US20210380702A1 - Human antibodies to pd-l1 - Google Patents

Human antibodies to pd-l1 Download PDF

Info

Publication number
US20210380702A1
US20210380702A1 US17/399,829 US202117399829A US2021380702A1 US 20210380702 A1 US20210380702 A1 US 20210380702A1 US 202117399829 A US202117399829 A US 202117399829A US 2021380702 A1 US2021380702 A1 US 2021380702A1
Authority
US
United States
Prior art keywords
antigen
antibody
binding
antibodies
amino acid
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
US17/399,829
Inventor
Nicholas J. Papadopoulos
Andrew J. Murphy
Gavin Thurston
Ella Ioffe
Elena Burova
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.)
Regeneron Pharmaceuticals Inc
Original Assignee
Regeneron Pharmaceuticals 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 Regeneron Pharmaceuticals Inc filed Critical Regeneron Pharmaceuticals Inc
Priority to US17/399,829 priority Critical patent/US20210380702A1/en
Publication of US20210380702A1 publication Critical patent/US20210380702A1/en
Assigned to REGENERON PHARMACEUTICALS, INC. reassignment REGENERON PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MURPHY, ANDREW J., BUROVA, Elena, IOFFE, ELLA, THURSTON, GAVIN, PAPADOPOULOS, NICHOLAS J.
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
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • 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
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins 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
    • C07K2317/75Agonist effect on antigen
    • 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

Definitions

  • the present invention is related to human antibodies and antigen-binding fragments of human antibodies that specifically bind to the immunomodulatory receptor ligand programmed death-ligand 1 (PD-L1), and therapeutic and diagnostic methods of using those antibodies.
  • PD-L1 immunomodulatory receptor ligand programmed death-ligand 1
  • PD-L1 Programmed death-ligand 1
  • B7-H1 or CD274 is a 290 amino acid protein receptor ligand expressed widely on both lymphoid and non-lymphoid tissues such as CD4 and CD8 T-cells, macrophage lineage cells, peripheral tissues as well as on tumor cells, and virally-infected cells (Dong et al 1999, Nature Med.).
  • PD-L1 binds to receptors PD-1 and B7-1 which belong to the CD28/CTLA-4 (cytotoxic T lymphocyte antigen)/ICOS (inducible co-stimulator) family of T-cell co-inhibitory receptors (Chen et al 2013, Nature Rev. Immunol.
  • PD-L1 binding to PD-1 or B7-1 results in decreased T-cell proliferation and cytokine secretion, compromising humoral and cellular immune responses in diseases such as cancer, and viral infection.
  • PD-L1 The expression of PD-L1 on tumor cells and virally-infected cells is exploited by tumors and chronic viral infections to evade immune response.
  • PD-L1 is expressed on a wide variety of tumors and studies on animal models have shown that PD-L1 on tumors inhibits T-cell activation and lysis of tumor cells and may lead to increased death of tumor-specific T-cells.
  • chronic viral infections PD-L1 expressed on virally-infected cells binds to PD-1 on virus-specific T-cells and these T-cells become “exhausted” with loss of effector functions and proliferative capacity (Freeman 2008, PNAS 105: 10275-10276).
  • the PD-1: PD-L1 system also plays an important role in induced T-regulatory (Treg) cell development and in sustaining Treg function (Francisco et al 2010, Immunol. Rev. 236: 219-242).
  • PD-L1 plays an important role in tumor immunity and infectious immunity, it is an ideal target for immunotherapy.
  • Monoclonal antibodies to PD-L1 are known in the art and have been described, for example, in US Patent/Publication Nos. 7943742, 8383796, 8217149, 20090055944, 20120003056, 20130034559, 20130045200, 20130045201, 20130045202, and in WO2007005874, WO2011066389, WO2010077634, EP1907424, and EP1899379.
  • the present invention provides antibodies and antigen-binding fragments thereof that bind PD-L1.
  • the antibodies of the present invention are useful, inter alia, for targeting cells expressing PD-L1 such as cancer cells or virally-infected cells, and for modulating PD-L1 activity.
  • the antibodies of the invention are useful for inhibiting or neutralizing PD-L1 activity and for stimulating T cell activation, e.g., under circumstances where T cell-mediated killing is beneficial or desirable.
  • the anti-PD-L1 antibodies of the invention may be included as part of a multi-specific antigen-binding molecule, for example, to modulate the immune response and/or to target the antibodies to a specific cell type, such as a tumor cell, or a virally infected cell.
  • the antibodies are useful in treating a disease or disorder such as cancer and viral infection.
  • the antibodies of the invention can be full-length (for example, an IgG1 or IgG4 antibody) or may comprise only an antigen-binding portion (for example, a Fab, F(ab′) 2 or scFv fragment), and may be modified to affect functionality, e.g., to eliminate residual effector functions (Reddy et al., 2000, J. Immunol. 164:1925-1933).
  • the antibodies may be bispecific.
  • the present invention provides isolated monoclonal antibodies or antigen-binding fragments thereof that bind specifically to PD-L1.
  • Exemplary anti-PD-L1 antibodies of the present invention are listed in Tables 1 and 2 herein.
  • Table 1 sets forth the amino acid sequence identifiers of the heavy chain variable regions (HCVRs), light chain variable regions (LCVRs), heavy chain complementarity determining regions (HCDR1, HCDR2 and HCDR3), and light chain complementarity determining regions (LCDR1, LCDR2 and LCDR3) of the exemplary anti-PD-L1 antibodies.
  • Table 2 sets forth the nucleic acid sequence identifiers of the HCVRs, LCVRs, HCDR1, HCDR2 HCDR3, LCDR1, LCDR2 and LCDR3 of the exemplary anti-PD-L1 antibodies.
  • the present invention provides antibodies, or antigen-binding fragments thereof, comprising an HCVR comprising an amino acid sequence selected from any of the HCVR amino acid sequences listed in Table 1, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • the present invention also provides antibodies, or antigen-binding fragments thereof, comprising an LCVR comprising an amino acid sequence selected from any of the LCVR amino acid sequences listed in Table 1, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • the present invention also provides antibodies, or antigen-binding fragments thereof, comprising an HCVR and an LCVR amino acid sequence pair (HCVR/LCVR) comprising any of the HCVR amino acid sequences listed in Table 1 paired with any of the LCVR amino acid sequences listed in Table 1.
  • the present invention provides antibodies, or antigen-binding fragments thereof, comprising an HCVR/LCVR amino acid sequence pair contained within any of the exemplary anti-PD-L1 antibodies listed in Table 1.
  • the HCVR/LCVR amino acid sequence pair is selected from the group consisting of SEQ ID NOs: 2/10, 18/26, 34/42, 50/58, 66/74, 82/90, 98/106, 114/122, 130/138, 146/154, 162/170, 178/170, 186/194, 202/210, 218/226, 234/242, 250/258, 266/274, 282/274, 290/274, 298/274, 306/274, 314/274, 322/274, 330/274, and 338/274.
  • the HCVR/LCVR amino acid sequence pair is selected from one of SEQ ID NOs: 82/90 (e.g., H2M8314N), 162/170 (e.g., H2M8718N), 306/274 (e.g., H1H9364P2), and 314/274 (e.g., H1H9373P2).
  • SEQ ID NOs: 82/90 e.g., H2M8314N
  • 162/170 e.g., H2M8718N
  • 306/274 e.g., H1H9364P2
  • 314/274 e.g., H1H9373P2
  • the HCVR/LCVR amino acid sequence pair is selected from one of SEQ ID NOs: 98/106 (e.g., H2M8316N), 146/154 (e.g., H2M8323N), 290/274 (e.g., H1H9351P2), and 330/274 (e.g., H1H9387P2).
  • SEQ ID NOs: 98/106 e.g., H2M8316N
  • 146/154 e.g., H2M8323N
  • 290/274 e.g., H1H9351P2
  • 330/274 e.g., H1H9387P2
  • the present invention also provides antibodies, or antigen-binding fragments thereof, comprising a heavy chain CDR1 (HCDR1) comprising an amino acid sequence selected from any of the HCDR1 amino acid sequences listed in Table 1 or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity.
  • HCDR1 heavy chain CDR1
  • the present invention also provides antibodies, or antigen-binding fragments thereof, comprising a heavy chain CDR2 (HCDR2) comprising an amino acid sequence selected from any of the HCDR2 amino acid sequences listed in Table 1 or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity.
  • HCDR2 heavy chain CDR2
  • the present invention also provides antibodies, or antigen-binding fragments thereof, comprising a heavy chain CDR3 (HCDR3) comprising an amino acid sequence selected from any of the HCDR3 amino acid sequences listed in Table 1 or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity.
  • HCDR3 heavy chain CDR3
  • the present invention also provides antibodies, or antigen-binding fragments thereof, comprising a light chain CDR1 (LCDR1) comprising an amino acid sequence selected from any of the LCDR1 amino acid sequences listed in Table 1 or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity.
  • LCDR1 light chain CDR1
  • the present invention also provides antibodies, or antigen-binding fragments thereof, comprising a light chain CDR2 (LCDR2) comprising an amino acid sequence selected from any of the LCDR2 amino acid sequences listed in Table 1 or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity.
  • LCDR2 light chain CDR2
  • the present invention also provides antibodies, or antigen-binding fragments thereof, comprising a light chain CDR3 (LCDR3) comprising an amino acid sequence selected from any of the LCDR3 amino acid sequences listed in Table 1 or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity.
  • LCDR3 light chain CDR3
  • the present invention also provides antibodies, or antigen-binding fragments thereof, comprising an HCDR3 and an LCDR3 amino acid sequence pair (HCDR3/LCDR3) comprising any of the HCDR3 amino acid sequences listed in Table 1 paired with any of the LCDR3 amino acid sequences listed in Table 1.
  • the present invention provides antibodies, or antigen-binding fragments thereof, comprising an HCDR3/LCDR3 amino acid sequence pair contained within any of the exemplary anti-PD-L1 antibodies listed in Table 1.
  • the HCDR3/LCDR3 amino acid sequence pair is selected from the group consisting of SEQ ID NOs: 88/96 (e.g., H2M8314N), 168/176 (e.g., H2M8718N), 312/280 (e.g., H1H9364P2), and 320/280 (e.g., H1H9373P2).
  • the HCDR3/LCDR3 amino acid sequence pair is selected from the group consisting of SEQ ID NOs: 104/112 (e.g., H2M8316N), 152/160 (e.g., H2M8323N), 296/280 (e.g., H1H9351P2), and 336/280 (e.g., H1H9387P2).
  • SEQ ID NOs: 104/112 e.g., H2M8316N
  • 152/160 e.g., H2M8323N
  • 296/280 e.g., H1H9351P2
  • 336/280 e.g., H1H9387P2
  • the present invention also provides antibodies, or antigen-binding fragments thereof, comprising a set of six CDRs HCDR1-HCDR2-HCDR3-LCDR1-LCDR2-LCDR3) contained within any of the exemplary anti-PD-L1 antibodies listed in Table 1.
  • the HCDR1-HCDR2-HCDR3-LCDR1-LCDR2-LCDR3 amino acid sequence set is selected from the group consisting of SEQ ID NOs: 84-86-88-92-94-96 (e.g., H2M8314N); 164-166-168-172-174-176 (e.g., H2M8718N); 308-310-312-276-278-280 (e.g., H1H9364P2); and 316-318-320-276-278-280 (e.g., H1H9373P2).
  • the HCDR1-HCDR2-HCDR3-LCDR1-LCDR2-LCDR3 amino acid sequence set is selected from the group consisting of SEQ ID NOs: 100-102-104-108-110-112 (e.g., H2M8316N); 148-150-152-156-158-160 (e.g., H2M8323N); 292-294-296-276-278-280 (e.g., H1H9351P2); and 332-334-336-276-278-280 (e.g., H1H9387P2).
  • the present invention provides antibodies, or antigen-binding fragments thereof, comprising a set of six CDRs HCDR1-HCDR2-HCDR3-LCDR1-LCDR2-LCDR3) contained within an HCVR/LCVR amino acid sequence pair as defined by any of the exemplary anti-PD-L1 antibodies listed in Table 1.
  • the present invention includes antibodies, or antigen-binding fragments thereof, comprising the HCDR1-HCDR2-HCDR3-LCDR1-LCDR2-LCDR3 amino acid sequences set contained within an HCVR/LCVR amino acid sequence pair selected from the group consisting of SEQ ID NOs: 82/90 (e.g., H2M8314N), 98/106 (e.g., H2M8316N), 146/154 (e.g., H2M8323N), 162/170 (e.g., H2M8718N), 290/274 (e.g., H1H9351P2), 306/274 (e.g., H1H9364P2), 314/274 (e.g., H1H9373P2) and 330/274 (e.g., H1H9387P2).
  • SEQ ID NOs: 82/90 e.g., H2M8314N
  • 98/106 e.g., H2M8316N
  • CDRs within HCVR and LCVR amino acid sequences are well known in the art and can be used to identify CDRs within the specified HCVR and/or LCVR amino acid sequences disclosed herein.
  • Exemplary conventions that can be used to identify the boundaries of CDRs include, e.g., the Kabat definition, the Chothia definition, and the AbM definition.
  • the Kabat definition is based on sequence variability
  • the Chothia definition is based on the location of the structural loop regions
  • the AbM definition is a compromise between the Kabat and Chothia approaches. See, e.g., Kabat, “Sequences of Proteins of Immunological Interest,” National Institutes of Health, Bethesda, Md.
  • the present invention includes anti-PD-L1 antibodies having a modified glycosylation pattern.
  • modification to remove undesirable glycosylation sites may be useful, or an antibody lacking a fucose moiety present on the oligosaccharide chain, for example, to increase antibody dependent cellular cytotoxicity (ADCC) function (see Shield et al. (2002) JBC 277:26733).
  • ADCC antibody dependent cellular cytotoxicity
  • modification of galactosylation can be made in order to modify complement dependent cytotoxicity (CDC).
  • the present invention also provides for antibodies and antigen-binding fragments thereof that compete for specific binding to PD-L1 with an antibody or antigen-binding fragment thereof comprising the CDRs of a HCVR and the CDRs of a LCVR, wherein the HCVR and LCVR each has an amino acid sequence selected from the HCVR and LCVR sequences listed in Table 1.
  • the present invention also provides isolated antibodies and antigen-binding fragments thereof that block PD-L1 binding to PD-1 or to B7-1.
  • the antibody or antigen-binding fragment thereof that blocks PD-L1 binding to PD-1 or to B7-1 may bind to the same epitope on PD-L1 as PD-1/B7-1 or may bind to a different epitope on PD-L1 as PD-1/B7-1.
  • the antibodies of the invention that block PD-L1 binding to PD-1 or to B7-1 comprise the CDRs of an HCVR having an amino acid sequence selected from the group consisting of HCVR sequences listed in Table 1; and the CDRs of a LCVR having an amino acid sequence selected from the group consisting of LCVR sequences listed in Table 1.
  • the present invention provides antibodies and antigen-binding fragments thereof that do not block PD-L1 binding to PD-1 or to B7-1.
  • the present invention provides isolated antibodies or antigen-binding fragments thereof that bind PD-L1, wherein the antibodies or antigen-binding fragments thereof enhance PD-L1 binding to PD-1 or to B7-1.
  • the isolated antibodies or antigen-binding fragments thereof that enhance PD-L1 binding to PD-1/B7-1 comprise the CDRs of a HCVR, wherein the HCVR has an amino acid sequence selected from the group consisting of SEQ ID NOs: 18, 66, 114, 130, 202, 218, 266, 282, 298, 322 and 338; and the CDRs of a LCVR, wherein the LCVR has an amino acid sequence selected from the group consisting of SEQ ID NOs: 26, 74, 122, 138, 210, 226, and 274.
  • the isolated antibodies or antigen-binding fragments thereof comprise an HCVR/LCVR amino acid sequence pair selected from the group consisting of SEQ ID NOs: 18/26 (e.g., H2M8307N), 66/74 (e.g., H2M8312N), 114/122 (e.g., H2M8317N), 130/138 (e.g., H2M8321N), 202/210 (e.g., H1H9323P), 218/226 (e.g., H1H9327P), 266/274 (e.g., H1H9344P2), 282/274 (e.g., H1H9345P2), 298/274 (e.g., H1H9354P2), 322/274 (e.g., H1H9382P2), and 338/274 (e.g., H1H9396P2).
  • SEQ ID NOs: 18/26 e.g., H2M8307N
  • the present invention also provides antibodies and antigen-binding fragments thereof that bind specifically to PD-L1 from human or other species.
  • the antibodies may bind to human PD-L1 and/or to cynomolgus PD-L1.
  • the present invention also provides antibodies and antigen-binding fragments thereof that cross-compete for binding to PD-L1 with a reference antibody or antigen-binding fragment thereof comprising the CDRs of a HCVR and the CDRs of a LCVR, wherein the HCVR and LCVR each has an amino acid sequence selected from the HCVR and LCVR sequences listed in Table 1.
  • the invention provides an isolated antibody or antigen-binding fragment that has one or more of the following characteristics: (a) blocks the binding of PD-L1 to PD-1 or to B7-1; (b) binds specifically to human PD-L1 and/or cynomolgus PD-L1; (c) inhibits T-cell proliferation in a mixed lymphocyte reaction (MLR) assay; and (d) increases IL-2 and/or interferon-gamma secretion in a MLR assay.
  • MLR mixed lymphocyte reaction
  • the antibody or antigen binding fragment thereof may bind specifically to PD-L1 in an agonist manner, i.e., it may enhance or stimulate PD-L1 binding and/or activity; in other embodiments, the antibody may bind specifically to PD-L1 in an antagonist manner, i.e., it may block PD-L1 from binding to its receptor.
  • the antibodies or antigen-binding fragments of the present invention are bispecific comprising a first binding specificity to PD-L1 and a second binding specificity for a second target epitope.
  • the second target epitope may be another epitope on PD-L1 or on a different protein such as a T-cell co-inhibitor.
  • the target epitope may be on a different cell including e.g., a different T-cell, a B-cell, a tumor cell, an autoimmune tissue cell or a virally infected cell.
  • the present invention provides nucleic acid molecules encoding anti-PD-L1 antibodies or portions thereof.
  • the present invention provides nucleic acid molecules encoding any of the HCVR amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the HCVR nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • the present invention also provides nucleic acid molecules encoding any of the LCVR amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the LCVR nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • the present invention also provides nucleic acid molecules encoding any of the HCDR1 amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the HCDR1 nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • the present invention also provides nucleic acid molecules encoding any of the HCDR2 amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the HCDR2 nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • the present invention also provides nucleic acid molecules encoding any of the HCDR3 amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the HCDR3 nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • the present invention also provides nucleic acid molecules encoding any of the LCDR1 amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the LCDR1 nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • the present invention also provides nucleic acid molecules encoding any of the LCDR2 amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the LCDR2 nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • the present invention also provides nucleic acid molecules encoding any of the LCDR3 amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the LCDR3 nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • the present invention also provides nucleic acid molecules encoding an HCVR, wherein the HCVR comprises a set of three CDRs (i.e., HCDR1-HCDR2-HCDR3), wherein the HCDR1-HCDR2-HCDR3 amino acid sequence set is as defined by any of the exemplary anti-PD-L1 antibodies listed in Table 1.
  • the present invention also provides nucleic acid molecules encoding an LCVR, wherein the LCVR comprises a set of three CDRs (i.e., LCDR1-LCDR2-LCDR3), wherein the LCDR1-LCDR2-LCDR3 amino acid sequence set is as defined by any of the exemplary anti-PD-L1 antibodies listed in Table 1.
  • the LCVR comprises a set of three CDRs (i.e., LCDR1-LCDR2-LCDR3), wherein the LCDR1-LCDR2-LCDR3 amino acid sequence set is as defined by any of the exemplary anti-PD-L1 antibodies listed in Table 1.
  • the present invention also provides nucleic acid molecules encoding both an HCVR and an LCVR, wherein the HCVR comprises an amino acid sequence of any of the HCVR amino acid sequences listed in Table 1, and wherein the LCVR comprises an amino acid sequence of any of the LCVR amino acid sequences listed in Table 1.
  • the nucleic acid molecule comprises a polynucleotide sequence selected from any of the HCVR nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto, and a polynucleotide sequence selected from any of the LCVR nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • the nucleic acid molecule encodes an HCVR and LCVR, wherein the HCVR and LCVR are both derived from the same anti-PD-L1 antibody listed in Table 1.
  • the present invention provides recombinant expression vectors capable of expressing a polypeptide comprising a heavy or light chain variable region of an anti-PD-L1 antibody.
  • the present invention includes recombinant expression vectors comprising any of the nucleic acid molecules mentioned above, i.e., nucleic acid molecules encoding any of the HCVR, LCVR, and/or CDR sequences as set forth in Table 1.
  • host cells into which such vectors have been introduced as well as methods of producing the antibodies or portions thereof by culturing the host cells under conditions permitting production of the antibodies or antibody fragments, and recovering the antibodies and antibody fragments so produced.
  • the present invention provides multi-specific antigen-binding molecules and antigen-binding fragments thereof comprising a first antigen-binding specificity that binds specifically to PD-L1 and a second antigen-binding specificity that binds specifically to an antigen selected from the group consisting of PD-L1, a tumor cell-specific antigen, an infected-cell-specific antigen, and a T-cell co-inhibitor.
  • the first antigen-binding specificity may comprise three CDRs derived from a HCVR with an amino acid sequence selected from the HCVR sequences in Table 1 and three CDRs derived from a LCVR with an amino acid sequence selected from the LCVR sequences in Table 1.
  • the first antigen-binding specificity may comprise an extracellular domain of PD-1 or of B7-1, or a fragment thereof.
  • the second antigen-binding specificity may target an antigen on the same cell as PD-L1 or on a different cell of the same tissue type or of a different tissue type.
  • the multi-specific antigen-binding molecule may bind to a T-cell wherein the first antigen-binding specificity may bind specifically to PD-L1 and the second antigen-binding specificity may bind to a T-cell co-inhibitor on the T-cell.
  • the first antigen-binding specificity binds specifically to PD-L1 on a T-cell and the second antigen-binding specificity is targeted to an antigen/receptor on a B-cell or a macrophage or antigen-presenting cell.
  • the second antigen-binding specificity is directed to an antigen on a tumor cell, or on a cell infected with a virus.
  • the first antigen-binding specificity comprises an extracellular domain of PD-1 and the second antigen-binding specificity binds to a different epitope on PD-L1.
  • the first antigen-binding specificity binds to PD-L1 with a lower affinity, for example, with a K D more than 10 ⁇ 8 M, more than 10 ⁇ 7 M, more than 10 ⁇ 6 M, or more than 10 ⁇ 5 M.
  • the invention provides a pharmaceutical composition comprising a recombinant human antibody or antigen-binding fragment thereof which specifically binds PD-L1 and a pharmaceutically acceptable carrier.
  • the invention features a composition which is a combination of an anti-PD-L1 antibody and a second therapeutic agent.
  • the second therapeutic agent is any agent that is advantageously combined with an anti-PD-L1 antibody.
  • Exemplary agents that may be advantageously combined with an anti-PD-L1 antibody include, without limitation, other agents that bind and/or modulate PD-L1 signaling (including other antibodies or antigen-binding fragments thereof, etc.) and/or agents which do not directly bind PD-L1 but nonetheless modulate immune cell activation. Additional combination therapies and co-formulations involving the anti-PD-L1 antibodies and multi-specific antigen-binding molecules of the present invention are disclosed elsewhere herein.
  • the invention provides methods to modulate the immune response in a subject, the method comprising administering a therapeutically effective amount of an anti-PD-L1 antibody or antigen-binding fragment thereof of the invention to the subject in need thereof.
  • the invention provides methods to enhance the immune response in a subject, the methods comprising administering to the subject an effective amount of an antibody or fragment thereof of the invention that binds PD-L1 and blocks PD-L1 binding to PD-1 or to B7-1.
  • the invention provides a method to stimulate or enhance T-cell activation in a subject, the method comprising administering a blocking antibody or antigen-binding fragment thereof of the invention to the subject in need thereof.
  • the invention provides methods to inhibit a T-regulatory (Treg) cell in a subject, the methods comprising administering a blocking antibody or antigen-binding fragment thereof of the invention to the subject in need thereof.
  • the subject in need thereof may suffer from a disease or disorder such as cancer or viral infection.
  • the present invention provides methods to inhibit T-cell activation, the methods comprising administering an activating antibody or antigen-binding fragment thereof of the invention to a subject in need thereof.
  • the subject in need thereof may suffer from an autoimmune disease.
  • the invention provides therapeutic methods for treating a disease or disorder, for example, cancer or viral infection, in a subject using an anti-PD-L1 antibody or antigen-binding portion of an antibody of the invention, wherein the therapeutic methods comprise administering a therapeutically effective amount of a pharmaceutical composition comprising an antibody or fragment of an antibody of the invention to the subject in need thereof.
  • the disorder treated is any disease or condition which is improved, ameliorated, inhibited or prevented by stimulation or inhibition of PD-L1 binding or activity.
  • the antibody or antigen-binding fragment thereof the invention is administered in combination with a second therapeutic agent to the subject in need thereof.
  • the second therapeutic agent may be selected from the group consisting of an antibody to a T-cell co-inhibitor, an antibody to a tumor cell antigen, an antibody to a T-cell receptor, an antibody to a Fc receptor, an antibody to an epitope on a virally infected cell, an antibody to PD-1, a cytotoxic agent, an anti-cancer drug, an anti-viral drug, an anti-inflammatory drug (e.g., corticosteroids), a VEGF antagonist, and any other drug or therapy known in the art.
  • the second therapeutic agent may be an agent that helps to counteract or reduce any possible side effect(s) associated with an antibody or antigen-binding fragment thereof of the invention, if such side effect(s) should occur.
  • the present invention provides methods for suppressing tumor growth. In certain embodiments, the present invention provides methods to enhance survival of cancer patients.
  • cancer include, but are not limited to, primary and/or recurrent cancer, including brain cancer (e.g., glioblastoma multiforme), lung cancer (e.g., non-small cell lung cancer), squamous cell carcinoma of head and neck, renal cell carcinoma, melanoma, multiple myeloma, prostate cancer, and colon cancer.
  • the methods comprise administering a pharmaceutical composition comprising a therapeutically effective amount of an anti-PD-L1 antibody of the present invention in combination with a second therapeutic agent selected from the group consisting of a vascular endothelial growth factor (VEGF) antagonist (e.g., aflibercept, bevacizumab), an angiopoietin-2 (Ang2) inhibitor (e.g., an anti-Ang2 antibody such as nesvacumab), a lymphocyte activation gene 3 (LAG-3) inhibitor, a cytotoxic T-lymphocyte antigen 4 (CTLA-4) inhibitor (e.g., ipilimumab), a CCR4 inhibitor (e.g., mogamulizumab), a chemotherapeutic agent, and radiation therapy.
  • VEGF vascular endothelial growth factor
  • Ang2 angiopoietin-2
  • LAG-3 lymphocyte activation gene 3
  • CTLA-4 inhibitor e.g., ipilim
  • the antibody or fragment thereof may be administered subcutaneously, intravenously, intradermally, intraperitoneally, orally, intramuscularly, or intracranially.
  • the antibody or fragment thereof may be administered at a dose of about 0.1 mg/kg of body weight to about 100 mg/kg of body weight of the subject.
  • the present invention also includes use of an anti-PD-L1 antibody or antigen-binding fragment thereof of the invention in the manufacture of a medicament for the treatment of a disease or disorder (e.g., cancer, and chronic viral infection) that would benefit from the blockade or enhancement of PD-L1 binding and/or signaling.
  • a disease or disorder e.g., cancer, and chronic viral infection
  • FIG. 1 is a schematic of the luciferase-based PD-L1 bioassay described in Example 8 herein.
  • Panel A Inactive Jurkat cells
  • Panel B Jurkat cells are activated by T-cell receptor (TCR) clustering through the CD3 ⁇ CD20 bispecific antibody
  • Panel C PD-1 activation attenuates response in activated Jurkat cells
  • Panel D Blocking PD-L1 rescues the response in activated Jurkat cells.
  • FIG. 2 illustrates tumor growth and survival results for mice implanted with Colon-26 tumor cells at Day 0 and treated with the indicated combinations of molecules by injection at Days 3, 6, 10, 13 and 19 (“early-treatment tumor model”).
  • the graph depicts tumor volume (in mm 3 ) for the different experimental groups at various time points after implantation. Upward arrows along the X-axis indicate the timing of treatment injections.
  • “mlgG2a” is IgG2 isotype control
  • Fc is human Fc control
  • VEGF Trap” is aflibercept
  • anti-PD-1 is anti-mouse PD-1 clone RPMI-14
  • anti-PD-L1 is an anti-PD-L1 monoclonal antibody as described elsewhere herein.
  • FIG. 3 illustrates tumor growth and survival results for mice implanted with Colon-26 tumor cells at Day 0 and treated with the indicated combinations of molecules by injection at Days 3, 6, 10, 13 and 19 (“early-treatment tumor model”).
  • the graph shows the tumor volume (in mm 3 ) of individual mice in each experimental group at Day 28 after implantation.
  • “mlgG2a” is IgG2 isotype control
  • “Fc” is human Fc control
  • VEGF Trap” is aflibercept
  • anti-PD-1 is anti-mouse PD-1 clone RPMI-14
  • anti-PD-L1 is an anti-PD-L1 monoclonal antibody as described elsewhere herein.
  • the term “PD-L1” refers to programmed death-ligand 1, also known as CD274 and B7H1.
  • the amino acid sequence of full-length PD-L1 is provided in GenBank as accession number NP 054862.1 and is also referred to herein as SEQ ID NO: 351.
  • the term “PD-L1” also includes protein variants of PD-L1 having the amino acid sequence of SEQ ID NOs: 345, 346, 347 or 348.
  • the term “PD-L1” includes recombinant PD-L1 or a fragment thereof.
  • the term also encompasses PD-L1 or a fragment thereof coupled to, for example, histidine tag, mouse or human Fc, or a signal sequence such as ROR1.
  • the term includes sequences exemplified by SEQ ID NOs: 347 or 348, comprising a mouse Fc (mlgG2a) or human Fc (hIgG1) at the C-terminal, coupled to amino acid residues 19-239 of full-length PD-L1 (SEQ ID NO: 351; NP_054862.1).
  • Protein variants as exemplified by SEQ ID NO: 345 comprise a histidine tag at the C-terminal, coupled to amino acid residues 19-239 of NP_054862.1.
  • PD-L1 means human PD-L1.
  • PD-L1 is a 290 amino acid protein with extracellular IgV-like and IgC-like domains (amino acids 19-239 of full length PD-L1), a transmembrane domain and an intracellular domain of approximately 30 amino acids.
  • PD-L1 is constitutively expressed on many cells such as antigen presenting cells (e.g., dendritic cells, macrophages, and B-cells) and on hematopoietic and non-hematopoietic cells (e.g., vascular endothelial cells, pancreatic islets, and sites of immune privilege).
  • antigen presenting cells e.g., dendritic cells, macrophages, and B-cells
  • hematopoietic and non-hematopoietic cells e.g., vascular endothelial cells, pancreatic islets, and sites of immune privilege.
  • PD-L1 is also expressed on a wide variety of tumors, and virally-infected cells and is a component of the immunosuppressive milieu (Ribas 2012, NEJM 366: 2517-2519). PD-L1 binds to one of two T-cell co-inhibitors PD-1 and B7-1.
  • PD-1 refers to the programmed death-1 protein, a T-cell co-inhibitor, also known as CD279.
  • the amino acid sequence of full-length PD-1 is provided in GenBank as accession number NP 005009.2 and is also referred to herein as SEQ ID NO: 352.
  • the term also encompasses PD-1 or a fragment thereof coupled to, for example, histidine tag, mouse or human Fc, or a signal sequence such as ROR1.
  • the term includes sequences exemplified by SEQ ID NOs: 349 or 350, comprising a mouse Fc (mlgG2a) or human Fc (hIgG1) at the C-terminal, coupled to amino acid residues 25-170 of NP_005009.2 with a C93S change.
  • PD-1 is a member of the CD28/CTLA-4/ICOS family of T-cell co-inhibitors.
  • PD-1 is a 288-amino acid protein with an extracellular N-terminal domain which is IgV-like, a transmembrane domain and an intracellular domain containing an immunoreceptor tyrosine-based inhibitory (ITIM) motif and an immunoreceptor tyrosine-based switch (ITSM) motif (Chattopadhyay et al 2009, Immunol. Rev.).
  • ITIM immunoreceptor tyrosine-based inhibitory
  • ITSM immunoreceptor tyrosine-based switch
  • the PD-1 receptor has two ligands, PD-L1 and PD-L2.
  • B7-1 refers to the T-lymphocyte activation antigen, also known as costimulatory factor CD80.
  • B7-1 is a 288 amino acid membrane receptor with an extracellular N-terminal domain which comprises IgV-like (aa 37-138) and IgC-like (aa 154-232) regions, a transmembrane domain (aa 243-263) and a C-terminal intracellular region (aa 263-288).
  • the amino acid sequence of full-length B7-1 is provided in GenBank as accession number NP_005182.1.
  • T-cell co-inhibitor refers to a ligand and/or receptor which modulates the immune response via T-cell activation or suppression.
  • T-cell co-inhibitor also known as T-cell co-signaling molecule, includes, but is not limited to, PD-1, lymphocyte activation gene 3 protein (LAG-3, also known as CD223), cytotoxic T-lymphocyte antigen-4 (CTLA-4), B and T lymphocyte attenuator (BTLA), CD-28, 2B4, LY108, T-cell immunoglobulin and mucin-3 (TIM3), T-cell immunoreceptor with immunoglobulin and ITIM (TIGIT; also known as VSIG9), leucocyte associated immunoglobulin-like receptor 1 (LAIR1; also known as CD305), inducible T-cell costimulator (ICOS; also known as CD278), B7-1 (CD80), and CD160.
  • LAIR1 leucocyte associated immunoglobulin-like receptor 1
  • ITIM
  • antibody is intended to refer to immunoglobulin molecules comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds (i.e., “full antibody molecules”), as well as multimers thereof (e.g. IgM) or antigen-binding fragments thereof.
  • Each heavy chain is comprised of a heavy chain variable region (“HCVR” or “V H ”) and a heavy chain constant region (comprised of domains C H 1, C H 2 and C H 3).
  • Each light chain is comprised of a light chain variable region (“LCVR or “V L ”) and a light chain constant region (CL).
  • V H and V L regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each V H and V L is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the FRs of the antibody (or antigen binding fragment thereof) may be identical to the human germline sequences, or may be naturally or artificially modified.
  • An amino acid consensus sequence may be defined based on a side-by-side analysis of two or more CDRs.
  • CDR residues not contacting antigen can be identified based on previous studies (for example residues H60-H65 in CDRH2 are often not required), from regions of Kabat CDRs lying outside Chothia CDRs, by molecular modeling and/or empirically. If a CDR or residue(s) thereof is omitted, it is usually substituted with an amino acid occupying the corresponding position in another human antibody sequence or a consensus of such sequences. Positions for substitution within CDRs and amino acids to substitute can also be selected empirically. Empirical substitutions can be conservative or non-conservative substitutions.
  • the fully human anti-PD-L1 monoclonal antibodies disclosed herein may comprise one or more amino acid substitutions, insertions and/or deletions in the framework and/or CDR regions of the heavy and light chain variable domains as compared to the corresponding germline sequences. Such mutations can be readily ascertained by comparing the amino acid sequences disclosed herein to germline sequences available from, for example, public antibody sequence databases.
  • the present invention includes antibodies, and antigen-binding fragments thereof, which are derived from any of the amino acid sequences disclosed herein, wherein one or more amino acids within one or more framework and/or CDR regions are mutated to the corresponding residue(s) of the germline sequence from which the antibody was derived, or to the corresponding residue(s) of another human germline sequence, or to a conservative amino acid substitution of the corresponding germline residue(s) (such sequence changes are referred to herein collectively as “germline mutations”).
  • germline mutations such sequence changes are referred to herein collectively as “germline mutations”.
  • all of the framework and/or CDR residues within the V H and/or V L domains are mutated back to the residues found in the original germline sequence from which the antibody was derived.
  • only certain residues are mutated back to the original germline sequence, e.g., only the mutated residues found within the first 8 amino acids of FR1 or within the last 8 amino acids of FR4, or only the mutated residues found within CDR1, CDR2 or CDR3.
  • one or more of the framework and/or CDR residue(s) are mutated to the corresponding residue(s) of a different germline sequence (i.e., a germline sequence that is different from the germline sequence from which the antibody was originally derived).
  • the antibodies of the present invention may contain any combination of two or more germline mutations within the framework and/or CDR regions, e.g., wherein certain individual residues are mutated to the corresponding residue of a particular germline sequence while certain other residues that differ from the original germline sequence are maintained or are mutated to the corresponding residue of a different germline sequence.
  • antibodies and antigen-binding fragments that contain one or more germline mutations can be easily tested for one or more desired property such as, improved binding specificity, increased binding affinity, improved or enhanced antagonistic or agonistic biological properties (as the case may be), reduced immunogenicity, etc.
  • Antibodies and antigen-binding fragments obtained in this general manner are encompassed within the present invention.
  • the present invention also includes fully human anti-PD-L1 monoclonal antibodies comprising variants of any of the HCVR, LCVR, and/or CDR amino acid sequences disclosed herein having one or more conservative substitutions.
  • the present invention includes anti-PD-L1 antibodies having HCVR, LCVR, and/or CDR amino acid sequences with, e.g., 10 or fewer, 8 or fewer, 6 or fewer, 4 or fewer, etc. conservative amino acid substitutions relative to any of the HCVR, LCVR, and/or CDR amino acid sequences disclosed herein.
  • human antibody is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • the human mAbs of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3.
  • the term “human antibody”, as used herein is not intended to include mAbs in which CDR sequences derived from the germline of another mammalian species (e.g., mouse), have been grafted onto human FR sequences.
  • the term includes antibodies recombinantly produced in a non-human mammal, or in cells of a non-human mammal.
  • the term is not intended to include antibodies isolated from or generated in a human subject.
  • multi-specific antigen-binding molecules refers to bispecific, tri-specific or multi-specific antigen-binding molecules, and antigen-binding fragments thereof. Multi-specific antigen-binding molecules may be specific for different epitopes of one target polypeptide or may contain antigen-binding domains specific for epitopes of more than one target polypeptide.
  • a multi-specific antigen-binding molecule can be a single multifunctional polypeptide, or it can be a multimeric complex of two or more polypeptides that are covalently or non-covalently associated with one another.
  • multi-specific antigen-binding molecules includes antibodies of the present invention that may be linked to or co-expressed with another functional molecule, e.g., another peptide or protein.
  • another functional molecule e.g., another peptide or protein.
  • an antibody or fragment thereof can be functionally linked (e.g., by chemical coupling, genetic fusion, non-covalent association or otherwise) to one or more other molecular entities, such as a protein or fragment thereof to produce a bi-specific or a multi-specific antigen-binding molecule with a second binding specificity.
  • the term “multi-specific antigen-binding molecules” also includes bi-specific, tri-specific or multi-specific antibodies or antigen-binding fragments thereof.
  • an antibody of the present invention is functionally linked to another antibody or antigen-binding fragment thereof to produce a bispecific antibody with a second binding specificity. Bispecific and multi-specific antibodies of the present invention are described elsewhere herein.
  • the term “specifically binds,” or “binds specifically to”, or the like, means that an antibody or antigen-binding fragment thereof forms a complex with an antigen that is relatively stable under physiologic conditions. Specific binding can be characterized by an equilibrium dissociation constant of at least about 1 ⁇ 10 ⁇ 8 M or less (e.g., a smaller K D denotes a tighter binding). Methods for determining whether two molecules specifically bind are well known in the art and include, for example, equilibrium dialysis, surface plasmon resonance, and the like. As described herein, antibodies have been identified by surface plasmon resonance, e.g., BIACORETM, which bind specifically to PD-L1. Moreover, multi-specific antibodies that bind to one domain in PD-L1 and one or more additional antigens or a bi-specific that binds to two different regions of PD-L1 are nonetheless considered antibodies that “specifically bind”, as used herein.
  • high affinity antibody refers to those mAbs having a binding affinity to PD-L1, expressed as K D , of at least 10 ⁇ 8 M; preferably 10 ⁇ 9 M; more preferably 10 ⁇ 10 M, even more preferably 10 ⁇ 11 M, even more preferably 10 ⁇ 12 M, as measured by surface plasmon resonance, e.g., BIACORETM or solution-affinity ELISA.
  • slow off rate an antibody that dissociates from PD-L1, with a rate constant of 1 ⁇ 10 ⁇ 3 s ⁇ 1 or less, preferably 1 ⁇ 10 ⁇ 4 s ⁇ 1 or less, as determined by surface plasmon resonance, e.g., BIACORETM.
  • antigen-binding portion of an antibody, “antigen-binding fragment” of an antibody, and the like, as used herein, include any naturally occurring, enzymatically obtainable, synthetic, or genetically engineered polypeptide or glycoprotein that specifically binds an antigen to form a complex.
  • antigen-binding fragment of an antibody, or “antibody fragment”, as used herein, refers to one or more fragments of an antibody that retain the ability to bind to PD-L1.
  • antibody or antibody fragments of the invention may be conjugated to a moiety such a ligand or a therapeutic moiety (“immunoconjugate”), such as an antibiotic, a second anti-PD-L1 antibody, or an antibody to another antigen such a tumor-specific antigen, a virally-infected cell antigen, or a T-cell co-inhibitor, or an immunotoxin, or any other therapeutic moiety useful for treating a disease or condition including e.g., cancer, chronic viral infection or autoimmune disease.
  • a moiety such as an antibiotic, a second anti-PD-L1 antibody, or an antibody to another antigen such a tumor-specific antigen, a virally-infected cell antigen, or a T-cell co-inhibitor, or an immunotoxin, or any other therapeutic moiety useful for treating a disease or condition including e.g., cancer, chronic viral infection or autoimmune disease.
  • an “isolated antibody”, as used herein, is intended to refer to an antibody that is substantially free of other antibodies (Abs) having different antigenic specificities (e.g., an isolated antibody that specifically binds PD-L1, or a fragment thereof, is substantially free of Abs that specifically bind antigens other than PD-L1.
  • blocking antibody or a “neutralizing antibody”, as used herein (or an “antibody that neutralizes PD-L1 activity” or an “antagonist antibody”), is intended to refer to an antibody whose binding to PD-L1 results in inhibition of at least one biological activity of PD-L1.
  • an antibody of the invention may prevent or block PD-L1 binding to PD-1 or to B7-1.
  • an “activating antibody” or an “enhancing antibody”, as used herein (or an “agonist antibody”) is intended to refer to an antibody whose binding to PD-L1 results in increasing or stimulating at least one biological activity of PD-L1.
  • an antibody of the invention may increase or enhance PD-L1 binding to PD-1 or to B7-1.
  • surface plasmon resonance refers to an optical phenomenon that allows for the analysis of real-time biomolecular interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIACORETM system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.).
  • K D is intended to refer to the equilibrium dissociation constant of a particular antibody-antigen interaction.
  • epitope refers to an antigenic determinant that interacts with a specific antigen binding site in the variable region of an antibody molecule known as a paratope.
  • a single antigen may have more than one epitope. Thus, different antibodies may bind to different areas on an antigen and may have different biological effects.
  • epitope also refers to a site on an antigen to which B and/or T cells respond. It also refers to a region of an antigen that is bound by an antibody.
  • Epitopes may be defined as structural or functional. Functional epitopes are generally a subset of the structural epitopes and have those residues that directly contribute to the affinity of the interaction.
  • Epitopes may also be conformational, that is, composed of non-linear amino acids.
  • epitopes may include determinants that are chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl groups, or sulfonyl groups, and, in certain embodiments, may have specific three-dimensional structural characteristics, and/or specific charge characteristics.
  • nucleic acid or fragment thereof indicates that, when optimally aligned with appropriate nucleotide insertions or deletions with another nucleic acid (or its complementary strand), there is nucleotide sequence identity in at least about 90%, and more preferably at least about 95%, 96%, 97%, 98% or 99% of the nucleotide bases, as measured by any well-known algorithm of sequence identity, such as FASTA, BLAST or GAP, as discussed below.
  • a nucleic acid molecule having substantial identity to a reference nucleic acid molecule may, in certain instances, encode a polypeptide having the same or substantially similar amino acid sequence as the polypeptide encoded by the reference nucleic acid molecule.
  • the term “substantial similarity” or “substantially similar” means that two peptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least 90% sequence identity, even more preferably at least 95%, 98% or 99% sequence identity.
  • residue positions, which are not identical differ by conservative amino acid substitutions.
  • a “conservative amino acid substitution” is one in which an amino acid residue is substituted by another amino acid residue having a side chain (R group) with similar chemical properties (e.g., charge or hydrophobicity). In general, a conservative amino acid substitution will not substantially change the functional properties of a protein.
  • the percent or degree of similarity may be adjusted upwards to correct for the conservative nature of the substitution. Means for making this adjustment are well known to those of skill in the art. See, e.g., Pearson (1994) Methods Mol. Biol. 24: 307-331, which is herein incorporated by reference.
  • Examples of groups of amino acids that have side chains with similar chemical properties include 1) aliphatic side chains: glycine, alanine, valine, leucine and isoleucine; 2) aliphatic-hydroxyl side chains: serine and threonine; 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; 5) basic side chains: lysine, arginine, and histidine; 6) acidic side chains: aspartate and glutamate, and 7) sulfur-containing side chains: cysteine and methionine.
  • Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-glutamine.
  • a conservative replacement is any change having a positive value in the PAM250 log-likelihood matrix disclosed in Gonnet et al. (1992) Science 256: 1443 45, herein incorporated by reference.
  • a “moderately conservative” replacement is any change having a nonnegative value in the PAM250 log-likelihood matrix.
  • Sequence similarity for polypeptides is typically measured using sequence analysis software. Protein analysis software matches similar sequences using measures of similarity assigned to various substitutions, deletions and other modifications, including conservative amino acid substitutions.
  • GCG software contains programs such as GAP and BESTFIT which can be used with default parameters to determine sequence homology or sequence identity between closely related polypeptides, such as homologous polypeptides from different species of organisms or between a wild type protein and a mutein thereof. See, e.g., GCG Version 6.1. Polypeptide sequences also can be compared using FASTA with default or recommended parameters; a program in GCG Version 6.1.
  • FASTA e.g., FASTA2 and FASTA3
  • FASTA2 and FASTA3 provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (Pearson (2000) supra).
  • Another preferred algorithm when comparing a sequence of the invention to a database containing a large number of sequences from different organisms is the computer program BLAST, especially BLASTP or TBLASTN, using default parameters. See, e.g., Altschul et al. (1990) J. Mol. Biol. 215: 403-410 and (1997) Nucleic Acids Res. 25:3389-3402, each of which is herein incorporated by reference.
  • terapéuticaally effective amount is meant an amount that produces the desired effect for which it is administered. The exact amount will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, for example, Lloyd (1999) The Art, Science and Technology of Pharmaceutical Compounding).
  • the term “subject” refers to an animal, preferably a mammal, in need of amelioration, prevention and/or treatment of a disease or disorder such as chronic viral infection, cancer or autoimmune disease.
  • anti-cancer drug means any agent useful to treat cancer including, but not limited to, cytotoxins and agents such as antimetabolites, alkylating agents, anthracyclines, antibiotics, antimitotic agents, procarbazine, hydroxyurea, asparaginase, corticosteroids, mytotane (O,P′-(DDD)), interferons and radioactive agents.
  • cytotoxins agents such as antimetabolites, alkylating agents, anthracyclines, antibiotics, antimitotic agents, procarbazine, hydroxyurea, asparaginase, corticosteroids, mytotane (O,P′-(DDD)), interferons and radioactive agents.
  • a cytotoxin or cytotoxic agent means any agent that is detrimental to cells.
  • Taxol® paclitaxel
  • cytochalasin B gramicidin D
  • ethidium bromide emetine
  • mitomycin etoposide
  • tenoposide vincristine
  • vinbiastine coichicin
  • doxorubicin daunorubicin
  • dihydroxy anthracin dione mitoxantrone
  • mithramycin actinomycin D
  • 1-dehydrotestosterone glucocorticoids
  • procaine tetracaine
  • lidocaine lidocaine
  • propranolol puromycin and analogs or homologs thereof.
  • anti-viral drug refers to any drug or therapy used to treat, prevent, or ameliorate a viral infection in a host subject.
  • anti-viral drug includes, but is not limited to zidovudine, lamivudine, abacavir, ribavirin, lopinavir, efavirenz, cobicistat, tenofovir, rilpivirine, analgesics and corticosteroids.
  • the viral infections include long-term or chronic infections caused by viruses including, but not limited to, human immunodeficiency virus (HIV), hepatitis B virus (HBV), hepatitis C virus (HCV), human papilloma virus (HPV), lymphocytic choriomeningitis virus (LCMV), and simian immunodeficiency virus (SIV).
  • viruses including, but not limited to, human immunodeficiency virus (HIV), hepatitis B virus (HBV), hepatitis C virus (HCV), human papilloma virus (HPV), lymphocytic choriomeningitis virus (LCMV), and simian immunodeficiency virus (SIV).
  • viruses including, but not limited to, human immunodeficiency virus (HIV), hepatitis B virus (HBV), hepatitis C virus (HCV), human papilloma virus (HPV), lymphocytic choriomeningitis
  • the antibodies and antigen-binding fragments of the present invention specifically bind to PD-L1 and modulate the interaction of PD-L1 with PD-1 or with B7-1.
  • the anti-PD-L1 antibodies may bind to PD-L1 with high affinity or with low affinity.
  • the antibodies of the present invention are blocking antibodies wherein the antibodies bind to PD-L1 and block the interaction of PD-L1 with PD-1 or with B7-1.
  • the blocking antibodies of the invention block the binding of PD-L1 to PD-1 or to B7-1 and/or stimulate or enhance T-cell activation.
  • the blocking antibodies are useful for stimulating or enhancing the immune response and/or for treating a subject suffering from cancer, or a chronic viral infection.
  • the antibodies when administered to a subject in need thereof may reduce the chronic infection by a virus such as HIV, LCMV or HBV in the subject. They may be used to inhibit the growth of tumor cells in a subject. They may be used alone or as adjunct therapy with other therapeutic moieties or modalities known in the art for treating cancer, or viral infection.
  • the anti-PD-L1 antibodies that bind to PD-L1 with a low affinity are used as multi-specific antigen-binding molecules wherein the first binding specificity binds to PD-L1 with a low affinity and the second binding specificity binds to an antigen selected from the group consisting of a different epitope of PD-L1, a T-cell co-inhibitor such as PD-1, a tumor specific antigen and an infected-cell-specific antigen.
  • the antibodies of the present invention are agonist antibodies, wherein the antibodies bind to PD-L1 and enhance the interaction of PD-L1 and PD-1/B7-1.
  • the activating antibodies enhance binding of PD-L1 to PD-1 or to B7-1 and/or inhibit or suppress T-cell activation.
  • the activating antibodies of the present invention may be useful for inhibiting the immune response in a subject and/or for treating autoimmune disease.
  • the anti-PD-L1 antibodies are multi-specific antigen-binding molecules, wherein they comprise a first binding specificity to PD-L1 and a second binding specificity to an antigen selected from the group consisting of a different epitope of PD-L1, a T-cell co-inhibitor such as PD-1, a tumor specific antigen and an infected-cell-specific antigen.
  • the first binding specificity binds to PD-L1 with low affinity, e.g., with a K D of 10 ⁇ 8 M, 10 ⁇ 7 M or more.
  • Certain anti-PD-L1 antibodies of the present invention are able to bind to and neutralize the activity of PD-L1, as determined by in vitro or in vivo assays.
  • the ability of the antibodies of the invention to bind to and neutralize the activity of PD-L1 may be measured using any standard method known to those skilled in the art, including binding assays, or activity assays, as described herein.
  • Example 3 Non-limiting, exemplary in vitro assays for measuring binding activity are illustrated in Example 3, herein.
  • Example 3 the binding affinities and kinetic constants of human anti-PD-L1 antibodies for human PD-L1 and cynomolgus PD-L1 were determined by surface plasmon resonance and the measurements were conducted on a T200 Biacore instrument.
  • blocking assays were used to determine the ability of the anti-PD-L1 antibodies to block PD-L1-binding ability of PD-1 or to B7-1 in vitro.
  • Example 6 blocking assays were used to determine cross-competition between different anti-PD-L1 antibodies.
  • Example 7 describes the binding of the antibodies to cells overexpressing PD-L1.
  • Example 8 a luciferase assay was used to determine the ability of anti-PD-L1 antibodies to antagonize PD-1/PD-L1 signaling in T-cells.
  • the antibodies of the present invention are able to enhance or stimulate T-cell activation in vitro and in a subject with cancer or in a subject infected with a virus such as LCMV.
  • the antibodies of the present invention are used in combination with a second therapeutic agent, such as an antibody to a tumor-specific antigen or a T-cell co-inhibitor, to enhance the immune response and inhibit tumor growth in a subject.
  • the agonist antibodies of the invention able to enhance PD-L1 binding to PD-1 or to B7-1 and may inhibit T-cell activation in vitro and/or in a subject with autoimmune disease.
  • the antibodies specific for PD-L1 may contain no additional labels or moieties, or they may contain an N-terminal or C-terminal label or moiety.
  • the label or moiety is biotin.
  • the location of a label may determine the orientation of the peptide relative to the surface upon which the peptide is bound. For example, if a surface is coated with avidin, a peptide containing an N-terminal biotin will be oriented such that the C-terminal portion of the peptide will be distal to the surface.
  • the label may be a radionuclide, a fluorescent dye or a MRI-detectable label. In certain embodiments, such labeled antibodies may be used in diagnostic assays including imaging assays.
  • antibody shall be understood to encompass antibody molecules comprising two immunoglobulin heavy chains and two immunoglobulin light chains (i.e., “full antibody molecules”) as well as antigen-binding fragments thereof.
  • full antibody molecules immunoglobulin heavy chains and two immunoglobulin light chains
  • antigen-binding portion of an antibody, “antigen-binding fragment” of an antibody, and the like, as used herein, include any naturally occurring, enzymatically obtainable, synthetic, or genetically engineered polypeptide or glycoprotein that specifically binds an antigen to form a complex.
  • antigen-binding fragment of an antibody, or “antibody fragment”, as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to PD-L1.
  • an antibody fragment may include a Fab fragment, a F(ab′) 2 fragment, a Fv fragment, a dAb fragment, a fragment containing a CDR, or an isolated CDR.
  • the term “antigen-binding fragment” refers to a polypeptide or fragment thereof of a multi-specific antigen-binding molecule.
  • the term “antigen-binding fragment” includes, e.g., an extracellular domain of PD-1 which binds specifically to PD-L1.
  • Antigen-binding fragments of an antibody may be derived, e.g., from full antibody molecules using any suitable standard techniques such as proteolytic digestion or recombinant genetic engineering techniques involving the manipulation and expression of DNA encoding antibody variable and (optionally) constant domains.
  • DNA is known and/or is readily available from, e.g., commercial sources, DNA libraries (including, e.g., phage-antibody libraries), or can be synthesized.
  • the DNA may be sequenced and manipulated chemically or by using molecular biology techniques, for example, to arrange one or more variable and/or constant domains into a suitable configuration, or to introduce codons, create cysteine residues, modify, add or delete amino acids, etc.
  • Non-limiting examples of antigen-binding fragments include: (i) Fab fragments; (ii) F(ab′)2 fragments; (iii) Fd fragments; (iv) Fv fragments; (v) single-chain Fv (scFv) molecules; (vi) dAb fragments; and (vii) minimal recognition units consisting of the amino acid residues that mimic the hypervariable region of an antibody (e.g., an isolated complementarity determining region (CDR) such as a CDR3 peptide), or a constrained FR3-CDR3-FR4 peptide.
  • CDR complementarity determining region
  • engineered molecules such as domain-specific antibodies, single domain antibodies, domain-deleted antibodies, chimeric antibodies, CDR-grafted antibodies, diabodies, triabodies, tetrabodies, minibodies, nanobodies (e.g. monovalent nanobodies, bivalent nanobodies, etc.), small modular immunopharmaceuticals (SMIPs), and shark variable IgNAR domains, are also encompassed within the expression “antigen-binding fragment,” as used herein.
  • SMIPs small modular immunopharmaceuticals
  • an antigen-binding fragment of an antibody will typically comprise at least one variable domain.
  • the variable domain may be of any size or amino acid composition and will generally comprise at least one CDR, which is adjacent to or in frame with one or more framework sequences.
  • the V H and V L domains may be situated relative to one another in any suitable arrangement.
  • the variable region may be dimeric and contain V H -V H , V H -V L or V L -V L dimers.
  • the antigen-binding fragment of an antibody may contain a monomeric V H or V L domain.
  • an antigen-binding fragment of an antibody may contain at least one variable domain covalently linked to at least one constant domain.
  • variable and constant domains that may be found within an antigen-binding fragment of an antibody of the present invention include: (i) V H —C H 1; (ii) V H —C H 2; (iii) V H —C H 3; (iv) V H —C H 1-C H 2; (v) V H —C H 1-C H 2-C H 3; (vi) V H —C H 2-C H 3, V H —CL, V L —C H 1; (ix) V L -C H 2, (x) V L -C H 3; (xi) V L -C H 1-C H 2; (xii) V L -C H 1-C H 2-C H 3; V L -C H 2-C H 3; and (xiv) V L -CL.
  • variable and constant domains may be either directly linked to one another or may be linked by a full or partial hinge or linker region.
  • a hinge region may consist of at least 2 (e.g., 5, 10, 15, 20, 40, 60 or more) amino acids, which result in a flexible or semi-flexible linkage between adjacent variable and/or constant domains in a single polypeptide molecule.
  • an antigen-binding fragment of an antibody of the present invention may comprise a homo-dimer or hetero-dimer (or other multimer) of any of the variable and constant domain configurations listed above in non-covalent association with one another and/or with one or more monomeric V H or V L domain (e.g., by disulfide bond(s)).
  • antigen-binding fragments may be mono-specific or multi-specific (e.g., bi-specific).
  • a multi-specific antigen-binding fragment of an antibody will typically comprise at least two different variable domains, wherein each variable domain is capable of specifically binding to a separate antigen or to a different epitope on the same antigen.
  • Any multi-specific antibody format, including the exemplary bi-specific antibody formats disclosed herein, may be adapted for use in the context of an antigen-binding fragment of an antibody of the present invention using routine techniques available in the art.
  • an immunogen comprising any one of the following can be used to generate antibodies to PD-L1.
  • the antibodies of the invention are obtained from mice immunized with a primary immunogen, such as a full length PD-L1 [See GenBank accession number NP_054862.1 (SEQ ID NO: 351)] or with a recombinant form of PD-L1 or modified human PD-L1 fragments (SEQ ID NOs: 345, 347 or 348) or with modified cynomolgus PD-L1 fragments (SEQ ID NO: 346), followed by immunization with a secondary immunogen, or with an immunogenically active fragment of PD-L1.
  • a primary immunogen such as a full length PD-L1 [See GenBank accession number NP_054862.1 (SEQ ID NO: 351)] or with a recombinant form of PD-L1 or modified human PD-L1 fragments (SEQ ID NOs: 345, 347 or 34
  • the immunogen may be a peptide from the N terminal or C terminal end of PD-L1.
  • the immunogen is the extracellular IgV-like and/or IgC-like domain of PD-L1.
  • the immunogen is a fragment of PD-L1 that ranges from about amino acid residues 19-239 of SEQ ID NO: 351 (NP_054862.1).
  • the immunogen may be a recombinant PD-L1 peptide expressed in E. coli or in any other eukaryotic or mammalian cells such as Chinese hamster ovary (CHO) cells.
  • antibodies that bind specifically to PD-L1 may be prepared using fragments of the above-noted regions, or peptides that extend beyond the designated regions by about 5 to about 20 amino acid residues from either, or both, the N or C terminal ends of the regions described herein. In certain embodiments, any combination of the above-noted regions or fragments thereof may be used in the preparation of PD-L1 specific antibodies.
  • VELOCIMMUNE® technology see, for example, U.S. Pat. No. 6,596,541, Regeneron Pharmaceuticals, VELOCIMMUNE®
  • VELOCIMMUNE® technology involves generation of a transgenic mouse having a genome comprising human heavy and light chain variable regions operably linked to endogenous mouse constant region loci such that the mouse produces an antibody comprising a human variable region and a mouse constant region in response to antigenic stimulation.
  • the DNA encoding the variable regions of the heavy and light chains of the antibody are isolated and operably linked to DNA encoding the human heavy and light chain constant regions.
  • the DNA is then expressed in a cell capable of expressing the fully human antibody.
  • the anti-PD-L1 antibodies and antibody fragments of the present invention encompass proteins having amino acid sequences that vary from those of the described antibodies, but that retain the ability to bind PD-L1.
  • Such variant antibodies and antibody fragments comprise one or more additions, deletions, or substitutions of amino acids when compared to parent sequence, but exhibit biological activity that is essentially equivalent to that of the described antibodies.
  • the antibody-encoding DNA sequences of the present invention encompass sequences that comprise one or more additions, deletions, or substitutions of nucleotides when compared to the disclosed sequence, but that encode an antibody or antibody fragment that is essentially bioequivalent to an antibody or antibody fragment of the invention.
  • Two antigen-binding proteins, or antibodies are considered bioequivalent if, for example, they are pharmaceutical equivalents or pharmaceutical alternatives whose rate and extent of absorption do not show a significant difference when administered at the same molar dose under similar experimental conditions, either single dose or multiple doses.
  • Some antibodies will be considered equivalents or pharmaceutical alternatives if they are equivalent in the extent of their absorption but not in their rate of absorption and yet may be considered bioequivalent because such differences in the rate of absorption are intentional and are reflected in the labeling, are not essential to the attainment of effective body drug concentrations on, e.g., chronic use, and are considered medically insignificant for the particular drug product studied.
  • two antigen-binding proteins are bioequivalent if there are no clinically meaningful differences in their safety, purity, or potency.
  • two antigen-binding proteins are bioequivalent if a patient can be switched one or more times between the reference product and the biological product without an expected increase in the risk of adverse effects, including a clinically significant change in immunogenicity, or diminished effectiveness, as compared to continued therapy without such switching.
  • two antigen-binding proteins are bioequivalent if they both act by a common mechanism or mechanisms of action for the condition or conditions of use, to the extent that such mechanisms are known.
  • Bioequivalence may be demonstrated by in vivo and/or in vitro methods.
  • Bioequivalence measures include, e.g., (a) an in vivo test in humans or other mammals, in which the concentration of the antibody or its metabolites is measured in blood, plasma, serum, or other biological fluid as a function of time; (b) an in vitro test that has been correlated with and is reasonably predictive of human in vivo bioavailability data; (c) an in vivo test in humans or other mammals in which the appropriate acute pharmacological effect of the antibody (or its target) is measured as a function of time; and (d) in a well-controlled clinical trial that establishes safety, efficacy, or bioavailability or bioequivalence of an antibody.
  • Bioequivalent variants of the antibodies of the invention may be constructed by, for example, making various substitutions of residues or sequences or deleting terminal or internal residues or sequences not needed for biological activity.
  • cysteine residues not essential for biological activity can be deleted or replaced with other amino acids to prevent formation of unnecessary or incorrect intramolecular disulfide bridges upon renaturation.
  • bioequivalent antibodies may include antibody variants comprising amino acid changes, which modify the glycosylation characteristics of the antibodies, e.g., mutations that eliminate or remove glycosylation.
  • anti-PD-L1 antibodies comprising an Fc domain comprising one or more mutations which enhance or diminish antibody binding to the FcRn receptor, e.g., at acidic pH as compared to neutral pH.
  • the present invention includes anti-PD-L1 antibodies comprising a mutation in the C H 2 or a C H 3 region of the Fc domain, wherein the mutation(s) increases the affinity of the Fc domain to FcRn in an acidic environment (e.g., in an endosome where pH ranges from about 5.5 to about 6.0).
  • Such mutations may result in an increase in serum half-life of the antibody when administered to an animal.
  • Non-limiting examples of such Fc modifications include, e.g., a modification at position 250 (e.g., E or Q); 250 and 428 (e.g., L or F); 252 (e.g., L/Y/F/W or T), 254 (e.g., S or T), and 256 (e.g., S/R/Q/E/D or T); or a modification at position 428 and/or 433 (e.g., H/L/R/S/P/Q or K) and/or 434 (e.g., A, W, H, F or Y [N434A, N434W, N434H, N434F or N434Y]); or a modification at position 250 and/or 428; or a modification at position 307 or 308 (e.g., 308F, V308F), and 434.
  • a modification at position 250 e.g., E or Q
  • 250 and 428 e.g., L or F
  • the modification comprises a 428L (e.g., M428L) and 434S (e.g., N434S) modification; a 428L, 259I (e.g., V259I), and 308F (e.g., V308F) modification; a 433K (e.g., H433K) and a 434 (e.g., 434Y) modification; a 252, 254, and 256 (e.g., 252Y, 254T, and 256E) modification; a 250Q and 428L modification (e.g., T250Q and M428L); and a 307 and/or 308 modification (e.g., 308F or 308P).
  • the modification comprises a 265A (e.g., D265A) and/or a 297A (e.g., N297A) modification.
  • the present invention includes anti-PD-L1 antibodies comprising an Fc domain comprising one or more pairs or groups of mutations selected from the group consisting of: 250Q and 248L (e.g., T250Q and M248L); 252Y, 254T and 256E (e.g., M252Y, S254T and T256E); 428L and 434S (e.g., M428L and N434S); 257I and 311I (e.g., P257I and Q311I); 257I and 434H (e.g., P257I and N434H); 376V and 434H (e.g., D376V and N434H); 307A, 380A and 434A (e.g., T307A, E380A and N434A); and 433K and 434F (e.g., H433K and N434F).
  • 250Q and 248L e.g., T250Q and M
  • the present invention includes anti-PD-L1 antibodies comprising an Fc domain comprising a S108P mutation in the hinge region of IgG4 to promote dimer stabilization. All possible combinations of the foregoing Fc domain mutations, and other mutations within the antibody variable domains disclosed herein, are contemplated within the scope of the present invention.
  • the present invention also includes anti-PD-L1 antibodies comprising a chimeric heavy chain constant (C H ) region, wherein the chimeric C H region comprises segments derived from the C H regions of more than one immunoglobulin isotype.
  • the antibodies of the invention may comprise a chimeric C H region comprising part or all of a C H 2 domain derived from a human IgG1, human IgG2 or human IgG4 molecule, combined with part or all of a C H 3 domain derived from a human IgG1, human IgG2 or human IgG4 molecule.
  • the antibodies of the invention comprise a chimeric C H region having a chimeric hinge region.
  • a chimeric hinge may comprise an “upper hinge” amino acid sequence (amino acid residues from positions 216 to 227 according to EU numbering) derived from a human IgG1, a human IgG2 or a human IgG4 hinge region, combined with a “lower hinge” sequence (amino acid residues from positions 228 to 236 according to EU numbering) derived from a human IgG1, a human IgG2 or a human IgG4 hinge region.
  • the chimeric hinge region comprises amino acid residues derived from a human IgG1 or a human IgG4 upper hinge and amino acid residues derived from a human IgG2 lower hinge.
  • An antibody comprising a chimeric C H region as described herein may, in certain embodiments, exhibit modified Fc effector functions without adversely affecting the therapeutic or pharmacokinetic properties of the antibody. (See, e.g., U.S. Ser. No. 14/170,166, filed Jan. 31, 2014, the disclosure of which is hereby incorporated by reference in its entirety).
  • the antibodies of the present invention function by binding to PD-L1.
  • the present invention includes anti-PD-L1 antibodies and antigen-binding fragments thereof that bind soluble monomeric or dimeric PD-L1 molecules with high affinity.
  • the present invention includes antibodies and antigen-binding fragments of antibodies that bind monomeric PD-L1 (e.g., at 25° C. or at 37° C.) with a K D of less than about 318 pM as measured by surface plasmon resonance, e.g., using the assay format as defined in Example 3 herein.
  • the antibodies or antigen-binding fragments thereof bind monomeric PD-L1 with a K D of less than about 300 pM, less than about 250 pM, less than about 150 pM, less than about 100 pM, or less than about 50 pM, as measured by surface plasmon resonance, e.g., using the assay format as defined in Example 3 herein, or a substantially similar assay.
  • the present invention also includes antibodies and antigen-binding fragments thereof that bind dimeric PD-L1 (e.g., at 25° C. or at 37° C.) with a K D of less than about 15 pM as measured by surface plasmon resonance, e.g., using the assay format as defined in Example 3 herein.
  • the antibodies or antigen-binding fragments thereof bind dimeric PD-L1 with a K D of less than about 12 pM, less than about 10 pM, less than about 8 pM, or less than about 5 pM, as measured by surface plasmon resonance, e.g., using the assay format as defined in Example 3 herein, or a substantially similar assay.
  • the present invention also includes antibodies or antigen-binding fragments thereof that bind cynomolgus ( Macaca fascicularis ) PD-L1 (e.g., at 25° C. or at 37° C.) with a K D of less than about 28 nM as measured by surface plasmon resonance, e.g., using the assay format as defined in Example 3 herein.
  • the antibodies or antigen-binding fragments thereof bind cynomolgus PD-L1 with a K D of less than about 25 nM, less than about 20 nM, less than about 15 nM, less than about 10 nM, or less than about 5 nM, as measured by surface plasmon resonance, e.g., using the assay format as defined in Example 3 herein, or a substantially similar assay.
  • the present invention also includes antibodies and antigen-binding fragments thereof that bind PD-L1 with a dissociative half-life (t1 ⁇ 2) of greater than about 1 minute as measured by surface plasmon resonance at 25° C. or 37° C., e.g., using an assay format as defined in Example 3 herein, or a substantially similar assay.
  • t1 ⁇ 2 dissociative half-life
  • the antibodies or antigen-binding fragments of the present invention bind PD-L1 with a t % of greater than about 5 minutes, greater than about 10 minutes, greater than about 30 minutes, greater than about 50 minutes, greater than about 60 minutes, greater than about 70 minutes, greater than about 80 minutes, greater than about 90 minutes, greater than about 100 minutes, greater than about 200 minutes, greater than about 300 minutes, greater than about 400 minutes, greater than about 500 minutes, greater than about 600 minutes, greater than about 700 minutes, or greater than about 800 minutes, as measured by surface plasmon resonance at 25° C. or 37° C., e.g., using an assay format as defined in Example 3 herein (e.g., mAb-capture or antigen-capture format), or a substantially similar assay.
  • an assay format as defined in Example 3 herein (e.g., mAb-capture or antigen-capture format), or a substantially similar assay.
  • the present invention also includes antibodies or antigen-binding fragments thereof that block PD-L1 binding to PD-1 with an IC 50 of less than about 770 pM as determined using a ELISA-based immunoassay assay, e.g., as shown in Example 4, or a substantially similar assay.
  • the present invention also includes antibodies or antigen-binding fragments thereof that block PD-L1 binding to B7-1 with an IC 50 of less than about 10 nM as determined using a ELISA-based immunoassay assay, e.g., as shown in Example 4, or a substantially similar assay.
  • the present invention also includes antibodies and antigen-binding fragments thereof that bind to PD-L1 and enhance the binding of PD-L1 to PD-1 or to B7-1.
  • the antibodies of the present invention may bind to the extracellular domain of PD-L1 or to a fragment of the domain. In some embodiments, the antibodies of the present invention may bind to more than one domain (cross-reactive antibodies). In certain embodiments, the antibodies of the present invention may bind to an epitope located in the extracellular domain comprising amino acid residues 19-239 of NP_054862.1 (SEQ ID NO: 351). In some embodiments, the antibodies may bind to an epitope comprising one or more amino acids selected from the group consisting of amino acid residues 1-221 of SEQ ID NOs: 345-348, or 353.
  • the antibodies of the present invention may function by blocking or inhibiting the PD-1-binding or the B7-1-binding activity associated with PD-L1 by binding to any other region or fragment of the full length protein, the amino acid sequence of which is shown in SEQ ID NO: 351.
  • the antibodies may attenuate or modulate the interaction between PD-L1 and PD-1/B7-1.
  • the antibodies of the present invention may be bi-specific antibodies.
  • the bi-specific antibodies of the invention may bind one epitope in one domain and may also bind a second epitope in a different domain of PD-L1.
  • the bi-specific antibodies of the invention may bind two different epitopes in the same domain.
  • the multi-specific antigen-binding molecule comprises a first antigen-binding specificity wherein the first binding specificity comprises the extracellular domain or fragment thereof of PD-1; and a second antigen-binding specificity to another epitope of PD-L1.
  • the multi-specific antigen-binding molecule comprises a first antigen-binding specificity wherein the first binding specificity comprises the extracellular domain or fragment thereof of B7-1; and a second antigen-binding specificity to another epitope of PD-L1.
  • the invention provides a fully human monoclonal antibody or antigen-binding fragment thereof that binds to PD-L1, wherein the antibody or fragment thereof exhibits one or more of the following characteristics: (i) comprises a HCVR having an amino acid sequence selected from the group consisting of SEQ ID NO: 2, 18, 34, 50, 66, 82, 98, 114, 130, 146, 162, 178, 186, 202, 218, 234, 250, 258, 266, 274, 282, 290, 298, 306, 314, 322, 330 and 338, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; (ii) comprises a LCVR having an amino acid sequence selected from the group consisting of SEQ ID NO: 10, 26, 42, 58, 74, 90, 106, 122, 138, 154, 170, 194, 210, 226, 242, 258, and 274, or a substantially similar sequence thereof having at least 90%, at least 95%
  • the invention provides a fully human monoclonal antibody or antigen-binding fragment thereof that blocks PD-L1 binding to PD-1 or to B7-1, wherein the antibody or fragment thereof exhibits one or more of the following characteristics: (i) comprises a HCVR having an amino acid sequence selected from the group consisting of SEQ ID NO: 82, 98, 146, 162, 290, 306, 314, and 330, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; (ii) comprises a LCVR having an amino acid sequence selected from the group consisting of SEQ ID NO: 90, 106, 154, 170, and 274, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; (iii) comprises a HCDR3 domain having an amino acid sequence selected from the group consisting of SEQ ID NO: 88, 104, 152, 168, 296, 312, 320,
  • the antibodies of the present invention may possess one or more of the aforementioned biological characteristics, or any combinations thereof. Other biological characteristics of the antibodies of the present invention will be evident to a person of ordinary skill in the art from a review of the present disclosure including the working Examples herein.
  • the anti-PD-L1 antibodies bind to human PD-L1 but not to PD-L1 from other species.
  • the anti-PD-L1 antibodies of the invention in certain embodiments, bind to human PD-L1 and to PD-L1 from one or more non-human species.
  • the anti-PD-L1 antibodies of the invention may bind to human PD-L1 and may bind or not bind, as the case may be, to one or more of mouse, rat, guinea pig, hamster, gerbil, pig, cat, dog, rabbit, goat, sheep, cow, horse, camel, cynomolgus, marmoset, rhesus or chimpanzee PD-L1.
  • the anti-PD-L1 antibodies of the invention may bind to human and cynomolgus PD-L1 with the same affinities or with different affinities.
  • the present invention includes anti-PD-L1 antibodies which interact with one or more amino acids found within one or more domains of the PD-L1 molecule including, e.g., extracellular (IgV-like) domain, the extracellular IgC-like domain, a transmembrane domain, and an intracellular domain.
  • the epitope to which the antibodies bind may consist of a single contiguous sequence of 3 or more (e.g., 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more) amino acids located within any of the aforementioned domains of the PD-L1 molecule (e.g. a linear epitope in a domain).
  • the epitope may consist of a plurality of non-contiguous amino acids (or amino acid sequences) located within either or both of the aforementioned domains of the PD-L1 molecule (e.g. a conformational epitope).
  • exemplary techniques can be used to determine whether an antibody “interacts with one or more amino acids” within a polypeptide or protein.
  • Exemplary techniques include, for example, routine cross-blocking assays, such as that described in Antibodies, Harlow and Lane (Cold Spring Harbor Press, Cold Spring Harbor, N.Y.).
  • Other methods include alanine scanning mutational analysis, peptide blot analysis (Reineke (2004) Methods Mol. Biol. 248: 443-63), peptide cleavage analysis crystallographic studies and NMR analysis.
  • methods such as epitope excision, epitope extraction and chemical modification of antigens can be employed (Tomer (2000) Prot. Sci. 9: 487-496).
  • the hydrogen/deuterium exchange method involves deuterium-labeling the protein of interest, followed by binding the antibody to the deuterium-labeled protein. Next, the protein/antibody complex is transferred to water and exchangeable protons within amino acids that are protected by the antibody complex undergo deuterium-to-hydrogen back-exchange at a slower rate than exchangeable protons within amino acids that are not part of the interface. As a result, amino acids that form part of the protein/antibody interface may retain deuterium and therefore exhibit relatively higher mass compared to amino acids not included in the interface.
  • the target protein After dissociation of the antibody, the target protein is subjected to protease cleavage and mass spectrometry analysis, thereby revealing the deuterium-labeled residues which correspond to the specific amino acids with which the antibody interacts. See, e.g., Ehring (1999) Analytical Biochemistry 267: 252-259; Engen and Smith (2001) Anal. Chem. 73: 256A-265A.
  • epitope refers to a site on an antigen to which B and/or T cells respond.
  • B-cell epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents, whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents.
  • An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation.
  • MAP Modification-Assisted Profiling
  • SAP Antigen Structure-based Antibody Profiling
  • mAbs monoclonal antibodies
  • Each category may reflect a unique epitope either distinctly different from or partially overlapping with epitope represented by another category. This technology allows rapid filtering of genetically identical antibodies, such that characterization can be focused on genetically distinct antibodies.
  • MAP may facilitate identification of rare hybridoma clones that produce mAbs having the desired characteristics.
  • MAP may be used to sort the antibodies of the invention into groups of antibodies binding different epitopes.
  • the anti-PD-L1 antibodies or antigen-binding fragments thereof bind an epitope within any one or more of the regions exemplified in PD-L1, either in natural form, as exemplified in SEQ ID NO: 351, or recombinantly produced, as exemplified in SEQ ID NOS: 345-348, or to a fragment thereof.
  • the antibodies of the invention bind to an extracellular region comprising one or more amino acids selected from the group consisting of amino acid residues 19-239 of PD-L1.
  • the antibodies of the invention bind to a region comprising one or more amino acids selected from the group consisting of amino acid residues 1-221 of cynomolgus PD-L1, as exemplified by SEQ ID NO: 346.
  • the antibodies of the invention interact with at least one amino acid sequence selected from the group consisting of amino acid residues ranging from about position 19 to about position 130 of SEQ ID NO: 351; or amino acid residues ranging from about position 130 to about position 153 of SEQ ID NO: 351; or amino acid residues ranging from about position 153 to about position 210 of SEQ ID NO: 351; or to amino acid residues ranging from about position 210 to about position 239 of SEQ ID NO: 351.
  • amino acid sequence selected from the group consisting of amino acid residues ranging from about position 19 to about position 130 of SEQ ID NO: 351; or amino acid residues ranging from about position 130 to about position 153 of SEQ ID NO: 351; or amino acid residues ranging from about position 153 to about position 210 of SEQ ID NO: 351; or to amino acid residues ranging from about position 210 to about position 239 of SEQ ID NO: 351.
  • the present invention includes anti-PD-L1 antibodies that bind to the same epitope, or a portion of the epitope, as any of the specific exemplary antibodies described herein in Table 1, or an antibody having the CDR sequences of any of the exemplary antibodies described in Table 1.
  • the present invention also includes anti-PD-L1 antibodies that compete for binding to PD-L1 or a PD-L1 fragment with any of the specific exemplary antibodies described herein in Table 1, or an antibody having the CDR sequences of any of the exemplary antibodies described in Table 1.
  • the present invention includes anti-PD-L1 antibodies that cross-compete for binding to PD-L1 with one or more antibodies as defined in Example 6 herein (e.g., H2aM8309N, H1H9329P, H1H9336P, H2aM8314N, H2aM8316N, H2AM8718N, H1H9387P2, H1H9351P2, H1H9364P2, H1H9373P2, and H2aM8306N).
  • H2aM8309N, H1H9329P, H1H9336P, H2aM8314N, H2aM8316N, H2AM8718N, H1H9387P2, H1H9351P2, H1H9364P2, H1H9373P2, and H2aM8306N e.g., H2aM8309N, H1H9329P, H1H9336P, H2aM8314N, H2aM8316N, H2AM87
  • the present invention also includes anti-PD-L1 antibodies that cross-compete for binding to PD-L1 with one or more antibodies as defined in Example 6 herein (e.g., H1H9396P2, H2aM8317N, H2aM8321N, H1H9323P, H1H9382P2, H1H9344P2, H1H9345P2 and H1H9354P2).
  • anti-PD-L1 antibodies that cross-compete for binding to PD-L1 with one or more antibodies as defined in Example 6 herein (e.g., H1H9396P2, H2aM8317N, H2aM8321N, H1H9323P, H1H9382P2, H1H9344P2, H1H9345P2 and H1H9354P2).
  • test antibody may bind to the same epitope as the epitope bound by the reference anti-PD-L1 antibody of the invention.
  • the above-described binding methodology is performed in two orientations: In a first orientation, the reference antibody is allowed to bind to a PD-L1 protein under saturating conditions followed by assessment of binding of the test antibody to the PD-L1 molecule. In a second orientation, the test antibody is allowed to bind to a PD-L1 molecule under saturating conditions followed by assessment of binding of the reference antibody to the PD-L1 molecule. If, in both orientations, only the first (saturating) antibody is capable of binding to the PD-L1 molecule, then it is concluded that the test antibody and the reference antibody compete for binding to PD-L1.
  • an antibody that competes for binding with a reference antibody may not necessarily bind to the identical epitope as the reference antibody, but may sterically block binding of the reference antibody by binding an overlapping or adjacent epitope.
  • Two antibodies bind to the same or overlapping epitope if each competitively inhibits (blocks) binding of the other to the antigen. That is, a 1-, 5-, 10-, 20- or 100-fold excess of one antibody inhibits binding of the other by at least 50% but preferably 75%, 90% or even 99% as measured in a competitive binding assay (see, e.g., Junghans et al., Cancer Res. 1990 50:1495-1502).
  • two antibodies have the same epitope if essentially all amino acid mutations in the antigen that reduce or eliminate binding of one antibody reduce or eliminate binding of the other.
  • Two antibodies have overlapping epitopes if some amino acid mutations that reduce or eliminate binding of one antibody reduce or eliminate binding of the other.
  • Additional routine experimentation e.g., peptide mutation and binding analyses
  • peptide mutation and binding analyses can then be carried out to confirm whether the observed lack of binding of the test antibody is in fact due to binding to the same epitope as the reference antibody or if steric blocking (or another phenomenon) is responsible for the lack of observed binding.
  • steric blocking or another phenomenon
  • this sort can be performed using ELISA, RIA, surface plasmon resonance, flow cytometry or any other quantitative or qualitative antibody-binding assay available in the art.
  • the invention encompasses a human anti-PD-L1 monoclonal antibody conjugated to a therapeutic moiety (“immunoconjugate”), such as a cytotoxin or a chemotherapeutic agent to treat cancer.
  • a therapeutic moiety such as a cytotoxin or a chemotherapeutic agent to treat cancer.
  • immunoconjugate refers to an antibody which is chemically or biologically linked to a cytotoxin, a radioactive agent, a cytokine, an interferon, a target or reporter moiety, an enzyme, a toxin, a peptide or protein or a therapeutic agent.
  • the antibody may be linked to the cytotoxin, radioactive agent, cytokine, interferon, target or reporter moiety, enzyme, toxin, peptide or therapeutic agent at any location along the molecule so long as it is able to bind its target.
  • immunoconjugates include antibody drug conjugates and antibody-toxin fusion proteins.
  • the agent may be a second different antibody to PD-L1.
  • the antibody may be conjugated to an agent specific for a tumor cell or a virally infected cell.
  • the type of therapeutic moiety that may be conjugated to the anti-PD-L1 antibody and will take into account the condition to be treated and the desired therapeutic effect to be achieved. Examples of suitable agents for forming immunoconjugates are known in the art; see for example, WO 05/103081.
  • the antibodies of the present invention may be mono-specific, bi-specific, or multi-specific. Multi-specific antibodies may be specific for different epitopes of one target polypeptide or may contain antigen-binding domains specific for more than one target polypeptide. See, e.g., Tutt et al., 1991, J. Immunol. 147:60-69; Kufer et al., 2004, Trends Biotechnol. 22:238-244.
  • the present invention includes multi-specific antigen-binding molecules or antigen-binding fragments thereof wherein one antigen-binding specificity of an immunoglobulin is specific for an epitope within the extracellular domain of PD-L1 (e.g., in the IgV-like region), or a fragment thereof, and the other antigen-binding specificity of the immunoglobulin is specific for binding to a different epitope in the extracellular domain of PD-L1 (e.g., in the IgC-like region), or a second therapeutic target, or is conjugated to a therapeutic moiety.
  • the first antigen-binding specificity may comprise PD-1 or B7-1 or a fragment thereof.
  • the first antigen-binding specificity that binds to PD-L1 comprises the extracellular domain of PD-1.
  • one antigen-binding specificity of an immunoglobulin is specific for an epitope within amino acid residues 19-239 of PD-L1 (SEQ ID NO: 351) or a fragment thereof, and the other specificity of the immunoglobulin is specific for a second target antigen.
  • the second target antigen may be on the same cell as PD-L1 or on a different cell.
  • the second target cell is on an immune cell other than a T-cell such as a B-cell, antigen-presenting cell, monocyte, macrophage, or dendritic cell.
  • the second target antigen may be present on a tumor cell or on a virally infected cell.
  • the invention provides multi-specific antigen-binding molecules or antigen-binding fragments thereof comprising a first antigen-binding specificity that binds to PD-L1 and a second antigen-binding specificity that binds specifically to a target antigen on a tumor cell.
  • the tumor-specific antigen is one of CA9, CA125, melanoma-associated antigen (MAGE), carcinoembryonic antigen (CEA), vimentin, tumor-M2-PK, prostate-specific antigen (PSA), MART-1, or CA19-9.
  • Non-limiting examples of other specific tumor-associated antigens include, e.g., AFP, ALK, BAGE proteins, ⁇ -catenin, brc-abl, BRCA1, BORIS, carbonic anhydrase IX, caspase-8, CCR5, CD19, CD20, CD30, CD40, CDK4, CTLA4, cyclin-B1, CYP1B1, EGFR, EGFRvIII, ErbB2/Her2, ErbB3, ErbB4, ETV6-AML, EpCAM, EphA2, Fra-1, FOLR1, GAGE proteins (e.g., GAGE-1, -2), GD2, GD3, GloboH, glypican-3, GM3, gp100, Her2, HLA/B-raf, HLA/k-ras, HLA/MAGE-A3, hTERT, LMP2, MAGE proteins (e.g., MAGE-1, -2,-3, -4, -6, and -12), HLA-A2, MART
  • the second antigen-binding specificity binds to a tumor antigen that is present on tumor cells specific to, but not limited to, renal cell carcinoma, prostate cancer, colorectal cancer, melanoma, breast cancer, kidney cancer, ovarian cancer, and pancreatic cancer.
  • the antibodies of the invention in this aspect, may inhibit the activity of PD-L1.
  • the invention provides multi-specific antigen-binding molecules or antigen-binding fragments thereof wherein the second antigen-binding specificity binds to an antigen specific to a virally-infected cell.
  • the second antigen-binding specificity binds to an antigen specific to a cell infected with a virus selected from the group consisting of HIV, HBV, HCV, HPV, LCMV and SIV.
  • the invention provides multi-specific antigen-binding molecules or antigen-binding fragments thereof comprising a first antigen-binding specificity that binds to PD-L1 and a second antigen-binding specificity that binds to a T-cell co-inhibitor such as PD-1, LAG-3, TIM3, B7-1, CTLA-4, BTLA, CD-28, 2B4, LY108, TIGIT, LAIR1, ICOS and CD160.
  • a T-cell co-inhibitor such as PD-1, LAG-3, TIM3, B7-1, CTLA-4, BTLA, CD-28, 2B4, LY108, TIGIT, LAIR1, ICOS and CD160.
  • any of the multi-specific antigen-binding molecules, or variants thereof, may be constructed using standard molecular biological techniques (e.g., recombinant DNA and protein expression technology), as will be well known to a person of ordinary skill in the art.
  • PD-L1-specific antibodies are generated in a bi-specific format (a “bi-specific”) in which variable regions binding to distinct domains of PD-L1 are linked together to confer dual-domain specificity within a single binding molecule.
  • bi-specifics may enhance overall PD-L1 inhibitory efficacy through increasing both specificity and binding avidity.
  • Variable regions with specificity for individual domains, (e.g., segments of the N-terminal domain), or that can bind to different regions within one domain are paired on a structural scaffold that allows each region to bind simultaneously to the separate epitopes, or to different regions within one domain.
  • V H heavy chain variable regions
  • V L light chain variable regions
  • V L segment Use of a single V L segment reduces the complexity of the system and thereby simplifies and increases efficiency in cloning, expression, and purification processes used to generate the bi-specific (See, for example, U.S. Ser. No. 13/022,759 and US2010/0331527).
  • antibodies that bind more than one domains and a second target may be prepared in a bi-specific format using techniques described herein, or other techniques known to those skilled in the art.
  • Antibody variable regions binding to distinct regions may be linked together with variable regions that bind to relevant sites on, for example, the extracellular domain of PD-L1, to confer dual-antigen specificity within a single binding molecule.
  • Appropriately designed bi-specifics of this nature serve a dual function.
  • Variable regions with specificity for the extracellular domain are combined with a variable region with specificity for outside the extracellular domain and are paired on a structural scaffold that allows each variable region to bind to the separate antigens.
  • An exemplary bi-specific antibody format that can be used in the context of the present invention involves the use of a first immunoglobulin (Ig) C H 3 domain and a second Ig C H 3 domain, wherein the first and second Ig C H 3 domains differ from one another by at least one amino acid, and wherein at least one amino acid difference reduces binding of the bi-specific antibody to Protein A as compared to a bi-specific antibody lacking the amino acid difference.
  • the first Ig C H 3 domain binds Protein A and the second Ig C H 3 domain contains a mutation that reduces or abolishes Protein A binding such as an H95R modification (by IMGT exon numbering; H435R by EU numbering).
  • the second C H 3 may further comprise a Y96F modification (by IMGT; Y436F by EU). Further modifications that may be found within the second C H 3 include: D16E, L18M, N44S, K52N, V57M, and V821 (by IMGT; D356E, L358M, N384S, K392N, V397M, and V4221 by EU) in the case of IgG1 antibodies; N44S, K52N, and V821 (IMGT; N384S, K392N, and V4221 by EU) in the case of IgG2 antibodies; and Q15R, N44S, K52N, V57M, R69K, E79Q, and V821 (by IMGT; Q355R, N384S, K392N, V397M, R409K, E419Q, and V4221 by EU) in the case of IgG4 antibodies. Variations on the bi-specific antibody format described above are contemplate
  • Bispecific antibodies can also be constructed using peptide/nucleic acid conjugation, e.g., wherein unnatural amino acids with orthogonal chemical reactivity are used to generate site-specific antibody-oligonucleotide conjugates which then self-assemble into multimeric complexes with defined composition, valency and geometry. (See, e.g., Kazane et al., J. Am. Chem. Soc . [Epub: Dec. 4, 2012]).
  • the invention provides therapeutic compositions comprising the anti-PD-L1 antibodies or antigen-binding fragments thereof of the present invention.
  • Therapeutic compositions in accordance with the invention will be administered with suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • a multitude of appropriate formulations can be found in the formulary known to all pharmaceutical chemists: Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa.
  • formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTINTM), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. See also Powell et al. “Compendium of excipients for parenteral formulations” PDA (1998) J Pharm Sci Technol 52:238-311.
  • the dose of antibody may vary depending upon the age and the size of a subject to be administered, target disease, conditions, route of administration, and the like.
  • an antibody of the present invention is used for treating a disease or disorder in an adult patient, or for preventing such a disease, it is advantageous to administer the antibody of the present invention normally at a single dose of about 0.1 to about 100 mg/kg body weight, more preferably about 5 to about 80, about 10 to about 60, or about 20 to about 50 mg/kg body weight.
  • the frequency and the duration of the treatment can be adjusted.
  • the antibody or antigen-binding fragment thereof of the invention can be administered as an initial dose of at least about 0.1 mg to about 800 mg, about 1 to about 500 mg, about 5 to about 300 mg, or about 10 to about 200 mg, to about 100 mg, or to about 50 mg.
  • the initial dose may be followed by administration of a second or a plurality of subsequent doses of the antibody or antigen-binding fragment thereof in an amount that can be approximately the same or less than that of the initial dose, wherein the subsequent doses are separated by at least 1 day to 3 days; at least one week, at least 2 weeks; at least 3 weeks; at least 4 weeks; at least 5 weeks; at least 6 weeks; at least 7 weeks; at least 8 weeks; at least 9 weeks; at least 10 weeks; at least 12 weeks; or at least 14 weeks.
  • Various delivery systems are known and can be used to administer the pharmaceutical composition of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the mutant viruses, receptor mediated endocytosis (see, e.g., Wu et al. (1987) J. Biol. Chem. 262:4429-4432).
  • Methods of introduction include, but are not limited to, intradermal, transdermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural and oral routes.
  • the composition may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • the pharmaceutical composition can be also delivered in a vesicle, in particular a liposome (see, for example, Langer (1990) Science 249:1527-1533).
  • Nanoparticles to deliver the antibodies of the present invention is also contemplated herein.
  • Antibody-conjugated nanoparticles may be used both for therapeutic and diagnostic applications. Antibody-conjugated nanoparticles and methods of preparation and use are described in detail by Arruebo, M., et al. 2009 (“Antibody-conjugated nanoparticles for biomedical applications” in J. Nanomat. Volume 2009, Article ID 439389, 24 pages, doi: 10.1155/2009/439389), incorporated herein by reference. Nanoparticles may be developed and conjugated to antibodies contained in pharmaceutical compositions to target tumor cells or autoimmune tissue cells or virally infected cells. Nanoparticles for drug delivery have also been described in, for example, U.S. Pat. No. 8,257,740, or U.S. Pat. No. 8,246,995, each incorporated herein in its entirety.
  • the pharmaceutical composition can be delivered in a controlled release system.
  • a pump may be used.
  • polymeric materials can be used.
  • a controlled release system can be placed in proximity of the composition's target, thus requiring only a fraction of the systemic dose.
  • the injectable preparations may include dosage forms for intravenous, subcutaneous, intracutaneous, intracranial, intraperitoneal and intramuscular injections, drip infusions, etc. These injectable preparations may be prepared by methods publicly known. For example, the injectable preparations may be prepared, e.g., by dissolving, suspending or emulsifying the antibody or its salt described above in a sterile aqueous medium or an oily medium conventionally used for injections.
  • aqueous medium for injections there are, for example, physiological saline, an isotonic solution containing glucose and other auxiliary agents, etc., which may be used in combination with an appropriate solubilizing agent such as an alcohol (e.g., ethanol), a polyalcohol (e.g., propylene glycol, polyethylene glycol), a nonionic surfactant [e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)], etc.
  • an alcohol e.g., ethanol
  • a polyalcohol e.g., propylene glycol, polyethylene glycol
  • a nonionic surfactant e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil
  • oily medium there are employed, e.g., sesame oil, soybean oil, etc., which may be used in combination with a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc.
  • a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc.
  • a pharmaceutical composition of the present invention can be delivered subcutaneously or intravenously with a standard needle and syringe.
  • a pen delivery device readily has applications in delivering a pharmaceutical composition of the present invention.
  • Such a pen delivery device can be reusable or disposable.
  • a reusable pen delivery device generally utilizes a replaceable cartridge that contains a pharmaceutical composition. Once all of the pharmaceutical composition within the cartridge has been administered and the cartridge is empty, the empty cartridge can readily be discarded and replaced with a new cartridge that contains the pharmaceutical composition. The pen delivery device can then be reused.
  • a disposable pen delivery device there is no replaceable cartridge. Rather, the disposable pen delivery device comes prefilled with the pharmaceutical composition held in a reservoir within the device. Once the reservoir is emptied of the pharmaceutical composition, the entire device is discarded.
  • Numerous reusable pen and autoinjector delivery devices have applications in the subcutaneous delivery of a pharmaceutical composition of the present invention.
  • Examples include, but certainly are not limited to AUTOPENTM (Owen Mumford, Inc., Woodstock, UK), DISETRONICTM pen (Disetronic Medical Systems, Burghdorf, Switzerland), HUMALOG MIX 75/25TM pen, HUMALOGTM pen, HUMALIN 70/30TM pen (Eli Lilly and Co., Indianapolis, Ind.), NOVOPENTM I, II and III (Novo Nordisk, Copenhagen, Denmark), NOVOPEN JUNIORTM (Novo Nordisk, Copenhagen, Denmark), BDTM pen (Becton Dickinson, Franklin Lakes, N.J.), OPTIPENTM, OPTIPEN PROTM, OPTIPEN STARLETTM, and OPTICLIKTM (Sanofi-Aventis, Frankfurt, Germany), to name only a few.
  • AUTOPENTM Owen Mumford, Inc., Woodstock, UK
  • Examples of disposable pen delivery devices having applications in subcutaneous delivery of a pharmaceutical composition of the present invention include, but certainly are not limited to the SOLOSTARTM pen (Sanofi-Aventis), the FLEXPENTM (Novo Nordisk), and the KWIKPENTM (Eli Lilly), the SURECLICKTM Autoinjector (Amgen, Thousand Oaks, Calif.), the PENLETTM (Haselmeier, Stuttgart, Germany), the EPIPEN (Dey, L. P.) and the HUMIRATM Pen (Abbott Labs, Abbott Park, Ill.), to name only a few.
  • the pharmaceutical compositions for oral or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the active ingredients.
  • dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, etc.
  • the amount of the antibody contained is generally about 5 to about 500 mg per dosage form in a unit dose; especially in the form of injection, it is preferred that the antibody is contained in about 5 to about 100 mg and in about 10 to about 250 mg for the other dosage forms.
  • the antibodies of the present invention are useful for the treatment, prevention, and/or amelioration of disease or disorder or condition such as cancer, autoimmune disease or a viral infection and/or for ameliorating at least one symptom associated with such disease, disorder or condition.
  • the antibodies described herein are useful for treating subjects suffering from primary or recurrent cancer, including for example, renal cell carcinoma, prostate cancer, ovarian cancer, kidney cancer, colorectal cancer, gastric cancer, breast cancer, head and neck cancer, non-small-cell lung cancer, brain cancer, multiple myeloma, and melanoma.
  • the antibodies may be used to treat early stage or late-stage symptoms of cancer.
  • an antibody or fragment thereof of the invention may be used to treat metastatic cancer.
  • the antibodies are useful in reducing or inhibiting or shrinking tumor growth of both solid tumors and blood cancers.
  • the antibodies may be used to prevent relapse of a tumor.
  • treatment with an antibody or antigen-binding fragment thereof of the invention may lead to more than 50% regression, more than 60% regression, more than 70% regression, more than 80% regression or more than 90% regression of a tumor in a subject.
  • the antibodies may be used to increase survival of a subject suffering from cancer.
  • the antibodies of the invention are useful to treat subjects suffering from a chronic viral infection. In some embodiments, the antibodies of the invention are useful in decreasing viral titers in the host and/or rescuing exhausted T-cells.
  • an antibody or antigen-binding fragment thereof the invention may be administered at a therapeutic dose to a patient with an infection by human immunodeficiency virus (HIV) or human papilloma virus (HPV) or hepatitis B/C virus (HBV/HCV).
  • HCV human immunodeficiency virus
  • HPV human papilloma virus
  • HBV/HCV hepatitis B/C virus
  • an antibody or antigen-binding fragment thereof of the invention may be used to treat an infection by simian immunodeficiency virus (SIV) in a simian subject such as cynomolgus.
  • an antibody or fragment thereof of the invention may be used to treat chronic viral infection by lymphocytic choriomeningitis virus (LCMV
  • a blocking antibody of the present invention may be administered in a therapeutically effective amount to a subject suffering from cancer or a viral infection.
  • the antibodies of the invention are useful for treating an autoimmune disease, including but not limited to, alopecia areata, autoimmune hepatitis, celiac disease, Graves' disease, Guillain-Barre syndrome, Hashimoto's disease, hemolytic anemia, inflammatory bowel disease, inflammatory myopathies, multiple sclerosis, primary biliary cirrhosis, psoriasis, rheumatoid arthritis, scleroderma, Sjögren's syndrome, systemic lupus erthyematosus, vitiligo, autoimmune pancreatitis, autoimmune urticaria, autoimmune thrombocytopenic purpura, Crohn's disease, diabetes type I, eosinophilic fasciitis, eosinophilic enterogastritis, Goodpasture's syndrome, myasthenia gravis, psoriatic arthritis, rheumatic fever, ulcerative colitis, vas
  • One or more antibodies of the present invention may be administered to relieve or prevent or decrease the severity of one or more of the symptoms or conditions of the disease or disorder.
  • one or more antibodies of the present invention prophylactically to patients at risk for developing a disease or disorder such as cancer, and chronic viral infection.
  • the present antibodies are used for the preparation of a pharmaceutical composition for treating patients suffering from cancer, autoimmune disease or viral infection.
  • the present antibodies are used as adjunct therapy with any other agent or any other therapy known to those skilled in the art useful for treating cancer, autoimmune disease or viral infection.
  • Combination therapies may include an anti-PD-L1 antibody of the invention and any additional therapeutic agent that may be advantageously combined with an antibody of the invention, or with a biologically active fragment of an antibody of the invention.
  • the antibodies of the present invention may be combined synergistically with one or more anti-cancer drugs or therapy used to treat cancer, including, for example, renal cell carcinoma, ovarian cancer, prostate cancer, colorectal cancer, non-small-cell lung cancer, and melanoma. It is contemplated herein to use anti-PD-L1 antibodies of the invention in combination with immunostimulatory and/or immunosupportive therapies to inhibit tumor growth, and/or enhance survival of cancer patients.
  • the immunostimulatory therapies include direct immunostimulatory therapies to augment immune cell activity by either “releasing the brake” on suppressed immune cells or “stepping on the gas” to activate an immune response. Examples include targeting other checkpoint receptors, vaccination and adjuvants.
  • the immunosupportive modalities may increase antigenicity of the tumor by promoting immunogenic cell death, inflammation or have other indirect effects that promote an anti-tumor immune response. Examples include radiation, chemotherapy, anti-angiogenic agents, and surgery.
  • one or more antibodies of the present invention may be used in combination with a second antibody to PD-L1, an antibody to PD-1 (e.g., nivolumab), a LAG-3 inhibitor, a CTLA-4 inhibitor (e.g., ipilimumab), a TIM3 inhibitor, a BTLA inhibitor, a TIGIT inhibitor, a CD47 inhibitor, an antagonist of another T-cell co-inhibitor or ligand (e.g., an antibody to CD-28, 2B4, LY108, LAIR1, ICOS, CD160 or VISTA), an indoleamine-2,3-dioxygenase (IDO) inhibitor, a vascular endothelial growth factor (VEGF) antagonist [e.g., a “VEGF-Trap” such as aflibercept or other VEGF-inhibiting fusion protein as set forth in U.S.
  • VEGF vascular endothelial growth factor
  • an anti-VEGF antibody or antigen binding fragment thereof e.g., bevacizumab, or ranibizumab
  • a small molecule kinase inhibitor of VEGF receptor e.g., sunitinib, sorafenib, or pazopanib
  • an Ang2 inhibitor e.g., nesvacumab
  • TGF ⁇ transforming growth factor beta
  • EGFR epidermal growth factor receptor
  • an agonist to a co-stimulatory receptor e.g., an agonist to glucocorticoid-induced TNFR-related protein
  • an antibody to a tumor-specific antigen e.g., CA9, CA125, melanoma-associated antigen 3 (MAGE3), carcinoembryonic antigen (CEA), vimentin, tumor-M2-PK, prostate-specific antigen
  • a tumor-specific antigen e.g., CA9, CA125, melanoma-associated antigen 3 (
  • the anti-PD-L1 antibodies of the present invention may be used in combination with cancer vaccines including dendritic cell vaccines, oncolytic viruses, tumor cell vaccines, etc. to augment the anti-tumor response.
  • cancer vaccines that can be used in combination with anti-PD-L1 antibodies of the present invention include MAGE3 vaccine for melanoma and bladder cancer, MUC1 vaccine for breast cancer, EGFRv3 (e.g., Rindopepimut) for brain cancer (including glioblastoma multiforme), or ALVAC-CEA (for CEA+ cancers).
  • the anti-PD-L1 antibodies of the present invention may be used in combination with a dietary supplement such as anti-oxidants or any palliative care to treat cancer.
  • the anti-PD-L1 antibodies of the invention may be administered in combination with radiation therapy in methods to generate long-term durable anti-tumor responses and/or enhance survival of patients with cancer.
  • the anti-PD-L1 antibodies of the invention may be administered prior to, concomitantly or after administering radiation therapy to a cancer patient.
  • radiation therapy may be administered in one or more doses to tumor lesions followed by administration of one or more doses of anti-PD-L1 antibodies of the invention.
  • radiation therapy may be administered locally to a tumor lesion to enhance the local immunogenicity of a patient's tumor (adjuvinating radiation) and/or to kill tumor cells (ablative radiation) followed by systemic administration of an anti-PD-L1 antibody of the invention.
  • intracranial radiation may be administered to a patient with brain cancer (e.g., glioblastoma multiforme) along with systemic administration of an anti-PD-L1 antibody of the invention.
  • the anti-PD-L1 antibodies of the invention may be administered in combination with radiation therapy and a chemotherapeutic agent (e.g., temozolomide) or a VEGF antagonist (e.g., aflibercept).
  • a chemotherapeutic agent e.g., temozolomide
  • VEGF antagonist e.g., aflibercept
  • the antibodies or fragments thereof of the invention may be administered in combination with one or more anti-viral drugs known in the art, including but not limited to, zidovudine, lamivudine, abacavir, ribavirin, lopinavir, efavirenz, cobicistat, tenofovir, rilpivirine and corticosteroids.
  • anti-viral drugs known in the art, including but not limited to, zidovudine, lamivudine, abacavir, ribavirin, lopinavir, efavirenz, cobicistat, tenofovir, rilpivirine and corticosteroids.
  • the anti-PD-L1 antibodies of the invention may be administered in combination with a LAG3 inhibitor, a CTLA-4 inhibitor, a PD-1 inhibitor or any antagonist of another T-cell co-inhibitor to treat chronic viral infection.
  • the antibodies of fragments thereof of the invention may be used in combination with any drug or therapy known in the art (e.g., corticosteroids and other immunosuppressants) to treat an autoimmune disease or disorder including, but not limited to, alopecia areata, autoimmune hepatitis, celiac disease, Graves' disease, Guillain-Barre syndrome, Hashimoto's disease, hemolytic anemia, inflammatory bowel disease, inflammatory myopathies, multiple sclerosis, primary biliary cirrhosis, psoriasis, rheumatoid arthritis, scleroderma, Sjögren's syndrome, systemic lupus erthyematosus, vitiligo, autoimmune pancreatitis, autoimmune urticaira, autoimmune thrombocytopenic purpura, Crohn's disease, diabetes type I, eosinophilic fasciitis, eosinophilic enterogastritis, Goodpas
  • the additional therapeutically active component(s) may be administered prior to, concurrent with, or after the administration of the anti-PD-L1 antibody of the present invention.
  • administration regimens are considered the administration of an anti-PD-L1 antibody “in combination with” a second therapeutically active component.
  • the additional therapeutically active component(s) may be administered to a subject prior to administration of an anti-PD-L1 antibody of the present invention.
  • a first component may be deemed to be administered “prior to” a second component if the first component is administered 1 week before, 72 hours before, 60 hours before, 48 hours before, 36 hours before, 24 hours before, 12 hours before, 6 hours before, 5 hours before, 4 hours before, 3 hours before, 2 hours before, 1 hour before, 30 minutes before, 15 minutes before, 10 minutes before, 5 minutes before, or less than 1 minute before administration of the second component.
  • the additional therapeutically active component(s) may be administered to a subject after administration of an anti-PD-L1 antibody of the present invention.
  • a first component may be deemed to be administered “after” a second component if the first component is administered 1 minute after, 5 minutes after, 10 minutes after, 15 minutes after, 30 minutes after, 1 hour after, 2 hours after, 3 hours after, 4 hours after, 5 hours after, 6 hours after, 12 hours after, 24 hours after, 36 hours after, 48 hours after, 60 hours after, 72 hours after administration of the second component.
  • the additional therapeutically active component(s) may be administered to a subject concurrent with administration of an anti-PD-L1 antibody of the present invention.
  • Constant administration includes, e.g., administration of an anti-PD-L1 antibody and an additional therapeutically active component to a subject in a single dosage form (e.g., co-formulated), or in separate dosage forms administered to the subject within about 30 minutes or less of each other.
  • each dosage form may be administered via the same route (e.g., both the anti-PD-L1 antibody and the additional therapeutically active component may be administered intravenously, subcutaneously, etc.); alternatively, each dosage form may be administered via a different route (e.g., the anti-PD-L1 antibody may be administered intravenously, and the additional therapeutically active component may be administered subcutaneously).
  • administering the components in a single dosage from, in separate dosage forms by the same route, or in separate dosage forms by different routes are all considered “concurrent administration,” for purposes of the present disclosure.
  • administration of an anti-PD-L1 antibody “prior to”, “concurrent with,” or “after” (as those terms are defined herein above) administration of an additional therapeutically active component is considered administration of an anti-PD-L1 antibody “in combination with” an additional therapeutically active component).
  • the present invention includes pharmaceutical compositions in which an anti-PD-L1 antibody of the present invention is co-formulated with one or more of the additional therapeutically active component(s) as described elsewhere herein using a variety of dosage combinations.
  • an anti-PD-L1 antibody of the invention is administered in combination with a VEGF antagonist (e.g., a VEGF trap such as aflibercept), including administration of co-formulations comprising an anti-PD-L1 antibody and a VEGF antagonist
  • a VEGF antagonist e.g., a VEGF trap such as aflibercept
  • the individual components may be administered to a subject and/or co-formulated using a variety of dosage combinations.
  • the anti-PD-L1 antibody may be administered to a subject and/or contained in a co-formulation in an amount selected from the group consisting of 0.01 mg, 0.02 mg, 0.03 mg, 0.04 mg, 0.05 mg, 0.1 mg, 0.2 mg, 0.3 mg, 0.4 mg, 0.5 mg, 0.6 mg, 0.7 mg, 0.8 mg, 0.9 mg, 1.0 mg, 1.5 mg, 2.0 mg, 2.5 mg, 3.0 mg, 3.5 mg, 4.0 mg, 4.5 mg, 5.0 mg, 6.0 mg, 7.0 mg, 8.0 mg, 9.0 mg, and 10.0 mg; and the VEGF antagonist (e.g., a VEGF trap such as aflibercept) may be administered to the subject and/or contained in a co-formulation in an amount selected from the group consisting of 0.1 mg, 0.2 mg, 0.3 mg, 0.4 mg, 0.5 mg, 0.6 mg, 0.7 mg, 0.8 mg, 0.9 mg, 1.0 mg, 1.1 mg, 1.2 mg, 1.3 mg,
  • the combinations/co-formulations may be administered to a subject according to any of the administration regimens disclosed elsewhere herein, including, e.g., twice a week, once every week, once every 2 weeks, once every 3 weeks, once every month, once every 2 months, once every 3 months, once every 4 months, once every 5 months, once every 6 months, etc.
  • multiple doses of an anti-PD-L1 antibody may be administered to a subject over a defined time course.
  • the methods according to this aspect of the invention comprise sequentially administering to a subject multiple doses of an anti-PD-L1 antibody of the invention.
  • sequentially administering means that each dose of anti-PD-L1 antibody is administered to the subject at a different point in time, e.g., on different days separated by a predetermined interval (e.g., hours, days, weeks or months).
  • the present invention includes methods which comprise sequentially administering to the patient a single initial dose of an anti-PD-L1 antibody, followed by one or more secondary doses of the anti-PD-L1 antibody, and optionally followed by one or more tertiary doses of the anti-PD-L1 antibody.
  • the anti-PD-L1 antibody may be administered at a dose of between 0.1 mg/kg to about 100 mg/kg.
  • the terms “initial dose,” “secondary doses,” and “tertiary doses,” refer to the temporal sequence of administration of the anti-PD-L1 antibody of the invention.
  • the “initial dose” is the dose which is administered at the beginning of the treatment regimen (also referred to as the “baseline dose”);
  • the “secondary doses” are the doses which are administered after the initial dose;
  • the “tertiary doses” are the doses which are administered after the secondary doses.
  • the initial, secondary, and tertiary doses may all contain the same amount of anti-PD-L1 antibody, but generally may differ from one another in terms of frequency of administration.
  • the amount of anti-PD-L1 antibody contained in the initial, secondary and/or tertiary doses varies from one another (e.g., adjusted up or down as appropriate) during the course of treatment.
  • two or more (e.g., 2, 3, 4, or 5) doses are administered at the beginning of the treatment regimen as “loading doses” followed by subsequent doses that are administered on a less frequent basis (e.g., “maintenance doses”).
  • each secondary and/or tertiary dose is administered 1 to 26 (e.g., 1, 1%, 2, 2%, 3, 3%, 4, 4%, 5, 5%, 6, 6%, 7, 7%, 8, 8%, 9, 9%, 10, 10%, 11, 11%, 12, 12%, 13, 13%, 14, 14%, 15, 15%, 16, 16%, 17, 17%, 18, 18%, 19, 19%, 20, 20%, 21, 21%, 22, 22%, 23, 23%, 24, 24%, 25, 25%, 26, 26%, or more) weeks after the immediately preceding dose.
  • the phrase “the immediately preceding dose,” as used herein, means, in a sequence of multiple administrations, the dose of anti-PD-L1 antibody which is administered to a patient prior to the administration of the very next dose in the sequence with no intervening doses.
  • the methods according to this aspect of the invention may comprise administering to a patient any number of secondary and/or tertiary doses of an anti-PD-L1 antibody.
  • any number of secondary and/or tertiary doses of an anti-PD-L1 antibody may comprise administering to a patient any number of secondary and/or tertiary doses of an anti-PD-L1 antibody.
  • only a single secondary dose is administered to the patient.
  • two or more (e.g., 2, 3, 4, 5, 6, 7, 8, or more) secondary doses are administered to the patient.
  • only a single tertiary dose is administered to the patient.
  • two or more (e.g., 2, 3, 4, 5, 6, 7, 8, or more) tertiary doses are administered to the patient.
  • each secondary dose may be administered at the same frequency as the other secondary doses. For example, each secondary dose may be administered to the patient 1 to 2 weeks or 1 to 2 months after the immediately preceding dose. Similarly, in embodiments involving multiple tertiary doses, each tertiary dose may be administered at the same frequency as the other tertiary doses. For example, each tertiary dose may be administered to the patient 2 to 12 weeks after the immediately preceding dose.
  • the frequency at which the secondary and/or tertiary doses are administered to a patient can vary over the course of the treatment regimen. The frequency of administration may also be adjusted during the course of treatment by a physician depending on the needs of the individual patient following clinical examination.
  • the present invention includes administration regimens in which 2 to 6 loading doses are administered to a patient at a first frequency (e.g., once a week, once every two weeks, once every three weeks, once a month, once every two months, etc.), followed by administration of two or more maintenance doses to the patient on a less frequent basis.
  • a first frequency e.g., once a week, once every two weeks, once every three weeks, once a month, once every two months, etc.
  • the maintenance doses may be administered to the patient once every five weeks, once every six weeks, once every seven weeks, once every eight weeks, once every ten weeks, once every twelve weeks, etc.).
  • the anti-PD-L1 antibodies of the present invention may be used to detect and/or measure PD-L1 in a sample, e.g., for diagnostic purposes. Some embodiments contemplate the use of one or more antibodies of the present invention in assays to detect a disease or disorder such as cancer, autoimmune disease or chronic viral infection. Exemplary diagnostic assays for PD-L1 may comprise, e.g., contacting a sample, obtained from a patient, with an anti-PD-L1 antibody of the invention, wherein the anti-PD-L1 antibody is labeled with a detectable label or reporter molecule or used as a capture ligand to selectively isolate PD-L1 from patient samples.
  • an unlabeled anti-PD-L1 antibody can be used in diagnostic applications in combination with a secondary antibody which is itself detectably labeled.
  • the detectable label or reporter molecule can be a radioisotope, such as 3 H, 14 C, 32 P, 35 S, or 125 I; a fluorescent or chemiluminescent moiety such as fluorescein isothiocyanate, or rhodamine; or an enzyme such as alkaline phosphatase, ⁇ -galactosidase, horseradish peroxidase, or luciferase.
  • Specific exemplary assays that can be used to detect or measure PD-L1 in a sample include enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), and fluorescence-activated cell sorting (FACS).
  • Samples that can be used in PD-L1 diagnostic assays according to the present invention include any tissue or fluid sample obtainable from a patient, which contains detectable quantities of either PD-L1 protein, or fragments thereof, under normal or pathological conditions.
  • levels of PD-L1 in a particular sample obtained from a healthy patient e.g., a patient not afflicted with cancer or an autoimmune disease
  • This baseline level of PD-L1 can then be compared against the levels of PD-L1 measured in samples obtained from individuals suspected of having a cancer-related condition, or symptoms associated with such condition.
  • the antibodies specific for PD-L1 may contain no additional labels or moieties, or they may contain an N-terminal or C-terminal label or moiety.
  • the label or moiety is biotin.
  • the location of a label may determine the orientation of the peptide relative to the surface upon which the peptide is bound. For example, if a surface is coated with avidin, a peptide containing an N-terminal biotin will be oriented such that the C-terminal portion of the peptide will be distal to the surface.
  • aspects of the invention relate to use of the disclosed antibodies as markers for predicting prognosis of cancer or an autoimmune disorder in patients.
  • Antibodies of the present invention may be used in diagnostic assays to evaluate prognosis of cancer in a patient and to predict survival.
  • Human antibodies to PD-L1 were generated using a fragment of PD-L1 that ranges from about amino acids 19-239 of SEQ ID NO: 351 (Genbank Accession No. NP_054862.1).
  • the immunogen was administered directly, with an adjuvant to stimulate the immune response, to a VELOCIMMUNE® mouse comprising DNA encoding human Immunoglobulin heavy and kappa light chain variable regions.
  • the antibody immune response was monitored by a PD-L1-specific immunoassay. When a desired immune response was achieved splenocytes were harvested and fused with mouse myeloma cells to preserve their viability and form hybridoma cell lines.
  • the hybridoma cell lines were screened and selected to identify cell lines that produce PD-L1-specific antibodies.
  • several anti-PD-L1 chimeric antibodies i.e., antibodies possessing human variable domains and mouse constant domains
  • exemplary antibodies generated in this manner were designated as H2M8306N, H2M8307N, H2M8309N, H2M8310N, H2M8312N, H2M8314N, H2M8316N, H2M8317N, H2M8321N, H2M8323N, H2M8718N, H2M8718N2, and H2M8719N.
  • Anti-PD-L1 antibodies were also isolated directly from antigen-positive B cells without fusion to myeloma cells, as described in U.S. 2007/0280945A1, herein specifically incorporated by reference in its entirety. Using this method, several fully human anti-PD-L1 antibodies (i.e., antibodies possessing human variable domains and human constant domains) were obtained; exemplary antibodies generated in this manner were designated as follows: H1H9323P, H1H9327P, H1H9329P, H1H9336P, H1H9344P2, H1H9345P2, H1H9351P2, H1H9354P2, H1H9364P2, H1H9373P2, H1H9382P2, H1H9387P2, and H1H9396P2.
  • Table 1 sets forth the amino acid sequence identifiers of the heavy and light chain variable regions and CDRs of selected anti-PD-L1 antibodies of the invention.
  • the corresponding nucleic acid sequence identifiers are set forth in Table 2.
  • Antibodies are typically referred to herein according to the following nomenclature: Fc prefix (e.g. “H1H,” “H2M,” “H2aM,” etc.), followed by a numerical identifier (e.g. “8306,” “9323,” etc., as shown in Table 1), followed by a “P,” “N,” “P2,” or “N2” suffix.
  • Fc prefix e.g. “H1H,” “H2M,” “H2aM,” etc.
  • a numerical identifier e.g. “8306,” “9323,” etc., as shown in Table 1
  • an “H1H” antibody has a human IgG1 Fc
  • an “HIM” antibody has a mouse IgG1 Fc
  • an “H2M” or “H2aM” antibody has a mouse IgG2 Fc, (all variable regions are fully human as denoted by the first ‘H’ in the antibody designation).
  • an antibody having a particular Fc isotype can be converted to an antibody with a different Fc isotype (e.g., an antibody with a mouse IgG1 Fc can be converted to an antibody with a human IgG4, etc.), but in any event, the variable domains (including the CDRs)—which are indicated by the numerical identifiers shown in Table 1—will remain the same, and the binding properties to antigen are expected to be identical or substantially similar regardless of the nature of the Fc domain.
  • Binding association and dissociation rate constants (k a and k d , respectively), equilibrium dissociation constants and dissociation half-lives (K D and t 1/2 , respectively) of antigen binding to purified anti-PD-L1 monoclonal antibodies were determined using a real-time surface plasmon resonance biosensor assay on a Biacore 4000 instrument.
  • the Biacore sensor surface was either derivatized with polyclonal rabbit anti-mouse antibody (GE, #BR-1008-38) or with monoclonal mouse anti-human Fc antibody (GE, #BR-1008-39) to capture approximately 200-300 RUs of anti-PD-L1 monoclonal antibodies, expressed with either a mouse Fc or with human Fc, respectively.
  • the PD-L1 reagents tested for binding to the anti-PD-L1 antibodies included recombinant human PD-L1 (amino acids 19-239 of accession number NP_054862.1) expressed with a C-terminal myc-myc-hexahistidine tag (hPD-L1-MMH; SEQ ID: 345), recombinant cynomolgus monkey PD-L1 expressed with a C-terminal myc-myc-hexahistidine tag (MfPD-L1-MMH; SEQ ID: 346), recombinant human PD-L1 (amino acids 19-239 of accession number NP_054862.1) expressed with either a C-terminal human IgG1 Fc tag (hPD-L1-hFc; SEQ ID: 347) or with a C-terminal mouse IgG2a Fc tag (hPD-L1-mFc; SEQ ID: 348), and recomb
  • PD-L1 reagents Different concentrations of PD-L1 reagents were injected over the anti-PD-L1 monoclonal antibody captured surface at a flow rate of 30 ⁇ L/min.
  • the binding of the PD-L1 reagents to the captured monoclonal antibodies was monitored for 3 to 4 minutes while the dissociation of antibody bound PD-L1 reagents was monitored for 10 minutes in HBST running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.05% v/v Surfactant tween-20). Experiments were performed at either 25° C. or 37° C.
  • Binding kinetics parameters for different anti-PD-L1 monoclonal antibodies binding to different PD-L1 reagents at 25° C. and 37° C. are tabulated in Tables 3-8.
  • Nonspecific binding sites were subsequently blocked using a 0.5% (w/v) solution of BSA in PBS.
  • a constant amount of 0.5 nM or 8.0 nM of a dimeric hPD-L1 protein comprised of a portion of the human PD-L1 extracellular domain that was expressed with a C-terminal mFc tag (hPD-L1-mFc; SEQ ID: 348) was separately titrated with concentrations of anti-PD-1 antibodies and isotype control antibodies ranging between 0-210 nM in serial dilution. These antibody-protein complexes were then incubated for 1 hour at room temperature (RT).
  • the absorbance measured at the constant amount of hPD-L1 on the dose curve is defined as 0% blocking and the absorbance with no added hPD-L1 is defined as 100% blocking.
  • the absorbance values of the wells containing the highest concentration for each antibody were used to determine the blocking percent at maximum concentration antibody tested.
  • Antibodies with a maximum percent blockade below 25% were characterized as non-blockers, and their IC 50 values were not reported in Table 11.
  • Antibodies with a maximum percent blockade below ⁇ 25% were characterized as non-blockers/enhancers.
  • 19 of the 25 anti-PD-L1 antibodies of the invention blocked 0.5 nM of hPD-L1-mFc from binding to hPD-1-hFc with IC 50 values ranging from less than 250 pM to 770 pM with maximum percent blockade ranging from 26% to 100%.
  • IC 50 values ranging from less than 250 pM to 770 pM with maximum percent blockade ranging from 26% to 100%.
  • Four of the 25 anti-PD-L1 antibodies tested were characterized as non-blockers of hPD-L1-mFc binding to hPD-1-hFc, while one antibody tested (H1H9327P) was characterized as a non-blocker/enhancer of hPD-L1-mFc binding to hPD-1-hFc.
  • One antibody (H2aM8307N) demonstrated weak blocking of hPD-L1-mFc binding to hPD-1-hFc with a maximum percent blockade of 29%; however the
  • 14 of the 25 anti-PD-L1 antibodies of the invention blocked 8 nM of hPD-L1-mFc from binding to hB7-1-hFc-6His with IC 50 values ranging from ⁇ 4 nM to 10 nM with maximum percent blockade ranging from 57% to 101%.
  • Two of the 25 anti-PD-L1 antibodies tested were characterized as non-blockers of hPD-L1-mFc binding to hB7-1-hFc-6His, while 8 antibodies tested were characterized as non-blockers/enhancers of hPD-L1-mFc binding to hB7-1-hFc-6His.
  • H1H9327P demonstrated weak blocking of hPD-L1-mFc binding to hB7-1-hFc-6His with a maximum percent blockade of 41%; however the IC 50 value could not be determined for this sample.
  • the Octet biosensors captured with hPD-1-hFc were subsequently submerged into wells containing the mixture of hPD-L1-mFc and different anti-PD-L1 monoclonal antibodies.
  • the entire experiment was performed at 25° C. in Octet HBST buffer (0.01 M HEPES pH7.4, 0.15M NaCl, 3 mM EDTA, 0.05% v/v Surfactant P20, 0.1 mg/mL BSA) with a plate shaking at a speed of 1000 rpm.
  • the biosensors were washed in Octet HBST buffer in between each step of the experiment.
  • the real-time binding response was monitored during the course of the experiment and the binding response at the end of every step was recorded. Binding of hPD-L1-mFc to the captured hPD-1-hFc was compared in the presence and absence of different anti-PD-L1 monoclonal antibodies and was used to determine the blocking behavior of the tested antibodies as shown in Table
  • Biacore 3000 instrument 100 nM of recombinant human PD-L1 expressed with a C-terminal human IgG1 Fc tag (hPD-L1-hFc; SEQ ID: 350) was incubated with 500 nM of each anti-PD-L1 monoclonal antibody for at least 1 hour before running the inhibition assay.
  • a CM5 Biacore sensor surface was first derivatized with anti-mouse IgG2a specific polyclonal antibody (Southern Biotech, #1080-01) using the standard EDC-NHS chemistry.
  • hPD-1-mFc C-terminal mouse IgG2a Fc tag
  • SEQ ID: 3408 C-terminal mouse IgG2a Fc tag
  • the entire experiment was performed at 25° C. in HBST running buffer (0.01 M HEPES pH7.4, 0.15M NaCl, 3 mM EDTA, 0.05% v/v Surfactant P20).
  • the real-time binding responses were monitored during the entire course of the experiment and the binding response at the end of every step was recorded. Binding of hPD-L1-hFc to the captured hPD-1-mFc was compared in the presence and absence of different anti-PD-L1 monoclonal antibodies and was used to determine the blocking behavior of the tested antibodies as shown in Table 13.
  • Binding competition between anti-PD-L1 monoclonal antibodies was determined using a real time, label-free bio-layer interferometry assay on an Octet RED384 biosensor (Pall ForteBio Corp.). The entire experiment was performed at 25° C. in Octet HBST buffer (0.01 M HEPES pH7.4, 0.15M NaCl, 3 mM EDTA, 0.05% v/v Surfactant P20, 0.1 mg/mL BSA) with the plate shaking at the speed of 1000 rpm.
  • hPD-L1-MMH C-terminal myc-myc-hexahistidine tag
  • the antigen captured biosensor tips were then saturated with first anti-PD-L1 monoclonal antibody (subsequently referred to as mAb-1) by dipping into wells containing 50 ⁇ g/mL solution of mAb-1 for 5 minutes.
  • the biosensor tips were then subsequently dipped into wells containing 50 ⁇ g/mL solution of a second anti-PD-L1 monoclonal antibody (subsequently referred to as mAb-2).
  • the biosensor tips were washed in Octet HBST buffer in between every step of the experiment.
  • the real-time binding response was monitored during the course of the experiment and the binding response at the end of every step was recorded as shown in FIG. 1 .
  • the response of mAb-2 binding to hPD-L1-MMH pre-complexed with mAb-1 was compared and competitive/non-competitive behavior of different anti-PD-L1 monoclonal antibodies was determined.
  • H2aM8307N when applied first competed with H2aM8309N, H1H9329P, H1H9336P, H2aM8314N, H2aM8316N, H2aM8718N, H1H9387P2, H1H9351P2, H1H9364P2, H1H9373P2, and H2aM8306N; however, in the opposite orientation, H2aM8309N, H1H9329P, H1H9336P, H2aM8314N, H2aM8316N, H2aM8718N, H1H9387P2, H1H9351P2, H1H9364P2, H1H9373P2, and H2aM8306N when applied first did not compete with H2aM8307N.
  • adherent cells were detached using enzyme-free dissociation buffer and blocked with complete medium. Cells were centrifuged and resuspended at a concentration of 2.8 ⁇ 10 ⁇ circumflex over ( ) ⁇ 6 cells/mL in cold PBS containing 2% FBS. HEK293 parental and HEK293/hPD-L1 cells were then incubated for 15 to 30 minutes on ice with 100 nM of each anti-PD-L1 antibody or an isotype control antibody.
  • Unbound antibodies were removed by washing with D-PBS containing 2% FBS, and cells were subsequently incubated with a phycoerythrin-conjugated secondary Fc ⁇ fragment specifically recognizing either human Fc (Jackson ImmunoResearch, #109-116-170) or mouse Fc (Jackson ImmunoResearch, #115-115-164) for 15 to 30 minutes on ice.
  • Cells were washed with D-PBS containing 2% FBS to remove unbound secondary detection reagents and fluorescence measurements were acquired using a HyperCyte (IntelliCyt, Inc.) flow cytometer. Data was analyzed using HyperCyte software.
  • Example 8 Blocking of PD-L1-Induced T-Cell Down-Regulation in a T-Cell/APC Luciferase Reporter Assay
  • T-cell activation is achieved by stimulating T-cell receptors (TcR) that recognize specific peptides presented by major histocompatibility complex class I or II proteins on antigen-presenting cells (APC).
  • TcRs T-cell receptors
  • APC antigen-presenting cells
  • Activated TcRs in turn initiate a cascade of signaling events that can be monitored by reporter genes driven by transcription factors such as activator-protein 1 (AP-1), Nuclear Factor of Activated T-cells (NFAT) or Nuclear factor kappa-light-chain-enhancer of activated B cells (NF ⁇ b).
  • T-cell response is modulated via engagement of co-receptors expressed either constitutively or inducibly on T-cells.
  • PD-1 programmed cell death protein 1
  • PD-1 programmed cell death protein 1
  • PD-1 interacts with its ligand, PD-L1, which is expressed on target cells including APCs or cancer cells, and this interaction results in the delivery of inhibitory signals by recruiting phosphatases to the TcR signalosome, resulting in the suppression of positive signaling.
  • a bioassay was developed to measure T cell signaling induced by interaction between APC and T cells by utilizing a mixed culture derived from two mammalian cell lines: Jurkat cells (an immortalized T cell line) and Raji cells (a B cell line) ( FIG. 1 ).
  • Jurkat cells an immortalized T cell line
  • Raji cells a B cell line
  • FIG. 1 For the first component of the bioassay, Jurkat Clone E6-1 cells (ATCC, #TIB-152) were transduced with the Cignal Lenti AP-1 Luc Reporter (Qiagen—Sabiosciences, #CLS-011L) as per the manufacturer's instructions.
  • the lentivirus encodes the firefly luciferase gene under the control of a minimal CMV promoter, tandem repeats of the TPA-inducible transcriptional response element (TRE) and a puromycin resistance gene.
  • the engineered Jurkat cell line was subsequently transduced with a PD-1 chimera comprising the extracellular domain of human PD-1 (amino acids from 1 to 170 of human PD1; accession number NP_005009.2) and the trans-membrane and cytoplasmic domains of human CD300a (amino acids from 181 to 299 of human CD300a; accession number NP_009192.2).
  • the resulting stable cell line (Jurkat/AP1-Luc/hPD1-hCD300a) was selected and maintained in RPMI/10% FBS/penicillin/streptomycin/glutamine supplemented with 500 ug/mL G418+1 ug/mL puromycin.
  • Raji cells (ATCC, #CCL-86) were transduced with human PD-L1 gene (amino acids 1-290 of accession number NP_054862.1) that had been cloned into a lentiviral (pLEX) vector system (Thermo Scientific Biosystems, #OHS4735).
  • Raji cells, positive for PD-L1 (Raji/hPD-L1) were isolated by FACS using a PD-L1 antibody and maintained in Iscove/10% FBS/penicillin/streptomycin/glutamine supplemented with 1 ug/mL puromycin.
  • a bispecific antibody composed of one Fab arm that bindings to CD3 on T cells and the other one Fab arm binding that binds to CD20 on Raji cells (CD3 ⁇ CD20 bispecific antibody; e.g., as disclosed in US20140088295) was utilized.
  • the presence of the bispecific molecule in the assay results in the activation of the T cell and APC by bridging the CD3 subunits on T-cells to CD20 endogenously expressed on Raji cells.
  • Ligation of CD3 with anti-CD3 antibodies has been demonstrated to lead to activation of T cells.
  • antibodies blocking the PD1/PD-L1 interaction rescue T-cell activity by disabling the inhibitory signaling and subsequently leading to increased AP1-Luc activation.
  • RPMI1640 supplemented with 10% FBS and penicillin/streptomycin/glutamine was used as assay medium to prepare cell suspensions and antibody dilutions to carry out the screening of anti-PD-L1 monoclonal antibodies (mAbs).
  • mAbs monoclonal antibodies
  • EC 50 values of anti-PD-L1 mAbs in the presence of a fixed concentration of CD3 ⁇ CD20 bispecific antibody (30 pM), as well as the EC 50 of the bispecific antibody alone, were determined.
  • cells and reagents were added to 96 well white, flat-bottom plates.
  • the anti-PD-L1 mAb EC 50 determinations first a fixed concentration of CD3 ⁇ CD20 bispecific antibody (final 30 pM) was prepared and added to the microtiter plate wells. Twelve-point serial dilutions of anti-PD-L1 mAbs and controls were then added (final concentrations ranging from 1.7 pM to 100 nM; plus wells with assay medium alone). For the bispecific antibody (alone) EC 50 determination, the bispecific antibody, at final concentrations ranging from 0.17 pM to 10 nM (plus wells with assay medium alone), was added to the microtiter plate wells.
  • a 2.5 ⁇ 10 ⁇ circumflex over ( ) ⁇ 6/mL Raji/hPD-L1 cell suspension was prepared and 20 uL per well was added (final cell number/well 5 ⁇ 10 ⁇ circumflex over ( ) ⁇ 4 cells). Plates were left at room temperature (15-20 minutes), while a suspension of 2.5 ⁇ 10 ⁇ circumflex over ( ) ⁇ 6/mL of Jurkat/AP1-Luc/hPD1(ecto)-hCD300a(TM-Cyto) was prepared. 20 uL of the Jurkat suspension (final cell number/well 5 ⁇ 10 ⁇ circumflex over ( ) ⁇ 4 cells) was added per well. Plates containing the co-culture were incubated for 5 to 6 hours at 37° C.
  • Luciferase activity was then detected after the addition of ONE-GloTM (Promega, #E6051) reagent and relative light units (RLUs) were measured on a Victor luminometer. All samples were tested in duplicates.
  • RLU values for each screened antibody were normalized by setting the assay condition with fixed (30 pM) concentration of the CD3/CD20 bispecific antibody, but without anti-PD-L1 antibody to 100%. This condition corresponds to the maximal AP1-Luc response elicited by the bispecific molecule in the presence of the PD-1/PD-L1 inhibitory signal. Upon addition of the anti-PD-L1 antibody, the inhibitory signal is suppressed, and the increased stimulation is shown here as E max , the percentage increase in the signal in the presence of the highest antibody dose tested (100 nM).
  • mice that were homozygous for the expression of the extracellular domain of human PD-L1 in place of extracellular domain of mouse PD-L1 (PD-L1 HumIn mice) on a 75% C57/BI6/25% 129 strain background.
  • MC38.Ova mouse colon adenocarcinoma cells were engineered to express chicken ovalbumin in order to increase tumor immunogenicity, and to allow monitoring of the T-cell immune responses to well-defined antigenic ovalbumin peptides.
  • MC38.Ova cells were transduced with a lentiviral vector expressing hPD-L1 under SFFV promoter.
  • MC38.Ova cells positive for hPD-L1 (MC38.Ova/hPD-L1) were isolated by FACS using hPD-L1-specific antibody. The cells were found to express low level of endogenous mouse PD-L1.
  • Isotype control 1 500 ⁇ g Days 3, 7, 10, 14, 17 H1H8314N 500 ⁇ g Days 3, 7, 10, 14, 17 H1H9364P2 500 ⁇ g Days 3, 7, 10, 14, 17 H1H9373P2 500 ⁇ g Days 3, 7, 10, 14, 17 Isotype control 2 500 ⁇ g Days 3, 7, 10, 14, 17 2
  • Isotype control 1 10 mg/kg Days 3,7, 10, 14, 17 H1H8314N 10 mg/kg Days 3, 7, 10, 14, 17 H1H8314N 5 mg/kg Days 3, 7, 10, 14, 17 H1H9364P2 10 mg/kg Days 3, 7, 10, 14, 17 H1H9364P2 5 mg/kg Days 3, 7, 10, 14, 17 H1H9373P2 10 mg/kg Days 3, 7, 10, 14, 17 H1H9373P2 5 mg/kg Days 3, 7, 10, 14, 17 H1H9373P2 10 mg/kg Days 3, 7, 10, 14, 17 H1H9373P2 5 mg/kg Days 3, 7, 10, 14, 17
  • An early-treatment tumor model was developed to test the efficacy of a combination of an anti-PD-L1 antibody and a VEGF antagonist.
  • the combination therapy is administered shortly after tumor implantation.
  • the experiment also used an anti-PD-1 antibody alone and in combination with the VEGF antagonist.
  • the anti-PD-L1 antibody used in this experiment was an anti-PD-L1 monoclonal antibody with V H /V L sequences of antibody “YW243.55S70” according to US20100203056A1 (Genentech, Inc.), with mouse IgG2a and which was cross-reactive with mouse PD-L1.
  • the VEGF antagonist used in this experiment was aflibercept (a VEGF receptor-based chimeric molecule, also known as “VEGF-trap” or “VEGFR1R2-Fc ⁇ C1(a),” a full description of which is provided elsewhere herein).
  • the anti-PD-1 antibody used in this experiment was anti-mouse PD-1 clone “RPMI-14” with rat IgG2b (Bio X Cell, West Riverside, N.H.).
  • mice were treated with one of the mono- or combination therapies, or control combination, as set forth in Table 19.
  • IgG2a isotype hFc control Combination control 250 ⁇ g, IP
  • VEGF Trap IgG2a isotype Aflibercept only control 250 ⁇ g, IP
  • anti-PD-1 anti-PD-1 mAb hFc control only RPMI-14 250 ⁇ g, IP
  • anti-PD-L1 anti-PD-L1 mAb hFc control only 250 ⁇ g, IP
  • VEGF Trap + anti-PD-1 mAb Aflibercept anti-PD-1 RPMI-14 250 ⁇ g, IP) (10 mg/kg, SC)
  • the various therapies were administered at five different time points over a two week period (i.e., injections at Day 3, Day 6, Day 10, Day 13 and Day 19).
  • Tumor growth was substantially reduced in animals treated with the combination of VEGF Trap+ anti-PD-L1 antibody as compared with treatment regimens involving either therapeutic agent alone (see FIGS. 2 and 3 ). Furthermore, survival was substantially increased in the VEGF Trap+ anti-PD-L1 antibody group, with 90% of animals surviving to at least day 50 after tumor implantation. By contrast, for the anti-PD-L1 and VEGF Trap monotherapy groups, survival to Day 50 was only 50% and 30% respectively (see FIG. 3 and Table 20).

Abstract

The present invention provides antibodies that bind to the T-cell co-inhibitor ligand programmed death-ligand1 (PD-L1) protein, and methods of use. In various embodiments of the invention, the antibodies are fully human antibodies that bind to PD-L1. In certain embodiments, the present invention provides multi-specific antigen-binding molecules comprising a first binding specificity that binds to PD-L1 and a second binding specificity that binds to a tumor cell antigen, an infected cell-specific antigen, or a T-cell co-inhibitor. In some embodiments, the antibodies of the invention are useful for inhibiting or neutralizing PD-L1 activity, thus providing a means of treating a disease or disorder such as cancer or viral infection.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. patent application Ser. No. 15/905,653 filed Feb. 26, 2018, which is a continuation of U.S. patent application Ser. No. 14/603,808 filed Jan. 23, 2015 (now U.S. Pat. No. 9,938,345), which claims the benefit of priority under 35 U.S.C. § 119(e) of U.S. Provisional Application No. 61/930,582 filed Jan. 23, 2014 and U.S. Provisional Application No. 62/089,549 filed Dec. 9, 2014, the disclosures of all of which are hereby incorporated by reference in their entireties.
  • FIELD OF THE INVENTION
  • The present invention is related to human antibodies and antigen-binding fragments of human antibodies that specifically bind to the immunomodulatory receptor ligand programmed death-ligand 1 (PD-L1), and therapeutic and diagnostic methods of using those antibodies.
  • STATEMENT OF RELATED ART
  • Programmed death-ligand 1 (PD-L1) (also called B7-H1 or CD274) is a 290 amino acid protein receptor ligand expressed widely on both lymphoid and non-lymphoid tissues such as CD4 and CD8 T-cells, macrophage lineage cells, peripheral tissues as well as on tumor cells, and virally-infected cells (Dong et al 1999, Nature Med.). PD-L1 binds to receptors PD-1 and B7-1 which belong to the CD28/CTLA-4 (cytotoxic T lymphocyte antigen)/ICOS (inducible co-stimulator) family of T-cell co-inhibitory receptors (Chen et al 2013, Nature Rev. Immunol. 13: 227-242) and attenuates the immune response by inhibiting T-cell activation. PD-L1 binding to PD-1 or B7-1 results in decreased T-cell proliferation and cytokine secretion, compromising humoral and cellular immune responses in diseases such as cancer, and viral infection.
  • The expression of PD-L1 on tumor cells and virally-infected cells is exploited by tumors and chronic viral infections to evade immune response. PD-L1 is expressed on a wide variety of tumors and studies on animal models have shown that PD-L1 on tumors inhibits T-cell activation and lysis of tumor cells and may lead to increased death of tumor-specific T-cells. In chronic viral infections, PD-L1 expressed on virally-infected cells binds to PD-1 on virus-specific T-cells and these T-cells become “exhausted” with loss of effector functions and proliferative capacity (Freeman 2008, PNAS 105: 10275-10276). The PD-1: PD-L1 system also plays an important role in induced T-regulatory (Treg) cell development and in sustaining Treg function (Francisco et al 2010, Immunol. Rev. 236: 219-242).
  • Since PD-L1 plays an important role in tumor immunity and infectious immunity, it is an ideal target for immunotherapy. Blocking PD-L1 with antagonists, including monoclonal antibodies, has been studied in treatments of cancer and chronic viral infections (Ribas 2012, NEJM 366: 2517-2519; Freeman 2008, PNAS 105: 10275-10276; Sheridan 2012, Nature Biotechnology 30: 729-730).
  • Monoclonal antibodies to PD-L1 are known in the art and have been described, for example, in US Patent/Publication Nos. 7943742, 8383796, 8217149, 20090055944, 20120003056, 20130034559, 20130045200, 20130045201, 20130045202, and in WO2007005874, WO2011066389, WO2010077634, EP1907424, and EP1899379.
  • BRIEF SUMMARY OF THE INVENTION
  • The present invention provides antibodies and antigen-binding fragments thereof that bind PD-L1. The antibodies of the present invention are useful, inter alia, for targeting cells expressing PD-L1 such as cancer cells or virally-infected cells, and for modulating PD-L1 activity. In certain embodiments, the antibodies of the invention are useful for inhibiting or neutralizing PD-L1 activity and for stimulating T cell activation, e.g., under circumstances where T cell-mediated killing is beneficial or desirable. The anti-PD-L1 antibodies of the invention, or antigen-binding portions thereof, may be included as part of a multi-specific antigen-binding molecule, for example, to modulate the immune response and/or to target the antibodies to a specific cell type, such as a tumor cell, or a virally infected cell. The antibodies are useful in treating a disease or disorder such as cancer and viral infection.
  • The antibodies of the invention can be full-length (for example, an IgG1 or IgG4 antibody) or may comprise only an antigen-binding portion (for example, a Fab, F(ab′)2 or scFv fragment), and may be modified to affect functionality, e.g., to eliminate residual effector functions (Reddy et al., 2000, J. Immunol. 164:1925-1933). In certain embodiments, the antibodies may be bispecific.
  • In a first aspect, the present invention provides isolated monoclonal antibodies or antigen-binding fragments thereof that bind specifically to PD-L1. Exemplary anti-PD-L1 antibodies of the present invention are listed in Tables 1 and 2 herein. Table 1 sets forth the amino acid sequence identifiers of the heavy chain variable regions (HCVRs), light chain variable regions (LCVRs), heavy chain complementarity determining regions (HCDR1, HCDR2 and HCDR3), and light chain complementarity determining regions (LCDR1, LCDR2 and LCDR3) of the exemplary anti-PD-L1 antibodies. Table 2 sets forth the nucleic acid sequence identifiers of the HCVRs, LCVRs, HCDR1, HCDR2 HCDR3, LCDR1, LCDR2 and LCDR3 of the exemplary anti-PD-L1 antibodies.
  • The present invention provides antibodies, or antigen-binding fragments thereof, comprising an HCVR comprising an amino acid sequence selected from any of the HCVR amino acid sequences listed in Table 1, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • The present invention also provides antibodies, or antigen-binding fragments thereof, comprising an LCVR comprising an amino acid sequence selected from any of the LCVR amino acid sequences listed in Table 1, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • The present invention also provides antibodies, or antigen-binding fragments thereof, comprising an HCVR and an LCVR amino acid sequence pair (HCVR/LCVR) comprising any of the HCVR amino acid sequences listed in Table 1 paired with any of the LCVR amino acid sequences listed in Table 1. According to certain embodiments, the present invention provides antibodies, or antigen-binding fragments thereof, comprising an HCVR/LCVR amino acid sequence pair contained within any of the exemplary anti-PD-L1 antibodies listed in Table 1. In certain embodiments, the HCVR/LCVR amino acid sequence pair is selected from the group consisting of SEQ ID NOs: 2/10, 18/26, 34/42, 50/58, 66/74, 82/90, 98/106, 114/122, 130/138, 146/154, 162/170, 178/170, 186/194, 202/210, 218/226, 234/242, 250/258, 266/274, 282/274, 290/274, 298/274, 306/274, 314/274, 322/274, 330/274, and 338/274. In certain embodiments, the HCVR/LCVR amino acid sequence pair is selected from one of SEQ ID NOs: 82/90 (e.g., H2M8314N), 162/170 (e.g., H2M8718N), 306/274 (e.g., H1H9364P2), and 314/274 (e.g., H1H9373P2). In certain other embodiments, the HCVR/LCVR amino acid sequence pair is selected from one of SEQ ID NOs: 98/106 (e.g., H2M8316N), 146/154 (e.g., H2M8323N), 290/274 (e.g., H1H9351P2), and 330/274 (e.g., H1H9387P2).
  • The present invention also provides antibodies, or antigen-binding fragments thereof, comprising a heavy chain CDR1 (HCDR1) comprising an amino acid sequence selected from any of the HCDR1 amino acid sequences listed in Table 1 or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity.
  • The present invention also provides antibodies, or antigen-binding fragments thereof, comprising a heavy chain CDR2 (HCDR2) comprising an amino acid sequence selected from any of the HCDR2 amino acid sequences listed in Table 1 or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity.
  • The present invention also provides antibodies, or antigen-binding fragments thereof, comprising a heavy chain CDR3 (HCDR3) comprising an amino acid sequence selected from any of the HCDR3 amino acid sequences listed in Table 1 or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity.
  • The present invention also provides antibodies, or antigen-binding fragments thereof, comprising a light chain CDR1 (LCDR1) comprising an amino acid sequence selected from any of the LCDR1 amino acid sequences listed in Table 1 or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity.
  • The present invention also provides antibodies, or antigen-binding fragments thereof, comprising a light chain CDR2 (LCDR2) comprising an amino acid sequence selected from any of the LCDR2 amino acid sequences listed in Table 1 or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity.
  • The present invention also provides antibodies, or antigen-binding fragments thereof, comprising a light chain CDR3 (LCDR3) comprising an amino acid sequence selected from any of the LCDR3 amino acid sequences listed in Table 1 or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity.
  • The present invention also provides antibodies, or antigen-binding fragments thereof, comprising an HCDR3 and an LCDR3 amino acid sequence pair (HCDR3/LCDR3) comprising any of the HCDR3 amino acid sequences listed in Table 1 paired with any of the LCDR3 amino acid sequences listed in Table 1. According to certain embodiments, the present invention provides antibodies, or antigen-binding fragments thereof, comprising an HCDR3/LCDR3 amino acid sequence pair contained within any of the exemplary anti-PD-L1 antibodies listed in Table 1. In certain embodiments, the HCDR3/LCDR3 amino acid sequence pair is selected from the group consisting of SEQ ID NOs: 88/96 (e.g., H2M8314N), 168/176 (e.g., H2M8718N), 312/280 (e.g., H1H9364P2), and 320/280 (e.g., H1H9373P2). In certain other embodiments, the HCDR3/LCDR3 amino acid sequence pair is selected from the group consisting of SEQ ID NOs: 104/112 (e.g., H2M8316N), 152/160 (e.g., H2M8323N), 296/280 (e.g., H1H9351P2), and 336/280 (e.g., H1H9387P2).
  • The present invention also provides antibodies, or antigen-binding fragments thereof, comprising a set of six CDRs HCDR1-HCDR2-HCDR3-LCDR1-LCDR2-LCDR3) contained within any of the exemplary anti-PD-L1 antibodies listed in Table 1. In certain embodiments, the HCDR1-HCDR2-HCDR3-LCDR1-LCDR2-LCDR3 amino acid sequence set is selected from the group consisting of SEQ ID NOs: 84-86-88-92-94-96 (e.g., H2M8314N); 164-166-168-172-174-176 (e.g., H2M8718N); 308-310-312-276-278-280 (e.g., H1H9364P2); and 316-318-320-276-278-280 (e.g., H1H9373P2). In certain other embodiments, the HCDR1-HCDR2-HCDR3-LCDR1-LCDR2-LCDR3 amino acid sequence set is selected from the group consisting of SEQ ID NOs: 100-102-104-108-110-112 (e.g., H2M8316N); 148-150-152-156-158-160 (e.g., H2M8323N); 292-294-296-276-278-280 (e.g., H1H9351P2); and 332-334-336-276-278-280 (e.g., H1H9387P2).
  • In a related embodiment, the present invention provides antibodies, or antigen-binding fragments thereof, comprising a set of six CDRs HCDR1-HCDR2-HCDR3-LCDR1-LCDR2-LCDR3) contained within an HCVR/LCVR amino acid sequence pair as defined by any of the exemplary anti-PD-L1 antibodies listed in Table 1. For example, the present invention includes antibodies, or antigen-binding fragments thereof, comprising the HCDR1-HCDR2-HCDR3-LCDR1-LCDR2-LCDR3 amino acid sequences set contained within an HCVR/LCVR amino acid sequence pair selected from the group consisting of SEQ ID NOs: 82/90 (e.g., H2M8314N), 98/106 (e.g., H2M8316N), 146/154 (e.g., H2M8323N), 162/170 (e.g., H2M8718N), 290/274 (e.g., H1H9351P2), 306/274 (e.g., H1H9364P2), 314/274 (e.g., H1H9373P2) and 330/274 (e.g., H1H9387P2). Methods and techniques for identifying CDRs within HCVR and LCVR amino acid sequences are well known in the art and can be used to identify CDRs within the specified HCVR and/or LCVR amino acid sequences disclosed herein. Exemplary conventions that can be used to identify the boundaries of CDRs include, e.g., the Kabat definition, the Chothia definition, and the AbM definition. In general terms, the Kabat definition is based on sequence variability, the Chothia definition is based on the location of the structural loop regions, and the AbM definition is a compromise between the Kabat and Chothia approaches. See, e.g., Kabat, “Sequences of Proteins of Immunological Interest,” National Institutes of Health, Bethesda, Md. (1991); Al-Lazikani et al., J. Mol. Biol. 273:927-948 (1997); and Martin et al., Proc. Natl. Acad. Sci. USA 86:9268-9272 (1989). Public databases are also available for identifying CDR sequences within an antibody.
  • The present invention includes anti-PD-L1 antibodies having a modified glycosylation pattern. In some embodiments, modification to remove undesirable glycosylation sites may be useful, or an antibody lacking a fucose moiety present on the oligosaccharide chain, for example, to increase antibody dependent cellular cytotoxicity (ADCC) function (see Shield et al. (2002) JBC 277:26733). In other applications, modification of galactosylation can be made in order to modify complement dependent cytotoxicity (CDC).
  • The present invention also provides for antibodies and antigen-binding fragments thereof that compete for specific binding to PD-L1 with an antibody or antigen-binding fragment thereof comprising the CDRs of a HCVR and the CDRs of a LCVR, wherein the HCVR and LCVR each has an amino acid sequence selected from the HCVR and LCVR sequences listed in Table 1.
  • The present invention also provides isolated antibodies and antigen-binding fragments thereof that block PD-L1 binding to PD-1 or to B7-1. In some embodiments, the antibody or antigen-binding fragment thereof that blocks PD-L1 binding to PD-1 or to B7-1 may bind to the same epitope on PD-L1 as PD-1/B7-1 or may bind to a different epitope on PD-L1 as PD-1/B7-1. In certain embodiments, the antibodies of the invention that block PD-L1 binding to PD-1 or to B7-1 comprise the CDRs of an HCVR having an amino acid sequence selected from the group consisting of HCVR sequences listed in Table 1; and the CDRs of a LCVR having an amino acid sequence selected from the group consisting of LCVR sequences listed in Table 1.
  • In alternate embodiments, the present invention provides antibodies and antigen-binding fragments thereof that do not block PD-L1 binding to PD-1 or to B7-1. In certain embodiments, the present invention provides isolated antibodies or antigen-binding fragments thereof that bind PD-L1, wherein the antibodies or antigen-binding fragments thereof enhance PD-L1 binding to PD-1 or to B7-1. In some embodiments, the isolated antibodies or antigen-binding fragments thereof that enhance PD-L1 binding to PD-1/B7-1 comprise the CDRs of a HCVR, wherein the HCVR has an amino acid sequence selected from the group consisting of SEQ ID NOs: 18, 66, 114, 130, 202, 218, 266, 282, 298, 322 and 338; and the CDRs of a LCVR, wherein the LCVR has an amino acid sequence selected from the group consisting of SEQ ID NOs: 26, 74, 122, 138, 210, 226, and 274. In some embodiments, the isolated antibodies or antigen-binding fragments thereof comprise an HCVR/LCVR amino acid sequence pair selected from the group consisting of SEQ ID NOs: 18/26 (e.g., H2M8307N), 66/74 (e.g., H2M8312N), 114/122 (e.g., H2M8317N), 130/138 (e.g., H2M8321N), 202/210 (e.g., H1H9323P), 218/226 (e.g., H1H9327P), 266/274 (e.g., H1H9344P2), 282/274 (e.g., H1H9345P2), 298/274 (e.g., H1H9354P2), 322/274 (e.g., H1H9382P2), and 338/274 (e.g., H1H9396P2).
  • The present invention also provides antibodies and antigen-binding fragments thereof that bind specifically to PD-L1 from human or other species. In certain embodiments, the antibodies may bind to human PD-L1 and/or to cynomolgus PD-L1.
  • The present invention also provides antibodies and antigen-binding fragments thereof that cross-compete for binding to PD-L1 with a reference antibody or antigen-binding fragment thereof comprising the CDRs of a HCVR and the CDRs of a LCVR, wherein the HCVR and LCVR each has an amino acid sequence selected from the HCVR and LCVR sequences listed in Table 1.
  • In one embodiment, the invention provides an isolated antibody or antigen-binding fragment that has one or more of the following characteristics: (a) blocks the binding of PD-L1 to PD-1 or to B7-1; (b) binds specifically to human PD-L1 and/or cynomolgus PD-L1; (c) inhibits T-cell proliferation in a mixed lymphocyte reaction (MLR) assay; and (d) increases IL-2 and/or interferon-gamma secretion in a MLR assay.
  • In some embodiments, the antibody or antigen binding fragment thereof may bind specifically to PD-L1 in an agonist manner, i.e., it may enhance or stimulate PD-L1 binding and/or activity; in other embodiments, the antibody may bind specifically to PD-L1 in an antagonist manner, i.e., it may block PD-L1 from binding to its receptor.
  • In certain embodiments, the antibodies or antigen-binding fragments of the present invention are bispecific comprising a first binding specificity to PD-L1 and a second binding specificity for a second target epitope. The second target epitope may be another epitope on PD-L1 or on a different protein such as a T-cell co-inhibitor. In certain embodiments, the target epitope may be on a different cell including e.g., a different T-cell, a B-cell, a tumor cell, an autoimmune tissue cell or a virally infected cell.
  • In a second aspect, the present invention provides nucleic acid molecules encoding anti-PD-L1 antibodies or portions thereof. For example, the present invention provides nucleic acid molecules encoding any of the HCVR amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the HCVR nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • The present invention also provides nucleic acid molecules encoding any of the LCVR amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the LCVR nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • The present invention also provides nucleic acid molecules encoding any of the HCDR1 amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the HCDR1 nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • The present invention also provides nucleic acid molecules encoding any of the HCDR2 amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the HCDR2 nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • The present invention also provides nucleic acid molecules encoding any of the HCDR3 amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the HCDR3 nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • The present invention also provides nucleic acid molecules encoding any of the LCDR1 amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the LCDR1 nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • The present invention also provides nucleic acid molecules encoding any of the LCDR2 amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the LCDR2 nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • The present invention also provides nucleic acid molecules encoding any of the LCDR3 amino acid sequences listed in Table 1; in certain embodiments the nucleic acid molecule comprises a polynucleotide sequence selected from any of the LCDR3 nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto.
  • The present invention also provides nucleic acid molecules encoding an HCVR, wherein the HCVR comprises a set of three CDRs (i.e., HCDR1-HCDR2-HCDR3), wherein the HCDR1-HCDR2-HCDR3 amino acid sequence set is as defined by any of the exemplary anti-PD-L1 antibodies listed in Table 1.
  • The present invention also provides nucleic acid molecules encoding an LCVR, wherein the LCVR comprises a set of three CDRs (i.e., LCDR1-LCDR2-LCDR3), wherein the LCDR1-LCDR2-LCDR3 amino acid sequence set is as defined by any of the exemplary anti-PD-L1 antibodies listed in Table 1.
  • The present invention also provides nucleic acid molecules encoding both an HCVR and an LCVR, wherein the HCVR comprises an amino acid sequence of any of the HCVR amino acid sequences listed in Table 1, and wherein the LCVR comprises an amino acid sequence of any of the LCVR amino acid sequences listed in Table 1. In certain embodiments, the nucleic acid molecule comprises a polynucleotide sequence selected from any of the HCVR nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto, and a polynucleotide sequence selected from any of the LCVR nucleic acid sequences listed in Table 2, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity thereto. In certain embodiments according to this aspect of the invention, the nucleic acid molecule encodes an HCVR and LCVR, wherein the HCVR and LCVR are both derived from the same anti-PD-L1 antibody listed in Table 1.
  • In a related aspect, the present invention provides recombinant expression vectors capable of expressing a polypeptide comprising a heavy or light chain variable region of an anti-PD-L1 antibody. For example, the present invention includes recombinant expression vectors comprising any of the nucleic acid molecules mentioned above, i.e., nucleic acid molecules encoding any of the HCVR, LCVR, and/or CDR sequences as set forth in Table 1. Also included within the scope of the present invention are host cells into which such vectors have been introduced, as well as methods of producing the antibodies or portions thereof by culturing the host cells under conditions permitting production of the antibodies or antibody fragments, and recovering the antibodies and antibody fragments so produced.
  • In a third aspect, the present invention provides multi-specific antigen-binding molecules and antigen-binding fragments thereof comprising a first antigen-binding specificity that binds specifically to PD-L1 and a second antigen-binding specificity that binds specifically to an antigen selected from the group consisting of PD-L1, a tumor cell-specific antigen, an infected-cell-specific antigen, and a T-cell co-inhibitor. In certain embodiments, the first antigen-binding specificity may comprise three CDRs derived from a HCVR with an amino acid sequence selected from the HCVR sequences in Table 1 and three CDRs derived from a LCVR with an amino acid sequence selected from the LCVR sequences in Table 1. In one embodiment, the first antigen-binding specificity may comprise an extracellular domain of PD-1 or of B7-1, or a fragment thereof. The second antigen-binding specificity may target an antigen on the same cell as PD-L1 or on a different cell of the same tissue type or of a different tissue type. For example, the multi-specific antigen-binding molecule may bind to a T-cell wherein the first antigen-binding specificity may bind specifically to PD-L1 and the second antigen-binding specificity may bind to a T-cell co-inhibitor on the T-cell. Alternatively, in another embodiment, the first antigen-binding specificity binds specifically to PD-L1 on a T-cell and the second antigen-binding specificity is targeted to an antigen/receptor on a B-cell or a macrophage or antigen-presenting cell. In certain embodiments, the second antigen-binding specificity is directed to an antigen on a tumor cell, or on a cell infected with a virus. In one embodiment, the first antigen-binding specificity comprises an extracellular domain of PD-1 and the second antigen-binding specificity binds to a different epitope on PD-L1. In certain embodiments, the first antigen-binding specificity binds to PD-L1 with a lower affinity, for example, with a KD more than 10−8 M, more than 10−7 M, more than 10−6 M, or more than 10−5 M.
  • In a fourth aspect, the invention provides a pharmaceutical composition comprising a recombinant human antibody or antigen-binding fragment thereof which specifically binds PD-L1 and a pharmaceutically acceptable carrier. In a related aspect, the invention features a composition which is a combination of an anti-PD-L1 antibody and a second therapeutic agent. In one embodiment, the second therapeutic agent is any agent that is advantageously combined with an anti-PD-L1 antibody. Exemplary agents that may be advantageously combined with an anti-PD-L1 antibody include, without limitation, other agents that bind and/or modulate PD-L1 signaling (including other antibodies or antigen-binding fragments thereof, etc.) and/or agents which do not directly bind PD-L1 but nonetheless modulate immune cell activation. Additional combination therapies and co-formulations involving the anti-PD-L1 antibodies and multi-specific antigen-binding molecules of the present invention are disclosed elsewhere herein.
  • In a fifth aspect, the invention provides methods to modulate the immune response in a subject, the method comprising administering a therapeutically effective amount of an anti-PD-L1 antibody or antigen-binding fragment thereof of the invention to the subject in need thereof. In certain embodiments, the invention provides methods to enhance the immune response in a subject, the methods comprising administering to the subject an effective amount of an antibody or fragment thereof of the invention that binds PD-L1 and blocks PD-L1 binding to PD-1 or to B7-1. In one embodiment, the invention provides a method to stimulate or enhance T-cell activation in a subject, the method comprising administering a blocking antibody or antigen-binding fragment thereof of the invention to the subject in need thereof. In one embodiment, the invention provides methods to inhibit a T-regulatory (Treg) cell in a subject, the methods comprising administering a blocking antibody or antigen-binding fragment thereof of the invention to the subject in need thereof. In certain embodiments, the subject in need thereof may suffer from a disease or disorder such as cancer or viral infection. In alternate embodiments, the present invention provides methods to inhibit T-cell activation, the methods comprising administering an activating antibody or antigen-binding fragment thereof of the invention to a subject in need thereof. In certain further embodiments, the subject in need thereof may suffer from an autoimmune disease.
  • In a sixth aspect, the invention provides therapeutic methods for treating a disease or disorder, for example, cancer or viral infection, in a subject using an anti-PD-L1 antibody or antigen-binding portion of an antibody of the invention, wherein the therapeutic methods comprise administering a therapeutically effective amount of a pharmaceutical composition comprising an antibody or fragment of an antibody of the invention to the subject in need thereof. The disorder treated is any disease or condition which is improved, ameliorated, inhibited or prevented by stimulation or inhibition of PD-L1 binding or activity. In certain embodiments, the antibody or antigen-binding fragment thereof the invention is administered in combination with a second therapeutic agent to the subject in need thereof. The second therapeutic agent may be selected from the group consisting of an antibody to a T-cell co-inhibitor, an antibody to a tumor cell antigen, an antibody to a T-cell receptor, an antibody to a Fc receptor, an antibody to an epitope on a virally infected cell, an antibody to PD-1, a cytotoxic agent, an anti-cancer drug, an anti-viral drug, an anti-inflammatory drug (e.g., corticosteroids), a VEGF antagonist, and any other drug or therapy known in the art. In certain embodiments, the second therapeutic agent may be an agent that helps to counteract or reduce any possible side effect(s) associated with an antibody or antigen-binding fragment thereof of the invention, if such side effect(s) should occur.
  • In certain embodiments, the present invention provides methods for suppressing tumor growth. In certain embodiments, the present invention provides methods to enhance survival of cancer patients. Examples of cancer include, but are not limited to, primary and/or recurrent cancer, including brain cancer (e.g., glioblastoma multiforme), lung cancer (e.g., non-small cell lung cancer), squamous cell carcinoma of head and neck, renal cell carcinoma, melanoma, multiple myeloma, prostate cancer, and colon cancer. The methods comprise administering a pharmaceutical composition comprising a therapeutically effective amount of an anti-PD-L1 antibody of the present invention in combination with a second therapeutic agent selected from the group consisting of a vascular endothelial growth factor (VEGF) antagonist (e.g., aflibercept, bevacizumab), an angiopoietin-2 (Ang2) inhibitor (e.g., an anti-Ang2 antibody such as nesvacumab), a lymphocyte activation gene 3 (LAG-3) inhibitor, a cytotoxic T-lymphocyte antigen 4 (CTLA-4) inhibitor (e.g., ipilimumab), a CCR4 inhibitor (e.g., mogamulizumab), a chemotherapeutic agent, and radiation therapy. Additional examples of additional therapies/therapeutic agents that can be used in combination with an anti-PD-L1 antibody of the invention for use in treating cancer are described elsewhere herein.
  • The antibody or fragment thereof may be administered subcutaneously, intravenously, intradermally, intraperitoneally, orally, intramuscularly, or intracranially. The antibody or fragment thereof may be administered at a dose of about 0.1 mg/kg of body weight to about 100 mg/kg of body weight of the subject.
  • The present invention also includes use of an anti-PD-L1 antibody or antigen-binding fragment thereof of the invention in the manufacture of a medicament for the treatment of a disease or disorder (e.g., cancer, and chronic viral infection) that would benefit from the blockade or enhancement of PD-L1 binding and/or signaling.
  • Other embodiments will become apparent from a review of the ensuing detailed description.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 is a schematic of the luciferase-based PD-L1 bioassay described in Example 8 herein. Panel A: Inactive Jurkat cells; Panel B: Jurkat cells are activated by T-cell receptor (TCR) clustering through the CD3×CD20 bispecific antibody; Panel C: PD-1 activation attenuates response in activated Jurkat cells; Panel D: Blocking PD-L1 rescues the response in activated Jurkat cells.
  • FIG. 2 illustrates tumor growth and survival results for mice implanted with Colon-26 tumor cells at Day 0 and treated with the indicated combinations of molecules by injection at Days 3, 6, 10, 13 and 19 (“early-treatment tumor model”). The graph depicts tumor volume (in mm3) for the different experimental groups at various time points after implantation. Upward arrows along the X-axis indicate the timing of treatment injections. “mlgG2a” is IgG2 isotype control; “Fc” is human Fc control; “VEGF Trap” is aflibercept; “anti-PD-1” is anti-mouse PD-1 clone RPMI-14; “anti-PD-L1” is an anti-PD-L1 monoclonal antibody as described elsewhere herein.
  • FIG. 3 illustrates tumor growth and survival results for mice implanted with Colon-26 tumor cells at Day 0 and treated with the indicated combinations of molecules by injection at Days 3, 6, 10, 13 and 19 (“early-treatment tumor model”). The graph shows the tumor volume (in mm3) of individual mice in each experimental group at Day 28 after implantation. “mlgG2a” is IgG2 isotype control; “Fc” is human Fc control; “VEGF Trap” is aflibercept; “anti-PD-1” is anti-mouse PD-1 clone RPMI-14; “anti-PD-L1” is an anti-PD-L1 monoclonal antibody as described elsewhere herein.
  • DETAILED DESCRIPTION
  • Before the present methods are described, it is to be understood that this invention is not limited to particular methods, and experimental conditions described, as such methods and conditions may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference in their entirety.
  • The term “PD-L1” refers to programmed death-ligand 1, also known as CD274 and B7H1. The amino acid sequence of full-length PD-L1 is provided in GenBank as accession number NP 054862.1 and is also referred to herein as SEQ ID NO: 351. The term “PD-L1” also includes protein variants of PD-L1 having the amino acid sequence of SEQ ID NOs: 345, 346, 347 or 348. The term “PD-L1” includes recombinant PD-L1 or a fragment thereof. The term also encompasses PD-L1 or a fragment thereof coupled to, for example, histidine tag, mouse or human Fc, or a signal sequence such as ROR1. For example, the term includes sequences exemplified by SEQ ID NOs: 347 or 348, comprising a mouse Fc (mlgG2a) or human Fc (hIgG1) at the C-terminal, coupled to amino acid residues 19-239 of full-length PD-L1 (SEQ ID NO: 351; NP_054862.1). Protein variants as exemplified by SEQ ID NO: 345 comprise a histidine tag at the C-terminal, coupled to amino acid residues 19-239 of NP_054862.1. Unless specified as being from a non-human species, the term “PD-L1” means human PD-L1.
  • PD-L1 is a 290 amino acid protein with extracellular IgV-like and IgC-like domains (amino acids 19-239 of full length PD-L1), a transmembrane domain and an intracellular domain of approximately 30 amino acids. PD-L1 is constitutively expressed on many cells such as antigen presenting cells (e.g., dendritic cells, macrophages, and B-cells) and on hematopoietic and non-hematopoietic cells (e.g., vascular endothelial cells, pancreatic islets, and sites of immune privilege). PD-L1 is also expressed on a wide variety of tumors, and virally-infected cells and is a component of the immunosuppressive milieu (Ribas 2012, NEJM 366: 2517-2519). PD-L1 binds to one of two T-cell co-inhibitors PD-1 and B7-1.
  • The term “PD-1” refers to the programmed death-1 protein, a T-cell co-inhibitor, also known as CD279. The amino acid sequence of full-length PD-1 is provided in GenBank as accession number NP 005009.2 and is also referred to herein as SEQ ID NO: 352. The term also encompasses PD-1 or a fragment thereof coupled to, for example, histidine tag, mouse or human Fc, or a signal sequence such as ROR1. For example, the term includes sequences exemplified by SEQ ID NOs: 349 or 350, comprising a mouse Fc (mlgG2a) or human Fc (hIgG1) at the C-terminal, coupled to amino acid residues 25-170 of NP_005009.2 with a C93S change.
  • PD-1 is a member of the CD28/CTLA-4/ICOS family of T-cell co-inhibitors. PD-1 is a 288-amino acid protein with an extracellular N-terminal domain which is IgV-like, a transmembrane domain and an intracellular domain containing an immunoreceptor tyrosine-based inhibitory (ITIM) motif and an immunoreceptor tyrosine-based switch (ITSM) motif (Chattopadhyay et al 2009, Immunol. Rev.). The PD-1 receptor has two ligands, PD-L1 and PD-L2.
  • The term “B7-1” refers to the T-lymphocyte activation antigen, also known as costimulatory factor CD80. B7-1 is a 288 amino acid membrane receptor with an extracellular N-terminal domain which comprises IgV-like (aa 37-138) and IgC-like (aa 154-232) regions, a transmembrane domain (aa 243-263) and a C-terminal intracellular region (aa 263-288). The amino acid sequence of full-length B7-1 is provided in GenBank as accession number NP_005182.1.
  • As used herein, the term “T-cell co-inhibitor” refers to a ligand and/or receptor which modulates the immune response via T-cell activation or suppression. The term “T-cell co-inhibitor”, also known as T-cell co-signaling molecule, includes, but is not limited to, PD-1, lymphocyte activation gene 3 protein (LAG-3, also known as CD223), cytotoxic T-lymphocyte antigen-4 (CTLA-4), B and T lymphocyte attenuator (BTLA), CD-28, 2B4, LY108, T-cell immunoglobulin and mucin-3 (TIM3), T-cell immunoreceptor with immunoglobulin and ITIM (TIGIT; also known as VSIG9), leucocyte associated immunoglobulin-like receptor 1 (LAIR1; also known as CD305), inducible T-cell costimulator (ICOS; also known as CD278), B7-1 (CD80), and CD160.
  • The term “antibody”, as used herein, is intended to refer to immunoglobulin molecules comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds (i.e., “full antibody molecules”), as well as multimers thereof (e.g. IgM) or antigen-binding fragments thereof. Each heavy chain is comprised of a heavy chain variable region (“HCVR” or “VH”) and a heavy chain constant region (comprised of domains C H1, CH2 and CH3). Each light chain is comprised of a light chain variable region (“LCVR or “VL”) and a light chain constant region (CL). The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. In certain embodiments of the invention, the FRs of the antibody (or antigen binding fragment thereof) may be identical to the human germline sequences, or may be naturally or artificially modified. An amino acid consensus sequence may be defined based on a side-by-side analysis of two or more CDRs.
  • Substitution of one or more CDR residues or omission of one or more CDRs is also possible. Antibodies have been described in the scientific literature in which one or two CDRs can be dispensed with for binding. Padlan et al. (1995 FASEB J. 9:133-139) analyzed the contact regions between antibodies and their antigens, based on published crystal structures, and concluded that only about one fifth to one third of CDR residues actually contact the antigen. Padlan also found many antibodies in which one or two CDRs had no amino acids in contact with an antigen (see also, Vajdos et al. 2002 J Mol Biol 320:415-428).
  • CDR residues not contacting antigen can be identified based on previous studies (for example residues H60-H65 in CDRH2 are often not required), from regions of Kabat CDRs lying outside Chothia CDRs, by molecular modeling and/or empirically. If a CDR or residue(s) thereof is omitted, it is usually substituted with an amino acid occupying the corresponding position in another human antibody sequence or a consensus of such sequences. Positions for substitution within CDRs and amino acids to substitute can also be selected empirically. Empirical substitutions can be conservative or non-conservative substitutions.
  • The fully human anti-PD-L1 monoclonal antibodies disclosed herein may comprise one or more amino acid substitutions, insertions and/or deletions in the framework and/or CDR regions of the heavy and light chain variable domains as compared to the corresponding germline sequences. Such mutations can be readily ascertained by comparing the amino acid sequences disclosed herein to germline sequences available from, for example, public antibody sequence databases. The present invention includes antibodies, and antigen-binding fragments thereof, which are derived from any of the amino acid sequences disclosed herein, wherein one or more amino acids within one or more framework and/or CDR regions are mutated to the corresponding residue(s) of the germline sequence from which the antibody was derived, or to the corresponding residue(s) of another human germline sequence, or to a conservative amino acid substitution of the corresponding germline residue(s) (such sequence changes are referred to herein collectively as “germline mutations”). A person of ordinary skill in the art, starting with the heavy and light chain variable region sequences disclosed herein, can easily produce numerous antibodies and antigen-binding fragments which comprise one or more individual germline mutations or combinations thereof. In certain embodiments, all of the framework and/or CDR residues within the VH and/or VL domains are mutated back to the residues found in the original germline sequence from which the antibody was derived. In other embodiments, only certain residues are mutated back to the original germline sequence, e.g., only the mutated residues found within the first 8 amino acids of FR1 or within the last 8 amino acids of FR4, or only the mutated residues found within CDR1, CDR2 or CDR3. In other embodiments, one or more of the framework and/or CDR residue(s) are mutated to the corresponding residue(s) of a different germline sequence (i.e., a germline sequence that is different from the germline sequence from which the antibody was originally derived). Furthermore, the antibodies of the present invention may contain any combination of two or more germline mutations within the framework and/or CDR regions, e.g., wherein certain individual residues are mutated to the corresponding residue of a particular germline sequence while certain other residues that differ from the original germline sequence are maintained or are mutated to the corresponding residue of a different germline sequence. Once obtained, antibodies and antigen-binding fragments that contain one or more germline mutations can be easily tested for one or more desired property such as, improved binding specificity, increased binding affinity, improved or enhanced antagonistic or agonistic biological properties (as the case may be), reduced immunogenicity, etc. Antibodies and antigen-binding fragments obtained in this general manner are encompassed within the present invention.
  • The present invention also includes fully human anti-PD-L1 monoclonal antibodies comprising variants of any of the HCVR, LCVR, and/or CDR amino acid sequences disclosed herein having one or more conservative substitutions. For example, the present invention includes anti-PD-L1 antibodies having HCVR, LCVR, and/or CDR amino acid sequences with, e.g., 10 or fewer, 8 or fewer, 6 or fewer, 4 or fewer, etc. conservative amino acid substitutions relative to any of the HCVR, LCVR, and/or CDR amino acid sequences disclosed herein.
  • The term “human antibody”, as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences. The human mAbs of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3. However, the term “human antibody”, as used herein, is not intended to include mAbs in which CDR sequences derived from the germline of another mammalian species (e.g., mouse), have been grafted onto human FR sequences. The term includes antibodies recombinantly produced in a non-human mammal, or in cells of a non-human mammal. The term is not intended to include antibodies isolated from or generated in a human subject.
  • The term “multi-specific antigen-binding molecules”, as used herein refers to bispecific, tri-specific or multi-specific antigen-binding molecules, and antigen-binding fragments thereof. Multi-specific antigen-binding molecules may be specific for different epitopes of one target polypeptide or may contain antigen-binding domains specific for epitopes of more than one target polypeptide. A multi-specific antigen-binding molecule can be a single multifunctional polypeptide, or it can be a multimeric complex of two or more polypeptides that are covalently or non-covalently associated with one another. The term “multi-specific antigen-binding molecules” includes antibodies of the present invention that may be linked to or co-expressed with another functional molecule, e.g., another peptide or protein. For example, an antibody or fragment thereof can be functionally linked (e.g., by chemical coupling, genetic fusion, non-covalent association or otherwise) to one or more other molecular entities, such as a protein or fragment thereof to produce a bi-specific or a multi-specific antigen-binding molecule with a second binding specificity. According to the present invention, the term “multi-specific antigen-binding molecules” also includes bi-specific, tri-specific or multi-specific antibodies or antigen-binding fragments thereof. In certain embodiments, an antibody of the present invention is functionally linked to another antibody or antigen-binding fragment thereof to produce a bispecific antibody with a second binding specificity. Bispecific and multi-specific antibodies of the present invention are described elsewhere herein.
  • The term “specifically binds,” or “binds specifically to”, or the like, means that an antibody or antigen-binding fragment thereof forms a complex with an antigen that is relatively stable under physiologic conditions. Specific binding can be characterized by an equilibrium dissociation constant of at least about 1×10−8 M or less (e.g., a smaller KD denotes a tighter binding). Methods for determining whether two molecules specifically bind are well known in the art and include, for example, equilibrium dialysis, surface plasmon resonance, and the like. As described herein, antibodies have been identified by surface plasmon resonance, e.g., BIACORE™, which bind specifically to PD-L1. Moreover, multi-specific antibodies that bind to one domain in PD-L1 and one or more additional antigens or a bi-specific that binds to two different regions of PD-L1 are nonetheless considered antibodies that “specifically bind”, as used herein.
  • The term “high affinity” antibody refers to those mAbs having a binding affinity to PD-L1, expressed as KD, of at least 10−8 M; preferably 10−9 M; more preferably 10−10 M, even more preferably 10−11 M, even more preferably 10−12 M, as measured by surface plasmon resonance, e.g., BIACORE™ or solution-affinity ELISA.
  • By the term “slow off rate”, “Koff” or “kd” is meant an antibody that dissociates from PD-L1, with a rate constant of 1×10−3 s−1 or less, preferably 1×10−4s−1 or less, as determined by surface plasmon resonance, e.g., BIACORE™.
  • The terms “antigen-binding portion” of an antibody, “antigen-binding fragment” of an antibody, and the like, as used herein, include any naturally occurring, enzymatically obtainable, synthetic, or genetically engineered polypeptide or glycoprotein that specifically binds an antigen to form a complex. The terms “antigen-binding fragment” of an antibody, or “antibody fragment”, as used herein, refers to one or more fragments of an antibody that retain the ability to bind to PD-L1.
  • In specific embodiments, antibody or antibody fragments of the invention may be conjugated to a moiety such a ligand or a therapeutic moiety (“immunoconjugate”), such as an antibiotic, a second anti-PD-L1 antibody, or an antibody to another antigen such a tumor-specific antigen, a virally-infected cell antigen, or a T-cell co-inhibitor, or an immunotoxin, or any other therapeutic moiety useful for treating a disease or condition including e.g., cancer, chronic viral infection or autoimmune disease.
  • An “isolated antibody”, as used herein, is intended to refer to an antibody that is substantially free of other antibodies (Abs) having different antigenic specificities (e.g., an isolated antibody that specifically binds PD-L1, or a fragment thereof, is substantially free of Abs that specifically bind antigens other than PD-L1.
  • A “blocking antibody” or a “neutralizing antibody”, as used herein (or an “antibody that neutralizes PD-L1 activity” or an “antagonist antibody”), is intended to refer to an antibody whose binding to PD-L1 results in inhibition of at least one biological activity of PD-L1. For example, an antibody of the invention may prevent or block PD-L1 binding to PD-1 or to B7-1.
  • An “activating antibody” or an “enhancing antibody”, as used herein (or an “agonist antibody”), is intended to refer to an antibody whose binding to PD-L1 results in increasing or stimulating at least one biological activity of PD-L1. For example, an antibody of the invention may increase or enhance PD-L1 binding to PD-1 or to B7-1.
  • The term “surface plasmon resonance”, as used herein, refers to an optical phenomenon that allows for the analysis of real-time biomolecular interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIACORE™ system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.).
  • The term “KD”, as used herein, is intended to refer to the equilibrium dissociation constant of a particular antibody-antigen interaction.
  • The term “epitope” refers to an antigenic determinant that interacts with a specific antigen binding site in the variable region of an antibody molecule known as a paratope. A single antigen may have more than one epitope. Thus, different antibodies may bind to different areas on an antigen and may have different biological effects. The term “epitope” also refers to a site on an antigen to which B and/or T cells respond. It also refers to a region of an antigen that is bound by an antibody. Epitopes may be defined as structural or functional. Functional epitopes are generally a subset of the structural epitopes and have those residues that directly contribute to the affinity of the interaction. Epitopes may also be conformational, that is, composed of non-linear amino acids. In certain embodiments, epitopes may include determinants that are chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl groups, or sulfonyl groups, and, in certain embodiments, may have specific three-dimensional structural characteristics, and/or specific charge characteristics.
  • The term “substantial identity” or “substantially identical,” when referring to a nucleic acid or fragment thereof, indicates that, when optimally aligned with appropriate nucleotide insertions or deletions with another nucleic acid (or its complementary strand), there is nucleotide sequence identity in at least about 90%, and more preferably at least about 95%, 96%, 97%, 98% or 99% of the nucleotide bases, as measured by any well-known algorithm of sequence identity, such as FASTA, BLAST or GAP, as discussed below. A nucleic acid molecule having substantial identity to a reference nucleic acid molecule may, in certain instances, encode a polypeptide having the same or substantially similar amino acid sequence as the polypeptide encoded by the reference nucleic acid molecule.
  • As applied to polypeptides, the term “substantial similarity” or “substantially similar” means that two peptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least 90% sequence identity, even more preferably at least 95%, 98% or 99% sequence identity. Preferably, residue positions, which are not identical, differ by conservative amino acid substitutions. A “conservative amino acid substitution” is one in which an amino acid residue is substituted by another amino acid residue having a side chain (R group) with similar chemical properties (e.g., charge or hydrophobicity). In general, a conservative amino acid substitution will not substantially change the functional properties of a protein. In cases where two or more amino acid sequences differ from each other by conservative substitutions, the percent or degree of similarity may be adjusted upwards to correct for the conservative nature of the substitution. Means for making this adjustment are well known to those of skill in the art. See, e.g., Pearson (1994) Methods Mol. Biol. 24: 307-331, which is herein incorporated by reference. Examples of groups of amino acids that have side chains with similar chemical properties include 1) aliphatic side chains: glycine, alanine, valine, leucine and isoleucine; 2) aliphatic-hydroxyl side chains: serine and threonine; 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; 5) basic side chains: lysine, arginine, and histidine; 6) acidic side chains: aspartate and glutamate, and 7) sulfur-containing side chains: cysteine and methionine. Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-glutamine. Alternatively, a conservative replacement is any change having a positive value in the PAM250 log-likelihood matrix disclosed in Gonnet et al. (1992) Science 256: 1443 45, herein incorporated by reference. A “moderately conservative” replacement is any change having a nonnegative value in the PAM250 log-likelihood matrix.
  • Sequence similarity for polypeptides is typically measured using sequence analysis software. Protein analysis software matches similar sequences using measures of similarity assigned to various substitutions, deletions and other modifications, including conservative amino acid substitutions. For instance, GCG software contains programs such as GAP and BESTFIT which can be used with default parameters to determine sequence homology or sequence identity between closely related polypeptides, such as homologous polypeptides from different species of organisms or between a wild type protein and a mutein thereof. See, e.g., GCG Version 6.1. Polypeptide sequences also can be compared using FASTA with default or recommended parameters; a program in GCG Version 6.1. FASTA (e.g., FASTA2 and FASTA3) provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (Pearson (2000) supra). Another preferred algorithm when comparing a sequence of the invention to a database containing a large number of sequences from different organisms is the computer program BLAST, especially BLASTP or TBLASTN, using default parameters. See, e.g., Altschul et al. (1990) J. Mol. Biol. 215: 403-410 and (1997) Nucleic Acids Res. 25:3389-3402, each of which is herein incorporated by reference.
  • By the phrase “therapeutically effective amount” is meant an amount that produces the desired effect for which it is administered. The exact amount will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, for example, Lloyd (1999) The Art, Science and Technology of Pharmaceutical Compounding).
  • As used herein, the term “subject” refers to an animal, preferably a mammal, in need of amelioration, prevention and/or treatment of a disease or disorder such as chronic viral infection, cancer or autoimmune disease.
  • As used herein, “anti-cancer drug” means any agent useful to treat cancer including, but not limited to, cytotoxins and agents such as antimetabolites, alkylating agents, anthracyclines, antibiotics, antimitotic agents, procarbazine, hydroxyurea, asparaginase, corticosteroids, mytotane (O,P′-(DDD)), interferons and radioactive agents. As used herein, “a cytotoxin or cytotoxic agent” means any agent that is detrimental to cells. Examples include Taxol® (paclitaxel), cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinbiastine, coichicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • As used herein, the term “anti-viral drug” refers to any drug or therapy used to treat, prevent, or ameliorate a viral infection in a host subject. The term “anti-viral drug” includes, but is not limited to zidovudine, lamivudine, abacavir, ribavirin, lopinavir, efavirenz, cobicistat, tenofovir, rilpivirine, analgesics and corticosteroids. In the context of the present invention, the viral infections include long-term or chronic infections caused by viruses including, but not limited to, human immunodeficiency virus (HIV), hepatitis B virus (HBV), hepatitis C virus (HCV), human papilloma virus (HPV), lymphocytic choriomeningitis virus (LCMV), and simian immunodeficiency virus (SIV).
  • The antibodies and antigen-binding fragments of the present invention specifically bind to PD-L1 and modulate the interaction of PD-L1 with PD-1 or with B7-1. The anti-PD-L1 antibodies may bind to PD-L1 with high affinity or with low affinity. In certain embodiments, the antibodies of the present invention are blocking antibodies wherein the antibodies bind to PD-L1 and block the interaction of PD-L1 with PD-1 or with B7-1. In some embodiments, the blocking antibodies of the invention block the binding of PD-L1 to PD-1 or to B7-1 and/or stimulate or enhance T-cell activation. In some embodiments, the blocking antibodies are useful for stimulating or enhancing the immune response and/or for treating a subject suffering from cancer, or a chronic viral infection. The antibodies when administered to a subject in need thereof may reduce the chronic infection by a virus such as HIV, LCMV or HBV in the subject. They may be used to inhibit the growth of tumor cells in a subject. They may be used alone or as adjunct therapy with other therapeutic moieties or modalities known in the art for treating cancer, or viral infection. In certain embodiments, the anti-PD-L1 antibodies that bind to PD-L1 with a low affinity are used as multi-specific antigen-binding molecules wherein the first binding specificity binds to PD-L1 with a low affinity and the second binding specificity binds to an antigen selected from the group consisting of a different epitope of PD-L1, a T-cell co-inhibitor such as PD-1, a tumor specific antigen and an infected-cell-specific antigen.
  • In certain embodiments, the antibodies of the present invention are agonist antibodies, wherein the antibodies bind to PD-L1 and enhance the interaction of PD-L1 and PD-1/B7-1. In some embodiments, the activating antibodies enhance binding of PD-L1 to PD-1 or to B7-1 and/or inhibit or suppress T-cell activation. The activating antibodies of the present invention may be useful for inhibiting the immune response in a subject and/or for treating autoimmune disease.
  • In certain embodiments, the anti-PD-L1 antibodies are multi-specific antigen-binding molecules, wherein they comprise a first binding specificity to PD-L1 and a second binding specificity to an antigen selected from the group consisting of a different epitope of PD-L1, a T-cell co-inhibitor such as PD-1, a tumor specific antigen and an infected-cell-specific antigen. In certain embodiments, the first binding specificity binds to PD-L1 with low affinity, e.g., with a KD of 10−8 M, 10−7 M or more.
  • Certain anti-PD-L1 antibodies of the present invention are able to bind to and neutralize the activity of PD-L1, as determined by in vitro or in vivo assays. The ability of the antibodies of the invention to bind to and neutralize the activity of PD-L1 may be measured using any standard method known to those skilled in the art, including binding assays, or activity assays, as described herein.
  • Non-limiting, exemplary in vitro assays for measuring binding activity are illustrated in Example 3, herein. In Example 3, the binding affinities and kinetic constants of human anti-PD-L1 antibodies for human PD-L1 and cynomolgus PD-L1 were determined by surface plasmon resonance and the measurements were conducted on a T200 Biacore instrument. In Examples 4 and 5, blocking assays were used to determine the ability of the anti-PD-L1 antibodies to block PD-L1-binding ability of PD-1 or to B7-1 in vitro. In Example 6, blocking assays were used to determine cross-competition between different anti-PD-L1 antibodies. Example 7 describes the binding of the antibodies to cells overexpressing PD-L1. In Example 8, a luciferase assay was used to determine the ability of anti-PD-L1 antibodies to antagonize PD-1/PD-L1 signaling in T-cells.
  • In certain embodiments, the antibodies of the present invention are able to enhance or stimulate T-cell activation in vitro and in a subject with cancer or in a subject infected with a virus such as LCMV. In certain embodiments, the antibodies of the present invention are used in combination with a second therapeutic agent, such as an antibody to a tumor-specific antigen or a T-cell co-inhibitor, to enhance the immune response and inhibit tumor growth in a subject. In certain embodiments, the agonist antibodies of the invention able to enhance PD-L1 binding to PD-1 or to B7-1 and may inhibit T-cell activation in vitro and/or in a subject with autoimmune disease.
  • The antibodies specific for PD-L1 may contain no additional labels or moieties, or they may contain an N-terminal or C-terminal label or moiety. In one embodiment, the label or moiety is biotin. In a binding assay, the location of a label (if any) may determine the orientation of the peptide relative to the surface upon which the peptide is bound. For example, if a surface is coated with avidin, a peptide containing an N-terminal biotin will be oriented such that the C-terminal portion of the peptide will be distal to the surface. In one embodiment, the label may be a radionuclide, a fluorescent dye or a MRI-detectable label. In certain embodiments, such labeled antibodies may be used in diagnostic assays including imaging assays.
  • Antigen-Binding Fragments of Antibodies
  • Unless specifically indicated otherwise, the term “antibody,” as used herein, shall be understood to encompass antibody molecules comprising two immunoglobulin heavy chains and two immunoglobulin light chains (i.e., “full antibody molecules”) as well as antigen-binding fragments thereof. The terms “antigen-binding portion” of an antibody, “antigen-binding fragment” of an antibody, and the like, as used herein, include any naturally occurring, enzymatically obtainable, synthetic, or genetically engineered polypeptide or glycoprotein that specifically binds an antigen to form a complex. The terms “antigen-binding fragment” of an antibody, or “antibody fragment”, as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to PD-L1. An antibody fragment may include a Fab fragment, a F(ab′)2 fragment, a Fv fragment, a dAb fragment, a fragment containing a CDR, or an isolated CDR. In certain embodiments, the term “antigen-binding fragment” refers to a polypeptide or fragment thereof of a multi-specific antigen-binding molecule. In such embodiments, the term “antigen-binding fragment” includes, e.g., an extracellular domain of PD-1 which binds specifically to PD-L1. Antigen-binding fragments of an antibody may be derived, e.g., from full antibody molecules using any suitable standard techniques such as proteolytic digestion or recombinant genetic engineering techniques involving the manipulation and expression of DNA encoding antibody variable and (optionally) constant domains. Such DNA is known and/or is readily available from, e.g., commercial sources, DNA libraries (including, e.g., phage-antibody libraries), or can be synthesized. The DNA may be sequenced and manipulated chemically or by using molecular biology techniques, for example, to arrange one or more variable and/or constant domains into a suitable configuration, or to introduce codons, create cysteine residues, modify, add or delete amino acids, etc.
  • Non-limiting examples of antigen-binding fragments include: (i) Fab fragments; (ii) F(ab′)2 fragments; (iii) Fd fragments; (iv) Fv fragments; (v) single-chain Fv (scFv) molecules; (vi) dAb fragments; and (vii) minimal recognition units consisting of the amino acid residues that mimic the hypervariable region of an antibody (e.g., an isolated complementarity determining region (CDR) such as a CDR3 peptide), or a constrained FR3-CDR3-FR4 peptide. Other engineered molecules, such as domain-specific antibodies, single domain antibodies, domain-deleted antibodies, chimeric antibodies, CDR-grafted antibodies, diabodies, triabodies, tetrabodies, minibodies, nanobodies (e.g. monovalent nanobodies, bivalent nanobodies, etc.), small modular immunopharmaceuticals (SMIPs), and shark variable IgNAR domains, are also encompassed within the expression “antigen-binding fragment,” as used herein.
  • An antigen-binding fragment of an antibody will typically comprise at least one variable domain. The variable domain may be of any size or amino acid composition and will generally comprise at least one CDR, which is adjacent to or in frame with one or more framework sequences. In antigen-binding fragments having a VH domain associated with a VL domain, the VH and VL domains may be situated relative to one another in any suitable arrangement. For example, the variable region may be dimeric and contain VH-VH, VH-VL or VL-VL dimers. Alternatively, the antigen-binding fragment of an antibody may contain a monomeric VH or VL domain.
  • In certain embodiments, an antigen-binding fragment of an antibody may contain at least one variable domain covalently linked to at least one constant domain. Non-limiting, exemplary configurations of variable and constant domains that may be found within an antigen-binding fragment of an antibody of the present invention include: (i) VHC H1; (ii) VH—CH2; (iii) VH—CH3; (iv) VH—CH1-CH2; (v) VH—CH1-CH2-CH3; (vi) VH—CH2-CH3, VH—CL, VLC H1; (ix) VL-CH2, (x) VL-CH3; (xi) VL-CH1-CH2; (xii) VL-CH1-CH2-CH3; VL-CH2-CH3; and (xiv) VL-CL. In any configuration of variable and constant domains, including any of the exemplary configurations listed above, the variable and constant domains may be either directly linked to one another or may be linked by a full or partial hinge or linker region. A hinge region may consist of at least 2 (e.g., 5, 10, 15, 20, 40, 60 or more) amino acids, which result in a flexible or semi-flexible linkage between adjacent variable and/or constant domains in a single polypeptide molecule. Moreover, an antigen-binding fragment of an antibody of the present invention may comprise a homo-dimer or hetero-dimer (or other multimer) of any of the variable and constant domain configurations listed above in non-covalent association with one another and/or with one or more monomeric VH or VL domain (e.g., by disulfide bond(s)).
  • As with full antibody molecules, antigen-binding fragments may be mono-specific or multi-specific (e.g., bi-specific). A multi-specific antigen-binding fragment of an antibody will typically comprise at least two different variable domains, wherein each variable domain is capable of specifically binding to a separate antigen or to a different epitope on the same antigen. Any multi-specific antibody format, including the exemplary bi-specific antibody formats disclosed herein, may be adapted for use in the context of an antigen-binding fragment of an antibody of the present invention using routine techniques available in the art.
  • Preparation of Human Antibodies
  • Methods for generating human antibodies in transgenic mice are known in the art. Any such known methods can be used in the context of the present invention to make human antibodies that specifically bind to PD-L1.
  • An immunogen comprising any one of the following can be used to generate antibodies to PD-L1. In certain embodiments, the antibodies of the invention are obtained from mice immunized with a primary immunogen, such as a full length PD-L1 [See GenBank accession number NP_054862.1 (SEQ ID NO: 351)] or with a recombinant form of PD-L1 or modified human PD-L1 fragments (SEQ ID NOs: 345, 347 or 348) or with modified cynomolgus PD-L1 fragments (SEQ ID NO: 346), followed by immunization with a secondary immunogen, or with an immunogenically active fragment of PD-L1.
  • In certain embodiments, the immunogen may be a peptide from the N terminal or C terminal end of PD-L1. In one embodiment, the immunogen is the extracellular IgV-like and/or IgC-like domain of PD-L1. In certain embodiments of the invention, the immunogen is a fragment of PD-L1 that ranges from about amino acid residues 19-239 of SEQ ID NO: 351 (NP_054862.1).
  • In some embodiments, the immunogen may be a recombinant PD-L1 peptide expressed in E. coli or in any other eukaryotic or mammalian cells such as Chinese hamster ovary (CHO) cells.
  • In certain embodiments, antibodies that bind specifically to PD-L1 may be prepared using fragments of the above-noted regions, or peptides that extend beyond the designated regions by about 5 to about 20 amino acid residues from either, or both, the N or C terminal ends of the regions described herein. In certain embodiments, any combination of the above-noted regions or fragments thereof may be used in the preparation of PD-L1 specific antibodies.
  • Using VELOCIMMUNE® technology (see, for example, U.S. Pat. No. 6,596,541, Regeneron Pharmaceuticals, VELOCIMMUNE®) or any other known method for generating monoclonal antibodies, high affinity chimeric antibodies to PD-L1 are initially isolated having a human variable region and a mouse constant region. The VELOCIMMUNE® technology involves generation of a transgenic mouse having a genome comprising human heavy and light chain variable regions operably linked to endogenous mouse constant region loci such that the mouse produces an antibody comprising a human variable region and a mouse constant region in response to antigenic stimulation. The DNA encoding the variable regions of the heavy and light chains of the antibody are isolated and operably linked to DNA encoding the human heavy and light chain constant regions. The DNA is then expressed in a cell capable of expressing the fully human antibody.
  • Bioequivalents
  • The anti-PD-L1 antibodies and antibody fragments of the present invention encompass proteins having amino acid sequences that vary from those of the described antibodies, but that retain the ability to bind PD-L1. Such variant antibodies and antibody fragments comprise one or more additions, deletions, or substitutions of amino acids when compared to parent sequence, but exhibit biological activity that is essentially equivalent to that of the described antibodies. Likewise, the antibody-encoding DNA sequences of the present invention encompass sequences that comprise one or more additions, deletions, or substitutions of nucleotides when compared to the disclosed sequence, but that encode an antibody or antibody fragment that is essentially bioequivalent to an antibody or antibody fragment of the invention.
  • Two antigen-binding proteins, or antibodies, are considered bioequivalent if, for example, they are pharmaceutical equivalents or pharmaceutical alternatives whose rate and extent of absorption do not show a significant difference when administered at the same molar dose under similar experimental conditions, either single dose or multiple doses. Some antibodies will be considered equivalents or pharmaceutical alternatives if they are equivalent in the extent of their absorption but not in their rate of absorption and yet may be considered bioequivalent because such differences in the rate of absorption are intentional and are reflected in the labeling, are not essential to the attainment of effective body drug concentrations on, e.g., chronic use, and are considered medically insignificant for the particular drug product studied.
  • In one embodiment, two antigen-binding proteins are bioequivalent if there are no clinically meaningful differences in their safety, purity, or potency.
  • In one embodiment, two antigen-binding proteins are bioequivalent if a patient can be switched one or more times between the reference product and the biological product without an expected increase in the risk of adverse effects, including a clinically significant change in immunogenicity, or diminished effectiveness, as compared to continued therapy without such switching.
  • In one embodiment, two antigen-binding proteins are bioequivalent if they both act by a common mechanism or mechanisms of action for the condition or conditions of use, to the extent that such mechanisms are known.
  • Bioequivalence may be demonstrated by in vivo and/or in vitro methods. Bioequivalence measures include, e.g., (a) an in vivo test in humans or other mammals, in which the concentration of the antibody or its metabolites is measured in blood, plasma, serum, or other biological fluid as a function of time; (b) an in vitro test that has been correlated with and is reasonably predictive of human in vivo bioavailability data; (c) an in vivo test in humans or other mammals in which the appropriate acute pharmacological effect of the antibody (or its target) is measured as a function of time; and (d) in a well-controlled clinical trial that establishes safety, efficacy, or bioavailability or bioequivalence of an antibody.
  • Bioequivalent variants of the antibodies of the invention may be constructed by, for example, making various substitutions of residues or sequences or deleting terminal or internal residues or sequences not needed for biological activity. For example, cysteine residues not essential for biological activity can be deleted or replaced with other amino acids to prevent formation of unnecessary or incorrect intramolecular disulfide bridges upon renaturation. In other contexts, bioequivalent antibodies may include antibody variants comprising amino acid changes, which modify the glycosylation characteristics of the antibodies, e.g., mutations that eliminate or remove glycosylation.
  • Anti-PD-L1 Antibodies Comprising Fc Variants
  • According to certain embodiments of the present invention, anti-PD-L1 antibodies are provided comprising an Fc domain comprising one or more mutations which enhance or diminish antibody binding to the FcRn receptor, e.g., at acidic pH as compared to neutral pH. For example, the present invention includes anti-PD-L1 antibodies comprising a mutation in the CH2 or a CH3 region of the Fc domain, wherein the mutation(s) increases the affinity of the Fc domain to FcRn in an acidic environment (e.g., in an endosome where pH ranges from about 5.5 to about 6.0). Such mutations may result in an increase in serum half-life of the antibody when administered to an animal. Non-limiting examples of such Fc modifications include, e.g., a modification at position 250 (e.g., E or Q); 250 and 428 (e.g., L or F); 252 (e.g., L/Y/F/W or T), 254 (e.g., S or T), and 256 (e.g., S/R/Q/E/D or T); or a modification at position 428 and/or 433 (e.g., H/L/R/S/P/Q or K) and/or 434 (e.g., A, W, H, F or Y [N434A, N434W, N434H, N434F or N434Y]); or a modification at position 250 and/or 428; or a modification at position 307 or 308 (e.g., 308F, V308F), and 434. In one embodiment, the modification comprises a 428L (e.g., M428L) and 434S (e.g., N434S) modification; a 428L, 259I (e.g., V259I), and 308F (e.g., V308F) modification; a 433K (e.g., H433K) and a 434 (e.g., 434Y) modification; a 252, 254, and 256 (e.g., 252Y, 254T, and 256E) modification; a 250Q and 428L modification (e.g., T250Q and M428L); and a 307 and/or 308 modification (e.g., 308F or 308P). In yet another embodiment, the modification comprises a 265A (e.g., D265A) and/or a 297A (e.g., N297A) modification.
  • For example, the present invention includes anti-PD-L1 antibodies comprising an Fc domain comprising one or more pairs or groups of mutations selected from the group consisting of: 250Q and 248L (e.g., T250Q and M248L); 252Y, 254T and 256E (e.g., M252Y, S254T and T256E); 428L and 434S (e.g., M428L and N434S); 257I and 311I (e.g., P257I and Q311I); 257I and 434H (e.g., P257I and N434H); 376V and 434H (e.g., D376V and N434H); 307A, 380A and 434A (e.g., T307A, E380A and N434A); and 433K and 434F (e.g., H433K and N434F). In one embodiment, the present invention includes anti-PD-L1 antibodies comprising an Fc domain comprising a S108P mutation in the hinge region of IgG4 to promote dimer stabilization. All possible combinations of the foregoing Fc domain mutations, and other mutations within the antibody variable domains disclosed herein, are contemplated within the scope of the present invention.
  • The present invention also includes anti-PD-L1 antibodies comprising a chimeric heavy chain constant (CH) region, wherein the chimeric CH region comprises segments derived from the CH regions of more than one immunoglobulin isotype. For example, the antibodies of the invention may comprise a chimeric CH region comprising part or all of a CH2 domain derived from a human IgG1, human IgG2 or human IgG4 molecule, combined with part or all of a CH3 domain derived from a human IgG1, human IgG2 or human IgG4 molecule. According to certain embodiments, the antibodies of the invention comprise a chimeric CH region having a chimeric hinge region. For example, a chimeric hinge may comprise an “upper hinge” amino acid sequence (amino acid residues from positions 216 to 227 according to EU numbering) derived from a human IgG1, a human IgG2 or a human IgG4 hinge region, combined with a “lower hinge” sequence (amino acid residues from positions 228 to 236 according to EU numbering) derived from a human IgG1, a human IgG2 or a human IgG4 hinge region. According to certain embodiments, the chimeric hinge region comprises amino acid residues derived from a human IgG1 or a human IgG4 upper hinge and amino acid residues derived from a human IgG2 lower hinge. An antibody comprising a chimeric CH region as described herein may, in certain embodiments, exhibit modified Fc effector functions without adversely affecting the therapeutic or pharmacokinetic properties of the antibody. (See, e.g., U.S. Ser. No. 14/170,166, filed Jan. 31, 2014, the disclosure of which is hereby incorporated by reference in its entirety).
  • Biological Characteristics of the Antibodies
  • In general, the antibodies of the present invention function by binding to PD-L1. The present invention includes anti-PD-L1 antibodies and antigen-binding fragments thereof that bind soluble monomeric or dimeric PD-L1 molecules with high affinity. For example, the present invention includes antibodies and antigen-binding fragments of antibodies that bind monomeric PD-L1 (e.g., at 25° C. or at 37° C.) with a KD of less than about 318 pM as measured by surface plasmon resonance, e.g., using the assay format as defined in Example 3 herein. In certain embodiments, the antibodies or antigen-binding fragments thereof bind monomeric PD-L1 with a KD of less than about 300 pM, less than about 250 pM, less than about 150 pM, less than about 100 pM, or less than about 50 pM, as measured by surface plasmon resonance, e.g., using the assay format as defined in Example 3 herein, or a substantially similar assay.
  • The present invention also includes antibodies and antigen-binding fragments thereof that bind dimeric PD-L1 (e.g., at 25° C. or at 37° C.) with a KD of less than about 15 pM as measured by surface plasmon resonance, e.g., using the assay format as defined in Example 3 herein. In certain embodiments, the antibodies or antigen-binding fragments thereof bind dimeric PD-L1 with a KD of less than about 12 pM, less than about 10 pM, less than about 8 pM, or less than about 5 pM, as measured by surface plasmon resonance, e.g., using the assay format as defined in Example 3 herein, or a substantially similar assay.
  • The present invention also includes antibodies or antigen-binding fragments thereof that bind cynomolgus (Macaca fascicularis) PD-L1 (e.g., at 25° C. or at 37° C.) with a KD of less than about 28 nM as measured by surface plasmon resonance, e.g., using the assay format as defined in Example 3 herein. In certain embodiments, the antibodies or antigen-binding fragments thereof bind cynomolgus PD-L1 with a KD of less than about 25 nM, less than about 20 nM, less than about 15 nM, less than about 10 nM, or less than about 5 nM, as measured by surface plasmon resonance, e.g., using the assay format as defined in Example 3 herein, or a substantially similar assay.
  • The present invention also includes antibodies and antigen-binding fragments thereof that bind PD-L1 with a dissociative half-life (t½) of greater than about 1 minute as measured by surface plasmon resonance at 25° C. or 37° C., e.g., using an assay format as defined in Example 3 herein, or a substantially similar assay. In certain embodiments, the antibodies or antigen-binding fragments of the present invention bind PD-L1 with a t % of greater than about 5 minutes, greater than about 10 minutes, greater than about 30 minutes, greater than about 50 minutes, greater than about 60 minutes, greater than about 70 minutes, greater than about 80 minutes, greater than about 90 minutes, greater than about 100 minutes, greater than about 200 minutes, greater than about 300 minutes, greater than about 400 minutes, greater than about 500 minutes, greater than about 600 minutes, greater than about 700 minutes, or greater than about 800 minutes, as measured by surface plasmon resonance at 25° C. or 37° C., e.g., using an assay format as defined in Example 3 herein (e.g., mAb-capture or antigen-capture format), or a substantially similar assay.
  • The present invention also includes antibodies or antigen-binding fragments thereof that block PD-L1 binding to PD-1 with an IC50 of less than about 770 pM as determined using a ELISA-based immunoassay assay, e.g., as shown in Example 4, or a substantially similar assay. The present invention also includes antibodies or antigen-binding fragments thereof that block PD-L1 binding to B7-1 with an IC50 of less than about 10 nM as determined using a ELISA-based immunoassay assay, e.g., as shown in Example 4, or a substantially similar assay. The present invention also includes antibodies and antigen-binding fragments thereof that bind to PD-L1 and enhance the binding of PD-L1 to PD-1 or to B7-1.
  • In some embodiments, the antibodies of the present invention may bind to the extracellular domain of PD-L1 or to a fragment of the domain. In some embodiments, the antibodies of the present invention may bind to more than one domain (cross-reactive antibodies). In certain embodiments, the antibodies of the present invention may bind to an epitope located in the extracellular domain comprising amino acid residues 19-239 of NP_054862.1 (SEQ ID NO: 351). In some embodiments, the antibodies may bind to an epitope comprising one or more amino acids selected from the group consisting of amino acid residues 1-221 of SEQ ID NOs: 345-348, or 353.
  • In certain embodiments, the antibodies of the present invention may function by blocking or inhibiting the PD-1-binding or the B7-1-binding activity associated with PD-L1 by binding to any other region or fragment of the full length protein, the amino acid sequence of which is shown in SEQ ID NO: 351. In certain embodiments, the antibodies may attenuate or modulate the interaction between PD-L1 and PD-1/B7-1.
  • In certain embodiments, the antibodies of the present invention may be bi-specific antibodies. The bi-specific antibodies of the invention may bind one epitope in one domain and may also bind a second epitope in a different domain of PD-L1. In certain embodiments, the bi-specific antibodies of the invention may bind two different epitopes in the same domain. In one embodiment, the multi-specific antigen-binding molecule comprises a first antigen-binding specificity wherein the first binding specificity comprises the extracellular domain or fragment thereof of PD-1; and a second antigen-binding specificity to another epitope of PD-L1. In another embodiment, the multi-specific antigen-binding molecule comprises a first antigen-binding specificity wherein the first binding specificity comprises the extracellular domain or fragment thereof of B7-1; and a second antigen-binding specificity to another epitope of PD-L1.
  • In one embodiment, the invention provides a fully human monoclonal antibody or antigen-binding fragment thereof that binds to PD-L1, wherein the antibody or fragment thereof exhibits one or more of the following characteristics: (i) comprises a HCVR having an amino acid sequence selected from the group consisting of SEQ ID NO: 2, 18, 34, 50, 66, 82, 98, 114, 130, 146, 162, 178, 186, 202, 218, 234, 250, 258, 266, 274, 282, 290, 298, 306, 314, 322, 330 and 338, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; (ii) comprises a LCVR having an amino acid sequence selected from the group consisting of SEQ ID NO: 10, 26, 42, 58, 74, 90, 106, 122, 138, 154, 170, 194, 210, 226, 242, 258, and 274, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; (iii) comprises a HCDR3 domain having an amino acid sequence selected from the group consisting of SEQ ID NO: 8, 24, 40, 56, 72, 88, 104, 120, 136, 152, 168, 184, 192, 208, 224, 240, 256, 272, 280, 288, 296, 304, 312, 320, 328, 336 and 344, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; and a LCDR3 domain having an amino acid sequence selected from the group consisting of SEQ ID NO: 16, 32, 48, 64, 80, 96, 112, 128, 144, 160, 176, 200, 216, 232, 248, 264, and 280, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; (iv) comprises a HCDR1 domain having an amino acid sequence selected from the group consisting of SEQ ID NO: 4, 20, 36, 52, 68, 84, 100, 116, 132, 148, 164, 180, 188, 204, 220, 236, 252, 268, 284, 292, 300, 308, 316, 324, 332, and 340, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; a HCDR2 domain having an amino acid sequence selected from the group consisting of SEQ ID NO: 6, 22, 38, 54, 70, 86, 102, 118, 134, 150, 166, 182, 190, 206, 222, 238, 254, 270, 286, 294, 302, 310, 318, 326, 334, and 342, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; a LCDR1 domain having an amino acid sequence selected from the group consisting of SEQ ID NO: 12, 28, 44, 60, 76, 92, 108, 124, 140, 156, 172, 196, 212, 228, 244, 260, and 276, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; and a LCDR2 domain having an amino acid sequence selected from the group consisting of SEQ ID NO: 14, 30, 46, 62, 78, 94, 110, 126, 142, 158, 174, 198, 214, 230, 246, 262, and 278, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; (v) is a multi-specific antigen-binding molecule comprising a first binding specificity to PD-L1 and a second binding specificity to an antigen selected from the group consisting of PD-L1, a tumor specific antigen, a virally infected cell antigen, and a T-cell co-inhibitor; (vi) binds to human PD-L1 with a KD of about 4 pM to about 645 nM; (vii) binds to cynomolgus PD-L1 with a KD of about 70 pM to about 400 nM; (viii) blocks or enhances the binding of PD-L1 to PD-1 with an IC50≤770 pM; (ix) blocks or enhances the binding of PD-L1 to B7-1 with an IC50≤10 nM; (x) blocks PD-1-induced T-cell down-regulation and/or rescues T-cell signaling in a T-cell/APC luciferase reporter assay; (xi) stimulates T-cell proliferation and activity in a mixed lymphocyte reaction (MLR) assay; (xii) induces IL-2 and/or IFNγ production in a MLR assay; and (xiii) suppresses tumor growth and increases survival in subjects with cancer.
  • In one embodiment, the invention provides a fully human monoclonal antibody or antigen-binding fragment thereof that blocks PD-L1 binding to PD-1 or to B7-1, wherein the antibody or fragment thereof exhibits one or more of the following characteristics: (i) comprises a HCVR having an amino acid sequence selected from the group consisting of SEQ ID NO: 82, 98, 146, 162, 290, 306, 314, and 330, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; (ii) comprises a LCVR having an amino acid sequence selected from the group consisting of SEQ ID NO: 90, 106, 154, 170, and 274, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; (iii) comprises a HCDR3 domain having an amino acid sequence selected from the group consisting of SEQ ID NO: 88, 104, 152, 168, 296, 312, 320, and 336, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; and a LCDR3 domain having an amino acid sequence selected from the group consisting of SEQ ID NO: 96, 112, 160, 176, and 280, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; (iv) comprises a HCDR1 domain having an amino acid sequence selected from the group consisting of SEQ ID NO: 84, 100, 148, 164, 292, 308, 316, and 332, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; a HCDR2 domain having an amino acid sequence selected from the group consisting of SEQ ID NO: 86, 102, 150, 166, 294, 310, 318, and 334, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; a LCDR1 domain having an amino acid sequence selected from the group consisting of SEQ ID NO: 92, 108, 156, 172, and 276, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; and a LCDR2 domain having an amino acid sequence selected from the group consisting of SEQ ID NO: 94, 110, 158, 174, and 278, or a substantially similar sequence thereof having at least 90%, at least 95%, at least 98% or at least 99% sequence identity; (v) is a multi-specific antigen-binding molecule comprising a first binding specificity to PD-L1 and a second binding specificity to an antigen selected from the group consisting of a different epitope of PD-L1, a tumor specific antigen, a virally-infected cell antigen, and a T-cell co-inhibitor; (vi) binds to human PD-L1 with a KD≤10−10M; (vii) binds to cynomolgus PD-L1 with a KD≤10−7M; (viii) blocks the binding of PD-L1 to PD-1 or to B7-1; (ix) blocks PD-1-induced T-cell down-regulation and/or rescues T-cell signaling in a T-cell/APC luciferase reporter assay; (xi) stimulates T-cell proliferation and activity in a mixed lymphocyte reaction (MLR) assay; (xii) induces IL-2 and/or IFNγ production in a MLR assay; and (xiii) suppresses tumor growth and increases survival in subjects with cancer.
  • The antibodies of the present invention may possess one or more of the aforementioned biological characteristics, or any combinations thereof. Other biological characteristics of the antibodies of the present invention will be evident to a person of ordinary skill in the art from a review of the present disclosure including the working Examples herein.
  • Species Selectivity and Species Cross-Reactivity
  • According to certain embodiments of the invention, the anti-PD-L1 antibodies bind to human PD-L1 but not to PD-L1 from other species. Alternatively, the anti-PD-L1 antibodies of the invention, in certain embodiments, bind to human PD-L1 and to PD-L1 from one or more non-human species. For example, the anti-PD-L1 antibodies of the invention may bind to human PD-L1 and may bind or not bind, as the case may be, to one or more of mouse, rat, guinea pig, hamster, gerbil, pig, cat, dog, rabbit, goat, sheep, cow, horse, camel, cynomolgus, marmoset, rhesus or chimpanzee PD-L1. In certain embodiments, the anti-PD-L1 antibodies of the invention may bind to human and cynomolgus PD-L1 with the same affinities or with different affinities.
  • Epitope Mapping and Related Technologies
  • The present invention includes anti-PD-L1 antibodies which interact with one or more amino acids found within one or more domains of the PD-L1 molecule including, e.g., extracellular (IgV-like) domain, the extracellular IgC-like domain, a transmembrane domain, and an intracellular domain. The epitope to which the antibodies bind may consist of a single contiguous sequence of 3 or more (e.g., 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more) amino acids located within any of the aforementioned domains of the PD-L1 molecule (e.g. a linear epitope in a domain). Alternatively, the epitope may consist of a plurality of non-contiguous amino acids (or amino acid sequences) located within either or both of the aforementioned domains of the PD-L1 molecule (e.g. a conformational epitope).
  • Various techniques known to persons of ordinary skill in the art can be used to determine whether an antibody “interacts with one or more amino acids” within a polypeptide or protein. Exemplary techniques include, for example, routine cross-blocking assays, such as that described in Antibodies, Harlow and Lane (Cold Spring Harbor Press, Cold Spring Harbor, N.Y.). Other methods include alanine scanning mutational analysis, peptide blot analysis (Reineke (2004) Methods Mol. Biol. 248: 443-63), peptide cleavage analysis crystallographic studies and NMR analysis. In addition, methods such as epitope excision, epitope extraction and chemical modification of antigens can be employed (Tomer (2000) Prot. Sci. 9: 487-496). Another method that can be used to identify the amino acids within a polypeptide with which an antibody interacts is hydrogen/deuterium exchange detected by mass spectrometry. In general terms, the hydrogen/deuterium exchange method involves deuterium-labeling the protein of interest, followed by binding the antibody to the deuterium-labeled protein. Next, the protein/antibody complex is transferred to water and exchangeable protons within amino acids that are protected by the antibody complex undergo deuterium-to-hydrogen back-exchange at a slower rate than exchangeable protons within amino acids that are not part of the interface. As a result, amino acids that form part of the protein/antibody interface may retain deuterium and therefore exhibit relatively higher mass compared to amino acids not included in the interface. After dissociation of the antibody, the target protein is subjected to protease cleavage and mass spectrometry analysis, thereby revealing the deuterium-labeled residues which correspond to the specific amino acids with which the antibody interacts. See, e.g., Ehring (1999) Analytical Biochemistry 267: 252-259; Engen and Smith (2001) Anal. Chem. 73: 256A-265A.
  • The term “epitope” refers to a site on an antigen to which B and/or T cells respond. B-cell epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents, whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents. An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation.
  • Modification-Assisted Profiling (MAP), also known as Antigen Structure-based Antibody Profiling (ASAP) is a method that categorizes large numbers of monoclonal antibodies (mAbs) directed against the same antigen according to the similarities of the binding profile of each antibody to chemically or enzymatically modified antigen surfaces (see US 2004/0101920, herein specifically incorporated by reference in its entirety). Each category may reflect a unique epitope either distinctly different from or partially overlapping with epitope represented by another category. This technology allows rapid filtering of genetically identical antibodies, such that characterization can be focused on genetically distinct antibodies. When applied to hybridoma screening, MAP may facilitate identification of rare hybridoma clones that produce mAbs having the desired characteristics. MAP may be used to sort the antibodies of the invention into groups of antibodies binding different epitopes.
  • In certain embodiments, the anti-PD-L1 antibodies or antigen-binding fragments thereof bind an epitope within any one or more of the regions exemplified in PD-L1, either in natural form, as exemplified in SEQ ID NO: 351, or recombinantly produced, as exemplified in SEQ ID NOS: 345-348, or to a fragment thereof. In some embodiments, the antibodies of the invention bind to an extracellular region comprising one or more amino acids selected from the group consisting of amino acid residues 19-239 of PD-L1. In some embodiments, the antibodies of the invention bind to a region comprising one or more amino acids selected from the group consisting of amino acid residues 1-221 of cynomolgus PD-L1, as exemplified by SEQ ID NO: 346.
  • In certain embodiments, the antibodies of the invention, as shown in Table 1, interact with at least one amino acid sequence selected from the group consisting of amino acid residues ranging from about position 19 to about position 130 of SEQ ID NO: 351; or amino acid residues ranging from about position 130 to about position 153 of SEQ ID NO: 351; or amino acid residues ranging from about position 153 to about position 210 of SEQ ID NO: 351; or to amino acid residues ranging from about position 210 to about position 239 of SEQ ID NO: 351. These regions are partially exemplified in SEQ ID NOs: 345-348.
  • The present invention includes anti-PD-L1 antibodies that bind to the same epitope, or a portion of the epitope, as any of the specific exemplary antibodies described herein in Table 1, or an antibody having the CDR sequences of any of the exemplary antibodies described in Table 1. Likewise, the present invention also includes anti-PD-L1 antibodies that compete for binding to PD-L1 or a PD-L1 fragment with any of the specific exemplary antibodies described herein in Table 1, or an antibody having the CDR sequences of any of the exemplary antibodies described in Table 1. For example, the present invention includes anti-PD-L1 antibodies that cross-compete for binding to PD-L1 with one or more antibodies as defined in Example 6 herein (e.g., H2aM8309N, H1H9329P, H1H9336P, H2aM8314N, H2aM8316N, H2AM8718N, H1H9387P2, H1H9351P2, H1H9364P2, H1H9373P2, and H2aM8306N). The present invention also includes anti-PD-L1 antibodies that cross-compete for binding to PD-L1 with one or more antibodies as defined in Example 6 herein (e.g., H1H9396P2, H2aM8317N, H2aM8321N, H1H9323P, H1H9382P2, H1H9344P2, H1H9345P2 and H1H9354P2).
  • One can easily determine whether an antibody binds to the same epitope as, or competes for binding with, a reference anti-PD-L1 antibody by using routine methods known in the art. For example, to determine if a test antibody binds to the same epitope as a reference anti-PD-L1 antibody of the invention, the reference antibody is allowed to bind to a PD-L1 protein or peptide under saturating conditions. Next, the ability of a test antibody to bind to the PD-L1 molecule is assessed. If the test antibody is able to bind to PD-L1 following saturation binding with the reference anti-PD-L1 antibody, it can be concluded that the test antibody binds to a different epitope than the reference anti-PD-L1 antibody. On the other hand, if the test antibody is not able to bind to the PD-L1 protein following saturation binding with the reference anti-PD-L1 antibody, then the test antibody may bind to the same epitope as the epitope bound by the reference anti-PD-L1 antibody of the invention.
  • To determine if an antibody competes for binding with a reference anti-PD-L1 antibody, the above-described binding methodology is performed in two orientations: In a first orientation, the reference antibody is allowed to bind to a PD-L1 protein under saturating conditions followed by assessment of binding of the test antibody to the PD-L1 molecule. In a second orientation, the test antibody is allowed to bind to a PD-L1 molecule under saturating conditions followed by assessment of binding of the reference antibody to the PD-L1 molecule. If, in both orientations, only the first (saturating) antibody is capable of binding to the PD-L1 molecule, then it is concluded that the test antibody and the reference antibody compete for binding to PD-L1. As will be appreciated by a person of ordinary skill in the art, an antibody that competes for binding with a reference antibody may not necessarily bind to the identical epitope as the reference antibody, but may sterically block binding of the reference antibody by binding an overlapping or adjacent epitope.
  • Two antibodies bind to the same or overlapping epitope if each competitively inhibits (blocks) binding of the other to the antigen. That is, a 1-, 5-, 10-, 20- or 100-fold excess of one antibody inhibits binding of the other by at least 50% but preferably 75%, 90% or even 99% as measured in a competitive binding assay (see, e.g., Junghans et al., Cancer Res. 1990 50:1495-1502). Alternatively, two antibodies have the same epitope if essentially all amino acid mutations in the antigen that reduce or eliminate binding of one antibody reduce or eliminate binding of the other. Two antibodies have overlapping epitopes if some amino acid mutations that reduce or eliminate binding of one antibody reduce or eliminate binding of the other.
  • Additional routine experimentation (e.g., peptide mutation and binding analyses) can then be carried out to confirm whether the observed lack of binding of the test antibody is in fact due to binding to the same epitope as the reference antibody or if steric blocking (or another phenomenon) is responsible for the lack of observed binding. Experiments of this sort can be performed using ELISA, RIA, surface plasmon resonance, flow cytometry or any other quantitative or qualitative antibody-binding assay available in the art.
  • Immunoconjugates
  • The invention encompasses a human anti-PD-L1 monoclonal antibody conjugated to a therapeutic moiety (“immunoconjugate”), such as a cytotoxin or a chemotherapeutic agent to treat cancer. As used herein, the term “immunoconjugate” refers to an antibody which is chemically or biologically linked to a cytotoxin, a radioactive agent, a cytokine, an interferon, a target or reporter moiety, an enzyme, a toxin, a peptide or protein or a therapeutic agent. The antibody may be linked to the cytotoxin, radioactive agent, cytokine, interferon, target or reporter moiety, enzyme, toxin, peptide or therapeutic agent at any location along the molecule so long as it is able to bind its target. Examples of immunoconjugates include antibody drug conjugates and antibody-toxin fusion proteins. In one embodiment, the agent may be a second different antibody to PD-L1. In certain embodiments, the antibody may be conjugated to an agent specific for a tumor cell or a virally infected cell. The type of therapeutic moiety that may be conjugated to the anti-PD-L1 antibody and will take into account the condition to be treated and the desired therapeutic effect to be achieved. Examples of suitable agents for forming immunoconjugates are known in the art; see for example, WO 05/103081.
  • Multi-Specific Antibodies
  • The antibodies of the present invention may be mono-specific, bi-specific, or multi-specific. Multi-specific antibodies may be specific for different epitopes of one target polypeptide or may contain antigen-binding domains specific for more than one target polypeptide. See, e.g., Tutt et al., 1991, J. Immunol. 147:60-69; Kufer et al., 2004, Trends Biotechnol. 22:238-244.
  • In one aspect, the present invention includes multi-specific antigen-binding molecules or antigen-binding fragments thereof wherein one antigen-binding specificity of an immunoglobulin is specific for an epitope within the extracellular domain of PD-L1 (e.g., in the IgV-like region), or a fragment thereof, and the other antigen-binding specificity of the immunoglobulin is specific for binding to a different epitope in the extracellular domain of PD-L1 (e.g., in the IgC-like region), or a second therapeutic target, or is conjugated to a therapeutic moiety. In certain embodiments, the first antigen-binding specificity may comprise PD-1 or B7-1 or a fragment thereof. In one embodiment, the first antigen-binding specificity that binds to PD-L1 comprises the extracellular domain of PD-1. In certain embodiments of the invention, one antigen-binding specificity of an immunoglobulin is specific for an epitope within amino acid residues 19-239 of PD-L1 (SEQ ID NO: 351) or a fragment thereof, and the other specificity of the immunoglobulin is specific for a second target antigen. The second target antigen may be on the same cell as PD-L1 or on a different cell. In one embodiment, the second target cell is on an immune cell other than a T-cell such as a B-cell, antigen-presenting cell, monocyte, macrophage, or dendritic cell. In some embodiments, the second target antigen may be present on a tumor cell or on a virally infected cell.
  • In another aspect, the invention provides multi-specific antigen-binding molecules or antigen-binding fragments thereof comprising a first antigen-binding specificity that binds to PD-L1 and a second antigen-binding specificity that binds specifically to a target antigen on a tumor cell. In various embodiments, the tumor-specific antigen is one of CA9, CA125, melanoma-associated antigen (MAGE), carcinoembryonic antigen (CEA), vimentin, tumor-M2-PK, prostate-specific antigen (PSA), MART-1, or CA19-9. Non-limiting examples of other specific tumor-associated antigens include, e.g., AFP, ALK, BAGE proteins, β-catenin, brc-abl, BRCA1, BORIS, carbonic anhydrase IX, caspase-8, CCR5, CD19, CD20, CD30, CD40, CDK4, CTLA4, cyclin-B1, CYP1B1, EGFR, EGFRvIII, ErbB2/Her2, ErbB3, ErbB4, ETV6-AML, EpCAM, EphA2, Fra-1, FOLR1, GAGE proteins (e.g., GAGE-1, -2), GD2, GD3, GloboH, glypican-3, GM3, gp100, Her2, HLA/B-raf, HLA/k-ras, HLA/MAGE-A3, hTERT, LMP2, MAGE proteins (e.g., MAGE-1, -2,-3, -4, -6, and -12), HLA-A2, MART-1, mesothelin, ML-IAP, Muc1, Muc2, Muc3, Muc4, Muc5, Muc16 (CA-125), MUM1, NA17, NY-BR1, NY-BR62, NY-BR85, NY-ES01, OX40, p15, p53, PAP, PAX3, PAX5, PCTA-1, PLAC1, PRLR, PRAME, PSMA (FOLH1), RAGE proteins, Ras, RGS5, Rho, SART-1, SART-3, Steap-1, Steap-2, survivin, TAG-72, TGF-β, TMPRSS2, Tn, TRP-1, TRP-2, tyrosinase, and uroplakin-3. In other embodiments, the second antigen-binding specificity binds to a tumor antigen that is present on tumor cells specific to, but not limited to, renal cell carcinoma, prostate cancer, colorectal cancer, melanoma, breast cancer, kidney cancer, ovarian cancer, and pancreatic cancer. The antibodies of the invention, in this aspect, may inhibit the activity of PD-L1.
  • In another aspect, the invention provides multi-specific antigen-binding molecules or antigen-binding fragments thereof wherein the second antigen-binding specificity binds to an antigen specific to a virally-infected cell. In certain embodiments, the second antigen-binding specificity binds to an antigen specific to a cell infected with a virus selected from the group consisting of HIV, HBV, HCV, HPV, LCMV and SIV.
  • In another aspect, the invention provides multi-specific antigen-binding molecules or antigen-binding fragments thereof comprising a first antigen-binding specificity that binds to PD-L1 and a second antigen-binding specificity that binds to a T-cell co-inhibitor such as PD-1, LAG-3, TIM3, B7-1, CTLA-4, BTLA, CD-28, 2B4, LY108, TIGIT, LAIR1, ICOS and CD160.
  • Any of the multi-specific antigen-binding molecules, or variants thereof, may be constructed using standard molecular biological techniques (e.g., recombinant DNA and protein expression technology), as will be well known to a person of ordinary skill in the art.
  • In some embodiments, PD-L1-specific antibodies are generated in a bi-specific format (a “bi-specific”) in which variable regions binding to distinct domains of PD-L1 are linked together to confer dual-domain specificity within a single binding molecule. Appropriately designed bi-specifics may enhance overall PD-L1 inhibitory efficacy through increasing both specificity and binding avidity. Variable regions with specificity for individual domains, (e.g., segments of the N-terminal domain), or that can bind to different regions within one domain, are paired on a structural scaffold that allows each region to bind simultaneously to the separate epitopes, or to different regions within one domain. In one example for a bi-specific, heavy chain variable regions (VH) from a binder with specificity for one domain are recombined with light chain variable regions (VL) from a series of binders with specificity for a second domain to identify non-cognate VL partners that can be paired with an original VH without disrupting the original specificity for that VH. In this way, a single VL segment (e.g., VL1) can be combined with two different VH domains (e.g., V H1 and VH2) to generate a bi-specific comprised of two binding “arms” (VH1-V L1 and VH2-VL1). Use of a single VL segment reduces the complexity of the system and thereby simplifies and increases efficiency in cloning, expression, and purification processes used to generate the bi-specific (See, for example, U.S. Ser. No. 13/022,759 and US2010/0331527).
  • Alternatively, antibodies that bind more than one domains and a second target, such as, but not limited to, for example, a second different anti-PD-L1 antibody, may be prepared in a bi-specific format using techniques described herein, or other techniques known to those skilled in the art. Antibody variable regions binding to distinct regions may be linked together with variable regions that bind to relevant sites on, for example, the extracellular domain of PD-L1, to confer dual-antigen specificity within a single binding molecule. Appropriately designed bi-specifics of this nature serve a dual function. Variable regions with specificity for the extracellular domain are combined with a variable region with specificity for outside the extracellular domain and are paired on a structural scaffold that allows each variable region to bind to the separate antigens.
  • An exemplary bi-specific antibody format that can be used in the context of the present invention involves the use of a first immunoglobulin (Ig) CH3 domain and a second Ig CH3 domain, wherein the first and second Ig CH3 domains differ from one another by at least one amino acid, and wherein at least one amino acid difference reduces binding of the bi-specific antibody to Protein A as compared to a bi-specific antibody lacking the amino acid difference. In one embodiment, the first Ig CH3 domain binds Protein A and the second Ig CH3 domain contains a mutation that reduces or abolishes Protein A binding such as an H95R modification (by IMGT exon numbering; H435R by EU numbering). The second CH3 may further comprise a Y96F modification (by IMGT; Y436F by EU). Further modifications that may be found within the second CH3 include: D16E, L18M, N44S, K52N, V57M, and V821 (by IMGT; D356E, L358M, N384S, K392N, V397M, and V4221 by EU) in the case of IgG1 antibodies; N44S, K52N, and V821 (IMGT; N384S, K392N, and V4221 by EU) in the case of IgG2 antibodies; and Q15R, N44S, K52N, V57M, R69K, E79Q, and V821 (by IMGT; Q355R, N384S, K392N, V397M, R409K, E419Q, and V4221 by EU) in the case of IgG4 antibodies. Variations on the bi-specific antibody format described above are contemplated within the scope of the present invention.
  • Other exemplary bispecific formats that can be used in the context of the present invention include, without limitation, e.g., scFv-based or diabody bispecific formats, IgG-scFv fusions, dual variable domain (DVD)-Ig, Quadroma, knobs-into-holes, common light chain (e.g., common light chain with knobs-into-holes, etc.), CrossMab, CrossFab, (SEED)body, leucine zipper, Duobody, IgG1/IgG2, dual acting Fab (DAF)-IgG, and Mabe bispecific formats (see, e.g., Klein et al. 2012, mAbs 4:6, 1-11, and references cited therein, for a review of the foregoing formats). Bispecific antibodies can also be constructed using peptide/nucleic acid conjugation, e.g., wherein unnatural amino acids with orthogonal chemical reactivity are used to generate site-specific antibody-oligonucleotide conjugates which then self-assemble into multimeric complexes with defined composition, valency and geometry. (See, e.g., Kazane et al., J. Am. Chem. Soc. [Epub: Dec. 4, 2012]).
  • Therapeutic Administration and Formulations
  • The invention provides therapeutic compositions comprising the anti-PD-L1 antibodies or antigen-binding fragments thereof of the present invention. Therapeutic compositions in accordance with the invention will be administered with suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like. A multitude of appropriate formulations can be found in the formulary known to all pharmaceutical chemists: Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa. These formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTIN™), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. See also Powell et al. “Compendium of excipients for parenteral formulations” PDA (1998) J Pharm Sci Technol 52:238-311.
  • The dose of antibody may vary depending upon the age and the size of a subject to be administered, target disease, conditions, route of administration, and the like. When an antibody of the present invention is used for treating a disease or disorder in an adult patient, or for preventing such a disease, it is advantageous to administer the antibody of the present invention normally at a single dose of about 0.1 to about 100 mg/kg body weight, more preferably about 5 to about 80, about 10 to about 60, or about 20 to about 50 mg/kg body weight. Depending on the severity of the condition, the frequency and the duration of the treatment can be adjusted. In certain embodiments, the antibody or antigen-binding fragment thereof of the invention can be administered as an initial dose of at least about 0.1 mg to about 800 mg, about 1 to about 500 mg, about 5 to about 300 mg, or about 10 to about 200 mg, to about 100 mg, or to about 50 mg. In certain embodiments, the initial dose may be followed by administration of a second or a plurality of subsequent doses of the antibody or antigen-binding fragment thereof in an amount that can be approximately the same or less than that of the initial dose, wherein the subsequent doses are separated by at least 1 day to 3 days; at least one week, at least 2 weeks; at least 3 weeks; at least 4 weeks; at least 5 weeks; at least 6 weeks; at least 7 weeks; at least 8 weeks; at least 9 weeks; at least 10 weeks; at least 12 weeks; or at least 14 weeks.
  • Various delivery systems are known and can be used to administer the pharmaceutical composition of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the mutant viruses, receptor mediated endocytosis (see, e.g., Wu et al. (1987) J. Biol. Chem. 262:4429-4432). Methods of introduction include, but are not limited to, intradermal, transdermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural and oral routes. The composition may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local. The pharmaceutical composition can be also delivered in a vesicle, in particular a liposome (see, for example, Langer (1990) Science 249:1527-1533).
  • The use of nanoparticles to deliver the antibodies of the present invention is also contemplated herein. Antibody-conjugated nanoparticles may be used both for therapeutic and diagnostic applications. Antibody-conjugated nanoparticles and methods of preparation and use are described in detail by Arruebo, M., et al. 2009 (“Antibody-conjugated nanoparticles for biomedical applications” in J. Nanomat. Volume 2009, Article ID 439389, 24 pages, doi: 10.1155/2009/439389), incorporated herein by reference. Nanoparticles may be developed and conjugated to antibodies contained in pharmaceutical compositions to target tumor cells or autoimmune tissue cells or virally infected cells. Nanoparticles for drug delivery have also been described in, for example, U.S. Pat. No. 8,257,740, or U.S. Pat. No. 8,246,995, each incorporated herein in its entirety.
  • In certain situations, the pharmaceutical composition can be delivered in a controlled release system. In one embodiment, a pump may be used. In another embodiment, polymeric materials can be used. In yet another embodiment, a controlled release system can be placed in proximity of the composition's target, thus requiring only a fraction of the systemic dose.
  • The injectable preparations may include dosage forms for intravenous, subcutaneous, intracutaneous, intracranial, intraperitoneal and intramuscular injections, drip infusions, etc. These injectable preparations may be prepared by methods publicly known. For example, the injectable preparations may be prepared, e.g., by dissolving, suspending or emulsifying the antibody or its salt described above in a sterile aqueous medium or an oily medium conventionally used for injections. As the aqueous medium for injections, there are, for example, physiological saline, an isotonic solution containing glucose and other auxiliary agents, etc., which may be used in combination with an appropriate solubilizing agent such as an alcohol (e.g., ethanol), a polyalcohol (e.g., propylene glycol, polyethylene glycol), a nonionic surfactant [e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)], etc. As the oily medium, there are employed, e.g., sesame oil, soybean oil, etc., which may be used in combination with a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc. The injection thus prepared is preferably filled in an appropriate ampoule.
  • A pharmaceutical composition of the present invention can be delivered subcutaneously or intravenously with a standard needle and syringe. In addition, with respect to subcutaneous delivery, a pen delivery device readily has applications in delivering a pharmaceutical composition of the present invention. Such a pen delivery device can be reusable or disposable. A reusable pen delivery device generally utilizes a replaceable cartridge that contains a pharmaceutical composition. Once all of the pharmaceutical composition within the cartridge has been administered and the cartridge is empty, the empty cartridge can readily be discarded and replaced with a new cartridge that contains the pharmaceutical composition. The pen delivery device can then be reused. In a disposable pen delivery device, there is no replaceable cartridge. Rather, the disposable pen delivery device comes prefilled with the pharmaceutical composition held in a reservoir within the device. Once the reservoir is emptied of the pharmaceutical composition, the entire device is discarded.
  • Numerous reusable pen and autoinjector delivery devices have applications in the subcutaneous delivery of a pharmaceutical composition of the present invention. Examples include, but certainly are not limited to AUTOPEN™ (Owen Mumford, Inc., Woodstock, UK), DISETRONIC™ pen (Disetronic Medical Systems, Burghdorf, Switzerland), HUMALOG MIX 75/25™ pen, HUMALOG™ pen, HUMALIN 70/30™ pen (Eli Lilly and Co., Indianapolis, Ind.), NOVOPEN™ I, II and III (Novo Nordisk, Copenhagen, Denmark), NOVOPEN JUNIOR™ (Novo Nordisk, Copenhagen, Denmark), BD™ pen (Becton Dickinson, Franklin Lakes, N.J.), OPTIPEN™, OPTIPEN PRO™, OPTIPEN STARLET™, and OPTICLIK™ (Sanofi-Aventis, Frankfurt, Germany), to name only a few. Examples of disposable pen delivery devices having applications in subcutaneous delivery of a pharmaceutical composition of the present invention include, but certainly are not limited to the SOLOSTAR™ pen (Sanofi-Aventis), the FLEXPEN™ (Novo Nordisk), and the KWIKPEN™ (Eli Lilly), the SURECLICK™ Autoinjector (Amgen, Thousand Oaks, Calif.), the PENLET™ (Haselmeier, Stuttgart, Germany), the EPIPEN (Dey, L. P.) and the HUMIRA™ Pen (Abbott Labs, Abbott Park, Ill.), to name only a few.
  • Advantageously, the pharmaceutical compositions for oral or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the active ingredients. Such dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, etc. The amount of the antibody contained is generally about 5 to about 500 mg per dosage form in a unit dose; especially in the form of injection, it is preferred that the antibody is contained in about 5 to about 100 mg and in about 10 to about 250 mg for the other dosage forms.
  • Therapeutic Uses of the Antibodies
  • The antibodies of the present invention are useful for the treatment, prevention, and/or amelioration of disease or disorder or condition such as cancer, autoimmune disease or a viral infection and/or for ameliorating at least one symptom associated with such disease, disorder or condition. In some embodiments of the invention, the antibodies described herein are useful for treating subjects suffering from primary or recurrent cancer, including for example, renal cell carcinoma, prostate cancer, ovarian cancer, kidney cancer, colorectal cancer, gastric cancer, breast cancer, head and neck cancer, non-small-cell lung cancer, brain cancer, multiple myeloma, and melanoma. The antibodies may be used to treat early stage or late-stage symptoms of cancer. In one embodiment, an antibody or fragment thereof of the invention may be used to treat metastatic cancer. The antibodies are useful in reducing or inhibiting or shrinking tumor growth of both solid tumors and blood cancers. In certain embodiments, the antibodies may be used to prevent relapse of a tumor. In certain embodiments, treatment with an antibody or antigen-binding fragment thereof of the invention may lead to more than 50% regression, more than 60% regression, more than 70% regression, more than 80% regression or more than 90% regression of a tumor in a subject. In certain embodiments, the antibodies may be used to increase survival of a subject suffering from cancer.
  • In certain embodiments, the antibodies of the invention are useful to treat subjects suffering from a chronic viral infection. In some embodiments, the antibodies of the invention are useful in decreasing viral titers in the host and/or rescuing exhausted T-cells. In one embodiment, an antibody or antigen-binding fragment thereof the invention may be administered at a therapeutic dose to a patient with an infection by human immunodeficiency virus (HIV) or human papilloma virus (HPV) or hepatitis B/C virus (HBV/HCV). In a related embodiment, an antibody or antigen-binding fragment thereof of the invention may be used to treat an infection by simian immunodeficiency virus (SIV) in a simian subject such as cynomolgus. In another embodiment, an antibody or fragment thereof of the invention may be used to treat chronic viral infection by lymphocytic choriomeningitis virus (LCMV).
  • In certain embodiments, a blocking antibody of the present invention may be administered in a therapeutically effective amount to a subject suffering from cancer or a viral infection.
  • In certain embodiments, the antibodies of the invention are useful for treating an autoimmune disease, including but not limited to, alopecia areata, autoimmune hepatitis, celiac disease, Graves' disease, Guillain-Barre syndrome, Hashimoto's disease, hemolytic anemia, inflammatory bowel disease, inflammatory myopathies, multiple sclerosis, primary biliary cirrhosis, psoriasis, rheumatoid arthritis, scleroderma, Sjögren's syndrome, systemic lupus erthyematosus, vitiligo, autoimmune pancreatitis, autoimmune urticaria, autoimmune thrombocytopenic purpura, Crohn's disease, diabetes type I, eosinophilic fasciitis, eosinophilic enterogastritis, Goodpasture's syndrome, myasthenia gravis, psoriatic arthritis, rheumatic fever, ulcerative colitis, vasculitis and Wegener's granulomatosis. In certain embodiments, an activating antibody of the invention may be used to treat a subject suffering from autoimmune disease.
  • One or more antibodies of the present invention may be administered to relieve or prevent or decrease the severity of one or more of the symptoms or conditions of the disease or disorder.
  • It is also contemplated herein to use one or more antibodies of the present invention prophylactically to patients at risk for developing a disease or disorder such as cancer, and chronic viral infection.
  • In a further embodiment of the invention the present antibodies are used for the preparation of a pharmaceutical composition for treating patients suffering from cancer, autoimmune disease or viral infection. In another embodiment of the invention, the present antibodies are used as adjunct therapy with any other agent or any other therapy known to those skilled in the art useful for treating cancer, autoimmune disease or viral infection.
  • Combination Therapies
  • Combination therapies may include an anti-PD-L1 antibody of the invention and any additional therapeutic agent that may be advantageously combined with an antibody of the invention, or with a biologically active fragment of an antibody of the invention.
  • The antibodies of the present invention may be combined synergistically with one or more anti-cancer drugs or therapy used to treat cancer, including, for example, renal cell carcinoma, ovarian cancer, prostate cancer, colorectal cancer, non-small-cell lung cancer, and melanoma. It is contemplated herein to use anti-PD-L1 antibodies of the invention in combination with immunostimulatory and/or immunosupportive therapies to inhibit tumor growth, and/or enhance survival of cancer patients. The immunostimulatory therapies include direct immunostimulatory therapies to augment immune cell activity by either “releasing the brake” on suppressed immune cells or “stepping on the gas” to activate an immune response. Examples include targeting other checkpoint receptors, vaccination and adjuvants. The immunosupportive modalities may increase antigenicity of the tumor by promoting immunogenic cell death, inflammation or have other indirect effects that promote an anti-tumor immune response. Examples include radiation, chemotherapy, anti-angiogenic agents, and surgery.
  • In various embodiments, one or more antibodies of the present invention may be used in combination with a second antibody to PD-L1, an antibody to PD-1 (e.g., nivolumab), a LAG-3 inhibitor, a CTLA-4 inhibitor (e.g., ipilimumab), a TIM3 inhibitor, a BTLA inhibitor, a TIGIT inhibitor, a CD47 inhibitor, an antagonist of another T-cell co-inhibitor or ligand (e.g., an antibody to CD-28, 2B4, LY108, LAIR1, ICOS, CD160 or VISTA), an indoleamine-2,3-dioxygenase (IDO) inhibitor, a vascular endothelial growth factor (VEGF) antagonist [e.g., a “VEGF-Trap” such as aflibercept or other VEGF-inhibiting fusion protein as set forth in U.S. Pat. No. 7,087,411, or an anti-VEGF antibody or antigen binding fragment thereof (e.g., bevacizumab, or ranibizumab) or a small molecule kinase inhibitor of VEGF receptor (e.g., sunitinib, sorafenib, or pazopanib)], an Ang2 inhibitor (e.g., nesvacumab), a transforming growth factor beta (TGFβ) inhibitor, an epidermal growth factor receptor (EGFR) inhibitor (e.g., erlotinib, cetuximab), an agonist to a co-stimulatory receptor (e.g., an agonist to glucocorticoid-induced TNFR-related protein), an antibody to a tumor-specific antigen (e.g., CA9, CA125, melanoma-associated antigen 3 (MAGE3), carcinoembryonic antigen (CEA), vimentin, tumor-M2-PK, prostate-specific antigen (PSA), mucin-1, MART-1, and CA19-9), a vaccine (e.g., Bacillus Calmette-Guerin, a cancer vaccine), an adjuvant to increase antigen presentation (e.g., granulocyte-macrophage colony-stimulating factor), a bispecific antibody (e.g., CD3×CD20 bispecific antibody, PSMAxCD3 bispecific antibody), a cytotoxin, a chemotherapeutic agent (e.g., dacarbazine, temozolomide, cyclophosphamide, docetaxel, doxorubicin, daunorubicin, cisplatin, carboplatin, gemcitabine, methotrexate, mitoxantrone, oxaliplatin, paclitaxel, and vincristine), cyclophosphamide, radiotherapy, an IL-6R inhibitor (e.g., sarilumab), an IL-4R inhibitor (e.g., dupilumab), an IL-10 inhibitor, a cytokine such as IL-2, IL-7, IL-21, and IL-15, an antibody-drug conjugate (ADC) (e.g., anti-CD19-DM4 ADC, and anti-DS6-DM4 ADC), an anti-inflammatory drug (e.g., corticosteroids, and non-steroidal anti-inflammatory drugs), a dietary supplement such as anti-oxidants or any palliative care to treat cancer. In certain embodiments, the anti-PD-L1 antibodies of the present invention may be used in combination with cancer vaccines including dendritic cell vaccines, oncolytic viruses, tumor cell vaccines, etc. to augment the anti-tumor response. Examples of cancer vaccines that can be used in combination with anti-PD-L1 antibodies of the present invention include MAGE3 vaccine for melanoma and bladder cancer, MUC1 vaccine for breast cancer, EGFRv3 (e.g., Rindopepimut) for brain cancer (including glioblastoma multiforme), or ALVAC-CEA (for CEA+ cancers). In certain embodiments, the anti-PD-L1 antibodies of the present invention may be used in combination with a dietary supplement such as anti-oxidants or any palliative care to treat cancer.
  • In certain embodiments, the anti-PD-L1 antibodies of the invention may be administered in combination with radiation therapy in methods to generate long-term durable anti-tumor responses and/or enhance survival of patients with cancer. In some embodiments, the anti-PD-L1 antibodies of the invention may be administered prior to, concomitantly or after administering radiation therapy to a cancer patient. For example, radiation therapy may be administered in one or more doses to tumor lesions followed by administration of one or more doses of anti-PD-L1 antibodies of the invention. In some embodiments, radiation therapy may be administered locally to a tumor lesion to enhance the local immunogenicity of a patient's tumor (adjuvinating radiation) and/or to kill tumor cells (ablative radiation) followed by systemic administration of an anti-PD-L1 antibody of the invention. For example, intracranial radiation may be administered to a patient with brain cancer (e.g., glioblastoma multiforme) along with systemic administration of an anti-PD-L1 antibody of the invention. In certain embodiments, the anti-PD-L1 antibodies of the invention may be administered in combination with radiation therapy and a chemotherapeutic agent (e.g., temozolomide) or a VEGF antagonist (e.g., aflibercept).
  • The antibodies or fragments thereof of the invention may be administered in combination with one or more anti-viral drugs known in the art, including but not limited to, zidovudine, lamivudine, abacavir, ribavirin, lopinavir, efavirenz, cobicistat, tenofovir, rilpivirine and corticosteroids. In some embodiments, the anti-PD-L1 antibodies of the invention may be administered in combination with a LAG3 inhibitor, a CTLA-4 inhibitor, a PD-1 inhibitor or any antagonist of another T-cell co-inhibitor to treat chronic viral infection.
  • The antibodies of fragments thereof of the invention may be used in combination with any drug or therapy known in the art (e.g., corticosteroids and other immunosuppressants) to treat an autoimmune disease or disorder including, but not limited to, alopecia areata, autoimmune hepatitis, celiac disease, Graves' disease, Guillain-Barre syndrome, Hashimoto's disease, hemolytic anemia, inflammatory bowel disease, inflammatory myopathies, multiple sclerosis, primary biliary cirrhosis, psoriasis, rheumatoid arthritis, scleroderma, Sjögren's syndrome, systemic lupus erthyematosus, vitiligo, autoimmune pancreatitis, autoimmune urticaira, autoimmune thrombocytopenic purpura, Crohn's disease, diabetes type I, eosinophilic fasciitis, eosinophilic enterogastritis, Goodpasture's syndrome, myasthenia gravis, psoriatic arthritis, rheumatic fever, ulcerative colitis, vasculitis and Wegener's granulomatosis.
  • The additional therapeutically active component(s) may be administered prior to, concurrent with, or after the administration of the anti-PD-L1 antibody of the present invention. For purposes of the present disclosure, such administration regimens are considered the administration of an anti-PD-L1 antibody “in combination with” a second therapeutically active component.
  • The additional therapeutically active component(s) may be administered to a subject prior to administration of an anti-PD-L1 antibody of the present invention. For example, a first component may be deemed to be administered “prior to” a second component if the first component is administered 1 week before, 72 hours before, 60 hours before, 48 hours before, 36 hours before, 24 hours before, 12 hours before, 6 hours before, 5 hours before, 4 hours before, 3 hours before, 2 hours before, 1 hour before, 30 minutes before, 15 minutes before, 10 minutes before, 5 minutes before, or less than 1 minute before administration of the second component. In other embodiments, the additional therapeutically active component(s) may be administered to a subject after administration of an anti-PD-L1 antibody of the present invention. For example, a first component may be deemed to be administered “after” a second component if the first component is administered 1 minute after, 5 minutes after, 10 minutes after, 15 minutes after, 30 minutes after, 1 hour after, 2 hours after, 3 hours after, 4 hours after, 5 hours after, 6 hours after, 12 hours after, 24 hours after, 36 hours after, 48 hours after, 60 hours after, 72 hours after administration of the second component. In yet other embodiments, the additional therapeutically active component(s) may be administered to a subject concurrent with administration of an anti-PD-L1 antibody of the present invention. “Concurrent” administration, for purposes of the present invention, includes, e.g., administration of an anti-PD-L1 antibody and an additional therapeutically active component to a subject in a single dosage form (e.g., co-formulated), or in separate dosage forms administered to the subject within about 30 minutes or less of each other. If administered in separate dosage forms, each dosage form may be administered via the same route (e.g., both the anti-PD-L1 antibody and the additional therapeutically active component may be administered intravenously, subcutaneously, etc.); alternatively, each dosage form may be administered via a different route (e.g., the anti-PD-L1 antibody may be administered intravenously, and the additional therapeutically active component may be administered subcutaneously). In any event, administering the components in a single dosage from, in separate dosage forms by the same route, or in separate dosage forms by different routes are all considered “concurrent administration,” for purposes of the present disclosure. For purposes of the present disclosure, administration of an anti-PD-L1 antibody “prior to”, “concurrent with,” or “after” (as those terms are defined herein above) administration of an additional therapeutically active component is considered administration of an anti-PD-L1 antibody “in combination with” an additional therapeutically active component).
  • The present invention includes pharmaceutical compositions in which an anti-PD-L1 antibody of the present invention is co-formulated with one or more of the additional therapeutically active component(s) as described elsewhere herein using a variety of dosage combinations.
  • In exemplary embodiments in which an anti-PD-L1 antibody of the invention is administered in combination with a VEGF antagonist (e.g., a VEGF trap such as aflibercept), including administration of co-formulations comprising an anti-PD-L1 antibody and a VEGF antagonist, the individual components may be administered to a subject and/or co-formulated using a variety of dosage combinations. For example, the anti-PD-L1 antibody may be administered to a subject and/or contained in a co-formulation in an amount selected from the group consisting of 0.01 mg, 0.02 mg, 0.03 mg, 0.04 mg, 0.05 mg, 0.1 mg, 0.2 mg, 0.3 mg, 0.4 mg, 0.5 mg, 0.6 mg, 0.7 mg, 0.8 mg, 0.9 mg, 1.0 mg, 1.5 mg, 2.0 mg, 2.5 mg, 3.0 mg, 3.5 mg, 4.0 mg, 4.5 mg, 5.0 mg, 6.0 mg, 7.0 mg, 8.0 mg, 9.0 mg, and 10.0 mg; and the VEGF antagonist (e.g., a VEGF trap such as aflibercept) may be administered to the subject and/or contained in a co-formulation in an amount selected from the group consisting of 0.1 mg, 0.2 mg, 0.3 mg, 0.4 mg, 0.5 mg, 0.6 mg, 0.7 mg, 0.8 mg, 0.9 mg, 1.0 mg, 1.1 mg, 1.2 mg, 1.3 mg, 1.4 mg, 1.5 mg, 1.6 mg, 1.7 mg, 1.8 mg, 1.9 mg, 2.0 mg, 2.1 mg, 2.2 mg, 2.3 mg, 2.4 mg, 2.5 mg, 2.6 mg, 2.7 mg, 2.8 mg, 2.9 mg and 3.0 mg. The combinations/co-formulations may be administered to a subject according to any of the administration regimens disclosed elsewhere herein, including, e.g., twice a week, once every week, once every 2 weeks, once every 3 weeks, once every month, once every 2 months, once every 3 months, once every 4 months, once every 5 months, once every 6 months, etc.
  • Administrative Regimens
  • According to certain embodiments of the present invention, multiple doses of an anti-PD-L1 antibody (or a pharmaceutical composition comprising a combination of an anti-PD-L1 antibody and any of the additional therapeutically active agents mentioned herein) may be administered to a subject over a defined time course. The methods according to this aspect of the invention comprise sequentially administering to a subject multiple doses of an anti-PD-L1 antibody of the invention. As used herein, “sequentially administering” means that each dose of anti-PD-L1 antibody is administered to the subject at a different point in time, e.g., on different days separated by a predetermined interval (e.g., hours, days, weeks or months). The present invention includes methods which comprise sequentially administering to the patient a single initial dose of an anti-PD-L1 antibody, followed by one or more secondary doses of the anti-PD-L1 antibody, and optionally followed by one or more tertiary doses of the anti-PD-L1 antibody. The anti-PD-L1 antibody may be administered at a dose of between 0.1 mg/kg to about 100 mg/kg.
  • The terms “initial dose,” “secondary doses,” and “tertiary doses,” refer to the temporal sequence of administration of the anti-PD-L1 antibody of the invention. Thus, the “initial dose” is the dose which is administered at the beginning of the treatment regimen (also referred to as the “baseline dose”); the “secondary doses” are the doses which are administered after the initial dose; and the “tertiary doses” are the doses which are administered after the secondary doses. The initial, secondary, and tertiary doses may all contain the same amount of anti-PD-L1 antibody, but generally may differ from one another in terms of frequency of administration.
  • In certain embodiments, however, the amount of anti-PD-L1 antibody contained in the initial, secondary and/or tertiary doses varies from one another (e.g., adjusted up or down as appropriate) during the course of treatment. In certain embodiments, two or more (e.g., 2, 3, 4, or 5) doses are administered at the beginning of the treatment regimen as “loading doses” followed by subsequent doses that are administered on a less frequent basis (e.g., “maintenance doses”).
  • In certain exemplary embodiments of the present invention, each secondary and/or tertiary dose is administered 1 to 26 (e.g., 1, 1%, 2, 2%, 3, 3%, 4, 4%, 5, 5%, 6, 6%, 7, 7%, 8, 8%, 9, 9%, 10, 10%, 11, 11%, 12, 12%, 13, 13%, 14, 14%, 15, 15%, 16, 16%, 17, 17%, 18, 18%, 19, 19%, 20, 20%, 21, 21%, 22, 22%, 23, 23%, 24, 24%, 25, 25%, 26, 26%, or more) weeks after the immediately preceding dose. The phrase “the immediately preceding dose,” as used herein, means, in a sequence of multiple administrations, the dose of anti-PD-L1 antibody which is administered to a patient prior to the administration of the very next dose in the sequence with no intervening doses.
  • The methods according to this aspect of the invention may comprise administering to a patient any number of secondary and/or tertiary doses of an anti-PD-L1 antibody. For example, in certain embodiments, only a single secondary dose is administered to the patient. In other embodiments, two or more (e.g., 2, 3, 4, 5, 6, 7, 8, or more) secondary doses are administered to the patient. Likewise, in certain embodiments, only a single tertiary dose is administered to the patient. In other embodiments, two or more (e.g., 2, 3, 4, 5, 6, 7, 8, or more) tertiary doses are administered to the patient.
  • In embodiments involving multiple secondary doses, each secondary dose may be administered at the same frequency as the other secondary doses. For example, each secondary dose may be administered to the patient 1 to 2 weeks or 1 to 2 months after the immediately preceding dose. Similarly, in embodiments involving multiple tertiary doses, each tertiary dose may be administered at the same frequency as the other tertiary doses. For example, each tertiary dose may be administered to the patient 2 to 12 weeks after the immediately preceding dose. In certain embodiments of the invention, the frequency at which the secondary and/or tertiary doses are administered to a patient can vary over the course of the treatment regimen. The frequency of administration may also be adjusted during the course of treatment by a physician depending on the needs of the individual patient following clinical examination.
  • The present invention includes administration regimens in which 2 to 6 loading doses are administered to a patient at a first frequency (e.g., once a week, once every two weeks, once every three weeks, once a month, once every two months, etc.), followed by administration of two or more maintenance doses to the patient on a less frequent basis. For example, according to this aspect of the invention, if the loading doses are administered at a frequency of, e.g., once a month (e.g., two, three, four, or more loading doses administered once a month), then the maintenance doses may be administered to the patient once every five weeks, once every six weeks, once every seven weeks, once every eight weeks, once every ten weeks, once every twelve weeks, etc.).
  • Diagnostic Uses of the Antibodies
  • The anti-PD-L1 antibodies of the present invention may be used to detect and/or measure PD-L1 in a sample, e.g., for diagnostic purposes. Some embodiments contemplate the use of one or more antibodies of the present invention in assays to detect a disease or disorder such as cancer, autoimmune disease or chronic viral infection. Exemplary diagnostic assays for PD-L1 may comprise, e.g., contacting a sample, obtained from a patient, with an anti-PD-L1 antibody of the invention, wherein the anti-PD-L1 antibody is labeled with a detectable label or reporter molecule or used as a capture ligand to selectively isolate PD-L1 from patient samples. Alternatively, an unlabeled anti-PD-L1 antibody can be used in diagnostic applications in combination with a secondary antibody which is itself detectably labeled. The detectable label or reporter molecule can be a radioisotope, such as 3H, 14C, 32P, 35S, or 125I; a fluorescent or chemiluminescent moiety such as fluorescein isothiocyanate, or rhodamine; or an enzyme such as alkaline phosphatase, β-galactosidase, horseradish peroxidase, or luciferase. Specific exemplary assays that can be used to detect or measure PD-L1 in a sample include enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), and fluorescence-activated cell sorting (FACS).
  • Samples that can be used in PD-L1 diagnostic assays according to the present invention include any tissue or fluid sample obtainable from a patient, which contains detectable quantities of either PD-L1 protein, or fragments thereof, under normal or pathological conditions. Generally, levels of PD-L1 in a particular sample obtained from a healthy patient (e.g., a patient not afflicted with cancer or an autoimmune disease) will be measured to initially establish a baseline, or standard, level of PD-L1. This baseline level of PD-L1 can then be compared against the levels of PD-L1 measured in samples obtained from individuals suspected of having a cancer-related condition, or symptoms associated with such condition.
  • The antibodies specific for PD-L1 may contain no additional labels or moieties, or they may contain an N-terminal or C-terminal label or moiety. In one embodiment, the label or moiety is biotin. In a binding assay, the location of a label (if any) may determine the orientation of the peptide relative to the surface upon which the peptide is bound. For example, if a surface is coated with avidin, a peptide containing an N-terminal biotin will be oriented such that the C-terminal portion of the peptide will be distal to the surface.
  • Aspects of the invention relate to use of the disclosed antibodies as markers for predicting prognosis of cancer or an autoimmune disorder in patients. Antibodies of the present invention may be used in diagnostic assays to evaluate prognosis of cancer in a patient and to predict survival.
  • Examples
  • The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the methods and compositions of the invention, and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is average molecular weight, temperature is in degrees Centigrade, room temperature is about 25° C., and pressure is at or near atmospheric.
  • Example 1: Generation of Human Antibodies to PD-L1
  • Human antibodies to PD-L1 were generated using a fragment of PD-L1 that ranges from about amino acids 19-239 of SEQ ID NO: 351 (Genbank Accession No. NP_054862.1). The immunogen was administered directly, with an adjuvant to stimulate the immune response, to a VELOCIMMUNE® mouse comprising DNA encoding human Immunoglobulin heavy and kappa light chain variable regions. The antibody immune response was monitored by a PD-L1-specific immunoassay. When a desired immune response was achieved splenocytes were harvested and fused with mouse myeloma cells to preserve their viability and form hybridoma cell lines. The hybridoma cell lines were screened and selected to identify cell lines that produce PD-L1-specific antibodies. Using this technique, and the immunogen described above, several anti-PD-L1 chimeric antibodies (i.e., antibodies possessing human variable domains and mouse constant domains) were obtained; exemplary antibodies generated in this manner were designated as H2M8306N, H2M8307N, H2M8309N, H2M8310N, H2M8312N, H2M8314N, H2M8316N, H2M8317N, H2M8321N, H2M8323N, H2M8718N, H2M8718N2, and H2M8719N.
  • Anti-PD-L1 antibodies were also isolated directly from antigen-positive B cells without fusion to myeloma cells, as described in U.S. 2007/0280945A1, herein specifically incorporated by reference in its entirety. Using this method, several fully human anti-PD-L1 antibodies (i.e., antibodies possessing human variable domains and human constant domains) were obtained; exemplary antibodies generated in this manner were designated as follows: H1H9323P, H1H9327P, H1H9329P, H1H9336P, H1H9344P2, H1H9345P2, H1H9351P2, H1H9354P2, H1H9364P2, H1H9373P2, H1H9382P2, H1H9387P2, and H1H9396P2.
  • The biological properties of the exemplary antibodies generated in accordance with the methods of this Example are described in detail in the Examples set forth below.
  • Example 2: Heavy and Light Chain Variable Region Amino Acid and Nucleotide Sequences
  • Table 1 sets forth the amino acid sequence identifiers of the heavy and light chain variable regions and CDRs of selected anti-PD-L1 antibodies of the invention. The corresponding nucleic acid sequence identifiers are set forth in Table 2.
  • TABLE 1
    Amino Acid Sequence Identifiers
    Antibody SEQ ID NOs:
    Designation HCVR HCDR1 HCDR2 HCDR3 LCVR LCDR1 LCDR2 LCDR3
    H2M8306N 2 4 6 8 10 12 14 16
    H2M8307N 18 20 22 24 26 28 30 32
    H2M8309N 34 36 38 40 42 44 46 48
    H2M8310N 50 52 54 56 58 60 62 64
    H2M8312N 66 68 70 72 74 76 78 80
    H2M8314N 82 84 86 88 90 92 94 96
    H2M8316N 98 100 102 104 106 108 110 112
    H2M8317N 114 116 118 120 122 124 126 128
    H2M8321N 130 132 134 136 138 140 142 144
    H2M8323N 146 148 150 152 154 156 158 160
    H2M8718N 162 164 166 168 170 172 174 176
    H2M8718N2 178 180 182 184 170 172 174 176
    H2M8719N 186 188 190 192 194 196 198 200
    H1H9323P 202 204 206 208 210 212 214 216
    H1H9327P 218 220 222 224 226 228 230 232
    H1H9329P 234 236 238 240 242 244 246 248
    H1H9336P 250 252 254 256 258 260 262 264
    H1H9344P2 266 268 270 272 274 276 278 280
    H1H9345P2 282 284 286 288 274 276 278 280
    H1H9351P2 290 292 294 296 274 276 278 280
    H1H9354P2 298 300 302 304 274 276 278 280
    H1H9364P2 306 308 310 312 274 276 278 280
    H1H9373P2 314 316 318 320 274 276 278 280
    H1H9382P2 322 324 326 328 274 276 278 280
    H1H9387P2 330 332 334 336 274 276 278 280
    H1H9396P2 338 340 342 344 274 276 278 280
  • TABLE 2
    Nucleic Acid Sequence Identifiers
    Antibody SEQ ID NOs:
    Designation HCVR HCDR1 HCDR2 HCDR3 LCVR LCDR1 LCDR2 LCDR3
    H2M8306N 1 3 5 7 9 11 13 15
    H2M8307N 17 19 21 23 25 27 29 31
    H2M8309N 33 35 37 39 41 43 45 47
    H2M8310N 49 51 53 55 57 59 61 63
    H2M8312N 65 67 69 71 73 75 77 79
    H2M8314N 81 83 85 87 89 91 93 95
    H2M8316N 97 99 101 103 105 107 109 111
    H2M8317N 113 115 117 119 121 123 125 127
    H2M8321N 129 131 133 135 137 139 141 143
    H2M8323N 145 147 149 151 153 155 157 159
    H2M8718N 161 163 165 167 169 171 173 175
    H2M8718N2 177 179 181 183 169 171 173 175
    H2M8719N 185 187 189 191 193 195 197 199
    H1H9323P 201 203 205 207 209 211 213 215
    H1H9327P 217 219 221 223 225 227 229 231
    H1H9329P 233 235 237 239 241 243 245 247
    H1H9336P 249 251 253 255 257 259 261 263
    H1H9344P2 265 267 269 271 273 275 277 279
    H1H9345P2 281 283 285 287 273 275 277 279
    H1H9351P2 289 291 293 295 273 275 277 279
    H1H9354P2 297 299 301 303 273 275 277 279
    H1H9364P2 305 307 309 311 273 275 277 279
    H1H9373P2 313 315 317 319 273 275 277 279
    H1H9382P2 321 323 325 327 273 275 277 279
    H1H9387P2 329 331 333 335 273 275 277 279
    H1H9396P2 337 339 341 343 273 275 277 279
  • Antibodies are typically referred to herein according to the following nomenclature: Fc prefix (e.g. “H1H,” “H2M,” “H2aM,” etc.), followed by a numerical identifier (e.g. “8306,” “9323,” etc., as shown in Table 1), followed by a “P,” “N,” “P2,” or “N2” suffix. Thus, according to this nomenclature, an antibody may be referred to herein as, e.g., “H2M8306N,” “H1H9344P2,” etc. The H1H, H2M and H2aM prefixes on the antibody designations used herein indicate the particular Fc region isotype of the antibody. For example, an “H1H” antibody has a human IgG1 Fc, an “HIM” antibody has a mouse IgG1 Fc, and an “H2M” or “H2aM” antibody has a mouse IgG2 Fc, (all variable regions are fully human as denoted by the first ‘H’ in the antibody designation). As will be appreciated by a person of ordinary skill in the art, an antibody having a particular Fc isotype can be converted to an antibody with a different Fc isotype (e.g., an antibody with a mouse IgG1 Fc can be converted to an antibody with a human IgG4, etc.), but in any event, the variable domains (including the CDRs)—which are indicated by the numerical identifiers shown in Table 1—will remain the same, and the binding properties to antigen are expected to be identical or substantially similar regardless of the nature of the Fc domain.
  • Example 3: Antibody Binding to PD-L1 as Determined by Surface Plasmon Resonance
  • Binding association and dissociation rate constants (ka and kd, respectively), equilibrium dissociation constants and dissociation half-lives (KD and t1/2, respectively) of antigen binding to purified anti-PD-L1 monoclonal antibodies were determined using a real-time surface plasmon resonance biosensor assay on a Biacore 4000 instrument. The Biacore sensor surface was either derivatized with polyclonal rabbit anti-mouse antibody (GE, #BR-1008-38) or with monoclonal mouse anti-human Fc antibody (GE, #BR-1008-39) to capture approximately 200-300 RUs of anti-PD-L1 monoclonal antibodies, expressed with either a mouse Fc or with human Fc, respectively. The PD-L1 reagents tested for binding to the anti-PD-L1 antibodies included recombinant human PD-L1 (amino acids 19-239 of accession number NP_054862.1) expressed with a C-terminal myc-myc-hexahistidine tag (hPD-L1-MMH; SEQ ID: 345), recombinant cynomolgus monkey PD-L1 expressed with a C-terminal myc-myc-hexahistidine tag (MfPD-L1-MMH; SEQ ID: 346), recombinant human PD-L1 (amino acids 19-239 of accession number NP_054862.1) expressed with either a C-terminal human IgG1 Fc tag (hPD-L1-hFc; SEQ ID: 347) or with a C-terminal mouse IgG2a Fc tag (hPD-L1-mFc; SEQ ID: 348), and recombinant cynomolgus monkey PD-L1 expressed with a C-terminal mouse IgG2a Fc tag (MfPD-L1-mFc; SEQ ID: 353). Different concentrations of PD-L1 reagents were injected over the anti-PD-L1 monoclonal antibody captured surface at a flow rate of 30 μL/min. The binding of the PD-L1 reagents to the captured monoclonal antibodies was monitored for 3 to 4 minutes while the dissociation of antibody bound PD-L1 reagents was monitored for 10 minutes in HBST running buffer (0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.05% v/v Surfactant tween-20). Experiments were performed at either 25° C. or 37° C. Kinetic association (ka) and dissociation (kd) rate constants were determined by processing and fitting the data to a 1:1 binding model using Scrubber 2.0c curve fitting software. Binding dissociation equilibrium constants (KD) and dissociative half-lives (t1/2) were then calculated from the kinetic rate constants as: KD (M)=kd/ka and t1/2 (min)=[In2/(60*kd)]. Binding kinetics parameters for different anti-PD-L1 monoclonal antibodies binding to different PD-L1 reagents at 25° C. and 37° C. are tabulated in Tables 3-8.
  • TABLE 3
    Binding Kinetics parameters of anti-PD-L1
    monoclonal antibodies binding to
    hPD-L1-MMH at 25° C.
    hPD-L1-MMH Monomer Binding at 25° C.
    ka kd KD t1/2
    Antibody (1/Ms) (1/s) (M) (min)
    H2aM8306N 1.44E+05 1.00E−02 6.96E−08 1.2
    H2aM8307N 3.27E+04 1.47E−04 4.50E−09 79
    H2aM8309N 6.42E+05 1.04E−02 1.63E−08 1.1
    H2aM8310N 8.86E+04 3.10E−04 3.50E−09 37
    H2aM8312N 7.59E+04 7.22E−04 9.51E−09 16
    H2aM8314N 9.25E+05 5.17E−05 5.58E−11 224
    H2aM8316N 9.57E+05 1.12E−03 1.17E−09 10
    H2aM8317N 9.40E+04 3.95E−02 4.21E−07 0.3
    H2aM8321N 1.03E+05 3.59E−03 3.49E−08 3.2
    H2aM8323N 9.37E+05 2.23E−04 2.38E−10 52
    H2aM8718N 9.27E+05 8.01E−05 8.63E−11 144
    H2aM8719N 8.64E+04 2.26E−03 2.62E−08 5.1
    H1H9323P 5.51E+04 1.88E−02 3.41E−07 0.6
    H1H9327P 4.19E+05 2.59E−04 6.19E−10 45
    H1H9329P 1.77E+06 1.34E−01 7.58E−08 0.1
    H1H9336P 7.92E+05 3.90E−04 4.92E−10 30
    H1H9345P2 9.02E+04 1.49E−02 1.65E−07 0.8
    H1H9351P2 4.56E+05 8.96E−04 1.96E−09 13
    H1H9354P2 4.76E+04 3.83E−04 8.04E−09 30
    H1H9364P2 9.32E+05 2.99E−04 3.21E−10 39
    H1H9373P2 2.65E+06 2.91E−04 1.10E−10 40
    H1H9382P2 7.90E+04 4.79E−03 6.06E−08 2.4
    H1H9387P2 2.64E+06 5.82E−02 2.21E−08 0.2
    H1H9396P2 1.72E+05 2.18E−03 1.27E−08 5.3
  • TABLE 4
    Binding Kinetics parameters of anti-PD-L1 monoclonal
    antibodies binding to hPD-L1-MMH at 37° C.
    hPD-L1-MMH Monomer Binding at 37° C.
    ka kd KD t1/2
    Antibody (1/Ms) (1/s) (M) (min)
    H2aM8306N 1.80E+05 3.10E−02 1.72E−07 0.4
    H2aM8307N 5.35E+04 8.79E−04 1.64E−08 13
    H2aM8309N 1.06E+06 3.14E−02 2.97E−08 0.4
    H2aM8310N 1.32E+05 1.28E−03 9.70E−09 9.0
    H2aM8312N 8.89E+04 4.03E−03 4.53E−08 2.9
    H2aM8314N 1.07E+06 1.50E−04 1.40E−10 77
    H2aM8316N 1.01E+06 5.30E−03 5.24E−09 2.2
    H2aM8317N 9.03E+04 5.85E−02 6.47E−07 0.2
    H2aM8321N 1.01E+05 9.29E−03 9.16E−08 1.2
    H2aM8323N 1.38E+06 6.84E−04 4.97E−10 17
    H2aM8718N 1.08E+06 1.55E−04 1.44E−10 74
    H2aM8719N 1.50E+05 5.76E−03 3.84E−08 2.0
    H1H9323P 1.21E+05 4.25E−02 3.52E−07 0.3
    H1H9327P 5.21E+05 4.29E−04 8.24E−10 27
    H1H9329P 2.82E+06 3.29E−01 1.17E−07 0.04
    H1H9336P 1.07E+06 7.88E−04 7.33E−10 15
    H1H9345P2 1.72E+05 3.40E−02 1.97E−07 0.3
    H1H9351P2 6.82E+05 1.68E−03 2.47E−09 6.9
    H1H9354P2 7.39E+04 1.14E−03 1.54E−08 10
    H1H9364P2 1.35E+06 5.63E−04 4.17E−10 21
    H1H9373P2 3.09E+06 5.58E−04 1.80E−10 21
    H1H9382P2 9.97E+04 1.07E−02 1.07E−07 1.1
    H1H9387P2 3.49E+06 1.37E−01 3.91E−08 0.08
    H1H9396P2 3.44E+05 1.13E−02 3.30E−08 1.0
  • TABLE 5
    Binding Kinetics parameters of anti-PD-L1 monoclonal
    antibodies binding to human PD-L1 Dimer at 25° C.
    Human PD-L1 Dimer Binding at 25° C.
    ka kd KD t1/2
    Antibody (1/Ms) (1/s) (M) (min)
    H2aM8306N 7.39E+05 3.09E−04 4.18E−10 37
    H2aM8307N 1.89E+04 2.89E−05 1.54E−09 399
    H2aM8309N 3.36E+06 1.44E−04 4.29E−11 80
    H2aM8310N 3.13E+05 5.71E−05 1.83E−10 202
    H2aM8312N 2.47E+05 1.02E−04 4.13E−10 113
    H2aM8314N 3.16E+06 1.35E−05 4.26E−12 859
    H2aM8316N 3.08E+06 1.44E−04 4.68E−11 80
    H2aM8317N 3.59E+05 4.50E−04 1.25E−09 26
    H2aM8321N 8.13E+05 2.87E−04 3.53E−10 40
    H2aM8323N 2.91E+06 2.05E−05 7.04E−12 565
    H2aM8718N 3.20E+06 1.62E−05 5.06E−12 713
    H2aM8719N 3.42E+05 2.62E−04 7.67E−10 44
    H1H9323P 2.24E+05 1.69E−04 7.54E−10 68
    H1H9327P 4.66E+05 7.87E−05 1.69E−10 147
    H1H9329P 2.97E+06 7.68E−04 2.59E−10 15
    H1H9336P 1.38E+06 1.09E−04 7.86E−11 106
    H1H9345P2 5.00E+05 2.37E−04 4.74E−10 49
    H1H9351P2 9.16E+05 1.53E−04 1.67E−10 76
    H1H9354P2 1.68E+05 1.16E−04 6.89E−10 100
    H1H9364P2 2.42E+06 1.06E−04 4.37E−11 109
    H1H9373P2 4.08E+06 1.06E−04 2.60E−11 109
    H1H9382P2 2.23E+05 1.48E−04 6.63E−10 78
    H1H9387P2 5.07E+06 2.17E−04 4.27E−11 53
    H1H9396P2 7.76E+05 1.90E−04 2.45E−10 61
  • TABLE 6
    Binding Kinetics parameters of anti-PD-L1 monoclonal
    antibodies binding to human PD-L1 Dimer at 37° C.
    Human PD-L1 Dimer Binding at 37° C.
    ka kd KD t1/2
    Antibody (1/Ms) (1/s) (M) (min)
    H2aM8306N 9.97E+05 4.16E−04 4.17E−10 28
    H2aM8307N 4.41E+04 2.21E−04 5.00E−09 52
    H2aM8309N 4.41E+06 1.66E−04 3.76E−11 70
    H2aM8310N 4.81E+05 1.24E−04 2.58E−10 93
    H2aM8312N 3.57E+05 2.61E−04 7.32E−10 44
    H2aM8314N 3.89E+06 2.30E−05 5.91E−12 503
    H2aM8316N 4.06E+06 2.37E−04 5.85E−11 49
    H2aM8317N 4.81E+05 6.57E−04 1.36E−09 18
    H2aM8321N 9.46E+05 2.69E−04 2.85E−10 43
    H2aM8323N 4.32E+06 1.21E−04 2.80E−11 96
    H2aM8718N 3.72E+06 1.98E−05 5.32E−12 584
    H2aM8719N 4.37E+05 2.91E−04 6.66E−10 40
    H1H9323P 5.19E+05 2.03E−04 3.91E−10 57
    H1H9327P 6.83E+05 1.36E−04 2.00E−10 85
    H1H9329P 3.87E+06 2.67E−03 6.89E−10 4.3
    H1H9336P 2.75E+06 8.31E−05 3.02E−11 139
    H1H9345P2 6.82E+05 2.03E−04 2.97E−10 57
    H1H9351P2 1.25E+06 1.46E−04 1.17E−10 79
    H1H9354P2 4.56E+05 1.45E−04 3.17E−10 80
    H1H9364P2 3.34E+06 6.96E−05 2.08E−11 166
    H1H9373P2 5.12E+06 8.97E−05 1.75E−11 129
    H1H9382P2 4.92E+05 1.37E−04 2.78E−10 84
    H1H9387P2 6.12E+06 3.92E−04 6.39E−11 29
    H1H9396P2 1.09E+06 2.58E−04 2.37E−10 45
  • TABLE 7
    Binding Kinetics parameters of anti-PD-L1 monoclonal
    antibodies binding to MfPD-L1-MMH at 25° C.
    MfPD-L1-MMH Monomer Binding at 25° C.
    ka kd KD t1/2
    Antibody (1/Ms) (1/s) (M) (min)
    H2aM8306N 1.65E+05 7.88E−03 4.77E−08 1.5
    H2aM8307N 2.61E+04 1.07E−03 4.09E−08 11
    H2aM8309N 8.70E+05 1.30E−02 1.49E−08 0.9
    H2aM8310N 1.03E+05 3.06E−04 2.97E−09 38
    H2aM8312N 7.09E+04 5.97E−04 8.42E−09 19
    H2aM8314N 9.66E+05 7.00E−05 7.24E−11 165
    H2aM8316N 9.71E+05 1.64E−03 1.69E−09 7.0
    H2aM8317N 1.06E+05 2.38E−02 2.26E−07 0.5
    H2aM8321N 1.34E+05 4.02E−03 2.99E−08 2.9
    H2aM8323N 5.47E+05 8.68E−03 1.59E−08 1.3
    H2aM8718N 9.04E+05 6.64E−05 7.35E−11 174
    H2aM8719N 8.17E+04 2.68E−03 3.28E−08 4.3
    H1H9323P 8.22E+04 2.40E−02 2.92E−07 0.5
    H1H9327P 3.59E+05 3.33E−04 9.28E−10 35
    H1H9329P 1.76E+06 1.35E−01 7.69E−08 0.09
    H1H9336P 6.79E+05 5.94E−04 8.76E−10 19
    H1H9345P2 1.10E+05 8.50E−03 7.73E−08 1.4
    H1H9351P2 3.49E+05 1.11E−03 3.19E−09 10
    H1H9354P2 4.60E+04 3.05E−04 6.64E−09 38
    H1H9364P2 7.57E+05 3.12E−04 4.12E−10 37
    H1H9373P2 2.21E+06 2.82E−04 1.27E−10 41
    H1H9382P2 8.22E+04 1.29E−02 1.57E−07 0.9
    H1H9387P2 2.37E+06 6.04E−02 2.55E−08 0.2
    H1H9396P2 2.06E+05 2.52E−03 1.22E−08 4.6
  • TABLE 8
    Binding Kinetics parameters of anti-PD-L1 monoclonal
    antibodies binding to MfPD-L1-MMH at 37° C.
    MfPD-L1-MMH Monomer Binding at 37° C.
    ka kd KD t1/2
    Antibody (1/Ms) (1/s) (M) (min)
    H2aM8306N 2.74E+05 3.68E−02 1.34E−07 0.3
    H2aM8307N 1.87E+05 1.43E−03 7.63E−09 8.1
    H2aM8309N 1.07E+06 2.81E−02 2.63E−08 0.4
    H2aM8310N 4.71E+05 1.07E−03 2.27E−09 11
    H2aM8312N 1.01E+05 3.32E−03 3.27E−08 3.5
    H2aM8314N 1.07E+06 1.52E−04 1.42E−10 76
    H2aM8316N 1.02E+06 5.38E−03 5.27E−09 2.1
    H2aM8317N 2.66E+05 4.76E−02 1.79E−07 0.2
    H2aM8321N 1.59E+05 8.16E−03 5.11E−08 1.4
    H2aM8323N 9.56E+05 2.82E−02 2.95E−08 0.4
    H2aM8718N 1.10E+06 1.46E−04 1.33E−10 79
    H2aM8719N 1.35E+05 6.99E−03 5.19E−08 1.7
    H1H9323P 1.25E+05 4.99E−02 3.98E−07 0.2
    H1H9327P 4.77E+05 5.34E−04 1.12E−09 22
    H1H9329P 2.66E+06 3.64E−01 1.37E−07 0.03
    H1H9336P 9.09E+05 1.25E−03 1.38E−09 9.2
    H1H9345P2 1.64E+05 1.97E−02 1.21E−07 0.6
    H1H9351P2 5.60E+05 2.01E−03 3.59E−09 5.8
    H1H9354P2 8.59E+04 8.44E−04 9.82E−09 14
    H1H9364P2 1.12E+06 6.33E−04 5.66E−10 18
    H1H9373P2 2.81E+06 5.69E−04 2.03E−10 20
    H1H9382P2 1.30E+05 2.45E−02 1.89E−07 0.5
    H1H9387P2 3.20E+06 1.57E−01 4.89E−08 0.07
    H1H9396P2 3.94E+05 1.20E−02 3.05E−08 1.0
  • TABLE 9
    Binding Kinetics parameters of anti-PD-L1 monoclonal
    antibodies binding to monkey PD-L1-mFc at 25° C.
    MfPD-L1-mFc Binding at 25° C.
    ka kd KD t1/2
    Antibody (1/Ms) (1/s) (M) (min)
    H1H8314N 2.58E+06 6.77E−05 2.63E−11 171
    H1H8316N 2.71E+06 5.23E−05 1.93E−11 221
    H1H8323N 2.67E+06 6.16E−05 2.31E−11 188
    H1H9351P2 7.81E+05 7.19E−05 9.22E−11 161
    H1H9364P2 1.32E+06 7.75E−05 5.87E−11 149
    H1H9373P2 2.85E+06 4.96E−05 1.74E−11 233
    H1H9387P2 3.55E+06 1.61E−04 4.52E−11 72
    H1H9351P2 8.64E+05 7.80E−05 9.03E−11 148
    H1H9364P2 1.25E+06 5.80E−05 4.62E−11 199
    H1H9373P2 3.27E+06 6.55E−05 2.00E−11 176
    H1H9387P2 2.90E+06 2.12E−04 7.30E−11 55
    H1H9323P 2.80E+05 1.45E−04 5.19E−10 79
    H1H9327P 3.48E+05 1.02E−04 2.94E−10 113
    H1H9329P 1.74E+06 6.72E−04 3.86E−10 17
    H1H9336P 9.90E+05 6.16E−05 6.22E−11 188
    H1H9344P2 3.50E+05 1.38E−04 3.93E−10 84
    H1H9345P2 2.96E+05 1.51E−04 5.12E−10 76
    H1H9354P2 2.05E+05 9.14E−05 4.47E−10 126
    H1H9382P2 2.13E+05 1.33E−04 6.26E−10 87
    H1H9396P2 8.37E+05 1.88E−04 2.25E−10 61
  • TABLE 10
    Binding Kinetics parameters of anti-PD-L1 monoclonal
    antibodies binding to monkey PD-L1-mFc at 37° C.
    MfPD-L1-mFc Binding at 37° C.
    ka kd KD t1/2
    Antibody (1/Ms) (1/s) (M) (min)
    H1H8314N 2.89E+06 9.60E−05 3.33E−11 120
    H1H8316N 2.96E+06 6.16E−05 2.08E−11 187
    H1H8323N 2.99E+06 1.35E−04 4.51E−11 86
    H1H9351P2 1.06E+06 9.46E−05 8.91E−11 122
    H1H9364P2 2.36E+06 1.11E−04 4.71E−11 104
    H1H9373P2 3.15E+06 8.59E−05 2.73E−11 134
    H1H9387P2 3.41E+06 4.74E−04 1.39E−10 24
    H1H9351P2 1.61E+06 1.04E−04 6.47E−11 111
    H1H9364P2 2.41E+06 6.76E−05 2.80E−11 171
    H1H9373P2 3.86E+06 1.23E−04 3.19E−11 94
    H1H9387P2 2.90E+06 4.65E−04 1.61E−10 25
    H1H9323P 3.84E+05 2.44E−04 6.36E−10 47
    H1H9327P 7.64E+05 2.94E−04 3.85E−10 39
    H1H9329P 2.18E+06 1.54E−03 7.08E−10 8
    H1H9336P 1.86E+06 4.60E−05 2.47E−11 251
    H1H9344P2 9.05E+05 2.17E−04 2.40E−10 53
    H1H9345P2 8.61E+05 2.92E−04 3.39E−10 40
    H1H9354P2 2.72E+05 2.03E−04 7.46E−10 57
    H1H9382P2 2.84E+05 2.35E−04 8.25E−10 49
    H1H9396P2 1.57E+06 5.02E−04 3.19E−10 23
  • As shown in Table 3, at 25° C., all 25 anti-PD-L1 antibodies of the invention bound to hPD-L1-MMH with KD values ranging from 55.8 pM to 421 nM. As shown in Table 4, at 37° C., all 25 anti-PD-L1 antibodies of the invention bound to hPD-L1-MMH with KD values ranging from 140 pM to 647 nM. As shown in Table 5, at 25° C., all 25 anti-PD-L1 antibodies of the invention bound to hPD-L1 dimer with KD values ranging from 4.26 pM to 1.54 nM. As shown in Table 6, at 37° C., all 25 anti-PD-L1 antibodies of the invention bound to hPD-L1 dimer with KD values ranging from 5.32 pM to 5.0 nM. As shown in Table 7, at 25° C., all 25 anti-PD-L1 antibodies of the invention bound to MfPD-L1-MMH with KD values ranging from 72.4 pM to 292 nM. As shown in Table 8, at 37° C., all 25 anti-PD-L1 antibodies of the invention bound to MfPD-L1-MMH with KD values ranging from 133 pM to 398 nM. As shown in Table 9, at 25° C., all 20 anti-PD-L1 antibodies of the invention tested bound to MfPD-L1-mFc with KD values ranging from 17.4 pM to 626 pM. As shown in Table 10, at 37° C., all 20 anti-PD-L1 antibodies of the invention tested bound to MfPD-L1-mFc with KD values ranging from 20.8 pM to 825 pM.
  • Example 4: Blocking of PD-L1 Binding to PD-1 as Determined by ELISA
  • The ability of monoclonal anti-PD-L1 antibodies to block human PD-L1 from binding to its binding partners, the human PD-1 and the human B7-1 receptors, was measured using two competition sandwich ELISA formats.
  • A dimeric human PD-1 protein comprised of a portion of the extracellular domain (amino acids 25-170 of accession number NP_005009.2 with a C93S change) that was expressed with a C-terminal hFc tag (hPD-1-hFc; SEQ ID: 350) and a dimeric human B7-1 protein comprised of a portion of the extracellular domain expressed with C-terminal hFc tag and hexahistidine tags (hB7-1-hFc-6His; R&D Systems, #140-B1) were separately coated at 2 μg/mL on a 96-well microtiter plate in a PBS buffer overnight at 4° C. Nonspecific binding sites were subsequently blocked using a 0.5% (w/v) solution of BSA in PBS. A constant amount of 0.5 nM or 8.0 nM of a dimeric hPD-L1 protein comprised of a portion of the human PD-L1 extracellular domain that was expressed with a C-terminal mFc tag (hPD-L1-mFc; SEQ ID: 348) was separately titrated with concentrations of anti-PD-1 antibodies and isotype control antibodies ranging between 0-210 nM in serial dilution. These antibody-protein complexes were then incubated for 1 hour at room temperature (RT). Complexes with 0.5 nM constant hPD-L1-mFc were subsequently transferred to microtiter plates coated with hPD-1-hFc, and complexes with 8 nM constant hPD-L1-mFc were transferred to hB7-1-hFc-6His coated plates. The complexes were allowed to bind to the coated plates for 1 hour at RT. After the 1 hour incubation, the wells were washed and plate-bound hPD-L1-mFc was detected with an anti-mFc polyclonal antibody conjugated with horse-radish peroxidase (Jackson ImmunoResearch, #115-035-164). Samples were developed with a TMB solution (BD Biosciences, #51-2606KC and #51-2607KC) to produce a colorimetric reaction and neutralized with 1M sulfuric acid before measuring absorbance at 450 nm on a Victor X5 plate reader. Data analysis used a sigmoidal dose-response model within Prism™ software. The calculated IC50 value, defined as the concentration of antibody required to block 50% of hPD-L1-mFc binding to hPD-1-hFc or hB7-1-hFc-6His, was used as an indicator of blocking potency. Maximum blocking values represent the ability of the antibodies to block hPD-L1-mFc binding relative to baseline. The absorbance measured at the constant amount of hPD-L1 on the dose curve is defined as 0% blocking and the absorbance with no added hPD-L1 is defined as 100% blocking. The absorbance values of the wells containing the highest concentration for each antibody were used to determine the blocking percent at maximum concentration antibody tested. Antibodies with a maximum percent blockade below 25% were characterized as non-blockers, and their IC50 values were not reported in Table 11. Antibodies with a maximum percent blockade below −25% were characterized as non-blockers/enhancers.
  • TABLE 11
    ELISA blocking of hPD-L1-mFc binding to hPD-1-hFc and hB7-1-hFc-6His by anti-
    PD-L1 antibodies
    Blocking 8
    nM of
    Blocking 0.5 Highest hPD-L1-
    Highest Blocking 0.5 nM of Antibody Blocking 8 nM mFc
    Antibody nM of hPD-L1-mFc concentration of binding to
    concentration hPD-L1-mFc binding to (nM) tested hPD-L1-mFc hB7-1-hFc-
    (nM) tested binding to hPD-1-hFc, on hB7-1- binding to 6His, %
    on hPD-1- hPD-1-hFc, % maximum hFc-6His hB7-1-hFc- maximum
    Antibody hFc coat IC50 (M) blocking coat 6His, IC50 (M) blocking
    H2aM8306N 50 <2.5E−10 (*) 99 150 <4.0E−09 (*) 93
    H2aM8307N 50 IC 29 150 NBI/enhancer −44
    H2aM8309N 50 <2.5E−10 (*) 98 150 <4.0E−09 (*) 98
    H2aM8310N 50 3.9E−10  76 150 <4.0E−09 (*) 98
    H2aM8312N 50 NBI 23 150 <4.0E−09(*) 98
    H2aM8314N 50 <2.5E−10 (*) 93 150 <4.0E−09 (*) 97
    H2aM8316N 50 <2.5E−10 (*) 96 150 <4.0E−09 (*) 98
    H2aM8317N 50 NBI −11 150 NBI/enhancer −166
    H2aM8321N 50 NBI 10 150 NBI/enhancer −124
    H2aM8323N 50 <2.5E−10 (*) 100 150 <4.0E−09 (*) 99
    H2aM8718N 50 <2.5E−10 (*) 97 150 1.0E−08  57
    H2aM8719N 50 7.7E−10  95 150 NBI/enhancer −50
    H1H9323P 50 <2.5E−10 (*) 43 210 NBI/enhancer −25
    H1H9327P 50 NBI/enhancer −28 210 IC 41
    H1H9329P 50 <2.5E−10 (*) 100 210 4.9E−09  100
    H1H9336P 50 <2.5E−10 (*) 100 210 4.4E−09  99
    H1H9344P2 50 NBI 15 210 NBI/enhancer −51
    H1H9345P2 50 <2.5E−10 (*) 26 210 NBI/enhancer −34
    H1H9351P2 50 <2.5E−10 (*) 100 210 4.4E−09  100
    H1H9354P2 50 5.3E−10  34 210 NBI −13
    H1H9364P2 50 <2.5E−10 (*) 100 210 4.1E−09  101
    H1H9373P2 50 <2.5E−10 (*) 100 210 4.6E−09  101
    H1H9382P2 50 <2.5E−10 (*) 39 210 NBI/enhancer −30
    H1H9387P2 50 <2.5E−10 (*) 100 210 <4.0E−09 (*) 100
    H1H9396P2 50 <2.5E−10 (*) 59 210 NBI 8
    Isotype 50 NBI −11 210 NBI −3
    control-
    human IgG1
    Isotype 50 NBI 5 210 NBI −3
    control-
    mouse
    IgG2a
    (*) below theoretical bottom of the assay; Assay theoretical bottom is 2.5E−10 M for hPD-1-hFc coat and
    4.0E−09 M for hB7-1-hFc-6His coat;
    NBI—non-blocker;
    IC—inconclusive
  • As shown in Table 11, 19 of the 25 anti-PD-L1 antibodies of the invention blocked 0.5 nM of hPD-L1-mFc from binding to hPD-1-hFc with IC50 values ranging from less than 250 pM to 770 pM with maximum percent blockade ranging from 26% to 100%. Four of the 25 anti-PD-L1 antibodies tested were characterized as non-blockers of hPD-L1-mFc binding to hPD-1-hFc, while one antibody tested (H1H9327P) was characterized as a non-blocker/enhancer of hPD-L1-mFc binding to hPD-1-hFc. One antibody (H2aM8307N) demonstrated weak blocking of hPD-L1-mFc binding to hPD-1-hFc with a maximum percent blockade of 29%; however the IC50 value could not be determined for this sample.
  • Further, 14 of the 25 anti-PD-L1 antibodies of the invention blocked 8 nM of hPD-L1-mFc from binding to hB7-1-hFc-6His with IC50 values ranging from <4 nM to 10 nM with maximum percent blockade ranging from 57% to 101%. Two of the 25 anti-PD-L1 antibodies tested were characterized as non-blockers of hPD-L1-mFc binding to hB7-1-hFc-6His, while 8 antibodies tested were characterized as non-blockers/enhancers of hPD-L1-mFc binding to hB7-1-hFc-6His. One antibody (H1H9327P) demonstrated weak blocking of hPD-L1-mFc binding to hB7-1-hFc-6His with a maximum percent blockade of 41%; however the IC50 value could not be determined for this sample.
  • Example 5: Blocking of PD-L1 Binding to PD-1 as Determined by Biosensor Assay and by Surface Plasmon Resonance
  • Inhibition of human PD-L1 from binding to human PD-1 by different anti-PD-L1 monoclonal antibodies was studied either using real time bio-layer interferometry assay on an Octet Red96 biosensor instrument (Fortebio Inc.) or using a real-time surface plasmon resonance biosensor assay on a Biacore 3000 instrument.
  • Inhibition studies for anti-PD-L1 monoclonal antibodies expressed with mouse Fc were performed on an Octet Red96 instrument. First, 100 nM of a recombinant human PD-L1 expressed with a C-terminal mouse IgG2a Fc tag (hPD-L1-mFc; SEQ ID: 348) was incubated with 500 nM of each anti-PD-L1 monoclonal antibody for at least 1 hour before running the inhibition assay. Approximately 0.8 nm to 1.2 nm of recombinant human PD-1 expressed with a C-terminal human IgG1 Fc tag (hPD-1-hFc; SEQ ID: 350) was captured using anti-human IgG Fc capture biosensors. The Octet biosensors captured with hPD-1-hFc were subsequently submerged into wells containing the mixture of hPD-L1-mFc and different anti-PD-L1 monoclonal antibodies. The entire experiment was performed at 25° C. in Octet HBST buffer (0.01 M HEPES pH7.4, 0.15M NaCl, 3 mM EDTA, 0.05% v/v Surfactant P20, 0.1 mg/mL BSA) with a plate shaking at a speed of 1000 rpm. The biosensors were washed in Octet HBST buffer in between each step of the experiment. The real-time binding response was monitored during the course of the experiment and the binding response at the end of every step was recorded. Binding of hPD-L1-mFc to the captured hPD-1-hFc was compared in the presence and absence of different anti-PD-L1 monoclonal antibodies and was used to determine the blocking behavior of the tested antibodies as shown in Table 12.
  • TABLE 12
    Inhibition of human PD-1 binding to human PD-L1 by different anti-PD-L1 monoclonal
    antibodies expressed with mouse Fc performed on Octet Red96 instrument.
    Binding of the mixture of
    Anti-PD-L1 Amount of 100 nM hPD-L1-mFc and
    monoclonal hPD-1-hFc 500 nM anti-PD-L1 %
    antibody Captured (nm) monoclonal antibody (nm) Blocking
    No antibody 1.05 0.31 0
    H2aM8306N 0.98 0.02 94
    H2aM8307N 1.01 0.46 −48
    H2aM8309N 0.89 0.02 94
    H2aM8310N 0.95 0.13 58
    H2aM8312N 1.06 0.52 −68
    H2aM8314N 0.99 0.02 94
    H2aM8316N 1.06 0.01 97
    H2aM8317N 0.92 0.58 −87
    H2aM8321N 1.04 0.60 −94
    H2aM8323N 1.00 0.02 94
    H2aM8718N 1.08 0.01 97
    H2aM8719N 0.93 0.11 65
    Isotype control antibody 1.10 0.35 −13
  • As shown in Table 12, 8 of the 12 anti-PD-L1 antibodies tested on the Octet Red96 instrument demonstrated blocking of hPD-L1-mFc from binding to hPD-1-hFc ranging from 58% to 97%. Four anti-PD-L1 antibodies tested demonstrated the ability to enhance the binding of hPD-L1-mFc to hPD-1-hFc.
  • Next, inhibition studies for anti-PD-L1 monoclonal antibodies expressed with human Fc were performed on Biacore 3000 instrument. First, 100 nM of recombinant human PD-L1 expressed with a C-terminal human IgG1 Fc tag (hPD-L1-hFc; SEQ ID: 350) was incubated with 500 nM of each anti-PD-L1 monoclonal antibody for at least 1 hour before running the inhibition assay. A CM5 Biacore sensor surface was first derivatized with anti-mouse IgG2a specific polyclonal antibody (Southern Biotech, #1080-01) using the standard EDC-NHS chemistry. Around 230 RUs of recombinant human PD-1 expressed with a C-terminal mouse IgG2a Fc tag (hPD-1-mFc; SEQ ID: 348) was then captured and was followed by an injection of 100 nM of hPD-L1-hFc in the presence and absence of different anti-PD-L1 monoclonal antibodies at a flow rate of 25 μL/min for 2 minutes. The entire experiment was performed at 25° C. in HBST running buffer (0.01 M HEPES pH7.4, 0.15M NaCl, 3 mM EDTA, 0.05% v/v Surfactant P20). The real-time binding responses were monitored during the entire course of the experiment and the binding response at the end of every step was recorded. Binding of hPD-L1-hFc to the captured hPD-1-mFc was compared in the presence and absence of different anti-PD-L1 monoclonal antibodies and was used to determine the blocking behavior of the tested antibodies as shown in Table 13.
  • TABLE 13
    Inhibition of human PD-1 binding to human PD-L1 by different anti-PD-L1 monoclonal
    antibodies expressed with human Fc performed on Biacore 3000 instrument.
    Binding of the mixture of
    Anti-PD-L1 Amount of 100 nM hPD-L1-hFc and
    monoclonal hPD-1-mFc 500 nM anti-PD-L1 %
    antibody Captured (RU) monoclonal antibody (nm) Blocking
    No mAb 222 56 0
    H1H9323P 224 262 −368
    H1H9327P 226 172 −207
    H1H9329P 227 2 97
    H1H9336P 227 9 84
    H1H9345P2 229 292 −422
    H1H9351P2 227 6 90
    H1H9354P2 229 296 −428
    H1H9364P2 228 8 86
    H1H9373P2 227 6 89
    H1H9382P2 228 307 −448
    H1H9387P2 228 5 91
    H1H9396P2 228 164 −193
    Isotype control antibody 228 56 0
  • As shown in Table 13, 6 out of 12 anti-PD-L1 antibodies of the invention tested on the Biacore 3000 instrument demonstrated blocking of hPD-L1-hFc from binding to hPD-1-mFc ranging from 84% to 97%. Six anti-PD-L1 antibodies tested demonstrated the ability to enhance the binding of hPD-L1-hFc to hPD-1-mFc.
  • Example 6: Octet Cross-Competition Between Anti-PD-L1 Antibodies
  • Binding competition between anti-PD-L1 monoclonal antibodies was determined using a real time, label-free bio-layer interferometry assay on an Octet RED384 biosensor (Pall ForteBio Corp.). The entire experiment was performed at 25° C. in Octet HBST buffer (0.01 M HEPES pH7.4, 0.15M NaCl, 3 mM EDTA, 0.05% v/v Surfactant P20, 0.1 mg/mL BSA) with the plate shaking at the speed of 1000 rpm. To assess whether 2 antibodies were able to compete with one another for binding to their respective epitopes on the recombinant human PD-L1 expressed with a C-terminal myc-myc-hexahistidine tag (hPD-L1-MMH; SEQ ID: 345), around ˜0.3 nm of hPD-L1-MMH was first captured onto anti-Penta-His antibody coated Octet biosensor tips (Fortebio Inc, #18-5079) by submerging the tips for 5 minutes into well containing 20 μg/mL solution of hPD-L1-MMH. The antigen captured biosensor tips were then saturated with first anti-PD-L1 monoclonal antibody (subsequently referred to as mAb-1) by dipping into wells containing 50 μg/mL solution of mAb-1 for 5 minutes. The biosensor tips were then subsequently dipped into wells containing 50 μg/mL solution of a second anti-PD-L1 monoclonal antibody (subsequently referred to as mAb-2). The biosensor tips were washed in Octet HBST buffer in between every step of the experiment. The real-time binding response was monitored during the course of the experiment and the binding response at the end of every step was recorded as shown in FIG. 1. The response of mAb-2 binding to hPD-L1-MMH pre-complexed with mAb-1 was compared and competitive/non-competitive behavior of different anti-PD-L1 monoclonal antibodies was determined.
  • Under the experimental conditions used in this Example, (a) H2aM8309N, H1H9329P, H1H9336P, H2aM8314N, H2aM8316N, H2aM8718N, H1H9387P2, H1H9351P2, H1H9364P2, H1H9373P2, and H2aM8306N cross-competed with each other; (b) H2aM8310N, H2aM8321N and H2aM8312N cross-competed with each other; (c) H1H9396P2, H2aM8317N, H2aM8321N, H1H9323P, H1H9382P2, H1H9344P2, H1H9345P2, and H1H9354P2 cross-competed with each other; and (d) H1H9327P and H2aM8307N cross-competed with each other. In one instance, competition was observed in one orientation but not in the opposite orientation: i.e., H2aM8307N when applied first competed with H2aM8309N, H1H9329P, H1H9336P, H2aM8314N, H2aM8316N, H2aM8718N, H1H9387P2, H1H9351P2, H1H9364P2, H1H9373P2, and H2aM8306N; however, in the opposite orientation, H2aM8309N, H1H9329P, H1H9336P, H2aM8314N, H2aM8316N, H2aM8718N, H1H9387P2, H1H9351P2, H1H9364P2, H1H9373P2, and H2aM8306N when applied first did not compete with H2aM8307N.
  • Example 7: Antibody Binding to Cells Overexpressing PD-L1
  • The binding of anti-PD-L1 antibodies to a human embryonic kidney cell line (HEK293; ATCC, #CRL-1573) stably transfected with full length human PD-L1 (amino acids 1 to 290 of accession number NP_054862.1) (HEK293/hPD-L1) was determined by FACS.
  • For the assay, adherent cells were detached using enzyme-free dissociation buffer and blocked with complete medium. Cells were centrifuged and resuspended at a concentration of 2.8×10{circumflex over ( )}6 cells/mL in cold PBS containing 2% FBS. HEK293 parental and HEK293/hPD-L1 cells were then incubated for 15 to 30 minutes on ice with 100 nM of each anti-PD-L1 antibody or an isotype control antibody. Unbound antibodies were removed by washing with D-PBS containing 2% FBS, and cells were subsequently incubated with a phycoerythrin-conjugated secondary Fcγ fragment specifically recognizing either human Fc (Jackson ImmunoResearch, #109-116-170) or mouse Fc (Jackson ImmunoResearch, #115-115-164) for 15 to 30 minutes on ice. Cells were washed with D-PBS containing 2% FBS to remove unbound secondary detection reagents and fluorescence measurements were acquired using a HyperCyte (IntelliCyt, Inc.) flow cytometer. Data was analyzed using HyperCyte software.
  • TABLE 14
    FACS binding of anti-PD-L1 antibodies to
    HEK293/hPD-L1 cells and parental HEK293 cells
    FACS on FACS on Ratio of
    HEK293 HEK293/ HEK293/hPD-L1
    parental cells hPD-L1 cells to HEK293
    Antibody [MFI] [MFI] parental cells
    H1H9323P 1909 154992 81
    H1H9327P 2120 317592 150
    H1H9329P 1504 282088 188
    H1H9336P 2263 379009 168
    H1H9344P2 1691 200976 119
    H1H9345P2 1885 228406 121
    H1H9351P2 1685 289523 172
    H1H9354P2 2204 275839 125
    H1H9364P2 2066 323663 157
    H1H9373P2 2151 333236 155
    H1H9382P2 1473 205563 140
    H1H9387P2 1232 323793 263
    H1H9396P2 2340 227961 97
    H2aM8306N 1286 316485 246
    H2aM8307N 1382 73976 54
    H2aM8309N 1160 192678 166
    H2aM8310N 1357 14918 11
    H2aM8312N 1380 158331 115
    H2aM8314N 2053 194832 95
    H2aM8316N 1601 172104 108
    H2aM8317N 1270 67600 53
    H2aM8321N 1322 112495 85
    H2aM8323N 2250 163497 73
    H2aM8718N 2225 194341 87
    H2aM8719N 1272 133399 105
    mouse IgG 1273 1115 0.9
    Isotype control
    human IgG1 1179 1099 0.9
    Isotype control
    human IgG4 1991 1839 0.9
    Isotype control
  • As shown in Table 14, all 25 anti-PD-L1 antibodies of the invention showed strong binding to the HEK293/hPD-L1 cells compared to binding on the parental HEK293 line.
  • Example 8: Blocking of PD-L1-Induced T-Cell Down-Regulation in a T-Cell/APC Luciferase Reporter Assay
  • T-cell activation is achieved by stimulating T-cell receptors (TcR) that recognize specific peptides presented by major histocompatibility complex class I or II proteins on antigen-presenting cells (APC). Activated TcRs in turn initiate a cascade of signaling events that can be monitored by reporter genes driven by transcription factors such as activator-protein 1 (AP-1), Nuclear Factor of Activated T-cells (NFAT) or Nuclear factor kappa-light-chain-enhancer of activated B cells (NFκb). T-cell response is modulated via engagement of co-receptors expressed either constitutively or inducibly on T-cells. One such receptor is programmed cell death protein 1 (PD-1), a negative regulator of T-cell activity. PD-1 interacts with its ligand, PD-L1, which is expressed on target cells including APCs or cancer cells, and this interaction results in the delivery of inhibitory signals by recruiting phosphatases to the TcR signalosome, resulting in the suppression of positive signaling.
  • A bioassay was developed to measure T cell signaling induced by interaction between APC and T cells by utilizing a mixed culture derived from two mammalian cell lines: Jurkat cells (an immortalized T cell line) and Raji cells (a B cell line) (FIG. 1). For the first component of the bioassay, Jurkat Clone E6-1 cells (ATCC, #TIB-152) were transduced with the Cignal Lenti AP-1 Luc Reporter (Qiagen—Sabiosciences, #CLS-011L) as per the manufacturer's instructions. The lentivirus encodes the firefly luciferase gene under the control of a minimal CMV promoter, tandem repeats of the TPA-inducible transcriptional response element (TRE) and a puromycin resistance gene. The engineered Jurkat cell line was subsequently transduced with a PD-1 chimera comprising the extracellular domain of human PD-1 (amino acids from 1 to 170 of human PD1; accession number NP_005009.2) and the trans-membrane and cytoplasmic domains of human CD300a (amino acids from 181 to 299 of human CD300a; accession number NP_009192.2). The resulting stable cell line (Jurkat/AP1-Luc/hPD1-hCD300a) was selected and maintained in RPMI/10% FBS/penicillin/streptomycin/glutamine supplemented with 500 ug/mL G418+1 ug/mL puromycin.
  • For the second component of the bioassay, Raji cells (ATCC, #CCL-86) were transduced with human PD-L1 gene (amino acids 1-290 of accession number NP_054862.1) that had been cloned into a lentiviral (pLEX) vector system (Thermo Scientific Biosystems, #OHS4735). Raji cells, positive for PD-L1 (Raji/hPD-L1) were isolated by FACS using a PD-L1 antibody and maintained in Iscove/10% FBS/penicillin/streptomycin/glutamine supplemented with 1 ug/mL puromycin.
  • To simulate the APC/T cell interaction, a bispecific antibody composed of one Fab arm that bindings to CD3 on T cells and the other one Fab arm binding that binds to CD20 on Raji cells (CD3×CD20 bispecific antibody; e.g., as disclosed in US20140088295) was utilized. The presence of the bispecific molecule in the assay results in the activation of the T cell and APC by bridging the CD3 subunits on T-cells to CD20 endogenously expressed on Raji cells. Ligation of CD3 with anti-CD3 antibodies has been demonstrated to lead to activation of T cells. In this bioassay, antibodies blocking the PD1/PD-L1 interaction rescue T-cell activity by disabling the inhibitory signaling and subsequently leading to increased AP1-Luc activation.
  • In the luciferase-based bioassay, RPMI1640 supplemented with 10% FBS and penicillin/streptomycin/glutamine was used as assay medium to prepare cell suspensions and antibody dilutions to carry out the screening of anti-PD-L1 monoclonal antibodies (mAbs). On the day of the screening, EC50 values of anti-PD-L1 mAbs, in the presence of a fixed concentration of CD3×CD20 bispecific antibody (30 pM), as well as the EC50 of the bispecific antibody alone, were determined. In the following order, cells and reagents were added to 96 well white, flat-bottom plates. For the anti-PD-L1 mAb EC50 determinations, first a fixed concentration of CD3×CD20 bispecific antibody (final 30 pM) was prepared and added to the microtiter plate wells. Twelve-point serial dilutions of anti-PD-L1 mAbs and controls were then added (final concentrations ranging from 1.7 pM to 100 nM; plus wells with assay medium alone). For the bispecific antibody (alone) EC50 determination, the bispecific antibody, at final concentrations ranging from 0.17 pM to 10 nM (plus wells with assay medium alone), was added to the microtiter plate wells. Subsequently, a 2.5×10{circumflex over ( )}6/mL Raji/hPD-L1 cell suspension was prepared and 20 uL per well was added (final cell number/well 5×10{circumflex over ( )}4 cells). Plates were left at room temperature (15-20 minutes), while a suspension of 2.5×10{circumflex over ( )}6/mL of Jurkat/AP1-Luc/hPD1(ecto)-hCD300a(TM-Cyto) was prepared. 20 uL of the Jurkat suspension (final cell number/well 5×10{circumflex over ( )}4 cells) was added per well. Plates containing the co-culture were incubated for 5 to 6 hours at 37° C. in 5% CO2. Luciferase activity was then detected after the addition of ONE-Glo™ (Promega, #E6051) reagent and relative light units (RLUs) were measured on a Victor luminometer. All samples were tested in duplicates.
  • RLU values for each screened antibody were normalized by setting the assay condition with fixed (30 pM) concentration of the CD3/CD20 bispecific antibody, but without anti-PD-L1 antibody to 100%. This condition corresponds to the maximal AP1-Luc response elicited by the bispecific molecule in the presence of the PD-1/PD-L1 inhibitory signal. Upon addition of the anti-PD-L1 antibody, the inhibitory signal is suppressed, and the increased stimulation is shown here as Emax, the percentage increase in the signal in the presence of the highest antibody dose tested (100 nM). To compare potency of the anti-PD-L1 antibodies tested, the concentration of antibody at which the normalized RLU value reached 125% activation was determined from a four-parameter logistic equation over a 12-point response curve using GraphPad Prism. The results are summarized in Table 15.
  • TABLE 15
    Anti-PD-L1 antibody blocking PD-1/PD-L1
    dependent inhibition of AP1-Luc signaling
    Antagonistic assay Antagonistic assay
    Concentration [M] of Emax mean
    Antibody at 125% [%] @
    Antibody activation 100 nM
    H2aM8306N 3.78E−09 166.8
    H2aM8307N N/A 87.7
    H2aM8309N 3.09E−10 180.9
    H2aM8310N N/A 112.8
    H2aM8312N N/A 75.6
    H2aM8314N 1.44E−11 234.9
    H2aM8316N 1.47E−10 177.4
    H2aM8317N N/A 109.2
    H2aM8321N N/A 116.8
    H2aM8323N 2.20E−10 173.3
    H2aM8718N 1.51E−10 182.0
    H1H9323P N/A 101.1
    H1H9327P N/A 77.1
    H1H9329P N/A 124.5
    H1H9336P 9.81E−11 162.6
    H1H9344P2 N/A 98.3
    H1H9345P2 N/A 89.2
    H1H9351P2 3.44E−10 154.4
    H1H9354P2 N/A 89.9
    H1H9364P2 7.93E−11 164.5
    H1H9373P2 1.34E−10 150.2
    H1H9382P2 N/A 86.5
    H1H9387P2 1.86E−11 141.2
    H1H9396P2 N/A 102.6
    H1H8314N 6.57E−11 147.9
    H1H9364P2 1.62E−10 158.1
    H1H9373P2 7.07E−11 152.5
    mlgG2a isotype control N/A 80.2
    hlgG1 isotype control N/A 96.8
    hlgG4 isotype control N/A 87.6
    N/A = not applicable because at the concentrations tested these antibodies did not activate 125%
  • As shown in Table 15, 14 out of the 27 anti-PD-L1 antibodies of the invention tested blocked PD-1/PD-L1 inhibition with Emax values ranging from 234.9 to 138.1. Thirteen out of the 27 anti-PD-L1 antibodies of the invention did not demonstrate substantial blockade of PD1/PD-L1 interaction when tested in this assay. Isotype controls did not interfere with the PD1/PD-L1 interaction.
  • Example 9: In Vivo Efficacy of Anti-PD-L1 Antibodies
  • To determine the effect of a select number of anti-PD-L1 antibodies of the invention in a relevant in vivo model, an MC38.ova tumor growth study, involving subcutaneous injection of tumor cells and started on different days, was conducted in mice that were homozygous for the expression of the extracellular domain of human PD-L1 in place of extracellular domain of mouse PD-L1 (PD-L1 HumIn mice) on a 75% C57/BI6/25% 129 strain background. MC38.Ova (mouse colon adenocarcinoma) cells were engineered to express chicken ovalbumin in order to increase tumor immunogenicity, and to allow monitoring of the T-cell immune responses to well-defined antigenic ovalbumin peptides. In a second step, MC38.Ova cells were transduced with a lentiviral vector expressing hPD-L1 under SFFV promoter. MC38.Ova cells positive for hPD-L1 (MC38.Ova/hPD-L1) were isolated by FACS using hPD-L1-specific antibody. The cells were found to express low level of endogenous mouse PD-L1.
  • For a first study (study #1), mice were divided evenly according to body weight into 5 treatment or control groups (n=5 to 8 mice per group). At day 0, mice were anesthetized by isoflurane inhalation and then injected subcutaneously into the right flank with 1×106 MC38.ova/hPD-L1 cells in suspension of 100 uL of DMEM. Treatment groups were intraperitoneally injected with 500 ug of either one of three anti-PD-L1 antibodies of the invention, or one of two isotype control antibodies with irrelevant specificity on days 3, 7, 10, 14, and 17 of the experiment, while one group of mice was left untreated.
  • In a second study (study #2), PD-L1 humanized mice were randomized into 7 treatment groups (n=5 to 6 mice). On day 0, mice were subcutaneously implanted with 1×10{circumflex over ( )}6 MC38.Ova/hPD-L1 cells. Mice were intraperitoneally administered with REGN a-PD-L1 ab (H1H8314N or H1H9364P2 or H1H9373P2), or isotype control abs hIgG4 mut or hIgG1 at doses of 10 mg/kg or 5 mg/kg. Groups of mice were administered antibody on days 3, 7, 10, 14, and 17. Tumor volumes were monitored by caliper measurement twice per week for the duration of the experiment (21 days). Experimental dosing and treatment protocol for groups of mice are shown in Table 16.
  • TABLE 16
    Experimental dosing and treatment protocol for groups of mice
    Samples Dosage amount at each
    Study # tested dosage time point Dosing interval
    1 Isotype control 1 500 μg Days 3, 7, 10, 14, 17
    H1H8314N 500 μg Days 3, 7, 10, 14, 17
    H1H9364P2 500 μg Days 3, 7, 10, 14, 17
    H1H9373P2 500 μg Days 3, 7, 10, 14, 17
    Isotype control 2 500 μg Days 3, 7, 10, 14, 17
    2 Isotype control 1 10 mg/kg Days 3,7, 10, 14, 17
    H1H8314N 10 mg/kg Days 3, 7, 10, 14, 17
    H1H8314N 5 mg/kg Days 3, 7, 10, 14, 17
    H1H9364P2 10 mg/kg Days 3, 7, 10,
    14, 17
    H1H9364P2 5 mg/kg Days 3, 7, 10,
    14, 17
    H1H9373P2 10 mg/kg Days 3, 7, 10,
    14, 17
    H1H9373P2 5 mg/kg Days 3, 7, 10,
    14, 17
  • For the studies, average tumor volumes were monitored by caliper measurement twice per week for the duration of the experiment (17 days) and percent survival was recorded at the end of the experiment. In addition, the number of tumor-free mice was also assessed at the end of the study. Results, expressed as mean tumor volume (mm3)(±SD), percent survival, and number of tumor-free mice are shown in Tables 17 and 18.
  • TABLE 17
    Mean tumor volume, percent survival and numbers of
    tumor-free mice in each treatment group from study #1
    Tumor Volume, mm3 Tumor-Free
    mean (±SD) Survival, % Mice
    Antibody Days
    10 Day 17 Day 10 Day 17 Day 17
    Isotype Control 1 65 (±27) 148 (±109) 100% 100% 0/5 (0%)
    Isotype Control 2 54 (±44) 80 (±63) 100% 100% 0/5 (0%)
    H1H8314N  6 (±10) 2 (±5) 100% 100%  4/5 (80%)
    H1H9364P2 16 (±17) 0 (±0) 100% 100%   5/5 (100%)
    H1H9373P2 13 (±14) 0 (±0) 100% 100%   5/5 (100%)
  • As shown in Table 17 for study #1, all three anti-PD-L1 antibodies of the invention were efficacious in promoting tumor regression at the dosage of 500 ug/mouse with all mice from treatment groups that received two of the antibodies, H1H9364P2 and H1H9373P2, being tumor free at day 17. In the treatment group that received one of the anti-PD-L1 antibodies of the invention, H1H8314N, 4 out of 5 mice were tumor free by day 17, whereas 0 out of 5 animals were tumor-free in the isotype control groups. One-way ANOVA with Dunnett's multiple comparison post-test revealed a significant difference in tumor volumes between treatments with anti-PD-L1 antibodies of the invention and the isotype control antibody with a p value <0.05.
  • TABLE 18
    Mean tumor volume, percent survival and numbers of tumor-free mice in each
    treatment group from study #2
    Tumor Volume, mm3 Tumor- Tumor-
    Mean (SD) Survival, % free mice free mice
    Day
    10 Day 10 Day 21 Day 21 Day 10 Day 10 Day 21 Day 21 Day 21 Day 21
    Antibody 10 MPK 5 MPK 10 MPK 5 MPK 10 MPK 5 MPK 10 MPK 5 MPK 10 MPK 5 MPK
    Isotype 55 (37) N/A 534 N/A 100% N/A 100% N/A 0/6 (0%) N/A
    control 1 (356)
    H1H8314N 14 (15) 17 (4)  19 108 100% 100% 100% 100% 3/6 2/5 (40%)
    922) (101) (50%)
    H1H9364P2 18 (10) 23 (10) 34 (81) 231 100% 100% 100% 100% 5/6 1/5 (20%)
    (238) (83%)
    H1H9373P2 10 (8)  25 929) 7 (16) 37 (59) 100% 100% 100% 100% 5/6 3/5 (60%)
    (83%)
  • As shown in Table 18, in study #2, administration of the selected anti-PD-L1 antibodies resulted in inhibition of tumor growth promoting tumor regression. All three anti-PD-L1 antibodies were efficacious at the 10 mg/kg dose and 5 mg/kg dose and promoted tumor regression in treated mice in a dose dependent manner throughout the course of the experiment, whereas 0 out of 5 animals were tumor-free in the control group. One-way ANOVA with Tukey's multiple comparison post-test revealed a significant difference in tumor volumes between treatments with the anti-PD-L1 antibodies and isotype control antibody with p value <0.05 or lower.
  • Example 10: Anti-Tumor Effects of a Combination of an Anti-PD-L1 Antibody and a VEGF Antagonist in a Mouse Early-Treatment Tumor Model
  • An early-treatment tumor model was developed to test the efficacy of a combination of an anti-PD-L1 antibody and a VEGF antagonist. In this model, the combination therapy is administered shortly after tumor implantation. The experiment also used an anti-PD-1 antibody alone and in combination with the VEGF antagonist. The anti-PD-L1 antibody used in this experiment was an anti-PD-L1 monoclonal antibody with VH/VL sequences of antibody “YW243.55S70” according to US20100203056A1 (Genentech, Inc.), with mouse IgG2a and which was cross-reactive with mouse PD-L1. The VEGF antagonist used in this experiment was aflibercept (a VEGF receptor-based chimeric molecule, also known as “VEGF-trap” or “VEGFR1R2-FcΔC1(a),” a full description of which is provided elsewhere herein). The anti-PD-1 antibody used in this experiment was anti-mouse PD-1 clone “RPMI-14” with rat IgG2b (Bio X Cell, West Lebanon, N.H.).
  • For this experimental model, 1.0×106 Colon-26 tumor cells were implanted sub-cutaneously into BALB/c mice at Day 0. Starting on Day 3, prior to the establishment of measurable tumors, mice were treated with one of the mono- or combination therapies, or control combination, as set forth in Table 19.
  • TABLE 19
    Experimental Dosing and Treatment Groups
    Treatment Group First Agent Second Agent
    Control IgG2a isotype hFc control
    Combination control (250 μg, IP) (250 μg, SC)
    VEGF Trap IgG2a isotype Aflibercept
    only control (250 μg, IP) (10 mg/kg, SC)
    anti-PD-1 anti-PD-1 mAb hFc control
    only RPMI-14 (250 μg, IP) (250 μg, SC)
    anti-PD-L1 anti-PD-L1 mAb hFc control
    only (250 μg, IP) (250 μg, SC)
    VEGF Trap + anti-PD-1 mAb Aflibercept
    anti-PD-1 RPMI-14 (250 μg, IP) (10 mg/kg, SC)
    VEGF Trap + anti-PD-L1 mAb Aflibercept
    anti-PD-L1 (250 μg, IP) (10 mg/kg, SC)
  • The various therapies were administered at five different time points over a two week period (i.e., injections at Day 3, Day 6, Day 10, Day 13 and Day 19).
  • Animals in each therapy group were evaluated in terms of tumor incidence, tumor volume, median survival time, and number of tumor-free animals at Day 50. The extent of tumor growth is summarized in FIG. 2 (tumor growth curves) and FIG. 3 (tumor volume at Day 28). Results are also summarized in Table 20.
  • TABLE 20
    Tumor-free mice upon treatment
    No. of Tumor-Free
    Treatment Group Animals by Day 50
    Control Combination 0/10
    VEGF Trap only 3/10
    anti-PD-1 only 4/10
    anti-PD-L1 only 5/10
    VEGF Trap + anti-PD-1 7/10
    VEGF Trap + anti-PD-L1 9/10
  • Tumor growth was substantially reduced in animals treated with the combination of VEGF Trap+ anti-PD-L1 antibody as compared with treatment regimens involving either therapeutic agent alone (see FIGS. 2 and 3). Furthermore, survival was substantially increased in the VEGF Trap+ anti-PD-L1 antibody group, with 90% of animals surviving to at least day 50 after tumor implantation. By contrast, for the anti-PD-L1 and VEGF Trap monotherapy groups, survival to Day 50 was only 50% and 30% respectively (see FIG. 3 and Table 20).
  • The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and the accompanying figures. Such modifications are intended to fall within the scope of the appended claims.

Claims (42)

What is claimed is:
1. An isolated antibody or antigen-binding fragment thereof that competes for binding to human programmed death-ligand 1 (PD-L1) protein with a reference antibody comprising the complementarity determining regions (CDRs) of a heavy chain variable region (HCVR), wherein the HCVR has an amino acid sequence selected from the group consisting of HCVR sequences listed in Table 1; and the CDRs of a light chain variable region (LCVR), wherein the LCVR has an amino acid sequence selected from the group consisting of LCVR sequences listed in Table 1 and wherein the antibody has one or more of the following properties:
(a) binds monomeric PD-L1 with a binding dissociation equilibrium constant (KD) of less than about 310 pM as measured in a surface plasmon resonance assay at 37° C.;
(b) binds monomeric human PD-L1 with a KD less than about 180 pM in a surface plasmon resonance assay at 25° C.;
(c) binds dimeric human PD-L1 with a KD of less than about 15 pM as measured in a surface plasmon resonance assay at 37° C.; and
(d) binds dimeric human PD-L1 with a KD less than about 8 pM in a surface plasmon resonance assay at 25° C.
2. The isolated antibody or antigen-binding fragment of claim 1, wherein the reference antibody or antigen-binding fragment thereof comprises an HCVR/LCVR amino acid sequence pair as set forth in Table 1.
3. The isolated antibody or antigen-binding fragment of claim 2, wherein the reference antibody comprises an HCVR/LCVR amino acid sequence pair selected from the group consisting of SEQ ID NOs: 82/90, 98/106, 146/154, 162/170, 290/274, 306/274, 314/274 and 330/274.
4. An isolated human monoclonal antibody or antigen-binding fragment thereof that binds specifically to human PD-L1, wherein the antibody or antigen-binding fragment thereof comprises a HCVR having an amino acid sequence selected from the group consisting of HCVR sequences listed in Table 1.
5. An isolated human monoclonal antibody or antigen-binding fragment thereof that binds specifically to human PD-L1, wherein the antibody or antigen-binding fragment thereof comprises a LCVR having an amino acid sequence selected from the group consisting of LCVR sequences listed in Table 1.
6. An isolated human monoclonal antibody or antigen-binding fragment thereof that binds specifically to human PD-L1, wherein the antibody or antigen-binding fragment thereof comprises (a) a HCVR having an amino acid sequence selected from the group consisting of HCVR sequences listed in Table 1; and (b) a LCVR having an amino acid sequence selected from the group consisting of LCVR sequences listed in Table 1.
7. The isolated antibody or antigen-binding fragment thereof of claim 6, wherein the antibody or antigen-binding fragment thereof comprises three heavy chain CDRs (HCDR1, HCDR2 and HCDR3) contained within any one of the HCVR sequences listed in Table 1; and three light chain CDRs (LCDR1, LCDR2 and LCDR3) contained within any one of the LCVR sequences listed in Table 1.
8. The isolated antibody or antigen-binding fragment thereof of claim 7, comprising:
(a) a HCDR1 domain having an amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 20, 36, 52, 68, 84, 100, 116, 132, 148, 164, 180, 188, 204, 220, 236, 252, 268, 284, 292, 300, 308, 316, 324, 332, and 340;
(b) a HCDR2 domain having an amino acid sequence selected from the group consisting of SEQ ID NOs: 6, 22, 38, 54, 70, 86, 102, 118, 134, 150, 166, 182, 190, 206, 222, 238, 254, 270, 286, 294, 302, 310, 318, 326, 334, and 342;
(c) a HCDR3 domain having an amino acid sequence selected from the group consisting of SEQ ID NOs: 8, 24, 40, 56, 72, 88, 104, 120, 136, 152, 168, 184, 192, 208, 224, 240, 256, 272, 288, 296, 304, 312, 320, 328, 336, and 344;
(d) a LCDR1 domain having an amino acid sequence selected from the group consisting of SEQ ID NOs: 12, 28, 44, 60, 76, 92, 108, 124, 140, 156, 172, 196, 212, 228, 244, 260, and 276;
(e) a LCDR2 domain having an amino acid sequence selected from the group consisting of SEQ ID NOs: 14, 30, 46, 62, 78, 94, 110, 126, 142, 158, 174, 198, 214, 230, 246, 262, and 278; and
(f) a LCDR3 domain having an amino acid sequence selected from the group consisting of SEQ ID NOs: 16, 32, 48, 64, 80, 96, 112, 128, 144, 160, 176, 200, 216, 232, 248, 264, and 280.
9. The isolated antibody or antigen-binding fragment of claim 8, comprising a HCVR/LCVR amino acid sequence pair selected from the group consisting of SEQ ID NOs: 82/90, 98/106, 146/154, 162/170, 290/274, 306/274, 314/274 and 330/274.
10. An isolated antibody or antigen-binding fragment thereof that blocks PD-L1 binding to one of PD-1 or B7-1, the isolated antibody or antigen-binding fragment thereof comprising the CDRs of a HCVR, wherein the HCVR has an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 34, 50, 82, 98, 146, 162, 178, 186, 234, 250, 266, 290, 306, 314, and 330; and the CDRs of a LCVR, wherein the LCVR has an amino acid sequence selected from the group consisting of SEQ ID NOs: 10, 42, 58, 90, 106, 154, 170, 194, 242, 258, and 274.
11. An isolated antibody or antigen-binding fragment thereof that binds PD-L1, wherein the antibody or antigen-binding fragment thereof enhances PD-L1 binding to one of PD-1 or B7-1.
12. The isolated antibody or antigen-binding fragment thereof of claim 11, wherein the antibody comprises the CDRs of a HCVR, wherein the HCVR has an amino acid sequence selected from the group consisting of SEQ ID NOs: 18, 66, 114, 130, 202, 218, 282, 298, 322, and 338; and the CDRs of a LCVR, wherein the LCVR has an amino acid sequence selected from the group consisting of SEQ ID NOs: 26, 74, 122, 138, 210, 226, and 274.
13. The isolated antibody or antigen-binding fragment thereof of claim 11 or 12, wherein the antibody comprises an HCVR/LCVR amino acid sequence pair selected from the group consisting of SEQ ID NOs: 18/26, 66/74, 114/122, 130/138, 202/210, 218/226, 282/274, 298/274, 322/274, and 338/274.
14. The antibody or antigen-binding fragment thereof of any one of claims 1-13, wherein the antibody is a multi-specific antigen-binding molecule.
15. A pharmaceutical composition comprising an isolated antibody or antigen-binding fragment thereof that binds to PD-L1 according to any one of claims 1-14 and a pharmaceutically acceptable carrier or diluent.
16. An isolated polynucleotide molecule comprising a polynucleotide sequence that encodes a HCVR of an antibody as set forth in any one of claims 1-7 or 9-14.
17. An isolated polynucleotide molecule comprising a polynucleotide sequence that encodes a LCVR of an antibody as set forth in any one of claims 1-6 or 8-14.
18. A vector comprising the polynucleotide sequence of claim 16 or 17.
19. A cell expressing the vector of claim 18.
20. A multi-specific antigen-binding molecule or fragment thereof comprising a first antigen-binding specificity that binds specifically to PD-L1 and a second antigen-binding specificity that binds specifically to an antigen selected from the group consisting of a tumor-cell-specific antigen, an antigen specific to a virally-infected cell, and a T-cell co-inhibitor.
21. The multi-specific antigen-binding molecule or fragment thereof of claim 20, wherein the first antigen-binding specificity comprises three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within any one of the heavy chain variable region (HCVR) sequences listed in Table 1; and three light chain CDRs (LCDR1, LCDR2 and LCDR3) contained within any one of the light chain variable region (LCVR) sequences listed in Table 1.
22. The multi-specific antigen-binding molecule or fragment thereof of claim 21, wherein the first antigen-binding specificity comprises a HCVR having an amino acid sequence selected from the group consisting of HCVR sequences listed in Table 1; and a LCVR having an amino acid sequence selected from the group consisting of LCVR sequences listed in Table 1.
23. The multi-specific antigen-binding molecule or fragment thereof of claim 22, wherein the first antigen-binding specificity comprises a HCVR/LCVR amino acid sequence pair selected from the group consisting of SEQ ID NOs: 82/90, 98/106, 146/154, 162/170, 290/274, 306/274, 314/274 and 330/274.
24. The multi-specific antigen-binding molecule or fragment thereof of claim 20, wherein the first antigen-binding specificity comprises an extracellular domain of one of PD-1 or B7-1, or fragment thereof.
25. The multi-specific antigen-binding molecule or fragment thereof of claim 20, wherein the second antigen-binding specificity binds specifically to a T-cell co-inhibitor selected from the group consisting of LAG3, TIM3, B7-1, CTLA-4, BTLA, CD28, 2B4, LY108, TIGIT, ICOS, and CD160.
26. The multi-specific antigen-binding molecule or fragment thereof of any one of claims 20-24, wherein the second antigen-binding specificity binds specifically to a tumor cell-specific antigen selected from the group consisting of CA9, CA125, melanoma-associated antigen (MAGE), carcinoembryonic antigen (CEA), vimentin, tumor-M2-PK, prostate-specific antigen (PSA), MART-1, and CA19-9.
27. The multi-specific antigen-binding molecule or fragment thereof of any one of claims 20-24, wherein the second antigen-binding specificity binds specifically to an antigen from a cell infected with a virus selected from the group consisting of human immunodeficiency virus (HIV), hepatitis C virus (HCV), human papilloma virus (HPV), lymphocytic choriomeningitis virus (LCMV), and simian immunodeficiency virus (SIV).
28. The multi-specific antigen-binding molecule or fragment thereof of any one of claims 20-26 for use in the treatment of a cancer selected from the group consisting of renal cell carcinoma, colorectal cancer, ovarian cancer, prostate cancer, breast cancer, colon cancer, non-small-cell lung cancer and melanoma.
29. The multi-specific antigen-binding molecule or fragment thereof any one of claim 20-25 or 27 for use in the treatment of a chronic viral infection caused by a virus selected from the group consisting of HIV, HPV, HBV, HCV, LCMV and SIV.
30. A multi-specific antigen-binding molecule or antigen-binding fragment thereof comprising a first binding specificity that binds specifically to PD-L1 and a second binding specificity that comprises an extracellular domain of PD-1 or B7-1, or fragment thereof.
31. The multi-specific antigen-binding molecule or fragment thereof of claim 30 for use in the treatment of a cancer selected from the group consisting of renal cell carcinoma, colorectal cancer, ovarian cancer, prostate cancer, breast cancer, colon cancer, non-small-cell lung cancer and melanoma.
32. The multi-specific antigen-binding molecule or fragment thereof of claim 30 for use in the treatment of a chronic viral infection caused by a virus selected from the group consisting of HIV, HPV, HBV, HCV, LCMV and SIV.
33. A method of enhancing an immune response in a subject, the method comprising administering a pharmaceutical composition comprising an isolated antibody or antigen-binding fragment thereof according to any one of claims 1-14; or a multi-specific antigen-binding molecule or fragment thereof according to any one of claim 20-27 or 30.
34. A method of inhibiting a T-regulatory (Treg) cell in a subject comprising administering a pharmaceutical composition comprising an isolated human antibody or antigen-binding fragment thereof according to any one of claims 1-14; or a multi-specific antigen-binding molecule or fragment thereof according to any one of claim 20-27 or 30.
35. A method of enhancing T-cell activation in a subject, the method comprising administering a pharmaceutical composition comprising an antibody or antigen-binding fragment thereof according to any one of claims 1-14; or a multi-specific antigen-binding molecule or fragment thereof according to any one of claim 20-27 or 30.
36. The method of any one of claims 33-35, wherein the subject has a disease or disorder selected from the group consisting of renal cell carcinoma, ovarian cancer, prostate cancer, non-small-cell lung cancer, colorectal cancer, and melanoma.
37. The method of any one of claims 33-35, wherein the subject has a chronic viral infection caused by a virus selected from the group consisting of HIV, HCV, HBV, HPV, LCMV and SIV.
38. A method of inhibiting growth of a tumor or a tumor cell, the method comprising contacting the tumor or tumor cell with a therapeutically effective amount of the antibody of any one of claim 1-14 or 20-26.
39. The method of any one of claims 33-38, wherein the antibody or antigen-binding fragment thereof, or the pharmaceutical composition comprising the antibody or antigen-binding fragment thereof, is administered to the subject in combination with a second therapeutic agent.
40. The method of claim 39, wherein the second therapeutic agent is selected from the group consisting of a NSAID, a corticosteroid, an antibody to a T-cell co-inhibitor, an antibody to a tumor specific antigen, an antibody to an autoimmune tissue antigen, an antibody to a virally-infected-cell antigen, an antibody to PD-1, a dietary supplement such an antioxidant, a VEGF antagonist, a chemotherapeutic agent, a cytotoxic agent, and an anti-viral drug.
41. The method of any one of claims 33-40, wherein the antibody or antigen-binding fragment thereof is administered subcutaneously, intravenously, intradermally, intraperitoneally, orally, intramuscularly or intracranially.
42. The method of any one of claims 33-41, wherein the antibody or antigen-binding fragment is administered at a dose of about 0.1 mg/kg of body weight to about 60 mg/kg of body weight of the subject.
US17/399,829 2014-01-23 2021-08-11 Human antibodies to pd-l1 Abandoned US20210380702A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/399,829 US20210380702A1 (en) 2014-01-23 2021-08-11 Human antibodies to pd-l1

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201461930582P 2014-01-23 2014-01-23
US201462089549P 2014-12-09 2014-12-09
US14/603,808 US9938345B2 (en) 2014-01-23 2015-01-23 Human antibodies to PD-L1
US15/905,653 US11117970B2 (en) 2014-01-23 2018-02-26 Human antibodies to PD-L1
US17/399,829 US20210380702A1 (en) 2014-01-23 2021-08-11 Human antibodies to pd-l1

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/905,653 Continuation US11117970B2 (en) 2014-01-23 2018-02-26 Human antibodies to PD-L1

Publications (1)

Publication Number Publication Date
US20210380702A1 true US20210380702A1 (en) 2021-12-09

Family

ID=52463178

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/603,808 Active 2035-08-08 US9938345B2 (en) 2014-01-23 2015-01-23 Human antibodies to PD-L1
US15/905,653 Active 2036-01-30 US11117970B2 (en) 2014-01-23 2018-02-26 Human antibodies to PD-L1
US17/399,829 Abandoned US20210380702A1 (en) 2014-01-23 2021-08-11 Human antibodies to pd-l1

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US14/603,808 Active 2035-08-08 US9938345B2 (en) 2014-01-23 2015-01-23 Human antibodies to PD-L1
US15/905,653 Active 2036-01-30 US11117970B2 (en) 2014-01-23 2018-02-26 Human antibodies to PD-L1

Country Status (29)

Country Link
US (3) US9938345B2 (en)
EP (2) EP3097120B1 (en)
JP (2) JP2017507650A (en)
KR (1) KR102182925B1 (en)
CN (1) CN106103482B (en)
AU (1) AU2015209238B2 (en)
BR (1) BR112016016207B1 (en)
CA (1) CA2937343C (en)
CL (1) CL2016001879A1 (en)
CY (2) CY1122005T1 (en)
DK (2) DK3473648T3 (en)
EA (1) EA034695B1 (en)
ES (2) ES2711450T3 (en)
HR (1) HRP20190749T1 (en)
HU (2) HUE055465T2 (en)
IL (1) IL246661B (en)
LT (1) LT3097120T (en)
ME (1) ME03642B (en)
MX (1) MX2016009444A (en)
MY (1) MY188355A (en)
PH (1) PH12016501384B1 (en)
PL (2) PL3473648T3 (en)
PT (2) PT3473648T (en)
RS (1) RS58638B1 (en)
SG (1) SG11201605908WA (en)
SI (1) SI3097120T1 (en)
TW (1) TWI680138B (en)
UY (1) UY35965A (en)
WO (1) WO2015112805A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023215799A1 (en) * 2022-05-04 2023-11-09 Janux Therapeutics, Inc. Tumor activated multispecific antibodies for targeting cd28 and pd-l1 and methods of use thereof

Families Citing this family (267)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BRPI0917592B1 (en) 2008-12-09 2021-08-17 Genentech, Inc ANTI-PD-L1 ANTIBODY, COMPOSITION, MANUFACTURED ARTICLES AND USES OF A COMPOSITION
HUE052198T2 (en) 2011-07-21 2021-04-28 Sumitomo Dainippon Pharma Oncology Inc Heterocyclic protein kinase inhibitors
PL2928474T3 (en) 2012-12-07 2019-05-31 Chemocentryx Inc Diazole lactams
AR095196A1 (en) 2013-03-15 2015-09-30 Regeneron Pharma SERUM FREE CELL CULTIVATION MEDIA
AR097584A1 (en) 2013-09-12 2016-03-23 Hoffmann La Roche ANTIBODY COMBINATION THERAPY AGAINST HUMAN CSF-1R AND ANTIBODIES AGAINST HUMAN PD-L1
EP3757130A1 (en) 2013-09-26 2020-12-30 Costim Pharmaceuticals Inc. Methods for treating hematologic cancers
MA38960A1 (en) 2013-09-27 2017-10-31 Genentech Inc Anti-pdl1 antibody formulations
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
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
WO2016030455A1 (en) * 2014-08-28 2016-03-03 Medimmune Limited Anti-b7-h1 and anti-ctla-4 antibodies for treating non-small lung cancer
EP3191097B1 (en) 2014-09-13 2019-10-23 Novartis AG Combination therapies
MX2017004810A (en) 2014-10-14 2017-10-16 Novartis Ag Antibody molecules to pd-l1 and uses thereof.
RS61218B1 (en) 2014-12-09 2021-01-29 Regeneron Pharma Non-human animals having a humanized cluster of differentiation 274 gene
IL292449B2 (en) 2015-03-13 2024-02-01 Cytomx Therapeutics Inc Nucleic acids encoding anti-pdl1 antibodie and methods of producing same
CN113603784A (en) 2015-05-29 2021-11-05 艾吉纳斯公司 anti-CTLA-4 antibodies and methods of use thereof
TW202340452A (en) 2015-08-04 2023-10-16 美商再生元醫藥公司 Taurine supplemented cell culture medium and methods of use
EP3353212B1 (en) 2015-09-23 2021-11-03 Regeneron Pharmaceuticals, Inc. Optimized anti-cd3 bispecific antibodies and uses thereof
CN108135934A (en) 2015-10-19 2018-06-08 永恒生物科技股份有限公司 Pass through the method for combination therapy to treat entity or lympha tumour
JP2019500892A (en) 2015-11-03 2019-01-17 ヤンセン バイオテツク,インコーポレーテツド Antibodies that specifically bind to TIM-3 and uses thereof
CN106939047B (en) * 2016-01-04 2021-08-31 江苏怀瑜药业有限公司 PD-L1 antibody and preparation method thereof
EP3964529A1 (en) * 2016-01-22 2022-03-09 Mabquest SA Non-blocking pd1 specific antibodies
US20190048085A1 (en) * 2016-02-25 2019-02-14 Cell Medica Switzerland Ag Modified cells for immunotherapy
TWI752012B (en) 2016-03-04 2022-01-11 美商Jn生物科學有限責任公司 Antibodies to tigit
CN107151269B (en) 2016-03-04 2021-07-27 四川科伦博泰生物医药股份有限公司 PDL-1 antibody, pharmaceutical composition and application thereof
US11497781B2 (en) 2016-03-10 2022-11-15 Cg Oncology, Inc. Methods of treating bladder cancer by combination therapy comprising the oncolytic adenovirus CG0070 and an immune checkpoint inhibitor
ES2904286T3 (en) * 2016-03-15 2022-04-04 Chugai Pharmaceutical Co Ltd Cancer Treatment Methods Using PD-1 Axis Binding Antagonists and Anti-GPC3 Antibodies
WO2017165683A1 (en) * 2016-03-23 2017-09-28 Novartis Ag Cell secreted minibodies and uses thereof
EP3439653B1 (en) 2016-04-07 2021-01-20 ChemoCentryx, Inc. Reducing tumor burden by administering ccr1 antagonists in combination with pd-1 inhibitors or pd-l1 inhibitors
HUE055972T2 (en) * 2016-05-09 2022-01-28 Igm Biosciences Inc Anti-pd-l1 antibodies
TWI822521B (en) 2016-05-13 2023-11-11 美商再生元醫藥公司 Methods of treating skin cancer by administering a pd-1 inhibitor
TWI781934B (en) 2016-05-27 2022-11-01 美商艾吉納斯公司 Anti-tim-3 antibodies and methods of use thereof
KR20190079713A (en) * 2016-06-13 2019-07-05 아이-맵 Anti-pd-l1 antibodies and uses thereof
CN109715196A (en) 2016-06-13 2019-05-03 转矩医疗股份有限公司 For promoting the composition and method of immune cell function
US9567399B1 (en) 2016-06-20 2017-02-14 Kymab Limited Antibodies and immunocytokines
WO2018029474A2 (en) 2016-08-09 2018-02-15 Kymab Limited Anti-icos antibodies
RU2769282C2 (en) 2016-06-20 2022-03-30 Кимаб Лимитед Anti-pd-l1 and il-2 cytokines
CA3027204A1 (en) * 2016-06-29 2018-01-04 Checkpoint Therapeutics, Inc. Pd-l1-specific antibodies and methods of using the same
CN109843921B (en) * 2016-07-07 2023-05-26 艾欧凡斯生物治疗公司 Programmed death 1 ligand 1 (PD-L1) binding proteins and methods of use thereof
ES2899036T3 (en) * 2016-08-04 2022-03-09 Innovent Biologics Suzhou Co Ltd Anti-PD-L1 nanobody and its use
JP7198752B2 (en) 2016-08-09 2023-01-04 カイマブ・リミテッド Anti-ICOS antibody
EP3504328A1 (en) 2016-08-24 2019-07-03 Regeneron Pharmaceuticals, Inc. Host cell protein modification
WO2018036852A1 (en) 2016-08-25 2018-03-01 F. Hoffmann-La Roche Ag Intermittent dosing of an anti-csf-1r antibody in combination with macrophage activating agent
TW202246349A (en) 2016-10-11 2022-12-01 美商艾吉納斯公司 Anti-lag-3 antibodies and methods of use thereof
HUE057559T2 (en) 2016-11-02 2022-06-28 Jounce Therapeutics Inc Antibodies to pd-1 and uses thereof
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
WO2018094275A1 (en) 2016-11-18 2018-05-24 Tolero Pharmaceuticals, Inc. Alvocidib prodrugs and their use as protein kinase inhibitors
AU2017367734A1 (en) * 2016-12-01 2019-06-20 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-PD-l1 antibodies for immuno-PET imaging
MD3551660T2 (en) 2016-12-07 2024-03-31 Agenus Inc Anti-CTLA-4 antibodies and methods of use thereof
BR112019011582A2 (en) 2016-12-07 2019-10-22 Agenus Inc. antibodies and their methods of use
EP3552126A1 (en) 2016-12-09 2019-10-16 Regeneron Pharmaceuticals, Inc. Systems and methods for sequencing t cell receptors and uses thereof
US10611823B2 (en) 2016-12-14 2020-04-07 Hanssen Biotech, Inc CD137 binding fibronectin type III domains
AU2017378226A1 (en) 2016-12-14 2019-06-20 Janssen Biotech, Inc. CD8A-binding fibronectin type III domains
US10597438B2 (en) 2016-12-14 2020-03-24 Janssen Biotech, Inc. PD-L1 binding fibronectin type III domains
CN108204958A (en) * 2016-12-19 2018-06-26 伊缪泰普有限公司 binding assay
EP3558360A1 (en) * 2016-12-22 2019-10-30 F. Hoffmann-La Roche AG Treatment of tumors with an anti-csf-1r antibody in combination with an anti-pd-l1 antibody after failure of anti-pd-l1/pd1 treatment
CN108264561B (en) * 2016-12-30 2021-09-10 惠和生物技术(上海)有限公司 Tri-functional molecule combining CD19, CD3 and T cell negative co-stimulatory molecule and application thereof
JP2020503883A (en) 2017-01-13 2020-02-06 アジェナス インコーポレイテッド T-cell receptor binding to NY-ESO-1 and method of using same
CN110167967B (en) * 2017-01-18 2022-08-05 豪夫迈·罗氏有限公司 Idiotypic antibodies directed against anti-PD-L1 antibodies and uses thereof
SG11201907208XA (en) 2017-02-10 2019-09-27 Regeneron Pharma Radiolabeled anti-lag3 antibodies for immuno-pet imaging
WO2018150224A1 (en) * 2017-02-16 2018-08-23 Shenzhen Runshin Bioscience Anti-programmed death-ligand 1 (pd-l1) antibodies and therapeutic uses thereof
AU2018225102A1 (en) 2017-02-21 2019-09-05 Regeneron Pharmaceuticals, Inc. Anti-PD-1 antibodies for treatment of lung cancer
BR112019017628A2 (en) 2017-02-24 2020-07-07 Macrogenics, Inc. cd137 x ta binding molecule, pharmaceutical compositions, use of cd137 x ta binding molecule, cd137 binding molecule, use of cd137 binding molecule, her2 / neu binding molecule, use of her2 binding molecule / neu, and use of a composition
EP3366703B1 (en) * 2017-02-28 2019-04-03 Ralf Kleef Immune checkpoint therapy with hyperthermia
US11603407B2 (en) 2017-04-06 2023-03-14 Regeneron Pharmaceuticals, Inc. Stable antibody formulation
TW201841942A (en) 2017-04-13 2018-12-01 美商艾吉納斯公司 Anti-CD137 antibodies and methods of use thereof
RU2665790C1 (en) * 2017-04-17 2018-09-04 Закрытое Акционерное Общество "Биокад" Monoclonal pd-l1 antibody
AR111651A1 (en) 2017-04-28 2019-08-07 Novartis Ag CONJUGATES OF ANTIBODIES THAT INCLUDE TOLL TYPE RECEIVER AGONISTS AND COMBINATION THERAPIES
RS64576B1 (en) 2017-05-01 2023-10-31 Agenus Inc Anti-tigit antibodies and methods of use thereof
AU2018278327B2 (en) 2017-06-01 2023-03-16 Cytomx Therapeutics, Inc. Activatable anti-pdl1 antibodies and methods of use thereof
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
GB201709808D0 (en) 2017-06-20 2017-08-02 Kymab Ltd Antibodies
US20200172628A1 (en) 2017-06-22 2020-06-04 Novartis Ag Antibody molecules to cd73 and uses thereof
US20190048072A1 (en) 2017-06-22 2019-02-14 Novartis Ag USE OF IL-1beta BINDING ANTIBODIES
CN110785187B (en) 2017-06-22 2024-04-05 诺华股份有限公司 Antibody molecules against CD73 and uses thereof
WO2018235056A1 (en) 2017-06-22 2018-12-27 Novartis Ag Il-1beta binding antibodies for use in treating cancer
CN110831972B (en) * 2017-06-25 2023-05-12 西雅图免疫公司 anti-PD-L1 antibodies and methods of making and using the same
JP2020525483A (en) 2017-06-27 2020-08-27 ノバルティス アーゲー Dosing regimens for anti-TIM-3 antibodies and uses thereof
EA202090003A1 (en) * 2017-07-06 2020-06-18 Мерус Н.В. BINDING MOLECULES MODULATING THE BIOLOGICAL ACTIVITY THAT A CELL MANIFESTATES
MX2020000228A (en) 2017-07-06 2020-08-10 Regeneron Pharma Cell culture process for making a glycoprotein.
AU2018302283A1 (en) 2017-07-20 2020-02-06 Novartis Ag Dosage regimens of anti-LAG-3 antibodies and uses thereof
US10730944B2 (en) 2017-07-24 2020-08-04 Regeneron Pharmaceuticals, Inc. Anti-CD8 antibodies and uses thereof
TWI799432B (en) * 2017-07-27 2023-04-21 美商再生元醫藥公司 Anti-ctla-4 antibodies and uses thereof
KR20200041897A (en) 2017-08-01 2020-04-22 에이비 스튜디오 인코포레이티드 Bispecific antibodies and uses thereof
SG11202001319QA (en) 2017-09-04 2020-03-30 Agenus Inc T cell receptors that bind to mixed lineage leukemia (mll)-specific phosphopeptides and methods of use thereof
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
EP3684955A1 (en) 2017-09-20 2020-07-29 Regeneron Pharmaceuticals, Inc. Immunotherapy methods for patients whose tumors carry a high passenger gene mutation burden
JP7382922B2 (en) * 2017-09-20 2023-11-17 中外製薬株式会社 Dosing regimen for combination therapy using PD-1 system binding antagonists and GPC3 targeting agents
MA50665A (en) 2017-09-25 2020-08-05 Chemocentryx Inc POLYTHERAPY USING A CHEMIOKIN RECEPTOR 2 (CCR2) ANTAGONIST AND A PD-1 / PD-L1 INHIBITOR
CN111225926A (en) * 2017-10-10 2020-06-02 努玛治疗有限公司 Antibodies targeting PDL1 and methods of use thereof
US20210040205A1 (en) 2017-10-25 2021-02-11 Novartis Ag Antibodies targeting cd32b and methods of use thereof
MX2020004567A (en) 2017-11-06 2020-08-13 Genentech Inc Diagnostic and therapeutic methods for cancer.
TW201925782A (en) 2017-11-30 2019-07-01 瑞士商諾華公司 BCMA-targeting chimeric antigen receptor, and uses thereof
WO2019122882A1 (en) 2017-12-19 2019-06-27 Kymab Limited Bispecific antibody for icos and pd-l1
GB201721338D0 (en) 2017-12-19 2018-01-31 Kymab Ltd Anti-icos Antibodies
EP3727629A1 (en) 2017-12-22 2020-10-28 Regeneron Pharmaceuticals, Inc. System and method for characterizing drug product impurities
CN109970857B (en) 2017-12-27 2022-09-30 信达生物制药(苏州)有限公司 anti-PD-L1 antibodies and uses thereof
CN109970856B (en) 2017-12-27 2022-08-23 信达生物制药(苏州)有限公司 anti-LAG-3 antibodies and uses thereof
KR20200104886A (en) * 2017-12-28 2020-09-04 난징 레전드 바이오테크 씨오., 엘티디. Antibodies and variants against PD-L1
WO2019136210A1 (en) * 2018-01-04 2019-07-11 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
EP3737408A1 (en) 2018-01-08 2020-11-18 Novartis AG Immune-enhancing rnas for combination with chimeric antigen receptor therapy
CA3087765A1 (en) 2018-01-08 2019-07-11 Chemocentryx, Inc. Methods of treating solid tumors with ccr2 antagonists
JP7349998B2 (en) 2018-01-31 2023-09-25 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Systems and methods for characterizing size-variant and charge-variant drug product impurities
WO2019152660A1 (en) 2018-01-31 2019-08-08 Novartis Ag Combination therapy using a chimeric antigen receptor
TWI786265B (en) 2018-02-02 2022-12-11 美商再生元醫藥公司 System and method for characterizing protein dimerization
WO2019160956A1 (en) 2018-02-13 2019-08-22 Novartis Ag Chimeric antigen receptor therapy in combination with il-15r and il15
GB201802573D0 (en) * 2018-02-16 2018-04-04 Crescendo Biologics Ltd Therapeutic molecules that bind to LAG3
MX2020008988A (en) 2018-02-28 2020-09-28 Regeneron Pharma Systems and methods for identifying viral contaminants.
TW202323813A (en) 2018-03-19 2023-06-16 美商再生元醫藥公司 Microchip capillary electrophoresis assays and reagents
EP3768719A4 (en) * 2018-03-19 2022-04-27 Abeome Corporation High affinity neutralizing monoclonal antibodies to programmed death ligand 1 (pd-l1) and uses thereof
WO2019195452A1 (en) 2018-04-04 2019-10-10 Bristol-Myers Squibb Company Anti-cd27 antibodies and uses thereof
US20210147547A1 (en) 2018-04-13 2021-05-20 Novartis Ag Dosage Regimens For Anti-Pd-L1 Antibodies And Uses Thereof
CA3096460A1 (en) * 2018-04-15 2019-10-24 Immvira Co., Limited Antibodies binding pd-1 and uses thereof
AU2019256383A1 (en) 2018-04-17 2020-11-26 Celldex Therapeutics, Inc. Anti-CD27 and anti-PD-L1 antibodies and bispecific constructs
CA3096909A1 (en) 2018-04-26 2019-10-31 Agenus Inc. Heat shock protein-binding peptide compositions and methods of use thereof
TW202016125A (en) 2018-05-10 2020-05-01 美商再生元醫藥公司 Systems and methods for quantifying and modifying protein viscosity
KR20210011999A (en) * 2018-05-24 2021-02-02 얀센 바이오테크 인코포레이티드 Monospecific and multispecific anti-TMEFF2 antibodies and uses thereof
TW202015726A (en) 2018-05-30 2020-05-01 瑞士商諾華公司 Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
WO2019232244A2 (en) 2018-05-31 2019-12-05 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2019232319A1 (en) 2018-05-31 2019-12-05 Peloton Therapeutics, Inc. Compositions and methods for inhibiting cd73
EP3802611A2 (en) 2018-06-01 2021-04-14 Novartis AG Binding molecules against bcma and uses thereof
BR112020026384A2 (en) 2018-06-23 2021-03-30 Genentech, Inc. METHODS FOR TREATING AN INDIVIDUAL WITH LUNG CANCER AND FOR TREATING AN INDIVIDUAL WITH SMALL CELL LUNG CANCER, KITS, ANTIBODY ANTI-PD-L1 AND COMPOSITION
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
IL278951B (en) 2018-07-10 2022-08-01 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
CA3104147A1 (en) 2018-07-18 2020-01-23 Genentech, Inc. Methods of treating lung cancer with a pd-1 axis binding antagonist, an antimetabolite, and a platinum agent
US11034771B2 (en) 2018-07-25 2021-06-15 I-Mab Biopharma Us Limited Anti-CD73 anti-PD-L1 bispecific antibodies
MX2021002279A (en) 2018-08-27 2021-05-27 Regeneron Pharma Use of raman spectroscopy in downstream purification.
EP3775926A1 (en) 2018-08-30 2021-02-17 Regeneron Pharmaceuticals, Inc. Methods for characterizing protein complexes
WO2020051099A1 (en) 2018-09-03 2020-03-12 Genentech, Inc. Carboxamide and sulfonamide derivatives useful as tead modulators
WO2020083277A1 (en) * 2018-10-22 2020-04-30 上海一宸医药科技有限公司 Bispecific antibody
WO2020089811A1 (en) 2018-10-31 2020-05-07 Novartis Ag Dc-sign antibody drug conjugates
JP2022513029A (en) 2018-11-14 2022-02-07 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Intralesional administration of PD-1 inhibitors to treat skin cancer
CN113490499A (en) 2018-12-04 2021-10-08 大日本住友制药肿瘤公司 CDK9 inhibitors and polymorphs thereof as active agents for the treatment of cancer
BR112021011224A2 (en) 2018-12-11 2021-08-24 Theravance Biopharma R&D Ip, Llc alk5 inhibitors
MX2021007392A (en) 2018-12-20 2021-08-24 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives.
JP2022514087A (en) 2018-12-21 2022-02-09 ノバルティス アーゲー Use of IL-1β binding antibody
WO2020128637A1 (en) 2018-12-21 2020-06-25 Novartis Ag Use of il-1 binding antibodies in the treatment of a msi-h cancer
US20220025036A1 (en) 2018-12-21 2022-01-27 Novartis Ag Use of il-1beta binding antibodies
AU2019408408A1 (en) 2018-12-21 2021-06-03 Valerio Therapeutics New conjugated nucleic acid molecules and their uses
SG11202104699TA (en) 2018-12-21 2021-07-29 Novartis Ag Use of il-1 beta antibodies in the treatment or prevention of myelodysplastic syndrome
KR20210121047A (en) * 2018-12-27 2021-10-07 기가젠, 인코포레이티드 Anti-PD-L1 binding proteins and methods of use thereof
CN111378044B (en) * 2018-12-28 2022-07-15 长春金赛药业有限责任公司 Antibody fusion protein, preparation method and application thereof
TW202043272A (en) 2019-01-14 2020-12-01 美商建南德克公司 Methods of treating cancer with a pd-1 axis binding antagonist and an rna vaccine
WO2020150492A1 (en) 2019-01-16 2020-07-23 Regeneron Pharmaceuticals, Inc. Methods for identifying free thiols in proteins
TWI756621B (en) * 2019-01-25 2022-03-01 大陸商信達生物製藥(蘇州)有限公司 Novel bispecific antibody molecules and bispecific antibodies that simultaneously bind pd-l1 and lag-3
JP2022520361A (en) 2019-02-12 2022-03-30 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 Pharmaceuticals containing heterocyclic protein kinase inhibitors
JP2022520811A (en) 2019-02-15 2022-04-01 ノバルティス アーゲー 3- (1-oxo-5- (piperidine-4-yl) isoindoline-2-yl) piperidine-2,6-dione derivative and its use
KR20210129672A (en) 2019-02-15 2021-10-28 노파르티스 아게 Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
CN113490529A (en) 2019-02-28 2021-10-08 瑞泽恩制药公司 Administration of PD-1 inhibitors for the treatment of skin cancer
CA3129963A1 (en) 2019-03-06 2020-09-10 Regeneron Pharmaceuticals, Inc. Il-4/il-13 pathway inhibitors for enhanced efficacy in treating cancer
WO2020191326A1 (en) 2019-03-20 2020-09-24 Sumitomo Dainippon Pharma Oncology, Inc. Treatment of acute myeloid leukemia (aml) with venetoclax failure
AU2020245437A1 (en) 2019-03-22 2021-09-30 Sumitomo Pharma Oncology, Inc. Compositions comprising PKM2 modulators and methods of treatment using the same
CN109929037B (en) * 2019-04-01 2023-03-17 华博生物医药技术(上海)有限公司 Conjugates to programmed death ligands and uses thereof
WO2020214995A1 (en) 2019-04-19 2020-10-22 Genentech, Inc. Anti-mertk antibodies and their methods of use
CN112996815B (en) * 2019-04-26 2024-02-20 天境生物科技(上海)有限公司 Human PD-L1 antibodies
CN113785203A (en) 2019-05-13 2021-12-10 里珍纳龙药品有限公司 Improved competitive ligand binding assays
CN111606995B (en) * 2019-06-04 2021-02-12 优睿赛思(武汉)生物科技有限公司 Anti-human PD-L1 monoclonal antibody and application thereof
EP3983450A4 (en) * 2019-06-12 2023-07-05 Nanjing GenScript Biotech Co., Ltd. Anti-pd-l1/anti-lag-3 multiple antigen binding proteins and methods of use thereof
AU2020296181A1 (en) 2019-06-21 2021-12-16 Regeneron Pharmaceuticals, Inc. Use of bispecific antigen-binding molecules that bind PSMA and CD3 in combination with 4-1BB co-stimulation
US20200399371A1 (en) 2019-06-21 2020-12-24 Regeneron Pharmaceuticals, Inc. Use of bispecific antigen-binding molecules that bind muc16 and cd3 in combination with 4-1bb co-stimulation
CA3145864A1 (en) 2019-07-03 2021-01-07 Sumitomo Dainippon Pharma Oncology, Inc. Tyrosine kinase non-receptor 1 (tnk1) inhibitors and uses thereof
CN110305906B (en) * 2019-07-18 2021-11-12 山东大学第二医院 PDL 1-targeted lentiviral vector of CAR chimeric receptor and PDL1-CAR-T cell
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
CN112409484B (en) * 2019-08-22 2023-03-21 盛禾(中国)生物制药有限公司 Multifunctional antibodies, their preparation and uses
SG11202103221QA (en) * 2019-08-29 2021-04-29 Remegen Co Ltd Anti pd-l1 antibody and use thereof
JP2022545741A (en) 2019-08-30 2022-10-28 アジェナス インコーポレイテッド ANTI-CD96 ANTIBODY AND METHODS OF USE THEREOF
US20220348651A1 (en) 2019-09-18 2022-11-03 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
TW202124446A (en) 2019-09-18 2021-07-01 瑞士商諾華公司 Combination therapies with entpd2 antibodies
EP4034870A1 (en) 2019-09-24 2022-08-03 Regeneron Pharmaceuticals, Inc. Systems and methods for chromatography use and regeneration
JP2022550420A (en) * 2019-09-30 2022-12-01 ハーバー・バイオメド・(シャンハイ)・カンパニー・リミテッド ANTI-PD-L1 ANTIGEN-BINDING PROTEIN AND APPLICATION THEREOF
US11781138B2 (en) 2019-10-14 2023-10-10 Aro Biotherapeutics Company FN3 domain-siRNA conjugates and uses thereof
US11628222B2 (en) 2019-10-14 2023-04-18 Aro Biotherapeutics Company CD71 binding fibronectin type III domains
CN114786679A (en) 2019-10-21 2022-07-22 诺华股份有限公司 Combination therapy with Vernetork and TIM-3 inhibitors
TW202128191A (en) 2019-10-21 2021-08-01 瑞士商諾華公司 Tim-3 inhibitors and uses thereof
JP2023500156A (en) * 2019-11-08 2023-01-04 チャンスー シムサー ファーマシューティカル カンパニー リミテッド Anti-human programmed cell death ligand-1 (PD-L1) antibody and uses thereof
CN112858678B (en) * 2019-11-12 2023-10-27 上海奥普生物医药股份有限公司 Creatine kinase isoenzyme detection kit, preparation method and application
CA3155989A1 (en) 2019-11-13 2021-05-20 Jason Robert ZBIEG Therapeutic compounds and methods of use
WO2021102343A1 (en) 2019-11-22 2021-05-27 Sumitomo Dainippon Pharma Oncology, Inc. Solid dose pharmaceutical composition
TW202132297A (en) 2019-11-22 2021-09-01 美商施萬生物製藥研發Ip有限責任公司 Substituted pyridines and methods of use
US11730793B2 (en) 2019-11-25 2023-08-22 Regeneron Pharmaceuticals, Inc. Sustained release formulations using non-aqueous emulsions
WO2021123902A1 (en) 2019-12-20 2021-06-24 Novartis Ag Combination of anti tim-3 antibody mbg453 and anti tgf-beta antibody nis793, with or without decitabine or the anti pd-1 antibody spartalizumab, for treating myelofibrosis and myelodysplastic syndrome
IL293752A (en) 2020-01-17 2022-08-01 Novartis Ag Combination comprising a tim-3 inhibitor and a hypomethylating agent for use in treating myelodysplastic syndrome or chronic myelomonocytic leukemia
BR122023021668A2 (en) 2020-01-21 2024-01-09 Regeneron Pharmaceuticals, Inc. METHOD FOR DETERMINING THE STABILITY OF A PROTEIN OF INTEREST
KR20210095781A (en) 2020-01-24 2021-08-03 주식회사 에이프릴바이오 A multi-specific antibody comprising a fusion construct consisting of a Fab and a bioactive effector moiety
CA3161513A1 (en) 2020-01-28 2021-08-05 Irwin DAVIDSON Antisense oligonucleotide targeting linc00518 for treating melanoma
CN116650628A (en) 2020-01-31 2023-08-29 基因泰克公司 Method for inducing neoepitope specific T cells with PD-1 axis binding antagonists and RNA vaccines
CN115443269A (en) 2020-03-31 2022-12-06 施万生物制药研发Ip有限责任公司 Substituted pyrimidines and methods of use
JP7240512B2 (en) 2020-05-26 2023-03-15 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Methods of treating cervical cancer by administering a PD-1 inhibitor
MX2022015157A (en) 2020-06-02 2023-01-16 Arcus Biosciences Inc Antibodies to tigit.
EP4165041A1 (en) 2020-06-10 2023-04-19 Theravance Biopharma R&D IP, LLC Naphthyridine derivatives useful as alk5 inhibitors
TW202214857A (en) 2020-06-19 2022-04-16 法商昂席歐公司 New conjugated nucleic acid molecules and their uses
JP2023531676A (en) 2020-06-23 2023-07-25 ノバルティス アーゲー Dosing Regimens Containing 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione Derivatives
EP4178611A1 (en) 2020-07-07 2023-05-17 BioNTech SE Therapeutic rna for hpv-positive cancer
TW202216778A (en) 2020-07-15 2022-05-01 美商安進公司 Tigit and cd112r blockade
US11787775B2 (en) 2020-07-24 2023-10-17 Genentech, Inc. Therapeutic compounds and methods of use
JP2023536164A (en) 2020-08-03 2023-08-23 ノバルティス アーゲー Heteroaryl-substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
CA3192204A1 (en) 2020-08-19 2022-02-24 Xencor, Inc. Anti-cd28 and/or anti-b7h3 compositions
MX2023002123A (en) 2020-08-26 2023-03-15 Regeneron Pharma Methods of treating cancer by administering a pd-1 inhibitor.
KR20230058094A (en) 2020-08-31 2023-05-02 리제너론 파아마슈티컬스, 인크. Asparagine supply strategies to improve cell culture performance and mitigate asparagine sequence variants
WO2022051448A1 (en) 2020-09-03 2022-03-10 Regeneron Pharmaceuticals, Inc. Methods of treating cancer pain by administering a pd-1 inhibitor
AR123855A1 (en) 2020-10-20 2023-01-18 Genentech Inc PEG-CONJUGATED ANTI-MERTK ANTIBODIES AND METHODS OF USE
WO2022093981A1 (en) 2020-10-28 2022-05-05 Genentech, Inc. Combination therapy comprising ptpn22 inhibitors and pd-l1 binding antagonists
WO2022098638A2 (en) 2020-11-04 2022-05-12 Genentech, Inc. Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies
WO2022098628A2 (en) 2020-11-04 2022-05-12 Genentech, Inc. Subcutaneous dosing of anti-cd20/anti-cd3 bispecific antibodies
IL302217A (en) 2020-11-04 2023-06-01 Genentech Inc Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies and anti-cd79b antibody drug conjugates
EP4240491A1 (en) 2020-11-06 2023-09-13 Novartis AG Cd19 binding molecules and uses thereof
WO2022095970A1 (en) * 2020-11-06 2022-05-12 百奥泰生物制药股份有限公司 Bispecific antibody and use thereof
JP2023551446A (en) 2020-11-25 2023-12-08 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Sustained release formulation using non-aqueous film emulsification
WO2022119830A1 (en) 2020-12-02 2022-06-09 Genentech, Inc. Methods and compositions for neoadjuvant and adjuvant urothelial carcinoma therapy
TW202237119A (en) 2020-12-10 2022-10-01 美商住友製藥腫瘤公司 Alk-5 inhibitors and uses thereof
KR20230121854A (en) 2020-12-17 2023-08-21 리제너론 파아마슈티컬스, 인크. Fabrication of protein-encapsulated microgels
TW202245808A (en) 2020-12-21 2022-12-01 德商拜恩迪克公司 Therapeutic rna for treating cancer
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
WO2022135457A1 (en) * 2020-12-23 2022-06-30 广东菲鹏制药股份有限公司 Anti-pd-l1 antibody and use thereof
KR20230134117A (en) 2021-01-20 2023-09-20 리제너론 파마슈티칼스 인코포레이티드 Methods for improving protein titer in cell culture
EP4284510A1 (en) 2021-01-29 2023-12-06 Novartis AG Dosage regimes for anti-cd73 and anti-entpd2 antibodies and uses thereof
WO2022173931A1 (en) 2021-02-11 2022-08-18 Regeneron Pharmaceuticals, Inc. Methods of treating cancer by administering a neoadjuvant pd-1 inhibitor
CA3170733A1 (en) 2021-02-23 2022-09-01 Petra RIETSCHEL Methods of treating lung cancer by administering a pd-1 inhibitor
JP2024512299A (en) 2021-03-03 2024-03-19 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Systems and methods for quantifying and modifying protein viscosity
JP2024511373A (en) 2021-03-18 2024-03-13 ノバルティス アーゲー Biomarkers and their use for cancer
JP2024511106A (en) 2021-03-23 2024-03-12 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Method of treating cancer in immunosuppressed or immunocompromised patients by administering PD-1 inhibitors
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
WO2022204728A1 (en) 2021-03-26 2022-09-29 Regeneron Pharmaceuticals, Inc. Methods and systems for developing mixing protocols
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
TW202309022A (en) 2021-04-13 2023-03-01 美商努法倫特公司 Amino-substituted heterocycles for treating cancers with egfr mutations
EP4330436A1 (en) 2021-04-30 2024-03-06 Genentech, Inc. Therapeutic and diagnostic methods and compositions for cancer
CN117321078A (en) 2021-04-30 2023-12-29 豪夫迈·罗氏有限公司 Administration for combination therapy with anti-CD 20/anti-CD 3 bispecific antibody and anti-CD 79B antibody drug conjugates
WO2022243378A1 (en) 2021-05-18 2022-11-24 Kymab Limited Uses of anti-icos antibodies
AR125874A1 (en) 2021-05-18 2023-08-23 Novartis Ag COMBINATION THERAPIES
WO2022251359A1 (en) 2021-05-26 2022-12-01 Theravance Biopharma R&D Ip, Llc Bicyclic inhibitors of alk5 and methods of use
IL308864A (en) 2021-06-01 2024-01-01 Regeneron Pharma Microchip capillary electrophoresis assays and reagents
GB202107994D0 (en) 2021-06-04 2021-07-21 Kymab Ltd Treatment of cancer
KR20240028452A (en) 2021-07-02 2024-03-05 제넨테크, 인크. Methods and compositions for treating cancer
AU2022312698A1 (en) 2021-07-13 2024-01-25 BioNTech SE Multispecific binding agents against cd40 and cd137 in combination therapy for cancer
US20230139492A1 (en) 2021-07-19 2023-05-04 Regeneron Pharmaceuticals, Inc. Combination of checkpoint inhibitors and an oncolytic virus for treating cancer
AU2022317820A1 (en) 2021-07-28 2023-12-14 F. Hoffmann-La Roche Ag Methods and compositions for treating cancer
WO2023010094A2 (en) 2021-07-28 2023-02-02 Genentech, Inc. Methods and compositions for treating cancer
WO2023039457A1 (en) 2021-09-08 2023-03-16 Regeneron Pharmaceuticals, Inc. A high-throughput and mass-spectrometry-based method for quantitating antibodies and other fc-containing proteins
WO2023044139A1 (en) 2021-09-20 2023-03-23 Regeneron Pharmaceuticals, Inc. Methods of controlling antibody heterogeneity
WO2023051926A1 (en) 2021-09-30 2023-04-06 BioNTech SE Treatment involving non-immunogenic rna for antigen vaccination and pd-1 axis binding antagonists
TW202321308A (en) 2021-09-30 2023-06-01 美商建南德克公司 Methods for treatment of hematologic cancers using anti-tigit antibodies, anti-cd38 antibodies, and pd-1 axis binding antagonists
CA3230985A1 (en) 2021-10-07 2023-04-13 Ross BROWNE Ph meter calibration and correction
US20230116199A1 (en) 2021-10-07 2023-04-13 Regeneron Pharmaceuticals, Inc. Systems and methods of ph modeling and control
TW202333802A (en) 2021-10-11 2023-09-01 德商拜恩迪克公司 Therapeutic rna for lung cancer
WO2023076340A1 (en) 2021-10-26 2023-05-04 Regeneron Pharmaceuticals, Inc. Systems and methods for generating laboratory water and distributing laboratory water at different temperatures
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
TW202340212A (en) 2021-11-24 2023-10-16 美商建南德克公司 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
WO2023159102A1 (en) 2022-02-17 2023-08-24 Regeneron Pharmaceuticals, Inc. Combinations of checkpoint inhibitors and oncolytic virus for treating cancer
WO2023174210A1 (en) 2022-03-14 2023-09-21 Laekna Limited Combination treatment for cancer
US20230296559A1 (en) 2022-03-18 2023-09-21 Regeneron Pharmaceuticals, Inc. Methods and systems for analyzing polypeptide variants
WO2023191816A1 (en) 2022-04-01 2023-10-05 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023187460A1 (en) * 2022-04-01 2023-10-05 Mabtree Biologics Ag Human antibody or antigen binding fragment thereof specific against pd-l1 to enhance t-cell function
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
WO2023222854A1 (en) 2022-05-18 2023-11-23 Kymab Limited Uses of anti-icos antibodies
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
WO2024020432A1 (en) 2022-07-19 2024-01-25 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024049949A1 (en) 2022-09-01 2024-03-07 Genentech, Inc. Therapeutic and diagnostic methods for bladder cancer
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

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9657102B2 (en) * 2012-09-21 2017-05-23 Regeneron Pharmaceuticals, Inc. Anti-CD3 antibodies, bispecific antigen-binding molecules that bind CD3 and CD20, and uses thereof
US9987500B2 (en) * 2014-01-23 2018-06-05 Regeneron Pharmaceuticals, Inc. Human antibodies to PD-1
US10143186B2 (en) * 2010-02-08 2018-12-04 Regeneron Pharmaceuticals, Inc. Common light chain mouse
US10736976B2 (en) * 2016-12-01 2020-08-11 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-PD-L1 antibodies for immuno-PET imaging
US20200283518A1 (en) * 2019-03-06 2020-09-10 Regeneron Pharmaceuticals, Inc. Il-4/il-13 pathway inhibitors for enhanced efficacy in treating cancer
US11117970B2 (en) * 2014-01-23 2021-09-14 Regeneron Pharmaceuticals, Inc. Human antibodies to PD-L1
US20210388091A1 (en) * 2018-11-14 2021-12-16 Regeneron Pharmaceuticals, Inc. Intralesional administration of pd-1 inhibitors for treating skin cancer
US20220259313A1 (en) * 2019-02-28 2022-08-18 Regeneron Pharmaceuticals, Inc. Administration of a pd-1 inhibitor for for treating skin cancer

Family Cites Families (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH07291996A (en) 1994-03-01 1995-11-07 Yuu Honshiyo Polypeptide related to programmed cell death in human, dna coding the same, vector consisting of the same dna, host cell transformed with the same vector, antibody of the same polypeptide and pharmaceutical composition containing the same polypeptide or the same antibody
GB9809951D0 (en) 1998-05-08 1998-07-08 Univ Cambridge Tech Binding molecules
US7087411B2 (en) 1999-06-08 2006-08-08 Regeneron Pharmaceuticals, Inc. Fusion protein capable of binding VEGF
IL148021A0 (en) 1999-08-23 2002-09-12 Dana Farber Cancer Inst Inc Novel b7-4 molecules and uses therefor
PT1210428E (en) 1999-08-23 2015-07-21 Genetics Inst Llc Pd-1, a receptor for b7-4, and uses therefor
CA2392477A1 (en) 1999-11-30 2001-06-07 Mayo Foundation For Medical Education And Research B7-h1, a novel immunoregulatory molecule
AU7309601A (en) 2000-06-28 2002-01-08 Genetics Inst Pd-l2 molecules: novel pd-1 ligands and uses therefor
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
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
US7794710B2 (en) 2001-04-20 2010-09-14 Mayo Foundation For Medical Education And Research Methods of enhancing T cell responsiveness
EP1456652A4 (en) 2001-11-13 2005-11-02 Dana Farber Cancer Inst Inc Agents that modulate immune cell activation and methods of use thereof
JP4409430B2 (en) 2002-07-03 2010-02-03 小野薬品工業株式会社 Immunostimulatory composition
JP2004104681A (en) 2002-09-12 2004-04-02 Renesas Technology Corp Input buffer circuit
US20040101920A1 (en) 2002-11-01 2004-05-27 Czeslaw Radziejewski Modification assisted profiling (MAP) methodology
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
EP2053062A1 (en) 2004-03-24 2009-04-29 Xencor, Inc. Immunoglobin variants outside the Fc region
EP2067789A1 (en) 2004-04-13 2009-06-10 F. Hoffmann-La Roche Ag Anti-P selectin antibodies
EP1740946B1 (en) 2004-04-20 2013-11-06 Genmab A/S Human monoclonal antibodies against cd20
US8257740B1 (en) 2011-08-15 2012-09-04 Gp Medical, Inc. Pharmaceutical composition of nanoparticles
CN109485727A (en) 2005-05-09 2019-03-19 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
US8246995B2 (en) 2005-05-10 2012-08-21 The Board Of Trustees Of The Leland Stanford Junior University Hydrophobic nanotubes and nanoparticles as transporters for the delivery of drugs into cells
KR20080032097A (en) 2005-06-20 2008-04-14 메다렉스, 인코포레이티드 Cd19 antibodies and their uses
DK1907424T3 (en) 2005-07-01 2015-11-09 Squibb & Sons Llc HUMAN MONOCLONAL ANTIBODIES TO PROGRAMMED death ligand 1 (PD-L1)
CN104072614B (en) 2005-07-08 2017-04-26 生物基因Ma公司 Anti-alpha[v]beta[6] antibodies and uses thereof
EP1820513A1 (en) 2006-02-15 2007-08-22 Trion Pharma Gmbh Destruction of tumor cells expressing low to medium levels of tumor associated target antigens by trifunctional bispecific antibodies
US8216996B2 (en) 2006-03-03 2012-07-10 Ono Pharmaceutical Co., Ltd. Multimer of extracellular domain of cell surface functional molecule
WO2007143168A2 (en) 2006-06-02 2007-12-13 Regeneron Pharmaceuticals, Inc. High affinity antibodies to human il-6 receptor
ES2437327T3 (en) 2007-06-18 2014-01-10 Merck Sharp & Dohme B.V. Antibodies for the human programmed PD-1 receptor of programmed death
WO2009014708A2 (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
US9243052B2 (en) 2007-08-17 2016-01-26 Daniel Olive Method for treating and diagnosing hematologic malignancies
WO2009030285A1 (en) 2007-09-07 2009-03-12 Ablynx N.V. Binding molecules with multiple binding sites, compositions comprising the same and uses thereof
US8747847B2 (en) 2008-02-11 2014-06-10 Curetech Ltd. Monoclonal antibodies for tumor treatment
EP2262837A4 (en) 2008-03-12 2011-04-06 Merck Sharp & Dohme Pd-1 binding proteins
PE20110435A1 (en) 2008-08-25 2011-07-20 Amplimmune Inc ANTAGONIST COMPOSITIONS OF PD-1
JP2012501670A (en) 2008-09-12 2012-01-26 アイシス・イノベーション・リミテッド 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
KR102097887B1 (en) 2008-09-26 2020-04-06 다나-파버 캔서 인스티튜트 인크. Human anti-pd-1, pd-l1, and pd-l2 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
BRPI0917592B1 (en) * 2008-12-09 2021-08-17 Genentech, Inc ANTI-PD-L1 ANTIBODY, COMPOSITION, MANUFACTURED ARTICLES AND USES OF A COMPOSITION
EP3192811A1 (en) 2009-02-09 2017-07-19 Université d'Aix-Marseille Pd-1 antibodies and pd-l1 antibodies and uses thereof
CN102471378B (en) 2009-06-26 2014-04-02 瑞泽恩制药公司 Readily isolated bispecific antibodies with native immuneoglobulin format
EP3375791A1 (en) 2009-09-30 2018-09-19 Memorial Sloan Kettering Cancer Center Combination immunotherapy for the treatment of cancer
RS56469B1 (en) * 2009-11-24 2018-01-31 Medimmune Ltd Targeted binding agents against b7-h1
WO2011066342A2 (en) 2009-11-24 2011-06-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
TW201134488A (en) 2010-03-11 2011-10-16 Ucb Pharma Sa PD-1 antibodies
CA2791930A1 (en) 2010-03-11 2011-09-15 Kerry Louise Tyson Pd-1 antibody
JP2013532153A (en) 2010-06-18 2013-08-15 ザ ブリガム アンド ウィメンズ ホスピタル インコーポレイテッド Bispecific antibodies against TIM-3 and PD-1 for immunotherapy against chronic immune disease
DE102010025653B4 (en) 2010-06-30 2018-09-20 Kennametal Inc. Rotary cutting tool
EP2910572B1 (en) 2010-11-11 2017-09-06 Versitech Limited Soluble pd-1 variants, fusion constructs, and uses thereof
KR101970025B1 (en) * 2011-04-20 2019-04-17 메디뮨 엘엘씨 Antibodies and other molecules that bind b7-h1 and pd-1
US8686119B2 (en) 2011-07-24 2014-04-01 Curetech Ltd. Variants of humanized immunomodulatory monoclonal antibodies
CN103842030B (en) 2011-08-01 2018-07-31 霍夫曼-拉罗奇有限公司 Use the method for PD-1 axis binding antagonists and mek inhibitor treating cancer
DK2785375T3 (en) 2011-11-28 2020-10-12 Merck Patent Gmbh ANTI-PD-L1 ANTIBODIES AND USES THEREOF
GB201120527D0 (en) 2011-11-29 2012-01-11 Ucl Business Plc Method
AU2013211824B2 (en) 2012-01-27 2017-06-01 Gliknik Inc. Fusion proteins comprising IgG2 hinge domains
CA2865643A1 (en) * 2012-03-16 2013-09-19 Regeneron Pharmaceuticals, Inc. Histidine engineered light chain antibodies and genetically modified non-human animals for generating the same
CA2871751C (en) * 2012-05-04 2021-08-24 Dana-Farber Cancer Institute, Inc. Affinity matured anti-ccr4 humanized monoclonal antibodies and methods of use
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
US20130303250A1 (en) 2012-05-11 2013-11-14 Ryan Moore Method of Playing a Card Game
WO2013173223A1 (en) 2012-05-15 2013-11-21 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting pd-1/pd-l1 signaling
SG10201603055WA (en) 2012-05-31 2016-05-30 Genentech Inc Methods Of Treating Cancer Using PD-L1 Axis Binding Antagonists And VEGF Antagonists
UY34887A (en) * 2012-07-02 2013-12-31 Bristol Myers Squibb Company Una Corporacion Del Estado De Delaware OPTIMIZATION OF ANTIBODIES THAT FIX THE LYMPHOCYTE ACTIVATION GEN 3 (LAG-3) AND ITS USES
RS56624B1 (en) 2012-10-02 2018-03-30 Bristol Myers Squibb Co Combination of anti-kir antibodies and anti-pd-1 antibodies to treat cancer
US10034939B2 (en) 2012-10-26 2018-07-31 The University Of Chicago Synergistic combination of immunologic inhibitors for the treatment of cancer
TWI682941B (en) 2013-02-01 2020-01-21 美商再生元醫藥公司 Antibodies comprising chimeric constant domains
DK2958588T3 (en) 2013-02-22 2017-11-20 Curevac Ag Combination of vaccination and inhibition of PD-1 pathway
EP3305812B1 (en) 2013-03-14 2020-06-17 Bristol-Myers Squibb Company Combination of dr5 agonist and anti-pd-1 antagonist and methods of use
KR102158924B1 (en) 2013-03-15 2020-09-22 제넨테크, 인크. Biomarkers and methods of treating pd-1 and pd-l1 related conditions
BR112015026823A2 (en) 2013-05-02 2017-09-05 Anaptysbio Inc ISOLATED IMMUNOGLOBULIN HEAVY CHAIN POLYPEPTIDE, ISOLATED IMMUNOGLOBULIN LIGHT CHAIN POLYPEPTIDE, ISOLATED OR PURIFIED NUCLEIC ACID SEQUENCE, VECTOR, PROGRAMMED DEATH-1 PROTEIN (PD-1) BINDING AGENT ISOLATED, CELL ISOLATED, COMPOSITION, AND USE OF THE BINDING AGENT OF THE ISOLATED PD-1
WO2014194293A1 (en) 2013-05-30 2014-12-04 Amplimmune, Inc. Improved methods for the selection of patients for pd-1 or b7-h4 targeted therapies, and combination therapies thereof
US20160145355A1 (en) 2013-06-24 2016-05-26 Biomed Valley Discoveries, Inc. Bispecific antibodies
ES2819209T3 (en) 2013-07-16 2021-04-15 Hoffmann La Roche Cancer treatment procedures using PD-1 axis binding antagonists and TIGIT inhibitors
WO2015016718A1 (en) 2013-08-02 2015-02-05 Bionovion Holding B.V. Combining cd27 agonists and immune checkpoint inhibition for immune stimulation
WO2015026634A1 (en) 2013-08-20 2015-02-26 Merck Sharp & Dohme Corp. Treating cancer with a combination of a pd-1 antagonist and dinaciclib
AR097306A1 (en) 2013-08-20 2016-03-02 Merck Sharp & Dohme MODULATION OF TUMOR IMMUNITY
EP3178849B1 (en) 2013-09-20 2019-03-20 Bristol-Myers Squibb Company Combination of anti-lag-3 antibodies and anti-pd-1 antibodies to treat tumors
EP3757130A1 (en) 2013-09-26 2020-12-30 Costim Pharmaceuticals Inc. Methods for treating hematologic cancers
MA38960A1 (en) 2013-09-27 2017-10-31 Genentech Inc Anti-pdl1 antibody formulations
MX2016007958A (en) 2013-12-17 2016-08-03 Genentech Inc Anti-cd3 antibodies and methods of use.
JOP20200094A1 (en) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc Antibody molecules to pd-1 and uses thereof
TWI754319B (en) 2014-03-19 2022-02-01 美商再生元醫藥公司 Methods and antibody compositions for tumor treatment
CA2949121A1 (en) 2014-05-15 2015-11-19 Bristol-Myers Squibb Company Treatment of lung cancer using a combination of an anti-pd-1 antibody and another anti-cancer agent
US10092645B2 (en) 2014-06-17 2018-10-09 Medimmune Limited Methods of treatment with antagonists against PD-1 and PD-L1 in combination with radiation therapy
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
MX2017004810A (en) 2014-10-14 2017-10-16 Novartis Ag Antibody molecules to pd-l1 and uses thereof.
IL292449B2 (en) * 2015-03-13 2024-02-01 Cytomx Therapeutics Inc Nucleic acids encoding anti-pdl1 antibodie and methods of producing same
US20180155429A1 (en) 2015-05-28 2018-06-07 Bristol-Myers Squibb Company Treatment of pd-l1 positive lung cancer using an anti-pd-1 antibody
KR20180040138A (en) 2015-07-13 2018-04-19 싸이톰스 테라퓨틱스, 인크. Anti-PD-1 antibodies, activatable anti-PD-1 antibodies, and methods of using them
US9830706B2 (en) 2015-09-17 2017-11-28 Skycatch, Inc. Generating georeference information for aerial images
PT3394103T (en) 2015-12-22 2023-09-04 Regeneron Pharma Combination of anti-pd-1 antibodies and bispecific anti-cd20/anti-cd3 antibodies to treat cancer
US20190048085A1 (en) * 2016-02-25 2019-02-14 Cell Medica Switzerland Ag Modified cells for immunotherapy

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10143186B2 (en) * 2010-02-08 2018-12-04 Regeneron Pharmaceuticals, Inc. Common light chain mouse
US9657102B2 (en) * 2012-09-21 2017-05-23 Regeneron Pharmaceuticals, Inc. Anti-CD3 antibodies, bispecific antigen-binding molecules that bind CD3 and CD20, and uses thereof
US9987500B2 (en) * 2014-01-23 2018-06-05 Regeneron Pharmaceuticals, Inc. Human antibodies to PD-1
US11117970B2 (en) * 2014-01-23 2021-09-14 Regeneron Pharmaceuticals, Inc. Human antibodies to PD-L1
US10736976B2 (en) * 2016-12-01 2020-08-11 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-PD-L1 antibodies for immuno-PET imaging
US20220184241A1 (en) * 2016-12-01 2022-06-16 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-pd-l1 antibodies for immuno-pet imaging
US20210388091A1 (en) * 2018-11-14 2021-12-16 Regeneron Pharmaceuticals, Inc. Intralesional administration of pd-1 inhibitors for treating skin cancer
US20220259313A1 (en) * 2019-02-28 2022-08-18 Regeneron Pharmaceuticals, Inc. Administration of a pd-1 inhibitor for for treating skin cancer
US20200283518A1 (en) * 2019-03-06 2020-09-10 Regeneron Pharmaceuticals, Inc. Il-4/il-13 pathway inhibitors for enhanced efficacy in treating cancer

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023215799A1 (en) * 2022-05-04 2023-11-09 Janux Therapeutics, Inc. Tumor activated multispecific antibodies for targeting cd28 and pd-l1 and methods of use thereof

Also Published As

Publication number Publication date
TWI680138B (en) 2019-12-21
PH12016501384A1 (en) 2016-08-15
HUE043672T2 (en) 2019-09-30
EP3097120B1 (en) 2019-01-23
US20180186883A1 (en) 2018-07-05
HUE055465T2 (en) 2021-11-29
PL3473648T3 (en) 2021-11-29
JP7106594B2 (en) 2022-07-26
SG11201605908WA (en) 2016-08-30
AU2015209238A1 (en) 2016-07-21
ES2878091T3 (en) 2021-11-18
CL2016001879A1 (en) 2017-08-25
KR102182925B1 (en) 2020-11-25
IL246661B (en) 2019-05-30
PT3097120T (en) 2019-05-08
HRP20190749T1 (en) 2019-08-23
NZ721656A (en) 2020-10-30
EP3473648A1 (en) 2019-04-24
CY1122005T1 (en) 2020-10-14
CY1124437T1 (en) 2022-07-22
JP2020115876A (en) 2020-08-06
EP3097120A1 (en) 2016-11-30
PH12016501384B1 (en) 2016-08-15
SI3097120T1 (en) 2019-03-29
UY35965A (en) 2015-08-31
JP2017507650A (en) 2017-03-23
IL246661A0 (en) 2016-08-31
WO2015112805A8 (en) 2016-07-28
KR20160128309A (en) 2016-11-07
LT3097120T (en) 2019-04-25
DK3097120T3 (en) 2019-03-18
CA2937343C (en) 2022-08-16
EA034695B1 (en) 2020-03-06
RS58638B1 (en) 2019-05-31
MX2016009444A (en) 2017-01-16
EA201691487A1 (en) 2016-12-30
DK3473648T3 (en) 2021-06-28
BR112016016207B1 (en) 2023-10-24
WO2015112805A1 (en) 2015-07-30
PL3097120T3 (en) 2019-07-31
ES2711450T3 (en) 2019-05-03
CA2937343A1 (en) 2015-07-30
US20150203580A1 (en) 2015-07-23
TW201620936A (en) 2016-06-16
EP3473648B1 (en) 2021-06-02
CN106103482A (en) 2016-11-09
BR112016016207A2 (en) 2017-10-03
US9938345B2 (en) 2018-04-10
US11117970B2 (en) 2021-09-14
ME03642B (en) 2020-07-20
AU2015209238B2 (en) 2019-09-12
PT3473648T (en) 2021-07-12
MY188355A (en) 2021-12-02
CN106103482B (en) 2021-04-20

Similar Documents

Publication Publication Date Title
US20210380702A1 (en) Human antibodies to pd-l1
US20200330794A1 (en) Human antibodies to pd-1
US11692032B2 (en) Anti-LAG3 antibodies and uses thereof
NZ721656B2 (en) Human antibodies to pd-l1

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION UNDERGOING PREEXAM PROCESSING

AS Assignment

Owner name: REGENERON PHARMACEUTICALS, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PAPADOPOULOS, NICHOLAS J.;MURPHY, ANDREW J.;THURSTON, GAVIN;AND OTHERS;SIGNING DATES FROM 20150622 TO 20150731;REEL/FRAME:058845/0972

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