US20220251195A1 - Anti-tigit antibodies and their use as therapeutics and diagnostics - Google Patents

Anti-tigit antibodies and their use as therapeutics and diagnostics Download PDF

Info

Publication number
US20220251195A1
US20220251195A1 US17/675,515 US202217675515A US2022251195A1 US 20220251195 A1 US20220251195 A1 US 20220251195A1 US 202217675515 A US202217675515 A US 202217675515A US 2022251195 A1 US2022251195 A1 US 2022251195A1
Authority
US
United States
Prior art keywords
seq
antibody
chain variable
amino acid
tigit
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/675,515
Inventor
Tong Zhang
Liu Xue
Qi Liu
Min Wei
Kang Li
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Beigene Switzerland GmbH
Original Assignee
Beigene Switzerland GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Beigene Switzerland GmbH filed Critical Beigene Switzerland GmbH
Priority to US17/675,515 priority Critical patent/US20220251195A1/en
Assigned to BEIGENE SWITZERLAND GMBH reassignment BEIGENE SWITZERLAND GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BEIGENE, LTD.
Assigned to BEIGENE, LTD. reassignment BEIGENE, LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LIU, QI, WEI, MIN, ZHANG, TONG, XUE, Liu, LI, KANG
Publication of US20220251195A1 publication Critical patent/US20220251195A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/54F(ab')2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins 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
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present application relates to antibodies that specifically bind to TIGIT (T cell immunoreceptor with Ig and ITIM domains) and uses of the same.
  • Tigit T cell immunoglobulin and ITIM domain
  • CD28 CD28 family of proteins that plays an important role in inhibiting T- and NK cell-mediated functional activities in anti-tumor immunity [Boles K S, et al., 2009 Eur Immunol, 39:695-703; Stanietsky N, et al., 2009 PNAS 106:17858-63; Yu X et al. 2009 Nat. Immunol, 10:48-57].
  • TIGIT The genes and cDNAs coding for TIGIT were cloned and characterized in mouse and human.
  • Full length human TIGIT has a sequence of 244 amino acids (SEQ ID NO: 1) in length, in which the first 21 amino acids consist a signal peptide.
  • the amino acid sequence of the mature human TIGIT contains 223 amino acid (aa) residues (NCBI accession number: NM_173799).
  • the extracellular domain (ECD) of mature human TIGIT consists of 120 amino acid residues (SEQ ID NO: 2, corresponding to amino acids 22-141 of SEQ ID NO: 1) with a V-type Ig-like domain (corresponding to amino acids 39-127 of SEQ ID NO: 1), followed by a 21 aa transmembrane sequence, and an 82 aa cytoplasmic domain with an immunoreceptor tyrosine-based inhibitory motif (ITIM) [Yu X et al. 2009 Nat. Immunol, 10:48-57: Stengel K F, et al. 2012 PNAS 109:5399-04].
  • ITIM immunoreceptor tyrosine-based inhibitory motif
  • TIGIT is expressed on T cells (including activated T cells, memory T cells, regulatory T (Treg) cells, and follicular T helper (Tfh) cells), and NK cells [Boles K S, et al., 2009 Eur J Immunol 39:695-703; Joller N, et al., 2014 Immunity 40:569-81; Levin S D, et al. 2011 Eur Immunol, 41:902-15; Stanietsky N, et al. 2009 PNAS 106:17858-63; Yu X et al. 2009 Nat. Immunol, 10:48-57].
  • CD155 also known as poliovirus receptor or PVR
  • CD112 also known as poliovirus receptor-related 2, PVRL2, nectin-2
  • APCs such as dendritic cells and macrophages
  • tumor cells [Casado J G, et al., 2009 Cancer Immunol Immunother 58:1517-26; Levin S D, et al., 2011 Eur J Immunol, 41:902-15; Mendelsohn C L et al., 1989 56:855-65; Stanietsky N, et al., 2009 PNAS 106:17858-63; Yu X et al.
  • Tigit initiates inhibitory signaling in immune cells when engaged by its ligands, CD155 and CD112.
  • the binding affinity of Tigit to CD155 (Kd: ⁇ 1 nM) is much higher than to CD112 and whether the TIGIT: CD112 interaction is functionally relevant in mediating inhibitory signals yet remain to be determined.
  • a co-stimulatory receptor, CD226 (DNAM-1), binds to the same ligands with lower affinity (Kd: ⁇ 100 nM), but delivers a positive signal [Bottino C, et al., 2003 JExp Med 198:557-67].
  • CD96 Tactile
  • a “Tigit-like” receptor also plays a similarly inhibitory role in the same pathway [Chan C J, et al., 2014 Nat. Immunol 15:431-8].
  • Tigit can inhibit immune responses through different mechanisms.
  • interaction between TIGIT and PVR on dendritic cells (DCs) could deliver a “reverse signaling” in DCs, leading to up-regulation of IL-10 and decrease of IL-12 secretion, thereby inhibiting T-cell activation [Yu X et al. Nat Immunol. 2009 10:48-57].
  • TIGIT binds to CD155 with higher affinity, thereby competing off DNAM-1-CD155 interaction.
  • direct ligation of TIGIT on T cells could down-regulate TCR-mediated activation and subsequent proliferation and engagement of TIGIT on NK cells block NK cell cytotoxicity [Joller N, et al.
  • Tregs has been associated with a highly activated and suppressive phenotype in tumor tissue and TIGIT signaling in Tregs may favor Treg stability [Joller N, et al. Immunity 2014 40:569-81; Kurtulus S, et al. J Clin Invest. 2015 125: 4053-4062].
  • TIGIT has an immunoglobulin tail tyrosine (ITT)-like motif followed by an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic tail [Yu X et al. Nat Immunol. 2009 10:48-57; Engels N, et al. Curr Opin Immunol 2011 23: 324-329].
  • ITIM immunoreceptor tyrosine-based inhibition motif
  • TILs tumor-infiltrating lymphocytes
  • PBMCs peripheral blood mononuclear cells
  • Tigit in AML is associated with poor prognosis of patient survival outcome [Kong Y et al. Clin Cancer Res, 2016 22:3057-66]. Not only does up-regulation of Tigit signaling play important roles in immune tolerance to cancer, but also to chronic viral infection. During HIV infection, expression of Tigit on T cells was significantly higher and positively correlated with viral loads and disease progression [Chew G M, et al., 2016 PLoS Pathog. 12:e1005349]. In addition, blockade of Tigit receptor alone or in combination with other blockade could rescue functionally “exhausted” T cells both in vitro and in vivo [Chauvin J M, et al., J Clin Invest.
  • Tigit-mediated inhibitory signaling may restore the functional activities of immune cells including T cells, NK cells and dendritic cells (DCs), therefore enhancing immunity against cancer or chronic viral infection.
  • modulation of Tigit signaling by antagonistic molecules may rescue immune cells from tolerance, inducing efficient immune responses to eradicate tumors or chronic viral infections.
  • the present invention is at least in part based on the discovery of a set of monoclonal antibodies (mAbs) which inhibit Tigit-mediated cellular signaling in immune cells, re-activate the immune cells and enhance immunity by specifically binding to Tigit.
  • mAbs monoclonal antibodies
  • the present application relates to an anti-Tigit antibody and antigen-binding fragment thereof which is capable of binding to human Tigit (SEQ ID NO: 1).
  • the present invention also relates to the humanized version of the anti-Tigit mAbs of the first aspect.
  • the antibody of the present application comprises a heavy chain variable region (VH) comprising one, two or three CDRs having an amino acid sequence selected from SEQ ID NOs: 3, 4, 5 or 13, or variants thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions in the amino acid sequences of SEQ ID NOs 3, 4, 5 or 13; and/or a light chain variable region (VL) comprising one, two or three CDRs having an amino acid sequence selected from SEQ ID NOs: 6, 7 or 8, or variants thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions in the amino acid sequences of SEQ ID NOs: 6, 7, or 8.
  • VH heavy chain variable region
  • VL light chain variable region
  • the antibody of the present application comprises a heavy chain variable region (VH) comprising a VH-CDRT having an amino acid sequence of SEQ ID NO: 3 or a variant thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions, a VH-CDR2 having an amino acid sequence of SEQ ID NO: 4 or SEQ ID NO: 13 or a variant thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions, and a VH-CDR3 having an amino acid sequence of SEQ ID NO: 5 or a variant thereof comprising one or more conservative substitutions, e.g.
  • VH heavy chain variable region
  • VL light chain variable region
  • VL-CDR1 having an amino acid sequence of SEQ ID NO: 6 or a variant thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions, a VL-CDR2 having an amino acid sequence of SEQ ID NO: 7 or a variant thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions, and a VL-CDR3 having an amino acid sequence of SEQ ID NO: 8 or a variant thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions.
  • VL-CDR1 having an amino acid sequence of SEQ ID NO: 6 or a variant thereof comprising one or more conservative substitutions
  • VL-CDR2 having an amino acid sequence of SEQ ID NO: 7 or a variant thereof comprising one or more conservative substitutions
  • VL-CDR3 having an amino acid sequence of SEQ ID NO: 8 or a variant thereof comprising one or more conservative substitutions, e.g. one or two conservative
  • the antibody or the antigen-binding fragment thereof of the present application is capable of binding to human Tigit and comprises a heavy chain variable region having an amino acid sequence selected from SEQ ID NO: 9, 14, 19, or a sequence having at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% sequence identity with SEQ ID NO: 9, 14, 19.
  • the difference in sequence lies in the framework region.
  • the antibody or the antigen-binding fragment thereof comprises a heavy chain variable region encoded by an nucleotide sequence selected from SEQ ID NO: 10, 15 or 20, or a variant thereof.
  • the antibody or the antigen-binding fragment thereof of the present application is capable of binding to human Tigit and comprises a heavy chain variable region having an amino acid sequence selected from SEQ ID NO: 11, 16, 21, or 24, or a sequence having at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% sequence identity with SEQ ID NO: 11, 16, 21 or 24.
  • the difference in sequence lies in the framework ⁇ region.
  • the antibody or the antigen-binding fragment thereof comprising a heavy chain variable region encoded by an nucleotide sequence selected from SEQ ID NO: 12, 17 or 22, or a variant thereof.
  • the antibody or antigen-binding fragment thereof is capable of binding to human Tigit with a Kd value of about 1 ⁇ 10 ⁇ 9 M to about 1 ⁇ 10 ⁇ 12 M.
  • the antibody or the antigen-binding fragment thereof is capable of binding to human Tigit with a Kd value less than about 1 ⁇ 10 ⁇ 9 M, less than about 1 ⁇ 10 ⁇ 10 M, less than about 1 ⁇ 10 ⁇ 11 M, or less than about 1 ⁇ 10 ⁇ 12 M.
  • the antibody or the antigen-binding fragment thereof comprises a heavy chain constant region of the subclass of IgG1, IgG2, IgG3, or IgG4 or a variant thereof, and a light chain constant region of the type of kappa or lambda or a variant thereof.
  • the Fe region of the antibody is human IgG1 Fe or a variant thereof, e.g. a Fe region of SEQ ID NO: 18.
  • the antibody or antigen-binding fragment thereof promotes the production of IFN- ⁇ by antigen-specific T cells. In a more specific embodiment, the antibody or antigen-binding fragment thereof promotes the production of IFN- ⁇ by antigen-specific T cells in a dose-dependent manner.
  • the antibody of the present application reduces the surface expression of Tigit receptor via Fc ⁇ R-mediated trogocytosis, particularly Fc ⁇ RIIB-mediated trogocytosis.
  • the antibody of the present application shows a pH-dependent antigen binding such that the antibody exhibits a stronger binding to human TIGIT at a mild acidic pH in tumor microenvironment (e.g. pH 6.0) as compared to the binding to human TIGIT at a physiologic pH (e.g. pH 7.4).
  • a mild acidic pH in tumor microenvironment e.g. pH 6.0
  • a physiologic pH e.g. pH 7.4
  • the antibody of the present application has (1) a K D ratio at pH 7.4/pH 6.0 of greater than 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, or more, and/or (2) a Rmax (RU) value at pH 6.0 which is at least 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold higher than the Rmax at pH 7.4, as measured by surface plasmon resonance (Biacore) or similar technology.
  • a K D ratio at pH 7.4/pH 6.0 of greater than 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, or more
  • a Rmax (RU) value at pH 6.0 which is at least 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold higher than
  • the anti-Tigit mAbs disclosed herein have potential therapeutic uses in treating cancer, controlling viral infections and other human diseases that are mechanistically involved in immune tolerance or “exhaustion”. Accordingly, in further embodiments, the anti-Tigit antibody of the present application is for use in treatment of cancer. In another specific embodiment, the anti-Tigit antibody of the present application is for use in treating an infection, treating an infectious disease and/or controlling viral infections. In another specific embodiment, the anti-Tigit antibody of the present application is for use in the treatment of other human diseases related to or caused by immune tolerance, or the treatment of a disease that can be improved by increasing immune cell activation.
  • the present application relates to a composition
  • a composition comprising the anti-Tigit antibody or antigen-binding fragment thereof and a therapeutically acceptable excipient.
  • FIG. 1 Schematic diagram of Tigit-mIgG2a (top) and Tigit-huIgG1 (bottom).
  • Tigit ECD tigit extracellular domain
  • N N-terminus
  • C C-terminus.
  • FIG. 2A, 2B Phylogenetic trees of anti-Tigit antibody Vh ( 2 A) and Vk ( 2 B) regions.
  • the Vh and Vk sequences of candidate anti-Tigit antibodies were aligned using DNASTAR's Megalign software. Sequence homology was displayed in phylogenetic trees.
  • FIG. 3 Affinity determination of purified murine anti-Tigit antibodies by surface plasmon resonance (SPR).
  • FIG. 4 Determination of Tigit binding by flow cytometry.
  • FIG. 5A A schematic diagram showing the inhibition of Tigit-ligand interactions by anti-Tigit mAbs.
  • FIG. 5B The binding of soluble Tigit (Tigit-huIgG1 fusion protein) to Tigit ligand-expressing HEK293 cells (HEK293/PVR or HEK293/PVR-L2) was determined by flow cytometry. The blockade of Tigit-ligand interaction was quantitatively measured by adding serially diluted anti-Tigit antibodies. Results were shown in mean ⁇ SD of duplicates.
  • FIG. 6 Activation of CMV-specific human T cells by anti-Tigit mAbs.
  • Human CMV peptide NLVPMVATV, 495-503-sensitized HLA-A2.1 + PBMCs (4 ⁇ 10 4 ) were stimulated with the CMV peptide-pulsed target cells HCT116 cells (10 4 ) overnight in the presence of anti-Tigit antibodies.
  • IFN- ⁇ in the culture supernatant was determined by ELISA. All conditions were performed in triplicates. Results were shown as mean ⁇ SD.
  • FIG. 7 Anti-Tigit mAbs promote NK cell-mediated cytotoxicity.
  • A Tigit and DNAM-1 expression on engineered NK92MI/Tigit-DNAM-1 stable cell line.
  • B Killing of NK92MI/Tigit-DNAM-1 cells against SK-MES-1/PVR cells in the presence of hu1217-2-2/IgG1mf (0.007-30 ⁇ g/ml) was determined by an LDH (lactate dehydrogenase) release assay as described in Example 8. Results were shown in mean ⁇ SD of triplicates.
  • FIG. 8 Anti-Tigit mAb hu1217-2-2/IgG1wt reduces the surface expression of Tigit receptor via Fc ⁇ R-mediated trogocytosis.
  • Jurkat/Tigit cells were incubated with Fc ⁇ R-expressing HEK293 cells in the presence of biotin-labeled anti-Tigit mAbs in complete media overnight.
  • 10% human AB serum was added to determine the effects of bulk human IgG on trogocytosis.
  • Surface expression of Tigit receptor was determined by staining with SA-APC (Biolegend). MFI was determined by flow cytometry. All data points were in duplicates. Results were shown in mean ⁇ SD.
  • FIG. 9 ADCC effects of anti-Tigit mAbs on human peripheral blood mononuclear cells (PBMCs).
  • PBMCs peripheral blood mononuclear cells
  • (B) ADCC assay was performed using a CD16 + human NK cell line NK92MI/CD16V as effector cells and PHA-stimulated PBMCs as target cells in the presence of Tigit mAbs (30 ⁇ g/mL) or control antibodies (OKT3 at 5 ⁇ g/ml as a positive control, and huIgG at 30 ⁇ g/mL as a negative control) for 42 hrs. Percentages of CD3 + , CD8 ⁇ T cells and Tregs were determined by flow cytometry.
  • FIG. 10 CDC effects of anti-Tigit mAbs on human PBMCs.
  • CDC assay was performed using PHA-stimulated PBMCs as target cells and autologous sera as the source of complements. After 3 days of co-culture of pre-activated PBMCs with antiTigit mAbs (0.01-100 ⁇ g/ml) in the final concentration of 15% autologous sera, percentage of CDC (y-axis) was measured by cell-titer glow assay, and calculated as described in Example 11. Data from donors A and B are shown. HuIgG were used as a negative control, whereas anti-MHC-A, B, C was used as a positive control.
  • amino acids are commonly known in the art and exemplarily shown in the table below. Generally, a conservative amino acid substitution means that an amino acid residue is replaced by another amino acid residue having a similar side chain.
  • Tigit includes various mammalian isoforms, e.g., human Tigit, orthologs of human Tigit, and analogs comprising at least one epitope within Tigit.
  • the amino acid sequence of Tigit, e.g., human Tigit, and the nucleotide sequence encoding the same, is known in the art.
  • administration when applied to an animal, human, experimental subject, cell, tissue, organ, or biological fluid, mean contact of an exogenous pharmaceutical, therapeutic, diagnostic agent, or composition to the animal, human, subject, cell, tissue, organ, or biological fluid.
  • Treatment of a cell encompasses contact of a reagent to the cell, as well as contact of a reagent to a fluid, where the fluid is in contact with the cell.
  • administration or “treatment” also includes in vitro and ex vivo treatments, e.g., of a cell, by a reagent, diagnostic, binding compound, or by another cell.
  • subject herein refers to any organism, preferably an animal, more preferably a mammal (e.g., rat, mouse, dog, cat, rabbit) and most preferably a human.
  • antibody molecules that bind to Tigit with high affinity and specificity.
  • the anti-Tigit antibody binds to human Tigit and includes at least one, two, three, four, five or six complementarity determining regions (CDR's) comprising an amino acid sequence SEQ ID NOs 3, 4, 5, 13 or SEQ ID NOs: 6, 7, 8.
  • the antibody of the present application comprises a heavy chain variable region (VH) comprising one, two or three CDRs having an amino acid sequence selected from SEQ, ID NOs: 3, 4, 5 or 13, or a variant thereof comprising one or more conservative substitutions; and/or a light chain variable region (VL) comprising one, two or three CDRs having an amino acid sequence selected from SEQ ID NOs: 6, 7, or 8, or a variant thereof comprising one or more conservative substitutions.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-Tigit antibody is an isolated antibody, a humanized antibody, a chimeric antibody or a recombinant antibody.
  • the anti-Tigit antibody comprises at least one antigen-binding site, or at least a variable region. In some embodiments, the anti-Tigit antibody comprises an antigen-binding fragment derived from an antibody described herein.
  • antibody herein is used in the broadest sense and specifically covers antibodies (including full length monoclonal antibodies) and antibody fragments so long as they recognize antigen, e.g., Tigit.
  • An antibody is usually monospecific, but may also be described as idiospecific, heterospecific, or polyspecific.
  • Antibody molecules bind by means of specific binding sites to specific antigenic determinants or epitopes on antigens.
  • mAb monoclonal antibody
  • mAb monoclonal antibody
  • Mab monoclonal antibody
  • conventional (polyclonal) antibody preparations typically include a multitude of different antibodies having different amino acid sequences in their variable domains, particularly their complementarity determining regions (CDRs), which are often specific for different epitopes.
  • CDRs complementarity determining regions
  • Monoclonal antibodies indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method, Monoclonal antibodies (mAbs) may be obtained by methods known to those skilled in the art. See, for example Kohler G et al., Nature 1975 256:495-497; U.S. Pat. No.
  • the mAbs disclosed herein may be of any immunoglobulin class including IgG, IgM, IgD, IgE, IgA, and any subclass thereof.
  • a hybridoma producing a mAb may be cultivated in vitro or in vivo.
  • High titers of mAbs can be obtained by in vivo production where cells from the individual hybridomas are injected intraperitoneally into mice, such as pristine-primed Balb/c mice to produce ascites fluid containing high concentrations of the desired mAbs, MAbs of isotype IgM or IgG may be purified from such ascites fluids, or from culture supernatants, using column chromatography methods well known to those of skill in the art.
  • the basic antibody structural unit comprises a tetramer.
  • Each tetramer includes two identical pairs of polypeptide chains, each pair having one “light chain” (about 25 kDa) and one “heavy chain” (about 50-70 kDa).
  • the amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the carboxy-terminal portion of the heavy chain may define a constant region primarily responsible for effector function.
  • human light chains are classified as kappa and lambda light chains.
  • human heavy chains are typically classified as ⁇ , ⁇ , ⁇ , ⁇ , or ⁇ , and define the antibody's isotypes as IgA, IgD, IgE, IgG, and IgM, respectively.
  • the variable and constant regions are joined by a “J” region of about 12 or more amino acids, with the heavy chain also including a “D” region of about 10 more amino acids.
  • variable regions of each light/heavy chain (VL/VH) pair form the antibody binding site.
  • an intact antibody has two binding sites.
  • the two binding sites are, in general, the same.
  • variable domains of both the heavy and light chains comprise three hypervariable regions, also called “complementarity determining regions (CDRs)”, which are located between relatively conserved framework regions (FR).
  • CDRs complementarity determining regions
  • FR framework regions
  • the CDRs are usually aligned by the framework regions, enabling binding to a specific epitope.
  • both light and heavy chain variable domains sequentially comprise FR-1 (or FR1), CDR-1 (or CDR1), FR-2 (FR2), CDR-2 (CDR2), FR-3 (or FR3) CDR-3 (CDR3) and FR-4 (or FR4).
  • hypervariable region means the amino acid residues of an antibody that are responsible for antigen-binding.
  • the hypervariable region comprises amino acid residues from a “CDR” (i.e., CDR1, VL-CDR2 and VL-CDR3 in the light chain variable domain and VH-CDR1, VH-CDR2 and VH-CDR3 in the heavy chain variable domain).
  • CDR i.e., CDR1, VL-CDR2 and VL-CDR3 in the light chain variable domain and VH-CDR1, VH-CDR2 and VH-CDR3 in the heavy chain variable domain.
  • antibody fragment or “antigen-binding fragment” means antigen binding fragments of antibodies, i.e. antibody fragments that retain the ability to bind specifically to the antigen bound by the full-length antibody, e.g, fragments that retain one or more CDR regions.
  • antigen binding fragments include, but not limited to, Fab, Fab′, F(ab′)2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules, e.g., single chain Fv (ScFv); nanobodies and multispecific antibodies formed from antibody fragments.
  • An antibody that binds to a specified target protein with specificity is also described as specifically binding to a specified target protein. This means the antibody exhibits preferential binding to that target as compared to other proteins, but this specificity does not require absolute binding specificity.
  • An antibody is considered “specific” for its intended target if its binding is determinative of the presence of the target protein in a sample, e.g. without producing undesired results such as false positives.
  • Antibodies or binding fragments thereof, useful in the present invention will bind to the target protein with an affinity that is at least two fold greater, preferably at least 10-times greater, more preferably at least 20-times greater, and most preferably at least 100-times greater than the affinity with non-target proteins.
  • An antibody herein is said to bind specifically to a polypeptide comprising a given amino acid sequence, e.g. the amino acid sequence of a mature human Tigit molecule, if it binds to polypeptides comprising that sequence but does not bind to proteins lacking that sequence.
  • pH-dependent binding binds to its target/antigen, namely human TIGIT, in a pH-dependent manner.
  • the antibody of the present application shows a higher binding affinity and/or binding signal to its antigen at a mild acidic pH, e.g. pH 6.0, which is usually found in tumor microenvironment, as compared to the binding affinity and/or binding signal at physiologic pH, e.g. pH 7.4.
  • the methods for determining the binding affinity and/or the intensity of binding signal of the antibody of the present application are well known in the art and include but not limited to surface plasmon resonance (Biacore) or similar technology. More specifically, the antibody of the present application has a K D ratio at pH 7.4/pH 6.0 of greater than 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, or more, as measured by surface plasmon resonance (Biacore) or similar technology.
  • the antibody of the present application has a Rmax (RU) value at pH 6.0 which is at least 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold higher than the Rmax at pH 7.4 as measured by surface plasmon resonance (Biacore) or similar technology.
  • the binding affinity of the antibody can be measured at 25° C. or 37° C. Tumor microenvironment has been found to show a relatively more acidic pH than physiological condition or normal tissues (Zhang et al. Focus on molecular Imaging 2010; Tannock and Rotin et al, Cancer Res 1989).
  • the antibody of the present application having above-mentioned pH-dependent binding is advantageous as an anti-TIGIT therapeutic agent for targeting TIGIT-positive lymphocytes in the tumor microenvironment with selectivity and having lower toxicity associated with periphery activation of lymphocytes.
  • human antibody herein means an antibody that comprises human immunoglobulin protein sequences only.
  • a human antibody may contain murine carbohydrate chains if produced in a mouse, in a mouse cell, or in a hybridoma derived from a mouse cell.
  • mouse antibody or “rat antibody” means an antibody that comprises only mouse or rat immunoglobulin protein sequences, respectively.
  • humanized antibody means forms of antibodies that contain sequences from non-human (e.g., murine) antibodies as well as human antibodies. Such antibodies contain minimal sequence derived from non-human immunoglobulin.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • the prefix “hum”, “hu, “Hu” or “h” is added to antibody clone designations when necessary to distinguish humanized antibodies from parental rodent antibodies.
  • the humanized forms of rodent antibodies will generally comprise the same CDR sequences of the parental rodent antibodies, although certain amino acid substitutions may be included to increase affinity, increase stability of the humanized antibody, or for other reasons.
  • cancer means or describes the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • cancer include but are not limited to, lung cancer (including small-cell lung cancer, or non-small cell lung cancer), adrenal cancer, liver cancer, stomach cancer, cervical cancer, melanoma, renal cancer, breast cancer, colorectal cancer, leukemia, bladder cancer, bone cancer, brain cancer, an endometrial cancer, head and neck cancer, lymphoma, ovarian cancer, skin cancer, thyroid tumor, or metastatic lesion of the cancer.
  • the antibody of the present application has potential therapeutic uses in controlling viral infections and other human diseases that are mechanistically involved in immune tolerance or “exhaustion”.
  • exhaust refers to a process which leads to a depleted ability of immune cells to respond to the infecting virus during a prolonged period of chronic viral infection.
  • compositions e.g., pharmaceutically acceptable compositions, which include an anti-Tigit-3 antibody described herein, formulated together with at least one pharmaceutically acceptable excipient.
  • pharmaceutically acceptable excipient includes any and all solvents, dispersion media, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • the excipient can be suitable for intravenous, intramuscular, subcutaneous, parenteral, rectal, spinal or epidermal administration (e.g. by injection or infusion).
  • compositions herein may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusion solutions), dispersions or suspensions, liposomes, and suppositories.
  • liquid solutions e.g., injectable and infusion solutions
  • dispersions or suspensions e.g., dispersions or suspensions
  • liposomes e.g., liposomes, and suppositories.
  • a suitable form depends on the intended mode of administration and therapeutic application. Typical suitable compositions are in the form of injectable or infusion solutions.
  • One suitable mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular).
  • the antibody is administered by intravenous infusion or injection.
  • the antibody is administered by intramuscular or subcutaneous injection.
  • terapéuticaally effective amount refers to the amount of an antibody that, when administered to a subject for treating a disease or a disorder, or at least one of the clinical symptoms of a disease or disorder, is sufficient to effect such treatment for the disease, disorder, or symptom.
  • the “therapeutically effective amount” can vary with the antibody, the disease, disorder, and/or symptoms of the disease or disorder, severity of the disease, disorder, and/or symptoms of the disease or disorder, the age of the subject. to be treated, and/or the weight of the subject to be treated. An appropriate amount in any given instance can be apparent to those skilled in the art or can be determined by routine experiments.
  • the “therapeutically effective amount” refers to the total amount of the active agents comprised in the combination for the effective treatment of a disease, a disorder or a condition.
  • the “subject” as used herein is a mammal, e.g., a rodent or a primate, preferably a higher primate, e.g., a human (e.g., a patient having, or at risk of having, a disorder described herein).
  • Anti-Tigit monoclonal antibodies were generated based on conventional hybridoma fusion technology [de StGroth and Sheidegger 1980 J Immunol Methods 35:1; Mechetner; 2007 Methods Mol Biol 378:1] with minor modifications.
  • the mAbs with high binding activity in enzyme-linked immunosorbent assay (ELISA) and fluorescence-activated cell sorting (FACS) assay were selected for further characterization.
  • the cDNA coding for the full-length human Tigit was synthesized by and purchased from Sino Biological (Beijing, China) based on its GenBank sequence (Accession No: NM_173799).
  • the coding region of extracellular domain (ECD) of the full-length human Tigit corresponding to the amino acid (AA) 1-141 of SEQ ID NO: 1 was PCR-amplified, and cloned into pcDNA3.1-based expression vector (Invitrogen, Carlsbad, Calif., USA) with C-terminus fused either to the Fc domain of mouse IgG2a or to the Fc domain of human IgG1 heavy chain, which resulted in two recombinant fusion protein expression plasmids, Tigit-mIgG2a and Tigit-huIgG1, respectively.
  • Tigit fusion proteins were shown in FIG. 1 .
  • Tigit-mIgG2a and Tigit-huIgG1 plasmids were transiently transfected into 293G cells (developed in-house) and cultured for 7 days in a C02 incubator equipped with rotating shaker. The supernatant containing the recombinant protein was collected and cleared by centrifugation.
  • Tigit-mIgG2a and Tigit-huIgG1 were purified using a Protein A column (Cat: 17127901, GE Life Sciences). Both Tigit-mIgG2a and Tigit-hulgG1 proteins were dialyzed against phosphate buffered saline (DPBS) and stored in ⁇ 80° C. freezer in small aliquots.
  • DPBS phosphate buffered saline
  • Tigit genes (synthesized by Genescript, Nanjing, China) were cloned into a retroviral vector pFB-Neo (Cat.: 217561, Agilent, USA). Dual-tropic retroviral vectors were generated according to a previous protocol [Zhang T, et al. 2005 , Blood ].
  • Vectors containing huTigit and mkTigit were transduced into Jurkat and NK92MI cells (ATCC, Manassas, Va., USA), respectively, to generate the cell lines, Jurka/huTigit and NK92MI/mkTigit.
  • the high expression cell lines were selected by cultivation in medium with G418 and PACS binding assay.
  • mice Eight to twelve week-old Balb/c mice (from HFK BIOSCIENCE CO., LTD, Beijing, China) were immunized intraperitoneally (i.p.) with 100 ⁇ L of antigen mixture containing 10 ⁇ g of Tigit-mIgG2a and a water-soluble adjuvant (Cat.: KX0210041, KangBiQuan, Beijing, China). The procedure was repeated three weeks later. Two weeks after the 2 nd immunization, mouse sera were evaluated for Tigit binding by ELISA and FACS. Ten days after serum screening, the mice with the highest anti-Tigit antibody serum titers were boosted via i.p. injection with 50 ⁇ g of Tigit-mIgG2a. Three days after boosting, the splenocytes were isolated and fused to the murine myeloma cell line, SP2/0 cells (ATCC), using the standard techniques [1977 Somat Cell Genet, 3:231].
  • the supernatants of hybridoma clones were initially screened by ELBA as described in “Methods in Molecular Biology (2007) 378:33-52” with some modifications. Briefly, Tigit-huIgG1 protein was coated in 96-well plates. The HRP-linked anti-mouse IgG antibody (Cat.: 7076S, Cell Signaling Technology, USA) and substrate (Cat.: 00-4201-56, eBioscience, USA) were used to develop color absorbance signal at the wavelength of 450 nm, which was measured by using a plate reader (SpectraMax Paradigm, Molecular Devices, USA).
  • ELISA-positive clones were further verified by FACS using either NK92MI/huTigit or NK92mi/mkTigit cells described above.
  • Tigit-expressing cells (10 5 cells/well) were incubated with ELISA-positive hybridoma supernatants, followed by binding with Alexa Fluro-647 labeled goat anti-mouse IgG antibody (Cat.: A0473, Beyotime Biotechnology, China). Cell fluorescence was quantified using a flow cytometer (Guava easyCyte 8HT, Merck-Millipore, USA).
  • the conditioned media from the hybridomas that showed positive signals in both ELISA and FACS screening were subjected to functional assays to identify antibodies with good functional activity in human immune cell-based assays (see following sections).
  • the antibodies with desired. functional activities were further sub-cloned and characterized.
  • the positive hybridoma clones were sub-clotted by the limiting dilution. Three positive subclones based on ELISA and FACS screening from each plate were selected and characterized by functional assays. The top antibody subclones verified through functional assays were adapted for growth in the CDM4MAb medium (Cat.: SH30801.02, Hyclone, USA) with 3% FBS.
  • Hybridoma cells or 293G cells transiently transfected with an antibody expression plasmid (Cat. No. R79007, Invitrogen) was cultured either in CDM4MAb medium (Cat.: SH30801.02, Hyclone) or in FreestyleTM 293 Expression medium (Cat.: 12338018, Invitrogen), and incubated in a C02 incubator for 5 to 7 days at 37° C.
  • the conditioned medium was collected through centrifugation and filtrated by passing a 0.22 ⁇ m membrane before purification.
  • Murine or recombinant antibodies containing supernatants were applied and bound to a Protein A column (Cat.: 17127901, GE Life Sciences-) following the manufacturer's guide.
  • the procedure usually yielded antibodies at purity above 90%.
  • the Protein A-affinity purified antibodies were either dialyzed against PBS or further purified using a HiLoad 16/60 Superdex200 column (Cat.: 17531801, GE Life Sciences) to remove aggregates. Protein concentrations were determined by measuring absorbance at 280 nm. The final antibody preparations were stored in aliquots in ⁇ 80° C. freezer.
  • Murine hybridoma clones were harvested to prepare total cellular RNAs using Ultrapure RNA kit (Cat.: 74104, QIAGEN, Germany) based on the manufacturer's protocol.
  • the 1st strand cDNAs were synthesized using a cDNA synthesis kit from Invitrogen (Cat.: 18080-051) and PCR amplification of the nucleotide sequences coding for heavy chain variable region (Vh) and kappa chain variable region (Vk) of murine mAbs was performed using a PCR kit (Cat.: CW0686, CWBio, Beijing, China).
  • the oligo primers used for antibody cDNAs cloning of Vh and Vk were synthesized by Invitrogen (Beijing, China) based on the sequences reported previously (Brocks et al. 2001 Mol Med 7:461). PCR products were then subcloned into the pEASY-Blunt cloning vector (Cat.: CB101-02, TransGen, China) and sequenced by Genewiz (Beijing, China). The amino acid sequences of Vh and Vk regions were deduced from the DNA sequencing results.
  • the murine mAbs were analyzed by comparing sequence homology and grouped based on sequence similarity ( FIG. 2 ).
  • Complementary determinant regions were defined based on the Kabat [FWu and Kabat 1970 J. Exp. Med. 132:211-250] and IMGT [Lefranc 1999 Nucleic Acids Research 27:209-212] system by sequence annotation and by internet-based sequence analysis in IMGT.
  • the amino acid sequences of a representative top clone mu1217 (Vh and Vk) were listed in Table 1 (SEQ ID NOs: 9 and 11).
  • the CDR sequences of mu1217 were listed in Table 2 (SEQ ID NOs: 3-8).
  • Tigit antibodies with high binding activities in ELISA and FACS, as well as with potent functional activities in the cell-based assays were characterized for their binding kinetics by SPR assays using BIAcoreTM T-200 (GE Life Sciences). Briefly, anti-human IgG antibody was immobilized on an activated CM5 biosensor chip (Cat. No.: BR100530, GE Life Sciences), Human Fc-tagged Tigit was flowed over the chip surface and captured by anti-human IgG antibody.
  • CM5 biosensor chip Cat. No.: BR100530, GE Life Sciences
  • human germline IgG genes were searched for sequences that share high degrees of homology to the cDNA sequences of mu1217 variable regions by blasting the human immunoglobulin gene database in IMGT and NCBI websites.
  • the human IGVH and IGVK genes that are present in human antibody repertoires with high frequencies (Glanville 2009 PNAS 106:20216-20221) and are highly homologous to mu1217 were selected as the templates for humanization.
  • Humanization was carried out by CDR-grafting (Methods in Molecular Biology, Vol 248: Antibody Engineering, Methods and Protocols, Humana Press) and the humanization antibodies (hu1217s) were engineered as the human IgG1mf format using an in-house developed expression vector.
  • CDRs of mu1217 V K were grafted into the framework of human germline variable gene IGV k 3-15 with 1 murine framework residue (V 58 ) retained, resulting in the humanized V K sequence of Hu1217-1-1 (SEQ ID NO: 16 for amino acid sequence and SEQ ID NO: 17 for nucleotide sequence).
  • N-terminal of H-CDR2 (SEQ ID NO: 4), H-CDRT and H-CDR3 (SEQ ID NOs: 3 and 5) of mu1217 Vh were grafted into the framework of human germline variable gene IGVH3-7 with two murine framework (T 24 and I 37 of SEQ NO: 10) residues retained.
  • Hu1217-1-1 were constructed as human full-length antibody format using in-house developed expression vectors that contain constant regions of a human IgG1 variant termed as IgG1mf (SEQ ID NO: 18) and kappa chain, respectively, with easy adapting sub-cloning sites.
  • Expression and preparation of hu1217-1-1 antibody was achieved by co-transfection of the above two constructs into 293G cells and by purification using a protein A column (Cat.: 17543802, GE Life Sciences). The purified antibodies were concentrated to 0.5-5 mg/mL in PBS and stored in aliquots in ⁇ 80° C. freezer.
  • hu1217-1-1 Based on hu1217-1-1 template, we made several single-mutations converting the retained murine residues in framework region of Vk to corresponding human germline residues, which include V581 in V K and in T24A and 137V Vh.
  • the resulted hu1217-2A-1 (T24A), hu1217-2B-1 (137V), and hu1217-1-2a (V58I) all had similar binding and functional activities to hu1217-1-1.
  • All humanization mutations were made using primers containing mutations at specific positions and a site directed mutagenesis kit (Cat. No. FM111-02, TransGen, Beijing, China). The desired mutations were verified by sequencing analysis. These hu1217-derived variant antibodies were tested in binding and functional assays as described previously.
  • Hu1217 antibodies were further engineered by introducing mutations in CDRs and framework regions to improve molecular and biophysical properties for therapeutic use in human.
  • the considerations include amino acid compositions, heat stability (T m ), surface hydrophobicity and isoelectronic points (pIs) while maintaining functional activities.
  • hu1217-2-2 SEQ ID NOs:3, 5-8, 13, and 19-21
  • the results showed both hu1217-2-2 and hu1217-1-1 were very similar in binding affinity and functional activities such as inhibiting the Tigit-mediated downstream signaling.
  • NK92mi cells were engineered to over-express human Tigit.
  • Living NK92mi/Tigit cells were seeded in 96-well plate, and were incubated with a series of dilutions of anti-Tigit antibodies, Goat anti-Human IgG was used as secondary antibody to detect antibody binding to the cell surface.
  • EC 50 values for dose-dependent binding to human native Tigit were determined by fitting the dose-response data to the four-parameter logistic model with GraphPad Prism. As show in FIG. 4 and Table 6. Both humanized 1217 antibodies, hu1217-1-1 and hu1217-2-2, showed good binding affinity to native Tigit on living cells.
  • Tigit binds to PVR with a high affinity (Kd: ⁇ 1 nM), which can compete against CD266-PVR interaction [Yu et al., 2009].
  • HEK293 cells were engineered to express high levels of MIR or PVR-L2.
  • the resultant cell lines were named HEK293/PVR and HEK293/PVR-L2, respectively.
  • the binding of soluble Tigit (Tigit-mIgG2a fusion protein) to PVR or PVR-L2 was determined by flow cytometry ( FIG. 5A ).
  • the blockade of Tigit-ligand interaction was quantitatively measured by adding serially diluted anti-Tigit antibodies. As shown in FIG.
  • hu1217-2-2/IgG1 (a humanized version comprising a wild-type IgG1 Fc region and having the same VH and VL sequences as hu1217-2-2/IgG1mf) and hu1217-2-2/IgG1mf could block Tigit binding to PVR in a dose-dependent manner with IC 50 at 0.64 and 0.55 ⁇ g/mL, respectively.
  • the IC 50 of hu1217-2-2/IgG1 and hu1217-2-2/IgG1mf in blocking Tigit-PVR-L2 interaction is 0.25 and 0.18 ⁇ g/mL, respectively.
  • the functional activity of the Tigit antibodies were further assessed using naturally derived T-cells that recognized human CMV PP65 peptide (NLVPMVATV, 495-503. HLA-A2.1-restricted) [Boeckh M, Boeckh M and Geballe A P, 2011 J Clin Invest. 121:1673-80]. Briefly, PBMCs from HLA-A2.1 + healthy donors were simulated with PP65 peptide (>98% purity, synthesized by GL Biochem, Shanghai) in the complete RPMI with 10% FBS for a week. The pp65-primed PBMCs were used as effector cells.
  • target cells Prior to assay, target cells, HCT116 cells (HLA-A2.1 + , 10 4 ), were pulsed with pp65 peptide (5 ⁇ g/mL) for 30 mins and co-cultured with equal numbers of pp65-sensitized PBMCs in 96-well plates overnight in the presence or absence of anti-Tigit antibodies or a blank control (medium only).
  • pp65 peptide 5 ⁇ g/mL
  • PBMCs PBMCs in 96-well plates overnight in the presence or absence of anti-Tigit antibodies or a blank control (medium only).
  • hu1217-2-2/IgG1 promoted pp65-specific T cells to secrete IFN- ⁇ in the cell culture supernatant in a dose-dependent manner for both donors.
  • Tigit is known to be constitutively expressed on natural killer (NK) cells at relatively higher levels and the interaction between Tigit and its ligands inhibits NK cell-mediated cytotoxicity [Wang F, et al. 2015 Eur J. Immunology 45:2886-97; Stanietsky N et al., 2009 Proc Natl Acad Sci USA 106:17858-63].
  • an NK cell line NK92MI was engineered to co-express both Tigit and DNAM-1 receptors (NK92MI/Tigit-DNAM-1) as an effector cell by retroviral transduction, according to the protocols described previously [Zhang et al, 2006 Cancer Res. 66: 5927-5933], A PVR-expressing lung cancer cell line SK-MES-1/PVR was established similarly as a target.
  • Cytotoxicity of NK92MI/Tigit-DNAM-1 cells against SK-MES-1/PVR cells was determined by an LDH release assay using the CytoTox 96 Non-Radioactive Cytotoxicity Assay kit (Promega, Madison, Wis.).
  • NK92MI/Tigit-DNAM-1 cells (8 ⁇ 10 5 ) were co-cultured with SK-MES-1/PVR cells (2 ⁇ 10 4 ) in the presence of anti-Tigit Abs (0.007-30 ⁇ g/mL) for 5 hr in 96-well V-bottom plates.
  • the results showed that anti-Tigit antibodies hu1217-2-2/IgG1mf enhances NK cell killing in a dose-dependent manner (EC 50 : 0.185 ⁇ g/mL) ( FIG. 7 ).
  • Trogocytosis is a phenomenon, in which cell surface molecules are transferred from donor cells to acceptor cells [Joly E, et al. 2003 Nat. Immunol : Machlenkin A. et al. 2008 Cancer Res .; Beum P V et al. 2008 J Immunol ; Rossi E A, et al. 2013 Blood ].
  • Antibody-induced trogocytosis via Fe ⁇ receptors (Fc ⁇ Rs) leads to down-modulation of receptors on the cell surface [Carlsten M, et al. 2016 Clin Cancer Res ; Beum P V et al. 2011 J. Immunology]. Therefore, down-regulation of target receptor by trogocytosis may cause dampened signaling.
  • hu1217-2-2/IgG1 might induce trogocytosis of Tigit receptor in the presence of Fc ⁇ R + cells, resulting in lower surface expression.
  • Jurkat/Tigit cells were incubated with HEK cells expressing various Fc ⁇ Rs (including Fc ⁇ RIIAH131, Fc ⁇ RIIB, Fc ⁇ RIIAvlss) with biotin-labeled hu1217-2-2/IgG1wt (a humanized antibody comprising the same VL and VH sequences as hu1217-2-2/IgG1mf and a wild-type IgG1 Fc region) or hu1217-2-2/IgG1mf overnight.
  • Fc ⁇ Rs including Fc ⁇ RIIAH131, Fc ⁇ RIIB, Fc ⁇ RIIAvlss
  • biotin-labeled hu1217-2-2/IgG1wt a humanized antibody comprising the same VL and VH sequences as h
  • Tigit receptor was determined by with SA-APC (Biolegend). As shown in FIG. 8 , hu1217-2-2/IgG1 but not hu1217-2-2/IgG1mf caused a significant reduction of Tigit surface expression compared to the negative control human IgG-treated cells, indicating that the reduction of surface Tigit on Jurkat/Tigit cells are Fc ⁇ R-binding dependent.
  • a flow cytometry-based. ADCC assay was set up to determine whether Tigit antibodies could induce ADCC in Tigit T + cells.
  • the assay effector cell line, NK92MI/CD16V cells was generated from NK92MI cells (ATCC) by co-transducing expression plasmids containing CD16 V158 (V158 allele) and Fc ⁇ R cDNAs.
  • Human PBMCs from healthy donors were stimulated with PHA (1 ⁇ g/ml) to up-regulate Tigit expression.
  • T cells including CD4 + effector (CD3 + CD4 + Foxp3 ⁇ ), CD8 + and regulator T cells (CD4 + Foxp3 + ) all expressed significant amounts of Tigit.
  • PBMCs from 3 healthy donors were used as target cells.
  • a fluorescent dye CFSE-labeled NK92MI/CD16V cells (5 ⁇ 10 4 ) were co-cultured with equal number of target cells, for 40 hours in the presence of Tigit antibodies (hu1217-2-2/IgG1mf or hu1217-2-2/IgG1wt, 30 ⁇ g/mL) or control antibodies (the positive control anti-CD3 antibody OKT3 (5 ⁇ g/ml, Biolegend) or a negative control human IgG, 30 ⁇ g/mL).
  • hu1217-2-2/IgG1wt could lead to moderate reduction of Tregs via ADCC.
  • no significant ADCC effects were observed in total T cells and CD8 + T cells ( FIG. 9 ).
  • Whether hu1217-2-2/IgG1mf and hu1217-2-2/IgG1wt would trigger CDC was determined by using pre-activated human PBMCs and fresh autologous sera from healthy donors. Cell lysis by CDC was determined by a Celltiter glo assay kit (Promega, Beijing, China). In brief, PBMCs were pre-activated with PHA (10 ⁇ g/mL) for 3 days, and then were incubated in RPMI1.640 plus autologous serum (15%) and anti-Tigit or control antibodies (0.01-100 ⁇ g/mL) for overnight at 37° C.
  • the cell death due to CDC was assayed by the decrease of ATP released from viable cells after cell lysis at the end of reaction.
  • Anti-MHC-1 A, B, C was used as a positive control.
  • the experimental results demonstrated that both hu1217-2-2/IgG1mf and hu1217-2-2/IgG1wt had no detactable CDC with PBMCs isolated from two different donors.
  • the positive control antibody, anti-MHC-1 induces significant CDC activity ( FIG. 10 ).
  • hu1217-2-2/IgG1 showed higher binding affinity (KD) and stronger binding signal (Rmax) against human TIGIT at PH 6.0 (an acidic pH which is similar to the pH of tumor microenvironment) as compared to the data obtained at pH 7.4 (physiologic pH).

Abstract

Provided are antibodies that specifically bind to TIGIT (T cell immunoreceptor with Ig and ITIM domains, WUCAM or Vstm3) and inhibit Tigit-mediated cellular signaling and activities in immune cells. The anti-TIGIT antibodies can be used to treat or diagnose cancer, infectious diseases or other pathological disorders that may be modulated by Tigit-mediated functions.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. patent application Ser. No. 17/534,916, filed Nov. 24, 2021, which is a divisional of U.S. patent application Ser. No. 16/958,262, filed Jun. 26, 2020 (now issued U.S. Pat. No. 11,214,616), which is a U.S. National Stage Application under 35 U.S.C. § 371 of International Application No. PCT/CN2018/125375, filed Dec. 29, 2018, which claims the benefit of priority to International patent application number PCT/CN2017/120392, filed on Dec. 30, 2017; the disclosures of each of which are incorporated herein by reference in their entireties for all purposes.
  • DESCRIPTION OF THE TEXT FILE SUBMITTED ELECTRONICALLY
  • The contents of the text file submitted electronically herewith are incorporated herein by reference in their entirety: A computer readable format copy of the Sequence Listing (filename: BEIG_033_03US_SeqList_ST25.txt, date recorded Feb. 18, 2022, file size 19.3 kilobytes).
  • FIELD OF THE INVENTION
  • The present application relates to antibodies that specifically bind to TIGIT (T cell immunoreceptor with Ig and ITIM domains) and uses of the same.
  • BACKGROUND OF THE INVENTION
  • Tigit (T cell immunoglobulin and ITIM domain) is a type I transmembrane protein, a member of the CD28 family of proteins that plays an important role in inhibiting T- and NK cell-mediated functional activities in anti-tumor immunity [Boles K S, et al., 2009 Eur Immunol, 39:695-703; Stanietsky N, et al., 2009 PNAS 106:17858-63; Yu X et al. 2009 Nat. Immunol, 10:48-57].
  • The genes and cDNAs coding for TIGIT were cloned and characterized in mouse and human. Full length human TIGIT has a sequence of 244 amino acids (SEQ ID NO: 1) in length, in which the first 21 amino acids consist a signal peptide. The amino acid sequence of the mature human TIGIT contains 223 amino acid (aa) residues (NCBI accession number: NM_173799). The extracellular domain (ECD) of mature human TIGIT consists of 120 amino acid residues (SEQ ID NO: 2, corresponding to amino acids 22-141 of SEQ ID NO: 1) with a V-type Ig-like domain (corresponding to amino acids 39-127 of SEQ ID NO: 1), followed by a 21 aa transmembrane sequence, and an 82 aa cytoplasmic domain with an immunoreceptor tyrosine-based inhibitory motif (ITIM) [Yu X et al. 2009 Nat. Immunol, 10:48-57: Stengel K F, et al. 2012 PNAS 109:5399-04]. Within the ECD, human TIGIT shares only 59% and 87% aa sequence identity with mouse and cynomolgus monkey, respectively.
  • TIGIT is expressed on T cells (including activated T cells, memory T cells, regulatory T (Treg) cells, and follicular T helper (Tfh) cells), and NK cells [Boles K S, et al., 2009 Eur J Immunol 39:695-703; Joller N, et al., 2014 Immunity 40:569-81; Levin S D, et al. 2011 Eur Immunol, 41:902-15; Stanietsky N, et al. 2009 PNAS 106:17858-63; Yu X et al. 2009 Nat. Immunol, 10:48-57].
  • So far, two Tigit ligands, CD155 (also known as poliovirus receptor or PVR) and CD112 (also known as poliovirus receptor-related 2, PVRL2, nectin-2), have been identified. These ligands are primarily expressed on APCs (such as dendritic cells and macrophages) and tumor cells [Casado J G, et al., 2009 Cancer Immunol Immunother 58:1517-26; Levin S D, et al., 2011 Eur J Immunol, 41:902-15; Mendelsohn C L et al., 1989 56:855-65; Stanietsky N, et al., 2009 PNAS 106:17858-63; Yu X et al. 2009 Nat. Immunol, 10:48-57]. As an immune “checkpoint” molecule, Tigit initiates inhibitory signaling in immune cells when engaged by its ligands, CD155 and CD112. The binding affinity of Tigit to CD155 (Kd: ˜1 nM) is much higher than to CD112 and whether the TIGIT: CD112 interaction is functionally relevant in mediating inhibitory signals yet remain to be determined. A co-stimulatory receptor, CD226 (DNAM-1), binds to the same ligands with lower affinity (Kd: ˜100 nM), but delivers a positive signal [Bottino C, et al., 2003 JExp Med 198:557-67]. In addition, CD96 (Tactile), a “Tigit-like” receptor, also plays a similarly inhibitory role in the same pathway [Chan C J, et al., 2014 Nat. Immunol 15:431-8].
  • Tigit can inhibit immune responses through different mechanisms. First, interaction between TIGIT and PVR on dendritic cells (DCs) could deliver a “reverse signaling” in DCs, leading to up-regulation of IL-10 and decrease of IL-12 secretion, thereby inhibiting T-cell activation [Yu X et al. Nat Immunol. 2009 10:48-57]. Second, TIGIT binds to CD155 with higher affinity, thereby competing off DNAM-1-CD155 interaction. Third, direct ligation of TIGIT on T cells could down-regulate TCR-mediated activation and subsequent proliferation and engagement of TIGIT on NK cells block NK cell cytotoxicity [Joller N, et al. 2011 186: 1338-42; Stanietsky N, et al., 2009 PNAS 106:17858-63]. Fourth, Tigit expression on Tregs has been associated with a highly activated and suppressive phenotype in tumor tissue and TIGIT signaling in Tregs may favor Treg stability [Joller N, et al. Immunity 2014 40:569-81; Kurtulus S, et al. J Clin Invest. 2015 125: 4053-4062].
  • TIGIT has an immunoglobulin tail tyrosine (ITT)-like motif followed by an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic tail [Yu X et al. Nat Immunol. 2009 10:48-57; Engels N, et al. Curr Opin Immunol 2011 23: 324-329]. These motifs could mediate recruitment of the phosphatase SHIP-1 and β-arrestin 2 [Li M, et al. J Biol Chem. 2014 289:17647-17657; Liu S, et al. Cell death and differentiation 2013 20: 456-464], thus providing a mechanism by which TIGIT can intrinsically deliver inhibitory signals to dampen activating signals.
  • Up-regulation of Tigit expression in tumor-infiltrating lymphocytes (TILs) and peripheral blood mononuclear cells (PBMCs) has been reported in many types of cancers such as lung [Tassi, et al., Cancer Res. 2017 77: 851-861], esophageal [Xie J, et al., Oncotarget 2016 7:63669-63678], breast [Gil Del Alcazar C R, et al. 2017 Cancer Discov.], acute myeloid leukemia (AML) [Kong Y et al., Clin Cancer Res. 2016 22:3057-66] and melanoma [Chauvin J M, et al., J Clin Invest. 2015 125:2046-2058]. The increased expression of Tigit in AML is associated with poor prognosis of patient survival outcome [Kong Y et al. Clin Cancer Res, 2016 22:3057-66]. Not only does up-regulation of Tigit signaling play important roles in immune tolerance to cancer, but also to chronic viral infection. During HIV infection, expression of Tigit on T cells was significantly higher and positively correlated with viral loads and disease progression [Chew G M, et al., 2016 PLoS Pathog. 12:e1005349]. In addition, blockade of Tigit receptor alone or in combination with other blockade could rescue functionally “exhausted” T cells both in vitro and in vivo [Chauvin J M, et al., J Clin Invest. 2015 125: 2046-2058; Chew G M, et al., 2016 PLoS Pathog. 12:e1005349: Johnston R J, et al., Cancer Cell 2014 26:923-937]. In the cases of cancer and viral infections, activation of Tigit signaling promotes immune cell dysfunction, leading to the cancer outgrowth or extended viral infection. Inhibition of Tigit-mediated inhibitory signaling by therapeutic agents may restore the functional activities of immune cells including T cells, NK cells and dendritic cells (DCs), therefore enhancing immunity against cancer or chronic viral infection.
  • Therefore, modulation of Tigit signaling by antagonistic molecules may rescue immune cells from tolerance, inducing efficient immune responses to eradicate tumors or chronic viral infections.
  • SUMMARY OF THE INVENTION
  • The present invention is at least in part based on the discovery of a set of monoclonal antibodies (mAbs) which inhibit Tigit-mediated cellular signaling in immune cells, re-activate the immune cells and enhance immunity by specifically binding to Tigit. Accordingly, in the first aspect, the present application relates to an anti-Tigit antibody and antigen-binding fragment thereof which is capable of binding to human Tigit (SEQ ID NO: 1). The present invention also relates to the humanized version of the anti-Tigit mAbs of the first aspect.
  • In particular embodiments, the antibody of the present application comprises a heavy chain variable region (VH) comprising one, two or three CDRs having an amino acid sequence selected from SEQ ID NOs: 3, 4, 5 or 13, or variants thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions in the amino acid sequences of SEQ ID NOs 3, 4, 5 or 13; and/or a light chain variable region (VL) comprising one, two or three CDRs having an amino acid sequence selected from SEQ ID NOs: 6, 7 or 8, or variants thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions in the amino acid sequences of SEQ ID NOs: 6, 7, or 8.
  • In a more specific embodiment, the antibody of the present application comprises a heavy chain variable region (VH) comprising a VH-CDRT having an amino acid sequence of SEQ ID NO: 3 or a variant thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions, a VH-CDR2 having an amino acid sequence of SEQ ID NO: 4 or SEQ ID NO: 13 or a variant thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions, and a VH-CDR3 having an amino acid sequence of SEQ ID NO: 5 or a variant thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions; and/or a light chain variable region (VL) comprising a VL-CDR1 having an amino acid sequence of SEQ ID NO: 6 or a variant thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions, a VL-CDR2 having an amino acid sequence of SEQ ID NO: 7 or a variant thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions, and a VL-CDR3 having an amino acid sequence of SEQ ID NO: 8 or a variant thereof comprising one or more conservative substitutions, e.g. one or two conservative substitutions.
  • The antibody or the antigen-binding fragment thereof of the present application is capable of binding to human Tigit and comprises a heavy chain variable region having an amino acid sequence selected from SEQ ID NO: 9, 14, 19, or a sequence having at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% sequence identity with SEQ ID NO: 9, 14, 19. In one embodiment, the difference in sequence lies in the framework region. In one embodiment, the antibody or the antigen-binding fragment thereof comprises a heavy chain variable region encoded by an nucleotide sequence selected from SEQ ID NO: 10, 15 or 20, or a variant thereof.
  • The antibody or the antigen-binding fragment thereof of the present application is capable of binding to human Tigit and comprises a heavy chain variable region having an amino acid sequence selected from SEQ ID NO: 11, 16, 21, or 24, or a sequence having at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% sequence identity with SEQ ID NO: 11, 16, 21 or 24. In one embodiment, the difference in sequence lies in the framework region. In one embodiment, the antibody or the antigen-binding fragment thereof comprising a heavy chain variable region encoded by an nucleotide sequence selected from SEQ ID NO: 12, 17 or 22, or a variant thereof.
  • In one embodiment, the antibody or antigen-binding fragment thereof is capable of binding to human Tigit with a Kd value of about 1×10−9 M to about 1×10−12 M. For example, the antibody or the antigen-binding fragment thereof is capable of binding to human Tigit with a Kd value less than about 1×10−9 M, less than about 1×10−10 M, less than about 1×10−11 M, or less than about 1×10−12 M.
  • In one embodiment, the antibody or the antigen-binding fragment thereof comprises a heavy chain constant region of the subclass of IgG1, IgG2, IgG3, or IgG4 or a variant thereof, and a light chain constant region of the type of kappa or lambda or a variant thereof. In a more specific embodiment, the Fe region of the antibody is human IgG1 Fe or a variant thereof, e.g. a Fe region of SEQ ID NO: 18.
  • In one embodiment, the antibody or antigen-binding fragment thereof promotes the production of IFN-γ by antigen-specific T cells. In a more specific embodiment, the antibody or antigen-binding fragment thereof promotes the production of IFN-γ by antigen-specific T cells in a dose-dependent manner.
  • In a more specific embodiment, the antibody of the present application reduces the surface expression of Tigit receptor via FcγR-mediated trogocytosis, particularly FcγRIIB-mediated trogocytosis.
  • In one embodiment, the antibody of the present application shows a pH-dependent antigen binding such that the antibody exhibits a stronger binding to human TIGIT at a mild acidic pH in tumor microenvironment (e.g. pH 6.0) as compared to the binding to human TIGIT at a physiologic pH (e.g. pH 7.4). In a more specific embodiment, the antibody of the present application has (1) a KD ratio at pH 7.4/pH 6.0 of greater than 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, or more, and/or (2) a Rmax (RU) value at pH 6.0 which is at least 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold higher than the Rmax at pH 7.4, as measured by surface plasmon resonance (Biacore) or similar technology.
  • The anti-Tigit mAbs disclosed herein have potential therapeutic uses in treating cancer, controlling viral infections and other human diseases that are mechanistically involved in immune tolerance or “exhaustion”. Accordingly, in further embodiments, the anti-Tigit antibody of the present application is for use in treatment of cancer. In another specific embodiment, the anti-Tigit antibody of the present application is for use in treating an infection, treating an infectious disease and/or controlling viral infections. In another specific embodiment, the anti-Tigit antibody of the present application is for use in the treatment of other human diseases related to or caused by immune tolerance, or the treatment of a disease that can be improved by increasing immune cell activation.
  • In a further aspect, the present application relates to a composition comprising the anti-Tigit antibody or antigen-binding fragment thereof and a therapeutically acceptable excipient.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 Schematic diagram of Tigit-mIgG2a (top) and Tigit-huIgG1 (bottom). Tigit ECD: tigit extracellular domain, N: N-terminus. C: C-terminus.
  • FIG. 2A, 2B Phylogenetic trees of anti-Tigit antibody Vh (2A) and Vk (2B) regions. The Vh and Vk sequences of candidate anti-Tigit antibodies were aligned using DNASTAR's Megalign software. Sequence homology was displayed in phylogenetic trees.
  • FIG. 3 Affinity determination of purified murine anti-Tigit antibodies by surface plasmon resonance (SPR).
  • FIG. 4 Determination of Tigit binding by flow cytometry.
  • FIG. 5A A schematic diagram showing the inhibition of Tigit-ligand interactions by anti-Tigit mAbs. FIG. 5B The binding of soluble Tigit (Tigit-huIgG1 fusion protein) to Tigit ligand-expressing HEK293 cells (HEK293/PVR or HEK293/PVR-L2) was determined by flow cytometry. The blockade of Tigit-ligand interaction was quantitatively measured by adding serially diluted anti-Tigit antibodies. Results were shown in mean±SD of duplicates.
  • FIG. 6 Activation of CMV-specific human T cells by anti-Tigit mAbs. Human CMV peptide (NLVPMVATV, 495-503)-sensitized HLA-A2.1+ PBMCs (4×104) were stimulated with the CMV peptide-pulsed target cells HCT116 cells (104) overnight in the presence of anti-Tigit antibodies. IFN-γ in the culture supernatant was determined by ELISA. All conditions were performed in triplicates. Results were shown as mean±SD.
  • FIG. 7 Anti-Tigit mAbs promote NK cell-mediated cytotoxicity. (A) Tigit and DNAM-1 expression on engineered NK92MI/Tigit-DNAM-1 stable cell line. (B) Killing of NK92MI/Tigit-DNAM-1 cells against SK-MES-1/PVR cells in the presence of hu1217-2-2/IgG1mf (0.007-30 μg/ml) was determined by an LDH (lactate dehydrogenase) release assay as described in Example 8. Results were shown in mean±SD of triplicates.
  • FIG. 8 Anti-Tigit mAb hu1217-2-2/IgG1wt reduces the surface expression of Tigit receptor via FcγR-mediated trogocytosis. Jurkat/Tigit cells were incubated with Fc γ R-expressing HEK293 cells in the presence of biotin-labeled anti-Tigit mAbs in complete media overnight. In some cases, 10% human AB serum was added to determine the effects of bulk human IgG on trogocytosis. Surface expression of Tigit receptor was determined by staining with SA-APC (Biolegend). MFI was determined by flow cytometry. All data points were in duplicates. Results were shown in mean±SD.
  • FIG. 9 ADCC effects of anti-Tigit mAbs on human peripheral blood mononuclear cells (PBMCs). (A) Tigit expression on PHA-stimulated PBMCs from healthy donors was determined by flow cytometry. CD4+ (CD4+Foxp3), CD8+ T effectors and regulatory T cells (Tregs, CD4+Foxp3+) all expressed significant levels of Tigit (18˜41%). Data shown are representative results from 3 healthy donors. (B) ADCC assay was performed using a CD16+ human NK cell line NK92MI/CD16V as effector cells and PHA-stimulated PBMCs as target cells in the presence of Tigit mAbs (30 μg/mL) or control antibodies (OKT3 at 5 μg/ml as a positive control, and huIgG at 30 μg/mL as a negative control) for 42 hrs. Percentages of CD3+, CD8 T cells and Tregs were determined by flow cytometry.
  • FIG. 10. CDC effects of anti-Tigit mAbs on human PBMCs. CDC assay was performed using PHA-stimulated PBMCs as target cells and autologous sera as the source of complements. After 3 days of co-culture of pre-activated PBMCs with antiTigit mAbs (0.01-100 μg/ml) in the final concentration of 15% autologous sera, percentage of CDC (y-axis) was measured by cell-titer glow assay, and calculated as described in Example 11. Data from donors A and B are shown. HuIgG were used as a negative control, whereas anti-MHC-A, B, C was used as a positive control.
  • DETAILED DESCRIPTION OF THE INVENTION Definitions
  • Conservative amino acid substitutions of amino acids are commonly known in the art and exemplarily shown in the table below. Generally, a conservative amino acid substitution means that an amino acid residue is replaced by another amino acid residue having a similar side chain.
  • Original amino One-letter and Conservative
    acid residue three-letter codes substitution(s)
    Alanine A or Ala Gly; Ser
    Arginine R or Arg Lys; His
    Asparagine N or Asn Gln; His
    Aspartic acid D or Asp Gln; Asn
    Cysteine C or Cys Ser; Ala
    Glutamine Q or Gln Asn
    Glutamic acid E or Glu Asp; Gln
    Glycine G or Gly Ala
    Histidine H or His Asn; Gln
    Isoleucine I or Ile Leu; Val
    Leucine L or Leu Ile; val
    Lysine K or Lys Arg; His
    Methionine M or Met Leu; Ile; Tyr
    Phenylalanine F or Phe Tyr; Met; Leu
    Proline P or Pro Ala
    Serine S or Ser Thr
    Threonine T or Thr Ser
    Tryptophan W or Trp Tyr; Phe
    Tyrosine Y or Tyr Trp; Phe
    Valine V or Val Ile; Leu
  • Unless specifically defined elsewhere in this document, all other technical and scientific terms used herein have the meaning commonly understood by one of ordinary skill in the art to which this invention belongs.
  • As used herein, including the appended claims, the singular forms of words such as “a”, “an”, and “the”, include their corresponding plural references unless the context clearly dictates otherwise.
  • The term “or” is used to mean, and is used interchangeably with, the term “and/or” unless the context clearly dictates otherwise.
  • Throughout this specification and 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 amino acid sequence, DNA, sequence, step or group thereof, but not the exclusion of any other amino acid sequence, DNA sequence, step. When used herein the term “comprising” can be substituted with the term “containing”, “including” or sometimes “having”.
  • The term “Tigit” includes various mammalian isoforms, e.g., human Tigit, orthologs of human Tigit, and analogs comprising at least one epitope within Tigit. The amino acid sequence of Tigit, e.g., human Tigit, and the nucleotide sequence encoding the same, is known in the art.
  • The terms “administration”, “administering”, “treating” and “treatment” as used herein, when applied to an animal, human, experimental subject, cell, tissue, organ, or biological fluid, mean contact of an exogenous pharmaceutical, therapeutic, diagnostic agent, or composition to the animal, human, subject, cell, tissue, organ, or biological fluid. Treatment of a cell encompasses contact of a reagent to the cell, as well as contact of a reagent to a fluid, where the fluid is in contact with the cell. The term “administration” or “treatment” also includes in vitro and ex vivo treatments, e.g., of a cell, by a reagent, diagnostic, binding compound, or by another cell. The term “subject” herein refers to any organism, preferably an animal, more preferably a mammal (e.g., rat, mouse, dog, cat, rabbit) and most preferably a human.
  • Antibody or Antibody Molecule
  • Disclosed herein are antibody molecules that bind to Tigit with high affinity and specificity.
  • In some embodiments, the anti-Tigit antibody binds to human Tigit and includes at least one, two, three, four, five or six complementarity determining regions (CDR's) comprising an amino acid sequence SEQ ID NOs 3, 4, 5, 13 or SEQ ID NOs: 6, 7, 8. In particular embodiments, the antibody of the present application comprises a heavy chain variable region (VH) comprising one, two or three CDRs having an amino acid sequence selected from SEQ, ID NOs: 3, 4, 5 or 13, or a variant thereof comprising one or more conservative substitutions; and/or a light chain variable region (VL) comprising one, two or three CDRs having an amino acid sequence selected from SEQ ID NOs: 6, 7, or 8, or a variant thereof comprising one or more conservative substitutions.
  • In some embodiments, the anti-Tigit antibody is an isolated antibody, a humanized antibody, a chimeric antibody or a recombinant antibody.
  • In some embodiments, the anti-Tigit antibody comprises at least one antigen-binding site, or at least a variable region. In some embodiments, the anti-Tigit antibody comprises an antigen-binding fragment derived from an antibody described herein.
  • The term “antibody” herein is used in the broadest sense and specifically covers antibodies (including full length monoclonal antibodies) and antibody fragments so long as they recognize antigen, e.g., Tigit. An antibody is usually monospecific, but may also be described as idiospecific, heterospecific, or polyspecific. Antibody molecules bind by means of specific binding sites to specific antigenic determinants or epitopes on antigens.
  • The term “monoclonal antibody” or “mAb” or “Mab” herein means a population of substantially homogeneous antibodies, i.e., the antibody molecules comprised in the population are identical in amino acid sequence except for possible naturally occurring mutations that may be present in minor amounts. In contrast, conventional (polyclonal) antibody preparations typically include a multitude of different antibodies having different amino acid sequences in their variable domains, particularly their complementarity determining regions (CDRs), which are often specific for different epitopes. The modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method, Monoclonal antibodies (mAbs) may be obtained by methods known to those skilled in the art. See, for example Kohler G et al., Nature 1975 256:495-497; U.S. Pat. No. 4,376,110; Ausubel F M et al., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY 1992; Harlow E et al., ANTIBODIES: A LABORATORY MANUAL, Cold spring Harbor Laboratory 1988; and Colligan J E et at., CURRENT PROTOCOLS IN IMMUNOLOGY 1993. The mAbs disclosed herein may be of any immunoglobulin class including IgG, IgM, IgD, IgE, IgA, and any subclass thereof. A hybridoma producing a mAb may be cultivated in vitro or in vivo. High titers of mAbs can be obtained by in vivo production where cells from the individual hybridomas are injected intraperitoneally into mice, such as pristine-primed Balb/c mice to produce ascites fluid containing high concentrations of the desired mAbs, MAbs of isotype IgM or IgG may be purified from such ascites fluids, or from culture supernatants, using column chromatography methods well known to those of skill in the art.
  • In general, the basic antibody structural unit comprises a tetramer. Each tetramer includes two identical pairs of polypeptide chains, each pair having one “light chain” (about 25 kDa) and one “heavy chain” (about 50-70 kDa). The amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The carboxy-terminal portion of the heavy chain may define a constant region primarily responsible for effector function. Typically, human light chains are classified as kappa and lambda light chains. Furthermore, human heavy chains are typically classified as α, δ, ε, γ, or μ, and define the antibody's isotypes as IgA, IgD, IgE, IgG, and IgM, respectively. Within light and heavy chains, the variable and constant regions are joined by a “J” region of about 12 or more amino acids, with the heavy chain also including a “D” region of about 10 more amino acids.
  • The variable regions of each light/heavy chain (VL/VH) pair form the antibody binding site. Thus, in general, an intact antibody has two binding sites. Except in bifunctional or bispecific antibodies, the two binding sites are, in general, the same.
  • Typically, the variable domains of both the heavy and light chains comprise three hypervariable regions, also called “complementarity determining regions (CDRs)”, which are located between relatively conserved framework regions (FR). The CDRs are usually aligned by the framework regions, enabling binding to a specific epitope. In general, from N-terminal to C-terminal, both light and heavy chain variable domains sequentially comprise FR-1 (or FR1), CDR-1 (or CDR1), FR-2 (FR2), CDR-2 (CDR2), FR-3 (or FR3) CDR-3 (CDR3) and FR-4 (or FR4). The assignment of amino acids to each domain is, generally, in accordance with the definitions of Sequences of Proteins of Immunological Interest, Kabat, et al., National Institutes of Health, Bethesda, Md.; 5<m> ed.; NIHPubl. No. 91-3242 (1991); Kabat (1978) Adv. Prot. Chem. 32: 1-75; Kabat, et al., (1977) J. Biol. Chem. 252:6609-6616; Chothia, et al, (1987) J Mol. Biol. 196:901-917 or Chothia, et al, (1989) Nature 342:878-883.
  • The term “hypervariable region” means the amino acid residues of an antibody that are responsible for antigen-binding. The hypervariable region comprises amino acid residues from a “CDR” (i.e., CDR1, VL-CDR2 and VL-CDR3 in the light chain variable domain and VH-CDR1, VH-CDR2 and VH-CDR3 in the heavy chain variable domain). See, Kabat et al. (1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (defining the CDR regions of an antibody by sequence); see also Chothia and Lesk (1987) J. Mol. Biol. 196: 901-917 (defining the CDR regions of an antibody by structure). The term “framework” or “FR” residues mean those variable domain residues other than the hypervariable region residues defined herein as CDR residues.
  • Unless otherwise indicated, “antibody fragment” or “antigen-binding fragment” means antigen binding fragments of antibodies, i.e. antibody fragments that retain the ability to bind specifically to the antigen bound by the full-length antibody, e.g, fragments that retain one or more CDR regions. Examples of antigen binding fragments include, but not limited to, Fab, Fab′, F(ab′)2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules, e.g., single chain Fv (ScFv); nanobodies and multispecific antibodies formed from antibody fragments.
  • An antibody that binds to a specified target protein with specificity is also described as specifically binding to a specified target protein. This means the antibody exhibits preferential binding to that target as compared to other proteins, but this specificity does not require absolute binding specificity. An antibody is considered “specific” for its intended target if its binding is determinative of the presence of the target protein in a sample, e.g. without producing undesired results such as false positives. Antibodies or binding fragments thereof, useful in the present invention will bind to the target protein with an affinity that is at least two fold greater, preferably at least 10-times greater, more preferably at least 20-times greater, and most preferably at least 100-times greater than the affinity with non-target proteins. An antibody herein is said to bind specifically to a polypeptide comprising a given amino acid sequence, e.g. the amino acid sequence of a mature human Tigit molecule, if it binds to polypeptides comprising that sequence but does not bind to proteins lacking that sequence.
  • The expressions “pH-dependent binding”, “pH-dependent target binding” and “pH-dependent antigen binding” are interchangeable in the present disclosure, indicating that the antibody of the present application binds to its target/antigen, namely human TIGIT, in a pH-dependent manner. Specifically, the antibody of the present application shows a higher binding affinity and/or binding signal to its antigen at a mild acidic pH, e.g. pH 6.0, which is usually found in tumor microenvironment, as compared to the binding affinity and/or binding signal at physiologic pH, e.g. pH 7.4. The methods for determining the binding affinity and/or the intensity of binding signal of the antibody of the present application are well known in the art and include but not limited to surface plasmon resonance (Biacore) or similar technology. More specifically, the antibody of the present application has a KD ratio at pH 7.4/pH 6.0 of greater than 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, or more, as measured by surface plasmon resonance (Biacore) or similar technology. Alternatively or additionally, the antibody of the present application has a Rmax (RU) value at pH 6.0 which is at least 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold higher than the Rmax at pH 7.4 as measured by surface plasmon resonance (Biacore) or similar technology. The binding affinity of the antibody can be measured at 25° C. or 37° C. Tumor microenvironment has been found to show a relatively more acidic pH than physiological condition or normal tissues (Zhang et al. Focus on molecular Imaging 2010; Tannock and Rotin et al, Cancer Res 1989). Therefore, the antibody of the present application having above-mentioned pH-dependent binding is advantageous as an anti-TIGIT therapeutic agent for targeting TIGIT-positive lymphocytes in the tumor microenvironment with selectivity and having lower toxicity associated with periphery activation of lymphocytes.
  • The term “human antibody” herein means an antibody that comprises human immunoglobulin protein sequences only. A human antibody may contain murine carbohydrate chains if produced in a mouse, in a mouse cell, or in a hybridoma derived from a mouse cell. Similarly, “mouse antibody” or “rat antibody” means an antibody that comprises only mouse or rat immunoglobulin protein sequences, respectively.
  • The term “humanized antibody” means forms of antibodies that contain sequences from non-human (e.g., murine) antibodies as well as human antibodies. Such antibodies contain minimal sequence derived from non-human immunoglobulin. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. The prefix “hum”, “hu, “Hu” or “h” is added to antibody clone designations when necessary to distinguish humanized antibodies from parental rodent antibodies. The humanized forms of rodent antibodies will generally comprise the same CDR sequences of the parental rodent antibodies, although certain amino acid substitutions may be included to increase affinity, increase stability of the humanized antibody, or for other reasons.
  • The antibody of the present application has potential therapeutic uses in treating cancer. The term “cancer” or “tumor” herein means or describes the physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include but are not limited to, lung cancer (including small-cell lung cancer, or non-small cell lung cancer), adrenal cancer, liver cancer, stomach cancer, cervical cancer, melanoma, renal cancer, breast cancer, colorectal cancer, leukemia, bladder cancer, bone cancer, brain cancer, an endometrial cancer, head and neck cancer, lymphoma, ovarian cancer, skin cancer, thyroid tumor, or metastatic lesion of the cancer.
  • Further, the antibody of the present application has potential therapeutic uses in controlling viral infections and other human diseases that are mechanistically involved in immune tolerance or “exhaustion”. in the context of the present application, the term “exhaustion” refers to a process which leads to a depleted ability of immune cells to respond to the infecting virus during a prolonged period of chronic viral infection.
  • Pharmaceutical Compositions and Kits
  • In some aspects, this disclosure provides compositions, e.g., pharmaceutically acceptable compositions, which include an anti-Tigit-3 antibody described herein, formulated together with at least one pharmaceutically acceptable excipient. As used herein, the term “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, isotonic and absorption delaying agents, and the like that are physiologically compatible. The excipient can be suitable for intravenous, intramuscular, subcutaneous, parenteral, rectal, spinal or epidermal administration (e.g. by injection or infusion).
  • The compositions herein may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusion solutions), dispersions or suspensions, liposomes, and suppositories. A suitable form depends on the intended mode of administration and therapeutic application. Typical suitable compositions are in the form of injectable or infusion solutions. One suitable mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular). in some embodiments, the antibody is administered by intravenous infusion or injection. In certain embodiments, the antibody is administered by intramuscular or subcutaneous injection.
  • The term “therapeutically effective amount” as herein used, refers to the amount of an antibody that, when administered to a subject for treating a disease or a disorder, or at least one of the clinical symptoms of a disease or disorder, is sufficient to effect such treatment for the disease, disorder, or symptom. The “therapeutically effective amount” can vary with the antibody, the disease, disorder, and/or symptoms of the disease or disorder, severity of the disease, disorder, and/or symptoms of the disease or disorder, the age of the subject. to be treated, and/or the weight of the subject to be treated. An appropriate amount in any given instance can be apparent to those skilled in the art or can be determined by routine experiments. In the case of combination therapy, the “therapeutically effective amount” refers to the total amount of the active agents comprised in the combination for the effective treatment of a disease, a disorder or a condition.
  • The “subject” as used herein is a mammal, e.g., a rodent or a primate, preferably a higher primate, e.g., a human (e.g., a patient having, or at risk of having, a disorder described herein).
  • EXAMPLE Example 1 Generation of Anti-Tigit Monoclonal Antibody
  • Anti-Tigit monoclonal antibodies (mAbs) were generated based on conventional hybridoma fusion technology [de StGroth and Sheidegger 1980 J Immunol Methods 35:1; Mechetner; 2007 Methods Mol Biol 378:1] with minor modifications. The mAbs with high binding activity in enzyme-linked immunosorbent assay (ELISA) and fluorescence-activated cell sorting (FACS) assay were selected for further characterization.
  • Tigit Recombinant Protein for Immunization and Binding Assays
  • The cDNA coding for the full-length human Tigit (SEQ ID NO: 1) was synthesized by and purchased from Sino Biological (Beijing, China) based on its GenBank sequence (Accession No: NM_173799). The coding region of extracellular domain (ECD) of the full-length human Tigit corresponding to the amino acid (AA) 1-141 of SEQ ID NO: 1 was PCR-amplified, and cloned into pcDNA3.1-based expression vector (Invitrogen, Carlsbad, Calif., USA) with C-terminus fused either to the Fc domain of mouse IgG2a or to the Fc domain of human IgG1 heavy chain, which resulted in two recombinant fusion protein expression plasmids, Tigit-mIgG2a and Tigit-huIgG1, respectively. The schematic presentation of Tigit fusion proteins were shown in FIG. 1. For the recombinant fusion protein production, Tigit-mIgG2a and Tigit-huIgG1 plasmids were transiently transfected into 293G cells (developed in-house) and cultured for 7 days in a C02 incubator equipped with rotating shaker. The supernatant containing the recombinant protein was collected and cleared by centrifugation. Tigit-mIgG2a and Tigit-huIgG1 were purified using a Protein A column (Cat: 17127901, GE Life Sciences). Both Tigit-mIgG2a and Tigit-hulgG1 proteins were dialyzed against phosphate buffered saline (DPBS) and stored in −80° C. freezer in small aliquots.
  • Stable Expression Cell Lines
  • To establish stable cell lines that express full-length human Tigit (huTigit) or monkey Tigit (mkTigit, accession #: XM_005548101.2), Tigit genes (synthesized by Genescript, Nanjing, China) were cloned into a retroviral vector pFB-Neo (Cat.: 217561, Agilent, USA). Dual-tropic retroviral vectors were generated according to a previous protocol [Zhang T, et al. 2005, Blood]. Vectors containing huTigit and mkTigit were transduced into Jurkat and NK92MI cells (ATCC, Manassas, Va., USA), respectively, to generate the cell lines, Jurka/huTigit and NK92MI/mkTigit. The high expression cell lines were selected by cultivation in medium with G418 and PACS binding assay.
  • Immunization, Hybridoma Fusion and Cloning
  • Eight to twelve week-old Balb/c mice (from HFK BIOSCIENCE CO., LTD, Beijing, China) were immunized intraperitoneally (i.p.) with 100 μL of antigen mixture containing 10 μg of Tigit-mIgG2a and a water-soluble adjuvant (Cat.: KX0210041, KangBiQuan, Beijing, China). The procedure was repeated three weeks later. Two weeks after the 2nd immunization, mouse sera were evaluated for Tigit binding by ELISA and FACS. Ten days after serum screening, the mice with the highest anti-Tigit antibody serum titers were boosted via i.p. injection with 50 μg of Tigit-mIgG2a. Three days after boosting, the splenocytes were isolated and fused to the murine myeloma cell line, SP2/0 cells (ATCC), using the standard techniques [1977 Somat Cell Genet, 3:231].
  • Assessment of Tigit Binding Activity of Antibodies by ELISA and FACS
  • The supernatants of hybridoma clones were initially screened by ELBA as described in “Methods in Molecular Biology (2007) 378:33-52” with some modifications. Briefly, Tigit-huIgG1 protein was coated in 96-well plates. The HRP-linked anti-mouse IgG antibody (Cat.: 7076S, Cell Signaling Technology, USA) and substrate (Cat.: 00-4201-56, eBioscience, USA) were used to develop color absorbance signal at the wavelength of 450 nm, which was measured by using a plate reader (SpectraMax Paradigm, Molecular Devices, USA). The ELISA-positive clones were further verified by FACS using either NK92MI/huTigit or NK92mi/mkTigit cells described above. Tigit-expressing cells (105 cells/well) were incubated with ELISA-positive hybridoma supernatants, followed by binding with Alexa Fluro-647 labeled goat anti-mouse IgG antibody (Cat.: A0473, Beyotime Biotechnology, China). Cell fluorescence was quantified using a flow cytometer (Guava easyCyte 8HT, Merck-Millipore, USA).
  • The conditioned media from the hybridomas that showed positive signals in both ELISA and FACS screening were subjected to functional assays to identify antibodies with good functional activity in human immune cell-based assays (see following sections). The antibodies with desired. functional activities were further sub-cloned and characterized.
  • Subcloning and Adaptation of Hybridomas Serum-Free or Low Serum Medium
  • After primary screening by ELISA, FACS and functional assays as described above, the positive hybridoma clones were sub-clotted by the limiting dilution. Three positive subclones based on ELISA and FACS screening from each plate were selected and characterized by functional assays. The top antibody subclones verified through functional assays were adapted for growth in the CDM4MAb medium (Cat.: SH30801.02, Hyclone, USA) with 3% FBS.
  • Expression and Purification of Monoclonal Antibodies
  • Hybridoma cells or 293G cells transiently transfected with an antibody expression plasmid (Cat. No. R79007, Invitrogen) was cultured either in CDM4MAb medium (Cat.: SH30801.02, Hyclone) or in Freestyle™ 293 Expression medium (Cat.: 12338018, Invitrogen), and incubated in a C02 incubator for 5 to 7 days at 37° C. The conditioned medium was collected through centrifugation and filtrated by passing a 0.22 μm membrane before purification. Murine or recombinant antibodies containing supernatants were applied and bound to a Protein A column (Cat.: 17127901, GE Life Sciences-) following the manufacturer's guide. The procedure usually yielded antibodies at purity above 90%. The Protein A-affinity purified antibodies were either dialyzed against PBS or further purified using a HiLoad 16/60 Superdex200 column (Cat.: 17531801, GE Life Sciences) to remove aggregates. Protein concentrations were determined by measuring absorbance at 280 nm. The final antibody preparations were stored in aliquots in −80° C. freezer.
  • Example 2 Cloning and Sequence Analysis of Tigit Antibodies
  • Murine hybridoma clones were harvested to prepare total cellular RNAs using Ultrapure RNA kit (Cat.: 74104, QIAGEN, Germany) based on the manufacturer's protocol. The 1st strand cDNAs were synthesized using a cDNA synthesis kit from Invitrogen (Cat.: 18080-051) and PCR amplification of the nucleotide sequences coding for heavy chain variable region (Vh) and kappa chain variable region (Vk) of murine mAbs was performed using a PCR kit (Cat.: CW0686, CWBio, Beijing, China). The oligo primers used for antibody cDNAs cloning of Vh and Vk were synthesized by Invitrogen (Beijing, China) based on the sequences reported previously (Brocks et al. 2001 Mol Med 7:461). PCR products were then subcloned into the pEASY-Blunt cloning vector (Cat.: CB101-02, TransGen, China) and sequenced by Genewiz (Beijing, China). The amino acid sequences of Vh and Vk regions were deduced from the DNA sequencing results.
  • The murine mAbs were analyzed by comparing sequence homology and grouped based on sequence similarity (FIG. 2). Complementary determinant regions (CDRs) were defined based on the Kabat [FWu and Kabat 1970 J. Exp. Med. 132:211-250] and IMGT [Lefranc 1999 Nucleic Acids Research 27:209-212] system by sequence annotation and by internet-based sequence analysis in IMGT. The amino acid sequences of a representative top clone mu1217 (Vh and Vk) were listed in Table 1 (SEQ ID NOs: 9 and 11). The CDR sequences of mu1217 were listed in Table 2 (SEQ ID NOs: 3-8).
  • TABLE 1
    Amino acid sequences of mu 1217 Vh and Vk regions
    mu1217 Vh SEQ ID NO 9
    mu1217 Vk SEQ ID NO 11
  • TABLE 2
    CDR sequences (amino acids) of Mn 1217 Vh and Vk regions
    mAbs CDR1 CDR2 CDR3
    mu1217, Vh SEQ ID NO 3 SEQ ID NO 4 SEQ ID NO 5
    mu1217, Vk SEQ ID NO 6 SEQ ID NO 7 SEQ ID NO 8
    Note:
    CDR sequences are defined based on Kabat system
  • Example 3 Affinity Determination of Purified Murine Anti-Tigit Antibodies by SPR
  • The Tigit antibodies with high binding activities in ELISA and FACS, as well as with potent functional activities in the cell-based assays (described in Examples 1 and 2) were characterized for their binding kinetics by SPR assays using BIAcore™ T-200 (GE Life Sciences). Briefly, anti-human IgG antibody was immobilized on an activated CM5 biosensor chip (Cat. No.: BR100530, GE Life Sciences), Human Fc-tagged Tigit was flowed over the chip surface and captured by anti-human IgG antibody. Then a serial dilution (0.12 nM to 10 nM) of purified murine antibodies were flowed over the chip surface and changes in surface plasmon resonance signals were analyzed to calculate the association rates (kon) and dissociation rates (koff) by using the one-to-one Langmuir binding model (BIA Evaluation Software, GE Life Sciences). The equilibrium dissociation constant (KD) was calculated as the ratio koff/kon. The binding affinity profiles of top mAbs including mu1217, mu1257, mu1226 and mu242, were shown in FIG. 3 and Table 3.
  • TABLE 3
    Binding affinities of hybridoma antibodies by SPR
    Antibodies kon (M−1 s−1) koff(s−1) ko(nM)
    mu1217 4.33E+06 3.96E−05 9.15E−12
    mu1257 3.99E+06 4.20E−05 1.05E−11
    mu1266 1.07E+07 8.49E−05 7.94E−12
    mu242 5.12E+06 7.13E−05 1.39E−11
  • Example 4 Humanization of the Murine Anti-Human Tigit mAb Mu1217
  • mAb Humanization and Engineering
  • For humanization of the mu1217, human germline IgG genes were searched for sequences that share high degrees of homology to the cDNA sequences of mu1217 variable regions by blasting the human immunoglobulin gene database in IMGT and NCBI websites. The human IGVH and IGVK genes that are present in human antibody repertoires with high frequencies (Glanville 2009 PNAS 106:20216-20221) and are highly homologous to mu1217 were selected as the templates for humanization.
  • Humanization was carried out by CDR-grafting (Methods in Molecular Biology, Vol 248: Antibody Engineering, Methods and Protocols, Humana Press) and the humanization antibodies (hu1217s) were engineered as the human IgG1mf format using an in-house developed expression vector. In the initial round of humanization, mutations from murine to human amino acid residues in framework regions were guided by the simulated 3D structure, and the murine framework residues of structural importance for maintaining the canonical structures of CDRs were retained in the 1st version of humanization antibody 1217 (hu1217-1-1, with six CDRs having amino acid sequences of SEQ ID NOs: 3, 13, 5 (heavy chain CDRs) and SEQ ID NOs: 6, 7, 8 (light chain CDRs), a heavy chain variable region having an amino acid sequence of SEQ ID NO: 14 and encoded by a nucleotide sequence of SEQ ID NO: 15, and a light chain variable region having an amino acid sequence of SEQ ID No:16 and encoded by a nucleotide sequence of SEQ NO: 17). Specifically, CDRs of mu1217 VK (SEQ ID NO: 6-8) were grafted into the framework of human germline variable gene IGVk3-15 with 1 murine framework residue (V58) retained, resulting in the humanized VK sequence of Hu1217-1-1 (SEQ ID NO: 16 for amino acid sequence and SEQ ID NO: 17 for nucleotide sequence). N-terminal of H-CDR2 (SEQ ID NO: 4), H-CDRT and H-CDR3 (SEQ ID NOs: 3 and 5) of mu1217 Vh were grafted into the framework of human germline variable gene IGVH3-7 with two murine framework (T24 and I37 of SEQ NO: 10) residues retained. In the hu1217 humanization variants, only the N-terminal half of Kabat H-CDR2 was grafted, as only the N-terminal half was predicted to be important for antigen binding according to the simulated 3D structure. The amino acid sequence and nucleotide sequence of the resultant humanized Vh sequence of Hu1217-1-1 are shown in SEQ ID NO: 14 and SEQ ID NO: 15, respectively.
  • Hu1217-1-1 were constructed as human full-length antibody format using in-house developed expression vectors that contain constant regions of a human IgG1 variant termed as IgG1mf (SEQ ID NO: 18) and kappa chain, respectively, with easy adapting sub-cloning sites. Expression and preparation of hu1217-1-1 antibody was achieved by co-transfection of the above two constructs into 293G cells and by purification using a protein A column (Cat.: 17543802, GE Life Sciences). The purified antibodies were concentrated to 0.5-5 mg/mL in PBS and stored in aliquots in −80° C. freezer.
  • Based on hu1217-1-1 template, we made several single-mutations converting the retained murine residues in framework region of Vk to corresponding human germline residues, which include V581 in VK and in T24A and 137V Vh. The resulted hu1217-2A-1 (T24A), hu1217-2B-1 (137V), and hu1217-1-2a (V58I) all had similar binding and functional activities to hu1217-1-1. All humanization mutations were made using primers containing mutations at specific positions and a site directed mutagenesis kit (Cat. No. FM111-02, TransGen, Beijing, China). The desired mutations were verified by sequencing analysis. These hu1217-derived variant antibodies were tested in binding and functional assays as described previously.
  • Hu1217 antibodies were further engineered by introducing mutations in CDRs and framework regions to improve molecular and biophysical properties for therapeutic use in human. The considerations include amino acid compositions, heat stability (Tm), surface hydrophobicity and isoelectronic points (pIs) while maintaining functional activities.
  • Taken together, a well-engineered version of humanized monoclonal antibody, hu1217-2-2 (SEQ ID NOs:3, 5-8, 13, and 19-21), was derived from the mutation process described as above, and characterized in detail. The results showed both hu1217-2-2 and hu1217-1-1 were very similar in binding affinity and functional activities such as inhibiting the Tigit-mediated downstream signaling.
  • For affinity determination, antibodies were captured by anti-human Fc surface, and used in the affinity-assay based on surface plasmon resonance (SPR) technology. The results of SPR-determined binding profiles of anti-Tigit antibodies were summarized in Table 4. Hu1217-2-2 and hu1217-1-1 showed very similar binding profiles with average dissociation constant at 0.415 nM and 0.266 nM, respectively, which are close to that of ch1217.
  • TABLE 4
    Binding affinities of hu1217 antibodies by SPR
    Test
    1 Test 2 Mean
    Kon Koff KD Kon Koff KD KD
    Antibodies (M−1s−1) (s−1) (nM) (M−1s−1) (s−1) (nM) (nM)
    ch1217* 1.56 × 106 4.43 × 10−4 0.283 NA**
    hu1217-1-1 1.45 × 106 4.48 × 10−4 0.309 1.33 × 106 6.94 × 10−4 0.520 0.415
    hu1217-2-2 1.80 × 105 2.29 × 10−4 0.127 1.50 × 106 6.08 × 10−4 0.404 0.266
    *ch1217 is comprised of mu1217 variable domains fused to human IgG1mf/kappa constant regions
    **NA: not available.
  • TABLE 5
    CDRs of hu1217 antibodies
    Antibodies CDR1 CDR2 CDR3
    hu1217-1.-1, Vh SEQ ID NO 3 SEQ ID NO 13 SEQ ID NO 5
    hu121.7-2-2, Vh SEQ ID NO 3 SEQ ID NO 13 SEQ ID NO 5
    hu1217-1-1, Vκ SEQ ID NO 6 SEQ ID NO 7 SEQ ID NO 8
    hu1217-2-2, Vκ SEQ ID NO 6 SEQ ID NO 7 SEQ ID NO 8
  • All the humanization antibodies shown above were also confirmed for functional activities on primary human immune cells isolated from healthy donors (described in Example 7).
  • Example 5 Binding Activities of Different Versions of 1217 to Native Tigit
  • To evaluate the binding activity of anti-Tigit antibodies to native Tigit on living cells, NK92mi cells were engineered to over-express human Tigit. Living NK92mi/Tigit cells were seeded in 96-well plate, and were incubated with a series of dilutions of anti-Tigit antibodies, Goat anti-Human IgG was used as secondary antibody to detect antibody binding to the cell surface. EC50 values for dose-dependent binding to human native Tigit were determined by fitting the dose-response data to the four-parameter logistic model with GraphPad Prism. As show in FIG. 4 and Table 6. Both humanized 1217 antibodies, hu1217-1-1 and hu1217-2-2, showed good binding affinity to native Tigit on living cells.
  • TABLE 6
    EC50 of dose-dependent binding of humanized
    1217 variants to native Tigit
    EC50(ug/mL)
    Antibodies Test 1 Test 2
    Ch1217 0.100
    hu1217-1-1 0.114 0.084
    hu1217-2-2 0.068
  • Example 6 Anti-Tigit Antibodies Block the Interactions of Tigit with its Ligands PVR and PVR-L2
  • Tigit binds to PVR with a high affinity (Kd: ˜1 nM), which can compete against CD266-PVR interaction [Yu et al., 2009].
  • To determine whether anti-Tigit antibodies could block Tigit-PVR and Tigit-PVR-L2 interactions, HEK293 cells were engineered to express high levels of MIR or PVR-L2. The resultant cell lines were named HEK293/PVR and HEK293/PVR-L2, respectively. The binding of soluble Tigit (Tigit-mIgG2a fusion protein) to PVR or PVR-L2 was determined by flow cytometry (FIG. 5A). The blockade of Tigit-ligand interaction was quantitatively measured by adding serially diluted anti-Tigit antibodies. As shown in FIG. 5B, hu1217-2-2/IgG1 (a humanized version comprising a wild-type IgG1 Fc region and having the same VH and VL sequences as hu1217-2-2/IgG1mf) and hu1217-2-2/IgG1mf could block Tigit binding to PVR in a dose-dependent manner with IC50 at 0.64 and 0.55 μg/mL, respectively. Similarly, the IC50 of hu1217-2-2/IgG1 and hu1217-2-2/IgG1mf in blocking Tigit-PVR-L2 interaction is 0.25 and 0.18 μg/mL, respectively.
  • Example 7 Activation of CMV-Specific Human T Cells by Anti-Tigit Antibodies
  • The functional activity of the Tigit antibodies were further assessed using naturally derived T-cells that recognized human CMV PP65 peptide (NLVPMVATV, 495-503. HLA-A2.1-restricted) [Boeckh M, Boeckh M and Geballe A P, 2011 J Clin Invest. 121:1673-80]. Briefly, PBMCs from HLA-A2.1+ healthy donors were simulated with PP65 peptide (>98% purity, synthesized by GL Biochem, Shanghai) in the complete RPMI with 10% FBS for a week. The pp65-primed PBMCs were used as effector cells. Prior to assay, target cells, HCT116 cells (HLA-A2.1+, 10 4), were pulsed with pp65 peptide (5 μg/mL) for 30 mins and co-cultured with equal numbers of pp65-sensitized PBMCs in 96-well plates overnight in the presence or absence of anti-Tigit antibodies or a blank control (medium only). As shown in FIG. 6, hu1217-2-2/IgG1 promoted pp65-specific T cells to secrete IFN-γ in the cell culture supernatant in a dose-dependent manner for both donors.
  • Example 8 Anti-Tigit Antibodies Enhanced NK Cell-Mediated Cytotoxicity
  • Tigit is known to be constitutively expressed on natural killer (NK) cells at relatively higher levels and the interaction between Tigit and its ligands inhibits NK cell-mediated cytotoxicity [Wang F, et al. 2015 Eur J. Immunology 45:2886-97; Stanietsky N et al., 2009 Proc Natl Acad Sci USA 106:17858-63].
  • To confirm whether humanized anti-Tigit antibodies could promote NK-mediated cytotoxicity, an NK cell line NK92MI was engineered to co-express both Tigit and DNAM-1 receptors (NK92MI/Tigit-DNAM-1) as an effector cell by retroviral transduction, according to the protocols described previously [Zhang et al, 2006 Cancer Res. 66: 5927-5933], A PVR-expressing lung cancer cell line SK-MES-1/PVR was established similarly as a target.
  • Cytotoxicity of NK92MI/Tigit-DNAM-1 cells against SK-MES-1/PVR cells was determined by an LDH release assay using the CytoTox 96 Non-Radioactive Cytotoxicity Assay kit (Promega, Madison, Wis.). In brief, NK92MI/Tigit-DNAM-1 cells (8×105) were co-cultured with SK-MES-1/PVR cells (2×104) in the presence of anti-Tigit Abs (0.007-30 μg/mL) for 5 hr in 96-well V-bottom plates. LDH-release assay Specific lysis was determined using the following equation: percentage of specific lysis=[(experimental-effector spontaneous-target spontaneous)/(target maximum−target spontaneous)]×100. The results showed that anti-Tigit antibodies hu1217-2-2/IgG1mf enhances NK cell killing in a dose-dependent manner (EC50: 0.185 μg/mL) (FIG. 7).
  • Example 9 Anti-Tigit Antibodies can Reduce the Surface Expression of Tigit Receptor Via FcγR-Mediated Trogocytosis
  • Trogocytosis is a phenomenon, in which cell surface molecules are transferred from donor cells to acceptor cells [Joly E, et al. 2003 Nat. Immunol: Machlenkin A. et al. 2008 Cancer Res.; Beum P V et al. 2008 J Immunol; Rossi E A, et al. 2013 Blood]. Antibody-induced trogocytosis via Fe γ receptors (FcγRs) leads to down-modulation of receptors on the cell surface [Carlsten M, et al. 2016 Clin Cancer Res; Beum P V et al. 2011 J. Immunology]. Therefore, down-regulation of target receptor by trogocytosis may cause dampened signaling. In view of these observations, it would be possible that hu1217-2-2/IgG1 might induce trogocytosis of Tigit receptor in the presence of FcγR+ cells, resulting in lower surface expression. To address this possibility, Jurkat/Tigit cells were incubated with HEK cells expressing various FcγRs (including FcγRIIAH131, FcγRIIB, FcγRIIAvlss) with biotin-labeled hu1217-2-2/IgG1wt (a humanized antibody comprising the same VL and VH sequences as hu1217-2-2/IgG1mf and a wild-type IgG1 Fc region) or hu1217-2-2/IgG1mf overnight. Surface expression of Tigit receptor was determined by with SA-APC (Biolegend). As shown in FIG. 8, hu1217-2-2/IgG1 but not hu1217-2-2/IgG1mf caused a significant reduction of Tigit surface expression compared to the negative control human IgG-treated cells, indicating that the reduction of surface Tigit on Jurkat/Tigit cells are FcγR-binding dependent. In addition, presence of 10% human serum (containing high-level of endogenous IgG) could partially reduce FcγRIIAH131- or FcγRIIIA58-, but not FcγRIIB-mediated trogocytosis of Tigit receptor, suggesting that FcγRIIB could play a critical role reducing Tigit surface expression by anti-Tigit mAbs (e.g., hu1217-2-2/IgG1wt) in vivo. These observations are also consistent with previous findings [Gonesan L P, et al 2012 J Immunol 189:4981-8; Taylor R P, et al. 2015 Blood 125:762-6].
  • Example 10 ADCC and CDC Effector Functions of Anti-Tigit Antibodies
  • The abilities of anti-Tigit antibodies to induce ADCC and CDC in human primary PBMCs were determined using in vitro assay as described below.
  • ADCC Using Human PBMCs as Target Cells
  • A flow cytometry-based. ADCC assay was set up to determine whether Tigit antibodies could induce ADCC in Tigit T+ cells. The assay effector cell line, NK92MI/CD16V cells, was generated from NK92MI cells (ATCC) by co-transducing expression plasmids containing CD16V158 (V158 allele) and FcγR cDNAs. Human PBMCs from healthy donors were stimulated with PHA (1 μg/ml) to up-regulate Tigit expression. As shown in FIG. 9, T cells, including CD4+ effector (CD3+CD4+Foxp3), CD8+ and regulator T cells (CD4+Foxp3+) all expressed significant amounts of Tigit. These activated PBMCs (from 3 healthy donors) were used as target cells. A fluorescent dye CFSE-labeled NK92MI/CD16V cells (5×104) were co-cultured with equal number of target cells, for 40 hours in the presence of Tigit antibodies (hu1217-2-2/IgG1mf or hu1217-2-2/IgG1wt, 30 μg/mL) or control antibodies (the positive control anti-CD3 antibody OKT3 (5 μg/ml, Biolegend) or a negative control human IgG, 30 μg/mL). Compared with human IgG and hu1217-2-2/IgG1mf, hu1217-2-2/IgG1wt could lead to moderate reduction of Tregs via ADCC. However, no significant ADCC effects were observed in total T cells and CD8+ T cells (FIG. 9).
  • CDC Using Human PBMCs as Target Cells
  • Whether hu1217-2-2/IgG1mf and hu1217-2-2/IgG1wt would trigger CDC was determined by using pre-activated human PBMCs and fresh autologous sera from healthy donors. Cell lysis by CDC was determined by a Celltiter glo assay kit (Promega, Beijing, China). In brief, PBMCs were pre-activated with PHA (10 μg/mL) for 3 days, and then were incubated in RPMI1.640 plus autologous serum (15%) and anti-Tigit or control antibodies (0.01-100 μg/mL) for overnight at 37° C. The cell death due to CDC was assayed by the decrease of ATP released from viable cells after cell lysis at the end of reaction. Anti-MHC-1 A, B, C was used as a positive control. The fluorescence readout was conducted using a 96-well fluorometer (PHERA Star FS, BMG LABTECH), and the CDC activities were calculated from the relative fluorescence unit (RFU) readout as follows: % CDC activity=[(RFU test−RFU background) (RFU at total cell lysis RFU background)]×100. The experimental results demonstrated that both hu1217-2-2/IgG1mf and hu1217-2-2/IgG1wt had no detactable CDC with PBMCs isolated from two different donors. In contrast, the positive control antibody, anti-MHC-1, induces significant CDC activity (FIG. 10).
  • Example 11 pH Dependent Binding Affinity of Hu1217-2-2/IgG1
  • To investigate whether pH would influence the binding property of hu1217-2-2/IgG1, target binding SPR tests were performed in running buffers at pH 7.4 and at pH 6.0 for comparison. The antibody hu1217-2-2/IgG1 was immobilized to a CM5 chip (GE). Serial dilutions of TIGIT-his were flown over the immobilized hu1217-2-2/IgG1 in running buffer HBS at pH 7.4 or pH 6.0.
  • As shown by the results listed in Table 7 below, hu1217-2-2/IgG1 showed higher binding affinity (KD) and stronger binding signal (Rmax) against human TIGIT at PH 6.0 (an acidic pH which is similar to the pH of tumor microenvironment) as compared to the data obtained at pH 7.4 (physiologic pH). These results indicate a potential advantage of the antibody as a therapeutic agent targeting TIGIT-positive lymphocytes in the tumor environment, since hu1217-2-2/IgG1 might more selectively target the TIGIT-positive lymphocytes in the tumor microenvironment while have lower potential toxicity associated with activation of periphery lymphocytes.
  • TABLE 7
    Binding affinities of hu1217-2-2/IgG1
    at pH 7.4 and PH 6.0 by SPR
    pH kon (M−1 s−1) koff (s−1) KD (M) Rmax(RU)
    7.4 4.34E+05 9.53E−05 2.19E−10 21
    6.0 2.54E+06 7.60E−05 2.99E−11 37

Claims (23)

1. A pharmaceutical composition comprising an antibody or an antigen-binding fragment thereof, which is capable of binding to human Tigit, comprising:
(a) a heavy chain variable region (VH) comprising heavy chain complementarity determining region (CDR)1, CDR2, and CDR3 comprising amino acid sequences of SEQ ID NOs: 3, 13, and 5, respectively; and
(b) a light chain variable region (VL) comprising light chain CDR1, CDR2, and CDR3 comprising amino acid sequences of SEQ ID NOs: 6, 7 and 8, respectively;
wherein the composition is in a liquid solution form.
2. The pharmaceutical composition of claim 1, wherein the antibody or fragment thereof is a humanized antibody molecule.
3. The pharmaceutical composition of claim 1, wherein the antibody or fragment thereof comprises a heavy chain variable domain having at least 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO 14, and a light chain variable domain having at least 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO 16.
4. The pharmaceutical composition of claim 1, wherein the antibody or fragment thereof comprises a heavy chain variable domain having at least 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO 19, and a light chain variable domain having at least 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO 21.
5. The pharmaceutical composition of claim 1, wherein the antibody or fragment thereof comprises:
(a) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO 14, and a light chain variable domain comprising the amino acid sequence of SEQ ID NO 16; or
(b) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO 19, and a light chain variable domain comprising the amino acid sequence of SEQ ID NO 21.
6. The pharmaceutical composition of claim 1, wherein the antibody or fragment thereof comprises one or more of:
(a) a heavy chain variable domain with a T to A mutation at position 24 of SEQ ID NO 14;
(b) a heavy chain variable domain with a I to V mutation at position 37 of SEQ ID NO 14;
(c) a light chain variable domain with a V to I mutation at position 58 of SEQ ID NO 16.
7. The pharmaceutical composition of claim 1, wherein the antigen-binding fragment is a Fab, F(ab′)2, Fv, or a single chain Fv(ScFv).
8. The pharmaceutical composition of claim 1, wherein the antibody comprises a heavy chain constant region of the subclass of IgG1, IgG2, IgG3, or IgG4 or a variant thereof, and a light chain constant region of the type of kappa or lambda or a variant thereof.
9. The pharmaceutical composition of claim 1, wherein the composition is suitable for parenteral administration.
10. The pharmaceutical composition of claim 9, wherein the composition is suitable for intravenous administration.
11. The pharmaceutical composition of claim 1, further comprising second therapeutic agent.
12. The pharmaceutical composition of claim 11, wherein the second therapeutic agent is selected from a chemotherapy, an oncolytic drug, a cytotoxic agent, an immune-based therapy, a cytokine, an activator of a costimulatory molecule, an inhibitor of an inhibitory molecule, a vaccine, or a cellular immunotherapy.
13. A pharmaceutical composition comprising an antibody or an antigen-binding fragment thereof, which is capable of binding to human Tigit, comprising:
(a) a heavy chain variable region (VH) comprising heavy chain complementarity determining region (CDR)1, CDR2, and CDR3 comprising amino acid sequences of SEQ ID NOs: 3, 13, and 5, respectively; and
(b) a light chain variable region (VL) comprising light chain CDR1, CDR2, and CDR3 comprising amino acid sequences of SEQ ID NOs: 6, 7 and 8, respectively;
wherein the composition is an injectable or infusion solution.
14. The pharmaceutical composition of claim 13, wherein the antibody or fragment thereof is a humanized antibody molecule.
15. The pharmaceutical composition of claim 13, wherein the antibody or fragment thereof comprises a heavy chain variable domain having at least 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO 14, and a light chain variable domain having at least 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO 16.
16. The pharmaceutical composition of claim 13, wherein the antibody or fragment thereof comprises a heavy chain variable domain having at least 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO 19, and a light chain variable domain having at least 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO 21.
17. The pharmaceutical composition of claim 13, wherein the antibody or fragment thereof comprises:
(a) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO 14, and a light chain variable domain comprising the amino acid sequence of SEQ ID NO 16; or
(b) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO 19, and a light chain variable domain comprising the amino acid sequence of SEQ ID NO 21.
18. The pharmaceutical composition of claim 13, wherein the antibody or fragment thereof comprises one or more of:
(a) a heavy chain variable domain with a T to A mutation at position 24 of SEQ ID NO 14;
(b) a heavy chain variable domain with a I to V mutation at position 37 of SEQ ID NO 14;
(c) a light chain variable domain with a V to I mutation at position 58 of SEQ ID NO 16.
19. The pharmaceutical composition of claim 13, wherein the antigen-binding fragment is a Fab, F(ab′)2, Fv, or a single chain Fv(ScFv).
20. The pharmaceutical composition of claim 13, wherein the antibody comprises a heavy chain constant region of the subclass of IgG1, IgG2, IgG3, or IgG4 or a variant thereof, and a light chain constant region of the type of kappa or lambda or a variant thereof.
21. The pharmaceutical composition of claim 13, wherein the composition is suitable for intravenous injection or intravenous infusion.
22. The pharmaceutical composition of claim 13, further comprising second therapeutic agent.
23. The pharmaceutical composition of claim 22, wherein the second therapeutic agent is selected from a chemotherapy, an oncolytic drug, a cytotoxic agent, an immune-based therapy, a cytokine, an activator of a costimulatory molecule, an inhibitor of an inhibitory molecule, a vaccine, or a cellular immunotherapy.
US17/675,515 2017-12-30 2022-02-18 Anti-tigit antibodies and their use as therapeutics and diagnostics Pending US20220251195A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/675,515 US20220251195A1 (en) 2017-12-30 2022-02-18 Anti-tigit antibodies and their use as therapeutics and diagnostics

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CNPCT/CN2017/120392 2017-12-30
CN2017120392 2017-12-30
PCT/CN2018/125375 WO2019129261A1 (en) 2017-12-30 2018-12-29 Anti-tigit antibodies and their use as therapeutics and diagnostics
US202016958262A 2020-06-26 2020-06-26
US17/534,916 US20220153837A1 (en) 2017-12-30 2021-11-24 Anti-tigit antibodies and their use as therapeutics and diagnostics
US17/675,515 US20220251195A1 (en) 2017-12-30 2022-02-18 Anti-tigit antibodies and their use as therapeutics and diagnostics

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US17/534,916 Continuation US20220153837A1 (en) 2017-12-30 2021-11-24 Anti-tigit antibodies and their use as therapeutics and diagnostics

Publications (1)

Publication Number Publication Date
US20220251195A1 true US20220251195A1 (en) 2022-08-11

Family

ID=67063240

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/958,262 Active US11214616B2 (en) 2017-12-30 2018-12-29 Anti-TIGIT antibodies and their use as therapeutics and diagnostics
US17/534,916 Pending US20220153837A1 (en) 2017-12-30 2021-11-24 Anti-tigit antibodies and their use as therapeutics and diagnostics
US17/675,515 Pending US20220251195A1 (en) 2017-12-30 2022-02-18 Anti-tigit antibodies and their use as therapeutics and diagnostics

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US16/958,262 Active US11214616B2 (en) 2017-12-30 2018-12-29 Anti-TIGIT antibodies and their use as therapeutics and diagnostics
US17/534,916 Pending US20220153837A1 (en) 2017-12-30 2021-11-24 Anti-tigit antibodies and their use as therapeutics and diagnostics

Country Status (14)

Country Link
US (3) US11214616B2 (en)
EP (1) EP3731868A4 (en)
JP (2) JP7287963B2 (en)
KR (1) KR20200105849A (en)
CN (2) CN111526888B (en)
AU (1) AU2018393448A1 (en)
BR (1) BR112020012647A8 (en)
CA (1) CA3086935A1 (en)
EA (1) EA202091587A1 (en)
IL (1) IL274934A (en)
MX (1) MX2020006896A (en)
SG (1) SG11202005213WA (en)
TW (2) TWI816729B (en)
WO (1) WO2019129261A1 (en)

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HUE062927T2 (en) 2017-05-01 2023-12-28 Agenus Inc Anti-tigit antibodies and methods of use thereof
TWI816729B (en) 2017-12-30 2023-10-01 英屬開曼群島商百濟神州有限公司 Anti-tigit antibodies and their use as therapeutics and diagnostics
KR102224556B1 (en) * 2018-02-06 2021-03-09 아이-맵 바이오파마 유에스 리미티드 Antibodies against T cell immune receptors (TIGIT) with IG and ITIM domains and their use
CN113614109A (en) 2018-12-21 2021-11-05 Ose免疫疗法公司 Bifunctional anti-PD-1/IL-7 molecules
WO2020165374A1 (en) 2019-02-14 2020-08-20 Ose Immunotherapeutics Bifunctional molecule comprising il-15ra
TW202128755A (en) * 2019-09-27 2021-08-01 英商葛蘭素史密斯克藍智慧財產發展有限公司 Antigen binding proteins
US20230057899A1 (en) 2019-12-05 2023-02-23 Compugen Ltd. Anti-pvrig and anti-tigit antibodies for enhanced nk-cell based tumor killing
WO2021122866A1 (en) 2019-12-17 2021-06-24 Ose Immunotherapeutics Bifunctional molecules comprising an il-7 variant
US20230258645A1 (en) 2020-05-07 2023-08-17 Institut Curie Antxr1 as a biomarker of immunosuppressive fibroblast populations and its use for predicting response to immunotherapy
WO2021247591A1 (en) 2020-06-02 2021-12-09 Arcus Biosciences, Inc. Antibodies to tigit
KR20230024368A (en) 2020-06-18 2023-02-20 제넨테크, 인크. Treatment with anti-TIGIT antibodies and PD-1 axis binding antagonists
CN111718415B (en) * 2020-07-03 2021-02-23 上海洛启生物医药技术有限公司 anti-TIGIT nano antibody and application thereof
TWI815220B (en) * 2020-11-23 2023-09-11 大陸商廣東菲鵬制藥股份有限公司 Anti-tigit antibody or antigen-binding fragment thereof, method of producing and application for the same, and nucleic acid, vector, cells, pharmacuetical composition and test kit comprising the same
WO2022112198A1 (en) 2020-11-24 2022-06-02 Worldwide Innovative Network Method to select the optimal immune checkpoint therapies
WO2022148781A1 (en) 2021-01-05 2022-07-14 Institut Curie Combination of mcoln activators and immune checkpoint inhibitors
EP4320156A1 (en) 2021-04-09 2024-02-14 Ose Immunotherapeutics Scaffold for bifunctioanl molecules comprising pd-1 or cd28 and sirp binding domains
AU2022253351A1 (en) 2021-04-09 2023-10-12 Ose Immunotherapeutics New scaffold for bifunctional molecules with improved properties
TW202304965A (en) 2021-05-04 2023-02-01 美商艾吉納斯公司 Anti-tigit antibodies, anti-cd96 antibodies, and methods of use thereof
WO2022240159A1 (en) * 2021-05-10 2022-11-17 메디맵바이오 주식회사 Anti-tigit antibodies and use thereof
WO2023010094A2 (en) 2021-07-28 2023-02-02 Genentech, Inc. Methods and compositions for treating cancer
WO2023056403A1 (en) 2021-09-30 2023-04-06 Genentech, Inc. Methods for treatment of hematologic cancers using anti-tigit antibodies, anti-cd38 antibodies, and pd-1 axis binding antagonists
WO2023215719A1 (en) 2022-05-02 2023-11-09 Arcus Biosciences, Inc. Anti-tigit antibodies and uses of the same
WO2023240058A2 (en) 2022-06-07 2023-12-14 Genentech, Inc. Prognostic and therapeutic methods for cancer
WO2023241659A1 (en) * 2022-06-16 2023-12-21 Beigene, Ltd. Methods of treating lymphoma using anti-tigit antibodies
WO2024003360A1 (en) 2022-07-01 2024-01-04 Institut Curie Biomarkers and uses thereof for the treatment of neuroblastoma
WO2024012584A1 (en) * 2022-07-15 2024-01-18 Beigene Switzerland Gmbh Methods of cancer treatment using anti-tigit antibodies
WO2024028386A1 (en) 2022-08-02 2024-02-08 Ose Immunotherapeutics Multifunctional molecule directed against cd28
WO2024041639A1 (en) * 2022-08-26 2024-02-29 Nanjing Legend Biotech Co., Ltd. Antibodies targeting tigit and uses thereof

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI2925350T1 (en) * 2012-12-03 2019-04-30 Bristol-Myers Squibb Company Enhancing anti-cancer activity of immunomodulatory fc fusion proteins
KR20230118704A (en) * 2013-07-16 2023-08-11 제넨테크, 인크. Methods of treating cancer using pd-1 axis binding antagonists and tigit inhibitors
AU2015289621A1 (en) * 2014-07-16 2017-01-12 Genentech, Inc. Methods of treating cancer using TIGIT inhibitors and anti-cancer agents
UA122395C2 (en) 2014-08-19 2020-11-10 Мерк Шарп Енд Доум Корп. ANTIBODY AGAINST TIGIT
MX2017006323A (en) * 2014-11-21 2017-08-21 Bristol Myers Squibb Co Antibodies comprising modified heavy constant regions.
KR102644115B1 (en) * 2014-12-23 2024-03-05 브리스톨-마이어스 스큅 컴퍼니 Antibodies to tigit
EP3334757A4 (en) * 2015-08-14 2019-04-03 Merck Sharp & Dohme Corp. Anti-tigit antibodies
RU2732591C2 (en) * 2015-09-25 2020-09-21 Дженентек, Инк. Anti-tigit antibodies and methods of using
RU2729379C1 (en) * 2015-10-01 2020-08-06 Потенза Терапевтикс, Инк. Anti-tigit antigen-binding proteins and methods for use thereof
JP7317272B2 (en) 2017-09-29 2023-07-31 ジエンス ヘンルイ メデイシンカンパニー リミテッド TIGIT Antibodies, Antigen-Binding Fragments Thereof, and Medical Uses Thereof This application is based on and claims priority from Application No. CN201710908565.3 filed on September 29, 2019. The disclosure of which is incorporated herein by reference in its entirety.
TWI816729B (en) 2017-12-30 2023-10-01 英屬開曼群島商百濟神州有限公司 Anti-tigit antibodies and their use as therapeutics and diagnostics

Also Published As

Publication number Publication date
CN117186225A (en) 2023-12-08
MX2020006896A (en) 2020-09-07
EA202091587A1 (en) 2020-09-21
US20220153837A1 (en) 2022-05-19
US20200331999A1 (en) 2020-10-22
TW202400654A (en) 2024-01-01
JP2023109951A (en) 2023-08-08
AU2018393448A1 (en) 2020-07-09
CN111526888A (en) 2020-08-11
US11214616B2 (en) 2022-01-04
IL274934A (en) 2020-07-30
TWI816729B (en) 2023-10-01
CN111526888B (en) 2023-12-19
BR112020012647A8 (en) 2023-01-17
EP3731868A1 (en) 2020-11-04
WO2019129261A1 (en) 2019-07-04
CA3086935A1 (en) 2019-07-04
TW201930359A (en) 2019-08-01
JP7287963B2 (en) 2023-06-06
EP3731868A4 (en) 2021-09-01
BR112020012647A2 (en) 2021-01-12
SG11202005213WA (en) 2020-07-29
KR20200105849A (en) 2020-09-09
JP2021508676A (en) 2021-03-11

Similar Documents

Publication Publication Date Title
US20220251195A1 (en) Anti-tigit antibodies and their use as therapeutics and diagnostics
US20240076375A1 (en) Anti-TIM-3 Antibodies and Use Thereof
JP2022130393A (en) Anti-ctla4-anti-pd-1 bifunctional antibodies and pharmaceutical compositions and uses thereof
KR20180100238A (en) Anti-ROR1 antibody, ROR1 x CD3 bispecific antibody, and method of using same
KR20210124959A (en) antibody formulation
JP2024514246A (en) CLDN18.2 antigen binding protein and uses thereof
TWI764291B (en) Anti-tigit antibodies and methods of use
US20230391883A1 (en) Methods of Cancer Treatment Using Anti-TIGIT Antibodies in Combination with Anti-PD1 Antibodies
EA043681B1 (en) ANTI-TIGIT ANTIBODIES AND THEIR USE AS THERAPEUTIC AND DIAGNOSTIC AGENTS
CN116135884A (en) anti-TIGIT-anti-PD-L1 bispecific antibodies, pharmaceutical compositions and uses thereof
EA042181B1 (en) ANTI-TIM-3 ANTIBODIES AND THEIR USE
CN114641500A (en) Methods of treating cancer using anti-OX 40 antibodies in combination with anti-TIM 3 antibodies
CN116438198A (en) Antibodies capable of binding ROR2 and bispecific antibodies binding ROR2 and CD3

Legal Events

Date Code Title Description
AS Assignment

Owner name: BEIGENE SWITZERLAND GMBH, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BEIGENE, LTD.;REEL/FRAME:059052/0194

Effective date: 20220201

Owner name: BEIGENE, LTD., CAYMAN ISLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ZHANG, TONG;XUE, LIU;LIU, QI;AND OTHERS;SIGNING DATES FROM 20200324 TO 20200408;REEL/FRAME:059052/0184

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION