prediction of renal transporter mediated drug-drug...

10
1521-009X/43/3/325334$25.00 http://dx.doi.org/10.1124/dmd.114.059618 DRUG METABOLISM AND DISPOSITION Drug Metab Dispos 43:325334, March 2015 Copyright ª 2015 by The American Society for Pharmacology and Experimental Therapeutics Prediction of Renal Transporter Mediated Drug-Drug Interactions for Pemetrexed Using Physiologically Based Pharmacokinetic Modeling Maria M. Posada, James A. Bacon, Karen B. Schneck, Rommel G. Tirona, Richard B. Kim, J. William Higgins, Y. Anne Pak, Stephen D. Hall, and Kathleen M. Hillgren Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.M.P., J.A.B., K.B.S., J.W.H., Y.A.P., S.D.H., K.M.H.); and Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.G.T., R.B.K.) Received July 21, 2014; accepted December 12, 2014 ABSTRACT Pemetrexed, an anionic anticancer drug with a narrow therapeutic index, is eliminated mainly by active renal tubular secretion. The in vitro to in vivo extrapolation approach used in this work was developed to predict possible drug-drug interactions (DDIs) that may occur after coadministration of pemetrexed and nonsteroidal anti-inflammatory drugs (NSAIDs), and it included in vitro assays, risk assessment models, and physiologically based pharmacoki- netic (PBPK) models. The pemetrexed transport and its inhibition parameters by several NSAIDs were quantified using HEK-PEAK cells expressing organic anion transporter (OAT) 3 or OAT4. The NSAIDs were ranked according to their DDI index, calculated as the ratio of their maximum unbound concentration in plasma over the concentration inhibiting 50% (IC 50 ) of active pemetrexed transport. A PBPK model for ibuprofen, the NSAID with the highest DDI index, was built incorporating active renal secretion in Simcyp Simulator. The bottom-up model for pemetrexed underpredicted the clear- ance by 2-fold. The model we built using a scaling factor of 5.3 for the maximal uptake rate (V max ) of OAT3, which estimated using plasma concentration profiles from patients given a 10-minute infusion of 500 mg/m 2 of pemetrexed supplemented with folic acid and vitamin B 12 , recovered the clinical data adequately. The observed/predicted increases in C max and the area under the plasma-concentration time curve (AUC 0inf ) of pemetrexed when ibuprofen was coadministered were 1.1 and 1.0, respectively. The coadministration of all other NSAIDs was predicted to have no significant impact on the AUC 0inf based on their DDI indexes. The PBPK model reasonably reproduced pemetrexed concentration time profiles in cancer patients and its interaction with ibuprofen. Introduction Pemetrexed supplemented with folic acid and vitamin B 12 is used to treat nonsmall cell lung cancer and malignant mesothelioma, alone or in combination with cisplatin (Shih et al., 1997; Bajetta et al., 2003; Scagliotti et al., 2003; Latz et al., 2006; Takimoto et al., 2007). Pemetrexed is a hydrophilic anionic compound eliminated unchanged in the urine (7090%) primarily by active tubular secretion, presumably by organic anion transporters (OATs) (Rinaldi, 1999; Kurata et al., 2014). The structurally related drug methotrexate, one of the first antifolates used for the treatment of cancer, is predominantly eliminated unchanged in urine, with 80% of the dose being secreted actively by the kidney. The administration of inhibitors of the OATs, such as probenecid and nonsteroidal anti-inflammatory drugs (NSAIDs), reduces the clearance of methotrexate. For example, Tracy et al. showed that naproxen, trisalicylate, and ibuprofen decreased the plasma clearance by 1.28-, 1.31-, and 1.66-fold, respectively (Cassano, 1989; Frenia and Long, 1992; Tracy et al., 1992). Additionally, the coadministration of high doses of methotrexate and ketoprofen was shown cause severe toxicities and even deaths (Thyss et al., 1986). Consequently the use of NSAIDs with high doses of methotrexate is not recommended. Because patients undergoing chemotherapy often take nonsteroidal anti-inflammatory drugs (NSAIDS) for pain control and given that methotrexate and pemetrexed rely on renal transporters for their elimination, it was hypothesized that pemetrexed would experience the same drug interaction as methotrexate. Clinical studies demonstrated no interaction with aspirin, but a 20% increase in the area under the plasma-concentration time curve (AUC 0inf ) of pemetrexed was observed when patients with normal renal function (creatinine clearance .80 ml/min) were cotreated with 400 mg of ibuprofen 4 times a day (Sweeney et al., 2006). Although it is statistically significant, this increase does not require a dose adjustment in patients with normal renal function (creatinine clearance .80 ml/min). However, because renal insufficiency increases pemetrexed exposure, caution should be used when administering NSAIDs concurrently with pemetrexed to patients with mild and moderate renal insufficiency (creatinine clearance from 45 to 79 ml/min) (Mita et al., 2006). Although not studied clinically, it is recommended that NSAIDs with short elimination half-life should be avoided for the period of 2 days before, the day of, and 2 days after administration of pemetrexed in patients with mild to moderate renal insufficiency. For NSAIDs with longer elimination half-lives, patient taking these NSAIDs should interrupt dosing for at least 5 days before, the day of, and 2 days after pemetrexed administration [Alimta (pemetrexed), Eli Lilly and Company, Indianapolis, IN; package insert available at http://labels.fda.gov/]. dx.doi.org/10.1124/dmd.114.059618. ABBREVIATIONS: AUC, area under the plasma-concentration time curve; DDI, drug-drug interaction; E3S, [ 3 H]estrone-3-sulfate; LC-MS/MS, liquid chromatography with tandem mass spectrometry; NSAID, nonsteroidal anti-inflammatory drug; OAT, organic anion transporter; PBPK, physiologically based pharmacokinetic; REF/RAF, relative expression factor/ relative activity factor. 325 at ASPET Journals on September 26, 2020 dmd.aspetjournals.org Downloaded from

Upload: others

Post on 25-Jul-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Prediction of Renal Transporter Mediated Drug-Drug ...dmd.aspetjournals.org/content/dmd/43/3/325.full.pdf · cells expressing organic anion transporter (OAT) 3 or OAT4. The NSAIDs

1521-009X/43/3/325–334$25.00 http://dx.doi.org/10.1124/dmd.114.059618DRUG METABOLISM AND DISPOSITION Drug Metab Dispos 43:325–334, March 2015Copyright ª 2015 by The American Society for Pharmacology and Experimental Therapeutics

Prediction of Renal Transporter Mediated Drug-Drug Interactions forPemetrexed Using Physiologically Based Pharmacokinetic Modeling

Maria M. Posada, James A. Bacon, Karen B. Schneck, Rommel G. Tirona, Richard B. Kim,J. William Higgins, Y. Anne Pak, Stephen D. Hall, and Kathleen M. Hillgren

Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (M.M.P., J.A.B., K.B.S., J.W.H., Y.A.P., S.D.H., K.M.H.); andDivision of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University,

London, Ontario, Canada (R.G.T., R.B.K.)

Received July 21, 2014; accepted December 12, 2014

ABSTRACT

Pemetrexed, an anionic anticancer drug with a narrow therapeuticindex, is eliminated mainly by active renal tubular secretion. The invitro to in vivo extrapolation approach used in this work wasdeveloped to predict possible drug-drug interactions (DDIs) thatmay occur after coadministration of pemetrexed and nonsteroidalanti-inflammatory drugs (NSAIDs), and it included in vitro assays,risk assessment models, and physiologically based pharmacoki-netic (PBPK) models. The pemetrexed transport and its inhibitionparameters by several NSAIDs were quantified using HEK-PEAKcells expressing organic anion transporter (OAT) 3 or OAT4. TheNSAIDs were ranked according to their DDI index, calculated as theratio of their maximum unbound concentration in plasma over theconcentration inhibiting 50% (IC50) of active pemetrexed transport.A PBPK model for ibuprofen, the NSAID with the highest DDI index,

was built incorporating active renal secretion in Simcyp Simulator.The bottom-up model for pemetrexed underpredicted the clear-ance by 2-fold. The model we built using a scaling factor of 5.3 forthe maximal uptake rate (Vmax) of OAT3, which estimated usingplasma concentration profiles from patients given a 10-minuteinfusion of 500 mg/m2 of pemetrexed supplemented with folic acidand vitamin B12, recovered the clinical data adequately. Theobserved/predicted increases in Cmax and the area under theplasma-concentration time curve (AUC0–inf) of pemetrexed whenibuprofen was coadministered were 1.1 and 1.0, respectively. Thecoadministration of all other NSAIDs was predicted to have nosignificant impact on the AUC0–inf based on their DDI indexes. ThePBPK model reasonably reproduced pemetrexed concentrationtime profiles in cancer patients and its interaction with ibuprofen.

Introduction

Pemetrexed supplemented with folic acid and vitamin B12 is used totreat non–small cell lung cancer and malignant mesothelioma, alone orin combination with cisplatin (Shih et al., 1997; Bajetta et al., 2003;Scagliotti et al., 2003; Latz et al., 2006; Takimoto et al., 2007).Pemetrexed is a hydrophilic anionic compound eliminated unchanged inthe urine (70–90%) primarily by active tubular secretion, presumably byorganic anion transporters (OATs) (Rinaldi, 1999; Kurata et al., 2014).The structurally related drug methotrexate, one of the first antifolatesused for the treatment of cancer, is predominantly eliminated unchangedin urine, with 80% of the dose being secreted actively by the kidney.The administration of inhibitors of the OATs, such as probenecid andnonsteroidal anti-inflammatory drugs (NSAIDs), reduces the clearanceof methotrexate. For example, Tracy et al. showed that naproxen,trisalicylate, and ibuprofen decreased the plasma clearance by 1.28-,1.31-, and 1.66-fold, respectively (Cassano, 1989; Frenia and Long,1992; Tracy et al., 1992). Additionally, the coadministration of highdoses of methotrexate and ketoprofen was shown cause severe toxicitiesand even deaths (Thyss et al., 1986). Consequently the use of NSAIDswith high doses of methotrexate is not recommended.

Because patients undergoing chemotherapy often take nonsteroidalanti-inflammatory drugs (NSAIDS) for pain control and given thatmethotrexate and pemetrexed rely on renal transporters for theirelimination, it was hypothesized that pemetrexed would experience thesame drug interaction as methotrexate. Clinical studies demonstrated nointeraction with aspirin, but a 20% increase in the area under theplasma-concentration time curve (AUC0–inf) of pemetrexed wasobserved when patients with normal renal function (creatinine clearance.80 ml/min) were cotreated with 400 mg of ibuprofen 4 times a day(Sweeney et al., 2006). Although it is statistically significant, thisincrease does not require a dose adjustment in patients with normal renalfunction (creatinine clearance .80 ml/min). However, because renalinsufficiency increases pemetrexed exposure, caution should be usedwhen administering NSAIDs concurrently with pemetrexed to patientswith mild and moderate renal insufficiency (creatinine clearance from45 to 79 ml/min) (Mita et al., 2006). Although not studied clinically, it isrecommended that NSAIDs with short elimination half-life should beavoided for the period of 2 days before, the day of, and 2 days afteradministration of pemetrexed in patients with mild to moderate renalinsufficiency. For NSAIDs with longer elimination half-lives, patienttaking these NSAIDs should interrupt dosing for at least 5 days before,the day of, and 2 days after pemetrexed administration [Alimta(pemetrexed), Eli Lilly and Company, Indianapolis, IN; package insertavailable at http://labels.fda.gov/].dx.doi.org/10.1124/dmd.114.059618.

ABBREVIATIONS: AUC, area under the plasma-concentration time curve; DDI, drug-drug interaction; E3S, [3H]estrone-3-sulfate; LC-MS/MS,liquid chromatography with tandem mass spectrometry; NSAID, nonsteroidal anti-inflammatory drug; OAT, organic anion transporter; PBPK,physiologically based pharmacokinetic; REF/RAF, relative expression factor/ relative activity factor.

325

at ASPE

T Journals on Septem

ber 26, 2020dm

d.aspetjournals.orgD

ownloaded from

Page 2: Prediction of Renal Transporter Mediated Drug-Drug ...dmd.aspetjournals.org/content/dmd/43/3/325.full.pdf · cells expressing organic anion transporter (OAT) 3 or OAT4. The NSAIDs

OATs are a group of membrane transporter proteins expressed inseveral epithelial tissues of the body, including the proximal tubularcells of the kidney (Erdman et al., 2006; Burckhardt, 2012). Thesetransporters play an important role in renal elimination, by activelysecreting a wide variety of endogenous and exogenous hydrophilicorganic anions, including p-aminohippuric acid, prostaglandins, ceph-alosporins, antivirals, and anticancer drugs (Burckhardt, 2012). In thekidney, OAT1 (SLC22A6) and OAT3 (SLC22A8) are expressed on thebasolateral membrane of the proximal tubular cells, where they importorganic anions in an exchange of dicarboxylates such as succinate anda-ketoglutarate (Motohashi et al., 2002; Bakhiya et al., 2003; Sweetet al., 2003). OAT4 (SLC22A11) is expressed on the apical membraneof the apical tubular cells and functions as a bidirectional transporter,moving solutes from the cell into the tubular fluid and vice versa in anexchange of dicarboxylates, urate, and other organic anions (Motohashiet al., 2002; Hagos et al., 2007a; Hagos et al., 2007b). NSAIDs area well characterized group of OAT inhibitors that are generally betterinhibitors of OAT1 and OAT3 than OAT4. Although ibuprofen anddiclofenac are effective inhibitors of OAT1 and OAT3, with con-centration inhibiting 50% of the active transport (IC50) values in thelower micromolar range, acetaminophen, aspirin, and salicylate are low-affinity inhibitors (Khamdang et al., 2002).

Our objectives were to 1) identify the transporters responsible for thecellular transport of pemetrexed, 2) determine the kinetic parameters ofpemetrexed transport, 3) quantify the inhibitory potency of a subset ofNSAIDs toward these transporters, and finally 4) establish a mechanisticphysiologically based pharmacokinetic (PBPK) model of pemetrexedand its interaction with ibuprofen that also could be applied to otherpotential inhibitors of pemetrexed renal secretion.

Materials and Methods

Chemicals. Pemetrexed was synthesized at Eli Lilly and Company.Radiolabeled [14C]ibuprofen was purchased from American Radiochemicals(St. Louis, MO). Radiolabeled [3H]estrone-3-sulfate (E3S) was purchased fromPerkin Elmer (Boston, MA). The internal standard ([13C5

15N]pemetrexed) andcelecoxib were synthesized at Eli Lilly and Company. All other chemicals werepurchased from Sigma-Aldrich (St. Louis, MO).

Preliminary In Vitro Substrate Transporter Screen. The preliminaryuptake screen was performed according to a transient transporter expressionmethod previously described elsewhere using HeLa cells transfected with cDNAsof human transporters OAT1 (SLC22A6), OAT2 (SLC22A7), OAT3(SCL22A8), OAT4 (SLC22A9), OCT1 (SLC22A1), OCT2 (SLC22A2), OCTN1(SLC22A4), OATP1A2 (SLCO1A2), OATP1B1 (SLCO1B1), OATP1B3(SLCO1B3), OATP2B1 (SLCO2B1), OATP4C1 (SLCO4C1), ASBT

TABLE 1

Physicochemical and biochemical parameters used in the pemetrexed PBPK model

Property Value Source

Compound name PemetrexedCompound type Diprotic acid Shih et al., 1997Molecular weight 427.4 Shih et al., 1997Molecular formula C20H21N5O6 Shih et al., 1997Fraction unbound plasma (fu) 0.19 MeasuredBlood to plasma ratio (B:P) 0.55 PredictedcLogP 20.797 Predicted chemaxonpKa 1 (acid) 3.23 MeasuredpKa 2 (acid) 4.4 MeasuredDose (mg/m2) 500Route of administration Infusion i.v.Infusion time (min) 10CLint passive (ml/min per 106 cells) 8.0 1026 MeasuredKm OAT3 (mM) 18.2 MeasuredVmax OAT3 (pmol/min per 106 cells) 185 MeasuredIntrinsic uptake clearance (CLint,T) OAT4 (ml/min per 106 cells) 0.99 MeasuredREF OAT3 5.3 Fitted

TABLE 2

Ibuprofen physicochemical and biochemical parameters used for the PBPK model

Property Value Source

Compound name IbuprofenCompound type Monoprotic acid Davies, 1998Molecular weight 206.3 Davies, 1998Molecular formula C13H18O2 Davies, 1998Fraction unbound in plasma (fu) 0.012 Lockwood et al., 1983bLogP 4.13 Avdeef et al., 1999Blood to plasma ratio (B:P) 0.55 Default in SimcyppKa (acid) 4.42 Avdeef et al., 1999; Avdeef et al., 2000Clearance/F (l/h) 3.84 Davies, 1998Papp (cm/sec) 52.2 1026 Yee, 1997Human effective jejunal permeability (Peff) (cm/sec) 5.69 1024 Predicted based on Papp; Yee, 1997Fraction unbound in gut (fugut) 1 Default in SimcypIntrinsic solubility (mg/ml) 0.049 Avdeef et al., 1999Ki OAT3 (mM) 2.1 MeasuredKi OAT4 (mM) 18.6 Measured valueFraction unbound incubation (fu,inc) 0.89 Measured

326 Posada et al.

at ASPE

T Journals on Septem

ber 26, 2020dm

d.aspetjournals.orgD

ownloaded from

Page 3: Prediction of Renal Transporter Mediated Drug-Drug ...dmd.aspetjournals.org/content/dmd/43/3/325.full.pdf · cells expressing organic anion transporter (OAT) 3 or OAT4. The NSAIDs

(SLC10A2), PEPT1 (SLC15A1), and PEPT2 (SLC15A2) (Cvetkovic et al.,1999). Drug uptake was evaluated using [14C]pemetrexed (60 mM) in medium for10 minutes at 37�C. The reaction was terminated using ice-cold buffer, and thecells were lysed with 1% SDS. The samples were added to a vial containingUltima Gold scintillation cocktail (Perkin Elmer), and the amount of pemetrexedin the cells was measured using a scintillation counter (Tri-Carb 2900TR; PerkinElmer).

In Vitro Transporter Kinetic Studies. HEK-PEAK cells were transfectedwith plasmids containing human OAT3 and OAT4 following previously publishedprotocols (Godinot et al., 2003). In short, human OAT3 (NM_004254) and OAT4(NM_080866) genes, purchased from GeneArt (Regensburg, Germany), wereinserted into EW1969 plasmid before transfection of HEK-PEAK cells using anEffectene Transfection Reagent kit (Qiagen, Valencia, CA).

In vitro uptake experiments were performed based on reports published byNozaki et al. (2004) with some modifications. HEK-PEAK cells from passages36–75 were used 72 hours after seeding on CellBIND 12-well plates (CorningLife Sciences, Tewksbury, MA) at a density of 150,000 cells/well. Thereactions were performed in uptake buffer consisting of Krebs‑Henseleit bufferat pH 7.4 supplemented with 2.5 mM CaCl2 and 10 mM HEPES. Proteinquantification was performed using a BCA protein assay (Sigma-Aldrich)where bovine serum albumin was used as the standard.

OAT3 and OAT4 qualification studies were performed to assess thefunctionality of the HEK-PEAK transfected cells. The cells were treated with0.5 ml of buffer containing 1 mM E3S for 2 minutes in the presence or absence of200 mM probenecid, a known inhibitor of the OATs, at room temperature. Theincubations were stopped by the addition of ice-cold buffer, followed by twowashes with buffer. Cells were lysed in 0.3 ml of 0.2 N NaOH overnight at 4�C.A 0.2-ml aliquot was added to a vial containing scintillation cocktail (ScintSafe30%; Thermo Fisher Scientific, Waltham, MA), and the cellular uptake of E3Swas measured using a scintillation counter. Protein quantification was performedas mentioned previously with the remaining sample.

All pemetrexed uptake and inhibition studies were performed at roomtemperature with 0.5 ml of pemetrexed in Krebs‑Henseleit buffer. Before thebeginning of the experiment, the vector control, OAT3 cells, and OAT4 cellswere washed twice with buffer and allowed to equilibrate for 5 minutes. Time-dependent uptake studies were performed with 39 mM pemetrexed and stoppedat the specified times up to 15 minutes. Concentration-dependent uptake studieswere performed for 5 minutes with pemetrexed concentrations ranging from0.01 to 117 mM. All reactions were stopped with the addition of cold buffer,and the cells were lysed in a 1:1 methanol/water (v/v) for 3 minutes. Theconcentration of pemetrexed was quantified using liquid chromatography withtandem mass spectrometry (LC-MS/MS).

In Vitro Transporter Inhibition Studies. The inhibitory effects ofindividual NSAIDs and probenecid on pemetrexed uptake were evaluatedusing vector control and OAT3 and OAT4 transfected cells incubated at roomtemperature with 0.5 ml of pemetrexed (19.5 mM) in uptake buffer. Ibuprofen,diclofenac, naproxen, and celecoxib were added to the cells in concentrationsranging from 0.1 to 200 mM. The incubations were stopped at 5 minutes; thecells were rinsed twice with ice-cold buffer, and they were treated with 0.3 mlof 1:1 methanol/water (volume/volume) to extract the pemetrexed for LC-MS/MS quantification.

Fraction Unbound in Incubation. The fraction unbound of ibuprofen in theincubations (fu,inc) was measured in vector control cells using [14C]ibuprofen.The conditions for this experiment were the same as those described earlier forthe transport experiments. Briefly, the HEK-PEAK vector control cells werewashed twice with buffer and incubated at room temperature for 5 minutes with0.5 ml of 10 mM ibuprofen in buffer. Samples of the incubation media (200 ml)were collected at 0 and 5 minutes, placed in a vial containing 10 ml ofscintillation cocktail, and quantified using a scintillation counter. The fractionof pemetrexed unbound in the incubation was assumed to be 1.

Analytic Method. The concentration of pemetrexed in the cells was quantifiedby LC-MS/MS using [13C5

15N]pemetrexed as an internal standard. Reverse-phasechromatography was used with a flow rate of 0.6 ml/min and an injection volumeof 20 ml. The analytes were chromatographically separated using a Synergi MAX-RP 80A 5m 100 � 2.0 mm high-pressure liquid chromatography column(Phenomenex, Torrance, CA), with a gradient liquid chromatography systemcomposed of 0.2% formic acid in water (mobile phase A) and 0.2% formic acid inmethanol (mobile phase B). The gradient profile was 10% B from 0 to 0.30

minutes, 70% B at 0.70 to 2.00 minutes, and returned to 10% B at 2.20 minutes.The flow rate was 0.6 ml/min, and the analysis was performed at ambientlaboratory conditions with the effluent directed to the mass spectrometer between0.10 and 2.40 minutes.

Pemetrexed was detected in positive ion mode using a Sciex API 4000 triplequadruple mass spectrometer equipped with a Turbo Ion Spray interface andAnalyst 1.4 software (Applied Biosystems/MDS, Foster City, CA). The mass/charge ratio for pemetrexed and [13C5

15N]pemetrexed was 426 → 297 m/z and432 → 297 m/z, respectively. The collision energies were -30 and -36 forpemetrexed and [13C5

15N]pemetrexed, respectively. The dynamic range of theassay was between 0.77 and 395 ng/ml, and the samples with pemetrexedconcentrations above the upper limit of quantification were diluted with bufferto concentrations within the assay range.

Three individual batches of quality control samples at four separate con-centrations (0.98, 3.9, 125, and 500 ng/ml) were examined with four to sixsamples tested at each concentration. The results for 0.98, 3.9, 125, and 500 ng/mlsamples showed an interbatch accuracy of 4.3, 3.3, 4.7, and 7.7%, respectively,and an interbatch precision of 11.1, 8.3, 7.9, and 8.4%, respectively.

Data Analysis. The Michaelis-Menten kinetic parameters for pemetrexedtransport were estimated by fitting the pemetrexed uptake data using nonlinearregression (WinNonlin v5.3; Pharsight, Sunnyvale, CA), according to thefollowing formula:

v ¼ Vmax � S

Km þ Sþ Kd � S ð1Þ

where, v is the initial uptake (pmol/min per mg of protein), Vmax is the maximaluptake rate (pmol/min per mg of protein), S is the concentration of substrate(mM), Km (mM) is the Michaelis-Menten kinetic constant, and Kd (ml/min per

TABLE 3

Characteristics of the population used in the simulation

Property Mean (S.D.) Range

Age (yr) 60.8 (12) 35–80Body Surface area (m2) 1.89 (0.222) 1.4–2.2Weight (kg) 76.2 (15.8) 49–100.2Creatinine clearance (ml/min)a 115 (31.6) 66.1–192.5% Female 37 NA

NA, not available.aEstimated from serum creatinine concentration using the Cockcroft-Gault equation (Cockcroft

and Gault, 1976).

Fig. 1. Results of the in vitro screening assay. Expression of drug uptake transporterswas performed using a recombinant vaccinia system in HeLa cells. Uptake of[14C]pemetrexed, 60 mM (n = 3–6), at the 10-minute time point. The relative uptakewas compared with HeLa cells transfected with a vector lacking transporter cDNA(vector control). Data are presented as mean 6 S.D. (n = 3). ***P , 0.001.

Predicting Renal OAT-Mediated DDIs for Pemetrexed 327

at ASPE

T Journals on Septem

ber 26, 2020dm

d.aspetjournals.orgD

ownloaded from

Page 4: Prediction of Renal Transporter Mediated Drug-Drug ...dmd.aspetjournals.org/content/dmd/43/3/325.full.pdf · cells expressing organic anion transporter (OAT) 3 or OAT4. The NSAIDs

mg of protein) is the passive permeability constant. The Kd for pemetrexed wascalculated experimentally by fitting the uptake velocity of pemetrexed in theHEK-PEAK vector control cells. The passive intrinsic clearance (CLint,passive)of pemetrexed used in the model was calculated by multiplying the Kd by thenumber of cells in 1 milligram of protein.

The inhibition constant (Ki) of the NSAIDs was calculated according to thefollowing formula, assuming competitive inhibition:

Ki ¼ IC50

1þ ½S�Km

ð2Þ

where IC50 is the concentration of NSAIDs necessary to inhibit 50% of theuptake of pemetrexed, [S] is the concentration of pemetrexed in the medium,and Km is the affinity of pemetrexed for the particular organic anion transporter.The IC50 for pemetrexed was calculated with the following formula:

Percent of control ¼ 100*1

1þ ½I�IC50

ð3Þ

where [I] is the concentration of the NSAID, and percent of control is thepercentage of pemetrexed taken up by the cells in the presence of the inhibitorcompared with the cells treated with only pemetrexed.

Statistical Analysis. The in vitro experimental data are presented as mean6S.E. or mean (% coefficient of variation). To test for significant differencesbetween multiple groups, a one-way analysis of variance was used followed byeither Dunnett’s or Bonferroni’s test. P # 0.05 was considered statisticallysignificant. The software used was GraphPad Prism 6 (GraphPad Software, LaJolla, CA).

Modeling Strategy. The modeling strategy employed in this project was thefollowing. 1) The active secretion of pemetrexed was defined to be via OAT3

and OAT4 on the basolateral and apical membrane of the proximal tubularcells, respectively. 2) A pemetrexed PBPK model was developed using in vitroand in silico data. 3) The model was optimized by using pemetrexed plasmaconcentration-time profiles from cancer patients. 4) The model was tested bychanging the pemetrexed dose from 50 to 700 mg/m2, and the results werecompared with other clinical studies. 5) A PBPK model for ibuprofen wasdeveloped using literature data and measured Ki and fu,inc. The model wasverified using the results of the clinical interaction studies (Sweeney et al.,2006). This modeling strategy is described in detail herein.

Pemetrexed Clinical Data. Human plasma concentration-time profiles afterthe administration of a 10-minute infusion of 500 mg/m2 of pemetrexed withvitamin B12 and folic acid from a published study (Sweeney et al., 2006) wereused to build the model. In this study, serial blood samples were obtainedpredose, immediately before the end of infusion, and at 0.25, 0.5, 1, 2, 4, 6, 8,12, 24, and 48 hours after termination of the infusion. Results from publishedstudies were used to test and verify the model (Latz et al., 2006; Mita et al.,2006; Nakagawa et al., 2006; Sweeney et al., 2006).

Pemetrexed PBPK Model. All simulations were performed using PKPDProfiles in the Simcyp Simulator (version 12.1; Simcyp Limited, Sheffield,United Kingdom). A full PBPK model for pemetrexed was built using measuredand predicted physicochemical and biologic data (Table 1) based on the Rodgerset al. method with rapid equilibrium between blood and tissues except for thekidney, which was set up as a permeability-limited tissue (Rodgers et al., 2005;Rodgers and Rowland, 2006). The multicompartment kidney model built intoSimcyp was used to incorporate the glomerular filtration and active renalsecretion of pemetrexed. This mechanistic kidney model consists of three majorcompartments: blood, renal cells, and tubular fluid, all subdivided into severalregions to approximate the anatomy and physiology of the kidney.

Two active transport processes were enabled in the mechanistic kidneymodel, one on the basolateral membrane of the proximal tubular cells to

Fig. 2. In vitro uptake studies. (A) The uptake of [3H]estrone-3-sulfate (E3S) with or without probenecid, into vector control (VC), OAT3, and OAT4-transfected HEK-PEAKcells. The cell cultures were treated with 1 mME3S, alone (u), or in combination with 200 mM probenecid (j) for 2 minutes at room temperature. Data are presented as mean6 S.D. (n = 3). (B) Time-dependent uptake of pemetrexed (39 mM) in VC (m), OAT3 (j), and OAT4 (d) transfected HEK-PEAK cells. Experiments were performed at roomtemperature. Data are presented as mean6 S.D. (n = 3). (C) Uptake of pemetrexed into OAT3-transfected HEK-PEAK cells treated with increasing concentrations of pemetrexedfor 5 minutes. (j) Mean6 S.D. of the uptake in HEK OAT3; (m) mean6 S.D. of the uptake in vector control cells. (D) Uptake of pemetrexed into OAT4-transfected HEK-PEAKcells treated with increasing concentrations of pemetrexed for 5 minutes; (j) mean 6 S.D. of the uptake in HEK OAT4; (m) mean 6 S.D. of the uptake in vector control cells.

328 Posada et al.

at ASPE

T Journals on Septem

ber 26, 2020dm

d.aspetjournals.orgD

ownloaded from

Page 5: Prediction of Renal Transporter Mediated Drug-Drug ...dmd.aspetjournals.org/content/dmd/43/3/325.full.pdf · cells expressing organic anion transporter (OAT) 3 or OAT4. The NSAIDs

simulate OAT3-mediated uptake of pemetrexed, and a second on the apicalmembrane of the proximal tubular cells to simulate OAT4-mediated efflux ofpemetrexed into the renal tubules. The values for the passive and activemembrane transport of pemetrexed were assumed to be the equal in all threecompartments of the proximal tubular cells and were taken from the in vitroexperiments previously described.

All other parameters used in the mechanistic kidney model were defaultSimcyp Simulator values with no modification unless specifically stated. Thepemetrexed model assumptions were the following: 1) pemetrexed was notmetabolized, and the only site of elimination was the kidney; 2) the passivepermeability of pemetrexed in the vector control cell was representative of thepassive permeability of pemetrexed in the kidney cells; 3) OAT3 ishomogeneously expressed in the basolateral membrane of the three segmentsof the proximal tubular cells, and it only transports pemetrexed in one direction,from the blood into the cells; 4) the efflux of pemetrexed from the proximaltubular cells is governed by only one transporter, OAT4, which is expresseduniformly on the apical membrane of the three segments of proximal tubularcells and transports pemetrexed only in one direction, from inside the cells outto into the tubular fluid compartment.

Ibuprofen PBPK Model. A full PBPK model was built for ibuprofen in theSimcyp Simulator (version 12.1) using the Rodgers and Rowland (2006) methodwith rapid equilibrium between plasma and tissues. The parameters used in thePBPK model for ibuprofen can be found in Table 2. The dosing regimen ofibuprofen consisted of nine doses of 400 mg, taken every 6 hours for 2 daysbefore the administration of pemetrexed, and a tenth dose of ibuprofen taken 1hour before pemetrexed infusion (Sweeney et al., 2006). The model was based onthe physicochemical properties and pharmacokinetic parameters of ibuprofencollected from published data (Yee, 1997; Davies, 1998; Chen et al., 2012). Theabsorption of ibuprofen was modeled using the advanced absorption anddissolution model in the Simcyp Simulator, where the human effective jejunalpermeability (Peff) was predicted from Caco-2 permeability values (Yee, 1997).The intrinsic solubility of ibuprofen was 0.06 mg/ml, and the model assumed animmediate release formulation (Shaw et al., 2005).

Virtual Population. The virtual population used in the simulations was builtto replicate the cancer population in pemetrexed published clinical studies(Sweeney et al., 2006) (Table 3). The population was based on the northEuropean Caucasian with an average body weight of 76.8 kg, age ranging from34 to 80 years old, and 30% of the subjects were female. The relative abundancesand corresponding coefficient of variation (60%) of the transporters in the threesegments of the proximal tubules were the same. No genetic polymorphisms wereconsidered.

Parameter Sensitivity Analysis and Optimization. A parameter sensitivityanalysis was performed to evaluate the effect of model parameter values on theAUC0–inf and Cmax of pemetrexed. The first analysis was run for those parameterswith high uncertainty or variability, such as the difference in expression oractivity of the transporters in vitro versus in vivo. The model was optimizedaccording to the results of the sensitivity analysis using the plasma concentration-time profiles of 25 patients who were administered 500 mg/m2 of pemetrexedsupplemented with folic acid and vitamin B12. The parameter estimation wasperformed using the hybrid minimization method in the Simcyp Simulator andweighted least squares as the objective function.

Model Verification. The PBPK model predictions for AUC0–inf and Cmax

over a range of pemetrexed doses from 50 to 600 mg/m2 were compared with theobserved ones in published clinical studies (Latz et al., 2006; Mita et al., 2006;

Nakagawa et al., 2006; Sweeney et al., 2006). Predicted AUC0–inf was calculatedby dividing the dose by the predicted clearance. The observed pharmacokineticparameters were calculated using noncompartmental analysis, according to thepublished clinical studies.

Drug-Drug Interaction Predictions. The interaction between ibuprofen andpemetrexed occurred at both OAT3 and OAT4. The Ki and fu,inc values wereobtained from in vitro experiments (discussed earlier) and are listed in Table 2. Toevaluate the effect of the interaction between pemetrexed and ibuprofen, wecalculated and compared the observed and predicted AUC0–inf and Cmax ratios.

The AUC0–inf ratio is defined as the pemetrexed AUC0–inf when pemetrexedis coadministered with ibuprofen, over the AUC0–inf of pemetrexed whenpemetrexed is administered alone. Likewise, the Cmax ratio is defined as themaximum concentration of pemetrexed when ibuprofen is coadministered overthe pemetrexed Cmax the when pemetrexed is administered alone.

For the interaction model, the assumptions were the following. 1) The interactionoccurs via OAT3 and OAT4 in the proximal tubular cells of the kidney. 2)Ibuprofen exerts its inhibition in a competitive manner for both OAT3 and OAT4.3) Both the S and R enantiomers of ibuprofen have the same affinity for OATtransporters. 5) Ibuprofen metabolites do not interact with OAT3 and OAT4.

Results

Preliminary In Vitro Screen. [14C]Pemetrexed uptake wassignificantly higher in OAT3 and OAT4 transfected HeLa cells comparedwith the control (Fig. 1). The uptake of [14C]pemetrexed by all othertransporters tested was not significantly different than the control. Basedon this, all subsequent in vitro uptake and inhibition studies wereperformed in OAT3, OAT4, and vector control transfected cells.In Vitro Transporter Uptake and Inhibition Studies. The results

of the qualification experiments, shown in Fig. 2A, demonstrated thatthe transfected HEK-PEAK hOAT3 and hOAT4 were functional, andthe uptake of E3S, a known substrate for these transporters, is at least100-fold higher than the vector control cells. When the OAT3 andOAT4 transfected cells were cotreated with probenecid, the uptake ofE3S via OAT3 and OAT4 was substantially reduced by 97 and 74%,respectively.To probe the active and passive uptake of pemetrexed, kinetic

studies were performed in HEK-PEAK vector control, OAT3, and

TABLE 4

IC50 values of the inhibition of pemetrexed OAT-mediated uptake by differentNSAIDs in OAT3 and OAT4 transfected HEK-PEAK cells

The results are expressed as mean (% coefficient of variation), n = 3.

InhibitorIC50

OAT3 Mean OAT3 Mean % CV OAT4 Mean OAT4 Mean (% CV)

mM mg/ml mM mg/ml

Ibuprofen 4.15 0.86 23.5 37.2 7.67 45Diclofenac 3.7 0.76 21 2.63 0.54 9.4Naproxen 27.9 5.76 24 39 8.05 10.3Celecoxib 14.2 2.93 45 21 4.33 18.4

TABLE 5

DDI indexes for OAT3 and OAT4 used to rank and predict the risk of the interaction between pemetrexed anddifferent NSAIDs

Cmax values and unbound fractions were taken were taken from Goodman and Gilman (1941).

Compound Cmax [Iu] Unbound Cmax DDI Index OAT3 [Iu]/IC50 DDI Index OAT4 [Iu]/IC50

mM

Ibuprofen 155.1 1.55 0.38 0.042Diclofenac 6.3 0.03 0.01 0.012Naproxen 239.7 0.72 0.03 0.018Celecoxib 1.85 0.06 0.004 0.003

Predicting Renal OAT-Mediated DDIs for Pemetrexed 329

at ASPE

T Journals on Septem

ber 26, 2020dm

d.aspetjournals.orgD

ownloaded from

Page 6: Prediction of Renal Transporter Mediated Drug-Drug ...dmd.aspetjournals.org/content/dmd/43/3/325.full.pdf · cells expressing organic anion transporter (OAT) 3 or OAT4. The NSAIDs

OAT4 cells over a concentration range of 1.6 and 117 mM ofpemetrexed. As seen in Fig. 2B, the uptake of pemetrexed was linearup to 10 minutes in the conditions tested. Figure 2C shows saturableOAT3-mediated uptake of pemetrexed with a Vmax and Km of 185pmol/mg per million cells and 18.2 mM, respectively. The passiveintrinsic clearance of pemetrexed was estimated to be 3.98� 1023 ml/minper million cells. The uptake of pemetrexed in OAT4-transfected cellsdid not reach saturation in the conditions tested, so the Vmax and Km

values could not be calculated (Fig. 2D). As a result, the intrinsic uptakeclearance of OAT4 (CLint,T) was calculated by dividing the initial uptakeof pemetrexed by its corresponding concentration, and was estimated tobe 0.99 ml/min per million cells.The inhibition of pemetrexed uptake by several NSAIDs was studied

over a concentration range of 0.1 to 200 mM for the NSAIDs and 19.5 mMfor pemetrexed. Table 4 shows the IC50 values for OAT3 and OAT4 forall the tested inhibitors. Table 5 shows the reported maximum plasmaconcentration of the tested inhibitors, fraction unbound in plasma (fu),and calculated [Iu]/IC50 values for the NSAIDs (Goodman and Gilman,1941). The U.S. Food and Drug Administration’s 2012 draft guidelineon drug-drug interactions (DDIs) (http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm292362.pdf)concludes that an [Iu]/IC50 value higher than 0.1 indicates a high risk ofclinical drug interaction and low potential for false negative results.Ibuprofen was the only tested NSAID with an [Iu]/IC50 value above 0.1,so PBPK models were not built for the other NSAIDs.Pemetrexed PBPK Model. The bottom-up pemetrexed PBPK

model was built using the measured in vitro transport kinetic datawithout any scaling factors. The simulations predicted an AUC0–inf of362 mg/ml∙h (2.2-fold higher than the observed value), and a clearance

of 2.5 l/h (55% smaller than the observed value). Because thedifference between the in vitro and the in vivo amount and activity(relative expression factor/relative activity factor, REF/RAF) of thetransporters is unknown, the model was optimized by estimating theseparameters using clinical data. The REF/RAF value for OAT3 wasestimated to 5.3 using human pemetrexed plasma concentration-timeprofiles published by Sweeney et al. (2006). The REF/RAF for OAT4was set to 5.3 as well, which resulted in a lack of cellular accumulationand immediate excretion of pemetrexed in the urine. The optimizedPBPK model was able to recover the observed plasma concentration-time profiles of pemetrexed.Figure 3 shows the mean plasma concentrations of the predicted and

the observed individual plasma concentration-time profiles of pemetrexedafter a single intravenous 10-minute infusion. The lines for the 5 and 95%confidence intervals are also shown in the graph. The observed/predictionratios for the Cmax, AUC0–inf , and clearance were all within 0.91 and1.04. For AUC0–inf, the observed and predicted geometric means were164.7 and 158 mg/ml per hour, respectively. The observed and predictedgeometric means for Cmax were 102.1 and 111.6 mg/ml, respectively. Forplasma clearance, the observed and predicted geometric mean were 5.6and 5.7 l/h, respectively.Impact of OAT3 and OAT4 on the PBPK Model Predictions.

The impact of adding the kidney transporters to the PBPK model wasevaluated by comparing the predicted against the observed pemetrexedpharmacokinetic parameters (Rinaldi et al., 1999). As shown onTable 6, when OAT3 is not added to the basolateral membrane of theproximal tubular cells in the model, the clearance of pemetrexed wasunderpredicted by 74% and the predicted AUC0–inf was 3.8-foldhigher. When OAT3 was present but OAT4 was not added to theapical membrane of the proximal tubular cells in the kidney model, theplasma concentrations were not affected, but the fraction excreted inurine unchanged (fe,0–24) was underpredicted by 70%. The predictedmaximum intracellular drug concentration in the proximal tubular cellswas 1000-fold in the absence of OAT4 compared with the model thathad this transporter on the apical membrane.Model Verification. The model was tested by comparing the

predicted versus the observed Cmax and AUC0–inf for a range of pemetrexeddoses between 50 and 700 mg/m2. The observed pharmacokineticvalues were extracted from a phase 1 single-dose escalation study(Rinaldi et al., 1999). Figure 4A shows the predicted and observedAUC0–inf versus pemetrexed dose in mg/m2. In both the predictedand observed settings, the AUC0–inf increased linearly with dose. Theslopes of the lines are very similar, 0.4283 and 0.4896 for the predictedand observed values, respectively (observed/predicted ratio = 1.14).Figure 4B shows the predicted and observed maximum plasma con-centration versus pemetrexed dose in mg/m2. Again, the Cmax changedlinearly with the dose, and the model predicts a similar slope of 0.243and 0.219 for the observed and predicted values, respectively (observed/predicted ratio = 1.11).

Fig. 3. Results of the pemetrexed PBPK model simulation. Observed and predictedplasma concentration-time profiles after the administration of a single intravenousinfusion of 500 mg/m2 of pemetrexed. The points represent the individual measuredplasma concentrations of pemetrexed. The solid black line represents the predictedmean concentrations, and the thin gray lines represent the 5 and 95% confidenceintervals of the prediction.

TABLE 6

Effect of the addition of OAT3 and OAT4 to the kidney model

Observed values are from Sweeney et al. (2006).

Parameter Observed ValuePredicted

Both OAT3 and OAT4 No OAT3 No OAT4

Cmax (mg/ml) 102.1 111.6 116.6 111.5AUC0–inf (mg/ml per hour) 164.7 158 618.6 159.2CL plasma (l/h) 5.6 5.7 1.48 5.8fe,0–24 0.8–0.9 1 0.7 0.3Maximum amount of drug in renal cells (mg) NA 0.01 0.01 667.2

330 Posada et al.

at ASPE

T Journals on Septem

ber 26, 2020dm

d.aspetjournals.orgD

ownloaded from

Page 7: Prediction of Renal Transporter Mediated Drug-Drug ...dmd.aspetjournals.org/content/dmd/43/3/325.full.pdf · cells expressing organic anion transporter (OAT) 3 or OAT4. The NSAIDs

Drug-Drug Interaction Predictions. Ibuprofen plasma concen-trations were not measured during the clinical study, so the predictedplasma concentration-time profiles of ibuprofen at the time ofpemetrexed administration were compared against data from theliterature (Aarons et al., 1983a,b; Grennan et al., 1983; Lockwoodet al., 1983a,b; Albert et al., 1984; Wagner et al., 1984; Davies, 1998).The pharmacokinetics of ibuprofen depend on several factors(i.e., formulation, fasted/fed state, physiology, and disease state), soa meta-analysis was performed to extract pharmacokinetic parametersfrom studies where ibuprofen was dosed at 400 mg in both healthysubjects and patients. Table 7 lists the reported and predicted steady-state Cmax, time of maximum concentration (Tmax), and AUC values foribuprofen dosed at 400 mg every 6 hours. The predicted Tmax and Cmax

of ibuprofen at steady state were 3 and 7%, respectively, lower than theaverage of the values found in the literature (Aarons et al., 1983a;Grennan et al., 1983; Ragni et al., 1992; Davies, 1998). The predictedplasma exposure of ibuprofen between 42 and 48 hours (AUC42–48h)was 1.01-fold higher than the reported value.Figure 5A shows the predicted mean plasma concentrations and the 5

and 95% confidence intervals of ibuprofen. The results of the interactionmodel between ibuprofen and pemetrexed are shown in Fig. 5B. Thepemetrexed AUC ratio was predicted to be 1.23 compared with 1.2

(1.11–1.31) observed, and the predicted Cmax ratio was 1.01 comparedwith 1.15 (1.03–1.28) observed in patients.

Discussion

The goal of this study was to develop a strategy to predict clinicallyrelevant transporter-mediated DDIs for pemetrexed using in vitro data,and to build confidence for the future prediction of drug interactionsfor renally cleared drugs. The first step in the strategy involved the invitro identification of the transporters responsible for the active tubularsecretion of pemetrexed. This was performed using a single time andsingle concentration assay in transiently transfected HeLa cells. OAT3and OAT4 were identified as the transporters likely to be involved inthe active tubular secretion of pemetrexed.Under initial rate conditions in HEK-PEAK cells stably transfected

with OAT3 and OAT4, we determined the Vmax, Km, and CLint, andemployed these parameters in a PBPK model to reproduce thedisposition of pemetrexed in humans. The initial bottom-up PBPKmodel performed well and predicted the shape of the concentration-timecurve, but the predicted systemic clearance was 55% lower than theobserved. A parameter sensitivity analysis was conducted to identifypossible causes of the under prediction of pemetrexed clearance.Based on the sensitivity analysis and the expected uncertainties, we

hypothesized that the REF/RAF of OAT3 was the most likelycontributor to this underprediction. The REF/RAF scaling factor wasestimated to be 5.3 using a hybrid minimization method and weightedleast squares as the objective function. The middle-out approach thatemployed the scaling factor recovered the shape of the concentration-time profile and systemic clearance accurately (Table 6).The need of scaling factors for transport parameters obtained in vitro

is well recognized in the prediction of drug absorption, distribution, andelimination via transporters (Bouzom et al., 2012; Tsamandouras et al.,2013; Hsu et al., 2014). In some cases, the scaling factor may representthe differences in transporter protein abundance between in vitro and invivo, but it may also reflect the dissimilarities in activity per milligramof protein owing to several factors, including membrane environment ordifferences in posttranslational modifications (Straumann et al., 2006;Croset et al., 2012). Is important to note that in this study the Vmax andKm values were estimated from in vitro experiments performed at roomtemperature and not at 37�C. Rates of transport at room temperature aregenerally lower than those observed at 37�C, so the scaling factorshould be lower when parameters are obtained at 37�C. This wasdemonstrated by Jackson and Halestrap (1996) for the active uptake ofL-lactate into hepatocytes by the hepatic monocarboxylate transporter.The model encompassed several important assumptions. First, it was

assumed that the OATs identified in the transporter screen were the onlytransporters involved in the tubular secretion of pemetrexed. We assumedthat the uptake of pemetrexed into the tubular cells was unidirectionaland driven by blood concentrations and OAT3 intrinsic clearance. It wasalso assumed that the concentrations of the cotransported dicarboxylates,such as a-ketoglutarate and succinate, were not rate limiting. We alsoassumed unidirectional flow of pemetrexed from the cells into the tubular

Fig. 4. (A) Effect of increasing dose on pemetrexed plasma AUC0–inf. The opencircles represent the predicted mean AUC0–inf, and the solid circles represent theobserved values. Data are represented as mean6 S.D. (B) Effect of increasing dose onpemetrexed plasma Cmax. (s) Predicted mean plasma Cmax values and (d) observedvalues. Data are represented as mean 6 S.D.

TABLE 7

Observed and predicted ibuprofen pharmacokinetic parameters at steady state

Steady-state (ss) values obtained via Aarons et al. (1983a).

ParameterObserved ValueMean (% CV)

Predicted ValueMean (% CV)

Observed/Predicted

Tmax,ss (hour) 1.3 (14) 1.3 (19.5) 1.05Cmax,ss (mg/ml) 29.7 (19) 28.0 (22.7) 1.06AUCss (mg/ml per hour) 108.1 (12.3) 119.6 (35.3) 0.90

Predicting Renal OAT-Mediated DDIs for Pemetrexed 331

at ASPE

T Journals on Septem

ber 26, 2020dm

d.aspetjournals.orgD

ownloaded from

Page 8: Prediction of Renal Transporter Mediated Drug-Drug ...dmd.aspetjournals.org/content/dmd/43/3/325.full.pdf · cells expressing organic anion transporter (OAT) 3 or OAT4. The NSAIDs

fluid mediated by OAT4. Despite the fact that pemetrexed has beenshown to be a substrate of MRP2 and MRP5, OAT4 was chosen as theonly mechanism of active transport across the apical membrane giventhat its pemetrexed intrinsic clearance (2.5 ml/min per mg protein) is atleast 10-fold higher than the reported ones for MRP2 (0.27 ml/min permg protein) and MRP5 (0.04 ml/min per mg protein) (Pratt et al., 2002).It was also assumed that the passive permeability of pemetrexed wasnegligible based on the in vitro measurements. Under these conditionsthe OAT3-mediated step in the tubular secretion was an identifiableparameter and could be estimated using plasma curves. In comparison,the OAT4-mediated step in the model is nonidentifiable because we donot have tubular concentrations nor do we have urinary excretion curveswith samples collected over short intervals. Therefore, OAT4 wasnot scaled, resulting in complete urinary excretion and no cellularaccumulation of pemetrexed. This is consistent with the absence of

renal cytotoxicity and the 70–90% observed urinary recovery ofpemetrexed (Rinaldi, 1999; Rinaldi et al., 1999).One of the objectives of the current work was to test whether the in

vitro inhibition potency of OATs could be confidently employed to predictthe DDI observed in vivo. Using the transfected OAT3 and OAT4 HEK-PEAK cells, we determined the IC50 values for commonly used NSAIDs.Of the NSAIDs tested, ibuprofen was identified as the most likely to causea DDI with pemetrexed using the ratio of the unbound plasma Cmax to theIC50 (Table 5). To foresee the effect of ibuprofen on the tubular secretionof pemetrexed, a PBPK model was constructed for ibuprofen based onpharmacokinetic data from the literature (Table 7). The combination of thepredicted unbound ibuprofen plasma concentration-time profile and the invitro inhibition constants (Ki) for OAT3 and OAT4 resulted in an accurateprediction of the increase in pemetrexed exposure.It is important to note that although the model included the in-

hibition of both OAT3 and OAT4, the predicted plasma clearance ofpemetrexed was only affected by inhibition of OAT3. The inhibitionof OAT4 by ibuprofen was shown to have no effect on the plasmaclearance of pemetrexed (Fig. 6). This is not surprising because theplasma clearance will be dependent on the uptake into the proximaltubular cells and not on the efflux of the drug from the cells into thetubules (Fig. 6). Another important assumption of the current in vitroto in vivo extrapolation approach was that only the parent drug wasresponsible for inhibiting the active transport, and that ibuprofenmetabolites played no significant role in the inhibition. We believe thisassumption is valid because the levels of ibuprofen in plasma areconsiderably higher (approximately 10-fold) than the levels of its twoprimary metabolites (Mills et al., 1973).An important strength of mechanistic PBPK modeling is the ability

to simulate the outcome of different physiologic or pathologic changesthat have not been specifically studied. This is illustrated in the case of

Fig. 5. Results of the PBPK simulations for ibuprofen and for interaction betweenpemetrexed and ibuprofen. (A) Predicted plasma concentration-time profiles ofibuprofen after oral administration of multiple doses of 400 mg the NSAID. The thickblack line represents the predicted mean concentrations in plasma, and the thin bluelines represent the 5 and 95% confidence intervals of the predictions. (B) Predictedpemetrexed plasma concentration-time profiles after a single intravenous 10-minuteinfusion of 500 mg/m2 of pemetrexed without (red lines) and with (blue lines)coadministration of ibuprofen in clinical subjects. The thick lines represent the meanvalues and the thin dotted lines represent the 5 and 95% confidence intervals,respectively. Blue closed diamonds represent the observed pemetrexed concentration-time points when pemetrexed was coadministered with ibuprofen. The red crossesrepresent the observed concentration-time points when pemetrexed was given alone.

Fig. 6. Parameter sensitivity analyses for pemetrexed plasma clearance. (A) Effectof the relative expression factors/activities of OAT3 and OAT4 on the plasmaclearance of pemetrexed. (B) Effect of the inhibition constant of ibuprofen (Ki)OAT3 and OAT4 on the plasma clearance of pemetrexed.

332 Posada et al.

at ASPE

T Journals on Septem

ber 26, 2020dm

d.aspetjournals.orgD

ownloaded from

Page 9: Prediction of Renal Transporter Mediated Drug-Drug ...dmd.aspetjournals.org/content/dmd/43/3/325.full.pdf · cells expressing organic anion transporter (OAT) 3 or OAT4. The NSAIDs

pemetrexed by examining the effect of renal impairment on the plasmaclearance. Under the assumptions that the glomerular filtration rateand active tubular secretion intrinsic clearance decrease proportionallyin renal insufficiency and that the plasma binding of pemetrexed is notaltered, we predict a plasma clearance of 53.8 ml/min in patients withmoderate renal insufficiency (glomerular filtration rate = 40–59 ml/min).However, in this case, a clinical study had been performed to determinethe effect of renal insufficiency on pemetrexed pharmacokinetics incancer patients (Mita et al., 2006). In that study, the observed plasmaclearance of pemetrexed in the moderate renal insufficiency group was54.7 ml/min (34% coefficient of variation), which is in good agreementwith the prediction. The recruitment of the severe renal insufficiency(glomerular filtration rate ,20 ml/min) group was problematic in thatstudy, resulting in a single patient being studied. This is an examplewhere mechanistic PBPK modeling could be used to estimate the effectof altered physiology/pathology in situations that are difficult to study.Similarly, scenarios involving the combinations of DDIs and diseasestates could be studied.One the main challenges of mechanistic PBPK modeling of renal

elimination is the lack of information on the intrinsic clearance of effluxtransporters such as OAT4 because plasma concentration-time curvesare insensitive to this parameter (Fig. 6). To resolve this shortcoming,the measurement of pemetrexed concentrations in tubular cells ortubular fluid is necessary. A potential surrogate would be urineconcentrations if the study design were customized for this purpose andurine is collected quantitatively, at sufficient frequency, and at earlytime intervals, but this is rarely achieved in a clinical study. To date, thesuccessful implementation of this approach is lacking; consequently, thequantitation of renal tubular efflux transporter intrinsic clearance willusually be poorly defined.In conclusion, we demonstrated for the first time that pemetrexed is

transported by OAT3 and OAT4, and not by other solute carriertransporters that are involved in active secretion in the kidney.Importantly, we detail how the in vitro transport parameters for OAT3 andOAT4 can define the structure of a PBPK model that will predict the renalclearance of pemetrexed within 2-fold of the observed value. Once thestructure of the model was defined, we demonstrated for the first time thatthe decrease in the pemetrexed OAT3-mediated uptake and therefore therenal clearance could be predicted using in vitro inhibition parameters forOAT3. In this translational framework, bottom-up PBPK models area powerful tool for the prediction of pharmacokinetics in the presence oftransporter-mediated drug interactions and will be enhanced when theaccurate quantitation of protein concentration in in vitro and in vivosystems is available. However, it is likely the in vitro systems will notcompletely reproduce the transport rates and affinities that exist in vivoand the middle-out parameter optimization approach will be a necessarystep in model verification. Ongoing work is designed to determinewhether the current model paradigm accurately predicts the pharmaco-kinetics of other OAT substrates and the interactions with inhibitors.

Authorship ContributionsParticipated in research design: Posada, Bacon, Kim, Hall, Hillgren.Conducted experiments: Posada, Bacon, Tirona, Pak.Performed data analysis: Posada, Bacon, Pak, Higgins, Schneck.Wrote or contributed to the writing of the manuscript: Posada, Bacon,

Tirona, Hall, Hillgren,

References

Aarons L, Grennan DM, Rajapakse C, Brinkley J, Siddiqui M, Taylor L, and Higham C (1983a)Anti-inflammatory (ibuprofen) drug therapy in rheumatoid arthritis—rate of response and lackof time dependency of plasma pharmacokinetics. Br J Clin Pharmacol 15:387–388.

Aarons L, Grennan DM, and Siddiqui M (1983b) The binding of ibuprofen to plasma proteins.Eur J Clin Pharmacol 25:815–818.

Albert KS, Gillespie WR, Wagner JG, Pau A, and Lockwood GF (1984) Effects of age on theclinical pharmacokinetics of ibuprofen. Am J Med 77 (1A):47–50.

Avdeef A, Berger CM, and Brownell C (2000) pH-metric solubility. 2: correlation between theacid-base titration and the saturation shake-flask solubility-pH methods. Pharm Res 17:85–89.

Avdeef A, Box KJ, Comer JE, Gilges M, Hadley M, Hibbert C, Patterson W, and Tam KY (1999)PH-metric log P 11. pKa determination of water-insoluble drugs in organic solvent-watermixtures. J Pharm Biomed Anal 20:631–641.

Bajetta E, Celio L, Buzzoni R, Ferrari L, Marchiano A, Martinetti A, Longarini R, Becerra C,Ilardi C, and John W (2003) Phase II study of pemetrexed disodium (Alimta) administered withoral folic acid in patients with advanced gastric cancer. Ann Oncol 14:1543–1548.

Bakhiya A, Bahn A, Burckhardt G, and Wolff N (2003) Human organic anion transporter 3(hOAT3) can operate as an exchanger and mediate secretory urate flux. Cell Physiol Biochem13:249–256.

Bouzom F, Ball K, Perdaems N, and Walther B (2012) Physiologically based pharmacokinetic(PBPK) modelling tools: how to fit with our needs? Biopharm Drug Dispos 33:55–71.

Burckhardt G (2012) Drug transport by organic anion transporters (OATs). Pharmacol Ther 136:106–130.

Cassano WF (1989) Serious methotrexate toxicity caused by interaction with ibuprofen. Am JPediatr Hematol Oncol 11:481–482.

Chen Y, Jin JY, Mukadam S, Malhi V, and Kenny JR (2012) Application of IVIVE and PBPKmodeling in prospective prediction of clinical pharmacokinetics: strategy and approach duringthe drug discovery phase with four case studies. Biopharm Drug Dispos 33:85–98.

Cockcroft DW and Gault MH (1976) Prediction of creatinine clearance from serum creatinine.Nephron 16:31–41.

Croset A, Delafosse L, Gaudry JP, Arod C, Glez L, Losberger C, Begue D, Krstanovic A, RobertF, and Vilbois F, et al. (2012) Differences in the glycosylation of recombinant proteinsexpressed in HEK and CHO cells. J Biotechnol 161:336–348.

Cvetkovic M, Leake B, Fromm MF, Wilkinson GR, and Kim RB (1999) OATP and P-glycoprotein transporters mediate the cellular uptake and excretion of fexofenadine. DrugMetab Dispos 27:866–871.

Davies NM (1998) Clinical pharmacokinetics of ibuprofen. The first 30 years. Clin Pharmaco-kinet 34:101–154.

Erdman AR, Mangravite LM, Urban TJ, Lagpacan LL, Castro RA, de la Cruz M, Chan W, HuangCC, Johns SJ, and Kawamoto M, et al. (2006) The human organic anion transporter 3 (OAT3;SLC22A8): genetic variation and functional genomics. Am J Physiol Renal Physiol 290:F905–F912.

Frenia ML and Long KS (1992) Methotrexate and nonsteroidal antiinflammatory drug inter-actions. Ann Pharmacother 26:234–237.

Godinot N, Iversen PW, Tabas L, Xia X, Williams DC, Dantzig AH, and Perry WL, 3rd (2003)Cloning and functional characterization of the multidrug resistance-associated protein (MRP1/ABCC1) from the cynomolgus monkey. Mol Cancer Ther 2:307–316.

Goodman LS and Gilman A (1941) The Pharmacological Basis of Therapeutics: A Textbook ofPharmacology, Toxicology and Therapeutics for Physicians and Medical Students, Macmillan,New York.

Grennan DM, Aarons L, Siddiqui M, Richards M, Thompson R, and Higham C (1983) Dose-response study with ibuprofen in rheumatoid arthritis: clinical and pharmacokinetic findings. BrJ Clin Pharmacol 15:311–316.

Hagos Y, Bahn A, Vormfelde SV, Brockmöller J, and Burckhardt G (2007a) Torasemidetransport by organic anion transporters contributes to hyperuricemia. J Am Soc Nephrol 18:3101–3109.

Hagos Y, Stein D, Ugele B, Burckhardt G, and Bahn A (2007b) Human renal organic aniontransporter 4 operates as an asymmetric urate transporter. J Am Soc Nephrol 18:430–439.

Hsu V, de L T Vieira M, Zhao P, Zhang L, Zheng JH, Nordmark A, Berglund EG, GiacominiKM, and Huang SM (2014) Towards quantitation of the effects of renal impairment andprobenecid inhibition on kidney uptake and efflux transporters, using physiologically basedpharmacokinetic modelling and simulations. Clin Pharmacokinet 53:283–293.

Jackson VN and Halestrap AP (1996) The kinetics, substrate, and inhibitor specificity of themonocarboxylate (lactate) transporter of rat liver cells determined using the fluorescent in-tracellular pH indicator, 29,79-bis(carboxyethyl)-5(6)-carboxyfluorescein. J Biol Chem 271:861–868.

Khamdang S, Takeda M, Noshiro R, Narikawa S, Enomoto A, Anzai N, Piyachaturawat P,and Endou H (2002) Interactions of human organic anion transporters and human organiccation transporters with nonsteroidal anti-inflammatory drugs. J Pharmacol Exp Ther 303:534–539.

Kurata T, Iwamoto T, Kawahara Y, and Okuda M (2014) Characteristics of pemetrexed transport byrenal basolateral organic anion transporter hOAT3. Drug Metab Pharmacokinet 29:148–153.

Latz JE, Chaudhary A, Ghosh A, and Johnson RD (2006) Population pharmacokinetic analysis often phase II clinical trials of pemetrexed in cancer patients. Cancer Chemother Pharmacol 57:401–411.

Lockwood GF, Albert KS, Gillespie WR, Bole GG, Harkcom TM, Szpunar GJ, and Wagner JG(1983a) Pharmacokinetics of ibuprofen in man. I. Free and total area/dose relationships. ClinPharmacol Ther 34:97–103.

Lockwood GF, Albert KS, Szpunar GJ, and Wagner JG (1983b) Pharmacokinetics of ibuprofen inman—III: Plasma protein binding. J Pharmacokinet Biopharm 11:469–482.

Mills RF, Adams SS, Cliffe EE, Dickinson W, and Nicholson JS (1973) The metabolism ofibuprofen. Xenobiotica 3:589–598.

Mita AC, Sweeney CJ, Baker SD, Goetz A, Hammond LA, Patnaik A, Tolcher AW, Villalona-Calero M, Sandler A, and Chaudhuri T, et al. (2006) Phase I and pharmacokinetic study ofpemetrexed administered every 3 weeks to advanced cancer patients with normal and impairedrenal function. J Clin Oncol 24:552–562.

Motohashi H, Sakurai Y, Saito H, Masuda S, Urakami Y, Goto M, Fukatsu A, Ogawa O, and InuiK (2002) Gene expression levels and immunolocalization of organic ion transporters in thehuman kidney. J Am Soc Nephrol 13:866–874.

Nakagawa K, Kudoh S, Matsui K, Negoro S, Yamamoto N, Latz JE, Adachi S, and Fukuoka M(2006) A phase I study of pemetrexed (LY231514) supplemented with folate and vitamin B12in Japanese patients with solid tumours. Br J Cancer 95:677–682.

Nozaki Y, Kusuhara H, Endou H, and Sugiyama Y (2004) Quantitative evaluation of the drug-drug interactions between methotrexate and nonsteroidal anti-inflammatory drugs in the renaluptake process based on the contribution of organic anion transporters and reduced folatecarrier. J Pharmacol Exp Ther 309:226–234.

Predicting Renal OAT-Mediated DDIs for Pemetrexed 333

at ASPE

T Journals on Septem

ber 26, 2020dm

d.aspetjournals.orgD

ownloaded from

Page 10: Prediction of Renal Transporter Mediated Drug-Drug ...dmd.aspetjournals.org/content/dmd/43/3/325.full.pdf · cells expressing organic anion transporter (OAT) 3 or OAT4. The NSAIDs

Pratt SE, Emerick RM, Horwit L, Gallery M, Lesoon A, Jink S, Chen VJ, and Dantzig AH (2002)Multidrug resistance proteins (MRP) 2 and 5 transport and confer resistance to Alimta (Ab-stract). Proc Am Assoc Cancer Res 43:782.

Ragni MV, Miller BJ, Whalen R, and Ptachcinski R (1992) Bleeding tendency, platelet function,and pharmacokinetics of ibuprofen and zidovudine in HIV(+) hemophilic men. Am J Hematol40:176–182.

Rinaldi DA (1999) Overview of phase I trials of multitargeted antifolate (MTA, LY231514).Semin Oncol 26(2, Suppl 6)82–88.

Rinaldi DA, Kuhn JG, Burris HA, Dorr FA, Rodriguez G, Eckhardt SG, Jones S, Woodworth JR,Baker S, and Langley C, et al. (1999) A phase I evaluation of multitargeted antifolate (MTA,LY231514), administered every 21 days, utilizing the modified continual reassessment methodfor dose escalation. Cancer Chemother Pharmacol 44:372–380.

Rodgers T, Leahy D, and Rowland M (2005) Physiologically based pharmacokinetic modeling 1:predicting the tissue distribution of moderate-to-strong bases. J Pharm Sci 94:1259–1276.

Rodgers T and Rowland M (2006) Physiologically based pharmacokinetic modelling 2: predicting thetissue distribution of acids, very weak bases, neutrals and zwitterions. J Pharm Sci 95:1238–1257.

Scagliotti GV, Shin DM, Kindler HL, Vasconcelles MJ, Keppler U, Manegold C, Burris H,Gatzemeier U, Blatter J, and Symanowski JT, et al. (2003) Phase II study of pemetrexed withand without folic acid and vitamin B12 as front-line therapy in malignant pleural mesotheli-oma. J Clin Oncol 21:1556–1561.

Shaw LR, Irwin WJ, Grattan TJ, and Conway BR (2005) The effect of selected water-solubleexcipients on the dissolution of paracetamol and Ibuprofen. Drug Dev Ind Pharm 31:515–525.

Shih C, Chen VJ, Gossett LS, Gates SB, MacKellar WC, Habeck LL, Shackelford KA, MendelsohnLG, Soose DJ, and Patel VF, et al. (1997) LY231514, a pyrrolo[2,3-d]pyrimidine-based antifolatethat inhibits multiple folate-requiring enzymes. Cancer Res 57:1116–1123.

Straumann N, Wind A, Leuenberger T, and Wallimann T (2006) Effects of N-linked glycosylationon the creatine transporter. Biochem J 393:459–469.

Sweeney CJ, Takimoto CH, Latz JE, Baker SD, Murry DJ, Krull JH, Fife K, Battiato L, Cleverly A,and Chaudhary AK, et al. (2006) Two drug interaction studies evaluating the pharmacokinetics

and toxicity of pemetrexed when coadministered with aspirin or ibuprofen in patients with ad-vanced cancer. Clin Cancer Res 12:536–542.

Sweet DH, Chan LM, Walden R, Yang XP, Miller DS, and Pritchard JB (2003) Organic aniontransporter 3 (Slc22a8) is a dicarboxylate exchanger indirectly coupled to the Na+ gradient. AmJ Physiol Renal Physiol 284:F763–F769.

Takimoto CH, Hammond-Thelin LA, Latz JE, Forero L, Beeram M, Forouzesh B, de Bono J,Tolcher AW, Patnaik A, and Monroe P, et al. (2007) Phase I and pharmacokinetic study ofpemetrexed with high-dose folic acid supplementation or multivitamin supplementation inpatients with locally advanced or metastatic cancer. Clin Cancer Res 13:2675–2683.

Thyss A, Milano G, Kubar J, Namer M, and Schneider M (1986) Clinical and pharmacokineticevidence of a life-threatening interaction between methotrexate and ketoprofen. Lancet 1:256–258.

Tracy TS, Krohn K, Jones DR, Bradley JD, Hall SD, and Brater DC (1992) The effects ofa salicylate, ibuprofen, and naproxen on the disposition of methotrexate in patients withrheumatoid arthritis. Eur J Clin Pharmacol 42:121–125.

Tsamandouras N, Rostami-Hodjegan A, and Aarons L (2013) Combining the “bottom-up” and“top-down” approaches in pharmacokinetic modelling: fitting PBPK models to observedclinical data. Br J Clin Pharmacol DOI: 10.1111/bcp.12234 [published ahead of print].

Wagner JG, Albert KS, Szpunar GJ, and Lockwood GF (1984) Pharmacokinetics of ibuprofen inman IV: absorption and disposition. J Pharmacokinet Biopharm 12:381–399.

Yee S (1997) In vitro permeability across Caco-2 cells (colonic) can predict in vivo (smallintestinal) absorption in man—fact or myth. Pharm Res 14:763–766.

Address correspondence to: Dr. Maria M. Posada, Lilly Research Laboratories,Eli Lilly and Company, Indianapolis, IN 46285. E-mail: [email protected]

334 Posada et al.

at ASPE

T Journals on Septem

ber 26, 2020dm

d.aspetjournals.orgD

ownloaded from