throbber
RESEARCH PAPER
`
`Multiple Sclerosis 2008; 14: 749–758
`
`Multiple sclerosis: glatiramer acetate induces
`anti-inflammatory T cells in the cerebrospinal fluid
`
`ALK Hestvik1, G Skorstad2, DA Price3, F Vartdal1 and T Holmoy1,2
`
`Glatiramer acetate (GA) is believed to induce GA-reactive T cells that secrete anti-inflammatory cyto-
`kines at the site of inflammation in multiple sclerosis (MS). However, GA-reactive T cells have not been
`established from the intrathecal compartment of MS patients, and intrathecal T cells may differ from T
`cells in blood. Here, we compared the phenotype of GA-reactive T cells from the cerebrospinal fluid
`(CSF) and blood of five MS patients treated with GA for 3-36 months, and in three of these patients
`also before treatment. From the CSF of these patients, all 22 T cell lines generated before and all 38 T
`cell lines generated during treatment were GA-reactive. GA treatment induced a more pronounced
`anti-inflammatory profile of GA-reactive T cell lines from CSF than from blood. While GA-reactive T cell
`clones from CSF were restricted by either human leukocyte antigen (HLA) -DR or HLA-DP, only HLA-
`DR restricted GA-reactive T cell clones were detected in blood. No cross reactivity with myelin proteins
`was detected in GA-reactive T cell lines or clones from CSF. These results suggest that a selected
`subset of GA-reactive T cells are present in the intrathecal compartment, and support an anti-inflam-
`matory mechanism of action for GA. Multiple Sclerosis 2008; 14: 749–758. http://msj.sagepub.com
`
`Key words: multiple sclerosis; immunology; glatiramer acetate; disease modifying therapies
`
`Introduction
`
`Glatiramer acetate (GA) has been shown to reduce
`both the relapse rate and the appearance of new
`central nervous system (CNS) lesions in multiple
`sclerosis (MS) [1,2]. GA is a polymer of the four
`amino acids most prevalent in myelin basic protein
`(MBP) and has been shown to bind several different
`human leukocyte antigen (HLA)-DR molecules [3].
`Antigen presenting cells (APCs) within or outside
`the CNS may be the primary target of GA-
`mediated immune modulation [4–6]. The mecha-
`nism of GA is thought to involve induction of
`anti-inflammatory GA-reactive T cells in secondary
`lymphoid organs [7]. Anergy induction of activated
`and potentially pathogenic T cells is thought to
`play a role as long-term GA treatment leads to a
`drop in the precursor frequency of GA-reactive
`T cells [8,9]. Within the CNS, GA-reactive T cells
`may cross-react with myelin proteins
`[10–12],
`dampen the inflammatory response by secretion of
`anti-inflammatory Th2 cytokines such as interleu-
`
`kin (IL)-4, IL-5, IL-10, and IL-13, [10,13,14], induce
`regulatory T cells [5], and stimulate reparative pro-
`cesses through secretion of neurotrophic factors
`[15–18].
`For these mechanisms to be relevant in MS, GA-
`reactive T cells must gain access to the CNS. Studies
`in mice have shown that GA-reactive T cells
`induced in the periphery penetrate the blood-brain
`barrier [13,14], and human GA-reactive T cells have
`been shown to migrate across an artificial blood-
`brain barrier in vitro [19]. However, because of diffi-
`culties in investigating migration of human T cells
`in vivo, current knowledge of human GA-reactive
`T cells stems from analyses conducted with lym-
`phocytes derived from blood.
`Studies in MS and other immune-mediated dis-
`eases have shown that T cells from the diseased
`organ may differ from T cells in blood [20–23]. To
`understand the effects of GA in MS, it is therefore
`important to investigate GA-reactive T cells from
`the intrathecal compartment. The aim of the pres-
`ent study was to compare GA-reactive T cells from
`
`1Faculty of Medicine, Institute of Immunology, Rikshospitalet-Radiumhospitalet, Oslo, Norway
`2Department of Neurology, Ullevål University Hospital, Oslo, Norway
`3Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
`Correspondence to: Anne Lise K Hestvik, Institute of Immunology, University of Oslo, 0027 Oslo, Norway.
`Email: a.l.k.hestvik@medisin.uio.no
`Received 9 August 2007; revised 2 December 2007; accepted 18 January 2008
`
`© SAGE Publications 2008
`Los Angeles, London, New Delhi and Singapore
`
`10.1177/1352458508089411
`
`Page 1 of 10
`
`YEDA EXHIBIT NO. 2059
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`750
`
`ALK Hestvik et al.
`
`blood and cerebrospinal fluid (CSF) in terms of
`cytokine profile, HLA restriction,
`and cross-
`recognition properties before and after initiation of
`GA treatment.
`
`Materials and methods
`
`Patients
`
`Five patients (four women and one man) with
`relapsing-remitting MS (Expanded Disability Status
`Scale 1–2) gave informed consent to participate in
`this study. Study participants were treated by neu-
`rologists with no other involvement in the study
`and had received 20 mg GA daily for 0–36 months
`at inclusion. The study was approved by the local
`ethics committee.
`
`Antigens
`
`GA (copaxone©) was obtained from Teva Pharma-
`ceuticals Ltd (London, UK). In all, 531 15-mer pep-
`tides, overlapping by 10 amino acids spanning
`the complete sequences of MBP (18.5 kDa and
`21.5 kDa isoforms), protelipid protein (PLP), myelin-
`oligodendrocyte glycoprotein (MOG;
`including
`the β2 isoform), myelin-associated oligodendrocytic
`basic
`protein, myelin-associated
`glycoprotein,
`oligodendrocyte myelin glycoprotein, αβ-crystallin,
`S100β,
`and
`2′,
`3′-cyclic
`nucleotide
`3’-
`phosphodiesterase (partly a gift from N. Karandikar,
`UT Southwestern Medical Center, Dallas, Texas,
`USA) were synthesized using standard techniques
`(BioSynthesis, Texas, USA) at 44.8–100% purity
`(0.5% were below 50% pure and 12% were above
`90% pure) [24]. Peptides were dissolved in dimethyl
`sulfoxide (DMSC) at a concentration of 50 mg/mL
`and tested in pools of 8–10 peptides; each peptide
`was tested at a concentration of 2.5 μM. A chymo-
`trypsin digest of gluten extracted from wheat flour
`(gift from O. Molberg, Institute of Immunology, Rik-
`shospitalet
`Radiumhospitalet, Oslo, Norway),
`human MBP (Chemicon, California, USA), and a pep-
`tide from glutamic acid decarboxylase (GAD57–72)
`served as control antigens.
`
`Generation of T cell lines and T cell clones
`
`In all, 40,000–160,000 cells were obtained from
`23 mL CSF collected by lumbar puncture. No bleed-
`ing was observed during the procedures. To further
`avoid blood contamination, the first 2 mL of CSF
`were discharged. To minimize differences related
`to in vitro conditions, T cell lines from CSF and
`blood were cultured in parallel using identical pro-
`
`cedures. Briefly, 1 × 105 peripheral blood mononu-
`clear cells (PBMC) or 4000–6000 CSF cells were
`incubated with 105 irradiated (25 Gy) autologous
`PBMC, which had been pre-incubated overnight
`with 50 or 200 μg/mL GA. Cells were stimulated
`with 10 IU/mL IL-2 (RnDSystems, Minnesota, USA)
`on day 7 and restimulated on day 14 with 105 irra-
`diated autologous PBMC and 50 or 200 μg/mL GA.
`The T cell lines were expanded with irradiated het-
`erologous PBMC, 1 μg/mL phytohemagglutinin
`(Remel, Kansas, USA), and 10 IU/mL IL-2 on day
`20 and tested for specificity and cytokine produc-
`tion on day 28 with autologous PBMC preincubated
`overnight with 50 or 200 μg/mL GA. T cell cloning
`and determination of the HLA-isotype presenting
`GA were performed using procedures and monoclo-
`nal antibodies described previously [25].
`
`Proliferation assays
`
`T cells were tested by incubating 105 T cells sus-
`pended in 200 μL medium with 105 irradiated autol-
`ogous PBMC, which had been pre-incubated with
`antigen overnight in 100 μL medium. T cells were
`cultured for 72 h and proliferation tested on the
`basis of 3H-thymidine incorporation as described
`previously [25]. A proliferative response was scored
`as positive if counts per minute (cpm) of stimulated
`wells minus cpm of unstimulated wells (Δcpm)
`exceeded 1000 and the stimulatory index (SI)
`exceeded 3 [23].
`
`Cytokine measurement
`
`Supernatants of T cells were frozen at −80°C for
`48 h after antigen stimulation. Concentrations of
`IL-4, IL-5, IL-10, IL-13, IL-17, tumor necrosis factor
`(TNF)-α, and interferon (IFN)-γ were measured with
`a custom-made multiplex antibody bead kit (Bio-
`source International
`Inc., Camarillo, California,
`USA) for Luminex™, according to the manufac-
`turer’s recommendations. The detection level for
`all cytokines was 3–5 pg/mL. Unstimulated T cells
`incubated with irradiated PBMC in the absence of
`antigen served as control. Irradiated PBMC incu-
`bated with antigen overnight produced no detect-
`able levels of the cytokines. Cytokine production
`was calculated as cytokine concentrations in super-
`natants from antigen-stimulated cells (T + APC + GA)
`minus unstimulated cells (T + APC).
`
`Statistics
`
`Cytokine ratios were compared with a two-way
`ANOVA analysis with patients as random effect in
`
`Multiple Sclerosis 2008; 14: 749–758
`
`http://msj.sagepub.com
`
`Page 2 of 10
`
`YEDA EXHIBIT NO. 2059
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Statistical Package for the Social Sciences (SPS) version
`15 (SPSS Inc., Chicago, Illinois, USA). To correct for
`deviation from the normal distribution, data were
`log transformed before analysis. The proportion of
`Th1-biased and Th2-biased GA-reactive T cell lines
`was analyzed using generalized estimating equation
`regression models in Statistical Analysis System (SAS)
`version 9.1 (SAS Inc., Cary, North Carolina, USA).
`
`Results
`
`GA-reactive T cells are present in the CSF and blood
`before and during GA treatment
`
`To establish whether GA-reactive T cells are present
`in the CSF of patients before and after initiation of
`treatment, 22 T cell lines from CSF and 36 T cell
`lines from blood were generated from patients 1–3
`before they started GA treatment, and 28 T cell
`lines from CSF and 31 T cell lines from blood of
`the same patients were generated after 3–6 months
`on treatment. All T cell lines generated both before
`and during treatment from both blood and CSF
`responded vigorously to GA both by proliferation
`
`GA-reactive T cells from the CSF of MS patient
`
`751
`
`and cytokine production (Figure 1). Although the
`number of CSF cells is too low to allow calculation
`of the frequency of GA-specific T cells, the observa-
`tion that all T cell lines were GA-reactive indicates
`that the precursor frequency of GA-specific T cells
`in CSF both before and after initiation of GA treat-
`ment exceeds 1:5000.
`
`Th2-bias is more pronounced in GA-reactive T cell
`lines from CSF than from blood
`
`To establish cytokine profiles of GA-reactive T cells,
`concentrations of pro- and anti-inflammatory cyto-
`kines were measured in supernatants of proliferat-
`ing GA-reactive T cell lines. Using an IL-5/IFN-γ
`ratio of 1 as the cut-off between a predominant
`Th1 versus Th2 phenotype, 13 of 22 GA-reactive
`T cell lines from CSF and 11 of 36 GA-reactive
`T cell lines from blood were Th2-biased before treat-
`ment. After treatment, the proportion of predomi-
`nantly Th2-biased GA reactive T cell lines from the
`same patients was 25 of 28 (CSF) and 21 of 31
`(blood). Thus, short-term GA-treatment induced
`an increase in the ratio of Th2-biased cell lines in
`both CSF and blood. The increase was only
`
`Figure 1 Glatiramer acetate (GA)-reactive T cell lines (TCL) can be generated from the CSF of GA-treated and treatment
`naive individuals. Proliferative responses (Δcpm) against four-fold serial dilutions of GA from 800–0.2 μg/mL GA in three rep-
`resentative CSF T cell lines from patient 1 generated before GA-treatment (CSF TCL 1–3) and after 3 months on GA treatment
`(CSF TCL 4–6) are shown (A). None of the T cell lines displayed any proliferation in response to 20–0.003 μg/mL myelin basic
`protein or gluten in the same experiment (data not shown). Production of IL-5 and IFN-γ is shown for all cell lines (B).
`
`http://msj.sagepub.com
`
`Multiple Sclerosis 2008; 14: 749–758
`
`Page 3 of 10
`
`YEDA EXHIBIT NO. 2059
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`752
`
`ALK Hestvik et al.
`
`significant in the CSF (P = 0.03), where a shift
`towards almost total dominance of Th2-biased GA-
`reactive T cells was observed.
`No consensus exists regarding the threshold ratios
`that define human T cells as Th1 or Th2. In addition
`to comparisons based on a potentially arbitrary clas-
`sification into Th1-biased and Th2-biased T cell lines
`it is therefore also important to compare absolute
`ratios of pro- and anti-inflammatory cytokines pro-
`duced by GA-reactive T cell lines from blood and
`CSF before and during GA treatment. Figure 2
`depicts the secretion of IL-5, IL-10, and IL-13 com-
`pared with IFN-γ produced by GA-reactive T cell
`
`lines before and during treatment for patients 1–3.
`Results for the secretion of IL-5, IL-10, and IL-13
`compared with IFN-γ and TNF-α in patients 1–3 com-
`bined are given in Figure 3. These results show that i)
`in T cells from CSF, GA treatment induced a highly
`significant increase in the secretion of IL-5, IL-10,
`and IL-13 compared with IFN-γ and TNF-α; ii) in T
`cells from blood, GA-treatment induced a highly sig-
`nificant increase in the secretion of IL-5, IL-10, and
`IL-13 compared with IFN-γ but not compared with
`TNF-α; iii) after 3–6 months on GA treatment, there
`is a prominent and significant Th2-polarization of
`GA-reactive T cells from both CSF and blood,
`
`Figure 2 Short-term glatiramer acetate (GA) treatment induces a Th2-bias of GA-reactive T cells, which is most prominent in
`CSF. Each data point represents the indicated cytokine ratio for individual GA-reactive T cell lines before and after 3 months
`(patient 1) or 6 months (patient 2 and 3) of treatment.
`
`Multiple Sclerosis 2008; 14: 749–758
`
`http://msj.sagepub.com
`
`Page 4 of 10
`
`YEDA EXHIBIT NO. 2059
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`GA-reactive T cells from the CSF of MS patient
`
`753
`
`Figure 3 Glatiramer acetate (GA) treatment induces a pronounced and significant increase in the ratio between anti- and
`pro-inflammatory cytokines produced by GA-reactive T cells from CSF and blood. GA-reactive T cell lines were established
`from CSF and blood of patients 1–3 before and 3–6 months after initiation of GA treatment. Each data point represents the
`indicated cytokine ratio for one GA-reactive T cell
`line 48 h after GA stimulation. Lines represent mean ratios. *P < 0.05;
`**P < 0.01; ***P < 0.005. The absolute cytokine concentrations (pg/mL, mean ± SD) in all cell lines (CSF and blood, before
`and after combined) were 2339, 3045 (interferon [IFN-γ]), 1981, 2703 (tumor necrosis factor [TNF-α]), 3024, 4209 (IL-5),
`1188, 1385 (IL-10), and 3680, 3916 (IL-13). The mean absolute value for unstimulated cells, all cytokines combined was
`15 pg/mL, SD = 31 pg/mL.
`
`which is more pronounced in T cells from CSF. The
`Th2-polarization of GA-reactive T cells was evident
`in each of the three patients.
`IL-17 has recently been identified as an impor-
`tant pro-inflammatory cytokine [26]. In CSF, IL-17
`production was detected in 5 of 21 GA-reactive
`T cell lines during GA treatment compared with 10
`of 28 before treatment. In blood, IL-17 production
`was detected in 11 of 31 GA-reactive T cell lines dur-
`ing GA treatment compared with 18 of 37 before
`treatment. These differences were not significant
`(results not shown).
`Patients 1–3 had only received GA for 3–6 months.
`To assess the impact of prolonged treatment, we estab-
`lished 10 GA-reactive T cell lines from CSF and 23 GA-
`reactive T cell lines from blood obtained from two
`patients who had received GA for 24 and 36 months
`(patients 4 and 5). Interestingly, GA-reactive T cell
`lines from both CSF and blood of these patients dis-
`played a significantly stronger Th2-bias than GA-
`reactive T cells from the short-term treated patients
`
`(Figure 4a). Although based on few patients, these
`results
`suggest
`that
`long-term treatment
`further
`enhances the Th2-polarization of GA-reactive T cells,
`which is most pronounced in CSF.
`Combining results for all GA-reactive T cell lines
`generated after initiation of GA treatment from
`patients 1–5 further confirms that GA-reactive
`T cells from CSF display a more pronounced anti-
`inflammatory profile than GA-reactive T cells from
`the blood (Figure 4b). In sum, the mean IL-5/IFN-γ
`ratio of GA-reactive T cells from CSF was 35.8 (95%
`CI: 20.4–51.6), compared with 9.6 (95% CI:
`6.3–12.8)
`for GA-reactive T cells
`from blood
`(P < 0.005).
`
`GA-specific T cells from CSF are restricted by either
`HLA-DR or HLA-DP
`
`To investigate the characteristics of individual GA-
`specific T cells, we cloned GA-reactive T cell lines
`
`http://msj.sagepub.com
`
`Multiple Sclerosis 2008; 14: 749–758
`
`Page 5 of 10
`
`YEDA EXHIBIT NO. 2059
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`754
`
`ALK Hestvik et al.
`
`Figure 4 Glatiramer acetate (GA)-reactive T cell lines from patients treated long-term (patients 4 and 5) display more pro-
`nounced Th2-bias than GA-reactive T cell
`lines from patients treated short-term (patients 1–3) with GA (A). Following
`3–36 months of GA treatment, GA-reactive T cells from the CSF of patients 1–5 display a more pronounced Th2-bias than
`T cells from blood (B). Concentrations of cytokines were measured in supernatants of proliferating GA-reactive T cell lines
`48 h after stimulation with GA. Each data point represents the IL-5/IFN-γ ratio for one cell
`line. *P < 0.05; **P < 0.01;
`***P < 0.005. The absolute IFN-γ and IL-5 concentrations (pg/mL, mean/SD) for all CSF and blood T cell lines from patient 4
`and 5 were 800, 1253 and 5361, 1874, respectively (For patients 1–3, see legend Figure 3).
`
`from patients 4 and 5 by limiting dilution. The
`cloning frequency was less than 5% making it
`more than 95% probable that each cell culture was
`monoclonal [27]. Forty-eight of 90 T cell clones
`from patient 4 and 154 of 379 T cell clones
`from patient 5 specifically recognized GA (mean
`Δcpm = 130239 and mean SI = 341 for 20 tested
`clones). Proliferative responses of representative
`T cell clones with and without HLA-blocking anti-
`bodies and HLA restriction are shown in Figure 5.
`Interestingly, all 10 tested T cell clones from blood
`were restricted by HLA-DR, whereas HLA-DP
`restricted T cells were cloned from CSF of both
`patients. One of five CSF T cell clones from patient
`4 and three of five CSF T cell clones from patient 5
`were restricted by HLA-DP.
`
`Cross-reactivity with myelin proteins
`
`GA-reactive T cells from mice and human blood
`have been reported to cross-recognize GA and mye-
`lin proteins, especially MBP [10,12,28,29]. To our
`knowledge, cross-reactivity in human T cells has
`not been confirmed at a clonal level. To investigate
`whether cross-reactivity can be detected at a clonal
`level, 20 GA-specific T cell clones (10 from CSF and
`10 from blood) from patient 4 and 5 were tested for
`proliferation against 531 peptides spanning most
`myelin proteins suggested to be associated with
`MS [30] and for cytokine production against 146
`peptides spanning MBP, MOG, and PLP. Because all
`T cell lines previously had produced either IFN-γ or
`IL-5 in response to GA, these two cytokines were
`used to assess cross-reactivity at the cytokine level.
`
`None of the 20 T cell clones showed any signs of
`proliferation or cytokine production in response to
`the peptide library. The T cells responded vigor-
`ously to GA in the same experiment (mean Δcpm:
`64,098; mean IFN-γ concentration 3400 pg/mL and
`mean IL-5 concentration 2330 pg/mL). The T cell
`clones were also tested against complete MBP.
`MBP did not elicit any proliferation, but we were
`able to detect a small amount of IFN-γ production
`in one T cell clone from blood (10 pg/mL elicited by
`MBP versus 350 pg/mL elicited by GA). MBP did not
`elicit any detectable cytokine production in T cell
`clones from CSF.
`GA-reactive T cell clones may represent a biased
`subpopulation of the total GA-reactive T cell reper-
`toire. To test if cross-reactive T cells were repre-
`sented in the polyclonal GA-reactive T cell lines,
`all 117 GA-reactive T cell lines from patients 1–3
`were tested for cross-reactivity against complete
`MBP. Neither MPB nor the control protein (gluten)
`elicited any detectable proliferation or cytokine
`production. The T cells responded vigorously to
`GA in the same experiment (mean IL-5 concen-
`tration 3900 pg/mL; mean IFN-γ concentration
`1880 pg/mL; mean Δcpm > 100,000).
`
`Discussion
`
`In the current study, we examined GA-reactive T
`cells from the CSF of humans and provided longitu-
`dinal information about the intrathecal immune
`response against GA at the clonal level. The princi-
`pal findings are that (i) GA-reactive T cells are pres-
`ent in the intrathecal compartment of MS patients
`
`Multiple Sclerosis 2008; 14: 749–758
`
`http://msj.sagepub.com
`
`Page 6 of 10
`
`YEDA EXHIBIT NO. 2059
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`GA-reactive T cells from the CSF of MS patient
`
`755
`
`Figure 5 Glatiramer acetate (GA)-specific T cells cloned from CSF and blood of patients treated long-term with GA (patients
`4 and 5) display divergent HLA-restriction. Proliferative responses (in Δcpm) against GA and a control peptide GAD57–72 of
`two representative CSF T cell clones are given in Figure 5A. Figure 5B shows proliferative responses to GA (in cpm) of the
`same T cell clones with or without monoclonal antibodies against HLA- DR, -DQ, -DP and HLA class I. Medium control repre-
`sents T cells and antigen-presenting cells in the absence of GA.
`
`before and after initiation of GA-treatment; (ii) GA
`treatment induces a Th2-bias of GA-reactive T cells
`from CSF and blood; and iii) following treatment,
`the Th2-bias is more pronounced in GA-reactive T
`cells from CSF than from blood. These results sup-
`port an anti-inflammatory T cell-mediated mecha-
`nism of action for GA.
`Resting T cells are not able to penetrate the intact
`blood-brain barrier, whereas activated T cells seem
`to gain access to the CNS regardless of their specific-
`ity [31]. The finding that GA-reactive T cells were
`present in the CSF before treatment, therefore,
`suggests that some GA-reactive T cells exist in an
`activated state in the absence of treatment. Accord-
`ingly, previous studies have shown high frequen-
`cies of GA-reactive memory T cells in the blood of
`treatment-naive individuals [32,11].
`GA is quickly degraded upon subcutaneous
`administration, making it unlikely that GA reaches
`the CNS in sufficient concentrations to induce Th2-
`polarization within the CNS [33]. Although GA-
`
`reactive T cells were present in the CSF before treat-
`ment, the observation that GA treatment induced
`an increasing Th2-polarization in both CSF and
`blood is compatible with the notion that a Th2-
`shift is induced in the periphery, and that a subpop-
`ulation of GA-reactive T cells subsequently gains
`access to the CNS. An alternative or additional
`explanation is the migration of type II monocytes
`to the CNS and induction of Th2 T cells at the site
`of inflammation [5,6]. The Th2-shift in blood of the
`short-term treated patients may be less pronounced
`than previously observed in long-term treated
`patients [12], and it is therefore possible that the
`more pronounced Th2 induction observed in CSF
`is a transient phenomenon. However, the marked
`Th2-bias in CSF compared with blood in the two
`long-term treated patients suggests that there is a
`persistent difference in favor of a more pronounced
`Th2-bias in CSF compared with blood.
`intrathecal
`Several observations
`suggest
`that
`GA-reactive T cells are not a random selection of
`
`http://msj.sagepub.com
`
`Multiple Sclerosis 2008; 14: 749–758
`
`Page 7 of 10
`
`YEDA EXHIBIT NO. 2059
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`756
`
`ALK Hestvik et al.
`
`GA-reactive T cells from blood. First, GA-reactive
`T cells from CSF were more Th2-biased than
`GA-reactive T cells from blood, possibly reflecting
`that GA induces Th2-skewing earlier in CSF than
`in blood or a preferential migration of strongly
`Th2-biased T cells to the CSF. Second, GA treatment
`imposed a more pronounced reduction in TNF-α
`secretion in GA-reactive T cells from CSF than
`from blood. Third, GA-specific T cells from the
`CSF were restricted by either HLA-DP or HLA-DR,
`whereas only HLA-DR restricted T cells were
`detected in blood. HLA restriction of GA-specific
`T cells from blood has previously been extensively
`studied, and HLA-DP restriction has to our knowl-
`edge not been reported [3,34]. The finding of
`HLA-DP restricted GA-reactive T cells in CSF was,
`therefore, highly unexpected and supports that a
`subset of GA-reactive T cells selectively accesses
`the intrathecal compartment.
`In this study, we only detected GA-reactive CD4+
`T cells. GA may also have an effect on CD8+ T cells
`as shown by clonal expansion of GA-reactive CD8+
`T cells from the blood of patients receiving GA [35].
`This difference may be related to different method-
`ological approaches. There are few reports on
`cloned GA-specific T cells [34] and to our knowl-
`edge no GA-specific CD8+ T cell clones have been
`established. The method applied in this study has
`previously allowed generation of T cell lines and
`clones with several specificities from the limited
`number of CD4+ T cells in CSF of MS patients
`[22,36], but there is currently no adequate method
`to establish antigen specific CD8+ T cell lines and
`clones from CSF and we can, therefore, not exclude
`that CD8+ GA-reactive T cells are present in the
`intrathecal compartment.
`The dissimilarities between GA-reactive T cells
`from CSF and blood can hardly be explained by dif-
`ferences in APCs or other in vitro factors because
`identical procedures were used to generate T cells
`from both compartments. The mechanisms recruit-
`ing a particular subset of GA-reactive T cells to the
`CSF are not clear, but several possibilities exist.
`Restimulation of GA-reactive T cells by cross-
`recognition of myelin antigens within the CNS
`could explain expansion of a particular subset of
`GA-reactive T cells within the CNS [13]. However,
`GA-reactive T cells from blood and CSF in this
`study displayed almost no cross-reactivity. Previous
`studies have shown that a proportion of human
`GA-reactive T cell lines from blood cross-recognize
`MBP [11,32]. Moreover, GA treatment has been
`found to induce degenerated T cell responses with
`broad
`cross-reactivity,
`suggesting
`that
`cross-
`recognition might also occur outside the CNS [12].
`Cross-recognition of myelin proteins can, therefore,
`hardly explain the differences between GA-reactive
`T cells from CSF and blood. Another possibility is
`
`that a selected subpopulation of GA-reactive T cells
`penetrates the blood-brain barrier. T cells from CSF
`display a more activated phenotype than T cells
`from blood [20,23], and differences in cytokine pro-
`files could reflect differences in activation status.
`However, a putative difference in activation status
`between GA-reactive T cells in blood and CSF
`would be expected to decline during prolonged
`treatment and could hardly explain the selective
`finding of HLA-DP restricted T cells in CSF. Further
`studies of expression of chemokine receptors and
`adhesion molecules on T cells from blood and CSF
`may contribute to our understanding of
`these
`intriguing findings. The expression patterns of
`such molecules are, however, sensitive to in vitro
`changes during cell culture and are difficult to
`establish directly ex vivo with the limited numbers
`of T cells available from CSF.
`
`Several studies on T cells from mice and human
`blood have shown cross-reactivity between GA and
`MBP. In one study, some GA-reactive T cell lines
`proliferated vigorously against MBP [32]. In other
`studies, evidence of cross-reactivity has been absent
`or limited to cytokine secretion [10,12,28,29,37]. In
`this study, we primarily studied cross-reactivity
`between GA and myelin proteins at a clonal level,
`whereas previous studies have studied polyclonal
`T cell lines [11,12]. The strict approach adopted
`here may have contributed to the finding of very
`limited cross-reactivity in this study. By definition,
`cross-reactivity of T cells means that one T cell
`receptor
`recognizes two different antigens and
`must, therefore, be established at a clonal
`level
`[38,39]. Because of limited cell numbers,
`it was
`only possible to test the extensive panel of myelin
`protein peptides at one concentration, and cross-
`reactivity between GA and myelin protein peptides
`with low affinity for the relevant HLA molecule or
`GA-reactive T cell receptor can, therefore, not be
`excluded. It must, however, be kept in mind that
`only 10–17% of polyclonal GA-reactive T cell lines
`from MS patients cross-recognized MBP in previous
`studies
`[11,32]. The limited finding of cross-
`reactivity at the clonal
`level
`in this study was,
`therefore, not unexpected and agrees with recent
`findings showing that cross-reactivity of adoptively
`transferred T cells
`from GA-treated mice was
`not necessary for the therapeutic benefit in recipi-
`ent mice [5]. These findings support previous
`suggestions that GA may also be effective in inflam-
`matory diseases outside the CNS [40,41].
`
`an anti-
`support
`In summary, our data
`inflammatory mechanism of action for GA at the
`site of the pathogenic process in MS and highlight
`the need to study intrathecal T cells in MS patients.
`
`Multiple Sclerosis 2008; 14: 749–758
`
`http://msj.sagepub.com
`
`Page 8 of 10
`
`YEDA EXHIBIT NO. 2059
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Acknowledgements
`
`The project has received financial support from
`Sanofi Aventis, The Odd Fellow Organization of
`Norway, The Multiple Sclerosis Society of Great
`Britain and Northern Ireland (grant 589–00), The
`National MS Society, and The National Institute of
`Health (grant AI53439 and AI65463). Professor
`Magne Thoresen at Department of Biostatistics at
`the University of Oslo is acknowledged for statisti-
`cal support. Nitin J. Karandikar at UT Southwestern
`Medical Center, Dallas, USA is acknowledged for
`providing synthetic peptides and purity data.
`Danny Douek is acknowledged for provision of
`reagents for the peptide synthesis.
`
`Reference
`
`1. Johnson, KP, Brooks, BR, Cohen, JA, et al. Copolymer 1
`reduces relapse rate and improves disability in relapsing-
`remitting multiple sclerosis: results of a phase III multi-
`center, double-blind placebo-controlled trial. The Copol-
`ymer 1 Multiple Sclerosis Study Group. Neurology 1995;
`45: 1268–1276.
`2. Johnson, KP, Brooks, BR, Cohen, JA, et al. Extended use
`of glatiramer acetate (Copaxone) is well tolerated and
`maintains its clinical effect on multiple sclerosis relapse
`rate and degree of disability. Copolymer 1 Multiple Scle-
`rosis Study Group. Neurology 1998; 50: 701–708.
`3. Fridkis-Hareli, M, Strominger, JL. Promiscuous binding of
`synthetic copolymer 1 to purified HLA-DR molecules. J
`Immunol 1998; 160: 4386–4397.
`4. Sanna, A, Fois, ML, Arru, G, et al. Glatiramer acetate
`reduces lymphocyte proliferation and enhances IL-5 and
`IL-13 production through modulation of monocyte-
`derived dendritic cells in multiple sclerosis. Clin Exp
`Immunol 2006; 143: 357–362.
`5. Weber, MS, Prod’homme, T, Youssef, S, et al. Type II
`monocytes modulate T cell-mediated central nervous
`system autoimmune disease. Nat Med 2007; 13: 935–943.
`6. Kim, HJ, Ifergan, I, Antel, JP, et al. Type 2 monocyte and
`microglia differentiation mediated by glatiramer acetate
`therapy in patients with multiple sclerosis. J Immunol
`2004; 172: 7144–7153.
`7. Ziemssen, T, Schrempf, W. Glatiramer acetate: mechan-
`isms of action in multiple sclerosis. Int Rev Neurobiol
`2007; 79: 537–570.
`8. Ragheb, S, Abramczyk, S, Lisak, D, Lisak, R. Long-term
`therapy with glatiramer acetate in multiple sclerosis:
`effect on T cells. Mult Scler 2001; 7: 43–47.
`9. Chen, M, Conway, K, Johnson, KP, Martin, R, Dhib-
`Jalbut, S. Sustained immunological effects of Glatiramer
`acetate in patients with multiple sclerosis treated for over
`6 years. J Neurol Sci 2002; 201: 71–77.
`10. Aharoni, R, Teitelbaum, D, Sela, M, Arnon, R. Copolymer
`1 induces T cells of the T helper type 2 that crossreact
`with myelin basic protein and suppress experimental
`autoimmune encephalomyelitis. Proc Natl Acad Sci U S A
`1997; 94: 10821–10826.
`11. Neuhaus, O, Farina, C, Yassouridis, A, et al. Multiple scle-
`rosis: comparison of copolymer-1-reactive T cell lines
`from treated and untreated subjects reveals cytokine
`shift from T helper 1 to T helper 2 cells. Proc Natl Acad
`Sci U S A 2000; 97: 7452–7457.
`
`GA-reactive T cells from the CSF of MS patient
`
`757
`
`12. Duda, PW, Schmied, MC, Cook, SL, Krieger, JI, Hafler,
`DA. Glatiramer acetate (Copaxone) induces degenerate,
`Th2-polarized immune responses in patients with multi-
`ple sclerosis. J Clin Invest 2000; 105: 967–976.
`13. Aharoni, R, Teitelbaum, D, Leitner, O, Meshorer, A, Sela,
`M, Arnon, R. Specific Th2 cells accumulate in the central
`nervous system of mice protected against experimental
`autoimmune encephalomyelitis by copolymer 1. Proc
`Natl Acad Sci U S A 2000; 97: 11472–11477.
`14. Aharoni, R, Meshorer, A, Sela, M, Arnon, R. Oral treat-
`ment of mice with copolymer 1 (glatiramer acetate)
`results in the accumulation of specif

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