throbber
Journal of the Neurological Sciences 259 (2007) 27 – 37
`
`www.elsevier.com/locate/jns
`
`Pharmacokinetics and pharmacodynamics of the interferon-betas,
`glatiramer acetate, and mitoxantrone in multiple sclerosis
`Oliver Neuhaus ⁎, Bernd C. Kieseier, Hans-Peter Hartung
`
`Department of Neurology, Heinrich Heine University, Moorenstraße 5, D-40225 Düsseldorf, Germany
`
`Received 2 March 2006; received in revised form 25 April 2006; accepted 1 May 2006
`Available online 27 March 2007
`
`Abstract
`
`Five disease-modifying agents are currently approved for long-term treatment of multiple sclerosis (MS), namely three interferon-beta
`preparations, glatiramer acetate, and mitoxantrone1. Pharmacokinetics describes the fate of drugs in the human body by studying their
`absorption, distribution, metabolism and excretion. Pharmacodynamics is dedicated to the mechanisms of action of drugs. The understanding
`of the pharmacokinetics and pharmacodynamics of the approved disease-modifying agents against MS is of importance as it might contribute
`to the development of future derivatives with a potentially higher efficacy and a more favourable safety profile. This article reviews data thus
`far present both on the pharmacokinetics as well as on the putative mechanisms of action of the interferon-betas, glatiramer acetate, and
`mitoxantrone in the immunopathogenesis of MS.
`© 2007 Elsevier B.V. All rights reserved.
`
`Keywords: Multiple sclerosis; Immunotherapy; Immunomodulation; Immunosuppression; Interferon-beta; Glatiramer acetate; Mitoxantrone
`
`1. Introduction
`
`Multiple sclerosis (MS) is the most common chronic
`central nervous system (CNS) disorder of younger adults and
`a major cause of lasting neurological disability [1].
`Pharmacological therapeutic approaches in MS are based
`on three principles. Apart from (i) treatment of relapses by
`corticosteroids and, in some cases, plasma exchange, and (ii)
`symptomatic treatment, (iii) immunomodulatory or immu-
`nosuppressive long-term treatment with the aim of modify-
`ing the disease course represents the mainstay. However, as
`MS is a chronic disorder requiring chronic therapy, the
`optimal treatment choice for individual patients is often
`difficult to make and in many cases is discussed controver-
`sially [2].
`
`⁎ Corresponding author. Tel.: +49 211 81 17880; fax: +49 211 81 18469.
`E-mail address: oliver.neuhaus@uni-duesseldorf.de (O. Neuhaus).
`1 A sixth disease-modifying agent, natalizumab, has been approved for
`treatment of multiple sclerosis after acceptance of this manuscript. Its
`pharmacokinetics and pharmacodynamics are not subject of this review.
`
`0022-510X/$ - see front matter © 2007 Elsevier B.V. All rights reserved.
`doi:10.1016/j.jns.2006.05.071
`
`The rationale to influence immunological activity in MS
`is to suppress ongoing inflammation with an aim to prevent
`myelin and axonal damage and thus to prevent clinical
`disability. For years, a number of immunosuppressive agents
`(i.e. inhibitors of crucial components of the immune system
`causing generalized immune dysfunction) were used off
`label, the potential beneficial effects of which in MS were
`limited by systemic adverse effects, such as increased risk of
`cancer or infection. Examples are azathioprine (licensed for
`MS treatment in some countries) or cyclophosphamide. In
`the 1990s, two classes of immunomodulatory agents were
`approved for the treatment of relapsing–remitting MS,
`namely interferon (IFN)-beta (IFN-β1a or IFN-β1b) as
`well as glatiramer acetate (GA) [3–5]. Immunomodulators –
`without generally suppressing immunological properties –
`shift immune responses from pro-inflammatory autoimmune
`conditions (mediated by TH-1 cytokines released from
`autoreactive T cells) towards a more beneficial anti-inflam-
`matory environment (mediated through TH-2 cytokines
`secreted by regulatory T cells) [6]. Both IFN-β and GA have
`been proven at least partially effective in relapsing–remitting
`MS as assessed in pivotal and subsequent trials [7–9]. In
`
`Page 1 of 11
`
`YEDA EXHIBIT NO. 2012
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`
`28
`
`O. Neuhaus et al. / Journal of the Neurological Sciences 259 (2007) 27–37
`
`secondary progressive MS, treatment options are limited [10].
`All IFN-β preparations have been investigated in phase 3
`clinical trials, but only IFN-β1b, as administered in a European
`Trial [11], in which many of the patients included exhibited
`clinical relapses, significantly delayed time to onset of
`sustained progression of disability [12] as measured by the
`Expanded Disability Status Scale (EDSS) [13]. The more
`recent approval of the immunosuppressant mitoxantrone in
`MS gave clinicians another therapeutical tool for treatment
`of active forms of relapsing–remitting and secondary pro-
`gressive MS [14].
`
`2. Pharmacokinetics
`
`Pharmacokinetics describes absorption, distribution, me-
`tabolism and elimination of a given drug. The action and fate
`of this drug in the body over a period of time is investigated,
`including the processes of permeation through barriers
`between compartments, localization in tissues and biotrans-
`formation. The pharmacokinetics of the drug is directly
`related to its pharmacodynamics, i.e. its interaction with the
`body.
`
`3. Pharmacokinetics of interferon-beta 1a and
`interferon-beta 1b
`
`Human IFN-β is a polypeptide naturally produced by
`human fibroblasts (predominantly but not exclusively, as
`there are reports of other sources of natural IFN-β, such as
`dendritic cells or
`retinal glial cells; upon stimulation,
`
`virtually all mammalian cells can produce IFN-β). IFN-β
`is highly species-specific, i.e. it is effective only in the
`human organism. It consists of 166 amino acids forming five
`α helices (Fig. 1). IFN-β1a is produced in Chinese hamster
`ovary cells and is pharmacologically identical with the natu-
`ral form, i.e. it is glycosylated by oligosaccharides at Asn 80;
`IFN-β1b is produced in E. coli and has slight differences
`compared to the natural form: it is not glycosylated, Met 1 is
`lacking (i.e. IFN-β1b has 165 amino acids), and Cys 17 is
`replaced by Ser. In both IFN-β forms, a disulphide bridge
`extends between Cys 31 and Cys 141. The optimal dosage
`and route of administration (subcutaneously vs. intramus-
`cularly) and the resulting pharmacodynamic properties of
`IFN-β are subjects of controversial discussion [15–19].
`IFN-β1a is a lyophilized glycoprotein produced in
`mammalian cells using the natural human gene sequence
`[20]. Two preparations are licensed for treatment of MS,
`30 μg once a week administered by intramuscular injection,
`and 22 or 44 μg administered three times a week
`subcutaneously. In a comparative study of three routes of
`administration of 60 μg IFN-β1a in healthy donors, the
`intramuscular route was observed to induce the highest area
`under curve (AUC) for serum IFN activity as compared to
`subcutaneous and intravenous routes [20]. This putative
`disadvantage of the subcutaneous route is thought to be
`circumvented by the higher weekly dose. However,
`the
`results of direct dosing comparison studies are contradictory:
`in the pivotal trial of IFN-β1a administered subcutaneously,
`the 44 μg three times a week group was shown to exhibit
`more beneficial effects as compared to the 22 μg three times
`
`Fig. 1. Structure of interferon-beta. Two recombinant interferon (IFN)-β preparations are approved for MS treatment. IFN-β1a is pharmacologically identical
`with the natural form, i.e. it is glycosylated by oligosaccharides at Asn 80; IFN-β1b has slight differences compared to the natural form: it is not glycosylated,
`Met 1 is lacking (i.e. IFN-β1b has 165 amino acids), and Cys 17 is replaced by Ser. In both IFN-β forms, a disulphide bridge extends between Cys 31 and Cys
`141. Structurally important amino acids are highlighted in red.
`
`Page 2 of 11
`
`YEDA EXHIBIT NO. 2012
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`
`O. Neuhaus et al. / Journal of the Neurological Sciences 259 (2007) 27–37
`
`29
`
`Fig. 2. Structure of glatiramer acetate. Glatiramer acetate (GA) is the acetate salt of a standardized, randomized mixture of synthetic polypeptides consisting of
`L-glutamic acid, L-lysine, L-alanine, and L-tyrosine with a defined molar residue ratio of 0.14 : 0.34 : 0.43 : 0.09 and an average length of 45 to 100 amino acids.
`One possible example sequence is shown here.
`
`a week group [9]. A similar dose-dependent effect was
`observed in a study comparing 44 μg vs. 22 μg subcutane-
`ously once a week (a dose that is not licensed for treatment of
`MS) [21]. In contrast, a study comparing 30 μg vs. 60 μg
`(double dose) once a week intramuscularly did not reveal
`any differences [22]. Taken together, the optimal route, dose
`and frequency of IFN-β1a in MS remains elusive.
`IFN-β1b is a lyophilized protein produced by DNA
`recombinant technology using E. coli and has a molecular
`weight of 18.5 kDa. It is combined with mannitol and human
`albumin in order to have a neutral pH of 7.2. The approved
`dose is 250 μg every other day administered by subcutane-
`ous injection [23]. A comparative phase III trial assessing the
`double dose of 500 μg every other day subcutaneously is
`currently underway.
`
`4. Neutralizing antibodies to interferon-beta
`
`A major issue of the pharmacokinetics of IFN-β (and also
`affecting its pharmacodynamics)
`is the relevance of
`neutralizing antibodies [15,24–27]. Although it is estab-
`lished that the induction of neutralizing antibodies in patients
`treated with IFN-β reduces its clinical effects and may
`accelerate disease progression [27], the mode of measuring
`the presence of neutralizing antibodies as well as the optimal
`time point (after a given treatment duration vs. when clinical
`non-responsiveness to IFN-β occurs) and the clinical
`consequences are still a matter of debate [26,28].
`
`5. Pharmacokinetics of glatiramer acetate
`
`GA is the acetate salt of a standardized, randomized
`mixture of synthetic polypeptides consisting of L-glutamic
`acid, L-lysine, L-alanine, and L-tyrosine with a defined molar
`residue ratio of 0.14:0.34:0.43:0.09 and an average length of
`45 to 100 amino acids (Fig. 2). The approved dose is 20 mg
`applied subcutaneously every day. After subcutaneous
`administration, GA is quickly degraded to free amino acids
`and small oligopeptides with only 10% remaining at the
`injection site after 1 h [29]. No systemic plasma concentra-
`tions nor any urinary or faecal excretion are detectable. Due
`to its high polarity and hydrophilic nature, the penetration of
`GA through the blood–brain barrier is impeded [29]. Thus,
`GA is most unlikely to reach the central nervous system and
`most probably initiates its major immunological effects in
`the periphery [30].
`
`6. Pharmacokinetics of mitoxantrone
`
`Mitoxantrone has a molecular mass of 517.4 Da and is a
`synthetic anthracenedione derivative related to the anthracy-
`clins doxorubicin and daunorubicin (Fig. 3). The agent
`interacts with topoisomerase-2 and causes single and double
`strand breaks by intercalating the DNA. Due to the rapid
`diffusion of mitoxantrone into different tissue compartments
`followed by a relatively slow elimination phase, its report-
`ed maximum plasma half-life varies between 25 and 215 h
`
`Fig. 3. Structure of mitoxantrone. Mitoxantrone is an anthracenedione derivative related to the anthracyclins doxorubicin and daunorubicin.
`
`Page 3 of 11
`
`YEDA EXHIBIT NO. 2012
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`
`30
`
`O. Neuhaus et al. / Journal of the Neurological Sciences 259 (2007) 27–37
`
`[31–36]. Mitoxantrone is eliminated both by the kidney and
`the liver, although less than 11% of mitoxantrone was shown to
`be recovered in urine and less than 25% in faeces within 5 days
`after administration [31]. Furthermore, it has been reported to
`persist in the body for up to 9 months [37]. Reports on the
`ability of mitoxantrone to cross the blood–brain barrier are
`contradictory [37–39]. After intravenous administration, there
`is a linear relationship between the dose and the AUC.
`Mitoxantrone is 78% bound to plasma proteins [40].
`
`7. Pharmacodynamics
`
`Pharmacodynamics assesses the action of a given drug in
`an organism, i.e. the interaction between a drug and the
`body. It is the study of the biochemical and physiological
`effects and the mechanisms of drug action as well as the
`relationship between drug concentration and effect.
`On the molecular level, the binding to and activation or
`deactivation of receptors, signal transduction, induction of
`biosynthesis, impact on transport processes contribute to the
`pharmacodynamics of a drug.
`
`8. Current view on the immunopathogenesis of multiple
`sclerosis
`
`In order to understand the pharmacodynamics of disease-
`modifying treatments in MS,
`the current concept of its
`pathogenetic background needs to be highlighted. Deviations
`of immune responses play a central role in the pathogenesis
`of MS [41] by contributing to the formation and perturbation
`of the MS lesion [42]. The initial inflammatory phase is
`characterized by selective demyelination, and eventually
`subsides to a neurodegenerative phase with axonal loss and
`gliosis [41,43]. With ever novel scientific information
`emerging, concepts regarding the pathogenesis of MS are in
`constant flux and shown in Fig. 4 [44]. In genetically
`susceptible individuals an activation of antigen-specific,
`encephalitogenic T cells occurs in response to a yet
`unidentified trigger, probably in the periphery. Once activated,
`T lymphocytes are able to migrate across the blood–brain
`barrier and invade the CNS . Reactivated there, these T cells
`release pro-inflammatory cytokines and orchestrate destruc-
`tion of the myelin sheath by various immune cell types.
`Pathologically, four different pathologic patterns have been
`described [42]: (i) T-cell and macrophage-mediated demye-
`lination; (ii) antibody-mediated demyelination involving
`complement activation; (iii) distal oligodendrogliopathy and
`oligodendrocyte apoptosis; (iv) primary oligodendrocyte
`degeneration. Interestingly,
`the presence of pattern (iii),
`oligodendrocyte apoptosis, has recently been demonstrated
`in very early MS lesions [45]. In parallel to the autoaggressive
`inflammatory phase causing demyelination, recent evidence
`suggests that axonal loss responsible for irreversible disability
`occurs already early in the disease course and – as the disease
`evolves – predominates in the underlying pathogenetic
`mechanisms [41,46,47].
`
`9. Pharmacodynamics of interferon-beta 1a and
`interferon-beta 1b
`
`IFN-β exerts its immunological effects antigen-indepen-
`dently through reducing the secretion of proteolytic matrix
`metalloproteinases that mediate the migration of T cells
`across biological barriers [48,49], as well as possibly
`downregulating MHC class II on various antigen presenting
`cells (APC) [50,51]. IFN-β has immunomodulatory proper-
`ties, antiviral and antiproliferative effects and promotes cell
`differentiation [52]. Although the mechanisms of action of
`IFN-β are not yet finally understood, there is agreement that the
`major effects are mediated by a transmembranous IFN receptor
`leading to upregulation or downregulation of target genes
`[53,54]. IFN-β shares the receptor with IFN-α as so-called
`“type I interferons” (in contrast to the “type II interferon”, IFN-
`γ). The receptor consists of two chains, IFN-αR1 and IFN-
`αR2, or several subvariants. Ligand binding of IFN-β to the
`extracellular domain of the receptor induces an intracellular
`signal transduction cascade by (i) recruitment and activation of
`the cytoplasmic tyrosine kinase (Tyk)-2 (by IFN-αR1) and
`Janus kinase (Jak)-1 (by IFN-αR12); (ii) subsequent phos-
`phorylation and recruitment of “signal
`transducers and
`activators of transcription” (Stat)-1 and Stat-2 forming a Stat-
`1/Stat-2 heterodimer; (iii) migration of the Stat-1/Stat-2
`heterodimer to the nucleus; (iv) association with the p48
`protein forming the active “IFN-stimulated gene factor-3”; (v)
`binding to promoter elements and initiation of the transcription
`of target genes. The variations of this rough scheme of signal
`transduction – that are based on the wide complexity in the
`single interaction steps – are still not completely identified [53].
`IFN-β exerts a wide and ever broadening range of
`immune effects [52,55]. Among other effects, it
`
`• suppresses T-cell proliferation [56,57];
`• diminishes IFN-γ-induced upregulation of MHC class II
`expression [58,59];
`• downregulates matrix metalloproteinases (MMP) [48,49,
`60]; decreases surface-expressed adhesion molecules [61]
`and increases release of soluble adhesion molecules [60]
`(the latter three actions in concert reduce the T-cell
`migratory potential);
`• induces the production of TH2 cytokines and conversely
`reduces synthesis of TH1 cytokines [62,63];
`• inhibits activation of monocytes [64].
`
`Comparing the three interferon preparations, apart from
`clinical parameters as assessed in the pivotal and subsequent
`trials [7–9], some pharmacodynamical differences have been
`demonstrated. In an open trial comparing the cytokine profiles of
`short-term peripheral blood lymphocyte cultures in patients
`treated with IFN-β1a i.m. and IFN-β1b, respectively, IFN-β1a
`has been shown to increase the concentrations of the TH2
`cytokines IL-4 and IL-10 whereas IFN-β1b reduced the
`concentration of the TH1 cytokine IFN-γ [65]. However, the
`TH net effect of both treatments putatively remained comparable.
`
`Page 4 of 11
`
`YEDA EXHIBIT NO. 2012
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`
`O. Neuhaus et al. / Journal of the Neurological Sciences 259 (2007) 27–37
`
`31
`
`Fig. 4. Hypothetical pathogenesis of MS. Via their T-cell receptor (TCR), pro-inflammatory T cells are activated in the periphery by foreign or self antigens (Ag) presented on major histocompatibility complex class II
`(MHC-II) by antigen-presenting cells (APC), including dendritic cells and B cells. Activated T cells migrate to, adhere at and penetrate through the blood–brain barrier, a step mediated by adhesion molecules,
`proteases and chemokines. Inside the central nervous system (CNS), T cells are reactivated in the context of MHC-II on resident APC, predominantly microglia cells. These reactivated T cells secrete pro-
`inflammatory cytokines, such as interferon (IFN)-γ or interleukin (IL)-2 and induce CNS inflammation by subsequent activation of macrophages, other T cells and B cells as effector cells. Macrophages and T cells
`attack the oligodendrocytic myelin sheath by cytotoxic mediators, mainly tumor necrosis factor (TNF)-α, oxygen (O2) radicals and nitric oxide (NO). B cells differentiate to plasma cells that secrete demyelinating
`antibodies. They can guide and activate macrophages or ignite the complement cascade with assembly of the membrane attack complex which causes pore formation in myelin membranes. Modified from Ref. [44],
`with permission from Elsevier.
`
`Page 5 of 11
`
`YEDA EXHIBIT NO. 2012
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`
`32
`
`O. Neuhaus et al. / Journal of the Neurological Sciences 259 (2007) 27–37
`
`10. Pharmacodynamics of glatiramer acetate
`
`GA acts on “signal one” of T cell activation by binding to
`MHC class II molecules irrespective of haplotype [66], and
`possibly crossreacts with several CNS antigens. It also serves
`as an altered peptide ligand (APL) [67] and induces anti-
`inflammatory regulatory GA-reactive T cells capable of a
`“bystander suppression” once reactivated inside the CNS
`[68].
`In contrast to the multiple immunological effects of IFN-β
`that are antigen-nonspecific, the immunomodulatory potential
`of GA is based on immune cells specific for myelin basic
`protein (MBP) and probably other myelin antigens [52,68,69].
`Four major mechanisms of GA have been identified, namely
`
`• competition of GA with MBP for binding to MHC
`molecules [66];
`• competition of the GA/MHC complex with MBP/MHC
`for binding to the T-cell receptor (TCR) [70];
`• activation and tolerance induction of MBP-specific T
`cells through an altered peptide ligand [67];
`• induction of GA-reactive TH2-like regulatory cells
`mediating local bystander suppression [71,72].
`
`Whereas the first two effects are supposed to take place
`only in vitro, the latter two mechanisms are most likely to
`occur also in vivo and could contribute to the anti-
`inflammatory effects of GA [68].
`More recently, a series of hints have emerged that GA may
`also exert neuroprotective effects, although the final proof of this
`hypothesis is still pending:
`
`• GA significantly reduces the number of T1 black holes in
`magnetic resonance imaging (MRI) in MS [73];
`• MR spectroscopy revealed a higher N-acetyl-aspartate
`(NAA)-creatine-ratio in GA-treated than in untreated
`patients [74];
`• GA-treated patients have been observed to exhibit lower
`brain atrophy than patients treated with placebo [75];
`• Human activated GA-reactive T lymphocytes secrete
`brain derived neurotrophic factor (BDNF), that exerts
`neuroprotective properties [76]. T cells with other
`specificity secrete BDNF as well, although at
`lower
`concentrations. In addition, GA-reactive anti-inflamma-
`tory TH2 cells have been shown to secrete higher
`amounts of BDNF than pro-inflammatory TH1 cells [77].
`• In an adoptive transfer mouse model, GA-specific T
`lymphocytes have been demonstrated to migrate to the
`CNS and express BDNF in situ [78]. In a transgenic
`experimental autoimmune encephalomyelitis (EAE)
`model, MBP-specific T cells secreting the neurotrophic
`factor nerve growth factor (NGF) induced a significantly
`milder course of EAE than MBP-specific cells not
`secreting NGF [79]. In addition, other neurotrophic
`factors have been demonstrated to be beneficial in EAE,
`both clinically and pathologically [80,81].
`
`• In another EAE model the burden of axonal damage has
`been observed to be diminished by GA treatment [82].
`• In non-inflammatory animal models for motoneuron
`diseases or trauma, beneficial effects of GA (plus complete
`Freund's adjuvant) have been demonstrated [83].
`
`11. Pharmacodynamics of mitoxantrone
`
`Mitoxantrone interacts with topoisomerase-2 and causes
`both crosslinking and single and double strand breaks by
`intercalating the DNA [84]. Apart
`from the cytotoxic
`efficacy of mitoxantrone, immunosuppressive effects and
`even antiviral and antibiotic effects have been observed
`[85,86]. More recently, immunomodulatory properties have
`been suggested, as a number of distinct
`immunological
`effects have been described [40,87,88]. Still, more research
`is warranted to illuminate the immunological effects of
`mitoxantrone in MS, as its specific mechanisms of action
`targeting the immune system still widely remain unclear.
`
`11.1. Immunosuppressive properties
`
`Mitoxantrone is a potent immunosuppressive agent target-
`ing proliferating immune cells [89–92]. It inhibits proliferation
`of macrophages, B lymphocytes and T lymphocytes [89,91].
`
`11.2. Effects on helper and suppressor T cells
`
`In an in vitro system testing an anti-sheep red blood cell
`response, mitoxantrone was observed to inhibit T helper
`activity and – conversely – to enhance T suppressor functions
`[90]. In contrast, in an in vivo mouse model, the induction of
`suppressor T cells was also abrogated by mitoxantrone [90]. T
`helper cells were indirectly inhibited by mitoxantrone-induced
`macrophages.
`
`11.3. Induction of cell death
`
`Mitoxantrone was shown to induce apoptosis of B
`lymphocytes [93] and other types of antigen-presenting cells
`[94]. Comparison of peripheral blood mononuclear cells
`(PBMC) obtained from MS patients before and immediately
`after application of mitoxantrone exhibited a decreased
`proliferation of PBMC based on necrotic cell death, predom-
`inantly in B cells [95]. Thus, there is apparently a bimodal
`mechanism of cell death induced by mitoxantrone: apoptosis at
`lower concentrations and cell lysis at higher concentrations.
`Previous pharmacokinetic studies in oncology revealed maxi-
`mum serum concentrations of mitoxantrone between 308 and
`839 ng/ml and terminal half-lifes between 38.4 and 71.5 h [33–
`36]. Thus, in the first approximately 10 days after infusion of
`12 mg/m2 body surface, maximum serum concentrations
`exceed 20 ng/ml (a putative threshold between induction of
`necrosis and apoptosis [94]), whereas the latter 80 days of a
`three-month dosage regime will have concentrations below
`20 pg/ml. Thus, mitoxantrone apparently may act via short-term
`
`Page 6 of 11
`
`YEDA EXHIBIT NO. 2012
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`
`O. Neuhaus et al. / Journal of the Neurological Sciences 259 (2007) 27–37
`
`33
`
`Fig. 5. Putative action sites of interferon-beta, glatiramer acetate, and mitoxantrone in the pathogenesis of MS. The action sites of the currently approved disease-modifying drugs are highlighted (see Fig. 4).
`(a) Antigen presentation by antigen-presenting cells is inhibited. (b) Proliferation of autoreactive T cells is blocked or tolerance is induced. (c) T-cell migration, adhesion and penetration are abrogated. (d) T-cell
`reactivation inside the central nervous system (CNS) is blocked. (e) Release of pro-inflammatory cytokines by autoreactive T cells is diminished. (f ) A cytokine shift from pro-inflammatory T-helper (TH)-1 to anti-
`inflammatory TH2 cytokines is induced. (g) A special mechanism of action of glatiramer acetate (GA) is the induction of T cells cross-reactive with both GA and CNS antigens (CNS Ag) which after reactivation
`inside the CNS exert a local bystander suppression effect through the release of TH2 cytokines. (h) Activation of macrophages or secretion of pro-inflammatory mediators are curtailed. (i) Apoptosis of autoreactive T
`cells is induced. ( j) Activation of B cells and their differentiation to plasma cells is blocked. (k) Axonal damage may be reduced. Further abbreviations: IFN, interferon; IL, interleukin; MHC, major histocompatibility
`complex; TCR, T- cell receptor. Modified from Ref. [44], with permission from Elsevier.
`
`Page 7 of 11
`
`YEDA EXHIBIT NO. 2012
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`
`34
`
`O. Neuhaus et al. / Journal of the Neurological Sciences 259 (2007) 27–37
`
`immunosuppressive effects by the induction of cell lysis leading
`to blood leukocyte reduction postinfusion and to inhibition of
`proliferation of all types of immune cells demonstrated in vitro
`[89,90,95]. In addition, a long-term immunological impact of
`mitoxantrone is considered to occur at lower concentrations by
`the induction of programmed cell death in antigen-presenting
`cells [94]. Consistent with this hypothesis, the clinical effects of
`mitoxantrone in MS have been suggested to last up to 1 year
`posttreatment [96].
`
`11.4. Effects on the cytokine network
`
`Recent ex vivo analysis of the cytokine profile of im-
`mune cells obtained from patients before and during treat-
`ment with mitoxantrone revealed a decrease of monocytes
`expressing the anti-inflammatory cytokine IL-10 and of
`T lymphocytes expressing IL-2R-β1 after 6 months of
`treatment [97].
`An overview of the immunological effects of IFN-β, GA,
`and mitoxantrone is given in Fig. 5 and Table 1.
`
`12. What do we learn from pharmacokinetics and
`pharmacodynamics?
`
`In recent years there have been major advances in
`understanding how cellular and humoral immune responses
`
`contribute to the pathogenesis of MS. In parallel, the knowledge
`on the mechanisms of action of agents aimed to modify the
`disease course has been growing continuously. As the efficacy
`of the currently available drugs is often unsatisfactory, new
`developments of therapeutic strategies against MS are highly
`warranted. Principally, this can be pursued by five ways:
`
`• the optimization of dose and route of administration of the
`currents drugs by learning from their pharmacokinetics
`(e.g. higher doses such as IFN-β1b 500 μg vs. 250 μg or
`GA 40 mg vs. 20 mg; new galenics (lipid micelles; oral or
`intranasal applications [98]);
`• the development of derivatives of the current drugs by
`learning from their pharmacodynamics (e.g. pixantrone
`instead of mitoxantrone with the aim of reducing drug-
`related cardiotoxicity [99]);
`• the evaluation of agents with other
`indications but
`reasonable for treatment of MS based on their known
`pharmacodynamics including immunomodulatory prop-
`erties (e.g. statins [100], or hormones [101,102]);
`• the design of fully new agents based on pathogenetical
`concepts (natalizumab as a selective adhesion molecule
`inhibitor is an example [103,104]);
`• the combination of available substances by learning from
`the single agents' pharmacokinetics and pharmacody-
`namics [105].
`
`Table 1
`Immunological effects of IFN-β, glatiramer acetate, and mitoxantrone [52,55,68,88]
`IFN-β
`
`Immune mechanism
`
`Glatiramer acetate
`↓ Proliferation of MBP-specific T cells
`
`No known effect
`No known effect
`
`Mitoxantrone
`
`Antigen-independent
`suppression of T-cell proliferation
`↓ Activation of B lymphocytes
`No known effect
`
`Direct and promiscuous binding to HLA-DR molecules No known effect
`↓ T-cell migration
`↓ Migration of PBL from GA-treated patients (unknown
`mechanism)
`No known effect
`
`No known effect
`
`No known effect
`No known effect
`
`No known effect
`No known effect
`
`↑ IL-10 in serum and of mRNA for TGF-β and IL-4; ↓
`mRNA for TNF-α in PBL; shift of GA-reactive T cells
`from TH1 towards TH2 during GA treatment
`Induction of anergy in MBP-specific T-cell clones
`
`↓ IL-10 in monocytes; ↓ IL-2R-β1
`in T lymphocytes during
`mitoxantrone treatment
`No known effect
`
`Proliferation of T cells
`in vitro
`B-cell activation
`Regulation of MHC
`expression
`MHC binding
`T-cell migration
`
`Adhesion molecule
`expression
`
`MMP expression
`Chemokine and chemokine
`receptor expression
`Cytokine shift in PBL
`
`APL effect on CNS-
`specific T cells
`Effects on APC
`
`Antigen-independent suppression
`of T-cell proliferation
`No known effect
`↓ IFN-γ-induced upregulation of
`MHC class II expression
`No known effect
`↓ T-cell migration
`
`↑ Soluble adhesion molecules
`(sICAM-1, sVCAM-1), ↓ surface-
`expressed adhesion molecules
`(VLA-4)
`↓ Expression of MMP-9
`↓ Secretion of chemokines, ↓
`expression of chemokine receptors
`Induction of TH2 cytokines and
`reduction of TH1 cytokines in
`PBL
`No known effect
`
`↓ IFN-γ induced FcγRI
`expression in monocytes
`
`↓ TNF-α and cathepsin-B production in a monocytic cell
`line, ↓ monocyte and dendritic cell functions
`
`↓ Proliferation of monocytes and
`macrophages; induction of
`apoptosis
`No known effect
`
`Neuroprotection
`
`Secretion of BDNF by GA-reactive T cells;
`Positive clinical and MRI effects
`neuroprotection in EAE
`on disease progression
`↑, increase; ↓, inhibitory effect; APC, antigen presenting cells; APL, altered peptide ligand; BDNF, brain-derived neurotrophic factor; CNS, central nervous
`system; EAE, experimental autoimmune encephalomyelitis; GA, glatiramer acetate; HLA, human leukocyte antigen; ICAM, intercellular adhesion molecule;
`IFN, interferon; IL, interleukin; MBP, myelin-basic protein; MHC, major histocompatibility complex; MRI, magnetic resonance imaging; MMP, matrix
`metalloproteinase; PBL, peripheral blood lymphocytes; sICAM, soluble intercellular cell adhesion molecule; sVCAM, soluble vascular cell adhesion molecule;
`TGF, transforming growth factor; TH, T helper function; TNF, tumor necrosis factor; VLA, very late antigen.
`
`Page 8 of 11
`
`YEDA EXHIBIT NO. 2012
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`
`O. Neuhaus et al. / Journal of the Neurological Sciences 259 (2007) 27–37
`
`35
`
`An overview on future developments in MS is given in Ref.
`[106]. The primary goal remains targeting of key elements of
`the immunological cascade that culminates in neural and glial
`tissue damage through disease-modifying agents.
`One step behind the horizon is the development of further
`therapeutic strategies against the demyelination as well as the
`axonal damage in MS, the latter of which is known to be
`associated with disability progression [46,47]. And further steps
`behind, i.e. neuroregenerative strategies, are still speculative.
`
`References
`
`[1] Noseworthy JH, Lucchinetti C, Rodriguez M, Weinshenker BG.
`Medical progress: multiple sclerosis. N Engl J Med 2000;343:
`938–52.
`[2] Multiple Sclerosis Therapy Consensus Group. Escalating immu-
`notherapy of multiple sclerosis. New aspects and practical applica-
`tion. J Neurol 2004;251:1329–39.
`[3] Paty DW. Interferon beta-1b. In: Hawkins CP, Wolinsky JS, editors.
`Principles of treatments in multiple sclerosis. Oxford, Woburn:
`Butterworth-Heinemann; 2000. p. 24–37.
`[4] Wolinsky JS. Glatiramer acetate. In: Hawkins CP, Wolinsky JS,
`editors. Principles of
`treatments in multiple sclerosis. Oxford,
`Woburn: Butterworth-Heinemann; 2000. p. 71–94.
`[5] Goodin DS. Therapeutic developments in multiple sclerosis. Expert
`Opin Investig Drugs 2000;9:655–70.
`[6] Hartung HP, Bar-Or A, Zoukos Y. What do we know about the
`mechanism of action of disease-modifying treatments in MS? J Neurol
`2004;251(Supplement 5):V/12–29.
`[7] The IFNB Multiple Sclerosis Study Group. Interferon beta-1b is
`effective in relapsing–remitting multiple sclerosis: I. Clinical results
`of a multicent

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