throbber
Translational Therapies for Immune-based Disorders Review Series
`
`Guest Editor: D. Pozo
`
`Systems biology approaches for the study of multiple sclerosis
`
`J. Cell. Mol. Med. Vol 12, No 4, 2008 pp. 1087-1093
`
`Francisco J. Quintana *, Mauricio F. Farez, Howard L. Weiner
`
`Center for Neurologic Diseases, Harvard Medical School, Boston, MA, USA
`
`Received: April 1, 2008; Accepted: May 10, 2008
`
`• Introduction
`• Genomics
`• Transcriptomics
`- Characterization of the MS lesion
`- Characterization of the immune response
`• Proteomics
`-
`Identification of new pathogenic processes
`
`Abstract
`
`- Role of environmental triggers in MS
`- Characterization of the autoimmune response:
`antibodies
`• Metabolomics
`• Integration of data from different ‘omics’ approaches
`• New experimental models
`• Conclusions
`
`Multiple sclerosis (MS) is a progressive neurological disease caused by an autoimmune attack to the central nervous system (CNS). MS
`is thought to result from a complex interaction between genetic and environmental factors. In this review, we analyse the contribution
`of genomics, trancriptomics and proteomics in delineating these factors, as well as their utility for the monitoring of disease progres-
`sion, the identification of new targets for therapeutic intervention and the early detection of individuals at risk.
`
`Keywords: multiple sclerosis • systems biology • autoimmunity
`
`Introduction
`Multiple sclerosis (MS) is an autoimmune disorder in which the
`central nervous system (CNS) is targeted by the dysregulated activ-
`ity of the immune system, resulting in progressive neurological
`dysfunction. A variety of symptoms characterize MS, among them
`are visual and motor problems, changes in sensation in the arms,
`legs or face and weakness. At the onset of the disease, 85–90% of
`the patients present a clinical course characterized by discrete
`attacks followed by periods of partial or total recovery (relapsing-
`remitting MS, RRMS); 10% of the patients present a slowly accu-
`mulating disability over time (primary progressive MS, PPMS). A
`total of 40% of the patients initially diagnosed with RRMS eventu-
`ally become progressive (secondary progressive MS, SPMS).
`The term MS refers to the scars (scleroses or plaques) that
`characterize the white matter of the brain and spinal cord of MS
`patients. The autoimmune attack that drives MS is thought to
`cause these scars, characterized by a perivascular infiltration by
`inflammatory cells (B and T lymphocytes among them) [1, 2]. In
`addition, demyelination, astrogliosis and axonal injury are also
`detected [1, 2]. Different mechanisms contribute to axonal dam-
`age, including the direct effects of pro-inflammatory cytokines,
`complement fixation, apoptosis, cell-mediated cytotoxicity and
`neurodegeneration [1, 2]. Pathological findings suggest that the
`
`relative contribution of each one of these processes in disease
`progression differs in each patient [3].
`The autoimmune response in MS targets components of the
`myelin sheaths surrounding neuronal axons, interfering with the
`neurons’ ability to conduct electrical signals and probably leading to
`their death. Several CNS proteins are targeted by the immune system
`in MS, among them are myelin oligodendrocyte glycoprotein [4],
`oligodendrocyte-specific protein [5], myelin basic protein [6],
`myelin-associated glycoprotein [7], 2⬘,3⬘-cyclic nucleotide 3⬘ phos-
`phodiesterase [8] and ␣␤-crystallin [9]. It is believed that the clinical
`symptoms that characterize MS result from the blockade in axonal
`transmission that follows axonal demyelination or axonal loss [1, 2].
`Epidemiological studies have suggested the contribution of sev-
`eral environmental factors to the susceptibility of a specific individ-
`ual to MS. Several viral, bacterial and parasitic infections have been
`classically linked to MS onset and progression [10–15], but no sin-
`gle environmental agent can be singled out as a ‘cause’ of MS.
`MS is heterogeneous in its rate of progression, clinical symp-
`toms, the specificity of the immune response and the pathology of
`the CNS lesions, reflecting the contribution of different factors to
`a pathogenic autoimmune response [16]. In this review, we
`analyse the contribution of genomics, trancriptomics, proteomics
`
`*Correspondence to: Francisco J. QUINTANA, Ph.D.,
`Center for Neurologic Diseases, Harvard Medical School,
`77 Ave. L. Pasteur HIM720, Boston 02115, MA, USA.
`
`Tel.: +617-525-5311
`Fax: +617-525-5305
`E-mail: fquintana@rics.bwh.harvard.edu
`
`© 2008 The Authors
`Journal compilation © 2008 Foundation for Cellular and Molecular Medicine/Blackwell Publishing Ltd
`
`doi:10.1111/j.1582-4934.2008.00375.x
`
`
`Page 1 of 7
`
`YEDA EXHIBIT NO. 2002
`MYLAN PHARM. v. YEDA
`IPR2014-00644
`
`

`

`and metabolomics in delineating these factors, as well as their utility
`for the monitoring of disease progression, the identification of
`new targets for therapeutic intervention and the early detection of
`individuals at risk.
`
`Genomics
`
`MS is considered a complex genetic disease in which many poly-
`morphic genes have small effects on the overall disease risk, its
`severity, rate of progression and age of onset among several clin-
`ical outcomes. To date, the strongest chromosomal region linked
`to MS is the major histocompatiblity complex (MHC) locus on
`chromosome 6p21 [17–19]. In addition, several non-MHC candi-
`date loci have also been linked to MS [19], but it has proven diffi-
`cult to validate their association in independent studies. The diffi-
`culty in the identification of non-MHC genes associated to MS
`might reflect the genetic heterogeneity existing among MS
`patients, which results in different combinations of gene alleles
`leading to the same end phenotype. Nevertheless, polymorphisms
`in the ␣ chain of the IL-7 receptor (IL-7R␣) have been recently
`associated with MS [20–22]. These polymorphisms make only a
`small contribution to the genetic susceptibility to MS but are a sig-
`nificant step towards the identification of genetic determinants for
`MS outside the MHC locus. The IL-7R␣ allele associated with MS
`favours a relative decrease in the membrane-bound IL-7R␣ [21].
`IL-7 is produced by stromal cells in lymphoid tissues, its availabil-
`ity is controlled through its uptake by the membrane-bound IL-7R
`on T cells [23]. Thus, considering the positive effects that IL-7 has
`on lymphocyte survival and proliferation [23], the decrease in
`membrane IL-7R might result in increased levels of IL-7 available
`to fuel the inflammatory T-cell response in MS.
`The ␣ chain IL-2 receptor (IL-2R␣) gene has also been recent-
`ly linked to MS [22]. IL-2R␣ allelic variation has been previously
`associated to other autoimmune diseases such as type I diabetes,
`but at a different genomic position [24]. IL-2 is required for the
`development of regulatory T cells (Treg) [25, 26], and indeed,
`deficits in Treg activity characterize RRMS [27]; thus the IL-2R␣
`polymorphisms might be related to the immune dysregulation
`observed in MS. Notably, IL-2R␣-specific antibodies have shown
`promising beneficial effects for the treatment of MS on phase 2 clin-
`ical trials [28, 29]. Although the link between IL-2R␣ polymorphisms
`and MS is still awaiting further validation, the association of IL-7R␣
`and IL-2R␣ variants to MS supports the use of genome-wide stud-
`ies to delineate pathways contributing to disease pathogenesis.
`
`Transcriptomics
`
`Characterization of the MS lesion
`
`Large-scale studies of mRNA expression have been directed at
`characterizing either the lesion or the immune response in MS.
`Lock and coworkers found that ␣4-integrin was found to be
`
`elevated in MS lesions [30]. ␣4-integrin mediates the interaction
`of T cells with the endothelium in the inflamed CNS, a required
`step for the migration of the self-reactive T cells into the brain and
`spinal cord in MS [31]. Antibodies to ␣4-integrin reverse and
`reduce the rate of relapse in relapsing-remitting experimental
`autoimmune encephalomyelitis (EAE) an animal model for MS
`[32], and a humanized version of this antibody showed positive
`effects in the treatment of RRMS [33].
`In a separate study, the large-scale sequencing of non-normal-
`ized cDNA libraries derived from MS plaques revealed an increased
`expression of osteopontin (OPN) in the CNS of MS and EAE sam-
`ples [34]. The up-regulation of OPN levels in MS plaques [35] and
`in the circulation [36–38] of MS patients was replicated in inde-
`pendent studies, prompting the search for polymorphisms in the
`opn gene associated with MS. Although some controversy still
`remains [39], polymorphisms in the opnlocus have been associat-
`ed with increased levels of circulating OPN and the clinical course
`of MS [40]. To study the mechanism of action of OPN in MS, OPN-
`deficient mice were generated, which showed a reduced severity in
`EAE [34]. OPN-triggered signalling is thought to contribute to MS
`pathogenesis by increasing the pro-inflammatory phenotype and
`survival of pathogenic myelin-specific T cells [41]. In addition, OPN
`interacts with the ␣4-integrin and is also involved in cell migration
`into the inflamed CNS [42]. Neutralization of OPN with neutralizing
`antibodies results in the amelioration of EAE [43]. Thus, OPN is
`therefore an example of how results obtained in transcriptomics
`studies might lead to the identification of mechanisms of disease
`pathogenesis and new therapeutic targets for MS.
`
`Characterization of the immune response
`
`Transcriptional profiling has also been used to study the peripheral
`immune response in MS. Two limitations, however, should be kept
`in mind when using cDNA arrays for the analysis of the immune
`response in MS patients: First, these studies assume that changes
`in the peripheral immune system somehow reflect the immune
`response within the CNS. Second, the results of these studies are
`influenced by factors such as gender, age or changes in the relative
`proportion of different blood cell subsets that occur through the
`course of the disease. Nevertheless, two areas show significant
`progress in the transcriptional profiling of the immune response in
`MS: the follow-up of disease activity and the response to therapy.
`Follow-up of disease activity: Achiron and coworkers charac-
`terized the transcriptional activity in peripheral blood mononuclear
`cells (PBMC) from RRMS patients during the course of the dis-
`ease [44]. The authors identified a transcriptional signature asso-
`ciated to the relapse, that included genes involved in the recruit-
`ment of immune cells, epitope spreading and escape from
`immune-regulation. Although encouraging, these results should
`be validated using an independent set of samples and in longitu-
`dinal studies to assess their predictive value.
`Response to therapy: ␤–Interferon (␤–IFN) is widely used for
`the treatment of MS [45], however, biomarkers that would allow
`
`1088
`
`© 2008 The Authors
`Journal compilation © 2008 Foundation for Cellular and Molecular Medicine/Blackwell Publishing Ltd
`
`
`Page 2 of 7
`
`YEDA EXHIBIT NO. 2002
`MYLAN PHARM. v. YEDA
`IPR2014-00644
`
`

`

`the identification of patients that would benefit from treatment
`with ␤–IFN are still not available. Weinstock-Guttman and col-
`leagues used cDNA micro-arrays to study the effects of ␤–IFN
`therapy on monocyte-depleted PBMC [46]. They found significant
`changes in the expression of genes involved in the anti-viral
`response, ␤–IFN signalling and markers of lymphocyte activation.
`These studies provided a molecular description of the effects of
`␤–IFN on RRMS patients and were later on extended to identify
`transcriptional signatures associated to a favourable response to
`treatment with ␤–IFN [47]. Based on these observations,
`Oksenberg and coworkers constructed a classifier for the identifi-
`cation of MS patients likely to respond to treatment with ␤–IFN
`[48]. The work of Oksenberg and coworkers is remarkable for two
`reasons: First, it demonstrates that gene expression profiling can
`be helpful in the selection of therapeutic regimes for the manage-
`ment of MS. Second, it uses a technology (real-time PCR) acces-
`sible to clinical laboratories, facilitating the translation of their
`results into daily medical practice.
`
`Proteomics
`
`Proteomic studies in MS have been shown to identify new
`processes contributing to disease pathology and also, biomarkers
`for the early diagnosis and monitoring of MS patients.
`
`Identification of new pathogenic processes
`
`Recently a large proteomic study of MS lesions by Steinman and
`coworkers have identified tissue factor and protein C inhibitor
`expression within chronic active plaque samples, suggesting that
`the dysregulation of the clotting cascade contributes to MS patho-
`genesis [49]. The authors went on to investigate the potential ther-
`apeutic use of their findings on EAE, concluding that the coagula-
`tion cascade is an attractive therapeutic target in MS.
`
`Role of environmental triggers in MS
`
`Epidemiological studies suggest that environmental factors con-
`tribute to MS susceptibility. As a result, several groups are active-
`ly searching for microbial triggers for MS [10–15]. One of these
`putative triggers is the Epstein-Barr Virus (EBV) [50]. The link
`between EBV infection and MS has been recently strengthened by
`the work of Cepok and coworkers, who used protein expression
`arrays to characterize the antibody reactivity in the cerebrospinal
`fluid (CSF) of MS patients, most of those antibodies recognized
`EBV epitopes [51]. These results, together with the detection of
`EBV reactivation in active MS lesions [52], suggest that EBV might
`elicit an abnormal immune response in susceptible individuals
`that contributes to MS [53].
`
`J. Cell. Mol. Med. Vol 12, No 4, 2008
`
`Characterization of the autoimmune response:
`antibodies
`
`The autoimmune nature of MS suggests that the study of the
`immune response should be useful for the early diagnosis, prog-
`nosis and monitoring of MS patients. T cells are thought to make
`a major contribution to MS immuno-pathology [16], but the stan-
`dardized characterization of the T-cell response has proven diffi-
`cult in MS. Antibodies might also have a pathological role [54].
`Moreover, the activation of antibody-producing B cells is con-
`trolled by T cells, thus antibody response is thought to reflect the
`activity of the T-cell compartment [55]. Since it is easier to assay
`antibody reactivity than to follow antigen-specific T-cell respons-
`es, new technologies have been developed for monitoring the
`humoural response in MS patients and autoimmunity [56, 57].
`Antigen arrays have been shown to detect changes in the
`repertoire of antibodies reflecting the antigen spreading that
`accompanies EAE progression [58]. The information obtained
`about the antigen spreading was used to design tailored immuno-
`modulatory vaccines to control EAE [58]. Of note, these vaccines
`showed promising results in a phase 1/2 human clinical trial [59].
`Future experiments should study the antibody response in the
`serum of MS patients, searching for patterns of antibody reactivity
`that predict the progression of MS or the response to therapy, as it
`was shown for other autoimmune disorders, such as rheumatoid
`arthritis [60], autoimmune diabetes [57] and systemic lupus erythe-
`matosus [61]. Indeed, our own data suggest that antigen arrays
`might be used to identify antibody patterns linked to the different
`forms of MS and identify pathogenic mechanisms and therapeutic
`targets (F. J. Quintana et al., submitted). Thus, antigen arrays
`are promising platforms for the identification of patients at risk of
`developing MS, before the overt onset of the symptoms [57].
`
`Metabolomics
`
`The metabolome is defined as ‘the complete set of small-molecule
`metabolites (such as metabolic intermediates, hormones and other
`signalling molecules, and secondary metabolites) to be found within
`a biological sample, such as a single organism’ [62]. Although initial
`studies aimed at studying the metabolome in simple organisms like
`the yeast [63], the study of a limited subset of the human
`metabolome in health and disease is well underway [64, 65]. Several
`groups have undertaken the study of metabolomic aspects of MS.
`During the course of MS, macrophages and astrocytes pro-
`duce nitric oxide, a metabolite that is thought to contribute to sev-
`eral aspects of MS pathology such as the disruption of the
`blood–brain barrier, oligodendrocyte injury and demyelination,
`axonal degeneration [66]. Nitric oxide metabolites can be detect-
`ed in CSF, serum and urine of MS patients, and their levels seem
`to reflect the activity of inflammatory processes that contribute to
`the pathology of the disease [66–68].
`
`© 2008 The Authors
`Journal compilation © 2008 Foundation for Cellular and Molecular Medicine/Blackwell Publishing Ltd
`
`1089
`
`
`Page 3 of 7
`
`YEDA EXHIBIT NO. 2002
`MYLAN PHARM. v. YEDA
`IPR2014-00644
`
`

`

`N-acetylaspartate (NAA) is only present in living mature neu-
`rons, thus decreases in NAA levels indicate neuronal loss.
`Magnetic resonance (MR)-based approaches have been success-
`fully used to measure the levels of NAA and other metabolites
`[69–72] in vivo, providing a novel non-invasive method for the
`acquisition of real-time data about the state of the CNS in MS. The
`power of this approach is highlighted by new data showing that
`the precise measurement of NAA levels by a combination of in vivo
`proton MR spectroscopic imaging with segmented, high-resolu-
`tion MR imaging can identify RRMS patients in their transition to
`the SPMS form of the disease [73]. Although preliminary, these
`results suggest that metabolomics might provide sensitive bio-
`markers to follow up changes in the neurodegenerative and
`inflammatory processes that contribute to MS pathogenesis [74].
`
`Integration of data from different
`‘omics’ approaches
`
`The combination of the data generated in transcriptomics and
`genomics studies can be an invaluable source of information and
`new hypotheses. Aune et al. compared the genes differentially
`expressed by lymphocytes in rheumatoid arthritis, systemic lupus
`erythematosus, insulin-dependent diabetes mellitus and MS, con-
`cluding that they are clustered within chromosomal domains in
`the genome [75]. Strikingly, they found that the chromosomal
`domains containing the genes differentially expressed in autoim-
`mune disorders could be mapped to disease susceptibility loci
`associated to those diseases by genetic linkage studies [75].
`These results suggest that the expression of disease-associated
`genes is co-regulated as a result of shared genetic regulatory ele-
`ments or local patterns of chromatin condensation. Recently,
`Baranzini and coworkers studied the genetic concordance
`between gene expression and genetic linkage in MS [76]. They
`first compiled the data on gene expression available for MS and
`EAE, and superimposed it with all the known susceptibility loci
`identified in MS and EAE. In their study, Baranzini and coworkers
`identified the MS susceptibility genes located in the MHC locus as
`overlapping with clusters of differentially expressed genes in MS
`and murine EAE. However, they could also identify an interesting
`region on chromosome X that might contribute to the sexual
`dimorphism observed in MS. The integration of the data generat-
`ed by different platforms, like transcriptomics, genomics and pro-
`teomics, is therefore likely to deepen our understanding of the
`mechanisms driving MS.
`
`New experimental models
`
`Screenings aimed at identifying genes or drugs controlling the
`immune response cannot be easily undertaken in mice because
`they are based on crossing, maintaining and screening large num-
`
`bers of animals, an expensive time-, space- and labour-intensive
`task; new experimental models are needed. Our current knowl-
`edge on innate immunity originated from pioneering studies that
`used flies and worms to carry out genetic studies and identify
`pathways controlling the response to microbes [77]. Invertebrates
`lack adaptive immunity, but the zebrafish (Danio rerio) harbours
`an adaptive immune system that resembles the mammalian
`immune system [78] and offers several experimental advantages
`for the study of pathways controlling vertebrate processes of inter-
`est. As part of our work on the zebrafish to identify pathways con-
`trolling immunity, we have characterized the zebrafish homologues
`of the transcription factors autoimmune regulator [79] and Foxp3
`[80, 81], pivotal for central and peripheral tolerance, respectively.
`Our work on zebrafish Foxp3 led us to identify the ligand-acti-
`vated transcription factor aryl hydrocarbon receptor (AHR) as a
`regulator of the expression of mammalian Foxp3 [82]. Upon acti-
`vation by its ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD),
`AHR interacts with its binding sites on the Foxp3 gene and stimu-
`lates its transcription. AHR activation by TCDD generates function-
`al Treg that inhibit the development of EAE by a transforming
`growth factor ␤1-dependent mechanism. Surprisingly, AHR acti-
`vation by an alternative ligand, 6-formylindolo[3,2-b]carbazole,
`interferes with Treg differentiation, boosts TH17 differentiation and
`worsens EAE. Thus, AHR regulates both Treg and TH17 differentia-
`tion in a ligand-specific fashion, constituting a unique target for
`therapeutic immuno-modulation. In addition, our findings suggest
`that the experimental advantages offered by the zebrafish can be
`exploited to characterize metabolic pathways controlling immunity in
`vertebrates and to identify new targets for therapeutic intervention.
`
`Conclusions
`
`How can we apply the information provided by genomics, tran-
`scriptomics and proteomics to the early diagnosis, prevention,
`monitoring and therapy of MS? A first step is the establishment of
`experimental models where biologic problems of interest can be
`investigated through genomic, transcriptomic and proteomic
`approaches simultaneously. The zebrafish, with its experimental
`advantages for the study of vertebrate-specific processes [78],
`might turn into a platform, where to identify pathways contribut-
`ing to MS pathology and therapeutic targets. Our findings on the
`control of the immune response by AHR support this view.
`Another step should be the adaptation of genomic, transcrip-
`tomic and proteomic technologies to a clinical setup. The person-
`al genome project, for example, aims at developing fast and reli-
`able methods to sequence the individual human genomes for
`US$1000 or less [83]. However, the data generated by these high-
`throughput approaches should be integrated to understand how
`the genomics, transcriptomics and proteomics of an individual
`influence each other. This would require the development of com-
`putational tools for the integration of networks and pathways into
`accurate quantitative models [84, 85]; with user interfaces aimed
`at facilitating its exploration and modification [86].
`
`1090
`
`© 2008 The Authors
`Journal compilation © 2008 Foundation for Cellular and Molecular Medicine/Blackwell Publishing Ltd
`
`
`Page 4 of 7
`
`YEDA EXHIBIT NO. 2002
`MYLAN PHARM. v. YEDA
`IPR2014-00644
`
`

`

`J. Cell. Mol. Med. Vol 12, No 4, 2008
`
`MS results from a complex dialogue between a susceptible
`individual and a fostering environment, a dialogue unique to each
`individual. The combination of genomic, transcriptomic and pro-
`teomic techniques might allow us to identify key elements in this
`dialogue to prevent, diagnose and cure MS.
`
`mental autoimmune encephalomyelitis (EAE), IL-7 receptor ␣ chain
`(IL-7R␣), IL-2 receptor ␣ chain (IL-2R␣), magnetic resonance (MR), major
`histocompatiblity complex (MHC), multiple sclerosis (MS), N-acetylaspartate
`(NAA), nitric oxide (NO), osteopontin (OPN), peripheral blood mononuclear
`cells (PBMC), primary progressive MS (PPMS), regulatory T cells (Treg),
`relapsing-remitting MS (RRMS), secondary progressive MS (SPMS).
`
`Abbreviations
`
`Acknowledgements
`
`2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), aryl hydrocarbon
`receptor (AHR), ␤–Interferon (␤–IFN), central nervous system
`(CNS), Cerebrospinal fluid (CSF), Epstein-Barr Virus (EBV), experi-
`
`FJQ is a recipient of a long-term fellowship from the Human Frontiers of
`Science Program Organization.
`
`References
`
`2.
`
`3.
`
`4.
`
`1. McFarland HF, Martin R. Multiple sclero-
`sis: a complicated picture of autoimmuni-
`ty. Nat Immunol.2007; 8: 913–9.
`Sospedra M, Martin R. Immunology of
`multiple sclerosis. Annu Rev Immunol.
`2005; 23: 683–747.
`Lucchinetti CF, Bruck W, Lassmann H.
`Evidence for pathogenic heterogeneity in
`multiple sclerosis. Ann Neurol.2004; 56: 308.
`Xiao BG, Linington C, Link H. Antibodies
`to myelin-oligodendrocyte glycoprotein in
`cerebrospinal fluid from patients with mul-
`tiple sclerosis and controls. J Neuroimmunol.
`1991; 31: 91–6.
`Bronstein JM, Lallone RL, Seitz RS,
`Ellison GW, Myers LW. A humoral
`response to oligodendrocyte-specific pro-
`tein in MS: a potential molecular mimic.
`Neurology.1999; 53: 154–61.
`6. Warren KG, Catz I, Steinman L. Fine speci-
`ficity of the antibody response to myelin
`basic protein in the central nervous system
`in multiple sclerosis: the minimal B-cell
`epitope and a model of its features. Proc
`Natl Acad Sci U S A.1995; 92: 11061–5.
`7. Moller JR, Johnson D, Brady RO,
`Tourtellotte WW, Quarles RH. Antibodies to
`myelin-associated glycoprotein (MAG) in
`the cerebrospinal fluid of multiple sclerosis
`patients. J Neuroimmunol.1989; 22: 55–61.
`8. Walsh MJ, Murray JM. Dual implication of
`2⬘,3⬘-cyclic nucleotide 3⬘ phosphodi-
`esterase as major autoantigen and C3
`complement-binding protein in the patho-
`genesis of multiple sclerosis. J Clin Invest.
`1998; 101: 1923–31.
`Celet B, Akman-Demir G, Serdaroglu P,
`Yentur SP, Tasci B, van Noort JM, Eraksoy
`M, Saruhan-Direskeneli G. Anti-alpha B-
`crystallin immunoreactivity in inflammatory
`
`5.
`
`9.
`
`nervous system diseases. J Neurol.2000;
`247: 935–9.
`10. Lipton HL, Liang Z, Hertzler S, Son KN. A
`specific viral cause of multiple sclerosis:
`one virus, one disease. Ann Neurol.2007;
`61: 514–23.
`11. Ascherio A, Munger KL. Environmental
`risk factors for multiple sclerosis. Part I:
`the role of infection. Ann Neurol.2007; 61:
`288–99.
`12. Giovannoni G, Cutter GR, Lunemann J,
`Martin R, Munz C, Sriram S, Steiner I,
`Hammerschlag MR, Gaydos
`CA.
`Infectious causes of multiple sclerosis.
`Lancet Neurol.2006; 5: 887–94.
`13. Gilden DH. Infectious causes of multiple
`sclerosis. Lancet Neurol.2005; 4: 195–202.
`14. Correale J, Farez M. Monocyte-derived
`dendritic cells in multiple sclerosis: the
`effect
`of
`bacterial
`infection. J
`Neuroimmunol.2007; 190: 177–89.
`15. Correale J, Farez M. Association between
`parasite infection and immune responses
`in multiple sclerosis. Ann Neurol. 2007;
`61: 97–108.
`16. Weiner HL. Multiple sclerosis is an
`inflammatory T-cell-mediated autoim-
`mune disease. Arch Neurol. 2004; 61:
`1613–5.
`Jersild C, Svejgaard A, Fog T. HL-A anti-
`gens and multiple sclerosis. Lancet.1972;
`1: 1240–1.
`18. GAMES; Transatlantic Multiple Sclerosis
`Genetics Cooperative. A meta-analysis of
`whole genome linkage screens in multiple
`sclerosis. J Neuroimmunol. 2003; 143:
`39–46.
`19. Oksenberg JR, Barcellos LF. Multiple scle-
`rosis genetics: leaving no stone unturned.
`Genes Immun.2005; 6: 375–87.
`
`17.
`
`20. Lundmark F, Duvefelt K, Iacobaeus E,
`Kockum I, Wallstrom E, Khademi M,
`Oturai A, Ryder LP, Saarela J, Harbo
`HF, Celius EG, Salter H, Olsson T,
`Hillert J. Variation in interleukin 7
`receptor alpha chain (IL7R) influences
`risk of multiple sclerosis. Nat Genet.
`2007; 39: 1108–13.
`21. Gregory SG, Schmidt S, Seth P,
`Oksenberg JR, Hart J, Prokop A, Caillier
`SJ, Ban M, Goris A, Barcellos LF, Lincoln
`R, McCauley JL, Sawcer SJ, Compston
`DA, Dubois B, Hauser SL, Garcia-Blanco
`MA, Pericak-Vance MA, Haines JL.
`Interleukin 7 receptor alpha chain (IL7R)
`shows allelic and functional association
`with multiple sclerosis. Nat Genet. 2007;
`39: 1083–91.
`22. Hafler DA, Compston A, Sawcer S, Lander
`ES, Daly MJ, De Jager PL, de Bakker PI,
`Gabriel SB, Mirel DB,
`Ivinson AJ,
`Pericak-Vance MA, Gregory SG, Rioux
`JD, McCauley JL, Haines JL, Barcellos
`LF, Cree B, Oksenberg JR, Hauser SL.
`Risk alleles for multiple sclerosis identified
`by a genomewide study. N Engl J Med.
`2007; 357: 851–62.
`23. Mazzucchelli R, Durum SK. Interleukin-7
`receptor expression: intelligent design. Nat
`Rev Immunol.2007; 7: 144–54.
`24. Lowe CE, Cooper JD, Brusko T, Walker
`NM, Smyth DJ, Bailey R, Bourget K,
`Plagnol V, Field S, Atkinson M, Clayton
`DG, Wicker LS, Todd JA. Large-scale
`genetic fine mapping and genotype-pheno-
`type associations implicate polymorphism
`in the IL2RA region in type 1 diabetes. Nat
`Genet.2007; 39: 1074–82.
`25. Malek TR. The biology of interleukin-2.
`Annu Rev Immunol.2008; 26: 453–79.
`
`© 2008 The Authors
`Journal compilation © 2008 Foundation for Cellular and Molecular Medicine/Blackwell Publishing Ltd
`
`1091
`
`
`Page 5 of 7
`
`YEDA EXHIBIT NO. 2002
`MYLAN PHARM. v. YEDA
`IPR2014-00644
`
`

`

`26. Suzuki H, Kundig TM, Furlonger C,
`Wakeham A, Timms E, Matsuyama T,
`Schmits R, Simard JJ, Ohashi PS, Griesser
`H, Taniguchi T, Paige CJ, Mak TW.
`Deregulated T cell activation and autoim-
`munity in mice lacking interleukin-2 recep-
`tor beta. Science.1995; 268: 1472–6.
`27. Viglietta V, Baecher-Allan C, Weiner HL,
`Hafler DA. Loss of functional suppression
`by CD4⫹CD25⫹ regulatory T cells in
`patients with multiple sclerosis. J Exp
`Med.2004; 199: 971–9.
`28. Rose JW, Watt HE, White AT, Carlson NG.
`Treatment of multiple sclerosis with an
`anti-interleukin-2 receptor monoclonal
`antibody. Ann Neurol.2004; 56: 864–7.
`29. Bielekova B, Richert N, Howard T, Blevins
`G, Markovic-Plese S, McCartin J, Frank
`JA, Wurfel J, Ohayon J, Waldmann TA,
`McFarland HF, Martin R. Humanized anti-
`CD25 (daclizumab) inhibits disease activi-
`ty in multiple sclerosis patients failing to
`respond to interferon beta. Proc Natl Acad
`Sci USA.2004; 101: 8705–8.
`30. Lock C, Hermans G, Pedotti R, Brendolan
`A, Schadt E, Garren H, Langer-Gould A,
`Strober S, Cannella B, Allard J,
`Klonowski P, Austin A, Lad N, Kaminski N,
`Galli SJ, Oksenberg JR, Raine CS, Heller
`R, Steinman L. Gene-microarray analysis of
`multiple sclerosis lesions yields new targets
`validated in autoimmune encephalomyelitis.
`Nat Med.2002; 8: 500–8.
`31. Rice GP, Hartung HP, Calabresi PA. Anti-
`alpha4 integrin therapy for multiple sclero-
`sis: mechanisms and rationale. Neurology.
`2005; 64: 1336–42.
`32. Yednock TA, Cannon C, Fritz LC, Sanchez-
`Madrid F, Steinman L, Karin N. Prevention
`of experimental autoimmune encephalo-
`myelitis by antibodies against alpha 4 beta
`1 integrin. Nature.1992; 356: 63–6.
`33. Polman CH, O’Connor PW, Havrdova E,
`Hutchinson M, Kappos L, Miller DH,
`Phillips JT, Lublin FD, Giovannoni G,
`Wajgt A, Toal M, Lynn F, Panzara MA,
`Sandrock AW. A randomized, placebo-con-
`trolled trial of natalizumab for relapsing
`multiple sclerosis. N Engl J Med. 2006;
`354: 899–910.
`34. Chabas D, Baranzini SE, Mitchell D,
`Bernard CC, Rittling SR, Denhardt DT,
`Sobel RA, Lock C, Karpuj M, Pedotti R,
`Heller R, Oksenberg JR, Steinman L.
`The influence of the proinflammatory
`cytokine, osteopontin, on autoimmune
`demyelinating disease. Science. 2001;
`294: 1731–5.
`35. Tajouri L, Mellick AS, Tourtellotte A,
`Nagra RM, Griffiths LR. An examination of
`
`MS candidate genes identified as differen-
`tially regulated in multiple sclerosis plaque
`tissue, using absolute and comparative
`real-time Q-PCR analysis. Brain Res Brain
`Res Protoc.2005; 15: 79–91.
`36. Comabella M, Pericot I, Goertsches R,
`Nos C, Castillo M, Blas Navarro J, Rio J,
`Montalban X. Plasma osteopontin levels in
`multiple sclerosis. J Neuroimmunol.2005;
`158: 231–9.
`37. Vogt MH, Floris S, Killestein J, Knol DL,
`Smits M, Barkhof F, Polman CH,
`Nagelkerken L. Osteopontin levels and
`increased disease activity in relapsing-
`remitting multiple sclerosis patients. J
`Neuroimmunol.2004; 155: 155–60.
`38. Vogt MH, Lopatinskaya L, Smits M,
`Polman CH, Nagelkerken L. Elevated
`osteopontin levels in active relapsing-
`remitting multiple sclerosis. Ann Neurol.
`2003; 53: 819–22.
`39. Caillier S, Barcellos LF, Baranzini SE,
`Swerdlin A, Lincoln RR, Steinman L, Martin
`E, Haines JL, Pericak-Vance M, Hauser SL,
`Oksenberg JR. Osteopontin polymorphisms
`and disease course in multiple sclerosis.
`Genes Immun.2003; 4: 312–5.
`40. Chiocchetti A, Comi C, Indelicato M,
`Castelli L, Mesturini R, Bensi T,
`Mazzarino MC, Giordano M, D’Alfonso S,
`Momigliano-Richiardi P, Liguori M,
`Zorzon M, Amoroso A, Trojano M, Monaco
`F, Leone M, Magnani C, Dianzani U.
`Osteopontin gene haplotypes correlate
`with multiple sclerosis development and
`progression. J Neuroimmunol.2005; 163:
`172–8.
`41. Hur EM, Youssef S, Haws ME, Zhang SY,
`Sobel RA, Steinman L. Osteopontin-
`induced relapse and progression of
`autoimmune brain disease
`through
`enhanced survival of activated T cells. Nat
`Immunol.2007; 8: 74–83.
`42. Bayless KJ, Meininger GA, Scholtz JM,
`Davis GE. Osteopontin is a ligand for the
`alpha4beta1 integrin. J Cell Sci.1998; 111:
`1165–74.
`43. Blom T, Franze

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