throbber

`olsELSEVIER
`
`TRENDSin Pharmacological Sciences Vol.24 No.3 March 2003
`
`131
`
`Immunomodulation tn multiple
`sclerosis: from immunosuppression
`to neuroprotection
`Oliver Neuhaus’, Juan J. Archelos” and Hans-Peter Hartung’
`
`'Department of Neurology, Heinrich-Heine-Universitat, Moorenstrasse 5, 40225 Diisseldorf, Germany
`2Multiple Sclerosis Research Group, Department of Neurology, Karl-Franzens-Universitat, Auenbruggerplatz 22, 8036 Graz, Austria
`
`Multiple sclerosis (MS) is the most common disabling
`neurological disease of young adulthood. Following
`advances in the understanding of the immunological
`mechanisms that underlie the pathogenesis of MS, a
`growing arsenal of immunomodulatory agentsis avail-
`able. Two classes of immunomodulators are approved
`for long-term treatment of MS,the efficacy of several
`promising new conceptsis beingtestedin clinical trials
`and classical immunosuppressive agents used in MS
`treatment have been shownto exert specific, immuno-
`modulatory effects. Furthermore,
`two recent obser-
`vations have changed our basic understanding of the
`pathogenesis of MS. First, immunecells in MS lesions
`have neuroprotective activity, which indicates a bene-
`ficial role of neuroinflammation. Second, there is evi-
`dence that axonal
`loss,
`rather than demyelination,
`underlies the progression of MS and,hence, constitutes
`a therapeutic target.
`
`have been approved for the treatment of MS [8-10].
`Immunomodulators, which do not cause general suppres-
`sion of the immunesystem, shift immuneresponses from
`pro-inflammatory autoimmune conditions [mediated by
`T helper 1 (Th1) cytokines that are released by auto-
`reactive T cells] towards more beneficial anti-inflamma-
`tory circumstances (mediated through Th2 cytokines that
`are secreted by regulatory T cells). Both IFN-8 and GA
`have been provento be partially effective in clinical trials
`[1,11—14]. In the search for moreefficacious agents, many
`new drugs are under investigation in preclinical and
`clinical trials, but several promising approaches have
`failed [15]. In parallel there have been advances in
`understanding the underlying pathogenesis of the disease
`as well as modesof action of the different agents.
`Here, we summarize current concepts about the mech-
`anismsofaction of therapies already approved for MS and
`the most promising future candidates.
`
`‘The surest way to lose a reputation in neurology is to
`advocate a treatment for multiple sclerosis.’ (H. Houston
`Merritt)
`
`Multiple sclerosis (MS) is the most common inflamma-
`tory disorder of the CNS and the leading cause of
`neurological disability in young adults [1]. Many immune
`abnormalities have been described in MS, which indicates
`that the immune system plays a central role in its
`pathogenesis [2—4]. Although immuneresponsescontrib-
`ute to the formation and maintenance of MSlesions [5],
`neuroinflammation might have neuroprotective effects
`[6,7]. This crucial role of the immune system in disease
`pathogenesis has important therapeutic implications. For
`a long while corticosteroids were the only proven therapy
`for MS. However, these only shorten an acute attack and
`effective, long-term drug treatment was not available.
`Although several immunosuppressive agents (i.e. inhibi-
`tors of crucial components of the immune system that
`cause generalized immune dysfunction) were used off-
`label, the adverse systemic effects, such as increased risk
`of cancer and infection, limited the potential benefits in
`MS. More recently,
`two classes of immunomodulatory
`agents, interferon B (IFN-8) and glatiramer acetate (GA),
`
`Disease-relevant immune processes
`Sincethefirst description of MS in 1835 by J. Cruveilhier
`as ‘sclérose en taches, en iles par masses disséminées’[16],
`the concepts of its pathogenesis have been adapted
`continuously [1,2,4,17]. Although unproven, the current
`consensus is that MS pathogenesis comprises an initial
`inflammatory phase, which fulfils the criteria for an
`autoimmunedisease [18], followed by a phaseof selective
`demyelination andlast, a neurodegenerative phase [4,17].
`Subjects with genetically determined susceptibility to MS
`[19] harbor T cells that react with CNS autoantigens.
`Although these can remain dormant for decades, at some
`point they are activated in the periphery, probably by
`molecular mimicry (i.e. recognition ofepitopes that are com-
`mon to autoantigens and microbial antigens as exogenous
`triggers [20,21]). This enables them to migrate through the
`blood—brain barrier to the brain and spinal cord. Reacti-
`vated in the CNS,these T cells of either CD4* helper or
`CD8* cytotoxic phenotype [22] release pro-inflammatory
`Th1 cytokines and orchestrate the destruction of the
`myelin sheath by various types of immunecells. Destruc-
`tion follows the first two of four pathological patterns [5]:
`(1) T-cell- and macrophage-mediated demyelination; (2)
`antibody-mediated demyelination that involves comple-
`Corresponding author: Oliver Neuhaus(oliver.neuhaus@uni-duesseldorf.de).
`mentactivation [3,23]; (3) distal oligodendrogliopathy and
`Merck 2018
`http://tips.trends.com 0165-6147/03/$ - see front matter © 2003 Elsevier ScienceLtd. All rights reserved. doi:10.1016/S0165-6147(03)00028-2
`TWiv Merck
`IPR2023-00050
`
`

`

`132
`
`TRENDSin Pharmacological Sciences Vol.24 No.3 March 2003
`
`Periphery
`Activation
`
`Macrophage
`
`Migration,
`adhesion,
`penetration
`
`Central nervous system
`Reactivation
`
`Inflammation
`
`Microglia
`
`Macrophage
`
`Demyelination
`patterns (1)—(4)
`Neuron Oligodendrocyte
`
`Axonal damage
`
`
`
`
`
`Antigen-
`
`presenting
`Adhesiesion
`cell
`TNF-o.,
`x
`
`MHC-II Jal molecules,|MHC-IIJal < Qo, NO
`
`
`
`Microbial Ag © proteases,|CNS Ag O Uf Activated S (1) T-cell and
`
`
`chemokines
`IFN-y ,
`T cell
`SS
`macrophage
`
`TCR
`TCR
`7
`attack
`1
`;
`’
`TNF=c.
`i
`vt
`
`
`Autoreactive oy(Oy)I Le >(1) ( Antibodyattack
`3
`\
`Complement
`4
`apoptosis
`2
`\
`~
`(4) Primary OG
`7
`Thi
`a
`IFN-y, ‘Activated
`Plasma
`F
`degeneration
`Axonalswelling,
`2
`4 Beell
`IL-2
`cell SQ
`lobulation,
`5
`<x!
`disconnection
`}
`©--Oz
`s
`= (2)
`a]
`8
`=f
`eS
`
`no Demyelinating_—Target cellsEffector cells
`antibodies
`
`--»>
`
`
`
`TRENDSin Pharmacological Sciences
`
`Fig. 1. Pro-inflammatoryTcells in the periphery are activated whenforeign antigens (Ags) andself-antigens that are presented on major histocompatibility complexclassII
`(MHC-Il) by Ag-presenting cells, such as macrophages,bind to T-cell receptors (TCRs). Activated T cells migrate to, adhere at and penetrate through the blood-brain bar-
`rier, steps that are mediated by adhesion molecules, proteases and chemokines. In the CNS,the T cells are reactivated by CNS Ags presented on MHC-II by other Ag-pre-
`senting cells, predominantly microglial cells. The reactivated T cells secrete pro-inflammatory cytokines, such as interferon y (IFN-y) or interleukin 2 (IL-2), which induce
`CNSinflammation by subsequent activation of macrophages,other T cells and B cells. MacrophagesandTcells attack the myelin sheath of oligodendrocytes (OGs) by
`cytotoxic mediators, mainly tumornecrosis factor « (TNF-«), Oz radicals and nitric oxide (NO). B cells differentiate into plasma cells. These secrete demyelinating antibodies
`that can guide and activate macrophages, and ignite the complement cascade, which causes assembly of the membrane attack complex and causes pore formation in
`myelin membranes. Demyelination occurs by four different pathological patterns (1-4), as described in the main text.
`
`oligodendrocyte apoptosis; and (4) primaryoligodendrocyte
`degeneration. The mechanismsofthe latter two patterns
`remain elusive.
`In addition to this autoaggressive inflammatory phase,
`axonal loss, which causes irreversible disability, occurs
`early in the course of the disease [24,25]. It is unclear
`whether axonal damageis the consequence of a primary
`active destructive process executed by,
`for example,
`macrophages and cytotoxic molecules derived from CD8
`cells [26], or a (patho)physiological response that occurs
`secondarily to demyelination and is based on increased
`vulnerability [24,27]. Axonal damage appears to be
`initiated by increased membrane permeability followed
`by enhanced Ca?" influx. Disruption of axonal transport
`alters the cytoskeleton and leads to axonal swelling,
`lobulation and, finally, disconnection [28].
`The different molecules involved in each phase of MS
`are summarized in Fig. 1 [1,17,29].
`
`MStherapeutics: immunomodulatory profile in vitro and
`in vivo
`The structural features of the therapeutic agents are
`shown in Fig. 2. Interferon Bla (IFN-Bla), IFN-B1b and
`glatirameracetate (GA) are approved for the treatment of
`relapsing-remitting (RR) MS,
`IFN-§1b for secondary-
`progressive (SP) MS, IFN-B1a for SP MS with super-
`imposedrelapses and mitoxantrone for worsening forms of
`RR and SP MS.
`
`Glucocorticosteroids
`Methylprednisolone and prednisoloneare the mainstays of
`treatment for acute attacks in MS [30]. Becauseof side-
`effects in long-term treatment regimens and superior
`efficacy compared with oral application, steroids are
`mostly delivered in intravenous pulses. Most of the
`immunologicaleffects of glucocorticosteroids are mediated
`by specific, ubiquitously distributed intracellular recep-
`tors. These form a multiprotein complex with two
`http://tips.trends.com
`
`molecules of heat shock protein 90 (HSP90), one molecule
`of HSP70, one molecule of HSP56, one so-called immuno-
`philin, andother, less well characterized, proteins. After
`binding lipophilic glucocorticosteroid, the receptor dis-
`sociates irreversibly from the rest of the complex, and the
`steroid—receptor complex ligates specific glucocorticoid-
`responsive elements in nuclear DNA.This either upregu-
`lates or downregulates the transcription of target genes.
`The nuclearfactor kB (NF-«B) transcription factor induces
`the expression of multiple inflammatory and immune
`genes [31]. Glucocorticosteroids inhibit NF-«B, both by
`direct binding of the activated receptor to NF-KB and by
`inducing expression of the specific inhibitory protein IkB
`in lymphocytes.
`effects of glucocorticosteroids
`The immunological
`include: (1) inhibition of T-cell activation and production
`of pro-inflammatory cytokines, such as interleukin 2 (IL-2)
`and IFN-y; (2) increased production of anti-inflammatory
`Th2 cytokines by T cells; (3) inhibition of IFN-y-induced
`major histocompatibility complex (MHC) class IT expres-
`sion on macrophages; (4) decreased production of pro-
`inflammatory cytokines, prostaglandins and leukotrienes
`by macrophages;(5) diminished adhesion of neutrophils to
`endothelial cells; and (6) inhibition of endothelial-cell
`activation, and expression of MHCclass II and adhesion
`molecules. Taken together,
`these actions underlie the
`sealing effect of steroids on the blood—brain barrier, which
`prevents further access of immunecells and molecules to
`the brain.
`In addition to the genomic effects mediated by steroid
`receptor activation, there is recent evidence that high
`doses of glucocorticosteroids induce apoptosis of target
`cells by a direct, non-genomic effect
`[32]. In this case,
`apoptosis is thought to be mediated by a direct effect on
`cellular membranes, which influences transmembranous
`ion transport and, subsequently, reduces the availability of
`ATP. Steroid-induced apoptosis of autoreactive T cells is
`important for the termination of an acute MSattack [32].
`
`

`

`TRENDSin Pharmacological Sciences Vol.24 No.3 March 2003
`
`133
`
`IFN-B
`IFN-8 is highly species specific. In humans, the IFN-B
`polypeptide is produced andsecreted by fibroblasts, but
`virtually all mammalian cells can produce IFN-f on stimu-
`lation. Two recombinant IFN-B preparations, IFN-B1a and
`IFN-81b, are approved for treatment of MS. The optimal
`dosage and route of administration (subcutaneously
`versus intramuscularly), the resulting pharmacodynamic
`properties of IFN-f and therole of neutralizing antibodies
`are controversial [33-37].
`IFN-8 has immunomodulatory properties, antiviral and
`anti-proliferative effects, and promotescell differentiation
`[38]. Although the mechanismsofaction of IFN-B are not
`fully understood, there is agreement that the majoreffects
`are mediated by activation of a transmembrane IFN
`receptor, which leads to either the upregulation or down-
`regulation of target genes [39,40]. Unlike IFN-vy, the ‘type
`II interferon’, IFN-8 and IFN-a (type I interferons) share
`a receptor, which consists of two chains, IFN-aR1 and
`IFN-aR2, or several subvariants. Binding of IFN-8 (and
`other type I interferons) to the extracellular domain of the
`receptor induces an intracellular signal
`transduction
`cascade that involves: (1) recruitment and activation of
`the cytoplasmic tyrosine kinase 2 by IFN-aR1, and Janus
`kinase 1 by IFN-aR2; (2) subsequent phosphorylation and
`recruitment of signal transducers andactivators of tran-
`scription (STAT1 and STAT2) to form a STAT1-—STAT2
`heterodimer; (3) migration of the STAT1—STAT2 hetero-
`dimer to the nucleus; (4) association of STAT1—STAT2
`with the p48 protein, to form the active ‘IFN-stimulated
`genefactor 3’; (5) binding of IFN-stimulated genefactor 3
`to promoter elements andinitiation of the transcription
`of target genes. The variations in this rough scheme
`ofsignal transduction thatleadto different effects ofIFN-B
`in different target cells are based on variations in each
`of the steps outlined above, which are not yet fully
`understood[39].
`The range ofimmuneeffects attributed to IFN-B is wide
`and ever broadening [38,41]. It suppresses T-cell pro-
`liferation, diminishes IFN-y-induced upregulation of MHC
`class II expression, induces the production of Th2 cyto-
`kines andreduces synthesis ofTh1 cytokines, and inhibits
`monocyte activation. In addition, IFN-8 downregulates
`matrix metalloproteinases (MMPs), decreases surface-
`expressed adhesion molecules and increases the release
`ofsoluble adhesion molecules, which combineto reduce the
`migratory potential of T cells.
`
`GA
`GA is the acetate salt of a standardized, randomized
`mixture of synthetic polypeptides. After subcutaneous
`administration, GA is quickly degradedto free aminoacids
`and small oligopeptides and, thus, most probablyinitiates
`its major immunological effects in the periphery. Unlike
`the multiple immunological effects of IFN-8, which are
`antigen nonspecific, the immunomodulatory potential of
`GAis based on immunecells that are specific for myelin
`basic protein (MBP)and, probably, other myelin antigens
`[38,42]. Four major mechanismsof GA activity have been
`identified:
`(1) competition between GA and MBP for
`binding to MHC molecules;
`(2) competition between
`http://tips.trends.com
`
`GA-MHCcomplexes and MBP—MHCcomplexesfor bind-
`ing to the T-cell receptor (TCR); (3) activation andtoler-
`ance induction in MBP-specific T cells through an altered
`peptide ligand; and (4) induction of GA-reactive, Th2-like
`regulatory cells that mediate local bystander suppression.
`Whereas the first two effects are thought to take
`place only in vitro, the latter two are also likely to occur
`in vivo and could contribute to the anti-inflammatory
`effects of GA [42].
`
`Intravenous immunoglobulins
`Intravenous immunoglobulins (IVIgs) are pooled, purified,
`human Igs with virtually unlimited specificities. Consist-
`ent with their composition, several mechanismsof action
`of IVIgs have been suggested [43,44]. These include:
`(1) anti-idiotype antibodies (binding and inactivation of
`pathogenic antibodies by IVIgs);
`(2) blockade of Fc
`receptors on mononuclear phagocytes; (3) downregulation
`of the endogenous production of Igs; (4) attenuation and
`abrogation of complement-mediated effects (partially by
`‘consumption’ of complement components); (5) neutraliz-
`ation of molecules (TCR, MHC, costimulatory molecules
`and cytokines)
`that are involved in inflammation;
`(6) induction ofanti-inflammatory cytokines; and (7) induc-
`tion of apoptosis. Experimental evidence indicates that
`IVIgs mightalso be involved in myelin repair, but clinical
`proofof this is lacking [44]. Currently, the role of IVIgs in
`treating MS is undecided.
`
`Immunosuppressive agents
`Mitoxantroneis effective in the treatment of severe active
`forms of MS [45,46]. Although mitoxantrone suppresses
`both T cells and B cells,
`in vitro experiments indicate
`that major sites of action in MS are antigen-presenting
`cells, which are induced to undergo apoptosis, and macro-
`phages, the major effector cells of demyelination, which
`are deactivated [47].
`Azathioprine is widely used in organ transplantation
`and autoimmunedisorders [48] and is considered a second-
`line drug in MS.It mainly targets the activation, pro-
`liferation and differentiation of both T cells and B cells by
`competition between its metabolites and DNA nucleotides.
`Specific immunomodulatory properties have not been
`reported to date.
`Cyclophosphamideis usedto treat severe and rapidly
`progressive forms of MS, although evidenceofits efficacy
`in clinical trials is conflicting. In addition to strong
`immunosuppression, cyclophosphamide exerts immuno-
`modulatory effects that shift immuneresponses from Th1
`towards Th2 by an unknown mechanism [49].
`One study has shown methotrexate to convey some
`therapeutic benefit in progressive forms of MS [10], but its
`mechanismsof action in autoimmunediseasesare largely
`unknown.
`
`Potential new agents
`Anti-adhesion molecules
`Adhesion molecules comprise several families of mol-
`ecules that are essential in virtually all cellular inter-
`actions ofimmunecells [50,51]. The selectins (e.g. L-selectin,
`E-selectin and P-selectin), which have a lectin-binding
`
`

`

`134
`
`TRENDSin Pharmacological Sciences Vol.24 No.3 March 2003
`
`
`
`(a) Glucocorticosteroids
`
`(g) Cyclophosphamide
`
`—C|
`
`
`
`HoCS~ cy,
`
`QQ. N—~cH,
`a
`O~
`~NH HeC~o,
`
`279.1 Da
`
`(h) Methotrexate
`
`H2N
`
`N
`
`NH»
`
`7 . aNn @Z
`
`N
`
`N'
`
` CH»NCH3
`
`Hooc
`HOOC~-CH)-CH2-CH—NH-CO
`454.4 Da
`
`(i) Anti-adhesion molecule antibodies
`150 kDa
`
`Murine
`Human
`
`150 kDa
`
`(j) Statins
`
`Simvastatin (418.6 Da)
`
`(k) Neurotrophic factors
`
`NH,
`
`COOH
`
`BDNF
`(13.5 kDa)
`
`Disulfide bridges:
`Cys13-80; Cys58—109; Cys68-111
`
`(I) Neuroprotective agents
`
`oO
`
`CF3~ CrN
`
`s
`
`Riluzole (234.2 Da)
`
`
`
`Methylprednisolone (374.5 Da)
`
`(b) Interferon B (IFN-B)
`
`
`Cys31 Cys17
`
`NH»)
`Met1
`
`E
`
`COOH
`Asn166
`
`Sugar
`residues
`Disulfide bridge:
`Cys31-141
`
`IFN-B1a
`(22.5kDa)
`
`Cys31
`
`Cys141
`
`Ser17
`
`A
`
`B
`
`D
`
`E
`
`NH
`Ser2
`
`COOH
`Asn166
`
`IFN-B1b (18.5 kDa)
`
`(c) Glatiramer acetate (GA)
`COLSON acid
`GOBHBOOGLLOBO
`oeDESAISUSENLSAIDC
`HOSASAAN
`CRDSADADESLSCNING
`SSIES
`et tat tet tet
`
`4.7-11.0 kDa
`
`(d) Intravenous immunoglobulins (IVIgs)
`
`Vv,
`
`Gy
`
`C
`
`L
`
`(\y,,
`
`Fab fragment<
`.
`:
`Hinge region
`
`Fc fragment
`
`~— Light chain
`<<— Heavy chain
`
`:
`,
`Variable region
`Constant region
`.
`.
`— Disulfide bridge
`
`150 kDa
`
`(e) Mitoxantrone
`
`OH
`
`© H-N-CH)-CH)-NH-CH)-CH,-OH
`
`e 2 HCI
`
`OH
`
`O H-N-CHj-CH2-NH-CH)-CH2-OH
`517.4 Da
`
`Azathioprine
`
`(fy
`
`—
`
`OxLO
`vsLy
`
`7
`
`H3c’
`
`N
`
`H
`NeN-N
`
`‘T
`
`277.3 Da
`
`TRENDSin Pharmacological Sciences
`
`http://tips.trends.com
`
`

`

`TRENDSin Pharmacological Sciences Vol.24 No.3 March 2003
`
`135
`
`[55]. In addition, statins
`transactivator IV promoter
`curtail T-cell proliferation, lower expression of activation
`surface markers and induce production of the cytokine
`IL-4 [56]. Currently, simvastatin is being tested in a Phase
`II clinical trial in MS. The oral administration, extensive
`safety data, possible effects synergistic with IFN-8 and
`simultaneous treatment of co-morbidity make statins
`particularly attractive candidate agents.
`
`domain that binds to glycosylated andsialylated ligands,
`are involved in the ‘rolling’ of leukocytes. Integrins
`comprise a4B1, «581, a681 integrins [also known as
`very late antigen 4 (VLA-4) VLA-5 and VLA-6, respec-
`tively] and leukocyte function antigen 1 (LFA-1). VLA-4
`and LFA-1 are present on circulating leukocytes and
`mediate their migration across the vascular endothelium;
`in addition, LFA-1 is a costimulatory molecule in T-cell
`activation. Cadherins
`(including classical cadherins,
`desmosomal cadherins and protocadherins) form molecu-
`lar links between adjacentcells by zipper-like structures.
`Members of the Ig superfamily, such as intercellular
`adhesion molecule 1 (ICAM-1),
`ICAM-2 and ICAM-3,
`vascular cell adhesion molecule (VCAM), and LFA-2 and
`LFA-3 have Ig-like domains. ICAM-1 and VCAM-1 are the
`counter-receptors of LFA-1 and VLA-4, respectively, and
`are thus involved in leukocyte migration.
`Antibodies against single adhesion molecules can
`potently inhibit crucial steps in the pathogenesis of MS,
`especially lymphocyte migration. Currently,
`the most
`promising candidate is natalizumab [52], a humanized
`monoclonal antibody against the a4 chain of 0481 integrin
`that has been effective in PhaseII clinical trials [53].
`‘Humanized’ meansthat a murine antibody clone has been
`Neuroprotective agents
`grafted toa human IgG4 framework at the complementary
`Recent evidence indicates that axonal and neuronal
`determining region to reduce its immunogenicity [52]. Two
`
`degeneration occur early in MS and-—as the disease
`large-scale, PhaseIII clinical trials in RR MS have been
`evolves — predominate the underlying pathogenetic
`initiated recently and small-molecule antagonists, which
`mechanisms [4,17,24]. This paradigm shift has obvious
`offer advantages over monoclonal antibodies (oral avail-
`therapeutic implications.
`In addition to neurotrophic
`ability and lack of antigenicity) will certainly be explored
`in the future [54].
`factors, other chemically defined neuroprotective agents
`that save neuronsfrom toxic stress [64], such asriluzole (a
`potent K* channel activator used in amyotrophiclateral
`sclerosis) are either being investigatedor are likely to be
`tested in clinical trials.
`
`Neurotrophic factors
`Neurotrophic factors are secreted proteins that regulate
`the survival anddifferentiation ofnervecells [59]. They act
`via specific neurotrophin receptors [60]. Neurotrophic
`factors have been observed to shift the CNS cytokine
`balance from Th1 to Th2 by an undefined mechanism [61].
`In addition, they might promote survival ofneurons in MS
`lesions [6] and pharmacological neuroprotection by the
`exogenous application of neurotrophic factors provides
`a promising therapeutic approach [59,62]. Insulin-like
`growth factor 1 is currently explored in a PhaseI/II study.
`Other attractive candidates are brain-derived neuro-
`trophic factor, glial growth factor and ciliary neurotrophic
`factor [63].
`
`Statins
`Statins are effective lipid-lowering agents. Recent findings
`indicate that they have additional immunomodulatory
`effects in vivo in the animal model experimental auto-
`immune encephalomyelitis (EAE), and in vitro [55-57].
`Oneproposed immunomodulatory mechanism ofstatins is
`basedon theselective inhibition of the adhesion molecule
`LFA-1, an integrin that is involved in inflammation [58].
`Furthermore,statins reduce IFN-y-induced MHCclassII
`expression by blocking transcription of the class II
`
`Similarities and peculiarities
`there are
`Given the complexity of MS pathogenesis,
`multiple sites where immunomodulatory agents, either
`alone or in combination, might be effective. Current
`knowledge of the sites of action obtained from in vitro
`and in vivo data is summarized in Fig. 3. Although the
`
`Fig. 2. Structures of immunomodulators. Interferon f (IFN-f), glatiramer acetate (GA) and mitoxantrone are currently approved for use in multiple sclerosis (MS). (a) Gluco-
`corticosteroids are derived from cortisol, a naturally occurring adrenal hormone. Methylprednisolone is commonly usedto treat acute relapses in MS.(b) IFN-8 contains
`166 residuesthat form five a-helices (A-E). Two preparations of recombinantIFN-B are approved for MStreatment. IFN-B1a is produced in Chinese hamsterovarycells and
`is pharmacologically identical to the natural form (i.e. it is glycosylated by oligosaccharides at Asn80). Compared with natural IFN-f, IFN-81b is not glycosylated, it lacks
`Met1 (i.e. IFN-81b has 165 residues) and thereis a Cys to Ser substitution at residue 17. In both forms of recombinant IFN-B,there is a disulfide bridge between Cys31 and
`Cys141. Structurally important amino acids are shownin green.(c) GAis the acetate salt of a standardized, randomized mixture of synthetic polypeptides (average length
`45-100 aminoacids) that consist of L-glutamic acid, L-lysine, L-alanine and L-tyrosine in the molar ratio of 0.14:0.34:0.43:0.09. An example sequence is shown. (d) Intra-
`venous immunoglobulins (IVIgs) are pooled, purified, human Igs prepared by cold ethanol fractionation of human plasma derived from 3000-10 000 donors. Igs share a
`commonY-shapedstructure, which consists of two heavy chains (50 < hsp sp = 0.25 > kDa each) and twolight chains (25 < hsp sp = 0.25 > kDa each) connectedbydisul-
`fide bridges. The variable region is responsible for antigen recognition. The five heavy-chain isotypes determine the immunoglobulin classes. IVigs contain ~ 95% IgG,
`2.5% IgA and a minority of IgM. (e) Mitoxantrone is an anthracenedionederivative that is related to the anthracyclins doxorubicine and daunorubicine.It interacts with
`topoisomerase-2 and causessingle and double string breaks by intercalating with DNA. (f) Azathioprine is a purine analoguethat is metabolized rapidly to the cytotoxic
`and immunosuppressantderivatives 6-mercaptopurine and thioinosine acid, the latter competes with DNAnucleotides. (g) Cyclophosphamideis an alkylating agent of the
`nitrogen mustard group;its active metabolites, formed by the activity of hepatic cytochrome P450, induce DNA-string breaks. (h) Methotrexate interferes with DNA syn-
`thesis by inhibiting dihydrofolate reductase and, thus, thymidine biosynthesis. It reduces 1-carbontransfers to purines. (i) Monoclonal antibodies against adhesion mol-
`ecules inhibit homing of T cells to the CNS. To reduce immunogenicity, newer approachesuse ‘humanized’ antibodies, which are chimeras of murine variable regions and
`humanconstant regions connected at the complementary determining region.(j) Statins inhibit 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase. Statins are
`effective lipid-lowering agents because HMG-CoAreductaseis essential to the cholesterol biosynthesis pathway, but they also have immunomodulatory properties. (k) The
`structure of neurotrophic factors is dominated by four antiparallel pairs of 8 strands (green arrows) held in place by three disulfide bridges. Neurotrophic factors are active
`as homodimersandact via specific neurotrophin receptors. Brain-derived neurotrophic factor (BDNF) is shown,with the position of the cysteine residues that form the dis-
`ulfide bridges in green.(1) Neuroprotective agents comprise a heterogeneousgroup thatis characterized by their ability to exert protection from neurotoxic stress. Riluzole
`acts by activating Kt channels.
`http://tips.trends.com
`
`

`

`136
`
`TRENDSin Pharmacological Sciences Vol.24 No.3 March 2003
`
`
`
`
`(Cc)T-cell migration, adhesion andpenetration
`Steroids, IFN-B, IVigs, anti-adhesion molecule
`antibodies, statins
`
`(@) Release of pro-inflammatory cytokines
`Steroids, IFN-B, IVigs
`
`(h)Effectorcell activation: macrophages
`Steroids, IFN-B, mitoxantrone
`
`
`
`
`
` (k) Axonal damage
`
`
`
`
`(a) Antigen-presenting cell (d)T-cell reactivation (i) Effectorcell activation:Tcells
`Steroids, IFN-B, IVigs, mitoxantrone,
`IFN-B, IVigs, azathioprine
`Steroids, IFN-B, IVigs, azathioprine
`Neurotrophins, neuroprotective agents
`statins
`
`
`
` TRENDSin Pharmacological Sciences
`
`
`
`
`
`(b) Autoreactive T cell
`IFN-B, GA, mitoxantrone,
`azathioprine, cyclophosphamide,
`methotrexate, statins
`
`
`
`(9)
`
`GA
`o=(©)
`TCR
`MHC-II
`
`The
`
`GA-reactive
`regulatory T cell
`
`
`
`
`
`
`(j)Effectorcellactivation:Bcells,
`.
`roan
`plasmacells and demyelinating antibodies
`IVIgs, mitoxantrone, azathioprine
`
`
`
`(I) Demyelination
`+ Remyelination as neuroconstructive approach?
`+ Stem cells as source?
`+ Schwanncell preparations?
`
`
`’
`’ Bystander
`suppression
`effect
`
`/
`
`/
`
`/
`
`v
`/
`7 IL-4
`‘CNS Ag
`Ee f
`
`TCR =
`MHC-II
`
`(f) Cytokineshift
`Th1 —> Th2
`Steroids, IFN-B,
`GA-reactive T cells,
`cyclophosphamide,
`factors
`
`statins,neurotrophic
`
`Fig. 3. lmmunomodulatory agents have commonsites of action in multiple sclerosis (MS) and interfere at other sites individually. (a) Antigen presentation by antigen-pre-
`senting cells is inhibited. (b) Proliferation of autoreactive T cells is blocked or tolerance is induced.(c) T-cell migration, adhesion and penetration into the CNS are abro-
`gated. (d) T-cell reactivation in the CNS is blocked. (e) Release of pro-inflammatory cytokines by autoreactive T cells is diminished. (f) Several agents induce a shift from
`pro-inflammatory Th1 to anti-inflammatory Th2 cytokines. (g) Uniquely, GA induces regulatory T cells that cross-react to both GA and CNSAgs.After reactivation inside
`the CNS,these regulatory T cells exert a local bystander suppression effect through the release of Th2 cytokines. (h) Activation of macrophagesorsecretion of pro-inflam-
`matory mediators are curtailed. (i) Apoptosis of autoreactive T cells is induced. (j) Activation of B cells and their differentiation to plasma cells is blocked; demyelinating
`antibodies are neutralized by several mechanisms. (k) Axonal damage is reduced by neuroprotective approaches.(I) Therapeutical approaches that target demyelination
`arestill at an experimental stage. Abbreviations: Ag, antigen; GA, glatirameracetate; IFN, interferon; IL, interleukin; IVigs, intravenous immunoglobulins; MHC, majorhisto-
`compatibility complex; TCR, T-cell receptor; Th1, T helper1.
`
`immunomodulatory agents presented share some common
`features, many differ markedly in their mechanisms and
`sites of action. General properties to be considered are
`nonspecific versus specific antigen effects (e.g.
`IFN-B
`versus GA), polydirectional versus unidirectional immuno-
`modulatory properties(e.g. the pleiotropic effects of IFN-B
`versus
`the monoclonal
`antibody directed against
`a4-integrin)
`and
`pure
`immunomodulators
`versus
`immunosuppressants with additional immunomodulatory
`properties (e.g. IFN-B versus mitoxantrone).
`The interactions between the various drugs arestill
`widely unknown [65,66]. However, gaining more know]-
`edge of the individual targets might allow combination
`therapies to be developed that have either additive or
`synergistic effects.
`
`Concluding remarks
`Although an increasing arsenal of immunomodulatory
`agents is available, many questions remain about each
`single approach [67]. These include whento initiate and
`whento stop treatment, the optimal dose, frequency and
`route ofadministration, the long-term effects oftreatment,
`the occurrence and relevanceof neutralizing antibodies to
`biological agents, and the cost utility.
`http://tips.trends.com
`
`It is likely that differential therapy in MS will gain in
`importance in the near future. As evidence mounts that
`MS comprises several distinct subforms[5], the choice of
`treatmentfor individual patients will, ideally, be determined
`by the knowledgeof the specific underlying pathomechan-
`ism and the respective optimal drug or combination ofdrugs.
`The goalis tailor-made immunotherapy.
`Recent evidenceindicates that autoimmunecells have
`neuroprotective properties that are mediated, at least in
`part, by neurotrophic factors [6,7]. Based on this, a novel
`therapeutic approach would be to ‘import’ neurotrophic
`factors into the CNS via immunecells [68]. However, these
`findings also provide an impetusto shift the rationale of
`nonselective immunosuppressive therapies towards a more
`selective immunomodulatory regimen, which would pre-
`serve or even enhance the neuroprotective functions of
`autoimmuneT cells[6].
`In a contrasting approach, bone marrow transplan-
`tation (the most aggressive strategy to eradicate a
`disordered immune system) is being tested clinically
`[69,70], but this remains controversial [71].
`Taking neurobiological approaches further, the next
`step on the horizon (which is being investigated in
`animal models and in vitro) is neuroregeneration. This
`
`

`

`TRENDSin Pharmacological Sciences Vol.24 No.3 March 2003
`
`137
`
`Box 1. Currentclinical trials in multiple sclerosis
`
`http.//www.nationalmssociety.org/pdf/research/agents.pdf
`http.//www.nationalmssociety.org/pdf/research/clinicaltrials. pdf
`
`neuroconstructive therapy encompassesinhibition of scar
`formation and promotion of axonal regrowth and remye-
`lination [72-74]. The contentious issue of the use of
`embryonic stem cells as source of progenitors for remye-
`lination must be discussed in this context. Alternatively,
`retrieval of multipotential neuronal stem cells from the
`adult brain has been suggested [75]. Recently, oligo-
`dendrocyte progenitors have been identified, even in
`chronic MS lesions [76]. Transplantation of peripheral
`Schwanncells is being studied in Phase II clinical trials
`anddefined chemical agents for CNS myelination, such as
`eliprodil (a high-affinity ligand of a so-called o-receptor
`that mediates neuroprotection) are being explored experi-
`mentally [77]. B

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