throbber
REVIEW ARTICLE
`
`Biodrugs 2010; 24 (4): 249-274
`1173-8804/10/0004-0249/$49.95/0
`ª 2010 Adis Data Information BV. All rights reserved.
`
`Therapeutic Approaches to Multiple Sclerosis
`An Update on Failed, Interrupted, or Inconclusive Trials of Immunomodulatory
`Treatment Strategies
`
`Jochen C. Ulzheimer,1,2 Sven G. Meuth,1,3 Stefan Bittner,1 Christoph Kleinschnitz,1 Bernd C. Kieseier4 and Heinz Wiendl3
`
`1 Department of Neurology, University of Wuerzburg, Wuerzburg, Germany
`2 Clinic of Neurology, Caritas Hospital Bad Mergentheim, Bad Mergentheim, Germany
`3 Department of Neurology Inflammatory Disorders of the Nervous System and Neurooncology, University of Muenster,
`Muenster, Germany
`4 Department of Neurology, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
`
`Contents
`
`4.
`
`Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249
`1.
`Immunopathology of Multiple Sclerosis and Therapeutic Targets . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250
`2. Modulation of T-Cell Differentiation and T Helper (Th)-1/Th2 Balance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250
`2.1 Interleukin-12/23: p40 Neutralizing Monoclonal Antibody (Ustekinumab) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251
`2.2 Phosphodiesterase Inhibitors (Ibudilast, Rolipram) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251
`2.3 HMG-CoA-Dependent T-Cell Signaling: Statins (Atorvastatin) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253
`3. Modulation of T-Cell Activation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253
`3.1 Cytotoxic T-Lymphocyte Antigen 4 (CTLA4): Chimeric CTLA4-Ig (Abatacept, RG2077) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 254
`3.2 CD40-CD40L: Blocking Monoclonal Anti-CD154 Antibody (Toralizumab) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 256
`3.3 Peroxisome Proliferator-Activated Receptor (PPAR)-g Agonists: Thiazolidinediones (Pioglitazone, Rosiglitazone). . . . . . . . . . . . 256
`Inhibition of Leukocyte Chemotaxis, Adhesion, and Migration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 257
`4.1 Chemokine Receptors: CCR1 Antagonists (BX-471, CP-481715) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 257
`4.2 Vascular Endothelial Cell Adhesion Molecules: Monoclonal Anti-LFA-1 (CD11/CD18) Antibody (Efalizumab, Hu23F2G) . . . . . . 259
`4.3 Matrix Metalloproteinases (Minocycline, Doxycycline) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 259
`Induction of Immunotolerance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 260
`5.1 Hematopoietic Stem Cell Transplantation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 261
`5.2 Mesenchymal Non-Hematopoietic Stem Cell Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 264
`5.3 Myelin Peptide Therapy: MBP-8298 (Dirucotide) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 264
`5.4 DNA Vaccination: Myelin Basic Protein DNA Vaccine (BHT-3009) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265
`6. Modulation of Antigen Recognition and Disease Triggers. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 266
`6.1 Putative Viral Triggers: Antiviral Agents (Acyclovir, Valacyclovir) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 266
`6.2 Putative Bacterial Triggers: Antibiotics (Rifampicin, Azithromycin) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 267
`6.3 Antigen Degradation: Hydrolytic Enzymes (Bromelain) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 267
`7. Other Immunosuppressants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 268
`7.1 Methotrexate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 268
`8. Concluding Remarks. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 269
`
`5.
`
`Abstract
`
`Multiple sclerosis (MS) continues to be a therapeutic challenge, and much effort is being made to develop
`new and more effective immune therapies. Particularly in the past decade, neuroimmunologic research has
`delivered new and highly effective therapeutic options, as seen in the growing number of immunotherapeutic
`agents and biologics in development. However, numerous promising clinical trials have failed to show
`
`Biogen Exhibit 2120
`Mylan v. Biogen
`IPR 2018-01403
`
`Page 1 of 26
`
`

`

`250
`
`Ulzheimer et al.
`
`efficacy or have had to be halted prematurely because of unexpected adverse events. Some others have
`shown results that are of unknown significance with regard to a reliable assessment of true efficacy versus
`safety. For example, studies of the highly innovative monoclonal antibodies that selectively target im-
`munologic effector molecules have not only revealed the impressive efficacy of such treatments, they have
`also raised serious concerns about the safety profiles of these antibodies. These results add a new dimension
`to the estimation of risk-benefit ratios regarding acute or long-term adverse effects.
`Therapeutic approaches that have previously failed in MS have indicated that there are discrepancies between
`theoretical expectations and practical outcomes of different compounds. Learning from these defeats helps to
`optimize future study designs and to reduce the risks to patients. This review summarizes trials on MS treatments
`since 2001 that failed or were interrupted, attempts to analyze the underlying reasons for failure, and discusses
`the implications for our current view of MS pathogenesis, clinical practice, and design of future studies. In order
`to maintain clarity, this review focuses on anti-inflammatory therapies and does not include studies on already
`approved and effective disease-modifying therapies, albeit used in distinct administration routes or under dif-
`ferent paradigms. Neuroprotective and alternative treatment strategies are presented elsewhere.
`
`1. Immunopathology of Multiple Sclerosis and
`Therapeutic Targets
`
`The therapeutic options for multiple sclerosis (MS) have
`been widened significantly over the past decade. However, the
`approved therapeutic agents (beta-interferons [IFNb], glatir-
`amer acetate, mitoxantrone, natalizumab) still have limited
`efficacy in preventing disease progression, and some of them
`are associated with either a considerable long-term toxicity or a
`still unclear risk-benefit ratio. There is a tremendous activity in
`the search for new therapeutics,[1,2] which is reflected by the
`soaring number of publications. However, one has to realisti-
`cally concede that few successful agents in MS stand apart from
`a large number of therapeutic disappointments.[3-5] Despite
`rational pathophysiologic concepts, conclusive data from
`animal models, promising phase I/II studies, and successful
`application in other autoimmune diseases, several trials testing
`new compounds in MS patients have shown no benefit. On the
`other hand, some effective treatments are associated with un-
`expected or unexpectedly severe adverse effects. Whereas pos-
`itive studies usually make it into prestigious journals, many
`negative trials are published merely as abstracts or are not
`published at all.[6] This is unfortunate because there is a lot to
`learn from negative results, and a critical reflection is highly
`important for understanding human MS immunopathogenesis
`and to help improve future clinical trial design.
`We here discuss the pathophysiologic rationale, the experi-
`mental basis, and the trial data of novel agents in MS therapy
`
`that were not effective and/or were associated with considerable
`unexpected adverse effects when tested in human phase I–III
`1
`This review focuses on
`studies in MS between 2001 and 2010.
`immunomodulatory strategies; neuroprotective and alternative
`treatment targets will be discussed in a separate article.
`
`2. Modulation of T-Cell Differentiation and T Helper
`(Th)-1/Th2 Balance
`
`One of the pivotal steps in the initial autoimmune inflam-
`matory pathogenesis of MS is the activation of autoreactive
`T cells in the periphery via T-cell receptor (TCR)-mediated
`recognition of major histocompatibility complex (MHC)-I
`presented antigens – possibly misled by antigenic mimicry.
`After transmigration across the blood-brain barrier and re-
`activation in the CNS, both CD4+ T helper (Th) cells and CD8+
`cytotoxic T cells trigger demyelination and primary axonal
`damage via a shift to a proinflammatory Th1 cytokine en-
`vironment.[7] This process seems to be perpetuated by dysre-
`gulation of apoptotic mechanisms in T cells. Therefore,
`modulation of T-cell differentiation and rebalancing of Th1 and
`Th2 response represents a critical mechanism for therapeutic
`intervention. Broad depletion of autoreactive T cells may be
`achieved by means of monoclonal antibodies against specific T-
`cell markers. After the overall negative results with the mono-
`clonal antibodies against CD3 (muromonab CD3) and CD4
`(priliximab) in MS trials,[5] other T-cell targets such as CD52
`
`1 Search strategy and selection criteria: studies were identified by a search of PubMed for publications published over the period January 2001 to
`March 2010, using the terms ‘multiple sclerosis’ and ‘therapy’ or ‘treatment’, and ‘trial’. Eligible studies were also identified from conference
`information and personal communications of the authors. Abstracts and reports from meetings were included, especially since failed trials are
`often not published in peer reviewed journals. Studies were excluded if they were not published in English.
`
`ª 2010 Adis Data Information BV. All rights reserved.
`
`Biodrugs 2010; 24 (4)
`
`Page 2 of 26
`
`

`

`Therapeutic Approaches to Multiple Sclerosis
`
`251
`
`and proinflammatory Th1 cytokines attracted considerable
`attention (table I).
`
`2.1 Interleukin-12/23: p40 Neutralizing Monoclonal Antibody
`(Ustekinumab)
`
`2.1.1 Background
`Two main proinflammatory populations of CD4+ T cells are
`Th1 and Th17 cells. Interleukin (IL)-12 and IL-23 are two
`cytokines involved in the differentiation of these two T-cell
`subsets. IL-12 and IL-23 are closely related, are secreted by
`myeloid cells and bind to specific receptors expressed on T cells.
`IL-12 has long been recognized as essential for generation of
`Th1 cells secreting interferon-g (IFNg), whereas IL-23 has re-
`cently been shown to induce a specific T-cell subset producing
`IL-17.[13,14] Both cytokines are heterodimers consisting of a
`common subunit (p40) and either p35 (IL-12) or p19 (IL-23).
`The common IL-12/IL-23 subunit p40 is detected in MS pla-
`ques, and administration of IL-12 can induce relapses in ex-
`perimental autoimmune encephalitis (EAE), an animal model
`of MS.[15] Therefore, p40 blockade has been expected to be a
`strategy for modulating autoimmune processes in EAE and
`MS.[16] Ustekinumab (CNTO-1275) is a human monoclonal
`antibody directed against the common IL-12/IL-23 p40 subunit
`that has been shown to prevent clinical disease and develop-
`ment of hyperintense lesions on a T2-weighted MRI in a mar-
`moset model of EAE.[17]
`
`2.1.2 Studies
`Tolerability of ustekinumab administered subcutaneously
`was proven in a phase I trial in relapsing-remitting MS (RRMS)
`patients.[8] Based on this study, Segal and coworkers[9] tested
`four widely spread doses (27–180 mg) of ustekinumab in 249
`patients with definite RRMS (Expanded Disability Status Scale
`[EDSS] range 0–6.5) over 19 weeks. Analysis of this random-
`ized, double-blind, placebo-controlled, dose-ranging, phase II
`study for new gadolinium-enhancing T1-weighted lesions
`(primary endpoint) revealed no significant difference compared
`with placebo, for any of the dosage regimens. Regarding clin-
`ical parameters, the authors observed no median change in
`EDSS from baseline to week 23, and most patients developed
`one relapse but not more than two relapses by week 23 without
`a significant difference across all subgroups. A high number of
`adverse events, predominantly infections, were reported in both
`arms (placebo 78% and verum 83%). However, serious adverse
`events occurred only in 3% and 2% of the patients treated with
`ustekinumab and placebo, respectively.[9]
`
`2.1.3 Comment
`One possible reason for the negative results found in this
`study might be a reduced CNS penetration of ustekinumab with
`subcutaneous administration, since the positive results in the
`marmoset model of EAE were obtained with intravenous ad-
`ministration. However, active MS lesions are associated with
`disruption of the blood-brain barrier, which should permit
`significant penetration of the antibody to potential sites of ac-
`tion. Alternatively, the neutralizing antibody might have been
`administered outside the therapeutic window, since IL-12 and
`IL-23 expression in CNS may precede opening of the blood-
`brain barrier. By contrast, ustekinumab seems to be active in
`EAE as well as in psoriasis and inflammatory bowel disease,
`suggesting pathogenic and immunologic differences in these
`entities. In general, basic differences in the immunopathogen-
`esis of EAE and MS have to be kept in mind, especially con-
`cerning the relative importance of Th17 cells in EAE versus
`MS.[18]
`It may also be possible that distinct actions of the IL-12
`heterodimer and its subunits at the IL-12 receptor (IL-12R)
`result in a lack of effect of this neutralizing antibody. As
`mentioned above, IL-12 consists of a p40 subunit (which is
`shared by IL-23) and a p35 subunit forming the IL-12 p70
`heterodimer. Levels of IL-12 p40 and IL-12 p70 are in-
`dependent, and p40 is produced in excess over IL-12 p70 by
`5- to 500-fold, which can result in the formation of IL-12 p80
`homodimers.[19] The IL-12R consists of a b1 and a b2 subunit,
`of which only the latter transmits intracellular signals upon
`ligation.[20] The IL-12 p40 subunit exclusively binds to the
`IL-12Rb1 subunit, which lacks signal transmission if the b2
`subunit is disengaged. Excess IL-12 p40 is thus able to com-
`petitively antagonize binding of IL-12 p70 heterodimer to its
`receptor.[21] In turn, IL-12 p80 homodimers are able to occupy
`IL-12R to a higher affinity and block signal transmission by
`noncompetitive antagonism to IL-12 p70 heterodimers[22] and
`thus act in an anti-inflammatory way. The functional relevance
`of antagonization of IL-12 p70 signaling by excess IL-12 p40
`monomers or homodimers has been shown in vitro.[23,24] Thus,
`neutralization of IL-12 p40 by ustekinumab might pre-
`dominantly block the IL-12 antagonizing activity of a p40 ex-
`cess and outscale the net blocking effect on proinflammatory
`IL-12 p70 signaling.
`
`2.2 Phosphodiesterase Inhibitors (Ibudilast, Rolipram)
`
`2.2.1 Background
`Phosphodiesterases (PDEs) are involved in the regulation of
`intracellular levels of the second messengers cyclic adenosine
`
`ª 2010 Adis Data Information BV. All rights reserved.
`
`Biodrugs 2010; 24 (4)
`
`Page 3 of 26
`
`

`

`252
`
`Ref./clinical
`trial ID
`[NCTy]
`
`Further/ongoing
`trials
`
`Comment
`
`Finished
`
`Negative results: no clinical
`efficacy
`
`8
`
`clinical adverse
`effects
`
`Ustekinumab was
`well tolerated
`
`Table I. Modulation of T-cell differentiation and T helper (Th)-1/Th2 balance
`
`Agent
`
`(Assumed) mechanism
`of action
`
`Characteristics
`
`Disease
`course
`
`Outcome
`
`MRI
`
`IL-12/-23 p40
`neutralizing
`mAb
`(ustekinumab,
`CNTO-1275)
`
`Blockade of
`differentiation of naive T
`cells to Th1 cells (IL-12),
`modulation of
`macrophage function by
`blockade of IL-23
`
`RRMS
`
`RRMS
`
`Phase I, db, pc,
`sequential dose
`escalation;
`20 pts
`
`Phase II, r, db,
`pc, mc; 249 pts;
`repeated SC
`administration
`
`High variability in T2
`lesion volume and
`total number of
`GdEL
`
`9 0
`
`0207727
`
`Finished
`
`Negative, new GdEL No significant
`differences for new
`GdEL T1 lesions
`and no significant
`effect on clinical
`parameters
`
`ol, co; 18 pts
`
`RRMS,
`SPMS
`
`Negative
`
`PDE inhibitors
`(ibudilast)
`
`Downregulation of
`inflammatory responses
`by changing levels of
`cAMP and cGMP;
`shifting the cytokine
`milieu towards Th2-
`driven responses
`
`Atorvastatin
`(alone or add-
`on to IFNb-1a
`SC)
`
`Modulation of HMG-CoA
`reductase-dependent
`T-cell signaling
`pathways, shift of Th1 to
`Th2 response
`
`Phase II, ol, btt;
`36 pts
`
`RRMS
`
`Reduction in number
`and volume of GdEL
`when all or IFN-
`treated pts were
`analyzed. No
`significant effect in
`pts without IFN
`
`In all pts and all
`strata, a
`significantly
`improved MSFC
`was found,
`whereas EDSS
`was not
`significantly
`influenced
`
`Terminated due
`to lack of clinical
`efficacy; dose-
`dependent
`adverse effects,
`e.g. nausea and
`emesis
`
`10
`
`Regarded as
`immunomodulating
`treatment; first-generation of
`PDE inhibitors with
`considerable adverse
`effects; second-generation
`compounds are improved in
`this regard
`
`Finished
`
`No parallel groups, effect
`possibly due to IFN
`treatment, short observation
`period with low number of pts
`
`11
`00616187
`
`Ulzheimer et al
`
`RRMS
`
`r, db, pc, 0 mg/
`40 mg/80 mg,
`add-on to IFN;
`26 pts
`
`Negative; more new
`T2 lesions or GdEL
`in verum group
`(8/17) vs placebo
`(1/9)
`btt = baseline-to-treatment; cAMP = cyclic adenosine monophosphate; cGMP = cyclic guanosine monophosphate; co = crossover; db = double-blind; EDSS = Expanded Disability Status
`Scale; GdEL = gadolinium-enhancing lesions; ID = identifier; IFN = interferon; IL = interleukin; mAb = monoclonal antibodies; mc = multicenter; mo = month(s); MRI = magnetic resonance
`imaging; MSFC = multiple sclerosis functional scale; ol = open-label; pc = placebo-controlled; PDE = phosphodiesterase; pt(s) = patient(s); r = randomized; Ref. = reference; RRMS =
`relapsing-remitting multiple sclerosis; SC = subcutaneous; SPMS = secondary-progressive multiple sclerosis.
`
`Finished
`
`Negative; more
`relapses in verum
`group (4/17) vs
`placebo (1/9)
`
`High number of pts not
`adhering to study drug
`protocol, low number of pts
`
`12
`
`ª 2010 Adis Data Information BV All rights reserved
`
`Biodrugs 2010; 24 (4)
`
`Page 4 of 26
`
`

`

`Therapeutic Approaches to Multiple Sclerosis
`
`253
`
`monophosphate (cAMP) and cyclic guanosine monophos-
`phate (cGMP) by hydrolysis of the respective cyclic nucleo-
`tides. The 11 known PDE subtypes differ in their substrate
`specificity and their pharmacologic properties, e.g. PDE-4 is
`activated by elevated levels of cAMP and inhibited by rolipram.
`Inhibition of PDE reduces tumor necrosis factor-a (TNFa)
`production by activated monocytes and macrophages, resulting
`in a lower immune response and a shift of the cytokine milieu to
`Th2-driven responses. Treatment with PDE inhibitors has
`previously shown clinical and histopathologic amelioration in
`several EAE models.[25] In human MS, the unspecific PDE
`inhibitor ibudilast was shown to influence cytokine production
`of T-cell lineages and natural killer cells.[10]
`
`pathways in T cells.[28,29] Simvastatin, for example, has been
`shown to interfere with IL-17 production of human T lym-
`phocytes,[30] and other statins shift the cytokine response to-
`wards a Th2 pattern.[31,32] Moreover, statins interfere with cell
`infiltration via the blood-brain barrier by downregulation of
`cell adhesion molecules like lymphocyte function-associated
`antigen-1 (LFA-1)[33] and reduction of chemokine produc-
`tion by endothelial cells.[34] Statins have also been shown to be
`effective in the EAE model,[31,35] and it is known from clinical
`practice that they generally have good tolerability and an ex-
`cellent safety record. It therefore seemed reasonable to in-
`vestigate a potential beneficial effect of statins on MS in clinical
`trials.
`
`2.2.2 Studies
`An open-label, crossover, phase I/II clinical trial of the
`specific PDE-4 inhibitor rolipram had to be terminated pre-
`maturely because of lack of clinical efficacy, after enrolling only
`eight MS patients. Unexpectedly, the number of contrast-
`enhancing lesions increased significantly (0.44–1.71 lesions/
`patient/month), while rolipram was otherwise immunologically
`active and inhibited Th1 and Th17 cells in MS patients.[26]
`Furthermore, the acceptance of the oral formulation was
`hampered by dose-dependent adverse effects, e.g. nausea and
`emesis.
`
`2.2.3 Comment
`The first clinical evaluations of PDE inhibitors showed that
`they have to be regarded as immunomodulating treatment, but
`they also display considerable adverse effects. However, the
`observed dissociation between expected immunologic effects
`and the negative clinical outcome measures raises concerns
`about the clinical perspective of these drugs. Besides the ob-
`served lack of efficacy in MS, clinical development has also
`been halted in depression, the other major target of PDE in-
`hibitors.[27] The second-generation compounds, which target
`only a subset of PDE-4 enzymes, are improved in this regard
`and could perhaps be worthwhile to try in a well designed MS
`treatment trial.
`
`2.3 HMG-CoA-Dependent T-Cell Signaling: Statins
`(Atorvastatin)
`
`2.3.1 Background
`HMG-CoA reductase inhibitors (statins) are known to
`have pleiotropic effects in vivo, and considerable experimen-
`tal evidence points towards an immunomodulatory influence
`of statins by influencing HMG-CoA-dependent signaling
`
`2.3.2 Clinical Trials
`An early open-label, single-arm, crossover study compared
`disease activity by MRI in 30 RRMS patients before and after
`6 months of treatment with simvastatin.[36] The number and
`volume of gadolinium-enhancing lesions declined by ~40%.
`Four years later, a randomized, double-blind pilot study was
`launched including 26 subjects with RRMS receiving atorva-
`statin versus placebo as add-on therapy to IFNb-1a therapy.[12]
`Surprisingly, statin-treated patients showed a significantly in-
`creased relapse rate and an increased number of new lesions as
`assessed by MRI. These unexpected results are in contrast to
`other clinical studies, which underline clinical safety and effi-
`cacy of statin treatment in MS.[11,36,37]
`
`2.3.3 Comment
`A possible explanation for these conflicting data on statins in
`neuroinflammation might be related to putative proinflam-
`matory and harmful effects of statins. They have been reported
`to increase IFNg, IL-12,[38] and IL-12p70[39] production, and to
`augment the proteolytic activity of matrix metalloproteinases
`(MMPs).[40] Moreover, statins were shown to hamper CNS
`remyelination by blocking oligodendrocyte progenitor cell
`differentiation[41] and mature oligodendrocyte function. In
`summary, oral add-on therapies with clinically approved agents
`in other indications, like statins, still represent an attractive
`strategy for improving MS therapy, but careful studies will be
`necessary to rule out a putative harmful interaction between
`statin and interferon treatment in MS.
`
`3. Modulation of T-Cell Activation
`
`Autoreactive T cells in the systemic immune compartment
`recognize specific autoantigens presented by MHC class II
`
`ª 2010 Adis Data Information BV. All rights reserved.
`
`Biodrugs 2010; 24 (4)
`
`Page 5 of 26
`
`

`

`254
`
`Ulzheimer et al.
`
`molecules on the surface of antigen-presenting cells. Activation
`of T cells as well as B cells requires a dual signaling, one signal of
`which comes from a ‘trimolecular complex’ consisting of the
`TCR complex and its co-receptors CD4 and CD8 bound to the
`antigenic peptide presented by the MHC molecule. The second
`signal originates from soluble factors such as IL-2, or from
`binding of co-stimulatory cell surface ligands.[42] These co-
`stimulatory signals seem to be critical for the regulation of
`T-cell activation and for the balance between Th1 and Th2
`cell differentiation.[43] Therefore, blockade of co-stimulatory
`pathways may be an interesting therapeutic approach for
`treating autoimmune disorders such as MS. A well character-
`ized co-stimulatory pathway includes CD28 on T cells, which
`has two ligands on antigen-presenting cells: B7-1 (CD80)
`and B7-2 (CD86). Alternatively, an additional counter-receptor
`on T cells, cytotoxic T-lymphocyte antigen 4 (CTLA4), can
`bind both CD80 and CD86, acting as a negative regulator of T-
`cell function. In this regard, mainly the CD28-CTLA-4/B-7 and
`the CD40-CD40-ligand system revealed a promising target,
`since in the EAE model, recombinant or pharmacologic mod-
`ulation of the CD28-B-7 network influenced disease develop-
`ment and progression.[44,45] Further, both systems contribute to
`the (dys)regulation of autoreactive T cells in MS[46-48] (table II).
`
`3.1 Cytotoxic T-Lymphocyte Antigen 4 (CTLA4): Chimeric
`CTLA4-Ig (Abatacept, RG2077)
`
`3.1.1 Background
`CTLA4 (CD152) is expressed on T cells and is similar to the
`co-stimulatory molecule CD28, as they both are able to bind to
`CD80 and CD86 on antigen-presenting cells. CTLA4, however,
`is known to be an important negative regulator of T-cell
`function[44,45] and has also been implicated in the mechanism of
`action of CD4+CD25+ regulatory T cells (Treg) cells.[55] In the
`EAE model, anti-CTLA4 treatment exacerbates the severity of
`the disease.[56-58] In addition, certain CTLA4 gene poly-
`morphisms seem to be associated with human MS.[48,59,60]
`Taken together, these observations highlight the potential role
`of CTLA4 in the control of autoimmunity.
`The B7/CD28-CTLA4 pathway can be modulated by
`CTLA4-Ig, a chimeric protein consisting of the extracellular
`domain of human CTLA4 fused to the Fc region of human
`IgG-1.[61,62] CTLA4-Ig inhibits T-cell activation by binding
`with higher affinity to B7-1 and B7-2 than CD28. In patients
`with refractory rheumatoid arthritis, the CTLA4-Ig protein
`abatacept (RG2077) demonstrated significant clinical benefits
`(for review see Westhovens and Verschueren[49]) and was thus
`approved by the US FDA in December 2005. It is also currently
`
`under investigation for the treatment of type 1 diabetes mellitus
`and for ulcerative colitis.
`
`3.1.2 Studies
`A small pilot study assessed the safety and immune mech-
`anisms of CTLA4-Ig (abatacept) and has recently been pub-
`lished.[50] In this study, 20 subjects with RRMS received either
`single intravenous infusions of abatacept 2, 10, 20, or 35 mg/kg
`or a multi-dose of 10 mg/kg. No major adverse effects were
`observed and immunologic assessment showed a reduction in
`the proliferation of myelin basic protein (MBP)-specific T-cell
`lines with a reduced production of IFNg.
`A double-blind, placebo-controlled, multicenter, phase II
`trial of abatacept included 219 patients with RRMS, random-
`ized between abatacept at one of two doses (2 or 10 mg/kg)
`or placebo, administered by infusion on days 1, 15, and 29,
`and then every 4 weeks until day 197.[51] Of the 219 patients in
`the study, 127 received at least one infusion of the study med-
`ication. The study, albeit thoroughly designed and adequately
`powered, was prematurely terminated by the safety board
`because of an increased relapse rate and inflammatory MRI
`activity in the low-dose verum group. Preliminary efficacy
`analysis revealed that, compared with patients in the 2 mg/kg
`arm and the placebo group, those in the 10 mg/kg group had
`fewer new gadolinium-T1 enhancing lesions and fewer relapses.
`
`3.1.3 Comment
`The abatacept trial in RRMS was halted prematurely be-
`cause of the occurrence of higher relapse rates in one of the two
`verum groups (lower dose). Initially, the investigators were
`concerned about disease (re)activation by CTLA4-Ig, similar to
`that seen earlier in the TNFa MS trial.[63] However, unblinding
`of the patient cohort revealed that the low-dose group already
`had higher disease activity at the time of study inclusion; for
`example, 80% of patients with more than 10 gadolinium-T1
`enhancing lesions were randomized to the low-dose abatacept
`treatment group. Therefore, rather than a true ‘treatment fail-
`ure’ or CTLA4-Ig-induced immune exacerbation, these results
`likely occurred because of a randomization problem. Final
`interpretation of the study is still pending and published data
`on CTLA4-Ig in MS is too scarce to allow any firm conclusions
`regarding its safety and potential efficacy. Trial experiences in
`other autoimmune diseases as well as in transplantation gen-
`erally indicate that this therapy has considerable potential.[62,64]
`The experience with CTLA4-Ig illustrates the risk of in-
`appropriately stopping a trial because of a small number of
`adverse events, which instead of being caused by the study drug
`
`ª 2010 Adis Data Information BV. All rights reserved.
`
`Biodrugs 2010; 24 (4)
`
`Page 6 of 26
`
`

`

`Therapeutic Approaches to Multiple Sclerosis
`
`Ref./clinical
`trial ID
`[NCTy]
`
`49,50
`
`51
`00035529
`
`Characteristics
`
`Disease
`course
`
`Outcome
`
`MRI
`
`clinical adverse effects
`
`Further/ongoing
`trials
`
`Comment
`
`RRMS
`
`RRMS
`
`Pilot study, 20 pts,
`single infusions (2, 10,
`20, 35 mg/kg) or multi-
`dose of 10 mg/kg
`
`db, pc, mc, phase II
`trial, 330 pts, (2 or
`10 mg/kg infusions on
`days 1, 15, 29 and
`then every 4 wk until
`day 197)
`
`No major adverse
`effect
`
`Finished
`
`Accumulation of
`inflammatory MRI
`activity (low-dose
`verum group); fewer
`new GdEL in
`10 mg/kg group
`
`Accumulation of
`relapses in low-dose
`verum group; less
`relapses in 10 mg/kg
`group
`
`Study was
`prematurely
`halted
`
`Reason for worse
`outcome in the verum
`group probable due to
`randomization failure; the
`clinical benefit in MS
`remains unclear
`
`Table II. Modulation of T-cell activation
`
`Agent
`
`CTLA4-Ig
`(abatacept,
`RG2077)
`
`(Assumed)
`mechanism of
`action
`
`Negative
`regulator of T-
`cell function;
`effects on
`CD4+CD25+
`Treg cells
`
`Anti-CD40L
`(anti-CD154,
`toralizumab)
`
`Antibody
`interacting
`with the
`co-stimulatory
`pathway
`CD40-CD40L
`
`Pilot study (IDEC-
`131) in 15 pts
`
`RRMS
`
`Positive
`
`No relapses for at least
`6 mo
`
`Finished
`
`Potential interference
`with the thrombocyte
`system
`
`RRMS
`
`46 pts, db, pc, phase II
`trial (15 mg/kg) IV for
`5 wk and then every
`mo for 3 mo
`
`Study was halted
`because 1 pt
`developed
`thromboembolism in
`another study of Crohn
`disease (later, 2 more
`pts)
`
`Stopped,
`although all pts
`had pre-existing
`risk factors for
`clotting
`
`PPAR-g agonists:
`thiazolidinediones
`(pioglitazone,
`rosiglitazone)
`
`Inhibition of T-
`cell activation,
`reduction of
`proinflammatory
`cytokines
`
`Phase I/II pilot study,
`21 pts taking IFNb-1a
`using pioglitazone.
`Observation period:
`1 y
`
`RRMS
`
`Positive
`
`Finished
`
`Clinical potential in MS
`unclear, broad
`experience in other
`disease entities,
`candidate for
`continuation
`
`52
`
`53
`
`54
`
`255
`
`db = double-blind; CTLA4 = cytotoxic T-lymphocyte antigen 4; GdEL = gadolinium-enhancing lesions; ID = identifier; IFN = interferon; IV = intravenous; mc = multicenter; mo = month(s);
`MRI = magnetic resonance imaging; MS = multiple sclerosis; pc = placebo-controlled; PPAR = peroxisome proliferator-activated receptor; pt(s) = patient(s); Ref. = reference; RRMS =
`= regulatory T cells; wk = week(s); y = year(s).
`relapsing-remitting MS; Treg
`
`ª 2010 Adis Data Information BV All rights reserved
`
`Biodrugs 2010; 24 (4)
`
`Page 7 of 26
`
`

`

`256
`
`Ulzheimer et al.
`
`could also be due to chance, unbalanced baseline character-
`istics, or complex dose-response interactions. This trial em-
`phasizes the need to utilize trial designs that protect against this
`occurrence (e.g. adaptive designs).
`
`3.2 CD40-CD40L: Blocking Monoclonal Anti-CD154
`Antibody (Toralizumab)
`
`3.2.1 Background
`CD40L (CD154) is a member of the TNF family of cell surface
`interaction molecules, which is expressed on CD4 T cells, B cells,
`macrophages, and dendritic cells.[65,66] The CD40-CD40L path-
`way has been shown to have multiple roles in the immune system,
`e.g. it enhances the antigen-specific T-cell response thr

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