throbber
APOTEX ET AL. - EXHIBIT 1068
`Apotex Inc. et al. v. Novartis AG
`IPR2017-00854
`
`

`

`W0 2008/156865
`
`PCT/US2008/007770
`
`JOINT DESTRUCTION BIOMARKERS FOR ANTI-IL-17A THERAPY
`
`OF INFLAMMATORY JOINT DISEASE
`
`The present application claims the benefit of US. Provisional Patent Application
`
`60/945239, filed 20 June 2007.
`
`FIELD OF THE INVENTION
`
`10
`
`The present invention relates generally to the treatment of inflammatory joint diseases
`
`with antagonists of interleukin-17A (IL-17A). More specifically, the invention relates to
`
`biomarkers that are correlated with the efficacy of 1L-17A antagonists for inhibiting joint
`
`destruction in rheumatoid arthritis and associated arthritides.
`
`15
`
`20
`
`25
`
`30
`
`BACKGROUND OF THE INVENTION
`
`Rheumatoid arthritis (RA) is an inflammatory disease caused by the dys-regulation of
`
`the immune system resulting in joint inflammation, causing joint pain, discomfort, swelling
`
`and stiffness, With progressive bone and cartilage erosion. The combination of inflammation
`
`and structural joint damage results in loss of function which can lead to permanent disability.
`
`1L—17A, which was originally named cytotoxic T-Lymphocyte-associated antigen 8
`
`(CTLA8) is a homodimeric cytokine that binds to lL-17RA (also known as ILl7R) and IL-
`
`17RC. The functional receptor for 1L-17A is believed to be a multimeric receptor complex
`
`comprising one or both of 1L—17RA and IL-17RC (e.g., an 1L-17RA homodimer, an 1L-17RC
`
`homodimer, or an 1L-17RA/1L-17RC heterodimer) and possibly a third, as yet unknown,
`
`protein (Toy, D. et al., (2006) J. oflmmunol. 177(1):36-39; unpublished data).
`
`1L-17A is produced by a subset of T cells known as Thl7 cells, whose differentiation
`
`is initiated by TGF-beta signaling in the context of proinflammatory cytokines, particularly IL-
`
`6,
`
`IL-l-beta and TNF-alpha, and whose maintenance and survival are dependent on
`
`interleukin-23 (IL-23) (Langrish, C.L. et al. (2005), J. Exp. Med 201:233-240; Harrington,
`
`L.E., et al., (2005), Nat. Immunol. 6:1123—1132; Veldhoen, M. et al., (2006) Immunity 24:179-
`
`189).
`
`IL-23 is a heterodimeric cytokine comprised of two subunits: p19, which is unique to
`
`IL-23; and p40, which is shared with 1L-12.
`
`IL-23 mediates signaling by binding to a
`
`heterodimeric receptor, comprised of 1L—23R and lL-lZRbetal (1Ll2RB1), which is shared by
`
`the 1L-12 receptor. Studies in murine disease models suggest that IL-23-dependent Th1 7 cells
`
`

`

`W0 2008/156865
`
`PCT/U82008/007770
`
`2
`
`play a pathogenic role in autoimmune and chronic inflammatory diseases (Langrish et a1.,
`
`supra, Park, H., et al. (2005), Nat. Immunol. 6:1133—1141).
`
`IL-17A is present in RA synovial fluid at the earliest stages of the disease along with
`
`other known inflammatory mediators such as TNF and IL-lB. Dys-regulated IL-17A
`
`expression within an inflamed joint singly, and in synergy with TNF and IL-IB, stimulates
`
`multiple downstream proteases, chemokines and pro-inflammatory cytokines that collectively
`
`contribute to cartilage and bone erosion. A variety of IL-17A biological antagonists used in
`
`multiple rodent arthritis models have demonstrated that E—17A blockade inhibits arthritis
`
`progression and the resulting joint destruction that occurs with a special emphasis on bone
`
`sparing (see, e.g., Koenders MI, et al., (2006) Ann. Rheum. Dis. 65 (Suppl. 3):29-33). At least
`
`one anti-IL-17A antibody is being tested in clinical trials of human RA patients.
`
`Currently, assessing the effect of anti-rheumatic drugs on the progression of joint
`
`destruction relies mainly on radiographic evaluation. However, how to use radiographic data
`
`in clinical trials is controversial (van der Heijde, D. et al, (2002) Arthritis Rheum 47;215-218).
`
`In addition to being time consuming, radiography is impractical in early stages of RA in which
`
`symptoms reflecting the inflammatory process often predominate over symptoms related to
`
`joint destruction (Morozzi, G., et al, (2007) Clin Rheumatol.)
`
`'Indeed, nonsteroidal anti-
`
`inflammatory drugs (NSAIDs), which have traditionally been used as first line therapies for
`
`RA, are reasonably effective at ameliorating the signs and symptoms of inflammation, but
`
`have little efficacy in retarding joint destruction, leading to speculation that inflammation and
`
`subsequent joint destruction can be uncoupled (van den Berg, WB (2001), Semin Arthritis
`
`Rheum. 30:7-16; Geusens, RB, et al. (2006), Arthritis & Rheumatism 54 (6):1772-1777).
`
`Thus, there is a well-established clinical need for better tools to predict the effect of anti-
`
`rheumatic drugs on structural joint damage, with recent development efforts focused on
`
`various markers of cartilage and/or bone metabolism that are elevated in the serum or urine of
`
`RA patients compared to normal subjects (kaic, M. et a1., (2003) Arthritis Res. Ther.
`
`5:R181-R185; Valleala, H., et a1. (2003) Eur. J. Endocrinol. 148:527-530); Ziolkowska, M.,
`
`et a1. (2002) Arthritis & Rheumatism 46(7):1744-1753).
`
`Articular cartilage in the joints is composed of the proteoglycan aggrecan, collagen
`
`(three a-chains form a triple helix), and other non—collagenous proteins (e.g., cartilage
`
`oligomer matrix protein (COMP) and human cartilage glycoprotein-39 (HC gp-39), which is
`
`also known as YKL-40). Type I collagen is a major component of bone and other tissues;
`
`whereas, type II collagen is specifically localized to articular cartilage of the joint. At the ends
`
`10
`
`15
`
`20
`
`25
`
`30
`
`

`

`W0 2008/156865
`
`’
`
`3
`
`PCT/US2008/007770
`
`of type I and H collagen helixes are short, non-helical N- and C-terminal
`
`telopeptides
`
`containing covalent cross-links that connect to other a-chains, both within the same trimer
`
`and to adjacent trimers. Physiological and pathological cleavage of collagen by MMPs or
`Cathepsin K results in the generation of degradation products or neo-epitopes (e.g. C2C,
`C1,2C, C-terminal cross-linking telopeptide of type I collagen (CTX-I), C-terminal cross-
`
`linking telopeptide of type [I collagen (CTX-II), N-terminal cross-linking telopeptide of type I
`
`collagen (NTX-I)), which are released into the synovial fluid, serum, and urine. Cleavage of
`
`the collagen triple helix also releases non-collagenous proteins (e.g. COMP, YKL-40,
`
`aggrecan) previously incorporated in the collagen fibrils. These molecules are elevated in
`
`synovial fluid and serum under conditions of normal remodeling and pathological cartilage
`
`destruction.
`
`Cartilage destruction also results in compensatory increased collagen synthesis by
`
`chondrocytes. Type I‘and [I collagen is synthesized as a pro-molecule and once outside the
`
`cell, cleavage of pro-collagen releases N-terminal and C-terminal pro-peptides. Type [I
`
`collagen C—terrninal pro-peptide (CPII) levels correlate with new type [I collagen synthesis.
`
`Cartilage destruction also increases aggrecan synthesis and the appearance of the “fetal form”
`
`of aggrecan that has the CS846 epitope.
`
`Increased CS846 levels in the serum reflect new
`
`aggrecan synthesis (versus cleavage of “old” aggrecan).
`
`Bone destruction occurs via the generation of excessive numbers of osteoclasts that
`
`resorb the mineralized bone and degrade the organic matrix of the de-mineralized bone.
`
`Receptor activator of NFKB (RANK) ligand (RANKL) is a cell-surface molecule expressed by
`
`activated T—cells,
`
`fibroblast-like synoviocytes (FLS), and osteoblasts that
`
`is critical
`
`in
`
`promoting the differentiation of pre-osteoclasts into mature osteoclasts, which are cells that
`
`can erode bone. RANKL can be shed by proteolytic cleavage (both cell surface and soluble
`
`RANKL are active), and is elevated in mouse arthritis and human RA serum. Membrane or
`
`soluble RANKL binds to RANK on pre-osteoclasts and delivers a differentiation signal.
`
`Osteoprotegn'n (OPG) is a natural antagonist of this system by binding to RANKL and
`
`preventing its interaction with RANK on pre-osteoclasts.
`
`Tartrate-resistant acid phosphatase (TRACP) isoform 5b is released into the serum by
`
`bone-resorbing osteoclasts as they transcytose degraded bone proteins fiom the resorbed bone
`
`surface to outside the bone. TRACP isoform 5b serum levels are elevated in bone resorption
`
`diseases.
`
`The amino acid sequence for TRACPS is
`
`found in Accession No.
`
`for
`
`NM 001102405, NM_001102404 or NM_007388.
`
`1O
`
`15
`
`20
`
`25
`
`30
`
`

`

`W0 2008/156865
`
`PCT/U82008/007770
`
`4
`
`‘Some of these serum markers of bone and cartilage metabolism (or destruction) are
`
`elevated in RA patients and have some prognostic value in identifying patients that are at a
`
`higher risk of having more aggressive bone destruction. For example, elevated levels of
`cartilage oligomeric matrix protein (COMP) have been associated with more aggressive
`
`radiographic progression (den Broeder, A. A., et a1. (2002) Ann Rheum Dis 61(4): 311-318;
`
`Wollheim, F. A., et a1. (1997) Br J Rheumatol 36(8): 847-8499; Skoumal, M., et al. (2003).
`
`ScandJRheumatoI 32(3):156-l6l; Mansson, B., et a1. (1995), J Clin Invest 95(3):1071-1077;
`
`Lindqvist, E., et a1. (2005) Ann Rheum Dis 64(2):]96-201; Forslind, K., et a1. (1992) Br J
`
`Rheumatol 31(9): 593-598; Fex, E., et a1. (1997) Br JRheumatoI 36(11):]161-1165. Also, a
`
`low OPG/RANKL ratio predicted increased five year radiographic progression (Geusens, P. P.
`
`et a1. (2006) Arthritis Rheum 54(6):]772—1777) and elevated CTX-I and CTX-II levels were
`
`associated with four year Sharp Score increase in early RA patients (Gamero, P., et al. (2002)
`
`Arthritis Rheum 46(11):2847-2856).
`
`However, the inventors herein are not aware of any published studies that conclude
`
`that blocking IL-17A can inhibit bone erosion and modulate serum levels of any of the above
`
`proteins in severely arthritic animals. Thus, a need exists to identify biomarkers that correlate
`
`with inhibition ofjoint destruction by anti-IL-17A therapy.
`
`SUMMARY OF THE INVENTION
`
`The present invention is based on the discovery described herein that COMP, OPG and
`
`RANKL serum levels in mice with collagen-induced arthritis (CIA) following treatment with
`
`an anti-IL-17A monoclonal antibody (Mab) are modulated by anti-IL-17A therapy. Also, the
`
`inventors have discovered that RANKL serum levels in CIA-mice decrease with increasing
`
`doses of the anti-IL-17A Mab, and reach normal levels at antibody doses that are effective at
`
`inhibiting joint destruction in the CIA mice as measured by traditional histological and u—CT-
`
`based techniques. Based on these results with COMP, OPG and RANKL in the mouse CIA
`
`arthritis model, the inventors herein believe that these markers are likely to be useful as
`
`surrogate markers, i.e., biomarkers, of the effect of anti—IL-17A therapy on joint destruction in
`
`inflammatory joint diseases such as RA and associated athritidies. Also, these data obtained
`
`in arthritic mice support the use of other markers of cartilage and bone metabolism that are
`
`elevated in human RA patients,
`
`including CTX—I, CTX-II, and HC gp-39, as surrogate
`
`markers for monitoring the effect of anti-IL-17A therapy on joint destruction in patients with
`
`inflammatory joint disease.
`
`10
`
`15
`
`20
`
`25
`
`30
`
`

`

`W0 2008/156865
`
`PCT/U82008/007770
`
`5
`
`In addition, it has been previously discovered that anti-IL-17A therapy is effective at
`
`inhibiting joint destruction in the CIA arthritis model, even when that therapy produces no
`
`apparent improvement in inflammation (WO 2008/021156). Thus, it is expected that anti-IL-
`
`17A therapy will be usefill to inhibit ongoing bone erosion in human patients who have been
`
`previously treated with a different anti-rheumatic agent, regardless of whether or not the
`
`previous agent had reduced the signs and symptoms of inflammation. Thus, the present
`
`discovery of non-inflammation related markers that are correlated with inhibition of bone
`
`erosion by IL-17A therapy provides novel methods and products for treating patients for bone
`
`erosion who are inflammatory nonresponders or inflammatory responders to previous anti-
`
`10
`
`rheumatic therapy.
`
`Accordingly, one aspect of the present invention is a method of selecting a patient with
`
`an inflammatory joint disease for treatment with an IL—17A antagonist. The patient selection
`
`method comprises comparing the level(s) of at least one joint destruction biomarker in a
`
`serum sample taken from the subject with the normal range of serum levels for the biomarker
`
`15
`
`and selecting the patient for treatment with the IL-17A antagonist if the level(s) of the joint
`
`destruction biomarker in the subject’s serum sample is outside of the normal range.
`
`In another aspect, the invention provides a method of predicting efficacy of an IL-17A
`
`antagonist in inhibiting bone erosion in a subject with an inflammatory joint disease. This
`
`efficacy prediction method comprises determining the levels of at least one joint destruction
`
`20
`
`biomarker in serum samples taken from the subject prior to and at the end of an initial
`
`treatment period with the IL-17A antagonist, and comparing the levels of the biomarker in
`
`these pre-treatment and post-treatment serum samples. A normalization in the level of the
`
`biomarker during the initial treatment period predicts that the IL—17A antagonist will likely be
`
`effective in inhibiting joint destruction in the subject.
`
`In a preferred embodiment,
`
`the
`
`prediction method firrther comprises determining the level of the biomarker in a third serum
`
`sample taken from the subject at the end of a subsequent treatment period with the IL-17A
`antagonist; if the level of the biomarker in the third serum sample is more normalized than the
`
`level of the biomarker in the second serum sample, then the IL-17A antagonist is predicted to
`
`be effective in inhibiting joint destruction in the subject. Preferred initial treatment periods
`
`are at least one week, at least two weeks, at least four weeks, at least eight weeks, at least
`
`twelve weeks, at least 24 weeks, or at least 48 weeks, while preferred subsequent treatment
`
`periods are at least 12 weeks, at least 24 weeks or at least 48 weeks.
`
`In some embodiments,
`
`the subject
`
`is a non—human animal with arthritis, which may be naturally present or
`
`25
`
`30
`
`

`

`W0 2008/156865
`
`PCT/U82008/007770
`
`6
`
`experimentally-induced.
`
`Preferred non-human subjects include CIA mice or rats with
`
`adjuvant-induced arthritis (AIA). In other embodiments, the subject is a human with arthritis.
`
`In yet a timber aspect, the present invention provides a method of treating a subject for
`
`an inflammatory joint disease with an IL-l7A antagonist. This treatment method comprises
`
`determining, in a first serum sample taken from the subject, the level of at least one joint
`
`destruction biomarker, administering the IL-l7A antagonist to the subject according to a first
`
`dosing regimen during an initial
`
`treatment period, determining the level of the selected
`
`biomarker(s) in at least a second serum sample taken from the patient at the end of the initial
`
`treatment period, and comparing the levels of the biomarker in the first and second serum
`
`10
`
`samples.
`
`If the level of the biomarker in the second serum sample is within a specified range,
`
`then the subject is treated with the IL-17A antagonist according to the first dosing regimen
`
`during at least one subsequent treatment period. However, if the level of the biomarker in the
`
`second serum sample is outside of the specified range, e.g., indicating that more aggressive
`
`therapy may be necessary to achieve inhibition of joint destruction, then the subject is treated
`
`with the IL-17A antagonist according to a second dosing regimen during at
`
`least one
`
`subsequent treatment period, wherein the second dosing regimen comprises administering a
`
`total amount of the IL-17A antagonist during the subsequent treatment period that is greater
`
`than the total amount administered during the initial treatment period.
`
`In one preferred embodiment of this treatment method, the specified range is the
`
`normal range, i.e., the range of serum levels of the biomarker found in a population of healthy,
`
`, gender- and age-matched subjects.
`
`In another preferred embodiment, the specified range for
`
`the serum level of the biomarker is defined by a confidence interval of at least 80% of the
`
`mean serum level of the biomarker in a population of subjects with the inflammatory disease
`
`who were treated with the same IL-17A antagonist according to the same dosing regimen for a
`
`time period equal
`
`to or longer than the initial
`
`treatment period, wherein the population
`
`exhibited inhibition of joint destruction following treatment with the IL-17A antagonist
`
`according to the first dosing regimen during a time period equal to or longer than the
`
`subsequent treatment period.
`
`In one preferred embodiment, when the biomarker level in the second serum sample
`
`indicates that more aggressive anti—IL-17A therapy is required, the subject is treated with a
`
`greater
`
`total amount of the antagonist during the subsequent
`
`treatment period(s) by
`
`15
`
`20
`
`25
`
`30
`
`

`

`WO 2008/156865
`
`PCT/US2008/007770
`
`7
`
`administering the IL-17A antagonist at higher doses and/or at more frequent intervals than the
`
`doses and intervals employed during the initial treatment period.
`
`In
`
`another preferred embodiment,
`
`the
`
`treatment method flirther
`
`comprises
`
`administering an IL-23 antagonist during each of the initial
`
`treatment and subsequent
`
`treatment periods, or during only a subsequent treatment period. The IL-23 antagonist may
`
`inhibit the expression of either subunit of the cytokine (IL-23p19 or p40), either subunit of the
`
`functional receptor (IL-23R or IL-12beta1), or may inhibit IL-23 signaling by directly or
`
`indirectly interacting with one or more of these polypeptides to prevent a functional ligand-
`
`receptor interaction.
`
`In some preferred embodiments, the IL-23 antagonist is an antibody or
`
`antibody fragment that binds to and inhibits the activity of either IL-23p19 or IL—23R. In one
`
`particularly preferred embodiment,
`
`the IL-23 antagonist
`
`is a monoclonal antibody that
`
`specifically binds to IL—23p19.
`
`The invention also provides a kit for treating an inflammatory joint disease. The kit
`
`comprises a pharmaceutical composition of an IL-17A antagonist and reagents for measuring
`
`the level of at least one joint destruction biomarker in a serum sample taken from a subject.
`
`The invention also provides a manufactured drug product for treating an inflammatory
`
`joint disease, which comprises a pharmaceutical formulation comprising an IL-17A antagonist
`
`and instructions for determining patient serum levels of at
`
`least one joint destruction
`
`biomarker before and during treatment with the IL-17A antagonist.
`
`Yet another aspect of the invention is the use of an IL-17A antagonist for preparing a
`
`medicament for treating a patient having an inflammatory joint disease to inhibit joint
`
`destruction, wherein the patient has an abnormal serum level of at least one joint destruction
`
`biomarker after previous treatment with a different anti-rheumatic therapy.
`
`In a preferred
`
`embodiment, the medicament is for administering the IL-17A antagonist according to any of
`
`10
`
`15
`
`20
`
`25
`
`the treatment regimens described herein.
`
`In each of the above described aspects of the invention, the IL-17A antagonist may
`
`inhibit the expression of IL-17A or IL-17RA or IL-17RC or may inhibit IL-17A signaling by
`
`directly or indirectly interacting with one or more of these polypeptides to prevent a functional
`
`ligand-receptor interaction.
`
`In some preferred embodiments, the IL-17A antagonist is an
`
`30
`
`antibody or antibody fragment that binds to and inhibits the activity of either IL-17A, IL-
`
`17RA or IL-17RC.
`
`In one particularly preferred embodiment, the IL-17A antagonist is a
`
`monoclonal antibody that specifically binds to IL-17A.
`
`In other preferred embodiments, the
`
`IL-17A antagonist is a bispecific antibody that binds to and inhibits the activity of IL-23p19
`
`

`

`W0 2008/156865
`
`PCT/U82008/007770
`
`8
`
`and H.-17A; H.—23p19 and H.—17RA; IL-23R and IL-17A; IL-23R and IL-17RA, IL—23p19 and
`
`IL-17RC; or H.-23R and IL-17RC.
`
`In another particularly preferred embodiment, the H.-17A
`
`antagonist is a bispecific antibody that binds to and inhibits the activity of IL-23p19 and IL-
`
`17A.
`
`In each of the above—described aspects of the invention, preferred joint destruction
`
`biomarkers are COMP, CTX-I, CTX-II, HC gp-39, OPG, RANKL, TRACP) isoform 5b and
`
`osteocalcin. Any one of these biomarkers or any combination of two or more, or all six, of
`
`these biomarkers may be employed. COMP, OPG and RANKL are more preferred
`
`biomarkers, with RANKL being the most preferred biomarker.
`
`Preferred inflammatory joint diseases that may be treated using any of the above
`
`aspects of the invention are rheumatoid arthritis (RA), psoriatic arthritis (PsA) or ankylosing
`
`spondylitis (AS), with rheumatoid arthritis being a particularly preferred inflammatory joint
`
`disease.
`
`Also,
`
`in each of the above-described aspects of the invention, the subject with-the
`
`inflammatory joint disease may be one that is an inflammatory nonresponder, an inflammatory
`
`responder, a moderate inflammatory responder, or a good inflammatory responder to the IL-
`
`17A antagonist or a different anti-rheumatic drug.
`
`BRIEF DESCRIPTION OF THE DRAWINGS
`
`Figure 1A shows the amino acid sequence of the light chain of humanized anti-IL—17A
`
`antibody 16C10 according to the present invention SEQ ID NO: 1. CDRs are indicated.
`
`Figure 1B shows amino acid sequence for the heavy chain of humanized anti-IL-17A
`
`antibody 16C10 according to the present invention SEQ ID NO: 2. CDRs are indicated.
`
`1O
`
`15
`
`20
`
`Figures 2A-2D shows the effects of anti-IL-17A antibody treatments on pathology in
`
`25
`
`the CIA mouse model of rheumatoid arthritis. Treatments include administration of anti-IL-
`
`17A antibody JL7.1D10 (at 28, 7, and 2 mg/kg) and administration of an isotype control (7
`
`mg/kg).
`
`Figure 2A presents visual disease severity score (DSS), a measure of visual paw
`
`swelling and redness, as a function of antibody treatment. Scoring is: O = paw appears the
`
`30
`
`same as control (untreated) paw;
`
`1 = inflammation of one finger on a given paw; 2 =
`
`inflammation of two fingers or the palm of a given paw; 3 = inflammation of the palm and
`
`finger(s) of a given paw.
`
`Figure 2B presents cartilage damage (by histopathology) as a function of antibody
`
`treatment. Scoring is: 0 = normal; 1 = minimal, 2 = mild; 3 = moderate; 4 = severe.
`
`

`

`W0 2008/156865
`
`PCT/US2008/007770
`
`9
`
`Figure 2C presents bone erosion (by histopathology) as a function of antibody
`
`treatment. Scoring is: 0 = normal; 1 = minimal, 2 = mild; 3 = moderate; 4 = severe.
`
`Figure 2D presents bone erosion (by histopathology) for paws from CIA mice that
`
`scored 2 or 3 in visual DSS, i.e. highly inflamed paws. rIgGl is an isotype control antibody.
`
`Scoring is: 0 = normal; 1 = minimal, 2 = mild; 3 = moderate; 4 = severe.
`
`Figures 3A—3B presents data showing the effect of an anti-IL-17A antibody on serum
`
`COMP levels in arthritic mice.
`
`Figure 3A presents serum COMP levels in mice treated with isotype control or an anti-
`
`IL-17A antibody (JL7.1D10). The solid horizontal bar denotes the average serum COMP
`
`level
`
`in non-diseased animals (grey circles on left side of graphs) and the two dotted
`
`horizontal bars denotes +/— 2 standard deviations from the non-diseased mice average.
`
`Figure 3AA presents serum COMP levels in non—diseased (normal) mice or in CIA
`
`mice treated weekly for five weeks with an isotype control or an anti—IL-17A antibody
`
`(JL7.1D10) at a dose of 28 mg/kg or 7 mg/kg.
`
`Figure 3B presents serum COMP levels in non-diseased (un-manipulated) and severely
`
`arthritic mice treated with short term isotype control (rIgGl) or an anti-IL-17A antibody
`
`(JL7.1D10).
`
`Figure 4 presents serum RANKL levels in arthritic mice who were untreated, treated
`
`with an isotype control, or treated with one of three doses of an anti-IL-17A antibody
`
`(JL7.1D10). The solid horizontal bar denotes the average serum COMP level in non-diseased
`
`animals (grey circles on left side of graphs) and the two dotted horizontal bars denotes +/— 2
`
`standard deviations from the non-diseased mice average.
`
`Figure 5 presents serum OPG levels in normal mice (grey circles) or in arthritic mice
`
`who were untreated (no dosing), treated with an isotype control (Rat IgGl), or treated with an
`
`10
`
`15
`
`20
`
`25
`
`anti-IL-17A antibody.
`
`Figure 6 presents serum RANKL and OPG levels in normal mice (grey circles) or in
`
`severely arthritic mice following short-term exposure to an isotype control (rIgGl) or an anti-
`
`IL-17A antibody (JL7.1D10).
`
`Figure 7 presents micro-CT X-rays of an un-inflamed mice paw, and of severely
`
`30
`
`inflamed paws from arthritic mice treated with an isotype control antibody or an anti-IL-17A
`
`antibody.
`
`

`

`W0 2008/156865
`
`PCT/US2008/007770
`
`l 0
`
`Figure 8 presents~ serum TRACP levels in non-diseased (un—manipulated) and severely
`
`arthritic mice treated with long term isotype control (rIgGl) or an anti-IL-17A antibody
`
`(JL7.1D10).
`
`Figure 9 presents serum osteocalcin levels in normal mice and in CIA mice treated
`
`with an anti-E-17A antibody (JL7.1D10) or an isotype control antibody
`
`Figure 10 shows the dynamic change in serum RANKL (left panel) and OPG (right
`
`panel) levels in a cohort of mice progressing through the mouse CIA mode, with horizontal
`
`lines indicating mean (unbroken line) +/— SD. (broken lines) in un-manipulated healthy mice
`
`and antibody dosing denoted as arrowheads.
`
`10
`
`Figure 11 shows the serum RANKL concentration versus total animal DSS from
`
`individual normal or CIA mice at each of five weeks of treatment with a rat IgGl isotype
`
`control antibody.
`
`Figure 12 shows the serum OPG concentration versus total animal DSS from
`
`individual normal or CIA mice at each of five weeks of treatment with a rat IgGl isotype
`
`15
`
`control antibody.
`
`Figure 13 presents the serum RANKL levels in individual CIA mice that were not
`
`treated or treated with five weekly subcutaneous doses of an isotype control antibody (rat
`
`IgGl) or an anti-E-17A antibody (JL7.1D10) at 2, 7, or 28 mg/kg.
`
`Figure 14 shows the serum OPG profiles for individual CIA mice that were not treated
`
`or treated with five weekly subcutaneous doses of 7 mg/kg of an isotype control antibody (rat
`
`IgGl) or 28 mg/kg of an anti-E-17A antibody (JL7.1D10).
`
`Figure 15 shows the thickness of paws in rats with adjuvant-induced arthritis (AIA)
`
`that were treated prior to adjuvant injection with an isotype antibody or with the indicated
`
`doses of an anti-rat IL-17A antibody (JL8.18E10).
`
`Figure 16 illustrates the effect of anti-E-l7A or anti-TNF therapy on serum RANKL
`
`levels at days 14 and 25 following drug exposure in individual rats with established AIA.
`
`1.
`
`Definitions.
`
`DETAILED DESCRIPTION
`
`So that the invention may be more readily understood, certain technical and scientific
`
`terms are specifically defined below. Unless specifically defined elsewhere in this document,
`
`all other technical and scientific terms used herein have the meaning commonly understood by
`
`one of ordinary skill in the art to which this invention belongs.
`
`20
`
`25
`
`30
`
`

`

`W0 2008/156865
`
`PCT/US2008/007770
`
`l 1
`
`As used herein, including the appended claims, the singular forms of words such as
`
`“a,” “an,” and “the,” include their corresponding plural references unless the context clearly
`
`dictates otherwise.
`
`“Abnormal” in the context of the serum level of a joint destruction biomarker means
`
`that the serum level is outside of the normal range for that biomarker.
`
`“Ankylosing spondylitis” or “AS” is a form of chronic inflammation of the spine and
`
`the sacroiliac joints, which are located in the low back where the sacrum (the bone directly
`
`above the tailbone) meets the iliac bones (bones on either side of the upper buttocks). Chronic
`
`inflammation in these areas causes pain and stiffness in and around the spine. Over time,
`
`10
`
`chronic spinal inflammation (spondylitis) can lead to a complete cementing together (fusion)
`
`of the vertebrae, a process referred to as ankylosis. Ankylosis leads to loss of mobility of the
`
`spine. Ankylosing spondylitis is also a systemic rheumatic disease, meaning it can affect
`
`other tissues throughout the body. Accordingly, it can cause inflammation in or injury to
`
`other joints away from the spine, as well as other organs, such as the eyes, heart, lungs, and
`
`15
`
`kidneys.
`
`“Antagonist” means any molecule that can prevent, neutralize, inhibit or reduce a
`
`targeted activity, i.e., the activity of a cytokine such as IL-17A, either in vitro or in vivo.
`
`Cytokine antagonists include, but are not limited to, antagonistic antibodies, peptides, peptide-
`
`mimetics, polypeptides, and small molecules that bind to a cytokine (or any of its subunits) or
`
`its functional receptor (or any of its subunits) in a manner that interferes with cytokine signal
`
`transduction and downstream activity. Examples of peptide and polypeptide antagonists
`
`include truncated versions or fragments of the cytokine receptor (e.g., soluble extracellular
`
`domains) that bind to the cytokine in a manner that either reduces the amount of cytokine
`
`available to bind to its functional receptor or otherwise prevents the cytokine from binding to
`
`its functional receptor. Antagonists also include molecules that prevent expression of any
`
`subunit
`
`that comprises the cytokine or
`
`its receptor, such as,
`
`for example, antisense
`
`oligonucleotides which target mRNA, andinterfering messenger RNA, (see, e.g., Arenz and
`
`Schepers (2003) Naturwz'ssenschaften 90:345-359; Sazani and K016 (2003) J. Clin. Invest.
`
`112:481-486; Pirollo, et al. (2003) Pharmacol. Therapeutics 99:55-77; Wang, et al. (2003)
`
`Antisense Nucl. Acid Drug Devel. 13:169-189). The inhibitory effect of an antagonist can be
`
`measured by routine techniques. For example, to assess the inhibitory effect on cytokine-
`
`induced activity, human cells expressing a functional receptor for a cytokine are treated with
`
`the cytokine and the expression of genes known to be activated or inhibited by that cytokine is
`
`20
`
`25
`
`30
`
`

`

`W0 2008/156865
`
`PCT/US2008/007770
`
`1 2
`
`measured in the presence or absence of a potential antagonist. Antagonists useful in the
`
`present invention inhibit the targeted activity by at least 25%, preferably by at least 50%, more
`
`preferably by at least 75%, and most preferably by at least 90%, when compared to a suitable
`
`control.
`
`"Antibody" refers to any form of antibody that exhibits the desired biological activity,
`
`such as inhibiting binding of a ligand to its receptor, or by inhibiting ligand—induced signaling
`
`of a receptor. Thus, “antibody” is used in the broadest sense and specifically covers, but is not
`
`limited to, monoclonal antibodies (including full length monoclonal antibodies), polyclonal
`
`antibodies, and multispecific antibodies (e. g., bispecific antibodies).
`
`“Antibody fragment” and “antibody binding fragment” mean antigen-binding
`
`fragments and analogues of an antibody, typically including at least a portion of the antigen
`
`binding or variable regions (e.g. one or more CDRs) of the parental antibody. An antibody
`
`fragment retains at least some of the binding specificity of the parental antibody. Typically,
`
`an antibody fragment retains at least 10% of the parental binding activity when that activity is
`
`expressed on a molar basis. Preferably, an antibody fragment retains at least 20%, 50%, 70%,
`
`80%, 90%, 95% or 100% or more of the parental antibody’s binding affinity for the target.
`
`Examples of antibody fragments include, but are not limited to, Fab, Fab', F(ab')2, and Fv
`
`fragments; diabodies;
`
`linear antibodies; single-chain antibody molecules, e.g., sc-Fv; and
`
`multispecific antibodies formed from antibody fragments. Engineered antibody variants are
`
`reviewed in Holliger and Hudson (2005) Nat. Biotechnol. 23: 1 126-1 136.
`
`A "Fab fragment" is comprised of one light chain and the CH1 and variable regions of
`
`one heavy chain. The heavy chain of a Fab molecule cannot form a disulfide bond with
`
`another heavy chain molecule.
`
`An "Fc" region contains two heavy chain fragments comprising the CH1 and CH2
`
`domains of an antibody. The two heavy chain fragments are held together by two or more
`
`disulfide bonds and by hydrophobic interactions of the CH3 domains.
`
`A "Fab’ fragment" contains one light chain and a portion of one heavy chain that
`
`contains the VH domain and the C H1 domain and also the region between the CH1 and C H2
`
`domains, such that an interchain disulfide bond can be formed between the two heavy chains
`
`10
`
`15
`
`20
`
`25
`
`30
`
`of two Fab' fragments to form a F(ab') 2 molecule.
`
`A "F(ab'); fragment" contains two light chains and two heavy chains containing a
`
`portion of the constant region between the CH1 and CH2 domains, such that an interchain
`
`

`

`W0 2008/156865
`
`PCT/US2008/007770
`

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