throbber
THERAPY REVIEW
`
`Biodrugs 2002; 16 (3): 183-200
`1173-8804/02/0003-0183/$25.00/0
`
`© Adis International Limited. All rights reserved.
`
`Therapeutic Approaches in Multiple Sclerosis
`Lessons from Failed and Interrupted Treatment Trials
`Heinz Wiendl1 and Reinhard Hohlfeld2,3
`1 Department of Neurology, School of Medicine, University of Tuebingen, Tuebingen, Germany
`2 Institute for Clinical Neuroimmunology, Klinikum Grosshadern, Munich, Germany
`3 Department of Neuroimmunology, Max-Planck-Institute for Neurobiology, Martinsried, Germany
`
`Contents
` . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 183
`Abstract
`1.
`Immunopathology of Multiple Sclerosis and Therapeutic Approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 184
`2. Modification of the Cytokine Pattern . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
`2.1 Tumour Necrosis Factor-α Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
`2.1.1 Infliximab (CA2) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 187
`2.1.2 Lenercept
` . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 187
`2.1.3 Commentary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 187
`2.2 Transforming Growth Factor-β2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190
`2.3 Interleukin-10 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190
`2.4 Interleukin-4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
`2.5 Commentary on Cytokine Modulators . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
`3. Various Immunosuppressants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 195
`3.1 Roquinimex . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
`3.2 Sulfasalazine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
`3.3 Gusperimus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 193
`3.4 Cladribine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 193
`4. Aspects of Remyelination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194
`4.1 Intravenous Immunoglobulins (IVIg) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194
`4.1.1 IVIg in Optic Neuritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194
`4.1.2 IVIg in Permanent Neurological Deficits . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194
`4.1.3 Commentary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194
`5. Antigen-Derived Therapies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194
`5.1 Oral Tolerance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194
`5.1.1 Oral Myelin (AI-100) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 195
`5.2 Altered Peptide Ligands . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 195
`5.2.1 Tiplimotide . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 195
`5.3 Commentary on Antigen-Derived Therapies
` . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 195
`6. Targeting Leucocyte Differentiation Molecules with Monoclonal Antibodies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 196
`6.1 Anti-CD3 (Muromonab-CD3) and Anti-CD4 (Priliximab) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 196
`Inactivation of Circulating T Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 196
`7.1 Extracorporeal Photopheresis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 196
`8. Concluding Remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 197
`
`7.
`
`Abstract
`
`The therapy for multiple sclerosis (MS) has changed dramatically over the past decade. Recent im-
`munobiological findings and current pathophysiological concepts together with advances in biotechnology,
`improvements in clinical trial design and development of magnetic resonance imaging have led to a variety of
`evaluable therapeutic approaches in MS. However, in contrast to the successfully introduced and established
`immunomodulatory therapies (e.g. interferon-β and glatiramer acetate), there have been a remarkable number
`of therapeutic failures as well. Despite convincing immunological concepts, impressive data from animal models
`and promising results from phase I/II studies, the drugs and strategies investigated showed no benefit or even
`turned out to have unexpectedly severe adverse effects.
`Although to date there is no uniformly accepted model for MS, there is agreement on the significance of
`
`Biogen Exhibit 2122
`Mylan v. Biogen
`IPR 2018-01403
`
`Page 1 of 18
`
`

`

`184
`
`Wiendl & Hohlfeld
`
`inflammatory events mediated by autoreactive T cells in the CNS. These can be modified therapeutically at the
`individual steps of a hypothetical pathogenetic cascade. Crucial corners like: (i) the prevalence and peripheral
`activation of CNS-autoreactive T cells in the periphery; (ii) adhesion and penetration of T cells into the CNS;
`(iii) local activation and proliferation and; (iv) de- and remyelination processes can be targeted through their
`putative mediators. Like a ‘specificity pyramid’, therapeutic approaches therefore cover from general immuno-
`suppression up to specific targeting of T-cell receptor peptide major histocompatibility (MHC) complex.
`We discuss in detail clinical MS trials that failed or were discontinued for other reasons. These trials include
`cytokine modulators [tumour necrosis factor (TNF)-α antagonists, interleukin-10, interleukin-4, transforming
`growth factor-β2], immunosuppressive agents (roquinimex, gusperimus, sulfasalazine, cladribine), inducers of
`remyelination [intravenous immunoglobulins (IVIg)], antigen-derived therapies [oral tolerance, altered peptide
`ligands (APL), MHC-Peptide blockade], T cell and T-cell receptor directed therapies (T cell vaccination, T-cell
`receptor peptide vaccination), monoclonal antibodies against leucocyte differentiation molecules (anti-CD3,
`anti-CD4), and inactivation of circulating T cells (extracorporeal photopheresis).
`The main conclusions that can be drawn from these ‘negative’ experiences are as follows. Theoretically
`promising agents may paradoxically increase disease activity (lenercept, infliximab), be associated with unfore-
`seen adverse effects (e.g. roquinimex) or short-term favourable trends may reverse with prolonged follow-up
`(e.g. sulfasalzine). One should not be too enthusiastic about successful trials in animal models (TNFα blockers;
`oral tolerance; remyelinating effect of IVIg) nor be irritated by non-scientific media hype (deoxyspergualine;
`bone marrow transplantation). More selectivity can imply less efficacy (APL, superselective interventions like
`T-cell receptor vaccination) and antigen-related therapies can stimulate rather than inhibit encephalitogenic
`cells. Failed strategies are of high importance for a critical revision of assumed immunopathological mecha-
`nisms, their neuroimaging correlates, and for future trial design. Since failed trials add to our growing under-
`standing of multiple sclerosis, ‘misses’ are nearly as important to the scientific process as the ‘hits’.
`
`in MS clinical trial methodology, which is largely based on ad-
`vances in nuclear MRI techniques as a ‘surrogate marker’ in the
`assessment of potential therapeutic drug effects.[5,6] However,
`despite rational therapeutic concepts, convincing preliminary an-
`imal experiments or positive experiences with other autoimmune
`diseases, some initial studies showed no proof of efficacy or
`failed because of unforeseen adverse effects (table II). Whereas
`the positive trials usually make it into prestigious journals, many
`negative trials are published merely as abstracts or not at all.[1]
`This is unfortunate, because there is a lot to learn from a negative
`result, and critical reflection is highly important for under-
`standing human MS immunopathogenesis and appropriate trial
`
`300
`
`250
`
`200
`
`150
`
`100
`
`50
`
`Number of publications
`
`0
`1965
`
`1970
`
`1975 1980
`
`1985 1990
`Year
`
`1995 2000
`
`Fig. 1. Number of yearly publications on multiple sclerosis therapy (data from
`Medline) [reproduced from Hohlfeld et al.,[1] with permission from copyright hold-
`ers, John Wiley & Sons, Inc.].
`
`1. Immunopathology of Multiple Sclerosis and
`Therapeutic Approaches
`
`Multiple sclerosis (MS) therapy has changed dramatically
`over the past decade. Based on the growing immunopathogenetic
`understanding of MS and with the assistance of modern biotech-
`nology, a growing arsenal of potential therapeutic drugs has been
`developed. Several agents have been approved and are now being
`widely used, and a whole battery of new immunomodulatory
`treatments is currently under development. The methodology of
`MS trials has evolved in parallel with the therapeutic agents,
`utilising magnetic resonance imaging (MRI) techniques to great
`advantage. The sense of excitement in the field of MS therapeu-
`tics is reflected by the soaring number of publications (figure 1).
`Although to date there is no uniformly accepted model for
`MS, there is agreement on the significance of inflammatory
`events mediated by autoreactive T cells in the CNS.[2] These can
`be modified therapeutically at the individual steps of a hypothet-
`ical pathogenetic cascade (figure 2). Crucial steps such as the
`prevalence and peripheral activation of CNS-autoreactive T cells
`in the periphery, adhesion and penetration of T cells into the CNS,
`local activation and proliferation, and de- and remyelination pro-
`cesses can be targeted through their putative mediators (table I).
`Like a ‘specificity pyramid’, therapeutic approaches therefore
`cover the field from general immunosuppression to specific tar-
`geting of T-cell receptor peptide major histocompatibility com-
`plex (MHC).[3,4] In addition, significant progress has been made
`
`© Adis International Limited. All rights reserved.
`
`Biodrugs 2002; 16 (3)
`
`Page 2 of 18
`
`

`

`185
`Therapeutic Approaches in Multiple Sclerosis
`
`
`
`Myelln sheath
`
` : } Myelln antigens
`
`—> Cell mlgratlon
`
`—> Soluble mediators, cytokhes.
`chemoklnes a pretenses
`
`Fig. 2. Cnicial steps in miltiple sclerosis pathogenesis. Pre-exist'ng autoreac'tive T cells are activabd outside the CNS. The activated T cells traverse the blood-brain
`barrier and are locdly re-activated when they recognise ’their‘ antigen on the surface at local antigen-presenting cells. The activated T cells secrete cytokines that
`stimulate microgia eels and astrocytes. recruit additional inflammatory cells, and induce antibody production by plasma cells. Antinyelin antbodies and activated
`macrophages/microglia cells are thought to cooperate in dernyelination. Dilterent steps at the putative immunopathological cascade and ditterent mediators can be
`targeted therapeutically (see table II) [reproduced from Hatfield,“ with permission from copyright holders. Oxford University Press]. MHC = rmior histocornpatibility
`corrpIex; TCR = T-cell receptor.
`
`design. In this review we discuss the immunobiological back-
`ground, the experimental basis and the clinical studies of some
`agents and therapeutic strategies in MS treatment which were not
`effective or led to early trial termination for other reasons.
`
`2. Modification ot the Cytoklne Pattern
`
`2.1 Tumour Necrosis Factor—a Antagonists
`
`Tumour necrosis factor (TNF)-0t, initially characterised for
`its tumoricidal activity, plays an important role in acute and
`
`chronic inflammation (reviewed by Aggarwal and Nataljan,[3°]
`Beutlerp”). TNFa, mainly produced by T cells and macro-
`phages, activates the vascular endothelium and increases penne-
`ability. Together with interferon (IFN)-'y, TNFa stimulates the
`production of nitric oxide (NO) and reactive oxygen derivates,
`the release of interleukin (IL)-1 and many other cytokines, as well
`
`OAdshtemotlondLhitedAl
`
`htS reserved
`
`Page 3 of 1
`
`as all metabolites of arachidonic acid. TNFa is one of at least ten
`
`(known) members of a ligand family that activates a correspond-
`
`ing family of structurally related receptorslm The receptors trig-
`ger signals for cell proliferation and apoptosis which play an
`important role in development as well as in the induction of an
`
`immune response.
`There are two types of TNF receptors: TNFRI-p55 and
`
`TNFRII—p75. They are found either in a transmembrane or in a
`secreted form, consisting of two subunits, which are stimulated
`
`not only by TNFa but also by lymphotoxin-a. Most known bio-
`
`logical effects are mediated by the TNFRI-p55 subunit, which
`
`binds ligands with a higher affinity than TNFRII-p75. It is im-
`
`portant to mention that the receptors are able to mediate different
`
`signalling pathways, which partly explains the pleiotropism and
`
`the dependence of TNF effects on the cellular context.
`Numerous investigations have identified TNFa as an essen-
`
`tial pathogenetic factor in different models of experimental aller-
`
`Blodrugszmz-Ioo)
`
`

`

`186
`
`Wiendl & Hohlfeld
`
`Table I. Examples of recent multiples sclerosis (MS) trials that did not show a convincing clinical benefit (adapted from Hohlfeld and Wiendl,[1] with permission
`from copyright holders, John Wiley & Sons, Inc.)
`
`Agent
`
`Mechanism of action
`
`MS type (no. of patients)
`[trial duration]
`
`References Outcome/MRI Clinical
`effect
`
`Problems
`
`Sulfasalazine
`
`Gusperimus
`
`Immunosuppressants
`Roquinimex
`Synthetic immunomodulator:
`inhibition of IFNγ and TNFα
`Anti-inflammatory and
`immunomodulatory properties
`Interaction with intracellular
`heat-shock protein (hsp 70) and
`activation of NF-κB
`Adenosindeaminase-resistant
`purine nucleoside: induction of
`long-lasting lymphopenia
`
`Cladribine
`
`Cytokine modulators
`Lenercept
`Soluble TNF-receptor p55:
`inhibition of TNFα-functions
`
`Infliximab
`
`TGFβ2
`
`IL-10
`
`IL-4 (BAY
`36-1677)
`
`TNFα neutralising antibody;
`human/murine chimeric IgG1:
`inhibition of TNFα-functions
`Immune suppression, pleiotropic
`growth factor
`Recombinant cytokine: inhibition
`of macrophage APC-function,
`up-regulation of Th2-cells
`Recombinant cytokine: mutein
`with 2 AA exchanges and
`selectivity for T, B cells and
`monocytes, up-regulation of
`Th2-cells
`
`Inducers of remyelination
`Diverse immunomodulatory
`IVIg
`(Gamimune® bN)
`effects; in addition, promotion of
`remyelination in animal model
`
`RR, SP (715)
`[terminated early]
`PP, RR, SP (199) [36mo] 9
`
`7,8
`
`RR, SP (236) [12mo]
`
`10,11
`
`Positive
`
`Positive
`
`Cardiopulmonary toxicity
`
`No sustained
`effect
`No effect
`
`No sustained
`effect
`No effect
`
`Initially positive effect,
`absence of long-term benefit
`Overall effects unconvincing
`
`PP, SP (159) [12mo]
`
`12-14
`
`Positivea
`
`No effect
`
`RR (168) [11mo]
`
`SP (2) [2mo]
`
`SP (11) [6mo]
`
`15
`
`16
`
`17
`
`No effect
`
`Worsening
`
`Worsening
`
`No effect on
`EDSS
`
`No effect
`
`No effect
`
`RR, SP [terminated]
`
`Unpublished
`
`Discrepancy between MRI
`and clinical effect; probably
`no effect on tissue injury
`
`Paradoxical effect of TNFα;
`discrepancy between MRI
`and clinical effects
`Paradoxical effect of TNFα
`
`Bioavailability in the CNS?;
`nephrotoxicity
`Insufficient efficacy; possible
`induction of exacerbations
`
`[terminated]
`
`Unpublished
`
`Insufficient efficacy
`
`SDON (55) [12mo]
`
`18
`
`Not done
`
`No overall
`effect
`
`Remyelination potential may
`depend on disease activity,
`timepoint, dose and duration
`of treatment
`
`Antigen-derived therapies
`AI-100
`Oral bovine MBP; induction of
`systemic tolerance via stimulation
`of antigen-specific regulatory
`(Th2-, Th3-) cells
`
`RR, SP (TND) (67) [6mo] 19
`RR (10) [6wk]
`20
`
`No effectc
`Not done
`
`No effect
`No effect
`
`RR (30) [12mo]
`RR (515) [24mo]
`
`21
`22,23
`
`Not done
`Not
`documented
`
`Possible
`No effect
`
`Tiplimotide
`
`Altered peptide ligand; peptide
`analogue of human MBP 83-99
`
`RR (8) [terminated,
`maximum 9mo]
`
`24
`
`Worsening
`
`Worsening
`
`AG284
`(DR2:MBP84-102)
`
`Soluble HLA-DR2 with a single
`noncovalently bound MBP peptide
`
`RR (142) [terminated,
`4mo planned]
`SP (33) [3mo]
`
`25
`
`26
`
`Positived
`
`No effect
`
`No effect
`
`No effect
`
`Interindividual differences in
`target epitopes (e.g. ‘epitope
`spreading’)?; unexpected
`effects on different T cell
`populations; allergic reactions
`
`© Adis International Limited. All rights reserved.
`
`Biodrugs 2002; 16 (3)
`
`Page 4 of 18
`
`

`

`Therapeutic Approaches in Multiple Sclerosis
`
`187
`
`Table I. Contd
`
`Agent
`
`Mechanism of action
`
`TCR-directed therapies
`T cell vaccination Attenuated autologous
`MBP-reactive T cell clones,
`induction of anticlonotypic T cell
`responses
`
`MS type (no. of patients)
`[trial duration]
`
`References Outcome/MRI Clinical
`effect
`
`Problems
`
`RR (8) [22-38mo]
`
`27
`
`Mixede
`
`Mixede
`
`TCR peptide
`vaccination
`
`TCR Vß5.2 (residues 38-58),
`induction of anti-TCR-regulatory
`effects
`
`PP, SP (23, all
`HLA-DRB1*1501
`positive) [12mo]
`
`28
`
`Not done
`
`No effect
`
`Small number of patients;
`complexity and diversity of
`human autoimmune T cells;
`role of MBP in MS
`pathogenesis?
`Small number of patients;
`marginal effect on disease
`progression; heterogeneity
`and individuality of
`TCR-repertoire and
`antigen-specificity
`
`T cell inactivation
`Extracorporeal
`Direct or indirect induction of
`photopheresis
`apoptosis on circulating T cells
`
`SP (16) [18mo]
`
`29
`
`No effect
`
`No effect
`
`Quantities of peripheral
`CNS-antigen reactive T cells
`in chronic MS? Relevance of
`CNS-specific milieu for
`perpetuation of immune
`response in chronic MS
`a Favourable effect on presence, number and volume of gadolinium-enhanced T1 brain lesions and T2-lesion load,[12] no effect on the T1(hypointense)-lesion volume,[13] or on
`whole brain volume changes in patients with progressive MS.[14]
`b Use of tradenames is for product identification only and does not imply endorsement
`c Only a small number of patients (5 of each group) underwent MRI.
`d Secondary analysis of patients completing the study and receiving the lowest dose (5mg).
`e Beneficial effects on MRI and/or clinical course in 5 patients, worsening of lesions and/or relapses in 3 patients.
`AA = amino acid; APC = antigen-presenting cell; EDSS = Expanded Disability Scale; IFN = Interferon; IgG = immunoglobulin G; IL = interleukin; IVIg =
`intravenous immunoglobulin; MBP = myelin basic protein; MRI = magnetic resonance imaging; NF = nuclear factor; PP = primary progressive MS; RR =
`relapsing-remitting MS; SDON = stable demyelinating optic neuritis; SP = secondary chronic progressive MS; TCR = T-cell receptor; TGF = transforming
`growth factor; Th = T helper cell; TND = targeted neurological deficit; TNF = tumour necrosis factor.
`
`gic encephalomyelitis (EAE) and MS. It has been detected in
`inflammatory CNS lesions; in active lesions it is involved in
`pathological tissue damage (inflammation as well as demyelin-
`ation).[33,34] In vitro TNFα is cytotoxic for oligodendrocytes. The
`elimination of TNF-producing macrophages, as well as an-
`tagonisation with TNF antibodies, administration of various ther-
`apeutic drugs affecting TNFα production (e.g. thalidomide, pen-
`toxifylline, rolipram), or doses of soluble TNF receptor
`(lenercept), clearly showed a positive effect on pathogenesis and
`demyelination in various animal models.[35,36]
`A series of studies in MS patients showed a correlation of
`TNF levels in blood, serum or cerebrospinal fluid (CSF) with the
`clinical course or disease activity.[37-43]
`
`2.1.1 Infliximab (CA2)
`In an open phase I study, two patients with a severe second-
`ary chronic progressive form of MS (SPMS) were treated with a
`monoclonal antibody against TNFα (infliximab).[16] Inflamma-
`
`tory activity as measured by MRI, CSF lymphocytic pleocytosis
`and IgG index was clearly increased after receiving the infusions.
`After 2 to 3 weeks, values dropped back to their initial level; the
`Expanded Disability Status Scale (EDSS) was not altered.
`
`2.1.2 Lenercept
`In a phase II study [168 patients with mainly relapsing-re-
`mitting MS (RRMS)], the effect of the soluble TNF-receptor im-
`munoglobulin fusion protein lenercept on the development of
`new lesions in MRI was examined.[15] In this four-armed study,
`patients received 10, 50 or 100mg of the drug or placebo every 4
`weeks (up to 12 months). Baseline MRI was taken as a reference
`and followed up every 4 weeks (up to week 24 of the study).
`MRI showed no significant difference between lenercept and
`placebo (primary endpoint: cumulative number of new active le-
`sions). However, the number of clinical exacerbations was sig-
`nificantly higher in the lenercept group (annual relapse rate was
`0.98 with placebo vs 1.64 with lenercept 50mg; p = 0.007). In the
`
`© Adis International Limited. All rights reserved.
`
`Biodrugs 2002; 16 (3)
`
`Page 5 of 18
`
`

`

`188
`
`Wiendl & Hohlfeld
`
`Table II. Immunotherapeutic strategies based on putative immunepathological mechanisms in multiple sclerosis (see also figure 2)
`
`Process in immunopathological cascade
`
`Key mediators
`
`Therapeutic approaches
`
`Pre-existing autoreactive T cells
`Nonselective activation of T cells
`
`T cells
`
`Selective activation of myelin-reactive T cells
`
`Molecular mimicry?
`Superantigens?
`Other factors?
`
`Adhesion and penetration of T cells into the CNS
`Adherence and diapedesis
`Selectins (L-, E-, P-)
`Integrins (VLA-4, VLA-5, VLA-6, LFA-1, MAC-1,
`CR4)
`Ig superfamily (ICAM-1, ICAM-2, ICAM-3, VCAM,
`LFA-2, LFA-3)
`MMPs
`Cytokines
`
`Blood-brain barrier disruption
`
`Cellular CNS invasion
`
`Cell recruitment in the CNS
`
`? Infection (viral, bacterial)
`? Reactive metabolites
`
`MMPs
`TIMPs
`Integrins (VLA-4, VLA-5, VLA-6, LFA-1, MAC-1,
`CR4)
`Chemokines
`
`Local activation and proliferation of immune cells in the CNS
`APC-T cell interaction
`
`interaction of the trimolecular complex
`
`MHC class II
`TCR
`
` activation of ‘second signals’
`
`Costimulatory molecules (B7-1, B7-2,
`CD40-CD40-L, CTLA-4, other CD-molecules)
`
`Cytokine network
`
`Th1-predominance, Th1/Th2-dysbalance
`
`Soluble toxic mediators
`
`TNFα, IFNγ, lymphotoxin
`
`Cell-mediated damage
`
`MMPs/proteases
`CD4+, CD8+ T cells, macrophages, microglia
`
`Humorally mediated damage
`
`B cells
`
`Antibodies
`
`Complement system
`
`Bone marrow transplantation, extracorporeal
`photopheresis
`T cell vaccination
`T-cell receptor (TCR) peptide vaccination
`Antigen-derived therapies
`
`Selectin inhibitors, antiselectin MAbs
`Integrin/ligand inhibitors, anti-integrin MAbs
`
`Antiadhesion molecule MAbs
`
`MMP inhibitors, anti-MMP MAbs
`Cytokine modulators (anticytokine drugs,
`anticytokine MAbs, cytokines)
`Antiviral/antimicrobial therapies
`Free radical scavengers, nitric oxide synthase
`inhibitors
`MMP inhibitors, anti-MMP MAbs
`TIMP promoters
`Integrin/ligand inhibitors, anti-integrin MAbs
`
`Chemokine inhibitors, antichemokine MAbs,
`Cytokine modulators (anticytokine drugs,
`anticytokine MAbs, cytokines)
`
`MHC peptide vaccine, anti-MHC MAbs
`TCR peptide vaccination, TCR MAbs, T cell
`vaccination, antigen-derived therapies (e.g. altered
`peptide ligands)
`Anti-B7 MAbs CD40/CD40-L interaction, CTLA-4-Ig
`fusion protein, anti-T cell MAbs (e.g. CD2), T cell
`vaccines
`Anti-Th1 cytokines, Th1 cytokine inhibitors, Th2
`cytokines (IL-4, IL-10, TGFβ), Th2 cytokine
`promoters (e.g. glatiramer acetate), shift Th1 to Th2
`(e.g. glatiramer acetate, drugs, manipulation of
`costimulation, etc.)
`Cytokine modulators (anticytokine drugs,
`anticytokine-MAbs, cytokines)
`MMP inhibitors, anti-MMP MAbs, TIMP promoters
`Immunosuppression, cell-specific MAbs, autologous
`stem cell transplantation
`Immunosuppression, cell-specific MAbs, autologous
`stem cell transplantation
`Anti-autoantibody MAbs, intravenous
`immunoglobulins (IVIg), plasmapheresis
`Complement inhibitors, MAC (C5b-C9 membrane
`attack complex inhibitors), CD59, CD46, DAF
`
`© Adis International Limited. All rights reserved.
`
`Biodrugs 2002; 16 (3)
`
`Page 6 of 18
`
`

`

`Therapeutic Approaches in Multiple Sclerosis
`
`189
`
`Table II. Contd
`
`Process in immunopathological cascade
`Other damage mechanisms
`
`Key mediators
`Glutamate
`Reactive oxygen species
`Reactive nitrogen species
`Apoptosis
`Viral infection
`Microbial infection
`
`Therapeutic approaches
`AMPA/kainate inhibitors
`Free radical scavengers
`Nitric oxide synthase inhibitors
`Fas/Fas-L and other receptor modulations
`Antiviral therapies
`Antibiotics
`
`Demyelination and remyelination
`
`Survival and maturation of oligodendrocyte
`precursors
`Availability of oligodendrocyte precursors
`
`BDNF, GDNF, CNTF, IGF-1, leukaemia inhibitory
`factor, neurotrophin-3, PDGF
`Stem cell transplantation, precursor cell
`implantation, immortalised cell transplantation,
`Schwann cell transplantation, xenotransplantation
`AMPA = alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate; APC = antigen-presenting cell; BDNF = brain-derived neurotrophic factor; CD = cluster
`of differentiation; CNTF = ciliary neurotrophic factor; CR = complement receptor; CTLA = cytotoxic T lymphocyte-associated molecule; DAF =
`decay-accelerating factor; GDNF = glial cell-derived growth factor; ICAM = intercellular adhesion molecule; IFNγ = interferon-γ; Ig = immunoglobulin; IGF
`= insulin-like growth factor; IL = interleukin; LFA = lymphocyte function-associated antigen; MAb = monoclonal antibody; MAC = membrane attack complex;
`MHC = major histocompatibility complex; MMP = matrix metalloproteinase; PDGF = platelet-derived growth factor; TGF = transforming growth factor; Th
`= T helper cell; TIMP = tissue inhibitor of MMP; TNFα = tumour necrosis factor-α; VCAM = vascular cell adhesion molecule; VLA = very late antigen.
`
`group receiving lenercept, exacerbations occurred earlier (p =
`0.006), duration of relapses was longer, time until appearance of
`clinical exacerbations was shortened and neurological deficits
`appeared to be more serious (not significant; EDSS not altered).
`Adverse events such as headaches, nausea, abdominal pain or hot
`flushes occurred more frequently with lenercept. Antibodies
`against lenercept were generated in 88 to 100% of the patients,
`which did not interfere with neutralisation of TNF but accelerated
`elimination of the drug. A first evaluation after all patients were
`treated for 24 weeks led to early termination of the study.
`
`2.1.3 Commentary
`Blockage of TNFα, a putative ‘key cytokine’ in MS, failed
`in two trials. In contrast, anti-TNF strategies showed impressive
`beneficial effects in other T cell-mediated autoimmune diseases,
`particularly rheumatoid arthritis.[44-48] The unexpected and sur-
`prising negative results for infliximab and lenercept require care-
`ful analysis, particularly as they question present concepts of MS
`pathogenesis. The discrepancy between clinical exacerbations
`and MRI findings in the lenercept study is remarkable. Whereas
`the increase in clinical exacerbation rate was overt, MRI findings
`showed only a trend towards increased activity during therapy.
`Detection of new active lesions in MRI is assumed to be a highly
`sensitive indicator for disease activity (approximately ten new
`lesions correspond to about one clinical exacerbation). MRI as-
`sessment as a primary endpoint in MS studies therefore can help
`
`in rapidly gaining information but using lower patient numbers.[5]
`However, an exact correlation of ‘positive’ MRI findings with
`clinical parameters is not guaranteed. In the lenercept study the
`number of new lesions did not differ significantly from the lesion
`number in the placebo group. It is not clear whether MRI as the
`primary endpoint parameter for the efficacy assessment was
`wrong, or whether ‘technical reasons’ in the study protocol could
`account for the difference. The MRI scans were taken before each
`intravenous infusion; in other words, 4 weeks after the last infu-
`sion. In contrast, in the infliximab study, where the increased
`MRI activity appeared, examinations were conducted shortly af-
`ter the infusions of the antibodies. After 2 to 3 weeks, MRI ac-
`tivity dropped back to its initial level. Obviously lenercept pro-
`moted the formation of clinically relevant lesions without
`detectable correlates in MRI.
`In almost all patients antibodies against the TNF receptor
`construct lenercept were generated. Although they did not inhibit
`binding to TNF, they accelerated elimination and thereby may
`have shortened the duration of the drug effect. Anti-TNF antibod-
`ies that bind TNF may, per se, act as a ‘TNF sink’, which may
`then release TNF later and induce exacerbation of disease.
`Lenercept itself also possesses the Fc portion of IgG immuno-
`globulins. The Ig backbone may increase the longevity of the
`antibody in the circulation and increase Fc binding events. It is
`likely that Fc fragments stimulated the formation of immune
`complexes as well as the activation of Fc receptors on lympho-
`
`© Adis International Limited. All rights reserved.
`
`Biodrugs 2002; 16 (3)
`
`Page 7 of 18
`
`

`

`190
`
`Wiendl & Hohlfeld
`
`cytes. This effect could potentially be a trigger for inflammatory
`events. However, the use of Fab fragments or the extracellular
`portions of receptors linked by a polyglycine/serine linker may
`confer a short half-life and reduce peripheral adverse effects.
`Studies determining the efficacy of a therapeutic agent often
`study its effect prior to the onset of disease. Very few EAE animal
`studies target therapy after the onset or following the acute phase
`of disease during remission. In the case of TNF, a study deliver-
`ing a dimeric TNF receptor to mice with EAE demonstrated that
`the anti-TNF treatment was more efficacious when delivered
`prior to onset rather than during remission, although therapy at
`both timepoints could significantly inhibit disease sever-
`ity.[49]Although mechanisms for this were not demonstrated, it
`was suggested that the secondary relapse phase may be less de-
`pendent on TNF than the initial priming event. This is also sug-
`gested by TNF gene-deleted mice, in which the onset of disease
`was delayed compared with that in wild-type mice, but EAE de-
`veloped in the absence

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