throbber
FTY720 Immunosuppression Impairs Effector T Cell
`Peripheral Homing Without Affecting Induction, Expansion,
`and Memory1
`
`Daniel D. Pinschewer,* Adrian F. Ochsenbein,* Bernhard Odermatt,† Volker Brinkmann,‡
`Hans Hengartner,* and Rolf M. Zinkernagel2*
`FTY720 (2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol hydrochloride) prolongs survival of solid organ allografts in animal
`models. Mechanisms of FTY720 immunomodulation were studied in mice infected with lymphocytic choriomeningitis virus
`(LCMV) to assess T cell responses or with vesicular stomatitis virus to evaluate Ab responses. Oral FTY720 (0.3 mg/kg/day) did
`not affect LCMV replication and specific CTL and B cells were induced and expanded normally. Moreover, the anti-viral humoral
`immune responses were normal. However, FTY720 treatment showed first a shift of overall distribution of CTL from the spleen
`to peripheral lymph nodes and lymphocytopenia was observed. This effect was reversible within 7–21 days. Together with un-
`impaired T and B cell memory after FTY720 treatment, this finding rendered enhancement of lymphocyte apoptosis by FTY720
`in vivo unlikely. Secondly, the delayed-type hypersensitivity reaction to a viral MHC class I-presented peptide was markedly
`reduced by FTY720. These results were supported by impaired circulation of LCMV specific TCR transgenic effector lymphocytes
`in the peripheral blood and reduced numbers of tissue infiltrating CTL in response to delayed-type hypersensitivity reaction.
`Thirdly, in a CD8ⴙ T cell-mediated diabetes model in a transgenic mouse expressing the LCMV glycoprotein in the islets of the
`pancreas, FTY720 delayed or prevented disease by reducing islet-infiltrating CTL. Thus, FTY720 effectively reduced recirculation
`of CD8ⴙ effector T cells and their recruitment to peripheral lesions without affecting the induction and expansion of immune
`responses in secondary lymphoid organs. These properties may offer the potential to treat ongoing organ-specific T cell-mediated
`immunopathologic disease. The Journal of Immunology, 2000, 164: 5761–5770.
`
`sible mechanism of action (7, 8). However, the concentrations
`needed for lymphocyte apoptosis in vitro were several orders of
`magnitude greater than the blood concentrations in rats given
`FTY720 at the therapeutic dose of 1 mg/kg (9). In vivo FTY720
`has been reported to shift the distribution of lymphocytes from
`spleen to Peyer’s patches and peripheral lymph nodes and accel-
`erate homing of naive lymphocytes to these organs (9). Another
`study suggested that low blood lymphocyte counts may reflect re-
`duced emigration of effector cells to the periphery (10), but they
`could not distinguish effects on the circulation of naive mature
`lymphocytes from specific effector cells. Whether FTY720 inter-
`feres with the generation of specific effector T cells has not been
`shown. Therefore, the mechanisms responsible for the reported
`reduced graft infiltration under FTY720 treatment (10) remain un-
`clear. In the present study we analyzed the immunosuppressive
`mechanisms of 0.3 mg/kg FTY720 daily as monotherapy on T cell
`responses to lymphocytic choriomeningitis virus (LCMV) and B
`cell responses to vesicular stomatitis virus (VSV) in the mouse.
`This dose reveals biological effects in vivo and may be given to
`transplantation patients in combination therapy with other immu-
`nosuppressants. An in vivo half-life around or below 24 h (data not
`shown) excludes the possibility of major drug accumulation.
`Primary infection with LCMV, a noncytopathic RNA virus, is
`controlled almost exclusively by CTL (11, 12). Immunopathology
`in this infection is also T cell-mediated (11, 13, 14), and ␣␤TCR
`transgenic CD8⫹ T cells are available against a major CTL target
`LCMV glycoprotein peptide 33–41 (GP33); so LCMV infection
`offers a good model to assess beneficial or detrimental effects of
`immunomodulation. On the other hand, VSV, a highly cytopatho-
`genic RNA virus, was used to study the effects of FTY720 on B
`cell responses. An early neutralizing Ab response against VSV is
`
`F TY720
`
`(2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol
`hydrochloride)3 is a chemical derivative of myriocin (ther-
`mozymocidin), a substance found in the fungi Myriococ-
`cum albomyces and Isaria sinclairii (1, 2). Despite the strong sup-
`pressive activity of myriocin in mixed lymphocyte reactions, this
`property is lost upon chemical modification to FTY720. FTY720
`has been shown to prevent graft rejection in various animal allo-
`transplantation models (3–5). Good synergy with cyclosporin and
`sirolimus has been reported (5, 6). The current strategy of immu-
`nosuppressive regimens is to combine different substances with
`distinct mechanisms of action to minimize the dose and toxicity of
`each individual therapeutic constituent.
`In vitro studies on FTY720 suggested that selective apoptotic
`cell death of lymphocytes, mainly of the T cell subset, was a pos-
`
`*Institute of Experimental Immunology and †Laboratory for Special Techniques, De-
`partment of Pathology, University Hospital, Zurich, Switzerland; and ‡Novartis
`Pharma AG, Basel, Switzerland
`Received for publication May 7, 1999 Accepted for publication March 21, 2000
`The costs of publication of this article were defrayed in part by the payment of page
`charges This article must therefore be hereby marked advertisement in accordance
`with 18 U S C Section 1734 solely to indicate this fact
`1 This work was supported by the Swiss National Science Foundation, the Kanton of
`Zurich, and Novartis Pharmaceuticals (Basel, Switzerland)
`2 Address correspondence and reprint requests to Dr Rolf M Zinkernagel, Institute of
`Experimental Immunology Schmelzbergstrasse 12, 8091 Zurich, Switzerland E-mail
`address: rolf zinkernagel@pty usz ch
`3 Abbreviations used in this paper: FTY720, 2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-
`propanediol hydrochloride; LCMV,
`lymphocytic choriomeningitis virus; CFSE,
`5-(and 6-)carboxyfluorescein diacetate succinimidyl ester; LCMV-WE, LCMV strain
`WE; LCMV-NP, nucleoprotein of LCMV; LCMV-GP33/GP33, peptide 33–41 of the
`LCMV glycoprotein; vaccinia-WR, vaccinia virus strain WR; VSV, vesicular stoma-
`titis virus; VSV-IND, VSV strain Indiana; VSV-NJ, VSV strain New Jersey; VSV-
`NP52/NP52, peptide 52–59 of the VSV nucleoprotein; tg, transgenic; DTH, delayed-
`type hypersensitivity reaction
`
`Copyright © 2000 by The American Association of Immunologists
`
`0022-1767/00/$02 00
`
`Apotex v. Novartis
`IPR2017-00854
`NOVARTIS 2049
`
`

`

`5762
`
`FTY720 IMPAIRS EFFECTOR T CELL PERIPHERAL HOMING
`
`required to prevent lethal encephalitis, and VSV infection induces
`lifelong protective B cell memory (15, 16).
`We now show that FTY720 does not impair efficient priming of
`T and B cell/Ab responses, but inhibits immune responses by pre-
`venting the homing of effector T cells to lesions in peripheral
`organs.
`Materials and Methods
`Mice
`C57BL/6 mice (H-2b), 318 TCR transgenic mice, expressing a TCR spe-
`cific for LCMV-GP33 together with H-2Db, and RIP-GP transgenic mice
`expressing the LCMV-GP on pancreatic islet cells were obtained from the
`breeding colony of the Institut fu¨r Labortierkunde, Veterinary Hospital
`(Zurich, Switzerland) Breeding was performed under specific pathogen-
`free conditions, and experiments were performed under conventional ani-
`mal facility conditions
`Viruses, virus detection, and inactivation
`Viruses. LCMV-WE was originally obtained from Dr F Lehmann-
`Grube (Heinrich Pette Institute, Hamburg, Germany) and was grown on
`L929 cells VSV Indiana (VSV-IND; Mudd-Summers isolate) and VSV
`New Jersey (VSV-NJ; Pringle isolate) were obtained from Dr D Kolak-
`ovsky (University of Geneva, Geneva, Switzerland) and were grown on
`BHK21 cells Vaccinia-WR was generously provided by Dr B Moss (Na-
`tional Institutes of Health, Bethesda, MD) and was propagated on BSC40
`cells
`Infectious LCMV in the organs was detected by focus-forming assays
`performed as previously described (17)
`For UV inactivation a small volume of high titer VSV preparation was
`exposed as a thin layer in a petri dish to a UV lamp (Philips, 15W) for 3
`min at a distance of 8 centimeters (18)
`Cell analysis and staining
`Cell analysis and counting of absolute numbers of transferred cells circu-
`lating in the blood were performed by FACS by mixing a defined number
`of autofluorescent beads with a known volume of blood to permit the
`calculation of positive cell number per microliter, using Becton Dickinson
`Trucount Tubes (Mountain View, CA) In addition to the manufacturer’s
`instructions, the absolute number of autofluorescent beads per sample was
`re-evaluated by FACS with the instrument settings used for the respective
`cell analysis Relative cell numbers varied within a range of ⫾2%; truly
`absolute numbers are, however, less precisely measurable
`For counting of transferred specific CTL present in secondary lymphoid
`organs single-cell suspensions were prepared as described for cytotoxicity
`assays The total number of lymphocytes per organ was assessed in a
`Neubauer chamber (improved bright-line, Brand Wertheim, Germany)
`The percentage of CD8⫹V␣2⫹V␤8⫹ triple-positive cells in the lympho-
`cyte population was evaluated by FACS and multiplied by the total number
`of lymphocytes to obtain the absolute number of CD8⫹V␣2⫹V␤8⫹ indi-
`cator CTLs per organ
`The fluorescent dye CFSE from Molecular Probes (Eugene, OR) was
`used to label cells before transfer as previously described (19)
`TCR transgenic CD8⫹ T cells were detected with anti-mouse CD8␣-
`TRI Ab from Caltag (Burlingame, CA), anti-mouse V␤8 1/8 2-FITC, and
`anti-mouse V␣2-PE Abs (PharMingen, San Diego, CA) CFSE-labeled
`CD4⫹ and CD8⫹ T cells were stained with anti-mouse CD4-TRI and anti-
`mouse CD8-TRI (both from Caltag), respectively
`Assessment of the primary footpad swelling reaction and of
`DTH/footpad infiltrating CTL
`LCMV-WE (200 PFU) in 30 ␮l of MEM supplemented with 2% FCS and
`GP33 (3 ␮g/ml) or NP52 (1 mg/ml) in 30 ␮l of balanced salts solution was
`injected into both hind footpads The footpad thickness was measured with
`a spring-loaded caliper (Kroeplein, Schluchtern, Hessen, Germany) Val-
`ues were taken as the mean of both hind footpads (20, 21)
`CTL assay and peptides
`Virus-specific cytotoxic T cells were assayed as described previously (22)
`Briefly, single-cell suspensions were prepared from the spleens and lymph
`nodes of mice at the indicated time points and used directly in a 51Cr
`release assay Target cells were either GP33-coated EL4 or vaccinia-WR-
`infected MC57 cells (multiplicity of infection of 3 for 2 h) The LCMV
`peptide GP33 and the VSV peptide NP52–59 were purchased from Neo-
`system Laboratoire (Strasbourg, France)
`
`Ab detection
`VSV-NJ and VSV-IND neutralizing Abs were determined in a neutraliza-
`tion assay as described previously (23) IgG was determined by its resis-
`tance to reduction by ␤-ME The difference between total Ig and IgG rep-
`resents IgM LCMV-NP-specific Abs were measured by ELISA using the
`following steps as described previously (24): 1) coating with baculovirus-
`derived LCMV-NP, 2) blocking with 2% BSA in PBS, 3) 10-fold predi-
`luted mouse serum was titrated in 3-fold dilutions over 12 steps, 4) IgM-
`or IgG-specific horseradish peroxidase-labeled goat anti-mouse Abs (0 5
`␮g/ml; Southern Biotechnology Associates, Birmingham, AL), 5) substrate
`2,2⬘-azinobis-3-ethylbenzthiazolazine sulfonic acid (Roche, Mannheim,
`Germany) and H2O2 (Fluka Buchs, Switzerland) Ab titers were deter-
`mined as the serum dilutions yielding an absorption (OD405) of twice back-
`ground levels
`Diabetes induction and assessment of blood glucose levels
`For the induction of an autoimmune diabetes, RIP-GP transgenic mice
`were infected i v with 200 PFU of LCMV-WE This elicits diabetes in
`RIP-GP transgenic but not in C57BL/6 mice (25) (see also Results) Glu-
`cose levels in the blood were measured using the Haemo-Glucotest color
`reaction sticks and were quantified with the reflection photometer Reflolux
`II (Roche) according to the manufacturer’s instructions
`Immunohistology
`Freshly removed organs were immersed in HBSS and snap-frozen in liquid
`nitrogen Tissue sections of 5-␮m thickness were cut in a cryostat, placed
`on siliconized glass slides, air-dried, fixed with acetone for 10 min, and
`stored at ⫺70°C For the staining of lymphocyte differentiation markers,
`rehydrated sections were incubated with primary rat mAbs against CD4
`and CD8 (YTS 191 and YTS 169) (26) Insulin was detected using affinity-
`purified guinea pig primary Abs (Dako, Glostrup, Denmark) Primary Abs
`were revealed by sequential incubation with either goat anti-rat Ig Abs
`(Tago, Burlingame, CA) and alkaline phosphatase-labeled donkey anti-
`goat Ig Abs or alkaline phosphatase-labeled rabbit anti-guinea pig Ig Abs
`and alkaline phosphatase-labeled goat anti-rabbit Ig Abs (Jackson Immu-
`noResearch Laboratories, West Grove, PA) Dilutions of the affinity-puri-
`fied secondary Abs were made in Tris-buffered saline containing 5% nor-
`mal mouse serum Alkaline phosphatase was visualized using naphthol
`AS-BI (6-bromo-2-hydroxy-3-naphtholic acid-2-methoxy anilide) phos-
`phate and new fuchsin as substrate Endogenous alkaline phosphatase was
`blocked by levamisole Color reactions were performed at room tempera-
`tures for 15 min with reagents from Sigma (St Louis, MO) Sections were
`counterstained with hemalum, and coverslips were mounted with glycerol
`and gelatin
`FTY720 administration
`FTY720 was obtained from Novartis Pharmaceuticals (Basel, Switzerland)
`and was dissolved in distilled water A dose of 0 3 mg/kg was given daily
`by gavage in a volume of 100 ␮l/10 g body weight
`Statistical analysis
`We performed repeated measures ANOVA with two between factors
`(group and trial) and one within factor (time point or E:T cell ratio, re-
`spectively) In experiments with two groups, post-hoc comparisons at dif-
`ferent time points were performed using unpaired t tests with Bonferroni
`correction In experiments with three groups, the Bonferroni-Dunn test was
`used for pairwise between-group comparisons Between-group compari-
`sons on different days were performed using a two-way ANOVA with
`post-hoc Bonferroni-Dunn test and an additional Bonferroni correction for
`tests on various days Cell counts were log-transformed to achieve an ap-
`proximate normal distribution and compared using a two-way ANOVA
`(factors trial and group) with Bonferroni correction for testing of multiple
`organs The figures show the data of only one representative experiment,
`while two or three comparable sets of experiments were used for statistical
`analysis as stated in the figure legends
`Results
`Reduced primary swelling reaction after LCMV infection
`Infection of C57BL/6 mice with 200 PFU of LCMV into the hind
`footpad causes a primary swelling reaction starting on day 6 and
`peaking around days 7–9. The first peak of the swelling reaction
`from days 6 to 8 is caused by CD8⫹ cells, whereas the following
`lesser swelling from days 9 to 14 is mainly CD4⫹ T cell dependent
`
`

`

`The Journal of Immunology
`
`5763
`
`FIGURE 1.
`Impairment of the primary footpad swelling reaction by
`FTY720 C57BL/6 mice were either treated from day ⫺1 until day 15 daily
`with 0 3 mg/kg FTY720 orally (f) or left untreated (䡺; control) On day
`0 they were immunized with 200 PFU of LCMV-WE diluted in a volume
`of 30 ␮l of MEM/2% FCS into the hind footpad Footpad swelling was
`monitored as described in Materials and Methods Each symbol represents
`a group of three mice One representative of two statistically analyzed
`experiments is shown Values indicate the mean ⫾ SD ⴱ, p ⬍ 0 005
`
`(21). This simple in vivo readout offers a reliable assessment of
`overall T cell-mediated immune responses. Immunosuppression
`by any mechanism modulating this response is readily revealed
`(27). One group of mice was treated daily with 0.3 mg/kg FTY720
`orally starting 1 day before infection, and the control group was
`left untreated. FTY720 treatment significantly reduced the swell-
`ing during the entire period ( p ⫽ 0.001; Fig. 1). This suggested
`that the early CD8⫹ as well as the CD4⫹ effector T cell activity
`were impaired at least as assessed locally in the footpad.
`
`Unimpaired induction and expansion of cytotoxic T cells
`To evaluate potential effects of FTY720 on T cell induction and
`expansion (28, 29) vs T cell effector function in the footpad, the
`following experiments were performed. Mice were immunized
`with 200 PFU of LCMV-WE i.v., and CTL activity was measured
`at its peak on day 8. Because a difference in the distribution of
`naive T cells had already been shown (9), not only splenocytes but
`also lymph node cells were tested as effectors in a 51Cr release
`assay (Fig. 2). An obvious decrease in cytolytic activity in the
`spleen ( p ⫽ 0.004; Fig. 2A) after FTY720 treatment contrasted
`with an increase in cytotoxic activity in peripheral lymph nodes
`( p ⫽ 0.01; Fig. 2B). This finding rendered a general reduction in
`the cytotoxic T cell population unlikely. Cells from pooled popli-
`teal, inguinal, mesenteric, para-aortic, axillary, and submandibular
`lymph nodes and spleen yielded the same cytotoxic activity in
`treated and untreated mice ( p ⫽ 0.74; Fig. 2C). The same exper-
`iment was repeated with vaccinia-WR with similar results (Fig. 2,
`D–F). FTY720-treated mice exhibited lower cytotoxic activity in
`the spleen ( p ⫽ 0.0001) and moderately more cytotoxicity in
`lymph nodes ( p ⫽ 0.01), but activity in pooled secondary lym-
`phoid organs was comparable to that in untreated control mice
`( p ⫽ 0.29). Despite an only moderate difference in lymph nodes
`(Fig. 2, B and E), statistical analysis of more than one comparable
`experiment resulted in overall significantly higher activity in
`lymph nodes of FTY720-treated mice. This finding should be
`viewed in the context of additional data presented below. Absolute
`numbers of lymphocytes found in pooled secondary lymphoid or-
`gans were about the same as those in controls (not shown). Taken
`together, these data show that a dose of 0.3 mg/kg FTY720 did not
`interfere significantly with the induction or expansion of specific
`cytotoxic T cells, but FTY720 apparently changed the distribution
`pattern of specific activated CD8⫹ effector T cells in a manner
`similar to that found for naive lymphocytes (9). To exclude effects
`
`FIGURE 2. Unimpaired induction and expansion of cytotoxic T cells
`under FTY720 treatment A–C, C57BL/6 mice were either treated with 0 3
`mg/kg FTY720 orally daily from day ⫺1 until the end of the experiment
`(f) or left untreated (䡺; control) After immunization with 200 PFU of
`LCMV-WE i v on day 0, mice were sacrificed on day 8, and cells from
`spleen and lymph nodes were tested in a primary in vitro cytotoxicity
`assay Specific cytotoxic activity against GP33-labeled EL-4 target cells
`was measured for spleen cells (A), for inguinal lymph node cells (B), and
`for pooled cells from popliteal, inguinal, mesenteric, para-aortal, axillary,
`and submandibular lymph nodes and spleen (C) Each symbol represents a
`group of three mice One representative of two statistically analyzed ex-
`periments is shown Spontaneous release was always ⬍20% Values indi-
`cate the mean ⫾ SD D–F, Mice were treated as described in A–C, but were
`immunized on day 0 with 2 ⫻ 106 PFU of vaccinia-WR i v On day 6 mice
`were sacrificed, and spleen and lymph node cells were tested in a primary
`in vitro cytotoxicity assay against vaccinia-WR-infected MC57 target cells
`(D, spleen cells; E, pooled lymph node cells; F, lymph node and spleen
`cells pooled) One representative of two or three statistically analyzed ex-
`periments is shown Spontaneous release was always ⬍20% Values indi-
`cate the mean ⫾ SD
`
`of FTY720 on viral replication, mice were infected with 200 PFU
`of LCMV i.v. Four days later viral titers were measured in the
`spleen and inguinal lymph nodes and were comparable in treated
`and untreated mice (data not shown).
`
`Induction of lymphocytopenia and its reversibility
`In vitro studies had suggested that specific apoptotic death of T
`cells was responsible for the immunosuppressive properties of
`FTY720 (7, 8, 30). Apoptosis might have been expected to reduce
`the cytotoxic effector population, but the results of overall unim-
`paired induction of CTL responses did not readily support this
`idea. In addition, previous studies showed a transient lymphocy-
`topenia after a single dose of FTY720 (3, 9), but did not evaluate
`
`

`

`5764
`
`FTY720 IMPAIRS EFFECTOR T CELL PERIPHERAL HOMING
`
`FIGURE 4.
`Impaired circulation of CD8⫹ cytotoxic T cells C57BL/6
`mice were either treated from day ⫺1 until day 15 daily with 0 3 mg/kg
`FTY720 orally (f) or left untreated (䡺; control) and infected with 200
`PFU of LCMV-WE i v on day 0 The mice had received 104 318-tgTCR
`V␣2⫹V␤8⫹ spleen cells specific for Db and LCMV-GP33 i v on day ⫺2
`tgTCR⫹ (V␣2⫹V␤8⫹) CD8⫹ T cells in the blood were measured at the
`time points indicated One representative experiment of three is shown
`Values indicate the mean ⫾ SD
`
`of specific effector T cells (31). To follow specific effector T cells
`in the blood of mice, 104 indicator spleen cells from mice with a
`transgenic TCR specific for Db and LCMV-GP33 (318-mouse)
`were transferred to naive recipients that were infected i.v. with 200
`PFU of LCMV-WE. It has previously been shown that this number
`of transferred cells replaces about 50% of the endogenous GP33-
`specific CD8⫹ T cells without changing the overall response
`against LCMV (32, 33). The number of specific circulating trans-
`genic TCR⫹ CD8⫹ T cells per microliter of blood was measured
`by FACS analysis. FTY720 treatment reduced the number of spe-
`cific circulating CD8⫹ T cells by more than a factor of 10 (Fig. 4).
`
`Comparable numbers, but changed distribution pattern, of
`specific CD8⫹ cytotoxic effector T cells
`Comparing the above finding with the data from Fig. 2, it was
`rather unlikely that a reduced population of specific CD8⫹ cyto-
`toxic effector T cells was the underlying reason for their reduced
`circulation in the peripheral blood (Fig. 4). To corroborate this
`conclusion and to further analyze the effect of FTY720 on the
`distribution pattern of specific CD8⫹ cytotoxic effector T cells,
`mice were treated as described in Fig. 4 and were sacrificed at the
`peak of the CTL response on day 8 after infection. Transgenic
`TCR⫹ CD8⫹ T cells were counted in blood, spleen, inguinal
`lymph node, and pooled secondary lymphoid organs (spleen
`pooled with popliteal, inguinal, mesenteric, para-aortal, axillary,
`and submandibular lymph nodes) as described in Materials and
`Methods (Fig. 5). Comparable numbers of indicator CTL were
`present in pooled secondary lymphoid organs of FTY720-treated
`and untreated mice ( p ⫽ 0.52), while drug treatment significantly
`reduced the number of CTL circulating in the blood ( p ⬍ 0.0001).
`Similar to the behavior of naive lymphocytes (9) and compatible
`with the data shown in Fig. 2, A and D, fewer specific CD8⫹
`cytotoxic effector T cells could be found in the spleen of FTY720-
`treated mice compared with untreated control mice ( p ⫽ 0.0003),
`while their number was increased in the inguinal lymph node ( p ⫽
`0.0001). This result taken together with the data from Figs. 1, 2,
`and 4 suggested that FTY720 impairs the circulation of effector
`CD8⫹ T cells, apparently by sequestering them in secondary lym-
`phoid organs, mainly peripheral lymph nodes, without impairing
`their induction or expansion.
`
`FIGURE 3.
`Induction of lymphocytopenia by FTY720 and its revers-
`ibility On day ⫺1, 3 ⫻ 107 naive CFSE-labeled splenocytes were adop-
`tively transferred i v into naive syngeneic C57BL/6 recipients The recip-
`ients were then treated with a single dose of 0 3 mg/kg FTY720 orally on
`day 0 (䡺), treated with the same dose daily from day 0 until day 6 (Œ), or
`left untreated (control ⫽ 100%) The numbers of transferred, CFSE-posi-
`tive circulating CD4⫹ (A) and CD8⫹ (B) T cells per microliter of blood
`were measured by FACS analysis; staining of transferred cells was per-
`formed as described in Materials and Methods Gated living white cells
`were analyzed for CFSE⫹CD4⫹ (A) and CFSE⫹CD8⫹ (B) double-positive
`cells, respectively The values are expressed as a percentage of the mean of
`the control group Each group consisted of four animals One representa-
`tive experiment of two is shown Values indicate the mean ⫾ SD
`
`recovery due to newly generated lymphocytes after FTY720 treat-
`ment was stopped. To address this question, naive spleen cells
`were labeled with the vital dye CFSE (19) and adoptively trans-
`ferred i.v. to naive recipients. The recipients were then treated
`either with a single dose of 0.3 mg/kg FTY720 or with the same
`dose for 7 consecutive days. The numbers of transferred CD4⫹
`(Fig. 3A) and CD8⫹ T cells (Fig. 3B) in the peripheral blood were
`measured by FACS and compared with those in untreated control
`mice. A marked drop in both populations was observed a few
`hours after a single dose of 0.3 mg/kg FTY720; this drop was more
`severe after repetitive treatments. Within 7 or 21 days after treat-
`ment was stopped, respectively, transferred CD4⫹ and CD8⫹ T
`cells in the blood had recovered levels within 10% of the untreated
`control values. The same recovery was found for B lymphocytes
`(data not shown). Additional experiments had shown that the cell
`numbers slowly but continuously recovered during the time span
`of 7 and 21 days, respectively (data not shown). Recovery due to
`proliferation could be excluded, because proliferation and the re-
`sulting CFSE dilution would have been easily detected by alter-
`ation of the fluorescence intensities (19). These results indicate that
`the recovery of circulating lymphocytes after transient FTY720
`application is not due to newly generated cells, suggesting that
`FTY720 probably causes sequestration of lymphocytes rather than
`significantly affecting their viability.
`
`Impaired circulation of specific CD8⫹ cytotoxic effector T cells
`The obvious discrepancy between the reduction of the primary
`swelling reaction and the overall efficient induction and expansion
`of cytotoxic T cells required analyses of the circulation properties
`
`

`

`The Journal of Immunology
`
`5765
`
`FIGURE 6. Reduced DTH reaction under FTY720 treatment C57BL/6
`mice were either treated daily with 0 3 mg/kg FTY720 orally starting on
`day ⫺1 (f) and on day 5 after infection (Œ), respectively, until the end of
`the experiment or were left untreated (䡺 control) Thirteen days after in-
`fection with 200 PFU of LCMV-WE i v on day 0, 30 ␮l of GP33 solution
`in a concentration of 3 ␮g/ml dissolved in BSS was injected into both hind
`footpads Footpad swelling was measured as described in Materials and
`Methods Each symbol represents a group of five mice One representative
`of two statistically analyzed experiments is shown Values indicate the
`mean ⫾ SD ⴱ, p ⬍ 0 0016 (for f vs 䡺 and for Œ vs 䡺 only, but not for
`f vs Œ)
`
`control, p ⫽ 0.0001) to the same extent as had been found in mice
`treated from day ⫺1 onward (FTY720 day 1 vs FTY720 day 5,
`p ⫽ 0.51; Fig. 6).
`
`Prevention of a CD8⫹ T cell-mediated autoimmune diabetes
`Interference with the homing of effector T cells to peripheral le-
`sions may offer new therapeutic possibilities against immuno-
`pathological or autoimmune disease. To examine this, we analyzed
`a transgenic mouse expressing the glycoprotein of LCMV in the
`islets of the pancreas (RIP-GP) (25). In this model the transgenic
`GP is ignored by CD8⫹ T cells in the naive mouse, but after
`infection with LCMV, CD8⫹ T cell-mediated autoimmune diabe-
`tes develops because potentially autoreactive T cells have not been
`deleted in the thymus or the periphery, and effector CTLs are ef-
`ficiently induced following LCMV infection of secondary lym-
`phoid organs (25). Consequent perforin-dependent killing by CTLs
`leads to islet destruction and hyperglycemia (34). In a total of three
`experiments, daily treatment with 0.3 mg/kg FTY720 starting 1
`
`FIGURE 7. Prevention of CD8⫹ T cell-mediated autoimmune diabetes
`RIP-GP transgenic mice were either treated with 0 3 mg/kg FTY720 orally
`daily from day ⫺1 until day 20 (f) or left untreated (䡺) On day 0 they
`were immunized with 200 PFU of LCMV-WE i v Glucose levels in the
`blood were measured as described in Materials and Methods Each symbol
`represents an individual mouse One representative experiment of three is
`shown
`
`FIGURE 5. Changed distribution pattern of specific cytotoxic T cells
`C57BL/6 mice were either treated from day ⫺1 until day 8 daily with 0 3
`mg/kg FTY720 orally (f) or left untreated (䡺) and were infected with 200
`PFU of LCMV-WE i v on day 0 The mice had received 104 318-tgTCR
`V␣2⫹V␤8⫹ spleen cells specific for Db and LCMV-GP33 i v on day ⫺2
`Mice were sacrificed at day 8 after infection, and tgTCR⫹ (V␣2⫹V␤8⫹)
`CD8⫹ T cells were counted in blood, spleen, inguinal lymph node, and
`pooled secondary lymphoid organs (pooled spleen and popliteal, inguinal,
`mesenteric, para-aortal, axillary, and submandibular lymph nodes) as de-
`scribed in Materials and Methods One representative of two statistically
`analyzed experiments is shown Values indicate the mean ⫾ SD
`
`Reduced LCMV-specific delayed-type hypersensitivity reaction
`(DTH)
`To study the recruitment of effector CD8⫹ T cells to s.c. sites with
`Ag, mice were immunized with 200 PFU of LCMV-WE i.v. and
`13 days later were treated with the immunodominant Db nonapep-
`tide, gp33–41 of LCMV-glycoprotein (GP33). This peptide elicits
`an exclusively CD8⫹ T cell-mediated DTH in LCMV-infected, but
`not in naı¨ve, C57BL/6 mice (20). Footpad injection of the unre-
`lated immunodominant Db binding octapeptide np52–59 of VSV-
`nucleoprotein (NP52), which causes a similar reaction in VSV-
`infected mice (20), did not lead to any footpad swelling (data not
`shown). Daily treatment with 0.3 mg/kg FTY720 starting 1 day
`before infection significantly reduced the CD8⫹ T cell-mediated
`DTH against GP33 (FTY720 day 1 vs control, p ⫽ 0.0001; Fig. 6).
`To avoid an effect of FTY720 on early phases of the anti-LCMV
`response, a third group of mice was only treated with FTY720
`from day 5 after infection onward. Consistent with earlier findings
`(4, 6), the evoked DTH reaction was reduced (FTY720 day 5 vs
`
`

`

`5766
`
`Fl'Y'lZO IMPAJRS EFFECTOR T CELL PERIPHERAL HOMING
`
`.‘
`‘
`V
`. m‘
`‘
`y
`-
`.
`fa.’§( 6.\.‘.‘; ANN-O, ‘\'“.\‘ :t'
`V
`-’ '-
`2 -
`
`
`‘
`'5'.
`"s-
`!v‘.
`‘ “a “‘ “2‘
`51-.
`337 ' fl’f‘naflhi w’ 3W u‘
`,
`.
`.
`,V 1,;
`. 1.. «.
`‘
`
`
`,
`
`

`

`The Journal of Immunology
`
`5767
`
`tration, the following experiments were performed. RIP-GP mice
`were treated as described above (Fig. 7) and sacrificed on days 9
`and 20 after infection for histological examination. On day 9 con-
`trol mice developed diabetes and exhibited considerable CD8⫹
`infiltrates (Fig. 8B), while only very few CD8⫹ T cells were visible
`around the pancreatic islets of FTY720-treated mice (Fig. 8F).
`Corresponding to the degree of diabetes they had developed (Fig.
`7), a similar reduction of CD8⫹ cytotoxic T cells could also be
`seen 20 days after infection (Fig. 8H, normoglycemic mouse) com-
`pared with dense aggregates in untreated control mice (Fig. 8D).
`Parallel to increased CD8⫹ T cell infiltration staining for insulin
`decreased (Fig. 8, A, C, E, andG).
`C57BL/6 mice were treated with 0.3 mg/kg FTY720 orally daily
`starting 1 day before infection with 200 PFU of LCMV-WE i.v. on
`day 0. On day 13 LCMV-GP33 was injected into the hind footpad,
`and the mice were sacrificed 1, 3, 8, and 18 h later. Histological
`sections of the footpad were analyzed for infiltrating CD8⫹ and
`CD4⫹ T cells. One hour after peptide injection, some perivascular
`CD8⫹ T cells could be found in the s.c. tissue of untreated control
`mice (Fig. 8I), while no infiltration was seen in FTY720-treated
`mice (Fig. 8M). Two hours later massive aggregates of perivascu-
`lar CD8⫹ T cells and a beginning infiltration of approximate sweat
`glands (Fig. 8J) were considerably reduced by FTY720 (Fig. 8N).
`Eight hours after peptide challenge FTY720 treatment reduced the
`extensive infiltration of sweat glands (Fig. 8, K and O) and muscle
`(not shown). Another 10 h later, decreasing numbers of CD8⫹ T
`cells had caused considerable destruction and edema in untreated
`mice (Fig. 8L), while in the footpad of FTY720-treated mice
`CD8⫹ T cells had caused correspondingly less damage (Fig. 8P).
`Only very few to no CD4⫹ T cells were found in the samples
`above (Fig. 8, Q, R, U, andV, and data not shown) independent of
`FTY720 treatment. Present mainly at later time points, CD4⫹ T
`cells were probably attracted by the local inflammatory process
`initiated by CTLs. Footpad injection of the Db binding immuno-
`dominant VSV peptide NP52 did not cause either swelling (not
`shown) or considerable infiltration of CD8⫹ (Fig. 8, S, T, W, and
`X) or CD4⫹ T cells (data not shown) independent of FTY720
`treatment. Similarly, GP33 did not cause infiltration in naive
`C57BL/6 mice (data not shown). This suggests that the infiltrates
`shown in Fig. 8,I—

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