throbber
Review
`
`TRENDS in Pharmacological Sciences Vol.24 No.3 March 2003
`
`131
`
`Immunomodulation in multiple
`sclerosis: from immunosuppression
`to neuroprotection
`Oliver Neuhaus1, Juan J. Archelos2 and Hans-Peter Hartung1
`
`1Department of Neurology, Heinrich-Heine-Universita¨ t, Moorenstrasse 5, 40225 Du¨ sseldorf, Germany
`2Multiple Sclerosis Research Group, Department of Neurology, Karl-Franzens-Universita¨ t, 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 agents is avail-
`able. Two classes of immunomodulators are approved
`for long-term treatment of MS, the efficacy of several
`promising new concepts is being tested in clinical trials
`and classical immunosuppressive agents used in MS
`treatment have been shown to exert specific, immuno-
`modulatory effects. Furthermore, two recent obser-
`vations have changed our basic understanding of the
`pathogenesis of MS. First, immune cells 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.
`
`‘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 immune responses contrib-
`ute to the formation and maintenance of MS lesions [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-b) and glatiramer acetate (GA),
`
`Corresponding author: Oliver Neuhaus (oliver.neuhaus@uni-duesseldorf.de).
`
`have been approved for the treatment of MS [8 – 10].
`Immunomodulators, which do not cause general suppres-
`sion of the immune system, shift immune responses 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-b and GA
`have been proven to be partially effective in clinical trials
`[1,11 – 14]. In the search for more efficacious 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 modes of action of the different agents.
`Here, we summarize current concepts about the mech-
`anisms of action of therapies already approved for MS and
`the most promising future candidates.
`
`Disease-relevant immune processes
`Since the first description of MS in 1835 by J. Cruveilhier
`as ‘scle´rose en taches, en ıˆles par masses disse´mine´es’ [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
`autoimmune disease [18], followed by a phase of selective
`demyelination and last, 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 of epitopes 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-

`helper or
`vated in the CNS, these T cells of either CD4

`CD8
`cytotoxic phenotype [22] release pro-inflammatory
`Th1 cytokines and orchestrate the destruction of the
`myelin sheath by various types of immune cells. 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-
`ment activation [3,23]; (3) distal oligodendrogliopathy and
`
`http://tips.trends.com 0165-6147/03/$ - see front matter q 2003 Elsevier Science Ltd. All rights reserved. doi:10.1016/S0165-6147(03)00028-2
`1
`
`Hopewell EX1007
`
`

`

`132
`
`Review
`
`TRENDS in Pharmacological Sciences Vol.24 No.3 March 2003
`
`Fig. 1. Pro-inflammatory T cells in the periphery are activated when foreign antigens (Ags) and self-antigens that are presented on major histocompatibility complex class II
`(MHC-II) 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 g (IFN-g) or interleukin 2 (IL-2), which induce
`CNS inflammation by subsequent activation of macrophages, other T cells and B cells. Macrophages and T cells attack the myelin sheath of oligodendrocytes (OGs) by
`cytotoxic mediators, mainly tumor necrosis factor a (TNF-a), O2 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) primary oligodendrocyte
`degeneration. The mechanisms of the 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 damage is 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 Ca2þ
`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].
`
`MS therapeutics: immunomodulatory profile in vitro and
`in vivo
`The structural features of the therapeutic agents are
`shown in Fig. 2. Interferon b1a (IFN-b1a), IFN-b1b and
`glatiramer acetate (GA) are approved for the treatment of
`relapsing-remitting (RR) MS, IFN-b1b for secondary-
`progressive (SP) MS, IFN-b1a for SP MS with super-
`imposed relapses and mitoxantrone for worsening forms of
`RR and SP MS.
`
`Glucocorticosteroids
`Methylprednisolone and prednisolone are the mainstays of
`treatment for acute attacks in MS [30]. Because of side-
`effects in long-term treatment regimens and superior
`efficacy compared with oral application, steroids are
`mostly delivered in intravenous pulses. Most of the
`immunological effects of glucocorticosteroids are mediated
`by specific, ubiquitously distributed intracellular recep-
`tors. These form a multiprotein complex with two
`
`http://tips.trends.com
`
`2
`
`molecules of heat shock protein 90 (HSP90), one molecule
`of HSP70, one molecule of HSP56, one so-called immuno-
`philin, and other, 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 nuclear factor kB (NF-kB) transcription factor induces
`the expression of multiple inflammatory and immune
`genes [31]. Glucocorticosteroids inhibit NF-kB, both by
`direct binding of the activated receptor to NF-kB and by
`inducing expression of the specific inhibitory protein IkB
`in lymphocytes.
`The immunological effects of glucocorticosteroids
`include: (1) inhibition of T-cell activation and production
`of pro-inflammatory cytokines, such as interleukin 2 (IL-2)
`and IFN-g; (2) increased production of anti-inflammatory
`Th2 cytokines by T cells; (3) inhibition of IFN-g-induced
`major histocompatibility complex (MHC) class II 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 MHC class II and adhesion
`molecules. Taken together, these actions underlie the
`sealing effect of steroids on the blood– brain barrier, which
`prevents further access of immune cells 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 MS attack [32].
`
`

`

`Review
`
`TRENDS in Pharmacological Sciences Vol.24 No.3 March 2003
`
`133
`
`IFN-b
`IFN-b is highly species specific. In humans, the IFN-b
`polypeptide is produced and secreted by fibroblasts, but
`virtually all mammalian cells can produce IFN-b on stimu-
`lation. Two recombinant IFN-b preparations, IFN-b1a and
`IFN-b1b, are approved for treatment of MS. The optimal
`dosage and route of administration (subcutaneously
`versus intramuscularly), the resulting pharmacodynamic
`properties of IFN-b and the role of neutralizing antibodies
`are controversial [33 – 37].
`IFN-b has immunomodulatory properties, antiviral and
`anti-proliferative effects, and promotes cell differentiation
`[38]. Although the mechanisms of action of IFN-b are not
`fully understood, there is agreement that the major effects
`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-g, the ‘type
`II interferon’, IFN-b 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-b (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 and activators 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
`gene factor 3’; (5) binding of IFN-stimulated gene factor 3
`to promoter elements and initiation of the transcription
`of target genes. The variations in this rough scheme
`of signal transduction that lead to different effects of IFN-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 of immune effects attributed to IFN-b is wide
`and ever broadening [38,41]. It suppresses T-cell pro-
`liferation, diminishes IFN-g-induced upregulation of MHC
`class II expression, induces the production of Th2 cyto-
`kines and reduces synthesis of Th1 cytokines, and inhibits
`monocyte activation. In addition, IFN-b downregulates
`matrix metalloproteinases (MMPs), decreases surface-
`expressed adhesion molecules and increases the release
`of soluble adhesion molecules, which combine to 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 degraded to free amino acids
`and small oligopeptides and, thus, most probably initiates
`its major immunological effects in the periphery. Unlike
`the multiple immunological effects of IFN-b, which are
`antigen nonspecific, the immunomodulatory potential of
`GA is based on immune cells that are specific for myelin
`basic protein (MBP) and, probably, other myelin antigens
`[38,42]. Four major mechanisms of GA activity have been
`identified: (1) competition between GA and MBP for
`binding to MHC molecules;
`(2) competition between
`
`http://tips.trends.com
`
`3
`
`GA – MHC complexes and MBP – MHC complexes for bind-
`ing to the T-cell receptor (TCR); (3) activation and toler-
`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 mechanisms of 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 of anti-inflammatory cytokines; and (7) induc-
`tion of apoptosis. Experimental evidence indicates that
`IVIgs might also be involved in myelin repair, but clinical
`proof of this is lacking [44]. Currently, the role of IVIgs in
`treating MS is undecided.
`
`Immunosuppressive agents
`Mitoxantrone is 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 autoimmune disorders [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.
`Cyclophosphamide is used to treat severe and rapidly
`progressive forms of MS, although evidence of its efficacy
`in clinical trials is conflicting. In addition to strong
`immunosuppression, cyclophosphamide exerts immuno-
`modulatory effects that shift immune responses 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
`mechanisms of action in autoimmune diseases are 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 of immune cells [50,51]. The selectins (e.g. L-selectin,
`E-selectin and P-selectin), which have a lectin-binding
`
`

`

`134
`
`Review
`
`TRENDS in Pharmacological Sciences Vol.24 No.3 March 2003
`
`(g) Cyclophosphamide
`
`Cl
`
`H2C
`
`O
`
`O
`
`CH2
`N
`
`P
`
`NH
`
`CH2
`H2C Cl
`
`279.1 Da
`
`CH2NCH3
`
`NN
`
`(h) Methotrexate
`
`H2N
`
`N
`
`N
`
`NH2
`
`HOOC
`
`HOOC
`CH2 CH2
`CH NH CO
`454.4 Da
`
`(i) Anti-adhesion molecule antibodies
`150 kDa
`
`Murine
`Human
`
`(j) Statins
`
`150 kDa
`
`O
`
`O
`
`O
`
`O
`
`(a) Glucocorticosteroids
`
`HO
`CH3
`
`H
`
`CH3
`
`H
`
`H
`
`O
`
`CH2OH
`C
`O
`OH
`
`CH3
`Methylprednisolone (374.5 Da)
`(b) Interferon β (IFN-β)
`Cys31
`
`Cys141
`
`Cys17
`
`A
`
`B
`
`C
`
`D
`
`E
`
`NH2
`Met1
`
`Asn80
`
`COOH
`Asn166
`Sugar
`residues
`Disulfide bridge:
`Cys31–141
`
`IFN-β1a
`(22.5 kDa)
`
`Cys31
`
`Cys141
`
`Ser17
`
`A
`
`B
`
`C
`
`D
`
`E
`
`NH2
`Ser2
`
`IFN-β1b (18.5 kDa)
`(c) Glatiramer acetate (GA)
`
`COOH
`Asn166
`
`O
`
`H
`
`Simvastatin (418.6 Da)
`
`(k) Neurotrophic factors
`
`NH2
`
`COOH
`
`BDNF
`(13.5 kDa)
`
`Disulfide bridges:
`Cys13–80; Cys58–109; Cys68–111
`
`(l) Neuroprotective agents
`
`NH2
`
`NS
`
`O
`
`CF3
`
`Riluzole (234.2 Da)
`
`TRENDS in Pharmacological Sciences
`
`Lys Ala
`Ala
`Lys
`Ala
`Lys
`Glu
`Ala
`Lys
`Tyr
`
`Ala
`Ala
`Ala Tyr
`Glu
`Ala
`Ala
`Lys
`Lys
`Glu
`Ala
`Ala Ala
`Lys
`Tyr
`Ala Ala
`Lys
`Lys Lys
`Glu
`Ala
`Ala Ala Ala
`
`Lys Lys
`Ala Ala Ala
`Lys
`Lys Lys
`Tyr
`Glu
`Ala Ala
`Ala
`Lys
`Lys
`Glu
`Ala
`Ala
`Lys
`Tyr
`Glu
`Ala
`Lys
`Lys
`Lys
`
`Glu
`Ala
`Tyr
`Glu
`Ala
`
`Lys
`Ala
`Lys
`Glu
`Ala
`Lys
`Lys Lys
`Ala
`Lys
`
`4.7–11.0 kDa
`
`(d) Intravenous immunoglobulins (IVIgs)
`
`Fab fragment
`
`Hinge region
`
`Fc fragment
`
`CL
`
`VL
`
`CH1
`
`VH
`
`CH2
`CH3
`
`150 kDa
`
`Light chain
`Heavy chain
`
`Variable region
`Constant region
`
`Disulfide bridge
`
`(e) Mitoxantrone
`
`OH
`
`O
`
`H
`
`N
`
`CH2
`
`CH2 NH CH2 CH2 OH
`
`2 HCI
`
`OH O
`
`H
`
`N CH2
`
`CH2 NH CH2 CH2 OH
`517.4 Da
`
`N
`
`HN
`
`O
`
`N
`
`H3C
`
`O−
`
`N+
`
`S
`
`N
`
`N
`
`N
`277.3 Da
`
`(f) Azathioprine
`
`http://tips.trends.com
`
`4
`
`

`

`Review
`
`TRENDS in Pharmacological Sciences Vol.24 No.3 March 2003
`
`135
`
`domain that binds to glycosylated and sialylated ligands,
`are involved in the ‘rolling’ of
`leukocytes. Integrins
`comprise a4b1, a5b1, a6b1 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 adjacent cells 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 a4b1 integrin
`that has been effective in Phase II clinical trials [53].
`‘Humanized’ means that a murine antibody clone has been
`grafted to a human IgG4 framework at the complementary
`determining region to reduce its immunogenicity [52]. Two
`large-scale, Phase III clinical trials in RR MS have been
`initiated recently and small-molecule antagonists, which
`offer advantages over monoclonal antibodies (oral avail-
`ability and lack of antigenicity) will certainly be explored
`in the future [54].
`
`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].
`One proposed immunomodulatory mechanism of statins is
`based on the selective inhibition of the adhesion molecule
`LFA-1, an integrin that is involved in inflammation [58].
`Furthermore, statins reduce IFN-g-induced MHC class II
`expression by blocking transcription of the class II
`
`transactivator IV promoter [55]. In addition, statins
`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-b and
`simultaneous treatment of co-morbidity make statins
`particularly attractive candidate agents.
`
`Neurotrophic factors
`Neurotrophic factors are secreted proteins that regulate
`the survival and differentiation of nerve cells [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 of neurons 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 Phase I/II study.
`Other attractive candidates are brain-derived neuro-
`trophic factor, glial growth factor and ciliary neurotrophic
`factor [63].
`
`Neuroprotective agents
`Recent evidence indicates that axonal and neuronal
`degeneration occur early in MS and – as the disease
`evolves – predominate the underlying pathogenetic
`mechanisms [4,17,24]. This paradigm shift has obvious
`therapeutic implications. In addition to neurotrophic
`factors, other chemically defined neuroprotective agents
`that save neurons from toxic stress [64], such as riluzole (a

`channel activator used in amyotrophic lateral
`potent K
`sclerosis) are either being investigated or are likely to be
`tested in clinical trials.
`
`Similarities and peculiarities
`Given the complexity of MS pathogenesis, there are
`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 b (IFN-b), 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 used to treat acute relapses in MS. (b) IFN-b contains
`166 residues that form five a-helices (A –E). Two preparations of recombinant IFN-b are approved for MS treatment. IFN-b1a is produced in Chinese hamster ovary cells and
`is pharmacologically identical to the natural form (i.e. it is glycosylated by oligosaccharides at Asn80). Compared with natural IFN-b, IFN-b1b is not glycosylated, it lacks
`Met1 (i.e. IFN-b1b has 165 residues) and there is 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 shown in green. (c) GA is the acetate salt of a standardized, randomized mixture of synthetic polypeptides (average length
`45 –100 amino acids) 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
`common Y-shaped structure, which consists of two heavy chains (50 , hsp sp ¼ 0.25 . kDa each) and two light chains (25 , hsp sp ¼ 0.25 . kDa each) connected by disul-
`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 anthracenedione derivative that is related to the anthracyclins doxorubicine and daunorubicine. It interacts with
`topoisomerase-2 and causes single and double string breaks by intercalating with DNA. (f) Azathioprine is a purine analogue that is metabolized rapidly to the cytotoxic
`and immunosuppressant derivatives 6-mercaptopurine and thioinosine acid, the latter competes with DNA nucleotides. (g) Cyclophosphamide is 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-carbon transfers to purines. (i) Monoclonal antibodies against adhesion mol-
`ecules inhibit homing of T cells to the CNS. To reduce immunogenicity, newer approaches use ‘humanized’ antibodies, which are chimeras of murine variable regions and
`human constant 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-CoA reductase is 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 b strands (green arrows) held in place by three disulfide bridges. Neurotrophic factors are active
`as homodimers and act 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. (l) Neuroprotective agents comprise a heterogeneous group that is characterized by their ability to exert protection from neurotoxic stress. Riluzole

`acts by activating K
`channels.
`
`http://tips.trends.com
`
`5
`
`

`

`136
`
`Review
`
`TRENDS in Pharmacological Sciences Vol.24 No.3 March 2003
`
`(c) T-cell migration, adhesion and penetration
`Steroids, IFN-β, IVIgs, anti-adhesion molecule
`antibodies, statins
`
`(e) Release of pro-inflammatory cytokines
`Steroids, IFN-β, IVIgs
`
`(h) Effector cell activation: macrophages
`Steroids, IFN-β, mitoxantrone
`
`(d) T-cell reactivation
`IFN-β, IVIgs, azathioprine
`
`(i) Effector cell activation: T cells
`Steroids, IFN-β, IVIgs, azathioprine
`
`(k) Axonal damage
`Neurotrophins, neuroprotectíve agents
`
`Central nervous system
`
`Reactivation
`
`Inflammation
`
`Demyelination
`
`Axonal damage
`
`(a) Antigen-presenting cell
`Steroids, IFN-β, IVIgs, mitoxantrone,
`statins
`
`Periphery
`
`Activation
`
`Migration,
`adhesion,
`penetration
`
`Antigen-
`presenting
`cell
`
`Autoreactive
`T cell
`
`Th1
`
`(b) Autoreactive T cell
`IFN-β, GA, mitoxantrone,
`azathioprine, cyclophosphamide,
`methotrexate, statins
`
`Bystander
`suppression
`effect
`
`(g)
`
`GA
`
`TCR
`MHC-II
`
`Th2
`
`GA-reactive
`regulatory T cell
`
`IL-4
`CNS Ag
`
`TCR
`MHC-II
`
`Effector cells
`
`Target cells
`
`(f) Cytokine shift
`Th1 Th2
`Steroids, IFN-β,
`GA-reactive T cells,
`cyclophosphamide,
`statins, neurotrophic
`factors
`
`(l) Demyelination
`• Remyelination as neuroconstructive approach?
`• Stem cells as source?
`• Schwann cell preparations?
`
`(j) Effector cell activation: B cells,
`plasma cells and demyelinating antibodies
`IVIgs, mitoxantrone, azathioprine
`
`TRENDS in Pharmacological Sciences
`
`Fig. 3. Immunomodulatory agents have common sites 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 CNS Ags. After reactivation inside
`the CNS, these regulatory T cells exert a local bystander suppression effect through the release of Th2 cytokines. (h) Activation of macrophages or secretion 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. (l) Therapeutical approaches that target demyelination
`are still at an experimental stage. Abbreviations: Ag, antigen; GA, glatiramer acetate; IFN, interferon; IL, interleukin; IVIgs, intravenous immunoglobulins; MHC, major histo-
`compatibility complex; TCR, T-cell receptor; Th1, T helper 1.
`
`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 are still
`widely unknown [65,66]. However, gaining more knowl-
`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 when to initiate and
`when to stop treatment, the optimal dose, frequency and
`route of administration, the long-term effects of treatment,
`the occurrence and relevance of neutralizing antibodies to
`biological agents, and the cost utility.
`
`http://tips.trends.com
`
`6
`
`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
`treatment for individual patients will, ideally, be determined
`by the knowledge of the specific underlying pathomechan-
`ism and the respective optimal drug or combination of drugs.
`The goal is tailor-made immunotherapy.
`Recent evidence indicates that autoimmune cells 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 immune cells [68]. However, these
`findings also provide an impetus to shift the rationale of
`nonselective immunosuppressive therapies towards a more
`selective immunomodulatory regimen, which would pre-
`serve or even enhance the neuroprotective functions of
`autoimmune T 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
`
`

`

`Review
`
`TRENDS in Pharmacological Sciences Vol.24 No.3 March 2003
`
`137
`
`Box 1. Current clinical trials in multiple sclerosis
`
`http.//www.nationalmssociety.org/pdf/research/agents.pdf
`http.//www.nationalmssociety.org/pdf/research/clinicaltrials.pdf
`
`neuroconstructive therapy encompasses inhibition 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
`Schwann cells is being studied in Phase II clinical trials
`and defined chemical agents for CNS myelination, such as
`eliprodil (a high-affinity ligand of a so-called s-receptor
`that mediates neuroprotection) are being explored experi-
`mentally [77]. Because activated immune cells commonly
`release glutamate, excitotoxicity induced by glutamate is
`thought to be involved in the formation of MS lesions and
`axonal damage. Hence, glutamate receptor antagonists
`are promising candidates [78].

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