throbber
Journal of the Neurological Sciences 201 (2002) 71 – 77
`
`www.elsevier.com/locate/jns
`
`Sustained immunological effects of Glatiramer acetate in patients with
`multiple sclerosis treated for over 6 years
`
`M. Chen a, K. Conway a, K.P. Johnson a, R. Martin b, S. Dhib-Jalbut a,c,*
`
`aUniversity of Maryland School of Medicine, Baltimore, MD 21201, USA
`bNeuroimmunology Branch, NINDS, National Institutes of Health, Bethesda, MD 20892, USA
`cBaltimore VA Medical Center, Baltimore, MD 21201, USA
`
`Received 6 February 2002; received in revised form 9 April 2002; accepted 10 June 2002
`
`Abstract
`
`The availability of a group of multiple sclerosis (MS) patients at the University of Maryland, who had participated in the pivotal
`CopaxoneR trial in the early 1990s, provided an opportunity to examine the long-term immunologic effects of Glatiramer acetate (GA)
`treatment in MS. Forty-eight GA-reactive T-cell lines (TCL) were generated from 10 MS patients who have been receiving GA treatment for
`6 – 9 years. Proliferative responses, cytokine production, and cross-reactivity with myelin basic protein (MBP) and the MBP im-
`munodominant peptide 83 – 99 were compared to responses obtained from 10 MS patients who were tested pretreatment and after a shorter
`period of treatment ranging from 1 to 10 months. The results indicate that while long-term treatment with GA results in a 2.9-fold decrease in
`the estimated precursor frequency of GA-reactive T-cells, the sustained response to GA remains Th2-biased and in part cross-reactive with
`MBP and MBP (83 – 99) as measured by proliferation and cytokine release assays. The results indicate that despite a drop in the precursor
`frequency of GA-reactive T-cells with long-term treatment, the sustained response remains predominantly Th2-biased and cross-reactive with
`MBP, which is consistent with the anti-inflammatory effects of the drug and bystander suppression.
`D 2002 Elsevier Science B.V. All rights reserved.
`
`Keywords: Multiple sclerosis; Glatiramer acetate; CopaxoneR; Immune deviation; Immunotherapy
`
`1. Introduction
`
`Glatiramer acetate (GA) (CopaxoneR), formerly known
`as Copolymer-1, is a synthetic amino acid copolymer that
`consists of four amino acids: L-alanine, L-lysine, L-glutamic
`acid, and L-tyrosine in fixed molar ratio and an average
`molecular weight of 4.7 – 11 kDa. The molecule was found
`to inhibit experimental allergic encephalomyelitis (EAE),
`and was subsequently developed for treating patients with
`relapsing – remitting multiple sclerosis (MS) [1 – 4]. When
`given subcutaneously in daily doses of 20 mg, the drug
`reduces the relapse rate, slows the accumulation of disabil-
`ity, and reduces MRI disease activity approximately 6
`months after treatment is initiated [5 – 8].
`
`* Corresponding author. Department of Neurology, University of
`Maryland Hospital, Rm. N4W46, 22 S. Greene Street, Baltimore, MD
`21201, USA. Tel.: +1-410-706-4216; fax: +1-410-706-0186.
`E-mail address: sjalbut@umaryland.edu (S. Dhib-Jalbut).
`
`Although the mechanism of action of GA is not fully
`understood, a number of studies both in EAE and in MS
`point to the induction of a GA-reactive T-cell repertoire with
`a protective Th2 anti-inflammatory phenotype as a likely
`mechanism of action [9 – 16]. The GA-reactive T-cells may
`exert their protective action by entering the CNS compart-
`ment and the production of anti-inflammatory cytokines in
`response to cross-recognition of myelin basic protein (MBP;
`bystander suppression) [17]. Additional mechanisms have
`been proposed, as a result of the ability of GA to bind
`promiscuously to HLA class II molecules associated with
`MS, including DR2 and DR4, on antigen presenting cells
`[18 – 20]. This includes inhibition of presentation of several
`myelin antigens to T-cells [4,11,15,21 – 24], induction of
`anergy or deletion of a population of GA-reactive T-cells
`[15,26].
`Although peripheral blood mononuclear cells (PBMC)
`from the majority of MS patients proliferate in response to
`GA, this response declines approximately 6 months after
`treatment is initiated [13,25]. The surviving GA-reactive T-
`
`0022-510X/02/$ - see front matter D 2002 Elsevier Science B.V. All rights reserved.
`PII: S 0 0 2 2 - 5 1 0 X ( 0 2 ) 0 0 2 0 1 - 0
`
`Page 1 of 7
`
`YEDA EXHIBIT NO. 2126
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`72
`
`M. Chen et al. / Journal of the Neurological Sciences 201 (2002) 71–77
`
`cells demonstrate a high degree of degeneracy, with a Th2-
`polarized response characterized by IL-5 and IL-13 secre-
`tion [13]. In view of the decline in the proliferative response
`to GA with time, we investigated whether MS patients who
`had received the drug for 6 – 9 years, continue to generate
`the anti-inflammatory GA-reactive Th2 cells associated with
`the therapeutic effect of the drug, and whether these cells
`have the characteristics of bystander suppression.
`
`2. Methods
`
`2.1. Antigens
`
`Glatiramer acetate (Copaxone lots 119135 and 119137)
`was from Teva Pharmaceutical Ind., Israel. Guinea pig MBP
`was purchased from Sigma (St. Louis, MO, USA). MBP
`peptide (83 – 99) (purity > 95%) was synthesized by Mixture
`Sciences (San Diego, CA, USA) using the simultaneous
`multiple synthesis methods [27]. Tetanus toxoid (TT) was
`obtained from Pasteur Merieux Connaught (North York,
`Ontario, Canada).
`
`2.2. Subjects
`
`Ten patients with clinically definite MS, who participated
`in the Copolymer-1 pivotal multicenter trial [7,8] and
`followed up at the Maryland Center for MS, were included
`in this study. Those patients received Glatiramer acetate
`treatment for 6 – 9 years and are termed the long-term
`treatment group. The clinical characteristics of the patients
`are presented in Table 1. Results were compared to those
`from another group of 10 MS patients who were studied 1 –
`10 months after they have been on Glatiramer acetate
`treatment (short-term treatment group) and on whom we
`have reported earlier [16]. The study was approved by the
`University of Maryland Institutional Review Board, and the
`subjects’ consent was obtained.
`
`Table 1
`Proliferative responses of GA – TCL from the long-term treatment group
`Relapsesa EDSSb
`
`Patient
`
`Duration of
`GA treatment
`(year)
`
`Precursor
`No.
`GA – TCL frequency
`
`Entry End
`
`2.3. Cells
`
`Approximately 100 cc of heparinized blood was obtained
`by venipuncture from the 10 MS patients at each time point.
`Peripheral blood mononuclear cells (PBMC) were prepared
`using a Ficoll-Hypaque gradient as described in the suppli-
`er’s protocol (ICN Biomedicals, OH, USA). For the gen-
`eration of antigen specific T-cell lines (TCL), cells were
`cultured without prior freezing.
`
`2.4. Generation of antigen-specific T-cell lines
`
`Antigen-specific TCL were generated by the split-well
`assay as described previously [28]. Briefly, PBMC were
`seeded in complete medium RPMI 1640 (Biofluid, Rock-
`ville, MD, USA) containing 5% human AB serum (Sigma),
`2 mM L-glutamine, 50 Ag/ml gentamicin, 100 U/ml pen-
`icillin/streptomycin, at 1 105 cells/well into 96-well U-
`bottom microtiter plates (Nunc, Roskilde, Denmark) and
`stimulated with antigens (20 Ag/ml of GA or 5 Ag/ml of TT).
`Thirty microtiter wells were prepared for each antigen.
`Human recombinant IL-2 (Biosource International Cama-
`rillo, CA, USA) was added to the culture on day 8 at a final
`concentration of 20 U/ml. On day 15, 50 Al of the cell
`suspension were transferred into each of two adjacent wells
`of a separate 96-well U-bottom microtiter plate, and 150 Al
`of complete medium containing 1 105 autologous irradi-
`ated PBMC (3000 rad) was added. One well was stimulated
`with 20 Ag/ml GA and the other with medium only. After 48
`h, 1 ACi/well of 3H-thymidine (Amersham Pharmacia Bio-
`tech, Piscataway, NJ, USA) was added. Eighteen hours later,
`cells were harvested on an automated cell harvester (Tom-
`tec, Hamden, CT, USA) and 3H-thymidine incorporation
`was measured using a Betaplate counter (Wallac, Gaithers-
`burg, MD, USA). Wells that showed stimulation index (SI,
`cpm of cells with antigen/cpm of cells without antigen) of
`>2, and background counts >200 cpm were further
`expanded and characterized.
`
`2.5. Precursor frequency estimation
`
`The precursor frequency of GA-reactive T-cells was
`estimated following the first stimulation cycle based on
`the number of proliferating microtiter wells and seeding
`105 cells/well; precursor frequency = number of GA-reactive
`TCL/number of wells plated 105.
`
`2.6. Cytokine production
`
`IFN-g and IL-5 were measured as markers of Th-1 and
`Th-2 phenotypes, respectively. Cells from GA-reactive
`wells identified after the first stimulation were restimulated
`with antigen, feeders, and IL-2 for 7 – 10 days. These cells
`were then harvested, washed, and cultured at 1 105 cells/
`well with 1 105/well of irradiated autologous PBMC with
`or without antigen for 48 h. Supernatants were harvested
`
`1
`1
`1
`7
`3
`7
`0
`1
`2
`1
`
`MS 12(503) 9
`MS 13(504) 6
`MS 14(525) 8
`MS 15(513) 6
`MS 16(524) 8
`MS 17(517) 8
`MS 18(518) 8
`MS 19(502) 9
`MS 20(519) 6
`MS 21(501) 6
`Total
`a Relapses during treatment period.
`b EDSS: expanded disability status scale at entry and end of the
`reporting period.
`
`1.5
`1.5
`2.5
`1.5
`5.0
`1.5
`1.5
`2.0
`2.5
`1.5
`
`1.0
`1.5
`2.5
`4.5
`4.0
`4.0
`1.0
`2.0
`2.5
`1.0
`
`5
`7
`4
`1
`17
`6
`4
`2
`2
`2
`48
`
`1.56/106
`1.09/106
`1.25/106
`0.33/106
`5.31/106
`1.87/106
`1.33/106
`0.67/106
`0.26/106
`0.33/106
`1.40/106
`
`Page 2 of 7
`
`YEDA EXHIBIT NO. 2126
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`M. Chen et al. / Journal of the Neurological Sciences 201 (2002) 71–77
`
`73
`
`and stored at 70 jC. IL-5 and IFN-g were measured by
`ELISA according to the manufacturer’s protocol (Bio-
`source). The sensitivity of the ELISA was < 4 pg/ml for
`both IL-5 and IFN-g. The GA-specific TCL were classified
`as Th0-, Th1- or Th2-biased based on the ratio of IFN-g/IL-
`5 secretion. A ratio >2 was arbitrarily defined as Th1 bias,
`< 0.5 as Th2 bias, and a value between 0.5 and 2 as Th0.
`
`2.7. Cross-reactivity studies
`
`Cross-reactivity of GA – TCL with MBP and MBP pep-
`tide 83 – 99 were examined using 10 Ag/ml of the antigens.
`These antigen concentrations had previously been found to
`give at least half-maximal stimulation in the vast majority of
`TCL [15]. Cross-reactivity of GA-reactive T-cells with MBP
`was determined by the split-well technique as described
`z
`above. Proliferative responses
`2 with a background of
`200 cpm or higher were considered cross-reactive. Cross-
`reactivity by cytokine release was performed as follows:
`1 105 cells/well of GA-reactive TCL were seeded in 96-
`well U-bottom microtiter plate together with 1 105 irradi-
`ated autologous PBMC in the absence of antigen or in the
`presence of GA, MBP, or MBP peptide 83 – 99 in triplicate
`wells. After cells were cultured for 48 h, 100-Al supernatants
`were removed from each well for cytokine level determi-
`nations. Cross-reactivity as measured by cytokine production
`was determined by pooling supernatants from triplicate wells
`and subsequent measurements of IL-5 and IFN-g levels.
`
`2.8. Statistical analysis
`
`A software package (Graphpad, Prismk) was used in the
`statistical analysis. Differences in lymphoproliferative
`responses and cytokine production between groups were
`compared using the Student’s t-test. A p-value < 0.05 was
`considered significant.
`
`3. Results
`
`3.1. Frequency of GA-reactive T-cells
`
`Forty-eight GA-reactive TLC were generated from 10
`MS patients who have been receiving GA treatment for 6 – 9
`
`Fig. 1. Regression lines for changes in the precursor frequency of GA and
`TT reactive T-cells with increasing GA treatment duration in MS patients.
`GA responses were obtained from 18 MS patients and TT responses from
`nine. The numbers adjacent to the regression lines indicate slopes (S) and p-
`values ( P).
`
`years (long-term treatment group; MS 12-21). The estimated
`precursor frequency of the GA-reactive T-cells ranged from
`0.26 to 5.31 in 1 million (Table 1). Precursor frequencies of
`GA-reactive T-cells generated from the long-term treatment
`group were compared to those generated from 10 MS
`patients who were studied pretreatment (81 TCL) and 1 –
`10 months after initiation of treatment with GA (130 TCL)
`(short-term treatment group) and on whom we have reported
`previously [16] (Table 2). The mean estimated precursor
`frequency of the GA-reactive TCL was 1.4/106 in the long-
`term treatment group, significantly lower than that of the
`short-term treatment group (4.03/106) ( p = 0.027). Regres-
`sion lines for the estimated precursor frequency of GA-
`reactive T-cells generated for the short-term treatment group
`z
`(1 – 6 months; n = 8) and long-term treatment group (
`6
`years; n = 10) demonstrated a significant drop in the esti-
`mated precursor frequency of GA – TCL with increasing
`treatment duration ( P = 0.010). In contrast, the estimated
`precursor frequency for the TT – TCL did not change sig-
`nificantly with long-term treatment (Fig. 1).
`
`3.2. GA-reactive TCL maintain a Th2 phenotype bias during
`long-term treatment with GA
`
`We and others have previously demonstrated that GA
`treatment induced an immune deviation in MS patients,
`characterized by a cytokine profile shift from Th1 pretreat-
`ment to Th2 during GA treatment [16]. In this study, we
`examined the cytokine secretion profile of 34 GA-reactive
`TCL generated from the long-term treatment group. A
`comparison of the average levels of IL-5 and IFN-g for
`GA and TT-reactive TCL from short- and long-term treat-
`ment groups is presented in Fig. 2. Mean IL-5 level was
`47.14 pg/ml pretreatment, 90.78 pg/ml for the short-term
`treatment group, and 94.69 pg/ml for the long-term treat-
`ment group. Mean IFN-g level was 171.2 pg/ml pretreat-
`
`Table 2
`Comparison of the estimated precursor frequency of GA-reactive T cells in
`the long-term and short-term treatment groups
`Precursor frequency/106
`Mean F S.D.
`2.66 F 3.32
`4.03 F 2.96
`1.40 F 1.48
`
`Range
`
`0.31 – 9.0
`0.94 – 9.67
`0.26 – 5.31
`
`P
`
`0.027
`
`GA – TCL
`
`Pre-Rx
`S.T. Rx
`L.T. Rx
`
`S.D.: standard deviation; Rx: treatment; S.T.: short-term; L.T: long-term;
`P: student t-test p-value.
`
`Page 3 of 7
`
`YEDA EXHIBIT NO. 2126
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`74
`
`M. Chen et al. / Journal of the Neurological Sciences 201 (2002) 71–77
`
`Fig. 2. Mean IL-5 (A and C) and IFN-g (B and D) produced by the GA-reactive (A and B) and TT-reactive (C and D) TCL pretreatment with GA, during short-
`term and long-term treatments.
`
`ment, 16.99 pg/ml for short-term treatment, and 31.19 pg/ml
`for long-term GA treatment. Both treatment groups had
`IFN-g levels significantly lower than pretreatment levels. In
`contrast, cytokine levels produced by TT-reactive TCL did
`not change significantly in both treatment groups compared
`to pretreatment levels.
`
`the Th1 and Th2 phenotype bias of the GA-
`Next,
`reactive TCL generated from the MS patients was analyzed.
`GA-reactive TCL producing IL-5 and IFN-g above the
`detection limits of the ELISA were classified as Th0-,
`Th1- or Th2-biased based on the ratio of IFN-g/IL-5
`secretion as described in Methods (Fig. 3). The mean ratio
`
`Fig. 3. Ratios of IFN-g to IL-5 levels for each of the GA-reactive T-cell lines pretreatment and in both treatment groups. The insert figure shows the mean ratios
`from the three groups.
`
`Page 4 of 7
`
`YEDA EXHIBIT NO. 2126
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`M. Chen et al. / Journal of the Neurological Sciences 201 (2002) 71–77
`
`75
`
`Table 3
`Comparison of cross-reactivity of GA – TCL with MBP in short-term and long-term treated groups
`
`MS patient
`
`Cross-reactive by proliferation
`
`Cross-reactive by cytokine secretion
`
`No. TCL
`
`tested
`
`No. TCL
`
`No. TCL
`
`No. TCL cross-reactive (%)
`
`cross-reactive (%)
`
`tested
`
`IL-5
`
`IFN-g
`
`Pre-Rx (n = 10)
`81
`10 (12.34%)
`51
`5 (9.80)
`4 (7.84)
`S.T. Rx (n = 10)
`130
`21 (16.15%)
`74
`23 (31.08)
`9 (12.16)
`L.T. Rx (n = 10)
`52
`6 (11.54%)
`32
`10 (31.25)
`2 (6.25)
`a Some TCL were cross-reactive for both IL-5 and IFN-g secretion, and therefore were counted as a single TCL in the ‘‘Total’’ column.
`
`Total
`
`8 (15.69)a
`24 (32.4)a
`12 (37.5)
`
`of IFN-g/IL-5 of GA-reactive TCL was 1.15 and 0.32 for
`long-term and short-term GA treatments, respectively; both
`significantly lower than the corresponding pretreatment
`ratio (2.76). It
`is also noteworthy that
`this ratio was
`significantly different between the two treatment groups
`with less Th2 bias in the long-term treatment group. The
`percentages of GA-reactive TCL classified as Th1, Th0, or
`Th2 were 20.59%, 23.53%, and 55.88% for the long-term
`treatment compared to 8%, 9%, and 83% for the short-term
`treatment group. In contrast, the Th1/Th0/Th2 distribution
`of TT-reactive TCL did not shift post-treatment in either
`group (data not shown).
`
`3.3. Cross-reactivity of GA-reactive TCL with MBP and
`MBP peptide 83 – 99
`
`We have previously reported cross-reactivity of GA-
`reactive TCL with MBP and MBP peptide 83 – 99 in the
`short-term treatment group [16]. Cross-reactivity was
`observed by proliferation for some GA – TCL and by
`cytokine secretion for others. In this study, we determined
`whether GA – TCL generated from the long-term treatment
`
`group manifest cross-reactivity with MBP and MBP (83 –
`99), since such cross-reactivity is a prerequisite for by-
`stander suppression. Cross-reactivity by proliferation
`(SI>2.0) was observed in 3 of the 10 patients, and in
`approximately 12% of the 52 GA – TCL examined (Table
`3). Cross-reactivity of the GA – TCL as measured by cyto-
`kine secretion was examined in 32 and 24 GA – TCL for
`MBP and MBP (83 – 99), respectively. Cytokine secretion
`(>50% over background levels) in response to MBP or MBP
`(83 – 99) was considered a cross-reactive response. Approx-
`imately 25% of the GA – TCL cross-reacted with MBP, and
`33% cross-reacted with MBP (83 – 99). The cytokines
`secreted by the cross-reactive GA – TCL in response to
`MBP or MBP (83 – 99) were biased in favor of the Th2
`phenotype (Fig. 4). Comparison of the cross-reactive
`responses in the long-term treatment group with that for
`the short-term treatment group is presented in Table 3.
`While the percentage of cross-reactive T-cell lines as meas-
`ured by proliferation was lower with long-term treatment,
`cross-reactivity as measured by cytokine secretion was not
`significantly different between the two groups. Interestingly,
`while 12% of the cross-reactive GA – TCL in the short-term
`
`Fig. 4. IL-5 and IFN-g levels produced by the MBP cross-reactive (A and B) and the MBP 83-99 cross-reactive (C and D) GA – TCL. A and C show the levels
`for each cross-reactive TCL. B and D show the mean levels for the cross-reactive TCL.
`
`Page 5 of 7
`
`YEDA EXHIBIT NO. 2126
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`76
`
`M. Chen et al. / Journal of the Neurological Sciences 201 (2002) 71–77
`
`treatment group produced IFN-g, only 6% did in the long-
`term treatment group, indicating a sustained Th2-polarized
`cross-reactive response in the long-term treatment group.
`
`4. Discussion
`
`A number of laboratories including ours reported on the
`generation of GA-reactive T-cells with a Th2-biased phe-
`notype as early as 1 month after initiation of GA treatment
`in MS patients [9,10,13 – 17,29]. Currently, it is believed
`that these cells mediate the therapeutic effect of GA in MS.
`Of some concern, however, is the observation that the robust
`proliferative response to GA observed early on during
`treatment declines after 6 months [13,25]. Therefore, the
`objective of this study was to examine the characteristics of
`the immune response to GA in patients who have been on
`the drug for several years and compare the findings to those
`from a group of MS patients who were studied in our
`laboratory before and after they had been on the drug for
`a shorter period (1 – 10 months). The availability of a group
`of MS patients at the University of Maryland who had
`participated in the pivotal CopaxoneR trial
`in the early
`1990s provided an opportunity to examine the long-term
`immunologic effects of Glatiramer acetate treatment in MS
`[30].
`First, we observed that the estimated precursor frequency
`of GA-reactive T-cells declined 2.9-fold compared to that
`during the first 10 months of treatment. Although the
`method we used is not a precise measure of precursor
`frequency, regression analysis supports such a decline with
`time. These observations are consistent with the findings of
`Ragheb et al. [26]. Since the peptide composition of GA is
`heterogeneous, these results may suggest that chronic stim-
`ulation with GA could result in the selection of a more
`restricted T-cell repertoire compared to that during early
`treatment. Alternatively, chronic administration of GA may
`result
`in the induction of anergy or deletion of highly
`reactive T-cells [15,26].
`The second notable observation is that the Th2 pheno-
`type of the GA-reactive T-cells remains dominant after
`many years of treatment. This is supported by the fact that
`significantly lower levels of IFN-g and higher IL-5 levels
`are produced by GA – TCL from both short-term and long-
`term treatment groups compared to pretreatment. Similarly,
`the IFN-g/IL-5 ratio remained Th2-biased in both groups.
`Therefore, despite a drop in the precursor frequency of GA-
`reactive T-cells with prolonged treatment,
`the sustained
`GA – T-cell response remains Th2-biased and therefore in
`favor of an anti-inflammatory response. The finding that
`long-term treatment with GA did not significantly affect the
`response to TT indicate that GA does not cause generalized
`immune suppression in MS patients. Of interest
`is the
`observation that some GA – TCL were Th1-biased despite
`prolonged treatment. One possible explanation is that in
`some individuals, GA induces CD8 + T-cells that produce
`
`IFN [25,31]. Another possibility is that GA constantly
`stimulates subpopulations of naı¨ve T-cells that
`initially
`produce IFN-g although our earlier studies indicate that
`the GA-reactive T-cells examined 1 week after the first
`
`/RO + ) [15]. It is
`stimulation are memory T-cells (CD45
`also conceivable that GA stimulates Th1-committed mem-
`ory cells with a low activation threshold in some donors and
`that these cells are expanded or at least perpetuated over
`time. The observation that the IFN-g/IL-5 ratio was signifi-
`cantly higher in the long-term treatment group (Fig. 3)
`seems to indicate that progressive deletion of Th1 cells as
`a result of chronic GA treatment is not a universal mech-
`anisms that accounts for the Th2-biased response. It is quite
`possible that the Th phenotype response is heterogeneous
`and is influenced by the genetic background of the patient
`including genes encoding for the HLA and T-cell receptor.
`Thirdly, cross-reactivity of some GA – TCL with MBP is
`observed after long-term treatment and the magnitude of this
`cross-reactivity is not compromised. More importantly, the
`cytokines released in response to the cross-reactive antigen
`remain Th2-biased. These findings are relevant
`to the
`mechanism of action of GA in MS as it is currently believed
`that GA-reactive T-cells cross the blood – brain barrier,
`cross-react with myelin antigens and consequently produce
`bystander suppression through the secretion of anti-inflam-
`matory cytokines.
`trial, who remained
`Patients enrolled in the pivotal
`clinically stable, continued participation in the open label
`phase which included the 10 patients examined in this study.
`Therefore, the patient population we examined is biased in
`favor of clinical responders as reflected by the stability of
`the EDSS scores over at least 6 years of treatment. In spite
`of the rather uniform clinical stability among the 10 patients,
`we observed heterogeneity in the precursor frequency and
`the phenotype of the GA – TCL response, and in cross-
`reactivity with myelin antigens. The two patients who had
`relatively higher relapse rate during the course of treatment
`(Table 1, cases MS 15 and MS 17) did not have a unique
`immunologic profile that distinguished them from the rest of
`the group. Currently, it is not known whether the immuno-
`logic responses to GA correlate with the clinical response.
`Although this study was not intended to address this issue
`because of the small sample size, the fact that there is
`substantial heterogeneity in the immune responses to GA
`warrants further investigations in a larger patient population,
`in order to determine if the immunologic parameters corre-
`late with the clinical response, and whether they can be used
`as surrogate markers of treatment response.
`
`Acknowledgements
`
`This work was supported by grants from TEVA
`pharmaceuticals, The National Institute of Neurological
`Disorders and Stroke (K-24-NS02082), and the Department
`of Veteran’s Affairs.
`
`Page 6 of 7
`
`YEDA EXHIBIT NO. 2126
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`M. Chen et al. / Journal of the Neurological Sciences 201 (2002) 71–77
`
`77
`
`References
`
`[1] Teitelbaum D, Meshorer A, Hirshfeld T, Arnon R, Sela M. Suppres-
`sion of experimental allergic encephalomyelitis by a synthetic poly-
`peptide. Eur J Immunol 1971;1:242 – 8.
`[2] Teitelbaum D, Webb C, Bree M, Meshorer A, Arnon R, Sela M.
`Suppression of experimental allergic encephalomyelitis in Rhesus
`monkeys by a synthetic basic copolymer. Clin Immunol Immunopa-
`thol 1974;3:256 – 62.
`[3] Lisak RP, Zweiman B, Blanchard N, Rorke LB. Effect of treatment
`with Copolymer 1 (Cop-1) on the in vivo and in vitro manifestations
`of experimental allergic encephalomyelitis (EAE). J Neurol Sci
`1983;62:281 – 93.
`[4] Teitelbaum D, Fridkis-Hareli M, Arnon R, Sela M. Copolymer 1
`inhibits chronic relapsing experimental allergic encephalomyelitis
`induced by proteolipid protein (PLP) peptides in mice and inter-
`feres with PLP-specific T cell responses. J Neuroimmunol 1996;64:
`209 – 17.
`[5] Bornstein MB, Miller A. Slagle sea. A pilot trial of COP 1 in ex-
`acerbating – remitting multiple sclerosis. N Engl J Med 1987;317:
`408 – 14.
`[6] Johnson KP, Brooks BR, Cohen JA, Ford CC, Goldstein J, Lisak RP,
`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. Neurology 1995;45:
`1268 – 76.
`[7] Johnson KP, Brooks BR, Cohen JA, Ford CC, Goldstein J, Lisak RP,
`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. The copolymer 1 multiple sclerosis study group.
`Neurology 1998;50:701 – 8.
`[8] Comi G, Filippi M, Wolinsky JS. The effects of glatiramer acetate on
`magnetic resonance imaging-measured disease activity and burden in
`patients with relapsing remitting multiple sclerosis. Ann Neurol
`2001;49:290 – 7.
`[9] Aharoni R, Teitelbaum D, Arnon R. T suppressor hybridomas and
`interleukin-2-dependent lines induced by copolymer 1 or by spinal
`cord homogenate down-regulate experimental allergic encephalomye-
`litis. Eur J Immunol 1993;23:17 – 25.
`[10] Aharoni R, Teitelbaum D, Sela M, Arnon R. Bystander suppression of
`experimental autoimmune encephalomyelitis by T cell lines and
`clones of the Th2 type induced by copolymer 1. J Neuroimmunol
`1998;91:135 – 46.
`[11] Aharoni R, Teitelbaum D, Arnon R, Sela M. Copolymer 1 acts against
`the immunodominant epitope 82 – 100 of myelin basic protein by T
`cell receptor antagonism in addition to major histocompatibility com-
`plex blocking. Proc Natl Acad Sci U S A 1999;96:634 – 9.
`[12] Miller A, Shapiro S, Gershtein R, Kintary A, Rawashden H, Honig-
`man S, et al. Treatment of Multiple sclerosis with copolymer-1
`(CopaxoneR): implicating mechanism of Th1 to Th2/Th3 immune-
`deviation. J Neuroimmunol 1998;92:113 – 21.
`[13] Duda PW, Schmied MC, Cook SL, Krieger JI, Hafler DA. Glatiramer
`acetate (CopaxoneR) induces degenerate, Th2-polarized immune re-
`sponses in patients with multiple sclerosis. J Clin Invest 2000;105:
`967 – 76.
`[14] Neuhaus O, Farina C, Yassouridis A, Wiendl H, Bergh FT, Dose T, et
`al. Multiple sclerosis: 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 – 7.
`
`[15] Gran B, Tranquil LR, Chen M, Bielekova B, Zhou W, Dhib-Jalbut S,
`et al. Mechanisms of immunomodulation by glatiramer acetate. Neu-
`rology 2000;55:1704 – 14.
`[16] Chen M, Gran B, Costello K, Johnson K, Martin R, Dhib-Jalbut S.
`Glatiramer acetate induces a Th2-biased response and cross-reactivity
`with myelin basic protein. Mult Scler 2001;7:209 – 19.
`[17] Miller A, Lider O, Weiner HL. Antigen-driven bystander suppression
`after oral administration of antigens. J Exp Med 1991;174:791 – 8.
`[18] Fridkis-Hareli M, Teitelbaum D, Gurevich E, Pecht I, Brautbar C,
`Kwott OJ, et al. Direct binding of myelin basic protein and synthetic
`copolymer I to class II major histocompatibility complex molecules
`on living antigen-presenting cells- specificity and promiscuity. Proc
`Natl Acad Sci U S A 1994;91:4872 – 6.
`[19] Fridkis-Hareli M, Teitelbaum D, Pecht I, Arnon R, Sela M. Binding of
`copolymer 1 and myelin basic protein leads to clustering of class II
`MHC molecules on antigen-presenting cells. Int Immunol 1997;9:
`925 – 34.
`[20] Fridkis-Hareli M, Strominger JL. Promiscuous binding of synthetic
`copolymer 1 to purified HLA-DR molecules. J Immunol 1998;160:
`4386 – 97.
`[21] Racke MK, Martin R, McFarland H, Fritz RB. Copolymer-1-induced
`inhibition of antigen-specific T cell activation: interference with anti-
`gen presentation. J Neuroimmunol 1992;37:75 – 84.
`[22] Teitelbaum D, Aharoni R, Arnon R, Sela M. Specific inhibition of the
`T-cell response to myelin basic protein by the synthetic copolymer
`Cop 1. Proc Natl Acad Sci U S A 1988;85:9724 – 8.
`[23] Teitelbaum D, Milo R, Arnon R, Sela M. Synthetic copolymer 1
`inhibits human T-cell lines specific for myelin basic protein. Proc Natl
`Acad Sci U S A 1992;89:137 – 41.
`[24] Ben-Nun A, Mendel I, Bakimer R, Fridkis-Hareli M, Teitelbaum D,
`Arnon R, et al. The autoimmune reactivity to myelin oligodendrocyte
`glycoprotein (MOG) in multiple sclerosis is potentially pathogenic:
`effect of copolymer 1 on MOG-induced disease. J Neurol 1996;243:
`S14 – 22.
`[25] Farina C, Then Bergh F, Albrecht H, Meinl E, Yassouridis A, Neuhaus
`O, et al. Treatment of multiple sclerosis with Copaxone (COP) Elispot
`assay detects COP-induced interleukin-4 and interferon-g response in
`blood cells. Brain 2001;124:705 – 19.
`[26] 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 – 7.
`[27] Houghten RA. General method for the rapid solid-phase synthesis of
`large numbers of peptides: specificity of antigen-antibody interaction
`at the level of individual amino acids. Proc Natl Acad Sci U S A
`1985;82:5131 – 5.
`[28] Martin R, Utz U, Coligan JE, Richert JR, Flerlage M, Robinson E, et al.
`Diversity in fine specificity and T cell receptor usage of the human
`CD4+ cytotoxic T cell response specific for the immunodominant mye-
`lin basic protein peptide 87-106. J Immunol 1992;148:1359 – 66.
`[29] Aharoni R, Teitelbaum D, Sela M, Arnon R. Copolymer 1 induces T
`cells of the T helper type 2 that cross-react with myelin basic protein
`and suppress experimental autoimmune encephalomyelitis. Proc Natl
`Acad Sci U S A 1997;94:10821 – 6.
`[30] Johnson KP, Brooks BR, Ford CC, Goodman A, Guarnaccia J, Lisak
`RP, et al. Sustained clinical benefits of glatiramer acetate in relapsing
`multiple sclerosis patients observed for 6 years. Mult Scler 2000;6:
`256 – 66.
`[31] Karandikar NJ, Crawford MP, Yan X, Ratts RB, Brenchley JM, Am-
`brozak DR, et al. Glatiramer acetate (Copaxone) therapy induces
`CD8(+) T cell responses in patients with multiple sclerosis. J Clin
`Invest 2002;109:641 – 9.
`
`Page 7 of 7
`
`YEDA EXHIBIT NO. 2126
`MYLAN PHARM. v YEDA
`IPR2015-00644

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