WO2018229163A1 - Methods of activating v delta 2 negative gamma delta t cells - Google Patents

Methods of activating v delta 2 negative gamma delta t cells Download PDF

Info

Publication number
WO2018229163A1
WO2018229163A1 PCT/EP2018/065730 EP2018065730W WO2018229163A1 WO 2018229163 A1 WO2018229163 A1 WO 2018229163A1 EP 2018065730 W EP2018065730 W EP 2018065730W WO 2018229163 A1 WO2018229163 A1 WO 2018229163A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
tigit
antibody
agent
inhibitor
Prior art date
Application number
PCT/EP2018/065730
Other languages
French (fr)
Inventor
Maria Luisa IANNITTO
Oliver Nussbaumer
Adrian HAYDAY
Original Assignee
King's College London
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 King's College London filed Critical King's College London
Publication of WO2018229163A1 publication Critical patent/WO2018229163A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • C12N5/0638Cytotoxic T lymphocytes [CTL] or lymphokine activated killer cells [LAK]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/74Inducing cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins 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
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2302Interleukin-2 (IL-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2304Interleukin-4 (IL-4)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2315Interleukin-15 (IL-15)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2321Interleukin-21 (IL-21)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/599Cell markers; Cell surface determinants with CD designations not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/998Proteins not provided for elsewhere

Definitions

  • the invention relates to methods for activating tissue-infiltrating gamma delta T cells, particularly V52 ⁇ gamma-delta T cells ( ⁇ T cells), in a mammalian subject by antagonizing TIGIT activity of V52 ⁇ ⁇ T cells.
  • ⁇ T cells intra-tumorai gamma delta T cells
  • TCR ⁇ T-cell receptor
  • ⁇ T cells represent a subset of T cells that express on their surface a distinct, defining ⁇ T-cell receptor (TCR).
  • the ⁇ TCR is made up of one gamma ( ⁇ ) and one delta ( ⁇ ) chain.
  • Human ⁇ T cells can be broadly classified into two types: peripheral blood-resident ⁇ T cells and non- hematopoietic, tissue-resident, ⁇ T cells.
  • ⁇ T cells Most blood-resident ⁇ T cells express a ⁇ 2 TCR; however, a smaller population of ⁇ 2 " ⁇ T cells can be detected among ⁇ T cells in blood. Conversely, tissue-resident ⁇ T cells more frequently use ⁇ and/or other ⁇ chains ( ⁇ 2 ⁇ ⁇ T cells); however, a smaller population of ⁇ 2 + ⁇ T cells can be detected among ⁇ T cells in tissue. Therefore, there is a need for novel methods for the specific activation and/or derepression of ⁇ T cells, and in particular, the ⁇ 2 ⁇ ⁇ T cells.
  • the application herein discloses methods of activating and/or de-repressing ⁇ 2 " gamma-delta T cells ( ⁇ T cells) in a mammalian subject, comprising antagonizing TIGIT activity of ⁇ 2 " ⁇ T cells present in the subject to an extent sufficient to activate the ⁇ 2 " ⁇ T cells in vivo.
  • the ⁇ 2 " ⁇ T cells are ⁇ 1 + cells.
  • the ⁇ 2 " ⁇ T cells are ⁇ ⁇ 2 " double negative (DN) cells.
  • antagonizing TIGIT activity comprises administering at least one TIGIT inhibitor to the subject.
  • the at least one TIGIT inhibitor is administered adjunctively with ⁇ 2 " ⁇ T cells that have been cultured ex vivo.
  • the at least one TIGIT inhibitor is added to the ⁇ 2 " ⁇ T cells during culturing ex vivo.
  • the cultured ⁇ 2 " ⁇ T cells have been expanded ex vivo.
  • the at least one TIGIT inhibitor is administered in a composition comprising the cultured ⁇ T cells.
  • the at least one TIGIT inhibitor is administered separately from the ex vivo expanded ⁇ T cells.
  • the methods further comprise administering to the subject at least one additional agent in an amount sufficient to activate ⁇ 2 " ⁇ T cells.
  • the at least one additional agent is an agonist of DNAM-1.
  • the at least one TIGIT inhibitor is selected from the group consisting of an antagonist of TIGIT activity, an antagonist of TIGIT interaction with Polio virus receptor (PVR), an agent that inhibits and/or blocks the interaction of TIGIT with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL2, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL3, an agent that inhibits and/or blocks the intracellular signaling mediated by PVR binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling mediated by PVRL2 binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling mediated by PVLR3 binding to TIGIT, and combinations thereof.
  • PVR Polio virus receptor
  • the TIGIT inhibitor is an antibody. In an aspect, the antibody binds specifically to TIGIT. In an aspect, the antibody binds to PVR to specifically inhibit PVR interaction with TIGIT. In an aspect, the antibody is a full length IgG antibody. In an aspect, the antibody is an antigen-binding antibody fragment. In an aspect, the antibody is a single domain antibody. In an aspect, the antibody is fully human. In an aspect, the antibody is humanized. In an aspect, the TIGIT inhibitor is a small molecule inhibitor.
  • the ex vivo expanded V52 " ⁇ T cells were obtained by culturing lymphocytes obtained from non-hematopoietic tissue of humans or non-human animals in the presence of interleukin-2 (IL-2) and/or interleukin-15 (IL-15), and not in direct contact with stromal or epithelial cells during culture.
  • the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non- hematopoietic tissue in the presence of IL-2.
  • the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of interleukin-15 (IL-15).
  • the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of IL-2 and IL-15. In an aspect, the culturing step comprises culturing the lymphocytes in the absence of TCR activation or co-stimulation signals. In an aspect, the culturing step comprises culturing the lymphocytes in the absence of a T cell receptor pathway agonist. In an aspect, the culturing step comprises culturing the lymphocytes in the absence of stromal or epithelial cells. In an aspect, stromal or epithelial cells are removed prior to culture. In an aspect, the lymphocytes are cultured in the absence of fibroblasts.
  • the lymphocytes have been obtained from skin, the gastrointestinal tract ⁇ e.g. colon), mammary gland tissue, lung, liver, pancreas or prostate.
  • the ex vivo expanded V52 " ⁇ T cells were obtained by culturing lymphocytes obtained from non-hematopoietic tissue of humans or non-human animals in the presence of IL-2, IL-15, and a factor selected from the group consisting of IL-4, IL-21, IL-6, IL-7, IL-8, IL-9, IL-12, IL-18, IL-33, IGF-1, IL- ⁇ , human platelet lysate (HPL), and stromal cell-derived factor- 1 (SDF-1) for at least 5 days to produce an expanded population of ⁇ T cells.
  • IL-2 IL-15
  • the expanded population of ⁇ T cells comprises at least 20-fold the number of ⁇ T cells as the ⁇ T cells obtained from a non-hematopoietic tissue. In an aspect, within 7 days of culture, the expanded population of ⁇ T cells comprises at least 2-fold the number of ⁇ T cells as the ⁇ T cells obtained from a non-hematopoietic tissue. In an aspect, the expanded population of ⁇ T cells is at least 50% ⁇ 1 + cells. In an aspect, the expanded population of ⁇ T cells is at least 70% ⁇ 1 + cells. In an aspect, the expanded population of ⁇ T cells is at least 90%> ⁇ 1 + cells. In an aspect, the subject has cancer.
  • the cancer has high infiltration of ⁇ T cells.
  • the application further discloses a TIGIT inhibitor as disclosed in any of the aspects and embodiments described herein for use in any of the methods described herein of activating and/or de-repressing ⁇ 2 " gamma-delta T cells ( ⁇ T cells) in a mammalian subject.
  • the application discloses herein a method of treating cancers having high infiltration of ⁇ T cells, comprising administering at least one TIGIT inhibitor to a patient with a cancer having high infiltration of ⁇ T cells, in an amount sufficient to activate tissue-infiltrating ⁇ T cells in the patient.
  • the cancer-infiltrating ⁇ T cells are ⁇ 2 " cells.
  • the ⁇ 2 " cells are ⁇ 1 + or DN cells.
  • the at least one TIGIT inhibitor is selected from the group consisting of an antagonist of TIGIT activity, an antagonist of TIGIT interaction with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL2, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL3, an agent that inhibits and/or blocks the intracellular signaling mediated by PVR binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling mediated by PVRL2 binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling mediated by PVLR3 binding to TIGIT, and combinations thereof.
  • the TIGIT inhibitor is an antibody. In an aspect, the antibody binds specifically to TIGIT. In an aspect, the antibody binds to PVR to specifically inhibit PVR interaction with TIGIT. In an aspect, the antibody is a full length IgG antibody. In an aspect, the antibody is an antigen-binding antibody fragment. In an aspect, the antibody is a single domain antibody. In an aspect, the antibody is fully human. In an aspect, the antibody is humanized. In an aspect, the TIGIT inhibitor is a small molecule inhibitor. In an aspect, the methods further comprise the antecedent step of selecting for treatment a patient whose cancer shows high levels of ⁇ T cell infiltration.
  • the ⁇ T cell infiltration is detected by analysis of a tumor biopsy by a method comprising an assay selected from the group consisting of immunohistochemistry, polymerase chain reaction, in situ hybridization, and combinations thereof.
  • the ⁇ T cell infiltration comprises ⁇ 1 + or double negative (DN) cells.
  • at least one additional therapeutic agent is administered.
  • the at least one or more additional agents comprises an agonist of DNAM-1.
  • the at least one or more additional agents comprises an anti-cancer agent.
  • the anti-cancer agent is selected from the group consisting of radiation, a chemotherapeutic or growth inhibitory agent, a targeted therapeutic agent, a small molecule inhibitor, a T cell expressing a chimeric antigen receptor, an antibody or antigen-binding fragment thereof, an antibody-drug conjugate, an angiogenesis inhibitor, an antineoplastic agent, a cancer vaccine, an adjuvant, and combinations thereof.
  • the anti-cancer agent is a chemotherapeutic.
  • the anti-cancer agent is a small molecule inhibitor.
  • the anti-cancer agent is an antibody.
  • the TIGIT inhibitor is administered continuously. In an aspect, the TIGIT inhibitor is administered intermittently.
  • the anti-cancer agent is administered continuously. In an aspect, the anti-cancer agent is administered intermittently. In an aspect, wherein the TIGIT inhibitor is administered before the anti-cancer agent. In an aspect, the TIGIT inhibitor is administered simultaneous with the anti-cancer agent. In an aspect, the TIGIT inhibitor is administered after the anticancer agent.
  • the application further discloses a TIGIT inhibitor as disclosed in any of the aspects and embodiments described herein for use in any of the methods described herein of treating cancers having high infiltration of ⁇ T cells.
  • the application discloses herein a method of increasing the number of tissue infiltrating ⁇ T cells in a mammalian subject recipient, comprising obtaining ⁇ T cells from tissue comprising ⁇ T cells form a donor; expanding the ⁇ T cells ex vivo; transplanting the expanded ⁇ T cells into a recipient, and administering at least one TIGIT inhibitor to the recipient.
  • the obtained and/or expanded ⁇ T cells are ⁇ 2 ⁇ .
  • the ⁇ 2 " cells are ⁇ 1 + or DN.
  • the at least one TIGIT inhibitor is selected from the group consisting of an antagonist of TIGIT activity, an antagonist of TIGIT interaction with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL2, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL3, an agent that inhibits and/or blocks the intracellular signaling mediated by PVR binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling mediated by PVRL2 binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling mediated by PVLR3 binding to TIGIT, or combinations thereof.
  • the at least one TIGIT inhibitor is an antibody.
  • the antibody binds specifically to TIGIT.
  • the antibody binds to PVR.
  • the antibody is a full length IgG antibody.
  • the antibody is an antigen- binding antibody fragment.
  • the antibody is a single domain antibody.
  • the antibody is fully human.
  • the antibody is humanized.
  • the at least one TIGIT inhibitor comprises a small molecule inhibitor.
  • the ex vivo expanded ⁇ 2 " ⁇ T cells were obtained by culturing lymphocytes obtained from non- hematopoietic tissue of humans or non-human animals in the presence of interleukin-2 (IL-2) and/or interleukin-15 (IL-15), and not in direct contact with stromal or epithelial cells during culture.
  • the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of IL-2.
  • the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of interleukin-15 (IL-15).
  • the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of IL-2 and IL-15. In an aspect, the culturing step comprises culturing the lymphocytes in the absence of TCR activation or co-stimulation signals. In an aspect, the culturing step comprises culturing the lymphocytes in the absence of a T cell receptor pathway agonist. In an aspect, the culturing step comprises culturing the lymphocytes in the absence of stromal or epithelial cells. In an aspect, stromal or epithelial cells are removed prior to culture. In as aspect, the lymphocytes are cultured in the absence of fibroblasts.
  • the lymphocytes have been obtained from skin, the gastrointestinal tract (e.g. colon), mammary gland tissue, lung, liver, pancreas or prostate.
  • the ex vivo expanded V52 " ⁇ T cells were obtained by culturing lymphocytes obtained from non- hematopoietic tissue of humans or non-human animals in the presence of IL-2, IL-15, and a factor selected from the group consisting of IL-4, IL-21, IL-6, IL-7, IL-8, IL-9, IL-12, IL-18, IL-33, IGF-1, IL- ⁇ , human platelet lysate (HPL), and stromal cell-derived factor-1 (SDF-1) for at least 5 days to produce an expanded population of ⁇ T cells.
  • the expanded population of ⁇ T cells comprises at least 20-fold the number of ⁇ T cells as the ⁇ T cells obtained from a non-hematopoietic tissue. In an aspect, within 7 days of culture, the expanded population of ⁇ T cells comprises at least 2-fold the number of ⁇ T cells as the ⁇ T cells obtained from a non-hematopoietic tissue. In an aspect, the expanded population of ⁇ T cells is at least 50% ⁇ 1 + cells. In an aspect, the expanded population of ⁇ T cells is at least 70% ⁇ 1 + cells. In an aspect, the expanded population of ⁇ T cells is at least 90% ⁇ 1 + cells.
  • the administration of the at least one TIGIT inhibitor is performed prior to, during, after the transplantation, or combinations thereof.
  • at least one additional agent is administered to the recipient.
  • the at least one additional agent is an anti-cancer agent.
  • the anti-cancer agent is selected from the group consisting of radiation, a chemotherapeutic or growth inhibitory agent, a targeted therapeutic agent, a small molecule inhibitor, a T cell expressing a chimeric antigen receptor, an antibody or antigen-binding fragment thereof, an antibody-drug conjugate, an angiogenesis inhibitor, an antineoplastic agent, a cancer vaccine, an adjuvant, and combinations thereof.
  • the recipient has cancer.
  • the ⁇ T cells are obtained from an autologous donor.
  • the non-hematopoietic tissue is tumor tissue.
  • the non-hematopoietic tissue is skin tissue.
  • the method further comprises separating ⁇ 1 + ⁇ T cells or DN cells from the ⁇ T cells prior to transplantation.
  • at least one additional checkpoint inhibitor is administered.
  • the additional checkpoint inhibitor does not inhibit PD1.
  • the additional checkpoint inhibitor inhibits TIM-3.
  • the additional checkpoint inhibitor inhibits LAG-3.
  • the administration of the TIGIT inhibitor results in elevated release of cytokines from the ⁇ T cells, selected from the group consisting of IFN- ⁇ , TNF-a, interleukins, and combinations thereof.
  • the application further discloses expanded ⁇ T cells obtained from tissue comprising ⁇ T cells from a donor as disclosed in any of the aspects and embodiments described herein and at least one TIGIT inhibitor as disclosed in any of the aspects and embodiments described herein for use in any of the methods described herein of increasing the number of tissue infiltrating ⁇ T cells in a mammalian subject recipient.
  • described herein are methods of treating cancer, comprising administering a therapeutically effective amount of a TIGIT inhibitor to a cancer patient who has been determined to have an elevated representation of ⁇ 1 + cells in a sample of peripheral blood.
  • the methods further comprise the prior step of determining the representation of ⁇ 1 + cells in the peripheral blood sample.
  • the methods further comprise the step, after determining the representation of ⁇ 1 + cells and before administering the TIGIT inhibitor, of selecting the patient for TIGIT inhibitor treatment if the patient's sample has been determined to have an elevated representation of ⁇ 1 + cells.
  • the elevated representation is an increase in ⁇ 1 + cells as a percentage of total lymphocytes in the sample.
  • greater than 0.5% of total lymphocytes in the sample are determined to be ⁇ 1 + cells. In certain aspects, greater than 1.0% of total lymphocytes are determined to be ⁇ 1 + cells. In certain aspects, greater than 2.0% of total lymphocytes are determined to be ⁇ 1 + cells. In certain aspects, wherein the elevated representation is an increase in ⁇ 1 + cells as a percentage of total ⁇ T cells in the sample. In certain aspects, greater than 10% of total ⁇ T cells in the sample are ⁇ 1 + cells. In certain aspects, greater than 15% of total ⁇ T cells in the sample are ⁇ 1 + cells. In certain aspects, the elevated representation is an increase in the ratio of ⁇ 1 + cells to ⁇ 2 + cells in the sample.
  • the ratio of ⁇ 1 + : ⁇ 2 + cells is greater than 1 :9. In certain aspects, the ratio of ⁇ 1 + : ⁇ 2 + cells is greater than 1 :8. In certain aspects, the ratio of ⁇ 1 + : ⁇ 2 + cells is greater than 1 :7. In certain aspects, the ratio of V51 + :V52 + cells is greater than 1 :6.
  • the TIGIT inhibitor is an antibody or antigen-binding antibody fragment that binds specifically to TIGIT.
  • the methods further comprise administration of at least one additional anti-cancer therapy. In certain aspects, at least one of the at least one additional anti-cancer therapy is administration of a non-TIGIT checkpoint inhibitor.
  • the non-TIGIT checkpoint inhibitor is an antibody that binds specifically to PD-1. In certain aspects, the non-TIGIT checkpoint inhibitor is an antibody that binds specifically to PDL1.
  • the cancer is a solid, non-hematopoietic, cancer. In certain aspects, the cancer is a breast cancer. In certain aspects, the cancer is a hematological cancer.
  • the application further discloses a TIGIT inhibitor as disclosed in any of the aspects and embodiments described herein for use in any of the methods described herein of treating cancer in a patient who has been determined to have an elevated representation of V51 + cells in a sample of peripheral blood.
  • Figure 1A provides a conceptual schema of sub-populations of T cells.
  • Figure IB shows results of flow cytometry performed on human peripheral blood mononuclear cells ("human blood”) and human skin-derived lymphocytes (“human tissue”) stained with antibodies specific for CD3, V51, and V52.
  • FIG. 1 illustrates PVR expression on Antigen Presenting Cells (APC) and antagonistic action on T Cells through DNAM1 and TIGIT.
  • APC Antigen Presenting Cells
  • FIG. 3 shows results of flow cytometry ("FACS") analysis for DNAM 1 expression of ⁇ T cells derived from human peripheral blood mononuclear cells (“PBMCs”) and human skin lymphocytes.
  • the shaded histogram is the signal of the Fluorescence minus one (FMO) control.
  • Percent of Max refers to the percentage of maximum count.
  • Figure 4 shows results of FACS analysis for TIGIT expression of ⁇ T cells derived from human PBMCs, both unstimulated (left panel) and stimulated for 72 hours with PHA.
  • Figure 5 shows results of FACS analysis for TIGIT expression on ⁇ T cells derived from human skin.
  • Figure 6 shows results of F ACS analysis for DNAM 1 and TIGIT expression on V51 + , V52 + , and DN ⁇ T cells derived from a representative donor of human PBMCs from peripheral blood (PB).
  • PB peripheral blood
  • FIG. 7 shows results of FACS analysis for DNAM 1 and TIGIT expression on V5T
  • V52 + , and DN ⁇ T cells derived from multiple donors of human PBMCs from PB are also known.
  • Figure 8 shows results of FACS analysis for DNAM 1 and TIGIT expression on ⁇ and DN ⁇ T cells derived from a representative donor of human skin leukocytes.
  • Figure 9 shows results of FACS analysis for DNAM 1 and TIGIT expression on V5T and DN ⁇ T cells derived from multiple donors of human skin leukocytes.
  • Figure 10 shows interferon gamma (IFNy) production in anti-CD3 stimulated V51 + .
  • PVR Fc Chimera Protein or an isotype control antibody (iso), demonstrating that PVR inhibits TCR stimulation in DN/ ⁇ T cells from human PBMCs.
  • FIG 11 shows results from FACS analysis of Interferon gamma (IFNy) and Tumor Necrosis Factor alpha (TNFa) production in anti-CD3 stimulated V51 + , V52 + , and DN ⁇ T cells derived from human PBMCs and human skin leukocytes upon exposure to PVR or an isotype control antibody (iso), demonstrating that PVR inhibits TCR stimulation in DN/V51 + T cells.
  • n 6 peripheral blood-derived ⁇ cell samples from different donors and 8 skin-derived ⁇ cell samples (2 out of 8 are sorted ⁇ cells).
  • n 3 peripheral blood-derived ⁇ cell samples and 1 skin-derived ⁇ cell samples.
  • the graphs for ⁇ 1 + and DN cell samples are generated from exactly all the same donors, while the graphs for V ⁇ 2 + are generated with a different set of donors.
  • the donors used for the V ⁇ 2 + graphs are all included among the donors for V ⁇ 1 + and DN samples.
  • Figure 12 shows results of FACS analysis for proliferation indicator/dye retention of anti-CD3 stimulated ⁇ 1 + and DN ⁇ T cells derived from human skin leukocytes upon exposure to PVR or an isotype control antibody, demonstrating the PVR inhibits TCR- induced proliferation in DN/V01 + T cells from human skin.
  • Figure 13 shows quantitation of results from FACS analysis for proliferation indicator/dye retention of anti-CD3 stimulated ⁇ 1 + and DN ⁇ T cells derived from human skin leukocytes upon exposure to PVR or an isotype control antibody, demonstrating that PVR inhibits TCR-induced proliferation in DN/V01 + T cells from human skin.
  • Figure 14 shows quantification of results from FACS analysis of Interferon gamma (IFNy) and Tumor Necrosis Factor alpha (TNFa) production in anti-CD3 stimulated V51 + and DN ⁇ T cells derived from human skin leukocytes upon exposure to an IgG control (mlgG), anti-CD3 (aCD3), and anti-CD3 plus PVR, demonstrating that PVR specifically inhibits TCR signaling.
  • IFNy Interferon gamma
  • TNFa Tumor Necrosis Factor alpha
  • Figure 15 shows quantification of results from FACS analysis of Interferon gamma (IFNy) production in anti-CD3 stimulated V51 + and DN ⁇ T cells derived from human skin leukocytes upon exposure to PVR and/or MICA, anti-DNAMl or an IgG control.
  • IFNy Interferon gamma
  • Figure 16 shows quantification of results from FACS analysis of Tumor Necrosis Factor alpha (TNFa) production in anti-CD3 stimulated V51 + and DN ⁇ T cells derived from human skin leukocytes upon exposure to PVR and/or MICA, anti-DNAMl or an IgG control, demonstrating that PVR specifically inhibits TCR signaling.
  • TNFa Tumor Necrosis Factor alpha
  • FIG. 17 shows quantification of results from FACS analysis of granulocyte- macrophage colony-stimulating factor (GMCSF), Interferon gamma (IFNy) and Tumor Necrosis Factor alpha (TNFa) production of ⁇ T cells from human skin lymphocytes of 5 different donors sorted with a pan ⁇ T cell antibody upon exposure to an IgG control (mlgG), anti-CD3 (aCD3), and anti-CD3 plus PVR, demonstrating that PVR is a general inhibitor of TCR stimulation.
  • GMCSF granulocyte- macrophage colony-stimulating factor
  • IFNy Interferon gamma
  • TNFa Tumor Necrosis Factor alpha
  • Figure 18 shows quantification of results from FACS analysis for TIGIT expression of tissue-derived V51 + and V53 + cells cultured in the presence of IL-2 and IL-15 (culture method 1), or in the presence of IL-2, IL-4, IL-15 and IL-21 (culture method 2), demonstrating that cells grown using culture method 2 express low levels of TIGIT.
  • Figure 19 are graphs showing quantification of results from FACS analysis of tissue- derived V51 + and V53 + cells from an individual donor cultured in the presence of IL-2, IL-4, IL-15 and IL-21 upon exposure to an IgG control (mlgG), anti-CD3 (aCD3), and anti-CD3 plus PVR, demonstrating that the PVR inhibition of INFy and TNFa was lost in V51 + and V53 + cells expressing low levels of TIGIT.
  • IgG control mlgG
  • aCD3 anti-CD3 plus PVR
  • Figure 20 are graphs showing quantification of results from FACS analysis of tissue- derived V51 + and V53 + cells from an individual donor cultured in the presence of IL-2, IL-4, IL-15 and IL-21 upon exposure to an IgG control (mlgG), anti-CD3 (aCD3), and anti-CD3 plus PVR, demonstrating that the PVR inhibition of INFy and TNFa was lost in V51 + and V53 + cells expressing low levels of TIGIT. 6.
  • IgG control mlgG
  • aCD3 anti-CD3 plus PVR
  • tissue-infiltrating ⁇ T cells particularly V52 " ⁇ T cells
  • methods for activating tissue-infiltrating ⁇ T cells, particularly V52 " ⁇ T cells in mammalian subjects, by antagonizing TIGIT activity of the V52 " ⁇ T cells, and methods for the treatment of cancer comprising administration of at least one TIGIT inhibitor.
  • V62 ⁇ T cells refers to any ⁇ T cell that does not express V52.
  • DN Double negative
  • DN cells can express, but are not limited to those that express, V53, V54, V55, V56, V57, and
  • activate a T cell includes activation, de-repression, and both activation and de-repression of the T cell.
  • TIGIT inhibitor refers to any agent or compound capable of disrupting TIGIT activity or disrupting TIGIT interaction with PVR.
  • a “TIGIT inhibitor” can increase PVR interaction with DNAMl .
  • a “TIGIT inhibitor” may block the interaction of PVR with TIGIT, leading to sequestration of TIGIT and increased binding of PVR to DNAMl.
  • a “TIGIT inhibitor” can be an antagonist of TIGIT activity, an antagonist of TIGIT interaction with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL2, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL3, an agent that inhibits and/or blocks the intracellular signaling within a V52 " ⁇ T cell mediated by PVR binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling within a V52 " ⁇ T cell mediated by PVRL2 binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling within a V52 " ⁇ T cell mediated by PVLR3 binding to TIGIT, and combinations thereof.
  • an “expanded population of ⁇ cells” refers to a population of cells that has been cultured under conditions and for a duration sufficient to cause proliferation of ⁇ cells within that population.
  • an “expanded population of V61 + T cells,” as used herein, refers to a population of cells that has been cultured under conditions and for a duration sufficient to cause proliferation of V51 + T cells within that population.
  • tissue-resident refers to cells that are located at non-vascular anatomical sites.
  • Antigen binding fragment of an antibody or “antigen binding antibody fragment” refers to any single chain or multiple chain portion of an antibody that is capable of binding specifically to an epitope of an antigen.
  • Antigen binding fragment includes, without limitation, Fab, F(ab)2, and single-chain Fv (scFv).
  • Human antibody unless otherwise indicated is one whose sequences correspond to (i.e., are identical in sequence to) an antibody that could be produced by a human and/or consists entirely of amino acid sequences that are encoded in the human genome.
  • a "human antibody” as used herein can be produced using various techniques known in the art, including phage-display libraries and by administering the antigen (e.g., TIGIT) to a transgenic animal that has been modified to produce such antibodies in response to antigenic challenge, but whose endogenous loci have been disabled, e.g., immunized Xenomice.
  • Humanized antibodies refer to non-human (e.g., murine) antibodies that are chimeric antibodies and contain minimal sequence derived from non-human immunoglobulin.
  • a humanized antibody is a human immunoglobulin (recipient antibody) in which residues from a hypervariable region (HVR) of the recipient are replaced by residues from a HVR of a non-human species (donor antibody) such as mouse, rat, rabbit, or nonhuman primate having the desired specificity, affinity, and/or capacity.
  • donor antibody such as mouse, rat, rabbit, or nonhuman primate having the desired specificity, affinity, and/or capacity.
  • FR residues of the human immunoglobulin variable domain are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. In an embodiment, the humanized antibodies do not comprise residues that are not found in the recipient antibody or in the donor antibody.
  • a 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 FRs are those of a human immunoglobulin sequence.
  • the humanized antibody optionally will also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. In an embodiment where the humanized antibodies do comprise residues that are not found in the recipient antibody or in the donor antibody, the Fc regions of the antibodies are modified.
  • Binds specifically refers to the property of an antibody or fragment of binding to the (specified) antigen with a dissociation constant that is ⁇ 1 ⁇ , preferably ⁇ 1 nM and most preferably ⁇ 10 pM.
  • the Kd of the antibody that binds specifically to TIGIT is 250-500 pM.
  • An epitope that "binds specifically "or "binds preferentially” (used
  • an antibody or a polypeptide is a term well understood in the art, and methods to determine such specific or preferential binding are also well known in the art.
  • a molecular entity is said to exhibit "specific binding” or “preferential binding” if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular cell or substance than it does with alternative cells or substances.
  • An antibody "specifically binds” or “preferentially binds" to a target if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other substances.
  • an antibody that specifically or preferentially binds to a TIGIT conformational epitope is an antibody that binds this epitope with greater affinity, avidity, more readily, and/or with greater duration than it binds to other TIGIT epitopes or non-TIGIT epitopes. It is also understood by reading this definition that, for example, an antibody (or moiety or epitope) that specifically or preferentially binds to a first target may or may not specifically or preferentially bind to a second target. As such, “specific binding” or “preferential binding” does not necessarily require (although it can include) exclusive binding.
  • cancers or tissues with "high ⁇ T cell infiltration” refers to tissues (e.g., tumor from tumor biopsies) wherein the fraction of ⁇ T cells to total leukocytes, either measured or estimated, is greater than the fraction of ⁇ T cells in a given reference tissue, such as non-tumor tissue of a similar origin.
  • tissues with "high ⁇ T cell infiltration” refers to tissues wherein the fraction of ⁇ T cells to total leukocytes is greater than 0.0001 , greater than 0.001 , greater than 0.01 , greater than 0.1 , greater than 0.2, greater than 0.3, greater than 0.4, or greater than 0.5.
  • tissues with "high ⁇ T cell infiltration” refers to tissues in which the fraction of ⁇ T R A to total leukocyte R A is greater than 0.0001, greater than 0.001, greater than 0.01 , or greater than 0.1.
  • T-cell checkpoints or “checkpoints” refers to any protein or gene that functions to inhibit or suppress the activation of a T-cell.
  • T-cell checkpoints include, but are not limited to: A2A , B7-H3, B7-H4, BTLA, CTLA-4, IDO, KIR, LAG3, PD-1, TIM-3 and VISTA.
  • T-cell checkpoint inhibitor or "checkpoint inhibitor” refers to any molecule that inhibits the activity of a T-cell checkpoint.
  • “Sufficient amount” means an amount sufficient to produce a desired effect, e.g., an amount sufficient to modulate ⁇ T cell activity.
  • Therapeutically effective amount is an amount that is effective to ameliorate a symptom of a disease.
  • a therapeutically effective amount can be a “prophylactically effective amount” as prophylaxis can be considered therapy.
  • adjunctive administration or “administered to the subject adjunctively” are used interchangeably herein to mean administering a second therapeutic agent in therapeutically effective temporal proximity to a first therapeutic agent.
  • adjunctive administration improves the effectiveness of the second therapeutic agent, the first therapeutic agent, or both the first and the second therapeutic agent as compared to administration of the agent alone.
  • ⁇ T cells refers to gamma delta T cells
  • TGIT refers to T-Cell-IG and ITIM domain
  • TCR refers to the T-cell Receptor
  • PVR refers to Poliovirus receptor
  • IL-2 refers to native or recombinant inter leukin 2
  • IL-15 refers to native or recombinant interleukin 15, etc.
  • PB refers to peripheral blood and "PBMC” refers to peripheral blood mononuclear cells.
  • antagonizing TIGIT activity comprises administering at least one TIGIT inhibitor to the subject. TIGIT inhibitors suitable for use in the methods are described below in Section 6.6.
  • the at least one TIGIT inhibitor is administered to the subject adjunctively with V52 " ⁇ T cells that have been cultured ex vivo.
  • the at least one TIGIT inhibitor is administered in a
  • composition that also comprises the cultured ⁇ T cells.
  • the at least one TIGIT inhibitor is added to the V52 " ⁇ T cells during culturing ex vivo. In certain embodiments, the at least one TIGIT inhibitor is added to the V52 " ⁇ T cells after culturing ex vivo.
  • the at least one TIGIT inhibitor is administered to the subject separately from, but adjunctively with, the ex vivo expanded ⁇ T cells.
  • ⁇ 2 + T cells predominate among the ⁇ cells in hematopoietic tissues
  • ⁇ 2 " cells are also present, albeit in lower numbers.
  • the ⁇ T cells are obtained for culture from hematopoietic tissues, such as peripheral blood.
  • ⁇ 2 " ⁇ T cells are obtained for culture from the hematopoietic tissues.
  • ⁇ T cells from hematopoietic tissues are stimulated to reduce ⁇ 2 expression and/or induce ⁇ expression and/or selectively increase proliferation of ⁇ 2 " cells.
  • the ⁇ T cells are stimulated according to a protocol described in WO 2012/156,958, the contents of which are incorporated herein by reference in their entirety.
  • ⁇ T cells obtained from hematopoietic tissues are stimulated with a ⁇ TCR antibody to expand ⁇ 2 " ⁇ T cells and/or ⁇ 1 + ⁇ cells ex-vivo.
  • ⁇ T cells obtained from hematopoietic tissues are stimulated with a ⁇ TCR antibody to increase TIGIT expression of the ⁇ T cells ex-vivo.
  • ⁇ T cells are obtained for culture from non-hematopoietic tissues.
  • Detailed methods for the isolation, culturing and expansion of ⁇ T cells from non- hematopoietic tissues are disclosed in WO 2017/072367, which is incorporated herein by reference in its entirety.
  • V52 " ⁇ T cells for culture are obtained from any human or non-human mammal non-hematopoietic tissue that can be removed from a subject.
  • the non-hematopoietic tissue from which the V52 " ⁇ T cells are obtained for culture is skin (e.g., human skin), which can be obtained by methods known in the art.
  • the skin is obtained by punch biopsy.
  • the V52 " ⁇ T cells are obtained for culture from the gastrointestinal tract (e.g., colon), mammary gland, lung, prostate, liver, spleen, and pancreas.
  • the V52 " ⁇ T cells are obtained for culture from non-neoplastic tissues.
  • the V52 " ⁇ T cells are obtained for culture from human cancer tissues, e.g., tumors of the breast, the prostate, or other solid tumors.
  • the ⁇ T cells that are dominant in the non-hematopoietic tissues are primarily V51 + T cells, such that V51 + T cells comprise about 70-80% of the non-hematopoietic tissue-resident ⁇ T cell population.
  • the V52 " ⁇ T cells that have been cultured ex vivo are V51 + .
  • V52 T cells that are resident in non-hematopoietic tissues express neither V51 nor V52 TCR and we have named them double negative (DN) ⁇ T cells.
  • V52 " ⁇ T cells that have been cultured ex vivo are V51 " V52 " double negative (DN) cells.
  • DN ⁇ T cells are likely to be mostly V53 -expressing with a minority of V55 -expressing T cells.
  • the cultured V52 " ⁇ T cells have been expanded ex vivo.
  • the ex vivo expanded V52 " ⁇ T cells are obtained by culturing lymphocytes obtained from non-hematopoietic tissue of humans or non-human animals in the presence of inter leukin-2 (IL-2) and/or interleukin-15 (IL-15), and not in direct contact with stromal or epithelial cells during culture.
  • IL-2 inter leukin-2
  • IL-15 interleukin-15
  • the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of IL-2. In certain embodiments, the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of interleukin-15 (IL- 15). In particular embodiments, the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of IL-2 and IL-15.
  • IL-2 interleukin-15
  • the culturing step comprises culturing the lymphocytes in the absence of TCR activation or co-stimulation signals. In some embodiments, the culturing step comprises culturing the lymphocytes in the absence of a T cell receptor pathway agonist. In some embodiments, the culturing step comprises culturing the lymphocytes in the absence of stromal or epithelial cells. In some embodiments, stromal or epithelial cells are removed prior to culture. In some embodiments, the lymphocytes are cultured in the absence of fibroblasts. In various embodiments, the lymphocytes have been obtained from skin, the gastrointestinal tract (e.g., colon), mammary gland tissue, lung, liver, pancreas or prostate.
  • the gastrointestinal tract e.g., colon
  • mammary gland tissue e.g., lung, liver, pancreas or prostate.
  • the ex vivo expanded V52 " ⁇ T cells are obtained by culturing lymphocytes obtained from non-hematopoietic tissue of humans or non-human animals in the presence of IL-2, IL-15, and a factor selected from the group consisting of IL-4, IL-21, IL-6, IL-7, IL-8, IL-9, IL-12, IL-18, IL-33, IGF-1, IL- ⁇ , human platelet lysate (HPL), and stromal cell-derived factor- 1 (SDF-1) for at least 5 days to produce an expanded population of ⁇ T cells.
  • IL-2 lymphocytes obtained from non-hematopoietic tissue of humans or non-human animals in the presence of IL-2, IL-15, and a factor selected from the group consisting of IL-4, IL-21, IL-6, IL-7, IL-8, IL-9, IL-12, IL-18, IL-33, IGF-1, IL- ⁇ , human platelet lysate
  • the ex vivo expanded V52 " ⁇ T cells are obtained by culturing lymphocytes obtained from non-hematopoietic tissue of humans or non-human animals under conditions in which, within 14 days of culture, the expanded population of ⁇ T cells comprises at least 20-fold the number of ⁇ T cells as the ⁇ T cells obtained from a non- hematopoietic tissue.
  • the ex vivo expanded ⁇ 2 " ⁇ T cells were obtained by culturing lymphocytes obtained from non-hematopoietic tissue of humans or non- human animals under conditions in which, within 7 days of culture, the expanded population of ⁇ T cells comprises at least 2-fold the number of ⁇ T cells as the ⁇ T cells obtained from a non-hematopoietic tissue.
  • the expanded population of ⁇ T cells is at least 50% ⁇ 1 + cells. In certain embodiments, the expanded population of ⁇ T cells is at least 70% ⁇ 1 + cells. In select embodiments, the expanded population of ⁇ T cells is at least 90%> ⁇ 1 + cells.
  • the method further comprises expanding non-hematopoietic tissue-resident ⁇ T cells (e.g., skin-derived ⁇ T cells and/or ⁇ - ⁇ 2 T cells, e.g., ⁇ T cells and/or DN T cells).
  • the non-hematopoietic tissue-resident ⁇ T cells are expanded from a population of ⁇ T cells that has been separated from non- hematopoietic tissue according to methods described above.
  • non-hematopoietic tissue-resident ⁇ T cells are capable of spontaneously expanding upon removal of physical contact with stromal cells (e.g., skin fibroblasts).
  • the scaffold-based culture methods described above can be used to induce such separation, resulting in de-repression of the ⁇ T cells to trigger expansion. Accordingly, in some embodiments, no substantial TCR pathway activation is present during the expansion step (e.g., no exogenous TCR pathway activators are included in the culture). In various embodiments, the methods of expanding non- hematopoietic tissue-resident ⁇ T cells omit contact with feeder cells, tumor cells, and/or antigen-presenting cells.
  • the amount of each of the above cytokines required to produce an expanded population of ⁇ T cells will depend of the concentrations of one or more of the other cytokines. For example, if the concentration of IL-2 is increased or decreased, the concentration of IL-15 may be accordingly decreased or increased,
  • the amount effective to produce an expanded population refers herein to composite effect of all factors on cell expansion.
  • the ⁇ T cells are exposed to certain factors prior to culture with other factors.
  • the expansion culture can be gradually supplied with additional factors over the course of expansion, or, alternatively, the ⁇ T cells can be transferred from a culture of one factor or group of factors to another.
  • the ⁇ T cells are expanded in culture for a period of several hours (e.g., about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 18, or 21 hours) to about 35 days (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, or 35 days).
  • the ⁇ T cells are expanded for a period of 14 to 21 days.
  • the separation and expansion steps in some embodiments, can last between 28 and 56 days, or about 41 days.
  • Methods of expansion provide an expanded population of ⁇ T cells that is greater in number than a reference population.
  • the expanded population of ⁇ T cells is greater in number than the separated population of ⁇ T cells prior to the expansion step (e.g., at least 2-fold in number, at least 3-fold in number, at least 4-fold in number, at least 5-fold in number, at least 6-fold in number, at least 7-fold in number, at least 8-fold in number, at least 9-fold in number, at least 10-fold in number, at least 15-fold in number, at least 20-fold in number, at least 25-fold in number, at least 30-fold 35 in number, at least 35-fold in number, at least 40-fold in number, at least 50-fold in number, at least 60- fold in number, at least 70- fold in number, at least 80-fold in number, at least 90-fold in number, at least 100-fold in number, at least 200-fold in number, at least 300-fold in number, at least 400-fold in number, at least 500-
  • the invention provides a means to produce large populations of non-hematopoietic tissue-derived ⁇ T cells (e.g., skin-derived ⁇ T cells and/or ⁇ - ⁇ 2 T cells, e.g., ⁇ T cells and/or DN T cells) at high rates (e.g., by removing stromal cell contact and/or TCR stimulation, or by culturing in the presence of an effective amount of factors).
  • the expansion step described herein expands the ⁇ T cells at a low population doubling time, which is given by the following equation:
  • non-hematopoietic tissue-derived ⁇ T cells e.g., skin-derived ⁇ T cells and/or ⁇ - ⁇ 2 T cells, e.g., ⁇ T cells and/or DN T cells
  • a population doubling time of less than 5 days e.g., less than 4.5 days, less than 4.0 days, less than 3.9 days, less than 3.8 days, less 15 than 3.7 days, less than 3.6 days, less than 3.5 days, less than 3.4 days, less than 3.3 days, less than 3.2 days, less than 3.1 days, less than 3.0 days, less than 2.9 days, less than 2.8 days, less than 2.7 days, less than 2.6 days, less than 2.5 days, less than 2.4 days, less than 2.3 days, less than 2.2 days, less than 2.1 days, less than 2.0 days, less than 46 hours, less than 42 hours, less than 38 hours, less than 35 hours, less than 32
  • the expanded population of ⁇ T cells comprises at least 10-fold the number of ⁇ T cells relative to the separated population of ⁇ T-cells prior to expansion (e.g., at least 20-fold, at least 30-fold, at least 40-fold, at least 50-fold, at least 60-fold, at least 70-fold, at least 80-fold, at least 90-fold, at least 100-fold, at least 150-fold, at least 200-fold, at least 300-fold, at least 400-fold, at least 500-fold, at least 600-fold, at least 700-fold, at least 800-fold, at least 900-fold, at least 1,000-fold, at least 2,000-fold, at least 3,000-fold, at least 4,000-fold, at least 5,000-fold, at least 6,000-fold, at least 7,000-fold, or at least 8,000- fold the number of ⁇ T cells relative to the separated population of ⁇ T cells prior to
  • the expanded population of ⁇ T cells comprises at least 20-fold the number of ⁇ T cells relative to the separated population of ⁇ T cells prior to expansion (e.g., at least 30-fold, at least 40-fold, at least 50-fold, at least 60-fold, at least 70-fold, at least 80-fold, at least 90-fold, at least 100-fold, at least 150-fold, at least 200-fold, at least 300-fold, at least 400-fold, at least 500-fold, at least 600-fold, at least 700-fold, at least 800-fold, at least 900-fold, at least 1,000-fold, at least 2,000-fold, at least 3,000-fold, at least 4,000-fold, at least 5,000-fold, at least 6,000-fold, at least 7,000-fold, at least 8,000-fold, at least 9,000- fold, or at least 10,000-fold the number of ⁇ T cells relative to
  • the expanded population of ⁇ T cells comprises at least 50-fold the number of ⁇ T cells relative to the separated population of ⁇ T cells prior to expansion (e.g., at least 60- fold, at least 70-fold, at least 80-fold, at least 90- fold, at least 100-fold, at least 150-fold, at least 200-fold, at least 300-fold, at least 400-fold, at least 500-fold, at least 600-fold, at least 700-fold, at least 800-fold, at least 900-fold, at least 1,000-fold, at least 2,000-fold, at least 3,000-fold, at least 4,000-fold, at least 5,000-fold, at least 6,000-fold, at least 7,000-fold, at least 8,000-fold, at least 9,000-fold, or least 10,000- fold the number of ⁇ T cells relative to the separated population of ⁇ T cells prior to expansion).
  • at least 50-fold the number of ⁇ T cells relative to the separated population of ⁇ T cells prior to expansion e.g., at least 60- fold, at least 70
  • the expanded population of ⁇ T cells comprises at least 100-fold the number of ⁇ T cells relative to the separated population of ⁇ T cells prior to expansion (e.g., at least 110-fold, at least 120-fold, at least 130-fold, at least 140-fold, at least 150-fold, at least 200-fold, at least 300-fold, at least 400-fold, at least 500-fold, at least 600-fold, at least 700-fold, at least 800-fold, at least 900-fold, at least 1,000-fold, at least 2,000-fold, at least 3,000-fold, at least 4,000-fold, at least 5,000-fold, at least 6,000-fold, at least 7,000-fold, at least 8,000-fold, at least 9,000-fold, at least 10,000-fold, at least 12,000-fold, or at least 15,000-fold the number of ⁇ T cells relative to the separated population of ⁇ T cells prior
  • Non-hematopoietic tissue-derived ⁇ T cells expanded by the methods provided herein can have a phenotype well-suited for anti-tumor efficacy.
  • the expanded population of ⁇ T cells e.g., skin-derived ⁇ T cells
  • the expanded population of ⁇ T cells has a mean expression of CD27 that is at least 2-fold relative to the separated population of ⁇ T cells (e.g., at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8- fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 40-fold, at least 50-fold, at least 60-fold, at least 70-fold, at least 80-fold, at least 90-fold, at least 100-fold, at least 150-fold, at least 200-fold, at least 300-fold, at least 400-fold, at least 500-fold, at least 600-fold, at least 700-fold, at least 800-fold, at least 900- fold, at least 1,000-fold, at least 5,000-fold, at least 10,000-fold, at least 20,000-fold, or more, relative to the separated population of ⁇ T cells).
  • An increase or decrease in expression of other markers can be additionally or alternatively used to characterize one or more expanded populations of non-hematopoietic tissue-derived ⁇ T cells (e.g., skin-derived ⁇ T cells and/or ⁇ - ⁇ 2 T cells, e.g., ⁇ T cells and/or DN T cells), including CD124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1, CD31, KLRG1,CD30, CD2, NKp44, NKp46, ICAM- 2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1, and CD64.
  • non-hematopoietic tissue-derived ⁇ T cells e.g., skin-derived ⁇ T cells and/or ⁇ - ⁇ 2 T cells, e.g., ⁇ T cells and/or DN T cells
  • the expanded population of ⁇ T cells has a greater mean expression of one or more of the markers selected from the group consisting of CD124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1, CD31 , KLRG1, CD30, and CD2, relative to the separated population of ⁇ T cells, e.g., prior to expansion.
  • the expanded population of ⁇ T cells may have a greater frequency of cells expressing one or more of the markers selected from the group consisting of CD 124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1, CD31, KLRG1, CD30, and CD2, relative to the separated population of ⁇ T cells.
  • the expanded population of ⁇ T cells (e.g., skin-derived ⁇ T cells and/or non- V52 T cells, e.g., V51 T cells and/or DN T cells) has a lower mean expression of one or more of the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1, and CD64, relative to the separated population of ⁇ T cells.
  • the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1, and CD64 relative to the separated population of ⁇ T cells.
  • the expanded population may similarly have a lower frequency of cells expressing one or more of the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1, and CD64, relative to the separated population of ⁇ T cells.
  • a non-hematopoietic tissue-resident ⁇ T cell produced by the method of the invention may thus have one or more of the following properties: (i) displays the phenotype CD69high, TIM3high and CD281ow/absent; (ii) upregulates of one or more of CCR3, CD39, CDl lb, and CD9; (iii) produces IFN- ⁇ in response to an NKG2D ligand in the absence of TCR agonists; (iv) produces IL-13 in the absence of TCR agonists; (v) produces one or more of IFN- ⁇ , TNF- ⁇ and GM-CSF in response to TCR activation; (vi) produces no or substantially no IL-17 in response to TCR activation; (vii) grows in culture medium containing IL-2 without additional growth factors; (viii) displays a cytotoxic T cell response in the absence of TCR agonists; and/or (ix) displays selective cytotoxicity for tumor cells over normal cells.
  • a non-hematopoietic tissue-resident ⁇ T cell produced by the method of the invention produces IL-13 in the absence of TCR agonists and/or produces IFN- ⁇ in response to an NKG2D ligand in the absence of TCR agonists.
  • basal culture media suitable for use in the proliferation of ⁇ T cells are available, in particular complete media, such as AIM-V, Iscoves medium and RPMI-1640 (Life Technologies).
  • the medium may be supplemented with other media factors, such as serum, serum proteins and selective agents, such as antibiotics.
  • RPMI-1640 medium containing 2 mM glutamine, 10% FBS, 10 mM HEPES, pH 7.2, 1% penicillin-streptomycin, sodium pyruvate (1 mM; Life Technologies), nonessential amino acids (e.g.
  • cells are cultured at 37°C in a humidified atmosphere containing 5% C02 in a suitable culture medium.
  • the ⁇ T cells may be cultured as described herein in any suitable system, including stirred tank fermenters, airlift fermenters, roller bottles, culture bags or dishes, and other bioreactors, in particular hollow fiber bioreactors. The use of such systems is well-known in the art. General methods and techniques for culture of lymphocytes are well-known in the art.
  • the methods described herein can include more than one selection step, e.g., more than one depletion step.
  • Enrichment of a T cell population by negative selection can be accomplished, e.g., with a combination of antibodies directed to surface markers unique to the negatively selected cells.
  • One method is cell sorting and/or selection via negative magnetic immuno adherence or flow cytometry that uses a cocktail of monoclonal antibodies directed to cell surface markers present on the cells negatively selected.
  • the ACT methods comprise, T cell immunotherapy wherein subjects are infused with heterologous, autologous or allogeneic ⁇ T cells to treat disease.
  • the ACT methods comprise infusion of subjects of V52 " ⁇ T cells to treat disease.
  • the ACT methods comprise infusion of subjects of V52 " ⁇ T cells that have been expanded ex vivo to treat disease.
  • the ACT methods comprise infusion of subjects of V52 " ⁇ T cells that have been expanded ex vivo to treat cancer.
  • V52 " ⁇ T cells are selected and expanded ex vivo to enrich for V52 " ⁇ T cells from a mixed population of immune cells.
  • ⁇ T cells are selected and/or engineered ex vivo to target specific antigens, such as tumor-associated antigens.
  • immune cells comprising ⁇ T cells are obtained from tissue of donor subjects.
  • ⁇ T cells are obtained from non-hematopoietic tissue of donor subjects.
  • ⁇ T cells are obtained from blood of donor subjects.
  • tumor infiltrating lymphocytes (“TIL"s) comprising ⁇ T cells are obtained from donor subjects.
  • TIL tumor infiltrating lymphocytes
  • ⁇ T cells are expanded in culture and selected for antigen specificity without altering their native specificity.
  • T lymphocytes comprising ⁇ T cells obtained from the donor are engineered ex vivo by transduction with viral expression vectors, to express chimeric antigen receptors ("CAR"s) of predetermined specificity.
  • CAR chimeric antigen receptors
  • Concentrations of cytokines and growth factors can range from about 0.1 ng/mL to about 500 ng/mL, from about 1 ng/mL to about 200 ng/mL, from about 10 ng/ml to 100 ng/ml, depending on the type of cytokine and growth factor. Appropriate combinations and concentrations of cytokines are disclosed in WO 2017/072367, which is incorporated herein by reference in its entirety.
  • the methods further comprise administering to the subject at least one additional agent in an amount sufficient to activate V52 " ⁇ T cells.
  • the at least one additional agent is an agonist of DNAM-1.
  • the mammalian subject has cancer.
  • the mammalian subject has cancer.
  • the cancer has high infiltration of ⁇ T cells.
  • the mammalian subject is a human patient.
  • the subject from whom ⁇ 2 " T cells are obtained for culture is the same individual as the ultimate recipient of the cells, i.e., an autologous donor.
  • Autologous ⁇ T cells which are removed and in some instances stored are transplanted back into the subject.
  • the autologous ⁇ T cells are subject to additional treatments prior to transplantation back into the recipient.
  • autologous ⁇ T cells obtained from the subject are further treated or purified to remove any disease cells, such as cancer cells, prior to transplantation back into the subject.
  • treatments and purification also referred to as "purging," of cell preparations to remove disease cell can include, among others, use of antibodies directed against cell surface markers expressed in disease cells and anti-cancer (i.e., chemotherapeutic) treatments.
  • the autologous ⁇ T cells are obtained following treatment of the subject for the underlying disease, thereby reducing the risk of presence of disease cells in the autologous ⁇ T cell preparations.
  • the subject can be treated with an antibody therapeutic targeting the disease or treated with anti-cancer agent that does not destroy or suppress the hematopoietic system prior to obtaining the ⁇ T cells for transplantation.
  • the purged preparation of ⁇ T cells can be further purified, such as by FACS or affinity selection (e.g., chromatography), to select ⁇ T cells or ⁇ 2 " ⁇ T cells from other cells (i.e., other immune cells or diseased cells).
  • the donor subject is not the same individual as the ultimate recipient of the cells, i.e., a heterologous or allogeneic donor.
  • the cells obtained from the donor subject are not screened or matched for the recipient.
  • ⁇ T cells are non-MHC restricted, they do not recognize a host into which they are transferred as foreign, which means that they are less likely to cause graft- versus-host disease. This means that they can be used "off the shelf and transferred into any recipient, e.g., for heterologous or allogeneic adoptive T cell therapy.
  • allogeneic immune cells comprising ⁇ T cells are obtained from donors who are selected based on matching at loci of the human lymphocyte antigen (HLA) complex.
  • the allogeneic immune cells comprising ⁇ T cells can be further isolated or purified, and/or subject to further manipulation.
  • the allogeneic immune cells comprising ⁇ T cells are subject to additional treatments to expand a population of immune cells or manipulated by recombinant methods to introduce heterologous genes or additional functionality to the allogeneic immune cells prior to transplantation into the recipient subject.
  • ⁇ T cells obtained from a donor can be subject to additional treatments prior to transplantation into a recipient subject.
  • the immune cells comprising ⁇ T cells are treated to expand the population of ⁇ T cells, for example by culturing in a suitable medium.
  • the method further comprises the step of detecting ⁇ T cell infiltration, or ⁇ 2 " ⁇ T cell infiltration, of tissues in a mammalian subject and selecting subjects who have ⁇ T cell infiltrates.
  • ⁇ T cell infiltration or ⁇ 2 " ⁇ T cell infiltration is detected in tumors of a mammalian subject. Detection of ⁇ T cell or ⁇ 2 " ⁇ T-cells may be performed by any method known in the art for the detection of subpopulations of immune cells in tissue. T cell infiltration in tissues ⁇ e.g., tumor from tumor biopsies) can be determined by
  • An increase or decrease in expression of other markers can be additionally or alternatively used to characterize ⁇ T cells from tissue or ⁇ T cells from tissue which have been expanded ex-vivo ⁇ e.g., skin-derived ⁇ T cells and/or ⁇ - ⁇ 2 T cells, e.g., ⁇ T cells and/or DN T cells), including CD124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1 , CD31 , KLRG1 , CD30, CD2, NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1 , and CD64.
  • ⁇ T cells from tissue or ⁇ T cells from tissue which have been expanded ex-vivo ⁇ e.g., skin-derived ⁇ T cells and/or ⁇ - ⁇ 2 T cells, e.g., ⁇ T cells and/or DN T cells
  • the expanded population of ⁇ T cells has a greater mean expression of one or more of the markers selected from the group consisting of CD124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1 , CD31 , KLRG1 , CD30, and CD2, relative to the separated population of ⁇ T cells, e.g., prior to expansion.
  • the expanded population of ⁇ T cells may have a greater frequency of cells expressing one or more of the markers selected from the group consisting of CD124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1 , CD31 , KLRG1 , CD30, and CD2, relative to the separated population of ⁇ T cells.
  • the expanded population of ⁇ T cells (e.g., skin-derived ⁇ T cells and/or non- ⁇ 2 T cells, e.g., ⁇ T cells and/or DN T cells) has a lower mean expression of one or more of the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1 , and CD64, relative to the separated population of ⁇ T cells.
  • the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1 , and CD64 relative to the separated population of ⁇ T cells.
  • the expanded population may similarly have a lower frequency of cells expressing one or more of the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1 , and CD64, relative to the separated population of ⁇ T cells.
  • the composition is co-administered with an additional agent for treating cancer (e.g., an anti-cancer agent).
  • an additional agent for treating cancer e.g., an anti-cancer agent.
  • the composition is coadministered with an as yet to be described therapeutic for treating cancer.
  • the agents may be administered concurrently or sequentially.
  • agents described herein are administered prior to the other active agent(s).
  • the pharmaceutical formulations and modes of administration may be any of, but not limited to, those described below. It is also anticipated that there could be two or more co-administered chemical agents, biological agents or radiation that may each be administered using different modes or different formulations. 6.3.1.4.2. Selection of subjects with high V62 " ⁇ T cells in peripheral blood
  • the method further comprises the step of detecting V52 " ⁇ in the blood of a mammalian subject, and selecting subjects who have high ⁇ 2 ⁇ ⁇ T cells in the blood.
  • the method comprises determining the ratio of ⁇ 1 + and ⁇ 2 + in a blood sample from a potential mammalian subject.
  • the method comprises determining the total number of ⁇ 1 + and ⁇ 2 + in a blood sample from a potential mammalian subject.
  • the method further comprises comparing the ratio of ⁇ 1 + and ⁇ 2 + in a blood sample from a subject with cancer to a healthy individual.
  • the method further comprises comparing the total number of ⁇ 1 + and ⁇ 2 + in a blood sample from a subject with cancer to a healthy individual.
  • the cancer has high infiltration of ⁇ T cells.
  • the methods comprise administering at least one TIGIT inhibitor to a patient with a cancer having high infiltration of ⁇ T cells, in an amount sufficient to activate tissue-infiltrating ⁇ T cells in the patient.
  • the cancer-infiltrating ⁇ T cells are ⁇ 2 " cells, and the amount of TIGIT inhibitor is sufficient to activate ⁇ 2 " cells.
  • the amount of TIGIT inhibitor is sufficient to activate ⁇ 1 + cells.
  • the amount of TIGIT inhibitor is sufficient to activate double negative (DN) cells.
  • the TIGIT inhibitor is an inhibitor as described in
  • the TIGIT inhibitor is an antibody.
  • the antibody binds specifically to TIGIT.
  • the method further comprises the earlier step of selecting for treatment a patient whose cancer shows high levels of ⁇ T cell infiltration.
  • the ⁇ T cell infiltration is detected by analysis of a tumor biopsy.
  • the analysis comprises an assay selected from
  • the cancer shows high levels of V51 + or double negative (DN) cell infiltrate.
  • the method further comprises the prior step of detecting ⁇ T cell infiltration, or V52 " ⁇ T cell infiltration, of tissues in a mammalian subject and selecting subjects who have ⁇ T cell infiltrates.
  • ⁇ T cell infiltration or V52 " ⁇ T cell infiltration is detected in tumors of a mammalian subject. Detection of ⁇ T cell or ⁇ 2 " ⁇ T-cells may be performed by any method known in the art for the detection of subpopulations of immune cells in tissue. T cell infiltration in tissues (e.g., tumor from tumor biopsies) can be determined by
  • An increase or decrease in expression of other markers can be additionally or alternatively used to characterize ⁇ T cells from tissue or ⁇ T cells from tissue which have been expanded ex-vivo (e.g., skin-derived ⁇ T cells and/or ⁇ - ⁇ 2 T cells, e.g., ⁇ T cells and/or DN T cells), including CD124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1, CD31, KLRG1, CD30, CD2, NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1, and CD64.
  • tissue or ⁇ T cells from tissue which have been expanded ex-vivo e.g., skin-derived ⁇ T cells and/or ⁇ - ⁇ 2 T cells, e.g., ⁇ T cells and/or DN T cells
  • the expanded population of ⁇ T cells has a greater mean expression of one or more of the markers selected from the group consisting of CD124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1, CD31, KLRG1, CD30, and CD2, relative to the separated population of ⁇ T cells, e.g., prior to expansion.
  • the expanded population of ⁇ T cells may have a greater frequency of cells expressing one or more of the markers selected from the group consisting of CD 124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1, CD31, KLRG1, CD30, and CD2, relative to the separated population of ⁇ T cells.
  • the expanded population of ⁇ T cells (e.g., skin-derived ⁇ T cells and/or non- ⁇ 2 T cells, e.g., ⁇ T cells and/or DN T cells) has a lower mean expression of one or more of the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1, and CD64, relative to the separated population of ⁇ T cells.
  • the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1, and CD64 relative to the separated population of ⁇ T cells.
  • the expanded population may similarly have a lower frequency of cells expressing one or more of the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1, and CD64, relative to the separated population of ⁇ T cells.
  • the TIGIT inhibitor is co-administered with an additional agent for treating cancer (e.g., an anti-cancer agent).
  • the composition is coadministered with an as yet to be described therapeutic for treating cancer.
  • the agents may be administered concurrently or sequentially.
  • agents described herein are administered prior to the other active agent(s).
  • the pharmaceutical formulations and modes of administration may be any of, but not limited to, those described below. It is also anticipated that there could be two or more co-administered chemical agents, biological agents or radiation that may each be administered using different modes or different formulations.
  • the method of treating cancer comprises administering a therapeutically effective amount of a TIGIT inhibitor to a cancer patient who has been determined to have an elevated representation of V51 + cells in a sample of peripheral blood.
  • the method further comprising the prior step of determining the representation of V51 + cells in the peripheral blood sample. In some embodiments, the method further comprises the step, after determining the representation of V51 + cells and before administering the TIGIT inhibitor, of selecting the patient for TIGIT inhibitor treatment if the patient's sample has been determined to have an elevated representation of V51 + cells.
  • the method comprises determining the total number of V51 + and V52 + in a blood sample from a potential mammalian subject. In particular embodiments, the method comprises determining the ratio of V51 + and V52 + in a blood sample from a potential mammalian subject. In certain embodiments, the method further comprises comparing the total number of V51 + and V52 + in a blood sample from a subject with cancer to a healthy individual. In certain embodiments, the method further comprises comparing the ratio of V51 + and V52 + in a blood sample from a subject with cancer to a healthy individual. [0108] In some embodiments, the elevated representation is an increase in V51 + cells as a percentage of total lymphocytes in the sample.
  • greater than 0.2% of total lymphocytes in the sample are determined to be V51 + cells. In particular embodiments, greater than 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9% or 1.0% of total lymphocytes in the sample are determined to be V51 + cells. In some embodiments, greater than 1.5%, 2.0%, 2.5%), 3.0%), 3.5%), 4.0%), or 5.0% of total lymphocytes in the sample are determined to be V51 + cells.
  • the elevated representation is an increase in V51 + cells as a percentage of total ⁇ T cells in the sample. In certain embodiments, greater than 10% of total ⁇ T cells in the sample are V51 + cells. In particular embodiments, greater than 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, or 20% of total ⁇ T cells in the sample are V51 + cells.
  • the elevated representation is an increase in the ratio of V51 + cells to V52 + cells in the sample.
  • the ratio of V51 + :V52 + cells is greater than 1 :9.
  • the ratio of V51 + :V52 + cells is greater than 1 :8, 1 :7, 1 :6, or 1 :5.
  • the TIGIT inhibitor is an antibody or antigen-binding antibody fragment that binds specifically to TIGIT.
  • the method further comprises administering at least one additional anti-cancer therapy.
  • at least one of the at least one additional anti-cancer therapy is a non-TIGIT checkpoint inhibitor.
  • the non-TIGIT checkpoint inhibitor is an antibody that binds specifically to PD- 1.
  • the non-TIGIT checkpoint inhibitor is an antibody that binds specifically to PDL 1.
  • the cancer is a solid, non-hematopoietic, cancer.
  • the cancer is a breast cancer.
  • the cancer is a hematological cancer.
  • the method further comprises administering at least one additional therapeutic agent.
  • the at least one additional therapeutic agent is an agonist of DNAM-1.
  • the at least one or more additional agent is an anti-cancer agent.
  • the anti-cancer agent is selected from the group consisting of radiation, a chemotherapeutic or growth inhibitory agent, a targeted therapeutic agent, a small molecule inhibitor, a T cell expressing a chimeric antigen receptor, an antibody or antigen- binding fragment thereof, an antibody-drug conjugate, an angiogenesis inhibitor, an
  • the anti-cancer agent is a chemotherapeutic agent.
  • the anti-cancer agent is a targeted therapeutic agent.
  • the anti-cancer agent is an antibody.
  • the anti-cancer agent is administered continuously. In some embodiments, the anti-cancer agent is administered intermittently.
  • the TIGIT inhibitor is administered before the anti-cancer agent. In some embodiments, the TIGIT inhibitor is administered simultaneous with the anticancer agent. In some embodiments, the TIGIT inhibitor is administered after the anti-cancer agent.
  • the additional therapeutic agent is a checkpoint inhibitor.
  • methods of increasing the number of tissue-infiltrating ⁇ T cells in a mammalian subject recipient comprise (z) obtaining ⁇ T cells from tissue of a donor, (ii) expanding the ⁇ T cells ex vivo, ⁇ Hi) transplanting the expanded ⁇ T cells into a recipient, and (iv) administering at least one TIGIT inhibitor to the recipient.
  • the obtained and/or expanded ⁇ T cells are ⁇ 2 " .
  • the ⁇ 2 " cells are ⁇ 1 + or DN T cells.
  • ⁇ 2 " cells comprise both ⁇ 1 + and DN T cells.
  • the ⁇ T cells are obtained from hematopoietic tissue described herein in Section 6.3.1.1. [0124] In some embodiments, the ⁇ T cells are obtained from non-hematopoietic tissue, described herein in Section 6.3.1.2.
  • the TIGIT inhibitor is an inhibitor described herein in Section 6.6.
  • the ex vivo expanded V52 " ⁇ T cells were obtained by:
  • lymphocytes obtained from non-hematopoietic tissue of humans or non-human animals in the presence of interleukin-2 (IL-2) and/or interleukin-15 (IL-15), and not in direct contact with stromal or epithelial cells during culture.
  • IL-2 interleukin-2
  • IL-15 interleukin-15
  • the methods culture and expansion methods are selected from those described in Section 6.3 above.
  • the TIGIT inhibitor is selected from an antagonist of TIGIT activity, an antagonist of TIGIT interaction with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL2, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL3, an agent that inhibits and/or blocks the intracellular signaling within a V52 " ⁇ T cell mediated by PVR binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling within a V52 " ⁇ T cell mediated by PVRL2 binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling within a V52 " ⁇ T cell mediated by PVLR3 binding to TIGIT, and combinations thereof.
  • the TIGIT inhibitor is an antagonist of TIGIT interaction with PVR.
  • the TIGIT inhibitor is an agent that inhibits and/or blocks the interaction of TIGIT with PVR.
  • the TIGIT inhibitor may block the interaction of PVR with TIGIT, leading to sequestration of TIGIT and increased binding of PVR to DNAM1.
  • the TIGIT inhibitor is an antibody that binds specifically to TIGIT or to PVR.
  • the TIGIT inhibitor is an antigen-binding fragment of an antibody that binds specifically to TIGIT or to PVR.
  • the TIGIT inhibitor is an antigen binding antibody fragment selected from Fab, Fab', F(ab') 2 , F d , F v , complementarity determining region (CDR) fragment, or single-chain antibody (scFv).
  • the TIGIT inhibitor is a single domain antibody or single-chain antibody.
  • the single domain antibody is a V H H domain.
  • the TIGIT inhibitor is an inhibitor described in
  • the TIGIT inhibitor is an inhibitor described in WO 2017/030823, WO 2016/028656,
  • the TIGIT inhibitor is a human antibody.
  • the human antibody has variable regions in which both the framework and CDR regions are sequences identical to those of human origin.
  • the TIGIT inhibitor is a humanized antibody.
  • the humanized antibody is a non-human (e.g., murine) antibody that is a chimeric antibody and contains minimal sequence derived from non-human immunoglobulin.
  • the humanized antibody is a human immunoglobulin (recipient antibody) in which residues from a hypervariable region (HVR) of the recipient are replaced by residues from a HVR of a non- human species (donor antibody) such as mouse, rat, rabbit, or nonhuman primate having the desired specificity, affinity, and/or capacity.
  • donor antibody such as mouse, rat, rabbit, or nonhuman primate having the desired specificity, affinity, and/or capacity.
  • FR residues of the human immunoglobulin variable domain are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. In an embodiment, the humanized antibodies do not comprise residues that are not found in the recipient antibody or in the donor antibody.
  • a 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 FRs are those of a human immunoglobulin sequence.
  • the humanized antibody optionally will also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • the Fc regions of the antibodies are modified.
  • the TIGIT inhibitor is a small molecule inhibitor. In certain aspects, the TIGIT inhibitor specifically binds TIGIT. In certain aspects, the TIGIT inhibitor binds PVR.
  • Methods of delivery include, but are not limited to, intra-arterial, intra-muscular, intravenous, intranasal, and oral routes.
  • Methods to enhance uptake can include, but are not limited to, encapsulation in liposomes, microparticles, microcapsules, receptor-mediated endocytosis, and the like.
  • the agents identified can be administered to subjects or individuals susceptible to or at risk of developing a variety of conditions benefiting from activation of ⁇ T cells.
  • the agent can be added to a pharmaceutically acceptable carrier and systemically or topically administered to the subject.
  • Therapeutic amounts are empirically determined and vary with the pathology being treated, the subject being treated and the efficacy and toxicity of the agent. When delivered to an animal, the method is useful to further confirm efficacy of the agent.
  • in vivo administration is effected in one dose, continuously or intermittently throughout the course of treatment.
  • Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and vary with the composition used for therapy, the purpose of therapy, the target cell being treated, and the subject being treated. Single or multiple administrations are carried out with the dose level and pattern being selected by the treating physician.
  • Suitable dosage formulations and methods of administering the agents are readily determined by those of skill in the art.
  • the compounds are administered at about 0.01 mg/kg to about 200 mg/kg, more preferably at about 0.1 mg/kg to about 100 mg/kg, even more preferably at about 0.5 mg/kg to about 50 mg/kg.
  • the effective amount may be more or less than when the agent is used alone.
  • compositions can be administered orally, intranasally, parenterally or by inhalation therapy, and may take the form of tablets, lozenges, granules, capsules, pills, ampoules, suppositories or aerosol form. They may also take the form of suspensions, solutions and emulsions of the active ingredient in aqueous diluents, nonaqueous diluents, syrups, granulates or powders.
  • the active ingredient in aqueous diluents, nonaqueous diluents, syrups, granulates or powders.
  • compositions can also contain other pharmaceutically active compounds or a plurality of compounds of the invention.
  • an agent of the present invention also referred to herein as the active ingredient, may be administered for therapy by any suitable route including, but not limited to, oral, rectal, nasal, topical (including, but not limited to, transdermal, aerosol, buccal and sublingual), vaginal, parental (including, but not limited to, subcutaneous, intramuscular, intravenous and intradermal) and pulmonary. It is also appreciated that the preferred route varies with the condition and age of the recipient, and the disease being treated.
  • the agent should be administered to achieve peak concentrations of the active compound at sites of disease. This may be achieved, for example, by the intravenous injection of the agent, optionally in saline, or orally administered, for example, as a tablet, capsule or syrup containing the active ingredient.
  • Desirable blood levels of the agent may be maintained by a continuous infusion to provide a therapeutic amount of the active ingredient within disease tissue.
  • the use of operative combinations is contemplated to provide therapeutic combinations requiring a lower total dosage of each component antiviral agent than may be required when each individual therapeutic compound or drug is used alone, thereby reducing adverse effects.
  • the present invention also includes methods involving co-administration of the TIGIT inhibitors described herein with one or more additional active agents. Indeed, it is a further aspect of this invention to provide methods for enhancing prior art therapies and/or pharmaceutical compositions by co-administering the agents of this invention.
  • the agents may be administered concurrently or sequentially.
  • the agents described herein are administered prior to the other active agent(s).
  • the pharmaceutical formulations and modes of administration may be any of those described above.
  • the two or more co-administered chemical agents, biological agents or radiation may each be administered using different modes or different formulations.
  • one or more agents provided herein and one or more therapeutic agents, TIGIT inhibitors, T-cell checkpoint inhibitors or anti-cancer agents are administered to a subject under one or more of the following
  • the TIGIT inhibitor is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-oxidethyl-N-oxidethyl
  • the TIGIT inhibitor is administered prior to an additional TIGIT inhibitor, T-cell checkpoint inhibitor, therapeutic or anti-cancer agent, e.g., 0.5, 1 , 2, 3, 4, 5, 10, 12, or 18 hours, 1, 2, 3, 4, 5, or 6 days, or 1, 2, 3, or 4 weeks prior to the administration of therapeutic or anti-cancer agent.
  • the TIGIT inhibitor is administered after an additional TIGIT inhibitor, T-cell checkpoint inhibitor, therapeutic or anti-cancer agent, e.g., 0.5, 1, 2, 3, 4, 5, 10, 12, or 18 hours, 1 , 2, 3, 4, 5, or 6 days, or 1, 2, 3, or 4 weeks after the administration of an additional TIGIT inhibitor, T-cell checkpoint inhibitor, therapeutic or anti-cancer agent.
  • the TIGIT inhibitor and an additional TIGIT inhibitor, T-cell checkpoint inhibitor, therapeutic or anti-cancer agent are administered concurrently but on different schedules, e.g., the TIGIT inhibitor is administered daily while the additional TIGIT inhibitor, T-cell checkpoint inhibitor, therapeutic or anti-cancer agent, is administered once a week, once every two weeks, once every three weeks, or once every four weeks. In other embodiments, the TIGIT inhibitor is administered once a week while the additional TIGIT inhibitor, T-cell checkpoint inhibitor, therapeutic or anti-cancer agent is administered daily, once a week, once every two weeks, once every three weeks, or once every four weeks.
  • TIGIT inhibitors are administered in combination with an additional T-cell checkpoint inhibitor.
  • the T-cell checkpoint inhibitor or checkpoint inhibitor is any molecule that inhibits the activity of a T-cell checkpoint.
  • the T-cell checkpoint inhibitor inhibits a protein or gene that functions to inhibit or suppress the activation of a T-cell.
  • the T-cell checkpoint inhibitor inhibits the expression or activity of, but not limited to: A2AR, B7-H3, B7-H4, BTLA, CTLA-4, IDO, KIR, LAG3, PD-1, TIM-3, VISTA and combinations thereof.
  • TIGIT inhibitors are administered in combination with anticancer agents.
  • suitable therapeutic or anticancer agents are contemplated for use in the methods provided herein. Indeed, the methods provided herein can include but are not limited to, administration of numerous therapeutic agents such as: agents that induce apoptosis; polynucleotides (e.g., anti-sense, ribozymes, siR A); polypeptides (e.g., enzymes and antibodies); biological mimetics; alkaloids; alkylating agents; antitumor antibiotics; antimetabolites; hormones; platinum compounds; monoclonal or polyclonal antibodies (e.g., antibodies conjugated with anticancer drugs, toxins, defensins), toxins; radionuclides;
  • therapeutic agents such as: agents that induce apoptosis; polynucleotides (e.g., anti-sense, ribozymes, siR A); polypeptides (e.g., enzymes and antibodies); biological mimetics;
  • biological response modifiers e.g., interferons (e.g., IFN-a) and interleukins (e.g., IL-2)
  • adoptive immunotherapy agents hematopoietic growth factors; agents that induce tumor cell differentiation (e.g., all-trans-retinoic acid); gene therapy reagents (e.g., antisense therapy reagents and nucleotides); tumor vaccines; angiogenesis inhibitors; proteasome inhibitors: NF- KB modulators; anti-CDK compounds; HDAC inhibitors; and the like.
  • therapeutic agents such as chemotherapeutic compounds and anticancer therapies suitable for co-administration with the disclosed compounds are known to those skilled in the art.
  • the TIGIT inhibitors for use in the methods described herein can be formulated in pharmaceutical compositions.
  • the ex vivo cultured, and optionally expanded, ⁇ T cells for use in certain of methods described herein may formulated as a medicament.
  • compositions comprising TIGIT inhibitors can comprise, in addition to one or more of the TIGIT inhibitors, one or more additional checkpoint inhibitors, and a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient.
  • a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient.
  • the precise nature of the carrier or other material can depend on the route of administration, e.g. oral, intravenous, cutaneous or subcutaneous, nasal, intramuscular, intraperitoneal routes.
  • compositions for oral administration can be in tablet, capsule, powder or liquid form.
  • a tablet can include a solid carrier such as gelatin or an adjuvant.
  • Liquid pharmaceutical compositions generally include a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol can be included.
  • the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen- free and has suitable pH, isotonicity and stability.
  • isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringer's Injection.
  • Preservatives, stabilizers, buffers, antioxidants and/or other additives can be included, as required.
  • the pharmaceutically useful compound according to the present invention that is to be given to an individual, administration is preferably in a "therapeutically effective amount” or “prophylactically effective amount”(as the case can be, although prophylaxis can be considered therapy), this being sufficient to show benefit to the individual.
  • a therapeutically effective amount or “prophylactically effective amount”(as the case can be, although prophylaxis can be considered therapy)
  • prophylaxis can be considered therapy
  • the actual amount administered, and rate and time-course of administration will depend on the nature and severity of the cancer being treated. Prescription of treatment, e.g. decisions on dosage etc, is within the responsibility of general practitioners and other medical doctors, and typically takes account of the disorder to be treated, the condition of the individual patient, the site of delivery, the method of administration and other factors known to practitioners.
  • a composition can be administered alone or in combination with other treatments (e.g., additional TIGIT inhibitors, T-Cell checkpoint inhibitors and/or anti-cancer agents), either simultaneously or sequentially dependent upon the cancer to be treated.
  • additional TIGIT inhibitors e.g., additional TIGIT inhibitors, T-Cell checkpoint inhibitors and/or anti-cancer agents
  • PB Peripheral Blood
  • PBMCs Peripheral Blood Mononuclear Cells
  • Ficoll Ficoll-Paque PLUS, GE Healthcare Life Sciences
  • centrifugation 400 g for 20 minutes to separate red blood cells, blood plasma, and white lymphocytes/monocytes.
  • White blood cells were carefully harvested through a stripette and washed four times in cold PBS.
  • the PBMCs were used either immediately for immunostaining and phenotypic analysis of PB- derived ⁇ + T cells or cultured to have PB-derived ⁇ + T cell line for functional assays.
  • cells were resuspended in RPMI-1640 medium (Life Technologies) with 10% heat-inactivated foetal bovine serum (Life Technologies), L-glutamine (292 ⁇ g/ml; Life Technologies), penicillin (100 units/ml; Life Technologies), streptomycin (100 ⁇ g/ml; Life Technologies), N-2-hydroxyethylpiperazine-N-2-ethane sulfonic acid (HEPES; 0.01 M; Life Technologies), sodium pyruvate (1 mM; Life Technologies), minimal essential media (MEM) non-essential amino acids (IX; Life Technologies) at a density of 1 x 10 6 per mL and supplemented with human recombinant IL-2 (100 IU/ml; PROLEUKIN®; Novartis
  • IL-15 human recombinant IL-15 (20 ng/ml; Biolegend).
  • Cells were transferred into a 24 well plate that was coated with a-pan ⁇ TCR monoclonal Antibody (mAb) (20 ⁇ g/ml; clone Bl; Biolegend) 90 minutes prior to cell transfer.
  • mAb monoclonal Antibody
  • Cells were grown for 14 days, at 37°C and 5% C0 2 , media changed ever 2-3 days and fresh cytokines added. Upon reaching confluence, cells were split 1 : 1.
  • ⁇ + T cells Under these conditions, after 14 days, the original minor population of ⁇ + T cells is normally highly activated through their TCR (as indicated by upregulation if CD69 and CD25) and largely enriched consisting of mainly ⁇ 2 + T cells but also ⁇ T cells (up to 30% of all ⁇ + T cells).
  • lymphocytes were counted by either; (1) trypan blue stain (0.4%) (Life Technologies) and haemocytometer, or (2) CASY® Model TT cell counter and analyzer (Roche).
  • IMDM Iscove's Modified Dulbecco's Medium
  • L-glutamine (292 ⁇ g/ml; Life Technologies), penicillin (100 units/ml; Life Technologies), streptomycin (100 ⁇ g/ml; Life Technologies), N-2-hydroxyethylpiperazine-N- 2-ethane sulfonic acid (HEPES; 0.01 M; Life Technologies), sodium pyruvate (1 mM; Life Technologies), minimal essential media (MEM) non-essential amino acids (lx; Life
  • the matrices and media were transferred to a 50 ml centrifuge tube (Corning) containing 10 ml Hanks Balanced Salt Solution (HBSS; Life Technologies) with 0.01 mM HEPES (up to 12 matrices/tube).
  • the matrices were rinsed with the cell suspension using a 10 ml pipette, and the cell suspension was passed through a 70 ⁇ filter (BD Biosciences) into a fresh 50 ml centrifuge tube. The rinsing of the matrices was repeated two additional times.
  • the media from the culture well was also aspirated and passed through a 70 ⁇ filter into a fresh 50 ml centrifuge tube.
  • Lymphocytes (0.5xl0 6 cells/well) were plated in a 96-U bottom well plate and washed in PBS. First, a live/dead staining was done with the Fixable Blue Dead Cell Stain Kit (Life Technologies) or the Fixable Viability Dye eFluor 770 NIR (eBioscience) according the manufacture instructions.
  • BV510 conjugated a-CD3 clone OKT3; Biolegend
  • PC7-conjugated a-pan ⁇ IMMU510 clone; Beckman Coulter
  • FITC- conjugated a-V51 TS8.2 clone; Life Technologies
  • PE-conjugated a-V52 B6 clone;
  • PB-derived ⁇ + T cell line or Skin-derived lymphocytes or sorted pan ⁇ + T cells were plated in a 96-flat bottom well plate with a density of 0.2x10 6 cells/well.
  • the plate was previously coated with different combination of the following proteins: a-CD3 Ab (5 ⁇ g/ml) (HIT3a clone; Biolegend) or its isotype control (MOPC-173; Biolegend), recombinant human PVR Fc Chimera Protein (rhPVR) (20 ⁇ g/ml) (Life Technologies), recombinant human MICA Fc Chimera Protein (rhMICA) (10 ⁇ g/ml) (R&D Systems) or their hlgGl control (Biolegend).
  • a-CD3 Ab 5 ⁇ g/ml
  • HIT3a clone HIT3a clone
  • MOPC-173 isotype control
  • rhPVR recombinant human PVR Fc Chimer
  • the blocking a-DNAMl Ab (DX11 clone; BD Biosciences) was added to the cell suspension (10 ⁇ g/ml).
  • the cells were stimulated for 6 hours in RPMI 10%> FCS supplemented with the intracellular protein transport inhibitor Brefeldin A (20 ⁇ g/ml)(Sigma-Aldrich), at 37°C and 5% C0 2 .
  • the cells were harvested and moved to a 96-U bottom well plate, extracellular stained and fixed as previously described for the phenotypic analysis.
  • the intracellular staining was performed in Permeabilization Buffer (eBioscience) according the manufacture instructions and the cytokine production was assessed using the following mAbs: eFluor450-conjugated ⁇ -IFNy (4S.B3; eBioscience) APC-conjugated a-TNFa
  • Pan ⁇ + T cells were purified from Skin-derived lymphocytes using a a-pan ⁇ mAb (IMMU510 clone; Beckman Coulter) and a FACS Aria cell sorting. They were then stained with the CellTrace Violet (CTV) Cell Proliferation Kit (Life Technologies) and cultured in a 96-flat bottom well plate for 1 week in complete medium supplemented with IL-2.
  • CTV CellTrace Violet
  • the plate was previously coated with different combination of the following proteins: a-CD3 Ab (50 ng/ml) (HIT3a clone; Biolegend) or its isotype control (MOPC-173; Biolegend), rhPVR (10 ⁇ g/ml) (Life Technologies) or its hlgGl control (Biolegend).
  • a-CD3 Ab 50 ng/ml
  • MOPC-173 isotype control
  • rhPVR 10 ⁇ g/ml
  • hlgGl control Biolegend
  • V51 + cells The initial population of V51 + cells was 1.12% of lymphocytes.
  • Cells were seeded at 2 x 10 5 cells/well into 96 well flat bottom plates (Corning) or at 2 x 106 cells/well into 24 well plates (Corning) for expansion and supplemented with factors at the following concentrations: IL-2 (100 U/ml), IL-15 (10 ng/ml), IL-4 (5 ng/ml) and IL-21 (10 ng/ml).
  • IL-2 100 U/ml
  • IL-15 10 ng/ml
  • IL-4 5 ng/ml
  • IL-21 10 ng/ml
  • Example 1 ⁇ + T cell subpopulations in human PBMC and skin
  • lymphocytes using somatic gene rearrangement to generate diverse surface receptors, B cells, ⁇ T cells and ⁇ T cells respectively. Only ⁇ T cells show compartmentalization, with V52 TCR positive cells dominating the human blood whilst T cells recombining other TCR genes (eg. V51, 53, 54, 55, 56, 57, 58) are predominantly found in human tissues such as the skin, the gut, the lungs, the liver and others ( Figure 1 A).
  • TCR genes eg. V51, 53, 54, 55, 56, 57, 58
  • Peripheral blood mononuclear cells from 11 donors and human skin derived lymphocytes from 27 donors were isolated as described above. Flow cytometry was performed using the following antibody-fluorochrome conjugates: V51-FITC, CD3-BV510 and V52-PE. Samples were also stained for viability using the eFluor 770 NIR live/dead dye. All T lymphocytes present were gated using the marker CD3, and within this population the presence of V52 + ⁇ T cells or V51 + ⁇ T cells made visible using the according antibodies. The results show that V52 TCR positive cells are dominating in the human blood with almost no V51 T cells being present.
  • PB-derived ⁇ + T cells and Skin-derived ⁇ + T cells were tested by FACS analysis for the presence of DNAM1 and TIGIT on their surface.
  • DNAM1 and TIGIT expression was evaluated on a pan ⁇ + T cell population.
  • DNAMl was expressed by both PB- derived ⁇ + T cells and Skin-derived ⁇ + T cells.
  • ⁇ - ⁇ + T cells expressed TIGIT only upon activation, obtained in this case with PHA treatment.
  • TIGIT was already expressed by unstimulated Skin-derived ⁇ + T cells.
  • DNAMl and TIGIT expression was assessed by staining PB-derived and Skin-derived lymphocytes with anti- ⁇ and anti-V02 Abs, in addition to the pan ⁇ Ab.
  • This staining strategy allowed to gate three different populations into the pan ⁇ + gate: ⁇ 1 + , ⁇ 2 + and V61 " V62 " (named here Double Negative, DN).
  • ⁇ - ⁇ + T cells are mostly represented by the ⁇ 2 + population, and in a smaller percentage by ⁇ 1 + or DN.
  • the ⁇ 2 + population largely expressed DNAMl, more than 70% in all donors, while only a small proportion, less than 25%, or none of them expressed TIGIT.
  • ⁇ 1 + and DN populations present in the PB expressed DNAMl, even if to a lower level compare to the ⁇ 2 + population, and shared TIGIT constitutive expression as the Skin-derived ⁇ + T cells.
  • Skin-derived ⁇ + T cells are composed by ⁇ 1 + and DN cells, and even in this second more detailed staining, we confirmed that ⁇ 1 + and DN cells constitutively expressed
  • DNAMl and TIGIT are expressed by more than 70% of ⁇ and DN cells in all donors, and TIGIT is expressed by more than 50%> of ⁇ 1 + cells.
  • DN T cells showed a more variable TIGIT expression because it is a heterogeneous population and it is represented by populations expressing TIGIT and populations not expressing TIGIT, in a different proportion in every donor.
  • Figure 3 shows ⁇ T cells derived either from PBMCs or skin tissue express DNAMl .
  • ⁇ T cells isolated from human PBMCs ( Figure 4) and lymphocytes isolated from human skin tissue ( Figure 5) were stained with anti-TIGIT antibodies and antibodies specific for either the ⁇ chain or ⁇ chain TCR and subjected to FACS analysis.
  • V52 + ⁇ T cells isolated form PBMCs expressed high levels of DNAM1 ( Figure 6) and low levels of TIGIT ( Figure 7) while V51 + ⁇ T cells expressed low levels of DNAMl ( Figure 6) and high levels of TIGIT ( Figure 7).
  • DN cells (V5T V52 " ) expressed moderate levels of DNAMl ( Figure 8) and high levels of TIGIT.
  • ⁇ ⁇ T cells and DN cells isolated from human skin leukocytes expressed high levels of DNAMl and TIGIT ( Figures 8 and 9).
  • Example 3 aCD3-induced cytokine production is inhibited by PVR in V51 + and DN ⁇ T cells.
  • soluble PVR was added to aCD3 -stimulated DN and V51 + ⁇ T cells isolated from either human PBMCs (figure 10 and 11) or human skin lymphocytes ( Figures 11). PB-derived and Skin-derived lymphocytes were stimulated for 6 hours by plate-bound aCD3 Ab in
  • FIG. 11 shows collective plots for IFNy and TNFa production in ⁇ T cells derived from PB and Skin
  • V51 and DN ⁇ T cells The aCD3-induced proliferation of V51 and DN ⁇ T cells was impaired in the presence of PVR compare to its control, further confirming the inhibitory role of the PVR/TIGIT axis on the TCR-induced activation of V51 + and DN T cells.
  • PVR when is not in the context of a TCR stimulation, doesn't work as an inhibitor through TIGIT but works as a costimulator through DNAM1, as demonstrated by the stimulation with PVR only: V51 + and DN T cells are induced to produce, even if in a small amount, IFNy and TNFa and this production is reverted by a blocking aDNAMl mAb ( Figures 15 and 16).
  • Example 6 PVR inhibitory effect is lost on V51 + and V53 + cells not expressing
  • Tissue-derived V51 + and V53 + cells cultured and expanded ex-vivo in the presence of IL-2 and IL-15 (culture method 1) expressed high amounts of TIGIT
  • V51 + and V53 + cells cultured in the presence of IL-2, IL-4, IL-15 and IL-21 (culture method 2), expressed low levels of TIGIT
  • Figure 18 FACS analysis of tissue-derived V51 + and V53 + cells from individual donors cultured in the presence of IL-2, IL-4, IL-15 and IL-21, demonstrated that PVR inhibition of INFy and TNFa production was lost in V51 + and V53 + cells expressing low levels of TIGIT ( Figures 19 and 20).
  • the TCR stimulation is a strong activator of V51+ and DN T cells, in terms of production of inflammatory cytokines like IFNy and TNFa but also in terms of proliferation.
  • the TCR-induced activation of V51 + and DN T cells is normally kept under control by the PVR/TIGIT axis.
  • TIGIT is constitutively expressed by PB- and Skin-derived V51 + and DN T cells.
  • the inhibitory receptor TIGIT like has been extensively demonstrated, has greater affinity for its ligand PVR compare to the activator receptor DNAM1.
  • PVR preferentially binds TIGIT and triggers its inhibitory signaling into the cells dampening the activation of V51 + and DN T cells.
  • PVR is available to bind the costimulatory receptor DNAM1 and help the activation of V51 + and DN T cells.

Abstract

Described herein are methods for activating Vδ2- gamma-delta T cells (γδ T cells) in a mammalian subject, comprising antagonizing TIGIT activity of Vδ2- γδ T cells. The invention also relates to methods of treating cancers with tissue-infiltrated γδ T cells and methods of treating cancers comprising administration of pharmaceutical compositions comprising Vδ2- γδ T cells.

Description

METHODS OF ACTIVATING V DELTA 2 NEGATIVE GAMMA DELTA T
CELLS
1. CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of U.S. Provisional Application No 62/519,714 filed June 14, 2017, which is hereby incorporated by reference in its entirety.
2. FIELD OF THE INVENTION
[0002] The invention relates to methods for activating tissue-infiltrating gamma delta T cells, particularly V52~ gamma-delta T cells (γδ T cells), in a mammalian subject by antagonizing TIGIT activity of V52~ γδ T cells.
3. BACKGROUND
[0003] The growing interest in T cell immunotherapy for cancer has focused on the evident capacity of subsets of CD8+ and CD4+ αβ T cells to recognize cancer cells and to mediate host-protective functions, particularly when de-repressed by clinically mediated antagonism of inhibitory pathways exerted by PD-1, CTLA-4, and other receptors. Nonetheless, problems remain. For example, there are clinical scenarios in which the efficacy of such treatments is poor. Moreover, there are often profound adverse events, the capacity to predict either efficacy or adverse events is extremely limited, and there is very little explanation of the interactions that allow the host to sense tumor cells ("immunogenicity") that precede the activation of conventional, antigen- specific CD8+ and CD4+ αβ T cell responses.
[0004] Upon global analysis of leukocyte populations and prognostic patterns of many cancer types (where higher levels of estimated T cell fractions were found to generally correlate with superior survival), intra-tumorai gamma delta T cells (γδ T cells) emerged as one of the most significant favorable prognostic leukocyte population (Gentles et al., 2015). γδ T cells represent a subset of T cells that express on their surface a distinct, defining γδ T-cell receptor (TCR). The γδ TCR is made up of one gamma (γ) and one delta (δ) chain. Human γδ T cells can be broadly classified into two types: peripheral blood-resident γδ T cells and non- hematopoietic, tissue-resident, γδ T cells. Most blood-resident γδ T cells express a νδ2 TCR; however, a smaller population of νδ2"γδ T cells can be detected among γδ T cells in blood. Conversely, tissue-resident γδ T cells more frequently use νδΐ and/or other νδ chains (νδ2~ γδ T cells); however, a smaller population of νδ2+γδ T cells can be detected among γδ T cells in tissue. Therefore, there is a need for novel methods for the specific activation and/or derepression of γδ T cells, and in particular, the νδ2~γδ T cells.
4. SUMMARY OF THE INVENTION
[0005] In certain embodiments, the application herein discloses methods of activating and/or de-repressing νδ2" gamma-delta T cells (γδ T cells) in a mammalian subject, comprising antagonizing TIGIT activity of νδ2" γδ T cells present in the subject to an extent sufficient to activate the νδ2" γδ T cells in vivo. In certain aspects, the νδ2" γδ T cells are νδ1+ cells. In certain aspects, the νδ2" γδ T cells are νδΓ νδ2" double negative (DN) cells. In certain aspects, antagonizing TIGIT activity comprises administering at least one TIGIT inhibitor to the subject. In an aspect, the at least one TIGIT inhibitor is administered adjunctively with νδ2" γδ T cells that have been cultured ex vivo. In certain aspects, the at least one TIGIT inhibitor is added to the νδ2" γδ T cells during culturing ex vivo. In certain aspects, the cultured νδ2" γδ T cells have been expanded ex vivo. In aspect, the at least one TIGIT inhibitor is administered in a composition comprising the cultured γδ T cells. In certain aspects, the at least one TIGIT inhibitor is administered separately from the ex vivo expanded γδ T cells. In certain aspects, the methods further comprise administering to the subject at least one additional agent in an amount sufficient to activate νδ2" γδ T cells. In an aspect, the at least one additional agent is an agonist of DNAM-1. In certain aspects, the at least one TIGIT inhibitor is selected from the group consisting of an antagonist of TIGIT activity, an antagonist of TIGIT interaction with Polio virus receptor (PVR), an agent that inhibits and/or blocks the interaction of TIGIT with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL2, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL3, an agent that inhibits and/or blocks the intracellular signaling mediated by PVR binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling mediated by PVRL2 binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling mediated by PVLR3 binding to TIGIT, and combinations thereof. In an aspect, the TIGIT inhibitor is an antibody. In an aspect, the antibody binds specifically to TIGIT. In an aspect, the antibody binds to PVR to specifically inhibit PVR interaction with TIGIT. In an aspect, the antibody is a full length IgG antibody. In an aspect, the antibody is an antigen-binding antibody fragment. In an aspect, the antibody is a single domain antibody. In an aspect, the antibody is fully human. In an aspect, the antibody is humanized. In an aspect, the TIGIT inhibitor is a small molecule inhibitor. In certain aspects, the ex vivo expanded V52" γδ T cells were obtained by culturing lymphocytes obtained from non-hematopoietic tissue of humans or non-human animals in the presence of interleukin-2 (IL-2) and/or interleukin-15 (IL-15), and not in direct contact with stromal or epithelial cells during culture. In an aspect, the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non- hematopoietic tissue in the presence of IL-2. In an aspect, the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of interleukin-15 (IL-15). In an aspect, the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of IL-2 and IL-15. In an aspect, the culturing step comprises culturing the lymphocytes in the absence of TCR activation or co-stimulation signals. In an aspect, the culturing step comprises culturing the lymphocytes in the absence of a T cell receptor pathway agonist. In an aspect, the culturing step comprises culturing the lymphocytes in the absence of stromal or epithelial cells. In an aspect, stromal or epithelial cells are removed prior to culture. In an aspect, the lymphocytes are cultured in the absence of fibroblasts. In an aspect, the lymphocytes have been obtained from skin, the gastrointestinal tract {e.g. colon), mammary gland tissue, lung, liver, pancreas or prostate. In an aspect, the ex vivo expanded V52" γδ T cells were obtained by culturing lymphocytes obtained from non-hematopoietic tissue of humans or non-human animals in the presence of IL-2, IL-15, and a factor selected from the group consisting of IL-4, IL-21, IL-6, IL-7, IL-8, IL-9, IL-12, IL-18, IL-33, IGF-1, IL-Ιβ, human platelet lysate (HPL), and stromal cell-derived factor- 1 (SDF-1) for at least 5 days to produce an expanded population of γδ T cells. In an aspect, within 14 days of culture, the expanded population of γδ T cells comprises at least 20-fold the number of γδ T cells as the γδ T cells obtained from a non-hematopoietic tissue. In an aspect, within 7 days of culture, the expanded population of γδ T cells comprises at least 2-fold the number of γδ T cells as the γδ T cells obtained from a non-hematopoietic tissue. In an aspect, the expanded population of γδ T cells is at least 50% νδ1+ cells. In an aspect, the expanded population of γδ T cells is at least 70% νδ1+ cells. In an aspect, the expanded population of γδ T cells is at least 90%> νδ1+ cells. In an aspect, the subject has cancer. In an aspect, the cancer has high infiltration of γδ T cells. The application further discloses a TIGIT inhibitor as disclosed in any of the aspects and embodiments described herein for use in any of the methods described herein of activating and/or de-repressing νδ2" gamma-delta T cells (γδ T cells) in a mammalian subject. [0006] In certain embodiments, the application discloses herein a method of treating cancers having high infiltration of γδ T cells, comprising administering at least one TIGIT inhibitor to a patient with a cancer having high infiltration of γδ T cells, in an amount sufficient to activate tissue-infiltrating γδ T cells in the patient. In an aspect, the cancer-infiltrating γδ T cells are νδ2" cells. In an aspect, the νδ2" cells are νδ1+ or DN cells. In an aspect, the at least one TIGIT inhibitor is selected from the group consisting of an antagonist of TIGIT activity, an antagonist of TIGIT interaction with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL2, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL3, an agent that inhibits and/or blocks the intracellular signaling mediated by PVR binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling mediated by PVRL2 binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling mediated by PVLR3 binding to TIGIT, and combinations thereof. In an aspect, the TIGIT inhibitor is an antibody. In an aspect, the antibody binds specifically to TIGIT. In an aspect, the antibody binds to PVR to specifically inhibit PVR interaction with TIGIT. In an aspect, the antibody is a full length IgG antibody. In an aspect, the antibody is an antigen-binding antibody fragment. In an aspect, the antibody is a single domain antibody. In an aspect, the antibody is fully human. In an aspect, the antibody is humanized. In an aspect, the TIGIT inhibitor is a small molecule inhibitor. In an aspect, the methods further comprise the antecedent step of selecting for treatment a patient whose cancer shows high levels of γδ T cell infiltration. In an aspect, the γδ T cell infiltration is detected by analysis of a tumor biopsy by a method comprising an assay selected from the group consisting of immunohistochemistry, polymerase chain reaction, in situ hybridization, and combinations thereof. In an aspect, the γδ T cell infiltration comprises νδ1+ or double negative (DN) cells. In an aspect, at least one additional therapeutic agent is administered. In an aspect, the at least one or more additional agents comprises an agonist of DNAM-1. In an aspect, the at least one or more additional agents comprises an anti-cancer agent. In an aspect, the anti-cancer agent is selected from the group consisting of radiation, a chemotherapeutic or growth inhibitory agent, a targeted therapeutic agent, a small molecule inhibitor, a T cell expressing a chimeric antigen receptor, an antibody or antigen-binding fragment thereof, an antibody-drug conjugate, an angiogenesis inhibitor, an antineoplastic agent, a cancer vaccine, an adjuvant, and combinations thereof. In an aspect, the anti-cancer agent is a chemotherapeutic. In an aspect, the anti-cancer agent is a small molecule inhibitor. In an aspect, the anti-cancer agent is an antibody. In an aspect, the TIGIT inhibitor is administered continuously. In an aspect, the TIGIT inhibitor is administered intermittently. In an aspect, the anti-cancer agent is administered continuously. In an aspect, the anti-cancer agent is administered intermittently. In an aspect, wherein the TIGIT inhibitor is administered before the anti-cancer agent. In an aspect, the TIGIT inhibitor is administered simultaneous with the anti-cancer agent. In an aspect, the TIGIT inhibitor is administered after the anticancer agent. The application further discloses a TIGIT inhibitor as disclosed in any of the aspects and embodiments described herein for use in any of the methods described herein of treating cancers having high infiltration of γδ T cells.
[0007] In certain embodiments, the application discloses herein a method of increasing the number of tissue infiltrating γδ T cells in a mammalian subject recipient, comprising obtaining γδ T cells from tissue comprising γδ T cells form a donor; expanding the γδ T cells ex vivo; transplanting the expanded γδ T cells into a recipient, and administering at least one TIGIT inhibitor to the recipient. In an aspect, the obtained and/or expanded γδ T cells are νδ2~. In an aspect, the νδ2" cells are νδ1+ or DN. In as aspect, the at least one TIGIT inhibitor is selected from the group consisting of an antagonist of TIGIT activity, an antagonist of TIGIT interaction with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL2, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL3, an agent that inhibits and/or blocks the intracellular signaling mediated by PVR binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling mediated by PVRL2 binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling mediated by PVLR3 binding to TIGIT, or combinations thereof. In an aspect, the at least one TIGIT inhibitor is an antibody. In as aspect, the antibody binds specifically to TIGIT. In an aspect, the antibody binds to PVR. In an aspect, the antibody is a full length IgG antibody. In an aspect, the antibody is an antigen- binding antibody fragment. In as aspect, the antibody is a single domain antibody. In an aspect, the antibody is fully human. In as aspect, the antibody is humanized. In an aspect, the at least one TIGIT inhibitor comprises a small molecule inhibitor. In an aspect, the ex vivo expanded νδ2" γδ T cells were obtained by culturing lymphocytes obtained from non- hematopoietic tissue of humans or non-human animals in the presence of interleukin-2 (IL-2) and/or interleukin-15 (IL-15), and not in direct contact with stromal or epithelial cells during culture. In an aspect, the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of IL-2. In an aspect, the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of interleukin-15 (IL-15). In an aspect, the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of IL-2 and IL-15. In an aspect, the culturing step comprises culturing the lymphocytes in the absence of TCR activation or co-stimulation signals. In an aspect, the culturing step comprises culturing the lymphocytes in the absence of a T cell receptor pathway agonist. In an aspect, the culturing step comprises culturing the lymphocytes in the absence of stromal or epithelial cells. In an aspect, stromal or epithelial cells are removed prior to culture. In as aspect, the lymphocytes are cultured in the absence of fibroblasts. In an aspect, the lymphocytes have been obtained from skin, the gastrointestinal tract (e.g. colon), mammary gland tissue, lung, liver, pancreas or prostate. In an aspect, the ex vivo expanded V52" γδ T cells were obtained by culturing lymphocytes obtained from non- hematopoietic tissue of humans or non-human animals in the presence of IL-2, IL-15, and a factor selected from the group consisting of IL-4, IL-21, IL-6, IL-7, IL-8, IL-9, IL-12, IL-18, IL-33, IGF-1, IL-Ιβ, human platelet lysate (HPL), and stromal cell-derived factor-1 (SDF-1) for at least 5 days to produce an expanded population of γδ T cells. In an aspect, within 14 days of culture, the expanded population of γδ T cells comprises at least 20-fold the number of γδ T cells as the γδ T cells obtained from a non-hematopoietic tissue. In an aspect, within 7 days of culture, the expanded population of γδ T cells comprises at least 2-fold the number of γδ T cells as the γδ T cells obtained from a non-hematopoietic tissue. In an aspect, the expanded population of γδ T cells is at least 50% νδ1+ cells. In an aspect, the expanded population of γδ T cells is at least 70% νδ1+ cells. In an aspect, the expanded population of γδ T cells is at least 90% νδ1+ cells. In an aspect, the administration of the at least one TIGIT inhibitor is performed prior to, during, after the transplantation, or combinations thereof. In an aspect, at least one additional agent is administered to the recipient. In an aspect, the at least one additional agent is an anti-cancer agent. In an aspect, the anti-cancer agent is selected from the group consisting of radiation, a chemotherapeutic or growth inhibitory agent, a targeted therapeutic agent, a small molecule inhibitor, a T cell expressing a chimeric antigen receptor, an antibody or antigen-binding fragment thereof, an antibody-drug conjugate, an angiogenesis inhibitor, an antineoplastic agent, a cancer vaccine, an adjuvant, and combinations thereof. In an aspect, the recipient has cancer. In an aspect, the γδ T cells are obtained from an autologous donor. In an aspect, the non-hematopoietic tissue is tumor tissue. In an aspect, the non-hematopoietic tissue is skin tissue. In an aspect, the method further comprises separating νδ1+γδ T cells or DN cells from the γδ T cells prior to transplantation. In an aspect, at least one additional checkpoint inhibitor is administered. In an aspect, the additional checkpoint inhibitor does not inhibit PD1. In an aspect, the additional checkpoint inhibitor inhibits TIM-3. In an aspect, the additional checkpoint inhibitor inhibits LAG-3. In an aspect of the methods, the administration of the TIGIT inhibitor results in elevated release of cytokines from the γδ T cells, selected from the group consisting of IFN-γ, TNF-a, interleukins, and combinations thereof. The application further discloses expanded γδ T cells obtained from tissue comprising γδ T cells from a donor as disclosed in any of the aspects and embodiments described herein and at least one TIGIT inhibitor as disclosed in any of the aspects and embodiments described herein for use in any of the methods described herein of increasing the number of tissue infiltrating γδ T cells in a mammalian subject recipient.
[0008] In certain embodiments, described herein are methods of treating cancer, comprising administering a therapeutically effective amount of a TIGIT inhibitor to a cancer patient who has been determined to have an elevated representation of νδ1+ cells in a sample of peripheral blood. In certain aspects, the methods further comprise the prior step of determining the representation of νδ1+ cells in the peripheral blood sample. In an aspect, the methods further comprise the step, after determining the representation of νδ1+ cells and before administering the TIGIT inhibitor, of selecting the patient for TIGIT inhibitor treatment if the patient's sample has been determined to have an elevated representation of νδ1+ cells. In certain aspects, the elevated representation is an increase in νδ1+ cells as a percentage of total lymphocytes in the sample. In certain aspects, greater than 0.5% of total lymphocytes in the sample are determined to be νδ1+ cells. In certain aspects, greater than 1.0% of total lymphocytes are determined to be νδ1+ cells. In certain aspects, greater than 2.0% of total lymphocytes are determined to be νδ1+ cells. In certain aspects, wherein the elevated representation is an increase in νδ1+ cells as a percentage of total γδ T cells in the sample. In certain aspects, greater than 10% of total γδ T cells in the sample are νδ1+ cells. In certain aspects, greater than 15% of total γδ T cells in the sample are νδ1+ cells. In certain aspects, the elevated representation is an increase in the ratio of νδ1+ cells to νδ2+ cells in the sample. In certain aspects, the ratio of νδ1+:νδ2+ cells is greater than 1 :9. In certain aspects, the ratio of νδ1+:Υδ2+ cells is greater than 1 :8. In certain aspects, the ratio of νδ1+:Υδ2+ cells is greater than 1 :7. In certain aspects, the ratio of V51+:V52+ cells is greater than 1 :6. In certain aspects, the TIGIT inhibitor is an antibody or antigen-binding antibody fragment that binds specifically to TIGIT. In certain aspects, the methods further comprise administration of at least one additional anti-cancer therapy. In certain aspects, at least one of the at least one additional anti-cancer therapy is administration of a non-TIGIT checkpoint inhibitor. In certain aspects, the non-TIGIT checkpoint inhibitor is an antibody that binds specifically to PD-1. In certain aspects, the non-TIGIT checkpoint inhibitor is an antibody that binds specifically to PDL1. In certain aspects, the cancer is a solid, non-hematopoietic, cancer. In certain aspects, the cancer is a breast cancer. In certain aspects, the cancer is a hematological cancer. The application further discloses a TIGIT inhibitor as disclosed in any of the aspects and embodiments described herein for use in any of the methods described herein of treating cancer in a patient who has been determined to have an elevated representation of V51+ cells in a sample of peripheral blood.
5. BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS
[0009] These and other features, aspects, and advantages of the present invention will become better understood with regard to the following description, and accompanying drawings, where:
[0010] Figure 1A provides a conceptual schema of sub-populations of T cells.
[0011] Figure IB shows results of flow cytometry performed on human peripheral blood mononuclear cells ("human blood") and human skin-derived lymphocytes ("human tissue") stained with antibodies specific for CD3, V51, and V52.
[0012] Figure 2 illustrates PVR expression on Antigen Presenting Cells (APC) and antagonistic action on T Cells through DNAM1 and TIGIT.
[0013] Figure 3 shows results of flow cytometry ("FACS") analysis for DNAM 1 expression of γδ T cells derived from human peripheral blood mononuclear cells ("PBMCs") and human skin lymphocytes. The shaded histogram is the signal of the Fluorescence minus one (FMO) control. "Percent of Max" refers to the percentage of maximum count.
[0014] Figure 4 shows results of FACS analysis for TIGIT expression of γδ T cells derived from human PBMCs, both unstimulated (left panel) and stimulated for 72 hours with PHA.
[0015] Figure 5 shows results of FACS analysis for TIGIT expression on γδ T cells derived from human skin. [0016] Figure 6 shows results of F ACS analysis for DNAM 1 and TIGIT expression on V51+, V52+, and DN γδ T cells derived from a representative donor of human PBMCs from peripheral blood (PB).
[0017] Figure 7 shows results of FACS analysis for DNAM 1 and TIGIT expression on V5T,
V52+, and DN γδ T cells derived from multiple donors of human PBMCs from PB.
[0018] Figure 8 shows results of FACS analysis for DNAM 1 and TIGIT expression on νδ and DN γδ T cells derived from a representative donor of human skin leukocytes.
[0019] Figure 9 shows results of FACS analysis for DNAM 1 and TIGIT expression on V5T and DN γδ T cells derived from multiple donors of human skin leukocytes.
[0020] Figure 10 shows interferon gamma (IFNy) production in anti-CD3 stimulated V51+,
V52+, and DN γδ T cells derived from human PBMCs upon exposure to a recombinant human
PVR Fc Chimera Protein (PVR) or an isotype control antibody (iso), demonstrating that PVR inhibits TCR stimulation in DN/νδ T cells from human PBMCs.
[0021] Figure 11 shows results from FACS analysis of Interferon gamma (IFNy) and Tumor Necrosis Factor alpha (TNFa) production in anti-CD3 stimulated V51+, V52+, and DN γδ T cells derived from human PBMCs and human skin leukocytes upon exposure to PVR or an isotype control antibody (iso), demonstrating that PVR inhibits TCR stimulation in DN/V51+ T cells. For the graphs showing V51+ and DN γδ T cells, n = 6 peripheral blood-derived γδ cell samples from different donors and 8 skin-derived γδ cell samples (2 out of 8 are sorted γδ cells). For the graphs showing νδ2+ γδ T cells, n = 3 peripheral blood-derived γδ cell samples and 1 skin-derived γδ cell samples. The graphs for νδ1+ and DN cell samples are generated from exactly all the same donors, while the graphs for V δ2+ are generated with a different set of donors. The donors used for the V δ2+ graphs are all included among the donors for V δ1+ and DN samples.
[0022] Figure 12 shows results of FACS analysis for proliferation indicator/dye retention of anti-CD3 stimulated νδ1+ and DN γδ T cells derived from human skin leukocytes upon exposure to PVR or an isotype control antibody, demonstrating the PVR inhibits TCR- induced proliferation in DN/V01+ T cells from human skin.
[0023] Figure 13 shows quantitation of results from FACS analysis for proliferation indicator/dye retention of anti-CD3 stimulated νδ1+ and DN γδ T cells derived from human skin leukocytes upon exposure to PVR or an isotype control antibody, demonstrating that PVR inhibits TCR-induced proliferation in DN/V01+ T cells from human skin. [0024] Figure 14 shows quantification of results from FACS analysis of Interferon gamma (IFNy) and Tumor Necrosis Factor alpha (TNFa) production in anti-CD3 stimulated V51+ and DN γδ T cells derived from human skin leukocytes upon exposure to an IgG control (mlgG), anti-CD3 (aCD3), and anti-CD3 plus PVR, demonstrating that PVR specifically inhibits TCR signaling.
[0025] Figure 15 shows quantification of results from FACS analysis of Interferon gamma (IFNy) production in anti-CD3 stimulated V51+ and DN γδ T cells derived from human skin leukocytes upon exposure to PVR and/or MICA, anti-DNAMl or an IgG control.
[0026] Figure 16 shows quantification of results from FACS analysis of Tumor Necrosis Factor alpha (TNFa) production in anti-CD3 stimulated V51+ and DN γδ T cells derived from human skin leukocytes upon exposure to PVR and/or MICA, anti-DNAMl or an IgG control, demonstrating that PVR specifically inhibits TCR signaling.
[0027] Figure 17 shows quantification of results from FACS analysis of granulocyte- macrophage colony-stimulating factor (GMCSF), Interferon gamma (IFNy) and Tumor Necrosis Factor alpha (TNFa) production of γδ T cells from human skin lymphocytes of 5 different donors sorted with a pan γδ T cell antibody upon exposure to an IgG control (mlgG), anti-CD3 (aCD3), and anti-CD3 plus PVR, demonstrating that PVR is a general inhibitor of TCR stimulation.
[0028] Figure 18 shows quantification of results from FACS analysis for TIGIT expression of tissue-derived V51+ and V53+ cells cultured in the presence of IL-2 and IL-15 (culture method 1), or in the presence of IL-2, IL-4, IL-15 and IL-21 (culture method 2), demonstrating that cells grown using culture method 2 express low levels of TIGIT.
[0029] Figure 19 are graphs showing quantification of results from FACS analysis of tissue- derived V51+ and V53+ cells from an individual donor cultured in the presence of IL-2, IL-4, IL-15 and IL-21 upon exposure to an IgG control (mlgG), anti-CD3 (aCD3), and anti-CD3 plus PVR, demonstrating that the PVR inhibition of INFy and TNFa was lost in V51+ and V53+ cells expressing low levels of TIGIT.
[0030] Figure 20 are graphs showing quantification of results from FACS analysis of tissue- derived V51+ and V53+ cells from an individual donor cultured in the presence of IL-2, IL-4, IL-15 and IL-21 upon exposure to an IgG control (mlgG), anti-CD3 (aCD3), and anti-CD3 plus PVR, demonstrating that the PVR inhibition of INFy and TNFa was lost in V51+ and V53+ cells expressing low levels of TIGIT. 6. DETAILED DESCRIPTION
6.1. Advantages and utility
[0031] Briefly, and as described in more detail below, described herein are methods for activating tissue-infiltrating γδ T cells, particularly V52" γδ T cells, in mammalian subjects, by antagonizing TIGIT activity of the V52" γδ T cells, and methods for the treatment of cancer comprising administration of at least one TIGIT inhibitor.
6.2. Definitions
[0032] Terms used herein have the meanings ascribed to them by those of skill in the art unless explicitly defined as set forth below.
[0033] "V62" γδ T cells" refers to any γδ T cell that does not express V52.
[0034] "Double negative (DN)" refers to γδ T cells that do not express either V51 or V52.
DN cells can express, but are not limited to those that express, V53, V54, V55, V56, V57, and
V58.
[0035] The phrase "activate a T cell" includes activation, de-repression, and both activation and de-repression of the T cell.
[0036] "TIGIT inhibitor" refers to any agent or compound capable of disrupting TIGIT activity or disrupting TIGIT interaction with PVR. A "TIGIT inhibitor" can increase PVR interaction with DNAMl . For example, a "TIGIT inhibitor" may block the interaction of PVR with TIGIT, leading to sequestration of TIGIT and increased binding of PVR to DNAMl. A "TIGIT inhibitor" can be an antagonist of TIGIT activity, an antagonist of TIGIT interaction with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL2, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL3, an agent that inhibits and/or blocks the intracellular signaling within a V52" γδ T cell mediated by PVR binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling within a V52" γδ T cell mediated by PVRL2 binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling within a V52" γδ T cell mediated by PVLR3 binding to TIGIT, and combinations thereof. [0037] As used herein, an "expanded population of γδ cells" refers to a population of cells that has been cultured under conditions and for a duration sufficient to cause proliferation of γδ cells within that population. Likewise, an "expanded population of V61+ T cells," as used herein, refers to a population of cells that has been cultured under conditions and for a duration sufficient to cause proliferation of V51+ T cells within that population.
[0038] "Tissue-resident", "tissue-infiltrated" and "non-hematopoietic" refers to cells that are located at non-vascular anatomical sites.
[0039] "Antigen binding fragment" of an antibody or "antigen binding antibody fragment" refers to any single chain or multiple chain portion of an antibody that is capable of binding specifically to an epitope of an antigen. Antigen binding fragment includes, without limitation, Fab, F(ab)2, and single-chain Fv (scFv).
[0040] "Human antibody" unless otherwise indicated is one whose sequences correspond to (i.e., are identical in sequence to) an antibody that could be produced by a human and/or consists entirely of amino acid sequences that are encoded in the human genome. A "human antibody" as used herein can be produced using various techniques known in the art, including phage-display libraries and by administering the antigen (e.g., TIGIT) to a transgenic animal that has been modified to produce such antibodies in response to antigenic challenge, but whose endogenous loci have been disabled, e.g., immunized Xenomice.
[0041] "Humanized antibodies" refer to non-human (e.g., murine) antibodies that are chimeric antibodies and contain minimal sequence derived from non-human immunoglobulin. In one embodiment, a humanized antibody is a human immunoglobulin (recipient antibody) in which residues from a hypervariable region (HVR) of the recipient are replaced by residues from a HVR of a non-human species (donor antibody) such as mouse, rat, rabbit, or nonhuman primate having the desired specificity, affinity, and/or capacity. In some instances, FR residues of the human immunoglobulin variable domain are replaced by corresponding non-human residues. These modifications may be made to further refine antibody performance. Furthermore, in a specific embodiment, humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. In an embodiment, the humanized antibodies do not comprise residues that are not found in the recipient antibody or in the donor antibody. In general, a 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 FRs are those of a human immunoglobulin sequence. The humanized antibody optionally will also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. In an embodiment where the humanized antibodies do comprise residues that are not found in the recipient antibody or in the donor antibody, the Fc regions of the antibodies are modified.
[0042] "Binds specifically", "capable of specifically binding", "specifically binds", or "preferentially binds" refers to the property of an antibody or fragment of binding to the (specified) antigen with a dissociation constant that is <1 μΜ, preferably <1 nM and most preferably <10 pM. In an embodiment, the Kd of the antibody that binds specifically to TIGIT is 250-500 pM. An epitope that "binds specifically "or "binds preferentially" (used
interchangeably herein) to an antibody or a polypeptide is a term well understood in the art, and methods to determine such specific or preferential binding are also well known in the art. A molecular entity is said to exhibit "specific binding" or "preferential binding" if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular cell or substance than it does with alternative cells or substances. An antibody "specifically binds" or "preferentially binds" to a target if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other substances. For example, an antibody that specifically or preferentially binds to a TIGIT conformational epitope is an antibody that binds this epitope with greater affinity, avidity, more readily, and/or with greater duration than it binds to other TIGIT epitopes or non-TIGIT epitopes. It is also understood by reading this definition that, for example, an antibody (or moiety or epitope) that specifically or preferentially binds to a first target may or may not specifically or preferentially bind to a second target. As such, "specific binding" or "preferential binding" does not necessarily require (although it can include) exclusive binding.
[0043] In certain embodiments, cancers or tissues with "high γδ T cell infiltration" refers to tissues (e.g., tumor from tumor biopsies) wherein the fraction of γδ T cells to total leukocytes, either measured or estimated, is greater than the fraction of γδ T cells in a given reference tissue, such as non-tumor tissue of a similar origin. In certain embodiments, tissues with "high γδ T cell infiltration" refers to tissues wherein the fraction of γδ T cells to total leukocytes is greater than 0.0001 , greater than 0.001 , greater than 0.01 , greater than 0.1 , greater than 0.2, greater than 0.3, greater than 0.4, or greater than 0.5. In certain embodiments, tissues with "high γδ T cell infiltration" refers to tissues in which the fraction of γδ T R A to total leukocyte R A is greater than 0.0001, greater than 0.001, greater than 0.01 , or greater than 0.1.
[0044] "T-cell checkpoints" or "checkpoints" refers to any protein or gene that functions to inhibit or suppress the activation of a T-cell. T-cell checkpoints include, but are not limited to: A2A , B7-H3, B7-H4, BTLA, CTLA-4, IDO, KIR, LAG3, PD-1, TIM-3 and VISTA.
[0045] "T-cell checkpoint inhibitor" or "checkpoint inhibitor" refers to any molecule that inhibits the activity of a T-cell checkpoint.
[0046] "Sufficient amount" means an amount sufficient to produce a desired effect, e.g., an amount sufficient to modulate γδ T cell activity.
[0047] "Therapeutically effective amount" is an amount that is effective to ameliorate a symptom of a disease. A therapeutically effective amount can be a "prophylactically effective amount" as prophylaxis can be considered therapy.
[0048] The phrases "adjunctive administration" or "administered to the subject adjunctively" are used interchangeably herein to mean administering a second therapeutic agent in therapeutically effective temporal proximity to a first therapeutic agent. In typical embodiments, adjunctive administration improves the effectiveness of the second therapeutic agent, the first therapeutic agent, or both the first and the second therapeutic agent as compared to administration of the agent alone.
[0049] Abbreviations used in this application include, but are not limited to, the following: "γδ T cells" refers to gamma delta T cells; "TIGIT" refers to T-Cell-IG and ITIM domain; TCR refers to the T-cell Receptor, "PVR" refers to Poliovirus receptor, "IL-2" refers to native or recombinant inter leukin 2; and "IL-15" refers to native or recombinant interleukin 15, etc., "PB" refers to peripheral blood and "PBMC" refers to peripheral blood mononuclear cells.
[0050] It must be noted that, as used in the specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise.
6.3. Methods for activating V62" gamma-delta T cells
[0051] In a first aspect, methods are presented for activating V52" gamma-delta T cells (γδ T cells) in a mammalian subject. The methods comprise antagonizing TIGIT activity of V52" γδ T cells present in the subject to an extent sufficient to activate the V52" γδ T cells in vivo. In some embodiments, the V52" γδ T cells are V51+ cells. In some embodiments, the V52" γδ T cells are V51" V52" double negative (DN) cells. [0052] In typical embodiments, antagonizing TIGIT activity comprises administering at least one TIGIT inhibitor to the subject. TIGIT inhibitors suitable for use in the methods are described below in Section 6.6.
6.3.1. Adjunctive administration with ex vivo cultured V62- γδ T cells
[0053] In a variety of embodiments, the at least one TIGIT inhibitor is administered to the subject adjunctively with V52" γδ T cells that have been cultured ex vivo.
[0054] In some embodiments, the at least one TIGIT inhibitor is administered in a
composition that also comprises the cultured γδ T cells. In certain of these embodiments, the at least one TIGIT inhibitor is added to the V52" γδ T cells during culturing ex vivo. In certain embodiments, the at least one TIGIT inhibitor is added to the V52" γδ T cells after culturing ex vivo.
[0055] In other embodiments, the at least one TIGIT inhibitor is administered to the subject separately from, but adjunctively with, the ex vivo expanded γδ T cells.
6.3.1.1. γδ T cells from hematopoietic tissue
[0056] Although νδ2+ T cells predominate among the γδ cells in hematopoietic tissues, νδ2" cells are also present, albeit in lower numbers. In some embodiments, the γδ T cells are obtained for culture from hematopoietic tissues, such as peripheral blood. In certain embodiments, νδ2" γδ T cells are obtained for culture from the hematopoietic tissues.
In certain embodiments, γδ T cells from hematopoietic tissues are stimulated to reduce νδ2 expression and/or induce νδΐ expression and/or selectively increase proliferation of νδ2" cells. In various embodiments, the γδ T cells are stimulated according to a protocol described in WO 2012/156,958, the contents of which are incorporated herein by reference in their entirety. In select embodiments, γδ T cells obtained from hematopoietic tissues are stimulated with a γδ TCR antibody to expand νδ2" γδ T cells and/or νδ1+Τ cells ex-vivo. In certain embodiments, γδ T cells obtained from hematopoietic tissues are stimulated with a γδ TCR antibody to increase TIGIT expression of the γδ T cells ex-vivo.
6.3.1.2. γδ T cells from non-hematopoietic tissue
[0057] In various embodiments, γδ T cells are obtained for culture from non-hematopoietic tissues. Detailed methods for the isolation, culturing and expansion of γδ T cells from non- hematopoietic tissues are disclosed in WO 2017/072367, which is incorporated herein by reference in its entirety.
[0058] In a variety of embodiments, V52" γδ T cells for culture are obtained from any human or non-human mammal non-hematopoietic tissue that can be removed from a subject.
[0059] In some embodiments, the non-hematopoietic tissue from which the V52" γδ T cells are obtained for culture is skin (e.g., human skin), which can be obtained by methods known in the art. In some embodiments, the skin is obtained by punch biopsy. In other embodiments, the V52" γδ T cells are obtained for culture from the gastrointestinal tract (e.g., colon), mammary gland, lung, prostate, liver, spleen, and pancreas. In some embodiments, the V52" γδ T cells are obtained for culture from non-neoplastic tissues. In some embodiments, the V52" γδ T cells are obtained for culture from human cancer tissues, e.g., tumors of the breast, the prostate, or other solid tumors.
[0060] The γδ T cells that are dominant in the non-hematopoietic tissues are primarily V51+ T cells, such that V51+ T cells comprise about 70-80% of the non-hematopoietic tissue-resident γδ T cell population. In various embodiments, the V52" γδ T cells that have been cultured ex vivo are V51+.
[0061] Some γδ T cells that are resident in non-hematopoietic tissues express neither V51 nor V52 TCR and we have named them double negative (DN) γδ T cells. In some embodiments, the V52" γδ T cells that have been cultured ex vivo are V51"V52" double negative (DN) cells. These DN γδ T cells are likely to be mostly V53 -expressing with a minority of V55 -expressing T cells.
6.3.1.2.1. Expansion of non-hematopoietic tissue- resident γδ T cells
[0062] In some embodiments, the cultured V52" γδ T cells have been expanded ex vivo.
[0063] In certain embodiments, the ex vivo expanded V52" γδ T cells are obtained by culturing lymphocytes obtained from non-hematopoietic tissue of humans or non-human animals in the presence of inter leukin-2 (IL-2) and/or interleukin-15 (IL-15), and not in direct contact with stromal or epithelial cells during culture.
[0064] In certain embodiments, the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of IL-2. In certain embodiments, the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of interleukin-15 (IL- 15). In particular embodiments, the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of IL-2 and IL-15.
[0065] In typical embodiments, the culturing step comprises culturing the lymphocytes in the absence of TCR activation or co-stimulation signals. In some embodiments, the culturing step comprises culturing the lymphocytes in the absence of a T cell receptor pathway agonist. In some embodiments, the culturing step comprises culturing the lymphocytes in the absence of stromal or epithelial cells. In some embodiments, stromal or epithelial cells are removed prior to culture. In some embodiments, the lymphocytes are cultured in the absence of fibroblasts. In various embodiments, the lymphocytes have been obtained from skin, the gastrointestinal tract (e.g., colon), mammary gland tissue, lung, liver, pancreas or prostate.
[0066] In some embodiments, the ex vivo expanded V52" γδ T cells are obtained by culturing lymphocytes obtained from non-hematopoietic tissue of humans or non-human animals in the presence of IL-2, IL-15, and a factor selected from the group consisting of IL-4, IL-21, IL-6, IL-7, IL-8, IL-9, IL-12, IL-18, IL-33, IGF-1, IL-Ιβ, human platelet lysate (HPL), and stromal cell-derived factor- 1 (SDF-1) for at least 5 days to produce an expanded population of γδ T cells.
[0067] In select embodiments, the ex vivo expanded V52" γδ T cells are obtained by culturing lymphocytes obtained from non-hematopoietic tissue of humans or non-human animals under conditions in which, within 14 days of culture, the expanded population of γδ T cells comprises at least 20-fold the number of γδ T cells as the γδ T cells obtained from a non- hematopoietic tissue. In some embodiments, the ex vivo expanded νδ2" γδ T cells were obtained by culturing lymphocytes obtained from non-hematopoietic tissue of humans or non- human animals under conditions in which, within 7 days of culture, the expanded population of γδ T cells comprises at least 2-fold the number of γδ T cells as the γδ T cells obtained from a non-hematopoietic tissue.
[0068] In particular embodiments, the expanded population of γδ T cells is at least 50% νδ1+ cells. In certain embodiments, the expanded population of γδ T cells is at least 70% νδ1+ cells. In select embodiments, the expanded population of γδ T cells is at least 90%> νδ1+ cells.
[0069] In certain embodiments, the method further comprises expanding non-hematopoietic tissue-resident γδ T cells (e.g., skin-derived γδ T cells and/or ηοη-νδ2 T cells, e.g., νδΐ T cells and/or DN T cells). In some embodiments, the non-hematopoietic tissue-resident γδ T cells are expanded from a population of γδ T cells that has been separated from non- hematopoietic tissue according to methods described above. In general, non-hematopoietic tissue-resident γδ T cells are capable of spontaneously expanding upon removal of physical contact with stromal cells (e.g., skin fibroblasts). Thus, the scaffold-based culture methods described above can be used to induce such separation, resulting in de-repression of the γδ T cells to trigger expansion. Accordingly, in some embodiments, no substantial TCR pathway activation is present during the expansion step (e.g., no exogenous TCR pathway activators are included in the culture). In various embodiments, the methods of expanding non- hematopoietic tissue-resident γδ T cells omit contact with feeder cells, tumor cells, and/or antigen-presenting cells.
[0070] It will be understood that the amount of each of the above cytokines required to produce an expanded population of γδ T cells will depend of the concentrations of one or more of the other cytokines. For example, if the concentration of IL-2 is increased or decreased, the concentration of IL-15 may be accordingly decreased or increased,
respectively. As noted above, the amount effective to produce an expanded population refers herein to composite effect of all factors on cell expansion.
[0071] In certain embodiments, the γδ T cells are exposed to certain factors prior to culture with other factors. For instance, the expansion culture can be gradually supplied with additional factors over the course of expansion, or, alternatively, the γδ T cells can be transferred from a culture of one factor or group of factors to another.
[0072] In some embodiments, the γδ T cells are expanded in culture for a period of several hours (e.g., about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 18, or 21 hours) to about 35 days (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, or 35 days). In one embodiment, the γδ T cells are expanded for a period of 14 to 21 days. Thus, including a separation culture period (e.g., of 1 to 40 days, e.g., 14 to 21 days), the separation and expansion steps, in some embodiments, can last between 28 and 56 days, or about 41 days.
[0073] Methods of expansion provide an expanded population of γδ T cells that is greater in number than a reference population. In some embodiments, the expanded population of γδ T cells is greater in number than the separated population of γδ T cells prior to the expansion step (e.g., at least 2-fold in number, at least 3-fold in number, at least 4-fold in number, at least 5-fold in number, at least 6-fold in number, at least 7-fold in number, at least 8-fold in number, at least 9-fold in number, at least 10-fold in number, at least 15-fold in number, at least 20-fold in number, at least 25-fold in number, at least 30-fold 35 in number, at least 35-fold in number, at least 40-fold in number, at least 50-fold in number, at least 60- fold in number, at least 70- fold in number, at least 80-fold in number, at least 90-fold in number, at least 100-fold in number, at least 200-fold in number, at least 300-fold in number, at least 400-fold in number, at least 500-fold in number, at least 600-fold in number, at least 700-fold in number, at least 800-fold in number, at least 900-fold in number, at least 1,000-fold in number at least 5,000- fold in number, at least 10,000-fold in number, or more relative to the separated population of γδ T cells prior to the expansion step). Thus, the invention provides a means to produce large populations of non-hematopoietic tissue-derived γδ T cells (e.g., skin-derived γδ T cells and/or ηοη-νδ2 T cells, e.g., νδΐ T cells and/or DN T cells) at high rates (e.g., by removing stromal cell contact and/or TCR stimulation, or by culturing in the presence of an effective amount of factors). In some embodiments, the expansion step described herein expands the γδ T cells at a low population doubling time, which is given by the following equation:
Doubling Time = Duration log2
Log (final concentration) - Log (initial concentration)
[0074] Given the information provided herein,, a skilled artisan will recognize that the invention provides methods of expanding non-hematopoietic tissue-derived γδ T cells (e.g., skin-derived γδ T cells and/or ηοη-νδ2 T cells, e.g., νδΐ T cells and/or DN T cells) at a population doubling time of less than 5 days (e.g., less than 4.5 days, less than 4.0 days, less than 3.9 days, less than 3.8 days, less 15 than 3.7 days, less than 3.6 days, less than 3.5 days, less than 3.4 days, less than 3.3 days, less than 3.2 days, less than 3.1 days, less than 3.0 days, less than 2.9 days, less than 2.8 days, less than 2.7 days, less than 2.6 days, less than 2.5 days, less than 2.4 days, less than 2.3 days, less than 2.2 days, less than 2.1 days, less than 2.0 days, less than 46 hours, less than 42 hours, less than 38 hours, less than 35 hours, less than 32 hours). In some embodiments, within 7 days of culture, the expanded population of γδ T cells (e.g., the expanded population of νδΐ T cells and/or DN T cells) comprises at least 10-fold the number of γδ T cells relative to the separated population of γδ T-cells prior to expansion (e.g., at least 20-fold, at least 30-fold, at least 40-fold, at least 50-fold, at least 60-fold, at least 70-fold, at least 80-fold, at least 90-fold, at least 100-fold, at least 150-fold, at least 200-fold, at least 300-fold, at least 400-fold, at least 500-fold, at least 600-fold, at least 700-fold, at least 800-fold, at least 900-fold, at least 1,000-fold, at least 2,000-fold, at least 3,000-fold, at least 4,000-fold, at least 5,000-fold, at least 6,000-fold, at least 7,000-fold, or at least 8,000- fold the number of γδ T cells relative to the separated population of γδ T cells prior to expansion).
[0075] In some embodiments, within 14 days of culture, the expanded population of γδ T cells (e.g., the expanded population of V51 T cells and/or DN T cells) comprises at least 20-fold the number of γδ T cells relative to the separated population of γδ T cells prior to expansion (e.g., at least 30-fold, at least 40-fold, at least 50-fold, at least 60-fold, at least 70-fold, at least 80-fold, at least 90-fold, at least 100-fold, at least 150-fold, at least 200-fold, at least 300-fold, at least 400-fold, at least 500-fold, at least 600-fold, at least 700-fold, at least 800-fold, at least 900-fold, at least 1,000-fold, at least 2,000-fold, at least 3,000-fold, at least 4,000-fold, at least 5,000-fold, at least 6,000-fold, at least 7,000-fold, at least 8,000-fold, at least 9,000- fold, or at least 10,000-fold the number of γδ T cells relative to the separated population of γδ T cells prior to expansion). In some embodiments, within 21 days of culture, the expanded population of γδ T cells (e.g., the expanded population of νδΐ T cells and/or DN T cells) comprises at least 50-fold the number of γδ T cells relative to the separated population of γδ T cells prior to expansion (e.g., at least 60- fold, at least 70-fold, at least 80-fold, at least 90- fold, at least 100-fold, at least 150-fold, at least 200-fold, at least 300-fold, at least 400-fold, at least 500-fold, at least 600-fold, at least 700-fold, at least 800-fold, at least 900-fold, at least 1,000-fold, at least 2,000-fold, at least 3,000-fold, at least 4,000-fold, at least 5,000-fold, at least 6,000-fold, at least 7,000-fold, at least 8,000-fold, at least 9,000-fold, or least 10,000- fold the number of γδ T cells relative to the separated population of γδ T cells prior to expansion).
[0076] In some embodiments, within 28 days of culture, the expanded population of γδ T cells (e.g., the expanded population of νδΐ T cells and/or DN T cells) comprises at least 100-fold the number of γδ T cells relative to the separated population of γδ T cells prior to expansion (e.g., at least 110-fold, at least 120-fold, at least 130-fold, at least 140-fold, at least 150-fold, at least 200-fold, at least 300-fold, at least 400-fold, at least 500-fold, at least 600-fold, at least 700-fold, at least 800-fold, at least 900-fold, at least 1,000-fold, at least 2,000-fold, at least 3,000-fold, at least 4,000-fold, at least 5,000-fold, at least 6,000-fold, at least 7,000-fold, at least 8,000-fold, at least 9,000-fold, at least 10,000-fold, at least 12,000-fold, or at least 15,000-fold the number of γδ T cells relative to the separated population of γδ T cells prior to expansion).
[0077] Non-hematopoietic tissue-derived γδ T cells (e.g., skin-derived γδ T cells and/or non- νδ2 T cells, e.g., νδΐ T cells and/or DN T cells) expanded by the methods provided herein can have a phenotype well-suited for anti-tumor efficacy. In some embodiments, the expanded population of γδ T cells (e.g., skin-derived νδΐ T cells) has a greater mean expression of CD27 than a reference population (e.g., the separated population of γδ T cells prior to the expansion step). In some embodiments, the expanded population of γδ T cells has a mean expression of CD27 that is at least 2-fold relative to the separated population of γδ T cells (e.g., at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8- fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 40-fold, at least 50-fold, at least 60-fold, at least 70-fold, at least 80-fold, at least 90-fold, at least 100-fold, at least 150-fold, at least 200-fold, at least 300-fold, at least 400-fold, at least 500-fold, at least 600-fold, at least 700-fold, at least 800-fold, at least 900- fold, at least 1,000-fold, at least 5,000-fold, at least 10,000-fold, at least 20,000-fold, or more, relative to the separated population of γδ T cells).
[0078] An increase or decrease in expression of other markers can be additionally or alternatively used to characterize one or more expanded populations of non-hematopoietic tissue-derived γδ T cells (e.g., skin-derived γδ T cells and/or ηοη-νδ2 T cells, e.g., νδΐ T cells and/or DN T cells), including CD124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1, CD31, KLRG1,CD30, CD2, NKp44, NKp46, ICAM- 2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1, and CD64. In some instances, the expanded population of γδ T cells (e.g., skin-derived γδ T cells and/or ηοη-νδ2 T cells, e.g., νδΐ T cells and/or DN T cells) has a greater mean expression of one or more of the markers selected from the group consisting of CD124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1, CD31 , KLRG1, CD30, and CD2, relative to the separated population of γδ T cells, e.g., prior to expansion. Additionally or alternatively, the expanded population of γδ T cells (e.g., skin-derived γδ T cells and/or non- νδ2 T cells, e.g., νδΐ T cells and/or DN T cells) may have a greater frequency of cells expressing one or more of the markers selected from the group consisting of CD 124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1, CD31, KLRG1, CD30, and CD2, relative to the separated population of γδ T cells. In some embodiments, the expanded population of γδ T cells (e.g., skin-derived γδ T cells and/or non- V52 T cells, e.g., V51 T cells and/or DN T cells) has a lower mean expression of one or more of the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1, and CD64, relative to the separated population of γδ T cells. The expanded population may similarly have a lower frequency of cells expressing one or more of the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1, and CD64, relative to the separated population of γδ T cells.
[0079] A non-hematopoietic tissue-resident γδ T cell produced by the method of the invention may thus have one or more of the following properties: (i) displays the phenotype CD69high, TIM3high and CD281ow/absent; (ii) upregulates of one or more of CCR3, CD39, CDl lb, and CD9; (iii) produces IFN-γ in response to an NKG2D ligand in the absence of TCR agonists; (iv) produces IL-13 in the absence of TCR agonists; (v) produces one or more of IFN-γ, TNF- α and GM-CSF in response to TCR activation; (vi) produces no or substantially no IL-17 in response to TCR activation; (vii) grows in culture medium containing IL-2 without additional growth factors; (viii) displays a cytotoxic T cell response in the absence of TCR agonists; and/or (ix) displays selective cytotoxicity for tumor cells over normal cells.
[0080] In some instances, a non-hematopoietic tissue-resident γδ T cell produced by the method of the invention produces IL-13 in the absence of TCR agonists and/or produces IFN- γ in response to an NKG2D ligand in the absence of TCR agonists.
[0081] Numerous basal culture media suitable for use in the proliferation of γδ T cells are available, in particular complete media, such as AIM-V, Iscoves medium and RPMI-1640 (Life Technologies). The medium may be supplemented with other media factors, such as serum, serum proteins and selective agents, such as antibiotics. For example, in some embodiments, RPMI-1640 medium containing 2 mM glutamine, 10% FBS, 10 mM HEPES, pH 7.2, 1% penicillin-streptomycin, sodium pyruvate (1 mM; Life Technologies), nonessential amino acids (e.g. 100 μΜ Gly, Ala, Asn, Asp, Glu, Pro and Ser; IX MEM nonessential amino acids Life Technologies), and 10 μΙ/L β-mercaptoethanol. Conveniently, cells are cultured at 37°C in a humidified atmosphere containing 5% C02 in a suitable culture medium. The γδ T cells may be cultured as described herein in any suitable system, including stirred tank fermenters, airlift fermenters, roller bottles, culture bags or dishes, and other bioreactors, in particular hollow fiber bioreactors. The use of such systems is well-known in the art. General methods and techniques for culture of lymphocytes are well-known in the art.
[0082] The methods described herein can include more than one selection step, e.g., more than one depletion step. Enrichment of a T cell population by negative selection can be accomplished, e.g., with a combination of antibodies directed to surface markers unique to the negatively selected cells. One method is cell sorting and/or selection via negative magnetic immuno adherence or flow cytometry that uses a cocktail of monoclonal antibodies directed to cell surface markers present on the cells negatively selected.
[0083] In certain embodiments, disclosed herein are adoptive cell transfer (ACT) methods. In certain embodiments, the ACT methods comprise, T cell immunotherapy wherein subjects are infused with heterologous, autologous or allogeneic γδ T cells to treat disease. In certain aspects, the ACT methods comprise infusion of subjects of V52" γδ T cells to treat disease. In certain aspects, the ACT methods comprise infusion of subjects of V52" γδ T cells that have been expanded ex vivo to treat disease. In certain aspects, the ACT methods comprise infusion of subjects of V52" γδ T cells that have been expanded ex vivo to treat cancer. In certain embodiments, V52" γδ T cells are selected and expanded ex vivo to enrich for V52" γδ T cells from a mixed population of immune cells.
[0084] In certain embodiments of the ACT methods, γδ T cells are selected and/or engineered ex vivo to target specific antigens, such as tumor-associated antigens. In certain embodiments of the ACT methods, immune cells comprising γδ T cells are obtained from tissue of donor subjects. In certain embodiments, γδ T cells are obtained from non-hematopoietic tissue of donor subjects. In certain embodiments, γδ T cells are obtained from blood of donor subjects. In certain embodiments, tumor infiltrating lymphocytes ("TIL"s) comprising γδ T cells are obtained from donor subjects. In certain embodiments γδ T cells are expanded in culture and selected for antigen specificity without altering their native specificity. In certain T cell immunotherapy methods, T lymphocytes comprising γδ T cells obtained from the donor are engineered ex vivo by transduction with viral expression vectors, to express chimeric antigen receptors ("CAR"s) of predetermined specificity.
[0085] Concentrations of cytokines and growth factors can range from about 0.1 ng/mL to about 500 ng/mL, from about 1 ng/mL to about 200 ng/mL, from about 10 ng/ml to 100 ng/ml, depending on the type of cytokine and growth factor. Appropriate combinations and concentrations of cytokines are disclosed in WO 2017/072367, which is incorporated herein by reference in its entirety.
6.3.1.3. Additional agents
[0086] In some embodiments, the methods further comprise administering to the subject at least one additional agent in an amount sufficient to activate V52" γδ T cells. In select embodiments, the at least one additional agent is an agonist of DNAM-1.
6.3.1.4. Subjects
[0087] In various embodiments, the mammalian subject has cancer. In particular
embodiments, the cancer has high infiltration of γδ T cells. In a variety of these embodiments, the mammalian subject is a human patient.
[0088] In certain embodiments, the subject from whom γδ2" T cells are obtained for culture (donor) is the same individual as the ultimate recipient of the cells, i.e., an autologous donor. Autologous γδ T cells, which are removed and in some instances stored are transplanted back into the subject. In certain embodiments, the autologous γδ T cells are subject to additional treatments prior to transplantation back into the recipient.
[0089] In some embodiments, for example, autologous γδ T cells obtained from the subject are further treated or purified to remove any disease cells, such as cancer cells, prior to transplantation back into the subject. Such treatments and purification, also referred to as "purging," of cell preparations to remove disease cell can include, among others, use of antibodies directed against cell surface markers expressed in disease cells and anti-cancer (i.e., chemotherapeutic) treatments. In some embodiments, the autologous γδ T cells are obtained following treatment of the subject for the underlying disease, thereby reducing the risk of presence of disease cells in the autologous γδ T cell preparations. Also referred to as "in vivo purging," the subject can be treated with an antibody therapeutic targeting the disease or treated with anti-cancer agent that does not destroy or suppress the hematopoietic system prior to obtaining the γδ T cells for transplantation. In certain embodiments, the purged preparation of γδ T cells can be further purified, such as by FACS or affinity selection (e.g., chromatography), to select γδ T cells or νδ2" γδ T cells from other cells (i.e., other immune cells or diseased cells).
[0090] In certain embodiments, the donor subject is not the same individual as the ultimate recipient of the cells, i.e., a heterologous or allogeneic donor. In certain heterologous embodiments, the cells obtained from the donor subject are not screened or matched for the recipient. As γδ T cells are non-MHC restricted, they do not recognize a host into which they are transferred as foreign, which means that they are less likely to cause graft- versus-host disease. This means that they can be used "off the shelf and transferred into any recipient, e.g., for heterologous or allogeneic adoptive T cell therapy.
[0091] In certain allogeneic embodiments, allogeneic immune cells comprising γδ T cells are obtained from donors who are selected based on matching at loci of the human lymphocyte antigen (HLA) complex. In some embodiments, the allogeneic immune cells comprising γδ T cells can be further isolated or purified, and/or subject to further manipulation. In some embodiments, the allogeneic immune cells comprising γδ T cells are subject to additional treatments to expand a population of immune cells or manipulated by recombinant methods to introduce heterologous genes or additional functionality to the allogeneic immune cells prior to transplantation into the recipient subject.
[0092] γδ T cells obtained from a donor, either autologous, heterologous or allogeneic, can be subject to additional treatments prior to transplantation into a recipient subject. In some embodiments, the immune cells comprising γδ T cells are treated to expand the population of γδ T cells, for example by culturing in a suitable medium.
6.3.1.4.1. Selection of subjects with high γδ T cell
infiltration
[0093] In certain embodiments, the method further comprises the step of detecting γδ T cell infiltration, or νδ2" γδ T cell infiltration, of tissues in a mammalian subject and selecting subjects who have γδ T cell infiltrates.
[0094] In certain embodiments, γδ T cell infiltration or νδ2" γδ T cell infiltration is detected in tumors of a mammalian subject. Detection of γδ T cell or νδ2" γδ T-cells may be performed by any method known in the art for the detection of subpopulations of immune cells in tissue. T cell infiltration in tissues {e.g., tumor from tumor biopsies) can be determined by
determining the estimated fraction of γδ T cells to total leukocytes compared to a reference tissue such non-tumor tissue of a similar origin.
[0095] An increase or decrease in expression of other markers can be additionally or alternatively used to characterize γδ T cells from tissue or γδ T cells from tissue which have been expanded ex-vivo {e.g., skin-derived γδ T cells and/or ηοη-νδ2 T cells, e.g., νδΐ T cells and/or DN T cells), including CD124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1 , CD31 , KLRG1 , CD30, CD2, NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1 , and CD64. In some instances, the expanded population of γδ T cells (e.g., skin-derived γδ T cells and/or non-5 V52 T cells, e.g., V51 T cells and/or DN T cells) has a greater mean expression of one or more of the markers selected from the group consisting of CD124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1 , CD31 , KLRG1 , CD30, and CD2, relative to the separated population of γδ T cells, e.g., prior to expansion. Additionally or alternatively, the expanded population of γδ T cells (e.g., skin-derived γδ T cells and/or ηοη-νδ2 T cells, e.g., νδΐ T cells and/or DN T cells) may have a greater frequency of cells expressing one or more of the markers selected from the group consisting of CD124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1 , CD31 , KLRG1 , CD30, and CD2, relative to the separated population of γδ T cells. In some embodiments, the expanded population of γδ T cells (e.g., skin-derived γδ T cells and/or non- νδ2 T cells, e.g., νδΐ T cells and/or DN T cells) has a lower mean expression of one or more of the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1 , and CD64, relative to the separated population of γδ T cells. The expanded population may similarly have a lower frequency of cells expressing one or more of the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1 , and CD64, relative to the separated population of γδ T cells.
[0096] In certain embodiments, the composition is co-administered with an additional agent for treating cancer (e.g., an anti-cancer agent). In an aspect, the composition is coadministered with an as yet to be described therapeutic for treating cancer. In coadministration procedures, the agents may be administered concurrently or sequentially. In another aspect, agents described herein are administered prior to the other active agent(s). The pharmaceutical formulations and modes of administration may be any of, but not limited to, those described below. It is also anticipated that there could be two or more co-administered chemical agents, biological agents or radiation that may each be administered using different modes or different formulations. 6.3.1.4.2. Selection of subjects with high V62" γδ T cells in peripheral blood
[0097] In certain embodiments, the method further comprises the step of detecting V52" γδ in the blood of a mammalian subject, and selecting subjects who have high νδ2~γδ T cells in the blood. In particular embodiments, the method comprises determining the ratio of νδ1+ and νδ2+ in a blood sample from a potential mammalian subject. In particular embodiments, the method comprises determining the total number of νδ1+ and νδ2+ in a blood sample from a potential mammalian subject. In certain aspects, the method further comprises comparing the ratio of νδ1+ and νδ2+ in a blood sample from a subject with cancer to a healthy individual. In certain aspects, the method further comprises comparing the total number of νδ1+ and νδ2+ in a blood sample from a subject with cancer to a healthy individual.
6.4. Methods of treating cancer [0098] In another aspect, methods for treating cancers are presented. 6.4.1. Cancers with γδ T cell infiltration
[0099] In various embodiments, the cancer has high infiltration of γδ T cells. The methods comprise administering at least one TIGIT inhibitor to a patient with a cancer having high infiltration of γδ T cells, in an amount sufficient to activate tissue-infiltrating γδ T cells in the patient. In typical embodiments, the cancer-infiltrating γδ T cells are νδ2" cells, and the amount of TIGIT inhibitor is sufficient to activate νδ2" cells. In some embodiments, the amount of TIGIT inhibitor is sufficient to activate νδ1+ cells. In certain embodiments, the amount of TIGIT inhibitor is sufficient to activate double negative (DN) cells.
[00100] In various embodiments, the TIGIT inhibitor is an inhibitor as described in
Section 6.6. In currently preferred embodiments, the TIGIT inhibitor is an antibody. In certain of these embodiments, the antibody binds specifically to TIGIT.
[00101] In some embodiments, the method further comprises the earlier step of selecting for treatment a patient whose cancer shows high levels of γδ T cell infiltration.
[00102] In various embodiments, the γδ T cell infiltration is detected by analysis of a tumor biopsy.
[00103] In typical embodiments, the analysis comprises an assay selected from
immunohistochemistry, in situ hybridization, polymerase chain reaction, R A seq, and combinations thereof. See, for example, Gentles, et al., 2015, incorporated herein by reference in its entirety.
[0100] In some embodiments, the cancer shows high levels of V51+ or double negative (DN) cell infiltrate.
[0101] In certain embodiments, the method further comprises the prior step of detecting γδ T cell infiltration, or V52" γδ T cell infiltration, of tissues in a mammalian subject and selecting subjects who have γδ T cell infiltrates.
[0102] In certain embodiments, γδ T cell infiltration or V52" γδ T cell infiltration is detected in tumors of a mammalian subject. Detection of γδ T cell or νδ2" γδ T-cells may be performed by any method known in the art for the detection of subpopulations of immune cells in tissue. T cell infiltration in tissues (e.g., tumor from tumor biopsies) can be determined by
determining the estimated fraction of γδ T cells to total leukocytes compared to a reference tissue such non-tumor tissue of a similar origin.
[0103] An increase or decrease in expression of other markers can be additionally or alternatively used to characterize γδ T cells from tissue or γδ T cells from tissue which have been expanded ex-vivo (e.g., skin-derived γδ T cells and/or ηοη-νδ2 T cells, e.g., νδΐ T cells and/or DN T cells), including CD124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1, CD31, KLRG1, CD30, CD2, NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1, and CD64. In some instances, the expanded population of γδ T cells (e.g., skin-derived γδ T cells and/or non-5 νδ2 T cells, e.g., νδΐ T cells and/or DN T cells) has a greater mean expression of one or more of the markers selected from the group consisting of CD124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1, CD31, KLRG1, CD30, and CD2, relative to the separated population of γδ T cells, e.g., prior to expansion. Additionally or alternatively, the expanded population of γδ T cells (e.g., skin-derived γδ T cells and/or ηοη-νδ2 T cells, e.g., νδΐ T cells and/or DN T cells) may have a greater frequency of cells expressing one or more of the markers selected from the group consisting of CD 124, CD215, CD360, CTLA4, CDlb, BTLA, CD39, CD45RA, Fas Ligand, CD25, ICAM-1, CD31, KLRG1, CD30, and CD2, relative to the separated population of γδ T cells. In some embodiments, the expanded population of γδ T cells (e.g., skin-derived γδ T cells and/or non- νδ2 T cells, e.g., νδΐ T cells and/or DN T cells) has a lower mean expression of one or more of the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1, and CD64, relative to the separated population of γδ T cells. The expanded population may similarly have a lower frequency of cells expressing one or more of the markers selected from the group consisting of NKp44, NKp46, ICAM-2, CD70, CD28, CD103, NKp30, LAG3, CCR4, CD69, PD-1, and CD64, relative to the separated population of γδ T cells.
[0104] In certain embodiments, the TIGIT inhibitor is co-administered with an additional agent for treating cancer (e.g., an anti-cancer agent). In an aspect, the composition is coadministered with an as yet to be described therapeutic for treating cancer. In coadministration procedures, the agents may be administered concurrently or sequentially. In another aspect, agents described herein are administered prior to the other active agent(s). The pharmaceutical formulations and modes of administration may be any of, but not limited to, those described below. It is also anticipated that there could be two or more co-administered chemical agents, biological agents or radiation that may each be administered using different modes or different formulations.
6.4.2. Cancer patients with elevated V62" γδ T cells in peripheral blood
[0105] In various embodiments, the method of treating cancer comprises administering a therapeutically effective amount of a TIGIT inhibitor to a cancer patient who has been determined to have an elevated representation of V51+ cells in a sample of peripheral blood.
[0106] In some embodiments, the method further comprising the prior step of determining the representation of V51+ cells in the peripheral blood sample. In some embodiments, the method further comprises the step, after determining the representation of V51+ cells and before administering the TIGIT inhibitor, of selecting the patient for TIGIT inhibitor treatment if the patient's sample has been determined to have an elevated representation of V51+ cells.
[0107] In particular embodiments, the method comprises determining the total number of V51+ and V52+ in a blood sample from a potential mammalian subject. In particular embodiments, the method comprises determining the ratio of V51+ and V52+ in a blood sample from a potential mammalian subject. In certain embodiments, the method further comprises comparing the total number of V51+ and V52+ in a blood sample from a subject with cancer to a healthy individual. In certain embodiments, the method further comprises comparing the ratio of V51+ and V52+ in a blood sample from a subject with cancer to a healthy individual. [0108] In some embodiments, the elevated representation is an increase in V51+ cells as a percentage of total lymphocytes in the sample. In certain embodiments, greater than 0.2% of total lymphocytes in the sample are determined to be V51+ cells. In particular embodiments, greater than 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9% or 1.0% of total lymphocytes in the sample are determined to be V51+ cells. In some embodiments, greater than 1.5%, 2.0%, 2.5%), 3.0%), 3.5%), 4.0%), or 5.0% of total lymphocytes in the sample are determined to be V51+ cells.
[0109] In some embodiments, the elevated representation is an increase in V51+ cells as a percentage of total γδ T cells in the sample. In certain embodiments, greater than 10% of total γδ T cells in the sample are V51+ cells. In particular embodiments, greater than 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, or 20% of total γδ T cells in the sample are V51+ cells.
[0110] In some embodiments, the elevated representation is an increase in the ratio of V51+ cells to V52+ cells in the sample. In certain embodiments, the ratio of V51+:V52+ cells is greater than 1 :9. In particular embodiments, the ratio of V51+:V52+ cells is greater than 1 :8, 1 :7, 1 :6, or 1 :5.
[0111] In typical embodiments, the TIGIT inhibitor is an antibody or antigen-binding antibody fragment that binds specifically to TIGIT.
6.4.3. Additional therapeutic agents
[0112] In various embodiments, the method further comprises administering at least one additional anti-cancer therapy. In some embodiments, at least one of the at least one additional anti-cancer therapy is a non-TIGIT checkpoint inhibitor. In particular
embodiments, the non-TIGIT checkpoint inhibitor is an antibody that binds specifically to PD- 1. In particular embodiments, the non-TIGIT checkpoint inhibitor is an antibody that binds specifically to PDL 1.
[0113] In various embodiments, the cancer is a solid, non-hematopoietic, cancer. In particular embodiments, the cancer is a breast cancer.
[0114] In various embodiments, the cancer is a hematological cancer.
[0115] In various embodiments, the method further comprises administering at least one additional therapeutic agent. [0116] In some embodiments, the at least one additional therapeutic agent is an agonist of DNAM-1.
[0117] In some embodiments, the at least one or more additional agent is an anti-cancer agent. In certain embodiments, the anti-cancer agent is selected from the group consisting of radiation, a chemotherapeutic or growth inhibitory agent, a targeted therapeutic agent, a small molecule inhibitor, a T cell expressing a chimeric antigen receptor, an antibody or antigen- binding fragment thereof, an antibody-drug conjugate, an angiogenesis inhibitor, an
antineoplastic agent, a cancer vaccine, an adjuvant, and combinations thereof. In particular embodiments, the anti-cancer agent is a chemotherapeutic agent. In particular embodiments, the anti-cancer agent is a targeted therapeutic agent. In certain embodiments, the anti-cancer agent is an antibody.
[0118] In various embodiments, the anti-cancer agent is administered continuously. In some embodiments, the anti-cancer agent is administered intermittently.
[0119] In some embodiments, the TIGIT inhibitor is administered before the anti-cancer agent. In some embodiments, the TIGIT inhibitor is administered simultaneous with the anticancer agent. In some embodiments, the TIGIT inhibitor is administered after the anti-cancer agent.
[0120] In some embodiments, the additional therapeutic agent is a checkpoint inhibitor.
6.5. Methods of increasing the number of tissue-infiltrating γδ T cells in a
mammalian subject
[0121] In another aspect, methods of increasing the number of tissue-infiltrating γδ T cells in a mammalian subject recipient are presented. The methods comprise (z) obtaining γδ T cells from tissue of a donor, (ii) expanding the γδ T cells ex vivo, {Hi) transplanting the expanded γδ T cells into a recipient, and (iv) administering at least one TIGIT inhibitor to the recipient.
[0122] In typical embodiments, the obtained and/or expanded γδ T cells are νδ2". In certain embodiments, the νδ2" cells are νδ1+ or DN T cells. In certain embodiments, νδ2" cells comprise both νδ1+ and DN T cells.
[0123] In some embodiments, the γδ T cells are obtained from hematopoietic tissue described herein in Section 6.3.1.1. [0124] In some embodiments, the γδ T cells are obtained from non-hematopoietic tissue, described herein in Section 6.3.1.2.
[0125] In typical embodiments, the TIGIT inhibitor is an inhibitor described herein in Section 6.6.
[0126] In some embodiments, the ex vivo expanded V52" γδ T cells were obtained by:
culturing lymphocytes obtained from non-hematopoietic tissue of humans or non-human animals in the presence of interleukin-2 (IL-2) and/or interleukin-15 (IL-15), and not in direct contact with stromal or epithelial cells during culture. In various embodiments, the methods culture and expansion methods are selected from those described in Section 6.3 above.
6.6. TIGIT Inhibitors
[0127] In various embodiments of the methods described herein, the TIGIT inhibitor is selected from an antagonist of TIGIT activity, an antagonist of TIGIT interaction with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL2, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL3, an agent that inhibits and/or blocks the intracellular signaling within a V52" γδ T cell mediated by PVR binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling within a V52" γδ T cell mediated by PVRL2 binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling within a V52" γδ T cell mediated by PVLR3 binding to TIGIT, and combinations thereof.
[0128] In some embodiments, the TIGIT inhibitor is an antagonist of TIGIT interaction with PVR. In certain embodiments, the TIGIT inhibitor is an agent that inhibits and/or blocks the interaction of TIGIT with PVR. The TIGIT inhibitor may block the interaction of PVR with TIGIT, leading to sequestration of TIGIT and increased binding of PVR to DNAM1. In particular embodiments, the TIGIT inhibitor is an antibody that binds specifically to TIGIT or to PVR. In particular embodiments, the TIGIT inhibitor is an antigen-binding fragment of an antibody that binds specifically to TIGIT or to PVR.
[0129] In certain embodiments, the TIGIT inhibitor is an antigen binding antibody fragment selected from Fab, Fab', F(ab')2, Fd , Fv, complementarity determining region (CDR) fragment, or single-chain antibody (scFv). In certain embodiments, the TIGIT inhibitor is a single domain antibody or single-chain antibody. In particular embodiments, the single domain antibody is a VHH domain.
[0130] In particular embodiments, the TIGIT inhibitor is an inhibitor described in
WO 2017/053748, WO 2016/073282, WO/2016/01 1264 or WO 2015/009856, the disclosures of which are incorporated herein by reference in their entireties. In particular embodiments, the TIGIT inhibitor is an inhibitor described in WO 2017/030823, WO 2016/028656,
WO 2016/106302, WO 2016/191643, WO 2017/059095, WO 2017/037707, of
WO 2017/048824, the disclosures of which are incorporated herein by reference in their entireties.
[0131] In certain aspects, the TIGIT inhibitor is a human antibody. In certain aspects, the human antibody has variable regions in which both the framework and CDR regions are sequences identical to those of human origin.
[0132] In certain aspects, the TIGIT inhibitor is a humanized antibody. In certain aspects, the humanized antibody is a non-human (e.g., murine) antibody that is a chimeric antibody and contains minimal sequence derived from non-human immunoglobulin. In one embodiment, the humanized antibody is a human immunoglobulin (recipient antibody) in which residues from a hypervariable region (HVR) of the recipient are replaced by residues from a HVR of a non- human species (donor antibody) such as mouse, rat, rabbit, or nonhuman primate having the desired specificity, affinity, and/or capacity. In some instances, FR residues of the human immunoglobulin variable domain are replaced by corresponding non-human residues. These modifications may be made to further refine antibody performance. Furthermore, in a specific embodiment, humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. In an embodiment, the humanized antibodies do not comprise residues that are not found in the recipient antibody or in the donor antibody. In general, a 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 FRs are those of a human immunoglobulin sequence. The humanized antibody optionally will also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. In an embodiment where the humanized antibodies do comprise residues that are not found in the recipient antibody or in the donor antibody, the Fc regions of the antibodies are modified. [0133] In certain aspects, the TIGIT inhibitor is a small molecule inhibitor. In certain aspects, the TIGIT inhibitor specifically binds TIGIT. In certain aspects, the TIGIT inhibitor binds PVR.
6.7. Dosage regimens
[0134] In the methods of activating V52" γδ T cells and methods of treating cancer described herein, various routes of administration of the TIGIT inhibitor and/or additional therapeutic agents are permissible.
[0135] Methods of delivery include, but are not limited to, intra-arterial, intra-muscular, intravenous, intranasal, and oral routes. Methods to enhance uptake can include, but are not limited to, encapsulation in liposomes, microparticles, microcapsules, receptor-mediated endocytosis, and the like. In specific embodiments, it may be desirable to administer the pharmaceutical compositions of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion during surgery, injection, or by means of a catheter. It is contemplated that the agents identified can be administered to subjects or individuals susceptible to or at risk of developing a variety of conditions benefiting from activation of γδ T cells. When the agent is administered to a subject, the agent can be added to a pharmaceutically acceptable carrier and systemically or topically administered to the subject.
[0136] Therapeutic amounts are empirically determined and vary with the pathology being treated, the subject being treated and the efficacy and toxicity of the agent. When delivered to an animal, the method is useful to further confirm efficacy of the agent.
[0137] In some embodiments, in vivo administration is effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and vary with the composition used for therapy, the purpose of therapy, the target cell being treated, and the subject being treated. Single or multiple administrations are carried out with the dose level and pattern being selected by the treating physician.
[0138] Suitable dosage formulations and methods of administering the agents are readily determined by those of skill in the art. Preferably, the compounds are administered at about 0.01 mg/kg to about 200 mg/kg, more preferably at about 0.1 mg/kg to about 100 mg/kg, even more preferably at about 0.5 mg/kg to about 50 mg/kg. When the compounds described herein are co-administered with another agent, the effective amount may be more or less than when the agent is used alone.
[0139] The pharmaceutical compositions can be administered orally, intranasally, parenterally or by inhalation therapy, and may take the form of tablets, lozenges, granules, capsules, pills, ampoules, suppositories or aerosol form. They may also take the form of suspensions, solutions and emulsions of the active ingredient in aqueous diluents, nonaqueous diluents, syrups, granulates or powders. In addition to an agent of the present invention, the
pharmaceutical compositions can also contain other pharmaceutically active compounds or a plurality of compounds of the invention.
[0140] More particularly, an agent of the present invention also referred to herein as the active ingredient, may be administered for therapy by any suitable route including, but not limited to, oral, rectal, nasal, topical (including, but not limited to, transdermal, aerosol, buccal and sublingual), vaginal, parental (including, but not limited to, subcutaneous, intramuscular, intravenous and intradermal) and pulmonary. It is also appreciated that the preferred route varies with the condition and age of the recipient, and the disease being treated.
[0141] Ideally, the agent should be administered to achieve peak concentrations of the active compound at sites of disease. This may be achieved, for example, by the intravenous injection of the agent, optionally in saline, or orally administered, for example, as a tablet, capsule or syrup containing the active ingredient.
[0142] Desirable blood levels of the agent may be maintained by a continuous infusion to provide a therapeutic amount of the active ingredient within disease tissue. The use of operative combinations is contemplated to provide therapeutic combinations requiring a lower total dosage of each component antiviral agent than may be required when each individual therapeutic compound or drug is used alone, thereby reducing adverse effects.
[0143] The present invention also includes methods involving co-administration of the TIGIT inhibitors described herein with one or more additional active agents. Indeed, it is a further aspect of this invention to provide methods for enhancing prior art therapies and/or pharmaceutical compositions by co-administering the agents of this invention. In coadministration procedures, the agents may be administered concurrently or sequentially. In one embodiment, the agents described herein are administered prior to the other active agent(s). The pharmaceutical formulations and modes of administration may be any of those described above. In addition, the two or more co-administered chemical agents, biological agents or radiation may each be administered using different modes or different formulations.
[0144] In certain embodiments of the methods provided herein, one or more agents provided herein and one or more therapeutic agents, TIGIT inhibitors, T-cell checkpoint inhibitors or anti-cancer agents are administered to a subject under one or more of the following
conditions: at different periodicities, at different durations, at different concentrations, by different administration routes, etc. In certain embodiments, the TIGIT inhibitor is
administered prior to an additional TIGIT inhibitor, T-cell checkpoint inhibitor, therapeutic or anti-cancer agent,, e.g., 0.5, 1 , 2, 3, 4, 5, 10, 12, or 18 hours, 1, 2, 3, 4, 5, or 6 days, or 1, 2, 3, or 4 weeks prior to the administration of therapeutic or anti-cancer agent. In certain embodiments, the TIGIT inhibitor is administered after an additional TIGIT inhibitor, T-cell checkpoint inhibitor, therapeutic or anti-cancer agent, e.g., 0.5, 1, 2, 3, 4, 5, 10, 12, or 18 hours, 1 , 2, 3, 4, 5, or 6 days, or 1, 2, 3, or 4 weeks after the administration of an additional TIGIT inhibitor, T-cell checkpoint inhibitor, therapeutic or anti-cancer agent. In certain embodiments, the TIGIT inhibitor and an additional TIGIT inhibitor, T-cell checkpoint inhibitor, therapeutic or anti-cancer agent, are administered concurrently but on different schedules, e.g., the TIGIT inhibitor is administered daily while the additional TIGIT inhibitor, T-cell checkpoint inhibitor, therapeutic or anti-cancer agent, is administered once a week, once every two weeks, once every three weeks, or once every four weeks. In other embodiments, the TIGIT inhibitor is administered once a week while the additional TIGIT inhibitor, T-cell checkpoint inhibitor, therapeutic or anti-cancer agent is administered daily, once a week, once every two weeks, once every three weeks, or once every four weeks.
[0145] In certain embodiments TIGIT inhibitors are administered in combination with an additional T-cell checkpoint inhibitor. In certain aspects, the T-cell checkpoint inhibitor or checkpoint inhibitor is any molecule that inhibits the activity of a T-cell checkpoint. In certain aspects the T-cell checkpoint inhibitor inhibits a protein or gene that functions to inhibit or suppress the activation of a T-cell. In certain aspects, the T-cell checkpoint inhibitor inhibits the expression or activity of, but not limited to: A2AR, B7-H3, B7-H4, BTLA, CTLA-4, IDO, KIR, LAG3, PD-1, TIM-3, VISTA and combinations thereof.
[0146] In certain embodiments TIGIT inhibitors are administered in combination with anticancer agents. A number of suitable therapeutic or anticancer agents are contemplated for use in the methods provided herein. Indeed, the methods provided herein can include but are not limited to, administration of numerous therapeutic agents such as: agents that induce apoptosis; polynucleotides (e.g., anti-sense, ribozymes, siR A); polypeptides (e.g., enzymes and antibodies); biological mimetics; alkaloids; alkylating agents; antitumor antibiotics; antimetabolites; hormones; platinum compounds; monoclonal or polyclonal antibodies (e.g., antibodies conjugated with anticancer drugs, toxins, defensins), toxins; radionuclides;
biological response modifiers (e.g., interferons (e.g., IFN-a) and interleukins (e.g., IL-2)); adoptive immunotherapy agents; hematopoietic growth factors; agents that induce tumor cell differentiation (e.g., all-trans-retinoic acid); gene therapy reagents (e.g., antisense therapy reagents and nucleotides); tumor vaccines; angiogenesis inhibitors; proteasome inhibitors: NF- KB modulators; anti-CDK compounds; HDAC inhibitors; and the like. Numerous other examples of therapeutic agents such as chemotherapeutic compounds and anticancer therapies suitable for co-administration with the disclosed compounds are known to those skilled in the art.
6.8. Pharmaceutical compositions
[0147] The TIGIT inhibitors for use in the methods described herein can be formulated in pharmaceutical compositions. The ex vivo cultured, and optionally expanded, γδ T cells for use in certain of methods described herein may formulated as a medicament.
[0148] Pharmaceutical compositions comprising TIGIT inhibitors can comprise, in addition to one or more of the TIGIT inhibitors, one or more additional checkpoint inhibitors, and a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient. The precise nature of the carrier or other material can depend on the route of administration, e.g. oral, intravenous, cutaneous or subcutaneous, nasal, intramuscular, intraperitoneal routes.
[0149] Pharmaceutical compositions for oral administration can be in tablet, capsule, powder or liquid form. A tablet can include a solid carrier such as gelatin or an adjuvant. Liquid pharmaceutical compositions generally include a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol can be included. [0150] For intravenous, cutaneous or subcutaneous injection, or injection at the site of affliction, the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen- free and has suitable pH, isotonicity and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringer's Injection.
Preservatives, stabilizers, buffers, antioxidants and/or other additives can be included, as required.
[0151] The pharmaceutically useful compound according to the present invention that is to be given to an individual, administration is preferably in a "therapeutically effective amount" or "prophylactically effective amount"(as the case can be, although prophylaxis can be considered therapy), this being sufficient to show benefit to the individual. The actual amount administered, and rate and time-course of administration, will depend on the nature and severity of the cancer being treated. Prescription of treatment, e.g. decisions on dosage etc, is within the responsibility of general practitioners and other medical doctors, and typically takes account of the disorder to be treated, the condition of the individual patient, the site of delivery, the method of administration and other factors known to practitioners.
[0152] A composition can be administered alone or in combination with other treatments (e.g., additional TIGIT inhibitors, T-Cell checkpoint inhibitors and/or anti-cancer agents), either simultaneously or sequentially dependent upon the cancer to be treated.
7. EXAMPLES
[0153] Below are examples of specific embodiments for carrying out the present invention. The examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperatures, etc.), but some experimental error and deviation should, of course, be allowed for.
[0154] The practice of the present invention will employ, unless otherwise indicated, conventional methods of protein chemistry, biochemistry, recombinant DNA techniques and pharmacology, within the skill of the art. Methods
T cell isolation from Human Peripheral Blood
[0155] Peripheral Blood (PB) from healthy volunteers was used to isolate Peripheral Blood Mononuclear Cells (PBMCs) by layering whole blood onto Ficoll (Ficoll-Paque PLUS, GE Healthcare Life Sciences) followed by centrifugation at 400 g for 20 minutes to separate red blood cells, blood plasma, and white lymphocytes/monocytes. White blood cells were carefully harvested through a stripette and washed four times in cold PBS.
The PBMCs were used either immediately for immunostaining and phenotypic analysis of PB- derived γδ+ T cells or cultured to have PB-derived γδ+ T cell line for functional assays. To this second aim, cells were resuspended in RPMI-1640 medium (Life Technologies) with 10% heat-inactivated foetal bovine serum (Life Technologies), L-glutamine (292 μg/ml; Life Technologies), penicillin (100 units/ml; Life Technologies), streptomycin (100 μg/ml; Life Technologies), N-2-hydroxyethylpiperazine-N-2-ethane sulfonic acid (HEPES; 0.01 M; Life Technologies), sodium pyruvate (1 mM; Life Technologies), minimal essential media (MEM) non-essential amino acids (IX; Life Technologies) at a density of 1 x 106 per mL and supplemented with human recombinant IL-2 (100 IU/ml; PROLEUKIN®; Novartis
Pharmaceutical UK Ltd) and human recombinant IL-15 (20 ng/ml; Biolegend). Cells were transferred into a 24 well plate that was coated with a-pan γδ TCR monoclonal Antibody (mAb) (20 μg/ml; clone Bl; Biolegend) 90 minutes prior to cell transfer. Cells were grown for 14 days, at 37°C and 5% C02, media changed ever 2-3 days and fresh cytokines added. Upon reaching confluence, cells were split 1 : 1. Under these conditions, after 14 days, the original minor population of γδ+ T cells is normally highly activated through their TCR (as indicated by upregulation if CD69 and CD25) and largely enriched consisting of mainly νδ2+ T cells but also νδΓ T cells (up to 30% of all γδ+ T cells).
[0156] When cell counts were required, lymphocytes were counted by either; (1) trypan blue stain (0.4%) (Life Technologies) and haemocytometer, or (2) CASY® Model TT cell counter and analyzer (Roche).
Isolation of non-haematopoietic tissue-resident T cells from Human Skin [0157] A three-dimensional skin explant protocol was established using the Clark protocol. Cellfoam Matrices (Cytomatrix Pty Ltd) having dimensions of 9 mm x 9 mm x 1.5 mm, were autoclaved and incubated in a solution of 100 mg/ml rat tail collagen I (BD Biosciences) in PBS for 30 minutes at room temperature, followed by one rinse in PBS. Samples of adult human skin were obtained within 3-6 hours of cutaneous surgery. Subcutaneous fat was removed and the remaining skin tissue was minced into fragments measuring approximately 1 mm x 1 mm. Approximately five skin fragments/explants were placed and pressed down onto the surface of each matrix. Each matrix was placed into a separate well of a 24-well plate (Corning) containing 2 ml of Skin-T media (Iscove's Modified Dulbecco's Medium (IMDM; Life Technologies) with 10% heat-inactivated fetal bovine serum (Life
Technologies), L-glutamine (292 μg/ml; Life Technologies), penicillin (100 units/ml; Life Technologies), streptomycin (100 μg/ml; Life Technologies), N-2-hydroxyethylpiperazine-N- 2-ethane sulfonic acid (HEPES; 0.01 M; Life Technologies), sodium pyruvate (1 mM; Life Technologies), minimal essential media (MEM) non-essential amino acids (lx; Life
Technologies) and 3.5 μΙ/L 2-mercaptoethanol (Life Technologies). For the first 7 days of culture Amphotericin (2.5 μg/ml; Life Technologies) was added to the media. The culture was kept at 37°C and 5% CO2 and media were refreshed three times per week by aspirating the upper 1 mL of media from each well and adding 1 ml of fresh medium. Human recombinant IL-2 (100 IU/mL; PROLEUKIN®; Novartis Pharmaceutical UK Ltd) and human recombinant IL-15 (20 ng/ml; Biolegend) were added at the initiation of culture and on each media refresh until the isolation of lymphocytes after 21-35 days. Up to 96 wells (four 24-well plates) were set up in culture for each donor.
[0158] To isolate the lymphocytes, the matrices and media were transferred to a 50 ml centrifuge tube (Corning) containing 10 ml Hanks Balanced Salt Solution (HBSS; Life Technologies) with 0.01 mM HEPES (up to 12 matrices/tube). The matrices were rinsed with the cell suspension using a 10 ml pipette, and the cell suspension was passed through a 70 μιη filter (BD Biosciences) into a fresh 50 ml centrifuge tube. The rinsing of the matrices was repeated two additional times. The media from the culture well was also aspirated and passed through a 70 μιη filter into a fresh 50 ml centrifuge tube. The wells were washed two further times with 1 ml of 0.01 mM HEPES/HBSS and passed through a 70 μιη filter. Cells were subsequently isolated by centrifugation (1600 rpm for 15 minutes). The pellet was re- suspended in Skin-T media. The final cell pellet was re-suspended in Skin-T media for subsequent flow cytometry analysis or functional studies. When cell counts were required, lymphocytes were counted at this stage by either; (1) trypan blue stain (0.4%) (Life
Technologies) and haemocytometer, or (2) CASY® Model TT cell counter and analyzer (Roche).
γδ+ T cell phenotypic analysis
[0159] Lymphocytes (0.5xl06 cells/well) were plated in a 96-U bottom well plate and washed in PBS. First, a live/dead staining was done with the Fixable Blue Dead Cell Stain Kit (Life Technologies) or the Fixable Viability Dye eFluor 770 NIR (eBioscience) according the manufacture instructions. Then the cells were washed in FACS buffer (PBS, 0.5%> BSA, 2 mM EDTA) and stained in ice with a mix of the following mAbs: BV510 conjugated a-CD3 (clone OKT3; Biolegend), PC7-conjugated a-pan γδ (IMMU510 clone; Beckman Coulter), FITC- conjugated a-V51 (TS8.2 clone; Life Technologies), PE-conjugated a-V52 (B6 clone;
Biolegend), APC-conjugated a-DNAMl (11A8 clone; Biolegend), PerCP eFluor710- conjugated a-TIGIT (MBSA43 clone; eBioscience). Finally, the cells were fixed in fixation buffer (eBioscience), resuspended in FACS buffer and acquired by a FACS Fortessa machine. Data were analysed through the Flowjo 9.9 Software.
T cell cytokine production assay
[0160] PB-derived γδ+ T cell line or Skin-derived lymphocytes or sorted pan γδ+ T cells were plated in a 96-flat bottom well plate with a density of 0.2x106 cells/well. The plate was previously coated with different combination of the following proteins: a-CD3 Ab (5 μg/ml) (HIT3a clone; Biolegend) or its isotype control (MOPC-173; Biolegend), recombinant human PVR Fc Chimera Protein (rhPVR) (20 μg/ml) (Life Technologies), recombinant human MICA Fc Chimera Protein (rhMICA) (10 μg/ml) (R&D Systems) or their hlgGl control (Biolegend). The blocking a-DNAMl Ab (DX11 clone; BD Biosciences) was added to the cell suspension (10 μg/ml). The cells were stimulated for 6 hours in RPMI 10%> FCS supplemented with the intracellular protein transport inhibitor Brefeldin A (20 μg/ml)(Sigma-Aldrich), at 37°C and 5% C02. [0161] At the end of the stimulation, the cells were harvested and moved to a 96-U bottom well plate, extracellular stained and fixed as previously described for the phenotypic analysis. The intracellular staining was performed in Permeabilization Buffer (eBioscience) according the manufacture instructions and the cytokine production was assessed using the following mAbs: eFluor450-conjugated α-IFNy (4S.B3; eBioscience) APC-conjugated a-TNFa
(MAbl 1; Bio legend). The cells were resuspended in FACS buffer and acquired by a FACS Fortessa machine. Data were analysed through the Flowjo 9.9 Software.
T cell proliferation assay
[0162] Pan γδ+ T cells were purified from Skin-derived lymphocytes using a a-pan γδ mAb (IMMU510 clone; Beckman Coulter) and a FACS Aria cell sorting. They were then stained with the CellTrace Violet (CTV) Cell Proliferation Kit (Life Technologies) and cultured in a 96-flat bottom well plate for 1 week in complete medium supplemented with IL-2. The plate was previously coated with different combination of the following proteins: a-CD3 Ab (50 ng/ml) (HIT3a clone; Biolegend) or its isotype control (MOPC-173; Biolegend), rhPVR (10 μg/ml) (Life Technologies) or its hlgGl control (Biolegend). At the end of the stimulation, the cells were harvested and moved to a 96-U bottom well plate, extracellular stained and fixed as previously described for the phenotypic analysis. The cells were resuspended in FACS buffer and acquired by a FACS Fortessa machine. Data were analysed through the Flowjo 9.9 Software.
Expansion of skin-derived yd T cells
[0163] Mixed lymphocytes were harvested after three to four weeks of scaffold culture, washed in PBS, spun down, and re-suspended in Roswell Park Memorial Institute 1640 medium (RPM1-1640; Life Technologies) with filtered 10% heat-inactivated fetal bovine serum (Life Technologies), L-glutamine (292 μg/ml; Life Technologies), penicillin (100 units/ml; Life Technologies), streptomycin (100 μg/ml; Life Technologies), N-2- hydroxyethylpiperazine-N-2-ethane sulfonic acid (HEPES; 0.01 M; Life Technologies), sodium pyruvate (1 mM; Life Technologies), minimal essential media (MEM) non-essential amino acids (IX; Life Technologies) and 50 μΜ 2-mercaptoethanol (Life Technologies) at a concentration of 1 x 106 cells/ml. The initial population of V51+ cells was 1.12% of lymphocytes. Cells were seeded at 2 x 105 cells/well into 96 well flat bottom plates (Corning) or at 2 x 106 cells/well into 24 well plates (Corning) for expansion and supplemented with factors at the following concentrations: IL-2 (100 U/ml), IL-15 (10 ng/ml), IL-4 (5 ng/ml) and IL-21 (10 ng/ml). Cells were monitored daily by microscopy and provided with fresh media and cytokines three times per week. Upon full confluence and cell aggregation, cells were split 1 : 1 into additional wells and plates, as required. After 21 days, cells were harvested using ACCUTASE® (eBioscience), counted, and analyzed using flow cytometry.
Example 1: γδ+ T cell subpopulations in human PBMC and skin
[0164] Within the human adaptive, cell based immune system, there are 3 types of
lymphocytes using somatic gene rearrangement to generate diverse surface receptors, B cells, αβ T cells and γδ T cells respectively. Only γδ T cells show compartmentalization, with V52 TCR positive cells dominating the human blood whilst T cells recombining other TCR genes (eg. V51, 53, 54, 55, 56, 57, 58) are predominantly found in human tissues such as the skin, the gut, the lungs, the liver and others (Figure 1 A).
[0165] Peripheral blood mononuclear cells from 11 donors and human skin derived lymphocytes from 27 donors were isolated as described above. Flow cytometry was performed using the following antibody-fluorochrome conjugates: V51-FITC, CD3-BV510 and V52-PE. Samples were also stained for viability using the eFluor 770 NIR live/dead dye. All T lymphocytes present were gated using the marker CD3, and within this population the presence of V52+ γδ T cells or V51+ γδ T cells made visible using the according antibodies. The results show that V52 TCR positive cells are dominating in the human blood with almost no V51 T cells being present. In human skin, the situation is inverted with V51 TCR positive cells being the signature γδ T cell population and almost no V52 TCR positive cells can be found (Figure IB). Example 2: Human V51+/DN and V52+ T cells have very distinct expression patterns of DNAM1 and TIGIT
[0166] PB-derived γδ+ T cells and Skin-derived γδ+ T cells were tested by FACS analysis for the presence of DNAM1 and TIGIT on their surface. On a first analysis, DNAM1 and TIGIT expression was evaluated on a pan γδ+ T cell population. DNAMl was expressed by both PB- derived γδ+ T cells and Skin-derived γδ+ T cells. As was reported for αβ T cells, ΡΒ-γδ+ T cells expressed TIGIT only upon activation, obtained in this case with PHA treatment. On the contrary, TIGIT was already expressed by unstimulated Skin-derived γδ+ T cells.
[0167] In a second step, DNAMl and TIGIT expression was assessed by staining PB-derived and Skin-derived lymphocytes with anti-νδΐ and anti-V02 Abs, in addition to the pan γδ Ab. This staining strategy allowed to gate three different populations into the pan γδ+ gate: νδ1+, νδ2+ and V61"V62" (named here Double Negative, DN). Like previously said, ΡΒ-γδ+ T cells are mostly represented by the νδ2+ population, and in a smaller percentage by νδ1+ or DN. The νδ2+ population largely expressed DNAMl, more than 70% in all donors, while only a small proportion, less than 25%, or none of them expressed TIGIT. The νδ1+ and DN populations present in the PB expressed DNAMl, even if to a lower level compare to the νδ2+ population, and shared TIGIT constitutive expression as the Skin-derived γδ+ T cells. Indeed, Skin-derived γδ+ T cells are composed by νδ1+ and DN cells, and even in this second more detailed staining, we confirmed that νδ1+ and DN cells constitutively expressed
DNAMl and TIGIT. Specifically, DNAMl is expressed by more than 70% of νδ and DN cells in all donors, and TIGIT is expressed by more than 50%> of νδ1+ cells. DN T cells showed a more variable TIGIT expression because it is a heterogeneous population and it is represented by populations expressing TIGIT and populations not expressing TIGIT, in a different proportion in every donor. Figure 3 shows γδ T cells derived either from PBMCs or skin tissue express DNAMl . γδ T cells isolated from human PBMCs (Figure 4) and lymphocytes isolated from human skin tissue (Figure 5) were stained with anti-TIGIT antibodies and antibodies specific for either the γ chain or δ chain TCR and subjected to FACS analysis. Only 1.51% of the δ T cells isolated from unstimulated PBMCs expressed TIGIT, whereas 30.3% of δ T cells isolated from PBMCs stimulated with phytohaemagglutinin (PHA) expressed TIGIT (Figure 4). However, nearly all of the γδ T cells isolated from unstimulated skin leukocytes expressed TIGIT (Figure 5). γδ T cells isolated from human PBMCs (Figures 6 and 7) and lymphocytes isolated from human skin tissue (Figures 8 and 9) were stained for DNAM1, TIGIT, γ chain or δ chain TCR, V51 , and V52 and subjected to FACS analysis. V52+ γδ T cells isolated form PBMCs expressed high levels of DNAM1 (Figure 6) and low levels of TIGIT (Figure 7) while V51+ γδ T cells expressed low levels of DNAMl (Figure 6) and high levels of TIGIT (Figure 7). DN cells (V5T V52") expressed moderate levels of DNAMl (Figure 8) and high levels of TIGIT. νδ γδ T cells and DN cells isolated from human skin leukocytes expressed high levels of DNAMl and TIGIT (Figures 8 and 9).
[0168] In summary, all γδ+ subpopulations express DNAMl . Skin-derived V51+ and DN T cells express higher DNAMl compare to PB-derived V51+ and DN T cells, very likely for the natural presence of IL15 in the skin that upregulates DNAMl . Of note, V51+ and DN T cells, in PB and Skin, already express TIGIT in unstimulated conditions, while V52+ T cells do not. .
Example 3: aCD3-induced cytokine production is inhibited by PVR in V51+ and DN νδ T cells.
[0169] To confirm TIGIT functionality in suppressing TCR stimulation in DN and V51+ γδ T cells, soluble PVR was added to aCD3 -stimulated DN and V51+ γδ T cells isolated from either human PBMCs (figure 10 and 11) or human skin lymphocytes (Figures 11). PB-derived and Skin-derived lymphocytes were stimulated for 6 hours by plate-bound aCD3 Ab in
combination with rhPVR or its isotype control, and cytokine production, specifically IFNy and TNFa, was assessed by intracellular staining and FACS analysis gating on V51+, V52+, or DN populations.
[0170] The cytokine production induced by TCR stimulation was strongly inhibited in V51+ and DN cells when rhPVR was also present, while the V52+ population, that is TIGIT", was unaffected. We didn't notice any difference whether we performed the assay with PB-derived or Skin-derived γδ T cells. Moreover, with 2 donors, we could perform the experiment with sorted pan γδ+ T cells and we confirmed the PVR-induced inhibition of the cytokine production, suggesting an intrinsic γδ+ T cell mechanism that doesn't need the presence of other immune cells. Figure 10 shows IFNy and TNFa production in a representative donor of PB-derived lymphocytes. Figure 11 shows collective plots for IFNy and TNFa production in γδ T cells derived from PB and Skin These results very strongly suggest that rhPVR engages TIGIT on νδ1+ and DN cells, triggering its inhibitory function and weakening their activation. Example 4: aCD3-induced proliferation is inhibited by PVR in V51+ and DN νδ T cells
[0171] Skin-derived γδ+ T cells, previously sorted and CTV-stained, were stimulated with plate-bound aCD3 Ab in combination with rhPVR or its isotype control. After 7 days of stimulation, the proliferation of V51+ and DN γδ T cells was evaluated by FACS analysis of the CTV dilution. Figures 12 and 13 show increased dye concentrations retained in a much higher percentage of DN and V51+ γδ T cells after exposure to PVR upon stimulation by aCD3 as compared to cells exposed to isotype control, demonstrating reduced cell proliferation in cells with higher dye concentrations. The aCD3-induced proliferation of V51 and DN γδ T cells was impaired in the presence of PVR compare to its control, further confirming the inhibitory role of the PVR/TIGIT axis on the TCR-induced activation of V51+ and DN T cells.
Example 5: PVR specifically inhibits TCR signaling
[00104] To confirm that PVR mediated inhibition of stimulation DN and V51+ γδ T cells was due specifically to inhibition of TCR signalling; skin-derived lymphocytes were stimulated by plate-bound isotype control or aCD3 Ab or MHC class I polypeptide-related sequence A (rhMICA), (ligand for the NKG2D receptor) in the presence of rhPVR or its control. After 6 hours of stimulation, V51+ and DN cell cytokine production was assessed by intracellular staining and FACS analysis gating on V51+, V52+, or DN populations. Like already demonstrated, aCD3 -induced IFNy and TNFa production is inhibited by the presence of PVR (Figure 14). In parallel, the same donor was stimulated with plate-bound MICA, an innate TCR-independent activator for V51+ and DN T cells. Of note, the presence of PVR does not inhibit MICA-induced cytokine production, rather increase the production of IFNy and TNFa by V51+ and DN T cells (Figures 15 and 16). Indeed, PVR, when is not in the context of a TCR stimulation, doesn't work as an inhibitor through TIGIT but works as a costimulator through DNAM1, as demonstrated by the stimulation with PVR only: V51+ and DN T cells are induced to produce, even if in a small amount, IFNy and TNFa and this production is reverted by a blocking aDNAMl mAb (Figures 15 and 16).
[00105] Production of another cytokine associated with TCR signaling, granulocyte- macrophage colony-stimulating factor (GMCSF), was also analyzed in response to PVR (Figure 17). In addition to ΙΚΝγ and TNFa, GMCSF also was inhibited by PVR, demonstrating that PVR is a general inhibitor of TCR stimulation.
Example 6: PVR inhibitory effect is lost on V51+ and V53+ cells not expressing
TIGIT
[00106] Tissue-derived V51+ and V53+ cells cultured and expanded ex-vivo in the presence of IL-2 and IL-15 (culture method 1) expressed high amounts of TIGIT, whereas V51+ and V53+ cells cultured in the presence of IL-2, IL-4, IL-15 and IL-21 (culture method 2), expressed low levels of TIGIT (Figure 18). FACS analysis of tissue-derived V51+ and V53+ cells from individual donors cultured in the presence of IL-2, IL-4, IL-15 and IL-21, demonstrated that PVR inhibition of INFy and TNFa production was lost in V51+ and V53+ cells expressing low levels of TIGIT (Figures 19 and 20).
[0172] In conclusion, the TCR stimulation is a strong activator of V51+ and DN T cells, in terms of production of inflammatory cytokines like IFNy and TNFa but also in terms of proliferation. On the other side, the TCR-induced activation of V51+ and DN T cells is normally kept under control by the PVR/TIGIT axis. Indeed TIGIT is constitutively expressed by PB- and Skin-derived V51+ and DN T cells. The inhibitory receptor TIGIT, like has been extensively demonstrated, has greater affinity for its ligand PVR compare to the activator receptor DNAM1. In the context of a TCR stimulation, PVR preferentially binds TIGIT and triggers its inhibitory signaling into the cells dampening the activation of V51+ and DN T cells. On the other hand, when not in a TCR activation context, PVR is available to bind the costimulatory receptor DNAM1 and help the activation of V51+ and DN T cells. By using molecules or mAbs blocking TIGIT, and its interaction with PVR and its other ligands PVRL2 and PVRL3, we could dampen its negative signalling and promote V51+ and DN T cell activation.
[0173] While the invention has been particularly shown and described with reference to a preferred embodiment and various alternate embodiments, it will be understood by persons skilled in the relevant art that various changes in form and details can be made therein without departing from the spirit and scope of the invention. [0174] All references, issued patents and patent applications cited within the body of the instant specification are hereby incorporated by reference in their entirety, for all purposes.
8. REFERENCES CITED
[0175] Gentles, A. J., et al, "The prognostic landscape of genes and infiltrating immune cells across human cancers"; Nature Medicine ; Vol 21; No. 8; August 2015; pp. 938-945.

Claims

1. A method of activating V52" gamma-delta T cells (γδ T cells) in a mammalian subject, comprising:
antagonizing TIGIT activity of V52" γδ T cells present in the subject to an extent sufficient to activate the V52" γδ T cells in vivo.
2. The method of claim 1, wherein the V52" γδ T cells are V51+ cells.
3. The method of claim 1, wherein the V52" γδ T cells are V51" V52" double negative (DN) cells.
4. The method of any one of claims 1 - 3, wherein antagonizing TIGIT activity comprises administering at least one TIGIT inhibitor to the subject.
5. The method of claim 4, wherein the at least one TIGIT inhibitor is administered adjunctively with V52" γδ T cells that have been cultured ex vivo.
6. The method of claim 5, wherein the at least one TIGIT inhibitor is added to the V52" γδ T cells during culturing ex vivo.
7. The method of claim 5 or 6, wherein the cultured V52" γδ T cells have been expanded ex vivo.
8. The method of any one of claims 5 - 7, wherein the at least one TIGIT inhibitor is administered in a composition comprising the cultured γδ T cells.
9. The method of any one of claims 5 - 7, wherein the at least one TIGIT inhibitor is administered separately from the ex vivo expanded γδ T cells.
10. The method of any one of claims 4 - 9, further comprising administering to the subject at least one additional agent in an amount sufficient to activate V52" γδ T cells.
11. The method of claim 10, wherein the at least one additional agent is an agonist of DNAM-1.
12. The method of any one of claims 4 - 11, wherein the at least one TIGIT inhibitor is selected from the group consisting of an antagonist of TIGIT activity, an antagonist of TIGIT interaction with Poliovirus receptor (PVR), an agent that inhibits and/or blocks the interaction of TIGIT with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL2, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL3, an agent that inhibits and/or blocks the intracellular signaling within a V52" γδ T cell mediated by PVR binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling within a V52" γδ T cell mediated by PVRL2 binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling within a V52" γδ T cell mediated by PVLR3 binding to TIGIT, and combinations thereof.
13. The method of claim 12, wherein the TIGIT inhibitor is an antibody.
14. The method of claim 13, wherein the antibody binds specifically to TIGIT.
15. The method of claim 13, wherein the antibody binds to PVR to specifically inhibit PVR interaction with TIGIT.
16. The method of any one of claims 13 - 15, wherein the antibody is a full length IgG antibody.
17. The method of any one of claims 13 - 16, wherein the antibody is an antigen-binding antibody fragment.
18. The method of any one of claims 13 - 16, wherein the antibody is a single domain antibody.
19. The method of any one of claims 13 - 19 wherein the antibody is fully human.
20. The method of any one of claims 13 - 20, wherein the antibody is humanized.
21. The method of claim 12, wherein the TIGIT inhibitor is a small molecule inhibitor.
22. The method of claim 7 or any one of claims 8 - 21 as depend from claim 7, wherein the ex vivo expanded V52" γδ T cells were obtained by: culturing lymphocytes obtained from non-hematopoietic tissue of humans or non- human animals in the presence of interleukin-2 (IL-2) and/or interleukin-15 (IL-15), and not in direct contact with stromal or epithelial cells during culture.
23. The method according to claim 22, wherein the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of IL-2.
24. The method according to claim 22 or 23, wherein the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of interleukin-15 (IL-15).
25. The method according to any one of claims 22 - 34, wherein the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non- hematopoietic tissue in the presence of IL-2 and IL-15.
26. The method according to any one of claims 22 - 25, wherein the culturing step comprises culturing the lymphocytes in the absence of TCR activation or co-stimulation signals.
27. The method according to any one of claims 22 - 26, wherein the culturing step comprises culturing the lymphocytes in the absence of a T cell receptor pathway agonist.
28. The method according to any one of claims 22 - 27, wherein the culturing step comprises culturing the lymphocytes in the absence of stromal or epithelial cells.
29. The method according to claim 28, wherein stromal or epithelial cells are removed prior to culture.
30. The method according to claim 28, wherein the lymphocytes are cultured in the absence of fibroblasts.
31. The method according to any one of claims 22 to 30, wherein the lymphocytes have been obtained from skin, the gastrointestinal tract (e.g., colon), mammary gland tissue, lung, liver, pancreas, prostate or reproductive tissue.
32. The method of any one of claims 22 - 31 , wherein the ex vivo expanded V52" γδ T cells were obtained by: culturing lymphocytes obtained from non-hematopoietic tissue of humans or non- human animals in the presence of IL-2, IL-15, and a factor selected from the group consisting ofIL-4, IL-21, IL-6, IL-7, IL-8, IL-9, IL-12, IL-18, IL-33, IGF-1, IL-Ιβ, human platelet lysate (HPL), and stromal cell-derived factor- 1 (SDF-1) for at least 5 days to produce an expanded population of γδ T cells.
33. The method of any one of claims 22 - 32, wherein, within 14 days of culture, the expanded population of γδ T cells comprises at least 20-fold the number of γδ T cells as the γδ T cells obtained from a non-hematopoietic tissue.
34. The method of claim 33, wherein, within 7 days of culture, the expanded population of γδ T cells comprises at least 2-fold the number of γδ T cells as the γδ T cells obtained from a non-hematopoietic tissue.
35. The method of any one of claims 22 - 34, wherein the expanded population of γδ T cells is at least 50% νδ1+ cells.
36. The method of claim 35, wherein the expanded population of γδ T cells is at least 70% νδ1+ cells.
37. The method of claim 36, wherein the expanded population of γδ T cells is at least 90%> νδ1+ cells.
38. The method of any one of claims 1 - 37, wherein the subject has cancer.
39. The method of claim 38, wherein the cancer has high infiltration of γδ T cells.
40. A method of treating cancers having high infiltration of γδ T cells, comprising: administering at least one TIGIT inhibitor to a patient with a cancer having high infiltration of γδ T cells, in an amount sufficient to activate tissue-infiltrating γδ T cells in the patient.
41. The method of claim 40, wherein the cancer-infiltrating γδ T cells are νδ2" cells.
42. The method of claim 40 or 41, wherein the νδ2" cells are νδ1+ or DN cells.
43. The method of any one of claims 40 - 42, wherein the at least one TIGIT inhibitor is selected from the group consisting of an antagonist of TIGIT activity, an antagonist of TIGIT interaction with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL2, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL3, an agent that inhibits and/or blocks the intracellular signaling within a V52" γδ T cell mediated by PVR binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling within a V52" γδ T cell mediated by PVRL2 binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling within a V52" γδ T cell mediated by PVLR3 binding to TIGIT, and combinations thereof.
44. The method of any one of claims 40 - 43, wherein the TIGIT inhibitor is an antibody.
45. The method of claim 44, wherein the antibody binds specifically to TIGIT.
46. The method of claim 44, wherein the antibody binds to PVR to specifically inhibit PVR interaction with TIGIT.
47. The method of any one of claims 44 - 46, wherein the antibody is a full length IgG antibody.
48. The method of any one of claims 44 - 46, wherein the antibody is an antigen-binding antibody fragment.
49. The method of any one of claims 44 - 46, wherein the antibody is a single domain antibody.
50. The method of any one of claims 44 - 49, wherein the antibody is fully human.
51. The method of any one of claims 44 - 49, wherein the antibody is humanized.
52. The method of any one of claims 40 - 43, wherein the TIGIT inhibitor is a small molecule inhibitor.
53. The method of any one of claims 40 - 52, further comprising the antecedent step of selecting for treatment a patient whose cancer shows high levels of γδ T cell infiltration.
54. The method of claim 53, wherein the γδ T cell infiltration is detected by analysis of a tumor biopsy by a method comprising an assay selected from the group consisting of immunohistochemistry, polymerase chain reaction, in situ hybridization, or combinations thereof.
55. The method of claim 53 or 54, wherein the γδ T cell infiltration comprises V51+ or double negative (DN) cells.
56. The method of any one of claims 40 - 55, wherein at least one additional therapeutic agent is administered.
57. The method of claim 56, wherein the at least one or more additional agent comprises an agonist of DNAM-1.
58. The method of claim 57, wherein the at least one or more additional agent comprises an anti-cancer agent.
59. The method of claim 58, wherein the anti-cancer agent is selected from the group consisting of radiation, a chemotherapeutic or growth inhibitory agent, a targeted therapeutic agent, a small molecule inhibitor, a T cell expressing a chimeric antigen receptor, an antibody or antigen-binding fragment thereof, an antibody-drug conjugate, an angiogenesis inhibitor, an antineoplastic agent, a cancer vaccine, an adjuvant, and combinations thereof.
60. The method of claim 59, wherein the anti-cancer agent is a chemotherapeutic.
61. The method of claim 59, wherein the anti-cancer agent is a small molecule inhibitor.
62. The method of claim 59, wherein the anti-cancer agent is an antibody.
63. The method of any one of claims 40 - 62, wherein the TIGIT inhibitor is administered continuously.
64. The method of any one of claims 40 - 62, wherein the TIGIT inhibitor is administered intermittently.
65. The method of any one of claims 58 - 64, wherein the anti-cancer agent is administered continuously.
66. The method of any one of claims 58 - 64, wherein the anti-cancer agent is
administered intermittently.
67. The method of any one of claims 58 - 64, wherein the TIGIT inhibitor is administered before the anti-cancer agent.
68. The method of any one of claims 58 - 64, wherein the TIGIT inhibitor is administered simultaneous with the anti-cancer agent.
69. The method of any one of claims 58 - 64, wherein the TIGIT inhibitor is administered after the anti-cancer agent.
70. A method of increasing the number of tissue infiltrating γδ T cells in a mammalian subject recipient, comprising:
obtaining γδ T cells from tissue comprising γδ T cells from a donor;
expanding the γδ T cells ex vivo;
transplanting the expanded γδ T cells into a recipient, and administering at least one TIGIT inhibitor to the recipient.
71. The method of claim 70, wherein the obtained and/or expanded γδ T cells are νδ2~.
72. The method of claim 71, wherein the νδ2" cells are νδ1+ or DN.
73. The method of any one of claims 70 - 72, wherein the at least one TIGIT inhibitor is selected from the group consisting of an antagonist of TIGIT activity, an antagonist of TIGIT interaction with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVR, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL2, an agent that inhibits and/or blocks the interaction of TIGIT with PVRL3, an agent that inhibits and/or blocks the intracellular signaling within a νδ2" γδ T cell mediated by PVR binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling within a νδ2" γδ T cell mediated by PVRL2 binding to TIGIT, an agent that inhibits and/or blocks the intracellular signaling within a νδ2" γδ T cell mediated by PVLR3 binding to TIGIT, and combinations thereof.
74. The method of any one of claims 70 - 73, wherein the at least one TIGIT inhibitor is an antibody.
75. The method of claim 74, wherein the antibody binds specifically to TIGIT.
76. The method of claim 74, wherein the antibody binds to PVR.
77. The method of any one of claims 74 - 76, wherein the antibody is a full length IgG antibody.
78. The method of any one of claims 74 - 76, wherein the antibody is an antigen-binding antibody fragment.
79. The method of any one of claims 74 - 76, wherein the antibody is a single domain antibody.
80. The method of any one of claims 74 - 79, wherein the antibody is fully human.
81. The method of any one of claims 74 - 79, wherein the antibody is humanized.
82. The method of any one of claims 70 - 73, wherein the at least one TIGIT inhibitor comprises a small molecule inhibitor.
83. The method of any one of claims 71 - 82, wherein the ex vivo expanded V52" γδ T cells were obtained by: culturing lymphocytes obtained from non-hematopoietic tissue of humans or non- human animals in the presence of interleukin-2 (IL-2) and/or mterleukin-15 (IL-15), and not in direct contact with stromal or epithelial cells during culture.
84. The method according to claim 83, wherein the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of IL-2.
85. The method according to claim 83 or 84, wherein the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non-hematopoietic tissue in the presence of interleukin-15 (IL-15).
86. The method according to any one of claims 81 - 85, wherein the culturing step comprises culturing the lymphocytes obtained from human or non-human animal non- hematopoietic tissue in the presence of IL-2 and IL-15.
87. The method according to any one of claims 83 - 86, wherein the culturing step comprises culturing the lymphocytes in the absence of TCR activation or co-stimulation signals.
88. The method according to any one of claims 83 - 87, wherein the culturing step comprises culturing the lymphocytes in the absence of a T cell receptor pathway agonist.
89. The method according to any one of claims 83 - 88, wherein the culturing step comprises culturing the lymphocytes in the absence of stromal or epithelial cells.
90. The method according to claim 89, wherein stromal or epithelial cells are removed prior to culture.
91. The method according to claim 89, wherein the lymphocytes are cultured in the absence of fibroblasts.
92. The method according to any one of claims 83 - 91, wherein the lymphocytes have been obtained from skin, the gastrointestinal tract (e.g., colon), mammary gland tissue, lung, liver, pancreas or prostate.
93. The method of any one of claims 83 - 92, wherein the ex vivo expanded V52" γδ T cells were obtained by: culturing lymphocytes obtained from non-hematopoietic tissue of humans or non- human animals in the presence of IL-2, IL-15, and a factor selected from the group consisting ofIL-4, IL-21, IL-6, IL-7, IL-8, IL-9, IL-12, IL-18, IL-33, IGF-1, IL-Ιβ, human platelet lysate (HPL), and stromal cell-derived factor- 1 (SDF-1) for at least 5 days to produce an expanded population of γδ T cells.
94. The method of any one of claims 90 - 93, wherein, within 14 days of culture, the expanded population of γδ T cells comprises at least 20-fold the number of γδ T cells as the γδ T cells obtained from a non-hematopoietic tissue.
95. The method of claim 94, wherein, within 7 days of culture, the expanded population of γδ T cells comprises at least 2-fold the number of γδ T cells as the γδ T cells obtained from a non-hematopoietic tissue.
96. The method of any one of claims 83 - 95, wherein the expanded population of γδ T cells is at least 50% νδΓ cells.
97. The method of claim 96, wherein the expanded population of γδ T cells is at least 70% Υδ1+ cells.
98. The method of claim 97, wherein the expanded population of γδ T cells is at least 90% V51+ cells.
99. The method of any one of claims 70 - 98, wherein the administration of the at least one TIGIT inhibitor is performed prior to, during, after the transplantation, or combinations thereof.
100. The method of any one of claims 70 - 99, wherein at least one additional agent is administered to the recipient.
101. The method of claim 100, wherein the at least one additional agent is an anti-cancer agent.
102. The method of claim 101, wherein the anti-cancer agent is selected from the group consisting of radiation, a chemotherapeutic or growth inhibitory agent, a targeted therapeutic agent, a small molecule inhibitor, a T cell expressing a chimeric antigen receptor, an antibody or antigen-binding fragment thereof, an antibody-drug conjugate, an angiogenesis inhibitor, an antineoplastic agent, a cancer vaccine, an adjuvant, and combinations thereof.
103. The method of any one of claims 70 - 102, wherein the recipient has cancer.
104. The method of any one of claims 70 - 103, wherein the γδ T cells are obtained from an autologous donor.
105. The method of any one of claims 83 - 104, wherein the non-hematopoietic tissue is tumor tissue.
106. The method of any one of claims 83 - 104, wherein the non-hematopoietic tissue is skin tissue.
107. The method of any one of claims 70 - 106, further comprising separating νδ1+γδ T cells or DN cells from the γδ T cells prior to transplantation.
108. The method of any one of the preceding claims, wherein at least one additional checkpoint inhibitor is administered.
109. The method of claim 108, wherein the additional checkpoint inhibitor does not inhibit PD1.
110. The method of claim 108, wherein the additional checkpoint inhibitor inhibits TIM-3
111. The method of claim 109, wherein the additional checkpoint inhibitor inhibits LAG-3.
112. The method of any one of the preceding claims, wherein the administration of the TIGIT inhibitor results in elevated release of cytokines from the γδ T cells, selected from the group consisting of IFN-γ, TNA-a, interleukins, and combinations thereof.
113. A method of treating cancer, comprising: administering a therapeutically effective amount of a TIGIT inhibitor to a cancer patient who has been determined to have an elevated representation of V51+ cells in a sample of peripheral blood.
114. The method of claim 113, further comprising the prior step of determining the representation of V51+ cells in the peripheral blood sample.
115. The method of claim 114, further comprising the step, after determining the representation of V51+ cells and before administering the TIGIT inhibitor, of selecting the patient for TIGIT inhibitor treatment if the patient's sample has been determined to have an elevated representation of V51+ cells.
116. The method of any one of claims 113 - 115, wherein the elevated representation is an increase in V51+ cells as a percentage of total lymphocytes in the sample.
117. The method of claim 116, wherein greater than 0.5% of total lymphocytes in the sample are determined to be V51+ cells.
118. The method of claim 117, wherein greater than 1.0% of total lymphocytes are determined to be V51+ cells.
119. The method of claim 118, wherein greater than 2.0% of total lymphocytes are determined to be V51+ cells.
120. The method of any one of claims 113 - 115, wherein the elevated representation is an increase in V51+ cells as a percentage of total γδ T cells in the sample.
121. The method of claim 120, wherein greater than 10% of total γδ T cells in the sample are V51+ cells.
122. The method of claim 121, wherein greater than 15% of total γδ T cells in the sample are V51+ cells.
123. The method of any one of claims 113 - 115, wherein the elevated representation is an increase in the ratio of V51+ cells to V52+ cells in the sample.
124. The method of claim 123, wherein the ratio of V51+:V52+ cells is greater than 1 :9.
125. The method of claim 124, wherein the ratio of V51+:V52+ cells is greater than 1 :8.
126. The method of claim 125, wherein the ratio of V51+:V52+ cells is greater than 1 :7.
127. The method of claim 126, wherein the ratio of V51+:V52+ cells is greater than 1 :6.
128. The method of any one of claims 113 - 127, wherein the TIGIT inhibitor is an antibody or antigen-binding antibody fragment that binds specifically to TIGIT.
129. The method of any one of claims 113 - 128, further comprising administration of at least one additional anti-cancer therapy.
130. The method of claim 129, wherein at least one of the at least one additional anti-cancer therapy is administration of a non-TIGIT checkpoint inhibitor.
131. The method of claim 130, wherein the non-TIGIT checkpoint inhibitor is an antibody that binds specifically to PD-1.
132. The method of claim 130, wherein the non-TIGIT checkpoint inhibitor is an antibody that binds specifically to PDL 1.
133. The method of any one of claims 1 - 132, wherein the cancer is a solid, non- hematopoietic, cancer.
134. The method of claim 133, wherein the cancer is a breast cancer.
135. The method of any one of claims 113 - 132, wherein the cancer is a hematological cancer.
136. The method of claim 7 or a ny one of claims 8 - 21 as depend from claim 7, wherein the ex vivo expanded V52" γδ T cells were obtained by: culturing lymphocytes obtained from hematopoietic tissue of humans or non-human animals in the presence of interleukin-2 (IL-2) and/or mterleukin-15 (IL-15), and not in direct contact with stromal or epithelial cells during culture.
PCT/EP2018/065730 2017-06-14 2018-06-13 Methods of activating v delta 2 negative gamma delta t cells WO2018229163A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762519714P 2017-06-14 2017-06-14
US62/519,714 2017-06-14

Publications (1)

Publication Number Publication Date
WO2018229163A1 true WO2018229163A1 (en) 2018-12-20

Family

ID=63036006

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2018/065730 WO2018229163A1 (en) 2017-06-14 2018-06-13 Methods of activating v delta 2 negative gamma delta t cells

Country Status (1)

Country Link
WO (1) WO2018229163A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110157686A (en) * 2019-05-22 2019-08-23 南京大学 A kind of novel immunologic test point activates science oncolytic adenovirus and its construction method and the application of immune costimulation
WO2020095058A1 (en) * 2018-11-08 2020-05-14 GammaDelta Therapeutics Limited Methods for isolating and expanding cells
US10988517B2 (en) 2019-01-07 2021-04-27 Shattuck Labs, Inc. Heterodimeric proteins for modulating gamma delta T cells
US11021537B2 (en) 2017-05-01 2021-06-01 Agenus Inc. Anti-TIGIT antibodies and methods of use thereof
WO2022221506A1 (en) * 2021-04-16 2022-10-20 Acepodia Biotechnologies Ltd. Novel compositions enriched in gamma delta t cells, methods of preparation, and uses thereof
US11643447B2 (en) 2019-01-07 2023-05-09 Shattuck Labs, Inc. Heterodimeric proteins for modulating gamma delta T cells
US11718669B2 (en) 2021-05-04 2023-08-08 Agenus Inc. Anti-TIGIT and anti-CD96 antibodies

Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012156958A2 (en) 2011-05-19 2012-11-22 Instituto De Medicina Molecular Cell line of lymphocytes comprising gamma-delta t cells, composition and production method thereof
WO2015009856A2 (en) 2013-07-16 2015-01-22 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and tigit inhibitors
WO2016011264A1 (en) 2014-07-16 2016-01-21 Genentech, Inc. Methods of treating cancer using tigit inhibitors and anti-cancer agents
WO2016028656A1 (en) 2014-08-19 2016-02-25 Merck Sharp & Dohme Corp. Anti-tigit antibodies
WO2016073282A1 (en) 2014-11-06 2016-05-12 Genentech, Inc. Combination therapy comprising ox40 binding agonists and tigit inhibitors
WO2016106302A1 (en) 2014-12-23 2016-06-30 Bristol-Myers Squibb Company Antibodies to tigit
WO2016191643A2 (en) 2015-05-28 2016-12-01 Oncomed Pharmaceuticals, Inc. Tigit-binding agents and uses thereof
WO2016198480A1 (en) * 2015-06-09 2016-12-15 Lymphact - Lymphocyte Activation Technologies, S.A. Methods for the production of tcr gamma delta+ t cells
WO2017021526A1 (en) * 2015-08-05 2017-02-09 Amgen Research (Munich) Gmbh Immune-checkpoint inhibitors for use in the treatment of blood-borne cancers
WO2017030823A2 (en) 2015-08-14 2017-02-23 Merck Sharp & Dohme Corp. Anti-tigit antibodies
WO2017037707A1 (en) 2015-09-02 2017-03-09 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Antibodies specific to human t-cell immunoglobulin and itim domain (tigit)
WO2017048824A1 (en) 2015-09-14 2017-03-23 Compass Therapeutics Llc COMPOSITIONS AND METHODS FOR TREATING CANCER VIA ANTAGONISM OF THE CD155/TIGIT PATHWAY AND TGF-β
WO2017053748A2 (en) 2015-09-25 2017-03-30 Genentech, Inc. Anti-tigit antibodies and methods of use
WO2017059095A1 (en) 2015-10-01 2017-04-06 Potenza Therapeutics, Inc. Anti-tigit antigen-binding proteins and methods of use thereof
WO2017072367A1 (en) 2015-10-30 2017-05-04 Cancer Research Technology Limited EXPANSION OF NON-HAEMATOPOIETIC TISSUE-RESIDENT γδ T CELLS AND USES OF THESE CELLS
WO2017152088A1 (en) * 2016-03-04 2017-09-08 JN Biosciences, LLC Antibodies to tigit
WO2017149538A1 (en) * 2016-03-01 2017-09-08 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Antibodies specific to human poliovirus receptor (pvr)
WO2018102536A1 (en) * 2016-11-30 2018-06-07 Oncomed Pharmaceuticals, Inc. Methods for treatment of cancer comprising tigit-binding agents

Patent Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012156958A2 (en) 2011-05-19 2012-11-22 Instituto De Medicina Molecular Cell line of lymphocytes comprising gamma-delta t cells, composition and production method thereof
WO2015009856A2 (en) 2013-07-16 2015-01-22 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and tigit inhibitors
WO2016011264A1 (en) 2014-07-16 2016-01-21 Genentech, Inc. Methods of treating cancer using tigit inhibitors and anti-cancer agents
WO2016028656A1 (en) 2014-08-19 2016-02-25 Merck Sharp & Dohme Corp. Anti-tigit antibodies
WO2016073282A1 (en) 2014-11-06 2016-05-12 Genentech, Inc. Combination therapy comprising ox40 binding agonists and tigit inhibitors
WO2016106302A1 (en) 2014-12-23 2016-06-30 Bristol-Myers Squibb Company Antibodies to tigit
WO2016191643A2 (en) 2015-05-28 2016-12-01 Oncomed Pharmaceuticals, Inc. Tigit-binding agents and uses thereof
WO2016198480A1 (en) * 2015-06-09 2016-12-15 Lymphact - Lymphocyte Activation Technologies, S.A. Methods for the production of tcr gamma delta+ t cells
WO2017021526A1 (en) * 2015-08-05 2017-02-09 Amgen Research (Munich) Gmbh Immune-checkpoint inhibitors for use in the treatment of blood-borne cancers
WO2017030823A2 (en) 2015-08-14 2017-02-23 Merck Sharp & Dohme Corp. Anti-tigit antibodies
WO2017037707A1 (en) 2015-09-02 2017-03-09 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Antibodies specific to human t-cell immunoglobulin and itim domain (tigit)
WO2017048824A1 (en) 2015-09-14 2017-03-23 Compass Therapeutics Llc COMPOSITIONS AND METHODS FOR TREATING CANCER VIA ANTAGONISM OF THE CD155/TIGIT PATHWAY AND TGF-β
WO2017053748A2 (en) 2015-09-25 2017-03-30 Genentech, Inc. Anti-tigit antibodies and methods of use
WO2017059095A1 (en) 2015-10-01 2017-04-06 Potenza Therapeutics, Inc. Anti-tigit antigen-binding proteins and methods of use thereof
WO2017072367A1 (en) 2015-10-30 2017-05-04 Cancer Research Technology Limited EXPANSION OF NON-HAEMATOPOIETIC TISSUE-RESIDENT γδ T CELLS AND USES OF THESE CELLS
WO2017149538A1 (en) * 2016-03-01 2017-09-08 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Antibodies specific to human poliovirus receptor (pvr)
WO2017152088A1 (en) * 2016-03-04 2017-09-08 JN Biosciences, LLC Antibodies to tigit
WO2018102536A1 (en) * 2016-11-30 2018-06-07 Oncomed Pharmaceuticals, Inc. Methods for treatment of cancer comprising tigit-binding agents

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
DE WITTE M A ET AL: "Early Reconstitution of NK and [gamma][delta] T Cells and Its Implication for the Design of Post-Transplant Immunotherapy.", BIOLOGY OF BLOOD AND MARROW TRANSPLANTATION, vol. 24, no. 6, June 2018 (2018-06-01), pages 1152 - 1162, XP055514409, ISSN: 1523-6536 *
DENIGER D C ET AL: "Activating and Propagating Polyclonal Gamma Delta T Cells with Broad Specificity for Malignancies", CLINICAL CANCER RESEARCH, vol. 20, no. 22, 15 November 2014 (2014-11-15), pages 5708 - 5719, XP055216815, ISSN: 1078-0432, DOI: 10.1158/1078-0432.CCR-13-3451 *
GENTLES, A. J. ET AL.: "The prognostic landscape of genes and infiltrating immune cells across human cancers", NATURE MEDICINE, vol. 21, no. 8, August 2015 (2015-08-01), pages 938 - 945
KABELITZ D: "Human [gamma][delta] T cells: From a neglected lymphocyte population to cellular immunotherapy: A personal reflection of 30years of [gamma][delta] T cell research", CLINICAL IMMUNOLOGY, vol. 172, 18 July 2016 (2016-07-18), pages 90 - 97, XP029789610, ISSN: 1521-6616, DOI: 10.1016/J.CLIM.2016.07.012 *
KNIGHT A ET AL: "Human Vdelta1 gamma-delta T cells exert potent specific cytotoxicity against primary multiple myeloma cells", CYTOTHERAPY, vol. 14, no. 9, October 2012 (2012-10-01), pages 1110 - 1118, XP009163614, ISSN: 1465-3249, DOI: 10.3109/14653249.2012.700766 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11021537B2 (en) 2017-05-01 2021-06-01 Agenus Inc. Anti-TIGIT antibodies and methods of use thereof
WO2020095058A1 (en) * 2018-11-08 2020-05-14 GammaDelta Therapeutics Limited Methods for isolating and expanding cells
US10988517B2 (en) 2019-01-07 2021-04-27 Shattuck Labs, Inc. Heterodimeric proteins for modulating gamma delta T cells
US11098093B2 (en) 2019-01-07 2021-08-24 Shattuck Labs, Inc. Heterodimeric proteins for modulating gamma delta T cells
US11643447B2 (en) 2019-01-07 2023-05-09 Shattuck Labs, Inc. Heterodimeric proteins for modulating gamma delta T cells
CN110157686A (en) * 2019-05-22 2019-08-23 南京大学 A kind of novel immunologic test point activates science oncolytic adenovirus and its construction method and the application of immune costimulation
CN110157686B (en) * 2019-05-22 2022-06-21 南京惟亚德生物医药有限公司 Replication type oncolytic adenovirus activated by immune checkpoint and immune co-stimulation and construction method and application thereof
WO2022221506A1 (en) * 2021-04-16 2022-10-20 Acepodia Biotechnologies Ltd. Novel compositions enriched in gamma delta t cells, methods of preparation, and uses thereof
US11718669B2 (en) 2021-05-04 2023-08-08 Agenus Inc. Anti-TIGIT and anti-CD96 antibodies

Similar Documents

Publication Publication Date Title
US10653756B2 (en) Identification of CD8+ T cells that are CD161hi and/or IL18R(α)hi and have rapid drug efflux capacity
WO2018229163A1 (en) Methods of activating v delta 2 negative gamma delta t cells
TW202039830A (en) Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy
TW202039829A (en) Selection of improved tumor reactive t-cells
KR20210033436A (en) Genetically engineered NK cell lines transduced with a polynucleotide encoding novel chimeric antigen receptor and use thereof
TW202208617A (en) Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy
TW202039831A (en) Treatment of nsclc patients refractory for anti-pd-1 antibody
TW202100746A (en) Expansion of tumor infiltrating lymphocytes from liquid tumors and therapeutic uses thereof
CN112426526B (en) Preparation method of NK (natural killer) cells and application of NK cells in treatment of cancers
KR20220005075A (en) Methods of Administration of Chimeric Antigen Receptor Immunotherapy
EP4262830A1 (en) Treatment of cancer with nk cells and a cd20 targeted antibody
US20210030793A1 (en) Methods and compositions for treating cd33+ cancers and improving in vivo persistence of chimeric antigen receptor t cells
TW202208616A (en) Selection of improved tumor reactive t-cells
WO2023288278A1 (en) Chimeric molecules providing targeted costimulation for adoptive cell therapy
EP4319768A1 (en) Chimeric antigen receptor comprising an anti-her2 antibody or antigen-binding fragment thereof and natural killer cells comprising the same
TW202305360A (en) Methods and compositions for t-cell coculture potency assays and use with cell therapy products
CA3200770A1 (en) Methods and reagents for characterizing car t cells for therapies
KR20220004076A (en) Rituximab-resistant chimeric antigen receptor and uses thereof
EP3191109B1 (en) A cell population for use in treating cancer
US20240058447A1 (en) Use of fusion constructs for il-2 independent t cell therapy
WO2024040135A2 (en) Methods of administering natural killer cells comprising an anti-human epidermal growth factor receptor 2 (her2) chimeric antigen receptor (car)
Cho Targeting CD155 interactions to enhance natural killer cell therapy for osteosarcoma
KR20220152220A (en) CD19-directed chimeric antigen receptor T cell composition and methods and uses thereof
WO2022216831A1 (en) Treatment of cancer with nk cells and a her2 targeted antibody
KR20220119080A (en) Improved Methods for Culturing Therapeutic Tumor-Infiltrating Lymphocytes

Legal Events

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

Ref document number: 18746097

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18746097

Country of ref document: EP

Kind code of ref document: A1