WO2019018640A1 - Régimes posologiques pour anticorps anti-gitr et utilisations associées - Google Patents

Régimes posologiques pour anticorps anti-gitr et utilisations associées Download PDF

Info

Publication number
WO2019018640A1
WO2019018640A1 PCT/US2018/042882 US2018042882W WO2019018640A1 WO 2019018640 A1 WO2019018640 A1 WO 2019018640A1 US 2018042882 W US2018042882 W US 2018042882W WO 2019018640 A1 WO2019018640 A1 WO 2019018640A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody molecule
dose
cancer
gitr antibody
inhibitor
Prior art date
Application number
PCT/US2018/042882
Other languages
English (en)
Inventor
Angelique BOURGOIN
Glenn Dranoff
Yan JI
Deborah Knee
Jürgen RECKTENWALD
Lisa NARDI
Original Assignee
Novartis Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis Ag filed Critical Novartis Ag
Publication of WO2019018640A1 publication Critical patent/WO2019018640A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies

Definitions

  • Glucocorticoid-induced TNFR-related protein is a member of the Tumor Necrosis Factor Superfamily (TNFRSF) which includes more than 20 type I transmembrane proteins, several splicing variants and several viral proteins, all of which have a cysteine-rich domain as a common structural feature.
  • TNFRSF Tumor Necrosis Factor Superfamily
  • the extracellular domain (ECD) of GITR consists of three 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.
  • 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. Various 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 with 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.
  • antibody molecules e.g., humanized antibody molecules
  • GITR Glucocorticoid-induced TNFR-related protein
  • Pharmaceutical compositions and dose formulations comprising the anti-GITR antibody molecules are also provided.
  • the anti-GITR antibody molecules disclosed herein can be used (alone or in combination with other therapeutic agents, procedures, or modalities, e.g., in combination with a Programmed Death 1 (PD-1) inhibitor, e.g. an anti-PD1 antibody molecule) to treat or prevent disorders, such as cancerous disorders (e.g., solid tumors and hematological cancers), as well as infectious diseases (e.g., chronic infectious disorders or sepsis).
  • PD-1 Programmed Death 1
  • an anti-PD1 antibody molecule an anti-PD1 antibody molecule
  • diseases e.g., chronic infectious disorders or sepsis
  • methods, including dosage regimens, for treating various disorders using the anti-GITR antibody molecules are disclosed herein.
  • the GITR antibody molecule is administered or used
  • the disclosure features a method of treating (e.g., inhibiting, reducing, ameliorating, or preventing) a cancer in a subject.
  • the method including administering to the subject an anti-GITR antibody molecule (e.g., an anti-GITR antibody molecule described herein) at a dose of about 2 mg to about 600 mg once every week, once every three weeks, or once every six weeks.
  • an anti-GITR antibody molecule e.g., an anti-GITR antibody molecule described herein
  • the anti-GITR antibody molecule includes: a heavy chain variable region (VH) including a VHCDR1 amino acid sequence of SEQ ID NO: 909, a VHCDR2 amino acid sequence of SEQ ID NO: 911, and a VHCDR3 amino acid sequence of SEQ ID NO: 913; and a light chain variable region (VL) including a VLCDR1 amino acid sequence of SEQ ID NO: 914, a VLCDR2 amino acid sequence of SEQ ID NO: 916, and a VLCDR3 amino acid sequence of SEQ ID NO: 918.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 600 mg once every week. In other embodiments, the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 600 mg once every three weeks. In other embodiments, the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 600 mg once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 600 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, or about 400 mg to about 600 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, or about 500 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 250 mg to about 500 mg, e.g., about 375 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 500 mg to about 1000 mg, e.g., about 750 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 800 mg to about 1600 mg, e.g., about 1200 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 600 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, or about 400 mg to about 600 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, or about 500 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 100 mg, e.g., about 10 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 600 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, or about 400 mg to about 600 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, or about 500 mg, once everysix weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 100 mg, e.g., about 10 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every six weeks.
  • the disclosure features a method of treating (e.g., inhibiting, reducing, ameliorating, or preventing) a cancer in a subject.
  • the method including administering to the subject an anti-GITR antibody molecule (e.g., an anti-GITR antibody molecule described herein) at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg once every week, once every three weeks, or once every six weeks.
  • an anti-GITR antibody molecule e.g., an anti-GITR antibody molecule described herein
  • the anti-GITR antibody molecule includes: a heavy chain variable region (VH) including a VHCDR1 amino acid sequence of SEQ ID NO: 909, a VHCDR2 amino acid sequence of SEQ ID NO: 911, and a VHCDR3 amino acid sequence of SEQ ID NO: 913; and a light chain variable region (VL) including a VLCDR1 amino acid sequence of SEQ ID NO: 914, a VLCDR2 amino acid sequence of SEQ ID NO: 916, and a VLCDR3 amino acid sequence of SEQ ID NO: 918.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg once every week. In other embodiments, the anti- GITR antibody molecule is administered at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg once every three weeks. In other embodiments, the anti-GITR antibody molecule is
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg , e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, about 250 mg to about 500 mg, about 400 mg to about 600 mg, about 500 mg to about 1000 mg, about 800 mg to about 1600 mg, about 1000 mg to about 1800 mg, about 1200 mg to about 1600 mg, about 1400 mg to about 1600 mg, about 1400 mg to about 1800 mg, about 1000 mg to about 2000 mg, about 2000 mg to about 2500 mg, about 2800 mg to about 3200 mg, or about 2500 mg to about 3500 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, about 375 mg, about 500 mg, about 750 mg, about 1200 mg, about 1500 mg, about 2250 mg, e.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 250 mg to about 500 mg, e.g., about 375 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 500 mg to about 1000 mg, e.g., about 750 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 800 mg to about 1600 mg, e.g., about 1200 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 2000 mg, e.g., about 1500 mg, once every week. In certain embodiments, the anti- GITR antibody molecule is administered at a dose of about 2000 mg to about 2500 mg, e.g., about 2250 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 2500 mg to about 3500 mg, e.g., about 3000 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, about 250 mg to about 500 mg, about 400 mg to about 600 mg, about 500 mg to about 1000 mg, about 800 mg to about 1600 mg, about 1000 mg to about 1800 mg, about 1200 mg to about 1600 mg, about 1400 mg to about 1600 mg, about 1400 mg to about 1800 mg, about 1000 mg to about 2000 mg, about 2000 mg to about 2500 mg, about 2800 mg to about 3200 mg, or about 2500 mg to about 3500 mg, e.g., about 5 mg, about 10 mg
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 250 mg to about 500 mg, e.g., about 375 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 500 mg to about 1000 mg, e.g., about 750 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 800 mg to about 1600 mg, e.g., about 1200 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 2000 mg, e.g., about 1500 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 2000 mg to about 2500 mg, e.g., about 2250 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2500 mg to about 3500 mg, e.g., about 3000 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, about 250 mg to about 500 mg, about 400 mg to about 600 mg, about 500 mg to about 1000 mg, about 800 mg to about 1600 mg, about 1000 mg to about 1800 mg, about 1200 mg to about 1600 mg, about 1400 mg to about 1600 mg, about 1400 mg to about 1800 mg, about 1000 mg to about 2000 mg, about 2000 mg to about 2500 mg, about 2800 mg to about 3200 mg, or about 2500 mg to about 3500 mg, e.g., about 5 mg, about 10
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 250 mg to about 500 mg, e.g., about 375 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 500 mg to about 1000 mg, e.g., about 750 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 800 mg to about 1600 mg, e.g., about 1200 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 2000 mg, e.g., about 1500 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 2000 mg to about 2500 mg, e.g., about 2250 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2500 mg to about 3500 mg, e.g., about 3000 mg, once every six weeks.In an embodiment, the anti-GITR antibody molecule is administered at a dose of about 375 mg once every week. In an embodiment, the anti-GITR antibody molecule is administered at a dose of about 375 mg once every three weeks. In an embodiment, the anti-GITR antibody molecule is administered at a dose of about 375 mg once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 750 mg once every week. In an embodiment, the anti-GITR antibody molecule is administered at a dose of about 750 mg once every three weeks. In an embodiment, the anti-GITR antibody molecule is administered at a dose of about 750 mg once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 1500 mg once every week. In an embodiment, the anti-GITR antibody molecule is administered at a dose of about 1500 mg once every three weeks. In an embodiment, the anti-GITR antibody molecule is administered at a dose of about 1500 mg once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2250 mg once every week. In an embodiment, the anti-GITR antibody molecule is administered at a dose of about 2250 mg once every three weeks. In an embodiment, the anti-GITR antibody molecule is administered at a dose of about 2250 mg once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 3000 mg once every week. In an embodiment, the anti-GITR antibody molecule is administered at a dose of about 3000 mg once every three weeks. In an embodiment, the anti-GITR antibody molecule is administered at a dose of about 3000 mg once every six weeks.
  • the subject receives three doses of the anti-GITR antibody molecule over a period of two to four weeks (e.g., three weeks) followed by a eight- to ten-week (e.g., nine-week) pause. In other embodiments, the subject receives four doses of the anti-GITR antibody molecule over a period of ten to fifteen weeks (e.g., twelve weeks) followed by a eight- to ten-week (e.g., nine-week) pause.
  • the subject receives four doses of the anti-GITR antibody molecule over a period of twenty to twenty-five weeks (e.g., twenty-one to twenty-four weeks, e.g., twenty-one or twenty-four weeks) followed by a eight- to ten-week (e.g., nine-week) pause.
  • the anti-GITR antibody molecule includes a VH including the amino acid sequence of SEQ ID NO: 901 and a VL including the amino acid sequence of SEQ ID NO: 902. In one embodiment, the anti-GITR antibody molecule includes a heavy chain including the amino acid sequence of SEQ ID NO: 903 and a light chain including the amino acid sequence of SEQ ID NO: 904.
  • the cancer cell can be, e.g., a cell from a cancer described herein, such as a solid tumor or a hematological cancer, e.g., 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 a small cell lung cancer (SCLC)), a head and neck cancer (e.g., squamous cell carcinoma of the head and neck (SCCHN)), a nasopharyngeal cancer (NPC), a skin cancer (e.g., a Merkel cell carcinoma or a melanoma (e.g., an advanced melanoma)), a neuroendocrine cancer, a neuroectodermal cancer, a kidney cancer (e.g., a renal cancer (e.g., a renal
  • the cancer is a solid tumor.
  • the cancer is a lung cancer, e.g., a NSCLC, e.g., a NSCLC with squamous and/or non-squamous histology, or a NSCLC adenocarcinoma, or a SCLC.
  • the cancer is a head and neck cancer, e.g., SCCHN.
  • the cancer is a NPC.
  • the cancer is a skin cancer, e.g., a Merkel cell carcinoma or a melanoma, e.g., an advanced melanoma.
  • the cancer is a neuroendocrine cancer.
  • the cancer is a neuroectodermal cancer.
  • the cancer is a kidney cancer, e.g., a renal cancer, e.g., a RCC.
  • the cancer is an endometrial cancer, e.g., an adenocarcinoma of the endometrium.
  • the cancer is a cervical cancer.
  • the cancer is a gastric cancer, e.g., a stomach cancer, an esophageal cancer, or a cancer of the gastroesophageal junction.
  • the cancer is a liver cancer, e.g., a HCC.
  • the cancer is a mesothelioma.
  • the cancer is a colorectal cancer, e.g., an MSI-H colorectal cancer.
  • the cancer is an ovarian cancer.
  • the cancer is a breast cancer, e.g., a TNBC.
  • the cancer is a urothelial cancer, e.g., a urothelial carcinoma.
  • the cancer is a bladder cancer, e.g. a transitional cell carcinoma.
  • the cancer is a hematological cancer.
  • the cancer is a lymphoma (e.g., T-cell lymphoma, B-cell lymphoma, or a non-Hogdkin lymphoma).
  • the cancer is an MSI-high cancer. In some embodiments, the cancer is a metastatic cancer. In other embodiments, the cancer is an advanced cancer. In other embodiments, the cancer is a relapsed or refractory cancer.
  • the anti-GITR antibody molecule is used in combination with a second therapeutic agent or modality. In certain embodiments, the anti-GITR antibody molecule is used in combination with a PD-1 inhibitor, e.g., a PD-1 inhibitor described herein. In some embodiments, the PD-1 inhibitor is chosen from PDR001, nivolumab, pembrolizumab, pidilizumab, MEDI0680,
  • the PD-1 inhibitor is used at a dose of about 50 mg to about 400 mg once every two weeks, once every three weeks, or once every six weeks. In some embodiments, the PD-1 inhibitor is used at a dose of about 50 mg to about 200 mg once every two weeks, once every three weeks, or once every six weeks. In some embodiments, the PD-1 inhibitor is used at a dose of about 100 mg to about 300 mg once every two weeks, once every three weeks, or once every six weeks. In some embodiments, the PD-1 inhibitor is used at a dose of about 100 mg to about 400 mg once every two weeks, once every three weeks, or once every six weeks. In some embodiments, the PD-1 inhibitor is used at a dose of about 200 mg to about 400 mg once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is used at a dose of about 2 mg to about 10 mg and the PD-1 inhibitor is used at a dose of about 50 mg to about 200 mg; the anti-GITR antibody molecule is used at a dose of about 5 mg to about 20 mg and the PD-1 inhibitor is used at a dose of about 50 mg to about 200 mg; the anti-GITR antibody molecule is used at a dose of about 5 mg to about 20 mg and the PD-1 inhibitor is used at a dose of about 100 mg to about 300 mg; the anti-GITR antibody molecule is used at a dose of about 20 mg to about 40 mg and the PD-1 inhibitor is used at a dose of about 50 mg to about 200 mg; the anti-GITR antibody molecule is used at a dose of about 20 mg to about 40 mg and the PD-1 inhibitor is used at a dose of about 100 mg to about 400 mg; the anti-GITR antibody molecule is used at a dose of about 50 mg to about 100 mg and the PD-1 inhibitor is used at a dose of a dose of
  • the anti-GITR antibody molecule is used at a dose of about 5 mg and the PD-1 inhibitor is used at a dose of about 100 mg; the anti-GITR antibody molecule is used at a dose of about 10 mg and the PD-1 inhibitor is used at a dose of about 100 mg; the anti-GITR antibody molecule is used at a dose of about 10 mg and the PD-1 inhibitor is used at a dose of about 200 mg; the anti-GITR antibody molecule is used at a dose of about 30 mg and the PD-1 inhibitor is used at a dose of about 100 mg; the anti-GITR antibody molecule is used at a dose of about 30 mg and the PD-1 inhibitor is used at a dose of about 300 mg; the anti-GITR antibody molecule is used at a dose of about 60 mg and the PD-1 inhibitor is used at a dose of about 100 mg; the anti-GITR antibody molecule is used at a dose of about 75 mg and the PD-1 inhibitor is used at a dose of about 300 mg; the anti-GITR
  • the anti-GITR antibody molecule is used at a dose of about 2 mg to about 10 mg and the PD-1 inhibitor is used at a dose of about 50 mg to about 200 mg; the anti-GITR antibody molecule is used at a dose of about 5 mg to about 20 mg and the PD-1 inhibitor is used at a dose of about 50 mg to about 200 mg; the anti-GITR antibody molecule is used at a dose of about 5 mg to about 20 mg and the PD-1 inhibitor is used at a dose of about 100 mg to about 300 mg; the anti-GITR antibody molecule is used at a dose of about 20 mg to about 40 mg and the PD-1 inhibitor is used at a dose of about 50 mg to about 200 mg; the anti-GITR antibody molecule is used at a dose of about 20 mg to about 40 mg and the PD-1 inhibitor is used at a dose of about 100 mg to about 400 mg; the anti-GITR antibody molecule is used at a dose of about 50 mg to about 100 mg and the PD-1 inhibitor is used at a dose of a dose of
  • the anti-GITR antibody molecule is used at a dose of about 5 mg and the PD-1 inhibitor is used at a dose of about 100 mg; the anti-GITR antibody molecule is used at a dose of about 10 mg and the PD-1 inhibitor is used at a dose of about 100 mg; the anti-GITR antibody molecule is used at a dose of about 10 mg and the PD-1 inhibitor is used at a dose of about 200 mg; the anti-GITR antibody molecule is used at a dose of about 30 mg and the PD-1 inhibitor is used at a dose of about 100 mg; the anti-GITR antibody molecule is used at a dose of about 30 mg and the PD-1 inhibitor is used at a dose of about 300 mg; the anti-GITR antibody molecule is used at a dose of about 60 mg and the PD-1 inhibitor is used at a dose of about 100 mg; the anti-GITR antibody molecule is used at a dose of about 75 mg and the PD-1 inhibitor is used at a dose of about 300 mg; the anti-GITR
  • the anti-GITR antibody molecule is used at a dose of about 2 mg to about 10 mg and the PD-1 inhibitor is used at a dose of about 50 mg to about 200 mg; the anti-GITR antibody molecule is used at a dose of about 5 mg to about 20 mg and the PD-1 inhibitor is used at a dose of about 50 mg to about 200 mg; the anti-GITR antibody molecule is used at a dose of about 5 mg to about 20 mg and the PD-1 inhibitor is used at a dose of about 100 mg to about 300 mg; the anti-GITR antibody molecule is used at a dose of about 20 mg to about 40 mg and the PD-1 inhibitor is used at a dose of about 50 mg to about 200 mg; the anti-GITR antibody molecule is used at a dose of about 20 mg to about 40 mg and the PD-1 inhibitor is used at a dose of about 100 mg to about 400 mg; the anti-GITR antibody molecule is used at a dose of about 50 mg to about 100 mg and the PD-1 inhibitor is used at a dose of a dose of
  • the anti-GITR antibody molecule is used at a dose of about 5 mg and the PD-1 inhibitor is used at a dose of about 100 mg; the anti-GITR antibody molecule is used at a dose of about 10 mg and the PD-1 inhibitor is used at a dose of about 100 mg; the anti-GITR antibody molecule is used at a dose of about 10 mg and the PD-1 inhibitor is used at a dose of about 200 mg; the anti-GITR antibody molecule is used at a dose of about 30 mg and the PD-1 inhibitor is used at a dose of about 100 mg; the anti-GITR antibody molecule is used at a dose of about 30 mg and the PD-1 inhibitor is used at a dose of about 300 mg; the anti-GITR antibody molecule is used at a dose of about 60 mg and the PD-1 inhibitor is used at a dose of about 100 mg; the anti-GITR antibody molecule is used at a dose of about 75 mg and the PD-1 inhibitor is used at a dose of about 300 mg; the anti-GITR
  • the anti-GITR antibody molecule is used at a dose of about 10 mg once every week and the PD-1 inhibitor is used at a dose of about 100 mg once every two weeks. In an embodiment, the anti-GITR antibody molecule is used at a dose of about 10 mg once every week and the PD-1 inhibitor is used at a dose of about 200 mg once every two weeks. In an embodiment, the anti-GITR antibody molecule is used at a dose of about 30 mg once every week and the PD-1 inhibitor is used at a dose of about 100 mg once every two weeks. In an embodiment, the anti-GITR antibody molecule is used at a dose of about 30 mg once every week and the PD-1 inhibitor is used at a dose of about 300 mg once every two weeks.
  • the anti-GITR antibody molecule is used at a dose of about 75 mg once every week and the PD-1 inhibitor is used at a dose of about 300 mg once every two weeks. In an embodiment, the anti-GITR antibody molecule is used at a dose of about 1500 mg once every week and the PD-1 inhibitor is used at a dose of about 300 mg once every two weeks.In an embodiment, the anti-GITR antibody molecule is used at a dose of about 10 mg once every three weeks and the PD-1 inhibitor is used at a dose of about 100 mg once every three weeks. In an embodiment, the anti-GITR antibody molecule is used at a dose of about 10 mg once every three weeks and the PD-1 inhibitor is used at a dose of about 200 mg once once every three weeks.
  • the anti-GITR antibody molecule is used at a dose of about 30 mg once every three weeks and the PD-1 inhibitor is used at a dose of about 100 mg once every three weeks. In an embodiment, the anti-GITR antibody molecule is used at a dose of about 30 mg once every three weeks and the PD-1 inhibitor is used at a dose of about 300 mg once every three weeks. In an embodiment, the anti-GITR antibody molecule is used at a dose of about 75 mg once every three weeks and the PD-1 inhibitor is used at a dose of about 300 mg once every three weeks. In an embodiment, the anti-GITR antibody molecule is used at a dose of about 1500 mg once every three weeks and the PD-1 inhibitor is used at a dose of about 300 mg once every three weeks.
  • the anti-GITR antibody molecule is used at a dose of about 10 mg once every six weeks and the PD-1 inhibitor is used at a dose of about 100 mg once every six weeks. In an embodiment, the anti-GITR antibody molecule is used at a dose of about 10 mg once every six weeks and the PD-1 inhibitor is used at a dose of about 200 mg once once every six weeks. In an embodiment, the anti-GITR antibody molecule is used at a dose of about 30 mg once every six weeks and the PD-1 inhibitor is used at a dose of about 100 mg once every six weeks. In an embodiment, the anti-GITR antibody molecule is used at a dose of about 30 mg once every six weeks and the PD-1 inhibitor is used at a dose of about 300 mg once every six weeks.
  • the anti-GITR antibody molecule is used at a dose of about 75 mg once every six weeks and the PD-1 inhibitor is used at a dose of about 300 mg once every six weeks. In an embodiment, the anti-GITR antibody molecule is used at a dose of about 1500 mg once every six weeks and the PD-1 inhibitor is used at a dose of about 300 mg once every six weeks.
  • the anti-GITR antibody molecule and the PD-1 inhibitor is used on the same day. In certain embodiments, the anti-GITR antibody molecule and the PD-1 inhibitor is used on different days. For example, the anti-PD-1 inhibitor can be given on days 1 and 15 with the anti-GITR antibody molecule.
  • the subject receives two doses of the PD-1 inhibitor over a period of three weeks followed by a eight- to ten-week (e.g., nine-week) pause. In other embodiments, the subject receives four doses of the PD-1 inhibitor over a period of twelve weeks followed by a eight- to ten-week (e.g., nine-week) pause. In other embodiments, the subject receives four doses of the PD-1 inhibitor over a period of twenty-one or twenty four weeks followed by a eight- to ten-week (e.g., nine-week) pause. Any of the doses disclosed herein can be repeated once, twice, three times, four times, five time, six time, seven time, eight times, nine times, ten times, or more.
  • the anti-GITR antibody molecule is used in combination with a PD-L1 inhibitor (e.g., a PD-L1 inhibitor as described herein), e.g., FAZ053, atezolizumab, avelumab, durvalumab, or BMS-936559.
  • a PD-L1 inhibitor e.g., a PD-L1 inhibitor as described herein
  • the anti-GITR antibody molecule is used in combination with a CTLA-4 inhibitor (e.g., a CTLA-4 inhibitor as described herein).
  • the anti-GITR antibody molecule is used in combination with a LAG-3 inhibitor (e.g., a LAG-3 inhibitor as described herein).
  • the anti-GITR antibody molecule is used in combination with a CSF1R inhibitor (e.g., a CSF1R inhibitor as described herein). In other embodiments, the anti-GITR antibody molecule is used in combination with a IL-15/IL-15Ra complex (e.g., a IL-15/IL- 15Ra complex as described herein).
  • a CSF1R inhibitor e.g., a CSF1R inhibitor as described herein
  • a IL-15/IL-15Ra complex e.g., a IL-15/IL- 15Ra complex as described herein.
  • the subject has, or is identified as having, a cancer that expresses one or more of: a marker chosen from FoxP3, CD8, CD56, PD-L1, CD68, CD4, or a combination thereof; a cytokine, e.g., INF- ⁇ , GM-CSF, TNF- ⁇ , IL-1, IL-2, IL-5, IL-6, IL-10, IL-12, IL-17, IL-18, VEGF, or a combination thereof, or chemokine; or an immune cell activation marker, e.g., CD3, CD8, HLA-DR, GITR, or a combination thereof.
  • the subject has, or is identified as having, a cancer that expresses FoxP3.
  • the subject has, or is identified as having, GITR expression in tumor-infiltrating lymphocytes (TILs).
  • TILs tumor-infiltrating lymphocytes
  • the subject has, or is identified as having, a tumor mutation load that is higher than a reference level.
  • the subject has, or is identified as having, a cancer that expresses PD-L1.
  • the subject had not received a therapy including a PD-1 inhibitor or a PD-L1 inhibitor before receiving the anti-GITR antibody molecule. In other embodiments, the subject had received a therapy including a PD-1 inhibitor or a PD-L1 inhibitor before receiving the anti-GITR antibody molecule.
  • the disclosure features a method of reducing an activity (e.g., growth, survival, or viability, or all), of a hyperproliferative (e.g., a cancer) cell. The method includes contacting the cell with an anti-GITR antibody molecule, e.g., an anti-GITR antibody molecule described herein.
  • the method can be performed in a subject, e.g., as part of a therapeutic protocol, e.g., at a dose of about 2 mg to about 600 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, or about 400 mg to about 600 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, or about 500 mg, of an anti-GITR antibody molecule, once every week, once every three weeks, or once every six weeks.
  • a dose of about 2 mg to about 600 mg e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, or about 400 mg to about 600 mg, e.g., about 5 mg, about 10
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 600 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, or about 400 mg to about 600 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, or about 500 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 600 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, or about 400 mg to about 600 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, or about 500 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 100 mg, e.g., about 10 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 600 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, or about 400 mg to about 600 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, or about 500 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 100 mg, e.g., about 10 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every six weeks.
  • the disclosure features a method of reducing an activity (e.g., growth, survival, or viability, or all), of a hyperproliferative (e.g., a cancer) cell.
  • the method includes contacting the cell with an anti-GITR antibody molecule, e.g., an anti-GITR antibody molecule described herein.
  • the method can be performed in a subject, e.g., as part of a therapeutic protocol, e.g., at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, about 250 mg to about 500 mg, about 400 mg to about 600 mg, about 500 mg to about 1000 mg, about 800 mg to about 1600 mg, about 1000 mg to about 1800 mg, about 1200 mg to about 1600 mg, about 1400 mg to about 1600 mg, about 1400 mg to about 1800 mg, about 1000 mg to about 2000 mg, about 2000 mg to about 2500 mg, about 2800 mg to about 3200 mg, or about 2500 mg to about 3500 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, about 375 mg, about 500 mg, about 750
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, about 250 mg to about 500 mg, about 400 mg to about 600 mg, about 500 mg to about 1000 mg, about 800 mg to about 1600 mg, about 1000 mg to about 1800 mg, about 1200 mg to about 1600 mg, about 1400 mg to about 1600 mg, about 1400 mg to about 1800 mg, about 1000 mg to about 2000 mg, about 2000 mg to about 2500 mg, about 2800 mg to about 3200 mg, or about 2500 mg to about 3500 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, about 375 mg, about 500 mg, about 750 mg, about 1200 mg, about 1500 mg, about 2250 mg,
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 250 mg to about 500 mg, e.g., about 375 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 500 mg to about 1000 mg, e.g., about 750 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 800 mg to about 1600 mg, e.g., about 1200 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 2000 mg, e.g., about 1500 mg, once every week. In certain embodiments, the anti- GITR antibody molecule is administered at a dose of about 2000 mg to about 2500 mg, e.g., about 2250 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 2500 mg to about 3500 mg, e.g., about 3000 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, about 250 mg to about 500 mg, about 400 mg to about 600 mg, about 500 mg to about 1000 mg, about 800 mg to about 1600 mg, about 1000 mg to about 1800 mg, about 1200 mg to about 1600 mg, about 1400 mg to about 1600 mg, about 1400 mg to about 1800 mg, about 1000 mg to about 2000 mg, about 2000 mg to about 2500 mg, about 2800 mg to about 3200 mg, or about 2500 mg to about 3500 mg, e.g., about 5 mg, about 10 mg
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 250 mg to about 500 mg, e.g., about 375 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 500 mg to about 1000 mg, e.g., about 750 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 800 mg to about 1600 mg, e.g., about 1200 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 2000 mg, e.g., about 1500 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 2000 mg to about 2500 mg, e.g., about 2250 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 2500 mg to about 3500 mg, e.g., about 3000 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, about 250 mg to about 500 mg, about 400 mg to about 600 mg, about 500 mg to about 1000 mg, about 800 mg to about 1600 mg, about 1000 mg to about 1800 mg, about 1200 mg to about 1600 mg, about 1400 mg to about 1600 mg, about 1400 mg to about 1800 mg, about 1000 mg to about 2000 mg, about 2000 mg to about 2500 mg, about 2800 mg to about 3200 mg, or about 2500 mg to about 3500 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, about 375 mg, about 500 mg, about 750 mg, about 1200 mg, about 1500 mg, about 2250 mg,
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 250 mg to about 500 mg, e.g., about 375 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 500 mg to about 1000 mg, e.g., about 750 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 800 mg to about 1600 mg, e.g., about 1200 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 2000 mg, e.g., about 1500 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 2000 mg to about 2500 mg, e.g., about 2250 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 2500 mg to about 3500 mg, e.g., about 3000 mg, once every six weeks.
  • the cancer cell can be, e.g., a cell from a cancer described herein, such as a solid tumor or a hematological cancer, e.g., 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 a small cell lung cancer (SCLC)), a head and neck cancer (e.g., squamous cell carcinoma of the head and neck (SCCHN)), a nasopharyngeal cancer (NPC), a skin cancer (e.g., a Merkel cell carcinoma or a melanoma (e.g., an advanced melanoma)), a neuroendocrine cancer, a neuroectodermal cancer, a kidney cancer (e.g., a renal cancer (e.g., a renal cell carcinoma (RCC
  • the cancer is an MSI-high cancer. In some embodiments, the cancer is a metastatic cancer. In other embodiments, the cancer is an advanced cancer. In other embodiments, the cancer is a relapsed or refractory cancer. In certain embodiments of the methods disclosed herein, the method further includes determining the level of GITR expression, e.g., in tumor infiltrating lymphocytes (TILs), in the subject. In other embodiments, the level of GITR expression is determined in a sample (e.g., a tumor biopsy) acquired from the subject (e.g., using immunohistochemistry).
  • TILs tumor infiltrating lymphocytes
  • the anti-GITR antibody molecule is administered.
  • the detection steps can also be used, e.g., to monitor the effectiveness of a therapeutic agent described herein.
  • the detection step can be used to monitor the effectiveness of the anti-GITR antibody molecule.
  • the disclosure features a pharmaceutical composition or dose formulation (e.g., one or more compositions or dosage forms), that includes an anti-GITR antibody molecule (e.g., an anti- GITR antibody molecule described herein), a buffering agent, a carbohydrate, and a surfactant.
  • the anti-GITR antibody molecule includes: a heavy chain variable region (VH) including a VHCDR1 amino acid sequence of SEQ ID NO: 909, a VHCDR2 amino acid sequence of SEQ ID NO: 911, and a VHCDR3 amino acid sequence of SEQ ID NO: 913; and a light chain variable region (VL) including a VLCDR1 amino acid sequence of SEQ ID NO: 914, a VLCDR2 amino acid sequence of SEQ ID NO: 916, and a VLCDR3 amino acid sequence of SEQ ID NO: 918.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-GITR antibody molecule includes a VH including the amino acid sequence of SEQ ID NO: 901 and a VL including the amino acid sequence of SEQ ID NO: 902. In one embodiment, the anti-GITR antibody molecule includes a heavy chain including the amino acid sequence of SEQ ID NO: 903 and a light chain including the amino acid sequence of SEQ ID NO: 904.
  • the pharmaceutical composition or dose formulation includes about 100 mg/mL to about 200 mg/mL of the anti-GITR antibody molecule.
  • the buffering agent is present at a concentration of about 10 mM to about 30 mM.
  • the buffering agent has a pH of about 5 to about 6.
  • the buffering agent includes a histidine buffer.
  • the carbohydrate e.g., sucrose
  • the surfactant e.g., polysorbate 20
  • the pharmaceutical composition or dose formulation includes about 100 mg/mL to about 200 mg/mL of the anti-GITR antibody molecule, a buffering agent present at a concentration of about 10 mM to about 30 mM and at a pH of about 5 to about 6, a carbohydrate present at a concentration of about 200 mM to about 250 mM, and a surfactant present at a concentration of about 0.03% to about 0.05%.
  • the pharmaceutical composition or dose formulation includes about 150 mg/mL of the anti-GITR antibody molecule, a histidine buffer present at a concentration of about 20 mM and at a pH of about 5.5, sucrose present at a concentration of about 220 mM, and polysorbate-20 present at a concentration of about 0.03% to about 0.05%.
  • the disclosure features a pharmaceutical composition or dose formulation that includes an anti-GITR antibody molecule (e.g., an anti-GITR antibody molecule described herein) for use at a dose of about 2 mg to about 600 mg once every week, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule includes: a heavy chain variable region (VH) including a VHCDR1 amino acid sequence of SEQ ID NO: 909, a VHCDR2 amino acid sequence of SEQ ID NO: 911, and a VHCDR3 amino acid sequence of SEQ ID NO: 913; and a light chain variable region (VL) including a VLCDR1 amino acid sequence of SEQ ID NO: 914, a VLCDR2 amino acid sequence of SEQ ID NO: 916, and a VLCDR3 amino acid sequence of SEQ ID NO: 918.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-GITR antibody molecule is used at a dose of about 2 mg to about 600 mg once every week. In other embodiments, the anti-GITR antibody molecule is used at a dose of about 2 mg to about 600 mg once every three weeks. In other embodiments, the anti-GITR antibody molecule is used at a dose of about 2 mg to about 600 mg once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 600 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, or about 400 mg to about 600 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, or about 500 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 600 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, or about 400 mg to about 600 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, or about 500 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 100 mg, e.g., about 10 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 600 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, or about 400 mg to about 600 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, or about 500 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 100 mg, e.g., about 10 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every six weeks.
  • the disclosure features a pharmaceutical composition or dose formulation that includes an anti-GITR antibody molecule (e.g., an anti-GITR antibody molecule described herein) for use at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg once every week, once every three weeks, or once every six weeks.
  • an anti-GITR antibody molecule e.g., an anti-GITR antibody molecule described herein
  • the anti-GITR antibody molecule includes: a heavy chain variable region (VH) including a VHCDR1 amino acid sequence of SEQ ID NO: 909, a VHCDR2 amino acid sequence of SEQ ID NO: 911, and a VHCDR3 amino acid sequence of SEQ ID NO: 913; and a light chain variable region (VL) including a VLCDR1 amino acid sequence of SEQ ID NO: 914, a VLCDR2 amino acid sequence of SEQ ID NO: 916, and a VLCDR3 amino acid sequence of SEQ ID NO: 918.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-GITR antibody molecule is used at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg once every week.
  • the anti-GITR antibody molecule is used at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg once every three weeks. In other embodiments, the anti-GITR antibody molecule is used at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, about 250 mg to about 500 mg, about 400 mg to about 600 mg, about 500 mg to about 1000 mg, about 800 mg to about 1600 mg, about 1000 mg to about 1800 mg, about 1200 mg to about 1600 mg, about 1400 mg to about 1600 mg, about 1400 mg to about 1800 mg, about 1000 mg to about 2000 mg, about 2000 mg to about 2500 mg, about 2800 mg to about 3200 mg, or about 2500 mg to about 3500 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, about 375 mg, about 500 mg, about 750 mg, about 1200 mg, about 1500 mg, about 2250 mg,
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every week. In certain embodiments, the anti- GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 250 mg to about 500 mg, e.g., about 375 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 500 mg to about 1000 mg, e.g., about 750 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 800 mg to about 1600 mg, e.g., about 1200 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 2000 mg, e.g., about 1500 mg, once every week. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 2000 mg to about 2500 mg, e.g., about 2250 mg, once every week.
  • the anti-GITR antibody molecule is administered at a dose of about 2500 mg to about 3500 mg, e.g., about 3000 mg, once every week. In some embodiments, the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, about 250 mg to about 500 mg, about 400 mg to about 600 mg, about 500 mg to about 1000 mg, about 800 mg to about 1600 mg, about 1000 mg to about 1800 mg, about 1200 mg to about 1600 mg, about 1400 mg to about 1600 mg, about 1400 mg to about 1800 mg, about 1000 mg to about 2000 mg, about 2000 mg to about 2500 mg, about 2800 mg to about 3200 mg, or about 2500 mg to about 3500 mg, e.g., about 5 mg, about 10 mg
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 250 mg to about 500 mg, e.g., about 375 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 500 mg to about 1000 mg, e.g., about 750 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 800 mg to about 1600 mg, e.g., about 1200 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 2000 mg, e.g., about 1500 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 2000 mg to about 2500 mg, e.g., about 2250 mg, once every three weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 2500 mg to about 3500 mg, e.g., about 3000 mg, once every three weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 2400 mg or about 2 mg to about 4000 mg, e.g., about 2 mg to about 10 mg, about 5 mg to about 20 mg, about 20 mg to about 40 mg, about 50 mg to about 100 mg, about 100 mg to about 200 mg, about 200 mg to about 400 mg, about 250 mg to about 500 mg, about 400 mg to about 600 mg, about 500 mg to about 1000 mg, about 800 mg to about 1600 mg, about 1000 mg to about 1800 mg, about 1200 mg to about 1600 mg, about 1400 mg to about 1600 mg, about 1400 mg to about 1800 mg, about 1000 mg to about 2000 mg, about 2000 mg to about 2500 mg, about 2800 mg to about 3200 mg, or about 2500 mg to about 3500 mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, about 150 mg, about 300 mg, about 375 mg, about 500 mg, about 750 mg, about 1200 mg, about 1500 mg, about 2250 mg,
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg, e.g., about 30 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg, e.g., about 60 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 250 mg to about 500 mg, e.g., about 375 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg, e.g., about 500 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 500 mg to about 1000 mg, e.g., about 750 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 800 mg to about 1600 mg, e.g., about 1200 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 2000 mg, e.g., about 1500 mg, once every six weeks. In certain embodiments, the anti-GITR antibody molecule is
  • the anti-GITR antibody molecule is administered at a dose of about 2000 mg to about 2500 mg, e.g., about 2250 mg, once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2500 mg to about 3500 mg, e.g., about 3000 mg, once every six weeks.
  • the anti-GITR antibody molecule includes a VH including the amino acid sequence of SEQ ID NO: 901 and a VL including the amino acid sequence of SEQ ID NO: 902. In one embodiment, the anti-GITR antibody molecule includes a heavy chain including the amino acid sequence of SEQ ID NO: 903 and a light chain including the amino acid sequence of SEQ ID NO: 904.
  • the pharmaceutical composition or dose formulation for use includes about 100 mg/mL to about 200 mg/mL of the anti-GITR antibody molecule, a buffering agent present at a concentration of about 10 mM to about 30 mM and at a pH of about 5 to about 6, a carbohydrate present at a concentration of about 200 mM to about 250 mM, and a surfactant present at a concentration of about 0.03% to about 0.05%.
  • the pharmaceutical composition or dose formulation for use includes about 150 mg/mL of the anti-GITR antibody molecule, a histidine buffer present at a concentration of about 20 mM and at a pH of about 5.5, sucrose present at a concentration of about 220 mM, and polysorbate-20 present at a concentration of about 0.03% to about 0.05%.
  • the pharmaceutical composition or dose formulation is for use in treating (e.g., inhibiting, reducing, ameliorating, or preventing) cancer.
  • the cancer cell can be, e.g., a cell from a cancer described herein, such as a solid tumor or a hematological cancer, e.g., 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 a small cell lung cancer (SCLC)), a head and neck cancer (e.g., squamous cell carcinoma of the head and neck (SCCHN)), a nasopharyngeal cancer (NPC), a skin cancer (e.g., a Merkel cell carcinoma or a melanoma (e.g., an advanced melanoma)), a skin cancer (e
  • a neuroendocrine cancer a neuroectodermal cancer, a kidney cancer (e.g., a renal cancer (e.g., a renal cell carcinoma (RCC))), an endometrial cancer (e.g., an adenocarcinoma of the endometrium), a cervical cancer, a gastric cancer (e.g., a stomach cancer, an esophageal cancer, or a cancer of the gastroesophageal junction), a liver cancer (e.g., a hepatocellular carcinoma (HCC)), a mesothelioma, a colorectal cancer (e.g., an microsatellite instability-high (MSI-H) colorectal cancer), an ovarian cancer, a breast cancer (e.g., a triple negative breast cancer (TNBC)), a urothelial cancer (e.g., a urothelial carcinoma), a bladder cancer (e.g.
  • a renal cancer
  • a transitional cell carcinoma e.g., T-cell lymphoma, B-cell lymphoma, or a non-Hogdkin lymphoma
  • a metastatic lesion of the cancer e.g., T-cell lymphoma, B-cell lymphoma, or a metastatic lesion of the cancer. Additional features or embodiments of the methods, compositions, dosage formulations, and kits described herein include one or more of the following.
  • Antibody Molecules to GITR e.g., T-cell lymphoma, B-cell lymphoma, or a non-Hogdkin lymphoma
  • the anti-GITR antibody molecule comprises at least one, two, three, four, five or six complementarity determining regions (CDRs) (or collectively all of the CDRs) from a heavy and light chain variable region comprising an amino acid sequence shown in Table 1 (e.g., from the heavy and light chain variable region sequences of MAB7 disclosed in Table 1), or encoded by a nucleotide sequence shown in Table 1.
  • CDRs are according to the Kabat definition (e.g., as set out in Table 1).
  • the CDRs are according to the Chothia definition (e.g., as set out in Table 1).
  • one or more of the CDRs (or collectively all of the CDRs) have one, two, three, four, five, six or more changes, e.g., amino acid substitutions (e.g., conservative amino acid substitutions) or deletions, relative to an amino acid sequence shown in Table 1, or encoded by a nucleotide sequence shown in Table 1.
  • the anti-GITR antibody molecule comprises a heavy chain variable region (VH) comprising a VHCDR1 amino acid sequence of SEQ ID NO: 909, a VHCDR2 amino acid sequence of SEQ ID NO: 911, and a VHCDR3 amino acid sequence of SEQ ID NO: 913; and a light chain variable region (VL) comprising a VLCDR1 amino acid sequence of SEQ ID NO: 914, a VLCDR2 amino acid sequence of SEQ ID NO: 916, and a VLCDR3 amino acid sequence of SEQ ID NO: 918, each disclosed in Table 1.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-GITR antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 901, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 901.
  • the anti-GITR antibody molecule comprises a VL comprising the amino acid sequence of SEQ ID NO: 902, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 902.
  • the anti-GITR antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 901 and a VL comprising the amino acid sequence of SEQ ID NO: 902.
  • the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 905, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 905. In one embodiment, the antibody molecule comprises a VL encoded by the nucleotide sequence of SEQ ID NO: 906, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 906. In one embodiment, the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 905 and a VL encoded by the nucleotide sequence of SEQ ID NO: 906.
  • the anti-GITR antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 903, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 903. In one embodiment, the anti-GITR antibody molecule comprises a light chain comprising the amino acid sequence of SEQ ID NO: 904, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 904. In one embodiment, the anti-GITR antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 903 and a light chain comprising the amino acid sequence of SEQ ID NO: 904.
  • the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 907, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 907. In one embodiment, the antibody molecule comprises a light chain encoded by the nucleotide sequence of SEQ ID NO: 908, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 908. In one embodiment, the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 907 and a light chain encoded by the nucleotide sequence of SEQ ID NO: 908.
  • the antibody molecules described herein can be made by vectors, host cells, and methods described in WO 2016/057846, incorporated by reference in its entirety. Table 1. Amino acid and nucleotide sequences of exemplary anti-GITR antibody molecule
  • the anti-GITR antibody molecule is BMS-986156 (Bristol-Myers Squibb), also known as BMS 986156 or BMS986156.
  • BMS-986156 and other anti-GITR antibodies are disclosed, e.g., in US 9,228,016 and WO 2016/196792, incorporated by reference in their entirety.
  • the anti-GITR antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of BMS-986156, e.g., as disclosed in Table 2. Table 2. Amino acid sequence of other exemplary anti-GITR antibody molecules.
  • the anti-GITR antibody molecule is MK-4166 or MK-1248 (Merck).
  • MK- 4166, MK-1248, and other anti-GITR antibodies are disclosed, e.g., in US 8,709,424, WO 2011/028683, WO 2015/026684, and Mahne et al. Cancer Res.2017; 77(5):1108-1118, incorporated by reference in their entirety.
  • the anti-GITR antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of MK-4166 or MK-1248.
  • the anti-GITR antibody molecule is TRX518 (Leap Therapeutics).
  • TRX518 and other anti-GITR antibodies are disclosed, e.g., in US 7,812,135, US 8,388,967, US 9,028,823, WO 2006/105021, and Ponte J et al. (2010) Clinical Immunology; 135:S96, incorporated by reference in their entirety.
  • the anti-GITR antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of TRX518.
  • the anti-GITR antibody molecule is INCAGN1876 (Incyte/Agenus).
  • INCAGN1876 and other anti-GITR antibodies are disclosed, e.g., in US 2015/0368349 and WO
  • the anti-GITR antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of INCAGN1876.
  • the anti-GITR antibody molecule is AMG 228 (Amgen).
  • AMG 228 and other anti-GITR antibodies are disclosed, e.g., in US 9,464,139 and WO 2015/031667, incorporated by reference in their entirety.
  • the anti-GITR antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of AMG 228.
  • the anti-GITR antibody molecule is INBRX-110 (Inhibrx).
  • INBRX-110 and other anti-GITR antibodies are disclosed, e.g., in US 2017/0022284 and WO 2017/015623, incorporated by reference in their entirety.
  • the GITR agonist comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of INBRX-110.
  • the GITR agonist (e.g., a fusion protein) is MEDI 1873 (MedImmune), also known as MEDI1873.
  • MEDI 1873 and other GITR agonists are disclosed, e.g., in US 2017/0073386, WO 2017/025610, and Ross et al. Cancer Res 2016; 76(14 Suppl): Abstract nr 561, incorporated by reference in their entirety.
  • the GITR agonist comprises one or more of an IgG Fc domain, a functional multimerization domain, and a receptor binding domain of a glucocorticoid-induced TNF receptor ligand (GITRL) of MEDI 1873.
  • GITRL glucocorticoid-induced TNF receptor ligand
  • GITR agonists include those described, e.g., in WO 2016/054638, incorporated by reference in its entirety.
  • the anti-GITR antibody is an antibody that competes for binding with, and/or binds to the same epitope on GITR as, one of the anti-GITR antibodies described herein.
  • An anti-GITR antibody molecule or a composition described herein can be formulated into a formulation (e.g., a dose formulation or dosage form) suitable for administration (e.g., intravenous administration) to a subject as described herein.
  • a formulation e.g., a dose formulation or dosage form
  • the formulation described herein can be a liquid formulation, a lyophilized formulation, or a reconstituted formulation.
  • the formulation is a liquid formulation.
  • the formulation e.g., liquid formulation
  • the formulation comprises an anti-GITR antibody molecule (e.g., an anti-GITR antibody molecule described herein) and a buffering agent.
  • the formulation (e.g., liquid formulation) comprises an anti-GITR antibody molecule present at a concentration of 25 mg/mL to 250 mg/mL, e.g., 50 mg/mL to 200 mg/mL, 60 mg/mL to 180 mg/mL, 70 mg/mL to 150 mg/mL, 80 mg/mL to 120 mg/mL, 90 mg/mL to 110 mg/mL, 50 mg/mL to 150 mg/mL, 50 mg/mL to 100 mg/mL, 150 mg/mL to 200 mg/mL, or 100 mg/mL to 200 mg/mL, e.g., 50 mg/mL, 60 mg/mL, 70 mg/mL, 80 mg/mL, 90 mg/mL, 100 mg/mL, 110 mg/mL, 120 mg/mL, 130 mg/mL, 140 mg/mL, 150 mg/mL, 160 mg/mL, 170 mg/mL, 180 mg/mL, 190 mg/mL
  • the formulation (e.g., liquid formulation) comprises a buffering agent comprising histidine (e.g., a histidine buffer).
  • the buffering agent e.g., histidine buffer
  • the buffering agent is present at a concentration of 1 mM to 100 mM, e.g., 2 mM to 50 mM, 5 mM to 40 mM, 10 mM to 30 mM, 15 to 25 mM, 5 mM to 40 mM, 5 mM to 30 mM, 5 mM to 20 mM, 5 mM to 10 mM, 40 mM to 50 mM, 30 mM to 50 mM, 20 mM to 50 mM, 10 mM to 50 mM, or 5 mM to 50 mM, e.g., 2 mM, 5 mM, 10 mM, 15 mM, 20 mM, 25 mM, 30 mM, 35 mM,
  • the buffering agent e.g., histidine buffer
  • the buffering agent is present at a concentration of 15 mM to 25 mM, e.g., 20 mM.
  • the buffering agent e.g., a histidine buffer
  • the buffering agent e.g., histidine buffer
  • the buffering agent comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5). In certain embodiments, the buffering agent comprises histidine and histidine-HCl at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., a pH of 5.5).
  • the formulation (e.g., liquid formulation) comprises an anti-GITR antibody molecule present at a concentration of 80 to 120 mg/mL, e.g., 100 mg/mL; and a buffering agent that comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5).
  • the formulation (e.g., liquid formulation) further comprises a
  • the carbohydrate is sucrose.
  • the carbohydrate (e.g., sucrose) is present at a concentration of 50 mM to 500 mM, e.g., 100 mM to 400 mM, 150 mM to 300 mM, 180 mM to 250 mM, 200 mM to 240 mM, 210 mM to 230 mM, 100 mM to 300 mM, 100 mM to 250 mM, 100 mM to 200 mM, 100 mM to 150 mM, 300 mM to 400 mM, 200 mM to 400 mM, or 100 mM to 400 mM, e.g., 100 mM, 150 mM, 180 mM, 200 mM, 220 mM, 250 mM, 300 mM, 350 mM, or 400 mM.
  • the formulation comprises a carbohydrate or sucrose present at a concentration of 200 mM to 250 mM, e.g., 220 mM. In certain embodiments, the formulation comprises sucrose present at a concentration of 200 mM to 250 mM, e.g., 220 mM.
  • the formulation (e.g., liquid formulation) comprises an anti-GITR antibody molecule present at a concentration of 80 to 120 mg/mL, e.g., 100 mg/mL; a buffering agent that comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5); and a carbohydrate or sucrose present at a concentration of 200 mM to 250 mM, e.g., 220 mM.
  • the formulation (e.g., liquid formulation) further comprises a surfactant.
  • the surfactant is polysorbate 20.
  • the surfactant or polysorbate 20) is present at a concentration of 0.005 % to 0.1% (w/w), e.g., 0.01% to 0.08%, 0.02% to 0.06%, 0.03% to 0.05%, 0.01% to 0.06%, 0.01% to 0.05%, 0.01% to 0.03%, 0.06% to 0.08%, 0.04% to 0.08%, or 0.02% to 0.08% (w/w), e.g., 0.01%, 0.02%, 0.03%, 0.04%, 0.05%, 0.06%, 0.07%, 0.08%, 0.09%, or 0.1% (w/w).
  • the formulation comprises a surfactant or polysorbate 20 present at a concentration of 0.03% to 0.05%, e.g., 0.04% (w/w). In some embodiments, the formulation comprises polysorbate 20 present at a concentration of 0.03% to 0.05%, e.g., 0.04% (w/w).
  • the formulation (e.g., liquid formulation, e.g., for intravenous infusion) comprises an anti-GITR antibody molecule present at a concentration of 100 to 200 mg/mL, e.g., 150 mg/mL; a buffering agent that comprises a histidine buffer (e.g., histidine/histidine-HCL) at a concentration of 20 mM) and has a pH of 5.5; a carbohydrate or sucrose present at a concentration of 220 mM; and a surfactant or polysorbate 20 present at a concentration of 0.04% (w/w).
  • a histidine buffer e.g., histidine/histidine-HCL
  • a surfactant or polysorbate 20 present at a concentration of 0.04% (w/w).
  • the formulation (e.g., liquid formulation, e.g., for intravenous infusion) comprises an anti-GITR antibody molecule present at a concentration of 150 mg/mL; a buffering agent that comprises a histidine buffer (e.g., histidine/histidine-HCL) at a concentration of 20 mM) and has a pH of 5.5; a carbohydrate or sucrose present at a concentration of 220 mM; and a surfactant or polysorbate 20 present at a concentration of 0.04% (w/w).
  • a histidine buffer e.g., histidine/histidine-HCL
  • a surfactant or polysorbate 20 present at a concentration of 0.04% (w/w).
  • the formulation (e.g., liquid formulation) comprises an anti-GITR antibody molecule present at a concentration of 80 to 120 mg/mL, e.g., 100 mg/mL; a buffering agent that comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5); a carbohydrate or sucrose present at a concentration of 200 mM to 250 mM, e.g., 220 mM; and a surfactant or polysorbate 20 present at a concentration of 0.03% to 0.05%, e.g., 0.04% (w/w).
  • a buffering agent that comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5)
  • a carbohydrate or sucrose present at a concentration of 200 m
  • the formulation (e.g., liquid formulation) comprises an anti-GITR antibody molecule present at a concentration of 100 mg/mL; a buffering agent that comprises a histidine buffer (e.g., histidine/histidine-HCL) at a concentration of 20 mM) and has a pH of 5.5; a carbohydrate or sucrose present at a concentration of 220 mM; and a surfactant or polysorbate 20 present at a
  • the liquid formulation is prepared by diluting a formulation comprising an anti-GITR antibody molecule described herein.
  • a drug substance formulation can be diluted with a solution comprising one or more excipients (e.g., concentrated excipients).
  • the solution comprises one, two, or all of histidine, sucrose, or polysorbate 20.
  • the solution comprises the same excipient(s) as the drug substance formulation.
  • excipients include, but are not limited to, an amino acid (e.g., histidine), a carbohydrate (e.g., sucrose), or a surfactant (e.g., polysorbate 20).
  • the liquid formulation is not a reconstituted lyophilized formulation. In other embodiments, the liquid formulation is a reconstituted lyophilized formulation. In some embodiments, the formulation is stored as a liquid. In other embodiments, the formulation is prepared as a liquid and then is dried, e.g., by lyophilization or spray-drying, prior to storage.
  • 0.5 mL to 10 mL e.g., 0.5 mL to 8 mL, 1 mL to 6 mL, or 2 mL to 5 mL, e.g., 1 mL, 1.2 mL, 1.5 mL, 2 mL, 3 mL, 4 mL, 4.5 mL, or 5 mL
  • 0.5 mL to 10 mL e.g., 0.5 mL to 8 mL, 1 mL to 6 mL, or 2 mL to 5 mL, e.g., 1 mL, 1.2 mL, 1.5 mL, 2 mL, 3 mL, 4 mL, 4.5 mL, or 5 mL
  • container e.g., vial
  • the liquid formulation is filled into a container (e.g., vial) such that an extractable volume of at least 1 mL (e.g., at least 1.2 mL, at least 1.5 mL, at least 2 mL, at least 3 mL, at least 4 mL, or at least 5 mL) of the liquid formulation can be withdrawn per container (e.g., vial).
  • the liquid formulation is extracted from the container (e.g., vial) without diluting at a clinical site.
  • the liquid formulation is diluted from a drug substance formulation and extracted from the container (e.g., vial) at a clinical site.
  • the formulation e.g., liquid formulation
  • the formulation is injected to an infusion bag, e.g., within 1 hour (e.g., within 45 minutes, 30 minutes, or 15 minutes) before the infusion starts to the patient.
  • a formulation described herein can be stored in a container.
  • the container used for any of the formulations described herein can include, e.g., a vial, and optionally, a stopper, a cap, or both.
  • the vial is a glass vial, e.g., a 6R white glass vial.
  • the stopper is a rubber stopper, e.g., a grey rubber stopper.
  • the cap is a flip-off cap, e.g., an aluminum flip-off cap.
  • the container comprises a 6R white glass vial, a grey rubber stopper, and an aluminum flip-off cap.
  • the container e.g., vial
  • the container is for a single-use container.
  • 25 mg/mL to 250 mg/mL e.g., 50 mg/mL to 200 mg/mL, 60 mg/mL to 180 mg/mL, 70 mg/mL to 150 mg/mL, 80 mg/mL to 120 mg/mL, 90 mg/mL to 110 mg/mL, 50 mg/mL to 150 mg/mL, 50 mg/mL to 100 mg/mL, 150 mg/mL to 200 mg/mL, or 100 mg/mL to 200 mg/mL, e.g., 50 mg/mL, 60 mg/mL, 70 mg/mL, 80 mg/mL, 90 mg/mL, 100 mg/mL, 110 mg/mL, 120 mg/mL, 130 mg/mL, 140 mg/mL, 150 mg/mL, 160 mg/mL, 170 mg/mL, 180 mg/mL, 190 mg/mL,
  • kits that include the anti-GITR antibody molecules, compositions, or formulations described herein, and instructions for use, e.g., in accordance with dosage regimens described herein.
  • the anti-GITR antibody molecules described herein can inhibit, reduce, or neutralize one or more activities of GITR, resulting in blockade or reduction of an immune checkpoint.
  • the anti-GITR antibody molecules described herein can be used to treat or prevent disorders (e.g., cancer), where enhancing an immune response in a subject is desired.
  • a method of modulating an immune response in a subject comprises administering to the subject an anti-GITR antibody molecule described herein in accordance with a dosage regimen described herein, alone or in combination with one or more therapeutic agents, procedures, or modalities, 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.
  • a method of treating e.g., one or more of reducing, inhibiting, or delaying progression
  • the method comprises administering to the subject an anti-GITR antibody molecule described herein in accordance with a dosage regimen described herein, alone or in combination with one or more therapeutic agents, procedures, or modalities.
  • the cancer treated with the anti-GITR antibody molecule includes but is not limited to, a solid tumor, a hematological cancer (e.g., leukemia, lymphoma, myeloma, e.g., multiple myeloma), and a metastatic lesion.
  • the cancer is a solid tumor.
  • solid tumors include malignancies, e.g., sarcomas and carcinomas, e.g., adenocarcinomas of the various organ systems, such as those affecting the lung, breast, ovarian, lymphoid, gastrointestinal (e.g., colon), anal, genitals and genitourinary tract (e.g., renal, urothelial, bladder cells, prostate), pharynx, CNS (e.g., brain, neural or glial cells), head and neck, skin (e.g., melanoma), and pancreas, as well as adenocarcinomas which include malignancies such as colon cancers, rectal cancer, renal cancer (e.g., renal-cell carcinoma (clear cell or non-clear cell renal cell carcinoma), liver cancer, lung cancer (e.g., non-small cell lung cancer (squamous or non-squamous non-small cell lung cancer)), cancer of the small intestine and cancer of the e
  • 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 a small cell lung cancer (SCLC)), a head and neck cancer (e.g., squamous cell carcinoma of the head and neck (SCCHN)), a nasopharyngeal cancer (NPC), a skin cancer (e.g., a Merkel cell carcinoma or a melanoma (e.g., an advanced melanoma)), a neuroendocrine cancer, a neuroectodermal cancer, a kidney cancer (e.g., a renal cancer (e.g., a renal cell carcinoma (RCC))), an endometrial cancer (e.g., an adenocarcinoma of the endometrium), a cervical cancer,
  • a transitional cell carcinoma a lymphoma (e.g., T-cell lymphoma, B-cell lymphoma (e.g., a diffuse large B cell lymphoma), or a non- Hogdkin lymphoma), or a metastatic lesion of the cancer.
  • a lymphoma e.g., T-cell lymphoma, B-cell lymphoma (e.g., a diffuse large B cell lymphoma), or a non- Hogdkin lymphoma
  • a metastatic lesion of the cancer e.g., T-cell lymphoma, B-cell lymphoma (e.g., a diffuse large B cell lymphoma), or a non- Hogdkin lymphoma)
  • the cancer is a solid tumor.
  • the cancer is a lung cancer, e.g., a NSCLC, e.g., a NSCLC with squamous and/or non-squamous histology, or a NSCLC adenocarcinoma, or a SCLC.
  • the cancer is a head and neck cancer, e.g., SCCHN.
  • the cancer is a NPC.
  • the cancer is a skin cancer, e.g., a Merkel cell carcinoma or a melanoma, e.g., an advanced melanoma.
  • the cancer is a neuroendocrine cancer.
  • the cancer is a neuroectodermal cancer.
  • the cancer is a kidney cancer, e.g., a renal cancer, e.g., a RCC.
  • the cancer is an endometrial cancer, e.g., an adenocarcinoma of the endometrium.
  • the cancer is a cervical cancer.
  • the cancer is a gastric cancer, e.g., a stomach cancer, an esophageal cancer, or a cancer of the gastroesophageal junction.
  • the cancer is a liver cancer, e.g., a HCC.
  • the cancer is a mesothelioma.
  • the cancer is a colorectal cancer, e.g., an MSI-H colorectal cancer.
  • the cancer is an ovarian cancer.
  • the cancer is a breast cancer, e.g., a TNBC.
  • the cancer is a urothelial cancer, e.g., a urothelial carcinoma.
  • the cancer is a bladder cancer, e.g. a transitional cell carcinoma.
  • the cancer is a hematological cancer.
  • the cancer is a lymphoma (e.g., T-cell lymphoma, B-cell lymphoma, or a non-Hogdkin lymphoma).
  • the cancer is a metastatic cancer, an advanced cancer, a relapsed cancer, a refractory cancer, a recurrent cancer, or an MSI-H cancer. In some embodiments, the cancer is a FoxP3- expressing cancer.
  • the cancer microenvironment has an elevated level of GITR expression. In one embodiment, the cancer microenvironment has an elevated level of PD-L1 expression. Alternatively, or in combination, the cancer microenvironment can have increased IFN ⁇ and/or CD8 expression.
  • the subject has, or is identified as having, a tumor that has one or more of high PD-L1 level or expression, or as being Tumor Infiltrating Lymphocyte (TIL)+ (e.g., as having an increased number of TILs), or both.
  • TIL Tumor Infiltrating Lymphocyte
  • the subject has, or is identified as having, a tumor that has high PD-L1 level or expression and that is TIL+.
  • the methods described herein further include identifying a subject based on having a tumor that has one or more of high PD-L1 level or expression, or as being TIL+, or both.
  • the methods described herein further include identifying a subject based on having a tumor that has high PD-L1 level or expression and as being TIL+.
  • tumors that are TIL+ are positive for CD8 and IFN ⁇ .
  • the subject has, or is identified as having, a high percentage of cells that are positive for one, two or more of PD-L1, CD8, and/or IFN ⁇ .
  • the subject has or is identified as having a high percentage of cells that are positive for all of PD-L1, CD8, and IFN ⁇ .
  • the methods described herein further include identifying a subject based on having a high percentage of cells that are positive for one, two or more of PD-L1, CD8, and/or IFN ⁇ . In certain embodiments, the methods described herein further include identifying a subject based on having a high percentage of cells that are positive for all of PD-L1, CD8, and IFN ⁇ .
  • the subject has, or is identified as having, one, two or more of PD-L1, CD8, and/or IFN ⁇ , and one or more of a lung cancer, e.g., squamous cell lung cancer or lung
  • adenocarcinoma e.g., an NSCLC
  • a head and neck cancer e.g., a cervical cancer
  • a gastric cancer e.g., a stomach cancer, an esophageal cancer, or a cancer of the gastroesophageal junction
  • a skin cancer e.g., a Merkel cell carcinoma or a melanoma
  • a breast cancer e.g., an TNBC
  • NPC nasopharyngeal cancer
  • the methods described herein further describe identifying a subject based on having one, two or more of PD-L1, CD8, and/or IFN ⁇ , and one or more of a lung cancer, e.g., squamous cell lung cancer or lung adenocarcinoma (e.g., an NSCLC); a head and neck cancer; a cervical cancer; a gastric cancer (e.g., a stomach cancer, an esophageal cancer, or a cancer of the gastroesophageal junction); a skin cancer (e.g., a Merkel cell carcinoma or a melanoma), a breast cancer (e.g., an TNBC), and/or a nasopharyngeal cancer (NPC).
  • a lung cancer e.g., squamous cell lung cancer or lung adenocarcinoma (e.g., an NSCLC); a head and neck cancer; a cervical cancer; a gastric cancer (e.g.,
  • compositions, and formulations disclosed herein are useful for treating metastatic lesions associated with the aforementioned cancers.
  • the disclosure provides a method of treating an infectious disease (e.g., an infectious disease described herein) in a subject, comprising administering to the subject an anti-GITR antibody molecule described herein in accordance with a dosage regimen described herein.
  • an infectious disease e.g., an infectious disease described herein
  • 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) an anti-GITR antibody molecule described herein, in accordance with a dosage regimen 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 anti-GITR antibody molecule 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 intravenously at a flat dose described herein.
  • anti-GITR antibody molecules described herein can be used in combination with other therapeutic agents, procedures or modalities.
  • the methods described herein include administering to the subject a combination comprising an anti-GITR antibody molecule described herein, in combination with a therapeutic agent, procedure, or modality, in an amount effective to treat or prevent a disorder.
  • the anti-GITR antibody molecule is administered or used in accordance with a dosage regimen described herein.
  • the antibody molecule is administered or used as a composition or formulation described herein.
  • the anti-GITR antibody molecule and the therapeutic agent, procedure, or modality can be administered or used simultaneously or sequentially in any order. Any combination and sequence of the anti-GITR antibody molecule and the therapeutic agent, procedure, or modality (e.g., as described herein) can be used.
  • the antibody molecule and/or the therapeutic agent, procedure or modality can be administered or used during periods of active disorder, or during a period of remission or less active disease.
  • the antibody molecule can be administered before, concurrently with, or after the treatment with the therapeutic agent, procedure or modality.
  • the anti-GITR antibody molecule described herein is administered in combination with one or more of other antibody molecules, chemotherapy, other anti-cancer therapy (e.g., targeted anti-cancer therapies, gene therapy, viral therapy, RNA therapy bone marrow transplantation, nanotherapy, or oncolytic drugs), cytotoxic agents, immune-based therapies (e.g., cytokines or cell-based immune therapies), surgical procedures (e.g., lumpectomy or mastectomy) or radiation procedures, or a combination of any of the foregoing.
  • the additional therapy may be in the form of adjuvant or neoadjuvant therapy.
  • the additional therapy is an enzymatic inhibitor (e.g., a small molecule enzymatic inhibitor) or a metastatic inhibitor.
  • exemplary cytotoxic agents that can be administered in combination with the anti-GITR antibody molecule include antimicrotubule agents, topoisomerase inhibitors, anti-metabolites, mitotic inhibitors, alkylating agents, anthracyclines, vinca alkaloids, intercalating agents, agents capable of interfering with a signal transduction pathway, agents that promote apoptosis, proteosome inhibitors, and radiation (e.g., local or whole body irradiation (e.g., gamma irradiation).
  • the additional therapy is surgery or radiation, or a combination thereof.
  • the additional therapy is a therapy targeting one or more of PI3K/AKT/mTOR pathway, an HSP90 inhibitor, or a tubulin inhibitor.
  • the anti-GITR antibody described herein can be administered or used in combination with, one or more of: an immunomodulator (e.g., an activator of a costimulatory molecule or an inhibitor of an inhibitory molecule, e.g., an immune checkpoint molecule); a vaccine, e.g., a therapeutic cancer vaccine; or other forms of cellular immunotherapy.
  • an immunomodulator e.g., an activator of a costimulatory molecule or an inhibitor of an inhibitory molecule, e.g., an immune checkpoint molecule
  • a vaccine e.g., a therapeutic cancer vaccine
  • the anti-GITR molecule described herein is administered or used in combination with a modulator of a costimulatory molecule or an inhibitory molecule, e.g., a co-inhibitory ligand or receptor.
  • the anti-GITR antibody molecule described herein is administered or used in combination with a modulator, e.g., agonist, of a costimulatory molecule.
  • a modulator e.g., agonist
  • 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, 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 anti-GITR antibody molecule described herein is administered or used in combination with a GITR agonist, e.g., an anti-GITR antibody molecule.
  • the anti-GITR antibody molecule described herein is administered or used in combination with an inhibitor of an inhibitory (or immune checkpoint) molecule chosen from PD-1, PD- L1, PD-L2, CTLA-4, TIM-3, LAG-3, CEACAM (e.g., CEACAM-1, CEACAM-3, and/or CEACAM-5), VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and/or TGF beta.
  • the inhibitor is a soluble ligand (e.g., a CTLA-4-Ig), or an antibody or antibody fragment that binds to PD-1, LAG-3, TIM- 3, PD-L1, PD-L2, or CTLA-4.
  • the anti-GITR antibody molecule described herein is administered or used in combination with a PD-1 inhibitor, e.g., an anti-PD-1 antibody molecule.
  • the anti-GITR antibody molecule described herein is administered or used in combination with a LAG-3 inhibitor, e.g., an anti-LAG-3 antibody molecule.
  • the anti-GITR antibody molecule described herein is administered or used in combination with a TIM-3 inhibitor, e.g., an anti- TIM-3 antibody molecule.
  • the anti-GITR antibody molecule described herein is administered or used in combination with a PD-L1 inhibitor, e.g., an anti-PD-L1 antibody molecule.
  • the anti-GITR antibody molecule described herein is administered or used in combination with a PD-1 inhibitor (e.g., an anti-PD-1 antibody molecule) and a LAG-3 inhibitor (e.g., an anti-LAG-3 antibody molecule).
  • a PD-1 inhibitor e.g., an anti-PD-1 antibody molecule
  • a PD-L1 inhibitor e.g., an anti-PD-L1 antibody molecule
  • the anti-GITR antibody molecule described herein is administered or used in combination with a LAG-3 inhibitor (e.g., an anti-LAG-3 antibody molecule) and a PD-L1 inhibitor (e.g., an anti-PD- L1 antibody molecule).
  • a LAG-3 inhibitor e.g., an anti-LAG-3 antibody molecule
  • a PD-L1 inhibitor e.g., an anti-PD- L1 antibody molecule
  • the anti-GITR antibody molecule described herein is administered or used in combination with a PD-1 inhibitor (e.g., an anti-PD-1 antibody molecule) and a TIM-3 inhibitor (e.g., an anti-TIM-3 antibody molecule).
  • the anti-GITR antibody molecule described herein is administered or used in combination with a LAG-3 inhibitor (e.g., an anti- LAG-3 antibody molecule) and a TIM-3 inhibitor (e.g., an anti-TIM-3 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
  • a PD-L1 inhibitor e.g., an anti-PD- L1 antibody molecule
  • the anti-GITR antibody molecule described herein is administered or used in combination with a CEACAM inhibitor (e.g., CEACAM-1, CEACAM-3, and/or CEACAM-5 inhibitor), e.g., an anti- CEACAM antibody molecule.
  • a CEACAM inhibitor e.g., CEACAM-1, CEACAM-3, and/or CEACAM-5 inhibitor
  • the anti-GITR antibody molecule is administered or used in combination with a CEACAM-1 inhibitor, e.g., an anti-CEACAM-1 antibody molecule.
  • the anti-GITR antibody molecule is administered or used in combination with a CEACAM-3 inhibitor, e.g., an anti-CEACAM-3 antibody molecule.
  • the anti-PD-1 antibody molecule is administered or used in combination with a CEACAM-5 inhibitor, e.g., an anti-CEACAM-5 antibody molecule.
  • the combination of antibody molecules disclosed herein can be administered separately, e.g., as separate antibody molecules, or linked, e.g., as a bispecific or trispecific antibody molecule.
  • a bispecific antibody that includes an anti-GITR antibody molecule and an anti-PD-1, anti- CEACAM (e.g., anti-CEACAM-1, CEACAM-3, and/or anti-CEACAM-5), anti-PD-L1, anti-TIM-3, or anti-LAG-3 antibody molecule, is administered.
  • the combination of antibodies disclosed herein is used to treat a cancer, e.g., a cancer as described herein (e.g., a solid tumor or a hematologic malignancy).
  • the anti-GITR antibody molecule is administered or used in combination with a cytokine.
  • the cytokine can be administered as a fusion molecule to the anti-GITR antibody molecule, or as separate compositions.
  • the anti-GITR antibody molecule is administered or used in combination with one, two, three or more cytokines, e.g., as a fusion molecule or as separate compositions.
  • the cytokine is an interleukin (IL) chosen from one, two, three or more of IL-1, IL-2, IL-12, IL-15 or IL-21.
  • a bispecific antibody molecule has a first binding specificity to a first target (e.g., to GITR), a second binding specificity to a second target (e.g., PD-1, LAG-3, TIM-3, or PD-L1), and is optionally linked to an interleukin (e.g., IL-12) domain e.g., full length IL-12 or a portion thereof.
  • a first target e.g., to GITR
  • a second binding specificity to a second target e.g., PD-1, LAG-3, TIM-3, or PD-L1
  • an interleukin e.g., IL-12 domain
  • the combination of anti-GITR antibody molecule and the cytokine described herein is used to treat a cancer, e.g., a cancer as described herein (e.g., a solid tumor).
  • the anti-GITR antibody molecule is administered or used in combination with an antibody specific against an HLA C, e.g., an antibody specific to Killer-cell Immunoglobulin-like Receptors (also referred to herein as an“anti-KIR antibody”).
  • an antibody specific against an HLA C e.g., an antibody specific to Killer-cell Immunoglobulin-like Receptors (also referred to herein as an“anti-KIR antibody”).
  • the combination of anti-GITR antibody molecule and anti-KIR antibody is used to treat a cancer, e.g., a cancer as described herein (e.g., a solid tumor, e.g., an advanced solid tumor).
  • the anti-GITR antibody molecule is administered or used in combination with a vaccine, e.g., a cancer vaccine, (e.g., a dendritic cell renal carcinoma (DC-RCC) vaccine).
  • a vaccine e.g., a cancer vaccine, (e.g., a dendritic cell renal carcinoma (DC-RCC) vaccine).
  • the vaccine is peptide-based, DNA-based, RNA-based, or antigen-based, or a combination thereof.
  • the vaccine comprises one or more peptides, nucleic acids (e.g., DNA or RNA), antigens, or a combination thereof.
  • the combination of anti-GITR antibody molecule and the DC-RCC vaccine is used to treat a cancer, e.g., a cancer as described herein (e.g., a RCC).
  • the anti-GITR antibody molecule is administered or used in combination with an adjuvant.
  • the anti-GITR antibody molecule is administered or used in combination with chemotherapy, and/or immunotherapy.
  • the anti-GITR antibody molecule can be used to treat a myeloma, alone or in combination with one or more of: chemotherapy or other anti-cancer agents (e.g., thalidomide analogs, e.g., lenalidomide), an anti-PD-1 antibody molecule, tumor antigen- pulsed dendritic cells, fusions (e.g., electrofusions) of tumor cells and dendritic cells, or vaccination with immunoglobulin idiotype produced by malignant plasma cells.
  • the anti-GITR antibody molecule is administered or used in combination with an anti-PD-1 antibody molecule to treat a myeloma, e.g., a multiple myeloma.
  • the anti-GITR antibody molecule is administered or used in combination with chemotherapy to treat a lung cancer, e.g., non-small cell lung cancer.
  • a lung cancer e.g., non-small cell lung cancer.
  • the anti-GITR antibody molecule is administered or used with standard lung, e.g., NSCLC, chemotherapy, e.g., platinum doublet therapy, to treat lung cancer.
  • the anti-GITR antibody molecule is administered or used in combination with an indoleamine-pyrrole 2,3-dioxygenase (IDO) inhibitor (e.g., (4E)-4-[(3-chloro-4-fluoroanilino)-nitrosomethylidene]-1,2,5-oxadiazol-3-amine (also known as INCB24360), indoximod (1-methyl-D-tryptophan), ⁇ -cyclohexyl-5H-Imidazo[5,1-a]isoindole- 5-ethanol (also known as NLG919), etc.) in a subject with a metastatic cancer, an advanced cancer, a relapsed cancer, a refractory cancer, a recurrent cancer, or an MSI-H cancer.
  • IDO indoleamine-pyrrole 2,3-dioxygenase
  • the anti-GITR antibody molecule is administered or used in combination with one or more of: an immune-based strategy (e.g., interleukin-2 or interferon- ⁇ ), a targeting agent (e.g., a VEGF inhibitor such as a monoclonal antibody to VEGF); a VEGF tyrosine kinase inhibitor such as sunitinib, sorafenib, axitinib and pazopanib; an RNAi inhibitor; or an inhibitor of a downstream mediator of VEGF signaling, e.g., an inhibitor of the mammalian target of rapamycin (mTOR), e.g., everolimus and temsirolimus.
  • an immune-based strategy e.g., interleukin-2 or interferon- ⁇
  • a targeting agent e.g., a VEGF inhibitor such as a monoclonal antibody to VEGF
  • a renal cancer e.g., renal cell carcinoma (RCC) (e.g., clear cell renal cell carcinoma (CCRCC) or a non- clear cell renal cell carcinoma (nccRCC) or metastatic RCC, or a liver cancer (e.g., a hepatocellular carcinoma).
  • RCC renal cell carcinoma
  • CCRCC clear cell renal cell carcinoma
  • nccRCC non- clear cell renal cell carcinoma
  • metastatic RCC e.g., a hepatocellular carcinoma
  • the anti-GITR antibody molecule is administered or used in combination with a MEK inhibitor (e.g., a MEK inhibitor as described herein).
  • a MEK inhibitor e.g., a MEK inhibitor as described herein.
  • the combination of the anti-GITR antibody molecule and the MEK inhibitor is used to treat a cancer (e.g., a cancer described herein).
  • the cancer treated with the combination is chosen from a melanoma, a colorectal cancer, a non-small cell lung cancer, an ovarian cancer, a breast cancer, a prostate cancer, a pancreatic cancer, a hematological malignancy or a renal cell carcinoma.
  • the cancer includes a BRAF mutation (e.g., a BRAF V600E mutation), a BRAF wildtype, a KRAS wildtype or an activating KRAS mutation.
  • the cancer may be at an early, intermediate or late stage.
  • the anti-GITR antibody molecule is administered or used in combination with one, two or all of oxaliplatin, leucovorin, or 5-FU (e.g., a FOLFOX co-treatment).
  • combination further includes a VEGF inhibitor (e.g., a VEGF inhibitor as disclosed herein).
  • the combination of the anti-GITR antibody molecule, the FOLFOX co- treatment, and the VEGF inhibitor is used to treat a cancer (e.g., a cancer described herein).
  • the cancer treated with the combination is chosen from a melanoma, a colorectal cancer, a non-small cell lung cancer, an ovarian cancer, a breast cancer, a prostate cancer, a pancreatic cancer, a hematological malignancy or a renal cell carcinoma.
  • the cancer may be at an early, intermediate or late stage.
  • the anti-GITR antibody molecule is administered or used with a tyrosine kinase inhibitor (e.g., axitinib) to treat renal cell carcinoma and other solid tumors.
  • a tyrosine kinase inhibitor e.g., axitinib
  • the anti-GITR antibody molecule is administered or used with a 4-1BB receptor targeting agent (e.g., an antibody that stimulates signaling through 4-1BB (CD-137), e.g., PF- 2566).
  • 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 or used in combination with a tyrosine kinase inhibitor (e.g., axitinib) and a 4-1BB receptor targeting agent.
  • the anti-GITR antibody molecule can be bound to a substance, e.g., a cytotoxic agent or moiety (e.g., a therapeutic drug; a compound emitting radiation; molecules of plant, fungal, or bacterial origin; or a biological protein (e.g., a protein toxin) or particle (e.g., a recombinant viral particle, e.g., via a viral coat protein).
  • a cytotoxic agent or moiety e.g., a therapeutic drug; a compound emitting radiation; molecules of plant, fungal, or bacterial origin; or a biological protein (e.g., a protein toxin) or particle (e.g., a recombinant viral particle, e.g., via a viral coat protein).
  • the antibody can be coupled to a radioactive isotope such as an ⁇ -, ⁇ -, or ⁇ - emitter, or a ⁇ -and ⁇ -emitter.
  • the immunomodulator is an inhibitor of an immune checkpoint molecule.
  • the immunomodulator is an inhibitor of PD-1, PD-L1, PD-L2, CTLA-4, LAG-3, TIM-3 CEACAM (e.g., CEACAM-1, -3 and/or -5), VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and/or TGF 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 molecule that binds to the inhibitory molecule; e.g., an antibody molecule that binds to PD-1, PD-L1, PD-L2, CEACAM (e.g., CEACAM-1, -3 and/or -5), CTLA-4, LAG-3, TIM-3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and/or TGF beta, or a combination thereof.
  • a polypeptide e.g., a soluble ligand (e.g., PD-1-Ig or CTLA-4 Ig), or an antibody molecule that binds to the inhibitory molecule; e.g., an antibody molecule that binds to PD-1, PD-L1, PD-L2, CEACAM (e.g., CEA
  • the anti-GITR antibody molecule is in the form of a bispecific or multispecific antibody molecule.
  • the bispecific antibody molecule has a first binding specificity to GITR and a second binding specificity, e.g., a second binding specificity to, PD-1, PD-L1, CEACAM (e.g., CEACAM-1, -3 and/or -5), LAG-3, TIM-3, or PD-L2.
  • the bispecific antibody molecule binds to (i) PD-1 or PD-L1 (ii) and GITR.
  • the bispecific antibody molecule binds to GITR and LAG-3.
  • the bispecific antibody molecule binds to GITR and TIM-3. In another embodiment, the bispecific antibody molecule binds to GITR and CEACAM (e.g., CEACAM-1, -3 and/or -5). In another embodiment, the bispecific antibody molecule binds to GITR and CEACAM-1. In still another embodiment, the bispecific antibody molecule binds to GITR and CEACAM-3. In yet another embodiment, the bispecific antibody molecule binds to GITR and CEACAM-5.
  • the anti-GITR antibody molecule is used in combination with a bispecific or multispecific antibody molecule.
  • the bispecific antibody molecule binds to PD-1 or PD-L1.
  • the bispecific antibody molecule binds to PD-1 and PD-L2.
  • the bispecific antibody molecule binds to CEACAM (e.g., CEACAM-1, -3 and/or -5) and LAG-3.
  • the bispecific antibody molecule binds to CEACAM (e.g., CEACAM-1, -3 and/or -5) and TIM-3.
  • any combination of the aforesaid molecules can be made in a multispecific antibody molecule, e.g., a trispecific antibody that includes a first binding specificity to GITR, and a second and third binding specificities to two or more of: PD-1, PD-L1, CEACAM (e.g., CEACAM-1, -3 and/or -5), LAG-3, TIM- 3, or PD-L2.
  • a multispecific antibody molecule e.g., a trispecific antibody that includes a first binding specificity to GITR, and a second and third binding specificities to two or more of: PD-1, PD-L1, CEACAM (e.g., CEACAM-1, -3 and/or -5), LAG-3, TIM- 3, or PD-L2.
  • the immunomodulator is an inhibitor of PD-1, e.g., human PD-1.
  • the immunomodulator is an inhibitor of PD-L1, e.g., human PD-L1.
  • the inhibitor of PD-1 or PD-L1 is an antibody molecule to PD-1 or PD-L1 (e.g., an anti-PD- 1 or anti-PD-L1 antibody molecule as described herein).
  • the combination of the PD-1 or PD-L1 inhibitor with the anti-GITR antibody molecule can further include one or more additional immunomodulators, e.g., in combination with an inhibitor of LAG- 3, TIM-3, CEACAM (e.g., CEACAM-1, -3 and/or -5) or CTLA-4.
  • the inhibitor of PD-1 or PD-L1 e.g., the anti-PD-1 or PD-L1 antibody molecule
  • a LAG-3 inhibitor e.g., an anti-LAG-3 antibody molecule.
  • the inhibitor of PD-1 or PD-L1 is administered in combination with the anti-GITR antibody molecule and a TIM-3 inhibitor (e.g., an anti- TIM-3 antibody molecule).
  • a TIM-3 inhibitor e.g., an anti- TIM-3 antibody molecule.
  • the inhibitor of PD-1 or PD-L1 is administered in combination with the anti-GITR antibody molecule and a CEACAM inhibitor (e.g., CEACAM-1, -3 and/or -5 inhibitor), e.g., an anti-CEACAM antibody molecule.
  • the inhibitor of PD-1 or PD-L1 is administered in combination with the anti-GITR antibody molecule and a CEACAM-1 inhibitor (e.g., an anti-CEACAM-1 antibody molecule).
  • the inhibitor of PD-1 or PD-L1 is administered in combination with the anti-GITR antibody molecule and a CEACAM-5 inhibitor (e.g., an anti-CEACAM-5 antibody molecule).
  • the inhibitor of PD-1 or PD-L1 is administered in combination with the anti-GITR antibody molecule, a LAG-3 inhibitor (e.g., an anti- LAG-3 antibody molecule), and a TIM-3 inhibitor (e.g., an anti-TIM-3 antibody molecule).
  • immunomodulators with the anti-GITR antibody molecule and a PD-1 inhibitor e.g., one or more of PD-L2, CTLA-4, LAG-3, TIM-3, CEACAM (e.g., CEACAM-1, -3 and/or -5), VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and/or TGF beta
  • a PD-1 inhibitor e.g., one or more of PD-L2, CTLA-4, LAG-3, TIM-3, CEACAM (e.g., CEACAM-1, -3 and/or -5), VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and/or TGF beta
  • a PD-1 inhibitor e.g., one or more of PD-L2, CTLA-4, LAG-3, TIM-3, CEACAM (e.g., CEACAM-1, -3 and/or -5), VISTA, BTLA, TIGIT, LAIR1, CD160,
  • 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).
  • CEACAM e.g., CEACAM-1, -3 and/or -5
  • 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 e.g., CEACAM-1, -3 and/or -5 is an antibody molecule to CEACAM (e.g., CEACAM-1, -3 and/or -5).
  • the combination of the CEACAM (e.g., CEACAM-1, -3 and/or -5) inhibitor and the anti-GITR antibody molecule can further include one or more additional 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 combination of the LAG-3 inhibitor and the anti-GITR antibody molecule can further include one or more additional immunomodulators, e.g., in combination with an inhibitor of CEACAM (e.g., CEACAM-1, -3 and/or -5), PD-1, PD-L1, TIM-3, or CTLA-4.
  • additional immunomodulators e.g., in combination with an inhibitor of CEACAM (e.g., CEACAM-1, -3 and/or -5), PD-1, PD-L1, TIM-3, 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 combination of the TIM- 3 inhibitor and the anti-GITR antibody molecule can further include one or more additional
  • immunomodulators e.g., in combination with an inhibitor of CEACAM (e.g., CEACAM-1, -3 and/or -5), PD-1, PD-L1, LAG-3, or CTLA-4.
  • CEACAM e.g., CEACAM-1, -3 and/or -5
  • PD-1 e.g., PD-L1, LAG-3, 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 a second GITR agonist.
  • the second GITR agonist is a second antibody molecule to GITR.
  • the second anti-GITR antibody molecule and the first anti-GITR antibody molecule may be in the form of separate antibody composition, or as a bispecific antibody molecule.
  • the combination of the second GITR agonist with the first anti-GITR antibody molecule can further include one or more additional 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 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.
  • CEACAM e.g., CEACAM-1, -3 and/or -5
  • TIM-3 e.g., TIM-3, or LAG-3.
  • the second GITR agonist can be administered in combination with one or more additional activators of costimulatory molecules, 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.
  • additional activators of costimulatory molecules 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 immunomodulator is an OX40 agonist.
  • the OX40 agonist is an antibody molecule to OX40.
  • 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 combination of the OX40 agonist with the anti-GITR antibody molecule can further include one or more additional 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 e.g., TIM-3
  • LAG-3 e.g., LAG-3
  • the anti-OX40 antibody molecule is a bispecific antibody that binds to OX40 and PD-1, PD-L1, CTLA-4, CEACAM (e.g., CEACAM-1, -3 and/or -5), TIM-3, or LAG-3.
  • the OX40 agonist can be administered in combination with other costimulatory molecule, e.g., an agonist of 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 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.
  • any of the methods disclosed herein further includes evaluating or monitoring the effectiveness of a therapy (e.g., a monotherapy or a combination therapy) described herein, in a subject (e.g., a subject having a cancer, e.g., a cancer described herein).
  • the method includes acquiring a value of effectiveness to the therapy, wherein said value is indicative of the effectiveness of the therapy.
  • the value of effectiveness to the therapy comprises a measure of one, two, three, four, five, six, seven, eight, nine or more (e.g., all) of the following:
  • TIL tumor infiltrating lymphocyte
  • the parameter of a TIL phenotype comprises the level or activity of one, two, three, four or more (e.g., all) of Hematoxylin and eosin (H&E) staining for TIL counts, FOXP3, CD8, CD56, PDL1, CD68, or CD4, in the subject, e.g., in a sample from the subject (e.g., a tumor sample).
  • H&E Hematoxylin and eosin
  • the parameter of a myeloid cell population comprises the level or activity of one or both of FOXP3, CD8, CD56, PDL1, CD68, or CD4, in the subject, e.g., in a sample from the subject (e.g., a tumor sample).
  • the parameter of a surface expression marker comprises the level or activity of one, two, three or more (e.g., all) of GITR (e.g., soluble GITR (sGITR)), PD-1, PD-L1, or LAG-3, in the subject, e.g., in a sample from the subject (e.g., a tumor sample).
  • the level of GITR e.g., sGITR
  • PD-1, PD-L1, or LAG-3 is determined by immunohistochemistry (IHC).
  • the level of soluble sGITR is determined.
  • the parameter of a biomarker of an immunologic response comprises the level or sequence of one or more nucleic acid-based markers, in the subject, e.g., in a sample from the subject (e.g., a tumor sample).
  • the parameter of systemic cytokine modulation comprises the level or activity of one, two, three, four, five, six, seven, eight, or more (e.g., all) of GM-CSF, IFN- ⁇ , TNF- ⁇ , IL- 1, IL-2, IL-5, IL-6, IL-10, IL-12, IL-17, IL-18, or VEGF, in the subject, e.g., in a sample from the subject (e.g., a blood sample, e.g., a plasma sample).
  • a sample from the subject e.g., a blood sample, e.g., a plasma sample.
  • the parameter of cfDNA comprises the sequence or level of one or more circulating tumor DNA (cfDNA) molecules, in the subject, e.g., in a sample from the subject (e.g., a blood sample, e.g., a plasma sample).
  • a sample from the subject e.g., a blood sample, e.g., a plasma sample.
  • the parameter of systemic immune-modulation comprises phenotypic characterization of an activated immune cell, e.g., a CD3-expressing cell, a CD8-expressing cell, a HLA- DR-expressing cell, a GITR-expressing cell, or a combination thereof, in the subject, e.g., in a sample from the subject (e.g., a blood sample, e.g., a PBMC sample).
  • a sample from the subject e.g., a blood sample, e.g., a PBMC sample.
  • the parameter of microbiome comprises the sequence or expression level of one or more genes in the microbiome, in the subject, e.g., in a sample from the subject (e.g., a stool sample).
  • the parameter of a marker of activation in a circulating immune cell comprises the level or activity of one, two, three, four, five or more (e.g., all) of circulating GM-CSF, IFN- ⁇ , TNF- ⁇ , IL-1, IL-2, IL-5, IL-6, IL-10, IL-12, IL-17, IL-18, or VEGF expressing cells, in a sample (e.g., a blood sample, e.g., a plasma sample).
  • a sample e.g., a blood sample, e.g., a plasma sample.
  • the parameter of a circulating cytokine comprises the level or activity of GM-CSF, IFN- ⁇ , TNF- ⁇ , IL-1, IL-2, IL-5, IL-6, IL-10, IL-12, IL-17, IL-18, or VEGF, in the subject, e.g., in a sample from the subject (e.g., a blood sample, e.g., a plasma sample).
  • a sample from the subject e.g., a blood sample, e.g., a plasma sample.
  • the cancer has high Treg conent, e.g. compared to a reference Treg content level.
  • the therapy comprises a combination of an anti-GITR antibody molecule described herein and a second inhibitor of an immune checkpoint molecule, e.g., an inhibitor of PD-1 (e.g., an anti-PD-1 antibody molecule) or an inhibitor of PD-L1 (e.g., an anti-PD-L1 antibody molecule).
  • an inhibitor of PD-1 e.g., an anti-PD-1 antibody molecule
  • PD-L1 e.g., an anti-PD-L1 antibody molecule
  • the measure of one or more of (i)- (x) is obtained from a sample acquired from the subject.
  • the sample is chosen from a tumor sample, a blood sample (e.g., a plasma sample or a PBMC sample), or a stool sample.
  • the subject is evaluated prior to receiving, during, or after receiving, the therapy.
  • the measure of one or more of (i)- (x) evaluates a profile for one or more of gene expression, flow cytometry or protein expression.
  • administering to the subject an additional agent (e.g., a therapeutic agent described herein) in combination with the therapy; or
  • any of the methods disclosed herein includes 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 CGPGRLLLGTGTDARCCRVHTTRCCRDYPGEECCSEWDC
  • 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.
  • 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.
  • 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. In some embodiments, provided is 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.
  • any of the methods disclosed herein includes 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
  • 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
  • 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
  • the CDR3 of the heavy chain comprises SEQ ID NO:29 or SEQ ID NO:109
  • 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
  • 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. In some embodiments, the antibody, antibody fragment or antigen binding molecule is encoded by a polynucleotide.
  • 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 compositions described herein can be used in accordance with dosage regimens described herein.
  • 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.
  • the composition further comprises an antagonist of CTLA4. In some embodiments, the composition further comprises an antagonist of LAG3. In some embodiments, the composition further comprises an antagonist of TIM3. In some embodiments, the composition further comprises an inhibitor of PD-1/PD-L1 (e.g., B7-H1 or analogue thereof, PD-1 antibody) interaction. In certain embodiments the composition further comprises an antagonist of PD-1. In certain embodiments the composition further comprises an antagonist of PD-L1. In a further aspect, the invention further provides kits comprising an antibody or antibody fragment of the invention, as described herein. The kits described herein can be used in accordance with dosage regimens 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 described herein, in accordance with a dosage regimen 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, in accordance with a dosage regimen described herein.
  • 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, in accordance with a dosage regimen described herein.
  • 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 described herein, in accordance with a dosage regimen 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, in accordance with a dosage regimen described herein.
  • 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, in accordance with a dosage regimen described herein.
  • 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 CTLA-4. In some embodiments, the anti-GITR agonist antibody, antibody fragment, or antigen binding molecule is co-administered with an antagonist of LAG3. 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 additional agent e.g., second or third agent
  • 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) a CDK
  • P450
  • 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
  • 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).
  • an agent that enhances antigen e.g., tumor antigen
  • an effector cell response e.g., B cell and/or T cell activation and/or mobilization
  • 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).
  • 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
  • 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, e.g., in accordance with a dosage regimen described herein), 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
  • any of the methods disclosed herein further includes identifying in a subject or a sample (e.g., a subject’s sample comprising cancer cells and/or immune cells such as TILs) the presence of GITR, thereby providing a value for GITR.
  • the method can further include comparing the GITR value to a reference value, e.g., a control value.
  • GITR value is greater than the reference value, e.g., the control value, administering a therapeutically effective amount of an anti-GITR antibody molecule described herein to the subject, and optionally, in combination with a second therapeutic agent, procedure, or modality described herein, thereby treating a cancer.
  • the reference value e.g., the control value
  • any of the methods disclosed herein further includes identifying in a subject or a sample (e.g., a subject’s sample comprising cancer cells and/or immune cells such as TILs) the presence of PD-L1, thereby providing a value for PD-L1.
  • the method can further include comparing the PD-L1 value to a reference value, e.g., a control value. If the PD-L1 value is greater than the reference value, e.g., the control value, administering a therapeutically effective amount of an anti-GITR antibody molecule described herein to the subject, and optionally, in combination with a second therapeutic agent, procedure, or modality described herein, thereby treating a cancer.
  • any of the methods disclosed herein further includes identifying in a subject or a sample (e.g., a subject’s sample comprising cancer cells and optionally immune cells such as TILs) the presence of one, two or all of PD-L1, CD8, or IFN- ⁇ , thereby providing a value for one, two or all of PD-L1, CD8, and IFN- ⁇ .
  • the method can further include comparing the PD-L1, CD8, and/or IFN- ⁇ values to a reference value, e.g., a control value.
  • the PD-L1, CD8, and/or IFN- ⁇ values are greater than the reference value, e.g., the control values, administering a therapeutically effective amount of an anti- GITR antibody molecule described herein to the subject, and optionally, in combination with a second therapeutic agent, procedure, or modality described herein, thereby treating a cancer.
  • the reference value e.g., the control values
  • the subject may have a cancer described herein, such as a solid tumor or a hematological cancer, e.g., 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 a small cell lung cancer (SCLC)), a head and neck cancer (e.g., squamous cell carcinoma of the head and neck (SCCHN)), a nasopharyngeal cancer (NPC), a skin cancer (e.g., a Merkel cell carcinoma or a melanoma (e.g., an advanced melanoma)), a neuroendocrine cancer, a neuroectodermal cancer, a kidney cancer (e.g., a renal cancer (e.g., a renal cell carcinoma (RCC))), an endometrial cancer (e.
  • a transitional cell carcinoma a lymphoma (e.g., T-cell lymphoma, B-cell lymphoma, or a non-Hogdkin lymphoma), or a metastatic lesion of the cancer.
  • lymphoma e.g., T-cell lymphoma, B-cell lymphoma, or a non-Hogdkin lymphoma
  • metastatic lesion of the cancer e.g., T-cell lymphoma, B-cell lymphoma, or a metastatic lesion of the cancer.
  • FIG.1 depicts the production of soluble mouse GITR (smGITR) in OT-II splenocytes treated with agonistic anti-mGITR antibodies (DTA-1) in pg/mL.
  • DTA-1 agonistic anti-mGITR antibodies
  • 9B8 an antagonistic hamster anti-mouse GITR antibody
  • the OT-II splenocyte media was assayed for the presence of smGITR at 2 h, 7 h, 24 h, 48 h, and 96 h.
  • FIG.2 depicts the production of smGITR in serum from mice treated with agonistic anti-mGITR antibodies (DTA-1) in pg/mL.
  • DTA-1 agonistic anti-mGITR antibodies
  • FIG.2 depicts the production of smGITR in serum from mice treated with agonistic anti-mGITR antibodies (DTA-1) in pg/mL.
  • Data shown are the mean ⁇ SEM of two mice.
  • FIG.3 depicts the production of smGITR in OT-II splenocytes incubated for 48 h or 96 h with 1 ⁇ g/mL DTA-1 or a rat IgG2b (control) in the presence or absence of 25 ⁇ M hydroxamicacid-based metalloprotease inhibitor GM6001 in pg/mL.
  • FIGS.4A-4B depict the in vivo efficacy of a single dose of DTA-1.mIgG2a in mice bearing A20 lymphoma allografts.
  • the mean tumor volume in mm 3 ⁇ SEM (FIG.4A) and the smGITR serum levels in pg/mL (FIG.4B) after treatment with 5 or 15 mg/kg DTA-1.mIgG2a or 15 mg/kg mIgG2a (control) over 15 days are shown.
  • *** indicates the day 15 significance of P ⁇ .001 between DTA-1.mIgG2a treated and mIgG2a control groups.
  • FIGS.5A-5C depict the absolute number of activated CD8+ T cells (FIG.5A), the absolute number of regulatory T cells (Tregs; FIG 5B), and the Teff/Treg ratio (FIG.5C) in A20 tumors treated with 1mg/kg DTA-1.mIgG2a, 5 mg/kg DTA-1.mIgG2a, or mIgG2a (control) at 7 days post- treatment.
  • FIGS.6A-6B depict the in vivo efficacy of a single dose of DTA-1.mIgG2a in mice bearing a Colon26 syngeneic tumor model.
  • the mean tumor volume in mm 3 ⁇ SEM (FIG.6A) and the smGITR serum levels in pg/mL (FIG.6B) after treatment with a single dose of 5 or 15 mg/kg DTA-1.mIgG2a or mIgG2a (control) over 15 days are shown.
  • * indicates the day 15 significance of P ⁇ .05 between DTA- 1.mIgG2a treated and mIgG2a control groups.
  • FIGS.7A-7C depict the absolute number of activated CD8+ T cells (FIG.7A), the absolute number of regulatory T cells (Tregs; FIG 7B), and the Teff/Treg ratio (FIG.7C) in Colon26 tumors treated with 1mg/kg DTA-1.mIgG2a, 5 mg/kg DTA-1.mIgG2a, or mIgG2a (control) at 3 days post- treatment.
  • FIGS.8A-8C depict the in vivo efficacy of a single dose of DTA-1.mIgG2a at dosages from 0.3 mg/kg to 15 mg/kg in mice bearing a Colon26 syngeneic tumor model.
  • FIGS.9A-9B depict the in vivo efficacy of a single dose of the agonistic anti-GITR antibody MAB7 in hGITR.hGITRL-double knock-in (dKI) mice bearing a Colon26 syngeneic tumor model.
  • the mean tumor volume in mm 3 ⁇ SEM (FIG.9A) and the soluble human GITR (shGITR) serum levels in pg/mL (FIG.9B) after treatment with a single dose of 15 mg/kg MAB7 or vehicle over 15 days are shown. * indicates the day 15 significance of P ⁇ .05 between MAB7 treated and vehicle control groups with unpaired T-test with two-tailed 95% confidence interval..
  • FIGS.10A-10B depict the absolute number of Tregs and the Teff/Treg ratio at 3 days (FIG.10B) and the activated CD8+ T cells, the absolute number of Tregs, and the Teff/Treg ratio at 15 days (FIG. 10B) after treatment with 15 mg/kg MAB7 in Colon26 tumors of hGITR.hGITRL-dKI mice.
  • FIG.11 depicts the leves of smGITR detected after dosing in the MC38 syngeneic murine colon tumor model with DTA-1.mIgG2a as a single agent or in combination the anti-PD-1 antibody RMP1-14.
  • FIG.12 depicts the levels of IL-2 produced in the Staph enterotoxin B (SEB) PBMC stimulation assay in the presence of titrated anti-GITR antibody MAB7 in combination the anti-PD-1 antibody PDR001 at 0.5 mg/mL, MAB7 in combination with an isotope control (human IgG1), or PDR001 in combination with an isotype control (human IgG4).
  • SEB Staph enterotoxin B
  • checkpoint inhibitors e.g., antagonist of the Programmed Cell Death Protein -1 (PD-1) protein
  • PD-1 Programmed Cell Death Protein -1
  • Glucocorticoid-induced TNFR-related protein is a distinct costimulatory receptor found on, e.g., regulatory T cells, which dampens immune responses. GITR ligation may lead to costimulation of effector T cells and thereby enhance tumor cell killing and immunological memory.
  • the interaction of PD-L1 on tumor cells and antigen presenting cells with PD-1 on effector T cells provides a distinct immune-dampening signal in the tumor microenvironment.
  • treatment with an agonistic antibody to GITR can overcome resistance to anti-tumor immunity resulting from, e.g., a PD-1 blockade, to enhance response rates.
  • antibody molecules e.g., humanized antibody molecules
  • Pharmaceutical compositions and dose formulations comprising the anti-GITR antibody molecules are also provided.
  • the anti- GITR antibody molecules disclosed herein can be used (alone or in combination with other therapeutic agents, procedures, or modalities) to treat or prevent disorders, such as cancerous disorders (e.g., solid tumors and hematological cancers), as well as infectious diseases (e.g., chronic infectious disorders or sepsis).
  • cancerous disorders e.g., solid tumors and hematological cancers
  • infectious diseases e.g., chronic infectious disorders or sepsis
  • methods, including dosage regimens, for treating various disorders using the anti- GITR antibody molecules are disclosed herein.
  • the anti- GITR antibody molecule is administered in combination with a PD-1 inhibitor, e.g., a anti-PD1 antibody molecule.
  • the articles“a” and“an” refer to one or to more than one (e.g., to at least one) of the grammatical object of the article.
  • “About” and“approximately” shall generally mean an acceptable degree of error for the quantity measured given the nature or precision of the measurements. Exemplary degrees of error are within 20 percent (%), typically, within 10%, and more typically, within 5% of a given value or range of values.
  • 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 (VL) and variable heavy chain (VH) 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.
  • An“antibody light chain” or an“antibody heavy chain” as used herein refers to a polypeptide comprising the VL or VH, respectively.
  • the endogenous VL is encoded by the gene segments V (variable) and J (junctional), and the endogenous VH by V, D (diversity), and J.
  • Each of VL or VH 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 VL or VH, as one skilled in the art will readily recognize.
  • 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 mimics that use non-immunoglobulin protein scaffolds, including but not limited to, adnectins, avimers, single chain polypeptide binding molecules, and antibody-like binding peptidomimetics.
  • 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 ⁇
  • 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.
  • 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.
  • 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.
  • A“conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain.
  • Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
  • VH and VL regions can be subdivided into regions of hypervariability, termed
  • CDR complementarity determining regions
  • CDR complementarity determining region
  • HCDR1, HCDR2, HCDR3 three CDRs in each heavy chain variable region
  • LCDR1, LCDR2, LCDR3 three CDRs in each light chain variable region
  • the precise amino acid sequence boundaries of a given CDR can be determined using any of a number of well-known schemes, including those described by Kabat et al. (1991),“Sequences of Proteins of Immunological Interest,” 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD (“Kabat” numbering scheme), Al-Lazikani et al., (1997) JMB 273,927-948 (“Chothia” numbering scheme). As used herein, the CDRs defined according to the“Chothia” number scheme are also sometimes referred to as“hypervariable loops.”
  • the CDR amino acid residues in the heavy chain variable domain (VH) are numbered 31-35 (HCDR1) (e.g., insertion(s) after position 35), 50-65 (HCDR2), and 95-102 (HCDR3); and the CDR amino acid residues in the light chain variable domain (VL) are numbered 24-34 (LCDR1) (e.g., insertion(s) after position 27), 50-56 (LCDR2), and 89-97 (LCDR3).
  • the CDR amino acids in the VH are numbered 26-32 (HCDR1) (e.g., insertion(s) after position 31), 52-56 (HCDR2), and 95-102 (HCDR3); and the amino acid residues in VL are numbered 26-32 (LCDR1) (e.g., insertion(s) after position 30), 50-52 (LCDR2), and 91-96 (LCDR3).
  • the CDRs comprise or consist of, e.g., 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.
  • each VH and VL typically includes three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes).
  • the length of a reference sequence aligned for comparison purposes is at least 30%, preferably at least 40%, more preferably at least 50%, 60%, and even more preferably at least 70%, 80%, 90%, 100% of the length of the reference sequence.
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared.
  • amino acid or nucleic acid “identity” is equivalent to amino acid or nucleic acid “homology”
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
  • the percent identity between two amino acid sequences is determined using the Needleman and Wunsch ((1970) J. Mol. Biol. 48:444-453 ) algorithm which has been incorporated into the GAP program in the GCG software package (available at www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.
  • the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package (available at www.gcg.com), using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6.
  • a particularly preferred set of parameters are a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
  • the percent identity between two amino acid or nucleotide sequences can be determined using the algorithm of E. Meyers and W. Miller ((1989) CABIOS, 4:11-17) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • nucleic acid and protein sequences described herein can be used as a "query sequence" to perform a search against public databases to, for example, identify other family members or related sequences.
  • Such searches can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) J. Mol. Biol.215:403-10.
  • Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res.25:3389-3402.
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • XBLAST and NBLAST can be used. See www.ncbi.nlm.nih.gov.
  • 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.
  • 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.
  • Complementarity-determining domains or“complementary-determining regions (“CDRs”) interchangeably refer to the hypervariable regions of V L and V H .
  • CDRs are the target protein-binding site of the antibody chains that harbors specificity for such target protein.
  • CDR1- 3 There are three CDRs (CDR1- 3, numbered sequentially from the N-terminus) in each human V L or V H , constituting about 15-20% of the variable domains.
  • 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.
  • IMGT international ImMunoGeneTics database
  • 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.
  • 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 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. Biol.2:593-596 (1992)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such humanized antibodies are chimeric antibodies (U.S. Patent 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.4:417-302001.
  • 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.
  • 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.
  • 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).
  • 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 biological sample e.g., a blood, serum, plasma or tissue 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 (KD, M) refers to the dissociation rate constant (kd, time -1 ) divided by the association rate constant (ka, 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 (KD) of about 1 nM or less.
  • the antibody or antibody fragment binds to human GITR with a KD that is less than 1 nM. In some embodiments, the antibody or antibody fragment binds to human GITR with a KD that is in the range of from about 0.5 nM to about 1.0 nM.
  • 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 ⁇ GenBank Accession Nos. NM_004195.2 ⁇ NP_004186.1 (isoform 1 precursor), SEQ ID NO:1:
  • 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.
  • 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 or“polynucleotide” 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.
  • conservatively modified variants thereof e.g., degenerate codon substitutions
  • alleles e.g., orthologs, SNPs, and complementary sequences as well as the sequence explicitly indicated.
  • 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
  • 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
  • HSPs high scoring sequence pairs
  • W short words of length W in the query sequence
  • T is referred to as the neighborhood word score threshold (Altschul et al., supra).
  • M forward score for a pair of matching residues; always > 0
  • N penalty score for mismatching residues; always ⁇ 0).
  • 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.
  • 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.
  • 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.
  • a compound to inhibit a measurable parameter e.g., cancer
  • a measurable parameter e.g., cancer
  • 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.
  • the term“antigen presenting cell” or“APC” refers to 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.
  • T-cells may recognize these complexes using their T-cell receptors (TCRs).
  • APCs process antigens and present them to T-cells.
  • 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,
  • 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 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.
  • “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, 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).
  • parenteral administration and“administered parenterally” as used herein refers to 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.
  • a “therapeutically effective amount” as used herein refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result.
  • the pharmaceutical compositions of the invention may include a “therapeutically effective amount” or a “prophylactically effective amount” of an antibody or antibody portion of the invention.
  • 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.
  • a measurable parameter e.g., tumor growth rate
  • 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.
  • 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.
  • 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 anti-GITR antibody molecules described herein can be administered according to a dosage regimen described herein to treat (e.g., inhibit, reduce, ameliorate, or prevent) 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 anti- GITR antibody molecule is administered to the subject at a dose of about 2 mg to about 600 mg, e.g., once every week, once every three weeks, or once every six weeks.
  • the anti- GITR antibody molecule is administered to the subject at a dose of about 2 mg to about 2400 mg, e.g., once every week, once every three weeks, or once every six weeks.
  • the anti- GITR antibody molecule is administered to the subject at a dose of about 2 mg to about 4000 mg, e.g., once every week, once every
  • the anti-GITR antibody molecule is administered at a dose of about 1 mg to about 1500 mg, about 2 mg to about 1400 mg, about 5 mg to about 1300 mg, about 10 mg to about 1300 mg, about 20 mg to about 1200 mg, about 30 mg to about 1000 mg, about 40 mg to about 800 mg, about 50 mg to about 700 mg, about 60 mg to about 600 mg, about 70 mg to about 500 mg, about 80 mg to about 300 mg, about 90 mg to about 200 mg, about 100 mg to about 150 mg, 1 mg to about 150 mg, about 2 mg to about 300 mg, about 5 mg to about 500 mg, about 10 mg to about 600 mg, about 20 mg to about 700 mg, about 30 mg to about 800 mg, about 40 mg to about 900 mg, about 50 mg to about 1000 mg, about 60 mg to about 1100 mg, about 70 mg to about 1200 mg, about 80 mg to about 1300 mg, about 90 mg to about 1400 mg, or about 100 mg to about 1500 mg, e.g., once every week, two weeks, three weeks, four weeks
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 3600 mg, about 2 mg to about 3400 mg, about 5 mg to about 3200 mg, about 10 mg to about 3300 mg, about 20 mg to about 3200 mg, about 30 mg to about 3000 mg, about 40 mg to about 2800 mg, about 50 mg to about 2700 mg, about 60 mg to about 2600 mg, about 70 mg to about 2500 mg, about 80 mg to about 2300 mg, about 90 mg to about 2200 mg, about 100 mg to about 2100 mg, 1 mg to about 1900 mg, about 2 mg to about 1800 mg, about 5 mg to about 1700 mg, about 10 mg to about 1600 mg, about 20 mg to about 1500 mg, about 30 mg to about 2400 mg, about 40 mg to about 2300 mg, about 50 mg to about 2200 mg, about 60 mg to about 2100 mg, about 70 mg to about 2000 mg, about 80 mg to about 1900 mg, about 90 mg to about 1800 mg, about 100 mg to about 1700 mg, about 2000 mg to about 2500 mg,
  • the anti-GITR antibody molecule is administered at a dose of about 1 mg to about 100 mg, 5 mg to about 90 mg, about 10 mg to about 80 mg, about 15 mg to about 75 mg, about 20 mg to about 70 mg, about 30 mg to about 60mg, about 40 mg to about 50mg, e.g., about 5 mg, about 10 mg, about 30 mg, about 60 mg, or about 100 mg, e.g., once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg, e.g., about 5 mg, once every week, three or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg, e.g., about 10 mg, once every week, three or six weeks. In other embodiments, the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 50 mg, e.g., about 30 mg, once every week, three or six weeks. In other embodiments, the anti-GITR antibody molecule is administered at a dose of about 30 mg to about 100 mg, e.g., about 60 mg, once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 300 mg, about 120 mg to about 280 mg, about 140 mg to about 250 mg, about 150 mg to about 240 mg, about 160 mg to about 200 mg, e.g., about 100 mg, about 130 mg, about 150 mg, about 200 mg, about 230 mg, about 240 mg, about 260 mg, about 280 mg, or about 300 mg, e.g., once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg, e.g., about 150 mg, once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 300 mg to about 1000 mg, about 400 mg to about 900 mg, about 500 mg and 800 mg, about 600 mg to about 700, about 300 mg to about 5400 mg, e.g., about 300 mg, about 400 mg, about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, or about 1000 mg, e.g., about 300 mg or about 500 mg, once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg, e.g., about 300 mg, once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 1000 mg, e.g., about 500 mg, once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 300 mg to about 3000 mg, about 400 mg to about 2900 mg, about 500 mg to about 2800 mg, about 600 mg to about 2700, about 300 mg to about 2500 mg, e.g., about 375 mg, about 750 mg, about 1200 mg, or about 1500 mg, e.g., once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 500 mg, e.g., about 375 mg, once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 1000 mg, e.g., about 750 mg, once every week, three weeks, or six weeks. In other embodiments, the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 2000 mg, e.g., about 1200 mg, once every week, three weeks, or six weeks. In other embodiments, the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 2000 mg, e.g., about 1500 mg, once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 1800 mg, about 250 mg to about 1275 mg, about 300 mg to about 1250 mg, about 325 mg to about 1225 mg, or about 350 mg to about 1200 mg, e.g., about 375 mg, about 750 mg, about 1200 mg, e.g., once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 500 mg, e.g., about 375 mg, once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 1000 mg, e.g., about 750 mg, once every week, three weeks, or six weeks. In other embodiments, the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 1500 mg, e.g., about 1200 mg, once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2000 mg to about 2500 mg about 2800 mg to about 3200 mg, or about 2500 mg to about 3500 mg, e.g., about 2250 mg or about 3000 mg, e.g., once every week, three weeks, or six weeks. In certain embodiments, the anti-GITR antibody molecule is administered at a dose of 2000 mg to about 2500 mg, e.g., about 2250 mg, once every week, three weeks, or six weeks. In other embodiments, the anti-GITR antibody molecule is administered at a dose of about 2500 mg to about 3500 mg, e.g., about 3000 mg, once every week, three weeks, or six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 500 mg, about 10 mg to about 300 mg, about 30 mg to about 300 mg, about 60 mg to about 150 mg, about 5 mg to about 60 mg, about 10 mg to about 150 mg, about 30 mg to about 500 mg, about 60 mg to about 150 mg, about 60 to about 300 mg, about 150 mg to about 500 mg, once every week, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 2000 mg, about 150 mg to about 1500 mg, about 200 mg to about 1400 mg, about 300 mg to about 1300 mg, about 350 mg to about 1100 mg, about 150 mg to about 1000 mg, about 200 mg to about 900 mg, about 250 mg to about 850 mg, about 275 mg to about 800 mg, about 350 mg to about 775 mg, or about 375 mg to about 750 mg, once every week, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 1500 mg or less, about 1400 mg or less, about 1300 mg or less, about 1200 mg or less, about 1100 mg or less, about 1000 mg or less, about 900 mg or less, about 800 mg or less, about 700 mg or less, about 600 mg or less, about 500 mg or less, about 450 mg or less, about 400 mg or less, about 350 mg or less, about 300 mg or less, about 250 mg or less, about 200 mg or less, about 150 mg or less, about 140 mg or less, about 130 mg or less, about 120 mg or less, about 100 mg or less, about 90 mg or less, about 80 mg or less, about 70 mg or less, about 60 mg or less, about 50 mg or less, about 40 mg or less, about 30 mg or less, about 20 mg or less, about 10 mg or less, or about 5 mg or less, once every week, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 4000 mg or less, about 3800 mg or less, about 3600 mg or less, about 3400 mg or less, about 3300 mg or less, about 3200 mg or less, about 3100 mg or less, about 3000 mg or less, about 2900 mg or less, about 2800 mg or less, about 2700 mg or less, about 2600 mg or less, about 2500 mg or less, about 2400 mg or less, about 2200 mg or less, about 2200 mg or less, about 2100 mg or less, about 2000 mg or less, about 1900 mg or less, about 1800 mg or less, about 1700 mg or less, about 1600 mg or less, about 1500 mg or less, about 1450 mg or less, about 1400 mg or less, about 1350 mg or less, about 1300 mg or less, about 1250 mg or less, about 1200 mg or less, about 1150 mg or less, about 1100 mg or less, about 1050 mg or less, about 1000 mg or less, about 950 mg or less, about 900
  • the anti-GITR antibody molecule is administered once every three weeks.
  • a once every three weeks dosing schedule can provide continuous depletion of regulatory T cells (Tregs) and costimulation of T effector cells (Teffs), e.g., based on pharmacokinetics.
  • this schedule can be used to provide continuous exposure for 3 months at a time.
  • the anti-GITR antibody molecule is administered once every six weeks or once every week.
  • a once every six weeks or once every week dosing schedule can be used as an option to address emerging safety signals and/or the need for higher antibody doses to attain more profound depletion of Tregs.
  • a once every six weeks dosing schedule may allow the antibody exposure to return
  • a once every week dosing schedule may provide additional tumor penetration and enhance Treg depletion, e.g., to provide high exposure in a short period of time to maximize tumor penetration and Treg elimination.
  • the subject receives three doses of the anti-GITR antibody molecule over a period of three weeks followed by a one-week pause, two-week pause, three-week pause, four-week pause, five-week pause, six-week pause, seven-week pause, eight-week pause, nine-week pause, or ten- week pause, e.g., a nine-week pause.
  • the subject receives four doses of the anti- GITR antibody molecule over a period of twelve weeks, sixteen weeks, twenty weeks, twenty-one weeks, or twenty four weeks followed by a one-week pause, two-week pause, three-week pause, four-week pause, five-week pause, six-week pause, seven-week pause, eight-week pause, nine-week pause, or ten- week pause, e.g., a nine-week pause.
  • a dosing pause can be used to reduce lymphocyte over-stimulation and exhaustion, e.g., exhaustion of T effector cells (Teffs).
  • a nine-week pause may allow a reasonably low trough in a number of different regimens.
  • the anti-GITR antibody molecule is administered at a dose that results in at least 50%, 60%, 70%, 80%, 90%, 95%, or 98% binding, e.g., occupancy, of GITR in a tumor in the subject, e.g., within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 weeks of administration.
  • the anti-GITR antibody molecule is administered at a dose that cause 5% or more, e.g., 10% or more, 15% or more, 20% or more, 25% or more, 30% or more, 35% or more, 40% or more, 45% or more, or 50% or more of the maximal response of soluble GITR (sGITR), in the subject, e.g., within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 weeks of administration.
  • sGITR soluble GITR
  • the disorder is a cancer, e.g., a cancer described herein.
  • the cancer is a solid tumor.
  • the cancer is a lung cancer, e.g., a small cell lung cancer (SCLC) or a non-small cell lung cancer (NSCLC).
  • SCLC small cell lung cancer
  • NSCLC non-small cell lung cancer
  • the cancer is a head and neck cancer (e.g., a head and neck squamous cell carcinoma (HNSCC)).
  • HNSCC head and neck squamous cell carcinoma
  • the cancer is a nasopharyngeal cancer (NPC). In other embodiments, the cancer is a lymphoma. In some embodiments, the cancer is an ovarian cancer. In other embodiments, the cancer is a mesothelioma. In other embodiments, the cancer is a skin cancer, e.g., a Merkel cell carcinoma or a melanoma. In other embodiments, the cancer is a kidney cancer, e.g., a renal cell carcinoma. In other embodiments, the cancer is a bladder cancer. In other embodiments, the cancer is a soft tissue sarcoma, e.g., a hemangiopericytoma (HPC).
  • HPC hemangiopericytoma
  • the cancer is a bone cancer, e.g., a bone sarcoma. In other embodiments, the cancer is a colorectal cancer. In other embodiments, the cancer is a pancreatic cancer. In other embodiments, the cancer is a breast cancer. In other embodiments, the cancer is a duodenal cancer. In other embodiments, the cancer is an endometrial cancer. In other embodiments, the cancer is an adenocarcinoma, e.g., an unknown adenocarcinoma. In other embodiments, the cancer is a liver cancer, e.g., a hepatocellular carcinoma. In other embodiments, the cancer is a
  • the cancer is a sarcoma.
  • the cancer is a myelodysplastic syndrome (MDS) (e.g., a high risk MDS).
  • MDS myelodysplastic syndrome
  • the cancer is a leukemia (e.g., an acute myeloid leukemia (AML), e.g., a relapsed or refractory AML or a de novo AML).
  • AML acute myeloid leukemia
  • the cancer is a myeloma.
  • the cancer is an MSI-high cancer.
  • the cancer is a metastatic cancer.
  • the cancer is an advanced cancer.
  • the cancer is a relapsed or refractory cancer.
  • the cancer is a Merkel cell carcinoma. In other embodiments, the cancer is a melanoma. In other embodiments, the cancer is a breast cancer, e.g., a triple negative breast cancer (TNBC) or a HER2-negative breast cancer. In other embodiments, the cancer is a renal cell carcinoma (e.g., a clear cell renal cell carcinoma (CCRCC) or a non-clear cell renal cell carcinoma (nccRCC)). In other embodiments, the cancer is a thyroid cancer, e.g., an anaplastic thyroid carcinoma (ATC).
  • TNBC triple negative breast cancer
  • HER2-negative breast cancer e.g., the cancer is a renal cell carcinoma (e.g., a clear cell renal cell carcinoma (CCRCC) or a non-clear cell renal cell carcinoma (nccRCC)).
  • the cancer is a thyroid cancer, e.g., an anaplastic thyroid carcinoma (ATC).
  • the cancer is a neuroendocrine tumor (NET), e.g., an atypical pulmonary carcinoid tumor or an NET in pancreas, gastrointestinal (GI) tract, or lung.
  • NET neuroendocrine tumor
  • the cancer is a non- small cell lung cancer (NSCLC) (e.g., a squamous NSCLC or a non-squamous NSCLC).
  • NSCLC non- small cell lung cancer
  • the cancer is a fallopian tube cancer.
  • the cancer is a microsatellite instability-high colorectal cancer (MSI-high CRC) or a microsatellite stable colorectal cancer (MSS CRC).
  • the anti-GITR antibody molecule is administered in combination with an anti-PD-1 antibody molecule (e.g., an anti-PD-1 antibody molecule described herein).
  • the anti-PD-1 antibody molecule is administered at a dose of about 50 mg to about 200 mg (e.g., about 100 mg) or about 100 mg to about 300 mg (e.g., about 200 mg), e.g., once every week, once every two weeks, once every three weeks, once every four weeks, once every five weeks, or once every six weeks (e.g., once every two weeks, once every three weeks, or once every six weeks).
  • the anti-PD-1 antibody molecule is administered at a dose of about 300 mg to about 400 mg (e.g., about 300 mg), e.g., once every week, once every two weeks, once every three weeks, once every four weeks, once every five weeks, or once every six weeks (e.g., once every two weeks, once every three weeks, or once every six weeks).
  • the anti-PD-1 antibody molecule is administered at a dose of about 100 mg to about 400 mg (e.g., about 250 mg), e.g., once every week, once every two weeks, once every three weeks, once every four weeks, once every five weeks, or once every six weeks (e.g., once every two weeks, once every three weeks, or once every six weeks).
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg (e.g., about 5 mg) once every week, once every three weeks, once every four weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 50 mg to about 200 mg (e.g., about 100 mg) once every week, once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 2 mg to about 10 mg (e.g., about 5 mg) once every week, once every three weeks, once every four weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 100 mg to about 300 mg (e.g., about 200 mg) once every week, once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg (e.g., about 10 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 200 mg to about 400 mg (e.g., about 300 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg (e.g., about 10 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 200 mg to about 400 mg (e.g., about 300 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg (e.g., about 10 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 200 mg to about 600 mg (e.g., about 400 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg (e.g., about 10 mg) once every week, once every three weeks, once every four weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 50 mg to about 200 mg (e.g., about 100 mg) once every week, once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 5 mg to about 20 mg (e.g., about 10 mg) once every week, once every three weeks, once every four weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 100 mg to about 300 mg (e.g., about 200 mg) once every week, once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg (e.g., about 30 mg) once every week, once every three weeks, once every four weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 50 mg to about 200 mg (e.g., about 100 mg) once every week, once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg (e.g., about 30 mg) once every week, once every three weeks, once every four weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 100 mg to about 300 mg (e.g., about 200 mg) once every week, once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 20 mg to about 40 mg (e.g., about 30 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 200 mg to about 600 mg (e.g., about 300 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg (e.g., about 60 mg) once every week, once every three weeks, once every four weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 50 mg to about 200 mg (e.g., about 100 mg) once every week, once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg (e.g., about 60 mg) once every week, once every three weeks, once every four weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 100 mg to about 300 mg (e.g., about 200 mg) once every week, once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 100 mg (e.g., about 60 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 200 mg to about 600 mg (e.g., about 300 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 150 mg (e.g., about 75 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 50 mg to about 200 mg (e.g., about 100 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 50 mg to about 150 mg (e.g., about 75 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 100 mg to about 300 mg (e.g., about 200 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti- GITR antibody molecule is administered at a dose of about 50 mg to about 150 mg (e.g., about 75 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 200 mg to about 600 mg (e.g., about 300 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg (e.g., about 150 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 50 mg to about 200 mg (e.g., about 100 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg (e.g., about 150 mg) once every week, once every three weeks, or once every six weeks and the anti- PD-1 antibody molecule is administered at a dose of about 100 mg to about 300 mg (e.g., about 200 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg (e.g., about 150 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 200 mg to about 600 mg (e.g., about 300 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg (e.g., about 300 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 50 mg to about 200 mg (e.g., about 100 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg (e.g., about 300 mg) once every week, once every three weeks, or once every six weeks and the anti- PD-1 antibody molecule is administered at a dose of about 100 mg to about 300 mg (e.g., about 200 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg (e.g., about 300 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 200 mg to about 600 mg (e.g., about 300 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 250 mg to about 500 mg (e.g., about 375 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 50 mg to about 200 mg (e.g., about 100 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 250 mg to about 500 mg (e.g., about 375 mg) once every week, once every three weeks, or once every six weeks and the anti- PD-1 antibody molecule is administered at a dose of about 100 mg to about 300 mg (e.g., about 200 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 250 mg to about 500 mg (e.g., about 375 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 200 mg to about 600 mg (e.g., about 300 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg (e.g., about 500 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 50 mg to about 200 mg (e.g., about 100 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg (e.g., about 500 mg) once every week, once every three weeks, or once every six weeks and the anti- PD-1 antibody molecule is administered at a dose of about 100 mg to about 300 mg (e.g., about 200 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 400 mg to about 600 mg (e.g., about 500 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 200 mg to about 600 mg (e.g., about 300 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 500 mg to about 1000 mg (e.g., about 750 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 50 mg to about 200 mg (e.g., about 100 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 500 mg to about 1000 mg (e.g., about 750 mg) once every week, once every three weeks, or once every six weeks and the anti- PD-1 antibody molecule is administered at a dose of about 100 mg to about 300 mg (e.g., about 200 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 500 mg to about 1000 mg (e.g., about 750 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 200 mg to about 600 mg (e.g., about 300 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 2000 mg (e.g., about 1500 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 50 mg to about 200 mg (e.g., about 100 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 2000 mg (e.g., about 1500 mg) once every week, once every three weeks, or once every six weeks and the anti- PD-1 antibody molecule is administered at a dose of about 100 mg to about 300 mg (e.g., about 200 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 1000 mg to about 2000 mg (e.g., about 1500 mg) once every week, once every three weeks, or once every six weeks and the anti-PD-1 antibody molecule is administered at a dose of about 200 mg to about 600 mg (e.g., about 300 mg) once every two weeks, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg (e.g., about 20 mg) once every two weeks and the anti-PD-1 antibody molecule is administered at a dose of about 50 mg to about 200 mg (e.g., about 80 mg) once every week, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 100 mg to about 200 mg (e.g., about 20 mg) once every two weeks and the anti-PD-1 antibody molecule is administered at a dose of about 100 mg to about 300 mg (e.g., about 200 mg) once every week, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg (e.g., about 80 mg) once every two weeks and the anti-PD-1 antibody molecule is administered at a dose of about 100 mg to about 300 mg (e.g., about 150 mg) once every week, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 200 mg to about 400 mg (e.g., about 300 mg) once every two weeks and the anti-PD-1 antibody molecule is administered at a dose of about 200 mg to about 400 mg (e.g., about 300 mg) once every week, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 400 mg to 600 mg (e.g., about 240 mg) once every two weeks and the anti-PD-1 antibody molecule is administered at a dose of about 100 mg to about 300 mg (e.g., about 200 mg) once every week, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule is administered at a dose of about 400 mg to 600 mg (e.g., about 240 mg) once every two weeks and the anti-PD-1 antibody molecule is administered at a dose of about 100 mg to about 400 mg (e.g., about 300 mg) once every week, once every three weeks, or once every six weeks.
  • the anti-GITR antibody molecule, the anti-PD-1 antibody molecule, or both is adminsetered via intravenous infusion, e.g., over 30-60 minutes.
  • the anti- GITR antibody molecule and the anti-PD-1 antibody molecule are administered with at least 30, 60, or 90 minutes break between the two antibody administrations.
  • the subject receives two doses of the anti-PD-1 antibody molecule over a period of three weeks followed by a one-week pause, two-week pause, three-week pause, four-week pause, five-week pause, six-week pause, seven-week pause, eight-week pause, nine-week pause, or ten- week pause, e.g., a nine-week pause.
  • the subject receives four doses of the anti- PD-1 antibody molecule over a period of twelve weeks, sixteen weeks, twenty weeks, twenty-one weeks, or twenty four weeks followed by a one-week pause, two-week pause, three-week pause, four-week pause, five-week pause, six-week pause, seven-week pause, eight-week pause, nine-week pause, or ten- week pause, e.g., a nine-week pause.
  • 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 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 5 and Table 6 below.
  • BSD binding sequence determinant
  • 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 5 and Table 6).
  • 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 VH33-13/30: EVQLVESGGGLVQPGGSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVAVIRYDGSNKYYADS VKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK (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:
  • the amino acids EIVMTQSPATLSVSPGERATLSCRASQSVSSNLAWYQQKPGQAPRLLIYGASTRATGIPDRFSGSG SGTDFTLTISRLEPEDFAVYYCQQYGSSP (SEQ ID NO:92).
  • 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
  • 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
  • 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.
  • 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
  • EVQLVESGGGLVQPGGSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVAVIRYDGSNKYYADS VKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK– YFDYWGQGTLVTVSS (SEQ ID NOS:89 and 91)(VH33-13/30+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 EIVMTQSPATLSVSPGERATLSCRASQSVSSNLAWYQQKPGQAPRLLIYGASTRATGIPDRFSGSG SGTDFTLTISRLEPEDFAVYYC–YTFGQGTKLEIK (SEQ ID NOS:98 and 94) (VKIII
  • the CDRs of the antibodies listed in Table 5 can be determined by well known numbering systems known in the art, including those described herein.
  • Table 6 listed the CDRs that are defined by (1) using the numbering system described in Kabat et al. (1991),“Sequences of Proteins of
  • 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 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
  • the light chain CDR3 comprises SEQ ID NO:34
  • the heavy chain CDR1 comprises SEQ ID NO:22
  • the heavy chain CDR2 comprises SEQ ID NO:26
  • 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
  • the light chain CDR3 comprises SEQ ID NO:34
  • 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:27, 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
  • 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.
  • 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
  • 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
  • RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDS TYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC SEQ ID NO:21.
  • amino acids within the constant regions are added, deleted, or substituted.
  • 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 (KD) 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.
  • KD equilibrium dissociation constant
  • the GITR agonist is an anti-GITR antibody molecule. In one embodiment, the GITR agonist is an anti-GITR antibody molecule as described in International Application No. WO 2016/05784, published on April 14, 2016, entitled“Compositions and Methods of Use for Augmented Immune Response and Cancer Therapy,” incorporated by reference in its entirety. In some embodiments, the GITR agonist is GWN323 (NVS), BMS-986156, MK-4166 or MK-1248 (Merck), TRX518 (Leap Therapeutics), INCAGN1876 (Incyte/Agenus), AMG 228 (Amgen) or INBRX-110 (Inhibrx).
  • exemplary anti-GITR antibody molecules are described, e.g., in WO 2006/105021, US 9,028,823, US 8,591,886, EP 1866339, WO 2009/009116, EP 2175884, WO 2011/028683, US 8,709,424, WO 2013/039954, US 2014/0072566, WO 2015/031667, WO 2011/071871, WO 2004/060319, WO 2011/09789, WO 2013/043569, WO 2014/012479, US 8,409,577, WO 2007/146968, WO 2010/003118, WO 2011/090754, US 8,193,322, WO 2013/142255, WO 2014/022592, WO 2013/049254, WO
  • the GITR agonist is a peptide that activates the GITR signaling pathway.
  • the GITR agonist is an immunoadhesin binding fragment (e.g., an immunoadhesin binding fragment comprising an extracellular or GITR binding portion of GITRL) fused to a constant region (e.g., an Fc region of an immunoglobulin sequence).
  • a constant region e.g., an Fc region of an immunoglobulin sequence
  • the anti-GITR antibody molecules described herein include 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 MCV
  • 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 anti-GITR antibody molecules described herein include 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. Engineered and Modified Antibodies
  • the anti-GITR antibody molecules described herein include engineered and/or modified antibodies.
  • 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.
  • 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
  • framework residues within VH and/or VL e.g. to improve the properties of the antibody.
  • 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.
  • 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 embodiements 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. Patent 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
  • the antibody is modified to increase its biological half-life.
  • 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. Patent 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. Patent 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 ⁇ Rl, 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 anti-GITR antibody molecules described herein include 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, MA), ankyrin (Molecular Partners AG, Zurich, Switzerland), domain antibodies (Domantis, Ltd., Cambridge, MA, and Ablynx nv, Zwijnaarde, Belgium), lipocalin (Pieris Proteolab AG, Freising, Germany), small modular immuno-pharmaceuticals (Trubion Pharmaceuticals Inc., Seattle, WA), maxybodies (Avidia, Inc., Mountain View, CA), Protein A (Affibody AG, Sweden), and affilin (gamma-crystallin or ubiquitin) (Scil Proteins GmbH, Halle, Germany).
  • fibronectin Compound Therapeutics, Inc., Waltham, MA
  • ankyrin Molecular Partners AG, Zurich, Switzerland
  • domain antibodies Domantis, Ltd., Cambridge, MA, and Ablynx nv, Zwijnaard
  • 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 US 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.2004/0175756; 2005/0053973; 2005/0048512; and 2006/0008844.
  • 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., US 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 Brassicae 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 anti-GITR antibody molecules described herein include 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.
  • the human GITR-binding antibodies can be generated using methods that are known in the art.
  • the Humaneered ® technology platform (KaloBios, Sout San Francisco, CA) was used to convert non-human antibodies into engineered human antibodies.
  • 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.
  • 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.
  • the anti-GITR antibody molecules described herein include camelid antibodies.
  • Antibody proteins obtained from members of the camel and dromedary (Camelus bactrianus and Calelus dromaderius) family including new world members such as llama species (e.g., Lama paccos, Lama glama, and Lama vicugna) have been characterized with respect to size, structural complexity and antigenicity for human subjects.
  • 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
  • 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”.
  • a“camelid nanobody” See U.S. patent number 5,759,808 issued June 2, 1998; see also Stijlemans, B. et al., 2004 J Biol Chem 279: 1256-1261; Dumoulin, M. et al., 2003 Nature 424: 783-788; Pleschberger, M. et al.2003 Bioconjugate Chem 14: 440-448; Cortez-Retamozo, V.
  • 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 August 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. Multivalent Antibodies
  • the anti-GITR antibody molecules described herein include multivalent molecules (monospecific, bispecific, or multispecific) comprising a GITR-binding antibody, or a fragment thereof, of the invention.
  • 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).
  • first and second epitopes overlap. In an embodiment the first and second epitopes do not overlap. In an embodiment 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. Patent 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.
  • 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 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
  • scDb can be expressed in bacteria in soluble, active monomeric form (see Holliger and Winter, 1997 Cancer Immunol. Immunother., 45(34): 128-30; Wu et al., 1996
  • 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-l-carboxylate (sulfo-SMCC) (see e.g., Karpovsky et al., 1984 J. Exp.
  • 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 x mAb, mAb x Fab, Fab x 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. Patent Number 5,260,203; U.S. Patent Number 5,455,030; U.S.
  • Patent Number 4,881,175 U.S. Patent Number 5,132,405; U.S. Patent Number 5,091,513; U.S. Patent Number 5,476,786; U.S. Patent Number 5,013,653; U.S. Patent Number 5,258,498; and U.S. Patent Number 5,482,858.
  • 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 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.
  • a labeled reagent e.g., an antibody
  • the anti-GITR antibody molecules described herein include 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.
  • PEG polyethyleneglycol
  • PSA polysialic acid
  • HES hydroxyethyl starch
  • albumin-binding ligands and carbohydrate shields
  • carbohydrate shields by genetic fusion to proteins binding
  • 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.
  • the term“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 0154316 by Nishimura et al. and EP 0401384 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.
  • HES hydroxyethyl starch
  • 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. Patent No.
  • 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 anti-GITR antibody molecules described herein include 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. Patent 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
  • 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. Patent Nos.5,605,793, 5,811,238, 5,830,721, 5,834,252, and 5,837,458; Patten et al., 1997, Curr.
  • 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, CA, 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
  • 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, 1- dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, puromycin, maytansinoids, e.g., maytansinol (see U.S. Pat.
  • 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,
  • 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
  • anthracyclinies e.g., daunorubicin (formerly daunomycin) and doxorubicin
  • antibiotics e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)
  • anti-mitotic agents e.g., vincristine, vinblastine, taxol and maytansinoids
  • Antibodies may also be attached to solid supports, which are particularly useful for
  • Such solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
  • An anti-GITR antibody molecules can be combined with an immune therapy, e.g., an inhibitor of a co-inhibitory molecule (e.g., a PD-1 inhibitor (e.g., an anti-PD-1 antibody molecule), a PD-L1 inhibitor (e.g., an anti-PD-L1 antibody molecule), a PD-L2 inhibitor (e.g., an anti-PD-L2 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)), a cytokine (e.g., IL-15 complexed with a soluble form of IL-15 receptor alpha (IL-15Ra)), or any combination thereof.
  • a PD-1 inhibitor e.g., an anti-PD-1 antibody molecule
  • a PD-L1 inhibitor e.g., an anti-PD-L1 antibody molecule
  • an anti-GITR antibody molecules is administered in combination with a PD-1 inhibitor, e.g., in accordance with dosage regimens described herein.
  • PD-1 is a naturally occurring immune inhibitory molecule and antibodies blocking PD-1 allow continued function of tumor-specific Teff cells expressing PD-1.
  • combining anti-PD-1 with anti-GITR can both remove another inhibitor stimulus (Tregs) and increase the activity of effector T-cells, which may enhance immune response beyond what is attainable with either single-agent.
  • the PD-1 inhibitor is chosen from PDR001 (Novartis), Nivolumab (Bristol-Myers Squibb), Pembrolizumab (Merck & Co), Pidilizumab (CureTech), MEDI0680
  • the PD-1 inhibitor is an anti-PD-1 antibody molecule. In one embodiment, the PD-1 inhibitor is an anti-PD-1 antibody molecule as described in US 2015/0210769, published on July 30, 2015, entitled“Antibody Molecules to PD-1 and Uses Thereof,” incorporated by reference in its entirety.
  • the anti-PD-1 antibody molecule comprises at least one, two, three, four, five or six complementarity determining regions (CDRs) (or collectively all of the CDRs) from a heavy and light chain variable region comprising an amino acid sequence shown in Table 3 (e.g., from the heavy and light chain variable region sequences of BAP049-Clone-E or BAP049-Clone-B disclosed in Table 3), or encoded by a nucleotide sequence shown in Table 3.
  • the CDRs are according to the Kabat definition (e.g., as set out in Table 3).
  • the CDRs are according to the Chothia definition (e.g., as set out in Table 3).
  • the CDRs are according to the combined CDR definitions of both Kabat and Chothia (e.g., as set out in Table 3).
  • the combination of Kabat and Chothia CDR of VH CDR1 comprises the amino acid sequence
  • one or more of the CDRs (or collectively all of the CDRs) have one, two, three, four, five, six or more changes, e.g., amino acid substitutions (e.g., conservative amino acid substitutions) or deletions, relative to an amino acid sequence shown in Table 3, or encoded by a nucleotide sequence shown in Table 3.
  • the anti-PD-1 antibody molecule comprises a heavy chain variable region (VH) comprising a VHCDR1 amino acid sequence of SEQ ID NO: 501, a VHCDR2 amino acid sequence of SEQ ID NO: 502, and a VHCDR3 amino acid sequence of SEQ ID NO: 503; and a light chain variable region (VL) comprising a VLCDR1 amino acid sequence of SEQ ID NO: 510, a VLCDR2 amino acid sequence of SEQ ID NO: 511, and a VLCDR3 amino acid sequence of SEQ ID NO: 512, each disclosed in Table 3.
  • VH heavy chain variable region
  • VL light chain variable region
  • the antibody molecule comprises a VH comprising a VHCDR1 encoded by the nucleotide sequence of SEQ ID NO: 524, a VHCDR2 encoded by the nucleotide sequence of SEQ ID NO: 525, and a VHCDR3 encoded by the nucleotide sequence of SEQ ID NO: 526; and a VL comprising a VLCDR1 encoded by the nucleotide sequence of SEQ ID NO: 529, a VLCDR2 encoded by the nucleotide sequence of SEQ ID NO: 530, and a VLCDR3 encoded by the nucleotide sequence of SEQ ID NO: 531, each disclosed in Table 3.
  • the anti-PD-1 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 506, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 506. In one embodiment, the anti-PD-1 antibody molecule comprises a VL comprising the amino acid sequence of SEQ ID NO: 520, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 520. In one embodiment, the anti-PD-1 antibody molecule comprises a VL comprising the amino acid sequence of SEQ ID NO: 516, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 516.
  • the anti-PD-1 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 506 and a VL comprising the amino acid sequence of SEQ ID NO: 520. In one embodiment, the anti-PD- 1 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 506 and a VL comprising the amino acid sequence of SEQ ID NO: 516.
  • the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 507, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 507. In one embodiment, the antibody molecule comprises a VL encoded by the nucleotide sequence of SEQ ID NO: 521 or 517, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 521 or 517. In one embodiment, the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 507 and a VL encoded by the nucleotide sequence of SEQ ID NO: 521 or 517.
  • the anti-PD-1 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 508, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 508. In one embodiment, the anti-PD-1 antibody molecule comprises a light chain comprising the amino acid sequence of SEQ ID NO: 522, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 522. In one embodiment, the anti-PD-1 antibody molecule comprises a light chain comprising the amino acid sequence of SEQ ID NO: 518, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 518.
  • the anti-PD-1 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 508 and a light chain comprising the amino acid sequence of SEQ ID NO: 522. In one embodiment, the anti-PD-1 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 508 and a light chain comprising the amino acid sequence of SEQ ID NO: 518.
  • the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 509, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 509.
  • the antibody molecule comprises a light chain encoded by the nucleotide sequence of SEQ ID NO: 523 or 519, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 523 or 519.
  • the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 509 and a light chain encoded by the nucleotide sequence of SEQ ID NO: 523 or 519.
  • the antibody molecules described herein can be made by vectors, host cells, and methods described in US 2015/0210769, incorporated by reference in its entirety. Table 3. Amino acid and nucleotide sequences of exemplary anti-PD-1 antibody molecules.
  • the anti-PD-1 antibody molecule is Nivolumab (Bristol-Myers Squibb), also known as MDX-1106, MDX-1106-04, ONO-4538, BMS-936558, or OPDIVO®. Nivolumab (clone 5C4) and other anti-PD-1 antibodies are disclosed in US 8,008,449 and WO 2006/121168, incorporated by reference in their entirety.
  • the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of Nivolumab, e.g., as disclosed in Table 4.
  • the anti-PD-1 antibody molecule is Pembrolizumab (Merck & Co), also known as Lambrolizumab, MK-3475, MK03475, SCH-900475, or KEYTRUDA®.
  • Pembrolizumab and other anti-PD-1 antibodies are disclosed in Hamid, O. et al. (2013) New England Journal of Medicine 369 (2): 134–44, US 8,354,509, and WO 2009/114335, incorporated by reference in their entirety.
  • the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of Pembrolizumab, e.g., as disclosed in Table 4.
  • the anti-PD-1 antibody molecule is Pidilizumab (CureTech), also known as CT-011. Pidilizumab and other anti-PD-1 antibodies are disclosed in Rosenblatt, J. et al. (2011) J Immunotherapy 34(5): 409-18, US 7,695,715, US 7,332,582, and US 8,686,119, incorporated by reference in their entirety.
  • the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of Pidilizumab, e.g., as disclosed in Table 4.
  • the anti-PD-1 antibody molecule is MEDI0680 (Medimmune), also known as AMP-514.
  • MEDI0680 and other anti-PD-1 antibodies are disclosed in US 9,205,148 and WO
  • the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of MEDI0680.
  • the anti-PD-1 antibody molecule is REGN2810 (Regeneron). In one embodiment, the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of REGN2810.
  • the anti-PD-1 antibody molecule is PF-06801591 (Pfizer). In one embodiment, the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of PF-06801591.
  • the anti-PD-1 antibody molecule is BGB-A317 or BGB-108 (Beigene). In one embodiment, the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of BGB-A317 or BGB-108.
  • the anti-PD-1 antibody molecule is INCSHR1210 (Incyte), also known as INCSHR01210 or SHR-1210. In one embodiment, the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of INCSHR1210.
  • the anti-PD-1 antibody molecule is TSR-042 (Tesaro), also known as ANB011.
  • the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of TSR-042.
  • anti-PD-1 antibodies include those described, e.g., in WO 2015/112800, WO 2016/092419, WO 2015/085847, WO 2014/179664, WO 2014/194302, WO 2014/209804, WO
  • the anti-PD-1 antibody is an antibody that competes for binding with, and/or binds to the same epitope on PD-1 as, one of the anti-PD-1 antibodies described herein.
  • the PD-1 inhibitor is a peptide that inhibits the PD-1 signaling pathway, e.g., as described in US 8,907,053, incorporated by reference in its entirety.
  • 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 WO 2010/027827 and WO 2011/066342, incorporated by reference in their entirety).
  • Table 4 Amino acid sequences of other exemplary anti-PD-1 antibody molecules
  • Anti-GITR antibodies, antigen binding molecules, and fragments thereof, described herein 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
  • 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
  • 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
  • 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
  • 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.
  • bacilli such as Bacillus subtilis
  • enterobacteriaceae such as Salmonella, Serratia, and various Pseudomonas species.
  • 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).
  • endogenous immunoglobulin genes e.g., the myeloma hybridoma clones as described in the Examples
  • an exogenous expression vector e.g., the SP2/0 myeloma cells exemplified below.
  • SP2/0 myeloma cells exemplified below
  • immunoglobulins have been developed, including the CHO cell lines, various Cos cell lines, HeLa cells, myeloma cell lines, transformed B-cells and hybridomas.
  • 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.
  • 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. Assays for Identifying Agonist Anti-GITR Antibodies
  • 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 the present anti-GITR antibodies or antigen-binding molecules formulated together with a pharmaceutically acceptable carrier.
  • the pharmaceutical compositions described herein can be used in accordance with the dosage regimens described herein.
  • 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.
  • compositions e.g., pharmaceutically acceptable compositions, which include an anti-GITR antibody molecule described herein, formulated together with a pharmaceutically acceptable carrier.
  • compositions described herein 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., dispersions or suspensions
  • 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 (e.g., intravenous)).
  • the antibody is administered by intravenous infusion or injection (e.g., intravenous infusion).
  • the antibody is administered by intramuscular or subcutaneous injection.
  • compositions typically should be sterile and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high antibody concentration.
  • Sterile injectable solutions can be prepared by incorporating the active compound (e.g., antibody or antibody portion) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.
  • 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).
  • An anti-GITR antibody molecule or a composition described herein can be formulated into a formulation (e.g., a dose formulation or dosage form) suitable for administration (e.g., intravenous administration) to a subject as described herein.
  • a formulation e.g., a dose formulation or dosage form
  • the formulation described herein can be a liquid formulation, a lyophilized formulation, or a reconstituted formulation.
  • the formulation is a liquid formulation.
  • the formulation e.g., liquid formulation
  • the formulation comprises an anti-GITR antibody molecule (e.g., an anti-GITR antibody molecule described herein) and a buffering agent.
  • the formulation (e.g., liquid formulation) comprises an anti-GITR antibody molecule present at a concentration of 25 mg/mL to 250 mg/mL, e.g., 50 mg/mL to 200 mg/mL, 60 mg/mL to 180 mg/mL, 70 mg/mL to 150 mg/mL, 80 mg/mL to 120 mg/mL, 90 mg/mL to 110 mg/mL, 50 mg/mL to 150 mg/mL, 50 mg/mL to 100 mg/mL, 150 mg/mL to 200 mg/mL, or 100 mg/mL to 200 mg/mL, e.g., 50 mg/mL, 60 mg/mL, 70 mg/mL, 80 mg/mL, 90 mg/mL, 100 mg/mL, 110 mg/mL, 120 mg/mL, 130 mg/mL, 140 mg/mL, or 150 mg/mL.
  • the anti-GITR antibody molecule is present at a concentration of 80 mg/mL
  • the formulation (e.g., liquid formulation) comprises a buffering agent comprising histidine (e.g., a histidine buffer).
  • the buffering agent e.g., histidine buffer
  • the buffering agent is present at a concentration of 1 mM to 100 mM, e.g., 2 mM to 50 mM, 5 mM to 40 mM, 10 mM to 30 mM, 15 to 25 mM, 5 mM to 40 mM, 5 mM to 30 mM, 5 mM to 20 mM, 5 mM to 10 mM, 40 mM to 50 mM, 30 mM to 50 mM, 20 mM to 50 mM, 10 mM to 50 mM, or 5 mM to 50 mM, e.g., 2 mM, 5 mM, 10 mM, 15 mM, 20 mM, 25 mM, 30 mM, 35 mM,
  • the buffering agent e.g., histidine buffer
  • the buffering agent is present at a concentration of 15 mM to 25 mM, e.g., 20 mM.
  • the buffering agent e.g., a histidine buffer
  • the buffering agent e.g., histidine buffer
  • the buffering agent comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5). In certain embodiments, the buffering agent comprises histidine and histidine-HCl at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., a pH of 5.5).
  • the formulation (e.g., liquid formulation) comprises an anti-GITR antibody molecule present at a concentration of 80 to 120 mg/mL, e.g., 100 mg/mL; and a buffering agent that comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5).
  • the formulation (e.g., liquid formulation) further comprises a
  • the carbohydrate is sucrose.
  • the carbohydrate (e.g., sucrose) is present at a concentration of 50 mM to 500 mM, e.g., 100 mM to 400 mM, 150 mM to 300 mM, 180 mM to 250 mM, 200 mM to 240 mM, 210 mM to 230 mM, 100 mM to 300 mM, 100 mM to 250 mM, 100 mM to 200 mM, 100 mM to 150 mM, 300 mM to 400 mM, 200 mM to 400 mM, or 100 mM to 400 mM, e.g., 100 mM, 150 mM, 180 mM, 200 mM, 220 mM, 250 mM, 300 mM, 350 mM, or 400 mM.
  • the formulation comprises a carbohydrate or sucrose present at a concentration of 200 mM to 250 mM, e.g., 220 mM. In certain embodiments, the formulation comprises sucrose present at a concentration of 200 mM to 250 mM, e.g., 220 mM.
  • the formulation (e.g., liquid formulation) comprises an anti-GITR antibody molecule present at a concentration of 80 to 120 mg/mL, e.g., 100 mg/mL; a buffering agent that comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5); and a carbohydrate or sucrose present at a concentration of 200 mM to 250 mM, e.g., 220 mM.
  • a buffering agent that comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5)
  • a carbohydrate or sucrose present at a concentration of 200 mM to 250 mM, e.g., 220 mM.
  • the formulation (e.g., liquid formulation) further comprises a surfactant.
  • the surfactant is polysorbate 20.
  • the surfactant or polysorbate 20) is present at a concentration of 0.005 % to 0.1% (w/w), e.g., 0.01% to 0.08%, 0.02% to 0.06%, 0.03% to 0.05%, 0.01% to 0.06%, 0.01% to 0.05%, 0.01% to 0.03%, 0.06% to 0.08%, 0.04% to 0.08%, or 0.02% to 0.08% (w/w), e.g., 0.01%, 0.02%, 0.03%, 0.04%, 0.05%, 0.06%, 0.07%, 0.08%, 0.09%, or 0.1% (w/w).
  • the formulation comprises a surfactant or polysorbate 20 present at a concentration of 0.03% to 0.05%, e.g., 0.04% (w/w). In some embodiments, the formulation comprises polysorbate 20 present at a concentration of 0.03% to 0.05%, e.g., 0.04% (w/w).
  • the formulation (e.g., liquid formulation, e.g., for intravenous infusion) comprises an anti-GITR antibody molecule present at a concentration of 100 to 200 mg/mL, e.g., 150 mg/mL; a buffering agent that comprises a histidine buffer (e.g., histidine/histidine-HCL) at a concentration of 20 mM) and has a pH of 5.5; a carbohydrate or sucrose present at a concentration of 220 mM; and a surfactant or polysorbate 20 present at a concentration of 0.04% (w/w).
  • a histidine buffer e.g., histidine/histidine-HCL
  • a surfactant or polysorbate 20 present at a concentration of 0.04% (w/w).
  • the formulation (e.g., liquid formulation, e.g., for intravenous infusion) comprises an anti-GITR antibody molecule present at a concentration of 150 mg/mL; a buffering agent that comprises a histidine buffer (e.g., histidine/histidine-HCL) at a concentration of 20 mM) and has a pH of 5.5; a carbohydrate or sucrose present at a concentration of 220 mM; and a surfactant or polysorbate 20 present at a concentration of 0.04% (w/w).
  • a histidine buffer e.g., histidine/histidine-HCL
  • a surfactant or polysorbate 20 present at a concentration of 0.04% (w/w).
  • the formulation (e.g., liquid formulation) comprises an anti-GITR antibody molecule present at a concentration of 80 to 120 mg/mL, e.g., 100 mg/mL; a buffering agent that comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5); a carbohydrate or sucrose present at a concentration of 200 mM to 250 mM, e.g., 220 mM; and a surfactant or polysorbate 20 present at a concentration of 0.03% to 0.05%, e.g., 0.04% (w/w).
  • a buffering agent that comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5)
  • a carbohydrate or sucrose present at a concentration of 200 m
  • the formulation (e.g., liquid formulation) comprises an anti-GITR antibody molecule present at a concentration of 100 mg/mL; a buffering agent that comprises a histidine buffer (e.g., histidine/histidine-HCL) at a concentration of 20 mM) and has a pH of 5.5; a carbohydrate or sucrose present at a concentration of 220 mM; and a surfactant or polysorbate 20 present at a
  • the liquid formulation is prepared by diluting a formulation comprising an anti-GITR antibody molecule described herein.
  • a drug substance formulation can be diluted with a solution comprising one or more excipients (e.g., concentrated excipients).
  • the solution comprises one, two, or all of histidine, sucrose, or polysorbate 20.
  • the solution comprises the same excipient(s) as the drug substance formulation.
  • excipients include, but are not limited to, an amino acid (e.g., histidine), a carbohydrate (e.g., sucrose), or a surfactant (e.g., polysorbate 20).
  • the liquid formulation is not a reconstituted lyophilized formulation. In other embodiments, the liquid formulation is a reconstituted lyophilized formulation. In some embodiments, the formulation is stored as a liquid. In other embodiments, the formulation is prepared as a liquid and then is dried, e.g., by lyophilization or spray-drying, prior to storage.
  • 0.5 mL to 10 mL e.g., 0.5 mL to 8 mL, 1 mL to 6 mL, or 2 mL to 5 mL, e.g., 1 mL, 1.2 mL, 1.5 mL, 2 mL, 3 mL, 4 mL, 4.5 mL, or 5 mL
  • 0.5 mL to 10 mL e.g., 0.5 mL to 8 mL, 1 mL to 6 mL, or 2 mL to 5 mL, e.g., 1 mL, 1.2 mL, 1.5 mL, 2 mL, 3 mL, 4 mL, 4.5 mL, or 5 mL
  • container e.g., vial
  • the liquid formulation is filled into a container (e.g., vial) such that an extractable volume of at least 1 mL (e.g., at least 1.2 mL, at least 1.5 mL, at least 2 mL, at least 3 mL, at least 4 mL, or at least 5 mL) of the liquid formulation can be withdrawn per container (e.g., vial).
  • the liquid formulation is extracted from the container (e.g., vial) without diluting at a clinical site.
  • the liquid formulation is diluted from a drug substance formulation and extracted from the container (e.g., vial) at a clinical site.
  • the formulation e.g., liquid formulation
  • the formulation is injected to an infusion bag, e.g., within 1 hour (e.g., within 45 minutes, 30 minutes, or 15 minutes) before the infusion starts to the patient.
  • a formulation described herein can be stored in a container.
  • the container used for any of the formulations described herein can include, e.g., a vial, and optionally, a stopper, a cap, or both.
  • the vial is a glass vial, e.g., a 6R white glass vial.
  • the stopper is a rubber stopper, e.g., a grey rubber stopper.
  • the cap is a flip-off cap, e.g., an aluminum flip-off cap.
  • the container comprises a 6R white glass vial, a grey rubber stopper, and an aluminum flip-off cap.
  • the container e.g., vial
  • the container is for a single-use container.
  • 25 mg/mL to 250 mg/mL e.g., 50 mg/mL to 200 mg/mL, 60 mg/mL to 180 mg/mL, 70 mg/mL to 150 mg/mL, 80 mg/mL to 120 mg/mL, 90 mg/mL to 110 mg/mL, 50 mg/mL to 150 mg/mL, 50 mg/mL to 100 mg/mL, 150 mg/mL to 200 mg/mL, or 100 mg/mL to 200 mg/mL, e.g., 50 mg/mL, 60 mg/mL, 70 mg/mL, 80 mg/mL, 90 mg/mL, 100 mg/mL, 110 mg/mL, 120 mg/mL, 130 mg/mL, 140 mg/mL, 150 mg/mL, 160 mg/mL, 170 mg/mL, 180 mg/mL, 190 mg/mL,
  • the formulation is a lyophilized formulation.
  • the lyophilized formulation is lyophilized or dried from a liquid formulation comprising an anti-GITR antibody molecule described herein.
  • a liquid formulation comprising an anti-GITR antibody molecule described herein.
  • 1 to 5 mL, e.g., 1 to 2 mL, of a liquid formulation can be filled per container (e.g., vial) and lyophilized.
  • the formulation is a reconstituted formulation.
  • the reconstituted formulation is reconstituted from a lyophilized formulation comprising an anti-GITR antibody molecule described herein.
  • a reconstituted formulation can be prepared by dissolving a lyophilized formulation in a diluent such that the protein is dispersed in the reconstituted formulation.
  • the lyophilized formulation is reconstituted with 1 mL to 5 mL, e.g., 1 mL to 2 mL, e.g., 1.2 mL, of water or buffer for injection.
  • the lyophilized formulation is reconstituted with 1 mL to 2 mL of water for injection, e.g., at a clinical site.
  • the reconstituted formulation comprises an anti-GITR antibody molecule (e.g., an anti-GITR antibody molecule described herein) and a buffering agent.
  • an anti-GITR antibody molecule e.g., an anti-GITR antibody molecule described herein
  • a buffering agent e.g., an anti-GITR antibody molecule described herein
  • the reconstituted formulation comprises an anti-GITR antibody molecule present at a concentration of 25 mg/mL to 250 mg/mL, e.g., 50 mg/mL to 200 mg/mL, 60 mg/mL to 180 mg/mL, 70 mg/mL to 150 mg/mL, 80 mg/mL to 120 mg/mL, 90 mg/mL to 110 mg/mL, 50 mg/mL to 150 mg/mL, 50 mg/mL to 100 mg/mL, 150 mg/mL to 200 mg/mL, or 100 mg/mL to 200 mg/mL, e.g., 50 mg/mL, 60 mg/mL, 70 mg/mL, 80 mg/mL, 90 mg/mL, 100 mg/mL, 110 mg/mL, 120 mg/mL, 130 mg/mL, 140 mg/mL, or 150 mg/mL.
  • the anti-GITR antibody molecule is present at a concentration of 80 mg/mL to 120 mg/mL
  • the reconstituted formulation comprises a buffering agent comprising histidine (e.g., a histidine buffer).
  • the buffering agent e.g., histidine buffer
  • the buffering agent is present at a concentration of 1 mM to 100 mM, e.g., 2 mM to 50 mM, 5 mM to 40 mM, 10 mM to 30 mM, 15 to 25 mM, 5 mM to 40 mM, 5 mM to 30 mM, 5 mM to 20 mM, 5 mM to 10 mM, 40 mM to 50 mM, 30 mM to 50 mM, 20 mM to 50 mM, 10 mM to 50 mM, or 5 mM to 50 mM, e.g., 2 mM, 5 mM, 10 mM, 15 mM, 20 mM, 25 mM, 30 mM, 35 mM, 40 mM,
  • the buffering agent e.g., histidine buffer
  • the buffering agent is present at a concentration of 15 mM to 25 mM, e.g., 20 mM.
  • the buffering agent e.g., a histidine buffer
  • the buffering agent e.g., histidine buffer
  • the buffering agent comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5).
  • the buffering agent comprises histidine and histidine-HCl.
  • the reconstituted formulation comprises an anti-GITR antibody molecule present at a concentration of 80 to 120 mg/mL, e.g., 100 mg/mL; and a buffering agent that comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5).
  • the reconstituted formulation further comprises a carbohydrate.
  • the carbohydrate is sucrose.
  • the carbohydrate (e.g., sucrose) is present at a concentration of 50 mM to 500 mM, e.g., 100 mM to 400 mM, 150 mM to 300 mM, 180 mM to 250 mM, 200 mM to 240 mM, 210 mM to 230 mM, 100 mM to 300 mM, 100 mM to 250 mM, 100 mM to 200 mM, 100 mM to 150 mM, 300 mM to 400 mM, 200 mM to 400 mM, or 100 mM to 400 mM, e.g., 100 mM, 150 mM, 180 mM, 200 mM, 220 mM, 250 mM, 300 mM, 350 mM, or 400 mM.
  • the carbohydrate is sucrose.
  • the carbohydrate
  • the reconstituted formulation comprises an anti-GITR antibody molecule present at a concentration of 80 to 120 mg/mL, e.g., 100 mg/mL; a buffering agent that comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5); and a carbohydrate or sucrose present at a concentration of 200 mM to 250 mM, e.g., 220 mM.
  • a buffering agent that comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5)
  • a carbohydrate or sucrose present at a concentration of 200 mM to 250 mM, e.g., 220 mM.
  • the reconstituted formulation further comprises a surfactant.
  • the surfactant is polysorbate 20.
  • the surfactant or polysorbate 20) is present at a concentration of 0.005 % to 0.1% (w/w), e.g., 0.01% to 0.08%, 0.02% to 0.06%, 0.03% to 0.05%, 0.01% to 0.06%, 0.01% to 0.05%, 0.01% to 0.03%, 0.06% to 0.08%, 0.04% to 0.08%, or 0.02% to 0.08% (w/w), e.g., 0.01%, 0.02%, 0.03%, 0.04%, 0.05%, 0.06%, 0.07%, 0.08%, 0.09%, or 0.1% (w/w).
  • the formulation comprises a surfactant or polysorbate 20 present at a concentration of 0.03% to 0.05%, e.g., 0.04% (w/w).
  • the reconstituted formulation comprises an anti-GITR antibody molecule present at a concentration of 80 to 120 mg/mL, e.g., 100 mg/mL; a buffering agent that comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5); a carbohydrate or sucrose present at a concentration of 200 mM to 250 mM, e.g., 220 mM; and a surfactant or polysorbate 20 present at a concentration of 0.03% to 0.05%, e.g., 0.04% (w/w).
  • a buffering agent that comprises a histidine buffer at a concentration of 15 mM to 25 mM (e.g., 20 mM) and has a pH of 5 to 6 (e.g., 5.5)
  • a carbohydrate or sucrose present at a concentration of 200 mM to 250 mM
  • the reconstituted formulation comprises an anti-GITR antibody molecule present at a concentration of 100 mg/mL; a buffering agent that comprises a histidine buffer (e.g., histidine/histidine-HCL) at a concentration of 20 mM) and has a pH of 5.5; a carbohydrate or sucrose present at a concentration of 220 mM; and a surfactant or polysorbate 20 present at a concentration of 0.04% (w/w).
  • a histidine buffer e.g., histidine/histidine-HCL
  • a carbohydrate or sucrose present at a concentration of 220 mM
  • a surfactant or polysorbate 20 present at a concentration of 0.04% (w/w).
  • the formulation is reconstituted such that an extractable volume of at least 1 mL (e.g., at least 1.2 mL, 1.5 mL, 2 mL, 2.5 mL, or 3 mL) of the reconstituted formulation can be withdrawn from the container (e.g., vial) containing the reconstituted formulation.
  • the formulation is reconstituted and/or extracted from the container (e.g., vial) at a clinical site.
  • the formulation e.g., reconstituted formulation
  • exemplary buffering agents that can be used in the formulation described herein include, but are not limited to, an arginine buffer, a citrate buffer, or a phosphate buffer.
  • exemplary carbohydrates that can be used in the formulation described herein include, but are not limited to, trehalose, mannitol, sorbitol, or a combination thereof.
  • the formulation described herein may also contain a tonicity agent, e.g., sodium chloride, and/or a stabilizing agent, e.g., an amino acid (e.g., glycine, arginine, methionine, or a combination thereof).
  • 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 (e.g., 30 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 10mg/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 antibody is infused over a period of about 30 min.
  • 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.
  • 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.
  • an antibody molecule can be orally administered, for example, with an inert diluent or an assimilable edible carrier.
  • the compound (and other ingredients, if desired) may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet.
  • the compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • To administer a compound of the invention by other than parenteral administration it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation.
  • Therapeutic compositions can also be administered with medical devices known in the art.
  • 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.
  • 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.
  • compositions are preferably manufactured under GMP conditions.
  • 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
  • Dosage regimens are adjusted to provide the desired response (e.g., a therapeutic response).
  • 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.
  • 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.
  • 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.
  • 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 10mg/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 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 cytokine, for example, IL-7, IL-12 or IL-15.
  • 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 cytokine
  • 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).
  • 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., mechlor
  • 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., allylamine
  • the anti-GITR compositions of the present invention can be provided in a kit, e.g., for use in accordance with a dosage regimen described herein.
  • 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, e.g., in accordance with dosage regimens described herein. 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,
  • 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
  • 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, rhino
  • immunodeficiency virus type I HIV I
  • human immunodeficiency virus type II HAV II
  • any picornaviridae enteroviruses, caliciviridae, any of the Norwalk group of viruses, togaviruses, such as alphaviruses, flaviviruses, coronaviruses, rabies virus, Marburg viruses, ebola viruses, parainfluenza virus, orthomyxoviruses, bunyaviruses, arenaviruses, reoviruses, rotaviruses, orbiviruses, human T cell leukemia virus type I, human T cell leukemia virus type II, simian immunodeficiency virus, lentiviruses, polyomaviruses, parvoviruses, Epstein Barr virus, human herpesvirus 6, cercopithecine herpes virus 1 (B virus), and poxviruses.
  • togaviruses such as alphaviruses, flaviviruses, corona
  • 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, e.g., in accordance with a dosage regimen described herein.
  • 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.
  • 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.
  • 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.
  • 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.
  • inhibitortion 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, or 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 in accordance with a dosage regimen described herein 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-L1nhibitor In another embodiment, the agonist of GITR, , e.g., the anti-GITR antibody molecule, is administered in combination with a LAG-3 inhibitor, e.g., an anti-LAG-3 antibody molecule. In another embodiment, 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 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).
  • CEACAM e.g., CEACAM-1, -3 and/or -5
  • 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 e.g., CEACAM-1, -3 and/or -5 is an antibody molecule to CEACAM (e.g., CEACAM-1, -3 and/or -5).
  • CEACAM e.g., CEACAM-1, -3 and/or -5
  • the CEACAM 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. Additional Combination Therapies
  • 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
  • the heavy chain CDR1 comprises SEQ ID NO:22, and
  • the heavy chain CDR2 comprises a sequence selected from any one of
  • the heavy chain CDR3 comprises SEQ ID NO:29 or SEQ ID NO:109; and (b) a light chain variable region, wherein
  • the light chain CDR1 comprises SEQ ID NO:30 or SEQ ID NO:31
  • the light chain CDR2 comprises SEQ ID NO:33
  • the light chain CDR3 comprises SEQ ID NO:34.
  • any of the combinations disclosed herein further includes one or more of the agents described in Table 7.
  • 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, 150mg, 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.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne des régimes posologiques et des compositions d'anticorps, comprenant, par exemple, des anticorps, des anticorps modifiés et des fragments d'anticorps qui se lient à un membre de la superfamille du récepteur du facteur de nécrose tumorale (c'est-à-dire, 18), et des compositions comprenant un ou plusieurs agents thérapeutiques supplémentaires. Les compositions de l'invention sont utiles dans l'amélioration des réponses de lymphocytes T CD4+ et CD8+, et dans le traitement, l'atténuation et la prévention de maladies qui peuvent être contrecarrées par une réponse immunitaire accrue, par exemple, des cancers. L'invention concerne également des procédés d'utilisation de combinaisons qui trouvent une utilisation dans le traitement ou la prévention d'affections et de troubles cancéreux ou infectieux.
PCT/US2018/042882 2017-07-21 2018-07-19 Régimes posologiques pour anticorps anti-gitr et utilisations associées WO2019018640A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201762535771P 2017-07-21 2017-07-21
US62/535,771 2017-07-21
US201762611702P 2017-12-29 2017-12-29
US62/611,702 2017-12-29

Publications (1)

Publication Number Publication Date
WO2019018640A1 true WO2019018640A1 (fr) 2019-01-24

Family

ID=63104148

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/042882 WO2019018640A1 (fr) 2017-07-21 2018-07-19 Régimes posologiques pour anticorps anti-gitr et utilisations associées

Country Status (1)

Country Link
WO (1) WO2019018640A1 (fr)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10472419B2 (en) 2014-01-31 2019-11-12 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
US10752687B2 (en) 2014-01-24 2020-08-25 Novartis Ag Antibody molecules to PD-1 and uses thereof
WO2022060831A1 (fr) * 2020-09-17 2022-03-24 Mirati Therapeutics, Inc. Polythérapies
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
WO2022140788A1 (fr) * 2020-12-23 2022-06-30 Spark Therapeutic, Inc. Procédés pour améliorer la thérapie génique non virale
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation
WO2023122062A1 (fr) * 2021-12-21 2023-06-29 Cornell University Procédés d'administration améliorée d'acide nucléique
EP4085074A4 (fr) * 2020-01-02 2024-05-15 Nanjing Genscript Biotech Co Ltd Anticorps anti-gitr et leurs utilisations

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016057841A1 (fr) * 2014-10-08 2016-04-14 Novartis Ag Compositions et procédés d'utilisation pour une réponse immunitaire accrue et une thérapie anticancéreuse
WO2017106656A1 (fr) * 2015-12-17 2017-06-22 Novartis Ag Molécules d'anticorps anti-pd-1 et leurs utilisations

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016057841A1 (fr) * 2014-10-08 2016-04-14 Novartis Ag Compositions et procédés d'utilisation pour une réponse immunitaire accrue et une thérapie anticancéreuse
WO2017106656A1 (fr) * 2015-12-17 2017-06-22 Novartis Ag Molécules d'anticorps anti-pd-1 et leurs utilisations

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CLINICALTRIALS.GOV: "Study of MK-4166 and MK-4166 in Combination With Pembrolizumab (MK-3475) in Participants With Advanced Solid Tumors (MK-4166-001) - Tabular View - ClinicalTrials.gov", 7 May 2014 (2014-05-07), XP055501245, Retrieved from the Internet <URL:https://clinicaltrials.gov/ct2/show/record/NCT02132754> [retrieved on 20180822] *
HENRY KOON: "First in human Phase 1 single-dose study of TRX-518, an anti-human glucocorticoid-induced tumor necrosis factor receptor (GITR) monoclonal antibody in adults with advanced solid tumors", ASCO ANNUAL MEETING 2016, 3 June 2016 (2016-06-03), XP055501255, Retrieved from the Internet <URL:https://www.leaptx.com/file.cfm/17/docs/TRX518%20Single%20Dose%20Solid%20Tumors%20ASCO%202016.pdf> [retrieved on 20180822], DOI: https://www.leaptx.com/file.cfm/17/docs/TRX518%20Single%20Dose%20Solid%20Tumors%20ASCO%202016.pdf *
KIM C. OHAEGBULAM ET AL: "Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway", TRENDS IN MOLECULAR MEDICINE, vol. 21, no. 1, 1 January 2015 (2015-01-01), GB, pages 24 - 33, XP055249717, ISSN: 1471-4914, DOI: 10.1016/j.molmed.2014.10.009 *
KNEE DEBORAH A ET AL: "Rationale for anti-GITR cancer immunotherapy", EUROPEAN JOURNAL OF CANCER, ELSEVIER, AMSTERDAM, NL, vol. 67, 31 August 2016 (2016-08-31), pages 1 - 10, XP029758970, ISSN: 0959-8049, DOI: 10.1016/J.EJCA.2016.06.028 *
SUKUMAR SELVAKUMAR ET AL: "Characterization of MK-4166, a Clinical Agonistic Antibody That Targets Human GITR and Inhibits the Generation and Suppressive Effects of T Regulatory Cells", CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 77, no. 16, 13 June 2017 (2017-06-13), pages 4378 - 4388, XP009507381, ISSN: 0008-5472, DOI: 10.1158/0008-5472.CAN-16-1439 *

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11708412B2 (en) 2013-09-26 2023-07-25 Novartis Ag Methods for treating hematologic cancers
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
US10752687B2 (en) 2014-01-24 2020-08-25 Novartis Ag Antibody molecules to PD-1 and uses thereof
US11827704B2 (en) 2014-01-24 2023-11-28 Novartis Ag Antibody molecules to PD-1 and uses thereof
US10981990B2 (en) 2014-01-31 2021-04-20 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US11155620B2 (en) 2014-01-31 2021-10-26 Novartis Ag Method of detecting TIM-3 using antibody molecules to TIM-3
US10472419B2 (en) 2014-01-31 2019-11-12 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation
EP4085074A4 (fr) * 2020-01-02 2024-05-15 Nanjing Genscript Biotech Co Ltd Anticorps anti-gitr et leurs utilisations
WO2022060831A1 (fr) * 2020-09-17 2022-03-24 Mirati Therapeutics, Inc. Polythérapies
WO2022140788A1 (fr) * 2020-12-23 2022-06-30 Spark Therapeutic, Inc. Procédés pour améliorer la thérapie génique non virale
WO2023122062A1 (fr) * 2021-12-21 2023-06-29 Cornell University Procédés d'administration améliorée d'acide nucléique

Similar Documents

Publication Publication Date Title
US20220153835A1 (en) Combination therapies comprising antibody molecules to lag-3
US20170306038A1 (en) Compositions and methods of use for augmented immune response and cancer therapy
US20220133889A1 (en) Combination therapies comprising antibody molecules to tim-3
CN108025051B (zh) 包含抗pd-1抗体分子的联合疗法
US20210284737A1 (en) Antibody molecules to pd-l1 and uses thereof
EP3389712B1 (fr) Molécules d&#39;anticorps anti-pd-1 et leurs utilisations
WO2019018640A1 (fr) Régimes posologiques pour anticorps anti-gitr et utilisations associées
RU2788092C2 (ru) Молекулы антител к pd-1 и их применения

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18750055

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18750055

Country of ref document: EP

Kind code of ref document: A1