quantitative determination of absorption and first-pass...

9
1521-009X/42/6/974982$25.00 http://dx.doi.org/10.1124/dmd.113.056713 DRUG METABOLISM AND DISPOSITION Drug Metab Dispos 42:974982, June 2014 Copyright ª 2014 by The American Society for Pharmacology and Experimental Therapeutics Quantitative Determination of Absorption and First-Pass Metabolism of Apicidin, a Potent Histone Deacetylase Inhibitor s Beom Soo Shin, Sun Dong Yoo, Tae Hwan Kim, Jurgen B. Bulitta, Cornelia B. Landersdorfer, Jeong Cheol Shin, Jin Ho Choi, Kwon-Yeon Weon, Sang Hoon Joo, and Soyoung Shin College of Pharmacy, Catholic University of Daegu, Gyeongsan-si, Gyeongbuk, South Korea (B.S.S., J.C.S., J.H.C., K.Y.W., S.H.J.); School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, South Korea (S.D.Y., T.H.K.); Centre for Medicine Use and Safety, Monash University, Parkville, Australia (J.B.B., C.B.L.); Department of Pharmacy, Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk, South Korea (S.S.) Received December 21, 2013; accepted March 18, 2014 ABSTRACT Apicidin, a potential oral chemotherapeutic agent, possesses potent anti-histone-deacetylase activity. After oral administration, the total bioavailability of apicidin is known to be low (14.2%19.3%). In the present study, we evaluated the factors contributing to the low bioavailability of apicidin by means of quantitative determination of absorption fraction and first-pass metabolism after oral administration. Apicidin was given to rats by five different routes: into the femoral vein, duodenum, superior mesenteric artery, portal vein, and carotid artery. Especially, the fraction absorbed (F X ) and the fraction that is not metabolized in the gut wall (F G ) were separated by injection of apicidin via superior mesenteric artery, which enables bypassing the permeability barrier. The F X was 45.9% 6 9.7%, the F G was 70.9% 6 8.1% and the hepatic bioavailability (F H ) was 70.6% 6 12.3%, while the pulmonary first- pass metabolism was minimal (F L = 102.8% 6 7.4%), indicating that intestinal absorption was the rate-determining step for oral absorption of apicidin. The low F X was further examined in terms of passive diffusion and transporter-mediated efflux by in vitro immobilized artificial membrane (IAM) chromatographic assay and in situ single-pass perfusion method, respectively. Although the passive diffusion potential of apicidin was high (98.01%) by the IAM assay, the in situ permeability was significantly enhanced by the presence of the P-glycoprotein (P-gp) inhibitor elacrider. These data suggest that the low bioavailability of apicidin was mainly attributed to the P-gp efflux consistent with the limited F X measured in vivo experiment. Introduction Inhibitors of histone deacetylases (HDACs) have been identified as a novel class of chemotherapeutics for the treatment of various cancers. To date, two of the HDAC inhibitors, Vorinostat (Merck & Co., Whitehouse Station, NJ) and Romidepsin (Gloucester Pharma- ceutical Inc., Cambridge, MA) have been approved by the U.S. Food and Drug Administration (FDA) for the treatment of cutaneous T-cell lymphoma. More than 15 HDAC inhibitors including Panobinostat (LBH589), Entinostat (MS-275), Belinostat (PXD101), Givinostat (ITF2357), Mocetinostat (MGCD0103), and Abexinostat (PCI-24781) have been tested in preclinical and clinical studies (Tan et al., 2010; Kim and Bae, 2011). Currently, 104 clinical studies involving HDAC inhibitors have been completed, and over 100 trials are ongoing to develop these inhibitors as a monotherapy or in combination with other anticancer agents. This mechanism of cancer treatment has been recognized as a promising new direction in developing novel anticancer agents. Moreover, some of these HDAC inhibitors are orally available, which is of particular merit in the new drug development. Apicidin is one of the HDAC inhibitors that has potential to be developed as an oral chemotherapeutic agent with potent antiprolifer- ative and cytodifferentiation activities (Darkin-Rattray et al., 1996). It has been shown to exhibit antitumor effects in several human cancer cell lines, including leukemia (Kwon et al., 2002), ovarian (Ahn et al., 2012), endometrial (Ahn et al., 2010), oral squamous (Ahn et al., 2011), cervical (Han et al., 2000; Luczak and Jagodzinski, 2008), and breast (Han et al., 2000) cancer cell lines in vitro and in vivo (Lai et al., 2009). The therapeutic potential of apicidin also appears promising in combination therapy with other agents such as docetaxel (Buoncervello et al., 2012), doxorubicin (Lai et al., 2009), and tumor necrosis factorrelated apoptosis-inducing ligand (Park et al., 2009). However, only a few reports are available about the pharmacokinetic characteristics of apicidin. In our previous study, the oral bio- availability of apicidin was found to be 19.3% and 14.2% in fasting and nonfasting rats, respectively (Shin et al., 2006). A fundamental understanding of the absorption processes as well as the factors responsible for the relatively low oral bioavailability is critical for apicidin to be developed as an orally available chemotherapeutic agent. This work was supported by the National Research Foundation of Korea (NRF) grants funded by the Korea government (MEST) [NRF-2012R1A1A2008588, NRF- 2012R1A1A1044923]. B.S.S. and S.D.Y. contributed equally to this work. dx.doi.org/10.1124/dmd.113.056713. s This article has supplemental material available at dmd.aspetjournals.org. ABBREVIATIONS: AUC, area under the time-concentration curve; CL, clearance; F X, fraction absorbed; F G, fraction not metabolized in the gut wall; F H , hepatic bioavailability; F L , pulmonary bioavailability; HDAC, histone deacetylase; HPLC, high-performance liquid chromatography; ITF2357, Givinostat; LBH589, Panobinostat; LC-MS/MS, liquid chromatography coupled with tandem mass spectrometry; MGCD0103, Mocetinostat; MRT, mean residence time; MS-275, Entinostat; PE, polyethylene; P-gp, P-glycoprotein; PCI-24781, Abexinostat; PXD101, Belinostat. 974 http://dmd.aspetjournals.org/content/suppl/2014/03/18/dmd.113.056713.DC1 Supplemental material to this article can be found at: at ASPET Journals on July 3, 2018 dmd.aspetjournals.org Downloaded from

Upload: trinhdiep

Post on 29-May-2018

226 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Quantitative Determination of Absorption and First-Pass ...dmd.aspetjournals.org/content/dmd/42/6/974.full.pdf · Quantitative Determination of Absorption and First ... by means of

1521-009X/42/6/974–982$25.00 http://dx.doi.org/10.1124/dmd.113.056713DRUG METABOLISM AND DISPOSITION Drug Metab Dispos 42:974–982, June 2014Copyright ª 2014 by The American Society for Pharmacology and Experimental Therapeutics

Quantitative Determination of Absorption and First-Pass Metabolismof Apicidin, a Potent Histone Deacetylase Inhibitor s

Beom Soo Shin, Sun Dong Yoo, Tae Hwan Kim, Jurgen B. Bulitta, Cornelia B. Landersdorfer,Jeong Cheol Shin, Jin Ho Choi, Kwon-Yeon Weon, Sang Hoon Joo, and Soyoung Shin

College of Pharmacy, Catholic University of Daegu, Gyeongsan-si, Gyeongbuk, South Korea (B.S.S., J.C.S., J.H.C., K.Y.W.,S.H.J.); School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, South Korea (S.D.Y., T.H.K.); Centre for MedicineUse and Safety, Monash University, Parkville, Australia (J.B.B., C.B.L.); Department of Pharmacy, Institute of Pharmaceutical

Research and Development, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk, South Korea (S.S.)

Received December 21, 2013; accepted March 18, 2014

ABSTRACT

Apicidin, a potential oral chemotherapeutic agent, possessespotent anti-histone-deacetylase activity. After oral administration,the total bioavailability of apicidin is known to be low (14.2%–

19.3%). In the present study, we evaluated the factors contributingto the low bioavailability of apicidin by means of quantitativedetermination of absorption fraction and first-pass metabolismafter oral administration. Apicidin was given to rats by five differentroutes: into the femoral vein, duodenum, superior mesentericartery, portal vein, and carotid artery. Especially, the fractionabsorbed (FX) and the fraction that is not metabolized in the gut wall(FG) were separated by injection of apicidin via superior mesentericartery, which enables bypassing the permeability barrier. The FXwas 45.9% 6 9.7%, the FG was 70.9% 6 8.1% and the hepatic

bioavailability (FH) was 70.6% 6 12.3%, while the pulmonary first-pass metabolism was minimal (FL = 102.8%6 7.4%), indicating thatintestinal absorption was the rate-determining step for oralabsorption of apicidin. The low FX was further examined in termsof passive diffusion and transporter-mediated efflux by in vitroimmobilized artificial membrane (IAM) chromatographic assay andin situ single-pass perfusion method, respectively. Although thepassive diffusion potential of apicidin was high (98.01%) by the IAMassay, the in situ permeability was significantly enhanced by thepresence of the P-glycoprotein (P-gp) inhibitor elacrider. Thesedata suggest that the low bioavailability of apicidin was mainlyattributed to the P-gp efflux consistent with the limited FX measuredin vivo experiment.

Introduction

Inhibitors of histone deacetylases (HDACs) have been identified asa novel class of chemotherapeutics for the treatment of variouscancers. To date, two of the HDAC inhibitors, Vorinostat (Merck &Co., Whitehouse Station, NJ) and Romidepsin (Gloucester Pharma-ceutical Inc., Cambridge, MA) have been approved by the U.S. Foodand Drug Administration (FDA) for the treatment of cutaneous T-celllymphoma. More than 15 HDAC inhibitors including Panobinostat(LBH589), Entinostat (MS-275), Belinostat (PXD101), Givinostat(ITF2357), Mocetinostat (MGCD0103), and Abexinostat (PCI-24781)have been tested in preclinical and clinical studies (Tan et al., 2010;Kim and Bae, 2011). Currently, 104 clinical studies involving HDACinhibitors have been completed, and over 100 trials are ongoing todevelop these inhibitors as a monotherapy or in combination withother anticancer agents. This mechanism of cancer treatment has been

recognized as a promising new direction in developing novel anticanceragents. Moreover, some of these HDAC inhibitors are orally available,which is of particular merit in the new drug development.Apicidin is one of the HDAC inhibitors that has potential to be

developed as an oral chemotherapeutic agent with potent antiprolifer-ative and cytodifferentiation activities (Darkin-Rattray et al., 1996). Ithas been shown to exhibit antitumor effects in several human cancercell lines, including leukemia (Kwon et al., 2002), ovarian (Ahn et al.,2012), endometrial (Ahn et al., 2010), oral squamous (Ahn et al.,2011), cervical (Han et al., 2000; Luczak and Jagodzinski, 2008), andbreast (Han et al., 2000) cancer cell lines in vitro and in vivo (Laiet al., 2009). The therapeutic potential of apicidin also appearspromising in combination therapy with other agents such as docetaxel(Buoncervello et al., 2012), doxorubicin (Lai et al., 2009), and tumornecrosis factor–related apoptosis-inducing ligand (Park et al., 2009).However, only a few reports are available about the pharmacokineticcharacteristics of apicidin. In our previous study, the oral bio-availability of apicidin was found to be 19.3% and 14.2% in fastingand nonfasting rats, respectively (Shin et al., 2006). A fundamentalunderstanding of the absorption processes as well as the factorsresponsible for the relatively low oral bioavailability is critical forapicidin to be developed as an orally available chemotherapeuticagent.

This work was supported by the National Research Foundation of Korea (NRF)grants funded by the Korea government (MEST) [NRF-2012R1A1A2008588, NRF-2012R1A1A1044923].

B.S.S. and S.D.Y. contributed equally to this work.dx.doi.org/10.1124/dmd.113.056713.s This article has supplemental material available at dmd.aspetjournals.org.

ABBREVIATIONS: AUC, area under the time-concentration curve; CL, clearance; FX, fraction absorbed; FG, fraction not metabolized in the gut wall;FH, hepatic bioavailability; FL, pulmonary bioavailability; HDAC, histone deacetylase; HPLC, high-performance liquid chromatography; ITF2357,Givinostat; LBH589, Panobinostat; LC-MS/MS, liquid chromatography coupled with tandem mass spectrometry; MGCD0103, Mocetinostat; MRT,mean residence time; MS-275, Entinostat; PE, polyethylene; P-gp, P-glycoprotein; PCI-24781, Abexinostat; PXD101, Belinostat.

974

http://dmd.aspetjournals.org/content/suppl/2014/03/18/dmd.113.056713.DC1Supplemental material to this article can be found at:

at ASPE

T Journals on July 3, 2018

dmd.aspetjournals.org

Dow

nloaded from

Page 2: Quantitative Determination of Absorption and First-Pass ...dmd.aspetjournals.org/content/dmd/42/6/974.full.pdf · Quantitative Determination of Absorption and First ... by means of

The absorption process can be affected by a number of factors suchas solubility, permeability, transporters, and metabolism. After oraladministration, a drug must pass through the gastrointestinal lumen,transit through the gut wall, and withstand the initial degradation byintestinal mucosal enzymes. It may be metabolized while penetratingthe gut wall, and the drug that reaches the portal vein may be affectedby hepatic metabolism or biliary extraction. The portion escaping thehepatic elimination may also be subject to extrahepatic metabolism.Moreover, efflux transporters and drug metabolizing enzymes oftenwork together for xenobiotics, which may confound the interpretationof the in vivo experiments. The relative contributory roles of thesefactors leading to the poor bioavailability of apicidin are still unclear.Especially, distinguishing between the contributions of gut wallabsorption and first-pass metabolism, which compose the gastrointes-tinal bioavailability, will provide useful information for the de-velopment of novel apicidin analogs with improved bioavailability.Nevertheless, experimental separation of the fraction absorbed—thatis, the net transport of unchanged drug into the gastrointestinal tract(FX)—and the fraction that is not metabolized through the gut wall(FG) in vivo is not easily achieved, and most of the literature reportsthem together as gastrointestinal bioavailability (FX·FG) (Raoof et al.,1996; Hashimoto et al., 1998; Mihara et al., 2001; Choi et al., 2006;Hanada et al., 2008; Bae et al., 2009; Gertz et al., 2011).Our study quantitatively evaluated the contributory roles of the

absorption process and first-pass metabolism of apicidin to its oralbioavailability after oral administration. We administered apicidin byvarious routes, including the femoral vein, duodenum, superiormesentery artery, portal vein, and carotid artery, to distinguish thefractions absorbed through the gut wall and metabolized at differentsites after oral administration. Particularly, superior mesenteric arterialinjection of apicidin allowed us to determine the in vivo gastrointes-tinal absorption and gut wall first-pass metabolism separately. To bestof our knowledge, this is the first report of the in vivo approach forthe quantitative determination of FX and FG by comparing the areaunder the time-concentration curve (AUC) obtained via differentroutes of administration. In vitro and in situ studies were also con-ducted to confirm the results from the in vivo experiment and tofurther assess the factors contributing to the in vivo absorption such as,passive diffusion, efflux by P-glycoprotein (P-gp), and gastrointestinalstability.

Materials and Methods

Chemicals and Reagents. Apicidin was prepared from Fusarium sp. strainKCTC 16677 according to a method described previously (Park et al., 1999).HPLC grade acetonitrile, ethanol, and t-butyl methyl ether were purchased fromJ.T. Baker (Phillipsburg, NJ). The other reagents were purchased from Sigma-Aldrich (St. Louis, MO).

Animals. Male Sprague-Dawley rats (8–10 weeks old, body weight 220–272 g) were kept in plastic cages with free access to standard rat diet (Samyang,Seoul, South Korea) and water. The animals were maintained at a temperatureof 22–24°C with a 12-hour light/dark cycle and relative humidity of 50% 610% before the experiment. All animal studies were approved by the ethicscommittee for the treatment of laboratory animals at Sungkyunkwan Universityand Catholic University of Daegu.

Determination of Passive Absorption Potential by Immobilized ArtificialImmobilized Artificial Membrane Chromatographic Assay. The absorptionpotential of apicidin by passive diffusion was evaluated by the immobilizedartificial membrane (IAM) chromatographic assay described previously else-where (Yoon et al., 2004). Briefly, high-performance liquid chromatography(HPLC) analysis was performed using a Waters 2695 separations module andWaters 2996 photo diode array detector (Waters Corporation, Milford, MA).The retention time of apicidin (tr) and the holdup time of the column (t0) weredetermined using the IAM.PC.DD 2 Drug-Discovery column (Regis Technologies,

Morton Grove, IL). The isocratic mobile phase consisted of acetonitrile andDulbecco’s phosphate-buffered saline (80:20 v/v). The flow rate of the mobilephase was 1.0 ml/min, and the column oven temperature was 37°C. Apicidin wasdetected at 210 nm. The IAM capacity factors (KIAM) were determined for 28commercial drugs, and the obtained KIAM was modified by the power of thecompound molecular weight (KIAM/MWn) to improve the correlation between theKIAM and the dose absorbed in humans (Fa). The human Fa was obtained fromthe literature (Worland et al., 1984; Sugano et al., 2001; Zhu et al., 2002). Therelationship between modified KIAM (KIAM/MWn) and Fa was best described bythe following sigmoid equation:

Fað%Þ ¼ 98:01��12 e2 4:30� KIAM

MW2:05

�3:00

ð1Þ

where KIAM is the IAM capacity factor and MW is the molecular weight of thecompound. The Fa for apicidin by passive diffusion was calculated using eq. 1.

Determination of Intestinal Permeability by Single-Pass Perfusion. Theeffect of P-gp on the intestinal absorption of apicidin was evaluated by meansof a single-pass perfusion method in the absence or presence of the P-gpinhibitor elacrider. After overnight fasting, the rats were anesthetized byintraperitoneal injection of Zoletil 50 (20 mg/kg; Virbac Laboratories, Carros,France). The abdomen was opened and the intestinal segment of the duodenum(10 cm length from the end of the stomach) was isolated and cannulated at bothends with a silicone tube (2 mm i.d.; Daihan Scientific Co., Wonjoo, SouthKorea). The duodenal segment was rinsed with 37°C saline to clear the segmentbefore perfusion. Perfusate was prepared by dissolving apicidin at a concen-tration of 10 mM in normal saline in the absence or presence of elacrider(1 mM). The perfusion rate was 0.2 ml/min, and the outlet perfusates werecollected at 15-minute intervals from 50 minutes to 120 minutes after initiationof infusion. Collected samples were stored at 220°C until analysis. To maintainanesthesia, Zoletil 50 was administered via the intraperitoneal route every 30minutes, and the animal was placed on a heating pad to maintain the bodytemperature. The abdominal incision area was stapled to prevent loss of fluid andhypothermia. The effective permeability (Peff) was calculated by the single-passperfusion approach:

Peff ¼�2Qin � Ln

�Cout

Cin

��2pRL ð2Þ

where Cin and Cout are the inlet and outlet concentrations at steady state, Qin isthe perfusion rate, R and L are the intestinal radius (1.375 mm) and length(10 cm), respectively, and 2pRL represents the available area for the membranepermeability (Kararli, 1995; Liao et al., 2005).

Stability of Apicidin in Artificial Gastric and Intestinal Juices. Simulatedgastric juice was prepared by dissolving 2 mg of sodium chloride and 35%hydrochloric acid in distilled water to a total volume of 1 liter. Simulatedintestinal juice was prepared by mixing 250 ml of 0.2 mol/l potassiumphosphate and 118 ml of 0.2 mol/l sodium hydroxide in distilled water to a totalvolume of 1 liter. Portions of the simulated gastric and intestinal juices (900 mleach) were preincubated in a water bath for 5 minutes at 37°C, and then 100 mlof apicidin dissolved in dimethylsulfoxide (100 g/ml) was added to each simulatedgastric juice and intestinal juices, separately. Samples were taken at 15, 30, 60, and120 minutes and kept at 220°C until analysis.

In Vivo Pharmacokinetic Study. The rats were anesthetized by intraperito-neal injection of ketamine and xylazine (90:10 mg/kg) and cannulated with apolyethylene (PE) tubing (0.58 mm i.d., 0.96 mm o.d.; Natsume, Tokyo, Japan) inthe right jugular vein and the intended administration routes of femoral vein,duodenum, superior mesenteric artery, portal vein, or carotid artery. Apicidin wasdissolved in dimethylsulfoxide:PEG400:isotonic saline mixture (15:65:20 v/v) andinjected into the different administration sites.

The femoral vein was cannulated in rats receiving i.v. bolus injection.Apicidin was injected at a dose of 2 mg/kg (1 ml/kg) into the femoral vein (n =4 rats). An abdominal incision was also made in these rats to preserve the sameexperimental conditions as in the other groups.

For intraduodenal administration, rats were anesthetized and cannulated after12 hours of fasting. The abdomen was opened, and a PE tubing (0.58 mmi.d., 0.96 mm o.d.; Natsume, Tokyo, Japan) was inserted through the stomach,with the end of the PE tubing located at the duodenum. To prevent reflux of the

Absorption and First-Pass Metabolism of Apicidin 975

at ASPE

T Journals on July 3, 2018

dmd.aspetjournals.org

Dow

nloaded from

Page 3: Quantitative Determination of Absorption and First-Pass ...dmd.aspetjournals.org/content/dmd/42/6/974.full.pdf · Quantitative Determination of Absorption and First ... by means of

administered apicidin, the pylorus was ligated. Apicidin was then injected atdoses of 5 and 10 mg/kg (1, 2 ml/kg) via the duodenal cannula (n = 4 and 5,respectively).

For superior mesenteric arterial injection, an abdominal incision was made toexpose the superior mesenteric artery. A 29-gauge needle bent at 60° angleattached to a PE tubing (0.28 mm i.d., 0.61 mm o.d.; Natsume) was insertedinto the superior mesenteric artery. Epoxy glue (Krazy Glue, Columbus, OH)was applied to prevent bleeding. Apicidin was injected at a dose of 2 mg/kg(1 ml/kg) into the superior mesenteric artery (n = 5).

For portal venous injection, an abdominal incision was made to expose theportal vein. A PE tube (0.4 mm i.d., 0.8 mm o.d.; Natsume) was inserted intothe portal vein. Epoxy glue (Krazy Glue) was applied to prevent bleeding.Apicidin was injected at a dose of 2 mg/kg (1 ml/kg) into the portal vein (n = 5).

For the intra-arterial injection, the left carotid artery was cannulated, andapicidin was injected at a dose of 2 mg/kg (1 ml/kg, n = 6). An abdominalincision was made to preserve the same experimental conditions as in the otherexperiments.

Venous blood samples were collected before and at 5, 10, 15, and 30minutes, and 1, 1.5, 2, 3, 4, and 6 hours after injection of apicidin. All ex-periments were performed under anesthetized conditions. To maintain anesthesia,ketamine and xylazine (22.5:2.5 mg/kg) were administered via the intraperitonealroute every 1 hour after initial anesthesia, and the animal was placed on a heatingpad to maintain body temperature. The abdominal incision area was stapledto prevent loss of fluid and hypothermia. Serum samples were obtained bycentrifugation of the blood samples at 1500g for 10 minutes and were stored at220°Cuntil analysis.

Quantification of Apicidin and Its Metabolites by Liquid Chromatog-raphy Coupled with Tandem Mass Spectrometry. Apicidin and its metab-olites demethylated apicidin, hydroxylated apicidin, dihydroxylated apicidin,glucuronidated apicidin, glucuronidated monohydroxlyapicidin, and glucu-ronidated dihydroxyapicidin were assayed by a modification of a liquid chro-matography coupled with tandem mass spectrometry (LC-MS/MS) methodpreviously reported elsewhere (Shin et al., 2005). Briefly, 100 ml of the samplewas mixed with 100 ml of the internal standard solution (trazodone 100 ng/mlin methanol) and precipitated by addition of 800 ml of the mobile phase(acetonitrile/water containing 10 mM ammonium acetate = 85:15 v/v). Thesamples were mixed on a vortex mixer for 10 minutes and centrifuged at 2840gfor 10 minutes. An aliquot of the supernatant (10 ml) was then injected into theLC-MS/MS.

The HPLC analysis was performed using a Shimadzu 10Avp HPLC system(Kyoto, Japan) consisting of SCL-10Avp system controller, LC-10ADvp pump,SIL-10ADvp auto sampler, CTO-10Avp column oven, and DGU-14A degasser.Analytes were separated on a Gemini C18 column (150 � 2.0 mm i.d., 5 mm)with a SecurityGuard column (4 � 2.0 mm i.d.) (Phenomenex, Torrance, CA).The isocratic mobile phase consisted of acetonitrile and 10 mM ammoniumacetate in water (85:15 v/v). The flow rate of the mobile phase was 0.2 ml/min,and the column oven temperature was 35°C. The total run time was 4 minutes.

The HPLC system was coupled to an API 2000 triple-quadrupole massspectrometer equipped with a turbo electrospray ionization (ESI) source (ABMDS Sciex, Toronto, Ontario, Canada). The electrospray ionization source wasoperated in a positive mode. The selected multiple reaction monitoringtransitions of the precursors to the product ion were 624.5 → 84.2 for apicidin,610.5 → 84.2 for demethylated apicidin, 640.5 → 84.2 for hydroxylatedapicidin, 656.5 → 84.2 for dihydroxylated apicidin, 800.5 → 84.2 forgulcuronidated apicidin, 816.5 → 84.2 for glucuronidated monohydroxyapici-din, 832.5 → 84.2 for glucuronidated dihydroxyapicidin, and 372.1 → 176.1for trazodone.

Pharmacokinetic Analysis. The serum concentration of apicidin versustime data was analyzed by the noncompartmental method using WinNonlin(Pharsight, Cary, NC). The absolute oral bioavailability (F) was calculated bythe following equation:

F ¼ Div

Dpo×AUCpo

AUCivð3Þ

The fraction absorbed (FX) was calculated as follows:

FX ¼ Dmesentary  artery

Dpo×

AUCpo

AUCmesentary  arteryð4Þ

Fractions of the administered dose that escaped the first-pass metabolism by gutwall (FG), liver (FH), and lung (FL) were calculated as follows:

FG ¼ Dportal  vei n

Dmesentary  artery×AUCmesentary  artery

AUCportal  vei nð5Þ

FH ¼ Div

Dportal  vein×AUCportal  vei n

AUCivð6Þ

FL ¼ Dia

Div×AUCiv

AUCiað7Þ

where AUC is the area under the serum apicidin concentration versus timecurve from time zero to infinity and D is the administered dose. The subscriptrefers to the route of administration.

Statistical Analysis. The obtained parameters were compared by unpairedt test between the two means for unpaired data or one-way analysis of variance(ANOVA) followed by Scheffe’s post hoc test among more than two means forunpaired data. P , 0.05 was considered statistically significant.

Results

Quantification of Apicidin and Its Metabolites in Biologic Fluidsby LC-MS/MS. The LC-MS/MS method used to determine apicidinconcentrations was optimized and applied to analyze apicidin and itsmetabolites in various biologic fluids. The biologic samples werepretreated with one-step protein precipitation, and the assay wasvalidated over a linear concentration range from 2–1000 ng/ml.After the rats were given apicidin, apicidin and its metabolites

including demethylated apicidin and hydroxylated apicidin were detectedin serum samples. The retention times of apicidin, demethylated apicidin,hydroxylated apicidin, and dihydroxylated apicidin were 2.81, 2.63, 2.34,and 2.07 minutes, respectively. None of the potential phase II metabolites,such as glucuronidated apicidin, glucuronidated monohydroxylapicidin,or glucuronidated dihydroxyapicidin, were detected in the serumsamples.Determination of Passive Absorption Potential of Apicidin

Using IAM Chromatography. The relationship between the fractionof dose absorbed in humans (Fa%) and the IAM capacity factors(KIAM) of 28 commercial drugs is shown in Fig. 1. Based on thisrelationship, the equation derived to predict the passive absorptionpotential had excellent correlation (eq. 1, r = 0.958). The passiveabsorption potential of apicidin was then calculated from thisequation. Apicidin was highly retained on the amphiphilic phospho-lipids covalently bound to aminopropyl silica particles on IAM (tr =190.7 minutes), suggesting that it would strongly interact with the cellmembrane and demonstrate a high intestinal permeability. Based onthe retention time, the IAM capacity factor of apicidin corrected by thepower of the compound molecular weight was calculated (KIAM/MW2.05 = 32.22), and the fraction of dose absorbed in humans bypassive diffusion (Fa%) was predicted as 98.01%, indicating that thepassive diffusion-mediated permeability of apicidin would be veryhigh.Determination of Intestinal Permeability of Apicidin Using

Single-Pass Perfusion. The role of P-gp in the intestinal permeabilityof apicidin was evaluated by in situ single-pass intestinal perfusion.The effective permeability (Peff) of apicidin through the duodenumwas compared in the presence or absence of the potent P-gp inhibitorelacridar. The Peff of apicidin in the duodenum in the presence of1 mM of elacridar was statistically significantly higher than that in theabsence of elacridar (17.836 4.68 � 1025 versus 5.086 4.11 � 1025

cm/s, P , 0.05). This finding suggests that apicidin is a substrate ofP-gp and that P-gp may play an important role in the intestinalpermeability of apicidin.

976 Shin et al.

at ASPE

T Journals on July 3, 2018

dmd.aspetjournals.org

Dow

nloaded from

Page 4: Quantitative Determination of Absorption and First-Pass ...dmd.aspetjournals.org/content/dmd/42/6/974.full.pdf · Quantitative Determination of Absorption and First ... by means of

Stability of Apicidin in Simulated Gastric and Intestinal Juices.The stability of apicidin in gastiric and intestinal fluids was examinedby monitoring changes in apicidin concentration during 2 hours ofincubation in simulated gastric and intestinal juices. Apicidin wasfound to be stable (99.5% 6 3.4%) in the simulated intestinal juiceover the 2-hour incubation period. Apicidin was also stable in thesimulated gastric juice (pH = 1.2) for 1 hour, but its concentrationdecreased to 85.0% at the end of the incubation.Pharmacokinetics of Apicidin and Its Metabolites in Anesthe-

tized Rats after Intravenous Bolus Injection. The average concen-tration of apicidin, demethylated apicidin, and hydroxylated apicidinversus time profiles after a single i.v. bolus injection of apicidin at2 mg/kg in anesthetized rats is shown in Fig. 2. The serum apicidinconcentration versus time profile shows multiexponential decline.Both demethylated apicidin and hydroxylated apicidin were de-tected from the first blood sampling time (5 minutes) after i.v.administration of apicidin. Although the demethylated apicidinconcentrations decreased in parallel to the apicidin concentrations,the hydroxylated apicidin concentrations increased initially,reached a maximum concentration at approximately 0.6 hours,and decreased at a slower rate (Fig. 2).The noncompartmental pharmacokinetic parameters of apicidin,

demethylated apicidin, and hydroxylated apicidin obtained after i.v.injection of apicidin are shown in Table 1. The average terminalelimination half-life (t1/2) of demethylated apicidin (1.8 6 0.3 hours)was comparable to that of apicidin (1.9 6 0.2 hours). The serumconcentrations of demethylated apicidin extrapolated to time zero (C0)and the AUC were 8.4% and 8.6% of the corresponding values foundfor apicidin. The t1/2 of hydroxylated apicidin (2.6 6 0.4 hours) wassignificantly longer than that of apicidin, indicating that the eliminationrate of hydroxylated apicidin was slower than the metabolite formationrate.Pharmacokinetics of Apicidin and Metabolites in Anesthetized

Rats after Intraduodenal Administration. The average apicidin andmetabolite concentrations versus time profiles obtained after intra-duodenal administration of apicidin at doses of 5 and 10 mg/kg in

anesthetized rats are shown in Fig. 3. Apicidin was detected in serumfrom the first blood sampling time (5 minutes) and reached itsmaximum concentration within 1 hour in both groups of rats. Theconcentration versus time profiles of demethylated apicidin were inparallel with those of apicidin. However, the decrease in serumhydroxylated apicidin concentrations appeared to be slower than thatof apicidin; this observation was similar to what was found after i.v.injection.The noncompartmental pharmacokinetic parameters of apicidin,

demethylated apicidin, and hydroxylated apicidin obtained afterintraduodenal administration of apicidin at doses of 5 and 10 mg/kgare summarized in Table 2. The maximum serum concentration (Cmax)and the AUC of apicidin increased as the dose was increased. Theobserved Cmax values were 341.6 6 85.2 and 924.4 6 121.9 ng/mland the AUCs were 725.7 6 152.5 and 2110.5 6 397.2 ng·h/ml afterintraduodenal administration of 5 and 10 mg/kg, respectively. The t1/2of apicidin after intraduodenal administration was not significantlydifferent from that obtained after i.v. injection (Tables 1-2). Thesystemic clearances after intraduodenal administration of apicidin(CLs/F) at doses of 5 or 10 mg/kg were 119.0 6 26.6 and 81.1 6 14.4ml/min/kg, respectively. The systemic clearance corrected by oralbioavailability (CLs) was not significantly different between the 5 and10 mg/kg doses (27.4 versus 27.1 ml/min/kg), and these values were

Fig. 1. Relationship between the fraction of dose absorbed in humans (Fa%) andKIAM/MW for 28 commercial drugs (s) and apicidin (d). Solid line represents therelationship between Fa% and KIAM expressed by the sigmoid equation. Fig. 2. Average serum concentration of apicidin, demethylated apicidin, and

hydroxylated apicidin versus time curves after i.v. injection of apicidin at 2 mg/kg inanesthetized rats. Data are presented as the average 6 S.D. (n = 4).

TABLE 1.

Noncompartmental pharmacokinetic parameters of apicidin, demethylated apicidin,and hydroxylated apicidin obtained after i.v. injection of apicidin (2 mg/kg) in

anesthetized rats (mean 6 S.D.).

ParameterApicidin(n = 4)

Demethylated apicidin(n = 4)

Hydroxylated apicidin(n = 4)

t1/2 (h) 1.9 6 0.2 1.8 6 0.3 2.6 6 0.4*C0 (ng/ml) 1585.9 6 362.5 133.8 6 26.2 46.6 6 26.1AUC} (ng·h/ml) 1262.5 6 73.8 108.5 6 17.8 181.1 6 37.2CLs (ml/min/kg) 26.5 6 1.6 — —

Vss (l/kg) 3.2 6 0.4 — —

Vz (l/kg) 4.3 6 0.5 — —

MRT} (h) 2.0 6 0.2 1.7 6 0.3 4.1 6 0.6

*P , 0.05 for apicidin versus hydroxylated apicidin.

Absorption and First-Pass Metabolism of Apicidin 977

at ASPE

T Journals on July 3, 2018

dmd.aspetjournals.org

Dow

nloaded from

Page 5: Quantitative Determination of Absorption and First-Pass ...dmd.aspetjournals.org/content/dmd/42/6/974.full.pdf · Quantitative Determination of Absorption and First ... by means of

comparable to that obtained after i.v. injection (26.56 1.6 ml/min/kg).The average absolute oral bioavailability of apicidin was 23.0% 64.8% and 33.4% 6 6.3% at 5 and 10 mg/kg doses, respectively.

At both doses, the serum concentrations of demethylated apicidindecreased in parallel with those of apicidin, and the t1/2 wascomparable between apicidin and demethylated apicidin. In contrast,the t1/2 of hydroxylated apicidin at 5 and 10 mg/kg doses (2.3 6 0.7and 3.1 6 0.9 hours, respectively) was longer than those of apicidin(1.3 6 0.4 and 1.8 6 0.7 hours) and demethylated apicidin (1.4 6 0.3and 2.0 6 0.6 hours). The Tmax of hydroxylated apicidin (1.1 6 0.3and 1.1 6 0.2 hours for 5 and 10 mg/kg dose, respectively) occurredlater than those of apicidin and demethylated apicidin. The AUC ofdemethylated and hydroxylated apicidin obtained after intraduodenaladministration of 5 mg/kg dose was 96.3 6 13.6 ng/ml and 199.2 633.2 ng·h/ml, respectively, and they were 13.3% and 27.5% ofapicidin, respectively. The AUC of demethylated and hydroxylatedapicidin after intraduodenal administration of 10 mg/kg dose was294.76 68.2 ng/ml and 576.26 182.0 ng·h/ml, respectively, and theywere 14.0% and 27.3% of apicidin, respectively, which was signif-icantly greater than obtained after i.v. injection.Pharmacokinetics of Apicidin in Anesthetized Rats after Superior

Mesenteric Arterial, Portal Venous, and Carotid Arterial Injection.The mean apicidin concentration versus time profiles obtained aftersuperior mesenteric arterial, portal venous, and intra-arterial (left carotidarterial) injection of apicidin at a dose of 2 mg/kg in anesthetized rats incomparison with that after i.v. injection are shown in Fig. 4. The serumconcentrations of apicidin after different routes of administrationdeclined in parallel with that after i.v. injection. The noncompartmentalpharmacokinetic parameters of apicidin obtained after portal venous,superior mesenteric arterial, and intra-arterial injection of apicidin ata dose of 2 mg/kg are shown in Table 3.The t1/2 obtained after superior mesenteric arterial, portal venous,

and arterial injection (1.4 6 0.5, 1.9 6 0.3, 1.3 6 0.4 hours,respectively) were all comparable with that found after i.v. injection(1.9 6 0.2 hours). However, there were significant differences betweenthe AUC values after intraduodenal versus superior mesenteric arterialinjection, superior mesenteric arterial versus portal venous injection, andportal venous versus i.v. injection. The AUC of apicidin obtained aftersuperior mesenteric arterial injection (891.1 6 155.3 ng·h/ml) wassignificantly lower than that found after portal venous injection. The C0

and AUC of apicidin obtained portal venous injection was significantlylower than those obtained after i.v. injection. The pharmacokineticparameters obtained after intra-arterial injection of apicidin were notsignificantly different from those after i.v. injection.Determination of the Gastrointestinal, Hepatic, and Pulmonary

First-Pass Metabolism and Overall Bioavailability. Table 4 summa-rizes the estimation of total (FTotal), absorption (FX), gut wall (FG),hepatic (FH), and pulmonary (FL) bioavailability and the first-passelimination fraction of apicidin. The calculated fraction absorbed (FX)was 45.9% 6 9.7%. The calculated fraction not metabolized in thesingle passage through the gut wall was 70.9% 6 8.1%; that is, 29.1%

Fig. 3. Average serum concentration of apicidin, demethylated apicidin, andhydroxylated apicidin versus time curves after intraduodenal administration ofapicidin at 5 mg/kg (n = 4, panel A) and 10 mg/kg (n = 5, panel B) in anesthetizedrats. Data are presented as the average 6 S.D.

TABLE 2.

Noncompartmental pharmacokinetic parameters of apicidin, demethylated apicidin, and hydroxylated apicidin obtained after intraduodenal administration of apicidin at 5 and10 mg/kg doses in anesthetized rats (mean 6 S.D.).

Parameter

Dose = 5 mg/kg Dose = 10 mg/kg

Apicidin(n = 4)

Demethylated Apicidin(n = 4)

Hydroxylated Apicidin(n = 4)

Apicidin(n = 5)

Demethylated Apicidin(n = 5)

Hydroxylated Apicidin(n = 5)

t1/2 (h) 1.3 6 0.4 1.4 6 0.3 2.3 6 0.7 1.8 6 0.7 2.0 6 0.6 3.1 6 0.9Tmax (h) 0.9 6 0.3 0.8 6 0.3 1.1 6 0.3 0.8 6 0.3 0.9 6 0.2 1.1 6 0.2Cmax (ng/ml) 341.6 6 85.2 48.9 6 9.0 57.3 6 22.9 924.4 6 121.9 133.3 6 12.5 131.1 6 23.9AUC‘ (ng·h/ml) 725.7 6 152.5 96.3 6 13.6 199.2 6 33.2 2110.5 6 397.2 294.7 6 68.2 576.2 6 182.0CL/F (ml/min/kg) 119.0 6 26.6 — — 81.1 6 14.4 — —

Vz/F (l/kg) 13.3 6 2.9 — — 12.5 6 3.7 — —

MRT‘ (h) 2.1 6 0.6 1.9 6 0.3 3.7 6 1.0 2.7 6 0.8 2.7 6 0.7 4.9 6 1.3

978 Shin et al.

at ASPE

T Journals on July 3, 2018

dmd.aspetjournals.org

Dow

nloaded from

Page 6: Quantitative Determination of Absorption and First-Pass ...dmd.aspetjournals.org/content/dmd/42/6/974.full.pdf · Quantitative Determination of Absorption and First ... by means of

of the permeated apicidin through gut wall was metabolized by the gutwall first-pass extraction. The calculated hepatic bioavailability was70.6% 6 12.3%. Conversely, 29.4% of apicidin given into the portalvein was eliminated by the liver first-pass metabolism. There was nosignificant difference in AUC between two different dosing routes,indicating that the pulmonary first-pass metabolism was negligible(FL = 102.8% 6 7.4%).Taken together, 45.9% of the administered apicidin was absorbed

into enterocytes, and 29.1% of the absorbed apicidin was metabolizedby the gut wall first-pass extraction upon duodenal administration.Only 32.5% of the administered dose reached the portal vein, and29.4% of that was extracted by the liver first-pass metabolism whilethe rest (70.6%) escaped the hepatic metabolism. As a consequence,the overall bioavailability was calculated to be 23.0% (0.459 � 0.709 �0.706 � 100%).

Discussion

This study examined the pharmacokinetics of apicidin and quantita-tively determined the factors contributing to its oral bioavailability,including the absorption and first-pass metabolism at different stagesof the oral absorption. Administering apicidin into different sites—that is, into the femoral vein, duodenum, superior mesenteric artery,portal vein, and carotid artery—allowed the estimation of the fractionabsorbed and first-pass metabolism in the gut, liver, and lung in vivo(Fig. 5).

The gastrointestinal bioavailability (FX·FG) consists of the fractionabsorbed (FX) and the fraction that is not metabolized during passagethrough the gut wall (FG). However, few in vivo experimentalstrategies are available to distinguish the absorption and first-passmetabolism in the gut. Most studies determined the total gastrointes-tinal bioavailability (FX·FG) (Raoof et al., 1996; Hashimoto et al.,1998; Mihara et al., 2001; Choi et al., 2006; Hanada et al., 2008; Baeet al., 2009; Gertz et al., 2011) or minimized the interference of theother by using a model drug of which metabolism is negligible (Kaganet al., 2010) or specific inhibitors (Shirasaka et al., 2011). Althoughpredictions can be made by in vitro methods, a simple extrapolationfrom in vitro to in vivo has considerable limitations because variousfactors may affect the absorption and metabolism in the gastrointes-tinal tract. Separate quantification of absorption and metabolism in thegut wall was enabled in the present study by comparing the drugexposure after intraduodenal and superior mesenteric arterial admin-istration of apicidin. The mesenteric artery is responsible for bloodsupply to the gastrointestinal tract. Especially, the superior mesentericartery supplies the arterial blood to major drug absorption sites,including the duodenum and jejunum. Thus, apicidin injected into themesenteric artery bypasses the permeation process and the apicidininjected via this route is considered to be fully absorbed into the gut.This experimental design allows the separate determination of thefraction absorbed into the gut and the fraction metabolized duringpassage through the gut wall (Kwan, 1997). The fraction of drugpassing through the gut wall without being metabolized (FG), separatefrom the effect of gastrointestinal absorption, could be obtained bydividing the AUC obtained after superior mesenteric arterial injectionby the AUC after portal venous injection. The FG was calculated to be70.9% 6 8.1%; 29.1% of absorbed apicidin was thought to bemetabolized by the gut wall first-pass extraction. The calculatedfraction absorbed (FX) was 45.9% 6 9.7%.To assess the factors contributing to the fraction absorbed (FX) of

apicidin, a series of in vitro and in situ studies were conducted.Membrane permeability through the gut wall is determined by variousmechanisms, including passive diffusion and carrier-mediated uptakeor efflux. The most important factor for passive diffusion is the affinitybetween drug and membrane. The IAM chromatographic assay usesthe interaction between drug and amphiphilic phospholipids co-valently bound to aminopropyl silica particles (Pidgeon et al., 1995;Yang and Lundahl, 1995; Ong et al., 1996; Caldwell et al., 1998;Taillardat-Bertschinger et al., 2003), which represent the cell membrane.The membrane permeability can be predicted from the chromatographicaffinity as represented by the IAM capacity factor (KIAM). Themembrane permeability of apicidin predicted by IAM chromatographicassay was 98.01%, which was higher than the value obtained by the invivo study (FX = 45.9% 6 9.7%). Although IAM chromatography isa simple and potentially useful method for screening a large number of

Fig. 4. Average serum concentration of apicidin versus time curves obtained aftersuperior mesenteric arterial, portal venous, and intra-arterial injections (2 mg/kgeach) and oral administration (5 mg/kg) in anesthetized rats. *Serum concentrationsobtained after oral administration was normalized to a 2 mg/kg dose. Data arepresented as the average 6 S.D.

TABLE 3.

Noncompartmental pharmacokinetic parameters of apicidin after portal venous, superior mesenteric arterial, andintra-arterial injection of apicidin (2 mg/kg) in anesthetized rats (mean 6 S.D.).

ParametersPortal Venous Injection

(n = 5)Mesenteric Arterial Injection

(n = 5)Intra-arterial Injection

(n = 5)

t1/2 (h) 1.9 6 0.3 1.4 6 0.5 1.3 6 0.4C0 (ng/ml) 957.8 6 384.0a 799.7 6 180.6 1616.7 6 418.3AUC‘ (ng·h/ml) 891.1 6 155.3a 632.0 6 72.5b 1233.3 6 84.4CLs (ml/min/kg) 38.6 6 8.3a 53.4 6 6.8b 27.1 6 2.0Vss (l/kg) 5.5 6 1.4a 4.9 6 1.1 2.3 6 0.5MRT‘ (h) 2.4 6 0.6 1.6 6 0.4b 1.4 6 0.4

aP , 0.05 versus i.v. injection.bP , 0.05 versus portal venous injection.

Absorption and First-Pass Metabolism of Apicidin 979

at ASPE

T Journals on July 3, 2018

dmd.aspetjournals.org

Dow

nloaded from

Page 7: Quantitative Determination of Absorption and First-Pass ...dmd.aspetjournals.org/content/dmd/42/6/974.full.pdf · Quantitative Determination of Absorption and First ... by means of

compounds, the limitation of this method is that the predictedmembrane permeability is mostly governed by the lipophilicity of thedrug. Thus, the permeability prediction for small-sized hydrophilicdrugs or substrates for membrane transporters may be limited (Balimaneet al., 2000). In case of apicidin, the significantly higher permeabilityprediction based on the IAM chromatographic assay than the in vivoestimation might be due to the involvement of efflux transporters suchas P-gp.To assess the potential involvement of P-gp in the absorption of

apicidin, a single-pass perfusion was performed in the presence orabsence of the P-gp inhibitor elacridar. Elacridar, an acridone-carboxamidederivative, is the third generation P-gp inhibitor developed by Glaxolaboratories. It binds to the allosteric site of P-gp at the nanomolar range.The effective permeability (Peff) of apicidin co-perfused with elacridarwas more than 3 times higher than that without elacridar, indicating thatP-gp-mediated efflux may be responsible for the limited in vivopermeability. This result is also consistent with the literature report ofthe involvement of P-gp in the resistance mechanism of apicidin inosteosarcoma cells in vitro (Okada et al., 2006).Another factor that may contribute to the lower fraction absorbed

(FX) is the potential degradation of apicidin in the gut. When thestability was determined in simulated gastric juice (pH = 1.2) andsimulated intestinal juice (pH = 6.8), apicidin was found stable for upto 2 hours in simulated intestinal fluid and 1 hour in simulated gastricfluid. After oral administration of apicidin, the average Tmax rangedfrom 0.8–0.9 hours; therefore, it is likely that the absorption ofapicidin was complete within 2 hours after oral administration.Moreover, given the gastric emptying half-life of 6.29 6 1.01 minutesin rats (Franklin, 1977), most apicidin in solution was quickly removedfrom stomach. Therefore, instability may not be a major contributingfactor to the absorption of apicidin. Taken together, the fractionabsorbed of apicidin (FX = 45.9% 6 9.7%) may be mainly limited byP-pg, although the permeability by passive diffusion is extensive.The drug that escapes the hepatic metabolism may be subject to

extrahepatic metabolism. One of the sites responsible for potentialextrahepatic metabolism is the lung (Heinemann and Fishman, 1969).The total systemic clearance of apicidin in conscious rat was reportedto be 15.7 ml/min (Shin et al., 2006), which was significantly higherthan the liver blood flow (11.9 ml/min) (Bernareggi and Rowland,

TABLE4.

Total

(FTotal),absorptio

n(F

X),gutwall(F

G),hepatic

(FH),andpulm

onary(F

L)bioavailabilityandthefirst-pass

elim

inationfractio

nof

apicidin

administeredduodenally

toanesthetized

rats.

FTotal

FX

FG

FH

FL

Calculatio

n

Bioavailability(%

)23.0

64.8

45.9

69.7

70.9

68.1

70.6

612.3

102.86

7.4

First-passelim

inationfractio

n(%

)77.0

54.1

29.1

29.4

�0

Fig. 5. Schematic representation of the absorption processes after oraladministration.

980 Shin et al.

at ASPE

T Journals on July 3, 2018

dmd.aspetjournals.org

Dow

nloaded from

Page 8: Quantitative Determination of Absorption and First-Pass ...dmd.aspetjournals.org/content/dmd/42/6/974.full.pdf · Quantitative Determination of Absorption and First ... by means of

1991). The role of biliary and urinary excretion of apicidin seems tobe minor as well (Shin et al., 2006). Therefore, an extrahepaticelimination such as pulmonary metabolism may be suspected. In thisstudy, the pulmonary bioavailability was calculated by dividing theAUC after i.v. injection by the AUC after intra-arterial injection. Nostatistically significant difference was found between the two dosingroutes, suggesting that the contribution of the lung metabolism maybe insignificant to the overall systemic clearance of apicidin (FL =102.8% 6 7.4%).To determine the extent of oral absorption, we introduced apicidin

into the duodenum. Most drug absorption takes place predominantlywithin the upper duodenum and jejunum (Thummel et al., 1996).Intraduodenal administration allows a direct contact of apicidin withthe main absorption site, and the absorption is less affected by otherfactors such as drug transit and the presence of food in thegastrointestinal tract. Interanimal variability is also thought to be less.After intraduodenal injection of apicidin at doses of 5 and 10 mg/kg,a trend of a more than dose-proportional increase in AUC values wasobserved, and the clearance (CLs/F) obtained after oral administrationof 5 mg/kg was statistically significantly higher than that of 10 mg/kg(119.9 6 26.6 versus 81.4 6 14.4 ml/min/kg). In our pervious study,however, no saturable disposition was observed over the i.v. doserange of 0.5–4.0 mg/kg (Shin et al., 2006). The serum concentrationsobtained after oral administration of 5 or 10 mg/kg in our presentstudy were within the ranges of the observed serum concentrationsin our previous study. It is possible that the absorption processesincluding gastrointestinal absorption, intestinal, or hepatic first-passmetabolism may be saturated at an oral dose of 10 mg/kg, whereassystemic metabolism may not. At higher oral doses, P-gp-mediatedefflux could be saturated, which leads to an increase in the absorptionfraction followed by higher drug exposure with dose increase. Thus,the AUC obtained after intraduodenal administration at 5 mg/kg wasused for calculating the oral bioavailability, because no or minimalabsorption saturation may occur at this dose.In this study, the superior mesenteric arterial and portal venous

injection studies were conducted in anesthetized rats. Literatureinformation on the pharmacokinetics of apicidin following i.v. andoral administration were available only from conscious rats (Shinet al., 2006). To compare the pharmacokinetics of apicidin under thesame conditions, i.v. and oral administration studies were conducted inanesthetized rats in the present study. Thus, it was possible to examinethe effect of anesthesia on the pharmacokinetics of apicidin. Uponi.v. injection of apicidin in anesthetized rats, a significantly longerterminal elimination half-life of apicidin with no changes in thevolume of distribution (i.e., a smaller systemic clearance) was observedin this study compared with that found in the previous report after i.v.administration of apicidin to conscious rats (Shin et al., 2006). In theliterature, anesthesia with ketamine/xylazine reduced the heart rate andblood pressure of ICR (Institute of Cancer Research) mice (Furukawaet al., 1998). Organ clearance is a product of the extraction ratio and theorgan blood flow. Thus, alterations in the organ blood flow may affectthe organ clearance such that systemic clearance and elimination half-life of apicidin might be changed. Comparison of the pharmacokineticprofiles and pharmacokinetic parameters of apicidin in anesthetizedversus conscious rats are presented in the supplemental data (SupplementalFig. 1; Supplemental Table 1).

Conclusions

The gastrointestinal absorption of apicidin and the sequential first-pass metabolism in the gut, liver, and lung were quantitativelydetermined after various routes of administration in rats. Our results

demonstrate that the absorption of apicidin is limited by P-gp-mediatedefflux as well as by first-pass gut wall and hepatic metabolism despitethe high potential of passive diffusion. The present study providesa useful experimental strategy to understand the absorption processes ofa drug and to guide development of new analogs with optimized oralbioavailability.

Authorship ContributionsParticipated in research design: B.S. Shin, Yoo, Kim, Joo, S. Shin.Conducted experiments: B.S. Shin, Yoo, Kim, J.C. Shin, Choi, S. Shin.Performed data analysis: B.S. Shin, Yoo, Kim, Bulitta, Landersdorfer,

J.C. Shin, Choi, Weon, Joo, S. Shin.Wrote or contributed to the writing of the manuscript: B.S. Shin, Yoo, Kim,

Bulitta, Landersdorfer, J.C. Shin, Choi, Weon, Joo, S. Shin.

References

Ahn MY, Ahn SG, and Yoon JH (2011) Apicidin, a histone deaceylase inhibitor, induces bothapoptosis and autophagy in human oral squamous carcinoma cells. Oral Oncol 47:1032–1038.

Ahn MY, Chung HY, Choi WS, Lee BM, Yoon S, and Kim HS (2010) Anti-tumor effect ofapicidin on Ishikawa human endometrial cancer cells both in vitro and in vivo by blockinghistone deacetylase 3 and 4. Int J Oncol 36:125–131.

Ahn MY, Kang DO, Na YJ, Yoon S, Choi WS, Kang KW, Chung HY, Jung JH, Min S, and KimHS (2012) Histone deacetylase inhibitor, apicidin, inhibits human ovarian cancer cell migrationvia class II histone deacetylase 4 silencing. Cancer Lett 325:189–199.

Bae SK, Yang KH, Aryal DK, Kim YG, and Lee MG (2009) Pharmacokinetics of amitriptylineand one of its metabolites, nortriptyline, in rats: little contribution of considerable hepatic first-pass effect to low bioavailability of amitriptyline due to great intestinal first-pass effect.J Pharm Sci 98:1587–1601.

Balimane PV, Chong S, and Morrison RA (2000) Current methodologies used for evaluation ofintestinal permeability and absorption. J Pharmacol Toxicol Methods 44:301–312.

Bernareggi A and Rowland M (1991) Physiologic modeling of cyclosporin kinetics in rat andman. J Pharmacokinet Biopharm 19:21–50.

Buoncervello M, Borghi P, Romagnoli G, Spadaro F, Belardelli F, Toschi E, and Gabriele L(2012) Apicidin and docetaxel combination treatment drives CTCFL expression and HMGB1release acting as potential antitumor immune response inducers in metastatic breast cancercells. Neoplasia 14:855–867.

Caldwell GW, Masucci JA, Evangelisto M, and White R (1998) Evaluation of the immobilizedartificial membrane phosphatidylcholine. Drug discovery column for high-performance liquidchromatographic screening of drug-membrane interactions. J Chromatogr A 800:161–169.

Choi YH, Kim SG, and Lee MG (2006) Dose-independent pharmacokinetics of metformin in rats:Hepatic and gastrointestinal first-pass effects. J Pharm Sci 95:2543–2552.

Darkin-Rattray SJ, Gurnett AM, Myers RW, Dulski PM, Crumley TM, Allocco JJ, Cannova C,Meinke PT, Colletti SL, and Bednarek MA, et al. (1996) Apicidin: a novel antiprotozoal agentthat inhibits parasite histone deacetylase. Proc Natl Acad Sci USA 93:13143–13147.

Franklin RA (1977) The influence of gastric emptying on plasma concentrations of the analgesic,meptazinol. Br J Pharmacol 59:565–569.

Furukawa S, MacLennan MJ, and Keller BB (1998) Hemodynamic response to anesthesia inpregnant and nonpregnant ICR mice. Lab Anim Sci 48:357–363.

Gertz M, Houston JB, and Galetin A (2011) Physiologically based pharmacokinetic modeling ofintestinal first-pass metabolism of CYP3A substrates with high intestinal extraction. DrugMetab Dispos 39:1633–1642.

Han JW, Ahn SH, Park SH, Wang SY, Bae GU, Seo DW, Kwon HK, Hong S, Lee HY, and LeeYW, et al. (2000) Apicidin, a histone deacetylase inhibitor, inhibits proliferation of tumor cellsvia induction of p21WAF1/Cip1 and gelsolin. Cancer Res 60:6068–6074.

Hanada K, Ikemi Y, Kukita K, Mihara K, and Ogata H (2008) Stereoselective first-pass me-tabolism of verapamil in the small intestine and liver in rats. Drug Metab Dispos 36:2037–2042.

Hashimoto Y, Sasa H, Shimomura M, and Inui K (1998) Effects of intestinal and hepatic me-tabolism on the bioavailability of tacrolimus in rats. Pharm Res 15:1609–1613.

Heinemann HO and Fishman AP (1969) Nonrespiratory Functions of Mammalian Lung. PhysiolRev 49:1–47. http://physrev.physiology.org/content/49/1/1.abstract.

Kagan L, Dreifinger T, Mager DE, and Hoffman A (2010) Role of p-glycoprotein in region-specific gastrointestinal absorption of talinolol in rats. Drug Metab Dispos 38:1560–1566.

Kararli TT (1995) Comparison of the gastrointestinal anatomy, physiology, and biochemistry ofhumans and commonly used laboratory animals. Biopharm Drug Dispos 16:351–380.

Kim HJ and Bae SC (2011) Histone deacetylase inhibitors: molecular mechanisms of action andclinical trials as anti-cancer drugs. Am J Transl Res 3:166–179.

Kwan KC (1997) Oral bioavailability and first-pass effects. Drug Metab Dispos 25:1329–1336.Kwon SH, Ahn SH, Kim YK, Bae GU, Yoon JW, Hong S, Lee HY, Lee YW, Lee HW, and HanJW (2002) Apicidin, a histone deacetylase inhibitor, induces apoptosis and Fas/Fas ligandexpression in human acute promyelocytic leukemia cells. J Biol Chem 277:2073–2080.

Lai JP, Sandhu DS, Moser CD, Cazanave SC, Oseini AM, Shire AM, Shridhar V, Sanderson SO,and Roberts LR (2009) Additive effect of apicidin and doxorubicin in sulfatase 1 expressinghepatocellular carcinoma in vitro and in vivo. J Hepatol 50:1112–1121.

Liao D, Zhao J, and Gregersen H (2005) Regional surface geometry of the rat stomach based onthree-dimensional curvature analysis. Phys Med Biol 50:231–246.

Luczak MW and Jagodzinski PP (2008) Apicidin down-regulates human papillomavirus type 16E6 and E7 transcripts and proteins in SiHa cervical cancer cells. Cancer Lett 272:53–60.

Mihara K, Matsumura M, Yoshioka E, Hanada K, Nakasa H, Ohmori S, Kitada M, and Ogata H(2001) Intestinal first-pass metabolism of eperisone in the rat. Pharm Res 18:1131–1137.

Okada T, Tanaka K, Nakatani F, Sakimura R, Matsunobu T, Li X, Hanada M, Nakamura T, OdaY, Tsuneyoshi M, and Iwamoto Y (2006) Involvement of P-glycoprotein and MRP1 in

Absorption and First-Pass Metabolism of Apicidin 981

at ASPE

T Journals on July 3, 2018

dmd.aspetjournals.org

Dow

nloaded from

Page 9: Quantitative Determination of Absorption and First-Pass ...dmd.aspetjournals.org/content/dmd/42/6/974.full.pdf · Quantitative Determination of Absorption and First ... by means of

resistance to cyclic tetrapeptide subfamily of histone deacetylase inhibitors in the drug-resistantosteosarcoma and Ewing’s sarcoma cells. Int J Cancer 118:90–97.

Ong S, Liu H, and Pidgeon C (1996) Immobilized-artificial-membrane chromatography: mea-surements of membrane partition coefficient and predicting drug membrane permeability.J Chromatogr A 728:113–128.

Park JS, Lee KR, Kim JC, Lim SH, Seo JA, and Lee YW (1999) A hemorrhagic factor (Apicidin)produced by toxic Fusarium isolates from soybean seeds. Applied and Environmental Micro-biology 65:126–130.

Park SJ, Kim MJ, Kim HB, Sohn HY, Bae JH, Kang CD, and Kim SH (2009) Cotreatment withapicidin overcomes TRAIL resistance via inhibition of Bcr-Abl signaling pathway in K562leukemia cells. Exp Cell Res 315:1809–1818.

Pidgeon C, Ong S, Liu H, Qiu X, Pidgeon M, Dantzig AH, Munroe J, Hornback WJ, Kasher JS,and Glunz L, et al. (1995) IAM chromatography: an in vitro screen for predicting drugmembrane permeability. J Med Chem 38:590–594.

Raoof AA, Augustijns PF, and Verbeeck RK (1996) In vivo assessment of intestinal, hepatic, andpulmonary first pass metabolism of propofol in the rat. Pharm Res 13:891–895.

Shin BS, Chang HS, Park EH, Yoon CH, Kim HY, Kim J, Ryu JK, Zee OP, Lee KC, and Cao D,et al. (2006) Pharmacokinetics of a novel histone deacetylase inhibitor, apicidin, in rats. Bio-pharm Drug Dispos 27:69–75.

Shin BS, Kim J, Yoon CH, Kim CH, Park EH, Han JW, and Yoo SD (2005) Development ofa liquid chromatography/electrospray tandem mass spectrometry assay for the quantification ofapicidin, a novel histone deacetylase inhibitor, in rat serum: application to a pharmacokineticstudy. Rapid Commun Mass Spectrom 19:408–414.

Shirasaka Y, Suzuki K, Shichiri M, Nakanishi T, and Tamai I (2011) Intestinal absorption ofHMG-CoA reductase inhibitor pitavastatin mediated by organic anion transporting polypeptideand P-glycoprotein/multidrug resistance 1. Drug Metab Pharmacokinet 26:171–179.

Sugano K, Hamada H, Machida M, Ushio H, Saitoh K, and Terada K (2001) Optimized conditions of bio-mimetic artificial membrane permeation assay. International Journal of Pharmaceutics 228:181–188.

Taillardat-Bertschinger A, Carrupt P-A, Barbato F, and Testa B (2003) Immobilized artificialmembrane HPLC in drug research. J Med Chem 46:655–665.

Tan J, Cang S, Ma Y, Petrillo RL, and Liu D (2010) Novel histone deacetylase inhibitors inclinical trials as anti-cancer agents. J Hematol Oncol 3:5.

Thummel KE, O’Shea D, Paine MF, Shen DD, Kunze KL, Perkins JD, and Wilkinson GR (1996)Oral first-pass elimination of midazolam involves both gastrointestinal and hepatic CYP3A-mediated metabolism [ast]. Clin Pharmacol Ther 59:491–502.

Worland PJ, Drummer OH, and Jarrott B (1984) Gastric and intestinal absorption of captopril inacutely and chronically treated rats: comparison with salicylic acid. Journal of PharmaceuticalSciences 73:1755–1758.

Yang Q and Lundahl P (1995) Immobilized proteoliposome affinity chromatography for quan-titative analysis of specific interactions between solutes and membrane proteins. Interactionof cytochalasin B and D-glucose with the glucose transporter Glut1. Biochemistry 34:7289–7294.

Yoon CH, Shin BS, Chang HS, Kwon LS, Kim HY, Yoo SE, and Yoo SD (2004) Rapidscreening of drug absorption potential using the immobilized artificial membrane phosphati-dylcholine column and molar volume. Chromatographia 60:399–404. DOI: 10.1365/s10337-004-0410-x.

Zhu C, Jiang L, Chen TM, and Hwang KK (2002) A comparative study of artificial membranepermeability assay for high throughput profiling of drug absorption potential. EuropeanJournal of Medicinal Chemistry 37:399–407.

Address correspondence to: Dr. Soyoung Shin, Department of Pharmacy, Collegeof Pharmacy, Wonkwang University, 460 Iksan-daero, Iksan City, Jeonbuk 570-749,South Korea. E-mail: [email protected]

982 Shin et al.

at ASPE

T Journals on July 3, 2018

dmd.aspetjournals.org

Dow

nloaded from