throbber
POINT OF VIEW
`
`How to Successfully Apply Animal Studies
`in Experimental Allergic Encephalomyelitis
`to Research on Multiple Sclerosis
`
`Lawrence Steinman, MD,1 and Scott S. Zamvil, MD, PhD2
`
`In their Point of View entitled “Experimental Allergic Encephalomyelitis: A Misleading Model of Multiple Sclerosis,” Sriram and
`Steiner1 wrote, “The most disappointing aspect of EAE [experimental allergic encephalomyelitis] as a potential model for MS is
`its almost total inability to point toward a meaningful therapy or therapeutic approach for MS.” Actually, EAE has led directly
`to the development of three therapies approved for use in multiple sclerosis (MS): glatiramer acetate, mitoxantrone, and natali-
`zumab. Several new approaches to MS are in clinical trials based on positive indications in preclinical work relying on EAE. New
`clues to the pathogenesis of MS and new potential surrogate markers for MS are shown from research involving EAE when it
`is critically coupled with actual findings in MS. There are pitfalls in overreliance on the EAE model, or on any animal model
`for any human disease. Nevertheless, over the past 73 years, the EAE model has proved itself remarkably useful for aiding
`research on MS.
`
`Any discussion of the pros and cons of the animal
`models of multiple sclerosis (MS), collectively known
`as experimental allergic encephalomyelitis (EAE), must
`address our present state of knowledge about MS. MS
`is a complicated disease, the cause and pathogenesis of
`which are incompletely understood. Though we have
`made progress in therapy of MS, treatment is imper-
`fect. Current therapies reduce the frequency of relapse,
`somewhere between 33 and 66%, and delay disease
`progression to a modest extent in relapsing-remitting
`and secondary progressive MS.2 There is no single test
`we can run to determine whether someone has “MS,”
`and there is no surrogate marker for us to measure to
`assess whether MS is worsening. Whether MS is actu-
`ally a single disease or whether it is primarily or ini-
`tially an “immune disease,” “an infectious disease,” “an
`inflammatory disease,” or a “degenerative disease,” or a
`combination of all of these types are all questions with
`answers that are currently unknown. A few genetic fac-
`tors have been associated with MS, most prominently
`genes
`of
`the major histocompatibility
`complex
`(MHC).3 A genetic basis for MS is clearly only part of
`the story because concordance in identical twins is less
`than even 50%. Many environmental factors have been
`associated with MS, although none can be considered
`definitively linked. Therefore, set in this context in
`
`From the 1Department of Neurology and Neurological Sciences,
`Stanford University, Stanford, CA; and 2Department of Neurology,
`University of California San Francisco, San Francisco, CA.
`Received May 2, 2006. Accepted for publication May 9, 2006.
`Published
`online
`June
`26,
`2006
`in Wiley
`InterScience
`(www.interscience.wiley.com). DOI: 10.1002/ana.20913
`
`Ann Neurol 2006;60:12–21
`
`which nearly all of the major questions about human
`MS remain unanswered, this critique addresses how
`our understanding of MS has been aided by studies on
`a collection of animal models known as EAE, first de-
`scribed almost three quarters of a century ago. Given
`all these uncertainties about MS, it is remarkable that
`studies on EAE have culminated thus far in three MS
`therapies and have led to a better understanding of the
`biology of MS. Clever applications of the EAE model
`will be a valuable tool for understanding the pathology
`of MS, for making better biomarkers for its diagnosis
`and prognosis, and for creating ever improved and safe
`therapies for this disease. To study a disease such as
`MS, without support from available animal models, is
`to unnecessarily create obstacles in a task that is com-
`plicated enough.
`
`A Brief History of Experimental Autoimmune
`Encephalomyelitis
`In the 1930s, workers at Rockefeller University discov-
`ered an animal model, now known as EAE.4 The first
`experiments were aimed at understanding episodes of
`paralysis that sometimes accompany vaccination. Three
`years ago, on the 70th anniversary of the first publica-
`tion on EAE, we wrote in the Journal of Experimental
`Medicine of Rockefeller University:
`
`Address correspondence to Dr Steinman, Beckman Center for Mo-
`lecular Medicine, Stanford University, Stanford, CA 94305.
`E-mail: steinman@stanford.edu
`
`12 © 2006 American Neurological Association
`Published by Wiley-Liss, Inc., through Wiley Subscription Services
`
`Apotex v. Novartis
`IPR2017-00854
`NOVARTIS 2051
`
`

`

`One of the most enduring models of human disease
`now celebrates the seventieth anniversary of its publi-
`cation in The
`Journal
`of Experimental Medicine.
`Thomas Rivers, working at the Hospital of the Rock-
`efeller Institute for Medical Research, along with his
`colleagues D.H. Sprunt and G.P. Berry, submitted the
`article entitled, “Observations on Attempts to Produce
`Disseminated Encephalomyelitis in Monkeys,” on Feb.
`21, 1933 (4). Rivers established this model to try to
`understand what caused neurological reactions to cer-
`tain viral infections like smallpox and in some circum-
`stances to vaccinations like rabies: the very first sen-
`tence
`of
`this
`landmark
`paper
`reads,
`“During
`convalescence from certain diseases notably smallpox,
`vaccinia and measles, and during or following vaccina-
`tion against
`rabies, an occasional patient develops
`symptoms and signs referable to the central nervous
`system.”5
`Thus, the EAE model was initially constructed to un-
`derstand acute disseminated encephalomyelitis, not
`MS. Acute paralysis was observed in the first reported
`models with inflammatory changes in the central ner-
`vous system. Later versions of more chronic EAE have
`been developed with pathology including demyelina-
`tion and axonal damage and clinical events such as re-
`lapsing and remitting episodes of paralysis,6 all of
`which are features common to MS. We must remem-
`ber then that EAE is a collection of various models
`reflecting features of acute disseminated encephalomy-
`elitis, as well as MS. The relation between acute dis-
`seminated encephalomyelitis
`and MS remains
`an
`enigma itself.
`Many refinements and variations have been devel-
`oped in the past 75 years. Even the name EAE has
`evolved from experimental allergic encephalomyelitis to
`experimental autoimmune encephalomyelitis. Research-
`ers have developed numerous variations of EAE includ-
`for optic neuritis,7 relapsing-remitting
`ing models
`MS,8 –10 and progressive MS.9 Some of these models
`reflect certain aspects of the pathology seen in MS in-
`cluding axonal degeneration together with demyelina-
`tion.10 Researchers have constructed EAE models with
`essential genes,
`such as human leukocyte antigen
`(HLA) DR2 associated with susceptibility to MS, in-
`stalled into mice as transgenes.11,12 Others have de-
`vised forms of EAE in nonhuman primates such as the
`marmoset that reflect essential aspects of the pathology
`of MS with high fidelity.13 Numerous mouse versions
`of EAE exist where important components of the im-
`mune system have been “knocked out” by homologous
`recombination.14 Thus, there is no single model of
`EAE that we refer to in this critique, rather, it is the
`ensemble of EAE models, which have been reported in
`more than 5,000 publications since 1933.
`
`Experimental Autoimmune Encephalomyelitis for the
`Development of Approved Therapies of Multiple
`Sclerosis: Three Case Studies
`Sriram and Steiner1 wrote, “The most disappointing
`aspect of EAE as a potential model for MS is its almost
`total inability to point toward a meaningful therapy or
`therapeutic approach for MS.” We take a position
`nearly diametrically opposite to that perspective: In-
`deed, six medications have received approval from the
`US Food and Drug Administration for treatment of
`MS, and three of them, glatiramer acetate, mitox-
`antrone and natalizumab, were developed after showing
`promise in EAE. Moreover, glatiramer acetate and na-
`talizumab were invented after a set of
`logical and
`forward-looking experiments in the EAE model, which
`elucidated key targets in the pathogenesis of MS. Here,
`we review how experiments in EAE led to the devel-
`opment of three approved drugs for MS and how the
`model has been useful in helping us to understand the
`disease. Approved therapies that have been developed
`with the EAE model and new targets of interest devel-
`oped using the EAE model are shown in the Figure.
`Michael Sela and his colleagues Ruth Arnon and
`Dvora Teitelbaum15–17 first conceived glatiramer ace-
`tate in the early 1970s. They made a series of random
`copolymers based on the molar ratios of four amino
`acids, glutamate, alanine, tyrosine, and lysine, that are
`present in myelin basic protein. Sela and McDevitt18
`had shown 5 years earlier that the antibody response to
`ordered copolymers of tyrosine and glutamate on a
`backbone of alanine and lysine was under strict genetic
`control linked to the MHC. McDevitt and Sela’s work
`opened the field of the genetic control of the immune
`response. Their discovery that such control was linked
`to the MHC had widespread implications for immu-
`nology. More than just a coincidence, genes within the
`MHC are the most critical for imparting genetic sus-
`ceptibility to MS. Moreover, the MHC HLA class I
`and class II gene products, HLA-A, -B, -DR, and -DQ,
`are the likely targets for glatiramer. Interactions of
`glatiramer with the MHC turned out to be critical in
`understanding its mechanism of action (see Fig).
`In 1971, Sela and colleagues15 showed that the ran-
`dom copolymer composed of glutamate, tyrosine, ala-
`nine, and lysine, termed Copolymer 1, was able to sup-
`press the induction of acute EAE. They then showed
`that Copolymer 1 blocked relapsing EAE in the guinea
`pig and EAE in the nonhuman primate.19 –21 Initial
`clinical testing of glatiramer was undertaken in Jerusa-
`lem under the direction of Abramsky22 in patients with
`MS and acute disseminated encephalomyelitis. Clinical
`testing of glatiramer by Bornstein and colleagues23
`showed that glatiramer was effective in reducing re-
`lapses in relapsing-remitting MS. A pivotal trial leading
`to FDA approval, under the leadership of Johnson,24
`showed that relapses were reduced by 29% in patients
`
`Steinman and Zamvil: Applying EAE Research to MS
`
`13
`
`

`

`Fig. General scheme for pathogenesis of MS. T and B cell homing to the central nervous system is followed by inflammation medi-
`ated by antibodies, complement and the toxic effect of cytokines. Medications approved for multiple sclerosis (MS) that arose from
`studies on experimental allergic encephalomyelitis (EAE) are shown in black. Promising therapies for MS elucidated from a creative
`interplay of work in MS and in EAE are shown in red. ECM ⫽ extracellular matrix; IFN ⫽ interferon; IL ⫽ interleukin;
`MHC ⫽ major histocompatibility complex; TNF ⫽ tumor necrosis factor; VCAM ⫽ vascular cell adhesion molecule.
`
`trade-
`receiving daily injections. Glatiramer acetate,
`marked Copaxone from the original name Copolymer
`1, was approved in 1996 for treatment of relapsing-
`remitting MS. Glatiramer acetate was thus first derived
`from a preclinical conception and invention in the
`acute EAE model, and was then taken through proof
`of concept in various acute and relapsing models of
`EAE. Success in the EAE model was followed by dem-
`onstration of clinical efficacy in relapsing-remitting
`MS. Glatiramer acetate currently is one of the most
`popular medications
`for
`treatment of
`relapsing-
`remitting MS, and more than 100,000 individuals with
`MS worldwide have received glatiramer acetate treat-
`ment.25 It took a quarter of a century for the develop-
`ment of glatiramer from the publications of the first
`results in EAE to its approval for relapsing-remitting
`MS!
`There are multiple mechanisms of action associated
`with glatiramer, and many of these mechanisms were
`first unveiled in the EAE model. Antigen-specific mod-
`ulation of the immune response to myelin basic pro-
`tein has been described.21,26 Modulation of the im-
`mune response with glatiramer leads to deviation of
`cytokine production in response to myelin basic pro-
`tein from so-called Th1 cytokines such as ␥ interferon
`
`14 Annals of Neurology Vol 60 No 1
`
`July 2006
`
`to Th2 cytokine production.26 Another mechanism of
`action centers on the random chemical structures in-
`herent in this random copolymer that allow it to bind
`to molecular targets with a wide combinatorial array of
`peptides based on four amino acids. Glatiramer binds
`to MHC molecules derived from most genetic back-
`grounds.27–29 With its capacity to bind to a broad ar-
`ray of MHC molecules, glatiramer could compete with
`many proteins for these critical molecules responsible
`for presentation of antigen to T lymphocytes.
`Given its widespread binding to diverse HLA mole-
`cules, it is not surprising that glatiramer may have non-
`specific effects on the immune system. Glatiramer has
`been shown not only to block EAE, but is active in
`preventing models of inflammatory bowel disease and
`even amyotrophic lateral sclerosis.30 –32 This implies
`that the effect on MS may not even be specific for this
`disease, but that it may have a more general effect on
`immune modulation. Even though there are multiple
`potential mechanisms of action for glatiramer, and
`even though this drug may have potential uses in other
`diseases, the undisputed history of the development of
`glatiramer shows that it emanated from experiments on
`a treatment for EAE. One might reasonably conclude
`
`

`

`that without EAE, there would not have been a glati-
`ramer for treatment of MS.
`Mitoxantrone was developed for treatment of MS,
`after promising results published in the mid-1980s in
`reversing paralysis in the EAE model. The first article
`on mitoxantrone in EAE described mitoxantrone as a
`novel “anthracenedione that has shown antineoplastic
`activity against a variety of experimental tumors.”33 At
`the time, there was great interest in this class of drugs,
`because numerous cytotoxic agents, including azathio-
`prine, had shown promise in MS.34 Thus, mitox-
`antrone was tried in the EAE model in the rat. Ongo-
`ing paralysis was reversed, and the number and extent
`of perivascular lesions in brain was reduced after treat-
`ment with mitoxantrone. Given its promise in the EAE
`model, clinical development of mitoxantrone was taken
`forward in the clinic, leading to its approval for use in
`MS. FDA approval in 2000 for its use in secondary
`progressive MS and progressive or worsening relapsing-
`remitting MS was granted for reducing frequency of
`relapses and slowing clinical progression of disease.
`Success in the EAE model clearly spurred translation of
`this approach to the clinic in MS.35,36
`Natalizumab is yet another example of a drug that
`was developed directly from work in the EAE model.
`In the early 1990s, immunologists had developed the
`working hypothesis that there were specific “molecular
`addresses” for lymphocyte homing to various organs.
`Some referred to this as the “Zip Code Hypothesis” for
`lymphocyte homing. In collaboration with Yednock
`and colleagues37 at a small biotechnology company,
`Athena Neurosciences,
`the Steinman laboratory at
`Stanford determined the precise molecule involved in
`lymphocyte adhesion to inflamed brains taken from
`rats with EAE.38 Using an assay where frozen sections
`were cut on brains with EAE, the researchers bound
`human and rodent lymphocytes to the inflamed EAE
`sections. Monoclonal antibodies were then applied to
`these sections to see whether this lymphocyte binding
`could be blocked, and by inference, what molecules
`were involved in the association of lymphocytes to in-
`flamed brain tissue. More than 20 monoclonal anti-
`bodies to most of the adhesion molecules known at
`that time were tried on these sections of EAE brain,
`and only monoclonal antibodies binding ␣4 or ␤1 in-
`tegrin molecules inhibited adhesion of lymphocytes to
`the inflamed blood vessels in the brain.37
`We then proceeded to test whether a monoclonal
`antibody to ␣4␤1 integrin could block paralysis in-
`duced by T-cell clones that recognized myelin basic
`protein. These clones caused clinical paralysis and brain
`inflammation in a classic acute EAE model. The anti-
`body to ␣4␤1 integrin inhibited the development of
`paralysis when given at a dose of 4 to 6.4mg/kg in the
`Lewis rat. An article published in 1992 in Nature
`stated:
`
`Previous work on alpha-4 beta-1-dependent cell adhe-
`sion has mainly involved studies with endothelium that
`has been grown and stimulated in culture. The in vitro
`section assay described here extends those observations
`by showing that alpha-4 beta-1 integrin is crucial for
`the adhesion of leukocytes to vessels that have been
`activated in vivo. Furthermore, in vivo administration
`of anti-alpha 4 integrin prevented paralysis associated
`with the pathogenic inflammation of EAE. Therapy
`based on inhibiting alpha-4 beta-1 integrin, or the li-
`gand for this receptor on brain endothelium may prove
`effective in treating inflammatory disease in CNS.37
`From these experiments in EAE, it was recognized that
`blockade of ␣4␤1 integrin might be useful for MS. In
`1995, pathologists demonstrated the vascular cell adhe-
`sion molecule-1, the binding partner for ␣4␤1 inte-
`grin, was expressed in MS lesions. Over the next 10
`years, clinical development of a humanized monoclonal
`antibody to ␣4␤1 integrin showed that it had remark-
`able efficacy in blocking relapses of MS and even de-
`laying disease progression.39,40 Phase 3 studies showed
`that over a 2-year period, injection of 300mg of mono-
`clonal ␣4␤1 integrin reduced the relapse rate by two
`thirds. The dose was directly in the range shown to be
`effective in the experiments
`in EAE reported in
`1992.37 Indeed, the development of natalizumab was a
`tangible result of research in the EAE model.38
`Natalizumab has had a bipolar existence: It was ap-
`proved in November 2004 for use after 1 year of data
`were available in the 2-year phase 3 clinical
`trial.
`Within 3 months, three cases of Progressive Multifocal
`Leukoencephalopathy (PML) were observed, with 2
`deaths. The drug was voluntarily withdrawn. Unfortu-
`nately, PML does not occur in the animal species used
`in the EAE model, and this usually fatal complication
`was neither observed nor could it have been even tested
`in any EAE model or any available animal model for
`that matter.
`Investigators
`searched to see whether
`blockade of ␣4␤1 was associated with increased risk for
`infection to microbes such as cytomegalovirus and
`Borna virus. No increased risk for opportunistic infec-
`tions with either of these viruses was observed. The
`EAE model has limitations, and it is not particularly
`useful for examining the issue of opportunistic infec-
`tions, especially when the microbe in question has a
`species barrier.
`It should also be mentioned that ␤ interferons, the
`other major category of drugs approved for treatment
`of MS, have shown success when tested in EAE mod-
`els.41 However, the ␤ interferons were not developed
`initially because they showed promise in EAE, but
`rather because of the interest in development of anti-
`viral therapies for MS. Therefore, we do not count the
`development of ␤ interferons for therapy of MS as a
`triumph for applications of research on EAE. We do,
`however, consider the development of glatiramer, mi-
`
`Steinman and Zamvil: Applying EAE Research to MS
`
`15
`
`

`

`toxantrone, and natalizumab a direct consequence of
`research in the EAE model. So far, research on EAE
`has given three gifts of new therapies for treatment of
`MS.
`
`Problems and Promise of Using Experimental
`Autoimmune Encephalomyelitis for
`Development of Therapies for
`Multiple Sclerosis
`There is a long list of drugs that have shown promise
`in EAE models that are now being taken forward into
`the clinic. Other approaches including an orally avail-
`able sphingosine inhibitor,42 statins,43– 46 an orally
`available carboxamide,47,48 and a monoclonal antibody
`to IL-2 receptor49 –51 have shown great promise in
`phase 2 trials based first on success in the EAE model
`(see Fig).
`Of course, there are numerous examples of drugs
`that are effective in EAE, only to fail when tested in
`MS. One conclusion from these negative studies is that
`EAE is a poor predictor of success in MS. One must,
`however, examine the process of drug development to
`realize that preclinical research in EAE is merely just
`“exploratory,” whereas human clinical trails in MS un-
`dergo a rigorous “developmental” process,
`involving
`three phases of testing. Thus, once success is seen in
`EAE, one has to contend with issues such as formula-
`tion, dosing, and unforeseen toxicities when a drug is
`taken forward into clinical testing on humans. Given
`the high costs of clinical development of therapies in
`MS, one must make astute choices, usually on the first
`try, when translating preclinical results in EAE to clin-
`ical trials of MS. Each attempt at refining therapy in
`human clinical trials of MS is often prohibitively ex-
`pensive, so second chances are undertaken only in rare
`circumstances. In the case of natalizumab, the dose of
`monoclonal antibody in which a successful outcome
`was achieved in EAE was precisely translated to success
`in human clinical trials. However,
`for other drugs,
`problems with selecting a correct dose and dose fre-
`quency have confounded development. The case of al-
`tered peptide ligands, known as APL, which have
`shown great promise in EAE, exemplifies this problem.
`An APL from the region of myelin basic protein
`p83-99 showed promise in reducing relapse rates and
`reversing paralysis in preclinical studies of EAE.52–55 A
`version of this APL, known as NBI 5788, was designed
`and taken into clinical
`testing in patients with
`relapsing-remitting MS. This APL had an alteration in
`the main contact residues with human T-cell receptors
`recognizing this epitope of myelin basic protein. When
`NBI 5788 was given at a dosage of 50mg/week subcu-
`taneously, it was associated with exacerbations in three
`MS patients in an open-label trial.56 However, dosages
`of 5mg/week of this drug were associated with reduc-
`tion in gadolinium-enhancing lesions on magnetic res-
`
`16 Annals of Neurology Vol 60 No 1
`
`July 2006
`
`onance imaging, and there was no evidence of disease
`exacerbations.57 However, weekly dosing, at 5, 20, and
`50mg,
`led to allergic-type hypersensitivity reactions.
`The basis for these hypersensitivity reactions, seen in
`MS patients in phase 2 clinical trials, was then inves-
`tigated in the EAE model. In this new model of EAE,
`it was discovered that self-peptides of the myelin sheath
`could trigger fatal anaphylactic reactions in mice. The
`implications of this finding (ie, that even self-peptides
`were allergic) raised new and challenging questions. “A
`new version of horror autotoxicus,”58 first described a
`century ago by Paul Ehrlich, was discovered from un-
`derstanding a problem in a clinical trial of an MS drug.
`When dosing of NBI 5788 in phase 2b trials on
`relapsing-remitting MS was reduced to 5mg once a
`month instead of once a week to attempt to mitigate
`these hypersensitivity reactions, both the desirable ac-
`tivity in reducing magnetic resonance activity and the
`undesirable hypersensitivity reactions seen with APL
`disappeared.
`Continued development of APL in humans would
`require further financial
`investment if this particular
`approach is to be pursued with different dosing sched-
`ules. Negative studies in humans, on novel drugs such
`as the APL, which so far have failed to translate into an
`approved drug for MS, may not be a fault of the EAE
`model per se, but rather a reality of the huge expenses
`required for development of new drugs. When going
`from animal to human studies, revisions in dosing and
`formulation may be required that are too expensive
`and time consuming to pursue, given competing prior-
`ities. It is worth noting that the EAE model was used
`to help understand one of the clinical complications of
`this approach, allergic-like hypersensitivity reactions to
`self-constituents,
`seen with administration of
`the
`APL.58
`Another area where the EAE model has been called
`into question has been its inefficiency in predicting
`how blockade of various cytokines would work in MS.
`In rheumatoid arthritis and Crohn’s disease, we have
`seen the major triumph in therapy with the class of
`drugs known as tumor necrosis factor (TNF) blockers.
`This stunning advance in therapy led to the award of a
`Lasker Prize in Clinical Medicine to Profs Marc Feld-
`mann and Taini Maini to honor their achievement for
`implementing this mode of therapy in rheumatoid ar-
`thritis and Crohn’s disease. More than one million pa-
`tients with rheumatoid arthritis and Crohn’s disease
`have benefited from this approach.59 However, TNF
`blockade has been associated with worsening of
`MS,59 – 61 and a “black box” label has been placed on
`these drugs, warning against their use in MS.59 Studies
`in EAE have been equivocal, where some published ex-
`periments have shown the virtues of TNF blockade
`with anti-TNF monoclonal antibodies and soluble
`TNF-receptor constructs62– 64; in contrast, other pub-
`
`

`

`Table. The Long and Winding Road from Proof of Concept in Experimental Autoimmune Encephalomyelitis to US Food and
`Drug Administration Approval: Ranging from a Quarter of a Century to More Than a Decade
`
`Therapeutic
`
`Glatiramer Acetate
`Mitoxantrone
`
`Natalizumab
`
`Year of Publication
`of Proof of Concept in EAE
`
`Year of FDA
`Approval
`
`1971
`1987
`
`1992
`
`1996
`2000
`
`2004
`2005 withdrawn
`2006 reinstated
`
`Approved Indication
`
`Approved for RR-MS
`Approved for secondary progressive MS,
`worsening RR-MS
`Approved for RR-MS
`
`EAE ⫽ experimental autoimmune encephalomyelitis; FDA ⫽ US Food and Drug Administration; RR-MS ⫽ relapsing-remitting multiple
`sclerosis; MS ⫽ multiple sclerosis.
`
`lications have demonstrated the virtues of TNF itself,
`thus highlighting the pitfalls of blockade.65 One reason
`for this confusion is that a molecule such as TNF may
`have “janus-like effects,” giving benefit in some aspects
`of the disease, whereas imparting risk during other as-
`pects of disease. TNF is a mediator of inflammation in
`human autoimmune diseases and in animal models,59
`as well as a mediator of repair, including oligodendro-
`cyte growth, and a mediator of oligodendroglial
`death.66 – 68 Studies on EAE, which are often short
`term, must be carefully interpreted so that we pay at-
`tention to the “janus-like” propensity of cytokines:
`TNF blockade may be beneficial in inhibiting some
`inflammatory pathways, yet deleterious in blocking the
`maintenance and repair of cells in the nervous system.
`We have seen similar problems with glucocorticoids,
`which have virtues as antiinflammatory drugs, but have
`problems in inhibiting wound healing together with
`their numerous other unfortunate side effects.
`Yes, the same molecule may have different effects in
`influencing different biochemical pathways. And yes,
`EAE has its limitations. Misinterpretation and over-
`interpretation of certain results from EAE experiments
`may lead to calamitous consequences. Translation of
`animal to human studies is filled with large uncertain-
`ties. But, this should not mean that we discard the
`EAE model because of our uncertainty and ignorance
`of biology. We are only 21st century medical scientists,
`after all. We must remain humble about our under-
`standing of disease processes. Our predecessors in pre-
`vious centuries appear to us now so quaintly ignorant.
`Even though we have sequenced the human genome,
`we need to know what the products of these genes are
`doing in health and in disease. We need ever more
`clever animal models to help us understand observa-
`tions made in direct studies on human disease.
`One of the exciting directions in the development of
`therapy for MS is consideration of various combina-
`tions of medications. The EAE model has demon-
`strated potential synergies between drugs such as statins
`and glatiramer, which combine in the EAE model to
`show efficacy when used at doses that are suboptimal
`
`for these drugs when used alone.69,70 Other combina-
`tions of therapies might be tried in the EAE model, to
`search for synergies or unexpected adverse interactions.
`Drug development is not only costly, it is remark-
`ably slow. Rip Van Winkle could have a good sleep in
`the time intervals involved in drug development. The
`three drugs glatiramer, mitoxantrone, and natalizumab
`that first showed promise in EAE each took more than
`a decade before they were actually approved to treat
`MS. Drug development is a time consuming and ex-
`pensive process (Table). The fault does not necessarily
`rest with EAE, but in the harsh reality of how difficult
`it is to develop a new drug for MS.
`
`Experimental Autoimmune Encephalomyelitis
`Provides Insights to Understand Pathology, to
`Identify Surrogate Biomarkers and
`Therapeutic Targets
`MS researchers are now using a variety of powerful
`tools to understand the pathology of MS. These tech-
`nologies include gene, protein, and lipid microarrays,
`robotic sequencers for analysis of gene expression in
`MS tissue, and mass spectroscopy to detect minute
`amounts of proteins and lipids
`in MS brain le-
`sions.71–73 Once genes, proteins, and lipids of interest
`have been identified in MS, then clever applications of
`the EAE model allow one to explore their biological
`roles. Take the case of osteopontin, first discovered as a
`major transcript in lesions of MS by the use of a robot
`to sequence gene transcripts isolated from lesion mate-
`rial.74 No one had ever considered a role for osteopon-
`tin in MS brain, and from the name of the molecule,
`its main activity suggested that it involved bone and
`not brain. However, robotic sequencing of genes ex-
`pressed in MS lesions indicated that osteopontin tran-
`scripts were highly elevated at the site of brain inflam-
`mation.
`Immunohistochemistry showed widespread
`expression of the protein in MS lesions. A mouse strain
`existed where the gene for osteopontin was deleted. It
`was discovered that when EAE was induced in this
`strain, disease was milder, disease progression was
`blunted, mortality was decreased, and the intensity of
`
`Steinman and Zamvil: Applying EAE Research to MS
`
`17
`
`

`

`inflammatory mediators such as ␥ interferon was re-
`duced in myelin reactive T cells. Osteopontin, a
`cytokine-like molecule, was thus shown to have impor-
`tant immunomodulatory properties in EAE.74
`Researchers
`then explored whether osteopontin
`might play a role in the progression of MS. Three ar-
`ticles have now reported that osteopontin is elevated in
`blood before a relapse of MS.75–77 In MS, there is no
`surrogate marker to follow disease activity, which is
`akin to measurements of C-reactive protein in rheuma-
`toid arthritis or C-peptide in type 1 diabetes mellitus.
`Research on the EAE model in conjunction with stud-
`ies on human MS brain material may be leading to the
`discovery of the first surrogate marker in blood for MS.
`There are many other examples where a clever inter-
`play of studies on MS material combined with work on
`EAE has been illuminating. For example, B cells posi-
`tive for CD20 are found in MS lesions, and immuno-
`globulin V gene transcripts are among the most highly
`expressed genes in lesions.78,79 In the marmoset model
`of EAE, CD20 cells are common in cortical lesions,80
`whereas in the mouse model of EAE, B cells are quite
`common.81 There is great interest in several aspects of
`the role of B cells. Researchers have used studies on
`EAE combined with parallel observations in MS to de-
`termine the nature of the immune response in the cen-
`tral nervous system in MS, and then to see whether
`such antibody responses may play a pathogenic role in
`MS. Studies on the biological roles of antibodies to
`myelin proteins82– 84 and to lipids such as galactocere-
`broside and sphingomyelin73,85 have shown that not
`only are such antibodies present in the brains and spi-
`nal fluid of MS patients, but such antibodies are crit-
`ical in worsening the severity of disease in EAE models.
`Yet another example of understanding the pathogen-
`esis of a molecule with an unknown role in MS comes
`from studies on cytokines in MS brain. IL-6 and IL-17
`were first reported in MS lesions using transcriptional
`profiling with gene microarrays.80 Studies in the EAE
`model have shown that IL-6 and IL-17 are critical in
`the pathogenesis of EAE.86 IL-6 governs the produc-
`tion of a key set of regulatory T cells that modulate
`another subset of effector T cells producing IL-17.86
`Insights into MS will be understood only after dissec-
`tion of the biology of these newly discovered mole-
`cules. The use of transgenics, genetic “knock-outs and
`knock-ins,” and application of gene silencing tech-
`niques of interfering RNA, all techniques that allow us
`to analyze the biology of molecules in unprecedented
`ways, can o

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