WO2021227326A1 - Compositions et méthodes pour le traitement du cancer - Google Patents

Compositions et méthodes pour le traitement du cancer Download PDF

Info

Publication number
WO2021227326A1
WO2021227326A1 PCT/CN2020/115795 CN2020115795W WO2021227326A1 WO 2021227326 A1 WO2021227326 A1 WO 2021227326A1 CN 2020115795 W CN2020115795 W CN 2020115795W WO 2021227326 A1 WO2021227326 A1 WO 2021227326A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
amino acid
seq
cancer
subject
Prior art date
Application number
PCT/CN2020/115795
Other languages
English (en)
Inventor
Peter Peizhi Luo
Guizhong Liu
Changhua GONG
Original Assignee
Adagene 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 Adagene Ag filed Critical Adagene Ag
Priority to US17/924,540 priority Critical patent/US20230133118A1/en
Priority to CN202180048790.9A priority patent/CN115768525A/zh
Priority to PCT/CN2021/093511 priority patent/WO2021228178A1/fr
Priority to TW110117374A priority patent/TW202200622A/zh
Priority to CA3177717A priority patent/CA3177717A1/fr
Priority to AU2021271971A priority patent/AU2021271971A1/en
Priority to EP21803953.5A priority patent/EP4149547A1/fr
Priority to JP2022568397A priority patent/JP2023525085A/ja
Publication of WO2021227326A1 publication Critical patent/WO2021227326A1/fr

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • 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
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2299/00Coordinates from 3D structures of peptides, e.g. proteins or enzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the present application is in the field of cancer therapeutics, and relates to compositions and methods for treating cancers using antibodies that bind to human CD137.
  • CD137 (also referred to as CD137 receptor, 4-1BB, TNFRSF9, etc. ) is a transmembrane protein of the Tumor Necrosis Factor Receptor Superfamily (TNFRS) .
  • TNFSF Tumor Necrosis Factor Receptor Superfamily
  • Current understanding of CD137 indicates that its expression is generally activation dependent and is present in a broad subset of immune cells including activated NK and NKT cells, regulatory T cells, dendritic cells (DC) , stimulated mast cells, differentiating myeloid cells, monocytes, neutrophils, and eosinophils (Wang, 2009, Immunological Reviews 229: 192-215) .
  • CD137 expression has also been demonstrated on tumor vasculature (Broll, 2001, Amer. J. Clin. Pathol.
  • CD137L CD137 Ligand
  • CD137 promotes enhanced cellular proliferation, survival, and cytokine production (Croft, 2009, Nat Rev Immunol 9: 271-285) .
  • CD137 monotherapy and combination therapy tumor models have established durable anti-tumor protective T cell memory responses (Lynch, 2008, Immunol Rev. 22: 277-286) .
  • CD137 agonists also have been shown to inhibit autoimmune reactions in a variety of art-recognized autoimmunity models (Vinay, 2006, J Mol Med 84: 726-736) .
  • This dual activity of CD137 offers the potential to provide anti-tumor activity while dampening autoimmune side effects that can be associated with immunotherapy approaches that break immune tolerance.
  • the present application provides methods for treating cancer with an anti-CD137 antibody, and biomarkers (e.g., prognostic biomarkers) for the methods described herein.
  • biomarkers e.g., prognostic biomarkers
  • the present invention in one aspect provides a method of treating a cancer in a subject, comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues within amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1, and wherein the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • 500 mg e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg
  • One aspect of the present application provides a method of treating a cancer in a subject, comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues within amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1, and wherein the cancer is resistant or refractory to a prior therapy (e.g., a prior immunotherapy) .
  • the prior therapy is an anti-CD20 antibody.
  • the prior therapy is rituximab.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • One aspect of the present application provides a method of treating a cancer in a subject, comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and wherein the subject has a high level in one or more biomarkers selected from the group consisting of total CD137, membrane bound CD137 (mCD137) , CD137 ligand (CD137L) and PD-L1 and/or a low level of CD8+ effector memory T (T em ) cells or natural killer (NK) cells compared to a reference level.
  • an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the method further comprises administering to the subject an effective amount of an immune checkpoint inhibitor, such as an anti-PD-1 antibody (e.g., toripalimab) .
  • an immune checkpoint inhibitor such as an anti-PD-1 antibody (e.g., toripalimab) .
  • One aspect of the present application provides a method of treating a cancer in a subject, comprising: (a) administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and (b) subsequently determining a level of one or more biomarkers selected from the group consisting of total CD137, membrane bound (mCD137) , soluble CD137 (sCD137) , CD137L, Ki67, NK cells, CD8+ effector memory T (T em ) cells, regulatory T (T reg ) cells and NK cells in a sample of the subject.
  • an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consist
  • an increased level of one or more biomarkers selected from the group consisting of total CD137, sCD137, Ki67, CD137L, NK cells, and CD8 T em cells, and/or a decreased level of one or more biomarkers selected from the group consisting of mCD137 and T reg cells after administration of the anti-CD137 antibody compared to the level of the one or more biomarkers before administration of the anti-CD137 antibody indicates that the subject may benefit from the administration of the ant-CD137 antibody.
  • the sample has an increased level of one or more biomarkers selected from the group consisting of total CD137, sCD137, Ki67, CD137L, NK cells, and CD8 T em cells, and/or a decreased level of one or more biomarkers selected from the group consisting of mCD137 and T reg cells after administration of the anti-CD137 antibody compared to the level of the one or more biomarkers before administration of the anti-CD137 antibody, the method further comprises administering to the subject an effective amount of the anti-CD137 antibody.
  • the level of one or more biomarkers comprises a level of CD137 (e.g., in a tumor biopsy sample) .
  • the level of one or more biomarkers comprises a level of sCD137 in a blood sample, such as a plasma sample. In some embodiments, the level of one or more biomarkers comprises a level of mCD137 on CD8+T cells. In some embodiments, the level of one or more biomarkers comprises a level of Ki67 on CD8+ T cells. In some embodiments, the CD8+ T cells are tumor infiltrating T cells. In some embodiments, the level of one or more biomarkers comprises a level of CD137L (e.g., in a tumor biopsy sample) . In some embodiments, the level of one or more biomarkers comprises a level of NK cells in a blood sample.
  • the sample is a tumor biopsy sample.
  • the sample is a formalin-fixed paraffin-embedded (FFPE) sample.
  • the level of the one or more biomarkers is detected by immunohistochemistry (IHC) .
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the method further comprises administering to the subject an effective amount of an immune checkpoint inhibitor, such as an anti-PD-1 antibody (e.g., toripalimab) .
  • an immune checkpoint inhibitor such as an anti-PD-1 antibody (e.g., toripalimab) .
  • One aspect of the present application provides a method of providing a prognosis for a subject who has been administered with an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; the method comprising determining a level of one or more biomarkers selected from the group consisting of total CD137, membrane bound (mCD137) , soluble CD137 (sCD137) , CD137L, Ki67, CD8+ effector memory T (T em ) cells, regulatory T (T reg ) cells and NK cells in a sample of the subject, wherein an increased level of one or more biomarkers selected from the group consisting of total CD137, sCD137, Ki67, CD137L, CD8 T em cells, and NK cells, and/or a
  • the level of one or more biomarkers comprises a level of CD137 (e.g., in a tumor biopsy sample) .
  • the level of one or more biomarkers comprises a level of sCD137 in a blood sample (e.g., a plasma sample) .
  • the level of one or more biomarkers comprises a level of mCD137 on CD8+ T cells.
  • the level of one or more biomarkers comprises a level of Ki67 on CD8+ T cells.
  • the CD8+T cells are tumor infiltrating T cells.
  • the level of one or more biomarkers comprises a level of CD137L (e.g., in a tumor biopsy sample) .
  • the level of one or more biomarkers comprises a level of NK cells in a blood sample.
  • the sample is a tumor biopsy sample.
  • the sample is a formalin-fixed paraffin-embedded (FFPE) sample.
  • the level of the one or more biomarkers is detected by immunohistochemistry (IHC) .
  • the sample is a tumor biopsy sample.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the method further comprises administering to the subject an effective amount of an immune checkpoint inhibitor, such as an anti-PD-1 antibody (e.g., toripalimab) .
  • an immune checkpoint inhibitor such as an anti-PD-1 antibody (e.g., toripalimab) .
  • the cancer is a solid cancer.
  • the cancer is selected from the group consisting of colon cancer (e.g., Sigmoid colon cancer) , breast cancer, lung cancer (e.g., non-small cell lung cancer or NSCLC) , esophageal cancer, endometrial cancer, gastrointestinal cancer (e.g., gastrointestinal neuroectodermal tumor) , cholangiocarcinoma, nasopharyngeal cancer (NPC) , adenoid cystic carcinoma (ACC) , melanoma, mesothelioma (e.g., malignant pleural mesothelioma or MPM) , mantle cell lymphoma, anal cancer, head and neck cancer (e.g., head and neck squamous cell carcinoma or HNSCC) and appendiceal and sebaceous cancer.
  • colon cancer e.g., Sigmoid colon cancer
  • breast cancer e.g., breast cancer
  • the cancer is a liquid cancer. In some embodiments, the cancer is follicular lymphoma. In some embodiments, the cancer is non-Hodgkin’s lymphoma (NHL) . In some embodiments, the cancer is T cell lymphoma, such as angioimmunoblastic T-cell lymphoma (AITL) or Peripheral T-cell lymphoma (PTCL) .
  • AITL angioimmunoblastic T-cell lymphoma
  • PTCL Peripheral T-cell lymphoma
  • One aspect of the present application provides a method of treating a cancer in a subject, comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues within amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1, and wherein the cancer is selected from the group consisting of follicular lymphoma, T cell lymphoma, and ACC.
  • the cancer is follicular lymphoma.
  • the cancer is T cell lymphoma.
  • the cancer is AITL or PTCL.
  • the cancer is ACC.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • One aspect of the present application provides a method of treating a lung cancer in a subject, comprising administering to the subject: (a) an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and (b) an effective amount of an immune checkpoint inhibitor.
  • the immune checkpoint inhibitor is an anti-PD-L1 antibody (e.g., atezolizumab) .
  • the immune checkpoint inhibitor is an anti-PD-1 antibody (e.g., toripalimab) .
  • the immune checkpoint inhibitor is an anti-CTLA-4 antibody (e.g., ADG116) .
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • One aspect of the present application provides method of treating a breast cancer in a subject, comprising administering to the subject: (a) an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and (b) an effective amount of an immune checkpoint inhibitor.
  • the breast cancer is triple-negative breast cancer.
  • the immune checkpoint inhibitor is an anti-PD-L1 antibody (e.g., atezolizumab) .
  • the immune checkpoint inhibitor is an anti-PD-1 antibody (e.g., toripalimab) .
  • the immune checkpoint inhibitor is an anti-CTLA-4 antibody (e.g., ADG116) .
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • One aspect of the present application provides a method of treating a lung cancer in a subject, comprising administering to the subject: (a) an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and (b) an effective amount of a chemotherapeutic agent.
  • the chemotherapeutic agent is docetaxel.
  • the chemotherapeutic agent is cisplatin.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • One aspect of the present application provides a method of treating a breast cancer in a subject, comprising administering to the subject: (a) an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and (b) an effective amount of a chemotherapeutic agent.
  • the breast cancer is triple-negative breast cancer.
  • the chemotherapeutic agent is docetaxel.
  • the chemotherapeutic agent is cisplatin.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • One aspect of the present application provides a method of treating a lung cancer in a subject, comprising administering to the subject: (a) an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and (b) an effective amount of an anti-CD20 antibody.
  • the anti-CD20 antibody is rituximab.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • One aspect of the present application provides a method of treating a colon cancer in a subject, comprising administering to the subject: (a) an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and (b) an effective amount of a radiation therapy.
  • the radiation therapy is local radiation.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the anti-CD137 antibody is administered at a dose at a dose of about 300 mg to about 400 mg. In some embodiments, the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg. In some embodiments, the anti-CD137 antibody is administered at a dose of about 3 mg/kg to about 8 mg/kg. In some embodiments, the anti-CD137 antibody is administered at a dose of about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously. In some embodiments, the anti-CD137 antibody is administered about once every three weeks. In some embodiments, the subject receives at least 2 cycles of treatment with the anti-CD137 antibody.
  • the cancer is advanced-stage cancer.
  • the cancer is metastatic cancer.
  • the cancer is resistant or refractory to a prior therapy.
  • the prior therapy is selected from the group consisting of viral gene therapy, immunotherapy, targeted therapy, radiation therapy, and chemotherapy.
  • the anti-CD137 antibody is cross-reactive with a CD137 polypeptide from at least one non-human species selected from the group consisting of cynomolgus monkey, mouse, rat and dog.
  • the anti-CD137 antibody binds to amino acid residues 51, 63-67, 69-73, 83, 89, 92, 98-104 and 112-114 of SEQ ID NO: 1.
  • the anti-CD137 antibody comprises a heavy chain variable region (VH) and a light chain variable region (VL) , wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 2, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 3, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 4; and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 5, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 6, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 7.
  • VH heavy chain variable region
  • VL light chain variable region
  • the VH comprises the amino acid sequence of SEQ ID NO: 8
  • the VL comprises the amino acid sequence of SEQ ID NO: 9.
  • the antibody comprises a heavy chain and a light chain, and wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 10, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 11.
  • the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14; and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 15, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 17.
  • the VH comprises the amino acid sequence of SEQ ID NO: 18, and/or the VL comprises the amino acid sequence of SEQ ID NO: 19.
  • the antibody comprises a heavy chain and a light chain, wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 20, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 21.
  • the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24; and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 25, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 26, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 27.
  • the VH comprises the amino acid sequence of SEQ ID NO: 28, and/or the VL comprises the amino acid sequence of SEQ ID NO: 29.
  • the antibody comprises a heavy chain and a light chain, wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 30, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 31.
  • the anti-CD137 antibody comprises a human IgG4 Fc region.
  • the human IgG4 Fc region comprises an S241P mutation, wherein numbering is according to Kabat.
  • the subject is a human subject.
  • the method further comprises administering to the subject a therapeutically effective amount of at least one additional therapeutic agent.
  • the at least one additional therapeutic agent is selected from the group consisting of viral gene therapy, immune checkpoint inhibitors, targeted therapies, radiation therapies, and chemotherapies.
  • compositions, kits, and articles of manufacture for use in any one of the methods described herein.
  • FIGs. 1A-1D show the alignment of epitopes on CD137 bound by CD137 ligand, ADG 106, Utomilumab, and Urelumab.
  • FIG. 2 shows treatment duration and response of patients treated with 0.03, 0.1, 0.3, 1, 3, 5, or 10 mg/kg ADG106.
  • FIG. 3 shows percentage changes of target lesions in patients before and after treatment with ADG106.
  • Each line represents an individual patient.
  • red line denotes patient with slight tumor shrinkage after initial enlargement;
  • yellow lines denote other patients.
  • FIG. 4 shows PET CT images of a patient with follicular lymphoma.
  • the left panel shows a PET CT image before treatment; the right panel shows a PET CT image at week six after treatment.
  • FIG. 5 shows receptor occupancy of ADG106 at different dose levels.
  • FIG. 6A shows mean serum concentrations of ADG106 in treatment cycle 1 in patients dosed with 0.03, 0.1, 0.3, 1, 3, 5, or 10 mg/kg ADG106 in the U.S. clinical study.
  • FIG. 6B shows mean serum concentrations of ADG106 in treatment cycle 1 in patients dosed with 0.1, 0.5, 1.5, 3, 5, or 10 mg/kg ADG106 in the Chinese clinical study.
  • FIG. 7 shows mean serum concentrations of ADG106 in male and female cynomolgus monkeys after weekly repeat doses of 50, 100, or 200 mg/kg ADG 106.
  • FIG. 8A-8B show kinetics of Ki-67 expression on peripheral CD4+ and CD8+ T cells of patient R011 in stable disease with total dose of 170 mg vs the similar curve by Pembrolizumab with a total dose of 200mg from literature report.
  • FIG. 8A show kinetics of Ki-67 expression on peripheral CD4+ and CD8+ T cells from patient R011 dosed at 3mg/kg with a body weight of 57 kg, with a total dose of 170mg per cycle of ADG106.
  • FIG. 8B show kinetics of Ki-67 expression on peripheral CD4+ and CD8+ T cells from one patient treated with a total dose of 200mg per cycle of Pembrolizumab.
  • FIGS. 9A-9B show increased Ki-67 expression on peripheral CD4+ and CD8+ T cells of certain patients treated with high dose levels of ADG106 ranging from 0.5 mg/kg to 10mg/kg.
  • FIGS. 10A-10B show CD137 expression on CD4+ and CD8+ T cells of patients treated with ADG106 at different doses.
  • FIG. 11 shows soluble CD137 (sCD137) levels pre-dose (day 0) and post-dose (21, 42, 63, 84, 105, and 126 days) in patients treated with 1.5, 3, or 5 mg/kg ADG106 each cycle once every three weeks (Q3W) . It is apparent that the soluble CD137 (sCD137) is induced and increased significantly and then level off as a function of dosage from 1.5 and 5 mg/kg with the patients.
  • FIGS. 12A-12B show changes of CD137 levels in patients before and after receiving treatment with ADG106.
  • FIG. 12A shows the changes of soluble CD137 (sCD137) levels in patients pre-dose and post-dose.
  • FIG. 12B shows the changes of membrane-bound CD137 (mCD137) levels in patients pre-dose and post-dose.
  • P denotes progression of diseases
  • SX denotes stable disease and followed by stable or progressive diseases, abbreviated as “SS” or “SP” respectively.
  • FIG. 13 shows changes of Ki67+ CD8+ T cells levels in patients before and after receiving treatment with ADG106.
  • P denotes progression of diseases
  • SX denotes stable disease, and followed by stable or progressive diseases, abbreviated as “SS” or “SP” respectively.
  • SS stable disease
  • SP stable or progressive diseases
  • FIGS. 14A-14C show the levels of CD8+ effector memory T cells (T em ) pre-dose (C0) and changes of CD8+ T em levels post-dose (C1) in patients receiving ADG106 treatment.
  • FIG. 14A shows the pre-dose (C0) levels of CD8+ T em in patients that showed progression of disease (P) and patients that showed stable disease (SX) .
  • FIG. 14B shows the ratio of pre-dose (C0) and post-dose (C1) levels of CD8+ T em in patients that showed progression of disease (P) and patients that showed stable disease (SX) .
  • FIG. 14C shows the pre-dose (C0) and post-dose (C1) levels of CD8+ T em in patients that showed progression of disease (P) and patients that showed stable disease (SX) , and followed by stable or progressive diseases, abbreviated in SS or SP etc.
  • FIGS. 15A-15D show changes of Ki+CD8+ T cell percentages, membrane-bound CD137 levels on CD8+ T cells, CD4+ Treg percentages, CD8+ effector memory T cells T em in a patient (R017) , who achieved >30%tumor size reduction ranging from 16%to 57%in 6 targeted lesions after receiving one cycle of ADG106 treatment.
  • FIG. 15A shows changes of Ki+CD8+ T cell percentages through three treatment cycles.
  • FIG. 15B shows changes of membrane-bound CD137 levels on CD8+ T cells through three treatment cycles.
  • FIG. 15C shows changes of CD4+ Treg percentages through three treatment cycles.
  • FIG. 15D shows changes of CD8+ effector memory T cells Tem through three treatment cycles.
  • FIG. 16A-B show a target lesion by CT images before and after only one administration of ADG106 for patient with a stage III angioimmunoblastic T cell lymphoma.
  • FIG. 16A show PET CT images before treatment (left panels) .
  • FIG. 16B show CT images at cycle 2 day 21 after only one administration of ADG106 (right panels) .
  • LDi denotes longest diameter
  • SDi denotes shortest diameter.
  • FIGS. 17A-17D show tumor volumes in L5178-R and L5178-S murine T cell lymphoma syngeneic models treated with isotype control antibody or ADG106.
  • FIG. 17A shows tumor volumes up to 11 days after the start of treatment in L5178-R murine T cell lymphoma syngeneic model with IgG4 isotype control antibody or ADG106 at a dose of 20 mg/kg.
  • FIG. 17B shows the staining patterns of the CD137 ligand expression in L5178-R cells.
  • FIG. 17C shows tumor volume up to 23 days after the start of treatment in L5178-S murine T cell lymphoma syngeneic model with IgG4 isotype control antibody or ADG106 at dose of 20 mg/kg.
  • FIG. 17D shows the staining patterns of the CD137 ligand expression in L5178-S cells.
  • FIG. 18 shows ADG106 exposure in relation to total dose.
  • FIGs. 19A-19B show in vivo treatment effects of isotype control antibody, ADG106, (atezolizumab) , or a combination of ADG106 and (atezolizumab) in mouse 3LL lung cancer model in C57BL/6 mice.
  • the number of days post inoculation is shown on the x-axis, and the tumor volume in mm 3 is shown on the y-axis.
  • FIG. 19A shows tumor growth curves of different treatment groups. Data points represent group mean, and error bars represent SEM.
  • FIG. 19B shows tumor growth curves of individual mice in each tested group.
  • FIGs. 20A-20B show in vivo treatment effects of vehicle, ADG106, an anti-PD-1 antibody, or a combination of ADG106 and anti-PD-1 antibody in mouse Lewis lung cancer model in C57BL/6 mice.
  • FIG. 20A shows tumor growth curves of different treatment groups. Data points represent group mean, and error bars represent SEM.
  • FIG. 20B shows tumor growth curves of individual mice in each tested group.
  • FIGs. 21A-21B show in vivo treatment effects of isotype control antibody, ADG106, ADG116 (anti-CTLA-4 antibody) , or a combination of ADG106 and ADG116 in mouse 4T1 breast cancer model in C57BL/6 mice.
  • FIG. 21A shows tumor growth curves of different treatment groups. Data points represent group mean, and error bars represent SEM.
  • FIG. 21B shows tumor growth curves of individual mice in each tested group.
  • FIGs. 22A-22B show in vivo treatment effects of vehicle, ADG106, docetaxel, or a combination of ADG106 and docetaxel in mouse 4T1 breast cancer model in C57BL/6 mice.
  • FIG. 22A shows tumor growth curves of different treatment groups. Data points represent group mean, and error bars represent SEM.
  • FIG. 22B shows tumor growth curves of individual mice in each tested group.
  • FIGs. 23A-23B show in vivo treatment effects of vehicle, ADG106, cisplatin, or a combination of ADG106 and cisplatin in mouse Lewis lung cancer model in C57BL/6 mice.
  • FIG. 23A shows tumor growth curves of different treatment groups. Data points represent group mean, and error bars represent SEM.
  • FIG. 23B shows tumor growth curves of individual mice in each tested group.
  • FIGs. 24A-24B show in vivo treatment effects of isotype control antibody, ADG106, anti-CD20 antibody (Rituximab) , or a combination of ADG106 and Rituximab in a mouse Lewis lung cancer model stably transfected with human CD20 in C57BL/6 mice.
  • the number of days post inoculation is shown on the x-axis, and the volume of the tumor in mm 3 is shown on the y-axis.
  • FIG. 24A shows tumor growth curves of different treatment groups. Data points represent group mean, and error bars represent SEM.
  • FIG. 24B shows tumor growth curves of individual mice in each tested group.
  • FIGs. 25A-25B show in vivo treatment effects of isotype control antibody, local radiation, ADG106, or a combination of local radiation and ADG106 in mouse MC38 colon cancer model in C57BL/6 mice.
  • the number of days after the start of treatment is shown on the x-axis, and the volume of the tumor in mm 3 is shown on the y-axis.
  • FIG. 25A shows tumor growth curves of different treatment groups. Data points represent group mean, and error bars represent SEM.
  • FIG. 25B shows tumor growth curves of individual mice in each tested group.
  • FIGs. 26A-26B show changes of NK cell percentages in patients treated with ADG106.
  • FIG. 26A shows NK cell percentages before and after ADG106 treatment.
  • FIG. 26B shows changes of NK cell percentages in patients dosed with 0.1, 0.5, 1.5, 3, 5, or 10 mg/kg ADG106.
  • FIGs. 27A-27B show comparison of the base line NK cell percentages and changes in NK cell percentages in patients whose cancer progressed (P) versus patients with stable disease (S) after treatment of ADG106.
  • FIG. 27A shows baseline base line NK cell percentages in patients whose cancer progressed versus patients with stable disease after treatment of ADG106.
  • FIG. 27B shows changes in NK cell percentages in patients whose cancer progressed versus patients with stable disease after treatment of ADG106.
  • FIGs. 28A-28B show correlations of sCD137 serum level in nasopharyngeal cancer (NPC) patients who showed progression of disease (P) , stable disease (S) , or partial response (PR) .
  • FIG. 28A shows changes of sCD137 expression levels, which are expressed as ratios of induced changes in soluble CD137 expression levels after treatment (C2-C1) to baseline sCD137 expression levels before treatment (C1) in patients who showed progression of disease (P) , stable disease (S) , or partial response (PR) after treatment of ADG106.
  • C2-C1 induced changes in soluble CD137 expression levels after treatment
  • C1 sCD137 expression levels after treatment
  • S stable disease
  • PR partial response
  • sCD137 expression levels which are expressed as ratios of induced changes in soluble CD137 expression levels after treatment (C2-C1) to baseline sCD137 expression levels (C1) before treatment in patients dosed with 1.5, 3, 5, or 10 mg/kg ADG106.
  • FIGs. 29A-29B show comparison of CD137L protein expression levels between patients that responded to ADG106 treatment (responders) and patients that did not respond to ADG106 treatment (non-responders) .
  • FIG. 29A shows formalin-fixed paraffin-embedded (FFPE) specimens staining of CD137L protein in a responder (left) versus a non-responder (right) .
  • FIG. 29B shows analysis of CD137L protein expression in the responders (diamonds) and non-responders (circles) . The dotted line shows the tentative cut-off value of 18%.
  • the outlier in the non-responder group was a patient with non-small cell lung cancer (NSCLC) who achieved stable disease after 0.5 mg/kg ADG106 treatment.
  • NSCLC non-small cell lung cancer
  • FIGs. 30A-30B show CT scans of liver and left ethmoid sinus lesions of a nasopharyngeal carcinoma (NPC) patient who had 40%tumor reduction after two cycles of ADG106 treatment.
  • FIG. 30A shows liver lesion of the patient before treatment (left) and after two cycles of ADG106 treatment at day 42 (right) .
  • FIG. 30B shows left ethmoid sinus lesion of the patient before treatment (left) and after two cycles of ADG106 treatment at day 42 (right) .
  • FIG. 31 shows CT scan of a lymphoma patient before treatment (left) and after two cycles of ADG106 treatment (right) .
  • the patient achieved 32%tumor reduction.
  • the present application provides a method of treating a cancer in a subject using an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137.
  • the anti-CD137 antibodies described herein specifically binds an epitope that mimics the binding site of CD137L.
  • Administration of the anti-CD137 antibody lead to high receptor occupancy at a therapeutically effective dose.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 300-400 mg) .
  • the methods described herein can be used to treat a variety of cancers, including follicular lymphoma, T cell lymphoma, and adenoid cystic carcinoma (ACC) .
  • ACC adenoid cystic carcinoma
  • the methods described herein can be used to treat cancers that are advanced-stage cancer, metastatic cancer, and/or resistant or refractory to standard therapies.
  • the method comprises determining the levels of one or more biomarkers (e.g., prognostic biomarkers) selected from the group consisting of total CD137, membrane bound (mCD137) , soluble CD137 (sCD137) , CD137L, Ki67, NK cells, CD8+ effector memory T (T em ) cells, and regulatory T (T reg ) cells.
  • biomarkers e.g., prognostic biomarkers
  • the present application in one aspect provides a method of treating a cancer in a subject, comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1, and wherein the anti-CD137 antibody is administered at a dose of no more than 500 mg.
  • a method of treating a cancer in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1, and wherein the cancer is resistant or refractory to a prior therapy, e.g., a prior immunotherapy such as an anti-CD20 antibody.
  • the prior therapy is rituximab.
  • a method of treating a cancer in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1, and wherein the cancer is selected from the group consisting of follicular lymphoma, T cell lymphoma, and ACC.
  • a method of treating a cancer comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1, and an immune checkpoint inhibitor (e.g., an anti-PD-1 antibody, an anti-PD-L1 antibody, or an anti- CTLA-4 antibody) .
  • an immune checkpoint inhibitor e.g., an anti-PD-1 antibody, an anti-PD-L1 antibody, or an anti- CTLA-4 antibody
  • the subject is selected for the treatment based on a level of one or more biomarkers selected from the group consisting of total CD137, membrane bound (mCD137) , soluble CD137 (sCD137) , CD137L, Ki67, NK cells, CD8+effector memory T (T em ) cells, regulatory T (T reg ) cells and NK cells in a sample.
  • biomarkers selected from the group consisting of total CD137, membrane bound (mCD137) , soluble CD137 (sCD137) , CD137L, Ki67, NK cells, CD8+effector memory T (T em ) cells, regulatory T (T reg ) cells and NK cells in a sample.
  • a method of treating a cancer comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1, and a chemotherapeutic agent (e.g., docetaxel or cisplatin) .
  • a chemotherapeutic agent e.g., docetaxel or cisplatin
  • CD137 and CD137 receptor are used interchangeably in the present application, and include the human CD137 receptor, as well as variants, isoforms, and species homologs thereof. Accordingly, a binding molecule, as defined and disclosed herein, may also bind CD137 from species other than human. In other cases, a binding molecule may be completely specific for the human CD137 and may not exhibit species or other types of cross-reactivity.
  • CD137 antibody refers to an antibody, as defined herein, capable of binding to human CD137 receptor.
  • antibody is used herein in the broadest sense and specifically covers monoclonal antibodies (including full length monoclonal antibodies) , polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies) , and antibody fragments (e.g., a single-chain variable fragment or scFv) so long as they exhibit the desired biological activity.
  • antibody is an art-recognized term and may refer to an antigen-binding protein (i.e., immunoglobulin) having a basic four-polypeptide chain structure consisting of two identical heavy (H) chains and two identical light (L) chains. Each L chain is linked to an H chain by one covalent disulfide bond, while the two H chains are linked to each other by one or more disulfide bonds depending on the H chain isotype.
  • Each heavy chain has, at the N-terminus, a variable region (abbreviated herein as VH) followed by a constant region.
  • the heavy chain constant region is comprised of three domains, CH1, CH2 and CH3.
  • Each light chain has, at the N-terminus, a variable region (abbreviated herein as VL) followed by a constant region at its other end.
  • the light chain constant region is comprised of one domain, CL.
  • the VL is aligned with the VH and the CL is aligned with the first constant domain of the heavy chain (CH1) .
  • CH1 first constant domain of the heavy chain
  • the pairing of a VH and VL together forms a single antigen-binding site.
  • An IgM antibody consists of 5 of the basic heterotetramer units along with an additional polypeptide called J chain, and therefore contains 10 antigen binding sites, while secreted IgA antibodies can polymerize to form polyvalent assemblages comprising 2-5 of the basic 4-chain units along with J chain.
  • hyper-variable regions HVRs
  • framework regions FW
  • Kabat CDR definition by Yvonne Chen, et al. Selection and Analysis of an Optimized Anti-VEGF Antibody: Crystal Structure of an Affinity-matured Fab in Complex with Antigen, J. Mol. Biol. (1999) 293, 865-881) is listed below.
  • Each VH and VL is composed of three HVRs and four FWs, arranged from amino-terminus to carboxy-terminus in the following order: FW1, HVR1, FW2, HVR2, FW3, HVR3, FW4.
  • the three HVRs of the heavy chain are referred to as HVR_H1, HVR_H2, and HVR_H3.
  • the three HVRs of the light chain are referred to as HVR_L1, HVR_L2, and HVR_L3.
  • CDR complementarity determining region
  • CDR complementarity determining region
  • variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
  • the variable and constant regions are joined by a “J” region of about 12 or more amino acids, with the heavy chain also including a “D” region of about 10 or more amino acids. See generally, Fundamental Immunology Ch. 7 (Paul, W., ed., 2 nd ed. Raven Press, N.Y. (1989) ) .
  • the L chain from any vertebrate species can be assigned to one of two clearly distinct types, called kappa and lambda, based on the amino acid sequences of their constant domains.
  • antibodies can be assigned to different classes or isotypes. There are five classes of antibodies: IgA, IgD, IgE, IgG, and IgM, having heavy chains designated ⁇ (alpha) , ⁇ (delta) , ⁇ (epsilon) , ⁇ (gamma) , and ⁇ (mu) , respectively.
  • the IgG class of antibody can be further classified into four subclasses IgG1, IgG2, IgG3, and IgG4 by the gamma heavy chains, Y1-Y4, respectively.
  • Fc region refers to the polypeptide comprising the constant region of an antibody heavy chain excluding the first constant region immunoglobulin domain.
  • the Fc region may comprise immunoglobulin domains CH2 and CH3 and the hinge between CH1 and CH2.
  • antibody derivative or “derivative” of an antibody refers to a molecule that is capable of binding to the same antigen (e.g., CD137) that the antibody binds to and comprises an amino acid sequence of the antibody linked to an additional molecular entity.
  • the amino acid sequence of the antibody that is contained in the antibody derivative may be a full-length heavy chain, a full-length light chain, any portion or portions of a full-length heavy chain, any portion or portions of the full-length light chain of the antibody, any other fragment (s) of an antibody, or the complete antibody.
  • the additional molecular entity may be a chemical or biological molecule. Examples of additional molecular entities include chemical groups, amino acids, peptides, proteins (such as enzymes, antibodies) , and chemical compounds.
  • the additional molecular entity may have any utility, such as for use as a detection agent, label, marker, pharmaceutical or therapeutic agent.
  • the amino acid sequence of an antibody may be attached or linked to the additional molecular entity by chemical coupling, genetic fusion, noncovalent association, or otherwise.
  • antibody derivative also encompasses chimeric antibodies, humanized antibodies, and molecules that are derived from modifications of the amino acid sequences of an antibody (e.g., a CD137 antibody) , such as conservation amino acid substitutions, additions, and insertions.
  • sequence identity between two polypeptide sequences indicates the percentage of amino acids that are identical between the sequences.
  • the amino acid sequence identity of polypeptides can be determined conventionally using known computer programs such as Bestfit, FASTA, or BLAST (see, e.g. Pearson, Methods Enzymol. 183: 63-98 (1990) ; Pearson, Methods Mol. Biol. 132: 185-219 (2000) ; Altschul et al., J. Mol. Biol. 215: 403-410 (1990) ; Altschul et al., Nucleic Acids Res. 25: 3389-3402 (1997) ) .
  • the parameters are set such that the percentage of identity is calculated over the full length of the reference amino acid sequence and that gaps in homology of up to 5%of the total number of amino acid residues in the reference sequence are allowed.
  • This aforementioned method in determining the percentage of identity between polypeptides is applicable to all proteins, fragments, or variants thereof disclosed herein.
  • antigen-binding fragment or “antigen binding portion” of an antibody refers to one or more portions of an antibody that retain the ability to bind to the antigen that the antibody bonds to (e.g., CD137) .
  • antigen-binding fragment include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F (ab′) 2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., Nature 341: 544-546 (1989) ) , which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR) .
  • CDR complementarity determining
  • binding molecule encompasses (1) antibody, (2) antigen-binding fragment of an antibody, and (3) derivative of an antibody, each as defined herein.
  • binding CD137 refers to the binding of a binding molecule, as defined herein, to the human CD137 in an in vitro assay, such as a Biacore assay, with an affinity (K D ) of 100 nM or less.
  • binding molecule as defined herein, (e.g., an antibody) with its binding partner (e.g., an antigen)
  • binding partner e.g., an antigen
  • a CD137 binding molecule is said to specifically bind to human CD137 if it binds to human CD137 at an EC50 that is below 50 percent of the EC50 at which it binds CD137 of rat or mouse as determined in an in vitro assay. Binding specificity of an antibody can be determined using methods known in the art. Examples of such methods include FACS using PHA stimulated primary cells, Western blots, ELISA-, RIA-, ECL-, IRMA-tests and peptide scans.
  • Compet for binding refers to the interaction of two antibodies in their binding to a binding target.
  • a first antibody competes for binding with a second antibody if binding of the first antibody with its cognate epitope is detectably decreased in the presence of the second antibody compared to the binding of the first antibody in the absence of the second antibody.
  • the alternative, where the binding of the second antibody to its epitope is also detectably decreased in the presence of the first antibody can, but need not, be the case. That is, a first antibody can inhibit the binding of a second antibody to its epitope without that second antibody inhibiting the binding of the first antibody to its respective epitope.
  • each antibody detectably inhibits the binding of the other antibody with its cognate epitope whether to the same, greater, or lesser extent, the antibodies are said to “cross-compete” with each other for binding of their respective epitope (s) .
  • epitope refers to a part of an antigen to which an antibody (or antigen-binding fragment thereof) binds.
  • Epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents.
  • An epitope can include various numbers of amino acids in a unique spatial conformation.
  • Methods of determining spatial conformation of epitopes include, for example, x-ray crystallography, 2-dimensional nuclear magnetic resonance, deuterium and hydrogen exchange in combination with mass spectrometry, or site-directed mutagenesis, or all methods used in combination with computational modeling of antigen and its complex structure with its binding antibody and its variants. See, e.g., Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, G.E. Morris, Ed. (1996) .
  • antibodies to that epitope can be generated, e.g., using the techniques described herein. The generation and characterization of antibodies may also elucidate information about desirable epitopes.
  • human antibody refers to an antibody in which the entire amino acid sequences of the light chains and heavy chains are from the human immunoglobulin genes.
  • a human antibody may contain murine carbohydrate chains if produced in a mouse, in a mouse cell or in a hybridoma derived from a mouse cell.
  • Human antibodies may be prepared in a variety of ways known in the art.
  • humanized antibody refers to a chimeric antibody that contains amino acid residues derived from human antibody sequences.
  • a humanized antibody may contain some or all of the CDRs or HVRs from a non-human animal or synthetic antibody while the framework and constant regions of the antibody contain amino acid residues derived from human antibody sequences.
  • chimeric antibody refers to an antibody that comprises amino acid sequences derived from different animal species, such as those having a variable region derived from a human antibody and a murine immunoglobulin constant region.
  • isolated antibody or “isolated binding molecule” refers to an antibody or a binding molecule, as defined herein, that: (1) is not associated with naturally associated components that accompany it in its native state; (2) is free of other proteins from the same species; (3) is expressed by a cell from a different species; or (4) does not occur in nature.
  • isolated antibodies include a CD137 antibody that has been affinity purified using CD137, a CD137 antibody that has been generated by hybridomas or other cell line in vitro, and a CD137 antibody derived from a transgenic animal.
  • isolated nucleic acid refers to a nucleic acid molecule of genomic, cDNA, or synthetic origin, or a combination thereof, which is separated from other nucleic acid molecules present in the natural source of the nucleic acid.
  • genomic DNA the term “isolated” includes nucleic acid molecules, which are separated from the chromosome with which the genomic DNA is naturally associated.
  • an “isolated” nucleic acid is free of sequences, which naturally flank the nucleic acid (i.e., sequences located at the 5′ and 3′ ends of the nucleic acid of interest.
  • mammals are a mammal, more preferably a human. Mammals also include, but are not limited to, farm animals, sport animals, pets (such as cats, dogs, and horses) , primates, mice and rats.
  • treat refers causing a desirable or beneficial effect in the mammal having the disease condition.
  • the desirable or beneficial effect may include reduced frequency or severity of one or more symptoms of the disease (i.e., tumor growth and/or metastasis, or other effect mediated by the numbers and/or activity of immune cells, and the like) , or arrest or inhibition of further development of the disease, condition, or disorder.
  • the desirable or beneficial effect may include inhibition of further growth or spread of cancer cells, death of cancer cells, inhibition of reoccurrence of cancer, reduction of pain associated with the cancer, or improved survival of the mammal.
  • the effect can be either subjective or objective.
  • the mammal is human
  • the human may note improved vigor or vitality or decreased pain as subjective symptoms of improvement or response to therapy.
  • the clinician may notice a decrease in tumor size or tumor burden based on physical exam, laboratory parameters, tumor markers or radiographic findings.
  • Some laboratory signs that the clinician may observe for response to treatment include normalization of tests, such as white blood cell count, red blood cell count, platelet count, erythrocyte sedimentation rate, and various enzyme levels.
  • the clinician may observe a decrease in a detectable tumor marker.
  • other tests can be used to evaluate objective improvement, such as sonograms, nuclear magnetic resonance testing and positron emissions testing.
  • prevent or “preventing, ” with reference to a certain disease condition in a mammal, refers to preventing or delaying the onset of the disease, or preventing the manifestation of clinical or subclinical symptoms thereof.
  • an “effective amount” refers to an amount of an agent or drug effective to treat a disease or disorder in a subject.
  • the effective amount of the agent may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer.
  • an effective amount of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition.
  • an “effective amount” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • recurrence refers to the return of a cancer or disease after clinical assessment of the disappearance of disease. A diagnosis of distant metastasis or local recurrence can be considered a relapse.
  • refractory or “resistant” refers to a cancer or disease that has not responded to treatment.
  • an “adverse event” or “AE” as used herein refers to any untoward medical occurrence in an individual receiving a marketed pharmaceutical product or in an individual who is participating on a clinical trial who is receiving an investigational or non-investigational pharmaceutical agent.
  • the AE does not necessarily have a causal relationship with the individual’s treatment. Therefore, an AE can be any unfavorable and unintended sign, symptom, or disease temporally associated with the use of a medicinal product, whether or not considered to be related to the medicinal product.
  • An AE includes, but is not limited to: an exacerbation of a pre-existing illness; an increase in frequency or intensity of a pre-existing episodic event or condition; a condition detected or diagnosed after study drug administration even though it may have been present prior to the start of the study; and continuously persistent disease or symptoms that were present at baseline and worsen following the start of the study.
  • An AE generally does not include: medical or surgical procedures (e.g., surgery, endoscopy, tooth extraction, or transfusion) ; however, the condition that leads to the procedure is an adverse event; pre-existing diseases, conditions, or laboratory abnormalities present or detected at the start of the study that do not worsen; hospitalizations or procedures that are done for elective purposes not related to an untoward medical occurrence (e.g., hospitalizations for cosmetic or elective surgery or social/convenience admissions) ; the disease being studied or signs/symptoms associated with the disease unless more severe than expected for the individual's condition; and overdose of study drug without any clinical signs or symptoms.
  • medical or surgical procedures e.g., surgery, endoscopy, tooth extraction, or transfusion
  • a “serious adverse event” or (SAE) as used herein refers to any untoward medical occurrence at any dose including, but not limited to, that: a) is fatal; b) is life-threatening (defined as an immediate risk of death from the event as it occurred) ; c) results in persistent or significant disability or incapacity; d) requires in-patient hospitalization or prolongs an existing hospitalization (exception: Hospitalization for elective treatment of a pre-existing condition that did not worsen during the study is not considered an adverse event.
  • AEs Complications that occur during hospitalization are AEs and if a complication prolongs hospitalization, then the event is serious) ; e) is a congenital anomaly/birth defect in the offspring of an individual who received medication; or f) conditions not included in the above definitions that may jeopardize the individual or may require intervention to prevent one of the outcomes listed above unless clearly related to the individual’s underlying disease.
  • “Lack of efficacy” progressive disease
  • the signs and symptoms or clinical sequelae resulting from lack of efficacy should be reported if they fulfill the AE or SAE definitions.
  • CR complete response
  • PR partial response
  • SD stable disease
  • PD progressive disease
  • response assessments may be used to evaluate a non-target lesion: “complete response” or “CR” refers to disappearance of all non-target lesions; “stable disease” or “SD” refers to the persistence of one or more non-target lesions not qualifying for CR or PD; and “progressive disease” or “PD” refers to the “unequivocal progression” of existing non-target lesion (s) or appearance of one or more new lesion (s) is considered progressive disease (if PD for the individual is to be assessed for a time point based solely on the progression of non-target lesion (s) , then additional criteria are required to be fulfilled.
  • Progression free survival indicates the length of time during and after treatment that the cancer does not grow. Progression-free survival includes the amount of time individuals have experienced a complete response or a partial response, as well as the amount of time individuals have experienced stable disease.
  • polypeptide ” “protein, ” and “peptide” are used interchangeably herein and may refer to polymers of two or more amino acids.
  • the nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase or by a synthetic reaction.
  • a polynucleotide may comprise modified nucleotides, such as methylated nucleotides and their analogs. If present, modification to the nucleotide structure may be imparted before or after assembly of the polymer.
  • the sequence of nucleotides may be interrupted by non-nucleotide components.
  • a polynucleotide may comprise modification (s) made after synthesis, such as conjugation to a label.
  • modifications include, for example, “caps, ” substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoamidates, carbamates, etc. ) and with charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.
  • those containing pendant moieties such as, for example, proteins (e.g., nucleases, toxins, antibodies, signal peptides, ply-L-lysine, etc. ) , those with intercalators (e.g., acridine, psoralen, etc. ) , those containing chelators (e.g., metals, radioactive metals, boron, oxidative metals, etc. ) , those containing alkylators, those with modified linkages (e.g., alpha anomeric nucleic acids, etc. ) , as well as unmodified forms of the polynucleotides (s) .
  • proteins e.g., nucleases, toxins, antibodies, signal peptides, ply-L-lysine, etc.
  • intercalators e.g., acridine, psoralen, etc.
  • those chelators e.g., metals, radioactive metals
  • any of the hydroxyl groups ordinarily present in the sugars may be replaced, for example, by phosphonate groups, phosphate groups, protected by standard protecting groups, or activated to prepare additional linkages to additional nucleotides, or may be conjugated to solid or semi-solid supports.
  • the 5’ and 3’ terminal OH can be phosphorylated or substituted with amines or organic capping group moieties of from 1 to 20 carbon atoms.
  • Other hydroxyls may also be derivatized to standard protecting groups.
  • Polynucleotides can also contain analogous forms of ribose or deoxyribose sugars that are generally known in the art, including, for example, 2’-O-methyl-, 2’-O-allyl-, 2’-fluoro-or 2’-azido-ribose, carbocyclic sugar analogs, ⁇ -anomeric sugars, epimeric sugars such as arabinose, xyloses or lyxoses, pyranose sugars, furanose sugars, sedoheptuloses, acyclic analogs, and basic nucleoside analogs such as methyl riboside.
  • One or more phosphodiester linkages may be replaced by alternative linking groups.
  • linking groups include, but are not limited to, embodiments wherein phosphate is replaced by P (O) S ( “thioate” ) , P (S) S ( “dithioate” ) , (O) NR2 ( “amidate” ) , P (O) R, P (O) OR’, CO, or CH2 ( “formacetal” ) , in which each R or R’ is independently H or substituted or unsubstituted alkyl (1-20 C) optionally containing an ether (-O-) linkage, aryl, alkenyl, cycloalkyl, cycloalkenyl or araldyl. Not all linkages in a polynucleotide need be identical. The preceding description applies to all polynucleotides referred to herein, including RNA and DNA.
  • biomarker refers generally to a molecule (e.g., pre-mRNA, mRNA, protein, etc. ) or cell population (e.g., effector memory T cell or T em cell, or regulatory T cell or T reg cell) , the level of which in or on a subject’s tissue (e.g., tumor) , or in case of a molecule, secreted by the subject’s tissue or cell, can be detected by known methods (or methods disclosed herein) and is predictive or can be used to predict (or aid prediction) for a subject’s sensitivity to, and in some embodiments, to predict (or aid prediction) a subject’s responsiveness to, treatment regimens (e.g., treatments with an anti-CD137 antibody) .
  • treatment regimens e.g., treatments with an anti-CD137 antibody
  • sample refers to a composition that is obtained or derived from a subject of interest that contains a cellular and/or other molecular entity that is to be characterized and/or identified, for example based on physical, biochemical, chemical and/or physiological characteristics.
  • tissue or cell sample refers to a collection of similar cells obtained from a tissue of a subject or patient.
  • the source of the tissue or cell sample may be solid tissue as from a fresh, frozen and/or preserved organ or tissue sample or biopsy or aspirate; blood or any blood constituents; bodily fluids such as cerebral spinal fluid, amniotic fluid, peritoneal fluid, or interstitial fluid; cells from any time in gestation or development of the subject.
  • the tissue sample may also be primary or cultured cells.
  • the tissue or cell sample is obtained from a disease tissue or organ.
  • the tissue sample may contain compounds, which are not naturally intermixed with the tissue in nature such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics, or the like.
  • a “reference value” or “reference level” may be an absolute value; a relative value; a value that has an upper and/or lower limit; a range of values; an average value; a median value; a mean value; or a value as compared to a particular level or baseline level.
  • reference to "not" a value or parameter generally means and describes "other than” a value or parameter.
  • the method is not used to treat cancer of type X means the method is used to treat cancer of types other than X.
  • a and/or B is intended to include both A and B; A or B; A (alone) ; and B (alone) .
  • the term “and/or” as used herein a phrase such as “A, B, and/or C” is intended to encompass each of the following embodiments: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone) ; B (alone) ; and C (alone) .
  • the present application provides methods for treating cancers using a binding molecule (e.g., an anti-CD137 antibody) that specifically binds to an extracellular domain of human CD137.
  • a binding molecule e.g., an anti-CD137 antibody
  • Any one of the anti-CD137 antibodies (including full-length antibodies and antigen-binding fragments thereof) in Section III “Anti-CD137 Antibodies” may be used in the methods described herein.
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is selected from the group consisting of colon cancer (e.g., Sigmoid colon cancer) , breast cancer, lung cancer (e.g., non-small cell lung cancer or NSCLC) , esophageal cancer, endometrial cancer, gastrointestinal cancer (e.g., gastrointestinal neuroectodermal tumor) , cholangiocarcinoma, nasopharyngeal cancer (NPC) , adenoid cystic carcinoma (ACC) , melanoma, mesothelioma (e.g., malignant pleural mesothelioma or MPM) , mantle cell lymphoma, anal cancer, head and neck cancer (e.g., head and neck squamous cell carcinoma or HNSCC) , appendiceal and sebaceous cancer, follicular lymphoma, non-Hodgkin’s lymphoma (NHL) , and T cell lymphoma (e.g.,
  • a method of treating a cancer in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to amino acid residues 51, 63-67, 69-73, 83, 89, 92, 98-104 and 112-114 of SEQ ID NO: 1, and wherein the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • 500 mg e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is selected from the group consisting of colon cancer (e.g., Sigmoid colon cancer) , breast cancer, lung cancer (e.g., NSCLC) , esophageal cancer, endometrial cancer, gastrointestinal cancer (e.g., gastrointestinal neuroectodermal tumor) , cholangiocarcinoma, NPC, ACC, melanoma, mesothelioma (e.g., MPM) , mantle cell lymphoma, anal cancer, head and neck cancer (e.g., HNSCC) , appendiceal and sebaceous cancer, follicular lymphoma, NHL, and T cell lymphoma (e.g., AITL, or PTCL) .
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • a method of treating a cancer in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the anti-CD137 antibody comprises a heavy chain variable region (VH) and a light chain variable region (VL) , wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 2, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 3, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 4, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 5, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 6, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 7; and wherein the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg
  • the VH comprises the amino acid sequence of SEQ ID NO: 8
  • the VL comprises the amino acid sequence of SEQ ID NO: 9.
  • the antibody comprises a heavy chain and a light chain, and wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 10, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 11.
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is selected from the group consisting of colon cancer (e.g., Sigmoid colon cancer) , breast cancer, lung cancer (e.g., NSCLC) , esophageal cancer, endometrial cancer, gastrointestinal cancer (e.g., gastrointestinal neuroectodermal tumor) , cholangiocarcinoma, NPC, ACC, melanoma, mesothelioma (e.g., MPM) , mantle cell lymphoma, anal cancer, head and neck cancer (e.g., HNSCC) , appendiceal and sebaceous cancer, follicular lymphoma, NHL, and T cell lymphoma (e.g., AITL, or PTCL) .
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is selected from the group consisting of colon cancer (e.g., Sigmoid colon cancer) , breast cancer, lung cancer (e.g., NSCLC) , esophageal cancer, endometrial cancer, gastrointestinal cancer (e.g., gastrointestinal neuroectodermal tumor) , cholangiocarcinoma, NPC, ACC, melanoma, mesothelioma (e.g., MPM) , mantle cell lymphoma, anal cancer, head and neck cancer (e.g., HNSCC) , appendiceal and sebaceous cancer, follicular lymphoma, NHL, and T cell lymphoma (e.g., AITL, or PTCL) .
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • a method of treating a cancer in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 25, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 26, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 27; and wherein the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is selected from the group consisting of colon cancer (e.g., Sigmoid colon cancer) , breast cancer, lung cancer (e.g., NSCLC) , esophageal cancer, endometrial cancer, gastrointestinal cancer (e.g., gastrointestinal neuroectodermal tumor) , cholangiocarcinoma, NPC, ACC, melanoma, mesothelioma (e.g., MPM) , mantle cell lymphoma, anal cancer, head and neck cancer (e.g., HNSCC) , appendiceal and sebaceous cancer, follicular lymphoma, NHL, and T cell lymphoma (e.g., AITL, or PTCL) .
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • a method of treating a cancer in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and wherein the cancer is resistant or refractory to a prior therapy (e.g., a prior immunotherapy such as an anti-CD20 antibody, for example, rituximab) .
  • a prior immunotherapy such as an anti-CD20 antibody, for example, rituximab
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) . In some embodiments, the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg. In some embodiments, the anti-CD137 antibody is administered intravenously. In some embodiments, the anti-CD137 antibody is administered about once every three weeks.
  • a method of treating a cancer in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to amino acid residues 51, 63-67, 69-73, 83, 89, 92, 98-104 and 112-114 of SEQ ID NO: 1, and wherein the cancer is resistant or refractory to a prior therapy (e.g., a prior immunotherapy such as an anti-CD20 antibody, for example, rituximab) .
  • a prior immunotherapy such as an anti-CD20 antibody, for example, rituximab
  • a method of treating a cancer in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the anti-CD137 antibody comprises a heavy chain variable region (VH) and a light chain variable region (VL) , wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 2, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 3, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 4, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 5, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 6, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 7; and wherein the cancer is resistant or refractory to a prior therapy (e.g., a prior immunotherapy such as an anti-CD
  • the VH comprises the amino acid sequence of SEQ ID NO: 8
  • the VL comprises the amino acid sequence of SEQ ID NO: 9.
  • the antibody comprises a heavy chain and a light chain, and wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 10, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 11.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is selected from the group consisting of colon cancer (e.g., Sigmoid colon cancer) , breast cancer, lung cancer (e.g., NSCLC) , esophageal cancer, endometrial cancer, gastrointestinal cancer (e.g., gastrointestinal neuroectodermal tumor) , cholangiocarcinoma, NPC, ACC, melanoma, mesothelioma (e.g., MPM) , anal cancer, head and neck cancer (e.g., HNSCC) , mantle cell lymphoma, appendiceal and sebaceous cancer, follicular lymphoma, NHL, and T cell lymphoma (e.g., AITL, or PTCL) .
  • colon cancer e.g., Sigmoid colon cancer
  • breast cancer e.g., breast cancer
  • lung cancer e.g., NSCLC
  • esophageal cancer endometrial cancer
  • gastrointestinal cancer
  • a method of treating a cancer in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 15, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 17; and wherein the cancer is resistant or refractory to a prior therapy (e.g., a prior immunotherapy such as an anti-CD20 antibody, for example, rituximab
  • the VH comprises the amino acid sequence of SEQ ID NO: 18, and/or the VL comprises the amino acid sequence of SEQ ID NO: 19.
  • the antibody comprises a heavy chain and a light chain, wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 20, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 21.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the cancer is selected from the group consisting of colon cancer (e.g., Sigmoid colon cancer) , breast cancer, lung cancer (e.g., NSCLC) , esophageal cancer, endometrial cancer, gastrointestinal cancer (e.g., gastrointestinal neuroectodermal tumor) , cholangiocarcinoma, NPC, ACC, melanoma, mesothelioma (e.g., MPM) , anal cancer, head and neck cancer (e.g., HNSCC) , mantle cell lymphoma, appendiceal and sebaceous cancer, follicular lymphoma, NHL, and T cell lymphoma (e.g., AITL, or PTCL) .
  • colon cancer e.g., Sigmoid colon cancer
  • breast cancer e.g., breast cancer
  • lung cancer e.g., NSCLC
  • esophageal cancer endometrial cancer
  • gastrointestinal cancer
  • a method of treating a cancer in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 25, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 26, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 27; and wherein the cancer is resistant or refractory to a prior therapy (e.g., a prior immunotherapy such as an anti-CD20 antibody, for example, rituximab
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • a method of treating a follicular lymphoma in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • a method of treating a follicular lymphoma in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to amino acid residues 51, 63-67, 69-73, 83, 89, 92, 98-104 and 112-114 of SEQ ID NO: 1.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • the VH comprises the amino acid sequence of SEQ ID NO: 8
  • the VL comprises the amino acid sequence of SEQ ID NO: 9.
  • the antibody comprises a heavy chain and a light chain, and wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 10, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 11.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • the VH comprises the amino acid sequence of SEQ ID NO: 18, and/or the VL comprises the amino acid sequence of SEQ ID NO: 19.
  • the antibody comprises a heavy chain and a light chain, wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 20, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 21.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • a method of treating a follicular lymphoma in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 25, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 26, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 27.
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • a method of treating a T cell lymphoma in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1.
  • the T cell lymphoma is AITL or PTCL.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • a method of treating a T cell lymphoma in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the anti-CD137 antibody comprises a heavy chain variable region (VH) and a light chain variable region (VL) , wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 2, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 3, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 4, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 5, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 6, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 7.
  • VH heavy chain variable region
  • VL light chain variable region
  • the VH comprises the amino acid sequence of SEQ ID NO: 8
  • the VL comprises the amino acid sequence of SEQ ID NO: 9.
  • the antibody comprises a heavy chain and a light chain, and wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 10, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 11.
  • the T cell lymphoma is AITL or PTCL.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • a method of treating a T cell lymphoma in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 15, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 17.
  • a method of treating a T cell lymphoma in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 25, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 26, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 27.
  • a method of treating an adenoid cystic carcinoma (ACC) in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to amino acid residues 51, 63-67, 69-73, 83, 89, 92, 98-104 and 112-114 of SEQ ID NO: 1.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • a method of treating an adenoid cystic carcinoma (ACC) in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the anti-CD137 antibody comprises a heavy chain variable region (VH) and a light chain variable region (VL) , wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 2, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 3, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 4, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 5, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 6, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 7.
  • VH heavy chain variable region
  • VL light chain variable region
  • the VH comprises the amino acid sequence of SEQ ID NO: 8
  • the VL comprises the amino acid sequence of SEQ ID NO: 9.
  • the antibody comprises a heavy chain and a light chain, and wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 10, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 11.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • the VH comprises the amino acid sequence of SEQ ID NO: 18, and/or the VL comprises the amino acid sequence of SEQ ID NO: 19.
  • the antibody comprises a heavy chain and a light chain, wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 20, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 21.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • Cancer treatments can be evaluated by, e.g., tumor regression, tumor weight or size shrinkage, time to progression, duration of survival, progression free survival, overall response rate, duration of response, quality of life, protein expression and/or activity.
  • Approaches to determining efficacy of therapy can be employed, including for example, measurement of response through radiological imaging.
  • enteral route refers to the administration via any part of the gastrointestinal tract.
  • enteral routes include oral, mucosal, buccal, and rectal route, or intragastric route.
  • Parenteral route refers to a route of administration other than enteral route.
  • parenteral routes of administration examples include intravenous, intramuscular, intradermal, intraperitoneal, intratumor, intravesical, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, transtracheal, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal, subcutaneous, or topical administration.
  • the antibodies and compositions of the disclosure can be administered using any suitable method, such as by oral ingestion, nasogastric tube, gastrostomy tube, injection, infusion, implantable infusion pump, and osmotic pump.
  • the suitable route and method of administration may vary depending on a number of factors such as the specific antibody being used, the rate of absorption desired, specific formulation or dosage form used, type or severity of the disorder being treated, the specific site of action, and conditions of the patient, and can be readily selected by a person skilled in the art.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered at a dose of any one of about 150 mg to about 200 mg, about 150 mg to about 300 mg, about 150 mg to about 400 mg, about 150 mg to about 500 mg, about 100 mg to about 200 mg, about 200 mg to about 300 mg, about 300 mg to about 400 mg, about 400 mg to about 500 mg, about 100 mg to about 300 mg, about 300 mg to about 500 mg, about 200 mg to about 400 mg, about 100 mg to about 250 mg, about 250 mg to about 500 mg, about 250 mg to about 400 mg, about 100 mg to about 400 mg, about 200 mg to about 500 mg, or about 100 mg to about 500 mg.
  • the doses described herein may refer to a suitable dose for a human, or an equivalent dose for the specific species of the subject.
  • the anti-CD137 antibody is administered at a dose of no more than any one of 10 mg/kg, 9 mg/kg, 8 mg/kg, 7 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, 1 mg/kg, 0.8 mg/kg, 0.6 mg/kg, 0.5 mg/kg, 0.4 mg/kg, 0.3 mg/kg, 0.2 mg/kg, 0.1 mg/kg, 0.08 mg/kg, 0.05 mg/kg, 0.04 mg/kg, or 0.03 mg/kg.
  • the dose of the anti-CD137 antibody is within any one of the following ranges, wherein the ranges have an upper limit of any one of: 10 mg/kg, 9 mg/kg, 8 mg/kg, 7 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, 1 mg/kg, 0.8 mg/kg, 0.6 mg/kg, 0.5 mg/kg, 0.4 mg/kg, 0.3 mg/kg, 0.2 mg/kg, 0.1 mg/kg, 0.08 mg/kg, 0.05 mg/kg, or 0.04 mg/kg, and an independently selected lower limit of any one of 9 mg/kg, 8 mg/kg, 7 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, 1 mg/kg, 0.8 mg/kg, 0.6 mg/kg, 0.5 mg/kg, 0.4 mg/kg, 0.3 mg/kg, 0.2 mg/kg, 0.1 mg/kg, 0.08 mg/kg, 0.05 mg/kg, or
  • the anti-CD137 antibody is administered at a dose of any one of about 0.03 mg/kg to about 10 mg/kg, about 0.1 mg/kg to about 10 mg/kg, about 0.3 mg/kg to about 10 mg/kg, about 1 mg/kg to about 10 mg/kg, about 3 mg/kg to about 10 mg/kg, about 5 mg/kg to about 10 mg/kg, about 0.03 mg/kg to about 0.1 mg/kg, about 0.1 mg/kg to about 0.3 mg/kg, about 0.3 mg/kg to about 1 mg/kg, about 1 mg/kg to about 3 mg/kg, about 3 mg/kg to about 5 mg/kg, about 0.1 mg/kg to about 3 mg/kg, or about 1 mg/kg to about 5 mg/kg.
  • the effective amount of the anti-CD137 antibody may be administered in a single dose or in multiple doses.
  • exemplary dosing frequencies include, but are not limited to, weekly, weekly without break, weekly for two out of three weeks, weekly for three out of four weeks, once every three weeks, once every two weeks, monthly, every six months, yearly, etc.
  • the anti-CD137 antibody is administered about weekly, once every 2 weeks, or once every 3 weeks.
  • the intervals between each administration are less than about any of 3 years, 2 years, 12 months, 11 months, 10 months, 9 months, 8 months, 7 months, 6 months, 5 months, 4 months, 3 months, 2 months, 1 month, 4 weeks, 3 weeks, 2 weeks, or 1 week. In some embodiments, the intervals between each administration are more than about any of 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, 2 years, or 3 years. In some embodiments, there is no break in the dosing schedule.
  • the anti-CD137 antibody is administered at a low frequency, for example, any one of no more frequent than once per week, once every other week, once per three weeks, once per month, once per 2 months, once per 3 months, once per 4 months, once per 5 months, once per 6 months, once per 7 months, once per 8 months, once per 9 months, once per 10 months, once per 11 months, once per year, or less.
  • the anti-CD137 antibody is administered in a single dose. In some embodiments, the anti-CD137 antibody is administered about once every three weeks.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg, such as no more than any one of 400 mg, 350 mg, 300 mg, 250 mg, 200 mg, 150 mg or 100 mg once every three weeks. In some embodiments, the anti-CD137 antibody is administered at a dose of about 150 mg to about 500 mg, such as about any one of 150 mg, 200 mg, 250 mg, 300 mg, 350 mg, or 400 mg, once every three weeks.
  • the anti-CD137 antibody is administered at a dose of no more than 10 mg/kg, such as no more than any one of 8 mg/kg, 5 mg/kg, 3 mg/kg, 2 mg/kg, or 1 mg/kg once every three weeks. In some embodiments, the anti-CD137 antibody is administered at a dose of about 0.03 mg/kg to about 10 mg/kg, such as about any one of 0.03, 0.1, 0.3, 1, 3, 5, 8, or 10mg/kg, once every three weeks.
  • the anti-CD137 antibody is administered for 2 or more cycles, such as about any one of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more cycles.
  • the administration of the anti-CD137 antibody can be extended over an extended period of time, such as from about a week to about a month, from about a month to about a year, from about a year to about several years.
  • the anti-CD137 antibody is administered over a period of at least any of about 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, 1 year, 2 years, 3 years, 4 years, or more.
  • the methods described herein are useful for treating a variety of cancers.
  • the cancer is a solid cancer.
  • the cancer is a liquid cancer.
  • a variety of cancers where CD137 is implicated, whether malignant or benign and whether primary or secondary, may be treated or prevented with a method provided by the disclosure.
  • lung cancers such as bronchogenic carcinoma (e.g., squamous cell carcinoma, small cell carcinoma, large cell carcinoma, and adenocarcinoma) , alveolar cell carcinoma, bronchial adenoma, chondromatous hamartoma (noncancerous) , and sarcoma (cancerous) ; heart cancer such as myxoma, fibromas, and rhabdomyomas; bone cancers such as osteochondromas, condromas, chondroblastomas, chondromyxoid fibromas, osteoid osteomas, giant cell tumors, chondrosarcoma, multiple myeloma, osteosarcoma, fibrosarcomas, malignant fibrous histiocytomas, Ewing's tumor (Ewing's sarcoma) , and reticulum cell sarcoma; brain cancer such as gliomas (e.g., glioblasto
  • the subject has been previously treated with a prior therapy. In some embodiments, the subject has previously received any one of 1, 2, 3, 4, or more prior therapies. In some embodiments, the subject has exhausted all other available therapies. In some embodiments, the subject is unresponsive or resistant to a prior therapy. In some embodiments, the subject has disease reoccurrence subsequent to a prior therapy. In some embodiments, the subject is refractory to a prior therapy. In some embodiments, the subject has failed a prior therapy within about 1 year, 6 months, 3 months or less. In some embodiments, the subject has not previously received a prior therapy.
  • the subject has been previously treated with a standard therapy for the cancer. In some embodiments, the subject is unresponsive or resistant to a standard therapy. In some embodiments, the subject has disease reoccurrence subsequent to a standard therapy. In some embodiments, the subject is refractory to a standard therapy. In some embodiments, the subject has failed a standard therapy within about 1 year, 6 months, 3 months or less. In some embodiments, the subject has not previously received a standard therapy. In some embodiments, the subject has refused or is ineligible for a standard therapy.
  • the prior therapy is selected from the group consisting of viral gene therapy, immunotherapy, targeted therapy, radiation therapy, and chemotherapy.
  • the prior therapy is selected from the group consisting of pomalyst, revlimid, lenalidomide, pomalidomide, thalidomide, a DNA-alkylating platinum-containing derivative, cisplatin, 5-fluorouracil, cyclophosphamide, an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-CD20 antibody, an anti-CD40 antibody, an anti-DR5 antibody, an anti-CD1d antibody, an anti-TIM3 antibody, an anti-SLAMF7 antibody, an anti-KIR receptor antibody, an anti-OX40 antibody, an anti-HER2 antibody, an anti-ErbB-2 antibody, an anti-EGFR antibody, cetuximab, rituximab, trastuzumab, pembrolizum
  • chemotherapeutic agent refers to a chemical or biological substance that can cause death of cancer cells, or interfere with growth, division, repair, and/or function of cancer cells.
  • chemotherapeutic agents include those that are disclosed in WO 2006/129163, and US 20060153808, the disclosures of which are incorporated herein by reference.
  • chemotherapeutic agents include: (1) alkylating agents, such as chlorambucil mcyclophosphamide ifosfamide mechlorethamine hydrochloride thiotepa streptozotocin carmustine ( GLIADEL ) , lomustine and dacarbazine (2) alkaloids or plant vinca alkaloids, including cytotoxic antibiotics, such as doxorubicin epirubicin daunorubicin nemorubicin, idarubicin ( PFS, ) , mitoxantrone dactinomycin (actinomycin D, ) , plicamycin mitomycin and bleomycin vinorelbine tartrate vinblastine vincristine and vindesine (3) antimetabolites, such as capecitabine cytarabine fludarabine gemcitabine hydroxyurea methotrexate nelarabine trimetrexate and pemetrexed (4) Pyrimidine antagonists, such as
  • immunotherapeutic agents refers to a chemical or biological substance that can enhance an immune response of a mammal.
  • immunotherapeutic agents include: bacillus Calmette-Guerin (BCG) ; cytokines such as interferons; vaccines such as MyVax personalized immunotherapy, Onyvax-P, Oncophage, GRNVAC1, Favld, Provenge, Lovaxin C, BiovaxID, GMXX, and NeuVax; and antibodies such as alemtuzumab bevacizumab cetuximab gemtuzunab ozogamicin ibritumomab tiuxetan panitumumab rituximab trastuzumab tositumomab ipilimumab tremelimumab, CAT-3888, agonist antibodies to OX40 receptor (such as those disclosed in WO2009/079335) , agonist antibodies to CD
  • hormone therapeutic agents include tamoxifen toremifene fulvestrant anastrozole exemestane letrozole megestrol acetate goserelin and leuprolide
  • non-drug hormone therapies such as (1) surgical methods that remove all or part of the organs or glands which participate in the production of the hormone, such as the ovaries, the testicles, the adrenal gland, and the pituitary gland, and (2) radiation treatment, in which the organs or glands of the patient are subjected to radiation in an amount sufficient to inhibit or eliminate the production of the targeted hormone.
  • Prior therapies also encompass surgery to remove a tumor and radiation therapy.
  • exemplary radiation therapies include, but are not limited to, ionizing (electromagnetic) radiotherapy (e.g., X-rays or gamma rays) and particle beam radiation therapy (e.g., high linear energy radiation) .
  • the source of radiation can be external or internal to the subject.
  • the subject has previously received an immunotherapy.
  • the subject is resistant or refractory to the immunotherapy.
  • the immunotherapy is an anti-CD20 antibody.
  • the immunotherapy is rituximab or a biosimilar thereof.
  • the cancer is selected from the group consisting of colon cancer (e.g., Sigmoid colon cancer) , breast cancer, lung cancer (e.g., non-small cell lung cancer or NSCLC) , esophageal cancer, endometrial cancer, gastrointestinal cancer (e.g., gastrointestinal neuroectodermal tumor) , cholangiocarcinoma, nasopharyngeal cancer (NPC) , adenoid cystic carcinoma (ACC) , melanoma, mesothelioma (e.g., malignant pleural mesothelioma or MPM) , anal cancer, head and neck cancer (e.g., head and neck squamous cell carcinoma or HNSCC) , mantle cell lymphoma, appendiceal and sebaceous cancer, follicular lymphoma, non-Hodgkin’s lymphoma (NHL) , and T cell lymphoma (e.g.,
  • the cancer is a non-Hodgkin’s lymphoma (NHL) .
  • the NHL arises from B-lymphocytes.
  • the cancer is a B cell lymphoma.
  • the cancer is a diffuse large B-cell lymphoma (DLBCL) .
  • the cancer is follicular lymphoma (FL) .
  • the cancer is a low-grade FL.
  • the cancer is a high-grade FL.
  • the cancer is early stage FL, non-metastatic FL, primary FL, advanced FL, locally advanced FL, metastatic FL, FL in remission, or recurrent FL.
  • the FL has metastasized to other organs (e.g., lung, lymph nodes, or bone) .
  • the FL is Grade 1-2, wherein the tumor has no more than 15 centroblasts per high-power field.
  • the FL is Grade 3A, wherein the tumor has more than 15 centroblasts per high-power field, and the tumor is a mix of centrocytes and centroblasts. In some embodiments, the FL is Grade 3B, wherein the tumor has more than 15 centroblasts per high-power field, and the tumor has sheets of large centroblasts and no centrocytes.
  • the subject having FL has previously received one or more prior therapies. In some embodiments, the subject is resistant to or refractory to the one or more prior therapies. In some embodiments, the subject has exhausted all available therapies for FL.
  • Exemplary therapies for FL include, but are not limited to, antibody treatment (e.g., rituximab and obinutuzumab) , chemotherapies (e.g., vinca alkaloids such as etoposide and vincristine, anthracyclines such as doxorubicin, alkylating agents such as bendamustine, cyclophosphamide and ifosfamide, or antimetabolites such as methotrexate and cytarabine) , corticosteroids (e.g., prednisone and dexamethasone) , kinase inhibitors (e.g., PI3K inhibitors such as idelalisib, duvelisib, and copalisib) , immunomodulators (e.g., lenalidomide) , radioimmunotherapy (e.g., ibritumomab tiuxetan) , CAR T-
  • the cancer is T cell lymphoma (TCL) .
  • T-lymphoblastic lymphoma or leukemia In some embodiments, the cancer is peripheral T-cell lymphoma.
  • the cancer is angioimmunoblastic T-cell lymphoma (AITL) .
  • AITL angioimmunoblastic T-cell lymphoma
  • the cancer is extranodal natural killer/T-cell lymphoma, e.g., nasal type.
  • the cancer is enteropathy-associated intestinal T-cell lymphoma (EATL) .
  • the cancer is lymph node/tonsil type of TCL.
  • the cancer is anaplastic large cell lymphoma (ALCL) .
  • the cancer is peripheral T-cell lymphoma (PTCL) .
  • the cancer is early stage TCL, non-metastatic TCL, primary TCL, advanced TCL, locally advanced TCL, metastatic TCL, TCL in remission, or recurrent TCL.
  • the TCL has metastasized to other organs (e.g., lung, lymph nodes, or bone) .
  • the TCL is Stage I, wherein the cancer is in only one cluster of lymph nodes.
  • the TCL is Stage II, wherein the cancer is in 2 or more clusters either above or below the diaphragm of the subject.
  • the TCL is Stage III, wherein the cancer is in lymph tissue on both sides of the diaphragm of the subject.
  • the cancer is Stage IV, wherein the cancer has widely spread outside the lymphatic system.
  • the subject having TCL has previously received one or more prior therapies.
  • the subject is resistant to or refractory to the one or more prior therapies, such as targeted therapy, chemotherapy, steroids, immunotherapy, radiation therapy, stem cell transplant, and combinations thereof.
  • the subject has exhausted all available therapies for TCL.
  • Exemplary therapies for TCL include, but are not limited to, alemtuzumab, belinostat, bendamustine hydrochloride, bortezomib, brentuximab vedotin, carboplatin, cisplatin, cyclophosphamide, cyclosporine, cytarabine, dexamethasone, doxorubicin, epirubicin, etoposide, gemcitabine, ifosfamide, lenalidomide, leucovorin calcium, mesna, methotrexate, methylprednisolone, oxaliplatin, pralatrexate, prednisone, romidepsin, vincristine sulfate, and combinations thereof.
  • TCL can be diagnosed and monitored using known methods in the art, for example, by imaging tests (e.g., CT scan or PET scan) , biopsy, hematopathology review, immunohistochemistry (IHC) panel or genetic tests, as described in the NCCN Guidelines for Peripheral T cell Lymphoma.
  • imaging tests e.g., CT scan or PET scan
  • biopsy e.g., biopsy, hematopathology review, immunohistochemistry (IHC) panel or genetic tests, as described in the NCCN Guidelines for Peripheral T cell Lymphoma.
  • IHC immunohistochemistry
  • the cancer is adenoid cystic carcinoma (ACC) .
  • the ACC is a head and neck ACC.
  • the ACC is in breast, skin, respiratory system, and/or a reproductive organ.
  • the cancer is early stage ACC, non-metastatic ACC, primary ACC, advanced ACC, locally advanced ACC, metastatic ACC, ACC in remission, or recurrent ACC.
  • the ACC has metastasized to other organs (e.g., lung, lymph nodes, or bone) .
  • the ACC is Grade I, wherein the subject has tumors having tubular and cribriform areas but without solid components.
  • the ACC is Grade II, wherein the subject has cribriform tumors that are either pure or mixed with less than 30%of solid areas. In some embodiments, the ACC is Grade III, wherein the subject has tumors with predominantly solid pattern. In some embodiments, the subject having ACC has previously received one or more prior therapies. In some embodiments, the subject is resistant to or refractory to the one or more prior therapies. In some embodiments, the subject has exhausted all available therapies for ACC.
  • Exemplary therapies for ACC include, but are not limited to, surgery, chemotherapies (e.g., cisplatin, doxorubicin, cyclophosphamide, 5-FU, mitoxantrone, epirubicin, vinorelbine, paclitaxel, gemcitabine) , targeted therapies (e.g., imatinib, dasatinib, cetuximab, gefitinib, lapatinib) , kinase inhibitors, immunomodulators, and radiation therapy, and combinations thereof.
  • chemotherapies e.g., cisplatin, doxorubicin, cyclophosphamide, 5-FU, mitoxantrone, epirubicin, vinorelbine, paclitaxel, gemcitabine
  • targeted therapies e.g., imatinib, dasatinib, cetuximab, gefitinib, lapatinib
  • a method of inhibiting cell proliferation comprising administering to the individual an effective amount of any one of the anti-CD137 antibodies described herein.
  • at least about 10% including for example at least about any of 20%, 30%, 40%, 60%, 70%, 80%, 90%, 95%or more) cell proliferation is inhibited.
  • a method of reducing such as eradicating) pre-existing tumor metastasis (such as metastasis to the lymph node) in an individual, comprising administering to the individual an effective amount of any one of the anti-CD137 antibodies described herein.
  • at least about 10% including for example at least about any of 20%, 30%, 40%, 60%, 70%, 80%, 90%, 95%or more) metastasis is reduced.
  • a method of reducing incidence or burden of preexisting tumor metastasis comprising administering to the individual an effective amount of any one of the anti-CD137 antibodies described herein.
  • a method of reducing tumor size in an individual comprising administering to the individual an effective amount of any one of the anti-CD137 antibodies described herein.
  • the method reduces tumor size by at least about 10% (including for example at least about any of 20%, 30%, 40%, 60%, 70%, 80%, 90%, 95%or more) .
  • a method of prolonging time to disease progression of cancer in an individual comprising administering to the individual an effective amount of any one of the anti-CD137 antibodies described herein. In some embodiments, the method prolongs the time to disease progression by at least any of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 16, 20, 24, 28, 32, 36, or more weeks.
  • a method of prolonging survival e.g., overall survival or progression-free survival of an individual having cancer, comprising administering to the individual an effective amount of any one of the anti-CD137 antibodies described herein.
  • the method prolongs the survival of the individual by at least any of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, or 24 months.
  • a method of alleviating one or more symptoms in an individual having cancer comprising administering to the individual an effective amount of any one of the anti-CD137 antibodies described herein.
  • compositions of any one of the anti-CD137 antibodies described herein for use in the methods described in this section, and use of the anti-CD137 antibodies in the manufacture of a medicament for treating cancer e.g., follicular lymphoma, T cell lymphoma, or ACC.
  • a method of treating a lung cancer in a subject comprising administering to the subject: (a) an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and (b) an effective amount of an immune checkpoint inhibitor.
  • a method of treating a breast cancer comprising administering to the subject: (a) an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and (b) an effective amount of an immune checkpoint inhibitor.
  • Immune system checkpoint molecules include, but are not limited to, cytotoxic T-lymphocyte antigen 4 (CTLA-4) , programmed cell death 1 protein (PD-1) , programmed cell death 1 ligand 1 (PD-L1) , programmed cell death 1 ligand 2 (PD-L2) , lymphocyte activation gene 3 (LAG3) , B7-1, B7-H3, B7-H4, T cell membrane protein 3 (TIM3) , B- and T-lymphocyte attenuator (BTLA) , V-domain immunoglobulin (Ig) -containing suppressor of T-cell activation (VISTA) , Killer-cell immunoglobulin-like receptor (KIR) , and A2A adenosine receptor (A2aR) .
  • CTLA-4 cytotoxic T-lymphocyte antigen 4
  • PD-1 programmed cell death 1 protein
  • P-L1 programmed cell death 1 ligand 1
  • ligand 2 ligand 2
  • LAG3 lymphocyte activ
  • checkpoint inhibitors include antagonists of CTLA-4, PD-1, PD-L1, PD-L2, LAG3, B7-1, B7-H3, B7-H4, BTLA, VISTA, KIR, A2aR, or TIM3.
  • CTLA-4, PD-1, PD-L1, PD-L2, LAG3, B7-1, B7-H3, B7-H4, BTLA, VISTA, KIR, A2aR, or TIM3 are checkpoint inhibitors.
  • any molecule e.g., peptide, nucleic acid, small molecule, etc.
  • a checkpoint inhibitor e.g., peptide, nucleic acid, small molecule, etc.
  • the immune checkpoint inhibitor is an antibody that specifically binds to an immune checkpoint molecule. In some embodiments, the immune checkpoint inhibitor is selected from the group consisting of an anti-PD-1 antibody, an anti-PD-L1 antibody, and an anti-CTLA-4 antibody.
  • the immune checkpoint inhibitor is an anti-PD-1 antibody.
  • anti-PD-1 antibodies include, but are not limited to, 2E5 (Cstone Pharmaceuticals) , tislelizumab (BGB-A317) , BGB-108, STI-A1110, AM0001, BI 754091, sintilimab (IBI308) , cetrelimab (JNJ-63723283) , toripalimab (JS-001) , camrelizumab (SHR-1210, INCSHR-1210, HR-301210) , MEDI-0680 (AMP-514) , MGA-012 (INCMGA 0012) , nivolumab (BMS-936558, MDX1106, ONO-4538) , spartalizumab (PDR00l) , pembrolizumab (MK-3475, SCH 900475) , PF-06801591, cemiplimab (REGN-2810
  • the antibodies that compete with any of these art-recognized antibodies for binding to PD-1 also can be used.
  • the immune checkpoint inhibitor is 2E5. 2E5 and related anti-PD-1 antibodies have been described, for example, in CN107840887A, which is incorporated herein by reference in its entirety.
  • the immune checkpoint inhibitor is toripalimab. Toripalimab and related anti-PD-1 antibodies have been described, for example, in US10066013B2, which is incorporated herein by reference in its entirety.
  • the immune checkpoint inhibitor is an anti-PD-L1 antibody.
  • anti-PD-L1 antibodies include, but are not limited to, atezolizumab, avelumab, durvalumab (imfinzi) , BGB-A333, SHR-1316 (HTI-1088) , CK-301, BMS-936559, envafolimab (KN035, ASC22) , CS1001, MDX-1105 (BMS-936559) , LY3300054, STI-A1014, FAZ053, CX-072, INCB086550, GNS-1480, CA-170, CK-301, M-7824, HTI-1088 (HTI-131 , SHR-1316) , MSB-2311, AK-106, AVA-004, BBI-801, CA-327, CBA-0710, CBT-502, FPT-155, IKT-201, IKT-703, 10-103, JS-003, KD-03
  • the immune checkpoint inhibitor is an anti-CTLA-4 antibody.
  • anti-CTLA-4 antibodies include, but are not limited to, ipilimumab (IBI310, BMS-734016, MDX010, MDX-CTLA4, MEDI4736) , tremelimumab (CP-675, CP-675, 206) , APL-509, AGEN1884, and CS1002, AGEN1181, Abatacept (Orencia, BMS-188667, RG2077) , BCD-145, ONC-392, ADU-1604, REGN4659, ADG116, KN044, KN046, biosimilars thereof and derivatives thereof.
  • art recognized anti-CTLA-4 antibodies can be used.
  • the antibodies that compete with any of these art-recognized antibodies for binding to CTLA-4 also can be used.
  • the anti-CTLA-4 antibody is ADG116.
  • ADG116 also known as TY21580
  • related anti-CTLA-4 antibodies have been described, for example, in WO2019/149281, which is incorporated herein by reference in its entirety.
  • a method of treating a lung cancer in a subject comprising administering to the subject: (a) an effective amount of any one of the anti-CD137 antibodies described herein, and (b) an effective amount of an anti-PD-L1 antibody.
  • the anti-PD-L1 antibody is atezolizumab, a biosimilar thereof, or a derivative thereof.
  • the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 2, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 3, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 4, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 5, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 6, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 7.
  • the anti-CD137 antibody comprises a VH comprising the amino acid sequence of SEQ ID NO: 8, and/or a VL comprises the amino acid sequence of SEQ ID NO: 9.
  • the anti-CD137 antibody comprises a heavy chain and a light chain, and wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 10, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 11.
  • the anti-PD-L1 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 56, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 57, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 58, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 59, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 60, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 61.
  • the anti-PD-L1 antibody comprises a VH comprising the amino acid sequence of SEQ ID NO: 62, and/or a VL comprising the amino acid sequence of SEQ ID NO: 63.
  • a method of treating a lung cancer in a subject comprising administering to the subject: (a) an effective amount of any one of the anti-CD137 antibodies described herein, and (b) an effective amount of an anti-PD-1 antibody.
  • the anti-PD-1 antibody is 2E5, a biosimilar thereof, or a derivative thereof.
  • the anti-PD-1 antibody is toripalimab, a biosimilar thereof, or a derivative thereof.
  • the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 2, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 3, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 4, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 5, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 6, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 7.
  • the anti-PD-1 antibody comprises a VH and a VL
  • the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 64, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 65, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 66
  • the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 67, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 68, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 69.
  • a method of treating a breast cancer comprising administering to the subject: (a) an effective amount of any one of the anti-CD137 antibodies described herein, and (b) an effective amount of an anti-CTLA-4 antibody.
  • the anti-CTLA-4 antibody is ADG116, a biosimilar thereof, or a derivative thereof.
  • the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 2, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 3, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 4, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 5, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 6, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 7.
  • the anti-CD137 antibody comprises a VH comprising the amino acid sequence of SEQ ID NO: 8, and/or a VL comprises the amino acid sequence of SEQ ID NO: 9.
  • the anti-CD137 antibody comprises a heavy chain and a light chain, and wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 10, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 11.
  • a method of treating a lung cancer in a subject comprising administering to the subject: (a) an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and (b) an effective amount of a chemotherapeutic agent.
  • a method of treating a breast cancer comprising administering to the subject: (a) an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and (b) an effective amount of a chemotherapeutic agent.
  • chemotherapeutic agents include, but are not limited to, taxoids, e.g., paclitaxel, docetaxel; and platinum coordination complexes, such as cisplatin, oxaliplatin, and carboplatin.
  • the chemotherapeutic agent is docetaxel (e.g., ) .
  • the chemotherapeutic agent is cisplatin (e.g., or ) .
  • a method of treating a breast cancer comprising administering to the subject: (a) an effective amount of any one of the anti-CD137 antibodies described herein, and (b) an effective amount of a taxoid (e.g., docetaxel) .
  • a taxoid e.g., docetaxel
  • the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 2, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 3, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 4, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 5, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 6, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 7.
  • a method of treating a lung cancer in a subject comprising administering to the subject: (a) an effective amount of any one of the anti-CD137 antibodies described herein, and (b) an effective amount of a platinum coordination complex (e.g., cisplatin) .
  • a platinum coordination complex e.g., cisplatin
  • the anti-CD137 antibody comprises a VH comprising the amino acid sequence of SEQ ID NO: 8, and/or a VL comprises the amino acid sequence of SEQ ID NO: 9.
  • the anti-CD137 antibody comprises a heavy chain and a light chain, and wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 10, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 11.
  • a method of treating a lung cancer in a subject comprising administering to the subject: (a) an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and (b) an effective amount of an anti-CD20 antibody.
  • a method of treating a lung cancer in a subject comprising administering to the subject: (a) an effective amount of any one of the anti-CD137 antibodies described herein, and (b) an effective amount of an anti-CD20 antibody (e.g., rituximab) .
  • an anti-CD20 antibody e.g., rituximab
  • the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 2, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 3, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 4, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 5, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 6, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 7.
  • the anti-CD137 antibody comprises a VH comprising the amino acid sequence of SEQ ID NO: 8, and/or a VL comprises the amino acid sequence of SEQ ID NO: 9.
  • the anti-CD137 antibody comprises a heavy chain and a light chain, and wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 10, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 11.
  • a method of treating a colorectal (e.g., colon) cancer in a subject comprising administering to the subject: (a) an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and (b) an effective amount of a radiation therapy.
  • Exemplary radiation therapies include, but are not limited to, ionizing (electromagnetic) radiotherapy (e.g., X-rays or gamma rays) and particle beam radiation therapy (e.g., high linear energy radiation) .
  • the source of radiation can be external or internal to the subject.
  • the radiation therapy is local radiation.
  • the radiation therapy is single dose radiation.
  • the radiation therapy is high-dose radiation, e.g., at least about any one of 10, 20, 30, 40, 50, or more Gy.
  • a method of treating a colon cancer in a subject comprising administering to the subject: (a) an effective amount of any one of the anti-CD137 antibodies described herein, and (b) an effective amount of a radiation therapy (e.g., high-energy x-rays, involved-site radiation therapy or ISRT) .
  • a radiation therapy e.g., high-energy x-rays, involved-site radiation therapy or ISRT
  • the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 2, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 3, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 4, and wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 5, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 6, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 7.
  • the anti-CD137 antibody comprises a VH comprising the amino acid sequence of SEQ ID NO: 8, and/or a VL comprises the amino acid sequence of SEQ ID NO: 9.
  • the anti-CD137 antibody comprises a heavy chain and a light chain, and wherein the heavy chain comprises the amino acid sequence of SEQ ID NO: 10, and/or the light chain comprises the amino acid sequence of SEQ ID NO: 11.
  • the radiation therapy is local radiation.
  • biomarkers which can be used in conjunction with any one of the methods of treatment described herein.
  • Suitable biomarkers include total CD137, membrane bound CD137 (mCD137) , soluble CD137 (sCD137) , CD137L, Ki67, PD-L1, Fc ⁇ RIIb, Microsatellite instability (MSI) , inflammatory cytokines (e.g., TNF ⁇ , IFN ⁇ , IL-2, IL-6, IL-10) , peripheral blood immune cell profiles, such as absolute cell counts for circulating T cells, natural killer (NK) cells, B cells, effector T cell subpopulations (e.g., CD4+ and CD8+ T cells) , and memory T cell subpopulations (e.g., effector memory T (T em ) cells) , and regulatory T (T reg ) cells.
  • mCD137 membrane bound CD137
  • sCD137 soluble CD137
  • CD137L Ki67
  • PD-L1 Ki67
  • a method of treating or delaying progression of cancer in a subject by administering an effective amount of an anti-CD137 antibody based on a level of one or more biomarkers selected from the group consisting of mCD137, sCD137, CD137L, Ki67, PD-L1, Fc ⁇ RIIb, MSI, TNF ⁇ , IFN ⁇ , IL-2, IL-6, IL-10, NK cells, T em cells and T reg cells in one or more samples obtained from the subject.
  • a method of determining whether a subject is likely to respond to a therapy comprising an anti-CD137 antibody by determining a level of one or more biomarkers selected from the group consisting of total CD137, mCD137, sCD137, CD137L, Ki67, PD-L1, Fc ⁇ RIIb, MSI, TNF ⁇ , IFN ⁇ , IL-2, IL-6, IL-10, NK cells, T em cells and T reg cells in one or more samples obtained from the subject.
  • a method of treating or delaying progression of cancer in a subject by administering an effective amount of an anti-CD137 antibody after it has been determined that the subject is likely to respond to the anti-CD137 antibody.
  • the therapy further comprises an immune checkpoint inhibitor, such as an anti-PD-1 antibody.
  • a method of predicting responsiveness and/or monitoring treatment and/or responsiveness of a subject to a therapy comprising an anti-CD137 antibody by determining a level of one or more biomarkers selected from the group consisting of total CD137, mCD137, sCD137, CD137L, Ki67, PD-L1, Fc ⁇ RIIb, MSI, TNF ⁇ , IFN ⁇ , IL-2, IL-6, IL-10, NK cells, T em cells and T reg cells in one or more samples obtained from the subject.
  • the therapy further comprises an immune checkpoint inhibitor, such as an anti-PD-1 antibody.
  • the treatment comprises administration of an anti-CD137 antibody.
  • the treatment further comprises administration of an immune checkpoint inhibitor, such as an anti-PD-1 antibody.
  • an assay for determining a level of one or more biomarkers selected from the group consisting of total CD137, mCD137, sCD137, CD137L, Ki67, PD-L1, Fc ⁇ RIIb, MSI, TNF ⁇ , IFN ⁇ , IL-2, IL-6, IL-10, NK cells, T em cells and T reg cells in one or more samples obtained from the subject.
  • the assay is an immunohistochemistry (IHC) assay.
  • the assay is a multiplex IHC assay capable of detecting two or more biomarkers.
  • the assay is an immunoassay, such as a Meso Scale Discovery (MSD) assay.
  • a method of treating a cancer in a subject comprising administering to the subject an effective amount of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1; and wherein the subject has a high level in one or more biomarkers selected from the group consisting of total CD137, membrane bound CD137 (mCD137) , CD137L and PD-L1 and/or a low level of CD8+ effector memory T (T em ) cells or natural killer (NK) cells compared to a reference level.
  • an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137, wherein the antibody binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89,
  • the level of one or more biomarkers comprises a level of CD137 (e.g., in a tumor biopsy sample) .
  • the level of one or more biomarkers comprises a level of sCD137 in a blood sample (e.g., a plasma sample) .
  • the level of one or more biomarkers comprises a level of mCD137 on CD8 + T cells.
  • the level of one or more biomarkers comprises a level of Ki67 on CD8 + T cells.
  • the CD8 + T cells are tumor infiltrating T cells.
  • the level of one or more biomarkers comprises a level of CD137L (e.g., in a tumor biopsy sample) .
  • the level of one or more biomarkers comprises a level of NK cells in a blood sample.
  • the sample is a tumor biopsy sample.
  • the sample is a FFPE sample.
  • the level of the one or more biomarkers is detected by IHC.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is selected from the group consisting of colon cancer (e.g., Sigmoid colon cancer) , breast cancer, lung cancer (e.g., non-small cell lung cancer or NSCLC) , esophageal cancer, endometrial cancer, gastrointestinal cancer (e.g., gastrointestinal neuroectodermal tumor) , cholangiocarcinoma, nasopharyngeal cancer (NPC) , adenoid cystic carcinoma (ACC) , melanoma, mesothelioma (e.g., malignant pleural mesothelioma or MPM) , anal cancer, head and neck cancer (e.g., head and neck squamous cell carcinoma or HNSCC) , appendiceal and sebaceous cancer, mantle cell lymphoma, follicular lymphoma, non-Hodgkin’s lymphoma (NHL) , and T cell lymphoma (e.g.,
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • the method further comprises administering to the subject an effective amount of an anti-PD-1 antibody (e.g., toripalimab) .
  • an anti-PD-1 antibody e.g., toripalimab
  • the level of one or more biomarkers comprises a level of CD137 (e.g., in a tumor biopsy sample) .
  • the level of one or more biomarkers comprises a level of sCD137 in a blood sample (e.g., a plasma sample) .
  • the level of one or more biomarkers comprises a level of mCD137 on CD8+ T cells.
  • the level of one or more biomarkers comprises a level of Ki67 on CD8+ T cells.
  • the CD8+ T cells are tumor infiltrating T cells.
  • the level of one or more biomarkers comprises a level of CD137L (e.g., in a tumor biopsy sample) .
  • the level of one or more biomarkers comprises a level of NK cells in a blood sample.
  • the sample is a tumor biopsy sample.
  • the sample is an FFPE sample.
  • the level of the one or more biomarkers is detected by IHC.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is selected from the group consisting of colon cancer (e.g., Sigmoid colon cancer) , breast cancer, lung cancer (e.g., non-small cell lung cancer or NSCLC) , esophageal cancer, endometrial cancer, gastrointestinal cancer (e.g., gastrointestinal neuroectodermal tumor) , cholangiocarcinoma, nasopharyngeal cancer (NPC) , adenoid cystic carcinoma (ACC) , melanoma, mesothelioma (e.g., malignant pleural mesothelioma or MPM) , anal cancer, head and neck cancer (e.g., head and neck squamous cell carcinoma or HNSCC) , appendiceal and sebaceous cancer, mantle cell lymphoma, follicular lymphoma, non-Hodgkin’s lymphoma (NHL) , and T cell lymphoma (e.g.,
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • a prior therapy such as an immunotherapy.
  • an increased level of one or more biomarkers selected from the group consisting of total CD137, sCD137, Ki67, CD137L, NK cells, and CD8 T em cells, and/or a decreased level of one or more biomarkers selected from the group consisting of mCD137 and T reg cells after administration of the anti-CD137 antibody compared to the level of the one or more biomarkers before administration of the anti-CD137 antibody indicates that the subject may benefit from the administration of the ant-CD137 antibody.
  • the method further comprises administering to the subject an effective amount of the anti-CD137 antibody.
  • the method further comprises administering to the subject an effective amount of an anti-PD-1 antibody (e.g., toripalimab) .
  • the level of one or more biomarkers comprises a level of CD137 (e.g., in a tumor biopsy sample) .
  • the level of one or more biomarkers comprises a level of sCD137 in a blood sample (e.g., a plasma sample) .
  • the level of one or more biomarkers comprises a level of mCD137 on CD8+ T cells.
  • the level of one or more biomarkers comprises a level of Ki67 on CD8+ T cells.
  • the CD8+T cells are tumor infiltrating T cells.
  • the level of one or more biomarkers comprises a level of CD137L (e.g., in a tumor biopsy sample) .
  • the level of one or more biomarkers comprises a level of NK cells in a blood sample.
  • the sample is a tumor biopsy sample.
  • the sample is an FFPE sample.
  • the level of the one or more biomarkers is detected by IHC.
  • the anti-CD137 antibody is administered at a dose of no more than 500 mg (e.g., about 150 mg to about 500 mg, about 150 mg to about 300 mg, or about 300 mg to about 400 mg) .
  • the anti-CD137 antibody is administered at a dose of about 0.1 mg/kg to about 10 mg/kg, such as about 3 mg/kg to about 8 mg/kg, e.g., about 3 mg/kg, about 5 mg/kg, or about 8 mg/kg.
  • the anti-CD137 antibody is administered intravenously.
  • the anti-CD137 antibody is administered about once every three weeks.
  • the cancer is selected from the group consisting of colon cancer (e.g., Sigmoid colon cancer) , breast cancer, lung cancer (e.g., non-small cell lung cancer or NSCLC) , esophageal cancer, endometrial cancer, gastrointestinal cancer (e.g., gastrointestinal neuroectodermal tumor) , cholangiocarcinoma, nasopharyngeal cancer (NPC) , adenoid cystic carcinoma (ACC) , melanoma, mesothelioma (e.g., malignant pleural mesothelioma or MPM) , anal cancer, head and neck cancer (e.g., head and neck squamous cell carcinoma or HNSCC) , appendiceal and sebaceous cancer, mantle cell lymphoma, follicular lymphoma, non-Hodgkin’s lymphoma (NHL) , and T cell lymphoma (e.g.,
  • the cancer is resistant or refractory to a prior therapy, such as an immunotherapy.
  • the method further comprises administering to the subject an effective amount of an anti-PD-1 antibody (e.g., toripalimab) .
  • an anti-PD-1 antibody e.g., toripalimab
  • the method comprises determining the level of total CD137 in a sample of the subject. In some embodiments, determining the level of CD137 in a sample comprises measuring the level of CD137 in a tumor biopsy sample, such as an FFPE sample. In some embodiments, determining the level of CD137 in a sample comprises measuring the level of protein expression of CD137. In some embodiments, an increase in the level of CD137 after receiving an anti-CD137 antibody in a subject indicates that the subject is likely to respond to the anti-CD137 antibody treatment, e.g., the subject is likely to have stable disease.
  • the level of CD137 increases by at least about 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 2 fold, 3 fold, 4 fold, 5 fold or more in a subject likely to respond to the anti-CD137 antibody treatment after the subject receives an anti-CD137 antibody than before the subject receives the anti-CD137 antibody.
  • the level of total CD137 is percentage of nucleated cells expressing CD137 as determined by an IHC assay.
  • an increase in the CD137 level of at least about 5%after the subject receives an anti-CD137 antibody than before the subject receives the anti-CD137 antibody indicates that the subject is likely to benefit from the anti-CD137 antibody treatment.
  • the method comprises determining the level of mCD137 in a sample of the subject. In some embodiments, determining the level of mCD137 in a sample comprises measuring the level of mCD137 on CD8 + T cells, such as tumor infiltrating T cells. In some embodiments, determining the level of mCD137 in a sample comprises measuring the level of protein expression of mCD137. In some embodiments, the amino acid sequence of human mCD137 is SEQ ID NO: 1. In some embodiments, a decrease in the level of mCD137 after receiving an anti-CD137 antibody in a subject indicates that the subject is likely to respond to the anti-CD137 antibody treatment, e.g., the subject is likely to have stable disease.
  • the level of mCD137 decreases by at least about 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%or more in a subject likely to respond to the anti-CD137 antibody treatment after the subject receives an anti-CD137 antibody than before the subject receives the anti-CD137 antibody.
  • a high level of mCD137 prior to receiving an anti-CD137 antibody in a subject compared to a reference level indicates that the subject is likely to respond to the anti-CD137 antibody treatment.
  • the high level is at least about any one of 50%, 100%, 150%, 2 fold, 2.5 fold, 3 fold, 4 fold, 5 fold or more than the reference level.
  • the method comprises determining the level of sCD137 in a sample of the subject. In some embodiments, determining the level of sCD137 in a sample comprises measuring the level of sCD137 in a blood sample such as a blood sample (e.g., a plasma sample) . In some embodiments, determining the level of sCD137 in a sample comprises measuring the level of protein expression of sCD137. In some embodiments, the amino acid sequence of human sCD137 is SEQ ID NO: 43. In some embodiments, an increase in the level of sCD137 after receiving an anti-CD137 antibody in a subject indicates that the subject is likely to respond to the anti-CD137 antibody treatment, e.g., the subject is likely to have stable disease.
  • the level of sCD137 increases by at least about 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 2 fold, 3 fold, 4 fold, 5 fold, 6 fold, 7 fold, 8 fold, 9 fold, 10 fold, 12 fold, 15 fold, 20 fold or more in a subject likely to respond to the anti-CD137 antibody treatment after the subject receives an anti-CD137 antibody than before the subject receives the anti-CD137 antibody.
  • the method comprises determining a ratio ( “induction ratio” ) between the difference in the sCD137 level after administration of the anti-CD137 antibody (C2) and the sCD137 level before administration of the anti-CD137 antibody (C1) and the sCD137 level before administration of the anti-CD137 antibody (C1) : (C2-C1) /C1.
  • an induction ratio of at least about any one of 5, 6, 7, 8, 9, 10 or more indicates a high likelihood of responding to the anti-CD137 antibody treatment, i.e., having stable disease (SD) or partial response (PR) after the treatment.
  • the method comprises determining the level of CD137L in a sample. In some embodiments, determining the level of CD137L in a sample comprises measuring the level of CD137L in a tumor tissue. In some embodiments, determining the level of CD137L in a sample comprises measuring the level of CD137L in a tumor biopsy sample, such as an FFPE sample. In some embodiments, determining the level of CD137L in a sample comprises measuring the level of expression of a nucleic acid molecule encoding CD137L (e.g., measuring the level of RNA (such as pre-mRNA or mRNA) transcript expression from a gene encoding CD137L) and/or measuring the level of protein expression of CD137L.
  • a nucleic acid molecule encoding CD137L e.g., measuring the level of RNA (such as pre-mRNA or mRNA) transcript expression from a gene encoding CD137L
  • the nucleic acid sequence encoding human CD137L is SEQ ID NO: 44.
  • the amino acid sequence of human CD137L is SEQ ID NO: 45.
  • Methods for measuring the level of CD137L have been described, for example, in WO2019105468A1, which is incorporated herein by reference in its entirety.
  • an increase in the level of CD137L after receiving an anti-CD137 antibody in a subject indicates that the subject is likely to respond to the anti-CD137 antibody treatment, e.g., the subject is likely to have stable disease.
  • the level of CD137L increases by at least about 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%or more in a subject likely to respond to the anti-CD137 antibody treatment after the subject receives an anti-CD137 antibody than before the subject receives the anti-CD137 antibody.
  • a high level of CD137L prior to receiving an anti-CD137 antibody in a subject compared to a reference level indicates that the subject is likely to respond to the anti-CD137 antibody treatment.
  • the high level is at least about any one of 50%, 100%, 150%, 2 fold, 2.5 fold, 3 fold, 4 fold, 5 fold or more than the reference level.
  • the level of CD137L is percentage of nucleated cells expressing CD137 as determined by an IHC assay. In some embodiments, a CD137L level of at least about 15%, such as at least about any one of 15%, 20%, 25%, 30%, 35%, 40%, 45%, or more, indicates that the subject is likely to respond to the anti-CD137 antibody treatment.
  • a method of selecting a subject for a therapy comprising administration of an anti-CD137 antibody comprising: a) determining a level of CD137L in a sample of the subject, and b) selecting the subject for the therapy if the level of CD137L is higher than a reference level.
  • the reference level is expression of CD137L in at least about 15%of nucleated cells in the sample.
  • the sample is a tumor biopsy sample (e.g., an FFPE sample) .
  • the level of CD137L is determined by IHC.
  • the therapy further comprises administering to the subject an effective amount of an anti-PD-1 antibody (e.g., toripalimab) .
  • a method of treating cancer in a subject comprising administering an effective amount of an anti-CD137 antibody to the subject, if the level of CD137L in a sample of the subject is higher than a reference level.
  • the reference level is expression of CD137L in at least about 15%of nucleated cells in the sample.
  • the sample is a tumor biopsy sample (e.g., an FFPE sample) .
  • the level of CD137L is determined by IHC.
  • the method further comprises administering to the subject an effective amount of an anti-PD-1 antibody (e.g., toripalimab) .
  • the method further comprises the steps of: a) obtaining the sample from the subject; and b) measuring the level of expression of CD137L in the sample prior to administration of the anti-CD137 antibody to the subject.
  • a method of treating cancer in a subject comprising administering an effective amount of an anti-CD137 antibody to the subject, wherein it has been determined that the subject is likely to respond to the anti-CD137 antibody when the level of CD137L in a sample of the subject is higher than a reference level.
  • the reference level is expression of CD137L in at least about 15%of nucleated cells in the sample.
  • the sample is a tumor biopsy sample (e.g., an FFPE sample) .
  • the level of CD137L is determined by IHC.
  • the method further comprises administering to the subject an effective amount of an anti-PD-1 antibody (e.g., toripalimab) .
  • a method of determining if a subject is likely to respond to a therapy comprising an anti-CD137 antibody comprising: a) obtaining a sample from the subject; b) measuring the level of expression of CD137L in the sample; and c) determining that the subject is likely to respond to the therapy when the level of expression of CD137L in the sample is higher than a reference level.
  • the reference level is expression of CD137L in at least about 15%of nucleated cells in the sample.
  • the sample is a tumor biopsy sample (e.g., an FFPE sample) .
  • the level of CD137L is determined by IHC.
  • the therapy further comprises administering to the subject an effective amount of an anti-PD-1 antibody (e.g., toripalimab) .
  • the CD137L expression level is determined using an anti-CD137L antibody.
  • anti-CD137L antibodies suitable for determination of the CD137L expression level have been described, for example, in PCT/CN2020/094371, which is incorporated herein by reference in its entirety.
  • the anti-CD137L antibody binds to an intracellular or transmembrane region of human CD137 ligand (CD137L) .
  • the anti-CD137L antibody specifically binds to a peptide comprising the amino acid sequence of MEYASDASLDPEAPWPPAPRARACRVLP (SEQ ID NO: 72) and/or binds to a peptide comprising the amino acid sequence of MEYASDASLDPEAPWPPAPRARA (SEQ ID NO: 73) .
  • the anti-CD137L antibody comprises a heavy chain variable region comprises an HVR-H1 comprising the amino acid sequence of SEQ ID NO: 74, an HVR-H2 comprising the amino acid sequence of SEQ ID NO: 75, and an HVR- H3 comprising the amino acid sequence of SEQ ID NO: 76, and a light chain variable region comprises an HVR-L1 comprising the amino acid sequence of SEQ ID NO: 77, an HVR-L2 comprising the amino acid sequence of SEQ ID NO: 78, and an HVR-L3 comprising the amino acid sequence of SEQ ID NO: 79.
  • the anti-CD137L antibody comprises a heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 80, and a light chain variable region comprises the amino acid sequence of SEQ ID NO: 81.
  • the anti-CD137L antibody is TY23561.
  • the CD137L expression level is determined by contacting a human tissue sample (e.g., a tumor biopsy sample) with the anti-CD137L antibody and detecting binding of the antibody or antigen-binding fragment to the sample by immunohistochemistry (IHC) , binding of the anti-CD137L antibody to the sample indicates the level of expression of human CD137L in the sample.
  • a human tissue sample e.g., a tumor biopsy sample
  • IHC immunohistochemistry
  • the method comprises determining the level of PD-L1 in a sample. In some embodiments, determining the level of PD-L1 in a sample comprises measuring the level of PD-L1 on tumor cells. In some embodiments, determining the level of PD-L1 in a sample comprises measuring the level of expression of a nucleic acid molecule encoding PD-L1 (e.g., measuring the level of RNA (such as pre-mRNA or mRNA) transcript expression from a gene encoding PD-L1) and/or measuring the level of protein expression of PD-L1. In some embodiments, the nucleic acid sequence encoding human PD-L1 has GenBank Accession No. NM_001267706.1.
  • the amino acid sequence of human PD-L1 is SEQ ID NO: 46.
  • a decrease in the level of PD-L1 after receiving an anti-CD137 antibody in a subject indicates that the subject is likely to respond to the anti-CD137 antibody treatment, e.g., the subject is likely to have stable disease.
  • the level of PD-L1 decreases by at least about 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%or more in a subject likely to respond to the anti-CD137 antibody treatment after the subject receives an anti-CD137 antibody than before the subject receives the anti-CD137 antibody.
  • a high level of PD-L1 prior to receiving an anti-CD137 antibody in a subject compared to a reference level indicates that the subject is likely to respond to the anti-CD137 antibody treatment.
  • the high level is at least about any one of 50%, 100%, 150%, 2 fold, 2.5 fold, 3 fold, 4 fold, 5 fold or more than the reference level.
  • the method comprises determining the level of Ki67 in a sample. In some embodiments, determining the level of Ki67 in a sample comprises measuring the level of Ki67 on CD8 + T cells, such as tumor infiltrating T cells. In some embodiments, determining the level of Ki67 in a sample comprises measuring the level of expression of a nucleic acid molecule encoding Ki67 (e.g., measuring the level of RNA (such as pre-mRNA or mRNA) transcript expression from a gene encoding Ki67) and/or measuring the level of protein expression of Ki67. In some embodiments, the nucleic acid sequence encoding human Ki67 has GenBank Accession No. NM_001145966.2.
  • the amino acid sequence of human Ki67 is SEQ ID NO: 47.
  • an increase in the level of Ki67 after receiving an anti-CD137 antibody in a subject indicates that the subject is likely to respond to the anti-CD137 antibody treatment, e.g., the subject is likely to have stable disease.
  • the level of Ki67 increases by at least about 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 2 fold, 3 fold, 4 fold, 5 fold or more in a subject likely to respond to the anti-CD137 antibody treatment after the subject receives an anti-CD137 antibody than before the subject receives the anti-CD137 antibody.
  • the level of a biomarker in a sample is measured by determining the level of RNA transcript expression of the biomarker. Suitable methods of measuring RNA transcript levels in a sample are known in the art, including, for example, by Northern blot analysis, nuclease protection assays, in situ hybridization, PCR analysis (e.g., qPCR, RT-PCR, RT-qPCR, etc. ) , and next generation sequencing (e.g., RNAseq) . In some embodiments, the level of transcript expression of the biomarker is measured by RT-PCR, in situ hybridization, and/or RNAseq.
  • the level of a biomarker in a sample is measured by determining the level of protein expression of the biomarker.
  • Suitable methods of measuring protein expression in a sample are known in the art, including, for example, immunoassays (e.g., Meso Scale Discovery or MSD assay) , immunohistochemistry (IHC) , PET imaging, Western blotting, enzyme-linked immunosorbent assays (ELISAs) , flow cytometry, and mass spectrometry.
  • the level of protein expression of the biomarker is measured by immunoassay, Western blotting, ELISA, IHC, and/or flow cytometry.
  • the biomarker is MSI.
  • MSI is determined from a plurality of loci that are associated with microsatellite instability. In some embodiments, the MSI is determined from at least about 50 loci, at least about 60 loci, at least about 70 loci, at least about 80 loci, at least about 90 loci, at least about 100 loci, or more.
  • the biomarker is a cell population.
  • Suitable methods for determining levels of cell populations in a sample are known in the art, including, for example, fluorescence-activated cell sorting (FACS) .
  • FACS fluorescence-activated cell sorting
  • the method comprises determining the level of NK cells in a sample, such as a blood sample, e.g., peripheral blood sample.
  • a sample such as a blood sample, e.g., peripheral blood sample.
  • an increase in the level of NK cells after receiving an anti-CD137 antibody in a subject indicates that the subject is likely to respond to the anti-CD137 antibody treatment, e.g., the subject is likely to have stable disease.
  • the level of NK cells increases by at least about 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%or more in a subject likely to respond to the anti-CD137 antibody treatment after the subject receives an anti-CD137 antibody than before the subject receives the anti-CD137 antibody.
  • the method comprises determining a ratio ( “induction ratio” ) between the difference in the NK cell level after administration of the anti-CD137 antibody (C2) and the NK cell level before administration of the anti-CD137 antibody (C1) and the NK cell level before administration of the anti-CD137 antibody (C1) : (C2-C1) /C1.
  • an induction ratio of at least about any one of 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more indicates a high likelihood of responding to the anti-CD137 antibody treatment, i.e., having stable disease (SD) or partial response (PR) after the treatment.
  • a low level of NK cells prior to receiving an anti-CD137 antibody in a subject compared to a reference level indicates that the subject is likely to respond to the anti-CD137 antibody treatment.
  • the low level is no more than any one of 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%or less than the reference level.
  • the method comprises determining the level of T em cells in a sample.
  • the T em cells are CD8+ T em cells.
  • the T em cells are CD45RO+ CCR7-L-selectin-T cells.
  • the T em cells have intermediate to high expression of CD44.
  • the level of T em cells increases by at least about 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 2 fold, 3 fold, 4 fold, 5 fold or more in a subject likely to respond to the anti-CD137 antibody treatment after the subject receives an anti-CD137 antibody than before the subject receives the anti-CD137 antibody.
  • a low level of T em cells prior to receiving an anti-CD137 antibody in a subject compared to a reference level indicates that the subject is likely to respond to the anti-CD137 antibody treatment.
  • the low level is no more than any one of 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%or less than the reference level.
  • the method comprises determining the level of T reg cells in a sample.
  • the T reg cells are CD4+ CD25+ FOXP3+ T cells.
  • a decrease in the level of T reg cells after receiving an anti-CD137 antibody in a subject indicates that the subject is likely to respond to the anti-CD137 antibody treatment, e.g., the subject is likely to have stable disease.
  • the level of T reg cells decreases by at least about 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%or more in a subject likely to respond to the anti-CD137 antibody treatment after the subject receives an anti-CD137 antibody than before the subject receives the anti-CD137 antibody.
  • the level of one or more biomarkers is measured in one or more (e.g., one or more, two or more, three or more, four or more, etc. ) samples obtained from a subject.
  • Any suitable sample in the form of tissues and/or fluids that are known or believed to contain diseased cells and/or the target of interest may be used in the methods described herein, including, for example, sputum, pleural fluid, lymph fluid, bone marrow, blood, plasma, serum, urine, tissue samples (samples known or expected to contain cancer cells) , tumor samples, tumor biopsies, etc.
  • the sample is a serum sample.
  • the sample is a tumor sample.
  • the sample is a tumor biopsy.
  • the sample comprises one or more cancer cells.
  • tissue and/or fluid samples e.g., methods that are appropriate for obtaining a representative sample from a particular type, location, disease tissue, etc.
  • suitable tissue and/or fluid samples include, for example, by resection, bone marrow biopsy or bone marrow aspiration, endoscopic biopsy or endoscopic aspiration (e.g., cystoscopy, bronchoscopy, colonoscopy, etc. ) , needle biopsy or needle aspiration (e.g., fine needle aspiration, core needle biopsy, vacuum-assisted biopsy, image-guided biopsy, etc. ) skin biopsy (e.g., shave biopsy, punch biopsy, incisional biopsy, excisional biopsy, etc. ) , various other surgical tissue (e.g., tumor tissue) biopsy and/or excision strategies, and fluid collections (e.g., collecting urine, blood, serum, plasma, sputum, etc. ) .
  • endoscopic biopsy or endoscopic aspiration e.g., cystoscopy, bronchos
  • the one or more samples obtained from the subject are enriched for diseased (e.g., cancerous) cells.
  • Methods of enriching a tissue or fluid preparation for diseased (e.g., cancerous) cells are known in the art, including, for example, by separating diseased (e.g., cancerous) cells from normal cells by flow cytometry.
  • the level of one or more biomarkers is measured in the enriched samples. In some embodiments, the level of one or more biomarkers is measured in samples that have not been enriched or otherwise altered after isolation.
  • the one or more samples are fixed (i.e. preserved) by conventional methodology (See e.g., “Manual of Histological Staining Method of the Armed Forces Institute of Pathology, ” 3 rd edition (1960) Lee G. Luna, HT (ASCP) Editor, The Blakston Division McGraw-Hill Book Company, New York; The Armed Forces Institute of Pathology Advanced Laboratory Methods in Histology and Pathology (1994) Ulreka V. Mikel, Editor, Armed Forces Institute of Pathology, American Registry of Pathology, Washington, D.C. ) .
  • the choice of a fixative may be determined by one of ordinary skill in the art for the purpose for which the sample is to be analyzed.
  • the length of fixation will depend upon the size and type of the tissue sample and the fixative used (e.g., neutral buffered formalin, paraformaldehyde, etc. ) , as will be appreciated by one of ordinary skill in the art.
  • the level of one or more biomarkers is measured in a sample that is fixed. In some embodiments, the level of one or more biomarkers is measured in samples that have not been fixed or otherwise altered after isolation.
  • one or more samples are obtained from the subject prior to administration with an anti-CD137 antibody. In some embodiments, one or more samples are obtained from the subject after administration of a first and/or subsequent dose of an anti-CD137 antibody. In some embodiments, one or more samples are obtained from the subject after completion of an anti-CD137 antibody therapy. In some embodiments, one or more samples are obtained from the subject, prior to, during, and after completion of an anti-CD137 antibody therapy.
  • the method comprises comparing the level of a biomarker in a sample obtained from a subject to a reference level of the biomarker.
  • the reference level is the level of the biomarker in a reference sample (e.g., a reference cell (such as a cell line, including but not limited to Raji (ATCC, CC-86) or Daudi (ATCC, CCL-213) cell lines) , a corresponding sample taken from one or more patients determined to be responsive to anti-CD137 antibody therapy, a corresponding sample taken from one or more patients determined to be non-responsive to anti-CD137 antibody therapy, a corresponding adjacent normal tissue, etc. ) .
  • a reference sample e.g., a reference cell (such as a cell line, including but not limited to Raji (ATCC, CC-86) or Daudi (ATCC, CCL-213) cell lines
  • the reference level is measured in the reference sample using the same method as was used to measure the level of the biomarker in the subject’s sample. In some embodiments, the reference level is measured in the reference sample using a different method than was used to measure the level of the biomarker in the subject’s sample.
  • the reference level is a pre-determined level of a biomarker (e.g., the average level of the biomarker in a database of diseased samples (such as tissue biopsies or serum samples) isolated from multiple reference patients; the average level of the biomarker in a database of samples (such as tissue biopsies or serum samples) isolated from multiple healthy reference patients; etc. ) .
  • a biomarker e.g., the average level of the biomarker in a database of diseased samples (such as tissue biopsies or serum samples) isolated from multiple reference patients; the average level of the biomarker in a database of samples (such as tissue biopsies or serum samples) isolated from multiple healthy reference patients; etc.
  • the reference level of a biomarker refers to a detectable level of expression. That is to say, in some embodiments, the level of a biomarker measured in the sample obtained from the subject is considered to be lower than a reference level when the level of the biomarker in the sample is undetectable, e.g., below the limit of detection.
  • the level of a biomarker measured in the sample obtained from the subject is considered to be lower than the reference level when the level of the biomarker in the sample is at least about 25%lower than the reference level.
  • the level of a biomarker measured in the sample obtained from the subject is considered to be lower than the reference level when the level of the biomarker in the sample is at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99%lower than the reference level.
  • the level of a biomarker measured in the sample obtained from the subject is considered to be lower than the reference level when the level of the biomarker in the sample is at least about 1-fold lower than the reference level.
  • the level of a biomarker measured in the sample obtained from the subject is considered to be lower than the reference level when the level of the biomarker in the sample is at least about 1-fold, at least about 1.5-fold, at least about 2-fold, at least about 2.5-fold, at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 4.5-fold, at least about 5-fold, at least about 5.5-fold, at least about 6-fold, at least about 6.5-fold, at least about 7-fold, at least about 7.5 fold, at least about 8-fold, at least about 8.5-fold, at least about 9-fold, at least about 9.5-fold, at least about 10-fold, at least about 100-fold, or at least about 1000-fold lower than the reference level.
  • the level of a biomarker in the sample obtained from the subject is below the limit of detection. In some embodiments, the level of a biomarker measured in the sample obtained from the subject is considered to be lower than the reference level when the level of the biomarker in the sample is below the limit of detection while the reference level is above the limit of detection, is detectable, and/or is not zero.
  • a level is considered to be below the limit of detection when the level does not give an appreciable signal, a detectable signal, and/or is not significantly different than an appropriate negative control when performing an assay for measuring the level of a biomarker (e.g., below the limit of detection of an assay measuring RNA transcript expression of the biomarker (such as RT-PCR, in situ hybridization, and/or next generation sequencing) , below the limit of detection of an assay measuring protein expression of a biomarker (such as an immunoassay, PET imaging, Western blotting, ELISA, immunohistochemistry, and/or flow cytometry) , etc. ) .
  • an assay for measuring the level of a biomarker e.g., below the limit of detection of an assay measuring RNA transcript expression of the biomarker (such as RT-PCR, in situ hybridization, and/or next generation sequencing)
  • an assay measuring protein expression of a biomarker such as an immunoassay, PET imaging, Western blotting,
  • a subject is administered an effective amount of an anti-CD137 antibody when the level of a biomarker in a sample obtained from the subject is lower than the reference level. In some embodiments, a subject is determined to be likely to respond to an anti-CD137 antibody when the level of the biomarker in a sample obtained from the subject is lower than the reference level. In some embodiments, a subject is administered an effective amount of an anti-CD137 antibody after the subject has been determined to be likely to respond to the anti-CD137 antibody. In some embodiments, a subject having cancer is selected for treatment with an anti-CD137 antibody when the level of the biomarker in a sample obtained from the subject is lower than the reference level.
  • a subject is positively stratified for enrollment into an anti-CD137 antibody therapy when the level of a biomarker in a sample obtained from the subject is lower than the reference level.
  • the anti-CD137 antibody is administered to the subject in combination with an anti-PD-1 antibody.
  • the level of a biomarker measured in the sample obtained from the subject is considered to be higher than the reference level when the level of the biomarker in the sample is at least about 5%higher than the reference level.
  • the level of a biomarker measured in the sample obtained from the subject is considered to be higher than the reference level when the level of the biomarker in the sample is at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99%higher than the reference level.
  • the level of a biomarker measured in the sample obtained from the subject is considered to be higher than the reference level when the level of the biomarker in the sample is at least about 1-fold higher than the reference level.
  • the level of a biomarker measured in the sample obtained from the subject is considered to be higher than the reference level when the level of the biomarker in the sample is at least about 1-fold, at least about 1.5-fold, at least about 2-fold, at least about 2.5-fold, at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 4.5-fold, at least about 5-fold, at least about 5.5-fold, at least about 6-fold, at least about 6.5-fold, at least about 7-fold, at least about 7.5 fold, at least about 8-fold, at least about 8.5-fold, at least about 9-fold, at least about 9.5-fold, at least about 10-fold, at least about 100-fold, or at least about 1000-fold higher than the reference level.
  • the level of a biomarker in the reference sample is below the limit of detection. In some embodiments, the level of a biomarker measured in the sample obtained from the subject is considered to be higher than the reference level when the level of the biomarker in the sample is above the limit of detection, is detectable, and/or is not zero while the level of the biomarker in the reference sample is below the limit of detection.
  • a level is considered to be below the limit of detection when the level does not give an appreciable signal, a detectable signal, and/or is not significantly different than an appropriate negative control when performing an assay for measuring the level of a biomarker (e.g., below the limit of detection of an assay measuring RNA transcript expression of the biomarker (such as RT-PCR, in situ hybridization, and/or next generation sequencing) , below the limit of detection of an assay measuring protein expression of the biomarker (such as an immunoassay, PET imaging, Western blotting, ELISA, immunohistochemistry, and/or flow cytometry) , etc. ) .
  • an assay for measuring the level of a biomarker e.g., below the limit of detection of an assay measuring RNA transcript expression of the biomarker (such as RT-PCR, in situ hybridization, and/or next generation sequencing)
  • an assay measuring protein expression of the biomarker such as an immunoassay, PET imaging, Western blotting, EL
  • a subject is administered an effective amount of an anti-CD137 antibody when the level of a biomarker in a sample obtained from the subject is higher than the reference level. In some embodiments, a subject is determined to be likely to respond to an anti-CD137 antibody when the level of a biomarker in a sample obtained from the subject is higher than the reference level. In some embodiments, a subject is administered an effective amount of an anti-CD137 antibody after the subject has been determined to be likely to respond to the anti-CD137 antibody. In some embodiments, a subject having cancer is selected for treatment with an anti-CD137 antibody when the level of expression of a biomarker in a sample obtained from the subject is higher than the reference level.
  • a subject is positively stratified for enrollment into an anti-CD137 antibody therapy when the level of a biomarker in a sample obtained from the subject is higher than the reference level.
  • the anti-CD137 antibody is administered to the subject in combination with an anti-PD-1 antibody.
  • the method comprises determining the level of a biomarker at two or more time points during the course of the anti-CD137 antibody treatment. In some embodiments, the method comprises determining the level of a biomarker (e.g., sCD137, CD137L, NK cell level) in a sample obtained from the subject prior to the administration of the anti-CD137 antibody. In some embodiments, the method comprises determining the level of a biomarker in a sample obtained from the subject after the administration of the anti-CD137 antibody. In some embodiments, the method comprises determining the level of a biomarker in a sample obtained from the subject after each cycle of anti-CD137 antibody treatment.
  • a biomarker e.g., sCD137, CD137L, NK cell level
  • the method comprises determining a ratio ( “induction ratio” ) between the difference in the level of the biomarker after administration of the anti-CD137 antibody (C2) and the level of the biomarker before administration of the anti-CD137 antibody (C1) and the level of the biomarker before administration of the anti-CD137 antibody (C1) : (C2-C1) /C1.
  • an induction ratio of at least about any one of 50%, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more indicates a high likelihood of responding to the anti-CD137 antibody treatment, i.e., having stable disease (SD) or partial response (PR) after the treatment.
  • the method described herein comprise administration of an anti-CD137 antibody that specifically binds to an extracellular domain of human CD137.
  • the anti-CD137 antibodies described herein include full-length anti-CD137 antibodies, antigen-binding fragments of the CD137 antibodies, and derivatives of the CD137 antibodies.
  • the anti-CD137 antibody is any one of the antibodies described herein, including antibodies described with reference to epitope binding and antibodies described with reference to specific amino acid sequences of CDRs, variable regions (VL, VH) , and IgG (e.g., IgG4) light and heavy chains.
  • the anti-CD137 antibody has at least one (e.g., at least one, at least two, at least three, at least four, at least five, at least six, at least seven, eight, or all nine) of the following functional properties: (a) bind to human CD137 with a KD of 500 nM or less; (b) have agonist activity on human CD137; (c) do not bind to human OX40, CD40, GITR and/or CD27 receptor at concentration up to 1000 nM; (d) is cross-reactive with monkey, mouse, rat, or dog CD137; (e) do not induce ADCC effects; (f) are capable of inhibiting tumor cell growth; (g) have therapeutic effect on a cancer; (h) blocks binding between CD137 and CD137L; and (i) blocks CD137 signaling stimulated by CD137L (e.g., CD137L-stimulated NF- ⁇ B-dependent transcription) in a cell that expresses CD137.
  • CD137L e.g., CD137L-
  • the antibodies disclosed herein can also block, e.g., completely block, the binding between CD137 and its ligand CD137L.
  • the anti-CD137 antibody is an antibody (or an antigen-binding fragment thereof) that cross-competes for binding to human CD137 with one or more of the antibodies or antigen-binding fragments as described herein.
  • Exemplary anti-CD137 antibodies that are suitable for the methods described herein have been described, for example, in US20190055314A1, WO2019036855A1, and WO2019037711A1, which are incorporated herein by reference in their entirety.
  • Human CD137 is a 255 amino acid protein (e.g., GenBank Accession No. NM_001561; NP_001552; SEQ ID NO.: 1) .
  • the protein comprises a signal sequence (amino acid residues 1-17) , followed by an extracellular domain (169 amino acids) , a transmembrane region (27 amino acids) , and an intracellular domain (42 amino acids) (Cheuk ATC et al. 2004 Cancer Gene Therapy 11: 215-226) .
  • the receptor is expressed on the cell surface in monomer and dimer forms and likely trimerizes with CD137 ligand to signal.
  • the anti-CD137 antibody specifically binds to one or more amino acid residues within amino acid residues 34-108 of SEQ ID NO: 1. In some embodiments, the anti-CD137 antibody specifically binds to one or more amino acid residues within amino acid residues 34-93 of SEQ ID NO: 1. In some embodiments, the anti-CD137 antibody specifically binds to one or more amino acid residues selected from the group consisting of amino acid residues 34-36, 53-55, and 92-93 of SEQ ID NO: 1. In some embodiments, the anti-CD137 antibody specifically binds to one or more of amino acid residues 34-36, one or more of amino acid residues 53-55, and one or more or amino acid residues 92-93 of SEQ ID NO: 1.
  • the anti-CD137 antibody does not bind to one or more of amino acid residues selected from the group consisting of amino acid residues 109-112, 125, 126, 135-138, 150 and 151 of SEQ ID NO: 1. In some embodiments, the anti-CD137 antibody specifically does not bind to amino acid residues 109-112, 125, 126, 135-138, 150 and 151 of SEQ ID NO: 1.
  • Methods of measuring an antibody or antigen-binding fragment’s ability to bind a target antigen may be carried out using any method known in the art, including for example, by surface plasmon resonance, an ELISA, isothermal titration calorimetry, a filter binding assay, an EMSA, etc., or based on the crystal structure of the anti-CD137 antibody with CD137.
  • the anti-CD137 antibody specifically binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1. In some embodiments, the anti-CD137 antibody specifically binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 53, 63-67, 69-73, 83, 89, 92, 98-104, and 112-116 of SEQ ID NO: 1.
  • the anti-CD137 antibody specifically binds to one or more amino acid residues selected from the group consisting of amino acid residues 51, 63-67, 69-73, 83, 89, 92, 98-104 and 112-114 of SEQ ID NO: 1.
  • the anti-CD137 antibody specifically binds to amino acid residues 51, 53, 62-73, 83, 89, 92, 95-104 and 112-116 of SEQ ID NO: 1. In some embodiments, the anti-CD137 antibody specifically binds to amino acid residues 51, 53, 63-67, 69-73, 83, 89, 92, 98-104, and 112-116 of SEQ ID NO: 1. In some embodiments, the anti-CD137 antibody specifically binds to amino acid residues 51, 63-67, 69-73, 83, 89, 92, 98-104 and 112-114 of SEQ ID NO: 1.
  • the anti-CD137 antibody specifically binds to human CD137 with a KD of about 500 nM or less (e.g., about 500 nM or less, about 400 nM or less, about 300 nM or less, about 200 nM or less, about 150 nM or less, about 100 nM or less, about 90 nM or less, about 80 nM or less, about 75 nM or less, about 70 nM or less, about 60 nM or less, about 50 nM or less, about 40 nM or less, about 30 nM or less, about 25 nM or less, about 20 nM or less, about 10 nM or less, about 1 nM or less, about 0.1 nM or less, etc.
  • a KD of about 500 nM or less (e.g., about 500 nM or less, about 400 nM or less, about 300 nM or less, about 200 nM or less, about 150 nM or less, about 100 nM or
  • the anti-CD137 antibody specifically binds to human CD137 with a KD of about 100 nM or less. In some embodiments, the anti-CD137 antibody specifically binds to human CD137 with a KD of about 50 nM or less.
  • Methods of measuring the KD of an antibody or antigen-binding fragment may be carried out using any method known in the art, including for example, by surface plasmon resonance, an ELISA, isothermal titration calorimetry, a filter binding assay, an EMSA, etc.
  • Anti-CD137 antibodies need to be cross-linked to become agonistic. For example, cross-linking is achieved in vivo through Fcgamma receptors, while typically polyclonal anti-Fc antibodies are used in cell-based experiments in vitro.
  • the anti-CD137 antibodies described herein have agonist activity on human CD137.
  • the anti-CD137 antibody induces one or more (e.g., one or more, two or more, three or more, etc. ) activities of human CD137 when a cell (e.g., a human cell) expressing human CD137 is contacted by the anti-CD137 antibody.
  • CD137 activities are known in the art and may include, without limitation, induction of NF- ⁇ B-dependent transcription, induction of T cell proliferation, prolonging T cell survival, co- stimulation of activated T cells, induction of cytokine secretion (such as IL-2) , and induction of monocyte activation .
  • the one or more CD137 activities is not CD137 binding to its ligand.
  • Methods of measuring CD137 activity e.g., the induction of NF- ⁇ B-dependent transcription and/or T cell proliferation, etc.
  • the anti-CD137 antibody increases NF- ⁇ B dependent transcription in cells (e.g., human cells) expressing human CD137.
  • NF- ⁇ B dependent transcription is increased by about 10%or more, about 20%or more, about 30%or more, about 40%or more, about 50%or more, about 60%or more, about 70%or more, about 80%or more, about 90%or more, or about 99%or more in cells (e.g., human cells) expressing CD137 contacted with the anti-CD137 antibody, relative to a corresponding cell not contacted with the anti-CD137 antibody (e.g., a corresponding cell not contacted with an antibody, or contacted with an isotype control antibody) .
  • cells e.g., human cells
  • a corresponding cell not contacted with the anti-CD137 antibody e.g., a corresponding cell not contacted with an antibody, or contacted with an isotype control antibody
  • NF- ⁇ B dependent transcription is increased by about 2-fold, 3-fold, 4-folr, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 100-fold, 1000-fold or more in cells (e.g., human cells) expressing CD137 contacted with the anti-CD137 antibody, relative to a corresponding cell not contacted with the anti-CD137 antibody (e.g., a corresponding cell not contacted with an antibody, or contacted with an isotype control antibody) .
  • cells e.g., human cells
  • a corresponding cell not contacted with the anti-CD137 antibody e.g., a corresponding cell not contacted with an antibody, or contacted with an isotype control antibody
  • the anti-CD137 antibody is cross-reactive with monkey (e.g., cynomolgus monkey) , mouse, rat, and/or dog CD137. In some embodiments, the anti-CD137 antibody is cross-reactive with monkey CD137. In some embodiments, the anti-CD137 antibody is cross-reactive with mouse CD137. In some embodiments, the anti-CD137 antibody is cross-reactive with rat CD137. In some embodiments, the anti-CD137 antibody is cross-reactive with dog CD137.
  • monkey e.g., cynomolgus monkey
  • mouse CD137 mouse CD137
  • the anti-CD137 antibody is cross-reactive with rat CD137.
  • the anti-CD137 antibody is cross-reactive with dog CD137.
  • the anti-CD137 antibody is cross-reactive with monkey and mouse CD137; monkey and rat CD137; monkey and dog CD137; mouse and rat CD137; mouse and dog CD137; rat and dog CD137; monkey, mouse, and rat CD137; monkey, mouse, and dog CD137; monkey, rat, and dog CD137; mouse, rat, and dog CD137; or monkey, mouse, rat, and dog CD137.
  • the anti-CD137 antibody is cross-reactive at about 100 nM (e.g., at about 1nM, at about 10nM, at about 25nM, at about 50nM, at about 75nM, at about 100nM) .
  • Methods of measuring antibody cross-reactivity are known in the art, including, without limitation, surface plasmon resonance, an ELISA, isothermal titration calorimetry, a filter binding assay, an EMSA, etc.
  • the anti-CD137 antibody does not induce ADCC effects.
  • Methods of measuring ADCC effects e.g., in vivo methods are known in the art.
  • the anti-CD137 antibody does not ADCC effects by more than about 10%(do not induce ADCC by more than about 10%, more than about 5%, more than about 1%, more than about 0.1%, more than about 0.01%) relative to a control.
  • the anti-CD137 antibody is capable of inhibiting tumor cell growth/proliferation.
  • the tumor cell growth/proliferation is inhibited by at least about 5% (e.g., at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 99%) when contacted with the anti-CD137 antibody relative to corresponding tumor cells not contacted with the anti-CD137 antibody.
  • the anti-CD137 antibody is capable of reducing tumor volume in a subject when the subject is administered the anti-CD137 antibody.
  • the anti-CD137 antibody is capable of reducing tumor volume in a subject by at least about 5% (e.g., at least about 5%, at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 99%) relative to the initial tumor volume in the subject (e.g., prior to administration of the anti-CD137 antibody) .
  • Methods of monitoring tumor cell growth/proliferation, tumor volume, and/or tumor inhibition are known in the art.
  • the anti-CD137 antibody has therapeutic effect on a cancer. In some embodiments, the anti-CD137 antibody reduces one or more signs or symptoms of a cancer. In some embodiments, a subject suffering from a cancer goes into partial or complete remission when administered the anti-CD137 antibody.
  • the anti-CD137 antibody is selected from the group consisting of AG10058, AG10059 and ADG106. In some embodiments, the anti-CD137 antibody competes or cross-competes for binding to human CD137 with any of the illustrative antibodies of the present application, such as AG10058, AG10059 and ADG106. In some embodiments, the anti-CD137 antibody is an antibody that competes or cross-competes for binding to the same epitope on human CD137 as AG10058, AG10059 or ADG106. The ability of an antibody to compete or cross-compete for binding with another antibody can be determined using standard binding assays known in the art, such as BIAcore analysis, ELISA assays, or flow cytometry.
  • an illustrative antibody of the disclosure can bind to human CD137 under saturating conditions and then measure the ability of the test antibody to bind to the CD137. If the test antibody is able to bind to the CD137 at the same time as the illustrative antibody, then the test antibody binds to a different epitope as the illustrative antibody. However, if the test antibody is not able to bind to the CD137 at the same time, then the test antibody binds to the same epitope, an overlapping epitope, or an epitope that is in close proximity to the epitope bound by the illustrative antibody. This experiment can be performed using various methods, such as ELISA, RIA, FACS or surface plasmon resonance.
  • the anti-CD137 antibody blocks the binding between CD137 and its ligand (e.g., human CD137 and human CD137L) . In some embodiments, the anti-CD137 antibody blocks the binding between CD137 and its ligand in vitro. In some embodiments, the anti-CD137 antibody has a half maximal inhibitory concentration (IC50) of about 500 nM or less (e.g., about 500 nM or less, about 400nM or less, about 300nM or less, about 200nM or less, about 100nM or less, about 50nM or less, about 25nM or less, about 10nM or less, about 1nM or less, etc. ) for blocking binding of CD137 its ligand.
  • IC50 half maximal inhibitory concentration
  • the anti-CD137 antibody has a half-maximal inhibitory concentration (IC50) of about 100 nM or less for blocking binding of CD137 its ligand. In some embodiments, the anti-CD137 antibody completely blocks binding of human CD137 to its ligand when provided at a concentration of about 100 nM or greater (e.g., about 100nM or greater, about 500nM or greater, about 1 ⁇ M or greater, about 10 ⁇ M or greater, etc. ) .
  • the term “complete blocking” or “completely blocks” refers to the antibody or antigen-binding fragment’s ability to reduce binding between a first protein and a second protein by at least about 80% (e.g., at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 99%, etc. ) .
  • Methods of measuring the ability of an antibody or antigen-binding fragment to block binding of a first protein (e.g., a CD137) and a second protein (e.g., CD137L) are known in the art, including, without limitation, via BIAcore analysis, ELISA assays, and flow cytometry.
  • the anti-CD137 antibody comprises a heavy chain variable region (VH) and a light chain variable region (VL) , a) wherein the VH comprises an HVR-H1, an HVR-H2, and an HVR-H3, wherein the HVR-H1 comprises an amino acid sequence according to a formula selected from the group consisting of: Formula (I) : X1TFX2X3YX4IHWV (SEQ ID NO: 32) , wherein X1 is F or Y, X2 is S or T, X3 is G, N, or S, and X4 is A, G, or W; Formula (II) : YSIX1SGX2X3WX4WI (SEQ ID NO: 33) , wherein X1 is S or T, X2 is H or Y, X3 is H or Y, and X4 is A, D, G, N, S, or T; and Formula (III) : FSLSTX1GVX2
  • the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 34, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 35, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 38; and/or wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 39, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 40, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 41.
  • Table B Exemplary anti-CD137 antibodies.
  • the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 2, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 3, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 4; and/or wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 5, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 6, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 7.
  • the anti-CD137 antibody comprises a VH comprising a heavy chain complementarity determining region (HC-CDR) 1, a HC-CDR2, and a HC-CDR3 of the amino acid sequence of SEQ ID NO: 8; and/or a VL comprising a light chain complementarity determining region (LC-CDR) 1, a LC-CDR2, and a LC-CDR3 of the amino acid sequence of SEQ ID NO: 9.
  • the anti-CD137 antibody comprises a heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 8, and/or a light chain variable region comprising the amino acid sequence of SEQ ID NO: 9.
  • the anti-CD137 antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 10, and/or a light chain comprising the amino acid sequence of SEQ ID NO: 11.
  • the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14; and/or wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 15, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 17.
  • the anti-CD137 antibody comprises a VH comprising a HC-CDR1, a HC-CDR2, and a HC-CDR3 of the amino acid sequence of SEQ ID NO: 18; and/or a VL comprising a LC-CDR1, a LC-CDR2, and a LC-CDR3 of the amino acid sequence of SEQ ID NO: 19.
  • the anti-CD137 antibody comprises a heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 18, and/or a light chain variable region comprising the amino acid sequence of SEQ ID NO: 19.
  • the anti-CD137 antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 20, and/or a light chain comprising the amino acid sequence of SEQ ID NO: 21.
  • the anti-CD137 antibody comprises a VH and a VL, wherein the VH comprises a HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22, a HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, and a HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24; and/or wherein the VL comprises a HVR-L1 comprising the amino acid sequence of SEQ ID NO: 25, a HVR-L2 comprising the amino acid sequence of SEQ ID NO: 26, and a HVR-L3 comprising the amino acid sequence of SEQ ID NO: 27.
  • the anti-CD137 antibody comprises a VH comprising a HC-CDR1, a HC-CDR2, and a HC-CDR3 of the amino acid sequence of SEQ ID NO: 28; and/or a VL comprising a LC-CDR1, a LC-CDR2, and a LC-CDR3 of the amino acid sequence of SEQ ID NO: 29.
  • the anti-CD137 antibody comprises heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 28, and/or a light chain variable region comprising the amino acid sequence of SEQ ID NO: 29.
  • the anti-CD137 antibody comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 30, and/or a light chain comprising the amino acid sequence of SEQ ID NO: 31.
  • the CD137 antibodies described herein can be in any class, such as IgG, IgM, IgE, IgA, or IgD. It is preferred that the CD137 antibodies are in the IgG class, such as IgG1, IgG2, IgG3, or IgG4 subclass.
  • a CD137 antibody can be converted from one class or subclass to another class or subclass using methods known in the art.
  • An exemplary method for producing an antibody in a desired class or subclass comprises the steps of isolating a nucleic acid encoding a heavy chain of an CD137 antibody and a nucleic acid encoding a light chain of a CD137 antibody, isolating the sequence encoding the VH region, ligating the VH sequence to a sequence encoding a heavy chain constant region of the desired class or subclass, expressing the light chain gene and the heavy chain construct in a cell, and collecting the CD137 antibody.
  • the anti-CD137 antibody comprises a human IgG4 Fc region.
  • the human IgG4 Fc region comprises an S241P mutation, wherein numbering is according to Kabat.
  • the anti-CD137 antibody is an antigen-binding fragment of any one of the anti-CD137 antibodies described herein.
  • the antigen-binding fragments of an CD137 antibody include: (i) a Fab fragment, which is a monovalent fragment consisting of the V L , V H , C L and C H 1 domains; (ii) a F (ab′) 2 fragment, which is a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the V H and C H 1 domains; (iv) a Fv fragment consisting of the V L and V H domains of a single arm of an antibody; (v) a dAb fragment (Ward et al., (1989) Nature 341: 544-546) , which consists of a V H domain; (vi) an isolated CDR, and (vii) single chain antibody (scFv) , which is a polypeptide comprising a V L region of an antibody linked to a V H region of an antibody. Bird et al., (1988) Science
  • the anti-CD137 antibody is a derivative of any one of the anti-CD137 antibodies described herein.
  • the antibody derivative is derived from modifications of the amino acid sequences of an illustrative antibody ( “parent antibody” ) of the disclosure while conserving the overall molecular structure of the parent antibody amino acid sequence.
  • Amino acid sequences of any regions of the parent antibody chains may be modified, such as framework regions, CDR regions, or constant regions. Types of modifications include substitutions, insertions, deletions, or combinations thereof, of one or more amino acids of the parent antibody.
  • the antibody derivative comprises a VH comprising an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to the amino acid sequence of SEQ ID NO: 8; and/or a VL comprising an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to the amino acid sequence of SEQ ID NO: 9.
  • the antibody derivative comprises a HVR-H1 amino acid sequence region that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 2.
  • the antibody derivative comprises a HVR-H2 amino acid sequence region that is at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 3.
  • the antibody derivative comprises a HVR-H3 amino acid sequence region that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 4.
  • the antibody derivative comprises a HVR-L1 amino acid sequence region that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 5.
  • the antibody derivative comprises a HVR-L2 amino acid sequence region that is at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 6.
  • the antibody derivative comprises a HVR-L3 amino acid sequence region that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 7.
  • the antibody derivative comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 conservative or non-conservative substitutions, and/or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 additions and/or deletions to an amino acid sequence as set forth in any of SEQ ID NOs: 8, 9, 10, and 11.
  • the antibody derivative comprises a VH comprising an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to the amino acid sequence of SEQ ID NO: 18; and/or a VL comprising an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to the amino acid sequence of SEQ ID NO: 19.
  • the antibody derivative comprises a HVR-H1 amino acid sequence region that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 12.
  • the antibody derivative comprises a HVR-H2 amino acid sequence region that is at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 13.
  • the antibody derivative comprises a HVR-H3 amino acid sequence region that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 14.
  • the antibody derivative comprises a HVR-L1 amino acid sequence region that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 15.
  • the antibody derivative comprises a HVR-L2 amino acid sequence region that is at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 16.
  • the antibody derivative comprises a HVR-L3 amino acid sequence region that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 17.
  • the antibody derivative comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 conservative or non-conservative substitutions, and/or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 additions and/or deletions to an amino acid sequence as set forth in any of SEQ ID NOs: 18, 19, 20, and 21.
  • the antibody derivative comprises a VH comprising an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to the amino acid sequence of SEQ ID NO: 28; and/or a VL comprising an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to the amino acid sequence of SEQ ID NO: 29.
  • the antibody derivative comprises a HVR-H1 amino acid sequence region that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 22.
  • the antibody derivative comprises a HVR-H2 amino acid sequence region that is at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 23.
  • the antibody derivative comprises a HVR-H3 amino acid sequence region that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 24.
  • the antibody derivative comprises a HVR-L1 amino acid sequence region that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 25.
  • the antibody derivative comprises a HVR-L2 amino acid sequence region that is at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 26.
  • the antibody derivative comprises a HVR-L3 amino acid sequence region that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%identical to an amino acid sequence as set forth in SEQ ID NO: 27.
  • the antibody derivative comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 conservative or non-conservative substitutions, and/or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 additions and/or deletions to an amino acid sequence as set forth in any of SEQ ID NOs: 28, 29, 30, and 31.
  • Amino acid substitutions encompass both conservative substitutions and non-conservative substitutions.
  • conservative amino acid substitution means a replacement of one amino acid with another amino acid where the two amino acids have similarity in certain physico-chemical properties such as polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved.
  • substitutions typically may be made within each of the following groups: (a) nonpolar (hydrophobic) amino acids, such as alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; (b) polar neutral amino acids, such as glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine; (c) positively charged (basic) amino acids, such as arginine, lysine, and histidine; and (d) negatively charged (acidic) amino acids, such as aspartic acid and glutamic acid.
  • nonpolar amino acids such as alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine
  • polar neutral amino acids such as glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine
  • the present disclosure provides an antibody derivative that contains the VH and VL CDR sequences of an illustrative antibody of this disclosure, yet contains framework sequences different from those of the illustrative antibody.
  • 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 Genbank database or in the “VBase” human germline sequence database (Kabat, E.A., et al., Sequences of Proteins of Immunological Interest, Fifth Edition, U.S.
  • Framework sequences that may be used in constructing an antibody derivative include those that are structurally similar to the framework sequences used by illustrative antibodies of the disclosure, e.g., similar to the VH 3-23 framework sequences and/or the VL ⁇ 3 or ⁇ 1-13 framework sequences used by illustrative antibodies of the disclosure.
  • the HVR-H1, HVR-H2, and HVR-H3 sequences, and the HVR-L1, HVR-L2, and HVR-L3 sequences of an illustrative antibody 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.
  • the antibody derivative is a chimeric antibody, which comprises an amino acid sequence of an illustrative antibody of the disclosure.
  • one or more CDRs from one or more illustrative human antibodies are combined with CDRs from an antibody from a non-human animal, such as mouse or rat.
  • all of the CDRs of the chimeric antibody are derived from one or more illustrative antibodies.
  • the chimeric antibody comprises one, two, or three CDRs from the heavy chain variable region or from the light chain variable region of an illustrative antibody. Chimeric antibodies can be generated using conventional methods known in the art.
  • Another type of modification is to mutate amino acid residues within the CDR regions of the VH and/or VL chain.
  • 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 known in the art. Typically, conservative substitutions are introduced.
  • the mutations may be amino acid additions and/or deletions. Moreover, typically no more than one, two, three, four or five residues within a CDR region are altered.
  • the antibody derivative comprises 1, 2, 3, or 4 amino acid substitutions in the heavy chain CDRs and/or in the light chain CDRs.
  • the amino acid substitution is to change one or more cysteines in an antibody to another residue, such as, without limitation, alanine or serine.
  • the cysteine may be a canonical or non-canonical cysteine.
  • the antibody derivative has 1, 2, 3, or 4 conservative amino acid substitutions in the heavy chain CDR regions relative to the amino acid sequences of an illustrative antibody.
  • Modifications may also be made to the framework residues within the VH and/or VL regions. Typically, such framework variants are made to decrease the immunogenicity of the antibody.
  • One approach is to “back mutate” 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 “back mutated” to the germline sequence by, for example, site-directed mutagenesis or PCR-mediated mutagenesis.
  • modifications may also be made within the Fc region of an illustrative antibody, 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.
  • the hinge region of CH1 is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased. This approach is described further in U.S. Pat. No. 5,677,425.
  • 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.
  • the Fc hinge region of an antibody is mutated to decrease the biological half-life of the antibody.
  • an antibody of the disclosure may be modified to alter its potential glycosylation site or pattern in accordance with routine experimentation known in the art.
  • the anti-CD137 antibody derivative contains at least one mutation in a variable region of a light chain or heavy chain that changes the pattern of glycosylation in the variable region.
  • Such an antibody derivative may have an increased affinity and/or a modified specificity for binding an antigen.
  • the mutations may add a novel glycosylation site in the V region, change the location of one or more V region glycosylation site (s) , or remove a pre-existing V region glycosylation site.
  • the anti-CD137 antibody derivative has a potential N-linked glycosylation site at asparagine in the heavy chain variable region, wherein the potential N-linked glycosylation site in one heavy chain variable region is removed. In some embodiments, the anti-CD137 antibody derivative has having a potential N-linked glycosylation site at asparagine in the heavy chain variable region, wherein the potential N-linked glycosylation site in both heavy chain variable regions is removed.
  • the antibody derivative is a CD137 antibody multimer, which is a multimeric form of a CD137 antibody, such as antibody dimers, trimers, or higher-order multimers of monomeric antibodies.
  • a CD137 antibody multimer is a multimeric form of a CD137 antibody, such as antibody dimers, trimers, or higher-order multimers of monomeric antibodies.
  • Individual monomers within an antibody multimer may be identical or different.
  • antibody homodimers may be formed through chemical linkage techniques known in the art, such as through using crosslinking agents.
  • Suitable crosslinkers include those that are heterobifunctional, having two distinctly reactive groups separated by an appropriate spacer (such as m-maleimidobenzoyl-N-hydroxysuccinimide ester, succinimidyl 4- (maleimidomethyl) cyclohexane-1-carboxylate, and N-succinimidyl S-acethylthio-acetate) or homobifunctional (such as disuccinimidyl suberate) .
  • an appropriate spacer such as m-maleimidobenzoyl-N-hydroxysuccinimide ester, succinimidyl 4- (maleimidomethyl) cyclohexane-1-carboxylate, and N-succinimidyl S-acethylthio-acetate
  • homobifunctional such as disuccinimidyl suberate
  • the anti-CD137 antibody is a multimeric antibody (e.g., a bispecific antibody) .
  • the anti-CD137 antibody is an IgM antibody, e.g., comprises an IgM Fc region (e.g., a human IgM Fc region) .
  • a “single-chain antibody” consists of a single polypeptide chain comprising a VL domain linked to a VH domain wherein VL domain and VH domain are paired to form a monovalent molecule.
  • Single chain antibody can be prepared according to method known in the art (see, for example, Bird et al., (1988) Science 242: 423-426 and Huston et al., (1988) Proc. Natl. Acad. Sci. USA 85: 5879-5883) .
  • a “diabody” consists of two chains, each chain comprising a heavy chain variable region connected to a light chain variable region on the same polypeptide chain connected by a short peptide linker, wherein the two regions on the same chain do not pair with each other but with complementary domains on the other chain to form a bispecific molecule.
  • Methods of preparing diabodies are known in the art (See, e.g., Holliger P. et al., (1993) Proc. Natl. Acad. Sci. USA 90: 6444-6448, and Poljak R.J. et al., (1994) Structure 2: 1121-1123) .
  • Domain antibodies are small functional binding units of antibodies, corresponding to the variable regions of either the heavy or light chains of antibodies. Domain antibodies are well expressed in bacterial, yeast, and mammalian cell systems. Further details of domain antibodies and methods of production thereof are known in the art (see, for example, U.S. Pat. Nos. 6,291,158; 6,582,915; 6,593,081; 6,172,197; 6,696,245; European Patents 0368684 &0616640; WO05/035572, WO04/101790, WO04/081026, WO04/058821, WO04/003019 and WO03/002609) . Unibodies consist of one light chain and one heavy chain of an IgG4 antibody. Unibodies may be made by the removal of the hinge region of IgG4 antibodies. Further details of unibodies and methods of preparing them may be found in WO2007/059782.
  • Antibodies of the present disclosure can be produced by techniques known in the art, including conventional monoclonal antibody methodology e.g., the standard somatic cell hybridization technique (See e.g., Kohler and Milstein, Nature 256: 495 (1975) , viral or oncogenic transformation of B lymphocytes, or recombinant antibody technologies as described in detail herein below.
  • conventional monoclonal antibody methodology e.g., the standard somatic cell hybridization technique (See e.g., Kohler and Milstein, Nature 256: 495 (1975) , viral or oncogenic transformation of B lymphocytes, or recombinant antibody technologies as described in detail herein below.
  • Hybridoma production is a very well established procedure.
  • the common animal system for preparing hybridomas is the murine system. Immunization protocols and techniques for isolation of immunized splenocytes for fusion are known in the art. Fusion partners (e.g., murine myeloma cells) and fusion procedures are also known.
  • One well-known method that may be used for making human CD137 antibodies provided by the present disclosure involves the use of a XENOMOUSE TM animal system.
  • XENOMOUSE TM mice are engineered mouse strains that comprise large fragments of human immunoglobulin heavy chain and light chain loci and are deficient in mouse antibody production.
  • the animal is immunized with a CD137 antigen.
  • the CD137 antigen is isolated and/or purified CD137, preferably CD137. It may be a fragment of CD137, such as the extracellular domain of CD137, particularly a CD137 extracellular domain fragment comprising amino acid resides 34-108 or 34-93 of SEQ ID NO: 1. Immunization of animals can be carried out by any method known in the art. See, e.g., Harlow and Lane, Antibodies: A Laboratory Manual, New York: Cold Spring Harbor Press, 1990.
  • the CD137 antigen may be administered with an adjuvant to stimulate the immune response.
  • adjuvants include complete or incomplete Freund's adjuvant, RIBI (muramyl dipeptides) or ISCOM (immunostimulating complexes) .
  • lymph node and/or splenic B cells are immortalized.
  • Methods of immortalizing cells include, but are not limited to, transferring them with oncogenes, inflecting them with the oncogenic virus cultivating them under conditions that select for immortalized cells, subjecting them to carcinogenic or mutating compounds, fusing them with an immortalized cell, e.g., a myeloma cell, and inactivating a tumor suppressor gene. See, e.g., Harlow and Lane, supra. If fusion with myeloma cells is used, the myeloma cells preferably do not secrete immunoglobulin polypeptides (a non-secretory cell line) .
  • Immortalized cells are screened using CD137, a portion thereof, or a cell expressing CD137.
  • CD137 antibody-producing cells e.g., hybridomas
  • Hybridomas can be expanded in vivo in syngeneic animals, in animals that lack an immune system, e.g., nude mice, or in cell culture in vitro. Methods of selecting, cloning and expanding hybridomas are well known to those of ordinary skill in the art.
  • Antibodies of the disclosure can also be prepared using phage display or yeast display methods.
  • display methods for isolating human antibodies are established in the art, such as Achim Knappik, et al., “Fully Synthetic Human Combinatorial Antibody Libraries (HuCAL) Based on Modular Consensus Frameworks and CDRs Randomized with Trinucleotides. ” J. Mol. Biol. (2000) 296, 57-86; and Michael J. Feldhaus, et al., “Flow-cytometric isolation of human antibodies from a non-immune Saccharomyces cerevisiae surface display library” Nat Biotechnol (2003) 21: 163-170.
  • the anti-CD137 antibody is prepared by expressing one or more nucleic acids encoding the anti-CD137 antibody or polypeptide chains thereof in a host cell.
  • the one or more nucleic acids is a DNA or RNA, and may or may not contain intronic sequences.
  • the nucleic acid is a cDNA molecule.
  • Nucleic acids of the disclosure can be obtained using any suitable molecular biology techniques.
  • cDNAs encoding the light and heavy chains of the antibody made by the hybridoma can be obtained by PCR amplification or cDNA cloning techniques.
  • antibodies obtained from an immunoglobulin gene library e.g., using phage display techniques
  • the nucleic acid encoding the antibody can be recovered from the library.
  • the isolated DNA encoding the VH region can be converted to a full-length heavy chain gene by operatively linking the VH-encoding DNA to another DNA molecule encoding heavy chain constant regions (CH1, CH2 and CH3) .
  • heavy chain constant regions CH1, CH2 and CH3 .
  • the sequences of human heavy chain constant region genes are known in the art (see e.g., Kabat et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242) and DNA fragments encompassing these regions can be obtained by standard PCR amplification.
  • the heavy chain constant region can be an IgG1, IgG2, IgG3, IgG4, IgA, IgE, IgM or IgD constant region, but most preferably is an IgG4 or IgG2 constant region without ADCC effect.
  • the IgG4 constant region sequence can be any of the various alleles or allotypes known to occur among different individuals. These allotypes represent naturally occurring amino acid substitution in the IgG4 constant regions.
  • the VH-encoding DNA can be operatively linked to another DNA molecule encoding only the heavy chain CH1 constant region.
  • the isolated DNA encoding the VL region can be converted to a full-length light chain gene (as well as a Fab light chain gene) by operatively linking the VL-encoding DNA to another DNA molecule encoding the light chain constant region, CL.
  • the sequences of human light chain constant region genes are known in the art (see e.g., Kabat et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242) and DNA fragments encompassing these regions can be obtained by standard PCR amplification.
  • the light chain constant region can be a kappa or lambda constant region.
  • the VH- and VL-encoding DNA fragments are operatively linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (Gly 4 -Ser) 3 , such that the VH and VL sequences can be expressed as a contiguous single-chain protein, with the VL and VH regions joined by the flexible linker (see e.g., Bird et al., Science 242: 423-426 (1988) ; Huston et al., Proc. Natl. Acad. Sci. USA 85: 5879-5883 (1988) ; and McCafferty et al., Nature 348: 552-554 (1990) ) .
  • a flexible linker e.g., encoding the amino acid sequence (Gly 4 -Ser) 3 , such that the VH and VL sequences can be expressed as a contiguous single-chain protein, with the VL and VH regions joined by the flexible linker (see
  • a vector that comprises one or more nucleic acid molecules encoding an anti-CD137 antibody described herein.
  • the vector is an expression vector useful for the expression of the anti-CD137 antibody.
  • a first vector comprises a polynucleotide sequence encoding a heavy chain variable region as described herein
  • a second vector comprises a polynucleotide sequence encoding a light chain variable region as described herein.
  • a single vector comprises polynucleotides encoding a heavy chain variable region as described herein and a light chain variable region as described herein.
  • DNAs encoding partial or full-length light and heavy chains are inserted into expression vectors such that the DNA molecules are operatively linked to transcriptional and translational control sequences.
  • operatively linked means that an antibody gene is ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended function of regulating the transcription and translation of the DNA molecule.
  • the expression vector and expression control sequences are chosen to be compatible with the expression host cell used.
  • the antibody light chain gene and the antibody heavy chain gene can be inserted into separate vector or, more typically, both genes are inserted into the same expression vector.
  • the antibody genes are inserted into the expression vector by any suitable methods (e.g., ligation of complementary restriction sites on the antibody gene fragment and vector, or homologous recombination-based DNA ligation) .
  • the light and heavy chain variable regions of the antibodies described herein can be used to create full-length antibody genes of any antibody isotype and subclass by inserting them into expression vectors already encoding heavy chain constant and light chain constant regions of the desired isotype and subclass such that the VH segment is operatively linked to the CH segment (s) within the vector and the VL segment is operatively linked to the CL segment within the vector.
  • the recombinant expression vector can encode a signal peptide that facilitates secretion of the antibody chain from a host cell.
  • the antibody chain gene can be cloned into the vector such that the signal peptide is linked in-frame to the amino terminus of the antibody chain gene.
  • the signal peptide can be an immunoglobulin signal peptide or a heterologous signal peptide (i.e., a signal peptide from a non-immunoglobulin protein) .
  • the expression vectors of the disclosure typically carry regulatory sequences that control the expression of the antibody chain genes in a host cell.
  • regulatory sequence is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the antibody chain genes.
  • Such regulatory sequences are described, for example, in Goeddel (Gene Expression Technology. Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990) ) . It will be appreciated by those skilled in the art that the design of the expression vector, including the selection of regulatory sequences, may depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc.
  • regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV) , Simian Virus 40 (SV40) , adenovirus, (e.g., the adenovirus major late promoter (AdMLP) and polyoma.
  • CMV cytomegalovirus
  • SV40 Simian Virus 40
  • AdMLP adenovirus major late promoter
  • nonviral regulatory sequences may be used, such as the ubiquitin promoter or ⁇ -globin promoter.
  • regulatory elements composed of sequences from different sources such as the SR promoter system, which contains sequences from the SV40 early promoter and the long terminal repeat of human T cell leukemia virus type 1 (Takebe, Y. et al. (1988) Mol. Cell. Biol. 8: 466-472) .
  • the expression vectors may carry additional sequences, such as sequences that regulate replication of the vector in host cells (e.g., origins of replication) and selectable marker genes.
  • the selectable marker gene facilitates selection of host cells into which the vector has been introduced (see, e.g., U.S. Pat. Nos. 4,399,216, 4,634,665 and 5,179,017, all by Axel et al. ) .
  • the selectable marker gene confers resistance to drugs, such as G418, hygromycin or methotrexate, on a host cell into which the vector has been introduced.
  • Selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr-host cells with methotrexate selection/amplification) and the neo gene (for G418 selection) .
  • DHFR dihydrofolate reductase
  • neo gene for G418 selection
  • the expression vector (s) encoding the heavy and light chains is transfected into a host cell by any suitable techniques.
  • the various forms of the term “transfection” are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like.
  • electroporation e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like.
  • it is possible to express the antibodies of the disclosure in either prokaryotic or eukaryotic host cells expression of antibodies in eukaryotic cells, and typically mammalian host cells, is most typical.
  • a host cell containing a nucleic acid molecule provided by the present disclosure.
  • the host cell can be virtually any cell for which expression vectors are available. It may be, for example, a higher eukaryotic host cell, such as a mammalian cell, a lower eukaryotic host cell, such as a yeast cell, and may be a prokaryotic cell, such as a bacterial cell.
  • Introduction of the recombinant nucleic acid construct into the host cell can be effected by calcium phosphate transfection, DEAE, dextran mediated transfection, electroporation or phage infection.
  • Suitable prokaryotic hosts for transformation include E. coli, Bacillus subtilis, Salmonella typhimurium and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus.
  • Mammalian host cells for expressing a binding molecule of the disclosure include, for example, Chinese Hamster Ovary (CHO) cells (including dhfr-CHO cells, described in Urlaub and Chasin, Proc. Natl. Acad. Sci. USA 77: 4216-4220 (1980) , used with a DHFR selectable marker, e.g., as described in Kaufman and Sharp, J. Mol. Biol. 159: 601-621 (1982) , NS0 myeloma cells, COS cells and Sp2 cells.
  • CHO Chinese Hamster Ovary
  • GS glucose synthetase gene expression system
  • WO 87/04462 WO 89/01036
  • EP 338, 841 GS gene expression system
  • the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or secretion of the antibody into the culture medium in which the host cells are grown.
  • Antibodies can be recovered from the culture medium using any suitable protein purification methods.
  • compositions comprising any one of the anti-CD137 antibodies described herein.
  • the composition is a pharmaceutical composition comprising the anti-CD137 antibody and a pharmaceutically acceptable carrier.
  • the compositions can be prepared by conventional methods known in the art.
  • pharmaceutically acceptable carrier refers to any inactive substance that is suitable for use in a formulation for the delivery of an active agent (e.g., the anti-CD137 antibody) .
  • a carrier may be an antiadherent, binder, coating, disintegrant, filler or diluent, preservative (such as antioxidant, antibacterial, or antifungal agent) , sweetener, absorption delaying agent, wetting agent, emulsifying agent, buffer, and the like.
  • Suitable pharmaceutically acceptable carriers include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like) dextrose, vegetable oils (such as olive oil) , saline, buffer, buffered saline, and isotonic agents such as sugars, polyalcohols, sorbitol, and sodium chloride.
  • compositions may be in any suitable forms, such as liquid, semi-solid, and solid dosage forms.
  • liquid dosage forms include solution (e.g., injectable and infusible solutions) , microemulsion, liposome, dispersion, or suspension.
  • solid dosage forms include tablet, pill, capsule, microcapsule, and powder.
  • a particular form of the composition suitable for delivering an anti-CD137 antibody is a sterile liquid, such as a solution, suspension, or dispersion, for injection or infusion.
  • Sterile solutions can be prepared by incorporating the antibody in the required amount in an appropriate carrier, followed by sterilization microfiltration.
  • dispersions are prepared by incorporating the antibody into a sterile vehicle that contains a basic dispersion medium and other carriers.
  • compositions for the preparation of sterile liquid, methods of preparation include vacuum drying and freeze-drying (lyophilization) to yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • methods of preparation include vacuum drying and freeze-drying (lyophilization) to yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the various dosage forms of the compositions can be prepared by conventional techniques known in the art.
  • the relative amount of an anti-CD137 antibody included in the composition will vary depending upon a number of factors, such as the specific anti-CD137 antibody and carriers used, dosage form, and desired release and pharmacodynamic characteristics.
  • the amount of an anti-CD137 antibody in a single dosage form will generally be that amount which produces a therapeutic effect, but may also be a lesser amount. Generally, this amount will range from about 0.01 percent to about 99 percent, from about 0.1 percent to about 70 percent, or from about 1 percent to about 30 percent relative to the total weight of the dosage form.
  • one or more additional therapeutic agents may be included in the composition.
  • additional therapeutic agents are described herein in the “Methods of Treatment” section, including the “Combination Therapy” subsection.
  • the suitable amount of the additional therapeutic agent to be included in the composition can be readily selected by a person skilled in the art, and will vary depending on a number of factors, such as the particular agent and carriers used, dosage form, and desired release and pharmacodynamic characteristics.
  • the amount of the additional therapeutic agent included in a single dosage form will generally be that amount of the agent, which produces a therapeutic effect, but may be a lesser amount as well.
  • an article of manufacture comprising materials useful for the treatment of a cancer.
  • the article of manufacture can comprise a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition, which is effective for treating a coronavirus infection, described herein, and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle) .
  • Package insert refers to instructions customarily included in commercial packages of therapeutic products that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products.
  • the package insert indicates that the composition is used for treating a cancer.
  • the label or package insert may further comprise instructions for administering the composition to a patient.
  • the article of manufacture may further comprise a second container comprising a pharmaceutically acceptable buffer, such as bacteriostatic water for injection (BWFI) , phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • a pharmaceutically acceptable buffer such as bacteriostatic water for injection (BWFI) , phosphate-buffered saline, Ringer's solution and dextrose solution.
  • Kits are also provided that are useful for various purposes, e.g., for treatment of a cancer described herein, optionally in combination with the articles of manufacture.
  • Kits of the present application include one or more containers comprising any one of the compositions described herein (or unit dosage form and/or article of manufacture) .
  • the kit further comprises other agents (e.g., one or more additional therapeutic agents) and/or instructions for use in accordance with any of the methods described herein.
  • the kit may further comprise a description of selection of individuals suitable for treatment.
  • kits of the present application are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit) , but machine-readable instructions (e.g., instructions carried on a magnetic or optical storage disk) are also acceptable.
  • a label or package insert e.g., a paper sheet included in the kit
  • machine-readable instructions e.g., instructions carried on a magnetic or optical storage disk
  • kits comprising a pharmaceutical composition comprising any one of the anti-CD137 antibodies described herein and a pharmaceutically acceptable carrier; and instructions for administering the pharmaceutical composition to a subject having a cancer.
  • the kit further comprises a pharmaceutical composition comprising an additional therapeutic agent, such as an immune checkpoint inhibitor or a chemotherapeutic agent.
  • the kit comprises one or more assays or reagents thereof for determining a level of one or more biomarkers described herein (e.g., total CD137, mCD137, sCD137, CD137L, Ki67, NK cells, T em and/or T reg ) .
  • the kit comprises an IHC assay for determining a level of CD137L in a sample.
  • the kit comprises an anti-CD137L antibody (e.g., TY23561) .
  • the IHC assay is a multiplex IHC assay for determining levels of two or more (e.g., 3, 4, 5, 6, or more) biomarkers, such as total CD137, CD137L, and PD-L1.
  • the kit comprises an immunoassay (e.g., MSD assay) for determining a level of sCD137.
  • kits of the present application are in suitable packaging.
  • suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags) , and the like. Kits may optionally provide additional components such as buffers and interpretative information.
  • the present application thus also provides articles of manufacture, which include vials (such as sealed vials) , bottles, jars, flexible packaging, and the like.
  • the containers may be unit doses, bulk packages (e.g., multi-dose packages) or sub-unit doses.
  • Kits may also include multiple unit doses of the pharmaceutical compositions and instructions for use and packaged in quantities sufficient for storage and use in pharmacies, for example, hospital pharmacies and compounding pharmacies.
  • the CD137L binding site on CD137 as well as the epitopes of Utomilumab and Urelumab are determined based on the crystal structures with PDB IDs 6BWV, 6A3W and 6MHR respectively.
  • the epitope of ADG106 is determined based on the crystal structure of CD137-ADG106 complex.
  • Crystals of the ADG106 fragment in complex with CD137 were obtained using sitting-drop vapor diffusion set-ups. Well diffracting crystals appeared within 4 days and grew to full size over 14 days. Crystals were cryo-protected by the addition of glycerol to a final concentration of 10% (v/v) to the crystallization drop before mounting. A complete data set of an ADG106 fragment/CD137 crystal was collected and the data were integrated, analyzed and scaled. Complex formation results in a buried accessible surface area of approximately 2200 between the ADG106 fragment and CD137, with the area being approximately equally distributed between the variable regions of the light and heavy chains of the ADG106 fragment. The complex is stabilized through the formation of the prominent direct hydrogen bonds listed in Table 1, in addition to a number of van der Waal’s interactions and water-mediated hydrogen bonds.
  • FIGS. 1A-1D show in grey amino acid residues in CD137 that are within or from one or more amino acid residues from CD137L, ADG106, Utomilumab or Urelumab.
  • the epitope of ADG106 include amino acid residues 51, 63-67, 69-73, 83, 89, 92, 98-104 and 112-114 on CD137, the majority of which are located in the CDR2 domain of CD137.
  • ADG106 has a distinct epitope compared to the epitopes of known anti-CD137 antibodies Utomilumab and Urelumab. The epitope of ADG106 resembles the binding site of CD137L.
  • the epitope mapped by domain swapping/deletion plus site-directed mutagenesis has shown the binding epitope of CD137 across human, monkey and mouse species, this epitope spans domain some c-terminal part of domain 1 and most of the domains 2 and 3 (see WO2019/037711) ; the X-ray crystal structure complex between human CD137 and ADG106 shows that the epitope contact of CD137 with ADG106 locates mostly in domain 2 and 3.
  • CD137 It is known the interaction between CD137 and its antibody in solution is dynamic in nature and would interchange between different conformations between CD137 and Adg106, although most of the contact sites in complex structure is consistent with epitope mapping by domain swapping/deletion plus site-directed mutagenesis, except for the C-terminal of domain one of CD137 which is not observed in the X-ray structure; however, using CD137 structure from its complex with its ligand in an alternative conformation, the C-terminal of domain of CRD1 of CD137 would interact with ADG106 at the previously mapped site by directed mutagenesis at 34 to 36 of CD137; therefore 34 to 36 residues would be considered to encompass CRD1 if CD137 dynamic fraying of its conformation is considered as shown by mutagenesis although the X-ray structure observed did not include them.
  • the information provide here offer a more comprehensive understanding in the dynamic nature of the CD137 and ADG106 interaction, which would be important for their functional interpretation
  • This example describes a phase 1, multicenter, open-label, dose-escalation and dose-expansion study to the safety and efficacy of ADG106 in patients with solid tumors and/or non-Hodgkin lymphoma.
  • the primary objective of the study is assessment of the safety and tolerability of ADG106.
  • the secondary objectives of the study are determination of the pharmacokinetic (PK) profile of ADG106, determination of the immunogenicity of ADG106, and evaluation of the antitumor activity of ADG106.
  • PK pharmacokinetic
  • ADG106 is a fully human agonistic anti-CD137 monoclonal IgG4 antibody.
  • ADG106 targets the evolutionally conserved epitope of CD137 with cross-species reactivity across mouse, rats, money and human CD137 and exhibits novel mechanism of action for CD137 agonism, CD137 antagonism and potent cross-linking via FcgRIIb.
  • WOCBP Women of childbearing potential
  • Active central nervous system primary or secondary malignancies, active seizure disorder, spinal cord compression, or carcinomatous meningitis.
  • HIV human immunodeficiency virus
  • HBV hepatitis B virus
  • HCV hepatitis C virus
  • Any previous anti-CD137 mAb e.g., Utomilumab, or Urelumab treatment.
  • the patient After receiving the last treatment (chemotherapy, radiotherapy, biotherapy, or other research drugs) , the patient had a washout period of at least 4 weeks or more than 5 half-lives and had recovered from any toxic reaction of the previous treatment to less than 1 degree.
  • WOCBP Women of childbearing potential
  • Coagulation function was basically normal, INR ⁇ 1.5.
  • HCV antibody active hepatitis B (HBV DNA ⁇ 10000 copies/mL or 2000 IU/mL)
  • active hepatitis B HBV DNA ⁇ 10000 copies/mL or 2000 IU/mL
  • positive hepatitis virus taking antiviral drugs.
  • HIV human immunodeficiency virus
  • Any active autoimmune disease, evidence-based autoimmune disease, or systemic syndrome requiring systemic steroids or immunosuppressive drugs except for inactive vitiligo, psoriasis, asthma/specific reactivity in children after treatment within two years, or thyroid diseases controlled by alternative therapy/non-immunosuppressive therapy) .
  • the residual toxicity of the patient's previous treatment was more than grade 1.
  • any uncontrollable serious clinical problems include but not limited to: evidence of severe or uncontrollable systemic diseases (such as unstable or uncompensated respiratory, cardiac, liver, or kidney diseases) ; any unstable systemic diseases (including active infections, refractory high or drug failure Controlled hypertension (>150/100 mmHg) , unstable angina pectoris, congestive heart failure, liver and kidney or metabolic diseases) .
  • severe or uncontrollable systemic diseases such as unstable or uncompensated respiratory, cardiac, liver, or kidney diseases
  • any unstable systemic diseases including active infections, refractory high or drug failure Controlled hypertension (>150/100 mmHg) , unstable angina pectoris, congestive heart failure, liver and kidney or metabolic diseases.
  • a clear history of neurological or psychiatric disorders including epilepsy or dementia.
  • Phase 1a Accelerated Titration and Conventional Dose Escalation Studies
  • phase 1a dose escalation study in US included accelerated titration (0.03, 0.1 and 0.3 mg/kg) and conventional dose escalation (1, 3, and 10 mg/kg) .
  • the phase 1a dose escalation study in China included accelerated titration (0.1 mg/kg) and conventional dose escalation (0.5, 1.5, 3, 5, and 10 mg/kg) .
  • ADG106 was administered by intravenous infusion over 60 minutes on Day 1 of each cycle once every three weeks (Q3W) .
  • phase 1a accelerated titration and conventional dose escalation studies in US and China, 33 patients in 9 cohorts were enrolled. There were 5 patients with adenoid cystic carcinoma, 5 patients with colon cancer, 5 patients with non-small cell lung cancer (NSCLC) , 2 patients with follicular lymphoma, 3 patients with nasopharyngeal carcinoma (NPC) , and one each with anal, fibro-lung, fusiform cell, malignant pleural mesothelioma, mantle cell lymphoma, ovarian, breast, esophageal, endometrial, gastrointestinal (GI) , cholangiocarcinoma, appendiceal, and sebaceous cancer.
  • NSCLC non-small cell lung cancer
  • NPC nasopharyngeal carcinoma
  • phase 1b dose expansion study in US and China, 7 patients in 2 cohorts were enrolled (Table 2) . There were two patients with nasopharyngeal caner (NPC) , and one each with head and neck squamous cell carcinoma (HNSCC) , mesothelioma, Sigmoid colon carcinoma, angioimmunoblastic T cell lymphoma, and melanoma.
  • NPC nasopharyngeal caner
  • HNSCC head and neck squamous cell carcinoma
  • mesothelioma mesothelioma
  • Sigmoid colon carcinoma mesothelioma
  • angioimmunoblastic T cell lymphoma angioimmunoblastic T cell lymphoma
  • melanoma melanoma
  • Table 3B shows demographics and major cancer types of patients enrolled in the Chinese studies. Among the 23 Chinese patients tested so far, median treatment duration was 18.35 weeks (with minimum of 12.1 weeks and maximum of 33.1 weeks) .
  • Efficacy of ADG106 was measured by the percentage of stable disease achieved and decline in standardized uptake values (SUV) on PET CT images. Among 40 patients in 7 cohorts, 17 (42.5%) patients achieved stable disease. Among the 17 patients who achieved stable diseases, 7 patients were observed to have tumor size reduction (Table 4) . Table 4. Over all ADG106 efficacy data of accelerated titration phase, dose escalation phase, and dose expansion phase studies.
  • FIG. 2 and FIG. 3 show combined efficacy data of patients from both Chinese and U.S. clinical studies.
  • FIG. 2 demonstrates a general trend of prolonged time on treatment and duration of response in patients who received higher dose levels of ADG106.
  • FIG. 3 shows tumor shrinkage in several patients. One patient who experienced initial tumor enlargement had slight tumor shrinkage after receiving ADG106 treatment.
  • FIGS. 16A-16B show PET CT images of a 46 years old male patient with stage III angioimmunoblastic T cell lymphoma, who was treated with ADG106.
  • Prior therapies included chemotherapy, folate analog metabolic inhibitor, and autologous hematopoietic stem cell transplantation.
  • the patient achieved stable disease (SD) while receiving ADG106, an overall 33%tumor shrinkage of the targeted lesions after receiving one dose ADG106 and are supported by the biomarker studies described in Figures 15A-D. Shrinkage of two tumors were observed with 52%and 16%decrease in the volume of each tumor after only one administration of ADG106.
  • TEAEs treatment emergent adverse events
  • the most common TEAEs were decreased appetite (10%) , anemia (10%) , arthralgia (10%) , lymphopenia (15%) , dyspnea (10%) , and respiratory failure (10%) .
  • G3 anemia at 10mg/kg cohort was drug related and rest G3 TEAEs were not related to the study treatment. There was no drug related death up to 10 mg/kg.
  • * represents two laboratory abnormalities/G3 occurred in one same patient.
  • the receptor occupancy of Urelumab at its maximal tolerated clinical dose is estimated to be 64%, whereas Utomilumab is 98%at its maximal administered clinical dose (MAD, 10mg/kg) . Therefore, ADG106 can achieve high receptor occupancy at a dose 10 times below its DLT dose.
  • Pharmacokinetic profiles of ADG106 in subjects at dose levels of 0.03 mg/kg, 0.1 mg/kg, 0.3 mg/kg, 0.5 mg/kg, 1 mg/kg, 1.5mg/kg, 3 mg/kg, 5 mg/kg, and 10 mg/kg were determined.
  • Blood samples for PK analysis were collected at Cycle 1 (Day 1: pre-dose and 2, 6, 12, and 24 hours after the end of infusion; once on Days 8, 15, and 22) , Day 1 pre-dose and 2 hours after the end of infusion of subsequent cycles, and at the End of Treatment.
  • the concentrations of ADG106 were determined from the serum samples using a validated ELISA method, in which an anti-ADG106 idiotype mouse monoclonal antibody was used to coated ELISA microplates for capture, and an HRP-labeled goat anti-hIgG4-Fc polyclonal antibody was used for detection. Mean serum concentrations of ADG106 in each dose group versus time were plotted. PK parameters were estimated using a non-compartmental method with WinNonlin.
  • FIGs. 6A-6B show dose-dependent proportional increase of serum ADG106 levels in response to systemic exposure.
  • the mean half-life of ADG106 at doses ⁇ 0.5mg/kg is around 7 days.
  • ADG106 achieved peak concentrations (Cmax) of 4.25, 4.18, 7.95, 23.3, and 95.03 ⁇ g/L for 0.03, 0.1, 0.3, 1, and 3 mg/kg dose levels (Table 10, FIG. 6) , respectively.
  • the AUC0-t values were 626, 668, 1387, 3876.7, and 18420 ⁇ g/L ⁇ h for 0.03, 0.1, 0.3, 1, and 3 mg/kg dose levels (Table 9) , respectively.
  • the ADG106 terminal phase half-life (t1/2) was 87, 108, 159, 146.3, and 149 hours for 0.03, 0.1, 0.3, 1, and 3 mg/kg dose levels, respectively.
  • ADG106 achieved peak concentrations (Cmax) of 2.3, 11.2, 27.2, 62.9, 155 and 255 ⁇ g/L for 0.1, 0.5, 1.5, 3, 5 and 10 mg/kg dose levels.
  • the AUC0-t values were 255, 1761, 4162, 10016, 20966, and 39658 ⁇ g/L ⁇ h for 0.1, 0.5, 1, 1.5, 3, 5, and 10 mg/kg dose levels (Table 10) , respectively.
  • the ADG106 terminal phase half-life (t1/2) was 3.7, 6.1, 7.5, 8.9, 7.2 and 5.5 days for 0.1, 0.5, 1.5, 3, 5, and 10 mg/kg dose levels, respectively.
  • Immunogenicity of ADG106 in subjects at dose levels of 0.03 mg/kg, 0.1 mg/kg, 0.3 mg/kg, 0.5mg/kg, 1 mg/kg, 1.5mg/kg, 3 mg/kg, 5 mg/kg, and 10 mg/kg were determined.
  • Blood samples were collected as described in the PK analysis.
  • the anti-drug-antibody (ADA) to ADG106 in human serum was measured using a validated Affinity Capture and Elution (ACE) based immunoassay. All samples were first analyzed for ADAs in a screening assay. Study samples with results below the screening cut-off were reported as negative for ADAs. In the event of a positive result in the screening assay, samples were analyzed in the confirmatory assay. All samples confirmed positive were reported as positive.
  • the immunogenicity studies show that treatment-induced anti-drug antibodies were developed in about 20%of the patients tested. ADG106 treatment emergent or boosted ADA occurred in 5 of 23 Chinese patients tested so far.
  • Blood samples for PK/TK analysis were collected at different time points in each dosing cycle.
  • concentrations of ADG106 were determined from the serum samples using a validated ELISA method, in which an anti-ADG106 idiotype mouse monoclonal antibody was used to coat ELISA microplates for capture, and an HRP-labeled goat anti-hIgG-Fc polyclonal antibody was used for detection.
  • Mean serum concentrations of ADG106 in different dosage groups versus time were plotted.
  • PK parameters were estimated using a non-compartmental method with WinNonlin.
  • ADA levels against ADG106 in monkey serum samples were measured using a validated electrochemiluminescence (ECL) assay. All samples were first analyzed for ADAs in a screening assay. Study samples with results below the screening cut-off were reported as negative for ADAs. In the event of a positive result in the screening assay, samples were analyzed in the confirmatory assay. All samples confirmed positive were reported as positive.
  • ECL electrochemiluminescence
  • ADAs were present and correlated with effects of increased ADG106 clearance and reduced exposure in individual animals. These ADA positive titers and higher ADG106 clearances were mainly observed at 50 mg/kg (4/10 animals) and in 1/10 animals at 100 mg/kg, but not in animals in the 200 mg/kg treated group.
  • peripheral blood was collected prior to treatment initiation and at each ADG106 treatment cycle.
  • T cell proliferation was examined by analyzing Ki-67 expression using flow cytometry analysis.
  • Soluble CD137 (sCD137) levels in plasma were also examined using a validated MSD-based electrochemiluminescence assay.
  • FIGs. 10A-10B show CD137+CD4+ and CD137+CD8+ T cell populations in certain individuals treated with various levels of ADG106.
  • Biomarkers include serum levels of pro-inflammatory cytokines (TNF ⁇ , IFN ⁇ , IL-2, IL-6, IL-10, etc. ) and soluble CD137.
  • Peripheral blood immune cell profiles such as absolute cell counts for circulating T cells, natural killer (NK) cells, B cells, effector T cell subpopulations, and memory T cell subpopulations, were analyzed by flow cytometry using respective antibodies for each biomarker.
  • the dynamic changes of peripheral T cell clones were analyzed through TCR sequencing. Expression of CD137 and its ligand, PD-L1, and/or tumor infiltrating lymphocytes, etc., were also analyzed by immunohistochemistry (IHC) on archival tumor tissue or fresh biopsy tissue using validated methods.
  • IHC immunohistochemistry
  • ADG106 treatment significantly increased the plasma levels of soluble CD137 (sCD137) in all patients tested, which suggests that ADG106 treatment induced CD137 expression in the patients.
  • sCD137 plasma levels increased more in patients with stable diseases than patients with progressive diseases
  • mCD137 plasma levels increased more in patients with progressive diseases than patients with stable disease after one cycle of ADG106 treatment.
  • the increase of CD137 expression upon ADG106 treatment indicates that ADG106 engages the activation of the CD137 signaling pathway in CD8+ T cells.
  • Ki67+ CD8+ T-cells tended to increase more after one cycle of ADG106 treatment in patients who had stable diseases, in comparison to patients who had progressive diseases.
  • basal levels of CD8+effector memory T (T em ) cells correlated with clinical outcome of ADG106 treatment, in which patients who achieved stable diseases after treatment had significantly lower pre-treatment levels of CD8+ T em cells than patients who had progressive diseases.
  • T em CD8+effector memory T
  • FIGS. 15A-15D show biomarker levels in a patient with angioimmunoblastic T cell lymphoma (Patient R017) who achieved 33%tumor shrinkage after four cycles of ADG106 treatment.
  • the patient had increased proliferation (Ki67) and decreased mCD137 levels of CD8+ T-cells, and decreased Treg cells, the levels of CD8+ T em cells increased initially upon ADG106 treatment.
  • mice were inoculated subcutaneously at the right flank with L5178-R or L5178-S murine T cell lymphoma tumor cells (1 x 10 5 ) in 0.1 ml of PBS for tumor development.
  • the animals were randomized and treatment started when tumor volumes reach 50-80 mm 3 , or on the day of cell inoculation (FIGs. 17A and 17C) .
  • Each group consisted of 8 tumor-bearing mice.
  • Isotype control or ADG106 at 20mg/kg dose was administered by intraperitoneal injection once every 3 days. Tumor growth and animal body weight were monitored every 2-3 days.
  • ADG106 has antitumor activity in the L5178-S T lymphoma model, whereas the L5178-R T lymphoma model is resistant to ADG106 treatment.
  • Expression of CD137 ligand was examined in the two murine T lymphoma L5178-R and L5178-S cells. These lymphoma cells were stained with PE-labeled anti-CD137 ligand or isotype control antibody by incubation for 30-60 min on ice in dark. After washing off unbound antibodies, the samples were analyzed by flow cytometry. FIGs.
  • 17B and 17D show the staining patterns of isotype control (black) and anti-CD137 ligand antibodies (grey) in L5178-R and L5178-S cells, respectively.
  • the results indicate that L-5178-R T lymphoma cells are positive for CD137 ligand expression, whereas L5178-S T lymphoma cells are negative for CD137 ligand expression.
  • L5178-R is resistant while L5178-S is sensitive to ADG106 treatment.
  • these two T cell lymphoma models differ in terms of the CD137 ligand expression status: L5178-R is positive whereas L5178-S is negative for CD137 ligand expression.
  • CD137 ligand is the natural agonist for CD137 and ADG106 works by the same mechanism to activate CD137 receptor
  • the resistance of L5178-R T lymphoma to ADG106 treatment is consistent with the hypothesis that tumors with overexpression of the CD137 ligand could have developed resistance to agonists that have similar mechanism of action in the same pathway, such as anti-CD137 antibodies like ADG106, during the course of tumorigenesis.
  • Such data support that CD137 ligand expression status in tumors may be useful for stratifying patients in treatment with CD137 agonists.
  • RP2D Recommended Phase 2 Dose
  • the following example describes in vivo therapeutic efficacy of the anti-CD137 antibody ADG106 in combination with various immune checkpoint inhibitors in murine cancer models.
  • 3LL 3LL
  • tumors were established (75 mm 3 )
  • treatment began with isotype control antibody, ADG106 (10mg/kg) , (atezolizumab, 10 mg/kg) , or the combination of ADG106 (10mg/kg) and (atezolizumab, 10 mg/kg) by intraperitoneal injection twice a week for 3 weeks. Tumor growth was monitored twice weekly and reported as mean tumor volume ⁇ SEM over time.
  • both ADG106 and (atezolizumab) monotherapies showed anti-tumor activity, and the combination of ADG106 with (atezolizumab) exhibited synergistic anti-tumor efficacy in the 3LL lung cancer model.
  • ADG106 and (atezolizumab) each as single agents could inhibit tumor growth at certain degrees.
  • Combination of ADG106 and (atezolizumab) further enhanced antitumor efficacy, leading to complete tumor regression in 7/10 mice, which suggests a synergistic effect between these two agents.
  • both ADG106 and anti-PD-1 antibody monotherapies showed anti-tumor activity, and the combination of ADG106 with anti-PD-1 antibody exhibited synergistic anti-tumor efficacy in the Lewis lung cancer model.
  • ADG106 and anti-PD-1 antibody 2E5 each as single agents could significantly inhibit tumor growth.
  • Combination of ADG106 and anti-PD-1 antibody 2E5 further enhanced antitumor efficacy, leading to complete tumor regression, which suggests a synergistic effect between these two agents.
  • both ADG106 and ADG116 monotherapies showed anti-tumor activity, and the combination of ADG106 with ADG116 exhibited synergistic anti-tumor efficacy in the 4T1 breast cancer model.
  • ADG106 and ADG116 each as single agents could inhibit 4T1 tumor growth.
  • Combination of ADG106 and ADG116 further enhanced antitumor efficacy, suggesting a synergistic effect between these two agents.
  • results demonstrate that ADG106 can synergize with various immune checkpoint inhibitors to enhance antitumor efficacy in different mouse syngeneic cancer models.
  • Example 8 ADG106 in combination with chemotherapeutic agents in murine cancer models
  • the following example describes in vivo therapeutic efficacy of the anti-CD137 antibody ADG106 in combination with chemotherapeutic agent docetaxel in 4T1 breast cancer model mice, and ADG106 in combination with chemotherapeutic agent cisplatin in Lewis lung cancer model mice.
  • ADG106 and Docetaxel mono-therapies were well tolerated by mice and showed marginal anti-tumor activity.
  • the combination of ADG106 with Docetaxel exhibited enhanced anti-tumor efficacy in the 4T1 breast cancer model. No obvious toxicity was observed during the study.
  • both ADG106 and cisplatin monotherapies showed anti-tumor activity, and the combination of ADG106 with cisplatin exhibited synergistic anti-tumor efficacy in the Lewis lung cancer model. No obvious toxicity was observed during the study.
  • results demonstrate that ADG106 can synergize with various chemotherapeutic agents to enhance antitumor efficacy in different mouse syngeneic cancer models.
  • Example 9 ADG106 in combination with anti-CD20 antibody in a murine lung cancer model
  • Example 10 ADG106 in combination with local radiation in a murine colon cancer model
  • FIGs. 25A-25B indicate that a single dose radiation on tumor significantly inhibited tumor growth, whereas ADG106 alone had marginal if any antitumor activity. Moreover, combination of these two treatments led to even stronger antitumor efficacy, suggesting that ADG106 could enhance the antitumor effect of radiation therapy in this colon cancer model.
  • Example 11 Identification of NK cell proliferation as a pharmacodynamic biomarker in first-in-human phase 1 trial with ADG106
  • TBNK flow cytometry assay was employed to measure the composition of T (CD3+, CD4+/CD8+ subsets) , B (CD19+) and NK (CD16+/CD56+) cells in peripheral blood samples. NK cell levels were determined as percentages of CD45+ lymphocytes and absolute cell counts.
  • the results shown in FIG. 26A demonstrate proliferation of NK cells in patients treated with ADG106 after the first cycle of treatment, the post-treatment NK cell levels (Post) were measured at Day 8/C1D8, Day 15/C1D15, and/or Day 21/C1D21 after the first treatment (Table 11) .
  • the post-treatment NK cell levels were measured at Day 8/C1D8, Day 15/C1D15, and/or Day 21/C1D21 after the first treatment (Table 11) .
  • 88% (28/32) patients showed an increase of the NK cell level in their peripheral blood, 6% (2/32) showed no change, and 6% (2/32) showed a decrease of the NK cell level upon the ADG106 treatment.
  • FIG. 26B demonstrate dose-dependent stimulations of NK cell proliferation by ADG106 treatment at doses of 0.1mg/kg, 0.5mg/kg, and 1.5 mg/kg. No significant difference in NK cell proliferation effect was observed at doses higher than 1.5 mg/kg, suggesting that ADG106 might have saturated CD137 receptors on NK cells at doses ⁇ 1.5 mg/kg.
  • Table 11 Percentage of NK cells (CD3-CD56+/CD16+) before and after ADG106 treatment.
  • NK cell proliferation is an indication for CD137 target engagement in cancer patients.
  • the results shown in FIG. 27A indicate that the average baseline percentage of NK cells was lower in patients who had a stable disease (S) than those whose cancer progressed (P) .
  • the results shown in FIG. 27B indicate that patients with a stable disease (S) had a higher average percentage increase in NK cells compared to patients whose disease progressed (P) after treatment with ADG106.
  • NK cell proliferation could serve as a pharmacodynamic biomarker for anti-CD137 antibody treatment in cancer patients.
  • Example 12 Predict patient response to anti-CD137 antibody treatment by measuring CD137 and CD137L expression levels
  • biomarker studies were conducted to explore predictive and pharmacodynamics (PD) biomarkers.
  • sCD137 levels in blood samples from patients treated with ADG106 in the Phase I clinical trial were detected using Human 4-1BB/TNFRSF9 DuoSet ELISA (R&D Cat#: DY838) and the expression levels were measured using a highly sensitive electrochemiluminescence Meso Scale Discovery (MSD) assay.
  • MSD electrochemiluminescence Meso Scale Discovery
  • FIG. 28A show a significant correlation between nasopharyngeal cancer (NPC) patients’ clinical outcome with changes in their soluble CD137 (sCD137) expression levels, which is expressed as ratios of induced changes in soluble CD137 levels (C2-C1) (i.e., sCD137 level after treatment at time point C2 subtracted by sCD137 level before treatment at time point C1) to baseline sCD137 expression levels (C1) (i.e., sCD137 level before treatment at time point C1) .
  • C2-C1 i.e., sCD137 level after treatment at time point C2 subtracted by sCD137 level before treatment at time point C1
  • baseline sCD137 expression levels C1
  • sCD137 level i.e., sCD137 level before treatment at time point C1 .
  • P progressive disease
  • S stable disease
  • PR partial response
  • results shown in FIG. 28B demonstrate dose-dependent induction of soluble CD137 expression by ADG106 treatment at doses of 3 mg/kg, 5 mg/kg, and 10 mg/kg.
  • results in FIGs. 28A-28B demonstrate that the ratios of induced changes in soluble sCD137 expression levels (C2-C1) to baseline sCD137 expression levels (C1) could serve as a good indicator for the selection of recommended phase 2 dose (RP2D) .
  • mIHC multiplex immunohistochemistry
  • FFPE formalin fixed and paraffin embedded
  • the tissue slides were heated in an oven for 30 min at 65°C, then stained with primary and secondary antibodies against CD137, CD137L, and PD-L1 using the preset staining program on the IHC stainer (Leica RX) . Then, the tissue slides were mounted by proper side mounting media and the whole stained sections were scanned and imaged by the PERKINELMER 3.0 Imaging system. The scanned images of the patient sections were then analyzed using the software package.
  • the CD137L protein expression level was quantified as percentage of CD137L expression or CD137L (%) , which is the number of CD137L positive cells divided by the number of all the nucleated cells.
  • CD137L expression levels were measured in formalin-fixed paraffin-embedded (FFPE) tumor biopsy specimens of 24 patients treated with ADG106. The correlation of CD137L expression levels with patients’ clinical response to the ADG106 treatment was analyzed.
  • FFPE formalin-fixed paraffin-embedded
  • FIGs. 29A-29C indicate that patients expressing high levels of CD137L had better response to the ADG106 treatment.
  • one nasopharyngeal carcinoma (NPC) patient with a 43.78%CD137L expression level experienced a 40%tumor reduction after two cycles of ADG106 treatment (FIGs. 30A- 30B) .
  • Two lymphoma patients with 23.08%and 20.54%CD137L expression levels had 36%and 32%tumor shrinkage respectively after two cycle of treatment (FIG. 31) .
  • a NSCLC patient with a 40%CD137L expression level only showed moderate response (i.e., Stable Disease) , which may be due to the lower ADG106 dose administered to the patient during dose escalation (Table 12) .
  • the results in Table 12 also demonstrate a correlation between the ratio of induced change of CD137 protein expression levels to baseline CD137 levels and patients’ clinical response.
  • Low CD137 baseline levels also correlated with a progressive disease (PD) response.
  • PD progressive disease
  • the correlation between the CD137L expression levels with patients’ clinical response is stronger in comparison.
  • the Phase 2 study is a biomarker-guided study that investigates treatment efficacy among patients with advanced solid tumors or hematological malignancies.
  • the first arm involves monotherapy of ADG106
  • the second arm involves the combination therapy of ADG106 and an anti-PD-1 antibody, Toripalimab (JUNSHI BIOSCIENCES) .
  • a 15-20%CD137L expression level is used as the initial cut-off value for designating patients as biomarker positive (i.e., higher expression level than the cut-off value) versus biomarker negative (i.e., lower expression level than the cut-off value) .
  • the CD137L expression level cut-off value is further validated with more patient data from a biomarker validation group.
  • a chromogenic IHC assay is developed and cross-validated with the fluorescence IHC assay described in Example 12. Either the chromogenic IHC assay or the fluorescence IHC assay may be used as a companion diagnostic test for ADG106 monotherapy, and the combination therapy of ADG106 and Toripalimab.
  • Part 1 of the Phase 2 study involves patient stratification.
  • An interim analysis is performed after Part 1 of the Phase 2 study. If the overall response rate (ORR) is higher than 30%and/or the ORR is higher in biomarker (+) patients, Part 2 of the study is initiated.
  • ORR overall response rate
  • Part 2 of the Phase 2 study involves patient pre-selection.
  • biomarker-positive patients are further divided into two arms: patients in the first arm receive ADG106 monotherapy, and patients in the second arm receive a combination therapy of ADG106 and Toripalimab.
  • the primary objectives of the Phase 2 study are to study the overall response rate (ORR) and duration of the response.
  • the secondary objectives of the Phase 2 study are to study the progression free survival (PFS) rate and overall survival rate.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Endocrinology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

L'invention concerne des compositions et des méthodes pour le traitement de cancers, notamment du lymphome folliculaire, du lymphome T et du carcinome adénoïde kystique, à l'aide d'un anticorps anti-CD137 qui se lie spécifiquement à un domaine extracellulaire du CD137 humain. Dans certains modes de réalisation, l'invention concerne des polythérapies comprenant l'anticorps anti-CD137 et un inhibiteur de point de contrôle immunitaire et/ou un agent chimiothérapeutique. L'invention concerne également des biomarqueurs tels que le CD137 total, le CD137 lié à la membrane (mCD137), le CD137 soluble (sCD137), le ligand CD137, le Ki67, des cellules mémoires effectrice T (Tem) CD8+, des cellules T régulatrices (Treg) et des niveaux de cellules tueuses naturelles (NK) pour lesdites méthodes de traitement.
PCT/CN2020/115795 2020-05-13 2020-09-17 Compositions et méthodes pour le traitement du cancer WO2021227326A1 (fr)

Priority Applications (8)

Application Number Priority Date Filing Date Title
US17/924,540 US20230133118A1 (en) 2020-05-13 2021-05-13 Compositions and methods for treating cancer
CN202180048790.9A CN115768525A (zh) 2020-05-13 2021-05-13 用于治疗癌症的组合物和方法
PCT/CN2021/093511 WO2021228178A1 (fr) 2020-05-13 2021-05-13 Compositions et méthodes pour le traitement du cancer
TW110117374A TW202200622A (zh) 2020-05-13 2021-05-13 用於治療癌症之組成物及方法
CA3177717A CA3177717A1 (fr) 2020-05-13 2021-05-13 Compositions et methodes pour le traitement du cancer
AU2021271971A AU2021271971A1 (en) 2020-05-13 2021-05-13 Compositions and methods for treating cancer
EP21803953.5A EP4149547A1 (fr) 2020-05-13 2021-05-13 Compositions et méthodes pour le traitement du cancer
JP2022568397A JP2023525085A (ja) 2020-05-13 2021-05-13 がんを治療するための組成物および方法

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CNPCT/CN2020/090073 2020-05-13
PCT/CN2020/090073 WO2021226883A1 (fr) 2020-05-13 2020-05-13 Compositions et procédés de traitement du cancer par soumission d'un listage des séquences sur un fichier texte ascii
PCT/CN2020/094278 WO2021227156A1 (fr) 2020-05-13 2020-06-04 Compositions et méthodes pour le traitement du cancer
CNPCT/CN2020/094278 2020-06-04

Publications (1)

Publication Number Publication Date
WO2021227326A1 true WO2021227326A1 (fr) 2021-11-18

Family

ID=78526081

Family Applications (3)

Application Number Title Priority Date Filing Date
PCT/CN2020/090073 WO2021226883A1 (fr) 2020-05-13 2020-05-13 Compositions et procédés de traitement du cancer par soumission d'un listage des séquences sur un fichier texte ascii
PCT/CN2020/094278 WO2021227156A1 (fr) 2020-05-13 2020-06-04 Compositions et méthodes pour le traitement du cancer
PCT/CN2020/115795 WO2021227326A1 (fr) 2020-05-13 2020-09-17 Compositions et méthodes pour le traitement du cancer

Family Applications Before (2)

Application Number Title Priority Date Filing Date
PCT/CN2020/090073 WO2021226883A1 (fr) 2020-05-13 2020-05-13 Compositions et procédés de traitement du cancer par soumission d'un listage des séquences sur un fichier texte ascii
PCT/CN2020/094278 WO2021227156A1 (fr) 2020-05-13 2020-06-04 Compositions et méthodes pour le traitement du cancer

Country Status (1)

Country Link
WO (3) WO2021226883A1 (fr)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114306606B (zh) * 2021-11-29 2023-05-26 江苏省人民医院(南京医科大学第一附属医院) cGAS抑制剂在制备治疗T细胞淋巴瘤的药物中的应用

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006029220A2 (fr) * 2004-09-08 2006-03-16 Ohio State University Research Foundation Therapie combinee utilisant des anticorps anti-ctla4 et anti-4-1bb
WO2006088447A1 (fr) * 2005-02-15 2006-08-24 Gtc Biotherapeutics, Inc. Anticorps anti-cd137 en tant qu'agent dans le traitement de cancers et variante de glycosylation dudit anticorps
US20080305113A1 (en) * 2007-06-05 2008-12-11 University Of Ulsan Foundation For Industry Cooperation Pharmaceutical Composition for Preventing or Treating Chronic Graft-Versus-Disease Comprising Anti-CD137 Monoclonal Antibody
US20120076722A1 (en) * 2009-05-14 2012-03-29 University Of Maryland, Baltimore Methods for treating cancers and diseases associated with 4-1bb (cd137) expression
WO2019037711A1 (fr) * 2017-08-21 2019-02-28 Adagene Inc. Molécules anti-cd137 et utilisation correspondante
WO2019105468A1 (fr) * 2017-12-01 2019-06-06 Adagene Inc. Méthodes d'utilisation du ligand cd137 comme biomarqueur pour le traitement avec un anticorps anti-cd137

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190331682A1 (en) * 2016-02-18 2019-10-31 Institut Gustave Roussy Methods and kits for predicting the sensitivity of a subject to immunotherapy
AU2018253176B2 (en) * 2017-04-13 2023-02-02 Agenus Inc. Anti-CD137 antibodies and methods of use thereof
NZ760841A (en) * 2017-07-11 2024-02-23 Compass Therapeutics Llc Agonist antibodies that bind human cd137 and uses thereof
WO2019148445A1 (fr) * 2018-02-02 2019-08-08 Adagene Inc. Anticorps activables dépendant de la précision/du contexte, et leurs procédés de fabrication et d'utilisation
WO2019148444A1 (fr) * 2018-02-02 2019-08-08 Adagene Inc. Anticorps anti-ctla4 et leurs procédés de fabrication et d'utilisation

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006029220A2 (fr) * 2004-09-08 2006-03-16 Ohio State University Research Foundation Therapie combinee utilisant des anticorps anti-ctla4 et anti-4-1bb
WO2006088447A1 (fr) * 2005-02-15 2006-08-24 Gtc Biotherapeutics, Inc. Anticorps anti-cd137 en tant qu'agent dans le traitement de cancers et variante de glycosylation dudit anticorps
US20080305113A1 (en) * 2007-06-05 2008-12-11 University Of Ulsan Foundation For Industry Cooperation Pharmaceutical Composition for Preventing or Treating Chronic Graft-Versus-Disease Comprising Anti-CD137 Monoclonal Antibody
US20120076722A1 (en) * 2009-05-14 2012-03-29 University Of Maryland, Baltimore Methods for treating cancers and diseases associated with 4-1bb (cd137) expression
WO2019037711A1 (fr) * 2017-08-21 2019-02-28 Adagene Inc. Molécules anti-cd137 et utilisation correspondante
WO2019105468A1 (fr) * 2017-12-01 2019-06-06 Adagene Inc. Méthodes d'utilisation du ligand cd137 comme biomarqueur pour le traitement avec un anticorps anti-cd137

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
A. YONEZAWA, S. DUTT, C. CHESTER, J. KIM, H. E. KOHRT: "Boosting Cancer Immunotherapy with Anti-CD137 Antibody Therapy", CLINICAL CANCER RESEARCH, ASSOCIATION FOR CANCER RESEARCH, vol. 21, no. 14, 15 July 2015 (2015-07-15), pages 3113 - 3120, XP055603460, ISSN: 1078-0432, DOI: 10.1158/1078-0432.CCR-15-0263 *
DINH-TOI CHU, NGUYEN DUY BAC, KHANH-HOANG NGUYEN, NGUYEN LE BAO TIEN, VO VAN THANH, VU THI NGA, VO TRUONG NHU NGOC, DUONG THI ANH : "An Update on Anti-CD137 Antibodies in Immunotherapies for Cancer", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 20, no. 8, pages 1822, XP055754446, DOI: 10.3390/ijms20081822 *
TAKUSHI MASU, MASANORI ATSUKAWA, KATSUHISA NAKATSUKA, MASUMI SHIMIZU, DAISHU MIURA, TAEANG ARAI, HIROTOMO HARIMOTO, CHISA KONDO, K: "Anti-CD137 monoclonal antibody enhances trastuzumab-induced, natural killer cell-mediated cytotoxicity against pancreatic cancer cell lines with low human epidermal growth factor-like receptor 2 expression", PLOS ONE, vol. 13, no. 12, pages e0200664, XP055574560, DOI: 10.1371/journal.pone.0200664 *
ZAPATA JUAN M., PEREZ-CHACON GEMA, CARR-BAENA PABLO, MARTINEZ-FORERO IVAN, AZPILIKUETA ARANTZA, OTANO ITZIAR, MELERO IGNACIO: "CD137 (4-1BB) Signalosome: Complexity Is a Matter of TRAFs", FRONTIERS IN IMMUNOLOGY, vol. 9, 15 November 2018 (2018-11-15), XP055867067, DOI: 10.3389/fimmu.2018.02618 *

Also Published As

Publication number Publication date
WO2021226883A1 (fr) 2021-11-18
WO2021227156A1 (fr) 2021-11-18

Similar Documents

Publication Publication Date Title
US20220204637A1 (en) Anti-cd137 molecules and use thereof
US11498972B2 (en) Anti-OX40 antibody and use thereof
WO2021228178A1 (fr) Compositions et méthodes pour le traitement du cancer
CN108235685A (zh) Pd-1拮抗剂与egfr抑制剂的组合
US20220154296A1 (en) Use of anti-pd-1 antibody in preparation of medicament for treating solid tumors
WO2020244574A1 (fr) Anticorps anti-cd137l et leurs procédés d'utilisation
WO2021227326A1 (fr) Compositions et méthodes pour le traitement du cancer
US20240209091A1 (en) Methods for treating cancer using anti-ctla4 antibodies
US20230242663A1 (en) Combination therapy comprising anti-cd137 antibodies
TW202313109A (zh) 用抗cd39抗體治療癌症之方法
US20230365691A1 (en) Use of anti-pd-1 antibody in treatment of nasopharyngeal carcinoma
WO2024097896A1 (fr) Méthodes de traitement du cancer à l'aide d'un anticorps anti-ctla4 en combinaison avec du pembrolizumab
WO2024050429A2 (fr) Méthodes de traitement d'un cancer à l'aide d'anticorps anti-ctla4

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: 20935614

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: 20935614

Country of ref document: EP

Kind code of ref document: A1