DMD 40:1515–1526, 2012 Metabolic Activation of Mefenamic...

12
Metabolic Activation of Mefenamic Acid Leading to Mefenamyl-S- Acyl-Glutathione Adduct Formation In Vitro and In Vivo in Rat Mark P. Grillo, Michelle Tadano Lohr, 1 and Jill C. M. Wait 2 Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, California Received April 5, 2012; accepted May 10, 2012 ABSTRACT: Carboxylic acid-containing nonsteroidal anti-inflammatory drugs (NSAIDs) can be metabolized to chemically reactive acyl glucuro- nide and/or S-acyl-CoA thioester metabolites capable of transacy- lating GSH. We investigated the metabolism of the NSAID mefe- namic acid (MFA) to metabolites that transacylate GSH, leading to MFA-S-acyl-GSH thioester (MFA-SG) formation in incubations with rat and human hepatocytes and in vivo in rat bile. Thus, incuba- tion of MFA (1–500 M) with rat hepatocytes led to the detection of MFA-1--O-acyl glucuronide (MFA-1--O-G), MFA-S-acyl- CoA (MFA-SCoA), and MFA-SG by liquid chromatography-tan- dem mass spectrometric analysis. The C max of MFA-SG (330 nM; 10-min incubation with 100 M MFA) was 120- to 1400-fold higher than the C max of drug S-acyl-GSH adducts detected from studies with other carboxylic acid drugs to date. MFA-SG was also detected in incubations with human hepatocytes, but at much lower concentrations. Inhibition of MFA acyl glucuronidation in rat hepatocytes had no effect on MFA-SG formation, whereas a 58 1.7% inhibition of MFA-SCoA formation led to a corresponding 66 3.5% inhibition of MFA-SG production. Reactivity comparisons with GSH in buffer showed MFA-SCoA to be 80-fold more reactive than MFA-1--O-G forming MFA-SG. MFA-SG was detected in MFA- dosed (100 mg/kg) rat bile, where 17.4 g was excreted after admin- istration. In summary, MFA exhibited bioactivation in rat and human hepatocytes and in vivo in rat, leading to reactive acylating derivatives that transacylate GSH. The formation of MFA-SG in hepatocytes was shown not to be mediated by reaction with MFA-1--O-G, and not solely by MFA-SCoA, but perhaps also by intermediary MFA-acyl- adenylate formation, which is currently under investigation. Introduction The fenamate-type nonsteroidal anti-inflammatory drug (NSAID) mefenamic acid [MFA; 2-(2,3-dimethylphenyl)aminobenzoic acid; Ponstel; Fig. 1], an inhibitor of cyclooxygenase used for the treatment of pain and inflammation (Winder et al., 1962), has been shown to lead to instances of rare nephrotoxicity and hepatotoxicity in dosed patients (Robertson et al., 1980; Chan et al., 1991; Somchit et al., 2004). A hypothesis used to explain drug-induced hepatotoxicity is that some toxic drugs become metabolically activated in the liver to reactive metabolites that bind covalently to tissue proteins, leading to cellular dysfunction or immunotoxicity (Nelson, 2001; Baillie, 2008). Carboxylic acid-containing drug-protein adducts are proposed to function as antigens that mediate the formation of idiosyncratic aller- gic reactions (Boelsterli et al., 1995; Zia-Amirhosseini et al., 1995). Therefore, a proposed mechanism for the onset of MFA-mediated toxicities suggests that MFA is bioactivated to chemically reactive metabolites that become covalently bound to protein in liver and kidney tissues, leading to adverse immunological responses (McGurk et al., 1996; Boelsterli, 2002). MFA is metabolized to mefenamyl-1- O-acyl glucuronide (MFA-1--O-G; Fig. 1), which has been proposed to play a key role in MFA-mediated idiosyncratic toxicity (Sato et al., 1993; McGurk et al., 1996). Results from in vitro studies with MFA- 1--O-G showed it to be very stable in phosphate buffer (16.5-h half-life at pH 7.4 and 37°C); however, it was also shown to be chemically reactive, leading to covalent binding to human serum albumin in vitro (McGurk et al., 1996). Acyl glucuronide metabolites of acidic drugs are known to bind covalently to protein by two different mechanisms. These mechanisms include transacylation re- actions of protein nucleophiles by the 1-O-acyl glucuronide isomer and by a glycation mechanism involving the reaction of the open- chain aldehyde form of the acyl migration glucuronide isomers with protein amino groups. Then it is proposed that drug-protein adducts are recognized by the immune system, resulting in an immune re- sponse leading to potential allergic reactions (Zia-Amirhosseini et al., 1995). In addition to reactive acyl glucuronides, thioester-linked acyl- CoA derivatives (Fig. 1) are also electrophilic and can transacylate biological nucleophiles, including protein and GSH, forming S-acyl- GSH thioester adducts (Boelsterli, 2002; Li et al., 2002; Skonberg et 1 Current affiliation: School of Pharmacy, University of California at San Francisco, San Francisco, California. 2 Current affiliation: BioMarin Pharmaceutical Inc., Novato, California. Parts of this work were previously presented as follows: Grillo MP, Tadano- Lohr M, and Wait JCM (2005) Bioactivation of mefenamic acid in rat hepatocytes. 13th North American ISSX/20th JSSX Meeting; 2005 Oct 23–27; Maui, Hawaii. International Society for the Study of Xenobiotics, Washington, DC. Article, publication date, and citation information can be found at http://dmd.aspetjournals.org. http://dx.doi.org/10.1124/dmd.112.046102. ABBREVIATIONS: NSAID, nonsteroidal anti-inflammatory drug; MFA, mefenamic acid; MFA-SCoA, MFA-S-acyl-CoA thioester; MFA-SG, mefe- namyl-S-acyl-glutathione thioester; MFA-1--O-G, mefenamyl-1--O-acyl glucuronide; I-SCoA, ibuprofen-S-acyl-CoA; I-SG, ibuprofen-S-acyl- GSH; D-SG, diclofenac-S-acyl-glutathione; CBZ, carbamazepine; CID, collision-induced dissociation; GST, glutathione S-transferase; LC-MS/ MS, liquid chromatography-tandem mass spectrometry; MRM, multiple reaction monitoring. 1521-009X/12/4008-1515–1526$25.00 DRUG METABOLISM AND DISPOSITION Vol. 40, No. 8 Copyright © 2012 by The American Society for Pharmacology and Experimental Therapeutics 46102/3783056 DMD 40:1515–1526, 2012 1515 at ASPET Journals on May 15, 2018 dmd.aspetjournals.org Downloaded from

Transcript of DMD 40:1515–1526, 2012 Metabolic Activation of Mefenamic...

Page 1: DMD 40:1515–1526, 2012 Metabolic Activation of Mefenamic ...dmd.aspetjournals.org/content/dmd/40/8/1515.full.pdf · Metabolic Activation of Mefenamic Acid ... The fenamate-type

Metabolic Activation of Mefenamic Acid Leading to Mefenamyl-S-Acyl-Glutathione Adduct Formation In Vitro and In Vivo in Rat

Mark P. Grillo, Michelle Tadano Lohr,1 and Jill C. M. Wait2

Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, California

Received April 5, 2012; accepted May 10, 2012

ABSTRACT:

Carboxylic acid-containing nonsteroidal anti-inflammatory drugs(NSAIDs) can be metabolized to chemically reactive acyl glucuro-nide and/or S-acyl-CoA thioester metabolites capable of transacy-lating GSH. We investigated the metabolism of the NSAID mefe-namic acid (MFA) to metabolites that transacylate GSH, leading toMFA-S-acyl-GSH thioester (MFA-SG) formation in incubations withrat and human hepatocytes and in vivo in rat bile. Thus, incuba-tion of MFA (1–500 �M) with rat hepatocytes led to the detectionof MFA-1-�-O-acyl glucuronide (MFA-1-�-O-G), MFA-S-acyl-CoA (MFA-SCoA), and MFA-SG by liquid chromatography-tan-dem mass spectrometric analysis. The Cmax of MFA-SG (330 nM;10-min incubation with 100 �M MFA) was 120- to 1400-foldhigher than the Cmax of drug S-acyl-GSH adducts detected fromstudies with other carboxylic acid drugs to date. MFA-SG was

also detected in incubations with human hepatocytes, but atmuch lower concentrations. Inhibition of MFA acyl glucuronidation inrat hepatocytes had no effect on MFA-SG formation, whereas a 58 �

1.7% inhibition of MFA-SCoA formation led to a corresponding 66 �

3.5% inhibition of MFA-SG production. Reactivity comparisons withGSH in buffer showed MFA-SCoA to be 80-fold more reactive thanMFA-1-�-O-G forming MFA-SG. MFA-SG was detected in MFA-dosed (100 mg/kg) rat bile, where 17.4 �g was excreted after admin-istration. In summary, MFA exhibited bioactivation in rat and humanhepatocytes and in vivo in rat, leading to reactive acylating derivativesthat transacylate GSH. The formation of MFA-SG in hepatocytes wasshown not to be mediated by reaction with MFA-1-�-O-G, and notsolely by MFA-SCoA, but perhaps also by intermediary MFA-acyl-adenylate formation, which is currently under investigation.

Introduction

The fenamate-type nonsteroidal anti-inflammatory drug (NSAID)mefenamic acid [MFA; 2-(2,3-dimethylphenyl)aminobenzoic acid;Ponstel; Fig. 1], an inhibitor of cyclooxygenase used for the treatmentof pain and inflammation (Winder et al., 1962), has been shown tolead to instances of rare nephrotoxicity and hepatotoxicity in dosedpatients (Robertson et al., 1980; Chan et al., 1991; Somchit et al.,2004). A hypothesis used to explain drug-induced hepatotoxicity isthat some toxic drugs become metabolically activated in the liver toreactive metabolites that bind covalently to tissue proteins, leading tocellular dysfunction or immunotoxicity (Nelson, 2001; Baillie, 2008).Carboxylic acid-containing drug-protein adducts are proposed tofunction as antigens that mediate the formation of idiosyncratic aller-gic reactions (Boelsterli et al., 1995; Zia-Amirhosseini et al., 1995).

Therefore, a proposed mechanism for the onset of MFA-mediatedtoxicities suggests that MFA is bioactivated to chemically reactivemetabolites that become covalently bound to protein in liver andkidney tissues, leading to adverse immunological responses (McGurket al., 1996; Boelsterli, 2002). MFA is metabolized to mefenamyl-1-O-acyl glucuronide (MFA-1-�-O-G; Fig. 1), which has been proposedto play a key role in MFA-mediated idiosyncratic toxicity (Sato et al.,1993; McGurk et al., 1996). Results from in vitro studies with MFA-1-�-O-G showed it to be very stable in phosphate buffer (16.5-hhalf-life at pH 7.4 and 37°C); however, it was also shown to bechemically reactive, leading to covalent binding to human serumalbumin in vitro (McGurk et al., 1996). Acyl glucuronide metabolitesof acidic drugs are known to bind covalently to protein by twodifferent mechanisms. These mechanisms include transacylation re-actions of protein nucleophiles by the 1-O-acyl glucuronide isomerand by a glycation mechanism involving the reaction of the open-chain aldehyde form of the acyl migration glucuronide isomers withprotein amino groups. Then it is proposed that drug-protein adductsare recognized by the immune system, resulting in an immune re-sponse leading to potential allergic reactions (Zia-Amirhosseini et al.,1995). In addition to reactive acyl glucuronides, thioester-linked acyl-CoA derivatives (Fig. 1) are also electrophilic and can transacylatebiological nucleophiles, including protein and GSH, forming S-acyl-GSH thioester adducts (Boelsterli, 2002; Li et al., 2002; Skonberg et

1 Current affiliation: School of Pharmacy, University of California at SanFrancisco, San Francisco, California.

2 Current affiliation: BioMarin Pharmaceutical Inc., Novato, California.Parts of this work were previously presented as follows: Grillo MP, Tadano-

Lohr M, and Wait JCM (2005) Bioactivation of mefenamic acid in rat hepatocytes.13th North American ISSX/20th JSSX Meeting; 2005 Oct 23–27; Maui, Hawaii.International Society for the Study of Xenobiotics, Washington, DC.

Article, publication date, and citation information can be found athttp://dmd.aspetjournals.org.

http://dx.doi.org/10.1124/dmd.112.046102.

ABBREVIATIONS: NSAID, nonsteroidal anti-inflammatory drug; MFA, mefenamic acid; MFA-SCoA, MFA-S-acyl-CoA thioester; MFA-SG, mefe-namyl-S-acyl-glutathione thioester; MFA-1-�-O-G, mefenamyl-1-�-O-acyl glucuronide; I-SCoA, ibuprofen-S-acyl-CoA; I-SG, ibuprofen-S-acyl-GSH; D-SG, diclofenac-S-acyl-glutathione; CBZ, carbamazepine; CID, collision-induced dissociation; GST, glutathione S-transferase; LC-MS/MS, liquid chromatography-tandem mass spectrometry; MRM, multiple reaction monitoring.

1521-009X/12/4008-1515–1526$25.00DRUG METABOLISM AND DISPOSITION Vol. 40, No. 8Copyright © 2012 by The American Society for Pharmacology and Experimental Therapeutics 46102/3783056DMD 40:1515–1526, 2012

1515

at ASPE

T Journals on M

ay 15, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 2: DMD 40:1515–1526, 2012 Metabolic Activation of Mefenamic ...dmd.aspetjournals.org/content/dmd/40/8/1515.full.pdf · Metabolic Activation of Mefenamic Acid ... The fenamate-type

al., 2008; Grillo, 2011). Before the present studies, experiments on thedetection of S-acyl-CoA and S-acyl-GSH metabolites formed in vivoor in vitro of fenamic acid-type NSAIDs, including MFA, have notbeen performed.

Thioester-linked GSH adducts are known metabolites of a range ofcarboxylic acid-containing drugs, e.g., clofibric acid (Grillo andBenet, 2002), zomepirac (Grillo and Hua, 2003), diclofenac (Grillo etal., 2003a), tolmetin (Olsen et al., 2007), flunoxaprofen (Grillo et al.,2010), and ibuprofen (Grillo and Hua, 2008). Mechanistic in vitrostudies performed to investigate the relative contribution of acylglucuronidation and S-acyl-CoA formation pathways leading to S-acyl-GSH thioester formation have indicated the S-acyl-CoA forma-tion pathway to predominate in this regard, and therefore it may alsobe the bioactivation pathway predominating in vivo, leading to theacylation of protein nucleophiles by varied carboxylic acid-containingdrugs.

MFA is known to be metabolized in humans by cytochrome P450on the 3-methyl group forming 3-hydroxy-MFA (Glazko, 1966).Subsequent oxidation of the 3-hydroxy group leads to the formation ofthe 3-carboxy-MFA metabolite. The major metabolites isolated fromhuman urine are the acyl glucuronides of MFA, 3-hydroxy-MFA, and3-carboxy-MFA (Sato et al., 1993). Thioether-linked GSH adducts ofMFA have recently been detected from incubations with human liver

microsomes fortified with NADPH by using hybrid triple quadruplelinear ion trap mass spectrometric techniques for screening and iden-tification of GSH-trapped reactive metabolites (Zheng et al., 2007).The formation of the reactive metabolites leading to GSH adductswith [M�H]� ions m/z 547 and m/z 563 was proposed to be mediatedby cytochrome P450-mediated bioactivation of MFA to epoxide andquinone imine intermediates, respectively, that react with GSH. Thepresent studies were designed to examine the contribution of acylglucuronidation and potential acyl-CoA formation pathways on thebioactivation of MFA to reactive intermediates that transacylate GSHin vitro in rat and human hepatocytes and in vivo in rat, leading to theformation of the thioester-linked GSH adduct mefenamyl-S-acyl-glu-tathione (MFA-SG; Fig. 1).

Materials and Methods

Materials. MFA, carbamazepine (CBZ), GSH, lauric acid sodium salt,(�)-borneol, (R)-ibuprofen, coenzyme A, L-glutamate, �-glucuronidase (typeIX-A from Escherichia coli), William’s media E, and glutathione S-transferase(GST; from rat liver, 31 units/mg protein) were purchased from Sigma-Aldrich(St. Louis, MO). MFA-SG (Khera et al., 2010), ibuprofen-S-acyl-glutathione(I-SG) and ibuprofen-S-acyl-CoA (I-SCoA) (Grillo and Hua, 2008), and di-clofenac-S-acyl-glutathione (D-SG) (Grillo et al., 2003a) were available fromprevious studies. Authentic standards of MFA-SCoA and MFA-1-�-O-G weresynthesized as described below. Rat hepatocytes were isolated as described

HN

OHOCH3

H3C

Mefenamic Acid (MFA)Acyl-CoAFormation

Acyl Glucuronidation

HN

SCoAOCH3

H3CHN

OOCH3

H3C

OCO2H

OHOH

OH

Transacylation of Glutathione

MFA-1- -O-acyl glucuronide(MFA 1- -O-G )

MFA-S-acy-CoA(MFA-SCoA)

GSH GSH

OH

HN O

O

OH

HN

SOCH3

H3C

NH

O NH2

OHO

OH

Mefenamyl-S-acyl-glutathione(MFA-SG)

FIG. 1. Proposed scheme for the metabo-lism of MFA by acyl glucuronidation andacyl-CoA formation leading to the transacy-lation of GSH forming MFA-SG.

1516 GRILLO ET AL.

at ASPE

T Journals on M

ay 15, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 3: DMD 40:1515–1526, 2012 Metabolic Activation of Mefenamic ...dmd.aspetjournals.org/content/dmd/40/8/1515.full.pdf · Metabolic Activation of Mefenamic Acid ... The fenamate-type

below. Human cryopreserved hepatocytes (pooled from 20 donors; productnumber X008000, lot DET) were purchased from Celsis In Vitro Technologies(Baltimore, MD). Human suspension hepatocytes (male, single donor) werepurchased from Invitrogen (Carlsbad, CA). All solvents used for liquid chro-matography-tandem mass spectrometry (LC-MS/MS) analyses were of chro-matographic grade. Stock solutions of MFA, MFA-SCoA, MFA-SG, MFA-1-�-O-G, D-SG, I-SCoA, and I-SG were freshly prepared as 1-mM solutions indimethyl sulfoxide for each experiment.

Instrumentation and Analytical Methods. MFA-SCoA, MFA-SG, andMFA-1-�-O-G derivatives were characterized by LC-MS/MS on a TSQ Quan-tum Discovery Max mass spectrometer (Thermo Fisher Scientific, Waltham,MA) linked to an Agilent 1100 HPLC (Agilent Technologies, Santa Clara, CA)and a CTC HTS PAL Autosampler (LEAP Technologies, Carrboro, NC).LC-MS/MS analysis of MFA-SCoA, MFA-SG, and MFA-1-�-O-G derivativeswas performed with a Luna reverse-phase column [5 �, C18(2), 100 Å, 150 �2.00 mm; Phenomenex, Torrance, CA] and eluted with a mobile-phase flowrate of 0.3 ml/min. The mobile phase used for the analysis of MFA-SCoAconsisted of ammonium acetate (10 mM, pH 6.0) in water (solvent A) andacetonitrile (solvent B). The mobile phase used for the analysis of MFA-SGand MFA-1-�-O-G derivates consisted of 0.1% formic acid in water (solventC) and 0.1% formic acid in acetonitrile (solvent D). For these LC-MS/MSanalyses, gradient elution was achieved by increasing solvent B, or solvent D,from 0 to 95% over 13 min. Electrospray ionization was used with the needlepotential held at 4.5 kV. The tandem MS/MS conditions used were 2 mTorrargon collision gas and a collision potential of 35 eV. Positive ion mode fullscan (m/z 50 to m/z 1000) LC-MS/MS analysis was conducted with a scan timeof 0.73 s and a source collision energy of 10 V. Xcalibur software (version 2.0;Thermo Fisher Scientific) was used to acquire all data. NMR spectra wereacquired in methanol-d4 on a 600-MHz spectrometer equipped with a 5-mmcryoprobe (Bruker, Newark, DE).

Synthesis of MFA-SCoA Thioester. MFA-SCoA thioester was obtained in30% yield by a synthetic procedure using ethyl chloroformate and analogousto that previously reported for the synthesis and purification of clofibryl-S-acyl-CoA thioester (Grillo and Benet, 2002). The MFA-SCoA thioester elutedat a retention time of 7.1 min and showed no detectable impurities whenanalyzed by both positive and negative ion LC-MS scan modes via reverse-phase gradient elution as described above. Tandem LC-MS/MS analysis ofsynthetic MFA-SCoA by collision-induced dissociation (CID) of the proton-ated molecular ion at m/z 991 yielded a product ion mass spectrum [m/z (%)]:m/z 768 (coenzyme A [CoASH�H]�, 3%), m/z 582 ([M�H-409]�, 58%), m/z508 ([adenosine triphosphate-2H]�, 21%), m/z 484 ([M�H-507]�, 100%), m/z428 ([adenosine diphosphate � 2H]�, 54%), m/z 382 ([M�H-609]�, 33%),m/z 224 ([MFA�H-H2O]�, 20%), and m/z 136 ([adenine�H]�, 8%).

Synthesis of MFA-1-�-O-G. MFA-1-�-O-G was synthesized by themethod of Baba and Yoshioka (2006) by using commercially available methyl2,3,4-tri-O-acetyl-1-bromo-1-deoxy-�-D-glucopyranuronate to obtain the methyl2,3,4-tri-O-acetyl derivative of MFA-1-�-O-acyl glucuronide, followed by chemo-selective enzymatic removal of the methyl- and acetyl-protecting groups. TandemLC-MS/MS analysis of high-performance liquid chromatography-purified syn-thetic MFA-1-�-O-G by CID of the protonated molecular ion at m/z 418yielded a product ion mass spectrum [m/z (%)]: m/z 242 ([MFA�H]�, 14%)and m/z 224 ([MFA�H-H2O]�, 100%). 1H NMR analysis of MFA-1-�-O-G(CD3OD) was as follows: � 2.16 (3H, s, 2-methyl), � 2.34 (3H, s, 3-methyl),� 3.50 to 3.69 (3H, m, glucuronosyl C2-, C3-, and C4-H), � 4.05 (1H, d, J 9.6,glucuronosyl C5-H), � 5.79 (1H, d, J 7.9, glucuronosyl C1-H), � 6.66 (1H, d,J 8.5, Ar-H), 6.74 (1H, dt, J 0.9 and 7.5, Ar-H), � 7.07 to 7.18 (3H, m, Ar-H),� 7.34 (1H, dt, J 7.9 and 1.6, Ar-H), � 8.08 (1H, dd, J 1.6 and 8.1, Ar-H), and� 9.14 (1H, s, NH). �-Glucuronidase enzyme (1000 units/ml, 30 min, 2-mltotal volume, pH 5.0 and 37°C, used as per the manufacturer’s instructions),which is known to specifically cleave 1-�-O-acyl-linked acyl glucuronides,and not the 2-, 3-, or 4-O-acyl glucuronide migration isomers, was used tofurther confirm the 1-�-O-acyl glucuronide linkage to MFA.

In Vitro Studies with Rat and Human Hepatocytes. Freshly isolatedhepatocytes were prepared and incubated according to the method of Moldeuset al. (1978). Hepatocytes were isolated from male Sprague-Dawley rats(250–300 g; Charles River Laboratories, Inc., Wilmington, MA), and �95%viability was achieved as assessed by trypan blue exclusion testing. Incuba-tions with human hepatocytes were conducted separately with cryopreserved

human hepatocytes (20-donor pooled, product number X008000, lot DET;Celsis In Vitro Technologies) and with human hepatocytes in suspension(single donor, male; Invitrogen). Cryopreserved human hepatocytes (one tube,5 million cells per tube) were thawed in a water bath at 37°C for 5 min withoutshaking. Human hepatocytes in suspension (60 million cells per tube) werewarmed to 37°C for 5 min without shaking. Cells were centrifuged (26g, 1min), and the isolated pellets were gently resuspended in William’s media Ebuffer (pH 7.4; containing 4 mM L-glutamate). Hepatocytes were washed twicemore in this fashion, with resuspension of the final cell pellets in William’smedia E buffer to a cell concentration of 2 million viable cells per millimeter.Greater than 95% viability was achieved as assessed by trypan blue exclusiontesting. Hepatocytes were warmed to 37°C in a water bath under an atmosphereof 95% O2 and 5% CO2 for 15 min before the initiation of metabolismexperiments. Incubations of hepatocytes (2 million viable cells per milliliter;0.5-ml total volume; n � 3) with MFA were performed in William’s media E(pH 7.4, containing 4 mM L-glutamate) in 96-well Costar standard nonsteriledeep-well polypropylene plates (2-ml well volume; Corning Life Sciences,Lowell, MA) and with continuous orbital shaking (20 rpm/min) under anatmosphere of 95% O2 and 5% CO2 at 37°C in a model ORS-200 Boekel/Grantorbital and reciprocating water bath (Expotech USA, Houston, TX).

For time-dependent experiments (n � 3 separate studies), hepatocytes wereincubated with MFA (100 �M, n � 3 incubations per time point) and analyzedfor MFA-SCoA, MFA-SG, and MFA-1-�-O-G formation over a 60-min timeperiod. For the analysis of MFA-SG and MFA-1-�-O-G derivatives, incuba-tions (500 �l) were quenched at 0, 1, 2, 4, 6, 8, 10, 20, 30, and 60 min witha quench solution (500 �l) consisting of acetonitrile, 3% formic acid, and 1�M CBZ internal standard and sealed with a silicone sealing mat for 96-welldeep-well plates (Thermo Fisher Scientific), followed by vortex mixing (10min) in a DVX-2500 Multi-tube Vortexer (VWR, West Chester, PA). Sampleswere then centrifuged (4500 rpm, 30 min, 8–10°C), and aliquots (300 �l) ofthe supernatants were transferred to SPEware 96-well plates (V bottom, 0.8 ml;Thermo Fisher Scientific) before LC-MS/MS analysis. Incubations (n � 3) of(R)-ibuprofen (100 �M, 10 min) with rat hepatocytes were performed andprocessed as described above for the quantification of I-SG.

For the analysis of MFA-SCoA formation, separate incubations were per-formed in triplicate as described above for MFA-SG except that incubationswere quenched with a solution consisting of acetonitrile (without formic acid,500 �l) and CBZ (1 �M), followed by the addition of hexane (500 �l). Thesample plates then were capped, vortex mixed (1 min), and centrifuged (4500rpm, 10 min, 8–10°C), and aliquots (300 �l) of the aqueous layer weretransferred to SPEware 96-well plates for LC-MS/MS analysis of MFA-SCoA.

Concentration-dependent experiments (n � 3 separate studies) were per-formed as described above with increasing concentrations of MFA (0.8, 1.5,3.9, 7.8, 15.6, 31.3, 62.5, 100, 125, 250, and 500 �M) incubated with rathepatocytes (2 million cells per milliliter) for 10 min and processed forLC-MS/MS analysis of MFA-SG, MFA-1-�-O-G, and MFA-SCoA derivativesas described above.

Inhibition experiments were performed with MFA (100 �M) incubated withrat hepatocytes (2 million cells per milliliter) in the presence or absence of(�)-borneol (1000 �M), for the inhibition of MFA-1-�-O-G formation, orlauric acid (1000 �M), for the inhibition of MFA-SCoA formation. Hepatocyteincubations (0.5-ml total volume; n � 3 incubations per treatment) wereperformed for 10 min and quenched as described above for the analysis ofMFA-SG, MFA-1-�-O-G, and MFA-SCoA derivatives. A stock solution of(�)-borneol (1000 mM) was prepared in dimethyl sulfoxide, and controlincubations included the same final concentration of dimethyl sulfoxide (0.1%,v/v). A stock solution of lauric acid sodium salt was prepared as a 100-mMsolution in distilled water, pH 7.

An in vitro experiment was performed to assess the stability of MFA-1-�-O-G and MFA-SG, I-SG, and D-SG thioesters (1 �M) in separate incubationswith rat hepatocytes (2 million cells per milliliter, n � 3 incubations per timepoint). Incubations (500 �l) were quenched at 0, 0.5, 1, 2, 3, 4, 5, 10, 20, and30 min and processed as described above for the LC-MS/MS analysis ofMFA-SG, MFA-1-�-O-G, I-SG, and D-SG as described below.

Stability of MFA-SG, I-SG, and D-SG in Buffer in the Presence andAbsence of GST. An in vitro study was performed to determine the stabilityof MFA-SG, I-SG, and D-SG in incubations with phosphate buffer (0.1 M, pH7.4, 37°C, 0.2-ml incubation volume) in the presence and absence of rat liver

1517MEFENAMIC ACID-S-ACYL-GLUTATHIONE FORMATION

at ASPE

T Journals on M

ay 15, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 4: DMD 40:1515–1526, 2012 Metabolic Activation of Mefenamic ...dmd.aspetjournals.org/content/dmd/40/8/1515.full.pdf · Metabolic Activation of Mefenamic Acid ... The fenamate-type

GST (3 units/ml). Incubations were performed in duplicate for each time pointin 96-well plates as described above and quenched with acetonitrile (contain-ing 3% formic acid, v/v, and 1 �M CBZ, 200 �l) at 0-, 5-, 10-, 20-, 30-, and60-min time points. The quenched incubation mixtures were analyzed directlyby LC-MS/MS for MFA-SG, I-SG, and D-SG as described below.

Identification and Quantification of MFA-SG. Extracts of MFA-treatedrat hepatocyte incubations were analyzed by LC-MS/MS for MFA-SG andCBZ by using the multiple reaction monitoring (MRM) transitions MH� m/z531 to m/z 224, for MFA-SG detection, and MH� m/z 237 to m/z 194, for CBZdetection, in the positive ion mode and using the chromatographic methoddescribed above. Authentic MFA-SG standard eluted at a retention time of 9.1min, whereas CBZ eluted at 8.4 min. The concentration of MFA-SG thioesterwas determined from a linear standard curve generated from MFA-SG/CBZpeak area ratios. The LC-MS/MS quantification of I-SG and D-SG wasperformed using published procedures (Grillo et al., 2003a; Grillo and Hua,2008) except using the LC-MS/MS system and chromatography parametersdescribed above. Under these conditions, the retention times of I-SG andD-SG were 7.0 and 7.2 min, respectively. The concentrations of I-SG andD-SG thioesters were determined from linear standard curves generatedfrom I-SG/CBZ and D-SG/CBZ peak area ratios.

Identification and Quantification of MFA-SCoA. Extracts of MFA-treated rat hepatocyte quenched incubations were analyzed by LC-MS/MS forMFA-SCoA and CBZ by using the MRM transitions MH� m/z 991 to m/z 224and m/z 484 and MH� m/z 237 to m/z 194, respectively, in the positive ionmode and by using the chromatographic method for the analysis of MFA-SCoA as described above. Authentic MFA-SCoA standard eluted at a retentiontime of 6.2 min, whereas CBZ eluted at 7.1 min. The concentration ofMFA-SCoA thioester was determined from a linear standard curve generatedfrom MFA-SCoA/CBZ peak area ratios.

Identification of MFA-1-O-G. Analysis for the formation of MFA-1-�-O-G in incubations of MFA with rat hepatocytes was performed by LC-MS/MS in the positive ion mode with the MRM transitions MH� m/z 418 tom/z 224 for MFA-1-�-O-G detection and MH� m/z 237 to m/z 194 for CBZdetection, using the same LC-MS/MS chromatography method as describedabove for the analysis of MFA-SG. Authentic MFA-1-�-O-G standard elutedat a retention time of 11 min. The concentration of MFA-1-�-O-G wasdetermined from a linear standard curve generated from MFA-1-�-O-G/CBZpeak area ratios.

Reactions of MFA-SCoA and MFA-1-�-O-G with GSH in Buffer. In-cubations (0.5 ml, n � 3) containing MFA-SCoA (1 �M) or MFA-1-�-O-G (1�M) were performed in phosphate buffer (0.1 M, pH 7.4) at 37°C and fortifiedwith 10 mM GSH in 96-well Costar standard nonsterile deep-well polypro-pylene plates (2-ml well volume; Corning Inc.). Corresponding incubationswere also performed with I-SCoA to compare relative reactivity with GSHforming I-SG. Incubations were conducted for 0, 15, 30, 45, and 60 minfollowed by quenching with acetonitrile (0.5 ml) containing 3% formic acid(v/v) and 1 �M CBZ. Corresponding incubations were also performed in thepresence of rat liver GST (3 units/ml). Quenched mixtures then were directlyanalyzed by LC-MS/MS for MFA-SG (as described above). An in vitro studyon the degradation of MFA-SCoA and MFA-1-�-O-G in phosphate buffer (0.1M, pH 7.4, 37°C) was also preformed where the time-dependent degradationof MFA-SCoA and MFA-1-�-O-G (1 �M, separate incubations) was exam-ined as above except in the absence of GSH. Incubations (n � 3 incubationsper time point) were quenched at the 0-, 5-, 8-, and 15-h time points andquantified as described above for analysis of MFA-SCoA and MFA-1-�-O-Gderivatives.

Identification and Quantification of MFA-SG Thioester in Rat Bile.Three male Sprague-Dawley rats (300–330 g) fitted with exteriorized bile ductcannulas (obtained from Charles River Laboratories, Inc.) were housed indi-vidually in metabolic cages for the collection of bile. Rats received MFA (100mg/kg, in water, pH 7.0) by intraperitoneal administration. An artificial bilesalt supplement (13.4 mg/ml taurocholic acid, 0.5 mg/ml KCl, 9.0 mg/mlNaCl) was infused (1 ml/h) via a duodenal cannula during a 6-h collectionperiod. Bile was collected in plastic 50-ml Corning clear polypropylene conicalbottom centrifuge tubes (Thermo Fisher Scientific) containing 600 �l ofphosphoric acid on wet ice. Collected bile samples were then stored frozen(�20°C) until further processing for LC-M/MS analysis. Frozen bile samples(�10–15-ml collections) were thawed on wet ice followed by the addition of

an equal volume of quench solution consisting of acetonitrile containing 3%formic acid (v/v) and 1 �M CBZ internal standard. The samples were thenvortex mixed and centrifuged (14,000 rpm, 10 min), and the supernatants wereanalyzed and quantified by LC-MS/MS for MFA-SG and MFA-1-�-O-Gderivatives as described above. MFA concentration in rat bile was also deter-mined from these same extracts by LC/MS positive ion scanning and extractedion analysis for the protonated molecular ion of MFA (MH� m/z 242) and froma linear standard curve generated from MFA/CBZ peak area ratios.

Results

Identification of MFA-SCoA. Analysis of incubation extracts byLC-MS/MS MRM detection allowed for the identification of MFA-SCoA formed in rat hepatocyte incubations (Fig. 2). The transitionsused for this analysis were MH� m/z 991 to m/z 224 and MH� m/z991 to m/z 484, which were chosen because of being the majorfragmentation pathways for authentic MFA-SCoA as assessed bypositive ion LC-MS/MS CID of the MH� ion (Fig. 2B). Reverse-phase LC-MS/MS analysis showed the presence of MFA-SCoA inincubations of MFA (100 �M) with rat hepatocytes, which coelutedwith authentic MFA-SCoA standard at a retention time of 7.1 min(Fig. 2A). LC-MS/MS analysis of MFA-SCoA formed in hepatocytesprovided a product ion spectrum from CID of the MH� ion at m/z 991that was identical to the authentic MFA-SCoA standard and consistentwith its chemical structure (Fig. 2, B and C; Sidenius et al., 2004).MFA-SCoA was not detected in incubations of MFA with humanhepatocytes.

Identification of MFA-SG. Analysis of extracts by a sensitiveLC-MS/MS MRM detection technique allowed for the identificationof MFA-SG formed in hepatocyte incubations (Figs. 3 and 4). Thetransition used for this analysis was MH� m/z 531 to m/z 224 and waschosen because it was the major fragmentation pathway for MFA-SGas assessed by positive ion LC-MS/MS CID of the MH� ion ofauthentic MFA-SG (Fig. 3B). LC-MS/MS analysis showed the pres-ence of MFA-SG in incubations of MFA (100 �M) with rat hepato-cytes, which coeluted with authentic MFA-SG standard at a retentiontime of 9.2 min (Fig. 3A). LC-MS/MS analysis of MFA-SG formed inhepatocytes provided a product ion spectrum that displayed fragmentions identical to the authentic MFA-SG standard and also consistentwith its chemical structure (Fig. 3, B and C; Baillie and Davis, 1993).The formation of MFA-SG in incubations with human hepatocyteswas also shown and provided LC-MS/MS properties in agreementwith standard MFA-SG (Fig. 4). Small differences in retention timeand tandem mass spectra between the LC-MS/MS data for theMFA-SG detected in rat and human hepatocytes were observed eventhough the same LC-MS/MS parameters and chromatography systemwere used, except that the two analyses were performed �4 monthsapart.

LC-MS/MS Detection of MFA-1-�-O-G. The 1-O-�-acyl gluc-uronide metabolite of MFA was detected by positive ion LC-MS/MSanalysis of rat (Fig. 5) and human (data not shown) hepatocyte MFAincubation extracts. Results from these analyses showed MFA-1-�-O-G eluting at the same retention time as authentic MFA-1-�-O-Gstandard (11 min; Fig. 5A). LC-MS/MS analysis of the MFA-1-�-O-G metabolite formed in hepatocytes incubated with MFA providedproduct ion spectra containing fragment ions that were consistent withits chemical structure and identical to the authentic MFA-1-�-O-Gstandard (Fig. 5, B and C). The product ion spectrum for MFA-1-�-O-G formed in hepatocytes showed that the major fragment ion(100% relative abundance) upon CID of the MH� ion at m/z 418 wasm/z 224. Evidence for acyl migration isomers was not obtained duringthe LC-MS/MS analysis of MFA rat hepatocyte incubation extracts,

1518 GRILLO ET AL.

at ASPE

T Journals on M

ay 15, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 5: DMD 40:1515–1526, 2012 Metabolic Activation of Mefenamic ...dmd.aspetjournals.org/content/dmd/40/8/1515.full.pdf · Metabolic Activation of Mefenamic Acid ... The fenamate-type

which is consistent with the known chemical stability of the MFA-1-�-O-G metabolite (McGurk et al., 1996).

Time Course of MFA-SCoA, MFA-SG, and MFA-1-�-O-G For-mation in Incubations with Rat and Human Hepatocytes. WhenMFA (100 �M) was incubated with rat hepatocytes for up to 60 min,the formation of MFA-SCoA was determined to reach a maximumconcentration (Cmax) of 15 � 3 nM after 30 min of incubation(Fig. 6A). This concentration was much lower than the Cmax observedfor S-acyl-CoA metabolites of (R)-ibuprofen (2600 nM; Grillo andHua, 2008) and phenylacetic acid (1300 nM; Grillo and Lohr, 2009)measured under similar incubation conditions with rat hepatocytes;however, it was similar to the Cmax of flunoxaprofen-S-acyl-CoA (42nM) detected in recent studies with rat hepatocytes incubated with 100�M (R)-flunoxaprofen (Grillo et al., 2010). In addition, the length of

incubation time leading to Cmax (Tmax) of MFA-SCoA was 30 min inthe present studies, whereas the reported Tmax of formation of I-SCoA, phenylacetyl-S-acyl-CoA, and flunoxaprofen-S-acyl-CoA was3- to 7.5-fold shorter at 10, 4, and 6 min, respectively (Grillo, 2011).The formation of MFA-SG thioester in rat hepatocyte incubationswith MFA (100 �M) was more rapid than MFA-SCoA formation andreached a 22-fold higher Cmax of 330 � 22 nM at the 10-min timepoint (Fig. 6A). This Cmax of MFA-SG was 120- to 1400-fold higherthan the Cmax of drug-S-acyl-GSH adducts detected from rat hepato-cyte incubations with other carboxylic acid drugs (100 �M) studied todate. For example, the Cmax of the S-acyl-GSH thioester detected inincubations with rat hepatocytes incubated with 100 �M carboxylicacid drugs from previous studies was 0.24 nM for zomepirac (Grilloand Hua, 2003), 0.8 nM for diclofenac (Grillo et al., 2003a), 1.3 nM

80

90

100 224484

80

90

100

BA

40

50

60

70

ativ

e Ab

unda

nce

42850

60

70

ve A

bund

ance

10

20

30

40

Rel

382

582991

261340

20

30

40

Rel

ati

100 200 300 400 500 600 700 800 900 1000m/z

0208 330

90

100 484

0 2 4 6 8 10 12 14 160

10

Retention Time (min)2 4 6 8 10 12 14 16

60

70

80

90

danc

e

582

D C

30

40

50

Rel

ativ

e A

bund 582

428

382

100 200 300 400 500 600 700 800 900 1000m/z

0

10

20 224 508

991136261

768330

FIG. 2. A, representative reverse-phase gradient LC-MS/MS MRM chromatograms of MFA-SCoA from the analysis of extracts from rat hepatocytes incubated for 10 minwith 100 �M MFA (solid line) and extract from rat hepatocytes spiked with MFA-SCoA authentic standard (10 nM; dashed line). The LC-MS/MS MRM transition usedfor the analysis of MFA-SCoA was MH� m/z 991 to m/z 224 and m/z 484. B, LC-MS/MS tandem mass spectrum of MFA-SCoA formed in rat hepatocyte incubations withMFA (100 �M, 10 min) obtained by CID of the protonated molecular ion at m/z 991. C, corresponding LC-MS/MS tandem mass spectrum of authentic MFA-SCoAstandard. D, the chemical structure and origins of the characteristic fragment ions for MFA-SCoA are as shown.

1519MEFENAMIC ACID-S-ACYL-GLUTATHIONE FORMATION

at ASPE

T Journals on M

ay 15, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 6: DMD 40:1515–1526, 2012 Metabolic Activation of Mefenamic ...dmd.aspetjournals.org/content/dmd/40/8/1515.full.pdf · Metabolic Activation of Mefenamic Acid ... The fenamate-type

for (R)-ibuprofen (Grillo and Hua, 2008), and 2.8 nM for (R)-flunoxaprofen (Grillo et al., 2010). After the 10-min time point, theconcentration of MFA-SG in rat hepatocyte incubations fell sharply ina fairly linear fashion, reaching �50 nM after 60 min of incubation.The time course of MFA-1-�-O-G formation observed from theanalysis of extracts from hepatocyte incubations with MFA (100 �M)showed that the concentration of MFA-1-�-O-G increased throughoutthe 60-min incubation period, reaching 1560 � 120 nM (Fig. 6A). Asignificant consumption of the initial MFA concentration was ob-served over the 60-min time period, leading to approximately 80 �Mremaining (data not shown) and where MFA-1-�-O-G, MFA-SG, andMFA-S-CoA contributed to roughly 7.8, 0.25, and 0.05% of the

amount of MFA consumed, respectively. Incubations of MFA (100�M) with single-donor male human hepatocytes in suspensionshowed a time-dependent formation of MFA-SG that reached a Cmax

of 0.83 � 0.03 nM at the 30-min time point (Fig. 7). This Cmax was�400-fold lower than the Cmax of MFA-SG observed in correspond-ing incubations with rat hepatocytes. In incubations with cryopre-served human hepatocytes (pooled from 20 donors), 10-min incuba-tions were conducted with 100 �M MFA and led to the formation of1.0 � 0.1 nM MFA-SG (data not shown). The MFA-1-�-O-G wasalso detected in the human hepatocyte incubations where a time-dependent formation led to 18.6 � 0.9 �M after 60 min of incubationwith the single-donor hepatocytes in suspension (Fig. 7). Incubations

80

90

100531

384

224

x10 x10

80

90

100

BA

50

60

70

ativ

e A

bund

ance

224

50

60

70

ve A

bund

ance

10

20

30

40

Rel

a

456232

513

20

30

40

Rel

ati

50 100 150 200 250 300 350 400 450 500 550 600m/z

0

10 513495327299242179 402

90

100531

224

x10 x10

0 2 4 6 8 10 12 14 16Retention Time (min)

0

10

0 2 4 6 8 10 12 14 16

D C

60

70

80

90

danc

e

384

D C

30

40

50

Rel

ativ

e A

bund

50 100 150 200 250 300 350 400 450 500 550 600m/z

0

10

20514

456327307 402242

FIG. 3. A, representative reverse-phase gradient LC-MS/MS MRM chromatograms of MFA-SG from the analysis of extracts from rat hepatocytes incubated for 10 minwith 100 �M MFA (solid line) and extract from rat hepatocytes spiked with MFA-SG authentic standard (500 nM; dashed line). The LC-MS/MS MRM transition usedfor the analysis of MFA-SG was MH� m/z 531 to m/z 224. B, LC-MS/MS tandem mass spectrum of MFA-SG formed in rat hepatocyte incubations with MFA (100 �M,10 min) obtained by CID of the protonated molecular ion at m/z 531. C, corresponding LC-MS/MS tandem mass spectrum of authentic MFA-SG standard. D, the chemicalstructure and origins of the characteristic fragment ions for MFA-SG are as shown.

1520 GRILLO ET AL.

at ASPE

T Journals on M

ay 15, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 7: DMD 40:1515–1526, 2012 Metabolic Activation of Mefenamic ...dmd.aspetjournals.org/content/dmd/40/8/1515.full.pdf · Metabolic Activation of Mefenamic Acid ... The fenamate-type

with the cryopreserved human hepatocytes for 10 min showed 3.5 �0.25 �M MFA-1-�-O-G formed (data not shown).

Concentration-Dependent Formation of MFA-SCoA, MFA-SG,and MFA-1-�-O-G in Incubations with Rat Hepatocytes.When rathepatocytes (2 million cells per milliliter) were incubated for 10min with increasing concentrations of MFA, results showed aconcentration-dependent formation of MFA-SCoA from 0.8 to15.6 �M MFA reaching 6 � 2 nM MFA-SCoA at 15.6 �M MFA(Fig. 6B). No significant change in MFA-SCoA formation wasobserved at MFA concentrations studied above 15.6 �M. In com-parison, from these same incubations, the concentration ofMFA-SG formation was also found to increase sharply with in-creasing concentration of MFA (Fig. 6B). The formation ofMFA-SG reached a Cmax of 308 � 6 nM at 100 �M MFA. Above100 �M MFA, MFA-SG concentration did not increase but de-

creased by 25% to 230 � 30 nM at the 500 �M MFA incubationconcentration. From incubations with (R)-ibuprofen (100 �M, 10min), the I-SG thioester was detected at 3 � 1 nM. The concen-tration of MFA-1-�-O-G in the rat hepatocyte incubations in-creased with increasing MFA concentration up to 700 � 80 nM at62.5 �M MFA but then decreased to 460 � 85 nM at the 500-�MMFA incubation concentration.

Time-Dependent Degradation of MFA-SG in Rat Hepatocytes.The time course of degradation of authentic MFA-SG (1 �M) inincubations with rat hepatocytes showed the thioester derivative tobe degraded in a fairly rapid fashion (t1/2 � 8.5 min), whereascorresponding incubations with MFA-1-�-O-G (1 �M) showedcomplete stability after 60 min of incubation (Fig. 8). Although theMFA-SG degradation t1/2 in incubations with rat hepatocytes wasrapid, it was approximately 2.5- to 10.5-fold longer than the

80

90

100

80

90

100224

253384

x3 x3

BA

40

50

60

70

ativ

e A

bund

ance

50

60

70

ive

Abun

danc

e 456

10

20

30

40

Rel

a

10

20

30

40

Rel

ati

331353493189 449

599124 531

0 2 4 6 8 10 120

10

Retention Time (min)50 100 150 200 250 300 350 400 450 500 550 600

m/z0

10 599124 531

100224

x3 x3

C

60

70

80

90

danc

e

456

D

30

40

50

60

Rel

ativ

e A

bund

384321

496275 514

50 100 150 200 250 300 350 400 450 500 550 600m/z

0

10

20

FIG. 4. A, representative reverse-phase gradient LC-MS/MS MRM chromatograms of MFA-SG from the analysis of extracts from human hepatocytes incubated for 10min with 100 �M MFA (solid line) and extract from rat hepatocytes spiked with MFA-SG authentic standard (1 nM; dashed line). The LC-MS/MS MRM transition usedfor the analysis of MFA-SG was MH� m/z 531 to m/z 224. B, LC-MS/MS tandem mass spectrum of MFA-SG formed in rat hepatocyte incubations with MFA (100 �M,10 min) obtained by CID of the protonated molecular ion at m/z 531. C, corresponding LC-MS/MS tandem mass spectrum of authentic MFA-SG standard. D, the chemicalstructure and origins of the characteristic fragment ions for MFA-SG are as shown.

1521MEFENAMIC ACID-S-ACYL-GLUTATHIONE FORMATION

at ASPE

T Journals on M

ay 15, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 8: DMD 40:1515–1526, 2012 Metabolic Activation of Mefenamic ...dmd.aspetjournals.org/content/dmd/40/8/1515.full.pdf · Metabolic Activation of Mefenamic Acid ... The fenamate-type

degradation t1/2 reported for zomepirac-SG (t1/2 � 0.8 min; Grilloand Hua, 2003), D-SG (t1/2 � 1.0 min; Grillo et al., 2003a),flunoxaprofen-SG (t1/2 � 1.5 min; Grillo et al., 2010), and I-SG(t1/2 � 4.0 min; Grillo and Hua, 2008). In the present S-acyl-GSHthioester stability experiments, we also tested for the degradationt1/2 of (R,S)-I-SG and D-SG in incubations with rat hepatocyteswhere results showed their degradation half-lives to be 2.0 and 0.4min, respectively. The degradation of (R)- and (S)-I-SG in incu-bations with rat hepatocytes is known not to be enantioselective(Grillo and Hua, 2008). The products of degradation of MFA-SGwere not determined in this study but are proposed to be due tohydrolysis of the thioester, similar to that determined for D-SG(Grillo et al., 2003a), and not due to �-glutamyltranspeptidaseactivity, which is known to be negligible in rat liver tissue (Hinch-

man and Ballatori, 1990). It is noteworthy that S-acyl-GSH thio-esters are known to be chemically reactive species that cantransacylate nucleophiles such as N-acetylcysteine and thus mightalso contribute to the transacylation of protein nucleophiles (Grilloand Benet, 2002); however, we did not characterize the chemicalreactivity of MFA-SG in the present studies.

Inhibition Study. An inhibition experiment was performed withMFA (100 �M) in incubations with rat hepatocytes (2 million cellsper milliliter, 10 min) in the presence or absence of (�)-borneol(1000 �M), for the inhibition of MFA-1-�-O-G formation (Wat-kins and Klaassen, 1982), or lauric acid (1000 �M), for theinhibition of MFA-SCoA formation (Xiaotao and Hall, 1993).Results showed that coincubation with (�)-borneol led to theinhibition of MFA-1-�-O-G formation by 91%; however, no inhi-

80

90

100

80

90

100224x50 x50

BA

50

60

70

ive

Abu

ndan

ce

50

60

70

ativ

e A

bund

ance

209

20

30

40

Rel

ati

10

20

30

40

Rel

a

248

307315242

267

0 2 4 6 8 10 12 14 160

10

Retention Time (min)

60 100 140 180 220 260 300 340 380 420m/z

0

10 267147 180126

100224x50 x50

D C

60

70

80

90

danc

e 209

D C

30

40

50

Rel

ativ

e Ab

und

248

60 100 140 180 220 260 300 340 380 420m/z

0

10

20242

270 296194 083201 408180119

FIG. 5. A, representative reverse-phase gradient LC-MS/MS MRM chromatograms of MFA-1-�-O-G from the analysis of extracts from rat hepatocytes incubated for 10min with 100 �M MFA (solid line) and extract from rat hepatocytes spiked with MFA-1-�-O-G authentic standard (1000 nM; dashed line). The LC-MS/MS MRM transitionused for the analysis of MFA-1-�-O-G was MH� m/z 418 to m/z 224. B, LC-MS/MS tandem mass spectrum of MFA-1-�-O-G formed in rat hepatocyte incubations withMFA (100 �M, 10 min) obtained by CID of the protonated molecular ion at m/z 418. C, corresponding LC-MS/MS tandem mass spectrum of authentic MFA-1-�-O-Gstandard. D, the chemical structure and origins of the characteristic fragment ions for MFA-1-�-O-G are as shown.

1522 GRILLO ET AL.

at ASPE

T Journals on M

ay 15, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 9: DMD 40:1515–1526, 2012 Metabolic Activation of Mefenamic ...dmd.aspetjournals.org/content/dmd/40/8/1515.full.pdf · Metabolic Activation of Mefenamic Acid ... The fenamate-type

bition of MFA-SG production was observed (Table 1). In contrast,coincubation of MFA with lauric acid led to a 58 � 2% inhibitionof MFA-SCoA formation and to a corresponding 66 � 4% inhi-bition MFA-SG formation (Table 1).

Reaction of GSH with MFA-SCoA and MFA-1-�-O-G in Buf-fer. Incubation of MFA-SCoA with GSH (10 mM) in buffer (0.5-mlincubation volume, 0.1 M potassium phosphate, pH 7.4, 37°C) re-sulted in a rapid transacylation of GSH (3.5 nM MFA-SG formed perminute) generating 208 � 13 nM MFA-SG after 60 min of incubation(Fig. 9A). Compared with I-SCoA, MFA-SCoA was shown to be�2-fold less reactive with GSH, where the reactivity of I-SCoA withGSH led to �440 nM I-SG formed after 60 min of incubation.Corresponding incubations with MFA-1-�-O-G (1�M) showed theacyl glucuronide to be 80-fold less reactive than MFA-SCoA withGSH (0.045 nM MFA-1-�-O-G formed/min), leading to 2.7 � 1.0 nMMFA-SG formed at the 60-min time point (Fig. 9A). In a separatestudy, the reaction of MFA-SCoA or MFA-1-�-O-G (1 �M) withGSH (10 mM) in incubations fortified with rat liver GST led to a 1.3-and 6-fold increase in formation of MFA-SG, respectively (results notshown). MFA-SCoA (1 �M) was found to be completely stable inincubations with buffer (0.1 M potassium phosphate, pH 7.4, 37°C) in

the absence of GSH, whereas MFA-1-�-O-G (1 �M) degraded in alinear fashion over time with a degradation t1/2 of 16.5 h (Fig. 9B).

Stability of MFA-SG, I-SG, and D-SG in buffer with andwithout GST. Results from an in vitro study to determine the abilityof rat liver GST (0.1 M potassium phosphate buffer, pH 7.4, 37°C) tohydrolyze MFA-SG showed MFA-SG to be very stable, where �10%was degraded after 30 min of incubation relative to incubations in theabsence of GST (Fig. 10). The thioester D-SG, in contrast, wascompletely degraded by the 30-min time point in incubations fortifiedwith GST. Corresponding incubations with I-SG showed this thioesterto be similarly as stable as MFA-SG in GST-fortified incubations(11% degraded at 30 min relative to incubations in buffer in theabsence of GST).

Identification of MFA-SG in Rat Bile. After administration ofMFA to male Sprague-Dawley rats (100 mg/kg i.p.), bile was col-lected and processed, and the extract was analyzed for MFA-SG byLC-MS/MS. Results showed the presence of MFA-SG, where 17.4�g were excreted in bile over 6 h of collection. The total amountsof MFA-1-�-O-G and MFA excreted in bile 6 h after administra-tion of MFA were also determined and shown to be 2000 and 4 �g,respectively.

Discussion

Herein, we report MFA to undergo bioactivation to reactivetransacylating species in rat and human hepatocytes and in vivo in rat

0 10 20 30 40 50 600.0

0.2

0.4

0.6

0.8

1.0

0

4000

8000

12000

16000

20000MFA-SGMFA-1-β-O-G

Incubation Time (min)

[MFA

-SG

], n

M

[MFA

-1-β -O-G

], nM

FIG. 7. Time course for the formation of MFA-SG (filled circles) and MFA-1-�-O-G (open triangles) in human hepatocytes (single donor, 2 million cells permilliliter) incubated with 100 �M MFA. Values are expressed as the mean � S.D.from a single experiment, and n � 3 incubations per time point. MFA-SCoA wasnot detected in extracts from human hepatocyte incubations with MFA.

Incubation Time (min)

% R

emai

ning

0 10 20 30 40 50 600

20

40

60

80

100

120

MFA-SGI-SGD-SG

MFA-1-β-O-G

FIG. 8. Time-dependent degradation of 1 �M MFA-SG, MFA-1-�-O-G, I-SG, andD-SG in incubations with rat hepatocytes. Values are expressed as the average oftwo incubations per time point.

Incubation Time (Min)

[MFA

-SC

oA, M

FA-S

G],

nM [MFA

-1-β -O-G

], nM

0 10 20 30 40 50 600

50

100

150

200

250

300

350

0

200

400

600

800

1000

1200

1400

1600

MFA-SG

MFA-1-β-O-GMFA-SCoA

Concentration (µM)[M

FA-S

CoA

, MFA

-SG

], nM [M

FA-1-β -O

-G], nM

010

020

030

040

050

00

50

100

150

200

250

300

350

0

100

200

300

400

500

600

700

800

MFA-SG

MFA-1-β-O-GMFA-SCoA

A B

FIG. 6. A, time course for the formation ofMFA-SCoA (open squares), MFA-SG(filled circles), and MFA-1-�-O-G (opentriangles) in rat hepatocytes (2 million cellsper milliliter) incubated with 100 �M MFA.B, MFA concentration-dependent formationof MFA-SCoA, MFA-SG, and MFA-1-�-O-G in incubation with rat hepatocytes for10 min. Values are expressed as the mean �S.D. from three separate experiments, andn � 3 incubations per time point and MFAconcentration.

1523MEFENAMIC ACID-S-ACYL-GLUTATHIONE FORMATION

at ASPE

T Journals on M

ay 15, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 10: DMD 40:1515–1526, 2012 Metabolic Activation of Mefenamic ...dmd.aspetjournals.org/content/dmd/40/8/1515.full.pdf · Metabolic Activation of Mefenamic Acid ... The fenamate-type

that transacylate the cysteinyl-thiol of GSH forming MFA-S-acyl-GSH thioester. Results from the present studies provide the firstevidence for the formation of S-acyl-CoA and S-acyl-GSH thioestermetabolites of a fenamic acid-containing compound. LC-MS/MSanalysis of extracts from MFA-treated rat hepatocyte incubations ledto the detection of the MFA-SCoA and MFA-SG thioesters, wheretheir tandem mass spectra showed product ions consistent with theirchemical structures (Figs. 2 and 3) and with LC-MS/MS product ionspectra of S-acyl-CoA and S-acyl-GSH derivatives in general (Side-nius et al., 2004; Grillo, 2011). MFA-SG, but not the MFA-SCoAthioester, was detected in incubations with human hepatocytes, but atmuch lower concentrations (Fig. 4). MFA-SG was also identified inMFA-dosed rat bile, where 17.4 �g was excreted in rat bile over a 6-hcollection period, which was �60- and 3-fold greater than the re-ported amount of biliary-excreted D-SG in 200 mg/kg diclofenac-dosed rats (Grillo et al., 2003b) and zomepirac-S-acyl-GSH in 100mg/kg zomepirac-dosed rats (Grillo and Hua, 2003).

Studies on the chemical reactivity of MFA-SCoA and MFA-1-�-O-G with GSH showed the S-acyl-CoA thioester to be �80-foldmore reactive than the acyl glucuronide leading to MFA-SG for-mation (Fig. 9), which is consistent with results from previousstudies comparing the relative chemical reactivity between S-acyl-CoA thioesters and their corresponding acyl glucuronides withGSH (Li et al., 2002; Grillo, 2011). The 1.3- and 6-fold GST-catalyzed increase in MFA-SG formation rates from reactions ofMFA-SCoA and MFA-1-�-O-G with GSH is consistent with the weakcatalysis observed in previous studies with clofibric acid derivatives,where rat liver GST-mediated catalysis was shown to occur inincubations of clofibryl-1-�-O-G (�8-fold; Shore et al., 1995) andclofibryl-S-acyl-CoA (�3-fold; Grillo and Benet, 2002) with GSHforming clofibryl-S-acyl-GSH thioester. In addition, we deter-mined MFA-SCoA to be chemically stable in buffer at pH 7.4 and37°C over 15 h of incubation, whereas MFA-1-�-O-G degradedwith a t1/2 of 16.5 h, which was the same as reported previously

(McGurk et al., 1996; Walker et al., 2007). The degradation ofMFA-1-�-O-G in buffer is known to occur almost exclusivelythrough acyl migration and not by hydrolysis (Walker et al., 2007).

In time-course rat hepatocyte studies, we determined the Cmax ofMFA-SCoA detected in incubations treated with 100 �M MFA to be15 nM after 30 min of incubation, which was 170- and 90-fold lowerthan I-SCoA and phenylacetyl-S-acyl-CoA detected in similar studieswith (R)-ibuprofen (Grillo and Hua, 2008) and phenylacetic acid(Grillo and Lohr, 2009), respectively. Differences in drug-S-acyl-CoAformation may be due to differences in the acyl-CoA synthetase(s)catalyzing their formation; however, the identity of the acyl-CoAsynthetase(s) that mediates the formation of MFA-SCoA is notknown. Thioester S-acyl-CoA formation is catalyzed by acyl-CoAsynthetases located in the endoplasmic reticulum, in the outer mem-brane of mitochondria (long-chain fatty acid acyl-CoA synthetases),and in the mitochondrial matrix (short-, medium-, and branched-chainacyl-CoA synthetases). Mitochondrial medium-chain acyl-CoA syn-thetase is primarily associated with the metabolism of salicylic acid,benzoic acid, and phenylacetic acid, whereas hepatic microsomal andperoxisomal long-chain acyl-CoA synthetases are involved in theformation of S-acyl-CoA thioesters of hypolipidemic and peroxisomalproliferating drugs (e.g., clofibric acid) and of 2-arylproprionic aciddrugs such as ibuprofen (Knights, 1998; Nelson and Cox, 2005).

From the same time-course experiments, incubations of 100 �MMFA with rat hepatocytes led to a MFA-SG Cmax of 330 nM after a10-min of incubation, which was 120- to 1400-fold higher than theCmax of drug-S-acyl-GSH adducts detected from rat hepatocyte incu-bations with other carboxylic acid drugs (100 �M) studied to date. Inthe present work, incubations with (R)-ibuprofen led to the detectionof 3 nM I-SG, which was �100-fold less than the Cmax of MFA-SGformed in the same experiments. In contrast to results from previousstudies with ibuprofen and flunoxaprofen, where the concentration-time profile of S-acyl-GSH formation correlated with S-acyl-CoAformation, the time course of MFA-SG formation was not consistent

0 15 30 45 600

50100150200250300350400450500

MFA-1-β-O-GMFA-SCoA

Incubation Time (min)

[MFA

-SG

or I

-SG

], nM I-SCoA

0 15 30 45 600

1

2

3

4 MFA-1-β-O-G

Incubation Time (min)

[MFA

-SG

], nM

0 5 10 150

102030405060708090

100

MFA-SCoAMFA-1-β-O-G(t1/2 = 16.5 h)

Incubation Time (h)

% R

emai

ning

BA

FIG. 9. A, time-dependent formation of MFA-SG and I-SG ob-tained from separate incubations of MFA-SCoA (1 �M), MFA-1-�-O-G (1 �M), and I-SCoA (1 �M) with GSH (10 mM) inpotassium phosphate buffer (0.1 M, pH 7.4, 37°C). B, time-depen-dent degradation of MFA-1-�-O-G and MFA-SCoA in correspond-ing incubations in buffer in the absence of GSH. Values are ex-pressed as the mean � S.D. from three incubations per time point.

TABLE 1

Effect of inhibitors of MFA-1-�-O-G and MFA-SCoA formation on the production of MFA-SG in incubations of MFA (100 �M) with rat hepatocytes (2 million cells/ml, 10-min incubation time period)

Values are expressed as the mean � S.D. from triplicate incubations.

InhibitorMFA-1-�-O-G MFA-SCoA MFA-SG

Concentration % Control Concentration % Control Concentration % Control

nM nM nM

Control 619 � 158 9.1 � 4.5 334 � 76(�)-Borneol, 1000 �M 55.1 � 7 8.9 � 1.2 8.8 � 1.0 97 � 11 387 � 22.6 116 � 6.8Lauric acid, 1000 �M 198 � 15 32 � 2.4 3.8 � 0.2 42 � 1.7 113 � 11.6 33.9 � 3.5

1524 GRILLO ET AL.

at ASPE

T Journals on M

ay 15, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 11: DMD 40:1515–1526, 2012 Metabolic Activation of Mefenamic ...dmd.aspetjournals.org/content/dmd/40/8/1515.full.pdf · Metabolic Activation of Mefenamic Acid ... The fenamate-type

with the time course of MFA-SCoA formation, where the Tmax ofMFA-SG formation occurred at 10 min of incubation (Fig. 5A) andthe Tmax for MFA-SCoA formation occurred at the 30-min incubationtime point. From these results alone, we proposed that MFA-SCoAwas not solely responsible for the transacylation of GSH leading toMFA-SG formation in rat hepatocytes. As expected, the time courseof MFA-1-�-O-G formation was not consistent with the time coursefor MFA-SG formation, where MFA-1-�-O-G increased in a time-dependent fashion throughout the entire incubation period, reaching1560 nM after 60 min of incubation (Fig. 5A), and MFA-SG concen-tration at 60 min decreased by �7-fold from its Cmax at 10 min. Thislack of acyl glucuronide/S-acyl-SG time-course correlation has beenobserved before from similar studies with ibuprofen (Grillo and Hua,2008), flunoxaprofen (Grillo et al., 2010), and diclofenac (Grillo et al.,2003a). We do not believe that the decrease in MFA-SG concentrationafter the 10-min time point was due to a decrease in intracellular GSHconcentration, because the method used for the incubation of rathepatocytes allowed for a maintained GSH concentration up to 3 h ofincubation (Nakagawa and Moldeus, 1998). Rather, we propose thatthe decrease in MFA-SG concentration observed after the 10-min timepoint occurred because of a decrease in the amount of reactivetransacylating metabolite formed after the 10-min time point. Corre-sponding studies with human hepatocytes showed that MFA-SG for-mation (Fig. 7) occurred at a 400-fold lower Cmax compared withincubations with rat hepatocytes (Fig. 6A). We currently do notunderstand the reason for the dramatic species difference in MFA-SGformation; however, similar studies with diclofenac showed no spe-cies difference, where the formation of D-SG was shown to reachmaximum concentrations of 1 and 0.8 nM after 4 min in incubationswith rat and human hepatocytes, respectively (Grillo et al., 2003).

Results from MFA concentration-dependent studies in rat hepato-cytes (Fig. 6B) showed that the 100-�M MFA concentration wheremaximum MFA-SG also occurred did not coincide with the Cmax ofMFA-SCoA formation (15.6 �M MFA), again demonstrating a lackof correlation of MFA-SG formation with MFA-CoA formation alone.

S-Acyl-GSH thioesters are degraded in incubations with rat hepa-tocytes to the corresponding free acids (Grillo, 2011). The degradationrates of MFA-SG, I-SG, and D-SG in rat hepatocytes were examinedin the present studies to determine whether MFA-SG might be hy-drolyzed at a significantly slower rate. We hypothesized that thesubstantially higher Cmax of MFA-SG in MFA-treated rat hepato-cytes, relative to other carboxylic acids examined to date, may havebeen due to a lower degradation rate. Results indicated that MFA-SG,I-SG, and D-SG were degraded with half-life values of 8.5, 2.0, and0.4 min, respectively (Fig. 8). From these data, we propose that the4.3-fold lower MFA-SG degradation rate did not contribute to the

�100-fold increase in MFA-SG concentration measured in rat hepa-tocyte incubations with MFA relative to (R)-ibuprofen in the presentexperiments.

S-Acyl-GSH thioesters are also known to be degraded to the cor-responding free acids by GST-mediated hydrolysis (Ibarra et al.,2003). Thus, we determined that MFA-SG and I-SG were relativelystable toward rat liver GST and degraded by only 10 to 11% after 30min, whereas D-SG was completely hydrolyzed after 30 min ofincubation (Fig. 10). Because MFA-SG and I-SG have similar stabil-ities in incubations with GST, the differences in their respectiveS-acyl-GSH formation in incubations with rat hepatocytes may not bedue to differences in rates of degradation catalyzed by intracellularGST.

Results from inhibition studies in rat hepatocytes where MFA-SCoAand MFA-1-�-O-G formation was inhibited by coincubation withlauric acid and (�)-borneol, respectively, showed that inhibition ofS-acyl-CoA formation led to a corresponding inhibition ofMFA-SG formation, whereas almost complete inhibition of acylglucuronidation had no effect on MFA-SG formation (Table 1).These results are consistent with results from similar studies withibuprofen and flunoxaprofen (Grillo and Hua, 2008; Grillo et al.,2010) and point to a general lack of importance for acyl glucuronidesmediating the transacylation of GSH. Potentially, metabolism of MFAby acyl-CoA synthetase(s) leads to a buildup of an intermediateMFA-acyl-adenylate. Xenobiotic and endogenous carboxylic acidshave been shown to be converted to chemically reactive mixed anhy-dride adenosine 5-monophosphate-linked intermediates detected dur-ing the formation of their corresponding S-acyl-CoA thioesters (Maoet al., 1992; Hall and Quan, 1994; Menzel et al., 1994; Ikegawa et al.,1999). Acyl-adenylate intermediates of endogenous bile acids, such ascholic acid (Ikegawa et al., 1999), are known to be reactive acylatingderivatives that undergo transacylation-type reactions with the aminogroup of taurine, with peptides and proteins, and with the thiol groupof GSH, leading to the formation of bile acid S-acyl-GSH conjugates(Goto et al., 2001).

In summary, from these data we propose that the formation ofMFA-SG is not mediated by MFA-1-�-O-G, and not solely by reac-tion with MFA-SCoA, but is also mediated by other MFA-linkedreactive acylating species that have not yet been identified. Investi-gations on the ability of MFA to form a reactive acyl-adenylateintermediate that might mediate the acylation of GSH in incubationswith rat and human hepatocytes are ongoing.

Acknowledgments

We thank Dr. Raju Subramanian (Pharmacokinetics and Drug Metabolism,Amgen, Thousand Oaks, CA) for the synthesis and NMR characterization ofmefenamyl-1-�-O-acyl glucuronide and Craig Uyeda and Stacy Fide (Phar-macokinetics and Drug Metabolism, Amgen, South San Francisco, CA) forassistance in the preparation of rat hepatocytes and rat bile collection studies.

Authorship Contributions

Participated in research design: Grillo.Conducted experiments: Grillo, Tadano Lohr, and Wait.Contributed new reagents or analytic tools: Grillo.Performed data analysis: Grillo, Tadano Lohr, and Wait.Wrote or contributed to the writing of the manuscript: Grillo.

References

Baba A and Yoshioka T (2006) Synthesis of 1-�-O-acyl glucuronides of diclofenac, mefenamicacid and (S)-naproxen by the chemo-selective enzymatic removal of protecting groups fromthe corresponding methyl acetyl derivatives. Org Biomol Chem 4:3303–3310.

Baillie TA (2008) Metabolism and toxicity of drugs. Two decades of progress in industrial drugmetabolism. Chem Res Toxicol 21:129–137.

Incubation Time (min)

[Acy

l-SG

add

uct],µM

0 5 10 15 20 25 300.0

0.2

0.4

0.6

0.8

1.0

1.2

D-SG, - GSTD-SG, + GSTI-SG, - GSTI-SG, + GSTMFA-SG, - GSTMFA-SG, + GST

FIG. 10. Time-dependent degradation of MFA-SG, I-SG, and D-SG (1 �M) inpotassium phosphate buffer (0.1 M, pH 7.4, 37°C) in the presence or absence ofGST (3 units/ml). Values are expressed as the average of two incubations per timepoint.

1525MEFENAMIC ACID-S-ACYL-GLUTATHIONE FORMATION

at ASPE

T Journals on M

ay 15, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 12: DMD 40:1515–1526, 2012 Metabolic Activation of Mefenamic ...dmd.aspetjournals.org/content/dmd/40/8/1515.full.pdf · Metabolic Activation of Mefenamic Acid ... The fenamate-type

Baillie TA and Davis MR (1993) Mass spectrometry in the analysis of glutathione conjugates.Biol Mass Spectrom 22:319–325.

Boelsterli UA (2002) Xenobiotic acyl glucuronides and acyl CoA thioesters as protein-reactivemetabolites with the potential to cause idiosyncratic drug reactions. Curr Drug Metab3:439–450.

Boelsterli UA, Zimmerman HJ, and Kretz-Rommel A (1995) Idiosyncratic liver toxicity ofnonsteroidal antiinflammatory drugs: molecular mechanisms and pathology. Crit Rev Toxicol25:207–235.

Chan JC, Lai FM, and Critchley JA (1991) A case of Stevens-Johnson syndrome, cholestatichepatitis and haemolytic anaemia associated with use of mefenamic acid. Drug Saf 6:230–234.

Glazko AJ (1966) Experimental observations on flufenamic, mefenamic and meclofenamic acids.3. Metabolic disposition. Ann Phys Med i (Suppl):23–36.

Goto J, Nagata M, Mano N, Kobayashi N, Ikegawa S, and Kiyonami R (2001) Bile acid acyladenylate: a possible intermediate to produce a protein-bound bile acid. Rapid Commun MassSpectrom 15:104–109.

Grillo MP (2011) Drug-S-acyl-glutathione thioesters: synthesis, bioanalytical properties, chem-ical reactivity, biological formation and degradation. Curr Drug Metab 12:229–244.

Grillo MP and Benet LZ (2002) Studies on the reactivity of clofibryl-S-acyl-CoA thioester withglutathione in vitro. Drug Metab Dispos 30:55–62.

Grillo MP and Hua F (2003) Identification of zomepirac-S-acyl-glutathione in vitro in incuba-tions with rat hepatocytes and in vivo in rat bile. Drug Metab Dispos 31:1429–1436.

Grillo MP and Hua F (2008) Enantioselective formation of ibuprofen-S-acyl-glutathione in vitroin incubations of ibuprofen with rat hepatocytes. Chem Res Toxicol 21:1749–1759.

Grillo MP and Lohr MT (2009) Covalent binding of phenylacetic acid to protein in incubationswith freshly isolated rat hepatocytes. Drug Metab Dispos 37:1073–1082.

Grillo MP, Hua F, Knutson CG, Ware JA, and Li C (2003a) Mechanistic studies on thebioactivation of diclofenac: identification of diclofenac-S-acyl-glutathione in vitro in incuba-tions with rat and human hepatocytes. Chem Res Toxicol 16:1410–1417.

Grillo MP, Knutson CG, Sanders PE, Waldon DJ, Hua F, and Ware JA (2003b) Studies on thechemical reactivity of diclofenac acyl glucuronide with glutathione: identification of diclofe-nac-S-acyl-glutathione in rat bile. Drug Metab Dispos 31:1327–1336.

Grillo MP, Wait JCM, Tadano-Lohr M, Khera S, and Benet LZ (2010) Stereoselective flunox-aprofen-S-acyl-glutathione thioester formation mediated by acyl-CoA formation in rat hepa-tocytes. Drug Metab Dispos 38:133–142.

Hall SD and Quan X (1994) The role of coenzyme A in the biotransformation of 2-arylpropionicacids. Chem Biol Interact 90:235–251.

Hinchman CA and Ballatori N (1990) Glutathione-degrading capacities of liver and kidney indifferent species. Biochem Pharmacol 40:1131–1135.

Ibarra C, Grillo MP, Lo Bello M, Nucettelli M, Bammler TK, and Atkins WM (2003) Explo-ration of in vitro pro-drug activation and futile cycling by glutathione S-transferases: thiol esterhydrolysis and inhibitor maturation. Arch Biochem Biophys 414:303–311.

Ikegawa S, Ishikawa H, Oiwa H, Nagata M, Goto J, Kozaki T, Gotowda M, and Asakawa N(1999) Characterization of cholyl-adenylate in rat liver microsomes by liquid chromatography/electrospray ionization-mass spectrometry. Anal Biochem 266:125–132.

Khera S, Grillo M, Schnier P, and Hollis S (2010) Application of diffusion-edited NMRspectroscopy for the structural characterization of drug metabolites in mixtures. J PharmBiomed Anal 51:164–169.

Knights KM (1998) Role of hepatic fatty acid:coenzyme A ligases in the metabolism ofxenobiotic carboxylic acids. Clin Exp Pharmacol Physiol 25:776–782.

Li C, Benet LZ, and Grillo MP (2002) Studies on the chemical reactivity of 2-phenylpropionicacid 1-O-acyl glucuronide and S-acyl-CoA thioester metabolites. Chem Res Toxicol 15:1309–1317.

Mao LF, Millington DS, and Schulz H (1992) Formation of a free acyl adenylate during the

activation of 2-propylpentanoic acid. Valproyl-AMP: a novel cellular metabolite of valproicacid. J Biol Chem 267:3143–3146.

McGurk KA, Remmel RP, Hosagrahara VP, Tosh D, and Burchell B (1996) Reactivity ofmefenamic acid 1-O-acyl glucuronide with proteins in vitro and ex vivo. Drug Metab Dispos24:842–849.

Menzel S, Waibel R, Brune K, and Geisslinger G (1994) Is the formation of R-ibuprofenyl-adenylate the first stereoselective step of chiral inversion? Biochem Pharmacol 48:1056–1058.

Moldeus P, Hogberg J, and Orrenius S (1978) Isolation and use of liver cells. Methods Enzymol52:60–71.

Nakagawa Y and Moldeus P (1998) Mechanism of p-hydroxybenzoate ester-induced mitochon-drial dysfunction and cytotoxicity in isolated rat hepatocytes. Biochem Pharmacol 55:1907–1914.

Nelson DL and Cox MM (2005) Lehninger’s Principles of Biochemistry, 4th ed, WH Freemanand Company, New York.

Nelson SD (2001) Molecular mechanisms of adverse drug reactions. Curr Ther Res 62:885–899.Olsen J, Li C, Skonberg C, Bjørnsdottir I, Sidenius U, Benet LZ, and Hansen SH (2007) Studies

on the metabolism of tolmetin to the chemically reactive acyl-coenzyme A thioester interme-diate in rats. Drug Metab Dispos 35:758–764.

Robertson CE, Ford MJ, Van Someren V, Dlugolecka M, and Prescott LF (1980) Mefenamicacid nephropathy. Lancet 2:232–233.

Sato J, Yamane Y, Ito K, and Bando H (1993) Structures of mefenamic acid metabolites fromhuman urine. Biol Pharm Bull 16:811–812.

Shore LJ, Fenselau C, King AR, and Dickinson RG (1995) Characterization and formation of theglutathione conjugate of clofibric acid. Drug Metab Dispos 23:119–123.

Sidenius U, Skonberg C, Olsen J, and Hansen SH (2004) In vitro reactivity of carboxylicacid-CoA thioesters with glutathione. Chem Res Toxicol 17:75–81.

Skonberg C, Olsen J, Madsen KG, Hansen SH, and Grillo MP (2008) Metabolic activation ofcarboxylic acids. Expert Opin Drug Metab Toxicol 4:425–438.

Somchit N, Sanat F, Gan EH, Shahrin IA, and Zuraini A (2004) Liver injury induced by thenon-steroidal anti-inflammatory drug mefenamic acid. Singapore Med J 45:530–532.

Walker GS, Atherton J, Bauman J, Kohl C, Lam W, Reily M, Lou Z, and Mutlib A (2007)Determination of degradation pathways and kinetics of acyl glucuronides by NMR spectros-copy. Chem Res Toxicol 20:876–886.

Watkins JB and Klaassen CD (1982) Effect of inducers and inhibitors of glucuronidation on thebiliary excretion and choleretic action of valproic acid in the rat. J Pharmacol Exp Ther220:305–310.

Winder CV, Wax J, Scotti L, Scherrer RA, Jones EM, and Short FW (1962) Anti-inflammatory,antipyretic and antinociceptive properties of N-(2,3-xylyl)anthranilic acid (mefenamic acid).J Pharmacol Exp Ther 138:405–413.

Xiaotao Q and Hall SD (1993) Modulation of enantioselective metabolism and inversion ofibuprofen by xenobiotics in isolated rat hepatocytes. J Pharmacol Exp Ther 266:845–851.

Zheng J, Ma L, Xin B, Olah T, Humphreys WG, and Zhu M (2007) Screening and identificationof GSH-trapped reactive metabolites using hybrid triple quadruple linear ion trap massspectrometry. Chem Res Toxicol 20:757–766.

Zia-Amirhosseini P, Harris RZ, Brodsky FM, and Benet LZ (1995) Hypersensitivity to non-steroidal anti-inflammatory drugs. Nat Med 1:2–4.

Address correspondence to: Mark P. Grillo, Pharmacokinetics and DrugMetabolism, Amgen Inc., 1120 Veterans Blvd., South San Francisco, CA 94080.E-mail: [email protected]

1526 GRILLO ET AL.

at ASPE

T Journals on M

ay 15, 2018dm

d.aspetjournals.orgD

ownloaded from