1
|
Conflitti P, Lyman E, Sansom MSP, Hildebrand PW, Gutiérrez-de-Terán H, Carloni P, Ansell TB, Yuan S, Barth P, Robinson AS, Tate CG, Gloriam D, Grzesiek S, Eddy MT, Prosser S, Limongelli V. Functional dynamics of G protein-coupled receptors reveal new routes for drug discovery. Nat Rev Drug Discov 2025:10.1038/s41573-024-01083-3. [PMID: 39747671 DOI: 10.1038/s41573-024-01083-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 01/04/2025]
Abstract
G protein-coupled receptors (GPCRs) are the largest human membrane protein family that transduce extracellular signals into cellular responses. They are major pharmacological targets, with approximately 26% of marketed drugs targeting GPCRs, primarily at their orthosteric binding site. Despite their prominence, predicting the pharmacological effects of novel GPCR-targeting drugs remains challenging due to the complex functional dynamics of these receptors. Recent advances in X-ray crystallography, cryo-electron microscopy, spectroscopic techniques and molecular simulations have enhanced our understanding of receptor conformational dynamics and ligand interactions with GPCRs. These developments have revealed novel ligand-binding modes, mechanisms of action and druggable pockets. In this Review, we highlight such aspects for recently discovered small-molecule drugs and drug candidates targeting GPCRs, focusing on three categories: allosteric modulators, biased ligands, and bivalent and bitopic compounds. Although studies so far have largely been retrospective, integrating structural data on ligand-induced receptor functional dynamics into the drug discovery pipeline has the potential to guide the identification of drug candidates with specific abilities to modulate GPCR interactions with intracellular effector proteins such as G proteins and β-arrestins, enabling more tailored selectivity and efficacy profiles.
Collapse
Affiliation(s)
- Paolo Conflitti
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
| | - Edward Lyman
- Department of Physics and Astronomy, University of Delaware, Newark, DE, USA
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Peter W Hildebrand
- Institute of Medical Physics and Biophysics, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Hugo Gutiérrez-de-Terán
- Department of Cell and Molecular Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Paolo Carloni
- INM-9/IAS-5 Computational Biomedicine, Forschungszentrum Jülich, Jülich, Germany
- Department of Physics, RWTH Aachen University, Aachen, Germany
| | - T Bertie Ansell
- Department of Biochemistry, University of Oxford, Oxford, UK
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Shuguang Yuan
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Patrick Barth
- Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Anne S Robinson
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - David Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen, Denmark
| | - Stephan Grzesiek
- Focal Area Structural Biology and Biophysics, Biozentrum, University of Basel, Basel, Switzerland
| | - Matthew T Eddy
- Department of Chemistry, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA
| | - Scott Prosser
- Department of Chemistry, University of Toronto, Mississauga, Ontario, Canada
| | - Vittorio Limongelli
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland.
| |
Collapse
|
2
|
Ye PP, Yao BF, Yang Y, Yang XM, Li Q, Song LL, Chen KG, Zhou HY, Shi JY, Zhang YH, Zhao FR, Guo ZJ, Xu SS, Chen J, Goh AH, Zhu SW, Zheng Y, Zhao W. Drug-drug interactions of simnotrelvir/ritonavir: an open-label, fixed-sequence, two-period clinical trial. Clin Microbiol Infect 2025; 31:101-107. [PMID: 39299559 DOI: 10.1016/j.cmi.2024.09.007] [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: 07/14/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVES Simnotrelvir is a small-molecule highly specific 3C-like protease inhibitor for anti-SARS-CoV-2 and was approved as a combination drug with ritonavir (simnotrelvir/ritonavir) in China. Simnotrelvir is a substrate of cytochrome P450 3A (CYP3A) and P-glycoprotein (P-gp), and a weak inhibitor of CYP3A. Ritonavir is a substrate and inhibitor of CYP3A and an inhibitor of P-gp. Hence, the drug-drug interaction potential of simnotrelvir/ritonavir should be investigated. METHODS This drug-drug interaction study was an open-label, fixed-sequence, two-period phase I clinical trial in Chinese healthy adult subjects, divided into three cohorts, including simnotrelvir/ritonavir co-administrated with a strong CYP3A and P-gp inhibitor (itraconazole) and inducer (rifampicin), and with a specific CYP3A substrate (midazolam). RESULTS The results demonstrated that compared with administration of simnotrelvir/ritonavir alone, the co-administration with itraconazole increased the geometric least-square mean ratio (GMR) of the expose (area under the plasma concentration-time curve from time zero to the lowest detectable plasma concentration [AUC0-t]) of simnotrelvir by 25% (GMR 125%, 90% CI 114-137%), whereas co-administration with rifampicin significantly decreased the AUC0-t of simnotrelvir by 81.5% (GMR 18.5%, 90% CI 16.4-20.9%). Notably, simnotrelvir/ritonavir increased the AUC0-t of midazolam by 16.69-fold (GMR 1769%, 90% CI 1551-2018%). The co-administration of simnotrelvir/ritonavir and rifampicin caused the increased amount and severity of treatment-emergent adverse events, especially hepatotoxicity. DISCUSSION The co-administration of simnotrelvir/ritonavir with CYP3A and P-gp inhibitors can be safely used, whereas the co-administration with CYP3A and P-gp strong inducer should be avoided to minimize the risk of under-exposure. Co-administration of midazolam with simnotrelvir/ritonavir increased systemic exposure of midazolam. CLINICALTRIALS gov Identifier: NCT05665647.
Collapse
Affiliation(s)
- Pan-Pan Ye
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Bu-Fan Yao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yang Yang
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China; Simcere Zaiming Pharmaceutical Co. Ltd., Nanjing, China
| | - Xin-Mei Yang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Qian Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Lin-Lin Song
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Ke-Guang Chen
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Hai-Yan Zhou
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Jin-Yi Shi
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Ye-Hui Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Fu-Rong Zhao
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Zi-Jia Guo
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China; Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing, China
| | - Shan-Sen Xu
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China; Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing, China
| | - Jia Chen
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China; Simcere Zaiming Pharmaceutical Co. Ltd., Nanjing, China
| | - Aik Han Goh
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China; Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing, China
| | - Shun-Wei Zhu
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China; Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing, China
| | - Yi Zheng
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhao
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China; Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
3
|
Alemayehu TT, Wassie YA, Bekalu AF, Tegegne AA, Ayenew W, Tadesse G, Getachew D, Yazie AS, Teketelew BB, Mekete MD, Fentahun S, Abebe TB, Minwagaw T, Geremew GW. Prevalence of potential drug‒drug interactions and associated factors among elderly patients in Ethiopia: a systematic review and meta-analysis. Glob Health Res Policy 2024; 9:46. [PMID: 39533381 PMCID: PMC11559191 DOI: 10.1186/s41256-024-00386-7] [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: 08/12/2024] [Accepted: 09/26/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The occurrence of potential drug‒drug interactions (pDDIs) is a serious global issue that affects all age groups, with the elderly population being the most vulnerable. This is due to their relatively high rates of comorbidity and polypharmacy, as well as physiological changes that can increase the potential for DDIs and the likelihood of adverse drug reactions. The aim of this study was to estimate the prevalence of pDDIs and associated factors among elderly patients in Ethiopia. METHODS A comprehensive literature search using the preferred reporting items for systematic review and meta-analysis statement was conducted on HINARI, Science Direct, Embase, PubMed/MEDLINE, Google Scholar, and Research Gate. Data were extracted via a Microsoft Excel spreadsheet and analyzed via STATA version 11.0. Egger regression tests and funnel plot analysis were used to check publication bias, and the I2 statistic was used to evaluate statistical heterogeneity. Sensitivity and subgroup analyses were also conducted to identify potential causes of heterogeneity. RESULTS Seven articles were analyzed, and a total of 1897 pDDIs were identified in 970 patients, resulting in an average of 1.97 DDIs per patient. The number of DDIs per patient ranged from 0.18 to 5.86. The overall prevalence of pDDIs among elderly patients was 50.69% (95% CI 18.77-82.63%). However, the prevalence of pDDIs ranged widely from 2.80 to 90.1%. When the severity of the interactions was considered, the prevalence of potential DDIs was found to be 28.74%, 70.68%, and 34.20% for major, moderate, and minor pDDIs, respectively. Polypharmacy and long hospital stays were identified as factors associated with pDDIs among elderly patients in Ethiopia. CONCLUSIONS The overall prevalence of pDDIs among elderly patients was high, with a wide range of prevalence rates. Moderate-severity interactions were the most prevalent. Polypharmacy and long hospital stays were identified as factors associated with pDDIs among elderly patients. The study suggests that DDIs identification database itself could have modified the DDIs prevalence rate. As a result, a single DDIs identification database needs to be authorized; otherwise, clinical knowledge should be taken into account when interpreting the information obtained.
Collapse
Affiliation(s)
- Tekletsadik Tekleslassie Alemayehu
- Department of Social and Administrative Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia.
| | - Yilkal Abebaw Wassie
- Department of Medical Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Abaynesh Fentahun Bekalu
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Addisu Afrassa Tegegne
- Department of Pharmaceutical Chemistry, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Wondim Ayenew
- Department of Social and Administrative Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Gebresilassie Tadesse
- Department of Psychiatry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Demis Getachew
- Department of Pharmacology, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Abebaw Setegn Yazie
- Department of Medical Parasitology, School of Biomedical and Laboratory Sciences College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Bisrat Birke Teketelew
- Department of Hematology and Immune Hematology, School of Laboratory, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Mekonnen Derese Mekete
- Department of Social and Administrative Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
- Department of Pharmacy, Debremarkos University, Debremarkos, Ethiopia
| | - Setegn Fentahun
- Department of Psychiatry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tesfaye Birhanu Abebe
- Department of Internal Medicine, School of Medicines College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tefera Minwagaw
- Department of Social and Administrative Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
- Department of Pharmacy, Bahir Dar University, Bahir Dar, Ethiopia
| | - Gebremariam Wulie Geremew
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
4
|
Kim DH, Hong JH, Jung WT, Nam KY, Roh JS, Lee HJ, Moon J, Kim KY, Jung JG, Sunwoo J. Pharmacokinetic Drug-Drug Interaction between Cilostazol and Rosuvastatin in Healthy Participants. Am J Cardiovasc Drugs 2024:10.1007/s40256-024-00686-w. [PMID: 39487337 DOI: 10.1007/s40256-024-00686-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND AND OBJECTIVES Cilostazol improves ischemic symptoms and prevents recurrence following cerebral infarction, and rosuvastatin reduces cholesterol levels. However, no reports exist on the pharmacokinetic interactions between these two drugs in healthy adults. This study evaluated the pharmacokinetic (PK) interactions and safety of cilostazol and rosuvastatin when co-administered to healthy male participants. METHODS A randomized, open-label, multiple-dosing, two-arm, two-period study was conducted. Arm A had 30 participants receiving 200 mg cilostazol daily and arm B had 27 participants receiving 20 mg rosuvastatin daily for 7 days. In period 2, both arms received a combination of 200 mg cilostazol and 20 mg rosuvastatin daily for 7 days following a 7-day washout period. Plasma concentrations of cilostazol, its metabolites, and rosuvastatin were quantified using liquid chromatography-tandem mass spectrometry. RESULTS Fifty-seven participants were randomized, and 44 completed the study. The geometric mean ratio (GMR) and 90% confidence intervals (CI) for maximum plasma concentration at steady state (Cmax,ss) and area under the plasma concentration-time curve during the dosing interval at steady state (AUCtau,ss) indicated no significant interaction between cilostazol and rosuvastatin. Safety assessments showed comparable profiles to individual drug administration, with no significant adverse events. CONCLUSION The repeated co-administration of cilostazol and rosuvastatin in healthy male participants resulted in minor PK interactions and exhibited a safety and tolerability profile similar to those of the individual drugs. This suggested that the combined regimen is well tolerated and does not necessitate dose adjustments. REGISTRATION ClinicalTrials.Gov identifier no. NCT06568133.
Collapse
Affiliation(s)
- Dong Ho Kim
- Clinical Trials Center, Chungnam National University Hospital, 266 Munhwa-ro, Jung-gu, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Jang Hee Hong
- Clinical Trials Center, Chungnam National University Hospital, 266 Munhwa-ro, Jung-gu, Daejeon, Republic of Korea
- Department of Pharmacology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Won Tae Jung
- Korea United Pharmaceutical Corporation, Seoul, Republic of Korea
| | - Kyu-Yeol Nam
- Korea United Pharmaceutical Corporation, Seoul, Republic of Korea
| | - Jae Seok Roh
- Korea United Pharmaceutical Corporation, Seoul, Republic of Korea
| | | | - JungHa Moon
- Caleb Multilab Inc., Seoul, Republic of Korea
| | | | - Jin-Gyu Jung
- Department of Family Medicine, Chungnam National University Hospital, 266 Munhwa-ro, Jung-gu, Daejeon, Republic of Korea.
| | - Jung Sunwoo
- Clinical Trials Center, Chungnam National University Hospital, 266 Munhwa-ro, Jung-gu, Daejeon, Republic of Korea.
| |
Collapse
|
5
|
de Almada-Vilhena AO, dos Santos OVM, Machado MDA, Nagamachi CY, Pieczarka JC. Prospecting Pharmacologically Active Biocompounds from the Amazon Rainforest: In Vitro Approaches, Mechanisms of Action Based on Chemical Structure, and Perspectives on Human Therapeutic Use. Pharmaceuticals (Basel) 2024; 17:1449. [PMID: 39598361 PMCID: PMC11597570 DOI: 10.3390/ph17111449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
The Amazon rainforest is an important reservoir of biodiversity, offering vast potential for the discovery of new bioactive compounds from plants. In vitro studies allow for the investigation of biological processes and interventions in a controlled manner, making them fundamental for pharmacological and biotechnological research. These approaches are faster and less costly than in vivo studies, providing standardized conditions that enhance the reproducibility and precision of data. However, in vitro methods have limitations, including the inability to fully replicate the complexity of a living organism and the absence of a complete physiological context. Translating results to in vivo models is not always straightforward, due to differences in pharmacokinetics and biological interactions. In this context, the aim of this literature review is to assess the advantages and disadvantages of in vitro approaches in the search for new drugs from the Amazon, identifying the challenges and limitations associated with these methods and comparing them with in vivo testing. Thus, bioprospecting in the Amazon involves evaluating plant extracts through bioassays to investigate pharmacological, antimicrobial, and anticancer activities. Phenolic compounds and terpenes are frequently identified as the main bioactive agents, exhibiting antioxidant, anti-inflammatory, and antineoplastic activities. Chemical characterization, molecular modifications, and the development of delivery systems, such as nanoparticles, are highlighted to improve therapeutic efficacy. Therefore, the Amazon rainforest offers great potential for the discovery of new drugs; however, significant challenges, such as the standardization of extraction methods and the need for in vivo studies and clinical trials, must be overcome for these compounds to become viable medications.
Collapse
Affiliation(s)
| | | | | | | | - Julio C. Pieczarka
- Center for Advanced Biodiversity Studies, Cell Culture Laboratory, Institute of Biological Sciences, Federal University of Pará/Guamá Science and Technology Park, Avenida Perimetral da Ciência Km 01—Guamá, Belém 66075-750, PA, Brazil; (A.O.d.A.-V.); (O.V.M.d.S.); (M.d.A.M.); (C.Y.N.)
| |
Collapse
|
6
|
Choi H, Jo EK, Kwon J, Kang D, Jeon J, Kho Y, Shin MY, Kim S. Identification of candidate exposure biomarkers for acetyl tributyl citrate and acetyl triethyl citrate using suspect screening in human liver microsomes. ENVIRONMENT INTERNATIONAL 2024; 192:108980. [PMID: 39307008 DOI: 10.1016/j.envint.2024.108980] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/02/2024] [Accepted: 08/22/2024] [Indexed: 10/26/2024]
Abstract
Acetyl tributyl citrate (ATBC) and acetyl triethyl citrate (ATEC) are increasingly used as alternatives to phthalates in various products, including food packaging, medical devices, and personal care items, raising concerns about their potential health impacts. This study aimed to investigate the in vitro human metabolism of ATBC and ATEC and identify potential exposure biomarkers applicable in human biomonitoring. Pooled human liver microsomes were utilized to conduct in vitro metabolism assays of deuterium labeled ATBC (ATBC-d3) and ATEC, and ultra performance liquid chromatography coupled to quadrupole-time-of-flight mass spectrometry (UPLC-qToF/MS) was employed for analysis. Suspect screening workflow and confidence level assignment were applied for metabolite identification. Time-course analysis revealed rapid metabolism of both compounds, with estimated apparent half-lives of approximately 5 min for ATBC-d3 and less than 15 min for ATEC. Eleven metabolites were identified for ATBC-d3 and six for ATEC. The predominant chemical reactions observed were carboxylic ester hydrolysis, deacetylation, and hydroxylation. Based on their abundance and specificity, MB1 (hydroxylated) and MB11 (hydrolyzed and hydroxylated) were proposed as candidate exposure biomarkers for ATBC, and ME1 (hydrolyzed and deacetylated) for ATEC. The identified metabolites and proposed sequences of kinetic process enhance our understanding of the fate of these compounds in the human body, potentially informing future toxicological assessments and guiding the development of more comprehensive human biomonitoring strategies.
Collapse
Affiliation(s)
- Hyojoo Choi
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Eu-Kyung Jo
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Jinhyun Kwon
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Daeho Kang
- Department of Environmental Engineering, Changwon National University, Gyeongsangnam-do, Republic of Korea
| | - Junho Jeon
- Department of Environmental Engineering, Changwon National University, Gyeongsangnam-do, Republic of Korea; School of Smart and Green Engineering, Changwon National University, Gyeongsangnam-do, Republic of Korea
| | - Younglim Kho
- Department of Health, Environment and Safety, Eulji University, Seongnam, Republic of Korea
| | - Mi-Yeon Shin
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea; Office of Dental Education, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Sungkyoon Kim
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea; Institute of Health and Environment, Graduate School of Public Health, Seoul National University, Seoul, South Korea.
| |
Collapse
|
7
|
Romaldini A, Spanò R, Veronesi M, Grimaldi B, Bandiera T, Sabella S. Human Multi-Lineage Liver Organoid Model Reveals Impairment of CYP3A4 Expression upon Repeated Exposure to Graphene Oxide. Cells 2024; 13:1542. [PMID: 39329726 PMCID: PMC11429598 DOI: 10.3390/cells13181542] [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: 07/08/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Three-dimensional hepatic cell cultures can provide an important advancement in the toxicity assessment of nanomaterials with respect to 2D models. Here, we describe liver organoids (LOs) obtained by assembling multiple cell lineages in a fixed ratio 1:1:0.2. These are upcyte® human hepatocytes, UHHs, upcyte® liver sinusoidal endothelial cells, LSECs, and human bone marrow-derived mesenchymal stromal cells, hbmMSCs. The structural and functional analyses indicated that LOs reached size stability upon ca. 10 days of cultivation (organoid maturation), showing a surface area of approximately 10 mm2 and the hepatic cellular lineages, UHHs and LSECs, arranged to form both primitive biliary networks and sinusoid structures, alike in vivo. LOs did not show signs of cellular apoptosis, senescence, or alteration of hepatocellular functions (e.g., dis-regulation of CYP3A4 or aberrant production of Albumin) for the entire culture period (19 days since organoid maturation). After that, LOs were repeatedly exposed for 19 days to a single or repeated dose of graphene oxide (GO: 2-40 µg/mL). We observed that the treatment did not induce any macroscopic signs of tissue damage, apoptosis activation, and alteration of cell viability. However, in the repeated dose regimen, we observed a down-regulation of CYP3A4 gene expression. Notably, these findings are in line with recent in vivo data, which report a similar impact on CYP3A4 when mice were repeatedly exposed to GO. Taken together, these findings warn of the potential detrimental effects of GO in real-life exposure (e.g., occupational scenario), where its progressive accumulation is likely expected. More in general, this study highlights that LOs formed by many cell lineages can enable repeated exposure regimens (suitable to mimic accumulation); thus, they can be suitably considered alternative or complementary in vitro systems to animal models.
Collapse
Affiliation(s)
- Alessio Romaldini
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Raffaele Spanò
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Marina Veronesi
- Structural Biophysics Facility, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
- D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Benedetto Grimaldi
- Molecular Medicine, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Tiziano Bandiera
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Stefania Sabella
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| |
Collapse
|
8
|
Pihlaja T, Kiiski I, Sikanen T. HLM chip - A microfluidic approach to study the mechanistic basis of cytochrome P450 inhibition using immobilized human liver microsomes. Eur J Pharm Sci 2024; 197:106773. [PMID: 38641124 DOI: 10.1016/j.ejps.2024.106773] [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: 01/10/2024] [Revised: 03/29/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Cytochrome P450 (CYP) system is a critical elimination route to most pharmaceuticals in human, but also prone to drug-drug interactions arising from the fact that concomitantly administered pharmaceuticals inhibit one another's CYP metabolism. The most severe form of CYP interactions is irreversible inhibition, which results in permanent inactivation of the critical CYP pathway and is only restored by de novo synthesis of new functional enzymes. In this study, we conceptualize a microfluidic approach to mechanistic CYP inhibition studies using human liver microsomes (HLMs) immobilized onto the walls of a polymer micropillar array. We evaluated the feasibility of these HLM chips for CYP inhibition studies by establishing the stability and the enzyme kinetics for a CYP2C9 model reaction under microfluidic flow and determining the half-maximal inhibitory concentrations (IC50) of three human CYP2C9 inhibitors (sulfaphenazole, tienilic acid, miconazole), including evaluation of their inhibition mechanisms and nonspecific microsomal binding on chip. Overall, the enzyme kinetics of CYP2C9 metabolism on the HLM chip (KM = 127 ± 55 µM) was shown to be similar to that of static HLM incubations (KM = 114 ± 14 µM) and the IC50 values toward CYP2C9 derived from the microfluidic assays (sulfaphenazole 0.38 ± 0.09 µM, tienilic acid 3.4 ± 0.6 µM, miconazole 0.54 ± 0.09 µM) correlated well with those determined using current standard IC50 shift assays. Most importantly, the HLM chip could distinguish between reversible (sulfaphenazole) and irreversible (tienilic acid) enzyme inhibitors in a single, automated experiment, indicating the great potential of the HLM chip to simplify current workflows used in mechanistic CYP inhibition studies. Furthermore, the results suggest that the HLM chip can also identify irreversible enzyme inhibitors, which are not necessarily resulting in a time-dependent inhibition (like suicide inhibitors), but whose inhibition mechanism is based on other kind of covalent or irreversible interaction with the CYP system. With our HLM chip approach, we could identify miconazole as such a compound that nonselectively inhibits the human CYP system with a prolonged, possibly irreversible impact in vitro, even if it is not a time-dependent inhibitor according to the IC50 shift assay.
Collapse
Affiliation(s)
- Tea Pihlaja
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Finland; Helsinki Institute of Sustainability Science, University of Helsinki, Finland
| | - Iiro Kiiski
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Finland
| | - Tiina Sikanen
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Finland; Helsinki Institute of Sustainability Science, University of Helsinki, Finland.
| |
Collapse
|
9
|
Geng K, Shen C, Wang X, Wang X, Shao W, Wang W, Chen T, Sun H, Xie H. A physiologically-based pharmacokinetic/pharmacodynamic modeling approach for drug-drug-gene interaction evaluation of S-warfarin with fluconazole. CPT Pharmacometrics Syst Pharmacol 2024; 13:853-869. [PMID: 38487942 PMCID: PMC11098157 DOI: 10.1002/psp4.13123] [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: 11/15/2023] [Revised: 01/17/2024] [Accepted: 02/07/2024] [Indexed: 05/18/2024] Open
Abstract
Warfarin is a widely used anticoagulant, and its S-enantiomer has higher potency compared to the R-enantiomer. S-warfarin is mainly metabolized by cytochrome P450 (CYP) 2C9, and its pharmacological target is vitamin K epoxide reductase complex subunit 1 (VKORC1). Both CYP2C9 and VKORC1 have genetic polymorphisms, leading to large variations in the pharmacokinetics (PKs) and pharmacodynamics (PDs) of warfarin in the population. This makes dosage management of warfarin difficult, especially in the case of drug-drug interactions (DDIs). This study provides a whole-body physiologically-based pharmacokinetic/PD (PBPK/PD) model of S-warfarin for predicting the effects of drug-drug-gene interactions on S-warfarin PKs and PDs. The PBPK/PD model of S-warfarin was developed in PK-Sim and MoBi. Drug-dependent parameters were obtained from the literature or optimized. Of the 34 S-warfarin plasma concentration-time profiles used, 96% predicted plasma concentrations within twofold range compared to observed data. For S-warfarin plasma concentration-time profiles with CYP2C9 genotype, 364 of 386 predicted plasma concentration values (~94%) fell within the twofold of the observed values. This model was tested in DDI predictions with fluconazole as CYP2C9 perpetrators, with all predicted DDI area under the plasma concentration-time curve to the last measurable timepoint (AUClast) ratio within twofold of the observed values. The anticoagulant effect of S-warfarin was described using an indirect response model, with all predicted international normalized ratio (INR) within twofold of the observed values. This model also incorporates a dose-adjustment method that can be used for dose adjustment and predict INR when warfarin is used in combination with CYP2C9 perpetrators.
Collapse
Affiliation(s)
- Kuo Geng
- Anhui Provincial Center for Drug Clinical EvaluationYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Wannan Medical CollegeWuhuAnhuiChina
| | - Chaozhuang Shen
- Department of Clinical Pharmacy and Pharmacy Administration, West China College of PharmacySichuan UniversityChengduSichuanChina
| | - Xiaohu Wang
- Department of PharmaceuticsChina Pharmaceutical UniversityNanjingChina
| | - Xingwen Wang
- Anhui Provincial Center for Drug Clinical EvaluationYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Wannan Medical CollegeWuhuAnhuiChina
| | - Wenxin Shao
- Anhui Provincial Center for Drug Clinical EvaluationYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Wannan Medical CollegeWuhuAnhuiChina
| | - Wenhui Wang
- Anhui Provincial Center for Drug Clinical EvaluationYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Wannan Medical CollegeWuhuAnhuiChina
| | - Tao Chen
- Anhui Provincial Center for Drug Clinical EvaluationYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
- Wannan Medical CollegeWuhuAnhuiChina
| | - Hua Sun
- Anhui Provincial Center for Drug Clinical EvaluationYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
| | - Haitang Xie
- Anhui Provincial Center for Drug Clinical EvaluationYijishan Hospital of Wannan Medical CollegeWuhuAnhuiChina
| |
Collapse
|
10
|
Wang Y, Zhou Q, Wang H, Song W, Wang J, Mamun AA, Geng P, Zhou Y, Wang S. Effect of P. corylifolia on the pharmacokinetic profile of tofacitinib and the underlying mechanism. Front Pharmacol 2024; 15:1351882. [PMID: 38650629 PMCID: PMC11033359 DOI: 10.3389/fphar.2024.1351882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/19/2024] [Indexed: 04/25/2024] Open
Abstract
This work aimed to explore the mechanisms underlying the interaction of the active furanocoumarins in P. corylifolia on tofacitinib both in vivo and in vitro. The concentration of tofacitinib and its metabolite M8 was determined using UPLC-MS/MS. The peak area ratio of M8 to tofacitinib was calculated to compare the inhibitory ability of furanocoumarin contained in the traditional Chinese medicine P. corylifolia in rat liver microsomes (RLMs), human liver microsomes (HLMs) and recombinant human CYP3A4 (rCYP3A4). We found that bergapten and isopsoralen exhibited more significant inhibitory activity in RLMs than other furanocoumarins. Bergapten and isopsoralen were selected to investigate tofacitinib drug interactions in vitro and in vivo. Thirty rats were randomly allocated into 5 groups (n = 6): control (0.5% CMC-Na), low-dose bergapten (20 mg/kg), high-dose bergapten (50 mg/kg), low-dose isopsoralen (20 mg/kg) and ketoconazole. 10 mg/kg of tofacitinib was orally intervented to each rat and the concentration level of tofacitinib in the rats were determined by UPLC-MS/MS. More imporrantly, the results showed that bergapten and isopsoralen significantly inhibited the metabolism of tofacitinib metabolism. The AUC(0-t), AUC(0-∞), MRT(0-t), MRT(0-∞) and Cmax of tofacitinib increased in varying degrees compared with the control group (all p < 0.05), but CLz/F decreased in varying degrees (p < 0.05) in the different dose bergapten group and isopsoralen group. Bergapten, isopsoralen and tofacitinib exhibit similar binding capacities with CYP3A4 by AutoDock 4.2 software, confirming that they compete for tofacitinib metabolism. P. corylifolia may considerably impact the metabolism of tofacitinib, which can provide essential information for the accurate therapeutic application of tofacitinib.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yunfang Zhou
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, Wenzhou Medical University Lishui Hospital, Lishui People’s Hospital, Lishui, Zhejiang, China
| | - Shuanghu Wang
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, Wenzhou Medical University Lishui Hospital, Lishui People’s Hospital, Lishui, Zhejiang, China
| |
Collapse
|
11
|
Van der Linden L, Vanassche T, Van Aelst L, Verhamme P. Comment on: Anti-Coagulant Treatment of Cancer-Associated Thrombosis in Frail Patients: Impact of Frailties on the Management of Drug-Drug Interactions. Clin Pharmacokinet 2024; 63:395-396. [PMID: 38236563 DOI: 10.1007/s40262-023-01344-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2023] [Indexed: 01/19/2024]
Affiliation(s)
- Lorenz Van der Linden
- Hospital Pharmacy Department, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| | - Thomas Vanassche
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Department of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Lucas Van Aelst
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Department of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Peter Verhamme
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Department of Cardiology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
12
|
Asfand-E-Yar M, Hashir Q, Shah AA, Malik HAM, Alourani A, Khalil W. Multimodal CNN-DDI: using multimodal CNN for drug to drug interaction associated events. Sci Rep 2024; 14:4076. [PMID: 38374325 PMCID: PMC10876630 DOI: 10.1038/s41598-024-54409-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/12/2024] [Indexed: 02/21/2024] Open
Abstract
Drug-to-drug interaction (DDIs) occurs when a patient consumes multiple drugs. Therefore, it is possible that any medication can influence other drugs' effectiveness. The drug-to-drug interactions are detected based on the interactions of chemical substructures, targets, pathways, and enzymes; therefore, machine learning (ML) and deep learning (DL) techniques are used to find the associated DDI events. The DL model, i.e., Convolutional Neural Network (CNN), is used to analyze the DDI. DDI is based on the 65 different drug-associated events, which is present in the drug bank database. Our model uses the inputs, which are chemical structures (i.e., smiles of drugs), enzymes, pathways, and the target of the drug. Therefore, for the multi-model CNN, we use several layers, activation functions, and features of drugs to achieve better accuracy as compared to traditional prediction algorithms. We perform different experiments on various hyperparameters. We have also carried out experiments on various iterations of drug features in different sets. Our Multi-Modal Convolutional Neural Network - Drug to Drug Interaction (MCNN-DDI) model achieved an accuracy of 90.00% and an AUPR of 94.78%. The results showed that a combination of the drug's features (i.e., chemical substructure, target, and enzyme) performs better in DDIs-associated events prediction than other features.
Collapse
Affiliation(s)
- Muhammad Asfand-E-Yar
- Department of Computer Science, CoE-AI, Center of Excellence Artificial Intelligence, Bahria University, Islamabad, Pakistan
| | - Qadeer Hashir
- Department of Computer Science, CoE-AI, Center of Excellence Artificial Intelligence, Bahria University, Islamabad, Pakistan
| | - Asghar Ali Shah
- Department of Computer Science, Bahria University, Islamabad , Pakistan
| | | | - Abdullah Alourani
- Department of Management Information Systems and Production Management, College of Business and Economics, Qassim University, Buraydah 51452, Saudi Arabia.
| | - Waqar Khalil
- Department of Computer Science, CoE-AI, Center of Excellence Artificial Intelligence, Bahria University, Islamabad, Pakistan
| |
Collapse
|
13
|
Yumoto Y, Endo T, Harada H, Kobayashi K, Nakabayashi T, Abe Y. High-throughput assay to simultaneously evaluate activation of CYP3A and the direct and time-dependent inhibition of CYP3A, CYP2C9, and CYP2D6 using liquid chromatography-tandem mass spectrometry. Xenobiotica 2024; 54:45-56. [PMID: 38265764 DOI: 10.1080/00498254.2024.2308818] [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: 10/25/2023] [Accepted: 01/18/2024] [Indexed: 01/25/2024]
Abstract
In the early stages of drug discovery, adequate evaluation of the potential drug-drug interactions (DDIs) of drug candidates is important. Several CYP3A activators are known to lead to underestimation of DDIs. These compounds affect midazolam 1'-hydroxylation but not midazolam 4-hydroxylation.We used both metabolic reactions of midazolam to evaluate the activation and inhibition of CYP3A activators simultaneously. For our CYP inhibition assay using cocktail probe substrates, simultaneous liquid chromatography-tandem mass spectrometry monitoring of 1'-hydroxymidazolam and 4-hydroxymidazolam for CYP3A was established in addition to monitoring of 4-hydroxydiclofenac and 1'-hydroxybufuralol for CYP2C9 and CYP2D6.The results of our cocktail inhibition assay were well correlated with those of a single inhibition assay, as were the estimated inhibition parameters for typical CYP3A inhibitors. In our assay, a proprietary compound that activated midazolam 1'-hydroxylation and tended to inhibit 4-hydroxylation was evaluated along with known CYP3A activators. All compounds were well characterised by comparison of the results of midazolam 1'- and 4-hydroxylation.In conclusion, our CYP cocktail inhibition assay can detect CYP3A activation and assess the direct and time-dependent inhibition potentials for CYP3A, CYP2C9, and CYP2D6. This method is expected to be very efficient in the early stages of drug discovery.
Collapse
Affiliation(s)
- Yu Yumoto
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Takuro Endo
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Hiroshi Harada
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Kaoru Kobayashi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Takeshi Nakabayashi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Yoshikazu Abe
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| |
Collapse
|
14
|
Lee J, Beers JL, Geffert RM, Jackson KD. A Review of CYP-Mediated Drug Interactions: Mechanisms and In Vitro Drug-Drug Interaction Assessment. Biomolecules 2024; 14:99. [PMID: 38254699 PMCID: PMC10813492 DOI: 10.3390/biom14010099] [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: 12/15/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Drug metabolism is a major determinant of drug concentrations in the body. Drug-drug interactions (DDIs) caused by the co-administration of multiple drugs can lead to alteration in the exposure of the victim drug, raising safety or effectiveness concerns. Assessment of the DDI potential starts with in vitro experiments to determine kinetic parameters and identify risks associated with the use of comedication that can inform future clinical studies. The diverse range of experimental models and techniques has significantly contributed to the examination of potential DDIs. Cytochrome P450 (CYP) enzymes are responsible for the biotransformation of many drugs on the market, making them frequently implicated in drug metabolism and DDIs. Consequently, there has been a growing focus on the assessment of DDI risk for CYPs. This review article provides mechanistic insights underlying CYP inhibition/induction and an overview of the in vitro assessment of CYP-mediated DDIs.
Collapse
Affiliation(s)
- Jonghwa Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.L.B.); (R.M.G.)
| | | | | | - Klarissa D. Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.L.B.); (R.M.G.)
| |
Collapse
|
15
|
Yamamiya I, Hunt A, Takenaka T, Sonnichsen D, Mina M, He Y, Benhadji KA, Gao L. Evaluation of the Cytochrome P450 3A and P-glycoprotein Drug-Drug Interaction Potential of Futibatinib. Clin Pharmacol Drug Dev 2023; 12:966-978. [PMID: 37132707 DOI: 10.1002/cpdd.1259] [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: 01/19/2023] [Accepted: 03/26/2023] [Indexed: 05/04/2023]
Abstract
Futibatinib, a selective, irreversible fibroblast growth factor receptor 1-4 inhibitor, is being investigated for tumors harboring FGFR aberrations and was recently approved for the treatment of FGFR2 fusion/rearrangement-positive intrahepatic cholangiocarcinoma. In vitro studies identified cytochrome P450 (CYP) 3A as the major CYP isoform in futibatinib metabolism and indicated that futibatinib is likely a P-glycoprotein (P-gp) substrate and inhibitor. Futibatinib also showed time-dependent inhibition of CYP3A in vitro. Phase I studies investigated the drug-drug interactions of futibatinib with itraconazole (a dual P-gp and strong CYP3A inhibitor), rifampin (a dual P-gp and strong CYP3A inducer), or midazolam (a sensitive CYP3A substrate) in healthy adult participants. Compared with futibatinib alone, coadministration of futibatinib with itraconazole increased futibatinib mean peak plasma concentration and area under the plasma concentration-time curve by 51% and 41%, respectively, and coadministration of futibatinib with rifampin lowered futibatinib mean peak plasma concentration and area under the plasma concentration-time curve by 53% and 64%, respectively. Coadministration of midazolam with futibatinib had no effect on midazolam pharmacokinetics compared with midazolam administered alone. These findings suggest that concomitant use of dual P-gp and strong CYP3A inhibitors/inducers with futibatinib should be avoided, but futibatinib can be concomitantly administered with other drugs metabolized by CYP3A. Drug-drug interaction studies with P-gp-specific substrates and inhibitors are planned.
Collapse
Affiliation(s)
| | | | - Toru Takenaka
- Taiho Pharmaceuticals Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Daryl Sonnichsen
- Sonnichsen Pharmaceutical Associates, LLC, Collegeville, Pennsylvania, USA
| | - Mark Mina
- Taiho Oncology, Inc., Princeton, New Jersey, USA
| | - Yaohua He
- Taiho Oncology, Inc., Princeton, New Jersey, USA
| | | | - Ling Gao
- Taiho Oncology, Inc., Princeton, New Jersey, USA
| |
Collapse
|
16
|
Oda FB, Carvalho FA, Yamamoto PA, de Oliveira JA, Peccinini RG, Zocolo GJ, Ribeiro PRV, de Moraes NV, Dos Santos AG. Metabolism Characterization and Chemical and Plasma Stability of Casearin B and Caseargrewiin F. PLANTA MEDICA 2023; 89:1097-1105. [PMID: 37084791 DOI: 10.1055/a-2078-5920] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Oral preparations of Casearia sylvestris (guacatonga) are used as antacid, analgesic, anti-inflammatory, and antiulcerogenic medicines. The clerodane diterpenes casearin B and caseargrewiin F are major active compounds in vitro and in vivo. The oral bioavailability and metabolism of casearin B and caseargrewiin F were not previously investigated. We aimed to assess the stability of casearin B and caseargrewiin F in physiological conditions and their metabolism in human liver microsomes. The compounds were identified by UHPLC-QTOF-MS/MS and quantified by validated LC-MS methods. The stability of casearin B and caseargrewiin F in physiological conditions was assessed in vitro. Both diterpenes showed a fast degradation (p < 0.05) in simulated gastric fluid. Their metabolism was not mediated by cytochrome P-450 enzymes, but the depletion was inhibited by the esterase inhibitor NaF. Both diterpenes and their dialdehydes showed a octanol/water partition coefficient in the range of 3.6 to 4.0, suggesting high permeability. Metabolism kinetic data were fitted to the Michaelis-Menten profile with KM values of 61.4 and 66.4 µM and Vmax values of 327 and 648 nmol/min/mg of protein for casearin B and caseargrewiin F, respectively. Metabolism parameters in human liver microsomes were extrapolated to predict human hepatic clearance, and suggest that caseargrewiin F and casearin B have a high hepatic extraction ratio. In conclusion, our data suggest that caseargrewiin F and casearin B present low oral bioavailability due to extensive gastric degradation and high hepatic extraction.
Collapse
Affiliation(s)
- Fernando Bombarda Oda
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, SP, Brazil
| | - Flávio Alexandre Carvalho
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, SP, Brazil
| | - Priscila Akemi Yamamoto
- Center of Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Palo (USP), Ribeirão Preto, SP, Brazil
| | - Jonata Augusto de Oliveira
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, SP, Brazil
| | - Rosângela Gonçalves Peccinini
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, SP, Brazil
| | - Guilherme Julião Zocolo
- Embrapa Agroindústria Tropical, Empresa Brasileira de Pesquisa Agropecuária (Embrapa), Fortaleza, CE, Brazil
| | | | - Natália Valadares de Moraes
- Center of Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - André Gonzaga Dos Santos
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, SP, Brazil
| |
Collapse
|
17
|
Gao N, Chen J, Li Y, Ding Y, Han Z, Xu H, Qiao H. The CYP2E1 inhibitor Q11 ameliorates LPS-induced sepsis in mice by suppressing oxidative stress and NLRP3 activation. Biochem Pharmacol 2023:115638. [PMID: 37290597 DOI: 10.1016/j.bcp.2023.115638] [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: 02/04/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/10/2023]
Abstract
Sepsis is an infection-induced, multi-organ system failure with a pathophysiology related to inflammation and oxidative stress. Increasing evidence indicates that cytochrome P450 2E1 (CYP2E1) is involved in the incidence and development of inflammatory diseases. However, a role for CYP2E1 in lipopolysaccharide (LPS)-induced sepsis has not been completely explored. Here we use Cyp2e1 knockout (cyp2e1-/-) mice to determine if CYP2E1 could be a therapeutic target for sepsis. We also evaluated the ability of Q11, a new specific CYP2E1 inhibitor, to prevent and ameliorate LPS-induced sepsis in mice and in LPS-treated J774A.1 and RAW264.7 cells. Cyp2e1 deletion significantly reduced hypothermia, multi-organ dysfunction and histological abnormalities in LPS-treated mice; consistent with this finding, the CYP2E1 inhibitor Q11 significantly prolonged the survival time of septic mice and ameliorated multi-organ injury induced by LPS. CYP2E1 activity in liver correlated with indicators of multi-organ injury, such as the level of lactate dehydrogenase (LDH) and blood urea nitrogen (BUN) (P<0.05). Q11 significantly suppressed the expression of NLRP3 in tissues after LPS injection; in vitro studies revealed that activation of NLRP3 signaling and increase of ROS was attenuated by Q11 in LPS-stimulated macrophages, which was reflected by reduced expression of caspase-1 and formation of ASC specks. Overall, our results indicate that Q11 improves the survival of mice with LPS-induced sepsis and attenuates sepsis-induced multiple-organ injury, suggesting that CYP2E1 could be a therapeutic target for sepsis.
Collapse
Affiliation(s)
- Na Gao
- Institute of Clinical Pharmacology, School of Medicine, Zhengzhou University, Zhengzhou, Henan, China.
| | - Jingjing Chen
- Institute of Clinical Pharmacology, School of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Yunchao Li
- Institute of Clinical Pharmacology, School of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Ding
- Institute of Clinical Pharmacology, School of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Zixinying Han
- Institute of Clinical Pharmacology, School of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Haiwei Xu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Hailing Qiao
- Institute of Clinical Pharmacology, School of Medicine, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
18
|
He RJ, Tian ZH, Huang J, Sun MR, Wei F, Li CY, Zeng HR, Zhang F, Guan XQ, Feng Y, Meng XM, Yang H, Ge GB. Rationally Engineered CYP3A4 Fluorogenic Substrates for Functional Imaging Analysis and Drug-Drug Interaction Studies. J Med Chem 2023; 66:6743-6755. [PMID: 37145039 DOI: 10.1021/acs.jmedchem.3c00101] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Cytochrome P450 3A4 (CYP3A4) is a key xenobiotic-metabolizing enzyme-mediated drug metabolism and drug-drug interaction (DDI). Herein, an effective strategy was used to rationally construct a practical two-photon fluorogenic substrate for hCYP3A4. Following two-round structure-based substrate discovery and optimization, we have successfully constructed a hCYP3A4 fluorogenic substrate (F8) with desirable features, including high binding affinity, rapid response, excellent isoform specificity, and low cytotoxicity. Under physiological conditions, F8 is readily metabolized by hCYP3A4 to form a brightly fluorescent product (4-OH F8) that can be easily detected by various fluorescence devices. The practicality of F8 for real-time sensing and functional imaging of hCYP3A4 has been examined in tissue preparations, living cells, and organ slices. F8 also demonstrates good performance for high-throughput screening of hCYP3A4 inhibitors and assessing DDI potentials in vivo. Collectively, this study develops an advanced molecular tool for sensing CYP3A4 activities in biological systems, which strongly facilitates CYP3A4-associated fundamental and applied research studies.
Collapse
Affiliation(s)
- Rong-Jing He
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhen-Hao Tian
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Jian Huang
- Pharmacology and Toxicology Division, Shanghai Institute of Food and Drug Control, Shanghai 201203, China
| | - Meng-Ru Sun
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Feng Wei
- School of Chemistry and Chemical Engineering & Center for Atomic Engineering of Advanced Materials & Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials, Anhui University, Hefei 230601, China
| | - Chun-Yu Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hai-Rong Zeng
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Feng Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao-Qing Guan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yan Feng
- School of Chemistry and Chemical Engineering & Center for Atomic Engineering of Advanced Materials & Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials, Anhui University, Hefei 230601, China
| | - Xiang-Ming Meng
- School of Chemistry and Chemical Engineering & Center for Atomic Engineering of Advanced Materials & Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials, Anhui University, Hefei 230601, China
- Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Guang-Bo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
19
|
Alshabi A, Shaik IH, Zhao Y, Pillai VC, Caritis S, Venkataramanan R. A cocktail probe approach to evaluate the effect of hormones on the expression and activity of CYP enzymes in human hepatocytes with conditions simulating late stage of pregnancy. Eur J Clin Pharmacol 2023; 79:815-827. [PMID: 37060457 PMCID: PMC10105140 DOI: 10.1007/s00228-023-03489-1] [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: 07/12/2022] [Accepted: 03/28/2023] [Indexed: 04/16/2023]
Abstract
PURPOSE Pregnancy-mediated physiological and biochemical changes contribute to alterations in the pharmacokinetics of certain drugs. There is a paucity of data on the systematic evaluation of the underlying mechanisms. The objective of the current study was to examine the impact of changes in circulating and tissue hormonal concentration during the late stage of pregnancy on the activity and expression of hepatic cytochrome P450 (CYP) enzymes using a cocktail probe approach. METHODS Freshly isolated primary human hepatocytes were incubated with third trimester physiologic (plasma) and projected liver (ten-fold higher) concentrations of female hormones: progesterone (2 µM), estradiol (0.3 µM), estriol (0.8 µM), estrone (0.2 µM), 17α-hydroxyprogesterone (0.1 µM), and human growth hormone (0.005 µM). The metabolic activity of the hepatocytes was assessed using a cocktail of isozyme-specific P450 probe substrates (CYP1A2 (phenacetin), CYP2C9 (diclofenac), CYP2C19 (S-mephenytoin), CYP2D6 (dextromethorphan), and CYP3A4 (testosterone)). A validated LC-MS/MS assay was used to measure the corresponding metabolite concentrations. CYP450 protein and mRNA levels were measured using western blot and qRT-PCR, respectively. RESULTS Female hormones at projected third-semester hepatic concentrations significantly enhanced mRNA and protein expression and increased the metabolic activity of CYP3A4. The expression and activity of other CYP450 enzymes studied were not affected by mixtures of female hormones at concentrations used. CONCLUSION The increased activity of CYP3A4 is consistent with the clinically observed increase in clearance of CYP3A4 substrates during pregnancy. Overall expression and activity of CYP450 isozymes are differentially regulated during pregnancy.
Collapse
Affiliation(s)
- Ali Alshabi
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, 15261, USA
- Clinical Pharmacy Department, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Imam H Shaik
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, 15261, USA
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, 15261, USA
| | - Yang Zhao
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, 15261, USA
| | - Venkateswaran C Pillai
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, 15261, USA
| | - Steve Caritis
- Department of Obstetrics and Gynecology, UPMC Magee Women's Hospital, Pittsburgh, PA, 15213, USA
| | - Raman Venkataramanan
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
20
|
Winters BR, Clapp PW, Simmons SO, Kochar TK, Jaspers I, Madden MC. E-Cigarette Liquids and Aldehyde Flavoring Agents Inhibit CYP2A6 Activity in Lung Epithelial Cells. ACS OMEGA 2023; 8:11261-11266. [PMID: 37008141 PMCID: PMC10061538 DOI: 10.1021/acsomega.2c08258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/28/2023] [Indexed: 06/19/2023]
Abstract
Certain e-liquids and aromatic aldehyde flavoring agents were previously identified as inhibitors of microsomal recombinant CYP2A6, the primary nicotine-metabolizing enzyme. However, due to their reactive nature, aldehydes may react with cellular components before reaching CYP2A6 in the endoplasmic reticulum. To determine whether e-liquid flavoring agents inhibited CYP2A6 in a cellular system, we investigated their effects on CYP2A6 using BEAS-2B cells transduced to overexpress CYP2A6. We demonstrated that two e-liquids and three aldehyde flavoring agents (cinnamaldehyde, benzaldehyde, and ethyl vanillin) exhibited dose-dependent inhibition of cellular CYP2A6.
Collapse
Affiliation(s)
- Brett R. Winters
- Curriculum
in Toxicology and Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Phillip W. Clapp
- Curriculum
in Toxicology and Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, United States
- Center
for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Steven O. Simmons
- Center
for Computational Toxicology and Exposure, ORD, US EPA, Research Triangle Park, Research Triangle Park, North Carolina 27711, United States
| | - Tavleen K. Kochar
- Department
of Chemistry, The University of North Carolina
at Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Ilona Jaspers
- Curriculum
in Toxicology and Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, United States
- Center
for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Michael C. Madden
- Formerly
Public Health and Integrative Toxicology Division, ORD, US EPA, Chapel Hill, North Carolina 27514, United States
| |
Collapse
|
21
|
Al-Enezi BF, Al-Hasawi N, Matar KM. Impact of valproic acid on busulfan pharmacokinetics: In vitro assessment of potential drug-drug interaction. PLoS One 2023; 18:e0280574. [PMID: 36696427 PMCID: PMC9876357 DOI: 10.1371/journal.pone.0280574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 01/03/2023] [Indexed: 01/26/2023] Open
Abstract
Busulfan (Bu) is an alkylating agent commonly used at high doses in the preparative regimens of hematopoietic stem cell transplantation (HSCT). It has been shown that such high doses of Bu are associated with generalized seizures which are usually managed by prophylactic antiepileptic drugs (AEDs) such as valproic acid (VPA). Being a strong enzyme inhibitor, VPA may inhibit Bu metabolism and thus increase its potential toxicity. Despite its clinical relevance, the potential interaction between Bu and VPA has not yet been evaluated. The aim of the present study was to assess and evaluate the potential drug-drug interaction (DDI) between Bu and VPA. This study was carried out by incubating Bu in laboratory-prepared rat liver-subcellular fractions including S9, microsomes, and cytosol, alone or in combination with VPA. The liver fractions were prepared by differential centrifugation of the liver homogenate. Analysis of Bu was employed using a fully validated LC-MS/MS method. The validation parameters were within the proposed limits of the international standards guidelines. Bu metabolic stability was assessed by incubating Bu at a concentration of 8 μg/ml in liver fractions at 37°C. There were significant reductions in Bu levels in S9 and cytosolic fractions, whereas these levels were not significantly (P ˃ 0.05) changed in microsomes. However, in presence of VPA, Bu levels in S9 fraction remained unchanged. These results indicated, for the first time, the potential metabolic interaction of Bu and VPA being in S9 only. This could be explained by inhibiting Bu cytosolic metabolism by the interaction with VPA either by sharing the same metabolic enzyme or the required co-factor. In conclusion, the present findings suggest, for the first time, a potential DDI between Bu and VPA in vitro using rat liver fractions. Further investigations are warranted in human-derived liver fractions to confirm such an interaction.
Collapse
Affiliation(s)
- Bashayer F. Al-Enezi
- Department of Pharmacology & Therapeutics, Faculty of Pharmacy, Kuwait University, Kuwait City, Kuwait
| | - Nada Al-Hasawi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kuwait University, Kuwait City, Kuwait
| | - Kamal M. Matar
- Department of Pharmacology & Therapeutics, Faculty of Pharmacy, Kuwait University, Kuwait City, Kuwait
- * E-mail: ,
| |
Collapse
|
22
|
Metabolic activation of drugs by cytochrome P450 enzymes: Biochemical insights into mechanism-based inactivation by fibroblast growth factor receptor inhibitors and chemical approaches to attenuate reactive metabolite formation. Biochem Pharmacol 2022; 206:115336. [DOI: 10.1016/j.bcp.2022.115336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
|
23
|
Ryu S, Woody N, Chang G, Mathialagan S, Varma MVS. Identification of Organic Anion Transporter 2 Inhibitors: Screening, Structure-Based Analysis, and Clinical Drug Interaction Risk Assessment. J Med Chem 2022; 65:14578-14588. [PMID: 36270005 DOI: 10.1021/acs.jmedchem.2c01079] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Organic anion transporter 2 (OAT2 or SLC22A7) plays an important role in the hepatic uptake and renal secretion of several endogenous compounds and drugs. The goal of this work is to understand the structure activity of OAT2 inhibition and assess clinical drug interaction risk. A single-point inhibition assay using OAT2-transfected HEK293 cells was employed to screen about 150 compounds; and concentration-dependent inhibition potency (IC50) was measured for the identified "inhibitors". Acids represented about 65% of all inhibitors, and the frequency of bases-plus-zwitterions approximately doubled for "non-inhibitors". Interestingly, 9 of 10 most potent inhibitors (low IC50) are acids (pKa ∼ 3-5). Additionally, inhibitors are significantly larger and lipophilic than non-inhibitors. In silico (binary) models were developed to identify inhibitors and non-inhibitors. Finally, in vivo risk assessed via static drug-drug interaction models identified several inhibitors with potential for renal and hepatic OAT2 inhibition at clinical doses. This is the first study assessing the global pattern of OAT2-ligand interactions.
Collapse
Affiliation(s)
- Sangwoo Ryu
- Medicine Design, Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Nathaniel Woody
- Medicine Design, Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - George Chang
- Medicine Design, Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Sumathy Mathialagan
- Medicine Design, Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Manthena V S Varma
- Medicine Design, Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
24
|
Topriceanu CC, Moon JC, Captur G, Perera B. The use of attention-deficit hyperactivity disorder medications in cardiac disease. Front Neurosci 2022; 16:1020961. [PMID: 36340760 PMCID: PMC9626759 DOI: 10.3389/fnins.2022.1020961] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/03/2022] [Indexed: 09/02/2023] Open
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a neurodevelopmental disorder with onset usually in childhood characterized by inattention, impulsivity, and hyperactivity causing a functional impairment. Untreated ADHD, or treatment delay is associated with adverse outcomes and poor quality of life. Although conservative management strategies such as behavioral and psychological interventions are important, pharmacological treatment has a strong evidence base with improved outcomes. ADHD medications are broadly divided into stimulant and non-stimulant medications. Stimulant medications are generally more effective than non-stimulants. Cardiovascular safety of ADHD medication has been a matter of debate for decades. Treatment guidelines advise the careful consideration of risks and benefits in people with cardiovascular diseases such as congenital heart disease or cardiomyopathy. Although stimulants can increase systemic blood pressure and heart rate, no significant associations were found between their use and serious cardiovascular events. Concerns regarding QT effects and attendant sudden cardiac death risks deter clinicians from initiating much-needed ADHD medications in patients with heart disease. This overly cautious approach is potentially depriving low-risk individuals from significant benefits associated with timely ADHD drug treatment. This review discusses the cardiovascular risks reportedly associated with ADHD medications, the evidence base for their safe usage in persons with established cardiovascular disease, and highlights future research directions.
Collapse
Affiliation(s)
- Constantin-Cristian Topriceanu
- Barnet, Enfield and Haringey Mental Health Trust, London, United Kingdom
- UCL Institute of Cardiovascular Science, University College London, London, United Kingdom
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, United Kingdom
- Cardiac MRI Unit, Barts Heart Centre, London, United Kingdom
- Centre for Inherited Cardiovascular Diseases, Great Ormond Street Hospital, London, United Kingdom
| | - James C. Moon
- UCL Institute of Cardiovascular Science, University College London, London, United Kingdom
- Cardiac MRI Unit, Barts Heart Centre, London, United Kingdom
| | - Gabriella Captur
- UCL Institute of Cardiovascular Science, University College London, London, United Kingdom
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, United Kingdom
- Department of Cardiology, Centre for Inherited Heart Muscle Conditions, The Royal Free Hospital, London, United Kingdom
| | - Bhathika Perera
- Barnet, Enfield and Haringey Mental Health Trust, London, United Kingdom
| |
Collapse
|
25
|
Atypical kinetics of cytochrome P450 enzymes in pharmacology and toxicology. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 95:131-176. [PMID: 35953154 DOI: 10.1016/bs.apha.2022.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Atypical kinetics are observed in metabolic reactions catalyzed by cytochrome P450 enzymes (P450). Yet, this phenomenon is regarded as experimental artifacts in some instances despite increasing evidence challenging the assumptions of typical Michaelis-Menten kinetics. As P450 play a major role in the metabolism of a wide range of substrates including drugs and endogenous compounds, it becomes critical to consider the impact of atypical kinetics on the accuracy of estimated kinetic and inhibitory parameters which could affect extrapolation of pharmacological and toxicological implications. The first half of this book chapter will focus on atypical non-Michaelis-Menten kinetics (e.g. substrate inhibition, biphasic and sigmoidal kinetics) as well as proposed underlying mechanisms supported by recent insights in mechanistic enzymology. In particular, substrate inhibition kinetics in P450 as well as concurrent drug inhibition of P450 in the presence of substrate inhibition will be further discussed. Moreover, mounting evidence has revealed that despite the high degree of sequence homology between CYP3A isoforms (i.e. CYP3A4 and CYP3A5), they have the propensities to exhibit vastly different susceptibilities and potencies of mechanism-based inactivation (MBI) with a common drug inhibitor. These experimental observations pertaining to the presence of these atypical isoform- and probe substrate-specific complexities in CYP3A isoforms by several clinically-relevant drugs will therefore be expounded and elaborated upon in the second half of this book chapter.
Collapse
|
26
|
Doran AC, Burchett W, Landers C, Gualtieri GM, Balesano A, Eng H, Dantonio AL, Goosen TC, Obach RS. Defining the Selectivity of Chemical Inhibitors Used for Cytochrome P450 Reaction Phenotyping: Overcoming Selectivity Limitations with a Six-Parameter Inhibition Curve-Fitting Approach. Drug Metab Dispos 2022; 50:DMD-AR-2022-000884. [PMID: 35777846 DOI: 10.1124/dmd.122.000884] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/22/2022] Open
Abstract
The utility of chemical inhibitors in cytochrome P450 (CYP) reaction phenotyping is highly dependent on their selectivity and potency for their target CYP isoforms. In the present study, seventeen inhibitors of CYP1A2, 2B6, 2C8, 2C9, 2C19, 2D6, and 3A4/5 commonly used in reaction phenotyping were evaluated for their cross-enzyme selectivity in pooled human liver microsomes. The data were evaluated using a statistical desirability analysis to identify (1) inhibitors of superior selectivity for reaction phenotyping and (2) optimal concentrations for each. Among the inhibitors evaluated, α-naphthoflavone, furafylline, sulfaphenazole, tienilic acid, N-benzylnirvanol, and quinidine were most selective, such that their respective target enzymes were inhibited by ~95% without inhibiting any other CYP enzyme by more than 10%. Other commonly employed inhibitors, such as ketoconazole and montelukast, among others, were of insufficient selectivity to yield a concentration that could adequately inhibit their target enzymes without affecting other CYP enzymes. To overcome these shortcomings, an experimental design was developed wherein dose response data from a densely sampled multi-concentration inhibition curve are analyzed by a six-parameter inhibition curve function, allowing accounting of the inhibition of off-target CYP isoforms inhibition and more reliable determination of maximum targeted enzyme inhibition. The approach was exemplified using rosiglitazone N-demethylation, catalyzed by both CYP2C8 and 3A4, and was able to discern the off-target inhibition by ketoconazole and montelukast from the inhibition of the targeted enzyme. This methodology yields more accurate estimates of CYP contributions in reaction phenotyping. Significance Statement Isoform-selective chemical inhibitors are important tools for identifying and quantifying enzyme contributions as part of a CYP reaction phenotyping assessment for projecting drug-drug interactions. However, currently employed practices fail to adequately compensate for shortcomings in inhibitor selectivity and the resulting confounding impact on estimates of the CYP enzyme contribution to drug clearance. In this report, we describe a detailed IC50 study design with 6-parameter modeling approach that yields more accurate estimates of enzyme contribution.
Collapse
Affiliation(s)
| | | | | | | | | | - Heather Eng
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Global Research and Development, United States
| | | | - Theunis C Goosen
- Pharmacokinetics, Dynamics & Metabolism, Pfizer, Inc, United States
| | | |
Collapse
|
27
|
Sechaud R, Sinclair K, Grosch K, Ouatas T, Pathak D. Evaluation of drug-drug interactions between midostaurin and strong CYP3A4 inhibitors in patients with FLT-3-mutated acute myeloid leukemia (AML). Cancer Chemother Pharmacol 2022; 90:19-27. [PMID: 35751657 DOI: 10.1007/s00280-022-04448-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 06/04/2022] [Indexed: 12/25/2022]
Abstract
PURPOSE Midostaurin, approved for the treatment of newly diagnosed, FLT3-mutated acute myeloid leukemia (AML), is metabolized by cytochrome P450 3A4 (CYP3A4). Midostaurin with concomitant strong CYP3A4 inhibitors use (e.g., antifungal azoles) may result in drug-drug interactions. This post hoc analysis of RATIFY phase 3 study data evaluated effects of strong CYP3A4 inhibitor use on the exposure and safety of midostaurin. METHODS Trough concentrations were used to assess midostaurin and metabolite exposure in the presence and absence of strong CYP3A4 inhibitors. Adverse event (AE) frequency was assessed in patients who received concomitant strong CYP3A4 inhibitors vs those who did not. Time to first clinically notable AE (CNAE) was also assessed in patients with high midostaurin plasma exposure vs those of matched placebo controls. RESULTS Use of concomitant strong CYP3A4 inhibitors was most frequent during the induction phase (60.8%). A 1.44-fold increase in midostaurin plasma exposure was observed in patients with concomitant strong CYP3A4 inhibitor use vs those without. Midostaurin-treated patients who received concomitant strong CYP3A4 inhibitors experienced grade 3/4 infection-related AEs more frequently vs those who did not. Patients with high levels of midostaurin exposure had a shorter median time to first grade 3/4 CNAE vs placebo controls (36 vs 41 days, respectively; P = .012). CONCLUSION Although concomitantly administered strong CYP3A4 inhibitors increased midostaurin exposure 1.44-fold, no clinically relevant differences in safety were noted. Midostaurin dose adjustment is not necessary with concomitant strong CYP3A4 inhibitors in patients with FLT3-mutated AML; however, caution is advised, and patients should be closely monitored.
Collapse
Affiliation(s)
| | | | - Kai Grosch
- Novartis Pharma AG, CH-4002, Basel, Switzerland
| | | | | |
Collapse
|
28
|
Martin P, Czerwiński M, Limaye PB, Muranjan S, Ogilvie BW, Smith S, Boyd B. In vitro evaluation of fenfluramine and norfenfluramine as victims of drug interactions. Pharmacol Res Perspect 2022; 10:e00958. [PMID: 35599345 PMCID: PMC9124820 DOI: 10.1002/prp2.958] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/06/2022] [Indexed: 12/27/2022] Open
Abstract
Fenfluramine (FFA) has potent antiseizure activity in severe, pharmacoresistant childhood‐onset developmental and epileptic encephalopathies (e.g., Dravet syndrome). To assess risk of drug interaction affecting pharmacokinetics of FFA and its major metabolite, norfenfluramine (nFFA), we conducted in vitro metabolite characterization, reaction phenotyping, and drug transporter−mediated cellular uptake studies. FFA showed low in vitro clearance in human liver S9 fractions and in intestinal S9 fractions in all three species tested (t1/2 > 120 min). Two metabolites (nFFA and an N‐oxide or a hydroxylamine) were detected in human liver microsomes versus six in dog and seven in rat liver microsomes; no metabolite was unique to humans. Selective CYP inhibitor studies showed FFA metabolism partially inhibited by quinidine (CYP2D6, 48%), phencyclidine (CYP2B6, 42%), and furafylline (CYP1A2, 32%) and, to a lesser extent (<15%), by tienilic acid (CYP2C9), esomeprazole (CYP2C19), and troleandomycin (CYP3A4/5). Incubation of nFFA with rCYP1A2, rCYP2B6, rCYP2C19, and rCYP2D6 resulted in 10%−20% metabolism and no clear inhibition of nFFA metabolism by any CYP‐selective inhibitor. Reaction phenotyping showed metabolism of FFA by recombinant human cytochrome P450 (rCYP) enzymes rCYP2B6 (10%–21% disappearance for 1 and 10 µM FFA, respectively), rCYP1A2 (22%−23%), rCYP2C19 (49%−50%), and rCYP2D6 (59%−97%). Neither FFA nor nFFA was a drug transporter substrate. Results show FFA metabolism to nFFA occurs through multiple pathways of elimination. FFA dose adjustments may be needed when administered with strong inhibitors or inducers of multiple enzymes involved in FFA metabolism (e.g., stiripentol).
Collapse
|
29
|
Tang LWT, Wu G, Chan ECY. Identification of Infigratinib as a Potent Reversible Inhibitor and Mechanism-Based Inactivator of CYP2J2: Nascent Evidence for a Potential In Vivo Metabolic Drug-Drug Interaction with Rivaroxaban. J Pharmacol Exp Ther 2022; 382:123-134. [PMID: 35640957 PMCID: PMC9639665 DOI: 10.1124/jpet.122.001222] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/04/2022] [Indexed: 11/22/2022] Open
Abstract
Infigratinib (INF) is a fibroblast growth factor receptor inhibitor that was recently FDA-approved for the treatment of advanced or metastatic cholangiocarcinoma. We previously established that INF inhibited and inactivated cytochrome P450 3A4 (CYP3A4). Here, in a follow-up to our previous study, we identified for the first time that INF also elicited potent competitive inhibition and mechanism-based inactivation (MBI) of CYP2J2 with kinetic parameters K i, K I, k inact, and partition ratio of 1.94 µM, 0.10 µM, 0.026 min-1 and ~3 respectively when rivaroxaban was harnessed as the probe substrate. Inactivation was revealed to exhibit cofactor-dependency and was attenuated by an alternative substrate (astemizole) and direct inhibitor (nilotinib) of CYP2J2. Additionally, the nature of inactivation was unlikely to be pseudo-irreversible and instead arose from covalent modification due to the lack of substantial enzyme activity recovery following dialysis and chemical oxidation as well as the lack of a resolvable Soret band in spectral scans. Glutathione trapping confirmed that the identity of the putative reactive intermediate implicated in the covalent inactivation of both CYP2J2 and CYP3A4 was identical and likely attributable to an electrophilic p-benzoquinonediimine intermediate of INF. Finally, mechanistic static modelling revealed that by integrating the previously arcane inhibition and inactivation kinetic parameters of CYP2J2-mediated rivaroxaban hydroxylation by INF illuminated in this work together with those previously documented for CYP3A4, a 49% increase in the systemic exposure of rivaroxaban was projected. Our modelling results predicted a potential risk of metabolic DDI between the clinically-relevant combination of rivaroxaban and INF in the setting of cancer. Significance Statement In this study, we reported that INF elicits potent reversible inhibition and MBI of CYP2J2. Furthermore, static modelling predicted that its coadministration with the direct oral anticoagulant rivaroxaban may potentially culminate in an metabolic DDI leading to an increased risk of major bleeding. As rivaroxaban is steadily gaining prominence as the anticoagulant of choice in the treatment of cancer-associated venous thromboembolism, the DDI projections reported here are clinically-relevant and warrants further investigation via physiologically-based pharmacokinetic modelling and simulation.
Collapse
Affiliation(s)
| | - Guoyi Wu
- National University of Singapore, Singapore
| | | |
Collapse
|
30
|
Romaldini A, Spanò R, Catalano F, Villa F, Poggi A, Sabella S. Sub-Lethal Concentrations of Graphene Oxide Trigger Acute-Phase Response and Impairment of Phase-I Xenobiotic Metabolism in Upcyte® Hepatocytes. Front Bioeng Biotechnol 2022; 10:867728. [PMID: 35662849 PMCID: PMC9161028 DOI: 10.3389/fbioe.2022.867728] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/27/2022] [Indexed: 11/21/2022] Open
Abstract
The impact of graphene oxide on hepatic functional cells represents a crucial evaluation step for its potential application in nanomedicine. Primary human hepatocytes are the gold standard for studying drug toxicity and metabolism; however, current technical limitations may slow down the large-scale diffusion of this cellular tool for in vitro investigations. To assess the potential hepatotoxicity of graphene oxide, we propose an alternative cell model, the second-generation upcyte® hepatocytes, which show metabolic and functional profiles akin to primary human hepatocytes. Cells were acutely exposed to sub-lethal concentrations of graphene oxide (≤80 μg/ml) for 24 h and stress-related cell responses (such as apoptosis, oxidative stress, and inflammatory response) were evaluated, along with a broad investigation of graphene oxide impact on specialized hepatic functions. Results show a mild activation of early apoptosis but not oxidative stress or inflammatory response in our cell model. Notably, while graphene oxide clearly impacted phase-I drug-metabolism enzymes (e.g., CYP3A4, CYP2C9) through the inhibition of gene expression and metabolic activity, conversely, no effect was observed for phase-II enzyme GST and phase-III efflux transporter ABCG2. The GO-induced impairment of CYP3A4 occurs concomitantly with the activation of an early acute-phase response, characterized by altered levels of gene expression and protein production of relevant acute-phase proteins (i.e., CRP, Albumin, TFR, TTR). These data suggest that graphene oxide induces an acute phase response, which is in line with recent in vivo findings. In conclusion, upcyte® hepatocytes appear a reliable in vitro model for assessing nanomaterial-induced hepatotoxicity, specifically showing that sub-lethal doses of graphene oxide have a negative impact on the specialized hepatic functions of these cells. The impairment of the cytochrome P450 system, along with the activation of an acute-phase response, may suggest potential detrimental consequences for human health, as altered detoxification from xenobiotics and drugs.
Collapse
Affiliation(s)
- A. Romaldini
- D3 PharmaChemistry, Istituto Italiano di Tecnologia, Genoa, Italy
| | - R. Spanò
- D3 PharmaChemistry, Istituto Italiano di Tecnologia, Genoa, Italy
| | - F. Catalano
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, Genoa, Italy
| | - F. Villa
- Unit of Molecular Oncology and Angiogenesis, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - A. Poggi
- Unit of Molecular Oncology and Angiogenesis, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - S. Sabella
- D3 PharmaChemistry, Istituto Italiano di Tecnologia, Genoa, Italy
- *Correspondence: S. Sabella,
| |
Collapse
|
31
|
Tang LWT, Fu J, Koh SK, Wu G, Zhou L, Chan ECY. Metabolic Activation of the Acrylamide Michael Acceptor Warhead in Futibatinib to an Epoxide Intermediate Engenders Covalent Inactivation of Cytochrome P450 3A. Drug Metab Dispos 2022; 50:931-941. [PMID: 35512804 DOI: 10.1124/dmd.122.000895] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/06/2022] [Indexed: 11/22/2022] Open
Abstract
Futibatinib (FUT) is a potent inhibitor of fibroblast growth factor receptor (FGFR) 1-4 that is currently under clinical investigation for intrahepatic cholangiocarcinoma. Unlike its predecessors, FUT possesses an acrylamide warhead which enables it to bind covalently to a free cysteine residue in the FGFR kinase domain. However, it remains uninterrogated if this electrophilic α,β-unsaturated carbonyl scaffold could also directly or indirectly engender off-target covalent binding to nucleophilic centres on other cellular proteins. Here, we discovered that FUT inactivated both cytochrome P450 3A (CYP3A) isoforms with K I, k inact, and partition ratio of 12.5 and 51.4 µM, 0.25 and 0.06 min-1 and ~52 and ~58 for CYP3A4 and CYP3A5, respectively. Along with its time-, concentration- and cofactor-dependent inhibitory profile, FUT also exhibited several cardinal features that were consistent with mechanism-based inactivation. Moreover, the nature of inactivation was unlikely to be pseudo-irreversible and instead arose from the covalent modification of the P450 apoprotein and/or its heme moiety due to the lack of substantial enzyme activity recovery following dialysis and chemical oxidation as well as the absence of the diagnostic Soret peak in spectral analyses. Finally, utilizing GSH trapping and high-resolution mass spectrometry, we illuminated that while the acrylamide moiety in FUT could nonenzymatically conjugate to GSH via Michael addition, it was not implicated in the covalent inactivation of CYP3A. Rather, we surmised that it likely stemmed from the metabolic activation of its acrylamide covalent warhead to a highly electrophilic epoxide intermediate that could covalently modify CYP3A and culminate in its catalytic inactivation. Significance Statement In this study, we reported for the first time the inactivation of CYP3A by FUT. Furthermore, using FUT as an exemplary targeted covalent inhibitor, our study revealed the propensity for its acrylamide Michael acceptor moiety to be metabolically activated to a highly electrophilic epoxide. Due to the growing resurgence of covalent inhibitors and the well-established toxicological ramifications associated with epoxides, we advocate that closer scrutiny be adopted when profiling the reactive metabolites of compounds possessing an α,β-unsaturated carbonyl scaffold.
Collapse
Affiliation(s)
| | - Jiaxin Fu
- National University of Singapore, Singapore
| | | | - Guoyi Wu
- National University of Singapore, Singapore
| | - Lei Zhou
- Singapore Eye Research Institute, Singapore
| | | |
Collapse
|
32
|
Establishment and validation of a UPLC-MS/MS bioassay for the quantification of infigratinib in rat plasma. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.103893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
33
|
Bulleyaconitine A is a sensitive substrate and competitive inhibitor of CYP3A4: One of the possible explanations for clinical adverse reactions. Toxicol Appl Pharmacol 2022; 445:116024. [DOI: 10.1016/j.taap.2022.116024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/30/2022] [Accepted: 04/11/2022] [Indexed: 11/23/2022]
|
34
|
Kontsioti E, Maskell S, Bensalem A, Dutta B, Pirmohamed M. Similarity and Consistency Assessment of Three Major Online Drug-Drug Interaction Resources. Br J Clin Pharmacol 2022; 88:4067-4079. [PMID: 35362214 PMCID: PMC9545693 DOI: 10.1111/bcp.15341] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 03/15/2022] [Accepted: 03/28/2022] [Indexed: 11/27/2022] Open
Abstract
AIM To explore the level of agreement on drug-drug interaction (DDI) information listed in three major online drug information resources (DIRs) in terms of: (1) interacting drug pairs; (2) severity rating; (3) evidence rating and (4) clinical management recommendations. METHODS We extracted information from the British National Formulary (BNF), Thesaurus, and Micromedex. Following drug name normalisation, we estimated the overlap of the DIRs in terms of DDI. We annotated clinical management recommendations either manually, where possible, or through application of a machine learning algorithm. RESULTS The DIRs contained 51,481 (BNF), 38,037 (Thesaurus), and 65,446 (Micromedex) drug pairs involved in DDIs. The number of common DDIs across the three DIRs was 6,970 (13.54% of BNF, 18.32% of Thesaurus, and 10.65% of Micromedex). Micromedex and Thesaurus overall showed higher levels of similarity in their severity ratings, while the BNF agreed more with Micromedex on the critical severity ratings and with Thesaurus on the least significant ones. Evidence rating agreement between BNF and Micromedex was generally poor. Variation in clinical management recommendations was also identified, with some categories (i.e. Monitor and Adjust dose) showing higher levels of agreement compared to others (i.e. Use with caution, Wash-out, Modify administration). CONCLUSIONS There is considerable variation in the DDIs included in the examined DIRs, together with variability in categorisation of severity and clinical advice given. DDIs labelled as critical were more likely to appear in multiple DIRs. Such variability in information could have deleterious consequences for patient safety, and there is a need for harmonisation and standardisation.
Collapse
Affiliation(s)
- Elpida Kontsioti
- Department of Electrical Engineering and Electronics, University of Liverpool, Liverpool, United Kingdom.,Institute for Risk and Uncertainty, University of Liverpool, Liverpool, United Kingdom
| | - Simon Maskell
- Department of Electrical Engineering and Electronics, University of Liverpool, Liverpool, United Kingdom
| | | | - Bhaskar Dutta
- Patient Safety Center of Excellence, AstraZeneca, Gaithersburg, MD, United States
| | - Munir Pirmohamed
- The Wolfson Centre for Personalized Medicine, MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
35
|
Novel Bruton's tyrosine kinase inhibitor remibrutinib: Assessment of drug-drug interaction potential as a perpetrator of cytochrome P450 enzymes and drug transporters and the impact of covalent binding on possible drug interactions. Eur J Pharm Sci 2022; 172:106155. [PMID: 35247543 DOI: 10.1016/j.ejps.2022.106155] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/28/2022] [Accepted: 02/25/2022] [Indexed: 01/08/2023]
Abstract
PURPOSE Pharmacokinetic drug-drug interactions (DDIs) are investigated to ensure safety for patients receiving concomitant medications. Here, we present a strategy to characterise the DDI potential of remibrutinib, as an inhibitor of drug-metabolising enzymes and drug transporters, and as an inducer. Initial in vitro studies were performed, followed by a biomarker-based assessment of induction in a first in human study, concluded by a clinical study to verify initial results. Remibrutinib is a covalent inhibitor of Bruton's Tyrosine kinase inhibitor (BTKi) carrying a reactive acrylamide moiety (warhead), thus the potential contribution of covalent binding (off-target) to observed interactions was investigated as this could lead to prolonged and more potent drug interactions. METHODS DDI assessment was focused on the putative inhibition of key metabolic enzymes (Cytochrome P450, CYP), drug transporters and a potential effect on oral contraceptives (OC) by induction of enzymes that are involved in their clearance (CYP3A4). The impact of covalent binding was assessed by synthesising an identical reference molecule but with an inactivated warhead. RESULTS An interaction potential of limited clinical relevance was revealed for remibrutinib for CYP enzymes and drug transporters. The reactive warhead of remibrutinib had no impact on CYP enzyme and transporter inhibition, including time-dependent inhibition of CYP3A4, but may increase the induction potential of remibrutinib. CONCLUSIONS Observed inhibition of metabolic enzymes indicated that remibrutinib is a weak inhibitor of CYP3A4 and CYP2C9 and is not a clinically relevant inhibitor of uptake and efflux transporters, except for intestinal P-glycoprotein and breast cancer resistance protein inhibition. OC may be safely administered and are effective when given with pharmacologically relevant doses of remibrutinib.
Collapse
|
36
|
Tang LWT, Wei W, Verma RK, Koh SK, Zhou L, Fan H, Chan ECY. Direct and Sequential Bioactivation of Pemigatinib to Reactive Iminium Ion Intermediates Culminate in Mechanism-Based Inactivation of Cytochrome P450 3A. Drug Metab Dispos 2022; 50:529-540. [DOI: 10.1124/dmd.121.000804] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/01/2022] [Indexed: 11/22/2022] Open
|
37
|
Opportunities and Considerations in the Application of Artificial Intelligence to Pharmacokinetic Prediction. Methods Mol Biol 2022; 2390:461-482. [PMID: 34731483 DOI: 10.1007/978-1-0716-1787-8_21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The improvement in the ability of the pharmaceutical industry to predict human pharmacokinetic behavior are attributable to major technological shifts from 1990 to the present day. The opportunity for the application of AI/ML based approaches in the pharmaceutical industry is driven by the abundance of data sets that exist within individual pharmaceutical and biotech companies and the availability, within these environments, of abundant computing power. This chapter seeks to describe opportunities for artificial intelligence to contribute to the assessment and evaluation of the dug metabolism and pharmacokinetic (DMPK) properties of novel compounds across the drug discovery and development continuum. Many initiatives are already underway with respect to the application of AI/ML in predicting pharmacokinetic profiles so the question is not whether AI will influence pharmacokinetic prediction but rather how to best utilize and incorporate this and how to evaluate the value added from these applications. Since our understanding of the underlying biology of the in vitro and in vivo systems with respect to ADME, one of the key challenges to AI-based methods will be the ability to adapt to data sets that change in quality over time.
Collapse
|
38
|
Guengerich FP. Inhibition of Cytochrome P450 Enzymes by Drugs-Molecular Basis and Practical Applications. Biomol Ther (Seoul) 2022; 30:1-18. [PMID: 34475272 PMCID: PMC8724836 DOI: 10.4062/biomolther.2021.102] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 07/22/2021] [Indexed: 11/05/2022] Open
Abstract
Drug-drug interactions are a major cause of hospitalization and deaths related to drug use. A large fraction of these is due to inhibition of enzymes involved in drug metabolism and transport, particularly cytochrome P450 (P450) enzymes. Understanding basic mechanisms of enzyme inhibition is important, particularly in terms of reversibility and the use of the appropriate parameters. In addition to drug-drug interactions, issues have involved interactions of drugs with foods and natural products related to P450 enzymes. Predicting drug-drug interactions is a major effort in drug development in the pharmaceutical industry and regulatory agencies. With appropriate in vitro experiments, it is possible to stratify clinical drug-drug interaction studies. A better understanding of drug interactions and training of physicians and pharmacists has developed. Finally, some P450s have been the targets of drugs in some cancers and other disease states.
Collapse
Affiliation(s)
- F. Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-0146, USA
| |
Collapse
|
39
|
The utility of endogenous glycochenodeoxycholate-3-sulfate and 4β-hydroxycholesterol to evaluate the hepatic disposition of atorvastatin in rats. Asian J Pharm Sci 2021; 16:519-529. [PMID: 34703500 PMCID: PMC8520055 DOI: 10.1016/j.ajps.2021.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 01/06/2021] [Accepted: 03/07/2021] [Indexed: 11/22/2022] Open
Abstract
The liver is an important organ for drugs disposition, and thus how to accurately evaluate hepatic clearance is essential for proper drug dosing. However, there are many limitations in drug dosage adjustment based on liver function and pharmacogenomic testing. In this study, we evaluated the ability of endogenous glycochenodeoxycholate-3-sulfate (GCDCA-S) and 4β-hydroxycholesterol (4β-HC) plasma levels to evaluate organic anion-transporting polypeptide (Oatps)-mediated hepatic uptake and Cyp3a-meidated metabolism of atorvastatin (ATV) in rats. The concentration of ATV and its metabolites, 2-OH ATV and 4-OH ATV, was markedly increased after a single injection of rifampicin (RIF), an inhibitor of Oatps. Concurrently, plasma GCDCA-S levels were also elevated. After a single injection of the Cyp3a inhibitor ketoconazole (KTZ), plasma ATV concentrations were significantly increased and 2-OH ATV concentrations were decreased, consistent with the metabolism of ATV by Cyp3a. However, plasma 4β-HC was not affected by KTZ treatment despite it being a Cyp3a metabolite of cholesterol. After repeated oral administration of RIF, plasma concentrations of ATV, 2-OH ATV and 4-OH ATV were markedly increased and the hepatic uptake ratio of ATV and GCDCA-S was decreased. KTZ did not affect plasma concentrations of ATV, 2-OH ATV and 4-OH ATV, but significantly decreased the metabolic ratio of total and 4-OH ATV. However, the plasma level and hepatic metabolism of 4β-HC were not changed by KTZ. The inhibition of hepatic uptake of GCDCA-S by RIF was fully reversed after a 7-d washout of RIF. Plasma concentration and hepatic uptake ratio of GCDCA-S were correlated with the plasma level and hepatic uptake of ATV in rats with ANIT-induced liver injury, respectively. These results demonstrate that plasma GCDCA-S is a sensitive probe for the assessment of Oatps-mediated hepatic uptake of ATV. However, Cyp3a-mediated metabolism of ATV was not predicted by plasma 4β-HC levels in rats.
Collapse
|
40
|
Nguyen-Vo TH, Trinh QH, Nguyen L, Nguyen-Hoang PU, Nguyen TN, Nguyen DT, Nguyen BP, Le L. iCYP-MFE: Identifying Human Cytochrome P450 Inhibitors Using Multitask Learning and Molecular Fingerprint-Embedded Encoding. J Chem Inf Model 2021; 62:5059-5068. [PMID: 34672553 DOI: 10.1021/acs.jcim.1c00628] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The human cytochrome P450 (CYP) superfamily holds responsibilities for the metabolism of both endogenous and exogenous compounds such as drugs, cellular metabolites, and toxins. The inhibition exerted on the CYP enzymes is closely associated with adverse drug reactions encompassing metabolic failures and induced side effects. In modern drug discovery, identification of potential CYP inhibitors is, therefore, highly essential. Alongside experimental approaches, numerous computational models have been proposed to address this biochemical issue. In this study, we introduce iCYP-MFE, a computational framework for virtual screening on CYP inhibitors toward 1A2, 2C9, 2C19, 2D6, and 3A4 isoforms. iCYP-MFE contains a set of five robust, stable, and effective prediction models developed using multitask learning incorporated with molecular fingerprint-embedded features. The results show that multitask learning can remarkably leverage useful information from related tasks to promote global performance. Comparative analysis indicates that iCYP-MFE achieves three predominant tasks, one equivalent task, and one less effective task compared to state-of-the-art methods. The area under the receiver operating characteristic curve (AUC-ROC) and the area under the precision-recall curve (AUC-PR) were two decisive metrics used for model evaluation. The prediction task for CYP2D6-inhibition achieves the highest AUC-ROC value of 0.93 while the prediction task for CYP1A2-inhibition obtains the highest AUC-PR value of 0.92. The substructural analysis preliminarily explains the nature of the CYP-inhibitory activity of compounds. An online web server for iCYP-MFE with a user-friendly interface was also deployed to support scientific communities in identifying CYP inhibitors.
Collapse
Affiliation(s)
- Thanh-Hoang Nguyen-Vo
- School of Mathematics and Statistics, Victoria University of Wellington, Kelburn Parade, Wellington 6140, New Zealand
| | - Quang H Trinh
- Computational Biology Center, International University-VNU HCMC, Ho Chi Minh City 700000, Vietnam
| | - Loc Nguyen
- Computational Biology Center, International University-VNU HCMC, Ho Chi Minh City 700000, Vietnam
| | - Phuong-Uyen Nguyen-Hoang
- Computational Biology Center, International University-VNU HCMC, Ho Chi Minh City 700000, Vietnam
| | - Thien-Ngan Nguyen
- Computational Biology Center, International University-VNU HCMC, Ho Chi Minh City 700000, Vietnam
| | - Dung T Nguyen
- School of Information and Communication Technology, Hanoi University of Science and Technology, Hanoi 100000, Vietnam
| | - Binh P Nguyen
- School of Mathematics and Statistics, Victoria University of Wellington, Kelburn Parade, Wellington 6140, New Zealand
| | - Ly Le
- Computational Biology Center, International University-VNU HCMC, Ho Chi Minh City 700000, Vietnam.,Vingroup Big Data Institute, Ha Noi 100000, Vietnam
| |
Collapse
|
41
|
Ji SB, Park SY, Bae S, Seo HJ, Kim SE, Lee GM, Wu Z, Liu KH. Comprehensive Investigation of Stereoselective Food Drug Interaction Potential of Resveratrol on Nine P450 and Six UGT Isoforms in Human Liver Microsomes. Pharmaceutics 2021; 13:pharmaceutics13091419. [PMID: 34575495 PMCID: PMC8470274 DOI: 10.3390/pharmaceutics13091419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/29/2021] [Accepted: 09/03/2021] [Indexed: 11/16/2022] Open
Abstract
The stereoselectivity of the food drug inhibition potential of resveratrol on cytochrome P450s and uridine 5'-diphosphoglucuronosyl transferases was investigated in human liver microsomes. Resveratrol enantiomers showed stereoselective inhibition of CYP2C9, CYP3A, and UGT1A1. The inhibitions of CYP1A2, CYP2B6, and CYP2C19 by resveratrol were stereo-nonselective. The estimated Ki values determined for CYP1A2 were 13.8 and 9.2 μM for trans- and cis-resveratrol, respectively. Trans-resveratrol noncompetitively inhibited CYP3A and UGT1A1 activities with Ki values of 23.8 and 27.4 μM, respectively. Trans-resveratrol inhibited CYP1A2, CYP2C19, CYP2E1, and CYP3A in a time-dependent manner with Ki shift values >2.0, while cis-resveratrol time-dependently inhibited CYP2C19 and CYP2E1. The time-dependent inhibition of trans-resveratrol against CYP3A4, CYP2E1, CYP2C19, and CYP1A2 was elucidated using glutathione as a trapping reagent. This information helped the prediction of food drug interaction potentials between resveratrol and co-administered drugs which are mainly metabolized by UGT1A1, CYP1A2, CYP2C19, CYP2E1, and CYP3A.
Collapse
Affiliation(s)
- Seung-Bae Ji
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, Daegu 41566, Korea; (S.-B.J.); (S.-Y.P.); (S.B.); (H.-J.S.); (S.-E.K.); (G.-M.L.)
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Daegu 41566, Korea
| | - So-Young Park
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, Daegu 41566, Korea; (S.-B.J.); (S.-Y.P.); (S.B.); (H.-J.S.); (S.-E.K.); (G.-M.L.)
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Daegu 41566, Korea
| | - Subin Bae
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, Daegu 41566, Korea; (S.-B.J.); (S.-Y.P.); (S.B.); (H.-J.S.); (S.-E.K.); (G.-M.L.)
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Daegu 41566, Korea
| | - Hyung-Ju Seo
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, Daegu 41566, Korea; (S.-B.J.); (S.-Y.P.); (S.B.); (H.-J.S.); (S.-E.K.); (G.-M.L.)
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Daegu 41566, Korea
| | - Sin-Eun Kim
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, Daegu 41566, Korea; (S.-B.J.); (S.-Y.P.); (S.B.); (H.-J.S.); (S.-E.K.); (G.-M.L.)
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Daegu 41566, Korea
| | - Gyung-Min Lee
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, Daegu 41566, Korea; (S.-B.J.); (S.-Y.P.); (S.B.); (H.-J.S.); (S.-E.K.); (G.-M.L.)
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Daegu 41566, Korea
| | - Zhexue Wu
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Daegu 41566, Korea
- Mass Spectrometry Based Convergence Research Institute, Kyungpook National University, Daegu 41566, Korea
- Correspondence: (Z.W.); (K.-H.L.); Tel.: +82-53-950-8567 (Z.W. & K.-H.L.); Fax: +82-53-950-8557 (Z.W. & K.-H.L.)
| | - Kwang-Hyeon Liu
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, Daegu 41566, Korea; (S.-B.J.); (S.-Y.P.); (S.B.); (H.-J.S.); (S.-E.K.); (G.-M.L.)
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Daegu 41566, Korea
- Mass Spectrometry Based Convergence Research Institute, Kyungpook National University, Daegu 41566, Korea
- Correspondence: (Z.W.); (K.-H.L.); Tel.: +82-53-950-8567 (Z.W. & K.-H.L.); Fax: +82-53-950-8557 (Z.W. & K.-H.L.)
| |
Collapse
|
42
|
Huth F, Schiller H, Jin Y, Poller B, Schuhler C, Weis W, Woessner R, Drollmann A, End P. Novel Bruton's Tyrosine Kinase inhibitor remibrutinib: Drug-drug interaction potential as a victim of CYP3A4 inhibitors based on clinical data and PBPK modeling. Clin Transl Sci 2021; 15:118-129. [PMID: 34432364 PMCID: PMC8742645 DOI: 10.1111/cts.13126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/30/2021] [Accepted: 07/14/2021] [Indexed: 12/13/2022] Open
Abstract
Remibrutinib, a novel oral Bruton’s Tyrosine Kinase inhibitor (BTKi) is highly selective for BTK, potentially mitigating the side effects of other BTKis. Enzyme phenotyping identified CYP3A4 to be the predominant elimination pathway of remibrutinib. The impact of concomitant treatment with CYP3A4 inhibitors, grapefruit juice and ritonavir (RTV), was investigated in this study in combination with an intravenous microtracer approach. Pharmacokinetic (PK) parameters, including the fraction absorbed, the fractions escaping intestinal and hepatic first‐pass metabolism, the absolute bioavailability, systemic clearance, volume of distribution at steady‐state, and the fraction metabolized via CYP3A4 were evaluated. Oral remibrutinib exposure increased in the presence of RTV 4.27‐fold, suggesting that remibrutinib is not a sensitive CYP3A4 substrate. The rich PK dataset supported the building of a robust physiologically‐based pharmacokinetic (PBPK) model, which well‐described the therapeutic dose range of 25–100 mg. Simulations of untested scenarios revealed an absence of drug‐drug interaction (DDI) risk between remibrutinib and the weak CYP3A4 inhibitor fluvoxamine (area under the concentration‐time curve ratio [AUCR] <1.25), and a moderate effect with the CYP3A4 inhibitor erythromycin (AUCR: 2.71). Predictions with the moderate and strong CYP3A4 inducers efavirenz and rifampicin, suggested a distinct remibrutinib exposure decrease of 64% and 89%. Oral bioavailability of remibrutinib was 34%. The inclusion of an intravenous microtracer allowed the determination of all relevant remibrutinib PK parameters, which facilitated construction of the PBPK model. This will provide guidance on the selection or restriction of comedications and prediction of DDI risks.
Collapse
Affiliation(s)
- Felix Huth
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Hilmar Schiller
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Yi Jin
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Birk Poller
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | | | - Ralph Woessner
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Anton Drollmann
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Peter End
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
43
|
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
|
44
|
Smith S, Lyman M, Ma B, Tweedie D, Menzel K. Reaction Phenotyping of Low-Turnover Compounds in Long-Term Hepatocyte Cultures Through Persistent Selective Inhibition of Cytochromes P450. Drug Metab Dispos 2021; 49:995-1002. [PMID: 34407991 DOI: 10.1124/dmd.121.000601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/10/2021] [Indexed: 11/22/2022] Open
Abstract
Recognizing the challenges of determining the relative contribution of different drug metabolizing enzymes to the metabolism of slowly metabolized compounds, a cytochrome P450 reaction phenotyping (CRP) method using cocultured human hepatocytes (HEPATOPAC) has been established. In this study, the emphasis on the relative contribution of different cytochrome P450 (P450) isoforms was assessed by persistently inhibiting P450 isoforms over 7 days with human HEPATOPAC. P450 isoform-selective inhibition was achieved with the chemical inhibitors furafylline (CYP1A2), tienilic acid (CYP2C9), (+)-N-3-benzylnirvanol (CYP2C19), paroxetine (CYP2D6), azamulin (CYP3A), and a combination of 1-aminobenzotriazole and tienilic acid (broad spectrum inhibition of P450s). We executed this CRP method using HEPATOPAC by optimizing for the choice of P450 inhibitors, their selectivity, and the temporal effect of inhibitor concentrations on maintaining selectivity of inhibition. In general, the established CRP method using potent and selective chemical inhibitors allows to measure the relative contribution of P450s and to calculate the fraction of metabolism (f m) of low-turnover compounds. Several low-turnover compounds were used to validate this CRP method by determining their hepatic intrinsic clearance and f m, with comparison with literature values. We established the foundation of a robust CRP for low-turnover compound test system which can be expanded to include inhibition of other drug metabolizing enzymes. This generic CRP assay, using human long-term hepatocyte cultures, will be an essential tool in drug development for new chemical entities in the quantitative assessment of the risk as a victim of drug-drug interactions. SIGNIFICANCE STATEMENT: An ongoing trend is to develop drug candidates which have limited metabolic clearance. The current studies report a generic approach to conducting reaction phenotyping studies with human HEPATOPAC, focusing on P450 metabolism of low-turnover compounds. Potent and selective chemical inhibitors were used to assess the relative contribution of the major human P450s. Validation was achieved by confirming hepatic intrinsic clearance and fraction of metabolism for previously reported low-turnover compounds. This approach is adaptable for assessment of all drug metabolizing enzymes.
Collapse
Affiliation(s)
- Sheri Smith
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, New Jersey
| | - Michael Lyman
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, New Jersey
| | - Bennett Ma
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, New Jersey
| | - Donald Tweedie
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, New Jersey
| | - Karsten Menzel
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, New Jersey
| |
Collapse
|
45
|
Gajula SNR, Nadimpalli N, Sonti R. Drug metabolic stability in early drug discovery to develop potential lead compounds. Drug Metab Rev 2021; 53:459-477. [PMID: 34406889 DOI: 10.1080/03602532.2021.1970178] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Knowledge of the metabolic stability of a new drug substance eliminated by biotransformation is essential for envisaging the pharmacokinetic parameters required for deciding drug dosing and frequency. Strategies aimed at modifying lead compounds may improve metabolic stability, thereby reducing the drug dosing frequency. Replacement of selective hydrogens with deuterium can effectively enhance the drug's metabolic stability by increasing the biological half-life. Further, cyclization, change in ring size, and chirality can substantially improve the metabolic stability of drugs. The microsomal t1/2 approach for measuring drug in vitro intrinsic clearance by automated LC-MS/MS offers sensitive high-throughput screens with reliable data. The obtained in vitro intrinsic clearance from metabolic stability data helps predict the drug's in vivo total clearance using different scaling factors and hepatic clearance models. This review summarizes all the recent approaches and technological advancements in metabolic stability studies for narrowing down the potential lead compounds in drug discovery. Further, we summarized the potential pitfalls and assumptions made during the in vivo intrinsic clearance estimation from in vitro intrinsic clearance.
Collapse
Affiliation(s)
- Siva Nageswara Rao Gajula
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Nimisha Nadimpalli
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Rajesh Sonti
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
46
|
Deisenroth C, DeGroot DE, Zurlinden T, Eicher A, McCord J, Lee MY, Carmichael P, Thomas RS. The Alginate Immobilization of Metabolic Enzymes Platform Retrofits an Estrogen Receptor Transactivation Assay With Metabolic Competence. Toxicol Sci 2021; 178:281-301. [PMID: 32991717 DOI: 10.1093/toxsci/kfaa147] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The U.S. EPA Endocrine Disruptor Screening Program utilizes data across the ToxCast/Tox21 high-throughput screening (HTS) programs to evaluate the biological effects of potential endocrine active substances. A potential limitation to the use of in vitro assay data in regulatory decision-making is the lack of coverage for xenobiotic metabolic processes. Both hepatic- and peripheral-tissue metabolism can yield metabolites that exhibit greater activity than the parent compound (bioactivation) or are inactive (bioinactivation) for a given biological target. Interpretation of biological effect data for both putative endocrine active substances, as well as other chemicals, screened in HTS assays may benefit from the addition of xenobiotic metabolic capabilities to decrease the uncertainty in predicting potential hazards to human health. The objective of this study was to develop an approach to retrofit existing HTS assays with hepatic metabolism. The Alginate Immobilization of Metabolic Enzymes (AIME) platform encapsulates hepatic S9 fractions in alginate microspheres attached to 96-well peg lids. Functional characterization across a panel of reference substrates for phase I cytochrome P450 enzymes revealed substrate depletion with expected metabolite accumulation. Performance of the AIME method in the VM7Luc estrogen receptor transactivation assay was evaluated across 15 reference chemicals and 48 test chemicals that yield metabolites previously identified as estrogen receptor active or inactive. The results demonstrate the utility of applying the AIME method for identification of false-positive and false-negative target assay effects, reprioritization of hazard based on metabolism-dependent bioactivity, and enhanced in vivo concordance with the rodent uterotrophic bioassay. Integration of the AIME metabolism method may prove useful for future biochemical and cell-based HTS applications.
Collapse
Affiliation(s)
- Chad Deisenroth
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Danica E DeGroot
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Todd Zurlinden
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Andrew Eicher
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - James McCord
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Mi-Young Lee
- Safety and Environmental Assurance Centre, Unilever, Colworth Science, Park, Bedford, Sharnbrook MK44 1LQ, UK
| | - Paul Carmichael
- Safety and Environmental Assurance Centre, Unilever, Colworth Science, Park, Bedford, Sharnbrook MK44 1LQ, UK
| | - Russell S Thomas
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| |
Collapse
|
47
|
Junaid M, Akter Y, Siddika A, Nayeem SMA, Nahrin A, Afrose SS, Ezaj MMA, Alam MS. Nature-derived hit, lead, and drug-like small molecules: Current status and future aspects against key target proteins of Coronaviruses. Mini Rev Med Chem 2021; 22:498-549. [PMID: 34353257 DOI: 10.2174/1389557521666210805113231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND COVID-19 pandemic, the most unprecedented event of the year 2020, has brought millions of scientists worldwide in a single platform to fight against it. Though several drugs are now in the clinical trial, few vaccines available on the market already but the lack of an effect of those is making the situation worse. AIM OF THE STUDY In this review, we demonstrated comprehensive data of natural antiviral products showing activities against different proteins of Human Coronaviruses (HCoV) that are responsible for its pathogenesis. Furthermore, we categorized the compounds into the hit, lead, and drug based on the IC50/EC50 value, drug-likeness, and lead-likeness test to portray their potentiality to be a drug. We also demonstrated the present status of our screened antiviral compounds with respect to clinical trials and reported the lead compounds that can be promoted to clinical trial against COVID-19. METHODS A systematic search strategy was employed focusing on Natural Products (NPs) with proven activity (in vitro, in vivo, or in silico) against human coronaviruses, in general, and data were gathered from databases like PubMed, Web of Science, Google Scholar, SciVerse, and Scopus. Information regarding clinical trials retrieved from the Clinical Trial database. RESULTS Total "245" natural compounds were identified initially from the literature study. Among them, Glycyrrhizin, Caffeic acid, Curcumin is in phase 3, and Tetrandrine, Cyclosporine, Tacrolimus, Everolimus are in phase 4 clinical trial. Except for Glycyrrhizin, all compounds showed activity against COVID-19. CONCLUSIONS In summary, our demonstrated specific small molecules with lead and drug-like capabilities clarified their position in the drug discovery pipeline and proposed their future research against COVID-19.
Collapse
Affiliation(s)
- Md Junaid
- Natural Products Research Division, Advanced Bioinformatics, Computational Biology and Data Science Laboratory. Bangladesh
| | - Yeasmin Akter
- Natural Products Research Division, Advanced Bioinformatics, Computational Biology and Data Science Laboratory. Bangladesh
| | - Aysha Siddika
- Natural Products Research Division, Advanced Bioinformatics, Computational Biology and Data Science Laboratory. Bangladesh
| | - S M Abdul Nayeem
- Natural Products Research Division, Advanced Bioinformatics, Computational Biology and Data Science Laboratory. Bangladesh
| | - Afsana Nahrin
- Department of Pharmacy, University of Science and Technology Chittagong. Bangladesh
| | - Syeda Samira Afrose
- Natural Products Research Division, Advanced Bioinformatics, Computational Biology and Data Science Laboratory. Bangladesh
| | - Md Muzahid Ahmed Ezaj
- Natural Products Research Division, Advanced Bioinformatics, Computational Biology and Data Science Laboratory. Bangladesh
| | | |
Collapse
|
48
|
Roberts O, Kinvig H, Owen A, Lamorde M, Siccardi M, Scarsi KK. In vitro assessment of the potential for dolutegravir to affect hepatic clearance of levonorgestrel. HIV Med 2021; 22:898-906. [PMID: 34328253 PMCID: PMC9363158 DOI: 10.1111/hiv.13136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 06/07/2021] [Indexed: 12/01/2022]
Abstract
Objectives: The World Health Organization recommends that all countries adopt dolutegravir-based antiretroviral therapy as the preferred regimen for all individuals living with HIV. Levonorgestrel is a commonly used hormonal contraceptive, which undergoes drug–drug interactions with some antiretrovirals, but the potential interaction between dolutegravir and levonorgestrel has not been examined. We aimed to evaluate cytochrome P450 (CYP)-mediated levonorgestrel metabolism and quantify the effects of dolutegravir on levonorgestrel apparent intrinsic clearance (CLint.app.) and CYP gene expression. Methods: In vitro CYP-mediated CLint.app. of levonorgestrel was quantified using a recombinant human CYP (rhCYP) enzyme system. A primary human hepatocyte model of drug metabolism was used to assess the effects of dolutegravir on (1) levonorgestrel CLint.app., using liquid chromatography-tandem mass spectrometry, and (2) the expression of specific CYP enzymes, using quantitative real-time polymerase chain reaction. Results: Levonorgestrel clearance was mediated by multiple rhCYPs, including rhCYP3A4. Under control conditions, levonorgestrel CLint.app. was 22.4 ± 5.0 μL/min/106 hepatocytes. Incubation with 43.1 nM of unbound dolutegravir elevated levonorgestrel CLint.app. to 31.4 ± 7.8 μL/min/106 hepatocytes (P = 0.168), while 142.23 nM increased levonorgestrel CLint.app. to 37.0 ± 2.9 μL/min/106 hepatocytes (P = 0.012). Unbound dolutegravir ≥ 431 nM induced expression of CYP3A4 (≥ two-fold) in a dose-dependent manner, while 1.44 μM of unbound dolutegravir induced CYP2B6 expression 2.2 ± 0.3-fold (P = 0.0004). Conclusions: In summary, this in vitro study suggests that dolutegravir has the potential to increase hepatic clearance of levonorgestrel by inducing both CYP3A and non-CYP3A enzymes. The observed in vitro dolutegravir–levonorgestrel drug–drug interaction should be further examined in clinical studies.
Collapse
Affiliation(s)
- Owain Roberts
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Hannah Kinvig
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Andrew Owen
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, Materials Innovation Factory, University of Liverpool, Liverpool, UK
| | - Mohammed Lamorde
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Marco Siccardi
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Kimberly K Scarsi
- College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
49
|
Tang LWT, Teng JW, Verma RK, Koh SK, Zhou L, Go ML, Fan H, Chan ECY. Infigratinib is a Reversible Inhibitor and Mechanism-based Inactivator of Cytochrome P450 3A4. Drug Metab Dispos 2021; 49:856-868. [PMID: 34326139 DOI: 10.1124/dmd.121.000508] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/08/2021] [Indexed: 11/22/2022] Open
Abstract
Infigratinib (INF) is a promising selective inhibitor of fibroblast growth factor receptors 1-3 that has recently been accorded both orphan drug designation and priority review status by the U.S Food and Drug Administration for the treatment of advanced cholangiocarcinoma. Its propensity to undergo bioactivation to electrophilic species was recently expounded upon. However, other than causing aberrant idiosyncratic toxicities, these reactive intermediates may elicit mechanism-based inactivation (MBI) of cytochrome P450 enzymes (CYP450). In this study, we investigated the interactions between INF and the most abundant hepatic cytochrome P450 3A4 (CYP3A4). Our findings revealed that apart from being a potent noncompetitive reversible inhibitor of CYP3A4, INF inactivated CYP3A4 in a time-, concentration- and NADPH-dependent manner with K I, k inact and partition ratio of 2.45 µM, 0.053 min-1 and 41 respectively when rivaroxaban was employed as the probe substrate. Co-incubation with testosterone (alternative CYP3A substrate) or ketoconazole (direct CYP3A inhibitor) attenuated the rate of inactivation whereas the inclusion of glutathione and catalase did not confer such protection. The lack of enzyme activity recovery following dialysis for 4 hours and oxidation with potassium ferricyanide, coupled with the absence of the characteristic Soret peak signature collectively substantiated that inactivation of CYP3A4 by INF was not mediated by the formation of quasi-irreversible metabolite-intermediate complexes but rather through irreversible covalent adduction to the prosthetic heme and/or apoprotein. Finally, glutathione trapping and high-resolution mass spectrometry experimental results unravelled two plausible bioactivation mechanisms of INF arising from the generation of a p-benzoquinone diimine and epoxide reactive intermediate. Significance Statement The potential of infigratinib (INF) to cause mechanism-based inactivation (MBI) of CYP3A4 was unknown. We report the reversible noncompetitive inhibition and irreversible covalent MBI of CYP3A4 by INF and proposed two potential bioactivation pathways implicating p-benzoquinone diimine and epoxide reactive intermediates. Findings from this study lay the groundwork for future investigation of clinically-relevant drug-drug interactions between INF and concomitant substrates of CYP3A4.
Collapse
Affiliation(s)
| | | | - Ravi Kumar Verma
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore
| | | | - Lei Zhou
- Singapore Eye Research Institute, Singapore
| | - Mei Lin Go
- National University of Singapore, Singapore
| | - Hao Fan
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore
| | | |
Collapse
|
50
|
Chen Y, Nagaraja NV, Fan B, Utley L, Lemieux RM, Popovici-Muller J, Dang L, Kim H, Yan L, Su SSM, Biller SA, Yang H. Preclinical Drug Metabolism, Pharmacokinetic, and Pharmacodynamic Profiles of Ivosidenib, an Inhibitor of Mutant IDH1 for Treatment of IDH1-Mutant Malignancies. Drug Metab Dispos 2021; 49:870-881. [PMID: 34321251 DOI: 10.1124/dmd.120.000234] [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: 08/28/2021] [Accepted: 07/22/2021] [Indexed: 11/22/2022] Open
Abstract
Point mutations in isocitrate dehydrogenase 1 (IDH1) result in conversion of α-ketoglutarate to the oncometabolite, D-2-hydroxyglutarate (2-HG). Ivosidenib is a once daily (q.d.), orally available, potent, mutant IDH1 (mIDH1) inhibitor approved for the treatment of patients with relapsed or refractory acute myeloid leukemia (AML) and intensive chemotherapy-ineligible newly diagnosed AML, with a susceptible IDH1 mutation. We characterized the protein binding, metabolism, metabolites, cell permeability, and drug-drug interaction potential of ivosidenib in humans, monkeys, dogs, rats, and/or mice in in vitro experiments. In vivo pharmacokinetic (PK) profiling and assessment of drug distribution and excretion was undertaken in rats, dogs, and monkeys administered single-dose ivosidenib. The PK/pharmacodynamic (PD) relationship between ivosidenib and 2-HG was analyzed in an mIDH1 xenograft mouse model. Ivosidenib was well absorbed, showed low clearance, and moderate to long terminal half-life (5.3-18.5 hours) in rats, dogs, and monkeys. Brain to plasma exposure ratio was low (2.3%), plasma protein binding was high, and oxidative metabolism was the major elimination pathway. Ivosidenib had high cell permeability and was identified as a substrate for P-glycoprotein. There was moderate induction of cytochrome P450 (CYP) enzymes CYP3A4 and CYP2B6 but minimal CYP inhibition or autoinduction. Tumor 2-HG reduction appeared to be dose and drug-exposure dependent. Ivosidenib showed a favorable PK profile in several animal species, along with a clear PK/PD relationship demonstrating 2-HG inhibition, that translated well to patients with AML. Significance Statement Ivosidenib is a mutant IDH1 (mIDH1) inhibitor approved for the treatment of certain patients with mIDH1 acute myeloid leukemia (AML). In Sprague-Dawley rats, beagle dogs, and cynomolgus monkeys, ivosidenib demonstrated a favorable pharmacokinetic profile, and in female BALB/c mice showed clear dose- and exposure-dependent inhibition of the oncometabolite, D-2-hydroxyglutarate, which is present at abnormal levels in mIDH1 tumors. These findings led to the further development of ivosidenib and are consistent with data from patients with mIDH1 cancers and healthy participants.
Collapse
Affiliation(s)
- Yue Chen
- Agios Pharmaceuticals, Inc., United States Minor Outlying Islands
| | | | - Bin Fan
- Agios Pharmaceuticals, Inc., United States
| | - Luke Utley
- Agios Pharmaceuticals, Inc., United States
| | | | | | - Lenny Dang
- Agios Pharmaceuticals, Inc., United States
| | | | | | | | | | - Hua Yang
- Agios Pharmaceuticals, Inc., United States
| |
Collapse
|