US20170306038A1 - Compositions and methods of use for augmented immune response and cancer therapy - Google Patents

Compositions and methods of use for augmented immune response and cancer therapy Download PDF

Info

Publication number
US20170306038A1
US20170306038A1 US15/517,872 US201515517872A US2017306038A1 US 20170306038 A1 US20170306038 A1 US 20170306038A1 US 201515517872 A US201515517872 A US 201515517872A US 2017306038 A1 US2017306038 A1 US 2017306038A1
Authority
US
United States
Prior art keywords
seq
antibody
inhibitor
amino acid
cancer
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
US15/517,872
Other languages
English (en)
Inventor
Jennifer Brogdon
Daniela CIPOLLETTA
Glenn Dranoff
Deborah A. Knee
Fei Wang
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.)
Novartis AG
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=54347879&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20170306038(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Priority to US15/517,872 priority Critical patent/US20170306038A1/en
Publication of US20170306038A1 publication Critical patent/US20170306038A1/en
Assigned to NOVARTIS AG reassignment NOVARTIS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC.
Assigned to NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC. reassignment NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BROGDON, Jennifer, CIPOLLETTA, Daniela, DRANOFF, GLENN
Assigned to NOVARTIS INSTITUTE FOR FUNCTIONAL GENOMICS, INC., reassignment NOVARTIS INSTITUTE FOR FUNCTIONAL GENOMICS, INC., ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KNEE, DEBORAH A., WANG, FEI
Assigned to NOVARTIS AG reassignment NOVARTIS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS INSTITUTE FOR FUNCTIONAL GENOMICS, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [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
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • 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
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/51Complete heavy chain or Fd fragment, i.e. VH + CH1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/515Complete light chain, i.e. VL + CL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/74Inducing cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • the present invention relates to antibodies, antibody fragments, and antigen binding molecules that bind to tumor necrosis factor receptor superfamily member 18/glucocorticoid induced TNFR-related protein (“GITR”), and more specifically that are agonists, stimulate signaling through the receptor and/or modulate immune response.
  • GITR tumor necrosis factor receptor superfamily member 18/glucocorticoid induced TNFR-related protein
  • GITR Glucocorticoid-induced TNFR-related protein
  • TNFRSF Tumor Necrosis Factor Superfamily
  • ECD extracellular domain of GITR consists of 3 cysteine-rich domains (CRDs), followed by a transmembrane domain (TM) and an intracellular domain (ICD).
  • GITR expression is detected constitutively on murine and human CD4+CD25+ regulatory T cells which can be further increased upon activation.
  • effector CD4+CD25 ⁇ T cells and CD8+CD25 ⁇ T cells express low to undetectable levels of GITR, which is rapidly upregulated following T cell receptor activation.
  • Expression of GITR has also been detected on activated NK cells, dendritic cells, and macrophages.
  • Signal transduction pathway downstream of GITR has been shown to involve MAPK and the canonical NF ⁇ B pathways.
  • TRAF family members have been implicated as signaling intermediates downstream of GITR (Nocentini et al. (2005) Eur. J. Immunol., 35:1016-1022).
  • an anti-mGITR which has functional Fc effector activity has been shown in some preclinical models to deplete regulatory T cells, as well as enhance T effector cell proliferation and cytokine secretion in select tumor environment.
  • GITR glucocorticoid-induced tumor necrosis factor receptor superfamily member 18
  • the invention provides isolated antibodies, antibody fragments, and antigen binding molecules that specifically bind to human GITR, wherein the antibody, antibody fragment, or the antigen binding molecule binds to an epitope comprising the cysteine-rich domain 1 (“CRD1”, SEQ ID NO:4: CGPGRLLLGTGTDARCCRVHTTRCCRDYPGEECCSEWDC) and the cysteine-rich domain 2 (“CRD2”, SEQ ID NO:5: MCVQPEFHCGDPCCTTCRHHPCPPGQGVQSQGKFSFGFQC), and wherein the antibody, antibody fragment, or the antigen binding molecule is an agonist of GITR, and wherein the antibody, antibody fragment, or the antigen binding molecule optionally has an intact or increased FcR effector function.
  • CCD1 cysteine-rich domain 1
  • SEQ ID NO:4 CGPGRLLLGTGTDARCCRVHTTRCCRDYPGEECCSEWDC
  • CCD2 cysteine-rich domain 2
  • the antibody, antibody fragment, or the antigen binding molecule binds to an epitope comprising SEQ ID NO:88) of human GITR. In some embodiments, the antibody, antibody fragment, or antigen binding molecule competes with an antibody or antibody fragment that binds to an epitope comprising SEQ ID NO:88 of human GITR. In some embodiments, the antibody, antibody fragment, or antigen binding molecule binds to at least one amino acid residue within SEQ ID NO:88 of human GITR, for example, the antibody, antibody fragment, or antigen binding molecule binds to an epitope that overlaps with SEQ ID NO:88 of human GITR.
  • the antibody, antibody fragment, or the antigen binding molecule binds to an epitope comprising CRD1 (residues 34-72, SEQ ID NO:4) and residue 78 of human GITR. In some embodiments, the antibody, antibody fragment, or antigen binding molecule competes with an antibody or antibody fragment that binds to an epitope within CRD1 (residues 34-72, SEQ ID NO:4) and residue 78 of human GITR.
  • the antibody, antibody fragment, or antigen binding molecule binds to at least one amino acid residue within CRD1 (residues 34-72, SEQ ID NO:4) and residue 78 of human GITR, for example, the antibody, antibody fragment, or antigen binding molecule binds to an epitope that overlaps with CRD1 (residues 34-72, SEQ ID NO:4) and residue 78 of human GITR.
  • the antibody, antibody fragment, or antigen binding molecule binds to SEQ ID NO:1 and comprises (a) a heavy chain variable region comprising a human heavy chain wherein: i) the heavy chain CDR1 comprises SEQ ID NO:22, and ii) the heavy chain CDR2 comprises a sequence selected from any one of SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, and SEQ ID NO:27, and iii) the heavy chain CDR3 comprises SEQ ID NO:29 or SEQ ID NO:109; and (b) a light chain variable region, wherein i) the light chain CDR1 comprises SEQ ID NO:30 or SEQ ID NO:31, and ii) the light chain CDR2 comprises SEQ ID NO:33, and iii) the light chain CDR3 comprises SEQ ID NO:34.
  • the antibody, antibody fragment, or antigen binding molecule binds to SEQ ID NO:88 and comprises (a) a heavy chain variable region comprising a human heavy chain wherein: i) the heavy chain CDR1 comprises SEQ ID NO:22, and ii) the heavy chain CDR2 comprises a sequence selected from any one of SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, and SEQ ID NO:27, and iii) the heavy chain CDR3 comprises SEQ ID NO:29 or SEQ ID NO:109; and (b) a light chain variable region, wherein i) the light chain CDR1 comprises SEQ ID NO:30 or SEQ ID NO:31, and ii) the light chain CDR2 comprises SEQ ID NO:33, and iii) the light chain CDR3 comprises SEQ ID NO:34.
  • the heavy chain variable region has at least 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to the variable region of SEQ ID NO:16 and the light chain variable region has at least 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to the variable region of SEQ ID NO:17.
  • the antibody, antibody fragment, or antigen binding molecule comprises a heavy chain comprising SEQ ID NO:16 and a light chain comprising SEQ ID NO:17.
  • the antibody, antibody fragment, or antigen binding molecule competes with an antibody that comprises a heavy chain comprising SEQ ID NO:16 and a light chain comprising SEQ ID NO:17.
  • the heavy chain FR4 is a human germline FR4. In particular embodiments, the heavy chain FR4 is SEQ ID NO:42.
  • the light chain FR4 is a human germline FR4. In particular embodiments, the light chain FR4 is SEQ ID NO:50.
  • an antibody, antibody fragment or antigen binding molecule wherein: i) the heavy chain CDR1 comprises SEQ ID NO:22 or SEQ ID NO:84; ii) the heavy chain CDR2 comprises SEQ ID NO:28 or SEQ ID NO:80; iii) the heavy chain CDR3 comprises SEQ ID NO:29 or SEQ ID NO:109; iv) the light chain CDR1 comprises SEQ ID NO:30 or SEQ ID NO:85; v) the light chain CDR2 comprises SEQ ID NO:33 or SEQ ID NO:82, and vi) the light chain CDR3 comprises SEQ ID NO:34 or SEQ ID NO:83.
  • an antibody, antibody fragment or antigen binding molecule wherein: wherein: i) the heavy chain CDR1 comprises SEQ ID NO:22; ii) the heavy chain CDR2 comprises SEQ ID NO:23; iii) the heavy chain CDR3 comprises SEQ ID NO:29; iv) the light chain CDR1 comprises SEQ ID NO:30; v) the light chain CDR2 comprises SEQ ID NO:33, and vi) the light chain CDR3 comprises SEQ ID NO:34.
  • an antibody, antibody fragment or antigen binding molecule wherein: wherein: i) the heavy chain CDR1 comprises SEQ ID NO:22; ii) the heavy chain CDR2 comprises SEQ ID NO:24; iii) the heavy chain CDR3 comprises SEQ ID NO:29; iv) the light chain CDR1 comprises SEQ ID NO:31; v) the light chain CDR2 comprises SEQ ID NO:33, and vi) the light chain CDR3 comprises SEQ ID NO:34.
  • an antibody, antibody fragment or antigen binding molecule wherein: wherein: i) the heavy chain CDR1 comprises SEQ ID NO:22; ii) the heavy chain CDR2 comprises SEQ ID NO:25; iii) the heavy chain CDR3 comprises SEQ ID NO:29; iv) the light chain CDR1 comprises SEQ ID NO:30; v) the light chain CDR2 comprises SEQ ID NO:33, and vi) the light chain CDR3 comprises SEQ ID NO:34.
  • an antibody, antibody fragment or antigen binding molecule wherein: wherein: i) the heavy chain CDR1 comprises SEQ ID NO:22; ii) the heavy chain CDR2 comprises SEQ ID NO:26; iii) the heavy chain CDR3 comprises SEQ ID NO:29; iv) the light chain CDR1 comprises SEQ ID NO:30; v) the light chain CDR2 comprises SEQ ID NO:33, and vi) the light chain CDR3 comprises SEQ ID NO:34.
  • an antibody, antibody fragment or antigen binding molecule wherein: wherein: i) the heavy chain CDR1 comprises SEQ ID NO:22; ii) the heavy chain CDR2 comprises SEQ ID NO:27; iii) the heavy chain CDR3 comprises SEQ ID NO:29; iv) the light chain CDR1 comprises SEQ ID NO:30; v) the light chain CDR2 comprises SEQ ID NO:33, and vi) the light chain CDR3 comprises SEQ ID NO:34.
  • an antibody, antibody fragment or antigen binding molecule wherein: wherein: i) the heavy chain CDR1 comprises SEQ ID NO:22; ii) the heavy chain CDR2 comprises SEQ ID NO:25; iii) the heavy chain CDR3 comprises SEQ ID NO:109; iv) the light chain CDR1 comprises SEQ ID NO:30; v) the light chain CDR2 comprises SEQ ID NO:33, and vi) the light chain CDR3 comprises SEQ ID NO:34.
  • the invention provides antibodies, antibody fragments, or antigen binding molecules that specifically bind GITR, wherein the antibody or antibody fragment comprises a heavy chain variable region and a light chain variable region wherein: i) the CDR1 of the heavy chain comprises an amino acid sequence selected from the group consisting of SEQ ID NO:22, SEQ ID NO: 79, or SEQ ID NO:84; ii) the CDR2 of the heavy chain comprises an amino acid sequence selected from the group consisting of SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:62, and SEQ ID NO:80; iii) the CDR3 of the heavy chain comprises SEQ ID NO:29 or SEQ ID NO:109; iv) the CDR1 of the light chain comprises an amino acid sequence selected from the group consisting of SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:63, SEQ ID NO:81, SEQ ID
  • the heavy chain variable region has at least 90%, 93%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to the variable region of a sequence selected from the group consisting of SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:99 and SEQ ID NO:105
  • the light chain variable region has at least 90%, 93%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to the variable region of a sequence selected from the group consisting of SEQ ID NO:9 and SEQ ID NO:7.
  • the isolated antibody, antibody fragment, or antigen binding molecule comprises a heavy chain variable domain comprising a sequence selected from any of SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:99 and SEQ ID NO:105; and light chain variable domain comprising SEQ ID NO:7 or SEQ ID NO:9.
  • the isolated antibody, antibody fragment, or antigen binding molecule comprises a heavy chain variable domain of SEQ ID NO:6 and a light chain variable domain of SEQ ID NO:7.
  • the isolated antibody or antibody fragment comprises a heavy chain variable domain comprising SEQ ID NO:8 and a light chain variable domain comprising SEQ ID NO:9.
  • the isolated antibody or antibody fragment comprises a heavy chain variable domain comprising SEQ ID NO:10 and a light chain variable domain comprising SEQ ID NO:7. In other embodiments, the isolated antibody or antibody fragment comprises a heavy chain variable domain comprising SEQ ID NO:12 and a light chain variable domain comprising SEQ ID NO:7. In other embodiments, the isolated antibody or antibody fragment comprises a heavy chain variable domain comprising SEQ ID NO:14 and a light chain variable domain comprising SEQ ID NO:7.
  • the heavy chain variable region has at least 90%, 93%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to the variable region of SEQ ID NO:99 and the light chain variable region has at least 90%, 93%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to the variable region of SEQ ID NO:7.
  • the isolated antibody or antibody fragment comprises a heavy chain variable domain comprising the SEQ ID NO:99 and a light chain variable domain comprising SEQ ID NO:7.
  • the heavy chain variable region has at least 90%, 93%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to the variable region of SEQ ID NO:105 and the light chain variable region has at least 90%, 93%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to the variable region of SEQ ID NO:7.
  • the isolated antibody or antibody fragment comprises a heavy chain variable domain comprising the SEQ ID NO:105 and a light chain variable domain comprising SEQ ID NO:7.
  • the antibody, antibody fragment, or antigen binding molecule that binds to GITR is humanized. In certain embodiments, the antibody or antibody fragment comprises a human constant region.
  • the antibody fragment is a Fab′ fragment. In some embodiments, the antibody fragment is a single chain antibody (scFv). In some embodiments, the antibody fragment is a single-domain antibody or nanobody.
  • the antibody or antibody fragment is cross-linked to a second antibody or antibody fragment. In some embodiments, the antibody is glycosylated.
  • the antibody, antibody fragment, or antigen binding molecule is an IgG. In certain embodiments the antibody, antibody fragment, or antigen binding molecule comprises an IgG isotype antibody Fc region. In particular embodiments the antibody, antibody fragment, or antigen binding molecule comprises an IgG1 or an IgG2 isotype antibody Fc region. In certain embodiments the antibody, antibody fragment, or antigen binding molecule is an IgG1 or an IgG2 antibody. In some embodiments, the antibody, antibody fragment, or antigen binding molecule contains at least one mutation that modulates (i.e., increases or decreases) binding of the antibody or antibody fragment to an Fc receptor.
  • the antibody, antibody fragment, or antigen binding molecule contains at least one mutation that modulates (i.e., increases or decreases) the antibody, antibody fragment, or antigen binding molecule to activate an Fc receptor.
  • the antibody, antibody fragment, or antigen binding molecule contains at least one mutation that increases binding of the antibody or antibody fragment to an Fc receptor.
  • the antibody, antibody fragment, or antigen binding molecule contains at least one mutation that increases the antibody, antibody fragment, or antigen binding molecule to activate an Fc receptor.
  • the antibody, antibody fragment, or antigen binding molecule cross-reacts with human and non-human primate GITR. In some embodiments, the antibody, antibody fragment, or antigen binding molecule does not cross-react with rodent GITR, e.g., rat GITR or mouse GITR.
  • the invention further provides polynucleotides encoding an antibody, antibody fragment or antigen binding molecule of the invention as described herein.
  • the polynucleotide encoding the light chain variable region has at least 90%, 93%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity to a nucleic acid sequence selected from SEQ ID NO:52, SEQ ID NO:54 and SEQ ID NO:102.
  • the polynucleotide encoding the heavy chain variable region has at least 90%, 93%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity to a nucleic acid sequence selected from SEQ ID NO:51, SEQ ID NO:53, SEQ ID NO:55, SEQ ID NO:56, SEQ ID NO:57, SEQ ID NO:101, and SEQ ID NO:107.
  • the polynucleotide encoding the light chain variable region has a nucleic acid sequence selected from SEQ ID NO:52, SEQ ID NO:54, and SEQ ID NO:102.
  • the polynucleotide encoding the heavy chain variable region has a nucleic acid sequence selected from SEQ ID NO:51, SEQ ID NO:53, SEQ ID NO:55, SEQ ID NO:56, SEQ ID NO:57 SEQ ID NO:101 and SEQ ID NO:107.
  • the invention further provides compositions comprising an antibody, antibody fragment, or antigen binding molecule of the invention, as described herein, and a pharmaceutically acceptable carrier.
  • the invention provides pharmaceutical compositions comprising an antibody, antibody fragment, or antigen binding molecule of the invention for administering to an individual.
  • the composition further comprises a target antigen, for example, a cancer-associated antigen or a tumor-associated antigen.
  • a target antigen for example, a cancer-associated antigen or a tumor-associated antigen.
  • the target antigen is a viral antigen, a bacterial antigen, a fungal antigen or a parasitic antigen.
  • kits comprising an antibody or antibody fragment of the invention, as described herein.
  • kits further comprise a second agent for co-administration with the antibody.
  • the second agent is a target antigen, for example, a cancer-associated antigen or a tumor-associated antigen.
  • the target antigen is a viral antigen, a bacterial antigen, a fungal antigen or a parasitic antigen.
  • the second agent is an antagonist of CTLA4. In some embodiments, the second agent is an antagonist of TIM3. In some embodiments, the second agent is an antagonist of LAG3. In some embodiments, the second agent is an inhibitor of PD-1/PD-L1 (e.g., B7-H1 or analogue thereof, PD-1 antibody) interaction. In certain embodiments the second agent is an antagonist of PD-1. In certain embodiments the second agent is an antagonist of PD-L1.
  • PD-1/PD-L1 e.g., B7-H1 or analogue thereof, PD-1 antibody
  • the antibody or antibody fragment and second agent are provided as a mixture.
  • the antibody or antibody fragment and the second agent are provided in separate formulations.
  • the invention provides methods of enhancing a T cell response in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of an anti-GITR agonist antibody or antibody fragment of the invention, as described herein.
  • the invention provides an anti-GITR agonist antibody or antibody fragment of the invention for use in enhancing a T cell response in an individual.
  • the invention provides a composition comprising an antibody or antibody fragment of the invention for use in enhancing a T cell response in an individual.
  • the invention provides methods of treating tumor growth of a cancer that expresses a tumor associated antigen in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of an anti-GITR agonist antibody, antibody fragment, or antigen binding molecule of the invention, as described herein.
  • the invention further provides an anti-GITR agonist antibody or antibody fragment of the invention for use in treating tumor growth of a cancer in an individual.
  • the invention further provides a composition comprising an antibody or antibody fragment of the invention for use in reducing, inhibiting or preventing tumor growth of a cancer that expresses a tumor associated antigen in an individual.
  • the anti-GITR agonist antibody, antibody fragment, or antigen binding molecule is co-administered with an antigen.
  • the antigen is a cancer-associated antigen or a tumor-associated antigen.
  • the anti-GITR agonist antibody or antibody fragment is co-administered with cancer cells from the patient, i.e., autologous cancer cells.
  • the anti-GITR agonist antibody, antibody fragment, or antigen binding molecule is co-administered with an antagonist of CTLA4. In some embodiments, the anti-GITR agonist antibody, antibody fragment, or antigen binding molecule is co-administered with an antagonist of LAGS. In some embodiments, the anti-GITR agonist antibody, antibody fragment, or antigen binding molecule is co-administered with an antagonist of TIM3. In some embodiments, the anti-GITR agonist antibody or antibody fragment is co administered with an inhibitor of PD-1/PD-L1 (e.g., B7-H1) interaction.
  • PD-1/PD-L1 e.g., B7-H1
  • the anti-GITR agonist antibody, antibody fragment, or antigen binding molecule is co-administered with an antagonist of PD-1. In certain embodiments, the anti-GITR agonist antibody, antibody fragment, or antigen binding molecule is co-administered with an antagonist of PD-L1.
  • the anti-GITR agonist antibody, antibody fragment, or antigen binding molecule is co-administered with a chemotherapeutic agent or a cytotoxin.
  • the T cell response is a CD8+ cytotoxic T lymphocyte (CTL) T cell response. In some embodiments, the T cell response is a CD4+ helper T cell (Th) response.
  • CTL cytotoxic T lymphocyte
  • Th CD4+ helper T cell
  • compositions e.g., anti-GITR agonist antibody, antibody fragment, or antigen binding molecules and methods of using them
  • the methods described herein include administering to the subject an anti-GITR agonist antibody, antibody fragment, or antigen binding molecule as described herein (optionally in combination with one or more inhibitors of PD-1, PD-L1, LAG-3, TIM-3, CEACAM (e.g., CEACAM-1, CEACAM-3, and/or CEACAM-5), or CTLA-4)), further include administration of a second therapeutic agent chosen from one or more of the agents listed in Table 6, in an amount effective to treat or prevent a disorder, e.g., a disorder as described herein, e.g., a cancer.
  • a disorder e.g., a disorder as described herein, e.g., a cancer.
  • the—anti-GITR agonist antibody, antibody fragment, or antigen binding molecule, the additional agent (e.g., second or third agent), or all can be administered in an amount or dose that is higher, lower or the same than the amount or dosage of each agent used individually, e.g., as a monotherapy.
  • the administered amount or dosage of the anti-GITR agonist antibody, antibody fragment, or antigen binding molecule, the additional agent (e.g., second or third agent), or all is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50%) than the amount or dosage of each agent used individually, e.g., as a monotherapy.
  • the amount or dosage of the anti-GITR agonist antibody, antibody fragment, or antigen binding molecule, the additional agent (e.g., second or third agent), or all, that results in a desired effect (e.g., treatment of cancer) is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50% lower).
  • the second therapeutic agent is chosen from one or more of the agents listed in Table 6
  • the cancer is chosen from a lung cancer (e.g., a non-small cell lung cancer (NSCLC) (e.g., a NSCLC with squamous and/or non-squamous histology, or a NSCLC adenocarcinoma), or disclosed in a publication listed in Table 6.
  • NSCLC non-small cell lung cancer
  • the second therapeutic agent is chosen from one or more of: 1) a protein kinase C (PKC) inhibitor; 2) a heat shock protein 90 (HSP90) inhibitor; 3) an inhibitor of a phosphoinositide 3-kinase (PI3K) and/or target of rapamycin (mTOR); 4) an inhibitor of cytochrome P450 (e.g., a CYP17 inhibitor or a 17alpha-Hydroxylase/C17-20 Lyase inhibitor); 5) an iron chelating agent; 6) an aromatase inhibitor; 7) an inhibitor of p53, e.g., an inhibitor of a p53/Mdm2 interaction; 8) an apoptosis inducer; 9) an angiogenesis inhibitor; 10) an aldosterone synthase inhibitor; 11) a smoothened (SMO) receptor inhibitor; 12) a prolactin receptor (PRLR) inhibitor; 13) a Wnt signaling inhibitor; 14)
  • PIC protein
  • the invention features a method of treating (e.g., inhibiting, reducing, ameliorating, or preventing) a disorder, e.g., a hyperproliferative condition or disorder (e.g., a cancer) in a subject.
  • a disorder e.g., a hyperproliferative condition or disorder (e.g., a cancer) in a subject.
  • Methods include administering to the subject a combination of two, three or more therapeutic agents chosen from one, two or all of the following categories (i)-(iii): (i) an agent that enhances antigen (e.g., tumor antigen) presentation; (ii) an agent that enhances an effector cell response (e.g., B cell and/or T cell activation and/or mobilization); or (iii) an agent that decreases tumor immunosuppression, thereby treating the disorder, e.g., the hyperproliferative condition or disorder (e.g., the cancer).
  • exemplary therapeutic agents of categories (i)-(iii) are chosen from one or more of the agents listed in Table 7 and further described herein.
  • the combination includes at least a GITR modulator (e.g., an nti-GITR agonist antibody, antibody fragment, or antigen binding molecule as described herein) and one or more additional therapeutic agents of categories (i)-(iii).
  • a GITR modulator e.g., an nti-GITR agonist antibody, antibody fragment, or antigen binding molecule as described herein
  • the cancer treated can be, e.g., a cancer described herein, such as lung cancer (squamous), lung cancer (adenocarcinoma), head and neck cancer, cervical cancer (squamous), stomach cancer, thyroid cancer, melanoma, nasopharyngeal cancer, or breast cancer.
  • the patient has a cancer that expresses a tumor associated antigen.
  • the cancer is selected from the group consisting of melanoma, ovarian cancer, colorectal cancer, prostate, non-small cell lung cancer (NSCLC) and breast cancer.
  • the type of cancer is selected from the group consisting of: pancreatic cancer, melanomas, breast cancer, lung cancer, bronchial cancer, colorectal cancer, prostate cancer, stomach cancer, ovarian cancer, urinary bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, cervical cancer, uterine or endometrial cancer, cancer of the oral cavity or pharynx, head and neck squamous cell carcinoma (HNSCC), liver cancer, kidney cancer, testicular cancer, biliary tract cancer, small bowel or appendix cancer, salivary gland cancer, thyroid gland cancer, adrenal gland cancer, osteosarcoma, chondrosarcoma, and cancer of hematological tissues.
  • pancreatic cancer melanomas
  • breast cancer breast cancer
  • lung cancer bronchial cancer
  • colorectal cancer prostate cancer
  • stomach cancer ovarian cancer
  • urinary bladder cancer brain or central nervous system cancer
  • peripheral nervous system cancer esophageal cancer
  • cervical cancer uterine
  • the patient has an infectious disease, for example, a viral infection, a bacterial infection, a fungal antigen or a parasitic antigen.
  • the anti-GITR agonist antibody is co-administered with a viral antigen (e.g., from HCV, HSV or HIV).
  • the anti-GITR agonist antibody is co-administered with a bacterial antigen.
  • the anti-GITR agonist antibody is co-administered with a fungal antigen.
  • the anti-GITR agonist antibody is co-administered with a parasitic antigen (e.g., filariasis).
  • an isolated antibody, antibody fragment, or antigen binding molecule for use in for use in therapy.
  • the antibody, antibody fragment or antigen binding molecule are provided for use enhancing a T cell response in an individual in need thereof.
  • the antibody, antibody fragment or antigen binding molecule are provided for use in the treatment of tumor growth in an individual in need thereof.
  • combinations disclosed herein can result in one or more of: an increase in antigen presentation, an increase in effector cell function (e.g., one or more of T cell proliferation, IFN- ⁇ secretion or cytolytic function), inhibition of regulatory T cell function, an effect on the activity of multiple cell types, such as regulatory T cell, effector T cells and NK cells), an increase in tumor infiltrating lymphocytes, an increase in T-cell receptor mediated proliferation, and a decrease in immune evasion by cancerous cells.
  • an increase in antigen presentation an increase in effector cell function (e.g., one or more of T cell proliferation, IFN- ⁇ secretion or cytolytic function), inhibition of regulatory T cell function, an effect on the activity of multiple cell types, such as regulatory T cell, effector T cells and NK cells), an increase in tumor infiltrating lymphocytes, an increase in T-cell receptor mediated proliferation, and a decrease in immune evasion by cancerous cells.
  • an increase in antigen presentation e.g., an increase in effector cell function
  • a method of modulating an immune response in a subject comprises administering to the subject a combination disclosed herein (e.g., a combination comprising a therapeutically effective amount of an anti-GITR antibody molecule), alone or in combination with one or more agents or procedures, such that the immune response in the subject is modulated.
  • the antibody molecule enhances, stimulates or increases the immune response in the subject.
  • the subject can be a mammal, e.g., a primate, preferably a higher primate, e.g., a human (e.g., a patient having, or at risk of having, a disorder described herein).
  • the subject is in need of enhancing an immune response.
  • the subject has, or is at risk of, having a disorder described herein, e.g., a cancer or an infectious disorder as described herein.
  • the subject is, or is at risk of being, immunocompromised.
  • the subject is undergoing or has undergone a chemotherapeutic treatment and/or radiation therapy.
  • the subject is, or is at risk of being, immunocompromised as a result of an infection.
  • an “antibody” refers to a polypeptide of the immunoglobulin family that is capable of noncovalently, reversibly, and specifically binding a corresponding antigen.
  • An exemplary antibody structural unit comprises a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” chain (about 25 kD) and one “heavy” chain (about 50-70 kD), connected through a disulfide bond. Recognized immunoglobulin genes include the ⁇ , ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ constant region genes, as well as the myriad immunoglobulin variable region genes. Light chains are classified as either ⁇ or ⁇ .
  • Heavy chains are classified as ⁇ , ⁇ , ⁇ , ⁇ , or ⁇ , which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD, and IgE, respectively.
  • Antibodies of the invention can be of any isotype/class (e.g., IgG, IgM, IgA, IgD, and IgE), or any subclass (e.g., IgG1, IgG2, IgG3, IgG4, IgA1, IgA2).
  • the N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the terms variable light chain (V L ) and variable heavy chain (V H ) refer to these regions of light and heavy chains respectively.
  • both heavy chains and light chains contain a constant (C) region or domain.
  • a secreted form of a immunoglobulin C region is made up of three C domains, CH1, CH2, CH3, optionally CH4 (C ⁇ ), and a hinge region.
  • a membrane-bound form of an immunglobulin C region also has membrane and intracellular domains.
  • Each light chain has a VL at the N-terminus followed by a constant domain (C) at its other end. The VL is aligned with the VH and the CL is aligned with the first constant domain of the heavy chain. The pairing of a VH and VL together forms a single antigen-binding site.
  • a “conventional antibody” IgG immunoglobulin as used herein refers to an antibody in a configuration that occurs in nature. Typically, a conventional antibody IgG has four chains, two identical heavy chains and two identical light chains linked together through disulfide bonds.
  • an “antibody” also encompasses variations of antibodies and conventional antibody structures that possess a particular binding specificity, i.e., for GITR. Thus, within the scope of this concept are full length antibodies, chimeric antibodies, and humanized antibodies, that possess a particular binding specificity for GITR.
  • Antibodies exist as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases.
  • pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)′ 2 , a dimer of Fab′ which itself is a light chain joined to V H -C H 1 by a disulfide bond.
  • the F(ab)′ 2 may be reduced under mild conditions to break the disulfide linkage in the hinge region, thereby converting the F(ab)′ 2 dimer into an Fab′ monomer.
  • the Fab′ monomer is essentially Fab with part of the hinge region. Paul, Fundamental Immunology 3d ed. (1993).
  • an “antibody fragment” refers to one or more portions of an antibody, either produced by the modification of whole antibodies, or those synthesized de novo using recombinant DNA methodologies, that retain binding specificity and agonist activity for GITR.
  • antibody fragments include Fv fragments, single chain antibodies (ScFv), Fab, Fab′, Fd (Vh and CH1 domains), dAb (Vh and an isolated CDR); and multimeric versions of these fragments (e.g., F(ab′) 2 ,) with the same binding specificity.
  • Antibody fragments can also be incorporated into single domain antibodies, maxibodies, minibodies, diabodies, triabodies, tetrabodies, vNAR, bis-scFv, and other variations of antibody-like compounds to achieve the binding specificity and activity provided in the present invention.
  • a “Fab” domain as used in the context of the invention comprises a heavy chain variable domain, a constant region CH1 domain, a light chain variable domain, and a light chain constant region CL domain. The interaction of the domains is stabilized by a disulfide bond between the CH1 and CL domains.
  • the heavy chain domains of the Fab are in the order, from N-terminus to C-terminus, VH-CH and the light chain domains of a Fab are in the order, from N-terminus to C-terminus, VL-CL.
  • the heavy chain domains of the Fab are in the order, from N-terminus to C-terminus, CH—VH and the light chain domains of the Fab are in the order CL-VL.
  • Fabs were historically identified by papain digestion of an intact immunoglobulin, in the context of this invention, a “Fab” is typically produced recombinantly by any method. Each Fab fragment is monovalent with respect to antigen binding, i.e., it has a single antigen-binding site.
  • An “Fc region” as used herein refers to the constant region of an antibody excluding the first constant region immunoglobulin domain.
  • Fc refers to the last two constant region immunoglobulin domains of IgA, IgD, and IgG, and the last three constant region immunoglobulin domains of IgE and IgM, and the flexible hinge N-terminal to these domains.
  • IgA and IgM Fc may include the J chain.
  • Fc comprises immunoglobulin domains C ⁇ 2 and C ⁇ 3 and the hinge between C ⁇ 1 and C ⁇ .
  • Fc region may vary, however, the human IgG heavy chain Fc region is usually defined to comprise residues C226 or P230 to its carboxyl-terminus, using the numbering is according to the EU index as in Kabat et al. (1991, NIH Publication 91-3242, National Technical Information Service, Springfield, Va.). “Fc region” may refer to this region in isolation or this region in the context of an antibody or antibody fragment. “Fc region” includes naturally occurring allelic variants of the Fc region, e.g., in the CH2 and CH3 region, as well as modifications that modulate effector function. Fc regions also include variants that don't result in alterations to biological function.
  • one or more amino acids can be deleted from the N-terminus or C-terminus of the Fc region of an immunoglobulin without substantial loss of biological function.
  • a C-terminal lysine may be modified replaced or removed.
  • one or more C-terminal residues in the Fc region is altered or removed.
  • one or more C-terminal residues in the Fc e.g., the terminal lysine
  • one or more C-terminal residues in the Fc is substituted with an alternate amino acid (e.g., the terminal lysine is replaced).
  • the Fc domain is the portion of the Ig recognized by cell receptors, such as the FcR, and to which the complement-activating protein, C1 q, binds.
  • the lower hinge region which is encoded in the 5′ portion of the CH2 exon, provides flexibility within the antibody for binding to FcR receptors.
  • CDRs complementarity-determining domains
  • CDRs complementary-determining regions
  • CDRs are structurally complementary to the epitope of the target protein and are thus directly responsible for the binding specificity.
  • the remaining stretches of the V L or V H the so-called framework regions, exhibit less variation in amino acid sequence (Kuby, Immunology, 4th ed., Chapter 4. W.H. Freeman & Co., New York, 2000).
  • Positions of the CDRs and framework regions can be determined using various well known definitions in the art, e.g., Kabat, Chothia, international ImMunoGeneTics database (IMGT) (on the worldwide web at imgt.cines.fr/), and AbM (see, e.g., Johnson et al., Nucleic Acids Res., 29:205-206 (2001); Chothia and Lesk, J. Mol. Biol., 196:901-917 (1987); Chothia et al., Nature, 342:877-883 (1989); Chothia et al., J. Mol. Biol., 227:799-817 (1992); Al-Lazikani et al., J. Mol.
  • CDR amino acid residues in the V H are numbered 31-35 (HCDR1), 50-65 (HCDR2), and 95-102 (HCDR3); and the CDR amino acid residues in the V L are numbered 24-34 (LCDR1), 50-56 (LCDR2), and 89-97 (LCDR3).
  • CDR amino acids in the V H are numbered 26-32 (HCDR1), 52-56 (HCDR2), and 95-102 (HCDR3); and the amino acid residues in V L are numbered 26-32 (LCDR1), 50-52 (LCDR2), and 91-96 (LCDR3).
  • the CDRs consist of amino acid residues 26-35 (HCDR1), 50-65 (HCDR2), and 95-102 (HCDR3) in human VH and amino acid residues 24-34 (LCDR1), 50-56 (LCDR2), and 89-97 (LCDR3) in human VL.
  • binding specificity determinant or “BSD” interchangeably refer to a minimum contiguous or non-contiguous amino acid sequence within a complementary determining region necessary for determining the binding specificity of an antibody.
  • a minimum binding specificity determinant can be within one or more CDR sequences.
  • the minimum binding specificity determinants reside within (i.e., are determined solely by) a portion or the full-length of the CDR3 sequences of the heavy and light chains of the antibody.
  • an “antibody light chain” or an “antibody heavy chain” as used herein refers to a polypeptide comprising the V L or V H , respectively.
  • the endogenous V L is encoded by the gene segments V (variable) and J (junctional), and the endogenous V H by V, D (diversity), and J.
  • Each of V L or V H includes the CDRs as well as the framework regions.
  • antibody light chains and/or antibody heavy chains may, from time to time, be collectively referred to as “antibody chains.” These terms encompass antibody chains containing mutations that do not disrupt the basic structure of V L or V H , as one skilled in the art will readily recognize.
  • valency refers to the number of potential target binding sites in a polypeptide. Each target binding site specifically binds one target molecule or specific site on a target molecule. When a polypeptide comprises more than one target binding site, each target binding site may specifically bind the same or different molecules (e.g., may bind to different molecules, e.g., different antigens, or different epitopes on the same molecule).
  • a conventional antibody for example, has two binding sites and is bivalent.
  • the antibodies, antigen binding molecules, and fragments thereof can be monovalent (i.e., bind one target molecule), bivalent, or multivalent (i.e., bind more than one target molecule).
  • any technique known in the art can be used (see, e.g., Kohler & Milstein, Nature 256:495-497 (1975); Kozbor et al., Immunology Today 4:72 (1983); Cole et al., Monoclonal Antibodies and Cancer Therapy, pp. 77-96. Alan R. Liss, Inc. 1985).
  • Techniques for the production of single chain antibodies (U.S. Pat. No. 4,946,778) can be adapted to produce antibodies to polypeptides of this invention.
  • transgenic mice, or other organisms such as other mammals may be used to express primatized or humanized antibodies.
  • phage display technology can be used to identify antibodies and heteromeric Fab fragments that specifically bind to selected antigens (see, e.g., McCafferty et al., supra; Marks et al., Biotechnology, 10:779-783, (1992)).
  • a primatized or humanized antibody has one or more amino acid residues introduced into it from a source which is non-primate or non-human. These non-primate or non-human amino acid residues are often referred to as import residues, which are typically taken from an import variable domain.
  • Humanization can be essentially performed following the method of Winter and co-workers (see, e.g., Jones et al., Nature 321:522-525 (1986); Riechmann et al., Nature 332:323-327 (1988); Verhoeyen et al., Science 239:1534-1536 (1988) and Presta, Curr. Op. Struct.
  • humanized antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species.
  • primatized or humanized antibodies are typically primate or human antibodies in which some complementary determining region (“CDR”) residues and possibly some framework (“FR”) residues are substituted by residues from analogous sites in an originating species (e.g., rodent antibodies) to confer binding specificity.
  • CDR complementary determining region
  • FR framework
  • a “chimeric antibody” is an antibody molecule in which (a) the constant region, or a portion thereof, is altered, replaced or exchanged so that the antigen binding site (variable region) is linked to a constant region of a different or altered class, effector function and/or species, or an entirely different molecule which confers new properties to the chimeric antibody, e.g., an enzyme, toxin, hormone, growth factor, and drug; or (b) the variable region, or a portion thereof, is altered, replaced or exchanged with a variable region having a different or altered antigen specificity.
  • Antibodies or antigen-binding molecules of the invention further include one or more immunoglobulin chains that are chemically conjugated to, or expressed as, fusion proteins with other proteins. It also includes bispecific antibody.
  • a bispecific or bifunctional antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
  • Other antigen-binding fragments or antibody portions of the invention include bivalent scFv (diabody), bispecific scFv antibodies where the antibody molecule recognizes two different epitopes, single binding domains (dAbs), and minibodies.
  • the various antibodies or antigen-binding fragments described herein can be produced by enzymatic or chemical modification of the intact antibodies, or synthesized de novo using recombinant DNA methodologies (e.g., single chain Fv), or identified using phage display libraries (see, e.g., McCafferty et al., Nature 348:552-554, 1990).
  • minibodies can be generated using methods described in the art, e.g., Vaughan and Sollazzo, Comb Chem High Throughput Screen. 4:417-30 2001.
  • Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab′ fragments. See, e.g., Songsivilai & Lachmann, Clin. Exp.
  • Single chain antibodies can be identified using phage display libraries or ribosome display libraries, gene shuffled libraries. Such libraries can be constructed from synthetic, semi-synthetic or native and immunocompetent sources.
  • antibody binding molecule or “non-antibody ligand” refers to antibody mimics that use non-immunoglobulin protein scaffolds, including but not limited to, adnectins, avimers, single chain polypeptide binding molecules, and antibody-like binding peptidomimetics.
  • variable region or “V-region” interchangeably refer to a heavy or light chain comprising FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4.
  • An endogenous variable region is encoded by immunoglobulin heavy chain V-D-J genes or light chain V-J genes.
  • a V-region can be naturally occurring, recombinant or synthetic.
  • variable segment or “V-segment” interchangeably refer to a subsequence of the variable region including FR1-CDR1-FR2-CDR2-FR3.
  • An endogenous V-segment is encoded by an immunoglobulin V-gene.
  • a V-segment can be naturally occurring, recombinant or synthetic.
  • J-segment refers to a subsequence of the variable region encoded comprising a C-terminal portion of a CDR3 and the FR4.
  • An endogenous J-segment is encoded by an immunoglobulin J-gene.
  • a J-segment can be naturally occurring, recombinant or synthetic.
  • a “humanized” antibody is an antibody that retains the reactivity (e.g., binding specificity, activity) of a non-human antibody while being less immunogenic in humans. This can be achieved, for instance, by retaining non-human CDR regions and replacing the remaining parts of the antibody with human counterparts. See, e.g., Morrison et al., Proc. Natl. Acad. Sci. USA, 81:6851-6855 (1984); Morrison and Oi, Adv. Immunol., 44:65-92 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988); Padlan, Molec. Immun., 28:489-498 (1991); Padlan, Molec. Immun., 31(3):169-217 (1994).
  • corresponding human germline sequence refers to the nucleic acid sequence encoding a human variable region amino acid sequence or subsequence that shares the highest determined amino acid sequence identity with a reference variable region amino acid sequence or subsequence in comparison to all other all other known variable region amino acid sequences encoded by human germline immunoglobulin variable region sequences.
  • the corresponding human germline sequence can also refer to the human variable region amino acid sequence or subsequence with the highest amino acid sequence identity with a reference variable region amino acid sequence or subsequence in comparison to all other evaluated variable region amino acid sequences.
  • the corresponding human germline sequence can be framework regions only, complementary determining regions only, framework and complementary determining regions, a variable segment (as defined above), or other combinations of sequences or subsequences that comprise a variable region. Sequence identity can be determined using the methods described herein, for example, aligning two sequences using BLAST, ALIGN, or another alignment algorithm known in the art.
  • the corresponding human germline nucleic acid or amino acid sequence can have at least about 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity with the reference variable region nucleic acid or amino acid sequence.
  • Corresponding human germline sequences can be determined, for example, through the publicly available international ImMunoGeneTics database (IMGT) (on the worldwide web at imgt.cines.fr/) and V-base (on the worldwide web at vbase.mrc-cpe.cam.ac.uk).
  • IMGT international ImMunoGeneTics database
  • V-base on the worldwide web at vbase.mrc-cpe.cam.ac.uk.
  • an antigen e.g., a protein
  • an antibody, antibody fragment, or antibody-derived binding agent refers to a binding reaction that is determinative of the presence of the antigen in a heterogeneous population of proteins and other biologics, e.g., in a biological sample, e.g., a blood, serum, plasma or tissue sample.
  • a biological sample e.g., a blood, serum, plasma or tissue sample.
  • the antibodies or binding agents with a particular binding specificity bind to a particular antigen at least two times the background and do not substantially bind in a significant amount to other antigens present in the sample.
  • the antibody or binding agents with a particular binding specificity bind to a particular antigen at least ten (10) times the background and do not substantially bind in a significant amount to other antigens present in the sample.
  • Specific binding to an antibody or binding agent under such conditions may require the antibody or agent to have been selected for its specificity for a particular protein (e.g., human GITR).
  • specific binding includes antibodies fragments thereof and binding molecules that selectively bind to human GITR and do not include antibodies that cross-react with, e.g., murine GITR molecules or other TNF receptor superfamily members.
  • antibodies or antibody fragments are selected that cross-react with non-human primate GITR (e.g., cynomolgus GITR).
  • immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein.
  • solid-phase ELISA immunoassays are routinely used to select antibodies specifically immunoreactive with a protein (see, e.g., Harlow & Lane, Using Antibodies, A Laboratory Manual (1998), for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity).
  • a specific or selective binding reaction will produce a signal at least twice over the background signal and more typically at least than 10 to 100 times over the background.
  • Equilibrium dissociation constant (K D , M) refers to the dissociation rate constant (k d , time ⁇ 1 ) divided by the association rate constant (k a , time ⁇ 1 , M ⁇ 1 ). Equilibrium dissociation constants can be measured using any known method in the art.
  • the antibodies of the present invention generally will have an equilibrium dissociation constant of less than about 10 ⁇ 7 or 10 ⁇ 8 M, for example, less than about 10 ⁇ 9 M or 10 ⁇ 10 M, in some embodiments, less than about 10 ⁇ 11 M, 10 ⁇ 12 M or 10 ⁇ 13 M.
  • the isolated antibody or antibody fragment binds to human GITR with an equilibrium dissociation constant (K D ) of about 1 nM or less. In some embodiments, the antibody or antibody fragment binds to human GITR with a K D that is less than 1 nM. In some embodiments, the antibody or antibody fragment binds to human GITR with a K D that is in the range of from about 0.5 nM to about 1.0 nM.
  • K D equilibrium dissociation constant
  • the term “antigen-binding region” refers to a domain of the GITR-binding molecule of this invention that is responsible for the specific binding between the molecule and GITR.
  • An antigen-binding region includes at least one antibody heavy chain variable region and at least one antibody light chain variable region. There are at least one such antigen-binding regions present in each GITR-binding molecule of this invention, and each of the antigen-binding regions may be identical or different from the others. In some embodiments, at least one of the antigen-binding regions of a GITR-binding molecule of this invention acts as an agonist of GITR.
  • antibody agonist or “agonist” interchangeably refer to an antibody capable of activating a receptor to induce a full or partial receptor-mediated response.
  • an agonist of GITR binds to GITR and induces GITR-mediated intracellular signaling (e.g., increased NF- ⁇ B expression activation).
  • the antibody agonist stimulates signaling through GITR similarly to the native ligand, GITR-L. Binding of GITR-L to GITR induces NF ⁇ B activation due to degradation of I ⁇ B.
  • a GITR antibody agonist can be identified by its ability to bind GITR and induce T cell (e.g., CD8 + CTLs or CD4 + Th cells) proliferation, survival, cytolytic activity and/or cytokine production (e.g., IFN ⁇ , IL-10, IL-13, TNF ⁇ ) or as otherwise described herein.
  • T cell e.g., CD8 + CTLs or CD4 + Th cells
  • cytolytic activity and/or cytokine production e.g., IFN ⁇ , IL-10, IL-13, TNF ⁇
  • GITR or “glucocorticoid-induced tumor necrosis factor receptor receptor” or “tumor necrosis factor receptor superfamily, member 18” or “TNFRSF18” interchangeably refer to a type I transmembrane protein that is a member of the TNF-receptor superfamily. GITR is expressed at high levels on CD4 + CD25 + and on activated effector CD4 + and CD8 + T cells. The nucleic acid and amino acid sequences of GITR are known, and have been published in
  • GenBank Accession Nos. NM_004195.2 ⁇ NP_004186.1 (isoform 1 precursor), SEQ ID NO: 1: 1 maqhgamgaf ralcglallc alslgqrptg gpgcgpgrll lgtgtdarcc rvhttrocrd 61 ypgeeccsew dcmcvqpefh cgdpccttcr hhpcppgqgv qsqgkfsfgf qcidcasgtf 121 sggheghckp wtdctqfgfl tvfpgnkthn avcvpgsppa eplgwltvvl lavaacvlll 181 tsaqlglhiw qlrsqcmwpr etqlllevpp stedarscqf peeergersa eekg
  • a GITR amino acid sequence is a type I transmembrane protein that is a member of the TNF-receptor superfamily having a signal peptide, an extracellular domain (ECD) comprising three cysteine-rich domains (CRDs) and has over its full length at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity with the amino acid sequence of GenBank accession numbers NP_004186.1 (SEQ ID NO:1), NP_683699.1 (SEQ ID NO:2), NP_683700.1 (SEQ ID NO:3), or NP_005083.2.
  • a GITR nucleic acid sequence has over its full length at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity with the nucleic acid sequence of GenBank accession numbers NM_004195.2, NM_148901.1, NM_148902.1, NM_005092 or SEQ ID NOs:1-4.
  • agonism of rodent GITR inhibits, at least transiently, suppressor activity of CD25 + regulatory T cells (Treg).
  • GITR agonism further enhances immunoactivity, e.g., proliferation, survival, cytokine production and cytolytic activity of activated effector CD4 + and CD8 + T cells.
  • Activity of a polypeptide of the invention refers to structural, regulatory, or biochemical functions of a polypeptide in its native cell or tissue. Examples of activity of a polypeptide include both direct activities and indirect activities. Exemplary activities of GITR agonism include intracellular signaling that results in increased activation of NF- ⁇ B, increased proliferation, survival, cytokine production (e.g., IFN ⁇ , IL-10, IL-13, TNF ⁇ ), and cytolytic activity of activated effector CD4 + and CD8 + T cells. Therapeutically, agonism of GITR augments antitumor and antiviral T-cell responses in vivo.
  • nucleic acid or protein when applied to a nucleic acid or protein, denotes that the nucleic acid or protein is essentially free of other cellular components with which it is associated in the natural state. It is preferably in a homogeneous state. It can be in either a dry or aqueous solution. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified. In particular, an isolated gene is separated from open reading frames that flank the gene and encode a protein other than the gene of interest. The term “purified” denotes that a nucleic acid or protein gives rise to essentially one band in an electrophoretic gel. Particularly, it means that the nucleic acid or protein is at least 85% pure, more preferably at least 95% pure, and most preferably at least 99% pure.
  • nucleic acid refers to deoxyribonucleic acids (DNA) or ribonucleic acids (RNA) and polymers thereof in either single- or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions), alleles, orthologs, SNPs, and complementary sequences as well as the sequence explicitly indicated.
  • DNA deoxyribonucleic acids
  • RNA ribonucleic acids
  • degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 (1985); and Rossolini et al., Mol. Cell. Probes 8:91-98 (1994)).
  • polypeptide “peptide,” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues.
  • the terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymer.
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, ⁇ -carboxyglutamate, and O-phosphoserine.
  • Amino acid analogs refer to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an ⁇ -carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.
  • “Conservatively modified variants” applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, conservatively modified variants refers to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG, and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide.
  • nucleic acid variations are “silent variations,” which are one species of conservatively modified variations. Every nucleic acid sequence herein which encodes a polypeptide also describes every possible silent variation of the nucleic acid.
  • each codon in a nucleic acid except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan
  • TGG which is ordinarily the only codon for tryptophan
  • amino acid sequences one of skill will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a “conservatively modified variant” where the alteration results in the substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are well known in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the invention.
  • the following eight groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Glycine (G); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W); 7) Serine (S), Threonine (T); and 8) Cysteine (C), Methionine (M) (see, e.g., Creighton, Proteins (1984)).
  • Percentage of sequence identity is determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (e.g., a polypeptide of the invention), which does not comprise additions or deletions, for optimal alignment of the two sequences.
  • the percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity.
  • nucleic acids or polypeptide sequences refer to two or more sequences or subsequences that are the same sequences.
  • Two sequences are “substantially identical” if two sequences have a specified percentage of amino acid residues or nucleotides that are the same (i.e., at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% sequence identity over a specified region, or, when not specified, over the entire sequence of a reference sequence), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection.
  • the invention provides polypeptides or polynucleotides that are substantially identical to the polypeptides or polynucleotides, respectively, exemplified herein (e.g., the variable regions exemplified in any one of SEQ ID NOS:6-10, 12, 14, 59 and 61; the variable segments exemplified in any one of SEQ ID NOS:16-17; the CDRs exemplified in any one of SEQ ID NOS:22-34; the FRs exemplified in any one of SEQ ID NOS:35-50; and the nucleic acid sequences exemplified in any on of SEQ ID NOS:51-58 and 60).
  • the identity exists over a region that is at least about 15, 25 or 50 nucleotides in length, or more preferably over a region that is 100 to 500 or 1000 or more nucleotides in length, or over the full length of the reference sequence.
  • identity or substantial identity can exist over a region that is at least 5, 10, 15 or 20 amino acids in length, optionally at least about 25, 30, 35, 40, 50, 75 or 100 amino acids in length, optionally at least about 150, 200 or 250 amino acids in length, or over the full length of the reference sequence.
  • shorter amino acid sequences e.g., amino acid sequences of 20 or fewer amino acids
  • substantial identity exists when one or two amino acid residues are conservatively substituted, according to the conservative substitutions defined herein.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated.
  • sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters.
  • a “comparison window”, as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Methods of alignment of sequences for comparison are well known in the art. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1970) Adv. Appl. Math. 2:482c, by the homology alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol.
  • BLAST and BLAST 2.0 algorithms Two examples of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1977) Nuc. Acids Res. 25:3389-3402, and Altschul et al. (1990) J. Mol. Biol. 215:403-410, respectively.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et al., supra).
  • initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them.
  • the word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always ⁇ 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
  • the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5787).
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • P(N) the smallest sum probability
  • a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, more preferably less than about 0.01, and most preferably less than about 0.001.
  • nucleic acid sequences or polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is immunologically cross reactive with the antibodies raised against the polypeptide encoded by the second nucleic acid, as described below.
  • a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative substitutions.
  • Another indication that two nucleic acid sequences are substantially identical is that the two molecules or their complements hybridize to each other under stringent conditions, as described below.
  • Yet another indication that two nucleic acid sequences are substantially identical is that the same primers can be used to amplify the sequence.
  • link when used in the context of describing how the antigen-binding regions are connected within a GITR-binding molecule of this invention, encompasses all possible means for physically joining the regions.
  • the multitude of antigen-binding regions are frequently joined by chemical bonds such as a covalent bond (e.g., a peptide bond or a disulfide bond) or a non-covalent bond, which can be either a direct bond (i.e., without a linker between two antigen-binding regions) or indirect bond (i.e., with the aid of at least one linker molecule between two or more antigen-binding regions).
  • the terms “subject,” “patient,” and “individual” interchangeably refer to a mammal, for example, a human or a non-human primate mammal.
  • the mammal can also be a laboratory mammal, e.g., mouse, rat, rabbit, hamster.
  • the mammal can be an agricultural mammal (e.g., equine, ovine, bovine, porcine, camelid) or domestic mammal (e.g., canine, feline).
  • the terms “treat,” “treating,” or “treatment” of any disease or disorder refer in one embodiment, to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof).
  • “treat,” “treating,” or “treatment” refers to alleviating or ameliorating at least one physical parameter including those which may not be discernible by the patient.
  • “treat,” “treating,” or “treatment” refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both.
  • “treat,” “treating,” or “treatment” refers to preventing or delaying the onset or development or progression of the disease or disorder.
  • therapeutically acceptable amount or “therapeutically effective dose” interchangeably refer to an amount sufficient to effect the desired result (i.e., a reduction in tumor size, inhibition of tumor growth, prevention of metastasis, inhibition or prevention of viral, bacterial, fungal or parasitic infection). In some embodiments, a therapeutically acceptable amount does not induce or cause undesirable side effects.
  • a therapeutically acceptable amount can be determined by first administering a low dose, and then incrementally increasing that dose until the desired effect is achieved.
  • a “prophylactically effective amount,” and a “therapeutically effective amount,” of a GITR agonizing antibody of the invention can prevent the onset of, or result in a decrease in severity of, respectively, disease symptoms, including symptoms associated with cancer or infectious disease.
  • a “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result.
  • a therapeutically effective amount of the modified antibody or antibody fragment may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the modified antibody or antibody fragment is outweighed by the therapeutically beneficial effects.
  • a “therapeutically effective dosage” preferably inhibits a measurable parameter, e.g., tumor growth rate by at least about 20%, more preferably by at least about 40%, even more preferably by at least about 60%, and still more preferably by at least about 80% relative to untreated subjects.
  • the ability of a compound to inhibit a measurable parameter, e.g., cancer can be evaluated in an animal model system predictive of efficacy in human tumors. Alternatively, this property of a composition can be evaluated by examining the ability of the compound to inhibit, such inhibition in vitro by assays known to the skilled practitioner.
  • co-administer refers to the simultaneous presence of two active agents in the blood of an individual. Active agents that are co-administered can be concurrently or sequentially delivered.
  • the phrase “consisting essentially of” refers to the genera or species of active pharmaceutical agents included in a method or composition, as well as any inactive carrier or excipients for the intended purpose of the methods or compositions. In some embodiments, the phrase “consisting essentially of” expressly excludes the inclusion of one or more additional active agents other than an agonist anti-GITR antibody of the invention. In some embodiments, the phrase “consisting essentially of” expressly excludes the inclusion of one or more additional active agents other than an agonist anti-GITR antibody of the invention and a second co-administered agent.
  • cancer refers to a disease characterized by the rapid and uncontrolled growth of aberrant cells. Cancer cells can spread locally or through the bloodstream and lymphatic system to other parts of the body. Examples of various cancers are described herein and include but are not limited to, breast cancer, prostate cancer, ovarian cancer, cervical cancer, skin cancer, pancreatic cancer, colorectal cancer, renal cancer, liver cancer, brain cancer, lymphoma, leukemia, lung cancer and the like.
  • tumor and “cancer” are used interchangeably herein, e.g., both terms encompass solid and liquid, e.g., diffuse or circulating, tumors. As used herein, the term “cancer” or “tumor” includes premalignant, as well as malignant cancers and tumors.
  • cancer-associated antigen or “tumor-associated antigen” or “tumor-specific marker” or “tumor marker” interchangeably refers to a molecule (typically protein, carbohydrate or lipid) that is preferentially expressed on the surface of a cancer cell in comparison to a normal cell, and which is useful for the preferential targeting of a pharmacological agent to the cancer cell.
  • a cancer-associated antigen is a cell surface molecule that is overexpressed in a cancer cell in comparison to a normal cell, for instance, 1-fold over expression, 2-fold overexpression, 3-fold overexpression or more in comparison to a normal cell.
  • a cancer-associated antigen is a cell surface molecule that is inappropriately synthesized in the cancer cell, for instance, a molecule that contains deletions, additions or mutations in comparison to the molecule expressed on a normal cell. Oftentimes, a cancer-associated antigen will be expressed exclusively on the cell surface of a cancer cell and not synthesized or expressed on the surface of a normal cell.
  • Exemplified cell surface tumor markers include the proteins c-erbB-2 and human epidermal growth factor receptor (HER) for breast cancer, PSMA for prostate cancer, and carbohydrate mucins in numerous cancers, including breast, ovarian and colorectal.
  • an immune system cell such as an accessory cell (e.g., a B-cell, a dendritic cell, and the like) that displays a foreign antigen complexed with major histocompatibility complexes (MHC's) on its surface.
  • MHC's major histocompatibility complexes
  • T-cells may recognize these complexes using their T-cell receptors (TCRs).
  • TCRs T-cell receptors
  • costimulatory molecule refers to the cognate binding partner on a T cell that specifically binds with a costimulatory ligand, thereby mediating a costimulatory response by the T cell, such as, but not limited to, proliferation.
  • Costimulatory molecules are cell surface molecules other than antigen receptors or their ligands that are required for an efficient immune response.
  • Costimulatory molecules include, but are not limited to an MHC class I molecule, TNF receptor proteins, Immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocytic activation molecules (SLAM proteins), activating NK cell receptors, BTLA, a Toll ligand receptor, OX40, CD2, CD7, CD27, CD28, CD30, CD40, CDS, ICAM-1, LFA-1 (CD11a/CD18), 4-1BB (CD137), B7-H3, CDS, ICAM-1, ICOS (CD278), GITR, BAFFR, LIGHT, HVEM (LIGHTR), KIRDS2, SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA
  • Immuno effector cell refers to a cell that is involved in an immune response, e.g., in the promotion of an immune effector response.
  • immune effector cells include T cells, e.g., alpha/beta T cells and gamma/delta T cells, B cells, natural killer (NK) cells, natural killer T (NKT) cells, mast cells, and myeloid-derived phagocytes.
  • Immuno effector or “effector” “function” or “response,” as that term is used herein, refers to function or response, e.g., of an immune effector cell, that enhances or promotes an immune attack of a target cell.
  • an immune effector function or response refers a property of a T or NK cell that promotes killing or the inhibition of growth or proliferation, of a target cell.
  • primary stimulation and co-stimulation are examples of immune effector function or response.
  • effector function refers to a specialized function of a cell. Effector function of a T cell, for example, may be cytolytic activity or helper activity including the secretion of cytokines.
  • the terms “first”, “second”, “third” and “fourth”, with respect to antigen binding moieties, e.g., Fabs, are used for convenience of distinguishing when there is more than one of each moiety. Use of these terms is not intended to confer a specific order or orientation of the antibody unless otherwise stated.
  • the present invention provides antibodies, antibody fragments, and antigen binding molecules that bind to and stimulate signaling through GITR and/or induce a potentiated immune response in vivo.
  • the antibodies, antibody fragments, and antigen binding molecules find uses in enhancing CD4+ T helper (Th) and/or CD8+ cytolytic T lymphocyte (CTL) responses against a target antigen. They also find uses in treating disease conditions whose progression can be reversed or inhibited by an effective immune response, including cancers and infectious diseases.
  • the antibodies, antibody fragments and antigen binding molecules of the present invention show suitable properties to be used in human patients, for example, they have low risk for immunogenicity issues for uses in human (they are encoded by human germline nucleic acid sequences, with the exception of the binding specificity determining regions (BSD), in particular at least CDR3); have high affinity to GITR (e.g., K D is at least less than 5 nM); do not cross-react with other members of the TNFR superfamily; cross-react with human and non-human primate GITR; and agonize GITR signaling at low doses (e.g., in concentrations of less than 5 nM in in vitro assays). Other activities and characteristics are also demonstrated throughout the specification.
  • BSD binding specificity determining regions
  • the present invention provides antibodies, antibody fragments, and antigen-binding molecules that are agonists of GITR.
  • Provided anti-GITR antibodies, antibody fragments, or antigen-binding molecules contain a minimum binding sequence determinant (BSD) within the CDR3 of the heavy and light chains derived from the originating or reference monoclonal antibody, for example, the antibodies described in Table 1 and Table 2 below.
  • the remaining sequences of the heavy chain and light chain variable regions (CDR and FR), e.g., V-segment and J-segment, are from corresponding human germline and affinity matured amino acid sequences.
  • the V-segments can be selected from a human V-segment library. Further sequence refinement can be accomplished by affinity maturation or other methods known in the art to optimize binding activity or activity of the antibodies, antibody fragments or antigen binding molecules of the invention.
  • heavy and light chains of the anti-GITR antibodies or antibody fragments contain a human V-segment from the corresponding human germline sequence (FR1-CDR1-FR2-CDR2-FR3), e.g., selected from a human V-segment library, and a CDR3-FR4 sequence segment from the originating monoclonal antibody (e.g., the antibodies as described in Table 1 and Table 2).
  • the CDR3-FR4 sequence segment can be further refined by replacing sequence segments with corresponding human germline sequences and/or by affinity maturation.
  • the FR4 and/or the CDR3 sequence surrounding the BSD can be replaced with the corresponding human germline sequence, while the BSD from the CDR3 of the originating monoclonal antibody is retained.
  • the corresponding human germline sequence for the heavy chain V-segment is VH3 3-13/30: EVQLVESGGGLVQPGGSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVAVIRYDGSN KYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK (SEQ ID NO:89).
  • the last amino acid in SEQ ID NO:89, lysine (“K”), is substituted with arginine (“R”).
  • the corresponding human germline sequence for the heavy chain J-segment is JH4.
  • the heavy chain J-segment comprises the human germline JH4 partial sequence WGQGTLVTVSS (SEQ ID NO:90).
  • the full-length J-segment from human germline JH4 is YFDYWGQGTLVTVSS (SEQ ID NO:91).
  • the variable region genes are referenced in accordance with the standard nomenclature for immunoglobulin variable region genes. Current immunoglobulin gene information is available through the worldwide web, for example, on the ImMunoGeneTics (IMGT), V-base and PubMed databases. See also, Lefranc, Exp Clin Immunogenet. 2001; 18(2):100-16; Lefranc, Exp Clin Immunogenet.
  • the corresponding human germline sequence for the light chain V-segment is VKIII L16/A27: EIVMTQSPATLSVSPGERATLSCRASQSVSSNLAWYQQKPGQAPRLLIYGASTRATGIPD RFSGSGSGTDFTLTISRLEPEDFAVYYCQQYGSSP (SEQ ID NO:92).
  • the corresponding human germline sequence for the light chain J-segment is JK2.
  • the light chain J-segment comprises the human germline Jk2 partial sequence FGQGTKLEIK (SEQ ID NO:93). The full-length J segment from human germline Jk2 is YTFGQGTKLEIK (SEQ ID NO:94).
  • the heavy chain V-segment has at least 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence
  • the light chain V-segment has at least 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence
  • the heavy chain CDR3 comprises the amino acid sequence HAYGHDGGFAMDY (SEQ ID NO:29) or NAYGHDGGFAMDY (SEQ ID NO:109); and ii) the light chain CDR3 variable region comprises the amino acid sequence GQSYSYPFT (SEQ ID NO:34), or SYSYPF (SEQ ID NO:83).
  • the antibodies or antibody fragments of the invention comprise a heavy chain variable region comprising a CDR1 comprising an amino acid sequence SYGVD (SEQ ID NO:22), or GFSLSSY (SEQ ID NO:84); a CDR2 comprising an amino acid sequence VIWGGGGTYY(A/T)(A/S)S(L/V)M(A/G) (SEQ ID NO:28), or WGGGG (SEQ ID NO:80); and a CDR3 comprising an amino acid sequence of HAYGHDGGFAMDY (SEQ ID NO:29) or NAYGHDGGFAMDY (SEQ ID NO:109).
  • the antibodies or antibody fragments of the invention comprise a light chain variable region comprising a CDR1 comprising an amino acid sequence RAS(E/Q)SVSSN(L/V)A (SEQ ID NO:32) or S(E/Q)SVSSN (SEQ ID NO:87); a CDR2 comprising an amino acid sequence GASNRAT (SEQ ID NO:33), or GAS (SEQ ID NO:82); and a CDR3 comprising an amino acid sequence of GQSYSYPFT (SEQ ID NO:34), or SYSYPF (SEQ ID NO:83).
  • the antibodies or antibody fragments of the invention comprise a heavy chain variable region comprising a CDR1 comprising an amino acid sequence SYGVD (SEQ ID NO:22), or GFSLSSY (SEQ ID NO:84); a CDR2 comprising an amino acid sequence VIWGGGGTYY(A/T)(A/S)S(L/V)M(A/G) (SEQ ID NO:28) or WGGGG (SEQ ID NO:80); and a CDR3 comprising an amino acid sequence of HAYGHDGGFAMDY (SEQ ID NO:29) or NAYGHDGGFAMDY (SEQ ID NO:109).
  • a CDR1 comprising an amino acid sequence SYGVD (SEQ ID NO:22), or GFSLSSY (SEQ ID NO:84)
  • a CDR2 comprising an amino acid sequence VIWGGGGTYY(A/T)(A/S)S(L/V)M(A/G) (SEQ ID NO:28) or WGGGG (SEQ ID
  • Such antibodies or antibody fragments of the invention further comprise a light chain variable region comprising a CDR1 comprising an amino acid sequence RAS(E/Q)SVSSN(L/V)A (SEQ ID NO:32), or S(E/Q)SVSSN (SEQ ID NO:87); a CDR2 comprising an amino acid sequence GASNRAT (SEQ ID NO:33), or GAS (SEQ ID NO:82); and a CDR3 comprising an amino acid sequence of GQSYSYPFT (SEQ ID NO:34), or SYSYPF (SEQ ID NO:83).
  • the antibodies or antibody fragments of the invention comprise a heavy chain variable region comprising a CDR1 comprising an amino acid sequence SYGVD (SEQ ID NO:22), or GFSLRSY (SEQ ID NO:79); a CDR2 comprising an amino acid sequence VIWGGGGTNYNSALMA (SEQ ID NO:62), or WGGGG (SEQ ID NO:80); and a CDR3 comprising an amino acid sequence of HAYGHDGGFAMDY (SEQ ID NO:29) or NAYGHDGGFAMDY (SEQ ID NO:109).
  • the antibodies or antibody fragments are humanized.
  • the antibodies or antibody fragments of the invention comprise a light chain variable region comprising a CDR1 comprising an amino acid sequence KASENVDTFVS (SEQ ID NO:63), or SENVDTF (SEQ ID NO:81); a CDR2 comprising an amino acid sequence GASNRYT (SEQ ID NO:64), or GAS (SEQ ID NO:82); and a CDR3 comprising an amino acid sequence of GQSYSYPFT (SEQ ID NO:34), or SYSYPF (SEQ ID NO:83).
  • the antibodies or antibody fragments are humanized.
  • the antibodies or antibody fragments of the invention comprise a heavy chain variable region comprising a CDR1 comprising an amino acid sequence SYGVD (SEQ ID NO:22), or GFSLRSY (SEQ ID NO:79); a CDR2 comprising an amino acid sequence VIWGGGGTNYNSALMA (SEQ ID NO:62), or WGGGG (SEQ ID NO:80); and a CDR3 comprising an amino acid sequence of HAYGHDGGFAMDY (SEQ ID NO:29) or NAYGHDGGFAMDY (SEQ ID NO:109).
  • Such antibodies or antibody fragments further comprise a light chain variable region comprising a CDR1 comprising an amino acid sequence KASENVDTFVS (SEQ ID NO:63), or SENVDTF (SEQ ID NO:81); a CDR2 comprising an amino acid sequence GASNRYT (SEQ ID NO:64), or GAS (SEQ ID NO:82); and a CDR3 comprising an amino acid sequence of GQSYSYPFT (SEQ ID NO:34), or SYSYPF (SEQ ID NO:83).
  • the antibodies or antibody fragments are humanized.
  • the heavy chain variable region comprises a FR1 comprising the amino acid sequence of (E/Q)VQLVESGGGLVQ(P/S)GGSLRLSCAASGFSLS (SEQ ID NO:37); a FR2 comprising the amino acid sequence of WVRQAPGKGLEW(L/V)G (SEQ ID NO:40); a FR3 comprising the amino acid sequence of RFTISRDNSKNTLYLQMNSLRAEDTAVYYCA(K/R) (SEQ ID NO:41); and a FR4 comprising the amino acid sequence of WGQGTLVTVSS (SEQ ID NO:42).
  • the heavy chain variable region comprises a FR1 comprising the amino acid sequence selected from QVQLVESGGGLVQPGGSLRLSCAASGFSLS (SEQ ID NO:35) and QVQLVESGGGLVQPGGSLRLSCAASGFSLS (SEQ ID NO:36); a FR2 comprising the amino acid sequence selected from WVRQAPGKGLEWVG (SEQ ID NO:38) and WVRQAPGKGLEWLG (SEQ ID NO:39); a FR3 comprising the amino acid sequence of SEQ ID NO:41; and a FR4 comprising the amino acid sequence of SEQ ID NO:42.
  • the identified amino acid sequences may have one or more substituted amino acids (e.g., from affinity maturation) or one or two conservatively substituted amino acids.
  • the light chain variable region comprises a FR1 comprising an amino acid sequence of EIVMTQSPATLSVSPGERATLSC (SEQ ID NO:43); a FR2 comprising the amino acid sequence of WYQQ(K/R)PGQAPRLLIY (SEQ ID NO:46); a FR3 comprising the amino acid sequence of GIP(A/D)RFSGSGSGTDFTLTISRLEPEDFAVYYC (SEQ ID NO:49); and a FR4 comprising the amino acid sequence of SEQ ID NO:50.
  • the light chain variable region comprises a FR1 comprising an amino acid sequence of SEQ ID NO:43; a FR2 comprising the amino acid sequence selected from WYQQRPGQAPRLLIY (SEQ ID NO:44) and WYQQKPGQAPRLLIY (SEQ ID NO:45); a FR3 comprising the amino acid sequence selected from GIPARFSGSGSGTDFTLTISRLEPEDFAVYYC (SEQ ID NO:47) and GIPDRFSGSGSGTDFTLTISRLEPEDFAVYYC (SEQ ID NO:48); and a FR4 comprising the amino acid sequence of FGQGTKLEIK (SEQ ID NO:50).
  • the identified amino acid sequences may have one or more substituted amino acids (e.g., from affinity maturation) or one or two conservatively substituted amino acids.
  • variable regions of the anti-GITR antibodies of the present invention generally will have an overall variable region (e.g., FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4) amino acid sequence identity of at least about 85%, for example, at least about 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% to the corresponding human germline variable region amino acid sequence.
  • overall variable region e.g., FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4 amino acid sequence identity of at least about 85%, for example, at least about 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% to the corresponding human germline variable region amino acid sequence.
  • the heavy chain of the anti-GITR antibodies can have at least about 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to the human germline variable region EVQLVESGGGLVQPGGSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVAVIRYDGSN KYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK-YFDYWGQGTLVTVSS (SEQ ID NOS:89 and 91)(VH3 3-13/30+CDR3+JH4, the hyphen represents CDR3, which may be variable in length).
  • the last amino acid in SEQ ID NO:89, lysine (K), is substituted with arginine (R).
  • the light chain of the anti-GITR antibodies can have at least about 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to the human germline variable region EIVMTQSPATLSVSPGERATLSCRASQSVSSNLAWYQQKPGQAPRLLIYGASTRATGIPD RFSGSGSGTDFTLTISRLEPEDFAVYYC-YTFGQGTKLEIK (SEQ ID NOS:98 and 94) (VKIII L16/A27+CDR3+JK2; the hyphen represents CDR3, which may be variable in length).
  • the sequence identity comparison excludes the CDR3.
  • the CDRs of the antibodies listed in Table 1 can be determined by well known numbering systems known in the art, including those described herein.
  • Table 2 listed the CDRs that are defined by (1) using the numbering system described in Kabat et al. (1991), “Sequences of Proteins of Immunological Interest,” 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (“Kabat” numbering scheme), NIH publication No. 91-3242; and (2) Chothia, see Al-Lazikani et al., (1997) “Standard conformations for the canonical structures of immunoglobulins,” J. Mol. Biol. 273:927-948.
  • the anti-GITR antibodies or antibody fragments of the invention that binds to GITR is selected from any one of: i) an antibody, antibody fragment, or antigen binding molecule wherein: the heavy chain CDR1 comprises SEQ ID NO:22, the heavy chain CDR2 comprises SEQ ID NO:23, the heavy chain CDR3 comprises SEQ ID NO:29, the light chain CDR1 comprises SEQ ID NO:30, the light chain CDR2 comprises SEQ ID NO:33, and the light chain CDR3 comprises SEQ ID NO:34; ii) an antibody, antibody fragment, or antigen binding molecule wherein: the heavy chain CDR1 comprises SEQ ID NO:22, the heavy chain CDR2 comprises SEQ ID NO:24, the heavy chain CDR3 comprises SEQ ID NO:29, the light chain CDR1 comprises SEQ ID NO:31, the light chain CDR2 comprises SEQ ID NO:33, and the light chain CDR3 comprises
  • the antibodies or antibody fragments are humanized. In particular embodiments the antibodies or antibody fragments comprise a human constant region. In some embodiments the antibodies or antibody fragments comprise an IgG Fc region. In certain embodiments the antibody or antigen binding fragment is glycosylated. In some embodiments the antibodies or antibody fragments are modified or expressed in a modified cell, wherein such modification results in increased FcR effector function of the antibody or antibody fragment. In certain embodiments the antibody or antigen fragment induces an elevated Teff:Treg ratio in vivo. In some embodiments the antibody or antibody fragment induces a potentiated immune response in vivo. In some embodiments when the antibody or antibody fragment is cross linked to a second antibody or antibody fragment it is an agonist of SEQ ID NO:1, SEQ ID NO:2 or SEQ ID NO3.
  • the anti-GITR antibodies or antibody fragments of the invention comprise a heavy chain variable region having at least 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:16 and comprise a light chain variable region having at least 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO:17.
  • the anti-GITR antibodies or antibody fragments of the invention comprise a heavy chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:6 and comprise a light chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO:7.
  • the anti-GITR antibodies or antibody fragments of the invention comprise a heavy chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:8 and comprise a light chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO:9.
  • the anti-GITR antibodies or antibody fragments of the invention comprise a heavy chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:10 and comprise a light chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO:7.
  • the anti-GITR antibodies or antibody fragments of the invention comprise a heavy chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:12 and comprise a light chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO:7.
  • the anti-GITR antibodies or antibody fragments of the invention comprise a heavy chain polypeptide having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:14 and comprise a light chain polypeptide having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO:7.
  • the anti-GITR antibodies or antibody fragments of the invention comprise a heavy chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:99 and comprise a light chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO:7.
  • the anti-GITR antibodies or antibody fragments of the invention comprise a heavy chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:105 and comprise a light chain variable region having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO:7.
  • the anti-GITR antibodies or antibody fragments of the invention comprise a heavy chain polypeptide having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to a heavy chain variable region of SEQ ID NO:61 and comprise a light chain polypeptide having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to a light chain variable region of SEQ ID NO:59.
  • the anti-GITR antibodies of the present invention generally can have an overall constant region (e.g., IgG1) amino acid sequence identity of at least about 85%, for example, at least about 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% to human constant region, e.g., IgG1/kappa constant region amino acid sequences.
  • an overall constant region e.g., IgG1 amino acid sequence identity of at least about 85%, for example, at least about 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% to human constant region, e.g., IgG1/kappa constant region amino acid sequences.
  • the heavy chain of the anti-GITR antibodies can have at least about 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to the human IgG1 constant region ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSS GLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGG PSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQY NSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSR EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDK SRWQQGNVFSCSVMHEAL
  • the last amino acid, lysine (K), is substituted with arginine (R).
  • the light chain of the anti-GITR antibodies can have at least about 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to the human kappa light chain constant region
  • such antibody is a human or humanized antibody.
  • the VH, VL, full length light chain, and full length heavy chain sequences (amino acid sequences and the nucleotide sequences encoding the amino acid sequences) can be “mixed and matched” to create other GITR-binding antibodies of the invention.
  • Such “mixed and matched” GITR-binding antibodies can be tested using the binding assays known in the art (e.g., ELISAs, and other assays described in the Example section) to confirm activity.
  • binding assays known in the art (e.g., ELISAs, and other assays described in the Example section) to confirm activity.
  • a VH sequence from a particular VH/VL pairing should be replaced with a structurally similar VH sequence.
  • a full length heavy chain sequence from a particular full length heavy chain/full length light chain pairing should be replaced with a structurally similar full length heavy chain sequence.
  • a VL sequence from a particular VH/VL pairing should be replaced with a structurally similar VL sequence.
  • a full length light chain sequence from a particular full length heavy chain/full length light chain pairing should be replaced with a structurally similar full length light chain sequence.
  • the invention provides an isolated monoclonal antibody or antibody fragment having: a heavy chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOS:6, 8, 10, 12, 14, 99 and 105; and a light chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOS:7 and 9; wherein the antibody specifically binds to GITR.
  • the anti-GITR antibodies or antibody fragments of the invention comprise a heavy chain polypeptide having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to a heavy chain sequence selected from any of SEQ ID NO:65, SEQ ID NO:69, SEQ ID NO:73, SEQ ID NO:75, SEQ ID NO:77, SEQ ID NO:100 and SEQ ID NO:106; and comprise a light chain polypeptide having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to a light chain of SEQ ID NO:66 or SEQ ID NO:70.
  • the anti-GITR antibodies or antibody fragments of the invention comprise a heavy chain polypeptide selected from any of SEQ ID NO:65, SEQ ID NO:69, SEQ ID NO:73, SEQ ID NO:75, SEQ ID NO:77, SEQ ID NO:100 and SEQ ID NO:106; and comprise a light chain polypeptide of SEQ ID NO:66 or SEQ ID NO:70.
  • Anti-GITR antibodies and antibody fragments of the present invention generally will bind GITR, including isoform 1(SEQ ID NO:1), isoform 2(SEQ ID NO:2) and isoform 3(SEQ ID NO:3), with an equilibrium dissociation constant (K D ) of less than about 10 ⁇ 8 M or 10 ⁇ 9 M, for example, or less than about 10 ⁇ 10 M or 10 ⁇ 11 M, and in some embodiments, less than about 10 ⁇ 12 M or 10 ⁇ 13 M.
  • K D equilibrium dissociation constant
  • the present invention provides antibodies and antibody fragments that bind to an epitope comprising the cysteine-rich domain 1 (“CRD1”, SEQ ID NO:4: CGPGRLLLGTGTDARCCRVHTTRCCRDYPGEECCSEWDC) and the cysteine-rich domain 2 (“CRD2”, SEQ ID NO:5: MCVQPEFHCGDPCCTTCRHHPCPPGQGVQSQGKFSFGFQC) of human GITR, and wherein the antibody, antibody fragment, or the antigen binding molecule is an agonist of hGITR, and wherein the antibody, antibody fragment, or the antigen binding molecule optionally has an intact or increased FcR effector function.
  • CCD1 cysteine-rich domain 1
  • CCD2 cysteine-rich domain 2
  • MCVQPEFHCGDPCCTTCRHHPCPPGQGVQSQGKFSFGFQC MCVQPEFHCGDPCC
  • an antibody, antibody fragment, or the antigen binding molecule binds to an epitope comprising SEQ ID NO:88) of human GITR.
  • an epitope comprises residues within SEQ ID NO:88.
  • an epitope comprises amino acid residues within residues 34-72 and 78 of human GITR, where such antibodies and antibody fragments are agonists of hGITR.
  • the present invention also provides antibodies and antibody fragments that bind to the same epitope as do the GITR-binding antibodies described in Table 1. Additional antibodies and antibody fragments can therefore be identified based on their ability to cross-compete (e.g., to competitively inhibit the binding of, in a statistically significant manner) with other antibodies of the invention in GITR binding assays.
  • test antibody to inhibit the binding of antibodies and antibody fragments of the present invention to a GITR protein (e.g., human GITR) demonstrates that the test antibody can compete with that antibody or antibody fragment for binding to hGITR; such an antibody may, according to non-limiting theory, bind to the same or a related (e.g., a structurally similar or spatially proximal) epitope on the GITR protein as the antibody or antibody fragment with which it competes.
  • the antibody that binds to the same epitope on hGITR as the antibodies or antibody fragments of the present invention is a human or humanized monoclonal antibody.
  • Such human or humanized monoclonal antibodies can be prepared and isolated as described herein.
  • An antibody or antibody fragment of the invention further can be prepared using an antibody having one or more of the CDRs and/or VH and/or VL sequences shown herein (e.g., Table 1) as starting material to engineer a modified antibody or antibody fragment, which modified antibody may have altered properties from the starting antibody.
  • An antibody or antibody fragment can be engineered by modifying one or more residues within one or both variable regions (i.e., VH and/or VL), for example within one or more CDR regions and/or within one or more framework regions. Additionally or alternatively, an antibody or antibody fragment can be engineered by modifying residues within the constant region(s), for example to alter the effector function(s) of the antibody.
  • CDR grafting One type of variable region engineering that can be performed is CDR grafting.
  • Antibodies interact with target antigens predominantly through amino acid residues that are located in the six heavy and light chain complementarity determining regions (CDRs). For this reason, the amino acid sequences within CDRs are more diverse between individual antibodies than sequences outside of CDRs. Because CDR sequences are responsible for most antibody-antigen interactions, it is possible to express recombinant antibodies that mimic the properties of a specific antibody by constructing expression vectors that include CDR sequences from the specific antibody grafted onto framework sequences from a different antibody with different properties (see, e.g., Riechmann, L. et al., 1998 Nature 332:323-327; Jones, P.
  • another embodiment of the invention pertains to an isolated monoclonal antibody, or an antigen binding fragment thereof, comprising a heavy chain variable region comprising CDR1 sequence having an amino acid sequence selected from the group consisting of SEQ ID NOS:22, 79, and 84; CDR2 sequences having an amino acid sequence selected from the group consisting of SEQ ID NOS:23, 24, 25, 26, 27, 62, and 80; CDR3 sequences having an amino acid sequence selected from the group consisting of SEQ ID NOS:29, 34 and 109, respectively; and a light chain variable region having CDR1 sequences having an amino acid sequence selected from the group consisting of SEQ ID NOS:30, 31, 63, 81, 85, and 86; CDR2 sequences having an amino acid sequence selected from the group consisting of SEQ ID NOS:33, 64, and 82; and CDR3 sequences consisting of an amino acid sequence selected from the group consisting of SEQ ID NOS:34 and 83; respectively.
  • the isolated antibodies or antibody fragments comprise sequences that have amino acid sequence identity of at least about 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% to the corresponding sequences in this paragraph.
  • Such framework sequences can be obtained from public DNA databases or published references that include germline antibody gene sequences.
  • germline DNA sequences for human heavy and light chain variable region genes can be found in the “VBase” human germline sequence database (available on the Internet at www.mrc-cpe.cam.ac.uk/vbase), as well as in Kabat, E. A., et al., 1991 Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242; Tomlinson, I. M., et al., 1992 J. fol. Biol. 227:776-798; and Cox, J. P. L. et al., 1994 Eur. J Immunol. 24:827-836.
  • framework sequences for use in the antibodies of the invention are those that are structurally similar to the framework sequences used by selected antibodies of the invention, e.g., consensus sequences and/or framework sequences used by monoclonal antibodies of the invention.
  • the VH CDR1, 2 and 3 sequences, and the VL CDR1, 2 and 3 sequences can be grafted onto framework regions that have the identical sequence as that found in the germline immunoglobulin gene from which the framework sequence derive, or the CDR sequences can be grafted onto framework regions that contain one or more mutations as compared to the germline sequences.
  • variable region modification is to mutate amino acid residues within the VH and/or VL CDR1, CDR2, and/or CDR3 regions to thereby improve one or more binding properties (e.g., affinity) of the antibody of interest, known as “affinity maturation.”
  • Site-directed mutagenesis or PCR-mediated mutagenesis can be performed to introduce the mutation(s) and the effect on antibody binding, or other functional property of interest, can be evaluated in in vitro or in vivo assays as described herein and provided in the Examples and/or alternative or additional assays known in the art.
  • Conservative modifications can be introduced.
  • the mutations may be amino acid substitutions, additions or deletions. Moreover, typically no more than one, two, three, four or five residues within a CDR region are altered.
  • Engineered antibodies or antibody fragments of the invention include those in which modifications have been made to framework residues within VH and/or VL, e.g. to improve the properties of the antibody. Typically such framework modifications are made to decrease the immunogenicity of the antibody.
  • one approach is to “backmutate” one or more framework residues to the corresponding germline sequence. More specifically, an antibody that has undergone somatic mutation may contain framework residues that differ from the germline sequence from which the antibody is derived. Such residues can be identified by comparing the antibody framework sequences to the germline sequences from which the antibody is derived. To return the framework region sequences to their germline configuration, the somatic mutations can be “backmutated” to the germline sequence by, for example, site-directed mutagenesis. Such “backmutated” antibodies are also intended to be encompassed by the invention.
  • Another type of framework modification involves mutating one or more residues within the framework region, or even within one or more CDR regions, to remove T cell epitopes to thereby reduce the potential immunogenicity of the antibody. This approach is also referred to as “deimmunization” and is described in further detail in U.S. Patent Publication No. 20030153043 by Carr et al.
  • the constant regions of the anti-GITR antibodies or antibody fragments can be any type or subtype, as appropriate, and can be selected to be from the species of the subject to be treated by the present methods (e.g., human, non-human primate or other mammal, for example, agricultural mammal (e.g., equine, ovine, bovine, porcine, camelid), domestic mammal (e.g., canine, feline) or rodent (e.g., rat, mouse, hamster, rabbit).
  • the anti-GITR antibodies are engineered to generate humanized or Humaneered® antibodies.
  • the constant region isotype is IgG, for example, IgG1, IgG2, IgG3, IgG4. In certain embodiments the constant region isotype is IgG 1 .
  • antibodies or antibody fragments of the invention may be engineered to include modifications within the Fc region, typically to alter one or more functional properties of the antibody, such as serum half-life, complement fixation, Fc receptor binding, and/or antigen-dependent cellular cytotoxicity.
  • modifications within the Fc region typically to alter one or more functional properties of the antibody, such as serum half-life, complement fixation, Fc receptor binding, and/or antigen-dependent cellular cytotoxicity.
  • an antibody or antibody fragment of the invention may be chemically modified (e.g., one or more chemical moieties can be attached to the antibody) or be modified to alter its glycosylation, again to alter one or more functional properties of the antibody or antibody fragment.
  • the hinge region of CH1 is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased.
  • This approach is described further in U.S. Pat. No. 5,677,425 by Bodmer et al.
  • the number of cysteine residues in the hinge region of CH1 is altered to, for example, facilitate assembly of the light and heavy chains or to increase or decrease the stability of the antibody or antibody fragment.
  • the Fc hinge region of an antibody is mutated to alter the biological half-life of the antibody. More specifically, one or more amino acid mutations are introduced into the CH2-CH3 domain interface region of the Fc-hinge fragment such that the antibody has impaired Staphylococcyl protein A (SpA) binding relative to native Fc-hinge domain SpA binding.
  • SpA Staphylococcyl protein A
  • the antibody is modified to increase its biological half-life.
  • Various approaches are possible. For example, one or more of the following mutations can be introduced: T252L, T254S, T256F, as described in U.S. Pat. No. 6,277,375 to Ward.
  • the antibody can be altered within the CH1 or CL region to contain a salvage receptor binding epitope taken from two loops of a CH2 domain of an Fc region of an IgG, as described in U.S. Pat. Nos. 5,869,046 and 6,121,022 by Presta et al.
  • the Fc region is altered by replacing at least one amino acid residue with a different amino acid residue to alter the effector functions of the antibody.
  • one or more amino acids can be replaced with a different amino acid residue such that the antibody has an altered affinity for an effector ligand but retains the antigen-binding ability of the parent antibody.
  • the effector ligand to which affinity is altered can be, for example, an Fc receptor (FcR) or the C1 component of complement. This approach is described in further detail in U.S. Pat. Nos. 5,624,821 and 5,648,260, both by Winter et al.
  • one or more amino acids selected from amino acid residues can be replaced with a different amino acid residue such that the antibody has altered C1q binding and/or reduced or abolished complement dependent cytotoxicity (CDC).
  • CDC complement dependent cytotoxicity
  • Antibodies containing such mutations mediate reduced or no antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC).
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxicity
  • amino acid residues L234 and L235 of the IgG1 constant region are substituted to Ala234 and Ala235.
  • amino acid residue N267 of the IgG1 constant region is substituted to Ala267.
  • one or more amino acid residues are altered to thereby alter the ability of the antibody to fix complement. This approach is described further in PCT Publication WO 94/29351 by Bodmer et al.
  • the Fc region is modified to increase the ability of the antibody to mediate antibody dependent cellular cytotoxicity (ADCC) and/or to increase the affinity of the antibody for an Fc ⁇ receptor by modifying one or more amino acids.
  • ADCC antibody dependent cellular cytotoxicity
  • This approach is described further in PCT Publication WO 00/42072 by Presta.
  • the binding sites on human IgG1 for Fc ⁇ R1, Fc ⁇ RII, Fc ⁇ RIII and FcRn have been mapped and variants with improved binding have been described (see Shields, R. L. et al., 2001 J. Biol. Chen. 276:6591-6604).
  • glycosylation of an antibody is modified.
  • an aglycoslated antibody can be made (i.e., the antibody lacks glycosylation).
  • Glycosylation can be altered to, for example, increase the affinity of the antibody for “antigen’.
  • Such carbohydrate modifications can be accomplished by, for example, altering one or more sites of glycosylation within the antibody sequence.
  • one or more amino acid substitutions can be made that result in elimination of one or more variable region framework glycosylation sites to thereby eliminate glycosylation at that site.
  • Such aglycosylation may increase the affinity of the antibody for antigen.
  • an antibody can be made that has an altered type of glycosylation, such as a hypofucosylated antibody having reduced amounts of fucosyl residues or an antibody having increased bisecting GlcNac structures.
  • altered glycosylation patterns have been demonstrated to increase the ADCC ability of antibodies.
  • carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies of the invention to thereby produce an antibody with altered glycosylation.
  • glycoprotein-modifying glycosyl transferases e.g., beta(1,4)-N acetylglucosaminyltransferase III (GnTIII)
  • GnTIII glycoprotein-modifying glycosyl transferases
  • antibody/immunoglobulin frameworks or scaffolds can be employed so long as the resulting polypeptide includes at least one binding region which specifically binds to GITR.
  • Such frameworks or scaffolds include the 5 main idiotypes of human immunoglobulins, or fragments thereof, and include immunoglobulins of other animal species, preferably having humanized aspects. Single heavy-chain antibodies such as those identified in camelids are of particular interest in this regard. Novel frameworks, scaffolds and fragments continue to be discovered and developed by those skilled in the art.
  • the invention pertains to generating non-immunoglobulin based antibodies using non-immunoglobulin scaffolds onto which CDRs of the invention can be grafted.
  • Known or future non-immunoglobulin frameworks and scaffolds may be employed, as long as they comprise a binding region specific for the target GITR protein (e.g., human and/or cynomolgus GITR).
  • Non-immunoglobulin frameworks or scaffolds include, but are not limited to, fibronectin (Compound Therapeutics, Inc., Waltham, Mass.), ankyrin (Molecular Partners AG, Zurich, Switzerland), domain antibodies (Domantis, Ltd., Cambridge, Mass., and Ablynx nv, Zwijnaarde, Belgium), lipocalin (Pieris Proteolab AG, Freising, Germany), small modular immuno-pharmaceuticals (Trubion Pharmaceuticals Inc., Seattle, Wash.), maxybodies (Avidia, Inc., Mountain View, Calif.), Protein A (Affibody AG, Sweden), and affilin (gamma-crystallin or ubiquitin) (Scil Proteins GmbH, Halle, Germany).
  • fibronectin Compound Therapeutics, Inc., Waltham, Mass.
  • ankyrin Molecular Partners AG, Zurich, Switzerland
  • domain antibodies Domantis, Ltd., Cambridge, Mass., and Ablynx nv
  • the fibronectin scaffolds are based on fibronectin type III domain (e.g., the tenth module of the fibronectin type III (10 Fn3 domain)).
  • the fibronectin type III domain has 7 or 8 beta strands which are distributed between two beta sheets, which themselves pack against each other to form the core of the protein, and further containing loops (analogous to CDRs) which connect the beta strands to each other and are solvent exposed. There are at least three such loops at each edge of the beta sheet sandwich, where the edge is the boundary of the protein perpendicular to the direction of the beta strands (see U.S. Pat. No. 6,818,418).
  • fibronectin-based scaffolds are not an immunoglobulin, although the overall fold is closely related to that of the smallest functional antibody fragment, the variable region of the heavy chain, which comprises the entire antigen recognition unit in camel and llama IgG. Because of this structure, the non-immunoglobulin antibody mimics antigen binding properties that are similar in nature and affinity to those of antibodies.
  • These scaffolds can be used in a loop randomization and shuffling strategy in vitro that is similar to the process of affinity maturation of antibodies in vivo.
  • These fibronectin-based molecules can be used as scaffolds where the loop regions of the molecule can be replaced with CDRs of the invention using standard cloning techniques.
  • the ankyrin technology is based on using proteins with ankyrin derived repeat modules as scaffolds for bearing variable regions which can be used for binding to different targets.
  • the ankyrin repeat module is a 33 amino acid polypeptide consisting of two anti-parallel ⁇ -helices and a ⁇ -turn. Binding of the variable regions is mostly optimized by using ribosome display.
  • Avimers are derived from natural A-domain containing protein such as LRP-1. These domains are used by nature for protein-protein interactions and in human over 250 proteins are structurally based on A-domains. Avimers consist of a number of different “A-domain” monomers (2-10) linked via amino acid linkers. Avimers can be created that can bind to the target antigen using the methodology described in, for example, U.S. Patent Application Publication Nos. 20040175756; 20050053973; 20050048512; and 20060008844.
  • Affibody affinity ligands are small, simple proteins composed of a three-helix bundle based on the scaffold of one of the IgG-binding domains of Protein A.
  • Protein A is a surface protein from the bacterium Staphylococcus aureus .
  • This scaffold domain consists of 58 amino acids, 13 of which are randomized to generate affibody libraries with a large number of ligand variants (See e.g., U.S. Pat. No. 5,831,012).
  • Affibody molecules mimic antibodies, they have a molecular weight of 6 kDa, compared to the molecular weight of antibodies, which is 150 kDa. In spite of its small size, the binding site of affibody molecules is similar to that of an antibody.
  • Anticalins are products developed by the company Pieris ProteoLab AG. They are derived from lipocalins, a widespread group of small and robust proteins that are usually involved in the physiological transport or storage of chemically sensitive or insoluble compounds. Several natural lipocalins occur in human tissues or body liquids. The protein architecture is reminiscent of immunoglobulins, with hypervariable loops on top of a rigid framework. However, in contrast with antibodies or their recombinant fragments, lipocalins are composed of a single polypeptide chain with 160 to 180 amino acid residues, being just marginally bigger than a single immunoglobulin domain. The set of four loops, which makes up the binding pocket, shows pronounced structural plasticity and tolerates a variety of side chains.
  • the binding site can thus be reshaped in a proprietary process in order to recognize prescribed target molecules of different shape with high affinity and specificity.
  • One protein of lipocalin family the bilin-binding protein (BBP) of Pieris Bras sicae has been used to develop anticalins by mutagenizing the set of four loops.
  • BBP bilin-binding protein
  • One example of a patent application describing anticalins is in PCT Publication No. WO 199916873.
  • Affilin molecules are small non-immunoglobulin proteins which are designed for specific affinities towards proteins and small molecules.
  • New affilin molecules can be very quickly selected from two libraries, each of which is based on a different human derived scaffold protein. Affilin molecules do not show any structural homology to immunoglobulin proteins.
  • two affilin scaffolds are employed, one of which is gamma crystalline, a human structural eye lens protein and the other is “ubiquitin” superfamily proteins. Both human scaffolds are very small, show high temperature stability and are almost resistant to pH changes and denaturing agents. This high stability is mainly due to the expanded beta sheet structure of the proteins. Examples of gamma crystalline derived proteins are described in WO200104144 and examples of “ubiquitin-like” proteins are described in WO2004106368.
  • PEM Protein epitope mimetics
  • the present invention provides engineered human antibodies that specifically bind to GITR protein (e.g., human GITR). Compared to the chimeric, primatized, or humanized antibodies, the human GITR-binding antibodies of the invention have further reduced antigenicity when administered to human subjects.
  • GITR protein e.g., human GITR
  • the human GITR-binding antibodies can be generated using methods that are known in the art.
  • the Humaneered® technology platform KeroBios, Sout San Francisco, Calif.
  • U.S. Patent Publication No. 20050008625 describes an in vivo method for replacing a nonhuman antibody variable region with a human variable region in an antibody while maintaining the same or providing better binding characteristics relative to that of the nonhuman antibody.
  • the method relies on epitope guided replacement of variable regions of a non-human reference antibody with a fully human antibody.
  • the resulting human antibody is generally unrelated structurally to the reference nonhuman antibody, but binds to the same epitope on the same antigen as the reference antibody.
  • the anti-GITR antibodies of the invention are based on engineered human antibodies with V-region sequences having substantial amino acid sequence identity to human germline V region sequences while retaining the specificity and affinity of a reference antibody. See, U.S. Patent Publication No. 2005/0255552 and U.S. Patent Publication No. 2006/0134098, both of which are hereby incorporated herein by reference.
  • the process of improvement identifies minimal sequence information required to determine antigen-binding specificity from the variable region of a reference antibody, and transfers that information to a library of human partial V-region gene sequences to generate an epitope-focused library of human antibody V regions.
  • a microbial-based secretion system can be used to express members of the library as antibody Fab fragments and the library is screened for antigen-binding Fabs, for example, using a colony-lift binding assay. See, e.g., U.S. Patent Publication No. 2007/0020685. Positive clones can be further characterized to identify those with the highest affinity.
  • the resultant engineered human Fabs retain the binding specificity of the parent, reference anti-GITR antibody, typically have equivalent or higher affinity for antigen in comparison to the parent antibody, and have V-regions with a high degree of sequence identity compared with human germ-line antibody V-regions.
  • the minimum binding specificity determinant (BSD) required to generate the epitope-focused library is typically represented by a sequence within the heavy chain CDR3 (“CDRH3”) and a sequence within the light chain of CDR3 (“CDRL3”).
  • the BSD can comprise a portion or the entire length of a CDR3.
  • the BSD can be comprised of contiguous or non-contiguous amino acid residues.
  • the epitope-focused library is constructed from human V-segment sequences linked to the unique CDR3-FR4 region from the reference antibody containing the BSD and human germ-line J segment sequences (see, U.S. Patent Publication No. 2005/0255552).
  • the human V segment libraries can be generated by sequential cassette replacement in which only part of the reference antibody V segment is initially replaced by a library of human sequences.
  • the identified human “cassettes” supporting binding in the context of residual reference antibody amino acid sequences are then recombined in a second library screen to generate completely human V segments (see, U.S. Patent Publication No. 2006/0134098).
  • paired heavy and light chain CDR3 segments, CDR3-FR4 segments, or J segments, containing specificity determinants from the reference antibody are used to constrain the binding specificity so that antigen-binders obtained from the library retain the epitope-specificity of the reference antibody. Additional maturational changes can be introduced in the CDR3 regions of each chain during the library construction in order to identify antibodies with optimal binding kinetics.
  • the resulting engineered human antibodies have V-segment sequences derived from the human germ-line libraries, retain the short BSD sequence from within the CDR3 regions and have human germ-line framework 4 (FR4) regions.
  • Certain IgG antibodies from this family of mammals as found in nature lack light chains, and are thus structurally distinct from the typical four chain quaternary structure having two heavy and two light chains, for antibodies from other animals. See PCT/EP93/02214 (WO 94/04678 published 3 Mar. 1994).
  • a region of the camelid antibody which is the small single variable domain identified as VHH can be obtained by genetic engineering to yield a small protein having high affinity for a target, resulting in a low molecular weight antibody-derived protein known as a “camelid nanobody”.
  • VHH camelid antibody
  • the camelid nanobody has a molecular weight approximately one-tenth that of a human IgG molecule, and the protein has a physical diameter of only a few nanometers.
  • One consequence of the small size is the ability of camelid nanobodies to bind to antigenic sites that are functionally invisible to larger antibody proteins, i.e., camelid nanobodies are useful as reagents detect antigens that are otherwise cryptic using classical immunological techniques, and as possible therapeutic agents.
  • a camelid nanobody can inhibit as a result of binding to a specific site in a groove or narrow cleft of a target protein, and hence can serve in a capacity that more closely resembles the function of a classical low molecular weight drug than that of a classical antibody.
  • camelid nanobodies being extremely thermostable, stable to extreme pH and to proteolytic digestion, and poorly antigenic. Another consequence is that camelid nanobodies readily move from the circulatory system into tissues, and even cross the blood-brain barrier and can treat disorders that affect nervous tissue. Nanobodies can further facilitated drug transport across the blood brain barrier. See U.S. patent application 20040161738 published Aug. 19, 2004. These features combined with the low antigenicity to humans indicate great therapeutic potential. Further, these molecules can be expressed in prokaryotic cells such as E. coli and are expressed as fusion proteins with bacteriophage and are functional.
  • a feature of the present invention is a camelid antibody or nanobody having high affinity for GITR.
  • the camelid antibody or nanobody is naturally produced in the camelid animal, i.e., is produced by the camelid following immunization with GITR or a peptide fragment thereof, using techniques described herein for other antibodies.
  • the GITR-binding camelid nanobody is engineered, i.e., produced by selection for example from a library of phage displaying appropriately mutagenized camelid nanobody proteins using panning procedures with GITR as a target as described in the examples herein.
  • Engineered nanobodies can further be customized by genetic engineering to have a half life in a recipient subject of from 45 minutes to two weeks.
  • the camelid antibody or nanobody is obtained by grafting the CDRs sequences of the heavy or light chain of the human antibodies of the invention into nanobody or single domain antibody framework sequences, as described for example in PCT/EP93/02214.
  • the present invention provides multivalent camelid antibody or nanobody, according the methods described below.
  • an antibody molecule is a multispecific antibody molecule, e.g., it comprises a plurality of immunoglobulin variable domains sequences, wherein a first immunoglobulin variable domain sequence of the plurality has binding specificity for a first epitope and a second immunoglobulin variable domain sequence of the plurality has binding specificity for a second epitope.
  • the first and second epitopes are on the same antigen, e.g., the same protein (or subunit of a multimeric protein).
  • the first and second epitopes overlap.
  • the first and second epitopes do not overlap.
  • the first and second epitopes are on different antigens, e.g., the different proteins (or different subunits of a multimeric protein).
  • An antibody of the invention can be derivatized or linked to another functional molecule, e.g., another peptide or protein (e.g., another antibody or ligand for a receptor) to generate a multivalent molecule that binds to at least two different binding sites (which may be the same or different target sites or molecules).
  • another functional molecule e.g., another peptide or protein (e.g., another antibody or ligand for a receptor) to generate a multivalent molecule that binds to at least two different binding sites (which may be the same or different target sites or molecules).
  • a “derivatized” antibody molecule is one that has been modified. Methods of derivatization include but are not limited to the addition of a fluorescent moiety, a radionucleotide, a toxin, an enzyme or an affinity ligand such as biotin.
  • an antibody molecule can be functionally linked (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate association of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • another antibody e.g., a bispecific antibody or a diabody
  • detectable agent e.g., a detectable agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate association of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • an antibody of the invention e.g., a monospecific, bispecific, or multispecific antibody molecule
  • another partner e.g., a protein e.g., one, two or more cytokines, e.g., as a fusion molecule for example a fusion protein.
  • the fusion molecule comprises one or more proteins, e.g., one, two or more cytokines.
  • the antibody of the invention is derivatized or functionally linked (e.g., by chemical coupling, genetic fusion, noncovalent association or otherwise) to more than one other functional molecule to generate multivalent molecules that bind to two or more different binding sites which are the same or different binding sites on the same target molecule.
  • the multivalent binding sites are the same.
  • the antibody of the invention is derivatized or linked to more than one other functional molecule to generate multi-specific molecules that bind two or more different binding sites on at least two target molecules; such multi-specific molecules are also intended to be encompassed by the term “bispecific molecule” or “multispecific” as used herein.
  • an antibody of the invention can be functionally linked (e.g., by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other binding molecules, such as another antibody, antibody fragment, peptide or binding mimetic, such that a multivalent molecule results.
  • the present invention includes bispecific molecules comprising at least one first binding specificity for GITR and a second binding specificity for a second target epitope.
  • the second target epitope is another epitope of GITR different from the first target epitope.
  • the molecule further includes a third binding specificity, in addition to the first and second target epitope.
  • a multispecific antibody molecule comprises a third, fourth or fifth immunoglobulin variable domain.
  • a multispecific antibody molecule is a bispecific antibody molecule, a trispecific antibody molecule, or tetraspecific antibody molecule.
  • the bispecific molecules of the invention comprise as a binding specificity at least one antibody, or an antibody fragment thereof, including, e.g., an Fab, Fab′, F(ab′)2, Fv, or a single chain Fv.
  • the antibody may also be a light chain or heavy chain dimer, or any minimal fragment thereof such as a Fv or a single chain construct as described in Ladner et al. U.S. Pat. No. 4,946,778.
  • Diabodies are bivalent, bispecific molecules in which VH and VL domains are expressed on a single polypeptide chain, connected by a linker that is too short to allow for pairing between the two domains on the same chain.
  • the VH and VL domains pair with complementary domains of another chain, thereby creating two antigen binding sites (see e.g., Holliger et al., 1993 Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak et al., 1994 Structure 2:1121-1123).
  • Diabodies can be produced by expressing two polypeptide chains with either the structure VHA-VLB and VHB-VLA (VH-VL configuration), or VLA-VHB and VLB-VHA (VL-VH configuration) within the same cell. Most of them can be expressed in soluble form in bacteria.
  • Single chain diabodies (scDb) are produced by connecting the two diabody-forming polypeptide chains with linker of approximately 15 amino acid residues (see Holliger and Winter, 1997 Cancer Immunol. Immunother., 45(3-4):128-30; Wu et al., 1996 Immunotechnology, 2(1):21-36).
  • scDb can be expressed in bacteria in soluble, active monomeric form (see Holliger and Winter, 1997 Cancer Immunol.
  • a diabody can be fused to Fc to generate a “di-diabody” (see Lu et al., 2004 J. Biol. Chem., 279(4):2856-65).
  • antibodies which can be employed in the bispecific molecules of the invention are murine, chimeric and humanized monoclonal antibodies.
  • the bispecific and/or multivalent molecules of the present invention can be prepared by conjugating the constituent binding specificities, using methods known in the art. For example, each binding specificity of the bispecific and/or multivalent molecule can be generated separately and then conjugated to one another. When the binding specificities are proteins or peptides, a variety of coupling or cross-linking agents can be used for covalent conjugation.
  • cross-linking agents examples include protein A, carbodiimide, N-succinimidyl-S-acetyl-thioacetate (SATA), 5,5′-dithiobis(2-nitrobenzoic acid) (DTNB), o-phenylenedimaleimide (oPDM), N-succinimidyl-3-(2-pyridyldithio)propionate (SPDP), and sulfosuccinimidyl 4-(N-maleimidomethyl) cyclohaxane-1-carboxylate (sulfo-SMCC) (see e.g., Karpovsky et al., 1984 J. Exp. Med.
  • Conjugating agents are SATA and sulfo-SMCC, both available from Pierce Chemical Co. (Rockford, Ill.).
  • binding specificities are antibodies, they can be conjugated by sulfhydryl bonding of the constant domain hinge regions of the two heavy chains.
  • the hinge region is modified to contain an odd number of sulfhydryl residues, for example one, prior to conjugation.
  • binding specificities can be encoded in the same vector and expressed and assembled in the same host cell.
  • This method is particularly useful where the bispecific and/or multivalent molecule is a mAb ⁇ mAb, mAb ⁇ Fab, Fab ⁇ F(ab′)2 or ligand x Fab fusion protein.
  • a bispecific and/or multivalent molecule of the invention can be a single chain molecule comprising one single chain antibody and a binding determinant, or a single chain bispecific molecule comprising two binding determinants.
  • Bispecific molecules may comprise at least two single chain molecules. Methods for preparing bispecific molecules are described for example in U.S. Pat. No. 5,260,203; U.S. Pat. No. 5,455,030; U.S. Pat. No.
  • Binding of bispecific and/or multivalent molecules to their specific targets can be confirmed by, for example, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (REA), FACS analysis, bioassay (e.g., growth inhibition), or Western Blot assay.
  • ELISA enzyme-linked immunosorbent assay
  • REA radioimmunoassay
  • FACS analysis FACS analysis
  • bioassay e.g., growth inhibition
  • Western Blot assay Western Blot assay.
  • Each of these assays generally detects the presence of protein-antibody complexes of particular interest by employing a labeled reagent (e.g., an antibody) specific for the complex of interest.
  • the present invention provides for antibodies and antibody fragments that specifically bind to GITR protein which have an extended half-life in vivo.
  • kidney filtration kidney filtration, metabolism in the liver, degradation by proteolytic enzymes (proteases), and immunogenic responses (e.g., protein neutralization by antibodies and uptake by macrophages and dentritic cells).
  • proteolytic enzymes proteolytic enzymes
  • immunogenic responses e.g., protein neutralization by antibodies and uptake by macrophages and dentritic cells.
  • a variety of strategies can be used to extend the half life of the antibodies of the present invention.
  • polyethyleneglycol PEG
  • PEG polyethyleneglycol
  • PSA polysialic acid
  • HES hydroxyethyl starch
  • albumin-binding ligands and carbohydrate shields
  • genetic fusion to proteins binding to serum proteins such as albumin, IgG, FcRn, and transferring
  • other binding moieties that bind to serum proteins, such as nanobodies, Fabs, DARPins, avimers, affibodies, and anticalins
  • genetic fusion to rPEG, albumin, domain of albumin, albumin-binding proteins, and Fc or by incorporation into nancarriers, slow release formulations, or medical devices.
  • inert polymer molecules such as high molecular weight PEG can be attached to the antibodies or a fragment thereof with or without a multifunctional linker either through site-specific conjugation of the PEG to the N- or C-terminus of the antibodies or via epsilon-amino groups present on lysine residues.
  • PEG polyethylene glycol
  • the antibody, or fragment thereof typically is reacted with polyethylene glycol (PEG), such as a reactive ester or aldehyde derivative of PEG, under conditions in which one or more PEG groups become attached to the antibody or antibody fragment.
  • the pegylation can be carried out by an acylation reaction or an alkylation reaction with a reactive PEG molecule (or an analogous reactive water-soluble polymer).
  • a reactive PEG molecule or an analogous reactive water-soluble polymer.
  • polyethylene glycol is intended to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (C1-C10) alkoxy- or aryloxy-polyethylene glycol or polyethylene glycol-maleimide.
  • the antibody to be pegylated is an aglycosylated antibody. Linear or branched polymer derivatization that results in minimal loss of biological activity will be used.
  • the degree of conjugation can be closely monitored by SDS-PAGE and mass spectrometry to ensure proper conjugation of PEG molecules to the antibodies. Unreacted PEG can be separated from antibody-PEG conjugates by size-exclusion or by ion-exchange chromatography. PEG-derivatized antibodies can be tested for binding activity as well as for in vivo efficacy using methods well-known to those of skill in the art, for example, by immunoassays described herein. Methods for pegylating proteins are known in the art and can be applied to the antibodies of the invention. See for example, EP 0 154 316 by Nishimura et al. and EP 0 401 384 by Ishikawa et al.
  • modified pegylation technologies include reconstituting chemically orthogonal directed engineering technology (ReCODE PEG), which incorporates chemically specified side chains into biosynthetic proteins via a reconstituted system that includes tRNA synthetase and tRNA.
  • ReCODE PEG chemically orthogonal directed engineering technology
  • This technology enables incorporation of more than 30 new amino acids into biosynthetic proteins in E. coli , yeast, and mammalian cells.
  • the tRNA incorporates a nonnative amino acid any place an amber codon is positioned, converting the amber from a stop codon to one that signals incorporation of the chemically specified amino acid.
  • Recombinant pegylation technology can also be used for serum halflife extension.
  • This technology involves genetically fusing a 300-600 amino acid unstructured protein tail to an existing pharmaceutical protein. Because the apparent molecular weight of such an unstructured protein chain is about 15-fold larger than its actual molecular weight, the serum halflife of the protein is greatly increased. In contrast to traditional PEGylation, which requires chemical conjugation and repurification, the manufacturing process is greatly simplified and the product is homogeneous.
  • PSA polymer polysialic acid
  • PSA is a polymer of sialic acid (a sugar).
  • sialic acid a sugar
  • polysialic acid provides a protective microenvironment on conjugation. This increases the active life of the therapeutic protein in the circulation and prevents it from being recognized by the immune system.
  • the PSA polymer is naturally found in the human body. It was adopted by certain bacteria which evolved over millions of years to coat their walls with it. These naturally polysialylated bacteria were then able, by virtue of molecular mimicry, to foil the body's defence system.
  • PSA nature's ultimate stealth technology, can be easily produced from such bacteria in large quantities and with predetermined physical characteristics. Bacterial PSA is completely non-immunogenic, even when coupled to proteins, as it is chemically identical to PSA in the human body.
  • HES hydroxyethyl starch
  • Another technology include the use of hydroxyethyl starch (“HES”) derivatives linked to antibodies.
  • HES is a modified natural polymer derived from waxy maize starch and can be metabolized by the body's enzymes.
  • HES solutions are usually administered to substitute deficient blood volume and to improve the rheological properties of the blood. Hesylation of an antibody enables the prolongation of the circulation half-life by increasing the stability of the molecule, as well as by reducing renal clearance, resulting in an increased biological activity.
  • a wide range of HES antibody conjugates can be customized.
  • Antibodies having an increased half-life in vivo can also be generated introducing one or more amino acid modifications (i.e., substitutions, insertions or deletions) into an IgG constant domain, or FcRn binding fragment thereof (preferably a Fc or hinge Fc domain fragment). See, e.g., International Publication No. WO 98/23289; International Publication No. WO 97/34631; and U.S. Pat. No. 6,277,375.
  • antibodies can be conjugated to albumin in order to make the antibody or antibody fragment more stable in vivo or have a longer half life in vivo.
  • the techniques are well-known in the art, see, e.g., International Publication Nos. WO 93/15199, WO 93/15200, and WO 01/77137; and European Patent No. EP 413,622.
  • the strategies for increasing half life is especially useful in nanobodies, fibronectin-based binders, and other antibodies or proteins for which increased in vivo half life is desired.
  • the present invention provides antibodies or fragments thereof that specifically bind to a GITR protein recombinantly fused or chemically conjugated (including both covalent and non-covalent conjugations) to a heterologous protein or polypeptide (or fragment thereof, preferably to a polypeptide of at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90, or at least 100 amino acids) to generate fusion proteins.
  • the invention provides fusion proteins comprising an antigen-binding fragment of an antibody described herein (e.g., a Fab fragment, Fd fragment, Fv fragment, F(ab)2 fragment, a VH domain, a VH CDR, a VL domain, or a VL CDR) and a heterologous protein, polypeptide, or peptide.
  • an antibody described herein e.g., a Fab fragment, Fd fragment, Fv fragment, F(ab)2 fragment, a VH domain, a VH CDR, a VL domain, or a VL CDR
  • Methods for fusing or conjugating proteins, polypeptides, or peptides to an antibody or an antibody fragment are known in the art. See, e.g., U.S. Pat. Nos. 5,336,603, 5,622,929, 5,359,046, 5,349,053, 5,447,851, and 5,112,946; European Patent Nos.
  • EP 307,434 and EP 367,166 International Publication Nos. WO 96/04388 and WO 91/06570; Ashkenazi et al., 1991, Proc. Natl. Acad. Sci. USA 88: 10535-10539; Zheng et al., 1995, J. Immunol. 154:5590-5600; and Vil et al., 1992, Proc. Natl. Acad. Sci. USA 89:11337-11341.
  • DNA shuffling may be employed to alter the activities of antibodies of the invention or fragments thereof (e.g., antibodies or fragments thereof with higher affinities and lower dissociation rates). See, generally, U.S. Pat. Nos. 5,605,793, 5,811,238, 5,830,721, 5,834,252, and 5,837,458; Patten et al., 1997, Curr. Opinion Biotechnol. 8:724-33; Harayama, 1998, Trends Biotechnol.
  • Antibodies or fragments thereof, or the encoded antibodies or fragments thereof may be altered by being subjected to random mutagenesis by error-prone PCR, random nucleotide insertion or other methods prior to recombination.
  • a polynucleotide encoding an antibody or fragment thereof that specifically binds to a GITR protein may be recombined with one or more components, motifs, sections, parts, domains, fragments, etc. of one or more heterologous molecules.
  • the antibodies or fragments thereof can be fused to marker sequences, such as a peptide to facilitate purification.
  • the marker amino acid sequence is a hexa-histidine (HHHHHH SEQ ID NO:11) peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, Calif., 91311), among others, many of which are commercially available.
  • hexa-histidine SEQ ID NO:11
  • peptide tags useful for purification include, but are not limited to, the hemagglutinin (“HA”) tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al., 1984, Cell 37:767), and the “flag” tag.
  • HA hemagglutinin
  • An antibody molecules may be conjugated to another molecular entity, typically a diagnostic, detectable, or a therapeutic (e.g., a cytotoxic or cytostatic) agent or moiety. Radioactive isotopes can be used in diagnostic or therapeutic applications.
  • antibodies of the present invention or fragments thereof conjugated to a diagnostic or detectable agent can be useful for monitoring or prognosing the onset, development, progression and/or severity of a disease or disorder as part of a clinical testing procedure, such as determining the efficacy of a particular therapy.
  • Such diagnosis and detection can accomplished by coupling the antibody to detectable substances including, but not limited to, various enzymes, such as, but not limited to, horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; prosthetic groups, such as, but not limited to, streptavidinlbiotin and avidin/biotin; fluorescent materials, such as, but not limited to, umbelliferone, fluorescein, fluorescein isothiocynate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; luminescent materials, such as, but not limited to, luminol; bioluminescent materials, such as but not limited to, luciferase, luciferin, and aequorin; radioactive materials, such as, but not limited to, iodine (131I, 125I, 123I, and 121
  • the present invention further encompasses an antibody or fragment thereof conjugated to a therapeutic moiety or drug moiety that modifies a given biological effect or response and uses of antibodies or fragments thereof conjugated to a therapeutic moiety.
  • Therapeutic moieties or drug moieties are not to be construed as limited to classical chemical therapeutic agents.
  • the drug moiety may be a protein, peptide, or polypeptide possessing a desired biological activity.
  • Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, cholera toxin, or diphtheria toxin; a protein such as tumor necrosis factor, ⁇ -interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, an anti-angiogenic agent; or, a biological response modifier such as, for example, a lymphokine.
  • a toxin such as abrin, ricin A, pseudomonas exotoxin, cholera toxin, or diphtheria toxin
  • a protein such as tumor necrosis factor, ⁇ -interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, an anti-angiogenic agent
  • a biological response modifier such as, for example, a lymphokine.
  • An antibody or fragment thereof may be conjugated to a therapeutic moiety such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters.
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to cells.
  • an antibody can be conjugated to therapeutic moieties such as a radioactive metal ion, such as alph-emiters such as 213Bi or macrocyclic chelators useful for conjugating radiometal ions, including but not limited to, 131In, 131LU, 131Y, 131Ho, 131Sm, to polypeptides.
  • the macrocyclic chelator is 1,4,7,10-tetraazacyclododecane-N,N′,N′′,N′′′-tetraacetic acid (DOTA) which can be attached to the antibody via a linker molecule.
  • linker molecules include for example, glycine linkers e.g., GGGGS (SEQ ID NO:15), which may optionally be repeated, e.g., GGGGSGGGGSGGGGS (SEQ ID NO:18), or other linkers are commonly known in the art and described in Denardo et al., 1998, Clin Cancer Res. 4(10):2483-90; Peterson et al., 1999, Bioconjug. Chem. 10(4):553-7; and Zimmerman et al., 1999, Nucl. Med. Biol. 26(8):943-50, each incorporated by reference in their entireties.
  • GGGGS SEQ ID NO:15
  • GGGGSGGGGGGSGGGGS SEQ ID NO:18
  • Examples of other therapeutic agents include taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicine, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, l-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, puromycin, maytansinoids, e.g., maytansinol (see U.S. Pat. No.
  • Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, CC-1065, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclinies (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (
  • antimetabolites e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil de
  • Antibodies may also be attached to solid supports, which are particularly useful for immunoassays or purification of the target antigen.
  • solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
  • Anti-GITR antibodies, antigen binding molecules, and fragments thereof can be produced by any means known in the art, including but not limited to, recombinant expression, chemical synthesis, and enzymatic digestion of antibody tetramers, whereas full-length monoclonal antibodies can be obtained by, e.g., hybridoma or recombinant production.
  • Recombinant expression can be from any appropriate host cells known in the art, for example, mammalian host cells, bacterial host cells, yeast host cells, insect host cells, etc.
  • the invention further provides polynucleotides encoding the antibodies described herein, e.g., polynucleotides encoding heavy or light chain variable regions or segments comprising the complementary determining regions as described herein.
  • the polynucleotide encoding the heavy chain variable regions comprises a sequence having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide selected from the group consisting of SEQ ID NO:51, SEQ ID NO:53, SEQ ID NO:55, SEQ ID NO:56, SEQ ID NO:57, SEQ ID NO:101, and SEQ ID NO:107.
  • the polynucleotide encoding the light chain variable regions comprises a sequence having at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide selected from the group consisting of SEQ ID NO:52, SEQ ID NO:54, and SEQ ID NO:102.
  • the polynucleotide encoding the heavy chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:67. In some embodiments, the polynucleotide encoding the light chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:68.
  • the polynucleotide encoding the heavy chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:72.
  • the polynucleotide encoding the light chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide selected of SEQ ID NO:73.
  • the polynucleotide encoding the heavy chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:74. In some embodiments, the polynucleotide encoding the light chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:68.
  • the polynucleotide encoding the heavy chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:76. In some embodiments, the polynucleotide encoding the light chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:68.
  • the polynucleotide encoding the heavy chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:78. In some embodiments, the polynucleotide encoding the light chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:68.
  • the polynucleotide encoding the heavy chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide selected from the group consisting of SEQ ID NO:103.
  • the polynucleotide encoding the light chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:104.
  • the polynucleotide encoding the heavy chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:108.
  • the polynucleotide encoding the light chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:104.
  • the polynucleotide encoding the heavy chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:60. In some embodiments, the polynucleotide encoding the light chain has at least 85%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% nucleic acid sequence identity with a polynucleotide of SEQ ID NO:58.
  • the polynucleotides of the invention can encode only the variable region sequence of an anti-GITR antibody. They can also encode both a variable region and a constant region of the antibody. Some of the polynucleotide sequences encode a polypeptide that comprises variable regions of both the heavy chain and the light chain of one of the exemplified mouse anti-GITR antibody. Some other polynucleotides encode two polypeptide segments that respectively are substantially identical to the variable regions of the heavy chain and the light chain of one of the mouse antibodies.
  • the polynucleotide sequences can be produced by de novo solid-phase DNA synthesis or by PCR mutagenesis of an existing sequence (e.g., sequences as described herein) encoding an anti-GITR antibody or its binding fragment.
  • Direct chemical synthesis of nucleic acids can be accomplished by methods known in the art, such as the phosphotriester method of Narang et al., Meth. Enzymol. 68:90, 1979; the phosphodiester method of Brown et al., Meth. Enzymol. 68:109, 1979; the diethylphosphoramidite method of Beaucage et al., Tetra. Lett., 22:1859, 1981; and the solid support method of U.S. Pat. No.
  • Nonviral vectors and systems include plasmids, episomal vectors, typically with an expression cassette for expressing a protein or RNA, and human artificial chromosomes (see, e.g., Harrington et al., Nat Genet 15:345, 1997).
  • nonviral vectors useful for expression of the anti-GITR polynucleotides and polypeptides in mammalian (e.g., human) cells include pThioHis A, B & C, pcDNA3.1/His, pEBVHis A, B & C (Invitrogen, San Diego, Calif.), MPSV vectors, and numerous other vectors known in the art for expressing other proteins.
  • Useful viral vectors include vectors based on retroviruses, adenoviruses, adenoassociated viruses, herpes viruses, vectors based on SV40, papilloma virus, HBP Epstein Barr virus, vaccinia virus vectors and Semliki Forest virus (SFV). See, Brent et al., supra; Smith, Annu. Rev. Microbiol. 49:807, 1995; and Rosenfeld et al., Cell 68:143, 1992.
  • expression vector depends on the intended host cells in which the vector is to be expressed.
  • the expression vectors contain a promoter and other regulatory sequences (e.g., enhancers) that are operably linked to the polynucleotides encoding an anti-GITR antibody chain or fragment.
  • an inducible promoter is employed to prevent expression of inserted sequences except under inducing conditions.
  • Inducible promoters include, e.g., arabinose, lacZ, metallothionein promoter or a heat shock promoter. Cultures of transformed organisms can be expanded under noninducing conditions without biasing the population for coding sequences whose expression products are better tolerated by the host cells.
  • promoters In addition to promoters, other regulatory elements may also be required or desired for efficient expression of an anti-GITR antibody chain or fragment. These elements typically include an ATG initiation codon and adjacent ribosome binding site or other sequences. In addition, the efficiency of expression may be enhanced by the inclusion of enhancers appropriate to the cell system in use (see, e.g., Scharf et al., Results Probl. Cell Differ. 20:125, 1994; and Bittner et al., Meth. Enzymol., 153:516, 1987). For example, the SV40 enhancer or CMV enhancer may be used to increase expression in mammalian host cells.
  • Expression vectors may also provide a secretion signal sequence position to form a fusion protein with polypeptides encoded by inserted anti-GITR antibody sequences. More often, the inserted anti-GITR antibody sequences are linked to a signal sequences before inclusion in the vector.
  • Vectors to be used to receive sequences encoding anti-GITR antibody light and heavy chain variable domains sometimes also encode constant regions or parts thereof. Such vectors allow expression of the variable regions as fusion proteins with the constant regions thereby leading to production of intact antibodies or fragments thereof. Typically, such constant regions are human.
  • Host cells for harboring and expressing the anti-GITR antibody chains can be either prokaryotic or eukaryotic.
  • E. coli is one prokaryotic host useful for cloning and expressing the polynucleotides of the present invention.
  • Other microbial hosts suitable for use include bacilli, such as Bacillus subtilis , and other enterobacteriaceae, such as Salmonella, Serratia , and various Pseudomonas species.
  • prokaryotic hosts one can also make expression vectors, which typically contain expression control sequences compatible with the host cell (e.g., an origin of replication).
  • any number of a variety of well-known promoters will be present, such as the lactose promoter system, a tryptophan (trp) promoter system, a beta-lactamase promoter system, or a promoter system from phage lambda.
  • the promoters typically control expression, optionally with an operator sequence, and have ribosome binding site sequences and the like, for initiating and completing transcription and translation.
  • Other microbes, such as yeast can also be employed to express anti-GITR polypeptides of the invention. Insect cells in combination with baculovirus vectors can also be used.
  • mammalian host cells are used to express and produce the anti-GITR polypeptides of the present invention.
  • they can be either a hybridoma cell line expressing endogenous immunoglobulin genes (e.g., the myeloma hybridoma clones as described in the Examples) or a mammalian cell line harboring an exogenous expression vector (e.g., the SP2/0 myeloma cells exemplified below). These include any normal mortal or normal or abnormal immortal animal or human cell.
  • a number of suitable host cell lines capable of secreting intact immunoglobulins have been developed, including the CHO cell lines, various Cos cell lines, HeLa cells, myeloma cell lines, transformed B-cells and hybridomas.
  • the use of mammalian tissue cell culture to express polypeptides is discussed generally in, e.g., Winnacker, From Genes to Clones, VCH Publishers, N.Y., N.Y., 1987.
  • Expression vectors for mammalian host cells can include expression control sequences, such as an origin of replication, a promoter, and an enhancer (see, e.g., Queen et al., Immunol. Rev.
  • expression vectors usually contain promoters derived from mammalian genes or from mammalian viruses. Suitable promoters may be constitutive, cell type-specific, stage-specific, and/or modulatable or regulatable.
  • Useful promoters include, but are not limited to, the metallothionein promoter, the constitutive adenovirus major late promoter, the dexamethasone-inducible MMTV promoter, the SV40 promoter, the MRP polIII promoter, the constitutive MPSV promoter, the tetracycline-inducible CMV promoter (such as the human immediate-early CMV promoter), the constitutive CMV promoter, and promoter-enhancer combinations known in the art.
  • Methods for introducing expression vectors containing the polynucleotide sequences of interest vary depending on the type of cellular host. For example, calcium chloride transfection is commonly utilized for prokaryotic cells, whereas calcium phosphate treatment or electroporation may be used for other cellular hosts (see generally Sambrook et al., supra).
  • Other methods include, e.g., electroporation, calcium phosphate treatment, liposome-mediated transformation, injection and microinjection, ballistic methods, virosomes, immunoliposomes, polycation:nucleic acid conjugates, naked DNA, artificial virions, fusion to the herpes virus structural protein VP22 (Elliot and O'Hare, Cell 88:223, 1997), agent-enhanced uptake of DNA, and ex vivo transduction. For long-term, high-yield production of recombinant proteins, stable expression will often be desired.
  • cell lines which stably express anti-GITR antibody chains or binding fragments can be prepared using expression vectors of the invention which contain viral origins of replication or endogenous expression elements and a selectable marker gene. Following introduction of the vector, cells may be allowed to grow for 1-2 days in an enriched media before they are switched to selective media.
  • the purpose of the selectable marker is to confer resistance to selection, and its presence allows growth of cells which successfully express the introduced sequences in selective media. Resistant, stably transfected cells can be proliferated using tissue culture techniques appropriate to the cell type.
  • Agonist anti-GITR antibodies bind to GITR and promote, induce, stimulate intracellular signaling through GITR.
  • Binding of the anti-GITR antibodies to GITR can be determined using any method known in the art. For example, binding to GITR can be determined using known techniques, including without limitation ELISA, Western blots, surface plasmon resonance (e.g., BIAcore), and flow cytometry.
  • binding to GITR can be determined using known techniques, including without limitation ELISA, Western blots, surface plasmon resonance (e.g., BIAcore), and flow cytometry.
  • Intracellular signaling through GITR can be measured using any method known in the art.
  • activation through GITR promotes NF ⁇ B and MAPK signaling.
  • Methods for measuring NF ⁇ B and MAPK activation are standard in the art (e.g., use of reporter gene assays, nuclear translocation of NF ⁇ B proteins, phosphorylation status of MAPK proteins).
  • Activation through GITR is a co-stimulatory signal that promotes proliferation of activated CD4 + and CD8 + T cells in the presence of activation through the T-cell receptor (e.g., in the presence of primary or target antigen).
  • Methods for measuring proliferation of cells are standard in the art (e.g., 3 H-thymidine incorporation assays, CFSE labeling).
  • GITR Signaling through GITR also co-stimulates activated CD4 + and CD8 + T cells in the presence of activation through the T-cell receptor to produce cytokines. Signaling through GITR also co-stimulates activated NK cells to produce cytokines.
  • the cytokines can be either or both Th1-type cytokines (e.g., interferon- ⁇ , IL-2 and TNF) and Th2-type cytokines (e.g., IL-4, IL-5, IL-10 and IL-13). Methods for measuring cytokine production are well known in the art (e.g., ELISA assays, ELISpot assays). Activation through GITR may also induce apoptosis.
  • Th1-type cytokines e.g., interferon- ⁇ , IL-2 and TNF
  • Th2-type cytokines e.g., IL-4, IL-5, IL-10 and IL-13.
  • test cells or culture supernatant from test cells contacted with the agonist anti-GITR antibodies can be compared to control cells or culture supernatants from control cells that have not been contacted with the agonist anti-GITR antibodies.
  • the GITR agonist functionalities of the present antibodies can also be measured in vivo.
  • Preferred agonist anti-GITR antibodies have the ability to activate and expand CD4 + and CD8 + T-cells.
  • the in vivo activation and expansion of CD4 + and CD8 + T-cells can be measured using any method known in the art, e.g., by flow cytometry.
  • Preferred agonist anti-GITR antibodies can be therapeutically useful in inhibiting tumor growth or promoting tumor retraction. Tumor growth, or inhibition thereof, can be measured using any method known in the art (e.g., visual inspection, calipers, weight, imaging techniques, including MRI).
  • Preferred agonist anti-GITR antibodies can be therapeutically useful in preventing, reducing, inhibiting or eliminating the causative factor of an infectious disease, e.g., a bacterial, fungal, viral or parasitic infection.
  • the efficacy of the agonist anti-GITR antibodies in augmenting a T-cell response or reducing the severity of a disease can be determined by administering a therapeutically effective amount of the antibody to a subject and comparing the subject before and after administration of the antibody.
  • Efficacy of the agonist anti-GITR antibodies in augmenting a T-cell response or reducing the severity of a disease also can be determined by administering a therapeutically effective amount of the antibody to a test subject and comparing the test subject to a control subject who has not been administered the antibody.
  • compositions comprising Agonist Anti-GITR Antibodies
  • compositions comprising the present anti-GITR antibodies or antigen-binding molecules formulated together with a pharmaceutically acceptable carrier.
  • pharmaceutical compositions can additionally contain other therapeutic agents that are suitable for treating or preventing a given disorder.
  • Pharmaceutically acceptable carriers enhance or stabilize the composition, or to facilitate preparation of the composition.
  • Pharmaceutically acceptable carriers include solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • the carrier can be suitable for intravenous, intramuscular, subcutaneous, parenteral, rectal, spinal or epidermal administration (e.g. by injection or infusion).
  • compositions e.g., pharmaceutically acceptable compositions, which include an antibody molecule described herein, formulated together with a pharmaceutically acceptable carrier.
  • the compositions of this invention may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, liposomes and suppositories.
  • liquid solutions e.g., injectable and infusible solutions
  • dispersions or suspensions e.g., injectable and infusible solutions
  • liposomes e.g., liposomes and suppositories.
  • the preferred form depends on the intended mode of administration and therapeutic application. Typical preferred compositions are in the form of injectable or infusible solutions.
  • the preferred mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular).
  • the antibody is administered by intravenous infusion or injection.
  • a pharmaceutical composition of the present invention can be administered by a variety of methods known in the art.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion. Route and/or mode of administration vary depending upon the desired results.
  • administration be intravenous, intramuscular, intraperitoneal, or subcutaneous, or administered proximal to the site of the target.
  • a pharmaceutically acceptable carrier should be suitable for intravenous, intramuscular, subcutaneous, parenteral, intranasal, inhalational, spinal or epidermal administration (e.g., by injection or infusion).
  • active compound e.g., antibody or antigen binding fragment or multivalent molecule of the invention (e.g., monospecific, bispecific or multispecific molecule), may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.
  • Aerosol formulations i.e., they can be “nebulized”) to be administered via inhalation.
  • Aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like.
  • the composition is sterile and fluid. Proper fluidity can be maintained, for example, by use of coating such as lecithin, by maintenance of required particle size in the case of dispersion and by use of surfactants.
  • isotonic agents for example, sugars, polyalcohols such as mannitol or sorbitol, and sodium chloride in the composition. Long-term absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate or gelatin.
  • compositions can be prepared for storage in a lyophilized form using appropriate excipients (e.g., sucrose)
  • compositions of the invention can be prepared in accordance with methods well known and routinely practiced in the art.
  • Pharmaceutically acceptable carriers are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions of the present invention.
  • a therapeutically effective dose or efficacious dose of the anti-GITR antibody is employed in the pharmaceutical compositions of the invention.
  • the anti-GITR antibodies are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art. Dosage regimens are adjusted to provide the desired response (e.g., a therapeutic response). In determining a therapeutically or prophylactically effective dose, a low dose can be administered and then incrementally increased until a desired response is achieved with minimal or no undesired side effects. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • Actual dosage levels of active ingredients in the pharmaceutical compositions of the present invention can be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level depends upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors.
  • the antibody molecules can be administered by a variety of methods known in the art, although for many therapeutic applications, the preferred route/mode of administration is intravenous injection or infusion.
  • the antibody molecules can be administered by intravenous infusion at a rate of more than 20 mg/min, e.g., 20-40 mg/min, and typically greater than or equal to 40 mg/min to reach a dose of about 35 to 440 mg/m 2 , typically about 70 to 310 mg/m 2 , and more typically, about 110 to 130 mg/m 2 .
  • the antibody molecules can be administered by intravenous infusion at a rate of less than 10 mg/min; preferably less than or equal to 5 mg/min to reach a dose of about 1 to 100 mg/m 2 , preferably about 5 to 50 mg/m 2 , about 7 to 25 mg/m 2 and more preferably, about 10 mg/m 2 .
  • the route and/or mode of administration will vary depending upon the desired results.
  • the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.
  • Dosage regimens are adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.
  • an exemplary, non-limiting range for a therapeutically or prophylactically effective amount of an antibody molecule is 0.1-30 mg/kg, more preferably 1-25 mg/kg. Dosages and therapeutic regimens of the anti-GITR antibody molecule can be determined by a skilled artisan.
  • the anti-GITR antibody molecule is administered by injection (e.g., subcutaneously or intravenously) at a dose of about 1 to 40 mg/kg, e.g., 1 to 30 mg/kg, e.g., about 5 to 25 mg/kg, about 10 to 20 mg/kg, about 1 to 5 mg/kg, 1 to 10 mg/kg, 5 to 15 mg/kg, 10 to 20 mg/kg, 15 to 25 mg/kg, or about 3 mg/kg.
  • the dosing schedule can vary from e.g., once a week to once every 2, 3, or 4 weeks.
  • the anti-GITR antibody molecule is administered at a dose from about 10 to 20 mg/kg every other week.
  • the antibody molecule can be administered by intravenous infusion at a rate of more than 20 mg/min, e.g., 20-40 mg/min, and typically greater than or equal to 40 mg/min to reach a dose of about 35 to 440 mg/m 2 , typically about 70 to 310 mg/m 2 , and more typically, about 110 to 130 mg/m 2 .
  • the infusion rate of about 110 to 130 mg/m 2 achieves a level of about 3 mg/kg.
  • the antibody molecule can be administered by intravenous infusion at a rate of less than 10 mg/min, e.g., less than or equal to 5 mg/min to reach a dose of about 1 to 100 mg/m 2 , e.g., about 5 to 50 mg/m 2 , about 7 to 25 mg/m 2 , or, about 10 mg/m 2 .
  • the antibody is infused over a period of about 30 min. It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated.
  • the pharmacological compositions comprise a mixture of the anti-GITR antibody or antigen binding molecule and a second pharmacological agent.
  • exemplary second agents for inclusion in mixtures with the present anti-GITR agonist antibody or antigen binding molecule include without limitation primary or target antigens, agents that increase the immunogenicity of a tumor cell, agents that inhibit or suppress co-inhibitory signals.
  • the anti-GITR antibodies or antigen binding molecules of the invention can be co-formulated (i.e., provided as a mixture or prepared in a mixture) with a primary or target antigen.
  • the target antigen, or vaccine will depend on the disease condition to be treated.
  • the target antigen may be from a tumor cell, a bacterial cell, a fungus, a virus or a parasite.
  • the target antigen can be in the form of a peptide, a polypeptide, a cell or a polynucleotide, as appropriate.
  • the target antigen is from a virus, e.g., selected from the group consisting of: hepatitis type A, hepatitis type B, hepatitis type C, influenza, varicella, adenovirus, herpes simplex type I (HSV I), herpes simplex type II (HSV II), rinderpest, rhinovirus, echovirus, rotavirus, respiratory syncytial virus, papilloma virus, papova virus, cytomegalovirus, echinovirus, arbovirus, hantavirus, coxsackie virus, mumps virus, measles virus, rubella virus, polio virus, human immunodeficiency virus type I (HIV I), and human immunodeficiency virus type II (HIV II), any picornaviridae, enteroviruses, caliciviridae, any of the Norwalk group of viruses, togaviruses, such as alphaviruses, flaviviruse
  • the target antigen is from a bacterium, e.g., selected from the group consisting of: Neisseria spp, Streptococcus spp, S. mutans, Haemophilus spp., Moraxella spp, Bordetella spp, Mycobacterium spp, Legionella spp, Escherichia spp, Vibrio spp, Yersinia spp, Campylobacter spp, Salmonella spp, Listeria spp., Helicobacter spp, Pseudomonas spp, Staphylococcus spp., Enterococcus spp, Clostridium spp., Bacillus spp, Corynebacterium spp., Borrelia spp., Ehrlichia spp, Rickettsia spp, Chlamydia spp., Leptospira spp.
  • the anti-GITR antibodies or antigen binding molecules are co-formulated in a mixture with a tumor-associated antigen (TAA).
  • TAA can be an isolated polypeptide or peptide, can be part of an intact cell or part of a tumor cell lysate.
  • the TAAs can be a polynucleotide, for example a naked plasmid or a viral vector comprising a polynucleotide encoding one or more TAAs.
  • TAAs examples include without limitation, melanoma associated antigens (MAGE-1, MAGE-3, TRP-2, melanosomal membrane glycoprotein gp100, gp75 and MUC-1 (mucin-1) associated with melanoma); CEA (carcinoembryonic antigen) which can be associated, e.g., with ovarian, melanoma or colon cancers; folate receptor alpha expressed by ovarian carcinoma; free human chorionic gonadotropin beta (hCG ⁇ ) subunit expressed by many different tumors, including but not limited to myeloma; HER-2/neu associated with breast cancer; encephalomyelitis antigen HuD associated with small-cell lung cancer; tyrosine hydroxylase associated with neuroblastoma; prostate-specific antigen (PSA) associated with prostate cancer; CA125 associated with ovarian cancer; and the idiotypic determinants of a B cell lymphoma can generate tumor-specific immunity (attributed to idiotype-specific humoral
  • antigens of human T cell leukemia virus type 1 have been shown to induce specific CTL responses and antitumor immunity against the virus-induced human adult T cell leukemia (ATL). See, e.g., Haupt, et al., Experimental Biology and Medicine (2002) 227:227-237; Ohashi, et al., Journal of Virology (2000) 74(20):9610-9616.
  • Other TAAs are known and find use for co-formulation with the anti-GITR antibodies.
  • the anti-GITR antibodies or antigen binding molecules are co-formulated with autologous tumor cells from the patient, or allogeneic tumor cells of the same tissue type from another patient.
  • the tumor cells can be in the form of intact cells, tumor cell lysate, apoptotic tumor cells or total tumor mRNA.
  • the tumor cells can be transfected to express a polypeptide that enhances or augments the immunogenity of the tumor cell in the patient, e.g., transfected to express granulocyte colony stimulating factor (GM-CSF).
  • GM-CSF granulocyte colony stimulating factor
  • the tumor cells can be from any cancerous tissue, including without limitation, epithelial cancers or carcinomas, as well as sarcomas and lymphomas.
  • the cancer is melanoma, ovarian cancer, renal cancer, colorectal cancer, prostate, lung cancer including non-small cell lung cancer (NSCLC), breast cancer, glioma, fibrosarcoma, hematologic cancer, or a head and neck squamous cell carcinoma (HNSCC).
  • NSCLC non-small cell lung cancer
  • HNSCC head and neck squamous cell carcinoma
  • the tumor cell is from, e.g., pancreatic cancer, melanomas, breast cancer, lung cancer, bronchial cancer, colorectal cancer, prostate cancer, stomach cancer, ovarian cancer, urinary bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, cervical cancer, uterine or endometrial cancer, cancer of the oral cavity or pharynx, liver cancer, kidney cancer, testicular cancer, biliary tract cancer, small bowel or appendix cancer, salivary gland cancer, thyroid gland cancer, adrenal gland cancer, osteosarcoma, chondrosarcoma, and cancer of hematological tissues.
  • pancreatic cancer melanomas, breast cancer, lung cancer, bronchial cancer, colorectal cancer, prostate cancer, stomach cancer, ovarian cancer, urinary bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, cervical cancer, uterine or endometrial cancer, cancer of the oral cavity or pharynx,
  • the anti-GITR antibodies or antigen binding molecules are co-formulated with a cytotoxic agent.
  • the anti-GITR antibodies or antigen binding molecules are co-formulated with an agonist antibody or antigen binding molecule that binds to and reduces or depletes CD4+CD25+ regulatory T cells (Treg).
  • Treg CD4+CD25+ regulatory T cells
  • Exemplary Treg cell-depleting antibodies or antigen binding molecules bind to CD25 or CCR4. See, Expert Opin Ther Patents (2007) 17(5):567-575, and the references discussed therein.
  • the anti-GITR antibodies or antigen binding molecules are co-formulated with an inhibitor of a co-inhibitory signal.
  • Exemplary inhibitors include inhibitors of CTLA-4 and inhibitors of the PD-1/PD-L1 (e.g., B7-H1) interaction.
  • the anti-GITR antibodies are co-formulated with an antibody that binds to and inhibits CTLA-4.
  • the anti-GITR antibodies are co-formulated with an antibody that binds to and inhibits TIM3.
  • the anti-GITR antibodies are co-formulated with an antibody that binds to and inhibits LAG3.
  • the anti-GITR antibodies are co-formulated with an antibody that binds to and inhibits PD-1.
  • the anti-GITR antibodies are co-formulated with an antibody that binds to and inhibits B7-H1. See, e.g., Expert Opin Ther Patents (2007) 17(5):567-575; and Melero, et al., Clin Cancer Res (2009) 15(5):1507-1509, and the references discussed therein.
  • formulations comprising a bispecific molecule including an anti-GITR antibody or antigen binding molecule and inhibitor of a co-inhibitory signal.
  • formulations comprise a bispecific molecule including an anti-GITR antibody or antigen binding molecule and an inhibitor of CTLA4.
  • formulations comprise a bispecific molecule including an anti-GITR antibody or antigen binding molecule and an inhibitor of TIM3. In some embodiments, formulations comprise a bispecific molecule including an anti-GITR antibody or antigen binding molecule and an inhibitor of LAG3. In some embodiments, formulations comprise a bispecific molecule including an anti-GITR antibody or antigen binding molecule and an inhibitor of PD-1/PD-L1. In some embodiments, formulations comprise a bispecific molecule including an anti-GITR antibody or antigen binding molecule and an inhibitor B7H1.
  • the anti-GITR antibodies or antigen binding molecules can also be co-formulated with one or more immunostimulatory agents.
  • the anti-GITR antibodies are co-formulated with an immunostimulatory cytokine, for example, IL-7, IL-12 or IL-15.
  • the anti-GITR antibodies or antigen binding molecules can be co-formulated with a second immunostimulatory antibody.
  • the anti-GITR antibodies or antigen binding molecules can also be co-formulated with an agonist antibody or antigen binding molecule of another member of the tumor necrosis factor receptor superfamily.
  • Exemplary secondary immunostimulatory targets include without limitation TNFRSF4 tumor necrosis factor receptor superfamily, member 4 (also known as OX40) or tumor necrosis factor receptor superfamily, member 9 (also known as TNFRSF9, 4-1BB or CD137). See, e.g., Expert Opin Ther Patents (2007) 17(5):567-575; Pardee, et al, Immunotherapy (2009) 1(2):249-264; and Melero, et al., Clin Cancer Res (2009) 15(5):1507-1509, and the references discussed therein.
  • the anti-GITR antibodies or antigen binding molecules can also be co-formulated with a chemotherapeutic agent.
  • the selected agent will depend on the condition to be treated, e.g., a cancer or an infectious disease, such as a bacterial infection, a fungal infection, a viral infection or a parasitic infection.
  • the anti-GITR antibodies or antigen binding molecules can be co-formulated with a chemotherapeutic known by those of skill to treat the disease condition being treated.
  • Chemotherapeutic agents e.g., for the treatment of cancers and infectious diseases are known in the art, and are described, e.g., in Goodman and Gilman's The Pharmacological Basis of Therapeutics, 11th Ed., Brunton, Lazo and Parker, Eds., McGraw-Hill (2006); 2010 Physicians' Desk Reference (PDR), 64th Edition, Thomson PDR.
  • the anti-GITR antibodies or antigen binding molecules can be co-formulated with an antineoplastic agent.
  • antineoplastic agents that find use for mixing in compositions with the anti-GITR antibodies include alkylating agents (e.g., nitrogen mustards, ethyleneimines and methylmelamines, methylhydrazine derivative, alkyl sulfonate, nitrosoureas, triazenes and platinum coordination complexes); antimetabolites (e.g., folic acid analogs, pyrimidine analogs, purine analogs; natural products (e.g., vinca alkaloids, taxanes, epipodophyllotoxins, camptothecins, antibiotics, and anthracenedione).
  • alkylating agents e.g., nitrogen mustards, ethyleneimines and methylmelamines, methylhydrazine derivative, alkyl sulfonate, nitrosoureas, triazenes and platinum coordination complexes
  • antimetabolites
  • the anti-GITR antibodies or antigen binding molecules are co-formulated with an antimetabolite antineoplastic agent, e.g., a folic acid analog (e.g., methotrexate, pemetrexed, trimetrexate), a pyrimidine analog (e.g., 5-fluorouracil, capecitabine, cytarabine, gemcitabine), a purine analog (e.g., mercaptopurine, pentostatin, cladribine fludarabine), or mixtures thereof.
  • an antimetabolite antineoplastic agent e.g., a folic acid analog (e.g., methotrexate, pemetrexed, trimetrexate), a pyrimidine analog (e.g., 5-fluorouracil, capecitabine, cytarabine, gemcitabine), a purine analog (e.g., mercaptopurine, pentostatin, cladribine fludarabine),
  • the anti-GITR antibodies or antigen binding molecules are co-formulated with an alkylating agent antineoplastic agent, e.g., nitrogen mustards (e.g., mechlorethamine, cyclophosphamide, ifosfamide, melphalan, chlorambucil), ethyleneimines (e.g., altretamine) and methylmelamines (e.g., thiotepa), methylhydrazine derivatives (e.g., procarbazine), alkyl sulfonate (e.g., busulfan), nitrosoureas (e.g., carmustine, streptozocin), triazenes (e.g., dacarbazine, temozolomide) and platinum coordination complexes (e.g., cisplatin, carboplatin, oxaliplatin).
  • antineoplastic agent e.g., nitrogen mustards (e.g., mechlore
  • the anti-GITR antibodies or antigen binding molecules can be co-formulated with an antiviral agent.
  • antiviral agents include without limitation anti-herpesvirus agents (e.g., acyclovir, cidofovir, famciclovir, foscarnet, thiovir, fomivirsen, ganciclovir, idoxuridine, penciclovir, trifluridine, valacyclovir, valgenciclovir, resiquimod); anti-influenza agents (e.g., amantadine, oseltamivir, rimantadine, zanamivir, peramivir, E-118958); anti-hepatitis agents (e.g., adeforvir dipivoxil, interferon-alpha, lamivudine, entecavir, clevudine, emtricitabine, telbivudine, tenofovir,
  • the antiviral agent can be an antiretroviral agent.
  • antiretroviral agents include without limitation zidovudine, didanosine, stavudine, zalcitabine, lamivudine, abacavir, tenofavir, emtricitabine, nevirapine, efavirenz, delavirdine, saquinavir, indinavir, ritonavir, nelfinavir, amprenavir, lopinavir, atazanavir, fosamprenavir and enfuvirtide.
  • the anti-GITR antibodies or antigen binding molecules can be co-formulated with an antibacterial agent.
  • antibacterial agents include without limitation sulfonamides (e.g., sulfanilamide, sulfadiazine, sulfamethoxazole, sulfisoxazole, sulfacetamide), trimethoprim, quinolones (e.g., nalidixic acid, cinoxacin, norfloxacin, ciprofloxacin, ofloxacin, sparfloxacin, fleroxacin, perloxacin, levofloxacin, garenoxacin and gemifloxacin), methenamine, nitrofurantoin, penicillins (e.g., penicillin G, penicillin V, methicilin oxacillin, cloxacillin, dicloxacillin, nafcilin, ampicillin, amoxicillin, carbenicillin, ticarcillin,
  • the anti-GITR antibodies or antigen binding molecules can be co-formulated with an anti-parasitic agent.
  • anti-parasitic agents include without limitation anti-malarial agents (e.g., quinolines including chloroquine, mefloquine, quinine, quinidine, and primaquine; diaminopyrimidines including pyrimethamine, sulfadoxine, tetracyclines, atovaquone, and proguanil); anti-protozoal agents including amphotericin, chloroquine, eflornithine, emetine, fumagillin, 8-hydroxyquinolines, melarsoprol, metronidazole, miltefosine, nifurtimox, nitazoxanide, paromomycin, pentamidine, sodium stibogluconate, and suramin.
  • anti-malarial agents e.g., quinolines including chloroquine, mefloqu
  • the anti-GITR antibodies or antigen binding molecules can be co-formulated with an anti-fungal agent.
  • anti-fungal agents include without limitation polyenes (e.g., natamycin, rimocidin, filipin, nystatin, amphotericin B, candicin, and hamycin), imidazoles (e.g., miconazole, ketoconazole, clotrimazole, econazole, bifonazole, butoconazole, fenticonazole, isoconazole, oxiconazole, sertaconazole, sulconazole, tioconazole), triazoles (e.g., fluconazole, itraconazole, isavuconazole, ravuconazole, posaconazole, voriconazole, terconazole), thiazoles (e.g., abafungin), allylamines (e.g., allylamines
  • the anti-GITR compositions of the present invention can be provided in a kit.
  • the anti-GITR antibody, antibody fragment, or antigen binding molecule is generally in a vial or a container.
  • the antibody can be in liquid or dried (e.g., lyophilized) form.
  • the kits can comprise an anti-GITR antibody, antibody fragment, or antigen binding molecule of the invention, as described herein, and optionally also contain a second or third agent.
  • the kits contain anti-GITR antibody, antibody fragment, or antigen binding molecule of the invention and a pharmaceutically acceptable diluent.
  • the anti-GITR antibodies, antibody fragments, or antigen binding molecules can be provided in the kit with the second or third agents in the same or separate formulations (e.g., as mixtures or in separate containers).
  • the kits can contain aliquots of the anti-GITR antibodies, antibody fragments, or antigen binding molecules that provide for one or more doses. If aliquots for multiple administrations are provided, the doses can be uniform or varied. Varied dosing regimens can be escalating or decreasing, as appropriate.
  • the dosages of the anti-GITR antibody, antibody fragment, or antigen binding molecule and the second agent can be independently uniform or varying.
  • the kit can include one or more other elements including: instructions for use; other reagents, e.g., a label, a therapeutic agent, or an agent useful for chelating, or otherwise coupling, an antibody to a label or therapeutic agent, or a radioprotective composition; devices or other materials for preparing the antibody for administration; pharmaceutically acceptable carriers; and devices or other materials for administration to a subject.
  • other reagents e.g., a label, a therapeutic agent, or an agent useful for chelating, or otherwise coupling, an antibody to a label or therapeutic agent, or a radioprotective composition
  • devices or other materials for preparing the antibody for administration e.g., a label, a therapeutic agent, or an agent useful for chelating, or otherwise coupling, an antibody to a label or therapeutic agent, or a radioprotective composition
  • devices or other materials for preparing the antibody for administration e.g., a label, a therapeutic agent, or an agent useful for chelating, or otherwise coupling, an antibody to
  • kits further contain a target antigen.
  • the target antigen, or vaccine will depend on the disease condition to be treated.
  • the target antigen may be from a tumor cell, a bacterial cell, a fungus, a parasite or a virus.
  • the target antigen can be in the form of a peptide, a polypeptide, a cell, a polynucleotide (e.g., naked plasmid or viral vector) as appropriate.
  • the target antigen is a tumor associated antigen. Exemplary target antigens are discussed herein; others known in the art also find use.
  • kits further contain a cytotoxic agent.
  • the kits can contain an agonist antibody or antigen binding molecule that binds to and reduces or depletes CD4+CD25+ regulatory T cells (Treg).
  • Treg CD4+CD25+ regulatory T cells
  • Exemplary Treg cell-depleting antibodies or antigen binding molecules bind to CD25 or CCR4. See, Expert Opin Ther Patents (2007) 17(5):567-575, and the references discussed therein.
  • kits further contain an inhibitor of a co-inhibitory signal.
  • Exemplary inhibitors include inhibitors of CTLA-4, LAG3, TIM3, and/or inhibitors of the PD-1/PD-L1 (e.g., B7-H1) interaction.
  • the kits further contain an antibody that binds to and inhibits CTLA-4.
  • the kits further contain an antibody that binds to and inhibits LAG3.
  • the kits further contain an antibody that binds to and inhibits TIM3.
  • the kits further contain an antibody that binds to and inhibits PD-1.
  • the kits further contain an antibody that binds to and inhibits B7-H1. See, e.g., Expert Opin Ther Patents (2007) 17(5):567-575; and Melero, et al., Clin Cancer Res (2009) 15(5):1507-1509, and the references discussed therein.
  • kits further contain one or more immunostimulatory agents.
  • the kits contain an immunostimulatory cytokine, for example, IL-7, IL-12 or IL-15.
  • the kits can contain a second immunostimulatory antibody.
  • the kits can contain an agonist antibody or antigen binding molecule of another member of the tumor necrosis factor receptor superfamily.
  • Exemplary secondary immunostimulatory targets include without limitation TNFRSF4 tumor necrosis factor receptor superfamily, member 4 (also known as OX40) or tumor necrosis factor receptor superfamily, member 9 (also known as TNFRSF9, 4-1BB or CD137).
  • kits further contain a chemotherapeutic agent.
  • the selected agent will depend on the condition to be treated, e.g., a cancer or an infectious disease, such as a bacterial infection, a fungal infection, a viral infection or a parasitic infection.
  • exemplary chemotherapy agents include any antineoplastic, antiviral, antibacterial, antiparasitic, and antifungal agents known in the art and described herein.
  • the anti-GITR agonist antibodies and antibody fragments of the invention find use in augmenting CD4 + T helper and CD8 + cytolytic T cell responses in a patient in need thereof. Therefore, the antibodies find use in enhancing or augmenting a T cell response in a patient, e.g., to effect the reduction, reversal, inhibition or prevention of a disease that can be counteracted with an enhanced or augmented immune response.
  • the invention provides methods of enhancing a T cell response in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of an anti-GITR agonist antibody or antibody fragment of the invention, as described herein.
  • the invention also provides in one aspect an anti-GITR agonist antibody or antibody fragment for use in enhancing a T cell response in an individual.
  • the invention provides a composition comprising such an antibody or antibody fragment for use in enhancing a T cell response in an individual.
  • Conditions subject to treatment include cancers and infectious disease.
  • the patient may have a cancer or tumor or an infectious disease, e.g., a bacterial, viral, fungal or parasitic infection.
  • an infectious disease e.g., a bacterial, viral, fungal or parasitic infection.
  • the patient may be in remission from a cancer or may anticipate being exposed to a bacterial, viral, fungal or parasitic infection.
  • the antibodies can also serve as an adjuvant to enhance or promote or boost an immune response against a primary antigen or a target antigen, e.g., a vaccine.
  • the patient has a cancer, is suspected of having a cancer, or is in remission from a cancer.
  • Cancers subject to treatment with the anti-GITR antibodies usually express a tumor-associated antigen (TAA), as described herein.
  • TAA tumor-associated antigen
  • Cancers subject to treatment include without limitation epithelial cancers or carcinomas, as well as sarcomas and lymphomas.
  • the cancer is melanoma, ovarian cancer, renal cancer, colorectal cancer, prostate, lung cancer including non-small cell lung cancer (NSCLC), breast cancer, glioma, or fibrosarcoma. See, e.g., Pardee, et al, Immunotherapy (2009) 1(2):249-264, and references discussed therein.
  • the type of cancer is selected from the group consisting of: pancreatic cancer, melanomas, breast cancer, lung cancer, bronchial cancer, colorectal cancer, prostate cancer, stomach cancer, ovarian cancer, urinary bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, cervical cancer, uterine or endometrial cancer, cancer of the oral cavity or pharynx, liver cancer, kidney cancer, testicular cancer, biliary tract cancer, small bowel or appendix cancer, salivary gland cancer, thyroid gland cancer, adrenal gland cancer, osteosarcoma, chondrosarcoma, cancer of hematological tissues and head and neck squamous cell carcinoma (HNSCC).
  • pancreatic cancer melanomas
  • breast cancer breast cancer
  • lung cancer bronchial cancer
  • colorectal cancer prostate cancer
  • stomach cancer ovarian cancer
  • urinary bladder cancer brain or central nervous system cancer
  • peripheral nervous system cancer esophageal cancer
  • cervical cancer uterine or
  • the invention provides methods of treating tumor growth of a cancer that expresses a tumor associated antigen in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of an anti-GITR agonist antibody or antibody fragment of the invention, as described herein.
  • the invention also provides an anti-GITR agonist antibody or antibody fragment of the invention for use in treating tumor growth of a cancer that expresses a tumor associated antigen in an individual.
  • the invention further provides a composition comprising an antibody or antibody fragment of the invention for use in reducing, inhibiting or preventing tumor growth of a cancer that expresses a tumor associated antigen in an individual.
  • methods for facilitating the diagnosis or prognosis of cancer in an individual comprising using an anti-GITR agonist antibody or antibody fragment of the invention for the detection of expression of GITR in or around a tumor in the individual.
  • the patient has an infectious disease, for example, a bacterial, viral, fungal or parasitic infection.
  • infectious disease for example, a bacterial, viral, fungal or parasitic infection.
  • the anti-GITR agonist antibodies find use in reducing, inhibiting and/or preventing parasites in, e.g., filariasis and leishmaniasis.
  • anti-GITR agonist antibodies find use in treatment of viral infections, including without limitation hepatitis virus infection, for example, chronic hepatitis C (HCV) infection, herpes simplex virus (HSV) infection or human immunodeficiency virus (HIV) infection.
  • hepatitis virus infection for example, chronic hepatitis C (HCV) infection, herpes simplex virus (HSV) infection or human immunodeficiency virus (HIV) infection.
  • HCV chronic hepatitis C
  • HSV herpes simplex virus
  • HSV human immunodeficiency virus
  • anti-GITR agonist antibodies find use in treating a viral infection selected from the group consisting of: hepatitis type A, hepatitis type B, hepatitis type C, influenza, varicella, adenovirus, herpes simplex type I (HSV I), herpes simplex type II (HSV II), rinderpest, rhinovirus, echovirus, rotavirus, respiratory syncytial virus, papilloma virus, papova virus, cytomegalovirus, echinovirus, arbovirus, hantavirus, coxsackie virus, mumps virus, measles virus, rubella virus, polio virus, human immunodeficiency virus type I (HIV I), and human immunodeficiency virus type II (HIV II), any picornaviridae, enteroviruses, caliciviridae, any of the Norwalk group of viruses, togaviruses, such as alphaviruses, flaviviruse
  • anti-GITR agonist antibodies find use in treating bacterial infections, including without limitation an infection of Neisseria spp, Streptococcus spp, S. mutans, Haemophilus spp., Moraxella spp, Bordetella spp, Mycobacterium spp, Legionella spp, Escherichia spp, Vibrio spp, Yersinia spp, Campylobacter spp, Salmonella spp, Listeria spp., Helicobacter spp, Pseudomonas spp, Staphylococcus spp., Enterococcus spp, Clostridium spp., Bacillus spp, Corynebacterium spp., Borrelia spp., Ehrlichia spp, Rickettsia spp, Chlamydia spp., Leptospira spp., Trepone
  • a physician or veterinarian can start doses of the antibodies or antibody fragments of the invention employed in the pharmaceutical composition at levels lower than that required to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • effective doses of the compositions of the present invention vary depending upon many different factors, including the specific disease or condition to be treated, means of administration, target site, physiological state of the patient, whether the patient is human or an animal, other medications administered, and whether treatment is prophylactic or therapeutic. Treatment dosages need to be titrated to optimize safety and efficacy.
  • the dosage ranges from about 0.0001 to 100 mg/kg, and more usually 0.01 to 5 mg/kg, of the host body weight.
  • dosages can be 1 mg/kg body weight or 10 mg/kg body weight or within the range of 1-10 mg/kg.
  • Dosing can be daily, weekly, bi-weekly, monthly, or more or less often, as needed or desired.
  • An exemplary treatment regime entails administration once weekly, once per every two weeks or once a month or once every 3 to 6 months.
  • an polynucleotide encoding an anti-GITR antibody, antibody fragment, or antigen binding molecule of the invention is administered.
  • the agent is a nucleic acid
  • typical dosages can range from about 0.1 mg/kg body weight up to and including about 100 mg/kg body weight, e.g., between about 1 mg/kg body weight to about 50 mg/kg body weight. In some embodiments, about 1, 2, 3, 4, 5, 10, 15, 20, 30, 40 or 50 mg/kg body weight.
  • the antibody or antibody fragment can be administered in single or divided doses.
  • Antibody or antibody fragment is usually administered on multiple occasions. Intervals between single dosages can be weekly, bi-weekly, monthly or yearly, as needed or desired. Intervals can also be irregular as indicated by measuring blood levels of anti-GITR antibody or antibody fragment in the patient. In some methods, dosage is adjusted to achieve a plasma antibody or antibody fragment concentration of 1-1000 ⁇ g/ml and in some methods 25-300 ⁇ g/ml. Alternatively, antibody or antibody fragment can be administered as a sustained release formulation, in which case less frequent administration is required. Dosage and frequency vary depending on the half-life of the antibody or antibody fragment in the patient. In general, humanized antibodies show longer half life than that of chimeric antibodies and nonhuman antibodies.
  • the dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic.
  • a relatively low dosage is administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives.
  • a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated, and preferably until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patient can be administered a prophylactic regime.
  • the anti-GITR antibody, antibody fragment, or antigen binding molecule is co-administered with a second or third pharmacological agent.
  • the anti-GITR antibody, antibody fragment, or antigen binding molecule and the second or agent can be administered as a mixture or in separate formulations.
  • the anti-GITR antibody, antibody fragment, or antigen binding molecule and the second or agent can be administered concurrently or sequentially.
  • the anti-GITR antibody, antibody fragment, or antigen binding molecule and the second or agent can be administered via the same route of administration or via different routes of administration, as appropriate.
  • Exemplary second agents and third agents for co-administration with the present anti-GITR agonist antibodies, antibody fragments, or antigen binding molecules include without limitation, primary or target antigens, agents that increase the immunogenicity of a tumor cell, agents that inhibit or suppress co-inhibitory signals.
  • the anti-GITR agonist antibodies, antibody fragments, or antigen binding molecules can also be co-administered with chemotherapeutic used to treat the disease condition being treated, e.g., to enhance the efficacy of the chemotherapeutic agent or to further enhance an immune response against a target antigen.
  • the anti-GITR agonist antibodies, antibody fragments, or antigen binding molecules also find use in combination therapies with established procedures for treating the designated disease condition, e.g., radiation or surgery.
  • a combination or “in combination with,” it is not intended to imply that the therapy or the therapeutic agents must be administered at the same time and/or formulated for delivery together, although these methods of delivery are within the scope described herein.
  • the therapeutic agents in the combination can be administered concurrently with, prior to, or subsequent to, one or more other additional therapies or therapeutic agents.
  • the therapeutic agents or therapeutic protocol can be administered in any order. In general, each agent will be administered at a dose and/or on a time schedule determined for that agent.
  • the additional therapeutic agent utilized in this combination may be administered together in a single composition or administered separately in different compositions. In general, it is expected that additional therapeutic agents utilized in combination be utilized at levels that do not exceed the levels at which they are utilized individually. In some embodiments, the levels utilized in combination will be lower than those utilized individually.
  • the additional therapeutic agent is administered at a therapeutic or lower-than therapeutic dose.
  • the concentration of the second therapeutic agent that is required to achieve inhibition, e.g., growth inhibition is lower when the second therapeutic agent is administered in combination with the first therapeutic agent, e.g., the anti-GITR antibody molecule, than when the second therapeutic agent is administered individually.
  • the concentration of the first therapeutic agent that is required to achieve inhibition, e.g., growth inhibition is lower when the first therapeutic agent is administered in combination with the second therapeutic agent than when the first therapeutic agent is administered individually.
  • the concentration of the second therapeutic agent that is required to achieve inhibition, e.g., growth inhibition is lower than the therapeutic dose of the second therapeutic agent as a monotherapy, e.g., 10-20%, 20-30%, 30-40%, 40-50%, 50-60%, 60-70%, 70-80%, or 80-90% lower.
  • the concentration of the first therapeutic agent that is required to achieve inhibition, e.g., growth inhibition is lower than the therapeutic dose of the first therapeutic agent as a monotherapy, e.g., 10-20%, 20-30%, 30-40%, 40-50%, 50-60%, 60-70%, 70-80%, or 80-90% lower.
  • inhibitor includes a reduction in a certain parameter, e.g., an activity, of a given molecule, e.g., an immune checkpoint inhibitor.
  • a certain parameter e.g., an activity, of a given molecule
  • an immune checkpoint inhibitor e.g., an enzyme inhibitor, e.g., a cell proliferation inhibitor, or a cell proliferation factor receptor mediated by a cell proliferation factor.
  • inhibition of an activity e.g., a coinhibitory activity, of at least 5%, 10%, 20%, 30%, 40% or more is included by this term. Thus, inhibition need not be 100%.
  • activation includes an increase in a certain parameter, e.g., an activity, of a given molecule, e.g., a costimulatory molecule.
  • a certain parameter e.g., an activity, of a given molecule
  • a costimulatory molecule e.g., a costimulatory molecule
  • increase of an activity, e.g., a costimulatory activity, of at least 5%, 10%, 25%, 50%, 75% or more is included by this term.
  • the anti-GITR antibodies, antibody fragments, or antigen binding molecules of the invention can be co-administered with a primary or target antigen.
  • the target antigen, or vaccine will depend on the disease condition to be treated.
  • the target antigen may be from a tumor cell, a bacterial cell, a fungus, a virus or a parasite.
  • the target antigen can be in the form of a peptide, a polypeptide, a cell or a polynucleotide, as appropriate.
  • the anti-GITR antibodies, antibody fragments, or antigen binding molecules are co-administered with a target antigen from a virus, e.g., selected from the group consisting of: hepatitis type A, hepatitis type B, hepatitis type C, influenza, varicella, adenovirus, herpes simplex type I (HSV I), herpes simplex type II (HSV II), rinderpest, rhinovirus, echovirus, rotavirus, respiratory syncytial virus, papilloma virus, papova virus, cytomegalovirus, echinovirus, arbovirus, hantavirus, coxsackie virus, mumps virus, measles virus, rubella virus, polio virus, human immunodeficiency virus type I (HIV I), and human immunodeficiency virus type II (HIV II), any picornaviridae, enteroviruses, caliciviridae, any of
  • the anti-GITR antibodies, antibody fragments, or antigen binding molecules are co-administered with target antigen from a bacterium, e.g., selected from the group consisting of: Neisseria spp, Streptococcus spp, S.
  • mutans Haemophilus spp., Moraxella spp, Bordetella spp, Mycobacterium spp, Legionella spp, Escherichia spp, Vibrio spp, Yersinia spp, Campylobacter spp, Salmonella spp, Listeria spp., Helicobacter spp, Pseudomonas spp, Staphylococcus spp., Enterococcus spp, Clostridium spp., Bacillus spp, Corynebacterium spp., Borrelia spp., Ehrlichia spp, Rickettsia spp, Chlamydia spp., Leptospira spp., Treponema spp.
  • the anti-GITR antibodies, antibody fragments, or antigen binding molecules are co-administered with a tumor-associated antigen (TAA).
  • TAA tumor-associated antigen
  • the TAA can be an isolated polypeptide or peptide, can be part of an intact cell or part of a tumor cell lysate. Exemplary TAAs are discussed above; others known in the art also find use.
  • the anti-GITR antibodies, antibody fragments, or antigen binding molecules are co-administered with autologous tumor cells from the patient, or allogeneic tumor cells of the same tissue type from another patient.
  • the tumor cells can be in the form of intact cells, tumor cell lysate, apoptotic tumor cells or total tumor mRNA.
  • the tumor cells can be transfected to express a polypeptide that enhances or augments the immunogenity of the tumor cell in the patient, e.g., transfected to express granulocyte colony stimulating factor (GM-CSF).
  • GM-CSF granulocyte colony stimulating factor
  • the tumor cells can be from any cancerous tissue, including without limitation, epithelial cancers or carcinomas, as well as sarcomas and lymphomas.
  • the cancer is melanoma, ovarian cancer, renal cancer, colorectal cancer, prostate, lung cancer including non-small cell lung cancer (NSCLC), breast cancer, glioma, or fibrosarcoma.
  • NSCLC non-small cell lung cancer
  • breast cancer glioma
  • fibrosarcoma See, e.g., Pardee, et al, Immunotherapy (2009) 1(2):249-264, and references discussed therein.
  • the tumor cell is from, e.g., pancreatic cancer, melanomas, breast cancer, lung cancer, bronchial cancer, colorectal cancer, prostate cancer, stomach cancer, ovarian cancer, urinary bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, cervical cancer, uterine or endometrial cancer, cancer of the oral cavity or pharynx, liver cancer, kidney cancer, testicular cancer, biliary tract cancer, small bowel or appendix cancer, salivary gland cancer, thyroid gland cancer, adrenal gland cancer, osteosarcoma, chondrosarcoma, cancer of hematological tissues and head and neck squamous cell carcinoma (HNSCC).
  • pancreatic cancer melanomas, breast cancer, lung cancer, bronchial cancer, colorectal cancer, prostate cancer, stomach cancer, ovarian cancer, urinary bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, cervical cancer, uterine or endometrial
  • an immunomodulator used in the combinations with an anti GITR antibody molecule of the invention is an inhibitor of an immune checkpoint molecule.
  • the immunomodulator is an inhibitor of PD-1, PD-L1, PD-L2, CTLA4, TIM-3, LAG-3, CEACAM (e.g., CEACAM-1, -3 and/or -5), VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and/or TGFR beta.
  • the inhibitor of an immune checkpoint molecule inhibits PD-1, PD-L1, LAG-3, TIM-3, CEACAM (e.g., CEACAM-1, -3 and/or -5), CTLA-4, or any combination thereof.
  • Inhibition of an inhibitory molecule can be performed at the DNA, RNA or protein level.
  • an inhibitory nucleic acid e.g., a dsRNA, siRNA or shRNA
  • a dsRNA, siRNA or shRNA can be used to inhibit expression of an inhibitory molecule.
  • the inhibitor of an inhibitory signal is, a polypeptide e.g., a soluble ligand (e.g., PD-1-Ig or CTLA-4 Ig), or an antibody or antigen-binding fragment thereof, that binds to the inhibitory molecule; e.g., an antibody or fragment thereof (also referred to herein as “an antibody molecule”) that binds to PD-1, PD-L1, PD-L2, CEACAM (e.g., CEACAM-1, -3 and/or -5), CTLA-4, TIM-3, LAG-3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and/or TGFR beta, or a combination thereof.
  • a polypeptide e.g., a soluble ligand (e.g., PD-1-Ig or CTLA-4 Ig), or an antibody or antigen-binding fragment thereof, that binds to the inhibitory molecule; e.g., an antibody or fragment
  • the GITR agonist can be administered alone, or in combination with other immunomodulators, e.g., in combination with an inhibitor of PD-1, PD-L1, CTLA-4, CEACAM (e.g., CEACAM-1, -3 and/or -5), TIM-3 or LAG-3.
  • the anti-GITR antibody molecule e.g., as described herein
  • the GITR antibody molecule and the anti-PD-1 antibody molecule may be in the form of separate antibody composition, or as a bispecific antibody molecule.
  • a GITR agonist can be administered in combination with other costimulatory molecule, e.g., an agonist of OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), 4-1BB (CD137), CD30, CD40, BAFFR, HVEM, CD7, LIGHT, NKG2C, SLAMF7, NKp80, CD160, B7-H3, or CD83 ligand.
  • costimulatory molecule e.g., an agonist of OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), 4-1BB (CD137), CD30, CD40, BAFFR, HVEM, CD7, LIGHT, NKG2C, SLAMF7, NKp80, CD160, B7-H3, or CD83 ligand.
  • the antibody molecule is in the form of a bispecific or multispecific antibody molecule.
  • the anti-GITR antibody molecule is a bispecific antibody that binds to GITR and PD-1, PD-L1, CTLA-4, CEACAM (e.g., CEACAM-1, -3 and/or -5), TIM-3 or LAG-3.
  • the bispecific antibody molecule has a first binding specificity to GITR and a second binding specifity, e.g., a second binding specificity to PD-1, PD-L1 TIM-3, CEACAM (e.g., CEACAM-1, -3 and/or -5), LAG-3, or PD-L2.
  • the immunomodulator is an agonist of GITR, e.g., human GITR (e.g., an antibody molecule as described herein).
  • the immunomodulator is an antagonist, e.g., human GITRL.
  • the agonist of GITR and/or antagonist of GITRL is an antibody molecule to GITR.
  • the GITR antibody can be administered alone, or in combination with other immunomodulators, e.g., in combination with an inhibitor of PD-1, PD-L1, LAG-3, TIM-3, CEACAM (e.g., CEACAM-1, -3 and/or -5) or CTLA-4.
  • the agonist of GITR e.g., the anti-GITR antibody molecule
  • a PD-1 or PD-L1 inhibitor e.g., a PD-1 or PD-L1 inhibitor.
  • the agonist of GITR e.g., the anti-GITR antibody molecule
  • a LAG-3 inhibitor e.g., an anti-LAG-3 antibody molecule.
  • the agonist of GITR e.g., the anti-GITR antibody molecule
  • is administered in combination with a TIM-3 inhibitor e.g., an anti-TIM-3 antibody molecule.
  • the agonist of GITR e.g., the anti-GITR antibody molecule
  • a CEACAM inhibitor e.g., CEACAM-1, -3 and/or -5 inhibitor
  • the agonist of GITR e.g., the anti-GITR antibody molecule
  • a CEACAM-1 inhibitor e.g., an anti-CEACAM-1 antibody molecule
  • the agonist of GITR e.g., the anti-GITR antibody molecule
  • is administered in combination with a CEACAM-5 inhibitor e.g., an anti-CEACAM-5 antibody molecule.
  • the agonist of GITR e.g., the anti-GITR antibody molecule
  • a LAG-3 inhibitor e.g., an anti-LAG-3 antibody molecule
  • a TIM-3 inhibitor e.g., an anti-TIM-3 antibody molecule
  • Other combinations of immunomodulators with a GITR modulator e.g., one or more of PD-L2, CTLA-4, TIM-3, LAG-3, CEACAM (e.g., CEACAM-1, -3 and/or -5), VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and/or TGFR
  • Any of the antibody molecules known in the art or disclosed herein can be used in the aforesaid combinations of inhibitors of checkpoint molecule.
  • the immunomodulator is an inhibitor of CEACAM (e.g., CEACAM-1, -3 and/or -5), e.g., human CEACAM (e.g., CEACAM-1, -3 and/or -5).
  • the immunomodulator is an inhibitor of CEACAM-1, e.g., human CEACAM-1.
  • the immunomodulator is an inhibitor of CEACAM-3, e.g., human CEACAM-3.
  • the immunomodulator is an inhibitor of CEACAM-5, e.g., human CEACAM-5.
  • the inhibitor of CEACAM is an antibody molecule to CEACAM (e.g., CEACAM-1, -3 and/or -5).
  • the CEACAM (e.g., CEACAM-1, -3 and/or -5) inhibitor can be administered alone, or in combination with other immunomodulators, e.g., in combination with an inhibitor of LAG-3, TIM-3, PD-1, PD-L1 or CTLA-4.
  • the immunomodulator is an inhibitor of LAG-3, e.g., human LAG-3.
  • the inhibitor of LAG-3 is an antibody molecule to LAG-3.
  • the LAG-3 inhibitor can be administered alone, or in combination with other immunomodulators, e.g., in combination with an inhibitor of CEACAM (e.g., CEACAM-1, -3 and/or -5), TIM-3, PD-1, PD-L1 or CTLA-4.
  • CEACAM e.g., CEACAM-1, -3 and/or -5
  • TIM-3 e.g., PD-1, PD-L1 or CTLA-4.
  • the immunomodulator is an inhibitor of TIM-3, e.g., human TIM-3.
  • the inhibitor of TIM-3 is an antibody molecule to TIM-3.
  • the TIM-3 inhibitor can be administered alone, or in combination with other immunomodulators, e.g., in combination with an inhibitor of CEACAM (e.g., CEACAM-1, -3 and/or -5), LAG-3, PD-1, PD-L1 or CTLA-4.
  • CEACAM e.g., CEACAM-1, -3 and/or -5
  • LAG-3 e.g., PD-1, PD-L1 or CTLA-4.
  • the immunomodulator used in the combinations disclosed herein is an activator or agonist of a costimulatory molecule.
  • the agonist of the costimulatory molecule is chosen from an agonist (e.g., an agonistic antibody or antigen-binding fragment thereof, or a soluble fusion) of OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), 4-1BB (CD137), GITR, CD30, CD40, BAFFR, HVEM, CD7, LIGHT, NKG2C, SLAMF7, NKp80, CD160, B7-H3, or CD83 ligand.
  • the immunomodulator is an activator of a costimulatory molecule (e.g., an OX40 agonist).
  • the OX40 agonist is an antibody molecule to OX40.
  • the OX40 agonist can be administered alone, or in combination with other immunomodulators, e.g., in combination with an inhibitor of PD-1, PD-L1, CTLA-4, CEACAM (e.g., CEACAM-1, -3 and/or -5), TIM-3 or LAG-3.
  • the anti-OX40 antibody molecule is a bispecific antibody that binds to GITR and PD-1, PD-L1, CTLA-4, CEACAM (e.g., CEACAM-1, -3 and/or -5), TIM-3 or LAG-3.
  • an OX40 antibody molecule is administered in combination with an anti-GITR antibody molecule (e.g., an anti-GITR molecule as described herein).
  • the OX40 antibody molecule and the anti-GITR antibody molecule may be in the form of separate antibody composition, or as a bispecific antibody molecule.
  • the OX40 agonist can be administered in combination with other costimulatory molecule, e.g., an agonist of GITR, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), 4-1BB (CD137), CD30, CD40, BAFFR, HVEM, CD7, LIGHT, NKG2C, SLAMF7, NKp80, CD160, B7-H3, or CD83 ligand.
  • costimulatory molecule e.g., an agonist of GITR, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), 4-1BB (CD137), CD30, CD40, BAFFR, HVEM, CD7, LIGHT, NKG2C, SLAMF7, NKp80, CD160, B7-H3, or CD83 ligand.
  • the immunomodulator is an activator of a costimulatory molecule (e.g., an 4-1BB (CD-137) agonist).
  • the 4-1BB (CD-137) agonist is an antibody molecule to 4-1BB (CD-137).
  • the combination disclosed herein e.g., a combination comprising an anti-GITR antibody molecule, is administered with a 4-1BB receptor targeting agent (e.g., an antibody that stimulates signaling through 4-1BB (CD-137), e.g., PF-2566).
  • the anti-GITR antibody molecule is administered in combination with a tyrosine kinase inhibitor (e.g., axitinib) and a 4-1BB receptor targeting agent.
  • the 4-1BB agonist can be combined alone, or in combination with other immunomodulators, e.g., in combination with an inhibitor of PD-1, PD-L1, CTLA-4, CEACAM (e.g., CEACAM-1, -3 and/or -5), TIM-3 or LAG-3.
  • the anti-4-1BB antibody molecule is a bispecific antibody that binds to GITR and PD-1, PD-L1, CTLA-4, CEACAM (e.g., CEACAM-1, -3 and/or -5), TIM-3 or LAG-3.
  • an 4-1BB antibody molecule is administered in combination with an anti-GITR antibody molecule (e.g., an anti-GITR molecule as described herein).
  • the 4-1BB antibody molecule and the anti-GITR antibody molecule may be in the form of separate antibody composition, or as a bispecific antibody molecule.
  • the 4-1BB agonist can be administered in combination with other costimulatory molecule, e.g., an agonist of GITR, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), 4-1BB (CD137), CD30, CD40, BAFFR, HVEM, CD7, LIGHT, NKG2C, SLAMF7, NKp80, CD160, B7-H3, or CD83 ligand.
  • costimulatory molecule e.g., an agonist of GITR, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), 4-1BB (CD137), CD30, CD40, BAFFR, HVEM, CD7, LIGHT, NKG2C, SLAMF
  • the anti-GITR antibodies, antibody fragments, or antigen binding molecules are co-administered with a cytotoxic agent.
  • the anti-GITR antibodies or antigen binding molecules are co-administered with an agonist antibody or antigen binding molecule that binds to and reduces or depletes CD4+CD25+ regulatory T cells (Treg).
  • Treg CD4+CD25+ regulatory T cells
  • Exemplary Treg cell-depleting antibodies or antigen binding molecules bind to CD25 or CCR4. See, Expert Opin Ther Patents (2007) 17(5):567-575, and the references discussed therein.
  • the anti-GITR antibodies, antibody fragments, or antigen binding molecules are co-administered with an inhibitor of a co-inhibitory signal.
  • exemplary inhibitors include inhibitors of CTLA-4, LAG3, TIM3 and/or inhibitors of the PD-1/PD-L1 (e.g., B7-H1) interaction.
  • the anti-GITR antibodies are co-administered with an antibody that binds to and inhibits CTLA-4.
  • the anti-GITR antibodies are co-administered with an antibody that binds to and inhibits TIM3.
  • the anti-GITR antibodies are co-administered with an antibody that binds to and inhibits LAG3.
  • the anti-GITR antibodies are co-administered with an antibody that binds to and inhibits PD-1. In some embodiments, the anti-GITR antibodies are co-administered with an antibody that binds to and inhibits B7-H1. See, e.g., Expert Opin Ther Patents (2007) 17(5):567-575; and Melero, et al., Clin Cancer Res (2009) 15(5):1507-1509, and the references discussed therein.
  • the anti-GITR antibodies, antibody fragments, or antigen binding molecules can also be co-administered with one or more immunostimulatory agents.
  • the anti-GITR antibodies or antibody fragments are co-administered with an immunostimulatory cytokine, for example, IL-7, IL-12 or IL-15.
  • the anti-GITR antibodies, antibody fragments, or antigen binding molecules can be co-administered with a second immunostimulatory antibody.
  • the anti-GITR antibodies, antibody fragments, or antigen binding molecules can also be co-administered with an agonist antibody, antibody fragment, or antigen binding molecule of another member of the tumor necrosis factor receptor superfamily.
  • Exemplary secondary immunostimulatory targets include without limitation TNFRSF4 tumor necrosis factor receptor superfamily, member 4 (also known as OX40) or tumor necrosis factor receptor superfamily, member 9 (also known as TNFRSF9, 4-1BB or CD137). See, e.g., Expert Opin Ther Patents (2007) 17(5):567-575; Pardee, et al, Immunotherapy (2009) 1(2):249-264; and Melero, et al., Clin Cancer Res (2009) 15(5):1507-1509, and the references discussed therein.
  • the anti-GITR antibodies, antibody fragments, or antigen binding molecules can also be co-administered with a chemotherapeutic agent.
  • a chemotherapeutic agent will depend on the condition to be treated, e.g., a cancer or an infectious disease, such as a bacterial infection, a fungal infection, a viral infection or a parasitic infection.
  • the anti-GITR antibodies, antibody fragments, or antigen binding molecules can be co-administered with a chemotherapeutic known by those of skill to treat the disease condition being treated. Exemplary chemotherapeutic agents are discussed above; others known in the art also find use.
  • the anti-GITR antibody molecule includes: an antibody or an antibody fragment, or antigen binding molecule thereof that binds to SEQ ID NO:1, and wherein the antibody, antibody fragment, or antigen binding molecule comprises
  • any of the combinations disclosed herein further includes one or more of the agents described in Table 6.
  • the additional therapeutic agent is chosen from one or more of: 1) a protein kinase C (PKC) inhibitor; 2) a heat shock protein 90 (HSP90) inhibitor; 3) an inhibitor of a phosphoinositide 3-kinase (PI3K) and/or target of rapamycin (mTOR); 4) an inhibitor of cytochrome P450 (e.g., a CYP17 inhibitor or a 17alpha-Hydroxylase/C17-20 Lyase inhibitor); 5) an iron chelating agent; 6) an aromatase inhibitor; 7) an inhibitor of p53, e.g., an inhibitor of a p53/Mdm2 interaction; 8) an apoptosis inducer; 9) an angiogenesis inhibitor; 10) an aldosterone synthase inhibitor; 11) a smoothened (SMO) receptor inhibitor; 12) a prolactin receptor (PRLR) inhibitor; 13) a Wnt signaling inhibitor; 14)
  • PIC protein
  • the additional therapeutic agent is chosen from one or more of: Compound A8, Compound A17, Compound A23, Compound A24, Compound A27, Compound A29, Compound A33, and Compound A13.
  • the additional therapeutic agent is chosen from one or more of: Compound A5, Compound A8, Compound A17, Compound A23, Compound A24, Compound A29, and Compound A40.
  • the additional therapeutic agent is chosen from one or more of: Compound A9, Compound A16, Compound A17, Compound A21, Compound A22, Compound A25, Compound A28, Compound A48, and Compound 49.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a PKC inhibitor, Sotrastaurin (Compound A1), or a compound disclosed in PCT Publication No. WO 2005/039549, to treat a disorder, e.g., a disorder described herein.
  • the PKC inhibitor is Sotrastaurin (Compound A1) or a compound disclosed in PCT Publication No. WO 2005/039549.
  • a GITR antibody molecule is used in combination with Sotrastaurin (Compound A1), or a compound as described in PCT Publication No.
  • WO 2005/039549 to treat a disorder such as a cancer, a melanoma, a non-Hodgkin lymphoma, an inflammatory bowel disease, transplant rejection, an ophthalmic disorder, or psoriasis.
  • Sotrastaurin (Compound A1) is administered at a dose of about 20 to 600 mg, e.g., about 200 to about 600 mg, about 50 mg to about 450 mg, about 100 mg to 400 mg, about 150 mg to 350 mg, or about 200 mg to 300 mg, e.g., about 50 mg, 100 mg, 150 mg, 200 mg, 300 mg, 400 mg, 500 mg, or 600 mg.
  • the dosing schedule can vary from e.g., every other day to daily, twice or three times a day.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as provided herein, is used in combination with a BCR-ABL inhibitor, TASIGNA (Compound A2), or a compound disclosed in PCT Publication No. WO 2004/005281, to treat a disorder, e.g., a disorder described herein.
  • the BCR-ABL inhibitor is TASIGNA, or a compound disclosed in PCT Publication No. WO 2004/005281.
  • a GITR antibody molecule is used in combination with TASIGNA (Compound A2), or a compound as described in PCT Publication No.
  • WO 2004/005281 to treat a disorder such as a lymphocytic leukemia, Parkinson's Disease, a neurologic cancer, a melanoma, a digestive/gastrointestinal cancer, a colorectal cancer, a myeloid leukemia, a head and neck cancer, or pulmonary hypertension.
  • a disorder such as a lymphocytic leukemia, Parkinson's Disease, a neurologic cancer, a melanoma, a digestive/gastrointestinal cancer, a colorectal cancer, a myeloid leukemia, a head and neck cancer, or pulmonary hypertension.
  • the BCR-ABL inhibitor or TASIGNA is administered at a dose of about 300 mg (e.g., twice daily, e.g., for newly diagnosed Ph+ CML-CP), or about 400 mg, e.g., twice daily, e.g., for resistant or intolerant Ph+ CML-CP and CML-AP).
  • BCR-ABL inhibitor or a Compound A2 is administered at a dose of about 300-400 mg.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as provided herein, is used in combination with an HSP90 inhibitor, such as 5-(2,4-dihydroxy-5-isopropylphenyl)-N-ethyl-4-(4-(morpholinomethyl)phenyl)isoxazole-3-carboxamide (Compound A3), or a compound disclosed in PCT Publication No. WO 2010/060937 or WO 2004/072051, to treat a disorder, e.g., a disorder described herein.
  • an HSP90 inhibitor such as 5-(2,4-dihydroxy-5-isopropylphenyl)-N-ethyl-4-(4-(morpholinomethyl)phenyl)isoxazole-3-carboxamide (Compound A3), or a compound disclosed in PCT Publication No. WO 2010/060937 or WO 2004/072051, to treat a disorder, e.g., a disorder described herein.
  • the HSP90 inhibitor is 5-(2,4-dihydroxy-5-isopropylphenyl)-N-ethyl-4-(4-(morpholinomethyl)phenyl)isoxazole-3-carboxamide (Compound A3), or a compound disclosed in PCT Publication No. WO 2010/060937 or WO 2004/072051.
  • a GITR antibody molecule is used in combination with 5-(2,4-dihydroxy-5-isopropylphenyl)-N-ethyl-4-(4-(morpholinomethyl)phenyl)isoxazole-3-carboxamide (Compound A3), or a compound as described in PCT Publication No.
  • WO 2010/060937 or WO 2004/072051 to treat a disorder such as a cancer, a multiple myeloma, a non-small cell lung cancer, a lymphoma, a gastric cancer, a breast cancer, a digestive/gastrointestinal cancer, a pancreatic cancer, a colorectal cancer, a solid tumor, or a hematopoiesis disorder.
  • a disorder such as a cancer, a multiple myeloma, a non-small cell lung cancer, a lymphoma, a gastric cancer, a breast cancer, a digestive/gastrointestinal cancer, a pancreatic cancer, a colorectal cancer, a solid tumor, or a hematopoiesis disorder.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with an inhibitor of PI3K and/or mTOR, Dactolisib (Compound A4) or 8-(6-Methoxy-pyridin-3-yl)-3-methyl-1-(4-piperazin-1-yl-3-trifluoromethyl-phenyl)-1,3-dihydro-imidazo[4,5-c]quinolin-2-one (Compound A41), or a compound disclosed in PCT Publication No. WO 2006/122806, to treat a disorder, e.g., a disorder described herein.
  • a disorder e.g., a disorder described herein.
  • the PI3K and/or mTOR inhibitor is Dactolisib (Compound A4), 8-(6-Methoxy-pyridin-3-yl)-3-methyl-1-(4-piperazin-1-yl-3-trifluoromethyl-phenyl)-1,3-dihydro-imidazo[4,5-c]quinolin-2-one (Compound A41), or a compound disclosed in PCT Publication No. WO 2006/122806.
  • a GITR antibody molecule is used in combination with Dactolisib (Compound A4), 8-(6-Methoxy-pyridin-3-yl)-3-methyl-1-(4-piperazin-1-yl-3-trifluoromethyl-phenyl)-1,3-dihydro-imidazo[4,5-c]quinolin-2-one (Compound A41), or a compound described in PCT Publication No.
  • WO 2006/122806 to treat a disorder such as a cancer, a prostate cancer, a leukemia (e.g., lymphocytic leukemia), a breast cancer, a brain cancer, a bladder cancer, a pancreatic cancer, a renal cancer, a solid tumor, or a liver cancer.
  • a leukemia e.g., lymphocytic leukemia
  • a breast cancer e.g., a breast cancer
  • a brain cancer e.g., a bladder cancer, a pancreatic cancer, a renal cancer, a solid tumor, or a liver cancer.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with an FGFR inhibitor, 3-(2,6-dichloro-3,5-dimethoxyphenyl)-1-(6-((4-(4-ethylpiperazin-1-yl)phenyl)amino)pyrimidin-4-yl)-1-methylurea (Compound A5) or a compound disclosed in U.S. Pat. No. 8,552,002, to treat a disorder, e.g., a disorder described herein.
  • FGFR inhibitor 3-(2,6-dichloro-3,5-dimethoxyphenyl)-1-(6-((4-(4-ethylpiperazin-1-yl)phenyl)amino)pyrimidin-4-yl)-1-methylurea
  • Compound A5 3-(2,6-dichloro-3,5-dimethoxyphenyl)-1-(6-((4-
  • the FGFR inhibitor is 3-(2,6-dichloro-3,5-dimethoxyphenyl)-1-(6-((4-(4-ethylpiperazin-1-yl)phenyl)amino)pyrimidin-4-yl)-1-methylurea (Compound A5) or a compound disclosed in U.S. Pat. No. 8,552,002.
  • a GITR antibody molecule is used in combination with Compound A5, or a compound as described in U.S. Pat. No. 8,552,002, to treat a disorder such as a digestive/gastrointestinal cancer, a hematological cancer, or a solid tumor.
  • the FGFR inhibitor or 3-(2,6-dichloro-3,5-dimethoxyphenyl)-1-(6-((4-(4-ethylpiperazin-1-yl)phenyl)amino)pyrimidin-4-yl)-1-methylurea is administered at a dose of about 100-125 mg (e.g., per day), e.g., about 100 mg or about 125 mg.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a PI3K inhibitor, Buparlisib (Compound A6), or a compound disclosed in PCT Publication No. WO 2007/084786, to treat a disorder, e.g., a disorder described herein.
  • the PI3K inhibitor is Buparlisib (Compound A6) or a compound disclosed in PCT Publication No. WO 2007/084786.
  • a GITR antibody molecule is used in combination with Buparlisib (Compound A6), or a compound disclosed in PCT Publication No.
  • WO 2007/084786 to treat a disorder such as, a prostate cancer, a non-small cell lung cancer, an endocrine cancer, a leukemia, an ovarian cancer, a melanoma, a bladder cancer, a breast cancer, a female reproductive system cancer, a digestive/gastrointestinal cancer, a colorectal cancer, a glioblastoma multiforme, a solid tumor, a non-Hodgkin lymphoma, a hematopoiesis disorder, or a head and neck cancer.
  • a disorder such as, a prostate cancer, a non-small cell lung cancer, an endocrine cancer, a leukemia, an ovarian cancer, a melanoma, a bladder cancer, a breast cancer, a female reproductive system cancer, a digestive/gastrointestinal cancer, a colorectal cancer, a glioblastoma multiforme, a solid tumor, a non-Hodgkin lymphoma, a hem
  • the PI3K inhibitor or Buparlisib (Compound A6) is administered at a dose of about 100 mg (e.g., per day).
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with an FGFR inhibitor, 8-(2,6-difluoro-3,5-dimethoxyphenyl)-N-(4-((dimethylamino)methyl)-1H-imidazol-2-yl)quinoxaline-5-carboxamide (Compound A7) or a compound disclosed in PCT Publication No. WO 2009/141386 to treat a disorder, e.g., a disorder described herein.
  • an FGFR inhibitor 8-(2,6-difluoro-3,5-dimethoxyphenyl)-N-(4-((dimethylamino)methyl)-1H-imidazol-2-yl)quinoxaline-5-carboxamide
  • Compound A7 8-(2,6-difluoro-3,5-dimethoxyphenyl)-N-(4-((dimethylamino)methyl
  • the FGFR inhibitor is 8-(2,6-difluoro-3,5-dimethoxyphenyl)-N-(4-((dimethylamino)methyl)-1H-imidazol-2-yl)quinoxaline-5-carboxamide(Compound A7) or a compound disclosed in a PCT Publication No. WO 2009/141386.
  • the FGFR inhibitor is 8-(2,6-difluoro-3,5-dimethoxyphenyl)-N-(4-((dimethylamino)methyl)-1H-imidazol-2-yl)quinoxaline-5-carboxamide(Compound A7).
  • a GITR antibody molecule is used in combination with 8-(2,6-difluoro-3,5-dimethoxyphenyl)-N-(4-((dimethylamino)methyl)-1H-imidazol-2-yl)quinoxaline-5-carboxamide(Compound A7), or a compound disclosed in PCT Publication No. WO 2009/141386, to treat a disorder such as a cancer characterized by angiogenesis.
  • the FGFR inhibitor or 8-(2,6-difluoro-3,5-dimethoxyphenyl)-N-(4-((dimethylamino)methyl)-1H-imidazol-2-yl)quinoxaline-5-carboxamide is administered at a dose of e.g., from approximately 3 mg to approximately 5 g, more preferably from approximately 10 mg to approximately 1.5 g per person per day, optionally divided into 1 to 3 single doses which may, for example, be of the same size.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a PI3K inhibitor, (S)—N1-(4-methyl-5-(2-(1,1,1-trifluoro-2-methylpropan-2-yl)pyridin-4-yl)thiazol-2-yl)pyrrolidine-1,2-dicarboxamide (Compound A8) or a compound disclosed PCT Publication No. WO 2010/029082 to treat a disorder, e.g., a disorder described herein.
  • a PI3K inhibitor e.g., (S)—N1-(4-methyl-5-(2-(1,1,1-trifluoro-2-methylpropan-2-yl)pyridin-4-yl)thiazol-2-yl)pyrrolidine-1,2-dicarboxamide
  • Compound A8 or a compound disclosed PCT Publication No. WO 2010/029082 to treat a disorder, e.g., a
  • the PI3K inhibitor is (S)—N1-(4-methyl-5-(2-(1,1,1-trifluoro-2-methylpropan-2-yl)pyridin-4-yl)thiazol-2-yl)pyrrolidine-1,2-dicarboxamide (Compound A8) or a compound disclosed PCT Publication No. WO 2010/029082.
  • a GITR antibody molecule is used in combination with (S)—N1-(4-methyl-5-(2-(1,1,1-trifluoro-2-methylpropan-2-yl)pyridin-4-yl)thiazol-2-yl)pyrrolidine-1,2-dicarboxamide (Compound A8), or a compound disclosed PCT Publication No. WO 2010/029082, to treat a disorder such as a gastric cancer, a breast cancer, a pancreatic cancer, a digestive/gastrointestinal cancer, a solid tumor, and a head and neck cancer.
  • the PI3K inhibitor or (S)—N1-(4-methyl-5-(2-(1,1,1-trifluoro-2-methylpropan-2-yl)pyridin-4-yl)thiazol-2-yl)pyrrolidine-1,2-dicarboxamide is administered at a dose of about 150-300, 200-300, 200-400, or 300-400 mg (e.g., per day), e.g., about 200, 300, or 400 mg.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with an inhibitor of cytochrome P450 (e.g., a CYP17 inhibitor) or a compound disclosed in PCT Publication No. WO 2010/149755, to treat a disorder, e.g., a disorder described herein.
  • the cytochrome P450 inhibitor e.g., the CYP17 inhibitor
  • a GITR antibody molecule is used in combination with a compound disclosed in PCT Publication No. WO 2010/149755, to treat prostate cancer.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with an HDM2 inhibitor, (S)-1-(4-chlorophenyl)-7-isopropoxy-6-methoxy-2-(4-(methyl(((1r,4S)-4-(4-methyl-3-oxopiperazin-1-yl)cyclohexyl)methyl)amino)phenyl)-1,2-dihydroisoquinolin-3(4H)-one(Compound A10) or a compound disclosed in PCT Publication No. WO 2011/076786 to treat a disorder, e.g., a disorder described herein).
  • an HDM2 inhibitor (S)-1-(4-chlorophenyl)-7-isopropoxy-6-methoxy-2-(4-(methyl(((1r,4S)-4-(4-methyl-3-oxopiperazin-1-yl)cyclohexyl)methyl)
  • the HDM2 inhibitor is (S)-1-(4-chlorophenyl)-7-isopropoxy-6-methoxy-2-(4-(methyl(((1r,4S)-4-(4-methyl-3-oxopiperazin-1-yl)cyclohexyl)methyl)amino)phenyl)-1,2-dihydroisoquinolin-3(4H)-one (Compound A10) or a compound disclosed in PCT Publication No. WO 2011/076786.
  • a GITR antibody molecule is used in combination with (S)-1-(4-chlorophenyl)-7-isopropoxy-6-methoxy-2-(4-(methyl(((1r,4S)-4-(4-methyl-3-oxopiperazin-1-yl)cyclohexyl)methyl)amino)phenyl)-1,2-dihydroisoquinolin-3(4H)-one (Compound A10), or a compound disclosed in PCTPublication No. WO 2011/076786, to treat a disorder such as a solid tumor.
  • the HDM2 inhibitor or (S)-1-(4-chlorophenyl)-7-isopropoxy-6-methoxy-2-(4-(methyl(((1r,4S)-4-(4-methyl-3-oxopiperazin-1-yl)cyclohexyl)methyl)amino)phenyl)-1,2-dihydroisoquinolin-3(4H)-one is administered at a dose of about 400 to 700 mg, e.g., administered three times weekly, 2 weeks on and one week off. In some embodiments, the dose is about 400, 500, 600, or 700 mg; about 400-500, 500-600, or 600-700 mg, e.g., administered three times weekly.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with an iron chelating agent, Deferasirox (also known as EXJADE; Compound A11), or a compound disclosed in PCT Publication No. WO 1997/049395 to treat a disorder, e.g., a disorder described herein.
  • an iron chelating agent Deferasirox (also known as EXJADE; Compound A11)
  • the iron chelating agent is Deferasirox or a compound disclosed in PCT Publication No. WO 1997/049395.
  • the iron chelating agent is Deferasirox (Compound A11).
  • a GITR antibody molecule is used in combination with Deferasirox (Compound A11), or a compound disclosed in PCT Publication No. WO 1997/049395, to treat iron overload, hemochromatosis, or myelodysplasia.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with an aromatase inhibitor, Letrozole (also known as FEMARA; Compound A12), or a compound disclosed in U.S. Pat. No. 4,978,672 to treat a disorder, e.g., a disorder described herein.
  • the aromatase inhibitor is Letrozole (Compound A12) or a compound disclosed in U.S. Pat. No. 4,978,672.
  • a GITR antibody molecule is used in combination with Letrozole (Compound A12), or a compound disclosed in U.S. Pat. No. 4,978,672, to treat a disorder such as a cancer, a leiomyosarcoma, an endometrium cancer, a breast cancer, a female reproductive system cancer, or a hormone deficiency.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein
  • a PI3K inhibitor e.g., a pan-PI3K inhibitor, (4S,5R)-3-(2′-amino-2-morpholino-4′-(trifluoromethyl)-[4,5′-bipyrimidin]-6-yl)-4-(hydroxymethyl)-5-methyloxazolidin-2-one (Compound A13) or a compound disclosed in PCT Publication No. WO2013/124826 to treat a disorder, e.g., a disorder described herein.
  • a PI3K inhibitor e.g., a pan-PI3K inhibitor, (4S,5R)-3-(2′-amino-2-morpholino-4′-(trifluoromethyl)-[4,5′-bipyrimidin]-6-yl)-4-(hydroxymethyl)-5-methyloxazolidin-2-one (Compound A13) or a compound disclosed in
  • the PI3K inhibitor is (4S,5R)-3-(2′-amino-2-morpholino-4′-(trifluoromethyl)-[4,5′-bipyrimidin]-6-yl)-4-(hydroxymethyl)-5-methyloxazolidin-2-one (Compound A13) or a compound disclosed in PCT Publication No. WO2013/124826.
  • a GITR antibody molecule is used in combination with (4S,5R)-3-(2′-amino-2-morpholino-4′-(trifluoromethyl)-[4,5′-bipyrimidin]-6-yl)-4-(hydroxymethyl)-5-methyloxazolidin-2-one (Compound A13), or a compound disclosed in PCT Publication No. WO2013/124826, to treat a disorder such as a cancer or an advanced solid tumor.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with an inhibitor of p53 and/or a p53/Mdm2 interaction, (S)-5-(5-chloro-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)-6-(4-chlorophenyl)-2-(2,4-dimethoxypyrimidin-5-yl)-1-isopropyl-5,6-dihydropyrrolo[3,4-d]imidazol-4(1H)-one (Compound A14), or a compound disclosed in PCT Publication No.
  • the p53 and/or a p53/Mdm2 interaction inhibitor is (S)-5-(5-chloro-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)-6-(4-chlorophenyl)-2-(2,4-dimethoxypyrimidin-5-yl)-1-isopropyl-5,6-dihydropyrrolo[3,4-d]imidazol-4(1H)-one (Compound A14) or a compound disclosed in PCT Publication No. WO2013/111105.
  • a GITR antibody molecule is used in combination with (S)-5-(5-chloro-1-methyl-2-oxo-1,2-dihydropyridin-3-yl)-6-(4-chlorophenyl)-2-(2,4-dimethoxypyrimidin-5-yl)-1-isopropyl-5,6-dihydropyrrolo[3,4-d]imidazol-4(1H)-one (Compound A14), or a compound disclosed in PCT Publication No. WO2013/111105, to treat a disorder such as a cancer or a soft tissue sarcoma.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a CSF-1R tyrosine kinase inhibitor, 4-((2-(((1R,2R)-2-hydroxycyclohexyl)amino)benzo[d]thiazol-6-yl)oxy)-N-methylpicolinamide (Compound A15), or a compound disclosed in PCT Publication No. WO 2005/073224 to treat a disorder, e.g., a disorder described herein.
  • a CSF-1R tyrosine kinase inhibitor 4-((2-(((1R,2R)-2-hydroxycyclohexyl)amino)benzo[d]thiazol-6-yl)oxy)-N-methylpicolinamide
  • Compound A15 4-((2-(((1R,2R)-2-hydroxycyclohexyl)amino)benzo[d]thiazol-6-y
  • the CSF-1R tyrosine kinase inhibitor is 4-((2-(((1R,2R)-2-hydroxycyclohexyl)amino)benzo[d]thiazol-6-yl)oxy)-N-methylpicolinamide (Compound A15) or a compound disclosed in PCT Publication No. WO 2005/073224.
  • a GITR antibody molecule is used in combination with 4-((2-(((1R,2R)-2-hydroxycyclohexyl)amino)benzo[d]thiazol-6-yl)oxy)-N-methylpicolinamide (Compound A15) or a compound disclosed in PCT Publication No. WO 2005/073224, to treat a disorder such as cancer.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with an apoptosis inducer and/or an angiogenesis inhibitor, such as Imatinib mesylate (also known as GLEEVEC; Compound A16) or a compound disclosed in PCT Publication No. WO1999/003854 to treat a disorder, e.g., a disorder described.
  • the apoptosis inducer and/or an angiogenesis inhibitor is Imatinib mesylate (Compound A16) or a compound disclosed in PCT Publication No. WO1999/003854.
  • a GITR antibody molecule is used in combination with Imatinib mesylate (Compound A16), or a compound disclosed in PCT Publication No. WO1999/003854, to treat a disorder such as a cancer, a multiple myeloma, a prostate cancer, a non-small cell lung cancer, a lymphoma, a gastric cancer, a melanoma, a breast cancer, a pancreatic cancer, a digestive/gastrointestinal cancer, a colorectal cancer, a glioblastoma multiforme, a liver cancer, a head and neck cancer, asthma, multiple sclerosis, allergy, Alzheimer's dementia, amyotrophic lateral sclerosis, or rheumatoid arthritis.
  • a disorder such as a cancer, a multiple myeloma, a prostate cancer, a non-small cell lung cancer, a lymphoma, a gastric cancer, a melanoma, a breast cancer,
  • Imatinib mesylate (Compound A16) is administered at a dose of about 100 to 1000 mg, e.g., about 200 mg to 800 mg, about 300 mg to 700 mg, or about 400 mg to 600 mg, e.g., about 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, or 700 mg.
  • the dosing schedule can vary from e.g., every other day to daily, twice or three times a day.
  • Imatinib mesylate is administered at an oral dose from about 100 mg to 600 mg daily, e.g., about 100 mg, 200 mg, 260 mg, 300 mg, 400 mg, or 600 mg daily.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a JAK inhibitor, 2-fluoro-N-methyl-4-(7-(quinolin-6-ylmethyl)imidazo[1,2-b][1,2,4]triazin-2-yl)benzamide (Compound A17), or a dihydrochloric salt thereof, or a compound disclosed in PCT Publication No. WO 2007/070514, to treat a disorder, e.g., a disorder described herein.
  • a JAK inhibitor 2-fluoro-N-methyl-4-(7-(quinolin-6-ylmethyl)imidazo[1,2-b][1,2,4]triazin-2-yl)benzamide
  • Compound A17 2-fluoro-N-methyl-4-(7-(quinolin-6-ylmethyl)imidazo[1,2-b][1,2,4]triazin-2-yl)benzamide
  • the JAK inhibitor is 2-fluoro-N-methyl-4-(7-(quinolin-6-ylmethyl)imidazo[1,2-b][1,2,4]triazin-2-yl)benzamide (Compound A17), or a dihydrochloric salt thereof, or a compound disclosed in PCT Publication No. WO 2007/070514.
  • a GITR antibody molecule is used in combination with 2-fluoro-N-methyl-4-(7-(quinolin-6-ylmethyl)imidazo[1,2-b][1,2,4]triazin-2-yl)benzamide (Compound A17), or a dihydrochloric salt thereof, or a compound disclosed in PCT Publication No. WO 2007/070514, to treat a disorder such as colorectal cancer, myeloid leukemia, hematological cancer, autoimmune disease, non-Hodgkin lymphoma, or thrombocythemia.
  • the JAK inhibitor or a 2-fluoro-N-methyl-4-(7-(quinolin-6-ylmethyl)imidazo[1,2-b][1,2,4]triazin-2-yl)benzamide (Compound A17), or a dihydrochloric salt thereof is administered at a dose of about 400-600 mg (e.g., per day), e.g., about 400, 500, or 600 mg, or about 400-500 or 500-600 mg.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a JAK inhibitor, Ruxolitinib Phosphate (also known as JAKAFI; Compound A18) or a compound disclosed in PCT Publication No. WO 2007/070514 to treat a disorder, e.g., a disorder described herein.
  • a JAK inhibitor is Ruxolitinib Phosphate (Compound A18) or a compound disclosed in PCT Publication No. WO 2007/070514.
  • a GITR antibody molecule is used in combination with Ruxolitinib Phosphate (Compound A18), or a compound disclosed in PCT Publication No. WO 2007/070514, to treat a disorder such as a prostate cancer, a lymphocytic leukemia, a multiple myeloma, a lymphoma, a lung cancer, a leukemia, cachexia, a breast cancer, a pancreatic cancer, rheumatoid arthritis, psoriasis, a colorectal cancer, a myeloid leukemia, a hematological cancer, an autoimmune disease, a non-Hodgkin lymphoma, or thrombocythemia.
  • a disorder such as a prostate cancer, a lymphocytic leukemia, a multiple myeloma, a lymphoma, a lung cancer, a leukemia, cachexia, a breast cancer, a pancreatic cancer,
  • the JAK inhibitor or Ruxolitinib Phosphate is administered at a dose of about 15-25 mg, e.g., twice daily. In some embodiments, the dose is about 15, 20, or 25 mg, or about 15-20 or 20-25 mg.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a deacetylase (DAC) inhibitor, Panobinostat (Compound A19), or a compound disclosed in PCT Publication No. WO 2014/072493 to treat a disorder, e.g., a disorder described herein.
  • the DAC inhibitor is Panobinostat (Compound A19) or a compound disclosed in PCT Publication No. WO 2014/072493.
  • a GITR antibody molecule is used in combination with Panobinostat (Compound A19), a compound disclosed in PCT Publication No.
  • WO 2014/072493 to treat a disorder such as a small cell lung cancer, a respiratory/thoracic cancer, a prostate cancer, a multiple myeloma, myelodysplastic syndrome, a bone cancer, a non-small cell lung cancer, an endocrine cancer, a lymphoma, a neurologic cancer, a leukemia, HIV/AIDS, an immune disorder, transplant rejection, a gastric cancer, a melanoma, a breast cancer, a pancreatic cancer, a colorectal cancer, a glioblastoma multiforme, a myeloid leukemia, a hematological cancer, a renal cancer, a non-Hodgkin lymphoma, a head and neck cancer, a hematopoiesis disorders, or a liver cancer.
  • a disorder such as a small cell lung cancer, a respiratory/thoracic cancer, a prostate cancer, a multiple myeloma, myelodysplastic
  • the DAC inhibitor or Panobinostat is administered at a dose of about 20 mg (e.g., per day).
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with an inhibitor of one or more of cytochrome P450 (e.g., 11B2), aldosterone or angiogenesis, Osilodrostat (Compound A20), or a compound disclosed in PCT Publication No. WO2007/024945 to treat a disorder, e.g., a disorder described herein.
  • the inhibitor of one or more of cytochrome P450 (e.g., 11B2), aldosterone or angiogenesis is Osilodrostat (Compound A20) or a compound disclosed in PCT Publication No. WO2007/024945.
  • a GITR antibody molecule is used in combination with Osilodrostat (Compound A20), or a compound disclosed in PCT Publication No. WO2007/024945, to treat a disorder such as Cushing's syndrome, hypertension, or heart failure therapy.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a IAP inhibitor, (S)—N—((S)-1-cyclohexyl-2-((S)-2-(4-(4-fluorobenzoyl)thiazol-2-yl)pyrrolidin-1-yl)-2-oxoethyl)-2-(methylamino)propanamide (Compound A21) or a compound disclosed in U.S. Pat. No. 8,552,003 to treat a disorder, e.g., a disorder described herein.
  • a disorder e.g., a disorder described herein.
  • the IAP inhibitor is (S)—N—((S)-1-cyclohexyl-2-((S)-2-(4-(4-fluorobenzoyl)thiazol-2-yl)pyrrolidin-1-yl)-2-oxoethyl)-2-(methylamino)propanamide (Compound A21) or a compound disclosed in U.S. Pat. No. 8,552,003.
  • a GITR antibody molecule is used in combination with (S)—N—((S)-1-cyclohexyl-2-((S)-2-(4-(4-fluorobenzoyl)thiazol-2-yl)pyrrolidin-1-yl)-2-oxoethyl)-2-(methylamino)propanamide (Compound A21), or a compound disclosed in U.S. Pat. No. 8,552,003, to treat a disorder such as a multiple myeloma, a breast cancer, an ovarian cancer, a pancreatic cancer, or a hematopoiesis disorder.
  • the IAP inhibitor or (S)—N—((S)-1-cyclohexyl-2-((S)-2-(4-(4-fluorobenzoyl)thiazol-2-yl)pyrrolidin-1-yl)-2-oxoethyl)-2-(methylamino)propanamide (Compound A21) or a compound disclosed in U.S. Pat. No. 8,552,003 is administered at a dose of approximately 1800 mg, e.g., once weekly.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination a Smoothened (SMO) inhibitor, Sonidegib phosphate (Compound A22), (R)-2-(5-(4-(6-benzyl-4,5-dimethylpyridazin-3-yl)-2-methylpiperazin-1-yl)pyrazin-2-yl)propan-2-ol (Compound A25), or a compound disclosed in PCT Publication No. WO 2007/131201 or WO 2010/007120 to treat a disorder, e.g., a disorder described herein.
  • SMO Smoothened
  • Compound A22 Sonidegib phosphate
  • Compound A25 a compound disclosed in PC
  • the SMO inhibitor is Sonidegib phosphate (Compound A22), (R)-2-(5-(4-(6-benzyl-4,5-dimethylpyridazin-3-yl)-2-methylpiperazin-1-yl)pyrazin-2-yl)propan-2-ol (Compound A25), or a compound disclosed in PCT Publication No. WO 2007/131201 or WO 2010/007120.
  • a GITR antibody molecule is used in combination with Sonidegib phosphate (Compound A22), (R)-2-(5-(4-(6-benzyl-4,5-dimethylpyridazin-3-yl)-2-methylpiperazin-1-yl)pyrazin-2-yl)propan-2-ol (Compound A25), or a compound disclosed in PCT Publication No. WO 2007/131201 or WO 2010/007120 to treat a disorder such as a cancer, a medulloblastoma, a small cell lung cancer, a prostate cancer, a basal cell carcinoma, a pancreatic cancer, or an inflammation.
  • a disorder such as a cancer, a medulloblastoma, a small cell lung cancer, a prostate cancer, a basal cell carcinoma, a pancreatic cancer, or an inflammation.
  • Sonidegib phosphate (Compound A22) is administered at a dose of about 20 to 500 mg, e.g., about 40 mg to 400 mg, about 50 mg to 300 mg, or about 100 mg to 200 mg, e.g., about 50 mg, 100 mg, 150 mg, 200 mg, 250 mg, or 300 mg.
  • the dosing schedule can vary from e.g., every other day to daily, twice or three times a day.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with an Alk inhibitor, ceritinib (also known as ZYKADIA; Compound A23) or a compound disclosed in PCT Publication No. WO 2007/131201 to treat a disorder, e.g., a disorder described herein.
  • the Alk inhibitor is ceritinib (Compound A23) or a compound disclosed in PCT Publication No. WO 2007/131201.
  • a GITR antibody molecule is used in combination with ceritinib (Compound A23), or a compound disclosed in PCT Publication No. WO 2007/131201, to treat a disorder such as non-small cell lung cancer or solid tumors.
  • the Alk inhibitor or ceritinib (Compound A23) is administered at a dose of approximately 750 mg, e.g., once daily.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a JAK and/or CDK4/6 inhibitor, 7-cyclopentyl-N,N-dimethyl-2-((5-(piperazin-1-yl)pyridin-2-yl)amino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxamide (Compound A24), or a compound disclosed in U.S. Pat. No. 8,415,355 or U.S. Pat. No. 8,685,980 to treat a disorder, e.g., a disorder described herein.
  • a JAK and/or CDK4/6 inhibitor 7-cyclopentyl-N,N-dimethyl-2-((5-(piperazin-1-yl)pyridin-2-yl)amino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxamide
  • Compound A24 7-cyclopentyl
  • the JAK and/or CDK4/6 inhibitor is 7-cyclopentyl-N,N-dimethyl-2-((5-(piperazin-1-yl)pyridin-2-yl)amino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxamide (Compound A24) or a compound disclosed in U.S. Pat. No. 8,415,355 or U.S. Pat. No. 8,685,980.
  • a GITR antibody molecule is used in combination with 7-cyclopentyl-N,N-dimethyl-2-((5-(piperazin-1-yl)pyridin-2-yl)amino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxamide (Compound A24), or a compound disclosed in U.S. Pat. No. 8,415,355 or U.S. Pat. No. 8,685,980, to treat a disorder such as a lymphoma, a neurologic cancer, a melanoma, a breast cancer, or a solid tumor.
  • a disorder such as a lymphoma, a neurologic cancer, a melanoma, a breast cancer, or a solid tumor.
  • the JAK and/or CDK4/6 inhibitor or 7-cyclopentyl-N,N-dimethyl-2-((5-(piperazin-1-yl)pyridin-2-yl)amino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxamide is administered at a dose of approximately 200-600 mg, e.g., per day. In one embodiment, the compound is administered at a dose of about 200, 300, 400, 500, or 600 mg, or about 200-300, 300-400, 400-500, or 500-600 mg.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination a prolactin receptor (PRLR) inhibitor, a human monoclonal antibody molecule (Compound A26) as disclosed in U.S. Pat. No. 7,867,493), to treat a disorder, e.g., a disorder described herein.
  • the PRLR inhibitor is a human monoclonal antibody (Compound A26) disclosed in U.S. Pat. No. 7,867,493.
  • a GITR antibody molecule is used in combination with human monoclonal antibody molecule (Compound A26) described in U.S. Pat. No. 7,867,493 to treat a disorder such as, a cancer, a prostate cancer, or a breast cancer.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a PIM Kinase inhibitor, N-(4-((1R,3S,5S)-3-amino-5-methylcyclohexyl)pyridin-3-yl)-6-(2,6-difluorophenyl)-5-fluoropicolinamide (Compound A27) or a compound disclosed in PCT Publication No. WO 2010/026124 to treat a disorder, e.g., a disorder described herein.
  • a PIM Kinase inhibitor N-(4-((1R,3S,5S)-3-amino-5-methylcyclohexyl)pyridin-3-yl)-6-(2,6-difluorophenyl)-5-fluoropicolinamide
  • Compound A27 N-(4-((1R,3S,5S)-3-amino-5-methylcyclohex
  • the PIM Kinase inhibitor is N-(4-((1R,3S,5S)-3-amino-5-methylcyclohexyl)pyridin-3-yl)-6-(2,6-difluorophenyl)-5-fluoropicolinamide (Compound A27) or a compound disclosed in PCT Publication No. WO 2010/026124.
  • a GITR antibody molecule is used in combination with N-(4-((1R,3S,5S)-3-amino-5-methylcyclohexyl)pyridin-3-yl)-6-(2,6-difluorophenyl)-5-fluoropicolinamide (Compound A27), or a compound disclosed in PCT Publication No. WO 2010/026124, to treat a disorder such as a multiple myeloma, myelodysplastic syndrome, a myeloid leukemia, or a non-Hodgkin lymphoma.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination a Wnt signaling inhibitor, 2-(2′,3-dimethyl-[2,4′-bipyridin]-5-yl)-N-(5-(pyrazin-2-yl)pyridin-2-yl)acetamide (Compound A28) or a compound disclosed in PCT publication No. WO 2010/101849 to treat a disorder, e.g., a disorder described herein.
  • a Wnt signaling inhibitor 2-(2′,3-dimethyl-[2,4′-bipyridin]-5-yl)-N-(5-(pyrazin-2-yl)pyridin-2-yl)acetamide
  • Compound A28 2-(2′,3-dimethyl-[2,4′-bipyridin]-5-yl)-N-(5-(pyrazin-2-yl)pyridin-2-yl)ace
  • the Wnt signaling inhibitor is 2-(2′,3-dimethyl-[2,4′-bipyridin]-5-yl)-N-(5-(pyrazin-2-yl)pyridin-2-yl)acetamide (Compound A28) or a compound disclosed in PCT publication No. WO 2010/101849. In one embodiment, the Wnt signaling inhibitor is 2-(2′,3-dimethyl-[2,4′-bipyridin]-5-yl)-N-(5-(pyrazin-2-yl)pyridin-2-yl)acetamide (Compound A28).
  • a GITR antibody molecule is used in combination with 2-(2′,3-dimethyl-[2,4′-bipyridin]-5-yl)-N-(5-(pyrazin-2-yl)pyridin-2-yl)acetamide (Compound A28), or a compound disclosed in PCT publication No. WO 2010/101849, to treat a disorder such as a solid tumor (e.g., a head and neck cancer, a squamous cell carcinoma, a breast cancer, a pancreatic cancer, or a colon cancer).
  • a solid tumor e.g., a head and neck cancer, a squamous cell carcinoma, a breast cancer, a pancreatic cancer, or a colon cancer.
  • 2-(2′,3-dimethyl-[2,4′-bipyridin]-5-yl)-N-(5-(pyrazin-2-yl)pyridin-2-yl)acetamide (Compound A28) is administered at a dose of about 1 to 50 mg, e.g., about 2 mg to 45 mg, about 3 mg to 40 mg, about 5 mg to 35 mg, 5 mg to 10 mg, or about 10 mg to 30 mg, e.g., about 2 mg, 5 mg, 10 mg, 20 mg, 30 mg, or 40 mg.
  • the dosing schedule can vary from e.g., every other day to daily, twice or three times a day.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a BRAF inhibitor, Encorafenib (Compound A29), or a compound disclosed in PCT Publication No. WO 2011/025927 to treat a disorder, e.g., a disorder described herein.
  • the BRAF inhibitor is Encorafenib (Compound A29) or a compound disclosed in PCT Publication No. WO 2011/025927.
  • a GITR antibody molecule is used in combination with Encorafenib (Compound A29), or a compound disclosed in PCT Publication No. WO 2011/025927, to treat a disorder such as a non-small cell lung cancer, a melanoma, or a colorectal cancer.
  • the BRAF inhibitor or Encorafenib (Compound A29) is administered at a dose of about 200-300, 200-400, or 300-400 mg, e.g., per day. In one embodiment, the compound is administered at a dose of about 200, about 300 or about 400 mg.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination a CDK4/6 inhibitor, 7-cyclopentyl-N,N-dimethyl-2-((5-((1R,6S)-9-methyl-4-oxo-3,9-diazabicyclo[4.2.1]nonan-3-yl)pyridin-2-yl)amino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxamide (Compound A30), or a compound disclosed in PCT publication No. WO 2011/101409 to treat a disorder, e.g., a disorder described herein.
  • a CDK4/6 inhibitor 7-cyclopentyl-N,N-dimethyl-2-((5-((1R,6S)-9-methyl-4-oxo-3,9-diazabicyclo[4.2.1]nonan-3-yl)pyridin-2-yl)amino)-7H-
  • the CDK4/6 inhibitor is 7-cyclopentyl-N,N-dimethyl-2-((5-((1R,6S)-9-methyl-4-oxo-3,9-diazabicyclo[4.2.1]nonan-3-yl)pyridin-2-yl)amino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxamide (Compound A30) or a compound disclosed in PCT publication No. WO 2011/101409.
  • a GITR antibody molecule is used in combination with 7-cyclopentyl-N,N-dimethyl-2-((5-((1R,6S)-9-methyl-4-oxo-3,9-diazabicyclo[4.2.1]nonan-3-yl)pyridin-2-yl)amino)-7H-pyrrolo[2,3-d]pyrimidine-6-carboxamide (Compound A30), or a compound disclosed in PCT publication No.
  • WO 2011/101409 to treat a disorder such as a cancer, a mantle cell lymphoma, a liposarcoma, a non-small cell lung cancer, a melanoma, a squamous cell esophageal cancer, or a breast cancer.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a HER3 inhibitor, Compound A31, or a compound disclosed in PCT Publication No. WO 2012/022814, to treat a disorder, e.g., a disorder described herein.
  • the HER3 inhibitor is Compound A31 or a compound disclosed in PCT Publication WO 2012/022814.
  • a GITR antibody molecule is used in combination with Compound A31, or a compound disclosed in PCT Publication WO 2012/022814, to treat a disorder such as a gastric cancer, an esophageal cancer, a head and neck cancer, a squamous cell carcinoma, a stomach cancer, a breast cancer (e.g., metastatic breast cancer), or a digestive/gastrointestinal cancer.
  • a disorder such as a gastric cancer, an esophageal cancer, a head and neck cancer, a squamous cell carcinoma, a stomach cancer, a breast cancer (e.g., metastatic breast cancer), or a digestive/gastrointestinal cancer.
  • Compound A31 is a human monoclonal antibody molecule.
  • the HER3 inhibitor or Compound A31 is administered at a dose of about 3, 10, 20, or 40 mg/kg, e.g., once weekly (QW). In one embodiment, the compound is administered at a dose of about 3-10, 10-20, or 20-40 mg/kg.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination an FGFR2 and/or FGFR4 inhibitor, Compound A32, or a compound disclosed in a publication PCT Publication No. WO 2014/160160 (e.g., an antibody molecule drug conjugate against an FGFR2 and/or FGFR4, e.g., mAb 12425), to treat a disorder, e.g., a disorder described herein.
  • the FGFR2 and/or FGFR4 inhibitor is Compound A32 or a compound disclosed in a publication PCT Publication No. WO 2014/160160.
  • a GITR antibody molecule is used in combination with Compound A32, or a compound as described in Table 6, to treat a disorder such as a cancer, a gastric cancer, a breast cancer, a rhabdomyosarcoma, a liver cancer, an adrenal cancer, a lung cancer, an esophageal cancer, a colon cancer, or an endometrial cancer.
  • a disorder such as a cancer, a gastric cancer, a breast cancer, a rhabdomyosarcoma, a liver cancer, an adrenal cancer, a lung cancer, an esophageal cancer, a colon cancer, or an endometrial cancer.
  • Compound A32 is an antibody molecule drug conjugate against an FGFR2 and/or FGFR4, e.g., mAb 12425.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination an M-CSF inhibitor, Compound A33, or a compound disclosed in PCT Publication No. WO 2004/045532 (e.g., an antibody molecule or Fab fragment against M-CSF), to treat a disorder, e.g., a disorder described herein.
  • a disorder e.g., a disorder described herein.
  • the M-CSF inhibitor is Compound A33 or a compound disclosed in PCT Publication No. WO 2004/045532.
  • a GITR antibody molecule is used in combination with Compound A33, or a compound as described in PCT Publication No. WO 2004/045532, to treat a disorder such as a cancer, a prostate cancer, a breast cancer, or pigmented villonodular synovitis (PVNS).
  • Compound A33 is a monoclonal antibody molecule against M-CSF or a fragment (e.g., Fab fragment) thereof.
  • the M-CSF inhibitor or Compound A33 is administered at an average dose of about 10 mg/kg.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a MEK inhibitor, Binimetinib (Compound A34), or a compound disclosed in PCT Publication No. WO 2003/077914 to treat a disorder, e.g., a disorder described herein.
  • the MEK inhibitor is Binimetinib (Compound A34), or a compound disclosed in PCT Publication No. WO 2003/077914.
  • a GITR antibody molecule is used in combination with Binimetinib (Compound A34), or a compound disclosed in PCT Publication No.
  • WO 2003/077914 to treat a disorder such as a non-small cell lung cancer, a multisystem genetic disorder, a melanoma, an ovarian cancer, a digestive/gastrointestinal cancer, a rheumatoid arthritis, or a colorectal cancer.
  • the MEK inhibitor or Binimetinib (Compound A34) is administered at a dose of about 45 mg, e.g., twice daily.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination an inhibitor of one or more of c-KIT, histamine release, Flt3 (e.g., FLK2/STK1) or PKC, Midostaurin (Compound A35) or a compound disclosed in PCT Publication No. WO 2003/037347 to treat a disorder, e.g., a disorder described herein.
  • the inhibitor is Midostaurin (Compound A35) or compound disclosed in PCT Publication No. WO 2003/037347.
  • the inhibitor of one or more of c-KIT, histamine release, Flt3 (e.g., FLK2/STK1) or PKC is Midostaurin.
  • a GITR antibody molecule is used in combination with Midostaurin (Compound A35), or compound disclosed in PCT Publication No. WO 2003/037347, to treat a disorder such as a cancer, a colorectal cancer, a myeloid leukemia, myelodysplastic syndrome, an age-related mascular degeration, a diabetic complication, or a dermatologic disorder.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a TOR inhibitor (e.g., mTOR inhibitor), Everolimus (also known as AFINITOR; Compound A36) or a Compound disclosed in PCT Publication No. WO 2014/085318 to treat a disorder, e.g., a disorder described herein).
  • a TOR inhibitor e.g., mTOR inhibitor
  • Everolimus also known as AFINITOR; Compound A36
  • a Compound disclosed in PCT Publication No. WO 2014/085318 to treat a disorder, e.g., a disorder described herein.
  • the TOR inhibitor is Everolimus (Compound A36) or a Compound disclosed in PCT Publication No. WO 2014/085318.
  • a GITR antibody molecule is used in combination with Everolimus (Compound A36) to treat a disorder such as an interstitial lung disease, a small cell lung cancer, a respiratory/thoracic cancer, a prostate cancer, a multiple myeloma, a sarcoma, an age-related macular degeneration, a bone cancer, tuberous sclerosis, a non-small cell lung cancer, an endocrine cancer, a lymphoma, a neurologic disorders, an astrocytoma, a cervical cancer, a neurologic cancer, a leukemia, an immune disorders, transplant rejection, a gastric cancer, a melanoma, epilepsy, a breast cancer, or a bladder cancer.
  • a disorder such as an interstitial lung disease, a small cell lung cancer, a respiratory/thoracic cancer, a prostate cancer, a multiple myeloma, a sarcoma, an age-related macular degeneration, a bone cancer, tuberous
  • the TOR inhibitor or Everolimusis administered at a dose of about 2.5-20 mg/day. In one embodiment, the compound is administered at a dose of about 2.5, 5, 10, or 20 mg/day, e.g., about 2.5-5, 5-10, or 10-20 mg/day.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination an inhibitor of one or more of VEGFR-2, PDGFRbeta, KIT or Raf kinase C, 1-methyl-5-((2-(5-(trifluoromethyl)-1H-imidazol-2-yl)pyridin-4-yl)oxy)-N-(4-(trifluoromethyl)phenyl)-1H-benzo[d]imidazol-2-amine (Compound A37) or a compound disclosed in PCT Publication No. WO 2007/030377 to treat a disorder, e.g., a disorder described herein.
  • the inhibitor of one or more of VEGFR-2, PDGFRbeta, KIT or Raf kinase C is 1-methyl-5-((2-(5-(trifluoromethyl)-1H-imidazol-2-yl)pyridin-4-yl)oxy)-N-(4-(trifluoromethyl)phenyl)-1H-benzo[d]imidazol-2-amine (Compound A37) or a compound disclosed in PCT Publication No. WO 2007/030377.
  • a GITR antibody molecule is used in combination with 1-methyl-5-((2-(5-(trifluoromethyl)-1H-imidazol-2-yl)pyridin-4-yl)oxy)-N-(4-(trifluoromethyl)phenyl)-1H-benzo[d]imidazol-2-amine (Compound A37), or a compound disclosed in PCT Publication No. WO 2007/030377, to treat a disorder such as a cancer, a melanoma, or a solid tumor.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination a somatostatin agonist and/or growth hormone release inhibitor, Pasireotide diaspartate (also known as SIGNIFOR; Compound A38) or a compound disclosed in PCT Publication No. WO2002/010192 or U.S. Pat. No. 7,473,761 to treat a disorder, e.g., a disorder described herein.
  • the somatostatin agonist and/or growth hormone release inhibitor is Pasireotide diaspartate (Compound A38) or a compound disclosed in PCT Publication No. WO2002/010192 or U.S. Pat. No.
  • a GITR antibody molecule is used in combination with Pasireotide diaspartate (Compound A38), or a compound disclosed in PCT Publication No. WO2002/010192 or U.S. Pat. No. 7,473,761, to treat a disorder such as a prostate cancer, an endocrine cancer, a nurologic cancer, a skin cancer (e.g., a melanoma), a pancreatic cancer, a liver cancer, Cushing's syndrome, a gastrointestinal disorder, acromegaly, a liver and biliary tract disorder, or liver cirrhosis.
  • a disorder such as a prostate cancer, an endocrine cancer, a nurologic cancer, a skin cancer (e.g., a melanoma), a pancreatic cancer, a liver cancer, Cushing's syndrome, a gastrointestinal disorder, acromegaly, a liver and biliary tract disorder, or liver cirrhosis.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination a signal transduction modulator and/or angiogenesis inhibitor, Dovitinib (Compound A39) or a compound disclosed in PCT Publication No. WO 2009/115562 to treat a disorder, e.g., a disorder described herein.
  • the signal transduction modulator and/or angiogenesis inhibitor is Dovitinib (Compound A39) or a compound disclosed in PCT Publication No. WO 2009/115562.
  • a GITR antibody molecule is used in combination with Dovitinib (Compound A39), or a compound disclosed in PCT Publication No.
  • WO 2009/115562 to treat a disorder such as a cancer, a respiratory/thoracic cancer, a multiple myeloma, a prostate cancer, a non-small cell lung cancer, an endocrine cancer, or a neurological genetic disorder.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with an EGFR inhibitor, (R,E)-N-(7-chloro-1-(1-(4-(dimethylamino)but-2-enoyl)azepan-3-yl)-1H-benzo[d]imidazol-2-yl)-2-methylisonicotinamide (Compound A40) or a compound disclosed in PCT Publication No. WO 2013/184757 to treat a disorder, e.g., a disorder described herein.
  • an EGFR inhibitor (R,E)-N-(7-chloro-1-(1-(4-(dimethylamino)but-2-enoyl)azepan-3-yl)-1H-benzo[d]imidazol-2-yl)-2-methylisonicotinamide
  • Compound A40 or a compound disclosed in PCT Publication No. WO 2013/1847
  • the EGFR inhibitor is (R,E)-N-(7-chloro-1-(1-(4-(dimethylamino) but-2-enoyl)azepan-3-yl)-1H-benzo[d]imidazol-2-yl)-2-methylisonicotinamide (Compound A40) or a compound disclosed in PCT Publication No. WO 2013/184757.
  • a GITR antibody molecule is used in combination with (R,E)-N-(7-chloro-1-(1-(4-(dimethylamino)but-2-enoyl)azepan-3-yl)-1H-benzo[d]imidazol-2-yl)-2-methylisonicotinamide (Compound A40), or a compound disclosed in PCT Publication No. WO 2013/184757, to treat a disorder such as a cancer, e.g., a solid tumor.
  • the EGFR inhibitor or (R,E)-N-(7-chloro-1-(1-(4-(dimethylamino)but-2-enoyl)azepan-3-yl)-1H-benzo[d]imidazol-2-yl)-2-methylisonicotinamide is administered at a dose of 150-250 mg, e.g., per day. In one embodiment, the compound is administered at a dose of about 150, 200, or 250 mg, or about 150-200 or 200-250 mg.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination an ALK inhibitor, N 6 -(2-isopropoxy-5-methyl-4-(1-methylpiperidin-4-yl)phenyl)-N 4 -(2-(isopropylsulfonyl)phenyl)-1H-pyrazolo[3,4-d]pyrimidine-4,6-diamine (Compound A42) or a compound disclosed in PCT Publication No. WO 2008/073687 to treat a disorder, e.g., a disorder described herein.
  • a disorder e.g., a disorder described herein.
  • the ALK inhibitor is N 6 -(2-isopropoxy-5-methyl-4-(1-methylpiperidin-4-yl)phenyl)-N 4 -(2-(isopropylsulfonyl)phenyl)-1H-pyrazolo[3,4-d]pyrimidine-4,6-diamine (Compound A42) or a compound disclosed in PCT Publication No. WO 2008/073687.
  • a GITR antibody molecule is used in combination with N 6 -(2-isopropoxy-5-methyl-4-(1-methylpiperidin-4-yl)phenyl)-N 4 -(2-(isopropylsulfonyl)phenyl)-1H-pyrazolo[3,4-d]pyrimidine-4,6-diamine (Compound A42), or a compound disclosed in PCT Publication No. WO 2008/073687, to treat a disorder such as a cancer, an anaplastic large-cell lymphoma (ALCL), a non-small cell lung carcinoma (NSCLC), or a neuroblastoma.
  • ACL anaplastic large-cell lymphoma
  • NSCLC non-small cell lung carcinoma
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination an IGF-1R inhibitor, 3-(4-(4-((5-chloro-4-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-2-yl)amino)-5-fluoro-2-methylphenyl)piperidin-1-yl)thietane 1,1-dioxide (Compound A43), 5-chloro-N 2 -(2-fluoro-5-methyl-4-(1-(tetrahydro-2H-pyran-4-yl)piperidin-4-yl)phenyl)-N 4 -(5-methyl-1H-pyrazol-3-yl)pyrimidine-2,4-diamine (Compound A44), or 5-chloro-N2-(4-(1-ethylpiperidin-4-yl)-2-fluoro-5-methylphenyl)-N 4
  • the IGF-1R inhibitor is 3-(4-(4-((5-chloro-4-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-2-yl)amino)-5-fluoro-2-methylphenyl)piperidin-1-yl)thietane 1,1-dioxide (Compound A43), 5-chloro-N 2 -(2-fluoro-5-methyl-4-(1-(tetrahydro-2H-pyran-4-yl)piperidin-4-yl)phenyl)-N 4 -(5-methyl-1H-pyrazol-3-yl)pyrimidine-2,4-diamine (Compound A44), 5-chloro-N2-(4-(1-ethylpiperidin-4-yl)-2-fluoro-5-methylphenyl)-N 4 -(5-methyl-1H
  • a GITR antibody molecule is used in combination with 3-(4-(4-((5-chloro-4-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-2-yl)amino)-5-fluoro-2-methylphenyl)piperidin-1-yl)thietane 1,1-dioxide (Compound A43), 5-chloro-N 2 -(2-fluoro-5-methyl-4-(1-(tetrahydro-2H-pyran-4-yl)piperidin-4-yl)phenyl)-N 4 -(5-methyl-1H-pyrazol-3-yl)pyrimidine-2,4-diamine (Compound A44), 5-chloro-N2-(4-(1-ethylpiperidin-4-yl)-2-fluoro-5-methylphenyl)-N 4 -(5-methyl-1H-pyrazol-3-yl)pyrimidine-2,4-diamine (Com
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination a P-Glycoprotein 1 inhibitor, Valspodar (also known as AMDRAY; Compound A46) or a compound disclosed in EP 296122 to treat a disorder, e.g., a disorder described herein.
  • the P-Glycoprotein 1 inhibitor is Valspodar (Compound A46) or a compound disclosed in EP 296122.
  • a GITR antibody molecule is used in combination with Valspodar (Compound A46), or a compound disclosed in EP 296122, to treat a disorder such as a cancer or a drug-resistant tumor.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination one or more of a VEGFR inhibitor, Vatalanib succinate (Compound A47) or a compound disclosed in EP 296122 to treat a disorder, e.g., a disorder described herein.
  • the VEGFR inhibitor is Vatalanib succinate (Compound A47) or a compound disclosed in EP 296122.
  • a GITR antibody molecule is used in combination with Vatalanib succinate (Compound A47), or a compound disclosed in EP 296122, to treat cancer.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with an IDH inhibitor or a compound disclosed in WO2014/141104 to treat a disorder, e.g., a disorder described herein.
  • the IDH inhibitor is a compound disclosed in PCT Publication No.
  • a GITR antibody molecule is used in combination with a compound disclosed in WO2014/141104 to treat a disorder such as a cancer.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a BCL-ABL inhibitor or a compound disclosed in PCT Publication No. WO2013/171639, WO2013/171640, WO2013/171641, or WO2013/171642 to treat a disorder, e.g., a disorder described herein.
  • the BCL-ABL inhibitor is a compound disclosed in PCT Publication No. WO2013/171639, WO2013/171640, WO2013/171641, or WO2013/171642.
  • a GITR antibody molecule is used in combination with a compound disclosed in PCT Publication No. WO2013/171639, WO2013/171640, WO2013/171641, or WO2013/171642 to treat a disorder such as a cancer.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with a c-RAF inhibitor or a compound disclosed in PCT Publication No. WO2014/151616 to treat a disorder, e.g., a disorder described herein.
  • the c-RAF inhibitor is Compound A50 or a compound disclosed in PCT Publication No. WO2014/151616.
  • a GITR antibody molecule is used in combination with a compound disclosed in PCT Publication No. WO2014/151616 to treat a disorder such as a cancer.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is used in combination with an ERK1/2 ATP competitive inhibitor or a compound disclosed in International Patent Application No. PCT/US2014/062913 to treat a disorder, e.g., a disorder described herein.
  • the ERK1/2 ATP competitive inhibitor is a compound disclosed in PCT Publication No. WO2015/066188.
  • a GITR antibody molecule is used in combination with Compound A51 or a compound disclosed in PCT Publication No. WO2015/066188 to treat a disorder such as a cancer.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is administerd in combination with one or more agents selected from, Compound A8, Compound A17, Compound A23, Compound A24, Compound A27, Compound A29, and Compound A33.
  • the combination e.g., a combination comprising an anti-GITR antibody molecule as described herein, is administered in combination with an anti-cancer agent having a known activity in an immune cell assay, e.g., in one or more of a huMLR assay, a T cell proliferation assay, and a B-cell proliferation assay. Exemplary assays are described below. Based on the assay, an IC50 for can be calculated for each test agent.
  • the anti-cancer agent has an IC50 of, e.g., 0-1 ⁇ M, 1-4 ⁇ M, or greater than 4 ⁇ M, e.g., 4-10 ⁇ M or 4-20 ⁇ M.
  • the second therapeutic agent is chosen from one or more of: Compound A9, Compound A16, Compound A17, Compound A21, Compound A22, Compound A25, Compound A28, Compound A48, and Compound 49.
  • the Compound A28 (or a compound related to Compound A28) is administered at a dose of approximately 5-10 or 10-30 mg. In some embodiments, the Compound A22 (or compound related to Compound A22) is administered at a dose of about 200 mg. In some embodiments, the Compound A17 (or compound related to Compound A17) is administered at a dose of approximately 400-600 mg. In some embodiments, the Compound A16 (or compound related to Compound A16) is administered at a dose of approximately 400-600 mg PO qDay. In some embodiments, the Compound A29 (or compound related to Compound A29) is administered at a dose of approximately 200-400 or 300-400 mg.
  • the Compound A24 (or compound related to Compound A24) is administered at a dose of approximately 200-600 mg. In some embodiments, the Compound A23 (ceritinib) (or compound related to ceritinib) is administered at a dose of approximately 750 mg once daily. In some embodiments, the Compound A8 (or compound related to Compound A8) is administered at a dose of approximately 200-400 or 300-400 mg. In some embodiments, the Compound A5 (or compound related to Compound A5) is administered at a dose of approximately 100-125 mg. In some embodiments, the Compound A6 (or compound related to Compound A6) is administered at a dose of about 100 mg.
  • the Compound A1 (or compound related to Compound A1) is administered at a dose of approximately 200-300 or 200-600 mg. In some embodiments, the Compound A40 (or compound related to Compound A40) is administered at a dose of approximately 150-250 mg. In embodiments, the Compound A10 (or compound related to Compound A10) is administered at a dose of approximately 400 to 700 mg, e.g., administered three times weekly, 2 weeks on and one week off. In embodiments, the BCR-ABL inhibitor is administered at a dose of approximately 20 mg bid-80 mg bid.
  • compositions comprising a combination of two, three or more therapeutic agents chosen from one, two, or all of the following categories (i)-(iii): (i) an agent that enhances antigen presentation (e.g., tumor antigen presentation); (ii) an agent that enhances an effector cell response (e.g., B cell and/or T cell activation and/or mobilization); or (iii) an agent that decreases tumor immunosuppression, wherein at least one therapeutic agent is a GITR antibody, antibody fragment or antigen binding agent provided herein.
  • an agent that enhances antigen presentation e.g., tumor antigen presentation
  • an agent that enhances an effector cell response e.g., B cell and/or T cell activation and/or mobilization
  • an agent that decreases tumor immunosuppression wherein at least one therapeutic agent is a GITR antibody, antibody fragment or antigen binding agent provided herein.
  • therapeutic approaches that enhance anti-tumor immunity work more effectively when the immune response is optimized by targeting multiple components at one or more stages of an immune response, e.g., an anti-tumor immune response.
  • approaches that enhance antigen presentation e.g., by activation and/or maturation of dendritic cells
  • approaches that enhance cellular and humoral immune responses e.g., by stimulating, e.g., disinhibiting, phagocytes and/or tumor infiltrating lymphocytes (e.g., NK cells and T cells)
  • tumor immunosuppressive signaling e.g., by increasing macrophage polarization, increasing T reg depletion and/or decreasing myeloid-derived suppressive cells (MDSCs)
  • MDSCs myeloid-derived suppressive cells
  • combination therapies that optimize one, two, or all of: (i) antigen presentation, e.g., increasing antigen presentation (e.g., by enhancing one or more of dendritic cell activity or maturation, antigen uptake, or antigen processing); (ii) effector cell response, e.g., increasing effector cell response (e.g., enhancing B cell and/or T cell activation and/or mobilization, e.g., in the lymph node); or (iii) tumor immunosuppression, e.g., decreasing tumor immunosuppression (e.g., increasing T cell infiltration and tumor cell killing).
  • antigen presentation e.g., increasing antigen presentation (e.g., by enhancing one or more of dendritic cell activity or maturation, antigen uptake, or antigen processing)
  • effector cell response e.g., increasing effector cell response (e.g., enhancing B cell and/or T cell activation and/or mobilization, e.g., in the lymph
  • the combinations described herein can provide a superior beneficial effect, e.g., in the treatment of a disorder, such as an enhanced anti-cancer effect, reduced toxicity and/or reduced side effects, compared to monotherapy administration of the therapeutic agents in the combination.
  • a superior beneficial effect e.g., in the treatment of a disorder, such as an enhanced anti-cancer effect, reduced toxicity and/or reduced side effects, compared to monotherapy administration of the therapeutic agents in the combination.
  • one or more of the therapeutic agents in the combination can be administered at a lower dosage, or for a shorter period of administration, than would be required to achieve the same therapeutic effect compared to the monotherapy administration.
  • compositions and methods for treating cancer and other immune disorders using the aforesaid combination therapies are disclosed.
  • the invention features a method of treating (e.g., inhibiting, reducing, ameliorating, or preventing) a disorder, e.g., a hyperproliferative condition or disorder (e.g., a cancer) in a subject.
  • a disorder e.g., a hyperproliferative condition or disorder (e.g., a cancer) in a subject.
  • the method includes administering to the subject a combination of two, three or more therapeutic agents chosen from one, two or all of the following categories (i)-(iii): (i) an agent that enhances antigen (e.g., tumor antigen) presentation; (ii) an agent that enhances an effector cell response (e.g., B cell and/or T cell activation and/or mobilization); or (iii) an agent that decreases tumor immunosuppression, thereby treating the disorder, e.g., the hyperproliferative condition or disorder (e.g., the cancer).
  • the combinations include a-GITR modulator (e.g., an anti-GITR antibody molecule as described herein).
  • the cancer treated can be, e.g., a cancer described herein, such as lung cancer (squamous), lung cancer (adenocarcinoma), head and neck cancer, cervical cancer (squamous), stomach cancer, thyroid cancer, melanoma, nasopharyngeal cancer, or breast cancer.
  • a cancer described herein such as lung cancer (squamous), lung cancer (adenocarcinoma), head and neck cancer, cervical cancer (squamous), stomach cancer, thyroid cancer, melanoma, nasopharyngeal cancer, or breast cancer.
  • the combination includes one, two, three, four or more therapeutic agents that enhance antigen (e.g., tumor antigen) presentation (referred to herein as an “antigen-presentation combination”) in addition to a GITR modulator (e.g., an anti-GITR antibody molecule as described herein).
  • a GITR modulator e.g., an anti-GITR antibody molecule as described herein.
  • the antigen presentation combination includes one or more of: an agent that enhances antigen presentation (e.g., a vaccine, e.g., a cell- or antigen-based vaccine); an agent that enhances lysis of tumor cells (e.g., an oncolytic virus); an agent that stimulates (e.g., disinhibits) a phagocyte, e.g., a Type I interferon (IFN) activator (e.g., a TLR agonist, a RIG-I-like receptor agonist (RLRs)), and/or an agent that activates and/or recruits a dendritic cell or a macrophage (e.g., a macrophage I), e.g., a bi- or tri-specific cell engager.
  • an agent that enhances antigen presentation e.g., a vaccine, e.g., a cell- or antigen-based vaccine
  • an agent that enhances lysis of tumor cells e.g., an onco
  • the antigen-presentation combination includes one, two, three, four, five or more therapeutic agents chosen from: (i) an agonist of Stimulator of Interferon Genes (a STING agonist), (ii) an agonist of a Toll-like receptor (TLR) (e.g., an agonist of TLR-3, -4, -5, -7, -8, or -9), (iii) a TIM-3 modulator (e.g., an anti-TIM-3 antibody molecule), (iv) a vascular endothelial growth factor receptor (VEGFR) inhibitor, (v) a c-Met inhibitor, (vi) a TGFb inhibitor (e.g., an anti-TGFb antibody), (vii) an IDO/TDO inhibitor, (viii) an A2AR antagonist, (ix) an oncolytic virus, (x) a vaccine (e.g., a scaffold vaccine), or (xi) a bi- or tri-specific cell engager.
  • TLR Toll-like receptor
  • the antigen-presentation combination includes a STING agonist.
  • the antigen-presentation combination includes a TLR agonist (e.g., a TLR7 agonist).
  • the antigen-presentation combination includes a STING agonist and a TLR agonist (e.g., a TLR7 agonist).
  • the antigen presentation combination is chosen from a STING agonist, a TLR agonist, an A2AR antagonist, or an oncolytic virus or a combination thereof, and optionally, one or more of (iii)-(vii) or (x)-(xi).
  • the antigen presentation combination is chosen from a STING agonist or a TLR agonist, or a combination of both, and optionally, one or more of (iii)-(xi).
  • the antigen-presentation combination includes a STING agonist, a TLR agonist (e.g., a TLR7 agonist) and a TIM-3 modulator (e.g., an anti-TIM-3 inhibitor).
  • the antigen-presentation combination includes a STING agonist, a TLR agonist (e.g., a TLR7 agonist) and a VEGFR inhibitor.
  • the antigen-presentation combination includes a STING agonist, a TLR agonist (e.g., a TLR7 agonist) and a c-MET inhibitor.
  • the antigen-presenting combination includes an oncolytic virus.
  • the antigen-presenting combination includes an oncolytic virus and a cytokine, e.g., an oncolytic virus expressing one or more of GM-CSF, or a CSF (e.g., CSF1, or CSF2).
  • the antigen-presenting combination includes a bi- or tri-specific cell engager, e.g., a bi- or tri-specific antibody molecule to CD47 and CD19, with or without an Fc domain.
  • the antigen-presenting combination includes a TGFb inhibitor (e.g., an anti-TGFb antibody).
  • the antigen-presenting combination includes an IDO/TDO inhibitor.
  • the antigen-presenting combination includes an A2AR antagonist.
  • the antigen-presenting combination includes a vaccine (e.g., IL-2 in combination with MUC1, or a dendritic cell based vaccine (e.g., Provenge®)).
  • the antigen-presenting combination includes a vaccine and a TLR agonist (e.g., a TLR agonist as described herein).
  • the antigen-presentation combination includes a vaccine and a STING agonist.
  • the antigen-presentation combination includes a vaccine, a STING agonist and a TLR agonist.
  • the combination includes one, two, three, four, five or more therapeutic agents that enhance an effector cell response (referred to herein as an “effector cell combination”) in addition to a GITR modulator (e.g., an anti-GITR antibody molecule as described herein).
  • the effector cell combination includes a lymphocyte activator, e.g., an NK cell activator and/or a T cell activator.
  • the effector cell combination activates (e.g., disinhibits) a tumor infiltrating lymphocyte (TIL), e.g., an NK cell or a T cell.
  • TIL tumor infiltrating lymphocyte
  • the effector cell combination includes an NK cell modulator chosen from a modulator (e.g., an antibody molecule) of an NK receptor (e.g., a modulator of one or more of NKG2A, KIR3DL, NKp46, MICA or CEACAM1); an interleukin or an interleukin variant (e.g., IL-2, IL-15, IL-21, IL-13R or IL-12 cytokine or variant thereof, or a combination thereof); a bi- or tri-specific cell engager (e.g., a bispecific antibody molecule of NKG2A and CD138, or a bispecific antibody molecule of CD3 and TCR); an NK cell therapy; or a vaccine that includes NK cells and an antigen/immune stimulant.
  • a modulator e.g., an antibody molecule
  • an NK receptor e.g., a modulator of one or more of NKG2A, KIR3DL, NKp46, MICA or CE
  • the effector cell combination includes an immunomodulator (e.g., one or more of: an activator of a costimulatory molecule or an inhibitor of an immune checkpoint molecule as described herein).
  • the effector cell combination includes a T cell modulator chosen from an inhibitor of a checkpoint inhibitor (e.g., an inhibitor of one or more of: PD-1, PD-L1, TIM-3, LAG-3, VISTA, DKG- ⁇ , B7-H3, B7-H4, TIGIT, CTLA4, BTLA, CD160, TIMI, IDO, LAIR1, IL-12, or a combination thereof, e.g., an inhibitor of PD-1 and TIM-3, or an inhibitor of PD-1 and LAG-3).
  • an inhibitor of PD-1 and TIM-3 e.g., an inhibitor of PD-1 and TIM-3, or an inhibitor of PD-1 and LAG-3.
  • the inhibitor of the checkpoint inhibitor is an antibody molecule (e.g., a mono- or bispecific antibody or fragment thereof as described herein).
  • the inhibitor of the checkpoint inhibitor is an antibody molecule against PD-1, PD-L1, TIM-3, LAG-3, VISTA, B7-H4, CTLA4 or TIGIT, or any combination thereof (e.g. a combination as described herein).
  • the effector cell combination includes a T cell modulator chosen from an agonist or an activator of a costimulatory molecule.
  • the agonist of the costimulatory molecule is chosen from an agonist (e.g., an agonistic antibody or antigen-binding fragment thereof, or a soluble fusion) of GITR, OX40, ICOS, SLAM (e.g., SLAMF7), HVEM, LIGHT, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), 4-1BB (CD137), CD30, CD40, BAFFR, CD7, NKG2C, NKp80, CD160, B7-H3, or CD83 ligand.
  • an agonist e.g., an agonistic antibody or antigen-binding fragment thereof, or a soluble fusion
  • GITR e.g., OX40, ICOS
  • SLAM e.g., SLAMF7
  • HVEM e.g., SLAMF7
  • HVEM HVEM
  • LIGHT LIGHT
  • the effector cell combination includes a bispecific T cell engager (e.g., a bispecific antibody molecule that binds to CD3 and a tumor antigen (e.g., EGFR, PSCA, PSMA, EpCAM, HER2 among others).
  • a bispecific T cell engager e.g., a bispecific antibody molecule that binds to CD3 and a tumor antigen (e.g., EGFR, PSCA, PSMA, EpCAM, HER2 among others).
  • the effector cell combination includes one, two, three, four, five or more therapeutic agents chosen from: (i) a GITR modulator (e.g., a GITR agonist (e.g., an anti-GITR antibody molecule as described herein)), (ii) a PD-1 inhibitor, (iii) a PD-L1 inhibitor, (iv) an inhibitor of IAP (Inhibitor of Apoptosis Protein), (v) an inhibitor of EGFR (Epidermal Growth Factor Receptor), (vi) an inhibitor of target of rapamycin (mTOR), (vii) IL-15 or a variant thereof, (viii) a CTLA-4 inhibitor, (ix) a bispecific T cell engager (e.g., a bispecific antibody molecule that binds to CD3 and a tumor antigen (e.g., EGFR, PSCA, PSMA, EpCAM, HER2 among others), (x) a CD40 agonist (e.g., an GITR modul
  • the effector cell combination includes a GITR agonist (e.g., an anti-GITR antibody molecule as described herein).
  • the effector cell combination includes a PD-1 inhibitor.
  • the effector cell combination includes a PD-L1 inhibitor.
  • the effector cell combination includes a GITR agonist (e.g., an anti-GITR antibody molecule as described herein) and a PD-1 inhibitor.
  • the effector cell combination includes a GITR agonist (e.g., an anti-GITR antibody molecule as described herein) and a PD-L1 inhibitor.
  • the effector cell combination includes a GITR agonist (e.g., an anti-GITR antibody molecule as described herein), a PD-1 inhibitor, and a PD-L1 inhibitor. In other embodiments, the effector cell combination includes a PD-1 inhibitor, and a PD-L1 inhibitor. In one embodiment, the effector cell combination includes a GITR agonist (e.g., an anti-GITR antibody molecule as described herein) and an inhibitor of IAP. In another embodiment, the effector cell combination includes a GITR agonist (e.g., an anti-GITR antibody molecule as described herein) and an inhibitor of an EGFR inhibitor.
  • a GITR agonist e.g., an anti-GITR antibody molecule as described herein
  • IAP an inhibitor of IAP
  • the effector cell combination includes a GITR agonist (e.g., an anti-GITR antibody molecule as described herein) and an inhibitor of an EGFR inhibitor.
  • the effector cell combination includes a GITR agonist (e.g., an anti-GITR antibody molecule as described herein) and an inhibitor of an mTOR inhibitor.
  • the effector cell combination includes IL-15 or a variant thereof.
  • the effector cell combination includes a CTLA-4 inhibitor.
  • the effector cell combination includes a bispecific T cell engager (e.g., a bispecific antibody molecule that binds to CD3 and a tumor antigen (e.g., EGFR, PSCA, PSMA, EpCAM, HER2 among others).
  • the effector cell combination includes a CD40 agonist (e.g., an anti-CD40 antibody molecule).
  • the effector cell combination includes an OX40 agonist (e.g., an anti-OX40 antibody molecule).
  • the effector cell combination includes a CD27 agonist (e.g., an anti-CD27 antibody molecule).
  • the combination includes one, two, three, four, five or more therapeutic agents that decrease tumor immunosuppression (referred to herein as an “anti-tumor immunosuppression combination”) in addition to a GITR modulator (e.g., an anti-GITR antibody molecule as described herein).
  • a GITR modulator e.g., an anti-GITR antibody molecule as described herein.
  • the combination modulates the activity or level of one or more of T reg , macrophage 2 or MDSCs.
  • the combination increases one or more of M2 polarization, T reg depletion, or T cell recruitment.
  • the anti-tumor immunosuppression combination includes one, two, three, four, five or more therapeutic agents chosen from: (i) an immunomodulator (e.g., one or more of: an activator of a costimulatory molecule (e.g., a GITR agonist), or an inhibitor of an immune checkpoint molecule (e.g., one or more of PD-1, PD-L1, LAG-3, TIM-3 or CTLA-4), as described herein), (ii) a CSF-1/1R inhibitor (e.g., an inhibitor of macrophage colony-stimulating factor (M-CSF)), (iii) an IL-17 inhibitor, (iv) an IL-1 ⁇ inhibitor, (v) a CXCR2 inhibitor, (vi) an inhibitor of a phosphoinositide 3-kinase (PI3K, e.g., PI3K ⁇ or PI3K ⁇ ), (vii) a BAFF-R inhibitor, (viii) a MALT
  • the immunomodulator is an inhibitor of an immune checkpoint molecule (e.g., an inhibitor of PD-1, PD-L1, LAG-3, TIM-3, CEACAM (e.g., CEACAM-1, -3 and/or -5), or CTLA-4, or any combination thereof). Any combination of the aforesaid agents can be used in the tumor immunosuppression combination.
  • an immune checkpoint molecule e.g., an inhibitor of PD-1, PD-L1, LAG-3, TIM-3, CEACAM (e.g., CEACAM-1, -3 and/or -5), or CTLA-4, or any combination thereof.
  • the anti-tumor immunosuppression combination includes one, two, three, four, five or more therapeutic agents chosen from a PD-1 inhibitor, a PD-L1 inhibitor, a LAG-3 inhibitor, a TIM-3 modulator (e.g., an anti-TIM-3 inhibitor), a GITR agonist (e.g., an anti-GITR antibody molecule as described herein), a CSF-1/1R inhibitor (e.g., an M-CSF inhibitor), an IL-17 inhibitor, an IL-1 ⁇ inhibitor, or a CXCR2 inhibitor.
  • a PD-1 inhibitor e.g., a PD-L1 inhibitor, a LAG-3 inhibitor, a TIM-3 modulator (e.g., an anti-TIM-3 inhibitor), a GITR agonist (e.g., an anti-GITR antibody molecule as described herein), a CSF-1/1R inhibitor (e.g., an M-CSF inhibitor), an IL-17 inhibitor, an IL-1 ⁇ inhibitor, or a CX
  • the anti-tumor immunosuppression combination includes one, two, or all of a GITR agonist (e.g., an anti-GITR antibody molecule as described herein), CSF-1/1R inhibitor (e.g., an M-CSF inhibitor), an IL-17 inhibitor, an IL-1 ⁇ inhibitor.
  • the anti-tumor immunosuppression combination includes a a GITR agonist (e.g., an anti-GITR antibody molecule as described herein), an IL-17 inhibitor, a CXCR2 inhibitor, a CRTH2 inhibitor, an A2AR antagonist, or a PFKFB3 inhibitor, or a combination thereof.
  • the combination includes one or more therapeutic agents of the antigen-presentation combination. In other embodiments, the combination includes one or more therapeutic agents of the effector cell combination. In yet other embodiments, the combination includes one or more therapeutic agents of the anti-tumor immunosuppression combination. In other embodiments, the combination includes one or more therapeutic agents of the antigen-presentation combination and one or more therapeutic agents of the effector cell combination. In other embodiments, the one or more therapeutic agents of the antigen-presentation combination and one or more therapeutic agents of the anti-tumor immunosuppression combination.
  • the combination includes one or more therapeutic agents of the antigen-presentation combination, one or more therapeutic agents of the effector cell combination and one or more therapeutic agents of the anti-tumor immunosuppression combination. In other embodiments, the combination includes one or more therapeutic agents of the antigen-presentation combination, one or more therapeutic agents of the effector cell combination and one or more therapeutic agents of the anti-tumor immunosuppression combination.
  • compositions that include a combination of one or more of: (i) an agent that enhances antigen (e.g., tumor antigen) presentation; (ii) an agent that enhances an effector cell response (e.g., B cell and/or T cell activation and/or mobilization); or (iii) an agent that decreases tumor immunosuppression, thereby treating the disorder, e.g., the hyperproliferative condition or disorder (e.g., the cancer).
  • a GITR modulator e.g., an anti-GITR antibody molecule as described herein.
  • Exemplary combinations of therapeutic agents from two or more of Antigen-Presentation Combinations category (A), Effector Cell Combinations category (B), and Anti-tumor Immunosuppression Combinations category (C) are provided in Table 7:
  • B Effector Cell Immunosuppression 1 STING agonist GITR agonist PD-1 inhibitor 2 TLR agonist PD-1 inhibitor PD-L1 inhibitor 3 TIM-3 modulator PD-L1 inhibitor LAG-3 inhibitor 4 VEGFR inhibitor IAP inhibitor TIM-3 inhibitor 5 c-MET inhibitor EGFR inhibitor GITR inhibitor 6 TGFb inhibitor mTOR inhibitor CSF-1/1R inhibitor 7 IDO/TDO inhibitor IL-15 agonist IL-17 inhibitor 8 A2AR antagonist CTLA-4 inhibitor IL-1 ⁇ inhibitor 9
  • Oncolytic viruses Bispecific T-cell CXCR2 inhibitor engagers 10 Scaffold vaccines CD40 agonist PI3K- ⁇ , - ⁇ inhibitor 11 Bispecific T-cell OX40 agonist BAFF-R inhibitor engagers 12 CD27 agonist MALT-1/BTK inhibitor 13 JAK inhibitor 14 CRTH2 inhibitor 15 VEGFR inhibitor 16 IL-15 agonist 17 Anti-TGFb inhibitor 18
  • the combinations of the present invention include one or more of the following (designations indicate combinations of agents provided in the matrix in Table 7): A1B1, A1B2, A1B3, A1B4, A1B5, A1B6, A1B7, A1B8, A1B9, A1B10, A1B11, A1B12, A2B1, A2B2, A2B3, A2B4, A2B5, A2B6, A2B7, A2B8, A2B9, A2B10, A2B11, A2B12, A3B1, A3B2, A3B3, A3B4, A3B5, A3B6, A3B7, A3B8, A3B9, A3B10, A3B11, A3B12, A4B1, A4B2, A4B3, A4B4, A4B5, A4B6, A4B7, A4B8, A4B9, A4B10, A4B11, A4B12, A5B1, A5B2, A
  • the combination includes a STING agonist.
  • the combination is used to treat a cancer, e.g., a cancer described herein e.g., a solid tumor (e.g., a breast cancer, a squamous cell carcinoma, a melanoma, an ovarian cancer, a fallopian tube carcinoma, a peritoneal carcinoma, a soft tissue sarcoma, a melanoma, a breast cancer, an esophageal cancer, a head and neck cancer, an endometrial cancer, a cervical cancer, or a basal cell carcinoma), e.g., a hematologic malignancy (e.g., a leukemia (e.g., a chronic lymphocytic leukemia (CLL), or a lymphoma (e.g., a marginal zone B-cell lymphoma, a small lymphocytic lymphoma, a follicular lymphom
  • a cancer
  • the STING agonist is cyclic dinucleotide, e.g., a cyclic dinucleotide comprising purine or pyrimidine nucleobases (e.g., adenosine, guanine, uracil, thymine, or cytosine nucleobases).
  • the nucleobases of the cyclic dinucleotide comprise the same nucleobase or different nucleobases.
  • the STING agonist comprises an adenosine or a guanosine nucleobase. In some embodiments, the STING agonist comprises one adenosine nucleobase and one guanosine nucleobase. In some embodiments, the STING agonist comprises two adenosine nucleobases or two guanosine nucleobases.
  • the STING agonist comprises a modified cyclic dinucleotide, e.g., comprising a modified nucleobase, a modified ribose, or a modified phosphate linkage.
  • the modified cyclic dinucleotide comprises a modified phosphate linkage, e.g., a thiophosphate.
  • the STING agonist comprises a cyclic dinucleotide (e.g., a modified cyclic dinucleotide) with 2′,5′ or 3′,5′ phosphate linkages. In some embodiments, the STING agonist comprises a cyclic dinucleotide (e.g., a modified cyclic dinucleotide) with Rp or Sp stereochemistry around the phosphate linkages.
  • the STING agonist is Rp,Rp dithio 2′,3′ c-di-AMP (e.g., Rp,Rp-dithio c-[A(2′,5′)pA(3′,5′)p]), or a cyclic dinucleotide analog thereof.
  • the STING agonist is a compound depicted in U.S. Patent Publication No. US2015/0056224 (e.g., a compound in FIG. 2 c , e.g., compound 21 or compound 22).
  • the STING agonist is c-[G(2′,5′)pG(3′,5′)p], a dithio ribose O-substituted derivative thereof, or a compound depicted in FIG. 4 of PCT Publication Nos. WO 2014/189805 and WO 2014/189806.
  • the STING agonist is c-[A(2′,5′)pA(3′,5′)p] or a dithio ribose 0-substitued derivative thereof, or is a compound depicted in FIG. 5 of PCT Publication Nos. WO 2014/189805 and WO 2014/189806.
  • the STING agonist is c-[G(2′,5′)pA(3′,5′)p], or a dithio ribose 0-substitued derivative thereof, or is a compound depicted in FIG. 5 of PCT Publication Nos. WO 2014/189805 and WO 2014/189806.
  • the STING agonist is 2′-O-propargyl-cyclic-[A(2′,5′)pA(3′,5′)p] (2′-O-propargyl-ML-CDA) or a compound depicted in FIG. 7 of PCT Publication No. WO 2014/189806.
  • STING agonists are disclosed, e.g., in PCT Publication Nos. WO 2014/189805 and WO 2014/189806, and U.S. Publication No. 2015/0056225.
  • a combination described herein includes a Toll-like receptor (TLR) agonist.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a breast cancer, a squamous cell carcinoma, a melanoma, an ovarian cancer, a fallopian tube carcinoma, a peritoneal carcinoma, a soft tissue sarcoma, a melanoma, a breast cancer, an esophageal cancer, a head and neck cancer, an endometrial cancer, a cervical cancer, or a basal cell carcinoma), e.g., a hematologic malignancy (e.g., a leukemia (e.g., a chronic lymphocytic leukemia (CLL), or a lymphoma (e.g., a marginal zone B-cell lymphoma, a small lymphocytic lympho
  • CLL chronic lymph
  • TLRs are a family of pattern recognition receptors that were initially identified as sensors of the innate immune system that recognize microbial pathogens.
  • the TLRs include TLR-1, TLR-2, TLR-3, TLR-4, TLR-5, TLR-6, TLR-7, TLR-8, TLR-9, and TLR-10.
  • TLR-1, -2, -4, -5, and -6, are expressed on the surface of cells and TLR-3, -7/8, and -9 are expressed with the ER compartment.
  • Human dendritic cell subsets can be identified on the basis of distinct TLR expression patterns.
  • the myeloid or “conventional” subset of human dendritic cells express TLRs 1-8 and the plasmacytoid subset of dendritic cells express only TLR-7 and TLR-9.
  • Ligand binding to TLRs invokes a cascade of intra-cellular signaling pathways that induce the production of factors involved in inflammation and immunity.
  • the myeloid subset and the plasmacytoid subset of human dendritic cells result in antigen-specific CD4+ and CD8+ T cell priming and activation of NK cells and T-cells, respectively.
  • the TLR agonist is chosen from one or more of a TLR-1 agonist, a TLR-2 agonist, a TLR-3 agonist, a TLR-4 agonist, a TLR-5 agonist, a TLR-6 agonist, a TLR-7 agonist, a TLR-8 agonist, a TLR-9 agonist, a TLR-10 agonist, a TLR-1/2 agonist, a TLR-2/6 agonist, or a TLR-7/8 agonist.
  • the TLR agonist is a TLR7 agonist.
  • the TLR agonist is imiquimod or 3-(2-Methylpropyl)-3,5,8-triazatricyclo[7.4.0.02,6]trideca-1(9),2(6),4,7,10,12-hexaen-7-amine.
  • Imiquimod or 3-(2-Methylpropyl)-3,5,8-triazatricyclo[7.4.0.02,6]trideca-1(9),2(6),4,7,10,12-hexaen-7-amine can bind to and activate TLR-7 and/or TLR-8.
  • the TLR agonist is 852A.
  • 852A is disclosed, e.g., in Inglefield et al. J Interferon Cytokine Res. 2008; 28(4):253-63. 852A can bind to and activate TLR-7 and/or TLR-8.
  • the TLR agonist is Bacille Calmette-Guérin (BCG). BCG can bind to and activate TLR-9.
  • the TLR agonist is EMD 120108.
  • EMD 120108 is a synthetic oligonucleotide containing phosphorothioate oligodeoxynucleotide.
  • EMD 1201081 can bind to and activate TLR-9, e.g, in monocytes/macrophages, plasmacytoid dendritic cells (DCs) and B cells, initiating immune signaling pathways, activating B cells and inducing T-helper cell cytokine production.
  • TLR-9 e.g, in monocytes/macrophages, plasmacytoid dendritic cells (DCs) and B cells, initiating immune signaling pathways, activating B cells and inducing T-helper cell cytokine production.
  • the TLR agonist is IMO-2055.
  • IMO-2055 is a synthetic oligonucleotide containing unmethylated CpG dinucleotides. Mimicking unmethylated CpG sequences in bacterial DNA, IMO-2055 can bind to and activate TLR-9, e.g., in monocytes/macrophages, plasmacytoid dendritic cells (DCs) and B cells, initiating immune signaling pathways and activating B cells and DCs and inducing T-helper cell cytokine production.
  • DCs plasmacytoid dendritic cells
  • TLR-1/2 agonists e.g., Pam3Cys
  • TLR-2 agonists e.g., CFA, MALP2, Pam2Cys, FSL-1, or Hib-OMPC
  • TLR-3 agonists e.g., polyribosinic:polyribocytidic acid (Poly I:C), polyadenosine-polyuridylic acid (poly AU), polyinosinic-polycytidylic acid stabilized with poly-L-lysine and carboxymethylcellulose (Hiltonol®)
  • TLR-4 agonists e.g., monophosphoryl lipid A (MPL), LPS, sialyl-Tn (STn)
  • TLR-5 agonists e.g., bacterial flagellin
  • TLR-7 agonists e.g., imiquimod
  • TLR-7/8 agonists e.g.
  • the TLR agonist is used in combination with a GITR agonist, e.g., as described in WO2004060319, and International Publication No.: WO2014012479.
  • a combination described herein includes a vascular endothelial growth factor (VEGF) receptor inhibitor (e.g., an inhibitor of one or more of VEGFR (e.g., VEGFR-1, VEGFR-2, VEGFR-3) or VEGF).
  • VEGF vascular endothelial growth factor
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a melanoma, a breast cancer, a colon cancer, an esophageal cancer, a gastrointestinal stromal tumor (GIST), a kidney cancer (e.g., a renal cell cancer), a liver cancer, a non-small cell lung cancer (NSCLC), an ovarian cancer, a pancreatic cancer, a prostate cancer, or a stomach cancer), e.g., a hematologic malignancy (e.g., a lymphoma).
  • a cancer described herein e.g., a solid tumor (e.g., a melanoma, a breast cancer, a colon cancer, an esophageal cancer, a gastrointestinal stromal tumor (GIST), a kidney cancer (e.g., a renal cell cancer), a liver cancer, a non-small cell
  • the VEGFR inhibitor is vatalanib succinate (Compound A47) or a compound disclosed in EP 296122.
  • the VEGFR inhibitor is an inhibitor of one or more of VEGFR-2, PDGFRbeta, KIT or Raf kinase C, 1-methyl-5-((2-(5-(trifluoromethyl)-1H-imidazol-2-yl)pyridin-4-yl)oxy)-N-(4-(trifluoromethyl)phenyl)-1H-benzo[d]imidazol-2-amine (Compound A37) or a compound disclosed in PCT Publication No. WO 2007/030377.
  • VEGFR pathway inhibitors that can be used in the combinations disclosed herein include, e.g., bevacizumab (AVASTIN®), axitinib (INLYTA®); brivanib alaninate (BMS-582664, (S)-((R)-1-(4-(4-Fluoro-2-methyl-1H-indol-5-yloxy)-5-methylpyrrolo[2,1-f][1,2,4]triazin-6-yloxy)propan-2-yl)2-aminopropanoate); sorafenib (NEXAVAR®); pazopanib (VOTRIENT®); sunitinib malate (SUTENT®); cediranib (AZD2171, CAS 288383-20-1); vargatef (BIBF1120, CAS 928326-83-4); Foretinib (GSK1363089); telatinib (BAY57-9352, CAS 332012-40
  • WO 02/066470 dovitinib dilactic acid (TKI258, CAS 852433-84-2); linfanib (ABT869, CAS 796967-16-3); cabozantinib (XL184, CAS 849217-68-1); lestaurtinib (CAS 111358-88-4); N-[5-[[[5-(1,1-dimethylethyl)-2-oxazolyl]methyl]thio]-2-thiazolyl]-4-piperidinecarboxamide (BMS38703, CAS 345627-80-7); (3R,4R)-4-amino-1-((4-((3-methoxyphenyl)amino)pyrrolo[2,1-f][1,2,4]triazin-5-yl)methyl)piperidin-3-ol (BMS690514); N-(3,4-Dichloro-2-fluorophenyl)-6-methoxy-7-[[(
  • anti-VEGF antibodies that can be used in the combinations disclosed herein include, e.g., a monoclonal antibody that binds to the same epitope as the monoclonal anti-VEGF antibody A4.6.1 produced by hybridoma ATCC HB 10709; a recombinant humanized anti-VEGF monoclonal antibody generated according to Presta et al. (1997) Cancer Res. 57:4593-4599.
  • the anti-VEGF antibody is Bevacizumab (BV), also known as rhuMAb VEGF or AVASTIN®.
  • antibodies include those that bind to a functional epitope on human VEGF comprising of residues F17, M18, D19, Y21, Y25, Q89, 191, K1 01, E1 03, and C104 or, alternatively, comprising residues F17, Y21, Q22, Y25, D63, 183 and Q89.
  • a combination described herein includes an inhibitor of c-MET.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a non-small cell lung cancer, a pancreatic cancer, a liver cancer, a thyroid cancer, a brain tumor (e.g., a glioblastoma), a kidney cancer (e.g., a renal cell carcinoma), a head and neck cancer (e.g., a head and neck squamous cell carcinoma).
  • a cancer described herein e.g., a solid tumor (e.g., a non-small cell lung cancer, a pancreatic cancer, a liver cancer, a thyroid cancer, a brain tumor (e.g., a glioblastoma), a kidney cancer (e.g., a renal cell carcinoma), a head and neck cancer (e.g., a head and neck squamous cell carcinoma).
  • the c-MET inhibitor is Compound A17 or a compound described in U.S. Pat. Nos. 7,767,675 and 8,420,645).
  • c-MET a receptor tyrosine kinase overexpressed or mutated in many tumor cell types, plays key roles in tumor cell proliferation, survival, invasion, metastasis, and tumor angiogenesis. Inhibition of c-MET may induce cell death in tumor cells overexpressing c-MET protein or expressing constitutively activated c-MET protein.
  • the c-MET inhibitor is JNJ-38877605.
  • JNJ-38877605 is an orally available, small molecule inhibitor of c-Met. JNJ-38877605 selectively binds to c-MET, thereby inhibiting c-MET phosphorylation and disrupting c-Met signal transduction pathways.
  • the c-Met inhibitor is AMG 208.
  • AMG 208 is a selective small-molecule inhibitor of c-MET. AMG 208 inhibits the ligand-dependent and ligand-independent activation of c-MET, inhibiting its tyrosine kinase activity, which may result in cell growth inhibition in tumors that overexpress c-Met.
  • the c-Met inhibitor is AMG 337.
  • AMG 337 is an orally bioavailable inhibitor of c-Met.
  • AMG 337 selectively binds to c-MET, thereby disrupting c-MET signal transduction pathways.
  • the c-Met inhibitor is LY2801653.
  • LY2801653 is an orally available, small molecule inhibitor of c-Met. LY2801653 selectively binds to c-MET, thereby inhibiting c-MET phosphorylation and disrupting c-Met signal transduction pathways.
  • c-Met inhibitor is MSC2156119J.
  • MSC2156119J is an orally bioavailable inhibitor of c-Met.
  • MSC2156119J selectively binds to c-MET, which inhibits c-MET phosphorylation and disrupts c-Met-mediated signal transduction pathways.
  • the c-MET inhibitor is capmatinib.
  • Capmatinib is also known as INCB028060.
  • Capmatinib is an orally bioavailable inhibitor of c-MET.
  • Capmatinib selectively binds to c-Met, thereby inhibiting c-Met phosphorylation and disrupting c-Met signal transduction pathways.
  • the c-MET inhibitor is crizotinib.
  • Crizotinib is also known as PF-02341066.
  • Crizotinib is an orally available aminopyridine-based inhibitor of the receptor tyrosine kinase anaplastic lymphoma kinase (ALK) and the c-Met/hepatocyte growth factor receptor (HGFR).
  • ALK receptor tyrosine kinase anaplastic lymphoma kinase
  • HGFR c-Met/hepatocyte growth factor receptor
  • Crizotinib in an ATP-competitive manner, binds to and inhibits ALK kinase and ALK fusion proteins.
  • crizotinib inhibits c-Met kinase, and disrupts the c-Met signaling pathway. Altogether, this agent inhibits tumor cell growth.
  • the c-MET inhibitor is golvatinib.
  • Golvatinib is an orally bioavailable dual kinase inhibitor of c-MET and VEGFR-2 with potential antineoplastic activity. Golvatinib binds to and inhibits the activities of both c-MET and VEGFR-2, which may inhibit tumor cell growth and survival of tumor cells that overexpress these receptor tyrosine kinases.
  • the c-MET inhibitor is tivantinib.
  • Tivantinib is also known as ARQ 197.
  • Tivantinib is an orally bioavailable small molecule inhibitor of c-MET. Tivantinib binds to the c-MET protein and disrupts c-Met signal transduction pathways, which may induce cell death in tumor cells overexpressing c-MET protein or expressing consitutively activated c-Met protein.
  • a combination described herein includes a transforming growth factor beta (TGF- ⁇ ) inhibitor.
  • TGF- ⁇ transforming growth factor beta
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a brain cancer (e.g., a glioma), a melanoma, a kidney cancer (e.g., a renal cell carcinoma), a pleural malignant mesothelioma (e.g., a relapsed pleural malignant mesothelioma), or a breast cancer (e.g., a metastatic breast cancer)).
  • a cancer described herein e.g., a solid tumor (e.g., a brain cancer (e.g., a glioma), a melanoma, a kidney cancer (e.g., a renal cell carcinoma), a pleural malignant mesotheli
  • the TGF- ⁇ inhibitor is fresolimumab (CAS Registry Number: 948564-73-6). Fresolimumab is also known as GC1008. Fresolimumab is a human monoclonal antibody that binds to and inhibits TGF-beta isoforms 1, 2 and 3.
  • Fresolimumab is disclosed, e.g., in WO 2006/086469, U.S. Pat. No. 8,383,780, and U.S. Pat. No. 8,591,901.
  • the TGF- ⁇ inhibitor is XOMA 089.
  • XOMA 089 is also known as XPA.42.089.
  • XOMA 089 is a fully human monoclonal antibody that specifically binds and neutralizes TGF-beta 1 and 2 ligands.
  • the heavy chain variable region of XOMA 089 has the amino acid sequence disclosed as SEQ ID NO: 6 in WO 2012/167143.
  • the light chain variable region of XOMA 089 has the amino acid sequence disclosed as SEQ ID NO: 8 in WO 2012/167143.
  • a combination described herein includes an inhibitor of indoleamine 2,3-dioxygenase (IDO) and/or tryptophan 2,3-dioxygenase (TDO).
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., melanoma, non-small cell lung cancer, colon cancer, squamous cell head and neck cancer, ovarian cancer, peritoneal cancer, fallopian tube cancer, breast cancer (e.g., metastatic or HER2-negative breast cancer)), e.g., a hematologic malignancy (e.g., a lymphoma, e.g., a non-Hodgkin's lymphoma or a Hodgkin's lymphoma (e.g., a diffuse large B-cell lymphoma (DLBCL))).
  • a cancer described herein e.g.,
  • the IDO/TDO inhibitor is chosen from (4E)-4-[(3-chloro-4-fluoroanilino)-nitrosomethylidene]-1,2,5-oxadiazol-3-amine (also known as INCB24360), indoximod (1-methyl-D-tryptophan), or ⁇ -cyclohexyl-5H-Imidazo[5,1-a]isoindole-5-ethanol (also known as NLG919).
  • the IDO/TDO inhibitor is epacadostat (CAS Registry Number: 1204669-58-8).
  • Epacadostat is also known as INCB24360 or INCB024360 (Incyte).
  • Epacadostat is a potent and selective indoleamine 2,3-dioxygenase (IDO1) inhibitor with IC50 of 10 nM, highly selective over other related enzymes such as IDO2 or tryptophan 2,3-dioxygenase (TDO).
  • the IDO/TDO inhibitor is indoximod (New Link Genetics).
  • Indoximod the D isomer of 1-methyl-tryptophan, is an orally administered small-molecule indoleamine 2,3-dioxygenase (IDO) pathway inhibitor that disrupts the mechanisms by which tumors evade immune-mediated destruction.
  • IDO indoleamine 2,3-dioxygenase
  • the IDO/TDO inhibitor is NLG919 (New Link Genetics).
  • NLG919 is a potent IDO (indoleamine-(2,3)-dioxygenase) pathway inhibitor with Ki/EC50 of 7 nM/75 nM in cell-free assays.
  • the IDO/TDO inhibitor is F001287 (Flexus/BMS).
  • F001287 is a small molecule inhibitor of indoleamine 2,3-dioxygenase 1 (IDO1).
  • a combination described herein includes an adenosine A2a receptor (A2aR) antagonist (e.g., an inhibitor of A2aR pathway, e.g., an adenosine inhibitor, e.g., an inhibitor of A2aR or CD-73).
  • A2aR adenosine A2a receptor
  • the combination is used to treat a cancer, e.g., a cancer described herein.
  • the A2aR antagonist is istradefylline (CAS Registry Number: 155270-99-8).
  • Istradefylline is also known as KW-6002 or 8-[(E)-2-(3,4-dimethoxyphenyl)vinyl]-1,3-diethyl-7-methyl-3,7-dihydro-1H-purine-2,6-dione.
  • Istradefylline is disclosed, e.g., in LeWitt et al. (2008) Annals of Neurology 63 (3): 295-302).
  • the A2aR antagonist is tozadenant (Biotie). Tozadenant is also known as SYN115 or 4-hydroxy-N-(4-methoxy-7-morpholin-4-yl-1,3-benzothiazol-2-yl)-4-methylpiperidine-1-carboxamide. Tozadenant blocks the effect of endogenous adenosine at the A2a receptors, resulting in the potentiation of the effect of dopamine at the D2 receptor and inhibition of the effect of glutamate at the mGluR5 receptor. e.g., In some embodiments, the A2aR antagonist is preladenant (CAS Registry Number: 377727-87-2).
  • Preladenant is also known as SCH 420814 or 2-(2-Furanyl)-7-[2-[4-[4-(2-methoxyethoxy)phenyl]-1-piperazinyl]ethyl]7H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidine-5-amine.
  • Preladenant was developed as a drug that acted as a potent and selective antagonist at the adenosine A2A receptor.
  • the A2aR antagonist is vipadenan. Vipadenan is also known as BIIB014, V2006, or 3-[(4-amino-3-methylphenyl)methyl]-7-(furan-2-yl)triazolo[4,5-d]pyrimidin-5-amine. e.g., In some embodiments, the A2aR antagonist is PBF-509 (Palobiofarma). e.g., In some embodiments, the A2aR antagonist, e.g., PBF-509 is administered at a daily dose of about 80 mg, 160 mg, or 240 mg.
  • A2aR antagonists include, e.g., ATL-444, MSX-3, SCH-58261, SCH-412,348, SCH-442,416, VER-6623, VER-6947, VER-7835, CGS-15943, or ZM-241,385.
  • the A2aR antagonist is an A2aR pathway antagonist (e.g., a CD-73 inhibitor, e.g., an anti-CD73 antibody) is MEDI9447.
  • MEDI9447 is a monoclonal antibody specific for CD73. Targeting the extracellular production of adenosine by CD73 may reduce the immunosuppressive effects of adenosine.
  • MEDI9447 was reported to have a range of activities, e.g., inhibition of CD73 ectonucleotidase activity, relief from AMP-mediated lymphocyte suppression, and inhibition of syngeneic tumor growth.
  • MEDI9447 can drive changes in both myeloid and lymphoid infiltrating leukocyte populations within the tumor microenvironment. These changes include, e.g., increases in CD8 effector cells and activated macrophages, as well as a reduction in the proportions of myeloid-derived suppressor cells (MDSC) and regulatory T lymphocytes.
  • MDSC myeloid-derived suppressor cells
  • a combination as described herein includes an oncolytic virus.
  • oncolytic viruses are capable of selectively replicating in and triggering the death of or slowing the growth of a cancer cell. In some cases, oncolytic viruses have no effect or a minimal effect on non-cancer cells.
  • An oncolytic virus includes but is not limited to an oncolytic adenovirus, oncolytic Herpes Simplex Viruses, oncolytic retrovirus, oncolytic parvovirus, oncolytic vaccinia virus, oncolytic Sinbis virus, oncolytic influenza virus, or oncolytic RNA virus (e.g., oncolytic reovirus, oncolytic Newcastle Disease Virus (NDV), oncolytic measles virus, or oncolytic vesicular stomatitis virus (VSV)).
  • oncolytic adenovirus e.g., oncolytic Herpes Simplex Viruses, oncolytic retrovirus, oncolytic parvovirus, oncolytic vaccinia virus, oncolytic Sinbis virus, oncolytic influenza virus, or oncolytic RNA virus (e.g., oncolytic reovirus, oncolytic Newcastle Disease Virus (NDV), oncolytic measles virus, or oncolytic vesicular stomatitis virus (V
  • the oncolytic virus is a virus, e.g., recombinant oncolytic virus, described in US2010/0178684 A1, which is incorporated herein by reference in its entirety.
  • a recombinant oncolytic virus comprises a nucleic acid sequence (e.g., heterologous nucleic acid sequence) encoding an inhibitor of an immune or inflammatory response, e.g., as described in US2010/0178684 A1, incorporated herein by reference in its entirety.
  • the recombinant oncolytic virus e.g., oncolytic NDV
  • a pro-apoptotic protein e.g., apoptin
  • a cytokine e.g., GM-CSF, CSF, interferon-gamma, interleukin-2 (IL-2), tumor necrosis
  • the oncolytic virus is a chimeric oncolytic NDV described in U.S. Pat. No. 8,591,881 B2, US 2012/0122185 A1, or US 2014/0271677 A1, each of which is incorporated herein by reference in their entireties.
  • the oncolytic virus comprises a conditionally replicative adenovirus (CRAd), which is designed to replicate exclusively in cancer cells. See, e.g., Alemany et al. Nature Biotechnol. 18(2000):723-27.
  • CRAd conditionally replicative adenovirus
  • an oncolytic adenovirus comprises one described in Table 1 on page 725 of Alemany et al., incorporated herein by reference in its entirety.
  • Exemplary oncolytic viruses include but are not limited to the following:
  • Group B Oncolytic Adenovirus (ColoAdl) (PsiOxus Therapeutics Ltd.) (see, e.g., Clinical Trial Identifier: NCT02053220);
  • ONCOS-102 (previously called CGTG-102), which is an adenovirus comprising granulocyte-macrophage colony stimulating factor (GM-CSF) (Oncos Therapeutics) (see, e.g., Clinical Trial Identifier: NCT01598129);
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • VCN-01 which is a genetically modified oncolytic human adenovirus encoding human PH20 hyaluronidase (VCN Biosciences, S.L.) (see, e.g., Clinical Trial Identifiers: NCT02045602 and NCT02045589);
  • Conditionally Replicative Adenovirus ICOVIR-5 which is a virus derived from wild-type human adenovirus serotype 5 (Had5) that has been modified to selectively replicate in cancer cells with a deregulated retinoblastoma/E2F pathway (Institut Catala d'Oncologia) (see, e.g., Clinical Trial Identifier: NCT01864759);
  • Celyvir which comprises bone marrow-derived autologous mesenchymal stem cells (MSCs) infected with ICOVIR5, an oncolytic adenovirus (Hospital Infantil Universitario Ni ⁇ o Jes ⁇ s, Madrid, Spain/Ramon Alemany) (see, e.g., Clinical Trial Identifier: NCT01844661);
  • CG0070 which is a conditionally replicating oncolytic serotype 5 adenovirus (Ad5) in which human E2F-1 promoter drives expression of the essential Ela viral genes, thereby restricting viral replication and cytotoxicity to Rb pathway-defective tumor cells (Cold Genesys, Inc.) (see, e.g., Clinical Trial Identifier: NCT02143804); or
  • DNX-2401 (formerly named Delta-24-RGD), which is an adenovirus that has been engineered to replicate selectively in retinoblastoma (Rb)-pathway deficient cells and to infect cells that express certain RGD-binding integrins more efficiently (Clinica Universidad de Navarra, Universidad de Navarra/DNAtrix, Inc.) (see, e.g., Clinical Trial Identifier: NCT01956734).
  • an oncolytic virus described herein is administering by injection, e.g., subcutaneous, intra-arterial, intravenous, intramuscular, intrathecal, or intraperitoneal injection.
  • an oncolytic virus described herein is administered intratumorally, transdermally, transmuco sally, orally, intranasally, or via pulmonary administration.
  • Vaccines e.g., Scaffold Vaccines
  • a combination described herein includes a vaccine, e.g., a scaffold vaccine.
  • the combination is used to treat a cancer, e.g., a cancer described herein.
  • Cancer vaccines are disclosed, e.g., in PCT Publication Nos. WO 2007/070660 and WO 2012/167230, EP 1960009 B1, U.S. Pat. No. 8,067,237 and U.S. Pat. No. 8,932,583, and U.S. Publication No. US 2011/0020216.
  • the components that can be used within cancer vaccines e.g., implantable scaffold materials
  • Methods that can be used for administration of cancer vaccines are disclosed, e.g., in PCT Publication Nos. WO 2013/158673, WO 2012/048165, and WO 2012/149358.
  • the cancer vaccine includes a macroporous scaffold comprising (i) cells or a cell recruitment composition, and (ii) a deployment signal capable of inducing or promoting migration of cells, and (iii) a bioactive composition coated or seeded onto/into the scaffold, which causes cells recruited into the scaffold be modified. Migration of the modified cells can be promoted by the open, interconnected macropores and the deployment signal.
  • the cancer vaccine induces an endogenous immune response to a cancer target via administration of a porous scaffold bearing a recruitment composition and a target antigen composition, wherein an endogenous antigen presenting cell is recruited into the scaffold to encounter antigen and where said cell resides until a deployment signal induces egress to a lymph node tissue outside the scaffold, thereby stimulating an endogenous immune response to said cancer target.
  • the cancer vaccine is used to remove a target cell from a mammal using a scaffold composition.
  • an in situ cancer vaccine is generated via recruitment of cancer cells to an implanted scaffold and destruction of the cells using a cytotoxic agent.
  • a cytosine-guanosine oligonucleotide (CpG-ODN) is used as a component of a scaffold, which can effectively reprogram and deploy dendritic cells recruited to the scaffold, and generate an effective anti-tumor response.
  • polyinosine-polycytidylic acid (poly I:C) and/or CpG ODN are used to exert a synergistic effect on tumor inhibition.
  • porous rods comprising an immune cell recruitment compound (e.g. GM-CSF) and an immune cell activation compound (e.g. CpG ODN), and optionally comprising an antigen such as a tumor lysate, are used, e.g., to elicit an immune response to a vaccine antigen.
  • an immune cell recruitment compound e.g. GM-CSF
  • an immune cell activation compound e.g. CpG ODN
  • an antigen such as a tumor lysate
  • pores that facilitate recruitment or release of cells are formed in situ within hydrogels following hydrogel injection.
  • injectable shape memory porous hydrogel polymer is used for administration.
  • the combinations disclosed herein include a cancer or tumor vaccine.
  • tumor vaccines that can be used include peptides of melanoma antigens, such as peptides of gp100, MAGE antigens, Trp-2, MART1 and/or tyrosinase, tumor cells transfected to express the cytokine GM-CSF, DNA-based vaccines, RNA-based vaccines, and viral transduction-based vaccines.
  • the cancer vaccine may be prophylactic or therapeutic.
  • a vaccine is prepared using autologous or allogeneic tumor cells. These cellular vaccines have been shown to be most effective when the tumor cells are transduced to express GM-CSF. GM-CSF has been shown to be a potent activator of antigen presentation for tumor vaccination (Dranoff et al. (1993) Proc. Natl. Acad. Sci. U.S.A. 90: 3539-43).
  • the combinations disclosed herein can be used in conjunction with a collection of recombinant proteins and/or peptides expressed in a tumor in order to generate an immune response to these proteins.
  • These proteins are normally viewed by the immune system as self antigens and are therefore tolerant to them.
  • the tumor antigen may also include the protein telomerase, which is required for the synthesis of telomeres of chromosomes and which is expressed in more than 85% of human cancers and in only a limited number of somatic tissues (Kim, N et al. (1994) Science 266: 2011-2013). (These somatic tissues may be protected from immune attack by various means).
  • Tumor antigen may also be “neo-antigens” expressed in cancer cells because of somatic mutations that alter protein sequence or create fusion proteins between two unrelated sequences (ie. bcr-abl in the Philadelphia chromosome), or idiotype from B cell tumors.
  • tumor vaccines may include the proteins from viruses implicated in human cancers such a Human Papilloma Viruses (HPV), Hepatitis Viruses (HBV and HCV), Kaposi's Herpes Sarcoma Virus (KHSV), and Epstein-Barr virus (EBV).
  • HPV Human Papilloma Viruses
  • HBV and HCV Hepatitis Viruses
  • KHSV Kaposi's Herpes Sarcoma Virus
  • EBV Epstein-Barr virus
  • Another form of tumor specific antigen which may be used in conjunction with a GITR agonist is purified heat shock proteins (HSP) isolated from the tumor tissue itself. These heat shock proteins contain fragments of proteins from the tumor cells and these HSPs are highly efficient at delivery to antigen presenting cells for eliciting tumor immunity (Suot, R & Srivastava, P (1995) Science 269:1585-1588; Tamura, Y. et al. (1997) Science 278:117
  • DC Dendritic cells
  • DC immunization may be effectively combined with other agent, e.g., a GITR agonist, to activate more potent anti-tumor responses.
  • a combination described herein includes a bispecific T-cell engager.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a gastrointestinal cancer, a melanoma, or a lung cancer) or a hematologic malignancy (e.g., a lymphoma (e.g., non-Hodgkin's lymphoma) or a leukemia (e.g., an acute lymphoblastic leukemia).
  • a cancer e.g., a cancer described herein, e.g., a solid tumor (e.g., a gastrointestinal cancer, a melanoma, or a lung cancer) or a hematologic malignancy (e.g., a lymphoma (e.g., non-Hodgkin's lymphoma) or a leukemia (e.g., an acute lymphoblastic
  • Bi-specific T-cell engagers are a class of artificial bispecific monoclonal antibodies that can direct a host's immune system, e.g., the T cells' cytotoxic activity, against cancer cells.
  • Bi-specific T-cell engagers can form a link between T cells and tumor cells, which causes T cells to exert cytotoxic activity on tumor cells by producing proteins like perforin and granzymes, independently of the presence of MHC I or co-stimulatory molecules. These proteins enter tumor cells and initiate the cell's apoptosis. This action mimics physiological processes observed during T cell attacks against tumor cells.
  • the bi-specific T-cell engager is a fusion protein comprising two single-chain variable fragments (scFvs) of different antibodies.
  • one of the scFvs binds to T cells, e.g., via the CD3 receptor, and the other to a tumor cell, e.g., via a tumor specific molecule.
  • the bi-specific T-cell engager is a bispecific antibody molecule of NKG2A and CD138, or a bispecific antibody molecule of CD3 and TCR. In some embodiments, the bispecific T-cell engager is a bispecific antibody molecule that binds to CD3 and a tumor antigen (e.g., EGFR, PSCA, PSMA, EpCAM, HER2 among others).
  • a tumor antigen e.g., EGFR, PSCA, PSMA, EpCAM, HER2 among others.
  • the bi-specific T-cell engager is blinatumomab (CAS Registry Number: 853426-35-4).
  • Blinatumomab is also known as MT103.
  • Blinatumomab specifically targets a CD3 site for T cells and a CD19 site for B cells.
  • the bi-specific T-cell engager is MT110.
  • MT110 is a single-chain antibody that targets EpCAM and CD3.
  • MT110 is disclosed, e.g., in Amann et al. J Immunother. 2009; 32(5):452-64.
  • the bi-specific T-cell engager targets melanoma-associated chondroitin sulfate proteoglycan (MCSP). In some embodiments, the bi-specific T-cell engager targets CD33. In some embodiments the bi-specific T-cell engager comprises trastuzumab (targeting HER2/neu), cetuximab, or panitumumab (both targeting the EGF receptor), a functional fragment thereof. In some embodiments, the bi-specific T-cell engager targets CD66e and EphA2.
  • the GITR modulator is an antibody molecule, e.g., an anti-GITR antibody molecule as provided herein.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor or a hematologic malignancy.
  • exemplary GITR modulators include, e.g., GITR fusion proteins and anti-GITR antibodies (e.g., bivalent anti-GITR antibodies), such as, a GITR fusion protein described in U.S. Pat. No. 6,111,090, European Patent No.: 0920505B1, U.S. Pat. No. 8,586,023, PCT Publication Nos.: WO 2010/003118 and 2011/090754, or an anti-GITR antibody described, e.g., in U.S. Pat. No. 7,025,962, European Patent No.: 1947183B1, U.S. Pat. No. 7,812,135, U.S. Pat. No. 8,388,967, U.S. Pat. No.
  • the GITR agonist is used in combination with a PD-1 inhibitor, e.g., as described in WO2015/026684.
  • the GITR agonist is used in combination with a TLR agonist, e.g., as described in WO2004060319, and International Publication No.: WO2014012479.
  • the PD-1 inhibitor is an anti-PD-1 antibody chosen from Nivolumab, Pembrolizumab or Pidilizumab.
  • the anti-PD-1 antibody is Nivolumab.
  • Alternative names for Nivolumab include MDX-1106, MDX-1106-04, ONO-4538, or BMS-936558.
  • the anti-PD-1 antibody is Nivolumab (CAS Registry Number: 946414-94-4).
  • Nivolumab is a fully human IgG4 monoclonal antibody which specifically blocks PD1.
  • Nivolumab (clone 5C4) and other human monoclonal antibodies that specifically bind to PD1 are disclosed in U.S. Pat. No. 8,008,449 and WO2006/121168.
  • the inhibitor of PD-1 is Nivolumab, and having a sequence disclosed therein (or a sequence substantially identical or similar thereto, e.g., a sequence at least 85%, 90%, 95% identical or higher to the sequence specified).
  • the anti-PD-1 antibody is Pembrolizumab.
  • Pembrolizumab also referred to as Lambrolizumab, MK-3475, MK03475, SCH-900475 or KEYTRUDA®; Merck
  • Pembrolizumab and other humanized anti-PD-1 antibodies are disclosed in Hamid, O. et al. (2013) New England Journal of Medicine 369 (2): 134-44, U.S. Pat. No. 8,354,509 and WO2009/114335.
  • the inhibitor of PD-1 is Pembrolizumab disclosed in, e.g., U.S. Pat. No. 8,354,509 and WO 2009/114335, and having a sequence disclosed therein (or a sequence substantially identical or similar thereto, e.g., a sequence at least 85%, 90%, 95% identical or higher to the sequence specified).
  • the anti-PD-1 antibody is Pidilizumab.
  • Pidilizumab (CT-011; Cure Tech) is a humanized IgGlk monoclonal antibody that binds to PD1.
  • Pidilizumab and other humanized anti-PD-1 monoclonal antibodies are disclosed in WO2009/101611.
  • anti-PD1 antibodies include AMP 514 (Amplimmune), among others, e.g., anti-PD1 antibodies disclosed in U.S. Pat. No. 8,609,089, US 2010028330, and/or US 20120114649.
  • the PD-1 inhibitor is an immunoadhesin (e.g., an immunoadhesin comprising an extracellular or PD-1 binding portion of PD-L1 or PD-L2 fused to a constant region (e.g., an Fc region of an immunoglobulin sequence).
  • the PD-1 inhibitor is AMP-224 (B7-DCIg; Amplimmune; e.g., disclosed in WO2010/027827 and WO2011/066342), is a PD-L2 Fc fusion soluble receptor that blocks the interaction between PD-1 and B7-H1.
  • a combination includes an anti-GITR antibody molecule, e.g., as described herein, and an anti-PD-1 antibody disclosed in, e.g., WO 2015/112900, and having a sequence disclosed therein (or a sequence substantially identical or similar thereto, e.g., a sequence at least 85%, 90%, 95% identical or higher to the sequence specified).
  • the PD-L1 inhibitor is an antibody molecule.
  • the anti-PD-L1 inhibitor is chosen from YW243.55.S70, MPDL3280A, MEDI-4736, MSB-0010718C, or MDX-1105.
  • the anti-PD-L1 antibody is MSB0010718C.
  • MSB0010718C (also referred to as A09-246-2; Merck Serono) is a monoclonal antibody that binds to PD-L1.
  • Pembrolizumab and other humanized anti-PD-L1 antibodies are disclosed in WO2013/079174, and having a sequence disclosed therein (or a sequence substantially identical or similar thereto, e.g., a sequence at least 85%, 90%, 95% identical or higher to the sequence specified).
  • the PD-L1 inhibitor is YW243.55.S70.
  • the YW243.55.S70 antibody is an anti-PD-L1 described in WO 2010/077634 (heavy and light chain variable region sequences shown in SEQ ID Nos. 20 and 21, respectively), and having a sequence disclosed therein (or a sequence substantially identical or similar thereto, e.g., a sequence at least 85%, 90%, 95% identical or higher to the sequence specified).
  • the PD-L1 inhibitor is MDX-1105.
  • MDX-1105 also known as BMS-936559, is an anti-PD-L1 antibody described in WO2007/005874, and having a sequence disclosed therein (or a sequence substantially identical or similar thereto, e.g., a sequence at least 85%, 90%, 95% identical or higher to the sequence specified).
  • the PD-L1 inhibitor is MDPL3280A (Genentech/Roche).
  • MDPL3280A is a human Fc optimized IgG1 monoclonal antibody that binds to PD-L1.
  • MDPL3280A and other human monoclonal antibodies to PD-L1 are disclosed in U.S. Pat. No. 7,943,743 and U.S Publication No.: 20120039906.
  • the PD-L2 inhibitor is AMP-224.
  • AMP-224 is a PD-L2 Fc fusion soluble receptor that blocks the interaction between PD1 and B7-H1 (B7-DCIg; Amplimmune; e.g., disclosed in WO2010/027827 and WO2011/066342).
  • a combination described herein includes a LAG-3 inhibitor.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor or a hematologic malignancy.
  • the anti-LAG-3 antibody is BMS-986016.
  • BMS-986016 also referred to as BMS986016; Bristol-Myers Squibb
  • BMS-986016 and other humanized anti-LAG-3 antibodies are disclosed in US 2011/0150892, WO2010/019570, and WO2014/008218.
  • the anti-LAG-3 antibody is a humanized anti-LAG3 antibody disclosed in WO2015/138920.
  • a combination described herein includes a TIM-3 inhibitor.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor or a hematologic malignancy.
  • anti-TIM-3 antibodies are disclosed in U.S. Pat. No. 8,552,156, WO 2011/155607, EP 2581113 and U.S Publication No.: 2014/044728.
  • the anti-TIM3 is a humanized ABTIM3 mAb disclosed in WO2015/117002.
  • a combination described herein includes a CTLA-4 inhibitor.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor or a hematologic malignancy.
  • Exemplary anti-CTLA4 antibodies include Tremelimumab (IgG2 monoclonal antibody available from Pfizer, formerly known as ticilimumab, CP-675,206); and Ipilimumab (CTLA-4 antibody, also known as MDX-010, CAS No. 477202-00-9).
  • Tremelimumab IgG2 monoclonal antibody available from Pfizer, formerly known as ticilimumab, CP-675,206
  • Ipilimumab CLA-4 antibody, also known as MDX-010, CAS No. 477202-00-9
  • the combination includes an anti-GITR antibody molecule, e.g., as described herein, and an anti-CTLA-4 antibody, e.g., ipilimumab.
  • an anti-GITR antibody molecule e.g., as described herein
  • an anti-CTLA-4 antibody e.g., ipilimumab
  • Exemplary doses that can be use include a dose of anti-GITR antibody molecule of about 1 to 10 mg/kg, e.g., 3 mg/kg, and a dose of an anti-CTLA-4 antibody, e.g., ipilimumab, of about 3 mg/kg.
  • anti-CTLA-4 antibodies are disclosed, e.g., in U.S. Pat. No. 5,811,097.
  • a combination described herein includes an inhibitor of Inhibitor of Apoptosis Protein (IAP).
  • IAP Inhibitor of Apoptosis Protein
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a breast cancer, an ovarian cancer, or a pancreatic cancer), e.g., a hematologic malignancy (e.g., a multiple myeloma).
  • the IAP inhibitor is (S)—N—((S)-1-cyclohexyl-2-((S)-2-(4-(4-fluorobenzoyl)thiazol-2-yl)pyrrolidin-1-yl)-2-oxoethyl)-2-(methylamino)propanamide (Compound A21) or a compound disclosed in U.S. Pat. No. 8,552,003.
  • the IAP inhibitor e.g., (S)—N—((S)-1-cyclohexyl-2-((S)-2-(4-(4-fluorobenzoyl)thiazol-2-yl)pyrrolidin-1-yl)-2-oxoethyl)-2-(methylamino)propanamide (Compound A21) or a compound disclosed in U.S. Pat. No. 8,552,003, is administered at a dose of approximately 1800 mg, e.g., once weekly.
  • a combination described herein includes an inhibitor of Epidermal Growth Factor Receptor (EGFR).
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a lung cancer (e.g., a non-small cell lung cancer), a pancreatic cancer, a breast cancer, or a colon cancer).
  • a cancer e.g., a cancer described herein, e.g., a solid tumor (e.g., a lung cancer (e.g., a non-small cell lung cancer), a pancreatic cancer, a breast cancer, or a colon cancer).
  • the EGFR inhibitor is (R,E)-N-(7-chloro-1-(1-(4-(dimethylamino)but-2-enoyl)azepan-3-yl)-1H-benzo[d]imidazol-2-yl)-2-methylisonicotinamide (Compound A40) or a compound disclosed in PCT Publication No. WO 2013/184757.
  • the EGFR inhibitor e.g., (R,E)-N-(7-chloro-1-(1-(4-(dimethylamino)but-2-enoyl)azepan-3-yl)-1H-benzo[d]imidazol-2-yl)-2-methylisonicotinamide (Compound A40) or a compound disclosed in PCT Publication No. WO 2013/184757, is administered at a dose of 150-250 mg, e.g., per day.
  • the EGFR inhibitor e.g., (R,E)-N-(7-chloro-1-(1-(4-(dimethylamino)but-2-enoyl)azepan-3-yl)-1H-benzo[d]imidazol-2-yl)-2-methylisonicotinamide (Compound A40) or a compound disclosed in PCT Publication No. WO 2013/184757, is administered at a dose of about 150, 200, or 250 mg, or about 150-200 or 200-250 mg.
  • the EGFR inhibitor is chosen from one of more of erlotinib, gefitinib, cetuximab, panitumumab, necitumumab, PF-00299804, nimotuzumab, or RO5083945.
  • a combination described herein includes an inhibitor of target of rapamycin (mTOR).
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a prostate cancer, a breast cancer, a brain cancer, a bladder cancer, a pancreatic cancer, a renal cancer, or a liver cancer, a lung cancer (e.g., a small cell lung cancer or a non-small cell lung cancer), a respiratory/thoracic cancer, a sarcoma, a bone cancer, a non-small cell lung cancer, an endocrine cancer, an astrocytoma, a cervical cancer, a neurologic cancer, a gastric cancer, or a melanoma), e.g., a hematologic malignancy (e.g., a leukemia (e.g., lymphocytic leukemia), e.g., a lympho
  • the mTOR inhibitor is dactolisib (Compound A4) or 8-(6-Methoxy-pyridin-3-yl)-3-methyl-1-(4-piperazin-1-yl-3-trifluoromethyl-phenyl)-1,3-dihydro-imidazo[4,5-c]quinolin-2-one (Compound A41), or a compound disclosed in PCT Publication No. WO 2006/122806.
  • the mTOR inhibitor is everolimus (also known as AFINITOR®; Compound A36) or a compound disclosed in PCT Publication No. WO 2014/085318.
  • the mTOR inhibitor e.g., everolimus (Compound A36) or a compound disclosed in PCT Publication No. WO 2014/085318
  • the TOR inhibitor e.g., everolimus (Compound A36) or a compound disclosed in PCT Publication No. WO 2014/085318
  • the mTOR inhibitor is chosen from one or more of rapamycin, temsirolimus (TORISEL®), AZD8055, BEZ235, BGT226, XL765, PF-4691502, GDC0980, SF1126, OSI-027, GSK1059615, KU-0063794, WYE-354, Palomid 529 (P529), PF-04691502, or PKI-587.
  • TORISEL® temsirolimus
  • AZD8055 BEZ235
  • ridaforolimus (formally known as deferolimus, (1R,2R,4S)-4-[(2R)-2 [(1R,9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28Z,30S,32S,35R)-1,18-dihydroxy-19,30-dimethoxy-15,17,21,23, 29,35-hexamethyl-2,3,10,14,20-pentaoxo-11,36-dioxa-4-azatricyclo[30.3.1.04,9] hexatriaconta-16,24,26,28-tetraen-12-yl]propyl]-2-methoxycyclohexyl dimethylphosphinate, also known as AP23573 and MK8669, and described in PCT Publication No.
  • WO 03/064383 everolimus (AFINITOR® or RAD001); rapamycin (AY22989, SIROLIMUS®); simapimod (CAS Registry Number: 164301-51-3); (5- ⁇ 2,4-Bis[(3S)-3-methylmorpholin-4-yl]pyrido[2,3-d]pyrimidin-7-yl ⁇ -2-methoxyphenyl)methanol (AZD8055); 2-Amino-8-[trans-4-(2-hydroxyethoxy)cyclohexyl]-6-(6-methoxy-3-pyridinyl)-4-methyl-pyrido[2,3-d]pyrimidin-7(8H)-one (PF04691502, CAS Registry Number: 1013101-36-4); N2-[1,4-dioxo-4-[[4-(4-oxo-8-phenyl-4H-1-benzopyran-2-yl)morpholinium-4-yl]methoxy]butyl]-L-arginy
  • exemplary mTOR Inhibitors include, but are not limited to, temsirolimus; ridaforolimus (1R,2R,4S)-4-[(2R)-2 [(1R,9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28Z,30S,32S,35R)-1,18-dihydroxy-19,30-dimethoxy-15,17,21,23,29,35-hexamethyl-2,3,10,14,20-pentaoxo-11,36-dioxa-4-azatricyclo[30.3.1.0 4,9 ] hexatriaconta-16,24,26,28-tetraen-12-yl]propyl]-2-methoxycyclohexyl dimethylphosphinate, also known as AP23573 and MK8669; everolimus (RAD001); rapamycin (AY22989); simapimod; (5- ⁇ 2,4-bis[
  • a combination described herein includes an interleukin-15 (IL-15) agonist.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a refractory solid tumor), (e.g., a melanoma (e.g., a metastatic or advanced melanoma), a kidney cancer (e.g., a renal cell cancer), a non-small cell lung cancer, a squamous cell head and neck cancer, or a bladder cancer (e.g., a non-muscle invasive bladder cancer)), e.g., a hematologic malignancy (e.g., a leukemia, e.g., an acute myelogenous leukemia (e.g., a refractory or relapsed acute myelogenous leukemia), e.g., a lymphoma, e.
  • IL-15 secreted by mononuclear phagocytes (and some other cell types) following viral infection, regulates T and natural killer cell activation and proliferation.
  • This cytokine induces activation of transcription activators STAT3, STATS, and STATE via JAK kinase signal transduction pathways in mast cells, T cells, and dendritic epidermal T cells.
  • IL-15 and interleukin-2 (IL-2) are structurally similar and share many biological activities; both may bind to common hematopoietin receptor subunits, negatively regulating each other's activity.
  • CD8+ memory T cell number can be regulated by a balance between IL-15 and IL-2.
  • the IL-15 agonist is a recombinant human IL-15 (rhIL-15), e.g., CYP0150 (Cytune).
  • CYP0150 is a recombinant protein consisting of a human IL-15 linked to the Sushi+ domain of the human alpha chain receptor (transpresentation).
  • CYP0150 is disclosed, e.g., in PCT Publication No. WO 2007/046006.
  • CYP0150 has the amino acid disclosed as SEQ ID NO: 60 in WO 2007/046006 or disclosed as SEQ ID NO: 62 in WO 2007/046006.
  • the IL-15 agonist is ALT-803 (Altor BioScience).
  • ALT-803 is an IL-15N72D:IL-15R ⁇ Su/Fc soluble complex, produced from a high-yield recombinant mammalian cell line that co-expresses IL-15N72D and IL-15R ⁇ Su/Fc fusion protein.
  • the IL-15 mutant (N72D) has enhanced IL-15 biological activity (Zhu et al. 2009 , J Immunol. 183:3598).
  • IL-15N72D mutant and the soluble domain of IL-15R ⁇ can form stable heterodimeric complexes in solution and this complex exhibits increased biological activity (approximately 25-fold more active) compared to the non-complexed IL-15.
  • ALT-803 is disclosed, e.g., in PCT Publication No. WO 2012/040323 and U.S. Pat. No. 8,507,222.
  • the IL-15 agonist is hetlL-15 (Admune).
  • HetIL-15 is a heterodimeric human IL-15 (IL-15/sIL-15Ra).
  • HetIL-15 is disclosed, e.g., in PCT Publication Nos. WO 2009/002562 and WO 2014/066527.
  • the combination includes a CD40 agonist.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a lung cancer, an esophageal carcinoma, a melanoma, or a renal cell carcinoma), e.g., a hematologic malignancy (e.g., a leukemia (e.g., a chronic lymphocytic leukemia (CLL)), e.g., a lymphoma (e.g., a non-Hodgkin's lymphoma), e.g., or a multiple myeloma).
  • a cancer e.g., a cancer described herein, e.g., a solid tumor (e.g., a lung cancer, an esophageal carcinoma, a melanoma, or a renal cell carcinoma), e.g., a hematologic mal
  • the CD40 agonist is ADC-1013 (Alligator/Biolnvent).
  • ADC-1013 is a fully human IgG agonistic monoclonal antibody against human CD40.
  • CD40 an integral membrane protein found on the surface of B lymphocytes, is a member of the tumor necrosis factor receptor superfamily and is highly expressed in a number of cancers such as B-cell malignancies.
  • CD40 agonists e.g., anti-CD40 antibodies, are able to substitute effectively for T cell helper activity (Ridge, J. et al. (1998) Nature 393: 474-478).
  • ADC-1013 is disclosed, e.g., in PCT Publication No. WO 2015/091853.
  • ADC-1013 clones include, e.g., 1136/1137, 1132/1133, 1148/1149, 1140/1135, 1134/1135, 1107/1108, 1142/1135, 1146/1147, and 1150/1151.
  • the heavy chain variable region of 1132/1133 has the amino acid sequence disclosed as SEQ ID NO: 65 in WO 2015/091853.
  • the light chain variable region of 1132/1133 has the amino acid sequence disclosed as SEQ ID NO: 66 in WO 2015/091853.
  • the heavy chain CDR1 of 1132/1133 has the amino acid sequence disclosed as SEQ ID NO: 13 in WO 2015/091853.
  • the heavy chain CDR2 of 1132/1133 has the amino acid sequence disclosed as SEQ ID NO: 14 in WO 2015/091853.
  • the heavy chain CDR3 of 1132/1133 has the amino acid sequence disclosed as SEQ ID NO: 15 in WO 2015/091853.
  • the light chain CDR1 of 1132/1133 has the amino acid sequence disclosed as SEQ ID NO: 16 in WO 2015/091853.
  • the light chain CDR2 of 1132/1133 has the amino acid sequence disclosed as SEQ ID NO: 17 in WO 2015/091853).
  • the light chain CDR3 of 1132/1133 has the amino acid sequence disclosed as SEQ ID NO: 18 in WO 2015/091853.
  • the heavy chain variable region of 1107/1108 has the amino acid sequence disclosed as SEQ ID NO: 79 in WO 2015/091853.
  • the light chain variable region of 1107/1108 has the amino acid sequence disclosed as SEQ ID NO: 80 in WO 2015/091853.
  • the heavy chain CDR1 of 1107/1108 has the amino acid sequence disclosed as SEQ ID NO: 55 in WO 2015/091853.
  • the heavy chain CDR2 of 1107/1108 has the amino acid sequence disclosed as SEQ ID NO: 56 in WO 2015/091853.
  • the heavy chain CDR3 of 1107/1108 has the amino acid sequence disclosed as SEQ ID NO: 57 in WO 2015/091853.
  • the light chain CDR1 of 1107/1108 has the amino acid sequence disclosed as SEQ ID NO: 58 in WO 2015/091853.
  • the light chain CDR2 of 1107/1108 has the amino acid sequence disclosed as SEQ ID NO: 59 in WO 2015/091853.
  • the light chain CDR3 of 1107/1108 has the amino acid sequence disclosed as SEQ ID NO: 60 in WO 2015/091853.
  • the CD40 agonist is ISF35.
  • ISF35 is a chimeric CD154. ISF is disclosed in PCT Publication Nos. WO 2003/099340 and WO 2008/070743.
  • the CD40 agonist is dacetuzumab.
  • Dacetuzumab is also known as SGN-40 or huS2C6.
  • Dacetuzumab is a humanized monoclonal antibody that targets CD40.
  • Dacetuzumab is disclosed, e.g., in Advani et al. J Clin Oncol. 2009; 27(26):4371-7; and Khubchandani et al. Curr Opin Investig Drugs. 2009; 10(6):579-87.
  • the CD40 agonist is lucatumumab (CAS Registry Number: 903512-50-5).
  • Lucatumumab is also known as CHIR-12.12 or HCD-122.
  • Lucatumumab binds to and inhibits CD40, thereby inhibiting CD40 ligand-induced cell proliferation and triggering cell lysis via antibody-dependent cellular cytotoxicity (ADCC) in cells overexpressing CD40.
  • ADCC antibody-dependent cellular cytotoxicity
  • Anti-CD40 antibodies are able to substitute effectively for T cell helper activity (Ridge, J. et al. (1998) Nature 393: 474-478) and can be used in conjunction with PD-1 antibodies (Ito, N. et al. (2000) Immunobiology 201 (5) 527-40).
  • a combination described herein includes an OX40 agonist.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a breast cancer, a melanoma, a head and neck cancer, or a prostate cancer), e.g., a hematologic malignancy (e.g., a lymphoma (e.g., a B-cell lymphoma)).
  • a cancer e.g., a cancer described herein, e.g., a solid tumor (e.g., a breast cancer, a melanoma, a head and neck cancer, or a prostate cancer), e.g., a hematologic malignancy (e.g., a lymphoma (e.g., a B-cell lymphoma)).
  • a cancer described herein e.g., a solid tumor (e.g.,
  • OX40 also known as CD134, is a cell surface glycoprotein and member of the tumor necrosis factor (TNF) receptor superfamily, is expressed on T-lymphocytes and provides a co-stimulatory signal for the proliferation and survival of activated T-cells. OX40 activation can induce proliferation of effector T-lymphocytes, which promotes an immune response against the tumor cells that express tumor-associated antigens (TAAs).
  • TNF tumor necrosis factor
  • the OX40 agonist is chosen from mAb 106-222, humanized 106-222 (Hu106), mAb 119-122, or humanized 119-122 (Hu119).
  • MAb 106-222, humanized 106-222 (Hu106), mAb 119-122, and humanized 119-122 (Hu119) are disclosed, e.g., in PCT Publication No. WO 2012/027328 and U.S. Pat. No. 9,006,399.
  • the amino acid sequence of the heavy chain variable region of mAb 106-222 is disclosed as SEQ ID NO: 4 in WO 2012/027328.
  • the amino acid sequence of the light chain variable region of mAb 106-222 is disclosed as SEQ ID NO: 10 in WO 2012/027328.
  • the amino acid sequence of the heavy chain variable region of humanized 106-222 (Hu106) is disclosed as SEQ ID NO: 5 in WO 2012/027328.
  • the amino acid sequence of the light chain variable region of humanized 106-222 (Hu106) is disclosed as SEQ ID NO: 11 in WO 2012/027328.
  • the amino acid sequence of the heavy chain variable region of mAb 119-122 is disclosed as SEQ ID NO: 16 in WO 2012/027328.
  • the amino acid sequence of the light chain variable region of mAb 119-122 is disclosed as SEQ ID NO: 22 in WO 2012/027328.
  • the amino acid sequence of the heavy chain variable region of humanized 119-122 (Hu119) is disclosed as SEQ ID NO: 17 in WO 2012/027328.
  • the amino acid sequence of the light chain variable region of humanized 119-122 (Hu119) is disclosed as SEQ ID NO: 23 in WO 2012/027328.
  • the OX40 agonist is a humanized monoclonal antibody disclosed in U.S. Pat. No. 7,959,925 and PCT Publication No. WO 2006/121810.
  • the OX40 agonist is chosen from MEDI6469, MEDI0562, or MEDI6383.
  • MEDI6469 is a murine monoclonal antibody against OX40.
  • MEDI0562 is a humanized monoclonal antibody against OX40.
  • MEDI6383 is a monoclonal antibody against OX40.
  • the OX40 agonist e.g., MEDI6469
  • a combination described herein includes a CD27 agonist.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a melanoma, a renal cell carcinoma, a hormone-refractory prostate adenocarcinoma, an ovarian cancer, a breast cancer, a colorectal adenocarcinoma, or a non-small cell lung cancer), e.g., a hematologic malignancy (e.g., a lymphoma (e.g., a Hodgkin's lymphoma, a Burkett's lymphoma, a mantle cell lymphoma, a primary lymphoma of the central nervous system, or a marginal zone B-cell lymphoma), or a leukemia (e.g., a chronic lymphocytic leukemia (CLL)).
  • a cancer described herein e
  • the CD27 agonist is Varlilumab (CAS Registry Number: 1393344-72-3).
  • Varlilumab is also known as CDX-1127 (Celldex) or 1F5.
  • Varlilumab is a fully human monoclonal antibody (mAb) that targets CD27, molecule in the activation pathway of lymphocytes.
  • CDX-1127 is an agonist anti-CD27 mAb that can activate human T cells in the context of T cell receptor stimulation and therefore mediate anti-tumor effects. CDX-1127 can also provide direct therapeutic effects against tumors with CD27 expression.
  • Varlilumab is disclosed, e.g., in Vitale et al., Clin Cancer Res. 2012; 18(14):3812-21, WO 2008/051424, and U.S. Pat. No. 8,481,029.
  • the CD27 agonist is BION-1402 (BioNovion).
  • BION-1402 is also known as hCD27.15.
  • BION-1402 is an anti-human CD27 monoclonal antibody.
  • BION-1402 can stimulate the proliferation and/or survival of CD27+ cells.
  • BION-1402 can activate human CD27 more effectively than its ligand CD70, which results in a significantly increased effect on proliferation of CD8+ and CD4+ T-cells.
  • BION-1402 is disclosed, e.g., as hCD27.15 in WO 2012/004367.
  • This antibody is produced by hybridoma hCD27.15, which was deposited with the ATCC in on Jun. 2, 2010 under number PTA-11008.
  • the heavy chain variable region of hCD27.15 has the amino acid sequence disclosed as SEQ ID NO: 3 in WO 2012/004367.
  • the light chain variable region of hCD27.15 has the amino acid sequence disclosed as SEQ ID NO: 4 in WO 2012/004367.
  • the heavy chain CDR1 of hCD27.15 has the amino acid sequence disclosed as SEQ ID NO: 5 in WO 2012/004367.
  • the heavy chain CDR2 of hCD27.15 has the amino acid sequence disclosed as SEQ ID NO: 6 in WO 2012/004367.
  • the heavy chain CDR3 of hCD27.15 has the amino acid sequence disclosed as SEQ ID NO: 7 in WO 2012/004367.
  • the light chain CDR1 of hCD27.15 has the amino acid sequence disclosed as SEQ ID NO: 8 in WO 2012/004367.
  • the light chain CDR2 of hCD27.15 has the amino acid sequence disclosed as SEQ ID NO: 9 in WO 2012/004367.
  • the light chain CDR3 of hCD27.15 has the amino acid sequence disclosed as SEQ ID NO: 10 in WO 2012/004367.
  • a combination described herein includes a CSF-1/1R binding agent.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a prostate cancer, a breast cancer, or pigmented villonodular synovitis (PVNS)).
  • a cancer e.g., a cancer described herein, e.g., a solid tumor (e.g., a prostate cancer, a breast cancer, or pigmented villonodular synovitis (PVNS)).
  • a cancer described herein e.g., a solid tumor (e.g., a prostate cancer, a breast cancer, or pigmented villonodular synovitis (PVNS)).
  • PVNS pigmented villonodular synovitis
  • the CSF-1/1R binding agent is an inhibitor of macrophage colony-stimulating factor (M-CSF).
  • M-CSF macrophage colony-stimulating factor
  • the CSF-1/1R binding agent is a CSF-1R tyrosine kinase inhibitor, 4-((2-(((1R,2R)-2-hydroxycyclohexyl)amino)benzo[d]thiazol-6-yl)oxy)-N-methylpicolinamide (Compound A15), or a compound disclosed in PCT Publication No. WO 2005/073224.
  • the CSF-1/1R binding agent is an M-CSF inhibitor, Compound A33, or a compound disclosed in PCT Publication No. WO 2004/045532 (e.g., an antibody molecule or Fab fragment against M-CSF).
  • the CSF-1/1R binding agent e.g., an M-CSF inhibitor, Compound A33, or a binding agent to CSF-1 disclosed in PCT Publication No. WO 2004/045532 or PCT Publication No. WO 2005/068503 including RX1 or 5H4 (e.g., an antibody molecule or Fab fragment against M-CSF), is administered at an average dose of about 10 mg/kg.
  • the CSF-1/1R binding agent is a CSF1R inhibitor or 4-(2-((1R,2R)-2-hydroxycyclohexylamino)benzothiazol-6-yloxy)-N-methylpicolinamide.
  • the CSF-1/1R binding agent is pexidartinib (CAS Registry Number 1029044-16-3).
  • Pexidrtinib is also known as PLX3397 or 5-((5-chloro-1H-pyrrolo[2,3-b]pyridin-3-yl)methyl)-N-((6-(trifluoromethyl)pyridin-3-yl)methyl)pyridin-2-amine.
  • Pexidartinib is a small-molecule receptor tyrosine kinase (RTK) inhibitor of KIT, CSF1R and FLT3.
  • FLT3, CSF1R and FLT3 are overexpressed or mutated in many cancer cell types and play major roles in tumor cell proliferation and metastasis.
  • PLX3397 can bind to and inhibit phosphorylation of stem cell factor receptor (KIT), colony-stimulating factor-1 receptor (CSF1R) and FMS-like tyrosine kinase 3 (FLT3), which may result in the inhibition of tumor cell proliferation and down-modulation of macrophages, osteoclasts and mast cells involved in the osteolytic metastatic disease.
  • the CSF-1/1R binding agent e.g., pexidartinib
  • a GITR agonist e.g., an anti-GITR antibody molecule provided herein.
  • the CSF-1/1R binding agent is emactuzumab.
  • Emactuzumab is also known as RG7155 or RO5509554.
  • Emactuzumab is a humanized IgG1 mAb targeting CSF1R.
  • the CSF-1/1R binding agent e.g., pexidartinib
  • a PD-L1 inhibitor e.g., an anti-PD-L1 antibody molecule described herein.
  • the CSF-1/1R binding agent is FPA008.
  • FPA008 is a humanized mAb that inhibits CSF1R.
  • the CSF-1/1R binding agent, e.g., FPA008 is used in combination with a GITR agonist, e.g., an anti-GITR antibody molecule described herein.
  • a combination described herein includes an interleukine-17 (IL-17) inhibitor.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor, e.g., breast cancer, lung cancer, or colon cancer.
  • the IL-17 inhibitor is secukinumab (CAS Registry Numbers: 875356-43-7 (heavy chain) and 875356-44-8 (light chain)).
  • Secukinumab is also known as AIN457 and COSENTYX®.
  • Secukinumab is a recombinant human monoclonal IgG1/ ⁇ antibody that binds specifically to IL-17A. It is expressed in a recombinant Chinese Hamster Ovary (CHO) cell line.
  • Secukinumab is described, e.g., in WO 2006/013107, U.S. Pat. No. 7,807,155, U.S. Pat. No. 8,119,131, U.S. Pat. No. 8,617,552, and EP 1776142.
  • the heavy chain variable region of secukinumab has the amino acid sequence disclosed as SEQ ID NO: 8 in WO 2006/013107.
  • the light chain variable region of secukinumab has the amino acid sequence disclosed as SEQ ID NO: 10 in WO 2006/013107.
  • the heavy chain CDR1 of secukinumab has the amino acid sequence disclosed as SEQ ID NO: 1 in WO 2006/013107.
  • the heavy chain CDR2 of secukinumab has the amino acid sequence disclosed as SEQ ID NO: 2 in WO 2006/013107.
  • the heavy chain CDR3 of secukinumab has the amino acid sequence disclosed as SEQ ID NO: 3 in WO 2006/013107.
  • the light chain CDR1 of secukinumab has the amino acid sequence disclosed as SEQ ID NO: 4 in WO 2006/013107.
  • the light chain CDR2 of secukinumab has the amino acid sequence disclosed as SEQ ID NO: 5 in WO 2006/013107.
  • the light chain CDR3 of secukinumab has the amino acid sequence disclosed as SEQ ID NO: 6 in WO 2006/013107.
  • the IL-17 inhibitor is CJM112.
  • CJM112 is also known as XAB4.
  • CJM112 is a fully human monoclonal antibody that targets IL-17A.
  • CJM112 is disclosed, e.g., in WO 2014/122613.
  • the heavy chain of CJM112 has the amino acid sequence disclosed as SEQ ID NO: 14 in WO 2014/122613.
  • the light chain of CJM112 has the amino acid sequence disclosed as SEQ ID NO: 44 in WO 2014/122613.
  • the IL-17 inhibitor is ixekizumab (CAS Registry Number: 1143503-69-8).
  • Ixekizumab is also known as LY2439821.
  • Ixekizumab is a humanized IgG4 monoclonal antibody that targets IL-17A.
  • Ixekizumab is described, e.g., in WO 2007/070750, U.S. Pat. No. 7,838,638, and U.S. Pat. No. 8,110,191.
  • the heavy chain variable region of ixekizumab has the amino acid sequence disclosed as SEQ ID NO: 118 in WO 2007/070750.
  • the light chain variable region of ixekizumab has the amino acid sequence disclosed as SEQ ID NO: 241 in WO 2007/070750.
  • the IL-17 inhibitor is brodalumab (CAS Registry Number: 1174395-19-7). Brodalumab is also known as AMG 827 or AM-14. Brodalumab binds to the interleukin-17 receptor A (IL-17RA) and prevents IL-17 from activating the receptor.
  • IL-17RA interleukin-17 receptor A
  • Brodalumab is disclosed, e.g., in WO 2008/054603, U.S. Pat. No. 7,767,206, U.S. Pat. No. 7,786,284, U.S. Pat. No. 7,833,527, U.S. Pat. No. 7,939,070, U.S. Pat. No. 8,435,518, U.S. Pat. No. 8,545,842, U.S. Pat. No. 8,790,648, and U.S. Pat. No. 9,073,999.
  • the heavy chain CDR1 of brodalumab has the amino acid sequence disclosed as SEQ ID NO: 146 in WO 2008/054603.
  • the heavy chain CDR2 of brodalumab has the amino acid sequence disclosed as SEQ ID NO: 147 in WO 2008/054603.
  • the heavy chain CDR3 of brodalumab has the amino acid sequence disclosed as SEQ ID NO: 148 in WO 2008/054603.
  • the light chain CDR1 of brodalumab has the amino acid sequence disclosed as SEQ ID NO: 224 in WO 2008/054603.
  • the heavy chain CDR2 of brodalumab has the amino acid sequence disclosed as SEQ ID NO: 225 in WO 2008/054603.
  • the heavy chain CDR3 of brodalumab has the amino acid sequence disclosed as SEQ ID NO: 226 in WO 2008/054603.
  • a combination described herein includes an interleukine-1 beta (IL-1 ⁇ ) inhibitor.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a hematologic malignancy (e.g., a lymphoma (e.g., Hodgkin lymphoma), a leukemia (e.g., an acute or chronic leukemia), or a multiple myeloma).
  • a cancer e.g., a cancer described herein, e.g., a hematologic malignancy (e.g., a lymphoma (e.g., Hodgkin lymphoma), a leukemia (e.g., an acute or chronic leukemia), or a multiple myeloma).
  • a cancer described herein e.g., a hematologic malignancy (e.g., a lymphoma (e.g., Hodgkin lymph
  • the IL-1 ⁇ inhibitor is canakinumab.
  • Canakinumab is also known as ACZ885 or ILARIS®.
  • Canakinumab is a human monoclonal IgG1/ ⁇ antibody that neutralizes the bioactivity of human IL-1 ⁇ .
  • Canakinumab is disclosed, e.g., in WO 2002/16436, U.S. Pat. No. 7,446,175, and EP 1313769.
  • the heavy chain variable region of canakinumab has the amino acid sequence disclosed as SEQ ID NO: 1 in U.S. Pat. No. 7,446,175.
  • the light chain variable region of canakinumab has the amino acid sequence disclosed as SEQ ID NO: 2 in U.S. Pat. No. 7,446,175.
  • a combination described herein includes an inhibitor of chemokine (C—X—C motif) receptor 2 (CXCR2) inhibitor.
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor, e.g., a breast cancer, a metastatic sarcoma, a pancreatic cancer, a melanoma, a renal cell carcinoma (RCC), a non-small cell lung cancer (NSCLC), or a pediatric tumor (e.g., a rhabdomyosarcoma).
  • a cancer described herein e.g., a solid tumor, e.g., a breast cancer, a metastatic sarcoma, a pancreatic cancer, a melanoma, a renal cell carcinoma (RCC), a non-small cell lung cancer (NSCLC), or a pediatric tumor (e.g., a rhabdomyosarcoma).
  • the CXCR2 inhibitor is danirixin (CAS Registry Number: 954126-98-8).
  • Danirixin is also known as GSK1325756 or 1-(4-chloro-2-hydroxy-3-piperidin-3-ylsulfonylphenyl)-3-(3-fluoro-2-methylphenyl)urea. Danirixin is disclosed, e.g., in Miller et al. Eur J Drug Metab Pharmacokinet (2014) 39:173-181; and Miller et al. BMC Pharmacology and Toxicology (2015), 16:18.
  • the CXCR2 inhibitor is reparixin (CAS Registry Number: 266359-83-5).
  • Reparixin is also known as repertaxin or (2R)-2-[4-(2-methylpropyl)phenyl]-N-methylsulfonylpropanamide.
  • Reparixin is a non-competitive allosteric inhibitor of CXCR1/2. Reparixin is disclosed, e.g., in Zarbock et al. British Journal of Pharmacology (2008), 1-8.
  • the CXCR2 inhibitor is navarixin.
  • Navarixin is also known as MK-7123, SCH 527123, PS291822, or 2-hydroxy-N,N-dimethyl-3-[[2-[[(1R)-1-(5-methylfuran-2-yl)propyl]amino]-3,4-dioxocyclobuten-1-yl]amino]benzamide.
  • Navarixin is disclosed, e.g., in Ning et al. Mol Cancer Ther. 2012; 11(6):1353-64.
  • a combination described herein includes an inhibitor of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K), e.g., phosphatidylinositol-4,5-bisphosphate 3-kinase gamma and/or delta (PI3K- ⁇ , ⁇ ).
  • PI3K phosphatidylinositol-4,5-bisphosphate 3-kinase
  • PI3K phosphatidylinositol-4,5-bisphosphate 3-kinase gamma and/or delta
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a prostate cancer, a breast cancer, a brain cancer, a bladder cancer, a pancreatic cancer, a renal cancer, a solid tumor, a liver cancer, a non-small cell lung cancer, an endocrine cancer, an ovarian cancer, a melanoma, a female reproductive system cancer, a digestive/gastrointestinal cancer, a glioblastoma multiforme, a head and neck cancer, or a colon cancer), e.g., a hematologic malignancy (e.g., a leukemia (e.g., a lymphocytic leukemia, e.g., chronic lymphocytic leukemia (CLL) (e.g., relapsed CLL)),e.g., a lymphoma (e.g., non-Hodgkin
  • the PI3K inhibitor is an inhibitor of delta and gamma isoforms of PI3K.
  • Exemplary PI3K inhibitors that can be used in combination are described in, e.g., WO 2010/036380, WO 2010/006086, WO 09/114870, WO 05/113556, GSK 2126458, GDC-0980, GDC-0941, Sanofi XL147, XL756, XL147, PF-46915032, BKM 120, CAL-101, CAL 263, SF1126, PX-886, and a dual PI3K inhibitor.
  • the PI3K- ⁇ , ⁇ inhibitor is idelalisib (CAS Registry Number: 870281-82-6).
  • Idelalisib is also known as ZYDELIG®, GS-1101, CAL-101, or 5-Fluoro-3-phenyl-2-[(1S)-1-(7H-purin-6-ylamino)propyl]-4(3H)-quinazolinone.
  • Idelalisib blocks P110 ⁇ , the delta isoform of PI3K. Idelalisib is disclosed, e.g., in Wu et al. Journal of Hematology & Oncology (2013) 6: 36.
  • the PI3K- ⁇ , ⁇ inhibitor is dactolisib (Compound A4) or 8-(6-Methoxy-pyridin-3-yl)-3-methyl-1-(4-piperazin-1-yl-3-trifluoromethyl-phenyl)-1,3-dihydro-imidazo[4,5-c]quinolin-2-one (Compound A41), or a compound disclosed in PCT Publication No. WO 2006/122806.
  • the PI3K- ⁇ , ⁇ inhibitor is buparlisib (Compound A6) or a compound disclosed in PCT Publication No. WO 2007/084786.
  • the PI3K- ⁇ , ⁇ inhibitor e.g., buparlisib (Compound A6) or a compound disclosed in PCT Publication No. WO 2007/084786, is administered at a dose of about 100 mg (e.g., per day).
  • PI3K- ⁇ , ⁇ inhibitors that can be used in the combination include, e.g., pictilisib (GDC-0941), LY294002, pilaralisib (XL147), PI-3065, PI-103, VS-5584 (SB2343), CZC24832, duvelisib (IPI-145, INK1197), TG100-115, CAY10505, GSK1059615, PF-04691502, AS-605240, voxtalisib (SAR245409, XL765), IC-87114, omipalisib (GSK2126458, GSK458), TG100713, gedatolisib (PF-05212384, PKI-587), PKI-402, XL147 analogue, PIK-90, PIK-293, PIK-294, 3-Methyladenine (3-MA), AS-252424, AS-604850, or apitolis
  • the PI3K inhibitor is Compound A8 or a compound described in PCT Publication No. WO2010/029082.
  • the PI3K inhibitor is a pan-PI3K inhibitor, (4S,5R)-3-(2′-amino-2-morpholino-4′-(trifluoromethyl)-[4,5′-bipyrimidin]-6-yl)-4-(hydroxymethyl)-5-methyloxazolidin-2-one (Compound A13) or a compound disclosed in PCT Publication No. WO2013/124826.
  • Exemplary PI3K- ⁇ , - ⁇ inhibitors include, but are not limited to, duvelisib and idelalisib.
  • Idelalisib also called GS-1101 or CAL-101; Gilead
  • GS-1101 or CAL-101 Gilead
  • Gilead is a small molecule that blocks the delta isoform of PI3K.
  • the structure of idelalisib (5-Fluoro-3-phenyl-2-[(1S)-1-(7H-purin-6-ylamino)propyl]-4(3H)-quinazolinone) is shown below.
  • Duvelisib (also called IPI-145; Infinity Pharmaceuticals and Abbvie) is a small molecule that blocks PI3K- ⁇ , ⁇ .
  • the structure of duvelisib (8-Chloro-2-phenyl-3-[(1S)-1-(9H-purin-6-ylamino)ethyl]-1(2H)-isoquinolinone) is shown below.
  • the inhibitor is a dual phosphatidylinositol 3-kinase (PI3K) and mTOR inhibitor selected from 2-Amino-8-[trans-4-(2-hydroxyethoxy)cyclohexyl]-6-(6-methoxy-3-pyridinyl)-4-methyl-pyrido[2,3-d]pyrimidin-7(8H)-one (PF-04691502); N-[4-[[4-(Dimethylamino)-1-piperidinyl]carbonyl]phenyl]-N-[4-(4,6-di-4-morpholinyl-1,3,5-triazin-2-yl)phenyl]urea (PF-05212384, PKI-587); 2-Methyl-2- ⁇ 4-[3-methyl-2-oxo-8-(quinolin-3-yl)-2,3-dihydro-1H-imidazo[4,5-c]quinolin-1-yl]phenyl ⁇ propan
  • a combination described herein includes a B-cell-activating factor receptor (BAFF-R) inhibitor.
  • BAFF-R B-cell-activating factor receptor
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a hematologic malignancy, e.g., a leukemia (e.g., chronic lymphocytic leukemia (CLL), e.g., relapsed or refractory chronic lymphocytic leukemia).
  • a cancer e.g., a cancer described herein, e.g., a hematologic malignancy, e.g., a leukemia (e.g., chronic lymphocytic leukemia (CLL), e.g., relapsed or refractory chronic lymphocytic leukemia).
  • CLL chronic lymphocytic leukemia
  • the BAFF-R inhibitor is VAY736.
  • VAY736 is a fully human combinatorial antibody library (HuCAL)-derived monoclonal antibody targeting BAFF-R.
  • BAFF-R also known as tumor necrosis factor receptor superfamily member 13C, is overexpressed in certain tumor cell types and autoimmune diseases.
  • VAY736 has both anti-inflammatory and antineoplastic activities. In cancer cells, BAFF-R plays a key role in B-cell proliferation and survival.
  • VAY736 targets and binds to BAFF-R, which inhibits both BAFF/BAFF-R interaction and BAFF-R-mediated signaling. This may decrease cell growth in tumor cells expressing BAFF-R.
  • VAY736 is disclosed, e.g., in U.S. Pat. No. 8,106,163.
  • the heavy chain CDR1 of VAY736 has the amino acid sequence disclosed as SEQ ID NO: 3 in U.S. Pat. No. 8,106,163.
  • the heavy chain CDR2 of VAY736 has the amino acid sequence disclosed as SEQ ID NO: 10 in U.S. Pat. No. 8,106,163.
  • the heavy chain CDR3 of VAY736 has the amino acid sequence disclosed as SEQ ID NO: 17 in U.S. Pat. No. 8,106,163.
  • the light chain CDR1 of VAY736 has the amino acid sequence disclosed as SEQ ID NO: 24 in U.S. Pat. No. 8,106,163.
  • the light chain CDR2 of VAY736 has the amino acid sequence disclosed as SEQ ID NO: 31 in U.S. Pat. No. 8,106,163.
  • the light chain CDR3 of VAY736 has the amino acid sequence disclosed as SEQ ID NO: 38 in U.S. Pat. No. 8,106,163.
  • the heavy chain variable region of VAY736 has the amino acid sequence disclosed as SEQ ID NO: 52 in U.S. Pat. No. 8,106,163.
  • the light chain variable region of VAY736 has the amino acid sequence disclosed as SEQ ID NO: 45 in U.S. Pat. No. 8,106,163.
  • the heavy chain of VAY736 has the amino acid sequence disclosed as SEQ ID NO: 75 in U.S. Pat. No. 8,106,163.
  • the light chain variable region of VAY736 has the amino acid sequence disclosed as SEQ ID NO: 71 in U.S. Pat. No. 8,106,163.
  • a combination described herein includes an inhibitor of MALT-1 and/or BTK. In some embodiments, the combination is used to treat a cancer, e.g., a cancer described herein.
  • Exemplary BTK inhibitors include, but are not limited to, ibrutinib (PCI-32765); GDC-0834; RN-486; CGI-560; CGI-1764; HM-71224; CC-292; ONO-4059; CNX-774; or LFM-A13.
  • the BTK inhibitor does not reduce or inhibit the kinase activity of interleukin-2-inducible kinase (ITK), e.g., is selected from GDC-0834; RN-486; CGI-560; CGI-1764; HM-71224; CC-292; ONO-4059; CNX-774; or LFM-A13.
  • ITK interleukin-2-inducible kinase
  • the kinase inhibitor is a BTK inhibitor, e.g., ibrutinib (PCI-32765).
  • BTK inhibitor e.g., ibrutinib (PCI-32765).
  • PCI-32765 BTK inhibitor
  • the structure of ibrutinib (1-[(3R)-3-[4-Amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl]piperidin-1-yl]prop-2-en-1-one) is shown below.
  • the BTK inhibitor is a BTK inhibitor described in International Application WO/2015/079417, which is herein incorporated by reference in its entirety.
  • the BTK inhibitor is a compound of formula (I) or a pharmaceutically acceptable salt thereof;
  • R1 is hydrogen, C1-C6 alkyl optionally substituted by hydroxy
  • R2 is hydrogen or halogen
  • R3 is hydrogen or halogen
  • R4 is hydrogen
  • R5 is hydrogen or halogen
  • R4 and R5 are attached to each other and stand for a bond, —CH2-, —CH2-CH2-, —CH ⁇ CH—, —CH ⁇ CH—CH2-; —CH2-CH ⁇ CH—; or —CH2-CH2-CH2-;
  • R6 and R7 stand independently from each other for H, C1-C6 alkyl optionally substituted by hydroxyl, C3-C6 cycloalkyl optionally substituted by halogen or hydroxy, or halogen;
  • R8, R9, R, R′, R10 and R11 independently from each other stand for H, or C1-C6 alkyl optionally substituted by C1-C6 alkoxy; or any two of R8, R9, R, R′, R10 and R11 together with the carbon atom to which they are bound may form a 3-6 membered saturated carbocyclic ring;
  • R12 is hydrogen or C1-C6 alkyl optionally substituted by halogen or C1-C6 alkoxy;
  • R12 and any one of R8, R9, R, R′, R10 or R11 together with the atoms to which they are bound may form a 4, 5, 6 or 7 membered azacyclic ring, which ring may optionally be substituted by halogen, cyano, hydroxyl, C1-C6 alkyl or C1-C6 alkoxy;
  • n 0 or 1
  • R13 is C2-C6 alkenyl optionally substituted by C1-C6 alkyl, C1-C6 alkoxy or N,N-di-C1-C6 alkyl amino; C2-C6 alkynyl optionally substituted by C1-C6 alkyl or C1-C6 alkoxy; or C2-C6 alkylenyl oxide optionally substituted by C1-C6 alkyl.
  • the BTK inhibitor of Formula I is chosen from: N-(3-(5-((1-Acryloylazetidin-3-yl)oxy)-6-aminopyrimidin-4-yl)-5-fluoro-2-methylphenyl)-4-cyclopropyl-2-fluorobenzamide; (E)-N-(3-(6-Amino-5-((1-(but-2-enoyl)azetidin-3-yl)oxy)pyrimidin-4-yl)-5-fluoro-2-methylphenyl)-4-cyclopropyl-2-fluorobenzamide; N-(3-(6-Amino-5-((1-propioloylazetidin-3-yl)oxy)pyrimidin-4-yl)-5-fluoro-2-methylphenyl)-4-cyclopropyl-2-fluorobenzamide; N-(3-(6-Amino-5-((1-(but-2-ynoyl)
  • a combination described herein includes an inhibitor of Janus kinase (JAK).
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., a colon cancer, a prostate cancer, a lung cancer, a breast cancer, or a pancreatic cancer), e.g., a hematologic malignancy (e.g., a leukemia (e.g., a myeloid leukemia or a lymphocytic leukemia), e.g., a lymphoma (e.g., a non-Hodgkin lymphoma), or e.g., a multiple myeloma.
  • a cancer e.g., a cancer described herein, e.g., a solid tumor (e.g., a colon cancer, a prostate cancer, a lung cancer, a breast cancer, or a pancreatic cancer), e
  • the JAK inhibitor is 2-fluoro-N-methyl-4-(7-(quinolin-6-ylmethyl)imidazo[1,2-b][1,2,4]triazin-2-yl)benzamide (Compound A17), or a dihydrochloric salt thereof, or a compound disclosed in PCT Publication No. WO 2007/070514.
  • the JAK inhibitor e.g., 2-fluoro-N-methyl-4-(7-(quinolin-6-ylmethyl)imidazo[1,2-b][1,2,4]triazin-2-yl)benzamide (Compound A17), or a dihydrochloric salt thereof, or a compound disclosed in PCT Publication No. WO 2007/070514, is administered at a dose of about 400-600 mg (e.g., per day), e.g., about 400, 500, or 600 mg, or about 400-500 or 500-600 mg.
  • the JAK inhibitor is ruxolitinib phosphate (also known as JAKAFI; Compound A18) or a compound disclosed in PCT Publication No. WO 2007/070514.
  • the JAK inhibitor e.g., ruxolitinib phosphate (also known as JAKAFI; Compound A18) or a compound disclosed in PCT Publication No. WO 2007/070514
  • ruxolitinib phosphate also known as JAKAFI; Compound A18
  • the dose is about 15, 20, or 25 mg, or about 15-20 or 20-25 mg.
  • a combination described herein includes an inhibitor of chemoattractant receptor homologous to the T helper 2 cell (CRTH2).
  • the combination is used to treat a cancer, e.g., a cancer described herein.
  • the CRTH2 inhibitor is QAV680 (CAS Registry Number: 872365-16-7).
  • QAV680 is also known as fevipiprant and 2-[2-methyl-1-[(4-methylsulfonylphenyl)methyl]pyrrolo[2,3-b]pyridin-3-yl]acetic acid.
  • QAV680 is disclosed, e.g., in Sandham et al. Bioorg Med Chem. 2013; 21(21):6582-91.
  • the CRTH2 inhibitor is QAW039 (CAS Number: 872365-14-5).
  • QAW039 is also known as [1-(4-Methanesulfonyl-2-trifluoromethyl-benzyl)-2-methyl-1H-pyrrolo[2,3-b]pyridin-3-yl]-acetic acid.
  • QAW039 is disclosed, e.g. in Sykes et al. European Respiratory Journal Sep. 1, 2014 vol. 44 no. Suppl 58 P4074.
  • CRTH2 inhibitors that can be used in the combination include, e.g., AZD1981, ARRY-502, setipiprant (ACT-453859), and ACT-129968.
  • a combination described herein includes an inhibitor of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3).
  • PFKFB3 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3
  • the combination is used to treat a cancer, e.g., a cancer described herein, e.g., a solid tumor (e.g., an advanced solid tumor).
  • the PFKFB3 inhibitor is PFK-158.
  • PFK-158 is also known as ACT-PFK-158 or (E)-1-(pyridyn-4-yl)-3-(7-(trifluoromethyl)quinolin-2-yl)-prop-2-en-1-one.
  • PFK-158 is a derivative of 3-(3-pyridinyl)-1-[4-pyridinyl]-2-propen-1-one (3P0).
  • PFKFB3 which catalyzes the conversion of fructose-6-phosphate to fructose-2,6-bisphosphate, is highly expressed and active in human cancer cells and plays a key role in increasing both glycolytic flux in and proliferation of cancer cells.
  • PFKFB3 inhibitors e.g., PFK-158
  • PFK-158 can bind to and inhibit the activity of PFKFB3, which leads to the inhibition of both the glycolytic pathway in and glucose uptake by cancer cells. This prevents the production of macromolecules and energy that causes the enhanced cellular proliferation in cancer cells as compared to that of normal, healthy cells. Depriving cancer cells of nutrients and energy leads to the inhibition of cancer cell growth.
  • PFK158 is disclosed, e.g., at page 5 of WO 2013/148228.
  • the PFKFB3 inhibitor has the following structure:
  • the invention features a composition (e.g., one or more compositions or dosage forms), that includes a combination of three or more therapeutic agents chosen from one, two or all of the following categories (i)-(iii): (i) an agent that enhances antigen (e.g., tumor antigen) presentation; (ii) an agent that enhances an effector cell response (e.g., activation and/or mobilization of B cell and/or T cell); or (iii) an agent that decreases tumor immunosuppression.
  • the combination includes a a-GITR modulator (e.g., an anti-GITR antibody molecule as described herein).
  • the invention features a composition (e.g., one or more compositions or dosage forms as described hereom), for use in treating a disorder, e.g., a cancer.
  • the composition for use includes a combination of two, three or more therapeutic agents chosen from one, two or all of the following categories (i)-(iii): (i) an agent that enhances antigen (e.g., tumor antigen) presentation; (ii) an agent that enhances an effector cell response (e.g., activation and/or mobilization of B cell and/or T cell); or (iii) an agent that decreases tumor immunosuppression.
  • the combination used includes a a-GITR modulator (e.g., an anti-GITR antibody molecule as described herein).
  • the cancer can be, e.g., a cancer described herein, such as lung cancer (squamous), lung cancer (adenocarcinoma), head and neck cancer, cervical cancer (squamous), stomach cancer, thyroid cancer, melanoma, nasopharyngeal cancer, or breast cancer.
  • the combinations of therapeutic agents disclosed herein include two or more therapeutic agents described herein.
  • the therapeutic agents in the combination can belong to the same category, e.g., two or more therapeutic agents of category (i), or can include at least one agent of two or more categories (e.g., a therapeutic agent of category (i) combined with a therapeutic agent of category (ii)), as described below.
  • Certain therapeutic agents can belong to two or more categories of categories (i)-(iii).
  • a therapeutic agent e.g., a GITR agonist, an IDO antagonist, a TGF-b inhibitor, among others
  • Formulations e.g., dosage formulations, and kits, e.g., therapeutic kits, that include a combination of two, three or more therapeutic agents chosen from one, two or all of the following categories (i)-(iii): (i) an agent that enhances antigen (e.g., tumor antigen) presentation; (ii) an agent that enhances an effector cell response (e.g., activation and/or mobilization of B cell and/or T cell); or (iii) an agent that decreases tumor immunosuppression, thereby reducing an activity in the cell, and (optionally) instructions for use, are also disclosed.
  • the combination includes a a-GITR modulator (e.g., an anti-GITR antibody molecule as described herein).
  • the combinations disclosed herein can be administered together in a single composition or administered separately in two or more different compositions, e.g., compositions or dosage forms as described herein.
  • the administration of the therapeutic agents can be in any order.
  • the first agent and the additional agents e.g., second, third agents
  • a first therapeutic agent can be administered concurrently with, prior to, or subsequent to, the additional agent.
  • a first agent is administered locally, e.g., a therapeutic agent of any of categories (i)-(iii) can be coupled to a tumor targeting agent, e.g., a tumor-targeting antibody (e.g., to form an antibody-drug conjugate), or any other delivery agent (e.g., a formulation such as a targeted formulation) such that administration of the first agent is localized to a desired site, e.g., a tumor site (e.g., a dendritic cell-enriched site).
  • a tumor targeting agent e.g., a tumor-targeting antibody (e.g., to form an antibody-drug conjugate)
  • any other delivery agent e.g., a formulation such as a targeted formulation
  • the therapeutic agent is an antigen (e.g., a vaccine, e.g., an in situ cancer vaccine), which is targeted to the tumor environment, thus resulting in activation of dendritic cells.
  • a vaccine e.g., an in situ cancer vaccine
  • the therapeutic agent also can be locally administered, e.g., injected, at a tumor site (e.g., intratumoral or peritumoral administration). Localized delivery or administration of the therapeutic agent can reduce one or more side effects or toxicities that would otherwise be associated with systemic administration of the therapeutic agent.
  • a therapeutic agent e.g., STING or a TLR
  • a tumor-binding antibody e.g., an antibody that binds a tumor associated antigen (TAA)
  • TAA tumor associated antigen
  • the combination includes a therapeutic agent from the antigen-presentation combination (e.g., one or more of a STING agonist, a TLR agonist, a vaccine or an oncolytic virus) in combination with a an anti-GITR antibody molecule as described herein and a therapeutic agent from the effector cell and/or anti-tumor immunosuppression combination (e.g., an inhibitor of a checkpoint inhibitor, e.g., an inhibitor of PD-1, PD-L1, LAG-3, TIM-3, CEACAM (e.g., CEACAM-1, -3 and/or -5), or CTLA-4, or any combination thereof for therapy.
  • a therapeutic agent from the antigen-presentation combination e.g., one or more of a STING agonist, a TLR agonist, a vaccine or an oncolytic virus
  • a therapeutic agent from the effector cell and/or anti-tumor immunosuppression combination e.g., an inhibitor of a checkpoint inhibitor, e.g
  • one or more of a STING agonist, a TLR agonist, a vaccine or an oncolytic virus is administered in combination with an anti-GITR antibody molecule as described herein.
  • a STING agonist and/or a vaccine is administered in combination with an anti-GITR antibody molecule as described herein.
  • an oncolytic virus is administered in combination with an anti-GITR antibody molecule as described herein.
  • the first agent, the additional agent (e.g., second or third agent), or all can be administered in an amount or dose that is higher, lower or the same than the amount or dosage of each agent used individually, e.g., as a monotherapy.
  • the administered amount or dosage of the first agent, the additional agent (e.g., second or third agent), or all is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50%) than the amount or dosage of each agent used individually, e.g., as a monotherapy.
  • the amount or dosage of the first agent, the additional agent (e.g., second or third agent), or all, that results in a desired effect is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50% lower).
  • Cancers subject to treatment include without limitation epithelial cancers or carcinomas, as well as sarcomas and lymphomas.
  • the cancer is melanoma, ovarian cancer, renal cancer, colorectal cancer, prostate, lung cancer including non-small cell lung cancer (NSCLC), breast cancer, glioma, or fibrosarcoma.
  • NSCLC non-small cell lung cancer
  • the type of cancer is selected from the group consisting of: pancreatic cancer, melanomas, breast cancer, lung cancer, bronchial cancer, colorectal cancer, prostate cancer, stomach cancer, ovarian cancer, urinary bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, cervical cancer, uterine or endometrial cancer, cancer of the oral cavity or pharynx, liver cancer, kidney cancer, testicular cancer, biliary tract cancer, small bowel or appendix cancer, salivary gland cancer, thyroid gland cancer, adrenal gland cancer, osteosarcoma, chondrosarcoma, cancer of hematological tissues and head and neck squamous cell carcinoma (HNSCC).
  • pancreatic cancer melanomas
  • breast cancer breast cancer
  • lung cancer bronchial cancer
  • colorectal cancer prostate cancer
  • stomach cancer ovarian cancer
  • urinary bladder cancer brain or central nervous system cancer
  • peripheral nervous system cancer esophageal cancer
  • cervical cancer uterine or
  • One or more combinations can be used to treat a cancer as described herein, such as pancreatic cancer, melanomas, breast cancer, lung cancer, bronchial cancer, colorectal cancer, prostate cancer, stomach cancer, ovarian cancer, urinary bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, cervical cancer, uterine or endometrial cancer, cancer of the oral cavity or pharynx, liver cancer, kidney cancer, testicular cancer, biliary tract cancer, small bowel or appendix cancer, salivary gland cancer, thyroid gland cancer, adrenal gland cancer, osteosarcoma, chondrosarcoma, cancer of hematological tissues and head and neck squamous cell carcinoma (HNSCC).
  • Methods and compositions disclosed herein are useful for treating metastatic lesions associated with the aforementioned cancers.
  • the invention provides a method of enhancing an immune response to an antigen in a subject, comprising administering to the subject: (i) the antigen; and (ii) a combination as described herein, e.g., a combination comprising a therapeutically effective amount of an anti-GITR antibody molecule described herein, such that an immune response to the antigen in the subject is enhanced.
  • the antigen can be, for example, a tumor antigen, a viral antigen, a bacterial antigen or an antigen from a pathogen.
  • the combinations as described herein can be administered to the subject systemically (e.g., orally, parenterally, subcutaneously, intravenously, rectally, intramuscularly, intraperitoneally, intranasally, transdermally, or by inhalation or intracavitary installation), topically, or by application to mucous membranes, such as the nose, throat and bronchial tubes.
  • the anti-GITR antibody molecule is administered by injection (e.g., subcutaneously or intravenously) at a dose of about 1 to 30 mg/kg, e.g., about 5 to 25 mg/kg, about 10 to 20 mg/kg, about 1 to 5 mg/kg, or about 3 mg/kg.
  • the dosing schedule can vary from e.g., once a week to once every 2, 3, or 4 weeks.
  • the anti-GITR antibody molecule is administered at a dose from about 10 to 20 mg/kg every other week.
  • FIG. 1A-1D illustrate epitope mapping of the GITR mAbs of the invention.
  • FIG. 1A depicts results of hydrogen/deuterium exchange coupled to mass spectrometry (HDXMS) analyses using Fc-GITR fusion (top and middle trace) and HIS-GITR (lower trace) fusion proteins and MAB1 parental Ab. Numbering reflects removal of native GITR signal peptide (AA 1-26) sequence.
  • FIG. 1B depicts a schematic of N-terminal deletion constructs prepared using the extracellular domain of human GITR (hGITR ECD).
  • FIG. 1C depicts results of binding of MAB4 and MAB5 to hGITR ECD constructs.
  • FIG. 1D depicts results of alanine scanning mutagenesis.
  • MAB7 bound to all mutant proteins with the exception of GITR mutant E78A. ForteBioTM binding analysis was carried out, and results also confirmed loss of MAB7 binding to hGITRE78A mutant protein (data not shown).
  • Results implicates a region of ECD of GITR including CRD1 and including E78 (SEQ ID NO:88: RPTGGPGCGPGRLLGTGTDARCCRVHTTRCCRDYPGEECCSEWDCMCVQPEFHCGD) as a region and potential epitope involved in binding MAB1 and MAB7 (parental mAb).
  • FIG. 2A-2E depicts results of binding experiments of anti-GITR MAB antibodies.
  • FIGS. 2A and 2B illustrates MAB4, and MAB5 specifically bind to GITR from human and cynomolgus monkeys (2A) but not from rodent (2B), as determined by ELISA assays.
  • FIG. 2C illustrates MAB7 shares a similar profile binding human and cyno GITR but not murine GITR by ELISA assay.
  • FIG. 2D illustrates MAB7 competes with GITR-ligand binding as determined by FACS competition analysis.
  • 2E illustrates results of ELISA assays showing that the anti-GITR antibodies of the invention (e.g., MAB4, MAB5) do not bind to other members of the TNF receptor superfamily (TNFRSF).
  • ProtagenTM chip assays also confirmed that the antibodies do not bind to other off-target proteins (not shown).
  • FIG. 3A-3D depict intracellular signaling in 293 cells that have been engineered to express GITR.
  • FIG. 3A illustrates that recombinant human GITR ligand (GITR-L) activates intracellular signaling in 293 cells that have been stably transfected to overexpress human GITR.
  • FIG. 3B illustrates that monoclonal antibodies MAB4 and MAB5 activate intracellular signaling in 293 cells that have been transfected to overexpress human GITR comparably to GITR-L when the antibodies are cross-linked (EC 50 for GITRL is about 65 nM versus EC 50 of about 2.5 nM for agonist antibodies in the presence of cross-linker).
  • FIG. 1A illustrates that recombinant human GITR ligand (GITR-L) activates intracellular signaling in 293 cells that have been stably transfected to overexpress human GITR.
  • FIG. 3B illustrates that monoclonal antibodies MAB4 and MAB5 activate
  • FIG. 3C illustrates that cross-linked MAB antibody activates intracellular signaling in cells, as MAB7 and MAB8 also promote NF ⁇ B activation in 293 cells stably transfected with human GITR and the NFkB-Luciferase reporter gene in a similar manner to cross-linked MAB4.
  • FIG. 3D illustrates that Cross-linked MAB4 and MAB5 promote NF ⁇ B activation in 293 cells that have been stably transfected with cyno GITR and the NF ⁇ B-Luciferase reporter gene. Similar activation was seen with cross-linked MAB7 (data not shown).
  • FIG. 4A-4C depicts in vitro co-stimulatory activity of MAB7 on T cells is dependent upon T cell activation.
  • Anti-CD3 (OKT3), anti-CD28 (CD28.2) and anti-GITR mAbs were cross-linked (at a ratio of 1:1:3) on beads and then incubated with PBMCs.
  • FIG. 4A illustrates MAB7 is a co-activator of CD4+ T cells and stimulates T cell proliferation in CD4+ Tcells.
  • FIG. 4B illustrates MAB7 is a co-activator of CD8+ T cells and stimulates T cell proliferation in CD8+ Tcells.
  • FIG. 4C illustrates cytokine production, e.g. IFN ⁇ production following TCR engagement is increased in conjunction with MAB7. Similar results were seen for MAB4 and 5 (data not shown).
  • FIG. 5A-D illustrates in vitro ADCC activity of MAB7 in GITR expressing cells at varying levels.
  • FIG. 5A , FIG. 5B , FIG. 5C , and FIG. 5D each depict results of ADCC activity using control or MAB7 antibody with various levels of GITR expression.
  • MAB7 is able to induce signaling through the FcgRllla, with increased activity upon increased levels of GITR signaling.
  • FIG. 6A-6D illustrates GITR is functional in hGITR-hGITRL knock-in mice.
  • Splenocytes were isolated from hGITR-hGITRL knock-in mice and cultured either unstimulated or stimulated with aCD3 and aCD8 antibodies for 48 hours, then pulsed with controls or MAB7 at varying concentrations for 30 minutes, then fixed and stained with fluorophore-conjugated antibodies and analyzed by flow cytometry.
  • FIG. 6A depicts results showing expression of hGITR is upregulated on stimulated CD8+ T cells.
  • FIG. 6B depicts results of anti hGITR antibody binding to T cells by hFc staining, showing MAB7 can bind to hGITR expressed on mouse CD8+ T cells.
  • FIGS. 6C and 6D depict MAB7 binding to stimulated CD8+ T cells correlates with increased T cell activation, as shown by intracellular pIKK staining ( 6 C) and T cell activation ( 6 D). *p ⁇ 0.05, **p ⁇ 0.005.
  • FIG. 7A-7C illustrates MAB7 is functional in vivo.
  • FIG. 7A depicts results of tumor measurements twice per week and tumor volume calculated using the equation (L ⁇ W 2 )/2. Data shown is from the fifteen (15)-day time point group.
  • FIGS. 7B and 7C depict results from whole blood and FIGS. 7C and 7D depict results from tumors were collected 3-days post-dose and analyzed by flow cytometry for cell surface hGITR expression on immune cells. (*p ⁇ 0.05, ****p ⁇ 0.00005).
  • FIG. 8A-8E illustrates MAB7 elicits an anti-tumor immune response to Colon26 tumors in vivo.
  • FIG. 8A depicts results of Tregulatory cells 3-days post-dose.
  • FIG. 8B-8C depict results of lymphocytes (8B) and activated CD8+ T cells (8C) present in tumor site following treatment levels in tumor 15-days post-dose. The absolute number of cells was normalized to tumor size to account for the significant difference in tumor size between Vehicle and MAB7 treated groups.
  • FIG. 8D depicts Teff/Treg ratio resulting in treated animals as determined by total intratumoral activated CD8+ T cells compared to CD4+ FOXP3+ Tregs to generate T eff /T reg ratios.
  • FIG. 8E depicts results of splenocyte assays from purified CD8+ T cells incubated with Colon26 tumor cells ex-vivo, and measuring CTL response using IFNg ELISPOT assay. (*p ⁇ 0.05, ***p ⁇ 0.0005).
  • FIG. 9A-9C illustrates PD-1 expression is upregulated on CD8+ T cells in Colon26 tumors as well as spleens after treatment with a murine surrogate GITR antibody, DTA-1. Single cell suspensions of whole tumors or whole spleens were profiled by flow cytometry following 2 doses of DTA-1.
  • FIG. 9A depicts results of PD-1 positive cells assessed as a percentage of total CD19-CD3+CD8+ T cells.
  • FIG. 9B depicts results of PD-1 positive cells normalized to tumor size by absolute number of PD-1+CD19-CD3+CD8+ T cells per mm 3 volume of tumor.
  • FIG. 9A-9C illustrates PD-1 expression is upregulated on CD8+ T cells in Colon26 tumors as well as spleens after treatment with a murine surrogate GITR antibody, DTA-1. Single cell suspensions of whole tumors or whole spleens were profiled by flow cytometry following 2 doses of DTA-1
  • 9C depicts results of PD-1 expression is upregulated on CD8+ T cells in spleens of Colon26 tumor bearing mice after treatment with DTA-1. PD-1 positive cells were assessed as a percentage of total CD19-CD3+CD8+ T cells. (*p ⁇ 0.05 and ****p ⁇ 0.0005).
  • FIG. 10 illustrates anti-GITR and anti-PD-1 combinations confer survival advantage compared to isotype control. Depicted are results in Colon26 mice models treated with anti-GITR (IgG2a-DTA-1) and anti-PD-1 (RMP1-14) individually or in combination as compared to isotype control.
  • FIG. 11 illustrates expression of LAG3(first column), TIM3(middle column), and PD1(right column) after treatment with anti-GITR, anti-PD-1 and anti-GITR/anti-PD-1 in combinations in mice with established Colon26 tumors as compared to treatment with isotype control Ab. Depicted are results in Colon26 mice models treated with anti-GITR (IgG2a-DTA-1) and anti-PD-1 (RMP1-14) individually or in combination as compared to isotype control. The top row demonstrates results in tumor samples, and the lower row depicts results in spleen samples.
  • LAG3, TIM3 and PD1 expression is upregulated on CD8+ T cells in Colon26 tumors after treatment with a-GITR
  • a-PD1 PD-1 expression is upregulated on CD8+ T cells in Colon26 tumors after treatment with anti-GITR/anti-PD-1 in combination.
  • MAB2, MAB3, MAB4, MAB5, MAB6, MAB7 and MAB8 were generated by engineering a murine monoclonal GITR agonist antibody MAB1 to have greater sequence homology to a human germline antibody.
  • MAB2, MAB3, MAB4, MAB5, MAB6, MAB7 and MAB8 retain the epitope specificity, affinity, and cynomolgus macaque GITR cross-reactivity of the parental murine antibody, MAB1.
  • MAB2, MAB3, MAB4, MAB5, MAB6, MAB7 and MAB8 have much higher homology to the human germline sequence than the original murine antibody and should therefore be better tolerated by the human immune system.
  • Mouse monoclonal MAB1 was engineered to bring its protein sequence closer to a human germline sequence and decrease its immunogenicity using the Humaneered® technology platform available through KaloBios, (South San Francisco, Calif. (on the worldwide web at kalobios.com)).
  • Humaneered® antibodies are very close to human antibodies with V-region sequences that have high homology to a human germline sequence while still retaining the specificity and affinity of the parent or reference antibody (U.S. Patent Publ. 2005/0255552 and 2006/0134098).
  • the process first identifies the minimum antigen binding specificity determinants (BSDs) in heavy and light chain variable regions of a reference Fab (typically sequences within the heavy chain CDR3 and the light chain CDR3). As these heavy and light chain BSDs are maintained in all libraries constructed during the process, each library is epitope-focused, and the resulting Humaneered® antibodies retain the epitope specificity of the original mouse antibody.
  • BSDs minimum antigen binding specificity determinants
  • full chain libraries in which an entire light or heavy chain variable region is replaced with a library of human sequences
  • cassette libraries in which a portion of the heavy or light chain variable region of the mouse Fab is replaced with a library of human sequences
  • a bacterial secretion system is used to express members of the library as antibody Fab fragments, and the library is screened for Fabs that bind antigen using a colony lift binding assay (CLBA). Positive clones are further characterized to identify those with the highest affinity.
  • Identified human cassettes supporting binding in the context of residual murine sequences are the combined in a final library screen to generate completely human V-regions.
  • Humaneered® antibody Fabs have V-segment sequences derived from human libraries, retain the short BSD sequences identified within the CDR3 regions, and have human germline Framework 4 regions. These Fabs are converted to full IgGs by cloning variable regions of the heavy and light chains into IgG expression vectors. Humaneered® antibodies generated in this process retain the binding specificity of the parent, murine antibody, typically having equivalent or higher affinity for antigen that the parent antibody, and have V-regions with a high degree of sequence identity compared with human germline antibody genes at the protein level.
  • Bcl-2 transgenic mice C57BL/6-Tgn (bcl-2) 22 wehi strain
  • bcl-2 transgenic mice C57BL/6-Tgn (bcl-2) 22 wehi strain
  • RIMMS Repetitive Immunization at Multiple Sites
  • a hybridoma secreting MAB1 was identified and selected using a sandwich ELISA against hGITR and an NF ⁇ B Reporter Gene Assay to confirm hGITR binding and agonist activity.
  • Variable region DNA from murine monoclonal MAB1 was amplified by RT-PCR from RNA obtained from the hybridoma cell line using standard methods. Heavy chain variable region was amplified from MAB1 cDNA with HV3 (5′-GGGTCTAGACACCATGGCTGTCTTGGGGCTGCTCTTC-3′ (SEQ ID NO:95)) and HCconstant (5′-GCGTCTAGAAYCTCCACACACAGGRRCCAGTGGATAGAC-3′ (SEQ ID NO:96)).
  • Light chain variable region was amplified from the same cDNA with LV3 (5′-GGGTCTAGACACCATGGAGWCACAKWCTCAGGTCTTTRTA-3′ (SEQ ID NO:97)) and LCconstant (5′-GCGTCTAGAACTGGATGGTGGGAAGATGG-3′ (SEQ ID NO:19)). Variable heavy and light chain products were inserted into a pcDNA3.1 vector and sequence verified.
  • the heavy and light vectors were used as templates for PCR incorporating restriction enzyme sites for cloning into KaloBios vectors: Vh into KB1292-His (modified version of KB1292 that encodes a C-terminal flexible linker and 6-His tag (SEQ ID NO:11) of amino acid sequence AAGASHHHHHH (SEQ ID NO:13) on CH1) at NcoI (5′) and Nhel (3′); Vk into KB1296 at NcoI (5′) and BsiWI (3′).
  • These separate heavy and light chain vectors were then combined into a single dicistronic KaloBios Fab expression vector by restriction digest with BssHII and ClaI and ligation. Fab fragments were expressed in E. coli from this vector. This Fab was tested for hGITR-antigen binding and is referred to as MAB1rFab.
  • Fab fragments were expressed by secretion from E. coli using KaloBios expression vectors. Cells were grown in 2 ⁇ YT medium to an OD500 of ⁇ 0.6. Expression was induced by adding IPTG to 100 ⁇ M and shaking for 4 hours at 33° C. Assembled Fab was obtained from periplasmic fractions by osmotic lysis and purification by affinity chromatography using Ni-NTA columns HisTrap HP columns; GE Healthcare catalog #17-5247-01) according to standard methods. Fabs were eluted in buffer containing 500 mM imidazole and thoroughly dialyzed against PBS pH7.4 without calcium and magnesium.
  • cassettes Two types were constructed by overlap PCR: front-end cassettes (8C1VK3FE-01, and MAB1VH3FE-01) containing human sequences in FR1, CDR1, and FR2, and FR3 cassettes (MAB1VK3FR3-01, and MAB1VH3FR3-01) containing human sequences in the FR3 were amplified using the above mentioned germline restricted KaloBios libraries.
  • each Vh cassette library was cloned into vector KB1292-His at NcoI (5′) and KpnI (3′); each Vk cassette library was cloned into vector KB1296-B (modified version of KaloBios vector KB1296 which has a silent HindIII site added in FR4) at NcoI (5′) and HindIII (3′).
  • Vh or Vk plasmid libraries were then combined with the complementary chain from the optimized reference Fab (MAB1opVK or MAB1 opVH (e.g., the Vh front-end library was combined with the optimized reference Vk vector) by digestion with BssHII and ClaI and subsequent ligation to create libraries of dicistronic vectors expressing full Fabs.
  • MAB1opVK or MAB1 opVH e.g., the Vh front-end library was combined with the optimized reference Vk vector
  • VH3 front-end library (MAB1VH3FE-02) was constructed.
  • This library contains human sequences in FR1, FR2, and a collection of CDR2s encoding either the parental murine residue or the selected human germline residue at all positions.
  • the FR3 region sequences of this library were from six clones selected from the VH3FR3 library (MAB1VH3FR3-01).
  • Vk full-chain library (MAB1VK3FCL-01) was constructed by combining clones from VK front-end and VKFR3 cassette libraries with mutagenic VK CDR2s that encodes either the parental murine or the selected human germline residue at all positions.
  • the resulting Vk full-chain library was cloned into KB1296b at NcoI and HindIII sites.
  • This VK full-chain library was paired with a number of selected VH3FR3 library clones to allow functional Fab expression and screened by CLBS.
  • the antigen specific clones were confirmed by human GITR specific ELISA and ranked by antigen affinity titration ELISA.
  • VH3 full-chain library (MAB1VH3FcL-01) was generated using the selected clones from the second VH3 front end library (MAB1VH3FE-02) with a collection of CDR2 sequences containing either the parental murine or human residue at each position.
  • This VH full-chain library was cloned into KB1292-his at NcoI and KpnI sites.
  • selected VK full-chain clones were combined with VH full-chain library at BssHII and ClaI sites.
  • hGITR-hFc Recombinant human GITR and human Fc fusion protein
  • hGITR-hFc antigen diluted in PBS pH 7.4 was bound to a 96-well microtiter plate at 200 ng/well by overnight incubation at 4° C. After being rinsed three times with PBST, the plate was blocked with a solution of 1% BSA in PBS for one hour at 37° C., and then rinsed once with PBST.
  • Fab-containing cell medium or diluted, purified Fab (50 ⁇ L) was then added to each well.
  • the plate was rinsed three times with PBST.
  • Anti-human-kappa chain HRP conjugate (Sigma #A7164) diluted 1:5000 in PBST (50 ⁇ L) was added to each well, and the plate was incubated for 45 min at room temperature.
  • the plate was washed three times with PBST, then 100 ⁇ L of SureBlue TMB substrate (KPL #52-00-03) was added to each well and the plate was incubated for about 10 min at room temperature.
  • the plate was read at 650 nm in a spectrophotometer.
  • an affinity titration ELISA was developed. This assay combines two consecutive ELISA steps: the first one, using goat anti-human Fab (Jackson ImmunoResearch Lab #109-005-097) capture and goat anti-human Kappa (Sigma #A7164) detection, measures Fab concentrations in cell culture medium to normalize the amount of Fab used in the second antigen titration ELISA; the second ELISA, a normal antigen specific ELISA, generates an antigen binding dilution curve with the same amount of starting Fab. By comparing the dilution curves of different clones the high affinity clones are identified.
US15/517,872 2014-10-08 2015-10-08 Compositions and methods of use for augmented immune response and cancer therapy Abandoned US20170306038A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/517,872 US20170306038A1 (en) 2014-10-08 2015-10-08 Compositions and methods of use for augmented immune response and cancer therapy

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201462061644P 2014-10-08 2014-10-08
US201562198673P 2015-07-29 2015-07-29
US201562220764P 2015-09-18 2015-09-18
US15/517,872 US20170306038A1 (en) 2014-10-08 2015-10-08 Compositions and methods of use for augmented immune response and cancer therapy
PCT/US2015/054770 WO2016057841A1 (en) 2014-10-08 2015-10-08 Compositions and methods of use for augmented immune response and cancer therapy

Publications (1)

Publication Number Publication Date
US20170306038A1 true US20170306038A1 (en) 2017-10-26

Family

ID=54347879

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/517,872 Abandoned US20170306038A1 (en) 2014-10-08 2015-10-08 Compositions and methods of use for augmented immune response and cancer therapy
US15/517,867 Active 2036-04-16 US10662247B2 (en) 2014-10-08 2015-10-08 Compositions and methods of use for augmented immune response and cancer therapy
US16/845,030 Abandoned US20210015860A1 (en) 2014-10-08 2020-04-09 Compositions and methods of use for augmented immune response and cancer therapy

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/517,867 Active 2036-04-16 US10662247B2 (en) 2014-10-08 2015-10-08 Compositions and methods of use for augmented immune response and cancer therapy
US16/845,030 Abandoned US20210015860A1 (en) 2014-10-08 2020-04-09 Compositions and methods of use for augmented immune response and cancer therapy

Country Status (22)

Country Link
US (3) US20170306038A1 (es)
EP (2) EP3204421A1 (es)
JP (1) JP6731403B2 (es)
KR (1) KR20170065029A (es)
CN (1) CN107001476B (es)
AU (2) AU2015330771B2 (es)
BR (1) BR112017007093A2 (es)
CA (1) CA2964146A1 (es)
CL (2) CL2017000836A1 (es)
CO (1) CO2017003408A2 (es)
CR (1) CR20170138A (es)
CU (1) CU24437B1 (es)
EA (1) EA036467B1 (es)
EC (1) ECSP17027870A (es)
IL (1) IL251435A0 (es)
MA (1) MA41044A (es)
MX (1) MX2017004669A (es)
PE (1) PE20171654A1 (es)
PH (1) PH12017500634A1 (es)
SG (1) SG11201702596YA (es)
TN (1) TN2017000120A1 (es)
WO (2) WO2016057841A1 (es)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10675358B2 (en) 2016-07-07 2020-06-09 The Board Of Trustees Of The Leland Stanford Junior University Antibody adjuvant conjugates
CN111467474A (zh) * 2020-04-07 2020-07-31 江苏大学 一种具有免疫调节和抗肿瘤作用的药物
WO2020197578A1 (en) * 2019-03-27 2020-10-01 Immunophotonics, Inc. Semi-synthetic biopolymers for use in stimulating the immune system
CN111918878A (zh) * 2018-04-20 2020-11-10 信达生物制药(苏州)有限公司 抗gitr抗体及其用途
US11364304B2 (en) 2016-08-25 2022-06-21 Northwestern University Crosslinked micellar spherical nucleic acids
US11400164B2 (en) 2019-03-15 2022-08-02 Bolt Biotherapeutics, Inc. Immunoconjugates targeting HER2
US11696954B2 (en) 2017-04-28 2023-07-11 Exicure Operating Company Synthesis of spherical nucleic acids using lipophilic moieties
US11773188B2 (en) 2012-01-20 2023-10-03 Immunophotonics, Inc Chitosan-derived compositions
US11773180B2 (en) * 2017-11-08 2023-10-03 Kyowa Kirin Co., Ltd. Bispecific antibody which binds to CD40 and EpCAM

Families Citing this family (93)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101566538B1 (ko) 2012-06-08 2015-11-05 국립암센터 신규한 Th17 세포 전환용 에피토프 및 이의 용도
CR20160319A (es) 2013-12-12 2016-11-08 Jiangsu Hengrui Medicine Co Anticuerpo pd-1, fragmento de union al antigeno de este y uso médico de este
CN106132439A (zh) 2014-03-31 2016-11-16 豪夫迈·罗氏有限公司 包含抗血管发生剂和ox40结合激动剂的组合疗法
NZ726513A (en) 2014-05-28 2023-07-28 Memorial Sloan Kettering Cancer Center Anti-gitr antibodies and methods of use thereof
MA41044A (fr) 2014-10-08 2017-08-15 Novartis Ag Compositions et procédés d'utilisation pour une réponse immunitaire accrue et traitement contre le cancer
MA41460A (fr) 2015-02-03 2017-12-12 Oncomed Pharm Inc Agents de liaison à la tnfrsf et leurs utilisations
CA2992298A1 (en) * 2015-07-23 2017-01-26 Inhibrx Lp Multivalent and multispecific gitr-binding fusion proteins
JP7034066B2 (ja) 2015-10-02 2022-03-11 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト 共刺激tnf受容体に対する二重特異性抗体
CA3007233A1 (en) 2015-12-02 2017-06-08 Agenus Inc. Antibodies and methods of use thereof
JP2019503349A (ja) 2015-12-17 2019-02-07 ノバルティス アーゲー Pd−1に対する抗体分子およびその使用
DK3458478T3 (da) 2016-05-18 2021-03-22 Boehringer Ingelheim Int Anti-pd-1- og anti-lag3-antistoffer til cancerbehandling
KR102495601B1 (ko) 2016-06-10 2023-02-06 리제너론 파마슈티칼스 인코포레이티드 항-gitr 항체 및 그것의 사용
AU2017297506A1 (en) 2016-07-14 2019-02-21 Bristol-Myers Squibb Company Antibodies against TIM3 and uses thereof
RU2019104896A (ru) * 2016-07-22 2020-08-24 Дана-Фарбер Кэнсер Инститьют, Инк. Антитела к глюкокортикоид-индуцированному рецептору фактора некроза опухоли (gitr) и способы их использования
CN109963871A (zh) 2016-08-05 2019-07-02 豪夫迈·罗氏有限公司 具有激动活性的多价及多表位抗体以及使用方法
MA46770A (fr) 2016-11-09 2019-09-18 Agenus Inc Anticorps anti-ox40, anticorps anti-gitr, et leurs procédés d'utilisation
GB201619652D0 (en) * 2016-11-21 2017-01-04 Alligator Bioscience Ab Novel polypeptides
CN110475787A (zh) * 2017-01-27 2019-11-19 超人壹有限公司 结合剂
WO2018146549A1 (en) 2017-02-10 2018-08-16 Eutilex Co., Ltd. IFN-γ-INDUCIBLE REGULATORY T CELL CONVERTIBLE ANTI-CANCER (IRTCA) ANTIBODY AND USES THEREOF
CN110431152A (zh) * 2017-03-03 2019-11-08 雷纳神经科学公司 抗gitr抗体及其使用方法
WO2018185618A1 (en) 2017-04-03 2018-10-11 Novartis Ag Anti-cdh6 antibody drug conjugates and anti-gitr antibody combinations and methods of treatment
AR111651A1 (es) 2017-04-28 2019-08-07 Novartis Ag Conjugados de anticuerpos que comprenden agonistas del receptor de tipo toll y terapias de combinación
UY37695A (es) 2017-04-28 2018-11-30 Novartis Ag Compuesto dinucleótido cíclico bis 2’-5’-rr-(3’f-a)(3’f-a) y usos del mismo
WO2018213297A1 (en) 2017-05-16 2018-11-22 Bristol-Myers Squibb Company Treatment of cancer with anti-gitr agonist antibodies
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
US20200172628A1 (en) 2017-06-22 2020-06-04 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2018237157A1 (en) 2017-06-22 2018-12-27 Novartis Ag CD73 BINDING ANTIBODY MOLECULES AND USES THEREOF
WO2018234879A1 (en) 2017-06-22 2018-12-27 Novartis Ag USE OF IL-1β BINDING ANTIBODIES IN THE TREATMENT OF CANCER
WO2018235056A1 (en) 2017-06-22 2018-12-27 Novartis Ag IL-1BETA BINDING ANTIBODIES FOR USE IN THE TREATMENT OF CANCER
CA3066747A1 (en) 2017-06-27 2019-01-03 Novartis Ag Dosage regimens for anti-tim-3 antibodies and uses thereof
CN111163798A (zh) 2017-07-20 2020-05-15 诺华股份有限公司 用于抗lag-3抗体的给药方案及其用途
WO2019018640A1 (en) * 2017-07-21 2019-01-24 Novartis Ag POSOLOGICAL REGIMES FOR ANTI-GITREN ANTIBODIES AND USES THEREOF
CA3075969A1 (en) * 2017-10-10 2019-04-18 Numab Therapeutics AG Multispecific antibody
EP3476390A1 (en) * 2017-10-24 2019-05-01 Dompé farmaceutici S.p.A. Il-8 inhibitors for use in the treatment of sarcomas
US20210040205A1 (en) 2017-10-25 2021-02-11 Novartis Ag Antibodies targeting cd32b and methods of use thereof
CA3081602A1 (en) 2017-11-16 2019-05-23 Novartis Ag Combination therapies
WO2019140229A1 (en) 2018-01-12 2019-07-18 Bristol-Myers Squibb Company Antibodies against tim3 and uses thereof
US20210085785A1 (en) * 2018-02-23 2021-03-25 Truebinding, Inc. Treating cancer by blocking the interaction of vista and its binding partner
AU2019239747A1 (en) 2018-03-21 2020-10-08 Bristol-Myers Squibb Company Antibodies binding to VISTA at acidic pH
WO2019191133A1 (en) 2018-03-27 2019-10-03 Bristol-Myers Squibb Company Real-time monitoring of protein concentration using ultraviolet signal
US20210147547A1 (en) 2018-04-13 2021-05-20 Novartis Ag Dosage Regimens For Anti-Pd-L1 Antibodies And Uses Thereof
KR102558418B1 (ko) * 2018-04-15 2023-07-24 임비라 컴퍼니 리미티드 Pd-1에 결합하는 항체 및 그 용도
US20210206857A1 (en) * 2018-05-21 2021-07-08 Bioprocessia Technologies Llc Multivalent protein complexes
AR126019A1 (es) 2018-05-30 2023-09-06 Novartis Ag Anticuerpos frente a entpd2, terapias de combinación y métodos de uso de los anticuerpos y las terapias de combinación
WO2019229699A1 (en) 2018-05-31 2019-12-05 Novartis Ag Hepatitis b antibodies
WO2019232244A2 (en) 2018-05-31 2019-12-05 Novartis Ag Antibody molecules to cd73 and uses thereof
CA3103629A1 (en) 2018-06-15 2019-12-19 Flagship Pioneering Innovations V, Inc. Increasing immune activity through modulation of postcellular signaling factors
AU2019302454A1 (en) 2018-07-09 2021-02-25 Bristol-Myers Squibb Company Antibodies binding to ILT4
AR116109A1 (es) 2018-07-10 2021-03-31 Novartis Ag Derivados de 3-(5-amino-1-oxoisoindolin-2-il)piperidina-2,6-diona y usos de los mismos
SG11202011872QA (en) 2018-07-10 2021-01-28 Novartis Ag 3-(5-hydroxy-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of ikaros family zinc finger 2 (ikzf2)-dependent diseases
WO2020014327A2 (en) 2018-07-11 2020-01-16 Five Prime Therapeutics, Inc. Antibodies binding to vista at acidic ph
WO2020021465A1 (en) 2018-07-25 2020-01-30 Advanced Accelerator Applications (Italy) S.R.L. Method of treatment of neuroendocrine tumors
WO2020053742A2 (en) 2018-09-10 2020-03-19 Novartis Ag Anti-hla-hbv peptide antibodies
EP3873532A1 (en) 2018-10-31 2021-09-08 Novartis AG Dc-sign antibody drug conjugates
CN113645992A (zh) * 2018-11-15 2021-11-12 转矩医疗股份有限公司 用于癌症免疫疗法的方法和组合物
JP2022514315A (ja) 2018-12-20 2022-02-10 ノバルティス アーゲー 3-(1-オキソイソインドリン-2-イル)ピペリジン-2,6-ジオン誘導体を含む投与計画及び薬剤組み合わせ
WO2020128637A1 (en) 2018-12-21 2020-06-25 Novartis Ag Use of il-1 binding antibodies in the treatment of a msi-h cancer
WO2020128620A1 (en) 2018-12-21 2020-06-25 Novartis Ag Use of il-1beta binding antibodies
EP3897613A1 (en) 2018-12-21 2021-10-27 Novartis AG Use of il-1beta binding antibodies
AU2019406840A1 (en) 2018-12-21 2021-06-03 Novartis Ag Use of IL-1 beta antibodies in the treatment or prevention of myelodysplastic syndrome
KR20210129672A (ko) 2019-02-15 2021-10-28 노파르티스 아게 치환된 3-(1-옥소이소인돌린-2-일)피페리딘-2,6-디온 유도체 및 이의 용도
CA3124935A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020172658A1 (en) 2019-02-24 2020-08-27 Bristol-Myers Squibb Company Methods of isolating a protein
CN111773377B (zh) * 2019-04-04 2023-03-14 中南大学 阿尼芬净在制备抗肿瘤药物中的应用及抗肿瘤药物
EP3962493A2 (en) 2019-05-03 2022-03-09 Flagship Pioneering Innovations V, Inc. Methods of modulating immune activity/level of irf or sting or of treating cancer, comprising the administration of a sting modulator and/or purinergic receptor modulator or postcellular signaling factor
CN114245869A (zh) 2019-05-23 2022-03-25 百时美施贵宝公司 监测细胞培养基的方法
EP4031578A1 (en) 2019-09-18 2022-07-27 Novartis AG Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
EP4031575A1 (en) 2019-09-19 2022-07-27 Bristol-Myers Squibb Company Antibodies binding to vista at acidic ph
BR112022007179A2 (pt) 2019-10-21 2022-08-23 Novartis Ag Inibidores de tim-3 e usos dos mesmos
KR20220103947A (ko) 2019-10-21 2022-07-25 노파르티스 아게 베네토클락스 및 tim-3 억제제를 사용한 조합 요법
JP2023509359A (ja) 2019-12-17 2023-03-08 フラグシップ パイオニアリング イノベーションズ ブイ,インコーポレーテッド 鉄依存性細胞分解の誘導物質との併用抗癌療法
CN115052662A (zh) 2019-12-20 2022-09-13 诺华股份有限公司 抗TGFβ抗体和检查点抑制剂用于治疗增殖性疾病的用途
MX2022008763A (es) 2020-01-17 2022-07-27 Novartis Ag Combinacion que comprende un inhibidor de tim-3 y un agente hipometilante para usarse en el tratamiento del sindrome mielodisplasico o leucemia mielomonocitica cronica.
MX2022010910A (es) 2020-03-06 2022-10-07 Regeneron Pharma Anticuerpos anti-gitr y usos de estos.
JP2023529211A (ja) 2020-06-11 2023-07-07 ノバルティス アーゲー Zbtb32阻害剤及びその使用
JP2023531676A (ja) 2020-06-23 2023-07-25 ノバルティス アーゲー 3-(1-オキソイソインドリン-2-イル)ピぺリジン-2,6-ジオン誘導体を含む投与レジメン
CN116096906A (zh) 2020-06-29 2023-05-09 旗舰创业创新五公司 工程化以促进萨诺传递的病毒及其在治疗癌症中的用途
CN116134027A (zh) 2020-08-03 2023-05-16 诺华股份有限公司 杂芳基取代的3-(1-氧代异吲哚啉-2-基)哌啶-2,6-二酮衍生物及其用途
EP4204020A1 (en) 2020-08-31 2023-07-05 Advanced Accelerator Applications International S.A. Method of treating psma-expressing cancers
WO2022043557A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
US20230374064A1 (en) 2020-10-05 2023-11-23 Bristol-Myers Squibb Company Methods for concentrating proteins
WO2022117572A2 (en) 2020-12-02 2022-06-09 Oncurious Nv An ltbr agonist in combination therapy against cancer
CA3214085A1 (en) 2021-03-31 2022-10-06 Darby Rye Schmidt Thanotransmission polypeptides and their use in treating cancer
TW202304979A (zh) 2021-04-07 2023-02-01 瑞士商諾華公司 抗TGFβ抗體及其他治療劑用於治療增殖性疾病之用途
AR125874A1 (es) 2021-05-18 2023-08-23 Novartis Ag Terapias de combinación
KR20240017912A (ko) * 2021-06-04 2024-02-08 암젠 인크 항-ccr8 항체 및 이의 용도
KR20240026507A (ko) 2021-06-29 2024-02-28 플래그쉽 파이어니어링 이노베이션스 브이, 인크. 타노트랜스미션을 촉진시키도록 엔지니어링된 면역 세포 및 이의 용도
WO2023173011A1 (en) 2022-03-09 2023-09-14 Bristol-Myers Squibb Company Transient expression of therapeutic proteins
WO2023170207A1 (en) * 2022-03-09 2023-09-14 Alderaan Biotechnology Anti-cd160 transmembrane isoform antibodies
WO2023175614A1 (en) 2022-03-15 2023-09-21 Yeda Research And Development Co. Ltd. Anti glucocorticoid-induced tnfr-related (gitr) protein antibodies and uses thereof
WO2024077191A1 (en) 2022-10-05 2024-04-11 Flagship Pioneering Innovations V, Inc. Nucleic acid molecules encoding trif and additionalpolypeptides and their use in treating cancer
CN115969971B (zh) * 2022-11-24 2023-08-18 珠海恩科生物科技有限公司 组合物在制备治疗肿瘤的药物中的应用
CN115819603A (zh) * 2022-12-26 2023-03-21 广西大学 一种猪IKKα多克隆抗体的制备方法和应用

Family Cites Families (276)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
DE3572982D1 (en) 1984-03-06 1989-10-19 Takeda Chemical Industries Ltd Chemically modified lymphokine and production thereof
US4978672A (en) 1986-03-07 1990-12-18 Ciba-Geigy Corporation Alpha-heterocyclc substituted tolunitriles
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US4881175A (en) 1986-09-02 1989-11-14 Genex Corporation Computer based system and method for determining and displaying possible chemical structures for converting double- or multiple-chain polypeptides to single-chain polypeptides
US5260203A (en) 1986-09-02 1993-11-09 Enzon, Inc. Single polypeptide chain binding molecules
WO1988007089A1 (en) 1987-03-18 1988-09-22 Medical Research Council Altered antibodies
US5013653A (en) 1987-03-20 1991-05-07 Creative Biomolecules, Inc. Product and process for introduction of a hinge region into a fusion protein to facilitate cleavage
US5091513A (en) 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5132405A (en) 1987-05-21 1992-07-21 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
JPH02500329A (ja) 1987-05-21 1990-02-08 クリエイテイブ・バイオマリキユールズ・インコーポレーテツド ターゲット化多機能蛋白質
ATE95193T1 (de) 1987-06-17 1993-10-15 Sandoz Ag Cyclosporine und deren benutzung als arzneimittel.
US5677425A (en) 1987-09-04 1997-10-14 Celltech Therapeutics Limited Recombinant antibody
US5336603A (en) 1987-10-02 1994-08-09 Genentech, Inc. CD4 adheson variants
KR900005995A (ko) 1988-10-31 1990-05-07 우메모또 요시마사 변형 인터류킨-2 및 그의 제조방법
EP0401384B1 (en) 1988-12-22 1996-03-13 Kirin-Amgen, Inc. Chemically modified granulocyte colony stimulating factor
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5112946A (en) 1989-07-06 1992-05-12 Repligen Corporation Modified pf4 compositions and methods of use
FR2650598B1 (fr) 1989-08-03 1994-06-03 Rhone Poulenc Sante Derives de l'albumine a fonction therapeutique
WO1991006570A1 (en) 1989-10-25 1991-05-16 The University Of Melbourne HYBRID Fc RECEPTOR MOLECULES
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5349053A (en) 1990-06-01 1994-09-20 Protein Design Labs, Inc. Chimeric ligand/immunoglobulin molecules and their uses
DE69123241T2 (de) 1990-12-14 1997-04-17 Cell Genesys Inc Chimärische ketten zur transduktion von rezeptorverbundenen signalwegen
US20030206899A1 (en) 1991-03-29 2003-11-06 Genentech, Inc. Vascular endothelial cell growth factor antagonists
US6582959B2 (en) 1991-03-29 2003-06-24 Genentech, Inc. Antibodies to vascular endothelial cell growth factor
WO1994004679A1 (en) 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
US5622929A (en) 1992-01-23 1997-04-22 Bristol-Myers Squibb Company Thioether conjugates
FR2686899B1 (fr) 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa Nouveaux polypeptides biologiquement actifs, leur preparation et compositions pharmaceutiques les contenant.
FR2686901A1 (fr) 1992-01-31 1993-08-06 Rhone Poulenc Rorer Sa Nouveaux polypeptides antithrombotiques, leur preparation et compositions pharmaceutiques les contenant.
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
CA2076465C (en) 1992-03-25 2002-11-26 Ravi V. J. Chari Cell binding agent conjugates of analogues and derivatives of cc-1065
US5447851B1 (en) 1992-04-02 1999-07-06 Univ Texas System Board Of Dna encoding a chimeric polypeptide comprising the extracellular domain of tnf receptor fused to igg vectors and host cells
WO1993022332A2 (en) 1992-04-24 1993-11-11 Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
DK1087013T3 (da) 1992-08-21 2009-05-11 Univ Bruxelles Immunoglobuliner uden lette kæder
ATE348110T1 (de) 1992-10-28 2007-01-15 Genentech Inc Hvegf rezeptor als vegf antagonist
EP0714409A1 (en) 1993-06-16 1996-06-05 Celltech Therapeutics Limited Antibodies
SE9400088D0 (sv) 1994-01-14 1994-01-14 Kabi Pharmacia Ab Bacterial receptor structures
US5605793A (en) 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US5837458A (en) 1994-02-17 1998-11-17 Maxygen, Inc. Methods and compositions for cellular and metabolic engineering
US5834252A (en) 1995-04-18 1998-11-10 Glaxo Group Limited End-complementary polymerase reaction
CA2196200A1 (en) 1994-07-29 1996-02-15 Michael Joseph Browne Novel compounds
WO2000053753A2 (en) 1999-03-08 2000-09-14 Genentech, Inc. Promotion or inhibition of angiogenesis and cardiovascularization
WO2000073452A2 (en) 1999-06-02 2000-12-07 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
IL117645A (en) 1995-03-30 2005-08-31 Genentech Inc Vascular endothelial cell growth factor antagonists for use as medicaments in the treatment of age-related macular degeneration
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
US5811097A (en) 1995-07-25 1998-09-22 The Regents Of The University Of California Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
WO1997034631A1 (en) 1996-03-18 1997-09-25 Board Of Regents, The University Of Texas System Immunoglobin-like domains with increased half lives
TW533205B (en) 1996-06-25 2003-05-21 Novartis Ag Substituted 3,5-diphenyl-l,2,4-triazoles and their pharmaceutical composition
US6111090A (en) 1996-08-16 2000-08-29 Schering Corporation Mammalian cell surface antigens; related reagents
WO1998006842A1 (en) 1996-08-16 1998-02-19 Schering Corporation Mammalian cell surface antigens; related reagents
WO1998023289A1 (en) 1996-11-27 1998-06-04 The General Hospital Corporation MODULATION OF IgG BINDING TO FcRn
US6277375B1 (en) 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
US20020032315A1 (en) 1997-08-06 2002-03-14 Manuel Baca Anti-vegf antibodies
DE69829891T2 (de) 1997-04-07 2005-10-06 Genentech, Inc., South San Francisco Anti-VEGF Antikörper
US6884879B1 (en) 1997-04-07 2005-04-26 Genentech, Inc. Anti-VEGF antibodies
PT1787999E (pt) 1997-04-07 2010-11-11 Genentech Inc Anticorpos anti-vegf
EP1724282B1 (en) 1997-05-21 2013-05-15 Merck Patent GmbH Method for the production of non-immunogenic proteins
US6165476A (en) 1997-07-10 2000-12-26 Beth Israel Deaconess Medical Center Fusion proteins with an immunoglobulin hinge region linker
CO4940418A1 (es) 1997-07-18 2000-07-24 Novartis Ag Modificacion de cristal de un derivado de n-fenil-2- pirimidinamina, procesos para su fabricacion y su uso
DE19742706B4 (de) 1997-09-26 2013-07-25 Pieris Proteolab Ag Lipocalinmuteine
US6503184B1 (en) 1997-10-21 2003-01-07 Human Genome Sciences, Inc. Human tumor necrosis factor receptor-like proteins TR11, TR11SV1 and TR11SV2
WO1999020758A1 (en) 1997-10-21 1999-04-29 Human Genome Sciences, Inc. Human tumor necrosis factor receptor-like proteins tr11, tr11sv1, and tr11sv2
US6689607B2 (en) 1997-10-21 2004-02-10 Human Genome Sciences, Inc. Human tumor, necrosis factor receptor-like proteins TR11, TR11SV1 and TR11SV2
JP2002502607A (ja) 1998-02-09 2002-01-29 ジェネンテク・インコーポレイテッド 新規な腫瘍壊死因子レセプター相同体及びそれをコードする核酸
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
EP1070085A1 (en) 1998-04-08 2001-01-24 G.D. SEARLE & CO. DUAL A v?b 3? AND METASTASIS-ASSOCIATED RECEPTOR LIGANDS
PT1071700E (pt) 1998-04-20 2010-04-23 Glycart Biotechnology Ag Modificação por glicosilação de anticorpos para melhorar a citotoxicidade celular dependente de anticorpos
AU5124699A (en) 1998-07-22 2000-02-14 Incyte Pharmaceuticals, Inc. Molecules associated with cell proliferation
WO2000009143A1 (en) 1998-08-13 2000-02-24 G.D. Searle & Co. Multivalent avb3 and metastasis-associated receptor ligands
AU3107000A (en) 1998-12-01 2000-06-19 Genentech Inc. Methods and compositions for inhibiting neoplastic cell growth
US6818418B1 (en) 1998-12-10 2004-11-16 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
ES2694002T3 (es) 1999-01-15 2018-12-17 Genentech, Inc. Polipéptido que comprende una región Fc de IgG1 humana variante
KR20010103046A (ko) 1999-03-08 2001-11-17 제넨테크, 인크. 면역 관련 질환 치료용 조성물 및 치료 방법
JP2003531811A (ja) 1999-03-08 2003-10-28 ジェネンテック・インコーポレーテッド 血管形成及び心臓血管新生の促進又は阻害
PT1914244E (pt) 1999-04-09 2013-07-26 Kyowa Hakko Kirin Co Ltd Processo para regular a actividade de moléculas funcionais sob o ponto de vista imunológico
US6703020B1 (en) 1999-04-28 2004-03-09 Board Of Regents, The University Of Texas System Antibody conjugate methods for selectively inhibiting VEGF
CA2378179A1 (en) 1999-07-12 2001-01-18 Genentech, Inc. Promotion or inhibition of angiogenesis and cardiovascularization by tumor necrosis factor ligand/receptor homologs
DE19932688B4 (de) 1999-07-13 2009-10-08 Scil Proteins Gmbh Design von Beta-Faltblatt-Proteinen des gamma-II-kristallins antikörperähnlichen
GB0018891D0 (en) 2000-08-01 2000-09-20 Novartis Ag Organic compounds
EP1252192B1 (en) 2000-02-11 2006-08-16 MERCK PATENT GmbH Enhancing the circulating half-life of antibody-based fusion proteins
CA2405709A1 (en) 2000-04-12 2001-10-25 Human Genome Sciences, Inc. Albumin fusion proteins
CA2441903C (en) 2000-05-26 2012-07-31 National Research Council Of Canada Single-domain brain-targeting antibody fragments derived from llama antibodies
WO2002008293A2 (en) 2000-07-25 2002-01-31 Immunomedics Inc. Multivalent target binding protein
GB0020685D0 (en) 2000-08-22 2000-10-11 Novartis Ag Organic compounds
EP2351838A1 (en) 2000-10-20 2011-08-03 Chugai Seiyaku Kabushiki Kaisha Crosslinking agonistic antibodies
US6995162B2 (en) 2001-01-12 2006-02-07 Amgen Inc. Substituted alkylamine derivatives and methods of use
US20050048512A1 (en) 2001-04-26 2005-03-03 Avidia Research Institute Combinatorial libraries of monomer domains
US20040175756A1 (en) 2001-04-26 2004-09-09 Avidia Research Institute Methods for using combinatorial libraries of monomer domains
US20050053973A1 (en) 2001-04-26 2005-03-10 Avidia Research Institute Novel proteins with targeted binding
WO2003006058A1 (en) 2001-07-12 2003-01-23 Wyeth Cd25+ differential markers and uses thereof
EP1443961B1 (en) 2001-10-25 2009-05-06 Genentech, Inc. Glycoprotein compositions
PT1441737E (pt) 2001-10-30 2006-12-29 Dana Farber Cancer Inst Inc Derivados de estrutura como inibidores da actividade do receptor de tirosina cinase flt3
CN100522967C (zh) 2002-02-01 2009-08-05 阿里亚德基因治疗公司 含磷化合物及其应用
PT2275102E (pt) 2002-03-13 2015-10-27 Array Biopharma Inc Derivados de benzimidazole alquilado n3 como inibidores de mek
US7495090B2 (en) 2002-05-23 2009-02-24 The Regents Of The University Of California Nucleic acids encoding chimeric CD154 polypeptides
GB0215676D0 (en) 2002-07-05 2002-08-14 Novartis Ag Organic compounds
AU2003291002A1 (en) 2002-11-15 2004-06-15 Chiron Corporation Methods for preventing and treating cancer metastasis and bone loss associated with cancer metastasis
WO2004058191A2 (en) 2002-12-20 2004-07-15 Biogen Idec Ma Inc. Multivalent lymphotoxin beta receptor agonists and therapeutic uses thereof
CN101899114A (zh) 2002-12-23 2010-12-01 惠氏公司 抗pd-1抗体及其用途
EP1578419A4 (en) 2002-12-30 2008-11-12 3M Innovative Properties Co IMMUNOSTIMULATING COMBINATIONS
CA2515726C (en) 2003-02-11 2012-07-10 Vernalis (Cambridge) Limited Isoxazole compounds
WO2004072266A2 (en) 2003-02-13 2004-08-26 Kalobios Inc. Antibody affinity engineering by serial epitope-guided complementarity replacement
EP1462114A1 (en) 2003-03-28 2004-09-29 Universiteit Utrecht Holding B.V. Methods and means to suppress symptons of an allergic disease by inhibiting the glucocorticoid-induced tumor necrosis factor receptor (GRIT or TNFRSF18)
CA2525717A1 (en) * 2003-05-23 2004-12-09 Wyeth Gitr ligand and gitr ligand-related molecules and antibodies and uses thereof
ZA200509143B (en) * 2003-05-23 2007-03-28 Wyeth Corp GITR ligand and GITR ligand-related molecules and antibodies and uses thereof
DE10324447A1 (de) 2003-05-28 2004-12-30 Scil Proteins Gmbh Generierung künstlicher Bindungsproteine auf der Grundlage von Ubiquitin
US20100069614A1 (en) 2008-06-27 2010-03-18 Merus B.V. Antibody producing non-human mammals
ME00425B (me) 2003-05-30 2011-10-10 Genentech Inc Liječenje sa anti-vegf antitijelima
EP1660126A1 (en) 2003-07-11 2006-05-31 Schering Corporation Agonists or antagonists of the clucocorticoid-induced tumour necrosis factor receptor (gitr) or its ligand for the treatment of immune disorders, infections and cancer
US20050106667A1 (en) 2003-08-01 2005-05-19 Genentech, Inc Binding polypeptides with restricted diversity sequences
WO2005044853A2 (en) 2003-11-01 2005-05-19 Genentech, Inc. Anti-vegf antibodies
US20050069521A1 (en) 2003-08-28 2005-03-31 Emd Lexigen Research Center Corp. Enhancing the circulating half-life of interleukin-2 proteins
US20070142401A1 (en) 2003-10-27 2007-06-21 Novartis Ag Indolyl-pyrroledione derivatives for the treatment of neurological and vascular disorders related to beta-amyloid generation and/or aggregation
WO2005055808A2 (en) 2003-12-02 2005-06-23 Genzyme Corporation Compositions and methods to diagnose and treat lung cancer
PT2311873T (pt) 2004-01-07 2018-11-20 Novartis Vaccines & Diagnostics Inc Anticorpo monoclonal específico para m-csf e respetivos usos
CA2553692C (en) 2004-01-20 2014-10-07 Kalobios, Inc. Antibody specificity transfer using minimal essential binding determinants
WO2005073224A2 (en) 2004-01-23 2005-08-11 Amgen Inc Quinoline quinazoline pyridine and pyrimidine counds and their use in the treatment of inflammation angiogenesis and cancer
CA2560742A1 (en) 2004-03-23 2005-10-06 Biogen Idec Ma Inc. Receptor coupling agents and therapeutic uses thereof
GB0409799D0 (en) 2004-04-30 2004-06-09 Isis Innovation Method of generating improved immune response
US7932260B2 (en) 2004-05-13 2011-04-26 Icos Corporation Quinazolinones as inhibitors of human phosphatidylinositol 3-kinase delta
WO2006083289A2 (en) 2004-06-04 2006-08-10 Duke University Methods and compositions for enhancement of immunity by in vivo depletion of immunosuppressive cell activity
US20060008844A1 (en) 2004-06-17 2006-01-12 Avidia Research Institute c-Met kinase binding proteins
GB0512324D0 (en) 2005-06-16 2005-07-27 Novartis Ag Organic compounds
US20060009360A1 (en) 2004-06-25 2006-01-12 Robert Pifer New adjuvant composition
GB0417487D0 (en) 2004-08-05 2004-09-08 Novartis Ag Organic compound
US20060099171A1 (en) 2004-11-05 2006-05-11 Masahide Tone Mouse glucocorticoid-induced TNF receptor ligand is costimulatory for T cells
WO2006055778A2 (en) 2004-11-16 2006-05-26 Kalobios, Inc. Immunoglobulin variable region cassette exchange
WO2006074399A2 (en) 2005-01-05 2006-07-13 Biogen Idec Ma Inc. Multispecific binding molecules comprising connecting peptides
ES2711213T3 (es) 2005-02-08 2019-04-30 Genzyme Corp Anticuerpos de TGFbeta
ES2432091T5 (es) 2005-03-25 2022-03-18 Gitr Inc Moléculas de unión GITR y usos de las mismas
EP1885396A2 (en) 2005-05-04 2008-02-13 Quark Pharmaceuticals, Inc. Recombinant antibodies against cd55 and cd59 and uses thereof
SI1877090T1 (sl) 2005-05-06 2014-12-31 Providence Health System Trimerni fuzijski protein OY40-imunoglobulina in postopki uporabe
CA2970873C (en) 2005-05-09 2022-05-17 E. R. Squibb & Sons, L.L.C. Human monoclonal antibodies to programmed death 1 (pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
GB0510390D0 (en) 2005-05-20 2005-06-29 Novartis Ag Organic compounds
WO2006132272A1 (ja) 2005-06-07 2006-12-14 The University Of Tokyo 抗体の作製方法
CN104356236B (zh) 2005-07-01 2020-07-03 E.R.施贵宝&圣斯有限责任公司 抗程序性死亡配体1(pd-l1)的人单克隆抗体
US8211648B2 (en) 2005-07-22 2012-07-03 Kalobios Pharmaceuticals, Inc. Secretion of antibodies without signal peptides from bacteria
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
GT200600381A (es) 2005-08-25 2007-03-28 Compuestos organicos
PE20070335A1 (es) 2005-08-30 2007-04-21 Novartis Ag Benzimidazoles sustituidos y metodos para su preparacion
WO2007044887A2 (en) 2005-10-11 2007-04-19 Transtarget, Inc. Method for producing a population of homogenous tetravalent bispecific antibodies
EP1777294A1 (en) 2005-10-20 2007-04-25 Institut National De La Sante Et De La Recherche Medicale (Inserm) IL-15Ralpha sushi domain as a selective and potent enhancer of IL-15 action through IL-15Rbeta/gamma, and hyperagonist (IL15Ralpha sushi -IL15) fusion proteins
AR056142A1 (es) 2005-10-21 2007-09-19 Amgen Inc Metodos para generar el anticuerpo igg monovalente
PL2251034T3 (pl) 2005-12-02 2018-07-31 Icahn School Of Medicine At Mount Sinai Chimeryczne wirusy prezentujące nienatywne białka powierzchniowe i zastosowania tych wirusów
PL2481753T3 (pl) 2005-12-13 2018-09-28 Eli Lilly And Company Przeciwciała anty-IL-17
TWI468162B (zh) 2005-12-13 2015-01-11 英塞特公司 作為傑納斯激酶(JANUS KINASE)抑制劑之經雜芳基取代之吡咯并〔2,3-b〕吡啶及吡咯并〔2,3-b〕嘧啶
PT2347775T (pt) 2005-12-13 2020-07-14 The President And Fellows Of Harvard College Estruturas em andaime para transplante celular
EP1981969A4 (en) 2006-01-19 2009-06-03 Genzyme Corp ANTI-GITRANT ANTIBODIES FOR THE TREATMENT OF CANCER
JO2660B1 (en) 2006-01-20 2012-06-17 نوفارتيس ايه جي Pi-3 inhibitors and methods of use
WO2007109254A2 (en) 2006-03-17 2007-09-27 Biogen Idec Ma Inc. Stabilized polypeptide compositions
ES2654847T3 (es) 2006-04-19 2018-02-15 Novartis Ag Compuestos de benzoxazol y benzotiazol sustituidos en 6-O y métodos para inhibir la señalización CSF-1R
UA93548C2 (uk) 2006-05-05 2011-02-25 Айерем Елелсі Сполуки та композиції як модулятори хеджхогівського сигнального шляху
MX363905B (es) 2006-06-12 2019-04-08 Aptevo Res & Development Llc Proteinas de union multivalentes monocatenarias con funcion efectora.
PE20110224A1 (es) 2006-08-02 2011-04-05 Novartis Ag PROCEDIMIENTO PARA LA SINTESIS DE UN PEPTIDOMIMETICO DE Smac INHIBIDOR DE IAP, Y COMPUESTOS INTERMEDIARIOS PARA LA SINTESIS DEL MISMO
WO2008073160A2 (en) 2006-08-17 2008-06-19 The Trustees Of Columbia University In The City Of New York Methods for converting or inducing protective immunity
ME00588A (en) 2006-08-18 2011-12-20 Prlr-specific antibody and uses thereof
US7833527B2 (en) 2006-10-02 2010-11-16 Amgen Inc. Methods of treating psoriasis using IL-17 Receptor A antibodies
GB0620894D0 (en) 2006-10-20 2006-11-29 Univ Southampton Human immune therapies using a CD27 agonist alone or in combination with other immune modulators
ES2689444T3 (es) 2006-11-22 2018-11-14 Incyte Holdings Corporation Imidazotriazinas e imidazopirimidinas como inhibidores de la quinasa
WO2008070743A2 (en) 2006-12-05 2008-06-12 Memgen Llc Methods of increasing cancer sensitivity to chemotherapeutic agents using chimeric isf35
EP2091918B1 (en) 2006-12-08 2014-08-27 Irm Llc Compounds and compositions as protein kinase inhibitors
WO2008140621A2 (en) 2006-12-21 2008-11-20 Mount Sinai School Of Medicine Of New York University Transgenic oncolytic viruses and uses thereof
JP2008278814A (ja) 2007-05-11 2008-11-20 Igaku Seibutsugaku Kenkyusho:Kk アゴニスティック抗ヒトgitr抗体による免疫制御の解除とその応用
EP3222634A1 (en) 2007-06-18 2017-09-27 Merck Sharp & Dohme B.V. Antibodies to human programmed death receptor pd-1
US9770535B2 (en) 2007-06-21 2017-09-26 President And Fellows Of Harvard College Scaffolds for cell collection or elimination
NZ599338A (en) 2007-06-27 2013-11-29 Marinepolymer Tech Inc Complexes of il-15 and il-15ralpha and uses thereof
ES2591281T3 (es) * 2007-07-12 2016-11-25 Gitr, Inc. Terapias de combinación que emplean moléculas de enlazamiento a GITR
US20090155275A1 (en) 2007-07-31 2009-06-18 Medimmune, Llc Multispecific epitope binding proteins and uses thereof
KR20100052545A (ko) 2007-08-28 2010-05-19 바이오겐 아이덱 엠에이 인코포레이티드 Igf―1r의 다중 에피토프에 결합하는 조성물
WO2009043051A2 (en) 2007-09-27 2009-04-02 Biogen Idec Ma Inc. Cd23 binding molecules and methods of use thereof
US9266967B2 (en) 2007-12-21 2016-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US8747847B2 (en) 2008-02-11 2014-06-10 Curetech Ltd. Monoclonal antibodies for tumor treatment
JP5690143B2 (ja) 2008-02-13 2015-03-25 プレジデント・アンド・フェロウズ・オブ・ハーバード・カレッジ 持続的細胞プログラミング装置
EP2262837A4 (en) 2008-03-12 2011-04-06 Merck Sharp & Dohme PD-1 BINDING PROTEINS
US8637542B2 (en) 2008-03-14 2014-01-28 Intellikine, Inc. Kinase inhibitors and methods of use
AR070924A1 (es) 2008-03-19 2010-05-12 Novartis Ag Formas cristalinas y dos formas solvatadas de sales del acido lactico de 4- amino -5- fluoro-3-(5-(4-metilpiperazin-1-il ) -1h- bencimidazol-2-il) quinolin -2-(1h) - ona
WO2009131239A1 (ja) 2008-04-25 2009-10-29 Kyowa Hakko Kirin Co Ltd 安定な多価抗体
SI2300455T1 (sl) 2008-05-21 2017-12-29 Incyte Holdings Corporation Soli 2-fluoro-n-metil-4-(7-(kinolin-6-il-metil)-imidazo(1,2-b)1,2,4) triazin-2-il)benzamid in postopki v zvezi z njihovo pripravo
EP2282995B1 (en) 2008-05-23 2015-08-26 Novartis AG Derivatives of quinolines and quinoxalines as protein tyrosine kinase inhibitors
UY31929A (es) 2008-06-25 2010-01-05 Irm Llc Compuestos y composiciones como inhibidores de cinasa
MX2011000039A (es) 2008-07-02 2011-05-31 Emergent Product Dev Seattle Proteinas antagonistas del factor-beta de crecimiento transformante (tgf-beta), que se unen a multiples objetivos.
BRPI0915231A2 (pt) 2008-07-08 2018-06-12 Intellikine Inc compostos inibidores de quinase e métodos de uso
JP5767109B2 (ja) 2008-07-17 2015-08-19 ノバルティス アーゲー 治療抗体を用いる組成物及び方法
US20100041663A1 (en) 2008-07-18 2010-02-18 Novartis Ag Organic Compounds as Smo Inhibitors
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
PL2331547T3 (pl) 2008-08-22 2015-01-30 Novartis Ag Związki pirolopirymidynowe jako inhibitory CDK
KR20110074850A (ko) 2008-08-25 2011-07-04 앰플리뮨, 인크. Pd-1 길항제 및 그의 사용 방법
WO2010027423A2 (en) 2008-08-25 2010-03-11 Amplimmune, Inc. Compositions of pd-1 antagonists and methods of use
CN103333157A (zh) 2008-09-02 2013-10-02 诺瓦提斯公司 作为激酶抑制剂的吡啶甲酰胺衍生物
WO2010028798A1 (en) 2008-09-10 2010-03-18 F. Hoffmann-La Roche Ag Multivalent antibodies
UA104147C2 (uk) 2008-09-10 2014-01-10 Новартис Аг Похідна піролідиндикарбонової кислоти та її застосування у лікуванні проліферативних захворювань
WO2010030002A1 (ja) 2008-09-12 2010-03-18 国立大学法人三重大学 外来性gitrリガンド発現細胞
US8703778B2 (en) 2008-09-26 2014-04-22 Intellikine Llc Heterocyclic kinase inhibitors
CN102227221A (zh) 2008-11-28 2011-10-26 诺瓦提斯公司 包含hsp90抑制剂和mtor抑制剂的药物组合
PE20120341A1 (es) 2008-12-09 2012-04-24 Genentech Inc Anticuerpos anti-pd-l1 y su uso para mejorar la funcion de celulas t
WO2010071880A1 (en) * 2008-12-19 2010-06-24 University Of Miami Tnfr25 agonists to enhance immune responses to vaccines
WO2010091262A1 (en) 2009-02-05 2010-08-12 Mount Sinai School Of Medicine Of New York University Chimeric newcastle disease viruses and uses thereof
WO2010089411A2 (en) 2009-02-09 2010-08-12 Universite De La Mediterranee Pd-1 antibodies and pd-l1 antibodies and uses thereof
UA103918C2 (en) 2009-03-02 2013-12-10 Айерем Элелси N-(hetero)aryl, 2-(hetero)aryl-substituted acetamides for use as wnt signaling modulators
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
US8703132B2 (en) 2009-06-18 2014-04-22 Hoffmann-La Roche, Inc. Bispecific, tetravalent antigen binding proteins
MY155570A (en) 2009-06-26 2015-10-30 Novartis Ag 1, 3-disubstituted imidazolidin-2-one derivatives as inhibitors of cyp 17
JO3002B1 (ar) 2009-08-28 2016-09-05 Irm Llc مركبات و تركيبات كمثبطات كيناز بروتين
US8709424B2 (en) 2009-09-03 2014-04-29 Merck Sharp & Dohme Corp. Anti-GITR antibodies
GB0919054D0 (en) 2009-10-30 2009-12-16 Isis Innovation Treatment of obesity
WO2011066342A2 (en) 2009-11-24 2011-06-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
JP5950824B2 (ja) 2009-12-07 2016-07-13 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー 抗腫瘍抗体療法を増強するための方法
US8440693B2 (en) 2009-12-22 2013-05-14 Novartis Ag Substituted isoquinolinones and quinazolinones
SI2519543T1 (sl) 2009-12-29 2016-08-31 Emergent Product Development Seattle, Llc Beljakovine, ki se vežejo s heterodimeri in njihova uporaba
UY33227A (es) 2010-02-19 2011-09-30 Novartis Ag Compuestos de pirrolopirimidina como inhibidores de la cdk4/6
IL300733A (en) 2010-03-05 2023-04-01 Univ Johns Hopkins Compositions and methods for antibodies and fusion proteins targeting immune modulation
KR101846590B1 (ko) 2010-06-11 2018-04-09 교와 핫꼬 기린 가부시키가이샤 항 tim-3 항체
AU2011275749C1 (en) 2010-07-09 2015-09-17 Aduro Biotech Holdings, Europe B.V. Agonistic antibody to CD27
US20120014947A1 (en) * 2010-07-16 2012-01-19 The University Of Chicago Methods and compositions to reduce liver damage associated with conditions or therapies that affect the immune system
EA036314B1 (ru) 2010-08-20 2020-10-26 Новартис Аг Выделенные антитела к рецептору эпидермального фактора роста-3 (her3) и их фрагменты, фармацевтическая композиция, содержащая эти антитела и фрагменты, и их применение для лечения рака
MY171312A (en) 2010-08-23 2019-10-08 Univ Texas Anti-ox40 antibodies and methods of using the same
WO2012025525A1 (en) 2010-08-24 2012-03-01 Roche Glycart Ag Activatable bispecific antibodies
CN103068846B9 (zh) 2010-08-24 2016-09-28 弗·哈夫曼-拉罗切有限公司 包含二硫键稳定性Fv片段的双特异性抗体
KR20140020228A (ko) 2010-09-21 2014-02-18 알토 바이오사이언스 코포레이션 다량체성 아이엘 15 용해성 융합 분자 및 그의 제조 및 사용 방법
WO2012048165A2 (en) 2010-10-06 2012-04-12 President And Fellows Of Harvard College Injectable, pore-forming hydrogels for materials-based cell therapies
US9409950B2 (en) 2010-12-23 2016-08-09 Biogen Ma Inc. Linker peptides and polypeptides comprising same
EP2701745B1 (en) 2011-04-28 2018-07-11 President and Fellows of Harvard College Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
US8846042B2 (en) 2011-05-16 2014-09-30 Fabion Pharmaceuticals, Inc. Multi-specific FAB fusion proteins and methods of use
KR102051014B1 (ko) 2011-06-03 2019-12-04 조마 테크놀로지 리미티드 Tgf-베타에 특이적인 항체
EP2714073B1 (en) 2011-06-03 2021-03-10 President and Fellows of Harvard College In situ antigen-generating cancer vaccine
WO2012178137A1 (en) 2011-06-24 2012-12-27 Gillies Stephen D Light chain immunoglobulin fusion proteins and methods of use thereof
WO2013039954A1 (en) 2011-09-14 2013-03-21 Sanofi Anti-gitr antibodies
AU2013201121A1 (en) 2011-09-20 2013-04-04 Vical Incorporated Synergistic anti-tumor efficacy using alloantigen combination immunotherapy
CA2849765C (en) 2011-09-26 2021-10-19 Jn Biosciences Llc Hybrid constant regions
US9382319B2 (en) 2011-09-26 2016-07-05 Jn Biosciences Llc Hybrid constant regions
HUE051439T2 (hu) 2011-10-11 2021-03-01 Viela Bio Inc CD40L-re specifikus, TN3-eredetû állványok és alkalmazásuk
LT2785375T (lt) 2011-11-28 2020-11-10 Merck Patent Gmbh Anti-pd-l1 antikūnai ir jų panaudojimas
CN109125718A (zh) 2012-01-13 2019-01-04 哈佛学院董事会 在结构聚合装置中tlr激动剂的控制传递
UY34591A (es) 2012-01-26 2013-09-02 Novartis Ag Compuestos de imidazopirrolidinona
UY34632A (es) 2012-02-24 2013-05-31 Novartis Ag Compuestos de oxazolidin- 2- ona y usos de los mismos
WO2013142255A2 (en) 2012-03-22 2013-09-26 University Of Miami Multi-specific binding agents
WO2013148228A1 (en) 2012-03-29 2013-10-03 Advanced Cancer Therapeutics, Llc Pfkfb3 inhibitor and methods of use as an anti-cancer therapeutic
CA2870309C (en) 2012-04-16 2024-02-20 President And Fellows Of Harvard College Mesoporous silica compositions for modulating immune responses
CA3085032A1 (en) 2012-04-17 2013-10-24 University Of Washington Through Its Center For Commercialization Hla class ii deficient cells, hla class i deficient cells capable of expressing hla class ii proteins, and uses thereof
PT2900637T (pt) 2012-05-15 2017-11-15 Novartis Ag Derivados de amida de pirimidina, piridina e pirazina substituídos com tiazole ou imidazole e compostos relacionados como inibidores de abl1, abl2 e bcr-abl1 para tratamento do cancro, infecções virais específicas e disturbios do snc específicos
KR20150008406A (ko) 2012-05-15 2015-01-22 노파르티스 아게 Abl1, abl2 및 bcr-abl1의 활성을 억제하기 위한 벤즈아미드 유도체
AP3613A (en) 2012-05-15 2016-02-29 Novartis Ag Benzamide derivatives for inhibiting the activity of abl1, abl2 and bcr-abl1 abl1abl2 bcr-abl1
MX2014013373A (es) 2012-05-15 2015-08-14 Novartis Ag Derivados de benzamida para inhibir la actividad de abl1, abl2 y bcr-abl1.
JO3300B1 (ar) 2012-06-06 2018-09-16 Novartis Ag مركبات وتركيبات لتعديل نشاط egfr
KR101566538B1 (ko) 2012-06-08 2015-11-05 국립암센터 신규한 Th17 세포 전환용 에피토프 및 이의 용도
UY34887A (es) 2012-07-02 2013-12-31 Bristol Myers Squibb Company Una Corporacion Del Estado De Delaware Optimización de anticuerpos que se fijan al gen de activación de linfocitos 3 (lag-3) y sus usos
CN112587658A (zh) 2012-07-18 2021-04-02 博笛生物科技有限公司 癌症的靶向免疫治疗
EP2880057A4 (en) 2012-08-02 2016-03-23 Jn Biosciences Llc MULTIMERIZED FUSION ANTIBODIES OR PROTEINS THROUGH THE MUTATION OF A CYSTEINE AND A PIECE OF TAIL
TWI472974B (zh) 2012-09-06 2015-02-11 Au Optronics Corp 多類物體觸控點偵測方法
AU2013334610B2 (en) 2012-10-24 2018-09-13 Novartis Ag IL-15R alpha forms, cells expressing IL-15R alpha forms, and therapeutic uses of IL-15R alpha and IL-15/IL-15R alpha complexes
CA2890663A1 (en) 2012-11-08 2014-05-15 Novartis Ag Pharmaceutical combination comprising a b-raf inhibitor and a histone deacetylase inhibitor and their use in the treatment of proliferative diseases
JP2016501221A (ja) 2012-11-28 2016-01-18 ノバルティス アーゲー 併用療法
EP3508215A3 (en) 2012-12-03 2019-10-02 Bristol-Myers Squibb Company Enhancing anti-cancer activity of immunomodulatory fc fusion proteins
WO2014116846A2 (en) 2013-01-23 2014-07-31 Abbvie, Inc. Methods and compositions for modulating an immune response
CA3185317A1 (en) 2013-02-08 2014-08-14 Novartis Ag Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
US9498532B2 (en) 2013-03-13 2016-11-22 Novartis Ag Antibody drug conjugates
BR112015021414B1 (pt) 2013-03-14 2020-11-10 Icahn School Of Medicine At Mount Sinai vírus da doença newcastle e seus usos
US9242969B2 (en) 2013-03-14 2016-01-26 Novartis Ag Biaryl amide compounds as kinase inhibitors
AP2015008707A0 (en) 2013-03-14 2015-09-30 Novartis Ag 3-pyrimidin-4-yl-oxazolidin-2-ones as inhibitors of mutant idh
AU2014232416B2 (en) 2013-03-15 2017-09-28 Xencor, Inc. Modulation of T Cells with Bispecific Antibodies and FC Fusions
JP6453855B2 (ja) 2013-05-18 2019-01-16 アドゥロ バイオテック,インク. 「インターフェロン遺伝子の刺激因子」依存性シグナル伝達を活性化するための組成物及び方法
JP6400082B2 (ja) 2013-05-18 2018-10-03 アデュロ バイオテック,インコーポレイテッド 「インターフェロン遺伝子の刺激因子」依存性シグナル伝達を抑制するための組成物および方法
AR097306A1 (es) 2013-08-20 2016-03-02 Merck Sharp & Dohme Modulación de la inmunidad tumoral
TW201605896A (zh) 2013-08-30 2016-02-16 安美基股份有限公司 Gitr抗原結合蛋白
MX355480B (es) 2013-11-01 2018-04-19 Novartis Ag Amino-heteroaril-benzamidas como inhibidores de cinasa.
US9512084B2 (en) 2013-11-29 2016-12-06 Novartis Ag Amino pyrimidine derivatives
GB201322583D0 (en) 2013-12-19 2014-02-05 Alligator Bioscience Ab Antibodies
JOP20200094A1 (ar) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc جزيئات جسم مضاد لـ pd-1 واستخداماتها
JOP20200096A1 (ar) 2014-01-31 2017-06-16 Children’S Medical Center Corp جزيئات جسم مضاد لـ tim-3 واستخداماتها
ME03558B (me) 2014-03-14 2020-07-20 Novartis Ag Molekuli anti-lag-3 antiтela i njihove upotrebe
SG11201610074YA (en) 2014-06-06 2016-12-29 Bristol Myers Squibb Co Antibodies against glucocorticoid-induced tumor necrosis factor receptor (gitr) and uses thereof
MA41044A (fr) 2014-10-08 2017-08-15 Novartis Ag Compositions et procédés d'utilisation pour une réponse immunitaire accrue et traitement contre le cancer

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Dixon (Proteins. 1997; Suppl 1: 198-204) *
Gussow et al. (Methods in Enzymology. 1991; 203: 99-121) *
Lensink et al. (Proteins. 2007; 69: 704-718) *
Pitt et al (I, 44:1255-1269, 2016) *
Tame (J. Comput. Aided Mol. Des. 1999 Mar; 13 (2): 99-108) *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11773188B2 (en) 2012-01-20 2023-10-03 Immunophotonics, Inc Chitosan-derived compositions
US10675358B2 (en) 2016-07-07 2020-06-09 The Board Of Trustees Of The Leland Stanford Junior University Antibody adjuvant conjugates
US11110178B2 (en) 2016-07-07 2021-09-07 The Board Of Trustees Of The Leland Standford Junior University Antibody adjuvant conjugates
US11547761B1 (en) 2016-07-07 2023-01-10 The Board Of Trustees Of The Leland Stanford Junior University Antibody adjuvant conjugates
US11364304B2 (en) 2016-08-25 2022-06-21 Northwestern University Crosslinked micellar spherical nucleic acids
US11696954B2 (en) 2017-04-28 2023-07-11 Exicure Operating Company Synthesis of spherical nucleic acids using lipophilic moieties
US11773180B2 (en) * 2017-11-08 2023-10-03 Kyowa Kirin Co., Ltd. Bispecific antibody which binds to CD40 and EpCAM
CN111918878A (zh) * 2018-04-20 2020-11-10 信达生物制药(苏州)有限公司 抗gitr抗体及其用途
US11400164B2 (en) 2019-03-15 2022-08-02 Bolt Biotherapeutics, Inc. Immunoconjugates targeting HER2
WO2020197578A1 (en) * 2019-03-27 2020-10-01 Immunophotonics, Inc. Semi-synthetic biopolymers for use in stimulating the immune system
CN111467474A (zh) * 2020-04-07 2020-07-31 江苏大学 一种具有免疫调节和抗肿瘤作用的药物

Also Published As

Publication number Publication date
CL2017000836A1 (es) 2017-09-15
IL251435A0 (en) 2017-05-29
PE20171654A1 (es) 2017-11-13
TN2017000120A1 (en) 2018-07-04
PH12017500634A1 (en) 2017-09-25
US20210015860A1 (en) 2021-01-21
ECSP17027870A (es) 2019-02-28
CO2017003408A2 (es) 2017-07-11
AU2019200346A1 (en) 2019-02-07
WO2016057841A1 (en) 2016-04-14
SG11201702596YA (en) 2017-04-27
US20170306037A1 (en) 2017-10-26
JP2017535257A (ja) 2017-11-30
EA201790799A1 (ru) 2017-08-31
MX2017004669A (es) 2017-06-19
US10662247B2 (en) 2020-05-26
CL2018001369A1 (es) 2018-07-27
EA036467B1 (ru) 2020-11-13
WO2016057846A1 (en) 2016-04-14
CR20170138A (es) 2017-08-21
CA2964146A1 (en) 2016-04-14
CN107001476B (zh) 2021-08-24
CU20170045A7 (es) 2017-09-06
KR20170065029A (ko) 2017-06-12
AU2015330771A1 (en) 2017-04-20
CU24437B1 (es) 2019-09-04
AU2019200346B2 (en) 2020-07-30
MA41044A (fr) 2017-08-15
EP3204419A1 (en) 2017-08-16
CN107001476A (zh) 2017-08-01
BR112017007093A2 (pt) 2018-03-06
EP3204421A1 (en) 2017-08-16
JP6731403B2 (ja) 2020-07-29
AU2015330771B2 (en) 2018-10-18

Similar Documents

Publication Publication Date Title
US20170306038A1 (en) Compositions and methods of use for augmented immune response and cancer therapy
US20220153835A1 (en) Combination therapies comprising antibody molecules to lag-3
US20220133889A1 (en) Combination therapies comprising antibody molecules to tim-3
US20210284737A1 (en) Antibody molecules to pd-l1 and uses thereof
CN108025051B (zh) 包含抗pd-1抗体分子的联合疗法
KR20180088907A (ko) Pd-1에 대한 항체 분자 및 그의 용도
WO2019018640A1 (en) POSOLOGICAL REGIMES FOR ANTI-GITREN ANTIBODIES AND USES THEREOF
US20240025993A1 (en) Cd19 binding molecules and uses thereof
RU2788092C2 (ru) Молекулы антител к pd-1 и их применения
EA040861B1 (ru) Молекулы антител к pd-l1 и их применение

Legal Events

Date Code Title Description
AS Assignment

Owner name: NOVARTIS AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC.;REEL/FRAME:044962/0552

Effective date: 20151029

Owner name: NOVARTIS AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS INSTITUTE FOR FUNCTIONAL GENOMICS, INC.;REEL/FRAME:044962/0574

Effective date: 20151027

Owner name: NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC.,

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BROGDON, JENNIFER;CIPOLLETTA, DANIELA;DRANOFF, GLENN;SIGNING DATES FROM 20151026 TO 20151027;REEL/FRAME:044962/0557

Owner name: NOVARTIS INSTITUTE FOR FUNCTIONAL GENOMICS, INC.,,

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KNEE, DEBORAH A.;WANG, FEI;REEL/FRAME:044962/0566

Effective date: 20151014

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: 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

STCB Information on status: application discontinuation

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