throbber
IMMUNOLOGY
`
`Glatiramer acetate increases IL-1 receptor antagonist
`but decreases T cell-induced IL-1␤ in human
`monocytes and multiple sclerosis
`
`Danielle Burgera,1, Nicolas Molnarfia,b, Martin S. Weberb,c, Karim J. Brandta, Mahdia Benkhouchad, Lyssia Gruaza,
`Michel Chofflone, Scott S. Zamvilb, and Patrice H. Lalived,e,f
`
`aDivision of Immunology and Allergy, Clinical Immunology Unit (Hans Wilsdorf Laboratory), Department of Internal Medicine, University Hospital and
`Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; bDepartment of Neurology and Program in Immunology, University of California, San
`Francisco, CA 94143; cDepartment of Neurology, Technische Universita¨ t Mu¨ nchen, D-80538 Munich, Germany; dDepartment of Pathology and Immunology,
`Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; and eDepartment of Neurosciences, Division of Neurology, Neuroimmunology
`Laboratory, and fDepartment of Genetics and Laboratory Medicine, Laboratory Medicine Service, University Hospital of Geneva, 1211 Geneva, Switzerland
`
`Edited by Michael Sela, Weizmann Institute of Science, Rehovot, Israel, and approved January 26, 2009 (received for review December 1, 2008)
`
`Mechanisms of action as well as cellular targets of glatiramer
`acetate (GA) in multiple sclerosis (MS) are still not entirely under-
`stood. IL-1␤ is present in CNS-infiltrating macrophages and micro-
`glial cells and is an important mediator of inflammation in exper-
`imental autoimmune encephalitis (EAE), the MS animal model. A
`inhibitor of IL-1␤, the secreted form of IL-1 receptor
`natural
`antagonist (sIL-1Ra) improves EAE disease course. In this study we
`examined the effects of GA on the IL-1 system.
`In vivo, GA
`treatment enhanced sIL-1Ra blood levels in both EAE mice and
`patients with MS, whereas IL-1␤ levels remained undetectable. In
`vitro, GA per se induced the transcription and production of sIL-1Ra
`in isolated human monocytes. Furthermore,
`in T cell contact-
`activated monocytes, a mechanism relevant to chronic inflamma-
`tion, GA strongly diminished the expression of IL-1␤and enhanced
`that of sIL-1Ra. This contrasts with the effect of GA in monocytes
`activated upon acute inflammatory conditions. Indeed, in LPS-
`activated monocytes, IL-1␤and sIL-1Ra production were increased
`in the presence of GA. These results demonstrate that, in chronic
`inflammatory conditions, GA enhances circulating sIL-1Ra levels
`and directly affects monocytes by triggering a bias toward a less
`inflammatory profile, increasing sIL-1Ra while diminishing IL-1␤
`production. This study sheds light on a mechanism that is likely to
`participate in the therapeutic effects of GA in MS.
`
`experimental autoimmune encephalitis 兩 cellular contact 兩
`inflammation 兩 autoimmune disease
`
`Glatiramer acetate (GA; copolymer-1; Copaxone) is composed
`
`of a mixture of synthetic peptides of 50 to 90 aa randomly
`composed of L-glutamic acid (E), L-lysine (K), L-alanine (A), and
`L-tyrosine (Y). Initially developed to mimic the myelin basic
`protein, a major component of the myelin sheath, and to induce
`experimental autoimmune encephalitis (EAE), GA unexpectedly
`inhibited EAE in both rodents and monkeys (1). In subsequent
`clinical trials, GA reduced relapse rate and progression of disability
`in patients with relapsing–remitting multiple sclerosis (MS; RRMS)
`leading to its approval in 1995 (2).
`A number of investigations in MS and EAE addressed the
`immunological basis of GA clinical effects. However, the mech-
`anisms of GA action are still elusive. Initial
`investigations
`attributed most GA activity to a preferential Th2-polarization of
`myelin-specific T cells, thus focusing on its effects on the
`adaptive immune response (3). However, recent reports indi-
`cated that GA treatment also exerts immunomodulatory activity
`on cells of the monocytic lineage, i.e., monocytes/macrophages
`and dendritic cells (4–9). For instance, monocytes from GA-
`treated patients with MS secrete less IL-12 and TNF in response
`to LPS stimulation compared with monocytes from healthy
`controls and untreated patients with MS (4). Accordingly,
`dendritic cells and monocytes isolated from GA-treated patients
`
`produce more anti-inflammatory IL-10 and less pro-inflamma-
`tory IL-12 (5, 9). Furthermore, GA promotes the development
`of anti-inflammatory type II monocytes in EAE, accompanied
`by induction of regulatory T cells and increased secretion of both
`IL-10 and TGF-␤ (10).
`IL-1␤ is a pleiotropic pro-inflammatory cytokine whose pro-
`duction is tightly controlled at several levels (11). Indeed, as
`recently reviewed, there are several roadblocks to the release of
`IL-1␤ beginning with the transcription of the IL1B gene and
`ending with the exit of the active cytokine from the cell. In the
`extracellular space, IL-1␤activity is mainly ruled by the secreted
`IL-1 receptor antagonist (sIL-1Ra), which binds type I IL-1
`receptor without triggering signals (12). As it potently inhibits
`the various effects of IL-1, sIL-1Ra is considered an important
`regulator of the inflammatory and overall immune response
`mediated by IL-1 (13). Because of its extreme efficacy as a
`pro-inflammatory mediator, if these intracellular and extracel-
`lular roadblocks are not enough to limit IL-1␤ activity, it may
`also be reduced by the preferential binding to the cell surface or
`soluble form of type II IL-1 receptor, preventing it from trig-
`gering the signal-transducing type I receptor (11). Finally, the
`facilitation of IL-1␤ processing by the caspase 1 inflammasome
`through ATP activation of the P2X7 receptor can also be viewed
`as a potential roadblock to the activity of IL-1␤ (11).
`IL-1␤ is mainly produced upon activation of cells of the mono-
`cytic lineage. In chronic/sterile immuno-inflammatory diseases, the
`factors triggering pro-inflammatory cytokine production are still
`elusive. T cells may exert a pathological effect through direct
`cellular contact with monocytes/macrophages, inducing massive
`up-regulation of IL-1␤ and TNF (14). Besides triggering pro-
`inflammatory cytokine production, contact-mediated activation of
`monocytes induces the production and/or shedding of cytokine
`inhibitors such as sIL-1Ra and soluble receptors of IL-1 and TNF
`(15). The importance of T cell contact-mediated activation of
`monocytic cells in MS was further demonstrated in vitro in co-
`cultures of T cells and microglial cells (16, 17).
`In MS, IL-1␤ is mainly expressed by microglial cells and
`infiltrating monocyte/macrophages throughout the white matter
`and in acute lesions (18). This assertion was further confirmed
`in EAE studies. Indeed, dark agouti rats treated with sIL-1Ra
`during the induction of EAE, or after adoptive transfer with
`myelin antigen-primed lymph node cells, develop milder signs of
`
`Author contributions: D.B., N.M., M.S.W., M.C., S.S.Z., and P.H.L. designed research; N.M.,
`M.S.W., K.J.B., M.B., and L.G. performed research; D.B. and P.H.L. analyzed data; and D.B.
`and P.H.L. wrote the paper.
`
`The authors declare no conflict of interest.
`
`This article is a PNAS Direct Submission.
`
`Freely available online through the PNAS open access option.
`1To whom correspondence should be addressed. E-mail: danielle.burger@hcuge.ch.
`
`www.pnas.org兾cgi兾doi兾10.1073兾pnas.0812183106
`
`PNAS 兩 March 17, 2009 兩 vol. 106 兩 no. 11 兩 4355– 4359
`
`1 of 5
`
`YEDA EXHIBIT NO. 2061
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Table 1. Clinical characteristics of patients with MS and healthy
`controls
`
`Sex
`
`Age (y)
`
`Disease
`duration
`(y)
`
`EDSS
`
`GA treatment
`duration (mo)
`
`31
`F
`34
`F
`36
`F
`29
`M
`24
`M
`24
`F
`30
`F
`35
`F
`44
`F
`40
`M
`— 32.7 ⫾ 6.4
`
`—
`—
`—
`—
`—
`—
`—
`—
`—
`—
`—
`
`—
`—
`—
`—
`—
`—
`—
`—
`—
`—
`—
`
`45
`F
`31
`F
`15
`M
`23
`F
`41
`F
`36
`F
`34
`F
`27
`M
`31
`F
`46
`F
`41
`M
`— 33.6 ⫾ 9.6
`
`17
`10
`1
`1
`6
`3
`5
`3
`10
`6
`6
`6.2 ⫾ 4.7
`
`3.0
`1.5
`1.5
`2.0
`7.0
`2.0
`2.0
`1.5
`1.0
`2.5
`4.0
`2.5 ⫾ 1.7
`
`—
`—
`—
`—
`—
`—
`—
`—
`—
`—
`—
`
`—
`—
`—
`—
`—
`—
`—
`—
`—
`—
`—
`—
`
`39
`M
`33
`F
`26
`M
`45
`F
`32
`F
`27
`F
`23
`M
`39
`M
`25
`F
`— 32.1 ⫾ 7.6
`
`6
`7
`8
`15
`11
`4
`2
`9
`5
`7.4 ⫾ 3.9
`
`2.5
`2.0
`1.5
`1.5
`2.0
`0
`1.0
`2.0
`1.0
`1.5 ⫾ 0.8
`
`56
`18
`60
`27
`13
`44
`29
`36
`19
`33.6 ⫾ 16.8
`
`Clinical
`category
`
`Healthy
`controls
`1
`2
`3
`4
`5
`6
`7
`8
`9
`10
`Mean ⫾
`SD
`Untreated
`RRMS
`1
`2
`3
`4
`5
`6
`7
`8
`9
`10
`11
`Mean ⫾
`SD
`GA-
`treated
`RRMS
`1
`2
`3
`4
`5
`6
`7
`8
`9
`Mean ⫾
`SD
`
`EDSS: Expanded Disability Status Score; RRMS: relapsing-remitting multiple
`sclerosis; GA: glatiramer acetate. Data expressed at time of sampling.
`
`were activated by increasing doses of GA. The production of
`sIL-1Ra was enhanced by GA in a dose-dependent manner,
`reaching a plateau at 25 ␮g/mL (Fig. 3A). The latter dose was used
`for the in vitro experiments described later. Noticeably, GA did not
`induce IL-1␤ production, demonstrating that GA triggers an anti-
`inflammatory bias in human monocyte cytokine production.
`To confirm that GA affected the IL-1 system, we assessed its
`effect on human monocytes activated upon chronic/sterile and
`acute/infectious inflammatory conditions as mimicked by direct
`cellular contact with stimulated T cells and LPS, respectively.
`Studies of cell-cell interactions such as those occurring in T cell
`contact activation of human monocytes are usually complicated
`by the simultaneous presence of at least 2 viable cell types. To
`obviate this problem, and possible interferences caused by the
`
`sIL-1Ra levels are elevated in sera of EAE mice treated with GA. (A) GA
`Fig. 1.
`ameliorates EAE. C57BL/6 mice were injected s.c. daily with GA (150 ␮g) or
`vehicle (PBS solution) 7 d before immunization with 10 ␮g myelin oligoden-
`drocyte glycoprotein 35–55 peptide (dpi, day post-immunization). (B) EAE
`mice treated (GA) or not (vehicle) were killed at disease peak and their serum
`analyzed for IL-1␤ and sIL-1Ra content. IL-1␤ was not detected (not shown).
`
`the disease (19). sIL-1Ra delivered by non-replicative HSV-1
`vectors in EAE C57BL/6 mice delays disease onset and decreases
`disease severity (20). In addition, IL-1␣/␤ double deficient
`(IL-1⫺/⫺) mice exhibit significant resistance to EAE induction
`with reduction in disease severity, whereas IL-1Ra⫺/⫺ mice are
`highly susceptible to EAE induction in the absence of pertussis
`toxin administration (21). These observations demonstrate that
`the IL-1/IL-1Ra system is crucial for autoantigen-specific T cell
`induction in mice and that sIL-1Ra efficiently blocks IL-1␤
`effects and ameliorates EAE disease course (19–22).
`In this study we addressed the question of the effects of GA
`on IL-1 system in vivo and in vitro. The results show that
`GA-treatment increases the circulating levels of sIL-1Ra in both
`EAE mice and patients with MS. This is reflected in vitro by the
`direct effect of GA on human blood monocytes. Indeed, GA
`induces the production of the cytokine inhibitor sIL-1Ra and
`diminishes the production of IL-1␤ in conditions related to
`chronic inflammation, i.e., in monocytes activated by direct
`contact with stimulated T cells.
`
`Results
`sIL-1Ra Serum Levels Are Elevated in GA-Treated EAE Mice. To assess
`whether GA-treatment affected sIL-1Ra levels in the MS animal
`model, EAE was induced in mice treated either with GA or PBS
`solution (i.e., vehicle). As shown in Fig. 1A, EAE severity was
`reduced in GA-treated mice, as previously demonstrated (10).
`At peak disease, mouse sera were analyzed for levels of sIL-1Ra
`and IL-1␤. IL-1␤ was not detectable in any of the sera (not
`shown). However, sIL-1Ra was significantly elevated in mice
`treated with GA (3,336 ⫾ 1,190 pg/mL sIL-1Ra, mean ⫾ SD)
`compared with animals that received vehicle as a control (1,296 ⫾
`657 pg/mL sIL-1Ra; Fig. 1B). This demonstrates that GA-treatment
`enhanced sIL-1Ra concentration in EAE mouse serum.
`
`sIL-1Ra Levels Are Elevated in Sera of Patients with MS Treated with
`GA. sIL-1Ra circulating levels in MS have been shown to vary as
`a function of clinical status and treatment, so we examined
`whether GA-treatment would affect sIL-1Ra levels in patients
`with MS. IL-1␤ and sIL-1Ra levels were measured in sera of
`patients with RRMS treated with GA or untreated, and in
`healthy controls (Table 1). IL-1␤was not detectable in any of the
`sera. As shown in Fig. 2, sIL-1Ra was significantly increased in
`serum of patients treated with GA (434 ⫾ 265 pg/mL sIL-1Ra)
`whereas there was no significant difference between untreated
`patients (218 ⫾ 60 pg/mL sIL-1Ra) and healthy controls (188 ⫾
`65 pg/ml sIL-1Ra). This demonstrates that GA treatment en-
`hances sIL-1Ra in the serum of patients with MS.
`
`GA Differentially Regulates IL-1␤ and sIL-1Ra Production in Human
`Monocytes. To assess whether GA per se would affect the IL-1
`system in human monocytes, freshly isolated human monocytes
`
`4356 兩 www.pnas.org兾cgi兾doi兾10.1073兾pnas.0812183106
`
`Burger et al.
`
`2 of 5
`
`YEDA EXHIBIT NO. 2061
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`IMMUNOLOGY
`
`Fig. 4. GA affects sIL-1Ra and IL-1␤ mRNA in both CEsHUT-activated and
`resting monocytes. (A) Monocytes (2 ⫻ 106 cells/well/3 mL) were preincubated
`for 1 h with the indicated dose of GA and then cultured for 3 h in thepresence
`of CEsHUT (6 ␮g/mL). Total mRNA was isolated and analyzed by duplex real-time
`quantitative PCR (see Materials and Methods) for the presence of IL-1␤(open
`circles) and sIL-1Ra (filled circles) transcripts. Results are presented as percent-
`age of mRNA expression level, 100% being transcript expression measured
`after 3 h ofmonocyte activation by CE sHUT in the absence of GA; mean ⫾ SD
`of 3 different experiments. (B) Monocytes (2 ⫻ 106 cells/well/3 mL) were
`activated for 18 h with the indicated dose of GA. Total mRNA was isolated and
`analyzed by duplex real-time quantitative PCR (see Materials and Methods)
`for the presence of IL-1␤ (open circles) and sIL-1Ra (filled circles) transcripts.
`Results are presented as percentage of mRNA expression level, 100% being
`the transcript level at 50 ␮g/mL GA; mean ⫾ SD of 3 different experiments, i.e.,
`monocytes prepared from 3 different blood donors.
`
`level, monocytes were incubated for 1 h withincreasing doses of
`GA and then activated by CEsHUT or not activated. As shown in
`Fig. 4A, GA,
`in the absence of other stimuli,
`induced the
`expression of sIL-1Ra transcript in a dose-dependent manner,
`whereas that of IL-1␤ was not induced. When monocytes were
`activated by CEsHUT, GA diminished IL-1␤mRNA expression by
`30% whereas it enhanced sIL-1Ra mRNA expression by 25% in
`monocytes activated by CEsHUT (Fig. 4B), corroborating the
`effects of GA on protein production (Fig. 3). Together, these
`data suggest that GA displays opposite activity toward IL-1
`system members in monocytes/macrophages by directly inducing
`sIL-1Ra expression and production and by modulating both
`IL-1␤ and sIL-1Ra expression/production induced by CEsHUT.
`
`Discussion
`The present study sheds light on mechanisms by which GA might
`exert beneficial effects in MS. GA treatment enhances sIL-1Ra
`blood levels in patients with MS and in EAE mice. This is likely
`to be the consequence of the direct triggering effect of GA on
`monocytic production of sIL-1Ra. In addition, GA diminishes
`monocytic IL-1␤ production induced by direct contact with
`stimulated T cells. Thus, through different mechanisms, GA
`dampens IL-1␤ activity, which correlates with disease severity.
`Recent insights derived from studies on the mechanism of
`action of GA show a pivotal role of monocytes in the modulation
`of the immune system and highlight the importance of these cells
`as a target for pharmacologic intervention in autoimmune
`diseases (4–10, 24). These results suggest that GA might be
`useful in autoimmune diseases other than MS, as suggested by its
`beneficial effect in animal models of autoimmune diseases such
`as uveoretinitis (25) and inflammatory bowel disease (26), and
`graft rejection (27), whereas its efficacy has not been demon-
`strated in animal models of systemic lupus erythematosus (28)
`and collagen-induced arthritis (29).
`The premise that GA enhances sIL-1Ra levels in treated
`patients with MS is reminiscent of observations made with
`another immunomodulator used in MS. Indeed, IFN␤ also
`increases circulating serum levels of sIL-1Ra in patients with MS
`(30). Interestingly, with both immunomodulators, the circulating
`levels of sIL-1Ra are doubled in treated patients compared with
`untreated individuals. Together these observations suggest that
`the enhancement of sIL-1Ra might be relevant to therapeutic
`effects of both GA and IFN␤. Indeed, sIL-1Ra is transported and
`expressed into the CNS, where it could inhibit
`the pro-
`
`sIL-1Ra levels are elevated in sera of patients with MS treated with GA.
`Fig. 2.
`The levels of sIL-1Ra and IL-1␤ were measured in sera of patients with RRMS
`treated with GA or not treated, and age-matched healthy controls, as de-
`scribed in Table 1. IL-1␤ was under the detection limit (15 pg/mL) in all
`individuals. There was no significant difference between healthy controls and
`untreated patients with RRMS. Results are presented as a box plot (GraphPad
`Prism 4).
`
`fact that target cells are potentially phagocytic, isolated mem-
`branes from stimulated HUT-78 cells were solubilized with
`CHAPS and used as a stimulus, referred to as CEsHUT (23). As
`shown in Fig. 3B, GA enhanced the production of sIL-1Ra in
`monocytes activated by CEsHUT and LPS in a similar manner,
`and GA-induced sIL-1Ra production was additive to that trig-
`gered by CEsHUT or LPS. In contrast, the production of IL-1␤
`induced by CEsHUT was inhibited by GA, whereas LPS-induced
`production of IL-1␤ was enhanced in the presence of GA (Fig.
`3C). These observations suggest that GA displays opposite
`effects on signaling events downstream of LPS and CEsHUT.
`
`GA Affects the Expression of Cytokine Transcripts. To assess whether
`GA affected the production of cytokines at the transcriptional
`
`Fig. 3. GA differentially regulates IL-1␤ and sIL-1Ra production in human
`monocytes. (A) Monocytes (5 ⫻ 104 cells/200 ␮L/well; 96-well plates) were
`activated with the indicated dose of GA. After 48 h, supernatants were
`harvested and the production of IL-1␤(open circles) and sIL-1Ra (filled circles)
`were measured in triplicate wells and represented as mean ⫾ SD. Results are
`representative of 3 different experiments. (B) Monocytes (5 ⫻ 104 cells/200
`␮L/well; 96-well plates) were preincubated for 1 h with or without 25 ␮g/mL
`GA and then cultured for 48 h in the presence or absence of CEsHUT (1 ␮g/mL)
`or LPS (100 ng/mL). sIL-1Ra was measured in culture supernatants (mean ⫾ SD,
`n ⫽ 3 different experiments). (C) Monocytes (5 ⫻ 104 cells/200 ␮L/well; 96-well
`plates) were preincubated for 1 h with or without 25 ␮g/mL GA and then cultured
`for 48 h in the presence or absence of CEsHUT (6 ␮g/mL) or LPS (100 ng/mL). IL-1␤
`was measured in culture supernatants (mean ⫾ SD, n ⫽ 3 different experiments,
`i.e., monocytes prepared from 3 different blood donors).
`
`Burger et al.
`
`PNAS 兩 March 17, 2009 兩 vol. 106 兩 no. 11 兩 4357
`
`3 of 5
`
`YEDA EXHIBIT NO. 2061
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`inflammatory activities of IL-1␤, whose expression is increased
`in MS lesions (18, 31). The efficiency of sIL-1Ra treatment was
`demonstrated in EAE animals, in which it results in delayed and
`milder disease (19, 22). Besides, polymorphisms encoded within
`the IL-1 gene cluster were associated with MS (32). In particular,
`mild/moderate disease has been correlated to allele 2 of the
`IL-1Ra gene (IL1RN) variable number of tandem repeats ge-
`notype, which favors the production of sIL-1Ra (12, 33). In
`addition, families displaying high innate IL-1␤/sIL-1Ra ratio are
`at increased risk to have a relative who develops MS (34).
`Together, these studies reinforce the potential clinical benefit of
`GA to selectively induce sIL-1Ra secretion by monocytes in MS,
`as demonstrated in the present study. Of note, direct treatment
`with the commercially available form of sIL-1Ra (Anakinra)
`may represent an alternative treatment for MS, although its short
`lifespan once injected in humans may limit its efficacy (13).
`Nevertheless, as demonstrated here, the enhancement of intrin-
`sic production of sIL-1Ra might be a mediator of the beneficial
`effects of GA in MS.
`Most studies have focused on the effects of GA treatment on
`the adaptive immune system, in particular on T cells. Recent data
`favor the view that primary immune modulation of APC directs
`T cell immune modulation as a secondary step. Indeed, in mice
`lacking mature B and T cells, the GA treatment effect on
`monocytes is unbowed as indicated by an anti-inflammatory
`‘‘type II’’ cytokine shift (10). This indicates that GA does not
`require T cells or T cell products to alter monocytic cytokine
`production. Furthermore, adoptive transfer of highly purified,
`GA-induced type II monocytes into mice with EAE triggers T
`cell immune modulation and ameliorates the disease course of
`recipient mice. However, MHC II-deficient type II monocytes
`were unable to mediate this effect on T cells or disease severity
`(10). Taken together, these observations indicate that in vivo GA
`treatment exerts a direct effect on APC, which rules T cell
`immune modulation as the effector arm of GA clinical benefit
`in CNS autoimmune disease. The present study confirm the
`direct effect of GA on cells of the monocytic lineage by
`demonstrating that it down-regulates T cell contact-induced
`IL-1␤ production and directly triggers the production of sIL-
`1Ra. Thus, GA directly affects both the antigen presentation and
`cytokine production of monocytic cells.
`Direct cellular contact with stimulated T cells is a major
`pathway for the production of IL-1␤ and TNF in monocytes/
`macrophages under sterile conditions (17, 35, 36). Indeed,
`contact-mediated activation of monocytes/macrophages by stim-
`ulated T lymphocytes is as potent as optimal doses of LPS to
`inducing IL-1␤and TNF production in monocytes (37, 38). This
`model was recently used to assess the potency of kinase inhib-
`itors in acute and chronic inflammatory conditions (39). It is thus
`likely that this mechanism is highly relevant to the pathogenesis
`and persistence of chronic/sterile inflammation in diseases such
`as MS. The effect of GA on cytokine production induced by
`contact with stimulated T cells in human microglial cells was
`previously demonstrated (16). Stimulated T cells that were
`pretreated with GA induced lower levels of TNF, IL-1␤, and IL-6
`in human microglial cells and phorbol 12-myristate 13-acetate
`(PMA)/IFN␥-treated U937 monocytic cells. However, in the
`latter study, GA was absent during microglial cell activation,
`implying that GA rather inhibited the ability of T cells to activate
`cytokine production by cells of the monocytic lineage. The
`present study demonstrates that GA influences cytokine pro-
`duction by acting directly on human monocytes. Together, these
`studies suggest that GA affects the activation stage of both T cells
`and monocytes/macrophages to diminish contact-induced pro-
`inflammatory cytokine production.
`GA displays opposite effects on monocytes activated by LPS
`and T cell contact. Indeed, in contrast with CEsHUT-activated
`
`monocytes, the production of the pro-inflammatory cytokine
`IL-1␤ was up-regulated when cells were activated by LPS. This
`result is in agreement with a previous study showing that GA
`enhances the production of IL-1␤ in the human monocytic cell
`line THP-1 when activated by LPS (40). Thus, GA displays
`opposite effects on cytokine production by monocytes activated
`upon acute/infectious (i.e., LPS) and chronic/sterile (i.e., CE-
`sHUT) inflammatory conditions. Therefore, the use of LPS as an
`in vitro stimulus should be used with caution to mimic inflam-
`matory conditions when chronic/sterile inflammatory diseases
`are investigated (39).
`In conclusion, this study demonstrates that GA directly affects
`monocytes/macrophages by triggering the production of the
`anti-inflammatory cytokine sIL-1Ra. As sIL-1Ra can be both
`transported through the blood–brain barrier and induced within
`the CNS, it might exert immunomodulatory effects in both
`systemic and CNS compartments. In the latter, GA may also
`dampen the production and activity of IL-1␤. Finally, the present
`data strengthen recent demonstrations that, in addition to the
`modulation of the adaptive immune system, GA significantly
`affects the innate immune system.
`
`Materials and Methods
`Patients. Patients and healthy volunteers were recruited at the University
`Hospital of Geneva in accordance with institutional guidelines, and ap-
`proval of the local ethical committee was obtained. Blood was drawn from
`10 healthy controls, 11 untreated patients with RRMS, and 9 GA-treated
`patients with RRMS (Table 1). Sex, age, clinical score, and disease duration
`were matched between groups. All enrolled patients had definite RRMS
`according to revised McDonald criteria (41). At the time of blood sampling,
`GA-treated patients received 20 mg of GA s.c. daily for at least 1 year, with
`mean treatment duration of 33.6 ⫾ 16.8 months (Table 1). None of the
`patients were receiving an immunomodulatory or immunosuppressive
`drug in addition to GA. Patients from the untreated group did not receive
`any immunosuppressive or immunomodulatory drug for at least 6 months
`preceding the study.
`
`EAE Induction and GA Treatment. EAE was induced in 6 C57BL/6 mice using 10
`␮g myelin oligodendrocyte glycoprotein 35–55 peptide in complete Freund
`adjuvant. After immunization and 48 h later, mice received an i.v. injection of
`200 ng pertussis toxin. Mice were scored as follows: 0, no symptoms; 1,
`decreased tail tone; 2, mild monoparesis or paraparesis; 3, severe paraparesis;
`4, paraplegia and/or quadriparesis; and 5, moribund condition or death. Mice
`received daily s.c. injections of 150 ␮g GA suspended in PBS solution (n ⫽ 3) or
`PBS solution alone (n ⫽ 3) starting 7 days before EAE induction as described
`elsewhere (42). All experiments were carried out in accordance with guide-
`lines prescribed by the Institutional Animal Care and Use Committee at the
`University of California, San Francisco.
`
`Materials. FCS, streptomycin, penicillin, L-glutamine, RPMI-1640, and PBS
`solution free of Ca2⫹ and Mg2⫹ were purchased from Gibco; purified phyto-
`hemagglutinin from EY Laboratories; Lymphoprep from Axis-Shield; PMA,
`polymyxin B sulfate, and mouse anti-␤-tubulin antibody from Sigma; and GA
`from Sanofi-Aventis. Other reagents were of analytical grade or better.
`
`Monocytes. Peripheral blood monocytes were isolated from buffy coats of
`blood of healthy volunteers as previously described (43). To avoid activation
`by endotoxin, polymyxin B sulfate (2 ␮g/mL) was added in all solutions during
`isolation procedure.
`
`T Cell Stimulation and Membrane Isolation. HUT-78, a human T cell line, was
`obtained from the American Type Culture Collection. Cells were maintained
`in RPMI-1640 medium supplemented with 10% heat-inactivated FCS, 50
`␮g/mL streptomycin, 50 IU/mL penicillin, and 2 mM L-glutamine in 5% CO2-air
`humidified atmosphere at 37 °C. HUT-78 cells (2 ⫻ 106 cells/mL) were stimu-
`lated for 6 h byphytohemagglutinin (1 ␮g/mL) and PMA (5 ng/mL). Plasma
`membranes of stimulated HUT-78 cells were prepared as previously described
`and solubilized in 8 mM CHAPS (23, 44). CHAPS extract of membranes of
`stimulated HUT-78 cells was referred as to CEsHUT. Previous studies demon-
`strated that fixed, stimulated HUT-78 cells, plasma membranes of the latter
`
`4358 兩 www.pnas.org兾cgi兾doi兾10.1073兾pnas.0812183106
`
`Burger et al.
`
`4 of 5
`
`YEDA EXHIBIT NO. 2061
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`IMMUNOLOGY
`
`cells, and CEsHUT display similar ability to induce cytokine production in human
`monocytes (15). To activate monocytes, CEsHUT was used at either 1 ␮g/mL or
`6 ␮g/mL proteins as previously determined (38, 44).
`
`mRNA Quantification. Monocytes (2 ⫻ 106 cells/well/3 mL) were cultured in
`6-well plates with the indicated stimulus for 3 h or 18 h.Total RNA was isolated
`and analyzed by quantitative real-time PCR as previously described (23).
`
`Cytokine Production. Monocytes (5 ⫻ 104 cells/well/200 ␮L) were activated with
`the indicated stimulus in RPMI medium 1640 supplemented with 10% heat-
`inactivated FCS, 50 ␮g/mL streptomycin, 50 U/mL penicillin, 2 mM L-glutamine,
`and 5 ␮g/mL polymyxin B sulfate in 96-well plates and cultured for 48 h. The
`production of sIL-1Ra and IL-1␤ was measured in culture supernatants and
`patients’ serum by commercially available enzyme immunoassay: IL-1␤ (Im-
`munotech) and sIL-1Ra (Quantikine; R&D Systems). IL-1␤and sIL-1Ra concen-
`trations in serum of patients with RRMS and healthy controls were determined
`by triplicate measurements of the same sample.
`
`Statistics. When required, significance of differences between groups was
`evaluated using the Student t test.
`
`ACKNOWLEDGMENTS. This work was supported by Swiss National Science
`Foundation Grants 320000–116259 (to D.B.) and 310000–113653 (to P.H.L.); Swiss
`Society for Multiple Sclerosis grants (to D.B. and P.H.L.); a Hans Wilsdorf Foun-
`dation grant (to D.B.); Swiss National Science Foundation Advanced Researcher
`Fellowship PA00A-119532 (N.M.); National Multiple Sclerosis Society Fellowship
`(M.S.W.); National Institutes of Health Grants RO1 AI509709 and RO1 AI073737
`(to S.S.Z.); National Multiple Sclerosis Society Grants RG 4124 and RG 3913 (to
`S.S.Z.); Maisin Foundation (S.S.Z.); and the Dana Foundation (S.S.Z.).
`
`1. Sela M, Teitelbaum D (2001) Glatiramer acetate in the treatment of multiple sclerosis.
`Expert Opin Pharmacother 2:1149 –1165.
`2. Johnson KP, et al. (1995) Copolymer 1 reduces relapse rate and improves disability in
`relapsing-remitting multiple sclerosis: results of a phase III multicenter, double-blind
`placebo-controlled trial. The Copolymer 1 Multiple Sclerosis Study Group. Neurology
`45:1268 –1276.
`3. Neuhaus O, Farina C, Wekerle H, Hohlfeld R (2001) Mechanisms of action of glatiramer
`acetate in multiple sclerosis. Neurology 56:702–708.
`4. Weber MS, et al. (2004) Multiple sclerosis: glatiramer acetate inhibits monocyte
`reactivity in vitro and in vivo. Brain 127:1370 –1378.
`5. Kim HJ, et al. (2004) Type 2 monocyte and microglia differentiation mediated by
`glatiramer acetate therapy in patients with multiple sclerosis. J Immunol 172:7144 –
`7153.
`6. Vieira PL, Heystek HC, Wormmeester J, Wierenga EA, Kapsenberg ML (2003) Glati-
`ramer acetate (copolymer-1, copaxone) promotes Th2 cell development and increased
`IL-10 production through modulation of dendritic cells. J Immunol 170:4483– 4488.
`7. Jung S, et al. (2004) Induction of IL-10 in rat peritoneal macrophages and dendritic cells
`by glatiramer acetate. J Neuroimmunol 148:63–73.
`8. Stasiolek M, et al. (2006) Impaired maturation and altered regulatory function of
`plasmacytoid dendritic cells in multiple sclerosis. Brain 129:1293–1305.
`9. Hussien Y, Sanna A, Soderstrom M, Link H, Huang YM (2001) Glatiramer acetate and
`IFN-beta act on dendritic cells in multiple sclerosis. J Neuroimmunol 121:102–110.
`10. Weber MS, et al. (2007) Type II monocytes modulate T cell-mediated central nervous
`system autoimmune disease. Nat Med 13:935–943.
`11. Dinarello CA (2007) Mutations in cryopyrin: bypassing roadblocks in the caspase 1
`inflammasome for interleukin-1beta secretion and disease activity. Arthritis Rheum
`56:2817–2822.
`12. Burger, D, Dayer, JM (2000) IL-1Ra. Cytokine Reference, eds Oppenheim JJ, Feldmann
`M (Academic, London), pp 319 –336.
`13. Burger D, Dayer JM, Palmer G, Gabay C (2006) Is IL-1 a good therapeutic target in the
`treatment of arthritis? Best Pract Res Clin Rheumatol 20:879 – 896.
`14. Burger D, Dayer JM (2002) The role of human T lymphocyte-monocyte contact in
`inflammation and tissue destruction. Arthritis Res 4(suppl 3):S169 –S176.
`15. Burger, D, Dayer, JM, Molnarfi, N (2007) Cell contact dependence of inflammatory
`events. Contemporary Targeted Therapies in Rheumatology, eds Smolen JS, Lipsky PE
`(Taylor & Francis Books, Abingdon, UK), pp 85–103.
`16. Chabot S, et al. (2002) Cytokine production in T lymphocyte-microglia interaction is
`attenuated by glatiramer acetate: a mechanism for therapeutic efficacy in multiple
`sclerosis. Mult Scler 8:299 –306.
`17. Dasgupta S, Jana M, Liu X, Pahan K (2003) Role of very-late antigen-4 (VLA-4) in myelin
`basic protein-primed T cell contact-induced expression of proinflammatory cytokines
`in microglial cells. J Biol Chem 278:22424 –22431.
`18. Cannella B, Raine CS (1995) The adhesion molecule and cytokine profile of multiple
`sclerosis lesions. Ann Neurol 37:424 – 435.
`19. Badovinac V, Mostarica-Stojkovic M, Dinarello CA, Stosic-Grujicic S (1998) Interleukin-1
`receptor antagonist suppresses experimental autoimmune encephalomyelitis (EAE) in
`rats by influencing the activation and proliferation of encephalitogenic cells. J Neu-
`roimmunol 85:87–95.
`20. Furlan R, et al. (2007) HSV-1-mediated IL-1 receptor antagonist gene therapy amelio-
`rates MOG(35–55)-induced experimental autoimmune encephalomyelitis in C57BL/6
`mice. Gene Ther 14:93–98.
`21. Matsuki T, Nakae S, Sudo K, Horai R, Iwakura Y (2006) Abnormal T cell activ

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