US20140079699A1 - Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r) - Google Patents

Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r) Download PDF

Info

Publication number
US20140079699A1
US20140079699A1 US14/014,446 US201314014446A US2014079699A1 US 20140079699 A1 US20140079699 A1 US 20140079699A1 US 201314014446 A US201314014446 A US 201314014446A US 2014079699 A1 US2014079699 A1 US 2014079699A1
Authority
US
United States
Prior art keywords
antibody
csf1r
mmp
tnf
seq
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
US14/014,446
Other languages
English (en)
Inventor
Brian Wong
Emma Masteller
Kris Reedquist
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.)
Five Prime Therapeutics Inc
Original Assignee
Five Prime Therapeutics Inc
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
Application filed by Five Prime Therapeutics Inc filed Critical Five Prime Therapeutics Inc
Priority to US14/014,446 priority Critical patent/US20140079699A1/en
Assigned to FIVE PRIME THERAPEUTICS, INC. reassignment FIVE PRIME THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: REEDQUIST, Kris, MASTELLER, EMMA, WONG, BRIAN
Publication of US20140079699A1 publication Critical patent/US20140079699A1/en
Priority to US15/154,822 priority patent/US10221243B2/en
Priority to US16/256,311 priority patent/US10822421B2/en
Priority to US17/026,853 priority patent/US20210107986A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • 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
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • 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
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • 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
    • 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
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/564Immunoassay; Biospecific binding assay; Materials therefor for pre-existing immune complex or autoimmune disease, i.e. systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, rheumatoid factors or complement components C1-C9
    • 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
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/95Proteinases, i.e. endopeptidases (3.4.21-3.4.99)
    • G01N2333/964Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue
    • G01N2333/96425Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals
    • G01N2333/96427Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general
    • G01N2333/9643Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general with EC number
    • G01N2333/96486Metalloendopeptidases (3.4.24)
    • G01N2333/96491Metalloendopeptidases (3.4.24) with definite EC number
    • G01N2333/96494Matrix metalloproteases, e. g. 3.4.24.7
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • CSF1R colony stimulating factor 1 receptor
  • Colony stimulating factor 1 receptor (referred to herein as CSF1R; also referred to in the art as FMS, FIM2, C-FMS, M-CSF receptor, and CD115) is a single-pass transmembrane receptor with an N-terminal extracellular domain (ECD) and a C-terminal intracellular domain with tyrosine kinase activity.
  • CSF1R Colony stimulating factor 1 receptor
  • CSF1 or the interleukin 34 ligand referred to herein as IL-34; Lin et al., Science 320: 807-11 (2008)
  • CSF1R the interleukin 34 ligand
  • IL-34 the interleukin 34 ligand
  • Both CSF1 and IL-34 stimulate monocyte survival, proliferation, and differentiation into macrophages, as well as other monocytic cell lineages such as osteoclasts, dendritic cells, and microglia.
  • CSF1R tumor-associated macrophages
  • TAMs tumor-associated macrophages
  • CSF 1R plays a role in osteolytic bone destruction in bone metastasis. See, e.g., Ohno et al., Mol. Cancer Ther. 5: 2634-43 (2006).
  • CSF1 and its receptor have also been found to be involved in various inflammatory and autoimmune diseases. See, e.g., Hamilton, Nat. Rev. 8: 533-44 (2008). For example, synovial endothelial cells from joints afflicted with rheumatoid arthritis have been found to produce CSF1, suggesting a role for CSF1 and its receptor in the disease. Blocking CSF1R activity with an antibody results in positive clinical effects in mouse models of arthritis, including a reduction in the destruction of bone and cartilage and a reduction in macrophage numbers. See, e.g., Kitaura et al., J. Clin. Invest. 115: 3418-3427 (2005).
  • Mature differentiated myeloid lineage cells such as macrophages, microglial cells, and osteoclasts contribute to pathology of various diseases such as rheumatoid arthritis, multiple sclerosis and diseases of bone loss.
  • Differentiated myeloid lineage cells are derived from peripheral blood monocyte intermediates.
  • CSF1R stimulation contributes to development of monocytes from bone marrow precursors, to monocyte proliferation and survival, and to differentiation of peripheral blood monocytes into differentiated myeloid lineage cells such as macrophages, microglial cells, and osteoclasts.
  • CSF1R stimulation thus contributes to proliferation, survival, activation, and maturation of differentiated myeloid lineage cells, and in the pathologic setting, CSF1R stimulation contributes to the ability of differentiated myeloid lineage cells to mediate disease pathology.
  • methods of reducing the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in a subject are provided.
  • a method comprises administering an effective amount of an antibody that binds colony stimulating factor 1 receptor (CSF1R) to the subject, wherein the antibody blocks binding of colony stimulating factor 1 (CSF1) to CSF1R and blocks binding of IL-34 to CSF1R.
  • CSF1R colony stimulating factor 1 receptor
  • the subject has an inflammatory condition.
  • the subject has a condition selected from rheumatoid arthritis, juvenile idiopathic arthritis, Castleman's disease, psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, ulcerative colitis, lupus erythematosus, inflammatory bowel disease, inflammatory arthritis, and CD16+ disorders.
  • the method comprises reducing the level of at least one, at least two, at least three, or four factors selected from IL-6, IL-1 ⁇ , TNF- ⁇ , and CXCL10. In some embodiments, the method comprises reducing the level of IL-6. In some such embodiments, the subject has a condition selected from rheumatoid arthritis, juvenile idiopathic arthritis, and Castleman's disease. In some embodiments, the method comprises reducing the level of TNF- ⁇ . In some such embodiments, the subject has a condition selected from rheumatoid arthritis, juvenile idiopathic arthritis, psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, and ulcerative colitis.
  • the method comprises reducing the level of IL-1 ⁇ .
  • the subject has a condition selected from rheumatoid arthritis and juvenile idiopathic arthritis.
  • the method comprises reducing the level of CXCL10.
  • the method comprises reducing the level of at least one, at least two, at least three, or four factors selected from IL-6, IL-1 ⁇ , TNF- ⁇ , and CXCL10.
  • the method comprises reducing the level of IL-6; or the method comprises reducing the level of TNF- ⁇ ; or the method comprises reducing the level of IL-1 ⁇ ; or the method comprises reducing the level of CXCL10; or the method comprises reducing the levels of IL-6 and TNF- ⁇ ; or the method comprises reducing the levels of IL-6 and IL-1 ⁇ ; or the method comprises reducing the levels of IL-6 and CXCL10; or the method comprises reducing the levels of TNF- ⁇ and IL-1 ⁇ ; or the method comprises reducing the levels of TNF- ⁇ and CXCL10; or the method comprises reducing the levels of IL-1 ⁇ and CXCL10; or the method comprises reducing the levels of IL-1 ⁇ and CXCL10; or the method comprises reducing the levels of IL-6, TNF- ⁇ , and IL-1 ⁇ ;
  • methods of treating conditions associated with an elevated level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 are provided.
  • a method comprises administering an effective amount of an antibody that binds colony stimulating factor 1 receptor (CSF1R) to a subject with the condition, wherein the antibody blocks binding of colony stimulating factor 1 (CSF1) to CSF1R and blocks binding of IL-34 to CSF1R.
  • the antibody reduces the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9.
  • the subject has a condition selected from rheumatoid arthritis, juvenile idiopathic arthritis, Castleman's disease, psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, and ulcerative colitis, lupus erythematosus, and inflammatory bowel disease.
  • a condition is associated with an elevated level of at least one, at least two, at least three, or four factors selected from IL-6, IL-1 ⁇ , TNF- ⁇ , and CXCL10.
  • a condition is associated with an elevated level of IL-6.
  • the condition is selected from rheumatoid arthritis, juvenile idiopathic arthritis, and Castleman's disease.
  • a condition is associated with an elevated level of TNF- ⁇ .
  • the condition is selected from rheumatoid arthritis, juvenile idiopathic arthritis, psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, and ulcerative colitis.
  • a condition is associated with an elevated level of IL-1 ⁇ .
  • the condition is selected from rheumatoid arthritis and juvenile idiopathic arthritis.
  • a condition is associated with an elevated level of CXCL10.
  • a method of treating inflammatory arthritis comprises administering an effective amount of an antibody that binds CSF1R to a subject with inflammatory arthritis, wherein the antibody blocks binding of CSF1 to CSF1R and blocks binding of IL-34 to CSF1R, and wherein the antibody reduces the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9.
  • the inflammatory arthritis is selected from rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, and juvenile idiopathic arthritis.
  • a method comprises administering an effective amount of an antibody that binds CSF1R to a subject with an inflammatory condition, wherein the antibody blocks binding of CSF1 to CSF1R and blocks binding of IL-34 to CSF1R, and wherein the antibody reduces the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9.
  • the inflammatory condition is selected from rheumatoid arthritis, juvenile idiopathic arthritis, Castleman's disease, psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, and ulcerative colitis, lupus erythematosus, and inflammatory bowel disease.
  • a method comprises administering an effective amount of an antibody that binds CSF1R to a subject with a CD16+ disorder, wherein the antibody blocks binding of CSF1 to CSF1R and blocks binding of IL-34 to CSF1R, and wherein the antibody reduces the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9.
  • the antibody reduces the level of at least one, at least two, at least three, or four factors selected from IL-6, IL-1 ⁇ , TNF- ⁇ , and CXCL10.
  • the CD16+ disorder is selected from rheumatoid arthritis, juvenile idiopathic arthritis, Castleman's disease, psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, and ulcerative colitis, lupus erythematosus, and inflammatory bowel disease.
  • the antibody substantially reduces the number of CD16+ monocytes. In some embodiments, the number of CD16 ⁇ monocytes are substantially unchanged following administration of the antibody.
  • the antibody may reduce the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in vitro.
  • the subject may have an elevated level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 prior to administration of the antibody.
  • a method further comprises administering at least one additional therapeutic agent selected from methotrexate, an anti-TNF agent, a glucocorticoid, cyclosporine, leflunomide, azathioprine, a JAK inhibitor, a SYK inhibitor, an anti-IL-6 agent, an anti-CD20 agent, an anti-CD19 agent, an anti-GM-CSF agent, an anti-IL-1 agent, and a CTLA4 agent.
  • at least one additional therapeutic agent selected from methotrexate, an anti-TNF agent, a glucocorticoid, cyclosporine, leflunomide, azathioprine, a JAK inhibitor, a SYK inhibitor, an anti-IL-6 agent, an anti-CD20 agent, an anti-CD19 agent, an anti-GM-CSF agent, an anti-IL-1 agent, and a CTLA4 agent.
  • the at least one additional therapeutic agent is selected from methotrexate, an anti-TNF- ⁇ antibody, a soluble TNF receptor, a glucocorticoid, cyclosporine, leflunomide, azathioprine, a JAK inhibitor, a SYK inhibitor, an anti-IL-6 antibody, an anti-IL-6 receptor antibody, an anti-CD20 antibody, an anti-CD19 antibody, an anti-GM-CSF antibody, and anti-GM-CSF receptor antibody, an anti-IL-1 antibody, an IL-1 receptor antagonist, and a CTLA4-Ig fusion molecule.
  • the condition is resistant to methotrexate.
  • a method of treating an inflammatory condition comprises (a) determining the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in a subject with the inflammatory condition; and (b) if the level of at least one of the factors is elevated in the subject, administering to the subject an effective amount of an antibody that binds CSF1R and blocks binding of IL-34 to CSF1R, wherein the antibody reduces the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1
  • a method of treating an inflammatory condition comprises (a) detecting an elevated level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in a subject with the inflammatory condition; and (b) administering to the subject an effective amount of an antibody that binds CSF1R and blocks binding of IL-34 to CSF1R, wherein the antibody reduces the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF
  • the antibody may reduce the level of IL-6; or the antibody may reduce the level of TNF- ⁇ ; or the antibody may reduce the level of IL-1 ⁇ ; or the antibody may reduce the level of CXCL10; or the antibody may reduce the levels of IL-6 and TNF- ⁇ ; or the antibody may reduce the levels of IL-6 and IL-1 ⁇ ; or the antibody may reduce the levels of IL-6 and CXCL10; or the antibody may reduce the levels of TNF- ⁇ and IL-1 ⁇ ; or the antibody may reduce the levels of TNF- ⁇ and CXCL10; or the antibody may reduce the levels of IL-1 ⁇ and CXCL10; or the antibody may reduce the levels of IL-6, TNF- ⁇ , and IL-1 ⁇ ; or the antibody may reduce the levels of IL-6, TNF- ⁇ , and CXCL10; or the antibody may reduce the levels of TNF- ⁇ , IL-1 ⁇ ; or the method comprises reducing the levels of IL-6, IL-1 ⁇ , and CXCL10; or the antibody may reduce the levels of TNF-
  • a method of identifying a subject who may benefit from an antibody that binds CSF1R comprising determining the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in the subject, wherein an elevated level of at least one of the factors in the subject indicates that the subject may benefit from the antibody that binds CSF1R.
  • the subject has a CD16+ disorder.
  • the subject has rheumatoid arthritis.
  • the subject has an elevated level of CD16+ monocytes
  • a method of predicting responsiveness in a subject suffering from an inflammatory condition to an antibody that binds CSF1R comprising determining the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in the subject, wherein an elevated level of at least one of the factors in the subject indicates that the subject is more likely to respond to the antibody that binds CSF1R.
  • the subject has a CD16+ disorder.
  • the subject has
  • a condition may be resistant to methotrexate and/or the subject may be a methotrexate inadequate responder. Further, in any of the embodiments described herein, a condition may be resistant to a TNF inhibitor and/or the subject may be a TNF inhibitor inadequate responder.
  • a method of treating a methotrexate inadequate responder comprising administering to the methotrexate inadequate responder an antibody that binds CSF1R, wherein the antibody blocks binding of CSF1 to CSF1R and blocks binding of IL-34 to CSF1R.
  • a method of treating a TNF inhibitor inadequate responder comprising administering to the TNF inhibitor inadequate responder an antibody that binds CSF1R, wherein the antibody blocks binding of CSF1 to CSF1R and blocks binding of IL-34 to CSF1R.
  • the inadequate responder has a CD16+ disorder.
  • the CD16+ disorder is rheumatoid arthritis.
  • the antibody substantially reduces the number of CD16+ monocytes. In some embodiments, the number of CD16 ⁇ monocytes are substantially unchanged following administration of the antibody. In some embodiments, the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in the methotrexate and/or TNF-inhibitor inadequate responder is reduced following administration of the antibody.
  • the antibody heavy chain and/or the antibody light chain may have the structure described below.
  • the antibody heavy chain may comprise a sequence that is at least 90%, at least 95%, at least 97%, at least 99%, or 100% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45.
  • the antibody light chain may comprise a sequence that is at least 90%, at least 95%, at least 97%, at least 99%, or 100% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52.
  • the antibody heavy chain may comprise a sequence that is at least 90%, at least 95%, at least 97%, at least 99%, or 100% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45
  • the antibody light chain may comprise a sequence that is at least 90%, at least 95%, at least 97%, at least 99%, or 100% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52.
  • the HC CDR1, HC CDR2, and HC CDR3 may comprise a set of sequences selected from: (a) SEQ ID NOs: 15, 16, and 17; (b) SEQ ID NOs: 21, 22, and 23; and (c) SEQ ID NOs: 27, 28, and 29.
  • the LC CDR1, LC CDR2, and LC CDR3 may comprise a set of sequences selected from: (a) SEQ ID NOs: 18, 19, and 20; (b) SEQ ID NOs: 24, 25, and 26; and (c) SEQ ID NOs: 30, 31, and 32.
  • the heavy chain may comprise an HC CDR1, HC CDR2, and HC CDR3, wherein the HC CDR1, HC CDR2, and HC CDR3 comprise a set of sequences selected from: (a) SEQ ID NOs: 15, 16, and 17; (b) SEQ ID NOs: 21, 22, and 23; and (c) SEQ ID NOs: 27, 28, and 29; and the light chain may comprise an LC CDR1, LC CDR2, and LC CDR3, wherein the LC CDR1, LC CDR2, and LC CDR3 comprise a set of sequences selected from: (a) SEQ ID NOs: 18, 19, and 20; (b) SEQ ID NOs: 24, 25, and 26; and (c) SEQ ID NOs: 30, 31, and 32.
  • the antibody that binds CSF1R may comprise: (a) a heavy chain comprising a sequence that is at least 95%, at least 97%, at least 99%, or 100% identical to SEQ ID NO: 9 and a light chain comprising a sequence that is at least 95%, at least 97%, at least 99%, or 100% identical to SEQ ID NO: 10; (b) a heavy chain comprising a sequence that is at least 95%, at least 97%, at least 99%, or 100% identical to SEQ ID NO: 11 and a light chain comprising a sequence that is at least 95%, at least 97%, at least 99%, or 100% identical to SEQ ID NO: 12; (c) a heavy chain comprising a sequence that is at least 95%, at least 97%, at least 99%, or 100% identical to SEQ ID NO: 13 and a light chain comprising a sequence that is at least 95%, at least 97%, at least 99%, or 100% identical to SEQ ID NO: 14; (d) a heavy chain comprising
  • the antibody may comprise: (a) a heavy chain comprising a heavy chain (HC) CDR1 having the sequence of SEQ ID NO: 15, an HC CDR2 having the sequence of SEQ ID NO: 16, and an HC CDR3 having the sequence of SEQ ID NO: 17, and a light chain comprising a light chain (LC) CDR1 having the sequence of SEQ ID NO: 18, a LC CDR2 having the sequence of SEQ ID NO: 19, and a LC CDR3 having the sequence of SEQ ID NO: 20; (b) a heavy chain comprising a heavy chain (HC) CDR1 having the sequence of SEQ ID NO: 21, an HC CDR2 having the sequence of SEQ ID NO: 22, and an HC CDR3 having the sequence of SEQ ID NO: 23, and a light chain comprising a light chain (LC) CDR1 having the sequence of SEQ ID NO: 24, a LC CDR2 having the sequence of SEQ ID NO: 25, and a LC CDR3 having the
  • the antibody may comprise: (a) a heavy chain comprising a sequence of SEQ ID NO: 53 and a light chain comprising a sequence of SEQ ID NO: 60; (b) a heavy chain comprising a sequence of SEQ ID NO: 53 and a light chain comprising a sequence of SEQ ID NO: 61; or (c) a heavy chain comprising a sequence of SEQ ID NO: 58 and a light chain comprising a sequence of SEQ ID NO: 65.
  • an antibody comprises a heavy chain and a light chain, wherein the antibody comprises: (a) a heavy chain consisting of the sequence of SEQ ID NO: 53 and a light chain consisting of the sequence of SEQ ID NO: 60; (b) a heavy chain consisting of the sequence of SEQ ID NO: 53 and a light chain consisting of the sequence of SEQ ID NO: 61; or (c) a heavy chain consisting of the sequence of SEQ ID NO: 58 and a light chain consisting of the sequence of SEQ ID NO: 65.
  • the antibody may be a humanized antibody. In any of the methods described herein, the antibody may be selected from a Fab, an Fv, an scFv, a Fab′, and a (Fab′) 2 . In any of the methods described herein, the antibody may be a chimeric antibody. In any of the methods described herein, the antibody may be selected from an IgA, an IgG, and an IgD. In any of the methods described herein, the antibody may be an IgG. In any of the methods described herein, the antibody may be an IgG4. In any of the methods described herein, the antibody may be an IgG4 comprising an S241P mutation in at least one IgG4 heavy chain constant region.
  • the antibody may bind to human CSF1R and/or binds to cynomolgus CSF1R. In any of the methods described herein, the antibody may block ligand binding to CSF1R. In any of the methods described herein, the antibody may block binding of CSF1 and/or IL-34 to CSF1R. In any of the methods described herein, the antibody may block binding of both CSF1 and IL-34 to CSF1R. In any of the methods described herein, the antibody may inhibit ligand-induced CSF1R phosphorylation. In any of the methods described herein, the antibody may inhibit CSF1- and/or IL-34-induced CSF1R phosphorylation.
  • the antibody may bind to human CSF1R with an affinity (K D ) of less than 1 nM. In any of the methods described herein, the antibody may inhibit monocyte proliferation and/or survival responses in the presence of CSF1 or IL-34.
  • a pharmaceutical composition comprising an antibody that binds CSF1R is provided.
  • antibodies that bind CSF1R and compositions comprising antibodies that bind CSF1R are provided for use in any of the methods of treatment described herein.
  • FIG. 1A-C show an alignment of the humanized heavy chain variable regions for each of humanized antibodies huAb1 to huAb16, as discussed in Example 1. Boxed residues are amino acids in the human acceptor sequence that were changed back to the corresponding mouse residue.
  • FIG. 2A-C show an alignment of the humanized light chain variable regions for each of humanized antibodies huAb1 to huAb16, as discussed in Example 1. Boxed amino acids are residues in the human acceptor sequence that were changed back to the corresponding mouse residue.
  • Methods of reducing the level of one or more factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in a subject comprising administering antibodies that bind CSF1R and block CSF1 and IL-34 ligand binding are provided.
  • antibodies that bind CSF1R and block CSF1 and IL-34 ligand binding are effective for reducing the levels of one or more factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 and treating conditions associated with elevated levels of those factors.
  • Exemplary such conditions include, but are not limited to, rheumatoid arthritis, juvenile idiopathic arthritis, Castleman's disease, psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, ulcerative colitis, lupus erythematosus, and inflammatory bowel disease.
  • the present inventors found that contacting synovial biopsy samples from rheumatoid arthritis patients with an antibody that binds CSF1R reduces the levels of IL-6, IL-1 ⁇ , IL-8, CCL2 (also referred to as MCP-1), CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9.
  • Exemplary techniques used in connection with recombinant DNA, oligonucleotide synthesis, tissue culture and transformation (e.g., electroporation, lipofection), enzymatic reactions, and purification techniques are known in the art. Many such techniques and procedures are described, e.g., in Sambrook et al. Molecular Cloning: A Laboratory Manual (2nd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), among other places.
  • exemplary techniques for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients are also known in the art.
  • nucleic acid molecule and “polynucleotide” may be used interchangeably, and refer to a polymer of nucleotides. Such polymers of nucleotides may contain natural and/or non-natural nucleotides, and include, but are not limited to, DNA, RNA, and PNA.
  • Nucleic acid sequence refers to the linear sequence of nucleotides that comprise the nucleic acid molecule or polynucleotide.
  • polypeptide and “protein” are used interchangeably to refer to a polymer of amino acid residues, and are not limited to a minimum length. Such polymers of amino acid residues may contain natural or non-natural amino acid residues, and include, but are not limited to, peptides, oligopeptides, dimers, trimers, and multimers of amino acid residues. Both full-length proteins and fragments thereof are encompassed by the definition.
  • the terms also include post-expression modifications of the polypeptide, for example, glycosylation, sialylation, acetylation, phosphorylation, and the like.
  • a “polypeptide” refers to a protein which includes modifications, such as deletions, additions, and substitutions (generally conservative in nature), to the native sequence, as long as the protein maintains the desired activity. These modifications may be deliberate, as through site-directed mutagenesis, or may be accidental, such as through mutations of hosts which produce the proteins or errors due to PCR amplification.
  • CSF1R refers herein to the full-length CSF1R, which includes the N-terminal ECD, the transmembrane domain, and the intracellular tyrosine kinase domain, with or without an N-terminal leader sequence.
  • the CSF1R is a human CSF1R having the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2.
  • CSF1R extracellular domain refers to a CSF1R polypeptide that lacks the intracellular and transmembrane domains.
  • CSF1R ECDs include the full-length CSF1R ECD and CSF1R ECD fragments that are capable of binding CSF1 and/or IL-34.
  • the human full-length CSF1R ECD is defined herein as comprising either amino acids 1 to 512 (i.e., including the leader sequence) or amino acids 20 to 512 (i.e., lacking the leader sequence) of SEQ ID NO: 2.
  • a human CSF1R ECD fragment comprises amino acids 20 to 506 of SEQ ID NO: 2 (see SEQ ID NO: 5).
  • a human CSF1R fragment ends at amino acid 507, 508, 509, 510, or 511.
  • a cyno CSF1R ECD comprises the sequence of SEQ ID NO: 7 (with leader sequence) or amino acids 20 to 506 of SEQ ID NO: 7 (without leader sequence).
  • a ligand such as CSF1 and/or IL-34
  • grammatical variants thereof are used to refer to the ability to inhibit the interaction between CSF1R and a CSF1R ligand, such as CSF1 and/or IL-34. Such inhibition may occur through any mechanism, including direct interference with ligand binding, e.g., because of overlapping binding sites on CSF1R, and/or conformational changes in CSF1R induced by the antibody that alter ligand affinity, etc.
  • Antibodies and antibody fragments referred to as “functional” are characterized by having such properties.
  • an “immunological” activity refers only to the ability to induce the production of an antibody against an antigenic epitope possessed by a native or naturally-occurring CSF1R polypeptide.
  • antibody refers to a molecule comprising at least complementarity-determining region (CDR) 1, CDR2, and CDR3 of a heavy chain and at least CDR1, CDR2, and CDR3 of a light chain, wherein the molecule is capable of binding to antigen.
  • CDR complementarity-determining region
  • the term antibody includes, but is not limited to, fragments that are capable of binding antigen, such as Fv, single-chain Fv (scFv), Fab, Fab′, and (Fab′) 2 .
  • the term antibody also includes, but is not limited to, chimeric antibodies, humanized antibodies, and antibodies of various species such as mouse, human, cynomolgus monkey, etc.
  • an antibody comprises a heavy chain variable region and a light chain variable region. In some embodiments, an antibody comprises at least one heavy chain comprising a heavy chain variable region and at least a portion of a heavy chain constant region, and at least one light chain comprising a light chain variable region and at least a portion of a light chain constant region. In some embodiments, an antibody comprises two heavy chains, wherein each heavy chain comprises a heavy chain variable region and at least a portion of a heavy chain constant region, and two light chains, wherein each light chain comprises a light chain variable region and at least a portion of a light chain constant region.
  • a single-chain Fv or any other antibody that comprises, for example, a single polypeptide chain comprising all six CDRs (three heavy chain CDRs and three light chain CDRs) is considered to have a heavy chain and a light chain.
  • the heavy chain is the region of the antibody that comprises the three heavy chain CDRs and the light chain in the region of the antibody that comprises the three light chain CDRs.
  • heavy chain variable region refers to a region comprising heavy chain CDR1, framework (FR) 2, CDR2, FR3, and CDR3.
  • a heavy chain variable region also comprises at least a portion of an FR1 and/or at least a portion of an FR4.
  • a heavy chain CDR1 corresponds to Kabat residues 26 to 35;
  • a heavy chain CDR2 corresponds to Kabat residues 50 to 65;
  • a heavy chain CDR3 corresponds to Kabat residues 95 to 102. See, e.g., Kabat Sequences of Proteins of Immunological Interest (1987 and 1991, NIH, Bethesda, Md.); and FIG. 1 .
  • heavy chain constant region refers to a region comprising at least three heavy chain constant domains, C H 1, C H 2, and C H 3.
  • Nonlimiting exemplary heavy chain constant regions include ⁇ , ⁇ , and ⁇ .
  • Nonlimiting exemplary heavy chain constant regions also include ⁇ and ⁇ .
  • Each heavy constant region corresponds to an antibody isotype.
  • an antibody comprising a ⁇ constant region is an IgG antibody
  • an antibody comprising a ⁇ constant region is an IgD antibody
  • an antibody comprising an ⁇ constant region is an IgA antibody.
  • an antibody comprising a ⁇ constant region is an IgM antibody
  • an antibody comprising an ⁇ constant region is an IgE antibody.
  • IgG antibodies include, but are not limited to, IgG1 (comprising a ⁇ 1 constant region), IgG2 (comprising a ⁇ 2 constant region), IgG3 (comprising a ⁇ 3 constant region), and IgG4 (comprising a ⁇ 4 constant region) antibodies;
  • IgA antibodies include, but are not limited to, IgA1 (comprising an ⁇ 1 constant region) and IgA2 (comprising an ⁇ 2 constant region) antibodies;
  • IgM antibodies include, but are not limited to, IgM1 and IgM2.
  • a heavy chain constant region comprises one or more mutations (or substitutions), additions, or deletions that confer a desired characteristic on the antibody.
  • a nonlimiting exemplary mutation is the S241P mutation in the IgG4 hinge region (between constant domains C H 1 and C H 2), which alters the IgG4 motif CPSCP to CPPCP, which is similar to the corresponding motif in IgG1. That mutation, in some embodiments, results in a more stable IgG4 antibody. See, e.g., Angal et al., Mol. Immunol. 30: 105-108 (1993); Bloom et al., Prot. Sci. 6: 407-415 (1997); Schuurman et al., Mol. Immunol. 38: 1-8 (2001).
  • heavy chain refers to a polypeptide comprising at least a heavy chain variable region, with or without a leader sequence.
  • a heavy chain comprises at least a portion of a heavy chain constant region.
  • full-length heavy chain refers to a polypeptide comprising a heavy chain variable region and a heavy chain constant region, with or without a leader sequence.
  • light chain variable region refers to a region comprising light chain CDR1, framework (FR) 2, CDR2, FR3, and CDR3.
  • a light chain variable region also comprises an FR1 and/or an FR4.
  • a light chain CDR1 corresponds to Kabat residues 24 to 34
  • a light chain CDR2 corresponds to Kabat residues 50 to 56
  • a light chain CDR3 corresponds to Kabat residues 89 to 97. See, e.g., Kabat Sequences of Proteins of Immunological Interest (1987 and 1991, NIH, Bethesda, Md.); and FIG. 1 .
  • light chain constant region refers to a region comprising a light chain constant domain, C L .
  • Nonlimiting exemplary light chain constant regions include ⁇ and ⁇ .
  • light chain refers to a polypeptide comprising at least a light chain variable region, with or without a leader sequence.
  • a light chain comprises at least a portion of a light chain constant region.
  • full-length light chain refers to a polypeptide comprising a light chain variable region and a light chain constant region, with or without a leader sequence.
  • a “chimeric antibody” as used herein refers to an antibody comprising at least one variable region from a first species (such as mouse, rat, cynomolgus monkey, etc.) and at least one constant region from a second species (such as human, cynomolgus monkey, etc.).
  • a chimeric antibody comprises at least one mouse variable region and at least one human constant region.
  • a chimeric antibody comprises at least one cynomolgus variable region and at least one human constant region.
  • a chimeric antibody comprises at least one rat variable region and at least one mouse constant region.
  • all of the variable regions of a chimeric antibody are from a first species and all of the constant regions of the chimeric antibody are from a second species.
  • a “humanized antibody” as used herein refers to an antibody in which at least one amino acid in a framework region of a non-human variable region has been replaced with the corresponding amino acid from a human variable region.
  • a humanized antibody comprises at least one human constant region or fragment thereof.
  • a humanized antibody is a Fab, an scFv, a (Fab′) 2 , etc.
  • CDR-grafted antibody refers to a humanized antibody in which the complementarity determining regions (CDRs) of a first (non-human) species have been grafted onto the framework regions (FRs) of a second (human) species.
  • a “human antibody” as used herein refers to antibodies produced in humans, antibodies produced in non-human animals that comprise human immunoglobulin genes, such as XenoMouse®, and antibodies selected using in vitro methods, such as phage display, wherein the antibody repertoire is based on a human immunoglobulin sequences.
  • leader sequence refers to a sequence of amino acid residues located at the N terminus of a polypeptide that facilitates secretion of a polypeptide from a mammalian cell.
  • a leader sequence may be cleaved upon export of the polypeptide from the mammalian cell, forming a mature protein.
  • Leader sequences may be natural or synthetic, and they may be heterologous or homologous to the protein to which they are attached.
  • Exemplary leader sequences include, but are not limited to, antibody leader sequences, such as, for example, the amino acid sequences of SEQ ID NOs: 3 and 4, which correspond to human light and heavy chain leader sequences, respectively.
  • Nonlimiting exemplary leader sequences also include leader sequences from heterologous proteins.
  • an antibody lacks a leader sequence.
  • an antibody comprises at least one leader sequence, which may be selected from native antibody leader sequences and heterologous leader sequences.
  • vector is used to describe a polynucleotide that may be engineered to contain a cloned polynucleotide or polynucleotides that may be propagated in a host cell.
  • a vector may include one or more of the following elements: an origin of replication, one or more regulatory sequences (such as, for example, promoters and/or enhancers) that regulate the expression of the polypeptide of interest, and/or one or more selectable marker genes (such as, for example, antibiotic resistance genes and genes that may be used in colorimetric assays, e.g., ⁇ -galactosidase).
  • expression vector refers to a vector that is used to express a polypeptide of interest in a host cell.
  • a “host cell” refers to a cell that may be or has been a recipient of a vector or isolated polynucleotide.
  • Host cells may be prokaryotic cells or eukaryotic cells.
  • Exemplary eukaryotic cells include mammalian cells, such as primate or non-primate animal cells; fungal cells, such as yeast; plant cells; and insect cells.
  • Nonlimiting exemplary mammalian cells include, but are not limited to, NSO cells, PER.C6® cells (Crucell), and 293 and CHO cells, and their derivatives, such as 293-6E and DG44 cells, respectively.
  • isolated refers to a molecule that has been separated from at least some of the components with which it is typically found in nature.
  • a polypeptide is referred to as “isolated” when it is separated from at least some of the components of the cell in which it was produced.
  • a polypeptide is secreted by a cell after expression, physically separating the supernatant containing the polypeptide from the cell that produced it is considered to be “isolating” the polypeptide.
  • a polynucleotide is referred to as “isolated” when it is not part of the larger polynucleotide (such as, for example, genomic DNA or mitochondrial DNA, in the case of a DNA polynucleotide) in which it is typically found in nature, or is separated from at least some of the components of the cell in which it was produced, e.g., in the case of an RNA polynucleotide.
  • a DNA polynucleotide that is contained in a vector inside a host cell may be referred to as “isolated” so long as that polynucleotide is not found in that vector in nature.
  • elevated level means a higher level of a protein, such as a cytokine or matrix metalloproteinase, in a particular tissue of a subject relative to the same tissue in a control, such as an individual or individuals who are not suffering from an inflammatory condition or other condition described herein.
  • the elevated level may be the result of any mechanism, such as increased expression, increased stability, decreased degradation, increased secretion, decreased clearance, etc., of the protein.
  • the term “reduce” or “reduces” means to lower the level of a protein, such as a cytokine or matrix metalloproteinase, in a particular tissue of a subject by at least 10%.
  • an agent such as an antibody that binds CSF1R
  • the level of a protein is reduced relative to the level of the protein prior to contacting with an agent, such as an antibody that binds CSF1R.
  • resistant when used in the context of resistance to a therapeutic agent, means a decreased response or lack of response to a standard dose of the therapeutic agent, relative to the subject's response to the standard dose of the therapeutic agent in the past, or relative to the expected response of a similar subject with a similar disorder to the standard dose of the therapeutic agent.
  • a subject may be resistant to therapeutic agent although the subject has not previously been given the therapeutic agent, or the subject may develop resistance to the therapeutic agent after having responded to the agent on one or more previous occasions.
  • subject and “patient” are used interchangeably herein to refer to a human.
  • methods of treating other mammals including, but not limited to, rodents, simians, felines, canines, equines, bovines, porcines, ovines, caprines, mammalian laboratory animals, mammalian farm animals, mammalian sport animals, and mammalian pets, are also provided.
  • sample refers to a composition that is obtained or derived from a subject that contains a cellular and/or other molecular entity that is to be characterized, quantitated, and/or identified, for example based on physical, biochemical, chemical and/or physiological characteristics.
  • An exemplary sample is a tissue sample.
  • tissue sample refers to a collection of similar cells obtained from a tissue of a subject.
  • the source of the tissue sample may be solid tissue as from a fresh, frozen and/or preserved organ or tissue sample or biopsy or aspirate; blood or any blood constituents; bodily fluids such as cerebral spinal fluid, amniotic fluid, peritoneal fluid, synovial fluid, or interstitial fluid; cells from any time in gestation or development of the subject.
  • a tissue sample is a synovial biopsy tissue sample and/or a synovial fluid sample.
  • a tissue sample is a synovial fluid sample.
  • the tissue sample may also be primary or cultured cells or cell lines.
  • the tissue sample is obtained from a disease tissue/organ.
  • the tissue sample may contain compounds that are not naturally intermixed with the tissue in nature such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics, or the like.
  • a “control sample” or “control tissue”, as used herein, refers to a sample, cell, or tissue obtained from a source known, or believed, not to be afflicted with the disease for which the subject is being treated.
  • a “section” of a tissue sample means a part or piece of a tissue sample, such as a thin slice of tissue or cells cut from a solid tissue sample.
  • rheumatoid arthritis refers to a recognized disease state that may be diagnosed according to the 1987, 2000, or 2010 criteria for the classification of RA (American Rheumatism Association or Americal College of Rheumatology), or any similar criteria.
  • rheumatoid arthritis refers to a chronic autoimmune disease characterized primarily by inflammation of the lining (synovium) of the joints, which can lead to joint damage, resulting in chronic pain, loss of function, and disability. Because RA can affect multiple organs of the body, including skin, lungs, and eyes, it is referred to as a systemic illness.
  • rheumatoid arthritis includes not only active and early RA, but also incipient RA, as defined below.
  • Physiological indicators of RA include, symmetric joint swelling which is characteristic though not invariable in RA. Fusiform swelling of the proximal interphalangeal (PIP) joints of the hands as well as metacarpophalangeal (MCP), wrists, elbows, knees, ankles, and metatarsophalangeal (MTP) joints are commonly affected and swelling is easily detected. Pain on passive motion is the most sensitive test for joint inflammation, and inflammation and structural deformity often limits the range of motion for the affected joint.
  • Typical visible changes include ulnar deviation of the fingers at the MCP joints, hyperextension, or hyperflexion of the MCP and PIP joints, flexion contractures of the elbows, and subluxation of the carpal bones and toes.
  • the subject with RA may be resistant to a disease-modifying anti-rheumatic drug (DMARD), and/or a non-steroidal anti-inflammatory drug (NSAID).
  • DMARD disease-modifying anti-rheumatic drug
  • NSAID non-steroidal anti-inflammatory drug
  • Nonlimiting exemplary “DMARDs” include hydroxychloroquine, sulfasalazine, methotrexate (MTX), leflunomide, etanercept, infliximab (plus oral and subcutaneous MTX), azathioprine, D-penicillamine, gold salts (oral), gold salts (intramuscular), minocycline, cyclosporine including cyclosporine A and topical cyclosporine, staphylococcal protein A (Goodyear and Silverman, J. Exp. Med., 197(9):1125-1139 (2003)), including salts and derivatives thereof, etc.
  • TNF inhibitors such as etanercept, infliximab, golimumab, certolizumab, and/or adalimumab because of toxicity, inadequate efficacy, and/or resistance.
  • a patient with “active rheumatoid arthritis” means a patient with active and not latent symptoms of RA.
  • Subjects with “early active rheumatoid arthritis” are those subjects with active RA diagnosed for at least 8 weeks but no longer than four years, according to the revised 1987, 2000, or 2010 criteria for the classification of RA (American Rheumatism Association or Americal College of Rheumatology).
  • Subjects with “early rheumatoid arthritis” are those subjects with RA diagnosed for at least eight weeks but no longer than four years, according to the revised 1987, 2000, or 2010 criteria for classification of RA (American Rheumatism Association or Americal College of Rheumatology).
  • RA includes, for example, juvenile-onset RA, juvenile idiopathic arthritis (JIA), or juvenile RA (JRA).
  • Patients with “incipient RA” have early polyarthritis that does not fully meet ACR criteria for a diagnosis of RA, in association with the presence of RA-specific prognostic biomarkers such as anti-CCP and shared epitope. They include patients with positive anti-CCP antibodies who present with polyarthritis, but do not yet have a diagnosis of RA, and are at high risk for going on to develop bona fide ACR criteria RA (95% probability).
  • inflammatory arthritis encompasses any arthritis caused by an autoimmune condition.
  • Nonlimiting examples of inflammatory arthritis and autoimmune conditions that may involve inflammatory arthritis include rheumatoid arthritis (including juvenile-onset RA, juvenile idiopathic arthritis (JIA), and juvenile rheumatoid arthritis (JRA)), ankylosing spondylitis, mixed connective tissue disease (MCTD), psoriatic arthritis, reactive arthritis, scleroderma, Still's disease, systemic lupus erythematosus, acute and chronic arthritis, rheumatoid synovitis, gout or gouty arthritis, acute immunological arthritis, chronic inflammatory arthritis, degenerative arthritis, type II collagen-induced arthritis, infectious arthritis, septic arthritis, Lyme arthritis, proliferative arthritis, vertebral arthritis, osteoarthritis, arthritis chronica progrediente, arthritis deformans, polyarthritis chronica primaria, reactive arthritis, menopausal arthritis, estrogen-depletion arthritis,
  • “Joint damage” is used in the broadest sense and refers to damage or partial or complete destruction to any part of one or more joints, including the connective tissue and cartilage, where damage includes structural and/or functional damage of any cause, and may or may not cause joint pain/arthalgia. It includes, without limitation, joint damage associated with or resulting from inflammatory joint disease as well as non-inflammatory joint disease. This damage may be caused by any condition, such as an autoimmune disease, especially inflammatory arthritis, and most especially rheumatoid arthritis.
  • joints are points of contact between elements of a skeleton (of a vertebrate such as an animal) with the parts that surround and support it and include, but are not limited to, for example, hips, joints between the vertebrae of the spine, joints between the spine and pelvis (sacroiliac joints), joints where the tendons and ligaments attach to bones, joints between the ribs and spine, shoulders, knees, feet, elbows, hands, fingers, ankles and toes, but especially joints in the hands and feet.
  • lupus as used herein is an autoimmune disease or disorder that in general involves antibodies that attack connective tissue.
  • the principal form of lupus is a systemic one, systemic lupus erythematosus (SLE), including cutaneous SLE and subacutecutaneous SLE, as well as other types of lupus (including nephritis, extrarenal, cerebritis, pediatric, non-renal, discoid, and alopecia).
  • systemic lupus erythematosus refers to a chronic autoimmune disease that can result in skin lesions, joint pain and swelling, kidney disease (lupus nephritis), fluid around the heart and/or lungs, inflammation of the heart, and various other systemic conditions.
  • the term “lupus nephritis” refers to inflammation of the kidneys that occurs in patients with SLE. Lupus nephritis may include, for example, glomerulonephritis and/or interstitial nephritis, and can lead to hypertension, proteinuria, and kidney failure.
  • Lupus nephritis may be classified based on severity and extent of disease, for example, as defined by the International Society of Nephrology/Renal/Pathology Society.
  • Lupus nephritis classes include class I (minimal mesangial lupus nephritis), class II (mesangial proliferative lupus nephritis), class III (focal lupus nephritis), class IV (diffuse segmental (IV-S) or diffuse global (IV-G) lupus nephritis), class V (membranous lupus nephritis), and class VI (advanced sclerosing lupus nephritis).
  • the term “lupus nephritis” encompasses all of the classes.
  • MS multiple sclerosis
  • PPMS primary progressive multiple sclerosis
  • RRMS relapsing-remitting multiple sclerosis
  • SPMS secondary progressive multiple sclerosis
  • PRMS progressive relapsing multiple sclerosis
  • PPMS Primary progressive multiple sclerosis
  • PPMS is the sub-type of MS that is least likely to show inflammatory (gadolinium enhancing) lesions on MRI scans.
  • the primary progressive form of the disease affects between 10 and 15% of all people with multiple sclerosis.
  • PPMS may be defined according to the criteria in McDonald et al. Ann Neurol 50:121-7 (2001).
  • the subject with PPMS treated herein is usually one with a probable or definitive diagnosis of PPMS.
  • RRMS Relapsing-remitting multiple sclerosis
  • RRMS Relapsing-remitting multiple sclerosis
  • RRMS is characterized by relapses (also known as exacerbations) during which time new symptoms can appear and old ones resurface or worsen. The relapses are followed by periods of remission, during which time the person fully or partially recovers from the deficits acquired during the relapse. Relapses can last for days, weeks, or months, and recovery can be slow and gradual or almost instantaneous.
  • the vast majority of people presenting with MS are first diagnosed with RRMS. This is typically when they are in their twenties or thirties, though diagnoses much earlier or later are known. Twice as many women as men present with this sub-type of MS.
  • myelin a protective insulating sheath around the nerve fibers (neurons) in the white matter regions of the central nervous system (CNS)
  • CNS central nervous system
  • an oligodendrocyte a special type of glial cell in the CNS
  • sponsors remyelination a process whereby the myelin sheath around the axon may be repaired. It is this remyelination that may be responsible for the remission.
  • Approximately 50% of patients with RRMS convert to SPMS within 10 years of disease onset. After 30 years, this figure rises to 90%.
  • the relapsing-remitting form of the disease accounts around 55% of all people with MS.
  • SPMS Secondary progressive multiple sclerosis
  • PRMS Progressive relapsing multiple sclerosis
  • CD16+ disorder means a disease in which CD16+ monocytes of a mammal cause, mediate or otherwise contribute to morbidity in the mammal. Also included are diseases in which reduction of CD16+ monocytes has an ameliorative effect on progression of the disease. Included within this term are CD16+ inflammatory diseases, infectious diseases, immunodeficiency diseases, neoplasia, etc. In some embodiments, CD16+ inflammatory diseases include inflammatory diseases that are not responsive to methotrexate therapy.
  • CD16+ inflammatory diseases include methotrexate-resistant rheumatoid arthritis, methotrexate-resistant multiple sclerosis, methotrexate-resistant lupus, methotrexate-resistant inflammatory bowel disease, methotrexate-resistant Crohn's disease, methotrexate-resistant asthma, and methotrexate-resistant psoriasis.
  • patients having methotrexate-resistant diseases, such as methotrexate-resistant rheumatoid arthritis are referred to as methotrexate incomplete responders or methotrexate inadequate responders.
  • a subject with a CD16+ disorder is a methotrexate inadequate responder.
  • TNF inhibitor-resistant diseases such as TNF inhibitor-resistant rheumatoid arthritis
  • TNF inhibitor incomplete responders or TNF inhibitor inadequate responders
  • a subject with a CD16+ disorder is a TNF inhibitor inadequate responder.
  • CD16+ disorders examples include, but are not limited to, systemic lupus erythematosus, lupus nephritis, rheumatoid arthritis, juvenile chronic arthritis, juvenile idiopathic arthritis (JIA) (including systemic JIA and polyarticular course JIA), psoriatic arthritis, polymyalgia rheumatic, osteoarthritis, adult-onset Still's disease, spondyloarthropathies, ankylosing spondylitis, systemic sclerosis (scleroderma), idiopathic inflammatory myopathies (dermatomyositis, polymyositis), Sjogren's syndrome, systemic vasculitis, sarcoidosis, autoimmune hemolytic anemia (immune pancytopenia, paroxysmal nocturnal hemoglobinuria), autoimmune thrombocytopenia (idiopathic thrombocytopenic purpura, immune
  • JIA juvenile chronic arthritis
  • panuveitis anterior aveitis, intermediate uveitis, scleritis, keratitis, orbital inflammation, optic neuritis, dry eye, diabetic retinopathy, proliferative vitreoretinopathy, postoperative inflammation), muscle atrophy, demyelinating diseases of the central and peripheral nervous systems such as multiple sclerosis, idiopathic demyelinating polyneuropathy or Guillain-Barre syndrome, and chronic inflammatory demyelinating polyneuropathy, hepatobiliary diseases such as infectious hepatitis (hepatitis A, B, C, D, E and other non-hepatotropic viruses), autoimmune chronic active hepatitis, primary biliary cirrhosis, granulomatous hepatitis, and sclerosing cholangitis, inflammatory bowel disease (IBD), including ulcerative colitis, Crohn's disease, gluten-sensitive enteropathy, and Whipple's disease, pancreatitis, islet
  • methotrexate inadequate responder refers to a subject who has experienced, or is experiencing, an inadequate response to methotrexate treatment, for example, because of toxicity and/or inadequate efficacy at standard doses.
  • a methotrexate inadequate responder has experienced, or is experiencing, an inadequate response to methotrexate after receiving a standard dose for at least two weeks, at least three weeks, at least four weeks, at least six weeks, or at least twelve weeks.
  • TNF inhibitor inadequate responder refers to a subject who has experienced, or is experiencing, an inadequate response to a TNF inhibitor, for example, because of toxicity and/or inadequate efficacy at standard doses.
  • a TNF inhibitor inadequate responder has experienced, or is experiencing, an inadequate response to a TNF inhibitor after receiving a standard dose for at least two weeks, at least three weeks, at least four weeks, at least six weeks, or at least twelve weeks.
  • a TNF inhibitor inadequate responder has experienced, or is experiencing, an inadequate response to a TNF inhibitor selected from infliximab, adalimumab, certolizumab pegol, golimumab, and etanercept.
  • the TNF inhibitor is a TNF- ⁇ inhibitor.
  • substantially reduced and “substantially decreased” and grammatical variants thereof, when used to refer to the level of a protein or cell type, such as CD16+ monocytes, denote a sufficiently high degree of difference between the levels being compared, e.g., as indicated by numeric values, such that one of skill in the art would consider the difference between the levels to be of biological and/or statistical significance.
  • an “anti-[factor] agent” or a “[factor] inhibitor” as used herein refer to an agent that antagonizes the factor activity, such as by binding to the factor or a receptor for the factor (if any), or by specifically inhibiting expression of the factor or a receptor for the factor (if any).
  • exemplary anti-[factor] agents include, but are not limited to, anti-[factor] antibodies, anti-[factor] receptor antibodies, soluble [factor] receptors that bind to the factor, small molecules that bind the [factor] or [factor] receptor, antisense oligonucleotides that are complementary to [factor] or [factor] receptor pre-mRNA or mRNA, etc.
  • Nonlimiting exemplary factors include TNF- ⁇ , IL-1, IL-6, CD20, CD19, and GM-CSF.
  • An agent “antagonizes” factor activity when the agent neutralizes, blocks, inhibits, abrogates, reduces, and/or interferes with the activity of the factor, including its binding to one or more receptors when the factor is a ligand.
  • Treatment refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) the targeted pathologic condition or disorder.
  • treatment covers any administration or application of a therapeutic for disease in a mammal, including a human, and includes inhibiting or slowing the disease or progression of the disease; partially or fully relieving the disease, for example, by causing regression, or restoring or repairing a lost, missing, or defective function; stimulating an inefficient process; or causing the disease plateau to have reduced severity.
  • treatment also includes reducing the severity of any phenotypic characteristic and/or reducing the incidence, degree, or likelihood of that characteristic. Those in need of treatment include those already with the disorder as well as those prone to have the disorder or those in whom the disorder is to be prevented.
  • Chronic administration refers to administration of an agent in a continuous mode as opposed to an acute mode, so as to maintain the initial therapeutic effect (activity) for an extended period of time.
  • Intermittent administration is treatment that is not consecutively done without interruption, but rather is cyclic in nature.
  • an effective amount refers to an amount of a drug effective to treat a disease or disorder in a subject.
  • an effective amount refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • a therapeutically effective amount of an anti-CSF1R antibody of the invention may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the anti-CSF1R antibody to elicit a desired response in the individual.
  • a therapeutically effective amount encompasses an amount in which any toxic or detrimental effects of the anti-CSF1R antibody are outweighed by the therapeutically beneficial effects.
  • the expression “effective amount” refers to an amount of the antibody that is effective for treating the CD16+ disorder.
  • the disorder is RA
  • such effective amount can result in one or more of: reducing the signs or symptoms of RA (e.g. achieving ACR20, ACR50, or ACR70 response at week 24 and/or week 48), reducing disease activity (e.g. Disease Activity Score, DAS28), slowing the progression of structural joint damage, improving physical function, etc.
  • prophylactically effective amount refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, but not necessarily, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount would be less than the therapeutically effective amount.
  • Administration “in combination with” one or more further therapeutic agents includes simultaneous (concurrent) and consecutive administration in any order.
  • a “pharmaceutically acceptable carrier” refers to a non-toxic solid, semisolid, or liquid filler, diluent, encapsulating material, formulation auxiliary, or carrier conventional in the art for use with a therapeutic agent that together comprise a “pharmaceutical composition” for administration to a subject.
  • a pharmaceutically acceptable carrier is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation.
  • the pharmaceutically acceptable carrier is appropriate for the formulation employed.
  • the carrier may be a gel capsule. If the therapeutic agent is to be administered subcutaneously, the carrier ideally is not irritable to the skin and does not cause injection site reaction.
  • Anti-CSF1R antibodies include, but are not limited to, humanized antibodies, chimeric antibodies, mouse antibodies, human antibodies, and antibodies comprising the heavy chain and/or light chain CDRs discussed herein.
  • humanized antibodies that bind CSF1R are provided.
  • Humanized antibodies are useful as therapeutic molecules because humanized antibodies reduce or eliminate the human immune response to non-human antibodies (such as the human anti-mouse antibody (HAMA) response), which can result in an immune response to an antibody therapeutic, and decreased effectiveness of the therapeutic.
  • HAMA human anti-mouse antibody
  • Nonlimiting exemplary humanized antibodies include huAb1 through huAb16, described herein.
  • Nonlimiting exemplary humanized antibodies also include antibodies comprising a heavy chain variable region of an antibody selected from huAb1 to huAb16 and/or a light chain variable region of an antibody selected from huAb1 to huAb16.
  • Nonlimiting exemplary humanized antibodies include antibodies comprising a heavy chain variable region selected from SEQ ID NOs: 39 to 45 and/or a light chain variable region selected from SEQ ID NOs: 46 to 52.
  • Exemplary humanized antibodies also include, but are not limited to, humanized antibodies comprising heavy chain CDR1, CDR2, and CDR3, and/or light chain CDR1, CDR2, and CDR3 of an antibody selected from 0301, 0302, and 0311.
  • a humanized anti-CSF1R antibody comprises heavy chain CDR1, CDR2, and CDR3 and/or a light chain CDR1, CDR2, and CDR3 of an antibody selected from 0301, 0302, and 0311.
  • Nonlimiting exemplary humanized anti-CSF1R antibodies include antibodies comprising sets of heavy chain CDR1, CDR2, and CDR3 selected from: SEQ ID NOs: 15, 16, and 17; SEQ ID NOs: 21, 22, and 23; and SEQ ID NOs: 27, 28, and 29.
  • Nonlimiting exemplary humanized anti-CSF1R antibodies also include antibodies comprising sets of light chain CDR1, CDR2, and CDR3 selected from: SEQ ID NOs: 18, 19, and 20; SEQ ID NOs: 24, 25, and 26; and SEQ ID NOs: 30, 31, and 32.
  • Nonlimiting exemplary humanized anti-CSF1R antibodies include antibodies comprising the sets of heavy chain CDR1, CDR2, and CDR3, and light chain CDR1, CDR2, and CDR3 in Table 1 (SEQ ID NOs shown; see Table 8 for sequences). Each row of Table 1 shows the heavy chain CDR1, CDR2, and CDR3, and light chain CDR1, CDR2, and CDR3 of an exemplary antibody.
  • a humanized anti-CSF1R antibody comprises a heavy chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45, and wherein the antibody binds CSF1R.
  • a humanized anti-CSF1R antibody comprises a light chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52, wherein the antibody binds CSF1R.
  • a humanized anti-CSF1R antibody comprises a heavy chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45; and a light chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52; wherein the antibody binds CSF1R.
  • whether a particular polypeptide is, for example, at least 95% identical to an amino acid sequence can be determined using, e.g., a computer program.
  • the percentage of identity is calculated over the full length of the reference amino acid sequence.
  • a humanized anti-CSF1R antibody comprises at least one of the CDRs discussed herein. That is, in some embodiments, a humanized anti-CSF1R antibody comprises at least one CDR selected from a heavy chain CDR1 discussed herein, a heavy chain CDR2 discussed herein, a heavy chain CDR3 discussed herein, a light chain CDR1 discussed herein, a light chain CDR2 discussed herein, and a light chain CDR3 discussed herein. Further, in some embodiments, a humanized anti-CSF1R antibody comprises at least one mutated CDR based on a CDR discussed herein, wherein the mutated CDR comprises 1, 2, 3, or 4 amino acid substitutions relative to the CDR discussed herein.
  • one or more of the amino acid substitutions are conservative amino acid substitutions.
  • One skilled in the art can select one or more suitable conservative amino acid substitutions for a particular CDR sequence, wherein the suitable conservative amino acid substitutions are not predicted to significantly alter the binding properties of the antibody comprising the mutated CDR.
  • Exemplary humanized anti-CSF1R antibodies also include antibodies that compete for binding to CSF1R with an antibody described herein.
  • a humanized anti-CSF1R antibody is provided that competes for binding to CSF1R with an antibody selected from Fabs 0301, 0302, and 0311; and bivalent (i.e., having two heavy chains and two light chains) antibody versions of those Fabs.
  • a humanized antibody described herein comprises one or more human constant regions.
  • the human heavy chain constant region is of an isotype selected from IgA, IgG, and IgD.
  • the human light chain constant region is of an isotype selected from ⁇ and ⁇ .
  • a humanized antibody described herein comprises a human IgG constant region.
  • a humanized antibody described herein comprises a human IgG4 heavy chain constant region.
  • a humanized antibody described herein comprises an S241P mutation in the human IgG4 constant region.
  • a humanized antibody described herein comprises a human IgG4 constant region and a human ⁇ light chain.
  • the choice of heavy chain constant region can determine whether or not an antibody will have effector function in vivo.
  • effector function includes antibody-dependent cell-mediated cytotoxicity (ADCC) and/or complement-dependent cytotoxicity (CDC), and can result in killing of the cell to which the antibody is bound.
  • cell killing may be desirable, for example, when the antibody binds to a cell that supports the maintenance or growth of the tumor.
  • Exemplary cells that may support the maintenance or growth of a tumor include, but are not limited to, tumor cells themselves, cells that aid in the recruitment of vasculature to the tumor, and cells that provide ligands, growth factors, or counter-receptors that support or promote tumor growth or tumor survival.
  • an anti-CSF1R antibody comprising a human IgG1 heavy chain or a human IgG3 heavy chain is selected.
  • effector function may not be desirable.
  • anti-CSF1R antibodies developed for the treatment of cancer may not be suitable for use in treatment of lupus and/or MS and/or RA and/or osteolysis.
  • an anti-CSF1R antibody that lacks significant effector function is used in treatment of lupus and/or MS and/or RA and/or osteolysis.
  • an anti-CSF1R antibody for treatment of lupus and/or MS and/or RA and/or osteolysis comprises a human IgG4 or IgG2 heavy chain constant region.
  • an IgG4 constant region comprises an S241P mutation.
  • An antibody may be humanized by any method.
  • Nonlimiting exemplary methods of humanization include methods described, e.g., in U.S. Pat. Nos. 5,530,101; 5,585,089; 5,693,761; 5,693,762; 6,180,370; Jones et al., Nature 321: 522-525 (1986); Riechmann et al., Nature 332: 323-27 (1988); Verhoeyen et al., Science 239: 1534-36 (1988); and U.S. Publication No. US 2009/0136500.
  • a humanized antibody is an antibody in which at least one amino acid in a framework region of a non-human variable region has been replaced with the amino acid from the corresponding location in a human framework region.
  • at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least 10, at least 11, at least 12, at least 15, or at least 20 amino acids in the framework regions of a non-human variable region are replaced with an amino acid from one or more corresponding locations in one or more human framework regions.
  • some of the corresponding human amino acids used for substitution are from the framework regions of different human immunoglobulin genes. That is, in some such embodiments, one or more of the non-human amino acids may be replaced with corresponding amino acids from a human framework region of a first human antibody or encoded by a first human immunoglobulin gene, one or more of the non-human amino acids may be replaced with corresponding amino acids from a human framework region of a second human antibody or encoded by a second human immunoglobulin gene, one or more of the non-human amino acids may be replaced with corresponding amino acids from a human framework region of a third human antibody or encoded by a third human immunoglobulin gene, etc.
  • all of the corresponding human amino acids being used for substitution in a single framework region need not be from the same human framework. In some embodiments, however, all of the corresponding human amino acids being used for substitution are from the same human antibody or encoded by the same human immunoglobulin gene.
  • an antibody is humanized by replacing one or more entire framework regions with corresponding human framework regions.
  • a human framework region is selected that has the highest level of homology to the non-human framework region being replaced.
  • such a humanized antibody is a CDR-grafted antibody.
  • one or more framework amino acids are changed back to the corresponding amino acid in a mouse framework region.
  • Such “back mutations” are made, in some embodiments, to retain one or more mouse framework amino acids that appear to contribute to the structure of one or more of the CDRs and/or that may be involved in antigen contacts and/or appear to be involved in the overall structural integrity of the antibody.
  • ten or fewer, nine or fewer, eight or fewer, seven or fewer, six or fewer, five or fewer, four or fewer, three or fewer, two or fewer, one, or zero back mutations are made to the framework regions of an antibody following CDR grafting.
  • a humanized antibody also comprises a human heavy chain constant region and/or a human light chain constant region.
  • an anti-CSF1R antibody is a chimeric antibody.
  • an anti-CSF1R antibody comprises at least one non-human variable region and at least one human constant region.
  • all of the variable regions of an anti-CSF1R antibody are non-human variable regions, and all of the constant regions of an anti-CSF1R antibody are human constant regions.
  • one or more variable regions of a chimeric antibody are mouse variable regions.
  • the human constant region of a chimeric antibody need not be of the same isotype as the non-human constant region, if any, it replaces. Chimeric antibodies are discussed, e.g., in U.S. Pat. No. 4,816,567; and Morrison et al. Proc. Natl. Acad. Sci. USA 81: 6851-55 (1984).
  • Nonlimiting exemplary chimeric antibodies include chimeric antibodies comprising the heavy and/or light chain variable regions of an antibody selected from 0301, 0302, and 0311. Additional nonlimiting exemplary chimeric antibodies include chimeric antibodies comprising heavy chain CDR1, CDR2, and CDR3, and/or light chain CDR1, CDR2, and CDR3 of an antibody selected from 0301, 0302, and 0311.
  • Nonlimiting exemplary chimeric anti-CSF1R antibodies include antibodies comprising the following pairs of heavy and light chain variable regions: SEQ ID NOs: 9 and 10; SEQ ID NOs: 11 and 12; and SEQ ID NOs: 13 and 14.
  • Nonlimiting exemplary anti-CSF1R antibodies include antibodies comprising a set of heavy chain CDR1, CDR2, and CDR3, and light chain CDR1, CDR2, and CDR3 shown above in Table 1.
  • a chimeric anti-CSF1R antibody comprises a heavy chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45, wherein the antibody binds CSF1R.
  • a chimeric anti-CSF1R antibody comprises a light chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52, wherein the antibody binds CSF1R.
  • a chimeric anti-CSF1R antibody comprises a heavy chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45; and a light chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52; wherein the antibody binds CSF1R.
  • a chimeric anti-CSF1R antibody comprises at least one of the CDRs discussed herein. That is, in some embodiments, a chimeric anti-CSF1R antibody comprises at least one CDR selected from a heavy chain CDR1 discussed herein, a heavy chain CDR2 discussed herein, a heavy chain CDR3 discussed herein, a light chain CDR1 discussed herein, a light chain CDR2 discussed herein, and a light chain CDR3 discussed herein. Further, in some embodiments, a chimeric anti-CSF1R antibody comprises at least one mutated CDR based on a CDR discussed herein, wherein the mutated CDR comprises 1, 2, 3, or 4 amino acid substitutions relative to the CDR discussed herein.
  • one or more of the amino acid substitutions are conservative amino acid substitutions.
  • One skilled in the art can select one or more suitable conservative amino acid substitutions for a particular CDR sequence, wherein the suitable conservative amino acid substitutions are not predicted to significantly alter the binding properties of the antibody comprising the mutated CDR.
  • Exemplary chimeric anti-CSF1R antibodies also include chimeric antibodies that compete for binding to CSF1R with an antibody described herein.
  • a chimeric anti-CSF1R antibody is provided that competes for binding to CSF1R with an antibody selected from Fabs 0301, 0302, and 0311; and bivalent (i.e., having two heavy chains and two light chains) antibody versions of those Fabs.
  • a chimeric antibody described herein comprises one or more human constant regions.
  • the human heavy chain constant region is of an isotype selected from IgA, IgG, and IgD.
  • the human light chain constant region is of an isotype selected from ⁇ and ⁇ .
  • a chimeric antibody described herein comprises a human IgG constant region.
  • a chimeric antibody described herein comprises a human IgG4 heavy chain constant region.
  • a chimeric antibody described herein comprises an S241P mutation in the human IgG4 constant region.
  • a chimeric antibody described herein comprises a human IgG4 constant region and a human ⁇ light chain.
  • effector function may depend on the particular method of treatment intended for an antibody.
  • a chimeric anti-CSF1R antibody comprising a human IgG1 heavy chain constant region or a human IgG3 heavy chain constant region is selected.
  • a chimeric anti-CSF1R antibody comprising a human IgG4 or IgG2 heavy chain constant region is selected.
  • Human antibodies can be made by any suitable method.
  • Nonlimiting exemplary methods include making human antibodies in transgenic mice that comprise human immunoglobulin loci. See, e.g., Jakobovits et al., Proc. Natl. Acad. Sci. USA 90: 2551-55 (1993); Jakobovits et al., Nature 362: 255-8 (1993); Lonberg et al., Nature 368: 856-9 (1994); and U.S. Pat. Nos. 5,545,807; 6,713,610; 6,673,986; 6,162,963; 5,545,807; 6,300,129; 6,255,458; 5,877,397; 5,874,299; and 5,545,806.
  • Nonlimiting exemplary methods also include making human antibodies using phage display libraries. See, e.g., Hoogenboom et al., J. Mol. Biol. 227: 381-8 (1992); Marks et al., J. Mol. Biol. 222: 581-97 (1991); and PCT Publication No. WO 99/10494.
  • a human anti-CSF1R antibody binds to a polypeptide having the sequence of SEQ ID NO: 1.
  • Exemplary human anti-CSF1R antibodies also include antibodies that compete for binding to CSF1R with an antibody described herein.
  • a human anti-CSF1R antibody is provided that competes for binding to CSF1R with an antibody selected from Fabs 0301, 0302, and 0311, and bivalent (i.e., having two heavy chains and two light chains) antibody versions of those Fabs.
  • a human anti-CSF1R antibody comprises one or more human constant regions.
  • the human heavy chain constant region is of an isotype selected from IgA, IgG, and IgD.
  • the human light chain constant region is of an isotype selected from ⁇ and ⁇ .
  • a human antibody described herein comprises a human IgG constant region.
  • a human antibody described herein comprises a human IgG4 heavy chain constant region.
  • a human antibody described herein comprises an S241P mutation in the human IgG4 constant region.
  • a human antibody described herein comprises a human IgG4 constant region and a human ⁇ light chain.
  • a human anti-CSF1R antibody comprising a human IgG1 heavy chain constant region or a human IgG3 heavy chain constant region is selected. In some embodiments, when effector function is not desirable, a human anti-CSF1R antibody comprising a human IgG4 or IgG2 heavy chain constant region is selected.
  • anti-CSF1R antibodies also include, but are not limited to, mouse, humanized, human, chimeric, and engineered antibodies that comprise, for example, one or more of the CDR sequences described herein.
  • an anti-CSF1R antibody comprises a heavy chain variable region described herein.
  • an anti-CSF1R antibody comprises a light chain variable region described herein.
  • an anti-CSF1R antibody comprises a heavy chain variable region described herein and a light chain variable region described herein.
  • an anti-CSF1R antibody comprises heavy chain CDR1, CDR2, and CDR3 described herein.
  • an anti-CSF1R antibody comprises light chain CDR1, CDR2, and CDR3 described herein. In some embodiments, an anti-CSF1R antibody comprises heavy chain CDR1, CDR2, and CDR3 described herein and light chain CDR1, CDR2, and CDR3 described herein.
  • an anti-CSF1R antibody comprises a heavy chain variable region of an antibody selected from Fabs 0301, 0302, and 0311.
  • Nonlimiting exemplary anti-CSF1R antibodies also include antibodies comprising a heavy chain variable region of an antibody selected from humanized antibodies huAb1 to huAb16.
  • Nonlimiting exemplary anti-CSF1R antibodies include antibodies comprising a heavy chain variable region comprising a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45.
  • an anti-CSF1R antibody comprises a light chain variable region of an antibody selected from Fabs 0301, 0302, and 0311.
  • Nonlimiting exemplary anti-CSF1R antibodies also include antibodies comprising a light chain variable region of an antibody selected from humanized antibodies huAb1 to huAb16.
  • Nonlimiting exemplary anti-CSF1R antibodies include antibodies comprising a light chain variable region comprising a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52.
  • an anti-CSF1R antibody comprises a heavy chain variable region and a light chain variable region of an antibody selected from Fabs 0301, 0302, and 0311.
  • Nonlimiting exemplary anti-CSF1R antibodies also include antibodies comprising a heavy chain variable region and a light chain variable region of an antibody selected from humanized antibodies huAb1 to huAb16.
  • Nonlimiting exemplary anti-CSF1R antibodies include antibodies comprising the following pairs of heavy and light chain variable regions: SEQ ID NOs: 9 and 10; SEQ ID NOs: 11 and 12; and SEQ ID NOs: 13 and 14; SEQ ID NOs: 39 and 40; SEQ ID NOs: 41 and 42; SEQ ID NOs: 43 and 44; SEQ ID NOs: 45 and 46; SEQ ID NOs: 47 and 48; SEQ ID NOs: 49 and 50; and SEQ ID NOs: 51 and 52.
  • Nonlimiting exemplary anti-CSF1R antibodies also include antibodies comprising the following pairs of heavy and light chains: SEQ ID NOs: 33 and 34; SEQ ID NOs: 35 and 36; and SEQ ID NOs: 37 and 38.
  • an anti-CSF1R antibody comprises heavy chain CDR1, CDR2, and CDR3 of an antibody selected from Fabs 0301, 0302, and 0311.
  • Nonlimiting exemplary anti-CSF1R antibodies include antibodies comprising sets of heavy chain CDR1, CDR2, and CDR3 selected from: SEQ ID NOs: 15, 16, and 17; SEQ ID NOs: 21, 22, and 23; and SEQ ID NOs: 27, 28, and 29.
  • an anti-CSF1R antibody comprises light chain CDR1, CDR2, and CDR3 of an antibody selected from Fabs 0301, 0302, and 0311.
  • Nonlimiting exemplary anti-CSF1R antibodies include antibodies comprising sets of light chain CDR1, CDR2, and CDR3 selected from: SEQ ID NOs: 18, 19, and 20; SEQ ID NOs: 24, 25, and 26; and SEQ ID NOs: 30, 31, and 32.
  • an anti-CSF1R antibody comprises heavy chain CDR1, CDR2, and CDR3, and light chain CDR1, CDR2, and CDR3 of an antibody selected from Fabs 0301, 0302, and 0311.
  • Nonlimiting exemplary anti-CSF1R antibodies include antibodies comprising the sets of heavy chain CDR1, CDR2, and CDR3, and light chain CDR1, CDR2, and CDR3 shown above in Table 1.
  • an anti-CSF1R antibody comprises a heavy chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45, wherein the antibody binds CSF1R.
  • an anti-CSF1R antibody comprises a light chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52, wherein the antibody binds CSF1R.
  • an anti-CSF1R antibody comprises a heavy chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45; and a light chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52; wherein the antibody binds CSF1R.
  • an anti-CSF1R antibody comprises at least one of the CDRs discussed herein. That is, in some embodiments, an anti-CSF1R antibody comprises at least one CDR selected from a heavy chain CDR1 discussed herein, a heavy chain CDR2 discussed herein, a heavy chain CDR3 discussed herein, a light chain CDR1 discussed herein, a light chain CDR2 discussed herein, and a light chain CDR3 discussed herein. Further, in some embodiments, an anti-CSF1R antibody comprises at least one mutated CDR based on a CDR discussed herein, wherein the mutated CDR comprises 1, 2, 3, or 4 amino acid substitutions relative to the CDR discussed herein.
  • one or more of the amino acid substitutions are conservative amino acid substitutions.
  • One skilled in the art can select one or more suitable conservative amino acid substitutions for a particular CDR sequence, wherein the suitable conservative amino acid substitutions are not predicted to significantly alter the binding properties of the antibody comprising the mutated CDR.
  • anti-CSF1R antibodies also include antibodies that compete for binding to CSF1R with an antibody described herein.
  • an anti-CSF1R antibody is provided that competes for binding to CSF1R with an antibody selected from Fabs 0301, 0302, and 0311, and bivalent (i.e., having two heavy chains and two light chains) antibody versions of those Fabs.
  • an antibody described herein comprises one or more human constant regions.
  • the human heavy chain constant region is of an isotype selected from IgA, IgG, and IgD.
  • the human light chain constant region is of an isotype selected from ⁇ and ⁇ .
  • an antibody described herein comprises a human IgG constant region.
  • an antibody described herein comprises a human IgG4 heavy chain constant region.
  • an antibody described herein comprises an S241P mutation in the human IgG4 constant region.
  • an antibody described herein comprises a human IgG4 constant region and a human ⁇ light chain.
  • effector function may depend on the particular method of treatment intended for an antibody.
  • an anti-CSF1R antibody comprising a human IgG1 heavy chain constant region or a human IgG3 heavy chain constant region is selected.
  • an anti-CSF1R antibody comprising a human IgG4 or IgG2 heavy chain constant region is selected.
  • anti-CSF1R antibody heavy chain variable regions are provided.
  • an anti-CSF1R antibody heavy chain variable region is a mouse variable region, a human variable region, or a humanized variable region.
  • An anti-CSF1R antibody heavy chain variable region comprises a heavy chain CDR1, FR2, CDR2, FR3, and CDR3.
  • an anti-CSF1R antibody heavy chain variable region further comprises a heavy chain FR1 and/or FR4.
  • Nonlimiting exemplary heavy chain variable regions include, but are not limited to, heavy chain variable regions having an amino acid sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45.
  • an anti-CSF1R antibody heavy chain variable region comprises a CDR1 comprising a sequence selected from SEQ ID NOs: 15, 21, and 27.
  • an anti-CSF1R antibody heavy chain variable region comprises a CDR2 comprising a sequence selected from SEQ ID NOs: 16, 22, and 28.
  • an anti-CSF1R antibody heavy chain variable region comprises a CDR3 comprising a sequence selected from SEQ ID NOs: 17, 23, and 29.
  • Nonlimiting exemplary heavy chain variable regions include, but are not limited to, heavy chain variable regions comprising sets of CDR1, CDR2, and CDR3 selected from: SEQ ID NOs: 15, 16, and 17; SEQ ID NOs: 21, 22, and 23; and SEQ ID NOs: 27, 28, and 29.
  • an anti-CSF1R antibody heavy chain comprises a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45, wherein the heavy chain, together with a light chain, is capable of forming an antibody that binds CSF1R.
  • an anti-CSF1R antibody heavy chain comprises at least one of the CDRs discussed herein. That is, in some embodiments, an anti-CSF1R antibody heavy chain comprises at least one CDR selected from a heavy chain CDR1 discussed herein, a heavy chain CDR2 discussed herein, and a heavy chain CDR3 discussed herein. Further, in some embodiments, an anti-CSF1R antibody heavy chain comprises at least one mutated CDR based on a CDR discussed herein, wherein the mutated CDR comprises 1, 2, 3, or 4 amino acid substitutions relative to the CDR discussed herein. In some embodiments, one or more of the amino acid substitutions are conservative amino acid substitutions. One skilled in the art can select one or more suitable conservative amino acid substitutions for a particular CDR sequence, wherein the suitable conservative amino acid substitutions are not predicted to significantly alter the binding properties of the heavy chain comprising the mutated CDR.
  • a heavy chain comprises a heavy chain constant region. In some embodiments, a heavy chain comprises a human heavy chain constant region. In some embodiments, the human heavy chain constant region is of an isotype selected from IgA, IgG, and IgD. In some embodiments, the human heavy chain constant region is an IgG constant region. In some embodiments, a heavy chain comprises a human igG4 heavy chain constant region. In some such embodiments, the human IgG4 heavy chain constant region comprises an S241P mutation.
  • a heavy chain when effector function is desirable, comprises a human IgG1 or IgG3 heavy chain constant region. In some embodiments, when effector function is less desirable, a heavy chain comprises a human IgG4 or IgG2 heavy chain constant region.
  • anti-CSF1R antibody light chain variable regions are provided.
  • an anti-CSF1R antibody light chain variable region is a mouse variable region, a human variable region, or a humanized variable region.
  • An anti-CSF1R antibody light chain variable region comprises a light chain CDR1, FR2, CDR2, FR3, and CDR3.
  • an anti-CSF1R antibody light chain variable region further comprises a light chain FR1 and/or FR4.
  • Nonlimiting exemplary light chain variable regions include light chain variable regions having an amino acid sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52.
  • an anti-CSF1R antibody light chain variable region comprises a CDR1 comprising a sequence selected from SEQ ID NOs: 18, 24 and 30.
  • an anti-CSF1R antibody light chain variable region comprises a CDR2 comprising a sequence selected from SEQ ID NOs: 19, 25, and 31.
  • an anti-CSF1R antibody light chain variable region comprises a CDR3 comprising a sequence selected from SEQ ID NOs: 20, 26, and 32.
  • Nonlimiting exemplary light chain variable regions include, but are not limited to, light chain variable regions comprising sets of CDR1, CDR2, and CDR3 selected from: SEQ ID NOs: 18, 19, and 20; SEQ ID NOs: 24, 25, and 26; and SEQ ID NOs: 30, 31, and 32.
  • an anti-CSF1R antibody light chain comprises a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52, wherein the light chain, together with a heavy chain, is capable of forming an antibody that binds CSF1R.
  • an anti-CSF1R antibody light chain comprises at least one of the CDRs discussed herein. That is, in some embodiments, an anti-CSF1R antibody light chain comprises at least one CDR selected from a light chain CDR1 discussed herein, a light chain CDR2 discussed herein, and a light chain CDR3 discussed herein. Further, in some embodiments, an anti-CSF1R antibody light chain comprises at least one mutated CDR based on a CDR discussed herein, wherein the mutated CDR comprises 1, 2, 3, or 4 amino acid substitutions relative to the CDR discussed herein. In some embodiments, one or more of the amino acid substitutions are conservative amino acid substitutions. One skilled in the art can select one or more suitable conservative amino acid substitutions for a particular CDR sequence, wherein the suitable conservative amino acid substitutions are not predicted to significantly alter the binding properties of the light chain comprising the mutated CDR.
  • a light chain comprises a human light chain constant region.
  • a human light chain constant region is selected from a human ⁇ and a human ⁇ light chain constant region.
  • additional molecules that bind CSF1R are provided.
  • Such molecules include, but are not limited to, non-canonical scaffolds, such as anti-calins, adnectins, ankyrin repeats, etc. See, e.g., Hosse et al., Prot. Sci. 15:14 (2006); Fiedler, M. and Skerra, A., “Non-Antibody Scaffolds,” pp. 467-499 in Handbook of Therapeutic Antibodies, Dubel, S., ed., Wiley-VCH, Weinheim, Germany, 2007.
  • an antibody having a structure described above binds to the CSF1R with a binding affinity (K D ) of less than 1 nM, blocks binding of CSF1 and/or IL-34 to CSF1R, and inhibits CSF1R phosphorylation induced by CSF1 and/or IL-34.
  • K D binding affinity
  • an anti-CSF1R antibody binds to the extracellular domain of CSF1R (CSF1R-ECD).
  • an anti-CSF1R antibody has a binding affinity (K D ) for CSF1R of less than 1 nM, less than 0.5 nM, less than 0.1 nM, or less than 0.05 nM.
  • an anti-CSF1R antibody has a K D of between 0.01 and 1 nM, between 0.01 and 0.5 nM, between 0.01 and 0.1 nM, between 0.01 and 0.05 nM, or between 0.02 and 0.05 nM.
  • an anti-CSF1R antibody blocks ligand binding to CSF1R. In some embodiments, an anti-CSF1R antibody blocks binding of CSF1 to CSF1R. In some embodiments, an anti-CSF1R antibody blocks binding of IL-34 to CSF1R. In some embodiments, an anti-CSF1R antibody blocks binding of both CSF1 and IL-34 to CSF1R. In some embodiments, an antibody that blocks ligand binding binds to the extracellular domain of CSF1R. In some embodiments, an antibody blocks ligand binding to CSF1R when it reduces the amount of detectable binding of a ligand to CSF1R by at least 50%, using the assay described, e.g., U.S.
  • an antibody reduces the amount of detectable binding of a ligand to CSF1R by at least 60%, at least 70%, at least 80%, or at least 90%. In some such embodiments, the antibody is said to block ligand binding by at least 50%, at least 60%, at least 70%, etc.
  • an anti-CSF1R antibody inhibits ligand-induced CSF1R phosphorylation. In some embodiments, an anti-CSF1R antibody inhibits CSF1-induced CSF1R phosphorylation. In some embodiments, an anti-CSF1R antibody inhibits IL-34-induced CSF1R phosphorylation. In some embodiments, an anti-CSF1R antibody inhibits both CSF1-induced and IL-34-induced CSF1R phosphorylation.
  • an antibody is considered to “inhibit ligand-induced CSF1R phosphorylation” when it reduces the amount of detectable ligand-induced CSF1R phosphorylation by at least 50%, using the assay described, e.g., U.S. Pat. No. 8,206,715 B2, Example 6, which is incorporated herein by reference for any purpose.
  • an antibody reduces the amount of detectable ligand-induced CSF1R phosphorylation by at least 60%, at least 70%, at least 80%, or at least 90%.
  • the antibody is said to inhibit ligand-induced CSF1R phosphorylation by at least at least 50%, at least 60%, at least 70%, etc.
  • an antibody inhibits monocyte proliferation and/or survival responses in the presence of CSF1 and/or IL-34.
  • an antibody is considered to “inhibit monocyte proliferation and/or survival responses” when it reduces the amount of monocyte proliferation and/or survival responses in the presence of CSF1 and/or IL-34 by at least 50%, using the assay described, e.g., U.S. Pat. No. 8,206,715 B2, Example 10, which is incorporated herein by reference for any purpose.
  • an antibody reduces the amount of monocyte proliferation and/or survival responses in the presence of CSF1 and/or IL-34 by at least 60%, at least 70%, at least 80%, or at least 90%.
  • the antibody is said to inhibit monocyte proliferation and/or survival responses by at least at least 50%, at least 60%, at least 70%, etc.
  • an anti-CSF1R antibody is conjugated to a label and/or a cytotoxic agent.
  • a label is a moiety that facilitates detection of the antibody and/or facilitates detection of a molecule to which the antibody binds.
  • Nonlimiting exemplary labels include, but are not limited to, radioisotopes, fluorescent groups, enzymatic groups, chemiluminescent groups, biotin, epitope tags, metal-binding tags, etc.
  • One skilled in the art can select a suitable label according to the intended application.
  • a cytotoxic agent is a moiety that reduces the proliferative capacity of one or more cells.
  • a cell has reduced proliferative capacity when the cell becomes less able to proliferate, for example, because the cell undergoes apoptosis or otherwise dies, the cell fails to proceed through the cell cycle and/or fails to divide, the cell differentiates, etc.
  • Nonlimiting exemplary cytotoxic agents include, but are not limited to, radioisotopes, toxins, and chemotherapeutic agents.
  • One skilled in the art can select a suitable cytotoxic according to the intended application.
  • a label and/or a cytotoxic agent is conjugated to an antibody using chemical methods in vitro.
  • Nonlimiting exemplary chemical methods of conjugation are known in the art, and include services, methods and/or reagents commercially available from, e.g., Thermo Scientific Life Science Research Produces (formerly Pierce; Rockford, Ill.), Prozyme (Hayward, Calif.), SACRI Antibody Services (Calgary, Canada), AbD Serotec (Raleigh, N.C.), etc.
  • the label and/or cytotoxic agent when a label and/or cytotoxic agent is a polypeptide, the label and/or cytotoxic agent can be expressed from the same expression vector with at least one antibody chain to produce a polypeptide comprising the label and/or cytotoxic agent fused to an antibody chain.
  • a suitable method for conjugating a label and/or cytotoxic agent to an antibody can be selected from the same expression vector with at least one antibody chain to produce a polypeptide comprising the label and/or cytotoxic agent fused to an antibody chain.
  • a leader sequence from a heterologous protein may be desirable.
  • a leader sequence is selected from SEQ ID NOs: 3 and 4, which are light chain and heavy chain leader sequences, respectively.
  • employing heterologous leader sequences may be advantageous in that a resulting mature polypeptide may remain unaltered as the leader sequence is removed in the ER during the secretion process.
  • the addition of a heterologous leader sequence may be required to express and secrete some proteins.
  • leader sequence sequences are described, e.g., in the online Leader sequence Database maintained by the Department of Biochemistry, National University of Singapore. See Choo et al., BMC Bioinformatics, 6: 249 (2005); and PCT Publication No. WO 2006/081430.
  • nucleic acid molecules comprising polynucleotides that encode one or more chains of anti-CSF1R antibodies are provided.
  • a nucleic acid molecule comprises a polynucleotide that encodes a heavy chain or a light chain of an anti-CSF1R antibody.
  • a nucleic acid molecule comprises both a polynucleotide that encodes a heavy chain and a polynucleotide that encodes a light chain, of an anti-CSF1R antibody.
  • a first nucleic acid molecule comprises a first polynucleotide that encodes a heavy chain and a second nucleic acid molecule comprises a second polynucleotide that encodes a light chain.
  • the heavy chain and the light chain are expressed from one nucleic acid molecule, or from two separate nucleic acid molecules, as two separate polypeptides.
  • a single polynucleotide encodes a single polypeptide comprising both a heavy chain and a light chain linked together.
  • a polynucleotide encoding a heavy chain or light chain of an anti-CSF1R antibody comprises a nucleotide sequence that encodes a leader sequence, which, when translated, is located at the N terminus of the heavy chain or light chain.
  • the leader sequence may be the native heavy or light chain leader sequence, or may be another heterologous leader sequence.
  • Nucleic acid molecules may be constructed using recombinant DNA techniques conventional in the art.
  • a nucleic acid molecule is an expression vector that is suitable for expression in a selected host cell.
  • Vectors comprising polynucleotides that encode anti-CSF1R heavy chains and/or anti-CSF1R light chains are provided. Vectors comprising polynucleotides that encode anti-CSF1R heavy chains and/or anti-CSF1R light chains are also provided. Such vectors include, but are not limited to, DNA vectors, phage vectors, viral vectors, retroviral vectors, etc.
  • a vector comprises a first polynucleotide sequence encoding a heavy chain and a second polynucleotide sequence encoding a light chain.
  • the heavy chain and light chain are expressed from the vector as two separate polypeptides. In some embodiments, the heavy chain and light chain are expressed as part of a single polypeptide, such as, for example, when the antibody is an scFv.
  • a first vector comprises a polynucleotide that encodes a heavy chain and a second vector comprises a polynucleotide that encodes a light chain.
  • the first vector and second vector are transfected into host cells in similar amounts (such as similar molar amounts or similar mass amounts).
  • a mole- or mass-ratio of between 5:1 and 1:5 of the first vector and the second vector is transfected into host cells.
  • a mass ratio of between 1:1 and 1:5 for the vector encoding the heavy chain and the vector encoding the light chain is used.
  • a mass ratio of 1:2 for the vector encoding the heavy chain and the vector encoding the light chain is used.
  • a vector is selected that is optimized for expression of polypeptides in CHO or CHO-derived cells, or in NSO cells. Exemplary such vectors are described, e.g., in Running Deer et al., Biotechnol. Frog. 20:880-889 (2004).
  • a vector is chosen for in vivo expression of anti-CSF1R heavy chains and/or anti-CSF1R light chains in animals, including humans.
  • expression of the polypeptide is under the control of a promoter that functions in a tissue-specific manner.
  • tissue-specific promoters are described, e.g., in PCT Publication No. WO 2006/076288.
  • anti-CSF1R heavy chains and/or anti-CSF1R light chains may be expressed in prokaryotic cells, such as bacterial cells; or in eukaryotic cells, such as fungal cells (such as yeast), plant cells, insect cells, and mammalian cells. Such expression may be carried out, for example, according to procedures known in the art.
  • exemplary eukaryotic cells that may be used to express polypeptides include, but are not limited to, COS cells, including COS 7 cells; 293 cells, including 293-6E cells; CHO cells, including CHO-S and DG44 cells; PER.C6® cells (Crucell); and NSO cells.
  • anti-CSF1R heavy chains and/or anti-CSF1R light chains may be expressed in yeast. See, e.g., U.S. Publication No. US 2006/0270045 A1.
  • a particular eukaryotic host cell is selected based on its ability to make desired post-translational modifications to the anti-CSF1R heavy chains and/or anti-CSF1R light chains.
  • CHO cells produce polypeptides that have a higher level of sialylation than the same polypeptide produced in 293 cells.
  • nucleic acids into a desired host cell may be accomplished by any method, including but not limited to, calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, etc.
  • Nonlimiting exemplary methods are described, e.g., in Sambrook et al., Molecular Cloning, A Laboratory Manual, 3 rd ed. Cold Spring Harbor Laboratory Press (2001).
  • Nucleic acids may be transiently or stably transfected in the desired host cells, according to any suitable method.
  • one or more polypeptides may be produced in vivo in an animal that has been engineered or transfected with one or more nucleic acid molecules encoding the polypeptides, according to any suitable method.
  • Anti-CSF1R antibodies may be purified by any suitable method. Such methods include, but are not limited to, the use of affinity matrices or hydrophobic interaction chromatography. Suitable affinity ligands include the CSF1R ECD and ligands that bind antibody constant regions. For example, a Protein A, Protein G, Protein A/G, or an antibody affinity column may be used to bind the constant region and to purify an anti-CSF1R antibody. Hydrophobic interactive chromatography, for example, a butyl or phenyl column, may also suitable for purifying some polypeptides. Many methods of purifying polypeptides are known in the art.
  • an anti-CSF1R antibody is produced in a cell-free system.
  • a cell-free system Nonlimiting exemplary cell-free systems are described, e.g., in Sitaraman et al., Methods Mol. Biol. 498: 229-44 (2009); Spirin, Trends Biotechnol. 22: 538-45 (2004); Endo et al., Biotechnol. Adv. 21: 695-713 (2003).
  • the present disclosure relates to methods of reducing one or more factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9.
  • a subject has an elevated level of one or more factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 prior to treatment with the antibodies described herein.
  • the level of a factor is determined by detecting the level of the protein.
  • Nonlimiting exemplary amino acid sequences for human IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-9, and MMP-2 are shown in SEQ ID NOs: 96 to 108, respectively.
  • Any native forms of the proteins including naturally-occurring variants, such as variants comprising substitutions and/or deletions (such as truncations), variants comprising post-translational modifications, splice variants, and allelic variants, are specifically contemplated.
  • the level of the factor is determined by detecting the level of the mRNA.
  • Exemplary nucleotide sequences for human IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 mRNA (or its complement, cDNA) are known in the art.
  • the level of an mRNA may correlate with the level of the encoded protein, so that detection of the mRNA level may be used to determine whether the level of the protein is, for example, elevated prior to treatment, or has been reduced following treatment.
  • the level of the protein is determined.
  • Any method of detecting the level of a protein in a sample is contemplated.
  • One skilled in the art can select a suitable method depending on the type of sample being analyzed and the identity and number of proteins being detected.
  • Nonlimiting exemplary such methods include immunohistochemistry, ELISA, Western blotting, multiplex analyte detection (using, for example, Luminex technology), mass spectrometry, etc.
  • any method of detecting the level of an mRNA in a sample is contemplated.
  • One skilled in the art can select a suitable method depending on the type of sample being analyzed and the identity and number of mRNAs being detected.
  • Nonlimiting exemplary such methods include RT-PCR, quantitative RT-PCR and microarray-based methods, etc.
  • any method of determining the levels of CD16+ and/or CD16 ⁇ monocytes is contemplated.
  • One skilled in the art can select a suitable method depending on the type of sample being analyzed.
  • Nonlimiting exemplary methods of determining the levels of CD16+ and/or CD16 ⁇ monocytes include methods provided by commercial kits, such as CD16+Moncyte Isolation Kit (Miltenyl Biotec, Bergisch Gladbach, Germany).
  • the method comprises reducing at least one, at least two, at least three, or four factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in a subject comprising administering an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding.
  • the method comprises reducing at least one, at least two, at least three, or four factors selected from IL-6, IL-1 ⁇ , TNF- ⁇ , and CXCL10.
  • native mature sequences For exemplary mature human IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-9, and MMP-2 are shown in Table 8 (Table of Sequences; SEQ ID NOs: 96 to 108, respectively). Additional native mature sequences may also exist. In some embodiments, native mature sequences have 1 to 10 or more amino acids deleted from the amino terminus of the mature sequences shown in Table 8. In some embodiments, native mature sequences have one or more amino acid additions, deletions, and/or substitutions in relative to the mature sequences shown in Table 8. All of the native mature forms of each factor are intended to be encompassed herein.
  • IL-6 IL-1 ⁇
  • IL-8 CCL2
  • CXCL10 TNF- ⁇
  • CCL7 CXCL5
  • CXCL9 CXCL9
  • MMP-7 MMP-2
  • MMP-9 MMP-9
  • Exemplary conditions that are associated with elevated levels of one or more of those factors include, but are not limited to, rheumatoid arthritis, juvenile idiopathic arthritis, Castleman's disease, psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, ulcerative colitis, lupus erythematosus, and inflammatory bowel disease.
  • the antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding is selected from huAb1 to huAb16, described herein. In some embodiments, the antibody is huAb1.
  • a method of reducing the level of IL-6 in a subject comprising administering to the subject an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding. Reducing the level of IL-6 is beneficial, in some embodiments, in the treatment of a condition associated with elevated IL-6, such as rheumatoid arthritis, juvenile idiopathic arthritis, and Castleman's disease.
  • a method of reducing the level of TNF- ⁇ in a subject comprising administering to the subject an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding.
  • Reducing the level of TNF- ⁇ is beneficial, in some embodiments, in the treatment of a condition associated with elevated TNF- ⁇ , such as rheumatoid arthritis, juvenile idiopathic arthritis, psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, and ulcerative colitis.
  • a method of reducing the level of IL-1 ⁇ in a subject comprising administering to the subject an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding.
  • the antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding may be selected from huAb1 to huAb16, described herein. In any of the embodiments herein, the antibody may be huAb1.
  • a method comprises reducing IL-6 and IL-1 ⁇ . In some embodiments, the method comprises reducing IL-6 and TNF- ⁇ . In some embodiments, a method comprises reducing IL-6 and CXCL10. In some embodiments, a method comprises reducing IL-1 ⁇ and TNF- ⁇ . In some embodiments, a method comprises reducing IL-1 ⁇ and CXCL10. In some embodiments, a method comprises reducing TNF- ⁇ and CXCL10. In some embodiments, a method comprises reducing IL-6, IL-1 ⁇ , and TNF- ⁇ . In some embodiments, a method comprises reducing IL-6, IL-1 ⁇ , and CXCL10.
  • a method comprises reducing IL-6, TNF- ⁇ , and CXCL10. In some embodiments, a method comprises reducing TNF- ⁇ , IL-1 ⁇ , and CXCL10. In some embodiments, a method comprises reducing IL-6, IL-1 ⁇ , TNF- ⁇ , and CXCL10.
  • Methods of treating an inflammatory condition comprising administering to a subject with an inflammatory condition an effective amount of an antibody that binds CSF1R and blocks CSF 1 and IL-34 ligand binding.
  • a method of treating an inflammatory condition comprises reducing the level of one or more factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in a subject with an inflammatory condition, comprising administering an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding.
  • the method comprises reducing at least one, at least two, at least three, or four factors selected from IL-6, IL-1 ⁇ , TNF- ⁇ , and CXCL10.
  • the antibody that binds CSF1R and blocks CSF 1 and IL-34 ligand binding is selected from huAb1 to huAb16, described herein. In some embodiments, the antibody is huAb1.
  • Nonlimiting exemplary inflammatory conditions include rheumatoid arthritis, juvenile idiopathic arthritis, Castleman's disease, psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, ulcerative colitis, lupus erythematosus, inflammatory bowel disease, inflammatory arthritis, and CD16+ disorders.
  • Methods of treating inflammatory arthritis comprising administering to a subject with an inflammatory arthritis an effective amount of an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding.
  • a method of treating inflammatory arthritis comprises reducing the level of one or more factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in a subject with inflammatory arthritis, comprising administering an effective amount of an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding.
  • the method comprises reducing at least one, at least two, at least three, or four factors selected from IL-6, IL-1 ⁇ , TNF- ⁇ , and CXCL10.
  • the antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding is selected from huAb1 to huAb16, described herein. In some embodiments, the antibody is huAb1.
  • administering in addition to reducing the level of one or more factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9, administering to a subject with inflammatory arthritis an effective amount of an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding, such as huAb1 to huAb16, reduces inflammation, reduces pannus formation, reduces cartilage damage, reduces bone resorption, reduces macrophage numbers in the joints, reduces autoantibody formation, and/or reduces bone loss.
  • an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding such as huAb1 to huAb16
  • administering in addition to reducing the level of one or more factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9, administering to a subject with inflammatory arthritis an effective amount of an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding, such as huAb1 to huAb16, reduces inflammation.
  • Reducing inflammation comprises reducing erythrocyte sedimentation rate and/or reducing the levels of C-reactive proteins in blood.
  • the erythrocyte sedimentation rate increases, possibly due to increased levels of fibrinogen in the blood.
  • the erythrocyte sedimentation rate may be determined by any method in the art, including, but not limited to, calculating the rate by measuring the change in height of anticoagulated erythrocytes in one hour in a Westergren tube. See also Procedures for the Erythrocyte Sedimentation Rate Test; Approved Standard—Fifth Edition . CLSI document H02-A5. Wayne, Pa.: Clinical and Laboratory Standards Institute; 2011. Levels of C-reactive protein in blood may be determined by any methods in the art, including but not limited to using the RAPITEX® CRP test kit (Siemens).
  • Reducing inflammation comprises reducing peripheral edema, which is tissue swelling due to the buildup of fluids.
  • Peripheral edema may occur, in some instances, in the ankles, feet, legs, and/or calves of a subject with rheumatoid arthritis.
  • Reducing inflammation comprises reducing infiltration of inflammatory cells in the synovium of one or more affected joints. Synovial fluid may be collected, in some embodiments, by athrocentesis.
  • administering in addition to reducing the level of one or more factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9, administering to a subject with inflammatory arthritis an effective amount of an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding, such as huAb1 to huAb16, reduces pannus formation.
  • Reducing pannus formation comprises reducing infiltration of pannus into cartilage and/or subchondrial bone, and/or reducing hard tissue destruction resulting from pannus infiltration.
  • Pannus formation can be measured by any method in the art, including, but not limited to, imaging one or more affected joints.
  • Nonlimiting exemplary imaging techniques for detecting pannus formation include magnetic resonance imaging (MRI), computed tomography (CT) scan, arthroscopy, ultrasonography, duplex ultrasonography, and power doppler imaging.
  • MRI magnetic resonance imaging
  • CT computed tomography
  • arthroscopy arthroscopy
  • ultrasonography ultrasonography
  • duplex ultrasonography duplex ultrasonography
  • power doppler imaging power doppler imaging.
  • the progression of pannus formation is slowed following administration of the antibody and/or during a particular time interval during which the subject is undergoing treatment with the antibody.
  • the treatment may be a single dose or multiple doses.
  • administering in addition to reducing the level of one or more factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9, administering to a subject with inflammatory arthritis an effective amount of an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding, such as huAb1 to huAb16, reduces cartilage damage.
  • Reducing cartilage damage comprises reducing chondrocyte loss, reducing collagen disruption, and/or reducing cartilage loss.
  • Cartilage damage can be measured by any method in the art, including, but not limited to, imaging one or more affected joints.
  • Nonlimiting exemplary imaging techniques for detecting cartilage damage include MRI, CT scan, arthroscopy, and x-ray imaging.
  • the progression of cartilage damage is slowed following administration of the antibody and/or during a particular time interval during which the subject is undergoing treatment with the antibody.
  • the treatment may be a single dose or multiple doses.
  • administering in addition to reducing the level of one or more factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9, administering to a subject with inflammatory arthritis an effective amount of an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding, such as huAb1 to huAb16, reduces bone resorption.
  • Reducing bone resorption comprises reducing the number of osteoclasts in joints affected by rheumatoid arthritis.
  • bone resorption may be measured by determining the level of TRAP5b in plasma from the subject, wherein an elevated level of TRAP5b indicates elevated bone resorption in the subject.
  • a reduced level of TRAP5b indicates a reduction in bone resorption.
  • TRAP5b levels may be determined, in certain instances, before and after treatment with an antibody that binds CSF1R, and/or may be determined periodically throughout the course of treatment to monitor the effectiveness of the treatment in reducing bone loss.
  • TRAP5b levels may be determined using any method in the art, including, but not limited to, ELISA (including FAICEA, or fragments absorbed immunocapture enzymatic assay; see, e.g., Quidel® TRAP5b assay, TECOmedical Group, Sissach, Switzerland).
  • ELISA including FAICEA, or fragments absorbed immunocapture enzymatic assay; see, e.g., Quidel® TRAP5b assay, TECOmedical Group, Sissach, Switzerland).
  • administering in addition to reducing the level of one or more factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9, administering to a subject with inflammatory arthritis an effective amount of an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding, such as huAb1 to huAb16, reduces bone loss.
  • an effective amount of an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding such as huAb1 to huAb16
  • Bone loss may be determined using any method in the art, including, but not limited to, x-ray imaging, MRI, CT, bone densitometry, single and dual photon absorptiometry (SPA, DPA), single and dual energy x-ray absorptiometry (SXA, DXA), ultrasonography, scintigraphy, and by measuring levels of serum markers of bone formation and resorption.
  • x-ray imaging MRI, CT, bone densitometry
  • SPA single and dual photon absorptiometry
  • SXA single and dual energy x-ray absorptiometry
  • ultrasonography scintigraphy
  • Serum osteocalcin Serum and urinary hydroxyproline (Hyp) Serum total alkaline Urinary total pyridinoline (Pyr) phosphatase (ALP) Serum bone specific alkaline Urinary total deoxypyridinoline (dPyr) phosphatase (BSAP, BALP, or B-ALP) Serum procollagen I Urinary free pyridinoline (f-Pyr, also carboxyterminal propeptide known as Pyrilinks ® (Metra (PICP) Biosystems)) Serum procollagen type 1 Urinary free deoxypyridinoline (f-dPyr, N-terminal propeptide also known as Pyrilinks-D ®) (PINP) Bone sialoprotein Serum and urinary collagen type I cross- linked N-telopeptide (NTx, also referred to as Osteomark) Serum osteocalcin (OC) Serum and urinary hydroxyproline (Hyp) Se
  • the progression of bone loss is slowed following administration of the antibody and/or during a particular time interval during which the subject is undergoing treatment with the antibody.
  • the treatment may be a single dose or multiple doses.
  • administering to a subject with inflammatory arthritis an effective amount of an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding, such as huAb1 to huAb16, reduces autoantibody levels.
  • the levels of autoantibodies may be determined by any method in the art. In some embodiments, autoantibody levels are determined by the level of rheumatoid factor (RF) and/or anti-citrullinated protein antibodies (ACPA) and/or anti-nuclear antibodies (ANA).
  • administering in addition to reducing the level of one or more factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9, administering to a subject with inflammatory arthritis an effective amount of an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding, such as huAb1 to huAb16, substantially reduces the number of monocyte lineage cells, such as macrophages and/or CD16+ monocytes, in joints (including synovial fluid) affected by the inflammatory arthritis.
  • an effective amount of an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding such as huAb1 to huAb16
  • administering in addition to reducing the level of one or more factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9, administering to a subject with inflammatory arthritis an effective amount of an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding, such as huAb1 to huAb16, substantially reduces the number of CD16+ monocytes.
  • the subject has an autoimmune condition selected from rheumatoid arthritis and SLE (lupus).
  • the number of CD16 ⁇ monocytes is substantially unchanged.
  • CD16+ monocytes are reduced to a greater extent than CD16 ⁇ monocytes are reduced when an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding is administered to the subject.
  • CD16+ monocytes are reduced by at least 20%, at least 30%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90%.
  • CD16 ⁇ monocytes are reduced by less than 30%, less than 20%, or less than 10%.
  • the CD16+ monocytes are CD16+ peripheral blood monocytes.
  • the CD16 ⁇ monocytes are CD16 ⁇ peripheral blood monocytes.
  • methods of treating a CD16+ disorder comprising administering to a subject with a CD16+ disorder an effective amount of an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding, such as huAb1 to huAb16, wherein the antibody reduces the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9.
  • an antibody that binds CSF1R and blocks CSF1 and IL-34 ligand binding such as huAb1 to huAb16
  • the antibody reduces the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven,
  • Nonlimiting exemplary CD16+ disorders include rheumatoid arthritis, juvenile idiopathic arthritis, Castleman's disease, psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, ulcerative colitis, lupus erythematosus, and inflammatory bowel disease.
  • a subject with a CD16+ disorder has an elevated level of CD16+ monocytes, compared to the level of CD16+ monocytes in a healthy individual or a pool of healthy individuals.
  • a subject with a CD16+ disorder has an elevated level of CD16+ monocytes, compared to the subject's CD16+ monocyte level prior to developing the CD16+ disorder (for example, in some embodiments, substantially prior to developing any symptoms of the CD16+ disorder such that the subject would, in retrospect, be considered “healthy” at the time).
  • a method comprises determining the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in the subject.
  • an elevated level of at least one of the factors in the subject indicates that the subject may benefit from the antibody that binds CSF1R.
  • the subject has a CD16+ disorder.
  • the subject has rheumatoid arthritis.
  • the subject has an elevated level of CD16+ monocytes.
  • methods of predicting responsiveness in a subject suffering from an inflammatory condition to an antibody that binds CSF1R wherein the antibody blocks binding of CSF1 to CSF1R and blocks binding of IL-34 to CSF1R (such as huAb1 to huAb16) are provided.
  • a method comprises determining the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in the subject.
  • an elevated level of at least one of the factors in the subject indicates that the subject is more likely to respond to the antibody that binds CSF1R.
  • the subject has a CD16+ disorder.
  • the subject has rheumatoid arthritis.
  • the subject has an elevated level of CD16+ monocytes.
  • the subject has a condition that is resistant to methotrexate (e.g., the subject is methotrexate inadequate responder).
  • a condition that is resistant to methotrexate such as a subject who is a methotrexate inadequate responder, may have previously responded to methotrexate, but may have become resistant to methotrexate, or the subject may have never responded to methotrexate.
  • Resistance to methotrexate means that aspects of the condition that would be expected to improve following a standard dose of methotrexate do not improve, and/or improvement only occurs if greater than a standard dose of methotrexate is administered.
  • a methotrexate inadequate responder has experienced, or is experiencing, an inadequate response to methotrexate after receiving a standard dose for at least two weeks, at least three weeks, at least four weeks, at least six weeks, or at least twelve weeks.
  • a “standard” dose is determined by a medical professional, and may depend on the subject's age, weight, healthy history, severity of disease, the frequency of dosing, etc.
  • the subject is a TNF inhibitor inadequate responder.
  • a subject who is a TNF inhibitor inadequate responder may have previously responded to a TNF inhibitor, but may have become less responsive to the TNF inhibitor, or the subject may have never responded to the TNF inhibitor.
  • Inadequate response to a TNF inhibitor means that aspects of the condition that would be expected to improve following a standard dose of the TNF inhibitor do not improve, and/or improvement only occurs if greater than a standard dose is administered.
  • a TNF inhibitor inadequate responder has experienced, or is experiencing, an inadequate response to the TNF inhibitor after receiving a standard dose for at least two weeks, at least three weeks, at least four weeks, at least six weeks, or at least twelve weeks.
  • a “standard” dose is determined by a medical professional, and may depend on the subject's age, weight, healthy history, severity of disease, the frequency of dosing, etc.
  • a TNF inhibitor inadequate responder has experienced, or is experiencing, an inadequate response to a TNF inhibitor selected from infliximab, adalimumab, certolizumab pegol, golimumab, and etanercept.
  • a method comprises administering to the methotrexate inadequate responder an antibody that binds CSF1R, wherein the antibody blocks binding of CSF1 to CSF1R and blocks binding of IL-34 to CSF1R, such as huAb1 to huAb16.
  • the inadequate responder has a CD16+ disorder.
  • the CD16+ disorder is selected from rheumatoid arthritis, juvenile idiopathic arthritis, Castleman's disease, psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, and ulcerative colitis, lupus erythematosus, and inflammatory bowel disease.
  • the CD16+ disorder is rheumatoid arthritis.
  • the antibody substantially reduces the number of CD16+ monocytes. In some embodiments, the number of CD16 ⁇ monocytes are substantially unchanged following administration of the antibody.
  • the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in the inadequate responder is reduced following administration of the antibody.
  • the inadequate responder has an elevated level of CD16+ monocytes, for example, as compared to the level of CD16+ monocytes in a healthy individual or pool of healthy individuals.
  • the antibody substantially reduces the number of CD16+ monocytes.
  • the number of CD16 ⁇ monocytes are substantially unchanged following administration of the antibody.
  • a method comprises administering to the TNF inhibitor inadequate responder an antibody that binds CSF1R, wherein the antibody blocks binding of CSF1 to CSF1R and blocks binding of IL-34 to CSF1R, such as huAb1 to huAb16.
  • the inadequate responder has a CD16+ disorder.
  • the CD16+ disorder is selected from rheumatoid arthritis, juvenile idiopathic arthritis, Castleman's disease, psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, and ulcerative colitis, lupus erythematosus, and inflammatory bowel disease.
  • the CD16+ disorder is rheumatoid arthritis.
  • the antibody substantially reduces the number of CD16+ monocytes. In some embodiments, the number of CD16 ⁇ monocytes are substantially unchanged following administration of the antibody.
  • the level of at least one, at least two, at least three, or at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten factors selected from IL-6, IL-1 ⁇ , IL-8, CCL2, CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, MMP-7, MMP-2, and MMP-9 in the inadequate responder is reduced following administration of the antibody.
  • the inadequate responder has an elevated level of CD16+ monocytes, for example, as compared to the level of CD16+ monocytes in a healthy individual or pool of healthy individuals.
  • the antibody substantially reduces the number of CD16+ monocytes.
  • the number of CD16 ⁇ monocytes are substantially unchanged following administration of the antibody.
  • anti-CSF1R antibodies may be administered in vivo by various routes, including, but not limited to, oral, intra-arterial, parenteral, intranasal, intramuscular, intracardiac, intraventricular, intratracheal, buccal, rectal, intraperitoneal, intradermal, topical, transdermal, and intrathecal, or otherwise by implantation or inhalation.
  • the subject compositions may be formulated into preparations in solid, semi-solid, liquid, or gaseous forms; including, but not limited to, tablets, capsules, powders, granules, ointments, solutions, suppositories, enemas, injections, inhalants, and aerosols.
  • a nucleic acid molecule encoding an anti-CSF1R antibody may be coated onto gold microparticles and delivered intradermally by a particle bombardment device, or “gene gun,” as described in the literature (see, e.g., Tang et al., Nature 356:152-154 (1992)).
  • a particle bombardment device or “gene gun,” as described in the literature (see, e.g., Tang et al., Nature 356:152-154 (1992)).
  • the appropriate formulation and route of administration may be selected according to the intended application.
  • compositions comprising anti-CSF1R antibodies are provided in formulations with a wide variety of pharmaceutically acceptable carriers (see, e.g., Gennaro, Remington: The Science and Practice of Pharmacy with Facts and Comparisons: Drugfacts Plus, 20 th ed. (2003); Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7 th ed., Lippencott Williams and Wilkins (2004); Kibbe et al., Handbook of Pharmaceutical Excipients, 3 rd ed., Pharmaceutical Press (2000)).
  • Various pharmaceutically acceptable carriers which include vehicles, adjuvants, and diluents, are available.
  • Non-limiting exemplary carriers include saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof.
  • compositions comprising anti-CSF1R antibodies may be formulated for injection, including subcutaneous administration, by dissolving, suspending, or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids, or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
  • the compositions may be formulated for inhalation, for example, using pressurized acceptable propellants such as dichlorodifluoromethane, propane, nitrogen, and the like.
  • compositions may also be formulated, in various embodiments, into sustained release microcapsules, such as with biodegradable or non-biodegradable polymers.
  • a non-limiting exemplary biodegradable formulation includes poly lactic acid-glycolic acid polymer.
  • a non-limiting exemplary non-biodegradable formulation includes a polyglycerin fatty acid ester. Certain methods of making such formulations are described, for example, in EP 1 125 584 A1.
  • compositions contained in the unit dosage may comprise saline, sucrose, or the like; a buffer, such as phosphate, or the like; and/or be formulated within a stable and effective Ph range.
  • the composition may be provided as a lyophilized powder that may be reconstituted upon addition of an appropriate liquid, for example, sterile water.
  • the composition comprises one or more substances that inhibit protein aggregation, including, but not limited to, sucrose and arginine.
  • a composition of the invention comprises heparin and/or a proteoglycan.
  • compositions are administered in an amount effective for treatment or prophylaxis of the specific indication.
  • the therapeutically effective amount is typically dependent on the weight of the subject being treated, his or her physical or health condition, the extensiveness of the condition to be treated, or the age of the subject being treated.
  • anti-CSF1R antibodies may be administered in an amount in the range of about 10 ⁇ g/kg body weight to about 100 mg/kg body weight per dose. In some embodiments, anti-CSF1R antibodies may be administered in an amount in the range of about 50 ⁇ g/kg body weight to about 5 mg/kg body weight per dose. In some embodiments, anti-CSF1R antibodies may be administered in an amount in the range of about 100 ⁇ g/kg body weight to about 10 mg/kg body weight per dose.
  • anti-CSF1R antibodies may be administered in an amount in the range of about 100 ⁇ g/kg body weight to about 20 mg/kg body weight per dose. In some embodiments, anti-CSF1R antibodies may be administered in an amount in the range of about 0.5 mg/kg body weight to about 20 mg/kg body weight per dose.
  • the anti-CSF1R antibody compositions may be administered as needed to subjects. Determination of the frequency of administration may be made by persons skilled in the art, such as an attending physician based on considerations of the condition being treated, age of the subject being treated, severity of the condition being treated, general state of health of the subject being treated and the like.
  • an effective dose of an anti-CSF1R antibody is administered to a subject one or more times.
  • an effective dose of an anti-CSF1R antibody is administered to the subject once a month, less than once a month, such as, for example, every two months or every three months.
  • an effective dose of an anti-CSF1R antibody is administered more than once a month, such as, for example, every three weeks, every two weeks or every week.
  • An effective dose of an anti-CSF1R antibody is administered to the subject at least once.
  • the effective dose of an anti-CSF1R antibody may be administered multiple times, including for periods of at least a month, at least six months, or at least a year.
  • Anti-CSF1R antibodies may be administered alone or with other modes of treatment. They may be provided before, substantially contemporaneous with, or after other modes of treatment, for example, surgery, chemotherapy, radiation therapy, or the administration of a biologic, such as another therapeutic antibody.
  • a biologic such as another therapeutic antibody.
  • anti-CSF1R antibodies may be administered with other therapeutic agents, for example, hydroxychloroquine (Plaquenil®); corticosteroids, such as prednisone, methylprednisone, and prednisolone; immunosuppressants, such as cyclophosphamide (Cytoxan®), azathioprine (Imuran®, Azasan®), mycophenolate (Cellcept®), leflunomide (Arava®), methotrexate (TrexallTM), and belimumab (Benlysta®).
  • hydroxychloroquine Plaquenil®
  • corticosteroids such as prednisone, methylprednisone, and prednisolone
  • immunosuppressants such as cyclophosphamide (Cytoxan®), azathioprine (Imuran®, Azasan®), mycophenolate (Cellcept®), leflunomide (A
  • anti-CSF1R antibodies may be administered with other therapeutic agents, for example, interferon alpha; interferon beta; prednisone; anti-alpha4 integrin antibodies such as Tysabri®; anti-CD20 antibodies such as Rituxan®; FTY720 (fingolimod; Gilenya®); and cladribine (Leustatin®).
  • other therapeutic agents for example, interferon alpha; interferon beta; prednisone; anti-alpha4 integrin antibodies such as Tysabri®; anti-CD20 antibodies such as Rituxan®; FTY720 (fingolimod; Gilenya®); and cladribine (Leustatin®).
  • FIGS. 1 and 2 The sequences for each of the humanized heavy chain variable regions and humanized light chain variable regions, aligned with the sequences of the parental chimeric antibody variable regions and the sequences of the human acceptor variable framework regions are shown in FIGS. 1 (heavy chains) and 2 (light chains).
  • the changes in humanized variable region sequences relative to the human acceptor variable framework region sequences are boxed.
  • Each of the CDRs for each of the variable regions is shown in a boxed region, and labeled as “CDR” above the boxed sequences.
  • Table 8 shows the full sequences for the humanized heavy chains and humanized light chains of antibodies huAb1 to huAb16.
  • the name and SEQ ID Nos of the humanized heavy chain and humanized light chain of each of those antibodies is shown in Table 3.
  • the 16 humanized antibodies were tested for binding to human, cynomolgus monkey, and mouse CSF1R ECD, as described previously. See, e.g., PCT Publication No. WO 2011/140249.
  • the antibodies were found to bind to both human and cynomolgus monkey CSF1R ECD, but not to mouse CSF1R ECD.
  • the humanized antibodies were also found to block binding of CSF1 and IL-34 to both human and mouse CSF1R and to inhibit CSF 1-induced and IL-34-induced phosphorylation of human CSF1R expressed in CHO cells. See, e.g., PCT Publication No. WO 2011/140249.
  • the k a , k d , and K D for binding to human CSF1R ECD were previously determined and are shown in Table 4. See, e.g., PCT Publication No. WO 2011/140249.
  • HuAb1 Alters Cytokine and Certain Matrix Metalloproteinase Production in Synovial Biopsy Explants
  • Synovial tissue samples were obtained from the joints of rheumatoid arthritis patients. Patients had clinically active disease and tissue was obtained from clinically active joints. All patients provided written informed consent and these studies were approved by the Medical Ethics Committee of the Academic Medical Center (AMC) at the University of Amsterdam. The clinical characteristics of the six patients from whom biopsy samples were taken are shown in Table 5.
  • Synovial biopsy samples with a volume of approximately 5 mm 3 were cultured in triplicate in complete medium comprising DMEM (Life Technologies, Grand Island, N.Y.) with 2 Mm L-glutamine, 100 U/ml penicillin, 50 mg/ml gentamicin, 20 Mm HEPES buffer, and 10% FCS. Cultures were performed at 37° C. in a 5% CO 2 /95% air-humidified environment. Synovial samples were cultured for 4 days in the absence or presence of increasing concentrations of huAb1 or control IgG4 antibody ET904 (Eureka Therapeutics, Emeryville, Calif.). Cell-free supernatants were collected and stored at ⁇ 80° C. in two separate aliquots.
  • IL-6 IL-6 by ELISA.
  • the other aliquot was evaluated for multiplex analysis of cytokine and matrix metalloproteinase production using Luminex® technology (Millipore, Billerica, Mass.; Catalog Nos. MPXHCYTO-60K-08, MPXHCYP2-62K-02, MPXHCYP3-63K-04, HMMP2-55K-03).
  • the three tissue fragments in each culture condition were pooled, snap-frozen and preserved for Mrna expression analysis.
  • IL-6 production was reduced in all samples after culturing for 4 days in the presence of 1 ⁇ g/ml or 10 ⁇ g/ml huAb1, as compared to incubation in the same concentration of control antibody.
  • IL-6 production was reduced in four of the six samples after culturing for 4 days in the presence of 0.1 ⁇ g/ml huAb1, as compared to incubation in 0.1 ⁇ g/ml of control antibody.
  • FIG. 3 shows a plot of the reduction in IL-6 production in the four samples after culturing for 4 days in the presence of 1 ⁇ g/ml control antibody or 1 ⁇ g/ml huAb1.
  • FIG. 4 shows the results of the multiplex analysis after culturing four of the synovial biopsy explants in 1 ⁇ g/ml huAb1 or control antibody for 4 days.
  • Levels of IL-6, IL-1 ⁇ , IL-8, CCL2 (also referred to as MCP-1), CXCL10, TNF- ⁇ , CCL7, CXCL5, CXCL9, CXCL6, and MMP-9 were reduced in all four samples after incubation with 1 ⁇ g/ml huAb1, relative to incubation with 1 ⁇ g/ml control antibody.
  • Levels of MMP-7 were reduced in the two samples with measurable levels of MMP-7 in the control antibody-treated groups.
  • Table 7 below shows the results of the multiplex analysis for the four synovial biopsy explants shown in FIG. 4 .
  • the average cytokine level in the four explants is shown after incubation in medium alone, 0.1 ⁇ g/ml, 1 ⁇ g/ml, or 10 ⁇ g/ml IgG4 control, or 0.1 ⁇ g/ml, 1 ⁇ g/ml, or 10 ⁇ g/ml huAb1.
  • MMP-2 and MMP-9 levels after culturing two additional synovial biopsy explants in the presence of medium alone, 0.1 ⁇ g/ml, 1 ⁇ g/ml, or 10 ⁇ g/ml IgG4 control, or 0.1 ⁇ g/ml, 1 ⁇ g/ml, or 10 ⁇ g/ml huAb1 was performed substantially as described above.
  • Table 8 below shows the results of the multiplex analysis for all six synovial biopsy explants tested for MMP-2 and MMP-9 levels.
  • MMP-7 levels after culturing one additional synovial biopsy explants in the presence of medium alone, 0.1 ⁇ g/ml, 1 ⁇ g/ml, or 10 ⁇ g/ml IgG4 control, or 0.1 ⁇ g/ml, 1 ⁇ g/ml, or 10 ⁇ g/ml huAb1 was performed substantially as described above.
  • Table 9 below shows the results of the multiplex analysis for all five synovial biopsy explants tested for MMP-7 levels.
  • Table 10 provides certain sequences discussed herein. All polypeptide and antibody sequences are shown without leader sequences, unless otherwise indicated.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Rheumatology (AREA)
  • Biochemistry (AREA)
  • Mycology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Hematology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Endocrinology (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Cell Biology (AREA)
  • Rehabilitation Therapy (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Pain & Pain Management (AREA)
US14/014,446 2012-08-31 2013-08-30 Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r) Abandoned US20140079699A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US14/014,446 US20140079699A1 (en) 2012-08-31 2013-08-30 Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
US15/154,822 US10221243B2 (en) 2012-08-31 2016-05-13 Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US16/256,311 US10822421B2 (en) 2012-08-31 2019-01-24 Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US17/026,853 US20210107986A1 (en) 2012-08-31 2020-09-21 Methods of Treating Conditions with Antibodies that Bind Colony Stimulating Factor 1 Receptor (CSF1R)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201261695641P 2012-08-31 2012-08-31
US201361767989P 2013-02-22 2013-02-22
US201361778706P 2013-03-13 2013-03-13
US14/014,446 US20140079699A1 (en) 2012-08-31 2013-08-30 Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/154,822 Division US10221243B2 (en) 2012-08-31 2016-05-13 Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (CSF1R)

Publications (1)

Publication Number Publication Date
US20140079699A1 true US20140079699A1 (en) 2014-03-20

Family

ID=50184395

Family Applications (4)

Application Number Title Priority Date Filing Date
US14/014,446 Abandoned US20140079699A1 (en) 2012-08-31 2013-08-30 Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
US15/154,822 Active US10221243B2 (en) 2012-08-31 2016-05-13 Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US16/256,311 Active US10822421B2 (en) 2012-08-31 2019-01-24 Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US17/026,853 Pending US20210107986A1 (en) 2012-08-31 2020-09-21 Methods of Treating Conditions with Antibodies that Bind Colony Stimulating Factor 1 Receptor (CSF1R)

Family Applications After (3)

Application Number Title Priority Date Filing Date
US15/154,822 Active US10221243B2 (en) 2012-08-31 2016-05-13 Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US16/256,311 Active US10822421B2 (en) 2012-08-31 2019-01-24 Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US17/026,853 Pending US20210107986A1 (en) 2012-08-31 2020-09-21 Methods of Treating Conditions with Antibodies that Bind Colony Stimulating Factor 1 Receptor (CSF1R)

Country Status (14)

Country Link
US (4) US20140079699A1 (ru)
EP (2) EP2890398A4 (ru)
JP (3) JP2015533787A (ru)
KR (2) KR20200140400A (ru)
CN (2) CN104684582A (ru)
AU (2) AU2013308635A1 (ru)
BR (1) BR112015004426A2 (ru)
CA (1) CA2882804A1 (ru)
HK (1) HK1212214A1 (ru)
IL (1) IL237351B (ru)
RU (1) RU2718751C2 (ru)
SG (2) SG10201906328RA (ru)
TW (2) TWI713436B (ru)
WO (1) WO2014036357A1 (ru)

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9192667B2 (en) 2013-04-22 2015-11-24 Hoffmann-La Roche Inc. Method of treating cancer by administering CSF-1R antibodies and a TLR9 agonist
US9200075B2 (en) 2010-05-04 2015-12-01 Five Prime Therapeutics, Inc. Nucleic acids encoding antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US9765147B2 (en) 2014-10-29 2017-09-19 Five Prime Therapeutics, Inc. Anti-CSFR1 antibody and anti PD-1 antibody combination therapy for cancer
US10040858B2 (en) 2014-12-22 2018-08-07 Five Prime Therapeutics, Inc. Anti-CSF1R antibodies for treating PVNS
US10077314B1 (en) 2009-12-10 2018-09-18 Hoffmann-La Roche Inc. Antibodies against human CSF-1R and uses thereof
US10336830B2 (en) 2011-12-15 2019-07-02 Hoffmann-La Roche Inc. Antibodies against human CSF-1R and uses thereof
WO2020010177A1 (en) 2018-07-06 2020-01-09 Kymera Therapeutics, Inc. Tricyclic crbn ligands and uses thereof
US10874743B2 (en) 2017-12-26 2020-12-29 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US10975153B2 (en) 2014-06-23 2021-04-13 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US10982001B2 (en) 2012-05-11 2021-04-20 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US11117889B1 (en) 2018-11-30 2021-09-14 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11267857B2 (en) * 2017-02-27 2022-03-08 Shattuck Labs, Inc. CSF1R-based chimeric proteins
US11358948B2 (en) 2017-09-22 2022-06-14 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
US11421034B2 (en) 2017-09-13 2022-08-23 Five Prime Therapeutics, Inc. Combination anti-CSF1R and anti-PD-1 antibody combination therapy for pancreatic cancer
US11485750B1 (en) 2019-04-05 2022-11-01 Kymera Therapeutics, Inc. STAT degraders and uses thereof
US11485743B2 (en) 2018-01-12 2022-11-01 Kymera Therapeutics, Inc. Protein degraders and uses thereof
US11498968B2 (en) 2016-12-22 2022-11-15 Hoffmann-La Roche Inc. Treatment of tumors with an anti-CSF-1R antibody in combination with an anti-PD-L1 antibody after failure of anti-PD-L1/PD1 treatment
US11512080B2 (en) 2018-01-12 2022-11-29 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
US11512133B2 (en) 2013-09-12 2022-11-29 Hoffmann-La Roche Inc. Methods for treating colon cancer or inhibiting cell proliferation by administering a combination of antibodies against human CSF-1R and antibodies against human PD-L1
US11542335B2 (en) 2016-08-25 2023-01-03 Hoffmann-La Roche Inc. Method of treating cancer in a patient by administering an antibody which binds colony stimulating factor-1 receptor (CSF-1R)
US11559583B2 (en) 2015-04-13 2023-01-24 Five Prime Therapeutics, Inc. Anti-CSF1R antibody and agonistic anti-CD40 antibody combination therapy for cancer
US11591332B2 (en) 2019-12-17 2023-02-28 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11623932B2 (en) 2017-09-22 2023-04-11 Kymera Therapeutics, Inc. Protein degraders and uses thereof
US11679109B2 (en) 2019-12-23 2023-06-20 Kymera Therapeutics, Inc. SMARCA degraders and uses thereof
US11685750B2 (en) 2020-06-03 2023-06-27 Kymera Therapeutics, Inc. Crystalline forms of IRAK degraders
US11707457B2 (en) 2019-12-17 2023-07-25 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11773103B2 (en) 2021-02-15 2023-10-03 Kymera Therapeutics, Inc. IRAK4 degraders and uses thereof
US11857535B2 (en) 2020-07-30 2024-01-02 Kymera Therapeutics, Inc. Methods of treating mutant lymphomas
US11932624B2 (en) 2020-03-19 2024-03-19 Kymera Therapeutics, Inc. MDM2 degraders and uses thereof

Families Citing this family (164)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2718751C2 (ru) 2012-08-31 2020-04-14 Файв Прайм Терапьютикс, Инк. Способы лечения патологических состояний антителами, которые связываются с рецептором колониестимулирующего фактора 1 (csf1r)
NZ722326A (en) 2013-12-24 2019-09-27 Bristol Myers Squibb Co Tricyclic compounds as anticancer agents
WO2015160786A1 (en) * 2014-04-14 2015-10-22 Brigham And Women's Hospital, Inc. Method of diagnosing, prognosing, and treating lupus nephritis
EA037006B1 (ru) 2014-06-06 2021-01-26 Бристол-Майерс Сквибб Компани Антитела к индуцируемому глюкокортикоидами рецептору фактора некроза опухолей (gitr) и их применения
EP3151820A4 (en) 2014-06-06 2017-11-22 Flexus Biosciences, Inc. Immunoregulatory agents
JP6769948B2 (ja) * 2014-07-17 2020-10-14 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル 移植拒絶の予防及び自己免疫疾患の治療に使用するための単離されたインターロイキン−34ポリペプチド
MX2017001293A (es) * 2014-07-28 2017-05-23 Nogra Pharma Ltd Metodos y composiciones para diagnosticar y tratar trastornos inflamatorios del intestino.
CN107074976B (zh) * 2014-11-04 2020-10-09 北京韩美药品有限公司 同时阻断b7/cd28和il6/il6r/gp130信号通路的重组融合蛋白
EA201790806A1 (ru) 2014-11-05 2017-11-30 Флексус Байосайенсиз, Инк. Иммунорегулирующие средства
AR102537A1 (es) 2014-11-05 2017-03-08 Flexus Biosciences Inc Agentes inmunomoduladores
UY36390A (es) 2014-11-05 2016-06-01 Flexus Biosciences Inc Compuestos moduladores de la enzima indolamina 2,3-dioxigenasa (ido), sus métodos de síntesis y composiciones farmacéuticas que los contienen
PL3221363T3 (pl) 2014-11-21 2021-01-11 Bristol-Myers Squibb Company Przeciwciała przeciwko cd73 i ich zastosowanie
TW201630907A (zh) 2014-12-22 2016-09-01 必治妥美雅史谷比公司 TGFβR拮抗劑
US10983128B2 (en) 2015-02-05 2021-04-20 Bristol-Myers Squibb Company CXCL11 and SMICA as predictive biomarkers for efficacy of anti-CTLA4 immunotherapy
ES2789331T3 (es) 2015-03-02 2020-10-26 Rigel Pharmaceuticals Inc Inhibidores de TGF-beta
KR20170134981A (ko) 2015-04-03 2017-12-07 브리스톨-마이어스 스큅 컴퍼니 암의 치료를 위한 인돌아민-2,3-디옥시게나제의 억제제
WO2016162505A1 (en) 2015-04-08 2016-10-13 F-Star Biotechnology Limited Her2 binding agent therapies
ES2815683T3 (es) 2015-05-11 2021-03-30 Bristol Myers Squibb Co Compuestos tricíclicos como agentes antineoplásicos
WO2016183115A1 (en) 2015-05-12 2016-11-17 Bristol-Myers Squibb Company 5h-pyrido[3,2-b]indole compounds as anticancer agents
US9725449B2 (en) 2015-05-12 2017-08-08 Bristol-Myers Squibb Company Tricyclic compounds as anticancer agents
RS63897B1 (sr) 2015-05-29 2023-02-28 Bristol Myers Squibb Co Antitela protiv ox40 i njihova primena
EA201890162A1 (ru) 2015-06-29 2018-07-31 Бристол-Маерс Сквибб Компани Антитела к cd40 с повышенной агонистической активностью
CN104911168B (zh) * 2015-06-30 2018-03-30 中国人民解放军第三军医大学第一附属医院 I型胶原蛋白高亲和力多肽及其应用
EA039894B1 (ru) * 2015-07-10 2022-03-24 Файв Прайм Терапьютикс, Инк. Способ лечения рака, композиция для лечения рака, применение композиции для получения лекарственного средства для лечения рака, характеризующегося присутствием макрофагов, экспрессирующих csf1r
JP2018526344A (ja) 2015-07-28 2018-09-13 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Tgfベータ受容体アンタゴニスト
JP2018525415A (ja) 2015-08-25 2018-09-06 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Tgfベータ受容体アンタゴニスト
US10584160B2 (en) 2015-09-23 2020-03-10 Bristol-Myers Squibb Company Glypican-3-binding fibronectin based scaffold molecules
EP3377532B1 (en) 2015-11-19 2022-07-27 Bristol-Myers Squibb Company Antibodies against glucocorticoid-induced tumor necrosis factor receptor (gitr) and uses thereof
MA44309A (fr) * 2015-11-25 2018-10-03 Nogra Pharma Ltd Oligonucléotides antisens il-34 et leurs procédés d'utilisation
JP6856648B2 (ja) 2015-12-15 2021-04-07 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Cxcr4受容体アンタゴニスト
BR112018067368A2 (pt) 2016-03-04 2019-01-15 Bristol-Myers Squibb Company terapia de combinação com anticorpos anti-cd73
AU2017252527A1 (en) 2016-04-18 2018-11-08 Celldex Therapeutics, Inc. Agonistic antibodies that bind human CD40 and uses thereof
WO2017192815A1 (en) 2016-05-04 2017-11-09 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
KR20190003685A (ko) 2016-05-04 2019-01-09 브리스톨-마이어스 스큅 컴퍼니 인돌아민 2,3-디옥시게나제의 억제제 및 그의 사용 방법
EP3452451A4 (en) 2016-05-04 2019-11-13 Bristol-Myers Squibb Company INDOLEAMINE 2,3-DIOXYGENASE INHIBITORS AND METHODS OF USE
US10544099B2 (en) 2016-05-04 2020-01-28 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
JP2019516682A (ja) 2016-05-04 2019-06-20 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company インドールアミン2,3−ジオキシゲナーゼ阻害剤およびその使用方法
US11623958B2 (en) 2016-05-20 2023-04-11 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
US10994033B2 (en) 2016-06-01 2021-05-04 Bristol-Myers Squibb Company Imaging methods using 18F-radiolabeled biologics
BR112018076260A2 (pt) 2016-06-20 2019-03-26 Kymab Limited anticorpo ou fragmento do mesmo que se liga especificamente a hpd-l1, anticorpo biespecífico ou proteína de fusão, uso de um anticorpo ou fragmento, método, composição farmacêutica, método de modulação, método de inibição, método de tratamento, ácido nucleico, vetor, hospedeiro e imunocitocina
UY37325A (es) 2016-07-14 2018-01-31 Bristol Myers Squibb Company Una Corporacion Del Estado De Delaware Anticuerpos monoclonales que se enlazan a tim3 para estimular respuestas inmunitarias y composiciones que los contienen
US20190292179A1 (en) 2016-07-21 2019-09-26 Bristol-Myers Squibb Company TGF Beta RECEPTOR ANTAGONISTS
JP2019528301A (ja) 2016-08-26 2019-10-10 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company インドールアミン2,3−ジオキシゲナーゼの阻害剤およびその使用方法
WO2018039518A1 (en) 2016-08-26 2018-03-01 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
US10660909B2 (en) 2016-11-17 2020-05-26 Syntrix Biosystems Inc. Method for treating cancer using chemokine antagonists
US10961239B2 (en) 2017-01-05 2021-03-30 Bristol-Myers Squibb Company TGF beta receptor antagonists
CN110214012B (zh) 2017-01-20 2023-05-09 艾库斯生物科学有限公司 用于治疗癌症相关疾病的唑嘧啶
TWI788340B (zh) 2017-04-07 2023-01-01 美商必治妥美雅史谷比公司 抗icos促效劑抗體及其用途
CA3058944A1 (en) 2017-04-19 2018-10-25 Elstar Therapeutics, Inc. Multispecific molecules and uses thereof
EP3612030A4 (en) 2017-04-21 2021-04-28 Ikena Oncology, Inc. AHR INDOLE INHIBITORS AND THEIR USES
EP3621994A4 (en) 2017-05-12 2020-12-30 Harpoon Therapeutics, Inc. MESOTHELINE BINDING PROTEINS
US11066392B2 (en) 2017-05-12 2021-07-20 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
ES2951809T3 (es) 2017-05-17 2023-10-25 Arcus Biosciences Inc Derivados de quinazolina-pirazol para el tratamiento de trastornos relacionados con el cáncer
EA202090119A1 (ru) 2017-06-30 2020-04-21 Бристол-Маерс Сквибб Компани Аморфные и кристаллические формы ido-ингибиторов
KR20200032180A (ko) 2017-07-28 2020-03-25 브리스톨-마이어스 스큅 컴퍼니 항암제로서의 시클릭 디뉴클레오티드
AU2018319016B2 (en) 2017-08-17 2023-08-31 Ikena Oncology, Inc. AHR inhibitors and uses thereof
EP3676278B1 (en) 2017-08-31 2023-04-12 Bristol-Myers Squibb Company Cyclic dinucleotides as anticancer agents
JP7316263B2 (ja) 2017-08-31 2023-07-27 ブリストル-マイヤーズ スクイブ カンパニー 抗癌剤としての環状ジヌクレオチド
WO2019046500A1 (en) 2017-08-31 2019-03-07 Bristol-Myers Squibb Company CYCLIC DINUCLEOTIDES AS ANTICANCER AGENTS
WO2019074822A1 (en) 2017-10-09 2019-04-18 Bristol-Myers Squibb Company INDOLEAMINE 2,3-DIOXYGENASE INHIBITORS AND METHODS OF USE
US11203592B2 (en) 2017-10-09 2021-12-21 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
CN111344297B (zh) 2017-10-10 2023-10-20 百时美施贵宝公司 作为抗癌剂的环二核苷酸
EP3694552A1 (en) 2017-10-10 2020-08-19 Tilos Therapeutics, Inc. Anti-lap antibodies and uses thereof
BR112020007249B1 (pt) 2017-10-13 2022-11-22 Harpoon Therapeutics, Inc Roteína de ligação a antígeno de maturação de célula b, proteína de ligação multiespecífica e uso das referidas proteínas
WO2019079261A1 (en) 2017-10-16 2019-04-25 Bristol-Myers Squibb Company CYCLIC DINUCLEOTIDES AS ANTICANCER AGENTS
JP2021501801A (ja) 2017-11-01 2021-01-21 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company 癌の処置に用いるための免疫刺激アゴニスト抗体
ES2925450T3 (es) 2017-11-06 2022-10-18 Bristol Myers Squibb Co Compuestos de isofuranona útiles como inhibidores de HPK1
US20190142722A1 (en) * 2017-11-10 2019-05-16 The Trustees Of Columbia University In The City Of New York Methods and compositions for promoting or inducing hair growth
EP3720881A1 (en) 2017-12-08 2020-10-14 Elstar Therapeutics, Inc. Multispecific molecules and uses thereof
CN111788227A (zh) 2017-12-27 2020-10-16 百时美施贵宝公司 抗cd40抗体及其用途
US11447449B2 (en) 2018-01-05 2022-09-20 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
US20220089720A1 (en) 2018-01-12 2022-03-24 Bristol-Myers Squibb Company Antibodies against tim3 and uses thereof
US20210077832A1 (en) 2018-01-26 2021-03-18 Celldex Therapeutics, Inc. Methods of treating cancer with dendritic cell mobilizing agents
CA3089769A1 (en) 2018-01-29 2019-08-01 Merck Patent Gmbh Gcn2 inhibitors and uses thereof
TWI816742B (zh) 2018-01-29 2023-10-01 美商維泰克斯製藥公司 Gcn2抑制劑及其用途
US10519187B2 (en) 2018-02-13 2019-12-31 Bristol-Myers Squibb Company Cyclic dinucleotides as anticancer agents
WO2019165315A1 (en) 2018-02-23 2019-08-29 Syntrix Biosystems Inc. Method for treating cancer using chemokine antagonists alone or in combination
CN111801341B (zh) 2018-03-08 2023-10-24 百时美施贵宝公司 作为抗癌剂的环二核苷酸
SG11202008593PA (en) 2018-03-21 2020-10-29 Five Prime Therapeutics Inc ANTIBODIES BINDING TO VISTA AT ACIDIC pH
AU2019236865A1 (en) 2018-03-23 2020-10-01 Bristol-Myers Squibb Company Antibodies against MICA and/or MICB and uses thereof
PE20210313A1 (es) 2018-03-28 2021-02-12 Bristol Myers Squibb Co Proteinas de fusion interleucina-2/receptor alfa de interleucina-2 y metodos de uso
WO2019200256A1 (en) 2018-04-12 2019-10-17 Bristol-Myers Squibb Company Anticancer combination therapy with cd73 antagonist antibody and pd-1/pd-l1 axis antagonist antibody
EP3781162A1 (en) 2018-04-16 2021-02-24 Arrys Therapeutics, Inc. Ep4 inhibitors and use thereof
WO2019213340A1 (en) 2018-05-03 2019-11-07 Bristol-Myers Squibb Company Uracil derivatives as mer-axl inhibitors
US11180531B2 (en) 2018-06-22 2021-11-23 Bicycletx Limited Bicyclic peptide ligands specific for Nectin-4
CA3104654A1 (en) 2018-06-27 2020-01-02 Bristol-Myers Squibb Company Substituted naphthyridinone compounds useful as t cell activators
HRP20230627T1 (hr) 2018-06-27 2023-09-29 Bristol-Myers Squibb Company Spojevi naftiridinona korisni kao aktivatori t ćelija
PE20211604A1 (es) 2018-07-09 2021-08-23 Five Prime Therapeutics Inc Anticuerpos de union a ilt4
US20220073617A1 (en) 2018-07-11 2022-03-10 Five Prime Therapeutics, Inc. Antibodies Binding to Vista at Acidic pH
WO2020023355A1 (en) 2018-07-23 2020-01-30 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
US20210355113A1 (en) 2018-07-23 2021-11-18 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
US10959986B2 (en) 2018-08-29 2021-03-30 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
US11253525B2 (en) 2018-08-29 2022-02-22 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
JP2022500499A (ja) 2018-09-07 2022-01-04 ピク セラピューティクス, インコーポレイテッド Eif4e阻害剤およびその使用
AU2019346466A1 (en) 2018-09-25 2021-05-20 Harpoon Therapeutics, Inc. DLL3 binding proteins and methods of use
EP3856350A1 (en) 2018-09-27 2021-08-04 Marengo Therapeutics, Inc. Csf1r/ccr2 multispecific antibodies
CN113164780A (zh) 2018-10-10 2021-07-23 泰洛斯治疗公司 抗lap抗体变体及其用途
MX2021005708A (es) 2018-11-16 2021-09-21 Bristol Myers Squibb Co Anticuerpos anti grupo 2a del receptor inhibidor de células asesinas naturales (anti-nkg2a) y usos de los mismos.
US20220016168A1 (en) 2018-12-11 2022-01-20 Celldex Therapeutics, Inc. Methods of using cd27 antibodies as conditioning treatment for adoptive cell therapy
EP3670659A1 (en) 2018-12-20 2020-06-24 Abivax Biomarkers, and uses in treatment of viral infections, inflammations, or cancer
EA202192555A1 (ru) 2019-03-19 2021-11-25 Фундасио Привада Институт Д'Инвестигасио Онколохика Де Валь Эброн Комбинированная терапия для лечения рака
EP3946462A1 (en) 2019-04-02 2022-02-09 BicycleTX Limited Bicycle toxin conjugates and uses thereof
US20230242478A1 (en) 2019-05-13 2023-08-03 Bristol-Myers Squibb Company AGONISTS OF ROR GAMMAt
WO2020231766A1 (en) 2019-05-13 2020-11-19 Bristol-Myers Squibb Company AGONISTS OF ROR GAMMAt
WO2020243423A1 (en) 2019-05-31 2020-12-03 Ikena Oncology, Inc. Tead inhibitors and uses thereof
US20220306630A1 (en) 2019-08-06 2022-09-29 Bristol-Myers Squibb Company AGONISTS OF ROR GAMMAt
WO2021024020A1 (en) 2019-08-06 2021-02-11 Astellas Pharma Inc. Combination therapy involving antibodies against claudin 18.2 and immune checkpoint inhibitors for treatment of cancer
AR119821A1 (es) 2019-08-28 2022-01-12 Bristol Myers Squibb Co Compuestos de piridopirimidinonilo sustituidos útiles como activadores de células t
BR112022004451A2 (pt) 2019-09-13 2022-06-21 Nimbus Saturn Inc Antagonistas de hpk1 e usos dos mesmos
JP2022548292A (ja) 2019-09-19 2022-11-17 ブリストル-マイヤーズ スクイブ カンパニー 酸性pHでVISTAと結合する抗体
CA3158976A1 (en) 2019-11-19 2021-05-27 Bristol-Myers Squibb Company Compounds useful as inhibitors of helios protein
JP2023502531A (ja) 2019-11-26 2023-01-24 イケナ オンコロジー, インコーポレイテッド 多形カルバゾール誘導体およびその使用
US20230052523A1 (en) 2019-11-26 2023-02-16 Bristol-Myers Squibb Company Salts/cocrystals of (r)-n-(4-chlorophenyl)-2-((1s,4s)-4-(6-fluoroquinolin-4-yl)cyclohexyl)propanamide
AR120823A1 (es) 2019-12-23 2022-03-23 Bristol Myers Squibb Co Compuestos bicíclicos sustituidos útiles como activadores de células t
CN115103841A (zh) 2019-12-23 2022-09-23 百时美施贵宝公司 可用作t细胞激活剂的经取代的喹啉酮基哌嗪化合物
CA3163003A1 (en) 2019-12-23 2021-07-01 Upender Velaparthi Substituted heteroaryl compounds useful as t cell activators
EP4081522A1 (en) 2019-12-23 2022-11-02 Bristol-Myers Squibb Company Substituted piperazine derivatives useful as t cell activators
CN114846007A (zh) 2019-12-23 2022-08-02 百时美施贵宝公司 可用作t细胞激活剂的经取代的喹唑啉基化合物
EP4087874A1 (en) 2020-01-06 2022-11-16 HiFiBiO (HK) Limited Anti-tnfr2 antibody and uses thereof
CA3163988A1 (en) 2020-01-07 2021-07-15 Germain MARGALL DUCOS Anti-galectin-9 antibody and uses thereof
CA3170411A1 (en) 2020-03-03 2021-09-10 Christopher L. Vandeusen Eif4e inhibitors and uses thereof
US20230140384A1 (en) 2020-03-09 2023-05-04 Bristol-Myers Squibb Company Antibodies to cd40 with enhanced agonist activity
CA3173831A1 (en) 2020-03-19 2021-09-23 Arcus Biosciences, Inc. Tetralin and tetrahydroquinoline compounds as inhibitors of hif-2alpha
TW202140441A (zh) 2020-03-23 2021-11-01 美商必治妥美雅史谷比公司 經取代之側氧基異吲哚啉化合物
WO2021207449A1 (en) 2020-04-09 2021-10-14 Merck Sharp & Dohme Corp. Affinity matured anti-lap antibodies and uses thereof
WO2021231732A1 (en) 2020-05-15 2021-11-18 Bristol-Myers Squibb Company Antibodies to garp
MX2022015157A (es) 2020-06-02 2023-01-16 Arcus Biosciences Inc Anticuerpos para tigit.
JP2023530456A (ja) 2020-06-17 2023-07-18 アーカス バイオサイエンシズ インコーポレイティド Cd73阻害剤の結晶形態及びその使用
AU2021306613A1 (en) 2020-07-07 2023-02-02 BioNTech SE Therapeutic RNA for HPV-positive cancer
KR20230077722A (ko) 2020-08-10 2023-06-01 지브이20 테라퓨틱스 엘엘씨 Igsf8을 표적화하여 자가면역 질환 및 암을 치료하기 위한 조성물 및 방법
AU2021327130A1 (en) 2020-08-17 2023-03-02 Bicycletx Limited Bicycle conjugates specific for Nectin-4 and uses thereof
KR20230131189A (ko) 2020-12-02 2023-09-12 이케나 온콜로지, 인코포레이티드 Tead 억제제 및 이의 용도
WO2022120353A1 (en) 2020-12-02 2022-06-09 Ikena Oncology, Inc. Tead inhibitors and uses thereof
WO2022135667A1 (en) 2020-12-21 2022-06-30 BioNTech SE Therapeutic rna for treating cancer
TW202245808A (zh) 2020-12-21 2022-12-01 德商拜恩迪克公司 用於治療癌症之治療性rna
WO2022135666A1 (en) 2020-12-21 2022-06-30 BioNTech SE Treatment schedule for cytokine proteins
WO2022148979A1 (en) 2021-01-11 2022-07-14 Bicycletx Limited Methods for treating cancer
MX2023009059A (es) 2021-02-02 2023-09-15 Liminal Biosciences Ltd Antagonistas de gpr84 y usos de estos.
AU2022216810A1 (en) 2021-02-02 2023-08-24 Liminal Biosciences Limited Gpr84 antagonists and uses thereof
US20240109899A1 (en) 2021-02-04 2024-04-04 Bristol-Myers Squibb Company Benzofuran compounds as sting agonists
CN117295737A (zh) 2021-03-05 2023-12-26 林伯士萨顿公司 Hpk1拮抗剂和其用途
US11918582B2 (en) 2021-03-15 2024-03-05 Rapt Therapeutics, Inc. Pyrazole pyrimidine compounds and uses thereof
JP2024514530A (ja) 2021-04-02 2024-04-02 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア 切断型cdcp1に対する抗体およびその使用
EP4320125A1 (en) 2021-04-05 2024-02-14 Bristol-Myers Squibb Company Pyridinyl substituted oxoisoindoline compounds for the treatment of cancer
US11718601B2 (en) 2021-04-06 2023-08-08 Bristol-Myers Squibb Company Pyridinyl substituted oxoisoindoline compounds
EP4323066A1 (en) 2021-04-16 2024-02-21 Ikena Oncology, Inc. Mek inhibitors and uses thereof
EP4341261A1 (en) 2021-05-21 2024-03-27 Arcus Biosciences, Inc. Axl compounds
EP4341262A1 (en) 2021-05-21 2024-03-27 Arcus Biosciences, Inc. Axl inhibitor compounds
IL309831A (en) 2021-07-13 2024-02-01 BioNTech SE Multispecific binding agents against CD40 and CD137 in combined cancer therapy
IL310924A (en) 2021-08-25 2024-04-01 Pic Therapeutics Inc EIF4E inhibitors and their uses
WO2023028238A1 (en) 2021-08-25 2023-03-02 PIC Therapeutics, Inc. Eif4e inhibitors and uses thereof
TW202333802A (zh) 2021-10-11 2023-09-01 德商拜恩迪克公司 用於肺癌之治療性rna(二)
WO2023114984A1 (en) 2021-12-17 2023-06-22 Ikena Oncology, Inc. Tead inhibitors and uses thereof
WO2023150186A1 (en) 2022-02-01 2023-08-10 Arvinas Operations, Inc. Dgk targeting compounds and uses thereof
WO2023173053A1 (en) 2022-03-10 2023-09-14 Ikena Oncology, Inc. Mek inhibitors and uses thereof
WO2023173057A1 (en) 2022-03-10 2023-09-14 Ikena Oncology, Inc. Mek inhibitors and uses thereof
WO2023211889A1 (en) 2022-04-25 2023-11-02 Ikena Oncology, Inc. Polymorphic compounds and uses thereof
WO2023215719A1 (en) 2022-05-02 2023-11-09 Arcus Biosciences, Inc. Anti-tigit antibodies and uses of the same
WO2023230205A1 (en) 2022-05-25 2023-11-30 Ikena Oncology, Inc. Mek inhibitors and uses thereof
WO2024028363A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Heteroaryl carboxamide and related gpr84 antagonists and uses thereof
WO2024028364A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Aryl-triazolyl and related gpr84 antagonists and uses thereof
WO2024028365A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Substituted pyridone gpr84 antagonists and uses thereof
WO2024036100A1 (en) 2022-08-08 2024-02-15 Bristol-Myers Squibb Company Substituted tetrazolyl compounds useful as t cell activators
WO2024036101A1 (en) 2022-08-09 2024-02-15 Bristol-Myers Squibb Company Tertiary amine substituted bicyclic compounds useful as t cell activators

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8206715B2 (en) * 2010-05-04 2012-06-26 Five Prime Therapeutics, Inc. Antibodies that bind colony stimulating factor 1 receptor (CSF1R)

Family Cites Families (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989003687A1 (en) 1987-10-23 1989-05-05 Genetics Institute, Inc. Composition and method for treating cancers characterized by over-expression of the c-fms proto-oncogene
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
US6713610B1 (en) 1990-01-12 2004-03-30 Raju Kucherlapati Human antibodies derived from immunized xenomice
US5874299A (en) 1990-08-29 1999-02-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US6300129B1 (en) 1990-08-29 2001-10-09 Genpharm International Transgenic non-human animals for producing heterologous antibodies
US6255458B1 (en) 1990-08-29 2001-07-03 Genpharm International High affinity human antibodies and human antibodies against digoxin
US5877397A (en) 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
JPH08507916A (ja) 1992-06-09 1996-08-27 カイロン コーポレイション M−csfの結晶化
US20020193575A1 (en) 1993-09-07 2002-12-19 Smithkline Beecham P.L.C. Recombinant IL4 antibodies useful in treatment of IL4 mediated disorders
US6972323B1 (en) 1997-04-01 2005-12-06 Sankyo Company, Limited Anti-Fas antibodies
US6342220B1 (en) 1997-08-25 2002-01-29 Genentech, Inc. Agonist antibodies
WO1999029345A1 (en) 1997-12-05 1999-06-17 La Jolla Institute For Experimental Medicine Inhibition of tumor growth by macrophage intervention
AU6366999A (en) 1998-10-30 2000-05-22 Takeda Chemical Industries Ltd. Betacellulin protein-containing preparations
PT1223980E (pt) 1999-10-28 2003-10-31 Reinhold Hofbauer Utilizacao de inibidores de csf-1
AU1759501A (en) 1999-11-08 2001-06-06 Government of The United States of America, as represented by The Secretary Department of Health & Human Services, The National Institutes of Health, The Method of treating a viral infection using antagonists of macrophage colony stimulating factor
US7108852B2 (en) 2000-03-20 2006-09-19 Warner-Lambert Company Llc Methods of treating inflammation using antibodies to M-CSF
US7455836B2 (en) 2000-05-08 2008-11-25 The University Of Melbourne Method of treatment and agents useful for same
US6773895B2 (en) 2000-09-01 2004-08-10 Boehringer Ingelheim Pharma Kg Method for identifying substances which positively influence inflammatory conditions of chronic inflammatory airway diseases
US7247618B2 (en) 2001-04-30 2007-07-24 Tripathi Rajavashisth Methods for inhibiting macrophage colony stimulating factor and c-FMS-dependent cell signaling
JP2005500034A (ja) 2001-06-20 2005-01-06 プロション バイオテク リミテッド 受容体型タンパク質チロシンキナーゼ活性化を遮断する抗体、そのスクリーニング方法、及びその使用
CN1787837A (zh) 2002-11-15 2006-06-14 希龙公司 防止和治疗癌转移以及与癌转移相关的骨质损失的方法
ES2630224T3 (es) 2003-10-22 2017-08-18 Keck Graduate Institute Métodos para sintetizar polipéptidos heteromultiméricos en levadura con el uso de una estrategia de apareamiento haploide
GB0325836D0 (en) 2003-11-05 2003-12-10 Celltech R&D Ltd Biological products
EP1706137A2 (en) 2004-01-21 2006-10-04 Chiron Corporation M-csf muteins and uses thereof
US7105183B2 (en) * 2004-02-03 2006-09-12 The Regents Of The University Of California Chlorite in the treatment of neurodegenerative disease
AU2005245896A1 (en) 2004-05-14 2005-12-01 Receptor Biologix, Inc. Cell surface receptor isoforms and methods of identifying and using the same
CA2574654C (en) 2004-07-22 2014-02-18 Five Prime Therapeutics, Inc. Compositions and methods of use for mgd-csf in disease treatment
WO2006076288A2 (en) 2005-01-11 2006-07-20 Five Prime Therapeutics, Inc. Dna constructs for long-term expression of intravascularly injected naked dna
CA2589895A1 (en) 2005-01-27 2006-08-03 Five Prime Therapeutics, Inc. Leader sequences for directing secretion of polypeptides and methods for production thereof
US8003108B2 (en) 2005-05-03 2011-08-23 Amgen Inc. Sclerostin epitopes
EP1777523A1 (en) 2005-10-19 2007-04-25 INSERM (Institut National de la Santé et de la Recherche Médicale) An in vitro method for the prognosis of progression of a cancer and of the outcome in a patient and means for performing said method
JP2009515521A (ja) 2005-11-10 2009-04-16 レセプター バイオロジックス, インコーポレイテッド レセプターアイソフォームおよびリガンドアイソフォームの産生のための方法
WO2007075933A2 (en) 2005-12-21 2007-07-05 Cell Signaling Technology, Inc. Translocation and mutant csf1r kinase in human leukemia
JP2009521685A (ja) 2005-12-22 2009-06-04 ノバルティス アーゲー 可溶性ヒトm−csf受容体およびその使用
US20090246208A1 (en) 2006-01-05 2009-10-01 Novartis Ag Methods for preventing and treating cancer metastasis and bone loss associated with cancer metastasis
WO2008042611A2 (en) 2006-09-29 2008-04-10 Centocor, Inc. Method of using il6 antagonists with mitoxantrone for prostate cancer
JP2010510219A (ja) 2006-11-17 2010-04-02 バイオジェン・アイデック・エムエイ・インコーポレイテッド アミロイド関連障害を処置するためのコロニー刺激因子の全身投与
EP2069404B1 (en) 2007-02-14 2011-01-05 Vaccinex, Inc. Humanized anti-cd100 antibodies
WO2008124858A2 (en) 2007-04-11 2008-10-23 F-Star Biotechnologische Forschungs- Und Entwicklungsges. M.B.H. Targeted receptor
WO2008150383A1 (en) 2007-05-30 2008-12-11 Albert Einstein College Of Medicine Of Yeshiva University Csf-1r mutants
PL2188313T3 (pl) 2007-08-21 2018-04-30 Amgen, Inc. Białka wiążące ludzki antygen c-fms
US7981415B2 (en) 2007-09-07 2011-07-19 Cisthera, Inc. Humanized PAI-1 antibodies
EP2215482A2 (en) 2007-10-31 2010-08-11 Janssen Pharmaceutica N.V. Biomarker for assessing response to fms treatment
WO2009075344A1 (ja) 2007-12-12 2009-06-18 Japan As Represented By Director General Of Agency Of National Cancer Center M-csf受容体を分子標的とするmll白血病及びmoz白血病治療剤、およびその利用
KR101633520B1 (ko) 2008-03-14 2016-06-24 트랜스진 에스.에이. Csf-1r에 대한 항체
US8470977B2 (en) 2008-03-14 2013-06-25 Transgene S.A. Antibody against the CSF-1R
WO2010030979A2 (en) 2008-09-12 2010-03-18 Xbiotech, Inc. Targeting pathogenic monocytes
US8183207B2 (en) 2008-11-26 2012-05-22 Five Prime Therapeutics, Inc. Treatment of osteolytic disorders and cancer using CSF1R extracellular domain fusion molecules
WO2010062399A2 (en) * 2008-11-26 2010-06-03 Five Prime Therapeutics, Inc. Csf1r extracellular domain fusion molecules and treatments using same
CN102740888B (zh) * 2009-11-24 2016-10-12 奥尔德生物制药公司 Il-6抗体及其用途
ES2557454T3 (es) 2009-12-10 2016-01-26 F. Hoffmann-La Roche Ag Anticuerpos que se unen al dominio extracelular 4 de CSF1R humana y su utilización
EP2542587A1 (en) 2010-03-05 2013-01-09 F. Hoffmann-La Roche AG Antibodies against human csf-1r and uses thereof
EP2542588A1 (en) 2010-03-05 2013-01-09 F. Hoffmann-La Roche AG Antibodies against human csf-1r and uses thereof
AR080698A1 (es) 2010-04-01 2012-05-02 Imclone Llc Anticuerpo o fragmento del mismo que especificamente enlaza la variante de csf -1r humano, composicion farmaceutica que lo comprende, su uso para la manufactura de un medicamento util para el tratamiento de cancer y metodo para determinar si un sujeto es candidato para tratamiento de cancer basado e
AR084234A1 (es) * 2010-12-13 2013-05-02 Novartis Ag Metodos predictivos y metodos para tratar artritis usando antagonistas de interleuquina 17 (il-17)
KR102061358B1 (ko) 2011-10-21 2019-12-31 트랜스진 에스.에이. 대식세포 활성화의 조절
WO2013057290A1 (en) 2011-10-21 2013-04-25 INSERM (Institut National de la Santé et de la Recherche Médicale) A m-dc8+ monocyte depleting agent for the prevention or the treatment of a condition associated with a chronic hyperactivation of the immune system
US20130302322A1 (en) 2012-05-11 2013-11-14 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
EP3539984A1 (en) 2012-05-11 2019-09-18 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
RU2718751C2 (ru) 2012-08-31 2020-04-14 Файв Прайм Терапьютикс, Инк. Способы лечения патологических состояний антителами, которые связываются с рецептором колониестимулирующего фактора 1 (csf1r)

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8206715B2 (en) * 2010-05-04 2012-06-26 Five Prime Therapeutics, Inc. Antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US8747845B2 (en) * 2010-05-04 2014-06-10 Five Prime Therapeutics, Inc. Methods of treatment by administering antibodies that bind colony stimulating factor 1 receptor (CSF1R)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Buch et al. (2011), Current Opinion in Rheumatology, Vol. 23, pp.245-251. *

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10287358B2 (en) 2009-12-10 2019-05-14 Hoffmann-La Roche Inc. Antibodies against human CSF-1R and uses thereof
US10077314B1 (en) 2009-12-10 2018-09-18 Hoffmann-La Roche Inc. Antibodies against human CSF-1R and uses thereof
US9200075B2 (en) 2010-05-04 2015-12-01 Five Prime Therapeutics, Inc. Nucleic acids encoding antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US9695243B2 (en) 2010-05-04 2017-07-04 Five Prime Therapeutics, Inc. Antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US11186646B2 (en) 2010-05-04 2021-11-30 Five Prime Therapeutics, Inc. Antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US9957327B2 (en) 2010-05-04 2018-05-01 Five Prime Therapeutics, Inc. Antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US10562970B2 (en) 2010-05-04 2020-02-18 Five Prime Therapeutics, Inc. Antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US10336830B2 (en) 2011-12-15 2019-07-02 Hoffmann-La Roche Inc. Antibodies against human CSF-1R and uses thereof
US10982001B2 (en) 2012-05-11 2021-04-20 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US9192667B2 (en) 2013-04-22 2015-11-24 Hoffmann-La Roche Inc. Method of treating cancer by administering CSF-1R antibodies and a TLR9 agonist
US11512133B2 (en) 2013-09-12 2022-11-29 Hoffmann-La Roche Inc. Methods for treating colon cancer or inhibiting cell proliferation by administering a combination of antibodies against human CSF-1R and antibodies against human PD-L1
US10975153B2 (en) 2014-06-23 2021-04-13 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US11566076B2 (en) 2014-10-29 2023-01-31 Five Prime Therapeutics, Inc. Anti-CSF1R antibody and anti-PD-1 antibody combination therapy for selected cancers
US10618967B2 (en) 2014-10-29 2020-04-14 Five Prime Therapeutics, Inc. Anti-CSF1R antibody and anti PD-1 antibody combination therapy for cancer
US9765147B2 (en) 2014-10-29 2017-09-19 Five Prime Therapeutics, Inc. Anti-CSFR1 antibody and anti PD-1 antibody combination therapy for cancer
US10221244B2 (en) 2014-10-29 2019-03-05 Five Prime Therapeutics, Inc. Anti-CSF1R antibody and anti PD-1 antibody combination therapy for cancer
US10040858B2 (en) 2014-12-22 2018-08-07 Five Prime Therapeutics, Inc. Anti-CSF1R antibodies for treating PVNS
US10730949B2 (en) 2014-12-22 2020-08-04 Five Prime Therapeutics, Inc. Method of treating PVNS with anti-CSF1R antibodies
US11559583B2 (en) 2015-04-13 2023-01-24 Five Prime Therapeutics, Inc. Anti-CSF1R antibody and agonistic anti-CD40 antibody combination therapy for cancer
US11542335B2 (en) 2016-08-25 2023-01-03 Hoffmann-La Roche Inc. Method of treating cancer in a patient by administering an antibody which binds colony stimulating factor-1 receptor (CSF-1R)
US11498968B2 (en) 2016-12-22 2022-11-15 Hoffmann-La Roche Inc. Treatment of tumors with an anti-CSF-1R antibody in combination with an anti-PD-L1 antibody after failure of anti-PD-L1/PD1 treatment
US11267857B2 (en) * 2017-02-27 2022-03-08 Shattuck Labs, Inc. CSF1R-based chimeric proteins
US11421034B2 (en) 2017-09-13 2022-08-23 Five Prime Therapeutics, Inc. Combination anti-CSF1R and anti-PD-1 antibody combination therapy for pancreatic cancer
US11358948B2 (en) 2017-09-22 2022-06-14 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
US11623932B2 (en) 2017-09-22 2023-04-11 Kymera Therapeutics, Inc. Protein degraders and uses thereof
US11318205B1 (en) 2017-12-26 2022-05-03 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US10874743B2 (en) 2017-12-26 2020-12-29 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11723980B2 (en) 2017-12-26 2023-08-15 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11485743B2 (en) 2018-01-12 2022-11-01 Kymera Therapeutics, Inc. Protein degraders and uses thereof
US11512080B2 (en) 2018-01-12 2022-11-29 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
US11932635B2 (en) 2018-01-12 2024-03-19 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
US11897882B2 (en) 2018-07-06 2024-02-13 Kymera Therapeutics, Inc. Tricyclic crbn ligands and uses thereof
US11292792B2 (en) 2018-07-06 2022-04-05 Kymera Therapeutics, Inc. Tricyclic CRBN ligands and uses thereof
WO2020010177A1 (en) 2018-07-06 2020-01-09 Kymera Therapeutics, Inc. Tricyclic crbn ligands and uses thereof
US11352350B2 (en) 2018-11-30 2022-06-07 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11807636B2 (en) 2018-11-30 2023-11-07 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11117889B1 (en) 2018-11-30 2021-09-14 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11746120B2 (en) 2019-04-05 2023-09-05 Kymera Therapeutics, Inc. Stat degraders and uses thereof
US11485750B1 (en) 2019-04-05 2022-11-01 Kymera Therapeutics, Inc. STAT degraders and uses thereof
US11707457B2 (en) 2019-12-17 2023-07-25 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11591332B2 (en) 2019-12-17 2023-02-28 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11779578B2 (en) 2019-12-17 2023-10-10 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11679109B2 (en) 2019-12-23 2023-06-20 Kymera Therapeutics, Inc. SMARCA degraders and uses thereof
US11932624B2 (en) 2020-03-19 2024-03-19 Kymera Therapeutics, Inc. MDM2 degraders and uses thereof
US11685750B2 (en) 2020-06-03 2023-06-27 Kymera Therapeutics, Inc. Crystalline forms of IRAK degraders
US11857535B2 (en) 2020-07-30 2024-01-02 Kymera Therapeutics, Inc. Methods of treating mutant lymphomas
US11773103B2 (en) 2021-02-15 2023-10-03 Kymera Therapeutics, Inc. IRAK4 degraders and uses thereof

Also Published As

Publication number Publication date
AU2013308635A1 (en) 2015-03-12
SG10201906328RA (en) 2019-08-27
RU2015111563A (ru) 2016-10-20
HK1212214A1 (en) 2016-06-10
US10822421B2 (en) 2020-11-03
TW201414492A (zh) 2014-04-16
JP7106386B2 (ja) 2022-07-26
US20190248908A1 (en) 2019-08-15
JP2015533787A (ja) 2015-11-26
EP2890398A1 (en) 2015-07-08
JP2021095415A (ja) 2021-06-24
TW201919691A (zh) 2019-06-01
CN107759690A (zh) 2018-03-06
WO2014036357A1 (en) 2014-03-06
BR112015004426A2 (pt) 2018-08-28
EP2890398A4 (en) 2016-03-09
AU2018204331B2 (en) 2020-03-12
IL237351A0 (en) 2015-04-30
US20160326254A1 (en) 2016-11-10
CN104684582A (zh) 2015-06-03
TWI713436B (zh) 2020-12-21
JP2018197250A (ja) 2018-12-13
CA2882804A1 (en) 2014-03-06
IL237351B (en) 2020-03-31
SG11201501413YA (en) 2015-03-30
KR20150047593A (ko) 2015-05-04
AU2018204331A1 (en) 2018-07-05
KR20200140400A (ko) 2020-12-15
US20210107986A1 (en) 2021-04-15
EP3679949A1 (en) 2020-07-15
US10221243B2 (en) 2019-03-05
RU2718751C2 (ru) 2020-04-14

Similar Documents

Publication Publication Date Title
US10822421B2 (en) Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US20210277130A1 (en) Methods of Treating Conditions with Antibodies that Bind Colony Stimulating Factor 1 Receptor (CSF1R)
AU2012374617B2 (en) Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (CSF1R)
US20210317219A1 (en) Methods of Treating Conditions with Antibodies that Bind Colony Stimulating Factor 1 Receptor (CSF1R)
AU2019204985B2 (en) Method of treating conditions with antibodies that bind colony stimulating factor 1 receptor (CSF1R)
NZ745504A (en) Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
NZ745504B2 (en) Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)

Legal Events

Date Code Title Description
AS Assignment

Owner name: FIVE PRIME THERAPEUTICS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WONG, BRIAN;MASTELLER, EMMA;REEDQUIST, KRIS;SIGNING DATES FROM 20130906 TO 20130911;REEL/FRAME:031190/0210

STCB Information on status: application discontinuation

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