WO2021129874A1 - Anticorps anti-ox40 et son utilisation - Google Patents

Anticorps anti-ox40 et son utilisation Download PDF

Info

Publication number
WO2021129874A1
WO2021129874A1 PCT/CN2020/140259 CN2020140259W WO2021129874A1 WO 2021129874 A1 WO2021129874 A1 WO 2021129874A1 CN 2020140259 W CN2020140259 W CN 2020140259W WO 2021129874 A1 WO2021129874 A1 WO 2021129874A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
cancer
seq
antigen
human
Prior art date
Application number
PCT/CN2020/140259
Other languages
English (en)
Chinese (zh)
Inventor
张宏恺
晋瑞娜
王媛
Original Assignee
南开大学
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 南开大学 filed Critical 南开大学
Priority to CN202080073615.0A priority Critical patent/CN114630839A/zh
Publication of WO2021129874A1 publication Critical patent/WO2021129874A1/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
    • 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
    • A61K39/39566Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against immunoglobulins, e.g. anti-idiotypic antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]

Definitions

  • the present invention relates in particular to a novel anti-OX40 antibody, a composition comprising the anti-OX40 antibody, a nucleic acid encoding the anti-OX40 antibody, a method for preparing the anti-OX40 antibody, and the use of the anti-OX40 antibody.
  • nivolumab nivolumab
  • pembrolizumab nivolumab
  • nivolumab nivolumab
  • pembrolizumab nivolumab
  • diseases such as unresectable or metastatic melanoma and metastatic non-small cell lung cancer.
  • Patients treated with these drugs have produced an anti-tumor response measured by improvement in progression-free survival and/or overall survival.
  • the field still needs more cancer treatment products and methods to supplement the existing standard care.
  • PD-1 and CTLA-4 play an immunosuppressive effect in the process of T cell activation, thereby inhibiting the immune killing function of T cells on tumor cells; therefore, blocking monoclonal antibodies against these two targets can relieve this immunosuppression , To restore the anti-tumor immune function of T cells.
  • activated immune checkpoint molecules are gradually becoming new targets for drug development.
  • Activated immune checkpoint molecules mainly refer to the costimulatory signal molecules of T cell activation-T cell costimulatory receptors, belonging to the tumor necrosis factor receptor (TNFR) family, used to regulate the proliferation and activation of T cells And differentiation, including OX40, CD40, 4-1BB and GITR.
  • TNFR tumor necrosis factor receptor
  • the OX40 receptor also known as CD134 and TNFRSF4 (Tumor Necrosis Factor Receptor Superfamily Member 4), is a member of the TNFR superfamily receptor, which is not constitutively expressed on resting naive T cells, unlike CD28.
  • OX40 is a secondary costimulatory immune checkpoint molecule that is expressed 24 to 72 hours after activation; its ligand OX40L (also known as CD252 and TNFSF4) is not expressed on resting antigen presenting cells, but is expressed after activation. The expression of OX40 depends on the complete activation of T cells.
  • OX40 and its ligand OX40L combine to deliver costimulatory signals.
  • the interaction of OX40 and OX40L can recruit TNFR-related (TRAFs) molecules in the intracellular region of OX40 to form a signaling complex containing IKK ⁇ and IKK ⁇ as well as PI3k and PKB (Akt);
  • TCR TNFR-related
  • OX40 also cooperates with TCR signaling through unknown The mechanism enhances intracellular Ca 2+ , thereby enhancing NFAT into the nucleus.
  • OX40 can activate the classic NF- ⁇ B1 pathway or the non-canonical NF- ⁇ B2 pathway, PI3k/PKB and NFAT pathways, thereby regulating the genes that control T cell division and survival, as well as promoting the transcription of cytokine genes and the expression of cytokine receptors , Is essential for cell survival.
  • OX40 signaling can cause down-regulation including CTLA-4 and Foxp3.
  • OX40 When OX40 is combined with its ligand OX40L, it helps to improve the response of the immune system: 1. Increase the survival and expansion of effector T cells and memory T cells, and increase cytokines (such as IL-2, IL-4, IL-5) , IFN- ⁇ ) secretion; 2. Reduce the immunosuppressive activity of regulatory T cells, and further amplify the effect of T cell activation. In the tumor microenvironment, immune activation can lead to the expression of OX40. It can enhance the activation and proliferation of effector T cells, and inhibit regulatory T cells, resulting in a complex anti-tumor immune response. At present, a number of clinical trials of anti-OX40 antibodies for cancer treatment can be retrieved on the Clinical Trials website.
  • the present invention meets the above needs by providing a novel anti-OX40 antibody that specifically binds and activates OX40.
  • the present invention provides an isolated anti-OX40 antibody or antigen-binding fragment thereof, comprising: a heavy chain CDR1 domain shown in SEQ ID NO: 1, and a heavy chain CDR2 domain shown in SEQ ID NO: 2. , And the heavy chain variable region of the heavy chain CDR3 domain shown in SEQ ID NO: 3; and having the light chain CDR1 domain shown in SEQ ID NO: 9, and the light chain CDR2 domain shown in SEQ ID NO: 10, and The light chain variable region of the light chain CDR3 domain shown in SEQ ID NO: 11.
  • the present invention provides an isolated anti-OX40 antibody or antigen-binding fragment thereof, comprising: a heavy chain CDR1 domain shown in SEQ ID NO: 17, and a heavy chain CDR2 domain shown in SEQ ID NO: 18 , And the heavy chain variable region of the heavy chain CDR3 domain shown in SEQ ID NO: 19; and having the light chain CDR1 domain shown in SEQ ID NO: 25, and the light chain CDR2 domain shown in SEQ ID NO: 26, and The light chain variable region of the light chain CDR3 domain shown in SEQ ID NO: 27.
  • the present invention provides an isolated anti-OX40 antibody or antigen-binding fragment thereof, comprising: a heavy chain CDR1 domain shown in SEQ ID NO: 33, and a heavy chain CDR2 domain shown in SEQ ID NO: 34 , And the heavy chain variable region of the heavy chain CDR3 domain shown in SEQ ID NO: 35; and having the light chain CDR1 domain shown in SEQ ID NO: 41, and the light chain CDR2 domain shown in SEQ ID NO: 42, and The light chain variable region of the light chain CDR3 domain shown in SEQ ID NO: 43.
  • the present invention provides an antibody-drug conjugate comprising the OX40 antibody or antigen-binding fragment thereof described herein and another therapeutic agent; preferably, the anti-OX40 antibody or antigen-binding fragment thereof is combined with The additional therapeutic agent is connected by a linker.
  • the present invention provides a nucleic acid that encodes the anti-OX40 antibody or antigen-binding fragment thereof described herein.
  • the present invention provides an expression vector comprising the nucleic acid described herein.
  • the present invention provides a host cell comprising the nucleic acid described herein or the expression vector described herein.
  • the present invention provides a method for producing the anti-OX40 antibody or antigen-binding fragment thereof described herein, which comprises culturing the host cell described herein under conditions suitable for the expression of the antibody or antigen-binding fragment thereof , And recover the expressed antibody or its antigen-binding fragment from the culture medium.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the anti-OX40 antibody or antigen-binding fragment thereof described herein, or the antibody-drug conjugate described herein, or the nucleic acid described herein, or Said expression vector, and pharmaceutically acceptable carrier.
  • the present invention provides the anti-OX40 antibody or antigen-binding fragment thereof described herein, or the antibody-drug conjugate described herein, or the pharmaceutical composition described herein, for use in the treatment of cancer.
  • the present invention provides a method for treating cancer, which comprises administering to a subject in need a therapeutically effective amount of the anti-OX40 antibody or antigen-binding fragment thereof described herein, or the antibody described herein- A drug conjugate, or a pharmaceutical composition as described herein, to treat the cancer.
  • the present invention provides the anti-OX40 antibody or antigen-binding fragment thereof described herein, or the antibody-drug conjugate described herein, or the pharmaceutical composition described herein, in the preparation of a drug for the treatment of cancer In the use.
  • the present invention provides the anti-OX40 antibody or antigen-binding fragment thereof described herein, or the antibody-drug conjugate described herein, or the pharmaceutical composition described herein, which is used in one of the following or Multiple: Inhibition of Treg function (for example, suppression of Treg suppressive function), killing of OX40-expressing cells (for example, cells expressing high levels of OX40), improvement of effector T cell function and/or improvement of memory T cell function, reduction of tumor immunity, Enhance T cell function and/or deplete OX40-expressing cells.
  • Treg function for example, suppression of Treg suppressive function
  • OX40-expressing cells for example, cells expressing high levels of OX40
  • improvement of effector T cell function and/or improvement of memory T cell function reduction of tumor immunity
  • Enhance T cell function and/or deplete OX40-expressing cells Enhance T cell function and/or deplete OX40-expressing cells.
  • the present invention provides an anti-OX40 antibody or antigen-binding fragment thereof as described herein, or an antibody-drug conjugate as described herein, or a pharmaceutical composition as described herein, in preparation for the treatment of the following: Use of one or more drugs: inhibiting Treg function (for example, inhibiting the suppressive function of Treg), killing OX40-expressing cells (for example, cells expressing high levels of OX40), improving effector T cell function and/or improving memory T Cell function, reduce tumor immunity, enhance T cell function and/or reduce cells expressing OX40.
  • Treg function for example, inhibiting the suppressive function of Treg
  • OX40-expressing cells for example, cells expressing high levels of OX40
  • improving effector T cell function and/or improving memory T Cell function reduce tumor immunity, enhance T cell function and/or reduce cells expressing OX40.
  • the present invention provides a pharmaceutical combination comprising the anti-OX40 antibody or antigen-binding fragment thereof described herein, or the antibody-drug conjugate described herein, or the pharmaceutical composition described herein, and One or more additional therapeutic agents.
  • the present invention provides a kit comprising the anti-OX40 antibody or antigen-binding fragment thereof described herein, or the antibody-drug conjugate described herein, or the pharmaceutical composition described herein, preferably It further includes a drug delivery device.
  • Figure 1 500nM Fc-ILZ-OX40L stimulated sorting of Jurkat/NF-KappaB-GFP+hOX40 reporter cell line monoclonal, flow cytometry analysis of the positive rate of GFP expression in cells after stimulation.
  • Phage ELISA method detects the binding of phage monoclonal to OX40 recombinant protein, and calculates the ratio of phage to OX40 and phage to BSA OD450nm.
  • FIG. 3 HEK293FT cell culture supernatants transfected with 4 scFv-Fc expression plasmids with or without cross-linking secondary antibodies stimulated Jurkat/NF- ⁇ B-GFP+hOX40 reporter cells, and flow cytometry analyzed the GFP positive rate of cells.
  • Figure 4 Analysis of aggregation of anti-OX40 antibodies.
  • Figure 5 Anti-tumor efficacy of anti-OX40 antibodies in vivo.
  • the anti-OX40 antibody significantly inhibited tumor growth compared to the control.
  • Figure 6 T cell regulation effect of anti-OX40 antibody. Compared with the control, the anti-OX40 antibody significantly upregulated helper CD4+T cells and cytotoxic CD8+T cells in the spleen and tumors.
  • Figure 7 Dose-dependent effects of cross-linked and soluble anti-OX40 antibodies.
  • the present invention provides an isolated anti-OX40 antibody or antigen-binding fragment thereof, comprising: a heavy chain CDR1 domain shown in SEQ ID NO: 1, and a heavy chain CDR2 domain shown in SEQ ID NO: 2. , And the heavy chain variable region of the heavy chain CDR3 domain shown in SEQ ID NO: 3; and having the light chain CDR1 domain shown in SEQ ID NO: 9, and the light chain CDR2 domain shown in SEQ ID NO: 10, and The light chain variable region of the light chain CDR3 domain shown in SEQ ID NO: 11.
  • the present invention provides an isolated anti-OX40 antibody or antigen-binding fragment thereof, comprising: a heavy chain CDR1 domain shown in SEQ ID NO: 17, and a heavy chain CDR2 domain shown in SEQ ID NO: 18 , And the heavy chain variable region of the heavy chain CDR3 domain shown in SEQ ID NO: 19; and having the light chain CDR1 domain shown in SEQ ID NO: 25, and the light chain CDR2 domain shown in SEQ ID NO: 26, and The light chain variable region of the light chain CDR3 domain shown in SEQ ID NO: 27.
  • the present invention provides an isolated anti-OX40 antibody or antigen-binding fragment thereof, comprising: a heavy chain CDR1 domain shown in SEQ ID NO: 33, and a heavy chain CDR2 domain shown in SEQ ID NO: 34 , And the heavy chain variable region of the heavy chain CDR3 domain shown in SEQ ID NO: 35; and having the light chain CDR1 domain shown in SEQ ID NO: 41, and the light chain CDR2 domain shown in SEQ ID NO: 42, and The light chain variable region of the light chain CDR3 domain shown in SEQ ID NO: 43.
  • the anti-OX40 antibody or antigen-binding fragment thereof described herein comprises the heavy chain variable region shown in SEQ ID NO: 4, or has at least 80%, 81%, and the sequence shown in SEQ ID NO: 4 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% , 99% or 100% identical heavy chain variable region; and the light chain variable region shown in SEQ ID NO: 12, or at least 80%, 81%, 82%, and the sequence shown in SEQ ID NO: 12 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% Or a light chain variable region with 100% identity.
  • the anti-OX40 antibody or antigen-binding fragment thereof described herein comprises the heavy chain variable region shown in SEQ ID NO: 20, or has at least 80%, 81%, and the sequence shown in SEQ ID NO: 20. 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% , 99% or 100% identical heavy chain variable region; and the light chain variable region shown in SEQ ID NO: 28, or at least 80%, 81%, 82%, and the sequence shown in SEQ ID NO: 28 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% Or a light chain variable region with 100% identity.
  • the anti-OX40 antibody or antigen-binding fragment thereof described herein comprises the heavy chain variable region shown in SEQ ID NO: 36, or has at least 80%, 81%, and the sequence shown in SEQ ID NO: 36. 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% , 99% or 100% identical heavy chain variable region; and the light chain variable region shown in SEQ ID NO: 44, or at least 80%, 81%, 82%, and the sequence shown in SEQ ID NO: 44 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% Or a light chain variable region with 100% identity.
  • the anti-OX40 antibody or antigen-binding fragment thereof described herein further comprises a heavy chain constant region and a light chain constant region; preferably, the heavy chain constant region is the heavy chain shown in SEQ ID NO: 5 or 21
  • the constant region or the sequence shown in SEQ ID NO: 5 or 21 has at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical heavy chain constant region; and/or preferably, the light chain constant region is The light chain constant region shown in SEQ ID NO: 13, or at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88% with the sequence shown in SEQ ID NO: 13 , 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical light chain constant region.
  • the anti-OX40 antibody or antigen-binding fragment thereof described herein further comprises a heavy chain signal peptide connected to the heavy chain variable region and/or a heavy chain signal connected to the light chain variable region
  • the heavy chain signal peptide is the heavy chain signal peptide shown in SEQ ID NO: 6, or has at least 80%, 81%, 82%, 83%, 84% with the sequence shown in SEQ ID NO: 6 , 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity
  • the light chain signal peptide is the light chain signal peptide shown in SEQ ID NO: 14, or is at least 80%, 81%, 82% with the sequence shown in SEQ ID NO: 14 %, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 9
  • the anti-OX40 antibody or antigen-binding fragment thereof described herein is an IgG antibody or an antigen-binding fragment thereof, preferably an IgG1 antibody or an antigen-binding fragment thereof.
  • the anti-OX40 antibody or antigen-binding fragment thereof described herein is a monoclonal antibody or antigen-binding fragment thereof.
  • the anti-OX40 antigen-binding fragment described herein is Fab, Fab', F(ab')2, Fv, scFv, or sdAb.
  • the present invention provides an antibody-drug conjugate comprising the anti-OX40 antibody or antigen-binding fragment thereof as described herein and another therapeutic agent; preferably, the anti-OX40 antibody or antigen-binding fragment thereof The fragment and the additional therapeutic agent are connected by a linker.
  • the present invention provides a nucleic acid that encodes the anti-OX40 antibody or antigen-binding fragment thereof described herein.
  • nucleic acid described herein comprises:
  • the nucleic acid further comprises the heavy chain constant region nucleotide coding sequence shown in SEQ ID NO: 50 or 54 and/or the light chain constant region nucleotide coding sequence shown in SEQ ID NO: 52.
  • the present invention provides an expression vector comprising the nucleic acid described herein.
  • the present invention provides a host cell comprising the nucleic acid or expression vector described herein.
  • a method for producing the anti-OX40 antibody or antigen-binding fragment thereof described herein which comprises culturing the host cell described herein under conditions suitable for the expression of the antibody or antigen-binding fragment thereof, And recover the expressed antibody or its antigen-binding fragment from the culture medium.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the anti-OX40 antibody or antigen-binding fragment thereof described herein, or the antibody-drug conjugate described herein, or the nucleic acid described herein, or Said expression vector, and pharmaceutically acceptable carrier.
  • the anti-OX40 antibody or antigen-binding fragment thereof described herein, or the antibody-drug conjugate described herein, or the pharmaceutical composition described herein is used for the treatment of cancer.
  • the cancer is selected from squamous cell carcinoma (e.g., epithelial squamous cell carcinoma), lung cancer (including small cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous cell carcinoma of the lung), peritoneal cancer , Hepatocellular carcinoma, gastric cancer (including gastrointestinal cancer and gastrointestinal stromal cancer), pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, urethral cancer, liver tumor, breast cancer, colon cancer, Rectal cancer, colorectal cancer, endometrial or uterine cancer, salivary gland cancer, kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, liver cancer, anal cancer, penile cancer, melanoma, superficial spreading mel
  • the present invention provides a method for treating cancer, which comprises administering to a subject in need a therapeutically effective amount of the anti-OX40 antibody or antigen-binding fragment thereof described herein, or the antibody described herein- A drug conjugate, or a pharmaceutical composition as described herein, to treat the cancer.
  • the cancer is selected from squamous cell carcinoma (e.g., epithelial squamous cell carcinoma), lung cancer (including small cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous cell carcinoma of the lung), peritoneal cancer , Hepatocellular carcinoma, gastric cancer (including gastrointestinal cancer and gastrointestinal stromal cancer), pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, urethral cancer, liver tumor, breast cancer, colon cancer, Rectal cancer, colorectal cancer, endometrial or uterine cancer, salivary gland cancer, kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, liver cancer, anal cancer, penile cancer, melanoma, superficial spreading melanoma, Malignant freckle melanoma, acral melanoma, nodular melanoma, multiple myeloma and B-cell lymphoma, chronic
  • the present invention provides the anti-OX40 antibody or antigen-binding fragment thereof described herein, or the antibody-drug conjugate described herein, or the pharmaceutical composition described herein, in the preparation of a drug for the treatment of cancer
  • the cancer is selected from squamous cell carcinoma (e.g., epithelial squamous cell carcinoma), lung cancer (including small cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous cell carcinoma of the lung), peritoneal cancer , Hepatocellular carcinoma, gastric cancer (including gastrointestinal cancer and gastrointestinal stromal cancer), pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, urethral cancer, liver tumor, breast cancer, colon cancer, Rectal cancer, colorectal cancer, endometrial or uterine cancer, salivary gland cancer, kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, liver cancer, anal cancer, pen
  • squamous cell carcinoma
  • Inhibit Treg function e.g., inhibit the suppressive function of Treg
  • kill OX40-expressing cells e.g., cells expressing high levels of OX40
  • improve effector T cell function and/or improve memory T cell function reduce tumor immunity
  • enhance T cells Function and/or deplete cells expressing OX40 e.g., deplete cells expressing OX40.
  • the anti-OX40 antibody or antigen-binding fragment thereof described herein, or the antibody-drug conjugate described herein, or the pharmaceutical composition described herein is prepared for the treatment of one of the following or Uses in a variety of drugs: inhibit Treg function (e.g., inhibit the suppressive function of Treg), kill OX40-expressing cells (e.g., cells expressing high levels of OX40), improve effector T cell function and/or improve memory T cell function , Reduce tumor immunity, enhance T cell function and/or reduce cells expressing OX40.
  • Treg function e.g., inhibit the suppressive function of Treg
  • kill OX40-expressing cells e.g., cells expressing high levels of OX40
  • improve effector T cell function and/or improve memory T cell function e.g., reduce tumor immunity, enhance T cell function and/or reduce cells expressing OX40.
  • the present invention provides a pharmaceutical combination comprising the anti-OX40 antibody or antigen-binding fragment thereof described herein, or the antibody-drug conjugate described herein, or the pharmaceutical composition described herein, and One or more additional therapeutic agents.
  • the present invention provides a kit comprising the anti-OX40 antibody or antigen-binding fragment thereof described herein, or the antibody-drug conjugate described herein, or the pharmaceutical composition described herein, preferably It further includes a drug delivery device.
  • comparison window refers to a segment having at least about 20 (usually 30 to about 75 or 40 to about 50) contiguous positions, where the two sequences can be compared with those having Compare the reference sequences of the same number of consecutive positions.
  • the optimized alignment of the sequences used for comparison can be performed using a program in a suite of bioinformatics software (Inc., Madison, WI), using default parameters.
  • This program implements several comparison schemes described in the following references: Dayhoff, MO, 1978, A model of evolutionary change in proteins-Matrices for detecting distant relationships.
  • the "percentage of sequence identity” is determined by comparing two optimized aligned sequences on a comparison window with at least 20 positions, where the polynucleotide or polypeptide sequence in the comparison window Part of the reference sequence (which does not contain additions or deletions) compared to the optimized alignment of the two sequences may contain 20% or less, usually 5% to 15% or 10% to 12% of additions or deletions (ie, gaps). ). The percentage is calculated by determining the number of positions where the same nucleic acid base or amino acid residue appears in both sequences to generate the number of matching positions, and dividing the number of matching positions by the total number of positions in the reference sequence (Ie, window size) and multiply the result by 100 to produce a percentage of sequence identity.
  • the variant may also be substantially homologous to the natural gene or part or complement thereof.
  • These polynucleotide variants can hybridize to naturally-occurring DNA sequences encoding natural antibodies (or complementary sequences) under moderately stringent conditions.
  • Suitable “moderately stringent conditions” include pre-washing in a solution of 5X SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0); hybridization at 50°C to 65°C, 5X SSC overnight; then at 65°C Wash twice, lasting 20 minutes, and use 2X, 0.5X and 0.2X SSC containing 0.1% SDS for each wash.
  • highly stringent conditions or “highly stringent conditions” are those of the following: (1) Use low ionic strength and high temperature for cleaning, such as 0.015M sodium chloride/0.0015M citric acid at 50°C Sodium/0.1% sodium lauryl sulfate; (2) Use denaturants such as formamide during hybridization, for example, with 0.1% bovine serum albumin/0.1% sucrose/0.1% polyvinylpyrrole at pH 6.5 Pyridone/50mM sodium phosphate buffer and 50% (v/v) formamide of 750mM sodium chloride and 75mM sodium citrate at 42°C; or (3) 50% formamide, 5X SSC at 42°C (0.75M NaCl, 0.075M sodium citrate), 50mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5X Denhardt hybridization solution, sonicated salmon sperm DNA (50 ⁇ g/mL), 0.1% SDS and 10% Dextran sulfate, and washed
  • nucleotide sequences encoding the polypeptides as described herein. Some of these polynucleotides have minimal homology with the nucleotide sequence of any natural gene. However, the present invention specifically contemplates polynucleotides that are altered due to differences in codon usage. In addition, alleles of genes comprising the polynucleotide sequences provided herein are within the scope of the present invention. Alleles are endogenous genes that change due to one or more mutations of nucleotides, such as deletions, additions, and/or substitutions. The resulting mRNA and protein may (but need not) have altered structure or function. Alleles can be identified using standard techniques such as hybridization, amplification, and/or database sequence comparison.
  • the polynucleotide of the present invention can be obtained using chemical synthesis, recombinant methods or PCR.
  • the method of chemical polynucleotide synthesis is well known in the art and need not be described in detail herein. Those skilled in the art can use the sequences provided herein and commercially available DNA synthesizers to generate the desired DNA sequence.
  • the polynucleotides containing the desired sequence can be inserted into a suitable vector, and the vector can be further introduced into a suitable host cell for replication and amplification, as described herein further discussion.
  • the polynucleotide can be inserted into the host cell by any means known in the art. Cells are transformed by direct uptake, endocytosis, transfection, F-hybridization or electroporation to introduce exogenous polynucleotides. Once introduced, the exogenous polynucleotide can be maintained in the cell as a non-integrating vector (such as a plasmid) or integrated into the host cell gene.
  • the polynucleotide thus amplified can be isolated from the host cell by methods well known in the art. See, for example, Sambrook et al., 1989.
  • PCR allows for the replication of DNA sequences. .
  • RNA can be obtained by using isolated DNA in a suitable vector and inserting it into a suitable host cell. When the cell replicates and the DNA is transcribed into RNA, the RNA can then be isolated using methods known to those skilled in the art.
  • Suitable cloning and expression vectors can include various components such as promoters, enhancers, and other transcriptional regulatory sequences.
  • the vector can also be constructed to allow subsequent cloning of antibody variable domains into different vectors.
  • Suitable cloning vectors can be constructed according to standard techniques or can be selected from a large number of cloning vectors available in the art. Although the selected cloning vector can vary depending on the host cell to be used, useful cloning vectors will generally have the ability to self-replicate, can have a single target for specific restriction endonucleases, and/or can carry targets that can be used for selection. The gene containing the marker in the clone of the vector.
  • Suitable examples include plasmids and bacterial viruses, for example, pUC18, pUC19, Bluescript (e.g., pBS SK+) and derivatives thereof, mp18, mp19, pBR322, pMB9, ColE1, pCR1, RP4, phage DNA and shuttle vectors (such as pSA3 and pAT28).
  • Bluescript e.g., pBS SK+
  • shuttle vectors such as pSA3 and pAT28.
  • the expression vector is usually a replicable polynucleotide construct, which contains the polynucleotide according to the present invention. It implies that the expression vector must be replicable in the host cell, in the form of an episomal gene or as an integral part of chromosomal DNA.
  • Suitable expression vectors include (but are not limited to) plasmids and viral vectors, including adenovirus, adeno-associated virus, retrovirus, cosmid and the expression vector disclosed in PCT Publication No. WO 87/04462.
  • Vector components may generally include (but are not limited to) one or more of the following: a signal sequence; an origin of replication; one or more marker genes; suitable transcription control components (such as promoters, enhancers, and terminator).
  • suitable transcription control components such as promoters, enhancers, and terminator.
  • transcription control components such as ribosome binding sites, translation initiation sites, and termination codons, are also usually required.
  • the vector containing the polynucleotide of interest and/or the polynucleotide itself can be introduced into the host cell by any of a number of appropriate methods, including electroporation, the use of calcium chloride, rubidium chloride, calcium phosphate, Transfection of DEAE-dextran or other substances; microprojectile bombardment; lipofection; and infection (for example, where the vector is an infectious agent, such as a pox virus).
  • electroporation the use of calcium chloride, rubidium chloride, calcium phosphate, Transfection of DEAE-dextran or other substances; microprojectile bombardment; lipofection; and infection (for example, where the vector is an infectious agent, such as a pox virus).
  • the choice of introducing a vector or polynucleotide will generally depend on the characteristics of the host cell.
  • the antibodies of the present invention include human antibodies prepared, expressed, produced or isolated by recombinant methods, such as antibodies expressed using recombinant expression vectors transfected into host cells (further described in the following section II), isolated from recombinant combinatorial human antibody libraries Antibodies (further described in section III below), antibodies isolated from human immunoglobulin gene transgenic animals (e.g. mice) (see e.g. (Taylor, LD et al. (1992) Nucl. Acids Res. 20: 6287-6295) Or antibodies prepared, expressed, produced or isolated by any other method involving the splicing of human immunoglobulin gene sequences into other DNA sequences.
  • Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences ( See Kabat, EA, et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, USDepartment of Health and Human Services, NIH Publication No. 91-3242).
  • the antibody or antibody portion of the present invention can be prepared by recombinantly expressing immunoglobulin light chain genes and heavy chain genes in host cells.
  • a host cell is transfected with one or more recombinant expression vectors carrying DNA fragments encoding the immunoglobulin light and heavy chains of the antibody, so that the light and heavy chains are expressed in the host cell, Furthermore, it is preferably secreted into a medium in which the host cell is cultured, from which the antibody can be recovered. Standard recombinant DNA methodology is used to obtain antibody heavy chain genes and antibody light chain genes, introduce these genes into a recombinant expression vector, and then introduce the vector into host cells.
  • Antibodies or antigen-binding fragments thereof can be recombinantly produced using suitable host cells.
  • the nucleic acid encoding the antibody or antigen-binding fragment thereof can be cloned into an expression vector, which can then be introduced into host cells such as E. coli cells, yeast cells, insect cells, apes COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells , Wherein the cell does not additionally produce immunoglobulin to obtain antibody synthesis in the recombinant host cell.
  • host cells include E. coli cells, yeast cells, insect cells, apes COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells , Wherein the cell does not additionally produce immunoglobulin to obtain antibody synthesis in the recombinant host cell.
  • preferred host cells include CHO cells, human embryonic kidney (HEK) 293 cells, or Sp2.0 cells.
  • Antibody fragments can be produced by proteolysis or other degradation of full-length antibodies by recombinant methods or by chemical synthesis.
  • Polypeptide fragments of antibodies (especially shorter polypeptides of up to about 50 amino acids) can be conveniently prepared by chemical synthesis. Methods for the chemical synthesis of proteins and peptides are known in the art and are commercially available.
  • the antibody or antigen-binding fragment thereof of the present invention can be affinity matured.
  • affinity matured antibodies can be obtained by procedures known in the art (Marks et al., 1992, Bio/Technology, 10:779-783; Barbas et al., 1994, Proc Nat. Acad. Sci, USA 91: 3809- 3813; Schier et al., 1995, Gene, 169:147-155; Yelton et al., 1995, J. Immunol., 155:1994-2004; Jackson et al., 1995, J. Immunol., 154(7): 3310 -9; Hawkins et al., 1992, J. Mol. Biol., 226:889-896; and WO2004/058184).
  • amino acid sequence variants of the antibodies provided herein are encompassed.
  • Amino acid sequence variants of the antibody can be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions, and/or insertions and/or substitutions of residues within the amino acid sequence of the antibody. Any combination of deletion, insertion, and substitution can be made to obtain the final construct, as long as the final construct possesses the desired characteristics, for example, antigen binding.
  • antibody variants with one or more amino acid substitutions are provided.
  • Sites of interest for alternative mutagenesis include HVR and FR.
  • Conservative substitutions are shown in Table A under the heading of "preferred substitutions”. More substantial changes are provided in Table A under the heading of "exemplary substitutions” and are described further below with reference to amino acid side chain categories.
  • Amino acid substitutions can be introduced into the antibody of interest, and the product screened for the desired activity, such as retained/improved antigen binding, reduced immunogenicity, or improved ADCC or CDC.
  • amino acids can be grouped as follows:
  • Non-conservative substitutions would require replacing members of one of these categories with members of another category.
  • substitution variant involves replacing one or more hypervariable region residues of a parent antibody (e.g., a humanized or human antibody).
  • a parent antibody e.g., a humanized or human antibody.
  • the resulting variants selected for further research will have certain biological changes (e.g. improvement) (e.g. increased affinity, decreased immunogenicity) relative to the parent antibody and/or will substantially retain the parent antibody Some of the biological characteristics.
  • An exemplary alternative variant is an affinity matured antibody, which can be conveniently generated, for example, using phage display-based affinity maturation techniques such as those described herein.
  • one or more HVR residues are mutated, and the variant antibodies are displayed on phage and screened for specific biological activities (such as binding affinity).
  • HVR Changes (e.g., substitutions) can be made to HVR, for example to improve antibody affinity.
  • HVR "hot spots", residues encoded by codons that undergo mutations at a high frequency during the somatic maturation process (see, for example, Chowdhury, Methods Mol. Biol. 207:179-196 (2008)), and/or Such changes are made to residues in contact with the antigen, wherein the resulting variant VH or VL is tested for binding affinity.
  • Affinity maturation through the construction and reselection of secondary libraries has been described in, for example, Hoogenboom et al., in Methods in Molecular Biology 178: 1-37 (edited by O'Brien et al., Human Press, Totowa, NJ, (2001)).
  • diversity is introduced as a variable gene for maturation selection by multiple methods (e.g., error-prone PCR, strand shuffling, or oligonucleotide-directed mutagenesis). Then, create a secondary library. Then, the library is screened to identify any antibody variants with the desired affinity.
  • Another method of introducing diversity involves an HVR-directed method, in which several HVR residues (eg, 4-6 residues at a time) are randomized. Alanine scanning mutagenesis or modeling can be used, for example, to specifically identify HVR residues involved in antigen binding. In particular, CDR-H3 and CDR-L3 are often targeted.
  • substitutions, insertions, or deletions can occur within one or more HVRs, as long as such changes do not substantially reduce the ability of the antibody to bind antigen.
  • conservative changes e.g., conservative substitutions, as provided herein
  • HVR that do not substantially reduce binding affinity.
  • such changes can be outside of the antigen contact residues in the HVR.
  • each HVR is unchanged or contains no more than 1, 2, or 3 amino acid substitutions.
  • alanine scanning mutagenesis One method that can be used to identify residues or regions in antibodies that can be targeted for mutagenesis is called “alanine scanning mutagenesis", as described by Cunningham and Wells (1989) Science, 244:1081-1085.
  • residues or groups of target residues for example, charged residues such as arg, asp, his, lys, and glu
  • neutral or negatively charged amino acids for example, alanine Acid or polyalanine
  • Further substitutions can be introduced at amino acid positions that indicate functional sensitivity to the initial substitution.
  • the crystal structure of the antigen-antibody complex is used to identify the contact points between the antibody and the antigen.
  • contact residues and neighboring residues can be targeted or eliminated.
  • the variants can be screened to determine whether they contain the desired properties.
  • Amino acid sequence insertions include amino and/or carboxyl terminal fusions ranging from one residue to a polypeptide containing 100 or more residues in length, and intra-sequence insertions of single or multiple amino acid residues.
  • terminal insertions include antibodies with N-terminal methionyl residues.
  • Other insertional variants of antibody molecules include fusions of the N- or C-terminus of the antibody with an enzyme (for example, for ADEPT) or a polypeptide that extends the serum half-life of the antibody.
  • the antibodies provided herein are modified to increase or decrease the degree of glycosylation of the antibody.
  • the addition or deletion of glycosylation sites of the antibody can be conveniently achieved by changing the amino acid sequence so that one or more glycosylation sites are created or eliminated.
  • the carbohydrate to which it is attached can be changed.
  • Natural antibodies produced by mammalian cells usually contain branched, biantennary oligosaccharides, which are generally attached to Asn297 of the CH2 domain of the Fc region through an N linkage. See, for example, Wright et al., TIBTECH 15:26-32 (1997).
  • Oligosaccharides may include various carbohydrates, for example, mannose, N-acetylglucosamine (GlcNAc), galactose, and sialic acid, and fucose attached to GlcNAc in the "stem" of the biantennary oligosaccharide structure.
  • the oligosaccharides in the antibodies of the invention can be modified to create antibody variants with certain improved properties.
  • antibody variants are provided that have a carbohydrate structure lacking fucose attached (directly or indirectly) to the Fc region.
  • the amount of fucose in such antibodies can be 1% to 80%, 1% to 65%, 5% to 65%, or 20% to 40%.
  • Determine the amount of fucose by calculating the average amount of fucose in the sugar chain at Asn297 relative to the sum of all sugar structures (for example, complex, hybrid, and high-mannose structures) attached to Asn297, such as by Measured by MALDI-TOF mass spectrometry, for example, as described in WO 2008/077546.
  • Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of residues in the Fc region); however, Asn297 can also be located at about ⁇ 2% upstream or downstream of position 297 due to minor sequence variations in the antibody. 3 amino acids, that is, between the 294th and 300th positions. Such fucosylation variants may have improved ADCC function. See, for example, US Patent Publication No. US 2003/0157108 (Presta, L.); US 2004/0093621 (Kyowa Hakko Kogyo Co., Ltd).
  • Examples of publications involving "defucosylated” or “fucose deficient” antibody variants include: US 2003/0157108; WO 2000/61739; WO 2001/29246; US 2003/0115614; US 2002 /0164328; US 2004/0093621; US 2004/0132140; US 2004/0110704; US 2004/0110282; US 2004/0109865; WO 2003/085119; WO 2003/084570; WO 2005/035586; TO 2005/035778; TO2005/ 053742; TO2002/031140; Okazaki et al., J. Mol. Biol. 336:1239-1249 (2004); Yamane-Ohnuki et al., Biotech. Bioeng.
  • Examples of cell lines capable of producing defucosylated antibodies include LecI3CHO cells deficient in protein fucosylation (Ripka et al., Arch. Biochem. Biophys. 249:533-545 (1986); U.S. Patent Application No. US 2003 /0157108A1, Presta, L; and WO 2004/056312A1, Adams, etc.), and knockout cell lines, such as ⁇ -1,6-fucosyltransferase gene FUT8 knockout CHO cells (see, for example, Yamane-Ohnuki, etc., Biotech. Bioeng. 87: 614 (2004); Kanda, Y. et al., Biotechnol. Bioeng., 94(4): 680-688 (2006); and WO2003/085107).
  • antibody variants having bipartite oligosaccharides for example, the biantennary oligosaccharide attached to the Fc region of the antibody is bisected by GlcNAc.
  • Such antibody variants may have reduced fucosylation and/or improved ADCC function. Examples of such antibody variants are described in, for example, WO2003/011878 (Jean-Mairet et al.); US Patent No. 6,602,684 (Umana et al.); and US 2005/0123546 (Umana et al.).
  • antibody variants having at least one galactose residue in the oligosaccharide attached to the Fc region Such antibody variants may have improved CDC function.
  • Such antibody variants are described in, for example, WO 1997/30087 (Patel et al.); WO 1998/58964 (Raju, S.); and WO 1999/22764 (Raju, S.).
  • one or more amino acid modifications can be introduced into the Fc region of the antibodies provided herein, thereby generating Fc region variants.
  • the Fc region variant may comprise a human Fc region sequence (e.g., a human IgGl, IgG2, IgG3, or IgG4 Fc region) that contains amino acid modifications (e.g., substitutions) at one or more amino acid positions.
  • the present invention encompasses antibody variants possessing some but not all effector functions that make them desirable candidates for applications where the in vivo half-life of the antibody is important, and some Effector functions (such as complement and ADCC) are unnecessary or harmful.
  • In vitro and/or in vivo cytotoxicity assays can be performed to confirm the reduction/decrease of CDC and/or ADCC activity.
  • an Fc receptor (FcR) binding assay can be performed to ensure that the antibody lacks Fc ⁇ R binding (and therefore may lack ADCC activity), but retains FcRn binding ability.
  • the main cells that mediate ADCC, NK cells only express FcyRIII, while monocytes express FcyRI, FcyRII and FcyRIII.
  • FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol. 9:457-492 (1991).
  • a non-limiting example of an in vitro assay for assessing the ADCC activity of a molecule of interest is described in U.S. Patent No. 5,500,362 (see, for example, Hellstrom, I. et al., Proc. Nat'I Acad. Sci USA 83:7059-7063 (1986)) And Hellstrom, I etc., Proc. Nat'I Acad. Sci. USA 82: 1499-1502 (1985); 5, 821, 337 (see Bruggemann, M. et al., J. Exp. Med.
  • a non-radioactive assay method can be used (see, for example, the ACT I TM non-radioactive cytotoxicity assay for flow cytometry (Cell Technology, Inc. Mountain View, CA; and the CytoTox96 non-radioactive cytotoxicity assay (Promega, Madison) , WI)). Effector cells useful for such assays include peripheral blood mononuclear cells (PBMC) and natural killer (NK) cells.
  • PBMC peripheral blood mononuclear cells
  • NK natural killer cells.
  • the ADCC activity of the molecule of interest can be assessed in vivo, for example in animal models Such as those disclosed in Clynes et al., Proc Nat'I Acad Sci USA 95:652-656 (1998).
  • Clq binding assays can also be performed to confirm that antibodies cannot bind to Clq and therefore lack CDC activity. See, for example, WO 2006/ 029879 and the Clq and C3c binding ELISA in WO 2005/100402.
  • CDC assays can be implemented (see, for example, Gazzano-Santoro et al., J. Immunol. Methods 202:163 (1996); Cragg, MS. et al., Blood 101:1045-1052 (2003); and Cragg, M ⁇ S ⁇ and MJ Glennie, Blood 103:2738-2743 (2004)).
  • Antibodies with reduced effector function include those with substitutions of one or more of Fc region residues 238, 265, 269, 270, 297, 327, and 329 (US Patent No. 6,737,056).
  • Fc mutants include Fc mutants having substitutions at two or more of amino acid positions 265, 269, 270, 297, and 327, including so-called "DANA" in which residues 265 and 297 are substituted with alanine.
  • Fc mutant U.S. Patent No. 7,332,581.
  • antibody variants comprise an Fc region with one or more amino acid substitutions that improve ADCC, such as substitutions at positions 298, 333, and/or 334 (EU numbering of residues) in the Fc region.
  • changes are made to the Fc region that result in altered (ie, improved or reduced) Clq binding and/or complement dependent cytotoxicity (CDC), for example, as described in U.S. Patent No. 6,194,551 , WO 99/51642, and Idusogie et al., J.Tmmunol.164:4178-4184 (2000).
  • CDC complement dependent cytotoxicity
  • FcRn neonatal Fc receptor
  • the neonatal Fc receptor (FcRn) is responsible for the transfer of maternal IgG to the fetus (Guyer et al., J Immunol. 117:587 (1976) and Kim et al. J. Immunol. 24:249 (1994)).
  • Those antibodies comprise an Fc region with one or more substitutions therein that improve the binding of the Fc region to FcRn.
  • Such Fc variants include those that have substitutions at one or more of Fc region residues 238,256, 265,272,286,303,305,307,311,312,317,340,356,360,362,376,378,380,382,413,424 or 434, for example, substitution of Fc region residue 434 (US Patent No. 7,371,826).
  • cysteine for example, "thioMAb” in which one or more residues of the antibody are replaced with cysteine residues.
  • the substituted residue is present in an accessible site of the antibody.
  • the reactive thiol group is thus positioned at the accessible site of the antibody, and can be used to conjugate the antibody with other modules, such as drug modules or linker-drug modules, to Create immunoconjugates as described further herein.
  • cysteine can be substituted for any one or more of the following residues: V205 of the light chain (Kabat numbering); A118 of the heavy chain (EU numbering); and the Fc region of the heavy chain S400 (EU numbering method).
  • Cysteine engineered antibodies can be generated as described in, for example, US Patent No. 7,521,541.
  • the antibodies provided herein can be further modified to contain additional non-proteinaceous modules known in the art and readily available.
  • Modules suitable for antibody derivatization include, but are not limited to, water-soluble polymers.
  • water-soluble polymers include, but are not limited to, polyethylene glycol (PEG), ethylene glycol/propylene glycol copolymer, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinylpyrrolidone, poly-1 , 3-dioxolane, poly-1,3,6-trioxane, ethylene/maleic anhydride copolymer, polyamino acid (homopolymer or random copolymer), and dextran or poly( ⁇ -ethylene Pyrrolidone) polyethylene glycol, propylene glycol homopolymer, propylene oxide/ethylene oxide copolymer, polyoxyethylated polyol (such as glycerin), polyvinyl alcohol and mixtures thereof.
  • PEG polyethylene glycol
  • polyethylene glycol propionaldehyde Due to its stability in water, polyethylene glycol propionaldehyde may have advantages in production.
  • the polymer can be of any molecular weight and can be branched or unbranched.
  • the number of polymers attached to the antibody can vary, and if more than one polymer is attached, they can be the same or different molecules. Generally speaking, the number and/or type of polymers used for derivatization can be determined based on the following considerations, including but not limited to the specific properties or functions of the antibody to be improved, whether the antibody derivative will be used for treatment under specified conditions, etc. .
  • conjugates of antibodies and non-proteinaceous moieties that can be selectively heated by exposure to radiation are provided.
  • the non-proteinaceous module is carbon nanotubes (Kam et al., Proc. Natl. Acad. Sci. USA 102:11600-11605 (2005)).
  • the radiation can be of any wavelength, and includes, but is not limited to, a wavelength that does not damage ordinary cells, but heats the non-proteinaceous module to a temperature at which cells near the antibody-non-proteinaceous module are killed.
  • anti-OX40 antibodies provided herein can be identified, screened, or characterized by their physical/chemical properties and/or biological activities by a variety of assays known in the art.
  • the antibody of the present invention is tested for its antigen binding activity, for example, by a known method such as ELISA, Western blot, and the like. Methods known in the art can be used to determine OX40 binding, and exemplary methods are disclosed herein.
  • radioimmunoassay is used to measure binding.
  • An exemplary radioimmunoassay is illustrated.
  • the OX40 antibody was iodinated, and a competition reaction mixture containing a fixed concentration of iodinated antibody and a decreasing concentration of serially diluted unlabeled OX40 antibody was prepared.
  • Cells expressing OX40 for example, BT474 cells stably transfected with human OX40 are added to the reaction mixture.
  • the cells are washed to separate the free iodinated OX40 antibody from the OX40 antibody bound to the cells.
  • the level of bound iodinated OX40 antibody is determined, for example, by counting the radioactivity associated with the cells, and using standard methods to determine the binding affinity.
  • flow cytometry is used to assess the ability of OX40 antibodies to bind surface-expressed OX40 (e.g., on a subset of T cells).
  • Obtain peripheral leukocytes for example from humans, cynomolgus monkeys, rats or mice), and block the cells with serum.
  • the labeled OX40 antibody was added in serial dilutions, and T cells were also stained to identify T cell subsets (using methods known in the art).
  • surface plasmon resonance can be used to analyze OX40 binding. An exemplary surface plasmon resonance method is illustrated.
  • a competition assay can be used to identify antibodies that compete with any of the anti-OX40 antibodies disclosed herein for binding to OX40.
  • such competitive antibodies bind to the same epitope (e.g., linear or conformational epitope) as bound by any of the anti-OX40 antibodies disclosed herein. See Morris (1996) “Epitope Mapping Protocols", Methods in Molecular Biology vol. 66 (Humana Press, Totowa, NJ) for detailed exemplary methods for locating epitopes bound by antibodies.
  • a competitive assay is exemplified.
  • a first labeled antibody which binds to OX, such as mab 1A7.gr.1, mab 3C8.gr5
  • a second unlabeled antibody which are tested to compete with the first antibody
  • the ability to bind to OX40 incubate the immobilized OX40 in a solution.
  • the second antibody may be present in the supernatant of the hybridoma.
  • the immobilized OX40 was incubated in a solution containing the first labeled antibody but not the second unlabeled antibody.
  • an assay method for identifying anti-OX40 antibodies with biological activity can include, for example, binding to OX40 (for example, binding to human and/or cyno OX40), increasing OX40-mediated signal transduction (for example, increasing NFkB-mediated transcription), and reducing cells expressing human OX40 (for example, T cells) ,
  • OX40 for example, binding to human and/or cyno OX40
  • increasing OX40-mediated signal transduction for example, increasing NFkB-mediated transcription
  • reducing cells expressing human OX40 for example, T cells
  • T effector cell function e.g. CD4+ effector T cells
  • enhance T effector T cells e.g. by increasing effector T cell proliferation and/or increasing effector T cell cytokine production (e.g.
  • interferon gamma interferon gamma
  • enhance the function of memory T cells e.g. CD4+ memory T cells
  • cytokine production of memory T cells e.g. gamma interferon
  • inhibiting the function of regulatory T cells e.g. by Treg suppression that reduces effector T cell function (eg CD4+ effector T cell function)
  • binds to human effector cells e.g. CD4+ effector T cell function
  • the antibodies of the invention are tested for such biological activity.
  • T cells e.g., memory or effector T cells
  • peripheral white blood cells e.g., separated from human whole blood using Ficoll gradient centrifugation.
  • Methods known in the art can be used to isolate memory T cells (e.g., CD4+ memory T cells) or effector T cells (e.g., CD4+ Teff cells) from PBMC.
  • memory T cells e.g., CD4+ memory T cells
  • effector T cells e.g., CD4+ Teff cells
  • MiItenyi CD4+ Memory T Cell Isolation Kit or Miltenyi Naive CD4+ T Cell Isolation Kit can be used.
  • the isolated T cells are cultured in the presence of antigen-presenting cells (for example, irradiated L cells expressing CD32 and CD80), and activated by adding anti-CD3 antibodies in the presence or absence of OX40 agonistic antibodies.
  • antigen-presenting cells for example, irradiated L cells expressing CD32 and CD80
  • OX40 agonistic antibodies The effects of agonistic OX40 antibodies on T cell proliferation can be measured using methods known in the art. For example, you can use the CellTiterGlo kit (Promega), and read the results on a multi-label reader (PerkinElmer).
  • the effect of agonistic OX40 antibody on T cell function can also be determined by analyzing the cytokines produced by T cells.
  • interferon gamma production by CD4+ T cells is measured, for example, by measuring interferon gamma in the cell culture supernatant. Methods for measuring interferon gamma are well known in the art.
  • T cells are isolated from human whole blood using methods known in the art (e.g., memory T cells or naive T cells).
  • the purified CD4+ naive T cells are labeled (for example, with CFSE), and the purified Treg cells are labeled with different reagents.
  • the irradiated antigen-presenting cells for example, L cells expressing CD32 and CD80 are co-cultured with labeled and purified naive CD4+ T cells and purified Treg.
  • Co-cultures were activated with anti-CD3 antibodies and tested in the presence or absence of agonistic OMO antibodies. After a suitable time (for example, 6 days of co-cultivation), FACS analysis is used to track the level of CD4+ naive T cell proliferation by dye dilution in reduced marker staining (for example, reduced CFSE marker staining).
  • FACS analysis is used to track the level of CD4+ naive T cell proliferation by dye dilution in reduced marker staining (for example, reduced CFSE marker staining).
  • a transgenic cell expressing human OX40 and a reporter gene comprising an NFkB promoter fused to a reporter gene (such as ⁇ -luciferase)
  • a reporter gene such as ⁇ -luciferase
  • Phagocytosis can be measured, for example, by using monocyte-derived macrophages or U937 cells (a human histiocytic lymphoma cell line with the morphology and characteristics of mature macrophages).
  • Cells expressing OX40 are added to monocyte-derived macrophages or U937 cells in the presence or absence of anti-OX40 agonistic antibodies. After the cells are cultured for a suitable period of time, the percentage of cells double-stained with markers for 1) macrophages or U937 cells and 2) cells expressing OX40 is checked, and this is divided by the markers for cells expressing OX40 ( For example, the total number of GFP) cells is used to determine the percentage of phagocytosis. It can be analyzed by flow cytometry. In another embodiment, the analysis can be performed by fluorescence microscopy analysis.
  • ADCC can be measured, for example, using methods known in the art. Exemplary methods are described in the definition section.
  • the level of OX40 on OX40-expressing cells used for testing in the ADCC assay is characterized. The cells are stained with a detectably labeled anti-OX40 antibody (for example, PE-labeled), and then the fluorescence level is measured using flow cytometry, and the results are presented as median fluorescence intensity (MFI).
  • MFI median fluorescence intensity
  • ADCC can be analyzed by the CellTiter Glo assay kit, and cell viability/cytotoxicity can be determined by chemiluminescence.
  • the corresponding recombinant Fcy receptors can be used to measure the binding affinity of various antibodies to FcyRIA, FcyRIIA, FcyRIIB, and the two allotypes of FcyRIIIA (F158 and V158) in an ELISA-based ligand binding assay.
  • the purified human Fc ⁇ receptor was expressed as a fusion protein containing the extracellular domain of the receptor ⁇ chain linked to the C-terminal Gly/6xHis/glutathione S-transferase (GST) polypeptide tag.
  • GST Gly/6xHis/glutathione S-transferase
  • Fc ⁇ RIIA CD32A
  • Fc ⁇ RIIB CD32B
  • the two allotypes of Fe ⁇ RIIIA CD16
  • F-158 and V-158 you can use goat anti-human kappa chain F(ab ') 2 fragment (ICN Biomedical; Irvine, CA) cross-linked (approximate molar ratio 1:3 antibody: cross-linking F(ab')2) as a multimer test antibody.
  • the plate was coated with anti-GST antibody (Genentech) and blocked with bovine serum albumin (BSA).
  • Fc ⁇ receptor was added to the plate at 25ng/well and incubated at room temperature 1 hour. After washing the plate, a serial dilution of the test antibody is added as a polymer complex, and the plate is incubated at room temperature for 2 hours.
  • PBS phosphate buffered saline
  • ELx405 TM plate washer Biotek Instruments; Winooski, VT
  • HRP horseradish peroxidase
  • TMB tetramethylbenzidine
  • the plate is incubated at room temperature for 5-20 minutes to allow color development. Stop the reaction with IM H3PO4, and use the microplate reader ( 190, Molecular Devices; Sunnyvale, CA) measured absorbance at 450 nm.
  • a dose-response binding curve was generated by plotting the mean absorbance values from duplicate antibody dilutions against antibody concentration. The binding curve was fitted with a four-parameter equation using SoftMax 190 (Molecular Devices) to determine the effective antibody concentration (EC50) at which 50% of the maximum response from the bound Fc ⁇ receptor was detected.
  • SoftMax 190 Molecular Devices
  • the loss of membrane integrity shown by, for example, propidium iodide (PI), trypan blue or 7AAD uptake can be assessed relative to a control.
  • the PI uptake assay can be performed in the absence of complement and immune effector cells.
  • the cells expressing OX40 are incubated in a separate medium or a medium containing a suitable monoclonal antibody at a concentration of, for example, about 10 ⁇ g/ml.
  • the cells are incubated for a certain period of time (for example, 1 or 3 days). After each treatment, the cells were washed and aliquoted.
  • the cells are aliquoted into 35mm strainer-capped 12x75 tubes (1 ml per tube, 3 tubes per treatment group) to remove cell clumps. Then PI (10 ⁇ g/ml) was added to the tube. The samples can be analyzed using a FACSCAN TM flow cytometer and FACSCONVERT TM CellQuest software (Becton Dickinson).
  • Cells for use in any of the above in vitro assays include cells or cell lines that naturally express OX40 or have been engineered to express OX40. Such cells include activated T cells that naturally express OX40, Treg cells, and activated memory T cells. Such cells also include cell lines that express OX40 and cell lines that do not normally express OX40 but have been transfected with nucleic acid encoding OX40. Exemplary cell lines provided herein for use in any of the above in vitro assays include transgenic BT474 cells expressing human OX40 (a human breast cancer cell line).
  • immunoconjugates of the invention can be used to replace or supplement the anti-OX40 antibody to perform any of the aforementioned assays.
  • anti-OX40 antibodies and other therapeutic agents can be used to perform any of the aforementioned assays.
  • the antibody or antigen-binding fragment thereof of the present invention can be formulated into a pharmaceutical composition.
  • the pharmaceutical composition may further comprise pharmaceutically acceptable carriers, excipients and/or stabilizers (Remington: The Science and practice of Pharmacy, 20th Edition, 2000, Lippincott Williams and Wilkins, Ed. KE Hoover), and lyophilized Formulation or aqueous solution.
  • Acceptable carriers, excipients or stabilizers are non-toxic to the recipient at the dose and concentration, and may contain buffers such as phosphoric acid, citric acid and other organic acids; antioxidants, including ascorbic acid and methionine; Preservatives (such as octadecyl dimethyl benzyl ammonium chloride; hexahydroxy quaternary ammonium chloride; algaecide; benzonine chloride; phenolic alcohol, butanol or benzyl alcohol; alkyl p-hydroxybenzoate , Such as methyl p-hydroxybenzoate or propyl p-hydroxybenzoate; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) Polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulin; hydrophilic polymers, such as polyvinylpyrrolidone; amino acids, such
  • the antibody or antigen-binding fragment thereof of the present invention can be used for various therapeutic or diagnostic purposes.
  • the antibody or antigen-binding fragment thereof of the present invention can be used as an affinity purification agent (for example, for in vitro purification); as a diagnostic agent (for example, for detecting expression in specific cells, tissues, or serum).
  • Exemplary therapeutic uses of the antibodies of the present invention or antigen-binding fragments thereof include the treatment of cancer.
  • the antibody or antigen-binding fragment thereof of the present invention can also be used for prophylactic treatment.
  • the antibodies or antigen-binding fragments of the present invention can be administered to mammals, especially humans, by conventional techniques, such as intravenous (as a bolus injection or by continuous infusion over time), intramuscular, Intra-abdominal, intracerebral, subcutaneous, intra-articular, intra-synovial, intrathecal, oral, topical or inhalation.
  • the antibody or antigen-binding fragment thereof of the present invention can also be appropriately administered via intratumor, peritumor, intralesional, or perilesional routes.
  • the antibodies of the invention or antigen-binding fragments thereof are administered subcutaneously. In certain embodiments, the antibodies of the invention or antigen-binding fragments thereof are administered intravenously.
  • the pharmaceutical composition can be administered to subjects in need at a frequency that can vary with the severity of the disease.
  • the frequency may vary depending on the subject's disease susceptibility or tendency.
  • composition can be administered as a bolus injection or by continuous infusion to patients in need.
  • bolus administration of antibodies presented as Fab fragments can be administered from an amount of 0.0025 to 100 mg/kg body weight, 0.025 to 0.25 mg/kg, 0.010 to 0.10 mg/kg, or 0.10 to 0.50 mg/kg.
  • antibodies presented as Fab fragments can be 0.001 to 100 mg/kg body weight/min, 0.0125 to 1.25 mg/kg/min, 0.010 to 0.75 mg/kg/min, 0.010 to 1.0 mg/kg/min, or It is administered in an amount of 0.10 to 0.50 mg/kg/min for 1 to 24 hours, 1 to 12 hours, 2 to 12 hours, 6 to 12 hours, 2 to 8 hours, or 1 to 2 hours.
  • the dose can be from about 1 mg/kg to about 10 mg/kg, from about 2 mg/kg to about 10 mg/kg, from about 3 mg/kg to About 10 mg/kg, from about 4 mg/kg to about 10 mg/kg, from about 5 mg/kg to about 10 mg/kg, from about 1 mg/kg to about 20 mg/kg, from about 2 mg/kg to about 20 mg/kg, from About 3mg/kg to about 20mg/kg, from about 4mg/kg to about 20mg/kg, from about 5mg/kg to about 20mg/kg, about 1mg/kg or more, about 2mg/kg or more, about 3mg /kg or more, about 4mg/kg or more, about 5mg/kg or more, about 6mg/kg or more, about 7mg/kg or more, about 8mg/kg or more, about 9mg/kg Or more, about 10 mg/kg or more, about 11 mg
  • the composition can be administered to the patient via subcutaneous injection.
  • the dose of 1 to 100 mg of anti-OX40 antibody can be injected subcutaneously or intravenously at twice a week, once a week, once every two weeks, once every three weeks, once every four weeks, once every five weeks, once every six weeks, every It is administered to the patient at a frequency of once every seven weeks, once every eight weeks, once every nine weeks, once every ten weeks, twice a month, once a month, once every two months, or once every three months.
  • the half-life of an anti-OX40 antibody in humans is about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days. Days, about 14 days, about 15 days, about 16 days, about 17 days, about 18 days, about 19 days, about 20 days, about 21 days, about 22 days, about 23 days, about 24 days, about 25 days, About 26 days, about 27 days, about 28 days, about 29 days, about 30 days, from about 5 days to about 40 days, from about 5 days to about 35 days, from about 5 days to about 30 days, from about 5 days From about 10 days to about 40 days, from about 10 days to about 35 days, from about 10 days to about 30 days, from about 10 days to about 25 days, from about 15 days to about 40 days , From about 15 days to about 35 days, from about 15 days to about 40 days , From about 15 days to about 35 days, from about 15 days to about 30 days, or from about 15 days to about 25 days.
  • the pharmaceutical composition is administered subcutaneously or intravenously every 2 to 6 weeks at the following dose: from about 0.1 mg/kg to about 10 mg/kg, from about 0.5 mg/kg to about 10 mg/kg kg, from about 1 mg/kg to about 10 mg/kg, from about 1.5 mg/kg to about 10 mg/kg, from about 2 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 8 mg/kg, from about 0.5mg/kg to about 8mg/kg, from about 1mg/kg to about 8mg/kg, from about 1.5mg/kg to about 8mg/kg, from about 2mg/kg to about 8mg/kg, from about 0.1mg/kg To about 5mg/kg, from about 0.5mg/kg to about 5mg/kg, from about 1mg/kg to about 5mg/kg, from about 1.5mg/kg to about 5mg/kg, from about 2mg/kg to about 5mg/kg kg, about 0.5mg/kg
  • the pharmaceutical composition is administered subcutaneously or intravenously at a dose of about 2.0 mg/kg every 2 to 6 weeks. In certain embodiments, the pharmaceutical composition is administered subcutaneously or intravenously every 2 to 6 weeks at a dose of from about 2.0 mg/kg to about 10.0 mg/kg.
  • the pharmaceutical composition is administered subcutaneously every 2 weeks.
  • the antibodies or antigen-binding fragments thereof of the present invention can be used as monotherapy or in combination with other therapies to treat cancer.
  • an "antigen-binding fragment" of an antibody refers to a fragment of a full-length antibody that retains the ability to specifically bind to an antigen (preferably, has substantially the same binding affinity).
  • antigen-binding fragments include (i) Fab fragments, which are monovalent fragments composed of VL, VH, CL and CH1 domains; (ii) F(ab')2 fragments, which are contained in the hinge region connected by disulfide bonds (Iii) Fd fragment composed of VH and CH1 domains; (iv) Fv fragment composed of VL and VH domains of one arm of an antibody; (v) dAb fragment (Ward et al.
  • VL and VH isolated complementarity determining regions
  • dsFv disulfide-linked Fvs
  • anti- Id antibodies and intracellular antibodies.
  • VL and VH isolated complementarity determining regions
  • the synthetic linker allows it to be made into a single protein chain, in which the VL and VH regions Pair to form a monovalent molecule (called single-chain Fv (scFv)); see, for example, Bird et al.
  • Diabodies are bivalent bispecific antibodies in which the VH and VL domains are expressed on a single polypeptide chain, but the use of a linker that is too short to allow pairing between two chains on the same chain is used to force all The domain is paired with the complementary domain of the other chain and creates two antigen-binding sites (see, for example, Holliger et al., Proc. Natl. Acad. Sci. USA 90:6444-6448 (1993); Poljak et al., 1994, Structure 2:1121-1123).
  • variable domain refers to the variable region of the antibody light chain (VL) or the variable region of the antibody heavy chain (VH), alone or in combination.
  • VL variable region of the antibody light chain
  • VH variable region of the antibody heavy chain
  • CDR complementarity determining regions
  • FR framework regions
  • the residues in the variable domains are numbered according to Kabat, which is the numbering system of the heavy chain variable domain or the light chain variable domain used for the compilation of antibodies. See Kabat et al., Sequences of Proteins of Immunological Interest, 5th edition, Public Health Service, National Institutes of Health, Bethesda, MD. (1991)). Using this numbering system, the actual linear amino acid sequence may contain fewer or additional amino acids, which correspond to the shortening or insertion of the FR or CDR of the variable domain.
  • the Kabat numbering of residues can be determined by aligning the antibody's sequence to regions of homology with the "standard" Kabat numbering sequence.
  • Various algorithms for assigning Kabat numbers are available. Unless otherwise stated in this article, this article uses the algorithm implemented in Abysis (www.abysis.org) released in 2012 to assign Kabat numbers to variable regions.
  • CDR complementarity determining region
  • the "complementarity determining region” can be identified according to the definition of the aggregation, AbM, contact and/or configuration of both Kabat, Chothia, Kabat and Chothia well known in the art or any method of CDR determination. See, for example, Kabat et al., 1991, Sequences of Proteins of Immunological Interest, 5th edition (highly variable regions); Chothia et al., 1989, Nature 342:877-883 (structural loop structure).
  • the AbM definition of CDR is a compromise between Kabat and Chothia and the use of Oxford Molecular's AbM antibody modeling software.
  • the "contact" definition of CDR is based on the definition described in MacCallum et al., 1996, J. Mol.
  • CDR may refer to a CDR defined by any method (including a combination of methods) known in the art.
  • Antigenic determinant refers to the range or region in the antigen (Ag) where the antibody specifically binds, for example, the range or region containing the amino acid residues that interact with the antibody (Ab).
  • the epitope can be linear or non-linear (e.g., conformational).
  • the antibody or antigen-binding fragment thereof When the binding of corresponding antibodies or antigen-binding fragments thereof is mutually exclusive, the antibody or antigen-binding fragment thereof basically binds to the same epitope as another antibody or antigen-binding fragment thereof. That is, the binding of one antibody or antigen-binding fragment thereof excludes simultaneous or continuous binding of other antibodies or antigen-binding fragments thereof. If the antigen can be adapted to the simultaneous binding of two corresponding antibodies or antigen-binding fragments thereof, the epitope is considered to be unique or not substantially the same.
  • paratope is derived from the above definition of "antigenic determinant” by twisting the angle, and refers to a range or region involved in antigen binding in an antibody molecule, for example, a range or region containing residues that interact with the antigen .
  • Paratopes can be linear or conformational (such as discrete residues in CDRs).
  • the epitope/paratope of a given antibody/antigen binding pair can be defined and characterized at different levels of detail using various experimental and computational epitope positioning methods. Experimental methods include mutagenesis, X-ray crystallography, nuclear magnetic resonance (NMR) spectroscopy, hydrogen/deuterium exchange mass spectrometry (HX-MS) and various competitive binding methods. Because each method relies on the principle of uniqueness, the description of the epitope is closely related to the method by which the epitope has been determined. Therefore, the epitope/paratope of a given antibody/antigen pair will be defined differently depending on the localization method employed.
  • the epitope/paratope used for the interaction between the antibody (Ab) and the antigen (Ag) can be defined by the spatial coordinates of the atomic contact existing in the Ag-Ab interaction and the binding of the pair. The relative contribution of thermodynamics is defined by the information.
  • epitope/paratope residues can be characterized by defining the spatial coordinates of the atomic contact between Ag and Ab.
  • the epitope/paratopic residues can be defined by specific criteria, such as the distance between atoms in Ab and Ag (e.g., from a heavy atom of a homologous antibody and a heavy atom of the antigen equal to or less than about approx.
  • epitope/paratope residues are characterized by participating in hydrogen bonds with homologous antibodies/antigens, or with water molecules that are also hydrogen-bonded to homologous antibodies/antibodies (via water-mediated hydrogen bonding) Interaction.
  • epitope/paratopic residues are characterized by forming salt bridges with homologous antibody/antigen residues.
  • epitope/paratopic residues can be characterized by Residues that have non-zero changes in the masked surface area (BSA) due to the interaction with the homologous antibody/antigen.
  • BSA masked surface area
  • epitopes/paratopes can be characterized by functions, for example, by Competitive binding with other Abs.
  • the epitope/paratope can also be more generally defined as containing amino acid residues, where the substitution of another amino acid will change the characteristics of the interaction between Ab and Ag (for example, C Amino acid scan).
  • epitopes Since the description and definition of epitopes depend on the epitope mapping method used and the facts obtained at different levels of detail, it is inferred that the comparison of epitopes of different Abs on the same Ag can be similar at different levels of detail To proceed. For example, if it is described at the amino acid level, such as an epitope determined from X-ray structure, if it contains the same amino acid residue group, it is considered to be the same. If the binding of the corresponding antibodies is mutually exclusive, that is, the binding of one antibody excludes the simultaneous or sequential binding of other antibodies, then the epitopes characterized by competitive binding are considered to be overlapping; and if the antigen can accommodate two corresponding If antibodies bind at the same time, it is considered that the epitopes are distinct (unique).
  • the epitope and paratope of a given antibody/antigen pair can be identified by routine methods. For example, the general location of the epitope can be determined by assessing the ability of the antibody to bind to different fragments or variant polypeptides, as described more fully previously herein.
  • the specific residues in OX40 that can be contacted with the specific residues in the antibody can also be determined using routine methods.
  • the antibody/antigen complex can be crystallized. The crystal structure can be determined and used to identify specific sites of interaction between the antibody and the antigen.
  • specific binding is a term well known in the art, and methods for determining these specific bindings are also well known in the art. If a molecule reacts or binds to a specific cell or substance more frequently, faster, has a longer duration, and/or has greater affinity than the molecule reacts or binds to a replacement cell or substance, it is considered that the molecule exhibits "specific binding". If the antibody or antigen-binding fragment thereof binds to the target with greater affinity, binding, easier and/or longer duration than other substances, the antibody or antigen-binding fragment thereof "specifically binds" to the target.
  • an antibody or antigen-binding fragment thereof that specifically binds to OX40 is that the antibody binds to its homologous antigen with greater affinity, binding, easier and/or longer duration than binding to other antigens.
  • an anti-OX40 antibody can specifically bind to human OX40 in a sample, but does not substantially recognize or bind to other molecules in the sample.
  • an antibody or antigen-binding fragment thereof that specifically binds to a first target may or may not specifically bind to a second target. Therefore, "specific binding” does not necessarily require (although it may include) exclusive binding. Usually, but not necessarily, the reference to "binding" means specific binding.
  • Various analysis modes can be used to select antibodies or antigen-binding fragments thereof that specifically bind to the molecule of interest.
  • solid-phase ELISA immunoassay, immunoprecipitation, Biacore TM (GE Healthcare), KinExA, fluorescence activated cell sorting (FACS), Octet TM (FortéBio, Inc.) and Western blot analysis can be used Recognize antibodies or antigen-binding fragments thereof that specifically bind to an antigen.
  • the specific binding will be at least twice the background signal or noise, more usually at least 10 times the background, at least 50 times the background, at least 100 times the background, at least 500 times the background, at least 1000 times the background, or background At least 10,000 times of that.
  • Antibody binding specificities can be K D K D value and comparing the value with K D values to a control antibody known not to bind to OX40 evaluated by measuring the specific binding between the antibody and OX40.
  • K D is about ⁇ 10 -5 M or less, it is considered that the antibody "specifically" binds to the antigen.
  • the antibody or its antigen-binding fragment When compared to an antibody or its antigen-binding fragment that binds to other antigens, the antibody or its antigen-binding fragment does not bind to an antigen with greater affinity, binding, easier and/or longer duration, the antibody or its antigen-binding fragment
  • the antigen-binding fragment "substantially does not bind" to the antigen.
  • the combination will not be more than twice the background signal or noise.
  • it combines with the K D of 1 ⁇ 10 -4 M or more, 1 ⁇ 10 -3 M or more, 1 ⁇ 10 -2 M or more, or 1 ⁇ 10 -1 M or more. antigen.
  • competitive as used herein with respect to antibodies means that the binding of a first antibody or antigen-binding portion thereof to an antigen reduces subsequent binding of a second antibody or antigen-binding portion thereof to the same antigen.
  • the binding of the first antibody produces steric hindrance, conformational change, or binding to a common epitope (or part thereof), so that the binding of the second antibody to the same antigen is reduced.
  • Standard competitive binding analysis can be used to determine whether two antibodies compete with each other.
  • Biacore technology which may use surface plasmon resonance (SPR) technology, usually using a biosensor system (such as a system) to measure the degree of interaction.
  • SPR can be used in an in vitro competitive binding inhibition assay to determine the ability of an antibody to inhibit the binding of a second antibody.
  • Another analysis for measuring antibody competition uses an ELISA-based method.
  • a high-throughput method for "grading" antibodies based on antibody competition is described in WO2003/48731. If one antibody or antigen-binding fragment thereof reduces the binding of another antibody or antigen-binding fragment to OX40, there is competition. For example, sequential binding competition analysis can be used, and different antibodies can be added sequentially.
  • the first antibody can be added to achieve near-saturated binding. Then, the second antibody is added. If the binding of the second antibody to OX40 cannot be detected or compared to the parallel analysis in the absence of the first antibody (where the value can be set to 100%), it is significantly reduced (e.g., at least about 10%, at least about 10%). 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%, or at least about 90% reduction), then the two antibodies are considered to be competing with each other .
  • K i Concentration at which 50% inhibition occurs is referred to as K i.
  • this K i is equal to K D.
  • the binding affinities associated with different molecular interactions can be compared by comparing the K D values for individual antibody/antigen complexes.
  • the K D value for antibodies or other binding partners can be determined using methods established in the art.
  • Fc fusion protein is a protein in which one or more polypeptides are operably linked to an Fc polypeptide.
  • Fc fusion combines the Fc region of an immunoglobulin and a fusion partner.
  • the "Fc region” can be a native sequence Fc region or a variant Fc region.
  • the human IgG heavy chain Fc region is usually defined as an amino acid residue at position Cys226 or extending from Pro230 to its carboxyl terminus.
  • the numbering of residues in the Fc region is the numbering with the EU index, as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th edition, Public Health Service, National Institutes of Health, Bethesda, Md., 1991.
  • the Fc region of immunoglobulin usually contains two constant domains (CH 2 and CH 3 ). As known in the art, the Fc region can exist in dimer or monomer form.
  • terapéuticaally effective amount means an amount of an anti-OX40 antibody or antigen-binding fragment thereof, or a combination comprising the antibody or antigen-binding fragment thereof, sufficient to achieve the intended purpose.
  • the precise amount will depend on many factors, including (but not limited to) the components and physical characteristics of the therapeutic composition, the expected patient population, individual patient precautions, etc., and can be determined by those skilled in the art.
  • treatment includes prophylactic and/or therapeutic treatment. If administered before the clinical manifestation of the disease, disorder, or condition, the treatment is considered prophylactic.
  • Therapeutic treatment includes, for example, reducing or attenuating the severity of or shortening the length of the disease, disorder, or condition.
  • any of the anti-human OX40 antibodies provided herein can be used in treatment methods.
  • an anti-human OX40 agonistic antibody is provided for use as a medicine.
  • anti-human OX40 agonistic antibodies are provided for use in the treatment of cancer.
  • anti-human OX40 agonistic antibodies are provided for use in methods of treatment.
  • an anti-human OX40 agonist antibody is provided for use in a method of treating an individual with cancer, comprising administering an effective amount of the anti-human OX40 agonist antibody to the individual. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, such as described below.
  • an anti-human OX40 agonistic antibody for use in enhancing immune function (for example, by up-regulating a cell-mediated immune response) in an individual with cancer, including administering an effective amount of the anti-human OX40 to the individual Agonistic antibodies.
  • an anti-human OX40 agonistic antibody for enhancing T cell function in an individual with cancer including administering an effective amount of the anti-human OX40 agonistic antibody to the individual.
  • an anti-human OX40 agonistic antibody which is used to deplete human OX40-expressing cells (for example, OX40-expressing T cells, such as OX40-expressing Treg), including administering an effective amount of the anti-human OX40 to the individual Agonistic antibodies.
  • the abatement is performed by ADCC.
  • depletion is by phagocytosis.
  • an anti-human OX40 agonistic antibody which is used to treat individuals with tumor immunity.
  • anti-human OX40 agonistic antibodies are provided for use in the treatment of infections (eg, bacterial or viral or other pathogen infections).
  • infections eg, bacterial or viral or other pathogen infections.
  • the present invention provides an anti-human OX40 agonist antibody for use in a method of treating an individual with an infection, comprising administering an effective amount of the anti-human OX40 agonist antibody to the individual.
  • the infection is a viral and/or bacterial infection.
  • the infection is a pathogen infection.
  • the present invention provides the use of anti-OX40 antibody to manufacture or prepare medicine.
  • the drug is used to treat cancer.
  • the medicament is used in a method of treating cancer, which comprises administering an effective amount of the medicament to an individual with cancer. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, such as described below.
  • the drug is used to enhance immune function in an individual with cancer (for example, by up-regulating a cell-mediated immune response), which includes administering an effective amount of the drug to the individual.
  • the drug is used to enhance T cell function in an individual with cancer, which includes administering an effective amount of the drug to the individual.
  • the T cell dysfunctional disorder is cancer.
  • the drug is used to deplete human OX40 expressing cells (eg, high OX40 expressing cells, such as OX40 expressing T cells), which includes administering an effective amount of the drug to the individual.
  • the abatement is performed by ADCC.
  • depletion is by phagocytosis.
  • the drug is used to treat individuals with tumor immunity.
  • medicaments are provided for use in the treatment of infections (e.g., bacterial or viral or other pathogen infections).
  • the method of treating an individual with an infection by the medicament comprises administering an effective amount of the medicament to the individual.
  • the infection is a viral and/or bacterial infection.
  • the infection is a pathogen infection.
  • the present invention provides methods for treating cancer.
  • the method includes administering an effective amount of an anti-OX40 antibody to an individual with such cancer.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, such as described below.
  • the "individual" according to any of the above embodiments may be a human.
  • a method for enhancing T cell function in an individual with cancer comprising administering to the individual an effective amount of the anti-human OX40 agonist antibody.
  • a method for depleting cells expressing human OX40 for example, cells expressing high levels of OX40, such as T cells expressing OX40
  • the abatement is performed by ADCC.
  • depletion is by phagocytosis.
  • an anti-human OX40 agonistic antibody which is used to treat individuals with tumor immunity.
  • examples of cancer further include, but are not limited to, B-cell lymphoma (including low-grade/follicular non-Hodgkin’s lymphoma (NHL), small lymphocytic (SL) NHU intermediate/follicular NHL , Intermediate diffuse NHL, high-grade immunoblastic NHL, high-grade lymphoblastic NHL, high-grade small nonnucleoblastic NHL, bulky disease NHL, mantle cell lymphoma, AIDS-related lymphoma, and Val Denstrom's (Waldenstrom) macroglobulinemia), chronic lymphocytic leukemia (CLL), acute lymphoblastic leukemia (ALL), hairy cell leukemia, chronic myeloblastic leukemia, and post-transplant lymph Proliferative disorders (PTLD), and abnormal blood vessel proliferation associated with phakomatoses, edema (such as those associated with brain tumors), B cell proliferative disorders, and Meigs syndrome.
  • B-cell lymphoma including low-grade/follicular non
  • More specific examples include, but are not limited to, relapsed or refractory NHL, front-line low-grade NHL, stage III/IV NHL, chemotherapy-resistant NHL, precursor B lymphoblastic leukemia and/or lymphoma, small lymphoma Cellular lymphoma, B-cell chronic lymphocytic leukemia and/or prolymphocytic leukemia and/or small lymphocytic lymphoma, B-cell prelymphocytic lymphoma, immunocytoma and/or lymphoplasmacytic ( lymphoplasmacytic lymphoma, lymphoplasmacytic lymphoma, marginal zone B-cell lymphoma, splenic marginal zone lymphoma, extranodal marginal zone_MALT lymphoma, nodal marginal zone lymphoma, hair Cellular Leukemia, Cytocytoma and/or Plasma Cell Myeloma, Low Grade/Follicular Lymphoma, Intermediate/Follicular NHL, Mantle Cell Lymphoma, Folli
  • examples of cancer further include, but are not limited to, B-cell proliferative disorders, which further include, but are not limited to, lymphoma (eg, B-cell non-Hodgkin's lymphoma (NHL)) and lymphocytic leukemia.
  • lymphoma eg, B-cell non-Hodgkin's lymphoma (NHL)
  • NHL lymphocytic leukemia
  • lymphomas and lymphocytic leukemias include, for example, a) follicular lymphoma, b) small Non-Cleaved Cell Lymphoma/Burkitt’s lymphoma (including endemic Burkitt’s lymphoma, sporadic Burkitt’s lymphoma and non-Burkitt’s lymphoma), c) marginal zone lymphoma (including extranodal marginal zone B-cell lymphoma (mucosa-associated lymphoid tissue lymphoma, MALT) ), nodal marginal zone B-cell lymphoma and splenic marginal zone lymphoma), d) mantle cell lymphoma (MCL), e) large cell lymphoma (including B-cell diffuse large cell lymphoma (DLCL), diffuse mixed Cell lymphoma, immunoblastic lymphoma, primary mediastinal B-cell lymphoma, angiocentric lymphoma-pulmonary B-cell lympho
  • the cancer is a B cell proliferative disorder.
  • the B-cell proliferative disorder is lymphoma, non-Hodgkin's lymphoma (NHL), aggressive NHL, recurrent aggressive NHL, recurrent painless NHL, refractory NHL , Refractory painless NHL, chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma, leukemia, hairy cell leukemia (HCL), acute lymphocytic leukemia (ALL), or mantle cell lymphoma.
  • NHL such as painless NHL and/or aggressive NHL.
  • the B-cell proliferative disorder is painless follicular lymphoma or diffuse large B-cell lymphoma.
  • the present invention provides a pharmaceutical formulation comprising any anti-OX40 antibody provided herein, for example for use in any of the above-mentioned treatment methods.
  • the pharmaceutical formulation comprises any anti-OX40 antibody provided herein and a pharmaceutically acceptable carrier.
  • the pharmaceutical formulation comprises any anti-OX40 antibody provided herein and at least one additional therapeutic agent, such as described below.
  • the anti-human OX40 agonist antibody kills OX40-expressing cells (for example, cells expressing high levels of OX40) by inhibiting Treg function (for example, inhibiting the suppressive function of Treg), and increasing Effector T cell function and/or improve memory T cell function to suppress tumor immunity.
  • the anti-human OX40 agonist antibody kills OX40-expressing cells (for example, cells expressing high levels of OX40) by inhibiting Treg function (for example, inhibiting the suppressive function of Treg), and increasing Effector T cell function and/or improve memory T cell function to treat cancer.
  • the anti-human OX40 agonist antibody kills OX40-expressing cells (for example, cells expressing high levels of OX40) by inhibiting Treg function (for example, inhibiting the suppressive function of Treg), and increasing Effector T cell function and/or improve memory T cell function to enhance immune function.
  • the anti-human OX40 agonist antibody kills OX40-expressing cells (for example, cells expressing high levels of OX40) by inhibiting Treg function (for example, inhibiting the suppressive function of Treg), and increasing Effector T cell function and/or improve memory T cell function to enhance T cell function.
  • the anti-human OX40 agonistic antibody is a subtractive anti-human OX40 agonistic antibody.
  • the anti-human OX40 agonist antibody treatment results in cell depletion (e.g., depletion of cells expressing OX40, such as depletion of cells expressing high levels of OX40).
  • the abatement is performed by ADCC.
  • depletion is by phagocytosis.
  • the anti-human OX40 agonist antibody may, for example, inhibit effector and/or memory T cell function (in some embodiments, effector T cell function) relative to the Treg function prior to administration of the OX40 agonist antibody. And/or memory T cell proliferation and/or cytokine secretion) Treg suppression to inhibit Treg function.
  • the anti-human OX40 agonist antibody increases effector T cell proliferation relative to effector T cell proliferation prior to administration of the OX40 agonist antibody.
  • the anti-human OX40 agonist antibody increases memory T cell proliferation relative to the memory T cell proliferation prior to administration of the OX40 agonist antibody.
  • the anti-human OX40 agonist antibody increases effector T cell cytokine production (eg, gamma-interferon production) relative to effector T cell cytokine production prior to administration of the OX40 agonist antibody. In some embodiments of any method, the anti-human OX40 agonist antibody increases memory T cell cytokine production (eg, ⁇ -interferon production) relative to memory T cell cytokine production prior to administration of the OX40 agonist antibody.
  • the anti-human OX40 agonist antibody increases CD4+ effector T cell proliferation and/or CD8+ relative to CD4+ effector T cell proliferation and/or CD8+ effector T cell proliferation prior to administration of the OX40 agonist antibody Effector T cell proliferation. In some embodiments of any method, the anti-human OX40 agonist antibody increases memory T cell proliferation (e.g., CD4+ memory T cell proliferation) relative to memory T cell proliferation prior to administration of the OX40 agonist antibody.
  • memory T cell proliferation e.g., CD4+ memory T cell proliferation
  • the CD4+ effector T cells in the individual have enhanced proliferation, cytokine secretion and/or lysis. Cell activity.
  • the number of CD4+ effector T cells is increased relative to before administration of the anti-human OX40 agonist antibody. In some embodiments, CD4+ effector T cell cytokine secretion is increased relative to before administration of the anti-human OX40 agonist antibody. In some embodiments of any method, the CD8+ effector T cells in the individual have enhanced proliferation, cytokine secretion, and/or lytic activity relative to prior to administration of the anti-human OX40 agonistic antibody. In some embodiments, the number of CD8+ effector T cells is increased relative to before administration of the anti-human OX40 agonistic antibody. In some embodiments, CD8+ effector T cell cytokine secretion is increased relative to before administration of the anti-human OX40 agonist antibody.
  • the anti-human OX40 agonist antibody binds to human effector cells, for example, to Fc ⁇ R expressed by human effector cells.
  • the human effector cell performs ADCC effector function.
  • the human effector cell performs phagocytic effector function.
  • the anti-human OX40 agonist antibody comprising a variant IgG1Fc polypeptide (which contains a mutation that eliminates binding to human effector cells, such as the DANA or N297G mutation) has a portion relative to the IgG1Fc portion containing the native sequence
  • the anti-human OX40 agonistic antibody reduces the activity (eg CD4+ effector T cell function, such as proliferation).
  • an anti-human OX40 agonist antibody comprising a variant IgG1Fc polypeptide (which contains a mutation that eliminates binding to human effector cells, such as the DANA or N297G mutation) does not possess substantial activity (e.g., CD4+ effector T cell function, Such as proliferation).
  • the anti-human OX40 agonistic antibody function requires antibody cross-linking.
  • the function is to stimulate the proliferation of CD4+ effector T cells.
  • antibody cross-linking is determined by providing an anti-human OX40 agonistic antibody adhered to a solid surface (e.g., cell culture plate).
  • antibody cross-linking is determined by introducing mutations (such as DANA or N297S mutations) in the IgG1 Fc portion of the antibody and testing the function of the mutant antibody.
  • the memory T cells in the individual have enhanced proliferation and/or cytokine secretion relative to prior to administration of the anti-human OX40 agonistic antibody.
  • the number of memory T cells is increased relative to before administration of the anti-human OX40 agonist antibody.
  • memory T cell cytokine secretion (level) is increased relative to before administration of the anti-human OX40 agonist antibody.
  • Tregs in the individual have reduced effector T cell function (e.g., proliferation and/or cytokine secretion) inhibition relative to prior to administration of the anti-human OX40 agonistic antibody.
  • the number of effector T cells is increased relative to before administration of the anti-human OX40 agonistic antibody. In some embodiments, effector T cell cytokine secretion (level) is increased relative to before administration of the anti-human OX40 agonist antibody.
  • the number of (infiltrating) CD4+ effector T cells in the tumor is relative to the administration of the anti-human OX40 Agonistic antibodies were previously elevated.
  • the number of (infiltrating) CD4+ effector T cells in the tumor expressing interferon-gamma e.g., total CD4+ cells expressing interferon-gamma, or, for example, total CD4+ cells
  • the percentage of CD4+ cells expressing ⁇ -interferon was increased relative to before administration of anti-human OX40 agonistic antibody.
  • the number of (infiltrating) CD8+ effector T cells in the tumor is relative to administration of anti-human OX40 agonism The antibody was previously elevated.
  • the number of (infiltrating) CD8+ effector T cells in a tumor expressing interferon-gamma is relative to Elevated before administration of anti-human OX40 agonistic antibody.
  • the number of (infiltrating) Tregs within the tumor is reduced relative to before administration of the anti-human OX40 agonistic antibody.
  • the administration of the anti-human OX40 agonistic antibody is combined with the administration of the tumor antigen.
  • the tumor antigen comprises a protein.
  • the tumor antigen comprises nucleic acid.
  • the tumor antigen is a tumor cell.
  • the cancer displays human effector cells (e.g., is infiltrated by human effector cells).
  • the methods used to detect human effector cells are well known in the art and include, for example, by IHC.
  • the cancer displays high levels of human effector cells.
  • the human effector cells are one or more of NK cells, macrophages, and monocytes.
  • the cancer is any cancer described herein.
  • the cancer is non-small cell lung cancer (NSCLC), glioblastoma, neuroblastoma, melanoma, breast cancer (e.g. triple negative breast cancer), gastric cancer, colorectal cancer (CRC), Or hepatocellular carcinoma.
  • the cancer displays FcR-expressing cells (e.g., is infiltrated by FcR-expressing cells).
  • Methods for detecting FcR are well known in the art and include, for example, by IHC.
  • the cancer displays high levels of FcR-expressing cells.
  • FcR is FcyR.
  • the FcR is an activating Fc ⁇ R.
  • the cancer is non-small cell lung cancer (NSCLC), glioblastoma, neuroblastoma, melanoma, breast cancer (e.g. triple negative breast cancer), gastric cancer, colorectal cancer (CRC), Or hepatocellular carcinoma.
  • NSCLC non-small cell lung cancer
  • glioblastoma glioblastoma
  • neuroblastoma melanoma
  • breast cancer e.g. triple negative breast cancer
  • CRC colorectal cancer
  • the "individual” according to any of the above embodiments is preferably a human.
  • the antibodies of the present invention can be used alone or in combination with other agents in therapy.
  • the antibody of the invention can be co-administered with at least one other therapeutic agent.
  • Such combination therapies documented above encompass combined administration (wherein two or more therapeutic agents are contained in the same formulation or separate formulations), and separate administration, in which case the administration of another therapeutic agent And/or the administration of the antibody of the present invention occurs before, at the same time, and/or after the agent.
  • the administration of the anti-OX40 antibody and the administration of the other therapeutic agent are within about one month, or within about one, two or three weeks, or within about 1, 2, 3, 4, 5, or 6 days of each other occur.
  • the antibodies of the present invention can also be used in combination with radiation therapy.
  • the anti-human OX40 agonist antibody can be administered in combination with chemotherapy or chemotherapeutic agents. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with radiotherapy or radiotherapy agents. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with a targeted therapy or a targeted therapeutic agent. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with immunotherapy or immunotherapeutic agents, such as monoclonal antibodies.
  • the anti-human OX40 agonist antibody can be combined with PARP inhibitors (eg Olaparanib, Rucaparib, Niraparib, Cediranib, BMN673, Veliparib), Trabectedin, nab-paclitaxel (albumin-bound paclitaxel, ABRAXANE), Trebananib , Pazopanib, Cediranib, Palbociclib, everolimus, fluorouracil (e.g.
  • the anti-human OX40 agonist antibody can be administered in combination with a PD-1 axis binding antagonist.
  • PD-1 axis binding antagonists include but are not limited to PD-1 binding antagonists, PD-L1 binding antagonists and PD-L2 binding antagonists.
  • Alternative names for "PD-1” include CD279 and SLEB2.
  • Alternative names for "PD-L1” include B7-H1, B7-4, CD274 and B7-H.
  • Alternative names for "PD-L2" include B7-DC, Btdc, and CD273.
  • PD-1, PD-L1, and PD-L2 are human PD-1, PD-L1 and PD-L2.
  • the PD-1 binding antagonist is a molecule that inhibits the binding of PD-1 to its ligand binding partner.
  • the PD-1 ligand binding partner is PD-L1 and/or PD-L2.
  • the PD-L1 binding antagonist is a molecule that inhibits the binding of PD-L1 to its binding partner.
  • the PD-L1 binding partner is PD-1 and/or B7-1.
  • the PD-L2 binding antagonist is a molecule that inhibits the binding of PD-L2 to its binding partner.
  • the PD-L2 binding partner is PD-1.
  • the antagonist can be an antibody, an antigen-binding fragment thereof, an immunoadhesin, a fusion protein, or an oligopeptide.
  • the PD-1 binding antagonist is an anti-PD-1 antibody (e.g., a human antibody, a humanized antibody, or a chimeric antibody).
  • the anti-PD-1 antibody is selected from the group consisting of MDX-1106 (nivolumab, OPDIVO), Merck 3475 (MK-3475, pembrolizumab, KEYTRUDAWPCT-011 (Pidilizumab).
  • PD-1 The binding antagonist is an immunoadhesin (e.g., an immunoadhesin comprising an extracellular or PD-1 binding portion of PD-L1 or PD-L2 fused to a constant region (e.g., the Fc region of an immunoglobulin sequence)).
  • the PD-1 binding antagonist is AMP-224.
  • the PD-L1 binding antagonist is an anti-PD-L1 antibody.
  • the anti-PD-L1 binding antagonist is selected From the following group: YW243.55.S70, MPDL3280A, MEDI4736 and MDX-1105.
  • MDX-1105 also known as BMS-936559
  • Antibody YW243.55.S70 is WO Anti-PD-L1 described in 2010/077634 A1.
  • MDX-1106 also known as MDX-1106-04, ONO-4538, BMS-936558 or nivolumab
  • Merck 3475 is also known as MK-3475, SCH-900475 or pembrolizumab, is the anti-PD-1 antibody described in WO2009/114335.
  • CT-011 also known as hBAT, hBAT-1 or pidil izumab
  • AMP-224 also known as B7-DCIg
  • the anti-PD-1 antibody is MDX-1106.
  • Alternative names for "MDX-1106” include MDX-1106-04, ON0-4538, BMS-936558 or nivoIumab.
  • the anti-PD-1 antibody is nivolumab (CAS Registry Number: 946414- 94-4).
  • the anti-human OX40 agonist antibody can be administered in combination with an agonist directed against an activating costimulatory molecule.
  • the activating costimulatory molecule may include CD40, CD226, CD28, GITR, CD137, CD27, HVEM, or CD127.
  • the agonist against the activating costimulatory molecule is an agonist antibody that binds CD40, CD226, CD28, OX40, GITR, CD137, CD27, HVEM, or CD127.
  • the anti-human OX40 agonist antibody can be administered in combination with an antagonist against an inhibitory costimulatory molecule.
  • inhibitory costimulatory molecules may include CTLA-4 (also known as CD152), PD-1,'I'IM-3, BTLA, VISTA, LAG-3, B7-H3, B7-H4, IDO, TIGIT, MICA/B, or arginase.
  • the antagonist for inhibitory costimulatory molecules binds CTLA-4, PD-1,'I'IM-3, BTLA, VISTA, LAG-3 (e.g., LAG-3-IgG fusion protein (IMP321 )), B7-H3, B7-H4, IDO, TIGIT, MICA/B, or arginase antagonist antibody.
  • the anti-human OX40 agonist antibody can be administered in combination with an antagonist against CTLA-4 (also known as CD152), such as a blocking antibody.
  • CTLA-4 also known as CD152
  • the anti-human OX40 agonist antibody can be combined with ipilimumab (also known as MDX-010, MDX-101, or ) Combined application.
  • the anti-human OX40 agonistic antibody can be administered in combination with tremelimumab (also known as ticilimumab or CP-675,206).
  • the anti-human OX40 agonist antibody can be administered in combination with an antagonist against B7-H3 (also known as CD276), such as a blocking antibody.
  • the anti-human OX40 agonistic antibody can be administered in combination with MGA271. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an antagonist against TGFP, such as metelimumaM (also known as CAT-192), fresolimumab (also known as GC1008), or LY2157299.
  • an antagonist against TGFP such as metelimumaM (also known as CAT-192), fresolimumab (also known as GC1008), or LY2157299.
  • the anti-human OX40 agonist antibody can be administered in combination with a treatment comprising adoptive transfer of T cells expressing chimeric antigen receptor (CAR) (eg, cytotoxic T cells or CTL).
  • CAR chimeric antigen receptor
  • the anti-human OX40 agonistic antibody can be administered in combination with UCART19.
  • the anti-human OX40 agonistic antibody can be administered in combination with WT128z.
  • the anti-human OX40 agonistic antibody can be administered in combination with KTE-C19 (Kite).
  • the anti-human OX40 agonistic antibody can be administered in combination with CTL019 (Novartis).
  • the anti-human OX40 agonist antibody can be administered in combination with a treatment comprising adoptive transfer of T cells containing a dominant negative TGFI3 receptor, for example, a dominant negative TGFWI type receptor.
  • the anti-human OX40 agonistic antibody can be administered in combination with treatments that include the HERCREEM regimen (see, for example, ClinicalTrials.gov Identifier NCT00889954).
  • the anti-human OX40 agonist antibody can be administered in combination with an antagonist against CD19. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with MOR00208. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an antagonist against CD38. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with daratumumab.
  • the anti-human OX40 agonist antibody can be administered in combination with an agonist against CD137 (also known as TNFRSF9, 4-1BB, or ILA), such as an activating antibody.
  • the anti-human OX40 agonistic antibody can be administered in combination with urelumab (also known as BMS-663513).
  • the anti-human OX40 agonist antibody can be administered in combination with an agonist against CD40, such as an activating antibody.
  • the anti-human OX40 agonistic antibody can be administered in combination with CP-870893.
  • an anti-human OX40 agonist antibody can be administered in combination with an agonist against OX40 (also known as CD134), such as an activating antibody.
  • anti-human OX40 agonistic antibodies can be administered in combination with different anti-OX40 antibodies (eg, AgonOX).
  • the anti-human OX40 agonist antibody can be administered in combination with an agonist against CD27, such as an activating antibody.
  • the anti-human OX40 agonistic antibody can be administered in combination with CDX-1127.
  • the anti-human OX40 agonist antibody can be administered in combination with an antagonist against indoleamine-2,3-dioxygenase (IDO).
  • the IDO antagonist is 1-methyl-D-tryptophan (also known as I-D-MT).
  • the anti-human OX40 agonist antibody can be administered in combination with an agonist against CD137 (also known as TNFRSF9, 4-1BB, or ILA), such as an activating antibody.
  • the anti-human OX40 agonistic antibody can be administered in combination with urelumab (also known as BMS-663513).
  • the anti-human OX40 agonist antibody can be administered in combination with an agonist against CD40, such as an activating antibody.
  • the anti-human OX40 agonistic antibody can be administered in combination with CP-870893 or R07009789.
  • an anti-human OX40 agonist antibody can be administered in combination with an agonist against OX40 (also known as CD134), such as an activating antibody.
  • the anti-human OX40 agonist antibody can be administered in combination with an agonist against CD27, such as an activating antibody.
  • the anti-human OX40 agonistic antibody can be administered in combination with CDX-1127 (also known as varlilumab).
  • the anti-human OX40 agonist antibody can be administered in combination with an antagonist against indoleamine-2,3-dioxygenase (IDO).
  • the IDO antagonist is 1-methyl-D-tryptophan (also known as 1-D-MT).
  • the IDO antagonist is the IDO antagonist shown in WO2010/005958 (the content is clearly included through the record here).
  • the IDO antagonist is 4-( ⁇ 2-[(aminosulfonyl)amino]ethyl ⁇ amino)-N_(3-bromo-4-fluorophenyl)-N'-hydroxy-1, 2,5-oxadiazole-3-carboxamidine (e.g. as described in Example 23 of WO2010/005958).
  • the IDO antagonist is 4-( ⁇ 2-[(aminosulfonyl)amino]ethyl ⁇ amino)-N_(3-bromo-4-fluorophenyl)-N'-hydroxy-1, 2,5-oxadiazole-3-carboxamidine (e.g. as described in Example 23 of WO2010/005958).
  • the IDO antagonist is
  • the IDO antagonist is INCB24360. In some embodiments, the IDO antagonist is Indoximod (the D isomer of 1-methyl-tryptophan).
  • the anti-human OX40 agonist antibody can be administered in combination with an antibody-drug conjugate. In some embodiments, the antibody-drug conjugate comprises mertansine or monomethyl auristatin E (MMAE). In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an anti-NaPi2b antibody-MMAE conjugate (also known as DNIB0600A, RG7599 or Iifastuzumab vedotin).
  • the anti-human OX40 agonistic antibody can be combined with trastuzumab emtansine (also known as T-DM1, ado_trastuzumab emtansine, or KADCYL. Genentech) co-administered.
  • the anti-human OX40 agonist antibody can be administered in combination with the anti-MUC16 antibody-MMAE conjugate, DMUC5754A.
  • the anti-human OX40 agonist antibody can be administered in combination with the anti-MUC16 antibody-MMAE conjugate, DMUC4064A.
  • the anti-human OX40 agonist antibody can be administered in combination with an antibody-drug conjugate that targets the endothelin B receptor (EDNBR), such as an antibody against EDNBR conjugated with MMAE.
  • EDNBR endothelin B receptor
  • the anti-human OX40 agonistic antibody can be combined with an antibody-drug conjugate targeting lymphocyte antigen 6 complex, locus E (Ly6E), such as an antibody against Ly6E (also called Ly6E) conjugated with MMAE As DLYE5953A) combined administration.
  • the anti-human OX40 agonistic antibody can be administered in combination with polatuzumab vedotin.
  • the anti-human OX40 agonist antibody can be administered in combination with an antibody-drug conjugate that targets CD30.
  • the anti-human OX40 agonistic antibody can be administered in combination with ADCETRIS (also known as brentuximab vedotin).
  • the anti-human OX40 agonistic antibody can be administered in combination with polatuzumab vedotin.
  • the anti-human OX40 agonist antibody can be administered in combination with an angiogenesis inhibitor.
  • anti-human OX40 agonistic antibodies can be administered in combination with antibodies directed against VEGF, such as VEGF-A.
  • the anti-human OX40 agonist antibody can be combined with bevacizumab (also known as Genentech) co-administered.
  • the anti-human OX40 agonistic antibody can be administered in combination with an antibody against Angiopoietin 2 (also known as Ang2).
  • the anti-human OX40 agonistic antibody can be administered in combination with MEDI3617.
  • an anti-human OX40 agonist antibody can be administered in combination with an antibody against VEGFR2. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with ramucirumab. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with a VEGF receptor fusion protein. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with aflibercept. In some embodiments, the anti-human OX40 agonistic antibody can interact with ziv-aflibercept (also known as VEGF trap or ) Combined application. In some embodiments, anti-human OX40 agonistic antibodies can be administered in combination with bispecific antibodies directed against VEGF and Ang2.
  • the anti-human OX40 agonistic antibody can be administered in combination with RG7221 (also known as vanucizumab).
  • an anti-human OX40 agonist antibody can be combined with an angiogenesis inhibitor and a PD-1 axis binding antagonist (for example, a PD-1 binding antagonist such as an anti-PD-1 antibody, a PD-L1 binding antagonist such as Anti-PD-L1 antibody, and PD-L2 binding antagonist (such as anti-PD-L2 antibody) are administered in combination.
  • a PD-1 binding antagonist such as an anti-PD-1 antibody
  • a PD-L1 binding antagonist such as Anti-PD-L1 antibody
  • PD-L2 binding antagonist such as anti-PD-L2 antibody
  • the anti-human OX40 agonist antibody can be combined with bevacizumab and PD-1 axis binding antagonist (for example, PD-1 binding antagonist such as anti-PD-1 antibody, PD-L1 binding antagonist such as anti-PD-1 PD-L1 antibody, and PD-L2 binding antagonist such as anti-PD-L2 antibody) are administered in combination.
  • PD-1 axis binding antagonist for example, PD-1 binding antagonist such as anti-PD-1 antibody, PD-L1 binding antagonist such as anti-PD-1 PD-L1 antibody, and PD-L2 binding antagonist such as anti-PD-L2 antibody
  • the anti-human OX40 agonistic antibody can be administered in combination with bevacizumab and MDX-1106 (nivolumab, OPDIVO).
  • the anti-human OX40 agonistic antibody can be administered in combination with bevacizumab and Merck 3475 (MK-3475, pembrolizumab, KEYTRUDA). In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with bevacizumab and CT-011 (Pidilizumab). In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with bevacizumab and YW243.55.S70. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with bevacizumab and MPDL3280A.
  • the anti-human OX40 agonist antibody can be administered in combination with bevacizumab and MEDI4736. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with bevacizumab and MDX-1105.
  • the anti-human OX40 agonist antibody can be administered in combination with an anti-tumor agent. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an agent that targets CSF-1R (also known as M-CSFR or CD115). In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an anti-CSF-1R antibody (also known as IMC-CS4 or LY3022855). In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with the anti-CSF-1R antibody, RG7155 (also known as R05509554 or emactuzumab).
  • anti-human OX40 agonistic antibodies can be administered in combination with interferons, such as interferon- ⁇ or interferon- ⁇ .
  • the anti-human OX40 agonistic antibody can be administered in combination with Roferon-A (also known as recombinant interferon a-2a).
  • the anti-human OX40 agonistic antibody can be combined with GM-CSF (also known as recombinant human granulocyte macrophage colony stimulating factor, rhu GM-CSF, sargramostim, or ) Combined application.
  • the anti-human OX40 agonistic antibody can interact with IL-2 (also known as aldesleukin or ) Combined application.
  • the anti-human OX40 agonistic antibody can be administered in combination with IL-12.
  • the anti-human OX40 agonistic antibody can be administered in combination with IL27.
  • the anti-human OX40 agonistic antibody can be administered in combination with IL-15.
  • the anti-human OX40 agonistic antibody can be administered in combination with ALT-803.
  • anti-human OX40 agonistic antibodies can be administered in combination with antibodies that target CD20.
  • the CD20-targeting antibody is onuzumab (also known as GAlO 1 or ) Or rituximab.
  • anti-human OX40 agonistic antibodies can be administered in combination with antibodies that target GITR.
  • the antibody that targets GITR is TRX518.
  • the antibody targeting GITR is MK04166 (Merck).
  • the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of Bruton's tyrosine kinase (BTK). In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with ibrutinib. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of vaccine dehydrogenase I (IDH1) and/or vaccine dehydrogenase 2 (IDH2). In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with AG-120 (Agios).
  • IDH1 vaccine dehydrogenase I
  • IDH2 vaccine dehydrogenase 2
  • the anti-human OX40 agonistic antibody can be administered in combination with AG-120 (Agios).
  • the anti-human OX40 agonist antibody can be combined with onuzumab and PD-1 axis binding antagonist (for example, PD-1 binding antagonist such as anti-PD-1 antibody, PD-L1 binding antagonist such as anti-PD-1 PD-L1 antibody, and PD-L2 binding antagonist such as anti-PD-L2 antibody) are administered in combination.
  • PD-1 axis binding antagonist for example, PD-1 binding antagonist such as anti-PD-1 antibody, PD-L1 binding antagonist such as anti-PD-1 PD-L1 antibody, and PD-L2 binding antagonist such as anti-PD-L2 antibody
  • anti-human OX40 agonistic antibodies can be administered in combination with cancer vaccines.
  • the cancer vaccine is a peptide cancer vaccine, which in some embodiments is a personalized peptide vaccine.
  • the peptide cancer vaccine is a multivalent long peptide, a multipeptide, a peptide mixture, a hybrid peptide, or a peptide-pulsed dendritic cell vaccine (see, for example, Yamada et al., Cancer Sci, 104:14- 21, 2013).
  • the anti-human OX40 agonist antibody can be administered in combination with an adjuvant.
  • the anti-human OX40 agonist antibody can be combined with a TLR agonist, such as Poly-ICLC (also known as ), LPS, MPL, or CpG ODN treatments are administered in combination.
  • TLR agonist such as Poly-ICLC (also known as ), LPS, MPL, or CpG ODN treatments are administered in combination.
  • the anti-human OX40 agonist antibody can be administered in combination with tumor necrosis factor (TNF) a.
  • TNF tumor necrosis factor
  • the anti-human OX40 agonistic antibody can be administered in combination with IL-1.
  • the anti-human OX40 agonistic antibody can be administered in combination with HMGB1.
  • the anti-human OX40 agonist antibody can be administered in combination with an IL-10 antagonist.
  • the anti-human OX40 agonist antibody can be administered in combination with an IL-4 antagonist. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an IL-13 antagonist. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an IL-17 antagonist. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an HVEM antagonist. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an ICOS agonist (for example, by administering ICOS-L, or an agonistic antibody against ICOS). In some embodiments, anti-human OX40 agonistic antibodies can be administered in combination with treatments that target CX3CL1.
  • anti-human OX40 agonistic antibodies can be administered in combination with treatments that target CXCL9. In some embodiments, anti-human OX40 agonistic antibodies can be administered in combination with treatments that target CXCL10. In some embodiments, anti-human OX40 agonistic antibodies can be administered in combination with treatments that target CCL5. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an LFA-I or ICAM1 agonist. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with a selectin agonist.
  • the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of B-Raf. In some embodiments, the anti-human OX40 agonistic antibody can be combined with Verofenib (also known as ) Combined application. In some embodiments, the anti-human OX40 agonistic antibody can be combined with dabrafenib (also known as ) Combined application. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with encorafenib (LGX818).
  • the anti-human OX40 agonist antibody can be administered in combination with an EGFR inhibitor. In some embodiments, the anti-human OX40 agonistic antibody can be combined with erlotinib (also known as ) Combined application. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of EGFR-T790M. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with gefitinib. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with afatinib. In some embodiments, the anti-human OX40 agonist antibody can be combined with cetuximab (also known as ) Combined application.
  • the anti-human OX40 agonist antibody can be combined with panitumumab (also known as ) Combined application. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with rociletinib. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with AZD9291. In some embodiments, anti-human OX40 agonistic antibodies can be administered in combination with MEK, such as MEK1 (also known as MAP2K1) and/or MEK2 (also known as MAP2K2) inhibitors.
  • MEK such as MEK1 (also known as MAP2K1) and/or MEK2 (also known as MAP2K2) inhibitors.
  • the anti-human OX40 agonistic antibody can be administered in combination with cobimetinib (also known as CDC-0973 or XL-518). In some embodiments, the anti-human OX40 agonistic antibody can be combined with trametinib (also known as ) Combined application. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with binimetinib.
  • the anti-human OX40 agonist antibody can be administered in combination with inhibitors of B-Raf (e.g., verofenib or dabrafenib) and inhibitors of MEK (e.g., MEK1 and/or MEK2) (e.g., cobimetinib or trametinib) .
  • the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of ERK (eg, ERK1/2).
  • the anti-human OX40 agonistic antibody can be administered in combination with GDC-0994.
  • the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of B-Raf, an inhibitor of MEK, and an inhibitor of ERK1/2. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of EGFR, an inhibitor of MEK, and an inhibitor of ERK1/2. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with one or more MAP kinase pathway inhibitors. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with CK127. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of K-Ras.
  • the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of c-Met. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with onartuzumab (also known as MetMAb). In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of anapatic lymphoma kinase (ALK). In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with AF802 (also known as CH5424802 or alectinib). In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with crizotinib.
  • ALK anapatic lymphoma kinase
  • AF802 also known as CH5424802 or alectinib
  • the anti-human OX40 agonistic antibody can be administered in combination with crizotinib.
  • the anti-human OX40 agonistic antibody can be administered in combination with ceritinib. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of phosphatidylinositol 3-kinase (PI3K). In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with buparlisib (BKM-120). In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with pictilisib (also known as GDC-0941). In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with buparlisib (also known as BKM-120).
  • PI3K phosphatidylinositol 3-kinase
  • the anti-human OX40 agonist antibody can be administered in combination with perifosine (also known as KRX-0401). In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with a delta selective inhibitor of phosphatidylinositol 3-kinase (PI3K). In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with idelalisib (also known as GS-1101 or CAL-101). In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with taselisib (also known as GDC-0032).
  • perifosine also known as KRX-0401
  • PI3K delta selective inhibitor of phosphatidylinositol 3-kinase
  • the anti-human OX40 agonistic antibody can be administered in combination with idelalisib (also known as GS-1101 or CAL-101). In some embodiments, the anti-human OX40
  • the anti-human OX40 agonistic antibody can be administered in combination with BYL-719. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of Akt. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with MK2206. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with GSK690693. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with ipatasertib (also known as CDC-0068). In some embodiments, anti-human OX40 agonistic antibodies can be administered in combination with inhibitors of mTOR.
  • the anti-human OX40 agonistic antibody can be administered in combination with sirolimus (also known as rapamycin). In some embodiments, the anti-human OX40 agonistic antibody can interact with temsirolimus (also known as CCI-779 or ) Combined application. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with everolimus (also known as RADOO1). In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with ridaforolimus (also known as AP-23573, MK_8669, or deforolimus). In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with OSI-027.
  • sirolimus also known as rapamycin
  • the anti-human OX40 agonistic antibody can interact with temsirolimus (also known as CCI-779 or ) Combined application.
  • the anti-human OX40 agonistic antibody can be administered in combination with everolimus
  • the anti-human OX40 agonistic antibody can be administered in combination with AZD8055. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with INK128. In some embodiments, anti-human OX40 agonistic antibodies can be administered in combination with dual PI3K/mTOR inhibitors. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with XL765. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with GDC-0980. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with BEZ235 (also known as NVP-BEZ235).
  • BEZ235 also known as NVP-BEZ235
  • the anti-human OX40 agonistic antibody can be administered in combination with BGT226. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with GSK2126458. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with PF-04691502. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with PF-05212384 (also known as PKI-587).
  • the anti-human OX40 agonistic antibody can be administered in combination with an agent that selectively degrades the estrogen receptor. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with GDC-0927. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of HER3. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with duligotuzumab. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of LSD1. In some embodiments, anti-human OX40 agonistic antibodies can be administered in combination with inhibitors of MDM2.
  • anti-human OX40 agonistic antibodies can be administered in combination with inhibitors of BCL2. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with venetoclax. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of CHK1. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with CDC-0575. In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with an inhibitor of the activated hedgehog signaling pathway. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with ERIVEDGE.
  • anti-human OX40 agonistic antibodies can be administered in combination with radiation therapy. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with gemcitabine. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with nab-pacIitaxel (ABRAXANE). In some embodiments, the anti-human OX40 agonist antibody can be administered in combination with trastuzumab. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with TVEC. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with IL27.
  • the anti-human OX40 agonist antibody can be administered in combination with cyclophosphamide. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with an agent that recruits T cells to the tumor. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with lirilumab (IPH2102/BMS-986015). In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with Idelalisib. In some embodiments, anti-human OX40 agonistic antibodies can be administered in combination with antibodies that target CD3 and CD20. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with REGN1979. In some embodiments, anti-human OX40 agonistic antibodies can be administered in combination with antibodies that target CD3 and CD19. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with blinatumomab.
  • the anti-human OX40 agonistic antibody can be administered in combination with an oncolytic virus. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with carboplatin and nab-paclitaxel. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with carboplatin and paclitaxel. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with cisplatin and pemetrexed. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with cisplatin and gemcitabine. In some embodiments, anti-human OX40 agonistic antibodies can be administered in combination with FOLFOX. In some embodiments, the anti-human OX40 agonistic antibody can be administered in combination with FOLFIRI.
  • Such combination therapies documented above encompass combined administration (wherein two or more therapeutic agents are contained in the same formulation or separate formulations), and separate administration, in which case the administration of another therapeutic agent And/or the adjuvant before, at the same time, and/or after the administration of the antibody of the invention occurs.
  • the antibodies of the present invention can also be used in combination with radiation therapy.
  • the antibodies of the present invention can be administered by any suitable means (including parenteral, intrapulmonary, and intranasal, and if desired for local treatment, intralesional administration).
  • Parenteral infusion includes intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
  • the dosing can be carried out by any suitable route (for example, by injection, such as intravenous or subcutaneous injection).
  • Various dosing schedules are encompassed herein, including but not limited to single administration or multiple administrations at multiple time points, bolus administration, and pulse infusion.
  • the antibody of the present invention will be formulated, dosed and administered in a manner consistent with excellent medical practice. Factors considered in this context include the specific condition being treated, the specific mammal being treated, the clinical condition of the individual patient, the cause of the condition, the location of drug delivery, the method of administration, the schedule of administration, and other factors known to the medical practitioner.
  • the antibody need not be but is optionally formulated with one or more agents currently used to prevent or treat the condition in question.
  • the effective amount of such other drugs depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors mentioned above. These are generally used at the same dosage and route of administration as described herein, or at about 1-99% of the dosage described herein, or at any dosage and any route determined empirically/clinically to be suitable.
  • the appropriate dose of the antibody of the present invention (when used alone or in combination with one or more other therapeutic agents) will depend on the type of disease to be treated, the type of antibody, the severity of the disease and The course of the disease, whether the antibody is administered for prevention or treatment, previous therapies, the patient's clinical history and response to the antibody, and the consideration of the attending physician.
  • the antibody is suitable for administration to a patient in one or a series of treatments. Depending on the type and severity of the disease, about 1 ⁇ g/kg to 40 mg/kg of antibody can be administered to the patient as an initial candidate dose, whether for example by one or more divided administrations or by continuous infusion.
  • a typical daily dose may be in the range of about 1 ⁇ g/kg to 100 mg/kg or more.
  • the treatment will usually continue until the desired suppression of disease symptoms occurs.
  • Such doses may be administered intermittently, for example every week or every three weeks (for example, so that the patient receives about 2 to about 20 doses, or for example about 6 doses of antibody).
  • a higher initial loading agent can be applied, followed by one or more lower doses.
  • other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • immunoconjugates of the present invention can be used in place of or in addition to anti-OX40 antibodies to implement any of the aforementioned formulations or therapeutic methods.
  • NF- ⁇ B transcription factor binding elements in the promoter of the NF- ⁇ B-GFP reporter gene lentiviral system (QIAGEN, CCS-013G), which can drive the expression of GFP.
  • QIAGEN NF- ⁇ B transcription factor binding elements in the promoter of the NF- ⁇ B-GFP reporter gene lentiviral system
  • Jurkat cells ATCC, TIB-152 were infected with NF- ⁇ B-GFP reporter lentivirus, and Jurkat cells infected with lentivirus were screened with puromycin (InvivoGen, ant-pr-1).
  • TNF ⁇ protein 10ng/ml TNF ⁇ protein (Sino Biological, 10602-HNAE) was added to stimulate the cells selected with puromycin, and 24 hours later, the cell subsets that showed GFP fluorescence were sorted by flow cytometer (BD, FACSAria III) to obtain Jurkat/ NF- ⁇ B-GFP cells.
  • the DNA sequences of the positive controls OX40L and HEL used in this article were synthesized by Jinweizhi Company.
  • OX40L and HEL purification insert the DNA sequence into the eukaryotic expression vector pFUSE (InvivoGen, pfuse-hg1fc1), and use standard procedures to transfect 293F cells with PEI (Thermo Fisher Scientific, R79007); use Freestyle medium (Lifetechnologies, 12338026) ) Culture the cells on a shaker at 37°C. After 7 days of culture, the supernatant was collected and purified on the AKTA system (GE) using a Superdex TM 200 Increase prepacked column (GE, 28-9909-44).
  • OX40 signal transduction can activate the NF- ⁇ B pathway. If OX40 is successfully expressed on the surface of the Jurkat/NF- ⁇ B-GFP cell membrane, OX40L stimulation will induce the expression of GFP.
  • the constructed Jurkat/NF- ⁇ B-GFP cells sorted as described above were infected with the human OX40 lentiviral supernatant obtained above. After 16 hours, 100nM of the OX40L protein obtained above (as a positive control) and HEL (as a negative control) were added. Control) was stimulated, and 24 hours later, the cells with the strongest GFP fluorescence were sorted into a 96-well plate using a flow cytometer for single cell sorting.
  • the cells in the above 96-well plate were cultured in RPMI 1640 medium (Lifetechnologies, C11875500CP) containing 10% fetal bovine serum (Biological Industries, 04-001-1A) for 2 weeks, and 100 nM of the above was added after the single cells grew up.
  • RPMI 1640 medium Lifetechnologies, C11875500CP
  • HEL human serum
  • NF- ⁇ B-GFP+hOX40 select OX40L strongly positive and HEL negative monoclonal as Jurkat/NF- ⁇ B-GFP+hOX40 (referred to as NF- ⁇ B-GFP+hOX40) cells
  • NF- ⁇ B-GFP+hOX40 select OX40L strongly positive and HEL negative monoclonal as Jurkat/NF- ⁇ B-GFP+hOX40 cells
  • NF- ⁇ B-GFP+hOX40 select OX40L strongly positive and HEL negative monoclonal as Jurkat/NF- ⁇ B-GFP+hOX40 cells
  • the peripheral blood of 30 healthy adults was purchased from Tianjin Blood Center and Shanghai Miaotong Biological Company, and PBMCs were obtained by centrifugation with ficoll separation solution (Haoyang, LDS1075). Centrifugation conditions: 20°C, 2000rpm, up 5 down 0 mode for 30 minutes.
  • the PBMC obtained as described above was used to construct a human natural antibody library, and the library was constructed by a conventional method (phage display, Tim Clackson and Henry B. Lowman).
  • the obtained antibody heavy chain and light chain variable regions are randomly combined and displayed on the N-terminus of phage capsid protein pIII in the form of single-chain antibody scFv to obtain a phage display antibody library with a storage capacity of 10 10 .
  • Phage screening is a conventional technology (Phage Display: A Laboratory Manual, Carlos F Barbas III).
  • the biotinylated OX40 protein (acrobiosystems) was incubated with the phage display antibody library obtained as described above for 2 hours at room temperature. After the incubation, add 150ul streptavidin magnetic beads Dynabeads M280 (Lifetechnologies, 11205D) directly, and incubate on a mixer at room temperature for 30 min.
  • PBS-tween PBS: Lifetechnologies, 70011044; Tween: Sigma-Aldrich, 9005-64-5
  • PBS Lifetechnologies, 70011044; Tween: Sigma-Aldrich, 9005-64-5
  • PBS Lifetechnologies, 70011044; Tween: Sigma-Aldrich, 9005-64-5
  • PH2.2 glycine-hydrochloric acid
  • Bacteriophage conjugated The eluted phage was used to infect E. coli XL1-blue (Agilent, 200236), and the helper phage VCSM13 (Agilent, 200251) was added for amplification and used in the next round of screening. A total of three rounds of screening were carried out. After three rounds, the antibody phages that bind to CD40 were enriched, and the screening results are shown in Table 1.
  • An ELISA plate (Corning, 3690) was coated with 1 ⁇ g/ml of human OX40 (Acrobiosystems, OX0-H5224), and incubated overnight at 4°C. The overnight induced phage supernatant was added to the ELISA plate after overnight incubation, incubated for 1 h at room temperature, and washed 8 times with 0.05% PBST.
  • BSA Solarbio, A8020-100
  • HRP-conjugated anti-M13 GE, 27-9421-01, M13 is phage capsid protein
  • a positive clone was defined as the OX40 binding signal more than 3 times that of the BSA control. The results are shown in Figure 2. The positive rates of the second and third rounds of screening were 4.5% and 40.9%, respectively.
  • lentiviral core plasmid and helper plasmids pPACKH1-GAG, pPACKH1-REV, and pVSVG were co-transfected into 293FT cells at a ratio of 1:1:1:1.
  • the medium was changed and cultured at 37°C containing 10% fetal bovine serum ( In the DMEM medium of Biological Industries, 04-001-1A), continue to culture for 48 hours to collect the supernatant containing the anti-OX40 antibody lentiviral library; infect HEK293 cells with 50,000 pg of the anti-OX40 antibody lentiviral library, and after 16 hours of infection, Centrifuge to remove the medium containing the lentivirus, add fresh medium, and sort the infected HEK293 cells into a 96-well plate using flow sorting technology, so that each well contains only one infected HEK293 cell, and culture the cells for 3 weeks.
  • 10% fetal bovine serum In the DMEM medium of Biological Industries, 04-001-1A
  • the supernatant was added with 3 ⁇ 10 5 Jurkat/NF- ⁇ B-GFP+hOX40 reporter cells obtained as described above, and 2.5 ⁇ g/ml goat anti-human Fc (SouthernBiotech, SBA-2048-01) secondary antibody was added at the same time.
  • the pFUSE vector was sequenced, and the sequences of the positive antibodies 11-1, 2-1 and 2-2-1 were obtained.
  • the heavy chain and light chain coding nucleotide sequences of the anti-OX40 antibody were synthesized (see SEQ ID NO:49-60 for the respective constant region and variable region coding nucleotide sequences) (gold Weizhi Company), cloned them into the vector pFUSE, and transiently co-transfected the plasmids containing the heavy chain and the light chain into 293F suspension cells with PEI at a ratio of 1:1 to express the full-length antibody.
  • the AKTA system was used for purification with Superdex TM 200 Increase pre-packed column.
  • Biacore T200 (GE Healthcare) is used to detect the affinity of anti-OX40 antibodies.
  • the antibody was flowed through the Protein A chip at 10 ⁇ L/min and captured on the chip.
  • Use Running buffer HBS-EP+, GE
  • the concentration is set to 2 ⁇ M, 1 ⁇ M, 500nM, 250nM, 125nM, 62.5nM, 31.25nM.
  • ELISA was used to evaluate the effect of anti-OX40 antibody on the binding of OX40L and OX40.
  • the tested anti-OX40 antibodies significantly inhibited tumor growth relative to the IgG1 control.
  • the spleen and tumor of the mouse were stripped, and the spleen was directly ground and filtered with a 200-mesh nylon mesh to prepare a single cell suspension.
  • the tumor tissue was cut into small pieces and placed in 2ml enzyme mixture (each ml enzyme contains 1500U collagenase, hyaluronic acid) Acidase 1000U and DNase 2.5ul), shake in an incubator at 150rpm and 37°C for 1h. Filter and grind the incompletely digested tissue pieces with a 200 mesh nylon mesh to prepare a single cell suspension, centrifuge at 350g for 7 minutes, resuspend the cells with 2ml of red blood cell lysate, incubate on ice for 15 minutes, wash twice with PBS and count.
  • CD45+CD3+CD4+ is defined as CD4 + T cells
  • CD45+CD3+CD8+ is defined as CD8 + T cells.
  • the tested anti-OX40 antibodies significantly up-regulated helper CD4+ T cells and cytotoxic CD8+ T cells in the spleen and tumor relative to the control.
  • the spleen and tumor of the mouse were stripped and a single cell suspension was prepared.
  • the CD45, CD3, CD4 and CD8 antibodies were stained, and the ratio of CD4+ and CD8+ T cells was analyzed by flow cytometry.
  • CD45+CD3+CD4+ is defined as CD4+ T cells.
  • CD45+CD3+CD8+ is defined as CD8+ T cells.
  • Anti-OX40 antibodies can be divided into two types according to their different ways of agonizing.
  • the first type of OX40 agonistic activity does not depend on the crosslinking of Fc receptors; the other type requires Fc receptor crosslinking to have CD40 agonistic activity.
  • Fc receptor crosslinking There are more tumor-associated inflammatory cells in tumor tissues and surrounding draining lymph nodes, and Fc receptor Fc ⁇ R2b gathers more around tumor cells. Therefore, the "cross-linked antibody” agonist has higher tissue selectivity.
  • the antibody can produce obvious agonistic effects in the tumor microenvironment, while in the normal tissues of the body, the ability to act will be kept at a low level, so that it can be Improve the safety window of treatment.

Landscapes

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

Abstract

L'invention concerne un nouvel anticorps anti-OX40, une composition comprenant l'anticorps anti-OX40, un acide nucléique codant pour l'anticorps anti-OX40, un procédé de préparation de l'anticorps anti-OX40, et l'utilisation de l'anticorps anti-OX40.
PCT/CN2020/140259 2019-12-27 2020-12-28 Anticorps anti-ox40 et son utilisation WO2021129874A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202080073615.0A CN114630839A (zh) 2019-12-27 2020-12-28 抗ox40抗体及其用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201911380303.XA CN113045654A (zh) 2019-12-27 2019-12-27 抗ox40抗体及其用途
CN201911380303.X 2019-12-27

Publications (1)

Publication Number Publication Date
WO2021129874A1 true WO2021129874A1 (fr) 2021-07-01

Family

ID=76506637

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/140259 WO2021129874A1 (fr) 2019-12-27 2020-12-28 Anticorps anti-ox40 et son utilisation

Country Status (2)

Country Link
CN (2) CN113045654A (fr)
WO (1) WO2021129874A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114591988A (zh) * 2022-03-30 2022-06-07 北京贝来生物科技有限公司 一种激活肿瘤免疫的基因修饰干细胞制备方法

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113045655A (zh) * 2019-12-27 2021-06-29 高诚生物医药(香港)有限公司 抗ox40抗体及其用途

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108623685A (zh) * 2017-03-25 2018-10-09 信达生物制药(苏州)有限公司 抗ox40抗体及其用途
US20190169302A1 (en) * 2017-12-06 2019-06-06 Sorrento Therapeutics, Inc. Variant antibodies that bind ox40
CN110004177A (zh) * 2019-04-22 2019-07-12 中国食品药品检定研究院 一种检测抗ox40抗体的方法及其应用
CN110003338A (zh) * 2019-04-16 2019-07-12 北京免疫方舟医药科技有限公司 抗ox40抗体及其应用
CN110078825A (zh) * 2019-01-23 2019-08-02 北京天广实生物技术股份有限公司 结合ox40的抗体及其用途
CN110551216A (zh) * 2018-05-31 2019-12-10 信达生物制药(苏州)有限公司 多价抗ox40抗体及其用途

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101293917B (zh) * 2008-06-16 2010-12-01 南开大学 与pkr激酶结构域特异性结合的多肽及其应用
CN109476748B (zh) * 2016-08-08 2023-05-23 豪夫迈·罗氏有限公司 用于癌症的治疗和诊断方法
JP2019526623A (ja) * 2016-08-08 2019-09-19 ソレント・セラピューティクス・インコーポレイテッドSorrento Therapeutics, Inc. 抗ox40結合タンパク質
CA3045940A1 (fr) * 2016-12-15 2018-06-21 Abbvie Biotherapeutics Inc. Anticorps anti-ox40 et leurs utilisations
CN108623686A (zh) * 2017-03-25 2018-10-09 信达生物制药(苏州)有限公司 抗ox40抗体及其用途
CA3103040A1 (fr) * 2018-05-11 2019-11-14 Wuxi Biologics (Shanghai) Co., Ltd. Anticorps entierement humains diriges contre ox40, procede de preparation correspondant, et utilisation associee

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108623685A (zh) * 2017-03-25 2018-10-09 信达生物制药(苏州)有限公司 抗ox40抗体及其用途
US20190169302A1 (en) * 2017-12-06 2019-06-06 Sorrento Therapeutics, Inc. Variant antibodies that bind ox40
CN110551216A (zh) * 2018-05-31 2019-12-10 信达生物制药(苏州)有限公司 多价抗ox40抗体及其用途
CN110078825A (zh) * 2019-01-23 2019-08-02 北京天广实生物技术股份有限公司 结合ox40的抗体及其用途
CN110003338A (zh) * 2019-04-16 2019-07-12 北京免疫方舟医药科技有限公司 抗ox40抗体及其应用
CN110004177A (zh) * 2019-04-22 2019-07-12 中国食品药品检定研究院 一种检测抗ox40抗体的方法及其应用

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
PENG WEIYI, WILLIAMS LEILA J., XU CHUNYU, MELENDEZ BRENDA, MCKENZIE JODI A., CHEN YUAN, JACKSON HEATHER L., VOO KUI S., MBOFUNG RI: "Anti-OX40 Antibody Directly Enhances The Function of Tumor-Reactive CD8 + T Cells and Synergizes with PI3Kβ Inhibition in PTEN Loss Melanoma", CLINICAL CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 25, no. 21, 1 November 2019 (2019-11-01), US, pages 6406 - 6416, XP055823827, ISSN: 1078-0432, DOI: 10.1158/1078-0432.CCR-19-1259 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114591988A (zh) * 2022-03-30 2022-06-07 北京贝来生物科技有限公司 一种激活肿瘤免疫的基因修饰干细胞制备方法

Also Published As

Publication number Publication date
CN114630839A (zh) 2022-06-14
CN113045654A (zh) 2021-06-29
TW202130661A (zh) 2021-08-16

Similar Documents

Publication Publication Date Title
TWI803637B (zh) 特異性針對gucy2c之抗體及其用途
AU2016359609B2 (en) FGFR2 inhibitors alone or in combination with immune stimulating agents in cancer treatment
AU2015338974B2 (en) Combination therapy for treatment of disease
RU2769569C2 (ru) Нейтрализация ингибиторных путей в лимфоцитах
TWI824217B (zh) 抗ox40抗體及其用途
BR112019018378A2 (pt) proteínas de ligação ao antígeno anti-tigit e métodos de utilização destas
KR20210030956A (ko) Pd-l1 및 cd137에 결합하는 항체 분자
KR20210031479A (ko) Cd137 및 ox40에 결합하는 항체 분자
WO2021129874A1 (fr) Anticorps anti-ox40 et son utilisation
TWI841816B (zh) 抗ox40抗體及其用途
WO2022133123A1 (fr) Molécules de liaison à cd40 et leurs utilisations
WO2023275616A1 (fr) Anticorps monoclonal anti-ox40 et ses procédés d'utilisation
KR20230154315A (ko) 항체의 신규한 조합물 및 이의 용도
EP4308160A2 (fr) Méthodes de traitement du cancer gynécologique à l'aide d'une polythérapie avec des anticorps multispécifiques anti-muc16 x cd3 et des inhibiteurs de vegf

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: 20904407

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: 20904407

Country of ref document: EP

Kind code of ref document: A1