US20210163589A1 - Antibodies Directed Against Programmed Death-1(PD-1) - Google Patents

Antibodies Directed Against Programmed Death-1(PD-1) Download PDF

Info

Publication number
US20210163589A1
US20210163589A1 US16/927,339 US202016927339A US2021163589A1 US 20210163589 A1 US20210163589 A1 US 20210163589A1 US 202016927339 A US202016927339 A US 202016927339A US 2021163589 A1 US2021163589 A1 US 2021163589A1
Authority
US
United States
Prior art keywords
seq
amino acid
acid sequence
residue
replaced
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/927,339
Inventor
David J. King
Marilyn Kehry
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.)
Anaptysbio Inc
Original Assignee
Anaptysbio Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Anaptysbio Inc filed Critical Anaptysbio Inc
Priority to US16/927,339 priority Critical patent/US20210163589A1/en
Assigned to ANAPTYSBIO, INC. reassignment ANAPTYSBIO, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KEHRY, MARILYN, KING, DAVID J.
Publication of US20210163589A1 publication Critical patent/US20210163589A1/en
Priority to US18/334,665 priority patent/US20230331843A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P31/12Antivirals
    • 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
    • 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
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • 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

  • PD-1 (also known as Programmed Cell Death 1) is a type I transmembrane protein of 268 amino acids originally identified by subtractive hybridization of a mouse T cell line undergoing apoptosis (Ishida et al., Embo J., 11: 3887-95 (1992)).
  • PD-1 is a member of the CD28/CTLA-4 family of T-cell regulators, and is expressed on activated T-cells, B-cells, and myeloid lineage cells (Greenwald et al., Annu. Rev. Immunol., 23: 515-548 (2005); and Sharpe et al., Nat. Immunol., 8: 239-245 (2007)).
  • PD-L1 PD ligand 1
  • PD-L2 PD ligand 2
  • B7 protein superfamily Greenwald et al., supra.
  • PD-L1 is expressed in a variety of cell types, including cells of the lung, heart, thymus, spleen, and kidney (see, e.g., Freeman et al., J. Exp. Med., 192(7): 1027-1034 (2000); and Yamazaki et al., J. Immunol., 169(10): 5538-5545 (2002)).
  • PD-L1 expression is upregulated on macrophages and dendritic cells (DCs) in response to lipopolysaccharide (LPS) and GM-CSF treatment, and on T-cells and B-cells upon signaling via T-cell and B-cell receptors.
  • DCs macrophages and dendritic cells
  • LPS lipopolysaccharide
  • GM-CSF GM-CSF
  • T-cells and B-cells upon signaling via T-cell and B-cell receptors.
  • PD-L1 also is expressed in a variety of murine tumor cell lines (see, e.g., Iwai et al., Proc. Natl. Acad. Sci. USA, 99(19): 12293-12297 (2002), and Blank et al., Cancer Res., 64(3): 1140-1145 (2004)).
  • PD-L2 exhibits a more restricted expression pattern and is expressed primarily by antigen presenting cells (e.g., dendritic cells and macrophages), and some tumor cell lines (see, e.g., Latchman et al., Nat. Immunol., 2(3): 261-238 (2001)).
  • PD-1 negatively regulates T-cell activation, and this inhibitory function is linked to an immunoreceptor tyrosine-based switch motif (ITSM) in the cytoplasmic domain (see, e.g., Greenwald et al., supra, and Parry et al., Mol. Cell. Biol., 25: 9543-9553 (2005)).
  • IRSM immunoreceptor tyrosine-based switch motif
  • PD-1 deficiency can lead to autoimmunity.
  • C57BL/6 PD-1 knockout mice have been shown to develop a lupus-like syndrome (see, e.g., Nishimura et al., Immunity, 11: 141-1151 (1999)).
  • a single nucleotide polymorphism in the PD-1 gene is associated with higher incidences of systemic lupus erythematosus, type 1 diabetes, rheumatoid arthritis, and progression of multiple sclerosis (see, e.g., Nielsen et al., Tissue Antigens, 62(6): 492-497 (2003); Bertsias et al., Arthritis Rheum., 60(1): 207-218 (2009); Ni et al., Hum. Genet., 121(2): 223-232 (2007); Tahoori et al., Clin. Exp. Rheumatol., 29(5): 763-767 (2011); and Kroner et al., Ann.
  • Abnormal PD-1 expression also has been implicated in T-cell dysfunctions in several pathologies, such as tumor immune evasion and chronic viral infections (see, e.g., Barber et al., Nature, 439: 682-687 (2006); and Sharpe et al., supra).
  • T-cell suppression induced by PD-1 also plays a role in the suppression of anti-tumor immunity.
  • PD-L1 is expressed on a variety of human and mouse tumors, and binding of PD-1 to PD-L1 on tumors results in T-cell suppression and tumor immune evasion and protection (Dong et al., Nat. Med., 8: 793-800 (2002)).
  • Expression of PD-L1 by tumor cells has been directly associated with their resistance to lysis by anti-tumor T-cells in vitro (Dong et al., supra; and Blank et al., Cancer Res., 64: 1140-1145 (2004)).
  • PD-1 knockout mice are resistant to tumor challenge (Iwai et al., Int.
  • T-cells from PD-1 knockout mice are highly effective in tumor rejection when adoptively transferred to tumor-bearing mice (Blank et al., supra).
  • Blocking PD-1 inhibitory signals using a monoclonal antibody can potentiate host anti-tumor immunity in mice (Iwai et al., supra; and Hirano et al., Cancer Res., 65: 1089-1096 (2005)), and high levels of PD-L1 expression in tumors are associated with poor prognosis for many human cancer types (Hamanishi et al., Proc. Natl. Acad. Sci. USA, 104: 3360-335 (2007), Brown et al., J. Immunol., 170: 1257-1266 (2003); and Flies et al., Yale Journal of Biology and Medicine, 84(4): 409-421 (2011)).
  • nivolumab also known as BMS-936558
  • MK-3575 is a humanized monoclonal antibody directed against PD-1 that has shown evidence of antitumor activity in Phase I clinical trials (see, e.g., Patnaik et al., 2012 American Society of Clinical Oncology ( ASCO ) Annual Meeting , Abstract #2512).
  • a PD-1-binding agent e.g., an antibody
  • the invention provides such PD-1-binding agents.
  • the invention provides an isolated immunoglobulin heavy chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, wherein optionally (a)residue 9 of SEQ ID NO: 1 is replaced with a different amino acid residue, (b) one or more of residues 7, 8, and 9 of SEQ ID NO: 2 is replaced with a different amino acid residue, (c) one or more of residues 1, 2, and 5 of SEQ ID NO: 3 is replaced with a different amino acid residue, or (d) any combination of (a)-(c).
  • CDR complementarity determining region 1
  • the invention provides an isolated immunoglobulin heavy chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, wherein optionally (a) residue 9 of SEQ ID NO: 12 is replaced with a different amino acid residue, (b) residue 8 and/or residue 9 of SEQ ID NO: 13 is replaced with a different amino acid residue, (c) residue 5 of SEQ ID NO: 14 is replaced with a different amino acid residue, or (d) any combination of (a)-(c).
  • CDR complementarity determining region 1
  • the invention provides an isolated immunoglobulin heavy chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 19, a CDR2 amino acid sequence of SEQ ID NO: 20, and a CDR3 amino acid sequence of SEQ ID NO: 21.
  • CDR complementarity determining region 1
  • the invention also provides an isolated immunoglobulin heavy chain polypeptide which comprises an amino acid sequence that is at least 90% identical to any one of SEQ ID NOs: 4-11, SEQ ID NOs: 15-18, and SEQ ID NOs: 22-25.
  • the invention provides an isolated immunoglobulin light chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 26 and a CDR2 amino acid sequence of SEQ ID NO: 27.
  • CDR complementarity determining region 1
  • the invention provides an isolated immunoglobulin light chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31, wherein optionally residue 12 of SEQ ID NO: 30 is replaced with a different amino acid residue.
  • CDR complementarity determining region 1
  • the invention provides an isolated immunoglobulin light chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37, wherein optionally (a) residue 5 of SEQ ID NO: 36 is replaced with a different amino acid residue, and/or (b) residue 4 of SEQ ID NO: 37 is replaced with a different amino acid residue.
  • CDR complementarity determining region 1
  • the invention provides an isolated immunoglobulin light chain polypeptide which comprises an amino acid sequence that is at least 90% identical to SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, or SEQ ID NO: 41.
  • the invention provides isolated or purified nucleic acid sequences encoding the foregoing immunoglobulin polypeptides, vectors comprising such nucleic acid sequences, isolated PD-1-binding agents comprising the foregoing immunoglobulin polypeptides, nucleic acid sequences encoding such PD-1-binding agents, vectors comprising such nucleic acid sequences, isolated cells comprising such vectors, compositions comprising such PD-1-binding agents or such vectors with a pharmaceutically acceptable carrier, and methods of treating cancer or infectious diseases in mammals by administering effective amounts of such compositions to mammals.
  • FIG. 1 is a diagram which schematically depicts different PD-1 antigen constructs utilized to generate anti-PD-1 monoclonal antibodies as described in Example 1.
  • FIG. 2 is a graph which illustrates experimental results demonstrating increased activity of an anti-TIM-3 antagonist antibody in a human CD4+ T-cell MLR assay in the presence of low levels of anti-PD-1 antibody APE2058.
  • FIG. 3 is a graph which illustrates experimental results demonstrating increased activity of an anti-LAG-3 antagonist antibody in a human CD4+ T-cell MLR assay in the presence of low levels of anti-PD-1 APE2058.
  • the invention provides an isolated immunoglobulin heavy chain polypeptide and/or an isolated immunoglobulin light chain polypeptide, or a fragment (e.g., antigen-binding fragment) thereof.
  • immunoglobulin or “antibody,” as used herein, refers to a protein that is found in blood or other bodily fluids of vertebrates, which is used by the immune system to identify and neutralize foreign objects, such as bacteria and viruses.
  • the polypeptide is “isolated” in that it is removed from its natural environment.
  • an immunoglobulin or antibody is a protein that comprises at least one complementarity determining region (CDR).
  • the CDRs form the “hypervariable region” of an antibody, which is responsible for antigen binding (discussed further below).
  • a whole immunoglobulin typically consists of four polypeptides: two identical copies of a heavy (H) chain polypeptide and two identical copies of a light (L) chain polypeptide.
  • Each of the heavy chains contains one N-terminal variable (V H ) region and three C-terminal constant (C H 1, C H 2, and C H 3) regions, and each light chain contains one N-terminal variable (V L ) region and one C-terminal constant (C L ) region.
  • the light chains of antibodies can be assigned to one of two distinct types, either kappa ( ⁇ ) or lambda ( ⁇ ), based upon the amino acid sequences of their constant domains.
  • each light chain is linked to a heavy chain by disulphide bonds, and the two heavy chains are linked to each other by disulphide bonds.
  • the light chain variable region is aligned with the variable region of the heavy chain, and the light chain constant region is aligned with the first constant region of the heavy chain.
  • the remaining constant regions of the heavy chains are aligned with each other.
  • variable regions of each pair of light and heavy chains form the antigen binding site of an antibody.
  • the V H and V L regions have the same general structure, with each region comprising four framework (FW or FR) regions.
  • framework region refers to the relatively conserved amino acid sequences within the variable region which are located between the hypervariable or complementary determining regions (CDRs).
  • CDRs hypervariable or complementary determining regions
  • the framework regions form the ⁇ sheets that provide the structural framework of the variable region (see, e.g., C. A. Janeway et al. (eds.), Immunobiology, 5 th Ed ., Garland Publishing, New York, N.Y. (2001)).
  • the framework regions are connected by three complementarity determining regions (CDRs).
  • CDRs complementarity determining regions
  • the three CDRs known as CDR1, CDR2, and CDR3, form the “hypervariable region” of an antibody, which is responsible for antigen binding.
  • the CDRs form loops connecting, and in some cases comprising part of, the beta-sheet structure formed by the framework regions.
  • the constant regions of the light and heavy chains are not directly involved in binding of the antibody to an antigen, the constant regions can influence the orientation of the variable regions.
  • the constant regions also exhibit various effector functions, such as participation in antibody-dependent complement-mediated lysis or antibody-dependent cellular toxicity via interactions with effector molecules and cells.
  • the isolated immunoglobulin heavy chain polypeptide and the isolated immunoglobulin light chain polypeptide of the invention desirably bind to PD-1.
  • programmed death 1 (PD-1) (also known as programmed cell death 1) is a 268 amino acid type I transmembrane protein (Ishida et al., supra).
  • PD-1 is a member of the CD28/CTLA-4 family of T-cell regulators and is expressed on activated T-cells, B-cells, and myeloid lineage cells (Greenwald et al., supra; and Sharpe et al., supra).
  • PD-1 includes an extracellular IgV domain followed by short extracellular stalk, a transmembrane region and an intracellular tail.
  • the intracellular tail contains two phosphorylation sites located in an immunoreceptor tyrosine-based inhibitory motif and an immunoreceptor tyrosine-based switch motif, which play a role in the ability of PD-1 to negatively regulate T-cell receptor signaling (see, e.g., Ishida et al., supra; and Blank et al., supra).
  • the inventive isolated immunoglobulin heavy chain polypeptide and the inventive isolated immunoglobulin light chain polypeptide can form an agent that binds to PD-1 and another antigen, resulting in a “dual reactive” binding agent (e.g., a dual reactive antibody).
  • the agent can bind to PD-1 and to another negative regulator of the immune system such as, for example, lymphocyte-activation gene 3 (LAG-3) and/or T-cell immunoglobulin domain and mucin domain 3 protein (TIM-3).
  • LAG-3 lymphocyte-activation gene 3
  • TIM-3 T-cell immunoglobulin domain and mucin domain
  • Antibodies which bind to PD-1, and components thereof, are known in the art (see, e.g., U.S. Pat. No. 8,168,757; Topalian et al., supra; and Patnaik et al., supra). Anti-PD-1 antibodies also are commercially available from sources such as, for example, Abcam (Cambridge, Mass.).
  • substitution refers to the replacement of one amino acid at a given position or residue by another amino acid at the same position or residue within a polypeptide sequence.
  • Amino acids are broadly grouped as “aromatic” or “aliphatic.”
  • An aromatic amino acid includes an aromatic ring.
  • aromatic amino acids include histidine (H or His), phenylalanine (F or Phe), tyrosine (Y or Tyr), and tryptophan (W or Trp).
  • Non-aromatic amino acids are broadly grouped as “aliphatic.”
  • “aliphatic” amino acids include glycine (G or Gly), alanine (A or Ala), valine (V or Val), leucine (L or Leu), isoleucine (I or Ile), methionine (M or Met), serine (S or Ser), threonine (T or Thr), cysteine (C or Cys), proline (P or Pro), glutamic acid (E or Glu), aspartic acid (A or Asp), asparagine (N or Asn), glutamine (Q or Gln), lysine (K or Lys), and arginine (R or Arg).
  • Aliphatic amino acids may be sub-divided into four sub-groups.
  • the “large aliphatic non-polar sub-group” consists of valine, leucine, and isoleucine.
  • the “aliphatic, slightly-polar sub-group” consists of methionine, serine, threonine, and cysteine.
  • the “aliphatic polar/charged sub-group” consists of glutamic acid, aspartic acid, asparagine, glutamine, lysine, and arginine.
  • the “small-residue sub-group” consists of glycine and alanine.
  • the group of charged/polar amino acids may be sub-divided into three sub-groups: the “positively-charged sub-group” consisting of lysine and arginine, the “negatively-charged sub-group” consisting of glutamic acid and aspartic acid, and the “polar sub-group” consisting of asparagine and glutamine.
  • Aromatic amino acids may be sub-divided into two sub-groups: the “nitrogen ring sub-group” consisting of histidine and tryptophan and the “phenyl sub-group” consisting of phenylalanine and tyrosine.
  • the amino acid replacement or substitution can be conservative, semi-conservative, or non-conservative.
  • the phrase “conservative amino acid substitution” or “conservative mutation” refers to the replacement of one amino acid by another amino acid with a common property.
  • a functional way to define common properties between individual amino acids is to analyze the normalized frequencies of amino acid changes between corresponding proteins of homologous organisms (Schulz and Schirmer, Principles of Protein Structure , Springer-Verlag, New York (1979)). According to such analyses, groups of amino acids may be defined where amino acids within a group exchange preferentially with each other, and therefore resemble each other most in their impact on the overall protein structure (Schulz and Schirmer, supra).
  • conservative amino acid substitutions include substitutions of amino acids within the sub-groups described above, for example, lysine for arginine and vice versa such that a positive charge may be maintained, glutamic acid for aspartic acid and vice versa such that a negative charge may be maintained, serine for threonine such that a free —OH can be maintained, and glutamine for asparagine such that a free —NH 2 can be maintained.
  • “Semi-conservative mutations” include amino acid substitutions of amino acids within the same groups listed above, but not within the same sub-group. For example, the substitution of aspartic acid for asparagine, or asparagine for lysine, involves amino acids within the same group, but different sub-groups. “Non-conservative mutations” involve amino acid substitutions between different groups, for example, lysine for tryptophan, or phenylalanine for serine, etc.
  • the invention provides an immunoglobulin heavy chain polypeptide that comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3.
  • CDR complementarity determining region 1
  • the isolated immunoglobulin heavy chain polypeptide comprises, consists of, or consists essentially of a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, wherein optionally (a) residue 9 of SEQ ID NO: 1 is replaced with a different amino acid residue, (b) one or more of residues 7, 8, and 9 of SEQ ID NO: 2 is replaced with a different amino acid residue, (c) one or more of residues 1, 2, and 5 of SEQ ID NO: 3 is replaced with a different amino acid residue, or (d) any combination of (a)-(c).
  • CDR complementarity determining region 1
  • inventive immunoglobulin heavy chain polypeptide consists essentially of a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3 and optional amino acid replacements
  • additional components can be included in the polypeptide that do not materially affect the polypeptide (e.g., protein moieties such as biotin that facilitate purification or isolation).
  • the inventive immunoglobulin heavy chain polypeptide consists of a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3 and optional amino acid replacements
  • the polypeptide does not comprise any additional components (i.e., components that are not endogenous to the inventive immunoglobulin heavy chain polypeptide).
  • the isolated immunoglobulin polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that (a) residue 9 of SEQ ID NO: 1 is replaced with a different amino acid residue, (b) one or more of residues 7, 8, and 9 of SEQ ID NO: 2 is replaced with a different amino acid residue, (c) one or more of residues 1, 2, and 5 of SEQ ID NO: 3 is replaced with a different amino acid residue, or (d) any combination of (a)-(c).
  • the isolated immunoglobulin heavy chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that residue 9 of SEQ ID NO: 1 is replaced with a different amino acid residue and one or more of residues 7, 8, and 9 of SEQ ID NO: 2 is replaced with a different amino acid residue.
  • the isolated immunoglobulin heavy chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that residue 9 of SEQ ID NO: 1 is replaced with a different amino acid residue, one or more of residues 7, 8, and 9 of SEQ ID NO: 2 is replaced with a different amino acid residue, and one or more of residues 1, 2, and 5 of SEQ ID NO: 3 is replaced with a different amino acid residue.
  • the isolated immunoglobulin heavy chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that one or more of residues 1, 2, and 5 of SEQ ID NO: 3 is replaced with a different amino acid residue.
  • Each of residue 9 of SEQ ID NO: 1, residues 7, 8, and 9 of SEQ ID NO: 2, and residues 1, 2, and 5 of SEQ ID NO: 3 can be replaced with any suitable amino acid residue that can be the same or different in each position.
  • the amino acid residue of a first position can be replaced with a first different amino acid residue
  • the amino acid residue of a second position can be replaced with a second different amino acid residue, wherein the first and second different amino acid residues are the same or different.
  • the isolated immunoglobulin heavy chain polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that residue 9 of SEQ ID NO: 1 is replaced with a methionine (M) residue.
  • M methionine
  • the isolated immunoglobulin heavy chain polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that (a) residue 7 of SEQ ID NO: 2 is replaced with an asparagine (N) residue, (b) residue 8 of SEQ ID NO: 2 is replaced with a serine (S) residue, (c) residue 9 of SEQ ID NO: 2 is replaced with a threonine (T) residue, or (d) any combination of (a)-(c).
  • the isolated immunoglobulin heavy chain polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that (a) residue 1 of SEQ ID NO: 3 is replaced with a glutamic acid (E) residue, (b) residue 2 of SEQ ID NO: 3 is replaced with a tyrosine (Y) residue, (c) residue 5 of SEQ ID NO: 3 is replaced with a serine (S) residue, or (d) any combination of (a)-(c).
  • immunoglobulin heavy chain polypeptides as described above can comprise any one of the following amino acid sequences: SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, or SEQ ID NO: 11.
  • the invention provides an isolated immunoglobulin heavy chain polypeptide comprises, consists essentially of, or consists of a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, wherein optionally (a) residue 9 of SEQ ID NO: 12 is replaced with a different amino acid residue, (b) residue 8 and/or residue 9 of SEQ ID NO: 13 is replaced with a different amino acid residue, (c) residue 5 of SEQ ID NO: 14 is replaced with a different amino acid residue, or (d) any combination of (a)-(c).
  • CDR complementarity determining region 1
  • inventive immunoglobulin heavy chain polypeptide consists essentially of a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14 and optional amino acid replacements
  • additional components can be included in the polypeptide that do not materially affect the polypeptide (e.g., protein moieties such as biotin that facilitate purification or isolation).
  • the inventive immunoglobulin heavy chain polypeptide consists of a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14 and optional amino acid replacements
  • the polypeptide does not comprise any additional components (i.e., components that are not endogenous to the inventive immunoglobulin heavy chain polypeptide).
  • the isolated immunoglobulin heavy chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that (a) residue 9 of SEQ ID NO: 12 is replaced with a different amino acid residue, (b) residue 8 and/or residue 9 of SEQ ID NO: 13 is replaced with a different amino acid residue, (c) residue 5 of SEQ ID NO: 14 is replaced with a different amino acid residue, or (d) any combination of (a)-(c).
  • the isolated immunoglobulin heavy chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that residue 9 of SEQ ID NO: 12 is replaced with a different amino acid residue, residue 8 of SEQ ID NO: 13, and residue 9 of SEQ ID NO: 13 is replaced with a different amino acid residue.
  • the isolated immunoglobulin heavy chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that residue 9 of SEQ ID NO: 12 is replaced with a different amino acid residue and residue 5 of SEQ ID NO: 14 is replaced with a different amino acid residue.
  • the isolated immunoglobulin heavy chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that residue 9 of SEQ ID NO: 12 is replaced with a different amino acid residue, residue 8 of SEQ ID NO: 13 is replaced with a different amino acid residue, residue 9 of SEQ ID NO: 13 is replaced with a different amino acid residue, and residue 5 of SEQ ID NO: 14 is replaced with a different amino acid residue.
  • residue 9 of SEQ ID NO: 12, residues 8 and 9 of SEQ ID NO: 13, and residue 5 of SEQ ID NO: 14 can be replaced with any suitable amino acid residue that can be the same or different in each position.
  • the amino acid residue of a first position can be replaced with a first different amino acid residue
  • the amino acid residue of a second position can be replaced with a second different amino acid residue, wherein the first and second different amino acid residues are the same or different.
  • the isolated immunoglobulin heavy chain polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that residue 9 of SEQ ID NO: 12 is replaced with a leucine (L) residue.
  • the isolated immunoglobulin heavy chain polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that (a) residue 8 of SEQ ID NO: 13 is replaced with a tyrosine (Y) residue, and/or (b) residue 9 of SEQ ID NO: 13 is replaced with an alanine (A) residue.
  • the isolated immunoglobulin heavy chain polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that residue 5 of SEQ ID NO: 14 is replaced with a threonine (T) residue.
  • T threonine
  • immunoglobulin heavy chain polypeptides as described above can comprise any one of the following amino acid sequences: SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, or SEQ ID NO: 18.
  • the invention provides an isolated immunoglobulin heavy chain polypeptide comprises, consists essentially of, or consists of a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 19, a CDR2 amino acid sequence of SEQ ID NO: 20, and a CDR3 amino acid sequence of SEQ ID NO: 21.
  • CDR complementarity determining region 1
  • inventive immunoglobulin heavy chain polypeptide consists essentially of a CDR1 amino acid sequence of SEQ ID NO: 19, a CDR2 amino acid sequence of SEQ ID NO: 20, and a CDR3 amino acid sequence of SEQ ID NO: 21, additional components can be included in the polypeptide that do not materially affect the polypeptide (e.g., protein moieties such as biotin that facilitate purification or isolation).
  • the inventive immunoglobulin heavy chain polypeptide consists of a CDR1 amino acid sequence of SEQ ID NO: 19, a CDR2 amino acid sequence of SEQ ID NO: 20, and a CDR3 amino acid sequence of SEQ ID NO: 21, the polypeptide does not comprise any additional components (i.e., components that are not endogenous to the inventive immunoglobulin heavy chain polypeptide).
  • Exemplary immunoglobulin heavy chain polypeptides as described above can comprise any one of the following amino acid sequences: SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, or SEQ ID NO: 25.
  • one or more amino acids can be inserted into the aforementioned immunoglobulin heavy chain polypeptides. Any number of any suitable amino acids can be inserted into the amino acid sequence of the immunoglobulin heavy chain polypeptide. In this respect, at least one amino acid (e.g., 2 or more, 5 or more, or 10 or more amino acids), but not more than 20 amino acids (e.g., 18 or less, 15 or less, or 12 or less amino acids), can be inserted into the amino acid sequence of the immunoglobulin heavy chain polypeptide. Preferably, 1-10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) are inserted into the amino acid sequence of the immunoglobulin heavy chain polypeptide.
  • amino acids e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids
  • the amino acid(s) can be inserted into any one of the aforementioned immunoglobulin heavy chain polypeptides in any suitable location.
  • the amino acid(s) are inserted into a CDR (e.g., CDR1, CDR2, or CDR3) of the immunoglobulin heavy chain polypeptide.
  • the invention provides an isolated immunoglobulin heavy chain polypeptide which comprises an amino acid sequence that is at least 90% identical (e.g., at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical) to any one of SEQ ID NOs: 4-11, SEQ ID NOs: 15-18, and SEQ ID NOs: 22-25.
  • Nucleic acid or amino acid sequence “identity,” as described herein, can be determined by comparing a nucleic acid or amino acid sequence of interest to a reference nucleic acid or amino acid sequence.
  • the percent identity is the number of nucleotides or amino acid residues that are the same (i.e., that are identical) as between the sequence of interest and the reference sequence divided by the length of the longest sequence (i.e., the length of either the sequence of interest or the reference sequence, whichever is longer).
  • a number of mathematical algorithms for obtaining the optimal alignment and calculating identity between two or more sequences are known and incorporated into a number of available software programs.
  • Such programs include CLUSTAL-W, T-Coffee, and ALIGN (for alignment of nucleic acid and amino acid sequences), BLAST programs (e.g., BLAST 2.1, BL2SEQ, and later versions thereof) and FASTA programs (e.g., FASTA3x, FASTM, and SSEARCH) (for sequence alignment and sequence similarity searches).
  • BLAST programs e.g., BLAST 2.1, BL2SEQ, and later versions thereof
  • FASTA programs e.g., FASTA3x, FASTM, and SSEARCH
  • Sequence alignment algorithms also are disclosed in, for example, Altschul et al., J. Molecular Biol., 215(3): 403-410 (1990), Beigert et al., Proc. Natl. Acad. Sci.
  • the invention provides an immunoglobulin light chain polypeptide that comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 26 and a CDR2 amino acid sequence of SEQ ID NO: 27.
  • the isolated immunoglobulin light chain polypeptide comprises, consists essentially of, or consists of a CDR1 amino acid sequence of SEQ ID NO: 26 and a CDR2 amino acid sequence of SEQ ID NO: 27.
  • inventive immunoglobulin light chain polypeptide consists essentially of a CDR1 amino acid sequence of SEQ ID NO: 26 and a CDR2 amino acid sequence of SEQ ID NO: 27.
  • additional components can be included in the polypeptide that do not materially affect the polypeptide (e.g., protein moieties such as biotin that facilitate purification or isolation).
  • the inventive immunoglobulin light chain polypeptide consists of a CDR1 amino acid sequence of SEQ ID NO: 26 and a CDR2 amino acid sequence of SEQ ID NO: 27, the polypeptide does not comprise any additional components (i.e., components that are not endogenous to the inventive immunoglobulin light chain polypeptide).
  • Exemplary immunoglobulin light chain polypeptides as described above can comprise SEQ ID NO: 28 or SEQ ID NO: 29.
  • the invention provides an isolated immunoglobulin light chain polypeptide comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31.
  • the isolated immunoglobulin light chain polypeptide comprises, consists of, or consists essentially of a CDR1 amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31, wherein optionally residue 12 of SEQ ID NO: 30 is replaced with a different amino acid residue.
  • inventive immunoglobulin light chain polypeptide consists essentially of a CDR1 amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31 and optional amino acid replacements
  • additional components can be included in the polypeptide that do not materially affect the polypeptide (e.g., protein moieties such as biotin that facilitate purification or isolation).
  • the inventive immunoglobulin light chain polypeptide consists of a CDR1 amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31 and optional amino acid replacements
  • the polypeptide does not comprise any additional components (i.e., components that are not endogenous to the inventive immunoglobulin light chain polypeptide).
  • the isolated immunoglobulin light chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31, except that residue 12 of SEQ ID NO: 30 is replaced with a different amino acid residue.
  • Residue 12 of SEQ ID NO: 30 can be replaced with any suitable amino acid residue.
  • the isolated immunoglobulin light chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31, except that residue 12 of SEQ ID NO: 30 is replaced with a threonine (T) residue.
  • Exemplary immunoglobulin light chain polypeptides as described above can comprise any one of the following amino acid sequences: SEQ ID NO: 32, SEQ ID NO: 33, or SEQ ID NO: 34.
  • the invention provides an isolated immunoglobulin light chain polypeptide comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37.
  • CDR complementarity determining region 1
  • the immunoglobulin light chain polypeptide comprises, consists essentially of, or consists of a CDR1 amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37, wherein optionally (a) residue 5 of SEQ ID NO: 36 is replaced with a different amino acid residue, and/or (b) residue 4 of SEQ ID NO: 37 is replaced with a different amino acid residue.
  • inventive immunoglobulin light chain polypeptide consists essentially of a CDR1 amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37 and optional amino acid replacements
  • additional components can be included in the polypeptide that do not materially affect the polypeptide (e.g., protein moieties such as biotin that facilitate purification or isolation).
  • the inventive immunoglobulin light chain polypeptide consists of a CDR1 amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37 and optional amino acid replacements
  • the polypeptide does not comprise any additional components (i.e., components that are not endogenous to the inventive immunoglobulin light chain polypeptide).
  • the isolated immunoglobulin light chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37.
  • the isolated immunoglobulin light chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37, except that (a) residue 5 of SEQ ID NO: 36 is replaced with a different amino acid residue, and/or (b) residue 4 of SEQ ID NO: 37 is replaced with a different amino acid residue.
  • residue 5 of SEQ ID NO: 36 and residue 4 of SEQ ID NO: 37 can be replaced with any suitable amino acid residue that can be the same or different in each position.
  • the amino acid residue of a first position can be replaced with a first different amino acid residue
  • the amino acid residue of a second position can be replaced with a second different amino acid residue, wherein the first and second different amino acid residues are the same or different.
  • the isolated immunoglobulin light chain polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37, except that (a) residue 5 of SEQ ID NO: 36 is replaced with a leucine (L) residue, and/or (b) residue 4 of SEQ ID NO: 37 is replaced with an asparagine (N) residue.
  • Exemplary immunoglobulin light chain polypeptides as described above can comprise any one of the following amino acid sequences: SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, or SEQ ID NO: 41.
  • one or more amino acids can be inserted into the aforementioned immunoglobulin light chain polypeptides. Any number of any suitable amino acids can be inserted into the amino acid sequence of the immunoglobulin light chain polypeptide. In this respect, at least one amino acid (e.g., 2 or more, 5 or more, or 10 or more amino acids), but not more than 20 amino acids (e.g., 18 or less, 15 or less, or 12 or less amino acids), can be inserted into the amino acid sequence of the immunoglobulin light chain polypeptide. Preferably, 1-10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) are inserted in to the amino acid sequence of the immunoglobulin light chain polypeptide.
  • amino acids e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids
  • the amino acid(s) can be inserted into any one of the aforementioned immunoglobulin light chain polypeptides in any suitable location.
  • the amino acid(s) are inserted into a CDR (e.g., CDR1, CDR2, or CDR3) of the immunoglobulin light chain polypeptide.
  • the invention provides an isolated immunoglobulin light chain polypeptide which comprises an amino acid sequence that is at least 90% identical (e.g., at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical) to any one of SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, and SEQ ID NO: 41.
  • Nucleic acid or amino acid sequence “identity,” as described herein, can be determined using the methods described herein.
  • the invention provides an isolated programmed death 1 (PD-1)-binding agent comprising, consisting essentially of, or consisting of the inventive isolated amino acid sequences described herein.
  • programmed death 1 (PD-1)-binding agent is meant a molecule, preferably a proteinaceous molecule, that binds specifically to the programmed death 1 protein (PD-1).
  • the PD-1-binding agent is an antibody or a fragment (e.g., immunogenic fragment) thereof.
  • the isolated PD-1-binding agent of the invention comprises, consists essentially of, or consists of the inventive isolated immunoglobulin heavy chain polypeptide and/or the inventive isolated immunoglobulin light chain polypeptide.
  • the isolated PD-1-binding agent comprises, consists essentially of, or consists of the inventive immunoglobulin heavy chain polypeptide or the inventive immunoglobulin light chain polypeptide. In another embodiment, the isolated PD-1-binding agent comprises, consists essentially of, or consists of the inventive immunoglobulin heavy chain polypeptide and the inventive immunoglobulin light chain polypeptide.
  • the invention is not limited to an isolated PD-1-binding agent that comprises, consists essentially of, or consists of an immunoglobulin heavy chain polypeptide and/or light chain polypeptide having replacements, insertions, and/or deletions of the specific amino acid residues disclosed herein.
  • any amino acid residue of the inventive immunoglobulin heavy chain polypeptide and/or the inventive immunoglobulin light chain polypeptide can be replaced, in any combination, with a different amino acid residue, or can be deleted or inserted, so long as the biological activity of the PD-1-binding agent is enhanced or improved as a result of the amino acid replacements, insertions, and/or deletions.
  • the “biological activity” of an PD-1-binding agent refers to, for example, binding affinity for PD-1 or a particular PD-1 epitope, neutralization or inhibition of PD-1 protein binding to its ligands PD-L1 and PD-L1, neutralization or inhibition of PD-1 protein activity in vivo (e.g., IC 50 ), pharmacokinetics, and cross-reactivity (e.g., with non-human homologs or orthologs of the PD-1 protein, or with other proteins or tissues).
  • Other biological properties or characteristics of an antigen-binding agent recognized in the art include, for example, avidity, selectivity, solubility, folding, immunotoxicity, expression, and formulation.
  • the aforementioned properties or characteristics can be observed, measured, and/or assessed using standard techniques including, but not limited to, ELISA, competitive ELISA, surface plasmon resonance analysis (BIACORETM), or KINEXATM, in vitro or in vivo neutralization assays, receptor-ligand binding assays, cytokine or growth factor production and/or secretion assays, and signal transduction and immunohistochemistry assays.
  • standard techniques including, but not limited to, ELISA, competitive ELISA, surface plasmon resonance analysis (BIACORETM), or KINEXATM, in vitro or in vivo neutralization assays, receptor-ligand binding assays, cytokine or growth factor production and/or secretion assays, and signal transduction and immunohistochemistry assays.
  • inhibitor or “neutralize,” as used herein with respect to the activity of a PD-1-binding agent, refer to the ability to substantially antagonize, prohibit, prevent, restrain, slow, disrupt, alter, eliminate, stop, or reverse the progression or severity of, for example, the biological activity of a PD-1 protein, or a disease or condition associated with an PD-1 protein.
  • the isolated PD-1-binding agent of the invention preferably inhibits or neutralizes the activity of a PD-1 protein by at least about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 100%, or a range defined by any two of the foregoing values.
  • the isolated PD-1-binding agent of the invention can be a whole antibody, as described herein, or an antibody fragment.
  • fragment of an antibody “antibody fragment,” and “functional fragment of an antibody” are used interchangeably herein to mean one or more fragments of an antibody that retain the ability to specifically bind to an antigen (see, generally, Holliger et al., Nat. Biotech., 23(9): 1126-1129 (2005)).
  • the isolated PD-1 binding agent can contain any PD-1-binding antibody fragment.
  • the antibody fragment desirably comprises, for example, one or more CDRs, the variable region (or portions thereof), the constant region (or portions thereof), or combinations thereof.
  • antibody fragments include, but are not limited to, (i) a Fab fragment, which is a monovalent fragment consisting of the V L , V H , C L , and CH 1 domains, (ii) a F(ab′) 2 fragment, which is a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region, (iii) a Fv fragment consisting of the V L and V H domains of a single arm of an antibody, (iv) a Fab′ fragment, which results from breaking the disulfide bridge of an F(ab′) 2 fragment using mild reducing conditions, (v) a disulfide-stabilized Fv fragment (dsFv), and (vi) a domain antibody (dAb), which is an antibody single variable region domain (VH or VL) polypeptide that specifically binds antigen.
  • a Fab fragment which is a monovalent fragment consisting of the V L , V H , C L , and CH 1 domains
  • the isolated PD-1-binding agent comprises a fragment of the immunoglobulin heavy chain or light chain polypeptide
  • the fragment can be of any size so long as the fragment binds to, and preferably inhibits the activity of, a PD-1 protein.
  • a fragment of the immunoglobulin heavy chain polypeptide desirably comprises between about 5 and 18 (e.g., about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or a range defined by any two of the foregoing values) amino acids.
  • a fragment of the immunoglobulin light chain polypeptide desirably comprises between about 5 and 18 (e.g., about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or a range defined by any two of the foregoing values) amino acids.
  • the antibody or antibody fragment desirably comprises a heavy chain constant region (F c ) of any suitable class.
  • F c heavy chain constant region
  • the antibody or antibody fragment comprises a heavy chain constant region that is based upon wild-type IgG1, IgG2, or IgG4 antibodies, or variants thereof.
  • the PD-1-binding agent also can be a single chain antibody fragment.
  • single chain antibody fragments include, but are not limited to, (i) a single chain Fv (scFv), which is a monovalent molecule consisting of the two domains of the Fv fragment (i.e., V L and V H ) joined by a synthetic linker which enables the two domains to be synthesized as a single polypeptide chain (see, e.g., Bird et al., Science, 242: 423-426 (1988); Huston et al., Proc. Natl. Acad. Sci. USA, 85: 5879-5883 (1988); and Osbourn et al., Nat.
  • scFv single chain Fv
  • a diabody which is a dimer of polypeptide chains, wherein each polypeptide chain comprises a V H connected to a V L by a peptide linker that is too short to allow pairing between the V H and V L on the same polypeptide chain, thereby driving the pairing between the complementary domains on different V H -V L polypeptide chains to generate a dimeric molecule having two functional antigen binding sites.
  • Antibody fragments are known in the art and are described in more detail in, e.g., U.S. Patent Application Publication 2009/0093024 A1.
  • the isolated PD-1-binding agent also can be an intrabody or fragment thereof.
  • An intrabody is an antibody which is expressed and which functions intracellularly. Intrabodies typically lack disulfide bonds and are capable of modulating the expression or activity of target genes through their specific binding activity. Intrabodies include single domain fragments such as isolated V H and V L domains and scFvs.
  • An intrabody can include sub-cellular trafficking signals attached to the N or C terminus of the intrabody to allow expression at high concentrations in the sub-cellular compartments where a target protein is located.
  • an intrabody Upon interaction with a target gene, an intrabody modulates target protein function and/or achieves phenotypic/functional knockout by mechanisms such as accelerating target protein degradation and sequestering the target protein in a non-physiological sub-cellular compartment.
  • Other mechanisms of intrabody-mediated gene inactivation can depend on the epitope to which the intrabody is directed, such as binding to the catalytic site on a target protein or to epitopes that are involved in protein-protein, protein-DNA, or protein-RNA interactions.
  • the isolated PD-1-binding agent also can be an antibody conjugate.
  • the isolated PD-1-binding agent can be a conjugate of (1) an antibody, an alternative scaffold, or fragments thereof, and (2) a protein or non-protein moiety comprising the PD-1-binding agent.
  • the PD-1-binding agent can be all or part of an antibody conjugated to a peptide, a fluorescent molecule, or a chemotherapeutic agent.
  • the isolated PD-1-binding agent can be, or can be obtained from, a human antibody, a non-human antibody, or a chimeric antibody.
  • chimeric is meant an antibody or fragment thereof comprising both human and non-human regions.
  • the isolated PD-1-binding agent is a humanized antibody.
  • a “humanized” antibody is a monoclonal antibody comprising a human antibody scaffold and at least one CDR obtained or derived from a non-human antibody.
  • Non-human antibodies include antibodies isolated from any non-human animal, such as, for example, a rodent (e.g., a mouse or rat).
  • a humanized antibody can comprise, one, two, or three CDRs obtained or derived from a non-human antibody.
  • CDRH3 of the inventive PD-1-binding agent is obtained or derived from a mouse monoclonal antibody, while the remaining variable regions and constant region of the inventive PD-1-binding agent are obtained or derived from a human monoclonal antibody.
  • a human antibody, a non-human antibody, a chimeric antibody, or a humanized antibody can be obtained by any means, including via in vitro sources (e.g., a hybridoma or a cell line producing an antibody recombinantly) and in vivo sources (e.g., rodents).
  • in vitro sources e.g., a hybridoma or a cell line producing an antibody recombinantly
  • in vivo sources e.g., rodents.
  • a human antibody or a chimeric antibody can be generated using a transgenic animal (e.g., a mouse) wherein one or more endogenous immunoglobulin genes are replaced with one or more human immunoglobulin genes.
  • transgenic mice wherein endogenous antibody genes are effectively replaced with human antibody genes include, but are not limited to, the Medarex HUMAB-MOUSETM, the Kirin TC MOUSETM, and the Kyowa Kirin KM-MOUSETM (see, e.g., Lonberg, Nat. Biotechnol., 23(9): 1117-25 (2005), and Lonberg, Handb. Exp. Pharmacol., 181: 69-97 (2008)).
  • a humanized antibody can be generated using any suitable method known in the art (see, e.g., An, Z. (ed.), Therapeutic Monoclonal Antibodies: From Bench to Clinic , John Wiley & Sons, Inc. Hoboken, N.J. (2009)), including, e.g., grafting of non-human CDRs onto a human antibody scaffold (see, e.g., Kashmiri et al., Methods, 36(1): 25-34 (2005); and Hou et al., J. Biochem., 144(1): 115-120 (2008)).
  • a humanized antibody can be produced using the methods described in, e.g., U.S. Patent Application Publication 2011/0287485 A1.
  • the PD-1-binding agent binds an epitope of a PD-1 protein which blocks the binding of PD-1 to PD-L1.
  • the invention also provides an isolated or purified epitope of a PD-1 protein which blocks the binding of PD-1 to PD-L1 in an indirect or allosteric manner.
  • the invention also provides one or more isolated or purified nucleic acid sequences that encode the inventive immunoglobulin heavy chain polypeptide, the inventive immunoglobulin light chain polypeptide, and the inventive PD-1-binding agent.
  • nucleic acid sequence is intended to encompass a polymer of DNA or RNA, i.e., a polynucleotide, which can be single-stranded or double-stranded and which can contain non-natural or altered nucleotides.
  • nucleic acid and polynucleotide refer to a polymeric form of nucleotides of any length, either ribonucleotides (RNA) or deoxyribonucleotides (DNA). These terms refer to the primary structure of the molecule, and thus include double- and single-stranded DNA, and double- and single-stranded RNA.
  • RNA or DNA made from nucleotide analogs and modified polynucleotides such as, though not limited to, methylated and/or capped polynucleotides.
  • Nucleic acids are typically linked via phosphate bonds to form nucleic acid sequences or polynucleotides, though many other linkages are known in the art (e.g., phosphorothioates, boranophosphates, and the like).
  • Nucleic acid sequences encoding the inventive immunoglobulin heavy chain polypeptides include, for example, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, and SEQ ID NO: 55.
  • Nucleic acid sequences encoding the inventive immunoglobulin light chain polypeptides include, for example, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 62, SEQ ID NO: 63, and SEQ ID NO: 64.
  • the invention further provides a vector comprising one or more nucleic acid sequences encoding the inventive immunoglobulin heavy chain polypeptide, the inventive immunoglobulin light chain polypeptide, and/or the inventive PD-1-binding agent.
  • the vector can be, for example, a plasmid, episome, cosmid, viral vector (e.g., retroviral or adenoviral), or phage.
  • Suitable vectors and methods of vector preparation are well known in the art (see, e.g., Sambrook et al., Molecular Cloning, a Laboratory Manual, 3 rd edition , Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (2001), and Ausubel et al., Current Protocols in Molecular Biology , Greene Publishing Associates and John Wiley & Sons, New York, N.Y. (1994)).
  • the vector preferably comprises expression control sequences, such as promoters, enhancers, polyadenylation signals, transcription terminators, internal ribosome entry sites (IRES), and the like, that provide for the expression of the coding sequence in a host cell.
  • expression control sequences are known in the art and described in, for example, Goeddel, Gene Expression Technology: Methods in Enzymology , Vol. 185, Academic Press, San Diego, Calif. (1990).
  • promoters including constitutive, inducible, and repressible promoters, from a variety of different sources are well known in the art.
  • Representative sources of promoters include for example, virus, mammal, insect, plant, yeast, and bacteria, and suitable promoters from these sources are readily available, or can be made synthetically, based on sequences publicly available, for example, from depositories such as the ATCC as well as other commercial or individual sources.
  • Promoters can be unidirectional (i.e., initiate transcription in one direction) or bi-directional (i.e., initiate transcription in either a 3′ or 5′ direction).
  • Non-limiting examples of promoters include, for example, the T7 bacterial expression system, pBAD (araA) bacterial expression system, the cytomegalovirus (CMV) promoter, the SV40 promoter, the RSV promoter.
  • Inducible promoters include, for example, the Tet system (U.S. Pat. Nos. 5,464,758 and 5,814,618), the Ecdysone inducible system (No et al., Proc. Natl. Acad.
  • Enhancers refers to a DNA sequence that increases transcription of, for example, a nucleic acid sequence to which it is operably linked. Enhancers can be located many kilobases away from the coding region of the nucleic acid sequence and can mediate the binding of regulatory factors, patterns of DNA methylation, or changes in DNA structure. A large number of enhancers from a variety of different sources are well known in the art and are available as or within cloned polynucleotides (from, e.g., depositories such as the ATCC as well as other commercial or individual sources). A number of polynucleotides comprising promoters (such as the commonly-used CMV promoter) also comprise enhancer sequences. Enhancers can be located upstream, within, or downstream of coding sequences.
  • the vector also can comprise a “selectable marker gene.”
  • selectable marker gene refers to a nucleic acid sequence that allow cells expressing the nucleic acid sequence to be specifically selected for or against, in the presence of a corresponding selective agent. Suitable selectable marker genes are known in the art and described in, e.g., International Patent Application Publications WO 1992/008796 and WO 1994/028143; Wigler et al., Proc. Natl. Acad. Sci. USA, 77: 3567-3570 (1980); O'Hare et al., Proc. Natl. Acad. Sci. USA, 78: 1527-1531 (1981); Mulligan & Berg, Proc. Natl.
  • the vector is an “episomal expression vector” or “episome,” which is able to replicate in a host cell, and persists as an extrachromosomal segment of DNA within the host cell in the presence of appropriate selective pressure (see, e.g., Conese et al., Gene Therapy, 11: 1735-1742 (2004)).
  • Representative commercially available episomal expression vectors include, but are not limited to, episomal plasmids that utilize Epstein Barr Nuclear Antigen 1 (EBNA1) and the Epstein Barr Virus (EBV) origin of replication (oriP).
  • the vectors pREP4, pCEP4, pREP7, and pcDNA3.1 from Invitrogen (Carlsbad, Calif.) and pBK-CMV from Stratagene (La Jolla, Calif.) represent non-limiting examples of an episomal vector that uses T-antigen and the SV40 origin of replication in lieu of EBNA1 and oriP.
  • Suitable vectors include integrating expression vectors, which may randomly integrate into the host cell's DNA, or may include a recombination site to enable the specific recombination between the expression vector and the host cell's chromosome. Such integrating expression vectors may utilize the endogenous expression control sequences of the host cell's chromosomes to effect expression of the desired protein.
  • integrating expression vectors may utilize the endogenous expression control sequences of the host cell's chromosomes to effect expression of the desired protein.
  • Examples of vectors that integrate in a site specific manner include, for example, components of the flp-in system from Invitrogen (Carlsbad, Calif.) (e.g., pcDNATM5/FRT), or the cre-lox system, such as can be found in the pExchange-6 Core Vectors from Stratagene (La Jolla, Calif.).
  • vectors that randomly integrate into host cell chromosomes include, for example, pcDNA3.1 (when introduced in the absence of T-antigen) from Invitrogen (Carlsbad, Calif.), UCOE from Millipore (Billerica, Mass.), and pCI or pFN10A (ACT) FLEXITM from Promega (Madison, Wis.).
  • Viral vectors also can be used.
  • Representative commercially available viral expression vectors include, but are not limited to, the adenovirus-based Per.C6 system available from Crucell, Inc. (Leiden, The Netherlands), the lentiviral-based pLP1 from Invitrogen (Carlsbad, Calif.), and the retroviral vectors pFB-ERV plus pCFB-EGSH from Stratagene (La Jolla, Calif.).
  • Nucleic acid sequences encoding the inventive amino acid sequences can be provided to a cell on the same vector (i.e., in cis).
  • a unidirectional promoter can be used to control expression of each nucleic acid sequence.
  • a combination of bidirectional and unidirectional promoters can be used to control expression of multiple nucleic acid sequences.
  • Nucleic acid sequences encoding the inventive amino acid sequences alternatively can be provided to the population of cells on separate vectors (i.e., in trans). Each of the nucleic acid sequences in each of the separate vectors can comprise the same or different expression control sequences. The separate vectors can be provided to cells simultaneously or sequentially.
  • the vector(s) comprising the nucleic acid(s) encoding the inventive amino acid sequences can be introduced into a host cell that is capable of expressing the polypeptides encoded thereby, including any suitable prokaryotic or eukaryotic cell.
  • the invention provides an isolated cell comprising the inventive vector.
  • Preferred host cells are those that can be easily and reliably grown, have reasonably fast growth rates, have well characterized expression systems, and can be transformed or transfected easily and efficiently.
  • suitable prokaryotic cells include, but are not limited to, cells from the genera Bacillus (such as Bacillus subtilis and Bacillus brevis ), Escherichia (such as E. coli ), Pseudomonas, Streptomyces, Salmonella , and Erwinia .
  • Particularly useful prokaryotic cells include the various strains of Escherichia coli (e.g., K12, HB101 (ATCC No. 33694), DH5 ⁇ , DH10, MC1061 (ATCC No. 53338), and CC102).
  • the vector is introduced into a eukaryotic cell.
  • Suitable eukaryotic cells include, for example, yeast cells, insect cells, and mammalian cells.
  • suitable yeast cells include those from the genera Kluyveromyces, Pichia, Rhinosporidium, Saccharomyces , and Schizosaccharomyces .
  • Preferred yeast cells include, for example, Saccharomyces cerivisae and Pichia pastoris.
  • Suitable insect cells are described in, for example, Kitts et al., Biotechniques, 14: 810-817 (1993); Lucklow, Curr. Opin. Biotechnol., 4: 564-572 (1993); and Lucklow et al., J. Virol., 67: 4566-4579 (1993).
  • Preferred insect cells include Sf-9 and HI5 (Invitrogen, Carlsbad, Calif.).
  • mammalian cells are utilized in the invention.
  • suitable mammalian host cells are known in the art, and many are available from the American Type Culture Collection (ATCC, Manassas, Va.).
  • suitable mammalian cells include, but are not limited to, Chinese hamster ovary cells (CHO) (ATCC No. CCL61), CHO DHFR-cells (Urlaub et al., Proc. Natl. Acad. Sci. USA, 97: 4216-4220 (1980)), human embryonic kidney (HEK) 293 or 293T cells (ATCC No. CRL1573), and 3T3 cells (ATCC No. CCL92).
  • CHO Chinese hamster ovary cells
  • CHO DHFR-cells Urlaub et al., Proc. Natl. Acad. Sci. USA, 97: 4216-4220 (1980)
  • human embryonic kidney (HEK) 293 or 293T cells ATCC No. CRL1573)
  • suitable mammalian cell lines are the monkey COS-1 (ATCC No. CRL1650) and COS-7 cell lines (ATCC No. CRL1651), as well as the CV-1 cell line (ATCC No. CCL70).
  • Further exemplary mammalian host cells include primate cell lines and rodent cell lines, including transformed cell lines. Normal diploid cells, cell strains derived from in vitro culture of primary tissue, as well as primary explants, are also suitable.
  • Other suitable mammalian cell lines include, but are not limited to, mouse neuroblastoma N2A cells, HeLa, mouse L-929 cells, and BHK or HaK hamster cell lines, all of which are available from the ATCC. Methods for selecting suitable mammalian host cells and methods for transformation, culture, amplification, screening, and purification of cells are known in the art.
  • the mammalian cell is a human cell.
  • the mammalian cell can be a human lymphoid or lymphoid derived cell line, such as a cell line of pre-B lymphocyte origin.
  • human lymphoid cells lines include, without limitation, RAMOS (CRL-1596), Daudi (CCL-213), EB-3 (CCL-85), DT40 (CRL-2111), 18-81 (Jack et al., Proc. Natl. Acad. Sci. USA, 85: 1581-1585 (1988)), Raji cells (CCL-86), and derivatives thereof.
  • a nucleic acid sequence encoding the inventive amino acid sequence may be introduced into a cell by “transfection,” “transformation,” or “transduction.”
  • “Transfection,” “transformation,” or “transduction,” as used herein, refer to the introduction of one or more exogenous polynucleotides into a host cell by using physical or chemical methods.
  • Many transfection techniques are known in the art and include, for example, calcium phosphate DNA co-precipitation (see, e.g., Murray E. J. (ed.), Methods in Molecular Biology, Vol.
  • Phage or viral vectors can be introduced into host cells, after growth of infectious particles in suitable packaging cells, many of which are commercially available.
  • the invention provides a composition comprising an effective amount of the inventive immunoglobulin heavy chain polypeptide, the inventive immunoglobulin light chain polypeptide, the inventive PD-1-binding agent, the inventive nucleic acid sequence encoding any of the foregoing, or the inventive vector comprising the inventive nucleic acid sequence.
  • the composition is a pharmaceutically acceptable (e.g., physiologically acceptable) composition, which comprises a carrier, preferably a pharmaceutically acceptable (e.g., physiologically acceptable) carrier, and the inventive amino acid sequences, antigen-binding agent, or vector.
  • a pharmaceutically acceptable composition which comprises a carrier, preferably a pharmaceutically acceptable (e.g., physiologically acceptable) carrier, and the inventive amino acid sequences, antigen-binding agent, or vector.
  • Any suitable carrier can be used within the context of the invention, and such carriers are well known in the art.
  • the choice of carrier will be determined, in part, by the particular site to which the composition may be administered and the particular method used to administer the composition.
  • the composition optionally can be sterile.
  • the composition can be frozen or lyophilized for storage and reconstituted in a suitable sterile carrier prior to use.
  • the compositions can be generated in accordance with conventional techniques described in, e.g., Remington: The Science and Practice of Pharmacy, 21 st Edition , Lippincott Williams & Wilkins, Philadelphia, Pa. (2001).
  • the invention further provides a method of treating any disease or disorder in which the improper expression (e.g., overexpression) or increased activity of a PD-1 protein causes or contributes to the pathological effects of the disease, or a decrease in PD-1 protein levels or activity has a therapeutic benefit in mammals, preferably humans.
  • the invention also provides a method of treating a cancer or an infectious disease in a mammal. The method comprises administering the aforementioned composition to a mammal having a cancer or an infectious disease, whereupon the cancer or infectious disease is treated in the mammal.
  • PD-1 is abnormally expressed in a variety of cancers (see, e.g., Brown et al., J.
  • inventive method can be used to treat any type of cancer known in the art, such as, for example, melanoma, renal cell carcinoma, lung cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, gall bladder cancer, laryngeal cancer, liver cancer, thyroid cancer, stomach cancer, salivary gland cancer, prostate cancer, pancreatic cancer, or Merkel cell carcinoma (see, e.g., Bhatia et al., Curr. Oncol. Rep., 13(6): 488-497 (2011)).
  • cancer known in the art, such as, for example, melanoma, renal cell carcinoma, lung cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, gall bladder cancer, laryngeal cancer, liver cancer, thyroid cancer, stomach cancer, salivary gland cancer, prostate cancer, pancreatic cancer, or Merkel cell carcinoma (see, e.g., Bhatia et al., Curr. Oncol. Rep., 13(6): 488-497 (2011)).
  • the inventive method can be used to treat any type of infectious disease a disease or disorder caused by a bacterium, a virus, a fungus, or a parasite).
  • infectious diseases that can be treated by the inventive method include, but are not limited to, diseases caused by a human immunodeficiency virus (HIV), a respiratory syncytial virus (RSV), an influenza virus, a dengue virus, a hepatitis B virus (HBV, or a hepatitis C virus (HCV)).
  • compositions comprising the inventive immunoglobulin heavy chain polypeptide, the inventive immunoglobulin light chain polypeptide, the inventive PD-1-binding agent, the inventive nucleic acid sequence encoding any of the foregoing, or the inventive vector comprising the inventive nucleic acid sequence induces an immune response against a cancer or infectious disease in a mammal.
  • An “immune response” can entail, for example, antibody production and/or the activation of immune effector cells (e.g., T-cells).
  • the terms “treatment,” “treating,” and the like refer to obtaining a desired pharmacologic and/or physiologic effect.
  • the effect is therapeutic, i.e., the effect partially or completely cures a disease and/or adverse symptom attributable to the disease.
  • the inventive method comprises administering a “therapeutically effective amount” of the PD-1-binding agent.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result.
  • the therapeutically effective amount may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the PD-1-binding agent to elicit a desired response in the individual.
  • a therapeutically effective amount of a PD-1-binding agent of the invention is an amount which decreases PD-1 protein bioactivity in a human and/or enhances the immune response against a cancer or infectious disease.
  • the pharmacologic and/or physiologic effect may be prophylactic, i.e., the effect completely or partially prevents a disease or symptom thereof.
  • the inventive method comprises administering a “prophylactically effective amount” of the PD-1-binding agent.
  • a “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired prophylactic result (e.g., prevention of disease onset).
  • a typical dose can be, for example, in the range of 1 pg/kg to 20 mg/kg of animal or human body weight; however, doses below or above this exemplary range are within the scope of the invention.
  • the daily parenteral dose can be about 0.00001 ⁇ g/kg to about 20 mg/kg of total body weight (e.g., about 0.001 ⁇ g/kg, about 0.1 ⁇ g/kg, about 1 ⁇ g/kg, about 5 ⁇ g/kg, about 10 ⁇ g/kg, about 100 ⁇ g/kg, about 500 ⁇ g/kg, about 1 mg/kg, about 5 mg/kg, about 10 mg/kg, or a range defined by any two of the foregoing values), preferably from about 0.1 ⁇ g/kg to about 10 mg/kg of total body weight (e.g., about 0.5 ⁇ g/kg, about 1 ⁇ g/kg, about 50 ⁇ g/kg, about 150 ⁇ g/kg, about 300 ⁇ g/kg, about 750 ⁇ g/kg,
  • Therapeutic or prophylactic efficacy can be monitored by periodic assessment of treated patients. For repeated administrations over several days or longer, depending on the condition, the treatment can be repeated until a desired suppression of disease symptoms occurs. However, other dosage regimens may be useful and are within the scope of the invention.
  • the desired dosage can be delivered by a single bolus administration of the composition, by multiple bolus administrations of the composition, or by continuous infusion administration of the composition.
  • composition comprising an effective amount of the inventive immunoglohulin heavy chain polypeptide, the inventive immunoglobulin light chain polypeptide, the inventive PD-1-binding agent, the inventive nucleic acid sequence encoding any of the foregoing, or the inventive vector comprising the inventive nucleic acid sequence can be administered to a mammal using standard administration techniques, including oral, intravenous, intraperitoneal, subcutaneous, pulmonary, transdermal, intramuscular, intranasal, buccal, sublingual, or suppository administration.
  • the composition preferably is suitable for parenteral administration.
  • parenteral includes intravenous, intramuscular, subcutaneous, rectal, vaginal, and intraperitoneal administration. More preferably, the composition is administered to a mammal using peripheral systemic delivery by intravenous, intraperitoneal, or subcutaneous injection.
  • the biological activity of the inventive PD-1-binding agent can be measured by any suitable method known in the art.
  • the biological activity can be assessed by determining the stability of a particular PD-1-binding agent.
  • the PD-1-binding agent e.g., an antibody
  • the PD-1-binding agent has an in vivo half life between about 30 minutes and 45 days (e.g., about 30 minutes, about 45 minutes, about 1 hour, about 2 hours, about 4 hours, about 6 hours, about 10 hours, about 12 hours, about 1 day, about 5 days, about 10 days, about 15 days, about 25 days, about 35 days, about 40 days, about 45 days, or a range defined by any two of the foregoing values).
  • the PD-1-binding agent has an in vivo half life between about 2 hours and 20 days (e.g., about 5 hours, about 10 hours, about 15 hours, about 20 hours, about 2 days, about 3 days, about 7 days, about 12 days, about 14 days, about 17 days, about 19 days, or a range defined by any two of the foregoing values).
  • the PD-1-binding agent has an in vivo half life between about 10 days and about 40 days (e.g., about 10 days, about 13 days, about 16 days, about 18 days, about 20 days, about 23 days, about 26 days, about 29 days, about 30 days, about 33 days, about 37 days, about 38 days, about 39 days, about 40 days, or a range defined by any two of the foregoing values).
  • the biological activity of a particular PD-1-binding agent also can be assessed by determining its binding affinity to a PD-1 protein or an epitope thereof.
  • affinity refers to the equilibrium constant for the reversible binding of two agents and is expressed as the dissociation constant (K D ).
  • Affinity of a binding agent to a ligand, such as affinity of an antibody for an epitope can be, for example, from about 1 picomolar (pM) to about 100 micromolar ( ⁇ M) (e.g., from about 1 picomolar (pM) to about 1 nanomolar (nM), from about 1 nM to about 1 micromolar ( ⁇ M), or from about 1 ⁇ M to about 100 ⁇ M).
  • the PD-1-binding agent can bind to an PD-1 protein with a K D less than or equal to 1 nanomolar (e.g., 0.9 nM, 0.8 nM, 0.7 nM, 0.6 nM, 0.5 nM, 0.4 nM, 0.3 nM, 0.2 nM, 0.1 nM, 0.05 nM, 0.025 nM, 0.01 nM, 0.001 nM, or a range defined by any two of the foregoing values).
  • 1 nanomolar e.g., 0.9 nM, 0.8 nM, 0.7 nM, 0.6 nM, 0.5 nM, 0.4 nM, 0.3 nM, 0.2 nM, 0.1 nM, 0.05 nM, 0.025 nM, 0.01 nM, 0.001 nM, or a range defined by any two of the foregoing values).
  • the PD-1-binding agent can bind to PD-1 with a K D less than or equal to 200 pM (e.g., 190 pM, 175 pM, 150 pM, 125 pM, 110 pM, 100 pM, 90 pM, 80 pM, 75 pM, 60 pM, 50 pM, 40 pM, 30 pM, 25 pM, 20 pM, 15 pM, 10 pM, 5 pM, 1 pM, or a range defined by any two of the foregoing values).
  • Immunoglobulin affinity for an antigen or epitope of interest can be measured using any art-recognized assay.
  • Such methods include, for example, fluorescence activated cell sorting (FACS), separable beads (e.g., magnetic beads), surface plasmon resonance (SPR), solution phase competition (KINEXATM), antigen panning, and/or ELISA (see, e.g., Janeway et al. (ed.s.), Immunobiology, 5th ed., Garland Publishing, New York, N.Y., 2001).
  • FACS fluorescence activated cell sorting
  • separable beads e.g., magnetic beads
  • SPR surface plasmon resonance
  • KINEXATM solution phase competition
  • the PD-1-binding agent of the invention may be administered alone or in combination with other drugs (e.g., as an adjuvant).
  • the PD-1-binding agent can be administered in combination with other agents for the treatment or prevention of the diseases disclosed herein.
  • the PD-1-binding agent can be used in combination with at least one other anticancer agent including, for example, any chemotherapeutic agent known in the art, ionization radiation, small molecule anticancer agents, cancer vaccines, biological therapies (e.g., other monoclonal antibodies, cancer-killing viruses, gene therapy, and adoptive T-cell transfer), and/or surgery.
  • the PD-1-binding agent can be administered in combination with at least one anti-bacterial agent or at least one anti-viral agent.
  • the anti-bacterial agent can be any suitable antibiotic known in the art.
  • the anti-viral agent can be any vaccine of any suitable type that specifically targets a particular virus (e.g., live-attenuated vaccines, subunit vaccines, recombinant vector vaccines, and small molecule anti-viral therapies (e.g., viral replication inhibitors and nucleoside analogs).
  • the inventive PD-1 binding agent can be administered in combination with other agents that inhibit immune checkpoint pathways.
  • the inventive PD-1 binding agent can be administered in combination with agents that inhibit or antagonize the CTLA-4, TIM-3 or LAG-3 pathways.
  • Combination treatments that simultaneously target two or more of these immune checkpoint pathways have demonstrated improved and potentially synergistic antitumor activity (see, e.g., Sakuishi et al., J. Exp. Med., 207: 2187-2194 (2010); Ngiow et al., Cancer Res., 71: 3540-3551 (2011); and Woo et al., Cancer Res., 72: 917-927 (2012)).
  • the inventive PD-1 binding agent is administered in combination with an antibody that binds to TIM-3 and/or an antibody that binds to LAG-3.
  • the inventive method of treating a cancer or an infectious disease in a mammal can further comprise administering to the mammal a composition comprising (i) an antibody that binds to a TIM-3 protein and (ii) a pharmaceutically acceptable carrier or a composition comprising (i) an antibody that binds to a LAG-3 protein and (ii) a pharmaceutically acceptable carrier.
  • the PD-1-binding agent described herein can be used in diagnostic or research applications.
  • the PD-1-binding agent can be used in a method to diagnose a cancer or infectious disease.
  • the PD-1-binding agent can be used in an assay to monitor PD-1 protein levels in a subject being tested for a disease or disorder that is associated with abnormal PD-1 expression.
  • Research applications include, for example, methods that utilize the PD-1-binding agent, and a label to detect a PD-1 protein in a sample, e.g., in a human body fluid or in a cell or tissue extract.
  • the PD-1-binding agent can be used with or without modification, such as covalent or non-covalent labeling with a detectable moiety.
  • the detectable moiety can be a radioisotope (e.g., 3 H, 14 C, 32 P, 35 S, or 125 I), a fluorescent or chemiluminescent compound (e.g., fluorescein isothiocyanate, rhodainine, or luciferin), an enzyme (e.g., alkaline phosphatase, beta-galactosidase, or horseradish peroxidase), or prosthetic groups.
  • a radioisotope e.g., 3 H, 14 C, 32 P, 35 S, or 125 I
  • a fluorescent or chemiluminescent compound e.g., fluorescein isothiocyanate, rhodainine, or luciferin
  • an enzyme e.g., alkaline phosphatase, beta
  • any method known in the art for separately conjugating an antigen-binding agent (e.g., an antibody) to a detectable moiety may be employed in the context of the invention (see, e.g., Hunter et al., Nature, 194: 495-496 (1962); David et al., Biochemistry, 13: 1014-1021 (1974); Pain et al., J. Immunol. Meth., 40: 219-230 (1981); and Nygren, J. Histochem. and Cytochem., 30: 407-412 (1982)).
  • PD-1 protein levels can be measured using the inventive PD-1-binding agent by any suitable method known in the art. Such methods include, for example, radioimmunoassay (RIA), and FACS.
  • RIA radioimmunoassay
  • Normal or standard expression values of PD-1 protein can be established using any suitable technique, e.g., by combining a sample comprising, or suspected of comprising, a PD-1 polypeptide with a PD-1-specific antibody under conditions suitable to form an antigen-antibody complex. The antibody is directly or indirectly labeled with a detectable substance to facilitate detection of the bound or unbound antibody.
  • Suitable detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, and radioactive materials (see, e.g., Zola, Monoclonal Antibodies: A Manual of Techniques , CRC Press, Inc. (1987)).
  • the amount of PD-1 polypeptide expressed in a sample is then compared with a standard value.
  • the PD-1-binding agent can be provided in a kit, i.e., a packaged combination of reagents in predetermined amounts with instructions for performing a diagnostic assay.
  • the kit desirably includes substrates and cofactors required by the enzyme (e.g., a substrate precursor which provides a detectable chromophore or fluorophore).
  • substrates and cofactors required by the enzyme e.g., a substrate precursor which provides a detectable chromophore or fluorophore.
  • other additives may be included in the kit, such as stabilizers, buffers (e.g., a blocking buffer or lysis buffer), and the like.
  • the relative amounts of the various reagents can be varied to provide for concentrations in solution of the reagents which substantially optimize the sensitivity of the assay.
  • the reagents may be provided as dry powders (typically lyophilized), including excipients which on dissolution will provide a reagent solution having the appropriate
  • This example demonstrates a method of generating monoclonal antibodies directed against human PD-1.
  • FIG. 1 Several forms of genes encoding human PD-1 and its ligands PD-L1 and PD-L2 were generated as antigens for use in mouse immunization, hybridoma screening, and affinity maturation of CDR-grafted antibodies, and are schematically depicted in FIG. 1 .
  • Full-length human and cynomolgus monkey PD-1 genes were expressed with their native leader sequence and no added tags using a ubiquitous chromatin opening element (UCOE) single expression vector with hygromycin selection (Millipore, Billerica, Mass.).
  • CHO-K1 cells were stably transfected with Lipofectamine LTX (Life Technologies, Carlsbad, Calif.) according to the manufacturer's instructions.
  • UCOE ubiquitous chromatin opening element
  • cells expressing PD-1 on the cell surface were identified by flow cytometry using a PE-conjugated mouse antibody to human PD-1 (BD Bioscience, Franklin Lakes, N.J.) and subcloned. Subclones were then selected for high-level and uniform PD-1 expression.
  • Nucleic acid sequences encoding soluble monomeric forms of the extracellular domain (ECD) of human and cynomolgus monkey PD-1 were constructed with His tags appended to the C-terminus of the ECD or as soluble dimeric fusion proteins with mouse IgG2a Fc as indicated in FIG. 1 .
  • Nucleic acid sequences encoding soluble dimeric forms of the ECDs of human PD-L1 and PD-L2 were constructed as fusion proteins with mouse IgG1 Fc as indicated in FIG. 1 . Soluble proteins were expressed transiently in HEK 293 cells or in stable CHO cell lines using standard techniques.
  • His-tagged proteins were purified from cell culture supernatant via Ni-affinity column chromatography followed by size exclusion chromatography. IgG-Fc fusion proteins were purified using protein A/G affinity chromatography. Purified proteins were analyzed by SDS-PAGE and size-exclusion chromatography to ensure homogeneity. Additionally, identity and size were confirmed by mass spectrometry.
  • purified proteins were labeled with biotin using an NHS ester crosslinker (Thermo-Fisher Scientific, Inc., Waltham, Mass.) or the fluorescent dye DyLight 650 (Thermo-Fisher Scientific, Inc., Waltham, Mass.) using standard techniques.
  • mice were immunized with either CHO cells expressing full-length PD-1 on the cell surface or the PD-1 ECD His protein.
  • female BALB/c mice (7 weeks old) were purchased from Harlan Laboratories, Inc. (Indianapolis, Ind.) and divided into two groups. After six days of acclimatization, one group of animals was immunized with four weekly doses of purified human PD-1 ECD-His at 50 ⁇ g/mouse, as a 1:1 emulsion with TITERMAX GOLDTM (Sigma Aldrich, St. Louis, Mo.). Immunization was carried out subcutaneously around the armpits and inguinal regions.
  • the second group of animals was injected with four weekly doses of CHO-K1 cells stably expressing full length human PD-1 (5 ⁇ 10 6 cells/mouse) subcutaneously around the inguinal regions. After ten days, animals were bled for measurement of the serum titer to PD-1, and one animal from each group was boosted with soluble human PD-1 after a 3-week rest. After three days, spleens, axillary/brachial lymph nodes, and inguinal lymph nodes were collected from each animal. Single cell suspensions of cells from all tissues collected from both animals were pooled and used for generation of hybridomas by cell fusion using standard techniques. Two different myeloma cell lines were used for fusion, F0 (as described in de St.
  • Hybridoma supernatants from ten 96-well plates were screened for binding to a CHO-K1 cell clone stably transfected with a nucleic acid sequence encoding full length human PD-1 and compared to binding to untransfected CHO-K1 cells. Specifically, hybridoma supernatants were diluted 1:1 with PBS/2% FBS and incubated with an equal volume of PD-1 CHO-K1 cells (2.5 ⁇ 10 5 cells in PBS, 2% FBS) for 30 minutes at 4° C.
  • antibodies were captured on a BIACORETM CM5 chip to which GE anti-mouse IgG was coupled.
  • PD-1-His monomer was flowed over the captured antibody using two- or three-fold serial dilutions beginning with 500 nM at the highest concentration.
  • the resulting sensorgrams were fit globally using a 1:1 binding model to calculate on- and off-rates and the subsequent affinities (K D ).
  • results of this example demonstrate a method of producing monoclonal antibodies that bind to human and cynomolgus monkey PD-1 and block PD-1 ligand binding.
  • This example describes the design and generation of CDR-grafted and chimeric anti-PD-1 monoclonal antibodies.
  • PCR amplification of the V L utilized a pool of 9 or 11 degenerate mouse V L forward primers (see Kontermann and Dubel, eds., Antibody Engineering , Springer-Verlag, Berlin (2001)) and a mouse ⁇ constant region reverse primer.
  • PCR amplification of the V H utilized a pool of 12 degenerate mouse V H forward primers (Kontermann and Dubel, supra) and a mouse ⁇ 1 or ⁇ 2a constant region reverse primer (based on isotyping of purified antibody from each clone) with the protocol recommended in the SUPERSCRIPTTM III First-Stand Synthesis System (Life Technologies, Carlsbad, Calif.).
  • PCR products were purified and cloned into pcDNA3.3-TOPO (Life Technologies, Carlsbad, Calif.). Individual colonies from each cell pellet (24 heavy chains and 48 light chains) were selected and sequenced using standard Sanger sequencing methodology (Genewiz, Inc., South Plainfield, N.J.). Variable region sequences were examined and aligned with the closest human heavy chain or light chain V-region germline sequence.
  • Three antibodies were selected for CDR-grafting: (1) 9A2, comprising a V H of SEQ ID NO: 4 and a V L of SEQ ID NO: 28, (2) 10B11, comprising a V H of SEQ ID NO: 15 and a V L of SEQ ID NO: 32, and (3) 6E9, comprising a V H of SEQ ID NO: 22 and a V L of SEQ ID NO: 38.
  • CDR-grafted antibody sequences were designed by grafting CDR residues from each of the above-described mouse antibodies into the closest human germline homologue.
  • CDR-grafted antibody variable regions were synthesized and expressed with human IgG1/ ⁇ constant regions for analysis.
  • mouse:human chimeric antibodies were constructed using the variable regions of the above-described mouse antibodies linked to human IgG1/ ⁇ constant regions. Chimeric and CDR-grafted antibodies were characterized for binding to human and cynomolgus monkey PD-1 antigens and for activity in the PD-1/PD-L1 blocking assay as described above.
  • the functional antagonist activity of chimeric and CDR-grafted antibodies also was tested in a human CD4 + T-cell mixed lymphocyte reaction (MLR) assay in which activation of CD4+ T-cells in the presence of anti-PD-1 antibodies is assessed by measuring IL-2 secretion. Because PD-1 is a negative regulator of T-cell function, antagonism of PD-1 was expected to result in increased T-cell activation as measured by increased IL-2 production.
  • the 9A2, 10B11, and 6E9 CDR-grafted antibodies demonstrated antagonistic activity and were selected for affinity maturation.
  • results of this example demonstrate a method of generating chimeric and CDR-grafted monoclonal antibodies that specifically bind to and inhibit PD-1.
  • This example demonstrates affinity maturation of monoclonal antibodies directed against PD-1.
  • Each antibody was displayed on the surface of HEK 293c18 cells using the SHM-XEL deciduous system (see Bowers et al., Proc. Natl. Acad. Sci. USA, 108: 20455-20460 (2011); and U.S. Patent Application Publication No. 2013/0035472).
  • AID activation-induced cytosine deaminase
  • a panel of six affinity-matured humanized heavy and light chain variable region sequences were paired (denoted APE1922, APE1923, APE1924, APE1950, APE1963 and APE2058) and selected for characterization, and are set forth in Table 1.
  • the PD-1 binding properties of each of these antibody sequences were assayed using surface plasmon resonance (SPR) and solution-based affinity analysis.
  • Antibodies were expressed from HEK 293 cells as human IgG1 antibodies and compared to the reference antibody, a human IgG1 version of BMS-936558, designated BMS.
  • monomeric soluble human PD1-Avi-His was flowed over captured antibody (300 second association, 300 second dissociation) using a three-fold serial dilution series from 500 nM to 2 nM. Captured antibody and antigen were removed between each cycle using 3 M MgCl 2 (60 second contact time) in order to ensure a fresh binding surface for each concentration of antigen. The resulting sensorgrams were fit globally using a 1:1 binding model in order to calculate on- and off-rates (k a and k d , respectively), as well as affinities (K D ).
  • the solution was rotated at room temperature for 2 hours, and beads were pelleted in a picofuge and washed twice with blocking solution (10 mg/mL BSA, 1 M Tris-HCl, pH 8.0). Beads were resuspended in blocking solution (1 mL), rotated at room temperature for 1 hour, and diluted in 25 volumes PBS/0.02% NaN 3 .
  • the secondary antibody was ALEXFLUORTM 647 dye-anti-human IgG (500 ng/mL). Sample antibody concentrations were held constant (50 pM or 75 pM), while antigen PD1-Avi-His was titrated using a three-fold dilutions series from 1 ⁇ M to 17 pM.
  • binding to CHO cells expressing either human or cnyomolgus monkey PD-1 was determined by flow cytometry analysis as described above.
  • blocking of the PD-1/PD-L1 interaction was assessed using DyL650 labeled PD-L1 (mouse IgG1 Fc fusion protein) and PD-1-expressing CHO cells as described above.
  • High binding affinities for cell-surface PD-1 were observed for all tested affinity-matured antibody sequences, with reactivity to cynomolgus monkey PD-1 within a factor of 3-4 fold of human.
  • Blocking of the PD-1/PD-L1 interaction was also efficient with all of the tested affinity-matured antibody sequences, with IC 50 values in the low nM range. These results were consistent with binding affinities assayed both by the BIACORETM and KINEXATM systems as well as cell surface EC 50 values.
  • Thermal stability of the selected antibodies was assessed using a Thermofluor assay as described in McConnell et al., Protein Eng. Des. Sel., 26: 151 (2013). This assay assesses stability through the ability of a hydrophobic fluorescent dye to bind to hydrophobic patches on the protein surface which are exposed as the protein unfolds. The temperature at which 50% of the protein unfolds is determined (Tm) to measure thermal stability. This assay demonstrated that all of the tested affinity-matured antibody sequences had high thermal stability, and all were more stable than the reference antibody. APE2058 was the most stable antibody, exhibiting a Tm more than 10° C. greater than the Tm of the IgG1 version of BMS-936558.
  • results of this example demonstrate a method of generating the inventive immunoglobulin heavy and light chain polypeptides, which exhibit thermostability and high affinity for PD-1.
  • the anti-PD-1 antibody APE2058 was titrated in a dose response in the human CD4+ T-cell MLR assay described above. Antagonism of PD-1 signaling resulted in increased T-cell activation and a corresponding 4- to 5-fold increase in the production of IL-2.
  • an EC 50 value of 20 ng/mL and a concentration 10-fold lower that represents an approximate EC 10 value (2 ng/mL) were selected for combination studies with antagonist antibodies to the TIM-3 or LAG-3 checkpoint molecules.
  • a fully human anti-TIM-3 antibody was characterized in a CD4+ T cell in vitro assay as having antagonist activity as measured by increased IL-2 production in the presence of low levels of anti -CD3 and anti-CD28 antibodies.
  • the anti-TIM-3 antibody demonstrated activity in the MLR assay with an EC 50 value of approximately 0.3 ⁇ g/mL, as shown in FIG. 2 and Table 4, which is approximately 15-fold less activity than the anti-PD-1 APE2058 antibody alone (EC 50 approximately 0.02 ⁇ g/mL).
  • the anti-TIM-3 antagonist antibody stimulated increased amounts of IL-2 production as compared to APE2058 or anti-TIM-3 alone, resulting in a 10-fold decrease in the EC 50 values, as shown in FIG. 2 and Table 4.
  • a fully human antagonist anti-LAG-3 antibody (described in U.S. Patent Application Publication 2011/0150892) has demonstrated potent activity in blocking binding of recombinant soluble LAG-3 to MHC Class II positive cells.
  • This antibody designated herein as APE03109, was evaluated for functional activity in the human CD4 + T-cell MLR assay.
  • APE03109 demonstrated activity in the MLR with an EC 50 value of approximately 0.05 ⁇ g/mL, as shown in FIG. 3 and Table 4, which was similar to the activity of the anti-PD-1 antibody alone.
  • the APE03109 antibody stimulated increased amounts of IL-2 production over APE2058 or APE03109 alone, resulting in a 5-fold decrease in the EC 50 values.
  • an irrelevant human IgG1 antibody was tested in combination with 0.02 ⁇ g/mL anti-PD-1 antibody APE2058. At the highest concentration tested (30 ⁇ g/mL), the APE0422 antibody exhibited no effect on IL-2 production over anti-PD-1 alone.
  • results of this example demonstrate that the inventive PD-1-binding agent combined with antagonistic antibodies directed against TIM-3 or LAG-3 enhances CD4 + T-cell activation in vitro.

Abstract

The invention relates to an isolated immunoglobulin heavy chain polypeptide and an isolated immunoglobulin light chain polypeptide that bind to a programmed death-1 (PD-1) protein. The invention provides a PD-1-binding agent that comprises the aforementioned immunoglobulin heavy chain polypeptide and immunoglobulin light chain polypeptide. The invention also provides related vectors, compositions, and methods of using the PD-1-binding agent to treat a cancer or an infectious disease.

Description

    INCORPORATION-BY-REFERENCE OF MATERIAL SUBMITTED ELECTRONICALLY
  • Incorporated by reference in its entirety herein is a computer-readable nucleotide/amino acid sequence listing submitted concurrently herewith and identified as follows: One 45,217 Byte ASCII (Text) file named “727424_ST25.TXT,” created on Dec. 20, 2016.
  • BACKGROUND OF THE INVENTION
  • Programmed Death 1 (PD-1) (also known as Programmed Cell Death 1) is a type I transmembrane protein of 268 amino acids originally identified by subtractive hybridization of a mouse T cell line undergoing apoptosis (Ishida et al., Embo J., 11: 3887-95 (1992)). PD-1 is a member of the CD28/CTLA-4 family of T-cell regulators, and is expressed on activated T-cells, B-cells, and myeloid lineage cells (Greenwald et al., Annu. Rev. Immunol., 23: 515-548 (2005); and Sharpe et al., Nat. Immunol., 8: 239-245 (2007)).
  • Two ligands for PD-1 have been identified, PD ligand 1 (PD-L1) and PD ligand 2 (PD-L2), both of which belong to the B7 protein superfamily (Greenwald et al., supra). PD-L1 is expressed in a variety of cell types, including cells of the lung, heart, thymus, spleen, and kidney (see, e.g., Freeman et al., J. Exp. Med., 192(7): 1027-1034 (2000); and Yamazaki et al., J. Immunol., 169(10): 5538-5545 (2002)). PD-L1 expression is upregulated on macrophages and dendritic cells (DCs) in response to lipopolysaccharide (LPS) and GM-CSF treatment, and on T-cells and B-cells upon signaling via T-cell and B-cell receptors. PD-L1 also is expressed in a variety of murine tumor cell lines (see, e.g., Iwai et al., Proc. Natl. Acad. Sci. USA, 99(19): 12293-12297 (2002), and Blank et al., Cancer Res., 64(3): 1140-1145 (2004)). In contrast, PD-L2 exhibits a more restricted expression pattern and is expressed primarily by antigen presenting cells (e.g., dendritic cells and macrophages), and some tumor cell lines (see, e.g., Latchman et al., Nat. Immunol., 2(3): 261-238 (2001)). High PD-L1 expression in tumors, whether on the tumor cell, stroma, or other cells within the tumor microenvironment, correlates with poor clinical prognosis, presumably by inhibiting effector T cells and upregulating regulatory T cells (Treg) in the tumor.
  • PD-1 negatively regulates T-cell activation, and this inhibitory function is linked to an immunoreceptor tyrosine-based switch motif (ITSM) in the cytoplasmic domain (see, e.g., Greenwald et al., supra, and Parry et al., Mol. Cell. Biol., 25: 9543-9553 (2005)). PD-1 deficiency can lead to autoimmunity. For example, C57BL/6 PD-1 knockout mice have been shown to develop a lupus-like syndrome (see, e.g., Nishimura et al., Immunity, 11: 141-1151 (1999)). In humans, a single nucleotide polymorphism in the PD-1 gene is associated with higher incidences of systemic lupus erythematosus, type 1 diabetes, rheumatoid arthritis, and progression of multiple sclerosis (see, e.g., Nielsen et al., Tissue Antigens, 62(6): 492-497 (2003); Bertsias et al., Arthritis Rheum., 60(1): 207-218 (2009); Ni et al., Hum. Genet., 121(2): 223-232 (2007); Tahoori et al., Clin. Exp. Rheumatol., 29(5): 763-767 (2011); and Kroner et al., Ann. Neurol., 58(1): 50-57 (2005)). Abnormal PD-1 expression also has been implicated in T-cell dysfunctions in several pathologies, such as tumor immune evasion and chronic viral infections (see, e.g., Barber et al., Nature, 439: 682-687 (2006); and Sharpe et al., supra).
  • Recent studies demonstrate that T-cell suppression induced by PD-1 also plays a role in the suppression of anti-tumor immunity. For example, PD-L1 is expressed on a variety of human and mouse tumors, and binding of PD-1 to PD-L1 on tumors results in T-cell suppression and tumor immune evasion and protection (Dong et al., Nat. Med., 8: 793-800 (2002)). Expression of PD-L1 by tumor cells has been directly associated with their resistance to lysis by anti-tumor T-cells in vitro (Dong et al., supra; and Blank et al., Cancer Res., 64: 1140-1145 (2004)). PD-1 knockout mice are resistant to tumor challenge (Iwai et al., Int. Immunol., 17: 133-144 (2005)), and T-cells from PD-1 knockout mice are highly effective in tumor rejection when adoptively transferred to tumor-bearing mice (Blank et al., supra). Blocking PD-1 inhibitory signals using a monoclonal antibody can potentiate host anti-tumor immunity in mice (Iwai et al., supra; and Hirano et al., Cancer Res., 65: 1089-1096 (2005)), and high levels of PD-L1 expression in tumors are associated with poor prognosis for many human cancer types (Hamanishi et al., Proc. Natl. Acad. Sci. USA, 104: 3360-335 (2007), Brown et al., J. Immunol., 170: 1257-1266 (2003); and Flies et al., Yale Journal of Biology and Medicine, 84(4): 409-421 (2011)).
  • In view of the foregoing, strategies for inhibiting PD-1 activity to treat various types of cancer and for immunopotentiation (e.g., to treat infectious diseases) have been developed (see, e.g., Ascierto et al., Clin. Cancer. Res., 19(5): 1009-1020 (2013)). In this respect, monoclonal antibodies targeting PD-1 have been developed for the treatment of cancer (see, e.g., Weber, Semin. Oncol., 37(5): 430-4309 (2010); and Tang et al., Current Oncology Reports, 15(2): 98-104 (2013)). For example, nivolumab (also known as BMS-936558) produced complete or partial responses in non-small-cell lung cancer, melanoma, and renal-cell cancer in a Phase I clinical trial (see, e.g., Topalian, New England J. Med., 366: 2443-2454 (2012)), and is currently in Phase III clinical trials. MK-3575 is a humanized monoclonal antibody directed against PD-1 that has shown evidence of antitumor activity in Phase I clinical trials (see, e.g., Patnaik et al., 2012 American Society of Clinical Oncology (ASCO) Annual Meeting, Abstract #2512). In addition, recent evidence suggests that therapies which target PD-1 may enhance immune responses against pathogens, such as HIV (see, e.g., Porichis et al., Curr. HIV/AIDS Rep., 9(1): 81-90 (2012)). Despite these advances, however, the efficacy of these potential therapies in humans may be limited.
  • Therefore, there is a need for a PD-1-binding agent (e.g., an antibody) that binds PD-1 with high affinity and effectively neutralizes PD-1 activity. The invention provides such PD-1-binding agents.
  • BRIEF SUMMARY OF THE INVENTION
  • The invention provides an isolated immunoglobulin heavy chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, wherein optionally (a)residue 9 of SEQ ID NO: 1 is replaced with a different amino acid residue, (b) one or more of residues 7, 8, and 9 of SEQ ID NO: 2 is replaced with a different amino acid residue, (c) one or more of residues 1, 2, and 5 of SEQ ID NO: 3 is replaced with a different amino acid residue, or (d) any combination of (a)-(c).
  • The invention provides an isolated immunoglobulin heavy chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, wherein optionally (a) residue 9 of SEQ ID NO: 12 is replaced with a different amino acid residue, (b) residue 8 and/or residue 9 of SEQ ID NO: 13 is replaced with a different amino acid residue, (c) residue 5 of SEQ ID NO: 14 is replaced with a different amino acid residue, or (d) any combination of (a)-(c).
  • The invention provides an isolated immunoglobulin heavy chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 19, a CDR2 amino acid sequence of SEQ ID NO: 20, and a CDR3 amino acid sequence of SEQ ID NO: 21.
  • The invention also provides an isolated immunoglobulin heavy chain polypeptide which comprises an amino acid sequence that is at least 90% identical to any one of SEQ ID NOs: 4-11, SEQ ID NOs: 15-18, and SEQ ID NOs: 22-25.
  • The invention provides an isolated immunoglobulin light chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 26 and a CDR2 amino acid sequence of SEQ ID NO: 27.
  • The invention provides an isolated immunoglobulin light chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31, wherein optionally residue 12 of SEQ ID NO: 30 is replaced with a different amino acid residue.
  • The invention provides an isolated immunoglobulin light chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37, wherein optionally (a) residue 5 of SEQ ID NO: 36 is replaced with a different amino acid residue, and/or (b) residue 4 of SEQ ID NO: 37 is replaced with a different amino acid residue.
  • The invention provides an isolated immunoglobulin light chain polypeptide which comprises an amino acid sequence that is at least 90% identical to SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, or SEQ ID NO: 41.
  • In addition, the invention provides isolated or purified nucleic acid sequences encoding the foregoing immunoglobulin polypeptides, vectors comprising such nucleic acid sequences, isolated PD-1-binding agents comprising the foregoing immunoglobulin polypeptides, nucleic acid sequences encoding such PD-1-binding agents, vectors comprising such nucleic acid sequences, isolated cells comprising such vectors, compositions comprising such PD-1-binding agents or such vectors with a pharmaceutically acceptable carrier, and methods of treating cancer or infectious diseases in mammals by administering effective amounts of such compositions to mammals.
  • BRIEF DESCRIPTION THE DRAWINGS
  • FIG. 1 is a diagram which schematically depicts different PD-1 antigen constructs utilized to generate anti-PD-1 monoclonal antibodies as described in Example 1.
  • FIG. 2 is a graph which illustrates experimental results demonstrating increased activity of an anti-TIM-3 antagonist antibody in a human CD4+ T-cell MLR assay in the presence of low levels of anti-PD-1 antibody APE2058.
  • FIG. 3 is a graph which illustrates experimental results demonstrating increased activity of an anti-LAG-3 antagonist antibody in a human CD4+ T-cell MLR assay in the presence of low levels of anti-PD-1 APE2058.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention provides an isolated immunoglobulin heavy chain polypeptide and/or an isolated immunoglobulin light chain polypeptide, or a fragment (e.g., antigen-binding fragment) thereof. The term “immunoglobulin” or “antibody,” as used herein, refers to a protein that is found in blood or other bodily fluids of vertebrates, which is used by the immune system to identify and neutralize foreign objects, such as bacteria and viruses. The polypeptide is “isolated” in that it is removed from its natural environment. In a preferred embodiment, an immunoglobulin or antibody is a protein that comprises at least one complementarity determining region (CDR). The CDRs form the “hypervariable region” of an antibody, which is responsible for antigen binding (discussed further below). A whole immunoglobulin typically consists of four polypeptides: two identical copies of a heavy (H) chain polypeptide and two identical copies of a light (L) chain polypeptide. Each of the heavy chains contains one N-terminal variable (VH) region and three C-terminal constant (C H1, C H2, and CH3) regions, and each light chain contains one N-terminal variable (VL) region and one C-terminal constant (CL) region. The light chains of antibodies can be assigned to one of two distinct types, either kappa (κ) or lambda (λ), based upon the amino acid sequences of their constant domains. In a typical immunoglobulin, each light chain is linked to a heavy chain by disulphide bonds, and the two heavy chains are linked to each other by disulphide bonds. The light chain variable region is aligned with the variable region of the heavy chain, and the light chain constant region is aligned with the first constant region of the heavy chain. The remaining constant regions of the heavy chains are aligned with each other.
  • The variable regions of each pair of light and heavy chains form the antigen binding site of an antibody. The VH and VL regions have the same general structure, with each region comprising four framework (FW or FR) regions. The term “framework region,” as used herein, refers to the relatively conserved amino acid sequences within the variable region which are located between the hypervariable or complementary determining regions (CDRs). There are four framework regions in each variable domain, which are designated FR1, FR2, FR3, and FR4. The framework regions form the β sheets that provide the structural framework of the variable region (see, e.g., C. A. Janeway et al. (eds.), Immunobiology, 5th Ed., Garland Publishing, New York, N.Y. (2001)).
  • The framework regions are connected by three complementarity determining regions (CDRs). As discussed, above, the three CDRs, known as CDR1, CDR2, and CDR3, form the “hypervariable region” of an antibody, which is responsible for antigen binding. The CDRs form loops connecting, and in some cases comprising part of, the beta-sheet structure formed by the framework regions. While the constant regions of the light and heavy chains are not directly involved in binding of the antibody to an antigen, the constant regions can influence the orientation of the variable regions. The constant regions also exhibit various effector functions, such as participation in antibody-dependent complement-mediated lysis or antibody-dependent cellular toxicity via interactions with effector molecules and cells.
  • The isolated immunoglobulin heavy chain polypeptide and the isolated immunoglobulin light chain polypeptide of the invention desirably bind to PD-1. As discussed above, programmed death 1 (PD-1) (also known as programmed cell death 1) is a 268 amino acid type I transmembrane protein (Ishida et al., supra). PD-1 is a member of the CD28/CTLA-4 family of T-cell regulators and is expressed on activated T-cells, B-cells, and myeloid lineage cells (Greenwald et al., supra; and Sharpe et al., supra). PD-1 includes an extracellular IgV domain followed by short extracellular stalk, a transmembrane region and an intracellular tail. The intracellular tail contains two phosphorylation sites located in an immunoreceptor tyrosine-based inhibitory motif and an immunoreceptor tyrosine-based switch motif, which play a role in the ability of PD-1 to negatively regulate T-cell receptor signaling (see, e.g., Ishida et al., supra; and Blank et al., supra). The inventive isolated immunoglobulin heavy chain polypeptide and the inventive isolated immunoglobulin light chain polypeptide can form an agent that binds to PD-1 and another antigen, resulting in a “dual reactive” binding agent (e.g., a dual reactive antibody). For example, the agent can bind to PD-1 and to another negative regulator of the immune system such as, for example, lymphocyte-activation gene 3 (LAG-3) and/or T-cell immunoglobulin domain and mucin domain 3 protein (TIM-3).
  • Antibodies which bind to PD-1, and components thereof, are known in the art (see, e.g., U.S. Pat. No. 8,168,757; Topalian et al., supra; and Patnaik et al., supra). Anti-PD-1 antibodies also are commercially available from sources such as, for example, Abcam (Cambridge, Mass.).
  • An amino acid “replacement” or “substitution” refers to the replacement of one amino acid at a given position or residue by another amino acid at the same position or residue within a polypeptide sequence.
  • Amino acids are broadly grouped as “aromatic” or “aliphatic.” An aromatic amino acid includes an aromatic ring. Examples of “aromatic” amino acids include histidine (H or His), phenylalanine (F or Phe), tyrosine (Y or Tyr), and tryptophan (W or Trp). Non-aromatic amino acids are broadly grouped as “aliphatic.” Examples of “aliphatic” amino acids include glycine (G or Gly), alanine (A or Ala), valine (V or Val), leucine (L or Leu), isoleucine (I or Ile), methionine (M or Met), serine (S or Ser), threonine (T or Thr), cysteine (C or Cys), proline (P or Pro), glutamic acid (E or Glu), aspartic acid (A or Asp), asparagine (N or Asn), glutamine (Q or Gln), lysine (K or Lys), and arginine (R or Arg).
  • Aliphatic amino acids may be sub-divided into four sub-groups. The “large aliphatic non-polar sub-group” consists of valine, leucine, and isoleucine. The “aliphatic, slightly-polar sub-group” consists of methionine, serine, threonine, and cysteine. The “aliphatic polar/charged sub-group” consists of glutamic acid, aspartic acid, asparagine, glutamine, lysine, and arginine. The “small-residue sub-group” consists of glycine and alanine. The group of charged/polar amino acids may be sub-divided into three sub-groups: the “positively-charged sub-group” consisting of lysine and arginine, the “negatively-charged sub-group” consisting of glutamic acid and aspartic acid, and the “polar sub-group” consisting of asparagine and glutamine.
  • Aromatic amino acids may be sub-divided into two sub-groups: the “nitrogen ring sub-group” consisting of histidine and tryptophan and the “phenyl sub-group” consisting of phenylalanine and tyrosine.
  • The amino acid replacement or substitution can be conservative, semi-conservative, or non-conservative. The phrase “conservative amino acid substitution” or “conservative mutation” refers to the replacement of one amino acid by another amino acid with a common property. A functional way to define common properties between individual amino acids is to analyze the normalized frequencies of amino acid changes between corresponding proteins of homologous organisms (Schulz and Schirmer, Principles of Protein Structure, Springer-Verlag, New York (1979)). According to such analyses, groups of amino acids may be defined where amino acids within a group exchange preferentially with each other, and therefore resemble each other most in their impact on the overall protein structure (Schulz and Schirmer, supra).
  • Examples of conservative amino acid substitutions include substitutions of amino acids within the sub-groups described above, for example, lysine for arginine and vice versa such that a positive charge may be maintained, glutamic acid for aspartic acid and vice versa such that a negative charge may be maintained, serine for threonine such that a free —OH can be maintained, and glutamine for asparagine such that a free —NH2 can be maintained.
  • “Semi-conservative mutations” include amino acid substitutions of amino acids within the same groups listed above, but not within the same sub-group. For example, the substitution of aspartic acid for asparagine, or asparagine for lysine, involves amino acids within the same group, but different sub-groups. “Non-conservative mutations” involve amino acid substitutions between different groups, for example, lysine for tryptophan, or phenylalanine for serine, etc.
  • The invention provides an immunoglobulin heavy chain polypeptide that comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3. In one embodiment of the invention, the isolated immunoglobulin heavy chain polypeptide comprises, consists of, or consists essentially of a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, wherein optionally (a) residue 9 of SEQ ID NO: 1 is replaced with a different amino acid residue, (b) one or more of residues 7, 8, and 9 of SEQ ID NO: 2 is replaced with a different amino acid residue, (c) one or more of residues 1, 2, and 5 of SEQ ID NO: 3 is replaced with a different amino acid residue, or (d) any combination of (a)-(c). When the inventive immunoglobulin heavy chain polypeptide consists essentially of a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3 and optional amino acid replacements, additional components can be included in the polypeptide that do not materially affect the polypeptide (e.g., protein moieties such as biotin that facilitate purification or isolation). When the inventive immunoglobulin heavy chain polypeptide consists of a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3 and optional amino acid replacements, the polypeptide does not comprise any additional components (i.e., components that are not endogenous to the inventive immunoglobulin heavy chain polypeptide).
  • In one embodiment of the invention, the isolated immunoglobulin polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that (a) residue 9 of SEQ ID NO: 1 is replaced with a different amino acid residue, (b) one or more of residues 7, 8, and 9 of SEQ ID NO: 2 is replaced with a different amino acid residue, (c) one or more of residues 1, 2, and 5 of SEQ ID NO: 3 is replaced with a different amino acid residue, or (d) any combination of (a)-(c). For example, the isolated immunoglobulin heavy chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that residue 9 of SEQ ID NO: 1 is replaced with a different amino acid residue and one or more of residues 7, 8, and 9 of SEQ ID NO: 2 is replaced with a different amino acid residue. Alternatively, the isolated immunoglobulin heavy chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that residue 9 of SEQ ID NO: 1 is replaced with a different amino acid residue, one or more of residues 7, 8, and 9 of SEQ ID NO: 2 is replaced with a different amino acid residue, and one or more of residues 1, 2, and 5 of SEQ ID NO: 3 is replaced with a different amino acid residue. In another embodiment, the isolated immunoglobulin heavy chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that one or more of residues 1, 2, and 5 of SEQ ID NO: 3 is replaced with a different amino acid residue. Each of residue 9 of SEQ ID NO: 1, residues 7, 8, and 9 of SEQ ID NO: 2, and residues 1, 2, and 5 of SEQ ID NO: 3 can be replaced with any suitable amino acid residue that can be the same or different in each position. For example, the amino acid residue of a first position can be replaced with a first different amino acid residue, and the amino acid residue of a second position can be replaced with a second different amino acid residue, wherein the first and second different amino acid residues are the same or different.
  • In one embodiment, the isolated immunoglobulin heavy chain polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that residue 9 of SEQ ID NO: 1 is replaced with a methionine (M) residue. In another embodiment, the isolated immunoglobulin heavy chain polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that (a) residue 7 of SEQ ID NO: 2 is replaced with an asparagine (N) residue, (b) residue 8 of SEQ ID NO: 2 is replaced with a serine (S) residue, (c) residue 9 of SEQ ID NO: 2 is replaced with a threonine (T) residue, or (d) any combination of (a)-(c). In another embodiment, the isolated immunoglobulin heavy chain polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that (a) residue 1 of SEQ ID NO: 3 is replaced with a glutamic acid (E) residue, (b) residue 2 of SEQ ID NO: 3 is replaced with a tyrosine (Y) residue, (c) residue 5 of SEQ ID NO: 3 is replaced with a serine (S) residue, or (d) any combination of (a)-(c).
  • Exemplary immunoglobulin heavy chain polypeptides as described above can comprise any one of the following amino acid sequences: SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, or SEQ ID NO: 11.
  • The invention provides an isolated immunoglobulin heavy chain polypeptide comprises, consists essentially of, or consists of a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, wherein optionally (a) residue 9 of SEQ ID NO: 12 is replaced with a different amino acid residue, (b) residue 8 and/or residue 9 of SEQ ID NO: 13 is replaced with a different amino acid residue, (c) residue 5 of SEQ ID NO: 14 is replaced with a different amino acid residue, or (d) any combination of (a)-(c). When the inventive immunoglobulin heavy chain polypeptide consists essentially of a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14 and optional amino acid replacements, additional components can be included in the polypeptide that do not materially affect the polypeptide (e.g., protein moieties such as biotin that facilitate purification or isolation). When the inventive immunoglobulin heavy chain polypeptide consists of a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14 and optional amino acid replacements, the polypeptide does not comprise any additional components (i.e., components that are not endogenous to the inventive immunoglobulin heavy chain polypeptide).
  • In one embodiment, the isolated immunoglobulin heavy chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that (a) residue 9 of SEQ ID NO: 12 is replaced with a different amino acid residue, (b) residue 8 and/or residue 9 of SEQ ID NO: 13 is replaced with a different amino acid residue, (c) residue 5 of SEQ ID NO: 14 is replaced with a different amino acid residue, or (d) any combination of (a)-(c). For example, the isolated immunoglobulin heavy chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that residue 9 of SEQ ID NO: 12 is replaced with a different amino acid residue, residue 8 of SEQ ID NO: 13, and residue 9 of SEQ ID NO: 13 is replaced with a different amino acid residue. Alternatively, the isolated immunoglobulin heavy chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that residue 9 of SEQ ID NO: 12 is replaced with a different amino acid residue and residue 5 of SEQ ID NO: 14 is replaced with a different amino acid residue. In another embodiment, the isolated immunoglobulin heavy chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that residue 9 of SEQ ID NO: 12 is replaced with a different amino acid residue, residue 8 of SEQ ID NO: 13 is replaced with a different amino acid residue, residue 9 of SEQ ID NO: 13 is replaced with a different amino acid residue, and residue 5 of SEQ ID NO: 14 is replaced with a different amino acid residue. Each of residue 9 of SEQ ID NO: 12, residues 8 and 9 of SEQ ID NO: 13, and residue 5 of SEQ ID NO: 14 can be replaced with any suitable amino acid residue that can be the same or different in each position. For example, the amino acid residue of a first position can be replaced with a first different amino acid residue, and the amino acid residue of a second position can be replaced with a second different amino acid residue, wherein the first and second different amino acid residues are the same or different. In one embodiment, the isolated immunoglobulin heavy chain polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that residue 9 of SEQ ID NO: 12 is replaced with a leucine (L) residue. In another embodiment, the isolated immunoglobulin heavy chain polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that (a) residue 8 of SEQ ID NO: 13 is replaced with a tyrosine (Y) residue, and/or (b) residue 9 of SEQ ID NO: 13 is replaced with an alanine (A) residue. In another embodiment, the isolated immunoglobulin heavy chain polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that residue 5 of SEQ ID NO: 14 is replaced with a threonine (T) residue.
  • Exemplary immunoglobulin heavy chain polypeptides as described above can comprise any one of the following amino acid sequences: SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, or SEQ ID NO: 18.
  • The invention provides an isolated immunoglobulin heavy chain polypeptide comprises, consists essentially of, or consists of a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 19, a CDR2 amino acid sequence of SEQ ID NO: 20, and a CDR3 amino acid sequence of SEQ ID NO: 21. When the inventive immunoglobulin heavy chain polypeptide consists essentially of a CDR1 amino acid sequence of SEQ ID NO: 19, a CDR2 amino acid sequence of SEQ ID NO: 20, and a CDR3 amino acid sequence of SEQ ID NO: 21, additional components can be included in the polypeptide that do not materially affect the polypeptide (e.g., protein moieties such as biotin that facilitate purification or isolation). When the inventive immunoglobulin heavy chain polypeptide consists of a CDR1 amino acid sequence of SEQ ID NO: 19, a CDR2 amino acid sequence of SEQ ID NO: 20, and a CDR3 amino acid sequence of SEQ ID NO: 21, the polypeptide does not comprise any additional components (i.e., components that are not endogenous to the inventive immunoglobulin heavy chain polypeptide). Exemplary immunoglobulin heavy chain polypeptides as described above can comprise any one of the following amino acid sequences: SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, or SEQ ID NO: 25.
  • In addition, one or more amino acids can be inserted into the aforementioned immunoglobulin heavy chain polypeptides. Any number of any suitable amino acids can be inserted into the amino acid sequence of the immunoglobulin heavy chain polypeptide. In this respect, at least one amino acid (e.g., 2 or more, 5 or more, or 10 or more amino acids), but not more than 20 amino acids (e.g., 18 or less, 15 or less, or 12 or less amino acids), can be inserted into the amino acid sequence of the immunoglobulin heavy chain polypeptide. Preferably, 1-10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) are inserted into the amino acid sequence of the immunoglobulin heavy chain polypeptide. In this respect, the amino acid(s) can be inserted into any one of the aforementioned immunoglobulin heavy chain polypeptides in any suitable location. Preferably, the amino acid(s) are inserted into a CDR (e.g., CDR1, CDR2, or CDR3) of the immunoglobulin heavy chain polypeptide.
  • The invention provides an isolated immunoglobulin heavy chain polypeptide which comprises an amino acid sequence that is at least 90% identical (e.g., at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical) to any one of SEQ ID NOs: 4-11, SEQ ID NOs: 15-18, and SEQ ID NOs: 22-25. Nucleic acid or amino acid sequence “identity,” as described herein, can be determined by comparing a nucleic acid or amino acid sequence of interest to a reference nucleic acid or amino acid sequence. The percent identity is the number of nucleotides or amino acid residues that are the same (i.e., that are identical) as between the sequence of interest and the reference sequence divided by the length of the longest sequence (i.e., the length of either the sequence of interest or the reference sequence, whichever is longer). A number of mathematical algorithms for obtaining the optimal alignment and calculating identity between two or more sequences are known and incorporated into a number of available software programs. Examples of such programs include CLUSTAL-W, T-Coffee, and ALIGN (for alignment of nucleic acid and amino acid sequences), BLAST programs (e.g., BLAST 2.1, BL2SEQ, and later versions thereof) and FASTA programs (e.g., FASTA3x, FASTM, and SSEARCH) (for sequence alignment and sequence similarity searches). Sequence alignment algorithms also are disclosed in, for example, Altschul et al., J. Molecular Biol., 215(3): 403-410 (1990), Beigert et al., Proc. Natl. Acad. Sci. USA, 106(10): 3770-3775 (2009), Durbin et al., eds., Biological Sequence Analysis: Probalistic Models of Proteins and Nucleic Acids, Cambridge University Press, Cambridge, UK (2009), Soding, Bioinformatics, 21(7): 951-960 (2005), Altschul et al., Nucleic Acids Res., 25(17): 3389-3402 (1997), and Gusfield, Algorithms on Strings, Trees and Sequences, Cambridge University Press, Cambridge UK (1997)).
  • The invention provides an immunoglobulin light chain polypeptide that comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 26 and a CDR2 amino acid sequence of SEQ ID NO: 27. In one embodiment of the invention, the isolated immunoglobulin light chain polypeptide comprises, consists essentially of, or consists of a CDR1 amino acid sequence of SEQ ID NO: 26 and a CDR2 amino acid sequence of SEQ ID NO: 27. When the inventive immunoglobulin light chain polypeptide consists essentially of a CDR1 amino acid sequence of SEQ ID NO: 26 and a CDR2 amino acid sequence of SEQ ID NO: 27, additional components can be included in the polypeptide that do not materially affect the polypeptide (e.g., protein moieties such as biotin that facilitate purification or isolation). When the inventive immunoglobulin light chain polypeptide consists of a CDR1 amino acid sequence of SEQ ID NO: 26 and a CDR2 amino acid sequence of SEQ ID NO: 27, the polypeptide does not comprise any additional components (i.e., components that are not endogenous to the inventive immunoglobulin light chain polypeptide). Exemplary immunoglobulin light chain polypeptides as described above can comprise SEQ ID NO: 28 or SEQ ID NO: 29.
  • The invention provides an isolated immunoglobulin light chain polypeptide comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31. In one embodiment of the invention, the isolated immunoglobulin light chain polypeptide comprises, consists of, or consists essentially of a CDR1 amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31, wherein optionally residue 12 of SEQ ID NO: 30 is replaced with a different amino acid residue. When the inventive immunoglobulin light chain polypeptide consists essentially of a CDR1 amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31 and optional amino acid replacements, additional components can be included in the polypeptide that do not materially affect the polypeptide (e.g., protein moieties such as biotin that facilitate purification or isolation). When the inventive immunoglobulin light chain polypeptide consists of a CDR1 amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31 and optional amino acid replacements, the polypeptide does not comprise any additional components (i.e., components that are not endogenous to the inventive immunoglobulin light chain polypeptide).
  • In this respect, for example, the isolated immunoglobulin light chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31, except that residue 12 of SEQ ID NO: 30 is replaced with a different amino acid residue. Residue 12 of SEQ ID NO: 30 can be replaced with any suitable amino acid residue. In one embodiment, the isolated immunoglobulin light chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31, except that residue 12 of SEQ ID NO: 30 is replaced with a threonine (T) residue. Exemplary immunoglobulin light chain polypeptides as described above can comprise any one of the following amino acid sequences: SEQ ID NO: 32, SEQ ID NO: 33, or SEQ ID NO: 34.
  • The invention provides an isolated immunoglobulin light chain polypeptide comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37. In one embodiment, the immunoglobulin light chain polypeptide comprises, consists essentially of, or consists of a CDR1 amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37, wherein optionally (a) residue 5 of SEQ ID NO: 36 is replaced with a different amino acid residue, and/or (b) residue 4 of SEQ ID NO: 37 is replaced with a different amino acid residue. When the inventive immunoglobulin light chain polypeptide consists essentially of a CDR1 amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37 and optional amino acid replacements, additional components can be included in the polypeptide that do not materially affect the polypeptide (e.g., protein moieties such as biotin that facilitate purification or isolation). When the inventive immunoglobulin light chain polypeptide consists of a CDR1 amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37 and optional amino acid replacements, the polypeptide does not comprise any additional components (i.e., components that are not endogenous to the inventive immunoglobulin light chain polypeptide). In this respect, for example, the isolated immunoglobulin light chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37. Alternatively, the isolated immunoglobulin light chain polypeptide can comprise a CDR1 amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37, except that (a) residue 5 of SEQ ID NO: 36 is replaced with a different amino acid residue, and/or (b) residue 4 of SEQ ID NO: 37 is replaced with a different amino acid residue. Each of residue 5 of SEQ ID NO: 36 and residue 4 of SEQ ID NO: 37 can be replaced with any suitable amino acid residue that can be the same or different in each position. For example, the amino acid residue of a first position can be replaced with a first different amino acid residue, and the amino acid residue of a second position can be replaced with a second different amino acid residue, wherein the first and second different amino acid residues are the same or different.
  • In one embodiment, the isolated immunoglobulin light chain polypeptide comprises a CDR1 amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37, except that (a) residue 5 of SEQ ID NO: 36 is replaced with a leucine (L) residue, and/or (b) residue 4 of SEQ ID NO: 37 is replaced with an asparagine (N) residue. Exemplary immunoglobulin light chain polypeptides as described above can comprise any one of the following amino acid sequences: SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, or SEQ ID NO: 41.
  • In addition, one or more amino acids can be inserted into the aforementioned immunoglobulin light chain polypeptides. Any number of any suitable amino acids can be inserted into the amino acid sequence of the immunoglobulin light chain polypeptide. In this respect, at least one amino acid (e.g., 2 or more, 5 or more, or 10 or more amino acids), but not more than 20 amino acids (e.g., 18 or less, 15 or less, or 12 or less amino acids), can be inserted into the amino acid sequence of the immunoglobulin light chain polypeptide. Preferably, 1-10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) are inserted in to the amino acid sequence of the immunoglobulin light chain polypeptide. In this respect, the amino acid(s) can be inserted into any one of the aforementioned immunoglobulin light chain polypeptides in any suitable location. Preferably, the amino acid(s) are inserted into a CDR (e.g., CDR1, CDR2, or CDR3) of the immunoglobulin light chain polypeptide.
  • The invention provides an isolated immunoglobulin light chain polypeptide which comprises an amino acid sequence that is at least 90% identical (e.g., at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical) to any one of SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, and SEQ ID NO: 41. Nucleic acid or amino acid sequence “identity,” as described herein, can be determined using the methods described herein.
  • The invention provides an isolated programmed death 1 (PD-1)-binding agent comprising, consisting essentially of, or consisting of the inventive isolated amino acid sequences described herein. By “programmed death 1 (PD-1)-binding agent” is meant a molecule, preferably a proteinaceous molecule, that binds specifically to the programmed death 1 protein (PD-1). Preferably, the PD-1-binding agent is an antibody or a fragment (e.g., immunogenic fragment) thereof. The isolated PD-1-binding agent of the invention comprises, consists essentially of, or consists of the inventive isolated immunoglobulin heavy chain polypeptide and/or the inventive isolated immunoglobulin light chain polypeptide. In one embodiment, the isolated PD-1-binding agent comprises, consists essentially of, or consists of the inventive immunoglobulin heavy chain polypeptide or the inventive immunoglobulin light chain polypeptide. In another embodiment, the isolated PD-1-binding agent comprises, consists essentially of, or consists of the inventive immunoglobulin heavy chain polypeptide and the inventive immunoglobulin light chain polypeptide.
  • The invention is not limited to an isolated PD-1-binding agent that comprises, consists essentially of, or consists of an immunoglobulin heavy chain polypeptide and/or light chain polypeptide having replacements, insertions, and/or deletions of the specific amino acid residues disclosed herein. Indeed, any amino acid residue of the inventive immunoglobulin heavy chain polypeptide and/or the inventive immunoglobulin light chain polypeptide can be replaced, in any combination, with a different amino acid residue, or can be deleted or inserted, so long as the biological activity of the PD-1-binding agent is enhanced or improved as a result of the amino acid replacements, insertions, and/or deletions. The “biological activity” of an PD-1-binding agent refers to, for example, binding affinity for PD-1 or a particular PD-1 epitope, neutralization or inhibition of PD-1 protein binding to its ligands PD-L1 and PD-L1, neutralization or inhibition of PD-1 protein activity in vivo (e.g., IC50), pharmacokinetics, and cross-reactivity (e.g., with non-human homologs or orthologs of the PD-1 protein, or with other proteins or tissues). Other biological properties or characteristics of an antigen-binding agent recognized in the art include, for example, avidity, selectivity, solubility, folding, immunotoxicity, expression, and formulation. The aforementioned properties or characteristics can be observed, measured, and/or assessed using standard techniques including, but not limited to, ELISA, competitive ELISA, surface plasmon resonance analysis (BIACORE™), or KINEXA™, in vitro or in vivo neutralization assays, receptor-ligand binding assays, cytokine or growth factor production and/or secretion assays, and signal transduction and immunohistochemistry assays.
  • The terms “inhibit” or “neutralize,” as used herein with respect to the activity of a PD-1-binding agent, refer to the ability to substantially antagonize, prohibit, prevent, restrain, slow, disrupt, alter, eliminate, stop, or reverse the progression or severity of, for example, the biological activity of a PD-1 protein, or a disease or condition associated with an PD-1 protein. The isolated PD-1-binding agent of the invention preferably inhibits or neutralizes the activity of a PD-1 protein by at least about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 100%, or a range defined by any two of the foregoing values.
  • The isolated PD-1-binding agent of the invention can be a whole antibody, as described herein, or an antibody fragment. The terms “fragment of an antibody,” “antibody fragment,” and “functional fragment of an antibody” are used interchangeably herein to mean one or more fragments of an antibody that retain the ability to specifically bind to an antigen (see, generally, Holliger et al., Nat. Biotech., 23(9): 1126-1129 (2005)). The isolated PD-1 binding agent can contain any PD-1-binding antibody fragment. The antibody fragment desirably comprises, for example, one or more CDRs, the variable region (or portions thereof), the constant region (or portions thereof), or combinations thereof. Examples of antibody fragments include, but are not limited to, (i) a Fab fragment, which is a monovalent fragment consisting of the VL, VH, CL, and CH1 domains, (ii) a F(ab′)2 fragment, which is a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region, (iii) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (iv) a Fab′ fragment, which results from breaking the disulfide bridge of an F(ab′)2 fragment using mild reducing conditions, (v) a disulfide-stabilized Fv fragment (dsFv), and (vi) a domain antibody (dAb), which is an antibody single variable region domain (VH or VL) polypeptide that specifically binds antigen.
  • In embodiments where the isolated PD-1-binding agent comprises a fragment of the immunoglobulin heavy chain or light chain polypeptide, the fragment can be of any size so long as the fragment binds to, and preferably inhibits the activity of, a PD-1 protein. In this respect, a fragment of the immunoglobulin heavy chain polypeptide desirably comprises between about 5 and 18 (e.g., about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or a range defined by any two of the foregoing values) amino acids. Similarly, a fragment of the immunoglobulin light chain polypeptide desirably comprises between about 5 and 18 (e.g., about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or a range defined by any two of the foregoing values) amino acids.
  • When the PD-1-binding agent is an antibody or antibody fragment, the antibody or antibody fragment desirably comprises a heavy chain constant region (Fc) of any suitable class. Preferably, the antibody or antibody fragment comprises a heavy chain constant region that is based upon wild-type IgG1, IgG2, or IgG4 antibodies, or variants thereof.
  • The PD-1-binding agent also can be a single chain antibody fragment. Examples of single chain antibody fragments include, but are not limited to, (i) a single chain Fv (scFv), which is a monovalent molecule consisting of the two domains of the Fv fragment (i.e., VL and VH) joined by a synthetic linker which enables the two domains to be synthesized as a single polypeptide chain (see, e.g., Bird et al., Science, 242: 423-426 (1988); Huston et al., Proc. Natl. Acad. Sci. USA, 85: 5879-5883 (1988); and Osbourn et al., Nat. Biotechnol., 16: 778 (1998)) and (ii) a diabody, which is a dimer of polypeptide chains, wherein each polypeptide chain comprises a VH connected to a VL by a peptide linker that is too short to allow pairing between the VH and VL on the same polypeptide chain, thereby driving the pairing between the complementary domains on different VH-VL polypeptide chains to generate a dimeric molecule having two functional antigen binding sites. Antibody fragments are known in the art and are described in more detail in, e.g., U.S. Patent Application Publication 2009/0093024 A1.
  • The isolated PD-1-binding agent also can be an intrabody or fragment thereof. An intrabody is an antibody which is expressed and which functions intracellularly. Intrabodies typically lack disulfide bonds and are capable of modulating the expression or activity of target genes through their specific binding activity. Intrabodies include single domain fragments such as isolated VH and VL domains and scFvs. An intrabody can include sub-cellular trafficking signals attached to the N or C terminus of the intrabody to allow expression at high concentrations in the sub-cellular compartments where a target protein is located. Upon interaction with a target gene, an intrabody modulates target protein function and/or achieves phenotypic/functional knockout by mechanisms such as accelerating target protein degradation and sequestering the target protein in a non-physiological sub-cellular compartment. Other mechanisms of intrabody-mediated gene inactivation can depend on the epitope to which the intrabody is directed, such as binding to the catalytic site on a target protein or to epitopes that are involved in protein-protein, protein-DNA, or protein-RNA interactions.
  • The isolated PD-1-binding agent also can be an antibody conjugate. In this respect, the isolated PD-1-binding agent can be a conjugate of (1) an antibody, an alternative scaffold, or fragments thereof, and (2) a protein or non-protein moiety comprising the PD-1-binding agent. For example, the PD-1-binding agent can be all or part of an antibody conjugated to a peptide, a fluorescent molecule, or a chemotherapeutic agent.
  • The isolated PD-1-binding agent can be, or can be obtained from, a human antibody, a non-human antibody, or a chimeric antibody. By “chimeric” is meant an antibody or fragment thereof comprising both human and non-human regions. Preferably, the isolated PD-1-binding agent is a humanized antibody. A “humanized” antibody is a monoclonal antibody comprising a human antibody scaffold and at least one CDR obtained or derived from a non-human antibody. Non-human antibodies include antibodies isolated from any non-human animal, such as, for example, a rodent (e.g., a mouse or rat). A humanized antibody can comprise, one, two, or three CDRs obtained or derived from a non-human antibody. In a preferred embodiment of the invention, CDRH3 of the inventive PD-1-binding agent is obtained or derived from a mouse monoclonal antibody, while the remaining variable regions and constant region of the inventive PD-1-binding agent are obtained or derived from a human monoclonal antibody.
  • A human antibody, a non-human antibody, a chimeric antibody, or a humanized antibody can be obtained by any means, including via in vitro sources (e.g., a hybridoma or a cell line producing an antibody recombinantly) and in vivo sources (e.g., rodents). Methods for generating antibodies are known in the art and are described in, for example, Köhler and Milstein, Eur. J. Immunol., 5: 511-519 (1976); Harlow and Lane (eds.), Antibodies: A Laboratory Manual, CSH Press (1988); and Janeway et al. (eds.), Immunobiology, 5th Ed., Garland Publishing, New York, N.Y. (2001)). In certain embodiments, a human antibody or a chimeric antibody can be generated using a transgenic animal (e.g., a mouse) wherein one or more endogenous immunoglobulin genes are replaced with one or more human immunoglobulin genes. Examples of transgenic mice wherein endogenous antibody genes are effectively replaced with human antibody genes include, but are not limited to, the Medarex HUMAB-MOUSE™, the Kirin TC MOUSE™, and the Kyowa Kirin KM-MOUSE™ (see, e.g., Lonberg, Nat. Biotechnol., 23(9): 1117-25 (2005), and Lonberg, Handb. Exp. Pharmacol., 181: 69-97 (2008)). A humanized antibody can be generated using any suitable method known in the art (see, e.g., An, Z. (ed.), Therapeutic Monoclonal Antibodies: From Bench to Clinic, John Wiley & Sons, Inc. Hoboken, N.J. (2009)), including, e.g., grafting of non-human CDRs onto a human antibody scaffold (see, e.g., Kashmiri et al., Methods, 36(1): 25-34 (2005); and Hou et al., J. Biochem., 144(1): 115-120 (2008)). In one embodiment, a humanized antibody can be produced using the methods described in, e.g., U.S. Patent Application Publication 2011/0287485 A1.
  • In a preferred embodiment, the PD-1-binding agent binds an epitope of a PD-1 protein which blocks the binding of PD-1 to PD-L1. The invention also provides an isolated or purified epitope of a PD-1 protein which blocks the binding of PD-1 to PD-L1 in an indirect or allosteric manner.
  • The invention also provides one or more isolated or purified nucleic acid sequences that encode the inventive immunoglobulin heavy chain polypeptide, the inventive immunoglobulin light chain polypeptide, and the inventive PD-1-binding agent.
  • The term “nucleic acid sequence” is intended to encompass a polymer of DNA or RNA, i.e., a polynucleotide, which can be single-stranded or double-stranded and which can contain non-natural or altered nucleotides. The terms “nucleic acid” and “polynucleotide” as used herein refer to a polymeric form of nucleotides of any length, either ribonucleotides (RNA) or deoxyribonucleotides (DNA). These terms refer to the primary structure of the molecule, and thus include double- and single-stranded DNA, and double- and single-stranded RNA. The terms include, as equivalents, analogs of either RNA or DNA made from nucleotide analogs and modified polynucleotides such as, though not limited to, methylated and/or capped polynucleotides. Nucleic acids are typically linked via phosphate bonds to form nucleic acid sequences or polynucleotides, though many other linkages are known in the art (e.g., phosphorothioates, boranophosphates, and the like). Nucleic acid sequences encoding the inventive immunoglobulin heavy chain polypeptides include, for example, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, and SEQ ID NO: 55. Nucleic acid sequences encoding the inventive immunoglobulin light chain polypeptides include, for example, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 62, SEQ ID NO: 63, and SEQ ID NO: 64.
  • The invention further provides a vector comprising one or more nucleic acid sequences encoding the inventive immunoglobulin heavy chain polypeptide, the inventive immunoglobulin light chain polypeptide, and/or the inventive PD-1-binding agent. The vector can be, for example, a plasmid, episome, cosmid, viral vector (e.g., retroviral or adenoviral), or phage. Suitable vectors and methods of vector preparation are well known in the art (see, e.g., Sambrook et al., Molecular Cloning, a Laboratory Manual, 3rd edition, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (2001), and Ausubel et al., Current Protocols in Molecular Biology, Greene Publishing Associates and John Wiley & Sons, New York, N.Y. (1994)).
  • In addition to the nucleic acid sequence encoding the inventive immunoglobulin heavy polypeptide, the inventive immunoglobulin light chain polypeptide, and/or the inventive PD-1-binding agent, the vector preferably comprises expression control sequences, such as promoters, enhancers, polyadenylation signals, transcription terminators, internal ribosome entry sites (IRES), and the like, that provide for the expression of the coding sequence in a host cell. Exemplary expression control sequences are known in the art and described in, for example, Goeddel, Gene Expression Technology: Methods in Enzymology, Vol. 185, Academic Press, San Diego, Calif. (1990).
  • A large number of promoters, including constitutive, inducible, and repressible promoters, from a variety of different sources are well known in the art. Representative sources of promoters include for example, virus, mammal, insect, plant, yeast, and bacteria, and suitable promoters from these sources are readily available, or can be made synthetically, based on sequences publicly available, for example, from depositories such as the ATCC as well as other commercial or individual sources. Promoters can be unidirectional (i.e., initiate transcription in one direction) or bi-directional (i.e., initiate transcription in either a 3′ or 5′ direction). Non-limiting examples of promoters include, for example, the T7 bacterial expression system, pBAD (araA) bacterial expression system, the cytomegalovirus (CMV) promoter, the SV40 promoter, the RSV promoter. Inducible promoters include, for example, the Tet system (U.S. Pat. Nos. 5,464,758 and 5,814,618), the Ecdysone inducible system (No et al., Proc. Natl. Acad. Sci., 93: 3346-3351 (1996)), the T-REX™ system (Invitrogen, Carlsbad, Calif.), LACSWITCH™ system (Stratagene, San Diego, Calif.), and the Cre-ERT tamoxifen inducible recombinase system (Indra et al., Nuc. Acid. Res., 27: 4324-4327 (1999); Nuc. Acid. Res., 28: e99 (2000); U.S. Pat. No. 7,112,715; and Kramer & Fussenegger, Methods Mol. Biol., 308: 123-144 (2005)).
  • The term “enhancer” as used herein, refers to a DNA sequence that increases transcription of, for example, a nucleic acid sequence to which it is operably linked. Enhancers can be located many kilobases away from the coding region of the nucleic acid sequence and can mediate the binding of regulatory factors, patterns of DNA methylation, or changes in DNA structure. A large number of enhancers from a variety of different sources are well known in the art and are available as or within cloned polynucleotides (from, e.g., depositories such as the ATCC as well as other commercial or individual sources). A number of polynucleotides comprising promoters (such as the commonly-used CMV promoter) also comprise enhancer sequences. Enhancers can be located upstream, within, or downstream of coding sequences.
  • The vector also can comprise a “selectable marker gene.” The term “selectable marker gene,” as used herein, refers to a nucleic acid sequence that allow cells expressing the nucleic acid sequence to be specifically selected for or against, in the presence of a corresponding selective agent. Suitable selectable marker genes are known in the art and described in, e.g., International Patent Application Publications WO 1992/008796 and WO 1994/028143; Wigler et al., Proc. Natl. Acad. Sci. USA, 77: 3567-3570 (1980); O'Hare et al., Proc. Natl. Acad. Sci. USA, 78: 1527-1531 (1981); Mulligan & Berg, Proc. Natl. Acad. Sci. USA, 78: 2072-2076 (1981); Colberre-Garapin et al., J. Mol. Biol., 150: 1-14 (1981); Santerre et al., Gene, 30: 147-156 (1984); Kent et al., Science, 237: 901-903 (1987); Wigler et al., Cell, 11: 223-232 (1977); Szybalska & Szybalski, Proc. Natl. Acad. Sci. USA, 48: 2026-2034 (1962); Lowy et al., Cell, 22: 817-823 (1980); and U.S. Pat. Nos. 5,122,464 and 5,770,359.
  • In some embodiments, the vector is an “episomal expression vector” or “episome,” which is able to replicate in a host cell, and persists as an extrachromosomal segment of DNA within the host cell in the presence of appropriate selective pressure (see, e.g., Conese et al., Gene Therapy, 11: 1735-1742 (2004)). Representative commercially available episomal expression vectors include, but are not limited to, episomal plasmids that utilize Epstein Barr Nuclear Antigen 1 (EBNA1) and the Epstein Barr Virus (EBV) origin of replication (oriP). The vectors pREP4, pCEP4, pREP7, and pcDNA3.1 from Invitrogen (Carlsbad, Calif.) and pBK-CMV from Stratagene (La Jolla, Calif.) represent non-limiting examples of an episomal vector that uses T-antigen and the SV40 origin of replication in lieu of EBNA1 and oriP.
  • Other suitable vectors include integrating expression vectors, which may randomly integrate into the host cell's DNA, or may include a recombination site to enable the specific recombination between the expression vector and the host cell's chromosome. Such integrating expression vectors may utilize the endogenous expression control sequences of the host cell's chromosomes to effect expression of the desired protein. Examples of vectors that integrate in a site specific manner include, for example, components of the flp-in system from Invitrogen (Carlsbad, Calif.) (e.g., pcDNA™5/FRT), or the cre-lox system, such as can be found in the pExchange-6 Core Vectors from Stratagene (La Jolla, Calif.). Examples of vectors that randomly integrate into host cell chromosomes include, for example, pcDNA3.1 (when introduced in the absence of T-antigen) from Invitrogen (Carlsbad, Calif.), UCOE from Millipore (Billerica, Mass.), and pCI or pFN10A (ACT) FLEXI™ from Promega (Madison, Wis.).
  • Viral vectors also can be used. Representative commercially available viral expression vectors include, but are not limited to, the adenovirus-based Per.C6 system available from Crucell, Inc. (Leiden, The Netherlands), the lentiviral-based pLP1 from Invitrogen (Carlsbad, Calif.), and the retroviral vectors pFB-ERV plus pCFB-EGSH from Stratagene (La Jolla, Calif.).
  • Nucleic acid sequences encoding the inventive amino acid sequences can be provided to a cell on the same vector (i.e., in cis). A unidirectional promoter can be used to control expression of each nucleic acid sequence. In another embodiment, a combination of bidirectional and unidirectional promoters can be used to control expression of multiple nucleic acid sequences. Nucleic acid sequences encoding the inventive amino acid sequences alternatively can be provided to the population of cells on separate vectors (i.e., in trans). Each of the nucleic acid sequences in each of the separate vectors can comprise the same or different expression control sequences. The separate vectors can be provided to cells simultaneously or sequentially.
  • The vector(s) comprising the nucleic acid(s) encoding the inventive amino acid sequences can be introduced into a host cell that is capable of expressing the polypeptides encoded thereby, including any suitable prokaryotic or eukaryotic cell. As such, the invention provides an isolated cell comprising the inventive vector. Preferred host cells are those that can be easily and reliably grown, have reasonably fast growth rates, have well characterized expression systems, and can be transformed or transfected easily and efficiently.
  • Examples of suitable prokaryotic cells include, but are not limited to, cells from the genera Bacillus (such as Bacillus subtilis and Bacillus brevis), Escherichia (such as E. coli), Pseudomonas, Streptomyces, Salmonella, and Erwinia. Particularly useful prokaryotic cells include the various strains of Escherichia coli (e.g., K12, HB101 (ATCC No. 33694), DH5α, DH10, MC1061 (ATCC No. 53338), and CC102).
  • Preferably, the vector is introduced into a eukaryotic cell. Suitable eukaryotic cells are known in the art and include, for example, yeast cells, insect cells, and mammalian cells. Examples of suitable yeast cells include those from the genera Kluyveromyces, Pichia, Rhinosporidium, Saccharomyces, and Schizosaccharomyces. Preferred yeast cells include, for example, Saccharomyces cerivisae and Pichia pastoris.
  • Suitable insect cells are described in, for example, Kitts et al., Biotechniques, 14: 810-817 (1993); Lucklow, Curr. Opin. Biotechnol., 4: 564-572 (1993); and Lucklow et al., J. Virol., 67: 4566-4579 (1993). Preferred insect cells include Sf-9 and HI5 (Invitrogen, Carlsbad, Calif.).
  • Preferably, mammalian cells are utilized in the invention. A number of suitable mammalian host cells are known in the art, and many are available from the American Type Culture Collection (ATCC, Manassas, Va.). Examples of suitable mammalian cells include, but are not limited to, Chinese hamster ovary cells (CHO) (ATCC No. CCL61), CHO DHFR-cells (Urlaub et al., Proc. Natl. Acad. Sci. USA, 97: 4216-4220 (1980)), human embryonic kidney (HEK) 293 or 293T cells (ATCC No. CRL1573), and 3T3 cells (ATCC No. CCL92). Other suitable mammalian cell lines are the monkey COS-1 (ATCC No. CRL1650) and COS-7 cell lines (ATCC No. CRL1651), as well as the CV-1 cell line (ATCC No. CCL70). Further exemplary mammalian host cells include primate cell lines and rodent cell lines, including transformed cell lines. Normal diploid cells, cell strains derived from in vitro culture of primary tissue, as well as primary explants, are also suitable. Other suitable mammalian cell lines include, but are not limited to, mouse neuroblastoma N2A cells, HeLa, mouse L-929 cells, and BHK or HaK hamster cell lines, all of which are available from the ATCC. Methods for selecting suitable mammalian host cells and methods for transformation, culture, amplification, screening, and purification of cells are known in the art.
  • Most preferably, the mammalian cell is a human cell. For example, the mammalian cell can be a human lymphoid or lymphoid derived cell line, such as a cell line of pre-B lymphocyte origin. Examples of human lymphoid cells lines include, without limitation, RAMOS (CRL-1596), Daudi (CCL-213), EB-3 (CCL-85), DT40 (CRL-2111), 18-81 (Jack et al., Proc. Natl. Acad. Sci. USA, 85: 1581-1585 (1988)), Raji cells (CCL-86), and derivatives thereof.
  • A nucleic acid sequence encoding the inventive amino acid sequence may be introduced into a cell by “transfection,” “transformation,” or “transduction.” “Transfection,” “transformation,” or “transduction,” as used herein, refer to the introduction of one or more exogenous polynucleotides into a host cell by using physical or chemical methods. Many transfection techniques are known in the art and include, for example, calcium phosphate DNA co-precipitation (see, e.g., Murray E. J. (ed.), Methods in Molecular Biology, Vol. 7, Gene Transfer and Expression Protocols, Humana Press (1991)); DEAE-dextran; electroporation; cationic liposome-mediated transfection; tungsten particle-facilitated microparticle bombardment (Johnston, Nature, 346: 776-777 (1990)); and strontium phosphate DNA co-precipitation (Brash et al., Mol. Cell Biol., 7: 2031-2034 (1987)). Phage or viral vectors can be introduced into host cells, after growth of infectious particles in suitable packaging cells, many of which are commercially available.
  • The invention provides a composition comprising an effective amount of the inventive immunoglobulin heavy chain polypeptide, the inventive immunoglobulin light chain polypeptide, the inventive PD-1-binding agent, the inventive nucleic acid sequence encoding any of the foregoing, or the inventive vector comprising the inventive nucleic acid sequence. Preferably, the composition is a pharmaceutically acceptable (e.g., physiologically acceptable) composition, which comprises a carrier, preferably a pharmaceutically acceptable (e.g., physiologically acceptable) carrier, and the inventive amino acid sequences, antigen-binding agent, or vector. Any suitable carrier can be used within the context of the invention, and such carriers are well known in the art. The choice of carrier will be determined, in part, by the particular site to which the composition may be administered and the particular method used to administer the composition. The composition optionally can be sterile. The composition can be frozen or lyophilized for storage and reconstituted in a suitable sterile carrier prior to use. The compositions can be generated in accordance with conventional techniques described in, e.g., Remington: The Science and Practice of Pharmacy, 21st Edition, Lippincott Williams & Wilkins, Philadelphia, Pa. (2001).
  • The invention further provides a method of treating any disease or disorder in which the improper expression (e.g., overexpression) or increased activity of a PD-1 protein causes or contributes to the pathological effects of the disease, or a decrease in PD-1 protein levels or activity has a therapeutic benefit in mammals, preferably humans. The invention also provides a method of treating a cancer or an infectious disease in a mammal. The method comprises administering the aforementioned composition to a mammal having a cancer or an infectious disease, whereupon the cancer or infectious disease is treated in the mammal. As discussed herein, PD-1 is abnormally expressed in a variety of cancers (see, e.g., Brown et al., J. Immunol., 170: 1257-1266 (2003); and Flies et. al., Yale Journal of Biology and Medicine, 84: 409-421 (2011)), and PD-L1 expression in some renal cell carcinoma patients correlates with tumor aggressiveness. The inventive method can be used to treat any type of cancer known in the art, such as, for example, melanoma, renal cell carcinoma, lung cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, gall bladder cancer, laryngeal cancer, liver cancer, thyroid cancer, stomach cancer, salivary gland cancer, prostate cancer, pancreatic cancer, or Merkel cell carcinoma (see, e.g., Bhatia et al., Curr. Oncol. Rep., 13(6): 488-497 (2011)). The inventive method can be used to treat any type of infectious disease a disease or disorder caused by a bacterium, a virus, a fungus, or a parasite). Examples of infectious diseases that can be treated by the inventive method include, but are not limited to, diseases caused by a human immunodeficiency virus (HIV), a respiratory syncytial virus (RSV), an influenza virus, a dengue virus, a hepatitis B virus (HBV, or a hepatitis C virus (HCV)). Administration of a composition comprising the inventive immunoglobulin heavy chain polypeptide, the inventive immunoglobulin light chain polypeptide, the inventive PD-1-binding agent, the inventive nucleic acid sequence encoding any of the foregoing, or the inventive vector comprising the inventive nucleic acid sequence induces an immune response against a cancer or infectious disease in a mammal. An “immune response” can entail, for example, antibody production and/or the activation of immune effector cells (e.g., T-cells).
  • As used herein, the terms “treatment,” “treating,” and the like refer to obtaining a desired pharmacologic and/or physiologic effect. Preferably, the effect is therapeutic, i.e., the effect partially or completely cures a disease and/or adverse symptom attributable to the disease. To this end, the inventive method comprises administering a “therapeutically effective amount” of the PD-1-binding agent. A “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result. The therapeutically effective amount may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the PD-1-binding agent to elicit a desired response in the individual. For example, a therapeutically effective amount of a PD-1-binding agent of the invention is an amount which decreases PD-1 protein bioactivity in a human and/or enhances the immune response against a cancer or infectious disease.
  • Alternatively, the pharmacologic and/or physiologic effect may be prophylactic, i.e., the effect completely or partially prevents a disease or symptom thereof. In this respect, the inventive method comprises administering a “prophylactically effective amount” of the PD-1-binding agent. A “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired prophylactic result (e.g., prevention of disease onset).
  • A typical dose can be, for example, in the range of 1 pg/kg to 20 mg/kg of animal or human body weight; however, doses below or above this exemplary range are within the scope of the invention. The daily parenteral dose can be about 0.00001 μg/kg to about 20 mg/kg of total body weight (e.g., about 0.001 μg/kg, about 0.1 μg/kg, about 1 μg/kg, about 5 μg/kg, about 10 μg/kg, about 100 μg/kg, about 500 μg/kg, about 1 mg/kg, about 5 mg/kg, about 10 mg/kg, or a range defined by any two of the foregoing values), preferably from about 0.1 μg/kg to about 10 mg/kg of total body weight (e.g., about 0.5 μg/kg, about 1 μg/kg, about 50 μg/kg, about 150 μg/kg, about 300 μg/kg, about 750 μg/kg, about 1.5 mg/kg, about 5 mg/kg, or a range defined by any two of the foregoing values), more preferably from about 1 μg/kg to 5 mg/kg of total body weight (e.g., about 3 μg/kg, about 15 μg/kg, about 75 μg/kg, about 300 μg/kg, about 900 μg/kg, about 2 mg/kg, about 4 mg/kg, or a range defined by any two of the foregoing values), and even more preferably from about 0.5 to 15 mg/kg body weight per day (e.g., about 1 mg/kg, about 2.5 mg/kg, about 3 mg/kg, about 6 mg/kg, about 9 mg/kg, about 11 mg/kg, about 13 mg/kg, or a range defined by any two of the foregoing values). Therapeutic or prophylactic efficacy can be monitored by periodic assessment of treated patients. For repeated administrations over several days or longer, depending on the condition, the treatment can be repeated until a desired suppression of disease symptoms occurs. However, other dosage regimens may be useful and are within the scope of the invention. The desired dosage can be delivered by a single bolus administration of the composition, by multiple bolus administrations of the composition, or by continuous infusion administration of the composition.
  • The composition comprising an effective amount of the inventive immunoglohulin heavy chain polypeptide, the inventive immunoglobulin light chain polypeptide, the inventive PD-1-binding agent, the inventive nucleic acid sequence encoding any of the foregoing, or the inventive vector comprising the inventive nucleic acid sequence can be administered to a mammal using standard administration techniques, including oral, intravenous, intraperitoneal, subcutaneous, pulmonary, transdermal, intramuscular, intranasal, buccal, sublingual, or suppository administration. The composition preferably is suitable for parenteral administration. The term “parenteral,” as used herein, includes intravenous, intramuscular, subcutaneous, rectal, vaginal, and intraperitoneal administration. More preferably, the composition is administered to a mammal using peripheral systemic delivery by intravenous, intraperitoneal, or subcutaneous injection.
  • Once administered to a mammal (e.g., a human), the biological activity of the inventive PD-1-binding agent can be measured by any suitable method known in the art. For example, the biological activity can be assessed by determining the stability of a particular PD-1-binding agent. In one embodiment of the invention, the PD-1-binding agent (e.g., an antibody) has an in vivo half life between about 30 minutes and 45 days (e.g., about 30 minutes, about 45 minutes, about 1 hour, about 2 hours, about 4 hours, about 6 hours, about 10 hours, about 12 hours, about 1 day, about 5 days, about 10 days, about 15 days, about 25 days, about 35 days, about 40 days, about 45 days, or a range defined by any two of the foregoing values). In another embodiment, the PD-1-binding agent has an in vivo half life between about 2 hours and 20 days (e.g., about 5 hours, about 10 hours, about 15 hours, about 20 hours, about 2 days, about 3 days, about 7 days, about 12 days, about 14 days, about 17 days, about 19 days, or a range defined by any two of the foregoing values). In another embodiment, the PD-1-binding agent has an in vivo half life between about 10 days and about 40 days (e.g., about 10 days, about 13 days, about 16 days, about 18 days, about 20 days, about 23 days, about 26 days, about 29 days, about 30 days, about 33 days, about 37 days, about 38 days, about 39 days, about 40 days, or a range defined by any two of the foregoing values).
  • The biological activity of a particular PD-1-binding agent also can be assessed by determining its binding affinity to a PD-1 protein or an epitope thereof. The term “affinity” refers to the equilibrium constant for the reversible binding of two agents and is expressed as the dissociation constant (KD). Affinity of a binding agent to a ligand, such as affinity of an antibody for an epitope, can be, for example, from about 1 picomolar (pM) to about 100 micromolar (μM) (e.g., from about 1 picomolar (pM) to about 1 nanomolar (nM), from about 1 nM to about 1 micromolar (μM), or from about 1 μM to about 100 μM). In one embodiment, the PD-1-binding agent can bind to an PD-1 protein with a KD less than or equal to 1 nanomolar (e.g., 0.9 nM, 0.8 nM, 0.7 nM, 0.6 nM, 0.5 nM, 0.4 nM, 0.3 nM, 0.2 nM, 0.1 nM, 0.05 nM, 0.025 nM, 0.01 nM, 0.001 nM, or a range defined by any two of the foregoing values). In another embodiment, the PD-1-binding agent can bind to PD-1 with a KD less than or equal to 200 pM (e.g., 190 pM, 175 pM, 150 pM, 125 pM, 110 pM, 100 pM, 90 pM, 80 pM, 75 pM, 60 pM, 50 pM, 40 pM, 30 pM, 25 pM, 20 pM, 15 pM, 10 pM, 5 pM, 1 pM, or a range defined by any two of the foregoing values). Immunoglobulin affinity for an antigen or epitope of interest can be measured using any art-recognized assay. Such methods include, for example, fluorescence activated cell sorting (FACS), separable beads (e.g., magnetic beads), surface plasmon resonance (SPR), solution phase competition (KINEXA™), antigen panning, and/or ELISA (see, e.g., Janeway et al. (ed.s.), Immunobiology, 5th ed., Garland Publishing, New York, N.Y., 2001).
  • The PD-1-binding agent of the invention may be administered alone or in combination with other drugs (e.g., as an adjuvant). For example, the PD-1-binding agent can be administered in combination with other agents for the treatment or prevention of the diseases disclosed herein. In this respect, the PD-1-binding agent can be used in combination with at least one other anticancer agent including, for example, any chemotherapeutic agent known in the art, ionization radiation, small molecule anticancer agents, cancer vaccines, biological therapies (e.g., other monoclonal antibodies, cancer-killing viruses, gene therapy, and adoptive T-cell transfer), and/or surgery. When the inventive method treats an infectious disease, the PD-1-binding agent can be administered in combination with at least one anti-bacterial agent or at least one anti-viral agent. In this respect, the anti-bacterial agent can be any suitable antibiotic known in the art. The anti-viral agent can be any vaccine of any suitable type that specifically targets a particular virus (e.g., live-attenuated vaccines, subunit vaccines, recombinant vector vaccines, and small molecule anti-viral therapies (e.g., viral replication inhibitors and nucleoside analogs).
  • In another embodiment, the inventive PD-1 binding agent can be administered in combination with other agents that inhibit immune checkpoint pathways. For example, the inventive PD-1 binding agent can be administered in combination with agents that inhibit or antagonize the CTLA-4, TIM-3 or LAG-3 pathways. Combination treatments that simultaneously target two or more of these immune checkpoint pathways have demonstrated improved and potentially synergistic antitumor activity (see, e.g., Sakuishi et al., J. Exp. Med., 207: 2187-2194 (2010); Ngiow et al., Cancer Res., 71: 3540-3551 (2011); and Woo et al., Cancer Res., 72: 917-927 (2012)). In one embodiment, the inventive PD-1 binding agent is administered in combination with an antibody that binds to TIM-3 and/or an antibody that binds to LAG-3. In this respect, the inventive method of treating a cancer or an infectious disease in a mammal can further comprise administering to the mammal a composition comprising (i) an antibody that binds to a TIM-3 protein and (ii) a pharmaceutically acceptable carrier or a composition comprising (i) an antibody that binds to a LAG-3 protein and (ii) a pharmaceutically acceptable carrier.
  • In addition to therapeutic uses, the PD-1-binding agent described herein can be used in diagnostic or research applications. In this respect, the PD-1-binding agent can be used in a method to diagnose a cancer or infectious disease. In a similar manner, the PD-1-binding agent can be used in an assay to monitor PD-1 protein levels in a subject being tested for a disease or disorder that is associated with abnormal PD-1 expression. Research applications include, for example, methods that utilize the PD-1-binding agent, and a label to detect a PD-1 protein in a sample, e.g., in a human body fluid or in a cell or tissue extract. The PD-1-binding agent can be used with or without modification, such as covalent or non-covalent labeling with a detectable moiety. For example, the detectable moiety can be a radioisotope (e.g., 3H, 14C, 32P, 35S, or 125I), a fluorescent or chemiluminescent compound (e.g., fluorescein isothiocyanate, rhodainine, or luciferin), an enzyme (e.g., alkaline phosphatase, beta-galactosidase, or horseradish peroxidase), or prosthetic groups. Any method known in the art for separately conjugating an antigen-binding agent (e.g., an antibody) to a detectable moiety may be employed in the context of the invention (see, e.g., Hunter et al., Nature, 194: 495-496 (1962); David et al., Biochemistry, 13: 1014-1021 (1974); Pain et al., J. Immunol. Meth., 40: 219-230 (1981); and Nygren, J. Histochem. and Cytochem., 30: 407-412 (1982)).
  • PD-1 protein levels can be measured using the inventive PD-1-binding agent by any suitable method known in the art. Such methods include, for example, radioimmunoassay (RIA), and FACS. Normal or standard expression values of PD-1 protein can be established using any suitable technique, e.g., by combining a sample comprising, or suspected of comprising, a PD-1 polypeptide with a PD-1-specific antibody under conditions suitable to form an antigen-antibody complex. The antibody is directly or indirectly labeled with a detectable substance to facilitate detection of the bound or unbound antibody. Suitable detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, and radioactive materials (see, e.g., Zola, Monoclonal Antibodies: A Manual of Techniques, CRC Press, Inc. (1987)). The amount of PD-1 polypeptide expressed in a sample is then compared with a standard value.
  • The PD-1-binding agent can be provided in a kit, i.e., a packaged combination of reagents in predetermined amounts with instructions for performing a diagnostic assay. If the PD-1-binding agent is labeled with an enzyme, the kit desirably includes substrates and cofactors required by the enzyme (e.g., a substrate precursor which provides a detectable chromophore or fluorophore). In addition, other additives may be included in the kit, such as stabilizers, buffers (e.g., a blocking buffer or lysis buffer), and the like. The relative amounts of the various reagents can be varied to provide for concentrations in solution of the reagents which substantially optimize the sensitivity of the assay. The reagents may be provided as dry powders (typically lyophilized), including excipients which on dissolution will provide a reagent solution having the appropriate concentration.
  • The following examples further illustrate the invention but, of course, should not be construed as in any way limiting its scope.
  • EXAMPLE 1
  • This example demonstrates a method of generating monoclonal antibodies directed against human PD-1.
  • Several forms of genes encoding human PD-1 and its ligands PD-L1 and PD-L2 were generated as antigens for use in mouse immunization, hybridoma screening, and affinity maturation of CDR-grafted antibodies, and are schematically depicted in FIG. 1. Full-length human and cynomolgus monkey PD-1 genes were expressed with their native leader sequence and no added tags using a ubiquitous chromatin opening element (UCOE) single expression vector with hygromycin selection (Millipore, Billerica, Mass.). CHO-K1 cells were stably transfected with Lipofectamine LTX (Life Technologies, Carlsbad, Calif.) according to the manufacturer's instructions. Following selection with hygromycin, cells expressing PD-1 on the cell surface were identified by flow cytometry using a PE-conjugated mouse antibody to human PD-1 (BD Bioscience, Franklin Lakes, N.J.) and subcloned. Subclones were then selected for high-level and uniform PD-1 expression.
  • Nucleic acid sequences encoding soluble monomeric forms of the extracellular domain (ECD) of human and cynomolgus monkey PD-1 were constructed with His tags appended to the C-terminus of the ECD or as soluble dimeric fusion proteins with mouse IgG2a Fc as indicated in FIG. 1. Nucleic acid sequences encoding soluble dimeric forms of the ECDs of human PD-L1 and PD-L2 were constructed as fusion proteins with mouse IgG1 Fc as indicated in FIG. 1. Soluble proteins were expressed transiently in HEK 293 cells or in stable CHO cell lines using standard techniques. His-tagged proteins were purified from cell culture supernatant via Ni-affinity column chromatography followed by size exclusion chromatography. IgG-Fc fusion proteins were purified using protein A/G affinity chromatography. Purified proteins were analyzed by SDS-PAGE and size-exclusion chromatography to ensure homogeneity. Additionally, identity and size were confirmed by mass spectrometry.
  • For FACS sorting experiments, purified proteins were labeled with biotin using an NHS ester crosslinker (Thermo-Fisher Scientific, Inc., Waltham, Mass.) or the fluorescent dye DyLight 650 (Thermo-Fisher Scientific, Inc., Waltham, Mass.) using standard techniques.
  • Mice were immunized with either CHO cells expressing full-length PD-1 on the cell surface or the PD-1 ECD His protein. Specifically, female BALB/c mice (7 weeks old) were purchased from Harlan Laboratories, Inc. (Indianapolis, Ind.) and divided into two groups. After six days of acclimatization, one group of animals was immunized with four weekly doses of purified human PD-1 ECD-His at 50 μg/mouse, as a 1:1 emulsion with TITERMAX GOLD™ (Sigma Aldrich, St. Louis, Mo.). Immunization was carried out subcutaneously around the armpits and inguinal regions. The second group of animals was injected with four weekly doses of CHO-K1 cells stably expressing full length human PD-1 (5×106 cells/mouse) subcutaneously around the inguinal regions. After ten days, animals were bled for measurement of the serum titer to PD-1, and one animal from each group was boosted with soluble human PD-1 after a 3-week rest. After three days, spleens, axillary/brachial lymph nodes, and inguinal lymph nodes were collected from each animal. Single cell suspensions of cells from all tissues collected from both animals were pooled and used for generation of hybridomas by cell fusion using standard techniques. Two different myeloma cell lines were used for fusion, F0 (as described in de St. Groth and Scheidegger, J. Immunol. Methods, 35: 1-21 (1980)) and P3X63Ag8.653 (as described in Kearney et al., J. Immunol., 123: 1548-1550 (1979)).
  • Hybridoma supernatants from ten 96-well plates were screened for binding to a CHO-K1 cell clone stably transfected with a nucleic acid sequence encoding full length human PD-1 and compared to binding to untransfected CHO-K1 cells. Specifically, hybridoma supernatants were diluted 1:1 with PBS/2% FBS and incubated with an equal volume of PD-1 CHO-K1 cells (2.5×105 cells in PBS, 2% FBS) for 30 minutes at 4° C. Cells were centrifuged, washed once with PBS/1% FBS, and incubated with APC-conjugated goat anti-mouse IgG (H+L) (Southern Biotechnology, Birmingham, Ala.) for 30 minutes at 4° C. Cells were washed twice in PBS/2% FBS, resuspended in PBS, 2% FBS, 1% paraformaldehyde, and fluorescence analyzed on a BD FACSARRAY™ Bioanalyzer (BD Biosciences, Franklin Lakes, N.J.). Mouse IgG levels were quantified by ELISA.
  • Based on strong binding to PD-1 CHO cells, 46 parental wells were expanded, and the supernatants were tested for the ability to block binding of DyL650-labeled PD-L1-mIgG1 Fc fusion protein to PD-1 CHO cells. Specifically, purified mouse monoclonal antibodies were incubated in a dose response with the EC30 concentration of PD-L1-DyL650 (10 nM), and inhibition was quantified by flow cytometry. Cells from wells showing the best PD-L1 blocking activity and highest levels of mouse IgG were subcloned for further analysis, including purification and heavy and light chain (VH and VL) sequencing. Eleven of the strongest blockers of PD-1/PD-L1 interaction were selected for subcloning. Following re-confirmation of PD-1 binding and PD-L1 blocking, selected subclones were scaled up, and supernatant was submitted for antibody purification. Purified antibodies were verified for binding to both human and cynomolgus monkey PD-1 and for PD-L1 blocking activity. KD values were determined by surface plasmon resonance on a BIACORE™ T200 instrument (GE Healthcare, Waukesha, Wis.), and kinetic constants were determined using the BIACORE™ T200 evaluation software (GE Healthcare, Waukesha, Wis.). In this respect, antibodies were captured on a BIACORE™ CM5 chip to which GE anti-mouse IgG was coupled. PD-1-His monomer was flowed over the captured antibody using two- or three-fold serial dilutions beginning with 500 nM at the highest concentration. The resulting sensorgrams were fit globally using a 1:1 binding model to calculate on- and off-rates and the subsequent affinities (KD).
  • The results of this example demonstrate a method of producing monoclonal antibodies that bind to human and cynomolgus monkey PD-1 and block PD-1 ligand binding.
  • EXAMPLE 2
  • This example describes the design and generation of CDR-grafted and chimeric anti-PD-1 monoclonal antibodies.
  • Subclones of the hybridomas which produced PD-1-binding antibodies with PD-L1 blocking activity as described in Example 1 were isotyped, subjected to RT-PCR for cloning the antibody heavy chain variable region (VH) and light chain variable region (VL), and sequenced. Specifically, RNA was isolated from cell pellets of hybridoma clones (5×105 cells/pellet) using the RNEASY™ kit (Qiagen, Venlo, Netherlands), and cDNA was prepared using oligo-dT-primed SUPERSCRIPT™ III First-Strand Synthesis System (Life Technologies, Carlsbad, Calif.). PCR amplification of the VL utilized a pool of 9 or 11 degenerate mouse VL forward primers (see Kontermann and Dubel, eds., Antibody Engineering, Springer-Verlag, Berlin (2001)) and a mouse κ constant region reverse primer. PCR amplification of the VH utilized a pool of 12 degenerate mouse VH forward primers (Kontermann and Dubel, supra) and a mouse γ1 or γ2a constant region reverse primer (based on isotyping of purified antibody from each clone) with the protocol recommended in the SUPERSCRIPT™ III First-Stand Synthesis System (Life Technologies, Carlsbad, Calif.). PCR products were purified and cloned into pcDNA3.3-TOPO (Life Technologies, Carlsbad, Calif.). Individual colonies from each cell pellet (24 heavy chains and 48 light chains) were selected and sequenced using standard Sanger sequencing methodology (Genewiz, Inc., South Plainfield, N.J.). Variable region sequences were examined and aligned with the closest human heavy chain or light chain V-region germline sequence. Three antibodies were selected for CDR-grafting: (1) 9A2, comprising a VH of SEQ ID NO: 4 and a VL of SEQ ID NO: 28, (2) 10B11, comprising a VH of SEQ ID NO: 15 and a VL of SEQ ID NO: 32, and (3) 6E9, comprising a VH of SEQ ID NO: 22 and a VL of SEQ ID NO: 38.
  • CDR-grafted antibody sequences were designed by grafting CDR residues from each of the above-described mouse antibodies into the closest human germline homologue. CDR-grafted antibody variable regions were synthesized and expressed with human IgG1/κ constant regions for analysis. In addition, mouse:human chimeric antibodies were constructed using the variable regions of the above-described mouse antibodies linked to human IgG1/κ constant regions. Chimeric and CDR-grafted antibodies were characterized for binding to human and cynomolgus monkey PD-1 antigens and for activity in the PD-1/PD-L1 blocking assay as described above.
  • The functional antagonist activity of chimeric and CDR-grafted antibodies also was tested in a human CD4+ T-cell mixed lymphocyte reaction (MLR) assay in which activation of CD4+ T-cells in the presence of anti-PD-1 antibodies is assessed by measuring IL-2 secretion. Because PD-1 is a negative regulator of T-cell function, antagonism of PD-1 was expected to result in increased T-cell activation as measured by increased IL-2 production. The 9A2, 10B11, and 6E9 CDR-grafted antibodies demonstrated antagonistic activity and were selected for affinity maturation.
  • The results of this example demonstrate a method of generating chimeric and CDR-grafted monoclonal antibodies that specifically bind to and inhibit PD-1.
  • EXAMPLE 3
  • This example demonstrates affinity maturation of monoclonal antibodies directed against PD-1.
  • CDR-grafted antibodies derived from the original murine monoclonal antibodies, (9A2, 10B11, and 6E9) were subjected to affinity maturation via in vitro somatic hypermutation. Each antibody was displayed on the surface of HEK 293c18 cells using the SHM-XEL deciduous system (see Bowers et al., Proc. Natl. Acad. Sci. USA, 108: 20455-20460 (2011); and U.S. Patent Application Publication No. 2013/0035472). After establishment of stable episomal lines, a vector for expression of activation-induced cytosine deaminase (AID) was transfected into the cells to initiate somatic hypermutation as described in Bowers et al., supra. After multiple rounds of FACS sorting under conditions of increasing antigen binding stringency, a number of mutations in the variable region of each antibody were identified and recombined to produce mature humanized antibodies with improved properties.
  • A panel of six affinity-matured humanized heavy and light chain variable region sequences were paired (denoted APE1922, APE1923, APE1924, APE1950, APE1963 and APE2058) and selected for characterization, and are set forth in Table 1. The PD-1 binding properties of each of these antibody sequences were assayed using surface plasmon resonance (SPR) and solution-based affinity analysis. Antibodies were expressed from HEK 293 cells as human IgG1 antibodies and compared to the reference antibody, a human IgG1 version of BMS-936558, designated BMS.
  • SPR analyses were carried out using a BIACORE™ T200 instrument, and kinetic constants were determined using the BIACORE™ T200 evaluation software. Experimental parameters were chosen to ensure that saturation would be reached at the highest antigen concentrations and that Rmax values would be kept under 30 RU. GE anti-Human IgG (Fc-specific, approximately 7,000 RU) was immobilized on a BIACORE™ CM5 chip using EDC-activated amine coupling chemistry. Antibodies (0.5 μg/mL, 60 second capture time) were then captured using this surface. Next, monomeric soluble human PD1-Avi-His was flowed over captured antibody (300 second association, 300 second dissociation) using a three-fold serial dilution series from 500 nM to 2 nM. Captured antibody and antigen were removed between each cycle using 3 M MgCl2 (60 second contact time) in order to ensure a fresh binding surface for each concentration of antigen. The resulting sensorgrams were fit globally using a 1:1 binding model in order to calculate on- and off-rates (ka and kd, respectively), as well as affinities (KD).
  • Solution-based affinity analyses were carried out using a KINEXA™ 3000 assay (Sapidyne Instruments, Boise, Id.), and results were analyzed using KINEXA™ Pro Software 3.2.6. Experimental parameters were selected to reach a maximum signal with antibody alone between 0.8 and 1.2 V, while limiting nonspecific binding signal with buffer alone to less than 10% of the maximum signal. Azlactone beads (50 mg) were coated with antigen by diluting in a solution of PD-1-Avi-His (50 μg/mL in 1 mL) in 50 mM Na2CO3. The solution was rotated at room temperature for 2 hours, and beads were pelleted in a picofuge and washed twice with blocking solution (10 mg/mL BSA, 1 M Tris-HCl, pH 8.0). Beads were resuspended in blocking solution (1 mL), rotated at room temperature for 1 hour, and diluted in 25 volumes PBS/0.02% NaN3. For affinity measurement, the secondary antibody was ALEXFLUOR™ 647 dye-anti-human IgG (500 ng/mL). Sample antibody concentrations were held constant (50 pM or 75 pM), while antigen PD1-Avi-His was titrated using a three-fold dilutions series from 1 μM to 17 pM. All samples were diluted in PBS, 0.2% NaN3, 1 mg/mL BSA and allowed to equilibrate at room temperature for 30 hours. Additionally, samples containing only antibody and only buffer were tested in order to determine maximum signal and nonspecific binding signal, respectively. The results of the affinity analyses are set forth in Table 1. All of the selected antibodies exhibited higher affinities for PD-1 than the BMS reference antibody, with the highest affinity antibody being APE2058.
  • TABLE 1
    VH SEQ VL SEQ BIACORE ™ BIACORE ™ BIACORE ™ KINEXA ™
    Antibody ID NO: ID NO: ka (Ms)−1 kd (s−1) KD (nM) KD (nM)
    BMS n/a n/a 8.8 × 104 2.1 × 10−3 23 2
    APE1922 6 29 1.3 × 105 1.8 × 10−3 15
    APE1923 7 29 1.9 × 105 1.7 × 10−3 9 1
    APE1924 8 29 1.8 × 105 1.8 × 10−3 10 −1
    APE1950 9 29 1.5 × 105 2.5 × 10−3 17
    APE1963 10 29 5.8 × 104 1.0 × 10−3 17
    APE2058 23 40 3.0 × 105 6.4 × 10−4 2 0.2
  • To assess binding of the antibodies to cell surface PD-1, binding to CHO cells expressing either human or cnyomolgus monkey PD-1 was determined by flow cytometry analysis as described above. In addition, blocking of the PD-1/PD-L1 interaction was assessed using DyL650 labeled PD-L1 (mouse IgG1 Fc fusion protein) and PD-1-expressing CHO cells as described above. High binding affinities for cell-surface PD-1 were observed for all tested affinity-matured antibody sequences, with reactivity to cynomolgus monkey PD-1 within a factor of 3-4 fold of human. Blocking of the PD-1/PD-L1 interaction was also efficient with all of the tested affinity-matured antibody sequences, with IC50 values in the low nM range. These results were consistent with binding affinities assayed both by the BIACORE™ and KINEXA™ systems as well as cell surface EC50 values.
  • Thermal stability of the selected antibodies was assessed using a Thermofluor assay as described in McConnell et al., Protein Eng. Des. Sel., 26: 151 (2013). This assay assesses stability through the ability of a hydrophobic fluorescent dye to bind to hydrophobic patches on the protein surface which are exposed as the protein unfolds. The temperature at which 50% of the protein unfolds is determined (Tm) to measure thermal stability. This assay demonstrated that all of the tested affinity-matured antibody sequences had high thermal stability, and all were more stable than the reference antibody. APE2058 was the most stable antibody, exhibiting a Tm more than 10° C. greater than the Tm of the IgG1 version of BMS-936558.
  • De-risking of potential issues related to in vivo pharmokinetics of the tested antibodies was undertaken through (a) assessment of non-specific binding to target negative cells (see, e.g., Hotzel et al., mAbs, 4: 753-760 (2012)) and (b) measurement of differential neonatal Fc receptor (FcRn) dissociation properties (see, e.g., Wang et al., Drug Metab. Disp., 39: 1469-1477 (2011)). To assess non-specific binding, antibodies were tested for binding to HEK 293f cells using a flow cytometry-based assay. The tested antibodies were compared to two FDA-approved antibodies, infliximab and denosumab. The results indicated that non-specific binding was low for all of the antibodies. To assess FcRn binding and dissociation, both human FcRn and cynomolgus FcRn were tested in a BIACORE™-based assay. Antibodies were bound to FcRn at pH 6.0, and after pH adjustment to 7.4, residual bound antibody was determined. The results of this assay are shown in Table 2.
  • TABLE 2
    % Residual Binding at pH 7.4
    Antibody Human FcRn Cyno FcRn
    BMS 2.0 1.7
    APE1922 2.7 2.9
    APE1923 4.0 5.0
    APE1924 3.6 4.0
    APE1950 34.0 36.5
    APE1963 9.0 11.9
    APE2058 2.1 2.0
  • The results of this example demonstrate a method of generating the inventive immunoglobulin heavy and light chain polypeptides, which exhibit thermostability and high affinity for PD-1.
  • EXAMPLE 4
  • This example demonstrates the activity of the inventive immunoglobulin heavy and light chain polypeptides in vitro.
  • Functional antagonist activity of the VH and VL sequences described in Example 3 was tested in a human CD4+ T-cell MLR assay as described above. For determination of functional potency, the EC50 for each antibody was determined in five separate experiments using different human donors. The results are shown in Table 3 and demonstrate potent activity for each of the selected antibodies, which was indistinguishable from the activity of the reference antibody.
  • TABLE 3
    EC50 Values (μg/mL)
    BMS
    Reference APE2058 APE1922 APE1923 APE1924 APE1950 APE1963
    0.01 0.01 0.01 0.01 0.01 0.01 0.01
    0.03 0.10 0.10 0.2 0.03 0.20 0.10
    0.02 0.04 0.03 0.01 0.04 0.02 0.02
    0.01 0.01 0.01 0.02 0.01 0.01 0.07
    0.01 0.01 0.01 0.01 0.01 0.01 0.01
    Each line represents an independent experiment using different human donors for the responder CD4+ T cells.
    Italicized line with one responder produced higher IL-2 levels in the presence of the affinity-matured mAbs than in the other experiments, artificially raising the EC50 values.
  • The results of this example demonstrate that the inventive immunoglobulin heavy and light chain polypeptides can antagonize PD-1 signaling, resulting in increased T-cell activation.
  • EXAMPLE 5
  • This example demonstrates that a combination of the inventive PD-1 binding agent and either an anti-LAG-3 antibody or an anti-TIM-3 antibody enhances T-cell activation in vitro.
  • To establish parameters for combination studies, the anti-PD-1 antibody APE2058 was titrated in a dose response in the human CD4+ T-cell MLR assay described above. Antagonism of PD-1 signaling resulted in increased T-cell activation and a corresponding 4- to 5-fold increase in the production of IL-2.
  • Based on the results from titrating the APE2058 antibody in multiple MLR assays, an EC50 value of 20 ng/mL and a concentration 10-fold lower that represents an approximate EC10 value (2 ng/mL) were selected for combination studies with antagonist antibodies to the TIM-3 or LAG-3 checkpoint molecules.
  • A fully human anti-TIM-3 antibody was characterized in a CD4+ T cell in vitro assay as having antagonist activity as measured by increased IL-2 production in the presence of low levels of anti -CD3 and anti-CD28 antibodies. The anti-TIM-3 antibody demonstrated activity in the MLR assay with an EC50 value of approximately 0.3 μg/mL, as shown in FIG. 2 and Table 4, which is approximately 15-fold less activity than the anti-PD-1 APE2058 antibody alone (EC50 approximately 0.02 μg/mL). In combination with 0.02 μg/mL of APE2058, the anti-TIM-3 antagonist antibody stimulated increased amounts of IL-2 production as compared to APE2058 or anti-TIM-3 alone, resulting in a 10-fold decrease in the EC50 values, as shown in FIG. 2 and Table 4. These results demonstrate that enhanced T-cell activation occurs with combination inhibition of the PD-1 and TIM-3 checkpoint pathways.
  • A fully human antagonist anti-LAG-3 antibody (described in U.S. Patent Application Publication 2011/0150892) has demonstrated potent activity in blocking binding of recombinant soluble LAG-3 to MHC Class II positive cells. This antibody, designated herein as APE03109, was evaluated for functional activity in the human CD4+ T-cell MLR assay. APE03109 demonstrated activity in the MLR with an EC50 value of approximately 0.05 μg/mL, as shown in FIG. 3 and Table 4, which was similar to the activity of the anti-PD-1 antibody alone. In combination with 0.02 μg/mL of the anti-PD-1 APE2058 antibody, the APE03109 antibody stimulated increased amounts of IL-2 production over APE2058 or APE03109 alone, resulting in a 5-fold decrease in the EC50 values.
  • A time course of IL-2 production with the anti-LAG-3 APE03109 antibody alone and the combination of APE2058 with APE03109 also was characterized in a human CD4+ MLR assay. A similar decrease in EC50 value for the combination of 0.02 μg/mL APE2058 and APE03109 was observed after 72 hours of culture, as shown in FIG. 3. After 96 hours of culture the differences in EC50 values were not as pronounced; however, the levels of IL-2 produced in the cultures treated with 0.02 μg/mL of the anti-PD-1 APE2058 antibody and the anti-LAG-3 APE03109 antibody almost doubled as compared to cultures treated with APE03109 alone (2,200 pg/mL versus 1,200 pg/mL). Consistent with the time course of LAG-3 expression, no increased IL-2 production from adding APE03109 to APE2058 was observed after 24 hours, although APE2058 alone produced a dose responsive increase in IL-2 production at this time. In separate MLR experiments it was also demonstrated that the combination of APE2058 and APE03109 enhanced the levels of production of the T-cell cytokine IFN-γ by over 50% after 48 hours.
  • To demonstrate that the combined effects of the anti-TIM-3 antibody or the anti-LAG-3 antibody in the CD4+ T-cell MLR were due to target specificity, an irrelevant human IgG1 antibody, APE0422, was tested in combination with 0.02 μg/mL anti-PD-1 antibody APE2058. At the highest concentration tested (30 μg/mL), the APE0422 antibody exhibited no effect on IL-2 production over anti-PD-1 alone.
  • TABLE 4
    MLR Assay EC50 MLR Assay EC50 MLR Assay EC50 Fold
    Antibody Single agent with 2 ng/mL anti-PD-1 with 20 ng/mL anti-PD-1 Improvement
    Anti-TIM-3 330 ng/mL 310 ng/mL 33 ng/mL 10
    Anti-LAG-3 53 ng/mL 44 ng/mL 11 ng/mL 4.8
  • The results of this example demonstrate that the inventive PD-1-binding agent combined with antagonistic antibodies directed against TIM-3 or LAG-3 enhances CD4+ T-cell activation in vitro.
  • All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
  • The use of the terms “a” and “an” and “the” and “at least one” and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The use of the term “at least one” followed by a list of one or more items (for example, “at least one of A and B”) is to be construed to mean one item selected from the listed items (A or B) or any combination of two or more of the listed items (A and B), unless otherwise indicated herein or clearly contradicted by context. The terms “comprising,” “having,” “including,” and “containing” are to be construed as open-ended terms (i.e., meaning “including, but not limited to,”) unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.
  • Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.

Claims (36)

1. An isolated immunoglobulin heavy chain or light chain polypeptide, wherein the polypeptide comprises:
(I) an immunoglobulin heavy chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, wherein optionally
(a) residue 9 of SEQ ID NO: 1 is replaced with a different amino acid residue,
(b) one or more of residues 7, 8, and 9 of SEQ ID NO: 2 is replaced with a different amino acid residue,
(c) one or more of residues 1, 2, and 5 of SEQ ID NO: 3 is replaced with a different amino acid residue, or
(d) any combination of (a)-(c);
(II) an isolated immunoglobulin heavy chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, wherein optionally
(a) residue 9 of SEQ ID NO: 12 is replaced with a different amino acid residue,
(b) residue 8 and/or residue 9 of SEQ ID NO: 13 is replaced with a different amino acid residue,
(c) residue of SEQ ID NO: 14 is replaced with a different amino acid residue, or
(d) any combination of (a)-(c);
or
(III) an isolated immunoglobulin heavy chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 19, a CDR2 amino acid sequence of SEQ ID NO: 20, and a CDR3 amino acid sequence of SEQ ID NO: 21;
or
(IV) an isolated immunoglobulin light chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 26 and a CDR2 amino acid sequence of SEQ ID NO: 27;
or
(V) an isolated immunoglobulin light chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31, wherein optionally residue 12 of SEQ ID NO: 30 is replaced with a different amino acid residue;
or
(VI) an isolated immunoglobulin light chain polypeptide which comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37, wherein optionally
(a) residue 5 of SEQ ID NO: 36 is replaced with a different amino acid residue, and/or
(b) residue 4 of SEQ ID NO: 37 is replaced with a different amino acid residue;
or
(VII) an isolated immunoglobulin heavy chain polypeptide which comprises an amino acid sequence that is at least 90% identical to any one of SEQ ID NOs: 4-11, SEQ ID NOs: 15-18, SEQ ID NOs: 22-25; SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, or SEQ ID NO: 41.
2. The isolated immunoglobulin heavy chain polypeptide of claim 1, which comprises a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that residue 9 of SEQ ID NO: 1 is replaced with a methionine (M) residue.
3. (canceled)
4. The isolated immunoglobulin heavy chain polypeptide of claim 1, which comprises a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that
(a) residue 7 of SEQ ID NO: 2 is replaced with an asparagine (N) residue,
(b) residue 8 of SEQ ID NO: 2 is replaced with a serine (S) residue,
(c) residue 9 of SEQ ID NO: 2 is replaced with a threonine (T) residue, or
(d) any combination of(a)-(c).
5. (canceled)
6. The isolated immunoglobulin heavy chain polypeptide of claim 1, which comprises a CDR1 amino acid sequence of SEQ ID NO: 1, a CDR2 amino acid sequence of SEQ ID NO: 2, and a CDR3 amino acid sequence of SEQ ID NO: 3, except that
(a) residue 1 of SEQ ID NO: 3 is replaced with a glutamic acid (E) residue,
(b) residue 2 of SEQ ID NO: 3 is replaced with a tyrosine (Y) residue,
(c) residue 5 of SEQ ID NO: 3 is replaced with a serine (S) residue, or
(d) any combination of (a)-(c).
7.-9. (canceled)
10. The isolated immunoglobulin heavy chain polypeptide of claim 1, which comprises a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that residue 9 of SEQ ID NO: 12 is replaced with a leucine (L) residue.
11. (canceled)
12. The isolated immunoglobulin heavy chain polypeptide of claim 1, which comprises a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that
(a) residue 8 of SEQ ID NO: 13 is replaced with a tyrosine (Y) residue, and/or
(b) residue 9 of SEQ ID NO: 13 is replaced with an alanine (A) residue.
13. (canceled)
14. The isolated immunoglobulin heavy chain polypeptide of claim 1, which comprises a CDR1 amino acid sequence of SEQ ID NO: 12, a CDR2 amino acid sequence of SEQ ID NO: 13, and a CDR3 amino acid sequence of SEQ ID NO: 14, except that residue 5 of SEQ ID NO: 3 is replaced with a threonine (T) residue.
15.-23. (canceled)
24. The isolated immunoglobulin light chain polypeptide of claim 1, wherein the polypeptide comprises a complementarity determining region 1 (CDR) amino acid sequence of SEQ ID NO: 30 and a CDR2 amino acid sequence of SEQ ID NO: 31, except that residue 12 of SEQ ID NO: 30 is replacedwith a threonine (T) residue.
25.-26. (canceled)
27. The isolated immunoglobulin light chain polypeptide of claim 1, which comprises a CDR1 amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37, except that residue 5 of SEQ ID NO: 36 is replaced with a leucine (L) residue.
28. (canceled)
29. The isolated immunoglobulin light chain polypeptide of claim 1, which comprises a CDR1 amino acid sequence of SEQ ID NO: 35, a CDR2 amino acid sequence of SEQ ID NO: 36, and a CDR3 amino acid sequence of SEQ ID NO: 37, except that residue 4 of SEQ ID NO: 37 is replaced with an asparagine (N) residue.
30. (canceled)
31. The isolated immunoglobulin heavy chain or light chain polypeptide of claim 1, which comprises an amino acid sequence of SEQ ID NOs: 4-11, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 38, SEQ IS NO. 39, SEQ ID NO: 40, or SEQ ID NO: 41.
32. (canceled)
33. An isolated or purified nucleic acid sequence encoding the isolated immunoglobulin heavy chain or light chain polypeptide of claim 1, optionally in a vector.
34.-35. (canceled)
36. An isolated programmed death-1 protein (PD-1)-binding agent comprising an immunoglobulin heavy chain polypeptide and an immunoglobulin light chain polypeptide of claim 1.
37. The isolated PD-1-binding agent of claim 36, which is an antibody, an antibody conjugate, or an antigen-binding fragment thereof.
38. The isolated PD-1-binding agent of claim 36, which is an antibody fragment selected from F(ab′)2, Fab′, Fab, Fv, scFv, dsFv, dAb, and a single chain binding polypeptide.
39. An isolated or purified nucleic acid sequence encoding the PD-1-binding agent of claim 36, optionally in a vector.
40. (canceled)
41. An isolated cell comprising the nucleic acid, optionally in a vector, of claim 39.
42. A composition comprising the isolated PD-1-binding agent of claim 36 and a pharmaceutically acceptable carrier.
43. A method of treating a cancer or an infectious disease in a mammal, which method comprises administering an effective amount of the composition of claim 42 to a mammal having a cancer or an infectious disease, whereupon the cancer or infectious disease is treated in the mammal.
44.-48. (canceled)
49. The method of claim 43, wherein (a) the half-life of the PD-1-binding agent in the mammal is between 30 minutes and 45 days; (b) the PD-1-binding agent binds to PD-1 with a KD between about 1 picomolar (pM) and about 100 micromolar (μM): or both (a) and (b).
50. (canceled)
51. The method of claim 43, further comprising administering to the mammal a composition comprising (i) an antibody that binds to a TIM-3 protein and (ii) a pharmaceutically acceptable carrier; or (i) an antibody that binds to a LAG-3 protein and (ii) a pharmaceutically acceptable carrier.
52. (canceled)
US16/927,339 2013-05-02 2020-07-13 Antibodies Directed Against Programmed Death-1(PD-1) Abandoned US20210163589A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/927,339 US20210163589A1 (en) 2013-05-02 2020-07-13 Antibodies Directed Against Programmed Death-1(PD-1)
US18/334,665 US20230331843A1 (en) 2013-05-02 2023-06-14 Antibodies Directed Against Programmed Death-1(PD-1)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201361818755P 2013-05-02 2013-05-02
PCT/US2014/036525 WO2014179664A2 (en) 2013-05-02 2014-05-02 Antibodies directed against programmed death-1 (pd-1)
US201514888557A 2015-11-02 2015-11-02
US15/726,287 US10738117B2 (en) 2013-05-02 2017-10-05 Antibodies directed against programmed death-1 (PD-1)
US16/927,339 US20210163589A1 (en) 2013-05-02 2020-07-13 Antibodies Directed Against Programmed Death-1(PD-1)

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/726,287 Continuation US10738117B2 (en) 2013-05-02 2017-10-05 Antibodies directed against programmed death-1 (PD-1)

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/334,665 Continuation US20230331843A1 (en) 2013-05-02 2023-06-14 Antibodies Directed Against Programmed Death-1(PD-1)

Publications (1)

Publication Number Publication Date
US20210163589A1 true US20210163589A1 (en) 2021-06-03

Family

ID=51844119

Family Applications (4)

Application Number Title Priority Date Filing Date
US14/888,557 Active 2034-05-23 US9815897B2 (en) 2013-05-02 2014-05-02 Antibodies directed against programmed death-1 (PD-1)
US15/726,287 Active 2035-01-10 US10738117B2 (en) 2013-05-02 2017-10-05 Antibodies directed against programmed death-1 (PD-1)
US16/927,339 Abandoned US20210163589A1 (en) 2013-05-02 2020-07-13 Antibodies Directed Against Programmed Death-1(PD-1)
US18/334,665 Pending US20230331843A1 (en) 2013-05-02 2023-06-14 Antibodies Directed Against Programmed Death-1(PD-1)

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US14/888,557 Active 2034-05-23 US9815897B2 (en) 2013-05-02 2014-05-02 Antibodies directed against programmed death-1 (PD-1)
US15/726,287 Active 2035-01-10 US10738117B2 (en) 2013-05-02 2017-10-05 Antibodies directed against programmed death-1 (PD-1)

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/334,665 Pending US20230331843A1 (en) 2013-05-02 2023-06-14 Antibodies Directed Against Programmed Death-1(PD-1)

Country Status (24)

Country Link
US (4) US9815897B2 (en)
EP (2) EP2992017B1 (en)
JP (2) JP6742903B2 (en)
KR (1) KR102243062B1 (en)
CN (1) CN105339389B (en)
AU (2) AU2014259719B2 (en)
BR (1) BR112015026823A2 (en)
CA (1) CA2910278C (en)
CY (1) CY1123935T1 (en)
DK (1) DK2992017T3 (en)
ES (1) ES2845609T3 (en)
HK (1) HK1219743A1 (en)
HR (1) HRP20210122T1 (en)
HU (2) HUE053069T2 (en)
LT (2) LT2992017T (en)
MX (1) MX2015015037A (en)
NL (1) NL301120I2 (en)
PL (1) PL2992017T3 (en)
PT (1) PT2992017T (en)
RS (1) RS61400B1 (en)
RU (1) RU2723050C2 (en)
SG (1) SG11201508528TA (en)
SI (1) SI2992017T1 (en)
WO (1) WO2014179664A2 (en)

Families Citing this family (363)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL3409278T3 (en) 2011-07-21 2021-02-22 Sumitomo Pharma Oncology, Inc. Heterocyclic protein kinase inhibitors
PT2992017T (en) * 2013-05-02 2021-01-29 Anaptysbio Inc Antibodies directed against programmed death-1 (pd-1)
EP3049442A4 (en) 2013-09-26 2017-06-28 Costim Pharmaceuticals Inc. Methods for treating hematologic cancers
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
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
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
CN114081946A (en) 2014-03-12 2022-02-25 耶达研究与开发有限公司 Treatment of CNS diseases and injuries by reducing the level or activity of systemic regulatory T cells
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
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
US9982052B2 (en) 2014-08-05 2018-05-29 MabQuest, SA Immunological reagents
ES2847311T3 (en) 2014-08-05 2021-08-02 MabQuest SA Immunological reagents that bind to PD-1
JO3663B1 (en) 2014-08-19 2020-08-27 Merck Sharp & Dohme Anti-lag3 antibodies and antigen-binding fragments
EP3659621A1 (en) 2014-09-13 2020-06-03 Novartis AG Combination therapies for cancer
JP6827415B2 (en) 2014-10-31 2021-02-10 メレオ バイオファーマ 5 インコーポレイテッド Combination therapy for the treatment of the disease
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
MA41867A (en) * 2015-04-01 2018-02-06 Anaptysbio Inc T-CELL IMMUNOGLOBULIN AND MUCINE PROTEIN 3 ANTIBODIES (TIM-3)
MX2017015046A (en) 2015-05-29 2018-05-17 Agenus Inc Anti-ctla-4 antibodies and methods of use thereof.
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
BR112018000768A2 (en) * 2015-07-13 2018-09-25 Cytomx Therapeutics Inc anti-pd-1 antibodies, activatable anti-pd-1 antibodies and methods of use
CA2991628C (en) 2015-07-16 2020-04-07 Bioxcel Therapeutics, Inc. A novel approach for treatment of cancer using immunomodulation
EA201890296A1 (en) * 2015-07-30 2018-08-31 Макродженикс, Инк. PD-1-binding molecules and methods of their application
WO2017024465A1 (en) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
EP3337826A1 (en) 2015-08-20 2018-06-27 Sutro Biopharma, Inc. Anti-tim-3 antibodies, compositions comprising anti-tim-3 antibodies and methods of making and using anti-tim-3 antibodies
CN114605548A (en) 2015-09-01 2022-06-10 艾吉纳斯公司 anti-PD-1 antibodies and methods of use thereof
CN108368175B (en) * 2015-09-29 2021-08-06 上海张江生物技术有限公司 PD-1 antibodies and uses thereof
AU2016332725A1 (en) 2015-09-29 2018-03-22 Celgene Corporation PD-1 binding proteins and methods of use thereof
JP2018529719A (en) * 2015-09-30 2018-10-11 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 Combination of PD-1 system binding antagonist and ALK inhibitor for treating ALK negative cancer
SG10202008325XA (en) 2015-10-02 2020-09-29 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
MA43186B1 (en) * 2015-11-03 2022-03-31 Janssen Biotech Inc Antibodies specifically binding to pd-1 and uses thereof
GEP20217328B (en) 2015-12-14 2021-12-10 Macrogenics Inc Bispecific molecules having immunoreactivity with pd-1 and ctla-4, and methods of use thereof
BR112018012352A2 (en) 2015-12-16 2018-12-11 Merck Sharp & Dohme Corp. anti-lag3 antibodies and antigen binding fragments
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
CN109069628A (en) * 2016-01-14 2018-12-21 Bps生物科学有限公司 Anti- PD-1 antibody and application thereof
EP3964529A1 (en) * 2016-01-22 2022-03-09 Mabquest SA Non-blocking pd1 specific antibodies
US11214617B2 (en) 2016-01-22 2022-01-04 MabQuest SA Immunological reagents
CU20180088A7 (en) 2016-02-17 2019-05-03 Novartis Ag ANTIBODIES ANTI TGFBETA 2
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
TWI755395B (en) 2016-05-13 2022-02-21 美商再生元醫藥公司 Combination of anti-pd-1 antibodies and radiation to treat cancer
DK3458478T3 (en) * 2016-05-18 2021-03-22 Boehringer Ingelheim Int ANTI-PD-1 AND ANTI-LAG3 ANTIBODIES FOR CANCER TREATMENT
CN106008714B (en) * 2016-05-24 2019-03-15 瑞阳(苏州)生物科技有限公司 Anti-human PD-1 Humanized monoclonal antibodies and its application
BR112018074463A2 (en) 2016-05-27 2019-03-06 Agenus Inc. anti-tim-3 antibodies and methods of use.
KR20190015377A (en) 2016-06-02 2019-02-13 브리스톨-마이어스 스큅 컴퍼니 PD-1 blockade using nobilvir in refractory Hodgkin's lymphoma
PT3464368T (en) 2016-06-02 2023-08-17 Bristol Myers Squibb Co Use of an anti-pd-1 antibody in combination with an anti-cd30 antibody in lymphoma treatment
JP2019517512A (en) 2016-06-03 2019-06-24 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Use of anti-PD-1 antibodies in the treatment of patients with colorectal cancer
EP3463454A1 (en) 2016-06-03 2019-04-10 Bristol-Myers Squibb Company Anti-pd-1 antibody for use in a method of treatment of recurrent small cell lung cancer
WO2017210624A1 (en) 2016-06-03 2017-12-07 Bristol-Myers Squibb Company Anti-pd-1 antibody for use in a method of treating a tumor
WO2017214182A1 (en) * 2016-06-07 2017-12-14 The United States Of America. As Represented By The Secretary, Department Of Health & Human Services Fully human antibody targeting pdi for cancer immunotherapy
JP2019517557A (en) 2016-06-10 2019-06-24 アイオー セラピューティクス インコーポレイテッド Receptor-selective retinoid and rexinoid compounds and immunomodulators for cancer immunotherapy
AU2017283480A1 (en) 2016-06-13 2019-01-24 Torque Therapeutics, Inc. Methods and compositions for promoting immune cell function
KR102531889B1 (en) 2016-06-20 2023-05-17 키맵 리미티드 Anti-PD-L1 and IL-2 cytokines
RU2656181C1 (en) * 2016-07-13 2018-05-31 Закрытое Акционерное Общество "Биокад" Anti-pd-1 antibodies, method for their production, and method of application
CN107814845B (en) * 2016-09-14 2021-02-09 浙江特瑞思药业股份有限公司 Novel anti-PD-1 nano antibody and application thereof
EP3515944A4 (en) 2016-09-19 2020-05-06 Celgene Corporation Methods of treating immune disorders using pd-1 binding proteins
JP2019534859A (en) 2016-09-19 2019-12-05 セルジーン コーポレイション Method for treating vitiligo using PD-1 binding protein
IL265800B2 (en) 2016-10-11 2023-10-01 Agenus Inc Anti-lag-3 antibodies and methods of use thereof
CN110099925A (en) 2016-10-28 2019-08-06 百时美施贵宝公司 Use the method for anti-PD-1 Antybody therapy urothelial cancer
CN110062885A (en) 2016-11-01 2019-07-26 安奈普泰斯生物有限公司 For the antibody of T cell immunoglobulin and mucin 3 (TIM-3)
EP3666794A1 (en) * 2016-11-01 2020-06-17 AnaptysBio, Inc. Antibodies directed against programmed death- 1 (pd-1)
MA46708B1 (en) 2016-11-02 2021-10-29 Jounce Therapeutics Inc Anti-pd1 antibodies and their uses
CN110072890B (en) 2016-11-03 2022-11-29 百时美施贵宝公司 Activatable anti-CTLA-4 antibodies and uses thereof
US11279694B2 (en) 2016-11-18 2022-03-22 Sumitomo Dainippon Pharma Oncology, Inc. Alvocidib prodrugs and their use as protein kinase inhibitors
WO2018098352A2 (en) 2016-11-22 2018-05-31 Jun Oishi Targeting kras induced immune checkpoint expression
AU2017368155B2 (en) 2016-11-30 2022-02-24 Oncomed Pharmaceuticals, Inc. Methods for treatment of cancer comprising TIGIT-binding agents
LT3551660T (en) 2016-12-07 2023-12-27 Agenus Inc. Anti-ctla-4 antibodies and methods of use thereof
KR102603681B1 (en) 2016-12-07 2023-11-17 아게누스 인코포레이티드 Antibodies and methods of using them
JOP20190133A1 (en) 2016-12-08 2019-06-02 Innovent Biologics Suzhou Co Ltd Anti-tim-3 antibodies for combination with anti-pd-1 antibodies
CN106519034B (en) 2016-12-22 2020-09-18 鲁南制药集团股份有限公司 anti-PD-1 antibodies and uses thereof
WO2018119474A2 (en) * 2016-12-23 2018-06-28 Remd Biotherapeutics, Inc. Immunotherapy using antibodies that bind programmed death 1 (pd-1)
MA47208A (en) * 2017-01-09 2019-11-13 Tesaro Inc CANCER TREATMENT METHODS USING ANTI-PD-1 ANTIBODIES
KR20190098263A (en) 2017-01-09 2019-08-21 테사로, 인코포레이티드 How to Treat Cancer with Anti-TIM-3 Antibodies
MX2019008346A (en) 2017-01-13 2019-09-09 Agenus Inc T cell receptors that bind to ny-eso-1 and methods of use thereof.
EP3570870A1 (en) 2017-01-20 2019-11-27 Novartis AG Combination therapy for the treatment of cancer
CN110291109B (en) 2017-01-20 2023-01-31 大有华夏生物医药集团有限公司 Monoclonal antibodies to human programmed death receptor PD-1 and fragments thereof
EP3579874B1 (en) 2017-02-10 2021-07-21 Novartis AG 1-(4-amino-5-bromo-6-(1 h-pyrazol-1-yl)pyrimidin-2-yl)-1 h-pyrazol-4-ol and use thereof in the treatment of cancer
TWI674261B (en) 2017-02-17 2019-10-11 美商英能腫瘤免疫股份有限公司 Nlrp3 modulators
WO2018156740A1 (en) 2017-02-24 2018-08-30 Macrogenics, Inc. Bispecific binding molecules that are capable of binding cd137 and tumor antigens, and uses thereof
AU2018229278A1 (en) 2017-02-28 2019-10-17 Sanofi Therapeutic RNA
EP3601355A1 (en) 2017-03-31 2020-02-05 Bristol-Myers Squibb Company Methods of treating tumor
CN117003887A (en) 2017-04-03 2023-11-07 豪夫迈·罗氏有限公司 Immunoconjugates of anti-PD-1 antibodies with mutant IL-2 or with IL-15
BR112019019821A2 (en) 2017-04-05 2020-04-22 Hoffmann La Roche bispecific antibody, polynucleotide, prokaryotic or eukaryotic host cell, methods of producing the bispecific antibody, treating an individual who has cancer or a chronic viral infection and inhibiting tumor cell growth, pharmaceutical composition and use of the bispecific antibody
US11603407B2 (en) 2017-04-06 2023-03-14 Regeneron Pharmaceuticals, Inc. Stable antibody formulation
TWI788340B (en) 2017-04-07 2023-01-01 美商必治妥美雅史谷比公司 Anti-icos agonist antibodies and uses thereof
KR102629972B1 (en) 2017-04-13 2024-01-29 아게누스 인코포레이티드 Anti-CD137 antibody and methods of using the same
CN106939049B (en) * 2017-04-20 2019-10-01 苏州思坦维生物技术股份有限公司 The monoclonal antibody and the preparation method and application thereof of antagonism inhibition people PD-1 antigen and its ligand binding
EP3615572A1 (en) 2017-04-27 2020-03-04 Tesaro Inc. Antibody agents directed against lymphocyte activation gene-3 (lag-3) and uses thereof
AR111651A1 (en) 2017-04-28 2019-08-07 Novartis Ag CONJUGATES OF ANTIBODIES THAT INCLUDE TOLL TYPE RECEIVER AGONISTS AND COMBINATION THERAPIES
CA3062061A1 (en) 2017-05-01 2018-11-08 Agenus Inc. Anti-tigit antibodies and methods of use thereof
MA49144A (en) 2017-05-18 2020-03-25 Tesaro Inc POLYTHERAPIES FOR THE TREATMENT OF CANCER
AR111760A1 (en) 2017-05-19 2019-08-14 Novartis Ag COMPOUNDS AND COMPOSITIONS FOR THE TREATMENT OF SOLID TUMORS THROUGH INTRATUMORAL ADMINISTRATION
MX2019012038A (en) 2017-05-30 2019-11-18 Bristol Myers Squibb Co Compositions comprising a combination of an anti-lag-3 antibody, a pd-1 pathway inhibitor, and an immunotherapeutic agent.
BR112019020610A2 (en) 2017-05-30 2020-04-22 Bristol-Myers Squibb Company treatment of positive tumors for lag-3
CA3065304A1 (en) 2017-05-30 2018-12-06 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
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
BR112019025035A2 (en) 2017-06-01 2020-06-30 Compugen Ltd. method to treat cancer
CN110678483B (en) 2017-06-01 2023-09-22 百时美施贵宝公司 Methods of treating tumors with anti-PD-1 antibodies
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
TW201904993A (en) 2017-06-22 2019-02-01 瑞士商諾華公司 Use of IL-1β binding antibody
EP3642240A1 (en) 2017-06-22 2020-04-29 Novartis AG Antibody molecules to cd73 and uses thereof
PE20200717A1 (en) 2017-06-22 2020-07-21 Novartis Ag ANTIBODY MOLECULES THAT BIND AND USES CD73
WO2018235056A1 (en) 2017-06-22 2018-12-27 Novartis Ag Il-1beta binding antibodies for use in treating cancer
CA3066747A1 (en) 2017-06-27 2019-01-03 Novartis Ag Dosage regimens for anti-tim-3 antibodies and uses thereof
KR20200029543A (en) 2017-07-13 2020-03-18 아이오 테라퓨틱스, 인크. Receptor subtypes and functional selective retinoids and rexinoid compounds combined with immunomodulators for cancer immunotherapy
SG11202000248UA (en) 2017-07-14 2020-02-27 Innate Tumor Immunity Inc Nlrp3 modulators
JP2020527572A (en) 2017-07-20 2020-09-10 ノバルティス アーゲー Anti-LAG-3 antibody dosage regimen and its use
ES2932354T3 (en) 2017-07-28 2023-01-18 Bristol Myers Squibb Co Cyclic dinucleotides as anticancer agents
SI3661954T1 (en) 2017-08-03 2022-05-31 Amgen Inc. Interleukin-21 muteins and methods of treatment
JP7316263B2 (en) 2017-08-31 2023-07-27 ブリストル-マイヤーズ スクイブ カンパニー Cyclic dinucleotides as anticancer agents
KR102651946B1 (en) 2017-08-31 2024-03-26 브리스톨-마이어스 스큅 컴퍼니 Cyclic dinucleotides as anticancer agents
WO2019046500A1 (en) 2017-08-31 2019-03-07 Bristol-Myers Squibb Company Cyclic dinucleotides as anticancer agents
WO2019046591A1 (en) 2017-08-31 2019-03-07 Io Therapeutics, Inc. Rar selective agonists in combination with immune modulators for cancer immunotherapy
CN111278854A (en) 2017-09-04 2020-06-12 艾吉纳斯公司 T cell receptors that bind Mixed Lineage Leukemia (MLL) -specific phosphopeptides and methods of use thereof
US11497756B2 (en) 2017-09-12 2022-11-15 Sumitomo Pharma Oncology, Inc. Treatment regimen for cancers that are insensitive to BCL-2 inhibitors using the MCL-1 inhibitor alvocidib
MA50657A (en) 2017-09-26 2020-08-05 Tesaro Inc NIRAPARIB FORMULATIONS
MX2020003770A (en) * 2017-09-30 2020-07-29 Tesaro Inc Combination therapies for treating cancer.
US11801240B2 (en) 2017-10-06 2023-10-31 Tesaro, Inc. Combination therapies and uses thereof
KR20200061399A (en) 2017-10-10 2020-06-02 브리스톨-마이어스 스큅 컴퍼니 Cyclic dinucleotide as an anticancer agent
US20200239577A1 (en) 2017-10-15 2020-07-30 Bristol-Myers Squibb Company Methods of treating tumor
JP7254821B2 (en) 2017-10-16 2023-04-10 ブリストル-マイヤーズ スクイブ カンパニー Cyclic dinucleotides as anticancer agents
EP3700933A1 (en) 2017-10-25 2020-09-02 Novartis AG Antibodies targeting cd32b and methods of use thereof
US20210292415A1 (en) 2017-11-06 2021-09-23 Bristol-Myers Squibb Company Methods of treating a tumor
CN111213059B (en) 2017-11-06 2024-01-09 豪夫迈·罗氏有限公司 Diagnostic and therapeutic methods for cancer
CA3081602A1 (en) 2017-11-16 2019-05-23 Novartis Ag Combination therapies
TW201925782A (en) 2017-11-30 2019-07-01 瑞士商諾華公司 BCMA-targeting chimeric antigen receptor, and uses thereof
WO2019117690A1 (en) * 2017-12-15 2019-06-20 경북대학교 산학협력단 Peptide bound to pl-l1 and use thereof
KR20200105862A (en) 2017-12-27 2020-09-09 테사로, 인코포레이티드 How to treat cancer
EP3737408A1 (en) 2018-01-08 2020-11-18 Novartis AG Immune-enhancing rnas for combination with chimeric antigen receptor therapy
BR112020014121A2 (en) 2018-01-12 2020-12-01 Amgen Inc. anti-pd-1 antibodies and treatment methods
KR20200109339A (en) 2018-01-16 2020-09-22 브리스톨-마이어스 스큅 컴퍼니 How to treat cancer using antibodies to TIM3
WO2019144126A1 (en) 2018-01-22 2019-07-25 Pascal Biosciences Inc. Cannabinoids and derivatives for promoting immunogenicity of tumor and infected cells
BR112020014574A2 (en) 2018-01-22 2020-12-08 Bristol-Myers Squibb Company COMPOSITIONS AND METHODS FOR THE TREATMENT OF CANCER
CN117402114A (en) 2018-01-26 2024-01-16 埃克塞里艾克西斯公司 Compounds for the treatment of kinase dependent disorders
IL302626A (en) 2018-01-26 2023-07-01 Exelixis Inc Compounds for the treatment of kinase-dependent disorders
JP2021511357A (en) 2018-01-26 2021-05-06 エグゼリクシス, インコーポレイテッド Compounds for treating kinase-dependent disorders
US20200354457A1 (en) 2018-01-31 2020-11-12 Hoffmann-La Roche Inc. Bispecific antibodies comprising an antigen-binding site binding to lag3
EP3746116A1 (en) 2018-01-31 2020-12-09 Novartis AG Combination therapy using a chimeric antigen receptor
CA3090479A1 (en) 2018-02-05 2019-08-08 Tesaro, Inc Pediatric niraparib formulations and pediatric treatment methods
EP3752203A1 (en) 2018-02-13 2020-12-23 Novartis AG Chimeric antigen receptor therapy in combination with il-15r and il15
US10519187B2 (en) 2018-02-13 2019-12-31 Bristol-Myers Squibb Company Cyclic dinucleotides as anticancer agents
WO2019161536A1 (en) * 2018-02-23 2019-08-29 Eucure (Beijing) Biopharma Co. , Ltd Anti-pd-1 antibodies and uses thereof
JP7250808B2 (en) 2018-03-08 2023-04-03 ブリストル-マイヤーズ スクイブ カンパニー Cyclic dinucleotides as anticancer agents
AU2019236865A1 (en) 2018-03-23 2020-10-01 Bristol-Myers Squibb Company Antibodies against MICA and/or MICB and uses thereof
JP2021519771A (en) 2018-03-30 2021-08-12 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company How to treat a tumor
JP2021520201A (en) 2018-04-04 2021-08-19 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Anti-CD27 antibody and its use
US20210147547A1 (en) 2018-04-13 2021-05-20 Novartis Ag Dosage Regimens For Anti-Pd-L1 Antibodies And Uses Thereof
EP3781202A4 (en) * 2018-04-15 2022-01-05 Immvira Co., Limited Antibodies binding pd-1 and uses thereof
BR112020021539A2 (en) 2018-04-25 2021-01-19 Innate Tumor Immunity, Inc. NLRP3 MODULATORS
EP3784688A2 (en) 2018-04-26 2021-03-03 Agenus Inc. Heat shock protein-binding peptide compositions and methods of use thereof
AR126019A1 (en) 2018-05-30 2023-09-06 Novartis Ag ANTIBODIES AGAINST ENTPD2, COMBINATION THERAPIES AND METHODS OF USE OF ANTIBODIES AND COMBINATION THERAPIES
WO2019232244A2 (en) 2018-05-31 2019-12-05 Novartis Ag Antibody molecules to cd73 and uses thereof
TW202016136A (en) 2018-06-01 2020-05-01 瑞士商諾華公司 Binding molecules against bcma and uses thereof
MA52968A (en) 2018-06-23 2021-04-28 Hoffmann La Roche METHODS OF TREATMENT OF LUNG CANCER USING A PD-1 AXIS ANTAGONIST, PLATINUM AGENT AND TOPOISOMERASE II INHIBITOR
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
AU2019301944B2 (en) 2018-07-10 2022-02-24 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
KR20210034622A (en) 2018-07-18 2021-03-30 제넨테크, 인크. Lung cancer treatment method using PD-1 axis binding antagonist, anti-metabolite, and platinum agent
US20210261665A1 (en) * 2018-07-19 2021-08-26 Tayu Huaxia Biotech Medical Group Co., Ltd. Anti-pd-1 antibodies, dosages and uses thereof
WO2020021465A1 (en) 2018-07-25 2020-01-30 Advanced Accelerator Applications (Italy) S.R.L. Method of treatment of neuroendocrine tumors
EP3826660A1 (en) 2018-07-26 2021-06-02 Bristol-Myers Squibb Company Lag-3 combination therapy for the treatment of cancer
CN112888677A (en) 2018-08-16 2021-06-01 先天肿瘤免疫公司 Substituted 4-amino-1H-imidazo [4,5-c ] quinoline compounds and improved process for their preparation
CN112996567A (en) 2018-08-16 2021-06-18 先天肿瘤免疫公司 Imidazo [4,5-C ] quinoline-derived NLRP 3-modulators
CN113038989A (en) 2018-08-16 2021-06-25 先天肿瘤免疫公司 Imidazo [4,5-C ] quinoline-derived NLRP3 modulators
WO2020044252A1 (en) 2018-08-31 2020-03-05 Novartis Ag Dosage regimes for anti-m-csf antibodies and uses thereof
TW202024023A (en) 2018-09-03 2020-07-01 瑞士商赫孚孟拉羅股份公司 Therapeutic compounds and methods of use
TW202024638A (en) * 2018-09-04 2020-07-01 美商泰沙羅公司 Methods of treating cancer
WO2020049534A1 (en) 2018-09-07 2020-03-12 Novartis Ag Sting agonist and combination therapy thereof for the treatment of cancer
US20210347760A1 (en) 2018-10-03 2021-11-11 Tesaro, Inc. Niraparib Salts
US20210347758A1 (en) 2018-10-03 2021-11-11 Tesaro, Inc. Crystalline Forms of Niraparib Freebase
CN112839962A (en) 2018-10-09 2021-05-25 百时美施贵宝公司 Anti-merk antibodies for the treatment of cancer
CN112867803A (en) 2018-10-16 2021-05-28 诺华股份有限公司 Tumor mutational burden alone or in combination with immune markers as biomarkers for predicting response to targeted therapy
CN113286611A (en) 2018-10-19 2021-08-20 百时美施贵宝公司 Combination therapy for melanoma
JP2022505647A (en) 2018-10-23 2022-01-14 ブリストル-マイヤーズ スクイブ カンパニー How to treat a tumor
WO2020089811A1 (en) 2018-10-31 2020-05-07 Novartis Ag Dc-sign antibody drug conjugates
AU2019380307A1 (en) 2018-11-16 2021-07-01 Bristol-Myers Squibb Company Anti-NKG2A antibodies and uses thereof
JP7469305B2 (en) * 2018-11-19 2024-04-16 バイオサイトジェン ファーマシューティカルズ (ベイジン) カンパニー リミテッド Anti-PD-1 antibodies and uses thereof
EP3890749A4 (en) 2018-12-04 2022-08-03 Sumitomo Dainippon Pharma Oncology, Inc. Cdk9 inhibitors and polymorphs thereof for use as agents for treatment of cancer
EP3891508A1 (en) 2018-12-04 2021-10-13 Bristol-Myers Squibb Company Methods of analysis using in-sample calibration curve by multiple isotopologue reaction monitoring
EP3666905A1 (en) 2018-12-11 2020-06-17 Sanofi E. coli positive for pks island as marker of positive response to anti-pd1 therapy in colorectal cancer
US20230029213A1 (en) 2018-12-13 2023-01-26 Exelixis, Inc. Crystalline Forms and Salt Forms of a Kinase Inhibitor
EP3897637A1 (en) 2018-12-20 2021-10-27 Novartis AG Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
CN113166762A (en) 2018-12-21 2021-07-23 欧恩科斯欧公司 Novel conjugated nucleic acid molecules and uses thereof
US20220025036A1 (en) 2018-12-21 2022-01-27 Novartis Ag Use of il-1beta binding antibodies
US20200369762A1 (en) 2018-12-21 2020-11-26 Novartis Ag Use of il-1beta binding antibodies
KR102371173B1 (en) 2018-12-21 2022-03-04 오제 이뮈노테라프틱스 Humanized Anti-Human-PD-1 Antibody
KR20210108422A (en) 2018-12-21 2021-09-02 노파르티스 아게 Uses of IL-1β Binding Antibodies
WO2020128637A1 (en) 2018-12-21 2020-06-25 Novartis Ag Use of il-1 binding antibodies in the treatment of a msi-h cancer
CN113228190A (en) 2018-12-23 2021-08-06 豪夫迈·罗氏有限公司 Tumor classification based on predicted tumor mutation burden
EP3911417B1 (en) 2019-01-14 2022-10-26 Innate Tumor Immunity, Inc. Heterocyclic nlrp3 modulators , for use in the treatment of cancer
CN113286786A (en) 2019-01-14 2021-08-20 先天肿瘤免疫公司 NLRP3 modulators
CN113710702A (en) 2019-01-14 2021-11-26 健泰科生物技术公司 Methods of treating cancer with PD-1 axis binding antagonists and RNA vaccines
CN113301963A (en) 2019-01-14 2021-08-24 先天肿瘤免疫公司 Substituted quinazolines as NLRP3 modulators for the treatment of cancer
JP2022517112A (en) 2019-01-14 2022-03-04 イネイト・テューマー・イミュニティ・インコーポレイテッド NLRP3 modulator
TW202043274A (en) 2019-01-21 2020-12-01 法商賽諾菲公司 Therapeutic rna and anti-pd1 antibodies for advanced stage solid tumor cancers
CA3126721A1 (en) 2019-01-25 2020-07-30 Exelixis, Inc. Compounds for the treatment of kinase-dependent disorders
CA3129031A1 (en) 2019-02-12 2020-08-20 Novartis Ag Pharmaceutical combination comprising tno155 and a pd-1 inhibitor
WO2020167990A1 (en) 2019-02-12 2020-08-20 Tolero Pharmaceuticals, Inc. Formulations comprising heterocyclic protein kinase inhibitors
EA202192019A1 (en) 2019-02-15 2021-11-02 Новартис Аг DERIVATIVES OF 3- (1-OXO-5- (PIPERIDIN-4-YL) ISOINDOLIN-2-YL) PIPERIDINE-2,6-DIONE AND ROUTES OF THEIR APPLICATION
CN113329792A (en) 2019-02-15 2021-08-31 诺华股份有限公司 Substituted 3- (1-oxoisoindolin-2-yl) piperidine-2, 6-dione derivatives and uses thereof
JP2022525149A (en) 2019-03-20 2022-05-11 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 Treatment of Acute Myeloid Leukemia (AML) with Venetoclax Failure
EP3941463A1 (en) 2019-03-22 2022-01-26 Sumitomo Dainippon Pharma Oncology, Inc. Compositions comprising pkm2 modulators and methods of treatment using the same
KR20210146349A (en) 2019-03-28 2021-12-03 브리스톨-마이어스 스큅 컴퍼니 how to treat a tumor
EP3946625A1 (en) 2019-03-28 2022-02-09 Bristol-Myers Squibb Company Methods of treating tumor
CA3134522A1 (en) 2019-04-19 2020-10-22 Genentech, Inc. Anti-mertk antibodies and their methods of use
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
EP3966782A2 (en) 2019-05-06 2022-03-16 Tesaro, Inc. Methods for characterizing and treating a cancer type using cancer images
US20230295087A1 (en) 2019-05-13 2023-09-21 Bristol-Myers Squibb Company AGONISTS OF ROR GAMMAt
WO2020231713A1 (en) 2019-05-13 2020-11-19 Bristol-Myers Squibb Company AGONISTS OF ROR GAMMAt
JP2022534967A (en) 2019-05-30 2022-08-04 ブリストル-マイヤーズ スクイブ カンパニー Multiple tumor gene signatures and their uses
JP2022534981A (en) 2019-05-30 2022-08-04 ブリストル-マイヤーズ スクイブ カンパニー Cellular localization signatures and combination treatments
EP3976832A1 (en) 2019-05-30 2022-04-06 Bristol-Myers Squibb Company Methods of identifying a subject suitable for an immuno-oncology (i-o) therapy
BR112021024300A2 (en) 2019-06-03 2022-01-11 Exelixis Inc Crystalline salt forms of a kinase inhibitor
AR119069A1 (en) 2019-06-04 2021-11-24 Exelixis Inc COMPOUNDS FOR THE TREATMENT OF KINASE-DEPENDENT DISORDERS
KR20220041079A (en) 2019-06-18 2022-03-31 얀센 사이언시즈 아일랜드 언리미티드 컴퍼니 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
WO2021003417A1 (en) 2019-07-03 2021-01-07 Sumitomo Dainippon Pharma Oncology, Inc. Tyrosine kinase non-receptor 1 (tnk1) inhibitors and uses thereof
TWI809286B (en) 2019-07-05 2023-07-21 日商小野藥品工業股份有限公司 Treatment of hematological cancer with pd-1/cd3 bispecific protein
WO2021024020A1 (en) 2019-08-06 2021-02-11 Astellas Pharma Inc. Combination therapy involving antibodies against claudin 18.2 and immune checkpoint inhibitors for treatment of cancer
US20220306630A1 (en) 2019-08-06 2022-09-29 Bristol-Myers Squibb Company AGONISTS OF ROR GAMMAt
EP4011918A4 (en) 2019-08-08 2023-08-23 ONO Pharmaceutical Co., Ltd. Dual-specific protein
CR20220076A (en) 2019-08-30 2022-06-24 Agenus Inc Anti-cd96 antibodies and methods of use thereof
TW202124446A (en) 2019-09-18 2021-07-01 瑞士商諾華公司 Combination therapies with entpd2 antibodies
EP4031578A1 (en) 2019-09-18 2022-07-27 Novartis AG Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
AU2020350795A1 (en) 2019-09-22 2022-03-31 Bristol-Myers Squibb Company Quantitative spatial profiling for LAG-3 antagonist therapy
CA3155341A1 (en) 2019-09-25 2021-04-01 Seagen Inc. Combination anti-cd30 adc, anti-pd-1 and chemotherapeutic for treatment of hematopoietic cancers
AU2020353079A1 (en) 2019-09-25 2022-04-14 Bristol-Myers Squibb Company Composite biomarker for cancer therapy
WO2021074695A1 (en) 2019-10-16 2021-04-22 Avacta Life Sciences Limited PD-L1 INHIBITOR - TGFβ INHIBITOR BISPECIFIC DRUG MOIETIES.
JP2022553306A (en) 2019-10-21 2022-12-22 ノバルティス アーゲー TIM-3 inhibitors and uses thereof
BR112022007376A2 (en) 2019-10-21 2022-07-05 Novartis Ag COMBINATION THERAPIES WITH VENETOCLAX AND TIM-3 INHIBITORS
EP4055392A1 (en) 2019-11-05 2022-09-14 Bristol-Myers Squibb Company M-protein assays and uses thereof
WO2021092220A1 (en) 2019-11-06 2021-05-14 Bristol-Myers Squibb Company Methods of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy
WO2021092221A1 (en) 2019-11-06 2021-05-14 Bristol-Myers Squibb Company Methods of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy
BR112022008191A2 (en) 2019-11-08 2022-07-12 Bristol Myers Squibb Co LAG-3 ANTAGONIST THERAPY FOR MELANOMA
TW202130618A (en) 2019-11-13 2021-08-16 美商建南德克公司 Therapeutic compounds and methods of use
EP4058465A1 (en) 2019-11-14 2022-09-21 Cohbar Inc. Cxcr4 antagonist peptides
US20230000864A1 (en) 2019-11-22 2023-01-05 Sumitomo Pharma Oncology, Inc. Solid dose pharmaceutical composition
JP2023506187A (en) 2019-12-09 2023-02-15 シージェン インコーポレイテッド Combination therapy with LIV1-ADC and PD-1 antagonists
MX2022007464A (en) 2019-12-18 2022-06-27 Tesaro Inc Biopharmaceutical compositions and related methods.
EP4077394A1 (en) 2019-12-19 2022-10-26 NGM Biopharmaceuticals, Inc. Ilt3-binding agents and methods of use thereof
MX2022006932A (en) 2019-12-19 2022-07-11 Bristol Myers Squibb Co Combinations of dgk inhibitors and checkpoint antagonists.
BR112022011902A2 (en) 2019-12-20 2022-09-06 Novartis Ag COMBINATION THERAPIES
US20230348458A1 (en) 2020-01-10 2023-11-02 Innate Tumor Immunity, Inc. Nlrp3 modulators
CA3167413A1 (en) 2020-01-17 2021-07-22 Novartis Ag Combination comprising a tim-3 inhibitor and a hypomethylating agent for use in treating myelodysplastic syndrome or chronic myelomonocytic leukemia
US20230067202A1 (en) 2020-01-28 2023-03-02 Glaxosmithkline Intellectual Property Development Limited Combination Treatments and Uses and Methods Thereof
AU2021213969A1 (en) 2020-01-30 2022-09-01 ONA Therapeutics S.L. Combination therapy for treatment of cancer and cancer metastasis
AU2021212197A1 (en) 2020-01-31 2022-08-04 BioNTech SE Methods of inducing neoepitope-specific T cells with a PD-1 axis binding antagonist and an RNA vaccine
CN115362167A (en) 2020-02-06 2022-11-18 百时美施贵宝公司 IL-10 and uses thereof
US11577665B2 (en) 2020-02-27 2023-02-14 Cpk Interior Products Urethane and graphene interior trim panel
CA3173356A1 (en) 2020-02-28 2021-09-02 Novartis Ag A triple pharmaceutical combination comprising dabrafenib, an erk inhibitor and a raf inhibitor or a pd-1 inhibitor.
US20230235073A1 (en) 2020-03-06 2023-07-27 Ona Therapeutics, S.L. Anti-cd36 antibodies and their use to treat cancer
MX2022010912A (en) 2020-03-06 2022-11-09 Celgene Quanticel Res Inc Combination of an lsd-1 inhibitor and nivolumab for use in treating sclc or sqnsclc.
JP2023519254A (en) 2020-03-23 2023-05-10 ブリストル-マイヤーズ スクイブ カンパニー Anti-CCR8 Antibodies for Treating Cancer
JP2023523193A (en) 2020-04-21 2023-06-02 ノバルティス アーゲー Dosing Regimens for Treating Diseases Modulated by CSF-1R
AU2021265801A1 (en) 2020-05-01 2022-11-17 Ngm Biopharmaceuticals, Inc. ILt-binding agents and methods of use thereof
BR112022022335A2 (en) 2020-05-05 2023-01-10 Teon Therapeutics Inc TYPE 2 CANNABINOID RECEPTOR MODULATORS AND USES THEREOF
AR122644A1 (en) 2020-06-19 2022-09-28 Onxeo NEW CONJUGATED NUCLEIC ACID MOLECULES AND THEIR USES
CA3187944A1 (en) 2020-06-22 2021-12-30 Ngm Biopharmaceuticals, Inc. Lair-1-binding agents and methods of use thereof
MX2022015852A (en) 2020-06-23 2023-01-24 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6- dione derivatives.
KR20230033647A (en) 2020-06-30 2023-03-08 멘두스 비.브이. Use of leukemia-derived cells in ovarian cancer vaccines
JP2023532768A (en) 2020-07-07 2023-07-31 バイオエヌテック エスエー RNA for treatment of HPV-positive cancer
WO2022009157A1 (en) 2020-07-10 2022-01-13 Novartis Ag Lhc165 and spartalizumab combinations for treating solid tumors
US11787775B2 (en) 2020-07-24 2023-10-17 Genentech, Inc. Therapeutic compounds and methods of use
JP2023536264A (en) 2020-07-31 2023-08-24 エグゼリクシス, インコーポレイテッド Combinations for the treatment of cancer
EP4188549A1 (en) 2020-08-03 2023-06-07 Novartis AG Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
US20230314440A1 (en) 2020-08-21 2023-10-05 Exelixis, Inc. Method of treating cancer
WO2022047189A1 (en) 2020-08-28 2022-03-03 Bristol-Myers Squibb Company Lag-3 antagonist therapy for hepatocellular carcinoma
EP4204453A1 (en) 2020-08-31 2023-07-05 Bristol-Myers Squibb Company Cell localization signature and immunotherapy
EP4204021A1 (en) 2020-08-31 2023-07-05 Advanced Accelerator Applications International S.A. Method of treating psma-expressing cancers
EP4204020A1 (en) 2020-08-31 2023-07-05 Advanced Accelerator Applications International S.A. Method of treating psma-expressing cancers
EP4208482A1 (en) 2020-09-02 2023-07-12 Pharmabcine Inc. Combination therapy of a pd-1 antagonist and an antagonist for vegfr-2 for treating patients with cancer
EP3970489A1 (en) 2020-09-18 2022-03-23 CpK Interior Products Inc. Graphene-based antiviral polymer
IL301533A (en) 2020-09-24 2023-05-01 Merck Sharp ַ& Dohme Llc Stable formulations of programmed death receptor 1 (pd-1) antibodies and hyaluronidase variants and fragments thereof and methods of use thereof
CN116406369A (en) 2020-10-05 2023-07-07 百时美施贵宝公司 Method for concentrating proteins
WO2022076596A1 (en) 2020-10-06 2022-04-14 Codiak Biosciences, Inc. Extracellular vesicle-aso constructs targeting stat6
AR123855A1 (en) 2020-10-20 2023-01-18 Genentech Inc PEG-CONJUGATED ANTI-MERTK ANTIBODIES AND METHODS OF USE
US20240101666A1 (en) 2020-10-23 2024-03-28 Bristol-Myers Squibb Company Lag-3 antagonist therapy for lung cancer
WO2022094567A1 (en) 2020-10-28 2022-05-05 Ikena Oncology, Inc. Combination of an ahr inhibitor with a pdx inhibitor or doxorubicine
WO2022093981A1 (en) 2020-10-28 2022-05-05 Genentech, Inc. Combination therapy comprising ptpn22 inhibitors and pd-l1 binding antagonists
AU2021374590A1 (en) 2020-11-04 2023-06-01 Genentech, Inc. Subcutaneous dosing of anti-cd20/anti-cd3 bispecific antibodies
EP4240493A2 (en) 2020-11-04 2023-09-13 Genentech, Inc. Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies and anti-cd79b antibody drug conjugates
IL302396A (en) 2020-11-04 2023-06-01 Genentech Inc Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies
WO2022097060A1 (en) 2020-11-06 2022-05-12 Novartis Ag Cd19 binding molecules and uses thereof
WO2022119830A1 (en) 2020-12-02 2022-06-09 Genentech, Inc. Methods and compositions for neoadjuvant and adjuvant urothelial carcinoma therapy
WO2022120179A1 (en) 2020-12-03 2022-06-09 Bristol-Myers Squibb Company Multi-tumor gene signatures and uses thereof
TW202237119A (en) 2020-12-10 2022-10-01 美商住友製藥腫瘤公司 Alk-5 inhibitors and uses thereof
CA3201844A1 (en) 2020-12-17 2022-06-23 William W. Bachovchin Fap-activated radiotheranostics, and uses related thereto
WO2022135666A1 (en) 2020-12-21 2022-06-30 BioNTech SE Treatment schedule for cytokine proteins
WO2022135667A1 (en) 2020-12-21 2022-06-30 BioNTech SE Therapeutic rna for treating cancer
TW202245808A (en) 2020-12-21 2022-12-01 德商拜恩迪克公司 Therapeutic rna for treating cancer
AU2021416156A1 (en) 2020-12-28 2023-06-22 Bristol-Myers Squibb Company Methods of treating tumors
JP2024503265A (en) 2020-12-28 2024-01-25 ブリストル-マイヤーズ スクイブ カンパニー Antibody compositions and methods of use thereof
CN116723854A (en) 2021-01-22 2023-09-08 门德斯有限公司 Tumor vaccination method
WO2022162569A1 (en) 2021-01-29 2022-08-04 Novartis Ag Dosage regimes for anti-cd73 and anti-entpd2 antibodies and uses thereof
WO2022169921A1 (en) 2021-02-04 2022-08-11 Bristol-Myers Squibb Company Benzofuran compounds as sting agonists
WO2022187270A1 (en) 2021-03-01 2022-09-09 Xilio Development, Inc. Combination of ctla4 and pd1/pdl1 antibodies for treating cancer
AU2022230384A1 (en) 2021-03-01 2023-09-07 Xilio Development, Inc. Combination of masked ctla4 and pd1/pdl1 antibodies for treating cancer
CA3212351A1 (en) 2021-03-12 2022-09-15 Mendus B.V. Methods of vaccination and use of cd47 blockade
CN117321418A (en) 2021-03-18 2023-12-29 诺华股份有限公司 Cancer biomarkers and methods of use thereof
TW202304506A (en) 2021-03-25 2023-02-01 日商安斯泰來製藥公司 Combination therapy involving antibodies against claudin 18.2 for treatment of cancer
IL307262A (en) 2021-03-29 2023-11-01 Juno Therapeutics Inc Methods for dosing and treatment with a combination of a checkpoint inhibitor therapy and a car t cell therapy
EP4314068A1 (en) 2021-04-02 2024-02-07 The Regents Of The University Of California Antibodies against cleaved cdcp1 and uses 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
KR20230170039A (en) 2021-04-13 2023-12-18 뉴베일런트, 아이엔씨. Amino-substituted heterocycles for treating cancer with EGFR mutations
JP2024516230A (en) 2021-04-30 2024-04-12 ジェネンテック, インコーポレイテッド Therapeutic and diagnostic methods and compositions for cancer
EP4330282A1 (en) 2021-04-30 2024-03-06 F. Hoffmann-La Roche AG Dosing for combination treatment with anti-cd20/anti-cd3 bispecific antibody and anti-cd79b antibody drug conjugate
WO2022234003A1 (en) 2021-05-07 2022-11-10 Avacta Life Sciences Limited Cd33 binding polypeptides with stefin a protein
WO2022240741A1 (en) 2021-05-12 2022-11-17 Dana-Farber Cancer Institute, Inc. Lag3 and gal3 inhibitory agents, xbp1, cs1, and cd138 peptides, and methods of use thereof
AR125874A1 (en) 2021-05-18 2023-08-23 Novartis Ag COMBINATION THERAPIES
CN117412767A (en) 2021-05-25 2024-01-16 雪绒花免疫公司 C-X-C motif chemokine receptor 6 (CXCR 6) binding molecules and methods of use thereof
CN113234160B (en) * 2021-05-26 2022-05-27 广州爱思迈生物医药科技有限公司 anti-PD-1 antibody and application thereof
WO2022256538A1 (en) 2021-06-03 2022-12-08 Synthorx, Inc. Head and neck cancer combination therapy comprising an il-2 conjugate and cetuximab
GB202107994D0 (en) 2021-06-04 2021-07-21 Kymab Ltd Treatment of cancer
BR112023026966A2 (en) 2021-07-02 2024-03-12 Hoffmann La Roche METHODS FOR TREATING AN INDIVIDUAL WITH MELANOMA, FOR ACHIEVING A CLINICAL RESPONSE, FOR TREATING AN INDIVIDUAL WITH NON-HODGKIN LYMPHOMA, FOR TREATING A POPULATION OF INDIVIDUALS WITH NON-HODGKIN LYMPHOMA, AND FOR TREATING AN INDIVIDUAL WITH METASTATIC COLORECTAL CANCER
WO2023285552A1 (en) 2021-07-13 2023-01-19 BioNTech SE Multispecific binding agents against cd40 and cd137 in combination therapy for cancer
WO2023010094A2 (en) 2021-07-28 2023-02-02 Genentech, Inc. Methods and compositions for treating cancer
CA3224180A1 (en) 2021-07-28 2023-02-02 F. Hoffmann-La Roche Ag Methods and compositions for treating cancer
AU2022320051A1 (en) 2021-07-30 2024-01-25 ONA Therapeutics S.L. Anti-cd36 antibodies and their use to treat cancer
TW202325306A (en) 2021-09-02 2023-07-01 美商天恩治療有限公司 Methods of improving growth and function of immune cells
WO2023056403A1 (en) 2021-09-30 2023-04-06 Genentech, Inc. Methods for treatment of hematologic cancers using anti-tigit antibodies, anti-cd38 antibodies, and pd-1 axis binding antagonists
WO2023051926A1 (en) 2021-09-30 2023-04-06 BioNTech SE Treatment involving non-immunogenic rna for antigen vaccination and pd-1 axis binding antagonists
WO2023057534A1 (en) 2021-10-06 2023-04-13 Genmab A/S Multispecific binding agents against pd-l1 and cd137 in combination
TW202334196A (en) 2021-10-07 2023-09-01 英商阿法克塔生命科學有限公司 Pd-l1 binding polypeptides
TW202333802A (en) 2021-10-11 2023-09-01 德商拜恩迪克公司 Therapeutic rna for lung cancer
AU2022375806A1 (en) 2021-10-29 2023-12-14 Bristol-Myers Squibb Company Lag-3 antagonist therapy for hematological cancer
WO2023081730A1 (en) 2021-11-03 2023-05-11 Teon Therapeutics, Inc. 4-hydroxy-2-oxo-1,2-dihydro-1,8-naphthyridine-3-carboxamide derivatives as cannabinoid cb2 receptor modulators for the treatment of cancer
WO2023080900A1 (en) 2021-11-05 2023-05-11 Genentech, Inc. Methods and compositions for classifying and treating kidney cancer
WO2023083439A1 (en) 2021-11-09 2023-05-19 BioNTech SE Tlr7 agonist and combinations for cancer treatment
TW202319073A (en) 2021-11-12 2023-05-16 瑞士商諾華公司 Combination therapy for treating lung cancer
WO2023097211A1 (en) 2021-11-24 2023-06-01 The University Of Southern California Methods for enhancing immune checkpoint inhibitor therapy
US20230203062A1 (en) 2021-11-24 2023-06-29 Genentech, Inc. Therapeutic compounds and methods of use
WO2023097195A1 (en) 2021-11-24 2023-06-01 Genentech, Inc. Therapeutic indazole compounds and methods of use in the treatment of 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
TW202340148A (en) 2021-12-22 2023-10-16 美商艾克塞里克斯公司 Crystalline forms and salt forms of a kinase inhibitor
WO2023122723A1 (en) 2021-12-23 2023-06-29 The Broad Institute, Inc. Panels and methods for diagnosing and treating lung cancer
WO2023147371A1 (en) 2022-01-26 2023-08-03 Bristol-Myers Squibb Company Combination therapy for hepatocellular carcinoma
WO2023164638A1 (en) 2022-02-25 2023-08-31 Bristol-Myers Squibb Company Combination therapy for colorectal carcinoma
WO2023168404A1 (en) 2022-03-04 2023-09-07 Bristol-Myers Squibb Company Methods of treating a tumor
WO2023170606A1 (en) 2022-03-08 2023-09-14 Alentis Therapeutics Ag Use of anti-claudin-1 antibodies to increase t cell availability
WO2023173091A1 (en) 2022-03-11 2023-09-14 Ngm Biopharmaceuticals, Inc. Osteoclast-associated ig-like receptor (oscar) and methods of use thereof
WO2023178329A1 (en) 2022-03-18 2023-09-21 Bristol-Myers Squibb Company Methods of isolating polypeptides
WO2023191816A1 (en) 2022-04-01 2023-10-05 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023196987A1 (en) 2022-04-07 2023-10-12 Bristol-Myers Squibb Company Methods of treating tumor
WO2023196964A1 (en) 2022-04-08 2023-10-12 Bristol-Myers Squibb Company Machine learning identification, classification, and quantification of tertiary lymphoid structures
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors
WO2023219613A1 (en) 2022-05-11 2023-11-16 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023218046A1 (en) 2022-05-12 2023-11-16 Genmab A/S Binding agents capable of binding to cd27 in combination therapy
WO2023218243A1 (en) 2022-05-12 2023-11-16 Avacta Life Sciences Limited Lag-3/pd-l1 binding fusion proteins
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
WO2023235847A1 (en) 2022-06-02 2023-12-07 Bristol-Myers Squibb Company Antibody compositions and methods of use thereof
WO2023240058A2 (en) 2022-06-07 2023-12-14 Genentech, Inc. Prognostic and therapeutic methods for cancer
WO2024015897A1 (en) 2022-07-13 2024-01-18 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024015372A1 (en) 2022-07-14 2024-01-18 Teon Therapeutics, Inc. Adenosine receptor antagonists and uses thereof
WO2024020432A1 (en) 2022-07-19 2024-01-25 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
EP4310197A1 (en) 2022-07-21 2024-01-24 Fundación para la Investigación Biomédica del Hospital Universitario Puerta de Hierro Majadahonda Method for identifying lung cancer patients for a combination treatment of immuno- and chemotherapy
WO2024023740A1 (en) 2022-07-27 2024-02-01 Astrazeneca Ab Combinations of recombinant virus expressing interleukin-12 with pd-1/pd-l1 inhibitors
WO2024040194A1 (en) 2022-08-17 2024-02-22 Capstan Therapeutics, Inc. Conditioning for in vivo immune cell engineering
WO2024049949A1 (en) 2022-09-01 2024-03-07 Genentech, Inc. Therapeutic and diagnostic methods for bladder cancer
WO2024069009A1 (en) 2022-09-30 2024-04-04 Alentis Therapeutics Ag Treatment of drug-resistant hepatocellular carcinoma
WO2024077095A1 (en) 2022-10-05 2024-04-11 Genentech, Inc. Methods and compositions for classifying and treating bladder cancer
WO2024077166A1 (en) 2022-10-05 2024-04-11 Genentech, Inc. Methods and compositions for classifying and treating lung cancer

Family Cites Families (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
DE69128037T2 (en) 1990-11-13 1998-05-07 Immunex Corp BIFUNCTIONAL SELECTABLE FUSION GENES
EP0804590A1 (en) 1993-05-21 1997-11-05 Targeted Genetics Corporation Bifunctional selectable fusion genes based on the cytosine deaminase (cd) gene
US5464758A (en) 1993-06-14 1995-11-07 Gossen; Manfred Tight control of gene expression in eucaryotic cells by tetracycline-responsive promoters
US5814618A (en) 1993-06-14 1998-09-29 Basf Aktiengesellschaft Methods for regulating gene expression
JP4896327B2 (en) 1999-08-23 2012-03-14 ダナ−ファーバー キャンサー インスティテュート,インコーポレイテッド PD-1, B7-4 receptors and uses thereof
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
FR2814642B1 (en) 2000-10-03 2005-07-01 Ass Pour Le Dev De La Rech En TRANSGENIC MOUSE FOR THE TARGETED RECOMBINATION MEDIATED BY THE MODIFIED CRE-ER
WO2002039813A1 (en) 2000-11-15 2002-05-23 Ono Pharmaceutical Co., Ltd. Pd-1-lacking mouse and use thereof
WO2002079499A1 (en) 2001-04-02 2002-10-10 Wyeth Pd-1, a receptor for b7-4, and uses therefor
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
ATE524495T1 (en) 2001-07-31 2011-09-15 Ono Pharmaceutical Co PD-1 SPECIFIC SUBSTANCE
WO2003042402A2 (en) 2001-11-13 2003-05-22 Dana-Farber Cancer Institute, Inc. Agents that modulate immune cell activation and methods of use thereof
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
CN1753912B (en) 2002-12-23 2011-11-02 惠氏公司 Antibodies against PD-1 and uses therefor
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
CN105315373B (en) * 2005-05-09 2018-11-09 小野药品工业株式会社 The human monoclonal antibodies of programmed death-1 (PD-1) and the method for carrying out treating cancer using anti-PD-1 antibody
PL2397156T3 (en) * 2005-06-08 2017-07-31 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
EP2937360A1 (en) * 2005-06-17 2015-10-28 Merck Sharp & Dohme Corp. Ilt3 binding molecules and uses therefor
JP5093097B2 (en) 2006-03-03 2012-12-05 小野薬品工業株式会社 Extracellular domain multimers of cell surface functional molecules
NZ629273A (en) 2006-12-27 2015-02-27 Harvard College Compositions and methods for the treatment of infections and tumors
WO2008103475A1 (en) 2007-02-20 2008-08-28 Anaptysbio, Inc. Somatic hypermutation systems
ES2623925T3 (en) 2007-05-30 2017-07-12 Postech Academy-Industry- Foundation Immunoglobulin Fusion Proteins
KR101562580B1 (en) 2007-06-18 2015-10-22 머크 샤프 앤 도메 비.브이. Antibodies to human programmed death receptor PD-1
US20090028857A1 (en) * 2007-07-23 2009-01-29 Cell Genesys, Inc. Pd-1 antibodies in combination with a cytokine-secreting cell and methods of use thereof
US20120269806A1 (en) 2007-08-21 2012-10-25 The General Hospital Corporation Methods of inducing tolerance
US8563314B2 (en) 2007-09-27 2013-10-22 Sangamo Biosciences, Inc. Methods and compositions for modulating PD1
WO2009114335A2 (en) * 2008-03-12 2009-09-17 Merck & Co., Inc. Pd-1 binding proteins
US8460886B2 (en) 2008-07-04 2013-06-11 Ono Pharmaceutical Co., Ltd. Use of an efficacy marker for optimizing therapeutic efficacy of an anti-human PD-1 antibody on cancers
AR072999A1 (en) 2008-08-11 2010-10-06 Medarex Inc HUMAN ANTIBODIES THAT JOIN GEN 3 OF LYMPHOCYTARY ACTIVATION (LAG-3) AND THE USES OF THESE
PL2350129T3 (en) 2008-08-25 2015-12-31 Amplimmune Inc Compositions of pd-1 antagonists and methods of use
EP2342229A1 (en) 2008-09-12 2011-07-13 ISIS Innovation Limited Pd-1 specific antibodies and uses thereof
CA2736816C (en) 2008-09-12 2018-05-22 Isis Innovation Limited Pd-1 specific antibodies and uses thereof
US8552154B2 (en) 2008-09-26 2013-10-08 Emory University Anti-PD-L1 antibodies and uses therefor
KR101050829B1 (en) * 2008-10-02 2011-07-20 서울대학교산학협력단 Anticancer agents comprising an anti-PD-1 antibody or an anti-PD-L1 antibody
CA2744449C (en) 2008-11-28 2019-01-29 Emory University Methods for the treatment of infections and tumors
EP3192811A1 (en) 2009-02-09 2017-07-19 Université d'Aix-Marseille Pd-1 antibodies and pd-l1 antibodies and uses thereof
CN105693861A (en) 2009-12-29 2016-06-22 新兴产品开发西雅图有限公司 Heterodimer binding protein and application thereof
US20130202623A1 (en) 2010-02-16 2013-08-08 Nicolas Chomont Pd-1 modulation and uses thereof for modulating hiv replication
CA2791930A1 (en) 2010-03-11 2011-09-15 Kerry Louise Tyson Pd-1 antibody
TW201134488A (en) 2010-03-11 2011-10-16 Ucb Pharma Sa PD-1 antibodies
WO2011115996A1 (en) 2010-03-17 2011-09-22 Anaptysbio, Inc. Method of producing transcripts using cryptic splice sites
WO2011159877A2 (en) 2010-06-18 2011-12-22 The Brigham And Women's Hospital, Inc. Bi-specific antibodies against tim-3 and pd-1 for immunotherapy in chronic immune conditions
CA2805564A1 (en) * 2010-08-05 2012-02-09 Stefan Jenewein Anti-mhc antibody anti-viral cytokine fusion protein
WO2012021851A2 (en) 2010-08-12 2012-02-16 Beth Israel Deaconess Medical Center Methods and compositions for diagnosing and treating lupus
US20130310266A1 (en) 2010-09-03 2013-11-21 Immport Therapeutics, Inc. Methods and Compositions For The Diagnosis And Treatment Of Cancer and Autoimmune Disorders
CN103261217B (en) 2010-11-11 2017-04-26 港大科桥有限公司 Soluble pd-1 variants, fusion constructs, and uses thereof
JP6130307B2 (en) 2011-03-17 2017-05-17 ザ ユニバーシティ オブ バーミンガム Redirected immunotherapy
WO2012135408A1 (en) 2011-03-31 2012-10-04 Merck Sharp & Dohme Corp. Stable formulations of antibodies to human programmed death receptor pd-1 and related treatments
CN107519486B (en) 2011-06-24 2021-06-11 台北荣民总医院 Method for enhancing immune response in the treatment of infectious and malignant diseases
ES2893855T3 (en) 2011-08-11 2022-02-10 Ono Pharmaceutical Co Therapeutic agent for autoimmune diseases comprising PD-1 agonist
CA2845536A1 (en) * 2011-08-15 2013-02-21 Amplimmune, Inc. Anti-b7-h4 antibodies and their uses
GB201203442D0 (en) 2012-02-28 2012-04-11 Univ Birmingham Immunotherapeutic molecules and uses
WO2013169693A1 (en) 2012-05-09 2013-11-14 Bristol-Myers Squibb Company Methods of treating cancer using an il-21 polypeptide and an anti-pd-1 antibody
SG10201700698WA (en) 2012-05-15 2017-02-27 Bristol Myers Squibb Co Cancer immunotherapy by disrupting pd-1/pd-l1 signaling
EP2852410A4 (en) 2012-05-21 2015-12-23 Mitchell S Felder Treatment of cancer by manipulating the immune system
WO2013174997A1 (en) 2012-05-25 2013-11-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of refractory haematological malignancies
ES2742379T3 (en) 2012-05-31 2020-02-14 Hoffmann La Roche Cancer treatment procedures using PD-1 axis binding antagonists and VEGF antagonists
PT2992017T (en) * 2013-05-02 2021-01-29 Anaptysbio Inc Antibodies directed against programmed death-1 (pd-1)
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
AU2015237864B2 (en) 2014-03-24 2020-12-03 Cancer Research Technology Limited Modified antibodies containing modified IgG2 domains which elicit agonist or antagonistic properties and use thereof
CA2951278A1 (en) 2014-06-19 2015-12-23 Regeneron Pharmaceuticals, Inc. Non-human animals having a humanized programmed cell death 1 gene
ES2847311T3 (en) 2014-08-05 2021-08-02 MabQuest SA Immunological reagents that bind to PD-1
US20170209574A1 (en) 2014-10-03 2017-07-27 Novartis Ag Combination therapies
JP6827415B2 (en) 2014-10-31 2021-02-10 メレオ バイオファーマ 5 インコーポレイテッド Combination therapy for the treatment of the disease
US20170340733A1 (en) 2014-12-19 2017-11-30 Novartis Ag Combination therapies
MA41218A (en) 2014-12-19 2017-10-24 Advaxis Inc LISTERIA VACCINE COMBINATION WITH ANTI-OX40 OR ANTI-GITR ANTIBODIES
WO2016106159A1 (en) 2014-12-22 2016-06-30 Enumeral Biomedical Holding, Inc. Anti-pd-1 antibodies
AU2015374385B2 (en) 2014-12-31 2020-08-06 Checkmate Pharmaceuticals, Inc. Combination tumor immunotherapy
TWI591075B (en) 2015-01-16 2017-07-11 中央研究院 Peptide core-based multi-arm linkers
EP3666794A1 (en) * 2016-11-01 2020-06-17 AnaptysBio, Inc. Antibodies directed against programmed death- 1 (pd-1)

Also Published As

Publication number Publication date
KR20160034247A (en) 2016-03-29
RU2015151505A3 (en) 2018-03-20
CN105339389B (en) 2021-04-27
JP2016523516A (en) 2016-08-12
US20230331843A1 (en) 2023-10-19
US20160075783A1 (en) 2016-03-17
US20180051082A1 (en) 2018-02-22
NL301120I1 (en) 2021-08-18
AU2014259719B2 (en) 2019-10-03
HUE053069T2 (en) 2021-06-28
AU2014259719A1 (en) 2015-12-17
SI2992017T1 (en) 2021-04-30
MX2015015037A (en) 2016-07-08
RU2015151505A (en) 2017-06-07
NZ714537A (en) 2021-09-24
LTPA2021011I1 (en) 2021-10-25
EP2992017A4 (en) 2017-03-29
JP7009543B2 (en) 2022-01-25
HRP20210122T1 (en) 2021-04-16
US10738117B2 (en) 2020-08-11
WO2014179664A2 (en) 2014-11-06
EP2992017B1 (en) 2020-11-18
JP6742903B2 (en) 2020-08-19
ES2845609T3 (en) 2021-07-27
NL301120I2 (en) 2021-11-15
WO2014179664A3 (en) 2015-02-19
DK2992017T3 (en) 2021-01-25
US9815897B2 (en) 2017-11-14
EP3770176A1 (en) 2021-01-27
PT2992017T (en) 2021-01-29
RU2723050C2 (en) 2020-06-08
SG11201508528TA (en) 2015-11-27
CY1123935T1 (en) 2022-03-24
JP2020115874A (en) 2020-08-06
CN105339389A (en) 2016-02-17
AU2019240593B2 (en) 2021-06-10
CA2910278C (en) 2021-09-28
EP2992017A2 (en) 2016-03-09
CA2910278A1 (en) 2014-11-06
PL2992017T3 (en) 2021-09-06
HK1219743A1 (en) 2017-04-13
HUS2100031I1 (en) 2021-09-28
KR102243062B1 (en) 2021-04-21
AU2019240593A1 (en) 2019-10-24
BR112015026823A2 (en) 2017-09-05
NZ753073A (en) 2021-11-26
LT2992017T (en) 2021-02-25
RS61400B1 (en) 2021-02-26

Similar Documents

Publication Publication Date Title
US20230331843A1 (en) Antibodies Directed Against Programmed Death-1(PD-1)
US20210095026A1 (en) Antibodies Directed Against Lymphocyte Activation Gene 3 (LAG-3)
AU2016242964B2 (en) Antibodies directed against T cell immunoglobulin and Mucin Protein 3 (TIM-3)
WO2014172448A2 (en) Antibodies directed against activin receptor type ii (actrii)
JP2022534803A (en) PD-1 agonists and methods of using them
TW201825512A (en) Antibodies directed against programmed death- 1 (pd-1)
NZ714537B2 (en) Antibodies directed against programmed death-1 (pd-1)
NZ753073B2 (en) Antibodies directed against programmed death-1 (PD-1)
EA040513B1 (en) ANTIBODIES DIRECTED AGAINST LYMPHOCYTE ACTIVATION GENE-3 (LAG-3)

Legal Events

Date Code Title Description
AS Assignment

Owner name: ANAPTYSBIO, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KING, DAVID J.;KEHRY, MARILYN;SIGNING DATES FROM 20160128 TO 20160129;REEL/FRAME:053326/0444

STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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