throbber
0090-9556/06/3409-1480–1487$20.00
`DRUG METABOLISM AND DISPOSITION
`Copyright © 2006 by The American Society for Pharmacology and Experimental Therapeutics
`DMD 34:1480–1487, 2006
`
`Vol. 34, No. 9
`9001/3131990
`Printed in U.S.A.
`
`Physiologically Based Pharmacokinetic Modeling of FTY720
`(2-Amino-2[2-(-4-octylphenyl)ethyl]propane-1,3-diol hydrochloride)
`in Rats After Oral and Intravenous Doses
`
`Guy M. L. Meno-Tetang,1 Hongshan Li, Suzette Mis, Nancy Pyszczynski, Peter Heining,
`Philip Lowe, and William J. Jusko
`
`Novartis Pharma AG, Basel, Switzerland (G.M.L.M.-T., P.H., P.L.); and Department of Pharmaceutical Sciences, School of
`Pharmacy and Pharmaceutical Sciences, State University of New York, Buffalo, New York (H.L., S.M., N.P., W.J.J.)
`
`Received December 16, 2005; accepted May 31, 2006
`
`ABSTRACT:
`
`FTY720 (2-amino-2[2-(-4-octylphenyl)ethyl]propane-1,3-diol hy-
`drochloride) is a new sphingosine-1-phosphate receptor agonist
`being developed for multiple sclerosis and prevention of solid
`organ transplant rejection. A physiologically based pharmacoki-
`netic model was developed to predict the concentration of FTY720
`in various organs of the body. Single oral and intravenous doses of
`FTY720 were administered to male Wistar rats, with blood and
`tissue sampling over 360 h analyzed by liquid chromatography/
`tandem mass spectrometry. A well stirred model (perfusion rate-
`limited) described FTY720 kinetics in heart, lungs, spleen, muscle,
`kidneys, bone, and liver, with a permeability rate-limited model
`being required for brain, thymus, and lymph nodes. Tissue-to-
`blood partition coefficients (RT) ranged from 4.69 (muscle) to 41.4
`(lungs). In lymph nodes and spleen, major sites for FTY720-induced
`changes in sequestration of lymphocytes, RT values were 22.9 and
`
`34.7, respectively. Permeability-surface area products for brain,
`thymus, and lymph nodes were 39.3, 122, and 176 ml/min. Intrinsic
`hepatic clearance was 23,145 l/h/kg for the free drug in blood (fub
`0.000333); systemic clearance was 0.748 l/h/kg and terminal half-
`life was 23.4 h. The fraction orally absorbed was 71%. The model
`characterized well FTY720 disposition for this extensive dosing
`and tissue collection study in the rat. On scaling the model to dogs
`and humans, good agreement was found between the actual and
`predicted blood concentration-time profiles. More importantly,
`brain concentrations in dogs were well predicted from those of the
`rat. In absolute terms, the predictions were slightly lower than
`observed values, just under a 1.5-fold deviation, but the model
`accurately predicted the terminal elimination of FTY720 from the
`brain.
`
`FTY720 (2-amino-2[2-(-4-octylphenyl)ethyl]propane-1,3-diol hy-
`drochloride) is a new sphingosine-1-phosphate receptor agonist that is
`being developed for prevention of solid organ transplant rejection
`(Napoli, 2000). FTY720 exerts its immunomodulatory actions by
`affecting lymphocyte production (Yagi et al., 2000),
`trafficking
`(Chiba et al., 1998; Brinkmann et al., 2000, 2001), infiltration (Yana-
`gawa et al., 2000), and apoptosis (Enosawa et al., 1996; Bohler et al.,
`2000; Nagahara et al., 2000). The maximum effects of FTY720 on
`these immune responses are achieved at doses smaller than those
`producing effects against graft rejection (Yanagawa et al., 1998).
`
`This work was supported by Novartis Pharma AG and in part by Grant GM
`24211 from the National Institutes of Health. The liquid chromatograph/tandem
`mass spectrometer was obtained through a shared instrumentation grant
`(S10RR14573) from the National Center for Research Resources, National Insti-
`tutes of Health.
`1 Current affiliation: Serono International S.A, Geneva, Switzerland.
`Article, publication date, and citation information can be found at
`http://dmd.aspetjournals.org.
`doi:10.1124/dmd.105.009001.
`
`Regulation of gene expression may also account for pharmacological
`and toxicological effects of FTY720: for instance, a 26-week phar-
`macology study in rats using gene chips showed that, at doses of 0.3
`and 1.5 mg/kg/day, genes of B and T lymphocyte markers in blood
`(CD79 and CD3) were down-regulated (Novartis Pharma AG, internal
`communication).
`The elimination of FTY720 from the body occurs mainly via
`metabolism. Two primary pathways metabolize FTY720: 1) phos-
`phorylation at one of its two hydroxy groups (yielding FTY720-P)
`and 2) hydroxylation at the terminal methyl group (M12) (Novartis
`Pharma AG, internal communication). The blood clearance values
`for FTY720 in dogs, monkeys, and humans are 0.0617, 0.113,
`and 0.0433 ml/h/kg, respectively. The elucidation of the mecha-
`nism of action and pharmacodynamics of FTY720 on immune cells
`will necessitate the characterization of its disposition not only in
`blood but also in target organs such as lymph nodes, spleen, and
`thymus.
`The objective of this work was to develop a physiologically based
`pharmacokinetic (PBPK) model in rats to characterize the kinetics of
`
`ABBREVIATIONS: FTY720, 2-amino-2[2-(-4-octylphenyl)ethyl]propane-1,3-diol hydrochloride; PBPK, physiologically based pharmacokinetic;
`Y32919, 2-amino-2-[2-(4-octyloxyphenyl)ethyl]propane-1,3-diol hydrochloride; HPLC, high-performance liquid chromatography; LN,
`lymph
`nodes; AUC, area under the concentration-time curve; RT, tissue-to-blood partition coefficient; PST, permeability-surface area product; CLint,
`intrinsic clearance.
`
`1480
`
`Apotex v. Novartis
`IPR2017-00854
`NOVARTIS 2055
`
`

`

`FTY720 PBPK MODEL IN RAT, DOG AND HUMAN
`
`1481
`
`FTY720 both in major organs and in lymphatic tissues such as spleen,
`thymus, and lymph nodes.
`
`Materials and Methods
`
`Chemicals. Analytical grade FTY720 and Y32919 (internal standard;
`2-amino-2-[2-(4-octyloxyphenyl)ethyl]propane-1,3-diol hydrochloride) were
`supplied by Novartis Pharma AG (Basel, Switzerland). HPLC-grade dichlo-
`romethane and water were purchased from Burdick and Jackson (Muskegon,
`MI). Methanol optima grade and glacial acetic acid HPLC grade were pur-
`chased from Fisher Scientific (Fairlawn, NJ). tert-Butyl-methyl ether HPLC
`grade was purchased from Sigma Aldrich (Milwaukee, WI) and ammonium
`acetate microselect was purchased from Fluka (Milwaukee, WI).
`Animals. Normal male Wistar rats, 80 to 90 days old, with body weights
`ranging from 300 to 375 g, were used in this study (Harlan Sprague-Dawley
`Inc., Indianapolis, IN). The animals were housed in pairs in stainless steel
`cages in a controlled environment with a 12-h light/dark cycle. Filtered tap
`water and food were available ad libitum. Before the dosing day, animals were
`kept in the animal facility for at least 7 days for acclimatization.
`Dosing and Sampling. Single i.v. bolus doses of FTY720 (0.3, 2, and 4
`mg/kg) were given to rats via penal vein injection. For each group, three rats
`were sacrificed by aortic exsanguination under ketamine/xylazine anesthesia
`just before dosing and 0.5, 1, 3, 6, 12, 24, 48, 72, 120, 176, 240, and 360 h
`postdose. Arterial blood was collected, and lungs, heart, brain, kidneys, thy-
`mus, spleen, liver, muscle, fat, inguinal lymph nodes (LN), mesenteric LN,
`axillary LN, cervical LN, popliteal LN, and Peyer’s patches were dissected.
`Tissue collection was performed only after the 0.3 and 2 mg/kg doses. Samples
`collected at each time point from each of the three animals were pooled in
`equal volumes, homogenized in physiological buffer (4 times tissue weight, pH
`7.4), and stored at ⫺20°C.
`Single oral doses of FTY720 (2.8 and 7.5 mg/kg) were also administered to
`rats. Serial blood sampling was performed for venous blood. For the dose of
`2.8 mg/kg, spleen samples were collected at 1, 8, 12, 24, 48, 72, and 120 h.
`After administration of 7.5 mg/kg, brain, liver, lungs, muscle, and heart tissue
`samples were collected at 8 and 72 h.
`Eighteen male beagle dogs (3 per time point) aged 7 months and weighing
`7.6 to 10.1 kg received single oral doses of 10 mg/kg. At necropsy, on days 1,
`3, 7, 14, 21, and 28 after drug administration, the frontal lobe of the brain was
`collected.
`Clinical Study. Healthy male volunteers aged between 20 and 39 years,
`weight 68 to 95 kg, were given 1-mg doses of FTY720. Venous blood samples
`were obtained before FTY720 administration and then 1, 2, 4, 6, 8, 12, 16, 20,
`24, 36, 48, 72, 96, 120, and 168 h postdose. The study was carried out in
`compliance with the protocol and according to Good Clinical Practice with full
`informed consent according to the Declaration of Helsinki. Further details are
`available in a prior publication (Kovarik et al., 2004).
`Bioanalytical Instruments and FTY720 Assay. The liquid chromatogra-
`phy/tandem mass spectrometry system was a PE Sciex API 3000 with heated
`nebulizer interface (Applied Biosystems, Foster City, CA) together with Agi-
`lent 1100 pumps, detector, and autosampler (Agilent Technologies, Palo Alto,
`CA). The HPLC column was a 3.5 ␮ Symmetry Shield RP8, 4.6 ⫻ 50 mm
`(Waters Corp., Milford, MA).
`After thawing and rehomogenization of samples, FTY720 was extracted as
`follows. Aliquots (0.1 ml) of calibration standards, quality controls, or un-
`knowns were added to 0.1 ml of 100 ng/ml Y32919 in methanol, 0.5 ml of 0.1
`N sodium hydroxide solution added, followed by 6 ml of tert-butyl-methyl
`ether and dichloromethane (75:25 v/v). After shaking for 45 min followed by
`centrifugation for 10 min at 2000g, the organic phase was transferred and
`evaporated under N2, reconstituted with 150 ␮l of HPLC eluent, sonicated for
`5 min, mixed, and centrifuged for 5 min at 15,000g, and then injected into the
`liquid chromatograph/tandem mass spectrometer.
`The chromatography system was eluted with 70% methanol plus 30% 0.02
`M ammonium acetate at 1 ml/min. The parent molecular ion for FTY720 is
`308.3 Da (⫹1⫹ proton) with a daughter ion of 255.3 m/z. Y32919 has a parent
`of 324.4 Da and daughter of 271.4 m/z. The integrity of the original compounds
`was checked using the mass spectrometer, and there was no carryover into
`either the FTY720 or the Y32919 detection channels. The lower limit of
`quantitation was 0.5 ng/ml (11.9% CV) and the upper limit 2000 ng/ml (2.33%
`CV). The recovery was 86.1% at 30 ng/ml and 95.8% at 1250 ng/ml. The
`
`intraday and interday CV% ranges were 4.15 to 5.94% and 5.01 to 5.96%,
`respectively.
`Partition Coefficients. The AUC (area under the concentration-time
`curves) of drug in blood and tissues was calculated by
`
`AUC ⫽冘
`
`n⫺1
`
`i⫽0
`
`关共Ci⫹1 ⫹ Ci兲 䡠 共ti⫹1 ⫺ ti兲/2兴 ⫹
`
`Cn
`␭z
`
`(1)
`
`where Ci is the drug concentration at t ⫽ ti and ␭Z is the terminal slope of the
`
`concentration-time curve. The tissue-to-blood partition coefficient (RT) for
`noneliminating tissues was obtained from
`
`RT ⫽
`
`AUCT
`AUCV
`
`⬇ AUCT
`AUCA
`
`(2)
`
`where AUCT, AUCV, and AUCart are derived from drug concentration versus
`time profiles for tissues and venous and arterial blood. This equation was used
`to calculate RT for lungs, heart, brain, kidneys, thymus, spleen, muscle, and
`lymph nodes for the 0.3 and 2 mg/kg i.v. doses.
`The volume of distribution at steady state (VB,ss) was calculated as
`
`VB,ss ⫽ 冘
`
`non-elim
`
`关RT/B 䡠 VT兴 ⫹ RH/B 䡠 Vliver 䡠 共1 ⫺ Eliver兲 ⫹ Vblood
`
`(3)
`
`Tissue
`
`where VT represents specific tissue volumes and Eliver is the hepatic extraction
`ratio.
`Compartmental Analysis. Arterial blood concentration-time data for
`FTY720 after i.v. administration of 0.3 and 2 mg/kg doses were analyzed by
`compartmental analysis using the computer software WinNonlin ver. 4.0
`(Pharsight Corporation, Mountain View, CA).
`Model Development. Local models were developed with blood and organ
`concentration-time data obtained after single i.v. doses of 0.3 and 2 mg/kg.
`Data from each tissue were first considered individually. The choice of final
`structural models was based upon inspection of quality of fits to the data and
`the Akaike information criterion. Parameter values obtained with local models
`were then used as initial estimates for the whole body model. The fitted
`parameters were tissue-to-blood partition coefficients (RT), permeability-sur-
`face area products (PST), and liver intrinsic clearance (CLint). The first-order
`absorption rate constant (kabs) and fraction of drug absorbed (Fabs) were
`determined by adapting the i.v. PBPK model to oral data obtained after the
`single doses of 2.8 and 7.5 mg/kg. For the oral PBPK model, all parameters
`(RT, PST, and CLint) were maintained as constant. Only parameters kabs and
`Fabs were optimized. To verify the linearity of FTY720 kinetics with respect to
`dose, blood and tissue data obtained after the single 4 mg/kg i.v. dose were
`superimposed on the corresponding simulated curve based upon the parameters
`determined with the 0.3 and 2 mg/kg doses. Single i.v. bolus doses were
`regarded as a 7.5-s i.v. infusion, and drug was assumed to distribute evenly in
`the whole vein pool once injected. Based on metabolism and clearance data
`(Novartis Pharma AG, internal communication), the liver was considered as
`the sole eliminating organ.
`Local Models. FTY720 concentration-time data after i.v. doses were fitted
`
`to
`
`Cart ⫽ A ⫻ e⫺␣⫻t ⫹ B ⫻ e⫺␤⫻t ⫹ C ⫻ e⫺␥⫻t
`
`(4)
`
`Equations for well stirred organs (heart, lungs, muscle, spleen, liver, and
`kidneys) were
`
`dXT
`dt
`
`⫽ QT ⫻
`
`Xart
`VT
`
`⫺ QT ⫻
`
`XT
`共RT ⫻ VT兲
`
`(5)
`
`Equations for permeability rate-limited transport (brain, thymus, lymph
`nodes) were:
`
`Blood compartment
`
`dXT1
`dt
`
`⫽ QT ⫻
`
`Xart
`VT1
`
`⫺ QT ⫻
`
`XT1
`VT1
`
`⫺ PST ⫻ fub ⫻
`
`⫹ PST ⫻冉fub
`
`RT
`
`冊 ⫻
`
`XT2
`VT1
`
`XT1
`VT1
`
`(6)
`
`

`

`1482
`
`MENO-TETANG ET AL.
`
`TABLE 1
`Standard physiologic parameters for rats weighing 250 g
`
`Organ
`
`Lungs
`Braina,b
`Heart
`Stomach
`Gut
`Pancreas
`Spleen
`Kidneys
`Thymusa,b
`
`Mass
`
`g
`1.0
`1.7
`0.8
`1.1
`10
`1.3
`0.6
`2.3
`0.7
`
`Q
`
`ml/min
`43.0
`1.33
`3.92
`1.13
`7.52
`0.52
`0.63
`9.23
`0.3
`
`Organ
`
`Muscle
`Lymph nodesa,b,c
`Liver
`Skin
`Bone
`Fat
`Arterial blood
`Venous blood
`
`Mass
`
`g or ml
`122
`0.9
`10.3
`40
`15.8
`10
`5.6
`11.3
`
`Q
`
`ml/min
`7.5
`0.16
`2.0
`5.83
`2.53
`0.4
`
`a Fractional volume of vascular space: brain, 0.014; thymus and lymph nodes, 0.009.
`b Fractional volume of interstitial space: brain, 0.188; thymus and lymph nodes, 0.150.
`c Sum of mesenteric, axillary, inguinal, cervical, Peyer’s patches, and popliteal lymph nodes.
`
`Interstitial and intracellular compartments
`
`dXT2
`dt
`
`⫽ PST ⫻ fub ⫻
`
`XT1
`VT2
`
`⫺ PST ⫻冉fub
`
`RT
`
`冊 ⫻
`
`XT2
`VT2
`
`(7)
`
`where QT is organ blood flow rate, VT is organ volume, Xart is input from
`arterial blood, PST is permeability-surface area product, RT is tissue-to-blood
`partition coefficient, fwT is fractional volume of vascular and/or interstitial
`space, VT1 is volume of blood ⫹ interstitial fluid (VT1
`⫽ fwT
`⫻ VT), VT2 is
`⫽ VT
`⫺ VT1), and fub is free fraction of
`volume of intracellular space (VT2
`FTY720 in blood.
`Organ volumes (Vi) were scaled from published data for a 250-g rat
`(Bernareggi and Rowland, 1991; Davies and Morris, 1993) using
`
`V2 ⫽
`
`BW2 䡠 V1
`BW1
`
`(8)
`
`where BW1 is the rat standard body weight (250 g) and BW2 is the actual body
`weight of animals used in the current study (mean 362 g). Blood flow of tissues
`except thymus and lymph nodes was similarly scaled using
`
`Q2 ⫽
`
`BW2 䡠 Q1
`BW1
`
`(9)
`
`Blood flow for thymus and lymph nodes was scaled from Cahill and Trnka
`(1978). All physiologic parameters used for this analysis are listed in Table 1.
`Drug concentrations for each tissue were converted to ng/ml from ng/g using
`tissue density.
`Model Equations for Whole Body Model. The whole body PBPK model
`for FTY720 is depicted in Fig. 1. It depicts the body as composed of 13 tissue
`compartments and 2 blood compartments (arterial and venous pools) with the
`lungs closing the loop. Drug is placed in the venous compartment. All equa-
`tions were solved simultaneously using the maximum likelihood estimator in
`ADAPT II.4 (D’Argenio and Schumitzky, 1997). Although FTY720 concen-
`trations in bone, gastrointestinal tract, and skin were not collected, these organs
`were included as lumped compartments representing the gastrointestinal
`tract—splanchnic compartment—and “rest-of-body”. It was assumed that
`these compartments obey the distribution characteristics of a well stirred
`model.
`Interspecies Scale-Up. The PBPK model developed for rat was scaled-up
`to dog and humans. Physiological values (organ volume and blood flow rate)
`were taken from the literature. Unbound equilibrium distribution ratio (RTu) in
`dogs and humans for each tissue was calculated from RT values obtained with
`the rat model and corresponding fub values for each species (fub values were
`0.000241 in dogs and 0.000361 in humans). The PST values for brain, thymus,
`and lymph nodes for dog and human were predicted from the measurement in
`rat by use of an allometric equation
`
`PST ⫽ A共M兲B
`
`(10)
`
`where M is organ mass (or weight) and A and B are allometric coefficients. The
`B value of 0.67 was fixed, assuming that permeability of tissue cellular
`membrane and organ structure is geometrically similar among mammals.
`
`FIG. 1. Whole body PBPK model for FTY720. Subscripts are: A, arterial blood; V,
`venous blood; b, brain; h, heart; li, liver; sp, spleen; lu, lungs; g, gut-stomach-
`pancreas; kd, kidney; f, fat; sk, skin; m, muscle; th, thymus; ln, lymph nodes; and
`bn, bone.
`
`To predict the blood concentration versus time profile for FTY720 in dogs,
`a first-order absorption rate constant (kabs) and bioavailability values were
`taken from the results of a compartmental analysis. Because no in vitro data
`were available for the biotransformation of FTY720 in dogs, the intrinsic
`clearance in dogs (CLintD) was calculated by nonlinear regression analysis with
`the whole body PBPK model, keeping all distribution parameters (RT and PST)
`identical to those from the rat after correction for binding in blood.
`To predict the concentration versus time profile for FTY720 in human
`blood, the first-order absorption rate constant (kabs) was taken from the results
`of compartmental analysis. Because no i.v. formulation for FTY720 is avail-
`able for humans, we assumed that humans had the same bioavailability (F) as
`rats. The intrinsic clearance in humans (CLint,H) value was calculated in two
`ways. First, CLint,H was predicted from the product of the human in vitro
`clearance determined with liver microsomes and the in vivo to in vitro CLint
`ratio from rats. Second, when human FTY720 concentration data became
`available, they were used to improve the estimate of human CLint,H, keeping all
`distribution parameters (RT and PST) identical to those from the rat after
`correction for binding in blood.
`
`Results
`Blood Profiles. Figures 2 and 3 depict blood concentration-time
`profiles for FTY720 after i.v. and oral administration. It appears from
`the graphs that the model captured the venous blood concentration-
`time profiles relatively well for the 0.3, 2, and 4 mg/kg i.v. doses. In
`contrast, arterial concentration-time profiles were somewhat overes-
`timated. Venous blood concentration-time profiles for the 2.8 and 7.5
`mg/kg oral doses were also well captured by the PBPK model. The
`model yielded a liver intrinsic clearance (CLint) value based upon free
`FTY720 concentrations in blood of 23,145 l/h/kg. The corresponding
`systemic clearance value, according to the well stirred model, was
`⫽ 0.000333). After oral administration, the first-
`0.748 l/h/kg (fub
`
`

`

`FTY720 PBPK MODEL IN RAT, DOG AND HUMAN
`
`1483
`
`FIG. 2. Time course of FTY720 concentrations in rat venous blood after the indicated i.v. and oral doses. Symbols are experimental values and lines are PBPK model
`predictions.
`
`FIG. 3. Time course of measured (symbols) and predicted (lines) FTY720 concentrations in rat arterial blood and lymph nodes.
`
`order absorption rate constant was 0.052 h⫺1 and fraction of drug
`absorbed was 0.45.
`Well Stirred Organs. Concentration versus time profiles for heart,
`lungs, liver, kidneys, muscle, fat, and spleen captured by the PBPK
`model are depicted in Figs. 4 to 7. Concentration-time curves of these
`organs declined in parallel with that of venous and arterial blood.
`Tissue-to-blood partition coefficients (RT) calculated by noncompart-
`mental analysis and those obtained with the PBPK model are reported
`in Tables 2 and 3. Model-defined RT values were obtained with
`relatively good precision, with CV% on parameters generally ⬍20%.
`In most organs, the deviation between the noncompartmental and
`PBPK-defined RT values never exceeded 3-fold. Among these well
`stirred organs, FTY720 distributed most extensively into lungs and
`liver. The extent of distribution into fat and skeletal muscle was
`moderate.
`Permeability-Limited Organs. The concentration-time curves for
`
`FTY720 in lymph nodes, brain, and thymus are shown in Figs. 3, 4,
`and 6. The model captured the observed data fairly well, with the
`exception of thymus, for which an overprediction of tissue concen-
`tration levels was noticed. Nevertheless, the model still captured the
`trend in the data as well as the terminal slope of the drug in that organ.
`To develop the organ model for lymph nodes, it was assumed that
`fractional volumes of vascular and interstitial spaces were identical to
`those of thymus because the two organs belong to the immune system.
`The PBPK model captured fairly well the features of the data. Tissue-
`to-blood partition coefficient values were 22.9 for lymph nodes, 27.1
`for brain, and 15.7 for thymus. The corresponding PST values were
`176, 39.3, and 122 ml/min (Table 3).
`Other Organs. Organs that were not sampled, such as bone and
`skin, were lumped into a single compartment labeled “rest of body”.
`A nonsampled splanchnic compartment comprising stomach and in-
`testinal tract was also incorporated to account for the flow of blood
`
`

`

`1484
`
`MENO-TETANG ET AL.
`
`FIG. 4. Time course of measured (symbols) and predicted (lines) FTY720 concen-
`trations in rat heart, lungs, and brain after i.v. and oral administration.
`
`FIG. 5. Time course of measured (symbols) and predicted (lines) FTY720 concen-
`trations in rat liver, kidneys, and muscle after i.v. and oral administration.
`
`through the liver. The model-predicted RT values were 50.9 for the
`rest-of-body and 11.1 for the splanchnic compartment. Being unob-
`served, these tissue-to-blood partition coefficients may not reflect the
`real extent of FTY720 distribution in these organs.
`Prediction of FTY720 Kinetics in Dogs and Humans. Figure 8
`depicts FTY720 blood concentration-time profile in the dog after
`i.v. (Fig. 8, top) and oral (Fig. 8, middle) administration. The
`PBPK model was able to capture the experimental data reasonably
`well. Despite an overestimation of Cmax after oral administration,
`the model described the terminal slope with good accuracy.
`FTY720 concentrations in dog brain were simulated for a single
`oral dose of 10 mg/kg. We used the appropriate dog physiology
`and the extrapolated organ distribution parameters from the whole
`body PBPK model. As shown in Fig. 8 (bottom), the predictions
`were slightly lower than the observed values, by just under 1.5-
`fold, but the model predicted accurately the slope of decay of
`FTY720.
`Figure 9a shows the FTY720 concentration-time profile in human
`blood after a single oral dose of 1 mg, as predicted from the rat PBPK
`model with the in vitro intrinsic clearance determined from human
`liver microsomes. Although the prediction was not perfect, it was
`within interindividual variability. It was noted that the predicted Cmax
`value for FTY720 was different from the observed values. Figure 9b
`shows the FTY720 concentrations in human blood obtained after
`optimizing the intrinsic clearance to fit the human data. A great
`improvement was observed and the observed concentration data were
`better described by the PBPK model.
`
`Discussion
`The present analysis characterizes the concentration-time profiles
`for FTY720 in venous and arterial blood and in an extensive array of
`organs after i.v. and oral administration to rats. Gaining insight into
`the pharmacokinetic behavior of a drug in different organs is highly
`desirable, especially in therapeutic target organs or those for which the
`compound may be potentially harmful. We were able to analyze
`FTY720 concentration versus time profiles in the typical major or-
`gans, but also in other rather unusual ones, viz. lymph nodes, spleen,
`and thymus. FTY720 is an immunomodulator that exerts its pharma-
`cological action by sequestering lymphocytes in secondary immune
`organs. Thus, being able to characterize the behavior of the drug in
`pharmacological targets such as spleen and lymph nodes constitutes a
`unique achievement.
`There are several advantages of developing PBPK models. Two
`obvious advantages are the possibility to investigate, in animals,
`organs that could otherwise never be assessed in humans, and the
`possibility to extrapolate the model to higher species by adjusting
`organ volumes and perfusion rates, and adjusting for differences in
`metabolism, plasma protein binding, and blood cell distribution be-
`tween species.
`The results of the investigation showed that FTY720 distributed
`extensively into various organs. More importantly, distribution and
`clearance appeared to be linear in the dose range of 0.3 to 4 mg/kg.
`The model-predicted RT values were similar to those calculated by
`noncompartmental analysis. On average, a 3-fold deviation was ob-
`served between the two analyses (Table 2 versus Table 3).
`
`

`

`FTY720 PBPK MODEL IN RAT, DOG AND HUMAN
`
`1485
`
`FIG. 6. Time course of measured (symbols) and predicted (lines) FTY720 concen-
`trations in rat fat and thymus after i.v. and oral administration.
`
`Brain, lymph nodes, and thymus concentration-time data could be
`described by the permeability-limited model only. Although this
`model seems natural for brain because of the blood-brain barrier to
`xenobiotics, the applicability to lymph nodes and thymus is less clear.
`To our knowledge, specific cell membrane resistance to drug transport
`into thymus or lymph nodes has not been documented. The PST values
`for lymph nodes, brain, and thymus translate to transfer clearance (i.e.,
`product of PST and fub) values of 0.058, 0.0131, and 0.048 ml/min,
`respectively. These transfer clearances are all smaller than the corre-
`sponding organ blood flows (Table 1). Thus, modeling FTY720
`kinetics in these tissues using a permeability-limited model was
`warranted.
`There were some discrepancies between tissue binding parameters
`obtained with local and whole body models. Several reasons may
`explain these differences. First, each organ in the body is, in fact, in
`equilibrium with the blood leaving the organ, rather than with the
`measured venous blood as assumed in the local models. This discrep-
`ancy is even more pronounced for the lungs, which are in equilibrium
`with the arterial blood. Second, in the determination of local organ
`parameters, we used observed venous blood concentration, whereas
`the whole body model takes into account the model-predicted blood
`concentration. As can be seen in Fig. 2, the predicted venous blood
`concentrations after the bolus administration are higher than the
`observed data, especially for the lower dose of 0.3 mg/kg.
`The volume of distribution at steady state estimated with eq. 3
`yielded a value of 8.61 l/kg. This figure is close to the one obtained
`by a standard compartmental analysis (13.7 l/kg, Table 4). The intrin-
`sic clearance estimated by the current model was 23,145 l/h/kg. This
`value seems very large. However, because FTY720 is highly bound to
`
`FIG. 7. Time course of measured (symbols) and predicted (lines) FTY720 concen-
`trations in rat spleen after i.v. and oral administration.
`
`TABLE 2
`Tissue-to-blood partition coefficient (RT) of FTY720 calculated from AUC ratios
`
`Organ
`
`Lungs
`Heart
`Brain
`Kidneys
`Thymus
`Spleen
`Liver
`Muscle
`Lymph nodesa
`
`Intravenous Doses
`
`0.3 mg/kg
`
`2 mg/kg
`
`89.9
`27.7
`53.4
`53.7
`19.6
`67.4
`83.0
`9.4
`28.6
`
`68.2
`17.4
`49.2
`35.8
`19.6
`62.1
`47.0
`10.5
`32.6
`
`a Pool of mesenteric, axillary, inguinal, cervical, and popliteal lymph nodes.
`
`plasma proteins (Novartis Pharma AG, internal communication) and
`distributes extensively into red blood cells, this intrinsic clearance
`corresponded to a systemic clearance value of 0.748 l/h/kg. This value
`was not too different from the average systemic clearance obtained by
`compartmental analysis (0.497 l/h/kg, Table 4). Due to strong binding
`to blood components, extensive distribution, and a relatively low
`systemic clearance, FTY720 has a half-life of about 20 h in rats
`(Table 4).
`In the development of the PBPK model, it was assumed that
`FTY720 is eliminated from the body by metabolic biotransformation
`occurring mostly in the liver. This assumption was based on the fact
`that no parent drug could be detected in urine samples. OneOne comp
`compli-li-
`
`compoundound underundergoesgoes revers
`
`
`
`ation inin FTY720FTY720 metabolismmetabolism isis thatthat thethe comp
`revers--
`ccation
`ibliblee metabmetabolismolism (Novart
`
`
`ication). In-In-arma AG,AG, internainternall commun
`(Novartisis PhPharma
`communication).
`
`
`
`
`terconversionion bbetweenetween FTY720FTY720 ananddiitsts phosphorylated metametabolitebolite
`terconvers
`phosphorylated
`
`
`
`FTY720-P) iiss catacatalyzedlyzed bbyytytypep 1e 1aandnd 22sphingosine kikinases.nases. ThThese
`((FTY720-P)
`sphingosine
`
`

`

`1486
`
`MENO-TETANG ET AL.
`
`TABLE 3
`Physiologic pharmacokinetic parameters for FTY720 in rats, dogs, and humans using the whole body PBPK model
`Parameters for dogs and humans were derived from those of rats using their respective drug free fractions in blood.
`
`Species
`
`Parameters
`
`BW (kg)
`fub
`CLint (␮l/min/mg protein)-microsomes
`a (l/h/kg)
`CLint
`Fabs
`kabs (h⫺1)
`Rlungs
`Rbrain
`Rheart
`Rspleen
`Rmuscle
`Rthymus
`Rkidneys
`RLN
`Rliver
`Rfat
`RGIT
`Rrest-of-body
`PSbrain (ml/min)
`PSthymus (ml/min)
`PSLN (ml/min)
`
`b
`
`Rat
`
`0.362
`0.000333
`88
`2315
`0.71
`0.0007
`41.4
`27.1
`13.8
`34.7
`4.69
`15.8
`22.3
`22.9
`34.9
`0.647
`11.1
`50.9
`39.3
`122
`176
`
`CV%
`
`(12.2)
`(18.5)
`(19.3)
`(12.5)
`(11.9)
`(15.0)
`(10.5)
`(14.1)
`(11.72)
`(9.3)
`(12.9)
`(14.6)
`(9.84)
`(54.4)
`(8.3)
`(12.1)
`(11.3)
`(16.9)
`
`Dog
`
`11.65
`0.000241
`N.A.
`1018
`0.6
`0.00843
`29.9
`19.5
`9.96
`25.0
`3.39
`11.4
`16.1
`16.5
`25.2
`0.47
`8.01
`36.8
`379
`572
`820
`
`Human
`
`70
`0.000361
`33
`202
`0.5
`0.000743
`44.8
`29.3
`14.9
`37.5
`5.08
`17.1
`24.1
`24.8
`37.8
`0.70
`12.0
`55.1
`3613
`1517
`2173
`
`N.A., not applicable.
`a Based on free fraction in blood.
`b Value for liver here is equal to Rli 䡠 (1 ⫺ CLH,int/Qhp) rather than Rli.
`
`FIG. 8. Time course of measured (symbols) and predicted (lines) FTY720 concen-
`trations in blood of dogs after oral (top) or i.v. (middle) dosing. The bottom panel
`shows predictions of brain concentrations.
`
`FIG. 9. Time course of measured (symbols) and predicted (lines) FTY720 concentra-
`tions in blood of humans. a represents predictions using in vitro microsomal CLint, and
`b represents predictions from whole body PBPK after optimization of CLint.
`
`

`

`FTY720 PBPK MODEL IN RAT, DOG AND HUMAN
`
`1487
`
`TABLE 4
`Pharmacokinetic parameters for FTY720 in blood after single i.v. doses in rats
`calculated by compartmental analysis
`
`Parameter
`
`t1/2 (h)
`CL (l/h/kg)
`Vss (l/kg)
`
`Dose
`
`Mean
`
`S.D.
`
`0.3 mg/kg
`
`2 mg/kg
`
`4 mg/kg
`
`18.5
`0.341
`5.61
`
`30.7
`0.462
`16.2
`
`20.9
`0.687
`19.4
`
`23.4
`0.497
`13.7
`
`6.43
`0.176
`7.22
`
`
`enzymes are found in numerous tissues including liver, eye, lung, andenzymes are found in numerous tissues including liver, eye, lung, and
`
`muscle. The characterization of reversible systems can be achievedmuscle.
`only after collecting concentration-time data of parent and metabolite
`after separate i.v. administration of parent and metabolite (Cheng and
`Jusko, 1993). No metabolite concentration data were measured in the
`Therefore, the present PBPK model may only be an
`present study. Therefore, the present PBPK model may only be an
`approximation of FTY720 kinetics in blood and organs because
`approximation of FTY720 kinetics in blood and organs because
`reversible metabolic processes were not taken into account.
`reversible metabolic processes were not taken into account. Interest in
`FTY720-P is high; it is the pharmacologically active moiety. How-
`ever, with an equilibrium between FTY720 and FTY720-P, RT and
`PST will be apparent parameters incorporating both partition-diffusion
`to and from tissues, together with reversible metabolic interconver-
`sion. The rapidity of the achievement of an equilibrium between FTY
`and FTY-P may explain why a model of the parent works well for
`characterizing the pharmacodynamics of lymphocyte trafficking in
`monkeys (Li et al., 2002).
`The PBPK model was able to mimic blood concentration-time
`profiles in dogs after oral and i.v. administration when optimizing
`only the intrinsic clearance and maintaining the tissue binding param-
`eters from the rat. In addition, we were able to test the model
`prediction of brain concentrations in dogs. Although the predictions
`were slightly lower than the observed values, the model predicted
`accurately the slope of decay of FTY720 in dog brain. This may allow
`the determinat

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket