US20150197572A1 - Immunopotentiative composition - Google Patents

Immunopotentiative composition Download PDF

Info

Publication number
US20150197572A1
US20150197572A1 US14/638,985 US201514638985A US2015197572A1 US 20150197572 A1 US20150197572 A1 US 20150197572A1 US 201514638985 A US201514638985 A US 201514638985A US 2015197572 A1 US2015197572 A1 US 2015197572A1
Authority
US
United States
Prior art keywords
cells
antibody
lung cancer
mice
human
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US14/638,985
Other versions
US9067999B1 (en
Inventor
Tasuku Honjo
Nagahiro Minato
Yoshiko Iwai
Shiro Shibayama
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ono Pharmaceutical Co Ltd
Dana Farber Cancer Institute Inc
Original Assignee
Ono Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=30117379&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20150197572(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Ono Pharmaceutical Co Ltd filed Critical Ono Pharmaceutical Co Ltd
Priority to US14/638,985 priority Critical patent/US9067999B1/en
Assigned to ONO PHARMACEUTICAL CO., LTD., TASUKU HONJO reassignment ONO PHARMACEUTICAL CO., LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HONJO, TASUKU, MINATO, NAGAHIRO, SHIBAYAMA, SHIRO, IWAI, YOSHIKO
Application granted granted Critical
Publication of US9067999B1 publication Critical patent/US9067999B1/en
Publication of US20150197572A1 publication Critical patent/US20150197572A1/en
Assigned to DANA-FARBER CANCER INSTITUTE, INC. reassignment DANA-FARBER CANCER INSTITUTE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FREEMAN, GORDON J.
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1774Immunoglobulin superfamily (e.g. CD2, CD4, CD8, ICAM molecules, B7 molecules, Fc-receptors, MHC-molecules)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70521CD28, CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • C12Q1/025Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/54F(ab')2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to immunopotentiation characterized by inhibiting immunosuppressive signals induced by PD-1, PD-L1 or PD-L2, compositions for cancer or infection treatment, and therapies that use them.
  • the present invention relates to the compositions for cancer or infection treatment through an immunopotentiation caused by inhibition of immunosuppressive signals induced by PD-1, PD-L1 or PD-L2, the therapies that use them, screening methods of immunopotentiative substances, or substances for cancer treatment or for infection treatment that are contained in the composition as an active ingredient, cell lines used for those screening methods, an evaluation that selects the substances for cancer treatment, and carcinoma cells transplanted mammals used for the evaluation.
  • Immunotherapies can reduce side reactions that can't be avoided easily in most chemotherapies, and is expected as a therapy with extremely high specificity.
  • the Immunotherapies can be executed in the purpose to recover patient's QOL by activating the immune reaction that human originally has by an extrinsic method and subrogating a part of the load by medication.
  • Immunopotentiation can be executed by methods of activating immune reaction of T lymphocytes. It is said that not only stimulation through antigen receptors (TCR) but also an additionally stimulative inducement through conjugated stimulative molecular groups (for example, CD28) could be necessary for the activation of T cells. However, it is reported that as the molecular groups with homologous structures to the conjugated stimulative molecular groups, CTLA-4 and PD-1 were discovered recently and give signals that suppress signals of antigen receptors (TCR). It is thought that a method of activating T cells could be an effective mean to suprress the function of those conjugated control molecules.
  • conjugated stimulative molecular groups for example, CD28
  • PD-1 was cloned as 55 kD of I type membrane protein that belong to an immunoglobulin family (The EMBO Journal (1992), vol. 11, issue 11, p. 3887-3895, JP5336973, JP7291996).
  • Human PD-1 cDNA is composed of the base sequence shown in EMBL/GenBank Acc. No. NM — 005018 and mouse PD-1 cDNA is composed of the base sequence shown in Acc. No. X67914, and those expression are observed when thymus cells differentiate from CD4-CD8-cell into CD4+CD8+ cell (International Immunology (1996), vol. 18, issue 5, p. 773-780., Journal of Experimental Medicine (2000), vol. 191, issue 5, p.
  • ITIM motifs Immunoreceptor Tyrosine-based Inhibitory Motif
  • PD-1-deficient mice develop lupus-like autoimmune disease such as glomerular nephritis and arthritis (for C57BL/6 gene background.)
  • International Immunology (1998), vol.10, issue 10, p. 1563-1572., Immunity (1999), vol. 11, issue 2, p. 141-151.) and a disease like dilated cardiomyopathy (for BALB/c gene background.)( Science (2001), vol. 291, issue 5502, p. 319-332.)
  • PD-1 could be a control factor of development of autoimmune disease, especially the peripheral self-tolerance.
  • PD-L1 human PD-L1 cDNA is composed of the base sequence shown by EMBUGenBank Acc. No. AF233516 and mouse PD-L1 cDNA is composed of the base sequence shown by NM — 021893.
  • antigen-presenting cells such as activated monocytes and dendritic cells ( Journal of Experimental Medicine (2000), vol. 19, issue 7, p. 1027-1034.).
  • These cells present interaction molecules that induce a variety of immuno-inductive signals to T lymphocytes, and PD-L1 is one of these molecules that induce the inhibitory signal by PD-1.
  • PD-L1 ligand stimulation suppressed the activation (cellular proliferation and induction of various cytokine production) of PD-1 expressing T lymphocytes.
  • PD-L1 expression has been confirmed in not only immunocompetent cells but also a certain kind of tumor cell lines (cell lines derived from monocytic leukemia, cell lines derived from mast cells, cell lines derived from hepatic carcinomas, cell lines derived from neuroblasts, and cell lines derived from breast carcinomas) ( Nature Immunology (2001), vol. 2, issue 3, p. 261-267.).
  • PD-L 2 human PD-L 2 cDNA is composed of the base sequence shown by EMBUGenBank Acc. No. NM — 025239 and mouse PD-L2 cDNA is composed of the base sequence shown by NM — 021896.
  • PD-L1 Nature Immunology (2001), vol. 2, issue 3, p. 261-267.
  • the inhibitory signals from the conjugated suppressive molecules represented by PD-1 could control abnormal immune reaction to autoantigen and immunological tolerance in lymphocyte generation or maturation by a mechanism that appropriately controls positive signals with antigen receptors (TCR) and conjugated stimulation molecules. It has been thought that a certain kind of tumour and virus could use those conjugated suppressive molecules to intercepte the activation and proliferation of T cells and weaken the host immunity reaction to oneself by a direct or indirect mechanism ( Cell (1992), vol. 71, issue 7, p. 1093-1102, Science (1993), vol. 259, issue 5093, p. 368-370.). It has been thought that those conjugated suppressive molecules could have caused the impairment of T cells in a part of disease thought to originate in impairment of T cells.
  • a problem of the present invention is to provide compositions to activate immunity by inhibiting the inhibitory signals of PD-1, PD-L1 or PD-L2 and compositions for cancer or infection treatment through this mechanism.
  • the present inventors paid attention to PD-1, PD-L1, or PD-L2 as a new target in cancer or infection treatment and found that substances that inhibit the inhibitory signals of PD-1, PD-L1 or PD-L2 inhibit cancer proliferation through the mechanism of the recovery and activation of immune function. Further, they found that PD-1 signal, concretely, an interaction of PD-1 and PD-L1 or PD-1 and PD-L2 took part in the exclusion of infectious virus. According to those facts, they found the substances that could inhibit the inhibitory signals of PD-1, PD-L1 or PD-L2 having therapeutic potential for cancer or infection and completed the present invention.
  • the present invention relates to
  • PD-1, PD-L1, or PD-L2 of the present invention includes each one derived from mammal, for example, mouse, rat, hamster, guinea pig, dog, pig, ape, or primate including human. They are suitable to be human PD-L1, PD-1, and PD-L2, respectively.
  • the immunosuppressive signal of PD-1, PD-L1, or PD-L2 in the present invention is at least composed of the interaction of PD-1 and PD-L1 or PD-1 and PD-L2, and the intracellular signalings of PD-1. Production of PD-1, PD-L1 or PD-L2 itself is included in them.
  • the immunosuppressive signal of PD-1, PD- L1 , or PD-L2 in the present invention is inhibited by direct or indirect inhibition of the interaction of PD-L1 or PD-1 and PD-L2 or the intracellular signalings of PD-1.
  • a substance that selectively binds to PD-1, PD-L1, or PD-L2 respectively is included as a substance with those inhibitory activities.
  • it is suitable to be protein, polypeptide or peptide, polynucleotide or polynucleoside, antibody or the derivative, organic synthesis compound, inorganic compound, or natural product.
  • an antibody to PD-1, PD-L1 or PD-L2 is enumerated as an excellent substance in specificity.
  • the immunosuppressive signals are inhibited by inhibiting production of PD-1, PD-L1 or PD-L2 itself.
  • antibodies to PD-1, PD-L1 or PD-L2 all antibodies derived from human, mouse, rat, rabbit, or goat which can inhibit the immunosuppressive signals by PD-1, PD-L1, or PD-L2, those polyclonal or monoclonal antibodies, complete or shorten (for example, F(ab′) 2, Fab′, Fab, or Fv fragment) antibodies, chimeric antibodies, humanized antibodies, or completely humanized antibodies will be acceptable.
  • Such antibodies can be manufactured using a partial protein of the extracellular region of PD-1, PD-L1, or PD-L2 as an antigen according to well-known production methods of antibody or antiserum.
  • the partial protein of the extracellular region can be prepared by well-known protein expression and purification techniques.
  • the polyclonal antibodies can be manufactured according to well-known methods. For example, they can be manufactured by separation and refinement of the antibody of which a mixture of an antigen and a carrier protein is immunized to suitable animal, and an antibody inclusion to the antigen is gathered from the immunized animal. As such animal, mouse, rat, sheep, goat, rabbit, and guinea pig are generally enumerated. To improve the antibody producibility, Freund's complete adjuvant or Freund's incomplete adjuvant can be administered with the antigen. The administering is usually executed once every two weeks about 3-10 times in total.
  • the polyclonal antibody can be gathered from the immunized animal's blood and peritoneal fluid, etc. by the above method.
  • the measurement of the polyclonal antibody's titer in antiserum can be measured by ELISA.
  • the separation and refinement of the polyclonal antibody can be executed by refining techniques that use active adsorbents such as antigen binding solid phase, protein A, or protein G, etc., salting-out, alcohol precipitation, isoelectric precipitation, electrophoresis, adsorption and desorption with ion exchanger, ultracentrifugation, or separation and refinement of immunoglobulins such as gel filtration technique, etc.
  • the monoclonal antibody or the modifier is more suitable.
  • the monoclonal antibody producing cells can be prepared as hybridomas to be possible to subculture which produce the monoclonal antibody by selecting the individual of which the antibody titre is confirmed in an antigen immunized animals, gathering the spleen or the lymph node on day 2-5 after the final immunization, and fusing the antibody producting cells included in them with homogeneous or heterozoic myeloma cells.
  • the antigen itself or with the carrier and the diluent is administered to the part in which the antibody production is possible.
  • Freund's complete adjuvant or Freund's incomplete adjuvant can be administered with the antigen. According to the method of calling “DNA immunization”, animals are immunized.
  • This method is a method using a phenomenon in which antigen-expressing vectors are introduced into the part and are taken into myocytes on the process of tissue repair, and expresses the antigenic protein ( Nature Immunology (2001), vol. 2, issue 3, p. 261-267 ) after Cardiotoxin is treated to immune animal's tibialis anterior muscle of hind leg.
  • mouse and rat are suitable as an immune animal, mouse, rat, sheep, goat, rabbit, or guinea pig can be used, mouse and rat are suitable.
  • the fusion operation can be executed by the method ( Nature (1975), vol. 256, issue 5517, p. 495-497.) of Kohler and Milstein, and as fusion accelerants, polyethylene glycol (PEG) and Sendai virus, etc. are used.
  • PEG polyethylene glycol
  • Sendai virus etc.
  • myeloma cells myeloma cells such as P3U1, NS1, SP2/0, and AP1 can be used, P3U1 are often used usually.
  • the monoclonal antibody producing cells can be selected by detecting by ELISA, etc.
  • the separation and refinement of the monoclonal antibody can be executed according to the separation refining method similar to the separation and refinement of immunoglobulin for the above polyclonal antibody. Concretely, it is anti-mouse PD-L1 antibody producted by hybridomas identified by International Trust Number FERM BP-8396 or anti-human PD-1 antibody producted by a hybridoma identified by International Trust Number FERM BP-8392.
  • the hybridomas identified by International Trust Number FERM BP-8392 had been deposited as Trust Number FERM P-19162 to National Institute of Advanced Industrial Science and Technology, International Patent Organism Depositary in Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, Japan (ZIP code 305-8566) at Dec. 19, 2002, and had been transferred to International Deposit at Jun. 5, 2003.
  • the hybridomas identified by International Trust Number FERM BP-8396 had been deposited as Trust Number FERM P-18908 at Jun. 25, 2002, and had been transferred to International Deposit at Jun. 11, 2003.
  • the antibody fragment means F(ab′) 2 , Fab′, Fab, or scFv antibody fragment and can be prepared by reducing optionally after processing with protease enzyme.
  • F(ab′) 2 antibody fragments can be purified by either method of affinity chromatography such as ion-exchange chromatography, gel filtration, or protein A or protein G column, etc. after the purified monoclonal antibody is completely digested by pepsin. Because digestive time by pepsin is different depending on Ig subtype, it is necessary to prepare it suitably.
  • Fab′ antibody fragment can be prepared by reducing F(ab′) 2 by 2-mercaptoethylamine partly.
  • Fab antibody fragment can be prepared by the direct digestion under the presence of cysteine by the digestive enzyme papain followed by refining.
  • the monoclonal antibody is prepared as a rearrangement antibody and a hybrid antibody modified by gene recombination technology using DNA sequence that codes the amino acid sequence of antibody isolated from the hybridomas.
  • it can be prepared as a single-chain antibody but not a usual complete type antibody.
  • scFv antibody single chain Fv
  • scFv antibody single chain Fv
  • the single-chain antibody with the characteristic and the affinity of an original antibody can be prepared by making expression an expression vector including a fused DNA of which DNA fragments respectively coding a variable region of the heavy chain and the light chain had been connected with a spacer coding neurtal amino acids (glycine or serine) in suitable host cells.
  • the humanized antibody modified to be acceptable for administering to human is the antibody which is modified so that the decrease of antigenicity or the blood movement may improve to extent that can be allowed in pharmacology when the antibody is administered to human.
  • Human PD-1 antibody or human PD-L1 antibody in specification of the present invention includes the humanized or the complete human type antibody, too.
  • the humanized antibody can be prepared by substituting a part of non-human antibody which was prepared by being immunized to mammals other than human for a part of human antibody. Concretely, it has been known to be able to be prepared by constructing a chimera with a gene coding a constant region of the human antibody (Proc. Natl. Acad. Sci. (USA) (1987), vol. 84, p. 3439-3443, Journal of Immunology (1987), vol. 139, issue 1, p. 3521.).
  • the DNA sequence of the human constant region has been described to a prior art and the constant region gene can be easily obtained from an already-known clone. Then, the DNA sequence coding the variable region of the antibody can be fused to the human constant region.
  • An isotype of the human constant region can be selected due to desire effective function or antibody dependent cytotoxicity.
  • a suitable isotype is IgG1, IgG3, and IgG4. Both human light-chain constant region, K chain, and A chain can be used. It is possible to make this humanized chimeric antibody expression by a routine method.
  • the complete human type antibody can be prepared by using mice (XenoMouse ( Chemical Biology (2000), vol. 7, issue 8, p. R185-6.), HuMAb-Mouse ( Infection and Immunity (2002), vol. 70, issue 2, p. 612-9.), TC mouse (Biotechnology and Genetics Enginnering Revew (2002),vol. 19, p. 73-82.), and KM mouse (Cloning Stem Cells (2002), vol. 4, issue 1, p. 91-102.)) of which a constant region gene of human immunoglobulin have been transferred, and a target antibody can be mass-produced by making the antibody production lymphocytes separated from mice to hybridomas. It can be prepared by phage display method ( FEBS Letter (1998), vol.
  • the human type antibody can be expressed on the surface of the phage as a form fused with coat protein of the phages.
  • Polypeptides or the derivatives that bind to PD-1, PD-L1, or PD-L2 include each partial proteins of PD-1, PD-L1 or PD-L2 of which the immunosuppressive signal is not induced.
  • the presence of PD-1 in the neighborhood of the antigen receptors is indispensable for the inducement of the immunosuppressive signal of PD-1, for that purpose it is necessary to be restrained by the interaction with PD-L1 or PD-L2 in antigen-presenting cells, tumours, or carcinoma cells. Therefore, soluble PD-L1 or soluble PD-L2 with a part which is only extracellular domains and interacts with PD-1can inhibit the immunosuppressive signal of PD-1.
  • soluble PD-1 with a part which has a similar structure and can interact with PD-L1 or PD-L2 can inhibit the immunosuppressive signal.
  • These soluble proteins have only to include an extracellular region which is necessary and sufficient to bind to PD-1, PD-L1, or PD-L2 and can be prepared by a well-known expression and refining technique.
  • an interaction inhibitor of PD-1 and PD-L1 or PD-1 and PD-L2 is a protein or a polypeptide and an essential area for the interaction is composed by only consecutive polypeptide, such a polypeptide fragment can become a mutual antagonist.
  • an antagonist with stronger activity can be identified from molecular groups of which this polypeptide fragment is chemically modified, or designed by computer based on the spatial structure of the polypeptide fragment.
  • the best antagonist can be more efficiently selected from molecular groups designed by computer based on protein stereoanalysis data of the interaction area.
  • a substance that inhibits the interaction of PD-1 and PD-L1, or PD-1 and PD-L2 can be screened directly.
  • Such a substance can be identified from each libraries of protein, polypeptide or peptide, polynucleotide or polynucleoside, non-peptide compound, organic synthesis compound, or natural product (for example, fermentational product, cell extract, plant extract, and animal tissue extract.).
  • inhibitory signals of PD-1 intracellular domain are caused by contacting dephosphorylation enzymes (for example, SHP-1 and 2 (Sathish J. G., Journal of Immunology (2001), vol. 166, issue 3, p. 1763-70.)) that bind to ITIM in PD-1 intracellular domain with an intracellular complex including an antigen receptor complex, they are generally inhibited by inhibiting the contact between the antigen receptor complex and PD-1.
  • dephosphorylation enzymes for example, SHP-1 and 2 (Sathish J. G., Journal of Immunology (2001), vol. 166, issue 3, p. 1763-70.)
  • the substance that inhibits the inhibitory signals includes a substance that directly inhibits the activity of dephosphorylation enzymes, a substance that inhibit phosphorylation of tyrosine residue in ITIM, a substance that inhibits bonds of dephosphorylation enzymes to ITIM, or a substance that directly inhibits activity of dephosphorylation enzymes, etc.
  • the antigen receptor complex includes T cell receptor complex or B cell receptor complex.
  • the production of PD-1, PD-L1, or PD-L2 is inhibited by specific polynucleotide or polynucleoside, organic synthesis compound, inorganic compound, or natural product, etc.
  • the suitable polynucleotide or polynucleoside includes antisense nucleotide derivative so-called ribozyme. This uses a mechanism of which the expressed mRNA is destroyed by which the polynucleotide derivative that is complement to mRNA of PD-1, PD-L1, or PD-L2 is transferred into subject cells.
  • the vector can be used for the gene manipulation to lymphocyte stem cells gathered from a patient so that it inhibits the expression of PD-1, and the manipulated cells can be used for cell medical treatment of which the cells will be proliferated, differentiated, activated, and administered to the patient again.
  • the manipulated cells can be used for cell medical treatment of which the cells will be proliferated, differentiated, activated, and administered to the patient again.
  • more specific and clonal lymphocytes to target cells can be prepared by adding a specific antigen of the target cells on the process of the maturation and activation of lymphocyte stem cells.
  • the screening method of the present invention can be executed by a method of measuring cell function.
  • the carcinoma cell lines for screening of which PD-L1 or PD-L2 used by the method is transformed to be expressed includes carcinoma cell lines transformed transitory or stably after expression vectors constructed to express PD-L1 or PD-L2 have been introduced to the cells by well-known methods.
  • carcinoma cell lines used ape COS-1 cells, COS-7 cells, Vero, chinese hamster CHO cells (hereafter, abbreviated with CHO cells.), dhfr gene deficient chinese hamster CHO cells (hereafter, abbreviated with CHO (dhfr-) cells.), mouse L cells, mouse AtT-20, mouse myeloma cells, rat GH3, HEK293 T cells, and human FL cells, etc. are used.
  • the transformation of animal cells can be executed according to methods described in, for example, Cell Tehcnology separate volume 8, New Experimental Protocol of Cell Technology (1995), vol. 263, published by SHUJUNSHA Co., Ltd., and Virology (1973), vol. 52, issue 456 .
  • Cells naturally expressing PD-L1 or PD-L2 can be also used. Such cells include leukocyte, suitablely, monocytes, macrophages or antigen-presenting cells, epithelial cells, tumor cells, carcinoma cells, or those cell lines, etc.
  • tumor cells or carcinoma cells for example, P38D1 cells, P815 cells, NB41A3 cells, MDA-231 cells, SKBR-3 cells, MCF-7 cells, BT474 cells, J558L cells, P3U1 cells, PAI cells, X63 cells, or SP2/0 cells can be used.
  • Cells infected with pathogens, which express PD-L1 or PD-L2 naturally or compellingly, can be used.
  • Infectious pathogens include human hepatitis virus (hepatitis B, hepatitis C, and hepatitis A) or hepatitis E), human retrovirus, human immunodeficiency virus (HIV1 and HIV2), human T cell leukemia virus, human T lymphocytic virus (HTLV1 and HTLV2), simple herpes virus type 1 or 2, Epstein-Barr virus, cytomegalovirus, varicella-zoster virus, human herpesvirus including human herpesvirus 6, poliovirus, measles virus, rubella virus, Japanese encephalitis virus, mumps virus, influenza virus, adenovirus, enterovirus, rhinovirus, virus developing severely acute respiratory syndrome (SARS), Ebola virus, West Nile virus, or these virus modified artificially.
  • human hepatitis virus hepatitis B, hepatitis C, and hepatitis A) or hepatitis E
  • human retrovirus human immunodeficiency virus (
  • pathogens include, for example, pathogenesis protozoan (for example, trypanosome, malaria, and toxoplasma), bacillus (for example, mycobacterium, salmonella, and listeria) or fungus (for example, candida), etc.
  • pathogenesis protozoan for example, trypanosome, malaria, and toxoplasma
  • bacillus for example, mycobacterium, salmonella, and listeria
  • fungus for example, candida
  • the lymphocytes used in the screening method of the present invention are T or B lymphocytes, and suitably cytotoxic T lymphocytes (CTL).
  • CTL cytotoxic T lymphocytes
  • the immune reaction of lymphocytes in the screening method of the present invention includes citotoxic response (for example, tumor immunity reaction), mixed lymphocyte reaction, production of cytokines, antibodies, complements or other cell surface antigens, or cellular proliferation, etc.
  • the screening method of an active ingredient contained in the composition for immunopotentiation or cancer treatment can be executed by measuring cytotoxic activity of cytotoxic T lymphocytes against subject cells followed by measuring an effect of a subject substance against the activity.
  • This method is an assay of recovery and reinforcement of cytotoxic activity by adding the subject substance to a culture mixed cytotoxic T lymphocytes (CTL) naturally expressing PD-1 or cell lines (for example, 2C cells) with cells naturally or compulsorily expressing PD-L1 or PD-L2 which are derived from syngeneic mouse.
  • CTL culture mixed cytotoxic T lymphocytes
  • PD-1 or cell lines for example, 2C cells
  • this method has a feature of which the recovery of cytotoxic activity (rising part) due to the subject substance can be measured more clearly.
  • the recovery of cytotoxicity due to the subject substance can be evaluated as an equivalent to inhibition of suppression of cytotoxicity. Further, it is preferable that the the cytotoxicity due to the subject substance is arbitrarily measured.
  • these cells tumor cell lines or carcinoma cell lines naturally expressing PD-L1 or PD-L2 ( Nature Immunology (2001), vol. 2, issue 3, p.
  • P38D1 cells for example, P38D1 cells, P815 cells, NB41A3 cells, MDA-231 cells, SKBR-3 cells, MCF-7 cells, BT474 cells, J558 L cells, P3U1 cells, PAI cells, X63 cells, or SP2/0 cells can be used, and tumor cell lines or carcinoma cell lines transformed so as to stably or transiently express PD-L1 or PD-L2 can be also used.
  • the cytotoxic lymphocytes are PD-1 expressing cells derived from syngeneic animal to targeted cells.
  • the screening method of an active ingredient contained in the composition for infection treatment is an assay of enhancement effect on immune reaction of lymphocytes to infected cells or inhibitory effect on proliferation activity of pathogen or virus by adding the subject substance to a culture mixed cytotoxic T lymphocytes (CTL) or cell lines (for example, 2C cells) naturally expressing
  • CTL cytotoxic T lymphocytes
  • 2C cells naturally expressing
  • PD-1 with cells naturally or compulsorily expressing PD-L1 or PD-L2 which are derived from syngeneic mouse and was infected with pathogen or virus.
  • a mammal created by transplanting the above carcinoma cell lines for screening which are transformed so as to express PD-L1 or PD-L2 or cells naturally expressing PD-L1 or PD-L2 to a syngeneic mammal can be used.
  • a process of transplanting cells and a process of breeding the mammal until becoming appropriate for the evaluation is indispensable.
  • This evaluation is characterized in evaluating proliferation of the transplant cells and the amount of production of various cytokines or cell surface antigens, especially, in case of carcinoma cells, proliferation, permeation, or histological analysis of metastasis of the cells, or survival rates of the transplanted mammal.
  • the cellular proliferation can be evaluated as the number of carcinoma cells per unit capacity in case of ascites tumors or blood cancers, or the size or the weight after removing in case of solid cancer.
  • the effect of the subject substance for cancer treatment in this method can be evaluated as an equivalent to effect due to inhibition of suppression of cytotoxicity caused in PD-L1 or PD-L2.
  • syngeneic cells to a mammal for transplant, with good proliferation are more suitable.
  • the mammal include s primates except human, mouse, rat, hamster, guinea pig, dog, pig, and ape.
  • each specific antibody anti-PD-1 antibody and anti-PD-L1 antibody
  • These antibodies showed actions that recover or reinforce cytotoxic activity that has relatively decreased by presenting PD-L1 ligand to PD-1-expressing CTL (cytotoxic T lymphocyte) (example 1 and FIG. 1 .). This suggests that the cytotoxic activity to carcinoma cells by CTL could be reinforced by administering these antibodies.
  • CTL cytotoxic T lymphocyte
  • the substances with such effects include, for example, anti-PD-L2 antibody, soluble PD-1, soluble PD-L1, soluble PD-L2, PD-1 antagonists, PD-L1 antagonists, PD-L2 antagonists, substances that inhibits interaction between PD-1 and PD-L1 or PD-1 and PD-L2, PD-1 production inhibitors, PD-L1 production inhibitors, PD-L2 production inhibitors, and intracellular inhibitory signal inhibitors by PD-1.
  • Cancer or tumour of which the therapeutic potential could be expected by administration of the composition for cancer treatment of the present invention include, for example, carcinoma, squamous carcinoma (for example, cervical canal, eyelid, tunica conjunctiva, vagina, lung, oral cavity, skin, urinary bladder, tongue, larynx, and gullet), and adenocarcinoma (for example, prostate, small intestine, endometrium, cervical canal, large intestine, lung, pancreas, gullet, intestinum rectum, uterus, stomach, mammary gland, and ovary). Further, they include sarcomata (for example, myogenic sarcoma), leukosis, neuroma, melanoma, and lymphoma.
  • carcinoma for example, cervical canal, eyelid, tunica conjunctiva, vagina, lung, oral cavity, skin, urinary bladder, tongue, larynx, and gullet
  • adenocarcinoma for example, prostate, small intestine
  • PD-L1 or PD-L2 expression can be identified by checking up surgically excised cancer, tumour mass, or lesioned part gathered outside of the body as a sample. Administration of the composition of the present invention will become an efficient and available method as a treatment after surgery for tumour or cancer patient that PD-L1 or PD-L2 is remarkably expressed.
  • PD-L1 or PD-L2 expression can be identified by, for example, immunochemical method using PD-L1 antibody or PD-L2 antibody, RT-PCR, or DNA array.
  • a side reaction violently decreasing lymphocytes proliferation in chemotherapy and radiotherapy for cancer is inevitable.
  • Administration of the composition of the present invention presents an effect on stimulating and proliferating the decreased lymphocytes and can suppress a fierce side reaction accompanied by usual chemotherapy to minimum. Further, it is similar as to radiotherapy.
  • Concomitant use with the composition of the present invention can greatly decrease the dose of chemotherapy drug or the exposure dose of irradiation from the dose or the exposure dose used usually.
  • composition for cancer treatment of the present invention can be used together with existing chemotherapy drugs or be made as a mixture with them.
  • a chemotherapy drug include, for example, alkylating agents, nitrosourea agents, antimetabolites, antitumor antibiotics, alkaloids derived from plant, topoisomerase inhibitors, hormone therapy medicines, hormone antagonists, aromatase inhibitors, P-glycoprotein inhibitors, platinum complex derivatives, other immunotherapeutic drugs, and other anticancer agents.
  • they can be used together with hypoleukocytosis (neutrophil) medicines that are cancer treatment adjuvant, thrombopenia medicines, antiemetic drugs, and cancer pain medicines for patient's QOL recovery or be made as a mixture with them.
  • the composition for cancer treatment of the present invention can be used together with immunopotentiative substances or be made as a mixture with them.
  • immunopotentiative substances include, for example, various cytokines and a tumor antigen, etc.
  • Cytokines that stimulate immune reactions include, for example, GM-CSF, M-CSF, G-CSF, interferon- ⁇ , ⁇ , ⁇ , IL-1, IL-2, IL-3, and IL-12, etc.
  • B7 ligand derivatives, anti-CD3 antibodies and anti-CD28 antibodies, and anti-CTLA-4 antibodies can also improve the immune reactions.
  • the tumor antigen can also improve a specific immune reaction against T lymphocytes against carcinoma cells and additionally or synergistically reinforce by using together with the composition for cancer treatment of the present invention.
  • the tumor antigen can be prepared as a purified protein in case of an already-known gene, or as a lysate of carcinoma cells in case of an unknown gene.
  • Such the tumor antigen includes, for example, HLA-A1 and HLA-A2 restrictive peptides of malignant melanoma MAGE-1 or MAGE-3, MART-1, and gp100. Further, they include HER2/neu peptide of mammary carcinomas and ovarian carcinomas, MUC-1 peptide of adenocarcinoma, and NY-ESO-1 of metastatic carcinomas.
  • viruses could use conjugate suppressive factors of T lymphocytes as one of methods to escape from host's immuno-protection. It is thought that a part of virus infection could attribute the escape function of such viruses and administration of the composition of the present invention could improve the immune reaction of T lymphocytes to viruses.
  • composition for infection treatment of the present invention is effective on treatment for, for example, human hepatitis virus (hepatitis B, Hepatitis C, hepatitis A, or hepatitis E), human retrovirus, human immunodeficiency virus(HIV1, HIV2), human T leukemia virus (HTLV1, HTLV2), or human lymphocytic cell type virus.
  • human hepatitis virus hepatitis B, Hepatitis C, hepatitis A, or hepatitis E
  • human retrovirus human immunodeficiency virus(HIV1, HIV2)
  • HTLV1, HTLV2 human T leukemia virus
  • lymphocytic cell type virus for example, human lymphocytic cell type virus.
  • herpes virus type 1 or 2 simple herpes virus type 1 or 2, epstein-barr virus, cytomegalovirus, varicella-zoster virus, human herpesvirus including human herpesvirus 6, poliovirus, measles virus, rubella virus, Japanese encephalitis virus, mumps virus, influenza virus, adenovirus, enterovirus, rhinovirus, virus developing severely acute respiratory syndrome (SARS), ebola virus, west nile virus, or these virus modified artificially.
  • simple herpes virus type 1 or 2 simple herpes virus type 1 or 2
  • epstein-barr virus cytomegalovirus
  • varicella-zoster virus human herpesvirus including human herpesvirus 6, poliovirus, measles virus, rubella virus, Japanese encephalitis virus, mumps virus, influenza virus, adenovirus, enterovirus, rhinovirus, virus developing severely acute respiratory syndrome (SARS), ebola virus, west nile virus, or these virus modified artificial
  • pathogenesis protozoan for example, trypanosome, malaria, and toxoplasma
  • bacillus for example, mycobacterium, salmonella, and listeria
  • fungus for example, candida
  • the composition for infection treatment of the present invention can be used together with existing anti-HIV drugs, antiviral agents, antibiotic agents, antimicrobial agents, or visceral mycosis medicines or be made as a mixture with them.
  • the anti-HIV drugs include, for example, reverse transcriptase inhibitors (for example, AZT, ddl, 3TC, and d4T), protease inhibitors (for example, saquinavir mesylate, ritonavir, nelfinavir mesylate, amprenavir, delavirdine mesylate, saquinavir, and lopinavir/ritonavir) or CCR5 receptor antagonists.
  • the antiviral agents include, for example, anti-herpesvirus agents, anti-influenza virus agents, interferon- ⁇ and ⁇ , or various immunoglobulins.
  • composition for infection treatment of the present invention can be used together with vaccines of virus or pathogen or be made as a formulation with them.
  • vaccines include, for example, poliovaccine, measles vaccine, Japanese encephalitis vaccine, BCG vaccine, triple vaccine, mumps virus vaccine, varicella virus vaccine, influenza vaccine, hepatitis A vaccine, hepatitis B vaccine, and cholera vaccine.
  • composition of the present invention is usually administered systemically or locally, and orally or parenterally.
  • the dosage is determined depending on medicines used for the present invention, age, body weight, symptom, therapeutic effect, administration route, and duration of the treatment, etc.
  • the dosage range from 1 ⁇ g to 100 mg per an adult is orally administered once to several times per day, or the dosage range from 0.1 ng to 10 mg per an adult is administered once to several times per day parenterally, suitably intravenously, and is intravenously administered for 1 to 24 hours per day continuously.
  • a concomitant drug of the composition of the present invention and other medicines When a concomitant drug of the composition of the present invention and other medicines is administered, it is used as solid medicines for internal use, and injections, external preparations, suppositoriums, inhalant, pernasal preparation, etc. for parenteral administration.
  • the solid medicines for oral administration include compressed tablets, pills, capsules, dispersing powders, granules, etc.
  • the capsules include hard capsules and soft capsules.
  • the tablets include sublingual tablets, intraoral strapping tablets, and intraoral speed disintegrators, etc.
  • one or more active compound(s) may be pharmaceutically manufactured as itself/themselves or a formulation with excipients (lactose, mannitol, glucose, microcrystal cellulose, and starch, etc.), binders (hydroxypropylcellulose, polyvinyl pyrrolidone, and magnesium metasilicate aluminate, etc.), disintegrators (cellulose calcium glycolate, etc.), lubricants (magnesium stearate etc.), stabilizers, or solubilizers (glutamate and aspartic acid, etc.), etc. according to usual methods.
  • excipients lactose, mannitol, glucose, microcrystal cellulose, and starch, etc.
  • binders hydroxypropylcellulose, polyvinyl pyrrolidone, and magnesium metasilicate aluminate, etc.
  • disintegrators cellulose calcium glycolate, etc.
  • lubricants magnesium stearate etc.
  • stabilizers or solub
  • compositions may be optionally coated by coatings (sucrose, gelatin, hydroxypropylcellulose, and hydroxypropylmethylcellulose phthalate, etc.) or coated in the layer of two or more.
  • capsules of absorbable materials such as gelatin may be included.
  • the sublingual tablets are manufactured according to a well-known method.
  • they may be pharmaceutically manufactured as a mixture one or more active compound(s) with excipients (lactose, mannitol, glucose, microcrystal cellulose, colloidal silica, and starch, etc.), binders (hydroxypropylcellulose, polyvinylpyrrolidone, and magnesium metasilicate aluminate, etc.), disintegrator (starch, L-hydroxypropylcellulose, carboxymethylcellulose, crosscarmellose sodium, and cellulose calcium glycolate, etc.), lubricants (magnesium stearate etc.), swelling agents (hydroxypropylcellulose, hydroxypropylmethylcellulose, carbopole, carboxymethylcellulose, polyvinyl alcohol, xanthan gum, and Cyamoposis Gum, etc.), swelling adjuvants (glucose, fructose, mannitol, xylitol, erythritol, maltose, tre
  • additives such as preservatives, anti-oxidants, coloring agents, and sweeteners used regularly may be optionally added.
  • the intraoral strapping tablets are manufactured according to a well-known method.
  • they may be pharmaceutically manufactured as a mixture one or more active compound(s) with excipients (lactose, mannitol, glucose, microcrystal cellulose, colloidal silica, and starch, etc.), binders (hydroxypropylcellulose, polyvinylpyrrolidone, and magnesium metasilicate aluminate, etc.), disintegrator (starch, L-hydroxypropylcellulose, carboxymethylcellulose, crosscarmellose sodium, and cellulose calcium glycolate, etc.), lubricants (magnesium stearate etc.), swelling agents (hydroxypropylcellulose, hydroxypropylmethylcellulose, carbopole, carboxymethylcellulose, polyvinyl alcohol, xanthan gum, and Cyamoposis Gum, etc.), swelling adjuvants (glucose, fructose, mannitol, xylitol, erythritol, maltose,
  • additives such as preservatives, anti-oxidants, coloring agents, and sweeteners used regularly may be optionally added.
  • the intraoral speed disintegrators are manufactured according to a well-known method.
  • they may be pharmaceutically manufactured as one or more active compound(s) itself/themselves or a mixture bulk or granulated bulk particle with suitable coatings (ethyl cellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, and acrylic acid methacrylate copolymer, etc.), plasticizers (polyethylene glycol and triethyl citrate, etc.), excipients (lactose, mannitol, glucose, crystallite cellulose, colloidal silica, and starch, etc.), binders (hydroxypropylcellulose, polyvinyl pyrrolidone, and magnesium metasilicate aluminate, etc.), disintegrators (starch, L-hydroxypropylcellulose, carboxymethylcellulose, crosscarmellose sodium, and cellulose calcium glycolate, etc.), lubricants (magnesium stearate etc.), dispersant adjuvants (glucose, fruct
  • additives such as preservatives, anti-oxidants, coloring agents, and sweeteners used regularly may be optionally added.
  • the liquid compositions for oral administration include pharmaceutically acceptable waters, suspensions, emulsions, syrups, and elixirs, etc.
  • one or more active compound(s) may be dissolved, suspended, or emulsified to generally used diluent (purified water, ethanol or those mixing liquids, etc.).
  • those liquid medicines may contain humectants, suspending agents, emulsifying agents, sweeteners, flavor agents, flavoring agents, preservatives, and buffers, etc.
  • the external preparations for parenteral administration include, for example, ointments, gels, creams, fomentations, patchs, embrocations, aerosols, inhalants, sprays, aerosols, eye drops, and nasal drops, etc. These include one or more activator(s) and are manufactured by a well-known method or the formula usually used.
  • the ointments include one or more activator(s) and are manufactured by a well-known method or the formula usually used. For example, they are manufactured by levigating and melting one or more activator(s) to basis.
  • the ointment bases are chosen from a well-known or the one usually used. They are used mixing one or two or more kinds chosen from, for example, higher fatty acid or higher fatty acid ester (adipic acid, myristic acid, palmitic acid, stearic acid, oleic acid, adipic acid ester, myristic acid ester, palmitate, stearic acid ester, and oleic acid ester, etc.), rows (yellow wax, spermaceti, and ceresin, etc.), surfactants (polyoxyethylene alkyl ether phosphate etc.), higher alcohols (cetanol, stearyl alcohol, and cetostearyl alcohol, etc.), silicone oils (dimethylpolysiloxane etc.), hydrocarbons (hydrophilic petrolatum, white petrolatum, purified lanolin, and liquid paraffin, etc.), glycols (ethylene glycol, diethylene glycol, propylene glycol, polyethylene glycol, and macrogol
  • the gels are manufactured by a well-known method or a formula usually used. For example, they are manufactured by levigating and melting one or more activator(s) to a basis.
  • the base is chosen from a well-known or the one usually used. It is used mixing one or two or more kinds chosen from, for example, lower alcohols (ethanol and isopropyl alcohol, etc.), gelatinizers (carboxymethylcellulose, hydroxyethyl cellulose, hydroxypropylcellulose, and ethyl cellulose, etc.), neutralizers (triethanolamine and diisopropanolamine, etc.), surfactants (mono-stearic acid polyethylene glycol, etc.), gums, water, absorption enhancers, and poisoned inhibitors. Further, they may include preservatives, anti-oxidants, and flavors, etc.
  • the creams are manufactured by a well-known method or a formula usually used. For example, they are manufactured by levigating one or more activator(s) to a basis and spreading and rolling on the support after kneading.
  • the base is chosen from a well-known or the one usually used.
  • the fomentations are manufactured by a well-known method or a formula usually used. For example, they are manufactured by levigating one or more activator(s) to a basis and spreading and rolling on the support after kneading.
  • the base is chosen from a well-known or the one usually used.
  • thickeners polyacrylic acid, polyvinyl pyrrolidone, arabic gum, starch, gelatin, and methyl cellulose, etc.
  • humectants urea, glycerin, and propylene glycol, etc.
  • fillers china clay, flower of zinc, talc, calcium, and magnesium, etc.
  • they may include preservatives, anti-oxidants, and flavors, etc.
  • the patchs are manufactured by a well-known method or a formula usually used. For example, they are manufactured by levigating one or more activator(s) to a basis and spreading and rolling on the support after kneading.
  • the base for the pacth is chosen from a well-known or the one usually used. It is used mixing one or two or more kinds chosen from, for example, high molecular basis, oils, fats, higher fatty acids, tackifiers, and poisoned inhibitors. Further, they include preservatives, anti-oxidants, and flavors, etc.
  • the liniments are manufactured by a well-known method or a formula usually used. For example, they are manufactured by dissolving, suspending, or emulsifying one or more activator(s) to one or two or more kinds chosen from water, alcohols (ethanol and polyethylene glycol, etc.), higher fatty acids, glycerins, soaps, emulsifiers, suspending agents, etc. Further, they may include preservatives, anti-oxidants, and flavors, etc.
  • the aerosols, the inhalants, and the sprays may contain stabilizers such as sodium hydrogen sulfite besides the diluent generally used, buffers giving isotonicity, and isotonic agents such as, for example, sodium chloride, sodium citrate, and citrates.
  • stabilizers such as sodium hydrogen sulfite besides the diluent generally used, buffers giving isotonicity, and isotonic agents such as, for example, sodium chloride, sodium citrate, and citrates.
  • the injections for parenteral administration include solid shots that are dissolved or suspended to solution, suspension, emulsion, or time of use solvent.
  • the injections are used by dissolving, levigating and melting one or more activator(s) to the solvent.
  • the solvent for example, water for injection, distilled saline, vegetable oil, propylene glycol, polyethylene glycol, alcohols such as ethanol, etc., and these combination are used.
  • this injection may include stabilizers, solubilizers (glutamate, aspartic acid, and polysorbate 80 (registered trademark), etc.), suspending agents, emulsifying agents, soothing agents, buffers, and preservatives, etc. These are sterilize in the final process or are manufactured from the aseptic manipulation.
  • Aseptic solid medicines may be manufactured as a freeze-drying product and may be used by making to be aseptic or dissolving to aseptic distilled water for injection or other solvents before use.
  • the inhalant for parenteral administration includes aerosols, inhalant powders, or inhalant solutions, which may be used by making to dissolve or suspend to water or other suitable media at the time of use. These inhalants are manufactured according to a well-known method.
  • the inhalant solutions are prepared by properly selecting from preservatives (benzalkonium chloride and paraben, etc.), coloring agent, buffers (sodium phosphate and sodium acetate, etc.), tonicity agents (sodium chloride and concentrated glycerin, etc.), thickeners (carboxyvinyl polymer, etc.), and absorption enhancers, etc., optionally.
  • the inhalant powders are prepared by properly selecting from lubricants (stearic acid and the salt, etc.), binders (starch and dextrin, etc.), excipients (lactose and cellulose, etc.), coloring agents, preservatives (benzalkonium chloride and paraben, etc.), and absorption enhancers, etc., optionally.
  • lubricants stearic acid and the salt, etc.
  • binders starch and dextrin, etc.
  • excipients lactose and cellulose, etc.
  • coloring agents benzalkonium chloride and paraben, etc.
  • preservatives benzalkonium chloride and paraben, etc.
  • absorption enhancers etc., optionally.
  • a sprayer atomizer and nebulizer
  • an inhalation administering machine for powder is usually used.
  • compositions for parenteral administration contain one or more activator(s) and include suppository for intrarectal administration which is prescribed according to a routine procedure and pessary, etc. for intravaginal administering.
  • the sprays may contain a stabilizer such as sodium hydrogen sulfite besides the diluent generally used, a buffer giving isotonicity, and an isotonic medicine such as, for example, sodium chloride, sodium citrate, or citrates.
  • a stabilizer such as sodium hydrogen sulfite
  • an isotonic medicine such as, for example, sodium chloride, sodium citrate, or citrates.
  • FIG. 1 shows a flow cytometry that presents PD-1 expression in H-2L d -specific 2C CTL clone and PD-L1 expression of P815 (cell line derived from mastocytoma) stable transfectant clone, and (B) shows a cytotoxic activity of 2C CTL cell line to PD-L1-expressing P815 cell lines and the effect on the cytotoxic activity of anti-PD-L1 antibody (anti-PD-L1 F(ab’) 2 IgG).
  • FIG. 2 shows tumor growth and infiltrating of PD-L1-expressing P815 cell lines transplanted into syngeneic mouse.
  • A shows the tumour volume (upper) of the transplanted PD-L1-expressing P815 tumours and the survival rate (under) after the transplant
  • B shows the tissue staining view of the transplanted PD-L1-expressing P815 tumour mass in syngeneic DBA/2 mouse (a shows a view ( ⁇ 40) that presents invasion of tumor cells across the abdominal wall and the peritoneum, b shows the same view ( ⁇ 400), c shows the view of invasion into spleen, and d shows the view of invasion into liver.).
  • FIG. 3 shows in vivo effect of anti-PD-L1 antibody for the tumor growth of the transplanted PD-L1-expressing P815 cell lines in syngeneic mice.
  • A shows in vivo effect of anti-PD-L1 antibody for IFN- ⁇ production from the tumor-specific CD8+ T cells in mice
  • B shows in vivo effect of anti-PD-L1 antibody for the tumour volume (upper) and the survival rate (under) of the transplanted PD-L1-expressing P815 tumors in mice (squares show control group (rat IgG administrated group) and circles show anti-PD-L1 antibody (anti-PD-L1 F(ab′) 2 IgG) administrated group in the figure.).
  • FIG. 4 shows proliferation suppression of PD-L1-expressing B16 melanomas in homo-deficient syngeneic mice (PD-1 ⁇ / ⁇ ).
  • FIG. 5 shows in vivo effect of anti-PD-L1 antibody to tumor growth of transplanted myeloma cell lines in syngeneic mouse (BALB/c) and the involvement of PD-1.
  • A shows a flow cytometry that shows PD-L1 expression in various myeloma cell lines
  • B shows in vivo effect of anti-PD-L1 antibody for tumor growth of J558L tumors transplanted in the above mouse
  • C shows comparison of tumor growth of J558L tumors transplanted in wild type and PD-1 gene deficient (PD-1 ⁇ / ⁇ ) syngeneic mice.
  • FIG. 6 shows PD-L1 expression in capillary endothelium.
  • A shows PD-L1 and PD-L2 expression in vascular endothelial cells of mice heart .
  • B shows tissue staining of PD-L1 expression in each tissue of mice.
  • Ch means choroidea
  • CV means middle cardiac vein
  • GI means glomerule
  • Re means retina.
  • Each arrow shows vascular endothelial cells.
  • Each staining image is enlargement view ( ⁇ 40).
  • FIG. 7 shows PD-L1 expression in liver non-parenchymal cells.
  • FIG. 8 shows cell surface molecule phenotype in Kupffer cells and liver sinusoidal endothelial cells (LSECs).
  • FIG. 9 shows cell surface molecule phenotype in CD 4 positive T cells of PD-1 gene homo-deficient mice (PD-1 ⁇ / ⁇ ) or wild type mice (wt).
  • FIG. 10 shows cell surface molecule phenotype in CD8 positive T cells of PD-1 gene homo-deficient mice (PD-1 ⁇ / ⁇ ) or wild type mice (wt).
  • FIG. 11 shows the effect of PD-L1 on T-cell proliferation in LNPC.
  • A shows cellular proliferation when stimulating each naive T cells of PD-1 ⁇ / ⁇ mice and wt mice.
  • B shows the effect of anti-PD-L1 antibody on cellular proliferation in the co-culture of T cells that has already been activated and derived from PD-1 ⁇ / ⁇ mice or wt mice and LNPC.
  • FIG. 12 shows the effect of PD-L1 on cytokine production in LNPC.
  • A shows cytokine production when stimulating each naive T cell of PD-1 ⁇ / ⁇ mice and wt mice.
  • B shows the effect of anti-PD-L1 antibody on cytokine production in the co-culture of T cells that has already been activated and derived from PD-1 ⁇ / ⁇ mice or wt mice and LNPC.
  • C shows cell division of the activated T cells from PD-1 ⁇ / ⁇ mice or wt mice in the co-culture with LNPC.
  • FIG. 13 shows the involvement of PD- 1 in T lymphocytes proliferation in virus infected mouse liver.
  • Cellular proliferation of CD19-and CD3-positive lymphoid cells in liver and spleen of PD-1 ⁇ / ⁇ mice or wt mice on (A) day 0 and (B) day 7 after adenovirus infection are shown.
  • FIG. 14 shows the involvement of PD-1 to cellular proliferation of T lymphocytes in virus infected mouse liver.
  • A shows cellular proliferation of CD4-and CD8-positive lymphoid cells in liver of PD-1 ⁇ / ⁇ mice or wt mice on day 7 after adenovirus infection.
  • B shows the ratio of various proliferative lymphocytes on day 7 after the infection.
  • FIG. 15 shows the involvement of PD- 1 in virus infection.
  • (a)-(d) show tissue staining images that presents each cellular proliferation in liver of PD-1 ⁇ / ⁇ mice and wt mice on day 0 and day 7 after adenovirus infection.
  • (e) and (f) show cellular proliferation of CD4-and CD8-positive T cells in PD-1 ⁇ / ⁇ mice on day 7 after adenovirus infection.
  • FIG. 16 shows the involvement of PD-1 to virus infection.
  • (g)-(n) show hematoxylin & eosin tissue staining images in liver of PD- 1 ⁇ / ⁇ mice and wt mice on day 0 and day 7 after adenovirus infection.
  • (o)-(r) show X-gal tissue staining images in liver of PD-1 ⁇ / ⁇ mice and wt mice on day 0 and day 7 after adenovirus infection.
  • FIG. 17 shows the enhancement of each material to cytotoxic activity.
  • (a) the enhancement of anti-mouse PD-1 antibody, (b) the enhancement of anti-mouse PD-L1 antibody, (c) the enhancement of mouse PD-1 Fc, and (d) the enhancement of human PD-1 Fc are shown.
  • a mouse PD-L1 expression vector was constructed by being inserted and ligated mouse PD-L1 cDNA (Journal of Experimental Medicine (2000), vol. 19, issue 7, p. 1027-1034.) digested with restriction enzyme EcoRl to pApuroXS expression vector ( The EMBO Journal (1994), vol. 13, issue 6, p. 1341-1349.).
  • the expression vector pApuroXS-PD-L1 was transfected into P815 cells by electroporation (360V, 500 ⁇ F).
  • the P815 cells were cultured in RPMI-1640 medium including FCS (10%), 2-mercaptoethanol (10 ⁇ 5 M), and various antibiotics.
  • FIG. 1(A) shows flow cytometries presenting (i) PD-1 expression of H-2L d -specific 2C CTL clones and (ii) PD-L1 expression in PD-L1 expressing stable transformant of P815 (cell line derived from mastocytoma).
  • Transformed B16 cell lines (B16/PD-L1) stably expressing PD-L1 were cloned by a similar method ( FIG. 1(A) (iii)-(v)).
  • pEFBOSneo-PD-L1 (Nucleic Acid Research (1990), vol. 18, issue 17, p. 5322.) constructed as a expression vector by a similar method was used, and G418 (0.5 mg/ml) was used for selective culture of the cell lines.
  • cDNA coding protein to connect tandem 6 ⁇ his peptide tag (His-Tag) to 3′ end side of total length mouse PD-L1 cDNA was inserted in expression vector pVL1393 (the trade name: purchase from Clonetech) after being digested by restriction enzyme EcoRl and Notl. Then, this expression vector was continuously introduced to SF9 insect cells (purchased from Invitrogen), and the inclusion body was collected. This inclusion body virus was infected to HiFive insect cells (purchased from Invitrogen) by culturing for 2 days at 27° C.
  • the purified PD-L1 protein that are used as antigen was obtained by processing cell lysate dissolved with buffer solution (Tris-HCl (50 mM, pH7, and 1% TritonX-100), EDTA (10 mM), and NaCl (150 mM), various protease inhibitors) with Ni-sepharose column chromatography.
  • buffer solution Tris-HCl (50 mM, pH7, and 1% TritonX-100), EDTA (10 mM), and NaCl (150 mM), various protease inhibitors
  • the dialyzed PD-L1 protein was immunized to female wister rat at 8 weeks age (purchased from SLC Japan) with a complete Freund adjuvant (purchased from Amersham) and after several days, 2 ⁇ 10 8 of cells collected from peripheral lymph node were fused with the same number of SP2/0 cells using PEG1500. Further, the fused cells were selected by culturing in RPMI1640 medium (HAT (purchased from Sigma), Origen (10%, purchased from Igen), and FCS (10%), 2-mercaptoethanol (10 ⁇ 5 M), and various antibiotics), and the presence of the antibody production was confirmed by flow cytometric analysis.
  • the monoclonal antibody (1-111) to PD-L1 was obtained by purification of the collected peritoneal fluid with protein G sepharose column chromatography after transfering the established hybridoma (hybridomas recognized with International Trust Number: FERM BP-8396) to Balb/C nu/nu mice.
  • biotinated antibodies using Sulfo-NHS-LC-biotin can be used.
  • anti-human PD-1 antibody (the monoclonal antibody producted from hybridomas recognized with International Trust Number: FERM BP-8392) was prepared according to a similar method.
  • Cytotoxicity assay was executed by 51 Cr (chromium) separation assay.
  • 2C cells Journal of Immunology (1996), vol. 157, issue 2, p. 670-678.) are (H-2L) d alloreactive cytotoxic T-cells derived from 2C transgenic B6 mice.
  • Anti-PD-L1 antibody (anti-PD-L1 F(ab′) 2 ) recovered the decreased cytotoxic activity of cytotoxic T lymphocytes. Those results suggested that inhibition of PD-1 and PD-L1 signal by inhibiting PD-L1 function could reinforce cytotoxic activity to carcinoma cells.
  • the result is shown in FIG. 2(A) .
  • circles show P815 cell lines transplanted group
  • squares and triangles show PD-L1-expressing P815 cell lines transplanted group respectively.
  • histological analysis of P815/PD-L1 cells transferred group was executed.
  • FIG. 2(B) shows staining images dyed with hematoxylin & eosin after being fixed by 10% formaldehyde and embedded into paraffin.
  • a shows a view ( ⁇ 40) that presents invasion of tumor cells across the abdominal wall and the peritoneum
  • b shows the same view ( ⁇ 400)
  • c shows the view of invasion into spleen
  • d shows the view of invasion into liver.
  • P815 cells proliferation was suppressed and 30 percent of mice in this group lived by 6-7 weeks, while in PD-L1-expressing P815 cells (P815/PD-L1) transplanted group, carcinoma cells proliferation was remarkable and all mice died by 2-4 weeks ( FIG. 2(A) ).
  • P815/PD-L1 cells were observed to permeate across the peritoneal cavity and the abdominal cavity, and further those metastases to liver and spleen ( FIG. 2(B) a-d).
  • IFN-y in culture supernatant at 24 hours was measured with ELISA kit (purchased from Bioscience). The result is shown in FIG. 3(A) .
  • Anti-PD-L1 antibody restored IFN-y production from cytotoxic T lymphocytes which had been suppressed by P815/PD-L1 ( FIG. 3(A) ).
  • Administration of anti-PD-L1 antibody suppressed carcinoma cell growth, and showed the clear survival effect ( FIG. 3(B) ). This result presents that administration of anti-PD-L1 antibody is effective on cancer treatment.
  • Vascular endothelial cells (hereafter, abbreviated as ECs) was gathered from mouse heart by the method of Marelli-Berg ( Journal of immunological methods (2000), vol. 244, issue 1-2, p. 205-215.). Concretely, heart tissue digested by collagenase was pre-cultured followed by being pre-cultured with mouse Ig and co-cultured together with FITC-modified anti-CD3 antibody, the same modified anti-CD105 antibody, the same modified anti-isolectin B4 antibody, and anti-FITC bead. These vascular endothelial cells were purified by positive selection using Magnetic-activated cell-sorting separation columns (the trade name: purchased from Miltenyi Biotec).
  • PD-L1 and PD-L2 expressions in the gathered vascular endothelial cells were confirmed by flow cytometry.
  • the cell labelling was executed using anti-PD-L1 antibody (the antibody name: 1-111), anti-PD-L2 antibody (the antibody name: #122), and fluorescent labelling second antibody ( FIG. 6(A) ).
  • the analysis was executed 10,000 events with Facscalibur (the equipment name: purchased from Becton Dickinson) using CellQuest software (purchased from Dickinson).
  • Facscalibur the equipment name: purchased from Becton Dickinson
  • CellQuest software purchased from Dickinson
  • PD-L1 or PD-L2 expression is shown as shedding curve, and control Ig is shown as filling curve.
  • FIG. 6(B) shows PD-L1 expression in (a) eyeball, (b) submaxillary gland, (c) heart, (d) lung, (e) liver, (f) kidney.
  • Ch presents choroid
  • CV presents vena centralis
  • GI presents glomerule
  • Re presents retina.
  • Each arrow presents vascular endothelial cells.
  • Each staining figure is 40 ⁇ enlargement view.).
  • PD-L1 expression was observed in capillary endothelium of heart, lung, kidney, stomach, small intestine, submandibular gland, eyeball, and liver. The expression in liver was localized in liver sinusoidal capillary.
  • FIG. 7(A) shows ICAM-1 expression
  • FIG. 7(B) shows PD-L1 expression
  • CV presents vena centralis.
  • TSA tyramide signal amplification
  • LNPCs were isolated from mouse liver according to the pronaseE method ( Experimental Cell Research (1976, vol. 99, p. 444-449.). Concretely, LNPCs obtained by which liver is circulated with pronaseE solution (Merck) were cultured and were separated by density gradient centrifugation. Relative distribution of Kupffer cells (CD54+, CD11bhigh) in the cell suspension is 20-25%, and that of liver perisinusoidal space endothelial cells (hereafter, abbreviated with LSECs.) (CD54+, CD11bhigh) is 75-80%.
  • Kupffer cells and LSECs were doubly stained using FITC-labeled anti-CD11b antibody, each biotinated monoclonal antibody to ICAM-1, PD-L1, B 7 -1, and B7-2, and PE-labeled Streptavidin, respectively. Kupffer cells and LSECs were gate as CD11b high and CD11b low cell, respectively ( FIG. 8 ).
  • PD-L1 expressed together with ICAM-1, B7-1, and B7-2 in Kupffer cells, while the expression was weak in LSECs ( FIG. 8 ).
  • Native T cells were refined (refining degree 90% or more) from spleen and lymphoid of PD-1 gene homo-deficient mice (PD-1 ⁇ / ⁇ ) or wild type C57BL/6 mice (wt) by a negative selection using T-cell enrichment column (the trade name: purchased from Genzyme). The cells cultured for 48 hours with 10 ⁇ g/ml of anti-CD3 monoclonal antibody (2C11) were activated.
  • the naive T cells activated by the above method were doubly stained using FITC-labeled anti-CD4 antibody or APC-labeled anti-CD8 antibody, PE-labeled anti-CD25 antibody, PE-labeled anti-CD 4 antibody, PE-labeled anti-CD69 antibody, or PE-labeled anti-CTLA-4 antibody, biotin-labeled anti-B7-1 (CD80) antibody or biotin-labeled anti-B7-2 (CD86) antibody, and anti-PD- 1 antibody (the antibody name: J43, monoclonal antibody producted from hybridomas recognized by International Trust Number FERM BP-8118) or anti-PD-L1 antibody (1-111), and each molecule expressions were analyzed by flow cytometry ( FIGS. 9 and 10 ).
  • the hybridomas identified by International Trust Number FERM BP-8118 had been deposited as Trust Number FERM P-18356 to National Institute of Advanced Industrial Science and Technology, International Patent Organism Depositary in Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, Japan (ZIP code 305-8566) at May 30, 2001, and was transferred to International Deposit at Jul. 16, 2002.
  • naive T cells activation of PD-1 gene homo-deficient mice (PD-1 ⁇ / ⁇ ) or wild type mice (wt) was executed by the method described in example 8. Proliferation of these activated cells was measured by BrdU incorporation method ( FIG. 11(A) ). The proliferation was measured by adding BrdU at the last 6 hours of 48 hours to label the cells and measuring using Proliferation ELISA kit (the trade name: Purchased from Roche). The amount of IFN- ⁇ production at this time was measured in ELISA Kit (the trade name: purchased from Genzyme) ( FIG. 12(A) ).
  • T cells derived from PD-1 gene homo-deficient mice (PD-1 ⁇ / ⁇ ) or wild type mice (wt) was beforehand activated by the method described in example 8.
  • the activated T cells were respectively cultured for 60 hours under the presence or the absence of Mitomycin C-processed LNPCs of wild type mice and under the presence or the absence of 30 ⁇ g/ml of anti-PD-L1 antibody (1-111) (rat IgG as control) and 20 ⁇ g/ml of CTLA4-Ig (Genzyme) (human IgG as control), and the cells proliferation for the last 12 hours was measured by BrdU incorporation method ( FIG. 11(B) ). Further, the amount of IFN- ⁇ production at 48 hours was measured ( FIG. 12(B) ).
  • CFSE 5-(6)-carboxy-fluorescein diacetate succinimidyl diester
  • PD-1 gene homo-deficient mice (PD-1 ⁇ / ⁇ ) or wild type mice (wt) (3 per a group) were infected in adenoviruses by intravenous administration of 10 9 ⁇ 10 1 PFU (plaque-forming units) of Ad-lacZ.
  • Ad-lacZ used here which are 5 type adenovirus with lacZ gene, lacks E1 and E3 region and can be purified by cesium chloride density-gradient centrifugation ( Nucleic Acid Research (1995), vol. 234,issue 19, p. 3816-3821.) after proliferating them in 293 cells.
  • spleen cells and intrahepatic lymphocytes that had been gathered after intravenous administration of 0.5 mg of BrdU (the trade name: purchased from Sigma) to the mice at 1 hour before slaughtering were doubly labeled by anti-BrdU antibody and anti-CD19 antibody, or anti-CD3 antibody ( FIG. 13 ).
  • FIG. 14(B) each bar graph shows the ratio of BrdU-positive cells.
  • liver slices that had been gathered after intravenous administration of 0.5 mg of BrdU (the trade name: purchased from Sigma) to the mice at 1 hour before slaughtering were doubly labeled by anti-BrdU antibody ( FIG. 15( a )-( d ), 20 ⁇ enlargement view.).
  • Liver slices of PD-1 gene homo-deficient mice (PD-1 ⁇ / ⁇ ) on day 7 after infection were doubly labeled by anti-BrdU antibody, anti-CD19 antibody, anti-CD3 antibody, anti-CD4 antibody, and anti-CD8 antibody ( FIG. 15( e ), ( f ), 40 ⁇ enlargement view.).
  • Liver slices of PD-1 gene homo-deficient mice (PD-1 ⁇ / ⁇ ) or wild type mice (wt) on day 7 or 30 after infection were stained by hematoxylin & eosin ( FIG. 16( g )-( j ), 20 ⁇ enlargement view, (k)-(n), 40 ⁇ enlargement view.) and X-gal ( FIG. 16( o )-( r ), 40 ⁇ enlargement view.).
  • P-815/PD-L1 cells compellingly expressing mouse PD-L1 were sown into culture flask and were cultured in usual medium including 5 ⁇ g/mL of puromycin (purchased from Sigma)(hereafter, abbreviated as selective medium) at 37° C. under 5% CO 2 /95% air until to be 50% -90% confluent.
  • Mouse cytotoxic T lymphocyte 2C cells were subcultured for several days in usual medium together with P-815 cells processed by MMC (Mitomycin C) and culture supernatant of ConA-stimulated rat splenic cells.
  • the collected P-815/PD-L1 cells were cultured for 15 minutes after adding 3 ⁇ L of BATDA Reagent of DELFIA EuTDA Cytotoxicity Reagents (purchased from PerkinElmer). Then, they were washed with PBS. 2C cells subcultured for 5-8 days after adding P-815 cells were used.
  • 20 ⁇ L(10 ng/mL) of anti-mouse PD-1 antibody ( FIG. 17 , anti-mPD-1Ab(J43)), anti-mouse PD-L1 antibody (anti-mPD-L1Ab(1-111) in this figure), mouse PD-1 Fc (mPD-1Fc in this figure), human PD-1 Fc (hPD-1Fc in this figure), mouse IgG2a ⁇ (Control Ig in this figure), or PBS were dispensed into 96 well plate, and then 50 ⁇ L of P-815/PD-L1 cells or usual medium were added. Further, 50 ⁇ L of 2C cells, usual medium, or usual medium including 1% Triton ⁇ 100 were added.
  • the evaluated group is composed of a subject substrate, P-815/PD-L1 cells, and 2C cells
  • the high control group is composed of PBS, P-815/PD-L1 cells, and usual medium including 1% Triton ⁇ 100
  • the low control group is composed of PBS, P-815/PD-L1 cells, and usual medium
  • the 2C cell control group is composed of PBS, usual medium, and 2C cells
  • the background group is composed of PBS, P-815/PD-L1 cells, and usual medium.
  • CTL activity (%) was calculated by the following formula. All values are the one that had been subtracted by the average of the background.
  • CTL activity (%) ([the measurement of the evaluation group] ⁇ [the measurement of the 2 C cell control group] ⁇ [the measurement of the low control group])/([the measurement of the high control group] ⁇ [the measurement of the low control group]) ⁇ 100
  • Anti-PD-1 antibody, anti-PD-L1 antibody, and PD-1 Fc have significantly reinforced CTL activity ( FIG. 17( a )-( d ), E: T ratio presents the mixing ratio of 2C cells and PD-L1/P815 cells.).
  • the inhibiting effect of anti-PD-1 antibody on cancer metastasis was evaluated by intraperitoneal administration of anti-mouse PD-1 monoclonal antibody to C57BLJ6 mice to which B16 melanoma cells had been transferred at intervals of 2 days followed by measuring the liver weight on day 18 after transfer.
  • liver weight of anti-PD-1 antibody administrated group was significantly suppressed than that in control IgG administrated control group (liver weight/carcinoma cell non-transferred group: 1.3 g, decrease from control group: 6.8 g to anti-PD-1 antibody administrated group: 3.5 g.).
  • the suppression of the increase in this weight presents that the metastasis of B16 melanoma cells is suppressed.

Abstract

Compositions for cancer or infection treatment via immunopotentiation caused by inhibition of immunosuppressive signal induced by PD-1, PD-L1, or PD-L2 and therapies using them, immunopotentiative substrates included as the active ingredient, screening methods of the substrates for cancer or infection treatment, cell lines used for the screening methods, evaluation methods that selects the substrates for cancer treatment, and carcinoma cell transplanted mammals used for the evaluation methods. The compositions of the present invention that inhibits the function of PD-1, PD-L1, or PD-L2 are useful for cancer or infection treatment.

Description

    TECHNICAL FIELD
  • The present invention relates to immunopotentiation characterized by inhibiting immunosuppressive signals induced by PD-1, PD-L1 or PD-L2, compositions for cancer or infection treatment, and therapies that use them.
  • More specifically, the present invention relates to the compositions for cancer or infection treatment through an immunopotentiation caused by inhibition of immunosuppressive signals induced by PD-1, PD-L1 or PD-L2, the therapies that use them, screening methods of immunopotentiative substances, or substances for cancer treatment or for infection treatment that are contained in the composition as an active ingredient, cell lines used for those screening methods, an evaluation that selects the substances for cancer treatment, and carcinoma cells transplanted mammals used for the evaluation.
  • BACKGROUND ART
  • Immunotherapies can reduce side reactions that can't be avoided easily in most chemotherapies, and is expected as a therapy with extremely high specificity. The Immunotherapies can be executed in the purpose to recover patient's QOL by activating the immune reaction that human originally has by an extrinsic method and subrogating a part of the load by medication.
  • Immunopotentiation can be executed by methods of activating immune reaction of T lymphocytes. It is said that not only stimulation through antigen receptors (TCR) but also an additionally stimulative inducement through conjugated stimulative molecular groups (for example, CD28) could be necessary for the activation of T cells. However, it is reported that as the molecular groups with homologous structures to the conjugated stimulative molecular groups, CTLA-4 and PD-1 were discovered recently and give signals that suppress signals of antigen receptors (TCR). It is thought that a method of activating T cells could be an effective mean to suprress the function of those conjugated control molecules.
  • PD-1 was cloned as 55 kD of I type membrane protein that belong to an immunoglobulin family (The EMBO Journal (1992), vol. 11, issue 11, p. 3887-3895, JP5336973, JP7291996). Human PD-1 cDNA is composed of the base sequence shown in EMBL/GenBank Acc. No. NM005018 and mouse PD-1 cDNA is composed of the base sequence shown in Acc. No. X67914, and those expression are observed when thymus cells differentiate from CD4-CD8-cell into CD4+CD8+ cell (International Immunology (1996), vol. 18, issue 5, p. 773-780., Journal of Experimental Medicine (2000), vol. 191, issue 5, p. 891-898.). It is reported that PD-1 expression in periphery is observed in myeloid cells including T cells or B lymphocytes activated by stimulation from antigen receptors, or activated macrophages (International Immunology (1996), vol. 18, issue 5, p. 765-772.).
  • In an.intracellular domain of PD-1, there are ITIM motifs (Immunoreceptor Tyrosine-based Inhibitory Motif) that could been thought to be a repression domain to immune reaction. Since PD-1-deficient mice develop lupus-like autoimmune disease such as glomerular nephritis and arthritis (for C57BL/6 gene background.) (International Immunology (1998), vol.10, issue 10, p. 1563-1572., Immunity (1999), vol. 11, issue 2, p. 141-151.) and a disease like dilated cardiomyopathy (for BALB/c gene background.)(Science (2001), vol. 291, issue 5502, p. 319-332.), it has been suggested that PD-1 could be a control factor of development of autoimmune disease, especially the peripheral self-tolerance.
  • PD-L1 (human PD-L1 cDNA is composed of the base sequence shown by EMBUGenBank Acc. No. AF233516 and mouse PD-L1 cDNA is composed of the base sequence shown by NM021893.) that is a ligand of PD-1; is expressed in so-called antigen-presenting cells such as activated monocytes and dendritic cells (Journal of Experimental Medicine (2000), vol. 19, issue 7, p. 1027-1034.). These cells present interaction molecules that induce a variety of immuno-inductive signals to T lymphocytes, and PD-L1 is one of these molecules that induce the inhibitory signal by PD-1. It has been revealed that PD-L1 ligand stimulation suppressed the activation (cellular proliferation and induction of various cytokine production) of PD-1 expressing T lymphocytes. PD-L1 expression has been confirmed in not only immunocompetent cells but also a certain kind of tumor cell lines (cell lines derived from monocytic leukemia, cell lines derived from mast cells, cell lines derived from hepatic carcinomas, cell lines derived from neuroblasts, and cell lines derived from breast carcinomas) (Nature Immunology (2001), vol. 2, issue 3, p. 261-267.).
  • Though PD-L2 (human PD-L2 cDNA is composed of the base sequence shown by EMBUGenBank Acc. No. NM025239 and mouse PD-L2 cDNA is composed of the base sequence shown by NM021896.) had been identified as a second ligand of PD-1, it has been reported that the expression and function are almost same as PD-L1 (Nature Immunology (2001), vol. 2, issue 3, p. 261-267.).
  • It has been thought that the inhibitory signals from the conjugated suppressive molecules represented by PD-1 could control abnormal immune reaction to autoantigen and immunological tolerance in lymphocyte generation or maturation by a mechanism that appropriately controls positive signals with antigen receptors (TCR) and conjugated stimulation molecules. It has been thought that a certain kind of tumour and virus could use those conjugated suppressive molecules to intercepte the activation and proliferation of T cells and weaken the host immunity reaction to oneself by a direct or indirect mechanism (Cell (1992), vol. 71, issue 7, p. 1093-1102, Science(1993), vol. 259, issue 5093, p. 368-370.). It has been thought that those conjugated suppressive molecules could have caused the impairment of T cells in a part of disease thought to originate in impairment of T cells.
  • DISCLOSURE OF THE INVENTION
  • A problem of the present invention is to provide compositions to activate immunity by inhibiting the inhibitory signals of PD-1, PD-L1 or PD-L2 and compositions for cancer or infection treatment through this mechanism.
  • The present inventors paid attention to PD-1, PD-L1, or PD-L2 as a new target in cancer or infection treatment and found that substances that inhibit the inhibitory signals of PD-1, PD-L1 or PD-L2 inhibit cancer proliferation through the mechanism of the recovery and activation of immune function. Further, they found that PD-1 signal, concretely, an interaction of PD-1 and PD-L1 or PD-1 and PD-L2 took part in the exclusion of infectious virus. According to those facts, they found the substances that could inhibit the inhibitory signals of PD-1, PD-L1 or PD-L2 having therapeutic potential for cancer or infection and completed the present invention.
  • That is, the present invention relates to
    • 1. an immunopotentiative composition containing an immunosuppressive signal inhibitor of PD-1, PD-L1 or PD-L2,
    • 2. a composition for cancer treatment containing the immunosuppressive signal inhibitor of PD-1, PD-L1 or PD-L2,
    • 3. the composition for cancer treatment of the subsection 2, which is a composition that suppresses cancer metastasis,
    • 4. a composition for infection treatment containing an immunosuppressive signal inhibitor of PD-1, PD-L1 or PD-L2,
    • 5. the composition for cancer treatment of the subsection 2 or 3, which is characterized by acting through immunopotentiation,
    • 6. the composition for infection treatment of the subsection 4, which are characterized by acting through immunopotentiation,
    • 7. the composition of either of the subsection 1 to 6, which the immunosuppressive signal inhibitor is/are one or more selected from an interaction inhibitor of PD-1 and PD-L1 or PD-1 and PD-L2, an intracellular signaling inhibitor of PD-1, and an inhibitory substance of PD-1, PD-L1 or PD-L2 production,
    • 8. the composition of the subsection 7, which is/are one or more of the interaction inhibitor(s) of PD-1 and PD-L1 selected from PD-1; antibody, PD-L1 antibody, soluble PD-1, and soluble PD-L1,
    • 9. the composition of the subsection 8, which is PD-1 antibody selected from anti-human PD-1 antibody of which hybridomas identified by International Trust Number FERM BP-8392 product, anti-PD-1 antibody humanized from non-human antibody, and human type anti-human PD-1 antibody,
    • 10. the composition of either of the subsection 1 to 6, which the immunosuppressive signal inhibitor is a lymphocyte of which PD-1 expression is inhibited by gene-recombination,
    • 11. the composition of the subsection 7, which the interaction inhibitor of PD-1 and PD-L1 or PD-1 and PD-L2, an intracellular signaling inhibitor of PD-1, or the inhibitory substance of PD-1, PD-L1 or PD-L2 production is one or more of substance(s) selected from protein, polypeptide or peptide, polynucleotide or polynucleoside, antibody or the derivative, organic synthesis compound, inorganic compound, and natural product,
    • 12. an immunopotentiative method containing a method of administering the immunosuppressive signal inhibitor of PD-1, PD-L1 or PD-L2,
    • 13. a method for cancer treatment containing a method of administering the immunosuppressive signal inhibitor of PD-1, PD-L1 or PD-L2,
    • 14. the method of cancer treatment of the subsection 13, which is a method of suppressing cancer metastasis,
    • 15. a method for infection treatment containing a method of administering the immunosuppressive signal inhibitor of PD-1, PD-L1 or PD-L2,
    • 16. the method for cancer treatment of the subsection 13 or 14, which is characterized by acting through immunopotentiation,
    • 17. the method for infection treatment of the subsection 15, which is characterized by acting through immunopotentiation,
    • 18. the method of either of the subsection 12 to 17, which the immunosuppressive signal inhibitor is/are one or more selected from the interaction inhibitor of PD-1 and PD-L1 or PD-1 and PD-L2, the intracellular signaling inhibitor of PD-1, and the inhibitory substance of PD-1, PD-L1 or PD-L2 production,
    • 19. the method of the subsection 18, which the interaction inhibitor is/are one or more selected from PD-1 antibody, PD-L1 antibody, soluble PD-1, and soluble PD-L1,
    • 20. the method of the subsection 19, which PD-1 antibody is an antibody selected from anti-human PD-1 antibody of which hybridomas identified by international trust number FERM BP-8392 product, anti-PD-1 antibody humanized from non-human antibody, and human type anti-human PD-1 antibody,
    • 21. the method of either of the subsection 12 to 17, which the immunosuppressive signal inhibitor is a lymphocyte of which PD-1 expression is inhibited by gene-recombination,
    • 22. the method of the subsection 18, which the interaction inhibitor of PD-1 and PD-L1 or PD-1 and PD-L2, the intracellular signaling inhibitor of PD-1, or the inhibitory substance of PD-1, PD-L1 or PD-L2 production are one or more of substance(s) selected from protein, polypeptide or peptide, polynucleotide or polynucleoside, antibody or the derivative, organic synthesis compound, inorganic compound, and natural product,
    • 23. use of the immunosuppressive signal inhibitor of PD-1, PD-L1 or PD-L2 to manufacture the immunopotentiative composition,
    • 24. use of the immunosuppressive signal inhibitor of PD-1, PD-L1 or PD-L2 to manufacture the composition for cancer treatment,
    • 25. use of the substance of the subsection 24, which the composition for cancer treatment is the composition for suppression of cancer metastasis,
    • 26. use of the immunosuppressive signal inhibitor of PD-1, PD-L1 or PD-L2 to manufacture the composition for infection treatment,
    • 27. carcinoma cell lines for screening, which are transformed to express PD-L1 or PD-L2, 28. a screening method for the immunopotentiative substance characterized by touching the cells of the subsection 27 to lymphocytes and a subject substance, followed by evaluating the enhancement of the subject substance for immune reaction of lymphocytes to the cells of the subsection 27 ,
    • 29. a screening method for a substance for cancer treatment characterized by touching the carcinoma cells of the subsection 27 to lymphocytes and a subject substance, followed by evaluating the enhancement of the subject substance for immune reaction of lymphocytes to carcinoma cells and inhibitory effect on the subject substance for carcinoma cells proliferation,
    • 30. a screening method for a substance for infection treatment characterized by touching the infected cells of the subsection 27 to lymphocytes and a subject substance, followed by evaluating the enhancement of the subject substance for immune reaction of lymphocytes to the infected cells and inhibitory effect on the subject substances for pathogens proliferation,
    • 31. a mammal created by transplant the carcinoma cell lines of the subsection 27, and
    • 32. a screening method for a substance for cancer treatment characterized by administering the subject substance to the mammal of of the subsection 31, followed by evaluating the inhibitory ratio of the subject substance for the transplanted carcinoma cells proliferation.
  • PD-1, PD-L1, or PD-L2 of the present invention includes each one derived from mammal, for example, mouse, rat, hamster, guinea pig, dog, pig, ape, or primate including human. They are suitable to be human PD-L1, PD-1, and PD-L2, respectively.
  • The immunosuppressive signal of PD-1, PD-L1, or PD-L2 in the present invention is at least composed of the interaction of PD-1 and PD-L1 or PD-1 and PD-L2, and the intracellular signalings of PD-1. Production of PD-1, PD-L1 or PD-L2 itself is included in them.
  • The immunosuppressive signal of PD-1, PD-L1, or PD-L2 in the present invention is inhibited by direct or indirect inhibition of the interaction of PD-L1 or PD-1 and PD-L2 or the intracellular signalings of PD-1. A substance that selectively binds to PD-1, PD-L1, or PD-L2 respectively is included as a substance with those inhibitory activities. For example, it is suitable to be protein, polypeptide or peptide, polynucleotide or polynucleoside, antibody or the derivative, organic synthesis compound, inorganic compound, or natural product. Especially, an antibody to PD-1, PD-L1 or PD-L2 is enumerated as an excellent substance in specificity.
  • The immunosuppressive signals are inhibited by inhibiting production of PD-1, PD-L1 or PD-L2 itself.
  • As an antibody to PD-1, PD-L1 or PD-L2, all antibodies derived from human, mouse, rat, rabbit, or goat which can inhibit the immunosuppressive signals by PD-1, PD-L1, or PD-L2, those polyclonal or monoclonal antibodies, complete or shorten (for example, F(ab′) 2, Fab′, Fab, or Fv fragment) antibodies, chimeric antibodies, humanized antibodies, or completely humanized antibodies will be acceptable.
  • Such antibodies can be manufactured using a partial protein of the extracellular region of PD-1, PD-L1, or PD-L2 as an antigen according to well-known production methods of antibody or antiserum. The partial protein of the extracellular region can be prepared by well-known protein expression and purification techniques.
  • The polyclonal antibodies can be manufactured according to well-known methods. For example, they can be manufactured by separation and refinement of the antibody of which a mixture of an antigen and a carrier protein is immunized to suitable animal, and an antibody inclusion to the antigen is gathered from the immunized animal. As such animal, mouse, rat, sheep, goat, rabbit, and guinea pig are generally enumerated. To improve the antibody producibility, Freund's complete adjuvant or Freund's incomplete adjuvant can be administered with the antigen. The administering is usually executed once every two weeks about 3-10 times in total. The polyclonal antibody can be gathered from the immunized animal's blood and peritoneal fluid, etc. by the above method. The measurement of the polyclonal antibody's titer in antiserum can be measured by ELISA. The separation and refinement of the polyclonal antibody can be executed by refining techniques that use active adsorbents such as antigen binding solid phase, protein A, or protein G, etc., salting-out, alcohol precipitation, isoelectric precipitation, electrophoresis, adsorption and desorption with ion exchanger, ultracentrifugation, or separation and refinement of immunoglobulins such as gel filtration technique, etc.
  • As an antibody preparation, the monoclonal antibody or the modifier is more suitable.
  • The monoclonal antibody producing cells can be prepared as hybridomas to be possible to subculture which produce the monoclonal antibody by selecting the individual of which the antibody titre is confirmed in an antigen immunized animals, gathering the spleen or the lymph node on day 2-5 after the final immunization, and fusing the antibody producting cells included in them with homogeneous or heterozoic myeloma cells. The antigen itself or with the carrier and the diluent is administered to the part in which the antibody production is possible. To improve the antibody producibility, Freund's complete adjuvant or Freund's incomplete adjuvant can be administered with the antigen. According to the method of calling “DNA immunization”, animals are immunized. This method is a method using a phenomenon in which antigen-expressing vectors are introduced into the part and are taken into myocytes on the process of tissue repair, and expresses the antigenic protein (Nature Immunology (2001), vol. 2, issue 3, p. 261-267) after Cardiotoxin is treated to immune animal's tibialis anterior muscle of hind leg.
  • As an immune animal, mouse, rat, sheep, goat, rabbit, or guinea pig can be used, mouse and rat are suitable. The fusion operation can be executed by the method (Nature (1975), vol. 256, issue 5517, p. 495-497.) of Kohler and Milstein, and as fusion accelerants, polyethylene glycol (PEG) and Sendai virus, etc. are used. As those myeloma cells, myeloma cells such as P3U1, NS1, SP2/0, and AP1 can be used, P3U1 are often used usually. The monoclonal antibody producing cells can be selected by detecting by ELISA, etc. executed by adding hybridoma culture supernatant to solid phase in which antigenic proteins are adsorbed direct or with carrier perhaps. Hybridoma culture supernatant's antibody titre can be measured by ELISA. The separation and refinement of the monoclonal antibody can be executed according to the separation refining method similar to the separation and refinement of immunoglobulin for the above polyclonal antibody. Concretely, it is anti-mouse PD-L1 antibody producted by hybridomas identified by International Trust Number FERM BP-8396 or anti-human PD-1 antibody producted by a hybridoma identified by International Trust Number FERM BP-8392.
  • The hybridomas identified by International Trust Number FERM BP-8392 had been deposited as Trust Number FERM P-19162 to National Institute of Advanced Industrial Science and Technology, International Patent Organism Depositary in Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, Japan (ZIP code 305-8566) at Dec. 19, 2002, and had been transferred to International Deposit at Jun. 5, 2003. The hybridomas identified by International Trust Number FERM BP-8396 had been deposited as Trust Number FERM P-18908 at Jun. 25, 2002, and had been transferred to International Deposit at Jun. 11, 2003.
  • The antibody fragment means F(ab′)2, Fab′, Fab, or scFv antibody fragment and can be prepared by reducing optionally after processing with protease enzyme.
  • F(ab′)2 antibody fragments can be purified by either method of affinity chromatography such as ion-exchange chromatography, gel filtration, or protein A or protein G column, etc. after the purified monoclonal antibody is completely digested by pepsin. Because digestive time by pepsin is different depending on Ig subtype, it is necessary to prepare it suitably. Fab′ antibody fragment can be prepared by reducing F(ab′)2 by 2-mercaptoethylamine partly. Fab antibody fragment can be prepared by the direct digestion under the presence of cysteine by the digestive enzyme papain followed by refining.
  • Further, the monoclonal antibody is prepared as a rearrangement antibody and a hybrid antibody modified by gene recombination technology using DNA sequence that codes the amino acid sequence of antibody isolated from the hybridomas. For example, it can be prepared as a single-chain antibody but not a usual complete type antibody. scFv antibody (single chain Fv) can be prepared by the method of Jost (Journal of Biological Chemistry (1994), vol. 269, issue 42, p. 26267-26273.). The single-chain antibody with the characteristic and the affinity of an original antibody can be prepared by making expression an expression vector including a fused DNA of which DNA fragments respectively coding a variable region of the heavy chain and the light chain had been connected with a spacer coding neurtal amino acids (glycine or serine) in suitable host cells.
  • When non-human antibody is used to treat for human, it is indispensable to decrease the antigenicity of the antibody. Since the immune reaction to patient's antibody often shortens an effective treatment period, the process of decreasing the antigenicity of the antibody by making the antibody humanize or completely human type is necessary. The humanized antibody modified to be acceptable for administering to human is the antibody which is modified so that the decrease of antigenicity or the blood movement may improve to extent that can be allowed in pharmacology when the antibody is administered to human.
  • Human PD-1 antibody or human PD-L1 antibody in specification of the present invention includes the humanized or the complete human type antibody, too.
  • The humanized antibody can be prepared by substituting a part of non-human antibody which was prepared by being immunized to mammals other than human for a part of human antibody. Concretely, it has been known to be able to be prepared by constructing a chimera with a gene coding a constant region of the human antibody (Proc. Natl. Acad. Sci. (USA) (1987), vol. 84, p. 3439-3443, Journal of Immunology (1987), vol. 139, issue 1, p. 3521.). The DNA sequence of the human constant region has been described to a prior art and the constant region gene can be easily obtained from an already-known clone. Then, the DNA sequence coding the variable region of the antibody can be fused to the human constant region. An isotype of the human constant region can be selected due to desire effective function or antibody dependent cytotoxicity. A suitable isotype is IgG1, IgG3, and IgG4. Both human light-chain constant region, K chain, and A chain can be used. It is possible to make this humanized chimeric antibody expression by a routine method.
  • The complete human type antibody can be prepared by using mice (XenoMouse (Chemical Biology (2000), vol. 7, issue 8, p. R185-6.), HuMAb-Mouse (Infection and Immunity (2002), vol. 70, issue 2, p. 612-9.), TC mouse (Biotechnology and Genetics Enginnering Revew (2002),vol. 19, p. 73-82.), and KM mouse (Cloning Stem Cells (2002), vol. 4, issue 1, p. 91-102.)) of which a constant region gene of human immunoglobulin have been transferred, and a target antibody can be mass-produced by making the antibody production lymphocytes separated from mice to hybridomas. It can be prepared by phage display method (FEBS Letter (1998), vol. 441, p. 20-24.). In this method, by using phages of which the human antibody gene have been incorporated into a cyclic single strand DNA, the human type antibody can be expressed on the surface of the phage as a form fused with coat protein of the phages.
  • Polypeptides or the derivatives that bind to PD-1, PD-L1, or PD-L2 include each partial proteins of PD-1, PD-L1 or PD-L2 of which the immunosuppressive signal is not induced. The presence of PD-1 in the neighborhood of the antigen receptors is indispensable for the inducement of the immunosuppressive signal of PD-1, for that purpose it is necessary to be restrained by the interaction with PD-L1 or PD-L2 in antigen-presenting cells, tumours, or carcinoma cells. Therefore, soluble PD-L1 or soluble PD-L2 with a part which is only extracellular domains and interacts with PD-1can inhibit the immunosuppressive signal of PD-1. On the other hand, soluble PD-1 with a part which has a similar structure and can interact with PD-L1 or PD-L2 can inhibit the immunosuppressive signal. These soluble proteins have only to include an extracellular region which is necessary and sufficient to bind to PD-1, PD-L1, or PD-L2 and can be prepared by a well-known expression and refining technique.
  • If an interaction inhibitor of PD-1 and PD-L1 or PD-1 and PD-L2 is a protein or a polypeptide and an essential area for the interaction is composed by only consecutive polypeptide, such a polypeptide fragment can become a mutual antagonist. Further, an antagonist with stronger activity can be identified from molecular groups of which this polypeptide fragment is chemically modified, or designed by computer based on the spatial structure of the polypeptide fragment. Also, the best antagonist can be more efficiently selected from molecular groups designed by computer based on protein stereoanalysis data of the interaction area.
  • A substance that inhibits the interaction of PD-1 and PD-L1, or PD-1 and PD-L2 can be screened directly. Such a substance can be identified from each libraries of protein, polypeptide or peptide, polynucleotide or polynucleoside, non-peptide compound, organic synthesis compound, or natural product (for example, fermentational product, cell extract, plant extract, and animal tissue extract.).
  • Since the inhibitory signals of PD-1 intracellular domain are caused by contacting dephosphorylation enzymes (for example, SHP-1 and 2 (Sathish J. G., Journal of Immunology (2001), vol. 166, issue 3, p. 1763-70.)) that bind to ITIM in PD-1 intracellular domain with an intracellular complex including an antigen receptor complex, they are generally inhibited by inhibiting the contact between the antigen receptor complex and PD-1. The substance that inhibits the inhibitory signals includes a substance that directly inhibits the activity of dephosphorylation enzymes, a substance that inhibit phosphorylation of tyrosine residue in ITIM, a substance that inhibits bonds of dephosphorylation enzymes to ITIM, or a substance that directly inhibits activity of dephosphorylation enzymes, etc. The antigen receptor complex includes T cell receptor complex or B cell receptor complex.
  • The production of PD-1, PD-L1, or PD-L2 is inhibited by specific polynucleotide or polynucleoside, organic synthesis compound, inorganic compound, or natural product, etc. Especially, the suitable polynucleotide or polynucleoside includes antisense nucleotide derivative so-called ribozyme. This uses a mechanism of which the expressed mRNA is destroyed by which the polynucleotide derivative that is complement to mRNA of PD-1, PD-L1, or PD-L2 is transferred into subject cells. Further, the vector can be used for the gene manipulation to lymphocyte stem cells gathered from a patient so that it inhibits the expression of PD-1, and the manipulated cells can be used for cell medical treatment of which the cells will be proliferated, differentiated, activated, and administered to the patient again. Especially, in immunotherapy for cancer, more specific and clonal lymphocytes to target cells can be prepared by adding a specific antigen of the target cells on the process of the maturation and activation of lymphocyte stem cells.
  • The screening method of the present invention can be executed by a method of measuring cell function. The carcinoma cell lines for screening of which PD-L1 or PD-L2 used by the method is transformed to be expressed includes carcinoma cell lines transformed transitory or stably after expression vectors constructed to express PD-L1 or PD-L2 have been introduced to the cells by well-known methods. As the carcinoma cell lines used, ape COS-1 cells, COS-7 cells, Vero, chinese hamster CHO cells (hereafter, abbreviated with CHO cells.), dhfr gene deficient chinese hamster CHO cells (hereafter, abbreviated with CHO (dhfr-) cells.), mouse L cells, mouse AtT-20, mouse myeloma cells, rat GH3, HEK293 T cells, and human FL cells, etc. are used. Especially, the transformation of animal cells can be executed according to methods described in, for example, Cell Tehcnology separate volume 8, New Experimental Protocol of Cell Technology (1995), vol. 263, published by SHUJUNSHA Co., Ltd., and Virology (1973), vol. 52, issue 456 .
  • Cells naturally expressing PD-L1 or PD-L2 can be also used. Such cells include leukocyte, suitablely, monocytes, macrophages or antigen-presenting cells, epithelial cells, tumor cells, carcinoma cells, or those cell lines, etc. As tumor cells or carcinoma cells, for example, P38D1 cells, P815 cells, NB41A3 cells, MDA-231 cells, SKBR-3 cells, MCF-7 cells, BT474 cells, J558L cells, P3U1 cells, PAI cells, X63 cells, or SP2/0 cells can be used. Cells infected with pathogens, which express PD-L1 or PD-L2 naturally or compellingly, can be used. Infectious pathogens include human hepatitis virus (hepatitis B, hepatitis C, and hepatitis A) or hepatitis E), human retrovirus, human immunodeficiency virus (HIV1 and HIV2), human T cell leukemia virus, human T lymphocytic virus (HTLV1 and HTLV2), simple herpes virus type 1 or 2, Epstein-Barr virus, cytomegalovirus, varicella-zoster virus, human herpesvirus including human herpesvirus 6, poliovirus, measles virus, rubella virus, Japanese encephalitis virus, mumps virus, influenza virus, adenovirus, enterovirus, rhinovirus, virus developing severely acute respiratory syndrome (SARS), Ebola virus, West Nile virus, or these virus modified artificially. Other pathogens include, for example, pathogenesis protozoan (for example, trypanosome, malaria, and toxoplasma), bacillus (for example, mycobacterium, salmonella, and listeria) or fungus (for example, candida), etc.
  • The lymphocytes used in the screening method of the present invention are T or B lymphocytes, and suitably cytotoxic T lymphocytes (CTL). The immune reaction of lymphocytes in the screening method of the present invention includes citotoxic response (for example, tumor immunity reaction), mixed lymphocyte reaction, production of cytokines, antibodies, complements or other cell surface antigens, or cellular proliferation, etc.
  • In the present invention, the screening method of an active ingredient contained in the composition for immunopotentiation or cancer treatment can be executed by measuring cytotoxic activity of cytotoxic T lymphocytes against subject cells followed by measuring an effect of a subject substance against the activity. This method is an assay of recovery and reinforcement of cytotoxic activity by adding the subject substance to a culture mixed cytotoxic T lymphocytes (CTL) naturally expressing PD-1 or cell lines (for example, 2C cells) with cells naturally or compulsorily expressing PD-L1 or PD-L2 which are derived from syngeneic mouse. Since the cytotoxic activity against PD-L1 or PD-L2 expressing cells is lower than that against PD-L1 or PD-L2 non-expressing cells, this method has a feature of which the recovery of cytotoxic activity (rising part) due to the subject substance can be measured more clearly. The recovery of cytotoxicity due to the subject substance can be evaluated as an equivalent to inhibition of suppression of cytotoxicity. Further, it is preferable that the the cytotoxicity due to the subject substance is arbitrarily measured. As these cells, tumor cell lines or carcinoma cell lines naturally expressing PD-L1 or PD-L2 (Nature Immunology (2001), vol. 2, issue 3, p. 261-267.), for example, P38D1 cells, P815 cells, NB41A3 cells, MDA-231 cells, SKBR-3 cells, MCF-7 cells, BT474 cells, J558 L cells, P3U1 cells, PAI cells, X63 cells, or SP2/0 cells can be used, and tumor cell lines or carcinoma cell lines transformed so as to stably or transiently express PD-L1 or PD-L2 can be also used.
  • On the other hand, it is preferable that the cytotoxic lymphocytes are PD-1 expressing cells derived from syngeneic animal to targeted cells.
  • In the present invention, the screening method of an active ingredient contained in the composition for infection treatment is an assay of enhancement effect on immune reaction of lymphocytes to infected cells or inhibitory effect on proliferation activity of pathogen or virus by adding the subject substance to a culture mixed cytotoxic T lymphocytes (CTL) or cell lines (for example, 2C cells) naturally expressing
  • PD-1 with cells naturally or compulsorily expressing PD-L1 or PD-L2 which are derived from syngeneic mouse and was infected with pathogen or virus.
  • Further, in evaluations using a similar principle, a mammal created by transplanting the above carcinoma cell lines for screening which are transformed so as to express PD-L1 or PD-L2 or cells naturally expressing PD-L1 or PD-L2 to a syngeneic mammal can be used. To a manufacture method of the mammal, a process of transplanting cells and a process of breeding the mammal until becoming appropriate for the evaluation is indispensable. This evaluation is characterized in evaluating proliferation of the transplant cells and the amount of production of various cytokines or cell surface antigens, especially, in case of carcinoma cells, proliferation, permeation, or histological analysis of metastasis of the cells, or survival rates of the transplanted mammal. The cellular proliferation can be evaluated as the number of carcinoma cells per unit capacity in case of ascites tumors or blood cancers, or the size or the weight after removing in case of solid cancer. The effect of the subject substance for cancer treatment in this method can be evaluated as an equivalent to effect due to inhibition of suppression of cytotoxicity caused in PD-L1 or PD-L2. As such cells, syngeneic cells to a mammal for transplant, with good proliferation are more suitable. The mammal include s primates except human, mouse, rat, hamster, guinea pig, dog, pig, and ape.
  • INDUSTRIAL AVAILABILITY
  • As a substance that demonstrates remarkable suppression of cancer proliferation and life prolongation of an individual by administering to the carcinoma cells-transplanted animal model, the present inventors invented each specific antibody (anti-PD-1 antibody and anti-PD-L1 antibody) that inhibited PD-1 and PD-L1 function respectively. These antibodies showed actions that recover or reinforce cytotoxic activity that has relatively decreased by presenting PD-L1 ligand to PD-1-expressing CTL (cytotoxic T lymphocyte) (example 1 and FIG. 1.). This suggests that the cytotoxic activity to carcinoma cells by CTL could be reinforced by administering these antibodies. In the carcinoma cells-transplanted animal model (Protein, Nucleic acid , and Enzyme (1981), vol. 26, issue 3, p. 208-22.) which uses syngeneic mouse of which cell lines artificially expressing PD-L1 derived from mastocytomas have been imported, administration of anti-PD-L1 antibody presented suppression of proliferation, invasion, and metastasis of carcinoma cells, and life prolongation of an individual (FIGS. 2 and 3.). It was suggested that inhibition of PD-1 function or production could achieve an effect similar to the effect on inhibition of PD-L1 function by this antibody. This is based on non-proliferation of imported carcinoma cells in the cancer imported model using PD-1-deficient mice, and presents that inhibition of PD-1 function or production could be effective on cancer treatment (Example 5 and FIG. 3.).
  • Actually, it had been proven that administration of anti-PD-1 antibody significantly suppressed metastasis of imported carcinoma cells to liver in the cancer imported animal model (Example 13).
  • The inventors presented that the substance that inhibits the immunosuppressive signal induced by PD-1, PD-L1 or PD-L2 was useful for infection treatment (Example 11, FIGS. 15 and 16).
  • These results experimentally presented by the present inventors prove that only PD-1 antibody or PD-L1 antibody don't presents aforementioned effect, but also any substance that can inhibit the immunosuppressive signal from PD-1, PD-L1, or PD-L2 present almost similar effect. The substances with such effects include, for example, anti-PD-L2 antibody, soluble PD-1, soluble PD-L1, soluble PD-L2, PD-1 antagonists, PD-L1 antagonists, PD-L2 antagonists, substances that inhibits interaction between PD-1 and PD-L1 or PD-1 and PD-L2, PD-1 production inhibitors, PD-L1 production inhibitors, PD-L2 production inhibitors, and intracellular inhibitory signal inhibitors by PD-1.
  • Cancer or tumour of which the therapeutic potential could be expected by administration of the composition for cancer treatment of the present invention include, for example, carcinoma, squamous carcinoma (for example, cervical canal, eyelid, tunica conjunctiva, vagina, lung, oral cavity, skin, urinary bladder, tongue, larynx, and gullet), and adenocarcinoma (for example, prostate, small intestine, endometrium, cervical canal, large intestine, lung, pancreas, gullet, intestinum rectum, uterus, stomach, mammary gland, and ovary). Further, they include sarcomata (for example, myogenic sarcoma), leukosis, neuroma, melanoma, and lymphoma.
  • The effect on the ones that remarkably express PD-L1 or PD-L2 in these cancers or tumours is remarkable. PD-L1 or PD-L2 expression can be identified by checking up surgically excised cancer, tumour mass, or lesioned part gathered outside of the body as a sample. Administration of the composition of the present invention will become an efficient and available method as a treatment after surgery for tumour or cancer patient that PD-L1 or PD-L2 is remarkably expressed. PD-L1 or PD-L2 expression can be identified by, for example, immunochemical method using PD-L1 antibody or PD-L2 antibody, RT-PCR, or DNA array.
  • A side reaction violently decreasing lymphocytes proliferation in chemotherapy and radiotherapy for cancer is inevitable. Administration of the composition of the present invention presents an effect on stimulating and proliferating the decreased lymphocytes and can suppress a fierce side reaction accompanied by usual chemotherapy to minimum. Further, it is similar as to radiotherapy. Concomitant use with the composition of the present invention can greatly decrease the dose of chemotherapy drug or the exposure dose of irradiation from the dose or the exposure dose used usually.
  • The composition for cancer treatment of the present invention can be used together with existing chemotherapy drugs or be made as a mixture with them. Such a chemotherapy drug include, for example, alkylating agents, nitrosourea agents, antimetabolites, antitumor antibiotics, alkaloids derived from plant, topoisomerase inhibitors, hormone therapy medicines, hormone antagonists, aromatase inhibitors, P-glycoprotein inhibitors, platinum complex derivatives, other immunotherapeutic drugs, and other anticancer agents. Further, they can be used together with hypoleukocytosis (neutrophil) medicines that are cancer treatment adjuvant, thrombopenia medicines, antiemetic drugs, and cancer pain medicines for patient's QOL recovery or be made as a mixture with them.
  • The composition for cancer treatment of the present invention can be used together with immunopotentiative substances or be made as a mixture with them. Such immunopotentiative substances include, for example, various cytokines and a tumor antigen, etc. Cytokines that stimulate immune reactions include, for example, GM-CSF, M-CSF, G-CSF, interferon-α, β, γ, IL-1, IL-2, IL-3, and IL-12, etc. B7 ligand derivatives, anti-CD3 antibodies and anti-CD28 antibodies, and anti-CTLA-4 antibodies can also improve the immune reactions.
  • Administration of the tumor antigen can also improve a specific immune reaction against T lymphocytes against carcinoma cells and additionally or synergistically reinforce by using together with the composition for cancer treatment of the present invention. The tumor antigen can be prepared as a purified protein in case of an already-known gene, or as a lysate of carcinoma cells in case of an unknown gene.
  • Such the tumor antigen includes, for example, HLA-A1 and HLA-A2 restrictive peptides of malignant melanoma MAGE-1 or MAGE-3, MART-1, and gp100. Further, they include HER2/neu peptide of mammary carcinomas and ovarian carcinomas, MUC-1 peptide of adenocarcinoma, and NY-ESO-1 of metastatic carcinomas.
  • It has been thought that viruses could use conjugate suppressive factors of T lymphocytes as one of methods to escape from host's immuno-protection. It is thought that a part of virus infection could attribute the escape function of such viruses and administration of the composition of the present invention could improve the immune reaction of T lymphocytes to viruses.
  • Administration of the composition for infection treatment of the present invention is effective on treatment for, for example, human hepatitis virus (hepatitis B, Hepatitis C, hepatitis A, or hepatitis E), human retrovirus, human immunodeficiency virus(HIV1, HIV2), human T leukemia virus (HTLV1, HTLV2), or human lymphocytic cell type virus. Further, it is thought to be effective on treatment for simple herpes virus type 1 or 2, epstein-barr virus, cytomegalovirus, varicella-zoster virus, human herpesvirus including human herpesvirus 6, poliovirus, measles virus, rubella virus, Japanese encephalitis virus, mumps virus, influenza virus, adenovirus, enterovirus, rhinovirus, virus developing severely acute respiratory syndrome (SARS), ebola virus, west nile virus, or these virus modified artificially.
  • It is thought to be also effective on infection treatment for other pathogens such as, for example, pathogenesis protozoan (for example, trypanosome, malaria, and toxoplasma), bacillus (for example, mycobacterium, salmonella, and listeria) or fungus (for example, candida), etc.
  • The composition for infection treatment of the present invention can be used together with existing anti-HIV drugs, antiviral agents, antibiotic agents, antimicrobial agents, or visceral mycosis medicines or be made as a mixture with them. The anti-HIV drugs include, for example, reverse transcriptase inhibitors (for example, AZT, ddl, 3TC, and d4T), protease inhibitors (for example, saquinavir mesylate, ritonavir, nelfinavir mesylate, amprenavir, delavirdine mesylate, saquinavir, and lopinavir/ritonavir) or CCR5 receptor antagonists. The antiviral agents include, for example, anti-herpesvirus agents, anti-influenza virus agents, interferon-αand β, or various immunoglobulins.
  • The composition for infection treatment of the present invention can be used together with vaccines of virus or pathogen or be made as a formulation with them. Such vaccines include, for example, poliovaccine, measles vaccine, Japanese encephalitis vaccine, BCG vaccine, triple vaccine, mumps virus vaccine, varicella virus vaccine, influenza vaccine, hepatitis A vaccine, hepatitis B vaccine, and cholera vaccine.
  • The composition of the present invention is usually administered systemically or locally, and orally or parenterally.
  • The dosage is determined depending on medicines used for the present invention, age, body weight, symptom, therapeutic effect, administration route, and duration of the treatment, etc. For oral administration, generally, the dosage range from 1 μg to 100 mg per an adult is orally administered once to several times per day, or the dosage range from 0.1 ng to 10 mg per an adult is administered once to several times per day parenterally, suitably intravenously, and is intravenously administered for 1 to 24 hours per day continuously.
  • Since the dosage changes depending on various conditions as described above, there are cases in which doses lower or greater than the above dosage may be used.
  • When a concomitant drug of the composition of the present invention and other medicines is administered, it is used as solid medicines for internal use, and injections, external preparations, suppositoriums, inhalant, pernasal preparation, etc. for parenteral administration.
  • The solid medicines for oral administration include compressed tablets, pills, capsules, dispersing powders, granules, etc. The capsules include hard capsules and soft capsules. The tablets include sublingual tablets, intraoral strapping tablets, and intraoral speed disintegrators, etc.
  • As to such the solid medicines, one or more active compound(s) may be pharmaceutically manufactured as itself/themselves or a formulation with excipients (lactose, mannitol, glucose, microcrystal cellulose, and starch, etc.), binders (hydroxypropylcellulose, polyvinyl pyrrolidone, and magnesium metasilicate aluminate, etc.), disintegrators (cellulose calcium glycolate, etc.), lubricants (magnesium stearate etc.), stabilizers, or solubilizers (glutamate and aspartic acid, etc.), etc. according to usual methods. Further, they may be optionally coated by coatings (sucrose, gelatin, hydroxypropylcellulose, and hydroxypropylmethylcellulose phthalate, etc.) or coated in the layer of two or more. Further, capsules of absorbable materials such as gelatin may be included.
  • The sublingual tablets are manufactured according to a well-known method. For example, they may be pharmaceutically manufactured as a mixture one or more active compound(s) with excipients (lactose, mannitol, glucose, microcrystal cellulose, colloidal silica, and starch, etc.), binders (hydroxypropylcellulose, polyvinylpyrrolidone, and magnesium metasilicate aluminate, etc.), disintegrator (starch, L-hydroxypropylcellulose, carboxymethylcellulose, crosscarmellose sodium, and cellulose calcium glycolate, etc.), lubricants (magnesium stearate etc.), swelling agents (hydroxypropylcellulose, hydroxypropylmethylcellulose, carbopole, carboxymethylcellulose, polyvinyl alcohol, xanthan gum, and Cyamoposis Gum, etc.), swelling adjuvants (glucose, fructose, mannitol, xylitol, erythritol, maltose, trehalose, phosphate salt, citrate, silicate, glycine, glutamate, and arginine, etc.), stabilizers, solubilizers (polyethylene glycol, propylene glycol, glutamate, and aspartic acid, etc.), or spices (orange, strawberry, mint, lemon, and vanilla, etc.), etc. according to usual methods. Further, they may be optionally coated by coatings (sucrose, gelatin, hydroxypropylcellulose, and hydroxypropylmethylcellulose phthalate, etc.) or coated in the layer of two or more. Additives such as preservatives, anti-oxidants, coloring agents, and sweeteners used regularly may be optionally added.
  • The intraoral strapping tablets are manufactured according to a well-known method. For example, they may be pharmaceutically manufactured as a mixture one or more active compound(s) with excipients (lactose, mannitol, glucose, microcrystal cellulose, colloidal silica, and starch, etc.), binders (hydroxypropylcellulose, polyvinylpyrrolidone, and magnesium metasilicate aluminate, etc.), disintegrator (starch, L-hydroxypropylcellulose, carboxymethylcellulose, crosscarmellose sodium, and cellulose calcium glycolate, etc.), lubricants (magnesium stearate etc.), swelling agents (hydroxypropylcellulose, hydroxypropylmethylcellulose, carbopole, carboxymethylcellulose, polyvinyl alcohol, xanthan gum, and Cyamoposis Gum, etc.), swelling adjuvants (glucose, fructose, mannitol, xylitol, erythritol, maltose, trehalose, phosphate salt, citrate, silicate, glycine, glutamate, and arginine, etc.), stabilizers, solubilizers (polyethylene glycol, propylene glycol, glutamate, and aspartic acid, etc.), or spices (orange, strawberry, mint, lemon, and vanilla, etc.), etc. according to usual methods. Further, they may be optionally coated by coatings (sucrose, gelatin, hydroxypropylcellulose, and hydroxypropylmethylcellulose phthalate, etc.) or coated in the layer of two or more. Additives such as preservatives, anti-oxidants, coloring agents, and sweeteners used regularly may be optionally added.
  • The intraoral speed disintegrators are manufactured according to a well-known method. For example, they may be pharmaceutically manufactured as one or more active compound(s) itself/themselves or a mixture bulk or granulated bulk particle with suitable coatings (ethyl cellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, and acrylic acid methacrylate copolymer, etc.), plasticizers (polyethylene glycol and triethyl citrate, etc.), excipients (lactose, mannitol, glucose, crystallite cellulose, colloidal silica, and starch, etc.), binders (hydroxypropylcellulose, polyvinyl pyrrolidone, and magnesium metasilicate aluminate, etc.), disintegrators (starch, L-hydroxypropylcellulose, carboxymethylcellulose, crosscarmellose sodium, and cellulose calcium glycolate, etc.), lubricants (magnesium stearate etc.), dispersant adjuvants (glucose, fructose, mannitol, xylitol, erythritol, maltose, trehalose, phosphate salt, citrate, silicate, glycine, glutamate, and arginine, etc.), stabilizers, solubilizer (polyethylene glycol, propylene glycol, glutamate, and aspartic acid, etc.),spices (orange, Strawberry, Mint, lemon, and vanilla, etc.) etc. according to usual methods. Further, they may be optionally coated by coatings (sucrose, gelatin, hydroxypropylcellulose, and hydroxypropylmethylcellulose phthalate, etc.) or coated in the layer of two or more. Additives such as preservatives, anti-oxidants, coloring agents, and sweeteners used regularly may be optionally added.
  • The liquid compositions for oral administration include pharmaceutically acceptable waters, suspensions, emulsions, syrups, and elixirs, etc. As to such liquid medicines, one or more active compound(s) may be dissolved, suspended, or emulsified to generally used diluent (purified water, ethanol or those mixing liquids, etc.). Further, those liquid medicines may contain humectants, suspending agents, emulsifying agents, sweeteners, flavor agents, flavoring agents, preservatives, and buffers, etc.
  • The external preparations for parenteral administration include, for example, ointments, gels, creams, fomentations, patchs, embrocations, aerosols, inhalants, sprays, aerosols, eye drops, and nasal drops, etc. These include one or more activator(s) and are manufactured by a well-known method or the formula usually used.
  • The ointments include one or more activator(s) and are manufactured by a well-known method or the formula usually used. For example, they are manufactured by levigating and melting one or more activator(s) to basis.
  • The ointment bases are chosen from a well-known or the one usually used. They are used mixing one or two or more kinds chosen from, for example, higher fatty acid or higher fatty acid ester (adipic acid, myristic acid, palmitic acid, stearic acid, oleic acid, adipic acid ester, myristic acid ester, palmitate, stearic acid ester, and oleic acid ester, etc.), rows (yellow wax, spermaceti, and ceresin, etc.), surfactants (polyoxyethylene alkyl ether phosphate etc.), higher alcohols (cetanol, stearyl alcohol, and cetostearyl alcohol, etc.), silicone oils (dimethylpolysiloxane etc.), hydrocarbons (hydrophilic petrolatum, white petrolatum, purified lanolin, and liquid paraffin, etc.), glycols (ethylene glycol, diethylene glycol, propylene glycol, polyethylene glycol, and macrogol, etc.), vegetable oils (castor oil, olive oil, sesame oil, and oil of turpentine, etc.), animal oils (mink oil, yolk oil, squalane, and squalene, etc.), water, absorption enhancer, and poisoned inhibitor. Further, they may include moisturizing agents, preservatives, stabilizing agents, anti-oxidants, and flavors, etc.
  • The gels are manufactured by a well-known method or a formula usually used. For example, they are manufactured by levigating and melting one or more activator(s) to a basis. The base is chosen from a well-known or the one usually used. It is used mixing one or two or more kinds chosen from, for example, lower alcohols (ethanol and isopropyl alcohol, etc.), gelatinizers (carboxymethylcellulose, hydroxyethyl cellulose, hydroxypropylcellulose, and ethyl cellulose, etc.), neutralizers (triethanolamine and diisopropanolamine, etc.), surfactants (mono-stearic acid polyethylene glycol, etc.), gums, water, absorption enhancers, and poisoned inhibitors. Further, they may include preservatives, anti-oxidants, and flavors, etc.
  • The creams are manufactured by a well-known method or a formula usually used. For example, they are manufactured by levigating one or more activator(s) to a basis and spreading and rolling on the support after kneading. The base is chosen from a well-known or the one usually used. It is used mixing one or two or more kinds chosen from, for example, higher fatty acid esters, lower alcohols, hydrocarbons, polyhydric alcohols (propylene glycol, 1,3-butylene glycol, etc.), higher alcohols (2-hexyl decanol and cetanol, etc.), and emulsifiers (polyoxyethylene alkyl ethers and fatty acid esters, etc.), water, absorption enhancers, and poisoned inhibitors. Further, they may include preservatives, anti-oxidants, and flavors, etc.
  • The fomentations are manufactured by a well-known method or a formula usually used. For example, they are manufactured by levigating one or more activator(s) to a basis and spreading and rolling on the support after kneading. The base is chosen from a well-known or the one usually used. It is used mixing one or two or more kinds chosen from, for example, thickeners (polyacrylic acid, polyvinyl pyrrolidone, arabic gum, starch, gelatin, and methyl cellulose, etc.), humectants (urea, glycerin, and propylene glycol, etc.), and fillers (china clay, flower of zinc, talc, calcium, and magnesium, etc.), water, absorption enhancers, and poisoned inhibitors. Further, they may include preservatives, anti-oxidants, and flavors, etc.
  • The patchs are manufactured by a well-known method or a formula usually used. For example, they are manufactured by levigating one or more activator(s) to a basis and spreading and rolling on the support after kneading. The base for the pacth is chosen from a well-known or the one usually used. It is used mixing one or two or more kinds chosen from, for example, high molecular basis, oils, fats, higher fatty acids, tackifiers, and poisoned inhibitors. Further, they include preservatives, anti-oxidants, and flavors, etc.
  • The liniments are manufactured by a well-known method or a formula usually used. For example, they are manufactured by dissolving, suspending, or emulsifying one or more activator(s) to one or two or more kinds chosen from water, alcohols (ethanol and polyethylene glycol, etc.), higher fatty acids, glycerins, soaps, emulsifiers, suspending agents, etc. Further, they may include preservatives, anti-oxidants, and flavors, etc.
  • The aerosols, the inhalants, and the sprays may contain stabilizers such as sodium hydrogen sulfite besides the diluent generally used, buffers giving isotonicity, and isotonic agents such as, for example, sodium chloride, sodium citrate, and citrates.
  • The injections for parenteral administration include solid shots that are dissolved or suspended to solution, suspension, emulsion, or time of use solvent. The injections are used by dissolving, levigating and melting one or more activator(s) to the solvent. As the solvent, for example, water for injection, distilled saline, vegetable oil, propylene glycol, polyethylene glycol, alcohols such as ethanol, etc., and these combination are used. Further, this injection may include stabilizers, solubilizers (glutamate, aspartic acid, and polysorbate 80 (registered trademark), etc.), suspending agents, emulsifying agents, soothing agents, buffers, and preservatives, etc. These are sterilize in the final process or are manufactured from the aseptic manipulation. Aseptic solid medicines may be manufactured as a freeze-drying product and may be used by making to be aseptic or dissolving to aseptic distilled water for injection or other solvents before use.
  • The inhalant for parenteral administration includes aerosols, inhalant powders, or inhalant solutions, which may be used by making to dissolve or suspend to water or other suitable media at the time of use. These inhalants are manufactured according to a well-known method.
  • For example, the inhalant solutions are prepared by properly selecting from preservatives (benzalkonium chloride and paraben, etc.), coloring agent, buffers (sodium phosphate and sodium acetate, etc.), tonicity agents (sodium chloride and concentrated glycerin, etc.), thickeners (carboxyvinyl polymer, etc.), and absorption enhancers, etc., optionally.
  • The inhalant powders are prepared by properly selecting from lubricants (stearic acid and the salt, etc.), binders (starch and dextrin, etc.), excipients (lactose and cellulose, etc.), coloring agents, preservatives (benzalkonium chloride and paraben, etc.), and absorption enhancers, etc., optionally.
  • When the inhalant solution is administered, a sprayer (atomizer and nebulizer) is usually used. When the inhalant powder is administered, an inhalation administering machine for powder is usually used.
  • Other compositions for parenteral administration contain one or more activator(s) and include suppository for intrarectal administration which is prescribed according to a routine procedure and pessary, etc. for intravaginal administering.
  • The sprays may contain a stabilizer such as sodium hydrogen sulfite besides the diluent generally used, a buffer giving isotonicity, and an isotonic medicine such as, for example, sodium chloride, sodium citrate, or citrates. Production methods of the sprays have been described in, for example, U.S. Pat. No. 2,868,691 specification and U.S. Pat. No. 3,095,355 specification in detail.
  • BRIEF DESCRIPTION OF DRAWINGS
  • FIG. 1 (A) shows a flow cytometry that presents PD-1 expression in H-2Ld-specific 2C CTL clone and PD-L1 expression of P815 (cell line derived from mastocytoma) stable transfectant clone, and (B) shows a cytotoxic activity of 2C CTL cell line to PD-L1-expressing P815 cell lines and the effect on the cytotoxic activity of anti-PD-L1 antibody (anti-PD-L1 F(ab’)2 IgG).
  • FIG. 2 shows tumor growth and infiltrating of PD-L1-expressing P815 cell lines transplanted into syngeneic mouse. (A) shows the tumour volume (upper) of the transplanted PD-L1-expressing P815 tumours and the survival rate (under) after the transplant, (B) shows the tissue staining view of the transplanted PD-L1-expressing P815 tumour mass in syngeneic DBA/2 mouse (a shows a view (×40) that presents invasion of tumor cells across the abdominal wall and the peritoneum, b shows the same view (×400), c shows the view of invasion into spleen, and d shows the view of invasion into liver.).
  • FIG. 3 shows in vivo effect of anti-PD-L1 antibody for the tumor growth of the transplanted PD-L1-expressing P815 cell lines in syngeneic mice. (A) shows in vivo effect of anti-PD-L1 antibody for IFN-γ production from the tumor-specific CD8+ T cells in mice, (B) shows in vivo effect of anti-PD-L1 antibody for the tumour volume (upper) and the survival rate (under) of the transplanted PD-L1-expressing P815 tumors in mice (squares show control group (rat IgG administrated group) and circles show anti-PD-L1 antibody (anti-PD-L1 F(ab′)2 IgG) administrated group in the figure.).
  • FIG. 4 shows proliferation suppression of PD-L1-expressing B16 melanomas in homo-deficient syngeneic mice (PD-1−/−).
  • FIG. 5 shows in vivo effect of anti-PD-L1 antibody to tumor growth of transplanted myeloma cell lines in syngeneic mouse (BALB/c) and the involvement of PD-1. (A) shows a flow cytometry that shows PD-L1 expression in various myeloma cell lines, (B) shows in vivo effect of anti-PD-L1 antibody for tumor growth of J558L tumors transplanted in the above mouse, and (C) shows comparison of tumor growth of J558L tumors transplanted in wild type and PD-1 gene deficient (PD-1−/−) syngeneic mice.
  • FIG. 6 shows PD-L1 expression in capillary endothelium. (A) shows PD-L1 and PD-L2 expression in vascular endothelial cells of mice heart . (B) shows tissue staining of PD-L1 expression in each tissue of mice. Each PD-L1 expression in (a): eyeball, (b): submandibular gland, (c): heart, (d): lungs, (e): liver, (f): kidney is shown. In the figure, Ch means choroidea, CV means middle cardiac vein, GI means glomerule, and Re means retina. Each arrow shows vascular endothelial cells. Each staining image is enlargement view (×40).
  • FIG. 7 shows PD-L1 expression in liver non-parenchymal cells.
  • FIG. 8 shows cell surface molecule phenotype in Kupffer cells and liver sinusoidal endothelial cells (LSECs).
  • FIG. 9 shows cell surface molecule phenotype in CD4 positive T cells of PD-1 gene homo-deficient mice (PD-1−/−) or wild type mice (wt).
  • FIG. 10 shows cell surface molecule phenotype in CD8 positive T cells of PD-1 gene homo-deficient mice (PD-1−/−) or wild type mice (wt).
  • FIG. 11 shows the effect of PD-L1 on T-cell proliferation in LNPC. (A) shows cellular proliferation when stimulating each naive T cells of PD-1−/−mice and wt mice. (B) shows the effect of anti-PD-L1 antibody on cellular proliferation in the co-culture of T cells that has already been activated and derived from PD-1−/−mice or wt mice and LNPC.
  • FIG. 12 shows the effect of PD-L1 on cytokine production in LNPC. (A) shows cytokine production when stimulating each naive T cell of PD-1−/−mice and wt mice. (B) shows the effect of anti-PD-L1 antibody on cytokine production in the co-culture of T cells that has already been activated and derived from PD-1−/−mice or wt mice and LNPC. (C) shows cell division of the activated T cells from PD-1−/−mice or wt mice in the co-culture with LNPC.
  • FIG. 13 shows the involvement of PD-1 in T lymphocytes proliferation in virus infected mouse liver. Cellular proliferation of CD19-and CD3-positive lymphoid cells in liver and spleen of PD-1−/−mice or wt mice on (A) day 0 and (B) day 7 after adenovirus infection are shown.
  • FIG. 14 shows the involvement of PD-1 to cellular proliferation of T lymphocytes in virus infected mouse liver. (A) shows cellular proliferation of CD4-and CD8-positive lymphoid cells in liver of PD-1−/−mice or wt mice on day 7 after adenovirus infection. (B) shows the ratio of various proliferative lymphocytes on day 7 after the infection.
  • FIG. 15 shows the involvement of PD-1 in virus infection. In this figure, (a)-(d) show tissue staining images that presents each cellular proliferation in liver of PD-1 −/−mice and wt mice on day 0 and day 7 after adenovirus infection. (e) and (f) show cellular proliferation of CD4-and CD8-positive T cells in PD-1−/−mice on day 7 after adenovirus infection.
  • FIG. 16 shows the involvement of PD-1 to virus infection. In this figure, (g)-(n) show hematoxylin & eosin tissue staining images in liver of PD-1 −/−mice and wt mice on day 0 and day 7 after adenovirus infection. (o)-(r) show X-gal tissue staining images in liver of PD-1−/−mice and wt mice on day 0 and day 7 after adenovirus infection.
  • FIG. 17 shows the enhancement of each material to cytotoxic activity. In this figure, (a) the enhancement of anti-mouse PD-1 antibody, (b) the enhancement of anti-mouse PD-L1 antibody, (c) the enhancement of mouse PD-1 Fc, and (d) the enhancement of human PD-1 Fc are shown.
  • BEST MODE FOR CARRYING OUT INVENTION
  • The following example explains the present invention more concretely, but do not limit the range of the present invention.
  • EXAMPLE 1
  • A mouse PD-L1 expression vector was constructed by being inserted and ligated mouse PD-L1 cDNA (Journal of Experimental Medicine (2000), vol. 19, issue 7, p. 1027-1034.) digested with restriction enzyme EcoRl to pApuroXS expression vector (The EMBO Journal (1994), vol. 13, issue 6, p. 1341-1349.). The expression vector pApuroXS-PD-L1 was transfected into P815 cells by electroporation (360V, 500 μF). The P815 cells were cultured in RPMI-1640 medium including FCS (10%), 2-mercaptoethanol (10−5M), and various antibiotics. The P815 cell lines which stably expressed mouse PD-L1 can be cloned by subculturing as resistant cells under medium including antibiotic puromycin (3 μg/ml). PD-L1 expression can be confirmed by flow cytometric analysis. FIG. 1(A) shows flow cytometries presenting (i) PD-1 expression of H-2Ld-specific 2C CTL clones and (ii) PD-L1 expression in PD-L1 expressing stable transformant of P815 (cell line derived from mastocytoma). Transformed B16 cell lines (B16/PD-L1) stably expressing PD-L1 were cloned by a similar method (FIG. 1(A) (iii)-(v)). pEFBOSneo-PD-L1 (Nucleic Acid Research (1990), vol. 18, issue 17, p. 5322.) constructed as a expression vector by a similar method was used, and G418 (0.5 mg/ml) was used for selective culture of the cell lines.
  • cDNA coding protein to connect tandem 6× his peptide tag (His-Tag) to 3′ end side of total length mouse PD-L1 cDNA was inserted in expression vector pVL1393 (the trade name: purchase from Clonetech) after being digested by restriction enzyme EcoRl and Notl. Then, this expression vector was continuously introduced to SF9 insect cells (purchased from Invitrogen), and the inclusion body was collected. This inclusion body virus was infected to HiFive insect cells (purchased from Invitrogen) by culturing for 2 days at 27° C. The purified PD-L1 protein that are used as antigen was obtained by processing cell lysate dissolved with buffer solution (Tris-HCl (50 mM, pH7, and 1% TritonX-100), EDTA (10 mM), and NaCl (150 mM), various protease inhibitors) with Ni-sepharose column chromatography.
  • The dialyzed PD-L1 protein was immunized to female wister rat at 8 weeks age (purchased from SLC Japan) with a complete Freund adjuvant (purchased from Amersham) and after several days, 2×10 8 of cells collected from peripheral lymph node were fused with the same number of SP2/0 cells using PEG1500. Further, the fused cells were selected by culturing in RPMI1640 medium (HAT (purchased from Sigma), Origen (10%, purchased from Igen), and FCS (10%), 2-mercaptoethanol (10−5M), and various antibiotics), and the presence of the antibody production was confirmed by flow cytometric analysis. The monoclonal antibody (1-111) to PD-L1 was obtained by purification of the collected peritoneal fluid with protein G sepharose column chromatography after transfering the established hybridoma (hybridomas recognized with International Trust Number: FERM BP-8396) to Balb/C nu/nu mice. As antibodies used by flow cytometry, etc., biotinated antibodies using Sulfo-NHS-LC-biotin (the trade name: purchased from Pierce) can be used.
  • Further, anti-human PD-1 antibody (the monoclonal antibody producted from hybridomas recognized with International Trust Number: FERM BP-8392) was prepared according to a similar method.
  • Cytotoxicity assay was executed by 51Cr (chromium) separation assay. 2C cells (Journal of Immunology (1996), vol. 157, issue 2, p. 670-678.) are (H-2L)d alloreactive cytotoxic T-cells derived from 2C transgenic B6 mice. After mixing 2C cells (E: effector) together with 51Cr-labeled P815 cells (T: target) (circle), three kinds of PD-L1-expressing P815 cells (P815/PD-L1) (square, diamond, and triangle) respectively, or further under the presence of 10 mg/ml rat anti-PD-L1 F(ab′)2 IgG (filled triangle), a result of measuring separated 51Cr for 4 hours with various E/T ratio is shown in FIG. 1 (B).
  • Anti-PD-L1 antibody (anti-PD-L1 F(ab′)2) recovered the decreased cytotoxic activity of cytotoxic T lymphocytes. Those results suggested that inhibition of PD-1 and PD-L1 signal by inhibiting PD-L1 function could reinforce cytotoxic activity to carcinoma cells.
  • EXAMPLE 2
  • The tumor growth and the survival rate of mice was evaluated by hypodermically transfering 1×10 6 of P815 cells (n=6) or P815/PD-L1 cells (n=6) to syngeneic DBA/2 mice respectively. The result is shown in FIG. 2(A). In this figure, circles show P815 cell lines transplanted group, and squares and triangles show PD-L1-expressing P815 cell lines transplanted group respectively. Further, histological analysis of P815/PD-L1 cells transferred group was executed. FIG. 2(B) shows staining images dyed with hematoxylin & eosin after being fixed by 10% formaldehyde and embedded into paraffin. In this figure, a shows a view (×40) that presents invasion of tumor cells across the abdominal wall and the peritoneum, b shows the same view (×400), c shows the view of invasion into spleen, and d shows the view of invasion into liver.
  • In the P815 cells transplanted group, P815 cells proliferation was suppressed and 30 percent of mice in this group lived by 6-7 weeks, while in PD-L1-expressing P815 cells (P815/PD-L1) transplanted group, carcinoma cells proliferation was remarkable and all mice died by 2-4 weeks (FIG. 2(A)). P815/PD-L1 cells were observed to permeate across the peritoneal cavity and the abdominal cavity, and further those metastases to liver and spleen (FIG. 2(B) a-d).
  • EXAMPLE 3
  • After co-culturing 2×10 6 of 2C cells togather with 5×10 6 of P815 cells or P815/PD-L1 cells only, or P815/PD-L1 cells under the presence of 10 mg/ml rat anti-PD-L1 F(ab′)2 IgG respectively, IFN-y in culture supernatant at 24 hours was measured with ELISA kit (purchased from Bioscience). The result is shown in FIG. 3(A).
  • FIG. 3(B) shows the result of evaluating of the tumor growth and the survival rate of mice of which anti-rat IgG (square) or anti-PD-L1 F(ab′)2 IgG (0.1 mg/mouse) (circle) was intraperitoneally administered to syngeneic DBA/2 mice(n=10) to which 3×10 6 of P815/PD-L1 cells had been hypodermically transferred on day 1, 3, 5, and 7 after the cell-transfer.
  • Anti-PD-L1 antibody restored IFN-y production from cytotoxic T lymphocytes which had been suppressed by P815/PD-L1 (FIG. 3(A)). Administration of anti-PD-L1 antibody suppressed carcinoma cell growth, and showed the clear survival effect (FIG. 3(B)). This result presents that administration of anti-PD-L1 antibody is effective on cancer treatment.
  • EXAMPLE 4
  • 10 6 of B16 melanomas (n=6) or B16/PD-L1 cells (n=6) were hypodermically transferred to B6 mice (n=6) respectively, and the same number of B16/PD-L1 cells were transferred to PD-1 transgenic B6 mice (n=5) and PD-1 gene homo-deficient B6 mice (PD−1/−(n=4)) (Science(2001), vol. 291, issue 5502, p. 319-332.), each tumor growth was measured by 25 days thereafter. FIG. 4 shows the result.
  • EXAMPLE 5
  • Tumor growth of which (n=9) anti-rat IgG or anti-PD-L1 F(ab′)2 IgG (0.1 mg/mouse) was intraperitoneally administered to syngeneic Balb/C mice to which 2.5×10 8 of J558 myeloma cells had been hypodermically transferred on day 3, 5, and 7 after the cell-transfer was evaluated. Each tumor growth in PD-1 gene homo-deficient Balb/C mice and Balb/C mice (n=4) to which J558 myeloma cells had been hypodermically transferred were compared (FIG. 5(B)).
  • Administration of anti-PD-L1 antibody suppressed PD-L1-expressing J558 carcinoma cells proliferation (flow cytometries of PD-L1 expression in various myeloma cell lines are shown in FIG. 5(A))(FIG. 5(B)). The transplanted tumor cells proliferation was completely inhibited in PD-1-deficient mice to which J558 cells had been transplanted (FIG. 5(c)). These results present that inhibition of PD-L1 or PD-1 is effective on cancer treatment.
  • EXAMPLE 6
  • Vascular endothelial cells (hereafter, abbreviated as ECs) was gathered from mouse heart by the method of Marelli-Berg (Journal of immunological methods (2000), vol. 244, issue 1-2, p. 205-215.). Concretely, heart tissue digested by collagenase was pre-cultured followed by being pre-cultured with mouse Ig and co-cultured together with FITC-modified anti-CD3 antibody, the same modified anti-CD105 antibody, the same modified anti-isolectin B4 antibody, and anti-FITC bead. These vascular endothelial cells were purified by positive selection using Magnetic-activated cell-sorting separation columns (the trade name: purchased from Miltenyi Biotec).
  • PD-L1 and PD-L2 expressions in the gathered vascular endothelial cells were confirmed by flow cytometry. The cell labelling was executed using anti-PD-L1 antibody (the antibody name: 1-111), anti-PD-L2 antibody (the antibody name: #122), and fluorescent labelling second antibody (FIG. 6(A)). The analysis was executed 10,000 events with Facscalibur (the equipment name: purchased from Becton Dickinson) using CellQuest software (purchased from Dickinson). PD-L1 or PD-L2 expression is shown as shedding curve, and control Ig is shown as filling curve.
  • PD-L1 expression in each mouse tissue was confirmed by tissue staining. Before 1 hour each tissue sampling, 100 μl of PBS which 100 μg of biotin-labelling anti-PD-L1 antibody (1-111) had been dissolved was intravenously administered to mouse. Then, 5 μm frozen section was fixed by 4% paraformaldehyde (PFA), and dyed with Streptavidin-FITC. Each section was countered staining by Phalloidin(FIG. 6(B) shows PD-L1 expression in (a) eyeball, (b) submaxillary gland, (c) heart, (d) lung, (e) liver, (f) kidney. In this figure, Ch presents choroid, CV presents vena centralis, GI presents glomerule, and Re presents retina. Each arrow presents vascular endothelial cells. Each staining figure is 40× enlargement view.). PD-L1 expression was observed in capillary endothelium of heart, lung, kidney, stomach, small intestine, submandibular gland, eyeball, and liver. The expression in liver was localized in liver sinusoidal capillary.
  • EXAMPLE 7
  • PD-L1 expression in liver non-parenchymal cells (hereafter, abbreviated with LNPCs) was confirmed by tissue staining (FIG. 7(A)) and flow cytometry (FIG. 7(B)). In the tissue staining, 5 μm liver frozen section fixed with 3% of PFA was preprocessed by rat serum followed by being reacted for 1 hour at room temperature using biotin-labeled anti-PD-L1 antibody (1-111) or biotin-labeled anti-ICAM-1 antibody (the trade name: purchased from BD Pharmingen), and then the biotin antibodies were visualized by tyramide signal amplification (TSA) fluorescence system (the equipment name: purchased from PerkinElmer Life Sciences) (FIG. 7(A) shows ICAM-1 expression, FIG. 7(B) shows PD-L1 expression, and CV presents vena centralis. Each staining figure is 40× enlargement view.).
  • LNPCs were isolated from mouse liver according to the pronaseE method (Experimental Cell Research (1976, vol. 99, p. 444-449.). Concretely, LNPCs obtained by which liver is circulated with pronaseE solution (Merck) were cultured and were separated by density gradient centrifugation. Relative distribution of Kupffer cells (CD54+, CD11bhigh) in the cell suspension is 20-25%, and that of liver perisinusoidal space endothelial cells (hereafter, abbreviated with LSECs.) (CD54+, CD11bhigh) is 75-80%. Kupffer cells and LSECs were doubly stained using FITC-labeled anti-CD11b antibody, each biotinated monoclonal antibody to ICAM-1, PD-L1, B7-1, and B7-2, and PE-labeled Streptavidin, respectively. Kupffer cells and LSECs were gate as CD11b high and CD11b low cell, respectively (FIG. 8).
  • PD-L1 expressed together with ICAM-1, B7-1, and B7-2 in Kupffer cells, while the expression was weak in LSECs (FIG. 8).
  • EXAMPLE 8
  • Native T cells were refined (refining degree 90% or more) from spleen and lymphoid of PD-1 gene homo-deficient mice (PD-1−/−) or wild type C57BL/6 mice (wt) by a negative selection using T-cell enrichment column (the trade name: purchased from Genzyme). The cells cultured for 48 hours with 10 μg/ml of anti-CD3 monoclonal antibody (2C11) were activated. The naive T cells activated by the above method were doubly stained using FITC-labeled anti-CD4 antibody or APC-labeled anti-CD8 antibody, PE-labeled anti-CD25 antibody, PE-labeled anti-CD4 antibody, PE-labeled anti-CD69 antibody, or PE-labeled anti-CTLA-4 antibody, biotin-labeled anti-B7-1 (CD80) antibody or biotin-labeled anti-B7-2 (CD86) antibody, and anti-PD-1 antibody (the antibody name: J43, monoclonal antibody producted from hybridomas recognized by International Trust Number FERM BP-8118) or anti-PD-L1 antibody (1-111), and each molecule expressions were analyzed by flow cytometry (FIGS. 9 and 10).
  • The hybridomas identified by International Trust Number FERM BP-8118 had been deposited as Trust Number FERM P-18356 to National Institute of Advanced Industrial Science and Technology, International Patent Organism Depositary in Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, Japan (ZIP code 305-8566) at May 30, 2001, and was transferred to International Deposit at Jul. 16, 2002.
  • EXAMPLE 9
  • The naive T cells activation of PD-1 gene homo-deficient mice (PD-1−/−) or wild type mice (wt) was executed by the method described in example 8. Proliferation of these activated cells was measured by BrdU incorporation method (FIG. 11(A)). The proliferation was measured by adding BrdU at the last 6 hours of 48 hours to label the cells and measuring using Proliferation ELISA kit (the trade name: Purchased from Roche). The amount of IFN-γ production at this time was measured in ELISA Kit (the trade name: purchased from Genzyme) (FIG. 12(A)).
  • T cells derived from PD-1 gene homo-deficient mice (PD-1−/−) or wild type mice (wt) was beforehand activated by the method described in example 8. The activated T cells were respectively cultured for 60 hours under the presence or the absence of Mitomycin C-processed LNPCs of wild type mice and under the presence or the absence of 30 μg/ml of anti-PD-L1 antibody (1-111) (rat IgG as control) and 20 μg/ml of CTLA4-Ig (Genzyme) (human IgG as control), and the cells proliferation for the last 12 hours was measured by BrdU incorporation method (FIG. 11(B)). Further, the amount of IFN-γ production at 48 hours was measured (FIG. 12(B)).
  • As to the amount of IFN-γ production when naive T cells were activated, a significant difference between PD-1−/− and wild type mice was not observed. On the other hand, in the activated T cells, the amount of IFN-γ production of T cells derived from wild type mice was significantly lower than that derived from PD-1−/− (FIG. 12). Therefore, it was suggested that the effect of PD-1 inhibitory action on the activated T cells could be higher than the effect on naive T cells activation.
  • In co-culture of the activated T cells derived from wild type mice and LNPCs, a significant difference in the cellular proliferation and the amount of IFN-γ production of these T cells was not observed, while in the co-culture of the activated T cells derived from PD-1−/−mice and LNPCs, a significant increase in the cellular proliferation of these T cells was observed (FIG. 11(B), FIG. 12(B)). In adding anti-PD-L1 antibody to the co-culture of the activated T cells derived from wild type mice and LNPCs, an increase in the cellular proliferation of these T cells was observed (FIG. 11(B)). These results suggest that PD-1 or PD-L1 of LNPCs could take part in suppression of T cells activation and lack of PD-1 or inhibition of interaction between PD-1 and PD-L1 could activate T cells.
  • The activated T cells of PD-1 gene homo-deficient mice (PD-1−/−) or wild type mice (wt) was labeled by 5 μM CFSE (5-(6)-carboxy-fluorescein diacetate succinimidyl diester) (the trade name: purchased from Molecular probes) and was co-cultured together with LNPCs for 48 hours. Cell division at this time was decided by CFSE activity measurement using FACS (FIG. 12(C)).
  • It was suggested that the cellular proliferation suppression of the activation T cells could cause in cytostasis suppression and PD-1 signal could suppress cell division of T cells (FIG. 12(C)).
  • EXAMPLE 10
  • PD-1 gene homo-deficient mice (PD-1−/−) or wild type mice (wt) (3 per a group) were infected in adenoviruses by intravenous administration of 10 910 1 PFU (plaque-forming units) of Ad-lacZ. Ad-lacZ used here, which are 5 type adenovirus with lacZ gene, lacks E1 and E3 region and can be purified by cesium chloride density-gradient centrifugation (Nucleic Acid Research (1995), vol. 234,issue 19, p. 3816-3821.) after proliferating them in 293 cells. On day 0 or 7 after infection, spleen cells and intrahepatic lymphocytes that had been gathered after intravenous administration of 0.5 mg of BrdU (the trade name: purchased from Sigma) to the mice at 1 hour before slaughtering were doubly labeled by anti-BrdU antibody and anti-CD19 antibody, or anti-CD3 antibody (FIG. 13).
  • The cells on day 7 after infection were doubly labeled by anti-BrdU antibody, anti-CD19 antibody, anti-CD3 antibody, anti-CD4 antibody, and anti-CD8 antibody (FIG. 14(B), each bar graph shows the ratio of BrdU-positive cells.).
  • In adenovirus infected PD-1−/−mice liver, the ratio of each proliferative (BrdU positive) lymphocyte (CD19 positive, CD3 positive, CD4 positive, or CD8 positive) had increased compared to that in similarly infected wild type mice liver. On the other hand, since such a phenomenon in spleen was not observed, it was suggested that PD-1 could inhibit T cells proliferation in inflammatory tissues (FIG. 14(B)).
  • EXAMPLE 11
  • On day 0 or 7 after 10 9-10 10 PFU of Ad-lacZ had been intravenously administered to PD-1 gene homo-deficient mice (PD-1−/−) or wild type mice (wt) (3 per a group), liver slices that had been gathered after intravenous administration of 0.5 mg of BrdU (the trade name: purchased from Sigma) to the mice at 1 hour before slaughtering were doubly labeled by anti-BrdU antibody (FIG. 15( a)-(d), 20× enlargement view.). Liver slices of PD-1 gene homo-deficient mice (PD-1−/−) on day 7 after infection were doubly labeled by anti-BrdU antibody, anti-CD19 antibody, anti-CD3 antibody, anti-CD4 antibody, and anti-CD8 antibody (FIG. 15( e), (f), 40× enlargement view.).
  • In wild type mice liver on day 30 after infection, the locally and medium cell invasion to sinusoidal capillary and non-parenchymal region was observed, but in PD-1−/−mice, hepatitis symptom was not observed (FIGS. 16( h), (i) and (i), (n)).
  • Liver slices of PD-1 gene homo-deficient mice (PD-1−/−) or wild type mice (wt) on day 7 or 30 after infection were stained by hematoxylin & eosin (FIG. 16( g)-(j), 20 × enlargement view, (k)-(n), 40× enlargement view.) and X-gal (FIG. 16( o)-(r), 40× enlargement view.). These results presented that PD-1 signal could take part in the exclusion of viruses by which the proliferation of effector T cells in virus infected tissue could be induced.
  • EXAMPLE 12
  • P-815/PD-L1 cells compellingly expressing mouse PD-L1 were sown into culture flask and were cultured in usual medium including 5 μg/mL of puromycin (purchased from Sigma)(hereafter, abbreviated as selective medium) at 37° C. under 5% CO2/95% air until to be 50% -90% confluent. Mouse cytotoxic T lymphocyte 2C cells were subcultured for several days in usual medium together with P-815 cells processed by MMC (Mitomycin C) and culture supernatant of ConA-stimulated rat splenic cells. The collected P-815/PD-L1 cells were cultured for 15 minutes after adding 3 μL of BATDA Reagent of DELFIA EuTDA Cytotoxicity Reagents (purchased from PerkinElmer). Then, they were washed with PBS. 2C cells subcultured for 5-8 days after adding P-815 cells were used.
  • As subject substrates, 20 μL(10 ng/mL) of anti-mouse PD-1 antibody (FIG. 17, anti-mPD-1Ab(J43)), anti-mouse PD-L1 antibody (anti-mPD-L1Ab(1-111) in this figure), mouse PD-1 Fc (mPD-1Fc in this figure), human PD-1 Fc (hPD-1Fc in this figure), mouse IgG2aκ(Control Ig in this figure), or PBS were dispensed into 96 well plate, and then 50 μL of P-815/PD-L1 cells or usual medium were added. Further, 50 μL of 2C cells, usual medium, or usual medium including 1% Triton ×100 were added. 50 μL of supernatants in wells which the usual medium were added were collected for background and other supernatants were preserved at 37° C. until being collected. The residual cells were cultured for 4 hours. The 96 well plates were centrifuged and the supernatants were collected. 200 μL of DELFIA Europium Solution of Cytotoxicity Reagents (purchased from PerkinElmer) were added to the collected supernatants, and were shaken for 15 minutes. After shaking, time resolved fluorescence measurement was executed with ARVOsx multi-label counter (WALLAC). The supernatants in wells that the usual medium including 1% Triton ×100 was added were used as a high control, and the one in wells that the usual medium were used as a low control.
  • The evaluated group is composed of a subject substrate, P-815/PD-L1 cells, and 2C cells, the high control group is composed of PBS, P-815/PD-L1 cells, and usual medium including 1% Triton ×100, and the low control group is composed of PBS, P-815/PD-L1 cells, and usual medium, the 2C cell control group is composed of PBS, usual medium, and 2C cells, and the background group is composed of PBS, P-815/PD-L1 cells, and usual medium. CTL activity (%) was calculated by the following formula. All values are the one that had been subtracted by the average of the background.

  • CTL activity (%)=([the measurement of the evaluation group]−[the measurement of the 2C cell control group]−[the measurement of the low control group])/([the measurement of the high control group]−[the measurement of the low control group])×100
  • Anti-PD-1 antibody, anti-PD-L1 antibody, and PD-1 Fc have significantly reinforced CTL activity (FIG. 17( a)-(d), E: T ratio presents the mixing ratio of 2C cells and PD-L1/P815 cells.).
  • EXAMPLE 13
  • The inhibiting effect of anti-PD-1 antibody on cancer metastasis was evaluated by intraperitoneal administration of anti-mouse PD-1 monoclonal antibody to C57BLJ6 mice to which B16 melanoma cells had been transferred at intervals of 2 days followed by measuring the liver weight on day 18 after transfer.
  • The increase in the liver weight of anti-PD-1 antibody administrated group was significantly suppressed than that in control IgG administrated control group (liver weight/carcinoma cell non-transferred group: 1.3 g, decrease from control group: 6.8 g to anti-PD-1 antibody administrated group: 3.5 g.). The suppression of the increase in this weight presents that the metastasis of B16 melanoma cells is suppressed.

Claims (31)

1-32. (canceled)
33. A method of treating a lung cancer comprising administering a composition comprising a human or humanized anti-PD-1 monoclonal antibody to a human with the lung cancer, wherein the administration of the composition treats the lung cancer in the human.
34. The method of claim 33, further comprising administering an anticancer agent.
35. The method of claim 34, wherein the anticancer agent is a chemotherapy drug.
36. The method of claim 35, wherein the chemotherapy drug is selected from an alkylating agent, a nitrosourea agent, an antimetabolite, an antitumor antibiotic, an alkaloid derived from a plant, a topoisomerase inhibitor, a hormone therapy medicine, a hormone antagonist, an aromatase inhibitor, a P-glycoprotein inhibitor and a platinum complex derivative.
37. The method of claim 34, wherein the anticancer agent is an anti-CTLA-4 antibody.
38. The method of claim 33, wherein the composition further comprises a solubilizer.
39. The method of claim 38, wherein the solubilizer comprises polysorbate 80.
40. The method of claim 33, wherein the composition is in a solution.
41. The method of claim 40, wherein the solution is a saline solution.
42. The method of claim 33, wherein the composition further comprises an excipient.
43. The method of claim 33, wherein the composition is administered parenterally.
44. The method of claim 43, wherein the composition is administered intravenously.
45. The method of claim 33, wherein the lung cancer is a squamous carcinoma.
46. The method of claim 33, wherein the lung cancer is an adenocarcinoma.
47. The method of claim 45, further comprising administering an anticancer agent.
48. The method of claim 46, further comprising administering an anticancer agent.
49. The method of claim 47, wherein the anticancer agent is an anti-CTLA-4 antibody.
50. The method of claim 48, wherein the anticancer agent is an anti-CTLA-4 antibody.
51. The method of claim 33, wherein the lung cancer expresses PD-L1.
52. The method of claim 33, wherein the lung cancer expresses PD-L2.
53. The method of claim 34, wherein the lung cancer expresses PD-L1.
54. The method of claim 34, wherein the lung cancer expresses PD-L2.
55. The method of claim 37, wherein the lung cancer expresses PD-L1 or PD-L2.
56. The method of claim 45, wherein the lung cancer expresses PD-L1 or PD-L2.
57. The method of claim 46, wherein the lung cancer expresses PD-L1.
58. The method of claim 51, wherein the PD-L1 expression is identified by immunohistochemistry.
59. The method of claim 52, wherein the PD-L2 expression is identified by immunohistochemistry.
60. The method of claim 55, wherein the PD-L1 or PD-L2 expression is identified by immunohistochemistry.
61. The method of claim 56, wherein the PD-L1 or PD-L2 expression is identified by immunohistochemistry.
62. The method of claim 57, wherein the PD-L1 expression is identified by immunohistochemistry.
US14/638,985 2002-07-03 2015-03-04 Immunopotentiative composition Expired - Lifetime US9067999B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/638,985 US9067999B1 (en) 2002-07-03 2015-03-04 Immunopotentiative composition

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
JP2002194491 2002-07-03
JP2002-194491 2002-07-03
JP2003-029846 2003-02-06
JP2003029846 2003-02-06
PCT/JP2003/008420 WO2004004771A1 (en) 2002-07-03 2003-07-02 Immunopotentiating compositions
US12/538,698 US8168179B2 (en) 2002-07-03 2009-08-10 Treatment method using anti-PD-L1 antibody
US12/959,307 US8728474B2 (en) 2002-07-03 2010-12-02 Immunopotentiative composition
US14/245,692 US9402899B2 (en) 2002-07-03 2014-04-04 Immunopotentiative composition
US14/638,985 US9067999B1 (en) 2002-07-03 2015-03-04 Immunopotentiative composition

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/245,692 Division US9402899B2 (en) 2002-07-03 2014-04-04 Immunopotentiative composition

Publications (2)

Publication Number Publication Date
US9067999B1 US9067999B1 (en) 2015-06-30
US20150197572A1 true US20150197572A1 (en) 2015-07-16

Family

ID=30117379

Family Applications (10)

Application Number Title Priority Date Filing Date
US10/519,925 Active 2024-08-08 US7595048B2 (en) 2002-07-03 2003-07-02 Method for treatment of cancer by inhibiting the immunosuppressive signal induced by PD-1
US12/538,698 Active US8168179B2 (en) 2002-07-03 2009-08-10 Treatment method using anti-PD-L1 antibody
US12/959,307 Active 2025-10-02 US8728474B2 (en) 2002-07-03 2010-12-02 Immunopotentiative composition
US14/245,692 Expired - Lifetime US9402899B2 (en) 2002-07-03 2014-04-04 Immunopotentiative composition
US14/550,585 Expired - Lifetime US9073994B2 (en) 2002-07-03 2014-11-21 Immunopotentiative composition
US14/638,985 Expired - Lifetime US9067999B1 (en) 2002-07-03 2015-03-04 Immunopotentiative composition
US15/000,869 Expired - Lifetime US9439962B2 (en) 2002-07-03 2016-01-19 Immunopotentiative composition
US15/000,863 Expired - Lifetime US9393301B2 (en) 2002-07-03 2016-01-19 Immunopotentiative composition
US15/226,848 Abandoned US20170051060A1 (en) 2002-07-03 2016-08-02 Immunopotentiative Composition
US16/459,392 Abandoned US20200062846A1 (en) 2002-07-03 2019-07-01 Immunopotentiative composition

Family Applications Before (5)

Application Number Title Priority Date Filing Date
US10/519,925 Active 2024-08-08 US7595048B2 (en) 2002-07-03 2003-07-02 Method for treatment of cancer by inhibiting the immunosuppressive signal induced by PD-1
US12/538,698 Active US8168179B2 (en) 2002-07-03 2009-08-10 Treatment method using anti-PD-L1 antibody
US12/959,307 Active 2025-10-02 US8728474B2 (en) 2002-07-03 2010-12-02 Immunopotentiative composition
US14/245,692 Expired - Lifetime US9402899B2 (en) 2002-07-03 2014-04-04 Immunopotentiative composition
US14/550,585 Expired - Lifetime US9073994B2 (en) 2002-07-03 2014-11-21 Immunopotentiative composition

Family Applications After (4)

Application Number Title Priority Date Filing Date
US15/000,869 Expired - Lifetime US9439962B2 (en) 2002-07-03 2016-01-19 Immunopotentiative composition
US15/000,863 Expired - Lifetime US9393301B2 (en) 2002-07-03 2016-01-19 Immunopotentiative composition
US15/226,848 Abandoned US20170051060A1 (en) 2002-07-03 2016-08-02 Immunopotentiative Composition
US16/459,392 Abandoned US20200062846A1 (en) 2002-07-03 2019-07-01 Immunopotentiative composition

Country Status (15)

Country Link
US (10) US7595048B2 (en)
EP (4) EP2243493A1 (en)
JP (9) JP4409430B2 (en)
AT (1) ATE481985T1 (en)
AU (1) AU2003281200A1 (en)
DE (2) DE60334303D1 (en)
DK (2) DK2206517T3 (en)
ES (2) ES2654064T3 (en)
FI (1) FI2206517T3 (en)
FR (2) FR15C0088I1 (en)
HU (3) HUE10161767T1 (en)
LU (2) LU92905I2 (en)
PT (2) PT1537878E (en)
SI (1) SI2206517T1 (en)
WO (1) WO2004004771A1 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9358289B2 (en) 2005-05-09 2016-06-07 Ono Pharmaceutical Co., Ltd. Methods for treating cancer using anti-PD-1 antibodies in combination with anti-CTLA-4 antibodies
US9393301B2 (en) 2002-07-03 2016-07-19 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
WO2018039131A1 (en) * 2016-08-22 2018-03-01 Protiva Biotherapeutics, Inc. Anti-pd-1 antibodies, or fragments thereof, for treating hepatitis b
US9907849B2 (en) 2014-07-18 2018-03-06 Advaxis, Inc. Combination of a PD-1 antagonist and a listeria-based vaccine for treating prostate cancer
US10081681B2 (en) 2013-09-20 2018-09-25 Bristol-Myers Squibb Company Combination of anti-LAG-3 antibodies and anti-PD-1 antibodies to treat tumors
US10266591B2 (en) 2012-07-02 2019-04-23 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (LAG-3), and uses thereof
US10344089B2 (en) 2008-08-11 2019-07-09 E.R. Squibb & Sons, L.L.C. Human antibodies that bind lymphocyte activation gene-3 (LAG-3), and uses thereof
US10392442B2 (en) 2015-12-17 2019-08-27 Bristol-Myers Squibb Company Use of anti-PD-1 antibody in combination with anti-CD27 antibody in cancer treatment
US11083790B2 (en) 2016-06-02 2021-08-10 Bristol-Myers Squibb Company Treatment of Hodgkin lymphoma using an anti-PD-1 antibody
US11299543B2 (en) 2016-06-02 2022-04-12 Bristol-Myers Squibb Company Use of an anti-PD-1 antibody in combination with an anti-CD30 antibody in cancer treatment
US11332529B2 (en) 2016-06-03 2022-05-17 Bristol-Myers Squibb Company Methods of treating colorectal cancer
US11723975B2 (en) 2017-05-30 2023-08-15 Bristol-Myers Squibb Company Compositions comprising an anti-LAG-3 antibody or an anti-LAG-3 antibody and an anti-PD-1 or anti-PD-L1 antibody
US11767361B2 (en) 2016-06-03 2023-09-26 Bristol-Myers Squibb Company Method of treating lung cancer
US11807686B2 (en) 2017-05-30 2023-11-07 Bristol-Myers Squibb Company Treatment of LAG-3 positive tumors

Families Citing this family (663)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HU228477B1 (en) 1999-08-23 2013-03-28 Dana Farber Cancer Inst Inc Pd-1, a receptor for b7-4, and uses therefor
AU784634B2 (en) 1999-11-30 2006-05-18 Mayo Foundation For Medical Education And Research B7-H1, a novel immunoregulatory molecule
US7030219B2 (en) 2000-04-28 2006-04-18 Johns Hopkins University B7-DC, Dendritic cell co-stimulatory molecules
AU2003270439B2 (en) 2002-09-11 2009-09-24 Genentech, Inc. Novel composition and methods for the treatment of immune related diseases
US7432351B1 (en) 2002-10-04 2008-10-07 Mayo Foundation For Medical Education And Research B7-H1 variants
JP5303146B2 (en) 2004-10-06 2013-10-02 メイヨ・ファウンデーション・フォー・メディカル・エデュケーション・アンド・リサーチ B7-H1 and methods for diagnosis, prognosis and treatment of cancer
EP1891107B1 (en) 2005-05-12 2011-07-06 ZymoGenetics, Inc. Compositions and methods for modulating immune responses
NZ593388A (en) 2005-06-08 2012-08-31 Dana Farber Cancer Inst Inc Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (pd-1) pathway
AU2012204032B2 (en) * 2005-06-08 2014-01-16 Dana-Farber Cancer Institute, Inc. Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (PD-1) pathway
AU2006265108C1 (en) 2005-07-01 2013-01-17 E. R. Squibb & Sons, L.L.C. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
RU2423375C2 (en) * 2005-07-26 2011-07-10 СиЭмЭс ПЕПТАЙДЗ ПЭЙТЕНТ ХОЛДИНГ КОМПАНИ ЛИМИТЕД New biologically active peptides and their new application
EP1777523A1 (en) * 2005-10-19 2007-04-25 INSERM (Institut National de la Santé et de la Recherche Médicale) An in vitro method for the prognosis of progression of a cancer and of the outcome in a patient and means for performing said method
WO2007082144A2 (en) * 2006-01-05 2007-07-19 Mayo Foundation For Medical Education And Research B7-h1 and survivin in cancer
US20090215084A1 (en) * 2006-01-05 2009-08-27 Mayo Foundation For Medical Education And Research B7-h1 and b7-h4 in cancer
US8216996B2 (en) 2006-03-03 2012-07-10 Ono Pharmaceutical Co., Ltd. Multimer of extracellular domain of cell surface functional molecule
WO2007124361A2 (en) * 2006-04-20 2007-11-01 Mayo Foundation For Medical Education And Research Soluble b7-h1
DK2772535T3 (en) 2006-05-31 2022-07-04 Childrens Medical Center ABC5-positive mesenchymal stem cells as immune modulators
AU2007342338A1 (en) * 2006-09-20 2008-07-17 The Johns Hopkins University Combinatorial therapy of cancer and infectious diseases with anti-B7-H1 antibodies
RS55740B1 (en) 2006-10-02 2017-07-31 Squibb & Sons Inc Human antibodies that bind cxcr4 and uses thereof
NZ619576A (en) * 2006-12-27 2014-07-25 Harvard College Compositions and methods for the treatment of infections and tumors
US7928202B2 (en) 2007-04-12 2011-04-19 The Brigham And Women's Hospital, Inc. Targeting ABCB5 for cancer therapy
DK2170959T3 (en) 2007-06-18 2014-01-13 Merck Sharp & Dohme ANTIBODIES AGAINST HUMAN PROGRAMMED DEATH RECEPTOR PD-1
EP2170946A2 (en) 2007-07-13 2010-04-07 The Johns Hopkins University B7-dc variants
WO2009101611A1 (en) 2008-02-11 2009-08-20 Curetech Ltd. Monoclonal antibodies for tumor treatment
US20110020325A1 (en) * 2008-02-29 2011-01-27 Kwon Eugene D Methods for reducing granulomatous inflammation
EP2262837A4 (en) * 2008-03-12 2011-04-06 Merck Sharp & Dohme Pd-1 binding proteins
CN104655854A (en) 2008-04-09 2015-05-27 健泰科生物技术公司 Novel compositions and methods for the treatment of immune related diseases
EP2853269B1 (en) 2008-05-19 2019-05-01 Advaxis, Inc. Dual delivery system for heterologous antigens comprising a recombinant Listeria strain attenuated by mutation of dal/dat and deletion of ActA comprising a nucleic acid molecule encoding an listeriolysin O - prostate specific anigen fusion protein
US9017660B2 (en) 2009-11-11 2015-04-28 Advaxis, Inc. Compositions and methods for prevention of escape mutation in the treatment of Her2/neu over-expressing tumors
US9650639B2 (en) 2008-05-19 2017-05-16 Advaxis, Inc. Dual delivery system for heterologous antigens
JP5945096B2 (en) * 2008-07-04 2016-07-05 小野薬品工業株式会社 Use of a determination marker for optimizing the therapeutic effect of anti-human PD-1 antibody on cancer
CN104740610A (en) * 2008-08-25 2015-07-01 安普利穆尼股份有限公司 PD-1 Antagonists and Methods for Treating Infectious Disease
MX2011002252A (en) 2008-08-25 2011-06-24 Amplimmune Inc Compositions of pd-1 antagonists and methods of use.
KR101050829B1 (en) 2008-10-02 2011-07-20 서울대학교산학협력단 Anticancer agents comprising an anti-PD-1 antibody or an anti-PD-L1 antibody
US11542328B2 (en) 2008-11-14 2023-01-03 The Brigham And Women's Hospital, Inc. Therapeutic and diagnostic methods relating to cancer stem cells
US20110287034A1 (en) 2008-11-14 2011-11-24 The Brigham And Womens Hospital, Inc. Therapeutic and diagnostic methods relating to cancer stem cells
US9598491B2 (en) * 2008-11-28 2017-03-21 Emory University Methods for the treatment of infections and tumors
DK2376535T3 (en) 2008-12-09 2017-06-12 Hoffmann La Roche ANTI-PD-L1 ANTIBODIES AND THEIR USE TO PROMOTE T CELL FUNCTION
CN107090437A (en) 2009-03-06 2017-08-25 国立大学法人三重大学 Method for strengthening T cell function
CA2775761C (en) 2009-09-30 2018-08-28 Memorial Sloan-Kettering Cancer Center Combination immunotherapy for the treatment of cancer
US10016617B2 (en) 2009-11-11 2018-07-10 The Trustees Of The University Of Pennsylvania Combination immuno therapy and radiotherapy for the treatment of Her-2-positive cancers
EP3279215B1 (en) 2009-11-24 2020-02-12 MedImmune Limited Targeted binding agents against b7-h1
TW201134488A (en) * 2010-03-11 2011-10-16 Ucb Pharma Sa PD-1 antibodies
JP5920929B2 (en) 2010-03-11 2016-05-18 ユセベ ファルマ ソシエテ アノニム PD-1 antibody
US20150231215A1 (en) 2012-06-22 2015-08-20 Randolph J. Noelle VISTA Antagonist and Methods of Use
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
BR112012024565B1 (en) 2010-03-26 2022-02-08 Trustees Of Dartmouth College FUSION PROTEIN VIEWED ISOLATED OR RECOMBINANT MULTIMERIC IMMUNOSUPPRESSANT AND COMPOSITION
JP2013532153A (en) 2010-06-18 2013-08-15 ザ ブリガム アンド ウィメンズ ホスピタル インコーポレイテッド Bispecific antibodies against TIM-3 and PD-1 for immunotherapy against chronic immune disease
US8907053B2 (en) 2010-06-25 2014-12-09 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
US9783578B2 (en) 2010-06-25 2017-10-10 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
US9226958B2 (en) 2010-10-01 2016-01-05 University Of Georgia Research Foundation, Inc. Use of Listeria vaccine vectors to reverse vaccine unresponsiveness in parasitically infected individuals
EP2625292B1 (en) 2010-10-07 2018-12-05 The General Hospital Corporation Biomarkers of cancer
WO2012125551A1 (en) 2011-03-11 2012-09-20 Advaxis Listeria-based adjuvants
EP3403672A1 (en) 2011-04-20 2018-11-21 Medlmmune, LLC Antibodies and other molecules that bind b7-h1 and pd-1
CN102250910B (en) * 2011-05-31 2013-01-30 苏州大学 Human soluble B7-DC quantitative detection kit
WO2012168944A1 (en) 2011-06-08 2012-12-13 Aurigene Discovery Technologies Limited Therapeutic compounds for immunomodulation
PT3409278T (en) 2011-07-21 2020-12-18 Sumitomo Dainippon Pharma Oncology Inc Heterocyclic protein kinase inhibitors
WO2013053775A1 (en) 2011-10-11 2013-04-18 Universität Zürich Prorektorat Mnw Combination medicament comprising il-12 and an agent for blockade of t-cell inhibitory molecules for tumour therapy
JP6259763B2 (en) * 2011-10-17 2018-01-10 ヘルレフ ホスピタルHerlev Hospital Immunotherapy based on PD-L1
US8466159B2 (en) 2011-10-21 2013-06-18 Abbvie Inc. Methods for treating HCV
US8492386B2 (en) 2011-10-21 2013-07-23 Abbvie Inc. Methods for treating HCV
WO2013059638A1 (en) 2011-10-21 2013-04-25 Abbvie Inc. Combination treatment (eg. with abt-072 or abt -333) of daas for use in treating hcv
EP2583677A3 (en) 2011-10-21 2013-07-03 Abbvie Inc. Methods for treating HCV comprising at least two direct acting antiviral agent, ribavirin but not interferon.
KR101981873B1 (en) * 2011-11-28 2019-05-23 메르크 파텐트 게엠베하 Anti-pd-l1 antibodies and uses thereof
GB201120779D0 (en) 2011-12-02 2012-01-11 Immodulon Therapeutics Ltd Cancer therapy
WO2013090552A1 (en) * 2011-12-13 2013-06-20 Yale University Compositions and methods for reducing ctl exhaustion
CN104245734B (en) 2012-01-20 2017-09-05 非营利性组织佛兰芒综合大学生物技术研究所 Target mutant alpha helical bundle cell factor
EP2814847B1 (en) 2012-02-16 2018-04-18 VLP Therapeutics, LLC Virus like particle composition
WO2013132317A1 (en) 2012-03-07 2013-09-12 Aurigene Discovery Technologies Limited Peptidomimetic compounds as immunomodulators
SG11201405605VA (en) 2012-03-12 2014-10-30 Advaxis Inc SUPPRESSOR CELL FUNCTION INHIBITION FOLLOWING <i>LISTERIA</i> VACCINE TREATMENT
US9422339B2 (en) 2012-03-29 2016-08-23 Aurigene Discovery Technologies Limited Immunomodulating cyclic compounds
EP2836234B1 (en) 2012-04-12 2019-06-12 Yale University Vehicles for controlled delivery of different pharmaceutical agents
SG10201700698WA (en) * 2012-05-15 2017-02-27 Bristol Myers Squibb Co Cancer immunotherapy by disrupting pd-1/pd-l1 signaling
MY180127A (en) * 2012-05-25 2020-11-23 Cellectis Methods for engineering allogeneic and immunosuppressive resistant t cell for immunotherapy
US9890215B2 (en) * 2012-06-22 2018-02-13 King's College London Vista modulators for diagnosis and treatment of cancer
CN112587658A (en) 2012-07-18 2021-04-02 博笛生物科技有限公司 Targeted immunotherapy for cancer
CN109793893B (en) * 2012-09-07 2023-05-26 达特茅斯大学理事会 VISTA modulators for diagnosis and treatment of cancer
EP2950814A4 (en) 2013-01-31 2016-06-08 Univ Jefferson Pd-l1 and pd-l2-based fusion proteins and uses thereof
WO2014122271A1 (en) 2013-02-07 2014-08-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for predicting the survival time of patients suffering from diffuse large b-cell lymphomas
US20150368316A1 (en) * 2013-02-07 2015-12-24 Albert Einstein College Of Medicine Of Yeshiva University A selective high-affinity immune stimulatory reagent and uses thereof
BR112015018989B1 (en) * 2013-02-22 2023-11-14 Curevac Ag COMBINATION VACCINE/PD-1 PATHWAY INHIBITOR, PD-1 PATHWAY INHIBITOR AND RNA VACCINE
US9302005B2 (en) 2013-03-14 2016-04-05 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
EP2983790A2 (en) 2013-04-09 2016-02-17 Boston Biomedical, Inc. Methods for treating cancer
AU2014259719B2 (en) 2013-05-02 2019-10-03 Anaptysbio, Inc. Antibodies directed against programmed death-1 (PD-1)
CN103242448B (en) * 2013-05-27 2015-01-14 郑州大学 Full-humanized anti-PD-1 monoclonal antibody and preparation method and application thereof
WO2014194293A1 (en) 2013-05-30 2014-12-04 Amplimmune, Inc. Improved methods for the selection of patients for pd-1 or b7-h4 targeted therapies, and combination therapies thereof
CN105683217B (en) 2013-05-31 2019-12-10 索伦托治疗有限公司 Antigen binding proteins that bind to PD-1
CN104250302B (en) 2013-06-26 2017-11-14 上海君实生物医药科技股份有限公司 The anti-antibody of PD 1 and its application
TWI676636B (en) 2013-07-12 2019-11-11 Vlp醫療股份有限公司 Virus like particle comprising pd-1 antigen or pd-1 ligand antigen
WO2015009856A2 (en) 2013-07-16 2015-01-22 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and tigit inhibitors
HUE048874T2 (en) 2013-09-06 2020-08-28 Aurigene Discovery Tech Ltd 1,2,4-oxadiazole derivatives as immunomodulators
SG11201601679TA (en) 2013-09-06 2016-04-28 Aurigene Discovery Tech Ltd 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators
EA201891818A3 (en) 2013-09-06 2019-03-29 Ауриген Дискавери Текнолоджиз Лимитед METHOD FOR PREPARING CONNECTIONS USED WHEN OBTAINING CYCLIC PEPTIDOMIMETIC COMPOUNDS
MX2016003292A (en) 2013-09-13 2016-06-24 Beigene Ltd Anti-pd1 antibodies and their use as therapeutics and diagnostics.
EP3757130A1 (en) 2013-09-26 2020-12-30 Costim Pharmaceuticals Inc. Methods for treating hematologic cancers
WO2015050663A1 (en) 2013-10-01 2015-04-09 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of bim
WO2015066535A1 (en) 2013-11-01 2015-05-07 Yale University Modular particles for immunotherapy
SG10201804945WA (en) 2013-12-12 2018-07-30 Shanghai hengrui pharmaceutical co ltd Pd-1 antibody, antigen-binding fragment thereof, and medical application thereof
GB201322626D0 (en) 2013-12-19 2014-02-05 Immutep S A Combined preparations for the treatment of cancer
GB201322725D0 (en) 2013-12-20 2014-02-05 Immodulon Therapeutics Ltd Cancer therapy
CA3154540A1 (en) * 2013-12-20 2015-06-25 Intervet International B.V. Caninized antibodies
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
KR102534681B1 (en) 2013-12-24 2023-05-18 잔센파마슈티카엔.브이. Anti-vista antibodies and fragments
EP3092254A4 (en) 2014-01-10 2017-09-20 Birdie Biopharmaceuticals Inc. Compounds and compositions for treating her2 positive tumors
LT3094351T (en) * 2014-01-15 2022-04-11 Kadmon Corporation, Llc Immunomodulatory agents
TWI680138B (en) 2014-01-23 2019-12-21 美商再生元醫藥公司 Human antibodies to pd-l1
TWI681969B (en) 2014-01-23 2020-01-11 美商再生元醫藥公司 Human antibodies to pd-1
JOP20200094A1 (en) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc Antibody molecules to pd-1 and uses thereof
JOP20200096A1 (en) 2014-01-31 2017-06-16 Children’S Medical Center Corp Antibody molecules to tim-3 and uses thereof
SG11201606428UA (en) 2014-02-04 2016-09-29 Incyte Corp Combination of a pd-1 antagonist and an ido1 inhibitor for treating cancer
US10899840B2 (en) 2014-02-04 2021-01-26 Pfizer Inc. Combination of a PD-1 antagonist and a 4-1BB agonist for treating cancer
TR201901507T4 (en) 2014-02-04 2019-02-21 Merck Sharp & Dohme Combination of a pd-1 antagonist and a vegfr inhibitor for treating cancer.
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
WO2015135558A1 (en) 2014-03-12 2015-09-17 Curevac Gmbh Combination of vaccination and ox40 agonists
US10618963B2 (en) 2014-03-12 2020-04-14 Yeda Research And Development Co. Ltd Reducing systemic regulatory T cell levels or activity for treatment of disease and injury of the CNS
US10144778B2 (en) 2014-03-12 2018-12-04 Yeda Research And Development Co. Ltd Reducing systemic regulatory T cell levels or activity for treatment of disease and injury of the CNS
US10519237B2 (en) 2014-03-12 2019-12-31 Yeda Research And Development Co. Ltd Reducing systemic regulatory T cell levels or activity for treatment of disease and injury of the CNS
US9394365B1 (en) 2014-03-12 2016-07-19 Yeda Research And Development Co., Ltd Reducing systemic regulatory T cell levels or activity for treatment of alzheimer's disease
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
CA2949998A1 (en) 2014-05-28 2015-12-03 Agenus Inc. Anti-gitr antibodies and methods of use thereof
WO2015181624A2 (en) 2014-05-28 2015-12-03 Idenix Pharmaceuticals, Inc Nucleoside derivatives for the treatment of cancer
WO2015191881A2 (en) 2014-06-11 2015-12-17 Green Kathy A Use of vista agonists and antagonists to suppress or enhance humoral immunity
TWI693232B (en) 2014-06-26 2020-05-11 美商宏觀基因股份有限公司 Covalently bonded diabodies having immunoreactivity with pd-1 and lag-3, and methods of use thereof
JP6526189B2 (en) 2014-07-03 2019-06-05 ベイジーン リミテッド Anti-PD-L1 antibodies and their use for therapy and diagnosis
EP3168236B1 (en) 2014-07-09 2019-09-04 Nippon Zenyaku Kogyo Co., Ltd. Anti-canine pd-1 antibody or anti-canine pd-l1 antibody
EP3166976B1 (en) 2014-07-09 2022-02-23 Birdie Biopharmaceuticals Inc. Anti-pd-l1 combinations for treating tumors
CN106999577B (en) 2014-07-16 2021-12-21 特兰斯吉恩股份有限公司 Oncolytic virus and immune checkpoint modulator combinations
ES2743873T3 (en) 2014-07-16 2020-02-21 Transgene Sa Oncolytic virus for the expression of immune control point modulators
ES2905301T3 (en) 2014-07-22 2022-04-07 Apollomics Inc Anti-PD-1 antibodies
EP3171896A4 (en) 2014-07-23 2018-03-21 Mayo Foundation for Medical Education and Research Targeting dna-pkcs and b7-h1 to treat cancer
US11186640B2 (en) * 2014-07-31 2021-11-30 The University Of Western Australia Method for the identification of immunotherapy-drug combinations using a network approach
CN105296433B (en) 2014-08-01 2018-02-09 中山康方生物医药有限公司 A kind of CTLA4 antibody, its medical composition and its use
KR102476226B1 (en) 2014-08-05 2022-12-12 아폴로믹스 인코포레이티드 Anti-pd-l1 antibodies
US9982052B2 (en) 2014-08-05 2018-05-29 MabQuest, SA Immunological reagents
JP6629321B2 (en) 2014-08-05 2020-01-15 マブクエスト エスエーMabQuest SA Immunological reagent
WO2016021720A1 (en) * 2014-08-07 2016-02-11 学校法人兵庫医科大学 Therapeutic agent for cancer which comprises combination of il-18 and molecule-targeting antibody
US10385101B2 (en) 2014-08-08 2019-08-20 Vlp Therapeutics, Llc Virus like particle comprising modified envelope protein E3
WO2016021209A1 (en) 2014-08-08 2016-02-11 Vlp Therapeutics, Llc Virus like particle comprising modified envelope protein e3
MX2017002134A (en) * 2014-08-19 2017-09-13 Nat Univ Corporation Okayama Univ Method for enhancing immune cell function and method for assessing immune cell multifunctionality.
US10695426B2 (en) 2014-08-25 2020-06-30 Pfizer Inc. Combination of a PD-1 antagonist and an ALK inhibitor for treating cancer
CN112546238A (en) 2014-09-01 2021-03-26 博笛生物科技有限公司 anti-PD-L1 conjugates for the treatment of tumors
KR102532832B1 (en) 2014-09-11 2023-05-16 브이엘피 테라퓨틱스 인코포레이티드 Flavivirus virus like particle
EP3191127A1 (en) 2014-09-13 2017-07-19 Novartis AG Combination therapies of egfr inhibitors
US10039564B2 (en) 2014-09-30 2018-08-07 Ethicon Llc Surgical devices having power-assisted jaw closure and methods for compressing and sensing tissue
JP6797111B2 (en) 2014-09-30 2020-12-09 インターベット インターナショナル ベー. フェー. PD-L1 antibody that binds to canine PD-L1
WO2016057898A1 (en) 2014-10-10 2016-04-14 Idera Pharmaceuticals, Inc. Treatment of cancer using tlr9 agonist with checkpoint inhibitors
JP6877339B2 (en) 2014-10-14 2021-05-26 ノバルティス アーゲー Antibody molecule against PD-L1 and its use
JP7305300B2 (en) 2014-11-05 2023-07-10 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Combination immunotherapy
KR20170101925A (en) * 2014-12-02 2017-09-06 셀진 코포레이션 Combination therapy
EP3226688B1 (en) 2014-12-05 2020-07-01 Merck Sharp & Dohme Corp. Tricyclic compounds as inhibitors of mutant idh enzymes
WO2016089833A1 (en) 2014-12-05 2016-06-09 Merck Sharp & Dohme Corp. Novel tricyclic compounds as inhibitors of mutant idh enzymes
AU2015357463B2 (en) 2014-12-05 2021-10-07 Immunext, Inc. Identification of VSIG8 as the putative vista receptor and its use thereof to produce vista/VSIG8 modulators
US10508108B2 (en) 2014-12-05 2019-12-17 Merck Sharp & Dohme Corp. Tricyclic compounds as inhibitors of mutant IDH enzymes
EP3229837A4 (en) * 2014-12-08 2018-05-30 Dana-Farber Cancer Institute, Inc. Methods for upregulating immune responses using combinations of anti-rgmb and anti-pd-1 agents
SG11201703931QA (en) * 2014-12-09 2017-06-29 Regeneron Pharma Non-human animals having a humanized cluster of differentiation 274 gene
TWI595006B (en) 2014-12-09 2017-08-11 禮納特神經系統科學公司 Anti-pd-1 antibodies and methods of use thereof
JP6767096B2 (en) * 2014-12-11 2020-10-14 リティックス バイオファーマ エイエス Combination of immune checkpoint inhibitors
US10849862B2 (en) 2014-12-26 2020-12-01 Theravalues Corporation Formulation of curcumin and anti-PD-1 antibody
EP3835312A1 (en) 2014-12-31 2021-06-16 Checkmate Pharmaceuticals, Inc. Combination tumor immunotherapy
GB201500374D0 (en) 2015-01-09 2015-02-25 Immutep S A Combined preparations for the treatment of cancer
GB201500319D0 (en) * 2015-01-09 2015-02-25 Agency Science Tech & Res Anti-PD-L1 antibodies
MA41414A (en) 2015-01-28 2017-12-05 Centre Nat Rech Scient ICOS AGONIST BINDING PROTEINS
EP3252469A1 (en) * 2015-01-29 2017-12-06 Konica Minolta, Inc. Method for simultaneous detection of blood cells having interacting molecules
MA41460A (en) 2015-02-03 2017-12-12 Oncomed Pharm Inc TNFRSF LIAISON AGENTS AND THEIR USES
EP3256123B1 (en) * 2015-02-12 2023-06-07 The Johns Hopkins University Inhibition of yap for breaking tumor immune tolerance
JP6885869B2 (en) 2015-02-26 2021-06-16 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung PD-1 / PD-L1 Inhibitors for Treating Cancer
AR103726A1 (en) 2015-02-27 2017-05-31 Merck Sharp & Dohme HUMAN ANTI-PD-1 MONOCLONAL ANTIBODY CRYSTALS
KR20170122810A (en) 2015-03-04 2017-11-06 머크 샤프 앤드 돔 코포레이션 A combination of PD-1 antagonist and eribulin for treating cancer
AU2015384801B2 (en) 2015-03-04 2022-01-06 Eisai R&D Management Co., Ltd. Combination of a PD-1 antagonist and a VEGFR/FGFR/RET tyrosine kinase inhibitor for treating cancer
CN114213356A (en) 2015-03-10 2022-03-22 奥瑞基尼探索技术有限公司 1,2, 4-oxadiazole and thiadiazole compounds as immunomodulators
EP3268392A2 (en) 2015-03-13 2018-01-17 CytomX Therapeutics, Inc. Anti-pdl1 antibodies, activatable anti-pdl1 antibodies, and methods of use thereof
MA53297A (en) 2015-03-23 2022-05-04 Jounce Therapeutics Inc ANTI-ICOS ANTIBODIES
ES2820768T3 (en) 2015-04-03 2021-04-22 Xoma Technology Ltd Cancer treatment using TGF-beta and PD-1 inhibitors
EA201792273A1 (en) 2015-04-17 2018-04-30 Бристол-Маерс Сквибб Компани COMPOSITIONS CONTAINING A COMBINATION OF ANTI-PD-1 ANTIBODIES AND OTHER ANTIBODIES
EP3988571A1 (en) 2015-04-28 2022-04-27 Bristol-Myers Squibb Company Treatment of pd-l1-negative melanoma using an anti-pd-1 antibody and an anti-ctla-4 antibody
US10815264B2 (en) 2015-05-27 2020-10-27 Southern Research Institute Nucleotides for the treatment of cancer
RU2017145558A (en) 2015-05-29 2019-07-01 Мерк Шарп И Доум Корп. COMBINATION OF ANTAGONIST PD-1 AND OLIGONUCLEOTIDE TYPE CPG-C FOR CANCER TREATMENT
LT3303394T (en) 2015-05-29 2020-10-12 Agenus Inc. Anti-ctla-4 antibodies and methods of use thereof
ES2752063T3 (en) * 2015-05-29 2020-04-02 Dynavax Tech Corp Intrapulmonary administration of toll 9 receptor polynucleotide agonists for the treatment of lung cancer
US11078278B2 (en) 2015-05-29 2021-08-03 Bristol-Myers Squibb Company Treatment of renal cell carcinoma
TW201717935A (en) 2015-06-03 2017-06-01 波士頓生醫公司 Compositions and methods for treating cancer
TWI773646B (en) 2015-06-08 2022-08-11 美商宏觀基因股份有限公司 Lag-3-binding molecules and methods of use thereof
TW201709929A (en) 2015-06-12 2017-03-16 宏觀基因股份有限公司 Combination therapy for the treatment of cancer
US20180141991A1 (en) 2015-06-12 2018-05-24 Mackay Medical Foundation The Presbyterian Church In Taiwan Mackay Memorial Hospital Methods and polypeptides for modulation of immunoresponse
KR20180018762A (en) 2015-06-16 2018-02-21 메르크 파텐트 게엠베하 Pd-l1 antagonist combination treatments
US11207405B2 (en) * 2015-06-24 2021-12-28 Immodulon Therapeutics Limited Checkpoint inhibitor and a whole cell Mycobacterium for use in cancer therapy
BR112017027870A2 (en) 2015-06-24 2018-08-28 Janssen Pharmaceutica Nv antibodies and anti-sight fragments
GB201511790D0 (en) 2015-07-06 2015-08-19 Iomet Pharma Ltd Pharmaceutical compound
JP7014706B2 (en) 2015-07-13 2022-02-01 サイトメックス セラピューティクス インコーポレイテッド Anti-PD-1 antibody, activating anti-PD-1 antibody, and how to use it
EP3322731B1 (en) 2015-07-14 2021-01-13 Bristol-Myers Squibb Company Method of treating cancer using immune checkpoint inhibitor; antibody that binds to programmed death-1 receptor (pd-1) or programmed death ligand 1 (pd-l1)
CN111375066B (en) 2015-07-16 2023-04-25 百欧肯治疗有限公司 Compositions and methods for treating cancer
DK3328419T3 (en) 2015-07-30 2021-10-11 Macrogenics Inc PD-1 BINDING MOLECULES AND METHODS FOR USING IT
EP3328896A4 (en) 2015-07-31 2019-08-07 University of Florida Research Foundation, Inc. Hematopoietic stem cells in combinatorial therapy with immune checkpoint inhibitors against cancer
RU2018107693A (en) 2015-08-04 2019-09-05 Глэксосмитклайн Интеллекчуал Проперти Дивелопмент Лимитед COMBINED TYPES OF TREATMENT AND THEIR OPTIONS AND METHODS
EP3331918A1 (en) 2015-08-04 2018-06-13 GlaxoSmithKline Intellectual Property Development Limited Combination treatments and uses and methods thereof
JP2018526989A (en) 2015-08-07 2018-09-20 ピエリス ファーマシューティカルズ ゲーエムベーハー Novel fusion polypeptide specific for LAG-3 and PD-1
EP3331919A1 (en) 2015-08-07 2018-06-13 GlaxoSmithKline Intellectual Property Development Limited Combination therapy comprising anti ctla-4 antibodies
WO2017024465A1 (en) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
SG10201914109VA (en) 2015-08-11 2020-02-27 Wuxi Biologics Cayman Inc Novel anti-pd-1 antibodies
US11453697B1 (en) 2015-08-13 2022-09-27 Merck Sharp & Dohme Llc Cyclic di-nucleotide compounds as sting agonists
EA034786B1 (en) 2015-08-13 2020-03-20 Мерк Шарп И Доум Корп. Cyclic di-nucleotide compounds as sting agonists
AR105654A1 (en) 2015-08-24 2017-10-25 Lilly Co Eli ANTIBODIES PD-L1 (LINKING 1 OF PROGRAMMED CELL DEATH)
CN114605548A (en) 2015-09-01 2022-06-10 艾吉纳斯公司 anti-PD-1 antibodies and methods of use thereof
CN108025069B (en) 2015-09-03 2021-11-12 小野药品工业株式会社 Cancer immunopotentiator using allergen-1 antagonist
WO2017049143A1 (en) * 2015-09-18 2017-03-23 Dana-Farber Cancer Institute, Inc. Methods of reducing liver pd-1-expressing cd8+t cells using pd-1 fc fusion proteins that bind fc receptors
EP3352858A4 (en) * 2015-09-21 2019-04-17 Merck Sharp & Dohme Corp. Antibody that binds to human programmed death ligand 2 (pd-l2) and uses thereof
CN108289966A (en) * 2015-09-24 2018-07-17 北卡罗来纳-查佩尔山大学 Method and composition for reducing transfer
MA42925A (en) 2015-09-25 2018-08-01 Hoffmann La Roche ANTI-TIGIT ANTIBODIES AND METHODS OF USE
CN108289953B (en) 2015-09-29 2022-03-11 细胞基因公司 PD-1 binding proteins and methods of use thereof
EP3356551B1 (en) 2015-09-29 2020-09-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for determining the metabolic status of b-lymphomas
BR112018006547A2 (en) * 2015-09-29 2018-12-11 Asia Biotech Pte. Ltd pd-1 antibodies and uses thereof
US20170247454A1 (en) 2015-10-02 2017-08-31 Hoffmann-La Roche Inc. Anti-pd1 antibodies and methods of use
ES2886569T3 (en) 2015-10-02 2021-12-20 Hoffmann La Roche Bispecific antibodies specific for PD1 and TIM3
RU2731202C2 (en) 2015-10-08 2020-08-31 Макродженикс, Инк. Combined therapy for cancer treatment
JP6954648B2 (en) 2015-10-19 2021-10-27 シージー オンコロジー, インコーポレイテッド Treatment of solid tumors or lymphoid tumors with combination therapy
CA2998208A1 (en) 2015-10-22 2017-04-27 Jounce Therapeutics, Inc. Gene signatures for determining icos expression
WO2017074774A1 (en) * 2015-10-28 2017-05-04 Yale University Humanized anti-dkk2 antibody and uses thereof
WO2017075045A2 (en) 2015-10-30 2017-05-04 Mayo Foundation For Medical Education And Research Antibodies to b7-h1
KR20180072821A (en) 2015-11-03 2018-06-29 얀센 바이오테크 인코포레이티드 Antibodies that specifically bind to PD-1 and uses thereof
EP3371311B1 (en) 2015-11-06 2021-07-21 Orionis Biosciences BV Bi-functional chimeric proteins and uses thereof
HRP20221167T1 (en) * 2015-11-12 2022-12-09 Hookipa Biotech Gmbh Arenavirus particles as cancer vaccines
TWI795347B (en) 2015-11-18 2023-03-11 美商必治妥施貴寶公司 Treatment of lung cancer using a combination of an anti-pd-1 antibody and an anti-ctla-4 antibody
WO2017093942A1 (en) 2015-12-01 2017-06-08 Glaxosmithkline Intellectual Property Development Limited Combination treatments and uses and methods thereof
MX2018006477A (en) 2015-12-02 2018-09-03 Agenus Inc Antibodies and methods of use thereof.
TWI704154B (en) 2015-12-03 2020-09-11 英商葛蘭素史克智慧財產發展有限公司 Novel compounds
WO2017098421A1 (en) 2015-12-08 2017-06-15 Glaxosmithkline Intellectual Property Development Limited Benzothiadiazine compounds
TW202208440A (en) 2015-12-14 2022-03-01 美商宏觀基因股份有限公司 Bispecific Molecules Having Immunoreactivity with PD-1 and CTLA-4, and Methods of Use Thereof
US10538497B2 (en) 2015-12-15 2020-01-21 Merck Sharp & Dohme Corp. Compounds as indoleamine 2,3-dioxygenase inhibitors
GB201522311D0 (en) 2015-12-17 2016-02-03 Photocure Asa Use
GB201522309D0 (en) 2015-12-17 2016-02-03 Photocure Asa Use
US11091556B2 (en) 2015-12-18 2021-08-17 Intervet Inc. Caninized human antibodies to human IL-4R alpha
ES2837155T3 (en) 2016-01-04 2021-06-29 Inst Nat Sante Rech Med Use of PD-1 and Tim-3 as a measure of CD8 + cells to predict and treat renal cell carcinoma
CN106943597A (en) 2016-01-07 2017-07-14 博笛生物科技(北京)有限公司 Anti-EGFR for treating tumour is combined
CN106943598A (en) 2016-01-07 2017-07-14 博笛生物科技(北京)有限公司 Anti- HER2 for treating tumour is combined
CN106943596A (en) 2016-01-07 2017-07-14 博笛生物科技(北京)有限公司 Anti-CD 20 for treating tumour is combined
EP3693019A1 (en) 2016-01-08 2020-08-12 Taiho Pharmaceutical Co., Ltd. Anti-tumor agent containing immunomodulator
EP3411409A2 (en) 2016-01-10 2018-12-12 Neotx Therapeutics Ltd. Immunopotentiator enhanced superantigen mediated cancer immunotherapy
AU2017208819B2 (en) 2016-01-22 2023-10-19 MabQuest SA PD1 specific antibodies
US11214617B2 (en) 2016-01-22 2022-01-04 MabQuest SA Immunological reagents
US11661455B2 (en) 2016-02-05 2023-05-30 Orionis Biosciences BV Chimeric protein comprising an interferon alpha 2mutant and a PD-L1 binding moiety
CN108883180B (en) 2016-02-05 2023-07-07 奥里尼斯生物科学私人有限公司 CLEC9A binding agents and uses thereof
BR112018016461A2 (en) 2016-02-12 2019-10-01 Janssen Pharmaceutica Nv antibodies and anti-sight fragments, their uses and their identification methods
US10358496B2 (en) 2016-03-01 2019-07-23 Ralf Kleef Low dose immune checkpoint blockade in metastatic cancer
US11883404B2 (en) 2016-03-04 2024-01-30 Taiho Pharmaceuticals Co., Ltd. Preparation and composition for treatment of malignant tumors
CA3016849A1 (en) 2016-03-07 2017-09-14 Vib Vzw Cd20 binding single domain antibodies
WO2017155981A1 (en) 2016-03-07 2017-09-14 Massachusetts Institute Of Technology Protein-chaperoned t-cell vaccines
WO2017153952A1 (en) 2016-03-10 2017-09-14 Glaxosmithkline Intellectual Property Development Limited 5-sulfamoyl-2-hydroxybenzamide derivatives
WO2017156349A1 (en) 2016-03-10 2017-09-14 Cold Genesys, Inc. Methods of treating solid or lymphatic tumors by combination therapy
WO2017160599A1 (en) 2016-03-14 2017-09-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Use of cd300b antagonists to treat sepsis and septic shock
PT3430141T (en) 2016-03-14 2021-02-25 Hoffmann La Roche Oligonucleotides for reduction of pd-l1 expression
AU2017234163B2 (en) 2016-03-15 2023-01-19 Mersana Therapeutics, Inc. NaPi2b-targeted antibody-drug conjugates and methods of use thereof
US10869923B2 (en) * 2016-03-30 2020-12-22 Microbio Co. Ltd. Combined cancer therapy with immune checkpoint modulators and fermentation products by symbiotic microbiota
EP3436066A1 (en) 2016-04-01 2019-02-06 Checkmate Pharmaceuticals, Inc. Fc receptor-mediated drug delivery
US11209441B2 (en) 2016-04-05 2021-12-28 Bristol-Myers Squibb Company Cytokine profiling analysis
PE20181884A1 (en) 2016-04-07 2018-12-07 Glaxosmithkline Ip Dev Ltd USEFUL HETEROCYCLIC AMIDES AS PROTEIN MODULATORS
KR20180132783A (en) 2016-04-07 2018-12-12 글락소스미스클라인 인털렉츄얼 프로퍼티 디벨로프먼트 리미티드 Heterocyclic amides useful as protein modulators
CR20180537A (en) 2016-04-15 2019-03-04 Immunext Inc ANTIHUMAN VIEW ANTIBODIES AND THEIR USE
US11192914B2 (en) 2016-04-28 2021-12-07 Emory University Alkyne containing nucleotide and nucleoside therapeutic compositions and uses related thereto
CN109328188A (en) 2016-05-05 2019-02-12 葛兰素史密斯克莱知识产权(第2 号)有限公司 2 inhibitor of ZESTE enhancer homologue
EP3455245A2 (en) 2016-05-13 2019-03-20 Orionis Biosciences NV Therapeutic targeting of non-cellular structures
EP3243832A1 (en) 2016-05-13 2017-11-15 F. Hoffmann-La Roche AG Antigen binding molecules comprising a tnf family ligand trimer and pd1 binding moiety
TWI822521B (en) 2016-05-13 2023-11-11 美商再生元醫藥公司 Methods of treating skin cancer by administering a pd-1 inhibitor
EP3454887B1 (en) 2016-05-13 2021-01-20 Orionis Biosciences BV Targeted mutant interferon-beta and uses thereof
WO2017201043A1 (en) * 2016-05-16 2017-11-23 Concert Pharmaceuticals, Inc. Combination therapy for treating cancer
KR102543118B1 (en) 2016-05-27 2023-06-14 아게누스 인코포레이티드 Anti-tim-3 antibodies and methods of use thereof
WO2017209155A1 (en) 2016-05-31 2017-12-07 大鵬薬品工業株式会社 Sulfonamide compound or salt thereof
EP3468960B1 (en) 2016-06-08 2022-03-23 GlaxoSmithKline Intellectual Property Development Limited Chemical compounds as atf4 pathway inhibitors
CN109563034A (en) 2016-06-08 2019-04-02 葛兰素史密斯克莱知识产权发展有限公司 Chemical compound
JP7185530B2 (en) 2016-06-13 2022-12-07 トルク セラピューティクス, インコーポレイテッド Methods and compositions for promoting immune cell function
BR112018076260A2 (en) 2016-06-20 2019-03-26 Kymab Limited antibody or fragment thereof that specifically binds to hpd-11, bispecific antibody or fusion protein, use of an antibody or fragment, method, pharmaceutical composition, modulation method, inhibition method, treatment method, nucleic acid, vector, host and immunocytocin
US9567399B1 (en) * 2016-06-20 2017-02-14 Kymab Limited Antibodies and immunocytokines
US10912287B2 (en) 2016-06-28 2021-02-09 Biocytogen Pharmaceuticals (Beijing) Co., Ltd Genetically modified mice expressing humanized PD-1
EP3478321A4 (en) 2016-06-30 2020-04-22 Oncorus, Inc. Pseudotyped oncolytic viral delivery of therapeutic polypeptides
NZ749997A (en) 2016-07-05 2022-11-25 Beigene Ltd Combination of a pd-l antagonist and a raf inhibitor for treating cancer
CN109789135A (en) 2016-07-20 2019-05-21 葛兰素史密斯克莱知识产权发展有限公司 Isoquinilone derivatives as PERK inhibitor
US11649289B2 (en) * 2016-08-04 2023-05-16 Glaxosmithkline Intellectual Property Development Limited Anti-ICOS and anti-PD-1 antibody combination therapy
CN116640214A (en) 2016-08-09 2023-08-25 科马布有限公司 Isolated antibodies and uses thereof
DK3500299T3 (en) 2016-08-19 2024-01-29 Beigene Switzerland Gmbh Combination of zanubrutinib with an anti-CD20 or an anti-PD-1 antibody for use in the treatment of cancer
WO2018035710A1 (en) 2016-08-23 2018-03-01 Akeso Biopharma, Inc. Anti-ctla4 antibodies
US10372100B2 (en) * 2016-08-29 2019-08-06 Ge Healthcare Bio-Sciences Corp. Manufacturing system for biopharmaceutical products
US10323248B2 (en) 2016-09-01 2019-06-18 Chimera Bioengineering, Inc. Gold optimized CAR T-cells
ES2837755T3 (en) 2016-09-14 2021-07-01 Abbvie Biotherapeutics Inc Anti-PD-1 antibodies
WO2018053434A1 (en) 2016-09-16 2018-03-22 The Johns Hopkins University Protein nanocages with enhanced mucus penetration for targeted tissue and intracellular delivery
JP2019531284A (en) 2016-09-19 2019-10-31 セルジーン コーポレイション Methods of treating immune disorders using PD-1 binding proteins
EP3515943A4 (en) 2016-09-19 2020-05-06 Celgene Corporation Methods of treating vitiligo using pd-1 binding proteins
JP7069177B2 (en) 2016-09-21 2022-05-17 ネクストキュア インコーポレイテッド Antibodies to Siglec-15 and how to use them
CA3037008A1 (en) 2016-09-27 2018-04-05 Oncologie, Inc. Methods for treating cancer with bavituximab based on levels of .beta.2-glycoprotein 1, and assays therefor
SG10202008647TA (en) 2016-10-04 2020-10-29 Merck Sharp & Dohme BENZO[b]THIOPHENE COMPOUNDS AS STING AGONISTS
KR20190062515A (en) 2016-10-06 2019-06-05 화이자 인코포레이티드 Usage of Abelipab for the Treatment of Cancer
CA3037380A1 (en) 2016-10-11 2018-04-19 Agenus Inc. Anti-lag-3 antibodies and methods of use thereof
BR112019007145A2 (en) 2016-10-14 2019-07-02 Eisai R&D Man Co Ltd combination of a pd-1 antagonist and eribulin for the treatment of urothelial cancer
EP3529264B1 (en) 2016-10-24 2022-03-09 Orionis Biosciences BV Targeted mutant interferon-gamma and uses thereof
WO2018079287A1 (en) 2016-10-26 2018-05-03 デンカ株式会社 MUTANT HUMAN DNA POLYMERASE ε
BR112019008859A2 (en) 2016-11-01 2019-07-09 Anaptysbio Inc antibodies directed against programmed death 1 (pd-1)
WO2018083087A2 (en) 2016-11-02 2018-05-11 Glaxosmithkline Intellectual Property (No.2) Limited Binding proteins
TW202321301A (en) 2016-11-02 2023-06-01 美商永斯醫療股份有限公司 Antibodies to pd-1 and uses thereof
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
US11471515B2 (en) 2016-11-09 2022-10-18 The Brigham And Women's Hospital, Inc. Restoration of tumor suppression using MRNA-based delivery system
US11279694B2 (en) 2016-11-18 2022-03-22 Sumitomo Dainippon Pharma Oncology, Inc. Alvocidib prodrugs and their use as protein kinase inhibitors
EP3541825A1 (en) 2016-11-21 2019-09-25 Idenix Pharmaceuticals LLC. Cyclic phosphate substituted nucleoside derivatives for the treatment of liver diseases
WO2018098352A2 (en) 2016-11-22 2018-05-31 Jun Oishi Targeting kras induced immune checkpoint expression
US11135307B2 (en) 2016-11-23 2021-10-05 Mersana Therapeutics, Inc. Peptide-containing linkers for antibody-drug conjugates
WO2018102427A1 (en) 2016-11-29 2018-06-07 Boston Biomedical, Inc. Naphthofuran derivatives, preparation, and methods of use thereof
WO2018102536A1 (en) 2016-11-30 2018-06-07 Oncomed Pharmaceuticals, Inc. Methods for treatment of cancer comprising tigit-binding agents
BR112019011350A2 (en) 2016-12-01 2019-10-22 Glaxosmithkline Ip Dev Ltd combination therapy
CA3045241A1 (en) 2016-12-01 2018-06-07 Glaxosmithkline Intellectual Property Development Limited Combination therapy
ES2963226T3 (en) 2016-12-07 2024-03-26 Agenus Inc ANTI-CTLA-4 antibodies and methods of their use
PE20190921A1 (en) 2016-12-07 2019-06-26 Agenus Inc ANTIBODIES AND METHODS OF THEIR USE
WO2018112364A1 (en) 2016-12-16 2018-06-21 Evelo Biosciences, Inc. Combination therapies for treating melanoma
WO2018112360A1 (en) 2016-12-16 2018-06-21 Evelo Biosciences, Inc. Combination therapies for treating cancer
US11371995B2 (en) 2016-12-28 2022-06-28 Daiichi Sankyo Company, Limited Method for selecting individuals to be administered immune checkpoint inhibitor
WO2018129559A1 (en) 2017-01-09 2018-07-12 Tesaro, Inc. Methods of treating cancer with anti-pd-1 antibodies
WO2018132739A2 (en) 2017-01-13 2018-07-19 Agenus Inc. T cell receptors that bind to ny-eso-1 and methods of use thereof
WO2018134279A1 (en) 2017-01-18 2018-07-26 Pieris Pharmaceuticals Gmbh Novel fusion polypeptides specific for lag-3 and pd-1
EP3573989A4 (en) 2017-01-25 2020-11-18 Beigene, Ltd. Crystalline forms of (s) -7- (1- (but-2-ynoyl) piperidin-4-yl) -2- (4-phenoxyphenyl) -4, 5, 6, 7-tetrahy dropyrazolo [1, 5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
WO2018141964A1 (en) 2017-02-06 2018-08-09 Orionis Biosciences Nv Targeted chimeric proteins and uses thereof
JP2020505955A (en) 2017-02-06 2020-02-27 オリオンズ バイオサイエンス インコーポレイテッド Targeted modified interferon and uses thereof
WO2018146074A1 (en) 2017-02-07 2018-08-16 Vib Vzw Immune-cell targeted bispecific chimeric proteins and uses thereof
EP3582855A1 (en) 2017-02-15 2019-12-25 GlaxoSmithKline Intellectual Property Development Limited Combination treatment for cancer
CA3052767A1 (en) 2017-02-27 2018-08-30 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
TW201834697A (en) 2017-02-28 2018-10-01 美商梅爾莎納醫療公司 Combination therapies of her2-targeted antibody-drug conjugates
CA3054733A1 (en) 2017-02-28 2018-09-07 Sanofi Therapeutic rna
EP3592769A1 (en) 2017-03-09 2020-01-15 Genmab A/S Antibodies against pd-l1
US20200150125A1 (en) 2017-03-12 2020-05-14 Yeda Research And Development Co., Ltd. Methods of diagnosing and prognosing cancer
WO2018167780A1 (en) 2017-03-12 2018-09-20 Yeda Research And Development Co. Ltd. Methods of prognosing and treating cancer
EP3600427A1 (en) 2017-03-24 2020-02-05 INSERM - Institut National de la Santé et de la Recherche Médicale Methods and compositions for treating melanoma
US11352408B2 (en) 2017-03-31 2022-06-07 Toagosei Co., Ltd. Antitumor peptide having PD-1 signal sequence and utilization thereof
CA3055769A1 (en) 2017-04-03 2018-10-11 Oncologie, Inc. Methods for treating cancer using ps-targeting antibodies with immuno-oncology agents
RU2761377C2 (en) 2017-04-03 2021-12-07 Ф. Хоффманн-Ля Рош Аг Immunoconjugates of antibody to pd-1 with il-2 or il-15 mutant
AU2018247794A1 (en) 2017-04-05 2019-08-22 F. Hoffmann-La Roche Ag Bispecific antibodies specifically binding to PD1 and LAG3
US11603407B2 (en) 2017-04-06 2023-03-14 Regeneron Pharmaceuticals, Inc. Stable antibody formulation
MA50956A (en) 2017-04-13 2020-10-14 Agenus Inc ANTI-CD137 ANTIBODIES AND RELATED METHODS OF USE
EP3612201B1 (en) 2017-04-21 2023-10-25 Sillajen, Inc. Oncolytic vaccinia virus and checkpoint inhibitor combination therapy
CN108794467A (en) 2017-04-27 2018-11-13 博笛生物科技有限公司 2- amino-quinoline derivatives
AR111651A1 (en) 2017-04-28 2019-08-07 Novartis Ag CONJUGATES OF ANTIBODIES THAT INCLUDE TOLL TYPE RECEIVER AGONISTS AND COMBINATION THERAPIES
ES2952961T3 (en) 2017-05-01 2023-11-07 Agenus Inc Anti-TIGIT antibodies and methods of their use
KR20200003107A (en) 2017-05-02 2020-01-08 머크 샤프 앤드 돔 코포레이션 Preparation of Anti-LAG3 Antibodies, and Co-Formulations of Anti-LAG3 Antibodies and Anti-PD-1 Antibodies
JOP20190260A1 (en) 2017-05-02 2019-10-31 Merck Sharp & Dohme Stable formulations of programmed death receptor 1 (pd-1) antibodies and methods of use thereof
WO2018208667A1 (en) 2017-05-12 2018-11-15 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as sting agonists
US10646464B2 (en) 2017-05-17 2020-05-12 Boston Biomedical, Inc. Methods for treating cancer
CN110913911A (en) 2017-05-18 2020-03-24 特沙诺有限公司 Combination therapy for the treatment of cancer
JP6738962B2 (en) 2017-05-26 2020-08-12 大鵬薬品工業株式会社 Antitumor effect enhancer using a novel biphenyl compound
JP6843979B2 (en) 2017-05-26 2021-03-17 大鵬薬品工業株式会社 New biphenyl compound or salt thereof
JOP20190279A1 (en) 2017-05-31 2019-11-28 Novartis Ag Crystalline forms of 5-bromo-2,6-di(1 h-pyrazol-1-yl)pyrimidin-4-amine and new salts
AU2018278327B2 (en) 2017-06-01 2023-03-16 Cytomx Therapeutics, Inc. Activatable anti-pdl1 antibodies and methods of use thereof
WO2018222989A1 (en) 2017-06-02 2018-12-06 The Penn State Research Foundation Ceramide nanoliposomes, compositions and methods of using for immunotherapy
WO2018225093A1 (en) 2017-06-07 2018-12-13 Glaxosmithkline Intellectual Property Development Limited Chemical compounds as atf4 pathway inhibitors
WO2018226336A1 (en) 2017-06-09 2018-12-13 Providence Health & Services - Oregon Utilization of cd39 and cd103 for identification of human tumor reactive cells for treatment of cancer
JP2020522555A (en) 2017-06-09 2020-07-30 グラクソスミスクライン、インテレクチュアル、プロパティー、ディベロップメント、リミテッドGlaxosmithkline Intellectual Property Development Limited Combination therapy
CA3066756A1 (en) 2017-06-15 2018-12-20 Cancer Advances Inc. Compositions and methods for inducing humoral and cellular immunities against tumors and cancer
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
GB201709808D0 (en) 2017-06-20 2017-08-02 Kymab Ltd Antibodies
US11312783B2 (en) 2017-06-22 2022-04-26 Novartis Ag Antibody molecules to CD73 and uses thereof
US20200172628A1 (en) 2017-06-22 2020-06-04 Novartis Ag Antibody molecules to cd73 and uses thereof
TW201904993A (en) 2017-06-22 2019-02-01 瑞士商諾華公司 Use of IL-1β binding antibody
WO2018235056A1 (en) 2017-06-22 2018-12-27 Novartis Ag Il-1beta binding antibodies for use in treating cancer
BR112019027025A2 (en) 2017-06-23 2020-06-30 Birdie Biopharmaceuticals, Inc. pharmaceutical compositions
EP3645569A4 (en) 2017-06-26 2021-03-24 BeiGene, Ltd. Immunotherapy for hepatocellular carcinoma
US20200223924A1 (en) 2017-06-27 2020-07-16 Novartis Ag Dosage regimens for anti-tim-3 antibodies and uses thereof
JP7264812B2 (en) 2017-06-30 2023-04-25 大鵬薬品工業株式会社 Cancer chemotherapy using azabicyclic compounds
CA3068753A1 (en) 2017-07-03 2019-01-10 Glaxosmithkline Intellectual Property Development Limited N-(3-(2-(4-chlorophenoxy)acetamido)bicyclo[1.1.1]pentan-1-yl)-2-cyclobutane-1-carboxamide derivatives and related compounds as atf4 inhibitors for treating cancer and other diseases
EP3649108A1 (en) 2017-07-03 2020-05-13 GlaxoSmithKline Intellectual Property Development Limited 2-(4-chlorophenoxy)-n-((1 -(2-(4-chlorophenoxy)ethynazetidin-3-yl)methyl)acetamide derivatives and related compounds as atf4 inhibitors for treating cancer and other diseases
CA3070095A1 (en) 2017-07-20 2019-01-24 Novartis Ag Dosage regimens of anti-lag-3 antibodies and uses thereof
WO2019021208A1 (en) 2017-07-27 2019-01-31 Glaxosmithkline Intellectual Property Development Limited Indazole derivatives useful as perk inhibitors
EP3661499A4 (en) 2017-08-04 2021-04-21 Merck Sharp & Dohme Corp. COMBINATIONS OF PD-1 ANTAGONISTS AND BENZO[b
SG11202000198QA (en) 2017-08-04 2020-02-27 Genmab As Binding agents binding to pd-l1 and cd137 and use thereof
CA3071538A1 (en) 2017-08-04 2019-02-07 Merck Sharp & Dohme Corp. Benzo[b]thiophene sting agonists for cancer treatment
US11498966B2 (en) 2017-08-09 2022-11-15 Orionis Biosciences Inc. PD-1 and PD-L1 binding agents
US11566072B2 (en) 2017-08-09 2023-01-31 Orionis Biosciences, Inc. CD8 binding agents
KR20200078481A (en) 2017-08-18 2020-07-01 아다스트라 파마수티컬스, 인코포레이티드 Polymorphic form of TG02
SG11202001319QA (en) 2017-09-04 2020-03-30 Agenus Inc T cell receptors that bind to mixed lineage leukemia (mll)-specific phosphopeptides and methods of use thereof
TW201922721A (en) 2017-09-07 2019-06-16 英商葛蘭素史克智慧財產發展有限公司 Chemical compounds
EP3679070A1 (en) 2017-09-07 2020-07-15 Augusta University Research Institute, Inc. Antibodies to programmed cell death protein 1
WO2019053617A1 (en) 2017-09-12 2019-03-21 Glaxosmithkline Intellectual Property Development Limited Chemical compounds
JP7196160B2 (en) 2017-09-12 2022-12-26 スミトモ ファーマ オンコロジー, インコーポレイテッド Treatment Regimens for Cancers Insensitive to BCL-2 Inhibitors Using the MCL-1 Inhibitor Albocidib
WO2019061324A1 (en) 2017-09-29 2019-04-04 Curis Inc. Crystal forms of immunomodulators
EP3697442A4 (en) 2017-09-30 2021-07-07 Tesaro, Inc. Combination therapies for treating cancer
US11377440B2 (en) 2017-10-05 2022-07-05 Glaxosmithkline Intellectual Property Development Limited Modulators of stimulator of interferon genes (STING)
AU2018344902B2 (en) 2017-10-05 2021-06-03 Glaxosmithkline Intellectual Property Development Limited Modulators of stimulator of interferon genes (STING) useful in treating HIV
JP2020536869A (en) 2017-10-06 2020-12-17 テサロ, インコーポレイテッド Combination therapy and its use
BR112020006669A2 (en) 2017-10-11 2020-09-24 Aurigene Discovery Technologies Limited crystalline forms of 3-substituted 1,2,4-oxadiazole
WO2019072241A1 (en) 2017-10-13 2019-04-18 Beijing Biocytogen Co., Ltd Genetically modified non-human animal with human or chimeric pd-1
JP2020536894A (en) 2017-10-15 2020-12-17 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Tumor treatment
EP3700933A1 (en) 2017-10-25 2020-09-02 Novartis AG Antibodies targeting cd32b and methods of use thereof
US11718679B2 (en) 2017-10-31 2023-08-08 Compass Therapeutics Llc CD137 antibodies and PD-1 antagonists and uses thereof
EP3703692A4 (en) 2017-11-01 2021-04-28 Merck Sharp & Dohme Corp. Novel substituted tetrahydroquinolin compounds as indoleamine 2,3-dioxygenase (ido) inhibitors
EP3703683A1 (en) 2017-11-03 2020-09-09 Aurigene Discovery Technologies Limited Dual inhibitors of tim-3 and pd-1 pathways
JP7378395B2 (en) 2017-11-06 2023-11-13 オーリジーン オンコロジー リミテッド Conjoint therapy for immunomodulation
WO2019093383A1 (en) * 2017-11-07 2019-05-16 ジェイファーマ株式会社 Method for predicting efficacy of anti-pd-1 antibody or anti-pd-l1 antibody therapy, method for evaluating cancer grade, and method for enhancing efficacy of anti-pd-1 antibody or anti-pd-l1 antibody therapy
EP3709986B1 (en) 2017-11-14 2023-11-01 Merck Sharp & Dohme LLC Novel substituted biaryl compounds as indoleamine 2,3-dioxygenase (ido) inhibitors
EP3710444B1 (en) 2017-11-14 2023-04-12 Merck Sharp & Dohme LLC Novel substituted biaryl compounds as indoleamine 2,3-dioxygenase (ido) inhibitors
WO2019104289A1 (en) 2017-11-27 2019-05-31 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
CN111801334B (en) 2017-11-29 2023-06-09 百济神州瑞士有限责任公司 Treatment of indolent or invasive B-cell lymphomas using combinations comprising BTK inhibitors
AU2018375738A1 (en) 2017-11-30 2020-06-11 Novartis Ag BCMA-targeting chimeric antigen receptor, and uses thereof
KR102214029B1 (en) 2017-12-01 2021-02-08 에스비아이 파마 가부시키가이샤 Pharmaceutical composition for enhancing anti-tumor effect by immune checkpoint inhibitor
US11946094B2 (en) 2017-12-10 2024-04-02 Augusta University Research Institute, Inc. Combination therapies and methods of use thereof
US11629189B2 (en) 2017-12-19 2023-04-18 Kymab Limited Bispecific antibody for ICOS and PD-L1
EA202091521A1 (en) 2017-12-19 2020-10-22 Дзе Рокфеллер Юниверсити HUMAN Fc IgG DOMAIN OPTIONS WITH IMPROVED EFFECTIVE FUNCTION
EP3727401A4 (en) 2017-12-20 2022-04-06 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as sting agonists
TW201929908A (en) 2017-12-21 2019-08-01 美商梅爾莎納醫療公司 Pyrrolobenzodiazepine antibody conjugates
WO2019133847A1 (en) 2017-12-29 2019-07-04 Oncorus, Inc. Oncolytic viral delivery of therapeutic polypeptides
WO2019134946A1 (en) 2018-01-04 2019-07-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma resistant
US11324774B2 (en) 2018-01-05 2022-05-10 Augusta University Research Institute, Inc. Compositions of oral alkaline salts and metabolic acid inducers and uses thereof
EP3737408A1 (en) 2018-01-08 2020-11-18 Novartis AG Immune-enhancing rnas for combination with chimeric antigen receptor therapy
EP3743447B1 (en) 2018-01-23 2024-03-27 Nextcure, Inc. B7-h4 antibodies and methods of use thereof
US20200354457A1 (en) 2018-01-31 2020-11-12 Hoffmann-La Roche Inc. Bispecific antibodies comprising an antigen-binding site binding to lag3
AU2019215031A1 (en) 2018-01-31 2020-08-20 Novartis Ag Combination therapy using a chimeric antigen receptor
AU2019217875A1 (en) 2018-02-06 2020-08-20 Icahn School Of Medicine At Mount Sinai Repeat RNA as biomarkers of tumor immune response
US20200399383A1 (en) 2018-02-13 2020-12-24 Novartis Ag Chimeric antigen receptor therapy in combination with il-15r and il15
US20210080467A1 (en) 2018-02-21 2021-03-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of sk1 as biomarker for predicting response to immunecheckpoint inhibitors
JP7110369B2 (en) 2018-02-23 2022-08-01 ユーキュア・(ベイジン)・バイオファーマ・カンパニー・リミテッド Anti-PD-1 antibody and use thereof
WO2019169229A1 (en) 2018-03-01 2019-09-06 Nextcure, Inc. Klrg1 binding compositions and methods of use thereof
NZ767596A (en) 2018-03-14 2022-01-28 Surface Oncology Inc Antibodies that bind cd39 and uses thereof
SG11202008435SA (en) 2018-03-19 2020-10-29 Taiho Pharmaceutical Co Ltd Pharmaceutical composition including sodium alkyl sulfate
WO2019183499A1 (en) 2018-03-22 2019-09-26 Surface Oncology, Inc. Anti-il-27 antibodies and uses thereof
WO2019195063A1 (en) 2018-04-03 2019-10-10 Merck Sharp & Dohme Corp. Aza-benzothiophene compounds as sting agonists
AU2019248545B2 (en) 2018-04-03 2022-08-11 Merck Sharp & Dohme Llc Benzothiophenes and related compounds as sting agonists
WO2019193540A1 (en) 2018-04-06 2019-10-10 Glaxosmithkline Intellectual Property Development Limited Heteroaryl derivatives of formula (i) as atf4 inhibitors
WO2019193541A1 (en) 2018-04-06 2019-10-10 Glaxosmithkline Intellectual Property Development Limited Bicyclic aromatic ring derivatives of formula (i) as atf4 inhibitors
US20210147547A1 (en) 2018-04-13 2021-05-20 Novartis Ag Dosage Regimens For Anti-Pd-L1 Antibodies And Uses Thereof
US20210198374A1 (en) 2018-04-17 2021-07-01 Celldex Therapeutics, Inc. Anti-cd27 and anti-pd-l1 antibodies and bispecific constructs
US10907161B2 (en) 2018-04-19 2021-02-02 Checkmate Pharmaceuticals, Inc. Synthetic RIG-I-like receptor agonists
US11542505B1 (en) 2018-04-20 2023-01-03 Merck Sharp & Dohme Llc Substituted RIG-I agonists: compositions and methods thereof
AU2019261451A1 (en) 2018-04-26 2020-12-03 Agenus Inc. Heat shock protein-binding peptide compositions and methods of use thereof
EP3802788A4 (en) 2018-05-15 2022-02-16 Duke University Systems and methods for genetic manipulation of akkermansia species
GB201807924D0 (en) 2018-05-16 2018-06-27 Ctxt Pty Ltd Compounds
UY38247A (en) 2018-05-30 2019-12-31 Novartis Ag ANTIBODIES AGAINST ENTPD2, COMBINATION THERAPIES AND METHODS OF USE OF ANTIBODIES AND COMBINATION THERAPIES
WO2019232319A1 (en) 2018-05-31 2019-12-05 Peloton Therapeutics, Inc. Compositions and methods for inhibiting cd73
WO2019232244A2 (en) 2018-05-31 2019-12-05 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2019231870A1 (en) 2018-05-31 2019-12-05 Merck Sharp & Dohme Corp. Novel substituted [1.1.1] bicyclo compounds as indoleamine 2,3-dioxygenase inhibitors
PE20210320A1 (en) 2018-06-01 2021-02-16 Novartis Ag BINDING MOLECULES AGAINST BCMA AND THE USES OF THEM
JP2021529814A (en) 2018-07-09 2021-11-04 グラクソスミスクライン、インテレクチュアル、プロパティー、ディベロップメント、リミテッドGlaxosmithkline Intellectual Property Development Limited Chemical compound
SG11202100208VA (en) 2018-07-09 2021-02-25 Precigen Inc Fusion constructs and methods of using thereof
JOP20210001A1 (en) 2018-07-10 2021-01-05 Novartis Ag 3-(5-hydroxy-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of ikaros family zinc finger 2 (ikzf2)-dependent diseases
AR116109A1 (en) 2018-07-10 2021-03-31 Novartis Ag DERIVATIVES OF 3- (5-AMINO-1-OXOISOINDOLIN-2-IL) PIPERIDINE-2,6-DIONA AND USES OF THE SAME
EP3827020A1 (en) 2018-07-24 2021-06-02 Amgen Inc. Combination of lilrb1/2 pathway inhibitors and pd-1 pathway inhibitors
WO2020021061A1 (en) 2018-07-26 2020-01-30 Pieris Pharmaceuticals Gmbh Humanized anti-pd-1 antibodies and uses thereof
WO2020030571A1 (en) 2018-08-06 2020-02-13 Glaxosmithkline Intellectual Property Development Limited Combinations of a pd-1 antibody and a tlr4 modulator and uses thereof
WO2020031107A1 (en) 2018-08-08 2020-02-13 Glaxosmithkline Intellectual Property Development Limited Chemical compounds
EP3843849A1 (en) 2018-08-27 2021-07-07 Pieris Pharmaceuticals GmbH Combination therapies comprising cd137/her2 bispecific agents and pd-1 axis inhibitors and uses thereof
WO2020044206A1 (en) 2018-08-29 2020-03-05 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors for use in the treatment cancer
US20210340279A1 (en) 2018-08-31 2021-11-04 Yale University Compositions and methods of using cell-penetrating antibodies in combination with immune checkpoint modulators
EP3858368B1 (en) 2018-09-25 2023-11-29 National Cerebral and Cardiovascular Center Antitumor effect potentiator
EP3865141A4 (en) 2018-10-09 2022-06-08 Biocomo Incorporation Anticancer agent, pharmaceutical composition for cancer treatment, and kit
JP2022504468A (en) 2018-10-17 2022-01-13 バイオラインアールエックス・リミテッド Treatment of metastatic pancreatic adenocarcinoma
CN113453715A (en) 2018-10-22 2021-09-28 葛兰素史克知识产权开发有限公司 Administration of drugs
AU2019369340A1 (en) 2018-10-29 2021-05-20 Mersana Therapeutics, Inc. Cysteine engineered antibody-drug conjugates with peptide-containing linkers
EP3873532A1 (en) 2018-10-31 2021-09-08 Novartis AG Dc-sign antibody drug conjugates
EP3873464A4 (en) 2018-11-01 2022-06-08 Merck Sharp & Dohme Corp. Novel substituted pyrazole compounds as indoleamine 2,3-dioxygenase inhibitors
EP3877366A4 (en) 2018-11-06 2022-08-24 Merck Sharp & Dohme Corp. Novel substituted tricyclic compounds as indoleamine 2,3-dioxygenase inhibitors
CA3118789A1 (en) 2018-11-06 2020-05-14 Genmab A/S Antibody formulation
KR20210107628A (en) 2018-11-13 2021-09-01 콤파스 테라퓨틱스 엘엘씨 Multispecific binding constructs for checkpoint molecules and uses thereof
US20210388091A1 (en) 2018-11-14 2021-12-16 Regeneron Pharmaceuticals, Inc. Intralesional administration of pd-1 inhibitors for treating skin cancer
TW202028222A (en) 2018-11-14 2020-08-01 美商Ionis製藥公司 Modulators of foxp3 expression
EP3880202A2 (en) 2018-11-16 2021-09-22 ArQule, Inc. Pharmaceutical combination for treatment of cancer
CN113227142B (en) 2018-11-19 2022-11-29 百奥赛图(北京)医药科技股份有限公司 anti-PD-1 antibodies and uses thereof
US20220040184A1 (en) 2018-11-20 2022-02-10 Merck Sharp Dohme Corp. Substituted amino triazolopyrimidine and amino triazolopyrazine adenosine receptor antagonists, pharmaceutical compositions and their use
WO2020104496A1 (en) 2018-11-20 2020-05-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Bispecific antibody targeting transferrin receptor 1 and soluble antigen
WO2020104479A1 (en) 2018-11-20 2020-05-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating cancers and resistant cancers with anti transferrin receptor 1 antibodies
US20210395255A1 (en) 2018-11-20 2021-12-23 Merck Sharp & Dohme Corp. Substituted amino triazolopyrimidine and amino triazolopyrazine adenosine receptor antagonists, pharmaceutical compositions and their use
WO2020112581A1 (en) 2018-11-28 2020-06-04 Merck Sharp & Dohme Corp. Novel substituted piperazine amide compounds as indoleamine 2, 3-dioxygenase (ido) inhibitors
AR117164A1 (en) 2018-11-30 2021-07-14 Merck Sharp & Dohme DERIVATIVES OF AMINO-TRIAZOLOQUINAZOLINE 9-SUBSTITUTED AS ANTAGONISTS OF THE ADENOSINE RECEPTOR, PHARMACEUTICAL COMPOSITIONS AND THEIR USE FOR THE TREATMENT OF VARIOUS TYPES OF CANCER
JP7406556B2 (en) 2018-11-30 2023-12-27 グラクソスミスクライン、インテレクチュアル、プロパティー、ディベロップメント、リミテッド Compounds useful in HIV therapy
WO2020117988A1 (en) 2018-12-04 2020-06-11 Tolero Pharmaceuticals, Inc. Cdk9 inhibitors and polymorphs thereof for use as agents for treatment of cancer
WO2020115261A1 (en) 2018-12-07 2020-06-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
TW202039459A (en) 2018-12-11 2020-11-01 美商施萬生物製藥研發 Ip有限責任公司 Alk5 inhibitors
WO2020120592A1 (en) 2018-12-12 2020-06-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for predicting and treating melanoma
GB201820547D0 (en) 2018-12-17 2019-01-30 Oxford Univ Innovation Modified antibodies
US20220064332A1 (en) 2018-12-19 2022-03-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating cancers by immuno-modulation using antibodies against cathespin-d
JP2022514315A (en) 2018-12-20 2022-02-10 ノバルティス アーゲー Dosage regimens and drug combinations containing 3- (1-oxoisoindoline-2-yl) piperidine-2,6-dione derivatives
WO2020128613A1 (en) 2018-12-21 2020-06-25 Novartis Ag Use of il-1beta binding antibodies
JP2022515188A (en) 2018-12-21 2022-02-17 エイム・イムノテック・インコーポレイテッド Compositions and Methods for Cancer Treatment
CA3119582A1 (en) 2018-12-21 2020-06-25 Novartis Ag Use of il-1.beta. binding antibodies
WO2020127885A1 (en) 2018-12-21 2020-06-25 INSERM (Institut National de la Santé et de la Recherche Médicale) Compositions for treating cancers and resistant cancers
CA3118182A1 (en) 2018-12-21 2020-06-25 Onxeo New conjugated nucleic acid molecules and their uses
WO2020128637A1 (en) 2018-12-21 2020-06-25 Novartis Ag Use of il-1 binding antibodies in the treatment of a msi-h cancer
KR20210107730A (en) 2018-12-21 2021-09-01 노파르티스 아게 Use of IL-1 beta antibodies in the treatment or prevention of myelodysplastic syndrome
CN109593135B (en) * 2018-12-29 2021-01-15 百奥赛图江苏基因生物技术有限公司 Anti-human PD-L1 monoclonal antibody and application thereof
CN111423510B (en) 2019-01-10 2024-02-06 迈威(上海)生物科技股份有限公司 Recombinant anti-human PD-1 antibody and application thereof
KR20210143718A (en) 2019-01-17 2021-11-29 조지아 테크 리서치 코포레이션 Drug Delivery Systems Containing Oxidized Cholesterol
KR20200089645A (en) 2019-01-17 2020-07-27 오토텔릭바이오 주식회사 A composition for treating cancer
EP3918332A1 (en) 2019-01-30 2021-12-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for identifying whether a subject suffering from a cancer will achieve a response with an immune-checkpoint inhibitor
EP3917964A1 (en) 2019-02-01 2021-12-08 GlaxoSmithKline Intellectual Property Development Limited Combination treatments for cancer comprising belantamab mafodotin and an anti ox40 antibody and uses and methods thereof
WO2020161083A1 (en) 2019-02-04 2020-08-13 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for modulating blood-brain barrier
WO2020167990A1 (en) 2019-02-12 2020-08-20 Tolero Pharmaceuticals, Inc. Formulations comprising heterocyclic protein kinase inhibitors
WO2020165370A1 (en) 2019-02-13 2020-08-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for selecting a cancer treatment in a subject suffering from cancer
EP3924521A4 (en) 2019-02-15 2023-03-29 IncellDx, Inc. Assaying bladder-associated samples, identifying and treating bladder-associated neoplasia, and kits for use therein
WO2020165834A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
CN113490528A (en) 2019-02-15 2021-10-08 诺华股份有限公司 3- (1-oxo-5- (piperidine-4-yl) isoindoline-2-yl) piperidine-2, 6-dione derivatives and uses thereof
MX2021010228A (en) 2019-02-28 2021-10-26 Regeneron Pharma Administration of pd-1 inhibitors for treating skin cancer.
KR20210136071A (en) 2019-03-06 2021-11-16 리제너론 파아마슈티컬스, 인크. IL-4/IL-13 Pathway Inhibitors for Enhanced Efficacy in Treating Cancer
WO2020183011A1 (en) 2019-03-14 2020-09-17 Institut Curie Htr1d inhibitors and uses thereof in the treatment of cancer
WO2020191326A1 (en) 2019-03-20 2020-09-24 Sumitomo Dainippon Pharma Oncology, Inc. Treatment of acute myeloid leukemia (aml) with venetoclax failure
JP2022519923A (en) 2019-03-22 2022-03-25 スミトモ ダイニッポン ファーマ オンコロジー, インコーポレイテッド Compositions comprising a PKM2 modulator and methods of treatment using it
MX2021011753A (en) 2019-03-26 2022-01-31 Univ Michigan Regents Small molecule degraders of stat3.
CN113767115A (en) 2019-03-28 2021-12-07 奥里尼斯生物科学股份有限公司 Therapeutic interferon alpha 1 proteins
JP2022527481A (en) 2019-03-29 2022-06-02 アンスティテュ・クリー Interleukin-2 variant with altered bioactivity
EP3947403A1 (en) 2019-03-29 2022-02-09 The Regents Of The University Of Michigan Stat3 protein degraders
US20220177465A1 (en) 2019-04-04 2022-06-09 Merck Sharp & Dohme Corp. Inhibitors of histone deacetylase-3 useful for the treatment of cancer, inflammation, neurodegeneration diseases and diabetes
EP3725370A1 (en) 2019-04-19 2020-10-21 ImmunoBrain Checkpoint, Inc. Modified anti-pd-l1 antibodies and methods and uses for treating a neurodegenerative disease
US20220220565A1 (en) 2019-04-30 2022-07-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
KR20220009428A (en) 2019-05-15 2022-01-24 네오티엑스 테라퓨틱스 엘티디. cancer treatment
JP2022533390A (en) 2019-05-16 2022-07-22 スティングセラ インコーポレイテッド Oxoacridinyl acetic acid derivative and method of use
US20220251079A1 (en) 2019-05-16 2022-08-11 Stingthera, Inc. Benzo[b][1,8]naphthyridine acetic acid derivatives and methods of use
US20220241412A1 (en) 2019-05-24 2022-08-04 Pfizer Inc. Combination therapies using cdk inhibitors
EP3978017A4 (en) 2019-05-29 2023-06-28 Taiho Pharmaceutical Co., Ltd. Combination cancer therapy using sulfonamide compound and immunomodulatory
US11246906B2 (en) 2019-06-11 2022-02-15 Alkermes Pharma Ireland Limited Compositions and methods for subcutaneous administration of cancer immunotherapy
CA3141414A1 (en) 2019-06-12 2020-12-17 Vanderbilt University Dibenzylamines as amino acid transport inhibitors
WO2020252353A1 (en) 2019-06-12 2020-12-17 Vanderbilt University Amino acid transport inhibitors and the uses thereof
CN114206379A (en) 2019-06-18 2022-03-18 爱尔兰詹森科学公司 Combination of Hepatitis B Virus (HBV) vaccine and anti-PD-1 antibody
WO2020255011A1 (en) 2019-06-18 2020-12-24 Janssen Sciences Ireland Unlimited Company Combination of hepatitis b virus (hbv) vaccines and anti-pd-1 or anti-pd-l1 antibody
JP7329173B2 (en) 2019-06-25 2023-08-18 東亞合成株式会社 Anti-tumor peptide and its use
BR112021025476A2 (en) 2019-06-26 2022-10-11 Glaxosmithkline Ip Dev Ltd IL1RAP-BINDING PROTEINS
EP3990635A1 (en) 2019-06-27 2022-05-04 Rigontec GmbH Design method for optimized rig-i ligands
EP3994132A1 (en) 2019-07-03 2022-05-11 Sumitomo Dainippon Pharma Oncology, Inc. Tyrosine kinase non-receptor 1 (tnk1) inhibitors and uses thereof
US20220356255A1 (en) * 2019-07-15 2022-11-10 Capella Bioscience Ltd Anti-pd-l1 antibodies
CN114127073A (en) 2019-07-16 2022-03-01 美国密歇根州立大学试剂中心 Imidazopyrimidines as EED inhibitors and uses thereof
GB201910304D0 (en) 2019-07-18 2019-09-04 Ctxt Pty Ltd Compounds
GB201910305D0 (en) 2019-07-18 2019-09-04 Ctxt Pty Ltd Compounds
US11083705B2 (en) 2019-07-26 2021-08-10 Eisai R&D Management Co., Ltd. Pharmaceutical composition for treating tumor
TW202120500A (en) 2019-08-02 2021-06-01 美商梅爾莎納醫療公司 Sting agonist compounds and methods of use
US20220296705A1 (en) 2019-08-09 2022-09-22 Riken Combined use of artificial adjuvant vector cells and an immunostimulant
CN114641337A (en) 2019-08-27 2022-06-17 密歇根大学董事会 CEREBLON E3 ligase inhibitors
TW202122420A (en) 2019-08-30 2021-06-16 美商艾吉納斯公司 Anti-cd96 antibodies and methods of use thereof
WO2021043961A1 (en) 2019-09-06 2021-03-11 Glaxosmithkline Intellectual Property Development Limited Dosing regimen for the treatment of cancer with an anti icos agonistic antibody and chemotherapy
WO2021048292A1 (en) 2019-09-11 2021-03-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
AU2020351122A1 (en) 2019-09-16 2022-02-17 Surface Oncology, Inc. Anti-CD39 antibody compositions and methods
US20220380314A1 (en) 2019-09-17 2022-12-01 Bial - R&D Investments, S.A. Substituted, saturated and unsaturated n-heterocyclic carboxamides and related compounds for their use in the treatment of medical disorders
EP4031531A1 (en) 2019-09-17 2022-07-27 Bial-R&D Investments, S.A. Substituted imidazole carboxamides and their use in the treatment of medical disorders
KR20220100859A (en) 2019-09-17 2022-07-18 비알 - 알&디 인베스트먼츠, 에스.에이. Substituted N-heterocyclic carboxamides as acid ceramidase inhibitors, and their use as medicaments
TW202124446A (en) 2019-09-18 2021-07-01 瑞士商諾華公司 Combination therapies with entpd2 antibodies
JP2022548881A (en) 2019-09-18 2022-11-22 ノバルティス アーゲー ENTPD2 Antibodies, Combination Therapy and Methods of Using Antibodies and Combination Therapy
CN115023267A (en) 2019-09-19 2022-09-06 密歇根大学董事会 Spirocyclic androgen receptor protein degrading agents
CA3151078A1 (en) 2019-09-25 2021-04-01 Surface Oncology, Inc. Anti-il-27 antibodies and uses thereof
JP7280387B2 (en) 2019-09-27 2023-05-23 グラクソスミスクライン、インテレクチュアル、プロパティー、ディベロップメント、リミテッド antigen binding protein
EP4037714A1 (en) 2019-10-03 2022-08-10 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for modulating macrophages polarization
CA3157889A1 (en) 2019-10-17 2021-04-22 Inserm (Institut National De La Sante Et De La Recherche Medicale) Methods for diagnosing nasal intestinal type adenocarcinomas
BR112022007376A2 (en) 2019-10-21 2022-07-05 Novartis Ag COMBINATION THERAPIES WITH VENETOCLAX AND TIM-3 INHIBITORS
EP4048285A1 (en) 2019-10-21 2022-08-31 Novartis AG Tim-3 inhibitors and uses thereof
EP4048304A1 (en) 2019-10-22 2022-08-31 Institut Curie Immunotherapy targeting tumor neoantigenic peptides
CA3155202A1 (en) 2019-10-23 2021-04-29 Arthur M. Krieg Synthetic rig-i-like receptor agonists
CN112724127B (en) 2019-10-28 2023-02-17 中国科学院上海药物研究所 Five-membered heterocyclic oxocarboxylic acid compound and medical application thereof
KR20220092540A (en) 2019-10-29 2022-07-01 에자이 알앤드디 매니지먼트 가부시키가이샤 Combination of a PD-1 antagonist, a VEGFR/FGFR/RET tyrosine kinase inhibitor and a CBP/beta-catenin inhibitor for treating cancer
EP4051286A1 (en) 2019-10-29 2022-09-07 Institut National de la Santé et de la Recherche Médicale (INSERM) Methods and compositions for treating uveal melanoma
IL291748A (en) 2019-11-07 2022-06-01 Oncxerna Therapeutics Inc Classification of tumor microenvironments
EP3824954A1 (en) 2019-11-22 2021-05-26 Centre National de la Recherche Scientifique Device, apparatus and method for minibeam radiation therapy
EP4061809A1 (en) 2019-11-22 2022-09-28 Theravance Biopharma R&D IP, LLC Substituted 1,5-naphthyridines or quinolines as alk5 inhibitors
JP2023502264A (en) 2019-11-22 2023-01-23 スミトモ ファーマ オンコロジー, インコーポレイテッド Solid dose pharmaceutical composition
US20210154281A1 (en) 2019-11-26 2021-05-27 Massachusetts Institute Of Technology Cell-based cancer vaccines and cancer therapies
EP4069683A1 (en) 2019-12-06 2022-10-12 Mersana Therapeutics, Inc. Dimeric compounds as sting agonists
US11897950B2 (en) 2019-12-06 2024-02-13 Augusta University Research Institute, Inc. Osteopontin monoclonal antibodies
US20230346901A1 (en) 2019-12-19 2023-11-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and vaccine compositions to treat cancers
IL293834A (en) 2019-12-20 2022-08-01 Novartis Ag Combination of anti tim-3 antibody mbg453 and anti tgf-beta antibody nis793, with or without decitabine or the anti pd-1 antibody spartalizumab, for treating myelofibrosis and myelodysplastic syndrome
KR20220128389A (en) 2020-01-17 2022-09-20 노파르티스 아게 A combination comprising a TIM-3 inhibitor and a hypomethylating agent for use in treating myelodysplastic syndrome or chronic myelomonocytic leukemia
US20230076415A1 (en) 2020-01-17 2023-03-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
US20230090446A1 (en) 2020-01-28 2023-03-23 Universite De Strasbourg Antisense oligonucleotide targeting linc00518 for treating melanoma
BR112022014562A2 (en) 2020-01-28 2022-09-13 Glaxosmithkline Ip Dev Ltd COMBINATION TREATMENTS, USES AND METHODS THEREOF
JP2023513896A (en) 2020-02-04 2023-04-04 ジェンマブ エー/エス Antibodies for use in therapy
WO2021156360A1 (en) 2020-02-05 2021-08-12 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for discontinuing a treatment with a tyrosine kinase inhibitor (tki)
WO2021170777A1 (en) 2020-02-28 2021-09-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for diagnosing, prognosing and managing treatment of breast cancer
IL296103A (en) 2020-03-05 2022-11-01 Neotx Therapeutics Ltd Methods and compositions for treating cancer with immune cells
US20230159573A1 (en) 2020-03-26 2023-05-25 The Regents Of The University Of Michigan Small molecule stat protein degraders
WO2021203131A1 (en) 2020-03-31 2021-10-07 Theravance Biopharma R&D Ip, Llc Substituted pyrimidines and methods of use
JP2023521228A (en) 2020-04-14 2023-05-23 グラクソスミスクライン、インテレクチュアル、プロパティー、ディベロップメント、リミテッド Cancer combination therapy
EP4136105A1 (en) 2020-04-14 2023-02-22 GlaxoSmithKline Intellectual Property Development Limited Combination treatment for cancer based upon an icos antibody and a pd-l1 antibody tgf-beta-receptor fusion protein
CA3171597A1 (en) 2020-04-14 2021-10-21 Glaxosmithkline Intellectual Property Development Limited Combination treatment for cancer
EP4139341A1 (en) 2020-04-21 2023-03-01 Regeneron Pharmaceuticals, Inc. Il-2 variants with reduced binding to il-2 receptor alpha and uses thereof
TW202206100A (en) 2020-04-27 2022-02-16 美商西健公司 Treatment for cancer
EP4137156A1 (en) 2020-05-01 2023-02-22 The University of Tokyo Prophylactic or therapeutic agent for at least one type of cancer selected from group consisting of pancreatic cancer, lung cancer, colorectal cancer, biliary tract cancer and liver cancer, prophylactic or therapeutic agent for said cancer which is used in combination drug in combination with said agent, combination drug comprising said agents, and method for screening for prophylactic or therapeutic agent for cancer
WO2021224186A1 (en) 2020-05-04 2021-11-11 Institut Curie New pyridine derivatives as radiosensitizers
PE20230680A1 (en) 2020-05-06 2023-04-21 Merck Sharp And Dohme Llc IL4I1 INHIBITORS AND METHODS OF USE
IL298019A (en) 2020-05-13 2023-01-01 Massachusetts Inst Technology Compositions of polymeric microdevices and their use in cancer immunotherapy
AU2021273009A1 (en) 2020-05-13 2022-12-15 Bonum Therapeutics, Inc. Compositions of protein complexes and methods of use thereof
CA3182333A1 (en) 2020-05-20 2021-11-25 Institut Curie Single domain antibodies and their use in cancer therapies
EP4157464A1 (en) 2020-05-26 2023-04-05 Regeneron Pharmaceuticals, Inc. Methods of treating cervical cancer by administering the pd-1 inhibitor antibody cemiplimab
US11767353B2 (en) 2020-06-05 2023-09-26 Theraly Fibrosis, Inc. Trail compositions with reduced immunogenicity
WO2021253041A1 (en) 2020-06-10 2021-12-16 Theravance Biopharma R&D Ip, Llc Naphthyridine derivatives useful as alk5 inhibitors
AR122644A1 (en) 2020-06-19 2022-09-28 Onxeo NEW CONJUGATED NUCLEIC ACID MOLECULES AND THEIR USES
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
US20230293530A1 (en) 2020-06-24 2023-09-21 Yeda Research And Development Co. Ltd. Agents for sensitizing solid tumors to treatment
WO2021263167A2 (en) 2020-06-26 2021-12-30 Amgen Inc. Il-10 muteins and fusion proteins thereof
WO2022011205A1 (en) 2020-07-10 2022-01-13 The Regents Of The University Of Michigan Androgen receptor protein degraders
WO2022011204A1 (en) 2020-07-10 2022-01-13 The Regents Of The University Of Michigan Small molecule androgen receptor protein degraders
WO2022023379A1 (en) 2020-07-28 2022-02-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for preventing and treating a cancer
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
JP2023540217A (en) 2020-08-26 2023-09-22 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Method of treating cancer by administering a PD-1 inhibitor
CA3189987A1 (en) 2020-09-02 2022-03-10 Pharmabcine Inc. Combination therapy of a pd-1 antagonist and an antagonist for vegfr-2 for treating patients with cancer
WO2022051448A1 (en) 2020-09-03 2022-03-10 Regeneron Pharmaceuticals, Inc. Methods of treating cancer pain by administering a pd-1 inhibitor
CN111920948B (en) * 2020-09-25 2021-02-02 安可瑞(山西)生物细胞有限公司 Pharmaceutical composition comprising immune cells for treating cancer
WO2022079270A1 (en) 2020-10-16 2022-04-21 Université D'aix-Marseille Anti-gpc4 single domain antibodies
EP4232453A1 (en) 2020-10-20 2023-08-30 Institut Curie Metallic trans-(n-heterocyclic carbene)-amine-platinum complexes and uses thereof for treating cancer
WO2022084531A1 (en) 2020-10-23 2022-04-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating glioma
TW202233185A (en) 2020-10-28 2022-09-01 日商衛材R&D企管股份有限公司 Pharmaceutical composition for treating tumors
WO2022097060A1 (en) 2020-11-06 2022-05-12 Novartis Ag Cd19 binding molecules and uses thereof
WO2022101481A1 (en) 2020-11-16 2022-05-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for predicting and treating uveal melanoma
WO2022101484A1 (en) 2020-11-16 2022-05-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for predicting and treating uveal melanoma
US20230416196A1 (en) 2020-11-18 2023-12-28 Institut Curie Dimer of biguanidines and their therapeutic uses
IL303424A (en) 2020-12-07 2023-08-01 Genmab As Antibody and taxane combination therapy
TW202237119A (en) 2020-12-10 2022-10-01 美商住友製藥腫瘤公司 Alk-5 inhibitors and uses thereof
WO2022130206A1 (en) 2020-12-16 2022-06-23 Pfizer Inc. TGFβr1 INHIBITOR COMBINATION THERAPIES
JP2024505428A (en) 2021-01-14 2024-02-06 アンスティテュ キュリー HER2 single domain antibody variants and their CARs
EP4284919A1 (en) 2021-01-29 2023-12-06 Iovance Biotherapeutics, Inc. Methods of making modified tumor infiltrating lymphocytes and their use in adoptive cell therapy
EP4284510A1 (en) 2021-01-29 2023-12-06 Novartis AG Dosage regimes for anti-cd73 and anti-entpd2 antibodies and uses thereof
WO2022165403A1 (en) 2021-02-01 2022-08-04 Yale University Chemotherapeutic bioadhesive particles with immunostimulatory molecules for cancer treatment
IL305427A (en) 2021-03-02 2023-10-01 Glaxosmithkline Ip Dev Ltd Substituted pyridines as dnmt1 inhibitors
WO2022187423A1 (en) 2021-03-03 2022-09-09 The Regents Of The University Of Michigan Cereblon ligands
WO2022187419A1 (en) 2021-03-03 2022-09-09 The Regents Of The University Of Michigan Small molecule degraders of androgen receptor
WO2022189618A1 (en) 2021-03-12 2022-09-15 Institut Curie Nitrogen-containing heterocycles as radiosensitizers
EP4308118A1 (en) 2021-03-17 2024-01-24 Institut National de la Santé et de la Recherche Médicale (INSERM) Methods and compositions for treating melanoma
WO2022195551A1 (en) 2021-03-18 2022-09-22 Novartis Ag Biomarkers for cancer and methods of use thereof
AU2022242000A1 (en) 2021-03-23 2023-09-14 Regeneron Pharmaceuticals, Inc. Methods of treating cancer in immunosuppressed or immunocompromised patients by administering a pd-1 inhibitor
KR20240000473A (en) 2021-03-25 2024-01-02 온세르나 테라퓨틱스, 인크. Targeted therapy for cancer
EP4313127A1 (en) 2021-03-29 2024-02-07 Juno Therapeutics, Inc. Methods for dosing and treatment with a combination of a checkpoint inhibitor therapy and a car t cell therapy
WO2022208353A1 (en) 2021-03-31 2022-10-06 Glaxosmithkline Intellectual Property Development Limited Antigen binding proteins and combinations thereof
TW202304979A (en) 2021-04-07 2023-02-01 瑞士商諾華公司 USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
AU2022254104A1 (en) 2021-04-08 2023-10-26 Nurix Therapeutics, Inc. Combination therapies with cbl-b inhibitor compounds
US20220363696A1 (en) 2021-04-13 2022-11-17 Nuvalent, Inc. Amino-substituted heterocycles for treating cancers with egfr mutations
EP4326903A1 (en) 2021-04-23 2024-02-28 Inserm (Institut National De La Sante Et De La Recherche Medicale) Methods and compositions for treating cell senescence accumulation related disease
WO2022227015A1 (en) 2021-04-30 2022-11-03 Merck Sharp & Dohme Corp. Il4i1 inhibitors and methods of use
US20220389089A1 (en) 2021-05-07 2022-12-08 Surface Oncology, Inc. Anti-il-27 antibodies and uses thereof
EP4337763A1 (en) 2021-05-10 2024-03-20 Institut Curie Methods for the treatment of cancer, inflammatory diseases and autoimmune diseases
AR125874A1 (en) 2021-05-18 2023-08-23 Novartis Ag COMBINATION THERAPIES
EP4346887A1 (en) 2021-05-25 2024-04-10 Edelweiss Immune Inc C-x-c motif chemokine receptor 6 (cxcr6) binding molecules, and methods of using the same
WO2022251359A1 (en) 2021-05-26 2022-12-01 Theravance Biopharma R&D Ip, Llc Bicyclic inhibitors of alk5 and methods of use
WO2022256534A1 (en) 2021-06-03 2022-12-08 Synthorx, Inc. Head and neck cancer combination therapy comprising an il-2 conjugate and pembrolizumab
GB202107994D0 (en) 2021-06-04 2021-07-21 Kymab Ltd Treatment of cancer
WO2022261018A1 (en) 2021-06-07 2022-12-15 Providence Health & Services - Oregon Cxcr5, pd-1, and icos expressing tumor reactive cd4 t cells and their use
AU2022302143A1 (en) 2021-07-02 2024-01-18 Venugopalareddy BOMMIREDDYVENKATA Compositions and methods for treating cancers
WO2023004287A1 (en) 2021-07-19 2023-01-26 Regeneron Pharmaceuticals, Inc. Combination of checkpoint inhibitors and an oncolytic virus for treating cancer
WO2023010080A1 (en) 2021-07-30 2023-02-02 Seagen Inc. Treatment for cancer
CN117794953A (en) 2021-08-03 2024-03-29 豪夫迈·罗氏有限公司 Bispecific antibodies and methods of use
CA3227880A1 (en) 2021-08-05 2023-02-09 Marios SOTIROPOULOS Scanning dynamic device for minibeams production
AU2022332285A1 (en) 2021-08-23 2024-02-15 Immunitas Therapeutics, Inc. Anti-cd161 antibodies and uses thereof
WO2023026881A1 (en) 2021-08-25 2023-03-02 東亞合成株式会社 Anti-pd-1 signal peptide antibody and use thereof
WO2023034864A1 (en) 2021-08-31 2023-03-09 Yale University Compositions and methods for treating cancers
WO2023041744A1 (en) 2021-09-17 2023-03-23 Institut Curie Bet inhibitors for treating pab1 deficient cancer
WO2023057882A1 (en) 2021-10-05 2023-04-13 Pfizer Inc. Combinations of azalactam compounds with a pd-1 axis binding antagonist for the treatment of cancer
WO2023057535A1 (en) 2021-10-06 2023-04-13 Genmab A/S Multispecific binding agents against pd-l1 and cd137 in combination with anti pd-1 antibodies for treating cancers
WO2023057534A1 (en) 2021-10-06 2023-04-13 Genmab A/S Multispecific binding agents against pd-l1 and cd137 in combination
WO2023079428A1 (en) 2021-11-03 2023-05-11 Pfizer Inc. Combination therapies using tlr7/8 agonist
WO2023078900A1 (en) 2021-11-03 2023-05-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating triple negative breast cancer (tnbc)
WO2023089032A1 (en) 2021-11-19 2023-05-25 Institut Curie Methods for the treatment of hrd cancer and brca-associated cancer
WO2023099763A1 (en) 2021-12-03 2023-06-08 Institut Curie Sirt6 inhibitors for use in treating resistant hrd cancer
WO2023111203A1 (en) 2021-12-16 2023-06-22 Onxeo New conjugated nucleic acid molecules and their uses
WO2023122573A1 (en) 2021-12-20 2023-06-29 Synthorx, Inc. Head and neck cancer combination therapy comprising an il-2 conjugate and pembrolizumab
WO2023118165A1 (en) 2021-12-21 2023-06-29 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
WO2023147488A1 (en) 2022-01-28 2023-08-03 Iovance Biotherapeutics, Inc. Cytokine associated tumor infiltrating lymphocytes compositions and methods
WO2023159102A1 (en) 2022-02-17 2023-08-24 Regeneron Pharmaceuticals, Inc. Combinations of checkpoint inhibitors and oncolytic virus for treating cancer
WO2023174210A1 (en) 2022-03-14 2023-09-21 Laekna Limited Combination treatment for cancer
WO2023180552A1 (en) 2022-03-24 2023-09-28 Institut Curie Immunotherapy targeting tumor transposable element derived neoantigenic peptides in glioblastoma
WO2023187024A1 (en) 2022-03-31 2023-10-05 Institut Curie Modified rela protein for inducing interferon expression and engineered immune cells with improved interferon expression
WO2023194607A1 (en) 2022-04-07 2023-10-12 Institut Curie Myeloid cells modified by chimeric antigen receptor with cd40 and uses thereof for anti-cancer therapy
WO2023194608A1 (en) 2022-04-07 2023-10-12 Institut Curie Myeloid cells modified by chimeric antigen receptor and uses thereof for anti-cancer therapy
WO2023213764A1 (en) 2022-05-02 2023-11-09 Transgene Fusion polypeptide comprising an anti-pd-l1 sdab and a member of the tnfsf
WO2023213763A1 (en) 2022-05-02 2023-11-09 Transgene Poxvirus encoding a binding agent comprising an anti- pd-l1 sdab
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors
WO2023230554A1 (en) 2022-05-25 2023-11-30 Pfizer Inc. Combination of a braf inhibitor, an egfr inhibitor, and a pd-1 antagonist for the treatment of braf v600e-mutant, msi-h/dmmr colorectal cancer
WO2023230541A1 (en) 2022-05-27 2023-11-30 Viiv Healthcare Company Piperazine derivatives useful in hiv therapy
WO2024003353A1 (en) 2022-07-01 2024-01-04 Transgene Fusion protein comprising a surfactant-protein-d and a member of the tnfsf
WO2024033399A1 (en) 2022-08-10 2024-02-15 Institut National de la Santé et de la Recherche Médicale Sigmar1 ligand for the treatment of pancreatic cancer
WO2024033400A1 (en) 2022-08-10 2024-02-15 Institut National de la Santé et de la Recherche Médicale Sk2 inhibitor for the treatment of pancreatic cancer
WO2024040264A1 (en) 2022-08-19 2024-02-22 Massachusetts Institute Of Technology Compositions and methods for targeting dendritic cell lectins
WO2024056716A1 (en) 2022-09-14 2024-03-21 Institut National de la Santé et de la Recherche Médicale Methods and pharmaceutical compositions for the treatment of dilated cardiomyopathy

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7595048B2 (en) * 2002-07-03 2009-09-29 Ono Pharmaceutical Co., Ltd. Method for treatment of cancer by inhibiting the immunosuppressive signal induced by PD-1

Family Cites Families (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE555319A (en) 1956-03-21 1900-01-01
US3095355A (en) 1961-10-12 1963-06-25 Revlon Aerosol composition
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
JP3454275B2 (en) 1992-06-05 2003-10-06 佑 本庶 Novel polypeptide associated with programmed cell death and DNA encoding the same
US5260074A (en) 1992-06-22 1993-11-09 Digestive Care Inc. Compositions of digestive enzymes and salts of bile acids and process for preparation thereof
EP0749475A4 (en) * 1992-08-26 1997-05-07 Harvard College Use of the cytokine ip-10 as an anti-tumor agent
IL108501A (en) 1994-01-31 1998-10-30 Mor Research Applic Ltd Antibodies and pharmaceutical compositions containing them
CA2143491C (en) 1994-03-01 2011-02-22 Yasumasa Ishida A novel peptide related to human programmed cell death and dna encoding it
DE4431401A1 (en) * 1994-08-24 1996-02-29 Max Delbrueck Centrum Live vaccine against tumor diseases
US6051227A (en) * 1995-07-25 2000-04-18 The Regents Of The University Of California, Office Of Technology Transfer Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
KR100308764B1 (en) 1995-08-29 2001-12-17 마나배게이사꾸 Chimeric Animals and How to Make them
US6632976B1 (en) 1995-08-29 2003-10-14 Kirin Beer Kabushiki Kaisha Chimeric mice that are produced by microcell mediated chromosome transfer and that retain a human antibody gene
PT1520588E (en) * 1998-07-13 2015-03-31 Univ Texas Uses of antibodies to aminophospholipids for cancer treatment
DK1137436T3 (en) * 1998-12-03 2008-10-13 Univ California Stimulation of T cells against self antigens using CTLA-4 blocking agents
US7041474B2 (en) 1998-12-30 2006-05-09 Millennium Pharmaceuticals, Inc. Nucleic acid encoding human tango 509
EP1053751A1 (en) * 1999-05-17 2000-11-22 Institut National De La Sante Et De La Recherche Medicale (Inserm) Compositions and methods for treating cell proliferation disorders
FR2794025A1 (en) * 1999-05-25 2000-12-01 Transgene Sa COMPOSITION FOR IMPLEMENTING ANTI-TUMOR OR ANTIVIRAL TREATMENT IN A MAMMAL
US6936704B1 (en) 1999-08-23 2005-08-30 Dana-Farber Cancer Institute, Inc. Nucleic acids encoding costimulatory molecule B7-4
HU228477B1 (en) * 1999-08-23 2013-03-28 Dana Farber Cancer Inst Inc Pd-1, a receptor for b7-4, and uses therefor
ATE354655T1 (en) 1999-08-24 2007-03-15 Medarex Inc HUMAN ANTIBODIES TO CTLA-4 AND THEIR USES
GB9927328D0 (en) * 1999-11-18 2000-01-12 Lorantis Ltd Immunotherapy
AU784634B2 (en) 1999-11-30 2006-05-18 Mayo Foundation For Medical Education And Research B7-H1, a novel immunoregulatory molecule
US6803192B1 (en) 1999-11-30 2004-10-12 Mayo Foundation For Medical Education And Research B7-H1, a novel immunoregulatory molecule
JP3904374B2 (en) * 2000-02-29 2007-04-11 独立行政法人科学技術振興機構 Lymphocytes with enhanced killer activity
US7030219B2 (en) 2000-04-28 2006-04-18 Johns Hopkins University B7-DC, Dendritic cell co-stimulatory molecules
NZ522844A (en) * 2000-06-28 2005-02-25 Brigham & Womens Hospital PD-L2 molecules: novel PD-1 ligands and methods to identify compounds to modulate T cell activation
US7132109B1 (en) * 2000-10-20 2006-11-07 University Of Connecticut Health Center Using heat shock proteins to increase immune response
CA2426366A1 (en) * 2000-10-20 2002-04-25 Tsuneya Ohno Fusion cells and cytokine compositions for treatment of disease
JP2002194491A (en) 2000-12-27 2002-07-10 Daido Steel Co Ltd Steel for spring
NZ528265A (en) 2001-04-02 2005-10-28 Wyeth Corp Screening of compounds which modulate PD-1 signalling by testing for compounds that modulate phosphorylation of SHP-2, ERK1 or ERK-2, and PKC-theta
AR036993A1 (en) * 2001-04-02 2004-10-20 Wyeth Corp USE OF AGENTS THAT MODULATE THE INTERACTION BETWEEN PD-1 AND ITS LINKS IN THE SUBMODULATION OF IMMUNOLOGICAL ANSWERS
AU2002258941A1 (en) * 2001-04-20 2002-11-05 Mayo Foundation For Medical Education And Research Methods of enhancing cell responsiveness
JP2003029846A (en) 2001-07-11 2003-01-31 Sanyo Electric Co Ltd Flow rate adjusting unit and beverage supplying device equipped with flow rate adjusting unit
WO2003006636A1 (en) 2001-07-12 2003-01-23 Genethor Gmbh Reduction of the stimulatory capacity of antigen-presenting cells
EP1445264B1 (en) 2001-07-31 2011-09-14 Ono Pharmaceutical Co., Ltd. Substance specific to pd-1
IL145926A0 (en) 2001-10-15 2002-07-25 Mor Research Applic Ltd Peptide epitopes of mimotopes useful in immunomodulation
WO2003042402A2 (en) * 2001-11-13 2003-05-22 Dana-Farber Cancer Institute, Inc. Agents that modulate immune cell activation and methods of use thereof
IL149820A0 (en) 2002-05-23 2002-11-10 Curetech Ltd Humanized immunomodulatory monoclonal antibodies for the treatment of neoplastic disease or immunodeficiency
CA2489803A1 (en) 2002-06-20 2003-12-31 The Regents Of The University Of California Compositions and methods for modulating lymphocyte activity
CN101899114A (en) * 2002-12-23 2010-12-01 惠氏公司 Anti-PD-1 antibody and uses thereof
JP4532409B2 (en) * 2003-01-23 2010-08-25 小野薬品工業株式会社 Substance with specificity for human PD-1
US7465446B2 (en) 2003-05-30 2008-12-16 Medarex, Inc. Surrogate therapeutic endpoint for anti-CTLA4-based immunotherapy of disease
EP1591627A1 (en) 2004-04-27 2005-11-02 Siemens Aktiengesellschaft Controlling arrangement for a compressor and use of a Bragg grating sensor in a controlling arrangement
US20090123413A1 (en) 2004-08-23 2009-05-14 Britta Hardy Use of bat monoclonal antibody for immunotherapy
US7423128B2 (en) 2004-11-03 2008-09-09 Amgen Fremont Inc. Anti-properdin antibodies, and methods for making and using same
CA2607147C (en) 2005-05-09 2018-07-17 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1 (pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
WO2006124269A2 (en) 2005-05-16 2006-11-23 Amgen Fremont Inc. Human monoclonal antibodies that bind to very late antigen-1 for the treatment of inflammation and other disorders
AU2006265108C1 (en) 2005-07-01 2013-01-17 E. R. Squibb & Sons, L.L.C. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
SG10201700698WA (en) 2012-05-15 2017-02-27 Bristol Myers Squibb Co Cancer immunotherapy by disrupting pd-1/pd-l1 signaling

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7595048B2 (en) * 2002-07-03 2009-09-29 Ono Pharmaceutical Co., Ltd. Method for treatment of cancer by inhibiting the immunosuppressive signal induced by PD-1
US8728474B2 (en) * 2002-07-03 2014-05-20 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
US20140314714A1 (en) * 2002-07-03 2014-10-23 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
US20150093380A1 (en) * 2002-07-03 2015-04-02 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9402899B2 (en) 2002-07-03 2016-08-02 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
US9439962B2 (en) 2002-07-03 2016-09-13 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
US9393301B2 (en) 2002-07-03 2016-07-19 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
US9358289B2 (en) 2005-05-09 2016-06-07 Ono Pharmaceutical Co., Ltd. Methods for treating cancer using anti-PD-1 antibodies in combination with anti-CTLA-4 antibodies
US9387247B2 (en) 2005-05-09 2016-07-12 Ono Pharmaceutical Co., Ltd. Monoclonal antibodies to programmed death 1 (PD-1)
US9492539B2 (en) 2005-05-09 2016-11-15 Ono Pharmaceutical Co., Ltd. Monoclonal antibodies to Programmed Death 1 (PD-1)
US9492540B2 (en) 2005-05-09 2016-11-15 Ono Pharmaceutical Co., Ltd. Methods for treating cancer using anti-PD-1 antibodies
US10441655B2 (en) 2005-05-09 2019-10-15 Ono Pharmaceutical Co., Ltd. Monoclonal antibodies to programmed death 1 (PD-1)
US11236164B2 (en) 2008-08-11 2022-02-01 E.R. Squibb & Sons, L.L.C. Human antibodies that bind lymphocyte activation gene-3 (LAG-3), and uses thereof
US11236165B2 (en) 2008-08-11 2022-02-01 E.R. Squibb & Sons, L.L.C. Human antibodies that bind Lymphocyte Activation Gene-3 (LAG-3), and uses thereof
US10988536B2 (en) 2008-08-11 2021-04-27 E.R. Squibb & Sons, L.L.C. Human antibodies that bind lymphocyte activation gene-3 (LAG-3), and uses thereof
US10344089B2 (en) 2008-08-11 2019-07-09 E.R. Squibb & Sons, L.L.C. Human antibodies that bind lymphocyte activation gene-3 (LAG-3), and uses thereof
US11236163B2 (en) 2008-08-11 2022-02-01 E.R. Squibb & Sons, L.L.C. Human antibodies that bind lymphocyte activation gene-3 (LAG-3), and uses thereof
US11512130B2 (en) 2008-08-11 2022-11-29 E.R. Squibb & Sons, L.L.C. Human antibodies that bind lymphocyte activation gene-3 (LAG-3), and uses thereof
US11530267B2 (en) 2008-08-11 2022-12-20 E.R. Squibb & Sons, L.L.C. Human antibodies that bind lymphocyte activation gene-3 (LAG-3), and uses thereof
US11001630B2 (en) 2008-08-11 2021-05-11 E.R. Squibb & Sons, L.L.C. Human antibodies that bind lymphocyte activation Gene-3 (LAG-3), and uses thereof
US10988535B2 (en) 2008-08-11 2021-04-27 E.R. Squibb & Sons, L.L.C. Human antibodies that bind lymphocyte activation gene-3 (LAG-3), and uses thereof
US10266591B2 (en) 2012-07-02 2019-04-23 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (LAG-3), and uses thereof
US11345752B2 (en) 2012-07-02 2022-05-31 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (LAG-3), and uses thereof
US10377824B2 (en) 2012-07-02 2019-08-13 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (LAG-3), and uses thereof
US11274152B2 (en) 2013-09-20 2022-03-15 Bristol-Myers Squibb Company Combination of anti-LAG-3 antibodies and anti-PD-1 antibodies to treat tumors
US10081681B2 (en) 2013-09-20 2018-09-25 Bristol-Myers Squibb Company Combination of anti-LAG-3 antibodies and anti-PD-1 antibodies to treat tumors
US9907849B2 (en) 2014-07-18 2018-03-06 Advaxis, Inc. Combination of a PD-1 antagonist and a listeria-based vaccine for treating prostate cancer
US10668152B2 (en) 2015-12-17 2020-06-02 Bristol-Myers Squibb Company Use of anti-PD-1 antibody in combination with anti-CD27 antibody in cancer treatment
US10392442B2 (en) 2015-12-17 2019-08-27 Bristol-Myers Squibb Company Use of anti-PD-1 antibody in combination with anti-CD27 antibody in cancer treatment
US11083790B2 (en) 2016-06-02 2021-08-10 Bristol-Myers Squibb Company Treatment of Hodgkin lymphoma using an anti-PD-1 antibody
US11299543B2 (en) 2016-06-02 2022-04-12 Bristol-Myers Squibb Company Use of an anti-PD-1 antibody in combination with an anti-CD30 antibody in cancer treatment
US11332529B2 (en) 2016-06-03 2022-05-17 Bristol-Myers Squibb Company Methods of treating colorectal cancer
US11767361B2 (en) 2016-06-03 2023-09-26 Bristol-Myers Squibb Company Method of treating lung cancer
WO2018039131A1 (en) * 2016-08-22 2018-03-01 Protiva Biotherapeutics, Inc. Anti-pd-1 antibodies, or fragments thereof, for treating hepatitis b
US11723975B2 (en) 2017-05-30 2023-08-15 Bristol-Myers Squibb Company Compositions comprising an anti-LAG-3 antibody or an anti-LAG-3 antibody and an anti-PD-1 or anti-PD-L1 antibody
US11807686B2 (en) 2017-05-30 2023-11-07 Bristol-Myers Squibb Company Treatment of LAG-3 positive tumors

Also Published As

Publication number Publication date
US20090297518A1 (en) 2009-12-03
FR15C0088I1 (en) 2016-02-12
ES2654064T3 (en) 2024-03-13
US9393301B2 (en) 2016-07-19
US20200062846A1 (en) 2020-02-27
JP2021080301A (en) 2021-05-27
HUS1600004I1 (en) 2016-02-29
JP2018039835A (en) 2018-03-15
US20150093380A1 (en) 2015-04-02
JP2009286795A (en) 2009-12-10
JP2014065748A (en) 2014-04-17
PT1537878E (en) 2010-11-18
JP6035372B2 (en) 2016-11-30
US20160158356A1 (en) 2016-06-09
AU2003281200A1 (en) 2004-01-23
EP2243493A1 (en) 2010-10-27
JP2015163631A (en) 2015-09-10
EP3287144A1 (en) 2018-02-28
US9402899B2 (en) 2016-08-02
JP2019094340A (en) 2019-06-20
US20160158355A1 (en) 2016-06-09
DK1537878T3 (en) 2011-01-24
JP5159730B2 (en) 2013-03-13
EP1537878A1 (en) 2005-06-08
JP2012236861A (en) 2012-12-06
DE10161767T1 (en) 2018-06-07
LU92940I2 (en) 2016-03-07
US20110081341A1 (en) 2011-04-07
US20140314714A1 (en) 2014-10-23
JP6559207B2 (en) 2019-08-14
US20060110383A1 (en) 2006-05-25
HUE10161767T1 (en) 2019-03-28
EP1537878A4 (en) 2006-11-29
US8728474B2 (en) 2014-05-20
US9439962B2 (en) 2016-09-13
JPWO2004004771A1 (en) 2005-11-04
FI2206517T3 (en) 2023-10-19
EP2206517A1 (en) 2010-07-14
LU92905I2 (en) 2016-02-10
US9073994B2 (en) 2015-07-07
DK2206517T3 (en) 2023-11-06
SI2206517T1 (en) 2023-12-29
JP6258428B2 (en) 2018-01-10
ATE481985T1 (en) 2010-10-15
US8168179B2 (en) 2012-05-01
US9067999B1 (en) 2015-06-30
JP5701266B2 (en) 2015-04-15
EP2206517B1 (en) 2023-08-02
DK2206517T1 (en) 2018-02-05
ES2350687T3 (en) 2011-01-26
ES2654064T1 (en) 2018-02-12
WO2004004771A1 (en) 2004-01-15
HUS1500066I1 (en) 2016-02-29
FR16C0001I1 (en) 2016-02-12
US7595048B2 (en) 2009-09-29
DE60334303D1 (en) 2010-11-04
PT2206517T (en) 2023-11-07
US20170051060A1 (en) 2017-02-23
JP4409430B2 (en) 2010-02-03
JP5885764B2 (en) 2016-03-15
JP2016210817A (en) 2016-12-15
EP1537878B1 (en) 2010-09-22

Similar Documents

Publication Publication Date Title
US20200062846A1 (en) Immunopotentiative composition
JP5726170B2 (en) Anti-CEACAM1 antibody and method of use thereof
US9828435B2 (en) Use of antibodies or antigen-binding fragments thereof that specifically bind semaphorin-4D to increase tumor infiltrating leukocyte frequency
JP2022511337A (en) Methods and Pharmaceutical Compositions for the Treatment of Cancers Resistant to Immune Checkpoint Treatment
WO2022083590A1 (en) Chimeric receptor containing dap 12 and co-stimulatory signal molecule signal domain, and method for using same

Legal Events

Date Code Title Description
AS Assignment

Owner name: ONO PHARMACEUTICAL CO., LTD., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HONJO, TASUKU;MINATO, NAGAHIRO;IWAI, YOSHIKO;AND OTHERS;SIGNING DATES FROM 20041222 TO 20041224;REEL/FRAME:035588/0973

Owner name: TASUKU HONJO, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HONJO, TASUKU;MINATO, NAGAHIRO;IWAI, YOSHIKO;AND OTHERS;SIGNING DATES FROM 20041222 TO 20041224;REEL/FRAME:035588/0973

STCF Information on status: patent grant

Free format text: PATENTED CASE

IPR Aia trial proceeding filed before the patent and appeal board: inter partes review

Free format text: TRIAL NO: IPR2016-01217

Opponent name: MERCK SHARP DOHME CORP. ANDMERCK CO., INC

Effective date: 20160629

Free format text: TRIAL NO: IPR2016-01218

Opponent name: MERCK SHARP DOHME CORP. ANDMERCK CO., INC

Effective date: 20160629

CC Certificate of correction
MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4

AS Assignment

Owner name: DANA-FARBER CANCER INSTITUTE, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:FREEMAN, GORDON J.;REEL/FRAME:050035/0010

Effective date: 20190806

CC Certificate of correction
MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8