Human carboxylesterase 1A plays a predominant role in hydrolysis of the anti-dyslipidemia agent fenofibrate in humans.

Fenofibrate, a marketed peroxisome proliferator-activated receptor-α (PPARα) agonist, has been widely used for treating severe hypertriglyceridemia and mixed dyslipidemia. As a canonical prodrug, fenofibrate can be rapidly hydrolyzed to release the active metabolite (fenofibric acid) in vivo, but the crucial enzyme(s) responsible for fenofibrate hydrolysis and the related hydrolytic kinetics have not been well-investigated. This study aimed to assign the key organs and crucial enzymes involved in fenofibrate hydrolysis in humans, as well as reveal the impact of fenofibrate hydrolysis on its non-PPAR mediated biological activities. Our results demonstrated that fenofibrate could be rapidly hydrolyzed in the preparations from both human liver and lung to release fenofibric acid. Reaction phenotyping assays coupling with chemical inhibition assays showed that human carboxylesterase 1A (hCES1A) played a predominant role in fenofibrate hydrolysis in human liver and lung, while human carboxylesterase 2A (hCES2A) and human monoacylglycerol esterase (hMAGL) contributed to a very lesser extent. Kinetic analyses showed that fenofibrate could be rapidly hydrolyzed by hCES1A in human liver preparations, while the inherent clearance of hCES1A-catalyzed fenofibrate hydrolysis is much higher (>200-fold) than much higher than that of hCES2A or hMAGL. Biological assays demonstrated that both fenofibrate and fenofibric acid showed very closed Nrf2 agonist effects, but fenofibrate hydrolysis strongly weaken its inhibitory effects against both hCES2A and hNtoum. Collectively, our findings reveal that the liver is the major organ and hCES1A is the predominant enzyme catalyzing fenofibrate hydrolysis in humans, while fenofibrate hydrolysis significantly reduces inhibitory effects of fenofibrate against serine hydrolases. Significance Statement Fenofibrate can be completely converted to fenofibric acid in humans and subsequently exert its pharmacological effects, but the hydrolytic pathways of fenofibrate in humans have not been well investigated. This study reported that the liver was the predominant organ and human carboxylesterase 1A was the crucial enzyme involved in fenofibrate hydrolysis in humans.

[1]  Hao Li,et al.  Pectolinarigenin ameliorates acetaminophen-induced acute liver injury via attenuating oxidative stress and inflammatory response in Nrf2 and PPARa dependent manners. , 2023, Phytomedicine : international journal of phytotherapy and phytopharmacology.

[2]  Zhanjun Jia,et al.  Anti-inflammatory role of fenofibrate in treating diseases , 2023, Biomolecules and biomedicine.

[3]  F. Cheng,et al.  Enzyme-activated prodrugs and their release mechanisms for the treatment of cancer. , 2022, Journal of materials chemistry. B.

[4]  Yui Machida,et al.  Functional and Structural Insights into Human PPARα/δ/γ Subtype Selectivity of Bezafibrate, Fenofibric Acid, and Pemafibrate , 2022, International journal of molecular sciences.

[5]  D. Iliopoulos,et al.  Anti-inflammatory drug combination therapy for atherosclerosis: colchicine and fenofibrate. , 2022, Current Medicinal Chemistry.

[6]  C. Marzolini,et al.  Magnitude of Drug–Drug Interactions in Special Populations , 2022, Pharmaceutics.

[7]  J. Zhang,et al.  High-throughput optical assays for sensing serine hydrolases in living systems and their applications , 2022, TrAC Trends in Analytical Chemistry.

[8]  Jennifer B Dennison,et al.  CES2 sustains HNF4α expression to promote pancreatic adenocarcinoma progression through an epoxide hydrolase-dependent regulatory loop , 2021, Molecular metabolism.

[9]  Feng Zhang,et al.  Human carboxylesterase 1A plays a predominant role in the hydrolytic activation of remdesivir in humans. , 2021, Chemico-biological interactions.

[10]  M. Coughtrie,et al.  Parameterization of Microsomal and Cytosolic Scaling Factors: Methodological and Biological Considerations for Scalar Derivation and Validation , 2020, European Journal of Drug Metabolism and Pharmacokinetics.

[11]  B. Warth,et al.  Drug-Exposome Interactions: The Next Frontier in Precision Medicine. , 2020, Trends in pharmacological sciences.

[12]  Zhongming Wu,et al.  Inhibition of ferroptosis by up-regulating Nrf2 delayed the progression of diabetic nephropathy. , 2020, Free radical biology & medicine.

[13]  D. Yang,et al.  Evaluation of the Pharmacokinetic Drug–Drug Interaction between Micronized Fenofibrate and Pitavastatin in Healthy Volunteers , 2020, Pharmaceutics.

[14]  L. Cai,et al.  The Role of Akt2 in the Protective Effect of Fenofibrate against Diabetic Nephropathy , 2020, International journal of biological sciences.

[15]  T. Ebner,et al.  Clinical Pharmacokinetics and Pharmacodynamics of Nintedanib , 2019, Clinical Pharmacokinetics.

[16]  J. Chen,et al.  Nevadensin is a naturally occurring selective inhibitor of human carboxylesterase 1. , 2018, International journal of biological macromolecules.

[17]  R. B. Sashidhar,et al.  Anti-inflammatory activity of anti-hyperlipidemic drug, fenofibrate, and its phase-I metabolite fenofibric acid: in silico, in vitro, and in vivo studies , 2017, Inflammopharmacology.

[18]  Jian-xing Ma,et al.  Fenofibrate ameliorates diabetic retinopathy by modulating Nrf2 signaling and NLRP3 inflammasome activation , 2018, Molecular and Cellular Biochemistry.

[19]  Guangbo Ge,et al.  Carboxylesterase Inhibitors: An Update. , 2017, Current medicinal chemistry.

[20]  Jarkko Rautio,et al.  Prodrugs in medicinal chemistry and enzyme prodrug therapies , 2017, Advanced drug delivery reviews.

[21]  Jinshun Zhao,et al.  Inhibition of JNK signalling mediates PPARα‐dependent protection against intrahepatic cholestasis by fenofibrate , 2017, British journal of pharmacology.

[22]  C. Jackson,et al.  The Synthesis of Certain Derivatives and Analogues of (-)- and (+)-Galanthamine and an Assessment of their Capacities to Inhibit Acetylcholine Esterase. , 2017, The Journal of organic chemistry.

[23]  R. Lehner,et al.  Carboxylesterases in lipid metabolism: from mouse to human , 2017, Protein & Cell.

[24]  Bhagwat Prasad,et al.  Age-Dependent Absolute Abundance of Hepatic Carboxylesterases (CES1 and CES2) by LC-MS/MS Proteomics: Application to PBPK Modeling of Oseltamivir In Vivo Pharmacokinetics in Infants , 2017, Drug Metabolism and Disposition.

[25]  M. Nobilis,et al.  In vitro metabolism of fenofibric acid by carbonyl reducing enzymes. , 2016, Chemico-biological interactions.

[26]  Guangbo Ge,et al.  Design, synthesis, and structure-activity relationship study of glycyrrhetinic acid derivatives as potent and selective inhibitors against human carboxylesterase 2. , 2016, European journal of medicinal chemistry.

[27]  Guangbo Ge,et al.  A bioluminescent sensor for highly selective and sensitive detection of human carboxylesterase 1 in complex biological samples. , 2016, Chemical communications.

[28]  H. Ginsberg,et al.  Targeted Proteomics Identifies Paraoxonase/Arylesterase 1 (PON1) and Apolipoprotein Cs as Potential Risk Factors for Hypoalphalipoproteinemia in Diabetic Subjects Treated with Fenofibrate and Rosiglitazone* , 2015, Molecular & Cellular Proteomics.

[29]  T. Fukami,et al.  Comparison of substrate specificity among human arylacetamide deacetylase and carboxylesterases. , 2015, European journal of pharmaceutical sciences : official journal of the European Federation for Pharmaceutical Sciences.

[30]  E. Wajs,et al.  Effect of canagliflozin on the pharmacokinetics of glyburide, metformin, and simvastatin in healthy participants , 2015, Clinical pharmacology in drug development.

[31]  W. Bilker,et al.  Pharmacoepidemiologic and in vitro evaluation of potential drug-drug interactions of sulfonylureas with fibrates and statins. , 2014, British journal of clinical pharmacology.

[32]  Yuichiro Sato,et al.  Simultaneous Absolute Protein Quantification of Carboxylesterases 1 and 2 in Human Liver Tissue Fractions using Liquid Chromatography-Tandem Mass Spectrometry , 2012, Drug Metabolism and Disposition.

[33]  T. Filippatos A Review of Time Courses and Predictors of Lipid Changes with Fenofibric Acid-Statin Combination , 2012, Cardiovascular Drugs and Therapy.

[34]  Armin Ruf,et al.  Comparative Molecular Profiling of the PPARα/γ Activator Aleglitazar: PPAR Selectivity, Activity and Interaction with Cofactors , 2012, ChemMedChem.

[35]  M. Kaushik,et al.  Fenofibric acid for hyperlipidemia , 2012, Expert opinion on pharmacotherapy.

[36]  G. Keating,et al.  Fenofibrate , 2011, Drugs.

[37]  J. Ansquer,et al.  Comparison of the Gastrointestinal Absorption and Bioavailability of Fenofibrate and Fenofibric Acid in Humans , 2010, Journal of clinical pharmacology.

[38]  I. Borecki,et al.  Fenofibrate and metabolic syndrome. , 2010, Endocrine, metabolic & immune disorders drug targets.

[39]  William F Bosron,et al.  Human carboxylesterases: an update on CES1, CES2 and CES3. , 2009, Protein and peptide letters.

[40]  V. Fischer,et al.  Absorption, Metabolism, and Excretion of [14C]Vildagliptin, a Novel Dipeptidyl Peptidase 4 Inhibitor, in Humans , 2009, Drug Metabolism and Disposition.

[41]  J. Blanco-Rivero,et al.  Long-term fenofibrate treatment impairs endothelium-dependent dilation to acetylcholine by altering the cyclooxygenase pathway. , 2007, Cardiovascular research.

[42]  B. Palmier,et al.  Neurological recovery-promoting, anti-inflammatory, and anti-oxidative effects afforded by fenofibrate, a PPAR alpha agonist, in traumatic brain injury. , 2007, Journal of neurotrauma.

[43]  Colleen D Lauster,et al.  Vildagliptin: a novel oral therapy for type 2 diabetes mellitus. , 2007, American journal of health-system pharmacy : AJHP : official journal of the American Society of Health-System Pharmacists.

[44]  R. Nave,et al.  The role of esterases in the metabolism of ciclesonide to desisobutyryl-ciclesonide in human tissue. , 2007, Biochemical pharmacology.

[45]  K. Chiba,et al.  Genomic Structure and Transcriptional Regulation of the Rat, Mouse, and Human Carboxylesterase Genes , 2007, Drug metabolism reviews.

[46]  Tetsuo Satoh,et al.  Structure, function and regulation of carboxylesterases. , 2006, Chemico-biological interactions.

[47]  D. Guay Update on fenofibrate. , 2006, Cardiovascular drug reviews.

[48]  G. Amidon,et al.  Nucleoside Ester Prodrug Substrate Specificity of Liver Carboxylesterase , 2006, Journal of Pharmacology and Experimental Therapeutics.

[49]  J. Auwerx,et al.  PPARα downregulates airway inflammation induced by lipopolysaccharide in the mouse , 2005, Respiratory research.

[50]  ReijiroArakawa,et al.  Fenofibric Acid, an Active Form of Fenofibrate, Increases Apolipoprotein A-I–Mediated High-Density Lipoprotein Biogenesis by Enhancing Transcription of ATP-Binding Cassette Transporter A1 Gene in a Liver X Receptor–Dependent Manner , 2005 .

[51]  D. Murry,et al.  Hydrolysis of Capecitabine to 5′-Deoxy-5-fluorocytidine by Human Carboxylesterases and Inhibition by Loperamide , 2005, Journal of Pharmacology and Experimental Therapeutics.

[52]  M. Redinbo,et al.  Mammalian carboxylesterases: from drug targets to protein therapeutics. , 2005, Drug discovery today.

[53]  F. Colpaert,et al.  Cardiovascular drugs inhibit MMP-9 activity from human THP-1 macrophages. , 2004, DNA and cell biology.

[54]  J. Yano,et al.  A Novel Selective Peroxisome Proliferator-Activated Receptor α Agonist, 2-Methyl-c-5-[4-[5-methyl-2-(4-methylphenyl)-4-oxazolyl]butyl]-1,3-dioxane-r-2-carboxylic acid (NS-220), Potently Decreases Plasma Triglyceride and Glucose Levels and Modifies Lipoprotein Profiles in KK-Ay Mice , 2004, Journal of Pharmacology and Experimental Therapeutics.

[55]  B. Ramjattan,et al.  Efficacy and tolerability of a "suprabioavailable" formulation of fenofibrate in patients with dyslipidemia: a pooled analysis of two open-label trials. , 2002, Clinical therapeutics.

[56]  アシュラフ・サエード,et al.  Peroxisome proliferator-activated receptor alpha agonist , 2001 .

[57]  P. Hubert,et al.  Determination of fenofibric acid in human plasma using automated solid-phase extraction coupled to liquid chromatography. , 2000, Journal of chromatography. B, Biomedical sciences and applications.

[58]  K. Kitamoto,et al.  Purification and characterization of isoamyl acetate-hydrolyzing esterase encoded by the IAH1 gene of Saccharomyces cerevisiae from a recombinant Escherichia coli , 2000, Applied Microbiology and Biotechnology.

[59]  T. Willson,et al.  The PPARs: from orphan receptors to drug discovery. , 2000, Journal of medicinal chemistry.

[60]  D. Guay Micronized Fenofibrate: A New Fibric Acid Hypolipidemic Agent , 1999, The Annals of pharmacotherapy.

[61]  F. Murad,et al.  Regional differences in endothelin converting enzyme activity in rat brain: inhibition by phosphoramidon and EDTA , 1992, British journal of pharmacology.

[62]  M. Chapman Pharmacology of fenofibrate. , 1987, The American journal of medicine.

[63]  R. Mentlein,et al.  Hydrolysis of ester- and amide-type drugs by the purified isoenzymes of nonspecific carboxylesterase from rat liver. , 1984, Biochemical pharmacology.

[64]  J. Lorsch Practical steady-state enzyme kinetics. , 2014, Methods in enzymology.

[65]  H. Milionis,et al.  Treatment of hyperlipidaemia with fenofibrate and related fibrates. , 2008, Expert opinion on investigational drugs.

[66]  Y. Altuntas,et al.  The effect of dual PPAR alpha/gamma stimulation with combination of rosiglitazone and fenofibrate on metabolic parameters in type 2 diabetic patients. , 2006, Diabetes Research and Clinical Practice.

[67]  S. Daskalopoulou,et al.  Fenofibrate: metabolic and pleiotropic effects. , 2005, Current vascular pharmacology.

[68]  G. Zhou,et al.  Activation of PPARalpha or gamma reduces secretion of matrix metalloproteinase 9 but not interleukin 8 from human monocytic THP-1 cells. , 2000, Biochemical and biophysical research communications.

[69]  J. Spence,et al.  Clinical Pharmacokinetics of Fibric Acid Derivatives (Fibrates) , 1998, Clinical pharmacokinetics.

[70]  D. Faulds,et al.  Micronised fenofibrate: a review of its pharmacodynamic properties and clinical efficacy in the management of dyslipidaemia. , 1997, Drugs.

[71]  N. Di Lascio,et al.  Tissue distribution of drug-metabolizing enzymes in humans. , 1988, Xenobiotica; the fate of foreign compounds in biological systems.