throbber
Neuroscience Letters 406 (2006) 270–275
`
`Effect of glatiramer acetate (Copaxone®) on the immunophenotypic and
`cytokine profile and BDNF production in multiple sclerosis:
`A longitudinal study
`Y. Blanco a,b, E.A. Moral c,d, M. Costa e, M. G´omez-Choco a,b, J.F. Torres-Peraza b,f,
`L. Alonso-Magdalena c,d, J. Alberch b,f, D. Jaraquemada e, T. Arbizu c,d,
`F. Graus a,b, A. Saiz a,b,∗
`
`a Service of Neurology, Hospital Cl´ınic, Universitat de Barcelona, Villarroel 170, 08036 Barcelona, Spain
`b Institut d’Investigaci´o Biom`edica August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villarroel 170, 08036 Barcelona, Spain
`c Service of Neurology, Hospital Universitario de Bellvitge, Avda. Feixa Llarga s/n, 08907, L’Hospitalet de Llobregat, Barcelona, Spain
`d Institut dˇıInvestigaci´o Biom`edica Bellvitge (IDIBELL), Avda. Feixa Llarga s/n, 08907, L’Hospitalet de Llobregat, Barcelona, Spain
`e Immunology Unit, Institut de Biotecnologia i Biomedicina, Universitat Aut`onoma de Barcelona, Spain
`f Department of Cell Biology and Pathology, Universitat de Barcelona, Barcelona, Spain
`Received 2 June 2006; received in revised form 11 July 2006; accepted 21 July 2006
`
`Abstract
`
`We assessed the effect of glatiramer acetate (GA) on the immunophenotypic and cytokine profile and the BDNF production by peripheral
`blood mononuclear cells, and their association with the clinical response in 19 na¨ıve-treated MS patients prospectively followed-up after GA
`therapy. Two patients withdrew the therapy. After a median follow-up of 21 months, twelve were considered responders and five as non-responders.
`Non-responder patients had significant longer disease duration and a higher EDSS score at baseline. In the responder group, a significant decrease
`in the percentage of INF-␥ producing total lymphocytes, CD4+ and CD8+ T cells, and reduced percentage of IL-2 producing CD4+ and CD8+ T
`cells were observed at 12, 18 and 24 months. These changes were associated with a significant increase in the percentage of CD3+, CD4+ and
`CD4+CD45RA+ T cells, and BDNF production from month 6 that remained significant throughout the study. We did not observe significant changes
`in the nonresponder group for any of the parameters studied. Our data suggest that GA treatment induces a downmodulation of proinflammatory
`cytokines associated with the regulation of the peripheral T cell compartment and with increased production of BDNF that might be related to the
`clinical response.
`© 2006 Elsevier Ireland Ltd. All rights reserved.
`
`Keywords: Multiple sclerosis; Glatiramer acetate; BDNF; Cytokines; Lymphocyte immunophenotyping; Immunomodulatory treatment
`
`Glatiramer acetate (GA, Copaxone®) is an approved agent for
`the immunomodulatory treatment of relapsing-remitting mul-
`tiple sclerosis (RRMS). Although the mechanisms of action
`of GA are not fully understood, recent studies support both
`immunomodulatory and neuroprotective effects of the drug
`[17,24]. Besides the induction of anergy in autoreactive T
`cells, GA induces a shift of GA-reactive T cells from a Th1
`to a Th2 phenotype [3,4,10,13,18] that are thought to release
`anti-inflammatory cytokines but also neurotrophic factors such
`
`∗
`
`Corresponding author at: Service of Neurology, Hospital Clinic, Villarroel
`170, Barcelona 08036, Spain. Tel.: +34 932275414; fax: +34 932275783.
`E-mail address: asaiz@clinic.ub.es (A. Saiz).
`
`0304-3940/$ – see front matter © 2006 Elsevier Ireland Ltd. All rights reserved.
`doi:10.1016/j.neulet.2006.07.043
`
`as brain-derived neurotrophic factor (BDNF) within the CNS
`[1,3,25]. However, most of these immunological effects are
`based on the results of the analysis of GA-specific T-cell lines
`and less is known on the ex vivo systemic effects on periph-
`eral blood mononuclear cells (PBMC) [9,11,15,16,23]. The aim
`of the present study was to assess the effect of GA on the
`immunophenotypic and cytokine profile and the BDNF produc-
`tion by PBMC, and to analyze the association of these param-
`eters with the clinical response in a group of RRMS patients
`prospectively followed-up after GA treatment.
`Nineteen na¨ıve-treated patients with RRMS according to the
`Poser criteria [19] from the MS Units of the two participant
`hospitals were included in the study. All patients began treat-
`ment with GA because they had active disease with two or
`
`Page 1 of 6
`
`YEDA EXHIBIT NO. 2125
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Y. Blanco et al. / Neuroscience Letters 406 (2006) 270–275
`
`271
`
`Table 1
`Baseline clinical characteristics of responder and non-responder patients to GA therapy
`
`Overall
`
`Responders
`
`Non-respondersa
`
`pb
`
`19
`14/5
`34.7± 9.4
`5.6± 3.3
`1.2± 0.6
`1.5± 1.3
`
`12
`9/3
`34.8± 8.0
`3.8± 0.9
`1.2± 0.6
`1.4± 0.8
`
`more relapses in the previous two years and with an Expanded
`Disability Status Scale (EDSS) score between 0 and 5.5. Base-
`line clinical characteristics are summarized in Table 1. Patients
`were regularly followed every 3 months and additional assess-
`ments were done in the event of a relapse. During the follow-up,
`patients who presented identical or higher annual relapse rate
`and/or increase of at least 1 EDSS point confirmed at six months,
`were defined as nonresponders to GA. PBMC were collected
`before treatment, at 6 months after GA therapy and then every 6
`months. In case of coincidental relapse, the sample was taken 1
`month after steroid administration. The study was approved by
`the Ethical Committee of the two participating hospitals.
`PBMC phenotypical characterization was analyzed using a
`FACSCalibur flow cytometer (Becton Dickinson Immunocy-
`tometry Sistems (BDIS), San Jos´e, CA, USA), as previously
`reported [2]. Monoclonal antibodies to the following surface
`markers were used: CD3, CD8, CD4, CD19, HLA-DR, CD62-
`L, CD45RA, CD45RO, (BD Biosciences, San Jose, CA, USA),
`and CD25 (BD, Pharmingen, San Diego, CA, USA).
`Intracellular cytokine production was detected by flow
`cytometry as previously described [2,7] with slight modifi-
`cations. Briefly, fresh peripheral blood was incubated for 4 h
`with phorbol 12-myristate (PMA) (25 ng/ml, Sigma–Aldrich,
`St. Louis) and ionomycin (1 ␮g/ml, Sigma–Aldrich) to stim-
`ulate cytokine production. Stimulated and unstimulated cells
`were treated with Brefeldin A (10 ␮g/ml, Sigma–Aldrich), an
`inhibitor of secretion, followed by superficial staining, cell fix-
`ation, permeabilization, and then intracytoplasmic staining for
`detection of the accumulated cytokines. T cells were first gated
`by their expression of CD8 or CD4 followed by the detection of
`IFN-␥, IL-2 (as markers of Th1 phenotype) and IL-4 (as marker
`of Th2 phenotype) (BD, Pharmingen, San Diego, CA, USA)
`positive cells.
`BDNF release levels were measured in the supernatants from
`PBMC unstimulated and stimulated with anti-CD3 and soluble
`anti-CD28 antibodies by using an ELISA kit (Promega, Madi-
`son, USA) as previously described [2].
`Non-parametric tests were used for all data comparisons:
`Friedman and Wilcoxon signed ranks tests for paired patients
`groups. Pearson’s correlation coefficient was used for the corre-
`lation analysis. Significance levels were set at 5% (p < 0.05).
`Two patients withdrew the treatment because of adverse
`events in the first 3 months. After a median follow-up of 21
`months, twelve patients were considered as responders and 5 as
`nonresponders. Responder patients had a decrease of the annu-
`
`5
`4/1
`40.2± 11.3
`8.1± 2.6
`1.0± 0.3
`3.1± 1.2
`
`ns
`ns
`<0.0001
`ns
`<0.0001
`
`Number of patients
`Gender (W/M)
`Age (mean± S.D.; years)
`Disease duration (mean± S.D.; years)
`Annual relapse rate (mean± S.D.; in prior 2 years)
`Baseline EDSS (mean± S.D.)
`a Two patients withdrew the treatment in the first 3 months.
`b Refers to comparison of clinical variables between responder and non-responder patients.
`alized relapse rate (0.3± 0.4 during treatment versus 1.2± 0.6
`before treatment) and did not present significant changes in the
`EDSS score (1.4± 0.8 at initiation of treatment versus 1.6± 0.8
`at last follow-up). Non-responders had a mean of 1.3± 0.9 annu-
`alized relapse rate (1.0± 0.3 before treatment), and a mean
`increase of 0.6± 0.9 in the EDSS score (3.1± 1.2 at initiation
`of treatment versus 4.1± 1.4 at last follow-up). The only sig-
`nificant difference between both groups was that nonresponders
`patients had longer disease duration and a higher EDSS score at
`initiation of GA therapy (p < 0.0001) (Table 1).
`Results of the immunophenotypic and cytokine profile along
`the evolution are presented as mean± standard deviation in
`Tables 2 and 3 for both groups. There were no significant
`baseline differences between both groups of patients for any
`of the parameters studied. After GA therapy a significant
`increase in the percentage of CD3+, CD4+ and CD4+CD45RA+
`(na¨ıve) T cells was seen at 6, 12, 18 and 24 months in
`the responder group of patients compared with that found
`before treatment (p =0.003, p = 0.012, p = 0.05; p = 0.002,
`p = 0.01, p = 0.038; p = 0.002, p = 0.005, p = 0.036; p = 0.021,
`p = 0.05, p = 0.028; respectively). The na¨ıve/memory CD4+ T
`cells (CD4+CD45RA+/CD4+CD45RO+) ratio also increased
`at 12 months and remained significant during the follow-up
`(p = 0.028, p = 0.026, p = 0.011). No significant changes were
`observed in the percentage of CD4+CD25+ T cells. No sig-
`nificant modification in T cells population frequencies were
`detected over the study period in the nonresponder group as
`compared with baseline values.
`A significant decrease in the number of IFN-␥ producing
`CD3+, CD4+ and CD8+ T cells was observed in the respon-
`der group at 12, 18 and 24 months after GA therapy, com-
`pared with pretreatment values (p =0.028, p = 0.008, p = 0.008;
`p = 0.024, p = 0.008, p = 0.011; p = 0.012, p = 0.012, p = 0.012;
`respectively) (see Fig. 1 and Table 1). In the same way, the num-
`ber of IL-2 producing CD4+ and CD8+ T cells also decreased
`(p =0.013, p = 0.021; p = 0.037, p = 0.037; p = 0.036, p = 0.028;
`respectively). No significant changes were observed in the num-
`ber of IL-4 producing CD4+ and CD8+ T cells. In the nonre-
`sponder patients’ group, no significant differences were found
`over the study period as compared with baseline values.
`A significant increase of mean levels of BDNF in the super-
`natants of unstimulated PBMC was observed in the responder
`group from month 6 and throughout the follow-up after GA
`therapy as compared with baseline (Fig. 2). Similar results
`were found after stimulation with anti-CD3 and soluble anti-
`
`Page 2 of 6
`
`YEDA EXHIBIT NO. 2125
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Y. Blanco et al. / Neuroscience Letters 406 (2006) 270–275
`
`Fig. 1. An example of the flow cytometry analysis of cytoplasmic IFN-␥ and IL-4 expression by CD4+ and CD8+ T cells of a responder patient before and after GA treatment, as described in methods. Quadrants
`were set based on the isotype control and on unstimulated samples. Values represent the percentage of positive cells.
`
`Page 3 of 6
`
`YEDA EXHIBIT NO. 2125
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Table 2
`Peripheral T cell population before and after GA treatment
`
`Y. Blanco et al. / Neuroscience Letters 406 (2006) 270–275
`
`273
`
`R
`NR
`
`R
`NR
`
`R
`NR
`
`R
`NR
`
`R
`NR
`
`R
`NR
`
`R
`NR
`
`R
`NR
`
`CD3+
`
`CD4+
`
`CD4+CD45RA+
`
`CD4+CD45RO+
`
`RA+/RO+ ratioa
`
`CD8+
`
`CD8+CD45RA+
`
`CD8+CD45RO+
`
`24 months
`18 months
`12 months
`6 months
`Baseline
`76.1 ± 3.3a
`71.0 ± 5.1
`75.7 ± 5.5a
`76.5 ± 4.8a
`75.2 ± 3.9a
`77.2 ± 1.2
`76.1 ± 1.1
`79.8 ± 3.6
`75.4 ± 3.3
`76.1 ± 7.4
`54.4 ± 7.8a
`53.7 ± 7.5a
`53.9 ± 7.7a
`53.3 ± 6.1a
`47.8 ± 9.1
`54.1 ± 9.9
`45.4 ± 11.9
`51.2 ± 8.9
`47.7 ± 5.2
`51.5 ± 6.1
`43.9 ± 10.1a
`35.3 ± 16.0
`42.9 ± 9.4a
`43.5 ± 10.2a
`44.8 ± 12.8a
`40.2 ± 18.4
`30.6 ± 21.5
`48.1 ± 17.5
`34.1 ± 12.2
`32.6 ± 15.0
`37.8 ± 9.0
`40.6 ± 9.1
`38.8 ± 8.1
`38.8 ± 6.5
`39.3 ± 8.0
`46.0 ± 9.4
`45.8 ± 11.0
`41.5 ± 17.8
`43.4 ± 7.4
`49.7 ± 14.0
`1.2 ± 0.4a
`1.1 ± 0.4a
`1.2 ± 0.4a
`1.2 ± 0.4
`0.9 ± 0.5
`0.9 ± 0.3
`0.7 ± 0.6
`1.1 ± 0.8
`0.7 ± 0.3
`0.8 ± 0.7
`21.3 ± 5.0
`21.2 ± 6.5
`21.0 ± 5.9
`22.5 ± 5.6
`21.0 ± 7.0
`24.1 ± 9.7
`30.7 ± 13.1
`27.4 ± 9.9
`26.2 ± 11.6
`26.7 ± 8.0
`57.8 ± 10.5
`57.2 ± 12.4
`60.2 ± 9.7
`6.3.3 ± 11.0
`58.4 ± 11.7
`59.6 ± 17.3
`49.0 ± 22.4
`54.5 ± 23.7
`58.1 ± 18.8
`53.7 ± 17.0
`23.5 ± 5.7
`24.8 ± 9.8
`21.7 ± 6.6
`24.2 ± 10.5
`23.9 ± 8.3
`23.0 ± 2.7
`19.8 ± 3.0
`17.8 ± 4.5
`22.9 ± 4.9
`23.2 ± 5.9
`31.3 ± 6.7
`28.5 ± 6.3
`28.7 ± 8.9
`31.5 ± 8.9
`31.7 ± 11.3
`R
`31.0 ± 5.1
`24.3 ± 6.4
`29.5 ± 4.8
`31.5 ± 8.5
`39.1 ± 6.3
`CD4+CD25+
`NR
`Results are expressed as mean± standard deviation of the percentage of positive cells for each surface marker; R = responder group; NR = non-responder group.
`a CD4+CD45RA+ to CD4+CD45RO+ T cells ratio.
`
`CD28 antibodies (data not shown). No significant changes were
`observed for the nonresponder group (Fig. 2).
`No significant correlation was found between basal immuno-
`logical data and clinical features (sex, age, disease duration,
`annualized relapse rate and EDSS score), nor there was any
`significant correlation between immunological measurements
`on treatment and clinical outcome (number of relapses, change
`of the annualized relapse rate, increase of the EDSS score
`and last EDSS score). None of the clinical and immunological
`data analyzed could significantly predict responsiveness to GA
`therapy.
`
`Our study shows that in vivo therapy with GA induces a down-
`modulation of proinflammatory cytokine-producing T cells in
`the periphery, and increases the BDNF production by unstimu-
`lated PBMC but also after stimulation involving T-cell receptor-
`mediated activation, and this effect is sustained over time but
`only observed in patients with clinical response to the drug.
`These data are in line with the results previously shown by
`the more complex method of analysis of GA-specific T-cell
`lines [1,3,4,10,13,18,25], although few immunological longi-
`tudinal studies with clinical correlation such as we present have
`been reported so far [22]. In this sense, other authors observed
`
`Table 3
`Intracellular cytokine staining profile before and after GA treatment
`
`12 m
`
`18 m
`
`IFN-␥ CD3+
`
`IFN-␥ CD4+
`
`IFN-␥ CD8+
`
`IL-2 CD4+
`
`IL-2 CD8+
`
`IL-4 CD4+
`
`R
`NR
`
`R
`NR
`
`R
`NR
`
`R
`NR
`
`R
`NR
`
`R
`NR
`
`6 m
`Baseline
`14.6 ± 5.5
`19.7 ± 9.8
`0.024
`0.028
`28.4 ± 11.4
`15.1 ± 12.6
`20.2 ± 17.1
`24.0 ± 17.6
`14.1 ± 8.7
`20.6 ± 9.4
`0.008
`0.008
`18.0 ± 5.8
`11.3 ± 2.5
`14.2 ± 8.9
`23.5 ± 12.4
`39.1 ± 14.8
`47.3 ± 18.1
`0.011
`0.008
`67.9 ± 14.8
`45.2 ± 16.3
`42.4 ± 30.2
`51.1 ± 27.5
`50.4 ± 15.4
`40.9 ± 17.5
`0.037
`0.013
`45.1 ± 12.4
`43.0 ± 14.6
`42.3 ± 5.6
`44.3 ± 11.9
`22.3 ± 9.6
`17.2 ± 11.5
`0.037
`0.021
`17.3 ± 3.2
`15.4 ± 2.8
`19.8 ± 3.5
`14.6 ± 2.6
`2.7 ± 1.1
`4.7 ± 2.5
`3.3 ± 0.8
`3.0 ± 2.0
`4.2 ± 2.5
`3.5 ± 1.1
`3.7 ± 3.4
`3.3 ± 1.3
`2.5 ± 1.5
`3.4 ± 2.7
`2.4 ± 1.4
`3.4 ± 1.2
`R
`2.9 ± 0.4
`1.8 ± 0.6
`2.5 ± 0.6
`2.0 ± 1.8
`IL-4 CD8+
`NR
`Results are expressed as mean± standard deviation of the percentage of cells staining positively. R = responder group; NR = non-responder group.
`
`24 m
`
`0.012
`20.9 ± 15.2
`0.012
`12.7 ± 9.7
`0.012
`49.9 ± 15.0
`0.036
`47.9 ± 11.7
`0.026
`13.9 ± 2.7
`1.1 ± 1.0
`3.8 ± 1.3
`1.7 ± 1.0
`1.0 ± 0.5
`
`Page 4 of 6
`
`YEDA EXHIBIT NO. 2125
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`274
`
`Y. Blanco et al. / Neuroscience Letters 406 (2006) 270–275
`
`Fig. 2. BDNF levels in the supernatant of unstimulated PBMC of responder (left graph) and non-responder (right graph) group before and after GA treatment.
`Results are expressed in pg/ml. Time is expressed in months; time 0 refers to baseline. p-values are calculated in comparison with baseline and corrected for multiple
`comparisons. S.D.: standard deviation.
`
`an increase in the IFN-␥ secretion in responders to treatment;
`however the study was cross-sectional and retrospective [12].
`Interesting, our significant immune changes started at month 6
`for the immunophenotypic change and BDNF production, and
`at month 12 for the Th1 cytokine reduction, consistent with a
`delayed mechanism of action and a radiologic response, as was
`seen in the serial magnetic resonance imaging-based clinical
`trial [6].
`However, the most interesting data we found was that the
`reduction of Th1-type cell number was accompanied by an in
`vivo change in the peripheral T cell compartment characterized
`by a significant increase of CD4+CD45RA+ (na¨ıve) T cell fre-
`quency associated with an increased RA+/RO+ (na¨ıve/memory)
`ratio. A finding that extends a previous in vitro study that showed
`that the response to GA was driven by the CD4+CD45RA+ T
`cell subpopulation [23].
`The relevance of this increased frequency of peripheral
`CD4+CD45RA+ T cells after GA therapy to the clinical response
`we observed is intriguing. Nevertheless, in MS patients the na¨ıve
`T cell frequency is lower than average [8], and have a reduced
`TREC (TCR excision circles) content in circulating T cells,
`especially for CD4+ T cells, [14], suggesting that the release of
`na¨ıve T cells from the thymus could be impaired in MS. Because
`CD4+CD45RA+ T cells number as well as TCR levels decline
`with age [14,21] it could be that the results we found were related
`to the different, although non significant, age of both groups of
`patients. However, it is unlikely because we did not observe an
`increase in the frequency of CD4+CD45RA+ T cells in 4 addi-
`tional RRMS patients followed at least for 1 year after treatment
`with IFN-␤ who had a similar age (mean, 34.25± 7.93 years)
`to the GA responder group. Interesting, all four patients were
`considered as responders to IFN-␤ by using the same clinical
`definition.
`Finally, if the differences we found on GA therapy merely
`reflect that the two patient groups were in different stages of the
`disease is unknown. However, it is likely because similar clin-
`
`ical results have been observed with other immunomodulatory
`therapies that showed higher therapeutic benefit in patients with
`shorter disease duration and lesser disability [20,5].
`In conclusion, our study suggests that in addition to down-
`modulation of proinflammatory cytokines and increased secre-
`tion of BDNF, GA therapy may induce the regulation of the
`peripheral T cell compartment. Whether this regulation con-
`tributes to the clinical efficacy of GA needs further confirmatory
`studies in a larger patient sample.
`
`Acknowledgements
`
`Supported in part by Red CIEN, and grant from Instituto
`Carlos III (PI040659). M.G.-C. is a recipient of a postresidency
`grant from the Hospital Clinic, and J.F.T.-P. is a fellow from
`Fundaci´o La Caixa.
`
`References
`
`[1] R. Aharoni, B. Kayhan, R. Eilam, M. Sela, R. Arnon, Glatiramer acetate-
`specific T cells in the brain express T helper 2/3 cytokines and brain-
`derived neurotrophic factor in situ, Proc. Natl. Acad. Sci. U.S.A. 100 (2003)
`14157–14162.
`[2] Y. Blanco, A. Saiz, M. Costa, J.F. Torres-Peraza, E. Carreras, J. Alberch,
`D. Jaraquemada, F. Graus, Evolution of brain-derived neurotrophic factor
`levels after autologous hematopoietic stem cell transplantation in multiple
`sclerosis, Neurosci. Lett. 380 (2005) 122–126.
`[3] M. Chen, K. Conway, K.P. Johnson, R. Martin, S. Dhib-Jalbut, Sustained
`immunological effects of galtiramer acetate in patients with multiple scle-
`rosis treated for over 6 years, J. Neurol. Sci. 201 (2002) 71–77.
`[4] M. Chen, R.M. Valenzuela, S. Dhib-Jalbut, Glatiramer acetate-reactive T
`cells produced brain-derived neurotrophic factor, J. Neurol. Sci 215 (2003)
`37–44.
`[5] A.J. Coles, J. Deans, A. Compston, Campath-1 H treatment of multiple
`sclerosis: lessons from the bedside for the bench, Clin. Neurol. Neurosurg.
`106 (2004) 270–274.
`[6] G. Comi, M. Filippi, J.S. Wolinsky, The European/Canadian Glatiramer
`Acetate Study Group, European/Canadian multicenter, double-blind, ran-
`domized, placebo-controlled study of the effects of glatiramer acetate
`
`Page 5 of 6
`
`YEDA EXHIBIT NO. 2125
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Y. Blanco et al. / Neuroscience Letters 406 (2006) 270–275
`
`275
`
`on magnetic resonance imaging-measured disease activity and burden in
`patients with relapsing multiple sclerosis, Ann. Neurol. 49 (2001) 290–297.
`[7] M. Costa, A. Saiz, R. Casmitjana, M.F. Casta˜ner, A. Sanmart´ı, F. Graus, D.
`Jaraquemada, T-cell reactivity to glutamic acid decarboxylase in stiff-man
`s´ındrome and cerebellar ataxia associated with polyendocrine autoimmu-
`nity, Clin. Exp. Immunol. 192 (2002) 471–478.
`[8] B. Crucian, P. Dunne, H. Friedman, R. Ragsdale, S. Pross, R. Widen,
`−
`/CD28+ suppressor cell precursor and
`Alterations in levels of CD28
`CD45RO+/CD4+ memory T lymphocytes in the peripheral blood of mul-
`tiple sclerosis patients, Clin. Diagn. Lab. Immunol. 2 (1995) 249–252.
`[9] S. Dhib-Jalbut, M. Chen, A. Said, M. Zhan, K.P. Johnson, R. Martin,
`Glatiramer acetate-reactive peripheral blood mononuclear cells respond
`to multiple sclerosis myelin antigens with a Th2-biased phenotype, J. Neu-
`roimmunol. 140 (2003) 163–171.
`[10] P.W. Duda, M.C. Schmied, S.L. Cook, J.I. Krieger, D.A. Hafler, Glatiramer
`acetate (Copaxone) induces degenerate, Th2-polarized immune responses
`in patients with multiple sclerosis, J. Clin. Invest. 105 (2000) 967–976.
`[11] C. Farina, F.T. Bergh, H. Albrecht, E. Meinl, A. Yassouridis, O. Neuhaus,
`R. Hohlfeld, Treatment of multiple sclerosis with Copaxone (COP): Elispot
`assay detects COP-induced interleukina-4 and interferon-␥ response in
`blood cells, Brain 124 (2001) 705–719.
`[12] C. Farina, S. Wagenpfeil, R. Hohlfeld, Immunological assay for assessing
`the efficacy of glatiramer acetate (Copaxone) in multiple sclerosis: a pilot
`study, J. Neurol. 249 (2002) 1587–1592.
`[13] B. Gran, L.R. Tranquill, M. Chen, B. Bielekova, W. Zhou, S. Dhib-Jalbut,
`R. Martin, Mechanisms of immunomodulation by glatiramer acetate, Neu-
`rology 55 (2000) 1704–1714.
`[14] A. Hug, M. Korporal, I. Schr¨oder, J. Haas, K. Glatz, B. Storch-Hagenlocher,
`B. Wildeman, Thymic export function and T cell homeostasis in patients
`with relapsing-remitting multiple sclerosis, J. Immunol. 170 (2003)
`432–437.
`[15] N.J. Karandikar, M.P. Crawford, X. Yan, R.B. Ratts, J.M. Brenchley, D.R.
`Ambrozak, A.E. Lovett-Racke, E.M. Frohman, P. Satstny, D.C. Douek,
`R.A. Koup, M.K. Racke, Glatiramer acetate (Copaxone) therapy induces
`CD8+ T cell responses in patients with multiple sclerosis, J. Clin. Invest.
`109 (2002) 641–649.
`
`[16] A. Miller, S. Shapiro, R. Gershtein, A. Kinarty, H. Rawahdeh, S. Honigman,
`N. Lahat, Treatment of multiple sclerosis with Copolymer-1 (Copaxone):
`implicating mechanisms of Th1 to Th2/Th3 immune-deviation, J. Neu-
`roimmunol. 92 (1998) 113–121.
`[17] O. Neuhaus, C. Farina, H. Wekerle, R. Hohlfeld, Mechanisms of action
`of glatiramer acetate in multiple sclerosis, Neurology 56 (2001) 702–
`706.
`[18] O. Neuhaus, C. Farina, A. Yassouridis, H. Wiendl, F.T. Bergh, T. Dose,
`H. Wekerle, R. Hohlfeld, 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. 97
`(2000) 7452–7457.
`[19] C.M. Poser, D.W. Paty, L. Scheinberg, W.I. McDonald, F.A. Davis, G.C.
`Ebers, K.P. Johnson, W.A. Sibley, D.H. Silberger, W.W. Tourtellote, New
`diagnostic criteria for multiple sclerosis: guidelines for research protocols,
`Ann. Neurol. 13 (1983) 227–231.
`[20] J.W. Rose, H.E. Watt, A.T. White, N.G. Carlson, Treatment of multiple
`sclerosis with an anti-interleukin-2 receptor monoclonal antibody, Ann.
`Neurol. 56 (2004) 864–867.
`[21] P. Saule, J. Trauet, V. Dutriez, V. Lekeux, J.P. Dessaint, M. Labalette, Accu-
`mulation of memory T cells from childhood to old age: central and effector
`memory cells in CD4+ versus effector memory and terminally differenti-
`ated memory cells in CD8+ compartment, Mech. Age. Dev. 127 (2006)
`274–281.
`[22] C. Weder, G.M. Baltariu, K.A. Wyler, H.J. Gober, C. Lienert, M. Schluep,
`E.W. Rad¨u, G. De Libero, L. Kappos, P.W. Duda, Clinical and immune
`responses correlate in glatiramer acetate therapy of multiple sclerosis, Eur.
`J. Neurol. 12 (2005) 869–878.
`[23] E. Wiesemann, J. Klatt, D. S¨onmez, R. Blasczyk, F. Heidenreich, A. Wind-
`hagen, Glatiramer acetate (GA) induces IL-13/IL-5 secretion in na¨ıve T
`cells, J. Neuroimmunol. 119 (2001) 137–144.
`[24] V.W. Yong, Differential mechanisms of action of interferon-␤ and glati-
`ramer acetate in MS, Neurology 59 (2002) 802–808.
`[25] T. Ziemssen, T. Kumpfel, W.E.F. Klinker, O. Neuhaus, R. Hohlfeld, Glati-
`ramer acetate-specific T-helper 1 and 2-type cell lines produce BDNF:
`implications for multiple sclerosis therapy, Brain 125 (2002) 2381–2391.
`
`Page 6 of 6
`
`YEDA EXHIBIT NO. 2125
`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