JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 JPET FastThis article Forward. has not beenPublished copyedited on and Marchformatted. 12, The 2004 final version as DOI:10.1124/jpet.103.063925 may differ from this version.

JPET #63925

Disposition Mechanisms of in the Human Intestinal Caco-2 Model

Eun Ju Jeong1, Huimin Lin1 and Ming Hu1, 2

Downloaded from

jpet.aspetjournals.org

at ASPET Journals on September 29, 2021

1

Copyright 2004 by the American Society for Pharmacology and Experimental Therapeutics. JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

1Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University,

Pullman, Washington

2Address correspondence to:

Ming Hu, Ph.D. Downloaded from

Department of Pharmaceutical Sciences

College of Pharmacy jpet.aspetjournals.org Washington State University

Pullman, WA 99164-6510

Tel: (509)-335-4184 at ASPET Journals on September 29, 2021

Fax :( 509)-335-5092

Email: [email protected]

ABBREVIATIONS: HBSS, Hank’s balanced salt solution; MRP, multidrug resistance related protein; OAT, organic anionic transporter; PAPS, 3’-phosphoadenosine 5’-phosphosulfate;

SERM, selective ; UGT, UDP-glucuronosyltransferases; PBA, secretory permeability; PAB, absorptive permeability

2 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

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 (PAB) of raloxifene was lower than its secretory permeability (PBA). As the concentration increased, the

efflux ratio (PBA/PAB) decreased, but PAB increased. PAB was also increased in the presence of Downloaded from verapamil and cyclosporine A, two p-glycoprotein inhbitors. Raloxifene was extensively metabolized into sulfated and glucuronidated conjugates. The extent of or jpet.aspetjournals.org was decreased as the concentration increased from 3.4 µM (96%) to 30 µM (22%). MRP inhibitors MK-571 and leukotriene C4 significantly decreased (maximal 80%) apical efflux of

both conjugates. They also significantly decreased (maximal 85%) basolateral efflux of at ASPET Journals on September 29, 2021 glucuronides but not sulfates. On the other hand, organic anion inhibitor (OAT) 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 OAT inhibitor 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 can result in increase in the transport of the intact compound. It also provides additional explanation why raloxifene has low but a long half-life.

3 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

Raloxifene, a selective modulator (SERM) (Duschek et al., 2003;

Colacurci et al., 2003), is used for the treatment of osteoporosis. Raloxifene blocks the adverse effects of estrogen in tissues and uterine 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 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 Downloaded from agent for (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 jpet.aspetjournals.org (Scott et al, 1999; Jordan and Morrow, 1999).

Raloxifene is reported by its manufacturer to be rapidly absorbed after , at ASPET Journals on September 29, 2021 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 with high within-subject variability (approximately 30% CV)

(Hochner-Celnikier, 1999). It undergoes extensive phase II biotransformation, primarily , but was not metabolized by the cytochrome P450s (Eli Lilly, 1998). Raloxifene has a plasma elimination half-life of approximately 27 hours, 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).

4 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

Although a variety of pharmacokinetic information has been provided by its manufacturer (as summarized previously), mechanisms responsible for absorption of the raloxifene, and of raloxifene conjugates have not been reported. Studies have not established the dynamic relationship between absorption, metabolism and excretion of raloxifene 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 Downloaded from human intestinal Caco-2 cell model using the cloned TC7 variant.

jpet.aspetjournals.org We chose Caco-2 TC7 cells, one of the cloned Caco-2 variants, for increased homogeneity and stability of cell population (Ranaldi et al., 2003, Caro et al., 1995). TC variant exhibits morphological characteristics similar to those of the parental Caco-2 cells (Gres et al., at ASPET Journals on September 29, 2021

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 if 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 (Liu and Hu, 2002), is analogous to enterohepatic recycling except the phase II conjugates are produced and excreted by the enterocytes instead of hepatocytes.

5 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

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 (Eli Lilly and Company, Indianapolis, IN) using 100% ethanol and concentration was then verified by using raloxifene hydrochloride purchased from National Cancer Institute Chemical Downloaded from

Standard Repository managed by Midwest Research Institute (Kansas City, MO). β-

Glucuronidase, sulfatase, Hank’s balanced salt solution (HBSS; powder form), cyclosporin A, jpet.aspetjournals.org verapamil, estrone glucuronide and estrone sulfate were purchased from Sigma (St. Louis, MO).

Leukotriene C4 and MK-571 were purchased from Biomol (Plymouth Meeting, PA). All other

materials were analytical grade or better and used as received. at ASPET Journals on September 29, 2021

Cell culture

Cell culture conditions for growing Caco-2 cells have been described previously (Liu and

Hu, 2002; Hu et al., 1994a,b; Chen et al., 2003). The seeding density was 100,000 cells/cm2 (4.2 cm2 per 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,b). Cell monolayers from 19 to 22 days past seeding were used for the experiments.

Transport Experiments in the Caco-2 Cell Model

6 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

The protocols for performing transport experiments were similar to those described previously (Liu and Hu, 2002). Briefly, the cell monolayers were washed three times with 37ºC

HBSS (pH 7.4). The transepithelial electrical resistance values were measured and those with the values less than 420 ohms x cm2 were discarded. Various concentrations (ranging from 1.5 to 30

µM) 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 Downloaded from samples (650 µl 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 is 2.5 ml) and same volume of jpet.aspetjournals.org donor solution (containing raloxifene) or transport medium was replaced after each sampling.

Forty-five µl of internal standard solution (100 µM in methanol) was immediately

µ added to 200 l of samples to stabilize them until analysis. at ASPET Journals on September 29, 2021

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 cm2, Nalge Nunc International, Rochester, NY). One ml of HBSS (pH 7.4) was added to each membrane and the solution was frozen (in liquid nitrogen) and thawed (in 37ºC water bath) for three times to disrupt cell membranes. After centrifugation at 13,000 rpm for 8 min, 0.5 ml of supernatant was collected. Half ml of methanol was added to the remaining and the tubes were centrifuged again to extract the remaining raloxifene.

7 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

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 37ºC 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 (4ºC) for 30 min as described previously (Hu et al., 2003). Afterwards, 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- Downloaded from

Rad, Hercules, CA).

jpet.aspetjournals.org 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 at ASPET Journals on September 29, 2021 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 µl) and the mixture was incubated in a 37ºC shaking (200 rpm) water bath for 4 h. The reaction was stopped by the addition of 50 µl solution of 94% acetonitril/6% glacial acetic acid containing 100 µM of 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 (PAPS) (0.1 mM) was added last to the reaction mixture

8 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

(total volume 200 µl) and the mixture was incubated in a 37ºC shaking (200 rpm) water bath for

4 h. The reaction was stopped by the addition of 50 µl solution of 94% acetonitril/6% glacial acetic acid containing 100 µM of testosterone as the internal standard.

Hydrolysis of Raloxifene Metabolites by Hydrolases.

A portion of 24 h samples of raloxifene (7 µM) transport and metabolism experiments was extracted with methylene chloride to remove intact raloxifene. The remaining aqueous phase Downloaded from was incubated with glucuronidase (20 units per reaction) or sulfatase (0.5 unit per reaction) at

37ºC for 4 h to reconvert conjugated raloxifene to raloxifene, which is used to identify the jpet.aspetjournals.org raloxifene peaks and amounts of metabolites (via reconversion) in HPLC chromatograms (Fig.1).

at ASPET Journals on September 29, 2021

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 µm, 150 X 4.60 mm (Phenomenex, Gilroy, 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% acetonitril; 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; injection volume, 200 µl. The retention times for raloxifene, glucuronide and sulfate and internal standard were 14.4, 9.7, 12.1 and 21.5 min, respectively (Fig.

1).

9 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

Data Analysis

Rates of transport (Bt) 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)

(equation [1]). Permeability (P) across a cellular membrane was calculated using the rate of transport divided by the surface area (A) of the monolayer and the initial concentration of raloxifene at the loading side (Ci) (equation [2]).

dC Downloaded from Bt = ×V (1) dt

B P = t (2) AC i jpet.aspetjournals.org

Apparent metabolic clearance (CL) of raloxifene was calculated using rate of excretion (Bex) divided by the initial concentration of raloxifene at the loading side (Ci) (equation [3]).

at ASPET Journals on September 29, 2021

B CL = ex (3) Ci

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.

10 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

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.2A, Fig. 2C). The amounts of metabolites in the donor side also increased linearly with Downloaded from time (Fig. 2B, Fig. 2D), but the amount of raloxifene in the donor side did not change significantly (Fig. 2B, Fig. 2D), since we replenished it after each sampling. jpet.aspetjournals.org

The rates of raloxifene transport were determined with respect to the direction of transport (Table 1). Secretory (basolateral to apical) rates of transport (BBA) were always higher at ASPET Journals on September 29, 2021 than absorptive (apical to basolateral) rates of transport (BAB) (P < 0.05) (Table 1). As concentration of raloxifene increased, the ratio of BBA to BAB decreased from 3.5 to 1.5. Pre- treatment of Caco-2 cells with 17 µM 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

-6 -6 Raloxifene has low absorptive permeability (PAB=0.39x10 ~ 4.1x10 , 1.5~30µM) when

-6 -6 compared with (PAB=31x10 , 35 µM) and (PAB=17x10 , 35 µM) (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).

11 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

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 Downloaded from high clearance value. The clearance values for apical efflux of glucuronide and sulfate (at a raloxifene concentration of 7 µM) were 10.04±1.37 µl/hr, and 26.57±1.12 µl/hr, respectively. jpet.aspetjournals.org For basolateral efflux, they were roughly equal, or about 11 µl/hr. Taken together, the total clearance values were 37.57 µl/hr for sulfates and 21 µl/hr for glucuronides, respectively. With a

cellular water volume of about 3.66 µl/mg protein, the total monolayer cellular water volume is at ASPET Journals on September 29, 2021 about 3.66 µl (Hu et al, 1994a). Therefore, the whole “cellular sulfate volume” is replaced about

10 times per hour or once every 6 min, whereas the whole “cellular glucuronide volume” is replaced about 5 times per hour 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. 4A, Fig.4B).

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

12 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925 basolateral clearance also decreased significantly (p < 0.05 according to one-way ANOVA) with an increase in raloxifene concentration (Fig. 4A, Fig.4B).

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 µM (p < 0.05) (Fig. 4A). However,

this difference in clearance decreased as raloxifene concentration increased, and eventually Downloaded from became minimal at 30 µM. On the other hand, basolateral clearance of the two metabolites was similar at all the concentrations measured (Fig. 4B). jpet.aspetjournals.org

Effect of Multidrug Resistance Related Protein (MRP) and p-Glycoprotein Inhibitors on the Transport of Raloxifene and Excretion of its Metabolites at ASPET Journals on September 29, 2021

Multidrug resistance related protein (MRP) inhibitor MK-571 (50 µM) and leukotriene

C4 (leukotriene C4, 0.1 µM) (Hu et al., 2003; Rappa et al., 1999; Wheeler et al., 2000) or p- glycoprotein inhibitors, cyclosporin A or verapamil, were added to both sides of Caco-2 cell monolayer to determine if MRP or P-glycoprotein is involved in the transport of raloxifene and the excretion of its metabolites. Previously, it was shown that leukotriene C4 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 PBA of raloxifene (7

µM) (p < 0.05) (Fig.5A), but had no effect on PAB of raloxifene (Fig. 5A). In contrast, P- glycoprotein inhibitors significantly increased PAB of raloxifene (7 µM) (p < 0.05) (Fig.5A), but had no effect on PBA of raloxifene (Fig. 5A).

13 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

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 apical side (Fig. 5C). However, they did not change the 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. 5B and 5C) except for cyclosporin A, which increased the basolateral clearance of sulfate when raloxifene was added to apical side (Fig 5C).

Downloaded from

Effect of Organic Anion Transporter (OAT) Inhibitors on the Transport of Raloxifene and

Excretion of its Metabolites jpet.aspetjournals.org Estrone sulfate (an organic anionic transporter or OAT substrate and inhibitor) 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 µM) or estrone sulfate (50 µM) was at ASPET Journals on September 29, 2021 used to study the possible involvement of OAT in the transport of raloxifene (7 µM) 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 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).

14 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

Effect of Estrone Sulfate on the Transport and Metabolism of Raloxifene as a Function of

Concentration

Increasing concentration of estrone sulfate (1~50 µM) significantly increased

(135~190%) the rate of transport of raloxifene (7 µM) through Caco-2 cells (p <0.05) (Fig. 7A) with an apparent EC50 value of 16.4 µM. On the other hand, increasing concentration of estrone sulfate significantly decreased the basolateral clearance of raloxifene glucuronide with a

maximum decrease of ≈40% but did not significantly affect the apical clearance of glucuronide Downloaded from

(Fig. 7B). Estrone sulfate inhibited both apical and basolateral clearance of raloxifene sulfate with a maximum decrease of ≈50% (Fig. 7C). jpet.aspetjournals.org

Effect of Estrone Sulfate on the Metabolism of Raloxifene by Caco-2 Cell Lysate

Twenty µM of estrone sulfate decreased formation of raloxifene glucuronide and at ASPET Journals on September 29, 2021 sulfate (using Caco-2 cell lysate) by about 30 % (p < 0.05) (Fig. 8). Excretion rates (Bex) 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 (Bmet) (Fig. 8). Estrone sulfate decreased the ratio of excretion rate over metabolism rate (Bex/ Bmet) 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 incubation with 6 µM 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 incubation compared to those after 4 h incubation,

15 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925 while the amount of sulfate found inside the cells was significantly increased after 24 h incubation (data not shown). The amount of raloxifene taken up by the Caco-2 cell increased significantly as concentrations increased (up to 7 µM), 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).

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 29, 2021

16 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

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, 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 Downloaded from metabolism and/or inhibition of efflux transporters responsible for pumping out the intracellular phase II metabolites. In this study, we investigated the mechanisms responsible for the jpet.aspetjournals.org 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 at ASPET Journals on September 29, 2021 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 (BBA/BAB) 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

17 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925 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 µM raloxifene, the total clearance value for both conjugates added together was about 59 µl/hr, which is enough to Downloaded from clear the cellular metabolite volume 16 times per hour. The extensive metabolism is consistent with reported extensive first-pass metabolism of raloxifene following oral administration jpet.aspetjournals.org

(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, at ASPET Journals on September 29, 2021 percentages of glucuronides increased as the concentration of raloxifene increased. Therefore, our results are consistent with 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), 2) clearances into apical and

18 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

basolateral media were changed in the presence of MK-571 and leukotriene C4, estrone glucuronide, or estrone sulfate (Fig.5 and Fig.6). Taken together, these results have elucidated for the first time the mechanisms responsible for efflux of a 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 co-worker 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). Downloaded from

We further analyzed the results of various chemical inhibitors on the excretion to jpet.aspetjournals.org 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 at ASPET Journals on September 29, 2021 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 to 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

19 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925 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 µM cyclosporine A using Caco-2 cell lysate (27% decrease), which suggest that cyclosporine A did not enhance the metabolite formation. The actual mechanism for sulfate efflux enhancement remains to be determined.

Downloaded from

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 jpet.aspetjournals.org 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 (Fig.6 and at ASPET Journals on September 29, 2021

Fig.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 get out of 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

20 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925 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 to 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 sulftases as

proposed (Hochner-Celnikier, 1999), resulting in higher than expected excretion of conjugates Downloaded from

(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 jpet.aspetjournals.org 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 at ASPET Journals on September 29, 2021 transporters.

The coupling of efflux transporters and conjugating enzymes also support our hypothesis that enteric recycling (Liu and Hu, 2002) is important for the 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 or be hydrolyzed by the microflora.

21 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

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 allow more intact Downloaded from 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 jpet.aspetjournals.org 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. at ASPET Journals on September 29, 2021

In conclusion, raloxifene and its phase II conjugates are excreted by the intestinal cells via the actions of 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 transporters 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.

22 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

REFERENCES

Bohets H, Annaert P, Mannens G, Van Beijsterveldt L, Anciaux K, Verboven P, Meuldermans

W, Lavrijsen K (2001) Strategies for absorption screening in drug discovery and

development. Curr Top Med Chem. 1: 367-83

Caro I, Boulenc X, Rousset M, Meunier V, Bourrie M, Julian B, Joyeux H, Roques C, Berger Y, Downloaded from

Zwebaum A and Fabre G (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 jpet.aspetjournals.org

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 at ASPET Journals on September 29, 2021

Colacurci N, Manzella D, Fornaro F, Carbonella M and Paolisso G (2003) Endothelial function

and : effects of raloxifene administration. J Clin Endocrinol Metab. 88:2135-40

Cummins CL, Jacobsen W and Benet LZ (2002) Unmasking the dynamic interplay 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 Eng J Med.

337:1641-1647

23 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

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, Fabre G Downloaded from

(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 jpet.aspetjournals.org line. Pharm Res. 15: 726-733

Hochner-Celnikier D (1999) Pharmacokinetics of raloxifene and its clinical application. Eur J at ASPET Journals on September 29, 2021

Obstet Gynecol Reprod Biol. 85:23-29

Hu M, Chen J, Tran D, Zhu Y and Leonardo G (1994b) 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, Zhu Y, Dantzig AH, Stratford RE and Kuhfeld MT Jr. (1994a) Mechanism and

kinetics of transcellular transport of a new β-lactam antibiotic loracarbef across a human

intestinal epithelial model system (Caco-2). Pharm Res. 11:1405-1413

24 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

Hu M, Li Y, Davitt CM, Huang SM, Thummel K, Penman BW, Crespi CL (1999) Transport and

metabolic characterization of Caco-2 cells expressing CYP3A4 and CYP3A4 plus

oxidoreductase. Pharm Res. 16: 1352-1359

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 Downloaded from

Jordan VC and Morrow M (1999) , raloxifene, and the prevention of breast cancer. jpet.aspetjournals.org Endocr Rev. 20:253-278

Kemp DC, Fan PW and Stevens JC (2002) Characterization of raloxifene glucuronidation in at ASPET Journals on September 29, 2021

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 in rats, dogs and monkeys. Xenobiotica. 14:841-847

Liu Y, Hu M (2002) Absorption and metabolism of flavonoids in the caco-2 cell culture model

and a perused rat intestinal model. Drug Metab Dispos. 30:370-7

MacGregor JI and Jordan C (1998) Basic guide to the mechanisms of antiestrogen action.

Pharmacol Rev. 50:151-196

25 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

Pfrunder A, Gutmann H, Beglinger C, 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, Arnaud P (2001) HT29-MTX and Caco-2/TC7

monolayers as predictive models for human intestinal absorption: role of the mucus layer. J Downloaded from

Pharm Sci. 90: 1608-1619

jpet.aspetjournals.org Ranaldi G, Consalvo R, Sambuy Y, 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 at ASPET Journals on September 29, 2021

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

26 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

Sabolovic N, Magdalou J, Netter P, Abid A (2000). Nonsteroidal anti-inflammatory 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, Walle T. (1999). Transport of the chrysin and its conjugated

metabolites by the human intestinal cell line Caco-2. Biochem Pharmacol. 58:431-8.

Downloaded from

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 jpet.aspetjournals.org resistance-associated protein. Int J Clin Pharmacol Ther. 38:122-129

at ASPET Journals on September 29, 2021

27 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

FOOTNOTES: This study was supported by National Institutes of Health (NIH) Grant CA 87779.

Downloaded from

jpet.aspetjournals.org

at ASPET Journals on September 29, 2021

28 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

LEGENDS FOR FIGURES:

Fig. 1. Representative HPLC profiles of raloxifene and its metabolites. Panel A shows the peaks of major metabolites (i.e., glucuronide and sulfate) after extracting raloxifene with methylene chloride from a 24 h Caco-2 incubation sample. Panel B shows disappearance of glucuronide peak and appearance of raloxifene peak after treatment with glucuronidase. Panel C shows

reduction of sulfate peak and appearance of raloxifene peak after treatment with sulfatase. Downloaded from

Fig 2. Transport and excretion of raloxifene and its metabolites in the Caco-2 model. Raloxifene jpet.aspetjournals.org (17 µM) was added to 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 at ASPET Journals on September 29, 2021 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 standard deviations of the mean. Each line represents linear regression of the data points. Panel A, R2=0.9000, 0.99421 and 0.9900 for raloxifene, glucuronide and sulfate; panel B, R2=0.9812 and 0.9805 for glucuronide and sulfate; panel C,

R2=0.9827, 0.9929 and 0.9902 for raloxifene, glucuronide and sulfate; panel D, R2=0.9972 and

0.9955 for glucuronide and sulfate, respectively.

Fig. 3. Effect of concentration on the absorptive transport of raloxifene. Raloxifene (1.5 ~30

µM) was added to apical (AP) side of the Caco-2 cell monolayer and the amount of raloxifene in

29 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925 receiver (BL) side were 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 the equation [1] and [2] as described under Materials and Methods. Each data point represents the mean of three determinations and the error bars are standard deviations of the mean. The line and curve represent linear and polynomial regression of the data points in panel A (R2=0.9790) and panel

B(R2=0.9985), respectively.

Downloaded from

Fig. 4. Effect of raloxifene concentration on the clearance of raloxifene metabolites to apical

(AP) or basolateral (BL) side of the Caco-2 cell monolayer. Raloxifene (1.5 ~30 µM) was added jpet.aspetjournals.org to apical side of the Caco-2 cell monolayer and the amount of raloxifene glucuronide and sulfate in receiver (basolateral) and donor (apical) side were determined at 1, 2, 3 and 4 h after incubation. Metabolic clearance (CL) of raloxifene was calculated according to the equation [3] at ASPET Journals on September 29, 2021 described under Materials and Methods. Each data point represents the mean of three determinations and the error bars are standard deviations of the mean. Each curve represents power regression of the data points using Microsoft Excel. Panel A, R2=0.8243 and 0.9563 for glucuronide and sulfate; panel B, R2=0.8973 and 0.8915 for glucuronide and sulfate, respectively.

The star symbol “*” indicates statistically significant differences (p < 0.05) between glucuronide and sulfate.

Fig. 5. Effect of multidrug resistance related protein (MRP) and P-glycoprotein inhibitors on the permeability of raloxifene and clearance of its metabolites in Caco-2 cells. Raloxifene (7 µM) was added to apical (AP) or basolateral (BL) side of the Caco-2 cell monolayer. Fifty µM MK-

571 plus 0.1 µM leukotriene C4, 20 µM cyclosporine A or 20 µM verapamil was added to both

30 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925 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 the equation [3] described under Materials and Methods. Each column represents the mean of three determinations and the error bars are standard deviations of the mean. Blank, solid, dashed and dotted columns represent the control, MK-571 plus leukotriene C4, cyclosporine A or verapamil treated group, respectively. The star symbol “*” indicates a statistically significant difference (p

< 0.05) compared to the control (i.e., raloxifene only). Downloaded from

Fig. 6. Effect of organic anionic transporter (OAT) inhibitors on the transport of raloxifene and jpet.aspetjournals.org clearance of its metabolites in Caco-2 cells. Raloxifene (7 µM) was added to apical side of the

Caco-2 cell monolayer. 50 µM estrone glucuronide (EG) or estrone sulfate (ES) was added to both sides of the Caco-2 cell monolayer. Rate of transport (Bt) and metabolic clearance (CL) of at ASPET Journals on September 29, 2021 raloxifene were calculated using the amounts of raloxifene and metabolites at 1, 2, 3 and 4 h after incubation according to the equation [1] and [3] described under Materials and Methods.

Each column represents the mean of three determinations and the error bars are standard deviations 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 Bt and CLs were 0.059±

0.013 nmol/h (panel A), 3.10 ± 0.10x10-6 ml/sec 11.16±0.36 x µl/hr (glucuronide, basolateral or

BL side, panel B), 10.04 ± 1.37 µl/hr (glucuronide, apical side or AP, panel B), 11.23 ± 0.47

µl/hr or (sulfate, basolateral side, panel C) and 26.57 ± 1.12 µl/hr (sulfate, apical side, panel C), respectively. The star symbol “*” indicates a statistically significant difference (p < 0.05) compared to the control.

31 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

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 µM) was added to apical

(AP) side of the Caco-2 cell monolayer. 1 ~ 50 µM estrone sulfate was added to both sides of the

Caco-2 cell monolayer. Rate of transport (B) 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 the equation [1] and [3] described under Materials and Methods. Each data point

represents the mean of three determinations and the error bars are standard deviations of the Downloaded from mean. . The mean control values (± S.D.) of Bt and CLs were 0.059± 0.013 nmol/h (panel A),

3.10 ± 0.10x10-6 ml/sec 11.16±0.36 x µl/hr (glucuronide, basolateral side, panel B), 10.04 ± 1.37 jpet.aspetjournals.org

µl/hr (glucuronide, apical side, panel B), 11.23 ± 0.47 µl/hr or (sulfate, basolateral side, panel C) and 26.57 ± 0.1.12 µl/hr (sulfate, apical side, panel C), respectively. Each curve represents

2 rectangular regression of the data points using SigmaPlot. Panel A, R =0.9680 and EC50=16.4 at ASPET Journals on September 29, 2021

µM; panel B, R2=0.9639 for basolateral side and maximum decrease of ≈40%; panel C,

R2=0.8435 and 0.8168 for basolateral and apical side and maximum decrease of ≈50% for both sides, respectively. The solid and open symbols indicate basolateral and apical side, respectively.

The star symbol “*” indicates a statistically significant difference (p < 0.05) compared to the control.

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 µM) and ES (20 µM) 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

32 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

metabolism (Bmet) 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 µM) was added to apical (AP) side and estrone sulfate (20 µM) was added to both sides of the Caco-2 cell monolayer. The sum of donor

(AP) side and receiver (basolateral, BL) side excretion rate (Bex) of each metabolite was calculated using the amounts of metabolites at 1, 2, 3 and 4 hour after incubation as described

under Materials and Methods. Each column represents the mean of three determinations and the Downloaded from error bars are standard deviations of the mean. Blank and solid columns represent rate of metabolism and excretion, respectively. Panel A, Bex/Bmet = 3.4 and 3.7 for the control and ES- jpet.aspetjournals.org treated group; panel B, Bex/Bmet = 1.3 and 0.8 for the control and ES-treated group, respectively.

The star symbol “*” and “**” indicate statistically significant differences (p < 0.05) between the control and ES-treated group and between metabolism and excretion, respectively. at ASPET Journals on September 29, 2021

Fig. 9. Effect of raloxifene concentration on the accumulation of raloxifene and its sulfate by the

Caco-2 cells. Raloxifene (1.5 ~18 µM) was added to 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 standard deviations of the mean. Each curve represents rectangular regression of the data points using SigmaPlot. Panel A, R2=0.9681; panel B, R2=0.9720. The star symbol “*” indicates a statistically significant difference (p <

0.05) compared to the lower concentration.

33 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

Fig. 10. Schematic representation of intestinal disposition of raloxifene. Absorbed raloxifene, in free or conjugated form, will be effluxed to the lumen (AP) or be transported into the 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-mediated, MRP-mediated or OAT-mediated efflux) responsible for Downloaded from intestinal disposition of raloxifene and/or its conjugates.

jpet.aspetjournals.org

at ASPET Journals on September 29, 2021

34 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

JPET #63925

Table 1. Absorptive (apical to basolateral: BAB) and secretory (basolateral to apical: BBA) 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, 2, 3 and 4 h) in the amount transported versus time plot. Some cells were treated with 17 µM raloxifene (both sides) for 24 h before the transport experiment. Data are presented as the mean ± standard deviation of three determinations.

Downloaded from Raloxifene BAB BBA Cell Ratio (µM) (nmol/h) (nmol/h)

17 Normal cell 0.498 ± 0.094 0.757 ± 0.098 152%* jpet.aspetjournals.org

7 Normal cell 0.088 ± 0.036 0.301 ± 0.011 344%*

7 Raloxifene, 24 h 0.093 ± 0.032 0.334 ± 0.016** 360%*

at ASPET Journals on September 29, 2021

* P < 0.05 between BAB and BBA

** P < 0.05 between normal and treated cell

Page of 3 35 JPET #63925

Table 2. Absorptive permeability, amounts transported intact and amounts of raloxifene metabolites formed after 2 h Drug was added to apical side of the Caco-2 monolayer. Permeability values were calculated using first four data (time= 1, 2, 3 and 4 This articlehasnotbeencopyeditedandformatted.Thefinalversionmaydifferfromthisversion. 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 JPET FastForward.PublishedonMarch12,2004asDOI:10.1124/jpet.103.063925 metabolites in the apical (AP) and basolateral (BL) media after 2 h-incubation were determined. Data are presented as the mean ± standard deviation of three determinations. Permeability Glucuronide (nmol) Sulfate (nmol) Total Amounts (nmol) % Metabolism Compound (10-7 cm/sec) AP side BL side AP side BL side Parent Conjugates Parent/(Total)

0.419 ± Raloxifene, 1.5 µM UDb UD UD UD - - - 0.052 Raloxifene, 3.4 µM 3.88 ± 0.83c 0.136 ± 0.159 ± 0.635 ± 0.172 ± 0.041 1.102 96.4 0.007 0.018 0.040 0.033 Raloxifene, 6 µM 7.87 ± 2.05 c 0.168 ± 0.321 ± 0.400 ± 0.232 ± 0.132 1.121 89.4 0.025 0.032 0.025 0.001 Raloxifene, 7 µM 8.66 ± 3.60 c 0.168 ± 0.190 ± 0.781 ± 0.296 ± 0.173 1.089 86.3 0.020 0.012 0.047 0.035 Raloxifene, 17 µM 17.8 ± 6.96 c 0.092± 0.240 ± 0.308 ± 0.338 ± 0.897 0.979 52.2 0.008 0.024 0.014 0.028 Raloxifene, 30 µM 41.4 ± 7.28 c 0.165 ± 0.307 ± 0.222 ± 0.392 ± 3.782 1.085 22.3 0.165 0.016 0.011 0.037 Apigenin, 35 µMa 169 ± 10 3.42 ± 4.09 ± 3.75 ± 3.21 ± 20.94 17.61 45.7 0.35 0.18 0.22 0.26 Genistein, 35 µMa 309 ± 90 0.867 ± 1.68 ± 0.314 ± 0.709 ± 33.39 3.9 10.5 0.014 0.04 0.021 0.017 a. Chen et al., 2003

Page of 3 36

Downloaded from from Downloaded jpet.aspetjournals.org at ASPET Journals on September 29, 2021 29, September on Journals ASPET at JPET #63925

b. Under detection limit c.

Significant difference with respect to raloxifene concentration according to one-way ANOVA (p < 0.05). This articlehasnotbeencopyeditedandformatted.Thefinalversionmaydifferfromthisversion.

JPET FastForward.PublishedonMarch12,2004asDOI:10.1124/jpet.103.063925

Page of 3 37

Downloaded from from Downloaded jpet.aspetjournals.org at ASPET Journals on September 29, 2021 29, September on Journals ASPET at JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

mAu Downloaded from raloxifene 8

C

6 jpet.aspetjournals.org

4 internal B standard

2 A at ASPET Journals on September 29, 2021 glucuronide sulfate 0 0 5 10 15 20min 25

Fig. 1 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

(A) A-->B Receiver (BL) 50 (B) A-->B Donor (AP) 5 5 40 4 4 30 3 3

20 2 2 raloxifene or or raloxifene raloxifene (nmol) 1 10 1 metabolites (nmol) metabolites (nmol)

0 0 0 1234 1234 Time (h) Time (h)

5 Downloaded from (C) B-->A Receiver (AP) 50 (D) B-->A Donor (BL) 5 4 40 4

3 30 3

2 20 2 raloxifene or raloxifene jpet.aspetjournals.org metabolites (nmol) 1 raloxifene (nmol) 10 1 metabolites (nmol)

0 0 0 1234 1234 Time (h) Time (h) at ASPET Journals on September 29, 2021

Fig. 2 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

50 A: Permeability

7 40

30

20

P (cm/sec) X 10 P (cm/sec) 10 Downloaded from 0 0 5 10 15 20 25 30 35 Raloxifene (µM)

3 jpet.aspetjournals.org B: Rate of Transport

2

B (nmol/h) 1 at ASPET Journals on September 29, 2021

0 0 5 10 15 20 25 30 35 Raloxifene (µM)

Fig. 3 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

60 A: AP sid e * Glucuronide 6 40 Sulfate * /sec)X10 3 20 * *

CL (cm CL * *

0 Downloaded from 0 20406080100 Raloxifene (µM)

12 B: BL side

6 Glucuronide 10 jpet.aspetjournals.org Sulfate 8 /sec)X10

3 6 4

CL (cm CL 2 at ASPET Journals on September 29, 2021 0 0 20406080100 Raloxifene (µM)

Fig. 4 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

300% * A: Raloxifene Raloxifene only * with LTC4 & MK-571 200% with cyclosporin A with verapamil

100%

P (% control) * Downloaded from

0% PAB PBA

300% B: Glucuronide

200% jpet.aspetjournals.org

100% CL (% control) CL * * * * 0% AP-->BL (BL side) AP-->BL (AP side) BL-->AP (AP side) BL-->AP (BL side)

300% C: Sulfate at ASPET Journals on September 29, 2021 *

200%

100%

CL control) (% * * 0% AP-->BL (BL side) AP-->BL (AP side) BL-->AP (AP side) BL-->AP (BL side)

Fig. 5 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

A: Raloxifene Control EG ES 300% * 200% *

100% B (% control) (% B

0% Downloaded from Control EG ES

300% B: Glucuronide

200% * jpet.aspetjournals.org

100% * CL (% control)

0% BL side AP side at ASPET Journals on September 29, 2021 150% C. Sulfate

100% * * * 50% * CL (% control) 0% BL side AP side

Fig. 6 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

250 A: Raloxifene * 200 * * 150 *

100 B (% B (% control) B (% B (% control) B (% B (% control) B (% B (% control) 50

0 Downloaded from 0 1020304050

200 B: Glucuronide 150 jpet.aspetjournals.org 100 CL (% control) (% CL CL (% control) (% CL CL (% control) (% CL 50 ***** 0 0 1020304050 at ASPET Journals on September 29, 2021 120 C: Sulfate 100

80 * * * 60 **

40 CL (% CLcontrol) (% CL (% CLcontrol) (% CL (% CLcontrol) (% CL (% CLcontrol) (% * * 20 * * *

0 0 1020304050 Estrone Sulfate (µM)

Fig.7 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

15 A: Glucuronide Metabolism Excretion ** ** 10

* Downloaded from 5

pmol/min/monolayer * 0 Control ES jpet.aspetjournals.org

20 B: Sulfate **

ayer 15 **

10

* at ASPET Journals on September 29, 2021 * 5 pmol/min/monol 0 Control ES

Fig. 8 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

50 A: Raloxifene 40

30 * 20 * Downloaded from

nmol / monolayer nmol / 10

0 0 102030405060 jpet.aspetjournals.org 10 B: Sulfate

8 * 6

4

* at ASPET Journals on September 29, 2021

nmol / monolayer / nmol 2

0 0 102030405060 Raloxifene (µM)

Fig. 9 JPET Fast Forward. Published on March 12, 2004 as DOI: 10.1124/jpet.103.063925 This article has not been copyedited and formatted. The final version may differ from this version.

LUMEN Microflora Phase II Conjugates (Apical) Raloxifene

MDR1 MRP2 OAT ? OAT MRP2 WALL UGT Glucuronide Raloxifene Downloaded from Sulfate SULT MRP3 ? OAT Basolateral jpet.aspetjournals.org

MESSENTARY

Portal Vein Bile LIVER at ASPET Journals on September 29, 2021

Heart SYSTEMIC CIRCULATION (Bioavailable)

Fig. 10