1
|
D’Erasmo M, Sharma SK, Pribut N, Basson A, Dasari M, Bartsch P, Iskandar SE, Giesler KE, Burton S, Derdeyn CA, Liotta DC, Miller EJ. Building Metabolically Stable and Potent Anti-HIV Thioether-Lipid Analogues of Tenofovir Exalidex: A thorough Pharmacological Analysis. J Med Chem 2024; 67:18204-18220. [PMID: 39411803 PMCID: PMC11513920 DOI: 10.1021/acs.jmedchem.4c01510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
Inherently limited by poor bioavailability, antiviral agent tenofovir (TFV) is administered to people living with HIV in prodrug form. However, current prodrugs are prematurely metabolized, compromising access to HIV-infected cells and inducing toxicity. Inspired by lipid conjugate TFV exalidex (TXL), we recently disclosed TXL analogs with potent activity and robust hepatic stability in vitro, as well as attractive oral PK profiles in vivo. In parallel, we discovered the equipotent and equally stable hexadecylthiopropyl (HTP) derivative of TXL (2a). Reported herein are the synthetic and bioanalytic efforts that led to potent, safe, and hepatically stable HTP derivatives. While HTP analog 16h showed the most attractive PK profile in mice (55% F) discrepancies in translating in vitro cell-based results to in vivo PK data, for certain prodrugs, indicated that further in vitro/in vivo optimization is required for continued advancement of this program.
Collapse
Affiliation(s)
- Michael
P. D’Erasmo
- Department
of Chemistry, Emory University College of
Arts & Sciences, Atlanta, Georgia 30322, United States
| | - Savita K. Sharma
- Department
of Chemistry, Emory University College of
Arts & Sciences, Atlanta, Georgia 30322, United States
| | - Nicole Pribut
- Department
of Chemistry, Emory University College of
Arts & Sciences, Atlanta, Georgia 30322, United States
| | - Adriaan Basson
- HIV
Pathogenesis Research Unit, Department of Molecular Medicine and Haematology, University of the Witwatersrand, Johannesburg 2000, Gauteng, South Africa
| | - Madhuri Dasari
- Department
of Chemistry, Emory University College of
Arts & Sciences, Atlanta, Georgia 30322, United States
| | - Perry Bartsch
- Department
of Chemistry, Emory University College of
Arts & Sciences, Atlanta, Georgia 30322, United States
| | - Sabrina E. Iskandar
- Department
of Chemistry, Emory University College of
Arts & Sciences, Atlanta, Georgia 30322, United States
| | - Kyle E. Giesler
- Department
of Chemistry, Emory University College of
Arts & Sciences, Atlanta, Georgia 30322, United States
| | - Samantha Burton
- Department
of Chemistry, Emory University College of
Arts & Sciences, Atlanta, Georgia 30322, United States
| | - Cindy A. Derdeyn
- Department
of Laboratory Medicine & Pathology, University of Washington School of Medicine, Seattle, Washington 98195, United States
| | - Dennis C. Liotta
- Department
of Chemistry, Emory University College of
Arts & Sciences, Atlanta, Georgia 30322, United States
| | - Eric J. Miller
- Department
of Pharmacology & Chemical Biology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| |
Collapse
|
2
|
Chen Y, Wang L, Wang Y, Fang Y, Shen W, Si Y, Zheng X, Zeng S. Integrative Analysis of Histone Acetylation Regulated CYP4F12 in Esophageal Cancer Development. Drug Metab Dispos 2024; 52:813-823. [PMID: 38811154 DOI: 10.1124/dmd.124.001674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/27/2024] [Accepted: 05/08/2024] [Indexed: 05/31/2024] Open
Abstract
Current therapeutic strategies for esophageal cancer (EC) patients have yielded limited improvements in survival rates. Recent research has highlighted the influence of drug metabolism enzymes on both drug response and EC development. Our study aims to identify specific drug metabolism enzymes regulated by histone acetylation and to elucidate its molecular and clinical features. CYP4F12 exhibited a notable upregulation subsequent to trichostatin A treatment as evidenced by RNA sequencing analysis conducted on the KYSE-150 cell line. The change in gene expression was associated with increased acetylation level of histone 3 K18 and K27 in the promoter. The regulation was dependent on p300. In silicon analysis of both The Cancer Genome Atlas esophageal carcinoma and GSE53624 dataset suggested a critical role of CYP4F12 in EC development, because CYP4F12 was downregulated in tumor tissues and predicted better disease-free survival. Gene ontology analysis has uncovered a robust correlation between CYP4F12 and processes related to cell migration, as well as its involvement in cytosine-mediated immune activities. Further investigation into the relationship between immune cells and CYP4F12 expression has indicated an increased level of B cell infiltration in samples with high CYP4F12 expression. CYP4F12 was also negatively correlated with the expression of inhibitory checkpoints. An accurate predictive nomogram model was established combining with clinical factors and CYP4F12 expression. In conclusion, CYP4F12 was crucial in EC development, and targeting CYP4F12 may improve the therapeutic efficacy of current treatment in EC patients. SIGNIFICANCE STATEMENT: CYP4F12 expression was downregulated in esophageal cancer (EC) patients and could be induced by trichostatin A. During EC development, CYP4F12 was linked to reduced cell migration and increased infiltration of B cells. CYP4F12 also is a biomarker as prognostic predictors and therapeutic guide in EC patients.
Collapse
Affiliation(s)
- Yanhong Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (Y.C., Y.W., Y.F., S.Z.); and Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China (L.W., W.S., Y.S., X.Z.)
| | - Li Wang
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (Y.C., Y.W., Y.F., S.Z.); and Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China (L.W., W.S., Y.S., X.Z.)
| | - Yuchen Wang
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (Y.C., Y.W., Y.F., S.Z.); and Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China (L.W., W.S., Y.S., X.Z.)
| | - Yanyan Fang
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (Y.C., Y.W., Y.F., S.Z.); and Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China (L.W., W.S., Y.S., X.Z.)
| | - Wenyang Shen
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (Y.C., Y.W., Y.F., S.Z.); and Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China (L.W., W.S., Y.S., X.Z.)
| | - Yingxue Si
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (Y.C., Y.W., Y.F., S.Z.); and Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China (L.W., W.S., Y.S., X.Z.)
| | - Xiaoli Zheng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (Y.C., Y.W., Y.F., S.Z.); and Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China (L.W., W.S., Y.S., X.Z.)
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (Y.C., Y.W., Y.F., S.Z.); and Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China (L.W., W.S., Y.S., X.Z.)
| |
Collapse
|
3
|
Cioffi F, Adam RHI, Bansal R, Broersen K. A Review of Oxidative Stress Products and Related Genes in Early Alzheimer's Disease. J Alzheimers Dis 2021; 83:977-1001. [PMID: 34420962 PMCID: PMC8543250 DOI: 10.3233/jad-210497] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oxidative stress is associated with the progression of Alzheimer’s disease (AD). Reactive oxygen species can modify lipids, DNA, RNA, and proteins in the brain. The products of their peroxidation and oxidation are readily detectable at incipient stages of disease. Based on these oxidation products, various biomarker-based strategies have been developed to identify oxidative stress levels in AD. Known oxidative stress-related biomarkers include lipid peroxidation products F2-isoprostanes, as well as malondialdehyde and 4-hydroxynonenal which both conjugate to specific amino acids to modify proteins, and DNA or RNA oxidation products 8-hydroxy-2’-deoxyguanosine (8-OHdG) and 8-hydroxyguanosine (8-OHG), respectively. The inducible enzyme heme oxygenase type 1 (HO-1) is found to be upregulated in response to oxidative stress-related events in the AD brain. While these global biomarkers for oxidative stress are associated with early-stage AD, they generally poorly differentiate from other neurodegenerative disorders that also coincide with oxidative stress. Redox proteomics approaches provided specificity of oxidative stress-associated biomarkers to AD pathology by the identification of oxidatively damaged pathology-specific proteins. In this review, we discuss the potential combined diagnostic value of these reported biomarkers in the context of AD and discuss eight oxidative stress-related mRNA biomarkers in AD that we newly identified using a transcriptomics approach. We review these genes in the context of their reported involvement in oxidative stress regulation and specificity for AD. Further research is warranted to establish the protein levels and their functionalities as well as the molecular mechanisms by which these potential biomarkers are involved in regulation of oxidative stress levels and their potential for determination of oxidative stress and disease status of AD patients.
Collapse
Affiliation(s)
- Federica Cioffi
- Department of Nanobiophysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Rayan Hassan Ibrahim Adam
- Department of Nanobiophysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Ruchi Bansal
- Department of Medical Cell Biophysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands.,Department of Pharmacokinetics, Toxicology, and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Kerensa Broersen
- Department of Applied Stem Cell Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| |
Collapse
|
4
|
Feng L, Ning J, Tian X, Wang C, Yu Z, Huo X, Xie T, Zhang B, James TD, Ma X. Fluorescent probes for the detection and imaging of Cytochrome P450. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2020.213740] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
5
|
Trivedi A, Wahlstrom J, Mackowski M, Dutta S, Lee E. Pharmacokinetics, Disposition, and Biotransformation of [ 14C]Omecamtiv Mecarbil in Healthy Male Subjects after a Single Intravenous or Oral Dose. Drug Metab Dispos 2021; 49:619-628. [PMID: 34011533 DOI: 10.1124/dmd.121.000444] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/26/2021] [Indexed: 11/22/2022] Open
Abstract
Omecamtiv mecarbil (OM) is a novel cardiac myosin activator that is currently in clinical development for the treatment of heart failure. The absorption and disposition of [14C]OM (60 µCi) were studied after a single intravenous infusion (35 mg over 1 hour) or oral solution dose (35 mg) in 14 healthy male subjects. Mean recovery of the administered [14C]OM dose was 85.1% and 86.5% over 336 hours for the intravenous and oral routes, respectively. After intravenous dosing, 47.8% and 37.3% of the dose was recovered in urine and feces, respectively; after oral dosing, 48.6% and 38.0% was recovered in urine and feces, respectively. Unchanged OM accounted for a minor percentage of radioactivity in urine (mean 7.7% of dose) and feces (mean 4.1% of dose) across all subjects. The major metabolites recovered in urine and feces were M3 (decarbamoylation product) and sequential metabolite M4 (lactam of M3), which accounted for means of 26.5% and 11.6% of the administered dose, respectively. The CYP4 family of enzymes was primarily responsible for the formation of M3 based on in vitro studies. Other metabolic pathways accounted for 14.9% of the administered dose. In pooled plasma, OM, M3, and M4 accounted for 83.8%, 6.0%, and 3.3% of the total [14C]OM-related materials. No other plasma metabolites constituted more than 3% of the administered dose. The bioavailability for OM solution was 93.5% after rapid and extensive absorption. SIGNIFICANCE STATEMENT: This study characterized the absorption and disposition of OM, a novel small molecule being developed for the treatment of heart failure. OM was primarily cleared through metabolism by the CYP4 family through oxidative cleavage of a terminal carbamate moiety that resembles hydrolysis.
Collapse
|
6
|
Khan SR, Al Rijjal D, Piro A, Wheeler MB. Integration of AI and traditional medicine in drug discovery. Drug Discov Today 2021; 26:982-992. [PMID: 33476566 DOI: 10.1016/j.drudis.2021.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/01/2020] [Accepted: 01/11/2021] [Indexed: 11/24/2022]
Abstract
AI integration in plant-based traditional medicine could be used to overcome drug discovery challenges.
Collapse
Affiliation(s)
- Saifur R Khan
- Endocrine and Diabetes Platform, Department of Physiology, University of Toronto, Medical Sciences Building, Room 3352, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Advanced Diagnostics, Metabolism, Toronto General Hospital Research Institute, Toronto, ON, Canada.
| | - Dana Al Rijjal
- Endocrine and Diabetes Platform, Department of Physiology, University of Toronto, Medical Sciences Building, Room 3352, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Advanced Diagnostics, Metabolism, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Anthony Piro
- Endocrine and Diabetes Platform, Department of Physiology, University of Toronto, Medical Sciences Building, Room 3352, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Advanced Diagnostics, Metabolism, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Michael B Wheeler
- Endocrine and Diabetes Platform, Department of Physiology, University of Toronto, Medical Sciences Building, Room 3352, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Advanced Diagnostics, Metabolism, Toronto General Hospital Research Institute, Toronto, ON, Canada
| |
Collapse
|
7
|
Uehara S, Uno Y, Yamazaki H. The marmoset cytochrome P450 superfamily: Sequence/phylogenetic analyses, genomic structure, and catalytic function. Biochem Pharmacol 2019; 171:113721. [PMID: 31751534 DOI: 10.1016/j.bcp.2019.113721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 11/14/2019] [Indexed: 12/23/2022]
Abstract
The common marmoset (Callithrix jacchus) is a New World monkey that has attracted much attention as a potentially useful primate model for preclinical testing. A total of 36 marmoset cytochrome P450 (P450) isoforms in the P450 1-51 subfamilies have been identified and characterized by the application of genome analysis and molecular functional characterization. In this mini-review, we provide an overview of the genomic structures, sequence identities, and substrate selectivities of marmoset P450s compared with those of human P450s. Based on the sequence identity, phylogeny, and genomic organization of marmoset P450s, orthologous relationships were established between human and marmoset P450s. Twenty-four members of the marmoset P450 1A, 2A, 2B, 2C, 2D, 2E, 3A, 4A, and 4F subfamilies shared high degrees of homology in terms of cDNA (>89%) and amino acid sequences (>85%) with the corresponding human P450s; P450 2C76 was among the exceptions. Phylogenetic analysis using amino acid sequences revealed that marmoset P450s in the P450 1-51 families were located in the same clades as their human and macaque P450 homologs. This finding underlines the evolutionary closeness of marmoset P450s to their human and macaque homologs. Most marmoset P450 1-4 enzymes catalyzed the typical drug-metabolizing reactions of the corresponding human P450 homologs, except for some differences of P450 2A6 and 2B6. Consequently, it appears that the substrate specificities of enzymes in the P450 1-4 families are generally similar in marmosets and humans. The information presented here supports a better understanding of the functional characteristics of marmoset P450s and their similarities and differences with human P450s. It is hoped that this mini-review will facilitate the successful use of marmosets as primate models in drug metabolism and pharmacokinetic studies.
Collapse
Affiliation(s)
- Shotaro Uehara
- Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Yasuhiro Uno
- Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima-city, Kagoshima 890-8580, Japan
| | - Hiroshi Yamazaki
- Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan.
| |
Collapse
|
8
|
Effects of dronedarone, amiodarone and their active metabolites on sequential metabolism of arachidonic acid to epoxyeicosatrienoic and dihydroxyeicosatrienoic acids. Biochem Pharmacol 2017; 146:188-198. [DOI: 10.1016/j.bcp.2017.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 09/22/2017] [Indexed: 12/23/2022]
|
9
|
Uehara S, Yuki Y, Uno Y, Inoue T, Sasaki E, Yamazaki H. Terfenadine t-butyl hydroxylation catalyzed by human and marmoset cytochrome P450 3A and 4F enzymes in livers and small intestines. Xenobiotica 2017; 48:342-347. [PMID: 28436281 DOI: 10.1080/00498254.2017.1321811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
1. Roles of human cytochrome P450 (P450) 3A4 in oxidation of an antihistaminic drug terfenadine have been previously investigated in association with terfenadine-ketoconazole interaction. Several antihistamine drugs have been recently identified as substrates for multiple P450 enzymes. In this study, overall roles of P450 3A4, 2J2, and 4F12 enzymes in terfenadine t-butyl hydroxylation were investigated in small intestines and livers from humans, marmosets, and/or cynomolgus monkeys. 2. Human liver microsomes and liver and small intestine microsomes from marmosets and cynomolgus monkeys effectively mediated terfenadine t-butyl hydroxylation. Ketoconazole and N-hydroxy-N'-(4-butyl-2-methylphenyl)-formamidine (a P450 4A/F inhibitor) almost completely and moderately inhibited these activities, respectively, in human liver microsomes; however, these chemicals did not show substantially suppression in marmoset liver. Anti-human P450 3A and 4F antibodies showed the roughly supportive inhibitory effects. 3. Recombinant P450 3A4/90 and 4F12 showed high terfenadine t-butyl hydroxylation activities with substrate inhibition constants of 84-144 μM (under 26-76 μM of Km values), in similar manners to liver and intestine microsomes. 4. These results suggest that human and marmoset P450 3A4/90 and 4F12 in livers or small intestines played important roles in terfenadine t-butyl hydroxylation. Marmosets could be a model for humans during first pass extraction of terfenadine and related substrates.
Collapse
Affiliation(s)
- Shotaro Uehara
- a Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Machida , Tokyo , Japan
| | - Yukako Yuki
- a Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Machida , Tokyo , Japan
| | - Yasuhiro Uno
- b Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd. , Kainan , Wakayama , Japan
| | - Takashi Inoue
- c Department of Applied Developmental Biology , Central Institute for Experimental Animals , Kawasaki , Japan , and
| | - Erika Sasaki
- c Department of Applied Developmental Biology , Central Institute for Experimental Animals , Kawasaki , Japan , and.,d Keio Advanced Research Center, Keio University , Minato-ku, Tokyo , Japan
| | - Hiroshi Yamazaki
- a Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Machida , Tokyo , Japan
| |
Collapse
|
10
|
Uno Y, Uehara S, Yamazaki H. Utility of non-human primates in drug development: Comparison of non-human primate and human drug-metabolizing cytochrome P450 enzymes. Biochem Pharmacol 2016; 121:1-7. [DOI: 10.1016/j.bcp.2016.06.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 06/14/2016] [Indexed: 01/15/2023]
|
11
|
Foti RS, Dalvie DK. Cytochrome P450 and Non-Cytochrome P450 Oxidative Metabolism: Contributions to the Pharmacokinetics, Safety, and Efficacy of Xenobiotics. ACTA ACUST UNITED AC 2016; 44:1229-45. [PMID: 27298339 DOI: 10.1124/dmd.116.071753] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/10/2016] [Indexed: 12/16/2022]
Abstract
The drug-metabolizing enzymes that contribute to the metabolism or bioactivation of a drug play a crucial role in defining the absorption, distribution, metabolism, and excretion properties of that drug. Although the overall effect of the cytochrome P450 (P450) family of drug-metabolizing enzymes in this capacity cannot be understated, advancements in the field of non-P450-mediated metabolism have garnered increasing attention in recent years. This is perhaps a direct result of our ability to systematically avoid P450 liabilities by introducing chemical moieties that are not susceptible to P450 metabolism but, as a result, may introduce key pharmacophores for other drug-metabolizing enzymes. Furthermore, the effects of both P450 and non-P450 metabolism at a drug's site of therapeutic action have also been subject to increased scrutiny. To this end, this Special Section on Emerging Novel Enzyme Pathways in Drug Metabolism will highlight a number of advancements that have recently been reported. The included articles support the important role of non-P450 enzymes in the clearance pathways of U.S. Food and Drug Administration-approved drugs over the past 10 years. Specific examples will detail recent reports of aldehyde oxidase, flavin-containing monooxygenase, and other non-P450 pathways that contribute to the metabolic, pharmacokinetic, or pharmacodynamic properties of xenobiotic compounds. Collectively, this series of articles provides additional support for the role of non-P450-mediated metabolic pathways that contribute to the absorption, distribution, metabolism, and excretion properties of current xenobiotics.
Collapse
Affiliation(s)
- Robert S Foti
- Pharmacokinetics and Drug Metabolism, Amgen, Cambridge, Massachusetts (R.S.F.); and Pharmacokinetics, Dynamics, and Metabolism, Pfizer, La Jolla, California (D.K.D.)
| | - Deepak K Dalvie
- Pharmacokinetics and Drug Metabolism, Amgen, Cambridge, Massachusetts (R.S.F.); and Pharmacokinetics, Dynamics, and Metabolism, Pfizer, La Jolla, California (D.K.D.)
| |
Collapse
|
12
|
Uehara S, Uno Y, Yuki Y, Inoue T, Sasaki E, Yamazaki H. A New Marmoset P450 4F12 Enzyme Expressed in Small Intestines and Livers Efficiently Metabolizes Antihistaminic Drug Ebastine. ACTA ACUST UNITED AC 2016; 44:833-41. [PMID: 27044800 DOI: 10.1124/dmd.116.070367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/01/2016] [Indexed: 01/08/2023]
Abstract
Common marmosets (Callithrix jacchus) are attracting attention as animal models in preclinical studies for drug development. However, cytochrome P450s (P450s), major drug-metabolizing enzymes, have not been fully identified and characterized in marmosets. In this study, based on the four novel P450 4F genes found on the marmoset genome, we successfully isolated P450 4F2, 4F3B, 4F11, and 4F12 cDNAs in marmoset livers. Deduced amino acid sequences of the four marmoset P450 4F forms exhibited high sequence identities (87%-93%) to the human and cynomolgus monkey P450 4F homologs. Marmoset P450 4F3B and 4F11 mRNAs were predominantly expressed in livers, whereas marmoset P450 4F2 and 4F12 mRNAs were highly expressed in small intestines and livers. Four marmoset P450 4F proteins heterologously expressed in Escherichia coli catalyzed the ω-hydroxylation of leukotriene B4 In addition, marmoset P450 4F12 effectively catalyzed the hydroxylation of antiallergy drug ebastine, a human P450 2J/4F probe substrate. Ebastine hydroxylation activities by small intestine and liver microsomes from marmosets and cynomolgus monkeys showed greatly higher values than those of humans. Ebastine hydroxylation activities by marmoset and cynomolgus monkey small intestine microsomes were inhibited (approximately 60%) by anti-P450 4F antibodies, unlike human small intestine microsomes, suggesting that contribution of P450 4F enzymes for ebastine hydroxylation in the small intestine might be different between marmosets/cynomolgus monkeys and humans. These results indicated that marmoset P450 4F2, 4F3B, 4F11, and 4F12 were expressed in livers and/or small intestines and were functional in the metabolism of endogenous and exogenous compounds, similar to those of cynomolgus monkeys and humans.
Collapse
Affiliation(s)
- Shotaro Uehara
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., Y.Y., H.Y.); Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama, Japan (Y.U.); Department of Applied Developmental Biology (T.I.) and Center of Applied Developmental Biology (E.S.), Central Institute for Experimental Animals, Kawasaki, Japan; and Keio Advanced Research Center, Keio University, Minato-ku, Tokyo, Japan (E.S.)
| | - Yasuhiro Uno
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., Y.Y., H.Y.); Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama, Japan (Y.U.); Department of Applied Developmental Biology (T.I.) and Center of Applied Developmental Biology (E.S.), Central Institute for Experimental Animals, Kawasaki, Japan; and Keio Advanced Research Center, Keio University, Minato-ku, Tokyo, Japan (E.S.)
| | - Yukako Yuki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., Y.Y., H.Y.); Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama, Japan (Y.U.); Department of Applied Developmental Biology (T.I.) and Center of Applied Developmental Biology (E.S.), Central Institute for Experimental Animals, Kawasaki, Japan; and Keio Advanced Research Center, Keio University, Minato-ku, Tokyo, Japan (E.S.)
| | - Takashi Inoue
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., Y.Y., H.Y.); Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama, Japan (Y.U.); Department of Applied Developmental Biology (T.I.) and Center of Applied Developmental Biology (E.S.), Central Institute for Experimental Animals, Kawasaki, Japan; and Keio Advanced Research Center, Keio University, Minato-ku, Tokyo, Japan (E.S.)
| | - Erika Sasaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., Y.Y., H.Y.); Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama, Japan (Y.U.); Department of Applied Developmental Biology (T.I.) and Center of Applied Developmental Biology (E.S.), Central Institute for Experimental Animals, Kawasaki, Japan; and Keio Advanced Research Center, Keio University, Minato-ku, Tokyo, Japan (E.S.)
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (S.U., Y.Y., H.Y.); Pharmacokinetics and Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama, Japan (Y.U.); Department of Applied Developmental Biology (T.I.) and Center of Applied Developmental Biology (E.S.), Central Institute for Experimental Animals, Kawasaki, Japan; and Keio Advanced Research Center, Keio University, Minato-ku, Tokyo, Japan (E.S.)
| |
Collapse
|
13
|
Foti RS, Isoherranen N, Zelter A, Dickmann LJ, Buttrick BR, Diaz P, Douguet D. Identification of Tazarotenic Acid as the First Xenobiotic Substrate of Human Retinoic Acid Hydroxylase CYP26A1 and CYP26B1. J Pharmacol Exp Ther 2016; 357:281-92. [PMID: 26937021 DOI: 10.1124/jpet.116.232637] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/26/2016] [Indexed: 11/22/2022] Open
Abstract
Cytochrome P450 (CYP) 26A1 and 26B1 are heme-containing enzymes responsible for metabolizing all-trans retinoic acid (at-RA). No crystal structures have been solved, and therefore homology models that provide structural information are extremely valuable for the development of inhibitors of cytochrome P450 family 26 (CYP26). The objectives of this study were to use homology models of CYP26A1 and CYP26B1 to characterize substrate binding characteristics, to compare structural aspects of their active sites, and to support the role of CYP26 in the metabolism of xenobiotics. Each model was verified by dockingat-RA in the active site and comparing the results to known metabolic profiles ofat-RA. The models were then used to predict the metabolic sites of tazarotenic acid with results verified by in vitro metabolite identification experiments. The CYP26A1 and CYP26B1 homology models predicted that the benzothiopyranyl moiety of tazarotenic acid would be oriented toward the heme of each enzyme and suggested that tazarotenic acid would be a substrate of CYP26A1 and CYP26B1. Metabolite identification experiments indicated that CYP26A1 and CYP26B1 oxidatively metabolized tazarotenic acid on the predicted moiety, with in vitro rates of metabolite formation by CYP26A1 and CYP26B1 being the highest across a panel of enzymes. Molecular analysis of the active sites estimated the active-site volumes of CYP26A1 and CYP26B1 to be 918 Å(3)and 977 Å(3), respectively. Overall, the homology models presented herein describe the enzyme characteristics leading to the metabolism of tazarotenic acid by CYP26A1 and CYP26B1 and support a potential role for the CYP26 enzymes in the metabolism of xenobiotics.
Collapse
Affiliation(s)
- Robert S Foti
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F.); Department of Pharmaceutics, University of Washington, Seattle, Washington (N.I., A.Z., L.J.D., B.R.B.); Core Laboratory for Neuromolecular Production, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana (P.D.); CNRS, Université Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Valbonne, France (D.D.)
| | - Nina Isoherranen
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F.); Department of Pharmaceutics, University of Washington, Seattle, Washington (N.I., A.Z., L.J.D., B.R.B.); Core Laboratory for Neuromolecular Production, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana (P.D.); CNRS, Université Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Valbonne, France (D.D.)
| | - Alex Zelter
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F.); Department of Pharmaceutics, University of Washington, Seattle, Washington (N.I., A.Z., L.J.D., B.R.B.); Core Laboratory for Neuromolecular Production, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana (P.D.); CNRS, Université Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Valbonne, France (D.D.)
| | - Leslie J Dickmann
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F.); Department of Pharmaceutics, University of Washington, Seattle, Washington (N.I., A.Z., L.J.D., B.R.B.); Core Laboratory for Neuromolecular Production, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana (P.D.); CNRS, Université Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Valbonne, France (D.D.)
| | - Brian R Buttrick
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F.); Department of Pharmaceutics, University of Washington, Seattle, Washington (N.I., A.Z., L.J.D., B.R.B.); Core Laboratory for Neuromolecular Production, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana (P.D.); CNRS, Université Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Valbonne, France (D.D.)
| | - Philippe Diaz
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F.); Department of Pharmaceutics, University of Washington, Seattle, Washington (N.I., A.Z., L.J.D., B.R.B.); Core Laboratory for Neuromolecular Production, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana (P.D.); CNRS, Université Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Valbonne, France (D.D.)
| | - Dominique Douguet
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F.); Department of Pharmaceutics, University of Washington, Seattle, Washington (N.I., A.Z., L.J.D., B.R.B.); Core Laboratory for Neuromolecular Production, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana (P.D.); CNRS, Université Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Valbonne, France (D.D.)
| |
Collapse
|
14
|
Zhu SL, Wang L, Cao ZY, Wang J, Jing MZ, Xia ZC, Ao F, Ye LB, Liu S, Zhu Y. Inducible CYP4F12 enhances Hepatitis C virus infection via association with viral nonstructural protein 5B. Biochem Biophys Res Commun 2016; 471:95-102. [PMID: 26845356 DOI: 10.1016/j.bbrc.2016.01.173] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/28/2016] [Indexed: 02/07/2023]
Abstract
Hepatitis C virus (HCV) nonstructural protein 5B (NS5B) functions as an RNA-dependent RNA polymerase in the HCV replication complex derived from the endoplasmic reticulum in hepatic cells. In this study, NS5B was used as bait in a yeast two-hybrid assay to screen a human liver cDNA library. We confirmed that CYP4F12, a member of the cytochrome P450 superfamily, interacted with NS5B. Furthermore, overexpression of CYP4F12 facilitated HCV replication. In contrast, knockdown of CYP4F12 by specific shRNA decreased HCV replication and viral protein expression. Moreover, our results demonstrated that HCV infection increased the binding of the transcription factor SREBP1 to the CYP4F12 promoter and activated the promoter activity, which indicated that HCV infection increased the expression of CYP4F12 through the SREBP1 pathway. Our results showed that HCV infection induced expression of CYP4F12 protein, which bound to the HCV replication complex to facilitate viral replication.
Collapse
Affiliation(s)
- Sheng-Li Zhu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Li Wang
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Zhong-Ying Cao
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Jun Wang
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Ming-Zhen Jing
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Zhang-Chuan Xia
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Fang Ao
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Lin-Bai Ye
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Shi Liu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Ying Zhu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
15
|
Johnson AL, Edson KZ, Totah RA, Rettie AE. Cytochrome P450 ω-Hydroxylases in Inflammation and Cancer. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2015; 74:223-62. [PMID: 26233909 DOI: 10.1016/bs.apha.2015.05.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cytochrome P450-dependent ω-hydroxylation is a prototypic metabolic reaction of CYP4 family members that is important for the elimination and bioactivation of not only therapeutic drugs, but also endogenous compounds, principally fatty acids. Eicosanoids, derived from arachidonic acid, are key substrates in the latter category. Human CYP4 enzymes, mainly CYP4A11, CYP4F2, and CYP4F3B, hydroxylate arachidonic acid at the omega position to form 20-HETE, which has important effects in tumor progression and on angiogenesis and blood pressure regulation in the vasculature and kidney. CYP4F3A in myeloid tissue catalyzes the ω-hydroxylation of leukotriene B4 to 20-hydroxy leukotriene B4, an inactivation process that is critical for the regulation of the inflammatory response. Here, we review the enzymology, tissue distribution, and substrate selectivity of human CYP4 ω-hydroxylases and their roles as catalysts for the formation and termination of the biological effects of key eicosanoid metabolites in inflammation and cancer progression.
Collapse
Affiliation(s)
- Amanda L Johnson
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Katheryne Z Edson
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, Washington, USA; Amgen Inc., Thousand Oaks, California, USA
| | - Rheem A Totah
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Allan E Rettie
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, Washington, USA.
| |
Collapse
|