throbber
APOTEX ET AL. - EXHIBIT 1057
`Apotex Inc. et al. v. Novartis AG
`IPR2017-00854
`
`

`

`
`
`.MEDIcALscIENCES
`
`
`
`Table 1. Anthropometric parameters, leptin, and IFN-y measurements in RRMS patients and
`NIND controls
`
`Parameter measured
`
`No. of patients
`Sex (male/female)‘
`Age, yr
`Height, m
`Mass, kg
`BMl, kg/rnZ
`Serum leptin, pg/ml
`Serum leptin/BMI
`CSF leptin, pg/ml
`CSF leptin/BMl
`CSF leptin/serum leptin
`CSF leptin/CSF albumin
`CSF leptin index’
`CSF lFN-y, pg/ml
`
`RRMS patients
`
`126
`58/68
`36.5 : 9.5
`1.65 : 0.07
`65.9 I 9.6
`24.1 : 3.3
`21,5170 : 15,6760
`900.0 : 650.0
`1,143.1 : 1,389.5
`47.7 : 57.3
`0.09 t 0.18
`5,7 t 6.5 X10"S
`20.7 : 53.4
`3.9 I 3.1
`
`NIND controls
`117
`52/65
`38.2 1 15.5
`1.66 I 0.07
`65.4 t 10.7
`23.4 : 3.2
`11,7270 2: 13.0570
`488.6 : 482.0
`205.3 : 222.5
`8.2 1 8.7
`0.03 i 0.06
`1.2 : 2.1 x 10‘6
`7.0 I 12.2
`0.45 : 1.3
`
`P
`
`0.30
`0.52
`0.65
`0.12
`0.0001
`0.0001
`0.0001
`0.0001
`0.001
`0.0001
`0.008
`0.0001
`
`*The CSF leptin index is a measure of in situ synthesis of leptin in the CNS, calculated with the following formula:
`(CSF leptin/CSF albumin)/(serum leptin/serum albumin).
`
`Flow Cytometry. Immunophenotypic analysis of peripheral blood
`from RRMS patients and healthy controls was performed with an
`EPICS XL flow cytometer (Beclunan Coulter) using the Beckman
`Coulter software program XL SYSTEM 11. Triple combinations of
`various anti—human mAbs were used (Coulter Immunotech, Mar-
`seille, France). All samples were analyzed within 3-4 h of sampling,
`and staining was performed according to standard procedures as
`described in ref. 17.
`‘
`
`CD4+CD25+ TchS in various mouse strains were analyzed by
`flow cytometry with a FACSCalibur flow cytometer (Becton
`Dickinson) and the Becton Dickinson software program
`CELLQUEST. mAbs were added to single cell suspensions of lym—
`phocytes obtained from spleens and lymph nodes after lysis with the
`ACK buffer [0.15 M NH4Cl/10 mM KHCO3/0.1 mM NazEDTA
`(pH 7.4)]. The analysis and quantification of the TRCgs population
`was obtained by gating on CD4‘ T cells.
`
`Human Myelin Basic Protein (hMBP) T Cell Lines. hMBP—specific
`short—term T cells lines were generated according to a method
`reported in ref. 18. The T cell lines were derived from peripheral
`blood lymphocytes of three naive-to-treatment RRMS patients.
`
`Proliferation and Suppression Assays. For in vitro blocking experi-
`ments, Abs against human leptin provided by Radek Sokol (Bio-
`Vendor, Brno, Czech Republic) and mAb against the human leptin
`receptor (R & D Systems) were used at a final concentration of
`10—25 ug/ml; the control was irrelevant IgG Ab (BioVendor).
`The in vitro suppressive capacity of TMS isolated from RRMS
`patients and healthy controls was measured after magnetic cell
`sorting by using the Dynal CD4‘CD25' Tch kit (Dynal, Oslo).
`Briefly, CD4‘CD25‘ T cells (5 ><'10" cells per well) were cocul-
`tured with CD41CD25+ (5 X 104 cells per well) in a 1:1 ratio (both
`98% pure) and stimulated for 3 d in the presence of anti-CD3/
`CD28 Dynabeads (0.1 bead per cell) (Dynal). In mice, TRCgs were
`isolated with the Regulatory T Cell Isolation kit (Miltenyi Biotec,
`Gladbach, Germany) and stimulated with anti-CD3 antibody (2C11
`hybridoma) at 200 ng/ml final concentration and irradiated (30 Gy)
`T cell—depleted syngeneic splenocytes (1:1 ratio, 5 X 104 cells per
`well).
`
`Immunocytochemistry. T cells cultured, or not, with hMBP were
`washed twice with PBS on d 5 of culture, spotted onto glass slides,
`and fixed with methanol for 2 min. Leptin and ObR were detected
`with polyclonal Abs (Santa Cruz Biotechnology) (3).
`
`Mice. Female ob/ob (C57BL6/J-0br/0b), WT controls (C57BL6/J-
`WT), female leptin—receptor-deficient (db/db) mice (C57BL—Ks—
`
`db/db), C57BL—Ks—db/+ controls (db/+), and SJL/J mice (all 6»8
`weeks old) were obtained from Harlan Italy (Corezzana, Italy).
`Experiments were performed following the guidelines of the Isti—
`tuto Superiore di Sanita, Rome.
`
`EAE Induction and Treatment with the Fusion Protein of ObR and the
`
`Fc Fragment of IgG (Obnch). The peptide used for EAE induction
`in SJL/J female mice was the proteolipid protein peptide
`(PLP)139_151) (HSLGKWLGHPDKF). The peptide was synthe-
`sized by INBIOS (Pozzuoli, Italy), purity was assessed by HPLC
`(>97% pure), and amino acid composition was verified by mass
`spectrometry. For EAE induction, mice were immunized so in the
`flank with 100 [.L1 of complete Freund’s adjuvant (Difco) emulsified
`with 100 ug of PLP139_151 peptide on d 0 and with 200 ng of pertussis
`toxin (Sigma) i.p. on d 0 and d 1. Mice were scored for clinical
`symptoms and weighed daily according to a system described in refs.
`2 and 3. Brains and spinal cords were dissected 1520 d after
`immunization and fixed in 10% formalin. Paraffin—embedded sec
`tions of 5 pm thickness were stained with hematoxylin/eosin, and
`sections from 4—10 segments per mouse were examined blindly for
`the number of inflammatory foci by using a scoring system de—
`scribed in ref. 3.
`
`The chimeric fusion protein OszFc (R & D Systems) in 200 [.L1
`of PBS was injected i.p. at a dose of 100 pg per mouse per day for
`three consecutive days. Thus, treatment with OszFc of SJL/J mice
`was performed on d ~1, d 0, and d +1 both before and after
`PLPthsl immunization. The same amount of control IgG was
`injected i.p. in the control SJL/J mice.
`
`Real-Time Quantitative PCR. mRNA was extracted from purified
`CD4‘CD25‘ cells (98% pure by FACS analysis) by using the
`MicroFastTrack 2.0 kit
`followed by cDNA synthesis with
`the SuperScript System (Invitrogen). Expression levels of the
`transcription factor FoxP3 were analyzed by real-time quan—
`titative PCR (TaqMan gene expression assay) by using an ABI
`PRISM 7700 thermal cycler (Applied Biosystems). TaqMan
`primers and probes for FoxP3 and for the housekeeping gene
`GAPDH were purchased as premade kits (Applied Biosys-
`tems). For relative quantitation of gene expression to the
`endogenous control, the comparative CT method was used in
`accordance with the manufacturer’s guidelines. Results are
`expressed as the percentage of FoxP3 increase compared with
`CD4‘CD25‘ effector T cells.
`
`Statistical Analysis. Nonparametric analyses were performed by
`using the Mann—Whitney U test for unrelated two-group analyses.
`The AN OVA test was used to assess differences between groups.
`
`Matarese et al,
`
`PNAS
`
`| April 5, 2005
`
`| Vol.102
`
`| no.14 l
`
`5151
`
`a 7
`
`1
`fl
`
`
`
`

`

`a
`RRMS Manta-125)
`mm} y-7ousxusom
`A
`3 n2=m-
`.
`E E6ml “‘01
`.
`3mg 19-,”
`
`'
`
`~-
`
`‘
`
`'
`
`mun comm (n I117)
`b
`10080 y-1mw’32usn
`sew viz-0.2
`Rum
`
`
`
`uts l7.5 20 225 25 27.5 30 32.5
`
`0‘5 175 20 R5 25 27.5 36 32.5 H 37.5
`35 37.5
`Body mass Index (mm?)
`
`C
`
`7WD:
`Emmi
`moi
`
`,
`
`_
`
`-
`
`.
`
`.
`
`.
`anus mum (n - ml
`y - ms: O um
`R - u:
`Iii-0.0“"
`.P; 0.1
`
`7m
`3cm
`50°”
`
`nun Willis-111’)
`y = 1515:4551:
`R - u
`RZIIM
`P- 0.0001
`
` 015 £75 20 22.5 25 275 30 32.5 35 375
`
`Body mass index (kglmz)
`
`.
`
`'
`;
`
`.
`
`'
`
`-
`
`'
`
`mm
`
`e
`9
`E
`gm
`5 1,0000
`.
`s
`5 30°00 .
`a 20900 ‘
`
`was WM! (n' m)
`y- m: . mom
`- R:- 0.1
`n - 0.3
`‘ P-nnooi /,
`‘
`.
`//’.
`.
`
`c
`
`f
`mom
`
`m
`5mm
`W
`
`
`
`
`NIND controls (n = in;
`yI31.7ZI 0 5214.4:
`R1 - 0.:
`n . 0.54
`P-um
`
`3am .
`
`2mm 100m
`
`
`20m1050.. ll!
`
`onlmmmrmmwmmm
`ODIWMM‘OOOSGOOWFW
`CSF lentil) (99ml
`
`Minn control: (n - 117}
`y amnion)
`R2 - (worm
`R - M17
`P - rm
`
`Runs mm in - m;
`y- 554.39"an
`iu - 91
`R . us
`P a «.0001
`
`.
`.v_,/
`
`.._/’/
`
`'
`
`52
`
`14
`
`15
`
`h
`mac
`sum
`SM
`mu
`
`3000
`
`CSF interferonq (pglml)
`
`Fig. 1. Naive‘to-therapy RRMS patients show an increased secretion of leptin in
`serum and CSF that correlates with lFN-y production in CSF. Statistical analyses of
`these data are summarized in Table 1. (a and b) Simple regression analysis
`between serum leptin and BMI in RRMS patients (n = 126) and NlND controls(n :
`117). The correlation was lost in RRMS patients, whereas correlation was main-
`tained in NlND controls. (cand d) The correlation between CSF leptin and BMI was
`lost in RRMS patients whereas correlation was very strong in NlND controls. (e and
`f) Significant correlation between serum and CSF leptin in both RRMS patients
`and NIND controls; the correlation was stronger in patients than in controls. (9
`and h) Simple regression analysis between the CSF leptin and the lFN—y levels in
`CSF in both RRMS patients and NIND controls. Only in RRMS patients was there a
`statistically significant positive correlation between the CSF leptin and the lFN—y
`levels in CSF ln NIND controls, the lFN»y average levels were very low (see Table
`1), and no correlation was observed with CSF leptin.
`
`Simple regression analysis and the Pearson’s correlation coeffi—
`cients were adopted to study the relationship between different
`variables. The program used was STATVIEW (Abacus Concepts,
`Cary, NC). Results are expressed as mean : SD; P < 0.05 was
`considered statistically significant.
`
`Results
`
`increased Serum and CSF Leptin in Naive-to-Treatment RRMS Patients
`Correlates with lFN-y Production in CSF.
`We found that leptin was increased in both serum and CSF of
`naive—to-therapy RRMS patients (Table 1 and Fig. 1 a—d). These
`
`5152
`
`i www.pnas.0rg/cgi/doi/‘l0.1073,i’pnas.0408995102
`
`differences were maintained even when serum and CSF leptin
`were normalized for BMI (Table 1 and Fig. 1 a—d). In addition,
`as expected, serum and CSF-leptin secretion positively corre—
`lated with BMI
`in NIND controls (Fig.
`1 b and d). This
`correlation was lost in RRMS patients (Fig.
`1 a and c). Con—
`versely, the correlation between serum leptin and CSF leptin was
`maintained in both RRMS patients and NIND controls; how?
`ever, this correlation was stronger in NIND controls than in
`RRMS patients (Fig. 1 e and f). We also compared the CSF-
`leptin/serum-leptin ratio and observed a statistically significant
`increase of this value in RRMS (Table 1). This evidence was
`further supported by the lack of increase of albumin in the CSF
`of RRMS patients, a marker of blood-brain-barrier (BBB)
`damage. In addition, we calculated the CSF-leptin/CSF-albumin
`ratio as a further indicator of BBB integrity and the CSF leptin
`index, calculated as the (CSF leptin/CSF albumin)/(serum
`leptin/serum albumin),
`to evaluate the in situ production of
`leptin by CNS. As shown in Table 1.
`the CSF-leptin/CSF-
`albumin ratio and the CSF leptin index were higher in RRMS
`patients (Table 1), suggesting the production of leptin by CNS in
`RRMS.
`
`Finally, we measured the amount of IFN-y and IL—4 in CSF and
`observed a significant increase in IFN-y (Table 1) and a positive
`correlation with CSF leptin secretion in RRMS patients only (Fig.
`1g and h). IL—4 did not show any significant increase in CSF, and
`the concentration of IL-4 was always below the detection limit of the
`assay in both RRMS and NIND controls (data not shown).
`
`anti-Leptin
`RRMS Patient '1 E150
`
`E100
`El
`50
`
`0
`
`g 200
`
`Medium
`
`memo pglml)
`
`Mun cum: Anson mm
`($5!qu (10W) (10149”!
`map (10ugiml)
`
`lines against hMBP derived from nai‘ve-to-treatment RRMS
`T cell
`Fig. 2.
`patients produce immunoreactive leptin, up-regulate the ObR, and are inhibe
`ited in their proliferation by anti-leptin or anti—Ieptin-receptor blocking an-
`tibodies, (a—c) Expression of leptin in T cells from a naive—to~treatment RRMS
`patient in the presence of medium alone (a) or after activation with hMBP (b
`and c). Leptin was detectable only after activation in the cytoplasm of T cells.
`(d—f) Expression of ObR on T cells in the presence of medium only (d) or after
`activation with hMBP (e and f). The ObR was expressed at very low levels
`before activation and was significantly up—regulated on the cell membrane
`after activation with hMBP (e and 1‘). All photos show immunoperoxidase
`staining with diaminobenzidine chromogen (brown) and hematoxylin coun-
`terstaining (violet). The open squares in b and e represent the zone of higher
`magnification shown in c and 1‘, respectively. (Magnification: a, b, d, and e,
`x100; c and 2‘, X400.) (9) Anti~hMBP short-term T cell lines secrete immuno—
`reactive leptin. (h) The anti-hMBP proliferative response ofT cells is inhibited
`by the addition to cell cultures of either of the anti‘Ob or anti-ObR antibodies.
`The data shown are from one representative experiment of three.
`
`Matarese et al.
`
`a a '
`
`3
`
`

`

`.9
`
`Et
`
`.
`8
`:5
`2
`s
`3
`o
`as
`
`a
`
`‘
`
`_r:
`§ 40000
`E 30000
`‘3 20000
`10000
`0
`
`b mono
`.
`A 60M I .
`-
`
`50000 ‘
`
`
`
`
`90
`so a
`70 o
`5°
`i:
`so 8
`‘°
`‘
`30 g
`20
`10
`a
`
`RRMS
`
`Healthy controls
`
`0
`
`.
`I
`
`RRISpitlmhtnIM}
`y.4_m.5u,15
`Rum
`Rae,”
`P-am
`
`
`
`0
`
`2.5
`
`I5 I75 20 22.5
`125
`10
`7.5
`5
`at, cmtcozsi oicm+ cells
`
`C mono
`A 60000
`E
`3 some
`E .0...
`5 some
`as
`20000
`10000
`
`0
`
`g
`
`Mycoritmbtn-m
`y-lmihrtifll
`m-m
`R=Ml
`P 0.
`'
`2
`

`
`0°

`
`0
`
`0
`
`e
`8
`0
`0
`
`ea c
`on o
`o
`o
`2.5
`5
`7.5
`in
`its 15
`17.5
`20
`22.5
`at cm’tcozs+ oi 004‘ cells
`
`d raccoo-
`
`4
`
`E
`% iCOO’fl
`9
`E
`%
`5 amour
`
`f
`T
`
`
`
`Q RRMS patieniur
`I RRMS patientitz
`I Healthy control
`I
`r-Wfi
`seam—“a
`r
`
`'52%
`
`
`CDd'CDZS' W’CDfi'
`cwcozs'
`carcms‘
`Beads aniiCDSICDZE slimulaflon
`
`
`
`
`
`
`in
`Inverse correlation between serum leptin and circulating TReg,
`Fig. 3.
`RRMS patients. (a) The immune phenotype of circulating lymphocytes in RRMS
`patients selected on the basis of their increase in serum leptin (RRMS patients
`with a serum leptin increaseto 22.5~fo|d higherthan the mean ofserum leptin
`observed in NIND and healthy controls) revealed a significant reduction in the
`percentage and the absolute number of circulating TReg,_ (t, P : 0.0001 and *,
`P : 0.0001, respectively). (b and c) Astatistically significant inverse correlation
`was observed between serum leptin and circulating TRegs in RRMS patients (b),
`whereas no correlation was observed in healthy controls (c). (d) Functional
`analysis of CD4 ‘CD25’ TR,395 of two RRMS patients selected on the basis of an
`increase in serum leptin. The proliferative response was inhibited upon addi~
`tion of CD4'CD25' cellsto the CD4tCD25‘ responder population at a 1:1 ratio
`in normal controls (black bars). CD4‘CD25' cells from two naive-to—therapy
`patients with RRMS exhibited significantly less suppressor activity (white and
`gray bars). *, P = 0.03. CD4’ CD25' cells alone were unresponsive upon
`stimulation as reported in ref. 9. The numbers above the bars represent the
`percent of inhibition of proliferation in the experiment. The data shown are
`from one representative experiment of five.
`
`OszFc Soluble Chimera Increases the Number of Thy, and Amelio-
`rates Clinical Course and Progression of Relapsing EAE (R-EAE). Treat—
`ment of normal R—EAE-susceptible SJL/J mice with anti-leptin
`
`PNAS
`
`[ April 5. 2005
`
`|
`
`vol. 102
`
`i
`
`no. 14
`
`|
`
`5153
`
`
`
` MEDICALsciences
`
`RRMS Patient-Derived T Cell Lines Activated with hMBP Produce
`
`Immunoreactive Leptin and Up-Regulate the ObR. To investigate
`whether leptin could be secreted by hMBP-activated autoreactive T
`cells present in the CNS, we generated short-term T cell lines from
`RRMS patients and stained them with leptin- and ObR-specific
`antibodies. As shown in Fig. 2 a—f, hMBP—activated T cells from
`three naive-to—therapy RRMS patients produced consistent
`amounts of leptin and up—regulated the ObR. The production of
`leptin was also confirmed with the measurement of immunoreac—
`tive leptin in the culture medium by a human-leptin-specific ELISA
`(Fig. 2g).
`
`Neutralization of Leptin or Its Receptor Inhibits T Cell Activation of
`hMBP-Specific T Cell Lines Derived from RRMS Patients. We measured
`the proliferative response against-hMBP on T cells from three
`naive-to-treatment RRMS patients and added either an anti-leptin
`or an anti-leptin-receptor blocking antibody to the culture medium
`(Fig. 211). We observed a significant reduction in the proliferative
`response of all three patients tested, ranging from 45% to 60%
`inhibition of proliferation (Fig. 211).
`
`in
`Inverse Correlation Between Serum~ Leptin and Circulating Tneg,
`Naive-to-Treatment RRMS Patients. The analysis of the immune
`phenotype was also performed on the peripheral blood of 31
`individuals from the naive-to—therapy RRMS patient population,
`selected on the basis of increase in serum leptin concentration (a
`serum leptin increase to 22.5—fold higher than the mean serum
`leptin observed in NIND and healthy controls). We compared these
`phenotypes with the immune phenotype of 27 healthy controls
`matched for age, sex, and BMI. The relative percentage and the
`absolute cell count per mm3 of the CD3‘, CD4‘, CD8‘, CD19*,
`CD3'CD16+CD56‘, and CD4+CD25+ TRegs subpopulations were
`performed (see Table 3, which is published as supporting informa-
`tion on the PNAS web site). Interestingly, naive-to—therapy RRMS
`patients, selected on the basis of their serum leptin increase, showed
`a significant reduction in the percentage and absolute number of
`TM, in the peripheral blood (Fig. 3a and Table 3), whereas no
`difference was observed in the frequency of the other cell subv
`populations (Table 3). TM, measurement in healthy controls was
`in agreement with that found in other studies (14). Regression
`analysis between serum leptin and the percentage of Tch, showed
`an inverse correlation in RRMS patients (Fig. 3b) but not in the
`controls (Fig. 3C). In vitro analysis Of ”FMS-mediated suppression in
`RRMS patients indicated a reduced ability to suppress T cell
`proliferation as compared with healthy controls (Fig. 3d), as
`reported in ref. 15. Moreover, the addition of leptin (100 ng/ml) to
`human '1"chS alone, or during coculture with CD4‘ CD25‘ effec~
`tors, did not alter significantly either proliferation or the suppressive
`capacity of Tchs (see Fig. 5 a and b, which is published as supporting
`information on the PNAS web site).
`
`ob/ob and db/db Mice Have Increased Tnegs. To analyze in more
`detail
`the effect of leptin on the generation of TM,
`in the
`periphery, we measured the effect of chronic leptin deficiency on
`the number of TRCE, in 0b/0b mice. These mice showed an increased
`frequency of TRCgS in lymphoid organs when compared with normal
`WT mice (10.4 : 3.7% vs. 4.7 i 1.7%, respectively; P < 0.02). In
`addition, we counted TRCgS in the lymphoid organs of db/db mice
`and, again, observed an increased percentage of TREE, when com-
`pared with db/+ heterozygote controls (13.9 : 1.9 vs. 7.9 i 0.9,
`respectively; P < 0.01). Finally, the suppressive capacity and phe—
`notype of TRegs from db/db mice were evaluated. No significant
`differences were observed in terms of either suppressive capacity or
`hyporesponsiveness of TRCg, (see Fig. 6 a—c, which is published as
`supporting information on the PNAS web site). In addition, ex~
`pression levels of FoxP3 in Tm, of 017/01) and rib/db mice were
`comparable to those in normal control mice (Fig. 6d).
`
`Matarese et at.
`
`a V
`
`A
`'3
`
`
`
`
`
`

`

`
`
`
`
`CTR-Ab ObR:Fc
`in viva treatment from day -1 to #1
`
`
`ill
`
`ill,
`
`
`
`
`
`Meanclinicalscore
`
`
`
`Percentageinitialbodyweight
`
`Fig. 4. Neutralization of leptin with ObR:Fc increases the number of TRegs
`and ameliorates the clinical course of R-EAE.
`(a) Treatment of R-EAE-
`susceptible SJL/J female mice with ObR:Fc induced a significant increase in the
`circulating TRegs- *, P = 0.01. (b) Mean clinical score (bars) and body weight
`(curves) of SJL/J female mice pretreated with the ObR:Fc (white bars and
`squares) or the CTR-Ab (black bars and squares) on d —1, d 0, and d +1 and
`immunized with the PLPiggzm on d 0. Statistical analyses of these data are
`summarized in Table 2. The data shown are from one representative experi-
`ment of two (r1 = 6 mice per group). t, P 2 0.01; H, P = 0.002.
`
`blocking ObR:Fc soluble chimera induced an increase of the
`percentage of TRC,gs in the periphery (Fig. 4a). To test whether this
`treatment could also modify the induction/progression of R-EAE,
`we pretreated SJL/J female mice with ObR:Fc chimera before
`immunization with the encephalitogenic peptide PLP139_151. The
`treatment was performed from d —1 to d +1 with i.p. injection of
`100 pg per day per mouse of ObR:Fc chimera dissolved in PBS (Fig.
`4b). ObR:Fc-treated mice showed a reduced peak clinical score, an
`improvement in disease relapses and progression, and a reduction
`in the percentage of body weight
`loss (Fig. 4b and Table 2).
`Moreover, a significant increase in body weight before/during the
`early phases of disease in ObR:Fc-treated mice (on d 9,
`the
`percentage of initial body weight
`in ObR:Fc-treated mice was
`112.4 ': 0.9% vs. 100.6 : 0.7% in control (CTR)—Ab-treated mice,
`P : 0.01) was observed, compared with a classical reduction in body
`
`weight preceding the onset of clinical symptoms in CTR-Abtreated
`mice In addition, ObR: Fc- treated mice showed very rapid reduc-
`tion of body weight after d 10 and a rapid recovery after d 13 of
`disease to a weight
`that was significantly higher than that of
`CTR-Ab-treated mice (Fig. 4b and Table 2). On the contrary,
`control mice showed a more stable body weight
`loss that was
`maintained over the disease course (Fig. 4b and Table 2). Finally,
`CNS inflammatory lesions were also significantly reduced in Ob—
`R:Fc-treated mice (Table 2). A significant increase in Tch5 was
`observed on d 15 of the disease course in mice pretreated with
`ObR:Fc (Table 2).
`
`Discussion
`
`In this report, we analyze the secretion of leptin in serum and CSF
`of naive-to—treatment RRMS patients in correlation with the se-
`cretion of IFN-y in CSF and the percentage of circulating Tchg. The
`data presented here provide evidence that a significant increase of
`leptin secretion occurs in the acute phase of MS and that this event
`positively correlates with the CSF production of IFN- y. Increased
`secretion is present in both the serum and CSF of RRMS patients
`and determines the loss of correlation between leptin and BMI (Fig.
`1 a and c). Moreover, the increase of leptin in the CSF is higher than
`that in the serum (a 5.6-fold increase in CSF leptin vs. a 18-fold
`increase in serum leptin, P = 00001, Table 1), possibly secondary
`to in situ synthesis of leptin in the CNS and/or an increased
`transport across the blood— brain barrier, upon enhanced systemic
`production. Indeed, the CSF-leptin/serum-leptin ratio, the CSF-
`leptin/serum--albumin ratio, and the CSF leptin index all signifi—
`cantly increase in RRMS patients when compared with NIND
`controls (Table 1).
`Recently, gene-microarray analysis of Th-1 lymphocytes and
`active MS lesions in humans revealed elevated transcripts of many
`genes of the neuroimmunoendocrine axis, including leptin (19, 20).
`Leptin’s transcript was also abundant in the gene—expression profile
`of human Th-I clones, demonstrating that the leptin gene is induced
`in and associated with polarization toward Th-l responses, com-
`monly involved in T cell-mediated autoimmune diseases such as MS
`(19, 20). We previously reported in situ leptin secretion by inflam»
`matory T cells and macrophages in active EAE lesions (3). Here, we
`show that autoreactive hMBP—specific T cells from RRMS patients
`can produce immunoreactive leptin and up—regulate the leptin
`receptor after activation (Fig. 2 a—f). possibly explaining, in part, the
`increased in situ CSF leptin levels in RRMS patients. Interestingly,
`both anti-leptin and anti—leptin-receptor blocking antibodies re-
`duced the proliferative responses of hMBP-specific T cell lines (Fig.
`2 h, j, and l), underscoring the possibilities of leptin-based inter-
`vention on this autocrine loop:
`Many questions need to be answered about whether and how
`TRCgs can regulate autoimmunity in humans. In animal models of
`autoimmune diseases, the role of TREE, has been demonstrated (21).
`More recently, a reduced function and/or generation of TRCg, in
`human autoimmune diseases such as systemic lupus erythematosus,
`type 1 diabetes, autoimmune polyglandular syndromc type II,
`
`Table 2. Effect of pretreatment with soluble ObR:Fc chimera on neurological impairment and percentage of CD4+CD25+ during active
`R-EAE induction with the PLP139.151 encephalitogenic peptide in SJL/J female mice
`
`Percentage of
`CD4*CD25+
`No. of
`Percentage of initial
`after
`inflammatory
`body weight at
`Peak clinical Average
`Day of onset
`lncidence,
`______«—___—_________....—__._.—.____—___—_———~—
`Antigen no./tota| (%)
`(range)
`score
`CDI"
`disease peak
`foci
`treatment
`Group of mice
`
`4.5 : 0.7
`308 : 1.8
`89.4 : 0:5
`42.7 t 7.9
`2.8 f 0.7
`8.1 I 0.4 (8—9)
`6/6 (100.0)
`PLPizem
`SlL/J CTR»Ab (d ‘1 to d+1)
`
`
`
`
`10.6 t 2.0 (8—13)6/6 (100.0)PLP1397151SJL/J ObR:Fc (d -1tO del) 11.3 : 4.3‘M1.9 t 0.7' 21.8 : 5.3‘ 108.2 f. 0.7‘ 15.0 :1.5‘
`
`
`
`
`
`The data shown are from one representative of two independent experiments shown in Fig. 4b. CTR»Ab, control Ab.
`*Curnulative disease index, sum of daily scores determined for each mouse of that group and averaged.
`'P = 0.01.
`‘P : 0.002.
`
`5154
`
`I www.pnas.org/‘cgi,I'doi/10.1073,r"pnas.0408995102
`
`Matarese et al.
`
`

`

`juvenile idiopathic arthritis, and MS has been described in refs.
`10—15. Recently, this reduction has'been shown, in RRMS, to be a
`functional defect of TRegg rather than a reduced number of Tchs in
`the periphery (15 ). To address whether leptin secretion could have
`an effect on TchS in RRMS patients, we measured the TchS
`frequency in the peripheral blood of nai‘ve-to—treatment RRMS
`patients selected on the basis of an increase in serum leptin to
`22.5—fold higher than levels measured in NIND and healthy
`controls. Here, we show that
`the average percentage and the
`absolute number of TchS in these RRMS patients were significantly
`lower than those of healthy controls (Fig. 3a and Table 3). No
`significant differences in CD3‘, CD4+, CD8+, CD19+, and
`CD3”CD16*CD56‘ cells were observed in either study group
`(Table 3). In addition, our functional data confirmed that, in our
`experimental conditions, RRMS patients showed a functional Tchs
`defect, confirming findings previously reported in ref. 15 (Fig. 3d).
`Administration of exogenous leptin to human Tch, or to suppres—
`sion assays did not alter hyporesponsiveness and suppressive ca—
`pacity (Fig. 5 a and b), suggesting that in vitro leptin is not
`responsible for impaired TchS function. Simple regression analysis
`showed an inverse correlation between systemic leptin concentra—
`tions and TRegs in the naive-to-treatment RRMS population (Fig. 3
`b and c). These data demonstrate an inverse relationship between
`leptin and TRegS in MS and may account for a reduced generation
`of TREES, at least early in the disease, in naive-to—treatment patients.
`Indeed, we hypothesize that, after therapy, these phenomena may
`be masked and overcome by therapy»induced effects. In fact, in the
`case of chronic leptin deficiency, such as in ob/ob mice, we found
`an increased number of circulating TM, and similar results were
`observed in db/db mice. This finding was also confirmed by
`experiments showing a higher recovery and percentage of TRCgs
`from R—EAE—susceptible SJL/J female mice treated with leptin—
`blocking ObR:Fc (Fig. 4a). Also, this pretreatment subsequently
`ameliorated R-EAE onset and progression (Fig. 4b and Table 2).
`The fact that TchS from db/db mice had a similar suppressive
`capacity and phenotype compared with Tch, from normal controls
`(Fig. 6 a—d) suggests that leptin does not affect in vitro suppressive
`funetion but, rather, in vivo expansion/proliferation of TRCES. Fur-
`ther studies need to address this point. Recent reports have shown
`increased secretion of serum leptin before relapses in RRMS
`patients during treatment with IFN-B and the capacity of leptin to
`enhance in vitro secretion of TNF—a, IL-6, and IL—10 by peripheral
`blood mononuclear cells of RRMS patients in the acute phase of
`the disease but not in patients in the stable phase (22, 23). In view
`of the above considerations, we suggest that, in MS, leptin may be
`part of a wider scenario in which several proinflammatory soluble
`factors may act in concert in driving the pathogenic (autoreactive)
`Th-l responses targeting neuroantigens (24). Recently, Hafler er al.
`
`(15) reported a decrease in the effector function and cloning
`frequency of TRegs from the peripheral blood of patients with MS.
`We show here that, in na'ive-to-therapy MS patients, not only the
`function but also the number of TRCgs
`is affected, and, more
`importantly, the finding inversely correlates with the concentration
`of serum leptin. It appears therefore that, early in the disease, the
`effects on Tchs in MS may be different from the effect observed
`after therapy has been initiated. Regarding the correlation with
`leptin,
`it is worth mentioning that strains of mice prone to the
`spontaneous development of autoimmune diseases, such as nono—
`bese-diabetic (NOD) and IL—2-deficient (IL-2””) mice, show re-
`duced frequency of Tchs in the periphery (9) associated with
`abnormal leptin responses due to increased serum leptin concen—
`trations (disproportionate to fat mass) (25, 26). NOD mice have
`higher basal serum leptin levels than normal
`age—,
`sex—, and
`fat-matched controls (25). IL—2‘/‘ mice are prone to spontaneous
`development of inflammatory bowel disease and other autoim—
`mune disorders (26). Whereas in normal mice, serum leptin de-
`creases with fat—mass loss, in IL—2‘/‘ mice there is a paradoxical rise
`in serum leptin compared with control mice, even after starvation,
`which reduces serum leptin (26). These data support the hypothesis
`that a disproportionate response in leptin secretion can correlate
`with a reduction in the periphery of the Tm,gs compartment in these
`two models.
`Because of the influence of leptin on food intake and metabo-
`lism, the findings reported here underscore the role of molecules at
`the interface between metabolism and immunity in the control of
`not only inflammation but also autoimmune reactivity (24, 27).
`Recently, molecules with orexigenic activity, such as ghrelin and
`neuropeptide Y (NPY), have been shown to mediate not only
`effects opposite to those of leptin on the hypothalamic control of
`food intake but also on peripheral immune responses (28, 29).
`Indeed, ghrelin blocks the leptin—induced secretion of proinflam-
`matory cytokines by human T cells (28), and NPY ameliorates the
`clinical course and progression of EAE (29). Given these consid-
`erations, we may envisage a situation in which the influenccs
`exerted by several metabolic regulators,
`including leptin, might
`broadly influence vital functions not limited to caloric tuning but,
`rather, affecting immune responses and the interaction of the
`individual with the environment. Although additional studies are
`needed, our data provide direct evidence of a negative association
`between leptin secretion and Tchs in the early stages of an
`autoimmune disease characterized by Th—1 autoreactivity, such
`as MS.
`
`This work is dedicated to the memory of Eugenia Papa. This work was
`supported by Fondazione Italiana Sclerosi Multipla Grants 2001/R/68
`and 2002/R/55 (to S.Z.) and by a National Institutes of Health grant (to
`A,L.C.).
`
`(St Matarcse, G. (2004) Nat. Rev. Immunul. 4, 371—379.
`1. La Cava, A.
`2. Matarcsc, G.. Di Giacomo, A., Sanna, V., Lord, G. M., Howard, ,1. K, Di Tuom, A., Bloom,
`S. R., lecblcr, R. 1., Zappacosta, S. & Fontana, S. (2001)]. Immunol. 166, 5909—5916.
`3. Sanna, V., Di Giacomo, A., La Cava, A., Lecblcr, R. 1., Fontana. 5., Zappacosta, S. &
`Malarcsc, G. (2003) J. Clin. Invest. 111, 241—250.
`4. Busso, N., 50, A., Chobaz-Pcclat, V., Morard, C., Martinez-Soriu, E., Talabot-Ayer, D. &
`Gabay, C. (2002)]. Immunol. 168, 875—882.
`_
`5. Sicgmund, 13., Lchr, H. A. & Fantuzzi, G. (2002) Gastroenlemlogy 122, 201172025.
`6 Siegmund, B.. Sennello. .1. A., Jones-Carson. 1.. Gamhnni-Robertson, F., Lchr, H A., Batra,
`A., Fedke, 1.. ZeitL. M.
`(i: Fantuzzi, G (2004) (£141 53, 9654972.
`7 Zamvll, S. S. St Slemmun, 1.. (199(0/1/11111 Rm‘
`lmmurml.
`11, 579-621
`8. Brown, A. M. & MeFarlin, D. (1981) Lab. Invest. 45, 278-281.
`‘) Sakaguchl. S. (21104)Arim/ Rm IH‘UYHHH), 22, 5.11562
`10. Kukreja, A.. Cost, 0., Marker, 1., Zliang, C. Sun, / LIn-Su, K., Ten, 5., Sanz, M., Exlcy,
`M., Wilson, B . ('l 11/. (2002) J. (Int. Invest. 109, 1317140.
`11. Baecher-Ailan, C. & Hafler, D. A. (2004)]. Exp. Med. 3, 273—276.
`12. Crispin, J C. Martinez, A. & Alcoccr-Varcla,1. (2003) J. Autoimmun. 21. 27L276.
`13. dc cher,1.M.,Weddcrhurn,L.R.,Taams,L.S.,Pate1,A.,Varsani,1—1.,chin. M., dc Jagcr,
`W., Pugayung, G., Giannoni, F., Rijkers, (1,211)]. (2004)]. Immunol. 172, 6435—6443.
`14. Kricgel, M. A., Lohmann, T.. Gabler, (7., Blank. N., Kalden,1. R. & Lorenz, H. M. (2004)
`J. Exp. Med. 9, 12854291.
`15. Viglictta, V., Baecher~Allan, C, Weincr, H. L. & Haflcr, D A. (2004) I Exp. Med. 5, 9717979
`16. McDonald, W. 1., Compston, A., Edan, G., Goodkin, D., Hartung, H. P., Luhlin, F. D.,
`McFarland. H. F., Paty, D. W., Polman, C. H., Reingold, S. C... 61 al. (2001)/im1. Neural.
`1, 1217127.
`
`l7. Pontieiellu, A., Pcrna, F., Sturkenboom, M. C.. Marcbctiello, 1., Bocchino, M. & Sanduzzi,
`A. (2001) Int. J. Tuberr. Lung Dir. 5, 114871155.
`18. Montanaro, D.. Sanna, V., Matarese, G., Lurby, B. 8., Racioppi, L., Carrieri, P. 13., Bruno.
`R., Davey, N. J., Zappacosta, S. & Fontana, S. (2001) Clin. Exp. Immunol 123, 2887203.
`19. Rogge, L.,Biancl1i.E., Biffi,

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