throbber
USOO8182814B2
`
`US 8,182,814 B2
`(10) Patent No.:
`(12) United States Patent
`
`Baca et al.
`(45) Date of Patent:
`May 22, 2012
`
`(54) METHODS OF TREATING INFLAMMATORY
`AIRWAY CONDITIONS BY INHIBITION OF
`IL-11 ACTIVITY
`
`(58) Field of Classification Search ........................ None
`See application file for complete search history.
`
`(75)
`
`Inventors: Manuel Baca, Gaithersburg, MD (US);
`Andrew Donald Nash, Kew (AU); Jack
`A. Elias, Woodbridge, CT (US)
`
`(73) Assignees: CSL Limited, Parkville, Vlctoria (AU);
`Yale University, New Haven, CT (US)
`
`( * ) Notice:
`
`Subject to any disclaimer, the term of this
`patent is extended or adjusted under 35
`U.S.C. 154(b) by 0 days.
`
`(21) App1.No.: 12/256,883
`
`(22)
`
`Filed:
`
`Oct. 23, 2008
`
`(65)
`
`Prior Publication Data
`
`US 2009/0202533 A1
`
`Aug. 13, 2009
`
`Related U.S. Application Data
`
`(60) Provisional application No. 61/000,588, filed on Oct.
`26, 2007.
`
`(51)
`
`Int. Cl.
`(2006.01)
`A61K 38/20
`(2006.01)
`C07K 14/715
`(2006.01)
`C07K 14/54
`(2006.01)
`C07K 16/28
`(2006.01)
`C07K 16/24
`(52) U.S. Cl.
`............... 424/1431; 424/1331; 424/139.1;
`424/141.1; 424/145.1; 530/351; 530/387.3;
`530/388.22; 530/388.23
`
`(56)
`
`References Cited
`
`U.S. PATENT DOCUMENTS
`
`5,679,339 A * 10/1997 Keith et 31.
`.................. 424/852
`6,540,993 B1 *
`4/2003 W3rne et 31.
`..... 424/852
`
`7,252,820 B2 *
`8/2007 Boodhoo et 31.
`424/9467
`7,393,532 B1 *
`7/2008 de Sauv3ge et 31.
`....... 424/1441
`7,612,181 B2 * 11/2009 Wu et 31.
`.................... 530/387.3
`
`OTHER PUBLICATIONS
`
`Molet et 31., J. Allergy Clin. Immunology 2001, 108:430-438.*
`Wang, J. et 31. IL-11 selectively inhibits 3er0311ergen-induced pul-
`m0n3ry eosinophilia 3nd Th2 cytokine production. J Immunol 165,
`2222-31 (2000).*
`Minsh311 et 31., J. Allergy Clin. Immunology 2000, 105:232-238.*
`Kyung Sun Lee et 31., “Cysteinyl leukotriene upregulates IL-11
`expression in allergic airway disease of mice”, J. Allergy Clin.
`Immunol., 119(1): 141-149 (J3n. 1997; published online on Oct. 27,
`2006).
`
`* cited by examiner
`
`Primary Examiner 7 Lorraine Spector
`(74) Attorney, Agent, or Firm 7 Scu11y, Scott, Murphy &
`Presser, PC.
`
`(57)
`
`ABSTRACT
`
`The present invention provides a method for the treatment or
`prophylaxis of T—helper type 2 (Th2)-mediated disorders
`using antagonists of IL-1 1.
`
`16 Claims, 4 Drawing Sheets
`
`Lassen — Exhibit 1005, p. 1
`
`Lassen - Exhibit 1005, p. 1
`
`

`

`US. Patent
`
`May 22, 2012
`
`Sheet 1 of4
`
`US 8,182,814 B2
`
`24 HOURS
`
`D'O'OzO{OQA
`
`o 0
`
`v‘
`
`1'}A
`
`
`
`
`
`BALCellRecovery(x1004)
`
`(”NOD
`
`coo
`
`50.0
`
`40.0
`
`30.0
`
`20.0
`
`10.0
`
`0.0
`
`TOTALCELLS MACROPHAGE
`
`EOSINOPHIL
`
`LYMPHOCYTES NEUTROPHILS
`
`|L-11raKO+OVA
`
`DWT+SaIine
`
`.WT+OVA
`
`|L-11raKO+Sa|ine
`
`FIG. 1
`
`Lassen — Exhibit 1005, p. 2
`
`Lassen - Exhibit 1005, p. 2
`
`

`

`US. Patent
`
`May 22, 2012
`
`Sheet 2 of4
`
`US 8,182,814 B2
`
`48 HOURS
`
`" 70.0
`
`x1004V
`
`
`
`BALCellRecovery
`
`1 o :35:A.0‘
`
`'4o9‘
`
`A A
`
`60.0
`
`50.0
`
`40.0
`
`30.0
`
`20.0
`
`10.0
`
`0.0
`
`TOTAL CELLS
`
`MACROPHAG
`
`EOSINOPHIL
`
`LYMPHOCYTES
`
`NEUTROPHILS
`
`|L-11ra KO+OVA
`
`FIG. 2
`
`Lassen — Exhibit 1005, p. 3
`
`Lassen - Exhibit 1005, p. 3
`
`

`

`US. Patent
`
`May 22, 2012
`
`Sheet 3 of4
`
`US 8,182,814 B2
`
`72 HOURS
`
`90.0
`
`
`
`
`
`BALCellRecovery(x1004)
`
`A
`
`80.0
`
`0103\l
`
`coo
`
`40.0
`
`30.0
`
`20.0
`
`10.0
`
`.
`
`FIG. 3
`
`Lassen — Exhibit 1005, p. 4
`
`Lassen - Exhibit 1005, p. 4
`
`

`

`US. Patent
`
`May 22, 2012
`
`Sheet 4 of4
`
`US 8,182,814 B2
`
`24 Hours
`
`,
`
`4—.
`a1.
`5—
`EC
`
`3DU '
`
`5U
`
`BAL Inflammation
`
`lclsaline
`I CIDVA
`[II] Misaline
`MIDVA
`
`MN“GU'IUDO:
`
`Figure 4
`
`Lassen — Exhibit 1005, p. 5
`
`Lassen - Exhibit 1005, p. 5
`
`

`

`US 8,182,814 B2
`
`1
`METHODS OF TREATING INFLAMMATORY
`AIRWAY CONDITIONS BY INHIBITION OF
`IL-11 ACTIVITY
`
`APPLICATION DATA
`
`This application claims priority from and the benefit of
`US. Provisional Patent Application No. 61/000,588, filed
`Oct. 26, 2007, the entire contents of which are incorporated
`herein by reference.
`
`FIELD
`
`The present invention provides a method for the treatment
`or prophylaxis of T—helper type 2 (Th2)-mediated disorders
`using antagonists of lL-l 1.
`
`BACKGROUND
`
`Bibliographic details of references provided in the subject
`specification are listed at the end of the specification.
`Reference to any prior art is not, and shouldnot be taken as,
`an acknowledgment of or any form of suggestion that this
`prior art forms part of the common general knowledge in any
`country.
`Th2 cytokines, lL-4, lL-5, lL-9 and IL- 1 3, are derived from
`T helper type 2 (Th2) cells, although they may also derive
`from other cell types. These Th2 cytokines play an important
`role in the pathophysiology of allergic diseases including
`asthma.
`Asthma is a chronic disease that involves inflammation of
`
`the pulmonary airways and bronchial hyper-responsiveness
`leading to reversible obstruction of the lower airways (re-
`viewed in Bousquet et al, Am JRespir Crit Care Med 161(5):
`1720-1745, 2000). In a diagnostic context bronchial hyper-
`responsiveness is evidenced by decreased bronchial airflow
`following exposure to methacholine or histamine. Natural
`triggers that provoke airway obstruction include respiratory
`allergens, cold air, exercise, viral upper respiratory infection,
`and cigarette smoke. Bronchial provocation with allergen
`induces a prompt early phase immunoglobulin E (lgE)-me-
`diated decrease in bronchial airflow followed in many
`patients by a late-phase lgE-mediated reaction with a
`decrease in bronchial airflow for 4-8 hours.
`
`Asthmatic airways display lung hyperinflation, smooth
`muscle hypertrophy,
`fibrosis in the lamina reticularis,
`mucosal edema, epithelial cell sloughing, cilia cell disrup-
`tion, and mucus gland hypersecretion. Microscopically,
`asthma is characterized by the presence of increased numbers
`of eosinophils, mast cells, neutrophils,
`lymphocytes, and
`plasma cells in the bronchial tissues, bronchial secretions,
`and mucus. Activated CD4 T—lymphocytes that produce a Th2
`pattern of cytokines appear to be central to the initiation,
`development and maintenance of the disease phenotype
`(Robinson et al, NEnglJMed 326(5):298-304, 1992; Wills-
`Karp et al, Science 282(5397):2258-2261, 1998; Hamid et al
`J Clin Invest 87(5):1541-1546, 1991; Ray and Cohn, J Clin
`Invest 104(8):985-993, 1999). For example, the cytokines
`produced by these cells (including lL-4, lL-5, lL-9 and lL-13)
`regulate infiltration and mediator release by inflammatory
`cells and allergen specific antibody isotype switching from
`lgM to lgE. The activity of non-hemopoietic cells, for
`example mucus hypersecretion by goblet cells, is also regu-
`lated by Th2 cytokines.
`Regardless ofthe triggers of asthma, the repeated cycles of
`inflammation in the lungs with injury to the pulmonary tis-
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`45
`
`50
`
`55
`
`60
`
`65
`
`2
`
`sues followed by repair may produce long-term structural
`changes (“remodeling”) of the airways.
`In the most widely used animal model of human asthma,
`mice are sensitized to ovalbumin (ova, formulated in alum
`adjuvant) via the intraperitoneal route on one or more occa-
`sions.An allergic airway response is subsequently induced by
`single or repeated exposure to aerosol ova (generated via and
`ultrasonic nebulizer). Response parameters assessed over the
`subsequent 24-72 hr period include, for example, the accu-
`mulation of inflammatory cells and mediators in bronchoal-
`veolar lavage (BAL)
`fluid, bronchorestriction following
`intravenous administration of methacholine (airway hyper-
`reactivity) and ova specific serum lgE. Histological demon-
`stration ofinflammatory cell accumulation in lung tissues and
`goblet cell hyperplasia/metaplasia and associated mucus
`hypersecretion are also key characteristics of the mouse air-
`way response to ova. Large animal models of asthma (for
`example non-human primates and sheep), where lung archi-
`tecture, circulation and innervation more closely resemble
`that of humans, have been described but are less widely used.
`At the time studies described in this specification were per-
`formed there were no reports of the analysis of lL-l 1 antago-
`nists in either small or large animal models of asthma.
`lL-l 1 is a pleiotropic cytokine produced by a wide variety
`of cell types including fibroblasts, epithelial cells, chondro-
`cytes, endothelial cells, osteoblasts and certain tumor cells
`and cell lines (reviewed in Neben and Turner, Stem Cells.
`Suppl 2:156-62 1993, Du and Williams, Blood. 83(8):2023-
`2030, 1994). Human lL-11 is synthesized as a 19 kDa 199
`amino acid precursor protein, with a 21 amino acid leader
`sequence that is removed to generate a mature secreted pro-
`tein of 178 amino acids. lL-11 is highly conserved across
`speciesithe mature human and murine proteins share 88%
`homology at the amino acid level, while human and non-
`human primate lL-11 share 94% homology. Although the
`crystal structure of lL-11 has not been solved a variety of
`approaches (e.g. computer modeling and alanine scanning
`mutagenesis) suggest a 4 ot-helical bundle structure typical of
`many cytokines (Czupryn et al, Ann N YAcad Sci 762: 152-
`1 64, 1995).
`lL-11 was originally described as a soluble factor derived
`from stromal cells, which was capable of stimulating plasma-
`cytoma cell proliferation (Paul et al, Proc. Nat. Acad Sci.
`87:7512-7516, 1990). A variety of diverse biological proper-
`ties have subsequently been ascribed to lL-11 including: the
`ability to stimulate hemopoiesis, thrombopoiesis, megakary-
`opoiesis (Nandurkar et al, Blood 90:2148, 1997; Nakashima
`et al, Semin Hematol35(3):210-221, 1998), and bone resorp-
`tion (Sims et al, JBone Miner Res 20(7):1093-1102, 2005);
`the regulation of macrophage differentiation (Romas et al, J
`Exp Med 183(6):2581-2591, 1996); the regulation of proin-
`flammatory cytokine synthesis including TNFot and lL-lB
`(Leng etal,JImmunol159(5):2161-2168, 1997; Hermann et
`al, Arthritis Rheum 41 (8):1388-1397, 1998; Trepicchio et al,
`JImmunol 159(11):5661-5670, 1997); the ability to confer
`mucosal protection after chemotherapy and radiation therapy
`(Orazi et al, Lab Invest 75(1):33-42, 1996); and as an absolute
`requirement for normal development ofplacentation and sur-
`vival to birth (Robb et al, Nat Med 4:303, 1998). A number of
`these biological properties have been exploited in the devel-
`opment of new therapeutic strategies. Recombinant human
`lL-11 has been approved as a treatment for chemotherapy
`induced thrombocytopenia (Tepler et al, Blood 87(9):3607-
`3614, 1996) and is currently being assessed as a new approach
`to the treatment of chemotherapy induced gastrointestinal
`mucositis (Herrlinger et al, Am J Gastroenterol 101(4):793-
`797, 2006). Treatment with recombinant lL-11 in a mouse
`
`Lassen — Exhibit 1005, p. 6
`
`Lassen - Exhibit 1005, p. 6
`
`

`

`US 8,182,814 B2
`
`3
`model of rheumatoid arthritis (collagen induced arthritis,
`CIA) caused a significant reduction in the severity of estab-
`lished disease, which was associated with protection from
`joint damage, as assessed by histology (Walmsley et al,
`Immunology 95(1):31-37, 1998). In a subsequent Phase l/ll
`clinical study patients receiving a once weekly dose of lL-l 1
`(15 ug/kg) demonstrated a significant reduction in the number
`of tender joints, although there was no overall benefit at the
`ACR criterion of a 20% response (Moreland et al, Arthritis
`Res 3(4):247-252, 2001). Similarly, lL-11 has shown thera-
`peutic benefit in animal models of inflammatory bowel dis-
`ease (IBD; Peterson et al, Lab Invest 78(12): 1503-1512,
`1998) and this prompted clinical studies to assess the safety
`and efficacy of lL-11 in patients with active Crohns disease.
`While lL-11 was well tolerated and provided some clinical
`benefit, it remained significantly inferior when compared
`with a standard steroid based therapy (Herrlinger et al, supra
`2006).
`In addition to arthritis and lBD, lL-11 has also been dem-
`onstrated to provide therapeutic benefit in mouse (Lai et al,
`Nephron Exp Nephrol 101 (4):e146-1 54, 2005) and rat (Lai et
`al, JAm Soc Nephrol 12(11):2310-2320, 2001) models of
`glomerulonephritis. In these models, inflammatory disease is
`induced via the administration of ‘nephrotoxic serum’ (gen-
`erated by immunization of donor animals, for example sheep,
`with mouse or rat glomeruli preparations) and is assessed
`through standard histological and urine analysis.
`lL-11
`therapy resulted in a significant reduction in albuminuria at 24
`hrs as well as a decrease in fibrinogen deposition and infil-
`trating inflammatory cells at 14 days post induction ofdisease
`(Lai et al, supra 2005).
`In addition, lL-l 1 has been suggested as a potential thera-
`peutic agent in various other inflammatory disorders includ-
`ing radiation-induced lung damage (Redlich et al, JImmunol
`157(4):1i05 10, 1996), sepsis (Chang et al, Blood Cells Mol
`Dis 22(1):57-67, 1996) and psoriasis (Trepicchio et al, JClin
`Invest 104(11):1527-1537, 1999). U.S. Pat. No. 6,270,759
`suggests that lL-l 1 may be therapeutically useful for a variety
`of inflammatory conditions including asthma and rhinitis.
`The biological properties of lL-11 (IL-11 activity) are
`mediated through a multimeric receptor complex that incor-
`porates lL-11, the lL-llRot chain and gp130 (reviewed in
`Taga, JNeurochem 67(1): 1-10, 1996) and referred to as the
`lL-l 1 receptor complex. The lL-l 1R0. chain binds directly to
`lL-11 with low affinity (kDa ~10 nM), is unique to the lL-11
`receptor complex and is responsible for conferring specific-
`ity. gp130 is a shared receptor component used by members
`ofthe lL-6 ligand family (IL-6, lL-11, LlF, OSM and CNTF)
`and is responsible for the activation of intracellular signal
`transduction, primarily via the JAK/STAT pathway. Recent
`data suggests that the lL-l 1 receptor complex is a high aflinity
`(kDa ~400-800 pM), hexameric complex that incorporates
`two molecules of lL-l 1, two molecules of lL-l 1R0. and two
`molecules of gp130 (Barton et al, J Biol Chem 275(46):
`36197-36203, 2000).
`In contrast to the potential therapeutic approaches using
`lL-11, antagonists oflL-11 or lL-l 1R have been suggested as
`potential
`therapeutics for the treatment of osteoporosis
`(WO9959608) and in view of the role of lL-11 in the devel-
`opment ofplacentation and survival to birth (Robb et al, supra
`1998)
`as
`a
`contraceptive
`agents
`(WO9827996
`and
`W003099322).
`The role of lL-11 as a mediator of airway inflammation
`(including asthma) has primarily been investigated in mouse
`models, where one approach has been to assess the impact of
`increasing local
`lL-11 concentrations, Strategies used to
`achieve such an increase have included the local administra-
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`45
`
`50
`
`55
`
`60
`
`65
`
`4
`
`tion of recombinant lL-11 protein or local de novo synthesis
`via a lung specific lL-11 transgene. The results of these stud-
`ies have not been definitive and, in the context of airways
`disease such as asthma, the potential oflL-l 1 either as a target
`or as a novel therapeutic has remained unclear.
`Einarsson et al, JClin Invest 97(4):915-924, 1996 demon-
`strated that respiratory pathogens linked to asthma exacerba-
`tion (in contrast to other viral and bacterial pathogens) were
`potent stimulators of lung stromal cell lL-11 production in
`vitro. Consistent with this observation, lL-11 was readily
`detectable in aspirates from children with upper respiratory
`tract infections but not in aspirates from uninfected chil-
`dreniinterestingly the highest levels of lL-l 1 were detected
`in aspirates from children with clinical bronchospasm. When
`instilled into the lungs of mice, recombinant lL-11 induced a
`marked increase in sensitivity to methacholine and a mild
`mononuclear inflammatory response. In a subsequent report
`Tang et al, J. Clin. Invest. 98:2845, 1996 generated transgenic
`mice in which constitutive over-expression of lL-11 was tar-
`geted to the lung using the CC-10 promoter (CC-10/lL-11 Tg
`mice). In contrast to wildtype (wt) littermate controls, the
`transgenic animals demonstrated a nodular peribronchiolar
`mononuclear infiltrate with significant airways remodeling
`and sub-epithelial fibrosis. Furthermore, by two months of
`age the transgenic mice demonstrated increased airways
`resistance and airways hyperresponsivness to methacholine
`when compared with their wt littermates.
`While the above studies suggest that lL-11 overexpression
`may contribute to the development of experimental airways
`inflammation, a potential role for lL-11 in the pathology of
`asthma is less clear. For example, cell populations known to
`be central to the development of asthma pathology such as
`eosinophils and mast cells were not detected in the infiltrates
`induced by lL-l 1. Nevertheless lL-l 1 mRNA and protein has
`been detected in the epithelial and sub-epithelial layers of
`human bronchial biopsies, with levels significantly greater in
`moderate and severe asthmatics compared to patients with
`mild disease and non-asthmatics (Minshall et al, J Allergy
`Clin Immunol 105(2 Pt 1):232-238, 2000).
`To address this particular issue more directly Wang et al, J.
`Immunol. 165:2222, 2000 assessed the development of
`experimental asthma (OVA sensitization model)
`in the
`CC-10/lL-11 Tg mice. As expected OVA challenge of sensi-
`tized wt mice caused airway eosinophilic inflammation, Th2
`cell accumulation, and mucus hypersecretion with mucus
`metaplasia. Increased levels of endothelial cell VCAM-1,
`mucin (Muc) 5ac gene expression and bronchoalveolar lav-
`age and lung lL-4, lL-5, and lL-13 protein and mRNA were
`also noted. In contrast, OVA challenged CC10/lL-11 Tg mice
`that overexpressed lL-11 in the lung demonstrated lower
`levels of tissue and bronchoalveolar lavage inflammation,
`eosinophilia, and Th2 cell accumulation, and significantly
`lower levels of VCAM-1 and lL-4, lL-5, and lL-13 mRNA
`and protein. These studies demonstrate that lL-11 selectively
`inhibits many of the hallmarks of asthma pathology and
`prompted the authors to suggest that recombinant lL-11
`might be used as a treatment for Th2 mediated disorders such
`as asthma.
`
`More recent studies in the development of Th2 mediated
`disease have only served to add an additional layer of com-
`plexity (Chen et al, JImmunol 174(4):2305-2313, 2005). The
`Th2 cytokine lL-13 has been demonstrated to be key to the
`development of several aspects of asthma pathology includ-
`ing eosinophillic inflammation, mucus hypersecretion, air-
`ways hyper-responsiveness and allergen specific lgE. In
`agreement with these data lung-specific transgenic overex-
`pression of lL-13 (CC-10/lL-13 Tg mice) results in the devel-
`
`Lassen — Exhibit 1005, p. 7
`
`Lassen - Exhibit 1005, p. 7
`
`

`

`US 8,182,814 B2
`
`5
`opment of a severe Th2/asthma-like phenotype (Zhu et al, J
`Clin Invest 103(6):779-788, 1999). To assess a putative role
`for IL-1 1 in IL-13 activity (Chen et al, supra 2005) compared
`the expression of IL-1 1, IL-11R0t, and gp130 in lungs from
`wild-type mice and CC-10/IL-13 Tg mice and characterized
`the effects of a null mutation of IL-1 1R0. on the development
`of lung pathology in CC-10/IL-13 Tg mice. IL-13 was dem-
`onstrated to be a potent stimulator of IL-11 and IL-11R0t.
`Furthermore many ofthe pathological consequences of IL- 1 3
`overexpression, including inflammation, fibrosis, and mucus
`metaplasia, were substantially ameliorated in the absence of
`IL-1 1Rot. This led to the conclusion that IL-1 1R0. plays a key
`role in the pathogenesis of IL-13-induced inflammation and
`remodeling.
`Accordingly, with respect to airway-inflammation the role
`of IL-1 1 remains unclear. In contrast, for non-airway inflam-
`matory disease, the use of recombinant IL-11 as a novel
`therapeutic agent is well supported by published data.
`There is a need to develop new treatments for Th2-medi-
`ated disorders such as asthma.
`
`SUMMARY
`
`Throughout this specification, unless the context requires
`otherwise, the word “comprise”, or variations such as “com-
`prises” or “comprising”, will be understood to imply the
`inclusion of a stated element or integer or group of elements
`or integers but not the exclusion of any other element or
`integer or group of elements or integers.
`Nucleotide and amino acid sequences are referred to herein
`by a sequence identifier number (SEQ ID NO:). The SEQ ID
`NOs: correspond numerically to the sequence identifiers
`<400>1 (SEQ ID N021), <400>2 (SEQ ID NO:2), etc. A
`summary of the sequence identifiers is provided in Table 1.A
`sequence listing is provided after the claims.
`The present
`invention relates generally to the use of
`antagonists of IL-11 or IL-11Rot in the treatment of Th2-
`mediated disorders. Th2-mediated disorders include inflam-
`
`matory disorders such as asthma, chronic obstructive pulmo-
`nary disease (COPD), rhinitis, allergies and atopic dermatitis.
`Inparticular, the present invention provides the use of antago-
`nists of IL-11 or IL-11Rot in the treatment of asthma.
`
`The present invention is predicated in part on the elucida-
`tion of the role of IL-1 1 in an animal model of Th2-mediated
`
`inflammatory disorders such as asthma, and the effects of an
`antagonist of IL-11 or IL-11Rot in that model. In accordance
`with the present invention, inhibiting the activity of IL-11 is
`proposed to be useful
`in the treatment of Th2-mediated
`inflammatory disorders such as asthma, COPD, rhinitis, aller-
`gies and atopic dermatitis.
`Accordingly, one aspect ofthe present invention provides a
`method for the treatment of a Th2 -mediated disorder in a
`
`subject, the method comprising administering to the subject
`an amount of an antagonist of IL-1 1 or IL-1 1R0t. Reference to
`“an amount” includes an effective amount or an amount suf-
`
`ficient to ameliorate the symptoms of the Th2-mediated
`inflammatory disorder.
`In a particular embodiment, the Th2-mediated disorder is
`asthma.
`
`In another aspect the present invention provides a method
`for the treatment of asthma in a subject, the method compris-
`ing administering to the subject an amount of an antagonist of
`IL-11 or IL-11Rot.
`
`Particular antagonists include an IL-1 1 mutein, an anti-IL-
`1 1 antibody, an anti-IL-l 1R0. antibody and a soluble
`
`10
`
`15
`
`20
`
`25
`
`30
`
`40
`
`45
`
`50
`
`55
`
`60
`
`65
`
`6
`IL-1 1R0. or functional part thereof. A “functional part” is that
`part of the antagonist that retains inhibitory activity towards
`IL-11 or IL-11Rot.
`
`Generally, the agent is administered in an amount and for a
`time and under conditions suflicient to ameliorate the symp-
`toms of the Th2-mediated inflammatory disorder.
`The administration may be systemic or local. Systemic
`administration is particularly useful. Reference to “systemic
`administration” includes intra-articular, intravenous, intrap-
`eritoneal, and subcutaneous injection, infusion, as well as
`administration via oral, rectal and nasal routes, or via inhala-
`tion. Administration by subcutaneous injection or via inhala-
`tion is particularly useful.
`The present invention further contemplates combination
`therapy such as targeting IL-11 and/or IL-11Rot and one or
`more other inflammatory targets.
`invention
`Accordingly, another aspect of the present
`relates to a method for the treatment of a Th2-mediated dis-
`
`order such as but not limited to asthma in a subject, the
`method comprising administering an antagonist of IL-11 or
`IL-11Rot and at least one other therapeutic agent such as an
`anti-inflammatory agent, a bronchodilator or an antibiotic.
`The co-administration may be simultaneous or sequential
`administration.
`
`Particular subjects are mammals such as humans.
`The present invention extends to the use of pharmaceutical
`compositions comprising antagonists of IL-11 or IL-11R0t.
`Useful compositions comprise an IL-11 mutein, an anti-IL-
`1 1 antibody, an anti-IL-l 1R0. antibody, or a soluble IL-1 1Rot.
`The present invention further provides the use ofan antago-
`nist of IL-1 1 or IL-1 1R0. in the manufacture of a medicament
`
`for the treatment of a Th2-mediated disorder in a subject.
`The present invention further provides the use ofan antago-
`nist of IL-1 1 or IL-1 1R0. in the manufacture of a medicament
`
`for the treatment of asthma in a subject.
`A medical kit is also provided comprising an antagonist of
`IL-11 or IL-11Rot
`together with instructions to use the
`antagonists in the treatment of a Th2 -mediated disorder such
`as asthma.
`
`A summary of sequence identifiers used throughout the
`subject specification is provided in Table 1.
`
`TABLE 1
`
`Summafl of sequence identifiers
`
`SEQUENCE
`ID NO:
`
`1
`2
`3
`4
`5
`6
`
`DESCRIPTION
`
`Murine IL- 11 mutein
`Murine IL- 11 mutein
`Murine IL- 11 mutein
`Human IL- 11 mutein
`Muc Sac primer for rtPCR
`Muc Sac primer for rtPCR
`
`BRIEF DESCRIPTION OF THE FIGURES
`
`FIGS. 1, 2 and 3 are graphical representations showing the
`cell population in bronchoalveolar lavage (BAL) samples
`from wild type and IL-1 1R0. null mice challenged with phos-
`phate buffered saline (PBS) control and with OVA at 24, 48
`and 72 hours following exposure.
`FIG. 4 is a graphical representation showing the cell popu-
`lation in BAL samples from wild type mice challenged with
`OVA and treated with either an antagonist of IL-11/IL-1 1R0.
`or with a control.
`
`Lassen — Exhibit 1005, p. 8
`
`Lassen - Exhibit 1005, p. 8
`
`

`

`US 8,182,814 B2
`
`7
`DETAILED DESCRIPTION
`
`The singular forms “a”, “an” and “the” include plural
`aspects unless the context clearly dictates otherwise. Thus,
`for example, reference to “a cytokine” includes a single
`cytokine as well as two or more cytokines; reference to “an
`antibody” includes a single antibody, as well as two or more
`antibodies; reference to “the invention” includes single and
`multiple aspects, of an invention; and so forth.
`The present invention relates to a method for the treatment
`and prophylaxis of inflammatory conditions. It is predicated
`in part on an analysis of the use of a murine OVA-model of
`allergic asthma as a model of Th2-mediated inflammatory
`disorders.
`
`In this model, parameters of Th2 lung inflammation,
`mucus metaplasia and total and antigen-specific serum IgE
`levels enable the determination of the effectiveness of poten-
`tial therapeutic approaches in suppressing some of the key
`features of asthma.
`
`Infiltration of inflammatory cells into the airways, in par-
`ticular eosinophils, is an indicator of airway inflammation
`and a feature of asthmatic airways. The murine OVA-model
`of asthma results in a significant increase in the numbers of
`eosinophils and to a lesser extent macrophages migrating into
`the airways which can be easily seen in cell counts of fluid
`lavaged from the bronchoalveolar.
`In accordance with the present invention, inhibition of
`IL-11 activity including signaling with a test antagonist sig-
`nificantly impacts on the numbers of eosinophils and mac-
`rophages migrating into the airways as determined by cell
`counts of fluid lavaged from the bronchoalveolar of OVA-
`challenged mice, indicating that the antagonism of IL-11
`through inhibition of the formation of the IL-11 receptor
`complex is a useful therapeutic approach. These results were
`supported by experiments comparing IL-11Rot1 null mice
`with wildtype mice in the OVA-model of allergic asthma. The
`IL-11Rot1 null exhibited a reduction in the inflammatory
`response further suggesting that an antagonist of IL-11 or
`IL-11Rot a useful therapeutic approach.
`Accordingly, the present invention provides a method for
`the treatment of a Th2-mediated disorder in a subject, the
`method comprising administering to the subject an amount of
`an antagonist of IL-11 or IL-11R0t.
`Th2-mediated disorders include asthma, COPD, rhinitis,
`allergies and atopic dermatitis. A particular Th2-mediated
`disorder is asthma. Hence, another aspect the present inven-
`tion is directed to a method for the treatment of asthma in a
`subject, the method comprising administering to the subject
`an amount of an antagonist of IL-11 or IL-1 1R0t.
`Particular antagonists of IL-11 or IL-11Rot include an
`IL-1 1 mutein and an antibody specific for IL-1 1 or specific for
`IL-11Rot and a soluble IL-11Rot or a functional part thereof.
`Such a part is functional in the sense that it can still inhibit
`IL-1 1-mediated signaling.
`Reference to “amount” includes an effective amount or an
`
`amount suflicient to ameliorate symptoms of the Th2-medi-
`ated disorder.
`
`55
`
`The term “an antagonist of IL-11 or IL-11Rot” as used
`herein means an agent that binds to IL-11 or IL-11Rot and
`directly inhibits the formation on cells of a multimeric recep-
`tor complex that incorporates IL-11, IL-11Rot and gp130,
`thus inhibiting IL-11 signaling through the IL-11 receptor
`complex. Such antagonists inhibit the action of IL-11 on
`IL-11 sensitive cells. Examples of antagonists of IL-11 or
`IL-11Rot are:
`a. an IL-11 mutein;
`b. an antibody specific for IL-11;
`c. an antibody specific for IL-1 IR; and
`d. a soluble IL-11Rot.
`
`60
`
`65
`
`8
`Antagonists of IL-11 or IL-11Rot may also include agents
`that specifically inhibit expression of IL-11 or IL-11R, for
`example antisense polynucleotides that specifically recogn-
`ise a polynucleotide encoding IL-11 or the IL-11 receptor,
`interfering RNA that disrupt expression of IL-1 1 or the IL-1 1
`receptor or ribozymes that specifically recognise a polynucle-
`otide encoding IL-11 or the IL-11 receptor.
`An antibody specific for IL-11Rot and a soluble IL-11Rot
`may directly bind IL-11 and thereby directly inhibit the for-
`mation on cells of a multimeric receptor complex.
`Antagonists of IL-11 or IL-1 1R0. are known in the art, for
`example US. Pat. No. 6,998,123 describes a soluble
`IL-1 1Rot, IL-1 1-binding portions thereof, and commercially
`available antibodies to IL-11 and demonstrate their antago-
`nist activity. Soluble forms of IL-1 1R0. are also described in
`US. Pat. No. 6,528,281. International Patent Publication No.
`W0 03/099322 describes certain IL-11 muteins and demon-
`
`10
`
`15
`
`strates their antagonist activity.
`The term “IL-1 1” or its full name “interleukin-1 1” as used
`
`20
`
`25
`
`30
`
`35
`
`herein includes all mature forms of wild type mammalian
`IL-11, including murine, macaque and human, IL-1 1, and all
`truncated forms of such IL-11 that retain IL-11 signaling
`activity, i.e. the ability to bind with IL-11Rot and form a
`functional receptor complex with gp130. Mature human
`IL-11 is a 178 amino acid protein (i.e. lacking the 21 amino
`acid leader sequence of NPi000632, NCBI protein database
`Accession Number), and mature murine IL-1 1 is a 178 amino
`acid protein (i.e. lacking the 21 amino acid leader sequence of
`NPi032376, NCBI protein database Accession Number).
`The term “IL-1 1R0.” or its full name “interleukin-1 1 recep-
`tor alpha” as used herein includes, but is not limited to, human
`IL-11Rot having the nucleotide and amino acid sequences
`disclosed in SEQ ID NOs:1 and 2 of International Patent
`Publication NO. WO 96/19574 and murine IL-1 1R0. having
`the nucleotide and amino acid sequences disclosed in SEQ ID
`NOs:2 and 3 of International Patent Publication No. WO
`96/07737. IL-11Rot is also known as IL-11Rot1 and IL-11R
`
`40
`
`and the terms may be used herein interchangeably.
`The term “IL-1 1 mutein” as used herein refers to modified
`
`forms of mature IL-11 in which the amino acid sequence has
`been altered to retain effective binding to IL-1 1R0. but inhibit
`the formation of an IL-1 1 receptor complex with gp130. Such
`muteins compete with IL-11 for IL-11Rot binding and
`antagonize IL-11 signaling thereby inhibiting IL-11 action.
`Alterations to the sequence to form a mutein include amino
`acid substitutions of important residues for receptor binding.
`Conveniently, the mutein is based on human or murine IL-1 1
`and particularly human IL-11. International Publication No.
`WO 03/099322 describes certain IL-11 muteins and demon-
`
`45
`
`50
`
`strates their antagonist activity. Muteins may be expressed in
`suitable host cells and purified using standard techniques.
`IL-111 muteins may be further modified, for example to
`increase their in vivo half life, including for example, by the
`attachment of other elements such as a PEG groups. Methods
`for the PEGylation of peptides are well known in the art.
`The terms “antagonist”, “agent”,
`“compound”, and
`“active” may be used interchangeably herein to refer to a
`substance that induces a desired pharmacological and/or
`physiological effect and may include the IL-11 and IL-1 1R0.
`antagonists herein described. The terms also encompass phar-
`maceutically acceptable and pharmacologically active forms
`thereof, including salts. The desired effect is the inhibition of
`IL-11 activity or IL-11 receptor complex signaling.
`The terms “antibody” and “antibodies” include polyclonal
`and monoclonal antibodies and all the various forms derived
`
`from monoclonal antibodies, including but not limited to
`
`Lassen — Exhibit 1005, p. 9
`
`Lassen - Exhibit 1005, p. 9
`
`

`

`US 8,182,814 B2
`
`9
`full-length antibodies (e.g. having an intact Fc region), anti-
`gen-binding fragments, including for example, Fv, Fab, Fab'
`and F(ab')2 fragments; and antibody-derived polypeptides
`produced using recombinant methods such as single chain
`antibodies. The terms “antibody” and “antibodies” as used
`herein also refer to human antibodies produce

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