throbber
Downloaded from
`
`http://brain.oxfordjournals.org/
`
` by guest on August 2, 2016
`
`doi:10.1093/brain/awl083
`
`Brain (2006), 129, 1940–1952
`
`R E V I E W A R T I C L E
`The value of animal models for drug development
`in multiple sclerosis
`
`Manuel A. Friese,1 Xavier Montalban,4 Nick Willcox,2 John I. Bell,1,3 Roland Martin4 and Lars Fugger1,5
`
`1MRC Human Immunology Unit and Department of Clinical Neurology, 2Neurosciences Group, Weatherall Institute of
`Molecular Medicine and 3Office of the Regius Professor, John Radcliffe Hospital, University of Oxford, Oxford, UK,
`4Edifici Escola D’infermeria, 2a Planta, Unitat de Neuroimmunologia Clı´nica, Hospital Universitari Vall d’Hebron, Barcelona,
`Spain and 5Department of Clinical Immunology, Aarhus University Hospital, Skejby Sygehus, Denmark
`
`Correspondence to: Lars Fugger, MRC Human Immunology Unit and Department of Clinical Neurology, Weatherall
`Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, United Kingdom or
`Roland Martin, Edifici Escola D’infermeria, 2a Planta, Unitat de Neuroimmunologia Clı´nica,
`Hospital Universitari Vall d’Hebron, Pg. Vall d’Hebron 119-129, 08035 Barcelona, Spain
`E-mail: lars.fugger@molecular-medicine.oxford.ac.uk
`
`The rodent model for multiple sclerosis, experimental allergic (autoimmune) encephalomyelitis (EAE), has been
`used to dissect molecular mechanisms of the autoimmune inflammatory response, and hence to devise and test
`new therapies for multiple sclerosis. Clearly, artificial immunization against myelin may not necessarily reproduce
`all the pathogenetic mechanisms operating in the human disease, but most therapies tested in multiple sclerosis
`patients are nevertheless based on concepts derived from studies in EAE. Unfortunately, several treatments, though
`successful in pre-clinical EAE trials, were either less effective in patients, worsened disease or caused unexpected,
`severe adverse events, as we review here. These discrepancies must, at least in part, be due to genetic and
`environmental differences, but the precise underlying reasons are not yet clear. Our understanding of EAE patho-
`genesis is still incomplete and so, therefore, are any implications for drug development in these models. Here, we
`suggest some potential explanations based on new thinking about key pathogenic concepts and differences that may
`limit extrapolation from EAE to multiple sclerosis. To try to circumvent these rodent–human dissimilarities more
`systematically, we propose that pre-clinical trials should be started in humanized mouse models.
`
`Keywords: animal models; experimental allergic encephalomyelitis; immunomodulation; multiple sclerosis; treatments
`
`Abbreviations: CFA = complete Freund’s adjuvant; EAE = experimental allergic (autoimmune) encephalomyelitis;
`HSCT = haematopoietic stem cell transplantation; MBP = myelin basic protein; PDEs = phosphodiesterases; PML = progressive
`multifocal leukoencephalopathy; PPARs = peroxisome proliferator-activated receptors; TCR = T-cell receptor;
`TH1 = T helper 1; TNF = tumour necrosis factor
`
`Received November 30, 2005. Revised March 5, 2006. Accepted March 14, 2006. Advance Access publication April 24, 2006
`
`Introduction
`Multiple sclerosis is the commonest neurological disease of
`young adults, afflicting at least 350 000 individuals in North
`America and 500 000 in Europe (Hafler et al., 2005; Sospedra
`and Martin, 2005). Although multiple sclerosis does not
`usually shorten life expectancy, its socio-economic burden
`in young adults is second only to trauma (Sospedra and
`Martin, 2005). Its clinical signs and symptoms are very
`variable and depend on the parts of the CNS it affects,
`
`that is, the brain and spinal cord, and include motor, sensory,
`autonomic and cognitive disabilities (Noseworthy et al.,
`2000a).
`It can run at
`least
`three clinical courses: (i)
`relapsing–remitting (RR) multiple sclerosis, which is most
`frequent (85%) and characterized by discrete attacks
`(exacerbations) and subsequent periods of clinical stability.
`In most relapsing multiple sclerosis patients, (ii) a secondary
`progressive (SP) phase ensues, with continuously increasing
`deficits. About 10–15% of multiple sclerosis patients develop
`
`# The Author (2006). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For Permissions, please email: journals.permissions@oxfordjournals.org
`
`Coalition Exhibit 1048
`Coalition v. Biogen
`IPR2015-01993
`
`Page 1 of 13
`
`

`

`Downloaded from
`
`http://brain.oxfordjournals.org/
`
` by guest on August 2, 2016
`
`The value of animal models for drug development in multiple sclerosis
`
`Brain (2006), 129, 1940–1952
`
`1941
`
`steadily increasing neurological deficits from onset, (iii) the
`primary-progressive subtype (Noseworthy et al., 2000a).
`Neuropathologically, CNS tissue from multiple sclerosis
`patients shows discrete lesions (predominantly in the white
`matter) with inflammatory infiltrates, demyelination, astro-
`gliosis and early axonal damage. Again, there is considerable
`heterogeneity in composition of cellular infiltrates and in
`involvement of antibodies and complement (Lassmann
`et al., 2001). Multiple sclerosis is widely considered an
`autoimmune demyelinating disease, and the inflammatory
`infiltrates as pathogenically primary events. Its aetiology
`remains a mystery, but infectious agents have long been
`suspected as triggers (Marrie, 2004). The evidence for an
`autoimmune reaction targeting myelin is strong but not
`definitive. There are, for example, descriptions of primary
`oligodendrocyte apoptosis with microglial activation in
`early multiple sclerosis lesions in the absence of lymphocytes
`or myelin phagocytosis (Barnett and Prineas, 2004). Further,
`the decreasing inflammatory activity that is seen by MRI during
`the SP phase has led to the assumption that the pathology is
`inflammatory at first and degenerative later. Despite these uncer-
`tainties, it is generally accepted that multiple sclerosis involves an
`autoimmune reaction by myelin-specific CD4+ T helper 1 (TH1)
`cells, which initiate the neuropathology (Hafler et al., 2005;
`Sospedra and Martin, 2005). This notion is based on the
`cellular composition of CNS- and CSF-infiltrating cells
`(Hauser et al., 1986), on genetic studies in multiple sclerosis
`(Dyment et al., 2004) and on one animal model of multiple
`sclerosis, experimental allergic (autoimmune) encephalomye-
`litis (EAE) (Zamvil and Steinman, 1990).
`Dissecting the pathogenesis of a complex disease in man is
`fraught with many problems, particularly those associated
`with clinical and genetic heterogeneity. Not surprisingly,
`most of our current
`thinking about multiple sclerosis
`stems from EAE. This model originated from vaccination
`with rabies-infected rabbit spinal cord by Louis Pasteur
`(from 1885). About 1 in 1000 vaccinees had ‘neuroparalytic
`incidents’; this acute demyelinating disorder later proved to
`be due to ‘contamination’ by spinal cord components in the
`inoculum. The EAE model has since evolved a long way;
`different variants, mice, rats or non-human primates are
`immunized with whole spinal cord, myelin proteins or
`even defined peptides, usually in complete Freund’s adjuvant
`(CFA). This immunization leads to a disease that shares clin-
`ical and neuropathological changes with multiple sclerosis
`(Steinman, 1999). The course it takes ranges from acute
`monophasic (or even lethal) to chronic progressive or
`relapsing–remitting (Steinman, 1999). Typical CD4+ TH1
`myelin-specific T cells have been implicated as
`the
`disease-initiating subset. In almost all models, they are suffi-
`cient to induce EAE; they can be isolated, cloned and used to
`transfer disease to naı¨ve healthy animals (Zamvil and
`Steinman, 1990). These various EAE models have been
`used to dissect molecular mechanisms of the autoimmune
`inflammatory response, and hence to devise and test new
`therapies for multiple sclerosis. It is clear, however, that
`
`the artificial induction of a myelin-specific immune response
`may by-pass key pathogenetic mechanisms operating in
`human disease, as we do not even know the key target auto-
`antigens in multiple sclerosis.
`
`Limitations of current EAE models
`Without doubt, EAE models are vital for studying general
`concepts as well as specific processes of autoimmunity, how-
`ever rarely they predict success in clinical trials (see below).
`Nevertheless, their value is further challenged by our rudi-
`mentary understanding of the key pathogenetic mechanisms
`in EAE models, and their failure to forewarn us of adverse
`effects (reviewed below). As with other murine disease
`models,
`including the NOD model of
`type 1 diabetes
`(Roep et al., 2004), it appears much easier to prevent, reverse
`or ameliorate EAE in mice than multiple sclerosis in man.
`Furthermore, since EAE almost always has to be induced, it
`cannot mimic a spontaneous disease. The most important
`component in the inducing adjuvant CFA is heat-inactivated
`Mycobacterium tuberculosis, which always induces a promi-
`nent CD4+ TH1 response by activating certain toll-like recep-
`tors (Su et al., 2005). This leaves little room for variability in
`disease pathways and certainly does not reflect heterogeneous
`inducing mechanisms in multiple sclerosis. Also, demyelina-
`tion is not obvious in all models. Moreover, the time courses
`are very different. Since EAE develops over days in most
`models, they seem more similar to post-infectious acute
`demyelinating events (Steinman, 1999). Indeed, the mice
`are rarely monitored for late relapses and fatal adverse effects,
`such as those noted in marmosets (Genain et al., 1996).
`Nevertheless, the same treatment can have a different degree
`of efficacy or even opposite effects at different stages in EAE,
`as has also been reported for other autoimmune models such
`as in NOD mice (Shoda et al., 2005). In contrast, multiple
`sclerosis usually manifests insidiously over years, for example,
`in its
`relapsing–remitting
`and later
`chronic
`forms
`(Noseworthy et al., 2000a), by when antibodies and comple-
`ment may also be more important than in most mouse mod-
`els.
`Indeed, many patients present after much more
`protracted epitope spreading than is usually seen in EAE
`mice (Vanderlugt and Miller, 2002). These and other obvious
`mouse : human differences are summarized in Table 1.
`Many aspects of pathology and immunology differ between
`multiple sclerosis and EAE. These differences are fundamen-
`tal, as ongoing imbalances in immune regulation must be
`crucial for the progression of multiple sclerosis; such orders
`of complexity have not yet been recapitulated in EAE models.
`
`What can we learn from failures or
`successes in adapting therapies from
`EAE to multiple sclerosis?
`Only very few therapeutics that were successful in pre-clinical
`EAE trials have shown similar efficacy in multiple sclerosis
`
`Page 2 of 13
`
`

`

`1942
`
`Brain (2006), 129, 1940–1952
`
`M. A. Friese et al.
`
`Table 1 Immunological differences between mouse and human relevant for testing multiple sclerosis therapeutics
`
`Mouse
`
`Human
`
`References
`
`General
`
`Inbred; homozygous
`Short lifespan: high fecundity
`Fixed diet; pathogen-free
`
`Clean environment
`
`Outbred; heterozygous
`Long lifespan: low fecundity
`Varied diet; carriers of potential pathogens,
`e.g. EBV, JCV etc
`Open access to new infections
`
`EAE and multiple sclerosis
`
`Induction
`Testing new therapeutics
`
`May be monophasic
`Mice tested while epitopes are
`spreading
`Usually with CFA
`Induction of EAE studied much
`more than ongoing disease
`Only a few dozen mice tested
`
`Less detailed
`Scrutiny
`Often short-term only
`Follow-up
`Molecular differences in immune response
`T-cell responses
`Often stereotypical
`
`75–90%
`
`Different subtypes, usually relapsing
`Epitopes must often have spread long
`before diagnosis
`Spontaneous
`Ongoing disease
`
`Hundreds of multiple sclerosis patients;
`some side-effects are too rare to be seen
`in mice
`Detailed, would be missed in mice
`Several years or life-long
`
`Usually idiosyncratic, even to
`recurring epitope(s)
`30–50%
`
`Lymphocytes in
`peripheral blood
`CD4+ expression
`CD8+ expression
`IL-10 expression
`IFN-a response
`IL-4 and IFN-g
`expression by TH
`CD28 expression
`MHC class II expression
`
`CD52 expression
`Glucocorticoid-sensitivity
`
`Lymphocytes
`Lymphocytes, dendritic cells
`TH2
`No preferential TH differentiation
`Exclusively one or the other
`100% of CD4+ and CD8+ T cells
`Absent on T cells and
`endothelial cells
`Not found in mice
`High
`
`Lymphocytes, macrophages
`Lymphocytes
`TH1 and TH2
`Promotes TH1 response
`Sometimes both
`80% of CD4+ T cells, 50% of CD8+ T cells
`Present on T cells and
`endothelial cells
`Lymphocytes
`Low and variable
`
`Downloaded from
`
`http://brain.oxfordjournals.org/
`
` by guest on August 2, 2016
`
`Vanderlugt and Miller
`(2002)
`
`Doeing et al. (2003)
`
`Crocker et al. (1987)
`Banchereau et al. (2000)
`Del Prete et al. (1993)
`Farrar et al. (2000)
`Gor et al. (2003)
`
`Lenschow et al. (1996)
`Choo et al. (1997),
`Taams et al. (1999)
`Tone et al. (1999)
`Claman (1972)
`
`patients; the majority of new treatments were either less
`effective in these patients, worsened disease or caused severe
`adverse events. In Table 2 we list a subset of these therapies
`reflecting this discrepancy.
`
`Antigen-specific therapies
`Only one licensed multiple sclerosis therapy (Glatiramer
`acetate, GA), a synthetic amino acid copolymer (Glu, Ala,
`Lys and Tyr), emerged from findings in EAE (Teitelbaum
`et al., 1971). It was designed to mimic encephalitogenic mye-
`lin basic protein (MBP) epitopes, but instead it suppresses
`EAE by other mechanisms in several species, and it reportedly
`reduces multiple sclerosis relapses by 30% (Johnson et al.,
`1995). GA has many biological activities including bystander
`suppression via induction of TH2 cells that partly cross-react
`with MBP, and/or upregulation of CNS growth factors
`(Arnon and Aharoni, 2004). However, its in vivo mechanisms
`are not clear and even its beneficial effects on the main out-
`come measures in multiple sclerosis (disease progression)
`have now been questioned in a systematic Cochrane review
`(Munari et al., 2004).
`A more specific therapeutic approach in EAE and multiple
`sclerosis has been based on an altered peptide ligand of MBP
`
`85–99 that was modified at its main T-cell receptor (TCR)
`contact sites (Brocke et al., 1996). Despite promising effects
`in EAE, subcutaneous administration of altered peptide
`ligand at high doses led to multiple sclerosis exacerbations
`in some patients, which could be linked to this treatment
`(Bielekova et al., 2000). A trend towards improved MRI
`parameters was observed in another phase II trial (Kappos
`et al., 2000), and an additional phase II study is under way. Its
`success in EAE may depend on the stereotyped TH responses
`of inbred mice.
`Oral administration of myelin antigens leads to specific
`immune hyporesponsiveness in mice. Different doses and
`feeding regimes have been demonstrated to induce different
`types of ‘oral tolerance’/degrees of immune suppression in
`different EAE models (Faria and Weiner, 2005). ‘Bystander
`suppression’ directed against one tolerogen may suppress
`reactions against other myelin antigens in situ, a major advan-
`tage where the key autoimmunizing antigen(s) are not
`known. However, a large double-blind phase III trial of a
`single oral dose of bovine myelin in RR multiple sclerosis
`did not show differences in the number of relapses between
`placebo and treated groups (Faria and Weiner, 2005). Treat-
`ment failure could have been due to the unexpectedly strong
`
`Page 3 of 13
`
`

`

`The value of animal models for drug development in multiple sclerosis
`
`Brain (2006), 129, 1940–1952
`
`1943
`
`Table 2 Some immunomodulatory approaches of multiple sclerosis and their development from EAE or in vitro studies to
`clinical application*
`
`Treatment
`approach
`
`Based on
`clear
`hypothesis
`
`Rationale
`confirmed
`
`Efficacy
`in EAE
`
`Efficacy
`in multiple
`sclerosis
`
`Adverse
`event
`profile
`
`Status of
`development
`
`Reference
`
`Downloaded from
`
`http://brain.oxfordjournals.org/
`
` by guest on August 2, 2016
`
`Approved
`No
`
`Johnson et al. (1995)
`Bielekova et al. (2000),Kappos
`et al. (2000)
`Faria and Weiner (2005)
`
`Not continued after phase III
`owing to lack of efficacy
`Miller et al. (2003)
`Taken off the market
`Dumont (2002)
`No
`van Oosten et al. (1997)
`Halted in phase II
`Approved for other indication Coles et al. (1999b)
`Approved for other indication Bielekova et al. (2004)
`In phase III
`Kremer (2004)
`Approved
`Paty and Li (1993)
`Stopped in phase I
`Panitch et al., 1987)
`Approved for other indication van Oosten et al. (1996)
`
`Approved for other indication The Lenercept Multiple Sclerosis
`Study Group and The University
`of British Columbia multiple
`sclerosis/MRI Analysis Group
`(1999)
`Calabresi et al. (1998)
`Wiendl et al. (2000)
`Frank et al. (2002)
`R. Martin et al. (unpublished data)
`Diab et al. (2002), Feinstein et al.
`(2002)
`Vollmer et al. (2004), Youssef
`et al. (2002)
`Hartung et al. (2002)
`Noseworthy et al. (2000c)
`
`Stopped in phase I
`Stopped in phase II
`Phase IIa, not continued
`Halted in phase II
`Not yet tested in multiple
`sclerosis
`Approved for other
`indication
`Approved
`Phase III stopped due to
`cardiotoxicity
`In phase II
`In phase II
`
`Polman et al. (2005)
`Gonsette (2004), Rausch et al.,
`2004)
`Wiendl and Hohlfeld (2002)
`
`Noseworthy et al. (1998)
`Hommes et al. (2004), Sorensen
`et al. (2002)
`Mancardi et al. (2005), Tyndall
`and Saccardi (2005)
`
`++
`++
`
`++; i.d.
`+
`++(+); i.d. +
`
`
`+
`
`6
`6
`
`+
`++
`
`After phase II stopped
`owing to lack of efficacy
`In phase III
`In phase II
`
`++;i.d.
`
`6; i.d.
`
`In phase III
`
`+++
`6
`
`+++
`
`6†
`6
`6
`+
`++
`n.k.
`++
`
`‡
` ‡
`
`
`i.d.
`++
`+
`n.a.
`
`+
`
`6
`6
`
`No
`No; i.d.
`
`Yes
`
`Yes
`Yes
`No
`Yes
`No
`No; i.d.
`No
`No
`No
`
`No
`
`++
`++
`
`++
`
`+
`; i.d.
`
`
`+++
`+++
`n.k.
`+++
`6
`+++
`n.a.
`++; i.d.
`6
`+++; i.d.
`n.k.
`+++
`+
`+
`
`++
`++(?)
`++(?)
`
`++
`6
`+(+)
`++
`++
`
`i.d.
`i.d.
`; i.d.
`; i.d.
`n.t.
`
`+++
`
`++; i.d.
`
`Yes
`No
`Yes
`Yes
`Yes
`
`Yes
`
`No§
`No
`
`No
`No
`
`No
`
`No
`No
`
`Yes
`
`++
`++
`
`++
`+++
`
`+
`
`6
`6
`
`+
`
`Glatiramer acetate No
`Altered peptide
`Yes
`ligand
`Oral myelin
`
`Yes
`
`Anti-a4 integrin
`Anti-CD40L
`Anti-CD4
`Anti-CD52
`Anti-CD25
`CTLA-4-Ig
`IFN-b
`IFN-g
`Anti-TNF
`antibodies
`TNFR-Ig fusion
`protein
`
`TGF-b2
`IL-10
`IGF-1
`PDE4 inhibitors
`PPARg agonists
`
`Statins
`
`Mitoxantrone
`Linomide
`
`Laquinimod
`FTY720/SP-1
`agonist
`Deoxyspergualin
`
`Sulphasalazine
`IVIG
`
`Haematopoietic
`stem cell transplant
`
`Yes
`Yes
`Yes
`Yes
`Yes
`Yes
`No
`No
`Yes
`
`Yes
`
`Yes
`Yes
`Yes
`Yes
`Yes
`
`Yes
`
`No§
`No
`
`No
`No
`
`No
`
`Yes
`No
`
`Yes
`
`i.d., insufficient data; n.a., not applicable; n.k., not known; n.t., not tested; *The table depicts whether a therapeutic approach was developed
`for multiple sclerosis on the basis of a clear and pre-formed hypothesis, whether the rationale for its clinical/EAE testing had later been
`shown and whether the therapy was effective in EAE and/or multiple sclerosis. Both the clinical efficacy and the tolerability and safety are
`depicted by + or signs. In the context of the adverse events, + indicates a favourable profile. The relative weighting reflects the subjective
`perception of the authors either from own experience or the published literature; †Reasonable safety profile, but one specific severe
`adverse event (PML). ‡Development of demyelinating episodes and diseases in RA- and Crohn’s patients; §Broad immunosuppressant; no
`specific target.
`
`effects in the placebo group, wrong dose or type of antigen, or
`route of administration.
`
`Adhesion molecules
`Another promising strategy, using a blocking anti-a4 integrin
`humanized antibody (natalizumab), emerged from EAE
`evidence that a4b1 integrin is critical for T cell and monocyte
`
`homing to the CNS (Yednock et al., 1992). This mAb was
`highly effective in pre-clinical EAE studies and successfully
`completed phase II and III testing in large numbers of multi-
`ple sclerosis patients. Because of its remarkable efficacy in
`multiple sclerosis (Miller et al., 2003), natalizumab was
`approved by the Food and Drug Administration even before
`phase III trial data had been published, but was taken off the
`market four months later because of rare but very severe
`
`Page 4 of 13
`
`

`

`Downloaded from
`
`http://brain.oxfordjournals.org/
`
` by guest on August 2, 2016
`
`1944
`
`Brain (2006), 129, 1940–1952
`
`M. A. Friese et al.
`
`adverse events. Three patients had developed progressive
`multifocal
`leukoencephalopathy (PML), an often lethal
`opportunistic infection of the CNS; two died, and one is
`recovering, though with considerable neurological deficits
`(Kleinschmidt-DeMasters and Tyler, 2005; Langer-Gould
`et al., 2005; Van Assche et al., 2005). A large post facto analysis
`estimated the risk of PML for a 2-year treatment period to
`1 in 1000 patients (Yousry et al., 2006). PML is caused by
`reactivation and mutation of the highly prevalent polyoma-
`virus JC (JCV), which destroys oligodendrocytes. PML is
`almost exclusively observed in immunosuppressed indivi-
`duals, and it is not clear what initiated its unexpected deve-
`lopment under natalizumab treatment.
`JCV persists
`in
`kidneys and lymphoid organs,
`including bone marrow
`(Monaco et al., 1998). During immunosuppression, latent
`infection can be reactivated, and JCV disseminates to the
`CNS (Tornatore et al., 1992). That might have resulted either
`from compromised T-cell surveillance of the CNS or from
`mobilization of stem cells and JCV from the bone marrow
`(Papayannopoulou and Nakamoto, 1993; Ransohoff, 2005),
`where a4b1 integrin serves as a retaining signal (Simmons
`et al., 1992). Since JCV is not found in rodents, this adverse
`event could not have been anticipated from pre-clinical inves-
`tigations. Therefore, this drug cannot be called a failure of
`prediction, especially as many thousand patients needed to be
`treated to unravel potential adverse effects. In addition, the
`recently published two-year phase III trials underline its com-
`pelling effects on relapse rate and clinical progression
`(Polman et al., 2006; Rudick et al., 2006). In March 2006,
`The Peripheral and CNS Drugs Advisory Committee, under
`The Food and Drug Administration, voted unanimously to
`recommend the return of natalizumab for the treatment of
`RR multiple sclerosis in a subset group of patients.
`
`Co-stimulatory molecules
`Despite its promise in EAE, anti-CD40 ligand (CD154)
`(Howard et al., 1999) was not developed because of its throm-
`boembolic complications in man (Kawai et al., 2000), which
`result from its expression on human but not murine platelets.
`Anti-CD4 therapy was effective in EAE (Waldor et al., 1985),
`but not
`in human studies (van Oosten et al., 1997).
`Anti-CD52, which depletes both CD8+ and CD4+ T-cells
`(Coles et al., 1999b), was never evaluated in EAE, but is
`very effective against new lesions in multiple sclerosis, though
`30% of treated multiple sclerosis patients develop auto-
`immune hyperthyroidism (Coles et al., 1999a). On the
`other hand, IL-2 receptor blockade with the humanized
`anti-CD25 antibody (daclizumab) caused impressive reduc-
`tions in MRI lesions and improvements in some clinical
`measures (Bielekova et al., 2004). In this case, the theoretical
`role of CD25 in promoting T regulatory cells, and equivocal
`EAE data (Engelhardt et al., 1989; Reddy et al., 2004), might
`have argued against its use in multiple sclerosis. Interestingly,
`there is little evidence that it perturbs T regulatory or TH
`function; indeed it may act by expanding immunoregulatory
`
`NK cells (Bielekova et al., in review). CTLA-4-Ig interferes
`with co-stimulation from CD80/CD86 molecules on antigen-
`presenting cells (APCs) to the stimulatory or inhibitory
`ligands CD28 and CTLA-4 (Alegre et al., 2001). Data in
`EAE indicate that CTLA-4-Ig is much more effective as a
`preventive pre-treatment (Cross et al., 1995) than in therapy
`of ongoing disease (Cross et al., 1999). Treatment with
`CTLA-4-Ig is also effective in other autoimmune diseases
`such as rheumatoid arthritis (Kremer et al., 2003), and is
`currently being tested in a phase III trial in multiple sclerosis.
`
`Cytokines
`Cytokines have different effects at different stages of patho-
`genesis, for example, in the induction phase and the chronic/
`relapsing phase in EAE. These differences suggest a pleiotro-
`pic role in CNS inflammation and might explain some of the
`below-described discrepancies between EAE and multiple
`sclerosis.
`Interferon-b ( IFN-b), the first drug approved for multiple
`sclerosis, had not been previously tested in EAE. It exerts a
`wide variety of effects on the immune system: it inhibits both
`leukocyte proliferation and antigen presentation; it biases
`towards production of anti-inflammatory cytokines and it
`inhibits T-cell migration across the blood-brain barrier
`(Billiau et al., 2004). Although widely used in multiple sclero-
`sis, its long-term effectiveness and side-effects are still uncer-
`tain (Filippini et al., 2003). With other cytokines, effects have
`seemed contradictory in EAE vis a` vis multiple sclerosis. In
`it was found that IFN-g knockout mice
`the mid-1990s,
`develop lethal EAE (Ferber et al., 1996), and IFN-g admin-
`istration in EAE showed a protective effect on disease severity
`(Krakowski and Owens, 1996). By then, its use in multiple
`sclerosis patients had already led to a modest increase in
`disease exacerbations (Panitch et al., 1987). Although this
`study is limited, it is unlikely that IFN-g will ever be tested
`again in multiple sclerosis.
`In contrast, tumour necrosis factor-a (TNF-a) has long
`been considered a key mediator of multiple sclerosis patho-
`genesis (Sharief and Hentges, 1991), and its blockade by
`antibodies or soluble TNF receptors prevents or reverses dis-
`ease in EAE models (Ruddle et al., 1990; Selmaj et al., 1991,
`1995). Paradoxically, this approach worsens disease in multi-
`ple sclerosis patients and had to be discontinued (The Lener-
`cept Multiple Sclerosis Study Group and The University of
`British Columbia Multiple Sclerosis/MRI Analysis Group,
`1999; van Oosten et al., 1996). Indeed, a substantial number
`of cases developed their first demyelinating event while being
`treated with anti-TNF-a agents for other diseases such as
`rheumatoid arthritis or Crohn’s disease (Hyrich et al.,
`2004). Despite the data that TNF-a is an important compo-
`nent in the pathogenesis in EAE, a precise role for TNF-a in
`multiple sclerosis remains unclear. However, subsequent EAE
`/
`experiments using TNF-a gene deleted mice (TNF-a
`)
`surprisingly showed that TNF-a/
`mice displayed profound
`neurological impairment and high mortality with extensive
`
`Page 5 of 13
`
`

`

`Downloaded from
`
`http://brain.oxfordjournals.org/
`
` by guest on August 2, 2016
`
`The value of animal models for drug development in multiple sclerosis
`
`Brain (2006), 129, 1940–1952
`
`1945
`
`demyelination and monocytic cell infiltration in comparison
`with control mice (Liu et al., 1998). Conversely, treatment of
`the TNF-a/
`mice but also wild-type mice with recombi-
`nant TNF-a reduced disease severity in afflicted mice and
`prevented development of EAE in pre-treated mice. These
`studies suggested that TNF-a may be protective in the
`CNS during the development of demyelinating disease also
`in mice and may serve to limit the extent of immune-
`mediated inflammation, as was implicated by the study in
`multiple sclerosis patients. Further, we can only speculate on
`the reasons for the adverse outcome of TNF-blocking
`approaches in multiple sclerosis, but it has been demon-
`strated that TNF signalling is also important for remyelina-
`tion (Arnett et al., 2001; Diemel et al., 2004).
`Transforming growth factor (TGF)-b2 ameliorates EAE
`and is a very potent immunosuppressive cytokine. However,
`it has never been approved as a treatment in multiple sclero-
`sis, as it is associated with nephrotoxicity in multiple sclerosis
`patients, which was not checked in EAE (Calabresi et al.,
`1998). Interleukin (IL)-10 is another important suppressive
`cytokine, produced mainly by regulatory CD4+ T cells
`(O’Garra et al., 2004). Its selective upregulation in the
`CNS during the recovery phase of EAE prompted the evalua-
`tion of IL-10 treatment in a Lewis rat EAE model. Systemic
`administration during the initiation phase suppressed EAE
`disease (Rott et al., 1994). Again IL-10 treatment in a phase II
`clinical trial in multiple sclerosis patients had to be stopped,
`owing to lack of efficacy (Wiendl et al., 2000).
`
`Neurotrophic factors
`Apart from cytokines, several neurotrophic factors have been
`proposed as a novel therapeutic in multiple sclerosis. These
`proteins regulate survival and differentiation of neurons by
`binding to specific neurotrophin receptors (Thoenen and
`Sendtner, 2002). They are involved in skewing the cytokine
`balance in the CNS from TH1 to TH2 responses (Villoslada
`et al., 2000), and neuroprotective effects have been proposed
`(Hohlfeld et al., 2005). The clinical potential of one such
`factor, insulin-like growth factor-1 (IGF-1), was first studied
`in a rat EAE model, in which myelin regeneration and clinical
`recovery was enhanced (Yao et al., 1995). In contrast, there
`was no clear effect in one phase-I/II study in seven patients
`(Frank et al., 2002).
`
`Anti-inflammatory substances,
`immunosuppression and
`immunomodulation
`Cytokine secretion and proliferation of T cells are regulated
`by intracellular cyclic AMP (cAMP) levels, which are reduced
`by phosphodiesterases (PDEs). The type 4 PDE (PDE4) is a
`cAMP-specific phosphodiesterase expressed in cells of the
`immune system and the CNS (Engels et al., 1994). Inhibition
`of PDE4 activity by rolipram ameliorates EAE severity
`(Sommer et al., 1995). It was originally developed and
`
`evaluated in clinical studies as an anti-depressant (Zeller
`et al., 1984). Since rolipram and other PDE4 inhibitors
`(Dinter et al., 2000) demonstrated efficacy in various EAE
`models, it seemed a promising candidate for clinical use in
`multiple sclerosis patients. However, a phase II clinical trial
`was halted owing to lack of efficacy on the primary outcome
`measure, that is, the reduction of gadolinium-enhancing
`inflammatory CNS lesions by MRI (Martin, R., Bielekova, B.,
`Stu¨rzebecher, C.S., Richert, N., Frank, J.A., Ohayon, J.,
`McCartin, J., McFarland, H.F., unpublished data).
`Peroxisome proliferator-activated receptors (PPARs) are
`members of the nuclear hormone receptor superfamily of
`ligand-activated transcriptional factors that include receptors
`for steroids, thyroid hormone, vitamin D and retinoic acid
`(Mangelsdorf et al., 1995). PPAR-g is expressed in adipose
`tissue, on macrophages, T cells, and endothelial and vascular
`smooth muscle cells. The natural 15-deoxy-D12,14-PGJ2
`(15d-PGJ2) and the synthetic anti-diabetic thiazolidinedione
`are PPAR-g ligands; their administration before and at the
`onset of clinical signs of EAE significantly reduced its severity
`(Diab et al., 2002). Another orally administered PPAR-g
`agonist pioglitazone reduced the incidence and severity in
`C57BL/6 EAE and B10.Pl murine models. Pioglitazone also
`reduced clinical signs when given after disease onset (Feinstein
`et al., 2002). So far there is only casuistic information about
`PPAR-g agonists in multiple sclerosis (Pershadsingh et al.,
`2004), but clinical trials are being planned.
`Statins block the enzyme 3-hydroxy-3-methylglutaryl-
`coenzyme A reductase; they are widely used to lower choles-
`terol
`levels and prevent cardiovascular disease. They are
`highly
`effective
`in reversing EAE,
`shifting the pro-
`inflammatory TH1-type cytokine profile to a TH2-type
`pattern and also interfering with antigen presentation
`(Youssef et al., 2002). In this case, exploratory trials in multi-
`ple sclerosis patients have shown promising results with up to
`44% reduction in the number of contrast-enhancing MRI
`lesions (Vollmer et al., 2004), so they are a solitary success.
`Mitoxantrone is a potent anti-inflammatory cytostatic
`anthracenedione, and suppresses both B and T lymphocytes
`and macrophages, resulting in down-modulation of the
`inflammatory cascade. Since it had already been approved
`for the therapy of malignancies, this immunosuppressant
`could logically have been assessed in multiple sclerosis with-
`out studying EAE. Not surprisingly, mitoxantrone has potent
`effects in EAE. It suppressed paralysis in acute EAE, prevented
`its development when administered during the induction
`period and still showed some effects when given after clinical
`signs and symptoms appeared (Ridge et al., 1985; Levine and
`Saltzman, 1986). These results parallelled recent proof of
`efficacy in multiple sclerosis (Hartung et al., 2002). However,
`cardiotoxicity was a reported adverse effect in humans;
`though not observed in the early EAE studies (Ridge et al.,
`1985; Levine and Saltzman, 1986), it was also detected in
`careful retrospective analysis in treated mice. Meanwhile,
`the more serious problem of inducing haematopoietic malig-
`nancies is predictable for a cytotoxic agent, and has already
`
`Page 6 of 13
`
`

`

`Downloaded from
`
`http://brain.oxfordjournals.org/
`
` by guest on August 2, 2016
`
`1946
`
`Brain (2006), 129, 1940–1952
`
`M. A.

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