US20180085472A1 - Combination therapy for cancer - Google Patents

Combination therapy for cancer Download PDF

Info

Publication number
US20180085472A1
US20180085472A1 US15/564,866 US201615564866A US2018085472A1 US 20180085472 A1 US20180085472 A1 US 20180085472A1 US 201615564866 A US201615564866 A US 201615564866A US 2018085472 A1 US2018085472 A1 US 2018085472A1
Authority
US
United States
Prior art keywords
antibody
cancer
csf1r
seq
sequence
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
US15/564,866
Other languages
English (en)
Inventor
Emma Masteller
Thomas Brennan
David Bellovin
Kevin Baker
Brian Wong
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 US15/564,866 priority Critical patent/US20180085472A1/en
Publication of US20180085472A1 publication Critical patent/US20180085472A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6875Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody being a hybrid immunoglobulin
    • A61K47/6879Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody being a hybrid immunoglobulin the immunoglobulin having two or more different antigen-binding sites, e.g. bispecific or multispecific immunoglobulin
    • 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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1084Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody the antibody being a hybrid immunoglobulin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • 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
    • A61K2039/507Comprising a combination of two or more separate 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

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 activation by CSF1 or IL-34 leads to the trafficking, survival, proliferation, and differentiation of monocytes and macrophages, as well as other monocytic cell lineages such as osteoclasts, dendritic cells, and microglia.
  • CSF1R tumor-associated macrophages
  • TAMs tumor-associated macrophages
  • CSF1R plays a role in osteolytic bone destruction in bone metastasis.
  • TAMs promote tumor growth, in part, by suppressing anti-tumor T cell effector function through the release of immunosuppressive cytokines and the expression of T cell inhibitory surface proteins.
  • antibodies that bind to CSF1R may be useful in methods of treating cancer.
  • Some tumor cells may escape detection by the immune system at least in part by suppressing the immune response, for instance by altering the expression of immune modulatory genes.
  • the relative concentrations of both immune stimulatory and immune inhibitory molecules in the body may modulate the adaptive immune response.
  • a relatively high level of expression of inhibitory molecules and/or reduced expression of certain stimulatory molecules may create a checkpoint or switch that down-regulates the adaptive immune response.
  • Agents that counteract this effect by up-regulating the adaptive immune response or by stimulating the innate immune response are potential cancer therapies.
  • a combination regimen of agents that modulate the immune response in cancer may increase the depth and durability of the response and may also broaden efficacy to patients who do not respond to a single agent alone.
  • methods of treating cancer in a subject comprising administering to the subject an anti-CSF1R antibody and at least one immune stimulating agent.
  • the at least one immune stimulating agent comprises an agonist of an immune-stimulatory molecule, including a co-stimulatory molecule, while in some embodiments, the at least one immune stimulating agent comprises an antagonist of an immune inhibitory molecule, including a co-inhibitory molecule.
  • the at least one immune stimulating agent comprises an agonist of an immune-stimulatory molecule, including a co-stimulatory molecule, found on immune cells, such as T cells.
  • the at least one immune stimulating agent comprises an antagonist of an immune-inhibitory molecule, including a co-inhibitory molecule, found on immune cells, such as T cells.
  • the at least one immune stimulating agent comprises an agonist of an immune-stimulatory molecule, including a co-stimulatory molecule, found on cells involved in innate immunity, such as NK cells.
  • the at least one immune stimulating agent comprises an antagonist of an immune-inhibitory molecule, including a co-inhibitory molecule, found on cells involved in innate immunity, such as NK cells.
  • the combination enhances the antigen-specific T cell response in the treated subject and/or enhances the innate immunity response in the subject.
  • the combination results in an improved anti-tumor response in an animal cancer model, such as a xenograft model, compared to administration of either the anti-CSF1R antibody or immune stimulating agent alone. In some embodiments, the combination results in a synergistic response in an animal cancer model, such as a xenograft model, compared to administration of either the anti-CSF1R antibody or immune stimulating agent alone.
  • the at least one immune stimulating agent comprises an antagonist of an inhibitor of the activation of T cells, while in some embodiments, the at least one immune stimulating agent comprises comprises an agonist of a stimulator of the activation of T cells.
  • the at least one immune stimulating agent comprises an antagonist of CTLA4, LAG-3, Galectin 1, Galectin 9, CEACAM-1, BTLA, CD25, CD69, TIGIT, CD113, GPR56, VISTA, B7-H3, B7-H4, 2B4, CD48, GARP, PD1H, LAIR1, TIM1, TIM3, TIM4, ILT4, IL-6, IL-10, TGF ⁇ , VEGF, KIR, LAG-3, adenosine A2A receptor, PI3Kdelta, or IDO.
  • the at least one immune stimulating agent comprises an agonist of B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD27, CD40, CD40L, DR3, CD28H, IL-2, IL-7, IL-12, IL-15, IL-21, IFN ⁇ , STING or a Toll-like receptor agonist such as a TLR2/4 agonist.
  • the at least one immune stimulating agent comprises an agent that binds to a member of the B7 family of membrane-bound proteins such as B7-1, B7-2, B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6.
  • B7-1, B7-2, B7-H2 ICOS-L
  • B7-H3, B7-H4, B7-H5 VISTA
  • B7-H6 B7-H6
  • the at least one immune stimulating agent comprises an agent that binds to a member of the TNF receptor family or a co-stimulatory or co-inhibitory molecule binding to a member of the TNF receptor family such as CD40, CD40L, OX40, OX40L, GITR, GITRL, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, EDA1, EDA2, TACI, APRIL, BCMA, LT ⁇ R, LIGHT, DeR3, HVEM, VEGL/TL1A, TRAMP/DR3, TNFR1, TNF ⁇ , TNFR2, TNF ⁇ , 1 ⁇ 2, FAS, FASL, RELT, DR6,
  • the at least one immune stimulating agent comprises an agent that antagonizes or inhibits a cytokine that inhibits T cell activation such as IL-6, IL-10, TGF ⁇ , VEGF.
  • the at least one immune stimulating agent comprises an antagonist of a chemokine, such as CXCR2, CXCR4, CCR2, or CCR4.
  • the at least one immune stimulating agent comprises an agonist of a cytokine that stimulates T cell activation such as IL-2, IL-7, IL-12, IL-15, IL-21, and IFN ⁇ .
  • the at least one immune stimulating agent comprises an antibody.
  • the at least one immune stimulating agent may comprise a vaccine, such as a mesothelin-targeting vaccine or attenuated listeria cancer vaccine such as CRS-207. Any one or more of the above antagonists, agonists, and binding agents may be combined with any one or more of the anti-CSF1R antibodies described herein.
  • the at least one immune stimulating agent comprises a CD40 agonist, optionally in combination with at least one other immune stimulating agent as listed above.
  • the CD40 agonist is an antibody.
  • the CD40 agonist is an anti-CD40 antibody.
  • the anti-CD40 antibody comprises the CDRs of an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4.
  • the anti-CD40 antibody comprises the heavy chain and light chain variable regions of an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4.
  • the anti-CD40 antibody is an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4.
  • the CD40 agonist is recombinant CD40L.
  • the at least one immune stimulating agent comprises a CD40 agonist and at least one additional immune stimulating agent from any of those described above.
  • any one or more of the above immune stimulating agents above may be combined with any one or more of the anti-CSF1R antibodies described herein as well as with a CD40 agonist, such as a CD40 agonist antibody or recombinant CD40L, such as any one of the anti-CD40 antibodies described above.
  • the anti-CSF1R antibody and the at least one immune stimulatory agent are administered concurrently or sequentially. In some embodiments, the anti-CSF1R antibody and the at least one immune stimulatory agent are administered concurrently. In some embodiments, one or more doses of the at least one immune stimulatory agent are administered prior to administering an anti-CSF1R antibody. In some embodiments, the subject received a complete course of therapy with the at least one immune stimulatory agent prior to administration of the anti-CSF1R antibody. In some embodiments, the anti-CSF1R antibody is administered during a second course of therapy with the at least one immune stimulatory agent.
  • the subject received at least one, at least two, at least three, or at least four doses of the at least one immune stimulatory agent prior to administration of the anti-CSF1R antibody. In some embodiments, at least one dose of the at least one immune stimulatory agent is administered concurrently with the anti-CSF1R inhibitor. In some embodiments, one or more doses of the anti-CSF1R antibody are administered prior to administering at least one immune stimulatory agent. In some embodiments, the subject received at least two, at least three, or at least four doses of the anti-CSF1R antibody prior to administration of at least one immune stimulatory agent. In some embodiments, at least one dose of the anti-CSF1R antibody is administered concurrently with the at least one immune stimulatory agent.
  • the cancer is selected from non-small cell lung cancer, melanoma, squamous cell carcinoma of the head and neck, ovarian cancer, pancreatic cancer, renal cell carcinoma, hepatocellular carcinoma, bladder cancer, endometrial cancer, Hodgkin's lymphoma, lung cancer, glioma, gioblastoma multiforme, colon cancer, breast cancer, bone cancer, skin cancer, uterince cancer, gastric cancer, stomach cancer, lymphoma, lymphocytic leukemia, multiple myeloma, prostate cancer, mesothelioma, and kidney cancer.
  • the cancer is recurrent or progressive after a therapy selected from surgery, chemotherapy, radiation therapy, or a combination thereof.
  • compositions comprising an anti-CSF1R antibody and at least one immune stimulatory agent.
  • the at least one immune stimulating agent comprises an antagonist of an inhibitor of the activation of T cells, while in some embodiments, the at least one immune stimulating agent comprises comprises an agonist of a stimulator of the activation of T cells.
  • the at least one immune stimulating agent comprises an antagonist of CTLA4, LAG-3, Galectin 1, Galectin 9, CEACAM-1, BTLA, CD25, CD69, TIGIT, CD113, GPR56, VISTA, B7-H3, B7-H4, 2B4, CD48, GARP, PD1H, LAIR1, TIM1, TIM3, TIM4, ILT4, IL-6, IL-10, TGF ⁇ , VEGF, KIR, LAG-3, adenosine A2A receptor, PI3Kdelta, or IDO.
  • the at least one immune stimulating agent comprises an agonist of B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD27, CD40, CD40L, DR3, CD28H, IL-2, IL-7, IL-12, IL-15, IL-21, IFN ⁇ , STING, or a Toll-like receptor agonist such as a TLR2/4 agonist.
  • the at least one immune stimulating agent comprises an agent that binds to a member of the B7 family of membrane-bound proteins such as B7-1, B7-2, B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6.
  • B7-1, B7-2, B7-H2 ICOS-L
  • B7-H3, B7-H4, B7-H5 VISTA
  • B7-H6 B7-H6
  • the at least one immune stimulating agent comprises an agent that binds to a member of the TNF receptor family or a co-stimulatory or co-inhibitory molecule binding to a member of the TNF receptor family such as CD40, CD40L, OX40, OX40L, GITR, GITRL, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, EDA1, EDA2, TACI, APRIL, BCMA, LT ⁇ R, LIGHT, DeR3, HVEM, VEGL/TL1A, TRAMP/DR3, TNFR1, TNF ⁇ , TNFR2, TNF ⁇ , 1 ⁇ 2, FAS, FASL, RELT, DR6,
  • the at least one immune stimulating agent comprises an agent that antagonizes or inhibits a cytokine that inhibits T cell activation such as IL-6, IL-10, TGF ⁇ , VEGF.
  • the at least one immune stimulating agent comprises an agonist of a cytokine that stimulates T cell activation such as IL-2, IL-7, IL-12, IL-15, IL-21, and IFN ⁇ .
  • the at least one immune stimulating agent comprises an antagonist of a chemokine, such as CXCR2, CXCR4, CCR2, or CCR4.
  • the at least one immune stimulating agent comprises an antibody.
  • the at least one immune stimulating agent may comprise a vaccine, such as a mesothelin-targeting vaccine or attenuated listeria cancer vaccine such as CRS-207.
  • compositions comprise any one or more of the above antagonists, agonists, and binding agents combined with any one or more of the anti-CSF1R antibodies described herein.
  • the compositions may include each therapeutic agent in a separate container or compartment or alternatively, may include two or more of the therapeutic agents mixed together.
  • the compositions comprise an anti-CSF1R antibody and a CD40 agonist, optionally along with at least one other immune stimulating agent as listed above.
  • the CD40 agonist is an anti-CD40 antibody.
  • the anti-CD40 antibody comprises the CDRs of an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4.
  • the anti-CD40 antibody comprises the heavy chain and light chain variable regions of an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4.
  • the anti-CD40 antibody is an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4.
  • the CD40 agonist is recombinant CD40L.
  • the compositions comprise any one or more of the above immune stimulating agents above combined with both any one or more of the anti-CSF1R antibodies described herein as well as with a CD40 agonist, such as a CD40 agonist antibody or recombinant CD40L, such as any one of the anti-CD40 antibodies described above.
  • the compositions may include each therapeutic agent in a separate container or compartment or alternatively, may include two or more of the therapeutic agents mixed together.
  • the antibody heavy chain and/or the antibody light chain of the anti-CSF1R antibody may have the structure described below.
  • the anti-CSF1R 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 anti-CSF1R 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 anti-CSF1R 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 anti-CSF1R 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 anti-CSF1R antibody 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 anti-CSF1R antibody 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 anti-CSF1R antibody 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 anti-CSF1R antibody 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; (a) a heavy chain comprising a sequence
  • the anti-CSF1R 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
  • the anti-CSF1R 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 anti-CSF1R antibody may be a humanized antibody. In any of the compositions or methods described herein, the anti-CSF1R antibody may be selected from a Fab, an Fv, an scFv, a Fab′, and a (Fab′) 2 . In any of the compositions or methods described herein, the anti-CSF1R antibody may be a chimeric antibody. In any of the compositions or methods described herein, the anti-CSF1R antibody may be selected from an IgA, an IgG, and an IgD. In any of the compositions or methods described herein, the anti-CSF1R antibody may be an IgG. In any of the methods described herein, the antibody may be an IgG1 or IgG2.
  • the anti-CSF1R antibody may bind to human CSF1R and/or binds to cynomolgus CSF1R. In any of the compositions or methods described herein, the anti-CSF1R antibody may block ligand binding to CSF1R. In any of the compositions or methods described herein, the anti-CSF1R antibody may block binding of CSF1 and/or IL-34 to CSF1R. In any of the compositions or methods described herein, the anti-CSF1R antibody may block binding of both CSF1 and IL-34 to CSF1R.
  • the anti-CSF1R antibody may inhibit ligand-induced CSF1R phosphorylation. In any of the compositions or methods described herein, the anti-CSF1R antibody may inhibit CSF1- and/or IL-34-induced CSF1R phosphorylation. In any of the compositions or methods described herein, the anti-CSF1R antibody may bind to human CSF1R with an affinity (KD) of less than 1 nM. In any of the compositions or methods described herein, the anti-CSF1R antibody may inhibit monocyte proliferation and/or survival responses in the presence of CSF1 or IL-34.
  • 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.
  • FIG. 3 shows that the combination of anti-CSF1R antibody and anti-CD40 antibody demonstrate greater tumor growth suppression in an MC38 tumor mouse model than either therapy alone.
  • FIG. 4A-B shows the tumor volume of individual mice at ( FIG. 4A ) day 11 and ( FIG. 4B ) day 13.
  • the combination of anti-CSF1R antibody and anti-CD40 antibody performed significantly better than either therapy alone at both time points.
  • FIG. 5 shows body weight in mice used in the study.
  • TAMs Tumor-associated macrophages
  • TAMs can suppress an anti-tumor responses through multiple mechanisms.
  • TAMs express anti-inflammatory cytokines such as TGF and IL-10 which acts to suppress the ability of intratumoral dendritic cells to stimulate cytotoxic T cell responses (Ruffell et al., 2014, Cancer Cell ).
  • TAMs also express chemokines which recruit immunosuppressive regulatory T cells into tumors (Curiel et al., 2004, Nature Med.; Mizukami et al., 2008, Int. J.
  • TAMs express the ligands for the T cell inhibitor receptors PD-1 and CTLA-4 which act to directly inhibit T cell activation and funnction.
  • Inhibition of CSF1R can reduce immunosuppressive TAMs in mouse models and human tumors. See, e.g., Ries et al., 2014, Cancer Cell, 25: 846-859; Pyontech et al., 2013, Nature Med., 19: 1264-1272; and Zhu et al., 2014, Cancer Res., 74: 5057-5069.
  • immune stimulating agents work by stimulating an immune response.
  • 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.
  • immune stimulating agent refers to a molecule that stimulates the immune system by either acting as an agonist of an immune-stimulatory molecule, including a co-stimulatory molecule, or acting as an antagonist of an immune inhibitory molecule, including a co-inhibitory molecule.
  • An immune stimulating agent may be a biologic, such as an antibody or antibody fragment, other protein, or vaccine, or may be a small molecule drug.
  • An “immune stimulatory molecule” includes a receptor or ligand that acts to enhance, stimulate, induce, or otherwise “turn-on” an immune response. Immune stimulatory molecules as defined herein include co-stimulatory molecules.
  • Immune inhibitory molecule includes a receptor or ligand that acts to reduce, inhibit, suppress, or otherwise “turn-off” an immune response.
  • Immune inhibitory molecules as defined herein include co-inhibitory molecules.
  • Such immune stimulatory and immune inhibitory molecules may be, for example, receptors or ligands found on immune cells such as a T cells, or found on cells involved in innate immunity such as NK cells.
  • B-cell surface antigen CD40 and “CD40” refer herein to the full-length CD40, which includes the N-terminal ECD, the transmembrane domain, and the intracellular domain, with or without an N-terminal leader sequence.
  • the CD40 is a human CD40 having the amino acid sequence of SEQ ID NO: 96 (precursor, with signal sequence) or SEQ ID NO: 97 (mature, without signal sequence).
  • CD40 agonist refers to a moiety that interacts with CD40 and enhances CD40 activity.
  • Nonlimiting exemplary CD40 activities include signaling through CD40, enhancement of antigen presenting activity, induction of proinflammatory cytokines, and induction of tumorcidal activity.
  • a CD40 agonist is an anti-CD40 antibody.
  • anti-CD40 antibody refers to an antibody that specifically binds to CD40. Unless specifically indicated otherwise, the term “anti-CD40 antibody” as used herein refers to an anti-CD40 agonist antibody.
  • 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.
  • 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 a constant region is an IgA antibody.
  • an antibody comprising a ⁇ constant region is an IgM antibody
  • an antibody comprising an c 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 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 cancer 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 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.
  • cancer is used herein to refer to a group of cells that exhibit abnormally high levels of proliferation and growth.
  • a cancer may be benign (also referred to as a benign tumor), pre-malignant, or malignant.
  • Cancer cells may be solid cancer cells or leukemic cancer cells.
  • cancer growth is used herein to refer to proliferation or growth by a cell or cells that comprise a cancer that leads to a corresponding increase in the size or extent of the cancer.
  • cancer examples include but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia. More particular nonlimiting examples of such cancers include squamous cell cancer, small-cell lung cancer, pituitary cancer, esophageal cancer, astrocytoma, soft tissue sarcoma, non-small cell lung cancer (including squamous cell non-small cell lung cancer), adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney cancer, renal cell carcinoma, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, brain cancer, endometrial cancer, testis cancer, cholangiocarcinom
  • recurrent cancer refers to a cancer that has returned after a previous treatment regimen, following which there was a period of time during which the cancer could not be detected.
  • progressive cancer is a cancer that has increased in size or tumor spread since the beginning of a treatment regimen.
  • a progressive cancer is a cancer that has incrased in size or tumor spread by at least 10%, at least 20%, at least 30%, at least 40%, or at least 50% since the beginning of a treatment regimen.
  • chemotherapeutic agent is a chemical compound useful in the treatment of cancer.
  • examples of chemotherapeutic agents include, but are not limited to, alkylating agents such as thiotepa and Cytoxan® cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic ana
  • dynemicin including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, Adriamycin® doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, es
  • chemotherapeutic agents include anti-hormonal agents that act to regulate or inhibit hormone action on cancers such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including Nolvadex® tamoxifen), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and Fareston® toremifene; aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, Megase® megestrol acetate, Aromasin® exemestane, formestanie, fadrozole, Rivisor® vorozole, Femara® letrozole, and Arimidex® anastrozole; and anti-androgens such as flutamide,
  • an “anti-angiogenesis agent” or “angiogenesis inhibitor” refers to a small molecular weight substance, a polynucleotide (including, e.g., an inhibitory RNA (RNAi or siRNA)), a polypeptide, an isolated protein, a recombinant protein, an antibody, or conjugates or fusion proteins thereof, that inhibits angiogenesis, vasculogenesis, or undesirable vascular permeability, either directly or indirectly.
  • RNAi or siRNA inhibitory RNA
  • the anti-angiogenesis agent includes those agents that bind and block the angiogenic activity of the angiogenic factor or its receptor.
  • an anti-angiogenesis agent is an antibody or other antagonist to an angiogenic agent, e.g., antibodies to VEGF-A (e.g., bevacizumab (Avastin®)) or to the VEGF-A receptor (e.g., KDR receptor or Flt-1 receptor), anti-PDGFR inhibitors such as Gleevec® (Imatinib Mesylate), small molecules that block VEGF receptor signaling (e.g., PTK787/ZK2284, SU6668, Sutent®/SU11248 (sunitinib malate), AMG706, or those described in, e.g., international patent application WO 2004/113304).
  • an angiogenic agent e.g., antibodies to VEGF-A (e.g., bevacizumab (Avastin®)) or to the VEGF-A receptor (e.g., KDR receptor or Flt-1 receptor), anti-PDGFR inhibitors such as Gleeve
  • Anti-angiogensis agents also include native angiogenesis inhibitors , e.g., angiostatin, endostatin, etc. See, e.g., Klagsbrun and D'Amore (1991) Annu. Rev. Physiol. 53:217-39; Streit and Detmar (2003) Oncogene 22:3172-3179 (e.g., Table 3 listing anti-angiogenic therapy in malignant melanoma); Ferrara & Alitalo (1999) Nature Medicine 5(12):1359-1364; Tonini et al. (2003) Oncogene 22:6549-6556 (e.g., Table 2 listing known anti-angiogenic factors); and, Sato (2003) Int. J. Clin. Oncol. 8:200-206 (e.g., Table 1 listing anti-angiogenic agents used in clinical trials).
  • native angiogenesis inhibitors e.g., angiostatin, endostatin, etc. See, e.g., Klags
  • a “growth inhibitory agent” as used herein refers to a compound or composition that inhibits growth of a cell (such as a cell expressing VEGF) either in vitro or in vivo.
  • the growth inhibitory agent may be one that significantly reduces the percentage of cells (such as a cell expressing VEGF) in S phase.
  • growth inhibitory agents include, but are not limited to, agents that block cell cycle progression (at a place other than S phase), such as agents that induce G1 arrest and M-phase arrest.
  • Classical M-phase blockers include the vincas (vincristine and vinblastine), taxanes, and topoisomerase II inhibitors such as doxorubicin, epirubicin, daunorubicin, etoposide, and bleomycin.
  • Those agents that arrest G1 also spill over into S-phase arrest, for example, DNA alkylating agents such as tamoxifen, prednisone, dacarbazine, mechlorethamine, cisplatin, methotrexate, 5-fluorouracil, and ara-C.
  • Taxanes are anticancer drugs both derived from the yew tree.
  • Docetaxel (Taxotere®, Rhone-Poulenc Rorer), derived from the European yew, is a semisynthetic analogue of paclitaxel (Taxol®, Bristol-Myers Squibb). Paclitaxel and docetaxel promote the assembly of microtubules from tubulin dimers and stabilize microtubules by preventing depolymerization, which results in the inhibition of mitosis in cells.
  • anti-neoplastic composition refers to a composition useful in treating cancer comprising at least one active therapeutic agent.
  • therapeutic agents include, but are not limited to, e.g., chemotherapeutic agents, growth inhibitory agents, cytotoxic agents, agents used in radiation therapy, anti-angiogenesis agents, cancer immunotherapeutic agents, apoptotic agents, anti-tubulin agents, and other-agents to treat cancer, such as anti-HER-2 antibodies, anti-CD20 antibodies, an epidermal growth factor receptor (EGFR) antagonist (e.g., a tyrosine kinase inhibitor), HER1/EGFR inhibitor (e.g., erlotinib (Tarceve), platelet derived growth factor inhibitors (e.g., Gleevec® (Imatinib Mesylate)), a COX-2 inhibitor (e.g., celecoxib), interferons, CTLA4 inhibitors (e.g., anti-CTLA antibody ipilimumab
  • 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.
  • 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 and/or an immune stimulating agent 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 antibody or antibodies to elicit a desired response in the individual.
  • a therapeutically effective amount encompasses an amount in which any toxic or detrimental effects of the antibody or antibodies are outweighed by the therapeutically beneficial effects.
  • the expression “effective amount” refers to an amount of the antibody that is effective for treating the cancer.
  • 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 (sequential) 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.
  • 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 (KD) 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.
  • KD binding affinity
  • an anti-CSF1R antibody binds to the extracellular domain of CSF1R (CSF1R-ECD).
  • an anti-CSF1R antibody has a binding affinity (KD) for CSF1R of less than 1 nM, less than 0.5 nM, less than 0.1 nM, or less than 0.05 nM.
  • KD binding affinity
  • an anti-CSF1R antibody has a KD 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.
  • Immune stimulating agents may include, for example, a small molecule drug, antibody or fragment thereof, or other biologic or small molecule.
  • biologic immune stimulating agents include, but are not limited to, antibodies, antibody fragments, fragments of receptor or ligand polypeptides, for example that block receptor-ligand binding, vaccines and cytokines.
  • the antibody is a monoclonal antibody. In certain aspects, the monoclonal antibody is humanized or human antibody.
  • the at least one immune stimulating agent comprises an agonist of an immune stimulatory molecule, including a co-stimulatory molecule, while in some embodiments, the at least one immune stimulating agent comprises an antagonist of an immune inhibitory molecule, including a co-inhibitory molecule. In some embodiments, the at least one immune stimulating agent comprises an agonist of an immune-stimulatory molecule, including a co-stimulatory molecule, found on immune cells, such as T cells. In some embodiments, the at least one immune stimulating agent comprises an antagonist of an immune inhibitory molecule, including a co-inhibitory molecule, found on immune cells, such as T cells.
  • the at least one immune stimulating agent comprises an agonist of an immune stimulatory molecule, including a co-stimulatory molecule, found on cells involved in innate immunity, such as NK cells.
  • the at least one immune stimulating agent comprises an antagonist of an immune inhibitory molecule, including a co-inhibitory molecule, found on cells involved in innate immunity, such as NK cells.
  • the combination enhances the antigen-specific T cell response in the treated subject and/or enhances the innate immunity response in the subject.
  • the combination results in an improved anti-tumor response in an animal cancer model, such as a xenograft model, compared to administration of either the anti-CSF1R antibody or immune stimulating agent alone.
  • the combination results in a synergistic response in an animal cancer model, such as a xenograft model, compared to administration of either the anti-CSF1R antibody or immune stimulating agent alone.
  • an immune stimulating agent targets a stimulatory or inhibitory molecule that is a member of the immunoglobulin super family (IgSF).
  • an immune stimulating agent may be an agent that targets (or binds specifically to) a member of the B7 family of membrane-bound ligands, which includes B7-1, B7-2, B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6, or a co-stimulatory or co-inhibitory receptor binding specifically to a B7 family member.
  • An immune stimulating agent may be an agent that targets a member of the TNF family of membrane bound ligands or a co-stimulatory or co-inhibitory receptor binding specifically to a member of the TNF family.
  • Exemplary TNF and TNFR family members that may be targeted by immune stimulating agents include CD40 and CD40L, OX-40, OX-40L, GITR, GITRL, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DRS, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LT ⁇ R, LIGHT, DcR3, HVEM, VEGI/TL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFR1,
  • an immune stimulating agent may comprise (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitor) such as CTLA-4, LAG-3, TIM3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, B7-H3, B7-H4, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, TIM-4, and ILT4 and/or may comprise (ii)an agonist of a protein that stimulates T cell activation such as B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD70, CD27, CD40, CD40L, DR3 and CD28H.
  • an antagonist of a protein that inhibits T cell activation e.g., immune checkpoint inhibitor
  • an immune stimulating agent may comprise an agent that inhibit or is an antagonist of a cytokine that inhibits T cell activation (e.g., IL-6, IL-10, TGF- ⁇ , VEGF, and other immunosuppressive cytokines), and it some embodiments an immune stimulating agent may comprise an agent that is an agonist of a cytokine, such as IL-2, IL-7, IL-12, IL-15, IL-21 and IFN ⁇ (e.g., the cytokine itself) that stimulates T cell activation.
  • immune stimulating agents may comprise an antagonist of a chemokine, such as CXCR2 (e.g., MK-7123), CXCR4 (e.g. AMD3100), CCR2, or CCR4 (mogamulizumab).
  • immune stimulating agents may include antagonists of inhibitory receptors on NK cells or agonists of activating receptors on NK cells.
  • an anti-CSF1R antibody can be combined with an antagonist of KIR, optionally along with at least one other immune stimulating agent such as an agonist of CD40.
  • Immune stimulating agents may also include agents that inhibit TGF- ⁇ signaling, agents that enhance tumor antigen presentation, e.g., dendritic cell vaccines, GM-CSF secreting cellular vaccines, CpG oligonucleotides,and imiquimod, or therapies that enhance the immunogenicity of tumor cells (e.g., anthracyclines).
  • agents that enhance tumor antigen presentation e.g., dendritic cell vaccines, GM-CSF secreting cellular vaccines, CpG oligonucleotides,and imiquimod
  • therapies that enhance the immunogenicity of tumor cells e.g., anthracyclines.
  • Immune stimulating agents may also include certain vaccines such as mesothelin-targeting vaccines or attenuated listeria cancer vaccines, such as CRS-207.
  • Immune stimulating agents may also comprise agents that deplete or block Treg cells, such as agents that specifically bind to CD25.
  • Immune stimulating agents may also comprise agents that inhibit a metabolic enzyme such as indoleamine dioxigenase (IDO), dioxigenase, arginase, or nitric oxide synthetase.
  • IDO indoleamine dioxigenase
  • dioxigenase dioxigenase
  • arginase arginase
  • nitric oxide synthetase a metabolic enzyme
  • Immune stimulating agents may also comprise agents that inhibit the formation of adenosine or inhibit the adenosine A2A receptor.
  • Immune stimulating agents may also comprise agents that reverse/prevent T cell anergy or exhaustion and agents that trigger an innate immune activation and/or inflammation at a tumor site.
  • An anti-CSF1R antibody may be combined with more than one immune stimulating agent, such as a CD40 agonist and at least one additional immune stimulating agent.
  • the anti-CSF1R antibody optionally along with a CD40 agonist, may be combined with a combinatorial approach that targets multiple elements of the immune pathway, such as one or more of the following: at least one agent that enhances tumor antigen presentation (e.g., dendritic cell vaccine, GM-CSF secreting cellular vaccines, CpG oligonucleotides, imiquimod); at least one agent that inhibits negative immune regulation e.g., by inhibiting CTLA-4 pathway and/or depleting or blocking Treg or other immune suppressing cells; a therapy that stimulates positive immune regulation, e.g., with agonists that stimulate the CD-137, OX-40 and/or GITR pathway and/or stimulate T cell effector function; at least one agent that increases systemically the frequency of anti-tumor T cells; a therapy that depletes or inhibits Tre
  • an anti-CSF1R antibody can be used with one or more agonistic agents that ligate positive costimulatory receptors; one or more antagonists (blocking agents) that attenuate signaling through inhibitory receptors, such as antagonists that overcome distinct immune suppressive pathways within the tumor microenvironment; one or more agents that increase systemically the frequency of anti-tumor immune cells, such as T cells, deplete or inhibit Tregs (e.g., by inhibiting CD25); one or more agents that inhibit metabolic enzymes such as IDO; one or more agents that reverse/prevent T cell anergy or exhaustion; and one or more agents that trigger innate immune activation and/or inflammation at tumor sites.
  • agonistic agents that ligate positive costimulatory receptors
  • antagonists blocking agents that attenuate signaling through inhibitory receptors, such as antagonists that overcome distinct immune suppressive pathways within the tumor microenvironment
  • agents that increase systemically the frequency of anti-tumor immune cells such as T cells, deplete or inhibit Tregs (e.
  • the at least one immune stimulating agent comprises a CTLA-4 antagonist, such as an antagonistic CTLA-4 antibody.
  • CTLA-4 antibodies include, for example, YERVOY (ipilimumab) or tremelimumab.
  • the at least one immune stimulating agent comprises a LAG-3 antagonist, such as an antagonistic LAG-3 antibody.
  • LAG3 antibodies include, for example, BMS-986016 (WO10/19570, WO14/08218), or IMP-731 or IMP-321 (WO08/132601, WO09/44273).
  • the at least one immune stimulating agent comprises a CD137 (4-1BB) agonist, such as an agonistic CD137 antibody.
  • Suitable CD137 antibodies include, for example, urelumab or PF-05082566 (WO12/32433).
  • the at least one immune stimulating agent comprises a GITR agonist, such as an agonistic GITR antibody.
  • GITR antibodies include, for example, TRX-518 (WO06/105021, WO09/009116), MK-4166 (WO11/028683) or a GITR antibody disclosed in WO2015/031667.
  • the at least one immune stimulating agent comprises an OX40 agonist, such as an agonistic OX40 antibody.
  • OX40 antibodies include, for example, MEDI-6383, MEDI-6469 or MOXRO916 (RG7888; WO06/029879).
  • the at least one immune stimulating agent comprises a CD27 agonist, such as an agonistic CD27 antibody.
  • Suitable CD27 antibodies include, for example, varlilumab (CDX-1127).
  • the at least one immune stimulating agent comprises MGA271, which targets B7H3 (WO11/109400).
  • the at least one immune stimulating agent comprises a MR antagonist, such as lirilumab.
  • the at least one immune stimulating agent comprises an IDO antagonist.
  • IDO antagonists include, for example, INCB-024360 (WO2006/122150, WO07/75598, WO08/36653, WO08/36642), indoximod, NLG-919 (WO09/73620, WO09/1156652, WO11/56652, WO12/142237) or F001287.
  • the at least one immune stimulating agent comprises a Toll-like receptor agonist, e.g., a TLR2/4 agonist (e.g., Bacillus Calmette-Guerin); a TLR7 agonist (e.g., Hiltonol or Imiquimod); a TLR7/8 agonist (e.g., Resiquimod); or a TLR9 agonist (e.g., CpG7909).
  • a TLR2/4 agonist e.g., Bacillus Calmette-Guerin
  • TLR7 agonist e.g., Hiltonol or Imiquimod
  • TLR7/8 agonist e.g., Resiquimod
  • a TLR9 agonist e.g., CpG7909
  • the at least one immune stimulating agent comprises a TGF- ⁇ inhibitor, e.g., GC1008, LY2157299, TEW7197 or IMC-TR1.
  • a TGF- ⁇ inhibitor e.g., GC1008, LY2157299, TEW7197 or IMC-TR1.
  • the at least one immune stimulating agent comprises a CD40 agonist, optionally together with at least one additional immune stimulating agent as described herein.
  • the cell surface molecule CD40 is a member of the tumor necrosis factor receptor superfamily and is expressed by antigen presenting cells such as dentritic cells, B cells, macrophages and monocytes and is also expressed on other cell types, including immune, hematopoietic, vascular, and epithelial cells, as well as on various tumor cells.
  • antigen presenting cells such as dentritic cells, B cells, macrophages and monocytes and is also expressed on other cell types, including immune, hematopoietic, vascular, and epithelial cells, as well as on various tumor cells.
  • CD40 signaling results in activation and upregulation of T cell costimulatory molecules and other critical immune mediators required for the induction of an immune response.
  • CD40-targeting therapies have undergone phase 1 clinical evaluation in advanced-stage cancer patients, and initial findings have shown efficacy in the absence of major toxicity.
  • CD40 for example, animal models have shown that ligation of CD40 on dendritic cells results in activation of cytotoxic T lymphocytes that mediate tumor killing (Marzo et al., 2000, J. Immunol.; Todryk et al., 2001, J. Immunol. Methods. ) Activation of CD40 on macrophages results in tumoralcidal activity (Beatty et al., 2011, Science ), and cytokines produced from CD40 stimulated antigen presenting cells leads to the activation of natural killer cells important for tumor eradication. Given the complex nature of an anti-tumor immune response, effective cancer therapy may require combining multiple immunotherapy agents.
  • tumors that have CSF1R-expressing TAMs may be sensitive to combination therapy with an anti-CSF1R antibody and a CD40 agonist.
  • Exemplary CD40 agonists of the compositions and methods of this invention include, for example, anti-CD40 antibodies that enhance CD40 activity.
  • Such antibodies may be humanized antibodies, chimeric antibodies, mouse antibodies, human antibodies, and antibodies comprising the heavy chain and/or light chain CDRs of an anti-CD40 antibody iscussed herein.
  • Nonlimiting exemplary agonist anti-CD40 antibodies include, but are not limited to, CP-870,893 (Pfizer and VLST; antibody 21.4.1 in EP 1 476 185 B1 and U.S. Pat. No. 7,338,660; see also clinicaltrials.gov/ct2/show/NCT02225002); dacetuzumab (Seattle Genetics; SEQ ID NOs: 98 and 99 herein; see also U.S. Pat. No. 6,946,129 and U.S. Pat. No.
  • Exemplary CD40 agonists also include recombinant CD40L.
  • an 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 an antibody are provided.
  • a nucleic acid molecule comprises a polynucleotide that encodes a heavy chain or a light chain of an antibody.
  • a nucleic acid molecule comprises both a polynucleotide that encodes a heavy chain and a polynucleotide that encodes a light chain, of an 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 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 antibody heavy chains and/or light chains are provided. Vectors comprising polynucleotides that encode antibody heavy chains and/or 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.
  • 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. Prog. 20:880-889 (2004).
  • a vector is chosen for in vivo expression of antibody heavy chains and/or antibody 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.
  • antibody heavy chains and/or 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.
  • antibody heavy chains and/or light chains may be expressed in yeast. See, e.g., U.S. Publication No.
  • a particular eukaryotic host cell is selected based on its ability to make desired post-translational modifications to the antibody heavy chains and/or 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.
  • 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 antigen 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 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 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).
  • methods for treating cancer comprising administering an effective amount of an anti-CSF1R antibody and an effective amount of at least one immune stimulating agent.
  • the anti-CSF1R antibody and the at least one immune stimulating agent are administered concurrently.
  • the therapeutics may be infused together or injected at roughly the same time.
  • the anti-CSF1R antibody and the at least one immune stimulating agent administered sequentially.
  • the anti-CSF1R antibody is administered sequentially before or after at least one immune stimulating agent such that the two therapeutics are administered 30 minutes, 60 minutes, 90 minutes, 120 minutes, 3 hours, 6 hours, 12 hours, 24 hours, 36 hours, 48 hours, 3 days, 5 days, 7 days, or two weeks apart.
  • At least one, at least two, at least three doses, at least five doses, or at least ten doses of an anti-CSF1R antibody is administered prior to administration of at least one immune stimulating agent. In some embodiments, at least one, at least two, at least three doses, at least five doses, or at least ten doses of at least one immune stimulating agent is administered prior to administration of an anti-CSF1R antibody. In some embodiments, the last dose of immune stimulating agent is administered at least one, two, three, five, days or ten, or one, two, three, five, twelve, or twenty four weeks prior to the first dose of CSFR1 inhibitor.
  • the last dose of CSFR1 inhibitor is administered at least one, two, three, five, days or ten, or one, two, three, five, twelve, or twenty four weeks prior to the first dose of at least one immune stimulating agent.
  • a subject has received, or is receiving, therapy with at least one immune stimulating agent, and an anti-CSF1R antibody is added to the therapeutic regimen.
  • a method of selecting a patient for combination therapy with an anti-CSF1R antibody and a at least one immune stimulating agent, such as a CD40 agonist comprising determining the levels of TAMs and/or CD8+ T cells in the patient. In some embodiments, if a patient's TAM levels are high, the patient is selected for combination therapy. In some embodiments, if a patient's TAM and CD8+ T cell levels are high, the patient is selected for combination therapy.
  • the level of TAMs or CD8+ T cells is considered “high” if it is at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 75%, or at least 100% higher than the level in an individual who does not have cancer.
  • the level of TAMs or CD8+ T cells is considered “high” if it is above the median level found in individuals with cancer. In some embodiments, if a patient's TAM levels are high and CD8+ T cell levels are low, the patient is selected for combination therapy with an anti-CSF1R antibody and at least one immune stimulating agent, such as a CD40 agonist.
  • the level of CD8+ T cells is considered “low” if it is at or below the median level found in individuals with cancer. In some embodiment, the level of CD8+ T cells is considered “low” if it is at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 75%, or at least 100% lower than the level in an individual who does not have cancer.
  • expression of CSF1R on the patient's TAMs is determined. In some embodiments, if the patient's TAMs express CSF1R, the patient is selected for combination therapy. In some embodiments, if the patient's TAMs express elevated levels of CSF1R, the patient is selected for combination therapy. In some embodiments, a patient's TAMs are considered to express “elevated” levels of CSF1R if the level of CSF1R is at or above the median level of CSF1R found expressed on TAMS in individuals with cancer. In some embodiments, if the patient's CSF1R expression shows a high correlation with the level of CD8+ T cells, the patient is selected for combination therapy. The correlation of the expressions is considered “high” if it is at or above the median level found in individuals with cancer.
  • TAMs levels of TAMs, CSF1R expression, CD8+ T cells, and/or regulatory T cells may be measured by methods in the art.
  • Nonexemplary methods include immunohistochemistry (IHC), fluorescence-activated cell sorting (FACS), protein arrays, and gene expression assays, such as RNA sequencing, gene arrays, and quantitative PCR.
  • IHC immunohistochemistry
  • FACS fluorescence-activated cell sorting
  • protein arrays such as RNA sequencing, gene arrays, and quantitative PCR.
  • gene expression assays such as RNA sequencing, gene arrays, and quantitative PCR.
  • one or more markers selected from CSF1R, CD68, CD163, CD8, and FoxP3 may be detected by IHC, FACS, or gene expression assay on tumor sections, or dissociated cells from tumor sections.
  • the cancer is selected from squamous cell cancer, small-cell lung cancer, pituitary cancer, esophageal cancer, astrocytoma, soft tissue sarcoma, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney cancer, renal cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, brain cancer, endometrial cancer, testis cancer, cholangiocarcinoma, gallbladder carcinoma, gastric cancer, melanoma, and various types of head and neck cancer.
  • lung cancer is non-small cell lung cancer or lung squamous cell carcinoma.
  • leukemia is acute myeloid leukemia or chronic lymphocytic leukemia.
  • breast cancer is breast invasive carcinoma.
  • ovarian cancer is ovarian serous cystadenocarcinoma.
  • kidney cancer is kidney renal clear cell carcinoma.
  • colon cancer is colon adenocarcinoma.
  • bladder cancer is bladder urothelial carcinoma.
  • the cancer is selected from bladder cancer, cervical cancer (such as squamous cell cervical cancer), head and neck squamous cell carcinoma, rectal adenocarcinoma, non-small cell lung cancer, endometrial cancer, prostate adenocarcinoma, colon cancer, ovarian cancer (such as serous epithelial ovarian cancer), and melanoma.
  • cervical cancer such as squamous cell cervical cancer
  • head and neck squamous cell carcinoma rectal adenocarcinoma
  • non-small cell lung cancer endometrial cancer
  • prostate adenocarcinoma colon cancer
  • ovarian cancer such as serous epithelial ovarian cancer
  • melanoma such as serous epithelial ovarian cancer
  • the anti-CSF1R antibody blocks binding of CSF1 and/or IL-34 to CSF1R and/or inhibits CSF1R phosphorylation induced by CSF1 and/or IL-34. In some embodiments, the anti-CSF1R antibody locks binding of CSF1 and IL-34 to CSF1R and/or inhibits CSF1R phosphorylation induced by CSF1 and/or IL-34. In some embodiments, the anti-CSF1R antibody comprises the CDRs of, or the variable regions of, an antibody selected from huAb1 to huAb16, described herein. In some embodiments, the anti-CSF1R antibody comprises the CDRs of, or the variable regions of, huAb1.
  • the at least one immune stimulating agent comprises an antagonist of an inhibitor of the activation of T cells, while in some embodiments, the at least one immune stimulating agent comprises comprises an agonist of a stimulator of the activation of T cells.
  • the at least one immune stimulating agent comprises an antagonist of CTLA4, LAG-3, Galectin 1, Galectin 9, CEACAM-1, BTLA, CD25, CD69, TIGIT, CD113, GPR56, VISTA, B7-H3, B7-H4, 2B4, CD48, GARP, PD1H, LAIR1, TIM1, TIM3, TIM4, ILT4, IL-6, IL-10, TGF ⁇ , VEGF, KIR, LAG-3, adenosine A2A receptor, PI3Kdelta, or IDO.
  • the at least one immune stimulating agent comprises an agonist of B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD27, CD40, CD40L, DR3, CD28H, IL-2, IL-7, IL-12, IL-15, IL-21, IFN ⁇ , STING, or a Toll-like receptor agonist such as a TLR2/4 agonist.
  • the at least one immune stimulating agent comprises an agent that binds to a member of the B7 family of membrane-bound proteins such as B7-1, B7-2, B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6.
  • B7-1, B7-2, B7-H2 ICOS-L
  • B7-H3, B7-H4, B7-H5 VISTA
  • B7-H6 B7-H6
  • the at least one immune stimulating agent comprises an agent that binds to a member of the TNF receptor family or a co-stimulatory or co-inhibitory molecule binding to a member of the TNF receptor family such as CD40, CD40L, OX40, OX40L, GITR, GITRL, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DRS, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, EDA1, EDA2, TACI, APRIL, BCMA, LT ⁇ R, LIGHT, DeR3, HVEM, VEGL/TL1A, TRAMP/DR3, TNFR1, TNF ⁇ , TNFR2, TNF ⁇ , 1 ⁇ 2, FAS, FASL, RELT, DR
  • the at least one immune stimulating agent comprises an agent that antagonizes or inhibits a cytokine that inhibits T cell activation such as IL-6, IL-10, TGF ⁇ , VEGF.
  • the at least one immune stimulating agent comprises an agonist of a cytokine that stimulates T cell activation such as IL-2, IL-7, IL-12, IL-15, IL-21, and IFN ⁇ .
  • the at least one immune stimulating agent comprises an antagonist of a chemokine, such as CXCR2, CXCR4, CCR2, or CCR4.
  • the at least one immune stimulating agent comprises an antibody.
  • the at least one immune stimulating agent may comprise a vaccine, such as a mesothelin-targeting vaccine or attenuated listeria cancer vaccine such as CRS-207.
  • the at least one immune stimulating agent comprises a CD40 agonist, for example, an anti-CD40 antibody.
  • a CD40 agonist for example, an anti-CD40 antibody.
  • Nonlimiting exemplary agonist anti-CD40 antibodies include CP-870,893 (Pfizer and VLST); dacetuzumab (Seattle Genetics); RO7009789 (Roche); ACD-1013 (Alligator Bioscience); SEA-CD40 (Seattle Genetics); and Chi Lob 7/4 (Univ.shire).
  • a CD40 agonist is recombinant CD40L.
  • 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 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”
  • the appropriate formulation and route of administration may be selected according to the intended application.
  • compositions comprising 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 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.
  • 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, 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, 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.
  • 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, 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 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 antibody is administered to a subject one or more times. In various embodiments, an effective dose of an 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. In other embodiments, an effective dose of an 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 antibody is administered once per 1, 2, 3, 4, or 5 weeks. In some embodiments, an effective dose of an antibody is administered twice or three times per week. An effective dose of an antibody is administered to the subject at least once. In some embodiments, the effective dose of an antibody may be administered multiple times, including for periods of at least a month, at least six months, or at least a year.
  • the above therapeutic combinations 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.
  • the cancer has recurred or progressed following a therapy selected from surgery, chemotherapy, and radiation therapy, or a combination thereof.
  • the combinations may be administered in conjunction with one or more additional anti-cancer agents, such as a chemotherapeutic agent, growth inhibitory agent, anti-cancer vaccine such as a gene therapy vaccine, anti-angiogenesis agent and/or anti-neoplastic composition.
  • additional anti-cancer agents such as a chemotherapeutic agent, growth inhibitory agent, anti-cancer vaccine, anti-angiogenesis agent and anti-neoplastic composition.
  • chemotherapeutic agent, growth inhibitory agent, anti-cancer vaccine, anti-angiogenesis agent and anti-neoplastic composition that can be used in combination with the antibodies of the present invention are provided herein under “Definitions.”
  • an anti-inflammatory drug may be administered with the combination, such as a steroid or a non-steroidal anti-inflammatory drug (NSAID).
  • NSAID non-steroidal anti-inflammatory drug
  • 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 cynomolgus CSF1R and to inhibit CSF1-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.
  • mice 6-8 week old female C57BL/6 mice are housed 5 animals per cage with access to food and water ad libitum. Mice are acclimated for at least 3 days after arrival in the vivarium. Mice are weighed and their flanks shaved prior to tumor cell line inoculation.
  • Murine colon adenocarcinoma cell the MC38 is cultured in RPMI+10% FBS+2 mM L-glutamine + antibiotic/antimycotic at 37° C. with 5% CO 2 .
  • Cells are suspended in a solution of 50%/v of DPBS and 50%/v of Matrigel at a concentration of 5 million cells/ml.
  • 100 ⁇ l of cell solution (0.5 million cells) are implanted on the right flank of the each mouse using a 27G1/2 needle. Cells are kept from settling to the bottom of the tube by using an 18G needle and syringe and by slight vortex. Mice are anesthetized using isofluorane to reduce stress and to allow for more precise tumor cell implantation.
  • mice were separated into the following groups and administered chimeric rat anti-mouse CSF1R antibody (mouse IgG1; referred to as “cmFPA008”) and/or anti-CD40 antibody FGK45 (Bio X Cell; see Rolink et al., 1996, Immunity 5:319-330) according to the dosing schedules described below.
  • cmFPA008 chimeric rat anti-mouse CSF1R antibody
  • FGK45 Bio X Cell; see Rolink et al., 1996, Immunity 5:319-330
  • Plasma is collected for pharmacokinetic (PK) analysis.
  • PK pharmacokinetic
  • whole blood is collected via intra-cardiac bleeds, and plasma is isolated for PK analysis (bioanalytical group).
  • PK analysis bioanalytical group
  • At least five (5) tumors from each group are collected for the following analyses.
  • Single-cell isolates of the tumors are generated by collagenase treatment, and FACS is used to examine the infiltration of immune cells into the tumor(s).
  • Tumor sections are also snap frozen in liquid nitrogen and stored at ⁇ 80° C. for protein and mRNA extraction.
  • Tumor sections are embedded in Optimum Cutting Temperature compound (OCT) and stored in ⁇ 80° C. Tumor sections are also placed in 10% buffered formalin overnight, and then transferred to 70% ethanol the following day.
  • OCT Optimum Cutting Temperature compound
  • FIGS. 3 and 4 The results of this experiment are shown in FIGS. 3 and 4 .
  • the combination of anti-CSF1R antibody and anti-CD40 antibody (high) resulted in early tumor regression followed by tumor stasis.
  • the combination of anti-CSF1R antibody and anti-CD40 antibody (low) also demonstrated greater efficacy than either treatment alone.
  • FIG. 4 shows individual tumor volumes at (A) day 11 and (B) day 13.
  • Table 6 shows the tumor volume statistics at day 11 and day 13 using one-way ANOVA.
  • the combination of anti-CSF1R antibody and anti-CD40 antibody (high) showed significantly better efficacy than either treatment alone.
  • FIG. 5 shows body weight for all animals in the study, measured at least twice per week. No significant different in weight was observed for any group relative to control.
  • 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)
  • Immunology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Physics & Mathematics (AREA)
  • Optics & Photonics (AREA)
  • Cell Biology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US15/564,866 2015-04-13 2016-04-12 Combination therapy for cancer Abandoned US20180085472A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/564,866 US20180085472A1 (en) 2015-04-13 2016-04-12 Combination therapy for cancer

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201562146766P 2015-04-13 2015-04-13
US201562190945P 2015-07-10 2015-07-10
US15/564,866 US20180085472A1 (en) 2015-04-13 2016-04-12 Combination therapy for cancer
PCT/US2016/027038 WO2016168149A1 (en) 2015-04-13 2016-04-12 Combination therapy for cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/027038 A-371-Of-International WO2016168149A1 (en) 2015-04-13 2016-04-12 Combination therapy for cancer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/786,158 Continuation US11559583B2 (en) 2015-04-13 2020-02-10 Anti-CSF1R antibody and agonistic anti-CD40 antibody combination therapy for cancer

Publications (1)

Publication Number Publication Date
US20180085472A1 true US20180085472A1 (en) 2018-03-29

Family

ID=55809223

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/564,866 Abandoned US20180085472A1 (en) 2015-04-13 2016-04-12 Combination therapy for cancer
US16/786,158 Active US11559583B2 (en) 2015-04-13 2020-02-10 Anti-CSF1R antibody and agonistic anti-CD40 antibody combination therapy for cancer

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/786,158 Active US11559583B2 (en) 2015-04-13 2020-02-10 Anti-CSF1R antibody and agonistic anti-CD40 antibody combination therapy for cancer

Country Status (21)

Country Link
US (2) US20180085472A1 (pt)
EP (2) EP3283527B1 (pt)
JP (1) JP6971850B2 (pt)
KR (1) KR20170135924A (pt)
CN (2) CN107709365A (pt)
AU (1) AU2016249981B2 (pt)
BR (1) BR112017020952A2 (pt)
CA (1) CA2980460A1 (pt)
CY (1) CY1123972T1 (pt)
DK (1) DK3283527T3 (pt)
ES (1) ES2857076T3 (pt)
HU (1) HUE053903T2 (pt)
IL (1) IL254705B (pt)
LT (1) LT3283527T (pt)
MX (1) MX2017013178A (pt)
PL (1) PL3283527T3 (pt)
PT (1) PT3283527T (pt)
RS (1) RS61531B1 (pt)
SI (1) SI3283527T1 (pt)
TW (1) TWI725966B (pt)
WO (1) WO2016168149A1 (pt)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10293058B2 (en) 2015-04-22 2019-05-21 Curevac Ag RNA containing composition for treatment of tumor diseases
WO2020178313A1 (en) * 2019-03-05 2020-09-10 INSERM (Institut National de la Santé et de la Recherche Médicale) New biomarkers and biotargets in renal cell carcinoma
US10857230B2 (en) 2017-03-03 2020-12-08 Janssen Biotech, Inc. Co-therapy comprising a small molecule CSF-1R inhibitor and an agonistic antibody that specifically binds CD40 for the treatment of cancer
US11661634B2 (en) 2015-05-08 2023-05-30 CureVac Manufacturing GmbH Method for producing RNA

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6621252B2 (ja) * 2015-06-17 2019-12-18 国立大学法人北海道大学 治療耐性がんに対する治療耐性低減剤
CN109475603B (zh) 2016-06-20 2023-06-13 科马布有限公司 抗pd-l1抗体
WO2018144334A1 (en) * 2017-02-02 2018-08-09 Imclone Llc Dosing regimen for anti-csf-1r antibody
WO2018157164A1 (en) * 2017-02-27 2018-08-30 Shattuck Labs, Inc. Csf1r-based chimeric proteins
WO2018183366A1 (en) * 2017-03-28 2018-10-04 Syndax Pharmaceuticals, Inc. Combination therapies of csf-1r or csf-1 antibodies and a t-cell engaging therapy
US20200031944A1 (en) * 2017-03-31 2020-01-30 Five Prime Therapeutics, Inc. Combination therapy for cancer using anti-gitr antibodies
AU2018256406A1 (en) * 2017-04-19 2019-10-17 Marengo Therapeutics, Inc. Multispecific molecules and uses thereof
EP3632932A4 (en) * 2017-06-01 2021-08-18 Jiangsu Hengrui Medicine Co. Ltd. ANTI-CD40 ANTIBODIES, ANTIGEN BINDING FRAGMENT OF THIS ANTIGEN AND ASSOCIATED MEDICAL USE
WO2019035938A1 (en) 2017-08-16 2019-02-21 Elstar Therapeutics, Inc. MULTISPECIFIC MOLECULES BINDING TO BCMA AND USES THEREOF
US11845797B2 (en) 2018-07-03 2023-12-19 Marengo Therapeutics, Inc. Anti-TCR antibody molecules and uses thereof
EP3833762A4 (en) 2018-08-09 2022-09-28 Verseau Therapeutics, Inc. OLIGONUCLEOTIDE COMPOSITIONS FOR TARGETING CCR2 AND CSF1R AND THEIR USES
AU2019346645A1 (en) * 2018-09-27 2021-04-29 Marengo Therapeutics, Inc. CSF1R/CCR2 multispecific antibodies
CN109364252B (zh) * 2018-11-21 2021-09-28 南京大学 抑制ifn-i至arg1诱导通路在制备抗肿瘤药物组合物中的应用
WO2020108611A1 (zh) * 2018-11-30 2020-06-04 江苏恒瑞医药股份有限公司 抗cd40抗体、其抗原结合片段及其医药用途
US20220324988A1 (en) * 2019-04-10 2022-10-13 Nankai University Anti-CD40 antibodies and uses thereof
CN110904229B (zh) * 2019-11-25 2020-09-29 济南市中心医院 Ilt4在肿瘤诊治中的应用
WO2021142573A1 (zh) * 2020-01-13 2021-07-22 丁邦 一种融合蛋白及其制法和用途
WO2022117870A1 (en) * 2020-12-04 2022-06-09 Universiteit Antwerpen Combination immunotherapy of il-15 and cd40 agonist in cancer treatment
CN112979749A (zh) * 2021-02-26 2021-06-18 深圳海创生物科技有限公司 一种活性环肽、活性环肽组合物及其在制备具有抗氧化作用或抗衰老作用的产品中的应用
GB2623199A (en) 2021-04-08 2024-04-10 Marengo Therapeutics Inc Multifunctional molecules binding to TCR and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013132044A1 (en) * 2012-03-08 2013-09-12 F. Hoffmann-La Roche Ag Combination therapy of antibodies against human csf-1r and uses thereof
WO2014036357A1 (en) * 2012-08-31 2014-03-06 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)

Family Cites Families (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
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
US6713610B1 (en) 1990-01-12 2004-03-30 Raju Kucherlapati Human antibodies derived from immunized xenomice
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
US6300129B1 (en) 1990-08-29 2001-10-09 Genpharm International Transgenic non-human animals for producing heterologous antibodies
US5874299A (en) 1990-08-29 1999-02-23 Genpharm International, Inc. Transgenic non-human animals capable of 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
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
CA2137793C (en) 1992-06-09 2003-04-22 Jayvardhan Pandit Crystallization of m-csf
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
WO2000025803A1 (fr) 1998-10-30 2000-05-11 Takeda Chemical Industries, Ltd. Preparations contenant une proteine betacelluline
US6946129B1 (en) 1999-06-08 2005-09-20 Seattle Genetics, Inc. Recombinant anti-CD40 antibody and uses thereof
WO2001030381A2 (de) 1999-10-28 2001-05-03 Hofbauer, Reinhold Verwendung von csf-1-inhibitoren
WO2001034177A2 (en) 1999-11-08 2001-05-17 The Government Of The United States Of America, A S Represented By The Secretary, Department Of Hea Lth & Human Services, The National Institutes Of Health 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
WO2002102972A2 (en) 2001-06-20 2002-12-27 Prochon Biotech Ltd. Antibodies that block receptor protein tyrosine kinase activation, methods of screening for and uses thereof
AR039067A1 (es) 2001-11-09 2005-02-09 Pfizer Prod Inc Anticuerpos para cd40
ATE466885T1 (de) 2002-11-15 2010-05-15 Novartis Vaccines & Diagnostic Methoden zur verhinderung und behandlung von krebs-metastasierung und mit krebs-metastasierung einhergehendem knochenverlust
US7807389B2 (en) 2003-03-14 2010-10-05 University Of Rochester Methods and compositions related to joint inflammation diseases
DK1638941T3 (da) 2003-05-22 2010-10-11 Abbott Lab Indazol-, benzisoxazol- og benzisothiazolkinaseinhibitorer
SI1678314T1 (sl) 2003-10-22 2013-01-31 Keck Graduate Institute Metode za sintetiziranje heteromultimernih polipeptidov pri kvasovkah s strategijo razmnoĺ˝evanja haploidnih celic
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
JP2008506366A (ja) 2004-05-14 2008-03-06 レセプター バイオロジックス インコーポレイテッド 細胞表面受容体アイソフォームならびにその同定および使用方法
US20070264277A1 (en) 2004-07-22 2007-11-15 Dirk Behrens Compositions and Methods of Use for Mgd-Csf in Disease Treatment
TWI309240B (en) 2004-09-17 2009-05-01 Hoffmann La Roche Anti-ox40l antibodies
WO2006076288A2 (en) 2005-01-11 2006-07-20 Five Prime Therapeutics, Inc. Dna constructs for long-term expression of intravascularly injected naked dna
EP1841874B1 (en) 2005-01-27 2010-01-06 Five Prime Therapeutics, Inc. Leader sequences for directing secretion of polypeptides and methods for production thereof
ES2657443T3 (es) 2005-03-25 2018-03-05 Gitr, Inc. Anticuerpos anti-GITR y usos de los mismos
US8003108B2 (en) 2005-05-03 2011-08-23 Amgen Inc. Sclerostin epitopes
SI2439273T1 (sl) 2005-05-09 2019-05-31 Ono Pharmaceutical Co., Ltd. Človeška monoklonska protitelesa za programirano smrt 1 (PD-1) in postopki za zdravljenje raka z uporabo protiteles proti PD-1 samostojno ali v kombinaciji z ostalimi imunoterapevtiki
SI1879573T1 (sl) 2005-05-10 2013-04-30 Incyte Corporation Experimental Station Modulatorji indolamin 2,3-dioksigenaze in postopki za uporabo le-te
EP1885399B1 (en) 2005-05-26 2010-10-20 Seattle Genetics, Inc. Humanized anti-cd40 antibodies and their methods of use
AU2006320858A1 (en) * 2005-11-10 2007-06-07 Receptor Biologix, Inc. Methods for production of receptor and ligand isoforms
ES2540561T3 (es) 2005-12-20 2015-07-10 Incyte Corporation N-hidroxiamidinoheterociclos como moduladores de indolamina 2,3-dioxigenasa
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
US20090074711A1 (en) 2006-09-07 2009-03-19 University Of Southhampton Human therapies using chimeric agonistic anti-human cd40 antibody
JP5319532B2 (ja) 2006-09-19 2013-10-16 インサイト・コーポレイション インドールアミン2,3−ジオキシゲナーゼのモジュレーターとしてのn−ヒドロキシアミジノヘテロサイクル
CL2007002650A1 (es) 2006-09-19 2008-02-08 Incyte Corp Compuestos derivados de heterociclo n-hidroxiamino; composicion farmaceutica, util para tratar cancer, infecciones virales y desordenes neurodegenerativos entre otras.
EA016527B1 (ru) * 2006-10-23 2012-05-30 ЭсДжиЭкс ФАРМАСЬЮТИКАЛЗ, ИНК. Триазолопиридазиновые модуляторы протеинкиназ
US20100266528A1 (en) 2006-11-17 2010-10-21 Biogen Idec Ma Inc. Systemic Administration of Colony Stimulating Factors to Treat Amyloid Associated Disorders
ATE494306T1 (de) 2007-02-14 2011-01-15 Vaccinex Inc Humanisierte anti-cd100-antikörper
WO2008124858A2 (en) 2007-04-11 2008-10-23 F-Star Biotechnologische Forschungs- Und Entwicklungsges. M.B.H. Targeted receptor
EP1987839A1 (en) 2007-04-30 2008-11-05 I.N.S.E.R.M. Institut National de la Sante et de la Recherche Medicale Cytotoxic anti-LAG-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
WO2008150383A1 (en) 2007-05-30 2008-12-11 Albert Einstein College Of Medicine Of Yeshiva University Csf-1r mutants
EP3124046B1 (en) 2007-07-12 2019-12-25 GITR, Inc. Combination therapies employing gitr binding molecules
WO2009026303A1 (en) 2007-08-21 2009-02-26 Amgen Inc. Human c-fms antigen binding proteins
US7981415B2 (en) 2007-09-07 2011-07-19 Cisthera, Inc. Humanized PAI-1 antibodies
EP2044949A1 (en) 2007-10-05 2009-04-08 Immutep Use of recombinant lag-3 or the derivatives thereof for eliciting monocyte immune response
JP2011502266A (ja) 2007-10-31 2011-01-20 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ Fms治療に対する応答を評価するバイオマーカー
CN101932325B (zh) 2007-11-30 2014-05-28 新联基因公司 Ido抑制剂
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
CA2711569C (en) 2008-01-03 2017-08-01 Universite De La Mediterannee, Aix-Marseille Ii Composition and processes for use in an anti-hiv treatment
BRPI0909044B8 (pt) 2008-03-14 2021-05-25 Transgene Sa anticorpo que se liga especificamente ao csf-1r humano,composição farmacêutica e uso dos mesmos
US8470977B2 (en) 2008-03-14 2013-06-25 Transgene S.A. Antibody against the CSF-1R
WO2009120905A2 (en) * 2008-03-26 2009-10-01 Cellerant Therapeutics, Inc. Immunoglobulin and/or toll-like receptor proteins associated with myelogenous haematological proliferative disorders and uses thereof
MX2010012064A (es) 2008-05-05 2010-12-06 Schering Corp Uso secuencial de agentes quimioterapeuticos citotoxicos para el tratamiento de cancer.
JP5945096B2 (ja) 2008-07-04 2016-07-05 小野薬品工業株式会社 抗ヒトpd−1抗体の癌に対する治療効果を最適化するための判定マーカーの使用
AR072999A1 (es) 2008-08-11 2010-10-06 Medarex Inc Anticuerpos humanos que se unen al gen 3 de activacion linfocitaria (lag-3) y los usos de estos
US8080246B2 (en) 2008-11-26 2011-12-20 Five Prime Therapeutics, Inc. Colony stimulating factor 1 receptor (CSF1R) extracellular domain fusion molecules
US8183207B2 (en) 2008-11-26 2012-05-22 Five Prime Therapeutics, Inc. Treatment of osteolytic disorders and cancer using CSF1R extracellular domain fusion molecules
EP2473531A4 (en) 2009-09-03 2013-05-01 Merck Sharp & Dohme ANTI-GITRANT ANTIBODIES
BR112012013717B1 (pt) * 2009-12-10 2020-01-28 Hoffmann La Roche anticorpos de ligação ao csf-1r humano, composição farmacêutica e usos do anticorpo
LT2542256T (lt) 2010-03-04 2019-10-25 Macrogenics Inc Su b7-h3 reaguojantys antikūnai, jų imunologiškai aktyvūs fragmentai ir jų naudojimas
CN102918060B (zh) 2010-03-05 2016-04-06 霍夫曼-拉罗奇有限公司 抗人csf-1r抗体及其用途
CA2789071C (en) 2010-03-05 2018-03-27 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
LT2566517T (lt) 2010-05-04 2019-01-10 Five Prime Therapeutics, Inc. Antikūnai, surišantys csf1r
RU2551963C2 (ru) 2010-09-09 2015-06-10 Пфайзер Инк. Молекулы, связывающиеся с 4-1вв
CN103261217B (zh) 2010-11-11 2017-04-26 港大科桥有限公司 可溶性 pd‑1变体、融合构建体及其用途
AR084234A1 (es) 2010-12-13 2013-05-02 Novartis Ag Metodos predictivos y metodos para tratar artritis usando antagonistas de interleuquina 17 (il-17)
NO2694640T3 (pt) 2011-04-15 2018-03-17
GB201115280D0 (en) 2011-09-05 2011-10-19 Alligator Bioscience Ab Antibodies, uses and methods
SG11201401639QA (en) 2011-10-21 2014-05-29 Transgene Sa Modulation of macrophage activation
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)
EP2847220A1 (en) 2012-05-11 2015-03-18 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
JP6448533B2 (ja) 2012-05-15 2019-01-09 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Pd−1/pd−l1シグナル伝達を破壊することによる癌免疫療法
AR091649A1 (es) 2012-07-02 2015-02-18 Bristol Myers Squibb Co Optimizacion de anticuerpos que se fijan al gen de activacion de linfocitos 3 (lag-3) y sus usos
CN105530916B (zh) 2013-07-11 2020-04-07 艾利斯达医药品公司 与间水杨酸形成的烟碱盐
TW201605896A (zh) 2013-08-30 2016-02-16 安美基股份有限公司 Gitr抗原結合蛋白
AR097584A1 (es) 2013-09-12 2016-03-23 Hoffmann La Roche Terapia de combinación de anticuerpos contra el csf-1r humano y anticuerpos contra el pd-l1 humano
AU2015339306B2 (en) 2014-10-29 2021-07-22 Bristol-Myers Squibb Company Combination therapy for cancer

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013132044A1 (en) * 2012-03-08 2013-09-12 F. Hoffmann-La Roche Ag Combination therapy of antibodies against human csf-1r and uses thereof
WO2014036357A1 (en) * 2012-08-31 2014-03-06 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10293058B2 (en) 2015-04-22 2019-05-21 Curevac Ag RNA containing composition for treatment of tumor diseases
US10869935B2 (en) 2015-04-22 2020-12-22 Curevac Ag RNA containing composition for treatment of tumor diseases
US10918740B2 (en) 2015-04-22 2021-02-16 Curevac Ag RNA containing composition for treatment of tumor diseases
US11661634B2 (en) 2015-05-08 2023-05-30 CureVac Manufacturing GmbH Method for producing RNA
US10857230B2 (en) 2017-03-03 2020-12-08 Janssen Biotech, Inc. Co-therapy comprising a small molecule CSF-1R inhibitor and an agonistic antibody that specifically binds CD40 for the treatment of cancer
WO2020178313A1 (en) * 2019-03-05 2020-09-10 INSERM (Institut National de la Santé et de la Recherche Médicale) New biomarkers and biotargets in renal cell carcinoma

Also Published As

Publication number Publication date
ES2857076T3 (es) 2021-09-28
IL254705A0 (en) 2017-11-30
TWI725966B (zh) 2021-05-01
EP3283527A1 (en) 2018-02-21
EP3964527A2 (en) 2022-03-09
US20200237927A1 (en) 2020-07-30
MX2017013178A (es) 2018-03-01
LT3283527T (lt) 2021-03-25
CN107709365A (zh) 2018-02-16
CA2980460A1 (en) 2016-10-20
AU2016249981B2 (en) 2022-02-17
SI3283527T1 (sl) 2021-04-30
HUE053903T2 (hu) 2021-07-28
BR112017020952A2 (pt) 2018-07-10
AU2016249981A1 (en) 2017-10-12
WO2016168149A1 (en) 2016-10-20
EP3964527A3 (en) 2022-06-15
KR20170135924A (ko) 2017-12-08
US11559583B2 (en) 2023-01-24
JP2018512430A (ja) 2018-05-17
DK3283527T3 (da) 2021-03-08
CN114681608A (zh) 2022-07-01
RS61531B1 (sr) 2021-04-29
TW201702270A (zh) 2017-01-16
IL254705B (en) 2021-07-29
EP3283527B1 (en) 2021-01-13
JP6971850B2 (ja) 2021-11-24
PT3283527T (pt) 2021-03-03
CY1123972T1 (el) 2022-05-27
PL3283527T3 (pl) 2021-06-14

Similar Documents

Publication Publication Date Title
US11559583B2 (en) Anti-CSF1R antibody and agonistic anti-CD40 antibody combination therapy for cancer
US20240041979A1 (en) Cd80 extracellular domain polypeptides and their use in cancer treatment
US11789010B2 (en) Methods of treatment with CD80 extracellular domain polypeptides
US20230416382A1 (en) Anti-c-c chemokine receptor 8 (ccr8) antibodies and methods of use thereof
US20230045791A1 (en) Anti-tnfr2 antibody and uses thereof
WO2023281313A1 (en) Anti-tnfr2 antibody and uses thereof
US20200031944A1 (en) Combination therapy for cancer using anti-gitr antibodies
EA039894B1 (ru) Способ лечения рака, композиция для лечения рака, применение композиции для получения лекарственного средства для лечения рака, характеризующегося присутствием макрофагов, экспрессирующих csf1r

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

STCB Information on status: application discontinuation

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