throbber
Glatiramer acetate-specific T-helper 1- and 2-type
`cell lines produce BDNF: implications for multiple
`sclerosis therapy
`
`Brain (2002), 125, 2381–2391
`
`Tjalf Ziemssen,1 Tania Ku¨mpfel,1,2 Wolfgang E. F. Klinkert,1 Oliver Neuhaus3 and
`Reinhard Hohlfeld1,2
`
`Correspondence to: Dr R. Hohlfeld, Institute for Clinical
`Neuroimmunology, Klinikum Grosshadern, Ludwig
`Maximilians University, Marchioninistrasse 15,
`D-81366 Munich, Germany
`E-mail: hohlfeld@neuro.mpg.de
`
`Downloaded from
`
`by guest on October 30, 2014
`
`cells produce BDNF, we selected four GA-specific, long-
`term T-cell
`lines (TCLs), which were characterized
`according to their cytokine profile by intracellular
`double-fluorescence flow cytometry. Three TCLs (isol-
`ated from a normal subject) had the phenotypes TH1,
`TH1/TH0, and TH0; the fourth, derived from a GA-
`treated patient, had the phenotype TH2. To demon-
`strate BDNF production, we used a combination of RT-
`PCR (reverse transcription-polymerase chain reaction)
`and two specially designed techniques for BDNF protein
`detection: one was based on ELISA (enzyme-linked
`immunosorbent assay) of supernatants from co-cultures
`of GA-specific TCLs plus GA-pulsed antigen-presenting
`cells, and the other on the direct intracellular staining
`of BDNF in individual T cells and flow cytometric
`analysis. The different assays and different TCLs
`yielded similar, consistent results. All four GA-specific
`T-cell lines, representing the major different TH pheno-
`types, could be stimulated to produce BDNF.
`
`1Department of Neuroimmunology, Max Planck Institute of
`Neurobiology, Martinsried, 2Institute for Clinical
`Neuroimmunology and Department of Neurology, Klinikum
`Grosshadern, Ludwig Maximilians University, Munich,
`Germany and 3Department of Neurology, Karl-Franzens-
`University Graz, Austria
`
`Summary
`The clinical effects of glatiramer acetate (GA), an
`approved therapy for multiple sclerosis, are thought to
`be largely mediated by a T-helper 1 (TH1) to T-helper 2
`(TH2)
`shift of GA-reactive T-lymphocytes. Current
`theories propose that activated GA-reactive TH2 cells
`penetrate the CNS, release anti-inflammatory cytokines
`such as interleukin (IL)-4, IL-5 and IL-10, and thus
`inhibit neighbouring inflammatory cells by a mechanism
`termed ‘bystander suppression’. We demonstrate that
`both GA-specific TH2 and TH1 cells produce the neuro-
`trophin brain-derived neurotrophic factor (BDNF). As
`the signal-transducing receptor for BDNF,
`the full-
`length 145 tyrosine kinase receptor (trk) B, is expressed
`in multiple sclerosis lesions, it is likely that the BDNF
`secreted by GA-reactive TH2 and TH1 has neuro-
`trophic effects in the multiple sclerosis target tissue.
`This may be an additional mechanism of action of GA,
`and may be relevant for therapies with altered peptide
`ligands in general. To demonstrate that GA-reactive T
`
`Keywords: multiple sclerosis; altered peptide ligand (APL); immunotherapy; neuroprotection; glatiramer acetate
`
`Abbreviations: APL = altered peptide ligand; APC = antigen presenting cell; BDNF = brain-derived neurotrophic factor;
`ELISA = enzyme-linked immunosorbent assay; FACS = fluorescence-activated cell sorter; FITC = fluorescein
`isothiocyanate; GA = glatiramer acetate; IL = interleukin; MBP = myelin basic protein; PBMC = peripheral blood
`mononuclear cell; PMA = phorbol 12-myristate 13-acetate; RT-PCR = reverse transcription-polymerase cell reaction;
`TCL = T-cell line; TCR = T-cell receptor; TH1 = T-helper 1; TH2 = T-helper 2; trk = tyrosine-receptor kinase
`
`Introduction
`Glatiramer acetate (GA, copolymer 1, Copaxone(cid:226)) is a
`heterogeneous but standardized mixture of synthetic poly-
`peptides consisting of L-glutamic acid, L-lysine, L-alanine
`and L-tyrosine (average molecular mass, 6400 Da). GA has
`been known for a long time to have suppressive and
`ª Guarantors of Brain 2002
`
`protective effects in experimental autoimmune encephalo-
`myelitis, which can be induced in different species by various
`encephalitogenic antigens (Teitelbaum et al., 1972; Webb
`et al., 1975; Teitelbaum et al., 1996; Sela, 1999). More
`recently, GA has also been shown to have beneficial effects
`
`Page 1 of 11
`
`YEDA EXHIBIT NO. 2066
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Downloaded from
`
`by guest on October 30, 2014
`
`2382
`
`T. Ziemssen et al.
`
`on the clinical course and MRI-defined brain lesions of
`patients with multiple sclerosis. As a result, GA is now
`approved for use in the immunomodulatory therapy of
`relapsing-remitting multiple sclerosis (Teitelbaum et al.,
`1997; Comi et al., 2001; Sela et al., 2001; Ziemssen et al.,
`2001)
`Among potential mechanisms, the initial T-helper 1 (TH1)
`type response of GA-treated patients was found to gradually
`shift to a T-helper 2 (TH2) type response (Miller et al., 1998;
`Duda et al., 2000; Gran et al., 2000; Neuhaus et al., 2000; Qin
`et al., 2000). TH1 cells characteristically produce a spectrum
`of ‘proinflammatory’ cytokines such as interferon (IFN)-g,
`interleukin (IL)-2 and IL-12. In contrast, TH2 cells produce
`TH2-type ‘anti-inflammatory’ cytokines, i.e. IL-4, IL-5, IL-6
`and IL-13 (Paul and Seder, 1994; Mosmann and Sad, 1996;
`Allen and Maizels, 1997). An intermediate type of T cell,
`called a TH0 cell, produces both TH1- and TH2-type
`cytokines. Current
`theories propose that
`the GA-specific
`TH2 cells, which are induced and constantly activated during
`treatment, migrate into the CNS and release their TH2-like
`cytokines locally (Aharoni et al., 2000). These cytokines are
`thought to have beneficial effects on the local inflammatory
`milieu and to inhibit the action of encephalitogenic T cells by
`‘bystander suppression’ (Neuhaus et al., 2001).
`Although plausible,
`this scenario may actually be an
`oversimplification as two unexpected and intriguing findings
`suggest. Human immune cells including T-lymphocytes,
`B-lymphocytes and monocytes can produce brain-derived
`neurotrophic factor
`(BDNF)
`(Besser and Wank, 1999;
`Kerschensteiner et al., 1999)—a potent neurotrophin that
`has profound effects on neuronal
`survival and repair
`(Thoenen, 1995; Barde, 1997). Moreover, the receptor for
`BDNF, gp145TrkB, is expressed in neurones and astrocytes
`in multiple sclerosis brain lesions (Stadelmann et al., 2002).
`These findings prompted us to ask the following questions:
`(i) can GA-reactive T lymphocytes produce BDNF, and if so,
`(ii) do TH1-type and TH2-type GA-specific T cells differ in
`their capacity to produce BDNF?
`To answer these questions, we first had to overcome two
`major technical obstacles: (i) adapting our culture system to
`prevent added GA from affecting the BDNF enzyme-linked
`immunosorbent assay (ELISA) and (ii) optimizing the
`intracellular detection of BDNF in individual T-lymphocytes.
`This enabled us to demonstrate formally that GA-specific
`TH1, TH2 and TH0 cells all have the capacity to produce
`BDNF. We therefore postulate that the beneficial effects of
`not only TH2-type, but also TH1-type GA-reactive T cells,
`might, at least partly, be due to their release of BDNF in
`multiple sclerosis lesions.
`
`Material and methods
`Subjects
`Blood samples were drawn from a GA-treated patient (B.K.)
`and a healthy donor (T.Z.) after their informed consent was
`
`given. The patient, a 47-year-old woman, had been diagnosed
`in 1993 to have relapsing-remitting multiple sclerosis. Her
`current Expanded Disability Status Scale (EDSS) (Kurtzke,
`1983) is 1. She has been essentially free of exacerbations
`since GA treatment was started in December 1998. Her
`human leukocyte antigen (HLA) class II phenotype is DR2/
`DR4. The HLA class II phenotype of the healthy volunteer
`(T.Z.), a 28-year-old postdoctoral fellow, is DR8/DR13. HLA
`typing was kindly performed by Drs E. Albert and S. Scholz,
`Department of
`Immunogenetics, University of Munich,
`Germany.
`
`Selection and culture of GA-specific T-cell lines
`(TCLs)
`GA-specific CD4+ TCLs were selected from peripheral blood
`mononuclear cells (PBMCs) using a split-well
`technique
`(Kitze et al., 1988; Pette et al., 1990; Neuhaus et al., 2000).
`GA (batch 242992899, average molecular mass 6400 Da) was
`obtained from Teva Pharmaceutical Industries, Petah Tiqva,
`Israel.
`the
`Four GA-specific CD4+ TCL representatives of
`phenotypes TH1 (TZ-COP-1), TH1/0 (TZ-COP-3), TH0
`(TZ-COP-5)
`and TH2
`(BK-M6-COP-7) were
`used
`(Neuhaus et al., 2000). The protocols for fluorescence-
`activated cell sorter (FACS) phenotyping of the TCLs are
`described below. The TCLs TZ-COP-1, TZ-COP-3 and TZ-
`COP-5 were obtained from the healthy untreated subject T.Z.
`The TH2-type TCL, BK-M6-COP-7, was obtained from the
`GA-treated patient B.K. This TCL was originally described
`by Neuhaus et al. (2000).
`
`Stimulation of GA-specific TCL with GA-pulsed
`antigen presenting cells (APCs)
`In pilot experiments, the GA in the culture supernatants was
`occasionally observed to interfere with the BDNF ELISA,
`especially in the low range of BDNF concentrations
`(M. Kerschensteiner, W. Klinkert and T. Ziemssen, unpub-
`lished data). We therefore established a rigorous antigen-
`pulsing protocol to minimize these soluble GA concentra-
`tions. This protocol was used in all the experiments reported
`here. Thrombocyte-depleted APCs, X-irradiated with 40 Gy
`(Stabiloplan 2; Siemens, Erlangen, Germany), were incu-
`bated (‘pulsed’) with GA at a final concentration of 400 mg/ml
`for 4 h. The GA-pulsed APCs were washed twice before
`being used to stimulate the GA-specific TCLs. The same
`protocol was used for parallel proliferation assays.
`Proliferation was measured by [3H]thymidine uptake as
`described previously (Neuhaus et al., 2000).
`For proliferation and BDNF secretion (ELISA) assays, 105
`washed GA-specific TCL cells were stimulated in RPMI 1640
`medium supplemented with 5% foetal calf serum (FCS), 1%
`glutamine and 1% penicillin/streptomycin (all from Gibco
`BRL, Gaithersburg, MD, USA) with 9 3 104 GA-pulsed
`
`Page 2 of 11
`
`YEDA EXHIBIT NO. 2066
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Downloaded from
`
`by guest on October 30, 2014
`
`APCs. Supernatants were removed after 72 h and analysed for
`BDNF concentrations by ELISA (see below). For prolifer-
`ation assays, parallel cultures were labelled after 48 h with
`[3H]thymidine (0.2–0.5 mCi per well; Amersham Buchler,
`Braunschweig, Germany; 1 mCi = 37 kBq) and harvested 16–
`18 h later. [3H]thymidine incorporation was measured with a
`direct b-counter
`(Matrix TM 96; Packard, Frankfurt,
`Germany). For reverse transcription-polymerase chain reac-
`tion (RT-PCR) analysis of BDNF transcription, RNA was
`extracted (see below) from cell pellets after 24 h of incuba-
`tion with GA (50 mg/ml).
`
`Phenotypic characterization of the GA-specific
`TCL by flow cytometry
`TCLs were stained with monoclonal antibodies directed
`against CD3 (mouse IgG1, biotinylated;
`Immunotech,
`Marseille, France) plus
`streptavidin-phycoerythrin (PE)
`(PharMingen, San Diego, CA, USA), CD4 (mouse IgG1,
`PE-labelled; PharMingen) and CD8 (mouse IgG1, fluorescein
`isothiocyanate (FITC)-labelled; Becton Dickinson, San Jose,
`CA, USA) or the corresponding non-immune isotype controls
`[mouse IgG1, biotin- or FITC-labelled (PharMingen); PE-
`labelled (Becton Dickinson)]. The T-cell receptor (TCR) Vb
`(variable region) repertoire was analysed using monoclonal
`antibodies that recognize the following subfamilies: Vb2,
`Vb3, Vb3.1, Vb5.3, Vb7, Vb7.1, Vb8, Vb9, Vb11, Vb12,
`Vb13.1, Vb13.2, Vb13.6, Vb14, Vb17, Vb18, Vb20,
`Vb21.3, Vb23 (Immunotech) and Vb5a, Vb5b, Vb6.7
`(T-cell Diagnostics, Woburn, MA, USA). Monoclonal anti-
`bodies and isotype controls [mouse IgG1 (Becton Dickinson);
`mouse IgG2a and IgG2b (Cymbus, Chandlers Ford,
`Hampshire, UK)] were visualized with a FITC-labelled goat
`anti-mouse IgG antibody (Jackson ImmunoResearch, West
`Grove, PA, USA). The stained cells were analysed using a
`FACScan (Becton Dickinson).
`
`Intracellular flow cytometry analysis of cytokine
`profile and BDNF production
`Intracellular flow cytometry of the TCLs was performed 8–
`10 days after restimulation in the absence of viable APCs.
`GA-specific TCL were stimulated with phorbol 12-myristate
`13-acetate (PMA, 2.0 mg/ml) and ionomycin (250 pg/ml)
`(both from Sigma, St Louis, MO, USA) for 3 h (cytokine
`profile) or 12 h (BDNF production); the last 2 h (cytokine
`profile) or 6 h (BDNF production) in the presence of
`monensin (2 mmol/l; Sigma). The T cells were then washed
`with phosphate-buffered saline (PBS) fixed with 4%
`paraformaldehyde (Merck, Darmstadt, Germany) and perme-
`abilized with 0.1% saponin/PBS (Sigma). For the character-
`ization of the cytokine profile, the T cells were then stained
`using appropriate concentrations of monoclonal antibody
`directed
`against
`IL-4
`(mouse
`IgG1,
`PE-labelled;
`PharMingen) and IFN-g
`(mouse IgG1, FITC-labelled;
`
`BDNF production by GA-specific TCL
`
`2383
`
`PharMingen) or the corresponding isotype controls [mouse
`IgG1, PE-labelled (Becton Dickinson); mouse IgG1, FITC-
`labelled (Immunotech)]. For the detection of intracellular
`BDNF production, activated and non-activated T cells were
`stained with a chicken IgY antibody against human BDNF or,
`as an isotype control, with a chicken control immunoglobulin
`IgY (both Promega, Madison, WI, USA). IgY, the 180 kDa
`chicken IgG homologue, can be produced in chickens against
`certain biological antigens that fail
`to elicit a humoral
`immune response in other mammals due to species related-
`ness. The antibody is highly specific for BDNF. A rabbit anti-
`chicken Ig antibody (FITC-labelled; Promega) was used as
`secondary antibody.
`The untransfected murine ecotropic packaging line
`GP+E86 was used as a negative control for intracellular
`BDNF FACS staining. The packaging line transfected with
`the retroviral vector pLXSN into which BDNF cDNA was
`cloned (kindly provided by R. Kramer, Max-Planck-Institute
`of Neurobiology, Martinsried, Germany) served as a positive
`control (Flugel et al., 2001).
`The stained cells were analysed using a FACScan (Becton
`Dickinson). On a dot plot showing forward and side scatter,
`lymphoid cells were gated for further analysis. Dead cells
`were excluded by gating.
`
`Quantification of BDNF protein secretion in
`culture supernatants by ELISA
`BDNF protein concentrations were determined in duplicate
`using
`a
`sensitive
`sandwich ELISA as
`described
`previously (Kerschensteiner et al., 1999). In brief, 96-well
`flat-bottomed plates were coated with the chicken anti-human
`BDNF IgY antibody (Promega) in 0.025M NaHCO3 and
`0.025M Na2CO3 (pH 8.2). Recombinant human BDNF
`(used as standard; Research Diagnostics, Flanders, PA,
`USA) was used in serial dilutions and cell supernatants in
`1 : 2 dilutions. Bound BDNF was detected by incubating
`plates with a mouse anti-human BDNF antibody (Research
`Diagnostics) followed by peroxidase-conjugated goat anti-
`mouse IgG (Dianova, Hamburg, Germany). The plates were
`developed using a 3,3¢,5,5¢-tetramethyl-benzidine liquid
`substrate system (Sigma); the optical density was determined
`at 450 nm.
`
`RT-PCR analysis of BDNF transcription
`Total cellular RNA was extracted using the RNA extraction
`system of Qiagen (Hilden, Germany) with DNase digestion.
`The RNA (1 mg) was transcribed with oligo(dt) primers,
`Superscript(cid:226) Reverse Transcriptase (both Gibco BRL) and
`dNTP (MBI Fermentas, St Leon-Rot, Germany). All PCR
`in a total volume of 50 ml
`reactions were carried out
`containing 2 U Taq polymerase (Qiagen), 200 mM of each
`dNTP, and 15 pmol of each primer for 35 PCR cycles with an
`annealing temperature of 60(cid:176)C. The correct size of the bands
`
`Page 3 of 11
`
`YEDA EXHIBIT NO. 2066
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`challenged with myelin basic protein (MBP) as was pre-
`viously reported for murine and human GA-specific T cells
`(data not shown) (Aharoni et al., 1998; Neuhaus et al., 2000).
`Irradiated APCs alone did not proliferate, regardless of
`whether they were pulsed with GA or not (Fig. 2). When T
`cells were added to unpulsed APCs,
`there was a small
`background proliferation that was at least six times lower than
`the proliferation in the presence of GA-pulsed APCs (Fig. 2,
`right columns).
`The results of the BDNF ELISA indicate that the irradiated
`APCs (that is, PBMCs containing monocytes, T cells and
`B cells) produced small but clearly detectable amounts of
`BDNF (Fig. 2, left columns in left panels), although they did
`not proliferate (Fig. 2, left columns in right panels). There
`was a tendency for higher BDNF production by GA-pulsed
`APCs alone. All GA-specific TCLs showed an increased
`BDNF production after incubation with GA-pulsed APCs
`(Fig. 2, right columns in left panels).
`
`Downloaded from
`
`by guest on October 30, 2014
`
`RT-PCR analysis of BDNF transcription in
`GA-specific TCL
`The transcription of BDNF mRNA in GA-specific T-cells
`was analysed by RT-PCR. In contrast to the BDNF protein
`secretion assay, the cells were harvested after a culture period
`of 24 h for RNA extraction and reverse transcription. The
`expression of BDNF was examined using RT-PCR in
`comparison with the housekeeping gene b-actin. There was
`no detectable contamination by genomic DNA, i.e. there were
`no bands in the negative BDNF and b-actin controls using
`RNA samples processed in the absence of reverse transcrip-
`tase (Fig. 3). Consistent with the BDNF protein data shown in
`Fig. 2, RT-PCR revealed weak bands in the absence of GA
`and stronger bands in the presence of GA (Fig. 3).
`
`Detection of BDNF protein in GA-specific
`T-cells by intracellular flow cytometry
`To confirm that the GA-specific T-cells are the source of the
`GA-induced BDNF release, we developed a new intracellular
`staining technique suitable for flow cytometry and FACS
`analysis of intracellular BDNF production. This method
`allows the analysis of BDNF production by individual
`unstimulated und stimulated T cells. Since the analysis was
`performed at least 8–10 days after the last restimulation with
`antigen and irradiated APCs, the cultures contained only T
`cells in the absence of viable APCs. Before intracellular
`staining and FACS analysis, the T cells were stimulated with
`ionomycin and PMA; this mode of stimulation does not
`require the presence of APCs (Dayton et al., 1994). A BDNF-
`transfected and non-transfected murine retroviral packaging
`line was used as the positive and negative controls (Fig. 4, top
`panels).
`This new method was able to detect BDNF produced by the
`GA-specific TCLs, even without stimulation (left panels in
`
`2384
`
`T. Ziemssen et al.
`
`was determined by comparison with a DNA mass standard
`(SM0403, MBI Fermentas). RNA samples incubated in the
`absence of reverse transcriptase were used as negative
`controls to exclude genomic contamination. The primer
`sequences were as follows: BDNF forward 5¢-AGCGTG-
`AATGGGCCCAAGGCA-3¢
`(position 208–228); BDNF
`5¢-TGTGACCGTCCCGCCCGACA-3¢
`reverse
`(position
`570–551); b-actin
`5¢-CCTCGCCTTTGCCGA-
`forward
`TCC-3¢ (position –8 to 9); and b-actin reverse 5¢-GGATCT-
`TCATGAGGTAGTCAGTC-3¢ (position 623–604).
`
`Results
`Phenotypic characterization of GA-specific
`TCLs
`To analyse the production of BDNF by GA-specific T cells
`after antigen challenge in vitro, four GA-specific long-term
`TCLs were used—one TCL obtained from the GA-treated
`multiple sclerosis patient (B.K.) and three TCLs from the
`healthy donor (T.Z.). The TCLs were selected with the split-
`well cloning technique (Kitze et al., 1988; Pette et al., 1990).
`FACS analysis showed that all GA-specific TCLs had a
`CD3+CD4+CD8– phenotype (data not shown). The TCL from
`the multiple sclerosis patient (BK-M6-COP-7) was pre-
`viously characterized as part of another study (Neuhaus et al.,
`2000). The other three TCLs were analysed as to their TCR
`Vb usage. A panel of 23 different anti-TCR-Vb antibodies
`was used to demonstrate the oligoclonal nature of the TCLs.
`As characteristically seen with TCLs selected with the split-
`well protocol (Kitze et al., 1988; Pette et al., 1990), each line
`reacted predominantly with one monoclonal antibody from
`the panel of anti-TCR Vb antibodies (Fig. 1). The three
`investigated TCLs used different Vb elements. The fact that
`the TCL populations predominantly stained positive for only
`the 23 Vb elements indicated that
`one of
`they were
`oligoclonal (Fig. 1).
`Fig. 1 shows the cytokine profile of the TCLs for IL-4 and
`IFN-g. Whereas the TCLs TZ-COP-1 and TZ-COP-3 dis-
`played a TH1 or TH1/TH0 phenotype, TZ-COP-5 cells
`produced both IL-4 and IFN-g (TH0 phenotype). As
`described previously (Neuhaus et al., 2000), the TCL from
`the GA-treated multiple sclerosis patient (BK-M6-COP-7)
`had a stable TH2 cytokine profile (Fig. 1).
`
`GA-induced proliferation and BDNF protein
`secretion by GA-specific TCLs
`To demonstrate the ability of GA-specific T cells to secrete
`BDNF upon stimulation, we quantified the amount of BDNF
`in supernatants of GA-stimulated T cells and, in parallel,
`assessed the proliferative response to GA (Fig. 2). All GA-
`specific TCLs showed a specific proliferative response to GA
`with stimulation indices ranging between 6.5 and 21.0 over
`several re-stimulations. None of the tested GA-specific TCLs
`showed a proliferative response or cytokine production when
`
`Page 4 of 11
`
`YEDA EXHIBIT NO. 2066
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Downloaded from
`
`by guest on October 30, 2014
`
`BDNF production by GA-specific TCL
`
`2385
`
`Fig. 1 Phenotypical characterization of the GA-specific TCLs (TZ-COP-1, TZ COP-3, TZ-COP-5, BK-M6-COP-7) as to their Vb TCR
`usage (left panels: fine lines represent isotype controls and bold lines represent the indicated TCR Vb antibodies) and their cytokine
`secretion profile analysed by intracellular double-fluorescence cytometry (middle panels with isotype controls; right panels with the
`cytokine profile). ND* = not done; the Vb TCR usage of the published GA-specific T cell line BK-M6-COP-7 was not determined
`(Neuhaus et al., 2000).
`
`Fig. 4; fine lines represent isotype controls). After stimulation
`with ionomycin and PMA,
`there was an increase of
`intracellular BDNF production (Fig. 4, right panels). The
`results of this intracellular assay of BDNF expression are
`consistent with the results of the ELISA and RT-PCR
`analysis.
`
`Discussion
`Technical aspects of the study
`We clearly show that GA-specific, activated TH0, TH1 and
`TH2 cells produce the neurotrophic factor BDNF—not only
`at the transcriptional (mRNA) level by RT-PCR, but also at
`the protein level—using the newly developed assays for
`
`Page 5 of 11
`
`YEDA EXHIBIT NO. 2066
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`2386
`
`T. Ziemssen et al.
`
`Downloaded from
`
`by guest on October 30, 2014
`
`Fig. 2 BDNF secretion (left panels) and proliferation (right panels) of the examined GA-specific TCLs (TZ-COP-1, TZ-COP-3, TZ-COP-
`5, BK-M6-COP-7) after restimulation with GA. Irradiated autologous unpulsed and GA-pulsed APCs were analysed alone and in co-
`culture with the examined TCL as described in Material and methods. Filled columns indicate mean of duplicates and vertical lines
`indicate individual measurements. Different ordinate scales are used. The statistical significance of the difference between BDNF
`production of (unpulsed APC+TCL) versus (GA-pulsed APC+TCL) was P < 0.0003 (t-test; all TCL included).
`
`Page 6 of 11
`
`YEDA EXHIBIT NO. 2066
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Downloaded from
`
`by guest on October 30, 2014
`
`BDNF production by GA-specific TCL
`
`2387
`
`To show conclusively that the BDNF is produced by the
`GA-specific T cells rather than the APCs, we optimized its
`detection in individual T cells in the absence of APCs. Similar
`assays are widely used for the intracellular staining of various
`cytokines, including IL-4 and IFN-g (see Fig. 1); the T cells
`are typically stimulated with ionomycin and PMA [an
`antigen-independent maximal stimulus for T cells (Dayton
`et al., 1994)] before being fixed and permeabilized. In
`preliminary experiments with a series of monoclonal and
`polyclonal antibodies to BDNF, we obtained the best results
`with a highly specific chicken anti-human BDNF antiserum
`(Fig. 4) that also detects BDNF in ELISA and specifically
`labels a murine retroviral packaging cell line transfected with
`BDNF-cDNA. Together with the results of RT-PCR and
`ELISA, these results formally establish that human GA-
`specific T cells of different TH types can indeed produce
`BDNF.
`
`Implications for the presumed mechanism of
`action of GA
`Our findings have obvious implications for the presumed
`mechanism of action of GA. According to current theory, the
`beneficial effects of GA are mainly mediated by a population
`of GA-reactive TH2 cells. 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., 2000a; Farina et al.,
`2001). Their constant activation by daily immunization
`enables them to enter the CNS (Wekerle et al., 1986;
`Hickey et al., 1999). Indeed, transferred GA-reactive T cells
`have been directly demonstrated in the CNS of recipient mice
`(Aharoni et al., 2000). It is further assumed, that after local
`recognition of cross-reactive myelin degradation products,
`the GA-specific TH2 cells are stimulated to secrete anti-
`inflammatory cytokines, which in turn induce bystander
`suppression in neighbouring encephalitogenic T cells
`(Aharoni et al., 1997; Aharoni et al., 1998; Neuhaus et al.,
`2001). Our present findings imply that the locally activated
`GA-reactive TH2 cells produce not only protective TH2
`cytokines, but also BDNF. They further indicate that GA-
`specific TH1 cells, which are reduced but are still present in
`GA-treated patients (Neuhaus et al., 2000; Farina et al.,
`2001), could also act as a local source of BDNF.
`BDNF is one of the most potent factors supporting
`neuronal survival and regulating neurotransmitter release
`and dendritic growth (Thoenen, 1995; Lewin and Barde,
`1996; Barde, 1997). Several studies have shown that BDNF
`can rescue injured or degenerating neurones and induce
`axonal outgrowth, remyelination and regeneration (Yan et al.,
`1992; Gravel et al., 1997). Moreover, it can also protect axons
`from elimination during development, or from degeneration
`after axotomy, or in experimental neurodegenerative disease
`(Mitsumoto et al., 1994). Most known BDNF functions are
`signalled via the full-length gp145 tyrosine kinase receptor
`(trk) B (Bothwell, 1995); intriguingly, it is found in neurones
`
`Fig. 3 The BDNF transcription by unstimulated and GA-
`stimulated GA-specific TCLs (TZ-COP-1, TZ-COP-3, TZ-COP-5,
`BK-M6-COP-7) was examined by RT-PCR analysis of BDNF
`transcripts. The transcription of the housekeeping gene b-actin was
`used for comparison. RNA samples transcribed in the absence of
`reverse transcriptase were used as negative controls to exclude
`genomic contamination.
`
`BDNF secretion and synthesis of BDNF in individual T cells.
`The results from all assays consistently showed a low level of
`basal secretion of BDNF by GA-specific T cells and an
`increase of BDNF production after stimulation. For our
`analysis, we selected four well-characterized prototypic GA-
`specific TCLs. Similar
`results were obtained with >20
`additional GA-specific TCLs (data not shown).
`In pilot experiments, the presence of soluble GA in culture
`supernatants sometimes caused elevated ELISA readings for
`BDNF (M. Kerschensteiner, W. Klinkert and T. Ziemssen,
`unpublished data). To prevent that, we pre-pulsed the APCs
`with GA and washed them, before co-culturing with our
`highly purified long-term GA-specific oligoclonal TCL. In
`this culture system, the irradiated PBMC produced small
`amounts of BDNF even in the absence of GA-specific TCLs
`(Fig. 2). This was expected because the irradiated PBMC
`preparations contained T cells, and GA strongly stimulates T
`cells from naı¨ve human subjects (Duda et al., 2000a; Duda
`et al., 2000b; Farina et al., 2001). Although proliferation of
`the irradiated PBMC was abolished completely, some GA-
`stimulated BDNF production evidently persists (Fig. 2).
`When the GA-specific TCLs were added, BDNF production
`increased and was highest in the presence of GA-pulsed
`APCs. Qualitatively similar results were obtained in the
`parallel proliferation assays. All four tested TCLs yielded
`consistent results in all assays (Fig. 2).
`
`Page 7 of 11
`
`YEDA EXHIBIT NO. 2066
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`2388
`
`T. Ziemssen et al.
`
`Downloaded from
`
`by guest on October 30, 2014
`
`Fig. 4 Intracellular BDNF production of unstimulated (left panel) and PMA- and ionomycin-stimulated (right panels) GA-specific TCLs
`(TZ-COP-1, TZ-COP-3, TZ-COP-5, BK-M6-COP-7). Fine lines represent isotype controls and bold lines represent the anti-BDNF-
`antibody. The untransfected and BDNF-transfected murine ecotropic packaging line GP+E86 was used as the negative and positive control
`for the intracellular BDNF FACS staining.
`
`in the immediate vicinity of multiple sclerosis plaques as well
`as
`in reactive astrocytes
`in multiple sclerosis lesions
`(Stadelmann et al., 2002). Therefore, T-cell-derived BDNF
`could directly act on target cells expressing the appropriate
`trkB receptor.
`
`BDNF has been immunolocalized in inflammatory cells in
`active multiple sclerosis lesions (Stadelmann et al., 2002).
`This indicates that, even in untreated multiple sclerosis
`patients, inflammatory cells might have a beneficial (neuro-
`protective) effect
`in addition to their
`(probably major)
`
`Page 8 of 11
`
`YEDA EXHIBIT NO. 2066
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Downloaded from
`
`by guest on October 30, 2014
`
`detrimental role (Hohlfeld et al., 2000). Indeed, the concept
`of ‘neuroprotective autoimmunity’ has attracted considerable
`attention and is supported by experimental evidence from
`various animal models (Kipnis et al., 2000; Schori et al.,
`2001; Schwartz, 2001). It is conceivable that
`there is a
`complex interplay between detrimental and beneficial factors,
`and mediators in the inflammatory milieu of multiple
`sclerosis lesions. BDNF imported by GA-reactive T cells
`might help tip the balance in favour of the beneficial,
`neuroprotective influences. Indeed, in a rat neurodegenerative
`model, Kipnis and colleagues showed that rat immune cells
`stimulated by immunization with GA secreted neurotrophic
`factors upon restimulation with GA (Kipnis et al., 2000).
`The following scenario would accommodate both the
`findings in experimental autoimmune encephalomyelitis and
`the observations in human multiple sclerosis. GA-specific
`activated T cells can pass through the blood-brain barrier.
`Inside the CNS, some GA-specific T cells cross-react with
`products of local myelin turnover presented by local APCs. In
`our previous studies, 8–15% of GA-reactive TCLs cross-
`reacted with MBP as indicated by cytokine secretion, but not
`proliferation (Neuhaus et al., 2000). Recognition of cross-
`reactive antigens at the lesion site seems to be necessary for
`reactivation of the protective T cells and for their neuropro-
`tective effect. In rat models, only MBP- and GA-specific, but
`not ovalbumin-specific, T cells confer neuroprotection
`(Moalem et al., 1999). After reactivation in situ, GA-specific
`TH2-like regulatory T cells would not only provide anti-
`inflammatory cytokines like IL-4, IL-5, IL-13, and trans-
`(TGF)-b,
`forming
`growth
`factor
`but
`also BDNF.
`Furthermore, TH1-like GA-reactive T cells, which are
`reduced but still present in GA-treated patients (Neuhaus
`et al., 2000; Farina et al., 2001), would also have the capacity
`to release BDNF in multiple sclerosis lesions.
`
`Implications for therapy with other altered
`peptide ligands
`The above concept may be extended to other types of
`immunomodulatory therapy, especially with ‘altered peptide
`ligands’
`(APLs). By definition,
`they are derived from
`immunogenic peptide antigens by the selective alteration of
`one or more T-cell receptor-contacting residues. A good
`example is an APL designed from the myelin basic protein
`peptide 83–99 (Bielekova et al., 2000; Genain and Zamvil,
`2000; Kappos et al., 2000). This APL has been tested in
`clinical trials in multiple sclerosis patients. Despite several
`adverse effects, there was some evidence for partial efficacy
`and for a TH1-to-TH2 shift during treatment with low doses
`of the APL (Bielekova et al., 2000; Crowe et al., 2000;
`Kappos et al., 2000). The rationales for APL and GA
`treatment are very similar: APLs are able to expand
`populations of TH0 and TH2 cells that have specificity for
`the APL itself, but they can also cross-react with the native
`peptide. Thus, T cells specific for an APL analogue of myelin
`
`BDNF production by GA-specific TCL
`
`2389
`
`antigen will be able to penetrate the CNS, where they can
`down-regulate encephalitogenic inflammatory cells via ‘by-
`stander
`suppression’
`(Steinman, 1996; Hohlfeld, 1997;
`Nicholson et al., 1997; Genain and Zamvil, 2000). It should
`be noted, however, that TH2 responses can in principle
`contribute to myelin damage (Genain et al., 1996).
`Extrapolating from our present results to APL therapies in
`general, we would expect that APL-specific TH2 and TH1
`cells are also capable of producing BDNF (as do GA-specific
`TH2 and TH1 cells). However, for APL therapy to work, it is
`obviously cru

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