throbber
DOI: 10.1093/brain/awh163
`
`Brain (2004), 127, 1370–1378
`
`Multiple sclerosis: glatiramer acetate inhibits
`monocyte reactivity in vitro and in vivo
`
`Martin S. Weber,1 Michaela Starck,2 Stefan Wagenpfeil,3 Edgar Meinl,1,4 Reinhard Hohlfeld1,4 and
`Cinthia Farina4
`
`Correspondence to: Dr R. Hohlfeld, Institute for Clinical
`Neuroimmunology, Marchioninistrasse 15, D-81377
`Munich, Germany
`E-mail: hohlfeld@neuro.mpg.de
`
`Downloaded from
`
`by guest on October 30, 2014
`
`(detected by cytofluorometry), and by production of
`(TNF)-a
`monocyte-derived tumour necrosis
`factor
`(detected by enzyme-linked immunospot assay). GA had
`a broad inhibitory effect on all measures of monocyte
`reactivity, regardless of which stimulator was used. It is
`unlikely that this reflects a simple toxic effect, because
`monocyte viability and CD14 expression were unaf-
`fected. In a second series of experiments, we investi-
`gated the properties of monocytes cultured ex vivo from
`eight GA-treated multiple
`sclerosis patients,
`eight
`untreated multiple sclerosis patients and eight healthy
`subjects. We found that LPS-induced SLAM expression
`and TNF-a production were significantly reduced in
`monocytes from GA-treated patients compared with
`controls. These results demonstrate for the first time
`that GA inhibits monocyte reactivity in vitro and in vivo,
`significantly extending the current concept of the mech-
`anism of action of GA.
`
`1Institute for Clinical Neuroimmunology, Klinikum
`Großhadern, Ludwig Maximilians University, Munich,
`2Marianne-Strauss-Klinik, Berg, 3Institute of Medical
`Statistics and Epidemiology, Technical University, Munich
`and 4Department of Neuroimmunology, Max Planck
`Institute of Neurobiology, Martinsried, Germany
`
`Summary
`It is widely assumed that glatiramer acetate (GA), an
`approved agent for the immunomodulatory treatment
`of multiple sclerosis, acts primarily as an antigen for T
`lymphocytes. Recent studies, however,
`indicated that
`in vitro, GA directly inhibits dendritic cells, a rare but
`potent
`type of professional antigen-presenting cell
`(APC). To investigate whether these in vitro observa-
`tions are relevant to the actions of GA in vivo, we
`studied the effects of GA on monocytes, the major type
`of circulating APC. In a first series of experiments, we
`investigated the effects of GA on monocyte reactivity
`in vitro. Monocytes were stimulated with ligands for
`Toll-like receptor (TLR)-2 (peptidoglycan and lipo-
`teichoic acid), TLR-4 [lipopolysaccharide (LPS)] and
`TLR-5 (flagellin), as well as two proinflammatory cyto-
`kines (interferon-g and granulocyte–monocyte colony-
`stimulating factor). Monocyte activation was measured
`by induction of the surface markers signalling lympho-
`cytic activation molecule (SLAM), CD25 and CD69
`
`Keywords: multiple sclerosis; glatiramer acetate; immunotherapy; monocyte; ex vivo assay
`
`Abbreviations: APC = antigen-presenting cell; DC = dendritic cell; EDSS = Expanded Disability Status Scale;
`Elispot = enzyme-linked immunospot; FACS = fluorescence activated cell sorting; GM-CSF = granulocyte–monocyte
`colony-stimulating factor; GA = glatiramer acetate; IFN = interferon; IL = interleukin; LPS = lipopolysaccharide;
`LTA = lipoteichoic acid; PBMC = peripheral blood mononuclear cell; PGN = peptidoglycan; SLAM = signalling
`lymphocytic activation molecule; TLR = Toll-like receptor; TNF = tumour necrosis factor
`
`Received December 3, 2003. Revised February 4, 2004. Accepted February 5, 2004. Advance Access publication April 16, 2004
`
`Introduction
`sclerosis lesions (Johnson et al., 1995, 2000; Mancardi et al.,
`Glatiramer acetate (GA, Copaxone), a random copolymer
`1998; Ge et al., 2000; Comi et al., 2001; Filippi et al., 2001;
`composed of alanine, glutamic acid, lysine and tyrosine, is an
`Wolinsky et al., 2001; Ziemssen et al., 2001). Over recent
`approved agent for the immunomodulatory treatment of
`years, a number of studies have addressed the immunological
`multiple sclerosis. Clinical and magnetic imaging studies
`basis of the clinical effects of GA in animal models and
`indicated that GA treatment reduces the activity of multiple
`Brain Vol. 127 No. 6 ª Guarantors of Brain 2004; all rights reserved
`
`Page 1 of 9
`
`YEDA EXHIBIT NO. 2076
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Downloaded from
`
`by guest on October 30, 2014
`
`human multiple sclerosis (reviewed in Aharoni et al., 2000;
`Neuhaus et al., 2001; Yong, 2002). These studies consistently
`showed that GA induces a ‘TH1 to TH2 cytokine shift’ in
`GA-reactive CD4+ T cells. In the immune system of untreated
`multiple sclerosis patients and healthy subjects, the majority
`of GA-reactive CD4+ T cells belong to the TH1 subset. These
`cells characteristically produce pro-inflammatory cytokines
`such as interleukin (IL)-2 and interferon-g (IFN-g) (Murphy
`and Reiner, 2003). During treatment with GA, the cytokine
`profile of the GA-reactive T cells shifts towards the TH2 type,
`which is characterized by production of anti-inflammatory
`‘TH2’ cytokines, including IL-4 (Neuhaus et al., 2000; Farina
`et al., 2001; Dhib-Jalbut et al., 2002), IL-5 (Duda et al., 2000)
`and IL-13 (Wiesemann et al., 2003).
`These observations laid the basis for the current concept of
`the mechanism of action of GA. It is thought that activated,
`GA-reactive TH2 T cells migrate across the blood barrier. In
`the CNS,
`they are re-activated locally by cross-reacting
`myelin auto-antigens (Neuhaus et al., 2001). After local re-
`stimulation, the GA-reactive TH2 cells release their anti-
`inflammatory cytokines, and also certain neurotrophic factors
`(Ziemssen et al., 2002). In this way, the recruited TH2 T cells
`can suppress neighbouring autoaggressive TH1 cells. This
`process is called ‘bystander suppression’ (Aharoni et al.,
`1998). In addition, the GA-reactive T cells can deliver brain-
`derived neurotrophic factor
`(BDNF)
`to neurons, which
`upregulate the corresponding full-length signalling tyrosine
`kinase receptor gp145 trkB in multiple sclerosis lesions
`(Stadelmann et al., 2002; Ziemssen et al., 2002).
`The mechanism(s) of the therapeutically induced TH1 to
`TH2 cytokine shift of GA-reactive T cells is unknown.
`Theoretically,
`there are two (not necessarily exclusive)
`possibilities. First, GA might have a primary effect on T
`cells, for example by virtue of its properties as an ‘altered
`peptide ligand’, or by the special conditions of GA presen-
`tation in the skin. Secondly, GA might exert a primary effect
`on antigen-presenting cells (APCs), e.g. by altering their
`properties in such a way that they preferentially induce TH2
`cells. Indeed, there is recent evidence that GA might affect
`the properties of APCs in vitro (Hussien et al., 2001; Vieira
`et al., 2003). In this regard, the findings reported by Vieira
`et al. (2003) are especially intriguing. Using in vitro cultures,
`these authors found that GA affects the T-cell-stimulating
`properties of dendritic cells (DCs). After in vitro treatment
`with GA, DCs have an impaired capacity to secrete TH1-
`polarizing factors, and therefore preferentially induce TH2
`cells (Vieira et al., 2003). Whether these observations extend
`to other types of APC, and whether they are relevant in vivo
`presently is unknown.
`The main aim of our present study was to search for
`evidence that GA affects the function of APCs in vivo. We
`focused our analysis on monocytes, because DCs constitute a
`very rare subset of APCs which can only be studied after
`prolonged expansion in culture, and which are therefore
`inaccessible for direct ex vivo analysis. In contrast, mono-
`cytes represent a major subset of professional APCs which
`
`Glatiramer acetate therapy affects monocytes
`
`1371
`
`can be easily obtained from peripheral blood and are readily
`accessible for ex vivo investigations.
`In a first step of analysis, we performed a detailed series of
`experiments to identify the possible effects of GA on different
`patterns of monocyte activation in vitro. For monocyte
`activation, we used (i) four ligands for three distinct Toll-like
`receptors (TLRs); and (ii) the proinflammatory cytokines
`IFN-g and granulocyte–monocyte colony-stimulating factor
`(GM-CSF). As markers of monocyte activation, we looked at
`(i) the expression of activation-related surface molecules
`including
`signalling
`lymphocytic
`activation molecule
`(SLAM) (CD150), CD25 and CD69; and (ii) production of
`tumour necrosis factor (TNF)-a. These experiments revealed
`that GA broadly affects monocyte activation by different
`ligands and pathways.
`In the second step, we addressed the possible effects of GA
`on monocytes in vivo. To this end, we compared the
`properties of ex vivo monocytes from untreated and GA-
`treated subjects. ‘Ex vivo’ means that the monocytes were
`exposed to GA only in vivo, but not in vitro. Based on the
`experiments from the first step, we chose lipopolysaccharide
`(LPS)-induced SLAM expression and TNF-a secretion as
`‘read-out’ to test the monocyte stimulation thresholds in vitro.
`Our main observation is that monocytes cultured ex vivo from
`GA-treated patients were indeed significantly less susceptible
`to activation than monocytes from untreated patients and
`normal controls. These results demonstrate, we believe for
`the first time, that GA treatment in vivo leads to a systemic
`alteration of the properties of circulating monocytes, raising
`important new questions regarding the mechanism of action
`of GA.
`
`Materials and methods
`Subjects and cell samples
`Blood was drawn from healthy individuals, GA-treated multiple
`sclerosis patients and untreated multiple sclerosis patients after their
`informed consent. This study has been approved by the local ethics
`commitee of the Ludwig Maximilians University of Munich. All
`patients had definite multiple sclerosis (McDonald et al., 2001). All
`GA-treated patients (n = 8) had a relapsing–remitting disease course.
`At
`the time of sampling,
`they had injected 20 mg of GA
`subcutaneously (s.c.) daily for at least 1 year, with a mean treatment
`duration of 34.4 6 16.9 months [mean Expanded Disability Status
`Scale (EDSS) at time of sampling 1.75 6 1.2; mean age 31.8 6 8.4
`years] (Table 1). The untreated group included four patients with
`relapsing–remitting multiple sclerosis, two with primary progressive
`multiple sclerosis and two with secondary progressive multiple
`sclerosis (five women and three men; mean EDSS 3.5 6 1.9; mean
`age 47.1 6 10.5 years). None of these patients was treated with
`immunosuppressive or immunomodulatory therapy during at least 3
`months preceding the study. The group of healthy donors included
`four men and four women with a mean age of 35.1 6 11.9 years.
`Peripheral blood mononuclear cells (PBMCs) were isolated on a
`discontinuous density gradient
`(Lymphoprep, Nycomed, Oslo,
`Norway). Viable cells were counted by trypan blue (Sigma-
`Aldrich) exclusion and resuspended in culture medium [RPMI
`
`Page 2 of 9
`
`YEDA EXHIBIT NO. 2076
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Downloaded from
`
`by guest on October 30, 2014
`
`1372
`
`M. S. Weber et al.
`
`Table 1 Characteristics of GA-treated MS patients and untreated MS patients.
`
`Subjects
`
`Gender
`
`Age
`(years)
`
`Year of
`diagnosis
`
`Disease
`course
`
`EDSS*
`
`GA treatment
`(months)*
`
`GA-treated multiple sclerosis patients
`MS-GA-1
`MS-GA-2
`MS-GA-3
`MS-GA-4
`MS-GA-5
`MS-GA-6
`MS-GA-7
`MS-GA-8
`Untreated multiple sclerosis patients
`MS-NT-1
`MS-NT-2
`MS-NT-3
`MS-NT-4
`MS-NT-5
`MS-NT-6
`MS-NT-7
`MS-NT-8
`
`F
`F
`F
`M
`F
`F
`F
`F
`
`F
`F
`F
`M
`F
`M
`M
`F
`
`34
`21
`13
`62
`28
`32
`28
`57
`
`43
`30
`33
`25
`21
`33
`45
`25
`
`31
`36
`47
`39
`54
`56
`57
`57
`
`1981
`1991
`1995
`1988
`1999
`1999
`1993
`1993
`
`1996
`2000
`1997
`1982
`1975
`1976
`1996
`2000
`
`RR
`RR
`RR
`RR
`RR
`RR
`RR
`RR
`
`RR
`RR
`RR
`RR
`SP
`SP
`PP
`PP
`
`3.5
`1.5
`1.0
`1.5
`3.5
`1.0
`2
`0
`
`2.0
`2.5
`2.0
`1.0
`4.5
`6.5
`5.5
`4.0
`
`*At the time of sampling; MS-GA = GA-treated multiple sclerosis patient; MS-NT = untreated multiple sclerosis patient; RR = relapsing–
`remitting multiple sclerosis; SP = secondary progressive multiple sclerosis; PP = primary progressive multiple sclerosis.
`
`1640 supplemented with 5% fetal calf serum (FCS), 1% glutamine
`and 1% penicillin/streptomycin; Gibco]. One batch of FCS was used
`throughout the study.
`
`Reagents and antibodies
`The following reagents were used: human IFN-g (Roche, Mannheim,
`Germany), GM-CSF (R&D, Wiesbaden-Nordenstadt, Germany),
`flagellin from Helicobacter pylori (IBT, Reutlingen, Germany),
`peptidoglycan (PGN) from Staphylococcus aureus (Fluka, Sigma-
`Aldrich, Schnelldorf, Germany),
`lipoteichoic acid (LTA) from
`Staphylococcus aureus, and LPS from Escherichia coli 0111:B4
`(Sigma-Aldrich). LPS-free reagents, water (BioWhittaker, Verviers,
`Belgium), phosphate-buffered saline (PBS; Gibco, Karlsruhe,
`Germany) and bovine serum albumin (BSA; Sigma) were used to
`prepare the aliquots. GA (Batch-No. 242992899) was from Teva
`Pharmaceutical Industries Ltd, Petah Tiqva, Israel.
`The following antibodies were used for fluorescence activated
`cell sorting (FACS) analyses: phycoerythrin (PE)-labelled anti-
`SLAM, peridinin chlorophyll protein (PerCP)-labelled anti-human
`CD14, and fluorescein isothiocyanate (FITC)-labelled anti-human
`CD25 and CD69 (all
`from Becton Dickinson, Heidelberg,
`Germany). All
`the corresponding isotype controls were from
`Becton Dickinson.
`
`Enzyme-linked immunospot (Elispot) assay
`96-well polyvinylidene difluoride plates (Millipore, Eschborn,
`Germany) were coated at 4(cid:176)C overnight with the capture antibody
`(anti-TNF-a antibody; Mabtech, Nacka, Sweden). After the wells
`were washed and blocked with culture medium for 1 h at 37(cid:176)C, the
`cells (1 3 103/well for the in vitro and 3 3 103/well for the ex vivo
`TNF-a assay) were seeded and stimulated for 18 h at 37(cid:176)C and 5%
`CO2. The experiments were performed in triplicate. After culture,
`the plates were washed and incubated first with the biotinylated
`detector antibody (Mabtech),
`then with streptavidin–alkaline
`phosphatase (Mabtech), and finally with BCIP/NBT (Sigma-
`Aldrich). The Elispot plates were analysed with an automated
`imaging system and appropriate computer software (KS ELISPOT
`automated image analysis system, Zeiss, Jena, Germany).
`
`Statistical analysis
`The GA dose dependence of the percentage of SLAM-positive
`monocytes and the frequency of TNF-a-producing cells were
`analysed with linear regression. The t test for independent samples
`was used to compare GA-treated multiple sclerosis patients,
`untreated multiple sclerosis patients and healthy controls. All P
`values given are two-sided and subject to a significance level of 5%.
`
`FACS staining and analysis
`The cells were labelled with the predetermined appropriate antibody
`dilution or with the corresponding isotype controls. FACS stainings
`were analysed on a FACScan using Cell-Quest software (Becton
`Dickinson). Monocytes were gated in forward/side scatter. The
`quadrants were set on the relative isotype controls. The in vitro
`experiments were repeated at least three times.
`
`Results
`In vitro culture with GA inhibits monocyte
`activation via different TLR ligands and
`inflammatory cytokines
`In previous experiments, we have characterized monocyte
`responses to (i) different bacterial TLR ligands (TLR-2
`ligands PGN and LTA; TLR-4 ligand LPS; and TLR-5 ligand
`
`Page 3 of 9
`
`YEDA EXHIBIT NO. 2076
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Glatiramer acetate therapy affects monocytes
`
`1373
`
`Fig. 1 GA-mediated inhibition of LPS-stimulated monocyte responses in vitro (dose–response analysis). In (A), monocyte reactivity was
`measured as LPS-induced TNF-a production by Elispot assay. In (B), monocyte reactivity was measured as LPS-induced SLAM
`expression by FACS.
`
`Downloaded from
`
`by guest on October 30, 2014
`
`Fig. 2 GA-mediated inhibition of monocyte responses stimulated
`with different TLR ligands (LTA, PGN and flagellin) or
`inflammatory cytokines (GM-CSF and IFN-g). Monocyte reactivity
`was measured as TNF-a production by Elispot assay.
`
`50 mg/ml GA, SLAM induction was inhibited by 66.8 6 37.4
`(PGN stimulation), 80.4 6 30.3 (flagellin) and 87 6 18.3%
`(LTA) (average values 6 SD; data not shown). Similarly, GA
`inhibited TNF-a production by monocytes stimulated with
`half-maximal doses of the different TLR ligands (Fig. 2;
`P = 0.002 for PGN, P < 0.0001 for LTA and flagellin).
`GA inhibited not only TLR-stimulated, but also IFN-g- and
`GM-CSF-stimulated TNF-a release (Fig. 2, P = 0.001 for
`IFN-g and P < 0.0001 for GM-CSF), although in this case
`high concentrations of inflammatory cytokines were neces-
`sary for monocyte stimulation. In three different experiments,
`the inhibitory effect of GA on TNF-a production was
`consistently greater when TLR ligands were used as
`activators (87–97% inhibition), compared with stimulation
`with IFN-g or GM-CSF (47–58% inhibition) (Fig. 2).
`We also investigated the effects of GA on two additional
`markers of monocyte activation, CD25 and CD69. These
`molecules are newly induced on monocytes by stimulation
`with TLR ligands or inflammatory cytokines. Table 2 shows
`the results of one (out of
`three)
`representative FACS
`experiment. Pre-incubation with 50 mg/ml GA reduced the
`
`flagellin), and (ii) inflammatory cytokines (IFN-g and GM-
`CSF). Monocyte activation was measured by (i) induction of
`surface activation markers (CD25, CD69 and SLAM); and
`(ii) cytokine [TNF-a production (Farina et al., 2004)]. We
`found that SLAM was induced preferentially by stimulation
`with TLR ligands but not inflammatory cytokines, whereas
`CD25 and CD69 were induced by both TLR ligands and
`inflammatory cytokines. Further, TNF-a production was
`induced more strongly by TLR ligands than by IFN-g and
`GM-CSF (Farina et al., 2004). These results laid the basis for
`our present study of
`the effects of GA on monocyte
`activation.
`In a typical dose–response curve, a plateau of activation
`was reached with LPS concentrations >1000 pg/ml. At
`plateau, GA had no detectable inhibitory effect on monocyte
`reactivity. For optimal detection of the inhibitory effect of
`GA, we found that low to intermediate concentrations (150–
`1000 pg/ml) of LPS had to be used. PBMCs from healthy
`donors were pre-incubated for 1.5 h with four different
`concentrations of GA, and stimulated overnight with different
`concentrations of LPS. Figure 1 shows one of
`three
`representative experiments for TNF-a production (Fig. 1A)
`and SLAM induction (Fig. 1B). At each LPS concentration,
`the percentage of SLAM-positive monocytes and the fre-
`quency of TNF-a-producing cells was strongly reduced
`(P < 0.05 for TNF-a production by 300, 600 and 1000 pg/ml
`LPS; P < 0.05 for SLAM induction by 600 and 1000 pg/ml
`LPS). As shown in Fig. 1, the inhibitory effect of GA was
`dose dependent. We conclude that GA inhibits monocyte
`responses induced with the TLR-4 ligand LPS.
`We also investigated the effects of GA on monocyte
`activation stimulated with the TLR-2 ligands PGN and LTA,
`and the TLR-5 ligand flagellin. As with LPS, GA had no
`detectable inhibitory effect at very high concentrations of
`these ligands. We measured dose–response curves for each
`ligand and determined the half-maximal stimulating concen-
`trations. Again, the monocyte response was measured in
`terms of SLAM induction and TNF-a production by FACS
`and Elispot assay, respectively. After pre-incubation with
`
`Page 4 of 9
`
`YEDA EXHIBIT NO. 2076
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Downloaded from
`
`by guest on October 30, 2014
`
`we assessed LPS-stimulated SLAM induction and TNF-a
`production by monocytes from GA-treated multiple sclerosis
`patients and controls. We compared PBMCs from eight GA-
`treated multiple sclerosis patients, eight untreated multiple
`
`1374
`
`M. S. Weber et al.
`
`fraction of activated monocytes expressing CD25 or CD69,
`regardless of which activator was used for stimulation
`(Table 2). Although the magnitudes of monocyte activation
`and of the inhibitory effect of GA varied between the
`different stimuli, GA consistently reduced the induction of
`CD25 and CD69 by all the different stimuli (CD69: 70%
`average inhibition for all activators; CD25: 70% average
`inhibition for LPS, LTA and PGN, 50% average inhibition for
`flagellin and IFN-g, 30% inhibition for GM-CSF). In contrast,
`CD14, which is constitutively expressed on monocytes, was
`unaffected by GA (Fig. 3).
`
`Treatment with GA in vivo leads to a reduction
`of monocyte reactivity ex vivo
`The experiments described in the previous section indicated
`that GA inhibits monocyte activation by TLR ligands and
`inflammatory cytokines in vitro. To investigate whether
`in vivo treatment with GA also affects monocyte properties,
`
`Table 2 GA blocks induction of activation markers on
`monocytes
`
`CD25*
`
`CD69*
`
`–GA
`
`+GA**
`
`–GA
`
`+GA**
`
`No stimulus
`LPS 0.15 ng/ml
`LTA 0.25 mg/ml
`PGN 0.25 mg/ml
`Flagellin 0.6 mg/ml
`IFN-g 1000 U/ml
`GM-CSF 100 ng/ml
`
`0.4%
`24.1%
`17.4%
`24.6%
`5.3%
`5.9 %
`19.8 %
`
`1.2%
`0.8%
`7.8%
`6.7%
`3.3%
`3.5 %
`15.3 %
`
`4.1%
`16.2%
`13.3%
`15.4%
`11.1%
`13.6 %
`8.5 %
`
`2.4%
`0%
`3.1%
`2.4%
`1.4%
`5.9 %
`0.5 %
`
`*The expression of the indicated activation markers on monocytes
`was determined by FACS and expressed as percentage of positive
`cells; **pre-incubation with 50 mg/ml GA.
`
`Fig. 3 GA-mediated effects on CD25 (A) and CD14 (B)
`expression by LPS-stimulated monocytes. LPS (0.15 ng/ml) and
`GA (50 mg/ml) were used. Upper panels (A) represent dot-blots of
`CD25 expression (ordinate) versus forward scatter (abscissa). GA
`reduced the proportion of CD25+ monocytes from 34.7% (without
`GA, upper left) to 3.1% (with GA, upper right). Lower panels (B)
`represent dot-blots of CD14 expression (ordinate) versus forward
`scatter (abscissa). There was no detectable change of CD14
`expression in the presence of GA (lower right). The two markers
`were analysed in parallel in one experiment using PBMCs from
`one healthy donor.
`
`Fig. 4 LPS-stimulated SLAM induction on monocytes cultured ex vivo from eight healthy donors, eight untreated multiple sclerosis
`patients and eight GA-treated multiple sclerosis patients. SLAM induction was measured by FACS. The monocytes had been exposed to
`GA only during in vivo treatment, not in vitro. Monocytes from GA-treated patients showed significantly reduced responses. *P = 0.002
`healthy controls versus GA-treated patients; P = 0.017 untreated patients versus GA-treated patients; P = 0.185 healthy controls versus
`untreated patients. The right panel shows means from each of the left panels. The P values at lower LPS concentrations were not
`significant.
`
`Page 5 of 9
`
`YEDA EXHIBIT NO. 2076
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Glatiramer acetate therapy affects monocytes
`
`1375
`
`Fig. 5 LPS-stimulated TNF-a production by monocytes cultured ex vivo from eight healthy donors, eight untreated multiple sclerosis
`patients and eight GA-treated multiple sclerosis patients. TNF-a production was measured by Elispot. The monocytes had been exposed to
`GA only during in vivo treatment, not in vitro. Monocytes from GA-treated patients showed significantly reduced responses. *P = 0.015
`healthy controls versus GA-treated patients; P = 0.013 untreated patients versus GA-treated patients; P = 0.69 healthy controls versus
`untreated patients; **P = 0.002 healthy controls versus GA-treated patients; P = 0.033 untreated patients versus GA-treated patients;
`P = 0.41 healthy controls versus untreated patients; ***P < 0.0001 healthy controls versus GA-treated patients; P = 0.001 untreated
`patients versus GA-treated patients; P = 0.29 healthy controls versus untreated patients. The right panel shows means from each of the left
`panels.
`
`Downloaded from
`
`by guest on October 30, 2014
`
`sclerosis patients and eight healthy donors. Note that in these
`experiments, the monocytes were not exposed to GA in vitro.
`Figure 4 compares LPS-induced SLAM expression on
`monocytes from each group of patients and control subjects.
`Despite some inter-individual variability, cells from GA-
`treated patients were clearly less susceptible to SLAM
`induction than cells from the control groups. For example,
`at 1000 pg/ml LPS, the mean proportion of SLAM-expressing
`monocytes was 22.1 6 4.4% (mean 6 SD) for GA-treated
`patients compared with 37.2 6 9.5% for healthy subjects
`(P = 0.002), and with 31.1 6 8.0% for the untreated multiple
`sclerosis group (P = 0.017). The two control groups were not
`significantly different (Fig. 4).
`We also compared LPS-induced TNF-a production by
`monocytes from the different groups. Figure 5 shows the
`dose–response curves for each subject as detected by Elispot
`assay. Spontaneous release was similar in the three groups
`(P = 0.41 for healthy controls versus GA-treated patients;
`P = 0.81 for healthy controls versus untreated patients;
`P = 0.23 for untreated patients versus GA-treated patients).
`However, PBMCs from GA-treated individuals were less
`susceptible to in vitro activation even at a low concentration
`of LPS (P = 0.015 and P = 0.013 at 150 pg/ml LPS when
`compared with healthy subjects and untreated multiple
`sclerosis patients, respectively). At increasing LPS concen-
`trations, the differences became more evident (P = 0.002 and
`P = 0.0001 when compared with healthy donors, P = 0.033
`and P = 0.001 when compared with untreated multiple
`sclerosis patients at 600 and 1000 pg/ml LPS, respectively).
`There were no significant differences between the two control
`groups at any of the different LPS concentrations (P values
`ranging from 0.29 to 0.69).
`the curves
`As shown in Figs 4 and 5 (right panels),
`representing the means for GA-treated patients were different
`from the control curves.
`
`Discussion
`The major new observation reported herein is that in treated
`multiple sclerosis patients, GA induces systemic functional
`changes of circulating monocytes. Monocytes from GA-
`treated multiple sclerosis patients are significantly less
`susceptible to LPS stimulation than monocytes
`from
`untreated multiple sclerosis patients and normal control
`subjects. This reduced monocyte reactivity was observed in
`terms of both LPS-induced expression of
`the surface
`activation molecule SLAM, and also LPS-induced secretion
`of the cytokine TNF-a.
`These changes were observed with ‘ex vivo’ monocytes,
`which had been exposed to GA only in vivo, but not in vitro.
`The results are fully consistent with our parallel observations
`on monocytes cultured in the presence of GA in vitro. After
`pre-incubation with GA in vitro, monocytes were less
`susceptible to a broad range of diverse stimuli, including
`ligands of different TLRs, and also different stimulatory
`cytokines (IFN-g and GM-CSF).
`structural
`recognition of
`TLRs are responsible for
`components conserved among classes of microorganisms
`(for a review see Underhill, 2003). Ligand binding induces
`the expression of various host defence molecules, including
`inflammatory cytokines, chemokines, co-stimulatory mol-
`ecules and major histocompatibility complex (MHC) mol-
`ecules. We recently have characterized the complex spectrum
`of human monocyte responses triggered by stimulation with
`TLRs, and compared it with the classical activation induced
`by inflammatory cytokines (Farina et al., 2004). We found
`that TLR ligands are stronger inducers of TNF-a production
`than are the cytokines IFN-g and GM-CSF, and that TLR
`ligands selectively trigger SLAM expression on the monocyte
`surface (Farina et al., 2004). Other activation markers such as
`CD25 and CD69 are induced both by TLR ligands and by
`inflammatory cytokines. In the present study, pre-incubation
`
`Page 6 of 9
`
`YEDA EXHIBIT NO. 2076
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Downloaded from
`
`by guest on October 30, 2014
`
`1376
`
`M. S. Weber et al.
`
`with GA blocked the induction of all investigated surface
`markers (SLAM, CD25 and CD69), and also of TNF-a
`release. Several experiments with enriched monocyte popu-
`lations showed that the inhibitory in vitro effect of GA on
`SLAM induction occurs in the absence of T cells.
`Taken together, these results suggest that GA broadly
`affects monocyte reactivity. The exact mechanism(s) of the
`inhibitory effect is presently unknown, and deserves further
`study. For several reasons, it appears unlikely that GA exerts
`a (trivial) toxic effect on monocytes. First, monocyte viability
`was not altered at the GA concentrations used in our in vitro
`experiments, as seen by propidium iodide uptake. Secondly,
`although GA reduced the induction of the activation-related
`surface molecules SLAM, CD25 and CD69, another surface
`marker of monocytes, CD14 [which is part of the LPS
`receptor complex (Underhill, 2003)] was completely un-
`affected by GA treatment in vitro. Furthermore, T cells from
`GA-treated patients showed the typical, previously described
`Elispot cytokine responses when stimulated with a wide range
`of GA concentrations in vitro (Farina et al., 2001, 2002).
`The new findings have obvious implications for our
`understanding of the mechanisms of action of GA. In several
`previous studies, part of these mechanisms was elucidated in
`some detail. It is widely assumed that the beneficial effects of
`GA are mediated mainly by GA-reactive TH2 cells. Previous
`studies showed that GA treatment induces a gradual shift of
`GA-reactive T cells from TH1 to TH2 (Miller et al., 1998;
`Neuhaus et al., 2000; Duda et al., 2000; Farina et al., 2001).
`Since the GA-reactive T cells are constantly activated by
`immunization, they can gain access to the CNS (Wekerle
`et al., 1986; Hickey, 1991). Indeed, GA-reactive T cells have
`been directly demonstrated in the CNS of adoptively
`transferred animals (Aharoni et al., 2000). It is assumed
`further that after local recognition of cross-reactive myelin
`degradation products, the GA-specific TH2 cells are stimu-
`lated to secrete TH2-like cytokines, which suppress neigh-
`bouring encephalitogenic T cells via ‘bystander suppression’
`(Aharoni et al., 1997, 1998; Neuhaus et al., 2001).
`Furthermore,
`the locally activated GA-reactive T cells
`produce the neurotrophic factor BDNF (Ziemssen et al.,
`2002). Because the BDNF receptor gp145 trk B is expressed
`in neurons in the immediate vicinity of multiple sclerosis
`plaques (Stadelmann et al., 2002), these neurons should be
`responsive to the T-cell-derived BDNF.
`The basic tenets of the above scenario, especially the GA-
`induced TH2 shift of T cells, are widely accepted. It is
`unclear, however, how the cytokine shift is brought about.
`The most widely held view is that GA acts mainly on T cells,
`and that several factors related to the special antigenic
`properties of GA contribute to the observed TH2 shift. These
`include the properties of GA as a co-polymer, its route of
`administration by frequent s.c. injection and perhaps its local
`presentation to T cells by dermal Langerhans cells. In
`contrast, our results indicate that GA directly affects the
`functional properties of ‘professional’ APCs. Consistent with
`our findings, several recent publications showed that GA had
`
`inhibitory effects on TNF-a production by a monocytic cell
`line (Li et al., 1998) and on IL-12 secretion by in vitro
`generated, monocyte-derived DCs (Hussien et al., 2001;
`Vieira et al., 2003). Furthermore, our results show, as we
`believe for the first time, that GA reduces monocyte reactivity
`in treated patients, and that this effect can be observed with
`‘ex vivo’ monocyte cultures.
`What are the implications of these observations? First, GA
`may alter the properties of APCs in such a way that they
`preferentially induce TH2 responses. This is supported by the
`study of Vieira et al. (2003) who demonstrated that GA-
`treated DCs induce IL-4-secreting TH2 cells in vitro. We
`investigated the effects of GA treatment on the TH2 shift in a
`subset of patients. Five of six patients showed a pronounced
`GA-induced IL-4 production by Elispot assay, defined
`according to Farina et al. (2001) (data not shown). These
`findings indicate that the GA-induced TH2 shift and the
`inhibition of monocyte reactivity usually occur concurrently.
`Secondly, monocyte inhibition per se could be beneficial in
`multiple sclerosis, independently of the TH2 shift of T cells.
`Monocytes and macrophages have an important role in the
`pathogenesis of multiple sclerosis (for a review see Izikson
`et al., 2002). Although the composition of the inflammatory
`infiltrate in the CNS varies depending on the type, stage and
`activity of multiple sclerosis, monocytes/macrophages are
`thought to be key effectors responsible for tissue damage.
`They predominate in active multiple sclerosis lesions, and the
`presence of myelin degradation products inside macrophages
`is one of the most reliable markers of lesional activity. Toxic
`products of activated monocytes/macrophages, such as TNF-
`a, reactive oxygen species and matrix metalloproteinases
`(Bar-Or et al., 2003), contribute to myelin injury (for a review
`see Lassmann et al., 2001). Blockade of monocytes is
`therefore an obvious therapeutic goal.
`Thirdly,
`the direct effect of GA on APCs makes it
`necessary to reconsider the concept of ‘antigen selectivity’
`of GA. Although not formally proven, it is widely believed
`that GA preferentially affects myelin basic protein (MBP)-
`specific T-cell responses, at l

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