Disposition Mechanisms of Raloxifene in the Human Intestinal Caco-2 Model

Similar documents
The Pharmacokinetics of Raloxifene and Its Interaction with Apigenin in Rat

Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Pullman, Washington

JPET Fast Forward. Published on May 5, 2004 as DOI: /jpet JPET #68403

DISPOSITION OF FLAVONOIDS VIA RECYCLING: COMPARISON OF INTESTINAL VERSUS HEPATIC DISPOSITION

Biopharmaceutics Drug Disposition Classification System (BDDCS) and Its Application in Drug Discovery and Development

Biopharmaceutics Drug Disposition Classification System (BDDCS) --- Its Impact and Application

Pharmacokinetics of Drugs. Assistant Prof. Dr. Najlaa Saadi PhD Pharmacology Faculty of Pharmacy University of Philadelphia

GRADIENT RP-HPLC METHOD FOR THE DETRMINATION OF PURITY AND ASSAY OF RALOXIFENE HYDROCHLORIDE IN BULK DRUG

ABSORPTION AND METABOLISM OF FLAVONOIDS IN THE CACO-2 CELL CULTURE MODEL AND A PERUSED RAT INTESTINAL MODEL

Define the terms biopharmaceutics and bioavailability.

Exploiting BDDCS and the Role of Transporters

TENOFOVIR TABLETS: Final text for addition to The International Pharmacopoeia (June 2010)

Application Note. Authors. Abstract. Food

The TC7 Cell Monolayer is a Valuable in vitro Intestinal Epithelial Model for Membrane Permeability Screening

Oroxcell Percutaneous and intestinal absorption

ARTESUNATE TABLETS: Final text for revision of The International Pharmacopoeia (December 2009) ARTESUNATI COMPRESSI ARTESUNATE TABLETS

Standardization of a LC/MS/MS Method for the Determination of Acyl Glucuronides and Their Isomers

Interaction of baicalin with transporters

Determination of Tetracyclines in Chicken by Solid-Phase Extraction and High-Performance Liquid Chromatography

4. ABSORPTION. Transport mechanisms. Absorption ABSORPTION MECHANISMS. Active transport. Active transport uses metabolic energy

RITONAVIRI COMPRESSI RITONAVIR TABLETS. Final text for addition to The International Pharmacopoeia (July 2012)

Click to edit Master title style

CYCLOSERINI CAPSULAE - CYCLOSERINE CAPSULES (AUGUST 2015)

LC-MS/MS Method for the Determination of Raloxifene and its Glucuronide Metabolites from Human Plasma Using SPE Micro Elution

For the quantitative measurement of ATP Synthase Specific activity in samples from Human, Rat and Cow

RP-HPLC Analysis of Temozolomide in Pharmaceutical Dosage Forms

PDF hosted at the Radboud Repository of the Radboud University Nijmegen

DRUG ELIMINATION II BILIARY EXCRETION MAMMARY, SALIVARY AND PULMONARY EXCRETION

HEK293 cells transfected with human MATE1, MATE2-K, or vector control were established by

Chapter 4. Drug Biotransformation

Determination of propranolol in dog plasma by HPLC method

SUPPLEMENTARY INFORMATION

Pelagia Research Library

ab ATP Synthase Enzyme Activity Microplate Assay Kit

High-density Lipoprotein Cholesterol (HDL-C) Assay Kit

Summary and general discussion

Unit 2b: EXCRETION OF DRUGS. Ms.M.Gayathri Mpharm (PhD) Department of Pharmaceutics Krishna Teja Pharmacy college Subject code: 15R00603 (BPPK)

DEVELOPMENT OF RP-HPLC METHOD FOR ESTIMATION OF DROTAVERINE HYDROCHLORIDE IN PHARMACEUTICAL FORMULATIONS

Determination of β2-agonists in Pork Using Agilent SampliQ SCX Solid-Phase Extraction Cartridges and Liquid Chromatography-Tandem Mass Spectrometry

III. TOXICOKINETICS. Studies relevant to the toxicokinetics of inorganic chloramines are severely

Chunying Gao, Hongjian Zhang, Zitao Guo, Tiangeng You, Xiaoyan Chen, and Dafang Zhong

Application Note. Abstract. Authors. Pharmaceutical

Improvement of Intracellular Glutathione Content. in Baker s Yeast. for Nutraceutical Application

WHY... 8/21/2013 LEARNING OUTCOMES PHARMACOKINETICS I. A Absorption. D Distribution DEFINITION ADME AND THERAPEUIC ACTION

ZIDOVUDINE, LAMIVUDINE AND ABACAVIR TABLETS Draft proposal for The International Pharmacopoeia (September 2006)

A HIGH PERFORMANCE LIQUID CHROMATOGRAPHIC ASSAY FOR LERCANIDIPINE HYDROCHLORIDE

ISSN: ; CODEN ECJHAO E-Journal of Chemistry 2011, 8(3),

The ADME properties of most drugs strongly depends on the ability of the drug to pass through membranes via simple diffusion.

Relative Measurement of Zeaxanthin Stereoisomers by Chiral HPLC

Transporters DDI-2018

Supporting Information for:

Pharmacokinetics of ibuprofen in man. I. Free and total

Compliance. Should you have any questions, please contact Behnaz Almasi, Associate Scientific Liaison ( or

retardation in infants. A wide variety of analytical methods for the analysis of

ASSAY OF SPHINGOMYELINASE ACTIVITY

Total Phosphatidic Acid Assay Kit

INTRODUCTION TO PHARMACOKINETICS

IN VITRO ANTICANCER ACTIVITY OF FLOWER EXTRACTS OF COUROUPITA GUIANENSIS

Short Communication. Abstract. Introduction

An Investigative Study of Reactions Involving Glucosinolates and Isothiocyanates

Biopharmaceutics Classification System: Defining a Permeability Class

Bioequivalence Studies of Two Formulations of Famciclovir Tablets by HPLC Method

Chapter 2 Transport Systems

Pharmacokinetics in Drug Development. Edward P. Acosta, PharmD Professor & Director Division of Clinical Pharmacology Director, CCC PK/PD Core

International Journal of Pharma and Bio Sciences DEVELOPMENT AND VALIDATION OF RP-HPLC METHOD FOR THE ESTIMATION OF STRONTIUM RANELATE IN SACHET

Application Note Soy for Isoflavones by HPLC. Botanical Name: Glycine max L. Common Names: Parts of Plant Used: Beans.

Development and Validation for Simultaneous Estimation of Sitagliptin and Metformin in Pharmaceutical Dosage Form using RP-HPLC Method

This revision also necessitates a change in the table numbering in the test for Organic Impurities.

STANDARD OPERATING PROTOCOL (SOP)

Leslie Z. Benet, PhD. Professor of Bioengineering and Therapeutic Sciences Schools of Pharmacy and Medicine University of California San Francisco

Hyderabad, India. Department of Pharmaceutical Chemistry, Glocal University, Saharanpur, India.

Application Note. Agilent Application Solution Analysis of ascorbic acid, citric acid and benzoic acid in orange juice. Author. Abstract.

D9G : Oro-Mucosal Dosage Forms Development Background Paper

Evaluation of the first-pass glucuronidation of selected flavones in gut by Caco-2 monolayer model.

IAM Chromatography. Introduction

Supplementary Material (ESI) for Chemical Communications This journal is (c) The Royal Society of Chemistry 2008

Basic Concepts in Pharmacokinetics. Leon Aarons Manchester Pharmacy School University of Manchester

Controlling ADME through Chemical Design. Marty Mulvihill Chris Vulpe

National Standard of the People s Republic of China. National food safety standard. Determination of pantothenic acid in foods for infants and

1. Gastric Emptying Time Anatomically, a swallowed drug rapidly reaches the stomach. Eventually, the stomach empties its content in the small

Flupyradifurone. HPLC Method

METHOD DEVELOPMENT AND VALIDATION BY RP-HPLC FOR ESTIMATION OF ZOLPIDEM TARTARATE

Analysis of Amino Acids Derived Online Using an Agilent AdvanceBio AAA Column

CHAPTER INTRODUCTION OF DOSAGE FORM AND LITERATURE REVIEW

1 Introduction: The Why and How of Drug Bioavailability Research

Hepatocyte Metabolic Kinetics Assays

CYCLOSERINE Proposal for revision of The International Pharmacopoeia (August 2012)

Pilot experiments to investigate the glucuronidation of axitinib with human liver microsomes.

Saliva Versus Plasma Bioequivalence of Rusovastatin in Humans: Validation of Class III Drugs of the Salivary Excretion Classification System

THIN LAYER CHROMATOGRAPHY

cationic molecule, paracellular diffusion would be thought of as its primary mode of transport across epithelial cells.

Permeation prediction of M using the parallel artificial membrane permeability assay.

ARTENIMOLUM ARTENIMOL. Adopted revised text for addition to The International Pharmacopoeia

Membrane Transport. Anatomy 36 Unit 1

Comprehensive Study of SLE as a Sample. Preparation Tool for Bioanalysis

INTERNATIONAL PHARMACOPOEIA MONOGRAPH ON LAMIVUDINE TABLETS

EVALUATION OF DRUG-DRUG INTERACTION POTENTIAL BETWEEN SACUBITRIL/VALSARTAN (LCZ696) AND STATINS USING A PHYSIOLOGICALLY- BASED PHARMACOKINETIC MODEL

Evaluation of the Percutaneous Absorption of Tramadol, In Lipoderm, Into Inner Ear Feline Skin, In Vitro, Using the Franz Skin Finite Dose Model

What location in the gastrointestinal (GI) tract has tight, or impermeable, junctions between the epithelial cells?

Transcription:

0022-3565/04/3101-376 385$20.00 THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS Vol. 310, No. 1 Copyright 2004 by The American Society for Pharmacology and Experimental Therapeutics 0/1153839 JPET 310:376 385, 2004 Printed in U.S.A. Disposition Mechanisms of Raloxifene in the Human Intestinal Caco-2 Model Eun Ju Jeong, Huimin Lin, and Ming Hu Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Pullman, Washington Received December 5, 2003; accepted March 11, 2004 ABSTRACT The purpose of this study was to determine the mechanisms responsible for transport of raloxifene and its hydrophilic conjugates. Human intestinal Caco-2 cell culture model and Caco-2 cell lysate were used for the studies. The results indicated that absorptive permeability (P AB ) of raloxifene was lower than its secretory permeability (P BA ). As the concentration increased, the efflux ratio (P BA /P AB ) decreased, but P AB increased. P AB was also increased in the presence of verapamil and cyclosporine A, two P-glycoprotein inhibitors. Raloxifene was extensively metabolized into sulfated and glucuronidated conjugates. The extent of metabolism or clearance was decreased as the concentration increased from 3.4 (96%) to 30 (22%) M. Multidrug resistance-related protein inhibitors MK- 571 (C 26 H 26 ClN 2 O 3 S 2 ) and leukotriene C 4 significantly decreased (maximal 80%) apical efflux of both conjugates. They This study was supported by National Institutes of Health Grant CA 87779. Article, publication date, and citation information can be found at http://jpet.aspetjournals.org. DOI: 10.1124/jpet.103.063925. also significantly decreased (maximal 85%) basolateral efflux of glucuronides but not sulfates. On the other hand, organic anion transporter (OAT) inhibitor estrone sulfate and estrone glucuronide significantly decreased (maximal 50%) the efflux of sulfate from both sides but had variable effects on glucuronide efflux. Inhibition of conjugate efflux with the OAT inhibitor estrone sulfate was concentration dependent, which resulted in increased transport of intact raloxifene (maximal 90%). This increase in raloxifene transport was also observed in the presence of another OAT inhibitor estrone glucuronide (70%). In conclusion, this is the first report that inhibition of an efflux transporter responsible for the transport of metabolites can result in increase in the transport of the intact compound. It also provides additional explanation why raloxifene has low bioavailability but a long half-life. Raloxifene, a selective estrogen receptor modulator (Colacurci et al., 2003; Duschek et al., 2003), is used for the treatment of osteoporosis. Raloxifene blocks the adverse effects of estrogen in breast tissues and uterine endometrium while mimicking the beneficial estrogen effects on bone and lipid metabolism (MacGregor and Jordan, 1998; Scott et al., 1999). Data from both animal and human studies demonstrated that raloxifene has minimal effects on the uterus and caused no significant changes in the histological appearance of the endometrium (Scott et al., 1999 and references therein). Raloxifene is being considered as a chemopreventive agent for breast cancer (Delmas et al., 1997). Short-term clinical trials showed that raloxifene did not increase the risk of breast cancer and long-term multicenter trials are currently ongoing (Jordan and Morrow, 1999; Scott et al., 1999). Raloxifene is reported by its manufacturer to be rapidly absorbed after oral administration, but its absolute bioavailability is only 2%. This poor bioavailability is thought to be the result of extensive phase II metabolism, since it is absorbed rapidly and approximately 60% of dose was absorbed after oral dosing (Hochner-Celnikier, 1999). According to its manufacturer, raloxifene exhibits linear pharmacokinetics with high within-subject variability (approximately 30% CV) (Hochner-Celnikier, 1999). It undergoes extensive phase II biotransformation, primarily glucuronidation, but was not metabolized by the cytochrome P450s (Eli Lilly, 1998). Raloxifene has a plasma elimination half-life of approximately 27 h, which is attributed to its reversible systemic metabolism and significant enterohepatic cycling (Eli Lilly, 1998). The major human intestinal enzymes responsible for its phase II conjugation have been shown to be UGT1A8 and UGT1A10 (Kemp et al., 2002). Although a variety of pharmacokinetic information has been provided by its manufacturer (as summarized previously), mechanisms responsible for absorption of the raloxifene and excretion of raloxifene conjugates have not been reported. Studies have not established the dynamic relationship among absorption, metabolism, and excretion of ralox- ABBREVIATIONS. HBSS, Hanks balanced salt solution; HPLC, high-performance liquid chromatography; ANOVA, analysis of variance; B BA, secretory (basolateral to apical) rates of transport; B AB, absorptive (apical to basolateral) rates of transport; MRP, multidrug resistance-related protein; P BA, secretory permeability; P AB, absorptive permeability; OAT, organic anionic transporter; MK-571, C 26 H 26 ClN 2 O 3 S 2. 376

ifene in the intestine, which deserves more attention since it is likely to be the main site of presystemic clearance (Kemp et al., 2002). In the present research, we focused on the study of absorption/excretion pathways responsible for intestinal disposition of raloxifene using the human intestinal Caco-2 cell model employing the cloned TC7 variant. We chose Caco-2 TC7 cells, one of the cloned Caco-2 variants, for increased homogeneity and stability of cell population (Caro et al., 1995; Ranaldi et al., 2003). TC variant exhibits morphological characteristics similar to those of the parental Caco-2 cells (Gres et al., 1998) and shows similar expression of MDR1 and MRP1 5 as human jejunal biopsies (Pfrunder et al., 2003). It has been used by many investigators to study human intestinal disposition (Hu et al., 1999, 2000, 2003; Sabolovic et al., 2000; Pontier et al., 2001; Bohets et al., 2001). Therefore, the objective of the present study was to determine whether enteric recycling and other intestinal disposition events could help explain why raloxifene has a low bioavailability but a long half-life. The process of enteric recycling, which was first proposed to explain the disposition of flavonoids (Liu and Hu, 2002), is analogous to enterohepatic recycling except the phase II conjugates are produced and excreted by the enterocytes instead of hepatocytes. Materials and Methods Materials. Cloned Caco-2 cells (TC7) were a kind gift from Dr. Moniqué Rousset (Institute National de la Santé et de la Recherche zu178, Villejuit, France). Raloxifene was extracted from Evista tablet (Eli Lilly & Co., Indianapolis, IN) using 100% ethanol, and concentration was then verified by using raloxifene hydrochloride purchased from National Cancer Institute Chemical Standard Repository managed by Midwest Research Institute (Kansas City, MO). -Glucuronidase, sulfatase, Hank s balanced salt solution (HBSS; powder form), cyclosporin A, verapamil, estrone glucuronide, and estrone sulfate were purchased from Sigma-Aldrich (St. Louis, MO). Leukotriene C 4 and MK-571 were purchased from BIOMOL Research Laboratories (Plymouth Meeting, PA). All other materials were analytical grade or better and used as received. Cell Culture. Cell culture conditions for growing Caco-2 cells have been described previously (Hu et al., 1994a, 1994b; Liu and Hu, 2002; Chen et al., 2003). The seeding density was 100,000 cells/cm 2 (4.2 cm 2 /monolayer), and Dulbecco s modified Eagle s medium supplemented with 10% fetal bovine serum was used as growth media. Quality control criteria were the same as described previously (Hu et al., 1994a, 1994b). Cell monolayers from 19 to 22 days past seeding were used for the experiments. Transport Experiments in the Caco-2 Cell Model. The protocols for performing transport experiments were similar to those described previously (Liu and Hu, 2002). In brief, the cell monolayers were washed three times with 37 C HBSS (ph 7.4). The transepithelial electrical resistance values were measured, and those with the values less than 420 /cm 2 were discarded. Various concentrations (ranging from 1.5 30 M) of raloxifene were loaded at the apical or basolateral side of the Caco-2 cell monolayer, and the concentration of raloxifene and its metabolites at both sides were measured by HPLC. Inhibitors, when used, were loaded at the apical, basolateral, or both sides of the monolayer. Six samples (650 l each) were taken at different times (0, 1, 2, 3, 4, and 7 or 24 h after incubation) from both donor and receiver side (total volume of each chamber was 2.5 ml), and the same volume of donor solution (containing raloxifene) or transport medium was replaced after each sampling. Forty-five microliters of internal standard solution (100 M testosterone in methanol) was immediately added to 200 l of samples to stabilize them until analysis. Raloxifene Transport in the Caco-2 Model 377 Extraction of Raloxifene from Caco-2 Cell Monolayers. After the transport experiments, cells were washed three times with ice-cold saline solution, and the polycarbonate membranes containing cells were removed from the insert (4.2 cm 2 ; Nalge Nunc International, Naperville, IL). One milliliter of HBSS (ph 7.4) was added to each membrane, and the solution was frozen (in liquid nitrogen) and thawed (in 37 C water bath) three times to disrupt cell membranes. After centrifugation at 13,000 rpm for 8 min, 0.5 ml of supernatant was collected. Methanol (0.5 ml) was added to the remaining suspension, and the tubes were centrifuged again to extract the remaining raloxifene. Preparation of Caco-2 Cell Lysate. After six mature (19 22 days post-seeding) Caco-2 cell monolayers were washed twice with 3 ml of 37 C HBSS (ph 7.4), they were cut out together with the porous polycarbonate membranes, immersed into 6 ml of 50 mm potassium phosphate buffer (ph 7.4), and sonicated in an ice bath (4 C) for 30 min as described previously (Hu et al., 2003). Afterward, the cell lysate was centrifuged at 1000 rpm for 5 min to remove the polycarbonate membrane. The protein concentration of the cell lysate was then determined using a commercial protein assay kit (Bio-Rad, Hercules, CA). Glucuronidation of Raloxifene in Caco-2 Cell Lysate. Glucuronidation of raloxifene by Caco-2 cell lysate was measured using procedures described previously (Hu et al., 2003). The cell lysate (final concentration 0.65 mg/ml) was mixed with magnesium chloride (0.88 mm), saccharolactone (4.4 mm), and alamethicin (0.022 mg/ml). Raloxifene or raloxifene plus estrone sulfate in 50 mm potassium phosphate buffer (ph 7.4) were then added. Uridine diphosphoglucuronic acid (3.5 mm) was added last to the reaction mixture (total volume 200 l), and the mixture was incubated in a 37 C shaking (200 rpm) water bath for 4 h. The reaction was stopped by the addition of 50 l solution of 94% acetonitrile/6% glacial acetic acid containing 100 M testosterone as the internal standard. Sulfation of Raloxifene in Caco-2 Cell Lysate. The cell lysate (final concentration of about 0.91 mg/ml) was mixed with raloxifene or raloxifene plus estrone sulfate in 50 mm potassium phosphate buffer (ph 7.4). The cofactor 3 -phosphoadenosine 5 -phosphosulfate (0.1 mm) was added last to the reaction mixture (total volume 200 l), and the mixture was incubated in a 37 C shaking (200 rpm) water bath for 4 h. The reaction was stopped by the addition of 50 l solution of 94% acetonitrile/6% glacial acetic acid containing 100 M testosterone as the internal standard. Hydrolysis of Raloxifene Metabolites by Hydrolases. A portion of 24-h samples of raloxifene (7 M) transport and metabolism experiments was extracted with methylene chloride to remove intact raloxifene. The remaining aqueous phase was incubated with glucuronidase (20 units per reaction) or sulfatase (0.5 unit per reaction) at 37 Cfor4htoreconvert conjugated raloxifene to raloxifene, which is used to identify the raloxifene metabolite peaks and amounts of metabolites (via reconversion) in HPLC chromatograms (Fig. 1). HPLC Analysis of Raloxifene and Its Conjugates. The HPLC conditions were as follows: system, Agilent 1090 controlled by Chemstation with a dioarray detector and an autosampler (Agilent Technologies, Palo Alto, CA); column, Aqua 5 m, 150 4.60 mm (Phenomenex, Torrance, CA); mobile phase A, 0.04% (v/v) phosphoric acid plus 0.06% (v/v) triethylamine in water (ph 2.8); mobile phase B, 100% acetonitrile; gradient, 0 to 3 min, 80% A, 3 to 22 min, 80% to 51.5% A, 22 to 25 min, 51.5% A, 25 to 26 min, 51.5% to 80% A, 26 to 28 min 80% A; wavelength, 288 nm for raloxifene and 254 nm for the internal standard; and injection volume, 200 l. The retention times for raloxifene, glucuronide, sulfate, and internal standard were 14.4, 9.7, 12.1, and 21.5 min, respectively (Fig. 1). Data Analysis. Rates of transport (B t ) were obtained using rate of change in concentration of transported raloxifene or its metabolites as a function of time and volume of the sampling chamber (V) (eq. 1). Permeability (P) across a cellular membrane was calculated using the rate of transport divided by the surface area (A) of the monolayer

378 Jeong et al. Fig. 1. Representative HPLC profiles of raloxifene and its metabolites. A, peaks of major metabolites (i.e., glucuronide and sulfate) after extracting raloxifene with methylene chloride from a 2-h Caco-2 incubation sample. B, disappearance of glucuronide peak and appearance of raloxifene peak after treatment with glucuronidase. C, reduction of sulfate peak and appearance of raloxifene peak after treatment with sulfatase. and the initial concentration of raloxifene at the loading side (C i ) (eq. 2). B t dc V (1) dt P B t AC i (2) Apparent metabolic clearance (CL) of raloxifene was calculated using rate of excretion (B ex ) divided by the initial concentration of raloxifene at the loading side (C i ) (eq. 3). CL B ex C i (3) This is based on the assumption that the rate of excretion is related to initial concentration of raloxifene. It is also based on practical consideration in that intracellular concentrations of metabolites (e.g., glucuronides) are sometimes too low to measure (see Results). Therefore, if the rate of excretion is always proportional to the initial concentration, the clearance will be a constant. Otherwise, the clearance will change as concentration changes. Statistical Analysis. One-way ANOVA or Student s t test (Microsoft Excel) was used to analyze the data. The prior level of significance was set at 5%, or P 0.05. Results Time Course of Transport and Metabolism of Raloxifene. We monitored amounts of raloxifene and its metabolites in the apical and basolateral media as a function of time for 4 h after apical or basolateral loading. The results indicated that amounts of raloxifene and its metabolites appeared in the receiver side increased linearly with time (Fig. 2, A and C). The amounts of metabolites in the donor side also increased linearly with time (Fig. 2, B and D), but the amount of raloxifene in the donor side did not change significantly (Fig. 2, B and D) since we replenished it after each sampling. Fig. 2. Transport and excretion of raloxifene and its metabolites in the Caco-2 model. Raloxifene (17 M) was added to the apical (AP) or basolateral (BL) side of the Caco-2 cell monolayer, and the amounts of raloxifene (circles), raloxifene glucuronide (squares), and raloxifene sulfate (triangles) in apical (open symbols and dotted lines) and basolateral (solid symbols and solid lines) were determined at 1, 2, 3, and 4 h after incubation as described under Materials and Methods. These symbols (e.g., solid for basolateral appearance, squares for glucuronide, and triangles for sulfate) are used throughout the paper. Each data point represents the mean of three determinations, and the error bars are S.D. of the mean. Each line represents linear regression of the data points. A, R 2 0.9000, 0.9942, and 0.9900 for raloxifene, glucuronide, and sulfate; B, R 2 0.9812 and 0.9805 for glucuronide and sulfate; C, R 2 0.9827, 0.9929, and 0.9902 for raloxifene, glucuronide, and sulfate; D, R 2 0.9972 and 0.9955 for glucuronide and sulfate, respectively.

Raloxifene Transport in the Caco-2 Model 379 The rates of raloxifene transport were determined with respect to the direction of transport (Table 1). Secretory (basolateral to apical) rates of transport (B BA ) were always higher than absorptive (apical to basolateral) rates of transport (B AB )(P 0.05) (Table 1). As concentration of raloxifene increased, the ratio of B BA to B AB decreased from 3.5 to 1.5. Pretreatment of Caco-2 cells with 17 M raloxifene to saturate the conjugating enzymes increased (P 0.05) the secretory rate of transport, but the extent of increase was small (Table 1). Effect of Raloxifene Concentration on the Transport of Raloxifene. Raloxifene has low absorptive permeability (P AB 0.39 10 6 4.1 10 6, 1.5 30 M) when compared with genistein (P AB 31 10 6,35 M) and apigenin (P AB 17 10 6,35 M) (Table 2), two compounds with extensive metabolism in the intestine (Chen et al., 2003; Hu et al., 2003). Permeability of raloxifene increased linearly with rising raloxifene concentration (Fig. 3A). On the other hand, the rate of transport increased with the square of concentration (Fig. 3B). Effect of Raloxifene Concentration on the Excretion of Its Metabolites. Significant amounts of raloxifene were metabolized into glucuronidated and sulfated metabolites during transport across the Caco-2 cell monolayers, with the sulfates being the major metabolites (Fig. 2; Table 2). The extent of metabolism decreased as the concentration of raloxifene increased (Table 2). The extensive metabolism of raloxifene at lower concentration can be demonstrated by its high clearance value. The clearance values for apical efflux of glucuronide and sulfate (at a raloxifene concentration of 7 M) were 10.04 1.37 and 26.57 1.12 l/h, respectively. For basolateral efflux, they were roughly equal, or about 11 l/h. Taken together, the total clearance values were 37.57 l/h for sulfates and 21 l/h for glucuronides, respectively. With a cellular water volume of about 3.66 l/mg protein, the total monolayer cellular water volume is about 3.66 l (Hu et al., 1994b). Therefore, the whole cellular sulfate volume is replaced about 10 times/h or once every 6 min, whereas the whole cellular glucuronide volume is replaced about 5 times/h or once every 12 min. Clearance of raloxifene glucuronide into both apical and basolateral sides were similar and both decreased significantly (P 0.05 according to one-way ANOVA) with an increase in raloxifene concentration (Fig. 4, A and B). Clearance of raloxifene sulfate, on the other hand, was polarized and mainly (up to 3.5 times more) to the apical side (P 0.05). Similar to clearance of glucuronides, both apical and basolateral clearance also decreased significantly (P 0.05 according to one-way ANOVA) with an increase in raloxifene concentration (Fig. 4, A and B). When comparing the clearance of these two conjugates from the same side of the cell monolayers, apical clearance of raloxifene sulfate was higher (2.7 23 times) than that of glucuronide at raloxifene concentration between 1.5 and 17 M(P 0.05) (Fig. 4A). However, this difference in clearance decreased as raloxifene concentration increased and eventually became minimal at 30 M. On the other hand, basolateral clearance of the two metabolites was similar at all the concentrations measured (Fig. 4B). Effect of Multidrug Resistance-Related Protein (MRP) and P-Glycoprotein Inhibitors on the Transport of Raloxifene and Excretion of Its Metabolites. MRP inhibitor MK-571 (50 M) and leukotriene C 4 (0.1 M) TABLE 1 Absorptive (apical to basolateral, B AB ) and secretory (basolateral to apical, B BA ) rate of raloxifene transport Raloxifene was added to apical or basolateral side of the Caco-2 cell monolayer. Rate of transport (B) values were calculated using first four data (time 1 4 h) in the amount transported versus time plot. Some cells were treated with 17 M raloxifene (both sides) for 24 h before the transport experiment. Data are presented as the mean S.D. of three determinations. Raloxifene Cell B AB B BA Ratio M nmol/h 17 Normal cell 0.498 0.094 0.757 0.098 1.52%* 7 Normal cell 0.088 0.036 0.301 0.011 3.44%* 7 Raloxifene, 24 h 0.093 0.032 0.334 0.016** 3.60%* *P 0.05 between B AB and B BA. **P 0.05 between normal and treated cell. TABLE 2 Absorptive permeability, amounts transported intact, and amounts of raloxifene metabolites formed after 2 h Drug was added to the apical side of the Caco-2 monolayer. Permeability values were calculated using first four data (time 1 4 h for raloxifene or time 0.5, 1, 1.5, and 2 h for apigenin and genistein) in the amount transported versus time plot. Amounts of metabolites in the apical (AP) and basolateral (BL) media after 2-h incubation were determined. Data are presented as the mean S.D. of three determinations. Compound Permeability Glucuronide Sulfate Total Amounts AP side BL side AP side BL side Parent Conjugates Metabolism: Parent/(Total) 10 7 cm/s nmol nmol % Raloxifene, 1.5 M UD b UD UD 0.419 0.052 UD Raloxifene, 3.4 M 3.88 0.83 c 0.136 0.007 0.159 0.018 0.635 0.040 0.172 0.033 0.041 1.102 96.4 Raloxifene, 6 M 7.87 2.05 c 0.168 0.025 0.321 0.032 0.400 0.025 0.232 0.001 0.132 1.121 89.4 Raloxifene, 7 M 8.66 3.60 c 0.168 0.020 0.190 0.012 0.781 0.047 0.296 0.035 0.173 1.089 86.3 Raloxifene, 17 M 17.8 6.96 c 0.092 0.008 0.240 0.024 0.308 0.014 0.338 0.028 0.897 0.979 52.2 Raloxifene, 30 M 41.4 7.28 c 0.165 0.165 0.307 0.016 0.222 0.011 0.392 0.037 3.782 1.085 22.3 Apigenin, 35 M a 169 10 3.42 0.35 4.09 0.18 3.75 0.22 3.21 0.26 20.94 17.61 45.7 Genistein, 35 M a 309 90 0.867 0.014 1.68 0.04 0.314 0.021 0.709 0.017 33.39 3.9 10.5 a Chen et al., 2003. b Under detection limit. c Significant difference with respect to raloxifene concentration according to one-way ANOVA (P 0.05).

380 Jeong et al. Fig. 3. Effect of concentration on the absorptive transport of raloxifene. Raloxifene (1.5 30 M) was added to the apical (AP) side of the Caco-2 cell monolayer, and the amount of raloxifene in the receiver (BL) side was determined at 1, 2, 3, and 4 h after incubation. Rates of transport (B) and permeability (P) across a Caco-2 cellular membrane were obtained according to eq. 1 and eq. 2 as described under Materials and Methods. Each data point represents the mean of three determinations, and the error bars are S.D. of the mean. The line and curve represent linear and polynomial regression of the data points in A (R 2 0.9790) and B (R 2 0.9985), respectively. (Rappa et al., 1999; Wheeler et al., 2000; Hu et al., 2003) or P-glycoprotein inhibitors cyclosporin A or verapamil were added to both sides of Caco-2 cell monolayer to determine whether MRP or P-glycoprotein is involved in the transport of raloxifene and the excretion of its metabolites. Previously, it was shown that leukotriene C 4 was effective in decreasing the efflux of apigenin sulfate and glucuronide (Hu et al., 2003), and MK 571 was effective in decreasing the efflux of both apigenin and chrysin conjugates from the Caco-2 cells (Walle et al., 1999; Hu et al., 2003). MRP inhibitors significantly decreased P BA of raloxifene (7 M) (P 0.05) (Fig. 5A) but had no effect on P AB of raloxifene (Fig. 5A). In contrast, P-glycoprotein inhibitors significantly increased P AB of raloxifene (7 M) (P 0.05) (Fig. 5A) but had no effect on P BA of raloxifene (Fig. 5A). In addition, MRP inhibitors also significantly decreased the clearance of glucuronide from both basolateral and apical sides (P 0.05) (Fig. 5B) and the clearance of sulfate from the apical side (Fig. 5C). However, they did not change the Fig. 4. Effect of raloxifene concentration on the clearance of raloxifene metabolites to the apical (AP) or basolateral (BL) side of the Caco-2 cell monolayer. Raloxifene (1.5 30 M) was added to the apical side of the Caco-2 cell monolayer, and the amount of raloxifene glucuronide and sulfate in the receiver (basolateral) and donor (apical) side was determined at 1, 2, 3, and 4 h after incubation. Metabolic clearance (CL) of raloxifene was calculated according to eq. 3 described under Materials and Methods. Each data point represents the mean of three determinations, and the error bars are S.D. of the mean. Each curve represents power regression of the data points using Microsoft Excel. A, R 2 0.8243 and 0.9563 for glucuronide and sulfate; B, R 2 0.8973 and 0.8915 for glucuronide and sulfate, respectively., statistically significant differences (P 0.05) between glucuronide and sulfate. basolateral clearance of sulfate (Fig. 5C). P-glycoprotein inhibitors had no significant effect on the clearance of glucuronides and sulfates from both sides of Caco-2 cells (Fig. 5, B and C) except for cyclosporin A, which increased the basolateral clearance of sulfate when raloxifene was added to the apical side (Fig. 5C). Effect of Organic Anion Transporter (OAT) Inhibitors on the Transport of Raloxifene and Excretion of Its Metabolites. Estrone sulfate (an organic anionic transporter or OAT substrate and inhibitor) (Sekine et al., 2000) was shown to be effective in decreasing the efflux of apigenin sulfate from the Caco-2 cells (Hu et al., 2003). Therefore, OAT inhibitors estrone glucuronide (50 M) or estrone sulfate (50 M) were used to study the possible involvement of OAT in the transport of raloxifene (7 M) and in the excretion of raloxifene metabolites. Both of them increased the rate of transport for raloxifene (70% and 90% for estrone glucuronide and estrone sulfate, respectively, P 0.05) when

Raloxifene Transport in the Caco-2 Model 381 Fig. 5. Effect of MRP and P-glycoprotein inhibitors on the permeability of raloxifene and clearance of its metabolites in Caco-2 cells. Raloxifene (7 M) was added to the apical (AP) or basolateral (BL) side of the Caco-2 cell monolayer. Fifty micromolar MK-571 plus 0.1 M leukotriene C 4,20 M cyclosporine A, or 20 M verapamil was added to both sides of the Caco-2 cell monolayer. Metabolic clearance (CL) of raloxifene was calculated using the amounts of metabolites at 1, 2, 3, and 4 h after incubation according to eq. 3 described under Materials and Methods. Each column represents the mean of three determinations, and the error bars are S.D. of the mean. Blank, solid, dashed, and dotted columns represent the control, MK-571 plus leukotriene C 4, and cyclosporine A- or verapamiltreated group, respectively., statistically significant difference (P 0.05) compared with the control (i.e., raloxifene only). they were added to both the apical and the basolateral media. There was no significant difference between the effects of these two OAT inhibitors (Fig. 6A). For raloxifene glucuronide, estrone sulfate significantly decreased (40%) the basolateral clearance without affecting its apical clearance, whereas estrone glucuronide significantly increased (75%) the apical clearance without affecting its basolateral clearance (Fig. 6B). For raloxifene sulfate, these two inhibitors significantly decreased its clearance from both sides of the Caco-2 cell monolayer (25% 50%) (Fig. 6C). Effect of Estrone Sulfate on the Transport and Metabolism of Raloxifene as a Function of Concentration. Increasing concentration of estrone sulfate (1 50 M) significantly increased (135% 190%) the rate of transport of raloxifene (7 M) through Caco-2 cells (P 0.05) (Fig. 7A) with an apparent EC 50 value of 16.4 M. On the other hand, increasing concentration of estrone sulfate significantly decreased the basolateral clearance of raloxifene glucuronide Fig. 6. Effect of OAT inhibitors on the transport of raloxifene and clearance of its metabolites in Caco-2 cells. Raloxifene (7 M) was added to the apical side of the Caco-2 cell monolayer. Fifty micromolar estrone glucuronide (EG) or estrone sulfate (ES) was added to both sides of the Caco-2 cell monolayer. Rate of transport (B t ) and metabolic clearance (CL) of raloxifene were calculated using the amounts of raloxifene and metabolites at 1, 2, 3, and 4 h after incubation according to eq. 1 and eq. 3 described under Materials and Methods. Each column represents the mean of three determinations, and the error bars are S.D. of the mean. Blank, solid, and dashed columns represent the control, EG-treated, and ES-treated groups, respectively. The mean control values ( S.D.) of B t and CLs were 0.059 0.013 nmol/h (A), 3.10 0.10 10 6 ml/s, 11.16 0.36 l/h [glucuronide, basolateral (BL) side, B], 10.04 1.37 l/h [glucuronide, apical side (AP), B], 11.23 0.47 l/h (sulfate, basolateral side, C), and 26.57 1.12 l/h (sulfate, apical side, C), respectively., statistically significant difference (P 0.05) compared with the control. with a maximum decrease of 40% but did not significantly affect the apical clearance of glucuronide (Fig. 7B). Estrone sulfate inhibited both apical and basolateral clearance of raloxifene sulfate with a maximum decrease of 50% (Fig. 7C). Effect of Estrone Sulfate on the Metabolism of Raloxifene by Caco-2 Cell Lysate. Twenty micromolar estrone sulfate decreased in vitro formation of raloxifene glucuronide and sulfate (using Caco-2 cell lysate) by about 30% (P 0.05)

382 Jeong et al. Fig. 7. Effect of estrone sulfate concentration on the rate of raloxifene transport and clearance of raloxifene metabolites across the Caco-2 cell monolayer. Raloxifene (7 M) was added to the apical (AP) side of the Caco-2 cell monolayer. Estrone sulfate (1 50 M) was added to both sides of the Caco-2 cell monolayer. Rate of transport (B t ) and metabolic clearance (CL) of raloxifene were calculated using the amounts of raloxifene and metabolites at 1, 2, 3, and 4 h after incubation according to eq. 1 and eq. 3 described under Materials and Methods. Each data point represents the mean of three determinations, and the error bars are S.D. of the mean. The mean control values ( S.D.) of B t and CLs were 0.059 0.013 nmol/h (A), 3.10 0.10 10 6 ml/s, 11.16 0.36 l/h (glucuronide, basolateral side, B), 10.04 1.37 l/h (glucuronide, apical side, B), 11.23 0.47 l/h (sulfate, basolateral side, C), and 26.57 0.1.12 l/h (sulfate, apical side, C), respectively. Each curve represents rectangular regression of the data points using SigmaPlot. A, R 2 0.9680 and EC 50 16.4 M; B, R 2 0.9639 for basolateral side and maximum decrease of 40%; C, R 2 0.8435 and 0.8168 for the basolateral and apical sides and maximum decrease of 50% for both sides, respectively. The solid and open symbols indicate the basolateral and apical sides, respectively., statistically significant difference (P 0.05) compared with the control. (Fig. 8). Excretion rates (B ex ) of both metabolites from the Caco-2 cells monolayer were higher (3.4 and 1.3 times for glucuronide and sulfate, respectively) than the rates of metabolism in the cell lysate (B met ) (Fig. 8). Estrone sulfate Fig. 8. Effect of estrone sulfate (ES) on the in vitro metabolism of raloxifene by Caco-2 cell lysate or excretion of metabolites from Caco-2 cell. In metabolic experiments, raloxifene (7 M) and ES (20 M) were incubated for 4 h with Caco-2 cell lysate (microsome-rich fraction) or supernatant of the cell lysate (cytosol-rich fraction) ( 1 mg/ml of protein), and the rate of metabolism (B met ) was obtained as described under Materials and Methods. Equivalent of amounts of organic solvent (0.5% in this case) were added instead of ES in the control experiment. In transport experiment, raloxifene (7 M) was added to the apical (AP) side, and estrone sulfate (20 M) was added to both sides of the Caco-2 cell monolayer. The sum of donor (AP) side and receiver (basolateral, BL) side excretion rate (B ex ) of each metabolite was calculated using the amounts of metabolites at 1, 2, 3, and 4 h after incubation as described under Materials and Methods. Each column represents the mean of three determinations, and the error bars are S.D. of the mean. Blank and solid columns represent rate of metabolism and excretion, respectively. A, B ex /B met 3.4 and 3.7 for the control and ES-treated group; B, B ex /B met 1.3 and 0.8 for the control and ES-treated group, respectively. and, statistically significant differences (P 0.05) between the control and ES-treated group and between metabolism and excretion, respectively. decreased the ratio of excretion rate over metabolism rate (B ex /B met ) for raloxifene sulfate by 40% (P 0.05) (Fig. 8B) but did not affect this ratio for raloxifene glucuronide (Fig. 8A). Cellular Accumulation of Raloxifene and Its Metabolites in Caco-2 Cells. Raloxifene was extensively bound to Caco-2 cells (up to 73% of dose after 24 h of incubation with 6 M raloxifene). Among the conjugated metabolites, only sulfate was detected in the cell, whereas glucuronide was not detected. The amount of raloxifene taken up by or bound to cells was significantly decreased after 24 h of incubation compared with those after 4 h of incubation, whereas the amount of sulfate found inside the cells was significantly increased after 24 h of incubation (data not shown). The amount of raloxifene taken up by the Caco-2 cell increased

significantly as concentrations increased (up to 7 M) but did not increase further after that concentration (Fig. 9A). The uptake trend of raloxifene sulfate was similar to raloxifene, but the extent of uptake was 3.5 times smaller (Fig. 9B). Discussion Raloxifene has the potential to become the drug for breast cancer prevention. However, raloxifene has poor bioavailability (Lindstrom et al., 1984). We hypothesized that extensive intestinal metabolism and enteric and enterohepatic recycling of raloxifene conjugates could explain why this drug has poor oral bioavailability but a long half-life (Fig. 10). We further hypothesized that bioavailability of raloxifene could be enhanced via inhibition of phase II metabolism and/or inhibition of efflux transporters responsible for pumping out Fig. 9. Effect of raloxifene concentration on the accumulation of raloxifene and its sulfate by the Caco-2 cells. Raloxifene (1.5 18 M) was added to the apical side of the Caco-2 cell monolayer, and the amounts of raloxifene and its metabolites inside the cell were determined at 24 h after incubation as described under Materials and Methods. Each data point represents the mean of three determinations, and the error bars are S.D. of the mean. Each curve represents rectangular regression of the data points using SigmaPlot. A, R 2 0.9681; B, R 2 0.9720., statistically significant difference (P 0.05) compared with the lower concentration. Raloxifene Transport in the Caco-2 Model 383 the intracellular phase II metabolites. In this study, we investigated the mechanisms responsible for the absorption and excretion of raloxifene and its metabolites in the Caco-2 cell culture model. Our results suggest that raloxifene is subjected to efflux because: 1) as concentration increased, the permeability of raloxifene across the Caco-2 cell monolayer increased and the rate of transport increased parabolically (Fig. 3), consistent with saturation of efflux transporters; 2) secretory transport of raloxifene was faster than absorptive transport (Table 1); and 3) the ratio of secretory to absorptive transport rate (B BA /B AB ) was decreased as the concentration of raloxifene increased (Table 1). The efflux appeared to be mediated by MRP and P-glycoprotein since secretory transport was diminished when MRP inhibitors were added to the media and absorptive transport was augmented when P-glycoprotein inhibitors were added to the media (Fig. 5). The decrease by MRP inhibitors was achieved even though blockage of conjugate efflux has the tendency to increase the amounts of intact compound transported. Therefore, our data showed for the first time that one of the reasons why intestinal absorption of raloxifene is incomplete was due to its efflux by MRP and P-glycoprotein. This represents an important discovery about the transport of raloxifene in the enterocytes. Previously, absorption mechanisms of raloxifene were unknown. Usually, the passive diffusion process was implied as the mechanism of absorption since there is no demonstrated nonlinear relationship between dose and absorption. Our results showed that raloxifene undergoes extensive intestinal metabolism during transport across the Caco-2 cells (Fig. 1; Table 2). At low concentration of 7 M raloxifene, the total clearance value for both conjugates added together was about 59 l/h, which is enough to clear the cellular metabolite volume 16 times/h. The extensive metabolism is consistent with reported extensive first pass metabolism of raloxifene following oral administration (Hochner- Celnikier, 1999). The major metabolites in the Caco-2 cells are mainly sulfates followed by glucuronides. In addition, sulfates are rapidly and preferentially excreted into the apical side, which diminishes their transport into the systemic circulation. Furthermore, percentages of glucuronides increased as the concentration of raloxifene increased. Therefore, our results are consistent with the earlier observation that the main metabolite in human plasma is the glucuronides including the primary metabolite raloxifene-4 -glucuronide (Hochner-Celnikier, 1999). They are also consistent with a recent discovery that intestinal glucuronidation is the most important contributor to the presystemic clearance of raloxifene (Kemp et al., 2002). We then proceeded to determine the mechanisms responsible for the efflux of raloxifene conjugates since metabolic clearance decreased as concentration increased (Fig. 4), suggesting the involvement of a saturable process. The results indicated that excretion of raloxifene glucuronide and sulfate was mediated by efflux transporters such as MRP and/or OAT because: 1) clearance of metabolites was polarized and saturable (Fig. 4), and 2) clearances into apical and basolateral media were changed in the presence of MK-571 and leukotriene C 4, estrone glucuronide, or estrone sulfate (Figs. 5 and 6). Taken together, these results have elucidated for the first time the mechanisms responsible for efflux of a

384 Jeong et al. Fig. 10. Schematic representation of intestinal disposition of raloxifene. Absorbed raloxifene, in free or conjugated form, will be effluxed to the lumen (AP) or transported into the liver via portal vein. After intact compounds are further metabolized in the liver, conjugates either enter into the systemic circulation or are secreted to the intestinal lumen via bile. The excretion of the phase II conjugates by the enterocytes and hepatocytes render enteric and enterohepatic recycling of raloxifene, which in turn increases its apparent half-life. The present study focuses on mechanisms (e.g., MDR-, MRP-, or OAT-mediated efflux) responsible for intestinal disposition of raloxifene and/or its conjugates. glucuronide and a sulfate metabolite of a drug from the intestinal cells. Previously, we have reported the efflux mechanism responsible for the efflux of apigenin sulfate and apigenin glucuronide (Hu et al., 2003), whereas Walle and his coworker reported efflux of chrysin conjugates were inhibited by MK-571, an inhibitor of both glucuronidation and MRP (Walle et al., 1999; Hu et al., 2003). We further analyzed the results of various chemical inhibitors on the excretion to determine which transporter(s) may be mainly responsible for the excretion of raloxifene sulfate and glucuronide. The main pathway for glucuronide (Fig. 10) appears to be MRP since use of two MRP inhibitors inhibited the excretion by more than 80% (Fig. 5), with minor contribution from basolateral OAT (Fig. 6). The main pathway for sulfate is less well defined since its excretion was never inhibited by more than 55%. Contributions from both apical MRP and OAT and basolateral OAT are likely, and additional contributions from other transporter(s) are possible (Fig. 10). In comparison with the efflux of apigenin conjugates (Hu et al., 2003), the efflux of raloxifene glucuronide is much better defined than apigenin conjugate (about 60% attributable to MRP and OAT), but the efflux of sulfate is less well defined than apigenin sulfate ( 80% attributable to MRP and OAT). Further analysis of the inhibitor results suggests that MDR1 or P-glycoprotein is not involved in limiting the efflux of conjugates since neither cyclosporine A nor verapamil decreased the efflux of any conjugates. However, it is not presently clear why cyclosporine A increased the basolateral efflux of sulfate. To probe the possible mechanisms, we determined if cyclosporine A would increase sulfate formation in Caco-2 cell lysate. The result indicated a slight decrease in the formation of raloxifene sulfate in the presence of 20 M cyclosporine A using Caco-2 cell lysate (27% decrease), which suggests that cyclosporine A did not enhance the metabolite formation. The actual mechanism for sulfate efflux enhancement remains to be determined. Our discovery that raloxifene and its metabolites are excreted by the enterocyte efflux transporters is novel. The excretion of raloxifene metabolites is the result of direct coupling of efflux transporter and phase II metabolic enzymes, which has not been demonstrated for the disposition of a drug in the intestine previously. The direct coupling is demonstrated by the fact that inhibition of conjugate efflux could result in increase in raloxifene transport (Figs. 6 and 7). The direct coupling is clearly the enabling process for enteric recycling and may also be used to explain why raloxifene has a low oral bioavailability but a long half-life. The coupling of conjugating enzymes that produce hydrophilic metabolites and efflux transporters is more complex than the coupling of CYP3A4 and P-glycoprotein (Cummins et al., 2002) because these conjugates cannot exit cells via passive diffusion. Hence, the action of the efflux transporters can change cellular equilibrium between parent compound and its conjugates. This change in equilibrium has at least two possible consequences, both of which could lead to a higher than expected excretion of raloxifene conjugates (Fig. 8). The first consequence is less accumulation of conjugates in the cells because of rapid excretion of conjugates that can inhibit phase II conjugation via product inhibition. In other words, when the efflux dominates the cellular excretion, very little conjugate is accumulated by the cells. In the present study, the excretion rate of raloxifene glucuronide was much faster (3.4 times) than its formation rate, resulting in no measurable cellular accumulation of this metabolite even after 24 h. On the other hand, sulfate excretion rate was comparable with sulfate formation rates, resulting in significant accumulation of sulfate in cells after 24 h (Fig. 9). The second consequence is less hydrolysis of the conjugates to parent drug via the action of glucuronidases and sulfatases as proposed (Hochner-Celnikier, 1999), resulting in higher than expected excretion of conjugates (Fig. 8). Taken together, these two consequences will result in higher apparent excretion rate than the formation rate when efflux inhibitor was absent (Fig. 8). When the efflux transporter function was inhibited by estrone sulfate, the excretion of sulfates became slower than metabolism of sulfate as expected. Taken together, these results support the coupling hypothesis that predicts the modulation of cellular excretion of hydrophilic conjugates by the efflux transporters.

The coupling of efflux transporters and conjugating enzymes also support our hypothesis that enteric recycling (Liu and Hu, 2002) is important for the in vivo metabolism of raloxifene. This coupling can explain why substantial amounts of raloxifene conjugates are produced and excreted by the enterocytes. Because this coupling appears to function better at the apical side, at least in the case of raloxifene, it increases the importance of enteric recycling in the disposition of raloxifene. Because the apical side was the preferred side of excretion for the raloxifene sulfate, it may be used to explain why smaller amounts of sulfated metabolites are recovered in plasma since excreted metabolites are expected to be eliminated in feces or hydrolyzed by the microflora. Lastly, we also hypothesized that we could increase the bioavailability of raloxifene by inhibiting the efflux transporters responsible for the excretion of raloxifene conjugates. OAT inhibitors we used are not known to interact with MRP, even though they could slightly decrease the formation rates of conjugates. Therefore, increase in the intestinal transport of intact raloxifene (Fig. 6) can be viewed mainly as the result of inhibiting the efflux of conjugated metabolites, which in turn decreased the amounts of metabolites formed and allowed more intact raloxifene to permeate the Caco-2 cell monolayers intact. This is an important discovery since we have shown that inhibition of an efflux transporter(s) that are tightly coupled to conjugating enzymes can decrease the extent of metabolism and improve the transport of the intact compound. If such an effect can be shown in vivo, it would provide a new mechanism to improve the oral bioavailability of drugs and to explain drug interactions in the clinic. In conclusion, raloxifene and its phase II conjugates are excreted by the intestinal cells via the actions of P-glycoprotein, MRP, and/or OAT. Coupling of efflux transporters and conjugating enzymes enhances intestinal excretion and renders the enteric recycling. On the other hand, the inhibition of OAT responsible for conjugate efflux can increase transport of intact raloxifene and has the potential to increase its bioavailability pending the actual demonstration in vivo. Taken together with enterohepatic recycling, intestinal disposition may be used to explain why this drug has low bioavailability but a long half-life. References Bohets H, Annaert P, Mannens G, Van Beijsterveldt L, Anciaux K, Verboven P, Meuldermans W, and Lavrijsen K (2001) Strategies for absorption screening in drug discovery and development. Curr Top Med Chem 1:367 383. Caro I, Boulenc X, Rousset M, Meunier V, Bourrie M, Julian B, Joyeux H, Roques C, Berger Y, Zwebaum A, et al. (1995) Characterization of a newly isolated Caco-2 clone (TC-7), as a model of transport processes and biotransformation of drugs. Int J Pharm 116:147 157. Chen J, Lin H, and Hu M (2003) Metabolism of flavonoids via enteric recycling: role of intestinal disposition. J Pharmacol Exp Ther 304:1228 1235. Colacurci N, Manzella D, Fornaro F, Carbonella M, and Paolisso G (2003) Endothelial function and menopause: effects of raloxifene administration. J Clin Endocrinol Metab 88:2135 2140. Cummins CL, Jacobsen W, and Benet LZ (2002) Unmasking the dynamic interplay Raloxifene Transport in the Caco-2 Model 385 between intestinal P-glycoprotein and CYP3A4. J Pharmacol Exp Ther 300:1036 1045. Delmas PD, Bjarnason NH, Mitlak BH, Ravoux AC, Shah AS, Huster WJ, Draper M, and Christiansen C (1997) Effect of raloxifene on bone mineral density, serum cholesterol concentrations and uterine endometrium in postmenopausal women. New Engl J Med 337:1641 1647. Duschek EJ, Stehouwer CD, de Valk-de Roo GW, Schalkwijk CG, Lambert J, and Netelenbos C (2003) Raloxifene, conjugated oestrogen and endothelial function in postmenopausal women. J Intern Med 254:85 94. Eli Lilly (1998) Raloxifene package insert. Gres MC, Julian B, Bourrie M, Meunier V, Roques C, Berger M, Boulenc X, Berger Y, and Fabre G (1998) Correlation between oral drug absorption in humans and apparent drug permeability in TC-7 cells, a human epithelial intestinal cell line: comparison with the parental Caco-2 cell line. Pharm Res 15:726 733. Hochner-Celnikier D (1999) Pharmacokinetics of raloxifene and its clinical application. Eur J Obstet Gynecol Reprod Biol 85:23 29. Hu M, Chen J, and Lin H (2003) Disposition of flavonoids via recycling: mechanistic studies of disposition of apigenin in the Caco-2 cell culture model. J Pharmacol Exp Ther 307:314 321. Hu M, Chen J, Tran D, Zhu Y, and Leonardo G (1994a) The Caco-2 cell monolayers as an intestinal metabolism model: metabolism of dipeptide Phe-Pro. J Drug Target 2:79 89. Hu M, Chen J, Zheng L, Dantzig AH, and Stratford RE Jr (2000) Uptake characteristics of loracarbef and cephalexin in the Caco-2 cell culture model: effects of the proton gradient and possible presence of a distinctive second component. J Pharm Sci 85:767 772. Hu M, Chen J, Zhu Y, Dantzig AH, Stratford RE, and Kuhfeld MT Jr (1994b) Mechanism and kinetics of transcellular transport of a new -lactam antibiotic loracarbef across a human intestinal epithelial model system (Caco-2). Pharm Res 11:1405 1413. Hu M, Li Y, Davitt CM, Huang SM, Thummel K, Penman BW, and Crespi CL (1999) Transport and metabolic characterization of Caco-2 cells expressing CYP3A4 and CYP3A4 plus oxidoreductase. Pharm Res 16:1352 1359. Jordan VC and Morrow M (1999) Tamoxifen, raloxifene and the prevention of breast cancer. Endocr Rev 20:253 278. Kemp DC, Fan PW, and Stevens JC (2002) Characterization of raloxifene glucuronidation in vitro: contribution of intestinal metabolism to presystemic clearance. Drug Metab Dispos 30:694 700. Lindstrom TD, Whitaker NG, and Whitaker GW (1984) Disposition and metabolism of a new benzothiophene antiestrogen in rats, dogs and monkeys. Xenobiotica 14:841 847. Liu Y and Hu M (2002) Absorption and metabolism of flavonoids in the caco-2 cell culture model and a perused rat intestinal model. Drug Metab Dispos 30:370 377. MacGregor JI and Jordan C (1998) Basic guide to the mechanisms of antiestrogen action. Pharmacol Rev 50:151 196. Pfrunder A, Gutmann H, Beglinger C, and Drewe J (2003) Gene expression of CYP3A4, ABC-transporters (MDR1 and MRP1-MRP5) and hpxr in three different human colon carcinoma cell lines. J Pharm Pharmacol 55:59 66. Pontier C, Pachot J, Botham R, Lenfant B, and Arnaud P (2001) HT29-MTX and Caco-2/TC7 monolayers as predictive models for human intestinal absorption: role of the mucus layer. J Pharm Sci 90:1608 1619. Ranaldi G, Consalvo R, Sambuy Y, and Scarino ML (2003) Permeability characteristics of parental and clonal human intestinal Caco-2 cell lines differentiated in serum-supplemented and serum-free media. Toxicol In Vitro 17:761 767. Rappa G, Finch RA, Sartorelli AC, and Lorico A (1999) New insights into the biology and pharmacology of the multidrug resistance protein (MRP) from gene knockout models. Biochem Pharmacol 58:557 562. Scott JA, Da Camara CC, and Early JE (1999) Raloxifene: a selective estrogen receptor modulator. Am Fam Physician 60:1131 1138. Sekine Y, Cha SH, and Endou H (2000) The multispecific organic anion transporter (OAT) family. Pflugers Arch 440:337 350. Sabolovic N, Magdalou J, Netter P, and Abid A (2000) Nonsteroidal antiinflammatory drugs and phenols glucuronidation in Caco-2 cells: identification of the UDP-glucuronosyltransferases UGT1A6, 1A3 and 2B7. Life Sci 67:185 196. Walle UK, Galijatovic A, and Walle T (1999) Transport of the flavonoid chrysin and its conjugated metabolites by the human intestinal cell line Caco-2. Biochem Pharmacol 58:431 438. Wheeler R, Neo SY, Chew J, Hladky SB, and Barrand MA (2000) Use of membrane vesicles to investigate drug interactions with transporter proteins, P-glycoprotein and multidrug resistance-associated protein. Int J Clin Pharmacol Ther 38:122 129. Address correspondence to: Dr. Ming Hu, Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Pullman, WA 99164-6510. E-mail: minghu@wsu.edu