throbber
ARTICLE
`
`Multiple Sclerosis 2007; 13: 754 762
`
`Clinical response to glatiramer acetate correlates with
`modulation of IFN-g and IL-4 expression in multiple
`sclerosis
`
`RM Valenzuela1, K Costello2, M Chen2, A Said2, KP Johnson2 and S Dhib-Jalbut1
`
`Objective To determine whether glatiramer acetate (GA)-induced lymphoproliferation and IFN-g and
`IL-4 modulation correlate with the clinical response in multiple sclerosis (MS).
`Background GA therapy involves the induction of anti-inflammatory cytokine shifts. However, it is
`not known whether this response correlates with the clinical outcome.
`Methods Thirty-six relapsing-remitting (RR) MS patients were treated with GA for at least two years,
`and classified clinically as GA-responders (GA-R/22) or hypo/non-responders (GA-HR/NR/14).
`Proliferation of peripheral blood mononuclear cells (PBMC) to GA and Tetanus toxoid (TT), as well as
`IL-4 and IFN-g ELISPOT, were performed.
`Findings There was no difference in PBMC proliferation to GA or TT between GA-R and GA-HR/NR
`before and during treatment (P/0.05). The mean number of IFN-g ELISPOTS in unstimulated, TT
`and anti-CD3/CD28-stimulated PBMC was lower among GA-R (unstimulated: GA-R/10.19/6.21
`(n/22) versus GA-HR/NR/17.89/12.7 (n/14), P/0.04; TT-GA-R/12.29/4.06 (n/12) versus
`GA-HR/NR/26.89/21.0 (n/8), P/0.028; anti-CD-3/CD28 GA-R/217.39/140.4 (n/22) versus
`GA-HR/NR/368.59/170.1 (n/14), P/0.006). In contrast, the number of IL-4 ELISPOTS remained
`unchanged in the GA-R group, but was progressively reduced in the GA-HR/NR group during GA
`therapy (GA-HR/NR IL-4: pre-Rx: 599/34 versus 229/11 at 12 months (n/6), P/0.0429). The IL-4/
`IFN-g ratio in anti-CD3/CD28-stimulated PBMC was significantly higher among GA-R compared to
`GA-HR/NR (P/0.0474).
`Interpretation Lymphoproliferation to GA did not differentiate GA-R from GA-HR/NR. However,
`reduced IFN-g expression and stable IL-4 expression in anti-CD3/CD28-stimulated PBMC, and an
`increased IL-4/IFN-g ratio was associated with favorable clinical response. More data are needed to
`validate the prospective use of IL-4/IFN-g expression in PBMC as a biomarker of clinical response to
`GA for individual patients. Multiple Sclerosis 2007; 13: 754 762. http://msj.sagepub.com
`
`Key words: multiple sclerosis; glatiramer acetate; interferon-gamma; Interleukin-4
`
`Introduction
`
`The mechanism of action of glatiramer acetate (GA)
`in the treatment of multiple sclerosis (MS) is not
`fully elucidated. The molecule binds to HLA class II
`molecules associated with MS,
`including DR2
`and DR4, on antigen presenting cells [1 5]. Recent
`studies, both in experimental autoimmune ence-
`[6 8,12] and in humans
`phalomyelitis
`(EAE)
`[7,9 14], indicate that a likely in vivo mechanism
`
`of action of GA involves the induction of immu-
`nomodulatory regulatory T-helper type 2 (Th2)
`cells. The GA-specific T cells may exert
`their
`protective action systemically and by entering the
`CNS compartment, as evidenced by inhibition of
`new magnetic resonance imaging (MRI) gadoli-
`nium-enhancing lesions progressing into black
`holes [15], and the production of anti-inflamma-
`tory cytokines and brain-derived neurotrophic fac-
`tor (BDNF) [16], in response to cross-recognition of
`
`1 UMDNJ-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
`2 School of Medicine, University of Maryland, Baltimore, MD, USA
`Author for correspondence: Suhayl Dhib-Jalbut, MD, Department of Neurology, 97 Paterson Street, Room 205, New
`Brunswick, NJ 08901, USA. E-mail: jalbutsu@umdnj.edu
`Received 24 March 2006; accepted 30 October 2006
`
`– 2007 SAGE Publications
`
`10.1177/1352458506074510
`
`Downloaded from
`
`msj.sagepub.com
`
` by guest on November 18, 2015
`
`Page 1 of 9
`
`YEDA EXHIBIT NO. 2124
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`myelin antigens (bystander suppression) [6,17]. We
`and others have demonstrated that GA treatment in
`MS results in the induction of GA-specific T cells
`with a predominant Th2 phenotype, both in re-
`sponse to GA and crossreactive myelin antigens
`[17,18]. These results strongly suggest that the
`mechanism of action of GA in MS involves the
`induction of crossreactive GA-specific T-cells with a
`predominant Th2 cytokine profile. Importantly,
`the induction of GA-specific Th2 cells was not
`universal
`in MS patients treated with the drug
`[19]. Since clinical response to GA varies among
`treated patients, this raises the possibility that the
`immunological effects of GA may correlate with the
`clinical response, and may distinguish responders
`from hypo or non-responders.
`Recently, Farina et al. [20], reported a triad of
`immune responses to GA that predict treated from
`untreated patients. This triad consisted of: (1) a
`decline in GA-induced proliferation; (2) positive IL-
`4 CD4 cells; and (3) positive IFNg CD8 cells in
`response to high dose of GA (100 mg/mL). Whether
`this triad correlates with the clinical response is
`not certain at present. In this study, we determined
`whether the lymphoproliferative response to GA
`and the GA-induced cytokine shifts in IL-4 and
`IFN-g correlate with the clinical response to GA.
`
`Materials and methods
`
`Subjects
`
`Clinical response to GA
`
`755
`
`Cells
`
`Approximately 60 cc of heparinised blood was
`obtained by venipuncture from each MS patient at
`different time points during treatment, as indicated
`in Table 1. In a subset of patients, blood samples
`were obtained before treatment, and at three, six,
`12 and 18 24 months during treatment. The study
`was approved by the University of Maryland In-
`stitutional Review Board, and informed consent
`was obtained from each patient enrolled in the
`study. Peripheral blood mononuclear cells (PBMC)
`were purified using a Ficoll-Hypaque gradient, as
`described in the supplier’s protocol (ICN Biomedi-
`cals Inc., OH, USA).
`
`Lymphoproliferation assay
`
`One /105 PBMC/well were seeded in a 96-well
`U-bottom microtiter plate together with 1/105
`irradiated autologus PBMC,
`in the presence of
`antigen in triplicate wells (glatiramer acetate at 40
`and 100 mg/mL, Teva Pharmaceutical Industries,
`Ltd., Israel). Tetanus toxoid (TT; 5 mg) obtained
`from Pasteur Merieux Connaught (North York,
`Ont., Canada) was used as a control antigen. After
`cells were cultured for 48 hours, 1 mCi 3H-thymin-
`dine was added. Cells were harvested 18 hours later
`using TOMTEC cell harvester (TOMTEC, Hamden,
`CT). Incorporated radioactivity was measured using
`a liquid Scintillation counter (Wallac MicroBeta
`Trilux, Perkin Elmer, Boston, MA).
`
`Thirty-six consecutive patients, diagnosed with
`relapsing-remitting (RR) MS and treated with GA
`for at least two years, were included in the study.
`Thirty-one patients were female with a mean age of
`44 years, and five were male with a mean age of 49
`years. Patients had at least one relapse during the
`year prior to initiation of GA therapy. All subjects
`were closely followed up at the University of Mary-
`land Center for MS. The clinical characteristics of
`the patients are presented in Table 1. After at least
`two years on GA therapy, patients were classified as
`GA-responders (R) (n/ 22) or hypo/non-responders
`(HR/NR) (n/ 14) based on criteria arbitrarily set for
`this study. We adopted a more conservative criteria
`for patient classification into GA-R and GA-HR/NR
`compared to those recently reported in the litera-
`ture [21,22]. A responder (R) is a patient with an
`annual relapse rate (ARR) B/0.5 and no evidence of
`disease progression, as measured by Expanded
`Disability Status Scale (EDSS). A HR/NR is a patient
`with an ARR /0.5 and/or with progression in the
`EDSS of at least 1 point sustained for six months.
`
`ELISPOT assay
`
`Human IFN-g (Th1 indicator) and IL-4 (Th2 indi-
`cator) were measured by the ELISPOT assay, accord-
`ing to the manufacturer’s protocol (BD Biosciences,
`San Diego, CA, USA). Briefly, ELISPOT plates (PVDF
`plate, Millipore Corp., MA, USA) were coated with
`100 mL/well of 5 mg/mL capture antibody at 48C
`overnight. The plates were then washed with PBS,
`and incubated with 200 mL/well of blocking solu-
`tion (culture medium) for 2 hours. PBMC (2/105
`cells/well for IFN-g and 4/105 cells/well for IL-4
`detection) and antigen (GA 40 and 100 mg/mL, TT 5
`mg/mL, CD3Ab (2.5 mg/mL)/CD28 Ab (1 mg/mL))
`was added and cultured overnight at 378C in a 5%
`CO2 incubator. Patients were tested in triplicate
`wells in response to each antigen. After overnight
`culture, the plates were washed twice with deio-
`nised water, and three times with PBS, then in-
`cubated with 100 mL of 2.0 mg/mL biotinylated
`antibody for 2 hours at room temperature. The
`plates were then washed three times with wash
`buffer (0.05% Tween-20 PBS), and incubated with
`
`http://msj.sagepub.com
`
`Multiple Sclerosis 2007; 13: 754 762
`
`Downloaded from
`
`msj.sagepub.com
`
` by guest on November 18, 2015
`
`Page 2 of 9
`
`YEDA EXHIBIT NO. 2124
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`756
`
`RM Valenzuela et al.
`
`Table 1 Characteristics of MS patients and time points during treatment when blood samples were obtained
`
`Time sample analysed (months)
`
`EDSS
`
`ARR
`
`ID No.
`
`Age/sex
`
`Disease duration (years)
`
`Rx duration (months)
`
`0
`
`3
`
`42
`39
`43
`44
`45
`46
`47
`48
`49
`9
`50
`52
`7
`53
`54
`55
`56
`57
`58
`59
`1
`2
`3
`4
`6
`7a
`8
`10
`11
`12
`14
`17
`18
`21
`22
`24
`
`29/F
`67/F
`36/F
`41/F
`39/F
`40/F
`37/M
`64/F
`46/F
`49/F
`44/F
`51/F
`52/F
`40/M
`46/F
`41/F
`22/F
`53/F
`48/F
`43/F
`32/F
`45/M
`45/F
`32/F
`60/F
`53/F
`53/F
`43/F
`52/F
`40/F
`51/F
`60/M
`29/F
`49/F
`63/M
`36/F
`
`4
`37
`5
`Uncertain
`18
`19
`13
`25
`12
`6
`15
`20
`5
`3
`5
`5
`2
`32
`5
`Uncertain
`5
`6
`23
`37
`5
`Uncertain
`18
`13
`25
`12
`21
`5
`5
`5
`6
`5
`
`35
`40
`55
`27
`60
`24
`56
`27
`24
`24
`45
`38
`38
`24
`30
`25
`24
`57
`40
`41
`36
`24
`24
`26
`48
`24
`29
`36
`24
`24
`24
`24
`24
`24
`27
`24
`
`*
`*
`
`*
`
`*
`
`*
`
`*
`*
`*
`
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`
`6
`
`*
`
`*
`
`*
`
`*
`
`*
`
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`
`12
`
`18 24
`
`Pre-Rx On-Rx
`
`Pre-Rx On-Rx
`
`Patient status
`
`1
`1.5
`0
`1.5
`6.5
`2
`1.5
`2
`2
`2.5
`1.5
`3
`1.5
`0
`2.5
`2.5
`2
`6
`0
`1.5
`1.5
`2
`1.5
`3.5
`7
`3.5
`2
`1.5
`2
`1.5
`1.5
`2.5
`1.5
`2
`2.5
`1.5
`
`0
`1.5
`0
`1
`6.5
`2
`1
`1.5
`1.5
`4
`1.5
`4
`2.5
`1.5
`2.5
`2.0
`0
`7
`1
`1.5
`1.5
`2.5
`1.5
`1.5
`1.5
`7
`2
`2.5
`6
`1
`2.5
`2
`1.5
`2
`2.5
`1.5
`
`1.5
`2
`1.5
`1.5
`3
`2
`1.5
`1
`1.5
`2
`1.5
`1.5
`1
`0.5
`1.5
`0.5
`2
`1
`1
`0.9
`2.5
`0.5
`2
`1.5
`3
`0.5
`1.5
`1
`1.5
`0.5
`1.5
`1
`1.5
`1
`2
`2
`
`0.3
`0
`0.9
`0.4
`0
`0
`0
`0
`0
`1.5
`0
`1.3
`0
`0
`0.4
`0
`0
`0.4
`0.6
`1.5
`0
`0.99
`0
`0.46
`0
`0.49
`0
`0.99
`0
`0
`1.33
`1.2
`0
`0
`0
`0
`
`R
`R
`NR
`R
`R
`R
`R
`R
`R
`NR
`R
`NR
`NR
`NR
`R
`R
`R
`NR
`NR
`NR
`R
`NR
`R
`R
`R
`NR
`R
`NR
`NR
`R
`NR
`NR
`R
`R
`R
`R
`
`*
`
`*
`
`*
`*
`*
`
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`
`*
`
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`*
`
`Rx: treatment; pre-Rx, pre-treatment; post-Rx, post-treatment; ARR, annual relapse rate; R, responder; HR/NR, hypo/non-responder; LV, last visit.
`*Blood sample obtained.
`
`Multiple Sclerosis 2007; 13: 754 762
`
`http://msj.sagepub.com
`
`Downloaded from
`
`msj.sagepub.com
`
` by guest on November 18, 2015
`
`Page 3 of 9
`
`YEDA EXHIBIT NO. 2124
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`100 mL of 1:100 dilution of Avidin-HRP for 1 hour
`at room temperature, and washed three times with
`wash buffer. Finally, 100 mL of AEC (3-amino
`-9-ethyl-carbazole) substrate was added, and the
`reaction was stopped by washing plates with dis-
`tilled water. Spots were counted and analysed by
`CTL Analysers LLC ELISPOT Plate Reader (Cleve-
`land, OH, USA).
`
`Statistical analysis
`
`A software package (Graphpad, PrismTM) was used in
`the statistical analysis. The differences in cytokine
`spots between responder and hypo/non-responder
`were compared using the Student’s t -test and
`Bonferroni’s multiple comparison test. A P value
`B/0.05 was considered significant. Although only
`2/105 cells/well were utilised for the IFN-g ELI-
`SPOT, the values were adjusted to 4/105 cells/well
`for both the IL-4 and IFN-g spots to account for the
`corrected dilution factor.
`
`Results
`
`Classification of clinical responders and hypo/non-
`responders
`
`The patients were classified as GA-responders or
`hypo/non-responders after being treated with GA
`for at least 24 months, based on their annual
`relapse rate and progression in disability while on
`treatment (Table 1). The mean treatment duration
`was 34 months. The GA-responder group had a
`mean ARR of 1.68 and a mean EDSS of 2.31 before
`treatment compared to 0.07 ARR and an EDSS of
`1.70 during treatment. The hypo/non-responder
`group had a mean ARR of 1.10 and a mean EDSS
`of 1.96 before treatment compared to 0.75 ARR and
`an EDSS of 3 during GA therapy. Overall, a 96%
`reduction in the ARR, and a 0.61 point decrease in
`the EDSS occurred in the GA-R, whereas a 31%
`reduction in the ARR, and a 1.14 point increase
`in the EDSS occurred in the GA-HR/NR during
`treatment.
`
`Clinical response to GA
`
`757
`
`anti-CD3/CD28-stimulated PBMC, was significantly
`lower among GA-R compared to GA-HR/NR
`(unstimulated: GA-R/10.19/8.21 (n/ 22) versus
`GA-HR/NR/17.89/12.7 (n/ 14), P/ 0.03; TT-GA-
`R/12.29/4.06 (n/ 12) versus GA-HR/NR/26.89/
`21.0 (n/ 8), P/ 0.028; anti-CD-3/CD28 GA-R/
`217.39/140.4 (n/ 22) versus GA-HR/NR/368.59/
`170.1 (n/ 14), P/ 0.006). The mean number of
`IL-4 spots
`in anti-CD3/CD28-stimulated PBMC
`was not significantly different between GA-R and
`GA-HR/NR (GA-R/92.09/84.9 (n/ 22) versus GA-
`HR/NR/80.89/80.6 (n/ 14), P/ 0.05). The IL-4/
`IFN-g ratio in anti-CD3/CD28-stimulated PBMC
`was significantly higher among GA-R compared to
`GA-HR/NR (0.939/1.02 versus 0.349/0.40, respec-
`tively; P/ 0.0474).
`In 16 of the 36 patients (10 GA-R and six GA-HR/
`NR) pre-treatment and during treatment IL-4 and
`IFN-g ELISPOTS were determined. Cytokine expres-
`sion studies in anti-CD3/CD28-stimulated PBMC
`were performed at serial time points, ranging from
`three to 24 months for this sub-group (Figure 1).
`Mean IL-4 spots did not change significantly during
`treatment compared to pre-treatment levels in the
`GA-R group (Figure 1a). In contrast, mean IL-4 spots
`decreased from pre-treatment levels in the GA-HR/
`NR group beginning at three months of GA ther-
`apy, and the decrease became statistically signifi-
`cant at 12 months and beyond (Figure 1b). The
`mean IFN-g spots steadily decreased during treat-
`ment
`in the GA-R group, beginning at
`three
`months, and this decrease became statistically
`significant at ]/12 months (Figure 1c). Although a
`decrease in IFN-g was observed in the GA-HR/NR
`group, this decrease was delayed (]/12 months),
`and did not reach statistical significance (Figure
`1d). The mean IL-4/IFN-g ratio increased steadily
`during treatment in the GA-R group, and became
`statistically significant at six months and later
`the mean IL-4/IFN-g
`(Figure 1e).
`In contrast,
`ratio remained unchanged at all time points in
`the GA-HR/NR group (Figure 1f).
`
`Cytokine shifts
`
`Correlation of IL-4/IFNg ratio with the clinical
`response
`
`Initially, expression of IFN-g and IL-4 was examined
`by ELISPOT in PBMC obtained ex vivo from GA-
`treated MS patients at random time points during
`treatment
`(Table 2). There was no significant
`difference in cytokine production between GA-R
`and GA-HR/NR in response to GA stimulation
`at 40 and 100 mg/mL. The mean number of
`IFN-g spots in unstimulated TT (5 mg/mL) and
`
`Linear regression analysis using Spearman correla-
`tion showed no significant correlation between the
`number of IL-4 spots and ARR (Figure 2a), while a
`significant positive correlation was observed be-
`tween the number of IFN-g spots and ARR (P/
`0.040) (Figure 2b). A significant negative correla-
`tion between ARR and IL-4/IFN-g ratio was observed
`(P/ 0.041) (Figure 2c).
`
`http://msj.sagepub.com
`
`Multiple Sclerosis 2007; 13: 754 762
`
`Downloaded from
`
`msj.sagepub.com
`
` by guest on November 18, 2015
`
`Page 4 of 9
`
`YEDA EXHIBIT NO. 2124
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`758
`
`RM Valenzuela et al.
`
`Table 2 IL-4 and IFN-g expression by Elispot obtained from MS patients treated with GA
`
`R
`
`HR/NR
`
`R
`
`HR/NR
`
`R
`
`HR/NR
`
`R
`
`HR/NR
`
`No. Ag
`
`GA (40 mg/mL)
`
`GA (100 mg/mL)
`
`TT (5 mg/mL)
`
`CD3/CD28
`(2.5 mg/1 mg/mL)
`
`5.409/3.90
`(n/22)
`9.279/5.54
`(n/12)
`11.319/6.45
`(n/12)
`10.969/5.79
`(n/12)
`92.09/84.95
`(n/22)
`
`4.009/2.56
`(n/14)
`8.069/2.84
`(n/8)
`11.899/5.14
`(n/8)
`8.339/3.23
`(n/8)
`80.89/80.6
`(n/14)
`
`10.19/6.21
`(n/22)
`10.839/7.2
`(n/12)
`14.09/8.04
`(n/12)
`12.29/4.06
`(n/12)
`217.39/140
`(n/22)
`
`17.89/12.7
`(n/14)
`13.09/9.53
`(n/8)
`16.59/9.41
`(n/8)
`26.89/21.0
`(n/8)
`368.59/170
`(n/14)
`
`0.59/0.60
`
`0.259/0.40 0.25 0.20
`
`1.779/2.15
`
`0.6589/0.29 0.73 0.60
`
`0.9679/0.57
`(n/12)
`1.359/0.94
`
`0.829/0.39 0.85 0.74
`
`0.769/0.27 1.12 0.41
`
`0.939/1.02
`
`0.3419/0.4
`
`0.39 0.15
`
`R, GA-responder; HR/NR, GA-hypo/non-responder; n, number of patients.
`*Mean number of spots per 4/105 cells/well.
`The mean IL-4 and IFN-g spots shown in columns 2 and 3 were calculated as follows: first, the mean number of spots during the three
`to 24 month-treatment period was determined for each individual. Subsequently, the average of the mean number of spots for each
`group was calculated. The IL-4/IFN-g ratio shown in column 4 represents the mean of the ratios for each group. Note that the ratio in
`column 4 is not the ratio of IL-4 and IFN-g means shown in column 2 and 3.
`
`Lymphoproliferative response
`
`The ex vivo PBMC proliferative response to GA and
`TT are presented in Figure 3 as DCPM (mean cpm
`in antigen stimulated minus mean cpm in unsti-
`mulated wells) and as stimulation indexes (SI/
`mean CPM in stimulated wells/mean CPM in
`unstimulated wells). Overall, there was an initial
`increase in the proliferative response to GA fol-
`lowed by a decline after six months of treatment,
`which was statistically significant at 12 months
`(P/0.04) and 18 24 months (P/0.01). An initial
`decrease in the proliferative response to TT was
`observed at three months (P/0.007), but this
`response recovered after six months, and was not
`significantly different from pre-treatment values.
`There was no difference in PBMC proliferation to
`GA or TT between GA-R and GA-HR/NR before and
`during treatment at three, six, 12 months or two
`years (P values at all time points were /0.05).
`
`Discussion
`
`Previous studies suggested that a decrease in the
`lymphoproliferative response to GA and changes in
`IL-4 and IFN-g expression are associated with GA
`treatment in MS [13,18,21]. The goal of this study
`was to determine whether the lymphoproliferative
`response to GA and GA-induced cytokine shifts
`correlate with the clinical response. While GA
`treatment initially induces a strong lymphoproli-
`ferative response during the first three months, this
`response declines over time [20,23]. It is believed
`that the residual T-cell response to GA after six
`months of treatment includes predominantly Th2-
`biased cells with significant degeneracy in their
`antigen recognition patterns [24]. These cells cross-
`react with myelin antigens [18], and are believed to
`
`mediate the GA-therapeutic effect through the
`release of anti-inflammatory Th2 cytokines, result-
`ing in bystander suppression and by secretion of
`brain-derived neurotrophic factor
`(BDNF)
`[16].
`However,
`it is not known at the present time
`whether GA-induced cytokine shifts correlate with
`the clinical response to treatment with GA.
`Earlier reports have shown a reduction in PBMC
`proliferation to GA among GA-treated patients
`[20,24,31]. However, these results were not corre-
`lated with patients’ clinical response to the drug.
`Subsequently, Farina et al. [20], and Weder et al.
`[31], examined the proliferative response to GA in
`GA-treated MS patients versus healthy controls
`and untreated MS patients, and found that T cells
`from GA-treated MS patients had a significant
`reduction in stimulation indexes. Although the
`PBMC proliferation data presented by Farina et al.
`showed a trend towards a higher GA-induced
`stimulation index among the GA non-responders,
`the difference between GA-responders and GA-
`non-responders was not statistically significant,
`and failed to differentiate the two groups.
`In
`addition, they found that a strong in vitro response
`for both IL-4 and IFN-g correlates with a successful
`clinical outcome [29]. Our data showed an initial
`increase in both the DCPM and stimulation in-
`dexes within three months of treatment. However,
`a significant reduction was observed in the DCPM
`and stimulation indexes after six months of GA
`therapy, which is consistent with the findings
`reported by Duda et al. [24]. Although our study
`and those of others suggest
`that
`the reduced
`proliferation is specific to GA, the proliferation of
`GA and TT-stimulated PBMCs did not differentiate
`GA-responders
`from the hypo/non-responders.
`This raises the possibility of a mechanism unre-
`lated to proliferation involved in the differential
`
`Multiple Sclerosis 2007; 13: 754 762
`
`http://msj.sagepub.com
`
`Downloaded from
`
`
`
` by guest on November 18, 2015msj.sagepub.com
`
`Page 5 of 9
`
`YEDA EXHIBIT NO. 2124
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Clinical response to GA
`
`759
`
`GA-HR/NR (n=10)
`
`*
`
`*
`
`Pre-Rx 3mo
`
`6mo 12mo 24mo
`
`on Rx
`
`Pre-Rx
`
`3mo
`
`6mo
`
`12mo
`
`24mo
`
`on Rx
`
`75
`
`50
`
`25
`
`0
`
`b
`
`IL-4 spots per 4x105 cells
`
`600
`
`500
`
`400
`
`300
`
`200
`
`100
`
`0
`
`d
`
`IFN-γ spots per 4x105 cells
`
`f
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`IL-4/IFN-γ Ratio
`
`aG
`
`A-R (n=10)
`
`Pre-Rx 3mo
`
`6mo 12mo 24mo
`on Rx
`
`*
`
`*
`
`Pre-Rx 3mo
`
`6mo
`
`12mo 24mo
`
`on Rx
`
`*
`
`*
`
`*
`
`75
`
`50
`
`25
`
`0
`
`IL-4 spots per 4x105 cells
`
`c
`
`600
`
`500
`
`400
`
`300
`
`200
`
`100
`
`0
`
`IFN-γ spots per 4x105 cells
`
`e
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`
`IL-4/IFN-γ Ratio
`
`Pre-Rx 3mo
`
`12mo
`6mo
`on Rx
`
`24mo
`
`0.0
`
`Pre-Rx
`
`3mo
`
`24mo
`
`12mo
`6mo
`on Rx
`Figure 1 Comparison of the mean IL-4 and IFN-g spots among GA-R (a, c) and GA-HR/NR (b, d) with anti-CD3/CD28
`stimulation before treatment and at three, six, 12 and 18 24 months during therapy. A comparison of the mean IL-4/IFN-g
`ratio before and during treatment among clinical responders (e) and non-responders (f) is also shown. Asterisk indicates
`statistically significant difference from pre-treatment (PB/0.05). The IL-4/IFN-g ratio was calculated as follows: first, the mean
`IL-4/IFN-g ratio at each time point during treatment for individual patient was determined. Then the average of the ratios at
`time points ranging from three to 24 months was determined for each patient. The IL-4/IFN-g ratio shown in (e) and
`(f) represent the mean of the ratios for each group.
`
`response to the drug among responders and hypo/
`non-responders.
`The cytokine shifts observed during GA treat-
`ment are possibly one of the drug’s key mechanisms
`of action [6,18,21,24 28]. Our data revealed that
`
`cytokine expression in unstimulated ex vivo PBMC
`did not differentiate the GA-responders from the
`hypo/non-responders, nor did ex vivo PBMCs sti-
`mulated in vitro with GA at concentrations of
`40 and 100 ug/mL, prior to and during treatment.
`
`http://msj.sagepub.com
`
`Multiple Sclerosis 2007; 13: 754 762
`
`Downloaded from
`
`
`
` by guest on November 18, 2015msj.sagepub.com
`
`Page 6 of 9
`
`YEDA EXHIBIT NO. 2124
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`anti-CD3/CD28 which induces a global T-cell res-
`ponse is more likely to reveal differences between
`GA-R and GA-HR/NR, as demonstrated in this study.
`The latter also suggest that GA-reactive T-cells pro-
`duce a systemic bystander effect on Th1 cells, since
`IFN-g expression was significantly reduced in anti-
`CD3/CD28-stimulated PBMC during treatment.
`In a sub-group of patients (GA-R/10 and GA-
`HR/NR/6) pre-treatment and post-treatment IL-4
`and IFN-g expression was compared. The cytokine
`changes observed among GA-R with anti-CD3/
`CD28 stimulation were predominantly a reduction
`in the frequency of IFNg spots without an increase
`in IL-4 expression. Consequently,
`the increase
`observed in the IL-4/IFNg ratio among GA-R was
`mainly driven by the reduction in the occurrence of
`IFN-g spots. It is noteworthy that IL-4 expression
`was reduced during treatment in the GA-HR/NR
`group, and was unchanged in the GA-R group.
`While the mechanism of
`this differential
`IL-4
`response to GA is not clear, it suggests heterogene-
`ity in the patients’ response to GA, which may
`underlie their clinical response to the drug. It also
`suggests that a Th2 skew favors a clinical response
`to GA. Hong et al. recently proposed a mechanism
`of action of GA that involves GA-induced regula-
`tory T-cells (CD4 CD25 FoxP3) [30]. These
`T-reg cells, induced in response to GA therapy, can
`potentially explain the reduction in the number of
`IFN-g positive ELISPOTS. Although we have not
`specifically examined these regulatory cells in our
`samples, it is possible that their induction may
`correlate with the clinical response status of the
`patients. In the absence of activation of IL-4, a Th2
`cytokine, a question arises as to the role of GA-
`specific induction of Th2 cells in the suppression of
`MS, and whether reduction in IFN-g plays a more
`important role in GA therapy. We believe that
`while our findings highlight the importance of
`IFN-g reduction in the clinical response, and the
`association of IL-4 suppression with hypo-respon-
`siveness, it does not preclude a role for GA-reactive
`Th2 cells in mediating the therapeutic effect, since
`GA-reactive Th2 cells have been demonstrated in
`multiple laboratories, and are likely to be present in
`low frequencies, that are undetectable in our assay.
`Since the goal of immunomodulation in MS is to
`prevent relapses and disability, we correlated the
`patients’ annual relapse rate with IL-4 and IFN-g
`expression and the IL-4/IFN-g ratio. The results
`showed no significant correlation between ARR
`and the Th2 cytokine IL-4. However, a positive
`correlation between ARR and the Th1 cytokine IFN-
`g, and a significant inverse correlation between ARR
`and IL-4/IFN-g ratio were observed. These findings
`suggest that modulation of IFN-g and IL-4 expres-
`sion contribute to the beneficial clinical effect of
`this therapy.
`
`760
`
`RM Valenzuela et al.
`
`p>0.05
`
`0
`
`50 100 150 200 250 300 350
`IL-4
`
`p = 0.040
`
`0
`
`100
`
`200
`IFN-γ
`
`300
`
`400
`
`p = 0.041
`
`0.5
`
`0.4
`
`0.3
`
`0.2
`
`0.1
`
`0.0
`
`a
`
`Annula Relapse Rate
`
`b
`
`1.75
`
`1.50
`
`1.25
`
`1.00
`
`0.75
`
`0.50
`
`0.25
`
`0.00
`
`1.75
`
`1.50
`
`1.25
`
`1.00
`
`0.75
`
`0.50
`
`0.25
`
`0.00
`
`Annula Relapse Rate
`
`c
`
`Annula Relapse Rate
`
`0.0
`
`0.5
`
`2.0
`1.5
`1.0
`IL-4/IFN-γ Ratio
`
`2.5
`
`3.0
`
`Figure 2 Linear regression analysis of ARR against (a) IL-4,
`(b) IFN-g and (c) IL-4/IFN-g ratio using Spearman correlation.
`
`However, the mean number of IL-4 and IFN-g spots
`in PBMC stimulated with anti-CD3/CD28 was dif-
`ferent between the two groups. A possible explana-
`tion for the unobtainable differences with GA
`stimulation is that the precursor frequency of
`GA-reactive T-cells is very low following chronic
`treatment, so that GA-stimulation ex vivo may
`not elicit detectable differences in cytokine expres-
`sion between GA-R and GA-HR/NR. Such differences
`may be detectable if one generates GA-reactive T-cell
`lines from GA-treated patients [18,19]. In contrast,
`
`Multiple Sclerosis 2007; 13: 754 762
`
`http://msj.sagepub.com
`
`Downloaded from
`
`msj.sagepub.com
`
` by guest on November 18, 2015
`
`Page 7 of 9
`
`YEDA EXHIBIT NO. 2124
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Clinical response to GA
`
`761
`
`*
`
`TT
`
`*
`
`GA
`
`*
`
`Pre-Rx
`N=16
`
`3mo
`24
`
`6mo
`21
`
`12mo
`17
`
`24mo
`22
`
`GLATIRAMER ACETATE
`
`GA-R
`
`GA-HR/NR
`
`b
`
`50
`
`40
`
`30
`
`20
`
`10
`
`0
`
`10.0
`
`7.5
`
`5.0
`
`2.5
`
`0.0
`
`STIMULATION INDEX
`
`d
`
`STIMULATION INDEX
`
`a
`
`50000
`
`40000
`
`30000
`
`20000
`
`10000
`
`0
`
`*
`
`TT
`
`*
`
`*
`
`GA
`
`Pre-Rx
`N=16
`
`3mo
`24
`
`6mo
`21
`
`12mo
`17
`
`24mo
`22
`
`GLATIRAMER ACETATE
`
`15000
`
`12000
`
`9000
`
`6000
`
`3000
`
`0
`
`GA-R
`
`GA-HR/NR
`
`∆CPM
`
`c
`
`∆CPM
`
`Pre-Rx 3mo 6mo 12mo 24mo
`
`Pre-Rx 3mo 6mo 12mo 24mo
`
`10
`R
`15
`14
`10
`13
`HR/NR 6
`9
`7
`7
`9
`Figure 3 Proliferative response of PBMC among MS patients expressed as DCPM (a, c) and as stimulation index (b, d) after
`stimulation with TT 5 mg/mL (a, b) and GA 40 mg/mL (a d) before treatment and at different time points during GA treatment.
`(a) and (b) show data for the entire patient group, while (c) and (d) compare the results between clinical responders (n/10)
`and hypo/non-responders (n/6).
`
`R
`15
`10
`HR/NR 6 9
`
`14
`7
`
`10
`7
`
`13
`9
`
`In the European/Canadian GA study group by
`Comi et al.
`[15],
`it was demonstrated that the
`number of enhancing lesions and accumulation of
`new T2 lesions begin to decrease at six months
`following GA treatment. Our findings of cytokine
`changes among GA-R showed a significant increase
`in the IL-4/IFN-g ratio starting six months after
`treatment, which was not observed among the GA-
`HR/NR group. Thus, the time course of the immu-
`nological changes induced by GA-therapy seem to
`parallel the time course of the changes on MRI. These
`observations further support the significance of the
`IL-4/IFN-g changes in the clinical response to GA.
`In summary, new findings have emerged from
`this study: First, the lymphoproliferative response
`to GA did not differentiate the GA-responders from
`the hypo/non-responders. Second, the Th1 cyto-
`kine IFN-g (in anti-CD3/CD28-stimulated PBMC)
`was significantly lower among the GA-responders
`compared to hypo/non-responders during GA ther-
`apy, and was the main driving force towards a
`significant difference in the IL-4/IFN-g ratio ob-
`served between GA-R and GA-HR/NR. Third, a
`reduction in the expression of the Th2 cytokine
`IL-4 seems to be associated with hypo/non-respon-
`ders to GA. Fourth, an increased IL-4/IFN-g ratio
`
`and a decreased IFN-g cytokine response correlate
`with a lower annual relapse rate.
`The mechanism underlying the differential re-
`sponse to GA between the responders and hypo/
`non-responders is not clear, but could involve
`transcriptional regulation of cytokine genes and
`the HLA background of the patients among other
`factors [23]. More data are needed to validate the
`prospective use of IL-4/IFN-g expression in PBMC as
`a biomarker of clinical response to GA for indivi-
`dual patients.
`
`Acknowledgements
`
`This study was supported by grants to Dr Dhib-
`Jalbut from The National Institutes of Health (K24-
`N502082), the Wadsworth Foundation and Teva
`Pharmaceuticals Inc.
`
`References
`
`1. Fridkis-Hareli M, Aharoni R, Teitelbaum D, Arnon
`R, Sela M, Strominger JL. Binding of random copoly-
`mers of three amino acids to class II MHC molecules. Int
`Immunol 1999; 11: 635 41.
`
`http://msj.sagepub.com
`
`Multiple Sclerosis 2007; 13: 754 762
`
`Downloaded from
`
`msj.sagepub.com
`
` by guest on November 18, 2015
`
`Page 8 of 9
`
`YEDA EXHIBIT NO. 2124
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`762
`
`RM Valenzuela et al.
`
`2. Fridkis-Hareli M, Neveu JM, Robinson RA, Lane
`WS, Gauthier L, Wucherpfennig KW et al . Binding
`motifs of copolymer 1 to multiple sclerosis and rheuma-
`toid arthritis-associated HLA-DR molecules. J Immunol
`1999; 162: 4697 704.
`Fugger L,
`3. Fridkis-Hareli M, Rosloniec EF,
`Strominger JL. Synthetic amino acid copolymers that
`bind to HLA-DR proteins and inhibit type II collagen-
`reactive T cell clones. Proc Natl Acad Sci USA 1998; 95:
`12528 31.
`4. Fridkis Hareli M, Santabrogio L, Stern JN, Fugger
`L, Brosnan C, Strominger JL. Novel synthetic amino
`acid copolymers that inhibit autoanitigen-specific T cell
`responses and suppresses EAE. J Clin Invest 2002; 109:
`1635 43.
`5. Fridkis-Hareli M, Stern JN, Fugger L, Strominger
`JL. Synthetic peptides that inhibit binding of myelin
`basic protein 85-89 epitope to MS associated HLA-DR2
`molecules and MBP-specific T cell responses. Hum Im-
`munol 2001; 62: 753 63.
`6. Aharoni R, Teitelbaum D, Sela M, Arnon R. By-
`stander suppression of experimental autoimmune ence-
`phalomyelitis by T-cell lines and clones of the Th2 type
`induced by copolymer 1. J Neuroimmunol 1998; 91: 1358.
`7. Aharoni R, Teitalbaum D, Sela M, Amon R. Copo-
`lymer 1 induces T cells of the T-helper type 2 that cross-
`react with myelin basic protein and suppress experimen-
`tal autoimmune encephalomyelitis. Proc Natl Acad Sci
`USA 1997; 94:10821.
`8. Arnon R. The development of Cop 1 (Copaxone), an
`innovative drug for the treatment of multiple sclerosis:
`personal reflections. Immunol Lett 1996; 50: 1 15.
`9. 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 experi-
`mental autoimmune encephalomyelitis by copolymer-1.
`Proc Natl Acad Sci USA 2000; 97: 11472 77.
`10. Dhib-Jalbut SS. Glatiramer acetate (Copaxone) therapy
`for multiple sclerosis. Pharmacol Therapeut 2003; 245 55.
`11. Gran B, Tranquill LR, Chen M, Bielekova B, Zhou
`W, Dhib-Jalbut SS et al . Mechanisms of immunomo-
`dulation by glatiramer acetate. Neurology 2000; 55:1704
`14.
`12. Johnson KP, Brooks BR, Cohen JA, Ford C, Gold-
`stein J, Lisak RR et al . Copolymer 1 reduces relapse rate
`and improves disability in relapsing-remitting multiple
`sclerosis: results of phase III multicenter, double-blind,
`placebo-controlled trial. Neurology 1995; 45: 105.
`13. Miller AS, Shapiro R, Gershtein A, Kinarty H,
`Rawashdeh S, Honigman S et al . Treatment of multi-
`ple sclerosis with copolymer-1 (Copaxone): implicating
`mechanisms of Th1 to Th2/Th3 immune deviation.
`J Neuroimmunol 1998; 92: 113 21.
`14. Qin Y, Zhang DQ, Prat A, Pouly S, Antel J.
`Characterization of T-cell lines derived from glatiramer
`acetate-treated multiple sclerosis patients. J

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