WO2016068801A1 - Anti-pd-1 antibodies - Google Patents

Anti-pd-1 antibodies Download PDF

Info

Publication number
WO2016068801A1
WO2016068801A1 PCT/SG2015/050413 SG2015050413W WO2016068801A1 WO 2016068801 A1 WO2016068801 A1 WO 2016068801A1 SG 2015050413 W SG2015050413 W SG 2015050413W WO 2016068801 A1 WO2016068801 A1 WO 2016068801A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
antigen binding
binding fragment
cdr3
Prior art date
Application number
PCT/SG2015/050413
Other languages
French (fr)
Inventor
Cheng-I Wang
Hsueh Ling Janice OH
Siok Ping YEO
Jianrong Lionel LOW
Hwee Ching TAN
Original Assignee
Agency For Science, Technology And Research
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
Priority to RU2017116847A priority Critical patent/RU2715628C2/en
Priority to JP2017540959A priority patent/JP6701213B2/en
Priority to KR1020177014305A priority patent/KR20170070243A/en
Priority to SG11201703344WA priority patent/SG11201703344WA/en
Application filed by Agency For Science, Technology And Research filed Critical Agency For Science, Technology And Research
Priority to MYPI2017701430A priority patent/MY191581A/en
Priority to CA2965623A priority patent/CA2965623A1/en
Priority to AU2015340054A priority patent/AU2015340054A1/en
Priority to CN201580071389.1A priority patent/CN107438621B/en
Priority to US15/521,434 priority patent/US10280224B2/en
Priority to EP15855595.3A priority patent/EP3212671A4/en
Publication of WO2016068801A1 publication Critical patent/WO2016068801A1/en
Priority to US15/495,048 priority patent/US9771425B2/en
Priority to IL251963A priority patent/IL251963A0/en
Priority to PH12017500803A priority patent/PH12017500803A1/en
Priority to US16/360,445 priority patent/US11072659B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/005Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies constructed by phage libraries
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • 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
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/51Complete heavy chain or Fd fragment, i.e. VH + CH1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/515Complete light chain, i.e. VL + CL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • 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
    • 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
    • C07K2317/74Inducing cell proliferation
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/70521CD28, CD152

Definitions

  • the present invention relates to antibodies that bind to programmed cell death 1 (PD-1).
  • PD-1 programmed cell death 1
  • T-cell exhaustion is a state of T-cell dysfunction that arises during many chronic infections and cancer. It is defined by poor T-cell effector function, sustained expression of inhibitory receptors and a transcriptional state distinct from that of functional effector or memory T- cells. Exhaustion prevents optimal control of infection and tumors. (E John Wherry., Nature Immunology 12, 492-499 (201 1)).
  • T-cell exhaustion is characterized by the stepwise and progressive loss of T-cell functions. Exhaustion is well-defined during chronic lymphocytic choriomeningitis virus infection and commonly develops under conditions of antigen-persistence, which occur following many chronic infections including hepatitis B virus, hepatitis C virus and human immunodeficiency virus infections, as well as during tumor metastasis. Exhaustion is not a uniformly disabled setting as a gradation of phenotypic and functional defects can manifest, and these cells are distinct from prototypic effector, memory and also anergic T cells. Exhausted T cells most commonly emerge during high-grade chronic infections, and the levels and duration of antigenic stimulation are critical determinants of the process. (Yi et al., Immunology Apr 2010; 129(4):474-481).
  • Circulating human tumor-specific CD8 + T cells may be cytotoxic and produce cytokines in vivo, indicating that self- and tumor-specific human CD8 + T cells can reach functional competence after potent immunotherapy such as vaccination with peptide, incomplete Freund's adjuvant (I FA), and CpG or after adoptive transfer.
  • I FA incomplete Freund's adjuvant
  • CpG after adoptive transfer.
  • T-cells from metastasis are functionally deficient, with abnormally low cytokine production and upregulation of the inhibitory receptors PD-1 , CTLA-4, and TIM-3.
  • T-cells isolated from melanoma tissue can restore IFN- ⁇ production after short-term in vitro culture.
  • this functional impairment involves further molecular pathways, possibly resembling T-cell exhaustion or anergy as defined in animal models.
  • PD-1 also called CD279, is a type I membrane protein encoded in humans by the PDCD1 gene. It has two ligands, PD-L1 and PD-L2.
  • the PD-1 pathway is a key immune-inhibitory mediator of T-cell exhaustion. Blockade of this pathway can lead to T-cell activation, expansion, and enhanced effector functions. As such, PD-1 negatively regulates T cell responses.
  • PD-1 has been identified as a marker of exhausted T cells in chronic disease states, and blockade of PD-1 :PD-1 L interactions has been shown to partially restore T cell function. (Sakuishi et al., JEM Vol. 207, September 27, 2010, pp2187-2194).
  • Nivolumab (BMS-936558) is an anti-PD-1 antibody that was approved for the treatment of melanoma in Japan in July 2014. Other anti-PD-1 antibodies are described in WO 2010/077634, WO 2006/121 168, WO2008/156712 and WO2012/135408.
  • T cell immunoglobulin mucin 3 (TIM-3) is an immune regulator identified as being upregulated on exhausted CD8 + T cells (Sakuishi et al., JEM Vol. 207, September 27, 2010, pp2187-2194). TIM-3 was originally identified as being selectively expressed on
  • Both TIM-3 and PD-1 can function as negative regulators of T cell responses and combined targeting of the TIM-3 and PD-1 pathways is more effective in controlling tumor growth than targeting either pathway alone.
  • the present invention is concerned with antibodies, or antigen binding fragments, that bind to PD-1.
  • Heavy and light chain polypeptides are also disclosed.
  • the antibodies, antigen binding fragments and polypeptides may be provided in isolated and/or purified form and may be formulated into compositions suitable for use in research, therapy and diagnosis.
  • the antibody, or antigen binding fragment, or polypeptide may be effective to restore T-cell function in T-cells, e.g. CD8 + T-cells, exhibiting T-cell exhaustion or T-cell anergy.
  • an antibody, or antigen binding fragment may comprise the amino acid sequences i) to iii), or the amino acid sequences iv) to vi), or preferably the amino acid sequences i) to vi): i) LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25) ii) LC-CDR2: SNNQRPS (SEQ ID NO:26) iii) LC-CDR3: X1X2WDDX3X4X5GX6X7 (SEQ ID NO:53) iv) HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR1 SYGMH (SEQ ID NO:89)
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • this sequence may be comprised in the larger sequence GFTFSSYGMH (SEQ ID NO:39).
  • LC-CDR3 is one of ASWDDVLYGSV (SEQ ID NO:27),
  • HC-CDR3 is one of
  • the antibody, or antigen binding fragment may comprise at least one light chain variable region incorporating the following CDRs:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 ASWDDVLYGSV (SEQ ID NO:27)
  • the antibody, or antigen binding fragment may comprise at least one light chain variable region incorporating the following CDRs:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 ASWDDYYYGTI (SEQ ID NO:28)
  • the antibody, or antigen binding fragment may comprise at least one light chain variable region incorporating the following CDRs:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 ASWDDYLRGTV (SEQ ID NO:29)
  • the antibody, or antigen binding fragment may comprise at least one light chain variable region incorporating the following CDRs:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 SAWDDYLHGTV (SEQ ID NO:30)
  • the antibody, or antigen binding fragment may comprise at least one light chain variable region incorporating the following CDRs:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 ASWDDYVRGTM (SEQ ID NO:31)
  • the antibody, or antigen binding fragment may comprise at least one light chain variable region incorporating the following CDRs:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • the antibody, or antigen binding fragment may comprise at least one light chain variable region incorporating the following CDRs:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 SSWDDDARGTI (SEQ ID NO:33)
  • the antibody, or antigen binding fragment may comprise at least one light chain variable region incorporating the following CDRs:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 AAWDDVYYGTI (SEQ ID NO:34)
  • the antibody, or antigen binding fragment may comprise at least one light chain variable region incorporating the following CDRs:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 ASWDDSLYGTV (SEQ ID NO:35)
  • the antibody, or antigen binding fragment may comprise at least one light chain variable region incorporating the following CDRs:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 AAWDDAYYGTI (SEQ ID NO:36)
  • the antibody, or antigen binding fragment may comprise at least one light chain variable region incorporating the following CDRs:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 ASWDDVYRGTV (SEQ ID NO:37)
  • the antibody, or antigen binding fragment may comprise at least one light chain variable region incorporating the following CDRs:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 SSWDDSLYGTI (SEQ ID NO:38)
  • the antibody, or antigen binding fragment may comprise at least one heavy chain variable region incorporating the following CDRs:
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGKDH (SEQ ID NO:41)
  • the antibody, or antigen binding fragment may comprise at least one heavy chain variable region incorporating the following CDRs:
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 D LG AG PYYYG KD V (SEQ ID NO:42)
  • the antibody, or antigen binding fragment may comprise at least one heavy chain variable region incorporating the following CDRs:
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 D YG AG PYYYG M D V (SEQ ID NO:43)
  • the antibody, or antigen binding fragment may comprise at least one heavy chain variable region incorporating the following CDRs:
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • the antibody, or antigen binding fragment may comprise at least one heavy chain variable region incorporating the following CDRs: HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGMDV (SEQ ID NO:45)
  • the antibody, or antigen binding fragment may comprise at least one heavy chain variable region incorporating the following CDRs:
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGMDV (SEQ ID NO:46)
  • the antibody, or antigen binding fragment may comprise at least one heavy chain variable region incorporating the following CDRs:
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGMDV (SEQ ID NO:47)
  • the antibody, or antigen binding fragment may comprise at least one heavy chain variable region incorporating the following CDRs:
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGMDV (SEQ ID NO:48)
  • the antibody, or antigen binding fragment may comprise at least one heavy chain variable region incorporating the following CDRs:
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGMDV (SEQ ID NO:49)
  • the antibody, or antigen binding fragment may comprise at least one heavy chain variable region incorporating the following CDRs:
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or SYGMH (SEQ ID NO: 89)
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DYGAGPYYYGMDV (SEQ ID NO:50)
  • the antibody, or antigen binding fragment may comprise at least one heavy chain variable region incorporating the following CDRs:
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGSGYYLYGMDV (SEQ ID NO:51)
  • the antibody, or antigen binding fragment may comprise at least one heavy chain variable region incorporating the following CDRs:
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGMDV (SEQ ID NO:52)
  • the antibody may comprise at least one light chain variable region incorporating the CDRs shown in Figures 1 or 3.
  • the antibody may comprise at least one heavy chain variable region incorporating the CDRs shown in Figures 2 or 3.
  • the antibody may comprise at least one light chain variable region (VL) comprising the amino acid sequence of one of SEQ ID NOs 1 , 25, 26, 27 or 2, 25, 26, 28, or 3, 25, 26, 29 or 4, 25, 26, 30, or 5, 25, 26, 31 or 6, 25, 26, 32 or 7, 25, 26, 33 or 8, 25, 26, 34 or 9, 25, 26, 35 or 10, 25, 26 36 or 11 , 25, 26, 37 or 12, 25, 26, 38, or one of the amino acid sequences shown in Figure 1 or an amino acid sequence having at least 70%, more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to one of SEQ ID NOs 1 , 25, 26, 27 or 2, 25, 26, 28, or 3, 25, 26,
  • the antibody may comprise at least one heavy chain variable region (VH) comprising the amino acid sequence of one of SEQ ID NOs 13, 39 or 89, 40, 41 or 14, 39 or 89, 40, 42, or 15, 39 or 89, 40, 43 or 16, 39 or 89, 40, 44 or 17, 39 or 89, 40, 45 or 18, 39 or 89, 40, 46 or 19, 39 or 89, 40, 47 or 20, 39 or 89, 40, 48 or 21 , 39 or 89, 40, 49 or 22, 39 or 89, 40, 50 or 23, 39 or 89, 40, 51 or 24, 39 or 89, 40, 52, or one of the amino acid sequences shown in Figure 2 or an amino acid sequence having at least 70%, more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to one of SEQ ID NOs 13, 39, 40, 41 or 14,
  • the antibody may comprise at least one light chain variable region comprising the amino acid sequence of one of SEQ ID NOs 1 , 25, 26, 27 or 2, 25, 26, 28, or 3, 25, 26, 29 or 4, 25, 26, 30, or 5, 25, 26, 31 or 6, 25, 26, 32 or 7, 25, 26, 33 or 8, 25, 26, 34 or 9, 25, 26, 35 or 10, 25, 26 36 or 1 1 , 25, 26, 37 or 12, 25, 26, 38, or to one of the amino acid sequences shown in Figure 1 (or an amino acid sequence having at least 70%, more preferably one of at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100%, sequence identity to one of SEQ ID NOs 1 , 25, 26, 27 or 2, 25, 26, 28, or 3, 25, 26, 29 or 4, 25, 26, 30, or 5, 25, 26, 31 or 6, 25, 26, 32 or 7, 25, 26, 33 or 8, 25, 26, 34 or 9, 25, 26, 35 or 10, 25, 26 36 or 1 1 , 25, 26, 37 or 12, 25, 26, 38, or to to one of the amino acid sequences of the VL chain amino acid sequence shown in Figure 1 ) and at least one heavy chain
  • the antibody may optionally bind PD-1.
  • the antibody may optionally have amino acid sequence components as described above.
  • the antibody may be an IgG.
  • an in vitro complex, optionally isolated, comprising an antibody, or antigen binding fragment, as described herein, bound to PD-1 is provided.
  • an isolated heavy chain variable region polypeptide comprising the following CDRs:
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DZ 1 GZ 2 GZ 3 YZ 4 YGZ 5 DZ 6 (SEQ ID NO:54)
  • Z ⁇ L or Y
  • Z 2 A or S
  • Z 3 P or Y
  • Z 4 Y or L
  • Z 5 K
  • Z 6 H or V.
  • HC-CDR3 is one of DLGAGPYYYGKDH (SEQ ID NO:41), D LG AG PYYYG KD V (SEQ ID NO:42), DYGAGPYYYGMDV (SEQ ID NO:43),
  • DYGAGPYYYGMDV (SEQ ID NO:50), DLGSGYYLYGMDV (SEQ ID NO:51), or
  • an antibody, or antigen binding fragment comprising a heavy chain and a light chain variable region sequence, wherein:
  • the heavy chain comprises a HC-CDR1 , HC-CDR2, HC-CDR3, having at least 85% overall sequence identity to
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or SYGMH (SEQ ID NO:89),
  • HC-CDR3 is one of DZ ⁇ ZzGZsYZ ⁇ GZsDZe (SEQ ID NO:54),
  • DLGAGPYYYGKDH (SEQ ID NO:41), D LGAG PYYYG KDV (SEQ ID NO:42), DYGAGPYYYGMDV (SEQ ID NO:43), DLGAGPYYYGLDV (SEQ ID NO:44), DLGAGPYYYGMDV (SEQ ID NO:45), DLGAGPYYYGMDV (SEQ ID NO:46),
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25),
  • LC-CDR2 SNNQRPS (SEQ ID NO:26),
  • LC-CDR3 is one of X 1 X 2 WDDX 3 X4X5GX6X7 (SEQ ID NO:53), ASWDDVLYGSV
  • SEQ ID NO:27 ASWDDYYYGTI (SEQ ID NO:28), ASWDDYLRGTV (SEQ ID NO:29), SAWDDYLHGTV (SEQ ID NO:30), ASWDDYVRGTM (SEQ ID NO:31), SSWDDFLRGTV (SEQ ID NO:32), SSWDDDARGTI (SEQ ID NO:33),
  • the degree of sequence identity may be one of 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
  • an antibody, or antigen binding fragment, optionally isolated comprising a heavy chain and a light chain variable region sequence, wherein:
  • the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence of one of SEQ ID NOs: 13 to 24 ( Figure 2), and
  • the light chain sequence has at least 85% sequence identity to the light chain sequence of one of: SEQ ID NOs:1 to 12 ( Figure 1).
  • the degree of sequence identity may be one of 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
  • the antibody, antigen binding fragment, or polypeptide further comprises variable region heavy chain framework sequences between the CDRs according to the arrangement HCFR1 :HC-CDR1 :HCFR2:HC-CDR2:HCFR3:HC- CDR3:HCFR4.
  • the framework sequences may be derived from human consensus framework sequences.
  • an isolated light chain variable region polypeptide optionally in combination with a heavy chain variable region polypeptide as described herein, is provided, the light chain variable region polypeptide comprising the following CDRs:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 is one of ASWDDVLYGSV (SEQ ID NO:27),
  • ASWDDYYYGTI (SEQ ID NO:28), ASWDDYLRGTV (SEQ ID NO:29), SAWDDYLHGTV (SEQ ID NO:30), ASWDDYVRGTM (SEQ ID NO:31), SSWDDFLRGTV (SEQ ID NO:32), SSWDDDARGTI (SEQ ID NO:33), AAWDDVYYGTI (SEQ ID NO:34), ASWDDSLYGTV (SEQ ID NO:35), AAWDDAYYGTI (SEQ ID NO:36), ASWDDVYRGTV (SEQ ID NO:37), or SSWDDSLYGTI (SEQ ID NO:38).
  • the antibody, antigen binding fragment, or polypeptide further comprises variable region light chain framework sequences between the CDRs according to the arrangement LCFR1 :LC-CDR1 :LCFR2:LC-CDR2:LCFR3:LC-CDR3:LCFR4.
  • the framework sequences may be derived from human consensus framework sequences.
  • the antibody, or antibody binding fragment may further comprise a human constant region.
  • a human constant region For example selected from one of lgG1 , lgG2, lgG3 and lgG4.
  • the antibody, or antibody binding fragment may further comprise a murine constant region.
  • a murine constant region selected from one of lgG1 , lgG2A, lgG2B and lgG3.
  • an antibody or antigen binding fragment is provided, optionally isolated, which is capable of binding to PD-1 , and which is a bispecific antibody or a bispecific antigen binding fragment.
  • the bispecific antibody or bispecific antigen binding fragment comprises an antigen binding fragment or polypeptide capable of binding to PD-1 as described herein, and additionally comprises an antigen binding domain which is capable of binding to another target protein, e.g. a target protein other than PD-1.
  • the target protein is a cell surface receptor. In some embodiments, the target protein is a cell surface receptor expressed on the cell surface of immune cells, e.g. T cells. In some embodiments, the target protein may be a member of the CD28 family. In some embodiments, the member of the CD28 family is selected from TIM-3, LAG3, ICOS, CTLA4, BTLA or CD28.
  • a composition e.g. a pharmaceutical composition or medicament, is provided.
  • the composition may comprise an antibody, antigen binding fragment, or polypeptide as described herein and at least one
  • an isolated nucleic acid encoding an antibody, antigen binding fragment, or polypeptide as described herein is provided.
  • the nucleic acid may have a sequence of one of SEQ ID NOs 55, 56, 58, 59, 60, 61 , 62, 63, 64, 65, 66, 67, 68, 69, 70, 71 , 72, 73, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 87, or 88 ( Figure 4), or a coding sequence which is degenerate as a result of the genetic code, or may have a nucleotide sequence having at least 70% identity thereto, optionally one of 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
  • a vector comprising a nucleic acid described herein.
  • a host cell comprising the vector.
  • the host cell may be eukaryotic, or mammalian, e.g. Chinese Hamster Ovary (CHO), or human or may be a prokaryotic cell, e.g. E. coli.
  • a method for making an antibody, or antigen binding fragment or polypeptide as described herein comprising culturing a host cell as described herein under conditions suitable for the expression of a vector encoding the antibody, or antigen binding fragment or polypeptide, and recovering the antibody, or antigen binding fragment or polypeptide.
  • an antibody, antigen binding fragment or polypeptide is provided for use in therapy, or in a method of medical treatment.
  • an antibody, antigen binding fragment or polypeptide as described herein is provided for use in the treatment of a T-cell dysfunctional disorder.
  • the use of an antibody, antigen binding fragment or polypeptide as described herein in the manufacture of a medicament or pharmaceutical composition for use in the treatment of a T-cell dysfunctional disorder is provided.
  • a method of enhancing T-cell function comprising administering an antibody, antigen binding fragment or polypeptide as described herein to a dysfunctional T-cell.
  • the method may be performed in vitro or in vivo.
  • a method of treating a T-cell dysfunctional disorder comprising administering an antibody, antigen binding fragment or polypeptide as described herein to a patient suffering from a T-cell dysfunctional disorder.
  • a method of modulating an immune response in a subject comprising administering to the subject an antibody, antigen binding fragment or polypeptide as described herein such that the immune response in the subject is modulated.
  • a method of inhibiting growth of tumor cells in a subject comprising administering to the subject a therapeutically effective amount of an antibody, antigen binding fragment or polypeptide as described herein.
  • a method comprising contacting a sample containing, or suspected to contain, PD-1 with an antibody or antigen binding fragment, as described herein, and detecting the formation of a complex of antibody, or antigen binding fragment, and PD-1.
  • a method of diagnosing a disease or condition in a subject comprising contacting, in vitro, a sample from the subject with an antibody, or antigen binding fragment, , as described herein, and detecting the formation of a complex of antibody, or antigen binding fragment, and PD-1.
  • a method of selecting or stratifying a subject for treatment with PD-1 targeted agents comprising contacting, in vitro, a sample from the subject with an antibody, or antigen binding fragment, according to the present invention and detecting the formation of a complex of antibody, or antigen binding fragment, and PD-1.
  • the use of an antibody, or antigen binding fragment, as described herein, for the detection of PD-1 in vitro is provided.
  • the use of an antibody, or antigen binding fragment, as described herein, as an in vitro diagnostic agent is provided.
  • a method for expanding a population of T cells wherein T cells are contacted in vitro or ex vivo with an antibody, antigen binding fragment or polypeptide according to the present invention.
  • a method of treatment of a subject having a T- cell dysfunctional disorder comprising culturing T cells obtained from a blood sample from a subject in the presence of an antibody, antigen binding fragment or polypeptide according to the present invention so as to expand the T cell population, collecting expanded T cells, and administering the expanded T cells to a subject in need of treatment.
  • the antibody, antigen binding fragment or polypeptide may be provided as a composition as described herein.
  • the antibody may be antibody clone A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4, or H9 as described herein.
  • Antibodies according to the present invention preferably bind to PD-1 (the antigen), preferably human or rhesus PD-1 , optionally with a Koin the range 0.1 to 2nM.
  • the antibody preferably specifically binds PD-1 (e.g. human or rhesus) over other members of the CD28 family (preferably from the same organism), such as one or more or each of TIM-3 (HAVCR2), LAG3 (CD223), ICOS (CD278), CTLA4 (CD152), BTLA (CD272) or CD28.
  • PD-1 e.g. human or rhesus
  • HAVCR2 TIM-3
  • LAG3 CD223)
  • ICOS CD278
  • CTLA4 CD152
  • BTLA CD272
  • CD28 CD28
  • Antibodies according to the present invention may be provided in isolated form. Antibodies according to the present invention may exhibit least one of the following properties:
  • a) binds to human PD-1 with a KD of 1 ⁇ or less, preferably one of ⁇ 10nM, ⁇ 1 nM, ⁇ 800pM, ⁇ 700pM, ⁇ 600pM, ⁇ 500pM, ⁇ 400pM, ⁇ 300pM, ⁇ 200pM or ⁇ 100pM; b) does not substantially bind to human TIM-3, LAG3, ICOS, CTLA4, BTLA or
  • MLR Mixed Lymphocyte Reaction
  • e increases interleukin-2 (IL-2) secretion in an MLR assay.
  • the antibodies may be capable of increasing interferon-gamma production in an MLR assay in a dose-dependent manner. In some embodiments, the antibodies may be capable of increasing interferon-gamma production in an MLR assay by lymphocytes expressing one or more markers of exhaustion, e.g. PD-1.
  • antibody we include a fragment or derivative thereof, or a synthetic antibody or synthetic antibody fragment.
  • the antigen-binding portion may be a part of an antibody (for example a Fab fragment) or a synthetic antibody fragment (for example a single chain Fv fragment [ScFv]).
  • Suitable monoclonal antibodies to selected antigens may be prepared by known techniques, for example those disclosed in “Monoclonal Antibodies: A manual of techniques ", H Zola (CRC Press, 1988) and in "Monoclonal
  • Hybridoma Antibodies Techniques and Applications ", J G R Hurrell (CRC Press, 1982). Chimaeric antibodies are discussed by Neuberger et al (1988, 8th International
  • Monoclonal antibodies are useful in the methods of the invention and are a homogenous population of antibodies specifically targeting a single epitope on an antigen.
  • Polyclonal antibodies are useful in the methods of the invention. Monospecific polyclonal antibodies are preferred. Suitable polyclonal antibodies can be prepared using methods well known in the art. Antigen binding fragments of antibodies, such as Fab and Fab2 fragments may also be used/provided as can genetically engineered antibodies and antibody fragments.
  • the variable heavy (V H ) and variable light (V L ) domains of the antibody are involved in antigen recognition, a fact first recognised by early protease digestion experiments. Further confirmation was found by "humanisation" of rodent antibodies. Variable domains of rodent origin may be fused to constant domains of human origin such that the resultant antibody retains the antigenic specificity of the rodent parented antibody (Morrison et al (1984) Proc. Natl. Acad. Sd. USA 81 , 6851-6855).
  • variable domains that antigenic specificity is conferred by variable domains and is independent of the constant domains is known from experiments involving the bacterial expression of antibody fragments, all containing one or more variable domains.
  • variable domains include Fab-like molecules (Better et al (1988) Science 240, 1041); Fv molecules (Skerra et al (1988) Science 240, 1038); single-chain Fv (ScFv) molecules where the VH and VL partner domains are linked via a flexible oligopeptide (Bird et al (1988) Science 242, 423; Huston et al (1988) Proc. Natl. Acad. Sd.
  • ScFv molecules we mean molecules wherein the VH and VL partner domains are covalently linked, e.g. by a flexible oligopeptide.
  • Fab, Fv, ScFv and dAb antibody fragments can all be expressed in and secreted from E. coli, thus allowing the facile production of large amounts of the said fragments.
  • the present application also provides an antibody or antigen binding fragment which is capable of binding to PD-1 , and which is a bispecific antibody or a bispecific antigen binding fragment.
  • the bispecific antibody or bispecific antigen binding fragment may be isolated.
  • the bispecific antibodies and bispecific antigen binding fragments comprise an antigen binding fragment or a polypeptide according to the present invention. In some embodiments, the bispecific antibodies and bispecific antigen binding fragments comprise an antigen binding domain capable of binding to PD-1 , wherein the antigen binding domain which is capable of binding to PD-1 comprises or consists of an antigen binding fragment or a polypeptide according to the present invention.
  • the bispecific antibodies and bispecific antigen binding fragments comprise an antigen binding domain capable of binding to PD-1 , and an antigen binding domain capable of binding to another target protein.
  • the antigen binding domain capable of binding to another target protein may be capable of binding to another protein other than PD-1.
  • the target protein is a cell surface receptor.
  • the target protein is a cell surface receptor expressed on the cell surface of immune cells, e.g. T cells.
  • the target protein may be a member of the CD28 family.
  • the target protein may be a member of the CD28 family such as TIM-3 (HAVCR2), LAG 3 (CD223), ICOS (CD278), CTLA4 (CD152), BTLA (CD272) or CD28.
  • the target protein may be CTLA4 or LAG3.
  • the antigen binding domain for TIM-3 may comprise the CDRs, light and heavy chain variable domains or other TIM-3 binding fragment of e.g. anti-TIM-3 antibody clone F38-2E2 (BioLegend), clone 2E2 (Merck Millipore), clone 6B6E2, clone 024 (Sino Biological) clone 344801 (R&D Systems), clone E-18, clone H-191 (Santa Cruz Biotechnology), or clone 13A224 (United States Biological).
  • the antigen binding domain for LAG3 may comprise the CDRs, light and heavy chain variable domains or other LAG3 binding fragment of e.g. anti-LAG3 antibody clone 17B4 (Enzo
  • the antigen binding domain for ICOS may comprise the CDRs, light and heavy chain variable domains or other ICOS binding fragment of e.g. anti-ICOS antibody clone ISA-3 (eBioscience), clone SP98 (Novus Biologicals), clone 1 G1 , clone 3G4 (Abnova Corporation), clone 669222 (R&D Systems), clone TQ09 (Creative
  • the antigen binding domain for CTLA4 may comprise the CDRs, light and heavy chain variable domains or other CTLA4 binding fragment of e.g. anti-CTLA4 antibody clone 2F1 , clone 1 F4 (Abnova Corporation), clone 9H10 (EMD Millipore), clone BNU3 (GeneTex), clone 1 E2, clone AS32 (Lifespan Biosciences) clone A3.4H2.H12 (Acris Antibodies), clone 060 (Sino Biological), clone BU5G3 (Creative Diagnostics), clone MIH8 (MBL International), clone A3.6B10.G1 , or clone L3D10 (BioLegend).
  • anti-CTLA4 antibody clone 2F1 e.g. anti-CTLA4 antibody clone 2F1 , clone 1 F4 (Abnova Corporation), clone 9H10 (EMD Mill
  • the antigen binding domain for BTLA may comprise the CDRs, light and heavy chain variable domains or other BTLA binding fragment of e.g. anti-BTLA antibody clone 1 B7, clone 2G8, clone 4C5 (Abnova Corporation), clone 4B8 (antibodies-online), clone MIH26 (Thermo Scientific Pierce Antibodies), clone UMAB61 (OriGene Technologies), clone 330104 (R&D
  • the antigen binding domain for CD28 may comprise the CDRs, light and heavy chain variable domains or other CD28 binding fragment of e.g. anti-CD28 antibody clone CD28.6 (eBioscience), clone CD28.2, clone JJ319 (Novus Biologicals), clone 204.12, clone B-23, clone 10F3 (Thermo Scientific Pierce Antibodies), clone 37407 (R&D Systems), clone 204-12 (Abnova Corporation), clone 15E8 (EMD Millipore), clone 204-12, clone YTH913.12 (AbD Serotec), clone B-T3 (Acris Antibodies), clone 9H6E2 (Sino Biological), clone C28/77 (MyBioSource.com), clone KOLT-2
  • anti-CD28 antibody clone CD28.6 eBioscience
  • An antigen binding domain of a bispecific antibody or bispecific antigen binding fragment according to the present invention may be any domain of a polypeptide which is capable of binding to an antigen.
  • an antigen binding domain comprises at least the three light chain CDRs (i.e. LC-CDR1 , LC-CDR2 and LC-CDR3) and three heavy chain CDRs (i.e. HC-CDR1 , HC-CDR2 and HC-CDR3) which together define the antigen binding region of an antibody or antigen binding fragment.
  • an antigen binding domain may comprise the light chain variable domain and heavy chain variable domain of an antibody or antigen binding fragment.
  • an antigen binding domain may comprise the light chain polypeptide and heavy chain polypeptide of an antibody or antigen binding fragment.
  • Bispecific antibodies and bispecific antigen binding fragments according to the invention may be provided in any suitable format, such as those formats described in Kontermann MAbs 2012, 4(2): 182-197, which is hereby incorporated by reference in its entirety.
  • a bispecific antibody or bispecific antigen binding fragment may be a bispecific antibody conjugate (e.g. an lgG2, F(ab')2 or CovX-Body), a bispecific IgG or IgG-like molecule (e.g.
  • bispecific antibodies and bispecific antigen binding fragments are designed and prepare bispecific antibodies and bispecific antigen binding fragments according to the present invention.
  • Methods for producing bispecific antibodies include chemically crosslinking of antibodies or antibody fragments, e.g. with reducible disulphide or non-reducible thioether bonds, for example as described in Segal and Bast, 2001. Production of Bispecific Antibodies.
  • A/-succinimidyl-3-(-2-pyridyldithio)- propionate can be used to chemically crosslink e.g. Fab fragments via hinge region SH- groups, to create disulfide-linked bispecific F(ab) 2 heterodimers.
  • bispecific antibodies include fusing antibody-producing hybridomas e.g. with polyethylene glycol, to produce a quadroma cell capable of secreting bispecific antibody, for example as described in D. M. and Bast, B. J. 2001.
  • Bispecific antibodies and bispecific antigen binding fragments according to the present invention can also be produced recombinantly, by expression from e.g. a nucleic acid construct encoding polypeptides for the antigen binding molecules, for example as described in Antibody Engineering: Methods and Protocols, Second Edition (Humana Press, 2012), at Chapter 40: Production of Bispecific Antibodies: Diabodies and Tandem scFv (Hornig and Farber-Schwarz), or French, How to make bispecific antibodies, Methods Mol. Med. 2000; 40:333-339, the entire contents of both of which are hereby incorporated by reference.
  • a DNA construct encoding the light and heavy chain variable domains for the two antigen binding domains i.e.
  • the light and heavy chain variable domains for the antigen binding domain capable of binding PD-1 , and the light and heavy chain variable domains for the antigen binding domain capable of binding to another target protein), and including sequences encoding a suitable linker or dimerization domain between the antigen binding domains can be prepared by molecular cloning techniques.
  • Recombinant bispecific antibody can thereafter be produced by expression (e.g. in vitro) of the construct in a suitable host cell (e.g. a mammalian host cell), and expressed recombinant bispecific antibody can then optionally be purified.
  • Antibodies may be produced by a process of affinity maturation in which a modified antibody is generated that has an improvement in the affinity of the antibody for antigen, compared to an unmodified parent antibody.
  • Affinity-matured antibodies may be produced by procedures known in the art, e.g., Marks et ai, Rio/Technology 10:779-783 (1992);
  • Antibodies according to the present invention preferably exhibit specific binding to PD-1.
  • An antibody that specifically binds to a target molecule preferably binds the target with greater affinity, and/or with greater duration than it binds to other targets.
  • the extent of binding of an antibody to an unrelated target is less than about 10% of the binding of the antibody to the target as measured, e.g., by ELISA, or by a radioimmunoassay (RIA).
  • the binding specificity may be reflected in terms of binding affinity where the anti-PD-1 antibody of the present invention binds to PD-1 with a KD that is at least 0.1 order of magnitude (i.e. 0.1 x 10", where n is an integer
  • the KD of the antibody towards another target molecule e.g. another member of the CD28 family.
  • This may optionally be one of at least 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.5, or 2.0.
  • Antibodies according to the present invention preferably have a dissociation constant (KD) of one of ⁇ 1 ⁇ , ⁇ 100nM, ⁇ 10nM, ⁇ 1 nM or ⁇ 100pM. Binding affinity of an antibody for its target is often described in terms of its dissociation constant (KD). Binding affinity can be measured by methods known in the art, such as by Surface Plasmon Resonance, or by a radiolabeled antigen binding assay (RIA) performed with the Fab version of the antibody and antigen molecule.
  • KD dissociation constant
  • Antibodies according to the present invention may be "antagonist" antibodies that inhibit or reduce a biological activity of the antigen to which it binds. Blocking of PD-1 assists in the restoration of T-cell function by inhibiting the immune-inhibitory signalling pathway mediated by PD-1.
  • the antibody is clone A3, or a variant of A3.
  • A3 comprises the following CDR sequences:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 ASWDDVLYGSV (SEQ ID NO:27)
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGKDH (SEQ ID NO:41 )
  • the antibody is clone A10, or a variant of A10.
  • A10 comprises the following CDR sequences:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 ASWDDYYYGTI (SEQ ID NO:28)
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGKDV (SEQ ID NO:42)
  • the antibody is clone B6, or a variant of B6.
  • B6 comprises the following CDR sequences:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 ASWDDYLRGTV (SEQ ID NO:29)
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DYGAGPYYYGMDV (SEQ ID NO:43)
  • the antibody is clone C4, or a variant of C4.
  • C4 comprises the following CDR sequences:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 SAWDDYLHGTV (SEQ ID NO:30)
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGLDV (SEQ ID NO:44)
  • the antibody is clone D4, or a variant of D4.
  • D4 comprises the following CDR sequences:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 ASWDDYVRGTM (SEQ ID NO:31 )
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGMDV (SEQ ID NO:45)
  • the antibody is clone E1 , or a variant of E1.
  • E1 comprises the following CDR sequences:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGMDV (SEQ ID NO:46)
  • the antibody is clone F2, or a variant of F2.
  • F2 comprises the following CDR sequences:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 SSWDDDARGTI (SEQ ID NO:33)
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGMDV (SEQ ID NO:47)
  • the antibody is clone G1 , or a variant of G1.
  • G1 comprises the following CDR sequences: Light chain:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 AAWDDVYYGTI (SEQ ID NO:34)
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGMDV (SEQ ID NO:48)
  • the antibody is clone G2, or a variant of G2.
  • G2 comprises the following CDR sequences:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 ASWDDSLYGTV (SEQ ID NO:35)
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGMDV (SEQ ID NO:49)
  • the antibody is clone G10, or a variant of G10.
  • G10 comprises the following CDR sequences:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 AAWDDAYYGTI (SEQ ID NO:36)
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR3 DYGAGPYYYGMDV (SEQ ID NO:50)
  • the antibody is clone H4, or a variant of H4.
  • H4 comprises the following CDR sequences:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 ASWDDVYRGTV (SEQ ID NO:37)
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGSGYYLYGMDV (SEQ ID NO:51 )
  • the antibody is clone H9, or a variant of H9.
  • H9 comprises the following CDR sequences:
  • LC-CDR1 SGSSSNIKFNSVN (SEQ ID NO:25)
  • LC-CDR2 SNNQRPS (SEQ ID NO:26)
  • LC-CDR3 SSWDDSLYGTI (SEQ ID NO:38)
  • HC-CDR1 GFTFSSYGMH (SEQ ID NO:39) or
  • HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40)
  • HC-CDR3 DLGAGPYYYGMDV (SEQ ID NO:52)
  • Antibodies according to the present invention may comprise the CDRs of one of A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4, or H9 or one of SEQ ID NOs 1 and 13, 2 and 14, 3 and 15, 4 and 16, 5 and 17, 6 and 18, 7 and 19, 8 and 20, 9 and 21 , 10 and 22, 11 and 23, or 12 and 24.
  • one or two or three or four of the six CDR sequences may vary.
  • a variant may have one or two amino acid substitutions in one or two of the six CDR sequences.
  • Amino acid sequences of the V H and V L chains of ant-PD-1 clones are shown in Figures 1 and 2.
  • the encoding nucleotide sequences are shown in Figure 4.
  • the light and heavy chain CDRs may also be particularly useful in conjunction with a number of different framework regions. Accordingly, light and/or heavy chains having LC- CDR1-3 or HC-CDR1-3 may possess an alternative framework region. Suitable framework regions are well known in the art and are described for example in M. Lefranc & G. Lefranc (2001) "The Immunoglobulin FactsBook", Academic Press, incorporated herein by reference.
  • antibodies may have VH and/or VL chains comprising an amino acid sequence that has a high percentage sequence identity to one or more of the VH and/or V L amino acid sequences of SEQ ID Nos 1 and 13, 2 and 14, 3 and 15, 4 and 16, 5 and 17, 6 and 18, 7 and 19, 8 and 20, 9 and 21 , 10 and 22, 1 1 and 23, or 12 and 24 respectively, or to one or the amino acid sequences shown in Figures 1 and 2.
  • antibodies according to the present invention include antibodies that bind PD-1 and have a VH or VL chain that comprises an amino acid sequence having at least 70%, more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the V H or V L chain amino acid sequence of one of SEQ ID NOs 1 to 24 or to one or the amino acid sequences shown in Figures 1 and 2.
  • Antibodies according to the present invention may be detectably labelled or, at least, capable of detection.
  • the antibody may be labelled with a radioactive atom or a coloured molecule or a fluorescent molecule or a molecule which can be readily detected in any other way. Suitable detectable molecules include fluorescent proteins, luciferase, enzyme substrates, and radiolabels.
  • the binding moiety may be directly labelled with a detectable label or it may be indirectly labelled.
  • the binding moiety may be an unlabelled antibody which can be detected by another antibody which is itself labelled.
  • the second antibody may have bound to it biotin and binding of labelled streptavidin to the biotin is used to indirectly label the first antibody.
  • Antibodies, or antigen binding fragments, described herein may be used in methods that involve the binding of the antibody or antigen binding fragment to PD-1. Such methods may involve detection of the bound complex of antibody, or antigen binding fragment, and PD-1. As such, in one embodiment a method is provided, the method comprising contacting a sample containing, or suspected to contain, PD-1 with an antibody or antigen binding fragment as described herein and detecting the formation of a complex of antibody, or antigen binding fragment, and PD-1. Suitable method formats are well known in the art, including immunoassays such as sandwich assays, e.g. ELISA.
  • the method may involve labelling the antibody, or antigen binding fragment, or PD-1 , or both, with a detectable label, e.g. fluorescent, luminescent or radio- label.
  • a detectable label e.g. fluorescent, luminescent or radio- label.
  • Such methods may involve determining the amount of PD-1 present in a patient sample.
  • the method may further comprise comparing the determined amount against a standard or reference value as part of the process of reaching a diagnosis.
  • Other diagnostic tests may be used in conjunction with those described here to enhance the accuracy of the diagnosis or prognosis or to confirm a result obtained by using the tests described here.
  • the level of PD-1 present in a patient sample may be indicative that a patient may respond to treatment with an anti-PD1 antibody.
  • the presence of a high level of PD-1 in a sample may be used to select a patient for treatment with an anti-PD1 antibody.
  • the antibodies of the present invention may therefore be used to select a patient for treatment with anti-PD-1 therapy.
  • Detection in a sample of PD-1 may be used for the purpose of diagnosis of a T-cell dysfunctional disorder or a cancerous condition in the patient, diagnosis of a
  • the diagnosis or prognosis may relate to an existing (previously diagnosed) cancerous condition, which may be benign or malignant, may relate to a suspected cancerous condition or may relate to the screening for cancerous conditions in the patient (which may be previously undiagnosed).
  • the level of PD-1 expression on CD8+ T cells may be detected in order to indicate the degree of T-cell exhaustion and severity of the disease state.
  • a sample may be taken from any tissue or bodily fluid.
  • the sample may comprise or may be derived from: a quantity of blood; a quantity of serum derived from the individual's blood which may comprise the fluid portion of the blood obtained after removal of the fibrin clot and blood cells; a tissue sample or biopsy; or cells isolated from said individual.
  • Methods according to the present invention are preferably performed in vitro.
  • the term "in vitro” is intended to encompass experiments with cells in culture whereas the term “in vivo” is intended to encompass experiments with intact multi-cellular organisms.
  • Antibodies, antigen binding fragments and polypeptides according to the present invention and compositions comprising such agents may be provided for use in methods of medical treatment.
  • Treatment may be provided to subjects having a disease or condition in need of treatment.
  • the disease or condition may be one of a T-cell dysfunctional disorder, including a T-cell dysfunctional disorder associated with a cancer, or a cancer, or a T-cell dysfunctional disorder associated with an infection, or an infection.
  • a T-cell dysfunctional disorder may be a disease or condition in which normal T-cell function is impaired causing downregulation of the subject's immune response to pathogenic antigens, e.g. generated by infection by exogenous agents such as microorganisms, bacteria and viruses, or generated by the host in some disease states such as in some forms of cancer (e.g. in the form of tumor associated antigens).
  • pathogenic antigens e.g. generated by infection by exogenous agents such as microorganisms, bacteria and viruses, or generated by the host in some disease states such as in some forms of cancer (e.g. in the form of tumor associated antigens).
  • the T-cell dysfunctional disorder may comprise T-cell exhaustion or T-cell anergy.
  • T-cell exhaustion comprises a state in which CD8 + T-cells fail to proliferate or exert T-cell effector functions such as cytotoxicity and cytokine (e.g. IFNv) secretion in response to antigen stimulation.
  • Exhausted T-cells may also be characterised by sustained expression of PD-1 , where blockade of PD-1 :PD-L1 interactions may reverse the T-cell exhaustion and restore antigen-specific T cell responses.
  • the T-cell dysfunctional disorder may be manifest as an infection, or inability to mount an effective immune response against an infection.
  • the infection may be chronic, persistent, latent or slow, and may be the result of bacterial, viral, fungal or parasitic infection.
  • treatment may be provided to patients having a bacterial, viral or fungal infection.
  • bacterial infections include infection with Helicobacter pylori.
  • viral infections include infection with HIV, hepatitis B or hepatitis C.
  • the T-cell dysfunctional disorder may be associated with a cancer, such as tumor immune escape.
  • a cancer such as tumor immune escape.
  • Many human tumors express tumor-associated antigens recognised by T cells and capable of inducing an immune response.
  • immune evasion is common and is believed to be mediated by a number of soluble factors, including PD-L1. As such, blocking the interaction of PD-1 and PD-L1 may inhibit this negative
  • Cancers may also be treated where there is no indication of a T-cell dysfunctional disorder such as T-cell exhaustion but the use of an antibody, antigen binding fragment or polypeptide according to the present invention allows the subject to suppress PD-1 signalling and mount an effective immune response with limited impairment, evasion or induction of tumor immune escape.
  • the antibody, antigen binding fragment or polypeptide may provide a treatment for cancer that involves prevention of the development of tumor immune escape.
  • the treatment may be aimed at prevention of the T-cell dysfunctional disorder, e.g.
  • the antibodies, antigen binding fragments and polypeptides may be used to formulate pharmaceutical compositions or medicaments and subjects may be prophylactically treated against development of a disease state. This may take place before the onset of symptoms of the disease state, and/or may be given to subjects considered to be at greater risk of infection or development of cancer.
  • Treatment may comprise co-therapy with a vaccine, e.g. T-cell vaccine, which may involve simultaneous, separate or sequential therapy, or combined administration of vaccine and antibody, antigen binding fragment or polypeptide in a single composition.
  • a vaccine e.g. T-cell vaccine
  • the antibody, antigen binding fragment or polypeptide may be provided as an adjuvant to the vaccine.
  • an antibody, antigen binding fragment or polypeptide is preferably in a "therapeutically effective amount", this being sufficient to show benefit to the individual.
  • the actual amount administered, and rate and time-course of administration, will depend on the nature and severity of the disease being treated. Prescription of treatment, e.g. decisions on dosage etc., is within the responsibility of general practitioners and other medical doctors, and typically takes account of the disorder to be treated, the condition of the individual patient, the site of delivery, the method of administration and other factors known to practitioners. Examples of the techniques and protocols mentioned above can be found in Remington's Pharmaceutical Sciences, 20th Edition, 2000, pub. Lippincott, Williams & Wilkins.
  • Antibodies, antigen binding fragments and polypeptides according to the present invention may be formulated as pharmaceutical compositions for clinical use and may comprise a pharmaceutically acceptable carrier, diluent, excipient or adjuvant.
  • methods are also provided for the production of pharmaceutically useful compositions, such methods of production may comprise one or more steps selected from: isolating an antibody, antigen binding fragment or polypeptide as described herein; and/or mixing an isolated antibody, antigen binding fragment or polypeptide as described herein with a pharmaceutically acceptable carrier, adjuvant, excipient or diluent.
  • a further aspect of the present invention relates to a method of formulating or producing a medicament or pharmaceutical composition for use in the treatment of a T- cell dysfunctional disorder, the method comprising formulating a pharmaceutical composition or medicament by mixing an antibody, antigen binding fragment or polypeptide as described herein with a pharmaceutically acceptable carrier, adjuvant, excipient or diluent.
  • An infection may be any infection or infectious disease, e.g. bacterial, viral, fungal, or parasitic infection. In some embodiments it may be particularly desirable to treat chronic/persistent infections, e.g. where such infections are associated with T cell dysfunction or T cell exhaustion.
  • T cell exhaustion is a state of T cell dysfunction that arises during many chronic infections (including viral, bacterial and parasitic), as well as in cancer (Wherry Nature Immunology ⁇ Vol.12, No.6, p492-499, June 201 1).
  • An infection or infectious disease may be one in which PD-1 is upregulated (e.g. as reported by Radziewicz H, et al., J Virol. 2007;81 (6):2545-2553 and Golden-Mason L et al., J Virol. 2007;81 (17):9249-9258).
  • bacterial infections examples include infection by Bacillus spp., Bordetella pertussis, Clostridium spp., Corynebacterium spp., Vibrio chloerae,
  • Staphylococcus spp. Streptococcus spp. Escherichia, Klebsiella, Proteus, Yersinia, Erwina, Salmonella, Listeria sp, Helicobacter pylori, mycobacteria (e.g. Mycobacterium tuberculosis) and Pseudomonas aeruginosa.
  • the bacterial infection may be sepsis or tuberculosis.
  • viral infections examples include infection by influenza virus, measles virus, hepatitis B virus (HBV), hepatitis C virus (HCV), human immunodeficiency virus (HIV), lymphocytic choriomeningitis virus (LCMV), Herpes simplex virus and human papilloma virus.
  • influenza virus measles virus
  • HBV hepatitis B virus
  • HCV hepatitis C virus
  • HMV human immunodeficiency virus
  • LCMV lymphocytic choriomeningitis virus
  • Herpes simplex virus Herpes simplex virus and human papilloma virus.
  • HCV hepatitis C virus
  • McMahan et al. The Journal of Clinical Investigation Vol. 120, No. 12 p4546-4557, December 2010.
  • McMahan et al found that the level of dual TIM-3 and PD-1 expression on HCV-specific CTLs predated the development of viral persistence, providing prognostic information.
  • Barber et al. (Nature Vol 439, No. 9 p682-687 Feb 2006) reported that PD-1 is upregulated during chronic viral infection.
  • mice infected with LCMV they reported that blockade of the PD-1/PD-L1 inhibitory pathway had a beneficial effect on CD8 T cells, restoring their ability to undergo proliferation, secrete cytokines, kill infected cells and decrease viral load.
  • PD-1 is also upregulated in HIV infection (Said et al., Nature Medicine Vol. 16, No.4 p452-460 April 2010). Blocking interaction between PD-1 and PD-L1 contributed to viral clearance and improved T cell function in animal models of chronic viral infection (Said et al., supra).
  • fungal infections examples include infection by Alternaria sp, Aspergillus sp, Candida sp and Histoplasma sp.
  • the fungal infection may be fungal sepsis or histoplasmosis.
  • Chang et al (Blockade of the negative co-stimulatory molecules PD-1 and CTLA-4 improves survival in primary and secondary fungal sepsis.
  • Critical Care 2013, 17:R85) reported that anti-PD1 antibodies were highly effective at improving survival in primary and secondary fungal sepsis.
  • Lazar-Molnar et al (The PD-1/PD-L costimulatory pathway critically affects host resistance to the pathogenic fungus Histoplasma capsulatum PNAS vol. 105, no.7, p2658-2663, 19 Feb 2008) reported that anti-PD-1 antibody significantly increased survival of mice infected with Histoplasma capsulatum. As such, the importance of T cell exhaustion in mediating fungal infection is well established.
  • parasitic infections examples include infection by Plasmodium species (e.g. Plasmodium falciparum, Plasmodium yoeli, Plasmodium ovale, Plasmodium vivax, or Plasmodium chabaudi chabaudi).
  • the parasitic infection may be a disease such as malaria, leishmaniasis and toxoplasmosis. Infection of humans with Plasmodium falciparum has been shown to result in higher expression of PD-1 and T cell exhaustion mice (Butler et al., Nature Immunology ' Vol.13, No.12, p 188-195 February 2012).
  • Blockade of PD-L1 and LAG-3 using anti-PD-L1 and anti-LAG-3 monoclonal antibodies in vivo contributed to the restoration of CD4 + T-cell function, amplification of the number of follicular helper T cells, germinal-center B cells and plasmablasts, enhanced protective antibodies and rapidly cleared blood-stage malaria in mice. It was also shown to block the development of chronic infection (Butler et al., supra). Cancer
  • a cancer may be any unwanted cell proliferation (or any disease manifesting itself by unwanted cell proliferation), neoplasm or tumor or increased risk of or predisposition to the unwanted cell proliferation, neoplasm or tumor.
  • the cancer may be benign or malignant and may be primary or secondary (metastatic).
  • a neoplasm or tumor may be any abnormal growth or proliferation of cells and may be located in any tissue. Examples of tissues include the adrenal gland, adrenal medulla, anus, appendix, bladder, blood, bone, bone marrow, brain, breast, cecum, central nervous system (including or excluding the brain) cerebellum, cervix, colon, duodenum, endometrium, epithelial cells (e.g.
  • kidney oesophagus
  • glial cells heart, ileum, jejunum, kidney, lacrimal glad, larynx, liver, lung, lymph, lymph node, lymphoblast, maxilla, mediastinum, mesentery, myometrium, nasopharynx, omentume, oral cavity, ovary, pancreas, parotid gland, peripheral nervous system, peritoneum, pleura, prostate, salivary gland, sigmoid colon, skin, small intestine, soft tissues, spleen, stomach, testis, thymus, thyroid gland, tongue, tonsil, trachea, uterus, vulva, white blood cells.
  • Tumors to be treated may be nervous or non-nervous system tumors.
  • Nervous system tumors may originate either in the central or peripheral nervous system, e.g. glioma, medulloblastoma, meningioma, neurofibroma, ependymoma, Schwannoma,
  • Non-nervous system Neurofibrosarcoma, astrocytoma and oligodendroglioma.
  • cancers/tumors may originate in any other non-nervous tissue, examples include melanoma, mesothelioma, lymphoma, myeloma, leukemia, Non-Hodgkin's lymphoma (NHL), Hodgkin's lymphoma, chronic myelogenous leukemia (CML), acute myeloid leukemia (AML), myelodysplasia syndrome (MDS), cutaneous T-cell lymphoma (CTCL), chronic lymphocytic leukemia (CLL), hepatoma, epidermoid carcinoma, prostate carcinoma, breast cancer, lung cancer , colon cancer, ovarian cancer, pancreatic cancer, thymic carcinoma, NSCLC, haematologic cancer and sarcoma.
  • the cancer is one or more of lung cancer, renal cancer and bladder cancer.
  • Adoptive T cell transfer therapy is one or more of lung cancer, renal cancer and bladder cancer.
  • Adoptive T cell transfer therapy generally refers to a process in which white blood cells are removed from a subject, typically by drawing a blood sample from which white blood cells are separated, expanded in vitro or ex vivo and returned either to the same subject or to a different subject.
  • the treatment is typically aimed at increasing the
  • Such treatment may be beneficial in subjects experiencing T cell exhaustion.
  • Antibodies capable of blocking the mechanism of T cell exhaustion, or reversing it, provide a means of enhancing T cell activity and promoting T cell expansion.
  • a method for expanding a population of T cells wherein T cells are contacted in vitro or ex vivo with an antibody, antigen binding fragment or polypeptide according to the present invention.
  • the method may optionally comprise one or more of the following steps: taking a blood sample from a subject; isolating T cells from the blood sample; culturing the T cells in in vitro or ex vivo cell culture (where they may be contacted with the antibody, antigen binding fragment or polypeptide), collecting an expanded population of T cells; mixing the T cells with an adjuvant, diluent, or carrier; administering the expanded T cells to a subject.
  • a method of treatment of a subject having a T-cell dysfunctional disorder comprising obtaining a blood sample from a subject in need of treatment, culturing T cells obtained from the blood sample in the presence of an antibody, antigen binding fragment or polypeptide according to the present invention so as to expand the T cell population, collecting expanded T cells, and administering the expanded T cells to a subject in need of treatment.
  • the T cells may be obtained from a subject requiring treatment, and may be isolated and/or purified. They may be a CD4 + and/or CD8 + T-cell population.
  • the T-cells may represent a population experiencing T cell exhaustion and may optionally have upregulated expression of PD-1.
  • T cells may be contacted with the antibody, antigen binding fragment or polypeptide under conditions and for a period of time suitable to allow expansion of the T cells to a desired number of cells. After a suitable period of time the T cells may be harvested, optionally concentrated, and may be mixed with a suitable carrier, adjuvant or diluent and returned to the subject's body. A subject may undergo one or more rounds of such therapy.
  • T cell expansion Methods of T cell expansion are well known in the art, such as those described in Kalamasz et al., J Immunother 2004 Sep-Oct; 27(5):405-18; Monies et al., Clin Exp Immunol 2005 Nov;142(2):292-302; Wolfl and Greenburg Nature Protocols 9 p950-966 27 March 2014; Trickett and Kwan Journal of Immunological Methods Vol. 275, Issues 1- 2, 1 April 2003, p251-255; Butler et al PLoSONE l ⁇ ) 12 Jan 2012.
  • compositions may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated.
  • an antibody, antigen binding fragment or polypeptide of the present invention and an anti-infective agent or chemotherapeutic agent may be administered simultaneously or sequentially.
  • treatment with an antibody, antigen binding fragment or polypeptide of the present invention may be accompanied by chemotherapy.
  • Simultaneous administration refers to administration of the antibody, antigen binding fragment or polypeptide and therapeutic agent together, for example as a pharmaceutical composition containing both agents (combined preparation), or immediately after each other and optionally via the same route of administration, e.g. to the same artery, vein or other blood vessel.
  • Sequential administration refers to administration of one of the antibody, antigen binding fragment or polypeptide or therapeutic agent followed after a given time interval by separate administration of the other agent. It is not required that the two agents are administered by the same route, although this is the case in some embodiments.
  • the time interval may be any time interval.
  • an antibody, antigen binding fragment or polypeptide of the present invention may be administered in combination with an anti-infective agent, as described above.
  • the anti-infective agent may be an agent known to have action against the microorganism or virus responsible for the infection.
  • Suitable anti-infective agents include antibiotics (such as penicillins, cephalosporins, rifamycins, lipiarmycins, quinolones, sulfonamides, macrolides, lincosamides, tetracyclines, cyclic lipopeptides, glycylcyclines, oxazolidinones, and lipiarmycins), antiviral agents (such as reverse transcriptase inhibitors, integrase inhibitors, transcription factor inhibitors, antisense and siRNA agents and protease inhibitors), anti-fungal agents (such as polyenes, imidiazoles, triazoles, thiazoles, allylamines, and echinocandins) and anti-parasitic agents (such as antinematode agents, anticestode agents, antitrematode agents, antiamoebic agents and antiprotozoal agents).
  • antibiotics such as penicillins, cephalosporins,
  • Chemotherapy refers to treatment of a cancer with a drug or with ionising radiation (e.g. radiotherapy using X-rays or ⁇ -rays).
  • chemotherapy refers to treatment with a drug.
  • the drug may be a chemical entity, e.g. small molecule pharmaceutical, antibiotic, DNA intercalator, protein inhibitor (e.g. kinase inhibitor), or a biological agent, e.g. antibody, antibody fragment, nucleic acid or peptide aptamer, nucleic acid (e.g. DNA, RNA), peptide, polypeptide, or protein.
  • the drug may be formulated as a pharmaceutical composition or medicament.
  • the formulation may comprise one or more drugs (e.g.
  • a treatment may involve administration of more than one drug.
  • a drug may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated.
  • the chemotherapy may be a co-therapy involving administration of two drugs, one or more of which may be intended to treat the cancer.
  • the chemotherapy may be administered by one or more routes of administration, e.g. parenteral, intravenous injection, oral, subcutaneous, intradermal or intratumoral.
  • the chemotherapy may be administered according to a treatment regime.
  • the treatment regime may be a pre-determined timetable, plan, scheme or schedule of chemotherapy administration which may be prepared by a physician or medical practitioner and may be tailored to suit the patient requiring treatment.
  • the treatment regime may indicate one or more of: the type of chemotherapy to administer to the patient; the dose of each drug or radiation; the time interval between administrations; the length of each treatment; the number and nature of any treatment holidays, if any etc.
  • a single treatment regime may be provided which indicates how each drug is to be administered.
  • Chemotherapeutic drugs and biologies may be selected from:
  • alkylating agents such as cisplatin, carboplatin, mechlorethamine,
  • alkaloids and terpenoids such as vinca alkaloids (e.g. vincristine, vinblastine, vinorelbine, vindesine), podophyllotoxin, etoposide, teniposide, taxanes such as paclitaxel (TaxolTM), docetaxel;
  • vinca alkaloids e.g. vincristine, vinblastine, vinorelbine, vindesine
  • podophyllotoxin etoposide
  • teniposide teniposide
  • taxanes such as paclitaxel (TaxolTM), docetaxel
  • topoisomerase inhibitors such as the type I topoisomerase inhibitors
  • camptothecins irinotecan and topotecan or the type II topoisomerase inhibitors amsacrine, etoposide, etoposide phosphate, teniposide;
  • ⁇ antitumor antibiotics e.g. anthracyline antibiotics
  • dactinomycin e.g. dactinomycin
  • doxorubicin (AdriamycinTM), epirubicin, bleomycin, rapamycin;
  • antibody based agents such as anti-TIM-3 antibodies, anti-CTLA-4, anti-LAG-3, anti-4-I BB, anti-GITR, anti-CD27, anti-BLTA, anti-OX40, anti-VEGF, anti-TNFa, anti-IL-2, antiGpllb/llla, anti-CD-52, anti-CD20, anti-RSV, anti-HER2/neu(erbB2), anti-TNF receptor, anti-EGFR antibodies, monoclonal antibodies or antibody fragments, examples include: cetuximab, panitumumab, infliximab, basiliximab, bevacizumab (Avastin®), abciximab, daclizumab, gemtuzumab, alemtuzumab, rituximab (Mabthera®), palivizumab, trastuzumab, etanercept, adalimumab, nimotuzumab
  • EGFR inihibitors such as erlotinib, cetuximab and gefitinib • anti-angiogenic agents such as bevacizumab (Avastin®)
  • cancer vaccines such as Sipuleucel-T (Provenge®)
  • the chemotherapeutic agent is an anti-TIM-3 antibody, anti-CTLA-4, anti-LAG3, anti-41 BB, anti-GITR, anti-CD27, anti-BLTA, anti-OX40, anti-VEGF, anti- TNFa, anti-IL2, anti-Gpllb/llla, anti-CD-52, anti-CD20, anti-RSV, anti-HER2/neu(erbB2), anti-TNF receptor, anti-EGFR or other antibody.
  • the chemotherapeutic agent is an anti-TIM-3 antibody, anti-CTLA-4, anti-LAG3, anti-41 BB, anti-GITR, anti-CD27, anti-BLTA, anti-OX40, anti-VEGF, anti- TNFa, anti-IL2, anti-Gpllb/llla, anti-CD-52, anti-CD20, anti-RSV, anti-HER2/neu(erbB2), anti-TNF receptor, anti-EGFR or other antibody.
  • chemotherapeutic agent is an immune checkpoint inhibitor or costimulation molecule.
  • chemotherapeutic drugs may be selected from: 13-cis-Retinoic Acid, 2- Chlorodeoxyadenosine, 5-Azacitidine 5-Fluorouracil, 6-Mercaptopurine, 6-Thioguanine, Abraxane, Accutane®, Actinomycin-D Adriamycin®, Adrucil®, Afinitor®, Agrylin®, Ala- Cort®, Aldesleukin, Alemtuzumab, ALIMTA, Alitretinoin, Alkaban-AQ®, Alkeran®, All- transretinoic Acid, Alpha Interferon, Altretamine, Amethopterin, Amifostine,
  • Aminoglutethimide Anagrelide, Anandron®, Anastrozole, Arabinosylcytosine, Aranesp®, Aredia®, Arimidex®, Aromasin®, Arranon®, Arsenic Trioxide, Asparaginase, ATRA Avastin®, Azacitidine, BCG, BCNU, Bendamustine, Bevacizumab, Bexarotene,
  • BEXXAR® Bicalutamide, BiCNU, Blenoxane®, Bleomycin, Bortezomib, Busulfan, Busulfex®, Calcium Leucovorin, Campath®, Camptosar®, Camptothecin-11 ,
  • Ixabepilone IxempraTM, Kidrolase, Lanacort®, Lapatinib, L-asparaginase, LCR,
  • Lenalidomide Letrozole, Leucovorin, Leukeran, LeukineTM, Leuprolide, Leurocristine, LeustatinTM, Liposomal Ara-C, Liquid Pred®, Lomustine, L-PAM, L-Sarcolysin, Lupron®, Lupron Depot®, Matulane®, Maxidex, Mechlorethamine, Mechlorethamine Hydrochloride, Medralone®, Medrol®, Megace®, Megestrol, Megestrol Acetate, Melphalan,
  • Methylprednisolone Meticorten®, Mitomycin, Mitomycin-C, Mitoxantrone, M-Prednisol®, MTC, MTX, Mustargen®, Mustine, Mutamycin®, Myleran®, MylocelTM, Mylotarg®, Navelbine®, Nelarabine, Neosar®, NeulastaTM, Neumega®, Neupogen®, Nexavar®, Nilandron®, Nilutamide, Nipent®, Nitrogen Mustard, Novaldex®, Novantrone®,
  • Antibodies, antigen binding fragments, polypeptides and other therapeutic agents, medicaments and pharmaceutical compositions according to aspects of the present invention may be formulated for administration by a number of routes, including but not limited to, parenteral, intravenous, intra-arterial, intramuscular, subcutaneous, intradermal, intratumoral and oral.
  • Antibodies, antigen binding fragments, polypeptides and other therapeutic agents may be formulated in fluid or solid form. Fluid formulations may be formulated for administration by injection to a selected region of the human or animal body. Dosage regime
  • Multiple doses of the antibody, antigen binding fragment or polypeptide may be provided.
  • One or more, or each, of the doses may be accompanied by simultaneous or sequential administration of another therapeutic agent.
  • Multiple doses may be separated by a predetermined time interval, which may be selected to be one of 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, or 31 days, or 1 , 2, 3, 4, 5, or 6 months.
  • doses may be given once every 7, 14, 21 or 28 days (plus or minus 3, 2, or 1 days).
  • kits of parts may have at least one container having a predetermined quantity of the antibody, antigen binding fragment or polypeptide.
  • the kit may provide the antibody, antigen binding fragment or polypeptide in the form of a medicament or pharmaceutical composition, and may be provided together with instructions for administration to a patient in order to treat a specified disease or condition.
  • the antibody, antigen binding fragment or polypeptide may be formulated so as to be suitable for injection or infusion to a tumor or to the blood.
  • the kit may further comprise at least one container having a predetermined quantity of another therapeutic agent (e.g. anti-infective agent or chemotherapy agent).
  • the kit may also comprise a second medicament or pharmaceutical composition such that the two medicaments or pharmaceutical compositions may be administered simultaneously or separately such that they provide a combined treatment for the specific disease or condition.
  • the therapeutic agent may also be formulated so as to be suitable for injection or infusion to a tumor or to the blood.
  • the subject to be treated may be any animal or human.
  • the subject is preferably mammalian, more preferably human.
  • the subject may be a non-human mammal, but is more preferably human.
  • the subject may be male or female.
  • the subject may be a patient.
  • a subject may have been diagnosed with a disease or condition requiring treatment, or be suspected of having such a disease or condition.
  • the polypeptide may be expressed from a nucleotide sequence.
  • the nucleotide sequence may be contained in a vector present in a cell, or may be incorporated into the genome of the cell.
  • a "vector” as used herein is an oligonucleotide molecule (DNA or RNA) used as a vehicle to transfer exogenous genetic material into a cell.
  • the vector may be an expression vector for expression of the genetic material in the cell.
  • Such vectors may include a promoter sequence operably linked to the nucleotide sequence encoding the gene sequence to be expressed.
  • a vector may also include a termination codon and expression enhancers.
  • Suitable vectors include plasmids, binary vectors, viral vectors and artificial chromosomes (e.g. yeast artificial chromosomes).
  • operably linked may include the situation where a selected nucleotide sequence and regulatory nucleotide sequence (e.g. promoter and/or enhancer) are covalently linked in such a way as to place the expression of the nucleotide sequence under the influence or control of the regulatory sequence (thereby forming an expression cassette).
  • a regulatory sequence is operably linked to the selected nucleotide sequence if the regulatory sequence is capable of effecting transcription of the nucleotide sequence.
  • the resulting transcript may then be translated into a desired protein or polypeptide.
  • the cell may be a prokaryote or eukaryote.
  • Suitable prokaryotic cells include E.coli.
  • Examples of eukaryotic cells include a yeast cell, a plant cell, insect cell or a mammalian cell.
  • the cell is not a prokaryotic cell because some prokaryotic cells do not allow for the same post-translational modifications as eukaryotes.
  • very high expression levels are possible in eukaryotes and proteins can be easier to purify from eukaryotes using appropriate tags.
  • Specific plasmids may also be utilised which enhance secretion of the protein into the media.
  • Methods of producing a polypeptide of interest may involve culture or fermentation of a cell modified to express the polypeptide.
  • the culture or fermentation may be performed in a bioreactor provided with an appropriate supply of nutrients, air/oxygen and/or growth factors.
  • Secreted proteins can be collected by partitioning culture media/fermentation broth from the cells, extracting the protein content, and separating individual proteins to isolate secreted polypeptide. Culture, fermentation and separation techniques are well known to those of skill in the art.
  • Bioreactors include one or more vessels in which cells may be cultured. Culture in the bioreactor may occur continuously, with a continuous flow of reactants into, and a continuous flow of cultured cells from, the reactor. Alternatively, the culture may occur in batches.
  • the bioreactor monitors and controls environmental conditions such as pH, oxygen, flow rates into and out of, and agitation within the vessel such that optimum conditions are provided for the cells being cultured.
  • any suitable method for separating polypeptides/proteins from cell culture known in the art may be used. In order to isolate a polypeptide/protein of interest from a culture, it may be necessary to first separate the cultured cells from media containing the polypeptide/protein of interest. If the polypeptide/protein of interest is secreted from the cells, the cells may be separated from the culture media that contains the secreted polypeptide/protein by centrifugation. If the polypeptide/protein of interest collects within the cell, it will be necessary to disrupt the cells prior to centrifugation, for example using sonification, rapid freeze-thaw or osmotic lysis. Centrifugation will produce a pellet containing the cultured cells, or cell debris of the cultured cells, and a supernatant containing culture medium and the polypeptide/protein of interest.
  • polypeptide/protein of interest may be desirable to isolate the polypeptide/protein of interest from the supernatant or culture medium, which may contain other protein and non-protein components.
  • a common approach to separating polypeptide/protein components from a supernatant or culture medium is by precipitation.
  • Polypeptides/proteins of different solubility are precipitated at different concentrations of precipitating agent such as ammonium sulfate.
  • precipitating agent such as ammonium sulfate
  • water soluble proteins are extracted.
  • Dialysis may be subsequently used to remove ammonium sulfate from the separated proteins.
  • polypeptide/protein of interest Once the polypeptide/protein of interest has been isolated from culture it may be necessary to concentrate the protein.
  • a number of methods for concentrating a protein of interest are known in the art, such as ultrafiltration or lyophilisation.
  • Alignment for purposes of determining percent amino acid or nucleotide sequence identity can be achieved in various ways known to a person of skill in the art, for instance, using publicly available computer software such as ClustalW 1.82. T-coffee or Megalign
  • the invention includes the combination of the aspects and preferred features described except where such a combination is clearly impermissible or expressly avoided.
  • Figure 3 Table showing light chain and heavy chain CDR sequences for anti-PD-1 antibody clones A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4, H9.
  • Figure 4. Nucleotide and encoded amino acid sequences of heavy and light chain variable domain sequences for anti-PD-1 antibody clones A3, A10, optimised A10, B6, optimised B6, C4, optimised C4, D4, E1 , F2, G1 , G2, G10, H4, optimised H4 and H9 (human lgG4).
  • Figure 5 Table showing binding affinity (K D , nM) of clones A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4 and H9, and control antibodies nivolumab and lambrolizumab for human PD-1.
  • Figure 6. Chart showing binding of anti-human PD-1 antibodies A3, A10, nivolumab and lambrolizumab to activated T cells.
  • FIG. 7 Chart showing binding of anti-human PD-1 antibodies A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4 and H9, nivolumab and lambrolizumab to activated T cells.
  • Figure 8 Chart showing reactivity of clones B6, C4, D4, E1 , F2, G1 , G2, G10, H4 and H9 with human PD-1 , rhesus PD-1 and human CTLA-4.
  • Figure 9 Chart showing allogeneic CD4 + T cell proliferation of exhausted T cells in response to antibodies A3, A10, nivolumab, lambrolizumab.
  • Figure 10 Chart showing allogeneic CD4 + T cell proliferation of exhausted T cells in response to antibodies A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4, H9, nivolumab, lambrolizumab.
  • FIG. 11 Chart showing IFNv secretion of exhausted T cells in response to antibodies A3, A10, nivolumab, lambrolizumab.
  • FIG. 12 Chart showing IFNv secretion of exhausted T cells in response to antibodies A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4, H9, nivolumab, lambrolizumab.
  • Figure 13 Chart showing specificity of binding of antibodies A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4, H9, for human and rhesus-PD-1 over other human (h) or murine (m) CD28 family members compared with nivolumab, and lambrolizumab, as determined by ELISA.
  • Figure 14 Chart showing allogeneic CD4 + T cell proliferation of exhausted T cells in response to antibodies A10 codon optimised, B6 codon optimised, C4 codon optimised, H4 codon optimised, nivolumab, lambrolizumab.
  • Figure 15 Chart showing IFNv secretion of exhausted T cells in response to antibodies A10 codon optimised, B6 codon optimised, C4 codon optimised, H4 codon optimised, nivolumab, lambrolizumab.
  • Figure 16. Chart showing the expression of exhaustion markers PD-1 , PD-L1 , TIM-3 and LAG-3 by lung tumour infiltrating lymphocytes. Approximately 2/3 of CD4+ and CD8+ lymphocytes in the tumour express PD-1.
  • FIG. 17 Charts showing the secretion of IFN- ⁇ by tumour infiltrating lymphocytes (A) after 7 days of culture in the presence or absence of anti-PD-1 antibodies (direct culture of tumour dissociated tissues), (B) after a mixed lymphocyte reaction in the presence or absence of anti-PD-1 antibodies.
  • Figure 18 Chart showing the secreted IFN- ⁇ after culture of PBMCs with Influenza virus infected dendritic cells in the presence or absence of anti-PD-1 antibodies.
  • Figure 19 Chart showing the expression of exhaustion markers PD-1 , PD-L1 , TIM-3, LAG-3 and CTLA4 by lung tumour infiltrating lymphocytes. Approximately 2/3 of CD4+ and CD8+ lymphocytes in the tumour express PD-1.
  • Figure 20 Charts showing IFN- ⁇ secretion by tumour infiltrating lymphocytes after a mixed lymphocyte reaction in the presence or absence of anti-PD-1 antibodies, for three patients.
  • A Patient #1
  • B Patient #2
  • C Patient #3. Shown are mean ⁇ SD from duplicates or triplicates.
  • Figure 21 Charts showing the expression of exhaustion markers PD-1 , PD-L1 , ⁇ - 3, LAG-3 and CTLA4 by (A) renal tumour infiltrating lymphocytes and (B) blood circulating lymphocytes from renal carcinoma patients. Approximately 2/3 of CD4+ and CD8+ lymphocytes in the tumour express PD-1 , while most of PBMC lymphocytes do not express PD-1.
  • Figure 22 Charts showing IFN- ⁇ secretion by tumour infiltrating lymphocytes after a mixed lymphocyte reaction in the presence or absence of anti-PD-1 antibodies, for three patients.
  • C) Patient #3 Shown are mean ⁇ SD from duplicates or triplicates.
  • Figure 23 Charts showing the expression of exhaustion markers PD-1 , PD-L1 , TIM- 3, LAG-3 and CTLA4 by (A) bladder tumour infiltrating lymphocytes and (B) blood circulating lymphocytes from bladder carcinoma patients. A majority of tumour infiltrating lymphocytes express PD-1 , while only a minority of PBMC lymphocytes express PD-1.
  • Figure 24 Chart showing IFN- ⁇ secretion by tumour infiltrating lymphocytes after a mixed lymphocyte reaction in the presence or absence of anti-PD-1 antibodies, for one patient. Shown are mean ⁇ SD from duplicates or triplicates. Examples
  • the inventors described in the following Examples the identification of nucleotide and amino-acid sequences of isolated antibodies, or the antigen-binding portions thereof, that specifically bind human and rhesus PD-1 , block the PD-1 pathway and restore exhausted T cell activity.
  • Anti-PD-1 antibodies were isolated from a human antibody phage display library via in vitro selection in a 4-round bio-panning process.
  • Streptavidin-magnetic beads were coated with biotinylated human PD-1 and used to fish- out anti-PD-1-specific phages using magnetic sorting. Some steps to remove of potential anti-biotin antibodies were added in the selection process.
  • Fab antibodies were originally identified by ELISA with human-PD-1 as the antigen.
  • a first clonality screening was performed by DNA fingerprinting; clonality was then confirmed by sequencing.
  • Affinity of A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4 and H9 for human PD-1 was measured by Surface Plasmon Resonance and compared to the affinity of nivolumab and lambrolizumab, two anti-PD-1 antibodies in late clinical development stage (Figure 5).
  • A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4 and H9 show affinity in the range 0.1 to 0.7 nM while nivolumab and lambrolizumab exhibit KD of 1.9 and 0.5 nM respectively ( Figure 5).
  • T cells isolated from a healthy donor were incubated in the presence of 5,000 monocyte-derived DCs from another donor for 7 days at 37°C.
  • Antibodies, expressed as lgG4 were incubated with such activated T cells for 30 minutes in PBS. After a wash in PBS, a fluorescent labelled secondary antibody was added for 30 minutes and then washed in PBS. Cells were resuspended in FACS buffer and binding of the antibody to the cells was monitored by flow cytometry ( Figures 6 and 7).
  • Affinity-matured clone Fabs were tested in ELISA for recognition of rhesus PD-1. Briefly, ELISA plates were coated with 350ng/well of human or rhesus PD-1 in carbonate buffer and then blocked with a solution of casein. After extensive washes in PBS Tween-20, antibody-containing supernatants were transferred into the ELISA plates in the presence of 7% milk in PBS. After 90 minutes at room temperature under agitation and extensive washes, a goat anti-human Fab antibody coupled to HRP was added. One hour later, plates were washed and TMB substrate added. The reaction was stopped with 1 M HCI and optical density measured at 450nm with a reference at 670nm ( Figure 8).
  • Anti-PD-1 antibodies were tested in a functional assay measuring 2 activity parameters of T cells: proliferation and secretion of IFN- ⁇ . Briefly, T cells were isolated from a healthy donor and cultured for 7 days in the presence of monocyte-derived dendritic cells from another donor (50,000 Tcells / 5,000 DCs). This continuous stimulation induces exhaustion of the T cells. Antibodies were then added to the cultures for 5 more days. After 4 days, supernatants were collected to measure IFN- ⁇ by ELISA, and T cells were cultured for 4 days, with addition of 1 ⁇ of tritiated thymidine for the last 18 hours. Cells were then harvested and proliferation measured with a ⁇ -counter.
  • Clones A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4 and H9 are able to restore proliferation of previously exhausted T cells and to restore their ability to secrete IFN- ⁇ ( Figures 9 to 12).
  • Anti-PD-1 antibodies were tested in an ELISA assay for their ability to bind to other members of the CD28 family.
  • ELISA plates were coated with 350ng/well of one of the following antigens coupled to human Fc in carbonate buffer: human PD-1 , human PD-L1 , human TIM-3, human LAG-3, human ICOS, human CTLA4, human BTLA, human CD28, mouse TIM-3, or rhesus PD-1.
  • the plate was then blocked with a solution of casein. After extensive washes in PBS Tween-20, antibodies were added into the ELISA wells in the presence of 7% milk in PBS. After 90 minutes at room temperature under agitation and extensive washes, a goat anti-human Fab antibody coupled to HRP was added. One hour later, plates were washed and TMB substrate added.
  • nivolumab cross-binds to PD-L1 , TIM-3, LAG-3, BTLA and CD28 (and
  • 1 D1 1 and 1G4 are specific to PD-1 only; showing only a very weak cross-recognition of other members than PD-1 ( Figure 13).
  • HEK-293.6E cells were transiently transfected to produce anti-PD-1 antibodies in lgG4 format; production yield in this system was compared.
  • Codon-optimised clones retain their ability to neutralise PD-1 and restore T cell activity (T cell proliferation and IFN- ⁇ secretion - Figures 14 and 15).
  • tumour dissociated mixture was cultured with anti-PD-1 codon-optimised antibodies A10, B6, C4 and H4 for 7 days prior to measurement of IFN- ⁇ in the supernatant by
  • Nivolumab and lambrolizumab were used as positive controls, an isotype antibody as a negative control.
  • Figure 17A presents the secretion of IFN- ⁇ by tumour infiltrating lymphocytes after 7 days of culture in the presence or absence of anti-PD-1 antibodies.
  • Anti-PD-1 antibodies were able to re-activate lymphocytes to secrete IFN- ⁇ in a dose-dependent manner.
  • Another fraction of the dissociated mixture was co-cultured with allogeneic dendritic cells (DCs) to initiate a mixed lymphocyte reaction (MLR).
  • DCs dendritic cells
  • MLR mixed lymphocyte reaction
  • Cells were first cultured with DCs for 7 days without antibodies and then restimulated with DCs for 7 days in the presence of anti-PD-1 or control antibodies. After these 2 rounds, IFN- ⁇ was assayed in supernatants by ELISA.
  • Figure 17B presents the secretion of IFN- ⁇ after the MLR in the presence or absence of anti-PD-1 antibodies.
  • Anti-PD-1 antibodies were able to restore the ability of lymphocytes located in the tumour site to secrete IFN- ⁇ in a dose-dependent manner.
  • anti-PD-1 antibodies to treat infections: autologous activation of T cells in the presence of Influenza
  • Monocyte-derived DCs were infected with influenza virus A/PR/8/34 (H1 N1). Infected DCs were then mixed to PBMCs from the same donor for a first round of culture of 5 days. Cells were then restimulated with Influenza- infected DCs and cultured for a second round of 5 days in the presence of anti- PD-1 antibodies. After these 2 rounds, most of the cells in culture are lnfluenza-spec ⁇ f c T cells. At the end of the 2 rounds of culture, IFN- ⁇ was assayed in supernatants by ELISA.
  • Figure 18 presents the secreted IFN- ⁇ after culture of PBMCs with Influenza virus infected DCs in the presence or absence of anti-PD-1 antibodies.
  • Anti-PD-1 antibodies were able to restore the capacity of lymphocytes to secrete IFN- ⁇ upon viral stimulation in a dose-dependent manner.
  • Lung cancer use of anti-PD-1 antibodies to re-activate lung tumour infiltrating lymphocytes lex vivo data
  • isolated cell mixture was washed once and passed through 70 ⁇ filter to obtain single cell suspension.
  • Cells were stained with antibodies against CD4, CD8, PD- 1 , PD-L1 , Tim-3, LAG-3 and CTLA-4; a live/dead marker was also used to exclude dead cells from the analysis. Cells were analysed by flow cytometry.
  • Figure 19 shows the expression of PD-1 , PD-L1 , TIM-3, LAG-3 and CTLA-4 by tumour infiltrating lymphocytes from 3 different patients (shown are mean ⁇ SD from 3
  • tumour dissociated mixture was co-cultured with allogeneic dendritic cells (DC) to initiate a mixed lymphocyte reaction (MLR).
  • DC dendritic cells
  • MLR mixed lymphocyte reaction
  • Cells were first cultured for 7 days without antibodies and then for 7 days in the presence of anti-PD-1 or control antibodies. After these 2 rounds, IFN- ⁇ was assayed in supernatants by ELISA.
  • Figures 20A-20C present the secretion of IFN- ⁇ after the MLR in the presence or absence of anti-PD-1 antibodies. Shown are mean ⁇ SD from triplicates in 3 independent experiments (cells from 3 different patients). Anti-PD-1 antibodies were able to restore the ability of lymphocytes located in the tumour site to secrete IFN- ⁇ in a dose-dependent manner.
  • Renal cell carcinoma use of anti-PD-1 antibodies to re-activate kidney tumour infiltrating lymphocytes lex vivo data
  • Kidney tumour samples were obtained from the National Cancer Centre Singapore after approval by the proper IRB. Samples were dissociated using a human tumour
  • tissue dissociation kit and a tissue dissociator device.
  • isolated cell mixture was washed once and passed through 70 ⁇ filter to obtain single cell suspension.
  • Cells were stained with antibodies against CD4, CD8, PD- 1 , PD-L1 , Tim-3, LAG-3 and CTLA-4; a live/dead marker was also used to exclude dead cells from the analysis. Cells were analysed by flow cytometry. As a comparison, same markers expression was assessed on PBMCs from the same patients.
  • Figures 21 A and 21 B show the expression of PD-1 , PD-L1 , TIM-3, LAG-3 and CTLA-4 by tumour infiltrating lymphocytes (Fig. 21 A) and on blood circulating lymphocytes (Fig. 21 B) from 3 different patients (shown are mean ⁇ SD from 3 independent experiments using cells from 3 different donors, all experiments done in triplicates). Approximately 2/3 of CD4+ and CD8+ lymphocytes in the tumour express PD-1 while most of PBMC lymphocytes do not express PD-1.
  • tumour dissociated mixture was co-cultured with allogeneic dendritic cells (DC) to initiate a mixed lymphocyte reaction (MLR).
  • DC dendritic cells
  • MLR mixed lymphocyte reaction
  • Cells were first cultured for 7 days without antibodies and then for 7 days in the presence of anti-PD-1 or control antibodies. After these 2 rounds, IFN- ⁇ was assayed in supernatants by ELISA.
  • Figures 22A-22C present the secretion of IFN- ⁇ after the MLR in the presence or absence of anti-PD-1 antibodies. Shown are mean ⁇ SD from duplicates or triplicates in 3 independent experiments (cells from 3 different patients). Anti-PD-1 antibodies were able to restore the ability of lymphocytes located in the tumour site to secrete IFN- ⁇ in a dose- dependent manner.
  • Bladder cancer use of anti-PD-1 antibodies to re-activate bladder tumour infiltrating lymphocytes lex vivo data
  • Bladder tumour samples were obtained from the National Cancer Centre Singapore after approval by the proper IRB. Samples were dissociated using a human tumour dissociation kit and a tissue dissociator device.
  • FIG. 23A and 23B show the expression of PD-1 , PD-L1 , TIM-3, LAG-3 and CTLA-4 by tumour infiltrating lymphocytes (Fig. 23A) and blood circulating lymphocytes (Fig.
  • tumour dissociated mixture was co-cultured with allogeneic dendritic cells (DC) to initiate a mixed lymphocyte reaction (MLR).
  • DC dendritic cells
  • MLR mixed lymphocyte reaction
  • Cells were first cultured for 7 days without antibodies and then for 7 days in the presence of anti-PD-1 or control antibodies. After these 2 rounds, IFN- ⁇ was assayed in supernatants by ELISA.
  • Figure 24 presents the secretion of IFN- ⁇ after the MLR in the presence or absence of anti-PD-1 antibodies. Shown are mean ⁇ SD from triplicates in 1 experiment. Anti-PD-1 antibodies were able to restore the ability of lymphocytes located in the tumour site to secrete IFN- ⁇ in a dose-dependent manner.

Abstract

Anti-PD-1 antibodies are disclosed. Also disclosed are pharmaceutical compositions comprising such antibodies, and methods of using such antibodies to restore T-cell function in T-cells. The antibodies may also be used in the treatment of cancer or infectious diseases.

Description

Anti-PD-1 Antibodies
Field of the Invention
The present invention relates to antibodies that bind to programmed cell death 1 (PD-1).
Background to the Invention
T-cell exhaustion is a state of T-cell dysfunction that arises during many chronic infections and cancer. It is defined by poor T-cell effector function, sustained expression of inhibitory receptors and a transcriptional state distinct from that of functional effector or memory T- cells. Exhaustion prevents optimal control of infection and tumors. (E John Wherry., Nature Immunology 12, 492-499 (201 1)).
T-cell exhaustion is characterized by the stepwise and progressive loss of T-cell functions. Exhaustion is well-defined during chronic lymphocytic choriomeningitis virus infection and commonly develops under conditions of antigen-persistence, which occur following many chronic infections including hepatitis B virus, hepatitis C virus and human immunodeficiency virus infections, as well as during tumor metastasis. Exhaustion is not a uniformly disabled setting as a gradation of phenotypic and functional defects can manifest, and these cells are distinct from prototypic effector, memory and also anergic T cells. Exhausted T cells most commonly emerge during high-grade chronic infections, and the levels and duration of antigenic stimulation are critical determinants of the process. (Yi et al., Immunology Apr 2010; 129(4):474-481).
Circulating human tumor-specific CD8+ T cells may be cytotoxic and produce cytokines in vivo, indicating that self- and tumor-specific human CD8+ T cells can reach functional competence after potent immunotherapy such as vaccination with peptide, incomplete Freund's adjuvant (I FA), and CpG or after adoptive transfer. In contrast to peripheral blood, T-cells from metastasis are functionally deficient, with abnormally low cytokine production and upregulation of the inhibitory receptors PD-1 , CTLA-4, and TIM-3.
Functional deficiency is reversible, since T-cells isolated from melanoma tissue can restore IFN-γ production after short-term in vitro culture. However, it remains to be determined whether this functional impairment involves further molecular pathways, possibly resembling T-cell exhaustion or anergy as defined in animal models. (Baitsch et a\., J CIin Invest. 201 1 ; 121 (6):2350-2360). Programmed cell death 1 (PD-1), also called CD279, is a type I membrane protein encoded in humans by the PDCD1 gene. It has two ligands, PD-L1 and PD-L2.
The PD-1 pathway is a key immune-inhibitory mediator of T-cell exhaustion. Blockade of this pathway can lead to T-cell activation, expansion, and enhanced effector functions. As such, PD-1 negatively regulates T cell responses. PD-1 has been identified as a marker of exhausted T cells in chronic disease states, and blockade of PD-1 :PD-1 L interactions has been shown to partially restore T cell function. (Sakuishi et al., JEM Vol. 207, September 27, 2010, pp2187-2194). Nivolumab (BMS-936558) is an anti-PD-1 antibody that was approved for the treatment of melanoma in Japan in July 2014. Other anti-PD-1 antibodies are described in WO 2010/077634, WO 2006/121 168, WO2008/156712 and WO2012/135408.
T cell immunoglobulin mucin 3 (TIM-3) is an immune regulator identified as being upregulated on exhausted CD8+ T cells (Sakuishi et al., JEM Vol. 207, September 27, 2010, pp2187-2194). TIM-3 was originally identified as being selectively expressed on
IFN-y-secreting Th1 and Tc1 cells. Interaction of TIM-3 with its ligand, galectin-9, triggers cell death in TIM-3* T ceils. Anti-TIM-3 antibodies are described in Ngiow et al (Cancer Res. 2011 May 15;71 (10):3540-51),and in US8,552,156
Both TIM-3 and PD-1 can function as negative regulators of T cell responses and combined targeting of the TIM-3 and PD-1 pathways is more effective in controlling tumor growth than targeting either pathway alone. (Sakuishi et al., JEM Vol. 207, September 27, 2010, pp2187-2194; and Ngiow et al Cancer Res. 201 1 May 15;71 (10):3540-51).
Summary of the Invention
The present invention is concerned with antibodies, or antigen binding fragments, that bind to PD-1. Heavy and light chain polypeptides are also disclosed. The antibodies, antigen binding fragments and polypeptides may be provided in isolated and/or purified form and may be formulated into compositions suitable for use in research, therapy and diagnosis. In some embodiments the antibody, or antigen binding fragment, or polypeptide may be effective to restore T-cell function in T-cells, e.g. CD8+ T-cells, exhibiting T-cell exhaustion or T-cell anergy. In one aspect of the present invention an antibody, or antigen binding fragment, is provided, the amino acid sequence of the antibody may comprise the amino acid sequences i) to iii), or the amino acid sequences iv) to vi), or preferably the amino acid sequences i) to vi): i) LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25) ii) LC-CDR2: SNNQRPS (SEQ ID NO:26) iii) LC-CDR3: X1X2WDDX3X4X5GX6X7 (SEQ ID NO:53) iv) HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
HC-CDR1 : SYGMH (SEQ ID NO:89) v) HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40) vi) HC-CDR3: DZ1GZ2GZ3YZ4YGZ5DZ6 (SEQ ID NO:54) or a variant thereof in which one or two or three amino acids in one or more of the sequences (i) to (vi) are replaced with another amino acid, where
Figure imgf000005_0001
A or S, X2= S or A, Xs= V, Y, F, D, S or A, X4= L, Y, V or A, Xs= Y, R or H, Xe= S, or T, X7= V, I, or M and Zi= L or Y, Z2= A or S, Z3= P or Y, Z4= Y or L, Z5= K, M or L, Z6= H or V.
In connection with all aspects of the present invention, in embodiments wherein HC- CDR1 : SYGMH (SEQ ID NO:89), this sequence may be comprised in the larger sequence GFTFSSYGMH (SEQ ID NO:39).
In some embodiments, LC-CDR3 is one of ASWDDVLYGSV (SEQ ID NO:27),
ASWDDYYYGTI (SEQ ID NO:28), ASWDDYLRGTV (SEQ ID NO:29), SAWDDYLHGTV (SEQ ID NO:30), ASWDDYVRGTM (SEQ ID NO:31), SSWDDFLRGTV (SEQ ID NO:32), SSWDDDARGTI (SEQ ID NO:33), AAWDDVYYGTI (SEQ ID NO:34), ASWDDSLYGTV (SEQ ID NO:35), AAWDDAYYGTI (SEQ ID NO:36), ASWDDVYRGTV (SEQ ID NO:37), or SSWDDSLYGTI (SEQ ID NO:38). In some embodiments HC-CDR3 is one of
DLGAGPYYYGKDH (SEQ ID NO:41), DLGAGPYYYGKDV (SEQ ID NO:42),
DYGAGPYYYGMDV (SEQ ID NO:43), DLGAGPYYYGLDV (SEQ ID NO:44),
D LGAG PYYYG M D V (SEQ ID NO:45), DLGAGPYYYGMDV (SEQ ID NO:46),
DLGAGPYYYGMDV (SEQ ID NO:47), DLGAGPYYYGMDV (SEQ ID NO:48),
DLGAGPYYYGMDV (SEQ ID NO:49), DYGAGPYYYGMDV (SEQ ID NO:50),
DLGSGYYLYGMDV (SEQ ID NO:51), or DLGAGPYYYGMDV (SEQ ID NO:52). In some embodiments the antibody, or antigen binding fragment, may comprise at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDVLYGSV (SEQ ID NO:27)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDYYYGTI (SEQ ID NO:28)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDYLRGTV (SEQ ID NO:29) In some embodiments the antibody, or antigen binding fragment, may comprise at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: SAWDDYLHGTV (SEQ ID NO:30)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDYVRGTM (SEQ ID NO:31)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25) LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: SSWDDFLRGTV (SEQ ID NO:32)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: SSWDDDARGTI (SEQ ID NO:33)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: AAWDDVYYGTI (SEQ ID NO:34)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDSLYGTV (SEQ ID NO:35)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: AAWDDAYYGTI (SEQ ID NO:36)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDVYRGTV (SEQ ID NO:37) In some embodiments the antibody, or antigen binding fragment, may comprise at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: SSWDDSLYGTI (SEQ ID NO:38)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGKDH (SEQ ID NO:41)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: D LG AG PYYYG KD V (SEQ ID NO:42)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: D YG AG PYYYG M D V (SEQ ID NO:43)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGLDV (SEQ ID NO:44) In some embodiments the antibody, or antigen binding fragment, may comprise at least one heavy chain variable region incorporating the following CDRs: HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:45)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:46)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:47)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:48)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:49)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DYGAGPYYYGMDV (SEQ ID NO:50)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGSGYYLYGMDV (SEQ ID NO:51)
In some embodiments the antibody, or antigen binding fragment, may comprise at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:52)
The antibody may comprise at least one light chain variable region incorporating the CDRs shown in Figures 1 or 3. The antibody may comprise at least one heavy chain variable region incorporating the CDRs shown in Figures 2 or 3. The antibody may comprise at least one light chain variable region (VL) comprising the amino acid sequence of one of SEQ ID NOs 1 , 25, 26, 27 or 2, 25, 26, 28, or 3, 25, 26, 29 or 4, 25, 26, 30, or 5, 25, 26, 31 or 6, 25, 26, 32 or 7, 25, 26, 33 or 8, 25, 26, 34 or 9, 25, 26, 35 or 10, 25, 26 36 or 11 , 25, 26, 37 or 12, 25, 26, 38, or one of the amino acid sequences shown in Figure 1 or an amino acid sequence having at least 70%, more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to one of SEQ ID NOs 1 , 25, 26, 27 or 2, 25, 26, 28, or 3, 25, 26, 29 or 4, 25, 26, 30, or 5, 25, 26, 31 or 6, 25, 26, 32 or 7, 25, 26, 33 or 8, 25, 26, 34 or 9, 25, 26, 35 or 10, 25, 26 36 or 1 1 , 25, 26, 37 or 12, 25, 26, 38, or to the amino acid sequence of the VL chain amino acid sequence shown in Figure 1. The antibody may comprise at least one heavy chain variable region (VH) comprising the amino acid sequence of one of SEQ ID NOs 13, 39 or 89, 40, 41 or 14, 39 or 89, 40, 42, or 15, 39 or 89, 40, 43 or 16, 39 or 89, 40, 44 or 17, 39 or 89, 40, 45 or 18, 39 or 89, 40, 46 or 19, 39 or 89, 40, 47 or 20, 39 or 89, 40, 48 or 21 , 39 or 89, 40, 49 or 22, 39 or 89, 40, 50 or 23, 39 or 89, 40, 51 or 24, 39 or 89, 40, 52, or one of the amino acid sequences shown in Figure 2 or an amino acid sequence having at least 70%, more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to one of SEQ ID NOs 13, 39, 40, 41 or 14, 39 or 89, 40, 42, or 15, 39 or 89, 40, 43 or 16, 39 or 89, 40, 44 or 17, 39 or 89, 40, 45 or 18, 39 or 89, 40, 46 or 19, 39 or 89, 40, 47 or 20, 39 or 89, 40, 48 or 21 , 39 or 89, 40, 49 or 22, 39 or 89, 40, 50 or 23, 39 or 89, 40, 51 or 24, 39 or 89, 40, 52, or to the amino acid sequence of the VH chain amino acid sequence shown in Figure 2.
The antibody may comprise at least one light chain variable region comprising the amino acid sequence of one of SEQ ID NOs 1 , 25, 26, 27 or 2, 25, 26, 28, or 3, 25, 26, 29 or 4, 25, 26, 30, or 5, 25, 26, 31 or 6, 25, 26, 32 or 7, 25, 26, 33 or 8, 25, 26, 34 or 9, 25, 26, 35 or 10, 25, 26 36 or 1 1 , 25, 26, 37 or 12, 25, 26, 38, or to one of the amino acid sequences shown in Figure 1 (or an amino acid sequence having at least 70%, more preferably one of at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100%, sequence identity to one of SEQ ID NOs 1 , 25, 26, 27 or 2, 25, 26, 28, or 3, 25, 26, 29 or 4, 25, 26, 30, or 5, 25, 26, 31 or 6, 25, 26, 32 or 7, 25, 26, 33 or 8, 25, 26, 34 or 9, 25, 26, 35 or 10, 25, 26 36 or 1 1 , 25, 26, 37 or 12, 25, 26, 38, or to to one of the amino acid sequences of the VL chain amino acid sequence shown in Figure 1 ) and at least one heavy chain variable region comprising the amino acid sequence of one of SEQ ID NOs 13, 39 or 89, 40, 41 or 14, 39 or 89, 40, 42, or 15, 39 or 89, 40, 43 or 16, 39 or 89, 40, 44 or 17, 39 or 89, 40, 45 or 18, 39 or 89, 40, 46 or 19, 39 or 89, 40, 47 or 20, 39 or 89, 40, 48 or 21 , 39 or 89, 40, 49 or 22, 39 or 89, 40, 50 or 23, 39 or 89, 40, 51 or 24, 39 or 89, 40, 52, or one of the amino acid sequence shown in Figure 2 (or an amino acid sequence having at least 70%, more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to one of SEQ ID NOs 13, 39 or 89, 40, 41 or 14, 39 or 89, 40, 42, or 15, 39 or 89, 40, 43 or 16, 39 or 89, 40, 44 or 17, 39 or 89, 40, 45 or 18, 39 or 89, 40, 46 or 19, 39 or 89, 40, 47 or 20, 39 or 89, 40, 48 or 21 , 39 or 89, 40, 49 or 22, 39 or 89, 40, 50 or 23, 39 or 89, 40, 51 or 24, 39 or 89, 40, 52, or to one of the amino acid sequences of the VH chain amino acid sequence shown in Figure 2). The antibody may optionally bind PD-1. The antibody may optionally have amino acid sequence components as described above. The antibody may be an IgG. In one embodiment an in vitro complex, optionally isolated, comprising an antibody, or antigen binding fragment, as described herein, bound to PD-1 is provided.
In one aspect of the present invention an isolated heavy chain variable region polypeptide is provided, the heavy chain variable region polypeptide comprising the following CDRs:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DZ1GZ2GZ3YZ4YGZ5DZ6 (SEQ ID NO:54)
where Z^= L or Y, Z2= A or S, Z3= P or Y, Z4= Y or L, Z5= K, M or L, Z6= H or V.
In some embodiments HC-CDR3 is one of DLGAGPYYYGKDH (SEQ ID NO:41), D LG AG PYYYG KD V (SEQ ID NO:42), DYGAGPYYYGMDV (SEQ ID NO:43),
DLGAGPYYYGLDV (SEQ ID NO:44), D LG AG PYYYG MDV (SEQ ID NO:45),
D LGAG PYYYG M D V (SEQ ID NO:46), DLGAG PYYYG MDV (SEQ ID NO:47),
D LGAG PYYYG M D V (SEQ ID NO:48), DLGAG PYYYG MDV (SEQ ID NO:49),
DYGAGPYYYGMDV (SEQ ID NO:50), DLGSGYYLYGMDV (SEQ ID NO:51), or
DLGAGPYYYGMDV (SEQ ID NO:52).
In one aspect of the present invention an antibody, or antigen binding fragment, is provided, the antibody, or antigen binding fragment, comprising a heavy chain and a light chain variable region sequence, wherein:
the heavy chain comprises a HC-CDR1 , HC-CDR2, HC-CDR3, having at least 85% overall sequence identity to
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or SYGMH (SEQ ID NO:89),
HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40),
HC-CDR3: is one of DZ^ZzGZsYZ^GZsDZe (SEQ ID NO:54),
DLGAGPYYYGKDH (SEQ ID NO:41), D LGAG PYYYG KDV (SEQ ID NO:42), DYGAGPYYYGMDV (SEQ ID NO:43), DLGAGPYYYGLDV (SEQ ID NO:44), DLGAGPYYYGMDV (SEQ ID NO:45), DLGAGPYYYGMDV (SEQ ID NO:46),
DLGAGPYYYGMDV (SEQ ID NO:47), DLGAGPYYYGMDV (SEQ ID NO:48), DLGAGPYYYGMDV (SEQ ID NO:49), DYGAGPYYYGMDV (SEQ ID NO:50), DLGSGYYLYGMDV (SEQ ID NO:51), or DLGAGPYYYGMDV (SEQ ID NO:52) respectively, where Z^= L or Y,∑2= A or S, Zz= P or Y, Z4= Y or L, Zs= K, M or L, Z6= H or V, and the light chain comprises a LC-CDR1 , LC-CDR2, LC-CDR3,, having at least 85% overall sequence identity to
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25),
LC-CDR2: SNNQRPS (SEQ ID NO:26),
LC-CDR3: is one of X1X2WDDX3X4X5GX6X7 (SEQ ID NO:53), ASWDDVLYGSV
(SEQ ID NO:27), ASWDDYYYGTI (SEQ ID NO:28), ASWDDYLRGTV (SEQ ID NO:29), SAWDDYLHGTV (SEQ ID NO:30), ASWDDYVRGTM (SEQ ID NO:31), SSWDDFLRGTV (SEQ ID NO:32), SSWDDDARGTI (SEQ ID NO:33),
AAWDDVYYGTI (SEQ ID NO:34), ASWDDSLYGTV (SEQ ID NO:35),
AAWDDAYYGTI (SEQ ID NO:36), ASWDDVYRGTV (SEQ ID NO:37), or
SSWDDSLYGTI (SEQ ID NO:38) respectively, where
Figure imgf000013_0001
V, Y, F, D, S or A, X4= L, Y, V or A, X5= Y, R or H, Xe= S, or T, X7= V, I, or M. In some embodiments the degree of sequence identity may be one of 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
In another aspect of the present invention an antibody, or antigen binding fragment, optionally isolated, is provided comprising a heavy chain and a light chain variable region sequence, wherein:
the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence of one of SEQ ID NOs: 13 to 24 (Figure 2), and
the light chain sequence has at least 85% sequence identity to the light chain sequence of one of: SEQ ID NOs:1 to 12 (Figure 1).
In some embodiments the degree of sequence identity may be one of 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
In some embodiments the antibody, antigen binding fragment, or polypeptide further comprises variable region heavy chain framework sequences between the CDRs according to the arrangement HCFR1 :HC-CDR1 :HCFR2:HC-CDR2:HCFR3:HC- CDR3:HCFR4. The framework sequences may be derived from human consensus framework sequences.
In one aspect of the present invention an isolated light chain variable region polypeptide, optionally in combination with a heavy chain variable region polypeptide as described herein, is provided, the light chain variable region polypeptide comprising the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25) LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: Xi X2 WD DX3X4X5GX6X7 (SEQ ID NO:53)
where
Figure imgf000014_0001
Y, R or H, X6=
S, or T, X7= V, I, or M.
In some embodiments LC-CDR3 is one of ASWDDVLYGSV (SEQ ID NO:27),
ASWDDYYYGTI (SEQ ID NO:28), ASWDDYLRGTV (SEQ ID NO:29), SAWDDYLHGTV (SEQ ID NO:30), ASWDDYVRGTM (SEQ ID NO:31), SSWDDFLRGTV (SEQ ID NO:32), SSWDDDARGTI (SEQ ID NO:33), AAWDDVYYGTI (SEQ ID NO:34), ASWDDSLYGTV (SEQ ID NO:35), AAWDDAYYGTI (SEQ ID NO:36), ASWDDVYRGTV (SEQ ID NO:37), or SSWDDSLYGTI (SEQ ID NO:38).
In some embodiments the antibody, antigen binding fragment, or polypeptide further comprises variable region light chain framework sequences between the CDRs according to the arrangement LCFR1 :LC-CDR1 :LCFR2:LC-CDR2:LCFR3:LC-CDR3:LCFR4. The framework sequences may be derived from human consensus framework sequences.
In some embodiments, the antibody, or antibody binding fragment, may further comprise a human constant region. For example selected from one of lgG1 , lgG2, lgG3 and lgG4.
In some embodiments, the antibody, or antibody binding fragment, may further comprise a murine constant region. For example, selected from one of lgG1 , lgG2A, lgG2B and lgG3. In another aspect of the present invention, an antibody or antigen binding fragment is provided, optionally isolated, which is capable of binding to PD-1 , and which is a bispecific antibody or a bispecific antigen binding fragment. In some embodiments, the bispecific antibody or bispecific antigen binding fragment comprises an antigen binding fragment or polypeptide capable of binding to PD-1 as described herein, and additionally comprises an antigen binding domain which is capable of binding to another target protein, e.g. a target protein other than PD-1. In some embodiments, the target protein is a cell surface receptor. In some embodiments, the target protein is a cell surface receptor expressed on the cell surface of immune cells, e.g. T cells. In some embodiments, the target protein may be a member of the CD28 family. In some embodiments, the member of the CD28 family is selected from TIM-3, LAG3, ICOS, CTLA4, BTLA or CD28. In another aspect of the present invention, a composition, e.g. a pharmaceutical composition or medicament, is provided. The composition may comprise an antibody, antigen binding fragment, or polypeptide as described herein and at least one
pharmaceutically-acceptable carrier, excipient, adjuvant or diluent.
In another aspect of the present invention an isolated nucleic acid encoding an antibody, antigen binding fragment, or polypeptide as described herein is provided. The nucleic acid may have a sequence of one of SEQ ID NOs 55, 56, 58, 59, 60, 61 , 62, 63, 64, 65, 66, 67, 68, 69, 70, 71 , 72, 73, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 87, or 88 (Figure 4), or a coding sequence which is degenerate as a result of the genetic code, or may have a nucleotide sequence having at least 70% identity thereto, optionally one of 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%. In one aspect of the present invention there is provided a vector comprising a nucleic acid described herein. In another aspect of the present invention, there is provided a host cell comprising the vector. For example, the host cell may be eukaryotic, or mammalian, e.g. Chinese Hamster Ovary (CHO), or human or may be a prokaryotic cell, e.g. E. coli.
In one aspect of the present invention a method for making an antibody, or antigen binding fragment or polypeptide as described herein is provided, the method comprising culturing a host cell as described herein under conditions suitable for the expression of a vector encoding the antibody, or antigen binding fragment or polypeptide, and recovering the antibody, or antigen binding fragment or polypeptide. In another aspect of the present invention an antibody, antigen binding fragment or polypeptide is provided for use in therapy, or in a method of medical treatment. In another aspect of the present invention an antibody, antigen binding fragment or polypeptide as described herein is provided for use in the treatment of a T-cell dysfunctional disorder. In another aspect of the present invention, the use of an antibody, antigen binding fragment or polypeptide as described herein in the manufacture of a medicament or pharmaceutical composition for use in the treatment of a T-cell dysfunctional disorder is provided.
In another aspect of the present invention a method of enhancing T-cell function comprising administering an antibody, antigen binding fragment or polypeptide as described herein to a dysfunctional T-cell is provided. The method may be performed in vitro or in vivo.
In another aspect of the present invention a method of treating a T-cell dysfunctional disorder is provided, the method comprising administering an antibody, antigen binding fragment or polypeptide as described herein to a patient suffering from a T-cell dysfunctional disorder.
In another aspect of the present invention a method of modulating an immune response in a subject is provided, the method comprising administering to the subject an antibody, antigen binding fragment or polypeptide as described herein such that the immune response in the subject is modulated.
In another aspect of the present invention a method of inhibiting growth of tumor cells in a subject is provided, the method comprising administering to the subject a therapeutically effective amount of an antibody, antigen binding fragment or polypeptide as described herein.
In another aspect of the present invention a method is provided, the method comprising contacting a sample containing, or suspected to contain, PD-1 with an antibody or antigen binding fragment, as described herein, and detecting the formation of a complex of antibody, or antigen binding fragment, and PD-1.
In another aspect of the present invention a method of diagnosing a disease or condition in a subject is provided, the method comprising contacting, in vitro, a sample from the subject with an antibody, or antigen binding fragment, , as described herein, and detecting the formation of a complex of antibody, or antigen binding fragment, and PD-1.
In a further aspect of the present invention a method of selecting or stratifying a subject for treatment with PD-1 targeted agents is provided, the method comprising contacting, in vitro, a sample from the subject with an antibody, or antigen binding fragment, according to the present invention and detecting the formation of a complex of antibody, or antigen binding fragment, and PD-1. In a further aspect of the present invention the use of an antibody, or antigen binding fragment, as described herein, for the detection of PD-1 in vitro is provided. In another aspect of the present invention the use of an antibody, or antigen binding fragment, as described herein, as an in vitro diagnostic agent is provided.
In a further aspect of the present invention a method for expanding a population of T cells is provided, wherein T cells are contacted in vitro or ex vivo with an antibody, antigen binding fragment or polypeptide according to the present invention. In a further aspect of the present invention a method of treatment of a subject having a T- cell dysfunctional disorder is provided, the method comprising culturing T cells obtained from a blood sample from a subject in the presence of an antibody, antigen binding fragment or polypeptide according to the present invention so as to expand the T cell population, collecting expanded T cells, and administering the expanded T cells to a subject in need of treatment.
In methods of the present invention the antibody, antigen binding fragment or polypeptide may be provided as a composition as described herein. In some embodiments the antibody may be antibody clone A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4, or H9 as described herein.
Description Antibodies
Antibodies according to the present invention preferably bind to PD-1 (the antigen), preferably human or rhesus PD-1 , optionally with a Koin the range 0.1 to 2nM.
In any aspect of the present invention the antibody preferably specifically binds PD-1 (e.g. human or rhesus) over other members of the CD28 family (preferably from the same organism), such as one or more or each of TIM-3 (HAVCR2), LAG3 (CD223), ICOS (CD278), CTLA4 (CD152), BTLA (CD272) or CD28.
Antibodies according to the present invention may be provided in isolated form. Antibodies according to the present invention may exhibit least one of the following properties:
a) binds to human PD-1 with a KD of 1 μΜ or less, preferably one of≤10nM,≤1 nM, ≤800pM,≤700pM,≤600pM,≤500pM,≤400pM,≤300pM,≤200pM or≤100pM; b) does not substantially bind to human TIM-3, LAG3, ICOS, CTLA4, BTLA or
CD28;
c) increases T -cell proliferation in an Mixed Lymphocyte Reaction (MLR) assay (e.g. see Bromelow et al J.Immunol Methods, 2001 Jan 1 ;247(1-2): 1-8);
d) increases interferon-gamma production in an MLR assay; or
e) increases interleukin-2 (IL-2) secretion in an MLR assay.
In some embodiments, the antibodies may be capable of increasing interferon-gamma production in an MLR assay in a dose-dependent manner. In some embodiments, the antibodies may be capable of increasing interferon-gamma production in an MLR assay by lymphocytes expressing one or more markers of exhaustion, e.g. PD-1.
By "antibody" we include a fragment or derivative thereof, or a synthetic antibody or synthetic antibody fragment. In view of today's techniques in relation to monoclonal antibody technology, antibodies can be prepared to most antigens. The antigen-binding portion may be a part of an antibody (for example a Fab fragment) or a synthetic antibody fragment (for example a single chain Fv fragment [ScFv]). Suitable monoclonal antibodies to selected antigens may be prepared by known techniques, for example those disclosed in "Monoclonal Antibodies: A manual of techniques ", H Zola (CRC Press, 1988) and in "Monoclonal
Hybridoma Antibodies: Techniques and Applications ", J G R Hurrell (CRC Press, 1982). Chimaeric antibodies are discussed by Neuberger et al (1988, 8th International
Biotechnology Symposium Part 2, 792-799). Monoclonal antibodies (mAbs) are useful in the methods of the invention and are a homogenous population of antibodies specifically targeting a single epitope on an antigen.
Polyclonal antibodies are useful in the methods of the invention. Monospecific polyclonal antibodies are preferred. Suitable polyclonal antibodies can be prepared using methods well known in the art. Antigen binding fragments of antibodies, such as Fab and Fab2 fragments may also be used/provided as can genetically engineered antibodies and antibody fragments. The variable heavy (VH) and variable light (VL) domains of the antibody are involved in antigen recognition, a fact first recognised by early protease digestion experiments. Further confirmation was found by "humanisation" of rodent antibodies. Variable domains of rodent origin may be fused to constant domains of human origin such that the resultant antibody retains the antigenic specificity of the rodent parented antibody (Morrison et al (1984) Proc. Natl. Acad. Sd. USA 81 , 6851-6855).
That antigenic specificity is conferred by variable domains and is independent of the constant domains is known from experiments involving the bacterial expression of antibody fragments, all containing one or more variable domains. These molecules include Fab-like molecules (Better et al (1988) Science 240, 1041); Fv molecules (Skerra et al (1988) Science 240, 1038); single-chain Fv (ScFv) molecules where the VH and VL partner domains are linked via a flexible oligopeptide (Bird et al (1988) Science 242, 423; Huston et al (1988) Proc. Natl. Acad. Sd. USA 85, 5879) and single domain antibodies (dAbs) comprising isolated V domains (Ward et al (1989) Nature 341 , 544). A general review of the techniques involved in the synthesis of antibody fragments which retain their specific binding sites is to be found in Winter & Milstein (1991) Nature 349, 293- 299.
By "ScFv molecules" we mean molecules wherein the VH and VL partner domains are covalently linked, e.g. by a flexible oligopeptide. Fab, Fv, ScFv and dAb antibody fragments can all be expressed in and secreted from E. coli, thus allowing the facile production of large amounts of the said fragments.
Whole antibodies, and F(ab')2 fragments are "bivalent". By "bivalent" we mean that the said antibodies and F(ab')2 fragments have two antigen combining sites. In contrast, Fab, Fv, ScFv and dAb fragments are monovalent, having only one antigen combining site. Synthetic antibodies which bind to PD-1 may also be made using phage display technology as is well known in the art.
The present application also provides an antibody or antigen binding fragment which is capable of binding to PD-1 , and which is a bispecific antibody or a bispecific antigen binding fragment. In some embodiments, the bispecific antibody or bispecific antigen binding fragment may be isolated.
In some embodiments, the bispecific antibodies and bispecific antigen binding fragments comprise an antigen binding fragment or a polypeptide according to the present invention. In some embodiments, the bispecific antibodies and bispecific antigen binding fragments comprise an antigen binding domain capable of binding to PD-1 , wherein the antigen binding domain which is capable of binding to PD-1 comprises or consists of an antigen binding fragment or a polypeptide according to the present invention.
In some embodiments the bispecific antibodies and bispecific antigen binding fragments comprise an antigen binding domain capable of binding to PD-1 , and an antigen binding domain capable of binding to another target protein. The antigen binding domain capable of binding to another target protein may be capable of binding to another protein other than PD-1. In some embodiments, the target protein is a cell surface receptor. In some embodiments, the target protein is a cell surface receptor expressed on the cell surface of immune cells, e.g. T cells. In some embodiments, the target protein may be a member of the CD28 family. In some embodiments, the target protein may be a member of the CD28 family such as TIM-3 (HAVCR2), LAG 3 (CD223), ICOS (CD278), CTLA4 (CD152), BTLA (CD272) or CD28. In particular embodiments, the target protein may be CTLA4 or LAG3.
In some embodiments, the antigen binding domain for TIM-3 may comprise the CDRs, light and heavy chain variable domains or other TIM-3 binding fragment of e.g. anti-TIM-3 antibody clone F38-2E2 (BioLegend), clone 2E2 (Merck Millipore), clone 6B6E2, clone 024 (Sino Biological) clone 344801 (R&D Systems), clone E-18, clone H-191 (Santa Cruz Biotechnology), or clone 13A224 (United States Biological). In some embodiments, the antigen binding domain for LAG3 may comprise the CDRs, light and heavy chain variable domains or other LAG3 binding fragment of e.g. anti-LAG3 antibody clone 17B4 (Enzo
Life Sciences), clone 333210 (R&D Systems), or clone 14L676 (United States Biological). In some embodiments, the antigen binding domain for ICOS may comprise the CDRs, light and heavy chain variable domains or other ICOS binding fragment of e.g. anti-ICOS antibody clone ISA-3 (eBioscience), clone SP98 (Novus Biologicals), clone 1 G1 , clone 3G4 (Abnova Corporation), clone 669222 (R&D Systems), clone TQ09 (Creative
Diagnostics), or clone C398.4A (BioLegend). In some embodiments, the antigen binding domain for CTLA4 may comprise the CDRs, light and heavy chain variable domains or other CTLA4 binding fragment of e.g. anti-CTLA4 antibody clone 2F1 , clone 1 F4 (Abnova Corporation), clone 9H10 (EMD Millipore), clone BNU3 (GeneTex), clone 1 E2, clone AS32 (Lifespan Biosciences) clone A3.4H2.H12 (Acris Antibodies), clone 060 (Sino Biological), clone BU5G3 (Creative Diagnostics), clone MIH8 (MBL International), clone A3.6B10.G1 , or clone L3D10 (BioLegend). In some embodiments, the antigen binding domain for BTLA may comprise the CDRs, light and heavy chain variable domains or other BTLA binding fragment of e.g. anti-BTLA antibody clone 1 B7, clone 2G8, clone 4C5 (Abnova Corporation), clone 4B8 (antibodies-online), clone MIH26 (Thermo Scientific Pierce Antibodies), clone UMAB61 (OriGene Technologies), clone 330104 (R&D
Systems), clone 1 B4 (Lifespan Biosciences), clone 440205, clone 5E7 (Creative
Diagnostics). In some embodiments, the antigen binding domain for CD28 may comprise the CDRs, light and heavy chain variable domains or other CD28 binding fragment of e.g. anti-CD28 antibody clone CD28.6 (eBioscience), clone CD28.2, clone JJ319 (Novus Biologicals), clone 204.12, clone B-23, clone 10F3 (Thermo Scientific Pierce Antibodies), clone 37407 (R&D Systems), clone 204-12 (Abnova Corporation), clone 15E8 (EMD Millipore), clone 204-12, clone YTH913.12 (AbD Serotec), clone B-T3 (Acris Antibodies), clone 9H6E2 (Sino Biological), clone C28/77 (MyBioSource.com), clone KOLT-2
(ALPCO), clone 152-2E10 (Santa Cruz Biotechnology), or clone XPH-56 (Creative Diagnostics).
An antigen binding domain of a bispecific antibody or bispecific antigen binding fragment according to the present invention may be any domain of a polypeptide which is capable of binding to an antigen. In some embodiments, an antigen binding domain comprises at least the three light chain CDRs (i.e. LC-CDR1 , LC-CDR2 and LC-CDR3) and three heavy chain CDRs (i.e. HC-CDR1 , HC-CDR2 and HC-CDR3) which together define the antigen binding region of an antibody or antigen binding fragment. In some embodiments, an antigen binding domain may comprise the light chain variable domain and heavy chain variable domain of an antibody or antigen binding fragment. In some embodiments, an antigen binding domain may comprise the light chain polypeptide and heavy chain polypeptide of an antibody or antigen binding fragment.
Bispecific antibodies and bispecific antigen binding fragments according to the invention may be provided in any suitable format, such as those formats described in Kontermann MAbs 2012, 4(2): 182-197, which is hereby incorporated by reference in its entirety. For example, a bispecific antibody or bispecific antigen binding fragment may be a bispecific antibody conjugate (e.g. an lgG2, F(ab')2 or CovX-Body), a bispecific IgG or IgG-like molecule (e.g. an IgG, scFv4-lg, IgG-scFv, scFv-lgG, DVD-lg, IgG-sVD, sVD-lgG, 2 in 1- IgG, mAb2, or Tandemab common LC), an asymmetric bispecific IgG or IgG-like molecule (e.g. a kih IgG, kih IgG common LC, CrossMab, kih IgG-scFab, mAb-Fv, charge pair or SEED-body), a small bispecific antibody molecule (e.g. a Diabody (Db), dsDb, DART, scDb, tandAbs, tandem scFv (taFv), tandem dAb/VHH, triple body, triple head, Fab-scFv, or F(ab')2-scFv2), a bispecific Fc and CH3 fusion protein (e.g. a taFv-Fc, Di-diabody, scDb- CH3, SCFV-FC-SCFV, HCAb-VHH, scFv-kih-Fc, or scFv-kih-CH3), or a bispecific fusion protein (e.g. a scFv2-albumin, scDb-albumin, taFv-toxin, DNL-Fab3, DNL-Fab4-lgG, DNL- Fab4-lgG-cytokine2). See in particular Figure 2 of Kontermann MAbs 2012, 4(2): 182-19.
The skilled person is able to design and prepare bispecific antibodies and bispecific antigen binding fragments according to the present invention. Methods for producing bispecific antibodies include chemically crosslinking of antibodies or antibody fragments, e.g. with reducible disulphide or non-reducible thioether bonds, for example as described in Segal and Bast, 2001. Production of Bispecific Antibodies.
Current Protocols in Immunology. 14:IV:2.13:2.13.1— 2.13.16, which is hereby
incorporated by reference in its entirety. For example, A/-succinimidyl-3-(-2-pyridyldithio)- propionate (SPDP) can be used to chemically crosslink e.g. Fab fragments via hinge region SH- groups, to create disulfide-linked bispecific F(ab)2 heterodimers.
Other methods for producing bispecific antibodies include fusing antibody-producing hybridomas e.g. with polyethylene glycol, to produce a quadroma cell capable of secreting bispecific antibody, for example as described in D. M. and Bast, B. J. 2001.
Production of Bispecific Antibodies. Current Protocols in Immunology. 14:IV:2.13:2.13.1— 2.13.16.
Bispecific antibodies and bispecific antigen binding fragments according to the present invention can also be produced recombinantly, by expression from e.g. a nucleic acid construct encoding polypeptides for the antigen binding molecules, for example as described in Antibody Engineering: Methods and Protocols, Second Edition (Humana Press, 2012), at Chapter 40: Production of Bispecific Antibodies: Diabodies and Tandem scFv (Hornig and Farber-Schwarz), or French, How to make bispecific antibodies, Methods Mol. Med. 2000; 40:333-339, the entire contents of both of which are hereby incorporated by reference. For example, a DNA construct encoding the light and heavy chain variable domains for the two antigen binding domains (i.e. the light and heavy chain variable domains for the antigen binding domain capable of binding PD-1 , and the light and heavy chain variable domains for the antigen binding domain capable of binding to another target protein), and including sequences encoding a suitable linker or dimerization domain between the antigen binding domains can be prepared by molecular cloning techniques. Recombinant bispecific antibody can thereafter be produced by expression (e.g. in vitro) of the construct in a suitable host cell (e.g. a mammalian host cell), and expressed recombinant bispecific antibody can then optionally be purified.
Antibodies may be produced by a process of affinity maturation in which a modified antibody is generated that has an improvement in the affinity of the antibody for antigen, compared to an unmodified parent antibody. Affinity-matured antibodies may be produced by procedures known in the art, e.g., Marks et ai, Rio/Technology 10:779-783 (1992);
Barbas et al. Proc Nat. Acad. Sci. USA 91 :3809-3813 (1994); Schier et al. Gene 169: 147- 155 (1995); Yelton et al. J. Immunol. 155: 1994-2004 (1995); Jackson et al., J. Immunol. 154(7):331 0-15 9 (1995); and Hawkins et al, J. Mol. Biol. 226:889-896 (1992). Antibodies according to the present invention preferably exhibit specific binding to PD-1. An antibody that specifically binds to a target molecule preferably binds the target with greater affinity, and/or with greater duration than it binds to other targets. In one embodiment, the extent of binding of an antibody to an unrelated target is less than about 10% of the binding of the antibody to the target as measured, e.g., by ELISA, or by a radioimmunoassay (RIA). Alternatively, the binding specificity may be reflected in terms of binding affinity where the anti-PD-1 antibody of the present invention binds to PD-1 with a KD that is at least 0.1 order of magnitude (i.e. 0.1 x 10", where n is an integer
representing the order of magnitude) greater than the KD of the antibody towards another target molecule, e.g. another member of the CD28 family. This may optionally be one of at least 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.5, or 2.0.
Antibodies according to the present invention preferably have a dissociation constant (KD) of one of≤ 1 μΜ,≤ 100nM,≤10nM,≤1 nM or≤100pM. Binding affinity of an antibody for its target is often described in terms of its dissociation constant (KD). Binding affinity can be measured by methods known in the art, such as by Surface Plasmon Resonance, or by a radiolabeled antigen binding assay (RIA) performed with the Fab version of the antibody and antigen molecule.
Antibodies according to the present invention may be "antagonist" antibodies that inhibit or reduce a biological activity of the antigen to which it binds. Blocking of PD-1 assists in the restoration of T-cell function by inhibiting the immune-inhibitory signalling pathway mediated by PD-1.
In some aspects, the antibody is clone A3, or a variant of A3. A3 comprises the following CDR sequences:
Light chain:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDVLYGSV (SEQ ID NO:27)
Heavy chain:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGKDH (SEQ ID NO:41 )
CDR sequences determined by Kabat definition.
In some aspects, the antibody is clone A10, or a variant of A10. A10 comprises the following CDR sequences:
Light chain:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDYYYGTI (SEQ ID NO:28)
Heavy chain:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGKDV (SEQ ID NO:42)
CDR sequences determined by Kabat definition. In some aspects, the antibody is clone B6, or a variant of B6. B6 comprises the following CDR sequences:
Light chain:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDYLRGTV (SEQ ID NO:29)
Heavy chain:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DYGAGPYYYGMDV (SEQ ID NO:43)
CDR sequences determined by Kabat definition.
In some aspects, the antibody is clone C4, or a variant of C4. C4 comprises the following CDR sequences:
Light chain:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: SAWDDYLHGTV (SEQ ID NO:30)
Heavy chain:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGLDV (SEQ ID NO:44)
CDR sequences determined by Kabat definition.
In some aspects, the antibody is clone D4, or a variant of D4. D4comprises the following CDR sequences:
Light chain:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDYVRGTM (SEQ ID NO:31 )
Heavy chain: HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:45)
CDR sequences determined by Kabat definition.
In some aspects, the antibody is clone E1 , or a variant of E1. E1 comprises the following CDR sequences:
Light chain:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: SSWDDFLRGTV (SEQ ID NO:32)
chain:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:46)
CDR sequences determined by Kabat definition.
In some aspects, the antibody is clone F2, or a variant of F2. F2 comprises the following CDR sequences:
Light chain:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: SSWDDDARGTI (SEQ ID NO:33)
Heavy chain:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:47)
CDR sequences determined by Kabat definition.
In some aspects, the antibody is clone G1 , or a variant of G1. G1 comprises the following CDR sequences: Light chain:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: AAWDDVYYGTI (SEQ ID NO:34)
Heavy chain:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:48)
CDR sequences determined by Kabat definition
In some aspects, the antibody is clone G2, or a variant of G2. G2 comprises the following CDR sequences:
Light chain:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDSLYGTV (SEQ ID NO:35)
Heavy chain:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:49)
CDR sequences determined by Kabat definition.
In some aspects, the antibody is clone G10, or a variant of G10. G10 comprises the following CDR sequences:
Light chain:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: AAWDDAYYGTI (SEQ ID NO:36)
Heavy chain:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89) HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DYGAGPYYYGMDV (SEQ ID NO:50)
CDR sequences determined by Kabat definition.
In some aspects, the antibody is clone H4, or a variant of H4. H4 comprises the following CDR sequences:
Light chain:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDVYRGTV (SEQ ID NO:37)
Heavy chain:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGSGYYLYGMDV (SEQ ID NO:51 )
CDR sequences determined by Kabat definition.
In some aspects, the antibody is clone H9, or a variant of H9. H9 comprises the following CDR sequences:
Light chain:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: SSWDDSLYGTI (SEQ ID NO:38)
Heavy chain:
HC-CDR1 : GFTFSSYGMH (SEQ ID NO:39) or
SYGMH (SEQ ID NO: 89)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:52)
CDR sequences determined by Kabat definition.
Antibodies according to the present invention may comprise the CDRs of one of A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4, or H9 or one of SEQ ID NOs 1 and 13, 2 and 14, 3 and 15, 4 and 16, 5 and 17, 6 and 18, 7 and 19, 8 and 20, 9 and 21 , 10 and 22, 11 and 23, or 12 and 24. In an antibody according to the present invention one or two or three or four of the six CDR sequences may vary. A variant may have one or two amino acid substitutions in one or two of the six CDR sequences.
Amino acid sequences of the VH and VL chains of ant-PD-1 clones are shown in Figures 1 and 2. The encoding nucleotide sequences are shown in Figure 4.
The light and heavy chain CDRs may also be particularly useful in conjunction with a number of different framework regions. Accordingly, light and/or heavy chains having LC- CDR1-3 or HC-CDR1-3 may possess an alternative framework region. Suitable framework regions are well known in the art and are described for example in M. Lefranc & G. Lefranc (2001) "The Immunoglobulin FactsBook", Academic Press, incorporated herein by reference.
In this specification, antibodies may have VH and/or VL chains comprising an amino acid sequence that has a high percentage sequence identity to one or more of the VH and/or VL amino acid sequences of SEQ ID Nos 1 and 13, 2 and 14, 3 and 15, 4 and 16, 5 and 17, 6 and 18, 7 and 19, 8 and 20, 9 and 21 , 10 and 22, 1 1 and 23, or 12 and 24 respectively, or to one or the amino acid sequences shown in Figures 1 and 2. For example, antibodies according to the present invention include antibodies that bind PD-1 and have a VH or VL chain that comprises an amino acid sequence having at least 70%, more preferably one of at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, sequence identity to the VH or VL chain amino acid sequence of one of SEQ ID NOs 1 to 24 or to one or the amino acid sequences shown in Figures 1 and 2.
Antibodies according to the present invention may be detectably labelled or, at least, capable of detection. For example, the antibody may be labelled with a radioactive atom or a coloured molecule or a fluorescent molecule or a molecule which can be readily detected in any other way. Suitable detectable molecules include fluorescent proteins, luciferase, enzyme substrates, and radiolabels. The binding moiety may be directly labelled with a detectable label or it may be indirectly labelled. For example, the binding moiety may be an unlabelled antibody which can be detected by another antibody which is itself labelled. Alternatively, the second antibody may have bound to it biotin and binding of labelled streptavidin to the biotin is used to indirectly label the first antibody. Methods of detection
Antibodies, or antigen binding fragments, described herein may be used in methods that involve the binding of the antibody or antigen binding fragment to PD-1. Such methods may involve detection of the bound complex of antibody, or antigen binding fragment, and PD-1. As such, in one embodiment a method is provided, the method comprising contacting a sample containing, or suspected to contain, PD-1 with an antibody or antigen binding fragment as described herein and detecting the formation of a complex of antibody, or antigen binding fragment, and PD-1. Suitable method formats are well known in the art, including immunoassays such as sandwich assays, e.g. ELISA. The method may involve labelling the antibody, or antigen binding fragment, or PD-1 , or both, with a detectable label, e.g. fluorescent, luminescent or radio- label. Methods of this kind may provide the basis of a method of diagnosis of a disease or condition requiring detection and or quantitation of PD-1. Such methods may be performed in vitro on a patient sample, or following processing of a patient sample. Once the sample is collected, the patient is not required to be present for the in vitro method of diagnosis to be performed and therefore the method may be one which is not practised on the human or animal body.
Such methods may involve determining the amount of PD-1 present in a patient sample. The method may further comprise comparing the determined amount against a standard or reference value as part of the process of reaching a diagnosis. Other diagnostic tests may be used in conjunction with those described here to enhance the accuracy of the diagnosis or prognosis or to confirm a result obtained by using the tests described here.
The level of PD-1 present in a patient sample may be indicative that a patient may respond to treatment with an anti-PD1 antibody. The presence of a high level of PD-1 in a sample may be used to select a patient for treatment with an anti-PD1 antibody. The antibodies of the present invention may therefore be used to select a patient for treatment with anti-PD-1 therapy.
Detection in a sample of PD-1 may be used for the purpose of diagnosis of a T-cell dysfunctional disorder or a cancerous condition in the patient, diagnosis of a
predisposition to a cancerous condition or for providing a prognosis (prognosticating) of a cancerous condition. The diagnosis or prognosis may relate to an existing (previously diagnosed) cancerous condition, which may be benign or malignant, may relate to a suspected cancerous condition or may relate to the screening for cancerous conditions in the patient (which may be previously undiagnosed).
In one embodiment the level of PD-1 expression on CD8+ T cells may be detected in order to indicate the degree of T-cell exhaustion and severity of the disease state.
A sample may be taken from any tissue or bodily fluid. The sample may comprise or may be derived from: a quantity of blood; a quantity of serum derived from the individual's blood which may comprise the fluid portion of the blood obtained after removal of the fibrin clot and blood cells; a tissue sample or biopsy; or cells isolated from said individual.
Methods according to the present invention are preferably performed in vitro. The term "in vitro" is intended to encompass experiments with cells in culture whereas the term "in vivo" is intended to encompass experiments with intact multi-cellular organisms.
Therapeutic applications
Antibodies, antigen binding fragments and polypeptides according to the present invention and compositions comprising such agents may be provided for use in methods of medical treatment. Treatment may be provided to subjects having a disease or condition in need of treatment. The disease or condition may be one of a T-cell dysfunctional disorder, including a T-cell dysfunctional disorder associated with a cancer, or a cancer, or a T-cell dysfunctional disorder associated with an infection, or an infection.
A T-cell dysfunctional disorder may be a disease or condition in which normal T-cell function is impaired causing downregulation of the subject's immune response to pathogenic antigens, e.g. generated by infection by exogenous agents such as microorganisms, bacteria and viruses, or generated by the host in some disease states such as in some forms of cancer (e.g. in the form of tumor associated antigens).
The T-cell dysfunctional disorder may comprise T-cell exhaustion or T-cell anergy. T-cell exhaustion comprises a state in which CD8+ T-cells fail to proliferate or exert T-cell effector functions such as cytotoxicity and cytokine (e.g. IFNv) secretion in response to antigen stimulation. Exhausted T-cells may also be characterised by sustained expression of PD-1 , where blockade of PD-1 :PD-L1 interactions may reverse the T-cell exhaustion and restore antigen-specific T cell responses.
The T-cell dysfunctional disorder may be manifest as an infection, or inability to mount an effective immune response against an infection. The infection may be chronic, persistent, latent or slow, and may be the result of bacterial, viral, fungal or parasitic infection. As such, treatment may be provided to patients having a bacterial, viral or fungal infection. Examples of bacterial infections include infection with Helicobacter pylori. Examples of viral infections include infection with HIV, hepatitis B or hepatitis C.
The T-cell dysfunctional disorder may be associated with a cancer, such as tumor immune escape. Many human tumors express tumor-associated antigens recognised by T cells and capable of inducing an immune response. However, immune evasion is common and is believed to be mediated by a number of soluble factors, including PD-L1. As such, blocking the interaction of PD-1 and PD-L1 may inhibit this negative
immunoregulatory signal to tumor cells and enhance tumor-specific CD8+ T-cell immunity.
Cancers may also be treated where there is no indication of a T-cell dysfunctional disorder such as T-cell exhaustion but the use of an antibody, antigen binding fragment or polypeptide according to the present invention allows the subject to suppress PD-1 signalling and mount an effective immune response with limited impairment, evasion or induction of tumor immune escape. In such treatments, the antibody, antigen binding fragment or polypeptide may provide a treatment for cancer that involves prevention of the development of tumor immune escape.
The treatment may be aimed at prevention of the T-cell dysfunctional disorder, e.g.
prevention of infection or of the development or progression of a cancer. As such, the antibodies, antigen binding fragments and polypeptides may be used to formulate pharmaceutical compositions or medicaments and subjects may be prophylactically treated against development of a disease state. This may take place before the onset of symptoms of the disease state, and/or may be given to subjects considered to be at greater risk of infection or development of cancer.
Treatment may comprise co-therapy with a vaccine, e.g. T-cell vaccine, which may involve simultaneous, separate or sequential therapy, or combined administration of vaccine and antibody, antigen binding fragment or polypeptide in a single composition. In this context, the antibody, antigen binding fragment or polypeptide may be provided as an adjuvant to the vaccine. Limited proliferative potential of exhausted T cells has been attributed as a main reason for failure of T-cell immunotherapy and combination an agent capable of blocking or reversing T cell exhaustion is a potential strategy for improving the efficacy of T-cell immunotherapy (Barber et al., Nature Vol 439, No. 9 p682-687 Feb 2006).
Administration of an antibody, antigen binding fragment or polypeptide is preferably in a "therapeutically effective amount", this being sufficient to show benefit to the individual. The actual amount administered, and rate and time-course of administration, will depend on the nature and severity of the disease being treated. Prescription of treatment, e.g. decisions on dosage etc., is within the responsibility of general practitioners and other medical doctors, and typically takes account of the disorder to be treated, the condition of the individual patient, the site of delivery, the method of administration and other factors known to practitioners. Examples of the techniques and protocols mentioned above can be found in Remington's Pharmaceutical Sciences, 20th Edition, 2000, pub. Lippincott, Williams & Wilkins.
Formulating pharmaceutically useful compositions and medicaments
Antibodies, antigen binding fragments and polypeptides according to the present invention may be formulated as pharmaceutical compositions for clinical use and may comprise a pharmaceutically acceptable carrier, diluent, excipient or adjuvant.
In accordance with the present invention methods are also provided for the production of pharmaceutically useful compositions, such methods of production may comprise one or more steps selected from: isolating an antibody, antigen binding fragment or polypeptide as described herein; and/or mixing an isolated antibody, antigen binding fragment or polypeptide as described herein with a pharmaceutically acceptable carrier, adjuvant, excipient or diluent.
For example, a further aspect of the present invention relates to a method of formulating or producing a medicament or pharmaceutical composition for use in the treatment of a T- cell dysfunctional disorder, the method comprising formulating a pharmaceutical composition or medicament by mixing an antibody, antigen binding fragment or polypeptide as described herein with a pharmaceutically acceptable carrier, adjuvant, excipient or diluent. Infection
An infection may be any infection or infectious disease, e.g. bacterial, viral, fungal, or parasitic infection. In some embodiments it may be particularly desirable to treat chronic/persistent infections, e.g. where such infections are associated with T cell dysfunction or T cell exhaustion.
It is well established that T cell exhaustion is a state of T cell dysfunction that arises during many chronic infections (including viral, bacterial and parasitic), as well as in cancer (Wherry Nature Immunology Vol.12, No.6, p492-499, June 201 1).
An infection or infectious disease may be one in which PD-1 is upregulated (e.g. as reported by Radziewicz H, et al., J Virol. 2007;81 (6):2545-2553 and Golden-Mason L et al., J Virol. 2007;81 (17):9249-9258).
Examples of bacterial infections that may be treated include infection by Bacillus spp., Bordetella pertussis, Clostridium spp., Corynebacterium spp., Vibrio chloerae,
Staphylococcus spp., Streptococcus spp. Escherichia, Klebsiella, Proteus, Yersinia, Erwina, Salmonella, Listeria sp, Helicobacter pylori, mycobacteria (e.g. Mycobacterium tuberculosis) and Pseudomonas aeruginosa. For example, the bacterial infection may be sepsis or tuberculosis.
Yao et al (PD-1 on dendritic cells impedes innate immunity against bacterial infection. Blood 1 13(23): 5811 -5818 Jun 4 2009) established PD-1 in the negative regulation of DC function during innate immune response to infection by Listeria monocytogenes.
Brahmamdam et al (Delayed administration of anti-PD-1 antibody reverses immune dysfunction and improves survival during sepsis. Journal of Leukocyte Biology vo.88, no.2 233-240, August 2010) reported that anti-PD-1 antibody administered 24 h after sepsis prevented sepsis-induced depletion of lymphocytes and DCs, increased Bcl-xL, blocked apoptosis and improved survival. Tim3:Galectin-9 interactions have been reported to mediate T cell exhaustion and mediate the innate and adaptive immune response to infection by Mycobacterium tuberculosis (Jayaraman et al., The Journal of Immunology 2012, 188, 70.6). Examples of viral infections that may be treated include infection by influenza virus, measles virus, hepatitis B virus (HBV), hepatitis C virus (HCV), human immunodeficiency virus (HIV), lymphocytic choriomeningitis virus (LCMV), Herpes simplex virus and human papilloma virus.
Chronic viral infections, such as those caused by HCV, HBV, and HIV commonly involve mechanisms to evade immune clearance. Expression of PD-1 and TIM-3 have been identified as correlating with defective T cell responses to hepatitis C virus (HCV) (McMahan et al., The Journal of Clinical Investigation Vol. 120, No. 12 p4546-4557, December 2010). In HCV, McMahan et al (supra) found that the level of dual TIM-3 and PD-1 expression on HCV-specific CTLs predated the development of viral persistence, providing prognostic information. Barber et al. (Nature Vol 439, No. 9 p682-687 Feb 2006) reported that PD-1 is upregulated during chronic viral infection. In mice infected with LCMV they reported that blockade of the PD-1/PD-L1 inhibitory pathway had a beneficial effect on CD8 T cells, restoring their ability to undergo proliferation, secrete cytokines, kill infected cells and decrease viral load. PD-1 is also upregulated in HIV infection (Said et al., Nature Medicine Vol. 16, No.4 p452-460 April 2010). Blocking interaction between PD-1 and PD-L1 contributed to viral clearance and improved T cell function in animal models of chronic viral infection (Said et al., supra).
Examples of fungal infections that may be treated include infection by Alternaria sp, Aspergillus sp, Candida sp and Histoplasma sp. The fungal infection may be fungal sepsis or histoplasmosis.
Chang et al (Blockade of the negative co-stimulatory molecules PD-1 and CTLA-4 improves survival in primary and secondary fungal sepsis. Critical Care 2013, 17:R85) reported that anti-PD1 antibodies were highly effective at improving survival in primary and secondary fungal sepsis. Lazar-Molnar et al (The PD-1/PD-L costimulatory pathway critically affects host resistance to the pathogenic fungus Histoplasma capsulatum PNAS vol. 105, no.7, p2658-2663, 19 Feb 2008) reported that anti-PD-1 antibody significantly increased survival of mice infected with Histoplasma capsulatum. As such, the importance of T cell exhaustion in mediating fungal infection is well established.
Examples of parasitic infections that may be treated include infection by Plasmodium species (e.g. Plasmodium falciparum, Plasmodium yoeli, Plasmodium ovale, Plasmodium vivax, or Plasmodium chabaudi chabaudi). The parasitic infection may be a disease such as malaria, leishmaniasis and toxoplasmosis. Infection of humans with Plasmodium falciparum has been shown to result in higher expression of PD-1 and T cell exhaustion mice (Butler et al., Nature Immunology ' Vol.13, No.12, p 188-195 February 2012). Blockade of PD-L1 and LAG-3 using anti-PD-L1 and anti-LAG-3 monoclonal antibodies in vivo contributed to the restoration of CD4+ T-cell function, amplification of the number of follicular helper T cells, germinal-center B cells and plasmablasts, enhanced protective antibodies and rapidly cleared blood-stage malaria in mice. It was also shown to block the development of chronic infection (Butler et al., supra). Cancer
A cancer may be any unwanted cell proliferation (or any disease manifesting itself by unwanted cell proliferation), neoplasm or tumor or increased risk of or predisposition to the unwanted cell proliferation, neoplasm or tumor. The cancer may be benign or malignant and may be primary or secondary (metastatic). A neoplasm or tumor may be any abnormal growth or proliferation of cells and may be located in any tissue. Examples of tissues include the adrenal gland, adrenal medulla, anus, appendix, bladder, blood, bone, bone marrow, brain, breast, cecum, central nervous system (including or excluding the brain) cerebellum, cervix, colon, duodenum, endometrium, epithelial cells (e.g. renal epithelia), gallbladder, oesophagus, glial cells, heart, ileum, jejunum, kidney, lacrimal glad, larynx, liver, lung, lymph, lymph node, lymphoblast, maxilla, mediastinum, mesentery, myometrium, nasopharynx, omentume, oral cavity, ovary, pancreas, parotid gland, peripheral nervous system, peritoneum, pleura, prostate, salivary gland, sigmoid colon, skin, small intestine, soft tissues, spleen, stomach, testis, thymus, thyroid gland, tongue, tonsil, trachea, uterus, vulva, white blood cells.
Tumors to be treated may be nervous or non-nervous system tumors. Nervous system tumors may originate either in the central or peripheral nervous system, e.g. glioma, medulloblastoma, meningioma, neurofibroma, ependymoma, Schwannoma,
neurofibrosarcoma, astrocytoma and oligodendroglioma. Non-nervous system
cancers/tumors may originate in any other non-nervous tissue, examples include melanoma, mesothelioma, lymphoma, myeloma, leukemia, Non-Hodgkin's lymphoma (NHL), Hodgkin's lymphoma, chronic myelogenous leukemia (CML), acute myeloid leukemia (AML), myelodysplasia syndrome (MDS), cutaneous T-cell lymphoma (CTCL), chronic lymphocytic leukemia (CLL), hepatoma, epidermoid carcinoma, prostate carcinoma, breast cancer, lung cancer , colon cancer, ovarian cancer, pancreatic cancer, thymic carcinoma, NSCLC, haematologic cancer and sarcoma. In some embodiments, the cancer is one or more of lung cancer, renal cancer and bladder cancer. Adoptive T cell transfer therapy
Adoptive T cell transfer therapy generally refers to a process in which white blood cells are removed from a subject, typically by drawing a blood sample from which white blood cells are separated, expanded in vitro or ex vivo and returned either to the same subject or to a different subject. The treatment is typically aimed at increasing the
amount/concentration of an active form of the required T cell population in the subject. Such treatment may be beneficial in subjects experiencing T cell exhaustion.
Antibodies capable of blocking the mechanism of T cell exhaustion, or reversing it, provide a means of enhancing T cell activity and promoting T cell expansion.
Accordingly, in a further aspect of the present invention a method is provided for expanding a population of T cells, wherein T cells are contacted in vitro or ex vivo with an antibody, antigen binding fragment or polypeptide according to the present invention. The method may optionally comprise one or more of the following steps: taking a blood sample from a subject; isolating T cells from the blood sample; culturing the T cells in in vitro or ex vivo cell culture (where they may be contacted with the antibody, antigen binding fragment or polypeptide), collecting an expanded population of T cells; mixing the T cells with an adjuvant, diluent, or carrier; administering the expanded T cells to a subject.
Accordingly, in some aspects of the present invention a method of treatment of a subject having a T-cell dysfunctional disorder is provided, the method comprising obtaining a blood sample from a subject in need of treatment, culturing T cells obtained from the blood sample in the presence of an antibody, antigen binding fragment or polypeptide according to the present invention so as to expand the T cell population, collecting expanded T cells, and administering the expanded T cells to a subject in need of treatment. The T cells may be obtained from a subject requiring treatment, and may be isolated and/or purified. They may be a CD4+ and/or CD8+ T-cell population. The T-cells may represent a population experiencing T cell exhaustion and may optionally have upregulated expression of PD-1.
During culture, T cells may be contacted with the antibody, antigen binding fragment or polypeptide under conditions and for a period of time suitable to allow expansion of the T cells to a desired number of cells. After a suitable period of time the T cells may be harvested, optionally concentrated, and may be mixed with a suitable carrier, adjuvant or diluent and returned to the subject's body. A subject may undergo one or more rounds of such therapy.
Methods of T cell expansion are well known in the art, such as those described in Kalamasz et al., J Immunother 2004 Sep-Oct; 27(5):405-18; Monies et al., Clin Exp Immunol 2005 Nov;142(2):292-302; Wolfl and Greenburg Nature Protocols 9 p950-966 27 March 2014; Trickett and Kwan Journal of Immunological Methods Vol. 275, Issues 1- 2, 1 April 2003, p251-255; Butler et al PLoSONE l^) 12 Jan 2012.
Simultaneous or Sequential Administration
Compositions may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated.
In this specification an antibody, antigen binding fragment or polypeptide of the present invention and an anti-infective agent or chemotherapeutic agent (therapeutic agent) may be administered simultaneously or sequentially. In some embodiments, treatment with an antibody, antigen binding fragment or polypeptide of the present invention may be accompanied by chemotherapy.
Simultaneous administration refers to administration of the antibody, antigen binding fragment or polypeptide and therapeutic agent together, for example as a pharmaceutical composition containing both agents (combined preparation), or immediately after each other and optionally via the same route of administration, e.g. to the same artery, vein or other blood vessel.
Sequential administration refers to administration of one of the antibody, antigen binding fragment or polypeptide or therapeutic agent followed after a given time interval by separate administration of the other agent. It is not required that the two agents are administered by the same route, although this is the case in some embodiments. The time interval may be any time interval.
Anti-infective agents
In treating infection, an antibody, antigen binding fragment or polypeptide of the present invention may be administered in combination with an anti-infective agent, as described above. The anti-infective agent may be an agent known to have action against the microorganism or virus responsible for the infection. Suitable anti-infective agents include antibiotics (such as penicillins, cephalosporins, rifamycins, lipiarmycins, quinolones, sulfonamides, macrolides, lincosamides, tetracyclines, cyclic lipopeptides, glycylcyclines, oxazolidinones, and lipiarmycins), antiviral agents (such as reverse transcriptase inhibitors, integrase inhibitors, transcription factor inhibitors, antisense and siRNA agents and protease inhibitors), anti-fungal agents (such as polyenes, imidiazoles, triazoles, thiazoles, allylamines, and echinocandins) and anti-parasitic agents (such as antinematode agents, anticestode agents, antitrematode agents, antiamoebic agents and antiprotozoal agents).
Chemotherapy
Chemotherapy refers to treatment of a cancer with a drug or with ionising radiation (e.g. radiotherapy using X-rays or γ-rays). In preferred embodiments chemotherapy refers to treatment with a drug. The drug may be a chemical entity, e.g. small molecule pharmaceutical, antibiotic, DNA intercalator, protein inhibitor (e.g. kinase inhibitor), or a biological agent, e.g. antibody, antibody fragment, nucleic acid or peptide aptamer, nucleic acid (e.g. DNA, RNA), peptide, polypeptide, or protein. The drug may be formulated as a pharmaceutical composition or medicament. The formulation may comprise one or more drugs (e.g. one or more active agents) together with one or more pharmaceutically acceptable diluents, excipients or carriers. A treatment may involve administration of more than one drug. A drug may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated. For example, the chemotherapy may be a co-therapy involving administration of two drugs, one or more of which may be intended to treat the cancer. The chemotherapy may be administered by one or more routes of administration, e.g. parenteral, intravenous injection, oral, subcutaneous, intradermal or intratumoral.
The chemotherapy may be administered according to a treatment regime. The treatment regime may be a pre-determined timetable, plan, scheme or schedule of chemotherapy administration which may be prepared by a physician or medical practitioner and may be tailored to suit the patient requiring treatment.
The treatment regime may indicate one or more of: the type of chemotherapy to administer to the patient; the dose of each drug or radiation; the time interval between administrations; the length of each treatment; the number and nature of any treatment holidays, if any etc. For a co-therapy a single treatment regime may be provided which indicates how each drug is to be administered. Chemotherapeutic drugs and biologies may be selected from:
• alkylating agents such as cisplatin, carboplatin, mechlorethamine,
cyclophosphamide, chlorambucil, ifosfamide;
• purine or pyrimidine anti-metabolites such as azathiopurine or mercaptopurine;
• alkaloids and terpenoids, such as vinca alkaloids (e.g. vincristine, vinblastine, vinorelbine, vindesine), podophyllotoxin, etoposide, teniposide, taxanes such as paclitaxel (Taxol™), docetaxel;
• topoisomerase inhibitors such as the type I topoisomerase inhibitors
camptothecins irinotecan and topotecan, or the type II topoisomerase inhibitors amsacrine, etoposide, etoposide phosphate, teniposide;
· antitumor antibiotics (e.g. anthracyline antibiotics) such as dactinomycin,
doxorubicin (Adriamycin™), epirubicin, bleomycin, rapamycin;
• antibody based agents, such as anti-TIM-3 antibodies, anti-CTLA-4, anti-LAG-3, anti-4-I BB, anti-GITR, anti-CD27, anti-BLTA, anti-OX40, anti-VEGF, anti-TNFa, anti-IL-2, antiGpllb/llla, anti-CD-52, anti-CD20, anti-RSV, anti-HER2/neu(erbB2), anti-TNF receptor, anti-EGFR antibodies, monoclonal antibodies or antibody fragments, examples include: cetuximab, panitumumab, infliximab, basiliximab, bevacizumab (Avastin®), abciximab, daclizumab, gemtuzumab, alemtuzumab, rituximab (Mabthera®), palivizumab, trastuzumab, etanercept, adalimumab, nimotuzumab
• EGFR inihibitors such as erlotinib, cetuximab and gefitinib • anti-angiogenic agents such as bevacizumab (Avastin®)
• cancer vaccines such as Sipuleucel-T (Provenge®)
In one embodiment the chemotherapeutic agent is an anti-TIM-3 antibody, anti-CTLA-4, anti-LAG3, anti-41 BB, anti-GITR, anti-CD27, anti-BLTA, anti-OX40, anti-VEGF, anti- TNFa, anti-IL2, anti-Gpllb/llla, anti-CD-52, anti-CD20, anti-RSV, anti-HER2/neu(erbB2), anti-TNF receptor, anti-EGFR or other antibody. In some embodiments, the
chemotherapeutic agent is an immune checkpoint inhibitor or costimulation molecule.
Further chemotherapeutic drugs may be selected from: 13-cis-Retinoic Acid, 2- Chlorodeoxyadenosine, 5-Azacitidine 5-Fluorouracil, 6-Mercaptopurine, 6-Thioguanine, Abraxane, Accutane®, Actinomycin-D Adriamycin®, Adrucil®, Afinitor®, Agrylin®, Ala- Cort®, Aldesleukin, Alemtuzumab, ALIMTA, Alitretinoin, Alkaban-AQ®, Alkeran®, All- transretinoic Acid, Alpha Interferon, Altretamine, Amethopterin, Amifostine,
Aminoglutethimide, Anagrelide, Anandron®, Anastrozole, Arabinosylcytosine, Aranesp®, Aredia®, Arimidex®, Aromasin®, Arranon®, Arsenic Trioxide, Asparaginase, ATRA Avastin®, Azacitidine, BCG, BCNU, Bendamustine, Bevacizumab, Bexarotene,
BEXXAR®, Bicalutamide, BiCNU, Blenoxane®, Bleomycin, Bortezomib, Busulfan, Busulfex®, Calcium Leucovorin, Campath®, Camptosar®, Camptothecin-11 ,
Capecitabine, Carac™, Carboplatin, Carmustine, Casodex®, CC-5013, CCI-779, CCNU, CDDP, CeeNU, Cerubidine®, Cetuximab, Chlorambucil, Cisplatin, Citrovorum Factor,
Cladribine, Cortisone, Cosmegen®, CPT-11 , Cyclophosphamide, Cytadren®, Cytarabine Cytosar-U®, Cytoxan®, Dacogen, Dactinomycin, Darbepoetin Alfa, Dasatinib,
Daunomycin, Daunorubicin, Daunorubicin Hydrochloride, Daunorubicin Liposomal, DaunoXome®, Decadron, Decitabine, Delta-Cortef®, Deltasone®, Denileukin, Diftitox, DepoCyt™, Dexamethasone, Dexamethasone Acetate, Dexamethasone Sodium
Phosphate, Dexasone, Dexrazoxane, DHAD, DIC, Diodex, Docetaxel, Doxil®,
Doxorubicin, Doxorubicin Liposomal, Droxia™, DTIC, DTIC-Dome®, Duralone®, Eligard™, Ellence™, Eloxatin™, Elspar®, Emcyt®, Epirubicin, Epoetin Alfa, Erbitux, Erlotinib, Erwinia L-asparaginase, Estramustine, Ethyol Etopophos®, Etoposide, Etoposide Phosphate, Eulexin®, Everolimus, Evista®, Exemestane, Faslodex®,
Femara®, Filgrastim, Floxuridine, Fludara®, Fludarabine, Fluoroplex®, Fluorouracil, Fluoxymesterone, Flutamide, Folinic Acid, FUDR®, Fulvestrant, Gefitinib, Gemcitabine, Gemtuzumab ozogamicin, Gleevec™, Gliadel® Wafer, Goserelin, Granulocyte - Colony Stimulating Factor, Granulocyte Macrophage Colony Stimulating Factor, Herceptin ®, Hexadrol, Hexalen®, Hexamethylmelamine, HMM, Hycamtin®, Hydrea®, Hydrocort Acetate®, Hydrocortisone, Hydrocortisone Sodium Phosphate, Hydrocortisone Sodium Succinate, Hydrocortone Phosphate, Hydroxyurea, Ibritumomab, Ibritumomab Tiuxetan, Idamycin®, Idarubicin, Ifex®, IFN-alpha, Ifosfamide, IL-11 , IL-2, Imatinib mesylate, Imidazole Carboxamide, Interferon alfa, Interferon Alfa-2b (PEG Conjugate), Interleukin - 2, lnterleukin-11 , Intron A® (interferon alfa-2b), Iressa®, Irinotecan, Isotretinoin,
Ixabepilone, Ixempra™, Kidrolase, Lanacort®, Lapatinib, L-asparaginase, LCR,
Lenalidomide, Letrozole, Leucovorin, Leukeran, Leukine™, Leuprolide, Leurocristine, Leustatin™, Liposomal Ara-C, Liquid Pred®, Lomustine, L-PAM, L-Sarcolysin, Lupron®, Lupron Depot®, Matulane®, Maxidex, Mechlorethamine, Mechlorethamine Hydrochloride, Medralone®, Medrol®, Megace®, Megestrol, Megestrol Acetate, Melphalan,
Mercaptopurine, Mesna, Mesnex™, Methotrexate, Methotrexate Sodium,
Methylprednisolone, Meticorten®, Mitomycin, Mitomycin-C, Mitoxantrone, M-Prednisol®, MTC, MTX, Mustargen®, Mustine, Mutamycin®, Myleran®, Mylocel™, Mylotarg®, Navelbine®, Nelarabine, Neosar®, Neulasta™, Neumega®, Neupogen®, Nexavar®, Nilandron®, Nilutamide, Nipent®, Nitrogen Mustard, Novaldex®, Novantrone®,
Octreotide, Octreotide acetate, Oncospar®, Oncovin®, Ontak®, Onxal™, Oprevelkin, Orapred®, Orasone®, Oxaliplatin, Paclitaxel, Paclitaxel Protein-bound, Pamidronate, Panitumumab, Panretin®, Paraplatin®, Pediapred®, PEG Interferon, Pegaspargase, Pegfilgrastim, PEG-INTRON™, PEG-L-asparaginase, PEMETREXED, Pentostatin, Phenylalanine Mustard, Platinol®, Platinol-AQ®, Prednisolone, Prednisone, Prelone®, Procarbazine, PROCRIT®, Proleukin®, Prolifeprospan 20 with Carmustine Implant Purinethol®, Raloxifene, Revlimid®, Rheumatrex®, Rituxan®, Rituximab, Roferon-A® (Interferon Alfa-2a), Rubex®, Rubidomycin hydrochloride, Sandostatin® Sandostatin LAR®, Sargramostim, Solu-Cortef®, Solu-Medrol®, Sorafenib, SPRYCEL™, STI-571 , Streptozocin, SU11248, Sunitinib, Sutent®, Tamoxifen, Tarceva®, Targretin®, Taxol®, Taxotere®, Temodar®, Temozolomide, Temsirolimus, Teniposide, TESPA, Thalidomide, Thalomid®, TheraCys®, Thioguanine, Thioguanine Tabloid®, Thiophosphoamide, Thioplex®, Thiotepa, TICE®, Toposar®, Topotecan, Toremifene, Torisel®, Tositumomab, Trastuzumab, Treanda®, Tretinoin, Trexall™, Trisenox®, TSPA, TYKERB®, VCR, Vectibix™, Velban®, Velcade®, VePesid®, Vesanoid®, Viadur™, Vidaza®, Vinblastine, Vinblastine Sulfate, Vincasar Pfs®, Vincristine, Vinorelbine, Vinorelbine tartrate, VLB, VM-26, Vorinostat, VP-16, Vumon®, Xeloda®, Zanosar®, Zevalin™, Zinecard®,
Zoladex®, Zoledronic acid, Zolinza, Zometa®. Routes of administration Antibodies, antigen binding fragments, polypeptides and other therapeutic agents, medicaments and pharmaceutical compositions according to aspects of the present invention may be formulated for administration by a number of routes, including but not limited to, parenteral, intravenous, intra-arterial, intramuscular, subcutaneous, intradermal, intratumoral and oral. Antibodies, antigen binding fragments, polypeptides and other therapeutic agents, may be formulated in fluid or solid form. Fluid formulations may be formulated for administration by injection to a selected region of the human or animal body. Dosage regime
Multiple doses of the antibody, antigen binding fragment or polypeptide may be provided. One or more, or each, of the doses may be accompanied by simultaneous or sequential administration of another therapeutic agent. Multiple doses may be separated by a predetermined time interval, which may be selected to be one of 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, or 31 days, or 1 , 2, 3, 4, 5, or 6 months. By way of example, doses may be given once every 7, 14, 21 or 28 days (plus or minus 3, 2, or 1 days).
Kits
In some aspects of the present invention a kit of parts is provided. In some embodiments the kit may have at least one container having a predetermined quantity of the antibody, antigen binding fragment or polypeptide. The kit may provide the antibody, antigen binding fragment or polypeptide in the form of a medicament or pharmaceutical composition, and may be provided together with instructions for administration to a patient in order to treat a specified disease or condition. The antibody, antigen binding fragment or polypeptide may be formulated so as to be suitable for injection or infusion to a tumor or to the blood.
In some embodiments the kit may further comprise at least one container having a predetermined quantity of another therapeutic agent (e.g. anti-infective agent or chemotherapy agent). In such embodiments, the kit may also comprise a second medicament or pharmaceutical composition such that the two medicaments or pharmaceutical compositions may be administered simultaneously or separately such that they provide a combined treatment for the specific disease or condition. The therapeutic agent may also be formulated so as to be suitable for injection or infusion to a tumor or to the blood.
Subjects
The subject to be treated may be any animal or human. The subject is preferably mammalian, more preferably human. The subject may be a non-human mammal, but is more preferably human. The subject may be male or female. The subject may be a patient. A subject may have been diagnosed with a disease or condition requiring treatment, or be suspected of having such a disease or condition.
Protein Expression
Molecular biology techniques suitable for the producing polypeptides according to the invention in cells are well known in the art, such as those set out in Sambrook et al., Molecular Cloning: A Laboratory Manual, New York: Cold Spring Harbor Press, 1989
The polypeptide may be expressed from a nucleotide sequence. The nucleotide sequence may be contained in a vector present in a cell, or may be incorporated into the genome of the cell. A "vector" as used herein is an oligonucleotide molecule (DNA or RNA) used as a vehicle to transfer exogenous genetic material into a cell. The vector may be an expression vector for expression of the genetic material in the cell. Such vectors may include a promoter sequence operably linked to the nucleotide sequence encoding the gene sequence to be expressed. A vector may also include a termination codon and expression enhancers. Any suitable vectors, promoters, enhancers and termination codons known in the art may be used to express polypeptides from a vector according to the invention. Suitable vectors include plasmids, binary vectors, viral vectors and artificial chromosomes (e.g. yeast artificial chromosomes). In this specification the term "operably linked" may include the situation where a selected nucleotide sequence and regulatory nucleotide sequence (e.g. promoter and/or enhancer) are covalently linked in such a way as to place the expression of the nucleotide sequence under the influence or control of the regulatory sequence (thereby forming an expression cassette). Thus a regulatory sequence is operably linked to the selected nucleotide sequence if the regulatory sequence is capable of effecting transcription of the nucleotide sequence. Where appropriate, the resulting transcript may then be translated into a desired protein or polypeptide.
Any cell suitable for the expression of polypeptides may be used for producing peptides according to the invention. The cell may be a prokaryote or eukaryote. Suitable prokaryotic cells include E.coli. Examples of eukaryotic cells include a yeast cell, a plant cell, insect cell or a mammalian cell. In some cases the cell is not a prokaryotic cell because some prokaryotic cells do not allow for the same post-translational modifications as eukaryotes. In addition, very high expression levels are possible in eukaryotes and proteins can be easier to purify from eukaryotes using appropriate tags. Specific plasmids may also be utilised which enhance secretion of the protein into the media.
Methods of producing a polypeptide of interest may involve culture or fermentation of a cell modified to express the polypeptide. The culture or fermentation may be performed in a bioreactor provided with an appropriate supply of nutrients, air/oxygen and/or growth factors. Secreted proteins can be collected by partitioning culture media/fermentation broth from the cells, extracting the protein content, and separating individual proteins to isolate secreted polypeptide. Culture, fermentation and separation techniques are well known to those of skill in the art.
Bioreactors include one or more vessels in which cells may be cultured. Culture in the bioreactor may occur continuously, with a continuous flow of reactants into, and a continuous flow of cultured cells from, the reactor. Alternatively, the culture may occur in batches. The bioreactor monitors and controls environmental conditions such as pH, oxygen, flow rates into and out of, and agitation within the vessel such that optimum conditions are provided for the cells being cultured.
Following culture of cells that express the polypeptide of interest, that polypeptide is preferably isolated. Any suitable method for separating polypeptides/proteins from cell culture known in the art may be used. In order to isolate a polypeptide/protein of interest from a culture, it may be necessary to first separate the cultured cells from media containing the polypeptide/protein of interest. If the polypeptide/protein of interest is secreted from the cells, the cells may be separated from the culture media that contains the secreted polypeptide/protein by centrifugation. If the polypeptide/protein of interest collects within the cell, it will be necessary to disrupt the cells prior to centrifugation, for example using sonification, rapid freeze-thaw or osmotic lysis. Centrifugation will produce a pellet containing the cultured cells, or cell debris of the cultured cells, and a supernatant containing culture medium and the polypeptide/protein of interest.
It may then be desirable to isolate the polypeptide/protein of interest from the supernatant or culture medium, which may contain other protein and non-protein components. A common approach to separating polypeptide/protein components from a supernatant or culture medium is by precipitation. Polypeptides/proteins of different solubility are precipitated at different concentrations of precipitating agent such as ammonium sulfate. For example, at low concentrations of precipitating agent, water soluble proteins are extracted. Thus, by adding increasing concentrations of precipitating agent, proteins of different solubility may be distinguished. Dialysis may be subsequently used to remove ammonium sulfate from the separated proteins.
Other methods for distinguishing different polypeptides/proteins are known in the art, for example ion exchange chromatography and size chromatography. These may be used as an alternative to precipitation, or may be performed subsequently to precipitation.
Once the polypeptide/protein of interest has been isolated from culture it may be necessary to concentrate the protein. A number of methods for concentrating a protein of interest are known in the art, such as ultrafiltration or lyophilisation.
Sequence Identity
Alignment for purposes of determining percent amino acid or nucleotide sequence identity can be achieved in various ways known to a person of skill in the art, for instance, using publicly available computer software such as ClustalW 1.82. T-coffee or Megalign
(DNASTAR) software. When using such software, the default parameters, e.g. for gap penalty and extension penalty, are preferably used. The default parameters of ClustalW 1.82 are: Protein Gap Open Penalty = 10.0, Protein Gap Extension Penalty = 0.2, Protein matrix = Gonnet, Protein/DNA ENDGAP = -1 , Protein/DNA GAPDIST = 4.
The invention includes the combination of the aspects and preferred features described except where such a combination is clearly impermissible or expressly avoided.
The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. Aspects and embodiments of the present invention will now be illustrated, by way of example, with reference to the accompanying figures. Further aspects and embodiments will be apparent to those skilled in the art. All documents mentioned in this text are incorporated herein by reference.
Throughout this specification, including the claims which follow, unless the context requires otherwise, the word "comprise," and variations such as "comprises" and
"comprising," will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.
It must be noted that, as used in the specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Ranges may be expressed herein as from "about" one particular value, and/or to "about" another particular value. When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by the use of the antecedent "about," it will be understood that the particular value forms another embodiment. Brief Description of the Figures
Embodiments and experiments illustrating the principles of the invention will now be discussed with reference to the accompanying figures in which: Figure 1. Light chain variable domain sequences for anti-PD-1 antibody clones A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4, H9 (human lgG4). CDRs are underlined and shown separately.
Figure 2. Heavy chain variable domain sequences for anti-PD-1 antibody A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4, H9 (human lgG4). CDRs are underlined and shown separately.
Figure 3. Table showing light chain and heavy chain CDR sequences for anti-PD-1 antibody clones A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4, H9. Figure 4. Nucleotide and encoded amino acid sequences of heavy and light chain variable domain sequences for anti-PD-1 antibody clones A3, A10, optimised A10, B6, optimised B6, C4, optimised C4, D4, E1 , F2, G1 , G2, G10, H4, optimised H4 and H9 (human lgG4).
Figure 5. Table showing binding affinity (KD, nM) of clones A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4 and H9, and control antibodies nivolumab and lambrolizumab for human PD-1. Figure 6. Chart showing binding of anti-human PD-1 antibodies A3, A10, nivolumab and lambrolizumab to activated T cells.
Figure 7. Chart showing binding of anti-human PD-1 antibodies A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4 and H9, nivolumab and lambrolizumab to activated T cells.
Figure 8. Chart showing reactivity of clones B6, C4, D4, E1 , F2, G1 , G2, G10, H4 and H9 with human PD-1 , rhesus PD-1 and human CTLA-4.
Figure 9. Chart showing allogeneic CD4+ T cell proliferation of exhausted T cells in response to antibodies A3, A10, nivolumab, lambrolizumab.
Figure 10. Chart showing allogeneic CD4+ T cell proliferation of exhausted T cells in response to antibodies A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4, H9, nivolumab, lambrolizumab.
Figure 11. Chart showing IFNv secretion of exhausted T cells in response to antibodies A3, A10, nivolumab, lambrolizumab.
Figure 12. Chart showing IFNv secretion of exhausted T cells in response to antibodies A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4, H9, nivolumab, lambrolizumab.
Figure 13. Chart showing specificity of binding of antibodies A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4, H9, for human and rhesus-PD-1 over other human (h) or murine (m) CD28 family members compared with nivolumab, and lambrolizumab, as determined by ELISA. Figure 14. Chart showing allogeneic CD4+ T cell proliferation of exhausted T cells in response to antibodies A10 codon optimised, B6 codon optimised, C4 codon optimised, H4 codon optimised, nivolumab, lambrolizumab.
Figure 15. Chart showing IFNv secretion of exhausted T cells in response to antibodies A10 codon optimised, B6 codon optimised, C4 codon optimised, H4 codon optimised, nivolumab, lambrolizumab. Figure 16. Chart showing the expression of exhaustion markers PD-1 , PD-L1 , TIM-3 and LAG-3 by lung tumour infiltrating lymphocytes. Approximately 2/3 of CD4+ and CD8+ lymphocytes in the tumour express PD-1.
Figure 17. Charts showing the secretion of IFN-γ by tumour infiltrating lymphocytes (A) after 7 days of culture in the presence or absence of anti-PD-1 antibodies (direct culture of tumour dissociated tissues), (B) after a mixed lymphocyte reaction in the presence or absence of anti-PD-1 antibodies.
Figure 18. Chart showing the secreted IFN-γ after culture of PBMCs with Influenza virus infected dendritic cells in the presence or absence of anti-PD-1 antibodies.
Figure 19. Chart showing the expression of exhaustion markers PD-1 , PD-L1 , TIM-3, LAG-3 and CTLA4 by lung tumour infiltrating lymphocytes. Approximately 2/3 of CD4+ and CD8+ lymphocytes in the tumour express PD-1.
Figure 20. Charts showing IFN-γ secretion by tumour infiltrating lymphocytes after a mixed lymphocyte reaction in the presence or absence of anti-PD-1 antibodies, for three patients. (A) Patient #1 , (B) Patient #2, (C) Patient #3. Shown are mean ± SD from duplicates or triplicates.
Figure 21. Charts showing the expression of exhaustion markers PD-1 , PD-L1 , ΤΊΜ- 3, LAG-3 and CTLA4 by (A) renal tumour infiltrating lymphocytes and (B) blood circulating lymphocytes from renal carcinoma patients. Approximately 2/3 of CD4+ and CD8+ lymphocytes in the tumour express PD-1 , while most of PBMC lymphocytes do not express PD-1. Figure 22. Charts showing IFN-γ secretion by tumour infiltrating lymphocytes after a mixed lymphocyte reaction in the presence or absence of anti-PD-1 antibodies, for three patients. (A) Patient #1 , (B) Patient #2, (C) Patient #3. Shown are mean ± SD from duplicates or triplicates.
Figure 23. Charts showing the expression of exhaustion markers PD-1 , PD-L1 , TIM- 3, LAG-3 and CTLA4 by (A) bladder tumour infiltrating lymphocytes and (B) blood circulating lymphocytes from bladder carcinoma patients. A majority of tumour infiltrating lymphocytes express PD-1 , while only a minority of PBMC lymphocytes express PD-1.
Figure 24. Chart showing IFN-γ secretion by tumour infiltrating lymphocytes after a mixed lymphocyte reaction in the presence or absence of anti-PD-1 antibodies, for one patient. Shown are mean ± SD from duplicates or triplicates. Examples
The inventors described in the following Examples the identification of nucleotide and amino-acid sequences of isolated antibodies, or the antigen-binding portions thereof, that specifically bind human and rhesus PD-1 , block the PD-1 pathway and restore exhausted T cell activity.
Isolation of anti-human PD-1 antibodies
Anti-PD-1 antibodies were isolated from a human antibody phage display library via in vitro selection in a 4-round bio-panning process.
Streptavidin-magnetic beads were coated with biotinylated human PD-1 and used to fish- out anti-PD-1-specific phages using magnetic sorting. Some steps to remove of potential anti-biotin antibodies were added in the selection process.
Specific Fab antibodies were originally identified by ELISA with human-PD-1 as the antigen. A first clonality screening was performed by DNA fingerprinting; clonality was then confirmed by sequencing.
Affinity maturation
Selected antibodies went through affinity maturation by CDR engineering. Selection of specific anti-PD-1 antibodies was conducted by phage display. More than 80 affinity- matured clones with ~100-fold improved affinity were generated. To select clones, thermostability assays were conducted. Antibodies were heated and their stability monitored based on their ability to bind human PD-1 in ELISA. Clones still stable at 71 °C were kept for further analyses, their affinity for human PD-1 was measured using a ProteOn bioanalyser (Biorad). Briefly, human-PD-1 coupled to Fc was immobilised on a sensor chip and a flow of antibody was applied; association and dissociation rates were measured and the affinity (KD) calculated. Twelve of the affinity-matured clones were designated A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4 and H9.
Affinity of the isolated anti-PD-1 antibodies
Affinity of A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4 and H9 for human PD-1 was measured by Surface Plasmon Resonance and compared to the affinity of nivolumab and lambrolizumab, two anti-PD-1 antibodies in late clinical development stage (Figure 5).
Briefly, human or mouse PD-1 coupled to human Fc was immobilised on a sensor chip compatible with the Proteon XPR36 bioanalyser (Biorad). Crude Fab extracts (or the control antibodies) were then flown onto the chip and the association/dissociation of each candidate Fab was then recorded and analysed, the affinity (KD) was calculated.
A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4 and H9 show affinity in the range 0.1 to 0.7 nM while nivolumab and lambrolizumab exhibit KD of 1.9 and 0.5 nM respectively (Figure 5).
Characterisation of the anti-human PD-1 antibodies:
Binding to PD-1 expressing cells
50,000 T cells isolated from a healthy donor were incubated in the presence of 5,000 monocyte-derived DCs from another donor for 7 days at 37°C. Antibodies, expressed as lgG4, were incubated with such activated T cells for 30 minutes in PBS. After a wash in PBS, a fluorescent labelled secondary antibody was added for 30 minutes and then washed in PBS. Cells were resuspended in FACS buffer and binding of the antibody to the cells was monitored by flow cytometry (Figures 6 and 7).
Cross-reactivity with rhesus PD-1
Affinity-matured clone Fabs were tested in ELISA for recognition of rhesus PD-1. Briefly, ELISA plates were coated with 350ng/well of human or rhesus PD-1 in carbonate buffer and then blocked with a solution of casein. After extensive washes in PBS Tween-20, antibody-containing supernatants were transferred into the ELISA plates in the presence of 7% milk in PBS. After 90 minutes at room temperature under agitation and extensive washes, a goat anti-human Fab antibody coupled to HRP was added. One hour later, plates were washed and TMB substrate added. The reaction was stopped with 1 M HCI and optical density measured at 450nm with a reference at 670nm (Figure 8).
In vitro functional activity
Anti-PD-1 antibodies were tested in a functional assay measuring 2 activity parameters of T cells: proliferation and secretion of IFN-γ. Briefly, T cells were isolated from a healthy donor and cultured for 7 days in the presence of monocyte-derived dendritic cells from another donor (50,000 Tcells / 5,000 DCs). This continuous stimulation induces exhaustion of the T cells. Antibodies were then added to the cultures for 5 more days. After 4 days, supernatants were collected to measure IFN-γ by ELISA, and T cells were cultured for 4 days, with addition of 1 μθϊ of tritiated thymidine for the last 18 hours. Cells were then harvested and proliferation measured with a β-counter.
Clones A3, A10, B6, C4, D4, E1 , F2, G1 , G2, G10, H4 and H9 are able to restore proliferation of previously exhausted T cells and to restore their ability to secrete IFN-γ (Figures 9 to 12).
Specificity for PD-1
Anti-PD-1 antibodies were tested in an ELISA assay for their ability to bind to other members of the CD28 family.
Briefly, ELISA plates were coated with 350ng/well of one of the following antigens coupled to human Fc in carbonate buffer: human PD-1 , human PD-L1 , human TIM-3, human LAG-3, human ICOS, human CTLA4, human BTLA, human CD28, mouse TIM-3, or rhesus PD-1. The plate was then blocked with a solution of casein. After extensive washes in PBS Tween-20, antibodies were added into the ELISA wells in the presence of 7% milk in PBS. After 90 minutes at room temperature under agitation and extensive washes, a goat anti-human Fab antibody coupled to HRP was added. One hour later, plates were washed and TMB substrate added. The reaction was stopped with 1 M HCI and optical density measured at 450nm with a reference at 670nm. While nivolumab cross-binds to PD-L1 , TIM-3, LAG-3, BTLA and CD28 (and
lambrolizumab in a very weak manner too), 1 D1 1 and 1G4 are specific to PD-1 only; showing only a very weak cross-recognition of other members than PD-1 (Figure 13).
Production yield
HEK-293.6E cells were transiently transfected to produce anti-PD-1 antibodies in lgG4 format; production yield in this system was compared.
Figure imgf000053_0001
All novel antibody clones showed better production yield than lambrolizumab.
Codon optimisation
In order to increase gene expression efficiency, A10, B6, C4 and H4 underwent codon optimisation.
Codon-optimised clones retain their ability to neutralise PD-1 and restore T cell activity (T cell proliferation and IFN-γ secretion - Figures 14 and 15).
Use of anti-PD-1 antibodies to treat tumours: ex vivo activation of tumour infiltrating lymphocytes Lung tumour samples were obtained from the National Cancer Centre Singapore after approval by the proper IRB. Samples were dissociated using a human tumour dissociation kit and a tissue dissociator device.
To confirm expression of exhaustion markers at the surface of the tumour infiltrating lymphocytes, isolated cell mixture was washed once and passed through 70μηι filter to obtain single cell suspension. Cells were stained with antibodies against CD4, CD8, PD- 1 , PD-L1 , TIM-3 and LAG-3; a live/dead marker was also used to exclude dead cells from the analysis. Cells were analysed by flow cytometry. Results are shown in Figure 16.
The tumour dissociated mixture was cultured with anti-PD-1 codon-optimised antibodies A10, B6, C4 and H4 for 7 days prior to measurement of IFN-γ in the supernatant by
ELISA. Nivolumab and lambrolizumab were used as positive controls, an isotype antibody as a negative control.
Figure 17A presents the secretion of IFN-γ by tumour infiltrating lymphocytes after 7 days of culture in the presence or absence of anti-PD-1 antibodies. Anti-PD-1 antibodies were able to re-activate lymphocytes to secrete IFN-γ in a dose-dependent manner.
Another fraction of the dissociated mixture was co-cultured with allogeneic dendritic cells (DCs) to initiate a mixed lymphocyte reaction (MLR). Cells were first cultured with DCs for 7 days without antibodies and then restimulated with DCs for 7 days in the presence of anti-PD-1 or control antibodies. After these 2 rounds, IFN-γ was assayed in supernatants by ELISA.
Figure 17B presents the secretion of IFN-γ after the MLR in the presence or absence of anti-PD-1 antibodies. Anti-PD-1 antibodies were able to restore the ability of lymphocytes located in the tumour site to secrete IFN-γ in a dose-dependent manner.
Use of anti-PD-1 antibodies to treat infections: autologous activation of T cells in the presence of Influenza
Blood was collected from /n//uenza-positive donors. Monocyte-derived DCs were infected with influenza virus A/PR/8/34 (H1 N1). Infected DCs were then mixed to PBMCs from the same donor for a first round of culture of 5 days. Cells were then restimulated with Influenza- infected DCs and cultured for a second round of 5 days in the presence of anti- PD-1 antibodies. After these 2 rounds, most of the cells in culture are lnfluenza-spec\f c T cells. At the end of the 2 rounds of culture, IFN-γ was assayed in supernatants by ELISA.
Figure 18 presents the secreted IFN-γ after culture of PBMCs with Influenza virus infected DCs in the presence or absence of anti-PD-1 antibodies. Anti-PD-1 antibodies were able to restore the capacity of lymphocytes to secrete IFN-γ upon viral stimulation in a dose-dependent manner.
Lung cancer: use of anti-PD-1 antibodies to re-activate lung tumour infiltrating lymphocytes lex vivo data)
Lung tumour samples were obtained from the National Cancer Centre Singapore after approval by the proper IRB. Samples were dissociated using a human tumour
dissociation kit and a tissue dissociator device.
To confirm expression of exhaustion markers at the surface of the tumour infiltrating lymphocytes, isolated cell mixture was washed once and passed through 70μηι filter to obtain single cell suspension. Cells were stained with antibodies against CD4, CD8, PD- 1 , PD-L1 , Tim-3, LAG-3 and CTLA-4; a live/dead marker was also used to exclude dead cells from the analysis. Cells were analysed by flow cytometry.
Figure 19 shows the expression of PD-1 , PD-L1 , TIM-3, LAG-3 and CTLA-4 by tumour infiltrating lymphocytes from 3 different patients (shown are mean ± SD from 3
independent experiments using cells from 3 different donors, all experiments done in triplicates). Approximately 2/3 of CD4+ and CD8+ lymphocytes in the tumour express PD- 1.
The tumour dissociated mixture was co-cultured with allogeneic dendritic cells (DC) to initiate a mixed lymphocyte reaction (MLR). Cells were first cultured for 7 days without antibodies and then for 7 days in the presence of anti-PD-1 or control antibodies. After these 2 rounds, IFN-γ was assayed in supernatants by ELISA.
Figures 20A-20C present the secretion of IFN-γ after the MLR in the presence or absence of anti-PD-1 antibodies. Shown are mean ± SD from triplicates in 3 independent experiments (cells from 3 different patients). Anti-PD-1 antibodies were able to restore the ability of lymphocytes located in the tumour site to secrete IFN-γ in a dose-dependent manner.
Renal cell carcinoma: use of anti-PD-1 antibodies to re-activate kidney tumour infiltrating lymphocytes lex vivo data)
Kidney tumour samples were obtained from the National Cancer Centre Singapore after approval by the proper IRB. Samples were dissociated using a human tumour
dissociation kit and a tissue dissociator device. To confirm expression of exhaustion markers at the surface of the tumour infiltrating lymphocytes, isolated cell mixture was washed once and passed through 70μηι filter to obtain single cell suspension. Cells were stained with antibodies against CD4, CD8, PD- 1 , PD-L1 , Tim-3, LAG-3 and CTLA-4; a live/dead marker was also used to exclude dead cells from the analysis. Cells were analysed by flow cytometry. As a comparison, same markers expression was assessed on PBMCs from the same patients.
Figures 21 A and 21 B show the expression of PD-1 , PD-L1 , TIM-3, LAG-3 and CTLA-4 by tumour infiltrating lymphocytes (Fig. 21 A) and on blood circulating lymphocytes (Fig. 21 B) from 3 different patients (shown are mean ± SD from 3 independent experiments using cells from 3 different donors, all experiments done in triplicates). Approximately 2/3 of CD4+ and CD8+ lymphocytes in the tumour express PD-1 while most of PBMC lymphocytes do not express PD-1.
The tumour dissociated mixture was co-cultured with allogeneic dendritic cells (DC) to initiate a mixed lymphocyte reaction (MLR). Cells were first cultured for 7 days without antibodies and then for 7 days in the presence of anti-PD-1 or control antibodies. After these 2 rounds, IFN-γ was assayed in supernatants by ELISA.
Figures 22A-22C present the secretion of IFN-γ after the MLR in the presence or absence of anti-PD-1 antibodies. Shown are mean ± SD from duplicates or triplicates in 3 independent experiments (cells from 3 different patients). Anti-PD-1 antibodies were able to restore the ability of lymphocytes located in the tumour site to secrete IFN-γ in a dose- dependent manner.
Bladder cancer: use of anti-PD-1 antibodies to re-activate bladder tumour infiltrating lymphocytes lex vivo data)
Bladder tumour samples were obtained from the National Cancer Centre Singapore after approval by the proper IRB. Samples were dissociated using a human tumour dissociation kit and a tissue dissociator device.
To confirm expression of exhaustion markers at the surface of the tumour infiltrating lymphocytes, isolated cell mixture was washed once and passed through 70μηι filter to obtain single cell suspension. Cells were stained with antibodies against CD4, CD8, PD- 1 , PD-L1 , Tim-3, LAG-3 and CTLA-4; a live/dead marker was also used to exclude dead cells from the analysis. Cells were analysed by flow cytometry. Figure 23A and 23B show the expression of PD-1 , PD-L1 , TIM-3, LAG-3 and CTLA-4 by tumour infiltrating lymphocytes (Fig. 23A) and blood circulating lymphocytes (Fig. 23B) from 2 different patients (shown are mean ± SD from 2 independent experiments using cells from 2 different donors, all experiments done in triplicates). A majority of tumour infiltrating lymphocytes express PD-1 , while only a minority of their blood circulating counterparts do so.
The tumour dissociated mixture was co-cultured with allogeneic dendritic cells (DC) to initiate a mixed lymphocyte reaction (MLR). Cells were first cultured for 7 days without antibodies and then for 7 days in the presence of anti-PD-1 or control antibodies. After these 2 rounds, IFN-γ was assayed in supernatants by ELISA.
Figure 24 presents the secretion of IFN-γ after the MLR in the presence or absence of anti-PD-1 antibodies. Shown are mean ± SD from triplicates in 1 experiment. Anti-PD-1 antibodies were able to restore the ability of lymphocytes located in the tumour site to secrete IFN-γ in a dose-dependent manner.

Claims

Claims:
1. An antibody, or antigen binding fragment which is capable of binding to PD-1 , optionally isolated, having the amino acid sequences i) to vi):
i) LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25) ii) LC-CDR2: SNNQRPS (SEQ ID NO:26) iii) LC-CDR3: X1X2WDDX3X4X5GX6X7 (SEQ ID NO:53) iv) HC-CDR1 : SYGMH (SEQ ID NO:89) or
GFTFSSYGMH (SEQ ID NO: 39) v) HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40) vi) HC-CDR3: DZ1GZ2GZ3YZ4YGZ5DZ6 (SEQ ID NO:54) or a variant thereof in which one or two or three amino acids in one or more of the sequences (i) to (vi) are replaced with another amino acid, where
Figure imgf000058_0001
A or S, X2= S or A, Xs= V, Y, F, D, S or A, X4= L, Y, V or A, Xs= Y, R or H, Xe= S, or T, X7= V, I , or M and Zi= L or Y, Z2= A or S, Z3= P or Y, Z4= Y or L, Z5= K, M or L, Z6= H or V.
2. The antibody, or antigen binding fragment, of claim 1 , wherein LC-CDR3 is one of ASWDDVLYGSV (SEQ ID NO:27), ASWDDYYYGTI (SEQ ID NO:28), ASWDDYLRGTV (SEQ ID NO:29), SAWDDYLHGTV (SEQ ID NO:30), ASWDDYVRGTM (SEQ ID NO:31), SSWDDFLRGTV (SEQ ID NO:32), SSWDDDARGTI (SEQ ID NO:33), AAWDDVYYGTI (SEQ ID NO:34), ASWDDSLYGTV (SEQ ID NO:35), AAWDDAYYGTI (SEQ ID NO:36), ASWDDVYRGTV (SEQ ID NO:37), or SSWDDSLYGTI (SEQ ID NO:38).
3. The antibody, or antigen binding fragment, of claim 1 or 2, wherein HC-CDR3 is one of DLGAGPYYYGKDH (SEQ ID NO:41), D LG AG PYYYG KD V (SEQ ID NO:42), DYGAGPYYYGMDV (SEQ ID NO:43), DLGAG PYYYG LDV (SEQ ID NO:44),
D LGAG PYYYG M D V (SEQ ID NO:45), D LG AG PYYYG MDV (SEQ ID NO:46),
D LGAG PYYYG M D V (SEQ ID NO:47), DLGAG PYYYG MDV (SEQ ID NO:48),
D LGAG PYYYG M D V (SEQ ID NO:49), DYGAGPYYYGMDV (SEQ ID NO:50),
DLGSGYYLYGMDV (SEQ ID NO:51), or D LGAG PYYYG M D V (SEQ ID NO:52).
4. The antibody, or antigen binding fragment, of any one of claims 1 to 3, having at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26) LC-CDR3: ASWDDVLYGSV (SEQ ID NO:27)
5. The antibody, or antigen binding fragment, of any one of claims 1 to 3, having at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDYYYGTI (SEQ ID NO:28)
6. The antibody, or antigen binding fragment, of any one of claims 1 to 3, having at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDYLRGTV (SEQ ID NO:29)
7. The antibody, or antigen binding fragment, of any one of claims 1 to 3, having at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: SAWDDYLHGTV (SEQ ID NO:30).
8. The antibody, or antigen binding fragment, of any one of claims 1 to 3, having at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDYVRGTM (SEQ ID NO:31).
9. The antibody, or antigen binding fragment, of any one of claims 1 to 3, having at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: SSWDDFLRGTV (SEQ ID NO:32).
10. The antibody, or antigen binding fragment, of any one of claims 1 to 3, having at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: SSWDDDARGTI (SEQ ID NO:33).
1 1. The antibody, or antigen binding fragment, of any one of claims 1 to 3, having at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: AAWDDVYYGTI (SEQ ID NO:34).
12. The antibody, or antigen binding fragment, of any one of claims 1 to 3, having at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDSLYGTV (SEQ ID NO:35).
13. The antibody, or antigen binding fragment, of any one of claims 1 to 3, having at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: AAWDDAYYGTI (SEQ ID NO:36).
14. The antibody, or antigen binding fragment, of any one of claims 1 to 3, having at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: ASWDDVYRGTV (SEQ ID NO:37).
15. The antibody, or antigen binding fragment, of any one of claims 1 to 3, having at least one light chain variable region incorporating the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26) LC-CDR3: SSWDDSLYGTI (SEQ ID NO:38)
16. The antibody, or antigen binding fragment, of any one of claims 1 to 15, having at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : SYGMH (SEQ ID NO:89) or
GFTFSSYGMH (SEQ ID NO:39)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGKDH (SEQ ID NO:41)
17. The antibody, or antigen binding fragment, of any one of claims 1 to 15, having at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : SYGMH (SEQ ID NO:89) or
GFTFSSYGMH (SEQ ID NO:39)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: D LG AG PYYYG KD V (SEQ ID NO:42).
18. The antibody, or antigen binding fragment, of any one of claims 1 to 15, having at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : SYGMH (SEQ ID NO:89) or
GFTFSSYGMH (SEQ ID NO:39)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: D YG AG PYYYG M D V (SEQ ID NO:43)
19. The antibody, or antigen binding fragment, of any one of claims 1 to 15, having at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : SYGMH (SEQ ID NO:89) or
GFTFSSYGMH (SEQ ID NO:39)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGLDV (SEQ ID NO:44).
20. The antibody, or antigen binding fragment, of any one of claims 1 to 15, having at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : SYGMH (SEQ ID NO:89) or
GFTFSSYGMH (SEQ ID NO:39) HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:45).
21. The antibody, or antigen binding fragment, of any one of claims 1 to 15, having at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : SYGMH (SEQ ID NO:89) or
GFTFSSYGMH (SEQ ID NO:39)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:46).
22. The antibody, or antigen binding fragment, of any one of claims 1 to 15, having at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : SYGMH (SEQ ID NO:89) or
GFTFSSYGMH (SEQ ID NO:39)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:47).
23. The antibody, or antigen binding fragment, of any one of claims 1 to 15, having at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : SYGMH (SEQ ID NO:89) or
GFTFSSYGMH (SEQ ID NO:39)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:48).
24. The antibody, or antigen binding fragment, of any one of claims 1 to 15, having at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : SYGMH (SEQ ID NO:89) or
GFTFSSYGMH (SEQ ID NO:39)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:49).
25. The antibody, or antigen binding fragment, of any one of claims 1 to 15, having at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : SYGMH (SEQ ID NO:89) or GFTFSSYGMH (SEQ ID NO:39)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DYGAGPYYYGMDV (SEQ ID NO:50).
26. The antibody, or antigen binding fragment, of any one of claims 1 to 15, having at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : SYGMH (SEQ ID NO:89) or
GFTFSSYGMH (SEQ ID NO:39)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGSGYYLYGMDV (SEQ ID NO:51).
27. The antibody, or antigen binding fragment, of any one of claims 1 to 15, having at least one heavy chain variable region incorporating the following CDRs:
HC-CDR1 : SYGMH (SEQ ID NO:89) or
GFTFSSYGMH (SEQ ID NO:39)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DLGAGPYYYGMDV (SEQ ID NO:52).
28. An antibody, or antigen binding fragment, optionally isolated, that specifically binds PD-1 (e.g. human or rhesus) over other members of the CD28 family, wherein the antibody is optionally as claimed in any one of claims 1 to 27.
29. The antibody, or antigen binding fragment, of any one of claims 1 to 28, wherein the antibody is effective to restore T-cell function in T-cells exhibiting T-cell exhaustion or T-cell anergy.
30. An in vitro complex, optionally isolated, comprising an antibody, or antigen binding fragment, according to any one of claims 1 to 29 bound to PD-1.
31. An isolated light chain variable region polypeptide comprising the following CDRs:
LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25)
LC-CDR2: SNNQRPS (SEQ ID NO:26)
LC-CDR3: Xi X2 WD DX3X4X5GX6X7 (SEQ ID NO:53)
where
Figure imgf000063_0001
Y, R or H, X6= S, or T, X7= V, I, or M.
32. The isolated light chain variable region polypeptide of claim 31 wherein LC-CDR3 is one of ASWDDVLYGSV (SEQ ID NO:27), ASWDDYYYGTI (SEQ ID NO:28),
ASWDDYLRGTV (SEQ ID NO:29), SAWDDYLHGTV (SEQ ID NO:30), ASWDDYVRGTM (SEQ ID NO:31), SSWDDFLRGTV (SEQ ID NO:32), SSWDDDARGTI (SEQ ID NO:33), AAWDDVYYGTI (SEQ ID NO:34), ASWDDSLYGTV (SEQ ID NO:35), AAWDDAYYGTI (SEQ ID NO:36), ASWDDVYRGTV (SEQ ID NO:37), or SSWDDSLYGTI (SEQ ID NO:38).
33. An isolated heavy chain variable region polypeptide comprising the following CDRs:
HC-CDR1 : SYGMH (SEQ ID NO:89) or
GFTFSSYGMH (SEQ ID NO:39)
HC-CDR2: VISYDGSNKYYADSVKG (SEQ ID NO:40)
HC-CDR3: DZ1GZ2GZ3YZ4YGZ5DZ6 (SEQ ID NO:54)
where Zi= L or Y, Z2= A or S, Z3= P or Y, Z4= Y or L, Z5= K, M or L, Z6= H or V.
34. The isolated heavy chain variable region polypeptide of claim 33 wherein HC- CDR3 is one of DLGAGPYYYGKDH (SEQ ID NO:41), D LG AG PYYYG KD V (SEQ ID NO:42), DYGAGPYYYGMDV (SEQ ID NO:43), DLGAGPYYYGLDV (SEQ ID NO:44), D LGAG PYYYG M D V (SEQ ID NO:45), D LG AG PYYYG M D V (SEQ ID NO:46),
D LGAG PYYYG M D V (SEQ ID NO:47), D LG AG PYYYG M D V (SEQ ID NO:48),
D LGAG PYYYG M D V (SEQ ID NO:49), DYGAGPYYYGMDV (SEQ ID NO:50),
DLGSGYYLYGMDV (SEQ ID NO:51), or D LGAG PYYYG M D V (SEQ ID NO:52).
35. An isolated light chain variable region polypeptide of claim 31 or 32 in combination with a heavy chain variable region polypeptide according to claim 33 or 34.
36. An antibody or antigen binding fragment which is capable of binding to PD-1 , comprising a heavy chain and a light chain variable region sequence, wherein:
the heavy chain comprises a HC-CDR1 , HC-CDR2, HC-CDR3, having at least 85% overall sequence identity to HC-CDR1 : SYGMH (SEQ ID NO:89) or
GFTFSSYGMH (SEQ ID NO: 39), HC-CDR2 VISYDGSNKYYADSVKG (SEQ ID NO:40), HC-CDR3: DZiGZ2GZ3YZ4YGZ5DZ6 (SEQ ID NO:54), respectively, where Zi= L or Y, Z2= A or S, Z3= P or Y, Z4= Y or L, Z5= K, M or L, Z6= H or V, and the light chain comprises a LC-CDR1 , LC-CDR2, LC-CDR3,, having at least 85% overall sequence identity to LC-CDR1 : SGSSSNIKFNSVN (SEQ ID NO:25), LC- CDR2: SNNQRPS (SEQ ID NO:26), LC-CDR3: X1X2WDDX3X4X5GX6X7 (SEQ ID NO:53), respectively, where
Figure imgf000065_0001
A or S, X2= S or A, X3= V, Y, F, D, S or A, X4= L, Y, V or A, Xs= Y, R or H, Xe= S, or T, X7= V, I, or M.
37. An antibody or antigen binding fragment which is capable of binding to PD-1 , optionally isolated, comprising a heavy chain and a light chain variable region sequence, wherein:
the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence of one of SEQ ID NOs: 13 to 24 (Figure 2), and
the light chain sequence has at least 85% sequence identity to the light chain sequence: SEQ ID NO:1 or 12 (Figure 1).
38. An antibody or antigen binding fragment, optionally isolated, which is capable of binding to PD-1 , which is a bispecific antibody or a bispecific antigen binding fragment comprising (i) an antigen binding fragment or polypeptide according to any of one of claims 1 to 37, and (ii) an antigen binding domain which is capable of binding to a target protein other than PD-1.
39. The antibody, or antigen binding fragment, of claim 38, wherein the antigen binding domain which is capable of binding to a target protein other than PD-1 is capable of binding to one of TIM-3, LAG 3, ICOS, CTLA4, BTLA or CD28.
40. A composition comprising the antibody, or antigen binding fragment, or polypeptide of any one of claims 1 to 39 and at least one pharmaceutically-acceptable carrier.
41. An isolated nucleic acid encoding the antibody, or antigen binding fragment or polypeptide of any of one of claims 1 to 39.
42. A vector comprising the nucleic acid of claim 41.
43. A host cell comprising the vector of claim 42.
44. A method for making an antibody, or antigen binding fragment or polypeptide of any of one of claims 1 to 39 comprising culturing the host cell of any of claim 43 under conditions suitable for the expression of a vector encoding the antibody, or antigen binding fragment or polypeptide, and recovering the antibody, or antigen binding fragment or polypeptide.
45. An antibody, antigen binding fragment or polypeptide according to any of one of claims 1 to 39 for use in therapy, or in a method of medical treatment.
46. An antibody, antigen binding fragment or polypeptide according to any of one of claims 1 to 39 for use in the treatment of a T-cell dysfunctional disorder.
47. An antibody, antigen binding fragment or polypeptide according to any of one of claims 1 to 39 for use in the treatment of cancer.
48. An antibody, antigen binding fragment or polypeptide according to any of one of claims 1 to 39 for use in the treatment of an infectious disease.
49. Use of an antibody, antigen binding fragment or polypeptide according to any of one of claims 1 to 39 in the manufacture of a medicament for use in the treatment of a T- cell dysfunctional disorder.
50. Use of an antibody, antigen binding fragment or polypeptide according to any of one of claims 1 to 39 in the manufacture of a medicament for use in the treatment of cancer.
51. Use of an antibody, antigen binding fragment or polypeptide according to any of one of claims 1 to 39 in the manufacture of a medicament for use in the treatment of an infectious disease.
52. A method, in vitro or in vivo, of enhancing T-cell function comprising administering an antibody, antigen binding fragment or polypeptide according to any one of claims 1 to 39 to a dysfunctional T-cell.
53. A method of treating a T-cell dysfunctional disorder comprising administering an antibody, antigen binding fragment or polypeptide according to any one of claims 1 to 39 to a patient suffering from a T-cell dysfunctional disorder.
54. A method of treating cancer comprising administering an antibody, antigen binding fragment or polypeptide according to any one of claims 1 to 39 to a patient suffering from a cancer.
55. A method of treating an infectious disease comprising administering an antibody, antigen binding fragment or polypeptide according to any one of claims 1 to 39 to a patient suffering from an infectious disease.
56. A method comprising contacting a sample containing, or suspected to contain, PD-1 with an antibody or antigen binding fragment according to any one of claims 1 to 39 and detecting the formation of a complex of antibody, or antigen binding fragment, and PD-1.
57. A method of diagnosing a disease or condition in a subject, the method comprising contacting, in vitro, a sample from the subject with an antibody, or antigen binding fragment, according to any one of claims 1 to 39 and detecting the formation of a complex of antibody, or antigen binding fragment, and PD-1.
58. A method of selecting or stratifying a subject for treatment with PD-1 targeted agents, the method comprising contacting, in vitro, a sample from the subject with an antibody, or antigen binding fragment, according to any one of claims 1 to 39 and detecting the formation of a complex of antibody, or antigen binding fragment, and PD-1.
59. Use of an antibody, or antigen binding fragment, according to any one of 1 to 39 for the detection of PD-1 in vitro.
60. Use of an antibody, or antigen binding fragment, according to any one of 1 to 39 as an in vitro diagnostic agent.
61. A method for expanding a population of T cells, wherein T cells are contacted in vitro or ex vivo with an antibody, antigen binding fragment or polypeptide according to any one of claims 1 to 39.
62. A method of treatment of a subject having a T-cell dysfunctional disorder, the method comprising culturing T cells obtained from a blood sample from a subject in the presence of an antibody, antigen binding fragment or polypeptide according to any one of claims 1 to 39 so as to expand the T cell population, collecting expanded T cells, and administering the expanded T cells to a subject in need of treatment.
PCT/SG2015/050413 2014-10-27 2015-10-27 Anti-pd-1 antibodies WO2016068801A1 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
CA2965623A CA2965623A1 (en) 2014-10-27 2015-10-27 Anti-pd-1 antibodies
KR1020177014305A KR20170070243A (en) 2014-10-27 2015-10-27 Anti-pd-1 antibodies
SG11201703344WA SG11201703344WA (en) 2014-10-27 2015-10-27 Anti-pd-1 antibodies
CN201580071389.1A CN107438621B (en) 2014-10-27 2015-10-27 anti-PD-1 antibodies
MYPI2017701430A MY191581A (en) 2014-10-27 2015-10-27 Anti-pd-1 antibodies
JP2017540959A JP6701213B2 (en) 2014-10-27 2015-10-27 Anti-PD-1 antibody
AU2015340054A AU2015340054A1 (en) 2014-10-27 2015-10-27 Anti-PD-1 antibodies
RU2017116847A RU2715628C2 (en) 2014-10-27 2015-10-27 Antibodies to pd-1
US15/521,434 US10280224B2 (en) 2014-10-27 2015-10-27 Anti-PD-1 antibodies
EP15855595.3A EP3212671A4 (en) 2014-10-27 2015-10-27 Anti-pd-1 antibodies
US15/495,048 US9771425B2 (en) 2014-10-27 2017-04-24 Anti-PD-1 antibodies
IL251963A IL251963A0 (en) 2014-10-27 2017-04-27 Anti-pd-1 antibodies
PH12017500803A PH12017500803A1 (en) 2014-10-27 2017-04-27 Anti-pd-1 antibodies
US16/360,445 US11072659B2 (en) 2014-10-27 2019-03-21 Anti-PD-1 antibodies

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1419084.7A GB201419084D0 (en) 2014-10-27 2014-10-27 Anti-PD-1 antibodies
GB1419084.7 2014-10-27

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US15/521,434 A-371-Of-International US10280224B2 (en) 2014-10-27 2015-10-27 Anti-PD-1 antibodies
US15/495,048 Continuation US9771425B2 (en) 2014-10-27 2017-04-24 Anti-PD-1 antibodies
US16/360,445 Continuation US11072659B2 (en) 2014-10-27 2019-03-21 Anti-PD-1 antibodies

Publications (1)

Publication Number Publication Date
WO2016068801A1 true WO2016068801A1 (en) 2016-05-06

Family

ID=52103452

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SG2015/050413 WO2016068801A1 (en) 2014-10-27 2015-10-27 Anti-pd-1 antibodies

Country Status (14)

Country Link
US (3) US10280224B2 (en)
EP (1) EP3212671A4 (en)
JP (1) JP6701213B2 (en)
KR (1) KR20170070243A (en)
CN (1) CN107438621B (en)
AU (1) AU2015340054A1 (en)
CA (1) CA2965623A1 (en)
GB (1) GB201419084D0 (en)
IL (1) IL251963A0 (en)
MY (1) MY191581A (en)
PH (1) PH12017500803A1 (en)
RU (1) RU2715628C2 (en)
SG (1) SG11201703344WA (en)
WO (1) WO2016068801A1 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017220990A1 (en) 2016-06-20 2017-12-28 Kymab Limited Anti-pd-l1 antibodies
WO2018013017A1 (en) * 2016-07-13 2018-01-18 Закрытое Акционерное Общество "Биокад" Anti-pd-1 antibodies, method for producing same and method for using same
JP2018029587A (en) * 2016-08-17 2018-03-01 治範 小田 Method of culturing lymphocyte having improved cytotoxic activity, and cellular immunological therapeutic agent containing lymphocyte having improved cytotoxic activity obtained by the method
US10160806B2 (en) 2014-06-26 2018-12-25 Macrogenics, Inc. Covalently bonded diabodies having immunoreactivity with PD-1 and LAG-3, and methods of use thereof
WO2019092181A1 (en) 2017-11-10 2019-05-16 Agency For Science, Technology And Research Il2rbeta/common gamma chain antibodies
US10344090B2 (en) 2013-12-12 2019-07-09 Shanghai Hangrui Pharmaceutical Co., Ltd. PD-1 antibody, antigen-binding fragment thereof, and medical application thereof
US10428145B2 (en) 2015-09-29 2019-10-01 Celgene Corporation PD-1 binding proteins and methods of use thereof
US10577422B2 (en) 2015-07-30 2020-03-03 Macrogenics, Inc. PD-1-binding molecules and methods of use thereof
WO2020094836A1 (en) 2018-11-09 2020-05-14 Euchloe Bio Pte. Ltd. Il2rbeta/common gamma chain antibodies
WO2020094834A1 (en) 2018-11-09 2020-05-14 Euchloe Bio Pte. Ltd. Il2rbeta/common gamma chain antibodies
US10751414B2 (en) 2016-09-19 2020-08-25 Celgene Corporation Methods of treating psoriasis using PD-1 binding antibodies
US10766958B2 (en) 2016-09-19 2020-09-08 Celgene Corporation Methods of treating vitiligo using PD-1 binding antibodies
WO2021006199A1 (en) 2019-07-05 2021-01-14 小野薬品工業株式会社 Treatment of hematologic cancer with pd-1/cd3 dual specificity protein
WO2021025140A1 (en) 2019-08-08 2021-02-11 小野薬品工業株式会社 Dual-specific protein
US10954301B2 (en) 2015-12-14 2021-03-23 Macrogenics, Inc. Bispecific molecules having immunoreactivity with PD-1 and CTLA-4, and methods of use thereof
US11072653B2 (en) 2015-06-08 2021-07-27 Macrogenics, Inc. LAG-3-binding molecules and methods of use thereof
US11161905B2 (en) 2017-03-04 2021-11-02 Xiangtan Tenghua Bioscience Recombinant antibodies to programmed death 1 (PD-1) and uses thereof
US11285207B2 (en) 2017-04-05 2022-03-29 Hoffmann-La Roche Inc. Bispecific antibodies specifically binding to PD1 and LAG3
WO2022254227A1 (en) 2021-06-04 2022-12-08 Kymab Limited Treatment of pd-l1 negative or low expressing cancer with anti-icos antibodies
US11629189B2 (en) 2017-12-19 2023-04-18 Kymab Limited Bispecific antibody for ICOS and PD-L1
EP4079763A4 (en) * 2019-12-20 2023-10-11 Guangdong Feipeng Pharmaceutical Co., Ltd Anti-human programmed death -1 (pd-1) monoclonal antibody
US11858996B2 (en) 2016-08-09 2024-01-02 Kymab Limited Anti-ICOS antibodies

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201419084D0 (en) 2014-10-27 2014-12-10 Agency Science Tech & Res Anti-PD-1 antibodies
BR112018003186A2 (en) 2015-09-01 2018-09-25 Agenus Inc. anti-pd-1 antibodies and their methods of use
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
MX2019012295A (en) 2017-04-14 2020-02-07 Tollnine Inc Immunomodulating polynucleotides, antibody conjugates thereof, and methods of their use.
WO2019083971A1 (en) 2017-10-23 2019-05-02 Children's Medical Center Corporation Methods of treating cancer using lsd1 inhibitors in combination with immunotherapy
KR102608723B1 (en) * 2018-02-23 2023-12-01 유큐(베이징) 바이오파마 코., 엘티디 Anti-PD-1 antibodies and uses thereof
CN110790839B (en) * 2018-08-03 2023-05-12 江苏恒瑞医药股份有限公司 anti-PD-1 antibody, antigen binding fragment thereof and medical application thereof
JP2022517228A (en) * 2019-01-11 2022-03-07 ザ ウィスター インスティテュート オブ アナトミー アンド バイオロジー DNA monoclonal antibody targeting PD-1 for the treatment and prevention of cancer
EP4110830A1 (en) 2020-02-28 2023-01-04 Tallac Therapeutics, Inc. Transglutaminase-mediated conjugation
JP2024505600A (en) 2021-02-03 2024-02-06 モーツァルト セラピューティクス, インコーポレイテッド Binders and how to use them
WO2023076876A1 (en) 2021-10-26 2023-05-04 Mozart Therapeutics, Inc. Modulation of immune responses to viral vectors

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004056875A1 (en) * 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
WO2006121168A1 (en) * 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
WO2008156712A1 (en) * 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
WO2011159877A2 (en) * 2010-06-18 2011-12-22 The Brigham And Women's Hospital, Inc. Bi-specific antibodies against tim-3 and pd-1 for immunotherapy in chronic immune conditions

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007525434A (en) 2003-03-19 2007-09-06 アブジェニックス インコーポレイテッド Antibodies to T cells, immunoglobulin domain and mucin domain 1 (TIM-1) antigen and uses thereof.
CN101613409B (en) 2005-06-17 2014-06-04 英克隆有限责任公司 PDGFR-alpha antibody
GB0615662D0 (en) 2006-08-07 2006-09-13 Affitech As Antibody
AU2008282152B2 (en) 2007-07-31 2013-12-19 Regeneron Pharmaceuticals, Inc. Human antibodies to human CD20 and method of using thereof
AU2013204861B2 (en) * 2008-09-26 2016-05-12 Dana-Farber Cancer Institute, Inc. Human anti-PD-1, PD-L1, and PD-L2 antibodies and uses therefor
BRPI0917592B1 (en) 2008-12-09 2021-08-17 Genentech, Inc ANTI-PD-L1 ANTIBODY, COMPOSITION, MANUFACTURED ARTICLES AND USES OF A COMPOSITION
EP2366714A1 (en) 2010-03-03 2011-09-21 Dr. Rentschler Holding GmbH & Co. KG Naturally occuring autoantibodies against alpha-synuclein that inhibit the aggregation and cytotoxicity of alpha-synuclein
TW201134488A (en) 2010-03-11 2011-10-16 Ucb Pharma Sa PD-1 antibodies
WO2012004773A1 (en) 2010-07-09 2012-01-12 Universite De Geneve New uses of nogo-a inhibitors and related methods
EP2663331A4 (en) 2011-01-10 2015-07-15 Glaxosmithkline Intellectual Property Man Ltd Novel uses
MX341076B (en) 2011-03-31 2016-08-04 Merck Sharp & Dohme Stable formulations of antibodies to human programmed death receptor pd-1 and related treatments.
KR101970025B1 (en) 2011-04-20 2019-04-17 메디뮨 엘엘씨 Antibodies and other molecules that bind b7-h1 and pd-1
CN102898528B (en) 2012-08-18 2015-06-10 三峡大学 Calreticulin-soluble programmed death receptor 1 fusion protein, and preparation method and purpose thereof
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
KR102229873B1 (en) 2012-10-12 2021-03-19 더 브리검 앤드 우먼즈 하스피털, 인크. Enhancement of the immune response
US10034939B2 (en) 2012-10-26 2018-07-31 The University Of Chicago Synergistic combination of immunologic inhibitors for the treatment of cancer
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
CN111423511B (en) * 2013-05-31 2024-02-23 索伦托药业有限公司 Antigen binding proteins that bind to PD-1
US20160145355A1 (en) 2013-06-24 2016-05-26 Biomed Valley Discoveries, Inc. Bispecific antibodies
WO2015016718A1 (en) 2013-08-02 2015-02-05 Bionovion Holding B.V. Combining cd27 agonists and immune checkpoint inhibition for immune stimulation
GB201419084D0 (en) 2014-10-27 2014-12-10 Agency Science Tech & Res Anti-PD-1 antibodies

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004056875A1 (en) * 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
WO2006121168A1 (en) * 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
WO2008156712A1 (en) * 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
WO2011159877A2 (en) * 2010-06-18 2011-12-22 The Brigham And Women's Hospital, Inc. Bi-specific antibodies against tim-3 and pd-1 for immunotherapy in chronic immune conditions

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BERGER R ET AL.: "Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies.", CLIN CANCER RES, vol. 14, no. 10, 15 May 2008 (2008-05-15), pages 3044 - 3051, XP002524925 *
CHEN Y ET AL.: "Generation and characterization of four novel monoclonal antibodies against human programmed death-1 molecule.", HYBRIDOMA (LARCHMT, vol. 29, no. 2, 29 April 2010 (2010-04-29), pages 153 - 160, XP055439163 *
CHU F ET AL.: "Anti-PD-1 antibodies for the treatment of B- cell lymphoma.", ONCOIMMUNOLOGY, vol. 3, no. 3, 14 February 2014 (2014-02-14), pages e28101 *
See also references of EP3212671A4 *

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11365255B2 (en) 2013-12-12 2022-06-21 Suzhou Suncadia Biopharmaceuticals Co., Ltd. PD-1 antibody, antigen-binding fragment thereof, and medical application thereof
US10344090B2 (en) 2013-12-12 2019-07-09 Shanghai Hangrui Pharmaceutical Co., Ltd. PD-1 antibody, antigen-binding fragment thereof, and medical application thereof
US10160806B2 (en) 2014-06-26 2018-12-25 Macrogenics, Inc. Covalently bonded diabodies having immunoreactivity with PD-1 and LAG-3, and methods of use thereof
US11098119B2 (en) 2014-06-26 2021-08-24 Macrogenics, Inc. Covalently bonded diabodies having immunoreactivity with PD-1 and LAG-3, and methods of use thereof
US11072653B2 (en) 2015-06-08 2021-07-27 Macrogenics, Inc. LAG-3-binding molecules and methods of use thereof
US11858991B2 (en) 2015-06-08 2024-01-02 Macrogenics, Inc. LAG-3-binding molecules and methods of use thereof
US11623959B2 (en) 2015-07-30 2023-04-11 Macrogenics, Inc. PD-1-binding molecules and methods of use thereof
US10577422B2 (en) 2015-07-30 2020-03-03 Macrogenics, Inc. PD-1-binding molecules and methods of use thereof
US10428145B2 (en) 2015-09-29 2019-10-01 Celgene Corporation PD-1 binding proteins and methods of use thereof
US11840571B2 (en) 2015-12-14 2023-12-12 Macrogenics, Inc. Methods of using bispecific molecules having immunoreactivity with PD-1 and CTLA-4
US10954301B2 (en) 2015-12-14 2021-03-23 Macrogenics, Inc. Bispecific molecules having immunoreactivity with PD-1 and CTLA-4, and methods of use thereof
WO2017220990A1 (en) 2016-06-20 2017-12-28 Kymab Limited Anti-pd-l1 antibodies
WO2017220988A1 (en) 2016-06-20 2017-12-28 Kymab Limited Multispecific antibodies for immuno-oncology
WO2017220989A1 (en) 2016-06-20 2017-12-28 Kymab Limited Anti-pd-l1 and il-2 cytokines
KR20190029641A (en) * 2016-07-13 2019-03-20 조인트 스탁 컴퍼니 "바이오케드" Anti-PD-1 antibody, its production method and use
RU2656181C1 (en) * 2016-07-13 2018-05-31 Закрытое Акционерное Общество "Биокад" Anti-pd-1 antibodies, method for their production, and method of application
WO2018013017A1 (en) * 2016-07-13 2018-01-18 Закрытое Акционерное Общество "Биокад" Anti-pd-1 antibodies, method for producing same and method for using same
JP6993992B2 (en) 2016-07-13 2022-01-14 ジョイント・ストック・カンパニー “バイオキャド” Anti-PD-1 antibody, its production method and its usage method
JP2019527543A (en) * 2016-07-13 2019-10-03 ジョイント ストック カンパニー ”バイオカッド” Anti-PD-1 antibody, production method thereof and use method thereof
KR102482710B1 (en) 2016-07-13 2023-01-02 조인트 스탁 컴퍼니 “바이오케드” Anti-PD-1 antibodies, methods of producing and using the same
CN110023335A (en) * 2016-07-13 2019-07-16 “比奥卡特”封闭式股份公司 Anti- PD-1 antibody, its production method and its application method
US11136408B2 (en) 2016-07-13 2021-10-05 Joint Stock Company “Biocad” Anti-PD-1 antibodies, method for producing same and method for using same
US11858996B2 (en) 2016-08-09 2024-01-02 Kymab Limited Anti-ICOS antibodies
JP2018029587A (en) * 2016-08-17 2018-03-01 治範 小田 Method of culturing lymphocyte having improved cytotoxic activity, and cellular immunological therapeutic agent containing lymphocyte having improved cytotoxic activity obtained by the method
JP6993812B2 (en) 2016-08-17 2022-01-14 治範 小田 A method for culturing lymphocytes with improved cytotoxic activity and a cell immunotherapeutic agent containing lymphocytes with improved cytotoxic activity obtained by the method.
US10766958B2 (en) 2016-09-19 2020-09-08 Celgene Corporation Methods of treating vitiligo using PD-1 binding antibodies
US10751414B2 (en) 2016-09-19 2020-08-25 Celgene Corporation Methods of treating psoriasis using PD-1 binding antibodies
US11161905B2 (en) 2017-03-04 2021-11-02 Xiangtan Tenghua Bioscience Recombinant antibodies to programmed death 1 (PD-1) and uses thereof
US11285207B2 (en) 2017-04-05 2022-03-29 Hoffmann-La Roche Inc. Bispecific antibodies specifically binding to PD1 and LAG3
WO2019092181A1 (en) 2017-11-10 2019-05-16 Agency For Science, Technology And Research Il2rbeta/common gamma chain antibodies
US11629189B2 (en) 2017-12-19 2023-04-18 Kymab Limited Bispecific antibody for ICOS and PD-L1
WO2020094834A1 (en) 2018-11-09 2020-05-14 Euchloe Bio Pte. Ltd. Il2rbeta/common gamma chain antibodies
WO2020094836A1 (en) 2018-11-09 2020-05-14 Euchloe Bio Pte. Ltd. Il2rbeta/common gamma chain antibodies
WO2021006199A1 (en) 2019-07-05 2021-01-14 小野薬品工業株式会社 Treatment of hematologic cancer with pd-1/cd3 dual specificity protein
WO2021025140A1 (en) 2019-08-08 2021-02-11 小野薬品工業株式会社 Dual-specific protein
EP4079763A4 (en) * 2019-12-20 2023-10-11 Guangdong Feipeng Pharmaceutical Co., Ltd Anti-human programmed death -1 (pd-1) monoclonal antibody
WO2022254227A1 (en) 2021-06-04 2022-12-08 Kymab Limited Treatment of pd-l1 negative or low expressing cancer with anti-icos antibodies

Also Published As

Publication number Publication date
CN107438621A (en) 2017-12-05
MY191581A (en) 2022-06-30
US20190292261A1 (en) 2019-09-26
RU2017116847A3 (en) 2019-04-22
US20170240635A1 (en) 2017-08-24
EP3212671A4 (en) 2018-05-30
US20170313774A1 (en) 2017-11-02
AU2015340054A1 (en) 2017-06-01
PH12017500803A1 (en) 2017-10-02
CA2965623A1 (en) 2016-05-06
RU2017116847A (en) 2018-11-30
IL251963A0 (en) 2017-06-29
JP2018500924A (en) 2018-01-18
CN107438621B (en) 2021-10-22
JP6701213B2 (en) 2020-05-27
US11072659B2 (en) 2021-07-27
KR20170070243A (en) 2017-06-21
US10280224B2 (en) 2019-05-07
EP3212671A1 (en) 2017-09-06
US9771425B2 (en) 2017-09-26
SG11201703344WA (en) 2017-05-30
GB201419084D0 (en) 2014-12-10
RU2715628C2 (en) 2020-03-02

Similar Documents

Publication Publication Date Title
US11072659B2 (en) Anti-PD-1 antibodies
US11142574B2 (en) Anti-TIM-3 antibodies
US11534489B2 (en) Anti-PD-L1 antibodies
US20200055935A1 (en) Anti-TIM-3 Antibodies
EP3455255B1 (en) Anti-ctla-4 antibodies
US20190040136A1 (en) Anti-lag-3 antibodies

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15855595

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
ENP Entry into the national phase

Ref document number: 2965623

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 15521434

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 11201703344W

Country of ref document: SG

ENP Entry into the national phase

Ref document number: 2017540959

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 251963

Country of ref document: IL

REEP Request for entry into the european phase

Ref document number: 2015855595

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20177014305

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017116847

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2015340054

Country of ref document: AU

Date of ref document: 20151027

Kind code of ref document: A