cells, which resulted in increased growth inhibition in dose-dependent manner. The molecular mechanism behind the cell viability defect mediated by lidocaine was found to be induction of G2/M cell cycle arrest and cellular apoptosis. Notably, lidocaine treatment of TNBC cells also resulted in downregulation of CDC20 gene expression. Thus, this study identifies lidocaine as a potential anti-neoplastic agent for TNBC cells emphasizing CDC20 as a suitable therapeutic target for breast cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03554-7.
Collapse
Affiliation(s)
- Amiya Das
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Sector 125, Noida, 201313 India
| | - Hitesh Kumar Sharma
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Sector 125, Noida, 201313 India
| | - Viney Lather
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Sector 125, Noida, 201313 India
| | - Deepti Pandita
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research (DIPSAR) Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, Government of NCT of Delhi, New Delhi, 110017 India
- Centre for Advanced Formulation Technology (CAFT), Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017 India
| | - Pallavi Agarwal
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Sector 125, Noida, 201313 India
| |
Collapse
|
15
|
Kasarla SS, Borse SP, Kumar Y, Sharma N, Dikshit M. In vitro effect of Withania somnifera, AYUSH-64, and remdesivir on the activity of CYP-450 enzymes: Implications for possible herb−drug interactions in the management of COVID-19. Front Pharmacol 2022; 13:973768. [PMID: 36313313 PMCID: PMC9597875 DOI: 10.3389/fphar.2022.973768] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/29/2022] [Indexed: 11/22/2022] Open
Abstract
Ayurvedic medicines Withania somnifera Dunal (ashwagandha) and AYUSH-64 have been used for the prevention and management of COVID-19 in India. The present study explores the effect of Ashwagandha and AYUSH-64 on important human CYP enzymes (CYP3A4, CYP2C8, and CYP2D6) to assess their interaction with remdesivir, a drug used for COVID-19 management during the second wave. The study also implies possible herb−drug interactions as ashwagandha and AYUSH-64 are being used for managing various pathological conditions. Aqueous extracts of ashwagandha and AYUSH-64 were characterized using LC-MS/MS. A total of 11 and 24 phytoconstituents were identified putatively from ashwagandha and AYUSH-64 extracts, respectively. In addition, in silico studies revealed good ADME properties of most of the phytoconstituents of these herbal drugs and suggested that some of these might possess CYP-450 inhibitory activity. In vitro CYP-450 studies with human liver microsomes showed moderate inhibition of CYP3A4, 2C8, and 2D6 by remdesivir, while ashwagandha had no inhibitory effect alone or in combination with remdesivir. AYUSH-64 also exhibited a similar trend; however, a moderate inhibitory effect on CYP2C8 was noticed. Thus, ashwagandha seems to be safe to co-administer with the substrates of CYP3A4, CYP2C8, and CYP2D6. However, caution is warranted in prescribing AYUSH-64 along with CYP2C8 substrate drugs. Furthermore, preclinical and clinical PK studies would be helpful for their effective and safer use in the management of various ailments along with other drugs.
Collapse
Affiliation(s)
- Siva Swapna Kasarla
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
- Spatial Metabolomics Group, Leibniz-Institut für Analytische Wissenschaften-ISAS-e V, Dortmund, Germany
| | - Swapnil P. Borse
- AYUSH - Center of Excellence (AYUSH-CoE), Center for Complementary and Integrative Health [CCIH], Interdisciplinary School of Health Sciences (ISHS), Savitribai Phule Pune University Pune (SPPU), Pune, India
- *Correspondence: Swapnil P. Borse, ; Yashwant Kumar, ; Madhu Dikshit,
| | - Yashwant Kumar
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
- *Correspondence: Swapnil P. Borse, ; Yashwant Kumar, ; Madhu Dikshit,
| | - Neha Sharma
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
- Department of Pharmaceutical Analysis, Delhi Pharmaceutical Science and Research University, Pushp Vihar, New Delhi, India
| | - Madhu Dikshit
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
- CSIR- Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- *Correspondence: Swapnil P. Borse, ; Yashwant Kumar, ; Madhu Dikshit,
| |
Collapse
|
16
|
Evaluation of vatiquinone drug-drug interaction potential in vitro and in a phase 1 clinical study with tolbutamide, a CYP2C9 substrate, and omeprazole, a CYP2C19 substrate, in healthy subjects. Eur J Clin Pharmacol 2022; 78:1823-1831. [PMID: 36166059 DOI: 10.1007/s00228-022-03393-0] [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: 07/19/2022] [Accepted: 09/15/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE In this study, the drug-drug interaction potential of vatiquinone with cytochrome P450 (CYP) substrates was investigated in both in vitro and clinical studies. METHODS The inhibitory potential of vatiquinone on the activity of CYPs 1A2, 2B6, 2C8, 2C9, 2C19, 2D6, and 3A4/5 was assessed in vitro. In an open-label, drug-drug interaction study in 18 healthy human subjects, a single oral dose of 500 mg tolbutamide and 40 mg omeprazole was administered on day 1, followed by a washout of 7 days. Multiple oral doses of 400 mg vatiquinone (three times a day [TID]) were administered from day 8 to day 13 with coadministration of a single oral dose of 500 mg tolbutamide and 40 mg omeprazole on day 12. RESULTS In vitro, vatiquinone inhibited CYP2C9 (IC50 = 3.7 µM) and CYP2C19 (IC50 = 5.4 µM). In the clinical study, coadministration of vatiquinone did not affect the pharmacokinetic (PK) profile of tolbutamide and omeprazole. The 90% confidence intervals (CIs) of geometric least-square mean ratios for maximum plasma concentration (Cmax), areas under the plasma concentration-time curve (AUC0-t), and AUC0-inf of tolbutamide and omeprazole were entirely contained within the 80 to 125% no effect limit, except a minor excursion observed for Cmax of omeprazole (geometric mean ratio [GMR], 94.09; 90% CI, 78.70-112.50). Vatiquinone was generally well tolerated, and no clinically significant findings were reported. CONCLUSION The in vitro and clinical studies demonstrated vatiquinone has a low potential to affect the pharmacokinetics of concomitantly administered medications that are metabolized by CYP enzymes.
Collapse
|
17
|
Bhatt S, Dhiman S, Kumar V, Gour A, Manhas D, Sharma K, Ojha PK, Nandi U. Assessment of the CYP1A2 Inhibition-Mediated Drug Interaction Potential for Pinocembrin Using In Silico, In Vitro, and In Vivo Approaches. ACS OMEGA 2022; 7:20321-20331. [PMID: 35721953 PMCID: PMC9202019 DOI: 10.1021/acsomega.2c02315] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/24/2022] [Indexed: 05/23/2023]
Abstract
Pinocembrin, a bioflavonoid, is extensively used in complementary/alternative medicine. It turns out as a promising candidate against neurodegenerative diseases because of its multifaceted pharmacological action toward neuroprotection. However, literature evidence is still lacking for its inhibitory action on CYP1A2, which is responsible for xenobiotic metabolism leading to the generation of toxic metabolites and bioactivation of procarcinogens. In the present study, our aim was to evaluate the CYP1A2 inhibitory potential of pinocembrin via in silico, in vitro, and in vivo investigations. From the results of in vitro studies, pinocembrin is found to be a potent and competitive inhibitor of CYP1A2. In vitro-in vivo extrapolation results indicate the potential of pinocembrin to interact with CYP1A2 substrate drugs clinically. Molecular docking-based in silico studies demonstrate the strong interaction of pinocembrin with human CYP1A2. In in vivo investigations using a rat model, pinocembrin displayed a marked alteration in the plasma exposure of CYP1A2 substrate drugs, namely, caffeine and tacrine. In conclusion, pinocembrin has a potent CYP1A2 inhibitory action to cause drug interactions, and further confirmatory study is warranted at the clinical level.
Collapse
Affiliation(s)
- Shipra Bhatt
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sumit Dhiman
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
| | - Vinay Kumar
- Drug Theoretics
and Chemoinformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Abhishek Gour
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Diksha Manhas
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kuhu Sharma
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
| | - Probir Kumar Ojha
- Drug Theoretics
and Chemoinformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Utpal Nandi
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
18
|
Martin P, Czerwiński M, Limaye PB, Ogilvie BW, Smith S, Boyd B. In vitro evaluation suggests fenfluramine and norfenfluramine are unlikely to act as perpetrators of drug interactions. Pharmacol Res Perspect 2022; 10:e00959. [PMID: 35599347 PMCID: PMC9124818 DOI: 10.1002/prp2.959] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022] Open
Abstract
Studies support the safety and efficacy of fenfluramine (FFA) as an antiseizure medication (ASM) in Dravet syndrome, Lennox-Gastaut syndrome, or CDKL5 deficiency disorder, all pharmacoresistant developmental and epileptic encephalopathies. However, drug-drug interactions with FFA in multi-ASM regimens have not been fully investigated. We characterized the perpetrator potential of FFA and its active metabolite, norfenfluramine (nFFA), in vitro by assessing cytochrome P450 (CYP450) inhibition in human liver microsomes, CYP450 induction in cultured human hepatocytes, and drug transporter inhibition potential in permeability or cellular uptake assays. Mean plasma unbound fraction was ~50% for both FFA and nFFA, with no apparent concentration dependence. FFA and nFFA were direct in vitro inhibitors of CYP2D6 (IC50 , 4.7 and 16 µM, respectively) but did not substantially inhibit CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, or CYP3A4/5. No time- or metabolism-dependent CYP450 inhibition occurred. FFA and nFFA did not induce CYP1A2; both induced CYP2B6 (up to 2.8-fold and up to 2.0-fold, respectively) and CYP3A4 (1.9- to 3.0-fold and 3.6- to 4.8-fold, respectively). Mechanistic static pharmacokinetic models predicted that neither CYP450 inhibition nor induction was likely to be clinically relevant at doses typically used for seizure reduction (ratio of area under curve [AUCR] for inhibition <1.25; AUCR for induction >0.8). Transporters OCT2 and MATE1 were inhibited by FFA (IC50 , 19.8 and 9.0 μM) and nFFA (IC50 , 5.2 and 4.6 μM) at concentrations higher than clinically achievable; remaining transporters were not inhibited. Results suggest that FFA and nFFA are unlikely drug-drug interaction perpetrators at clinically relevant doses of FFA (0.2-0.7 mg/kg/day).
Collapse
|
19
|
Andrade JM, Pachar P, Trujillo L, Cartuche L. Suillin: A mixed-type acetylcholinesterase inhibitor from Suillus luteus which is used by Saraguros indigenous, southern Ecuador. PLoS One 2022; 17:e0268292. [PMID: 35576219 PMCID: PMC9109927 DOI: 10.1371/journal.pone.0268292] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 04/27/2022] [Indexed: 11/19/2022] Open
Abstract
Suillus luteus (L.) Roussel is an edible mushroom commonly known as slippery jack or “Kallampa” by indigenous people from Loja province. It is used in traditional medicine to manage gastrointestinal disorders and headaches. In addition, edible mushrooms have been used for neurodegenerative diseases; however, there is no report about the anticholinesterase effect produced by this species. The aim of this work was to isolate the main secondary metabolite of Suillus luteus and characterize its inhibitory potential against acetylcholinesterase. Fruiting bodies were extracted with ethanol (EtOH) and ethyl acetate (EtOAc). From the EtOAc, suillin, is reported as the major compound. The cholinesterase inhibitory potential of extracts and the major isolated compound was assessed by Ellman´s method and progression curves were recorded at 405 nm for 60 min. Donepezil hydroclhoride was used as a positive control. The samples were dissolved in methanol at 10 mg/mL and two more 10× dilutions were included to obtain final concentrations of 1, 0.1 and 0.01 mg/mL at the mix of reaction. IC50, Km, Vmax, and Ki were calculated for suillin. Suillin (200 mg) along with linoleic acid, ergosterol peroxide and ergosterol were isolated. The EtOH and EtOAc extracts exerted a moderate inhibitory effect (IC50 > 200 μg/mL. In adittion, suillin exerted a non-competitive mixed mechanism. against AChE with an IC50 value of 31.50 μM and Ki of 17.25 μM. To the best of our knowledge, this is the first report of the anticholinesterase effect of Suillus luteus and suillin. The kinetic parameters and the moderate potency of the compound determined in this study, encourage us to propose suillin as a promising chemopreventing agent for the treatment of neurodegenerative diseases such as Alzheimer.
Collapse
Affiliation(s)
- José Miguel Andrade
- Departamento de Química, Universidad Técnica Particular de Loja (UTPL), Loja, Ecuador
| | - Pamela Pachar
- Departamento de Química, Universidad Técnica Particular de Loja (UTPL), Loja, Ecuador
| | - Luisa Trujillo
- Departamento de Química, Universidad Técnica Particular de Loja (UTPL), Loja, Ecuador
| | - Luis Cartuche
- Departamento de Química, Universidad Técnica Particular de Loja (UTPL), Loja, Ecuador
- * E-mail:
| |
Collapse
|
20
|
Kinetic Modeling of Time-Dependent Enzyme Inhibition by Pre-Steady-State Analysis of Progress Curves: The Case Study of the Anti-Alzheimer's Drug Galantamine. Int J Mol Sci 2022; 23:ijms23095072. [PMID: 35563466 PMCID: PMC9105972 DOI: 10.3390/ijms23095072] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 01/27/2023] Open
Abstract
The Michaelis–Menten model of enzyme kinetic assumes the free ligand approximation, the steady-state approximation and the rapid equilibrium approximation. Analytical methods to model slow-binding inhibitors by the analysis of initial velocities have been developed but, due to their inherent complexity, they are seldom employed. In order to circumvent the complications that arise from the violation of the rapid equilibrium assumption, inhibition is commonly evaluated by pre-incubating the enzyme and the inhibitors so that, even for slow inhibitors, the binding equilibrium is established before the reaction is started. Here, we show that for long drug-target residence time inhibitors, the conventional analysis of initial velocities by the linear regression of double-reciprocal plots fails to provide a correct description of the inhibition mechanism. As a case study, the inhibition of acetylcholinesterase by galantamine, a drug approved for the symptomatic treatment of Alzheimer’s disease, is reported. For over 50 years, analysis based on the conventional steady-state model has overlooked the time-dependent nature of galantamine inhibition, leading to an erroneous assessment of the drug potency and, hence, to discrepancies between biochemical data and the pharmacological evidence. Re-examination of acetylcholinesterase inhibition by pre-steady state analysis of the reaction progress curves showed that the potency of galantamine has indeed been underestimated by a factor of ~100.
Collapse
|
21
|
Bhatt S, Manhas D, Kumar V, Gour A, Sharma K, Dogra A, Ojha PK, Nandi U. Effect of Myricetin on CYP2C8 Inhibition to Assess the Likelihood of Drug Interaction Using In Silico, In Vitro, and In Vivo Approaches. ACS OMEGA 2022; 7:13260-13269. [PMID: 35474783 PMCID: PMC9026026 DOI: 10.1021/acsomega.2c00726] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/16/2022] [Indexed: 05/05/2023]
Abstract
Myricetin, a bioflavonoid, is widely used as functional food/complementary medicine and has promising multifaceted pharmacological actions against therapeutically validated anticancer targets. On the other hand, CYP2C8 is not only crucial for alteration in the pharmacokinetics of drugs to cause drug interaction but also unequivocally important for the metabolism of endogenous substances like the formation of epoxyeicosatrienoic acids (EETs), which are considered as signaling molecules against hallmarks of cancer. However, there is hardly any information known to date about the effect of myricetin on CYP2C8 inhibition and, subsequently, the CYP2C8-mediated drug interaction potential of myricetin at the preclinical/clinical level. We aimed here to explore the CYP2C8 inhibitory potential of myricetin using in silico, in vitro, and in vivo investigations. In the in vitro study, myricetin showed a substantial effect on CYP2C8 inhibition in human liver microsomes using CYP2C8-catalyzed amodiaquine-N-deethylation as an index reaction. Considering the Lineweaver-Burk plot, the Dixon plot, and the higher α-value, myricetin is found to be a mixed type of CYP2C8 inhibitor. Moreover, in vitro-in vivo extrapolation data suggest that myricetin is likely to cause drug interaction at the hepatic level. The molecular docking study depicted a strong interaction between myricetin and the active site of the human CYP2C8 enzyme. Moreover, myricetin caused considerable elevation in the oral exposure of amodiaquine as a CYP2C8 substrate via a slowdown of amodiaquine clearance in the rat model. Overall, the potent action of myricetin on CYP2C8 inhibition indicates that there is a need for further exploration to avoid drug interaction-mediated precipitation of obvious adverse effects as well as to optimize anticancer therapy.
Collapse
Affiliation(s)
- Shipra Bhatt
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Diksha Manhas
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Vinay Kumar
- Drug
Theoretics and Chemoinformatics Laboratory, Department of Pharmaceutical
Technology, Jadavpur University, Kolkata 700032, India
| | - Abhishek Gour
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kuhu Sharma
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
| | - Ashish Dogra
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Probir Kumar Ojha
- Drug
Theoretics and Chemoinformatics Laboratory, Department of Pharmaceutical
Technology, Jadavpur University, Kolkata 700032, India
| | - Utpal Nandi
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- ,
| |
Collapse
|
22
|
Physiologically based pharmacokinetic model predictions of natural product-drug interactions between goldenseal, berberine, imatinib and bosutinib. Eur J Clin Pharmacol 2022; 78:597-611. [DOI: 10.1007/s00228-021-03266-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 12/13/2021] [Indexed: 11/03/2022]
|
23
|
Study on the Effect of Three CYP2C9 Variants on Drug–Drug Interaction Related to Six Drugs In Vitro by LC–MS/MS Method. Chromatographia 2022. [DOI: 10.1007/s10337-021-04126-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
24
|
Fu S, Yu F, Hu Z, Sun T. Metabolism-Mediated Drug-Drug Interactions – Study Design, Data Analysis, and Implications for In Vitro Evaluations. MEDICINE IN DRUG DISCOVERY 2022. [DOI: 10.1016/j.medidd.2022.100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
25
|
Valicherla GR, Graebing P, Zhang J, Zheng R, Nuttall J, Silvera P, Rohan LC. Investigating the Contribution of Drug-Metabolizing Enzymes in Drug-Drug Interactions of Dapivirine and Miconazole. Pharmaceutics 2021; 13:2193. [PMID: 34959473 PMCID: PMC8706783 DOI: 10.3390/pharmaceutics13122193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 11/17/2022] Open
Abstract
Dapivirine (DPV) is a potent NNRTI used to prevent the sexual transmission of HIV. In a phase 1 trial (IPM 028), the concomitant use of a DPV vaginal ring and an antifungal miconazole (MIC) vaginal capsule was found to increase the systemic exposure to DPV in women, suggesting a potential for drug-drug interactions. This study's objective was to investigate the mechanism of DPV-MIC interactions using drug-metabolizing enzymes (DMEs; CYPs and UGTs) that are locally expressed in the female reproductive tract (FRT). In vitro studies were performed to evaluate the metabolism of DPV and its inhibition and induction potential with DMEs. In addition, the impact of MIC on DPV metabolism and the inhibitory potential of DPV with DMEs were studied. Our findings suggest that DPV is a substrate of CYP1A1 and CYP3A4 enzymes and that MIC significantly decreased the DPV metabolism by inhibiting these two enzymes. DPV demonstrated potent inhibition of CYP1A1 and moderate/weak inhibition of the six CYP and eight UGT enzymes evaluated. MIC showed potent/moderate inhibition of seven CYP enzymes and weak/no inhibition of eight UGT enzymes. The combination of DPV and MIC showed potent inhibition of seven CYP enzymes (1A1, 1A2, 1B1, 2B6, 2C8, 2C19, and 3A4) and four UGT enzymes (1A3, 1A6, 1A9, and 2B7). DPV was not an inducer of CYP1A2, CYP2B6, and CYP3A4 enzymes in primary human hepatocytes. Therefore, the increased systemic concentrations of DPV observed in IPM 028 were likely due to the reduced metabolism of DPV because of CYP1A1 and CYP3A4 enzymes inhibition by MIC in the FRT.
Collapse
Affiliation(s)
- Guru Raghavendra Valicherla
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA; (G.R.V.); (P.G.); (J.Z.); (R.Z.)
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Phillip Graebing
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA; (G.R.V.); (P.G.); (J.Z.); (R.Z.)
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Junmei Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA; (G.R.V.); (P.G.); (J.Z.); (R.Z.)
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Ruohui Zheng
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA; (G.R.V.); (P.G.); (J.Z.); (R.Z.)
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Jeremy Nuttall
- International Partnership for Microbicides, Silver Spring, MD 20910, USA;
| | - Peter Silvera
- Advanced Bioscience Laboratories, Rockville, MD 20850, USA;
| | - Lisa Cencia Rohan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA; (G.R.V.); (P.G.); (J.Z.); (R.Z.)
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
26
|
Hanapi NA, Chear NJY, Azizi J, Yusof SR. Kratom Alkaloids: Interactions With Enzymes, Receptors, and Cellular Barriers. Front Pharmacol 2021; 12:751656. [PMID: 34867362 PMCID: PMC8637859 DOI: 10.3389/fphar.2021.751656] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/26/2021] [Indexed: 01/11/2023] Open
Abstract
Parallel to the growing use of kratom, there is a wealth of evidence from self-report, preclinical, and early clinical studies on therapeutic benefits of its alkaloids in particular for treating pain, managing substance use disorder, and coping with emotional or mental health conditions. On the other hand, there are also reports on potential health risks concerning kratom use. These two aspects are often discussed in reviews on kratom. Here, we aim to highlight specific areas that are of importance to give insights into the mechanistic of kratom alkaloids pharmacological actions. This includes their interactions with drug-metabolizing enzymes and predictions of clinical drug-drug interactions, receptor-binding properties, interactions with cellular barriers in regards to barrier permeability, involvement of membrane transporters, and alteration of barrier function when exposed to the alkaloids.
Collapse
Affiliation(s)
- Nur Aziah Hanapi
- Centre for Drug Research, Universiti Sains Malaysia, Minden, Malaysia
| | | | - Juzaili Azizi
- Centre for Drug Research, Universiti Sains Malaysia, Minden, Malaysia
| | - Siti R Yusof
- Centre for Drug Research, Universiti Sains Malaysia, Minden, Malaysia
| |
Collapse
|
27
|
How Science Is Driving Regulatory Guidances. Methods Mol Biol 2021. [PMID: 34272707 DOI: 10.1007/978-1-0716-1554-6_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
This chapter provides regulatory perspectives on how to translate in vitro drug metabolism findings into in vivo drug-drug interaction (DDI) predictions and how this affects the decision of conducting in vivo DDI evaluation. The chapter delineates rationale and analyses that have supported the recommendations in the U.S. Food and Drug Administration (FDA) DDI guidances in terms of in vitro-in vivo extrapolation of cytochrome P450 (CYP) inhibition-mediated DDI potential for investigational new drugs and their metabolites as substrates or inhibitors. The chapter also describes the framework and considerations to assess UDP-glucuronosyltransferase (UGT) inhibition-mediated DDI potential for drugs as substrates or inhibitors. The limitations of decision criteria and further improvements needed are also discussed. Case examples are provided throughout the chapter to illustrate how decision criteria have been utilized to evaluate in vivo DDI potential from in vitro data.
Collapse
|
28
|
Xu Q, Yu F, Hao Z, Wu W, Sun Y, Wang T, Li G, Lv Q, Hu Z. Metabolism and transporter based drug–drug interaction of tacrolimus with nine co-medicated injections. MEDICINE IN DRUG DISCOVERY 2021. [DOI: 10.1016/j.medidd.2021.100091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
29
|
Wang X, Wang Z, Fan X, Yan M, Jiang L, Xia Y, Cao J, Liu Y. Comparison of the drug-drug interactions potential of ibrutinib and acalabrutinib via inhibition of UDP-glucuronosyltransferase. Toxicol Appl Pharmacol 2021; 424:115595. [PMID: 34038714 DOI: 10.1016/j.taap.2021.115595] [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: 01/31/2021] [Revised: 05/18/2021] [Accepted: 05/21/2021] [Indexed: 11/26/2022]
Abstract
Ibrutinib and acalabrutinib are two Bruton's tyrosine kinase (BTK) inhibitors which have gained Food and Drug Administration (FDA) approval for the treatment of various B cell malignancies. Herein, we investigated the effects of the two drugs on UDP-glucuronosyltransferase (UGT) activities to evaluate their potential risk for drug-drug interactions (DDIs) via UGT inhibition. Our data indicated that ibrutinib exerted broad inhibition on most of UGTs, including a potent competitive inhibition against UGT1A1 with a Ki value of 0.90 ± 0.03 μM, a noncompetitive inhibition against UGT1A3 and UGT1A7 with Ki values of 0.88 ± 0.03 μM and 2.52 ± 0.23 μM, respectively, while acalabrutinib only exhibited weak UGT inhibition towards all tested UGT isoforms. DDI risk prediction suggested that the inhibition against UGT1A1 and UGT1A3 by ibrutinib might bring a potential DDIs risk, while acalabrutinib was unlikely to trigger clinically significant UGT-mediated DDIs due to its weak effects. Our study raises an alarm bell about potential DDI risk associated with ibrutinib, however, the extrapolation from in vitro data to in vivo drug interactions should be taken with caution, and additional systemic study is needed.
Collapse
Affiliation(s)
- Xiaoyu Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Zhe Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Xiaoyu Fan
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Mingrui Yan
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Lili Jiang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Yangliu Xia
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Jun Cao
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian 116044, China.
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China.
| |
Collapse
|
30
|
Kumar S, Bouic PJ, Rosenkranz B. Investigation of CYP2B6, 3A4 and β-esterase interactions of Withania somnifera (L.) dunal in human liver microsomes and HepG2 cells. JOURNAL OF ETHNOPHARMACOLOGY 2021; 270:113766. [PMID: 33395575 DOI: 10.1016/j.jep.2020.113766] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 10/27/2020] [Accepted: 12/24/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Withania somnifera (L.) Dunal (Solanaceae) is a traditional herb, used in African indigenous systems of medicine for the treatment of various diseases (including HIV/AIDS and tuberculosis). The relevance of clinically significant interactions of Withania with ARVs and anti-TB drugs needs to be investigated. AIM OF THE STUDY This study evaluated the effects of its roots on cytochromes P450 (CYPs) 2B6, 3A4, and rifampicin metabolism pathway, using methanol, ethanol, aqueous, and ethyl acetate solvent extractions. MATERIALS AND METHODS The extracts were tested on human liver microsomes (HLM) for CYP inhibition, mRNA expression in HepG2 cells for CYP induction. Biochemical qualitative tests and LC-MS/MS methodology were used to determine active phytoconstituents. RESULTS The methanolic and ethyl acetate extracts inhibited CYP2B6 with IC50s 79.16 and 57.96 μg/ml respectively, while none of the extracts had any effect on rifampicin metabolism or showed time-dependant inhibition (TDI). All extracts were moderate inducers of CYP3A4; the aqueous extract exhibited 38%-fold shift induction of CYP3A4 compared to the control. The methanolic extract had the lowest CTC50 (50% of cytotoxicity inhibition) (67.13 ± 0.83 μg/ml). LC-MS/MS-PDA full scans were consistent with the presence of flavone salvigenin (m/z 327), alkaloid isopelletierine (m/z 133), steroidal lactone 2,3-dihydrowithaferin-A (m/z 472), and other withanolides including withaperuvin I (m/z 533), withaferin derivative (m/z 567), some of these compounds likely being responsible for the observed CYP2B6 inhibition and CYP3A4 induction. The putative gastrointestinal tract (GIT) concentration for the active extracts was 1800 μg/ml and the hepatic circulation concentrations were estimated at about 220 μg/ml and 13.5 μg/ml for the methanolic and ethyl acetate extracts, respectively. The extrapolated in vivo percentage of inhibition was at 85% for the methanolic extract against CYP2B6. CONCLUSIONS The findings reported in this study suggest that W. somnifera extracts have the potential of causing clinically significant herb-drug interactions (HDI) as moderate inducer of CYP3A4 and inhibitor of CYP2B6 metabolism pathway (methanol and ethyl acetate extracts).
Collapse
Affiliation(s)
- Saneesh Kumar
- Division of Clinical Pharmacology, University of Stellenbosch, Cape Town, South Africa.
| | - Patrick J Bouic
- Division of Medical Microbiology, University of Stellenbosch, Cape Town, South Africa; Synexa Life Sciences, Montague Gardens, Cape Town, South Africa.
| | - Bernd Rosenkranz
- Division of Clinical Pharmacology, University of Stellenbosch, Cape Town, South Africa.
| |
Collapse
|
31
|
Gonzalez SN, Mills JJ, Maugeri D, Olaya C, Laguera BL, Enders JR, Sherman J, Rodriguez A, Pierce JG, Cazzulo JJ, D'Antonio EL. Design, synthesis, and evaluation of substrate - analogue inhibitors of Trypanosoma cruzi ribose 5-phosphate isomerase type B. Bioorg Med Chem Lett 2021; 32:127723. [PMID: 33249135 DOI: 10.1016/j.bmcl.2020.127723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 11/30/2022]
Abstract
Ribose 5-phosphate isomerase type B (RPI-B) is a key enzyme of the pentose phosphate pathway that catalyzes the isomerization of ribose 5-phosphate (R5P) and ribulose 5-phosphate (Ru5P). Trypanosoma cruzi RPI-B (TcRPI-B) appears to be a suitable drug-target mainly due to: (i) its essentiality (as previously shown in other trypanosomatids), (ii) it does not present a homologue in mammalian genomes sequenced thus far, and (iii) it participates in the production of NADPH and nucleotide/nucleic acid synthesis that are critical for parasite cell survival. In this survey, we report on the competitive inhibition of TcRPI-B by a substrate - analogue inhibitor, Compound B (Ki = 5.5 ± 0.1 μM), by the Dixon method. This compound has an iodoacetamide moiety that is susceptible to nucleophilic attack, particularly by the cysteine thiol group. Compound B was conceived to specifically target Cys-69, an important active site residue. By incubating TcRPI-B with Compound B, a trypsin digestion LC-MS/MS analysis revealed the identification of Compound B covalently bound to Cys-69. This inhibitor also exhibited notable in vitro trypanocidal activity against T. cruzi infective life-stages co-cultured in NIH-3T3 murine host cells (IC50 = 17.40 ± 1.055 μM). The study of Compound B served as a proof-of-concept so that next generation inhibitors can potentially be developed with a focus on using a prodrug group in replacement of the iodoacetamide moiety, thus representing an attractive starting point for the future treatment of Chagas' disease.
Collapse
Affiliation(s)
- Soledad Natalia Gonzalez
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de General San Martín - CONICET (IIBio-UNSAM), Avenida 25 de Mayo y Francia CP (1650), San Martín (Buenos Aires), Argentina
| | - Jonathan J Mills
- Department of Chemistry, North Carolina State University, 2620 Yarbrough Drive, Box 8204, Raleigh, NC 27695, USA
| | - Dante Maugeri
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de General San Martín - CONICET (IIBio-UNSAM), Avenida 25 de Mayo y Francia CP (1650), San Martín (Buenos Aires), Argentina
| | - Christopher Olaya
- Department of Chemistry, North Carolina State University, 2620 Yarbrough Drive, Box 8204, Raleigh, NC 27695, USA
| | - Breana L Laguera
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| | - Jeffrey R Enders
- Molecular Education, Technology and Research Innovation Center (METRIC), North Carolina State University, Raleigh, NC 27695, USA
| | - Julian Sherman
- Department of Microbiology, New York University School of Medicine, 430 East 29(th) Street, New York, NY 10016, USA
| | - Ana Rodriguez
- Department of Microbiology, New York University School of Medicine, 430 East 29(th) Street, New York, NY 10016, USA
| | - Joshua G Pierce
- Department of Chemistry, North Carolina State University, 2620 Yarbrough Drive, Box 8204, Raleigh, NC 27695, USA
| | - Juan José Cazzulo
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de General San Martín - CONICET (IIBio-UNSAM), Avenida 25 de Mayo y Francia CP (1650), San Martín (Buenos Aires), Argentina
| | - Edward L D'Antonio
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA.
| |
Collapse
|
32
|
Vang JY, Her C, Krishnan VV. NMR based real-time enzyme kinetics on estimating the inhibitory effect of sucralose in the enzymatic conversion of sucrose. Biophys Chem 2020; 268:106495. [PMID: 33171432 DOI: 10.1016/j.bpc.2020.106495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/08/2020] [Accepted: 10/22/2020] [Indexed: 10/23/2022]
Abstract
Sucralose, one of the popular non-caloric artificial sweeteners, has been known to influence the enzymatic conversion of sucrose to glucose and fructose by invertase. In continuing the use of real-time NMR experiments and reaction progress curve analysis to measure enzyme kinetics, here we investigate the role of sucralose as an inhibitor. NMR based kinetic experiments were performed as a function of the substrate concentration for a range of sucralose concentrations, and the results were analyzed by fitting the progress curve to the Lambert-W function. The Michaelis-Menten parameters were then used to estimate the inhibitory constant of sucralose. To estimate the extent of sucralose inhibition on the enzymatic production of glucose, control experiments were performed with lactose as the inhibitor under similar experimental conditions. The results show that sucralose is a much more potent inhibitor than lactose, inhibiting the enzymatic conversion at least seven times more.
Collapse
Affiliation(s)
- Justin Y Vang
- Department of Chemistry, California State University, Fresno, CA 93740, United States of America
| | - Cheenou Her
- Department of Chemistry, California State University, Fresno, CA 93740, United States of America
| | - V V Krishnan
- Department of Chemistry, California State University, Fresno, CA 93740, United States of America; Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, CA 95616, United States of America.
| |
Collapse
|
33
|
Miners JO, Rowland A, Novak JJ, Lapham K, Goosen TC. Evidence-based strategies for the characterisation of human drug and chemical glucuronidation in vitro and UDP-glucuronosyltransferase reaction phenotyping. Pharmacol Ther 2020; 218:107689. [PMID: 32980440 DOI: 10.1016/j.pharmthera.2020.107689] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/26/2022]
Abstract
Enzymes of the UDP-glucuronosyltransferase (UGT) superfamily contribute to the elimination of drugs from almost all therapeutic classes. Awareness of the importance of glucuronidation as a drug clearance mechanism along with increased knowledge of the enzymology of drug and chemical metabolism has stimulated interest in the development and application of approaches for the characterisation of human drug glucuronidation in vitro, in particular reaction phenotyping (the fractional contribution of the individual UGT enzymes responsible for the glucuronidation of a given drug), assessment of metabolic stability, and UGT enzyme inhibition by drugs and other xenobiotics. In turn, this has permitted the implementation of in vitro - in vivo extrapolation approaches for the prediction of drug metabolic clearance, intestinal availability, and drug-drug interaction liability, all of which are of considerable importance in pre-clinical drug development. Indeed, regulatory agencies (FDA and EMA) require UGT reaction phenotyping for new chemical entities if glucuronidation accounts for ≥25% of total metabolism. In vitro studies are most commonly performed with recombinant UGT enzymes and human liver microsomes (HLM) as the enzyme sources. Despite the widespread use of in vitro approaches for the characterisation of drug and chemical glucuronidation by HLM and recombinant enzymes, evidence-based guidelines relating to experimental approaches are lacking. Here we present evidence-based strategies for the characterisation of drug and chemical glucuronidation in vitro, and for UGT reaction phenotyping. We anticipate that the strategies will inform practice, encourage development of standardised experimental procedures where feasible, and guide ongoing research in the field.
Collapse
Affiliation(s)
- John O Miners
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide, Australia.
| | - Andrew Rowland
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | | | | | | |
Collapse
|
34
|
Guo X, Malcolm JR, Ali MM, Ribeiro Morais G, Shnyder SD, Loadman PM, Patterson LH, Falconer RA. An efficient assay for identification and quantitative evaluation of potential polysialyltransferase inhibitors. Analyst 2020; 145:4512-4521. [PMID: 32412559 DOI: 10.1039/d0an00721h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The polysialyltransferases (polySTs) catalyse the polymerisation of polysialic acid, which plays an important role in tumour metastasis. While assays are available to assess polyST enzyme activity, there is no methodology available specifically optimised for identification and quantitative evaluation of potential polyST inhibitors. The development of an HPLC-fluorescence-based enzyme assay described within includes a comprehensive investigation of assay conditions, including evaluation of metal ion composition, enzyme, substrate and acceptor concentrations, temperature, pH, and tolerance to DMSO, followed by validation using known polyST inhibitors. Thorough analysis of each of the assay components provided a set of optimised conditions. Under these optimised conditions, the experimentally observed Ki value for CMP, a competitive polyST inhibitor, was strongly correlated with the predicted Ki value, based on the classical Cheng-Prusoff equation [average fold error (AFE) = 1.043]. These results indicate that this assay can provide medium-throughput analysis for enzyme inhibitors with high accuracy, through determining the corresponding IC50 values with substrate concentration at the KM, without the need to perform extensive kinetic studies for each compound. In conclusion, an in vitro cell-free assay for accurate assessment of polyST inhibition is described. The utility of the assay for routine identification of potential polyST inhibitors is demonstrated, allowing quantitative measurement of inhibition to be achieved, and exemplified through assessment of full competitive inhibition. Given the considerable and growing interest in the polySTs as important anti-metastatic targets in cancer drug discovery, this is a vital tool to enable preclinical identification and evaluation of novel polyST inhibitors.
Collapse
Affiliation(s)
- Xiaoxiao Guo
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Adiwidjaja J, Boddy AV, McLachlan AJ. Physiologically-Based Pharmacokinetic Predictions of the Effect of Curcumin on Metabolism of Imatinib and Bosutinib: In Vitro and In Vivo Disconnect. Pharm Res 2020; 37:128. [PMID: 32529309 DOI: 10.1007/s11095-020-02834-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 04/26/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE This study aimed to investigate the potential pharmacokinetic interactions between curcumin, imatinib and bosutinib, combining In Vitro and in silico methods. METHODS In Vitro metabolism of imatinib and bosutinib were investigated in pooled human liver microsomes and recombinant CYP3A4 enzyme in the presence and absence of curcumin and curcumin glucuronide using an LC-MS/MS assay for N-desmethyl metabolites. A physiologically-based pharmacokinetic (PBPK) model for curcumin formulated as solid lipid nanoparticles (SLN) was constructed using In Vitro glucuronidation kinetics and published clinical pharmacokinetic data. The potential effects of curcumin coadministration on systemic exposures of imatinib and bosutinib were predicted in silico using PBPK simulations. RESULTS Curcumin demonstrated potent reversible inhibition of cytochrome P450 (CYP)3A4-mediated N-demethylation of imatinib and bosutinib and CYP2C8-mediated metabolism of imatinib with inhibitory constants (ki,u) of ≤1.5 μmol. L-1. A confirmatory In Vitro study with paclitaxel, the 6α-hydroxylation of which is exclusively mediated by CYP2C8, was consistent with a potent inhibition of this enzyme by curcumin. Curcumin glucuronide also inhibited both CYP enzymes In Vitro, albeit to a lesser extent than that of curcumin. PBPK model simulations predicted that at recommended dosing regimens of SLN curcumin, coadministration would result in an increase in systemic exposures of imatinib and bosutinib of up to only 10%. CONCLUSION A PBPK model for curcumin in a SLN formulation was successfully developed. Although curcumin possesses a strong In Vitro inhibitory activity towards CYP3A4 and CYP2C8 enzymes, its interactions with imatinib and bosutinib were unlikely to be of clinical importance due to curcumin's poor bioavailability.
Collapse
Affiliation(s)
- Jeffry Adiwidjaja
- Sydney Pharmacy School, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Alan V Boddy
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5001, Australia
- University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA, 5000, Australia
| | - Andrew J McLachlan
- Sydney Pharmacy School, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
36
|
Truncation of Huia versabilis Bowman-Birk inhibitor increases its selectivity, matriptase-1 inhibitory activity and proteolytic stability. Biochimie 2020; 171-172:178-186. [PMID: 32169666 DOI: 10.1016/j.biochi.2020.03.006] [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: 09/30/2019] [Accepted: 03/04/2020] [Indexed: 01/14/2023]
Abstract
A gradual truncation of the primary structure of frog skin-derived Huia versabilis Bowman-Birk peptidic inhibitor (HV-BBI) resulted in 18-times stronger inhibitor of matriptase-1 (peptide 6, Ki = 8 nm) in comparison to the full-length HV-BBI (Ki = 155 nm). Analogous increase in the inhibitory activity in correlation with the peptide length reduction was not observed in case of other serine proteases, bovine trypsin (Ki = 151 nm for peptide 6 and Ki = 120 nm for HV-BBI) and plasmin (Ki = 120 nm for peptide 6 and 82 nm for HV-BBI). Weaker binding affinity to these enzymes emphasized an inhibitory specificity of peptide 6. Molecular dynamic analysis revealed that the observed variations in the binding affinity of peptide 6 and HV-BBI with matriptase-1 are associated with the entropic differences of the unbound peptides. Moreover, several aspects explaining differences in the inhibition of matriptase-1 by peptide 6 (bearing the C-terminal amide group) and its two analogues, peptide 6∗ (having the C-terminal carboxyl group, Ki = 473 nm) and cyclic peptide 6∗∗ (Ki = 533 nm), both exhibiting more than 50-fold reduced inhibitory potency, were discovered. It was also shown that peptide 6 presented significantly higher resistance to proteolytic degradation in human serum than HV-BBI. Additional investigations revealed that, in contrast to some amphibian-derived inhibitors, HV-BBI and its truncated analogues do not possess bactericidal activity, thus they cannot be considered as bifunctional agents.
Collapse
|
37
|
Biochemical characterisation of four rhamnosidases from thermophilic bacteria of the genera Thermotoga, Caldicellulosiruptor and Thermoclostridium. Sci Rep 2019; 9:15924. [PMID: 31685873 PMCID: PMC6828813 DOI: 10.1038/s41598-019-52251-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/23/2019] [Indexed: 01/19/2023] Open
Abstract
Carbohydrate active enzymes are classified in databases based on sequence and structural similarity. However, their function can vary considerably within a similarity-based enzyme family, which makes biochemical characterisation indispensable to unravel their physiological role and to arrive at a meaningful annotation of the corresponding genes. In this study, we biochemically characterised the four related enzymes Tm_Ram106B, Tn_Ram106B, Cb_Ram106B and Ts_Ram106B from the thermophilic bacteria Thermotoga maritima MSB8, Thermotoga neapolitana Z2706-MC24, Caldicellulosiruptor bescii DSM 6725 and Thermoclostridium stercorarium DSM 8532, respectively, as α-l-rhamnosidases. Cobalt, nickel, manganese and magnesium ions stimulated while EDTA and EGTA inhibited all four enzymes. The kinetic parameters such as Km, Vmax and kcat were about average compared to other rhamnosidases. The enzymes were inhibited by rhamnose, with half-maximal inhibitory concentrations (IC50) between 5 mM and 8 mM. The α-l-rhamnosidases removed the terminal rhamnose moiety from the rutinoside in naringin, a natural flavonone glycoside. The Thermotoga sp. enzymes displayed the highest optimum temperatures and thermostabilities of all rhamnosidases reported to date. The four thermophilic and divalent ion-dependent rhamnosidases are the first biochemically characterised orthologous enzymes recently assigned to glycoside hydrolase family 106.
Collapse
|
38
|
Evenseth LM, Warszycki D, Bojarski AJ, Gabrielsen M, Sylte I. In Silico Methods for the Discovery of Orthosteric GABA B Receptor Compounds. Molecules 2019; 24:E935. [PMID: 30866507 PMCID: PMC6429233 DOI: 10.3390/molecules24050935] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/20/2019] [Accepted: 03/01/2019] [Indexed: 12/27/2022] Open
Abstract
The GABAB receptor (GABAB-R) is a heterodimeric class C G protein-coupled receptor comprised of the GABAB1a/b and GABAB2 subunits. The endogenous orthosteric agonist γ-amino-butyric acid (GABA) binds within the extracellular Venus flytrap (VFT) domain of the GABAB1a/b subunit. The receptor is associated with numerous neurological and neuropsychiatric disorders including learning and memory deficits, depression and anxiety, addiction and epilepsy, and is an interesting target for new drug development. Ligand- and structure-based virtual screening (VS) are used to identify hits in preclinical drug discovery. In the present study, we have evaluated classical ligand-based in silico methods, fingerprinting and pharmacophore mapping and structure-based in silico methods, structure-based pharmacophores, docking and scoring, and linear interaction approximation (LIA) for their aptitude to identify orthosteric GABAB-R compounds. Our results show that the limited number of active compounds and their high structural similarity complicate the use of ligand-based methods. However, by combining ligand-based methods with different structure-based methods active compounds were identified in front of DUDE-E decoys and the number of false positives was reduced, indicating that novel orthosteric GABAB-R compounds may be identified by a combination of ligand-based and structure-based in silico methods.
Collapse
Affiliation(s)
- Linn M Evenseth
- Molecular Pharmacology and Toxicology, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, NO-9037 Tromsø, Norway.
| | - Dawid Warszycki
- Department of Medicinal Chemistry, Institute of Pharmacology, Polish Academy of Science, Smetna 12, 31-343 Kraków, Poland.
| | - Andrzej J Bojarski
- Department of Medicinal Chemistry, Institute of Pharmacology, Polish Academy of Science, Smetna 12, 31-343 Kraków, Poland.
| | - Mari Gabrielsen
- Molecular Pharmacology and Toxicology, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, NO-9037 Tromsø, Norway.
| | - Ingebrigt Sylte
- Molecular Pharmacology and Toxicology, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, NO-9037 Tromsø, Norway.
| |
Collapse
|
39
|
Kazmi F, Sensenhauser C, Greway T. Characterization of the In Vitro Inhibitory Potential of the Oligonucleotide Imetelstat on Human Cytochrome P450 Enzymes with Predictions of In Vivo Drug-Drug Interactions. Drug Metab Dispos 2019; 47:9-14. [PMID: 30389730 DOI: 10.1124/dmd.118.084103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022] Open
Abstract
Imetelstat, a 13-base oligonucleotide (5'-TAGGGTTAGACAA-3'), is a potent, investigational telomerase inhibitor in clinical development for the treatment of hematologic myeloid malignancies. Modifications to imetelstat oligonucleotide chemistry include an N3'-P5' thio-phosphoramidate backbone linkage to improve biologic stability and the addition of a palmitoyl tail at the 5'-position to enhance cellular membrane permeability. Other oligonucleotides have been previously shown to have in vitro test-system-dependent outcomes when potent cytochrome P450 inhibition in human liver microsomes (HLM) is observed, but such inhibition is not observed in cryopreserved human hepatocytes (CHH). Outcomes in CHH are consistent with clinical reports in which no interactions were reported. In the present study, imetelstat was evaluated for in vitro inhibition of eight P450 enzymes, namely CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4 in CHH (0.5 million cells/ml). Assays were performed using validated conditions, including short substrate times (10 minutes), and at the approximate substrate Km concentration. Imetelstat was found to have little to no inhibition of all P450 isoforms evaluated, with inhibitor concentration that causes 50% inhibition (IC50) values >100 μM. Maximum percent inhibition values for each P450 isoform at 100 μM imetelstat were <20% except for CYP2C8 activity, which was inhibited by 49%. Using a static mechanistic model, the predicted change in area under the curve of a victim drug coadministered with imetelstat was 1.04-fold, projecting no relevant clinical interaction. Overall, the results from this in vitro study suggest that clinical use of imetelstat is unlikely to affect the pharmacokinetics of concomitant therapies that undergo cytochrome P450-mediated metabolism.
Collapse
Affiliation(s)
- Faraz Kazmi
- Drug Metabolism and Pharmacokinetics, Janssen Research & Development, LLC, Spring House, Pennsylvania (F.K., C.S.) and Drug Metabolism and Pharmacokinetics, Janssen Research & Development, LLC., Raritan, New Jersey (T.G.)
| | - Carlo Sensenhauser
- Drug Metabolism and Pharmacokinetics, Janssen Research & Development, LLC, Spring House, Pennsylvania (F.K., C.S.) and Drug Metabolism and Pharmacokinetics, Janssen Research & Development, LLC., Raritan, New Jersey (T.G.)
| | - Tony Greway
- Drug Metabolism and Pharmacokinetics, Janssen Research & Development, LLC, Spring House, Pennsylvania (F.K., C.S.) and Drug Metabolism and Pharmacokinetics, Janssen Research & Development, LLC., Raritan, New Jersey (T.G.)
| |
Collapse
|
40
|
Tan BH, Ahemad N, Pan Y, Palanisamy UD, Othman I, Yiap BC, Ong CE. Cytochrome P450 2C9-natural antiarthritic interactions: Evaluation of inhibition magnitude and prediction from in vitro data. Biopharm Drug Dispos 2018; 39:205-217. [PMID: 29488228 DOI: 10.1002/bdd.2127] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/10/2018] [Accepted: 02/18/2018] [Indexed: 11/08/2022]
Abstract
Many dietary supplements are promoted to patients with osteoarthritis (OA) including the three naturally derived compounds, glucosamine, chondroitin and diacerein. Despite their wide spread use, research on interaction of these antiarthritic compounds with human hepatic cytochrome P450 (CYP) enzymes is limited. This study aimed to examine the modulatory effects of these compounds on CYP2C9, a major CYP isoform, using in vitro biochemical assay and in silico models. Utilizing valsartan hydroxylase assay as probe, all forms of glucosamine and chondroitin exhibited IC50 values beyond 1000 μM, indicating very weak potential in inhibiting CYP2C9. In silico docking postulated no interaction with CYP2C9 for chondroitin and weak bonding for glucosamine. On the other hand, diacerein exhibited mixed-type inhibition with IC50 value of 32.23 μM and Ki value of 30.80 μM, indicating moderately weak inhibition. Diacerein's main metabolite, rhein, demonstrated the same mode of inhibition as diacerein but stronger potency, with IC50 of 6.08 μM and Ki of 1.16 μM. The docking of both compounds acquired lower CDOCKER interaction energy values, with interactions dominated by hydrogen and hydrophobic bondings. The ranking with respect to inhibition potency for the investigated compounds was generally the same in both in vitro enzyme assay and in silico modeling with order of potency being diacerein/rhein > various glucosamine/chondroitin forms. In vitro-in vivo extrapolation of inhibition kinetics (using 1 + [I]/Ki ratio) demonstrated negligible potential of diacerein to cause interaction in vivo, whereas rhein was predicted to cause in vivo interaction, suggesting potential interaction risk with the CYP2C9 drug substrates.
Collapse
Affiliation(s)
- Boon Hooi Tan
- Division of Applied Biomedical Sciences and Biotechnology, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Yan Pan
- Department of Biomedical Science, University of Nottingham Malaysia Campus, Semenyih, Selangor, Malaysia
| | - Uma Devi Palanisamy
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Beow Chin Yiap
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Chin Eng Ong
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| |
Collapse
|
41
|
Ripp SL, Mukherjee A, Eng H, Stock T, Fleishaker D, Checchio T, Tammara B. In Vitro and In Vivo Investigation of Potential for Complex CYP3A Interaction for PF-00251802 (Dagrocorat), a Novel Dissociated Agonist of the Glucocorticoid Receptor. Clin Pharmacol Drug Dev 2017; 7:244-255. [PMID: 29112329 PMCID: PMC5900965 DOI: 10.1002/cpdd.411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 09/24/2017] [Indexed: 11/12/2022]
Abstract
The dissociated agonists of the glucocorticoid receptor are a novel class of agents in clinical development for rheumatoid arthritis. PF‐04171327 (fosdagrocorat) is a phosphate ester prodrug of PF‐00251802 (dagrocorat), a selective high‐affinity partial agonist of the glucocorticoid receptor, which is further metabolized to PF‐04015475. This study evaluated the cytochrome P450 (CYP)–mediated drug–drug interaction (DDI) potential of PF‐00251802 and PF‐04015475 in vitro and used model‐based prediction approaches to estimate clinical impact. PF‐00251802 is a reversible inhibitor of several CYPs, but modeling has suggested no clinically relevant interaction. PF‐00251802 and PF‐04015475 are time‐dependent inhibitors and inducers of CYP3A in vitro; PF‐00251802 is also a time‐dependent inhibitor of CYP2D6. Model‐based prediction suggested the potential for weak inhibition of CYP3A in vivo. A clinical DDI study was conducted with midazolam, a sensitive CYP3A substrate. A phase 1 open‐label, multiple‐dose study evaluated the effect of PF‐04171327 on midazolam pharmacokinetics and safety in 12 healthy volunteers. Administration of midazolam alone or concomitantly with PF‐04171327 resulted in equivalent pharmacokinetic profiles (AUCinf, 21.17 vs 20.28 ng·h/mL, respectively), indicating that PF‐04171327 had no net effect on CYP3A activity in vivo. These findings support the further development of PF‐00251802 and PF‐04171327 as potential treatments for patients with rheumatoid arthritis (NCT00987038).
Collapse
|
42
|
Spaggiari D, Daali Y, Rudaz S. An extensive cocktail approach for rapid risk assessment of in vitro CYP450 direct reversible inhibition by xenobiotic exposure. Toxicol Appl Pharmacol 2016; 302:41-51. [DOI: 10.1016/j.taap.2016.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 04/15/2016] [Accepted: 04/16/2016] [Indexed: 11/25/2022]
|
43
|
Development of an Excel-based laboratory information management system for improving workflow efficiencies in early ADME screening. Bioanalysis 2015; 8:99-110. [PMID: 26653172 DOI: 10.4155/bio.15.232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND There is a clear requirement for enhancing laboratory information management during early absorption, distribution, metabolism and excretion (ADME) screening. The application of a commercial laboratory information management system (LIMS) is limited by complexity, insufficient flexibility, high costs and extended timelines. RESULTS An improved custom in-house LIMS for ADME screening was developed using Excel. All Excel templates were generated through macros and formulae, and information flow was streamlined as much as possible. This system has been successfully applied in task generation, process control and data management, with a reduction in both labor time and human error rates. CONCLUSION An Excel-based LIMS can provide a simple, flexible and cost/time-saving solution for improving workflow efficiencies in early ADME screening.
Collapse
|