throbber
V/38_V/45_Ziemssen_Suppl_V_JON 18.10.2005 08:30 Uhr Seite 38
`
`J Neurol (2005) 252 [Suppl 5]: V/38–V/45
`DOI 10.1007/s00415-005-5007-2
`
`Tjalf Ziemssen
`
`Abstract Historically consid-
`ered to be an autoimmune de-
`myelinating disease, multiple scle-
`rosis is now recognized to be
`characterized by significant axonal
`and neuronal pathology. Address-
`ing this neurodegenerative compo-
`nent of the disease is an important
`treatment objective, since axonal
`injury is believed to underlie the
`accumulation of disability and dis-
`ease progression. The precise rela-
`tionship between the inflammatory
`and neurodegenerative compo-
`nents in multiple sclerosis remains
`
`T. Ziemssen (쾷)
`Neurological University Clinic
`Technical University of Dresden
`Medical Faculty “Carl Gustav Carus”
`Fetscherstr. 74
`01307 Dresden, Germany
`Tel.: + 49-351/458-6614
`Fax: +49-351/458-5873
`E-Mail: ziemssen@web.de
`
`Introduction
`
`Modulating processes within the central
`nervous system is central to therapeutic
`control of multiple sclerosis
`
`poorly elucidated, although neu-
`rodegeneration appears to be at
`least partially independent from
`neuroinflammation. The mecha-
`nisms underlying axonal injury ap-
`pear complex and are likely to be
`multifactorial. Specific treatment
`strategies need to be developed
`that act within the central nervous
`system to prevent neurodegenera-
`tion and need to be provided from
`the earliest stages of disease. It is
`likely that immunomodulatory
`treatments acting purely in the pe-
`riphery will provide only indirect
`and not direct neuroprotection. A
`promising approach is to enhance
`neuroprotective autoimmunity in-
`side the brain, believed to be medi-
`ated, at least in part, by the release
`of neurotrophic factors within the
`nervous system from infiltrating
`immune cells. Such a beneficial
`process would be inhibited by a
`non-selective immunosuppressive
`
`strategy. In summary, treatments of
`multiple sclerosis should take into
`account the heterogeneous patho-
`physiology of the disease. The
`pathogenic process in the central
`nervous system itself should be the
`major focus in multiple sclerosis
`therapy in order to protect against
`demyelination and axonal loss and
`to promote remyelination and re-
`generation directly in the target tis-
`sue, independently of peripheral
`immune status. In conclusion, se-
`lective treatment strategies aimed
`at preventing axonal injury within
`the central nervous system are re-
`quired to complement existing, pe-
`ripherally acting treatments target-
`ing the immune system.
`
`Key words multiple sclerosis ·
`neurodegeneration ·
`neuroprotection · inflammation ·
`treatment
`
`[42]. In the central nervous system, these cells infiltrate
`discrete areas of tissue, where they cause damage to
`oligodendrocytes (e.g., demyelination) and neurons
`(e.g., axonal transection), resulting in the formation of a
`sclerotic plaque. The interactions between these differ-
`ent immune cell populations itself and between immune
`cells, neurons and glia are highly complex. However, a
`better understanding of these interactions in the central
`nervous system itself is necessary for the development
`of more rational treatment strategies which can modu-
`late these interactions in a specific way and thereby pre-
`vent disease activity. Of particular interest is the poten-
`
`Multiple sclerosis is a complex autoimmune disease in-
`volving disturbances to the peripheral immune system,
`although the detailed pathogenic cascade remains un-
`known. Many different immune cells are believed to be
`involved in the disease process, including myelin-reac-
`tive CD4 + T cells which carry the immune response to
`the nervous system,CD25 + regulatory T cells,which can
`control autoreactive CD4 + cells, myelin-reactive B cells,
`CD8 + killer cells, macrophages and brain microglia
`
`JON 5007
`
`Page 1 of 8
`
`YEDA EXHIBIT NO. 2084
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`■
`■
`

`
`V/38_V/45_Ziemssen_Suppl_V_JON 18.10.2005 08:30 Uhr Seite 39
`
`V/39
`
`tial to prevent the neurodegenerative changes in the ner-
`vous system that are thought to be responsible for the
`accumulation of permanent neurological disability.
`
`Table 1 Comparison between outcomes of treatment in clinical trials in multiple
`sclerosis (MS) and
`in animal models of experimental autoimmune en-
`cephalomyelitis (EAE)
`
`Treatment targets in the periphery
`
`To date, treatments for multiple sclerosis have been de-
`veloped with the intention of intervening at the level of
`certain autoimmune responses in the periphery. This
`approach is hampered by limited knowledge of the
`pathogenic cascade in human multiple sclerosis, which
`compromises the development of rationally designed
`immune treatments. Peripherally acting drugs targeting
`the immune system in multiple sclerosis include
`immunosuppressants, such as mitoxantrone, which pro-
`duce a non-specific inhibition of immune cell function,
`or immunomodulators such as the beta-interferons,
`glatiramer acetate and natalizumab which target more
`or less well-defined processes involved in the assumed
`pathogenic autoimmune response. Unfortunately, since
`there is no specific immunological abnormality in pa-
`tients with multiple sclerosis,it is not possible to develop
`treatments that selectively target these processes. For
`this reason, a major limitation of such drugs is that the
`immune processes targeted are more or less non-spe-
`cific, leading to unwanted effects on immune function
`such as immunosuppression.
`Because of the limited pathogenetic data in multiple
`sclerosis, nearly all studies of agents targeting the dis-
`ease have been performed not in the human disease but
`in animal models. In particular, the experimental au-
`toimmune encephalomyelitis (EAE) model has been
`used widely to evaluate immunomodulatory treatments
`for multiple sclerosis as well as to explore the patho-
`physiology of the disease. This model involves the gen-
`eration of an autoimmune response in the immunolog-
`ical periphery by immunizing animals with myelin
`proteins such as myelin basic protein (MBP). In this
`quite simple model, the disease can also be transferred
`from affected animals to healthy recipients by adoptive
`transfer of myelin-reactive T cells. The animals develop
`a clinical and pathological pattern which is quite differ-
`ent from human multiple sclerosis (spinal cord lesions
`in EAE) although several histopathological hallmarks of
`multiple sclerosis, including focal inflammatory lesions
`in the nervous system can be found.
`As more and more scientific data from animal exper-
`iments accumulate, it is important to keep in mind that
`EAE is not human multiple sclerosis. In fact, each EAE
`experiment only represents a small part of the still un-
`known pathogenetic cascade of autoimmune demyeli-
`nation.It is perhaps for this reason that divergent results
`have been observed for a number of treatments assessed
`in both the EAE model and in clinical trials in multiple
`sclerosis [39] (Table 1). The most striking example is
`
`Therapy
`IFN-γ, systemic
`Anti-TNF-α, systemic
`IL-4 transduced T cells
`TNF-α transduced T cells
`Glatiramer acetate
`Beta-interferons
`Anti-α4 integrin antibodies
`
`MS
`
`Worsens
`Worsens
`Not tested
`Not tested
`Improves
`Improves
`Improves
`
`EAE
`
`Cures
`Cures
`Cures
`Worsens
`Cures
`Improves
`Cures
`
`perhaps that of lenercept, a recombinant TNF receptor
`p55 immunoglobulin fusion protein. In the EAE model,
`such TNF receptor fusion proteins ameliorate clinical
`symptoms [22, 23]. However, a double-blind, random-
`ized placebo-controlled trial in relapsing-remitting
`multiple sclerosis found that treatment with lenercept
`was associated with a higher proportion of patients ex-
`periencing relapses, a shorter time to first relapse and
`more severe neurological deficits [36]. Other examples
`include glatiramer acetate (GA) and natalizumab which
`provide complete abrogation of the disease process in
`the EAE model in contrast to their limited clinical
`benefit in MS patients.For these reasons,all information
`obtained from the EAE model should be interpreted
`with caution and within the context of what is known of
`the overall pathophysiology of multiple sclerosis.
`Developing treatments that target the immune re-
`sponse within the central nervous system may be more
`promising than peripherally acting treatments. Brain
`targets have the advantage over peripheral targets in
`that they directly address the core disease process and
`will have less non-selective effects on systemic immune
`function. Interesting potential targets include activation
`of microglia, antibody-mediated injury to myelin and
`axons and B cell interactions with oligodendrocyte pre-
`cursor cells [29]. A drug that appears to down-regulate
`microglial activation is minocycline [37] and this agent
`also slows the appearance of EAE and attenuates its
`severity [6, 31]. Preliminary clinical data in multiple
`sclerosis indicate that minocycline may reduce lesion
`activity [26] and these findings merit confirmation in a
`randomized controlled trial.
`
`The two faces of multiple sclerosis: inflammation
`and neurodegeneration
`
`It is now clear that the pathophysiology of multiple scle-
`rosis cannot be adequately explained uniquely by acute,
`focal inflammatory attack inside the central nervous
`system [21]. Our understanding also needs to take into
`
`Page 2 of 8
`
`YEDA EXHIBIT NO. 2084
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`V/38_V/45_Ziemssen_Suppl_V_JON 18.10.2005 08:30 Uhr Seite 40
`
`V/40
`
`account the neurodegenerative processes of axonal loss,
`which may occur to some extent independently from in-
`flammation, and certainly arise very early in the disease
`process.
`These two facets of the pathology of multiple sclero-
`sis provide an elegant model to explain the heteroge-
`neous clinical presentation and course of the disease
`[12].If it has been accepted for many years that the acute
`relapses observed in relapsing-remitting multiple scle-
`rosis reflect flairs of inflammatory activity, it is now
`thought that permanent clinical disability is mainly
`determined by the extent of axonal loss. Once axonal
`loss has reached a certain critical threshold, irreversible
`neurological deficits emerge. During the course of the
`disease, inflammatory events become rarer, whereas
`neurodegeneration continues or even becomes more
`prominent (Fig. 1). This picture would account for the
`transition from a relapsing-remitting to a secondary
`progressive form of the disease. However, if inflamma-
`tion and neurodegeneration are to some extent inde-
`pendent, the relative importance of the two processes in
`individual patients may account for the different pat-
`terns of clinical presentation seen between patients and
`explain the imperfect correlation between exacerba-
`tions, inflammatory activity in MRI and accumulation
`of disability.
`The biochemical events underlying the inflammatory
`
`and neurodegenerative phases of the disease are not
`known in detail and may be quite different [33]. Inflam-
`mation involves activation of T and B cells in the pe-
`riphery, crossing the blood brain barrier and homing
`to the lesion site. In the lesion, T cells are reactivated
`by myelin antigens, release cytokines that attract
`macrophages and activate microglia which start to de-
`stroy the myelin sheath. Anti-myelin antibodies bind
`complement, attract macrophages and stimulate op-
`sonization of myelin [42]. Demyelination leads to re-
`versible and to some extent irreversible impairment of
`function of the axon whose conduction properties are
`deteriorated, thus accounting for the clinical symptoms
`associated with relapses.
`Neurodegeneration is likely to be a complex process
`[11, 18], especially when it takes place in inflammatory
`disorders like multiple sclerosis. To a significant extent,
`axonal loss seems to be a major consequence of de-
`myelination and inflammation, for example by binding
`of CD8 + T cells to exposed axons and secretion of toxic
`factors. However, other mechanisms not directly related
`to demyelination and inflammation are also likely to be
`important. An example is excitotoxicity: glutamic acid
`can bind to excitatory amino acid receptors on the cell
`bodies, dendrites or axon terminals of neurons and ini-
`tiate a process of necrotic cell death [34]. Theoretically,
`there are several possible relationships between inflam-
`
`Fig. 1 Different clinical phenotypes of multiple sclerosis and the underlying pathology. Adapted from [13]
`
`Page 3 of 8
`
`YEDA EXHIBIT NO. 2084
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`V/38_V/45_Ziemssen_Suppl_V_JON 18.10.2005 08:30 Uhr Seite 41
`
`mation and neurodegeneration in multiple sclerosis
`(Fig. 2). Three exclusive hypotheses can be envisaged:
`(a) that neurodegeneration is entirely secondary to in-
`flammation, (b) that inflammation is entirely secondary
`to neurodegeneration or (c) that inflammation and neu-
`rodegeneration are entirely independent. On the other
`hand, non-exclusive hypotheses where neurodegenera-
`tion is partially dependent and partially independent of
`inflammation or vice versa can also be put forward, and
`these seem intuitively more likely.
`There are a number of clinical arguments in favor of
`some independence between the inflammatory and
`neurodegenerative processes. For example, in a clinical
`trial of alatuzemab (Campath-1H), a monoclonal anti-
`body directed against CD52 which leads to T cell deple-
`tion, in secondary progressive multiple sclerosis, a grad-
`ual extinction of exacerbations and lesion activity
`visible on MRI was demonstrated [12]. However, dis-
`ability continued to progress in about half the patients
`in whom progressive brain atrophy and axonal degener-
`ation could be observed using MRI and magnetic reso-
`nance spectroscopy (MRS). The investigators con-
`cluded, first that inflammation and demyelination were
`responsible for relapses of multiple sclerosis and could
`be prevented by alatuzemab treatment and, second that
`continuing axonal degeneration accounted for the pro-
`gressive phase of disability. Even though axonal injury
`may have been conditioned by prior inflammation, this
`process can continue despite complete suppression of
`inflammatory activity or it is for example the case in
`bone narrow transplanted MS patient.
`There is also neuropathological evidence for a disso-
`ciation between inflammatory demyelination and ax-
`onal injury from a series of 42 biopsy samples obtained
`from patients with multiple sclerosis [3]. Acute axonal
`injury was visualized by amyloid precursor protein
`
`Fig. 2 Possible hypotheses for the causal relationship between inflammation and
`neurodegeneration in the pathogenesis of multiple sclerosis
`
`V/41
`
`(APP) staining. There was no relationship between the
`expression of APP or axonal density and the extent of
`demyelination, with axonal injury being observed even
`in lesions that were successfully remyelinating. Simi-
`larly, there was no association between the extent of ax-
`onal injury and markers of acute inflammation such as
`TNF-α or inducible NO synthase. However, axonal in-
`jury was correlated to some extent with the extent of in-
`filtration by CD8 + T lymphocytes and by macrophages.
`A dissociation between neurodegeneration and inflam-
`matory demyelination is also observed in lesions within
`the cortex. These lesions are characterized by a signifi-
`cant degree of axonal transection and apoptosis of neu-
`ronal cell bodies. However, the extent of infiltration by T
`lymphocytes and macrophages and the expression of in-
`flammatory markers is low [4, 30].
`MRI studies in very early disease also suggest that
`inflammation and neuronal injury are not strictly re-
`lated. A study of 31 subjects presenting with a clinically
`isolated syndrome evaluated inflammatory lesion activ-
`ity with classical T2- and T1-weighted images after
`gadolinium enhancement and measured a surrogate
`marker of axonal injury,the size of the N-acetylaspartate
`peak (NAA) determined in the whole brain [14].In these
`patients the mean size of the NAA peak was some 20 %
`lower than that observed in matched controls. No corre-
`lation was observed between the size of the NAA peak
`and lesion volume on either T1 or T2 images. The inves-
`tigators concluded that significant axonal injury occurs
`early in the disease and that this is only indirectly linked
`to inflammatory activity.
`Studies such as these suggest that treatment strate-
`gies for multiple sclerosis need to address both the in-
`flammatory and neurodegenerative components of the
`disease, and that anti-inflammatory therapies may only
`be able to control the inflammation-related neurode-
`generative process adequately [11].
`
`The pathogenic process takes place in the brain
`
`Degeneration of oligodendrocytes, neurons and axons
`are the key pathological features of multiple sclerosis
`which are responsible for the irreversible neurological
`handicap that accumulates in the course of disease. For
`this reason, rational therapies need to target these core
`disease processes in a more specific and a more effective
`way than it is possible with only peripherally acting
`treatments [11]. Nevertheless, the processes underlying
`axonal damage in multiple sclerosis are extremely com-
`plex and certainly multifactorial [16, 18], which has
`hampered development of neuroprotective treatments
`in the past. However, recent developments have identi-
`fied several promising avenues of research for develop-
`ing such drugs.
`Treatments targeting the brain are also more appro-
`
`Page 4 of 8
`
`YEDA EXHIBIT NO. 2084
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`V/38_V/45_Ziemssen_Suppl_V_JON 18.10.2005 08:30 Uhr Seite 42
`
`V/42
`
`priate with respect to the phase of the disease at which
`treatment is initiated.Although the initial trigger of dis-
`ease in multiple sclerosis is likely to occur in the periph-
`eral immune system, by the time the disease is diag-
`nosed, it is already a localized disease in the central
`nervous system. The initial symptoms correspond to re-
`activation of autoimmune cells, their entry into the
`brain and the triggering of a local demyelinating event.
`However, at this stage, the disease process is already
`characterized by primary and secondary axonal and
`neuronal degeneration and the local environment has
`become to some extent inhospitable for repair and re-
`generation of axons and myelin. Due to the built-in re-
`dundancy of the nervous system, clinical manifestations
`are only apparent once a critical degree of neuronal
`damage has been reached. That is why the earliest stages
`of the disease are as a consequence clinically silent. This
`can be visualized by the reduced brain volume and NAA
`compartments observed by imaging studies performed
`at the first clinical presentation of disease.
`Therapeutic strategies need to take into account this
`burden of tissue damage inside the brain that is already
`present when treatment decisions are first being made.
`Although prevention of future flairs of inflammatory at-
`tack on the nervous system are obviously required, it is
`also important to address existing damage and vulnera-
`bility. For example, strategies could be implemented to
`promote oligodendrocyte survival and repair,to prevent
`further axonal degeneration and neuronal dysfunction
`and to promote axonal and neuronal regeneration. De-
`livery of growth factors to the areas of tissue damage
`would be an interesting possibility to achieve these ob-
`jectives.
`
`The neuroprotective side of neuroinflammation
`
`A promising avenue of research for drug development in
`multiple sclerosis is the concept of protective autoim-
`munity. Although the traditional view has been that
`autoimmune responses are exclusively deleterious,espe-
`cially inside the brain, it now appears clear that autoim-
`mune cells can, under certain conditions, promote
`neural repair. This was first demonstrated in an animal
`model of traumatic optic nerve injury [27] in rats. If the
`rats were injected with activated anti-MBP T cells, they
`retained three times as many retinal ganglion cells with
`functionally intact axons than did rats injected with ac-
`tivated T cells specific for other antigens. Since then, this
`concept has been extended to many other experimental
`paradigms and appears to be a universal principle for
`both immune and non-immune degenerative diseases of
`the central nervous system [32]. For example, in Parkin-
`son’s disease, activated microglia is present in the sub-
`stantia nigra. On the one hand, these cells may con-
`tribute to tissue damage by, for example, the release of
`
`reactive oxygen species or pro-inflammatory cytokines,
`but, on the other, they may protect neurons by the re-
`lease of neurotrophic factors or by removal of excito-
`toxic glutamic acid from the extracellular milieu [35].
`Regulation of activated microglia in Parkinson’s disease
`as a potential target for new neuroprotective therapies is
`an exciting new prospect which is receiving much inter-
`est.
`A possible explanation of this neuroprotective role of
`T cells may be the release of neurotrophic factors from
`immune cells that promote neuronal repair or protect
`against injury [20] (Fig. 3). In particular, brain-derived
`neurotrophic factor (BDNF) has been shown to be pro-
`duced and secreted by a variety of immunocompetent
`cells [2, 5, 19]. BDNF is a potent neurotrophic factor
`which can rescue neurons following axonal transection
`[17]. In autopsy material from multiple sclerosis pa-
`tients, BDNF is present in T cells and macrophages infil-
`trating the lesions [33]. BDNF expression is higher in
`immune cells from active lesions compared to inactive
`ones, consistent with the observation in vitro that ex-
`pression is up-regulated following activation of T cells
`[19]. In addition, the BDNF receptor trkB appears to be
`up-regulated in damaged neurons in the immediate
`vicinity of active lesions [33]. The cellular machinery is
`therefore in place for BDNF-mediated neuroprotective
`immunity in multiple sclerosis lesions.
`The potential neuroprotective effects of growth fac-
`tors have been evaluated extensively in the EAE model
`of human multiple sclerosis. These studies have con-
`cerned nerve growth factor (NGF), leukemia inhibitory
`factor (LIF), insulin-like growth factor-1 (IGF-1) and
`glial growth factor-2 (GGF-2), but not BDNF. In the ma-
`jority of studies, administration of growth factors either
`delayed the onset of disease or reduced the severity of
`the neurological deficit (Table 2).
`
`Fig. 3 Potential neuroprotective effects of neurotrophins released from immune
`cells in inflammatory diseases of the nervous system. Reproduced from [20] with
`permission
`
`Page 5 of 8
`
`YEDA EXHIBIT NO. 2084
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`V/38_V/45_Ziemssen_Suppl_V_JON 18.10.2005 08:30 Uhr Seite 43
`
`Table 2 Studies of growth factors in murine models
`of experimental autoimmune encephalitis (EAE)
`
`V/43
`
`Growth factor
`administration
`
`NGF (ICV pump)
`NGF (transduced T cells)
`NGF (IP injection)
`LIF (IP injection)
`IGF-1 (IV injection)
`
`IGF-1/ILGFBP3
`(IP injection)
`IGF-1 (SC injection)
`GGF-2 (SC injection)
`
`EAE model
`
`Clinical outcome
`
`Reference
`
`Delay of EAE onset
`Active MOG EAE
`Suppression of EAE
`Passive MPB EAE Lewis rat
`Delay of EAE onset
`Active and passive MBP EAE
`앗 EAE severity
`TCR-tg mice
`Passive MBP EAE Lewis rat 앗 Mean cumulative
`clinical score
`Delay of EAE onset
`앖 Severity of disease
`앗 Severity of acute EAE
`Delay of EAE onset
`
`Passive MBP EAE PLxSJL
`mice
`Passive MBP EAE SJL mice
`Passive MBP EAE SJL mice
`
`Villoslada [38]
`Flügel [15]
`Arredondo [1]
`Butzkueven [7]
`Liu [24]
`
`Lovett-Racke [25]
`Cannella [9]
`Cannella [8]
`
`GGF-2 glial growth factor-2; IGF-1 insulin-like growth factor-1; LIF leukemia inhibitory factor; MBP myelin basic
`protein; MOG myelin oligodendrocyte glycoprotein; NGF nerve growth factor; TCR-tg T cell receptor transgenic
`
`An interesting study with NGF [15] evaluated the ef-
`fect of transfecting MBP-reactive CD4 + T cells with a
`vector driving NGF expression and synthesis on the
`ability of these T cells to induce EAE when injected into
`rats. These transfected cells produced only a very mild
`EAE syndrome in recipient animals and when co-in-
`jected with non-transfected MBP-reactive T cells atten-
`uated significantly the symptoms of EAE induced by the
`latter (Fig. 4). This suppression of EAE by the trans-
`fected cells was associated with a general reduction of
`immune cells infiltrating the nervous system, notably
`macrophages. This study clearly demonstrates the ben-
`eficial effect of neurotrophins released from immune
`cells on clinical expression of disease.
`The ability of T cells to secrete BDNF when activated
`may be harnessed by GA to endow this drug with a neu-
`roprotective effect in multiple sclerosis that comple-
`
`ments its longer-established anti-inflammatory activity.
`If GA-reactive T cells are recovered from the circulation
`and activated in vitro with GA in the presence of anti-
`gen-presenting cells, secretion of BDNF is stimulated
`[10, 40]. Such BDNF-secreting, GA-reactive T cells can
`be recovered from the circulation of most patients
`treated with GA. These studies have important implica-
`tions for the mechanism of action of GA, supporting the
`hypothesis that GA-reactive T cells activated in the pe-
`riphery may enter the brain and release BDNF in the site
`of active lesions besides their anti-inflammatory factors,
`thus contributing to repair, regeneration and protection
`of axons and neurons (Fig. 5).
`
`Fig. 4 Clinical severity of experimental autoimmune encephalomyelitis in rats in-
`: control T cells; ■
`: T cells
`jected with myelin basic protein-reactive T lymphocytes. ●
`: co-injection of control and transfected
`transfected with nerve growth factor; ▲
`T cells. Adapted from [15] with permission
`
`Fig. 5 Increased production of brain-derived growth factor from GA-reactive T
`cells in the presence of antigen-presenting cells in vitro. BDNF secretion of the TH2-
`like GA-specific T cell line BK-M6-COP-7 of a GA-treated patient, measured by BDNF
`ELISA (top), by RT-PCR (middle) and by intracellular BDNF staining (bottom). Re-
`produced from [41] with permission
`
`Page 6 of 8
`
`YEDA EXHIBIT NO. 2084
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`V/38_V/45_Ziemssen_Suppl_V_JON 18.10.2005 08:30 Uhr Seite 44
`
`V/44
`
`Conclusions
`
`Multiple sclerosis is no longer considered to be simply
`an autoimmune demyelinating disease. In addition to
`neuroinflammation and demyelination, axonal injury
`and neuronal loss are now also recognized to be hall-
`marks of the disease, and these seem to be the major
`contributors to the accumulation of disability and dis-
`ease progression.
`The mechanism underlying axonal pathology has
`not been clarified so far, but does not appear to be sim-
`ple. The relationship between axonal damage and other
`components of the pathological process such as de-
`myelination, inflammation and remyelination are still
`under intense investigation. Especially in the later
`phases of diseases, most irreversible damage does not
`seem to result from active immune attack resulting in
`new lesions; consequently, reduction of inflammatory
`disease activity produces only marginal effects on ner-
`vous tissue degeneration in these patients. In addition,
`suppression of immune-mediated inflammation does
`not prevent progressive loss of neurological function in
`patients with primary progressive multiple sclerosis.
`Animal models, such as experimental autoimmune
`encephalomyelitis, have been used widely to decipher
`the pathophysiology of multiple sclerosis and predict re-
`sponses to therapy. However, numerous pieces of data
`which are incompatible with this simple autoimmune
`hypothesis have been accumulated over past decades.
`This would suggest that other immunological mecha-
`nisms apart from T cell activation, involving, for ex-
`ample, microglia, or a neurodegenerative component
`within the target tissue,might contribute significantly to
`the initiation, propagation and evolution of disease.
`
`The critical question at the moment is the relation-
`ship between inflammation and axonal injury. If inflam-
`mation is the only factor leading to neurodegeneration
`in multiple sclerosis, immunosuppressants should be
`able to cure multiple sclerosis, which of course is not the
`case with the currently available drugs. On the other
`hand, if inflammation and axonal injury are at least
`partly independent pathologies, axonal injury has to be
`addressed therapeutically in a separate and specific way.
`Treatment of the peripheral immune system (i.e., im-
`munosuppression) will probably only allow modest in-
`direct effects on the inflammatory component of the
`disease process in the brain, which may be of relevance
`in the early stages of the disease. However, a new poten-
`tially beneficial role for neuroinflammation has been
`proposed in some stages of disease, based on data show-
`ing that immune cells in multiple sclerosis lesions can
`demonstrate neuroprotective activity. Such a beneficial
`process would be inhibited by an non-selective im-
`munosuppressive strategy.
`In summary, treatments of multiple sclerosis should
`take into account the heterogeneous pathophysiology of
`the disease. The pathogenic process in the central ner-
`vous system itself should be the major focus in multiple
`sclerosis therapy in order to protect against demyelina-
`tion and axonal loss and to promote remyelination and
`regeneration directly in the target tissue, independently
`of peripheral immune status. In particular, in order to
`produce significant long-term effects in the treatment of
`MS patients, the modulation of processes within the
`central nervous system will be central to therapeutic
`control of the disease – including neuroinflammation
`and neurodegeneration which both take place inside the
`central nervous system.
`
`References
`
`1. Arredondo LR, Deng C, Ratts RB, et al.
`(2001) Role of nerve growth factor in
`experimental autoimmune en-
`cephalomyelitis. Eur J Immunol 31:
`625–633
`2. Besser M, Wank R (1999) Cutting edge:
`clonally restricted production of the
`neurotrophins brain-derived neuro-
`trophic factor and neurotrophin-3
`mRNA by human immune cells and
`Th1/Th2-polarized expression of their
`receptors. J Immunol 162:6303–6306
`3. Bitsch A, Schuchardt J, Bunkowski S,
`et al. (2000) Acute axonal injury in
`multiple sclerosis. Correlation with de-
`myelination and inflammation. Brain
`123:1174–1183
`4. Bo L,Vedeler CA, Nyland H, et al.
`(2003) Intracortical multiple sclerosis
`lesions are not associated with in-
`creased lymphocyte infiltration. Mult
`Scler 9:323–331
`
`5. Braun A, Lommatzsch M, Mannsfeldt
`A, et al. (1999) Cellular sources of en-
`hanced brain-derived neurotrophic
`factor production in a mouse model of
`allergic inflammation. Am J Respir Cell
`Mol Biol 21:537–546
`6. Brundula V, Rewcastle NB, Metz LM,
`et al. (2002) Targeting leukocyte MMPs
`and transmigration: minocycline as a
`potential therapy for multiple sclero-
`sis. Brain 125:1297–1308
`7. Butzkueven H, Zhang JG, Soilu-Hanni-
`nen M, et al. (2002) LIF receptor signal-
`ing limits immune-mediated demyeli-
`nation by enhancing oligodendrocyte
`survival. Nat Med 8:613–619
`8. Cannella B, Hoban CJ, Gao YL, et al.
`(1998) The neuregulin, glial growth
`factor 2, diminishes autoimmune de-
`myelination and enhances remyelina-
`tion in a chronic relapsing model for
`multiple sclerosis. Proc Nat Acad Sci
`USA 95:10100–10105
`
`9. Cannella B, Pitt D, Capello E, Raine CS
`(2000) Insulin-like growth factor-1
`fails to enhance central nervous sys-
`tem myelin repair during autoimmune
`demyelination. Am J Pathol 157:
`933–943
`10. Chen M,Valenzuela RM, Dhib-Jalbut S
`(2003) Glatiramer acetate-reactive T
`cells produce brain-derived neuro-
`trophic factor. J Neurol Sci 215:37–44
`11. Chitnis T, Imitola J, Khoury SJ (2005)
`Therapeutic strategies to prevent neu-
`rodegeneration and promote regenera-
`tion in multiple sclerosis. Curr Drug
`Targets Immune Endocr Metabol Dis-
`ord 5:11–26
`12. Coles AJ, Wing MG, Molyneux P, et al.
`(1999) Monoclonal antibody treatment
`exposes three mechanisms underlying
`the clinical course of multiple sclero-
`sis. Ann Neurol 46:296–304
`
`Page 7 of 8
`
`YEDA EXHIBIT NO. 2084
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`V/38_V/45_Ziemssen_Suppl_V_JON 18.10.2005 08:30 Uhr Seite 45
`
`13. Compston A, Coles A (2002) Multiple
`sclerosis. Lancet 359:1221–1231
`14. Filippi M, Bozzali M, Rovaris M, et al.
`(2003) Evidence for widespread axonal
`damage at the earliest clinical stage of
`multiple sclerosis. Brain 126:433–437
`15. Flügel A, Matsumuro K, Neumann H,
`et al. (2001) Anti-inflammatory activity
`of nerve growth factor in experimental
`autoimmune encephalomyelitis: inhi-
`bition of monocyte transendothelial
`migration. Eur J Immunol 31:11–22
`16. Giuliani F, Yong VW (2003) Immune-
`mediated neurodegeneration and neu-
`roprotection in MS. Int MS J 10:
`122–130
`17. Gravel C, Gotz R, Lorrain A, Sendtner
`M (1997) Adenoviral gene transfer of
`ciliary neurotrophic factor and brain-
`derived neurotrophic factor leads to
`long-term survival of axotomized mo-
`tor neurons. Nat Med 3:765–

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