throbber
0022-3565/07/3232-469–476$20.00
`THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS
`Copyright © 2007 by The American Society for Pharmacology and Experimental Therapeutics
`JPET 323:469–476, 2007
`
`Vol. 323, No. 2
`127183/3263802
`Printed in U S.A.
`
`Brain Penetration of the Oral Immunomodulatory Drug FTY720
`and Its Phosphorylation in the Central Nervous System during
`Experimental Autoimmune Encephalomyelitis: Consequences
`for Mode of Action in Multiple Sclerosis
`
`Carolyn A. Foster, Laurence M. Howard, Alain Schweitzer, Elke Persohn,
`Peter C. Hiestand, Bala´ zs Balatoni,1 Roland Reuschel, Christian Beerli, Manuela Schwartz,2
`and Andreas Billich
`Novartis Institutes for BioMedical Research, Vienna, Austria (C.A.F., L.M.H., B.B., R.R., M.S., A.B.); Novartis Institutes for
`BioMedical Research, Basel, Switzerland (P.C.H., C.B.); Novartis Pharma AG, Basel, Switzerland (A.S.); and Novartis Pharma
`AG, Muttenz, Switzerland (E.P.)
`Received June 24, 2007; accepted August 3, 2007
`
`ABSTRACT
`FTY720
`[2-amino-2-[2-(4-octylphenyl)ethyl]propane-1,3-diol
`hydrochloride]
`is an oral sphingosine-1-phosphate receptor
`modulator under development for the treatment of multiple
`sclerosis (MS). The drug is phosphorylated in vivo by sphin-
`gosine kinase 2 to its bioactive form, FTY720-P. Although treat-
`ment with FTY720 is accompanied by a reduction of the pe-
`ripheral lymphocyte count, its efficacy in MS and experimental
`autoimmune encephalomyelitis (EAE) may be due to additional,
`direct effects in the central nervous system (CNS). We now
`show that FTY720 localizes to the CNS white matter, preferen-
`tially along myelin sheaths. Brain trough levels of FTY720 and
`
`FTY720-P in rat EAE are of the same magnitude and dose
`dependently increase; they are in the range of 40 to 540 ng/g in
`the brain tissue at efficacious doses and exceed blood con-
`centrations severalfold. In a rat model of chronic EAE, pro-
`longed treatment with 0.03 mg/kg was efficacious, but limiting
`the dosing period failed to prevent EAE despite a significant
`decrease in blood lymphocytes. FTY720 effectiveness is likely
`due to a culmination of mechanisms involving reduction of
`autoreactive T cells, neuroprotective influence of FTY720-P
`in the CNS, and inhibition of inflammatory mediators in the
`brain.
`
`FTY720 is an oral sphingosine-1-phosphate (S1P) receptor
`modulator (Baumruker et al., 2007) under development for
`the treatment of multiple sclerosis (MS), representing the
`first of a new class of immunomodulatory agents. Promising
`results in phase II trials with relapsing MS patients (Kappos
`et al., 2006) mirror the striking efficacy of FTY720 in MS
`models of experimental autoimmune encephalomyelitis
`(EAE), shown by preventive and therapeutic treatment
`(Brinkmann et al., 2002; Fujino et al., 2003; Webb et al.,
`2004; Kataoka et al., 2005; Balatoni et al., 2007). FTY720 is
`converted in vivo to its biologically active phosphate ester
`
`This work was supported by Novartis Pharma AG.
`1 Current affiliation: Novartis Hungary Healthcare, Budapest, Hungary.
`2 Current affiliation: Evangelisches Krankenhaus, Vienna, Austria.
`Article, publication date, and citation information can be found at
`http://jpet.aspetjournals.org.
`doi:10.1124/jpet.107.127183.
`
`metabolite (FTY720-P), which acts as a high-affinity agonist
`for four of the five known G-protein-coupled S1P receptors,
`namely S1P1 and S1P3–5 (Brinkmann et al., 2002; Mandala
`et al., 2002). Sphingosine kinase (SPHK) 2 is the primary
`enzyme required for FTY720-P formation, as we and others
`subsequently confirmed in SPHK2 knockout mice (Kharel et
`al., 2005; Zemann et al., 2006). The fact that SPHK1 null
`mice become lymphopenic after FTY720 administration fur-
`ther supports the view that SPHK2 is sufficient for the func-
`tional activation of FTY720 (Allende et al., 2004).
`Emerging evidence suggests that the effectiveness of
`FTY720 in the central nervous system (CNS) extends beyond
`immunomodulation to encompass other aspects of MS patho-
`physiology, including an influence on the blood-brain barrier
`and glial repair mechanisms that could ultimately contribute
`to restoration of nerve function (Baumruker et al., 2007;
`
`ABBREVIATIONS: FTY720, fingolimod, 2-amino-2-[2-(4-octylphenyl)ethyl]propane-1,3-diol hydrochloride; MS, multiple sclerosis; EAE, experi-
`mental autoimmune encephalomyelitis; SPHK, sphingosine kinase; CNS, central nervous system; DA, Dark Agouti; CFA, complete Freund’s
`adjuvant; CsA, cyclosporine A; QWBA, quantitative whole-body autoradiography; CSF, cerebrospinal fluid; ANOVA, analysis of variance; AUC,
`area under the curve; FTY720-P, 2-amino-2-[2-(4-octylphenyl)ethyl)propane-1,3-diol-1-(dihydrogen phosphate); FK506, tacrolimus.
`
`469
`
`Apotex v. Novartis
`IPR2017-00854
`NOVARTIS 2038
`
`

`

`470
`
`Foster et al.
`
`Jung et al., 2007; Osinde et al., 2007). A key consideration
`behind this concept is the finding that FTY720 distributes to
`the brain (Meno-Tetang et al., 2006), which contains endog-
`enous SPHK2 for the phosphorylation of FTY720 (Billich et
`al., 2003). Moreover, neurons and glial cells (astrocytes, mi-
`croglia, oligodendrocytes) in the brain differentially express
`S1P receptors (Fig. 1), thus raising the possibility for recep-
`tor activation in situ by FTY720-P. So far, there is no infor-
`mation on the presence of FTY720-P in the brain or potential
`concentrations therein. Our primary aim was to investigate
`the distribution of FTY720 and its phosphorylated form in
`the CNS after clinically relevant doses in two different EAE
`models. We provide preclinical evidence that the bioactive
`metabolite FTY720-P distributes to the CNS white matter,
`suggesting the potential for functional interaction with glial
`cells bearing S1P receptors in the brain and spinal cord.
`
`Materials and Methods
`Animals. For EAE, female RT11 Lewis rats from Charles River
`(Sulzfeld, Germany) and Dark Agouti (DA) rats from Harlan Winkel-
`mann (Borchen, Germany) were kept under standardized light- and
`climate-controlled conditions with free access to food and water.
`Age-matched rats were acclimatized for at least 1 week before dis-
`tribution into the experimental groups. For autoradiography, male
`pigmented LE/CR WIGA rats (Charles River; 198–238 g) were
`housed individually in metabolism cages. All experiments conformed
`to Novartis animal care regulations and were approved by the Aus-
`trian and Swiss health authorities in compliance with international
`animal welfare standards according to the European Communities
`Council Directive and the guidelines set forth in the National Insti-
`tutes of Health Guide for the Care and Use of Laboratory Animals
`(Institute of Laboratory Animal Resources, 1996).
`EAE Induction and Clinical Scoring. Animals were lightly
`anesthetized by isoflurane inhalation (0.5% Forane; Abbott Labora-
`tories, Vienna, Austria) and given a single intradermal 200-␮l inoc-
`ulation in the dorsal base of the tail root. The immunization mixture
`for DA rats consisted of syngeneic CNS antigen (4 parts brain to 6
`parts of spinal cord) in phosphate-buffered saline emulsified 1:1 in
`incomplete Freund’s adjuvant supplemented with 200 ␮g of heat-
`inactivated Mycobacterium tuberculosis (strain H37 RA; DIFCO, BD
`Diagnostics, Oxford, UK); the adjuvant is henceforth referred to as
`complete Freund’s adjuvant (CFA). For Lewis rats, the inoculum
`contained guinea pig spinal cord in phosphate-buffered saline emul-
`sified 1:1 in CFA. As an adjuvant control for all EAE studies, animals
`were injected with CFA alone and vehicle-treated. The rats were
`weighed every other day and scored daily for neurological signs as
`
`follows: 0, no clinical deficit; 1, complete loss of tail tonus; 2, limb
`weakness or ataxia; 3, full paralysis of hind or forelimbs; or 4,
`tetraparalysis or moribund. Animals with a score of 4 were sacrificed
`if weight loss indicated little chance of recovery, in accordance with
`animal welfare standards. Mortality due to sacrifice or spontaneous
`EAE-related death was indicated (†) and recorded as a 4 on thegiven
`day; this death score continued to be included in the clinical assess-
`ment, but body weight measurements were not carried forward.
`Test Compounds for in Vivo Evaluation. Unlabeled and 14C-
`labeled FTY720, as well as cyclosporine A (CsA), were supplied by
`Novartis Pharma AG (Basel, Switzerland). The radiochemical purity
`of [14C]FTY720, which was labeled in position 2, was shown by
`high-pressure liquid chromatography to be 98% with a specific ac-
`tivity of 35 ␮Ci/mg. FTY720 was dissolved in water, and CsA was
`dosed in the Neoral vehicle. Both drugs were freshly prepared and
`given p.o. once daily by gavage at a dosing volume of 5 ml/kg body
`weight. For prophylactic and therapeutic treatment, oral dosing
`started on day 0 at immunization and at the peak of disease in fully
`established EAE, respectively.
`Peripheral Leukocyte Counts. Rats were lightly anesthetized
`by isoflurane inhalation and 100 ␮l of blood from the retro-orbital
`venous plexus was collected in EDTA-coated tubes (Sarstedt AG,
`Nu¨ mbrecht, Germany). Automated differential leukocyte analysis
`was performed on the HESKA Vet ABC-Diff Hematology Analyzer
`(Heska Corp., Fort Collins, CO).
`Autoradiography of 14C-Labeled FTY720. Quantitative whole-
`body autoradiography (QWBA) and light microscopic autoradiogra-
`phy were performed to assess the uptake and tissue distribution of
`[14C]FTY720 radioactivity in male pigmented rats (n ⫽ 6) following
`seven oral doses at 7.5 mg/kg/d. At 8, 24, and 168 h after the last
`dose, the animals were deeply anesthetized with isoflurane and
`submerged in a dry ice-hexane bath at ⫺70°C for at least 20 min. The
`frozen carcasses were rapidly shaven and stored below ⫺20°C until
`embedment in an ice-cold aqueous solution of 2% carboxymethylcel-
`lulose. They were then frozen for approximately 30 min in a dry
`ice-hexane mixture at ⫺70°C, followed by an overnight stabilization
`at ⫺20°C. Lengthwise sections (40 ␮m thick) were obtained in a
`cryomicrotome (Leica Microsystems, Nussloch, Germany) at ⫺20°C.
`Whole-body autoradiograms were obtained by autoradioluminogra-
`phy. Briefly, sections with a paper backing were placed on Fuji
`BASIII imaging plates (Fuji Photo Film, Tokyo, Japan) for 1 day at
`room temperature in a lead shielding box. After exposure (detection
`of approximately 1.5 dpm/mg), the imaging plates were first kept in
`the dark for 3 to 5 min and then transferred to a Fuji BAS 2000 TR
`phosphorimaging device (Fuji Photo Film) for scanning at a 100-␮m
`step with a 1024 gradation. Images were prepared by re-exposing the
`sections
`onto Super Resolution storage
`phosphor
`screens
`(PerkinElmer, Shelton, CT) for 1 day at room temperature and
`
`Fig. 1. Cartoon depicting hierarchy of S1P receptor
`expression on rat glial subpopulations (Rao et al., 2003;
`Tham et al., 2003; Toman et al., 2004; Yu et al., 2004):
`implications for FTY720-P-mediated repair in the CNS.
`bFGF, basic fibroblast growth factor; CSF, colony-stim-
`ulating factor; GDNF, glial-derived neurotrophic factor;
`IFN␥, interferon ␥; IL1, interleukin-1; IL12, interleu-
`kin-12; MA, macrophage/microglia; NGF, nerve growth
`factor; NO, nitric oxide; TNF␣, tumor necrosis factor ␣.
`
`

`

`scanning at a 42-␮m step (Cyclone PhosphorImager; Packard Instru-
`ment, Meriden, CT). The image files were processed using Photoshop
`Elements 2.0 software (Adobe Systems, San Jose, CA). Levels of
`radioactivity in the tissues were determined by comparative densi-
`tometry, as described previously (Schweitzer et al., 1987).
`For light microscopy, brain and spinal cord samples from all
`animals were fixed in 3% glutaraldehyde in 0.1 M cacodylate buffer,
`pH 7.4, for 2 days at 4°C. Postfixation was performed with 1%
`osmium tetroxide in 0.1 M cacodylate buffer, pH 7.4, for 2 h at4°C.
`The tissues were dehydrated in graded acetone solutions and em-
`bedded in Epon. Semithin sections were dipped in Ilford L4 emulsion
`(Ilford, Mobberley, Cheshire, UK) in the dark at 4°C. Dipped sections
`were developed in Kodak D19 (Eastman Kodak, Rochester, NY) after
`31 weeks exposure, stopped in distilled water, fixed in Ilford Hypam
`rapid fixer (Ilford), and counterstained with toluidine blue. Light
`microscopic examination of sections was performed independently by
`three pathologists. Labeling was identified by more silver grains over
`the cells than the background without tissue.
`Quantification of FTY720 and FTY720-P in the Blood,
`Brain, and Cerebrospinal Fluid. Plasma, whole blood, brain, and
`cerebrospinal fluid (CSF) were collected from EAE rats at 24 h after
`the last FTY720 dose to obtain trough levels. Concentrations of
`FTY720 and FTY720-P were determined by high-pressure liquid
`chromatography (Agilent 1100; Agilent, Waldbronn, Germany) with
`mass spectrometric detection as described previously for serum and
`other tissues (Zemann et al., 2006). For measurements in plasma,
`heparinized blood, or CSF, 20- to 100-␮l aliquots were spiked with
`internal standards (final concentration, 0.5 ␮g/ml) and extracted
`with chloroform/methanol at acidic pH; extracts were dried and
`reconstituted in methanol/0.2% formic acid. Samples were chromato-
`graphed on a Luna C8 column (3 ␮, 2⫻ 50 mm; Phenomenex,
`Torrence, CA) equipped with a C4 wide-bore precolumn. The ana-
`lytes were eluted with a gradient (eluent A, 10 mM ammonium
`acetate containing 0.08% HCOOH in water; eluent B, 10 mM ammo-
`nium acetate containing 0.08% HCOOH in MeOH; 50–98% B in 14
`min) at a flow of 0.4 ml/min at 40°C. Analytes were detected by
`electrospray-ionization liquid chromatography with tandem mass
`spectroscopy using an API 4000 QTrap instrument (MDS Sciex,
`Concord, ON, Canada). The optimal collision energies for FTY720
`and FTY720-P were 23 and 25 V, respectively. The multiple reaction
`monitoring transitions monitored for FTY720 and FTY720-P were
`m/z 308/255 and 388/255, respectively.
`For CNS determination, half of a rat brain (approximately 750 mg)
`was emulsified 1:1 in 2 ml of MeOH/H2O (1:1) with a glass homog-
`enizer (Potter S; Braun, Melsungen, Germany). The homogenate was
`divided for separate determination of FTY720 and FTY720-P, then
`aliquots were spiked with their respective internal standards (35 ␮l)
`to give a final concentration of 0.5 ␮g/ml. Methanol (400 ␮l) was
`added to 0.5 ml of homogenate, followed by 20 min of mixing on a
`
`TABLE 1
`FTY720 therapeutic treatment effect in DA rat model of EAEa
`
`FTY720 Exposure in CNS
`
`471
`
`rotary shaker and 5 min of sonication at room temperature. Super-
`natants were harvested after centrifugation at 12,000g; the pellet
`was extracted once with 200 ␮l of methanol, then the supernatant
`was harvested after centrifugation. Combined supernatants were
`subjected to solid-phase extraction using STRATA-X-C 33 ␮m Cation
`Mixed-Mode Polymer Phase columns (Phenomenex). The columns
`were eluted with 3 ml of MeOH/1% NH4OH. The eluate was dried, its
`residue was dissolved in a 35:65 mixture of eluents A and B (see
`above), and then samples were chromatographed as described above.
`An alternative method for blood or homogenized brain comprised
`two extractions with acetonitrile/methanol/ethyl acetate (5:3:2), dis-
`solution of the residue of the evaporated organic phase in methanol/
`water/ammonia (50:48.75:1.25), and chromatography on a Zorbax
`SB-C8 0.5- ⫻ 75-mm column (3.5 ␮m; Agilent Technologies) at 50°C
`with a linear gradient from 0 to 95% B within 10 min at 20 ␮l/min
`(solvent A, 0.2% HCOOH in water; B, 0.2% HCOOH in acetonitrile).
`In this case, a Micromass Micro triple quadrupole mass spectrometer
`with electrospray source (Waters AG, Rupperswil, Switzerland) was
`used, and multiple reaction monitoring transitions were monitored
`for FTY720 and FTY720-P at m/z 308/105 and 388/255, respectively.
`Both analytical methods yielded identical results.
`Statistical Analysis. A one-way analysis of variance (ANOVA)
`was used to compare all data sets using SigmaStat for Windows,
`version 3.11 (Systat Software Inc., Richmond, CA). Differences be-
`tween groups were analyzed using the post hoc Tukey test for pair-
`wise multiple comparison. For EAE, area under the curve (AUC)
`values for body weight loss and clinical grade scores were evaluated
`during the entire prophylactic treatment period or after the initia-
`tion of therapeutic dosing. Probabilities (p) ⱕ 0.05 were considered to
`be statistically significant.
`
`Results
`FTY720 Provides Sustained Protection in EAE. Two-
`week therapeutic treatment with 0.03 to 0.9 mg/kg FTY720
`dose dependently inhibited progression of established dis-
`ease in the DA rat model of chronic EAE compared with
`vehicle controls, which exhibited sustained neurological def-
`icits throughout the 2-month observation (Table 1; Fig. 2).
`Evidence for rapid and full disease suppression was consis-
`tently observed after administration of 0.3 mg/kg FTY720,
`whereas the minimum effective dose providing almost com-
`plete protection even 1 month after discontinuation was 0.1
`mg/kg (p ⫽ 0.00002). A plateau in efficacy appeared to be
`reached by 0.3 mg/kg since there was no difference in the
`cumulative disease score between this dose and the 3-fold
`higher one of 0.9 mg/kg (Table 1). The very low dose of 0.03
`
`Treatment Groups: Oral Dose:
`from Days 12 to 25
`
`mg/kg
`Vehicle (n ⫽ 17)
`FTY720, 0.03 mg (n ⫽ 8)
`FTY720, 0.1 mg (n ⫽ 19)
`FTY720, 0.3 mg (n ⫽ 19)
`FTY720, 0.9 mg (n ⫽ 9)
`CsA, 25 mg (n ⫽ 18)
`
`EAE
`Onsetb
`
`Day
`
`8.4 ⫾ 0.4
`8.8 ⫾ 0.3
`8.4 ⫾ 0.3
`9.0 ⫾ 0.3
`8.3 ⫾ 0.2
`9.0 ⫾ 0.3
`
`Maximum Weight Lossc
`
`Maximum Diseased
`
`Cumulative Disease Score from
`Days 13 to 56e
`
`%
`
`Day
`
`Score
`
`Day
`
`AUC
`
`p vs. Vehicle
`
`23.6 ⫾ 0.02
`16.4 ⫾ 0.03
`12.0 ⫾ 0.02
`9.6 ⫾ 0.01
`15.4 ⫾ 0.02
`16.9 ⫾ 0.02
`
`20.7 ⫾ 0.7
`21.9 ⫾ 1.0
`20.4 ⫾ 0.2
`20.2 ⫾ 0.2
`20.2 ⫾ 0.1
`28.4 ⫾ 2.4
`
`2.9 ⫾ 0.2
`2.3 ⫾ 0.4
`1.2 ⫾ 0.3
`0.3 ⫾ 0.1
`0.6 ⫾ 0.4
`3.3 ⫾ 0.1
`
`22.6 ⫾ 1.5
`28.5 ⫾ 2.6
`24.0 ⫾ 1.9
`24.2 ⫾ 2.0
`25.7 ⫾ 3.7
`33.9 ⫾ 0.6
`
`79.0 ⫾ 12.6
`64.0 ⫾ 14.7
`17.0 ⫾ 3.7
`7.8 ⫾ 2.5
`7.7 ⫾ 3.3
`52.8 ⫾ 6.7
`
`N.S.
`0.00002
`⬍0.00001
`0.0005
`N.S.
`
`n, number of rats per group; N.S., not significant.
`a Syngeneic antigen-induced EAE (Fig. 2); data shown as mean ⫾ S.E.M.
`b Based on disease score ⱖ 1.
`c Highest weight loss starting 1 week after dosing (days 20–56) compared with adjuvant control.
`d Initial peak of disease severity starting 1 week after dosing (days 20–56).
`e Level of significance (p) determined by ANOVA of AUC values for clinical scores from days 13 to 56 compared with the positive control.
`
`

`

`472
`
`Foster et al.
`
`Fig. 2. Sustained efficacy of FTY720 after cessation in DA rat model of
`EAE. Representative disease course in syngeneic antigen-induced EAE in
`DA rats, depicted by the vehicle control (f; n ⫽ 17) and shown as mean
`score ⫾ S.E. Oral therapeutic dosing started at the peak of established
`disease on day 12 and continued for 2 weeks. CsA at 25 mg/kg (䡺; n ⫽ 18)
`suppressed EAE signs during treatment, but animals became severely
`paralyzed, and 22% died (†) upon drug discontinuation. In sharp contrast,
`FTY720 at 0.1 (E; n ⫽ 19) and 0.3 mg/kg (F; n ⫽ 19) significantly
`prevented wasting and recurrence of neurological deficits, as detailed in
`Table 1. Although 0.03 mg/kg FTY720 (ⴱ; n ⫽ 8) tended to reduce the
`disease burden, it was not statistically different to the vehicle (Table 1).
`Nevertheless, this very low dose as well as the other FTY720 treatments
`completely protected against EAE-related deaths, compared with three in
`the vehicle (days 15, 30, and 42) and four in CsA-treated animals (days
`34, 35, 40, and 41).
`
`mg/kg also tended to diminish the overall disease burden and
`prevent a marked rebound. In contrast, severe paralysis re-
`occurs following cessation of classic immunosuppressive
`agents, such as CsA at 25 mg/kg (Fig. 2) or FK506 at 4 mg/kg
`(data not shown), that were used as reference compounds. All
`doses of FTY720 completely prevented moribund incidence
`compared with 18 and 22% with vehicle and CsA, respec-
`tively (Fig. 2).
`Prolongation of Low-Dose FTY720 Steadily Reduces
`EAE Signs after Therapeutic Treatment. To further ex-
`plore the long-term efficacy of low-dose FTY720 in a more
`clinically relevant setting, 0.03 to 0.3 mg/kg was adminis-
`tered for 3 weeks in DA rats with established EAE (Fig. 3).
`By the last week of therapeutic treatment, three different
`studies consistently demonstrated a significant decrease in
`disease signs with 0.03 mg/kg versus the vehicle. Higher
`doses of 0.1 mg/kg (data not shown) and 0.3 mg/kg were even
`more efficacious. Moreover, the mortality rate was markedly
`reduced with FTY720 at 0.03 mg/kg (8.9% death), with com-
`plete protection at 0.1 and 0.3 mg/kg compared with 29.2%
`deaths in the positive control.
`In contrast, 2-week prophylactic treatment with 0.03
`mg/kg FTY720 in the Lewis rat model of acute EAE merely
`delayed the onset of paralysis by approximately 1 day but
`had no protective effect on disease development (Fig. 4).
`Increasing the 2-week dose to 0.3 mg/kg markedly prevented
`neurological deficits during the 1-month study (p ⫽ 0.0003);
`however, restricting the treatment to days 0 to 6 failed to
`stop EAE induction, with clinical signs appearing 1 week
`after drug cessation.
`Influence of FTY720 on Circulating Lymphocyte
`Counts during EAE. Given that the above disparities in
`
`Fig. 3. Prolonged therapeutic treatment with low-dose FTY720 progres-
`sively reduces EAE burden in DA rats. Change in clinical scores, along
`with EAE-related deaths (†), during the disease course in DA rats immu-
`nized with syngeneic CNS antigens. Data were pooled from three EAE
`studies and expressed as mean ⫾ S.E. Animals received daily treatment
`p.o. with vehicle (f; n ⫽ 48) or FTY720 for 3 weeks, starting from day 11.
`Prolongation of 0.03 mg/kg FTY720 dosing (O, n ⫽ 45) gradually reduced
`the EAE burden and weight loss such that animals no longer exhibited
`ataxia or limb paralysis (p ⫽ ⬍0.001), in contrast with vehicle controls,
`which displayed sustained disease throughout the 5-week observation
`period. As expected, the 0.3 mg/kg dose (F; n ⫽ 33) rapidly suppressed the
`EAE grade to a level near baseline, similar to the adjuvant controls that
`were injected with CFA alone and vehicle-treated (data not shown). Both
`doses of FTY720 profoundly prevented mortalities, except for 4 in the 0.03
`mg/kg group (days 15 and 21), compared with 14 in the vehicle (days 13,
`14, 17, 18, 19, 20, 22, 23, 25, and 26). Level of significance was determined
`by ANOVA of AUC values from days 12 to 33 versus the positive control.
`Blood was collected on days 14 and 33 (Œ) for differential leukocyte
`analysis, as shown in Fig. 5B. ⴱⴱⴱ, p ⬍ 0.001.
`
`Fig. 4. Efficacy of FTY720 prophylaxis in EAE is more dependent on dose
`and duration compared with therapeutic treatment. Clinical scores ⫾
`S.E. in Lewis rat model of EAE (nine animals per group) showing typical
`monophasic disease in the vehicle control (f). Preventive therapy with
`FTY720 was effective when 0.3 mg/kg was given for 2 weeks (F) but not
`when dosing was restricted to the first 7 days (E). A 10-fold lower dose of
`0.03 mg/kg (⽧) failed to prevent disease development. As expected, 25
`mg/kg CsA (ⴱ) was fully protective during treatment, yet paralysis oc-
`curred almost 2 weeks after drug cessation, resulting in a death (†). Blood
`was collected at 0, 6, and 24 h and on 3 subsequent days (Œ) for differ-
`ential leukocyte analysis, as shown in Fig. 5A.
`
`EAE-efficacy appeared to be less related to the FTY720 dose
`but more to its duration, we sought to investigate the tem-
`poral relationship between peripheral lymphocyte counts
`and EAE treatment regimens. Earlier studies showed that
`
`

`

`0.03 and 0.3 mg/kg FTY720 can decrease the peripheral
`lymphocyte count by approximately 20 and 70%, respec-
`tively, within 6 h after a single oral dose in naive Lewis rats
`(Brinkmann et al., 2002), resulting in an ED50 of 0.09 ⫾ 0.01
`mg/kg by 48 h. Likewise during EAE, 0.3 mg/kg FTY720
`already reached maximum reduction (approximately 90%) of
`lymphocytes by 6 h in theLewis rat (Fig. 5A). Furthermore,
`it was already recognized that daily doses of 0.03 mg/kg
`FTY720 for 1 week can reduce lymphocyte counts by up to
`80% compared with placebo-treated Lewis rats in a heart
`allograft model (Nikolova et al., 2000). We have extended
`these findings to EAE and demonstrate that 0.03 mg/kg
`therapeutic dosing for 3 weeks in DA rats leads to a highly
`significant reduction in circulating lymphocytes by days 14
`and 33 versus vehicle (Fig. 5B), i.e., 52 and 67%, respectively.
`At the early time point, 0.3 mg/kg FTY720 led to over twice
`the reduction in lymphocyte numbers compared with 0.03
`mg/kg (p ⬍ 0.001), but by day 33, there was no difference
`between these two doses (Fig. 5B).
`Taking into account that 7-day preventive dosing with 0.3
`mg/kg FTY720 failed to suppress EAE (Fig. 4), yet lympho-
`cytes were reduced by 75% versus vehicle (Fig. 5A), we agree
`with previous suggestions (Webb et al., 2004) that FTY720 or
`its phosphate are apt to exert additional effects beyond the
`induction of peripheral lymphopenia. For example, although
`0.03 mg/kg FTY720 decreased the circulating lymphocytes by
`at least 50% shortly after drug initiation (Fig. 5B), this low
`dose failed to significantly prevent EAE signs when treat-
`ment is limited to 2 weeks in a prophylactic setting (Fig. 4).
`To explore whether additional effects of FTY720 and
`FTY720-P in the brain are possible at all, based on available
`drug concentrations in that tissue, we examined the distri-
`bution of FTY720 in the rat and determined levels of
`FTY720/FTY720-P in the brain.
`Quantitative Whole-Body Autoradiography and My-
`elin Sheath Distribution of [14C]FTY720. First, QWBA
`was used to investigate the distribution of [14C]FTY720-
`related radioactivity in vivo and, in particular, its uptake
`into the CNS. Pigmented rats received [14C]FTY720 for 1
`week at a high oral dose of 7.5 mg/kg/d. By 24 h after the
`seventh and last dose (Fig. 6A), elevated amounts of ex-
`travascular radioactivity were detected in the adrenal cortex,
`kidney (cortex-medullary junction), nasal turbinates, pitu-
`
`FTY720 Exposure in CNS
`
`473
`
`itary gland, preputial gland, and stomach (glandular mu-
`cosa); maximal levels of radioactivity occurred in the brain,
`epididymis, eye (ocular membranes, vitreous body), and tes-
`tis. At 168 h, residual radioactivity was still observed in most
`of these tissues, equivalent to approximately 1.4% of the
`administered dose, but the highest concentrations were
`found in the brain (reticular nucleus, corpus callosum, cere-
`bellar white matter), preputial gland, and spinal cord (Fig.
`6B). The distribution pattern after multiple doses was simi-
`lar to that after a single dose (data not shown), especially the
`preferential localization to brain and spinal cord at 168 h.
`The accumulation factor (i.e., ratio between the tissue con-
`centration after multiple versus single dosing) was 3.2 for
`brain and 3.7 for spinal cord at 24 h after the seventh dose
`compared with the single dose.
`Light microscopic evaluation showed that the 14C labeling
`in brain and spinal cord was confined to the myelin sheets
`(Fig. 7). Neurons were free of grains, except for some back-
`ground labeling.
`Levels of FTY720 and FTY720-P in Blood and Brain.
`Given that exposure of the CNS after oral dosage of FTY720
`to normal rats was shown in the above QWBA study and in a
`previous pharmacokinetic analysis (Meno-Tetang et al.,
`2006), we next sought to determine concentrations of FTY720
`and its phosphorylated form in blood and brain of rats dis-
`eased with EAE. In fact, levels of FTY720-P in brain have so
`far not been reported at all. Samples from both acute and
`chronic models of EAE (in Lewis and DA rats, respectively)
`were obtained 24 h after the last dose to determine trough
`levels. FTY720 and FTY720-P were found both in the blood
`and brain, with ratios between approximately 0.8 and 3 (Ta-
`ble 2). Brain levels of both forms were considerably higher
`(by factors of 10 to 27 after 23 doses) than those in blood, as
`seen before for FTY720 in normal rats (Meno-Tetang et al.,
`2006). Brain concentrations of the two forms were of compa-
`rable magnitude. The observed amounts of both FTY720 and
`its phosphorylated form in the DA rat brain increased with
`the oral dose of FTY720, as did the blood levels; thus, the
`brain/blood ratio was relatively constant in DA rats (21–27
`for FTY720; 14–17 for FTY720-P). We also followed the time
`course of FTY720 and FTY720-P concentrations in blood and
`brain (Table 2). Although blood levels were constant over 21
`
`Fig. 5. FTY720 rapidly reduces the peripheral
`lymphocyte count, with sustained activity at a low
`subtherapeutic dose during EAE. A, time course of
`lymphocyte count in the Lewis rat model of EAE
`following prophylactic treatment from days 0 to 13
`with vehicle (f), 0.3 mg/kg FTY720 (E), or 25
`mg/kg CsA (ⴱ); n ⫽ 6 per group, represented as
`mean ⫾ S.D. At 6 h postimmunization, FTY720
`had already reduced the lymphocyte number to
`the maximal extent. CsA, despite full efficacy dur-
`ing treatment (Fig. 4), did not significantly alter
`the lymphocyte count. Level of significance was
`based on ANOVA comparison with naive animals
`at day 0. B, reduction of peripheral lymphocytes
`during early and late EAE in DA rats therapeuti-
`cally treated from days 11 to 33 with vehicle (u);
`n ⫽ 21) or FTY720 at 0.03 (o; n ⫽ 26) and 0.3 (f;
`n ⫽ 13) mg/kg. Data were pooled from three EAE
`studies (Fig. 3) and expressed as mean percent-
`age ⫾ S.E. of lymphocytes in the adjuvant control
`(䡺; n ⫽ 13). ANOVA was performed against the
`vehicle of antigen-immunized animals. ⴱ, p ⱕ 0.05;
`ⴱⴱ, p ⱕ 0.01; ⴱⴱⴱ, p ⱕ 0.001.
`
`

`

`474
`
`Foster et al.
`
`Fig. 6. High-dose [14C]FTY720 autoradiography in rats.
`Representative whole-body, midsagittal autoradiolumino-
`grams taken 24 (A) and 168 (B) h after administration of
`seven daily oral doses of 7.5 mg/kg [14C]FTY720 to pig-
`mented rats. White lines (B) point to increased label in the
`brain and spinal cord by 168 h after the last dose.
`
`FTY720 within the CNS. Interestingly, the brain/blood ratio
`remained relatively constant (21–27 and 14–17 for FTY720
`and FTY720-P, respectively) after 3 weeks of treatment de-
`spite the dose range (0.03–0.3 mg/kg; Table 2).
`Regardless of how the FTY720-P levels are attained in the
`brain, we speculate that this phosphorylated metabolite may
`promote endogenous repair mechanisms in the CNS via S1P
`receptors on glial and/or neuronal cells. It is noteworthy that
`the nervous system is a major locus for constitutive S1P
`receptor expression in glial cells and neurons (Ishii et al.,
`2004). Four of the five known S1P receptor subtypes display
`a distinct distribution pattern within specific brain regions
`and cell lineages, as illustrated in Fig. 1. CNS expression of
`S1P5, for example, is restricted to oligodendrocytes (Terai et
`al., 2003) and expressed throughout development to the ma-
`ture myelin-forming cell. Subsequent to the discovery that
`S1P acts as an important regulator of cell growth, it has
`become increasingly clear that this sphingolipid mediator
`may induce the survival of such cells in the CNS (Ishii et al.,
`2004).
`Indeed, recent studies have demonstrated that
`FTY720-P promotes the survival of oligodendroglial lineage
`cells in vitro (Jung et al., 2007). Moreover, FTY720-P ligation
`of S1P receptors on astrocytes (Osinde et al., 2007) could
`contribute to its known enhancement of endothelial barrier
`function (Abbott et al., 2006; Baumruker et al., 2007) and
`possibly to myelination (Talbott et al., 2005; Ishibashi et al.,
`2006). Further studies are needed to directly elucidate the in
`vivo consequences of FTY720-P signaling on CNS cells, which
`is underscored by the recent in vitro observation that S1P
`activation of S1P1 and S1P3 receptors can inhibit gap junc-
`tions in astrocytes (Rouach et al., 2006).
`The signature feature of FTY720 is its ability to rapidly
`reduce blood lymphocytes as a consequence of S1P1-mediated
`retention in the peripheral lymph nodes (Lo et al., 2005). It is
`notable that FTY720 not only spares CD4⫹CD25⫹ T-regula-
`tory cells (Treg) but also induces their functional activity
`(Daniel et al., 2007). Other mechanisms that act indepen-
`dently of S1P receptors may also be part of the activity of
`FTY720, such as suppression of eicosanoid production due to
`inhibition of cytosolic phospholipase A2 (Payne et al., 2007).
`Importantly, FTY720 neither inhibits the activation of lym-
`phocytes at therapeutically relevant concentrations nor
`overtly alters their effector function, including antibody re-
`
`Fig. 7. Light microscopic [14C]FTY720 autoradiography in rats. Semithin
`Epon-embedded sections of spinal cord, counterstained with toluidine
`blue, at 24 h (A) and 7 days (B) after the last dose of [14C]FTY720. Black
`autoradiography granules (arrows) are primarily localized along the my-
`elin sheets. Neurons (N) and axons (A) are free of grains, except for
`occas

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