US20080199460A1 - Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease - Google Patents

Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease Download PDF

Info

Publication number
US20080199460A1
US20080199460A1 US11/512,622 US51262206A US2008199460A1 US 20080199460 A1 US20080199460 A1 US 20080199460A1 US 51262206 A US51262206 A US 51262206A US 2008199460 A1 US2008199460 A1 US 2008199460A1
Authority
US
United States
Prior art keywords
antagonist
monoclonal antibody
treatment period
antibody fragment
activity
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/512,622
Other languages
English (en)
Inventor
Daniel J. Cua
Robert A. Kastelein
Van T. Tsai
Rachel Caspi
Phyllis Silver
Dror Luger
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
NATIONAL INSTITUTES OF HEALTH - OFFICE OF TECHNOLOGY TRANSFER
US Department of Health and Human Services
Merck Sharp and Dohme Corp
Original Assignee
US Department of Health and Human Services
Schering Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=37686002&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20080199460(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by US Department of Health and Human Services, Schering Corp filed Critical US Department of Health and Human Services
Priority to US11/512,622 priority Critical patent/US20080199460A1/en
Assigned to SCHERING CORPORATION reassignment SCHERING CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CUA, DANIEL J., KASTELEIN, ROBERT A., TSAI, VAN T.
Assigned to NATIONAL INSTITUTES OF HEALTH - OFFICE OF TECHNOLOGY TRANSFER reassignment NATIONAL INSTITUTES OF HEALTH - OFFICE OF TECHNOLOGY TRANSFER ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CASPI, RACHEL, LUGER, DROR, SILVER, PHYLLIS
Publication of US20080199460A1 publication Critical patent/US20080199460A1/en
Priority to US12/643,152 priority patent/US8524230B2/en
Priority to US12/643,166 priority patent/US20100111954A1/en
Priority to US13/015,900 priority patent/US20110142831A1/en
Priority to US13/960,441 priority patent/US20130323251A1/en
Priority to US14/274,905 priority patent/US20140248279A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention relates generally to the modulation of immune responses in the eye. More specifically, the invention relates to the use of antagonists of interleukin-23 (IL-23) and interleukin-17 (IL-17) to treat autoimmune ocular inflammatory disease.
  • IL-23 interleukin-23
  • IL-17 interleukin-17
  • Ocular inflammatory disease is a general term embracing a number of diseases and conditions in which inflammation affects the eye or surrounding tissues.
  • the diagnostic name given to an OID is typically based on the location of the ocular inflammation.
  • uveitis inflammation in the uveal tract
  • scleritis inflammation of the sclera
  • pars planitis inflammation of the pars plana, and so forth.
  • OIDs cause pain, irritation, and watering, and may result in loss of visual function.
  • uveitis is the third leading cause of blindness in the developed world.
  • OIDs can be caused by infections, malignancy, exposure to toxins, response to surgery or injury, and autoimmune disorders.
  • AOID autoimmune-mediated OID
  • S—Ag retinal arrestin
  • IRB interphotoreceptor retinoid binding protein
  • GP100 GP100, MART1, TRP1 and TRP2
  • OID is a manifestation of a systemic autoimmune disease
  • the eye is one of a variety of organs throughout the body that are being attacked.
  • systemic autoimmune diseases include rheumatoid arthritis, systemic lupus erythematosus, polyarteritis nodosa, relapsing polychondritis, Wegener's granulomatosis, scleroderma, Behcet's disease, Reiter's disease, inflammatory bowel disease (ulcerative colitis and Crohn's disease) and ankylosing spondylitis.
  • the eye may be the specific and only target affected in autoimmune diseases such as ocular cicatricial pemphigoid, Mooren's corneal ulcer, and various forms of uveitis.
  • AOIDs such as uveitis have been treated by various classes of compounds including steroids and nonsteroidal anti-inflammatory agents such as dexamethasone, flurometholone, prednisolone, indomethacin, aspirin, flubiprofen and diclofenac.
  • nonsteroidal anti-inflammatory agents such as dexamethasone, flurometholone, prednisolone, indomethacin, aspirin, flubiprofen and diclofenac.
  • these drugs are associated with serious side effects such as cataracts, glaucoma, delayed wound healing, altered prostaglandin production, corneal complications, increased ocular pressure, superinfections, and reduced immunity to infection (see, e.g., Id., at 181; Guidera, A. C., et al. (2001) Ophthalmology 108:936-944; Olsen, E. G. & Davanger M. (1984) Acta Ophtalmol. 62:893-899).
  • EAU experimental autoimmune uveitis
  • a retinal antigen shown to be reactive in uveitis patients e.g., arrestin, IRBP, rhodopsin/opsin, phosducin, recoverin
  • Studies using the EAU model provided apparently contradictory evidence about the mechanisms for induction and progression of this disease.
  • IFN- ⁇ deficient knock-out mice were susceptible for EAU, that EAU is exacerbated by neutralization of endogenous IFN- ⁇ , and that elevated levels of IFN- ⁇ were protective against EAU in wild-type mice (Caspi, R. R. et al. (1994) J. Immunol. 152:890-899; Jones et al., J. Immunol. 158:5997-6005; Tarrant, T. K., et al. (1999) J. Exp. Med. 189:219-230.
  • the present invention is based on the discoveries that (1) blocking interleukin-23 (IL-23) or interleukin-17 (IL-17) activity prevents induction of EAU; (2) after induction, neutralization of IL-17 activity inhibits or reverses progression of EAU, but neutralization of IL-23 activity has little to no effect; and (3) IL-17 activity is not necessary for induction of EAU.
  • the present invention uses IL-23 and/or IL-17 antagonists in methods and compositions for treating or preventing autoimmune inflammatory disease. These antagonists antagonize either the target cytokine itself or a functional receptor for the target cytokine.
  • IL-23 is a heterodimeric cytokine comprised of two subunits: p19, which is unique to IL-23; and p40, which is shared with IL-12.
  • IL-23 mediates signaling by binding to a heterodimeric receptor, comprised of IL-23R and IL-12Rbeta1 (IL12RB1), which is shared by the IL-12 receptor.
  • IL-12R IL-12Rbeta1
  • a recent paper reported that IL-23 promotes a T cell population characterized by the production of IL-17, IL-17F, TNF, IL-6 and other factors, and named these cells “Th 17 cells” (Langrish et al. (2005) J. Exp. Med. 201:233-240)).
  • IL-17 which was originally named cytotoxic T-Lymphocyte-associated antigen 8 (CTLA8) is a homodimeric cytokine that binds to IL-17RA (also known as IL17R) and IL-17C.
  • CTLA8 cytotoxic T-Lymphocyte-associated antigen 8
  • the functional receptor for IL-17 is believed to be a multimeric receptor complex comprising one or both of IL-17RA and IL-17RC (e.g., an IL-17RA homodimer, an IL-17RC homodimer, or an IL-17RA/IL-17RC heterodimer) and possibly a third, as yet unknown, protein (Toy, D. et al., (2006) J. of Immunol. 177(1):36-39; unpublished data).
  • the invention provides a method of treating a patient with an autoimmune ocular inflammatory disease, comprising administering to the patient an IL-17 antagonist.
  • an AOID need not be directly diagnosed, but may be inferred by a diagnosis that the patient has an ocular inflammation that is of putative autoimmune etiology and/or that exhibits one or more characteristics of an autoimmune response.
  • a particularly preferred AOID is autoimmune uveitis, e.g., uveitis without an infectious etiology.
  • the IL-17 antagonist may inhibit the expression of IL-17 or IL-17R or IL-17RC or may inhibit IL-17 signaling by directly or indirectly interacting with one or more of these polypeptides to prevent a functional ligand-receptor interaction.
  • the IL-17 antagonist is an antibody or antibody fragment that binds to and inhibits the activity of either IL-17, IL17R or IL17C.
  • the IL-17 antagonist is a monoclonal antibody that specifically binds to IL-17.
  • the IL-17 antagonist is a bispecific antibody that binds to and inhibits the activity of IL-23p19 and IL-17; IL-23p19 and IL-17RA; IL-23R and IL-17; or IL-23R and IL-17RA.
  • the IL-17 antagonist is a bispecific antibody that binds to and inhibits the activity of IL-23p19 and IL-17.
  • the IL-17 antagonist is administered according to a specified treatment regimen.
  • a specified dose of the antagonist is administered at a specified interval during a first treatment period, which may end after disappearance of one or more symptoms of the AOID, or within a specified period of time.
  • the treatment regimen further comprises gradually reducing the dose of the IL-17 antagonist during a second treatment period that begins upon the end of the first treatment period and ends when therapy with the IL-17 antagonist is stopped.
  • the duration of the second treatment period is typically between one and twelve months, one and nine months, one and six months, or one and three months.
  • the specified treatment regimen also comprises administration of an IL-23 antagonist to the patient during each of the first and second treatment periods, or during only the second 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.
  • the IL-23 antagonist is an antibody or antibody fragment that binds to and inhibits the activity of either IL-23p19 or IL-23R.
  • the IL-23 antagonist is a monoclonal antibody that specifically binds to IL-23p19.
  • the IL-23 antagonist may be administered at a specified dose at a specified interval during one or both of the first and second treatment periods.
  • the dose of the IL-23 antagonist administered in the second treatment period may be lower than the dose administered in the first period.
  • the doses of the IL-17 and IL-23 antagonists may be the same or different from each other.
  • the two antagonists may be administered at the same or different intervals during each treatment period.
  • the dose of the IL-17 antagonist may be reduced while the dose of the IL-23 antagonist is held constant, or the dose of each antagonist may be gradually reduced.
  • the dose of the IL-23 antagonist is held constant during the second treatment regimen and therapy with the IL-23 antagonist is continued during a third treatment period that begins upon the end of the second treatment period (i.e., when therapy with the IL-17 antagonist is stopped).
  • the IL-23 antagonist may be administered at the same dose and interval as in the second treatment period or may be administered at a lower dose and/or less frequent interval than used in the previous period.
  • the dose of the IL-23 antagonist may also be gradually reduced during the third treatment period.
  • the duration of the third treatment period is typically between one and twelve months, one and nine months, one and six months, or one and three months.
  • the specified treatment regimen also comprises administering a therapeutic agent that does not antagonize IL-17 or IL-23 activity but is capable of alleviating at least one symptom of the AOID or at least one side effect of the IL-17 or IL-23 antagonists during any or all of the treatment periods.
  • the therapeutic agent is a steroid or a nonsteroidal anti-inflammatory agent (e.g., NSAID) that is known to have efficacy in treating uveitis.
  • the therapeutic agent targets a cytokine that promotes the Th1 response.
  • Another aspect of the invention provides a method of prophylactically treating a patient who is diagnosed as being susceptible for an autoimmune ocular inflammatory disease, which comprises administering to the patient an antagonist of one or both of IL-23 and IL-17.
  • the susceptibility diagnosis is based on the patient having a previous incidence of ocular inflammation.
  • the susceptibility diagnosis is based on the patient having a systemic autoimmune disease.
  • the antagonist may be administered in a specified dose at a specified interval during a first treatment period, which typically ends after three months, six months, nine months or after two years of therapy with the antagonist.
  • the dose of the antagonist is gradually reduced during a second treatment period that begins upon the end of the first treatment period, and typically has a duration of between one and three months.
  • the invention provides a method of treating a patient for an autoimmune ocular inflammatory disease, comprising administering to the patient an IL-23 antagonist.
  • the IL-23 antagonist may be administered at a specified interval during a first treatment period, which is followed by a second treatment period in which the IL-23 antagonist is administered at a lower dose or at less frequent intervals, or at gradually reduced doses.
  • Therapy with the Il-23 antagonist will typically continue for at least three to six months and may continue for as many as 12 months, 18 months or 24 months.
  • Another aspect of the invention is the use of an IL-17 antagonist or an IL-23 antagonist for the preparation of a pharmaceutical composition for the treatment or prevention of an autoimmune ocular inflammatory disease (AOID) in a patient.
  • the pharmaceutical composition is for administering the antagonist according to any of the treatment regimens described herein.
  • the invention provides a manufactured drug product for treating an autoimmune ocular inflammatory disease.
  • the drug product comprises (i) a first pharmaceutical formulation comprising an IL-17 antagonist; and (ii) a second pharmaceutical formulation comprising an IL-23 antagonist.
  • the drug product includes product information which comprises instructions for administering the pharmaceutical formulations according to any of the treatment regimens described herein.
  • “Antagonist” means any molecule that can prevent, neutralize, inhibit or reduce a targeted activity, i.e., the activity of a cytokine such as IL-17 or IL-23, 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.
  • 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, and interfering messenger RNA, (see, e.g., Arenz and Schepers (2003) Naturwissenschaften 90:345-359; Sazani and Kole (2003) J. Clin. Invest.
  • 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 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.
  • 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.
  • an antibody fragment retains at least 10% of the parental binding activity when that activity is expressed on a molar basis.
  • 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.
  • 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. 23:1126-1136.
  • a “Fab fragment” is comprised of one light chain and the C H 1 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 C H 1 and C H 2 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 V H domain and the C H 1 domain and also the region between the C H 1 and C H 2 domains, such that an interchain disulfide bond can be formed between the two heavy chains of two Fab′ fragments to form a F(ab′) 2 molecule.
  • a “F(ab′) 2 fragment” contains two light chains and two heavy chains containing a portion of the constant region between the C H 1 and C H 2 domains, such that an interchain disulfide bond is formed between the two heavy chains.
  • a F(ab′) 2 fragment thus is composed of two Fab′ fragments that are held together by a disulfide bond between the two heavy chains.
  • the “Fv region” comprises the variable regions from both the heavy and light chains, but lacks the constant regions.
  • a “single-chain Fv antibody refers to antibody fragments comprising the V H and V L domains of an antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the V H and V L domains which enables the scFv to form the desired structure for antigen binding.
  • scFv see Pluckthun (1994) THE PHARMACOLOGY OF MONOCLONAL ANTIBODIES, vol. 113, Rosenburg and Moore eds. Springer-Verlag, New York, pp. 269-315. See also, International Patent Application Publication No. WO 88/01649 and U.S. Pat. Nos. 4,946,778 and 5,260,203.
  • a “diabody” is a small antibody fragment with two antigen-binding sites.
  • the fragments comprises a heavy chain variable domain (V H ) connected to a light chain variable domain (V L ) in the same polypeptide chain (V H -V L or V L -V H ).
  • V H heavy chain variable domain
  • V L light chain variable domain
  • the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites.
  • Diabodies are described more fully in, e.g., EP 404,097; WO 93/11161; and Holliger et al. (1993) Proc. Natl. Acad. Sci. USA 90: 6444-6448.
  • a “domain antibody fragment” is an immunologically functional immunoglobulin fragment containing only the variable region of a heavy chain or the variable region of a light chain.
  • two or more V H regions are covalently joined with a peptide linker to create a bivalent domain antibody fragment.
  • the two V H regions of a bivalent domain antibody fragment may target the same or different antigens.
  • AOID Autoimmune-mediated ocular inflammatory disease
  • AOID means any disease or condition in which (a) inflammation is present in any part of the eye or surrounding tissues (including the optic nerve, blood vessels, muscles) and (b) the inflammation is part of an immune response that requires or is promoted by one or both of IL-23 and IL-17. Intraocular inflammation without an infectious etiology is typically considered an AOID.
  • Nonlimiting examples of AOIDs are listed below.
  • BSRC Birdshot retinochoriodopathy
  • OCP Ocular cicatricial pemphigoid
  • Keratitis is inflammation of the cornea, the outer, transparent, dome-like structure that forms the anterior most part of the outer coat of the eye. If ulcers develop in the peripheral cornea, it is referred to as peripheral ulcerative Keratitis.
  • “Sympathetic ophtahlmia” is an AOID in which a trauma to one eye precipitates at a later time a destructive inflammation in the other (“sympathizing”) eye, apparently due to an autoimmune response to antigens released from the injured eye.
  • Vogt-Koyanagi Harada VKH
  • Vogt-Koyanagi-Harada syndrome VKH
  • uveomenigitic syndrome Severe bilateral panuveitis associated with subretinal fluid accumulation is the hallmark of ocular VKH.
  • Fuchs' heterochromic iridocyclitis A chronic, unilateral anterior uveitis characterized by iris heterochromia, a condition in which one eye is a different color from the other. The uveitis typically occurs in the lighter colored eye of a young adult.
  • Binding compound refers to a molecule, small molecule, macromolecule, antibody, a fragment or analogue thereof, or soluble receptor, capable of binding to a specified target. “Binding compound” also may refer to any of the following that are capable of binding to the specified target: a complex of molecules (e.g., a non-covalent molecular complex); an ionized molecule; and a covalently or non-covalently modified molecule (e.g., modified by phosphorylation, acylation, cross-linking, cyclization, or limited cleavage). In cases where the binding compound can be dissolved or suspended in solution, “binding” may be defined as an association of the binding compound with a target where the association results in reduction in the normal Brownian motion of the binding compound.
  • Binding composition refers to a binding compound in combination with at least one other substance, such as a stabilizer, excipient, salt, buffer, solvent, or additive.
  • Bispecific antibody means an antibody that has two antigen binding sites having specificities for two different epitopes, which may be on the same antigen, or on two different antigens.
  • Bispecific antibodies include bispecific antibody fragments. See, e.g., Hollinger, et al. (1993) Proc. Natl. Acad. Sci. U.S.A. 90: 6444-48, Gruber, et al., J. Immunol. 152: 5368 (1994).
  • cytokine which consists essentially of a recited amino acid sequence may also include one or more amino acids that do not materially affect the properties of the cytokine.
  • Interleukin-12R beta1 or “IL12RB1” means a single polypeptide chain consisting essentially of the sequence of the mature form of human IL12RB1 as described in NCBI Protein Sequence Database Accession Numbers NP714912, NP005526 or naturally occurring variants thereof.
  • Interleukin-17 means a protein consisting of one or two polypeptide chains, with each chain consisting essentially of the sequence of the mature form of human IL17A as described in any of NCBI Protein Sequence Database Accession Numbers NP002181, AAH67505, AAH67503, AAH67504, AAH66251, AAH66252 or naturally occurring variants thereof.
  • IL-17R or “IL-17RA” means a single polypeptide chain consisting essentially of the sequence of the mature form of human IL-17RA as described in WO 96/29408 or in any of NCBI Protein Sequence Database Accession Numbers: NP 055154, Q96F46, CAJ86450, or naturally occurring variants of these sequences.
  • IL-17RC means a single polypeptide chain consisting essentially of the sequence of the mature form of human IL-17RC as described in WO 238764A2 or in any of NCBI Protein Sequence Database Accession Numbers NP703191, NP703190 and NP 116121, or naturally occurring variants of these sequences.
  • Interleukin-23 means a protein consisting of two polypeptide chains. One chain consists essentially of the sequence of the mature form of human IL23, subunit p19 (also known as IL23A) as described in any of NCBI Protein Sequence Database Accession Numbers NP057668, AAH67511, AAH66267, AAH66268, AAH66269, AAH667512, AAH67513 or naturally occurring variants of these sequences.
  • the other chain consists essentially of the sequence of the mature form of human IL12, subunit p40 (also known as IL12B and IL23, subunit p40) as described in any of NCBI Protein Sequence Database Accession Numbers NP002178, P29460, AAG32620, AAH74723, AAH67502, AAH67499, AAH67498, AAH67501 or naturally occurring variants of these sequences.
  • Interleukin-23R or “IL-23R” means a single polypeptide chain consisting essentially of the sequence of the mature form of human IL23R as described in NCBI Protein Sequence Database Accession Number NP653302 or naturally occurring variants thereof.
  • “Monoclonal antibody” or “mAb” means an antibody obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • Parental administration means an intravenous, subcutaneous, or intramuscular injection.
  • Small molecule means a molecule with a molecular weight that is less than 10 kD, typically less than 2 kD, and preferably less than 1 kD.
  • Small molecules include, but are not limited to, inorganic molecules, organic molecules, organic molecules containing an inorganic component, molecules comprising a radioactive atom, synthetic molecules, peptide mimetics, and antibody mimetics.
  • Peptide mimetics of antibodies and cytokines are known in the art. See, e.g., Casset, et al. (2003) Biochem. Biophys. Res. Commun. 307:198-205; Muyldermans (2001) J. Biotechnol. 74:277-302; Li (2000) Nat.
  • one member of a binding pair has a significantly greater affinity for the other member of the binding pair than for irrelevant proteins.
  • an antibody is considered to be specific for a particular protein if it binds to that protein with an affinity that is at least 10-fold, and preferably 50-fold higher than its affinity for a different protein.
  • an antibody that “specifically binds” to a protein comprising a particular epitope does not bind to any measurable degree to proteins that do not comprise that epitope.
  • an antibody that is specific for a target protein will have an affinity toward the target protein that is greater than about 10 9 liters/mol, as determined, e.g., by Scatchard analysis (Munsen, et al. (1980) Analyt. Biochem. 107:220-239).
  • Treat” or “Treating” means to administer a therapeutic agent, such as a composition containing any of the IL-17 and IL-23 antagonists described herein, internally or externally to a patient in need of the therapeutic agent.
  • a therapeutic agent such as a composition containing any of the IL-17 and IL-23 antagonists described herein.
  • the agent is administered in an amount effective to prevent or alleviate one or more disease symptoms, or one or more adverse effects of treatment with a different therapeutic agent, whether by preventing the development of, inducing the regression of, or inhibiting the progression of such symptom(s) or adverse effect(s) by any clinically measurable degree.
  • the amount of a therapeutic agent that is effective to alleviate any particular disease symptom or adverse effect may vary according to factors such as the disease state, age, and weight of the patient, and the ability of the therapeutic agent to elicit a desired response in the patient. Whether a disease symptom or adverse effect has been alleviated can be assessed by any clinical measurement typically used by physicians or other skilled healthcare providers to assess the severity or progression status of that symptom or adverse effect.
  • a therapeutically effective amount will typically result in a reduction of the measured symptom by at least 5%, usually by at least 10%, more usually at least 20%, most usually at least 30%, preferably at least 40%, more preferably at least 50%, most preferably at least 60%, ideally at least 70%, more ideally at least 80%, and most ideally at least 90%.
  • an embodiment of the present invention may not be effective in preventing or alleviating the target disease symptom(s) or adverse effect(s) in every patient, it should alleviate such symptom(s) or effect(s) in a statistically significant number of patients as determined by any statistical test known in the art such as the Student's t-test, the chi 2 -test, the U-test according to Mann and Whitney, the Kruskal-Wallis test (H-test), Jonckheere-Terpstra-test and the Wilcoxon-test.
  • any statistical test known in the art such as the Student's t-test, the chi 2 -test, the U-test according to Mann and Whitney, the Kruskal-Wallis test (H-test), Jonckheere-Terpstra-test and the Wilcoxon-test.
  • Uveitis means inflammation affecting one or more of the three parts of the eye that make up the uvea: the iris (the colored part of the eye), the ciliary body (behind the iris, responsible for manufacturing the fluid inside the eye) and the choroid (the vascular lining tissue underneath the retina).
  • Panuveitis denotes the presence of inflammation in multiple parts of the same eye (anterior, intermediate, and posterior sections).
  • Uveitis can be either acute or chronic.
  • the chronic form is more often associated with systemic disorders including ankylosing spondylitis, Behcet's syndrome, inflammatory bowel disease, juvenile rheumatoid arthritis, Reiter's syndrome, sarcoidosis, syphilis, tuberculosis, and Lyme disease.
  • Anterior uveitis which involves inflammation in the front part of the eye, is the most common form of uveitis.
  • the inflammation is usually isolated to the iris; thus, anterior uveitis is often called ulceris.
  • anterior uveitis may be associated with the presence of an autoimmune disease such as rheumatoid arthritis or ankylosing spondylitis, but most cases of anterior uveitis occur in otherwise healthy people and do not indicate an underlying systemic disease.
  • This OID may affect only one eye and is most common in young and middle-aged people.
  • a history of an autoimmune disease is a risk factor. Most attacks of anterior uveitis last from a few days to weeks with treatment, but relapses are common.
  • Intermediate uveitis denotes an idiopathic inflammatory syndrome mainly involving the anterior vitreous, peripheral retina, and ciliary body, with minimal or no anterior segment or chorioretinal inflammatory signs.
  • Pars planitis is inflammation of the pars plana, a narrow area between the iris and the choroid. Pars planitis usually occurs in young men and is generally not associated with any other disease. However, there have been a few case reports of an association with Crohn's disease and some experts suggest a possible association with multiple sclerosis. For this reason, these experts recommend that patients over 25 years old diagnosed with pars planitis receive an MRI of their brain and spine.
  • Posterior uveitis affects the back portion of the uveal tract and involves primarily the choroid. This is called choroiditis.
  • Posterior uveitis is characterized by inflammation of the layer of blood vessels underlying the retina, and usually of the retina as well. If the adjacent retina is also involved, the condition is typically called chorioretinitis.
  • Posterior uveitis may follow a systemic infection or occur in association with an autoimmune disease. In posterior uveitis, the inflammation may last from months to years and may cause permanent vision damage, even with treatment.
  • the present invention provides methods of using antagonists of IL-17 and IL-23 activity to treat autoimmune ocular inflammatory disease.
  • IL17 activity which is reviewed in Kolls, J. et al. (2004) Immunity Vol. 21, 467-476, includes promoting accumulation of neutrophils in a localized area and the activation of neutrophils.
  • IL17 can induce or promote the production of any of the following proinflammatory and neutrophil-mobilizing cytokines, depending on the cell type: IL-6, MCP-1, CXCL8 (IL-8), CXCL1, CXCL6, TNF ⁇ , IL-1 ⁇ , G-CSF, GM-CSF, MMP-1, and MMP-13.
  • IL-23 activity includes inducing the proliferation of memory T cells, PHA blasts, CD45RO T cells, CD45RO T cells; and enhance production of interferon-gamma (IFN ⁇ ) by PHA blasts or CD45RO T cells.
  • IFN ⁇ interferon-gamma
  • IL-23 preferentially stimulates memory as opposed to na ⁇ ve T cell populations in both human and mouse.
  • IL-23 activates a number of intracellular cell-signaling molecules, e.g., Jak2, Tyk2, Stat1, Stat2, Stat3, and Stat4.
  • IL-12 activates this same group of molecules, but Stat4 response to IL-23 is relatively weak, while Stat4 response to IL-12 is strong (Oppmann, et al., supra; Parham, et al.
  • IL-23 has also been implicated in the maintenance and proliferation of IL-17 producing cells, also known as Th 17 cells (see, Cua and Kastelein (2006) Nature Immunology 7:557-559).
  • Antagonists useful in the present invention include a soluble receptor comprising the extracellular domain of a functional receptor for IL-17 or IL-23.
  • Soluble receptors can be prepared and used according to standard methods (see, e.g., Jones, et al. (2002) Biochem. Biophys. Acta 1592:251-263; PrudAppel, et al. (2001) Expert Opinion Biol. Ther. 1:359-373; Fernandez-Botran (1999) Crit. Rev. Clin. Lab Sci. 36:165-224).
  • Preferred IL-17 antagonists for use in the present invention are antibodies that specifically bind to, and inhibit the activity of, any of IL-17, IL-17RA, IL-17RC, and a heteromeric complex comprising IL-17RA and IL-17RC. More preferably, the target of the IL-17 antagonist is IL-17 or IL-17RA. Particularly preferred IL-17 antagonists specifically bind to, and inhibit the activity of IL-17.
  • IL-17 antagonist for use in the present invention is a bispecific antibody, or bispecific antibody fragment, which also antagonizes IL-23 activity.
  • bispecific antagonists specifically bind to, and inhibits the activity of, each member in any of the following combinations: IL-17 and IL-23; IL-17 and IL-23p19; IL-17 and IL-12p40; IL-17 and an IL-23R/IL12RB1 complex; IL-17 and IL-23R; IL-17 and IL12RB1; IL17RA and IL-23; IL-17RA and IL-23p19; IL-17RA and IL-12p40; IL-17RA and an IL-23R/IL12RB1 complex; IL-17RA and IL-23R; IL-17RA and IL12RB1; IL17RC and IL-23; IL-17RC and IL-23p19; IL-17RC and IL-12p40; IL-17RC and an IL-23R/IL12RB1 complex;
  • bispecific antibodies used in the present invention are: IL-17 and IL-23; IL-17 and IL-23p19; IL17RA and IL-23; and IL-17RA and IL-23p19.
  • a particularly preferred bispecific antibody specifically binds to, and inhibits the activity of, each of IL-17 and IL-23p19.
  • Preferred IL-23 antagonists are antibodies that bind to, and inhibit the activity of, any of IL-23, IL-23p19, IL-12p40, IL23R, IL12RB1, and an IL-23R/IL12RB1 complex.
  • Another preferred IL-23 antagonist is an IL-23 binding polypeptide which consists essentially of the extracellular domain of IL-23R, e.g., amino acids 1-353 of GenBankAAM44229, or a fragment thereof.
  • Antibody antagonists for use in the invention may be prepared by any method known in the art for preparing antibodies.
  • the preparation of monoclonal, polyclonal, and humanized antibodies is described in Sheperd and Dean (eds.) (2000) Monoclonal Antibodies , Oxford Univ. Press, New York, N.Y.; Kontermann and Dubel (eds.) (2001) Antibody Engineering , Springer-Verlag, New York; Harlow and Lane (1988) Antibodies A Laboratory Manual , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., pp. 139-243; Carpenter, et al. (2000) J. Immunol. 165:6205; He, et al. (1998) J. Immunol.
  • the eliciting antigen may be a peptide containing a single epitope or multiple epitopes, or it may be the entire protein alone or in combination with one or more immunogenicity enhancing agents known in the art.
  • the peptide may be conjugated to a carrier protein.
  • the antigen may also be an isolated full-length protein, a cell surface protein (e.g., immunizing with cells transfected with at least a portion of the antigen), or a soluble protein (e.g., immunizing with only the extracellular domain portion of the protein).
  • the antigen may be expressed by a genetically modified cell, in which the DNA encoding the antigen is genomic or non-genomic (e.g., on a plasmid).
  • a peptide consisting essentially of a region of predicted high antigenicity can be used for antibody generation.
  • regions of high antigenicity of human p19 occur at amino acids 16-28; 57-87; 110-114; 136-154; and 182-186 of GenBank AAQ89442 (gi:37183284) and regions of high antigenicity of human IL-23R occur at amino acids 22-33; 57-63; 68-74; 101-112; 117-133; 164-177; 244-264; 294-302; 315-326; 347-354; 444-473; 510-530; and 554-558 of GenBank AAM44229 (gi: 21239252), as determined by analysis with a Parker plot using Vector NTI® Suite (Informax, Inc, Bethesda, Md.).
  • Any suitable method of immunization can be used. Such methods can include use of adjuvants, other immunostimulants, repeated booster immunizations, and the use of one or more immunization routes. Immunization can also be performed by DNA vector immunization, see, e.g., Wang, et al. (1997) Virology 228:278-284. Alternatively, animals can be immunized with cells bearing the antigen of interest, which may provide superior antibody generation than immunization with purified antigen (Kaithamana, et al. (1999) J. Immunol. 163:5157-5164).
  • Preferred antibody antagonists are monoclonal antibodies, which may be obtained by a variety of techniques familiar to skilled artisans. Methods for generating monoclonal antibodies are generally described in Stites, et al. (eds.) BASIC AND CLINICAL IMMUNOLOGY (4th ed.) Lange Medical Publications, Los Altos, Calif., and references cited therein; Harlow and Lane (1988) ANTIBODIES: A LABORATORY MANUAL CSH Press; Goding (1986) MONOCLONAL ANTIBODIES: PRINCIPLES AND PRACTICE (2d ed.) Academic Press, New York, N.Y.
  • splenocytes isolated from an immunized mammalian host are immortalized, commonly by fusion with a myeloma cell to produce a hybridoma.
  • splenocytes isolated from an immunized mammalian host are immortalized, commonly by fusion with a myeloma cell to produce a hybridoma.
  • Alternative methods of immortalization include transformation with Epstein Barr Virus, oncogenes, or retroviruses, or other methods known in the art. See, e.g., Doyle, et al. (eds.
  • CELL AND TISSUE CULTURE LABORATORY PROCEDURES, John Wiley and Sons, New York, N.Y.
  • Colonies arising from single immortalized cells are screened for production of antibodies of the desired specificity, affinity and inhibiting activity using suitable binding and biological assays.
  • antibody to target binding properties can be measured, e.g., by surface plasmon resonance (Karlsson, et al. (1991) J. Immunol. Methods 145:229-240; Neri, et al. (1997) Nat. Biotechnol. 15:1271-1275; Jonsson, et al. (1991) Biotechniques 11:620-627) or by competition ELISA (Friguet, et al. (1985) J. Immunol. Methods 77:305-319; Hubble (1997) Immunol. Today 18:305-306).
  • Other suitable techniques involve screening phage antibody display libraries. See, e.g., Huse et al., Science 246:1275-1281 (1989); and Ward et al., Nature 341:544-546 (1989); Clackson et al. (1991) Nature 352: 624-628 and Marks et al. (1991) J. Mol. Biol. 222: 581-597; Presta (2005) J. Allergy Clin. Immunol. 116:731.
  • Preferred monoclonal antibodies for use in the present invention are “chimeric” antibodies (immunoglobulins) in which the variable domain is from the parental antibody generated in an experimental mammalian animal, such as a rat or mouse, and the constant domains are obtained from a human antibody, so that the resulting chimeric antibody will be less likely to elicit an adverse immune response in a human subject than the parental mammalian antibody.
  • a monoclonal antibody used in the present invention is a “humanized antibody”, in which all or substantially all of the hypervariable loops (e.g., the complementarity determining regions or CDRs) in the variable domains correspond to those of a non-human immunoglobulin, and all or substantially all of the framework (FR) regions in the variable domains are those of a human immunoglobulin sequence.
  • a particularly preferred monoclonal antibody for use in the present invention is a “fully human antibody”, e.g., an antibody that comprises human immunoglobulin protein sequences only.
  • a fully human antibody may contain carbohydrate chains from the cell species in which it is produced, e.g., if produced in a mouse, in a mouse cell, or in a hybridoma derived from a mouse cell, a fully human antibody will typically contain murine carbohydrate chains.
  • Monoclonal antibodies used in the present invention may also include camelized single domain antibodies. See, e.g., Muyldermans et al. (2001) Trends Biochem. Sci. 26:230; Reichmann et al. (1999) J. Immunol. Methods 231:25; WO 94/04678; WO 94/25591; U.S. Pat. No. 6,005,079.
  • the antagonistic antibodies used in the present invention may have modified (or blocked) Fc regions to provide altered effector functions. See, e.g., U.S. Pat. No. 5,624,821; WO2003/086310; WO2005/120571; WO2006/0057702. Alterations of the Fc region include amino acid changes (substitutions, deletions and insertions), glycosylation or deglycosylation, and adding multiple Fc. Changes to the Fc can alter the half-life of therapeutic antibodies, enabling less frequent dosing and thus increased convenience and decreased use of material. See Presta (2005) J. Allergy Clin. Immunol. 116:731 at 734-35.
  • the antibodies may also be conjugated (e.g., covalently linked) to molecules that improve stability of the antibody during storage or increase the half-life of the antibody in vivo.
  • molecules that increase the half-life are albumin (e.g., human serum albumin) and polyethylene glycol (PEG).
  • Albumin-linked and PEGylated derivatives of antibodies can be prepared using techniques well known in the art. See, e.g., Chapman, A. P. (2002) Adv. Drug Deliv. Rev. 54:531-545; Anderson and Tomasi (1988) J. Immunol. Methods 109:37-42; Suzuki, et al. (1984) Biochem. Biophys. Acta 788:248-255; and Brekke and Sandlie (2003) Nature Rev. 2:52-62).
  • Bispecific antibodies that antagonize both IL-17 and IL-23 activity can be produced by any technique known in the art.
  • bispecific antibodies can be produced recombinantly using the co-expression of two immunoglobulin heavy chain/light chain pairs. See, e.g., Milstein et al. (1983) Nature 305: 537-39.
  • bispecific antibodies can be prepared using chemical linkage. See, e.g., Brennan, et al. (1985) Science 229: 81.
  • bifunctional antibodies can also be prepared by disulfide exchange, production of hybrid-hybridomas (quadromas), by transcription and translation to produce a single polypeptide chain embodying a bispecific antibody, or transcription and translation to produce more than one polypeptide chain that can associate covalently to produce a bispecific antibody.
  • the contemplated bispecific antibody can also be made entirely by chemical synthesis.
  • the bispecific antibody may comprise two different variable regions, two different constant regions, a variable region and a constant region, or other variations.
  • Antibodies used in the present invention will usually bind with at least a K D of about 10 ⁇ 3 M, more usually at least 10 ⁇ 6 M, typically at least 10 ⁇ 7 M, more typically at least 10 ⁇ 8 M, preferably at least about 10 ⁇ 9 M, and more preferably at least 10 ⁇ 10 M, and most preferably at least 10 ⁇ 11 M (see, e.g., Presta, et al. (2001) Thromb. Haemost. 85:379-389; Yang, et al. (2001) Crit. Rev. Oncol. Hematol. 38:17-23; Carnahan, et al. (2003) Clin. Cancer Res. (Suppl.) 9:3982s-3990s).
  • IL-17 antagonists and IL-23 antagonists are typically administered to a patient as a pharmaceutical composition in which the antagonist is admixed with a pharmaceutically acceptable carrier or excipient, see, e.g., Remington's Pharmaceutical Sciences and U.S. Pharmacopeia: National Formulary , Mack Publishing Company, Easton, Pa. (1984).
  • the pharmaceutical composition may be formulated in any manner suitable for the intended route of administration. Examples of pharmaceutical formulations include lyophilized powders, slurries, aqueous solutions, suspensions and sustained release formulations (see, e.g., Hardman, et al.
  • the route of administration will depend on the properties of the antagonist or other therapeutic agent used in the pharmaceutical composition.
  • a possible administration route is to administer the pharmaceutical composition topically to the eye in the form of an ointment, gel or droppable liquids using an ocular delivery system known to the art such as an applicator or eyedropper.
  • the pharmaceutical composition may be administered intraocularly via an polymer implant that is placed under the under the conjunctiva of the eye or through injection directly into the eye.
  • compositions containing IL-17 antagonists and IL-23 antagonists are administered systemically by oral ingestion, injection or infusion by intravenous, intraperitoneal, intracerebral, intramuscular, intraocular, intraarterial, intracerebrospinal, intralesional, or pulmonary routes, or by sustained release systems such as implants.
  • Injection of gene transfer vectors into the central nervous system has also been described (see, e.g., Cua, et al. (2001) J. Immunol. 166:602-608; Sidman et al. (1983) Biopolymers 22:547-556; Langer, et al. (1981) J. Biomed. Mater. Res.
  • compositions used in the invention may be administered according to any treatment regimen that ameliorates or prevents one or more symptoms of the AOID. Selecting the treatment regimen will depend on several composition-dependent and patient-dependent factors, including but not limited to the half-life of the antagonist, the severity of the patient's symptoms, and the type or length of any adverse effects. Preferably, an administration regimen maximizes the amount of therapeutic agent delivered to the patient consistent with an acceptable level of side effects. Guidance in selecting appropriate doses of therapeutic antibodies and small molecules is available (see, e.g., Wawrzynczak (1996) Antibody Therapy , Bios Scientific Pub.
  • Biological antagonists such as antibodies may be provided by continuous infusion, or by doses at intervals of, e.g., once per day, once per week, or 2 to 7 times per week, once every other week, or once per month.
  • a total weekly dose for an antibody is generally at least 0.05 ⁇ g/kg body weight, more generally at least 0.2 ⁇ g/kg, most generally at least 0.5 ⁇ g/kg, typically at least 1 ⁇ g/kg, more typically at least 10 ⁇ g/kg, most typically at least 100 ⁇ g/kg, preferably at least 0.2 mg/kg, more preferably at least 1.0 mg/kg, most preferably at least 2.0 mg/kg, optimally at least 10 mg/kg, more optimally at least 25 mg/kg, and most optimally at least 50 mg/kg (see, e.g., Yang, et al.
  • the desired dose of a small molecule therapeutic is about the same as for an antibody or polypeptide, on a moles/kg basis.
  • Determination of the appropriate dose is made by the clinician, e.g., using parameters or factors known or suspected in the art to affect treatment or predicted to affect treatment. Generally, the beginning dose is an amount somewhat less than the optimum dose and the dose is increased by small increments thereafter until the desired or optimum effect is achieved relative to any negative side effects.
  • Treatment regimens using IL-17 or IL-23 antagonists will typically be determined by the treating physician and will take into account the patient's age, medical history, disease symptoms, and tolerance for different types of medications and dosing regimens.
  • the treatment regimen is designed to suppress the overly aggressive immune system, allowing the body to eventually re-regulate itself, with the result often being that after the patient has been kept on systemic medications to suppress the inappropriate immune response for a finite length of time (for example, one year), medication can then be tapered and stopped without recurrence of the autoimmune attack. Sometimes resumption of the attack does occur, in which case the patient must be re-treated.
  • the physician may prescribe the patient a certain number of doses of the antagonist to be taken over a prescribed time period, after which therapy with the antagonist is discontinued.
  • the physician will continue the agonist therapy for some period of time, in which the amount and/or frequency of antagonist administered is gradually reduced before treatment is stopped.
  • the present invention also contemplates treatment regimens in which an IL-17 antagonist is used in combination with an IL-23 antagonist.
  • Such regimens may be especially useful in treating the acute phase of AOID, in which the IL-17 antagonist inhibits the activity of existing Th 17 cells, while the IL-23 antagonist prevents the generation of new Th 17 cells.
  • Such combination therapy may provide effective treatment of AOID using a lower dose of the IL-17 antagonist and/or administering the IL-17 antagonist for a shorter period of time.
  • therapy with IL-17 antagonist is preferably discontinued, while administration of the IL-23 antagonist is continued to prevent generation of new autoreactive Th 17 cells that could lead to recurrence of the disease.
  • the two antagonists may be administered at the same time in a single composition, or in separate compositions. Alternately, the two antagonists may be administered at separate intervals. Different doses of the antagonists may also be used. Similarly, a bispecific antagonist may also be administered during the acute phase and gradually withdrawn, followed by treatment with an IL-23 antagonist to maintain repression of the disease.
  • the treatment regimen may also include use of other therapeutic agents, to ameliorate one or more symptoms of the AOID or to prevent or ameliorate adverse effects from the antagonist therapy.
  • therapeutic agents that have been used to treat AOID symptoms are steroids and other anti-inflammatories.
  • steroids such as dexamethasone, flurometholone, and prednisolone
  • non-steroidal anti-inflammatories such as indomethacin, aspirin, flubiprofen and diclofenac
  • antimetabolites e.g., methotrexate, azathioprine
  • inhibitors of transcription factors e.g., cyclosporine, tacrolimus
  • DNA cross-linking agents e.g., cyclophosphamide, chlorambucil.
  • TNF inhibitors such as Infliximab (Remicade®, Centocor, Malvern, Pa.), Etanercept (Enbrel®, Amgen, Thousand Oaks, Calif.), and Adalimumab (Humira®, Abbott Laboratories, Abbott Park, Ill.) and specific inhibitors of IL-2 signaling, including Daclizumab (Zenapax®, Roche Laboratories, Nutley, N.J.) and Basiliximab (Simulect®, Novartis Pharmaceutical Co., East Hanover, N.J.).
  • TNF inhibitors such as Infliximab (Remicade®, Centocor, Malvern, Pa.), Etanercept (Enbrel®, Amgen, Thousand Oaks, Calif.), and Adalimumab (Humira®, Abbott Laboratories, Abbott Park, Ill.) and specific inhibitors of IL-2 signaling, including Daclizumab (Zenapax®, Roche Laboratories, Nutley, N.
  • any of the therapies described herein in which two or more different therapeutic substances are used e.g., an IL-17 antagonist and an IL-23 antagonist, or an IL-17 antagonist and a therapeutic agent that does not antagonize IL-17 or IL-23 activity
  • the different therapeutic substances are administered in association with each other, that is, they may be administered concurrently in the same pharmaceutical composition or as separate compositions or the substances may be administered at separate times, and in different orders.
  • Diagnosing the presence of an AOID in a patient will typically involve examining the patient for symptoms known to be consistent with such diseases.
  • the typical presentation of anterior uveitis involves pain, photophobia, and hyperlacrimation. Patients report a deep, dull, aching of the involved eye and surrounding orbit. Associated sensitivity to lights may be severe. Excessive tearing occurs secondary to increased neural stimulation of the lacrimal gland and the patient does not report a foreign-body sensation.
  • Visual acuity is variable ranging from mild blur to significant vision loss if synechiae or cyclitic membranes are present.
  • An examination may reveal mild to moderate lid swelling resulting in pseudoptosis.
  • a deep, perilimbal injection of the conjunctiva and episclera is typical, although the palpebral conjunctiva is characteristically normal.
  • the cornea may display mild edema.
  • Iris findings may include adhesions to the lens capsule (posterior synechiae) or, less commonly, to the peripheral cornea (anterior synechiae). Additionally, granulomatous nodules may appear on the surface of the iris.
  • Intraocular pressure (IOP) is initially reduced in the involved eye due to secretory hypotony of the ciliary body. However, as the reaction persists, inflammatory by-products may accumulate in the trabeculum. If this debris builds significantly, and if the ciliary body resumes its normal secretory output, IOP can rise sharply resulting in a secondary uveitic glaucoma.
  • Identifying patients who are susceptible for an AOID will typically taking a personal and family medical history, and may include genetic testing. For example, some individuals will have genetic predisposition to uveitis which is related to autoimmune disease processes. The most common of these susceptibility genes is the HLA B27 haplotype, which can predispose to uveitis alone or also to the Seronegative Spondyloarthropathies and the enteropathic arthropathies. Examples are ankylosing spondylitis, reactive arthritis (Reiters syndrome), psoriatic arthritis, irritable bowel disease and Crohn's disease. A patient may also be diagnosed as susceptible for an AOID if there was a family history of any of these autoimmune diseases, or the patient has already been diagnosed with such a disease.
  • the effectiveness of the antagonist therapy for preventing or treating AOID in a particular patient can be determined using diagnostic measures such as reduction or occurrence of inflammatory symptoms of, e.g., the amount of ocular inflammation or level of inflammatory cytokines in the affected eye(s).
  • diagnostic measures such as reduction or occurrence of inflammatory symptoms of, e.g., the amount of ocular inflammation or level of inflammatory cytokines in the affected eye(s).
  • the symptoms of ocular inflammation for the most part depend on the affected area of the eye. Most common signs and symptoms are: pain redness, floaters, decreased vision, and light sensitivity.
  • the level of inflammatory cytokines can be measured, e.g, by contacting a binding compound for the inflammatory cytokine of interest with a sample from the patient's eye as well as with a sample from a control subject or from unaffected tissue or fluid from the patient, and then comparing the cytokine levels detected by the binding compound.
  • Expression or activity from a control subject or control sample can be provided as a predetermined value, e.g., acquired from a statistically appropriate group of control subjects.
  • the present invention is based upon studies in IL-23p19 knockout (KO) mice and administration of anti-IL-23p19 and anti-IL-17 antibodies to murine models of autoimmune uveitis. These experiments were performed according to the Materials and Methods described in Section II below.
  • mice In the experiments involving IL-23p19 KO mice, the EAU susceptibility of IL-23p19 KO (IL-23 deficient) mice were compared to the EAU susceptibility of IL-12p35 KO (IL-12 deficient) and IL-12p40 KO (IL-12 and IL-23 deficient) mice. All mice were on the C57BL/6 background and the EAU induction and scoring was as described in General Methods below. It was found that IL-12p35 is not required for generation of IRBP-specific eye tissue destruction. In contrast, IL-23p19 is essential for development of EAU (Table 1).
  • Cytokine analysis of lymph node cell cultures derived from IRBP-immunized mice showed that the EAU susceptible IL-12 deficient mice (IL-12p35KO) had elevated levels of IFN- ⁇ , IL-6, IL-17 and IL-18, compared to IL-23 deficient mice (IL-23p19KO and IL-12p40KO).
  • Delayed hypersensitivity (DTH) responses to IRBP of the 3 KO strains examined by the ear swelling assay, showed that DTH response to IRBP was well correlated with the EAU scores or the respective mice, with significantly lower responses for p19 and p40 KO and significantly higher responses in p35 KO compared to wild-type (WT).
  • mice received 500 ⁇ g of the indicated antibodies every other day, starting the day before immunization, and the eyes and lymphoid organs were collected 17 days after immunization, or 6-7 days after disease onset in controls.
  • cytokine protein expression in the lymph nodes of these mice was assessed by multiplex ELISA. These data show that treatment with IL-23 antagonists lessens the production of Th1 and pro-inflammatory cytokines. The data are shown in Table 3.
  • mice were treated with 500 ⁇ g of anti-IL-23p19 antibody every other day starting 7 days after immunization and the disease was compared to mice that were treated from day before immunization (as above). EAU could be prevented by early treatment with either anti-p19 or anti-p40 antibodies. However, when treatment was started 7 days after immunization, a time point when uveitogenic effector T cells have already been primed and can be isolated from the LN and spleen, EAU development could not be aborted and the disease scores developed by treated mice were similar to control. This suggests that the requirement for IL-23 occurs at an early stage of disease pathogenesis. The data are shown in Table 4.
  • IL-17A ⁇ / ⁇ mice were immunized with a uveitogenic regimen of IRBP. Inhibition of EAU by genetic IL-17 deficiency was only partial (Table 5). The relatively modest reduction of EAU scores in IL-17 ⁇ / ⁇ mice might be explained by the fact that these mice are deficient for the IL-17A isoform of the cytokine, and under conditions of congenital deficiency might compensate with the usually less abundantly produced IL-17F isoform.
  • Fluorescent reagents suitable for modifying nucleic acids including nucleic acid primers and probes, polypeptides, and antibodies, for use, e.g., as diagnostic reagents, are available (Molecular Probes (2003) Catalogue , Molecular Probes, Inc., Eugene, Oreg.; Sigma-Aldrich (2003) Catalogue, St. Louis, Mo.).
  • IL-23 KO (p19 KO) was described in Cua, et al. (2003) Nature 421:744-748.
  • IL-17 ⁇ / ⁇ mice were produced as described in Nakae, et al. (2002) Immunity 17:375-387.
  • IL-12p35 KO (P35 KO)
  • IL-12p40 KO (P40 KO)
  • IFN- ⁇ KO (GKO) (all on C57BL/6 background) and C57BL/6 and B10RIII
  • mice were purchased from Jackson Laboratories. Animals were kept in a specific pathogen-free facility and given water and standard laboratory chow ad libitum. Animal care and use were in compliance with institutional guidelines and with the Association for Research in Vision and Ophthalmology Statement for the Use of Animals in Ophthalmic and Vision Research.
  • IRBP was isolated from bovine retinas, as described previously, using Con A-Sepharose affinity chromatography and fast performance liquid chromatography (see, e.g., Pepperberg et al. (1991) Photochem Photobiol 54:1057-1060). IRBP preparations were aliquoted and stored at ⁇ 70° C. Human IRBP-derived peptide 161-180 (Karabezekian, Z. et al., (2005) Invest Ophthalmol Vis Sci. 46(10):3769-76) was synthesized by Fmoc chemistry (model 432A peptide synthesizer; Applied Biosystems, Foster City, Calif.).
  • Neutralizing anti-mouse IL-23 and anti-mouse IL-17A antibodies were provided by Schering-Plough Biopharma (Palo Alto, Calif.). Anti-mouse IL-23 was described previously (see, e.g., Langrish et al. (2005) J Exp Med 201:233-240). The C17.8 (anti-IL-12p40, rat IgG2a) hybridoma was provided by the Wistar Institute, Philadelphia, Pa. Monoclonal antibody was produced in ascites and purified by ion exchange HPLC by Harlan Bioproducts for Science (Indianapolis, Ind.).
  • FITC-labeled anti-mouse CD4 (clone-L3T4)
  • PE-labeled anti-mouse IL-17 (clone-TC11-18H10)
  • APC-labeled anti-IFN- ⁇ (clone-XMG1.2)
  • cytokine secretion blocker (GolgiStopTM)
  • PMA lonomycin were purchased from LC Laboratories (Boston, Mass.).
  • EAU was induced by active immunization with 150 ⁇ g of IRBP for C57BL/6 mice and with 7 ⁇ g IRBP peptide 161-180 for B10RIII mice (Jackson Labs, Maine).
  • Bordetella pertussis toxin 0.5 ⁇ g/mouse
  • PBS containing 2% normal mouse serum was given by intraperitoneal injection concurrently with immunization and in some experiments the IRBP was spiked with 500 ⁇ g of IRBP peptide 1-20 (Avichezer, D. et al. (2000), Invest Ophthalmol Vis Sci. 41(1): 127-31) to enhance the usually modest disease scores seen in this strain.
  • Antigen solution was emulsified 1:1 v/v in CFA that had been supplemented with Mycobacterium tuberculosis strain H37RA to 2.5 mg/ml. A total of 200 ⁇ l of emulsion was injected s.c., divided into 3 sites (base of the tail and both thighs).
  • EAU was induced by adoptive transfer of a uveitogenic T cell line (see below). 1-2 million cells, freshly stimulated with antigen, were injected intraperitoneally. Clinical EAU was evaluated by fundoscopy under a binocular microscope after dilation of the pupil and was graded on a scale of 0-4 using criteria based on the extent of inflammatory lesions, as described in detail elsewhere (see, e.g., Agarwal and Caspi, (2004) Methods Mol Med 102:395-419; and Chan et al. (1990) J Autoimmun 3:247-255).
  • Delayed Type Hypersensitivity (DTH) to IRBP was evaluated by the ear swelling assay (see, e.g., Tarrant et al. (1998) J Immunol 161:122-127).
  • DTH Delayed Type Hypersensitivity
  • the spleen and draining lymph nodes (inguinal and iliac) (5 per group) were collected at the end of each experiment as indicated. Lymphoid cells were pooled within the group, and were incubated with graded doses of Ag in triplicate 0.2-ml cultures, essentially as described (see, e.g., Avichezer et al. (2000) Invest Ophthalmol Vis Sci 41:127-131). Proliferation was determined by [ 3 H]thymidine uptake.
  • Cytokines were quantitated in 48-h Ag-stimulated supernatants using the Pierce Multiplex SearchLight Arrays technology (see, e.g., Moody et al. (2001) Biotechniques 31:186-190, 192-184).
  • mice were immunized with IRBP or IRBP uveitogenic peptide (161-180) as indicated. Mice were injected intraperitoneally with 0.5 mg per dose of anti-p19, anti-p40, or anti-IL-17. Treatment was given every other day starting on day -1 through day 15 after immunization, covering both priming and effector phase (prevention protocol) or starting day 7 through day 15, covering the effector phase only (treatment). Controls were given the same regimen of isotype (rat IgG1). Eyes and lymphoid organs were harvested on day 17, 6-7 days after disease onset.
  • the uveitogenic Th1 cell line specific to a peptide of human IRBP has been described (see, e.g., Silver et al. (1995) Invest Ophthalmol Vis Sci 36:946-954). Briefly, the line was derived from draining lymph nodes of B10.RIII mice immunized with human IRBP peptide 161-180, polarized in vitro toward the Th1 phenotype by culture in the presence of antigen, IL-12, and anti-IL-4.
  • the cells were maintained by alternating cycles of expansion in IL-2 and restimulation with 1 ⁇ g/ml of p161-180 every 2 to 3 weeks in the presence of syngeneic splenocytes, irradiated with 3000 rads, as APCs.
  • syngeneic splenocytes irradiated with 3000 rads, as APCs.
  • EAU induction cells freshly stimulated with Ag for 48 h were injected i.p. into naive syngeneic recipients.
  • T cell line was stimulated with 1 ⁇ g/ml IRBP peptide 161-180 in the presence of irradiated APCs for 24 h with the addition of GolgiStopTM protein transfer inhibitor(BD Biosciences, San Jose, Calif.) at the last 4 h. Thereafter, cells were separated on Ficoll, washed and stained for extracellular CD4. Than cells were washed, fixed, permeabilized with Cytofix/CytopermTM fixation and permeabilization buffer (BD Biosciences) and stained with PE-conjugated anti Il-17 and APC-conjugated anti IFN- ⁇ for FACS analysis.
  • GolgiStopTM protein transfer inhibitor BD Biosciences, San Jose, Calif.
  • T cell line was stimulated for 5 days with antigen (1 ⁇ g/ml IRBP peptide 161-180) or antigen+rIL-23 (10 ng/ml) or antigen+IL-23+anti IFN- ⁇ (10 ⁇ g/ml) in the presence of irradiated APCs. During the last 4 h of incubation cells were stimulated with PMA and lonomycin with the addition of GolgiStopTM protein transfer inhibitor (BD Biosciences). Thereafter cells were treated and stained for intracellular IL-17 and IFN- ⁇ as mentioned above.
  • the T cell line was adoptively transferred (2 ⁇ 10 6 /mouse) i.v. to na ⁇ ve Thy1.1/.2 heterozygous mice.
  • spleens were harvested and splenocytes were stimulated with IRBP peptide 161-180 for 24 h with the presence of PMA, lonomycin and GolgiStopTM protein transfer inhibitor (BD Biosciences) at the last 4 h. Thereafter cells were treated and stained for intracellular IL-17 and IFN- ⁇ as mentioned above.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Rheumatology (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Endocrinology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Pulmonology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Communicable Diseases (AREA)
  • Pain & Pain Management (AREA)
  • Transplantation (AREA)
  • Oncology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Steroid Compounds (AREA)
US11/512,622 2005-09-01 2006-08-30 Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease Abandoned US20080199460A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US11/512,622 US20080199460A1 (en) 2005-09-01 2006-08-30 Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease
US12/643,152 US8524230B2 (en) 2005-09-01 2009-12-21 Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease
US12/643,166 US20100111954A1 (en) 2005-09-01 2009-12-21 Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
US13/015,900 US20110142831A1 (en) 2005-09-01 2011-01-28 Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
US13/960,441 US20130323251A1 (en) 2005-09-01 2013-08-06 Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
US14/274,905 US20140248279A1 (en) 2005-09-01 2014-05-12 Use of il-23 and il-17 antiagonists to treat autoimmune ocular inflammatory disease

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US71379205P 2005-09-01 2005-09-01
US83731206P 2006-08-11 2006-08-11
US11/512,622 US20080199460A1 (en) 2005-09-01 2006-08-30 Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US12/643,166 Continuation US20100111954A1 (en) 2005-09-01 2009-12-21 Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
US12/643,152 Division US8524230B2 (en) 2005-09-01 2009-12-21 Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease
US13/015,900 Continuation US20110142831A1 (en) 2005-09-01 2011-01-28 Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease

Publications (1)

Publication Number Publication Date
US20080199460A1 true US20080199460A1 (en) 2008-08-21

Family

ID=37686002

Family Applications (6)

Application Number Title Priority Date Filing Date
US11/512,622 Abandoned US20080199460A1 (en) 2005-09-01 2006-08-30 Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease
US12/643,166 Abandoned US20100111954A1 (en) 2005-09-01 2009-12-21 Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
US12/643,152 Expired - Fee Related US8524230B2 (en) 2005-09-01 2009-12-21 Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease
US13/015,900 Abandoned US20110142831A1 (en) 2005-09-01 2011-01-28 Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
US13/960,441 Abandoned US20130323251A1 (en) 2005-09-01 2013-08-06 Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
US14/274,905 Abandoned US20140248279A1 (en) 2005-09-01 2014-05-12 Use of il-23 and il-17 antiagonists to treat autoimmune ocular inflammatory disease

Family Applications After (5)

Application Number Title Priority Date Filing Date
US12/643,166 Abandoned US20100111954A1 (en) 2005-09-01 2009-12-21 Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
US12/643,152 Expired - Fee Related US8524230B2 (en) 2005-09-01 2009-12-21 Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease
US13/015,900 Abandoned US20110142831A1 (en) 2005-09-01 2011-01-28 Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
US13/960,441 Abandoned US20130323251A1 (en) 2005-09-01 2013-08-06 Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
US14/274,905 Abandoned US20140248279A1 (en) 2005-09-01 2014-05-12 Use of il-23 and il-17 antiagonists to treat autoimmune ocular inflammatory disease

Country Status (22)

Country Link
US (6) US20080199460A1 (pt)
EP (3) EP1933869B1 (pt)
JP (3) JP2009507023A (pt)
CN (1) CN101296706B (pt)
AT (1) ATE445415T1 (pt)
AU (1) AU2006284841B2 (pt)
BR (1) BRPI0615297A2 (pt)
CA (1) CA2621086A1 (pt)
CY (1) CY1110606T1 (pt)
DE (1) DE602006009834D1 (pt)
DK (1) DK1933869T3 (pt)
ES (1) ES2333260T3 (pt)
HK (1) HK1119578A1 (pt)
HR (1) HRP20100014T1 (pt)
NO (1) NO20081574L (pt)
NZ (2) NZ566424A (pt)
PL (1) PL1933869T3 (pt)
PT (1) PT1933869E (pt)
RS (1) RS51142B (pt)
SI (1) SI1933869T1 (pt)
TW (2) TWI372061B (pt)
WO (1) WO2007027761A2 (pt)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100129355A1 (en) * 2007-07-26 2010-05-27 Osaka University Therapeutic agents for ocular inflammatory disease comprising interleukin 6 receptor inhibitor as active ingredient
US20100183587A1 (en) * 2009-01-09 2010-07-22 Schepens Eye Research Institute Therapeutic Compositions for Treatment of Corneal Disorders
US20100215666A1 (en) * 2004-08-05 2010-08-26 Di Padova Franco E Il-17 antagonistic antibodies
US20110104236A1 (en) * 2008-01-09 2011-05-05 Reza Dana Therapeutic compositions for treatment of ocular inflammatory disorders
WO2011159655A2 (en) 2010-06-16 2011-12-22 Allergan, Inc. IL23p19 ANTIBODY INHIBITOR FOR TREATING OCULAR AND OTHER CONDITIONS
US20120014970A1 (en) * 2009-01-09 2012-01-19 Reza Dana Therapeutic Compositions for Treatment of Corneal Disorders
US8137671B2 (en) 2009-05-05 2012-03-20 Genentech, Inc. Anti-IL-17F antibodies
US8715669B2 (en) 2008-05-05 2014-05-06 Novimmune Sa Anti-IL-17A/IL-17F cross-reactive antibodies
US8778346B2 (en) 2010-11-04 2014-07-15 Boehringer Ingelheim International Gmbh Anti-IL-23 antibodies
US8853150B2 (en) 2010-07-29 2014-10-07 Eleven Biotherapeutics, Inc. Chimeric IL-1 receptor type I antagonists
US9284283B2 (en) 2012-02-02 2016-03-15 Ensemble Therapeutics Corporation Macrocyclic compounds for modulating IL-17
US10059763B2 (en) 2014-09-03 2018-08-28 Boehringer Ingelheim International Gmbh Compound targeting IL-23A and TNF-alpha and uses thereof
US10507241B2 (en) 2014-07-24 2019-12-17 Boehringer Ingelheim International Gmbh Biomarkers useful in the treatment of IL-23A related diseases
US10799589B2 (en) 2013-03-13 2020-10-13 Buzzard Pharmaceuticals AB Chimeric cytokine formulations for ocular delivery
US11078265B2 (en) 2012-05-03 2021-08-03 Boehringer Ingelheim International Gmbh Anti-IL-23 antibodies

Families Citing this family (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009518314A (ja) 2005-12-02 2009-05-07 ジェネンテック・インコーポレーテッド Il−22に結合する抗体およびil−22rに結合する抗体を含む、サイトカインシグナリングに関連する疾患および障害の処置のための組成物および方法
US7910703B2 (en) 2006-03-10 2011-03-22 Zymogenetics, Inc. Antagonists to IL-17A, IL-17F, and IL-23P19 and methods of use
US7790862B2 (en) 2006-06-13 2010-09-07 Zymogenetics, Inc. IL-17 and IL-23 antagonists and methods of using the same
US7833527B2 (en) 2006-10-02 2010-11-16 Amgen Inc. Methods of treating psoriasis using IL-17 Receptor A antibodies
NZ579297A (en) * 2007-02-28 2012-03-30 Schering Corp Combination therapy comprising an il-23 antagonist and a cytokine antagonist for treatment of immune disorders
EP2150564A2 (en) * 2007-04-27 2010-02-10 ZymoGenetics, Inc. Antagonists to il-17a, il-17f, and il-23p19 and methods of use
WO2009023267A2 (en) * 2007-08-15 2009-02-19 President And Fellows Of Harvard College Methods for modulating development and expansion of il-17 expressing cells
WO2009068630A1 (en) 2007-11-27 2009-06-04 Ablynx N.V. Immunoglobulin constructs
WO2009082624A2 (en) * 2007-12-10 2009-07-02 Zymogenetics, Inc. Antagonists of il-17a, il-17f, and il-23 and methods of using the same
BRPI0907196B1 (pt) 2008-02-21 2021-05-25 Kirin-Amgen Inc Uso de um antagonista de il-17ra-il-17rb
KR101766927B1 (ko) * 2009-04-01 2017-08-09 제넨테크, 인크. 인슐린 저항성 장애의 치료
EP2480251A1 (en) 2009-09-21 2012-08-01 Peptinov SAS Carrier conjugates of il-23-peptides and their induced antibodies
MY182680A (en) 2010-01-15 2021-01-29 Amgen K A Inc Antibody formulation and therapeutic regimens
EP4137514A1 (en) * 2010-10-08 2023-02-22 Novartis AG Methods of treating psoriasis using il-17 antagonists
GB201112091D0 (en) 2011-07-14 2011-08-31 Gt Biolog Ltd Bacterial strains isolated from pigs
GB201117313D0 (en) 2011-10-07 2011-11-16 Gt Biolog Ltd Bacterium for use in medicine
WO2013059644A1 (en) * 2011-10-21 2013-04-25 Baylor College Of Medicine Method to measure inflammation in the conjunctiva of patients with tear dysfunction
BR112014024903A2 (pt) 2012-04-05 2017-07-11 Hoffmann La Roche anticorpos biespecíficos contra tweak humanao e il17 humana e seus usos
US9708401B2 (en) 2012-05-22 2017-07-18 Bristol-Myers Squibb Company IL-17A/F cross-reactive monoclonal antibodies and methods of using the same
WO2014093206A1 (en) * 2012-12-13 2014-06-19 Merck Sharp & Dohme Corp. Lyophilized spherical pellets of anti-il-23 antibodies
WO2014107737A2 (en) * 2013-01-07 2014-07-10 Eleven Biotherapeutics, Inc. Local delivery of il-17 inhibitors for treating ocular disease
GB201306536D0 (en) 2013-04-10 2013-05-22 Gt Biolog Ltd Polypeptide and immune modulation
US9603882B2 (en) 2013-08-13 2017-03-28 Industrial Technology Research Institute Method for modulating Th17 cells and method for treating a disease related to modulation of Th17 cells
AR102417A1 (es) * 2014-11-05 2017-03-01 Lilly Co Eli Anticuerpos biespecíficos anti-tnf- / anti-il-23
SG11201704811YA (en) 2014-12-23 2017-07-28 4D Pharma Res Ltd Immune modulation
BR112017013274A2 (pt) 2014-12-23 2018-02-06 4D Pharma Research Limited polipeptídeo e imunomodulação
WO2016109836A2 (en) 2014-12-31 2016-07-07 Chad David Andresen Antenna assembly
JP2018506275A (ja) * 2015-01-28 2018-03-08 ジェネンテック, インコーポレイテッド 多発性硬化症の遺伝子発現マーカー及び治療
CN104725514A (zh) * 2015-02-06 2015-06-24 中国药科大学 新型il23拮抗剂
CN115364122A (zh) 2015-06-15 2022-11-22 4D制药研究有限公司 包含细菌菌株的组合物
NZ737752A (en) 2015-06-15 2022-02-25 4D Pharma Res Ltd Compositions comprising bacterial strains
MA41010B1 (fr) 2015-06-15 2020-01-31 4D Pharma Res Ltd Compositions comprenant des souches bactériennes
SI3307288T1 (sl) 2015-06-15 2019-11-29 4D Pharma Res Ltd Spojine, ki vsebujejo bakterijske seve
MA41060B1 (fr) 2015-06-15 2019-11-29 4D Pharma Res Ltd Compositions comprenant des souches bactériennes
JP6909208B2 (ja) * 2015-09-17 2021-07-28 アムジェン インコーポレイテッド Il23経路バイオマーカーを使用するil23アンタゴニストに対する臨床応答の予測
GB201520497D0 (en) 2015-11-20 2016-01-06 4D Pharma Res Ltd Compositions comprising bacterial strains
CN112569262A (zh) 2015-11-20 2021-03-30 4D制药研究有限公司 包含细菌菌株的组合物
GB201520638D0 (en) 2015-11-23 2016-01-06 4D Pharma Res Ltd Compositions comprising bacterial strains
GB201520631D0 (en) * 2015-11-23 2016-01-06 4D Pharma Res Ltd Compositions comprising bacterial strains
GB201612191D0 (en) 2016-07-13 2016-08-24 4D Pharma Plc Compositions comprising bacterial strains
CN108883139B (zh) 2016-03-04 2022-04-26 4D制药有限公司 包含细菌菌株的组合物
TW201821093A (zh) 2016-07-13 2018-06-16 英商4D製藥有限公司 包含細菌菌株之組合物
GB201621123D0 (en) 2016-12-12 2017-01-25 4D Pharma Plc Compositions comprising bacterial strains
MA48939B1 (fr) 2017-05-22 2021-06-30 4D Pharma Res Ltd Compositions comprenant des souches bactériennes
EP3630942B1 (en) 2017-05-24 2022-11-30 4D Pharma Research Limited Compositions comprising bacterial strain
RS61210B1 (sr) 2017-06-14 2021-01-29 4D Pharma Res Ltd Kompozicije koje sadrže bakterijske sojeve
JP6837581B2 (ja) 2017-06-14 2021-03-03 フォーディー ファーマ リサーチ リミテッド4D Pharma Research Limited Megasphaera属の細菌株を含む組成物及びその使用
WO2022031871A1 (en) 2020-08-05 2022-02-10 Synthekine, Inc. Compositions and methods related to il27 receptor binding
CA3233721A1 (en) * 2021-10-03 2023-04-06 Sa XIAO Methods of treating cancer and the pharmaceutical compositions thereof

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6680057B1 (en) * 1995-03-23 2004-01-20 Immunex Corporation Methods of treating autoimmune disease by administering interleukin-17 receptor
US20040156849A1 (en) * 2002-10-30 2004-08-12 Gurney Austin L. Inhibition of IL-17 production
US20040219150A1 (en) * 2003-02-06 2004-11-04 Cua Daniel J. Uses of mammalian cytokine; related reagents
US20060011429A1 (en) * 2004-07-13 2006-01-19 Lg Electronics Inc. Damper for damping vibration and washing machine having the same
US20060110429A1 (en) * 2004-11-24 2006-05-25 Therakine Corporation Implant for intraocular drug delivery
US20060135518A1 (en) * 2004-11-19 2006-06-22 Synta Pharmaceuticals Corp. Pyrimidine compounds and uses thereof
US20070009526A1 (en) * 2005-06-30 2007-01-11 Jacqueline Benson Anti-IL-23 antibodies, compositions, methods and uses

Family Cites Families (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
ATE87659T1 (de) 1986-09-02 1993-04-15 Enzon Lab Inc Bindungsmolekuele mit einzelpolypeptidkette.
US5260203A (en) 1986-09-02 1993-11-09 Enzon, Inc. Single polypeptide chain binding molecules
WO1988007089A1 (en) 1987-03-18 1988-09-22 Medical Research Council Altered antibodies
DE3920358A1 (de) 1989-06-22 1991-01-17 Behringwerke Ag Bispezifische und oligospezifische, mono- und oligovalente antikoerperkonstrukte, ihre herstellung und verwendung
ATE297465T1 (de) 1991-11-25 2005-06-15 Enzon Inc Verfahren zur herstellung von multivalenten antigenbindenden proteinen
US6005079A (en) 1992-08-21 1999-12-21 Vrije Universiteit Brussels Immunoglobulins devoid of light chains
DK1087013T3 (da) 1992-08-21 2009-05-11 Univ Bruxelles Immunoglobuliner uden lette kæder
DK0698097T3 (da) 1993-04-29 2001-10-08 Unilever Nv Produktion af antistoffer eller (funktionaliserede) fragmenter deraf afledt af Camelidae-immunoglobuliner med tung kæde
US6132764A (en) 1994-08-05 2000-10-17 Targesome, Inc. Targeted polymerized liposome diagnostic and treatment agents
JP4373495B2 (ja) 1995-03-23 2009-11-25 イミュネックス・コーポレーション Il−17受容体
US6056973A (en) 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
US6326482B1 (en) 1999-04-23 2001-12-04 Genentech, Inc. SH2 domain-containing peptides
DE69942671D1 (de) 1998-12-01 2010-09-23 Facet Biotech Corp Humanisierte antikoerper gegen gamma-interferon
JP2004504064A (ja) 2000-06-22 2004-02-12 アムジエン・インコーポレーテツド Il−17分子およびその用途
WO2002097048A2 (en) 2001-05-30 2002-12-05 Centocor, Inc. ANTI-p40 IMMUNOGLOBULIN DERIVED PROTEINS, COMPOSITIONS, METHODS AND USES
US20050154046A1 (en) 2004-01-12 2005-07-14 Longgui Wang Methods of treating an inflammatory-related disease
WO2003086310A2 (en) 2002-04-12 2003-10-23 Ramot At Tel Aviv University Ltd. Prevention of brain inflammation as a result of induced autoimmune response
DK1601694T3 (da) 2003-03-10 2009-12-14 Schering Corp Anvendelser af IL23-antagonister, relaterede reagenser
WO2004101750A2 (en) * 2003-05-09 2004-11-25 Centocor, Inc. IL-23p40 SPECIFIC IMMUNOGLOBULIN DERIVED PROTEINS, COMPOSITIONS, METHODS AND USES
US7410483B2 (en) 2003-05-23 2008-08-12 Novare Surgical Systems, Inc. Hand-actuated device for remote manipulation of a grasping tool
EP1641822B1 (en) 2003-07-08 2013-05-15 Genentech, Inc. Il-17 a/f heterologous polypeptides and therapeutic uses thereof
JP2007535930A (ja) 2004-05-03 2007-12-13 シェーリング コーポレイション 皮膚の炎症を予測するためのil−17発現の使用;処置方法
US20050287593A1 (en) 2004-05-03 2005-12-29 Schering Corporation Use of cytokine expression to predict skin inflammation; methods of treatment
WO2005120571A2 (en) 2004-06-07 2005-12-22 Ramot At Tel Aviv University Ltd. Method of passive immunization against disease or disorder characterized by amyloid aggregation with diminished risk of neuroinflammation
CA2572334A1 (en) * 2004-07-01 2006-01-19 New York University Compositions and methods for modulation of ror.gamma.t
GB0417487D0 (en) * 2004-08-05 2004-09-08 Novartis Ag Organic compound
AR051444A1 (es) 2004-09-24 2007-01-17 Centocor Inc Proteinas derivadas de inmunoglobulina especifica de il-23p40, composiciones, epitopos, metodos y usos
EP1853707A2 (en) * 2004-12-20 2007-11-14 Schering Corporation Uses of il-23 antagonists in the treatment of diabetes mellitus
NZ583153A (en) 2004-12-21 2011-06-30 Centocor Ortho Biotech Inc Anti-IL-12 antibodies, epitopes, compositions, methods and uses
AU2006214473A1 (en) * 2005-02-14 2006-08-24 Wyeth Interleukin-17F antibodies and other IL-17F signaling antagonists and uses therefor
CN103172737A (zh) 2005-06-30 2013-06-26 Abbvie公司 Il-12/p40结合蛋白
US7807160B2 (en) 2005-08-31 2010-10-05 Schering Corporation Engineered anti-IL-23 antibodies
MX2008008621A (es) 2005-12-29 2008-11-27 Centocor Inc Anticuerpos anti-il-23 humanos, composiciones, metodos y usos.
TW200815469A (en) * 2006-06-23 2008-04-01 Astrazeneca Ab Compounds

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6680057B1 (en) * 1995-03-23 2004-01-20 Immunex Corporation Methods of treating autoimmune disease by administering interleukin-17 receptor
US20040156849A1 (en) * 2002-10-30 2004-08-12 Gurney Austin L. Inhibition of IL-17 production
US20040219150A1 (en) * 2003-02-06 2004-11-04 Cua Daniel J. Uses of mammalian cytokine; related reagents
US20060011429A1 (en) * 2004-07-13 2006-01-19 Lg Electronics Inc. Damper for damping vibration and washing machine having the same
US20060135518A1 (en) * 2004-11-19 2006-06-22 Synta Pharmaceuticals Corp. Pyrimidine compounds and uses thereof
US20060110429A1 (en) * 2004-11-24 2006-05-25 Therakine Corporation Implant for intraocular drug delivery
US20070009526A1 (en) * 2005-06-30 2007-01-11 Jacqueline Benson Anti-IL-23 antibodies, compositions, methods and uses

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10344084B2 (en) 2004-08-05 2019-07-09 Novartis Ag IL-17 antagonistic antibodies
US9765140B2 (en) 2004-08-05 2017-09-19 Novartis Ag IL-17 antagonistic antibodies
US20100215666A1 (en) * 2004-08-05 2010-08-26 Di Padova Franco E Il-17 antagonistic antibodies
US8617552B2 (en) 2004-08-05 2013-12-31 Novarts Ag IL-17 antibodies
US8119131B2 (en) 2004-08-05 2012-02-21 Novartis Ag IL-17 antagonistic antibodies
US20160326255A1 (en) * 2007-07-26 2016-11-10 Osaka University Therapeutic agents for ocular inflammatory disease comprising interleukin 6 receptor inhibitor as active ingredient
US20100129355A1 (en) * 2007-07-26 2010-05-27 Osaka University Therapeutic agents for ocular inflammatory disease comprising interleukin 6 receptor inhibitor as active ingredient
US11241497B2 (en) 2008-01-09 2022-02-08 The Schepens Eye Research Institute, Inc. Therapeutic compositions for treatment of ocular inflammatory disorders
US20110104236A1 (en) * 2008-01-09 2011-05-05 Reza Dana Therapeutic compositions for treatment of ocular inflammatory disorders
US9872901B2 (en) 2008-01-09 2018-01-23 The Schepens Eye Research Institute, Inc. Therapeutic compositions for treatment of ocular inflammatory disorders
WO2009089036A3 (en) * 2008-01-09 2016-03-24 Schepens Eye Research Institute Therapeutic compositions for treatment of ocular inflammatory disorders
US9309313B2 (en) * 2008-01-09 2016-04-12 The Schepens Eye Research Institute, Inc. Therapeutic compositions for treatment of ocular inflammatory disorders
US8715669B2 (en) 2008-05-05 2014-05-06 Novimmune Sa Anti-IL-17A/IL-17F cross-reactive antibodies
US8771697B2 (en) 2008-05-05 2014-07-08 Novimmune Sa Methods of treatment using anti-IL-17A/IL-17F cross-reactive antibodies
US9650437B2 (en) 2008-05-05 2017-05-16 Novimmune S.A. Nucleic acid encoding and method of producing anti-IL-17A/IL-17F cross-reactive antibodies
US20120014970A1 (en) * 2009-01-09 2012-01-19 Reza Dana Therapeutic Compositions for Treatment of Corneal Disorders
US10117906B2 (en) * 2009-01-09 2018-11-06 The Schepens Eye Research Institute, Inc. Methods for reducing corneal nerves damage, corneal lymphangiogenesis or immunity to corneal antigens in dry-eye associated ocular surface diseases by IL-1Ra
US20100183587A1 (en) * 2009-01-09 2010-07-22 Schepens Eye Research Institute Therapeutic Compositions for Treatment of Corneal Disorders
US8609093B2 (en) 2009-05-05 2013-12-17 Novimmune S. A. Methods of treatment using anti-IL-17F antibodies
US8137671B2 (en) 2009-05-05 2012-03-20 Genentech, Inc. Anti-IL-17F antibodies
US9475873B2 (en) 2009-05-05 2016-10-25 Novimmune Sa Nucleic acids encoding anti-IL-17F antibodies and methods of use thereof
WO2011159655A2 (en) 2010-06-16 2011-12-22 Allergan, Inc. IL23p19 ANTIBODY INHIBITOR FOR TREATING OCULAR AND OTHER CONDITIONS
US9458216B2 (en) 2010-07-29 2016-10-04 Eleven Biotherapeutics, Inc. Nucleic acid encoding chimeric IL-1 receptor type I antagonists
US8853150B2 (en) 2010-07-29 2014-10-07 Eleven Biotherapeutics, Inc. Chimeric IL-1 receptor type I antagonists
US9441036B2 (en) 2010-11-04 2016-09-13 Boehringer Ingelheim International Gmbh Anti-IL-23 antibodies
US8778346B2 (en) 2010-11-04 2014-07-15 Boehringer Ingelheim International Gmbh Anti-IL-23 antibodies
US10202448B2 (en) 2010-11-04 2019-02-12 Boehringer Ingelheim International Gmbh Anti-IL-23 antibodies
US9284283B2 (en) 2012-02-02 2016-03-15 Ensemble Therapeutics Corporation Macrocyclic compounds for modulating IL-17
US11078265B2 (en) 2012-05-03 2021-08-03 Boehringer Ingelheim International Gmbh Anti-IL-23 antibodies
US10799589B2 (en) 2013-03-13 2020-10-13 Buzzard Pharmaceuticals AB Chimeric cytokine formulations for ocular delivery
US10507241B2 (en) 2014-07-24 2019-12-17 Boehringer Ingelheim International Gmbh Biomarkers useful in the treatment of IL-23A related diseases
US10793629B2 (en) 2014-09-03 2020-10-06 Boehringer Ingelheim International Gmbh Compound targeting IL-23A and TNF-alpha and uses thereof
US10059763B2 (en) 2014-09-03 2018-08-28 Boehringer Ingelheim International Gmbh Compound targeting IL-23A and TNF-alpha and uses thereof
US11680096B2 (en) 2014-09-03 2023-06-20 Boehringer Ingelheim International Gmbh Compound targeting IL-23A and TNF-alpha and uses thereof

Also Published As

Publication number Publication date
EP2116258A1 (en) 2009-11-11
US20100111954A1 (en) 2010-05-06
WO2007027761A3 (en) 2007-05-03
CA2621086A1 (en) 2007-03-08
JP2011184466A (ja) 2011-09-22
CY1110606T1 (el) 2015-04-29
US8524230B2 (en) 2013-09-03
AU2006284841B2 (en) 2012-11-08
HK1119578A1 (en) 2009-03-13
AU2006284841A1 (en) 2007-03-08
EP2322219A3 (en) 2013-06-05
TWI372061B (en) 2012-09-11
BRPI0615297A2 (pt) 2011-05-17
ES2333260T3 (es) 2010-02-18
CN101296706B (zh) 2011-11-30
TW201235050A (en) 2012-09-01
EP1933869A2 (en) 2008-06-25
US20130323251A1 (en) 2013-12-05
PT1933869E (pt) 2009-12-24
US20110142831A1 (en) 2011-06-16
DE602006009834D1 (de) 2009-11-26
PL1933869T3 (pl) 2010-06-30
WO2007027761A2 (en) 2007-03-08
NZ566424A (en) 2011-10-28
NZ595262A (en) 2013-03-28
US20100111950A1 (en) 2010-05-06
NO20081574L (no) 2008-05-30
US20140248279A1 (en) 2014-09-04
HRP20100014T1 (hr) 2010-02-28
TW200800259A (en) 2008-01-01
CN101296706A (zh) 2008-10-29
JP5462838B2 (ja) 2014-04-02
JP2009507023A (ja) 2009-02-19
EP2322219A2 (en) 2011-05-18
DK1933869T3 (da) 2010-03-01
SI1933869T1 (sl) 2010-02-26
TWI463993B (zh) 2014-12-11
ATE445415T1 (de) 2009-10-15
EP1933869B1 (en) 2009-10-14
JP2014005285A (ja) 2014-01-16
RS51142B (sr) 2010-10-31

Similar Documents

Publication Publication Date Title
US8524230B2 (en) Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease
EP2056838B1 (en) Combination therapy for treatment of immune disorders
KR20170045222A (ko) 항-tnf-알파 항체 요법을 받은 건선 환자의 치료 방법
US11279755B2 (en) Use of IL-20 antagonists for treating eye diseases
AU2014200946A1 (en) Combination therapy for treatment of immune disorders

Legal Events

Date Code Title Description
AS Assignment

Owner name: SCHERING CORPORATION, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CUA, DANIEL J.;KASTELEIN, ROBERT A.;TSAI, VAN T.;REEL/FRAME:018976/0026;SIGNING DATES FROM 20061104 TO 20061117

Owner name: NATIONAL INSTITUTES OF HEALTH - OFFICE OF TECHNOLO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CASPI, RACHEL;SILVER, PHYLLIS;LUGER, DROR;REEL/FRAME:018976/0034

Effective date: 20061130

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION