WO2024073111A2 - Affinity binding entities directed to b7h6 and methods of use thereof - Google Patents

Affinity binding entities directed to b7h6 and methods of use thereof Download PDF

Info

Publication number
WO2024073111A2
WO2024073111A2 PCT/US2023/034227 US2023034227W WO2024073111A2 WO 2024073111 A2 WO2024073111 A2 WO 2024073111A2 US 2023034227 W US2023034227 W US 2023034227W WO 2024073111 A2 WO2024073111 A2 WO 2024073111A2
Authority
WO
WIPO (PCT)
Prior art keywords
cell
seq
antibody
cells
set forth
Prior art date
Application number
PCT/US2023/034227
Other languages
French (fr)
Inventor
Blake AFTAB
Arun BHAT
Kevin Nishimoto
Alexander TEAGUE
Gauri LAMTURE
Beibei Ding
Nitya RAMADOSS
Melinda Au
Original Assignee
Adicet Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Adicet Therapeutics, Inc. filed Critical Adicet Therapeutics, Inc.
Publication of WO2024073111A2 publication Critical patent/WO2024073111A2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation

Definitions

  • Chimeric antigen receptors capable of binding B7H6, polynucleotides, host cells comprising the polynucleotides and/or CARs, and methods of treating disorders associated with B7H6 in a patient are provided.
  • BACKGROUND OF THE DISCLOSURE [0003] Adoptive cellular therapy has undergone near constant iteration for more than thirty (30) years, from early days focusing on basic lymphokine activation and/or tumor infiltration to more recent strategies engineering immune cells to express genetically engineered antigen receptors, such as chimeric antigen receptors (CARs). While there have been some hints and indications of the curative potential of these approaches along the way, much still remains to be done.
  • CAR-T lymphocytes In particular, successful tumor eradication by CAR-T lymphocytes depends on CAR-T cell persistence and effector function, but an excess of either can trigger graft-versus-host (GvH) effects in the patient. Additionally, while the adoptive transfer of T cells expressing CARs has shown some success in treating haematological malignancies, only limited efficacy has been shown in other cancer types, particularly solid tumors. Compared with haematological diseases, solid tumors present unique challenges, including but not limited to highly immunosuppressive and metabolically challenging tumor microenvironments.
  • B7 homolog 6 (B7H6), also known as NCR3LG1 (natural killer cell cytotoxicity receptor 3 ligand 1), is human-specific B7 family member that is a ligand of NKp30.
  • NKp30 a Page 1 of 203 1 D 0 o 9 c 7 u 6 m 5 e 7 n 1 t 6 2 9 ⁇ 1 ⁇ AMERICAS member of the CD28 family, is an activating receptor of natural killer (NK) cells.
  • B7H6 is typically not expressed on normal human tissues, but is expressed in many human tumor cell lines including melanoma, carcinomas, T and B lymphomas, and myeloid leukemias, as well as primary tumor blood and bone marrow cells (Brandt et al., J Exp Med.2009; 206: 1495-1503; Cao et al., J Biol Chem.2015; 290: 29964-29973). Aside from its expression in cancer, B7H6 is upregulated under inflammatory conditions such as atopic dermatitis (Salimi et al., J Immunol. 2016; 196: 45-54). The wide expression profile on tumors and the lack of expression on healthy tissues make B7H6 an attractive target for immunotherapeutic approaches.
  • ⁇ T cells which are known to be associated with high alloreactive potential in general, as well as a propensity to trigger complications such as cytokine release syndrome (CRS) and macrophage activation syndrome (MAS).
  • CRS cytokine release syndrome
  • MAS macrophage activation syndrome
  • Gamma delta ( ⁇ ) T cells are thymus-derived lymphocytes that differ from ⁇ T cells in their mechanism of activation and function, as well as in their anatomical distribution.
  • ⁇ T cells function exclusively in adaptive immunity
  • ⁇ T cells are innate-like immune cells that recognize malignant cells through a repertoire of activating receptors in a MHC- independent manner, similar to NK cells (Welsh et al., Immunol Rev.1997; 159: 79-93).
  • ⁇ T cells can potentially be used in an allogeneic setting without the risk of causing graft versus host disease (GvHD).
  • engineered ⁇ -T cells may produce less proinflammatory cytokines than ⁇ -T cells, which could thereby reduce the risk of CRS in patients (Harrer et al., BMC Cancer 2017; 17(1): 551).
  • an affinity binding entity comprises an antigen binding domain that specifically binds to B7 homolog 6 (B7H6).
  • a heavy chain variable region/light chain variable region (HCVR/LCVR) sequence pair selected from the group consisting of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, 15/16, 17/18, 19/20, 21/22, 23/24, 25/26, 27/28, 29/30, 31/32, 33/34, 35/36, 37/38, and 39/40; or the six CDRs of a HCVR/LCVR sequence pair selected from the group consisting of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, 15/16, 17/18, 19/20, 21/22, 23/24, 25/26, 27/28, 29/30, 31/32, 33/34, 35/36, 37/38, and 39/40.
  • the antigen binding domain comprises a HCVR/LCVR sequence pair selected from the group consisting of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, and 15/16; or the six CDRs of a HCVR/LCVR sequence pair selected from the group consisting of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, and 15/16.
  • the antigen binding domain comprises a HCVR/LCVR sequence pair of SEQ ID NOs: 1/2; or the six CDRs of a HCVR/LCVR sequence pair of SEQ ID NOs: 1/2.
  • antigen binding domain comprises a HCVR/LCVR sequence pair of SEQ ID NOs: 3/4; or the six CDRs of a HCVR/LCVR sequence pair of SEQ ID NOs: 3/4.
  • the affinity binding entity is an antibody, or an antibody fragment. In embodiments, the antibody or antibody fragment is bispecific. In embodiments, the antibody or antigen binding fragment is chimeric, humanized, or human. In embodiments, the antibody or antibody fragment is monoclonal. In embodiments, the affinity binding entity is selected from the group consisting of scFv, Fab, Fab’, Fv, F(ab’)2, dsFv, dAb, and any combination or plurality thereof.
  • a chimeric antigen receptor comprises any of the afore-mentioned affinity binding entities as disclosed herein.
  • the CAR comprises a hinge domain.
  • the hinge domain comprises a glycine polymer, glycine-serine polymer, glycine- alanine polymer, alanine-serine polymer, immunoglobulin heavy chain hinge, or receptor-derived Page 3 of 203 1097657169 ⁇ 1 ⁇ AMERICAS hinge.
  • the receptor-derived hinge is a CD8 alpha hinge domain.
  • the CD8 alpha hinge domain comprises an amino acid sequence set forth as SEQ ID NO: 209.
  • the CAR comprises a transmembrane (TM) domain.
  • the TM domain comprises a TM region of 4-1BB/CD137, activating NK cell receptors, an Immunoglobulin protein, B7-H3, BAFFR, BLAME (SLAMF8), BTLA, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, or CD154, CD100 (SEMA4D), CD103, CD160 (BY55), CD18, CD19, CD19a, CD2, CD247, CD27, CD276 (B7-H3), CD28, CD29, CD3 delta, CD3 epsilon, CD3 gamma, CD3 zeta, CD30, CD4, CD40, CD49a, CD49D, CD49f, CD69, CD7, CD84, CD8, CD8alpha, CD8bet
  • the TM domain comprises a TM domain of CD8, preferably wherein the CD8 TM domain is a TM domain of CD8 alpha.
  • the TM domain comprises the amino acid sequence set forth as SEQ ID NO: 211.
  • the CAR further comprises at least one costimulatory domain.
  • the costimulatory domain comprises a costimulatory domain of TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, CARD11, B7-H3, CEACAM1, CRTAM, CD2, CD3C, CD4, CD7, CD8 ⁇ , CD8 ⁇ , CD11a, CD11b, CD11c, CD11d, IL2R ⁇ , IL2 ⁇ , IL7R ⁇ , IL4R, IL7R, IL15R, IL21R, CD18, CD19, CD19a, CD27, CD28, CD29, CD30, CD40, CDS, CD49a, CD49D, CD49f, CD54 (ICAM), CD69, CD70, CD80, CD83, CD84, CD86, CD96 (Tactile), CD100 (SEMA4D), CD103, CD134 (OX40), CD137 (4-1BB), CD152 (CTLA-4), CD160 (BY55), CD162 (SEL
  • the costimulatory domain is a 4-1BB costimulatory domain.
  • the 4-1BB costimulatory domain comprises an amino acid sequence set forth as SEQ ID NO: 202.
  • the CAR further comprises one or more intracellular signaling domains.
  • the intracellular signaling domain is a CD3 ⁇ intracellular signaling domain.
  • the CD3 ⁇ intracellular signaling domain comprises an amino acid sequence set forth as SEQ ID NO: 229, 231, or 232.
  • the CAR further comprises a signal peptide.
  • the signal peptide comprises an amino acid sequence set forth as SEQ ID NO: 170.
  • an isolated polynucleotide comprising a nucleic acid sequence encoding an affinity binding entity as herein disclosed.
  • an expression vector comprises the isolated polynucleotide.
  • the expression vector is operably linked to a cis-acting regulatory element.
  • a cell comprises the affinity binding entity, the isolated polynucleotide, and/or the expression vector.
  • an isolated polynucleotide comprising a nucleic acid sequence encoding a CAR as herein disclosed.
  • the polynucleotide comprises a nucleic acid sequence encoding at least one multicistronic linker region.
  • the multicistronic linker region encodes a cleavage sequence.
  • the cleavage sequence is selected from T2A, F2A, P2A, E2A, furin, and furin-P2A (FP2A).
  • the multicistronic linker region encodes an internal ribosomal entry site (IRES).
  • the polynucleotide further comprises a nucleic acid encoding one or more additional polypeptides.
  • the one or more additional polypeptides is selected from the group comprising or consisting of lymphotoxin beta receptor (LTBR), low- Page 5 of 203 1097657169 ⁇ 1 ⁇ AMERICAS affinity nerve growth factor receptor (LNGFR), a dominant negative (dn) receptor for TGF-beta or Fas, a truncated form of the human epidermal growth factor receptor (EGFRt), and membrane- bound IL-12 (mbIL-12), or any combination thereof.
  • LTBR lymphotoxin beta receptor
  • LNGFR low- Page 5 of 203 1097657169 ⁇ 1 ⁇ AMERICAS affinity nerve growth factor receptor
  • dn dominant negative receptor for TGF-beta or Fas
  • EGFRt truncated form of the human epidermal growth factor receptor
  • mbIL-12 membrane- bound IL-12
  • the one or more additional polypeptides is selected from a fluorescent protein, a gamma chain cytokine, CD19, CD20, LNGFR, EGFRt, LTBR, dnTGF ⁇ R2, dominant negative Fas, membrane-bound IL-12, CAR that binds to CD70, or any combination thereof.
  • the one or more additional polypeptides is a dominant negative receptor for TGF ⁇ .
  • the dominant negative receptor for TGF ⁇ is dnTGF ⁇ R2, optionally wherein the dnTGF ⁇ R2 comprises an amino acid sequence set forth as SEQ ID NO: 241.
  • the one or more additional polypeptides is lymphotoxin beta receptor (LTBR), optionally wherein the LTBR comprises an amino acid sequence set forth as SEQ ID NO: 245.
  • the one or more additional polypeptides is a truncated form of the epidermal growth factor receptor (EGFRt), optionally wherein the EGFRt comprises an amino acid sequence set forth as SEQ ID NO: 237.
  • the one or more additional polypeptides is low- affinity nerve growth factor receptor (LNGFR), optionally wherein the LNGFR comprises an amino acid sequence set forth as SEQ ID NO: 249.
  • the one or more additional polypeptides is selected from the group comprising or consisting of lymphotoxin beta receptor (LTBR), a dominant negative (dn) receptor for TGF-beta or Fas, a truncated form of the human epidermal growth factor receptor (EGFRt), and membrane-bound IL-12 (mbIL-12).
  • LTBR lymphotoxin beta receptor
  • dn dominant negative receptor for TGF-beta or Fas
  • EGFRt human epidermal growth factor receptor
  • mbIL-12 membrane-bound IL-12
  • the one or more additional polypeptides is a dominant negative Fas (dnFas).
  • the one or more additional polypeptides is membrane-bound IL-12 (mbIL-12).
  • the one or more additional polypeptides is a CAR that binds to CD70.
  • the one or more additional polypeptides are operably linked to a nucleic acid sequence encoding a signal peptide.
  • the signal peptide comprises an amino acid sequence selected from SEQ ID NO: 235, SEQ ID NO: 239, SEQ ID NO: 243, SEQ ID NO: 247, and SEQ ID NO: 196.
  • the polynucleotide comprising a nucleic acid encoding a CAR as herein disclosed has a nucleic acid sequence selected from SEQ ID NO: 294, 298, 302, 306, 310, 314, 318, 322, 326, 330, or 334.
  • an expression vector comprising the polynucleotide encoding a CAR of the present disclosure, operably linked to a cis-acting regulatory element.
  • a ⁇ T cell comprising (a) a nucleic acid encoding a chimeric antigen receptor (CAR), the CAR comprising an antigen binding domain that specifically binds to B7 homolog 6 (B7H6); and/or (b) a polypeptide comprising a CAR comprising an amino acid sequence encoded by the nucleic acid of (a), wherein the ⁇ T cell functionally expresses the binding domain of the polypeptide or nucleic acid encoded CAR on the surface of the ⁇ T cell.
  • the ⁇ T cell comprises a CAR comprising an affinity binding entity as herein disclosed.
  • the nucleic acid comprises a polynucleotide encoding a CAR as herein disclosed, or an expression vector encoding a CAR as herein disclosed.
  • a modified immune cell comprises a CAR as herein disclosed.
  • the modified immune cell comprises a polynucleotide comprising a nucleic acid sequence encoding a CAR as herein disclosed.
  • the modified immune cell comprises an expression vector that encodes for a CAR as herein disclosed.
  • the modified immune cell is a ⁇ ⁇ ⁇ T cell, a ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ cell, an ⁇ T cell, a NK cell, a NKT cell, or a macrophage.
  • the modified immune cell is a ⁇ ⁇ ⁇ T cell.
  • the ⁇ ⁇ ⁇ T cell is a ⁇ 1, a ⁇ 2, a ⁇ 3, or a ⁇ 4 ⁇ T cell, preferably a ⁇ 2- ⁇ T cell, more preferably a ⁇ 1 ⁇ T cell.
  • the modified immune cell or ⁇ ⁇ ⁇ T cell exhibits in vitro and/or in vivo cell killing activity against a tumor cell that exhibits cell surface expression of B7H6.
  • the cell killing activity of the modified immune cell, or the ⁇ T cell is greater than an innate level of in vitro and/or in vivo tumor cell killing activity in a control modified immune cell or control ⁇ T cell of the same type that does not comprise a CAR construct.
  • the modified immune cell or ⁇ T cell proliferates in response to contact with the tumor cell that exhibits cell surface expression of B7H6.
  • the modified immune cell or ⁇ T cell exhibits increased proliferation in response to contact with the Page 7 of 203 1097657169 ⁇ 1 ⁇ AMERICAS tumor cell that exhibits cell surface expression of B7H6 as compared to a control immune cell or ⁇ T cell of the same type that does not comprise a CAR construct.
  • the modified immune cell or ⁇ T cell proliferates in a host organism that comprises a tumor cell that exhibits cell surface expression of B7H6.
  • the modified immune cell or the ⁇ T cell expresses pro-inflammatory cytokines after contact with a tumor cell that exhibits cell surface expression of B7H6.
  • the modified immune cell or ⁇ T cell comprises at least one disrupted gene.
  • the disrupted gene is cytokine inducible SH2-containing protein (CISH).
  • the at least one disrupted endogenous gene is Cbl proto-oncogene B (CBL-B).
  • the at least one disrupted endogenous gene is Zinc Finger Protein 91 (ZFP91).
  • the at least one disrupted endogenous gene is Roquin.
  • the at least one disrupted endogenous gene is CD58 and/or ICAM-1.
  • the plurality of modified immune cells, or the plurality of ⁇ T cells comprise a composition that is at least 60%, 80%, or from about 60% or 80% to about 90% or 95% ⁇ 1, ⁇ 2, ⁇ 3, or ⁇ 4 ⁇ T cells, preferably ⁇ 1 or ⁇ 2 ⁇ T cells, more preferably ⁇ 2- ⁇ T cells, most preferably ⁇ 1 ⁇ T cells.
  • the plurality of modified immune cells, or the plurality of ⁇ T cells comprises at least about 10 7 modified immune cells or ⁇ T cells, respectively, preferably from about 10 8 modified immune cells or ⁇ T cells to about 10 11 modified immune cells or ⁇ T cells, respectively.
  • the method comprises transfecting immune cell(s) or ⁇ T cell(s) with a construct comprising an expression vector encoding a CAR as herein disclosed, optionally wherein the cell(s) has/have at least one disrupted gene.
  • the method comprises retroviral transduction.
  • the method comprises ex vivo expansion of the immune cell(s) or ⁇ T cell(s), wherein the ex vivo expansion is performed before transfection and/or after transfection of the expression vector.
  • an antibody-drug conjugate comprising an affinity binding entity as herein disclosed.
  • Page 8 of 203 1097657169 ⁇ 1 ⁇ AMERICAS [0035]
  • a pharmaceutical composition comprising an affinity binding entity as herein disclosed, a modified immune or ⁇ T cell as herein disclosed, or an ADC as herein disclosed.
  • the pharmaceutical composition comprises a pharmaceutically acceptable carrier.
  • a method of inhibiting the growth of a cell that exhibits cell surface expression of B7H6, comprising contacting the cell with an affinity binding entity as herein disclosed, a modified immune cell(s) as herein disclosed, a ⁇ T cell(s) as herein disclosed, an ADC as herein disclosed, or a pharmaceutical composition as herein disclosed.
  • a method of killing a tumor cell that exhibits cell surface expression of B7H6 comprising contacting the cell with an affinity binding entity as herein disclosed, a modified immune cell(s) as herein disclosed, a ⁇ T cell(s) as herein disclosed, an ADC as herein disclosed, or a pharmaceutical composition as herein disclosed.
  • the method comprises contacting the tumor cell with a therapeutically effective amount of an affinity binding entity as herein disclosed, modified immune cell(s) as herein disclosed, ⁇ T cell(s) as herein disclosed, or a pharmaceutical composition as herein disclosed.
  • the method comprises introducing into a host organism comprising the tumor cell the therapeutically affective amount of the affinity binding entity, the modified immune cell(s), the ⁇ T cell(s), the ADC, or the pharmaceutical composition.
  • the method comprises introducing into the host organism comprising the tumor cell the therapeutically affective amount of the affinity binding entity, the modified immune cell(s), the ⁇ T cell(s), the ADC, or the pharmaceutical composition; and simultaneously or sequentially administering one or more methods to elevate common gamma chain cytokine(s).
  • the administering one or more methods to elevate common gamma chain cytokine(s) comprises simultaneously or sequentially administering an amount of common gamma chain cytokine(s) effective to increase proliferation, cytotoxic activity, persistence, or the combination thereof of the introduced modified immune cell(s) or ⁇ T cell(s).
  • the one or more methods to elevate common gamma chain cytokine(s) comprises lymphodepletion before introducing the modified immune cell(s) or the ⁇ T cell(s). In embodiments, the one or more methods to elevate common gamma chain cytokine(s) comprises secretion of one or more common gamma chain cytokine(s) from the introduced modified immune cell(s) or ⁇ T cell(s).
  • the method reduces the in vivo tumor burden in the host organism and/or increases the mean survival time of the host organism as compared to a control organism, wherein the control Page 9 of 203 1097657169 ⁇ 1 ⁇ AMERICAS organism is not treated with the affinity binding entity, the modified immune cell(s), ⁇ T cell(s), the ADC, or the pharmaceutical composition.
  • the host organism is human.
  • the method is a method of treating cancer in a subject in need thereof.
  • an affinity binding entity as herein disclosed, modified immune cell(s) as herein disclosed, ⁇ T cell(s) as herein disclosed, an ADC as herein disclosed, or a pharmaceutical composition as herein disclosed in the preparation of a medicament for the treatment of a disease or condition.
  • the disease or condition is cancer.
  • the ⁇ T cells may comprise a dual CAR binding to CD70 and B7H6, or the method may further comprise co-administering the ⁇ T cell according to the subject invention simultaneously or sequentially with an immune cell (e.g., a T cell or NK cell) comprising a CAR binding to B7H6 and a CAR binding to CD70.
  • an immune cell e.g., a T cell or NK cell
  • FIG.1 is a graph illustrating B7H6 binding specificity of phage-display derived antibody IgG clones against B7H6+ cell lines.
  • FIG.2 is a table showing EC50 values from phage-display derived antibody clones against recombinant B7H6 protein.
  • FIGS.3A-3C illustrate phage-display derived antibody IgG clones binding to the V- distal and C-proximal membrane domain of B7H6. Page 10 of 203 1097657169 ⁇ 1 ⁇ AMERICAS [0047]
  • FIG.4 is a graph illustrating B7H6 cell surface expression in different cancer cell lines.
  • FIGS.5A-5B illustrate NFAT activity of Jurkat Lucia cells modified with anti-B7H6 scFv clones formatted into a CAR (B7H6 CAR),
  • FIGS.6A-6C are graphs illustrating in vitro cytotoxicity of B7H6 CAR V ⁇ 1 T cells against various tumor cell lines.
  • FIGS.7A-7B are graphs illustrating in vitro cytokine polyfunctionality of B7H6 CAR V ⁇ 1 T cells stimulated with B7-H6+ target tumor cells.
  • FIGS.8A-8B depict data illustrating the T cell memory profile, chemokine receptors, and activation/exhaustion-associated marker expression on B7H6 CAR V ⁇ 1 T cells.
  • FIG.9 is a series of graphs illustrating in vitro proliferation of B7H6 CAR V ⁇ 1 T cells stimulated with B7H6+ target tumor cells.
  • FIGS.10A-10B are graphs illustrating that B7H6 CAR V ⁇ 1 T cells maintain in vitro cytotoxicity against B7-H6+ target tumor cells in the presence of soluble B7H6.
  • FIGS.11A-11B are graphs illustrating in vivo therapeutic efficacy of B7H6 CAR V ⁇ 1 T cells in a subcutaneous human xenograft HCT-15 cell NOD scid gamma (NSG) mouse model.
  • FIG.12 depicts plots illustrating armoring of B7H6 CAR V ⁇ 1 T cells with dominant negative TGFR ⁇ 2 (dnTGFR ⁇ 2).
  • FIG.13 shows graphs illustrating in vitro inhibition of TGF ⁇ 1-induced pSMAD2/3 expression in B7H6 CAR V ⁇ 1 T cells containing dnTGFR ⁇ 2.
  • FIG.14 is a graph illustrating enhanced in vitro cytotoxicity of B7H6 CAR V ⁇ 1 T cells containing dnTGFR ⁇ 2 against B7H6+ target tumor cells.
  • FIG.15 is a graph illustrating gene knockout efficiency of two different guide RNAs targeting cytokine inducible SH2 containing protein (CISH).
  • FIG.16A is a graph illustrating that V ⁇ 1 T cells with CISH knocked out can be enriched post-depletion of ⁇ T cells. Page 11 of 203 1097657169 ⁇ 1 ⁇ AMERICAS
  • FIG.16B is a graph illustrating viability of V ⁇ 1 T cells with CISH knocked out.
  • FIGS.17A-17B illustrate CISH KO enhanced the in vitro cytotoxicity of V ⁇ 1 T cells.
  • FIG.18 illustrates the CAR-mbIL-12 constructs designs.
  • FIGS.19A-19D illustrate the expansion and expression of the CAR-mbIL-12 in V ⁇ 1 T cells.
  • FIG.20 illustrates the enhanced in vitro cytotoxicity of CAR-mbIL-12 in V ⁇ 1 T cells.
  • FIG. 21 illustrates in vivo therapeutic efficacy of CAR-mbIL-12 in V ⁇ 1 T cells in a subcutaneous human xenograft Raji cell NSG mouse model.
  • FIGS.22A-22B illustrate CBL-B KO enhanced the in vitro cytotoxicity of V ⁇ 1 T cells.
  • FIGS.23A-23B illustrate Roquin KO enhanced the in vitro cytotoxicity of V ⁇ 1 T cells.
  • FIGS.24A-24B illustrate CD58 or ICAM-1 KO enhanced the in vitro cell survival of V ⁇ 1 T cells in an allogeneic MLR assay. DETAILED DESCRIPTION I. Definitions [0069] For purposes of interpreting this specification, the following definitions will apply, and whenever appropriate, terms used in the singular will also include the plural and vice versa. In the event that any definition set forth conflicts with any document incorporated herein by reference, the definition set forth below shall control.
  • a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 2.7, 3, 4, 5, 5.3, and 6. This applies regardless of the breadth of the range.
  • the terms “patient,” “subject,” “individual,” and the like are used interchangeably herein, and refer to any animal amenable to the methods described herein. In certain non- limiting embodiments, the patient, subject or individual is a human.
  • the term “diagnosis”, or “diagnosing” as used herein refers to the process of identifying a disease, such as cancer, by its signs, symptoms, and/or results of various tests. A conclusion reached through such a process is a diagnosis. Forms of testing commonly performed include blood tests, medical imaging, urinalysis, biopsy, and the like.
  • the term “agent” refers to any protein, nucleic acid molecule (including chemically modified nucleic acids), compound, antibody, small molecule, organic compound, inorganic compound, other molecule of interest, or cell (e.g., cell engineered to express a chimeric antigen receptor). Agent can include a therapeutic agent, a diagnostic agent or a pharmaceutical agent.
  • a therapeutic or pharmaceutical agent is one that alone or together with an additional agent induces the desired response (such as inducing a therapeutic or prophylactic effect when administered to a subject, including treating a subject suffering from cancer, or other disease/condition.
  • therapeutically effective amount refers to the amount of an agent or composition (e.g., composition comprising an agent) that will elicit a biological or medical response of a tissue, system, or subject that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • ⁇ T cells gamma delta T cells
  • TCR T cell receptor
  • ⁇ T cells specifically includes all subsets of ⁇ T cells, including, without limitation, V ⁇ 1 and V ⁇ 2, V ⁇ 3 ⁇ T cells, as well as na ⁇ ve, effector memory, central memory, and terminally differentiated ⁇ T cells.
  • ⁇ T cells includes V ⁇ 4, V ⁇ 5, V ⁇ 7, and V ⁇ 8 ⁇ T cells, as well as V ⁇ 2, V ⁇ 3, V ⁇ 5, V ⁇ 8, V ⁇ 9, V ⁇ 10, and V ⁇ 11 ⁇ T cells.
  • the ⁇ T cells are V ⁇ 1-, V ⁇ 2-, or V ⁇ 1- and V ⁇ 2-.
  • compositions and methods for making and using engineered and non-engineered ⁇ T cells and/or sub-types thereof include, without limitation, those described in US 2016/0175358; WO 2017/197347; US 9499788; US 2018/0169147; US 9907820; US 2018/0125889 and US 2017/0196910, the contents of each of which are incorporated by reference for all purposes, including the the compositions and methods for making and using engineered and non-engineered ⁇ T cells and/or sub-types thereof.
  • the present application further contemplates T cells, or other engineered leukocytes or lymphocytes, that express one ⁇ -chain or one ⁇ -chain, optionally in combination with a second polypeptide to form a functional TCR.
  • the ⁇ ⁇ T cells described herein can be ⁇ 1, ⁇ 2, ⁇ 3, or ⁇ 4 ⁇ ⁇ ⁇ T cells, or combinations thereof.
  • the ⁇ ⁇ T cells are mostly (>50%), substantially (>90%), essentially all, or entirely ⁇ 2 ⁇ ⁇ T cells.
  • the ⁇ ⁇ T cells are mostly (>50%), substantially (>90%), essentially all, or entirely ⁇ 1 ⁇ ⁇ T cells.
  • the ⁇ ⁇ T cells are mostly (>50%), substantially (>90%), essentially all, or entirely ⁇ ⁇ ⁇ ⁇ T cells.
  • ⁇ ⁇ T cells for use as described herein can be obtained from an allogeneic or an autologous donor.
  • the ⁇ ⁇ T cells can be, partially or entirely purified, or not purified, and expanded ex vivo. Methods and compositions for ex vivo expansion include, without limitation, those described in WO 2017/197347. The expansion may be performed before or after, or before and after, a CAR polypeptide of the present disclosure is introduced into the ⁇ ⁇ T cell(s).
  • ⁇ T cell refers to T cells expressing ⁇ and ⁇ chains of the TCR as part of a complex with CD3 chain molecules. Each ⁇ and ⁇ chain contains one variable and one constant domain.
  • NK cell Natural killer cell refers to CD56 + CD3 ⁇ granular lymphocytes that play important roles in immunity against viruses and in the immune surveillance of tumors, and constitute a critical cellular subset of the innate immune system (Godfrey J, et al. Leuk Lymphoma 2012 53:1666-1676). NK cells express a remarkably diverse repertoire of inhibitory and activating receptors on their cell surface, which regulates their immune responses.
  • NK cells can kill transformed or infected cells by the release of perforin and granzymes or by using effector molecules of the tumor necrosis factor (TNF) family, such as TNF, TNF-related apoptosis inducing ligand (TRAIL), and Fas ligand, which induce apoptosis in the target cells.
  • TNF tumor necrosis factor
  • TRAIL TNF-related apoptosis inducing ligand
  • Fas ligand Fas ligand
  • NK cells launch cytotoxicity against tumor cells without the requirement for prior sensitization, and can also eradicate MHC-I-negative cells (Narni-Mancinelli E, et al. Int Immunol 201123:427-431). NK cells are considered fairly safe effector cells, as they may avoid the potentially lethal complications of cytokine storms (Morgan R A, et al. Mol Ther 201018:843-851), tumor lysis syndrome (Porter D L, et al. N Engl J Med 2011365:725-733), and on-target, off-tumor effects. [0082] NK cells can be obtained from an allogeneic or an autologous donor.
  • the NK cells can be partially or entirely purified, or not purified, and expanded ex vivo.
  • Methods and compositions for ex vivo expansion include, without limitation, those described in Becker et al., (2016) Cancer Immunol. Immunother.65(4): 477-84).
  • the expansion may be performed before or after, or before and after, a CAR is introduced into the NK cell(s).
  • expansion of Page 15 of 203 1097657169 ⁇ 1 ⁇ AMERICAS NK cells can include the use of engineered feeder cells, cytokine cocktails (e.g., IL-2, IL-15), and/or aAPCs (Cortés-Selva, D et al., (2021) Trends Pharmacol Sci.42(1): 45-59).
  • cytokine cocktails e.g., IL-2, IL-15
  • aAPCs Cortés-Selva, D et al., (2021) Trends Pharmacol Sci.42(1): 45-59.
  • placental hematopoietic stem-cell derived natural killer (PNK) cells or immortalized cell lines e.g., NK-92
  • PNK hematopoietic stem-cell derived natural killer
  • NK cells that can be used for engineering the expression of CARs herein can be differentiated from human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs).
  • hESCs human embryonic stem cells
  • iPSCs induced pluripotent stem cells
  • NKT Natural killer T cells
  • NKT cells are T lineage cells that share morphological and functional characteristics with both T cells and NK cells. NKT cells are rapid responders of the innate immune system and mediate potent immunoregulatory and effector functions in a variety of disease settings.
  • NKT cells Ligand recognition in NKT cells leads to rapid secretion of proinflammatory cytokines (such as IFN- ⁇ and TNF- ⁇ ) and anti-inflammatory cytokines (such as IL-4, IL-10, and IL-13) that enhance the immune response to e.g., cancer by directly targeting tumor cells and by indirectly modulating the antitumor response through the release of diverse cytokines or by altering the TME.
  • proinflammatory cytokines such as IFN- ⁇ and TNF- ⁇
  • anti-inflammatory cytokines such as IL-4, IL-10, and IL-13
  • NKT cells In addition, many of the cytokines secreted by NKT cells have powerful effects on ⁇ T cell differentiation and function, linking NKT cells to adaptive defense. NKT cells bridge the adaptive immune system with the innate immune system. Unlike conventional T cells that recognize peptide antigens presented by major histocompatibility complex (MHC) molecules, NKT cells recognize glycolipid antigen presented by a molecule called CD1d. NKT cells can be obtained from an allogeneic or an autologous donor. The NKT cells can be partially or entirely purified, or not purified, and expanded ex vivo.
  • MHC major histocompatibility complex
  • NKT cells can be expanded via the use of ex vivo IL-2, and/or monoclonal antibodies specific for the TCR ⁇ -chain CDR3 loop (Cortés-Selva, D et al., (2021) Trends Pharmacol Sci.42(1): 45-59).
  • ⁇ natural killer T cells or “ ⁇ NKT cells” refers to iPSC- derived cells that express ⁇ TCRs and NK receptors, but lack the expression of hallmark ⁇ T cell markers (Cortés-Selva, D et al., (2021) Trends Pharmacol Sci. 42(1): 45-59).
  • CARs can be expressed in ⁇ NKT cells, in embodiments herein, for use in accordance with the methods disclosed herein.
  • myeloid cells refers to a subgroup of leukocytes represented by granulocytes, monocytes, macrophages, and dendritic cells (DCs).
  • myeloid cells can also be found in tissues under steady-state condition, where they control development, homeostasis, and tissue repair.
  • myeloid cells refers to highly plastic innate cells with functional and phenotypic signatures that can be shaped in response to various stimuli.
  • Macrophage polarization is broadly simplified into two different states, either a M1 phenotype (classically activated) in response to factors such as lipopolysaccharide (LPS) or IFN- ⁇ , or a M2 phenotype in response to cytokines such as IL-4, IL-5, and IL-13.
  • M1-like macrophages express iNOS and proinflammatory cytokines such as TNF- ⁇ , IL1- ⁇ , IL-6, IL-12, and IL-23.
  • An example of M2 macrophages exhibit increased expression of CD209, CD200R, CD1a, and CD1b in humans, and have been implicated in wound healing and antitumor responses.
  • macrophages to infiltrate solid tumors and be reprogrammed, as well as the antitumor effects associated with a switch to the M1 phenotype, render macrophages relevant to the present disclosure in terms of engineered macrophages that express a CAR described herein.
  • macrophages can be reprogrammed towards antitumor M1 phenotype cells that are capable of producing nitric oxide and inducing IL-12-dependent NK-mediated antitumor effects by inhibiting NK- ⁇ B signaling in a murine model of ovarian cancer (Zhang F et al., (2019) Nat Commun 10: 3974).
  • Macrophages can be obtained/derived from an allogeneic or an autologous donor.
  • the macrophages can be partially or entirely purified, or not purified, and cultured ex vivo (see, e.g., Davies JQ and Gordon A (2005) Methods Mol Biol 290:105016).
  • the present disclosure encompasses macrophages derived from hESCs (Karlsson, KR et al., (2008) Exp Hematol 36: 1167-1175), or iPSC-derived macrophages (Takata K. et al., (2017) Immunity 47: 183-198).
  • T lymphocyte or “T cell” refers to an immune cell that expresses or has expressed CD3 (CD3+) and a T Cell Receptor (TCR+). T cells play a central role in cell-mediated immunity. A T cell that “has expressed” CD3 and a TCR has been engineered to eliminate CD3 and/or TCR cell surface expression.
  • TCR or “T cell receptor” refers to a dimeric heterologous cell surface signaling protein forming an alpha-beta or gamma-delta receptor or combinations thereof.
  • MHC major histocompatibility complex
  • HLA human leukocyte antigen
  • activated T cells refers to, among other things, T cells that are undergoing cell division.
  • the “costimulatory domain” in the context of a chimeric receptor, also referred to herein as a chimeric antigen receptor (CAR), of the present disclosure enhances cell proliferation, cell survival and development of memory cells for cytotoxic cells that express the chimeric receptor.
  • CAR chimeric antigen receptor
  • the chimeric receptors of the invention may include one or more costimulatory domains selected from the costimulatory domains of proteins in the TNFR superfamily, CD28, CD137 (4- 1BB), CD134 (OX40), Dapl0, CD27, CD2, CD7, CD5, ICAM-1, LFA-1 (CD1 la/CD18), Lck, TNFR-I, PD-1, TNFR-II, Fas, CD30, CD40, ICOS LIGHT, NKG2C, B7-H3, or combinations thereof. If the chimeric receptor includes more than one costimulatory domain, these domains may be arranged in tandem, optionally separated by a linker.
  • costimulatory domain also encompasses any modifications thereof, examples of which are described in US Patent Application No.20200129554; US Patent Application No.20200317777; WO2019010383; Li, W., et al., (2020) Immunity 53: 456-470; and Li, G., et al., (2017) J Immunol 198(1 Supplement): 198.4, the contents of each of which are incorporated herein in their entirety.
  • the “intracellular signaling domain” in the context of a chimeric receptor of the present disclosure transduces the effector function signal and directs the cytotoxic cell to perform its specialized function, i.e., harming and/or destroying the target cells.
  • suitable intracellular signaling domains include, e.g., the ⁇ chain of the T cell receptor complex or any of its homologs, e.g., ⁇ chain, FcsRly and ⁇ chains, MB 1 (Iga) chain, B29 (Ig) chain, etc., human CD3 ⁇ chain, CD3 polypeptides ( ⁇ , ⁇ and ⁇ ), syk family tyrosine kinases (Syk, ZAP 70, etc.), src family tyrosine kinases (Lck, Fyn, Lyn, etc.) and other molecules involved in T cell transduction, such as CD2, CD5 and CD28.
  • ⁇ chain of the T cell receptor complex or any of its homologs e.g., ⁇ chain, FcsRly and ⁇ chains, MB 1 (Iga) chain, B29 (Ig) chain, etc.
  • human CD3 ⁇ chain CD3 polypeptides ( ⁇ , ⁇ and ⁇ )
  • the intracellular signaling domain of a chimeric receptor may be human CD3 ⁇ chain, FcyRIII, FcsRI, cytoplasmic tails of Fc receptors, an immunoreceptor tyrosine-based activation motif (ITAM) bearing cytoplasmic receptors and combinations thereof.
  • the intracellular signaling domains may include intracellular signaling domains of several types of various other immune signaling receptors, including, but not limited to, first, second, and third generation T cell signaling proteins including CD3, B7 family costimulatory, and Tumor Necrosis Factor Receptor (TNFR) superfamily receptors (Park et al., "Are all chimeric antigen receptors created equal?" J Clin Oncol., vol.
  • Additional intracellular signaling domains include signaling domains used by NK and NKT cells (Hermanson, et al., "Utilizing chimeric antigen receptors to direct natural killer cell activity," Front Immunol., vol. 6, p. 195, 2015) such as signaling domains of NKp30 (Zhang et al., "An NKp30-based chimeric antigen receptor promotes T cell effector functions and antitumor efficacy in vivo," J Immunol., vol. 189, pp.
  • DAP12 (Topfer et al., "DAP12-based activating chimeric antigen receptor for NK cell tumor immunotherapy,” J Immunol., vol. 194, pp. 3201- 3212, 2015), NKG2D, NKp44, NKp46, DAP10, and CD3z.
  • intracellular signaling domains also includes signaling domains of human Immunoglobulin receptors that contain immunoreceptor tyrosine based activation motif (ITAM) such as FcgammaRI, FcgammaRIIA, FcgammaRIIC, FcgammaRIIIA, FcRL5 (Gillis et al., "Contribution of Human Fc.gamma.Rs to Disease with Evidence from Human Polymorphisms and Transgenic Animal Studies," Front Immunol., vol.5, p.254, 2014).
  • ITAM immunoreceptor tyrosine based activation motif
  • the intracellular signaling domain includes a cytoplasmic signaling domain of TCR ⁇ , FcR ⁇ , FcR ⁇ , CD3 ⁇ , CD3 ⁇ , CD5, CD22, CD79a, CD79b, or CD66d.
  • the intracellular signaling domain in the chimeric receptor includes a Page 19 of 203 1097657169 ⁇ 1 ⁇ AMERICAS cytoplasmic signaling domain of human CD3 ⁇ .
  • the term “intracellular signaling domain” as used herein also encompasses any modifications thereof, examples of which are described in US Patent Application No.
  • affinity binding entity refers to a binding moiety which binds to a specific antigen with a higher affinity than to a non-specific antigen and is endowed with an affinity of at least 10 -6 M, as determined by assays which are well known in the art, including surface plasmon resonance (SPR).
  • the affinity is 500 nM-0.01 nM, 100 nM- 0.01 nM, 50 nM-0.01 nM, 10 nM-0.01 nM, 5 nM-0.01 nM.
  • the affinity binding entity is an antibody.
  • antibody is used in the broadest sense and specifically covers, for example, single anti-B7H6 monoclonal antibodies (including agonist, antagonist, neutralizing antibodies, full length or intact monoclonal antibodies), anti-B7H6 antibody compositions with polyepitopic specificity, polyclonal antibodies, multivalent antibodies, multispecific antibodies (e.g., bispecific antibodies so long as they exhibit the desired biological activity), formed from at least two intact antibodies, single chain anti-B7H6 antibodies, and fragments of anti-B7H6 antibodies (see below), including Fab, Fab’, F(ab’)2 and Fv fragments, diabodies, single domain antibodies (sdAbs), as long as they exhibit the desired biological or immunological activity.
  • single anti-B7H6 monoclonal antibodies including agonist, antagonist, neutralizing antibodies, full length or intact monoclonal antibodies
  • anti-B7H6 antibody compositions with polyepitopic specificity polyclonal antibodies
  • multivalent antibodies e.g., bispecific antibodies so long as they exhibit
  • anti-B7H6 antibodies are also included among anti-B7H6 antibodies, and among fragments in particular, are portions of anti-B7H6 antibodies (and combinations of portions of anti-B7H6 antibodies, for example, scFv) that may be used as targeting arms, directed to e.g., a B7H6 epitope, in chimeric antigenic receptors of the present disclosure.
  • Such fragments are not necessarily proteolytic fragments but rather portions of polypeptide sequences that can confer affinity for target.
  • immunoglobulin Ig
  • An antibody can be, for example, human, humanized and/or affinity matured. [0099] Methods of making antibodies and antibody fragments are known in the art.
  • Antibodies can be produced by the immunization of various animals, including mice, rats, rabbits, goats, primates, humans and chickens with a target antigen such as B7H6 or peptide fragments of B7H6 containing the anti-B7H6 epitope of the present disclosure. Antibodies may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352: 624-8 (1991) and Marks et al., J. Mol.
  • Antibodies or antigen-binding fragment of the present invention can be purified by methods known in the art, for example, gel filtration, ion exchange, affinity chromatography, etc. Affinity chromatography or any of a number of other techniques known in the art can be used to isolate polyclonal or monoclonal antibodies from, for example, serum, ascites fluid, or hybridoma supernatants. [00101]
  • the terms “anti-B7H6 antibody”, “B7H6 antibody”, and “an antibody that binds to B7H6” are used interchangeably.
  • Anti-B7H6 antibodies are preferably capable of binding with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent, whether in isolation or as part of fusion protein, cell, or cell composition.
  • An “isolated antibody” is one which has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials which would interfere with therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes.
  • the basic 4-chain antibody unit is a heterotetrameric glycoprotein composed of two identical light (L) chains and two identical heavy (H) chains. In the case of IgGs, the 4-chain unit is generally about 150,000 daltons.
  • Each L chain is linked to a H chain by one covalent disulfide bond, while the two H chains are linked to each other by one or more disulfide bonds depending on the H chain isotype.
  • Each H and L chain also has regularly spaced intrachain disulfide bridges.
  • Each H chain has at the N-terminus, a variable domain (VH) followed by three constant domains (CH) for each of the ⁇ and ⁇ chains and four CH domains for ⁇ and ⁇ isotypes.
  • Each L chain has at the N-terminus, a variable domain (VL) followed by a constant domain (CL) at its other end. The VL is aligned with the VH and the CL is aligned with the first constant domain of the heavy chain (CH1).
  • immunoglobulins can be assigned to different classes or isotypes. There are five classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, having heavy chains designated ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • the ⁇ and ⁇ classes are further divided into subclasses on the basis of relatively minor differences in CH sequence and function, e.g., humans express the following subclasses: IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2.
  • variable region refers to the amino-terminal domains of the heavy or light chain of the antibody.
  • variable domain of the heavy chain may be referred to as “VH” or “VH”
  • variable domain of the light chain may be referred to as “VL” or “V L ”. These domains are generally the most variable parts of an antibody and contain the antigen-binding sites.
  • variable refers to the fact that certain segments of the variable domains differ extensively in sequence among antibodies. The V domain mediates antigen binding and defines specificity of a particular antibody for its particular antigen. However, the variability is not evenly distributed across the 110-amino acid span of the variable domains.
  • variable regions consist of relatively invariant stretches called framework regions (FRs) of 15-30 amino acids separated by shorter regions of extreme variability called “hypervariable regions” that are each about 9-12 amino acids long.
  • the variable domains of native heavy and light chains each comprise four FRs, largely adopting a ⁇ -sheet configuration, connected by three hypervariable regions, which form loops connecting, and in some cases forming part of, the ⁇ -sheet structure.
  • the hypervariable regions in each chain are held together in close proximity by the FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed.
  • an “intact” antibody is one which comprises an antigen-binding site as well as a CL and at least heavy chain constant domains, CH1, CH2 and CH3.
  • the constant domains may be Page 22 of 203 1097657169 ⁇ 1 ⁇ AMERICAS native sequence constant domains (e.g., human native sequence constant domains) or amino acid sequence variant thereof.
  • the intact antibody has one or more effector functions.
  • “Antibody fragments” comprise a portion of an intact antibody, preferably the antigen binding or one or more variable regions of the intact antibody.
  • antibody fragments include Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies (see U.S. Patent No. 5,641,870, Example 2; Zapata et al., Protein Eng.8(10): 1057-62 (1995)); single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • an antibody fragment comprises an antigen binding site of the intact antibody and thus retains the ability to bind antigen.
  • anti-B7H6 antibody fragments are portions of anti- B7H6 antibodies (and combinations of portions of anti-B7H6 antibodies, for example, scFv) that may be used as targeting arms, directed to e.g., a B7H6 epitope, in chimeric antigenic receptors of the present disclosure.
  • Such fragments are not necessarily proteolytic fragments but rather portions of polypeptide sequences that can confer affinity for target.
  • Papain digestion of antibodies produces two identical antigen-binding fragments, called “Fab” fragments, and a residual “Fc” fragment, a designation reflecting the ability to crystallize readily.
  • the Fab fragment consists of an entire L chain along with the variable region domain of the H chain (VH), and the first constant domain of one heavy chain (CH1). Each Fab fragment is monovalent with respect to antigen binding, i.e., it has a single antigen-binding site. Pepsin treatment of an antibody yields a single large F(ab')2 fragment which roughly corresponds to two disulfide linked Fab fragments having divalent antigen-binding activity and is still capable of cross-linking antigen.
  • Fab’ fragments differ from Fab fragments by having additional few residues at the carboxy terminus of the CH1 domain including one or more cysteines from the antibody hinge region.
  • Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear a free thiol group.
  • F(ab')2 antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
  • the Fc fragment comprises the carboxy-terminal portions of both H chains held together by disulfides. The effector functions of antibodies are determined by sequences in the Fc region, which region is also the part recognized by Fc receptors (FcR) found on certain types of cells.
  • Fv is the minimum antibody fragment which contains a complete antigen-recognition and -binding site. This fragment consists of a dimer of one heavy- and one light-chain variable region domain in tight, non-covalent association.
  • scFv single-chain Fv
  • one heavy- and one light-chain variable domain can be covalently linked by a flexible peptide linker such that the light and heavy chains can associate in a “dimeric” structure analogous to that in a two-chain Fv species.
  • Single-chain Fv also abbreviated as “sFv” or “scFv” are antibody fragments that comprise the VH and VL antibody domains connected into a single polypeptide chain.
  • the sFv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the sFv to form a desired structure for antigen binding.
  • a polypeptide linker between the VH and VL domains which enables the sFv to form a desired structure for antigen binding.
  • the present disclosure also includes the alternate embodiment in which the order of VH and VL regions are reversed, e.g., in an scFv or CAR comprising an scFv binding domain.
  • description of a VH- VL order also describes the alternate VL-VH order, e.g., in an scFv or CAR comprising an scFv binding domain.
  • description of a VL-VH order also describes the alternate VH-VL order, e.g., in an scFv or a CAR comprising an scFv binding domain.
  • VH and VL regions are either joined directly or joined by a peptide-encoding linker, which connects the N-terminus of the VH with the C-terminus of the VL, or the C-terminus of the VH with the N-terminus of the VL.
  • the scFv linker is usually rich in glycine for flexibility, as well as serine or threonine for solubility.
  • the linker can link the heavy chain variable region and the light chain variable region of the extracellular antigen-binding domain.
  • Non-limiting examples of linkers are disclosed in Shen et al, Anal. Chem.80(6): 1910-1917 (2008) and WO 2014/087010, the contents of which are hereby incorporated by reference in their entireties.
  • GS linker sequences are known in the Page 24 of 203 1097657169 ⁇ 1 ⁇ AMERICAS art, including, without limitation, glycine serine (GS) linkers such as (GS)n, (GSGGS)n (SEQ ID NO: 174), (GGGS)n (SEQ ID NO: 175), and (GGGGS)n (SEQ ID NO: 176), where n represents an integer of at least 1.
  • GS glycine serine
  • Exemplary linker sequences can comprise amino acid sequences including, without limitation, GGSG (SEQ ID NO: 177), GGSGG (SEQ ID NO: 178), GSGSG (SEQ ID NO: 179), GSGGG (SEQ ID NO: 180), GGGSG (SEQ ID NO: 181), GSSSG (SEQ ID NO: 182), GGGGS (SEQ ID NO: 183), GGGGSGGGGSGGGGS (SEQ ID NO: 172) and the like.
  • GGSG SEQ ID NO: 177
  • GGSGG SEQ ID NO: 178
  • GSGSG SEQ ID NO: 179
  • GSGGG SEQ ID NO: 180
  • GGGSG SEQ ID NO: 181
  • GSSSG SEQ ID NO: 182
  • GGGGS SEQ ID NO: 183
  • GGGGSGGGGSGGGGS SEQ ID NO: 172
  • an antigen binding domain of the present invention comprises a heavy chain variable region (VH) and a light chain variable region (VL), wherein the VH and VL is separated by the linker sequence having the amino acid sequence GGGGSGGGGSGGGGS (SEQ ID NO: 172), which may be encoded by the nucleic acid sequence GGAGGCGGAGGATCTGGTGGTGGTGGATCTGGCGGCGGAGGCTCT (SEQ ID NO: 173).
  • the term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts.
  • Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations which include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they may be synthesized uncontaminated by other antibodies.
  • the modifier “monoclonal” is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies useful in the present invention may be prepared by the hybridoma methodology first described by Kohler et al., Nature, 256: 495 (1975), or may be made using recombinant DNA methods in bacterial, eukaryotic animal or plant cells (e.g., U.S. Patent No.4,816,567).
  • the “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352: 624-8 (1991) and Marks et al., J. Mol. Biol., 222: 581-97 (1991), for example.
  • hypervariable region when used herein refers to the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops.
  • antibodies comprise six hypervariable regions; three in the VH (H1, H2, H3), and three in the VL (L1, L2, L3).
  • a number of hypervariable region delineations Page 25 of 203 1097657169 ⁇ 1 ⁇ AMERICAS are in use and are encompassed herein.
  • the Kabat Complementarity Determining Regions CDRs are based on sequence variability and are the most commonly used (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed.
  • Chothia refers instead to the location of the structural loops (Chothia and Lesk J. Mol. Biol. 196:901-917 (1987)).
  • the end of the Chothia CDR-H1 loop when numbered using the Kabat numbering convention varies between H32 and H34 depending on the length of the loop (this is because the Kabat numbering scheme places the insertions at H35A and H35B; if neither 35A nor 35B is present, the loop ends at 32; if only 35A is present, the loop ends at 33; if both 35A and 35B are present, the loop ends at 34).
  • the AbM hypervariable regions represent a compromise between the Kabat CDRs and Chothia structural loops, and are used by Oxford Molecular’s AbM antibody modeling software.
  • the “contact” hypervariable regions are based on an analysis of the available complex crystal structures. The residues from each of these hypervariable regions are noted below.
  • Hypervariable regions may comprise “extended hypervariable regions” as follows: 24- 36 or 24-34 (L1), 46-56 or 50-56 (L2) and 89-97 (L3) in the VL and 26-35
  • variable domain residues are numbered according to Kabat et al., supra, for each of these definitions.
  • “Framework” or “FR” residues are those variable domain residues other than the hypervariable region residues herein defined.
  • a heavy chain variable domain may include a single amino acid insert (residue 52a according to Kabat) after residue 52 of H2 and inserted residues (e.g., residues 82a, 82b, and 82c, etc. according to Kabat) after heavy chain FR residue 82.
  • the Kabat numbering of residues may be determined for a given antibody by alignment at regions of homology of the sequence of the antibody with a “standard” Kabat numbered sequence. [00119]
  • the Kabat numbering system is generally used when referring to a residue in the variable domain (approximately residues 1-107 of the light chain and residues 1-113 of the heavy chain) (e.g, Kabat et al., supra).
  • EU numbering system or “EU index” is generally used when referring to a residue in an immunoglobulin heavy chain constant region (e.g., the EU index reported in Kabat et al., supra).
  • the “EU index as in Kabat” refers to the residue numbering of the human IgG1 EU antibody. Unless stated otherwise herein, references to residue numbers in the variable domain of antibodies means residue numbering by the Kabat numbering system.
  • a “blocking” antibody or an “antagonist” antibody is one which inhibits or reduces biological activity of the antigen it binds. Preferred blocking antibodies or antagonist antibodies substantially or completely inhibit the biological activity of the antigen.
  • an anti-B7H6 antibody is provided, which is an antagonist antibody.
  • An antibody that “binds” an antigen or epitope of interest is one that binds the antigen or epitope with sufficient affinity that is measurably different from a non-specific interaction. Specific binding can be measured, for example, by determining binding of a molecule compared to binding of a control molecule, which generally is a molecule of similar structure that does not have binding activity. Page 27 of 203 1097657169 ⁇ 1 ⁇ AMERICAS [00122]
  • the term “antigen” or “Ag” as used herein is defined as a molecule that provokes an immune response. This immune response may involve either antibody production, or the activation of specific immunologically-competent cells, or both.
  • epitope includes any protein determinant, lipid or carbohydrate determinant capable of specific binding to an immunoglobulin or T cell receptor. Epitopic determinants usually consist of active surface groupings of molecules such as amino acids, lipids or sugar side chains and usually have specific three-dimensional structural characteristics, as well as specific charge characteristics.
  • the term "specifically binds”, as used herein refers to a receptor (which can include but is not limited to an antibody or antibody fragment) which recognizes a specific molecule/ligand, but does not substantially recognize or bind other molecules in a sample.
  • a receptor that specifically binds to a molecule from one species may also bind to that molecule from one or more other species. But, such cross-species reactivity does not itself alter the classification as specific.
  • a receptor that specifically binds to a molecule may also bind to different allelic forms of the molecule. However, such cross reactivity does not itself alter the classification as specific.
  • the terms “specific binding” or “specifically binding,” can be used in reference to the interaction of a protein (or a peptide) with a second chemical species, to mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, a receptor recognizes and binds to a specific a structure rather than to proteins generally. If receptor is specific for epitope "A”, the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled "A” and the receptor, will reduce the amount of labeled A bound to the receptor.
  • a particular structure e.g., an antigenic determinant or epitope
  • specific binding can be characterized by an equilibrium dissociation constant of at least about 1x10 -8 M or less (e.g., a smaller KD denotes a tighter binding).
  • Methods for determining whether two molecules specifically bind are well known in the art and include, for example, equilibrium dialysis, surface plasmon resonance, and the like.
  • Page 28 of 203 1097657169 ⁇ 1 ⁇ AMERICAS [00126]
  • cancer and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include but are not limited to carcinoma, lymphoma, blastoma, sarcoma (including liposarcoma), neuroendocrine tumors, mesothelioma, schwanoma, meningioma, adenocarcinoma, melanoma, and leukemia or lymphoid malignancies.
  • Cancers can include but are not limited to colon cancer, pancreatic cancer, Non-Hodgkin Lymphoma, cervical cancer, hepatic cancer, gastro-intestinal cancer, lung cancer, ovarian cancer, glioblastoma, prostate cancer, breast cancer, ovarian cancer, kidney cancer, bladder cancer, melanoma, and glioma.
  • autologous is meant to refer to any material derived from an individual which is later to be re-introduced into the same individual.
  • allogeneic refers to material derived from an animal which is later introduced into a different animal of the same species.
  • a “modification” of an amino acid residue/position refers to a change of a primary amino acid sequence as compared to a starting amino acid sequence, wherein the change results from a sequence alteration involving the amino acid residue/positions.
  • typical modifications include substitution of the residue (or at the position) with another amino acid (e.g., a conservative or non-conservative substitution), insertion of one or more (generally fewer than 5 or 3) amino acids adjacent to the residue/position, and deletion of the residue/position.
  • An “amino acid substitution”, or variation thereof refers to the replacement of an existing amino acid residue in a predetermined (starting) amino acid sequence with a different amino acid residue.
  • the modification results in alteration in at least one physicobiochemical activity of the variant polypeptide compared to a polypeptide comprising the starting (or “wild type”) amino acid sequence.
  • a physicobiochemical activity that is altered can be binding affinity, binding capability and/or binding effect upon a target molecule.
  • Page 29 of 203 1097657169 ⁇ 1 ⁇ AMERICAS [00131]
  • a therapy treats a disease or condition by decreasing one or more signs or symptoms associated with the disease or condition, for example as compared to the response in the absence of the therapy.
  • administration of a therapeutic agent may provide an anti-tumor effect that decreases one or more signs or symptoms associated with cancer.
  • Treating or preventing can refer to delaying the onset of symptoms, reducing the severity of symptoms, reducing the severity of an acute episode, reducing the number of symptoms, reducing the incidence of disease-related symptoms, reducing the latency of symptoms, ameliorating symptoms, reducing secondary symptoms, reducing secondary infections, prolonging patient survival, preventing relapse to a disease, decreasing the number or frequency of relapse episodes, increasing latency between symptomatic episodes, increasing time to sustained progression, expediting remission, inducing remission, augmenting remission, speeding recovery, or increasing efficacy of or decreasing resistance to alternative therapeutics.
  • treating refers to both therapeutic treatment and prophylactic or preventive measures, wherein the object is to prevent or lessen the targeted pathologic condition or disorder as described herein.
  • administration means to provide or give a subject one or more agents, such as an agent that treats one or more signs or symptoms associated with a condition/disorder or disease including but not limited to cancer (e.g., lymphoma), viral infection, bacterial infection, etc., by any effective route.
  • routes of administration include, but are not limited to, injection (such as subcutaneous, intramuscular, intradermal, intraperitoneal, and intravenous), oral, sublingual, rectal, transdermal, intranasal, vaginal and inhalation routes.
  • compositions in combination with include simultaneous (concurrent) and sequential administration in any order.
  • pharmaceutically acceptable refers to a material, including but not limited, to a salt, carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively nontoxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • pharmaceutically acceptable carriers (vehicles) useful in this disclosure are conventional. Page 30 of 203 1097657169 ⁇ 1 ⁇ AMERICAS Remington's Pharmaceutical Sciences, by E. W.
  • parenteral formulations can include injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle.
  • pharmaceutical agents to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate, sodium lactate, potassium chloride, calcium chloride, and triethanolamine oleate.
  • auxiliary substances such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate, sodium lactate, potassium chloride, calcium chloride, and triethanolamine oleate.
  • the present invention provides a pharmaceutical composition comprising a pharmaceutically acceptable excipient and an, e.g., T cell, preferably a ⁇ T cell, engineered to express a CAR directed to B7H6, as described herein.
  • Encoding refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom.
  • a gene encodes a protein if transcription and translation of mRNA corresponding to that gene produces the protein in a cell or other biological system.
  • Both the coding strand, the nucleotide sequence of which is identical to the mRNA sequence and is usually provided in sequence listings, and the non-coding strand, used as the template for transcription of a gene or cDNA, can be referred to as encoding the protein or other product of that gene or cDNA.
  • “Isolated” means altered or removed from the natural state.
  • nucleic acid or a peptide naturally present in a living animal is not “isolated,” but the same nucleic acid or peptide partially or completely separated from the coexisting materials of its natural state is “isolated.”
  • An isolated nucleic acid or protein can exist in substantially purified form, or can exist in a non-native environment such as, for example, a host cell.
  • a “nucleotide sequence encoding an amino acid sequence” includes all nucleotide sequences that are degenerate versions of each other and that encode the Page 31 of 203 1097657169 ⁇ 1 ⁇ AMERICAS same amino acid sequence. Nucleotide sequences that encode proteins and RNA may include introns.
  • “patient,” “subject,” “individual,” and the like are used interchangeably herein, and refer to any animal, amenable to the methods described herein.
  • the patient, subject or individual is a human.
  • “Expression cassette” refers to a nucleic acid comprising expression control sequences operatively linked to a nucleic acid encoding a transcript or polypeptide to be expressed.
  • An expression cassette comprises sufficient cis-acting elements for expression; other elements for expression can be supplied by the host cell or in an in vitro expression system.
  • Expression cassettes can be a component of a vector such as a cosmid, a plasmid (e.g., naked or contained in a liposome), or a virus (e.g., lentivirus, retrovirus, adenovirus, and adeno-associated virus).
  • An expression cassette can be in a host cell, such as a ⁇ T cell.
  • Polyclonal Antibodies may be raised in animals by multiple subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen and an adjuvant. It may be useful to conjugate the relevant antigen (especially when synthetic peptides are used) to a protein that is immunogenic in the species to be immunized.
  • the antigen can be conjugated to keyhole limpet hemocyanin (KLH), serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor, using a bifunctional or derivatizing agent, e.g., maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (through lysine residues), glutaraldehyde, succinic anhydride, SOCl2, or R′N ⁇ C ⁇ NR, where R and R1 are different alkyl groups.
  • KLH keyhole limpet hemocyanin
  • serum albumin serum albumin
  • bovine thyroglobulin bovine thyroglobulin
  • soybean trypsin inhibitor e.g., a bifunctional or derivatizing agent, e.g., maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues),
  • Animals are immunized against the antigen, immunogenic conjugates, or derivatives by combining, e.g., 100 ⁇ g or 5 ⁇ g of the protein or conjugate (for rabbits or mice, respectively) with 3 volumes of Freund’s complete adjuvant and injecting the solution intradermally at multiple Page 32 of 203 1097657169 ⁇ 1 ⁇ AMERICAS sites.
  • the animals are boosted with 1 ⁇ 5 to 1/10 the original amount of peptide or conjugate in Freund’s complete adjuvant by subcutaneous injection at multiple sites.
  • the animals are bled and the serum is assayed for antibody titer. Animals are boosted until the titer plateaus.
  • a monoclonal antibody (mAb) to an antigen-of-interest can be prepared by using any technique known in the art. These include, but are not limited to, the hybridoma technique originally described by Kohler and Milstein (1975, Nature 256, 495-497), the human B cell hybridoma technique (Kozbor et al., 1983, Immunology Today 4: 72), and the EBV-hybridoma technique (Cole et al., 1985, Monoclonal Antibodies and Cancer Therapy, Alan R.
  • Selected Lymphocyte Antibody Method (SLAM) (Babcook, J.S., et al., A novel strategy for generating monoclonal antibodies from single, isolated lymphocytes producing antibodies of defined specificities. Proc Natl Acad Sci U S A, 1996. 93 (15): p. 7843-8. ) and (McLean G et al., 2005, J Immunol. 174(8): 4768-78.
  • Such antibodies may be of any immunoglobulin class including IgG, IgM, IgE, IgA, and IgD and any subclass thereof.
  • the hybridoma producing the mAbs of use in this invention may be cultivated in vitro or in vivo.
  • Monoclonal antibodies may be made using the hybridoma method first described by Kohler et al., Nature, 256: 495 (1975), or may be made by recombinant DNA methods (U.S. Pat. No.4,816,567).
  • a mouse or other appropriate host animal such as a hamster, is immunized as described above to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization.
  • lymphocytes may be immunized in vitro.
  • lymphocytes are isolated and then fused with a myeloma cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp.59-103 (Academic Press, 1986)).
  • a suitable fusing agent such as polyethylene glycol
  • the hybridoma cells thus prepared are seeded and grown in a suitable culture medium which may contain one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells (also referred to as fusion partner).
  • the selective culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • HGPRT hypoxanthine guanine phosphoribosyl transferase
  • Preferred fusion partner myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a selective medium that selects against the unfused parental cells.
  • Preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, Calif. USA, and SP-2 and derivatives e.g., X63-Ag8-653 cells available from the American Type Culture Collection, Manassas, Va., USA.
  • Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor, J.
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen.
  • the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunosorbent assay (ELISA).
  • the binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis described in Munson et al., Anal. Biochem.107: 220 (1980). [00149] Once hybridoma cells that produce antibodies of the desired specificity, affinity, and/or activity are identified, the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)). Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium.
  • the hybridoma cells may be grown in vivo as ascites tumors in an animal, e.g., by intraperitoneal injection of the cells into mice.
  • the monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by conventional antibody purification procedures such as, for example, affinity chromatography (e.g., using protein A or protein G-Sepharose) or ion- exchange chromatography, hydroxylapatite chromatography, gel electrophoresis, dialysis, etc.
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies).
  • the hybridoma cells serve as a preferred source of such DNA.
  • the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese Hamster Ovary (CHO) cells, or myeloma cells that do not otherwise produce antibody protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • monoclonal antibodies or antibody fragments can be isolated from antibody phage libraries generated using the techniques described in McCafferty et al., Nature, 348: 552-54 (1990). Clackson et al., Nature, 352: 624-28 (1991) and Marks et al., J. Mol. Biol., 222: 581-97 (1991) describe the isolation of murine and human antibodies, respectively, using phage libraries.
  • the DNA that encodes the antibody may be modified to produce chimeric or fusion antibody polypeptides, for example, by substituting human heavy chain and light chain constant domain (CH and CO sequences for the homologous murine sequences (U.S. Pat. No.4,816,567; and Morrison, et al., Proc. Natl. Acad. Sci. USA, 81: 6851 (1984)), or by fusing the immunoglobulin coding sequence with all or part of the coding sequence for a non- immunoglobulin polypeptide (heterologous polypeptide).
  • CH and CO sequences for the homologous murine sequences
  • heterologous polypeptide heterologous polypeptide
  • the non-immunoglobulin polypeptide sequences can substitute for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen.
  • Chimeric, Humanized, and Human Antibodies is a chimeric antibody. Certain chimeric antibodies are described, e.g., in U.S. Pat. No.4,816,567; and Morrison et al., Proc. Natl. Acad. Sci.
  • a chimeric antibody comprises a non-human variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) and a human constant region.
  • a chimeric antibody is a “class switched” antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen-binding fragments thereof.
  • a chimeric antibody is a humanized antibody.
  • a non-human antibody is humanized to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody.
  • a humanized antibody comprises one or more variable domains in which HVRs, e.g., CDRs, (or portions thereof) are derived from a non- human antibody, and FRs (or portions thereof) are derived from human antibody sequences.
  • HVRs e.g., CDRs
  • FRs or portions thereof
  • a humanized antibody optionally will also comprise at least a portion of a human constant region.
  • some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the CDR residues are derived), e.g., to restore or improve antibody specificity or affinity.
  • the anti-B7H6 antibodies of the invention may comprise humanized antibodies or human antibodies.
  • Humanized forms of non-human (e.g., murine or rabbit) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab')2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin.
  • Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity.
  • CDR complementary determining region
  • donor antibody such as mouse, rat or rabbit having the desired specificity, affinity and capacity.
  • Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • Humanized antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all Page 36 of 203 1097657169 ⁇ 1 ⁇ AMERICAS or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin (Jones et al., Nature, 321: 522-5 (1986); Riechmann et al., Nature, 332: 323-9 (1988); and Presta, Curr. Op. Struct. Biol., 2: 593-6 (1992)).
  • Fc immunoglobulin constant region
  • a humanized antibody of the invention may comprise one or more human and/or human consensus non-hypervariable region (e.g., framework) sequences in its heavy and/or light chain variable domain. In embodiments, one or more additional modifications are present within the human and/or human consensus non-hypervariable region sequences.
  • the heavy chain variable domain of an antibody of the invention comprises a human consensus framework sequence, which in one embodiment is the subgroup III consensus framework sequence.
  • an antibody of the invention comprises a variant subgroup III consensus framework sequence modified at least one amino acid position.
  • the amino acid position/boundary delineating a hypervariable region of an antibody can vary, depending on the context and the various definitions known in the art. Some positions within a variable domain may be viewed as hybrid hypervariable positions in that these positions can be deemed to be within a hypervariable region under one set of criteria while being deemed to be outside a hypervariable region under a different set of criteria.
  • these positions can also be found in extended hypervariable regions (as further defined below).
  • the invention provides antibodies comprising modifications in these hybrid hypervariable positions.
  • these hypervariable positions include one or more positions 26-30, 33-35B, 47-49, 57-65, 93, 94 and 101-102 in a heavy chain variable domain.
  • these hybrid hypervariable positions include one or more of positions 24-29, 35-36, 46-49, 56 and 97 in a light chain variable domain.
  • an antibody of the invention comprises a human variant human subgroup consensus framework sequence modified at one or more hybrid hypervariable positions.
  • An antibody of the invention can comprise any suitable human or human consensus light chain framework sequences, provided the antibody exhibits the desired biological characteristics (e.g., a desired binding affinity).
  • an antibody of the invention comprises at least a portion (or all) of the framework sequence of human ⁇ light chain.
  • an antibody of the invention comprises at least a portion (or all) of human ⁇ subgroup I framework consensus sequence.
  • Methods for humanizing non-human antibodies are well known in the art. As discussed, a humanized antibody generally has one or more amino acid residues introduced into it from a source which is non-human.
  • humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • HAMA response human anti- mouse antibody
  • the invention provides antibodies that are humanized such that HAMA response is reduced or eliminated.
  • Variants of these antibodies can further be obtained using routine methods known in the art, some of which are further described below.
  • the sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable domain sequences.
  • the human V domain sequence which is closest to that of the rodent is identified and the human framework region (FR) within it accepted for the humanized antibody (Sims et al., J. Immunol. 151: 2296 (1993); Chothia et al., J. Mol. Biol., 196: 901 (1987)).
  • Another method uses a particular framework Page 38 of 203 1097657169 ⁇ 1 ⁇ AMERICAS region derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains.
  • the same framework may be used for several different humanized antibodies (Carter et al., Proc. Natl. Acad. Sci. USA, 89: 4285 (1992); Presta et al., J. Immunol. 151: 2623 (1993)).
  • an amino acid sequence from an antibody as described herein can serve as a starting (parent) sequence for diversification of the framework and/or hypervariable sequence(s).
  • a selected framework sequence to which a starting hypervariable sequence is linked is referred to herein as an acceptor human framework.
  • acceptor human frameworks may be from, or derived from, a human immunoglobulin (the VL and/or VH regions thereof), preferably the acceptor human frameworks are from, or derived from, a human consensus framework sequence as such frameworks that have been demonstrated to have minimal, or no, immunogenicity in human patients.
  • the acceptor is derived from a human immunoglobulin, one may optionally select a human framework sequence that is selected based on its homology to the donor framework sequence by aligning the donor framework sequence with various human framework sequences in a collection of human framework sequences, and select the most homologous framework sequence as the acceptor.
  • human consensus frameworks herein are from, or derived from, VH subgroup III and/or VL kappa subgroup I consensus framework sequences.
  • the acceptor may be identical in sequence to the human framework sequence selected, whether that be from a human immunoglobulin or a human consensus framework, the present invention contemplates that the acceptor sequence may comprise pre-existing amino acid substitutions relative to the human immunoglobulin sequence or human consensus framework sequence. These pre-existing substitutions are preferably minimal; usually four, three, two or one amino acid differences only relative to the human immunoglobulin sequence or consensus framework sequence.
  • Hypervariable region residues of the non-human antibody are incorporated into the VL and/or VH acceptor human frameworks.
  • the extended hypervariable region residues as follows are Page 39 of 203 1097657169 ⁇ 1 ⁇ AMERICAS incorporated: 24-34 (L1), 50-56 (L2) and 89-97 (L3), 26-35B (H1), 50-65, 47-65 or 49-65 (H2) and 93-102, 94-102, or 95-102 (H3).
  • nucleic acid encoding the desired amino acid sequence can be generated by mutating nucleic acid encoding the mouse variable domain sequence so that the framework residues thereof are changed to acceptor human framework residues, or by mutating nucleic acid encoding the human variable domain sequence so that the hypervariable domain residues are changed to non-human residues, or by synthesizing nucleic acid encoding the desired sequence, etc.
  • hypervariable region-grafted variants may be generated by Kunkel mutagenesis of nucleic acid encoding the human acceptor sequences, using a separate oligonucleotide for each hypervariable region. Kunkel et al., Methods Enzymol. 154:367-382 (1987). Appropriate changes can be introduced within the framework and/or hypervariable region, using routine techniques, to correct and re-establish proper hypervariable region-antigen interactions.
  • Phage(mid) display also referred to herein as phage display in some contexts
  • phage display can be used as a convenient and fast method for generating and screening many different potential variant antibodies in a library generated by sequence randomization.
  • Phage(mid) display technology has provided a powerful tool for generating and selecting novel proteins which bind to a ligand, such as an antigen. Using the techniques of phage(mid) display allows the generation of large libraries of protein variants which can be rapidly sorted for those sequences that bind to a target molecule with high affinity. Nucleic acids encoding variant polypeptides are generally fused to a nucleic acid sequence encoding a viral coat protein, such as the gene III protein or the gene VIII protein.
  • Monovalent phagemid display systems where the nucleic acid sequence encoding the protein or polypeptide is fused to a nucleic acid sequence encoding a portion of the gene III protein have been developed. (Bass, S., Proteins, 8:309 (1990); Lowman and Wells, Methods: A Companion to Methods in Enzymology, 3:205 (1991)).
  • the gene fusion is expressed at low levels and wild type gene III proteins are also expressed so that infectivity of the particles is retained.
  • Methods of Page 40 of 203 1097657169 ⁇ 1 ⁇ AMERICAS generating peptide libraries and screening those libraries have been disclosed in many patents (e.g., U.S. Pat.
  • the library is then screened for expression of antibodies or antigen binding proteins with the desired characteristics.
  • Methods of substituting an amino acid of choice into a template nucleic acid are well established in the art, some of which are described herein.
  • methods for introducing modifications into nucleic acid sequences can include the use of various kits available for purchase (e.g., QuickChange Site Directed Mutagenesis Kit, Agilent, Santa Clara, CA).
  • hypervariable region residues can be substituted using the Kunkel method (e.g., Kunkel et al., Methods Enzymol.154:367-382 (1987)).
  • Kunkel method e.g., Kunkel et al., Methods Enzymol.154:367-382 (1987)
  • humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences.
  • Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art.
  • Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen.
  • FR residues can be selected and combined from the recipient and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved.
  • the hypervariable region residues are directly and most substantially involved in influencing antigen binding.
  • Page 41 of 203 1097657169 ⁇ 1 ⁇ AMERICAS [00174]
  • the humanized antibody may be an antibody fragment, such as a Fab.
  • the humanized antibody may be an intact antibody, such as an intact IgG1 antibody.
  • human antibodies can be generated. For example, it is now possible to produce transgenic animals (e.g., mice) that are capable, upon immunization, of producing a full repertoire of human antibodies in the absence of endogenous immunoglobulin production.
  • JH antibody heavy-chain joining region
  • phage display technology can be used to produce human antibodies and antibody fragments in vitro, from immunoglobulin variable (V) domain gene repertoires from unimmunized donors.
  • V domain genes are cloned in-frame into either a major or minor coat protein gene of a filamentous bacteriophage, such as M13 or fd, and displayed as functional antibody fragments on the surface of the phage particle.
  • the filamentous particle contains a single-stranded DNA copy of the phage genome, selections based on the functional properties of the antibody also result in selection of the gene encoding the antibody exhibiting those properties.
  • the phage mimics some of the properties of the B-cell.
  • Phage display can be performed in a variety of formats, reviewed in, e.g., Johnson, Kevin S, and Chiswell, David J., Current Opinion in Structural Biology 3:564-571 (1993).
  • V-gene segments can be used for phage display.
  • Clackson et al., Nature, 352:624-628 (1991) isolated a diverse array of anti-oxazolone antibodies from a small random combinatorial library of V genes derived from the spleens of immunized mice.
  • a repertoire of V genes from unimmunized human donors can be constructed and antibodies to a diverse array of antigens (including self-antigens) can be isolated essentially following the techniques described by Marks et al., J. Mol. Biol. 222:581-97 (1991), or Griffith et al., EMBO J. 12: 725-34 (1993) (see also, U.S. Pat. Nos.5,565,332 and 5,573,905).
  • Human antibodies may also be generated by in vitro activated B cells (see, e.g., U.S. Pat. Nos.5,567,610 and 5,229,275).
  • human monoclonal antibodies directed against B7H6 may be generated using transgenic or transchromosomic mice carrying parts of the human immune system rather than the mouse system.
  • the HuMAb MouseTM (Medarex, Inc.) contains human immunoglobulin gene miniloci that encode unrearranged human heavy ( ⁇ and ⁇ ) and ⁇ light chain immunoglobulin sequences, together with targeted mutations that inactivate the endogenous ⁇ and ⁇ chain loci (see e.g., Lonberg, et al. (1994) Nature 368(6474): 856-9). Accordingly, the mice exhibit reduced expression of mouse IgM or ⁇ , and in response to immunization, the introduced human heavy and light chain transgenes undergo class switching and somatic mutation to generate high affinity human IgG ⁇ monoclonal antibodies (Lonberg, N. et al. (1994), supra; reviewed in Lonberg, N.
  • human antibodies of this disclosure can be raised using a mouse referred to as “KM MouseTM” that carries human immunoglobulin sequences on transgenes and transchromosomes, as described in detail in PCT Publication WO 02/43478. Page 43 of 203 1097657169 ⁇ 1 ⁇ AMERICAS [00181]
  • an alternative transgenic system referred to as the Xenomouse (Abgenix, Inc.) can be used; such mice are described in, for example, U.S. Pat. Nos. 5,939,598; 6,075,181; 6,114,598; 6,150,584 and 6,162,963.
  • mice carrying both a human heavy chain transchromosome and a human light chain tranchromosome referred to as “TC mice” can be used; such mice are described in Tomizuka et al. (2000) Proc. Natl. Acad. Sci. USA 97: 722-7.
  • cows carrying human heavy and light chain transchromosomes have been described in the art (e.g., Kuroiwa et al. (2002) Nature Biotechnology 20: 889-94 and PCT application No.
  • WO 2002/092812 can be used to raise anti-B7H6 antibodies of this disclosure.
  • Additional examples of transgenic animals that can be used to produce anti-B7H6 antibodies include OmniRat TM and OmniMouse TM (see e.g., Osborn M., et al. (2013) Journal of Immunology 190: 1481-90; Ma B., et al. (2013) Journal of Immunological Methods 400-401: 78-86; Geurts A., et al. (2009) Science 325: 433, U.S. Pat. No. 8,907,157; European Pat. No. 2152880B1; European Pat. No. 2336329B1).
  • VELOCIMMUNE® Technology involves generation of a transgenic mouse having a genome comprising human heavy and light chain variable regions operably linked to endogenous mouse constant region loci such that the mouse produces an antigen-binding protein, e.g., antibody, comprising a human variable region and a mouse constant region in response to antigenic stimulation.
  • the DNA encoding the variable regions of the heavy and light chains of the antibody are isolated and operably linked to DNA encoding the human heavy and light chain constant regions.
  • the DNA is then expressed in a cell capable of expressing the fully human antibody. 4.
  • Embodiments of the present disclosure encompass antibody fragments.
  • Various techniques have been developed for the production of antibody fragments. Traditionally, these fragments were derived via proteolytic digestion of intact antibodies (see, e.g., Morimoto et al., Journal of Biochemical and Biophysical Methods 24:107-7 (1992); and Brennan et al., Science, 229: 81 (1985)). However, these fragments can now be produced directly by Page 44 of 203 1097657169 ⁇ 1 ⁇ AMERICAS recombinant host cells. Fab, Fv and scFv antibody fragments can all be expressed in and secreted from E. coli, thus allowing the facile production of large amounts of these fragments.
  • Antibody fragments can be isolated from the antibody phage libraries discussed above.
  • Fab′- SH fragments can be directly recovered from E. coli and chemically coupled to form F(ab′)2 fragments (Carter et al., Bio/Technology 10:163-7 (1992)).
  • F(ab′)2 fragments can be isolated directly from recombinant host cell culture.
  • Fab and F(ab′)2 fragment with increased in vivo half-life comprising salvage receptor binding epitope residues are described in U.S. Pat. No. 5,869,046. Other techniques for the production of antibody fragments will be apparent to the skilled practitioner.
  • the antibody of choice is a single chain Fv fragment (scFv) (see WO 93/16185; U.S. Pat. No.5,571,894; and U.S. Pat. No.5,587,458).
  • Fv and sFv are the only species with intact combining sites that are devoid of constant regions; thus, they are suitable for reduced nonspecific binding during in vivo use.
  • sFv fusion proteins may be constructed to yield fusion of an effector protein at either the amino or the carboxy terminus of an sFv (see Antibody Engineering, ed. Borrebaeck, supra.
  • the antibody fragment may also be a “linear antibody”, e.g., as described in U.S. Pat.
  • an anti-B7H6 antibody derived scFv is used in a CAR of the present disclosure. Included among anti-B7H6 antibody fragments are portions of anti-B7H6 antibodies (and combinations of portions of anti-B7H6 antibodies, for example, scFv) that may be used as targeting arms, directed to a B7H6 epitope, in CARs and CAR modified immune cells of the present disclosure. Such fragments are not necessarily proteolytic fragments but rather portions of polypeptide sequences that can confer affinity for target. 5.
  • an anti-B7H6 antibody provided herein is a multispecific antibody, for example, a bispecific antibody.
  • Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes.
  • Exemplary bispecific antibodies may bind to two different epitopes of a B7H6 protein as described herein.
  • Other such antibodies may combine a B7H6 binding site with a binding site for another protein.
  • an anti-B7H6 arm may be combined with an arm which binds to a triggering molecule on a leukocyte such as a T cell receptor molecule (e.g., CD3), or Fc receptors for IgG (Fc ⁇ R), such as Fc ⁇ RI (CD64), Fc ⁇ RII (CD32) and Fc ⁇ RIII (CD16), so as to focus and localize cellular defense Page 45 of 203 1097657169 ⁇ 1 ⁇ AMERICAS mechanisms to the B7H6-expressing cell.
  • Bispecific antibodies may also be used to localize cytotoxic agents to cells which express B7H6.
  • bispecific antibodies possess a B7H6-binding arm and an arm which binds the cytotoxic agent (e.g., saporin, anti-interferon- ⁇ , vinca alkaloid, ricin A chain, methotrexate or radioactive isotope hapten).
  • cytotoxic agent e.g., saporin, anti-interferon- ⁇ , vinca alkaloid, ricin A chain, methotrexate or radioactive isotope hapten.
  • Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g., F(ab')2 bispecific antibodies). [00187] Methods for making bispecific antibodies are known in the art. Traditional production of full length bispecific antibodies is based on the co-expression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities (Millstein et al., Nature 305: 537-9 (1983)).
  • two immunoglobulin polypeptides each comprise an interface.
  • An interface of one immunoglobulin polypeptide interacts with a corresponding interface on the other immunoglobulin polypeptide, thereby allowing the two immunoglobulin polypeptides to associate.
  • These interfaces may be engineered such that a “knob” or “protuberance” (these terms may be used interchangeably herein) located in the interface of one immunoglobulin polypeptide corresponds with a “hole” or “cavity” (these terms may be used interchangeably herein) located in the interface of the other immunoglobulin polypeptide.
  • the hole is of identical or similar size to the knob and suitably positioned such that when the two interfaces interact, the knob of one interface is positionable in the corresponding hole of the other interface.
  • this is thought to stabilize the heteromultimer and favor formation of the heteromultimer over other species, for example homomultimers.
  • this approach may be used to promote the heteromultimerization of two different immunoglobulin polypeptides, creating a bispecific antibody comprising two immunoglobulin polypeptides with binding specificities for different epitopes.
  • antibody variable domains with the desired binding specificities are fused to immunoglobulin constant domain sequences.
  • the fusion preferably is with an immunoglobulin heavy chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is typical to have the first heavy- chain constant region (CH1) containing the site necessary for light chain binding, present in at least one of the fusions.
  • DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain are inserted into separate expression vectors, and are co- transfected into a suitable host organism.
  • the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm.
  • the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture.
  • One interface comprises at least a part of the C H 3 domain of an antibody constant domain.
  • Bispecific antibodies include cross-linked or “heteroconjugate” antibodies.
  • one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin.
  • Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Pat. No.4,676,980), and for treatment of HIV infection (WO 91/00360, WO 92/200373, and EP 03089).
  • Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in U.S. Pat. No. 4,676,980, along with a number of cross-linking techniques. [00193] Techniques for generating bispecific antibodies from antibody fragments have also been described in the literature. For example, bispecific antibodies can be prepared using chemical linkage.
  • bispecific antibodies produced can be used as agents for the selective immobilization of enzymes.
  • Shalaby et al., J. Exp. Med., 175: 217-225 (1992) describe the production of a fully humanized bispecific antibody F(ab′)2 molecule.
  • Each Fab′ fragment was separately secreted from E. coli and subjected to directed chemical coupling in vitro to form the bispecific antibody.
  • Various techniques for making and isolating bispecific antibody fragments directly from recombinant cell culture have also been described. For example, bispecific antibodies have been produced using leucine zippers. Kostelny et al., J. Immunol., 148(5):1547-1553 (1992).
  • the leucine zipper peptides from the Fos and Jun proteins were linked to the Fab′ portions of two different antibodies by gene fusion.
  • the antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers.
  • the “diabody” technology described by Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments.
  • the fragments comprise a heavy-chain variable domain (V H ) connected to a light-chain variable domain (V L ) by a linker which is too short to allow pairing between the two domains on the same chain.
  • V H and Page 48 of 203 1097657169 ⁇ 1 ⁇ AMERICAS V L domains of one fragment are forced to pair with the complementary V L and V H domains of another fragment, thereby forming two antigen-binding sites.
  • Another strategy for making bispecific antibody fragments by the use of single-chain Fv (sFv) dimers has also been reported. See Gruber et al, J. Immunol, 152:5368 (1994).
  • sFv single-chain Fv
  • Another technique for making bispecific antibody fragments is the “bispecific T cell engager” or BiTE® approach (see, e.g., WO2004/106381, WO2005/061547, WO2007/042261, and WO2008/119567).
  • a single polypeptide chain includes two single chain Fv (scFv) fragments, each having a variable heavy chain (VH) and a variable light chain (VL) domain separated by a polypeptide linker of a length sufficient to allow intramolecular association between the two domains.
  • This single polypeptide further includes a polypeptide spacer sequence between the two scFv fragments.
  • Each scFv recognizes a different epitope, and these epitopes may be specific for different cell types, such that cells of two different cell types are brought into close proximity or tethered when each scFv is engaged with its cognate epitope.
  • One particular embodiment of this approach includes a scFv recognizing a cell-surface antigen expressed by an immune cell, e.g., a CD3 polypeptide on a T cell, linked to another scFv that recognizes a cell- surface antigen expressed by a target cell, such as a malignant or tumor cell.
  • an immune cell e.g., a CD3 polypeptide on a T cell
  • another scFv that recognizes a cell- surface antigen expressed by a target cell, such as a malignant or tumor cell.
  • the bispecific T cell engager may be expressed using any prokaryotic or eukaryotic cell expression system known in the art, e.g., a CHO cell line.
  • a solution containing secreted polypeptides is first subjected to a metal affinity chromatography, and polypeptides are eluted with a gradient of imidazole concentrations.
  • This eluate is further purified using anion exchange chromatography, and polypeptides are eluted using with a gradient of sodium chloride concentrations.
  • this eluate is subjected to size exclusion chromatography to separate monomers from multimeric species.
  • DARTs dual- affinity re-targeting proteins
  • TandAbs Tandem diabodies
  • a DART is composed of two Fv fragments, with two unique antigen-binding sites formed when two Fv fragments Page 49 of 203 1097657169 ⁇ 1 ⁇ AMERICAS heterodimerize (Holliger et al., Proc. Natl. Acad. Sci. USA.90:6444–6448 (1993).
  • Fv1 consists of a VH from antibody “A” and a VL from antibody “B”, while Fv2 is made from a VH from antibody “B” and VL from antibody “A”.
  • TandAbs are tetravalent bispecific antibodies provide two binding sites for each antigen to maintain the avidity of a natural bivalent antibody. Moreover, TandAbs have a molecular weight (approximately 105 kDa) exceeding the first-pass renal clearance threshold, thus offering a longer half-life compared to smaller antibody constructs (Reusch et al., Clin. Cancer Res. Off. J. Am. Assoc.
  • Anti-B7H6 antibody variants can be prepared by introducing appropriate nucleotide changes into the encoding DNA, and/or by synthesis of the desired antibody or polypeptide. Those skilled in the art will appreciate that amino acid changes may alter post-translational processes of the anti-B7H6 antibody, such as changing the number or position of glycosylation sites or altering the membrane anchoring characteristics. [00199] Variations in the anti-B7H6 antibodies described herein, can be made, for example, using any of the techniques and guidelines for conservative and non-conservative mutations set forth, for instance, in U.S. Patent No. 5,364,934.
  • Variations may be a substitution, deletion or insertion of one or more codons encoding the antibody or polypeptide that results in a change in the amino acid sequence as compared with the native sequence antibody or polypeptide.
  • the variation is by substitution of at least one amino acid with any other amino acid in one or more of the domains of the anti-B7H6 antibody.
  • Guidance in determining which amino acid residue may be inserted, substituted or deleted without adversely affecting the desired activity may be found Page 50 of 203 1097657169 ⁇ 1 ⁇ AMERICAS by comparing the sequence of the anti-B7H6 antibody with that of homologous known protein molecules and minimizing the number of amino acid sequence changes made in regions of high homology.
  • Amino acid substitutions can be the result of replacing one amino acid with another amino acid having similar structural and/or chemical properties, such as the replacement of a leucine with a serine, i.e., conservative amino acid replacements. Insertions or deletions may optionally be in the range of about 1 to 5 amino acids. The variation allowed may be determined by systematically making insertions, deletions or substitutions of amino acids in the sequence and testing the resulting variants for activity exhibited by the full-length or mature native sequence.
  • Anti-B7H6 antibody fragments are provided herein. Such fragments may be truncated at the N-terminus or C-terminus, or may lack internal residues, for example, when compared with a full-length native antibody or protein.
  • Anti-B7H6 antibody fragments may be prepared by any of a number of conventional techniques. Desired peptide fragments may be chemically synthesized.
  • An alternative approach involves generating antibody or polypeptide fragments by enzymatic digestion, e.g., by treating the protein with an enzyme known to cleave proteins at sites defined by particular amino acid residues, or by digesting the DNA with suitable restriction enzymes and isolating the desired fragment.
  • Yet another suitable technique involves isolating and amplifying a DNA fragment encoding a desired antibody or polypeptide fragment, by polymerase chain reaction (PCR).
  • Oligonucleotides that define the desired termini of the DNA fragment are employed at the 5' and 3' primers in the PCR.
  • anti-B7H6 antibody fragments share at least one biological and/or immunological activity with the native anti-B7H6 antibody disclosed herein.
  • conservative substitutions of interest are shown in Table 1 under the heading of preferred substitutions. If such substitutions result in a change in biological activity, then more substantial changes, denominated exemplary substitutions in Table 1, or as further described below in reference to amino acid classes, are introduced and the products screened.
  • Naturally occurring residues are divided into groups based on common side-chain properties: (1) hydrophobic: norleucine, met, ala, val, leu, ile; Page 52 of 203 1097657169 ⁇ 1 ⁇ AMERICAS (2) neutral hydrophilic: cys, ser, thr; (3) acidic: asp, glu; (4) basic: asn, gln, his, lys, arg; (5) residues that influence chain orientation: gly, pro; and (6) aromatic: trp, tyr, phe. [00204] Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • Such substituted residues also may be introduced into the conservative substitution sites or, more preferably, into the remaining (non-conserved) sites.
  • the variations can be made using methods known in the art such as oligonucleotide- mediated (site-directed) mutagenesis, alanine scanning, and PCR mutagenesis.
  • Site-directed mutagenesis (Carter et al., Nucl. Acids Res., 13: 4331 (1986); Zoller et al., Nucl. Acids Res., 10: 6487 (1987)), cassette mutagenesis (Wells et al., Gene, 34: 315 (1985)), restriction selection mutagenesis (Wells et al., Philos. Trans. R.
  • Scanning amino acid analysis can also be employed to identify one or more amino acids along a contiguous sequence.
  • preferred scanning amino acids are relatively small, neutral amino acids.
  • Such amino acids include alanine, glycine, serine, and cysteine.
  • Alanine is typically a preferred scanning amino acid among this group because it eliminates the side-chain beyond the beta-carbon and is less likely to alter the main-chain conformation of the variant (Cunningham and Wells, Science, 244: 1081-5 (1989)). Alanine is also typically preferred because it is the most common amino acid.
  • cysteine residues not involved in maintaining the proper conformation of the anti- B7H6 antibody also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant crosslinking. Conversely, cysteine bond(s) may be added to the anti-B7H6 antibody to improve its stability (particularly where the antibody is an antibody fragment such as an Fv fragment).
  • a particularly preferred type of substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g., a humanized or human antibody).
  • a parent antibody e.g., a humanized or human antibody.
  • the resulting variant(s) selected for further development will have improved biological properties relative to the parent antibody from which they are generated.
  • a convenient way for generating such substitutional variants involves affinity maturation using phage display. Briefly, several hypervariable region sites (e.g., 6-7 sites) are mutated to generate all possible amino substitutions at each site.
  • the antibody variants thus generated are displayed in a monovalent fashion from filamentous phage particles as fusions to the gene III product of M13 packaged within each particle.
  • the phage-displayed variants are then screened for their biological activity (e.g., binding affinity) as herein disclosed.
  • alanine scanning mutagenesis can be performed to identify hypervariable region residues contributing significantly to antigen binding.
  • the panel of variants is subjected to screening as described herein and antibodies with superior properties in one or more relevant assays may be selected for further development.
  • Nucleic acid molecules encoding amino acid sequence variants of the anti-B7H6 antibody are prepared by a variety of methods known in the art. These methods include, but are not limited to, isolation from a natural source (in the case of naturally occurring amino acid sequence variants) or preparation by oligonucleotide-mediated (or site-directed) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared variant or a non-variant version of the anti-B7H6 antibody.
  • b) Modifications [00210] Covalent modifications of anti-B7H6 antibodies are included within the scope of this invention.
  • One type of covalent modification includes reacting targeted amino acid residues of an anti-B7H6 antibody with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C- terminal residues of the anti-B7H6 antibody.
  • Derivatization with bifunctional agents is useful, for instance, for crosslinking anti-B7H6 antibody to a water-insoluble support matrix or surface for use in a method for purifying anti-B7H6 antibodies, and vice-versa.
  • crosslinking agents include, e.g., 1,1-bis(diazoacetyl)-2-phenylethane, glutaraldehyde, N-hydroxysuccinimide esters, for example, esters with 4-azidosalicylic acid, homobifunctional imidoesters, including disuccinimidyl esters such as 3,3'- dithiobis(succinimidylpropionate), bifunctional maleimides such as bis-N-maleimido-1,8-octane and agents such as methyl-3-[(p-azidophenyl)dithio]propioimidate.
  • 1,1-bis(diazoacetyl)-2-phenylethane glutaraldehyde
  • N-hydroxysuccinimide esters for example, esters with 4-azidosalicylic acid
  • homobifunctional imidoesters including disuccinimidyl esters such as 3,3'- dithiobis(succin
  • Another type of covalent modification of the anti-B7H6 antibody included within the scope of this invention comprises altering the native glycosylation pattern of the antibody or polypeptide.
  • “Altering the native glycosylation pattern” is intended for purposes herein to mean deleting one or more carbohydrate moieties found in native sequence anti-B7H6 antibody (either by removing the underlying glycosylation site or by deleting the glycosylation by chemical and/or enzymatic means), and/or adding one or more glycosylation sites that are not present in the native sequence anti-B7H6 antibody.
  • the phrase includes qualitative changes in the glycosylation of the native proteins, involving a change in the nature and proportions of the various carbohydrate moieties present.
  • N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
  • the tripeptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain.
  • O-linked glycosylation refers to the attachment of one of the sugars N-aceylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5- hydroxylysine may also be used.
  • Page 55 of 203 1097657169 ⁇ 1 ⁇ AMERICAS [00214] Addition of glycosylation sites to the anti-B7H6 antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites).
  • the alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original anti-B7H6 antibody (for O-linked glycosylation sites).
  • the anti-B7H6 antibody amino acid sequence may optionally be altered through changes at the DNA level, particularly by mutating the DNA encoding the anti-B7H6 antibody at preselected bases such that codons are generated that will translate into the desired amino acids.
  • Another means of increasing the number of carbohydrate moieties on the anti-B7H6 antibody is by chemical or enzymatic coupling of glycosides to the polypeptide.
  • Enzymatic cleavage of carbohydrate moieties on polypeptides can be achieved by the use of a variety of endo- and exo- glycosidases as described by Thotakura et al., Meth. Enzymol., 138:350 (1987).
  • c) Fc Region Variants It may be desirable to modify the antibody of the invention with respect to effector function, e.g., so as to enhance antigen-dependent cell-mediated cyototoxicity (ADCC) and/or complement dependent cytotoxicity (CDC) of the antibody. This may be achieved by introducing one or more amino acid substitutions in an Fc region of the antibody.
  • cysteine residue(s) may be introduced in the Fc region, thereby allowing interchain disulfide bond formation in this region.
  • the homodimeric antibody thus generated may have improved internalization capability and/or increased complement-mediated cell killing and antibody- dependent cellular cytotoxicity (ADCC) (see Caron et al., J. Exp Med.176: 1191-5 (1992); Shopes, B. J. Immunol.148: 2918-22 (1992).
  • ADCC antibody- dependent cellular cytotoxicity
  • Homodimeric antibodies with enhanced anti-tumor activity may also be prepared using heterobifunctional cross-linkers as described in Wolff et al., Cancer Page 56 of 203 1097657169 ⁇ 1 ⁇ AMERICAS Research 53: 2560-5 (1993).
  • an antibody can be engineered which has dual Fc regions and may thereby have enhanced complement lysis and ADCC capabilities. See Stevenson et al., Anti-Cancer Drug Design 3: 219-30 (1989).
  • a salvage receptor binding epitope into the antibody (especially an antibody fragment) as described in U.S. Patent 5,739,277, for example.
  • the term “salvage receptor binding epitope” refers to an epitope of the Fc region of an IgG molecule (e.g., IgG1, IgG2, IgG3, or IgG4) that is responsible for increasing the in vivo serum half-life of the IgG molecule.
  • cysteine engineered antibodies e.g.,“thioMAbs,” in which one or more residues of an antibody are substituted with cysteine residues.
  • the substituted residues occur at accessible sites of the antibody.
  • reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker-drug moieties, to create an immunoconjugate, as described further herein.
  • Cysteine engineered antibodies can be generated as described, e.g, in U.S. Patent No. 7,521,541.
  • Immunoconjugates which include an antibody, disclosed herein, conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, proteins, peptides, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • cytotoxic agents such as chemotherapeutic agents or drugs, growth inhibitory agents, proteins, peptides, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • an antibody of the disclosed subject matter can be functionally linked (e.g., by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other binding molecules, such as another antibody, antibody fragment, peptide or binding mimetic.
  • an immunoconjugate is an antibody-drug conjugate (ADC) in which an antibody of the present disclosure is conjugated to one or more drugs, including but not limited to, a maytansinoid (see U.S. Patent Nos.5,208,020, 5,416,064 and European Patent EP 0 425235 Bl); an auristatin such as monomethyl auri statin drug moieties DE and DF (MMAE and Page 57 of 203 1097657169 ⁇ 1 ⁇ AMERICAS MMAF) (see U.S. Patent Nos. 5,635,483 and 5,780,588, and 7,498,298); a dolastatin; a calicheamicin or derivative thereof (see U.S.
  • ADC antibody-drug conjugate
  • an anthracycline such as daunomycin or doxorubicin
  • an immunoconjugate includes an antibody as described herein conjugated to an enzymatically active toxin or fragment thereof, including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
  • an enzymatically active toxin or fragment thereof including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (
  • an immunoconjugate includes an antibody, as described herein, conjugated to a radioactive atom to form a radioconjugate.
  • a variety of radioactive isotopes are available for the production of radioconjugates. Non-limiting examples include At 211 , Ac 225 , 1 131 , 1 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu.
  • a radioconjugate When a radioconjugate is used for detection, it can include a radioactive atom for scintigraphic studies, for example tc99m or I 123 , or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, mri), such as iodine-123, iodine-131, indium-111, fluorine-19, carbon-13, nitrogen-15, oxygen-17, gadolinium, manganese or iron.
  • NMR nuclear magnetic resonance
  • Conjugates of an antibody fragment and cytotoxic agent can be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP), succinimidyl-4-(N-maleimidomethyl) cyclohexane- 1-carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HC1), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis- diazonium derivatives (such as bis- Page 58 of 203 1097657169 ⁇ 1 ⁇ AMERICAS (p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as toluene 2,
  • a ricin immunotoxin can be prepared as described in Vitetta et al., Science 238: 1098 (1987).
  • Carbon- 14- labeled 1- isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody.
  • the linker can be a“cleavable linker” facilitating release of a cytotoxic drug in the cell.
  • an acid-labile linker, peptidase-sensitive linker, photolabile linker, dimethyl linker or disulfide-containing linker (Chari et al., Cancer Res. 52: 127-131 (1992); U. S.
  • Patent No. 5,208,020 can be used.
  • linkers are disclosed above.
  • the immunuoconjugates disclosed herein expressly contemplate, but are not limited to such conjugates prepared with cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MBS, sulfo- SIAB, sulfo- SMCC, and sulfo-SMPB, and SVSB (succinimidyl-(4-vinylsulfone)benzoate) which are commercially available (e.g ., from Pierce Biotechnology, Inc., Rockford, IL., U.S.A).
  • the presently disclosed subject matter also encompasses antibody fusions.
  • proteins can be linked together either through chemical or genetic manipulation using methods known in the art. See, for example, Gillies et al., Proc. Nat’l Acad. Sci. USA 89: 1428- 1432 (1992) and US Patent No.5,650,150.
  • the present disclosure encompasses an anti-B7H6 antibody-cytokine fusion protein.
  • an anti-B7H6 antibody as herein disclosed can be fused to any cytokine via the use of recombinant molecular biological techniques.
  • the anti- B7H6 antibody may be fused to IL-2 (Gillies, S., Protein Engineering, Design and Selection 26(10): 561-569 (2013); Klein, C. et al., OncoImmunology 6:3 (2017).
  • the present disclosure encompasses an anti-B7H6 antibody-T-cell engager fusion protein.
  • anti-B7H6 antibody-T-cell engager fusion proteins comprise fusions between an anti-B7H6 antibody and a ligand for a receptor expressed on a T cell. Examples of such ligands include but are not limited to CD40L, OX40L, 4-1BBL, CD80/86, ICOSL, and the like.
  • the ligand is fused to an Fc portion of an anti-B7H6 antibody. In embodiments, the ligand is fused to a C-terminus of a light chain of an anti-B7H6 Page 59 of 203 1097657169 ⁇ 1 ⁇ AMERICAS antibody.
  • a similar approach can be used for generation of other antibody-T-cell engager fusion proteins.
  • Anti-B7H6 antibodies or antigen-binding fragments of the present disclosure may be produced using recombinant methods and compositions, for example as described in US. Patent No.4,816,567.
  • the invention also provides transformed cells and progeny thereof into which a nucleic acid molecule encoding an antibody or antigen-binding fragment, has been introduced by means of recombinant DNA techniques in vitro, ex vivo or in vivo.
  • the transformed cells eukaryotic or prokaryotic, may be used to produce recombinant antibody or antibody fragment for purification, or for in situ or secretory expression for various purposes, such as diagnosis or therapy for tumor.
  • the transformed cells can be propagated and the introduced nucleic acid transcribed, or encoded protein expressed. It is understood that a progeny cell may not be identical to the parental cell, since there may be mutations that occur during replication.
  • Transformed cells include but are not limited to prokaryotic and eukaryotic cells such as bacteria, fungi, plant, insect, and animal (e.g., mammalian, including human) cells. The cells may be present in culture, in a cell, tissue or organ ex vivo or present in a subject.
  • the antibody or antibody fragment is displayed on yeast cell surface; in another embodiment, the antibody or antibody fragment is coated on nanoparticle surface; in another embodiment, the antibody or antibody fragment is displayed on mammalian cell surface, such as T cells, NK cells or other human or other mammalian cells; in another embodiment, the antibody or antibody fragment is produced as secretory protein by yeast, E.coli or mammalian cells.
  • cell transformation employs a vector.
  • vector refers to, e.g., a plasmid, virus, such as a viral vector, or other vehicle known in the art that can be manipulated by insertion or incorporation of a nucleic acid, for genetic manipulation (i.e., "cloning vectors"), or can be used to transcribe or translate the inserted polynucleic acid (i.e., "expression vectors").
  • cloning vectors can be used to transcribe or translate the inserted polynucleic acid
  • expression vectors are useful for introducing nucleic acids, including a nucleic acid that encodes an antibody or antibody antigen-binding fragment operably linked with an expression control element, and expressing the encoded protein in vitro (e.g., in solution or in solid phase), in cells or in vivo.
  • the expression vector(s) is(are) transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce an antibody or antigen-binding fragment of the invention.
  • the invention includes host cells containing polynucleic acid(s) encoding an antibody of the invention (e.g., whole antibody, a heavy or light chain thereof, or portion thereof, or a single chain antibody, or a fragment or variant thereof), operably linked to a heterologous promoter.
  • vectors encoding both the heavy and light chains are co- expressed in the host cell for expression of the entire immunoglobulin molecule.
  • host-expression vector systems may be utilized to express the antibody molecules of the invention.
  • Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleic acid coding sequences, express an antibody molecule of the invention in situ.
  • These include, but are not limited to, bacteriophage particles engineered to express antibody fragments or variants thereof (single chain antibodies), microorganisms such as bacteria (e.g., E. coli, B.
  • subtilis transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3, NSO cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from
  • bacterial cells such as Escherichia coli, and more preferably, eukaryotic cells, especially for the expression of whole recombinant antibody molecule, are used for the expression of a recombinant antibody molecule.
  • mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., Gene 45: 101 (1986); Cockett et al., Bio/Technology 8:2 (11990); B Page 61 of 203 1097657169 ⁇ 1 ⁇ AMERICAS ebbington et al., Bio/Techniques 10: 169 (1992); Keen and Hale, Cytotechnology 18:207 (1996)).
  • a vector used to transform a cell or a host-expression vector generally contains at least an origin of replication for propagation in the cell.
  • Control elements, including expression control elements as set forth herein, present within a vector, are included to facilitate transcription and translation.
  • expression control element is intended to include, at a minimum, one or more components whose presence can influence expression, and can include components other than or in addition to promoters or enhancers, for example, leader sequences and fusion partner sequences, internal ribosome binding sites (IRES) elements for the creation of multigene, or polycistronic, messages, splicing signal for introns, maintenance of the correct reading frame of the gene to permit in-frame translation of mRNA, polyadenylation signal to provide proper polyadenylation of the transcript of a gene of interest, stop codons, etc.
  • Vectors can include a selection marker. As is known in the art, "selection marker" means a gene that allows for the selection of cells containing the gene.
  • “Positive selection” refers to a process whereby only cells that contain the selection marker will survive upon exposure to the positive selection.
  • Drug resistance is one example of a positive selection marker; cells containing the marker will survive in culture medium containing the selection drug, and cells which do not contain the marker will die.
  • markers include drug resistance genes such as neo, which confers resistance to G418, hygr, which confers resistance to hygromycin, or puro which confers resistance to puromycin, among others.
  • Other positive selection marker genes include genes that allow identification or screening of cells containing the marker. These genes include genes for fluorescent proteins (GFP), the lacZ gene, the alkaline phosphatase gene, and surface markers such as CD8, among others.
  • Vectors can contain negative selection markers.
  • “Negative selection” refers to a process whereby cells containing a negative selection marker are killed upon exposure to an appropriate negative selection agent.
  • cells which contain the herpes simplex virus-thymidine kinase (HSV-tk) gene (Wigler et al., Cell 11:223 (1977)) are sensitive to the drug gancyclovir (GANC).
  • GANC drug gancyclovir
  • the gpt gene renders cells sensitive to 6-thioxanthine.
  • Mammalian expression systems further include vectors specifically designed for in vivo and ex vivo expression. Such systems include adeno-associated virus (AAV) vectors (U.S. Page 62 of 203 1097657169 ⁇ 1 ⁇ AMERICAS Pat.
  • AAV adeno-associated virus
  • AAV vectors have previously been shown to provide expression of Factor IX in humans and in mice at levels sufficient for therapeutic benefit (Kay et al., Nat. Genet.24:257 (2000); Nakai et al., Blood 91 :4600 (1998)).
  • Adenoviral vectors U.S. Pat. Nos. 5,700,470, 5,731,172 and 5,928,944
  • herpes simplex virus vectors U.S. Pat. No. 5,501,979
  • retroviral e.g., lentivirus vectors are useful for infecting dividing as well as non-dividing cells and foamy virues
  • papilloma virus vectors e.g., human and bovine papilloma virus
  • Vectors also include cytomegalovirus (CMV) based vectors (U.S. Pat. No. 5,561,063).
  • CMV cytomegalovirus
  • Vectors that efficiently deliver genes to cells of the intestinal tract have been developed and also may be used (see, e.g., U.S. Pat. Nos. 5,821,235, 5,786,340 and 6,110,456).
  • phagemid vectors for use in the invention include any available in the art suitable for the production of the antibodies/antibody templates/FR libraries of the present invention and include phagemid vectors pCB04, pITl, pIT2, CANTAB 6, pComb 3 HS.
  • Filamentous vectors and methods of phagemid construction are described in, for example, U.S. Pat. No. 6,054,312 and U.S. Pat. No. 6,803,230, each incorporated herein by reference.
  • Bacteriophage display systems involving non-filamentous bacteriophage vectors known as cytoplasmic bacteriophage or lytic phage can also be utilized as described in for example, U.S. Pat. No.5,766,905, incorporated herein by reference.
  • Suitable bacterial expression constructs for use with the present invention include, but are not limited to the pCAL, pUC, pET, pETBlueTM (Novagen), pBAD, pLEX, pTrcHis2, pSE280, pSE380, pSE420 (Invitrogen), pKK223-2 (Clontech), pTrc99A, pKK223-3, pRIT2T, pMC1871, pEZZ 18 (Pharmacia), pBluescript II SK (Stratagene), pALTER-Exl, pALTER- Ex2, pGEMEX (Promega), pFivE (MBI), pQE (Qiagen) commercially available expression constructs, and their derivatives, and others known in the art.
  • the construct may also include, a virus, a plasmid, a bacmid, a phagemid, a cosmid, or a bacteriophage.
  • Page 63 of 203 1097657169 ⁇ 1 ⁇ AMERICAS [00236]
  • the use of liposomes for introducing various compositions into cells, including nucleic acids, is known to those skilled in the art (see, e.g., U.S. Pat. Nos.4,844,904, 5,000,959, 4,863,740, and 4,975,282).
  • a carrier comprising a natural polymer, or a derivative or a hydrolysate of a natural polymer, described in WO 94/20078 and U.S. Pat.
  • nucleic acid sequences can be "operably linked", i.e., positioned, to ensure the functioning of an expression control sequence.
  • expression constructs are typically replicable in the cells either as episomes or as integral parts of the cell's chromosomal DNA, and may contain appropriate origins of replication for the respective prokaryotic strain employed for expression.
  • expression constructs contain selection markers, such as for example, tetracycline resistance, ampicillin resistance, kanamycin resistance or chlormaphenicol resistance, facilitating detection and/or selection of those bacterial cells transformed with the desired nucleic acid sequences (see, e.g., U.S. Pat. No.4,704,362). These markers, however, are not exclusionary, and numerous others may be employed, as known to those skilled in the art.
  • expression constructs contain both positive and negative selection markers.
  • reporter genes may be incorporated within expression constructs to facilitate identification of transcribed products. Accordingly, in one embodiment of the present invention, reporter genes utilized are selected from the group consisting of ⁇ -galactosidase, chloramphenicol acetyl transferase, luciferase and a fluorescent protein. [00239] Prokaryotic promoter sequences regulate expression of the encoded polynucleic acid sequences, and in some embodiments of the present invention, are operably linked to polynucleic acids encoding the polypeptides of this invention.
  • these promoters are either constitutive or inducible, and provide a means of high and low levels of expression of the polypeptides of this invention, and in some embodiments, for regulated expression of multiple polypeptides of the invention, which in some embodiments are expressed as a fusion protein.
  • Page 64 of 203 1097657169 ⁇ 1 ⁇ AMERICAS [00240] Many well-known bacterial promoters, including the T7 promoter system, the lactose promoter system, tryptophan (Trp) promoter system, Trc/Tac Promoter Systems, beta-lactamase promoter system, tetA Promoter systems, arabinose regulated promoter system, Phage T5 Promoter, or a promoter system from phage lambda, may be employed, and others, as well, and comprise embodiments of the present invention.
  • the promoters will typically control expression, optionally with an operator sequence and may include ribosome binding site sequences for example, for initiating and completing transcription and translation.
  • the vector may also contain expression control sequences, enhancers that may regulate the transcriptional activity of the promoter, appropriate restriction sites to facilitate cloning of inserts adjacent to the promoter and other necessary information processing sites, such as RNA splice sites, polyadenylation sites and transcription termination sequences as well as any other sequence which may facilitate the expression of the inserted nucleic acid.
  • C. Purification of Anti-B7H6 Antibody [00241] Forms of anti-B7H6 antibody may be recovered from culture medium or from host cell lysates. If membrane-bound, it can be released from the membrane using a suitable detergent solution (e.g., Triton-X 100) or by enzymatic cleavage.
  • Cells employed in expression of anti-B7H6 antibody can be disrupted by various physical or chemical means, such as freeze-thaw cycling, sonication, mechanical disruption, or cell lysing agents. [00242] It may be desired to purify anti-B7H6 antibody from recombinant cell proteins or polypeptides.
  • the following procedures are exemplary of suitable purification procedures: by fractionation on an ion-exchange column; ethanol precipitation; reverse phase HPLC; chromatography on silica or on a cation-exchange resin such as DEAE; chromatofocusing; SDS- PAGE; ammonium sulfate precipitation; gel filtration using, for example, Sephadex G-75; protein A Sepharose columns to remove contaminants such as IgG; and metal chelating columns to bind epitope-tagged forms of the anti-B7H6 antibody.
  • the particulate debris either host cells or lysed fragments, are removed, for example, by centrifugation or ultrafiltration.
  • Carter et al., Bio/Technology 10: 163-7 (1992) describe a procedure for isolating antibodies which are secreted to the periplasmic space of E. coli. Briefly, cell paste is thawed in the presence of sodium acetate (pH 3.5), EDTA, and phenylmethylsulfonylfluoride (PMSF) over about 30 min. Cell debris can be removed by centrifugation.
  • sodium acetate pH 3.5
  • EDTA EDTA
  • PMSF phenylmethylsulfonylfluoride
  • supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit.
  • a protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants.
  • the antibody composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography, with affinity chromatography being the preferred purification technique.
  • affinity chromatography being the preferred purification technique.
  • the suitability of protein A as an affinity ligand depends on the species and isotype of any immunoglobulin Fc domain that is present in the antibody.
  • Protein A can be used to purify antibodies that are based on human ⁇ 1, ⁇ 2 or ⁇ 4 heavy chains (Lindmark et al., J. Immunol. Meth. 62: 1-13 (1983)). Protein G is recommended for all mouse isotypes and for human ⁇ 3 (Guss et al., EMBO J.5: 15671575 (1986)).
  • the matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose.
  • the Bakerbond ABXTMresin J. T. Baker, Phillipsburg, NJ
  • Other techniques for protein purification such as fractionation on an ion-exchange column, ethanol precipitation, Reverse Phase HPLC, chromatography on silica, chromatography on heparin SEPHAROSETM chromatography on an anion or cation exchange resin (such as a polyaspartic acid column), chromatofocusing, SDS- PAGE, and ammonium sulfate precipitation are also available depending on the antibody to be recovered.
  • the mixture comprising the antibody of interest and contaminants may be subjected to low pH hydrophobic interaction chromatography Page 66 of 203 1097657169 ⁇ 1 ⁇ AMERICAS using an elution buffer at a pH between about 2.5-4.5, and generally at low salt concentrations (e.g., from about 0-0.25M salt).
  • the antibody of the present invention may be employed in any known assay method, such as ELISA, competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays (Zola, (1987) Monoclonal Antibodies: A Manual of Techniques, pp.147-158, CRC Press, Inc.).
  • a detection label may be useful for localizing, visualizing, and quantitating a binding or recognition event.
  • the labelled antibodies of the invention can detect cell-surface receptors or antigens.
  • Another use for detectably labelled antibodies is a method of bead-based immunocapture comprising conjugating a bead with a fluorescent labelled antibody and detecting a fluorescence signal upon binding of a ligand. Similar binding detection methodologies utilize the surface plasmon resonance (SPR) effect to measure and detect antibody-antigen interactions.
  • SPR surface plasmon resonance
  • Detection labels such as fluorescent dyes and chemiluminescent dyes (Briggs et al (1997) J. Chem. Soc., Perkin-Trans.
  • the labels preferably (iii) have good water-solubility to achieve effective conjugate concentration and detection sensitivity and (iv) are non-toxic to living cells so as not to disrupt the normal metabolic processes of the cells or cause premature cell death.
  • Direct quantification of cellular fluorescence intensity and enumeration of fluorescently labelled events may be conducted on a system (FMAT® 8100 HTS System, Applied Biosystems, Foster City, Calif.) that automates mix-and-read, non-radioactive assays with live cells or beads (Miraglia, “Homogeneous cell- and bead-based assays for high throughput screening using fluorometric microvolume assay technology”, (1999) J. of Biomolecular Screening 4:193-204).
  • FMAT® 8100 HTS System Applied Biosystems, Foster City, Calif.
  • Fluorometric microvolume assay technology can be used to identify the up or down regulation by a molecule that is targeted to the cell surface (Swartzman, “A homogeneous and multiplexed immunoassay for high-throughput screening using fluorometric microvolume assay technology”, (1999) Anal. Biochem.271:143-51).
  • Labelled antibodies of the invention are useful as imaging biomarkers and probes by the various methods and techniques of biomedical and molecular imaging such as: (i) MRI (magnetic resonance imaging); (ii) MicroCT (computerized tomography); (iii) SPECT (single photon emission computed tomography); (iv) PET (positron emission tomography) Chen et al Bioconjugate Chem.15: 41-9 (2004); (v) bioluminescence; (vi) fluorescence; and (vii) ultrasound.
  • Immunoscintigraphy is an imaging procedure in which antibodies labeled with radioactive substances are administered to an animal or human patient and a picture is taken of sites in the body where the antibody localizes (US 6528624).
  • Imaging biomarkers may be objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacological responses to a therapeutic intervention.
  • Peptide labelling methods are well known (e.g., Haugland, 2003, Molecular Probes Handbook of Fluorescent Probes and Research Chemicals, Molecular Probes, Inc.; Brinkley, 1992, Bioconjugate Chem. 3:2; Garman, (1997) Non-Radioactive Labelling: A Practical Approach, Academic Press, London; Means (1990) Bioconjugate Chem.1:2; Glazer et al. (1975) Chemical Modification of Proteins. Laboratory Techniques in Biochemistry and Molecular Biology (T. S. Work and E.
  • the fluorescent reporter may be partially or significantly quenched by the quencher moiety in an intact peptide. Upon cleavage of the peptide by a peptidase or protease, a detectable increase in fluorescence may be measured (Knight, C. (1995) “Fluorimetric Assays of Proteolytic Enzymes”, Methods in Enzymology, Academic Press, 248:18- 34).
  • the labelled antibodies of the invention may also be used as an affinity purification agent. In this process, the labelled antibody is immobilized on a solid phase such a Sephadex resin or filter paper, using methods well known in the art.
  • assays are provided for identifying anti-B7H6 antibodies thereof having biological activity.
  • Biological activity may include, e.g., the ability to inhibit cell growth or proliferation (e.g., “cell killing” activity), or the ability to induce cell death, including programmed cell death (apoptosis).
  • an anti-B7H6 antibody is tested for its ability to inhibit cell growth or proliferation in vitro.
  • Assays for inhibition of cell growth or proliferation are well known in the art.
  • Certain assays for cell proliferation for example “cell killing” assays, measure cell viability.
  • One such assay is the CellTiter-GloTM Luminescent Cell Viability Assay, which is Page 69 of 203 1097657169 ⁇ 1 ⁇ AMERICAS commercially available from Promega (Madison, WI). That assay determines the number of viable cells in culture based on quantitation of ATP present, which is an indication of metabolically active cells.
  • the assay may be conducted in 96- or 384-well format, making it amenable to automated high-throughput screening (HTS) (see Cree et al (1995) AntiCancer Drugs 6: 398-404).
  • HTS high-throughput screening
  • the assay procedure involves adding a single reagent (CellTiter-Glo® Reagent) directly to cultured cells. This results in cell lysis and generation of a luminescent signal produced by a luciferase reaction. The luminescent signal is proportional to the amount of ATP present, which is directly proportional to the number of viable cells present in culture.
  • MTT colorimetric assay that measures the oxidation of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide to formazan by mitochondrial reductase.
  • MTT colorimetric assay that measures the oxidation of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide to formazan by mitochondrial reductase.
  • MTT colorimetric assay that measures the oxidation of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide to formazan by mitochondrial reductase.
  • this assay indicates the number of metabolically active cells present in a cell culture (see, e.g., Mosmann (1983) J. Immunol. Meth.65:55-63, and Zhang et al. (2005) Cancer Res.65: 3877-82).
  • an anti-B7H6 antibody is tested for its ability to induce cell death in vitro.
  • Assays for induction of cell death are well known in the art.
  • such assays measure, e.g., loss of membrane integrity as indicated by uptake of propidium iodide (PI), trypan blue (see Moore et al. Cytotechnology, 17: 1-11 (1995)), or 7AAD.
  • PI propidium iodide
  • trypan blue see Moore et al. Cytotechnology, 17: 1-11 (1995)
  • 7AAD propidium iodide
  • cells are cultured in Dulbecco’s Modified Eagle Medium (D-MEM):Ham’s F-12 (50:50) supplemented with 10% heat-inactivated FBS (Hyclone) and 2 mM L-glutamine.
  • the assay is performed in the absence of complement and immune effector cells.
  • Cells are seeded at a density of 3 x 10 6 per dish in 100 x 20 mm dishes and allowed to attach overnight.
  • the medium is removed and replaced with fresh medium alone or medium containing various concentrations of the antibody.
  • the cells are incubated for a 3-day time period. Following treatment, monolayers are washed with PBS and detached by trypsinization.
  • Cells are then centrifuged at 1200 rpm for 5 minutes at 4 oC, the pellet resuspended in 3 mL cold Ca2+ binding buffer (10 mM Hepes, pH 7.4, 140 mM NaCl, 2.5 mM CaCl2) and aliquoted into 35 mm strainer-capped 12 x 75 mm tubes (1 mL per tube, 3 tubes per treatment group) for removal of cell clumps. Tubes then receive PI (10 ⁇ g/mL). Samples are analyzed using a FACSCANTM flow cytometer and FACSCONVERTTM CellQuest software Page 70 of 203 1097657169 ⁇ 1 ⁇ AMERICAS (Becton Dickinson).
  • an anti-B7H6 antibody is tested for its ability to induce apoptosis (programmed cell death) in vitro.
  • An exemplary assay for antibodies that induce apoptosis is an annexin binding assay.
  • an exemplary annexin binding assay cells are cultured and seeded in dishes as discussed in the preceding paragraph. The medium is removed and replaced with fresh medium alone or medium containing 0.001 to 10 ⁇ g/mL of the antibody. Following a three-day incubation period, monolayers are washed with PBS and detached by trypsinization.
  • annexin V-FITC labeled annexin
  • Samples are analyzed using a FACSCANTM flow cytometer and FACSCONVERTTM CellQuest software (BD Biosciences). Antibodies that induce statistically significant levels of annexin binding relative to control are thus identified.
  • Another exemplary assay for antibodies that induce apoptosis is a histone DNA ELISA colorimetric assay for detecting internucleosomal degradation of genomic DNA.
  • Cells for use in any of the above in vitro assays include cells or cell lines that naturally express B7H6 or that have been engineered to express B7H6. Such cells include tumor cells that overexpress B7H6 relative to normal cells of the same tissue origin. Such cells also include cell lines (including tumor cell lines) that express B7H6 and cell lines that do not normally express B7H6 but have been transfected with nucleic acid encoding B7H6. [00260] In one aspect, an anti-B7H6 antibody thereof is tested for its ability to inhibit cell growth or proliferation in vivo.
  • an anti-B7H6 antibody thereof is tested for its ability to inhibit tumor growth in vivo.
  • In vivo model systems such as xenograft models, can be used for such testing.
  • human tumor cells are introduced into a suitably immunocompromised non-human animal, e.g., a SCID mouse.
  • An antibody of the invention is administered to the animal.
  • the ability of the antibody to inhibit or decrease tumor growth is measured.
  • the human tumor cells are tumor cells from a human patient.
  • the human tumor cells are Page 71 of 203 1097657169 ⁇ 1 ⁇ AMERICAS introduced into a suitably immunocompromised non-human animal by subcutaneous injection or by transplantation into a suitable site, such as a mammary fat pad.
  • a suitable site such as a mammary fat pad.
  • binding assays and other assays [00261]
  • an anti-B7H6 antibody is tested for its antigen binding activity.
  • an anti-B7H6 antibody is tested for its ability to bind to B7H6 expressed on the surface of a cell.
  • a FACS assay may be used for such testing.
  • competition assays may be used to identify a monoclonal antibody that competes with a monoclonal antibody comprising a HCVR/LCVR sequence pair of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, 15/16, 17/18, 19/20, 21/22, 23/24, 25/26, 27/28, 29/30, 31/32, 33/34, 35/36, 37/38, or 39/40; or that competes with a monoclonal antibody comprising the six CDRs of a HCVR/LCVR sequence pair selected from SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, 15/16, 17/18, 19/20, 21/22, 23/24, 25/26, 27/28, 29/30, 31/32, 33/34, 35/36, 37/38, or 39/40.
  • such a competing antibody binds to the same epitope (e.g., a linear or a conformational epitope) that is bound by a monoclonal antibody comprising a HCVR/LCVR sequence pair of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, 15/16, 17/18, 19/20, 21/22, 23/24, 25/26, 27/28, 29/30, 31/32, 33/34, 35/36, 37/38, or 39/40; or a monoclonal antibody comprising the six CDRs of a HCVR/LCVR sequence pair selected from SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, 15/16, 17/18, 19/20, 21/22, 23/24, 25/26, 27/28, 29/30, 31/32, 33/34, 35/36, 37/38, or 39/40.
  • a monoclonal antibody comprising the six CDRs of
  • Exemplary competition assays include, but are not limited to, routine assays such as those provided in Harlow and Lane (1988) Antibodies: A Laboratory Manual ch.14 (Cold Spring Harbor Laboratory, Cold Spring Harbor, NY). Detailed exemplary methods for mapping an epitope to which an antibody binds are provided in Morris (1996) “Epitope Mapping Protocols,” in Methods in Molecular Biology vol.66 (Humana Press, Totowa, NJ). Two antibodies are said to bind to the same epitope if each blocks binding of the other by 50% or more.
  • immobilized B7H6 is incubated in a solution comprising a first labeled antibody that binds to B7H6 and a second unlabeled antibody that is being tested for its ability to compete with the first antibody for binding to B7H6.
  • the second antibody may be present in a hybridoma supernatant.
  • immobilized B7H6 is incubated in a solution comprising the first labeled antibody but not the second unlabeled antibody. After Page 72 of 203 1097657169 ⁇ 1 ⁇ AMERICAS incubation under conditions permissive for binding of the first antibody to B7H6, excess unbound antibody is removed, and the amount of label associated with immobilized B7H6 is measured.
  • immobilized B7H6 is present on the surface of a cell or in a membrane preparation obtained from a cell expressing B7H6 on its surface.
  • purified anti-B7H6 antibodies can be further characterized by a series of assays including, but not limited to, N-terminal sequencing, amino acid analysis, non-denaturing size exclusion high pressure liquid chromatography (HPLC), mass spectrometry, ion exchange chromatography and papain digestion.
  • an epitope can include at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acids in a unique spatial conformation.
  • Epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids can be typically retained on exposure to denaturing solvents whereas epitopes formed by tertiary folding can be typically lost on treatment with denaturing solvents.
  • Epitope mapping can be performed to identify the linear or non-linear, discontinuous amino acid sequence(s), i.e. the epitope, that is (e.g., specifically) recognized by an anti-B7H6 antibody or antigen-binding fragment thereof.
  • a general approach for epitope mapping can require the expression of the full-length polypeptide sequence that is recognized by an antibody or ligand of interest, as well as various fragments, i.e., truncated forms of the polypeptide sequence, generally in a heterologous expression system.
  • a library of epitopes can be created by synthetically designing various portions of B7H6 as constructs and expressing them in a suitable system.
  • portions of B7H6 can be amplified out of Total RNA extracted from B7H6-expressing-cells isolated from human normal and/or malignant tissue.
  • the host system can be any suitable expression system such as 293 cells, insect cells, or a suitable in-vitro translation system.
  • the binding of an anti-B7H6 antibody or antigen-binding fragment thereof to one of the epitopes in the above-described library can be detected by contacting a labeled B7H6 antibody of the present disclosure with an epitope of the library and detecting a signal from the label.
  • For epitope mapping computational algorithms have also been developed which have been shown to map conformational discontinuous epitopes.
  • Conformational epitopes can be identified by determining spatial conformation of amino acids with methods that include, e.g., x- ray crystallography and 2-dimensional nuclear magnetic resonance.
  • Some epitope mapping methods such as, x-ray analyses of crystals of antigen: antibody complexes can provide atomic resolution of the epitope.
  • computational combinatorial methods for epitope mapping can be employed to model a potential epitope based on the sequence of the anti-B7H6 or antigen-binding fragment thereof. In such cases, the antigen binding portion of the antibody is sequenced, and computation models are used to reconstruct and predict a potential binding site of the antibody.
  • the disclosure provides a method of determining a B7H6 epitope, comprising: (a) preparing a library of epitopes from B7H6; (b) contacting the library of epitopes with an anti-B7H6 antibody; and (b) identifying the amino acid sequence of at least one epitope in the library of epitopes that is bound by the antibody.
  • the antibody is attached to a solid support.
  • the library of epitopes can comprise sequences that correspond to continuous and discontinuous epitopes of B7H6.
  • the library of epitopes comprises fragments from B7H6 ranging from about 10 amino acids to about 30 amino acids in length, from about 10 amino acids to about 20 amino acids in length, or from about 5 amino acids to about 12 amino acids in length.
  • the anti-B7H6 antibody or antigen-binding fragment thereof is labeled and the label is a radioactive molecule, a luminescent molecule, a fluorescent molecule, an enzyme, or biotin.
  • Phage panning may be used to identify B7H6-binding molecules that bind to B7H6 or one or more epitopes on B7H6.
  • phage panning using biotinylated recombinant human B7H6 protein bound to streptavidin beads is performed on highly diverse synthetic scFv phage display libraries using multiple (e.g., 5) rounds of selection, each with decreasing antigen concentration (e.g., starting at 100 pmol to 2 pmol) but increasing wash stringency.
  • an alternate panning strategy with alternating rounds of selection with 1e8 B7H6+ cells or antigen e.g., 100 pmol and 25 pmol, respectively
  • Antigen-bound phage may then be pulled down, eluted, amplified and screened using ELISAs for binder confirmation and selection.
  • the selected scFvs may be reformatted into IgGs, expressed, purified and characterized.
  • F. Chimeric Antigen Receptor (CAR) Constructs [00275] Aspects of the invention include nucleic acids encoding CARs, and constructs and vectors containing such nucleic acids.
  • the nucleic acid is a, e.g., heterologous, component of an expression cassette.
  • the nucleic acid is a, e.g., heterologous, component of a retroviral vector.
  • the nucleic acid is a, e.g., heterologous, component of an ⁇ ⁇ or ⁇ T cell, and preferably a ⁇ T cell. In embodiments, the nucleic acid is a, e.g., heterologous, component of an ⁇ + T cell and/or a ⁇ + T cell. In embodiments, the nucleic acid is a, e.g., heterologous, component of an ⁇ - T cell and/or a ⁇ - T cell.
  • a subject CAR of the invention comprises an antigen binding domain capable of specifically binding to B7H6.
  • the antigen binding domain may be operably linked to another domain of the CAR, for example a transmembrane domain, a costimulatory domain and/or an intracellular signaling domain, as described herein.
  • the antigen binding domains described herein can be combined with any of the transmembrane, costimulatory, and/or intracellular signaling domain(s) described herein, and/or any of the other domains described herein that may be included in a CAR of the present invention.
  • a subject CAR of the present invention may also include a hinge domain as described herein.
  • a subject CAR of the present invention may also include at least one spacer domain as described herein. 1.
  • the antigen binding domain can include any domain that binds to B7H6 and may include, but is not limited to, a monoclonal antibody, a polyclonal antibody, a synthetic antibody, a human antibody, a humanized antibody, a non-human antibody, a bispecific antibody, and any fragment thereof.
  • the antigen binding domain portion comprises a mammalian antibody or a fragment thereof. The choice of antigen binding domain may depend upon the type and number of antigens that are present on the surface of a target cell.
  • the antigen binding domain is selected from the group consisting of an antibody, an antigen binding fragment (Fab), and a single-chain variable fragment (scFv).
  • the present disclosure provides antibodies and CARs with “substantial identity” or “substantial similarity” to the sequences provided herein in the CDR or framework regions.
  • a nucleic acid molecule having substantial identity to a reference nucleic acid molecule may, in certain instances, encode a polypeptide having the same Page 76 of 203 1097657169 ⁇ 1 ⁇ AMERICAS or substantially similar amino acid sequence as the polypeptide encoded by the reference nucleic acid molecule.
  • the term "substantial similarity" or “substantially similar” means that two peptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, or 100% sequence identity.
  • residue positions, which are not identical differ by conservative amino acid substitutions.
  • a “conservative amino acid substitution” is one in which an amino acid residue is substituted by another amino acid residue having a side chain (R group) with similar chemical properties (e.g., charge or hydrophobicity).
  • R group side chain
  • a conservative amino acid substitution will not substantially change the functional properties of a protein.
  • the percent or degree of similarity may be adjusted upwards to correct for the conservative nature of the substitution. Means for making this adjustment are well known to those of skill in the art. See, e.g., Pearson (1994) Methods Mol. Biol.24: 307-331, which is herein incorporated by reference.
  • Examples of groups of amino acids that have side chains with similar chemical properties include 1) aliphatic side chains: glycine, alanine, valine, leucine and isoleucine; 2) aliphatic-hydroxyl side chains: serine and threonine; 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; 5) basic side chains: lysine, arginine, and histidine; 6) acidic side chains: aspartate and glutamate, and 7) sulfur-containing side chains: cysteine and methionine.
  • Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine- tyrosine, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-glutamine.
  • a conservative replacement is any change having a positive value in the PAM250 log-likelihood matrix disclosed in Gonnet et al. (1992) Science 256: 144345, herein incorporated by reference.
  • a "moderately conservative" replacement is any change having a nonnegative value in the PAM250 log-likelihood matrix.
  • Protein analysis software matches similar sequences using measures of similarity assigned to various substitutions, deletions and other modifications, including Page 77 of 203 1097657169 ⁇ 1 ⁇ AMERICAS conservative amino acid substitutions.
  • GCG software contains programs such as GAP and BESTFIT which can be used with default parameters to determine sequence homology or sequence identity between closely related polypeptides, such as homologous polypeptides from different species of organisms or between a wild type protein and a mutein thereof. See, e.g., GCG Version 6.1. Polypeptide sequences also can be compared using FASTA with default or recommended parameters; a program in GCG Version 6.1.
  • FASTA (e.g., FASTA2 and FASTA3) provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (Pearson (2000) supra). Sequences also can be compared using the Smith-Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 2, BLOSUM matrix of 62.
  • Another preferred algorithm when comparing a sequence disclosed herein to a database containing a large number of sequences from different organisms is the computer program BLAST, especially BLASTP or TBLASTN, using default parameters. See, e.g., Altschul et al. (1990) J. Mol.
  • anti-B7H6 CARs comprising variants of any of the HCVR, LCVR, and/or CDR amino acid sequences disclosed herein having one or more substitutions (e.g., conservative substitutions).
  • the present disclosure includes anti-B7H6 CARs having HCVR, LCVR, and/or CDR amino acid sequences with, e.g., 20 or fewer, 19 or fewer, 18 or fewer, 17 or fewer, 16 or fewer, 15 or fewer, 14 or fewer, 13 or fewer, 12 or fewer, 11 or fewer, 10 or fewer, 9 or fewer, 8 or fewer, 7 or fewer, 6 or fewer, 5 or fewer, 4 or fewer, 3 or fewer, 2 or fewer, or 1 amino acid substitutions relative to any of the HCVR, LCVR, and/or CDR (e.g., HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, or LCDR3) amino acid sequences disclosed herein.
  • CDR e.g., HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, or LCDR3 amino acid sequences disclosed herein.
  • an anti-B7H6 CAR can comprise 20, 19, 18, 17, 16, 15, 1413, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid substitutions (e.g., conservative amino acid substitutions) relative to any of the HCVR, LCVR, and/or CDR (e.g., HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, or LCDR3) amino acid sequences disclosed herein.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising an amino acid sequence selected from the group consisting of any one of SEQ ID NOs: 1-40.
  • the anti-B7H6 binding domain binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence selected from the group Page 78 of 203 1097657169 ⁇ 1 ⁇ AMERICAS consisting of any one of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, and 39.
  • the anti-B7H6 binding domain binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a LCVR amino acid sequence selected from the group consisting of any one of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, and 40.
  • the anti-B7H6 binding domain binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence selected from the group consisting of any one of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, and 15.
  • the anti-B7H6 binding domain binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a LCVR amino acid sequence selected from the group consisting of any one of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, and 16.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 amino acid sequence selected from the group consisting of SEQ ID NOs: 41- 60; a CDR2 amino acid sequence selected from the group consisting of SEQ ID NOs: 61-80; and a CDR3 amino acid sequence selected from the group consisting of SEQ ID NOs: 81-100.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a LCVR comprising a CDR1 amino acid sequence selected from the group consisting of SEQ ID NOs: 101-120; a CDR2 amino acid sequence selected from the group consisting of SEQ ID NOs: 121-140; and a CDR3 amino acid sequence selected from the group consisting of SEQ ID NOs: 141-160.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 amino acid sequence selected from the group consisting of SEQ ID NOs: 41- 48; a CDR2 amino acid sequence selected from the group consisting of SEQ ID NOs: 61-68; and a CDR3 amino acid sequence selected from the group consisting of SEQ ID NOs: 81-88.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a LCVR comprising a CDR1 amino acid sequence selected from the group consisting of SEQ ID NOs: 101-108; a CDR2 amino acid sequence selected from the group consisting of SEQ ID NOs: 121-128; and a CDR3 amino acid sequence selected from the group consisting of SEQ ID NOs: 141-148.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 1, and a LCVR amino acid sequence set forth at SEQ ID NO: 2.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 3, and a LCVR amino acid sequence set forth at SEQ ID NO: 4.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 5, and a LCVR amino acid sequence set forth at SEQ ID NO: 6.
  • an isolated nucleic acid encodes an anti- B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 7, and a LCVR amino acid sequence set forth at SEQ ID NO: 8.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 9, and a LCVR amino acid sequence set forth at SEQ ID NO: 10.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti- B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 11, and a LCVR amino acid sequence set forth at SEQ ID NO: 12.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 13, and a LCVR amino acid sequence set forth at SEQ ID NO: 14.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 15, and a LCVR amino acid sequence set forth at SEQ ID NO: 16.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 17, and a LCVR amino acid sequence set forth at SEQ ID NO: 18.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 19, and a LCVR amino acid sequence set forth at SEQ ID NO: 20.
  • an isolated nucleic Page 80 of 203 1097657169 ⁇ 1 ⁇ AMERICAS acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 21, and a LCVR amino acid sequence set forth at SEQ ID NO: 22.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 23, and a LCVR amino acid sequence set forth at SEQ ID NO: 24.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 25, and a LCVR amino acid sequence set forth at SEQ ID NO: 26.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 27, and a LCVR amino acid sequence set forth at SEQ ID NO: 28.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 29, and a LCVR amino acid sequence set forth at SEQ ID NO: 30.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 31, and a LCVR amino acid sequence set forth at SEQ ID NO: 32.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 33, and a LCVR amino acid sequence set forth at SEQ ID NO: 34.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 35, and a LCVR amino acid sequence set forth at SEQ ID NO: 36.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 37, and a LCVR amino acid sequence set forth at SEQ ID NO: 38.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 39, and a LCVR amino acid sequence set forth at SEQ ID NO: 40.
  • Preferred embodiments include those in which an isolated nucleic acid encodes an anti- B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 1, and a LCVR amino acid sequence set forth at SEQ ID NO: 2; a HCVR amino acid sequence set forth as SEQ ID NO: 3, and a LCVR amino acid sequence set forth at SEQ ID NO: 4; a HCVR amino acid sequence set forth as SEQ ID NO: 5, and a LCVR amino acid sequence set forth at SEQ ID NO: 6; a HCVR amino acid sequence set forth as SEQ ID NO: 7, and a LCVR amino acid sequence set forth at SEQ ID NO: 8; a HCVR amino acid sequence set forth as SEQ ID NO: 9, and a LCVR amino acid sequence set forth at SEQ ID NO: 10;
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 41, a CDR2 sequence set forth as SEQ ID NO: 61, and a CDR3 sequence set forth as SEQ ID NO: 81; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 101, a CDR2 sequence set forth as SEQ ID NO: 121, and a CDR3 sequence set forth as SEQ ID NO: 141.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 42, a CDR2 sequence set forth as SEQ ID NO: 62, and a CDR3 sequence set forth as SEQ ID NO: 82; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 102, a CDR2 sequence set forth as SEQ ID NO: 122, and a CDR3 sequence set forth as SEQ ID NO: 142.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 43, a CDR2 sequence set forth as SEQ ID NO: 63, and a CDR3 sequence set forth as SEQ ID NO: 83; and/or comprising a LCVR comprising Page 82 of 203 1097657169 ⁇ 1 ⁇ AMERICAS a CDR1 sequence set forth as SEQ ID NO: 103, a CDR2 sequence set forth as SEQ ID NO: 123, and a CDR3 sequence set forth as SEQ ID NO: 143.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 44, a CDR2 sequence set forth as SEQ ID NO: 64, and a CDR3 sequence set forth as SEQ ID NO: 84; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 104, a CDR2 sequence set forth as SEQ ID NO: 124, and a CDR3 sequence set forth as SEQ ID NO: 144.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 45, a CDR2 sequence set forth as SEQ ID NO: 65, and a CDR3 sequence set forth as SEQ ID NO: 85; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 105, a CDR2 sequence set forth as SEQ ID NO: 125, and a CDR3 sequence set forth as SEQ ID NO: 145.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 46, a CDR2 sequence set forth as SEQ ID NO: 66, and a CDR3 sequence set forth as SEQ ID NO: 86; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 106, a CDR2 sequence set forth as SEQ ID NO: 126, and a CDR3 sequence set forth as SEQ ID NO: 146.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 47, a CDR2 sequence set forth as SEQ ID NO: 67, and a CDR3 sequence set forth as SEQ ID NO: 87; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 107, a CDR2 sequence set forth as SEQ ID NO: 127, and a CDR3 sequence set forth as SEQ ID NO: 147.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 48, a CDR2 sequence set forth as SEQ ID NO: 68, and a CDR3 sequence set forth as SEQ ID NO: 88; and/or comprising a LCVR comprising Page 83 of 203 1097657169 ⁇ 1 ⁇ AMERICAS a CDR1 sequence set forth as SEQ ID NO: 108, a CDR2 sequence set forth as SEQ ID NO: 128, and a CDR3 sequence set forth as SEQ ID NO: 148.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 49, a CDR2 sequence set forth as SEQ ID NO: 69, and a CDR3 sequence set forth as SEQ ID NO: 89; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 109, a CDR2 sequence set forth as SEQ ID NO: 129, and a CDR3 sequence set forth as SEQ ID NO: 149.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 50, a CDR2 sequence set forth as SEQ ID NO: 70, and a CDR3 sequence set forth as SEQ ID NO: 90; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 110, a CDR2 sequence set forth as SEQ ID NO: 130, and a CDR3 sequence set forth as SEQ ID NO: 150.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 51, a CDR2 sequence set forth as SEQ ID NO: 71, and a CDR3 sequence set forth as SEQ ID NO: 91; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 111, a CDR2 sequence set forth as SEQ ID NO: 131, and a CDR3 sequence set forth as SEQ ID NO: 151.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 52, a CDR2 sequence set forth as SEQ ID NO: 72, and a CDR3 sequence set forth as SEQ ID NO: 92; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 112, a CDR2 sequence set forth as SEQ ID NO: 132, and a CDR3 sequence set forth as SEQ ID NO: 152.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 53, a CDR2 sequence set forth as SEQ ID NO: 73, and a CDR3 sequence set forth as SEQ ID NO: 93; and/or comprising a LCVR comprising Page 84 of 203 1097657169 ⁇ 1 ⁇ AMERICAS a CDR1 sequence set forth as SEQ ID NO: 113, a CDR2 sequence set forth as SEQ ID NO: 133, and a CDR3 sequence set forth as SEQ ID NO: 153.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 54, a CDR2 sequence set forth as SEQ ID NO: 74, and a CDR3 sequence set forth as SEQ ID NO: 94; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 114, a CDR2 sequence set forth as SEQ ID NO: 134, and a CDR3 sequence set forth as SEQ ID NO: 154.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 55, a CDR2 sequence set forth as SEQ ID NO: 75, and a CDR3 sequence set forth as SEQ ID NO: 95; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 115, a CDR2 sequence set forth as SEQ ID NO: 135, and a CDR3 sequence set forth as SEQ ID NO: 155.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 56, a CDR2 sequence set forth as SEQ ID NO: 76, and a CDR3 sequence set forth as SEQ ID NO: 96; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 116, a CDR2 sequence set forth as SEQ ID NO: 136, and a CDR3 sequence set forth as SEQ ID NO: 156.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 57, a CDR2 sequence set forth as SEQ ID NO: 77, and a CDR3 sequence set forth as SEQ ID NO: 97; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 117, a CDR2 sequence set forth as SEQ ID NO: 137, and a CDR3 sequence set forth as SEQ ID NO: 157.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 58, a CDR2 sequence set forth as SEQ ID NO: 78, and a CDR3 sequence set forth as SEQ ID NO: 98; and/or comprising a LCVR comprising Page 85 of 203 1097657169 ⁇ 1 ⁇ AMERICAS a CDR1 sequence set forth as SEQ ID NO: 118, a CDR2 sequence set forth as SEQ ID NO: 138, and a CDR3 sequence set forth as SEQ ID NO: 158.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 59, a CDR2 sequence set forth as SEQ ID NO: 79, and a CDR3 sequence set forth as SEQ ID NO: 99; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 119, a CDR2 sequence set forth as SEQ ID NO: 139, and a CDR3 sequence set forth as SEQ ID NO: 159.
  • an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 60, a CDR2 sequence set forth as SEQ ID NO: 80, and a CDR3 sequence set forth as SEQ ID NO: 100; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 120, a CDR2 sequence set forth as SEQ ID NO: 140, and a CDR3 sequence set forth as SEQ ID NO: 160.
  • Preferred embodiments include those in which an isolated nucleic acid encodes an anti- B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 41, a CDR2 sequence set forth as SEQ ID NO: 61, and a CDR3 sequence set forth as SEQ ID NO: 81, and a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 101, a CDR2 sequence set forth as SEQ ID NO: 121, and a CDR3 sequence set forth as SEQ ID NO: 141; a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 42, a CDR2 sequence set forth as SEQ ID NO: 62, and a CDR3 sequence set forth as SEQ ID NO: 82, and a LCVR comprising a CDR1 sequence set forth as SEQ ID NO:
  • CARs of the present disclosure may comprise a transmembrane domain that couples the antigen binding domain of the CAR to one or more intracellular domains of the CAR.
  • the transmembrane domain of a CAR of the present disclosure is a region that is capable of spanning the plasma membrane of a cell (e.g., a ⁇ T cell).
  • the transmembrane domain is interposed between the antigen binding domain and the one or more intracellular domains of a CAR.
  • the transmembrane domain is naturally associated with one or more of the domains in the CAR.
  • the transmembrane domain can e selected or modified by one or more amino acid substitutions to avoid binding of such domains to the transmembrane domains of the same or different surface membrane proteins, to minimize interactions with other members of the receptor complex.
  • a transmembrane domain may be derived either from a natural or from a synthetic source. Where the source is natural, the domain may be derived from any membrane-bound or transmembrane protein.
  • Transmembrane regions of particular use Page 87 of 203 1097657169 ⁇ 1 ⁇ AMERICAS in this invention may be derived from (i.e.
  • CD13 comprise at least the transmembrane region(s) of) 4- 1BB/CD137, activating NK cell receptors, an Immunoglobulin protein, B7-H3, BAFFR, BLAME (SLAMF8), BTLA, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, or CD154, CD100 (SEMA4D), CD103, CD160 (BY55), CD18, CD19, CD19a, CD2, CD247, CD27, CD276 (B7-H3), CD28, CD29, CD3 delta, CD3 epsilon, CD3 gamma, CD3 zeta, CD30, CD4, CD40, CD49a, CD49D, CD49f, CD69, CD7, CD84, CD8, CD8alpha, CD8beta, CD96 (Tactile), CD11a, CD11b,
  • the transmembrane domain may be synthetic, in which case it will comprise predominantly hydrophobic residues such as leucine and valine. Preferably a triplet of phenylalanine, tryptophan and valine will be found at each end of a synthetic transmembrane domain.
  • the transmembrane domain comprises a transmembrane domain of CD8.
  • the transmembrane domain of CD8 is a transmembrane domain of CD8 ⁇ .
  • the transmembrane domain of CD8 comprises the amino acid sequence set forth in SEQ ID NO: 211.
  • the transmembrane domain comprises a transmembrane domain of CD28.
  • the transmembrane domain of CD28 comprises the amino acid sequence set forth in SEQ ID NO: 215. In certain embodiments, the transmembrane domain comprises a transmembrane domain of ICOS. In certain embodiments, the transmembrane domain of ICOS comprises the amino acid sequence set forth in SEQ ID NO: 216. Page 88 of 203 1097657169 ⁇ 1 ⁇ AMERICAS [00313] The transmembrane domains described herein can be combined with any of the antigen binding domains described herein, any of the intracellular domains described herein, or any of the other domains described herein that may be included in a subject CAR. [00314] In embodiments, the transmembrane domain further comprises a hinge region.
  • a subject CAR of the present invention may also include a hinge region.
  • the hinge region of the CAR is a hydrophilic region which is located between the antigen binding domain and the transmembrane domain. In embodiments, this domain facilitates proper protein folding for the CAR.
  • the hinge region is an optional component for the CAR.
  • the hinge region may include a domain selected from Fc fragments of antibodies, hinge regions of antibodies, CH2 regions of antibodies, CH3 regions of antibodies, artificial hinge sequences or combinations thereof.
  • hinge regions include, without limitation, a CD8 ⁇ hinge, CD8 ⁇ hinge, CD28 hinge, 4-1BB hinge, CD7 hinge, artificial hinges made of polypeptides which may be as small as, three glycines (Gly), as well as CHI and CH3 domains of IgGs (such as human IgG4). Naturally- occurring hinge domains may be used as wild-type hinge regions or the molecules may be altered.
  • a subject CAR of the present disclosure includes a hinge region that couples the antigen binding domain with the transmembrane domain, which, in turn, couples to one or more intracellular domain(s).
  • the hinge region is preferably capable of supporting the antigen binding domain to recognize and bind to the target antigen on the target cells (see, e.g., Hudecek et al., Cancer Immunol. Res. (2015) 3(2): 125-135).
  • the hinge region is a flexible domain, thus allowing the antigen binding domain to have a structure to optimally recognize the specific structure and density of the target antigens on a cell such as tumor cell (Hudecek et al., supra).
  • the flexibility of the hinge region permits the hinge region to adopt many different conformations.
  • the hinge region is an immunoglobulin heavy chain hinge region.
  • the hinge region is a hinge region polypeptide derived from a receptor (e.g., a CD8-derived hinge region).
  • the hinge region can have a length of from about 4 amino acids to about 50 amino acids, e.g., from about 4 aa to about 10 aa, from about 10 aa to about 15 aa, from about 15 aa to about 20 aa, from about 20 aa to about 25 aa, from about 25 aa to about 30 aa, from about 30 aa to about 40 aa, or from about 40 aa to about 50 aa.
  • the hinge region can have a length of greater than 5 aa, greater than 10 aa, greater than 15 aa, greater than 20 aa, greater than Page 89 of 203 1097657169 ⁇ 1 ⁇ AMERICAS 25 aa, greater than 30 aa, greater than 35 aa, greater than 40 aa, greater than 45 aa, greater than 50 aa, greater than 55 aa, or more.
  • Suitable hinge regions can be readily selected and can be of any of a number of suitable lengths, such as from 1 amino acid (e.g., Gly) to 20 amino acids, from 2 amino acids to 15 amino acids, from 3 amino acids to 12 amino acids, including 4 amino acids to 10 amino acids, 5 amino acids to 9 amino acids, 6 amino acids to 8 amino acids, or 7 amino acids to 8 amino acids, and can be 1, 2, 3, 4, 5, 6, or 7 amino acids.
  • Suitable hinge regions can have a length of greater than 20 amino acids (e.g., 30, 40, 50, 60 or more amino acids).
  • hinge regions include glycine polymers (G)n, glycine-serine polymers (including, for example, (GS)n, (GSGGS)n (SEQ ID NO: 174) and (GGGS)n (SEQ ID NO: 175), where n is an integer of at least one), glycine-alanine polymers, alanine-serine polymers, and other flexible linkers known in the art.
  • Glycine and glycine-serine polymers can be used; both Gly and Ser are relatively unstructured, and therefore can serve as a neutral tether between components.
  • Glycine polymers can be used; glycine accesses significantly more phi-psi space than even alanine, and is much less restricted than residues with longer side chains (see, e.g., Scheraga, Rev. Computational. Chem. (1992) 2: 73-142).
  • Exemplary hinge regions can comprise amino acid sequences including, but not limited to, GGSG (SEQ ID NO: 177), GGSGG (SEQ ID NO: 178), GSGSG (SEQ ID NO: 179), GSGGG (SEQ ID NO: 180), GGGSG (SEQ ID NO: 181), GSSSG (SEQ ID NO: 182), and the like.
  • the hinge region is an immunoglobulin heavy chain hinge region.
  • an immunoglobulin hinge region can include one of the following amino acid sequences: DKTHT (SEQ ID NO: 217); CPPC (SEQ ID NO: 218); CPEPKSCDTPPPCPR (SEQ ID NO: 219) (see, e.g., Glaser et al., J. Biol. Chem.
  • ELKTPLGDTTHT SEQ ID NO: 220
  • KSCDKTHTCP SEQ ID NO: 221
  • KCCVDCP SEQ ID NO: 222
  • KYGPPCP SEQ ID NO: 223
  • EPKSCDKTHTCPPCP SEQ ID NO: 224
  • human IgGl hinge human IgG2 hinge
  • ELKTPLGDTTHTCPRCP SEQ ID NO: 226
  • SPNMVPHAHHAQ SEQ ID NO: 227) (human IgG4 hinge); and the like.
  • the hinge region can comprise an amino acid sequence of a human IgGl, IgG2, IgG3, or IgG4, hinge region.
  • the hinge region can include one or more amino acid substitutions and/or insertions and/or deletions compared to a wild-type (naturally-occurring) hinge region.
  • His229 of human IgGl hinge can be substituted with Tyr, so that the hinge region comprises the sequence EPKSCDKTYTCPPCP (SEQ ID NO: 228); see, e.g., Yan et al., J. Biol. Chem. (2012) 287: 5891-5897.
  • the hinge region can comprise an amino acid sequence derived from human CD8, or a variant thereof.
  • the CAR comprises a CD8 alpha hinge sequence comprising the amino acid sequence set forth in SEQ ID NO: 209.
  • the CAR comprises a hinge and transmembrane domain sequence comprising the amino acid sequence set forth in SEQ ID NO: 213. 3.
  • a CAR encoded by a nucleic acid may further comprise at least one costimulatory domain, wherein the costimulatory domain comprises functional costimulatory signaling domain derived from e.g., a MHC class I molecule, TNF receptor proteins, immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocytic activation molecules (SLAM proteins), activating NK cell receptors, BTLA, a Toll ligand receptor, and the like.
  • the CAR can include 2, 3, 4 or more costimulatory domains.
  • the costimulatory domains may be the same, or they may be different.
  • the costimulatory domains are derived from one or more of TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, CARD11, B7-H3, CEACAM1, CRTAM, CD2, CD3C, CD4, CD7, CD8 ⁇ , CD8 ⁇ , CD11a, CD11b, CD11c, CD11d, IL2R ⁇ , IL2 ⁇ , IL7R ⁇ , IL4R, IL7R, IL15R, IL21R, CD18, CD19, CD19a, CD27, CD28, CD29, CD30, CD40, CDS, CD49a, CD49D, CD49f, CD54 (ICAM), CD69, CD70, CD80, CD83, CD84, CD86, CD96 (Tactile), CD100 (
  • a nucleic acid encoding a CAR encodes at least one 4-1BB costimulatory domain, and optionally a second costimulatory domain selected from 4-1BB, 2B4, ICOS, CD28, OX40, and CD27 costimulatory domains, or any of the above-mentioned costimulatory domains.
  • the nucleic acid encodes at least two 4-1BB costimulatory domains, or at least two 4-1BB costimulatory domains in combination with one, two, three, or four, or more, costimulatory domains selected from 4-1BB, ICOS, CD28, OX40, and CD27, or any of the above-mentioned costimulatory domains.
  • the 4-1BB costimulatory domain comprises an amino acid sequence set forth as SEQ ID NO: 202. In embodiments, the 4-1BB costimulatory domain comprises an amino acid sequence having at least one, at least two, or at least three or more modifications of an amino acid sequence of SEQ ID NO: 202. In embodiments, the 4-1BB costimulatory domain is substantially similar to the 4-1BB costimulatory domain comprising SEQ ID NO: 202.
  • a nucleic acid encoding a CAR encodes at least one CD27 costimulatory domain, and optionally at least one second costimulatory domain selected from 4- 1BB, ICOS, CD28, OX40, 2B4, and CD27 costimulatory domains, or any of the above-mentioned costimulatory domains.
  • the nucleic acid encodes at least one CD27 costimulatory domain, and a 4-IBB costimulatory domain.
  • the nucleic acid encodes two CD27 costimulatory domains, and at least one second costimulatory domain selected from a 4-1BB, ICOS, CD28, and CD27.
  • the CD27 costimulatory domain comprises SEQ ID NO: 208. In embodiments, the CD27 costimulatory domain comprises an amino acid sequence having at least one, at least two, at least three or more modifications of an amino acid sequence of SEQ ID NO: 208. In embodiments, the CD27 costimulatory domain is substantially similar to the CD27 costimulatory domain comprising SEQ ID NO: 208.
  • a nucleic acid encoding a CAR encodes at least one CD28 costimulatory domain, and optionally a second costimulatory domain selected from 4-1BB, 2B4, ICOS, CD28, OX40, and CD27 costimulatory domains, or any of the above-mentioned Page 92 of 203 1097657169 ⁇ 1 ⁇ AMERICAS costimulatory domains.
  • the nucleic acid encodes at least two CD28 costimulatory domains, or at least two CD28 costimulatory domains in combination with one, two, three, or four, or more, costimulatory domains selected from a 4-1BB, ICOS, CD28, OX40, and CD27, or any of the above-mentioned costimulatory domains.
  • the CD28 costimulatory domain comprises SEQ ID NO: 204.
  • the CD28 costimulatory domain comprises SEQ ID NO: 205. Included in SEQ ID NO: 204 and SEQ ID NO: 205 are three subdomains YMNM, PRRP, and PYAP, that are capable to regulate signaling pathways.
  • a disclosed CAR comprises mutation or deletion of one or more of the subdomains (see e.g., WO2019010383).
  • the CD28 costimulatory domain comprises an amino acid sequence having at least one, at least two, at least three or more modifications of an amino acid sequence of SEQ ID NO: 204, or an amino acid sequence of SEQ ID NO: 205.
  • the CD28 costimulatory domain is substantially similar to the CD28 costimulatory domain comprising SEQ ID NO: 204.
  • the CD28 costimulatory domain is substantially similar to the CD28 costimulatory domain comprising SEQ ID NO: 205.
  • a nucleic acid encoding a CAR encodes at least one ICOS costimulatory domain, and optionally a second costimulatory domain selected from 4-1BB, 2B4, ICOS, CD28, OX40, and CD27 costimulatory domains, or any of the above-mentioned costimulatory domains.
  • the nucleic acid encodes at least two ICOS costimulatory domains, or at least two ICOS costimulatory domains in combination with one, two, three, or four, or more, costimulatory domains selected from 4-1BB, ICOS, CD28, OX40, and CD27, or any of the above-mentioned costimulatory domains.
  • the ICOS costimulatory domain comprises SEQ ID NO: 206. In embodiments, the ICOS costimulatory domain comprises an amino acid sequence having at least one, at least two, at least three or more modifications of an amino acid sequence of SEQ ID NO: 206 (see e.g., US20170209492). In embodiments, the ICOS costimulatory domain is substantially similar to the ICOS costimulatory domain comprising SEQ ID NO: 206.
  • a nucleic acid encoding a CAR encodes at least one OX40 costimulatory domain, and optionally a second costimulatory domain selected from 4-1BB, 2B4, ICOS, CD28, OX40, and CD27 costimulatory domains, or any of the above-mentioned costimulatory domains.
  • the nucleic acid encodes at least two OX40 costimulatory domains, or at least two OX40 costimulatory domains in combination with one, two, three, or four, Page 93 of 203 1097657169 ⁇ 1 ⁇ AMERICAS or more, costimulatory domains selected from 4-1BB, ICOS, CD28, OX40, and CD27, or any of the above-mentioned costimulatory domains.
  • the OX40 costimulatory domain comprises SEQ ID NO: 207.
  • the OX40 costimulatory domain comprises an amino acid sequence having at least one, at least two, at least three or more modifications of an amino acid sequence of SEQ ID NO: 207.
  • the OX40 costimulatory domain is substantially similar to the OX40 costimulatory domain comprising SEQ ID NO: 207. 4.
  • Intracellular Signaling Domain a nucleic acid encoding a CAR encodes at least one intracellular signaling domain. In embodiments, the at least one intracellular signaling domain is additional to one or more costimulatory domains. In embodiments, the one or more intracellular signaling domains are included to increase proliferation, persistence, and/or cytotoxic activity of the host cell, preferably a ⁇ cell, harboring the CAR as herein disclosed.
  • the intracellular signaling domain(s) comprise CD3 ⁇ , repeat (e.g., 2-5) DAP10 YINM motifs, signaling domains derived from LFA-1, DAP12, FcR ⁇ , FcR ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD79a, CD79b, CD5, CD22, Fc ⁇ RI, CD66d, and the like.
  • the endodomain of a disclosed CAR can include a plurality (e.g., 2, 3, 4, or more) of intracellular signaling domains. In a case where more than one intracellular signaling domain is included, the intracellular signaling domains may be the same, or they may be different.
  • an intracellular signaling domain of a disclosed CAR is or comprises a CD3 ⁇ signaling domain.
  • a CD3 ⁇ signaling domain is or comprises the amino acid sequence set forth in SEQ ID NO: 229, 231, or 232. 5. Additional Polypeptides [00330]
  • an isolated nucleic acid encoding a CAR of the subject invention can also encode for one or more multicistronic linker region(s) configured to facilitate translation of the CAR polypeptide and one or more additional polypeptides.
  • nucleic acids encoding the one or more additional polypeptides and associated linker region can be positioned at the 3’ end of the isolated nucleic acid, or at the 5’ end of the isolated nucleic acid, or in some examples at both the 5’ end and the 3’ end of the isolated nucleic acid.
  • the linker region(s) can encode a self-cleavage and/or a cleavage polypeptide sequence.
  • the self-cleavage sequence is a 2A self-cleaving sequence (e.g., T2A, P2A, E2A, F2A) Page 94 of 203 1097657169 ⁇ 1 ⁇ AMERICAS which can induce ribosomal skipping during translation of the CAR.
  • the cleavage sequence is a furin sequence.
  • the cleavage sequence e.g., furin cleavage sequence as set forth in SEQ ID NO: 190
  • the multicistronic linker region encodes an internal ribosome entry site.
  • the addition of an optional linker “GSG” or “SGSG” and the like can improve cleavage efficiency. In this way, the one or more additional polypeptides can be release from the CAR and directed to the secretory pathway.
  • the cleavage sequence is the FP2A amino acid sequence as set forth in SEQ ID NO: 184.
  • the cleavage sequence is a P2A amino acid sequence as set forth in SEQ ID NO: 186, or SEQ ID NOs: 188-189.
  • the cleavage sequence is a furin amino acid sequence as set forth in SEQ ID NO: 190.
  • the cleavage sequence is a F2A amino acid sequence as set forth in SEQ ID NO: 191.
  • the cleavage sequence is a E2A amino acid sequence as set forth in SEQ ID NO: 192.
  • the cleavage sequence is a T2A amino acid sequence as set forth in SEQ ID NO: 193.
  • multiple cleavage and/or self-cleavage sequences can be encoded carboxy-terminal to signaling and/or costimulatory domain(s) and amino-terminal to an encoded one or more additional polypeptide.
  • one or more self-cleavage sequences and one or more sequences cleaved by an endogenous protease are encoded in a construct described herein.
  • an endogenous protease recognition site is encoded amino terminal to a self- cleavage sequence.
  • the multi-cistronic linker region encodes an internal ribosome entry site.
  • An exemplary internal ribosome entry site is encoded by the nucleotide sequence set forth in SEQ ID NO: 194.
  • Another exemplary internal ribosome entry site is encoded by the nucleotide sequence set forth in SEQ ID NO: 195.
  • Further suitable internal ribosome entry sites include, but are not limited to, those disclosed e.g., in Nucleic Acids Res.2010 Jan;38(Database issue):D131- 6. doi: 10.1093/nar/gkp981. Epub 2009 Nov 16, those described at iresite.org, those described in WO 2018/215787, the sequence described in GenBank accession No.
  • the one or more additional polypeptides include one or more soluble gamma chain cytokines expressed as separate polypeptides from the CAR.
  • the one or more soluble common gamma chain cytokines can include but are not limited to IL-2, IL-4, IL-7, IL-9, IL-15, IL-21, IL-23.
  • the common gamma chain cytokine is selected from IL-2, IL-7, and IL-15. In embodiments, the common gamma chain cytokine is IL-15.
  • IL-15 sequences, including codon optimized nucleic acid sequences encoding soluble IL-15 (sIL-15) are disclosed herein and in WO 2007/037780.
  • the one or more additional polypeptides include one or more labels or markers, for example to facilitate an ability to monitor CAR expression level, serve as an internal control, and the like.
  • an isolated nucleic acid encoding a CAR encodes for a fluorescent protein, examples of which include but are not limited to green fluorescent protein (GFP), red fluorescent protein (RFP), enhanced GFP (EGFP), enhanced cyan fluorescent protein (ECFP), enhanced yellow fluorescent protein (EYFP), and the like.
  • GFP green fluorescent protein
  • RFP red fluorescent protein
  • EGFP enhanced GFP
  • ECFP enhanced cyan fluorescent protein
  • EYFP enhanced yellow fluorescent protein
  • Other examples can include but are not limited to chloramphenicol acetyltransferase, beta-galactosidase, beta-glucuronidase, beta-lactamase, luciferase, and the like.
  • the one or more additional polypeptides include a protein that is expressed on a cell surface to facilitate detection and/or isolation of cells expressing the protein, e.g., via fluorescent activated cell sorting (FACS); or for enrichment through positive selection using an antibody specific to the encoded protein, e.g., use of an antibody to purify or enrich the cells product on a column or apparatus; or for in vivo binding of an antibody to the protein to enhance or eliminate activity, e.g., to facilitate removal of cells expressing the protein in patients as a safety consideration.
  • FACS fluorescent activated cell sorting
  • Exemplary proteins useful for these purposes include, e.g., CD19, CD20 (Rituxumab recognition domain), RQR8, LNGFR, a truncated form of the human epidermal growth factor receptor (EGFRt), and the like.
  • EGFRt can be targeted by a clinical stage antibody, where such treatment of a patient with the antibody results in elimination of cells containing an isolated nucleic acid encoding a CAR and/or the CAR as disclosed herein. See, e.g., Wang et al.
  • the one or more additional polypeptides include a protein that functions to increase resistance to exhaustion and activation-induced apoptosis and/or upregulate one or more proinflammatory cytokines, costimulatory molecules and/or antigen presentation machinery.
  • LTBR lymphotoxin beta receptor
  • TGF- ⁇ transforming growth factor beta
  • an isolated nucleic acid may encode a dominant negative receptor for TGF-beta (dnTGF ⁇ R2), e.g., as described in Foster et al., J Immunother. (2008); 31: 500-505, WO2019/173324A1, WO2020/183131A1, and WO2020042647A1.
  • dnTGF ⁇ R2 dominant negative receptor for TGF-beta
  • Incorporation of such a dominant negative receptor for TGF-beta may provide a functional advantage over control cells that lack such a dominant negative receptor for TGF-beta in the presence of a TGF-beta-secreting tumor, including enhanced anti-tumor activity.
  • an isolated nucleic acid encodes a signal peptide operably linked to facilitate directing of the one or more additional polypeptides to the secretory pathway.
  • Such one or more additional polypeptides can be those that reside inside certain organelles, are secreted from the host cell, or are inserted into cellular membranes.
  • the signal peptide comprises or consists of the amino acid sequence set forth as SEQ ID NO: 170.
  • the signal peptide comprises or consists of the amino acid sequence set forth as SEQ ID NO: 196.
  • the signal peptide comprises or consists of the amino acid sequence set forth as SEQ ID NO: 235.
  • the signal peptide comprises or consists of the amino acid Page 97 of 203 1097657169 ⁇ 1 ⁇ AMERICAS sequence set forth as SEQ ID NO: 239. In embodiments, the signal peptide comprises or consists of the amino acid sequence set forth as SEQ ID NO: 243. In embodiments, the signal peptide comprises or consists of the amino acid sequence set forth as SEQ ID NO: 247. [00339] In embodiments, the one or more additional polypeptides comprise or consist of the EGFRt amino acid sequence as set forth in SEQ ID NO: 237.
  • the one or more additional polypeptides comprise or consist of the dominant-negative TGF ⁇ receptor II (dnTGF ⁇ R2) amino acid sequence as set forth in SEQ ID NO: 241.
  • the signal peptide comprising or consisting of the amino acid sequence set forth as SEQ ID NO: 239 is operably linked to SEQ ID NO: 241.
  • the one or more additional polypeptides includes the full-length LTBR amino acid sequence as set forth in SEQ ID NO: 245.
  • the signal peptide comprising or consisting of the amino acid sequence set forth as SEQ ID NO: 243 is operably linked to SEQ ID NO: 245.
  • the one or more additional polypeptides includes the LNGFR amino acid sequence as set forth in SEQ ID NO: 249.
  • the signal peptide comprising or consisting of the amino acid sequence set forth as SEQ ID NO: 247 is operably linked to SEQ ID NO: 249.
  • the one or more additional polypeptides includes the sIL-15 amino acid sequence as set forth in SEQ ID NO: 197.
  • the signal peptide comprising or consisting of the amino acid sequence as set forth in SEQ ID NO: 196 is operably linked to SEQ ID NO: 197.
  • the one or more additional polypeptides includes a chimeric switch receptor comprising an extracellular domain of a TGF ⁇ receptor for binding to TGF ⁇ (e.g., TGF ⁇ RI and/or TGF ⁇ RII), and an intracellular domain of a cytokine receptor.
  • the chimeric switch receptors can convert a TGF ⁇ signal into a cytokine signal that promotes cytotoxicity. Examples of such chimeric switch receptors include those descried in WO2012138858, WO2016122738, WO2018094244, WO2014172584, WO2019109980, and WO2022037562, each of which is incorporated by reference in its entirety.
  • the one or more additional polypeptides includes a dominant negative Fas (dnFas).
  • dnFas a dominant negative Fas
  • Incorporation of such a dominant negative Fas in a T cell may provide a functional advantage over control cells that lack such a dominant negative Fas in the prevention of Fas ligand- induced apoptosis and allowing for T cell persistence and antitumor efficacy.
  • the dnFas include that described in Yamamoto TN et al., T cells genetically engineered to overcome Page 98 of 203 1097657169 ⁇ 1 ⁇ AMERICAS death signaling enhance adoptive cancer immunotherapy, J Clin Invest.2019 Feb 25;129(4):1551- 1565, which is incorporated by reference herein in its entirety.
  • the one or more additional polypeptides includes a membrane-bound IL-12 (mbIL-12).
  • mbIL-12 membrane-bound IL-12
  • Incorporation of such a mbIL-12 in a T cell may provide a functional advantage over control cells that lack such a mbIL-12 in enhancing effector functions of the T cells and/or limiting the systemic toxicity associated with IL-12.
  • the mbIL-12 include those described in Hu J. et al., Cell membrane-anchored and tumor-targeted IL-12 (attIL12)-T cell therapy for eliminating large and heterogeneous solid tumors, J Immunother Cancer. 2022 Jan;10(1):e003633; Hombach A.
  • the one or more additional polypeptides includes an antibody or fragment thereof that bind to CD70, or a CAR comprising such antibody or fragment.
  • CD70-binding molecule incorporation of such a CD70-binding molecule in a T cell may provide a functional advantage over control cells that lack the CD70-binding molecule in reducing HvG alloreactivity by targeting CD70+ activated T cells.
  • Examples of such CD70-binding molecules include those described in PCT/US2023/29047, which is incorporated by reference herein in its entirety. 6.
  • Exemplary CARs [00344] The present invention provides nucleic acid molecules encoding one or more CAR constructs described herein.
  • the nucleic acid molecule is provided as a messenger RNA transcript.
  • nucleic acid molecule is provided as a DNA construct.
  • an isolated nucleic acid encodes SEQ ID NO: 293, a CAR polypeptide PL754 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3 ⁇ signaling domain.
  • a nucleic acid encoding a PL754 CAR comprises the sequence of SEQ ID NO: 294.
  • Table 2 below provides annotation of the nucleotide sequence of SEQ ID NO: 294. Page 99 of 203 1097657169 ⁇ 1 ⁇ AMERICAS Table 2.
  • an isolated nucleic acid encodes SEQ ID NO: 297, a CAR polypeptide PL755 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3 ⁇ signaling domain.
  • a nucleic acid encoding a PL755 CAR comprises the sequence of SEQ ID NO: 298. Table 3 below provides annotation of the nucleotide sequence of SEQ ID NO: 298. Table 3.
  • an isolated nucleic acid encodes SEQ ID NO: 301, a CAR polypeptide PL758 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3 ⁇ signaling domain.
  • Page 100 of 203 1097657169 ⁇ 1 ⁇ AMERICAS [00350]
  • a nucleic acid encoding a PL758 CAR comprises the sequence of SEQ ID NO: 302. Table 4 below provides annotation of the nucleotide sequence of SEQ ID NO: 302. Table 4.
  • an isolated nucleic acid encodes SEQ ID NO: 305, a CAR polypeptide PL874 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3 ⁇ signaling domain.
  • a nucleic acid encoding a PL874 CAR comprises the sequence of SEQ ID NO: 306. Table 5 below provides annotation of the nucleotide sequence of SEQ ID NO: 306. Table 5.
  • an isolated nucleic acid encodes SEQ ID NO: 309, a CAR polypeptide PL875 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a Page 101 of 203 1097657169 ⁇ 1 ⁇ AMERICAS CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3 ⁇ signaling domain.
  • a nucleic acid encoding a PL875 CAR comprises the sequence of SEQ ID NO: 310. Table 6 below provides annotation of the nucleotide sequence of SEQ ID NO: 310. Table 6.
  • an isolated nucleic acid encodes SEQ ID NO: 313, a CAR polypeptide PL876 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3 ⁇ signaling domain.
  • a nucleic acid encoding a PL876 CAR comprises the sequence of SEQ ID NO: 314. Table 7 below provides annotation of the nucleotide sequence of SEQ ID NO: 314. Table 7.
  • an isolated nucleic acid encodes SEQ ID NO: 317, a CAR polypeptide PL1007 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3 ⁇ signaling domain.
  • a nucleic acid encoding a PL1007 CAR comprises the sequence of SEQ ID NO: 318. Table 8 below provides annotation of the nucleotide sequence of SEQ ID NO: 318. Table 8.
  • an isolated nucleic acid encodes SEQ ID NO: 321, a CAR polypeptide PL1012 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3 ⁇ signaling domain.
  • a nucleic acid encoding a PL1012 CAR comprises the sequence of SEQ ID NO: 322. Table 9 below provides annotation of the nucleotide sequence of SEQ ID NO: 322. Table 9.
  • an isolated nucleic acid encodes SEQ ID NO: 325, a CAR polypeptide PL1019 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3 ⁇ signaling domain.
  • a nucleic acid encoding a PL1019 CAR comprises the sequence of SEQ ID NO: 326.
  • an isolated nucleic acid encodes SEQ ID NO: 329, a CAR polypeptide PL1025 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3 ⁇ signaling domain.
  • a nucleic acid encoding a PL1025 CAR comprises the sequence of SEQ ID NO: 330.
  • an isolated nucleic acid encodes SEQ ID NO: 333, a CAR polypeptide PL1026 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3 ⁇ signaling domain.
  • a nucleic acid encoding a PL1026 CAR comprises the sequence of SEQ ID NO: 334.
  • Table 12 below provides annotation of the nucleotide sequence of SEQ ID NO: 334.
  • Table 12. PL1026 Nucleotide Sequence Annotation [00367]
  • the above-mentioned CARs and nucleic acids encoding the same include specific signal peptides. In embodiments, it may be desirable to substitute one signal peptide for another in a CAR of the present disclosure.
  • an isolated nucleic acid comprising SEQ ID NO: 296 encodes SEQ ID NO: 295 comprising PL754 minus signal peptide; Page 105 of 203 1097657169 ⁇ 1 ⁇ AMERICAS an isolated nucleic acid comprising SEQ ID NO: 300 encodes SEQ ID NO: 299 comprising PL755 minus signal peptide; an isolated nucleic acid comprising SEQ ID NO: 304 encodes SEQ ID NO: 303 comprising PL758 minus signal peptide; an isolated nucleic acid comprising SEQ ID NO: 308 encodes SEQ ID NO: 307 comprising PL874 minus signal peptide; a nucleic acid comprising SEQ ID NO: 312 encodes SEQ ID NO: 311 comprising PL875 minus signal peptide; an isolated nucleic acid comprising SEQ ID NO: 316 encodes SEQ ID NO: 315 comprising PL876 minus signal peptide; a nucleic acid comprising SEQ ID NO:
  • any nucleic acid sequence encoding a desired signal peptide can be substituted in any of the above-referenced nucleic acid sequences which lack signal peptides, to thereby encode a CAR comprising the desired signal peptide.
  • any of the above-mentioned isolated nucleic acids encoding the particular CAR polypeptides can further encode one or more additional polypeptides, as discussed herein.
  • any of the above-mentioned nucleic acids encoding the particular CARs can include at least one multicistronic linker and a polynucleic acid encoding a dnTGF ⁇ R2 polypeptide. 7.
  • the present invention encompasses a DNA construct comprising sequences of a CAR.
  • the nucleic acid sequences coding for the desired molecules can be obtained using recombinant methods known in the art, such as, for example by screening libraries from cells expressing the gene, by deriving the gene from a vector known to include the same, or by isolating directly from cells and tissues containing the same, using standard techniques. Alternatively, the gene of interest can be produced synthetically, rather than cloned.
  • the present invention provides vectors in which a DNA of the present invention is inserted.
  • Vectors derived from retroviruses such as the lentivirus are suitable tools to achieve long- term gene transfer since they allow long-term, stable integration of a transgene and its propagation Page 106 of 203 1097657169 ⁇ 1 ⁇ AMERICAS in daughter cells.
  • Lentiviral vectors have the added advantage over vectors derived from onco- retroviruses such as murine leukemia viruses in that they can transduce non-proliferating cells, such as hepatocytes. They also have the added advantage of low immunogenicity.
  • the vector comprising the nucleic acid encoding the desired CAR of the invention is an adenoviral vector (A5/35).
  • nucleic acids encoding CARs can be accomplished using of transposons such as sleeping beauty, crisper, CAS9, and zinc finger nucleases.
  • expression of natural or synthetic nucleic acids encoding CARs is typically achieved by operably linking a nucleic acid encoding the CAR polypeptide or portions thereof to a promoter and incorporating the construct into an expression vector.
  • the vectors can be suitable for replication and integration eukaryotes.
  • Typical cloning vectors contain transcription and translation terminators, initiation sequences, and promoters useful for regulation of the expression of the desired nucleic acid sequence.
  • the expression constructs of the present invention may also be used for nucleic acid immunization and gene therapy, using standard gene delivery protocols. Methods for gene delivery are known in the art (e.g., U.S. Pat. Nos.5,399,346, 5,580,859, 5,589,466, incorporated by reference herein in their entireties).
  • the invention provides a gene therapy vector.
  • the nucleic acid can be cloned into a number of types of vectors.
  • the nucleic acid can be cloned into a vector including, but not limited to a plasmid, a phagemid, a phage derivative, an animal virus, and a cosmid.
  • Vectors of particular interest include expression vectors, replication vectors, probe generation vectors, and sequencing vectors.
  • the expression vector may be provided to a cell in the form of a viral vector.
  • Viral vector technology is well known in the art and is described, for example, in Sambrook et al. (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York), and in other virology and molecular biology manuals.
  • Viruses, which are useful as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpes viruses, and lentiviruses.
  • a suitable vector contains an origin of replication functional in at least one organism, a promoter sequence, convenient restriction endonuclease sites, and one or more selectable markers, (e.g., WO 01/96584; WO 01/29058; and U.S. Pat. No.6,326,193).
  • selectable markers e.g., WO 01/96584; WO 01/29058; and U.S. Pat. No.6,326,193
  • a number Page 107 of 203 1097657169 ⁇ 1 ⁇ AMERICAS of viral based systems have been developed for gene transfer into mammalian cells.
  • retroviruses provide a convenient platform for gene delivery systems.
  • a selected gene can be inserted into a vector and packaged in retroviral particles using techniques known in the art. The recombinant virus can then be isolated and delivered to cells of the subject either in vivo or ex vivo.
  • adenovirus vectors are used.
  • a number of adenovirus vectors are known in the art.
  • lentivirus vectors are used.
  • Additional promoter elements e.g., enhancers, regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upstream of the start site, although a number of promoters have recently been shown to contain functional elements downstream of the start site as well. The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another.
  • tk thymidine kinase
  • the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline.
  • individual elements can function either cooperatively or independently to activate transcription.
  • CMV immediate early cytomegalovirus
  • EF-1 ⁇ Elongation Growth Factor-1 ⁇
  • constitutive promoter sequences may also be used, including, but not limited to the simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, an avian leukemia virus promoter, an Epstein-Barr virus immediate early promoter, a Rous sarcoma virus promoter, as well as human gene promoters such as, but not limited to, the actin promoter, the myosin promoter, the hemoglobin promoter, and the creatine kinase promoter. Further, the invention should not be limited to the use of constitutive promoters.
  • inducible promoters are also contemplated as part of the invention.
  • the use of an inducible promoter provides a molecular switch capable of turning on expression of the polynucleotide sequence which it is operatively linked when such expression is desired or turning off the expression when expression is not desired.
  • inducible promoters include, but are not limited to a metallothionine promoter, a glucocorticoid promoter, a progesterone promoter, and a tetracycline promoter.
  • the expression vector to be introduced into a cell can also contain either a selectable marker gene or a reporter gene or both to facilitate identification and selection of expressing cells from the population of cells sought to be transfected or infected through viral vectors.
  • the selectable marker may be carried on a separate piece of DNA and used in a co-transfection procedure. Both selectable markers and reporter genes may be flanked with appropriate regulatory sequences to enable expression in the host cells.
  • Useful selectable markers include, for example, antibiotic-resistance genes, such as neo and the like.
  • Reporter genes are used for identifying potentially transfected cells and for evaluating the functionality of regulatory sequences.
  • a reporter gene is a gene that is not present in or expressed by the recipient organism or tissue and that encodes a polypeptide whose expression is manifested by some easily detectable property, e.g., enzymatic activity. Expression of the reporter gene is assayed at a suitable time after the DNA has been introduced into the recipient cells.
  • Suitable reporter genes may include genes encoding luciferase, beta-galactosidase, chloramphenicol acetyl transferase, secreted alkaline phosphatase, or the green fluorescent protein gene (e.g., Ui-Tei et al., 2000 FEBS Letters 479: 79-82).
  • Suitable expression systems are well known and may be prepared using known techniques or obtained commercially.
  • the construct with the minimal 5' flanking region showing the highest level of expression of reporter gene is identified as the promoter.
  • Such promoter regions may be linked to a reporter gene and used to evaluate agents for the ability to modulate promoter-driven transcription.
  • the vector can be readily introduced into a host cell, e.g., mammalian, bacterial, yeast, or insect cell by any method in the art.
  • the expression vector can be transferred into a host cell by physical, chemical, or biological means.
  • Physical methods for introducing a polynucleotide into a host cell include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and the like. Methods for producing cells comprising vectors and/or exogenous nucleic acids are well- known in the art. See, for example, Sambrook et al. (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York).
  • Biological methods for introducing a polynucleotide of interest into a host cell include the use of DNA and RNA vectors.
  • Viral vectors, and especially retroviral vectors have become the most widely used method for inserting genes into mammalian, e.g., human cells.
  • Other viral vectors can be derived from lentivirus, poxviruses, herpes simplex virus I, adenoviruses and adeno- associated viruses, and the like. See, for example, U.S. Pat.
  • Chemical means for introducing a polynucleotide into a host cell include colloidal dispersion systems, such as macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes.
  • An exemplary colloidal system for use as a delivery vehicle in vitro and in vivo is a liposome (e.g., an artificial membrane vesicle).
  • an exemplary delivery vehicle is a liposome.
  • the nucleic acid may be associated with a lipid.
  • the nucleic acid associated with a lipid may be encapsulated in the aqueous interior of a liposome, interspersed within the lipid bilayer of a liposome, attached to a liposome via a linking molecule that is associated with both the liposome and the oligonucleotide, entrapped in a liposome, complexed with a liposome, dispersed in a solution containing a lipid, mixed with a lipid, combined with a lipid, contained as a suspension in a lipid, contained or complexed with a micelle, or otherwise associated with a lipid.
  • Lipid, lipid/DNA or lipid/expression vector associated compositions are not limited to any particular structure in solution. For example, they may be present in a bilayer structure, as micelles, or with a “collapsed” structure. They may also simply be interspersed in a solution, possibly forming aggregates that are not uniform in size or shape.
  • Lipids are fatty substances which may be naturally occurring or synthetic lipids.
  • lipids include the fatty droplets that naturally occur in the cytoplasm as well as the class of compounds which contain long-chain aliphatic hydrocarbons and their derivatives, such as fatty acids, alcohols, amines, amino alcohols, and aldehydes. [00385] Lipids suitable for use can be obtained from commercial sources.
  • dimyristyl phosphatidylcholine can be obtained from Sigma, St. Louis, Mo.
  • dicetyl phosphate can be obtained from K & K Laboratories (Plainview, N.Y.); cholesterol (“Choi”) can be obtained from Calbiochem-Behring
  • dimyristyl phosphatidylglycerol (“DMPG”) Page 110 of 203 1097657169 ⁇ 1 ⁇ AMERICAS and other lipids may be obtained from Avanti Polar Lipids, Inc. (Birmingham, AL).
  • Stock solutions of lipids in chloroform or chloroform/methanol can be stored at about -20.degree. C.
  • Liposome is a generic term encompassing a variety of single and multilamellar lipid vehicles formed by the generation of enclosed lipid bilayers or aggregates. Liposomes can be characterized as having vesicular structures with a phospholipid bilayer membrane and an inner aqueous medium. Multilamellar liposomes have multiple lipid layers separated by aqueous medium. They form spontaneously when phospholipids are suspended in an excess of aqueous solution.
  • compositions that have different structures in solution than the normal vesicular structure are also encompassed.
  • the lipids may assume a micellar structure or merely exist as nonuniform aggregates of lipid molecules.
  • lipofectamine-nucleic acid complexes are also contemplated.
  • assays include, for example, “molecular biological” assays well known to those of skill in the art, such as Southern and Northern blotting, RT-PCR and PCR; “biochemical” assays, such as detecting the presence or absence of a particular peptide, e.g., by immunological means (ELISAs and Western blots) or by assays described herein to identify agents falling within the scope of the invention. 8.
  • CAR polypeptides of the present disclosure may be expressed via their corresponding nucleic acid constructs in a wide variety of host cells.
  • the host cells are mammalian cells.
  • Host cells, as described herein, can be stored, e.g., cryopreserved, for use in adoptive cell transfer.
  • the host cells are stored prior to engineering the cells to express a CAR polypeptide.
  • the cells are engineered to express a CAR polypeptide and then the cells are stored.
  • Page 111 of 203 1097657169 ⁇ 1 ⁇ AMERICAS [00388]
  • Preferred host cells for use with the CARs of the present disclosure comprise immune cells.
  • Such cells may be obtained from the subject to be treated (i.e. are autologous) or, alternatively, immune cell lines or donor immune cells (allogeneic, syngeneic) can be used.
  • Immune cells can be obtained from a number of sources, including from peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors.
  • Immune cells can be obtained from blood collected from a subject using any number of techniques known to the skilled artisan, such as FicollTM separation. For example, cells from the circulating blood of an individual may be obtained by apheresis.
  • immune cells are isolated from peripheral blood lymphocytes by lysing the red blood cells and depleting the monocytes, for example, by centrifugation through a PERCOLLTM gradient or by counterflow centrifugal elutriation.
  • a specific subpopulation of immune cells can be further isolated by positive or negative selection techniques.
  • immune cells can be isolated using a combination of antibodies directed to surface markers unique to the positively selected cells, e.g., by incubation with antibody- conjugated beads for a time period sufficient for positive selection of the desired immune cells.
  • enrichment of immune cell populations can be accomplished by negative selection using a combination of antibodies directed to surface markers unique to the negatively selected cells.
  • Other specific manners of isolation and/or enrichment are disclosed herein.
  • the immune cells comprise any leukocyte involved in defending the body against infectious disease and foreign materials.
  • the immune cells can comprise lymphocytes, monocytes, macrophages, dendritic cells, mast cells, neutrophils, basophils, eosinophils, or any combinations thereof.
  • immune cells relevant to the present disclosure can include but are not limited to ⁇ T cells, ⁇ ⁇ ⁇ T cells, NK cells, NKT cells, ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ cells, B cells, innate lymphoid cells (ILCs), cytokine induced killer (CIK) cells, cytotoxic T lymphocytes (CTLs), lymphokine activated killer (LAK) cells, regulatory T cells, and the like.
  • preferred immune cells comprise ⁇ T cells, ⁇ ⁇ ⁇ T cells, NK cells, NKT cells, ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ cells, and/or, in some examples, macrophages.
  • preferred immune cells comprise ⁇ ⁇ ⁇ T cells.
  • the immune cells relevant to the present disclosure comprise allogeneic cells, autologous cells, or syngeneic cells.
  • Page 112 of 203 1097657169 ⁇ 1 ⁇ AMERICAS Page 112 of 203 1097657169 ⁇ 1 ⁇ AMERICAS
  • aspects of the invention include host cells, ⁇ T cells in some preferred embodiments, that functionally express an isolated nucleic acid described herein, and thereby express a CAR on the surface of the cell.
  • Aspects of the invention can additionally or alternatively include host cells, preferably ⁇ T cells, having in vitro or in vivo cytotoxic activity against a tumor cell that exhibits cell surface expression of B7H6.
  • the cytotoxic activity is innate activity. In some cases, the cytotoxicity is at least in part, significantly (> about 25%), or entirely, due to the presence of a CAR construct having a binding domain that specifically binds B7H6 expressed on the surface of the tumor cell.
  • the host cells preferably ⁇ T cells, exhibit tumor cell killing activity that is greater than an innate level of in vitro and/or in vivo tumor cell killing activity in a control cell of the same cell type. In some cases, the control cell does not comprise a CAR construct.
  • control cell comprises a CAR construct lacking a binding domain described herein, a hinge region described herein, a transmembrane domain described herein, an intracellular signaling domain described herein, and/or a costimulatory domain described herein.
  • the cytotoxicity is at least in part, significantly (> about 25%), or entirely, due to the presence of a CAR construct having a binding domain that specifically binds B7H6 or an epitope within B7H6.
  • the host cells preferably ⁇ T cells, functionally express a B7H6-specific CAR encoded by an isolated nucleic acid described herein.
  • the ⁇ T cells can exhibit HLA- restricted (e.g., HLA class I restricted) cytotoxicity. In other embodiments, most (>50%), substantially all (>90%), or all of the cytotoxic activity is not HLA-restricted (e.g., HLA class I restricted). HLA-restricted cytotoxic activity can be assessed by comparing in vitro cytotoxicity against an HLA (e.g., HLA class I) (null) tumor cell line versus in vitro cytotoxicity against an HLA+ (e.g., HLA class I + ) tumor cell line.
  • HLA HLA- restricted
  • HLA+ HLA+
  • the HLA-restricted cytotoxic activity is at least in part, significantly (>25%), or entirely, provided by the use of a T cell Receptor-like binding domain.
  • T cell receptor like binding domains are binding domains that specifically recognize the antigen when presented on the surface of a cell in complex with an MHC molecule.
  • T cell Receptor-like binding domains are further described, e.g., in WO 2016/199141. Page 113 of 203 1097657169 ⁇ 1 ⁇ AMERICAS [00395]
  • Host cells described herein, preferably ⁇ T cells can exhibit robust and/or persistent tumor cell killing activity.
  • the tumor cell killing activity can persist for at least about 6 days to 120 days, or for at least about 6 days to 180 days, from first contact with a tumor cell.
  • the tumor cell killing activity of a host cell described herein, preferably a ⁇ T cell, or a progeny thereof can persist for at least about 6 days to 120 days, or for at least about 6 days to 180 days, from first contact with a tumor cell, or from administration of the host cell. This persistent tumor cell killing activity can be exhibited in vitro, in vivo, or both in vitro and in vivo.
  • aspects of the invention can additionally or alternatively include host cells, preferably ⁇ T cells, that proliferate in response to contact with cells that exhibit cell surface expression, or overexpression, of B7H6.
  • the cells that exhibit cell surface expression, or overexpression, of B7H6 can be tumor cells or can be non-tumor cells.
  • the proliferation is an innate activity. In some cases, the proliferation is at least in part, significantly (> about 20% or > about 25%), or entirely, due to the presence of a CAR construct having a binding domain that specifically binds B7H6 expressed on the surface of a tumor cell.
  • the host cells preferably ⁇ T cells
  • the control cell does not comprise a CAR construct.
  • the control cell comprises a CAR construct lacking a binding domain described herein, a hinge region described herein, a transmembrane domain described herein, an intracellular signaling domain described herein, and/or a costimulatory domain described herein.
  • Host cells as described herein, preferably ⁇ T cells can exhibit robust and/or persistent proliferation in a host organism that comprises a cell, for example a tumor cell, that exhibits cell surface expression, or overexpression, of B7H6.
  • the proliferation can persist for at least about 6 days to 120 days, or for at least about 6 days to 180 days, from first contact with a tumor cell or from a date of administration of the host cell, preferably a ⁇ T cell, to the host organism.
  • the proliferation of a host cell, preferably a ⁇ T cell described herein, or a progeny thereof, in the host organism that comprises the cell that exhibits cell surface expression, or overexpression, of B7H6 can persist for at least about 6 days to 120 days, or for at least about 6 days to 180 days, from first contact with a B7H6-expressing cell or from the date of first administration of the host cell, preferably a ⁇ T cell, to the host organism.
  • the proliferation in the host organism is at least in part, significantly (> about 20% or > about 25%), Page 114 of 203 1097657169 ⁇ 1 ⁇ AMERICAS or entirely, due to the presence of a CAR construct having a binding domain that specifically binds B7H6 or an epitope within B7H6.
  • host cells preferably ⁇ T cells, exhibiting proliferation in the host organism comprising a cell that exhibits cell surface expression of B7H6 functionally express a B7H6 specific CAR encoded by an isolated nucleic acid described herein.
  • the host cells preferably ⁇ T cells described herein, express, or persistently express, pro-inflammatory cytokines such as tumor necrosis factor alpha or interferon gamma after contact with a B7H6-expressing cell.
  • the host cells described herein, or progeny thereof express, or persistently express, pro-inflammatory cytokines such as tumor necrosis factor alpha or interferon gamma after contact with the B7H6-expressing cell, e.g., in a host organism comprising the B7H6-expressing cell.
  • a ⁇ T cell, or a pharmaceutical composition containing the ⁇ T cell exhibits essentially no, or no graft versus host response when introduced into an allogeneic host.
  • the ⁇ T cell, or a pharmaceutical composition containing the ⁇ T cell exhibits a clinically acceptable level of graft versus host response when introduced into an allogeneic host.
  • a clinically acceptable level is an amount of graft versus host response that does not require cessation of a ⁇ T cell treatment to achieve a therapeutically effective treatment.
  • a clinically acceptable level of graft versus host response is an acute response that is less severe than Grade C according to an applicable IBMTR grading scale.
  • the severity of acute graft versus host response is determined by an assessment of the degree of involvement of the skin, liver, and gastrointestinal tract. The stages of individual organ involvement are combined to produce an overall grade, which has prognostic significance.
  • Grade I(A) GvHD is characterized as mild disease, grade II(B) GvHD as moderate, grade III(C) as severe, and grade IV(D) life-threatening.
  • the IBMTR grading system defines the severity of acute GvHD as follows (Rowlings et al., Br J Haematol 1997; 97:855): ⁇ Grade A – Stage 1 skin involvement alone (maculopapular rash over ⁇ 25 percent of the body) with no liver or gastrointestinal involvement ⁇ Grade B – Stage 2 skin involvement; Stage 1 to 2 gut or liver involvement ⁇ Grade C – Stage 3 involvement of any organ system (generalized erythroderma; bilirubin 6.1 to 15.0 mg/dL; diarrhea 1500 to 2000 mL/day) Page 115 of 203 1097657169 ⁇ 1 ⁇ AMERICAS ⁇ Grade D – Stage 4 involvement of any organ system (generalized erythroderma with bullous formation; bilirubin >15 mg/dL; diarrhea >2000 mL/day OR pain OR ileus).
  • a ⁇ T cell, or a pharmaceutical composition containing the ⁇ T cell exhibits reduced or substantially reduced graft versus host response when introduced into an allogeneic host as compared to a graft versus host response exhibited by control ⁇ T cells, or a control pharmaceutical composition comprising the control ⁇ T cells, administered to an allogeneic host.
  • the control ⁇ T cell is an allogeneic non-engineered control ⁇ T cell.
  • control ⁇ T cell does not comprise a CAR or does not comprise the same CAR as a reference ⁇ T cell.
  • host cells preferably ⁇ T cells, described herein can be modified to comprise one or more gene edits.
  • gene editing is a type of genetic engineering in which nucleotide(s)/nucleic acid(s) is/are inserted, deleted, and/or substituted in a DNA sequence, such as the genome of a ⁇ T cell. Targeted gene editing enables insertion, deletion, and/or substitution at pre-selected sites in the genome of a targeted cell.
  • a “disrupted gene” refers to a gene comprising an insertion, deletion or substitution relative to an endogenous gene such that expression of a functional protein from the endogenous gene is reduced or inhibited.
  • disrupting a gene refers to a method of inserting, deleting, or substituting at least one nucleotide/nucleic acid in an endogenous gene such that expression of a functional protein from the endogenous gene is reduced or inhibited. Methods of disrupting a gene are known to those of skill in the art, and described, e.g., in United States Patent No.11254912, incorporated herein by reference in its entirety.
  • a nuclease-dependent approach can be used to conduct targeted gene editing of a T cell. Such a nuclease-dependent approach can achieve targeted editing through the specific introduction of double strand breaks (DSBs) by specific endonucleases.
  • DSBs double strand breaks
  • Such nuclease- Page 116 of 203 1097657169 ⁇ 1 ⁇ AMERICAS dependent targeted editing utilizes DNA repair mechanisms, for example, non-homologous end joining (NHEJ), which occurs in response to DSBs.
  • NHEJ non-homologous end joining
  • DNA repair by NHEJ often leads to random insertions or deletions (indels) of a small number of endogenous nucleotides.
  • Indels random insertions or deletions
  • repair can also occur by a homology directed repair (HDR).
  • HDR homology directed repair
  • Available endonucleases capable of introducing specific and targeted DSBs include, but are not limited to, zinc-finger nucleases (ZFN), transcription activator-like effector nucleases (TALEN), and RNA-guided CRISPR-Cas9 nuclease (CRISPR/Cas9; Clustered Regular Interspaced Short Palindromic Repeats Associated 9).
  • ZFN zinc-finger nucleases
  • TALEN transcription activator-like effector nucleases
  • CRISPR/Cas9 Clustered Regular Interspaced Short Palindromic Repeats Associated 9
  • a CRISPR system can include a non-coding RNA molecule (e.g., guide RNA) that binds DNA and Cas proteins (e.g., Cas9) with nuclease functionality (Sander et al., Nature Biotechnology (2014); 32:347-355; Hsu et al., Cell (2014); 157(6):1262-1278).
  • a host cell preferably a ⁇ T cell, comprises one or more disrupted genes.
  • one or more genes whose expression is disrupted can comprise adenosine A2a receptor (ADORA), CD276, V-set domain containing T cell activation inhibitor 1 (VTCN1), B and T lymphocyte associated (BTLA), cytotoxic T-lymphocyte-associated protein 4 (CTLA4), indoleamine 2,3-dioxygenase 1 (IDO1), killer cell immunoglobulin-like receptor, three domains, long cytoplasmic tail, 1 (KIR3DL1), lymphocyte-activation gene 3 (LAG3), programmed cell death 1 (PD-1), hepatitis A virus cellular receptor 2 (HAVCR2), V-domain immunoglobulin suppressor of T-cell activation (VISTA), natural killer cell receptor 2B4 (CD244), cytokine inducible SH2-containing protein (CISH), hypoxanthine phosphoribosyltransferase 1 (HPRT), adeno-associated virus integration site (AAVS SITE (E.G.
  • ADORA
  • CCR5 chemokine (C—C motif) receptor 5 (gene/pseudogene) (CCR5), CD160 molecule (CD160), T- cell immunoreceptor with Ig and ITIM domains (TIGIT), CD96 molecule (CD96), cytotoxic and regulatory T-cell molecule (CRTAM), leukocyte associated immunoglobulin like receptor 1(LAIR1), sialic acid binding Ig like lectin 7 (SIGLEC7), sialic acid binding Ig like lectin 9 (SIGLEC9), tumor necrosis factor receptor superfamily member 10b (TNFRSF10B), tumor necrosis factor receptor superfamily member 10a (TNFRSF10A), caspase 8 (CASP8), caspase 10 (CASP10), caspase 3 (CASP3), caspase 6 (CASP6), caspase 7 (CASP7), Fas associated via death Page 117 of 203 1097657169 ⁇ 1 ⁇ AMERICAS domain (FADD)
  • CD160 CD160
  • a gene whose expression is disrupted is CISH, a negative regulator of TCR signaling.
  • Disruption of the CISH gene may provide a functional advantage over control cells that have an intact CISH gene in improving the sensitivity to certain cytokines (e.g., IL-2/IL- 15), increasing T cell proliferation, and/or limiting T cell exhaustion.
  • the CISH gene may be disrupted by methods described in Daher M. et al, Targeting a cytokine checkpoint enhances the fitness of armored cord blood CAR-NK cells, Blood. 2021 Feb 4;137(5):624-636, which is incorporated by reference herein in its entirety.
  • the CISH gene is disrupted by gene editing using an RNA-guided nuclease system comprising one or more guide RNAs comprising the sequences of any one of SEQ ID NOs: 198-201 and 339-340.
  • a gene whose expression is disrupted is CBL-B, a negative regulator of T cell activation. Disruption of the CBL-B gene may provide a functional advantage over control cells that have an intact CBL-B gene in enhancing T cell activation.
  • the CBL- B gene may be disrupted by methods described in Augustin R. et al., Targeting Cbl-b in cancer immunotherapy, J Immunother Cancer.
  • the CBL-B gene is disrupted by gene editing using a CRISPR-Cas system comprising one or more guide RNAs comprising the sequence of any one of SEQ ID NOs: 341-344.
  • a gene whose expression is disrupted is Roquin (e.g., Roquin-1). Disruption of the Roquin gene may provide a functional advantage over control cells that have an intact Roquin gene in increasing T cell proliferation and enhancing antitumor activity.
  • the Roquin gene may be disrupted by methods described in Mai D et al., Combined disruption of T cell inflammatory regulators Regnase-1 and Roquin-1 enhances antitumor activity of engineered human T cells, Proc Natl Acad Sci U S A. 2023 Mar 21;120(12):e2218632120, which is incorporated by reference herein in its entirety.
  • a gene whose expression is disrupted is ZFP91. Disruption of the ZFP91 gene may provide a functional advantage over control cells that have an intact ZFP91 gene in improving T cell glycolytic fitness and effector function.
  • the ZFP91 gene may be disrupted by method described in Wang F.
  • a gene whose expression is disrupted is CD58.
  • a gene whose expression is disrupted is ICAM-1.
  • both CD58 and ICAM-1 are disrupted. Disruption of the CD58 and/or ICAM-1 genes may provide a functional advantage over control cells that have an intact CD58 and/or ICAM-1 gene in disrupting T cell adhesion and co- stimulatory interactions to reduce Host vs Graft allocytotoxicity.
  • the ICAM-1 gene may be disrupted as described in Teo HY et al.
  • the ICAM-1 gene is disrupted by gene editing using an RNA-guided nuclease system (e.g., CRISPR-Cas or CRISPR-Mad7 system) comprising one or more guide RNAs comprising the sequences of any one of SEQ ID NOs: 349-350.
  • an RNA-guided nuclease system e.g., CRISPR-Cas or CRISPR-Mad7 system
  • guide RNAs comprising the sequences of any one of SEQ ID NOs: 349-350.
  • the CD58 gene is disrupted by gene editing using an RNA-guided nuclease system (e.g., CRISPR-Cas or CRISPR- Mad7 system) comprising one or more guide RNAs comprising the sequences of any one of SEQ ID NOs: 351-352.
  • RNA-guided nuclease system e.g., CRISPR-Cas or CRISPR- Mad7 system
  • host cells of the present disclosure preferably ⁇ T cells, can be modified to include a nucleic acid construct that encodes a protein which imparts a desired functionality to the host cells.
  • a nucleic acid may encode for a chimeric DAP10 adaptor polypeptide, described in U.S. Provisional Application No.
  • a chimeric DAP10 adaptor polypeptide is capable of associating with a chimeric antigen receptor of the present disclosure, for example a CAR of the present disclosure may comprise a DAP10- interacting domain.
  • the chimeric DAP10 adaptor polypeptide may also associate with one or more additional endogenous or exogenous polypeptides, for example endogenous or exogenous NKG2D.
  • a chimeric DAP10 adaptor polypeptide may not associate with a CAR of the present disclosure, but instead may interact with an endogenous or exogenous polypeptide (e.g., NKG2D) that includes a DAP10-interacting domain.
  • an endogenous or exogenous polypeptide e.g., NKG2D
  • NKG2D exogenous polypeptide
  • a chimeric DAP10 adaptor polypeptide may not associate with a CAR of the present disclosure, but instead may interact with an endogenous or exogenous polypeptide (e.g., NKG2D) that includes a DAP10-interacting domain.
  • an endogenous or exogenous polypeptide e.g., NKG2D
  • the antibody, antigen-binding fragments, or compositions provided herein can be used in diagnostic or therapeutic procedures.
  • One embodiment provided herein is directed to a method of diagnosing the presence of a tumor or a cancer growth in a subject.
  • a method includes determining the presence of B7H6 in a sample suspected of containing the B7H6 polypeptide.
  • the sample may contain cells, which may be cancer cells, suspected of expressing B7H6.
  • a method comprises exposing the sample to an antibody or antigen-binding fragment as herein disclosed, whereby specific binding of the antibody or antigen-binding fragment to the sample is indicative of the presence of a tumor or cancer growth in the subject.
  • the antibody employed in the method may optionally be detectably labeled, attached to a solid support, or the like.
  • Detectable labels can include but are not limited to a photoactivatable agent, a fluorophore, a radioisotope, a bioluminescent protein, a bioluminescent peptide, a fluorescent tag, a fluorescent protein, or a fluorescent peptide.
  • exemplary assays for achieving detection of a signal from a label includes assays routinely used in the art such as, but not limited to flow cytometry, ELISA, Western blotting, immunohistochemistry, membrane assays, and microscopic imaging. H.
  • the composition comprising an antibody or antigen-binding fragment thereof of the present invention is administered to a subject having a disease involving inappropriate expression of a target antigen, a protein or other molecule.
  • the composition comprising an antibody or antibody fragment that binds to B7H6 is administered to detect the presence, abundance, location, or combination thereof of B7H6 in the subject.
  • this is meant to include diseases and disorders characterized by aberrant proteins, due for example to alterations in the amount of a protein present, protein localization, posttranslational modification, conformational state, the presence of a mutant or pathogen protein, etc.
  • the disease or disorder may be characterized by alterations molecules including but not limited to polysaccharides and gangliosides.
  • An overabundance may be due to any cause, including but not limited to overexpression at the molecular level, prolonged or accumulated appearance at the site of action, or increased activity of a protein relative to normal. Included within this definition are diseases and disorders characterized by a reduction of a protein.
  • This reduction may be due to any cause, including but not limited to reduced expression at the molecular level, shortened or reduced appearance at the site of action, mutant forms of a protein, or decreased activity of a protein relative to normal.
  • Such an overabundance or reduction of a protein can be measured relative to normal expression, appearance, or activity of a protein, and the measurement may play an important role in the development and/or clinical testing of the antibodies of the present invention.
  • antibody or antibody fragment of the present invention binds the B7H6 antigen expressed on tumor cells when administrated in a subject; in another embodiment, antibody or antibody fragment of the present invention administrated in a subject binds the antigen expressed on hematological and/or solid tumor cells, including but not limited to colon cancer, pancreatic cancer, Non-Hodgkin Lymphoma, cervical cancer, hepatic cancer, gastro-intestinal cancer, lung cancer, ovarian cancer, glioblastoma, prostate cancer, breast cancer, ovarian cancer, kidney cancer, bladder cancer, melanoma, and glioma.
  • Detection methods for identification of binding can be direct or indirect and can include, for example, the measurement of light emission, radioisotopes, calorimetric dyes and fluorochromes.
  • Direct detection includes methods that operate without intermediates or secondary measuring procedures to assess the amount of bound antigen or ligand. Such methods generally employ ligands that are themselves labeled by, for example, radioactive, light emitting or Page 121 of 203 1097657169 ⁇ 1 ⁇ AMERICAS fluorescent moieties.
  • indirect detection includes methods that operate through an intermediate or secondary measuring procedure. These methods generally employ molecules that specifically react with the antigen or ligand and can themselves be directly labeled or detected by a secondary reagent.
  • an antibody specific for a ligand can be detected using a secondary antibody capable of interacting with the first antibody specific for the ligand, again using the detection methods described above for direct detection. Indirect methods can additionally employ detection by enzymatic labels. Moreover, for the specific example of screening for catalytic antibodies, the disappearance of a substrate or the appearance of a product can be used as an indirect measure of binding affinity or catalytic activity.
  • the antibodies, antigen-binding fragments thereof, and/or engineered cells of the present disclosure can be administered to a subject per se, or in a pharmaceutical composition where it is mixed with suitable carriers or excipients.
  • the antibodies and/or engineered cells of the invention may be administered by any route appropriate to the condition to be treated. Administration is typically parenterally, i.e. infusion, subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural.
  • Therapeutic formulations comprising an anti-B7H6 antibody used in accordance with the present invention are prepared for storage by mixing the antibody having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington’s Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as acetate, Tris, phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparag
  • compositions to be used for in vivo administration are generally sterile. This is readily accomplished by filtration through sterile filtration membranes.
  • the active ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano- particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano- particles and nanocapsules
  • Sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semi-permeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2- hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and ⁇ ethyl-L-glutamate non-degradable ethylene-vinyl acetate
  • degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT® (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate)
  • poly- D-(-)-3-hydroxybutyric acid While polymers such as ethylene-vinyl acetate and lactic acid- glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods.
  • encapsulated immunoglobulins When encapsulated immunoglobulins remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37oC, resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S-S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.
  • An antibody may be formulated in any suitable form for delivery to a target cell/tissue.
  • antibodies may be formulated as immunoliposomes.
  • a “liposome” is a small vesicle composed of various types of lipids, phospholipids and/or surfactant which is useful for delivery of a drug to a mammal. The components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes. Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci.
  • Liposomes with enhanced circulation time are disclosed in U.S. Patent No.5,013,556.
  • Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG- derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • Fab' fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et al., J. Biol. Chem. 257: 286-8 (1982) via a disulfide interchange reaction.
  • a chemotherapeutic agent is optionally contained within the liposome (See Gabizon et al., J. National Cancer Inst. 81(19): 1484 (1989)).
  • compositions for administration in the methods of the invention are known in the art (see, e.g., Remington's Pharmaceutical Sciences (1990) 18th ed., Mack Publishing Co., Easton, Pa.; The Merck Index (1996) 12th ed., Merck Publishing Group, Whitehouse, N.J.; and Pharmaceutical Principles of Solid Dosage Forms, Technonic Publishing Co., Inc., Lancaster, Pa., (1993)).
  • the pharmaceutical formulations can be packaged in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the pharmaceutical carrier or excipient.
  • Pharmaceutical compositions suitable for use in context of some embodiments of the invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. More specifically, a therapeutically effective amount means an Page 124 of 203 1097657169 ⁇ 1 ⁇ AMERICAS amount of active ingredients (anti-B7H6 antibody, antigen-binding fragment thereof, CAR- modified immune cells) effective to prevent, alleviate or ameliorate symptoms of a disorder (e.g., cancer) or prolong the survival of the subject being treated.
  • active ingredients anti-B7H6 antibody, antigen-binding fragment thereof, CAR- modified immune cells
  • the therapeutically effective amount or dose can be estimated initially from in vitro and cell culture assays.
  • a dose can be formulated in animal models to achieve a desired concentration or titer. Such information can be used to more accurately determine useful doses in humans.
  • Toxicity and therapeutic efficacy of the active ingredients described herein can be determined by standard pharmaceutical procedures in vitro, in cell cultures or experimental animals. The data obtained from these in vitro and cell culture assays and animal studies can be used in formulating a range of dosage for use in human.
  • the dosage may vary depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl, et al., 1975, in "The Pharmacological Basis of Therapeutics", Ch.1 p. l). [00426] Depending on the severity and responsiveness of the condition to be treated, dosing can be of a single or a plurality of administrations, with course of treatment lasting from several days to several weeks or until cure is effected or diminution of the disease state is achieved.
  • compositions comprising anti-B7H6 antibodies, antigen-binding fragments thereof, or CAR-modified immune cells comprising the same, as described herein, may be administered for prophylactic and/or therapeutic treatments.
  • An embodiment of the present invention provides a method of treating a condition associated with abnormal B7H6 expression, for example a B7H6-expressing cancer, in a subject.
  • the method may include targeting a B7H6-expressing cell, for example a B7H6-expressing tumor cell, with a therapeutically effective amount of the antibody or antigen-binding fragment.
  • the method comprises administering to the subject a composition comprising a therapeutically effective amount of anti-B7H6 antibodies, antigen- binding fragments thereof, or CAR-modified immune cells comprising the same.
  • the CAR-modified immune cells are CAR-modified ⁇ T cells.
  • the anti-B7H6 antibody is used in the form of an ADC.
  • the anti-B7H6 antibody may be coupled to a biologically active agent or a combination of such agents including but not limited to a radioisotope, a toxin, and the like.
  • a method of treating a hematological or solid tumor with abnormal B7H6 expression may comprise the step of targeting B7H6- expressing cells with a therapeutically effective amount of an anti-B7H6 antibody, antigen-binding fragment thereof, or CAR-modified immune cell comprising the same.
  • the CAR- modified immune cells are CAR-modified ⁇ T cells.
  • embodiments include administering to a subject having a tumor associated with abnormal B7H6 expression, an anti- B7H6 antibody, antigen-binding fragment thereof, or CAR-modified immune cell comprising the same.
  • the anti-B7H6 antibody is used in the form of an ADC.
  • a method of inhibiting cell growth or proliferation comprising exposing a B7H6-expressing cell to an anti-B7H6 antibody or antigen-binding fragment, or a CAR-modified immune cell comprising the same, thereof under conditions permissive for binding of the antibody or antigen-binding fragment to B7H6.
  • the CAR- modified immune cells are CAR-modified ⁇ T cells.
  • “Inhibiting cell growth or proliferation” refers to decreasing a cell’s growth or proliferation by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100%, and can include inducing cell death.
  • the B7H6- expressing cell is a tumor cell.
  • an anti-B7H6 antibody or antigen-binding fragment, or a CAR- modified immune cell comprising the same is used to treat or prevent a cell proliferative disorder.
  • the CAR-modified immune cells are CAR-modified ⁇ T cells.
  • the cell proliferative disorder is associated with abnormal expression and/or activity of B7H6.
  • the cell proliferative disorder is associated with Page 126 of 203 1097657169 ⁇ 1 ⁇ AMERICAS increased expression of B7H6 on the surface of a cell.
  • the cell proliferative disorder is a tumor or a cancer.
  • Pharmaceutical compositions comprising engineered host cells that express a CAR, and/or admixtures thereof, may be administered for prophylactic and/or therapeutic treatments.
  • a pharmaceutical composition comprises ⁇ T cells engineered to express a B7H6-targeting CAR.
  • An admixture may comprise non-engineered cells of a same or different type.
  • an admixture may comprise engineered cells of a same type that express a different B7H6-targeting CAR. Additionally or alternatively, an admixture may comprise engineered cells of a different type that express a same or different B7H6-targeting CAR. For example and without limitation, an admixture may comprise a population of ⁇ T cells engineered to express a B7H6-targeting CAR, and a population of non-engineered ⁇ T cells.
  • an admixture may comprise a population of ⁇ T cells engineered to express a B7H6-targeting CAR, and a population of engineered or non-engineered cells e.g., NK cells, NKT cells, ⁇ cells, ⁇ cells, etc.
  • the compositions can be administered to a subject already suffering from a disease or condition in an amount sufficient to decrease at least one sign or symptom associated with the disease or condition. In embodiments, the amount is sufficient to cure the disease or condition.
  • An engineered host cell population and/or admixtures thereof can also be administered to lessen a likelihood of developing, contracting, or worsening a condition.
  • Effective amount of a population of engineered host cells, non-engineered host cell, and/or admixtures thereof, for therapeutic use can vary based on the severity and course of the disease or condition, previous therapy, the subject’s health status, weight, and/or response to various drugs, and/or the judgement of a treating physician.
  • the one or multiple engineered host cell populations, non-engineered cells and/or admixtures thereof, of the present disclosure can be used to treat a subject in need of treatment for a condition. Examples of such conditions include but are not limited to cancer.
  • Subjects can be humans, non-human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the Page 127 of 203 1097657169 ⁇ 1 ⁇ AMERICAS like.
  • a subject can be of any age. Subjects can be, for example, elderly adults, adults, adolescents, pre-adolescents, children, toddlers, infants.
  • a method of treating a condition (e.g., ailment) in a subject may comprise administering to the subject a therapeutically-effective amount of one or more engineered host cell populations, preferably engineered ⁇ T cells, non-engineered cells and/or admixtures thereof.
  • the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof can be administered at various regimens (e.g., timing, concentration, dosage, spacing between treatment, and/or formulation).
  • a subject can also be preconditioned with, for example, chemotherapy, radiation, or a combination of both, prior to receiving the therapeutically-effective amount of one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof.
  • one or multiple engineered host cell populations, non- engineered cells, and/or admixtures thereof may be administered to a subject at a first regimen and the subject may be monitored to determine whether the treatment at the first regimen meets a given level of therapeutic efficacy.
  • the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof may be administered to the subject at a second regimen, based on information gleaned from providing the subject with the first regimen.
  • a pharmaceutical composition comprising at least one host cell engineered to express a CAR of the present disclosure, preferably a ⁇ T cell, may be administered in a first regimen.
  • the subject may be monitored, for example by a healthcare provider (e.g., treating physician or nurse).
  • the subject is monitored to determine or gauge an efficacy of the engineered host cell in treating the condition of the subject.
  • the subject may also be monitored to determine the in vivo expansion of an engineered host cell population in the subject.
  • Another pharmaceutical composition comprising at least one host cell engineered to express a CAR of the present disclosure may be administered to the subject in a second regimen.
  • the pharmaceutical composition administered in the second regimen may comprise a same type of host cell expressing a same CAR as that administered to the subject in the first regimen.
  • the pharmaceutical composition administered in the second regimen may comprise a different type of host cell, optionally expressing a different CAR.
  • the second regimen is not performed, for example if the first regimen is found to be effective (e.g., a single round of administration may Page 128 of 203 1097657169 ⁇ 1 ⁇ AMERICAS be sufficient to treat the condition).
  • a population of engineered host cells can be administered to various subjects (e.g., where the host cell has universal donor characteristics).
  • One or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof, having cytotoxic activity against a B7H6-expressing cell such as a tumor cell can be administered to a subject in any order or simultaneously.
  • the engineered host cell(s), and/or admixtures thereof, of the disclosure can be provided in a single, unified form, such as an intravenous injection, or in multiple forms, for example, as multiple intravenous infusions, s.c, injections or pills.
  • the one or multiple engineered host cell populations, non- engineered cells, and/or admixtures thereof of the disclosure can be packed together or separately, in a single package or in a plurality of packages.
  • One or all of the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof of the disclosure can be given in multiple doses.
  • an engineered host cell preferably an engineered ⁇ T cell, of the disclosure can proliferate within a subject's body, in vivo, after administration to a subject.
  • the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof of the present disclosure can be frozen to provide cells for multiple treatments with the same cell preparation.
  • the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof, and pharmaceutical compositions comprising the same, can be packaged as a kit.
  • a kit may include instructions (e.g., written instructions) on the use of the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof, and compositions comprising the same.
  • the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof, described herein can be administered before, during, or after the occurrence of a disease or condition, and the timing of administering a pharmaceutical composition containing the engineered host cell population can vary.
  • the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof can be used as a prophylactic and can be administered continuously to subjects with a propensity to conditions or diseases in order to lessen a likelihood of the occurrence of the disease or condition.
  • the initial administration can be via any route practical, such as by any route described herein using any formulation described herein.
  • the administration of the one or multiple engineered host cell Page 129 of 203 1097657169 ⁇ 1 ⁇ AMERICAS populations, non-engineered cells, and/or admixtures thereof of the disclosure is an intravenous administration.
  • One or multiple dosages of the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof can be administered as soon as is practicable after the onset of a particular condition (e.g., cancer) and for a length of time necessary for the treatment of the disease/condition, such as, for example, from about 24 hours to about 48 hours, from about 48 hours to about 1 week, from about 1 week to about 2 weeks, from about 2 weeks to about 1 month, from about 1 month to about 3 months.
  • one or multiple dosages of the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof can be administered years after onset of the disease/condition (e.g., cancer) and before or after other treatments.
  • the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof, of the disclosure is administered simultaneously or sequentially with one or more methods to elevate common gamma chain cytokine(s).
  • one or more methods to elevate common gamma chain cytokine(s) refers to a method, or combination of methods, that alters the physiological state of a subject, such that at least one common gamma chain cytokine level is elevated in the subject.
  • the method elevates the level of one or more common gamma chain cytokine(s) selected from the group consisting of IL-2, IL-4, IL-7, IL-15, and IL-21 in the subject.
  • the method comprises lymphodepletion.
  • the method comprises administering one or more common gamma chain cytokine(s) to the subject.
  • IL-2, IL-4, IL-7, IL-15, and/or IL-21 are administered.
  • the method comprises secreting common gamma chain cytokine(s) from an administered engineered host cell.
  • IL-2, IL-4, IL-7, IL-15, and/or IL-21 are secreted.
  • the administering one or more methods to elevate common gamma chain cytokine(s) comprises lymphodepletion before introducing the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof, of the disclosure.
  • the administering one or more methods to elevate common gamma chain cytokine(s) comprises administering simultaneously with introducing the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof, or sequentially an amount of common gamma chain cytokine(s) effective to increase proliferation, cytotoxic activity, persistence, or the combination thereof of the introduced one or multiple engineered host Page 130 of 203 1097657169 ⁇ 1 ⁇ AMERICAS cell populations, non-engineered cells, and/or admixtures thereof.
  • the amount of administered common gamma chain cytokine(s) can be an amount effective to increase proliferation, cytotoxic activity, persistence, or the combination thereof of the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof.
  • Exemplary amounts of IL-15 include, without limitation between 0.01 — 10 ⁇ g/kg/dose every 24 hours for IL-15.
  • Exemplary amounts of IL-2 include, without limitation, between about 3x10 6 and about 22x10 6 units every 8 - 48 hours.
  • the dosing regimen for IL2 in RCC is 600,000 International Units/kg (0.037 mg/kg) IV 48hr infused over 15 minutes for a maximum 14 doses.
  • the administering one or more methods to elevate common gamma chain cytokine(s) comprises lymphodepletion before administering the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof and administering simultaneously with introducing the one or multiple engineered host cell populations, non- engineered cells, and/or admixtures thereof, or sequentially, an amount of common gamma chain cytokine(s) effective to increase proliferation, cytotoxic activity, persistence, or the combination thereof of the introduced one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof.
  • elevating common gamma chain cytokine(s) is accomplished, at least in part, via the engineered host cell(s), where the common gamma chain cytokine(s) are expressed from CAR construct as disclosed herein.
  • the engineered host cell(s) where the common gamma chain cytokine(s) are expressed from CAR construct as disclosed herein.
  • one or more additional gamma chain cytokine(s) are additionally administered in a manner to elevate the additional gamma chain cytokine(s).
  • the ⁇ T cells of the subject invention can also be advantageously administered to a patient in conjunction with (e.g., before, simultaneously or following) any number of relevant treatment modalities (e.g., for treating cancer) including, e.g., chemotherapy, radiation therapy, or immunotherapy.
  • a patient can also be preconditioned with a therapeutically effective amount of ⁇ T cells prior to receiving the chemotherapy, radiation therapy, or immunotherapy (e.g., cell therapy).
  • Suitable immunotherapies for use in combination with ⁇ T cells include autologous and allogeneic cell therapies, engineered T and NK cells, immune engagers, fusion proteins, or other immune-oncology agents.
  • the subject ⁇ T cells can be administered in conjunction with an appropriate cellular immunotherapy for treating a disease such as cancer (e.g. CAR T or CAR NK cells, or Treg therapy).
  • a method of treatment e.g., for treating cancer
  • a conditioning step e.g. a preconditioning step, of administering a therapeutically effective amount of ⁇ T cells to a subject simultaneously or sequentially with administration of a cellular immunotherapy directed against the disease (e.g., cancer).
  • the cellular immunotherapy can comprise further administering an engineered T cell or NK cell therapy comprising a CAR binding to any tumor associated antigen of interest.
  • the subject ⁇ T cells may comprise a dual CAR binding to CD70 and B7H6.
  • the ⁇ T cells of the subject invention may be advantageously administered in conjunction with adoptive cell therapies (ACT) (for reviews of HSCT and adoptive cell therapy approaches, see, Rager & Porter, Ther Adv Hematol (2011) 2(6) 409-428; Roddie & Peggs, Expert Opin. Biol. Ther. (2011) 11(4):473-487; Wang et al. Int. J. Cancer: (2015)136, 1751-1768; and Chang, Y.
  • adoptive cell therapies for reviews of HSCT and adoptive cell therapy approaches, see, Rager & Porter, Ther Adv Hematol (2011) 2(6) 409-428; Roddie & Peggs, Expert Opin. Biol. Ther. (2011) 11(4):473-487; Wang et al
  • Such adoptive cell therapies include, but are not limited to, allogeneic and autologous hematopoietic stem cell transplantation, donor leukocyte (or lymphocyte) infusion (DLI), adoptive transfer of tumor infiltrating lymphocytes, or adoptive transfer of T cells or NK cells (including recombinant cells, e.g., CAR T, CAR NK). Beyond the necessity for donor-derived cells to reconstitute hematopoiesis after radiation and chemotherapy, immunologic reconstitution from transferred cells is important for the elimination of residual tumor cells.
  • the efficacy of ACT as a curative option for malignancies is influenced by a number of factors including the origin, composition and phenotype (lymphocyte subset, activation status) of the donor cells, the underlying disease, the pre-transplant conditioning regimen and post-transplant immune support (e.g., IL-2 therapy) and the graft-versus-tumor (GVT) effect mediated by donor cells within the graft. Additionally, these factors may be balanced against transplant-related mortality, typically arising from the conditioning regimen and/or excessive immune activity of donor cells within the host (i.e., graft-versus-host disease, cytokine release syndrome, etc.). [00449] K.
  • Another embodiment of the invention is an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of disease or condition associated with B7H6- expression.
  • the disease associated with B7H6 expression is selected from a proliferative disease such as a cancer or malignancy or a precancerous condition, or is a non-cancer related indication associated with expression of B7H6.
  • cancerous conditions relevant to the present disclosure include but are not limited to colon cancer, pancreatic cancer, Non-Hodgkin Lymphoma, cervical cancer, hepatic cancer, gastro-intestinal cancer, lung cancer, ovarian cancer, glioblastoma, prostate cancer, breast cancer, ovarian cancer, kidney cancer, bladder cancer, melanoma, and glioma.
  • the article of manufacture comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is effective for treating, preventing and/or diagnosing the condition associated with B7H6 expression and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is an anti-B7H6 antibody or antigen- binding fragment thereof of the invention, or a CAR-modified immune cell of the invention (preferably a CAR-modified ⁇ T cell, or a nucleic acid of the invention.
  • a composition further comprises a carrier, for example a pharmaceutically acceptable carrier.
  • the label or package insert indicates that the composition is used for treating the condition associated with B7H6 expression, for example cancer.
  • the label or package insert will further comprise instructions for administering the antibody composition to the patient.
  • the article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer’s solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • Kits are also provided that are useful for various purposes, e.g., for B7H6- expressing cell killing assays, for purification or immunoprecipitation of B7H6 polypeptide from cells.
  • the kit can contain an anti-B7H6 antibody coupled to beads (e.g., sepharose beads).
  • Kits can be provided which contain the antibodies for detection and quantitation of B7H6 polypeptide in vitro, e.g., in an ELISA or a Page 133 of 203 1097657169 ⁇ 1 ⁇ AMERICAS Western blot.
  • the kit comprises a container and a label or package insert on or associated with the container.
  • the container holds a composition comprising at least one anti-B7H6 antibody or antigen-binding fragment thereof, of the invention.
  • Additional containers may be included that contain, e.g., diluents and buffers, control antibodies.
  • kits can comprise a first container comprising a composition comprising one or more B7H6 antibodies or CAR modified immune cells of the invention; and a second container comprising a buffer.
  • the buffer may be pharmaceutically acceptable.
  • Example 1 Binding profiles of anti-B7H6 antibodies [00456] This Example demonstrates functionality of anti-B7H6 antibodies of the present disclosure. [00457] As shown at FIG.1, synthetic phage display libraries containing fully human antibody sequences were used to pan against recombinant human B7H6 (rhuB7H6) or B7H6+ cell lines.
  • Anti-B7H6 scFv clones derived from phage display panning were reformatted into human IgG antibodies (Abs) and tested using flow cytometry against HeLa cells with B7H6 knock-out (KO) and HeLa cells with B7H6 KO HeLa cells re-engineered to express B7H6 at higher levels (HeLa B7H6 high ).
  • the Ab clones only bound to HeLa B7H6 high cells compared to B7H6 KO HeLa cells.
  • Anti-B7H6 IgG1 Ab clones were evaluated against rhuB7H6 conjugated to MagPlex- C Microspheres.
  • FIG.3A depicted is an illustration of membrane domains of B7H6. Shown is full length B7H6 comprising of V-distal domain and C-proximal domain, V-distal domain with B7H6 stump region, and C-proximal domain with B7H6 stump region.
  • Binding of phage-display derived antibody IgG clones to the different membrane domains of B7H6 measured by flow cytometry are indicative of all antibody IgG clones binding to full-length B7H6 and majority binding to the distal region (FIG.3B).
  • Linear epitope mapping of phage-display derived antibody IgG clones performed using a chemifluorescent-based assay suggests that majority of the antibody clones bind to the membrane distal region (FIG.3C).
  • Each row indicates the IgG clone, and each column indicates an epitope’s sequence.
  • the color gradient for each cell of the heatmap plot represents the relative fluorescence unit, indicative of relative binding to the epitope sequence.
  • Target cell lines HeLa B7H6 KO (Synthego), HeLa, LoVo, Raji, HCT-15, and NCIH716 (ATCC) were stained with anti-B7H6 antibody (Biolegend) or isotype (R&D Systems) and were analyzed by flow cytometry. B7H6 expression in different cell lines was graphed as MFI (mean fluorescence intensity) ratio. The resulting ratio of antibody MFI to isotype MFI was graphed (FIG.4). Based on this data the cell lines could be classified as B7-H6 low expressing: HeLa; medium expressing: Lovo, Raji, HCT-15; and High expressing: NCIH716. [00461] Example 2.
  • phage display technology was used to screen for binders as mentioned above.
  • the screens incorporate recombinant proteins, cancer cell lines with variable target densities, recombinant lines expressing full-length and unique domains of interest (e.g., domains comprising post-translational modifications (PTMs) known to be relevant to a particular cancer type.
  • PTMs post-translational modifications
  • Phage panning using biotinylated recombinant human B7H6 protein (Acro Biosciences) bound to streptavidin beads was performed on highly diverse synthetic scFv phage display libraries (Twist Bio) using 5 rounds of selection, each with decreasing antigen concentration (starting at 100 pmol to 2 pmol) but increasing wash stringency.
  • An alternate panning strategy with alternating rounds of selection with 1e8 B7H6+ cells or antigen 100 pmol Page 135 of 203 1097657169 ⁇ 1 ⁇ AMERICAS and 25 pmol, respectively
  • Antigen-bound phage was pulled down, eluted, amplified and screened using ELISAs for binder confirmation and selection.
  • scFvs were reformatted into IgGs, expressed, purified and characterized.
  • Lead binders were reformatted into human immunoglobulins of appropriate isotype (e.g., IgG) or certain CAR surrogate formats (e.g., CAR diabodies) for further characterization and to assess a wide variety of potential liabilities. This is referred to as a “Molecular Assessment Approach,” which aims to optimize biophysical and chemical properties of molecules in early stages prior to CAR development.
  • Biochemical assays relied upon include but are not limited to ELISA, Western Blotting and/or flow cytometry assays, used to generate relative affinity data (e.g., EC50s) and maximum specific binding (Bmax) for the cohort and benchmark antibodies.
  • Benchmark CAR PL741 in which a B7H6 binding domain is derived from the 47.39 antibody (see WO 2013/169691) as herein described was included as a comparative CAR control.
  • a nucleic acid encodes SEQ ID NO: 337, a PL741 CAR comprising the following domains Page 136 of 203 1097657169 ⁇ 1 ⁇ AMERICAS in order: a signal peptide, a B7H6 binding domain, a CD8 hinge and transmembrane domain, a 4- 1BB costimulatory domain, and a CD3 ⁇ signaling domain.
  • a nucleic acid encoding the PL741 CAR comprises the sequence of SEQ ID NO: 338.
  • Table 13 below provides annotation of the nucleotide sequence of SEQ ID NO: 338. Table 13.
  • PL741 Nucleotide Sequence Annotation Annotation Numbering [00467]
  • specific assays for mapping epitopes bound by the disclosed antibodies and benchmark antibodies were also used, for example, via the use of overlapping peptides spanning the target, in an ELISA format.
  • recombinant cell lines were made expressing full-length target or domains known to have specific epitopes, for screening and binning.
  • hydrogen/deuterium exchange mass spectrometry HDX-MS
  • Lead binders were reformatted into CARs in both heavy-light (HL) and LH (light- heavy) orientations, and these CARs were further screened in Jurkat Lucia NFAT cells to assess CAR cell surface expression, tonic signaling, and activation in the context of target-based stimulation, in the form of recombinant protein or cancer cell lines expressing the B7H6 target.
  • CARs that showed robust activation in Jurkat cells were transduced into V ⁇ 1 ⁇ T cells, and potency against target-expressing cell lines was assessed using a stringent 120 hr in vitro cytotoxicity assay.
  • Example 3 Example 3.
  • Anti-B7H6 scFvs were formatted into CARs, inserted into a gammaretroviral vector, and transduced into Jurkat-Lucia TM NFAT cells (Invivogen).
  • CAR transgene activity in Jurkat- Page 137 of 203 1097657169 ⁇ 1 ⁇ AMERICAS Lucia cells was evaluated in a co-culture assay with HeLa or HeLa B7-H6 KO cells. Supernatants were collected after 18-hrs, was mixed with QUANTI-LucTM reagent (InvivoGen), and Lucia luciferase activity (NFAT activity) was detected using the CytationTM 5 imaging plate reader.
  • B7H6 CAR V ⁇ 1 ⁇ T cells were cocultured with Nuc-NIR expressing target cell lines at an effector to target ratio (E:T) of 1:1 for 120 hours.
  • E:T effector to target ratio
  • Viable Nuc-NIR expressing target cells were quantified every 4 hours by the Incucyte® SX5 (Sartorius). Cytotoxicity index was calculated by dividing the total NIR object area of each time point by the value of time 0 hours.
  • Target cells were modified to express Nuc-NIR by transducing them with Incucyte® Nuclight NIR lentivirus (Sartorius).
  • B7H6 CAR V ⁇ 1 ⁇ T cells showed strong cytotoxic activity against different cancer cell lines such as Raji (Lymphoma) (FIG. 6A), HeLa (cervical) (FIG.6B), and HCT-15 (colon) (FIG.6C).
  • the lower cytotoxicity index indicates better cytotoxic potential of B7H6 CAR V ⁇ 1 ⁇ T cells versus target cells alone (black diamonds).
  • PL741 (Benchmark, see Example 2 above) is a B7H6 CAR previously validated and the lead B7H6 CAR V ⁇ 1 ⁇ T cells outperform it.
  • B7H6 CAR V ⁇ 1 ⁇ T cells were evaluated for polyfunctionality by the Isolight single cell secretome assay (Isoplexis). Cells were stimulated with rhuB7-H6 (AcroBio) for 18 hrs prior to loading CAR T cells onto the IsoCode chip for single-cell multiplex cytokine analysis.
  • Polyfunctional strength index (PSI) (FIG.7A), is defined by the percentage of polyfunctional cells with the intensity of the cytokines measured.
  • Percent polyfunctionality is the percent of cells secreting 2 or more cytokines. Lead B7H6 CAR V ⁇ 1 ⁇ T cells show a higher degree of polyfunctionality as compared to PL741 (Benchmark) B7H6 CAR V ⁇ 1 ⁇ T cells. [00473]
  • Example 4. B7H6 CAR ⁇ T cell Phenotype B7H6 CAR V ⁇ 1 ⁇ T cells were stained for multiple phenotypic markers and were analyzed by flow cytometry. Expression profiles showed a predominant na ⁇ ve-like T cell memory phenotype of the candidate B7H6 CAR + V ⁇ 1 ⁇ T cells (FIG.8A).
  • B7H6 CAR ⁇ T cell Cytotoxic Potential [00478] Cytotoxic potentials of lead B7-H6 CAR V ⁇ 1 ⁇ T cells and Benchmark B7-H6 CAR V ⁇ 1 ⁇ T cells were evaluated against B7H6 + tumor cell line, HCT-15, in a 48-hour Incucyte Immune Cell Killing Assay in the presence and absence of exogenously added soluble B7-H6 (sB7-H6) (AcroBio). Low (FIG.10A) and high (FIG.10B) levels of spiked sB7H6 were used to simulate shed B7H6 levels in serum and tumor microenvironment, respectively.
  • sB7-H6 soluble B7-H6
  • Example 7 T cells were co- cultured with HCT-15 NucNIR-expressing target cells at a suboptimal E:T ratio of 1:1.
  • This Example demonstrates that sB7H6 interfered with the activity of Benchmark B7-H6 CAR V ⁇ 1 ⁇ T but did not impair the activity of the lead B7-H6 CAR V ⁇ 1 ⁇ T cells. [00479] Example 7.
  • B7H6 CAR ⁇ T cells The in vivo efficacy of B7H6 CAR V ⁇ 1 ⁇ T cells was assessed in a subcutaneous colon cancer model using HCT-15 cells.
  • Female NSG mice (The Jackson Laboratory) were implanted subcutaneously with 2 x 10 6 HCT-15 cells (ATCC), mixed with Matrigel (1:1 ratio) (Corning). Animals were randomized into groups of 5 when tumor volume reached an average of 100 mm 3 and were injected IV (tail vein) with 5 x 10 6 or 15 x 10 6 B7H6 CAR V ⁇ 1 ⁇ cells. Negative control group was tumor alone group (no treatment).
  • B7H6 CAR ⁇ T cells with dnTGF ⁇ RII Bolt-on CAR expression is not impacted by the expression of the dnTGF ⁇ RII Bolt-on in B7H6 CAR V ⁇ 1 ⁇ T cells.
  • B7-H6 CAR V ⁇ 1 ⁇ T cells with or without the dnTGF ⁇ RII Bolt-on were stained for TGF ⁇ RII (R&D Systems) and was only detected on the cell surface of B7H6 CAR V ⁇ 1 ⁇ T cells with the Bolt-on using flow cytometry (FIG.12).
  • B7H6 CAR V ⁇ 1 ⁇ T cells with the dnTGF ⁇ RII Bolt-on showed inhibition of pSMAD2/3 levels in the presence of 20ng/mL of TGF- ⁇ 1 (R&D Systems) using intracellular flow cytometry, confirming the functionality of the dnTGF ⁇ RII by inhibiting TGF- ⁇ 1 signaling.
  • B7H6 CAR V ⁇ 1 ⁇ T cells without the dnTGF ⁇ RII Bolt-on and untransduced V ⁇ 1 ⁇ T cells showed an increase in pSMAD2/3 after exposure to TGF- ⁇ 1 compared to no TGF- ⁇ 1 treatment (FIG.13).
  • Example 9 Gene editing of ⁇ T cells [00486] To knockout the CISH gene in V ⁇ 1 T cells, a ribonucleoprotein (RNP) complex comprised of CRISPR/Cas9 (Integrated DNA Technologies) and CISH sgRNA’s (Synthego Corporation) were combined with activated V ⁇ 1 T cells from PBMC’s (P3 Primary Cell 4D- Nucleofector X Kit L, Lonza).
  • RNP ribonucleoprotein
  • the RNP complex was delivered into V ⁇ 1 T cells by electroporation (Lonza 4D-Nucleofector). Post electroporation, the cells were transferred to T flasks (Corning) containing X-Vivo15 (Lonza) for cell recovery and expansion in a 37°C CO2 incubator (ThermoFisher).
  • T flasks Corning
  • X-Vivo15 Lithza
  • CISH KO cell pellets were collected following ⁇ T cell depletion (StemCell Technologies) on day 7 post gene edit and genomic DNA was extracted using Page 140 of 203 1097657169 ⁇ 1 ⁇ AMERICAS the NucleoSpin Tissue kit (MACHEREY-NAGEL).
  • the edited region of the CISH gene was amplified by PCR and Sanger sequenced (Sequetech).
  • Example 10 CISH KO enhances the function of V ⁇ 1 T cells [00489] Healthy donor peripheral blood mononuclear cells (PBMCs) were used to activate, expand, CRISPR-gene edit, and engineer V ⁇ 1 ⁇ T cells to express a CAR.
  • PBMCs peripheral blood mononuclear cells
  • a ribonucleoprotein (RNP) complex comprising CRISPR-Cas9 (Integrated DNA Technologies) or -MAD7 (Aldevron) and CISH sgRNA were combined with activated V ⁇ 1 T cells from PBMCs (P3 Primary Cell 4D-Nucleofector X Kit L, Lonza).
  • the RNP complex was delivered into V ⁇ 1 T cells by electroporation (Lonza 4D-Nucleofector). Post electroporation, the cells were transferred to T flasks (Corning) containing X-Vivo15 (Lonza) for cell recovery prior to transduction with gammaretroviral vector encoding the CAR and then were subsequently expanded.
  • CISH KO efficiency To evaluate CISH KO efficiency, cell pellets were collected, and genomic DNA was extracted using the NucleoSpin Tissue kit (MACHEREY-NAGEL). The edited region of the CISH gene was amplified by PCR and Sanger sequenced (Sequetech). Gene knockout efficiency was determined using the ICE Analysis tool (Synthego).
  • WT unedited
  • CISH KO CAR V ⁇ 1 T cells were incubated with and without IL- 2 (Peprotech) for 5 hours. Cell lysates were generated for western blot detection of CISH protein using anti-CISH Rabbit mAb (Cell Signaling).
  • FIGS. 17A- 17B An IRDye 800CW goat anti-Rabbit IgG secondary antibody (LI-COR) was used for detection of the primary antibody and the presence of the IRDye was imaged with the Odyssey imaging system (LI-COR). The results are shown in FIGS. 17A- 17B.
  • FIGS. 17A- 17B The results are shown in FIGS. 17A- 17B.
  • FIG. 17A WT CAR V ⁇ 1 T cells showed an increase in levels of CISH protein when Page 141 of 203 1097657169 ⁇ 1 ⁇ AMERICAS exposed to IL-2.
  • CISH KO CAR V ⁇ 1 T cells had substantially reduced levels of CISH protein in the presence of IL-2 supporting the KO of CISH.
  • FIG.17B To demonstrate enhanced function of CISH KO CAR-transduced and untransduced V ⁇ 1 T cells, the cytolytic activity against the B7-H6+ HCT-15 cell line (ATCC) was evaluated using an in vitro repetitive stimulation assay.
  • HCT-15 NIR fluorescently labeled cell line and effector cells were added into a 96-well plate at an E:T ratio of 5:1 with or without different concentrations of IL-2. Each stimulation was monitored for 3 days after which fresh target cells were plated in another 96-well plate. The effector cells from each stimulation were then transferred into the plate with fresh target cells. The co-cultures were monitored every 4 hours using Incucyte® SX5 (Satorius).
  • the Cytotoxicity Index was calculated based on the remaining viable target tumor cells by dividing the total NIR object area of each time point by the value at time 0 for each stimulation. The lower the cytotoxicity index, the better the cytotoxic potential.
  • CISH KO untransduced V ⁇ 1 T cells were associated with greater tumor cell killing compared to WT untransduced V ⁇ 1 T cells only in the presence of IL-2. Without IL-2, CISH KO CAR V ⁇ 1 T cells had superior tumor control compared to WT CAR V ⁇ 1 T cells. The results suggest CISH KO enhances the in vitro cytotoxicity of V ⁇ 1 T cells.
  • CAS9 CISH sgRNA TGTACAGCAGTGGCTGGTGG (AGG) (SEQ ID NO: 339)
  • MAD7 CISH sgRNA (TTTA) GGTGTACAGCAGTGGCTGGTG (SEQ ID NO: 340).
  • Example 11 Enhancement of CAR V ⁇ 1 T cells effector function using membrane bound IL-12 (mbIL-12)
  • CAR vector consisting of the anti-CD19 (clone FMC63) scFv (as described in Kochendefer JN et al., Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells, Blood.2010 Nov 11;116(19):3875-86), CD8 ⁇ hinge and transmembrane domains, and 4-1BB and CD3 ⁇ signaling domains followed by a self-cleaving P2A sequence that is used to separate the mbIL-12 molecule (as described in Lee et al., Antigen-dependent IL-12 signaling in CAR T cells promotes regional to systemic disease targeting, bioRxiv.2023 Jan 7;2023.01.06.522784), was constructed by cloning a DNA fragment containing all
  • FIG. 18 The Page 142 of 203 1097657169 ⁇ 1 ⁇ AMERICAS constructs design is shown in FIG. 18 (construct A) CAR (FMC63) construct, (construct B) CAR-mbIL-12 construct, and (construct C) mbIL-12 construct.
  • PBMCs peripheral blood mononuclear cells
  • FIGS. 19A-19D Healthy donor peripheral blood mononuclear cells (PBMCs) were used to activate, expand and engineer V ⁇ 1 ⁇ T cells to express the CD19 CAR-mbIL-12 transgene. The results are shown in FIGS. 19A-19D.
  • FIGS. 19A V ⁇ 1 ⁇ T cells (post-transduction) were sampled at different time points over the course of the expansion to determine cell counts.
  • V ⁇ 1 ⁇ T cell counts did not differ between CAR and CAR-mbIL-12, suggesting mbIL-12 has no negative impact on the expansion of V ⁇ 1 ⁇ T cells.
  • Cells were harvested, stained for CAR and mbIL-12 using anti-FMC63 antibody conjugated to PE (Acrobio Systems, Newark, DE) and anti-IL12 conjugated to APC (Miltenyi Biotec) respectively, and acquired on the Novocyte flow cytometer (Agilent Technologies) to determine the percentage of CAR and mbIL-12 positive V ⁇ 1 ⁇ T cells.
  • FIG.19B No difference in % CAR was observed between V ⁇ 1 ⁇ T cells expressing CAR and CAR-mbIL-12 at the end of the process to further support mbIL-12 did not affect CAR surface expression.
  • FIG. 19C Over the course of the expansion, the % mbIL-12 expressed on the cell surface of V ⁇ 1 ⁇ T cells decreased over time.
  • FIG.19D CAR-mbIL-12 V ⁇ 1 ⁇ T cells were co- cultured with and without CD19+ Raji cells for 18 hrs. After stimulation with target cells, mbIL- 12 increased on the cell surface of the V ⁇ 1 ⁇ T cells compared to unstimulated conditions suggesting the surface expression of mbIL-12 is dependent on CAR activation.
  • CAR-mbIL-12 cytolytic activity against CD19- positive cell lines using CAR, CAR-mbIL-12, and mbIL-12 transduced V ⁇ 1 ⁇ T cells was evaluated in an in vitro repetitive stimulation assay.
  • Raji-Nuc NIR fluorescently labeled cell line and effector cells were added into a 96-well plate at E:T ratios of 5:1, 2.5:1, and 1.25:1.
  • the first stimulation was monitored for 3 days after which fresh target cells were plated in another 96-well plate.
  • the effector cells from the first stimulation were then transferred into the plate with fresh target cells.
  • the second stimulation was monitored for an additional 3 days, and the same procedure was performed for a third stimulation.
  • the co-cultures were monitored every 4 hours using Incucyte® SX5 (Satorius).
  • the Cytotoxicity Index was calculated based on the remaining viable target tumor cells by dividing the total NIR object area of each time point by the value at time 0 for each stimulation. The lower the cytotoxicity index, the better the cytotoxic potential.
  • the CAR-mbIL-12 V ⁇ 1 ⁇ T cells were associated with greater tumor cell killing compared to CAR and mbIL12 V ⁇ 1 ⁇ T cells across the E:T ratios Page 143 of 203 1097657169 ⁇ 1 ⁇ AMERICAS tested.
  • Co-culture conditions with a Cytotoxicity Index > 1 (indication of incomplete tumor control), were not analyzed in the subsequent stimulation.
  • CAR-mbIL-12 V ⁇ 1 ⁇ T cells were evaluated in Raji cell subcutaneous xenograft tumor model at a suboptimal dose of 2.5e6 CAR+ viable cells.
  • V ⁇ 1 ⁇ T cells expressing CAR and CAR-mbIL-12 were administered on Day 0 as a single bolus dose.
  • a ribonucleoprotein (RNP) complex comprising CRISPR-Cas9 (Integrated DNA Technologies) and CBL-B sgRNAs were combined with activated V ⁇ 1 T cells from PBMCs (P3 Primary Cell 4D-Nucleofector X Kit L, Lonza).
  • the RNP complex was delivered into V ⁇ 1 T cells by electroporation (Lonza 4D-Nucleofector). Post electroporation, the cells were transferred to T flasks (Corning) containing X-Vivo15 (Lonza) for cell recovery prior to transduction with gammaretroviral vector encoding the CAR and then were subsequently expanded.
  • FIGS.22A-22B The results are shown in FIGS.22A-22B.
  • FIG. 22A To determine CBL-B KO efficiency, genomic DNA was extracted from cell pellets using the NucleoSpin Tissue kit (MACHARY-NAGEL). The edited region was amplified by PCR using previously in-house optimized primers. The PCR reaction was run on an agarose gel, and the prominent band excised, and gel extracted using NucleoSpin Gel and PCR Cleanup kit (MACHARY-NAGEL). The extracted DNA was sequenced by Sanger sequencing (Sequetech). % Indels were determined using the ICE Analysis tool (Synthego). (FIG.
  • Cas9 Multi-guide sgRNA sequences [00499] AAGACUCUUUAAAGAAGGCA (SEQ ID NO: 341) [00500] AGUACUCAUUCUCACUGAGU (SEQ ID NO: 342) [00501] CGUAAAUGCUGAUAUGUAUC (SEQ ID NO: 343) [00502] Cas9 Single-guide: TAATCTGGTGGACCTCATGA (AGG) (SEQ ID NO: 344).
  • Example 13 Roquin-1 KO enhances the function of V ⁇ 1 T cells
  • Healthy donor peripheral blood mononuclear cells PBMCs
  • PBMCs peripheral blood mononuclear cells
  • CRISPR-gene edit a CRISPR-gene edit
  • V ⁇ 1 ⁇ T cells a CAR-specific ribonucleoprotein (RNP) complex
  • RNP ribonucleoprotein
  • CRISPR-Cas9 Integrated DNA Technologies
  • Roquin-1 sgRNAs were combined with activated V ⁇ 1 T cells from PBMCs (P3 Primary Cell 4D-Nucleofector X Kit L, Lonza).
  • the RNP complex was delivered into V ⁇ 1 T cells by electroporation (Lonza 4D-Nucleofector). Post electroporation, the cells were transferred to T flasks (Corning) containing X-Vivo15 (Lonza) for cell recovery prior to transduction with gammaretroviral vector encoding the CAR and then were subsequently expanded. The results are shown in FIGS. 23A-23B. (FIG.23A). To determine the Roquin KO efficiency genomic DNA was extracted from cell pellets using the NucleoSpin Tissue kit (MACHARY-NAGEL). The edited region was amplified by PCR using previously in-house optimized primers.
  • the Cytotoxicity Index was calculated based on the remaining viable target tumor cells by dividing the total NIR object area of each time point by the value at time 0 for each stimulation.
  • Multi-guide sgRNA sequences [00506] AAGCCCAUCAGUUUGGGUUG (SEQ ID NO: 345) [00507] UGUACAAGCUCCACAAUGGA (SEQ ID NO: 346) [00508] CAAAUGGGCAAGCCUUGCGG (SEQ ID NO: 347) [00509] In-house optimized single gRNA sequence: CCTGAATAAACTCCACCGCA (AGG) (SEQ ID NO: 348).
  • Example 14 ICAM-1 and CD58 KO enhances the in vitro cell survival of CAR V ⁇ 1 T cells in the presence of allogeneic PBMCs
  • PBMCs Healthy donor peripheral blood mononuclear cells
  • a ribonucleoprotein (RNP) complex comprised of CRISPR-Cas9 (Integrated DNA Technologies) or -MAD7 (Aldevron) and ICAM-1 or CD58 sgRNA were combined with activated V ⁇ 1 T cells from PBMCs (P3 Primary Cell 4D-Nucleofector X Kit L, Lonza).
  • the RNP complex was delivered into V ⁇ 1 T cells by electroporation (Lonza 4D-Nucleofector).
  • the cells were transferred to T flasks (Corning) containing X-Vivo15 (Lonza) for cell recovery prior to transduction with gammaretroviral vector encoding the CAR and then were Page 146 of 203 1097657169 ⁇ 1 ⁇ AMERICAS subsequently expanded.
  • WT unedited
  • ICAM-1 KO ICAM-1 KO
  • CD58 KO CAR V ⁇ 1 ⁇ T cells were stained with anti-ICAM-1, anti-CD58, or isotype control antibody conjugated to PE (BioLegend) and acquired using the Novocyte flow cytometer (Agilent Technologies).
  • FIGS.24A-24B The results are shown in FIGS.24A-24B.
  • FIG.24A Greater than 80% KO efficiency was observed for both ICAM-1 and CD58 KO in CAR V ⁇ 1 ⁇ T cells.
  • FIG.24B To evaluate if the of ICAM-1 and CD58 can enhance the survival of CAR V ⁇ 1 T cells in the presence of allogeneic PBMCs, a 10-day mixed-lymphocyte reaction assay (MLR) was performed. WT (unedited), CISH KO, ICAM-1 KO, and CD58 KO CAR V ⁇ 1 ⁇ T cells (target cells) were co- cultured with allogeneic PBMCs (effector cells) at an E:T ratio of 10:1 in the presence of IL-2.
  • MLR mixed-lymphocyte reaction assay
  • the absolute count of WT CAR+ V ⁇ 1+ ⁇ T cells was compared to the KO CAR+ V ⁇ 1+ ⁇ T cell count conditions using the formula: [(WT CAR+ V ⁇ 1+ ⁇ T cells count – KO CAR+ V ⁇ 1+ ⁇ T cell count) ⁇ (WT CAR+ V ⁇ 1+ ⁇ T cells count)] x 100.
  • CAS9 ICAM-1 sgRNA AAAGUCAUCCUGCCCCGGGGGG (SEQ ID NO: 349)
  • MAD7 ICAM-1 sgRNA TTTG/AATAGCACATTGGTTGGCTAT (SEQ ID NO: 350)
  • CAS9 CD58 sgRNA AAAUAUAUGGUGUUGUGUAU (SEQ ID NO: 351)
  • MAD7 CD58 sgRNA TTTA/GACACTGTGTCAGGTAGCCTC (SEQ ID NO: 352).
  • the embodiments and examples described above are intended to be merely illustrative and non-limiting.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

Aspects of the disclosure include affinity-binding entities that target B7 homolog 6 (B7H6), chimeric antigen receptors (CARs) comprising the same, modified immune cells comprising said CARs, and compositions and methods comprising the same for the treatment of conditions associated with B7H6-expression. In embodiments, the modified immune cells are engineered γδ T cells.

Description

AFFINITY BINDING ENTITIES DIRECTED TO B7H6 AND METHODS OF USE THEREOF CROSS-REFERENCE TO RELATED APPLICATION [0001] This application claims the benefit of U.S. Provisional Application No.63/411,988, filed September 30, 2022, the disclosure of which is expressly incorporated by reference herein in its entirety. FIELD OF DISCLOSURE [0002] The present disclosure relates generally to affinity binding entities directed to B7 homolog 6 (B7H6). Chimeric antigen receptors (CARs) capable of binding B7H6, polynucleotides, host cells comprising the polynucleotides and/or CARs, and methods of treating disorders associated with B7H6 in a patient are provided. BACKGROUND OF THE DISCLOSURE [0003] Adoptive cellular therapy has undergone near constant iteration for more than thirty (30) years, from early days focusing on basic lymphokine activation and/or tumor infiltration to more recent strategies engineering immune cells to express genetically engineered antigen receptors, such as chimeric antigen receptors (CARs). While there have been some hints and indications of the curative potential of these approaches along the way, much still remains to be done. In particular, successful tumor eradication by CAR-T lymphocytes depends on CAR-T cell persistence and effector function, but an excess of either can trigger graft-versus-host (GvH) effects in the patient. Additionally, while the adoptive transfer of T cells expressing CARs has shown some success in treating haematological malignancies, only limited efficacy has been shown in other cancer types, particularly solid tumors. Compared with haematological diseases, solid tumors present unique challenges, including but not limited to highly immunosuppressive and metabolically challenging tumor microenvironments. [0004] B7 homolog 6 (B7H6), also known as NCR3LG1 (natural killer cell cytotoxicity receptor 3 ligand 1), is human-specific B7 family member that is a ligand of NKp30. NKp30, a Page 1 of 203 1 D0o9c7u6m5e7n1t629\1\AMERICAS member of the CD28 family, is an activating receptor of natural killer (NK) cells. B7H6 is typically not expressed on normal human tissues, but is expressed in many human tumor cell lines including melanoma, carcinomas, T and B lymphomas, and myeloid leukemias, as well as primary tumor blood and bone marrow cells (Brandt et al., J Exp Med.2009; 206: 1495-1503; Cao et al., J Biol Chem.2015; 290: 29964-29973). Aside from its expression in cancer, B7H6 is upregulated under inflammatory conditions such as atopic dermatitis (Salimi et al., J Immunol. 2016; 196: 45-54). The wide expression profile on tumors and the lack of expression on healthy tissues make B7H6 an attractive target for immunotherapeutic approaches. [0005] To date, B7H6-targeted CARs have been examined in the context of αβ T cells, which are known to be associated with high alloreactive potential in general, as well as a propensity to trigger complications such as cytokine release syndrome (CRS) and macrophage activation syndrome (MAS). [0006] Gamma delta (γδ) T cells are thymus-derived lymphocytes that differ from αβ T cells in their mechanism of activation and function, as well as in their anatomical distribution. In particular, while αβ T cells function exclusively in adaptive immunity, γδ T cells are innate-like immune cells that recognize malignant cells through a repertoire of activating receptors in a MHC- independent manner, similar to NK cells (Welsh et al., Immunol Rev.1997; 159: 79-93). As such, and in contrast to αβ T cells, γδ T cells can potentially be used in an allogeneic setting without the risk of causing graft versus host disease (GvHD). Moreover, recent studies have suggested that engineered γδ-T cells may produce less proinflammatory cytokines than αβ-T cells, which could thereby reduce the risk of CRS in patients (Harrer et al., BMC Cancer 2017; 17(1): 551). [0007] Despite rapid and intense development of CAR T therapies, optimal parameters for CAR T cell-target interactions that will result in in vivo and in-human efficacy are not yet well understood. Given the differences in mechanism of action and function for αβ T cells as compared to γδ T cells, demonstration of CAR function and effectiveness in the context of αβ T cells is not predictive of CAR function and effectiveness in the context of γδ T cells. Hence, the practical translation of B7H6-targeted CAR T therapeutic approaches to γδ T cells is at best uncertain. [0008] Accordingly, improved strategies are still clearly needed to improve the activity, survival, and/or expansion of the cells upon administration, while concurrently improving the safety of adoptive cellular therapeutic approaches targeting B7H6. Page 2 of 203 1097657169\1\AMERICAS SUMMARY OF DISCLOSURE [0009] The present disclosure provides affinity binding entities directed to B7H6, CARs capable of binding B7H6, polynucleotides, host cells comprising the polynucleotides and/or CARs, and methods of treating disorders associated with B7H6 in a patient. [0010] In one aspect, an affinity binding entity comprises an antigen binding domain that specifically binds to B7 homolog 6 (B7H6). In embodiments, a heavy chain variable region/light chain variable region (HCVR/LCVR) sequence pair selected from the group consisting of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, 15/16, 17/18, 19/20, 21/22, 23/24, 25/26, 27/28, 29/30, 31/32, 33/34, 35/36, 37/38, and 39/40; or the six CDRs of a HCVR/LCVR sequence pair selected from the group consisting of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, 15/16, 17/18, 19/20, 21/22, 23/24, 25/26, 27/28, 29/30, 31/32, 33/34, 35/36, 37/38, and 39/40. The amino acid sequences of the CDRs are shown in Tables 16 and 17. [0011] In embodiments, the antigen binding domain comprises a HCVR/LCVR sequence pair selected from the group consisting of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, and 15/16; or the six CDRs of a HCVR/LCVR sequence pair selected from the group consisting of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, and 15/16. In embodiments, the antigen binding domain comprises a HCVR/LCVR sequence pair of SEQ ID NOs: 1/2; or the six CDRs of a HCVR/LCVR sequence pair of SEQ ID NOs: 1/2. In embodiments, antigen binding domain comprises a HCVR/LCVR sequence pair of SEQ ID NOs: 3/4; or the six CDRs of a HCVR/LCVR sequence pair of SEQ ID NOs: 3/4. [0012] In embodiments, the affinity binding entity is an antibody, or an antibody fragment. In embodiments, the antibody or antibody fragment is bispecific. In embodiments, the antibody or antigen binding fragment is chimeric, humanized, or human. In embodiments, the antibody or antibody fragment is monoclonal. In embodiments, the affinity binding entity is selected from the group consisting of scFv, Fab, Fab’, Fv, F(ab’)2, dsFv, dAb, and any combination or plurality thereof. [0013] In an aspect, a chimeric antigen receptor (CAR) comprises any of the afore-mentioned affinity binding entities as disclosed herein. In embodiments, the CAR comprises a hinge domain. In embodiments, the hinge domain comprises a glycine polymer, glycine-serine polymer, glycine- alanine polymer, alanine-serine polymer, immunoglobulin heavy chain hinge, or receptor-derived Page 3 of 203 1097657169\1\AMERICAS hinge. In embodiments, the receptor-derived hinge is a CD8 alpha hinge domain. In embodiments, the CD8 alpha hinge domain comprises an amino acid sequence set forth as SEQ ID NO: 209. In embodiments, the CAR comprises a transmembrane (TM) domain. In embodiments, the TM domain comprises a TM region of 4-1BB/CD137, activating NK cell receptors, an Immunoglobulin protein, B7-H3, BAFFR, BLAME (SLAMF8), BTLA, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, or CD154, CD100 (SEMA4D), CD103, CD160 (BY55), CD18, CD19, CD19a, CD2, CD247, CD27, CD276 (B7-H3), CD28, CD29, CD3 delta, CD3 epsilon, CD3 gamma, CD3 zeta, CD30, CD4, CD40, CD49a, CD49D, CD49f, CD69, CD7, CD84, CD8, CD8alpha, CD8beta, CD96 (Tactile), CD11a, CD11b, CD11c, CD11d, CDS, CEACAM1, CRT AM, cytokine receptor, DAP10, DNAM1 (CD226), Fc gamma receptor, GADS, GITR, HVEM (LIGHTR), IA4, ICAM-1, Ig alpha (CD79a), IL-2R beta, IL-2R gamma, IL-7R alpha, inducible T cell costimulator (ICOS), integrins, ITGA4, ITGA6, ITGAD, ITGAE, ITGAL, ITGAM, ITGAX, ITGB2, ITGB7, ITGBl, KIRDS2, LAT, LFA-1, a ligand that specifically binds with CD83, LIGHT, LTBR, Ly9 (CD229), lymphocyte function-associated antigen-1 (LFA-1; CD11a/CD18), MHC class 1 molecule, NKG2C, NKG2D, NKp30, NKp44, NKp46, NKp80 (KLRF1), OX-40, PAG/Cbp, programmed death-1 (PD-1), PSGL1, SELPLG (CD162), Signaling Lymphocytic Activation Molecules (SLAM proteins), SLAM (SLAMF1; CD150; IPO-3), SLAMF4 (CD244; 2B4), SLAMF6 (NTB-A; Lyl08), SLAMF7, SLP-76, TNF receptor proteins, TNFR2, TNFSF14, a Toll ligand receptor, TRANCE/RANKL, VLA1, or VLA-6, or a fragment, truncation, or a combination thereof. In embodiments, the TM domain comprises a TM domain of CD8, preferably wherein the CD8 TM domain is a TM domain of CD8 alpha. In embodiments, the TM domain comprises the amino acid sequence set forth as SEQ ID NO: 211. [0014] In embodiments, the CAR further comprises at least one costimulatory domain. In embodiments, the costimulatory domain comprises a costimulatory domain of TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, CARD11, B7-H3, CEACAM1, CRTAM, CD2, CD3C, CD4, CD7, CD8α, CD8β, CD11a, CD11b, CD11c, CD11d, IL2Rβ, IL2γ, IL7Rα, IL4R, IL7R, IL15R, IL21R, CD18, CD19, CD19a, CD27, CD28, CD29, CD30, CD40, CDS, CD49a, CD49D, CD49f, CD54 (ICAM), CD69, CD70, CD80, CD83, CD84, CD86, CD96 (Tactile), CD100 (SEMA4D), CD103, CD134 (OX40), CD137 (4-1BB), CD152 (CTLA-4), CD160 (BY55), CD162 (SELPLG), CD244 (2B4), CD270 (HVEM), CD226 (DNAM1), CD229 Page 4 of 203 1097657169\1\AMERICAS (Ly9), CD278 (ICOS), ICAM-1, LFA-1 (CD11a/CD18), FcR, FcγRI, FcγRII, FcγRIII, LAT, NKG2C, SLP76, TRIM, ZAP70, GITR, BAFFR, LTBR, LAT, GADS, LIGHT, HVEM (LIGHTR), KIRDS2, ITGA4, ITGA6, ITGAD, ITGAE, ITGAL, ITGAM, ITGAX, ITGB1, ITGB2, ITGB7, NKG2C, NKG2D, IA4, VLA-1, VLA-6, SLAM (SLAMF1, CD150, IPO-3), SLAMF4, SLAMF6 (NTB-A, Ly108), SLAMF7, SLAMF8 (BLAME), SLP-76, PAG/Cbp, NKp80 (KLRF1), NKp44, NKp30, NKp46, BTLA, JAML, CD150, PSGL1, TSLP, TNFR2, and TRANCE/RANKL, or a portion thereof, and combinations thereof. In embodiments, the costimulatory domain is a 4-1BB costimulatory domain. In embodiments, the 4-1BB costimulatory domain comprises an amino acid sequence set forth as SEQ ID NO: 202. [0015] In embodiments, the CAR further comprises one or more intracellular signaling domains. In embodiments, the intracellular signaling domain is a CD3ζ intracellular signaling domain. In embodiments, the CD3ζ intracellular signaling domain comprises an amino acid sequence set forth as SEQ ID NO: 229, 231, or 232. [0016] In embodiments, the CAR further comprises a signal peptide. In embodiments, the signal peptide comprises an amino acid sequence set forth as SEQ ID NO: 170. [0017] According to an aspect, provided is an isolated polynucleotide comprising a nucleic acid sequence encoding an affinity binding entity as herein disclosed. In embodiments, an expression vector comprises the isolated polynucleotide. In embodiments, the expression vector is operably linked to a cis-acting regulatory element. In embodiments, a cell comprises the affinity binding entity, the isolated polynucleotide, and/or the expression vector. [0018] According to an aspect, provided is an isolated polynucleotide comprising a nucleic acid sequence encoding a CAR as herein disclosed. In embodiments, the polynucleotide comprises a nucleic acid sequence encoding at least one multicistronic linker region. In embodiments, the multicistronic linker region encodes a cleavage sequence. In embodiments, the cleavage sequence is selected from T2A, F2A, P2A, E2A, furin, and furin-P2A (FP2A). In embodiments, the multicistronic linker region encodes an internal ribosomal entry site (IRES). [0019] In embodiments, the polynucleotide further comprises a nucleic acid encoding one or more additional polypeptides. In embodiments, the one or more additional polypeptides is selected from the group comprising or consisting of lymphotoxin beta receptor (LTBR), low- Page 5 of 203 1097657169\1\AMERICAS affinity nerve growth factor receptor (LNGFR), a dominant negative (dn) receptor for TGF-beta or Fas, a truncated form of the human epidermal growth factor receptor (EGFRt), and membrane- bound IL-12 (mbIL-12), or any combination thereof. In embodiments, the one or more additional polypeptides is selected from a fluorescent protein, a gamma chain cytokine, CD19, CD20, LNGFR, EGFRt, LTBR, dnTGFβR2, dominant negative Fas, membrane-bound IL-12, CAR that binds to CD70, or any combination thereof. [0020] In embodiments, the one or more additional polypeptides is a dominant negative receptor for TGFβ. In embodiments, the dominant negative receptor for TGFβ is dnTGFβR2, optionally wherein the dnTGFβR2 comprises an amino acid sequence set forth as SEQ ID NO: 241. In embodiments, the one or more additional polypeptides is lymphotoxin beta receptor (LTBR), optionally wherein the LTBR comprises an amino acid sequence set forth as SEQ ID NO: 245. In embodiments, the one or more additional polypeptides is a truncated form of the epidermal growth factor receptor (EGFRt), optionally wherein the EGFRt comprises an amino acid sequence set forth as SEQ ID NO: 237. In embodiments, the one or more additional polypeptides is low- affinity nerve growth factor receptor (LNGFR), optionally wherein the LNGFR comprises an amino acid sequence set forth as SEQ ID NO: 249. In embodiments, the one or more additional polypeptides is selected from the group comprising or consisting of lymphotoxin beta receptor (LTBR), a dominant negative (dn) receptor for TGF-beta or Fas, a truncated form of the human epidermal growth factor receptor (EGFRt), and membrane-bound IL-12 (mbIL-12). In embodiments, the one or more additional polypeptides is a dominant negative Fas (dnFas). In embodiments, the one or more additional polypeptides is membrane-bound IL-12 (mbIL-12). In embodiments, the one or more additional polypeptides is a CAR that binds to CD70. [0021] In embodiments, the one or more additional polypeptides are operably linked to a nucleic acid sequence encoding a signal peptide. In embodiments, the signal peptide comprises an amino acid sequence selected from SEQ ID NO: 235, SEQ ID NO: 239, SEQ ID NO: 243, SEQ ID NO: 247, and SEQ ID NO: 196. [0022] In embodiments, the polynucleotide comprising a nucleic acid encoding a CAR as herein disclosed has a nucleic acid sequence selected from SEQ ID NO: 294, 298, 302, 306, 310, 314, 318, 322, 326, 330, or 334. Page 6 of 203 1097657169\1\AMERICAS [0023] In embodiments, provided is an expression vector comprising the polynucleotide encoding a CAR of the present disclosure, operably linked to a cis-acting regulatory element. [0024] According to an aspect, provided herein is a γδ T cell comprising (a) a nucleic acid encoding a chimeric antigen receptor (CAR), the CAR comprising an antigen binding domain that specifically binds to B7 homolog 6 (B7H6); and/or (b) a polypeptide comprising a CAR comprising an amino acid sequence encoded by the nucleic acid of (a), wherein the γδ T cell functionally expresses the binding domain of the polypeptide or nucleic acid encoded CAR on the surface of the γδ T cell. [0025] In embodiments, the γδ T cell comprises a CAR comprising an affinity binding entity as herein disclosed. In embodiments, the nucleic acid comprises a polynucleotide encoding a CAR as herein disclosed, or an expression vector encoding a CAR as herein disclosed. [0026] According to an aspect, provided is a modified immune cell. In embodiments, the modified immune cell comprises a CAR as herein disclosed. In embodiments, the modified immune cell comprises a polynucleotide comprising a nucleic acid sequence encoding a CAR as herein disclosed. In embodiments, the modified immune cell comprises an expression vector that encodes for a CAR as herein disclosed. [0027] In embodiments, the modified immune cell is a ^ ^ ^T cell, a ^ ^ ^ ^ ^ ^ ^cell, an αβ T cell, a NK cell, a NKT cell, or a macrophage. In embodiments, the modified immune cell is a ^ ^ ^T cell. In embodiments, the ^ ^ ^T cell is a δ1, a δ2, a δ3, or a δ4 γδ T cell, preferably a δ2- γδ T cell, more preferably a δ1 γδ T cell. [0028] In embodiments, the modified immune cell or ^ ^ ^T cell exhibits in vitro and/or in vivo cell killing activity against a tumor cell that exhibits cell surface expression of B7H6. In embodiments, the cell killing activity of the modified immune cell, or the γδ T cell, is greater than an innate level of in vitro and/or in vivo tumor cell killing activity in a control modified immune cell or control γδ T cell of the same type that does not comprise a CAR construct. [0029] In embodiments, the modified immune cell or γδ T cell proliferates in response to contact with the tumor cell that exhibits cell surface expression of B7H6. In embodiments, the modified immune cell or γδ T cell exhibits increased proliferation in response to contact with the Page 7 of 203 1097657169\1\AMERICAS tumor cell that exhibits cell surface expression of B7H6 as compared to a control immune cell or γδ T cell of the same type that does not comprise a CAR construct. [0030] In embodiments, the modified immune cell or γδ T cell proliferates in a host organism that comprises a tumor cell that exhibits cell surface expression of B7H6. In embodiments, the modified immune cell or the γδ T cell expresses pro-inflammatory cytokines after contact with a tumor cell that exhibits cell surface expression of B7H6. [0031] In embodiments, the modified immune cell or γδ T cell comprises at least one disrupted gene. In embodiments, the disrupted gene is cytokine inducible SH2-containing protein (CISH). In embodiments, the at least one disrupted endogenous gene is Cbl proto-oncogene B (CBL-B). In embodiments, the at least one disrupted endogenous gene is Zinc Finger Protein 91 (ZFP91). In embodiments, the at least one disrupted endogenous gene is Roquin. In embodiments, the at least one disrupted endogenous gene is CD58 and/or ICAM-1. [0032] According to an aspect, provided herein is a plurality of modified immune cells, or a plurality of modified γδ T cells, as herein disclosed. In embodiments, the plurality of modified immune cells, or the plurality of γδ T cells, comprise a composition that is at least 60%, 80%, or from about 60% or 80% to about 90% or 95% δ1, δ2, δ3, or δ4 γδ T cells, preferably δ1 or δ2 γδ T cells, more preferably δ2- γδ T cells, most preferably δ1 γδ T cells. In embodiments, the plurality of modified immune cells, or the plurality of γδ T cells comprises at least about 107 modified immune cells or γδ T cells, respectively, preferably from about 108 modified immune cells or γδ T cells to about 1011 modified immune cells or γδ T cells, respectively. [0033] According to an aspect, provided herein is a method of making a modified immune cell or modified γδ T cell as herein disclosed, or the plurality thereof, respectively. In embodiments, the method comprises transfecting immune cell(s) or γδ T cell(s) with a construct comprising an expression vector encoding a CAR as herein disclosed, optionally wherein the cell(s) has/have at least one disrupted gene. In embodiments, the method comprises retroviral transduction. In embodiments, the method comprises ex vivo expansion of the immune cell(s) or γδ T cell(s), wherein the ex vivo expansion is performed before transfection and/or after transfection of the expression vector. [0034] According to an aspect, provided herein is an antibody-drug conjugate (ADC), comprising an affinity binding entity as herein disclosed. Page 8 of 203 1097657169\1\AMERICAS [0035] According to an aspect, provided herein is a pharmaceutical composition comprising an affinity binding entity as herein disclosed, a modified immune or γδ T cell as herein disclosed, or an ADC as herein disclosed. In embodiments, the pharmaceutical composition comprises a pharmaceutically acceptable carrier. [0036] According to an aspect, provided herein is a method of inhibiting the growth of a cell that exhibits cell surface expression of B7H6, comprising contacting the cell with an affinity binding entity as herein disclosed, a modified immune cell(s) as herein disclosed, a γδ T cell(s) as herein disclosed, an ADC as herein disclosed, or a pharmaceutical composition as herein disclosed. [0037] According to an aspect, provided herein is a method of killing a tumor cell that exhibits cell surface expression of B7H6. In embodiments, the method comprises contacting the tumor cell with a therapeutically effective amount of an affinity binding entity as herein disclosed, modified immune cell(s) as herein disclosed, γδ T cell(s) as herein disclosed, or a pharmaceutical composition as herein disclosed. [0038] In embodiments, the method comprises introducing into a host organism comprising the tumor cell the therapeutically affective amount of the affinity binding entity, the modified immune cell(s), the γδ T cell(s), the ADC, or the pharmaceutical composition. [0039] In embodiments, the method comprises introducing into the host organism comprising the tumor cell the therapeutically affective amount of the affinity binding entity, the modified immune cell(s), the γδ T cell(s), the ADC, or the pharmaceutical composition; and simultaneously or sequentially administering one or more methods to elevate common gamma chain cytokine(s). In embodiments, the administering one or more methods to elevate common gamma chain cytokine(s) comprises simultaneously or sequentially administering an amount of common gamma chain cytokine(s) effective to increase proliferation, cytotoxic activity, persistence, or the combination thereof of the introduced modified immune cell(s) or γδ T cell(s). In embodiments, the one or more methods to elevate common gamma chain cytokine(s) comprises lymphodepletion before introducing the modified immune cell(s) or the γδ T cell(s). In embodiments, the one or more methods to elevate common gamma chain cytokine(s) comprises secretion of one or more common gamma chain cytokine(s) from the introduced modified immune cell(s) or γδ T cell(s). In embodiments, the method reduces the in vivo tumor burden in the host organism and/or increases the mean survival time of the host organism as compared to a control organism, wherein the control Page 9 of 203 1097657169\1\AMERICAS organism is not treated with the affinity binding entity, the modified immune cell(s), γδ T cell(s), the ADC, or the pharmaceutical composition. [0040] In embodiments of the methods disclosed herein, the host organism is human. In embodiments of the methods disclosed herein, the method is a method of treating cancer in a subject in need thereof. [0041] According to an aspect, provided is a use of an affinity binding entity as herein disclosed, modified immune cell(s) as herein disclosed, γδ T cell(s) as herein disclosed, an ADC as herein disclosed, or a pharmaceutical composition as herein disclosed in the preparation of a medicament for the treatment of a disease or condition. In embodiments, the disease or condition is cancer. [0042] In one aspect, provided is a method of reducing or inhibiting the graft vs. host response to an immune cell administered to a subject in need thereof, comprising administering a therapeutically effective amount of γδ T cells according to the subject invention. In embodiments, the γδ T cells may comprise a dual CAR binding to CD70 and B7H6, or the method may further comprise co-administering the γδ T cell according to the subject invention simultaneously or sequentially with an immune cell (e.g., a T cell or NK cell) comprising a CAR binding to B7H6 and a CAR binding to CD70. INCORPORATION BY REFERENCE [0043] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. BRIEF DESCRIPTION OF THE DRAWINGS [0044] FIG.1 is a graph illustrating B7H6 binding specificity of phage-display derived antibody IgG clones against B7H6+ cell lines. [0045] FIG.2 is a table showing EC50 values from phage-display derived antibody clones against recombinant B7H6 protein. [0046] FIGS.3A-3C illustrate phage-display derived antibody IgG clones binding to the V- distal and C-proximal membrane domain of B7H6. Page 10 of 203 1097657169\1\AMERICAS [0047] FIG.4 is a graph illustrating B7H6 cell surface expression in different cancer cell lines. [0048] FIGS.5A-5B illustrate NFAT activity of Jurkat Lucia cells modified with anti-B7H6 scFv clones formatted into a CAR (B7H6 CAR), [0049] FIGS.6A-6C are graphs illustrating in vitro cytotoxicity of B7H6 CAR Vδ1 T cells against various tumor cell lines. [0050] FIGS.7A-7B are graphs illustrating in vitro cytokine polyfunctionality of B7H6 CAR Vδ1 T cells stimulated with B7-H6+ target tumor cells. [0051] FIGS.8A-8B depict data illustrating the T cell memory profile, chemokine receptors, and activation/exhaustion-associated marker expression on B7H6 CAR Vδ1 T cells. [0052] FIG.9 is a series of graphs illustrating in vitro proliferation of B7H6 CAR Vδ1 T cells stimulated with B7H6+ target tumor cells. [0053] FIGS.10A-10B are graphs illustrating that B7H6 CAR Vδ1 T cells maintain in vitro cytotoxicity against B7-H6+ target tumor cells in the presence of soluble B7H6. [0054] FIGS.11A-11B are graphs illustrating in vivo therapeutic efficacy of B7H6 CAR Vδ1 T cells in a subcutaneous human xenograft HCT-15 cell NOD scid gamma (NSG) mouse model. [0055] FIG.12 depicts plots illustrating armoring of B7H6 CAR Vδ1 T cells with dominant negative TGFR ^2 (dnTGFR ^2). [0056] FIG.13 shows graphs illustrating in vitro inhibition of TGF ^1-induced pSMAD2/3 expression in B7H6 CAR Vδ1 T cells containing dnTGFR ^2. [0057] FIG.14 is a graph illustrating enhanced in vitro cytotoxicity of B7H6 CAR Vδ1 T cells containing dnTGFR ^2 against B7H6+ target tumor cells. [0058] FIG.15 is a graph illustrating gene knockout efficiency of two different guide RNAs targeting cytokine inducible SH2 containing protein (CISH). [0059] FIG.16A is a graph illustrating that Vδ1 T cells with CISH knocked out can be enriched post-depletion of αβ T cells. Page 11 of 203 1097657169\1\AMERICAS [0060] FIG.16B is a graph illustrating viability of Vδ1 T cells with CISH knocked out. [0061] FIGS.17A-17B illustrate CISH KO enhanced the in vitro cytotoxicity of Vδ1 T cells. [0062] FIG.18 illustrates the CAR-mbIL-12 constructs designs. [0063] FIGS.19A-19D illustrate the expansion and expression of the CAR-mbIL-12 in Vδ1 T cells. [0064] FIG.20 illustrates the enhanced in vitro cytotoxicity of CAR-mbIL-12 in Vδ1 T cells. [0065] FIG. 21 illustrates in vivo therapeutic efficacy of CAR-mbIL-12 in Vδ1 T cells in a subcutaneous human xenograft Raji cell NSG mouse model. [0066] FIGS.22A-22B illustrate CBL-B KO enhanced the in vitro cytotoxicity of Vδ1 T cells. [0067] FIGS.23A-23B illustrate Roquin KO enhanced the in vitro cytotoxicity of Vδ1 T cells. [0068] FIGS.24A-24B illustrate CD58 or ICAM-1 KO enhanced the in vitro cell survival of Vδ1 T cells in an allogeneic MLR assay. DETAILED DESCRIPTION I. Definitions [0069] For purposes of interpreting this specification, the following definitions will apply, and whenever appropriate, terms used in the singular will also include the plural and vice versa. In the event that any definition set forth conflicts with any document incorporated herein by reference, the definition set forth below shall control. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the disclosure pertains. [0070] “About” as used herein when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass variations of ±20% or ±10%, more preferably ±5%, even more preferably ±1%, and still more preferably ±0.1% from the specified value, as such variations are appropriate to perform the disclosed methods. [0071] As used herein, “w/v” refers to the weight of the component in a given volume of solution. Page 12 of 203 1097657169\1\AMERICAS [0072] “Ranges”: throughout this disclosure, various aspects of the disclosure can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the disclosure. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 2.7, 3, 4, 5, 5.3, and 6. This applies regardless of the breadth of the range. [0073] The terms “patient,” “subject,” “individual,” and the like are used interchangeably herein, and refer to any animal amenable to the methods described herein. In certain non- limiting embodiments, the patient, subject or individual is a human. [0074] The term “diagnosis”, or “diagnosing” as used herein refers to the process of identifying a disease, such as cancer, by its signs, symptoms, and/or results of various tests. A conclusion reached through such a process is a diagnosis. Forms of testing commonly performed include blood tests, medical imaging, urinalysis, biopsy, and the like. [0075] As used herein, the term “agent” refers to any protein, nucleic acid molecule (including chemically modified nucleic acids), compound, antibody, small molecule, organic compound, inorganic compound, other molecule of interest, or cell (e.g., cell engineered to express a chimeric antigen receptor). Agent can include a therapeutic agent, a diagnostic agent or a pharmaceutical agent. A therapeutic or pharmaceutical agent is one that alone or together with an additional agent induces the desired response (such as inducing a therapeutic or prophylactic effect when administered to a subject, including treating a subject suffering from cancer, or other disease/condition. [0076] The term "therapeutically effective amount", or simply “effective amount” refers to the amount of an agent or composition (e.g., composition comprising an agent) that will elicit a biological or medical response of a tissue, system, or subject that is being sought by the researcher, veterinarian, medical doctor or other clinician. The term "therapeutically effective amount" includes that amount of an agent, or a composition comprising an agent, that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more Page 13 of 203 1097657169\1\AMERICAS of the signs or symptoms of the disorder or disease (e.g., cancer) being treated. The therapeutically effective amount will vary depending on the composition, the disease and its severity and the age, weight, etc., of the subject to be treated. [0077] The term “γδ T cells (gamma delta T cells)” as used herein refers to a subset of T cells that express a distinct T cell receptor (TCR), namely γδTCR, on their surface, composed of one γ- chain and one δ-chain. The term “γδ T cells” specifically includes all subsets of γδ T cells, including, without limitation, Vδ1 and Vδ2, Vδ3 γδ T cells, as well as naïve, effector memory, central memory, and terminally differentiated γδ T cells. As a further example, the term “γδ T cells” includes Vδ4, Vδ5, Vδ7, and Vδ8 γδ T cells, as well as Vγ2, Vγ3, Vγ5, Vγ8, Vγ9, Vγ10, and Vγ11 γδ T cells. In embodiments, the γδ T cells are Vδ1-, Vδ2-, or Vδ1- and Vδ2-. Compositions and methods for making and using engineered and non-engineered γδ T cells and/or sub-types thereof include, without limitation, those described in US 2016/0175358; WO 2017/197347; US 9499788; US 2018/0169147; US 9907820; US 2018/0125889 and US 2017/0196910, the contents of each of which are incorporated by reference for all purposes, including the the compositions and methods for making and using engineered and non-engineered γδ T cells and/or sub-types thereof. The present application further contemplates T cells, or other engineered leukocytes or lymphocytes, that express one γ-chain or one δ-chain, optionally in combination with a second polypeptide to form a functional TCR. Such engineered leukocytes or lymphocytes, that express one γ-chain or one δ-chain may be used in the methods or present in the compositions described herein. [0078] The ^ ^ T cells described herein can be ^1, ^2, ^3, or ^4 ^ ^ ^T cells, or combinations thereof. In some cases, the ^ ^ T cells are mostly (>50%), substantially (>90%), essentially all, or entirely ^2 ^ ^ T cells. In some cases, the ^ ^ T cells are mostly (>50%), substantially (>90%), essentially all, or entirely ^1 ^ ^ T cells. In some cases, the ^ ^ T cells are mostly (>50%), substantially (>90%), essentially all, or entirely ^ ^ ^ ^ T cells. [0079] ^ ^ T cells for use as described herein can be obtained from an allogeneic or an autologous donor. The ^ ^ T cells can be, partially or entirely purified, or not purified, and expanded ex vivo. Methods and compositions for ex vivo expansion include, without limitation, those described in WO 2017/197347. The expansion may be performed before or after, or before and after, a CAR polypeptide of the present disclosure is introduced into the ^ ^ T cell(s). Other Page 14 of 203 1097657169\1\AMERICAS additional or alternative methods of expansion include the use of, e.g., artificial antigen-presenting cells (aAPCs), aminobisphosphonates, cytokine cocktails, and feeder cells (Cortés-Selva, D et al., (2021) Trends Pharmacol Sci.42(1): 45-59). [0080] As used herein, the term “αβ T cell” refers to T cells expressing α and β chains of the TCR as part of a complex with CD3 chain molecules. Each α and β chain contains one variable and one constant domain. αβ T cells primarily recognize peptide antigens presented by major histocompatibility complex (MHC) class I and class II molecules, where most of the receptor diversity is contained within the third complementarity determining region (CDR3) of the TCR α and β chains. [0081] As used herein, the term “Natural killer (NK) cell” refers to CD56+CD3 granular lymphocytes that play important roles in immunity against viruses and in the immune surveillance of tumors, and constitute a critical cellular subset of the innate immune system (Godfrey J, et al. Leuk Lymphoma 2012 53:1666-1676). NK cells express a remarkably diverse repertoire of inhibitory and activating receptors on their cell surface, which regulates their immune responses. NK cells can kill transformed or infected cells by the release of perforin and granzymes or by using effector molecules of the tumor necrosis factor (TNF) family, such as TNF, TNF-related apoptosis inducing ligand (TRAIL), and Fas ligand, which induce apoptosis in the target cells. Additionally, upon activation NK cells rapidly produce chemokines and cytokines, including interferon (IFN)- γ, GM-CSF, and IL-10, that recruit and affect the function of hematopoietic and nonhematopoietic cells in the host. Unlike cytotoxic CD8+ T lymphocytes, NK cells launch cytotoxicity against tumor cells without the requirement for prior sensitization, and can also eradicate MHC-I-negative cells (Narni-Mancinelli E, et al. Int Immunol 201123:427-431). NK cells are considered fairly safe effector cells, as they may avoid the potentially lethal complications of cytokine storms (Morgan R A, et al. Mol Ther 201018:843-851), tumor lysis syndrome (Porter D L, et al. N Engl J Med 2011365:725-733), and on-target, off-tumor effects. [0082] NK cells can be obtained from an allogeneic or an autologous donor. The NK cells can be partially or entirely purified, or not purified, and expanded ex vivo. Methods and compositions for ex vivo expansion include, without limitation, those described in Becker et al., (2016) Cancer Immunol. Immunother.65(4): 477-84). The expansion may be performed before or after, or before and after, a CAR is introduced into the NK cell(s). Briefly, and without limitation, expansion of Page 15 of 203 1097657169\1\AMERICAS NK cells can include the use of engineered feeder cells, cytokine cocktails (e.g., IL-2, IL-15), and/or aAPCs (Cortés-Selva, D et al., (2021) Trends Pharmacol Sci.42(1): 45-59). [0083] In some examples, placental hematopoietic stem-cell derived natural killer (PNK) cells or immortalized cell lines (e.g., NK-92) may be engineered to express chimeric adaptor polypeptides of the present disclosure. In other examples, NK cells that can be used for engineering the expression of CARs herein can be differentiated from human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs). As used herein, the term “Natural killer T (NKT) cells” are T lineage cells that share morphological and functional characteristics with both T cells and NK cells. NKT cells are rapid responders of the innate immune system and mediate potent immunoregulatory and effector functions in a variety of disease settings. Ligand recognition in NKT cells leads to rapid secretion of proinflammatory cytokines (such as IFN-γ and TNF-α) and anti-inflammatory cytokines (such as IL-4, IL-10, and IL-13) that enhance the immune response to e.g., cancer by directly targeting tumor cells and by indirectly modulating the antitumor response through the release of diverse cytokines or by altering the TME. Following activation, NKT cells can immediately commence cytokine secretion without first having to differentiate into effector cells. The rapidity of their response makes NKT cells important players in the very first lines of innate defense against some types of bacterial and viral infections. In addition, many of the cytokines secreted by NKT cells have powerful effects on αβ T cell differentiation and function, linking NKT cells to adaptive defense. NKT cells bridge the adaptive immune system with the innate immune system. Unlike conventional T cells that recognize peptide antigens presented by major histocompatibility complex (MHC) molecules, NKT cells recognize glycolipid antigen presented by a molecule called CD1d. NKT cells can be obtained from an allogeneic or an autologous donor. The NKT cells can be partially or entirely purified, or not purified, and expanded ex vivo. Briefly and without limitation, NKT cells can be expanded via the use of ex vivo IL-2, and/or monoclonal antibodies specific for the TCR α-chain CDR3 loop (Cortés-Selva, D et al., (2021) Trends Pharmacol Sci.42(1): 45-59). [0084] As used herein, the term “γδ natural killer T cells” or “γδ NKT cells” refers to iPSC- derived cells that express γδ TCRs and NK receptors, but lack the expression of hallmark γδ T cell markers (Cortés-Selva, D et al., (2021) Trends Pharmacol Sci. 42(1): 45-59). These cells have been shown to have anti-tumor activity against a broad number of cancer cell lines, but not against normal cells, and showed more potent killing than donor-derived γδ T cells or donor-derived NK Page 16 of 203 1097657169\1\AMERICAS cells (Zeng J et al., (2019) PLoS ONE 14(5): e0216815). CARs can be expressed in γδ NKT cells, in embodiments herein, for use in accordance with the methods disclosed herein. [0085] As used herein, the term “myeloid cells” refers to a subgroup of leukocytes represented by granulocytes, monocytes, macrophages, and dendritic cells (DCs). They circulate through the blood and lymphatic system and are rapidly recruited to sites of tissue damage and infection via various chemokine receptors. Within the tissues they are activated for phagocytosis as well as secretion of inflammatory cytokines, thereby playing major roles in protective immunity. Myeloid cells can also be found in tissues under steady-state condition, where they control development, homeostasis, and tissue repair. [0086] As used herein, the term “macrophages” refers to highly plastic innate cells with functional and phenotypic signatures that can be shaped in response to various stimuli. Macrophage polarization is broadly simplified into two different states, either a M1 phenotype (classically activated) in response to factors such as lipopolysaccharide (LPS) or IFN-γ, or a M2 phenotype in response to cytokines such as IL-4, IL-5, and IL-13. An example of M1-like macrophages express iNOS and proinflammatory cytokines such as TNF-α, IL1-β, IL-6, IL-12, and IL-23. An example of M2 macrophages exhibit increased expression of CD209, CD200R, CD1a, and CD1b in humans, and have been implicated in wound healing and antitumor responses. The ability of macrophages to infiltrate solid tumors and be reprogrammed, as well as the antitumor effects associated with a switch to the M1 phenotype, render macrophages relevant to the present disclosure in terms of engineered macrophages that express a CAR described herein. For example, it has been shown that macrophages can be reprogrammed towards antitumor M1 phenotype cells that are capable of producing nitric oxide and inducing IL-12-dependent NK-mediated antitumor effects by inhibiting NK-κB signaling in a murine model of ovarian cancer (Zhang F et al., (2019) Nat Commun 10: 3974). [0087] Macrophages can be obtained/derived from an allogeneic or an autologous donor. The macrophages can be partially or entirely purified, or not purified, and cultured ex vivo (see, e.g., Davies JQ and Gordon A (2005) Methods Mol Biol 290:105016). In embodiments, the present disclosure encompasses macrophages derived from hESCs (Karlsson, KR et al., (2008) Exp Hematol 36: 1167-1175), or iPSC-derived macrophages (Takata K. et al., (2017) Immunity 47: 183-198). Page 17 of 203 1097657169\1\AMERICAS [0088] As used herein, the term “T lymphocyte” or “T cell” refers to an immune cell that expresses or has expressed CD3 (CD3+) and a T Cell Receptor (TCR+). T cells play a central role in cell-mediated immunity. A T cell that “has expressed” CD3 and a TCR has been engineered to eliminate CD3 and/or TCR cell surface expression. [0089] As used herein, the term “TCR” or “T cell receptor” refers to a dimeric heterologous cell surface signaling protein forming an alpha-beta or gamma-delta receptor or combinations thereof. αβTCRs recognize an antigen presented by an MHC molecule, whereas γδTCR can recognize an antigen independently of MHC presentation. [0090] The term "MHC" (major histocompatibility complex) refers to a subset of genes that encodes cell-surface antigen-presenting proteins. In humans, these genes are referred to as human leukocyte antigen (HLA) genes. Herein, the abbreviations MHC or HLA are used interchangeably. [0091] “Activation”, as used herein, refers to the state of a T cell that has been sufficiently stimulated to induce detectable cellular proliferation. Activation can also be associated with induced cytokine production, and detectable effector functions. The term “activated T cells” refers to, among other things, T cells that are undergoing cell division. [0092] The “costimulatory domain” in the context of a chimeric receptor, also referred to herein as a chimeric antigen receptor (CAR), of the present disclosure enhances cell proliferation, cell survival and development of memory cells for cytotoxic cells that express the chimeric receptor. The chimeric receptors of the invention may include one or more costimulatory domains selected from the costimulatory domains of proteins in the TNFR superfamily, CD28, CD137 (4- 1BB), CD134 (OX40), Dapl0, CD27, CD2, CD7, CD5, ICAM-1, LFA-1 (CD1 la/CD18), Lck, TNFR-I, PD-1, TNFR-II, Fas, CD30, CD40, ICOS LIGHT, NKG2C, B7-H3, or combinations thereof. If the chimeric receptor includes more than one costimulatory domain, these domains may be arranged in tandem, optionally separated by a linker. [0093] The term “costimulatory domain” as used herein also encompasses any modifications thereof, examples of which are described in US Patent Application No.20200129554; US Patent Application No.20200317777; WO2019010383; Li, W., et al., (2020) Immunity 53: 456-470; and Li, G., et al., (2017) J Immunol 198(1 Supplement): 198.4, the contents of each of which are incorporated herein in their entirety. Page 18 of 203 1097657169\1\AMERICAS [0094] The “intracellular signaling domain” in the context of a chimeric receptor of the present disclosure transduces the effector function signal and directs the cytotoxic cell to perform its specialized function, i.e., harming and/or destroying the target cells. Examples of suitable intracellular signaling domains include, e.g., the ζ chain of the T cell receptor complex or any of its homologs, e.g., η chain, FcsRly and β chains, MB 1 (Iga) chain, B29 (Ig) chain, etc., human CD3 ζ chain, CD3 polypeptides (Δ, δ and ε), syk family tyrosine kinases (Syk, ZAP 70, etc.), src family tyrosine kinases (Lck, Fyn, Lyn, etc.) and other molecules involved in T cell transduction, such as CD2, CD5 and CD28. In embodiments, the intracellular signaling domain of a chimeric receptor may be human CD3 ζ chain, FcyRIII, FcsRI, cytoplasmic tails of Fc receptors, an immunoreceptor tyrosine-based activation motif (ITAM) bearing cytoplasmic receptors and combinations thereof. [0095] The intracellular signaling domains may include intracellular signaling domains of several types of various other immune signaling receptors, including, but not limited to, first, second, and third generation T cell signaling proteins including CD3, B7 family costimulatory, and Tumor Necrosis Factor Receptor (TNFR) superfamily receptors (Park et al., "Are all chimeric antigen receptors created equal?" J Clin Oncol., vol. 33, pp. 651-653, 2015). Additional intracellular signaling domains include signaling domains used by NK and NKT cells (Hermanson, et al., "Utilizing chimeric antigen receptors to direct natural killer cell activity," Front Immunol., vol. 6, p. 195, 2015) such as signaling domains of NKp30 (Zhang et al., "An NKp30-based chimeric antigen receptor promotes T cell effector functions and antitumor efficacy in vivo," J Immunol., vol. 189, pp. 2290-2299, 2012), and DAP12 (Topfer et al., "DAP12-based activating chimeric antigen receptor for NK cell tumor immunotherapy," J Immunol., vol. 194, pp. 3201- 3212, 2015), NKG2D, NKp44, NKp46, DAP10, and CD3z. Additionally intracellular signaling domains also includes signaling domains of human Immunoglobulin receptors that contain immunoreceptor tyrosine based activation motif (ITAM) such as FcgammaRI, FcgammaRIIA, FcgammaRIIC, FcgammaRIIIA, FcRL5 (Gillis et al., "Contribution of Human Fc.gamma.Rs to Disease with Evidence from Human Polymorphisms and Transgenic Animal Studies," Front Immunol., vol.5, p.254, 2014). [0096] In embodiments, the intracellular signaling domain includes a cytoplasmic signaling domain of TCR ζ, FcR γ, FcR β, CD3 γ, CD3 δ, CD3 ε, CD5, CD22, CD79a, CD79b, or CD66d. In exemplary embodiments the intracellular signaling domain in the chimeric receptor includes a Page 19 of 203 1097657169\1\AMERICAS cytoplasmic signaling domain of human CD3 ζ. The term “intracellular signaling domain” as used herein also encompasses any modifications thereof, examples of which are described in US Patent Application No. 2020/0317777, as well as Navarro, P., and Reyburn, HT., (2009) J Biol Chem 284(24): 16463-16472; Giurisato, E., et al., (2007) Mol Cell Biol 27(24): 8583-8599; and Wu, J., et al., (2000) J Exp Med 192(7): 1059-1068, the contents of each of which are incorporated herein in their entirety. [0097] The term “affinity binding entity” refers to a binding moiety which binds to a specific antigen with a higher affinity than to a non-specific antigen and is endowed with an affinity of at least 10-6 M, as determined by assays which are well known in the art, including surface plasmon resonance (SPR). According to a specific embodiment, the affinity is 500 nM-0.01 nM, 100 nM- 0.01 nM, 50 nM-0.01 nM, 10 nM-0.01 nM, 5 nM-0.01 nM. [0098] According to embodiments, the affinity binding entity is an antibody. The term “antibody” is used in the broadest sense and specifically covers, for example, single anti-B7H6 monoclonal antibodies (including agonist, antagonist, neutralizing antibodies, full length or intact monoclonal antibodies), anti-B7H6 antibody compositions with polyepitopic specificity, polyclonal antibodies, multivalent antibodies, multispecific antibodies (e.g., bispecific antibodies so long as they exhibit the desired biological activity), formed from at least two intact antibodies, single chain anti-B7H6 antibodies, and fragments of anti-B7H6 antibodies (see below), including Fab, Fab’, F(ab’)2 and Fv fragments, diabodies, single domain antibodies (sdAbs), as long as they exhibit the desired biological or immunological activity. Also included among anti-B7H6 antibodies, and among fragments in particular, are portions of anti-B7H6 antibodies (and combinations of portions of anti-B7H6 antibodies, for example, scFv) that may be used as targeting arms, directed to e.g., a B7H6 epitope, in chimeric antigenic receptors of the present disclosure. Such fragments are not necessarily proteolytic fragments but rather portions of polypeptide sequences that can confer affinity for target. The term “immunoglobulin” (Ig) is used interchangeably with antibody herein. An antibody can be, for example, human, humanized and/or affinity matured. [0099] Methods of making antibodies and antibody fragments are known in the art. (See for example, Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, New York, 1988, incorporated herein by reference). Page 20 of 203 1097657169\1\AMERICAS [00100] Antibodies can be produced by the immunization of various animals, including mice, rats, rabbits, goats, primates, humans and chickens with a target antigen such as B7H6 or peptide fragments of B7H6 containing the anti-B7H6 epitope of the present disclosure. Antibodies may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352: 624-8 (1991) and Marks et al., J. Mol. Biol., 222: 581-97 (1991), for example. Antibodies or antigen-binding fragment of the present invention can be purified by methods known in the art, for example, gel filtration, ion exchange, affinity chromatography, etc. Affinity chromatography or any of a number of other techniques known in the art can be used to isolate polyclonal or monoclonal antibodies from, for example, serum, ascites fluid, or hybridoma supernatants. [00101] The terms “anti-B7H6 antibody”, “B7H6 antibody”, and “an antibody that binds to B7H6” are used interchangeably. Anti-B7H6 antibodies are preferably capable of binding with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent, whether in isolation or as part of fusion protein, cell, or cell composition. [00102] An “isolated antibody” is one which has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials which would interfere with therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes. [00103] The basic 4-chain antibody unit is a heterotetrameric glycoprotein composed of two identical light (L) chains and two identical heavy (H) chains. In the case of IgGs, the 4-chain unit is generally about 150,000 daltons. Each L chain is linked to a H chain by one covalent disulfide bond, while the two H chains are linked to each other by one or more disulfide bonds depending on the H chain isotype. Each H and L chain also has regularly spaced intrachain disulfide bridges. Each H chain has at the N-terminus, a variable domain (VH) followed by three constant domains (CH) for each of the α and γ chains and four CH domains for μ and ε isotypes. Each L chain has at the N-terminus, a variable domain (VL) followed by a constant domain (CL) at its other end. The VL is aligned with the VH and the CL is aligned with the first constant domain of the heavy chain (CH1). Particular amino acid residues are believed to form an interface between the light chain and heavy chain variable domains. The pairing of a VH and VL together forms a single antigen-binding site. For the structure and properties of the different classes of antibodies, see, Page 21 of 203 1097657169\1\AMERICAS e.g., Basic and Clinical Immunology, 8th edition, Daniel P. Stites, Abba I. Terr and Tristram G. Parslow (eds.), Appleton & Lange, Norwalk, CT, 1994, at page 71 and Chapter 6. [00104] The L chain from any vertebrate species can be assigned to one of two clearly distinct types, called kappa and lambda, based on the amino acid sequences of their constant domains. Depending on the amino acid sequence of the constant domain of their heavy chains (CH), immunoglobulins can be assigned to different classes or isotypes. There are five classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, having heavy chains designated α, δ, ε, γ, and μ, respectively. The γ and α classes are further divided into subclasses on the basis of relatively minor differences in CH sequence and function, e.g., humans express the following subclasses: IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2. [00105] The “variable region” or “variable domain” of an antibody refers to the amino-terminal domains of the heavy or light chain of the antibody. The variable domain of the heavy chain may be referred to as “VH” or “VH” The variable domain of the light chain may be referred to as “VL” or “VL”. These domains are generally the most variable parts of an antibody and contain the antigen-binding sites. [00106] The term “variable” refers to the fact that certain segments of the variable domains differ extensively in sequence among antibodies. The V domain mediates antigen binding and defines specificity of a particular antibody for its particular antigen. However, the variability is not evenly distributed across the 110-amino acid span of the variable domains. Instead, the V regions consist of relatively invariant stretches called framework regions (FRs) of 15-30 amino acids separated by shorter regions of extreme variability called “hypervariable regions” that are each about 9-12 amino acids long. The variable domains of native heavy and light chains each comprise four FRs, largely adopting a β-sheet configuration, connected by three hypervariable regions, which form loops connecting, and in some cases forming part of, the β-sheet structure. The hypervariable regions in each chain are held together in close proximity by the FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991)). [00107] An “intact” antibody is one which comprises an antigen-binding site as well as a CL and at least heavy chain constant domains, CH1, CH2 and CH3. The constant domains may be Page 22 of 203 1097657169\1\AMERICAS native sequence constant domains (e.g., human native sequence constant domains) or amino acid sequence variant thereof. Preferably, the intact antibody has one or more effector functions. [00108] “Antibody fragments” comprise a portion of an intact antibody, preferably the antigen binding or one or more variable regions of the intact antibody. Examples of antibody fragments include Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies (see U.S. Patent No. 5,641,870, Example 2; Zapata et al., Protein Eng.8(10): 1057-62 (1995)); single-chain antibody molecules; and multispecific antibodies formed from antibody fragments. In one embodiment, an antibody fragment comprises an antigen binding site of the intact antibody and thus retains the ability to bind antigen. Also included among anti-B7H6 antibody fragments are portions of anti- B7H6 antibodies (and combinations of portions of anti-B7H6 antibodies, for example, scFv) that may be used as targeting arms, directed to e.g., a B7H6 epitope, in chimeric antigenic receptors of the present disclosure. Such fragments are not necessarily proteolytic fragments but rather portions of polypeptide sequences that can confer affinity for target. [00109] Papain digestion of antibodies produces two identical antigen-binding fragments, called “Fab” fragments, and a residual “Fc” fragment, a designation reflecting the ability to crystallize readily. The Fab fragment consists of an entire L chain along with the variable region domain of the H chain (VH), and the first constant domain of one heavy chain (CH1). Each Fab fragment is monovalent with respect to antigen binding, i.e., it has a single antigen-binding site. Pepsin treatment of an antibody yields a single large F(ab')2 fragment which roughly corresponds to two disulfide linked Fab fragments having divalent antigen-binding activity and is still capable of cross-linking antigen. Fab’ fragments differ from Fab fragments by having additional few residues at the carboxy terminus of the CH1 domain including one or more cysteines from the antibody hinge region. Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear a free thiol group. F(ab')2 antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known. [00110] The Fc fragment comprises the carboxy-terminal portions of both H chains held together by disulfides. The effector functions of antibodies are determined by sequences in the Fc region, which region is also the part recognized by Fc receptors (FcR) found on certain types of cells. Page 23 of 203 1097657169\1\AMERICAS [00111] “Fv” is the minimum antibody fragment which contains a complete antigen-recognition and -binding site. This fragment consists of a dimer of one heavy- and one light-chain variable region domain in tight, non-covalent association. In a single-chain Fv (scFv) species, one heavy- and one light-chain variable domain can be covalently linked by a flexible peptide linker such that the light and heavy chains can associate in a “dimeric” structure analogous to that in a two-chain Fv species. From the folding of these two domains emanate six hypervariable loops (3 loops each from the H and L chain) that contribute the amino acid residues for antigen binding and confer antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site. [00112] “Single-chain Fv” also abbreviated as “sFv” or “scFv” are antibody fragments that comprise the VH and VL antibody domains connected into a single polypeptide chain. In embodiments, the sFv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the sFv to form a desired structure for antigen binding. For a review of sFv, see, e.g., Pluckthun in The Pharmacology of Monoclonal Antibodies, vol.113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315 (1994); Borrebaeck 1995, infra. In one embodiment, an anti-B7H6 antibody derived scFv is used as the targeting arm of a CAR-modified immune cell as disclosed herein. In terms of scFv antibody fragments, where a certain order of VH and VL region in the binding domain is explicitly or implicitly described, the present disclosure also includes the alternate embodiment in which the order of VH and VL regions are reversed, e.g., in an scFv or CAR comprising an scFv binding domain. Thus, description of a VH- VL order also describes the alternate VL-VH order, e.g., in an scFv or CAR comprising an scFv binding domain. Moreover, description of a VL-VH order also describes the alternate VH-VL order, e.g., in an scFv or a CAR comprising an scFv binding domain. VH and VL regions are either joined directly or joined by a peptide-encoding linker, which connects the N-terminus of the VH with the C-terminus of the VL, or the C-terminus of the VH with the N-terminus of the VL. [00113] The scFv linker is usually rich in glycine for flexibility, as well as serine or threonine for solubility. The linker can link the heavy chain variable region and the light chain variable region of the extracellular antigen-binding domain. Non-limiting examples of linkers are disclosed in Shen et al, Anal. Chem.80(6): 1910-1917 (2008) and WO 2014/087010, the contents of which are hereby incorporated by reference in their entireties. Various linker sequences are known in the Page 24 of 203 1097657169\1\AMERICAS art, including, without limitation, glycine serine (GS) linkers such as (GS)n, (GSGGS)n (SEQ ID NO: 174), (GGGS)n (SEQ ID NO: 175), and (GGGGS)n (SEQ ID NO: 176), where n represents an integer of at least 1. Exemplary linker sequences can comprise amino acid sequences including, without limitation, GGSG (SEQ ID NO: 177), GGSGG (SEQ ID NO: 178), GSGSG (SEQ ID NO: 179), GSGGG (SEQ ID NO: 180), GGGSG (SEQ ID NO: 181), GSSSG (SEQ ID NO: 182), GGGGS (SEQ ID NO: 183), GGGGSGGGGSGGGGS (SEQ ID NO: 172) and the like. Those of skill in the art would be able to select the appropriate linker sequence for use in the present invention. In one embodiment, an antigen binding domain of the present invention comprises a heavy chain variable region (VH) and a light chain variable region (VL), wherein the VH and VL is separated by the linker sequence having the amino acid sequence GGGGSGGGGSGGGGS (SEQ ID NO: 172), which may be encoded by the nucleic acid sequence GGAGGCGGAGGATCTGGTGGTGGTGGATCTGGCGGCGGAGGCTCT (SEQ ID NO: 173). [00114] The term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations which include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they may be synthesized uncontaminated by other antibodies. The modifier “monoclonal” is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies useful in the present invention may be prepared by the hybridoma methodology first described by Kohler et al., Nature, 256: 495 (1975), or may be made using recombinant DNA methods in bacterial, eukaryotic animal or plant cells (e.g., U.S. Patent No.4,816,567). The “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352: 624-8 (1991) and Marks et al., J. Mol. Biol., 222: 581-97 (1991), for example. [00115] The term “hypervariable region”, “HVR”, or “HV”, when used herein refers to the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops. Generally, antibodies comprise six hypervariable regions; three in the VH (H1, H2, H3), and three in the VL (L1, L2, L3). A number of hypervariable region delineations Page 25 of 203 1097657169\1\AMERICAS are in use and are encompassed herein. The Kabat Complementarity Determining Regions (CDRs) are based on sequence variability and are the most commonly used (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991)). Chothia refers instead to the location of the structural loops (Chothia and Lesk J. Mol. Biol. 196:901-917 (1987)). The end of the Chothia CDR-H1 loop when numbered using the Kabat numbering convention varies between H32 and H34 depending on the length of the loop (this is because the Kabat numbering scheme places the insertions at H35A and H35B; if neither 35A nor 35B is present, the loop ends at 32; if only 35A is present, the loop ends at 33; if both 35A and 35B are present, the loop ends at 34). The AbM hypervariable regions represent a compromise between the Kabat CDRs and Chothia structural loops, and are used by Oxford Molecular’s AbM antibody modeling software. The “contact” hypervariable regions are based on an analysis of the available complex crystal structures. The residues from each of these hypervariable regions are noted below. Loop Kabat AbM Chothia Contact ---- ----- --- ------- ------- L1 L24-L34 L24-L34 L24-L34 L30-L36 L2 L50-L56 L50-L56 L50-L56 L46-L55 L3 L89-L97 L89-L97 L89-L97 L89-L96 H1 H31-H35B H26-H35B H26-H32..34 H30-H35B (Kabat Numbering) H1 H31-H35 H26-H35 H26-H32 H30-H35 (Chothia Numbering) H2 H50-H65 H50-H58 H52-H56 H47-H58 H3 H95-H102 H95-H102 H95-H102 H93-H101 [00116] Hypervariable regions may comprise “extended hypervariable regions” as follows: 24- 36 or 24-34 (L1), 46-56 or 50-56 (L2) and 89-97 (L3) in the VL and 26-35B (H1), 50-65, 47-65 Page 26 of 203 1097657169\1\AMERICAS or 49-65 (H2) and 93-102, 94-102 or 95-102 (H3) in the VH. The variable domain residues are numbered according to Kabat et al., supra, for each of these definitions. [00117] “Framework” or “FR” residues are those variable domain residues other than the hypervariable region residues herein defined. [00118] The term “variable domain residue numbering as in Kabat” or “amino acid position numbering as in Kabat”, and variations thereof, refers to the numbering system used for heavy chain variable domains or light chain variable domains of the compilation of antibodies in Kabat et al., supra. Using this numbering system, the actual linear amino acid sequence may contain fewer or additional amino acids corresponding to a shortening of, or insertion into, a FR or CDR of the variable domain. For example, a heavy chain variable domain may include a single amino acid insert (residue 52a according to Kabat) after residue 52 of H2 and inserted residues (e.g., residues 82a, 82b, and 82c, etc. according to Kabat) after heavy chain FR residue 82. The Kabat numbering of residues may be determined for a given antibody by alignment at regions of homology of the sequence of the antibody with a “standard” Kabat numbered sequence. [00119] The Kabat numbering system is generally used when referring to a residue in the variable domain (approximately residues 1-107 of the light chain and residues 1-113 of the heavy chain) (e.g, Kabat et al., supra). The “EU numbering system” or “EU index” is generally used when referring to a residue in an immunoglobulin heavy chain constant region (e.g., the EU index reported in Kabat et al., supra). The “EU index as in Kabat” refers to the residue numbering of the human IgG1 EU antibody. Unless stated otherwise herein, references to residue numbers in the variable domain of antibodies means residue numbering by the Kabat numbering system. [00120] A “blocking” antibody or an “antagonist” antibody is one which inhibits or reduces biological activity of the antigen it binds. Preferred blocking antibodies or antagonist antibodies substantially or completely inhibit the biological activity of the antigen. In one embodiment, an anti-B7H6 antibody is provided, which is an antagonist antibody. [00121] An antibody that “binds” an antigen or epitope of interest is one that binds the antigen or epitope with sufficient affinity that is measurably different from a non-specific interaction. Specific binding can be measured, for example, by determining binding of a molecule compared to binding of a control molecule, which generally is a molecule of similar structure that does not have binding activity. Page 27 of 203 1097657169\1\AMERICAS [00122] The term “antigen” or “Ag” as used herein is defined as a molecule that provokes an immune response. This immune response may involve either antibody production, or the activation of specific immunologically-competent cells, or both. The skilled artisan will understand that any macromolecule, including proteins or peptides, can serve as an antigen. [00123] The term "epitope" includes any protein determinant, lipid or carbohydrate determinant capable of specific binding to an immunoglobulin or T cell receptor. Epitopic determinants usually consist of active surface groupings of molecules such as amino acids, lipids or sugar side chains and usually have specific three-dimensional structural characteristics, as well as specific charge characteristics. [00124] The term "specifically binds”, as used herein refers to a receptor (which can include but is not limited to an antibody or antibody fragment) which recognizes a specific molecule/ligand, but does not substantially recognize or bind other molecules in a sample. For example, a receptor that specifically binds to a molecule from one species may also bind to that molecule from one or more other species. But, such cross-species reactivity does not itself alter the classification as specific. In another example, a receptor that specifically binds to a molecule may also bind to different allelic forms of the molecule. However, such cross reactivity does not itself alter the classification as specific. In some instances, the terms "specific binding" or "specifically binding," can be used in reference to the interaction of a protein (or a peptide) with a second chemical species, to mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, a receptor recognizes and binds to a specific a structure rather than to proteins generally. If receptor is specific for epitope "A", the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled "A" and the receptor, will reduce the amount of labeled A bound to the receptor. [00125] In embodiments, specific binding can be characterized by an equilibrium dissociation constant of at least about 1x10-8 M or less (e.g., a smaller KD denotes a tighter binding). Methods for determining whether two molecules specifically bind are well known in the art and include, for example, equilibrium dialysis, surface plasmon resonance, and the like. Page 28 of 203 1097657169\1\AMERICAS [00126] The term “anti-tumor effect” as used herein, refers to a biological effect which can be manifested by a decrease in tumor volume, a decrease in the number of tumor cells, a decrease in the number of metastases, an increase in life expectancy, or amelioration of various physiological symptoms associated with the cancerous condition. An “anti-tumor effect” can also be manifested by the ability of the peptides, polynucleotides, cells and antibodies of the invention in prevention of the occurrence of tumor in the first place. [00127] The term "cancer" and "cancerous" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include but are not limited to carcinoma, lymphoma, blastoma, sarcoma (including liposarcoma), neuroendocrine tumors, mesothelioma, schwanoma, meningioma, adenocarcinoma, melanoma, and leukemia or lymphoid malignancies. Cancers can include but are not limited to colon cancer, pancreatic cancer, Non-Hodgkin Lymphoma, cervical cancer, hepatic cancer, gastro-intestinal cancer, lung cancer, ovarian cancer, glioblastoma, prostate cancer, breast cancer, ovarian cancer, kidney cancer, bladder cancer, melanoma, and glioma. [00128] As used herein, the term “autologous” is meant to refer to any material derived from an individual which is later to be re-introduced into the same individual. [00129] As used herein, the term “allogeneic” refers to material derived from an animal which is later introduced into a different animal of the same species. [00130] A “modification” of an amino acid residue/position, as used herein, refers to a change of a primary amino acid sequence as compared to a starting amino acid sequence, wherein the change results from a sequence alteration involving the amino acid residue/positions. For example, typical modifications include substitution of the residue (or at the position) with another amino acid (e.g., a conservative or non-conservative substitution), insertion of one or more (generally fewer than 5 or 3) amino acids adjacent to the residue/position, and deletion of the residue/position. An “amino acid substitution”, or variation thereof, refers to the replacement of an existing amino acid residue in a predetermined (starting) amino acid sequence with a different amino acid residue. Generally, the modification results in alteration in at least one physicobiochemical activity of the variant polypeptide compared to a polypeptide comprising the starting (or “wild type”) amino acid sequence. For example, in the case of an antibody, a physicobiochemical activity that is altered can be binding affinity, binding capability and/or binding effect upon a target molecule. Page 29 of 203 1097657169\1\AMERICAS [00131] To “treat or prevent” a disease as the term is used herein, means to reduce the frequency or severity of at least one sign or symptom of a disease or disorder experienced by a subject. In one example, a therapy (e.g., administration of a therapeutic agent of the present disclosure) treats a disease or condition by decreasing one or more signs or symptoms associated with the disease or condition, for example as compared to the response in the absence of the therapy. For example, administration of a therapeutic agent may provide an anti-tumor effect that decreases one or more signs or symptoms associated with cancer. Treating or preventing can refer to delaying the onset of symptoms, reducing the severity of symptoms, reducing the severity of an acute episode, reducing the number of symptoms, reducing the incidence of disease-related symptoms, reducing the latency of symptoms, ameliorating symptoms, reducing secondary symptoms, reducing secondary infections, prolonging patient survival, preventing relapse to a disease, decreasing the number or frequency of relapse episodes, increasing latency between symptomatic episodes, increasing time to sustained progression, expediting remission, inducing remission, augmenting remission, speeding recovery, or increasing efficacy of or decreasing resistance to alternative therapeutics. In one embodiment, "treating" refers to both therapeutic treatment and prophylactic or preventive measures, wherein the object is to prevent or lessen the targeted pathologic condition or disorder as described herein. [00132] As used herein, the term “administration” means to provide or give a subject one or more agents, such as an agent that treats one or more signs or symptoms associated with a condition/disorder or disease including but not limited to cancer (e.g., lymphoma), viral infection, bacterial infection, etc., by any effective route. Exemplary routes of administration include, but are not limited to, injection (such as subcutaneous, intramuscular, intradermal, intraperitoneal, and intravenous), oral, sublingual, rectal, transdermal, intranasal, vaginal and inhalation routes. Administration “in combination with” one or more further therapeutic agents includes simultaneous (concurrent) and sequential administration in any order. [00133] The term “pharmaceutically acceptable”, as used herein, refers to a material, including but not limited, to a salt, carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively nontoxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained. The pharmaceutically acceptable carriers (vehicles) useful in this disclosure are conventional. Page 30 of 203 1097657169\1\AMERICAS Remington's Pharmaceutical Sciences, by E. W. Martin, Mack Publishing Co., Easton, Pa., 19th Edition (1995), describes compositions and formulations suitable for pharmaceutical delivery of one or more agents, such as one or more modulatory agents. In general, the nature of the carrier will depend on the particular mode of administration being employed. For instance, parenteral formulations can include injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle. In addition to biologically-neutral carriers, pharmaceutical agents to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate, sodium lactate, potassium chloride, calcium chloride, and triethanolamine oleate. For example, the present invention provides a pharmaceutical composition comprising a pharmaceutically acceptable excipient and an, e.g., T cell, preferably a γδ T cell, engineered to express a CAR directed to B7H6, as described herein. [00134] “Encoding” refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom. Thus, a gene encodes a protein if transcription and translation of mRNA corresponding to that gene produces the protein in a cell or other biological system. Both the coding strand, the nucleotide sequence of which is identical to the mRNA sequence and is usually provided in sequence listings, and the non-coding strand, used as the template for transcription of a gene or cDNA, can be referred to as encoding the protein or other product of that gene or cDNA. [00135] “Isolated” means altered or removed from the natural state. For example, a nucleic acid or a peptide naturally present in a living animal is not “isolated,” but the same nucleic acid or peptide partially or completely separated from the coexisting materials of its natural state is “isolated.” An isolated nucleic acid or protein can exist in substantially purified form, or can exist in a non-native environment such as, for example, a host cell. [00136] Unless otherwise specified, a “nucleotide sequence encoding an amino acid sequence” includes all nucleotide sequences that are degenerate versions of each other and that encode the Page 31 of 203 1097657169\1\AMERICAS same amino acid sequence. Nucleotide sequences that encode proteins and RNA may include introns. [00137] The terms “patient,” “subject,” “individual,” and the like are used interchangeably herein, and refer to any animal, amenable to the methods described herein. In certain non-limiting embodiments, the patient, subject or individual is a human. [00138] “Expression cassette” refers to a nucleic acid comprising expression control sequences operatively linked to a nucleic acid encoding a transcript or polypeptide to be expressed. An expression cassette comprises sufficient cis-acting elements for expression; other elements for expression can be supplied by the host cell or in an in vitro expression system. Expression cassettes can be a component of a vector such as a cosmid, a plasmid (e.g., naked or contained in a liposome), or a virus (e.g., lentivirus, retrovirus, adenovirus, and adeno-associated virus). An expression cassette can be in a host cell, such as a γδ T cell. II. Compositions and Methods of the Invention A. Anti-B7H6 Antibodies [00139] In one embodiment, the present invention provides anti-B7H6 antibodies which may find use herein as therapeutic agents. Exemplary antibodies include polyclonal, monoclonal, chimeric, humanized, and human antibodies. 1. Polyclonal Antibodies [00140] Polyclonal antibodies may be raised in animals by multiple subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen and an adjuvant. It may be useful to conjugate the relevant antigen (especially when synthetic peptides are used) to a protein that is immunogenic in the species to be immunized. For example, the antigen can be conjugated to keyhole limpet hemocyanin (KLH), serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor, using a bifunctional or derivatizing agent, e.g., maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (through lysine residues), glutaraldehyde, succinic anhydride, SOCl2, or R′N═C═NR, where R and R1 are different alkyl groups. [00141] Animals are immunized against the antigen, immunogenic conjugates, or derivatives by combining, e.g., 100 μg or 5 μg of the protein or conjugate (for rabbits or mice, respectively) with 3 volumes of Freund’s complete adjuvant and injecting the solution intradermally at multiple Page 32 of 203 1097657169\1\AMERICAS sites. One month later, the animals are boosted with ⅕ to 1/10 the original amount of peptide or conjugate in Freund’s complete adjuvant by subcutaneous injection at multiple sites. Seven to 14 days later, the animals are bled and the serum is assayed for antibody titer. Animals are boosted until the titer plateaus. Conjugates also can be made in recombinant cell culture as protein fusions. Also, aggregating agents such as alum are suitably used to enhance the immune response. 2. Monoclonal Antibodies [00142] A monoclonal antibody (mAb) to an antigen-of-interest can be prepared by using any technique known in the art. These include, but are not limited to, the hybridoma technique originally described by Kohler and Milstein (1975, Nature 256, 495-497), the human B cell hybridoma technique (Kozbor et al., 1983, Immunology Today 4: 72), and the EBV-hybridoma technique (Cole et al., 1985, Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96). The Selected Lymphocyte Antibody Method (SLAM) (Babcook, J.S., et al., A novel strategy for generating monoclonal antibodies from single, isolated lymphocytes producing antibodies of defined specificities. Proc Natl Acad Sci U S A, 1996. 93 (15): p. 7843-8. ) and (McLean G et al., 2005, J Immunol. 174(8): 4768-78. Such antibodies may be of any immunoglobulin class including IgG, IgM, IgE, IgA, and IgD and any subclass thereof. The hybridoma producing the mAbs of use in this invention may be cultivated in vitro or in vivo. [00143] Monoclonal antibodies may be made using the hybridoma method first described by Kohler et al., Nature, 256: 495 (1975), or may be made by recombinant DNA methods (U.S. Pat. No.4,816,567). [00144] In the hybridoma method, a mouse or other appropriate host animal, such as a hamster, is immunized as described above to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization. Alternatively, lymphocytes may be immunized in vitro. After immunization, lymphocytes are isolated and then fused with a myeloma cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp.59-103 (Academic Press, 1986)). [00145] The hybridoma cells thus prepared are seeded and grown in a suitable culture medium which may contain one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells (also referred to as fusion partner). For example, if the parental myeloma Page 33 of 203 1097657169\1\AMERICAS cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the selective culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells. [00146] Preferred fusion partner myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a selective medium that selects against the unfused parental cells. Preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, Calif. USA, and SP-2 and derivatives e.g., X63-Ag8-653 cells available from the American Type Culture Collection, Manassas, Va., USA. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor, J. Immunol., 133: 3001 (1984); and Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp.51-63 (Marcel Dekker, Inc., New York, 1987)). [00147] Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen. Preferably, the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunosorbent assay (ELISA). [00148] The binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis described in Munson et al., Anal. Biochem.107: 220 (1980). [00149] Once hybridoma cells that produce antibodies of the desired specificity, affinity, and/or activity are identified, the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)). Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium. In addition, the hybridoma cells may be grown in vivo as ascites tumors in an animal, e.g., by intraperitoneal injection of the cells into mice. [00150] The monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by conventional antibody purification procedures such as, for example, affinity chromatography (e.g., using protein A or protein G-Sepharose) or ion- exchange chromatography, hydroxylapatite chromatography, gel electrophoresis, dialysis, etc. Page 34 of 203 1097657169\1\AMERICAS [00151] DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). The hybridoma cells serve as a preferred source of such DNA. Once isolated, the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese Hamster Ovary (CHO) cells, or myeloma cells that do not otherwise produce antibody protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. Review articles on recombinant expression in bacteria of DNA encoding the antibody include Skerra et al., Curr. Opinion in Immunol.5: 256-62 (1993) and Plückthun, Immunol. Rev.130: 151- 88 (1992). [00152] In a further embodiment, monoclonal antibodies or antibody fragments can be isolated from antibody phage libraries generated using the techniques described in McCafferty et al., Nature, 348: 552-54 (1990). Clackson et al., Nature, 352: 624-28 (1991) and Marks et al., J. Mol. Biol., 222: 581-97 (1991) describe the isolation of murine and human antibodies, respectively, using phage libraries. Subsequent publications describe the production of high affinity (nM range) human antibodies by chain shuffling (Marks et al., Bio/Technology, 10: 779-83 (1992)), as well as combinatorial infection and in vivo recombination as a strategy for constructing very large phage libraries (Waterhouse et al., Nuc. Acids. Res.21: 2265-6 (1993)). Thus, these techniques are viable alternatives to traditional monoclonal antibody hybridoma techniques for isolation of monoclonal antibodies. [00153] The DNA that encodes the antibody may be modified to produce chimeric or fusion antibody polypeptides, for example, by substituting human heavy chain and light chain constant domain (CH and CO sequences for the homologous murine sequences (U.S. Pat. No.4,816,567; and Morrison, et al., Proc. Natl. Acad. Sci. USA, 81: 6851 (1984)), or by fusing the immunoglobulin coding sequence with all or part of the coding sequence for a non- immunoglobulin polypeptide (heterologous polypeptide). The non-immunoglobulin polypeptide sequences can substitute for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen. Page 35 of 203 1097657169\1\AMERICAS 3. Chimeric, Humanized, and Human Antibodies [00154] In embodiments, the anti-B7H6 antibody is a chimeric antibody. Certain chimeric antibodies are described, e.g., in U.S. Pat. No.4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA, 81: 6851-5 (1984)). In one example, a chimeric antibody comprises a non-human variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) and a human constant region. In a further example, a chimeric antibody is a “class switched” antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen-binding fragments thereof. [00155] In embodiments, a chimeric antibody is a humanized antibody. Typically, a non-human antibody is humanized to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody. Generally, a humanized antibody comprises one or more variable domains in which HVRs, e.g., CDRs, (or portions thereof) are derived from a non- human antibody, and FRs (or portions thereof) are derived from human antibody sequences. A humanized antibody optionally will also comprise at least a portion of a human constant region. In embodiments, some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the CDR residues are derived), e.g., to restore or improve antibody specificity or affinity. [00156] The anti-B7H6 antibodies of the invention may comprise humanized antibodies or human antibodies. Humanized forms of non-human (e.g., murine or rabbit) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab')2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity. In some instances, Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues. Humanized antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all Page 36 of 203 1097657169\1\AMERICAS or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin (Jones et al., Nature, 321: 522-5 (1986); Riechmann et al., Nature, 332: 323-9 (1988); and Presta, Curr. Op. Struct. Biol., 2: 593-6 (1992)). [00157] A humanized antibody of the invention may comprise one or more human and/or human consensus non-hypervariable region (e.g., framework) sequences in its heavy and/or light chain variable domain. In embodiments, one or more additional modifications are present within the human and/or human consensus non-hypervariable region sequences. In one embodiment, the heavy chain variable domain of an antibody of the invention comprises a human consensus framework sequence, which in one embodiment is the subgroup III consensus framework sequence. In one embodiment, an antibody of the invention comprises a variant subgroup III consensus framework sequence modified at least one amino acid position. [00158] As is known in the art, the amino acid position/boundary delineating a hypervariable region of an antibody can vary, depending on the context and the various definitions known in the art. Some positions within a variable domain may be viewed as hybrid hypervariable positions in that these positions can be deemed to be within a hypervariable region under one set of criteria while being deemed to be outside a hypervariable region under a different set of criteria. One or more of these positions can also be found in extended hypervariable regions (as further defined below). The invention provides antibodies comprising modifications in these hybrid hypervariable positions. In one embodiment, these hypervariable positions include one or more positions 26-30, 33-35B, 47-49, 57-65, 93, 94 and 101-102 in a heavy chain variable domain. In one embodiment, these hybrid hypervariable positions include one or more of positions 24-29, 35-36, 46-49, 56 and 97 in a light chain variable domain. In one embodiment, an antibody of the invention comprises a human variant human subgroup consensus framework sequence modified at one or more hybrid hypervariable positions. [00159] An antibody of the invention can comprise any suitable human or human consensus light chain framework sequences, provided the antibody exhibits the desired biological characteristics (e.g., a desired binding affinity). In one embodiment, an antibody of the invention comprises at least a portion (or all) of the framework sequence of human ^ light chain. In one Page 37 of 203 1097657169\1\AMERICAS embodiment, an antibody of the invention comprises at least a portion (or all) of human ^ subgroup I framework consensus sequence. [00160] Methods for humanizing non-human antibodies are well known in the art. As discussed, a humanized antibody generally has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as “import” residues, which are typically taken from an “import” variable domain. Humanization can be essentially performed following the method of Winter and co-workers (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting rodent CDRs for CDR sequences for the corresponding sequences of a human antibody. Accordingly, such “humanized” antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies. [00161] The choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is very important to reduce antigenicity and HAMA response (human anti- mouse antibody) when the antibody is intended for human therapeutic use. Reduction or elimination of a HAMA response is a significant aspect of clinical development of suitable therapeutic agents (see, e.g., Khaxzaeli et al., J. Natl. Cancer Inst. (1988), 80:937; Jaffers et al., Transplantation (1986), 41:572; Shawler et al., J. Immunol. (1985), 135:1530; Sears et al., J. Biol. Response Mod. (1984), 3:138; Miller et al., Blood (1983), 62:988; Hakimi et al., J. Immunol. (1991), 147:1352; Reichmann et al., Nature (1988), 332: 323; Junghans et al., Cancer Res. (1990), 50: 1495). As described herein, the invention provides antibodies that are humanized such that HAMA response is reduced or eliminated. Variants of these antibodies can further be obtained using routine methods known in the art, some of which are further described below. According to the so-called “best-fit” method, the sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable domain sequences. The human V domain sequence which is closest to that of the rodent is identified and the human framework region (FR) within it accepted for the humanized antibody (Sims et al., J. Immunol. 151: 2296 (1993); Chothia et al., J. Mol. Biol., 196: 901 (1987)). Another method uses a particular framework Page 38 of 203 1097657169\1\AMERICAS region derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same framework may be used for several different humanized antibodies (Carter et al., Proc. Natl. Acad. Sci. USA, 89: 4285 (1992); Presta et al., J. Immunol. 151: 2623 (1993)). [00162] For example, an amino acid sequence from an antibody as described herein can serve as a starting (parent) sequence for diversification of the framework and/or hypervariable sequence(s). A selected framework sequence to which a starting hypervariable sequence is linked is referred to herein as an acceptor human framework. While the acceptor human frameworks may be from, or derived from, a human immunoglobulin (the VL and/or VH regions thereof), preferably the acceptor human frameworks are from, or derived from, a human consensus framework sequence as such frameworks that have been demonstrated to have minimal, or no, immunogenicity in human patients. [00163] Where the acceptor is derived from a human immunoglobulin, one may optionally select a human framework sequence that is selected based on its homology to the donor framework sequence by aligning the donor framework sequence with various human framework sequences in a collection of human framework sequences, and select the most homologous framework sequence as the acceptor. [00164] In one embodiment, human consensus frameworks herein are from, or derived from, VH subgroup III and/or VL kappa subgroup I consensus framework sequences. [00165] While the acceptor may be identical in sequence to the human framework sequence selected, whether that be from a human immunoglobulin or a human consensus framework, the present invention contemplates that the acceptor sequence may comprise pre-existing amino acid substitutions relative to the human immunoglobulin sequence or human consensus framework sequence. These pre-existing substitutions are preferably minimal; usually four, three, two or one amino acid differences only relative to the human immunoglobulin sequence or consensus framework sequence. [00166] Hypervariable region residues of the non-human antibody are incorporated into the VL and/or VH acceptor human frameworks. For example, one may incorporate residues corresponding to the Kabat CDR residues, the Chothia hypervariable loop residues, the Abm residues, and/or contact residues. Optionally, the extended hypervariable region residues as follows are Page 39 of 203 1097657169\1\AMERICAS incorporated: 24-34 (L1), 50-56 (L2) and 89-97 (L3), 26-35B (H1), 50-65, 47-65 or 49-65 (H2) and 93-102, 94-102, or 95-102 (H3). [00167] While “incorporation” of hypervariable region residues is discussed herein, it will be appreciated that this can be achieved in various ways, for example, nucleic acid encoding the desired amino acid sequence can be generated by mutating nucleic acid encoding the mouse variable domain sequence so that the framework residues thereof are changed to acceptor human framework residues, or by mutating nucleic acid encoding the human variable domain sequence so that the hypervariable domain residues are changed to non-human residues, or by synthesizing nucleic acid encoding the desired sequence, etc. [00168] As described herein, hypervariable region-grafted variants may be generated by Kunkel mutagenesis of nucleic acid encoding the human acceptor sequences, using a separate oligonucleotide for each hypervariable region. Kunkel et al., Methods Enzymol. 154:367-382 (1987). Appropriate changes can be introduced within the framework and/or hypervariable region, using routine techniques, to correct and re-establish proper hypervariable region-antigen interactions. [00169] Phage(mid) display (also referred to herein as phage display in some contexts) can be used as a convenient and fast method for generating and screening many different potential variant antibodies in a library generated by sequence randomization. However, other methods for making and screening altered antibodies are available to the skilled person. [00170] Phage(mid) display technology has provided a powerful tool for generating and selecting novel proteins which bind to a ligand, such as an antigen. Using the techniques of phage(mid) display allows the generation of large libraries of protein variants which can be rapidly sorted for those sequences that bind to a target molecule with high affinity. Nucleic acids encoding variant polypeptides are generally fused to a nucleic acid sequence encoding a viral coat protein, such as the gene III protein or the gene VIII protein. Monovalent phagemid display systems where the nucleic acid sequence encoding the protein or polypeptide is fused to a nucleic acid sequence encoding a portion of the gene III protein have been developed. (Bass, S., Proteins, 8:309 (1990); Lowman and Wells, Methods: A Companion to Methods in Enzymology, 3:205 (1991)). In a monovalent phagemid display system, the gene fusion is expressed at low levels and wild type gene III proteins are also expressed so that infectivity of the particles is retained. Methods of Page 40 of 203 1097657169\1\AMERICAS generating peptide libraries and screening those libraries have been disclosed in many patents (e.g., U.S. Pat. No.5,723,286, U.S. Pat. No.5,432,018, U.S. Pat. No.5,580,717, U.S. Pat. No.5,427,908 and U.S. Pat. No.5,498,530). [00171] Libraries of antibodies or antigen binding polypeptides have been prepared in a number of ways including by altering a single gene by inserting random DNA sequences or by cloning a family of related genes. Methods for displaying antibodies or antigen binding fragments using phage(mid) display have been described in U.S. Pat. Nos. 5,750,373, 5,733,743, 5,837,242, 5,969,108, 6,172,197, 5,580,717, and 5,658,727. The library is then screened for expression of antibodies or antigen binding proteins with the desired characteristics. [00172] Methods of substituting an amino acid of choice into a template nucleic acid are well established in the art, some of which are described herein. For example, methods for introducing modifications into nucleic acid sequences can include the use of various kits available for purchase (e.g., QuickChange Site Directed Mutagenesis Kit, Agilent, Santa Clara, CA). As another example, hypervariable region residues can be substituted using the Kunkel method (e.g., Kunkel et al., Methods Enzymol.154:367-382 (1987)). [00173] It is important that antibodies be humanized with retention of high binding affinity for the antigen and other favorable biological properties. To achieve this goal, according to a preferred method, humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences. Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen. In this way, FR residues can be selected and combined from the recipient and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved. In general, the hypervariable region residues are directly and most substantially involved in influencing antigen binding. Page 41 of 203 1097657169\1\AMERICAS [00174] Various forms of a humanized anti-B7H6 antibody are contemplated. For example, the humanized antibody may be an antibody fragment, such as a Fab. Alternatively, the humanized antibody may be an intact antibody, such as an intact IgG1 antibody. [00175] As an alternative to humanization, human antibodies can be generated. For example, it is now possible to produce transgenic animals (e.g., mice) that are capable, upon immunization, of producing a full repertoire of human antibodies in the absence of endogenous immunoglobulin production. For example, it has been described that the homozygous deletion of the antibody heavy-chain joining region (JH) gene in chimeric and germ-line mutant mice results in complete inhibition of endogenous antibody production. Transfer of the human germ-line immunoglobulin gene array into such germ-line mutant mice will result in the production of human antibodies upon antigen challenge (see, e.g., Jakobovits et al., Proc. Natl. Acad. Sci. USA, 90: 2551 (1993); Jakobovits et al., Nature, 362: 255-8 (1993); Bruggemann et al., Year in Immuno. 7: 33 (1993); U.S. Pat. Nos.5,545,806, 5,569,825, 5,591,669; 5,545,807; and WO 97/17852). [00176] Alternatively, phage display technology (McCafferty et al., Nature 348: 552-53 (1990)) can be used to produce human antibodies and antibody fragments in vitro, from immunoglobulin variable (V) domain gene repertoires from unimmunized donors. According to this technique, antibody V domain genes are cloned in-frame into either a major or minor coat protein gene of a filamentous bacteriophage, such as M13 or fd, and displayed as functional antibody fragments on the surface of the phage particle. Because the filamentous particle contains a single-stranded DNA copy of the phage genome, selections based on the functional properties of the antibody also result in selection of the gene encoding the antibody exhibiting those properties. Thus, the phage mimics some of the properties of the B-cell. Phage display can be performed in a variety of formats, reviewed in, e.g., Johnson, Kevin S, and Chiswell, David J., Current Opinion in Structural Biology 3:564-571 (1993). Several sources of V-gene segments can be used for phage display. Clackson et al., Nature, 352:624-628 (1991) isolated a diverse array of anti-oxazolone antibodies from a small random combinatorial library of V genes derived from the spleens of immunized mice. A repertoire of V genes from unimmunized human donors can be constructed and antibodies to a diverse array of antigens (including self-antigens) can be isolated essentially following the techniques described by Marks et al., J. Mol. Biol. 222:581-97 (1991), or Griffith et al., EMBO J. 12: 725-34 (1993) (see also, U.S. Pat. Nos.5,565,332 and 5,573,905). Page 42 of 203 1097657169\1\AMERICAS [00177] Human antibodies may also be generated by in vitro activated B cells (see, e.g., U.S. Pat. Nos.5,567,610 and 5,229,275). [00178] Accordingly, in embodiments, human monoclonal antibodies directed against B7H6 may be generated using transgenic or transchromosomic mice carrying parts of the human immune system rather than the mouse system. [00179] The HuMAb Mouse™ (Medarex, Inc.) contains human immunoglobulin gene miniloci that encode unrearranged human heavy (µ and γ) and κ light chain immunoglobulin sequences, together with targeted mutations that inactivate the endogenous µ and κ chain loci (see e.g., Lonberg, et al. (1994) Nature 368(6474): 856-9). Accordingly, the mice exhibit reduced expression of mouse IgM or κ, and in response to immunization, the introduced human heavy and light chain transgenes undergo class switching and somatic mutation to generate high affinity human IgGκ monoclonal antibodies (Lonberg, N. et al. (1994), supra; reviewed in Lonberg, N. (1994) Handbook of Experimental Pharmacology 113: 49-101; Lonberg, N. and Huszar, D. (1995) Intern. Rev. Immunol. 13: 65-93, and Harding, F. and Lonberg, N. (1995) Ann. N.Y. Acad. Sci. 764: 536-46). Preparation and use of the HuMAb Mouse™, and the genomic modifications carried by such mice, is further described in Taylor, L. et al. (1992) Nucleic Acids Research 20:6287- 6295; Chen, J. et al. (1993) International Immunology 5: 647-656; Tuaillon et al. (1993) Proc. Natl. Acad. Sci. USA 90: 3720-4; Choi et al. (1993) Nature Genetics 4:117-23; Chen, J. et al. (1993) EMBO J. 12: 21-830; Tuaillon et al., (1994) J. Immunol. 152: 2912-20; Taylor, L. et al. (1994) International Immunology 6: 579-91; and Fishwild, D. et al. (1996) Nature Biotechnology 14: 845-51, the contents of all of which are hereby specifically incorporated by reference in their entirety. See further, U.S. Pat. Nos. 5,545,806; 5,569,825; 5,625,126; 5,633,425; 5,789,650; 5,877,397; 5,661,016; 5,814,318; 5,874,299; and 5,770,429; U.S. Pat. No. 5,545,807; PCT Publication Nos. WO 92/03918, WO 93/12227, WO 94/25585, WO 97/13852, WO 98/24884 and WO 99/45962; and PCT Publication No. WO 01/14424. [00180] In another embodiment, human antibodies of this disclosure can be raised using a mouse referred to as “KM Mouse™” that carries human immunoglobulin sequences on transgenes and transchromosomes, as described in detail in PCT Publication WO 02/43478. Page 43 of 203 1097657169\1\AMERICAS [00181] In another embodiment, an alternative transgenic system referred to as the Xenomouse (Abgenix, Inc.) can be used; such mice are described in, for example, U.S. Pat. Nos. 5,939,598; 6,075,181; 6,114,598; 6,150,584 and 6,162,963. [00182] Additional relevant transchromosomic animal systems expressing human immunoglobulin genes are available in the art and can be used to raise anti-B7H6 antibodies of this disclosure. For example, mice carrying both a human heavy chain transchromosome and a human light chain tranchromosome, referred to as “TC mice” can be used; such mice are described in Tomizuka et al. (2000) Proc. Natl. Acad. Sci. USA 97: 722-7. As another example, cows carrying human heavy and light chain transchromosomes have been described in the art (e.g., Kuroiwa et al. (2002) Nature Biotechnology 20: 889-94 and PCT application No. WO 2002/092812) and can be used to raise anti-B7H6 antibodies of this disclosure. Additional examples of transgenic animals that can be used to produce anti-B7H6 antibodies include OmniRatTM and OmniMouseTM (see e.g., Osborn M., et al. (2013) Journal of Immunology 190: 1481-90; Ma B., et al. (2013) Journal of Immunological Methods 400-401: 78-86; Geurts A., et al. (2009) Science 325: 433, U.S. Pat. No. 8,907,157; European Pat. No. 2152880B1; European Pat. No. 2336329B1). Yet another example includes the use of VELOCIMMUNE® Technology (see, for example, U.S. Pat. No. 6,596,541, Regeneron Pharmaceuticals, VELOCIMMUNE®. Briefly, the VELOCIMMUNE® technology involves generation of a transgenic mouse having a genome comprising human heavy and light chain variable regions operably linked to endogenous mouse constant region loci such that the mouse produces an antigen-binding protein, e.g., antibody, comprising a human variable region and a mouse constant region in response to antigenic stimulation. The DNA encoding the variable regions of the heavy and light chains of the antibody are isolated and operably linked to DNA encoding the human heavy and light chain constant regions. The DNA is then expressed in a cell capable of expressing the fully human antibody. 4. Antibody Fragments [00183] Embodiments of the present disclosure encompass antibody fragments. [00184] Various techniques have been developed for the production of antibody fragments. Traditionally, these fragments were derived via proteolytic digestion of intact antibodies (see, e.g., Morimoto et al., Journal of Biochemical and Biophysical Methods 24:107-7 (1992); and Brennan et al., Science, 229: 81 (1985)). However, these fragments can now be produced directly by Page 44 of 203 1097657169\1\AMERICAS recombinant host cells. Fab, Fv and scFv antibody fragments can all be expressed in and secreted from E. coli, thus allowing the facile production of large amounts of these fragments. Antibody fragments can be isolated from the antibody phage libraries discussed above. Alternatively, Fab′- SH fragments can be directly recovered from E. coli and chemically coupled to form F(ab′)2 fragments (Carter et al., Bio/Technology 10:163-7 (1992)). According to another approach, F(ab′)2 fragments can be isolated directly from recombinant host cell culture. Fab and F(ab′)2 fragment with increased in vivo half-life comprising salvage receptor binding epitope residues are described in U.S. Pat. No. 5,869,046. Other techniques for the production of antibody fragments will be apparent to the skilled practitioner. In other embodiments, the antibody of choice is a single chain Fv fragment (scFv) (see WO 93/16185; U.S. Pat. No.5,571,894; and U.S. Pat. No.5,587,458). Fv and sFv are the only species with intact combining sites that are devoid of constant regions; thus, they are suitable for reduced nonspecific binding during in vivo use. sFv fusion proteins may be constructed to yield fusion of an effector protein at either the amino or the carboxy terminus of an sFv (see Antibody Engineering, ed. Borrebaeck, supra. The antibody fragment may also be a “linear antibody”, e.g., as described in U.S. Pat. No.5,641,870 for example. [00185] In one embodiment, an anti-B7H6 antibody derived scFv is used in a CAR of the present disclosure. Included among anti-B7H6 antibody fragments are portions of anti-B7H6 antibodies (and combinations of portions of anti-B7H6 antibodies, for example, scFv) that may be used as targeting arms, directed to a B7H6 epitope, in CARs and CAR modified immune cells of the present disclosure. Such fragments are not necessarily proteolytic fragments but rather portions of polypeptide sequences that can confer affinity for target. 5. Multispecific Antibodies [00186] In any aspect of the present disclosure, an anti-B7H6 antibody provided herein is a multispecific antibody, for example, a bispecific antibody. Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes. Exemplary bispecific antibodies may bind to two different epitopes of a B7H6 protein as described herein. Other such antibodies may combine a B7H6 binding site with a binding site for another protein. In some examples, an anti-B7H6 arm may be combined with an arm which binds to a triggering molecule on a leukocyte such as a T cell receptor molecule (e.g., CD3), or Fc receptors for IgG (FcγR), such as FcγRI (CD64), FcγRII (CD32) and FcγRIII (CD16), so as to focus and localize cellular defense Page 45 of 203 1097657169\1\AMERICAS mechanisms to the B7H6-expressing cell. Bispecific antibodies may also be used to localize cytotoxic agents to cells which express B7H6. These antibodies possess a B7H6-binding arm and an arm which binds the cytotoxic agent (e.g., saporin, anti-interferon-α, vinca alkaloid, ricin A chain, methotrexate or radioactive isotope hapten). Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g., F(ab')2 bispecific antibodies). [00187] Methods for making bispecific antibodies are known in the art. Traditional production of full length bispecific antibodies is based on the co-expression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities (Millstein et al., Nature 305: 537-9 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity chromatography steps, is rather cumbersome, and the product yields are low. Similar procedures are disclosed in WO 93/08829, and in Traunecker et al., EMBO J. 10:3655-3659 (1991). [00188] Other approaches for making bispecific antibodies are known. One approach is the “knobs-into-holes” or “protuberance-into-cavity” approach (see, e.g., U.S. Pat. No.5,731,168). In this approach, two immunoglobulin polypeptides (e.g., heavy chain polypeptides) each comprise an interface. An interface of one immunoglobulin polypeptide interacts with a corresponding interface on the other immunoglobulin polypeptide, thereby allowing the two immunoglobulin polypeptides to associate. These interfaces may be engineered such that a “knob” or “protuberance” (these terms may be used interchangeably herein) located in the interface of one immunoglobulin polypeptide corresponds with a “hole” or “cavity” (these terms may be used interchangeably herein) located in the interface of the other immunoglobulin polypeptide. In embodiments, the hole is of identical or similar size to the knob and suitably positioned such that when the two interfaces interact, the knob of one interface is positionable in the corresponding hole of the other interface. Without wishing to be bound to theory, this is thought to stabilize the heteromultimer and favor formation of the heteromultimer over other species, for example homomultimers. In embodiments, this approach may be used to promote the heteromultimerization of two different immunoglobulin polypeptides, creating a bispecific antibody comprising two immunoglobulin polypeptides with binding specificities for different epitopes. Page 46 of 203 1097657169\1\AMERICAS [00189] According to a different approach, antibody variable domains with the desired binding specificities (antibody-antigen combining sites) are fused to immunoglobulin constant domain sequences. The fusion preferably is with an immunoglobulin heavy chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is typical to have the first heavy- chain constant region (CH1) containing the site necessary for light chain binding, present in at least one of the fusions. DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co- transfected into a suitable host organism. This provides for great flexibility in adjusting the mutual proportions of the three polypeptide fragments in embodiments when unequal ratios of the three polypeptide chains used in the construction provide the optimum yields. It is, however, possible to insert the coding sequences for two or all three polypeptide chains in one expression vector when the expression of at least two polypeptide chains in equal ratios results in high yields or when the ratios are of no particular significance. [00190] In one embodiment of this approach, the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. It was found that this asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, as the presence of an immunoglobulin light chain in only one half of the bispecific molecule provides for a facile way of separation. This approach is disclosed in WO 94/04690. For further details of generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 121:210 (1986). [00191] According to another approach described in WO96/27011, the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture. One interface comprises at least a part of the CH 3 domain of an antibody constant domain. In this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan). Compensatory “cavities” of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers. Page 47 of 203 1097657169\1\AMERICAS [00192] Bispecific antibodies include cross-linked or “heteroconjugate” antibodies. For example, one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Pat. No.4,676,980), and for treatment of HIV infection (WO 91/00360, WO 92/200373, and EP 03089). Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in U.S. Pat. No. 4,676,980, along with a number of cross-linking techniques. [00193] Techniques for generating bispecific antibodies from antibody fragments have also been described in the literature. For example, bispecific antibodies can be prepared using chemical linkage. Brennan et al., Science, 229: 81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab′)2 fragments. These fragments are reduced in the presence of the dithiol complexing agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The Fab′ fragments generated are then converted to thionitrobenzoate (TNB) derivatives. One of the Fab′-TNB derivatives is then reconverted to the Fab′-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab′-TNB derivative to form the bispecific antibody. The bispecific antibodies produced can be used as agents for the selective immobilization of enzymes. Shalaby et al., J. Exp. Med., 175: 217-225 (1992) describe the production of a fully humanized bispecific antibody F(ab′)2 molecule. Each Fab′ fragment was separately secreted from E. coli and subjected to directed chemical coupling in vitro to form the bispecific antibody. [00194] Various techniques for making and isolating bispecific antibody fragments directly from recombinant cell culture have also been described. For example, bispecific antibodies have been produced using leucine zippers. Kostelny et al., J. Immunol., 148(5):1547-1553 (1992). The leucine zipper peptides from the Fos and Jun proteins were linked to the Fab′ portions of two different antibodies by gene fusion. The antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers. The “diabody” technology described by Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments. The fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the VH and Page 48 of 203 1097657169\1\AMERICAS VL domains of one fragment are forced to pair with the complementary VL and VH domains of another fragment, thereby forming two antigen-binding sites. Another strategy for making bispecific antibody fragments by the use of single-chain Fv (sFv) dimers has also been reported. See Gruber et al, J. Immunol, 152:5368 (1994). [00195] Another technique for making bispecific antibody fragments is the “bispecific T cell engager” or BiTE® approach (see, e.g., WO2004/106381, WO2005/061547, WO2007/042261, and WO2008/119567). This approach utilizes two antibody variable domains arranged on a single polypeptide. For example, a single polypeptide chain includes two single chain Fv (scFv) fragments, each having a variable heavy chain (VH) and a variable light chain (VL) domain separated by a polypeptide linker of a length sufficient to allow intramolecular association between the two domains. This single polypeptide further includes a polypeptide spacer sequence between the two scFv fragments. Each scFv recognizes a different epitope, and these epitopes may be specific for different cell types, such that cells of two different cell types are brought into close proximity or tethered when each scFv is engaged with its cognate epitope. One particular embodiment of this approach includes a scFv recognizing a cell-surface antigen expressed by an immune cell, e.g., a CD3 polypeptide on a T cell, linked to another scFv that recognizes a cell- surface antigen expressed by a target cell, such as a malignant or tumor cell. [00196] As it is a single polypeptide, the bispecific T cell engager may be expressed using any prokaryotic or eukaryotic cell expression system known in the art, e.g., a CHO cell line. However, specific purification techniques (see, e.g., EP1691833) may be necessary to separate monomeric bispecific T cell engagers from other multimeric species, which may have biological activities other than the intended activity of the monomer. In one exemplary purification scheme, a solution containing secreted polypeptides is first subjected to a metal affinity chromatography, and polypeptides are eluted with a gradient of imidazole concentrations. This eluate is further purified using anion exchange chromatography, and polypeptides are eluted using with a gradient of sodium chloride concentrations. Finally, this eluate is subjected to size exclusion chromatography to separate monomers from multimeric species. [00197] Other relevant bispecific antibody fragment formats include but are not limited to dual- affinity re-targeting proteins (DARTs) and Tandem diabodies (TandAbs). A DART is composed of two Fv fragments, with two unique antigen-binding sites formed when two Fv fragments Page 49 of 203 1097657169\1\AMERICAS heterodimerize (Holliger et al., Proc. Natl. Acad. Sci. USA.90:6444–6448 (1993). Specifically, Fv1 consists of a VH from antibody “A” and a VL from antibody “B”, while Fv2 is made from a VH from antibody “B” and VL from antibody “A”. Unlike BiTE antibodies which are connected by a polypeptide linker, this combination allows DART to mimic natural interaction within an IgG molecule. Adding another cysteine residue to the end of each heavy-chain improves stability by forming a C-terminal disulfide bridge. TandAbs are tetravalent bispecific antibodies provide two binding sites for each antigen to maintain the avidity of a natural bivalent antibody. Moreover, TandAbs have a molecular weight (approximately 105 kDa) exceeding the first-pass renal clearance threshold, thus offering a longer half-life compared to smaller antibody constructs (Reusch et al., Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res.22:5829–5838 (2016); Reusch et al., MAbs. 6:728–739 (2014); Compte et al., Oncoimmunology. 3:e28810 (2014). For a recent review of common formats of bispecific antibodies including scFv-based and full-length IgG-like asymmetric antibodies, including methods of production thereof, see Wang et al., Antibodies (Basel), 8(3): 43 (2019). 6. Antibody Variants and Modifications a) Substitution, Insertion, and Deletion Variants [00198] In addition to the anti-B7H6 antibodies described herein, it is contemplated that anti- B7H6 antibody variants can be prepared. Anti-B7H6 antibody variants can be prepared by introducing appropriate nucleotide changes into the encoding DNA, and/or by synthesis of the desired antibody or polypeptide. Those skilled in the art will appreciate that amino acid changes may alter post-translational processes of the anti-B7H6 antibody, such as changing the number or position of glycosylation sites or altering the membrane anchoring characteristics. [00199] Variations in the anti-B7H6 antibodies described herein, can be made, for example, using any of the techniques and guidelines for conservative and non-conservative mutations set forth, for instance, in U.S. Patent No. 5,364,934. Variations may be a substitution, deletion or insertion of one or more codons encoding the antibody or polypeptide that results in a change in the amino acid sequence as compared with the native sequence antibody or polypeptide. Optionally the variation is by substitution of at least one amino acid with any other amino acid in one or more of the domains of the anti-B7H6 antibody. Guidance in determining which amino acid residue may be inserted, substituted or deleted without adversely affecting the desired activity may be found Page 50 of 203 1097657169\1\AMERICAS by comparing the sequence of the anti-B7H6 antibody with that of homologous known protein molecules and minimizing the number of amino acid sequence changes made in regions of high homology. Amino acid substitutions can be the result of replacing one amino acid with another amino acid having similar structural and/or chemical properties, such as the replacement of a leucine with a serine, i.e., conservative amino acid replacements. Insertions or deletions may optionally be in the range of about 1 to 5 amino acids. The variation allowed may be determined by systematically making insertions, deletions or substitutions of amino acids in the sequence and testing the resulting variants for activity exhibited by the full-length or mature native sequence. [00200] Anti-B7H6 antibody fragments are provided herein. Such fragments may be truncated at the N-terminus or C-terminus, or may lack internal residues, for example, when compared with a full-length native antibody or protein. Certain fragments lack amino acid residues that are not essential for a desired biological activity of the anti-B7H6 antibody. [00201] Anti-B7H6 antibody fragments may be prepared by any of a number of conventional techniques. Desired peptide fragments may be chemically synthesized. An alternative approach involves generating antibody or polypeptide fragments by enzymatic digestion, e.g., by treating the protein with an enzyme known to cleave proteins at sites defined by particular amino acid residues, or by digesting the DNA with suitable restriction enzymes and isolating the desired fragment. Yet another suitable technique involves isolating and amplifying a DNA fragment encoding a desired antibody or polypeptide fragment, by polymerase chain reaction (PCR). Oligonucleotides that define the desired termini of the DNA fragment are employed at the 5' and 3' primers in the PCR. Preferably, anti-B7H6 antibody fragments share at least one biological and/or immunological activity with the native anti-B7H6 antibody disclosed herein. [00202] In particular embodiments, conservative substitutions of interest are shown in Table 1 under the heading of preferred substitutions. If such substitutions result in a change in biological activity, then more substantial changes, denominated exemplary substitutions in Table 1, or as further described below in reference to amino acid classes, are introduced and the products screened. Table 1 Original Exemplary Preferred Page 51 of 203 1097657169\1\AMERICAS Residue Substitutions Substitutions Ala (A) val; leu; ile val Arg (R) lys; gln; asn lys Asn (N) gln; his; lys; arg gln Asp (D) glu glu Cys (C) ser ser Gln (Q) asn asn Glu (E) asp asp Gly (G) pro; ala ala His (H) asn; gln; lys; arg arg Ile (I) leu; val; met; ala; phe; norleucine leu Leu (L) norleucine; ile; val; met; ala; phe ile Lys (K) arg; gln; asn arg Met (M) leu; phe; ile leu Phe (F) leu; val; ile; ala; tyr leu Pro (P) ala ala Ser (S) thr thr Thr (T) ser ser Trp (W) tyr; phe tyr Tyr (Y) trp; phe; thr; ser phe Val (V) ile; leu; met; phe; ala; norleucine leu [00203] Substantial modifications in function or immunological identity of the anti-B7H6 antibody are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. Naturally occurring residues are divided into groups based on common side-chain properties: (1) hydrophobic: norleucine, met, ala, val, leu, ile; Page 52 of 203 1097657169\1\AMERICAS (2) neutral hydrophilic: cys, ser, thr; (3) acidic: asp, glu; (4) basic: asn, gln, his, lys, arg; (5) residues that influence chain orientation: gly, pro; and (6) aromatic: trp, tyr, phe. [00204] Non-conservative substitutions will entail exchanging a member of one of these classes for another class. Such substituted residues also may be introduced into the conservative substitution sites or, more preferably, into the remaining (non-conserved) sites. [00205] The variations can be made using methods known in the art such as oligonucleotide- mediated (site-directed) mutagenesis, alanine scanning, and PCR mutagenesis. Site-directed mutagenesis (Carter et al., Nucl. Acids Res., 13: 4331 (1986); Zoller et al., Nucl. Acids Res., 10: 6487 (1987)), cassette mutagenesis (Wells et al., Gene, 34: 315 (1985)), restriction selection mutagenesis (Wells et al., Philos. Trans. R. Soc. London SerA, 317: 415 (1986)) or other known techniques can be performed on the cloned DNA to produce the anti-B7H6 antibody variant DNA. [00206] Scanning amino acid analysis can also be employed to identify one or more amino acids along a contiguous sequence. Among the preferred scanning amino acids are relatively small, neutral amino acids. Such amino acids include alanine, glycine, serine, and cysteine. Alanine is typically a preferred scanning amino acid among this group because it eliminates the side-chain beyond the beta-carbon and is less likely to alter the main-chain conformation of the variant (Cunningham and Wells, Science, 244: 1081-5 (1989)). Alanine is also typically preferred because it is the most common amino acid. Further, it is frequently found in both buried and exposed positions (Creighton, The Proteins, (W.H. Freeman & Co., N.Y.); Chothia, J. Mol. Biol., 150: 1 (1976)). If alanine substitution does not yield adequate amounts of variant, an isoteric amino acid can be used. [00207] Any cysteine residue not involved in maintaining the proper conformation of the anti- B7H6 antibody also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant crosslinking. Conversely, cysteine bond(s) may be added to the anti-B7H6 antibody to improve its stability (particularly where the antibody is an antibody fragment such as an Fv fragment). Page 53 of 203 1097657169\1\AMERICAS [00208] A particularly preferred type of substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g., a humanized or human antibody). Generally, the resulting variant(s) selected for further development will have improved biological properties relative to the parent antibody from which they are generated. A convenient way for generating such substitutional variants involves affinity maturation using phage display. Briefly, several hypervariable region sites (e.g., 6-7 sites) are mutated to generate all possible amino substitutions at each site. The antibody variants thus generated are displayed in a monovalent fashion from filamentous phage particles as fusions to the gene III product of M13 packaged within each particle. The phage-displayed variants are then screened for their biological activity (e.g., binding affinity) as herein disclosed. In order to identify candidate hypervariable region sites for modification, alanine scanning mutagenesis can be performed to identify hypervariable region residues contributing significantly to antigen binding. Alternatively, or additionally, it may be beneficial to analyze a crystal structure of the antigen-antibody complex to identify contact points between the antibody and B7H6 polypeptide. Such contact residues and neighboring residues are candidates for substitution according to the techniques elaborated herein. Once such variants are generated, the panel of variants is subjected to screening as described herein and antibodies with superior properties in one or more relevant assays may be selected for further development. [00209] Nucleic acid molecules encoding amino acid sequence variants of the anti-B7H6 antibody are prepared by a variety of methods known in the art. These methods include, but are not limited to, isolation from a natural source (in the case of naturally occurring amino acid sequence variants) or preparation by oligonucleotide-mediated (or site-directed) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared variant or a non-variant version of the anti-B7H6 antibody. b) Modifications [00210] Covalent modifications of anti-B7H6 antibodies are included within the scope of this invention. One type of covalent modification includes reacting targeted amino acid residues of an anti-B7H6 antibody with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C- terminal residues of the anti-B7H6 antibody. Derivatization with bifunctional agents is useful, for instance, for crosslinking anti-B7H6 antibody to a water-insoluble support matrix or surface for use in a method for purifying anti-B7H6 antibodies, and vice-versa. Page 54 of 203 1097657169\1\AMERICAS Commonly used crosslinking agents include, e.g., 1,1-bis(diazoacetyl)-2-phenylethane, glutaraldehyde, N-hydroxysuccinimide esters, for example, esters with 4-azidosalicylic acid, homobifunctional imidoesters, including disuccinimidyl esters such as 3,3'- dithiobis(succinimidylpropionate), bifunctional maleimides such as bis-N-maleimido-1,8-octane and agents such as methyl-3-[(p-azidophenyl)dithio]propioimidate. [00211] Other modifications include deamidation of glutaminyl and asparaginyl residues to the corresponding glutamyl and aspartyl residues, respectively, hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the α-amino groups of lysine, arginine, and histidine side chains (T.E. Creighton, Proteins: Structure and Molecular Properties, W.H. Freeman & Co., San Francisco, pp.79-86 (1983)), acetylation of the N-terminal amine, and amidation of any C-terminal carboxyl group. [00212] Another type of covalent modification of the anti-B7H6 antibody included within the scope of this invention comprises altering the native glycosylation pattern of the antibody or polypeptide. “Altering the native glycosylation pattern” is intended for purposes herein to mean deleting one or more carbohydrate moieties found in native sequence anti-B7H6 antibody (either by removing the underlying glycosylation site or by deleting the glycosylation by chemical and/or enzymatic means), and/or adding one or more glycosylation sites that are not present in the native sequence anti-B7H6 antibody. In addition, the phrase includes qualitative changes in the glycosylation of the native proteins, involving a change in the nature and proportions of the various carbohydrate moieties present. [00213] Glycosylation of antibodies and other polypeptides is typically either N-linked or O- linked. N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue. The tripeptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain. Thus, the presence of either of these tripeptide sequences in a polypeptide creates a potential glycosylation site. O-linked glycosylation refers to the attachment of one of the sugars N-aceylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5- hydroxylysine may also be used. Page 55 of 203 1097657169\1\AMERICAS [00214] Addition of glycosylation sites to the anti-B7H6 antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites). The alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original anti-B7H6 antibody (for O-linked glycosylation sites). The anti-B7H6 antibody amino acid sequence may optionally be altered through changes at the DNA level, particularly by mutating the DNA encoding the anti-B7H6 antibody at preselected bases such that codons are generated that will translate into the desired amino acids. [00215] Another means of increasing the number of carbohydrate moieties on the anti-B7H6 antibody is by chemical or enzymatic coupling of glycosides to the polypeptide. Such methods are described in the art, e.g., in WO 87/05330 published 11 September 1987, and in Aplin and Wriston, CRC Crit. Rev. Biochem., pp.259-306 (1981). [00216] Removal of carbohydrate moieties present on the anti-B7H6 antibody may be accomplished chemically or enzymatically or by mutational substitution of codons encoding for amino acid residues that serve as targets for glycosylation. Chemical deglycosylation techniques are known in the art and described, for instance, by Hakimuddin, et al., Arch. Biochem. Biophys., 259:52 (1987) and by Edge et al., Anal. Biochem., 118:131 (1981). Enzymatic cleavage of carbohydrate moieties on polypeptides can be achieved by the use of a variety of endo- and exo- glycosidases as described by Thotakura et al., Meth. Enzymol., 138:350 (1987). c) Fc Region Variants [00217] It may be desirable to modify the antibody of the invention with respect to effector function, e.g., so as to enhance antigen-dependent cell-mediated cyototoxicity (ADCC) and/or complement dependent cytotoxicity (CDC) of the antibody. This may be achieved by introducing one or more amino acid substitutions in an Fc region of the antibody. Alternatively or additionally, cysteine residue(s) may be introduced in the Fc region, thereby allowing interchain disulfide bond formation in this region. The homodimeric antibody thus generated may have improved internalization capability and/or increased complement-mediated cell killing and antibody- dependent cellular cytotoxicity (ADCC) (see Caron et al., J. Exp Med.176: 1191-5 (1992); Shopes, B. J. Immunol.148: 2918-22 (1992). Homodimeric antibodies with enhanced anti-tumor activity may also be prepared using heterobifunctional cross-linkers as described in Wolff et al., Cancer Page 56 of 203 1097657169\1\AMERICAS Research 53: 2560-5 (1993). Alternatively, an antibody can be engineered which has dual Fc regions and may thereby have enhanced complement lysis and ADCC capabilities. See Stevenson et al., Anti-Cancer Drug Design 3: 219-30 (1989). To increase the serum half life of the antibody, one may incorporate a salvage receptor binding epitope into the antibody (especially an antibody fragment) as described in U.S. Patent 5,739,277, for example. As used herein, the term “salvage receptor binding epitope” refers to an epitope of the Fc region of an IgG molecule (e.g., IgG1, IgG2, IgG3, or IgG4) that is responsible for increasing the in vivo serum half-life of the IgG molecule. d) Cysteine Engineered Antibody Variants [00218] In certain embodiments, it may be desirable to create cysteine engineered antibodies, e.g.,“thioMAbs,” in which one or more residues of an antibody are substituted with cysteine residues. In particular embodiments, the substituted residues occur at accessible sites of the antibody. By substituting those residues with cysteine, reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker-drug moieties, to create an immunoconjugate, as described further herein. Cysteine engineered antibodies can be generated as described, e.g, in U.S. Patent No. 7,521,541. e) Immunoconjugates [00219] The presently disclosed subject matter also provides immunoconjugates, which include an antibody, disclosed herein, conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, proteins, peptides, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes. For example, an antibody of the disclosed subject matter can be functionally linked (e.g., by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other binding molecules, such as another antibody, antibody fragment, peptide or binding mimetic. [00220] In certain embodiments, an immunoconjugate is an antibody-drug conjugate (ADC) in which an antibody of the present disclosure is conjugated to one or more drugs, including but not limited to, a maytansinoid (see U.S. Patent Nos.5,208,020, 5,416,064 and European Patent EP 0 425235 Bl); an auristatin such as monomethyl auri statin drug moieties DE and DF (MMAE and Page 57 of 203 1097657169\1\AMERICAS MMAF) (see U.S. Patent Nos. 5,635,483 and 5,780,588, and 7,498,298); a dolastatin; a calicheamicin or derivative thereof (see U.S. Patent Nos. 5,712,374, 5,714,586, 5,739,116, 5,767,285, 5,770,701, 5,770,710, 5,773,001, and 5,877,296; Hinman et al., Cancer Res.53:3336- 3342 (1993); and Lode et al., Cancer Res. 58:2925-2928 (1998)); an anthracycline such as daunomycin or doxorubicin (see Kratz et al., Current Med. Chem.13:477-523 (2006); Jeffrey et al., Bioorganic & Med. Chem. Letters 16:358-362 (2006); Torgov et al., Bioconj. Chem.16:717- 721 (2005); Nagy et al., Proc. Natl. Acad. Sci. USA 97:829-834 (2000); Dubowchik et al., Bioorg. & Med. Chem. Letters 12: 1529-1532 (2002); King et al., J. Med. Chem.45:4336-4343 (2002); and U.S. Patent No. 6,630,579); methotrexate; vindesine; a taxane such as docetaxel, paclitaxel, larotaxel, tesetaxel, and ortataxel; a trichothecene; and CC1065. In certain embodiments, an immunoconjugate includes an antibody as described herein conjugated to an enzymatically active toxin or fragment thereof, including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes. [00221] In certain embodiments, an immunoconjugate includes an antibody, as described herein, conjugated to a radioactive atom to form a radioconjugate. A variety of radioactive isotopes are available for the production of radioconjugates. Non-limiting examples include At211, Ac225, 1131, 1125, Y90, Re186, Re188, Sm153, Bi212, P32, Pb212 and radioactive isotopes of Lu. When a radioconjugate is used for detection, it can include a radioactive atom for scintigraphic studies, for example tc99m or I123, or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, mri), such as iodine-123, iodine-131, indium-111, fluorine-19, carbon-13, nitrogen-15, oxygen-17, gadolinium, manganese or iron. [00222] Conjugates of an antibody fragment and cytotoxic agent can be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP), succinimidyl-4-(N-maleimidomethyl) cyclohexane- 1-carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HC1), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis- diazonium derivatives (such as bis- Page 58 of 203 1097657169\1\AMERICAS (p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate), and bis-active fluorine compounds (such as l,5-difluoro-2, 4-dinitrobenzene). For example, a ricin immunotoxin can be prepared as described in Vitetta et al., Science 238: 1098 (1987). Carbon- 14- labeled 1- isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. The linker can be a“cleavable linker” facilitating release of a cytotoxic drug in the cell. For example, an acid-labile linker, peptidase-sensitive linker, photolabile linker, dimethyl linker or disulfide-containing linker (Chari et al., Cancer Res. 52: 127-131 (1992); U. S. Patent No. 5,208,020) can be used. Non- limiting examples of linkers are disclosed above. The immunuoconjugates disclosed herein expressly contemplate, but are not limited to such conjugates prepared with cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MBS, sulfo- SIAB, sulfo- SMCC, and sulfo-SMPB, and SVSB (succinimidyl-(4-vinylsulfone)benzoate) which are commercially available (e.g ., from Pierce Biotechnology, Inc., Rockford, IL., U.S.A). f) Antibody Fusions [00223] The presently disclosed subject matter also encompasses antibody fusions. For example, proteins can be linked together either through chemical or genetic manipulation using methods known in the art. See, for example, Gillies et al., Proc. Nat’l Acad. Sci. USA 89: 1428- 1432 (1992) and US Patent No.5,650,150. [00224] In one example, the present disclosure encompasses an anti-B7H6 antibody-cytokine fusion protein. In principle, an anti-B7H6 antibody as herein disclosed can be fused to any cytokine via the use of recombinant molecular biological techniques. As one example, the anti- B7H6 antibody may be fused to IL-2 (Gillies, S., Protein Engineering, Design and Selection 26(10): 561-569 (2013); Klein, C. et al., OncoImmunology 6:3 (2017). [00225] In another example, the present disclosure encompasses an anti-B7H6 antibody-T-cell engager fusion protein. Discussed herein, anti-B7H6 antibody-T-cell engager fusion proteins comprise fusions between an anti-B7H6 antibody and a ligand for a receptor expressed on a T cell. Examples of such ligands include but are not limited to CD40L, OX40L, 4-1BBL, CD80/86, ICOSL, and the like. In embodiments, the ligand is fused to an Fc portion of an anti-B7H6 antibody. In embodiments, the ligand is fused to a C-terminus of a light chain of an anti-B7H6 Page 59 of 203 1097657169\1\AMERICAS antibody. Such an approach is described with regard to 4-1BBL (Dafne M. et al., Journal of Immunotherapy 38(8): 714-722 (2008), and a similar approach can be used for generation of other antibody-T-cell engager fusion proteins. B. Recombinant Methods and Compositions [00226] Anti-B7H6 antibodies or antigen-binding fragments of the present disclosure may be produced using recombinant methods and compositions, for example as described in US. Patent No.4,816,567. In embodiments, the invention also provides transformed cells and progeny thereof into which a nucleic acid molecule encoding an antibody or antigen-binding fragment, has been introduced by means of recombinant DNA techniques in vitro, ex vivo or in vivo. The transformed cells, eukaryotic or prokaryotic, may be used to produce recombinant antibody or antibody fragment for purification, or for in situ or secretory expression for various purposes, such as diagnosis or therapy for tumor. The transformed cells can be propagated and the introduced nucleic acid transcribed, or encoded protein expressed. It is understood that a progeny cell may not be identical to the parental cell, since there may be mutations that occur during replication. Transformed cells include but are not limited to prokaryotic and eukaryotic cells such as bacteria, fungi, plant, insect, and animal (e.g., mammalian, including human) cells. The cells may be present in culture, in a cell, tissue or organ ex vivo or present in a subject. In one embodiment, the antibody or antibody fragment is displayed on yeast cell surface; in another embodiment, the antibody or antibody fragment is coated on nanoparticle surface; in another embodiment, the antibody or antibody fragment is displayed on mammalian cell surface, such as T cells, NK cells or other human or other mammalian cells; in another embodiment, the antibody or antibody fragment is produced as secretory protein by yeast, E.coli or mammalian cells. [00227] Typically cell transformation employs a vector. The term "vector," refers to, e.g., a plasmid, virus, such as a viral vector, or other vehicle known in the art that can be manipulated by insertion or incorporation of a nucleic acid, for genetic manipulation (i.e., "cloning vectors"), or can be used to transcribe or translate the inserted polynucleic acid (i.e., "expression vectors"). Such vectors are useful for introducing nucleic acids, including a nucleic acid that encodes an antibody or antibody antigen-binding fragment operably linked with an expression control element, and expressing the encoded protein in vitro (e.g., in solution or in solid phase), in cells or in vivo. Page 60 of 203 1097657169\1\AMERICAS [00228] In one embodiment, the expression vector(s) is(are) transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce an antibody or antigen-binding fragment of the invention. Thus, the invention includes host cells containing polynucleic acid(s) encoding an antibody of the invention (e.g., whole antibody, a heavy or light chain thereof, or portion thereof, or a single chain antibody, or a fragment or variant thereof), operably linked to a heterologous promoter. In other embodiments, for the expression of entire antibody molecules, vectors encoding both the heavy and light chains are co- expressed in the host cell for expression of the entire immunoglobulin molecule. [00229] A variety of host-expression vector systems may be utilized to express the antibody molecules of the invention. Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleic acid coding sequences, express an antibody molecule of the invention in situ. These include, but are not limited to, bacteriophage particles engineered to express antibody fragments or variants thereof (single chain antibodies), microorganisms such as bacteria (e.g., E. coli, B. subtilis) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3, NSO cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter; CMV promoter or EFla promoter). Preferably, bacterial cells such as Escherichia coli, and more preferably, eukaryotic cells, especially for the expression of whole recombinant antibody molecule, are used for the expression of a recombinant antibody molecule. For example, mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., Gene 45: 101 (1986); Cockett et al., Bio/Technology 8:2 (11990); B Page 61 of 203 1097657169\1\AMERICAS ebbington et al., Bio/Techniques 10: 169 (1992); Keen and Hale, Cytotechnology 18:207 (1996)). These references are incorporated in their entireties by reference herein. [00230] A vector used to transform a cell or a host-expression vector generally contains at least an origin of replication for propagation in the cell. Control elements, including expression control elements as set forth herein, present within a vector, are included to facilitate transcription and translation. The term "expression control element" is intended to include, at a minimum, one or more components whose presence can influence expression, and can include components other than or in addition to promoters or enhancers, for example, leader sequences and fusion partner sequences, internal ribosome binding sites (IRES) elements for the creation of multigene, or polycistronic, messages, splicing signal for introns, maintenance of the correct reading frame of the gene to permit in-frame translation of mRNA, polyadenylation signal to provide proper polyadenylation of the transcript of a gene of interest, stop codons, etc. [00231] Vectors can include a selection marker. As is known in the art, "selection marker" means a gene that allows for the selection of cells containing the gene. "Positive selection" refers to a process whereby only cells that contain the selection marker will survive upon exposure to the positive selection. Drug resistance is one example of a positive selection marker; cells containing the marker will survive in culture medium containing the selection drug, and cells which do not contain the marker will die. Such markers include drug resistance genes such as neo, which confers resistance to G418, hygr, which confers resistance to hygromycin, or puro which confers resistance to puromycin, among others. Other positive selection marker genes include genes that allow identification or screening of cells containing the marker. These genes include genes for fluorescent proteins (GFP), the lacZ gene, the alkaline phosphatase gene, and surface markers such as CD8, among others. [00232] Vectors can contain negative selection markers. "Negative selection" refers to a process whereby cells containing a negative selection marker are killed upon exposure to an appropriate negative selection agent. For example, cells which contain the herpes simplex virus-thymidine kinase (HSV-tk) gene (Wigler et al., Cell 11:223 (1977)) are sensitive to the drug gancyclovir (GANC). Similarly, the gpt gene renders cells sensitive to 6-thioxanthine. [00233] Mammalian expression systems further include vectors specifically designed for in vivo and ex vivo expression. Such systems include adeno-associated virus (AAV) vectors (U.S. Page 62 of 203 1097657169\1\AMERICAS Pat. No.5,604,090). AAV vectors have previously been shown to provide expression of Factor IX in humans and in mice at levels sufficient for therapeutic benefit (Kay et al., Nat. Genet.24:257 (2000); Nakai et al., Blood 91 :4600 (1998)). Adenoviral vectors (U.S. Pat. Nos. 5,700,470, 5,731,172 and 5,928,944), herpes simplex virus vectors (U.S. Pat. No. 5,501,979) and retroviral (e.g., lentivirus vectors are useful for infecting dividing as well as non-dividing cells and foamy virues) vectors (U.S. Pat. Nos. 5,624,820, 5,693,508, 5,665,577, 6,013,516 and 5,674,703 and WIPO publications WO92/05266 and W092/14829) and papilloma virus vectors (e.g., human and bovine papilloma virus) have all been employed in gene therapy (U.S. Pat. No.5,719,054). Vectors also include cytomegalovirus (CMV) based vectors (U.S. Pat. No. 5,561,063). Vectors that efficiently deliver genes to cells of the intestinal tract have been developed and also may be used (see, e.g., U.S. Pat. Nos. 5,821,235, 5,786,340 and 6,110,456). In yeast, vectors that facilitate integration of foreign nucleic acid sequences into a chromosome, via homologous recombination, for example, are known in the art and can be used. Yeast artificial chromosomes (YAC) are typically used when the inserted nucleic acids are too large for more conventional vectors (e.g., greater than about 12 kb). [00234] In one embodiment, phagemid vectors for use in the invention include any available in the art suitable for the production of the antibodies/antibody templates/FR libraries of the present invention and include phagemid vectors pCB04, pITl, pIT2, CANTAB 6, pComb 3 HS. Filamentous vectors and methods of phagemid construction are described in, for example, U.S. Pat. No. 6,054,312 and U.S. Pat. No. 6,803,230, each incorporated herein by reference. Bacteriophage display systems involving non-filamentous bacteriophage vectors known as cytoplasmic bacteriophage or lytic phage can also be utilized as described in for example, U.S. Pat. No.5,766,905, incorporated herein by reference. [00235] Suitable bacterial expression constructs for use with the present invention include, but are not limited to the pCAL, pUC, pET, pETBlue™ (Novagen), pBAD, pLEX, pTrcHis2, pSE280, pSE380, pSE420 (Invitrogen), pKK223-2 (Clontech), pTrc99A, pKK223-3, pRIT2T, pMC1871, pEZZ 18 (Pharmacia), pBluescript II SK (Stratagene), pALTER-Exl, pALTER- Ex2, pGEMEX (Promega), pFivE (MBI), pQE (Qiagen) commercially available expression constructs, and their derivatives, and others known in the art. In embodiments of the present invention the construct may also include, a virus, a plasmid, a bacmid, a phagemid, a cosmid, or a bacteriophage. Page 63 of 203 1097657169\1\AMERICAS [00236] The use of liposomes for introducing various compositions into cells, including nucleic acids, is known to those skilled in the art (see, e.g., U.S. Pat. Nos.4,844,904, 5,000,959, 4,863,740, and 4,975,282). A carrier comprising a natural polymer, or a derivative or a hydrolysate of a natural polymer, described in WO 94/20078 and U.S. Pat. No.6,096,291, is suitable for mucosal delivery of molecules, such as polypeptides and polynucleic acids, piperazine based amphilic cationic lipids useful for gene therapy also are known (see, e.g., U.S. Pat. No.5,861,397). Cationic lipid systems also are known (see, e.g., U.S. Pat. No.5,459,127). Accordingly, viral and non-viral vector means of delivery into cells or tissue, in vitro, in vivo and ex vivo are included. [00237] In one embodiment, nucleic acid sequences can be "operably linked", i.e., positioned, to ensure the functioning of an expression control sequence. These expression constructs are typically replicable in the cells either as episomes or as integral parts of the cell's chromosomal DNA, and may contain appropriate origins of replication for the respective prokaryotic strain employed for expression. Commonly, expression constructs contain selection markers, such as for example, tetracycline resistance, ampicillin resistance, kanamycin resistance or chlormaphenicol resistance, facilitating detection and/or selection of those bacterial cells transformed with the desired nucleic acid sequences (see, e.g., U.S. Pat. No.4,704,362). These markers, however, are not exclusionary, and numerous others may be employed, as known to those skilled in the art. In another embodiment of the present invention expression constructs contain both positive and negative selection markers. [00238] Similarly, reporter genes may be incorporated within expression constructs to facilitate identification of transcribed products. Accordingly, in one embodiment of the present invention, reporter genes utilized are selected from the group consisting of β-galactosidase, chloramphenicol acetyl transferase, luciferase and a fluorescent protein. [00239] Prokaryotic promoter sequences regulate expression of the encoded polynucleic acid sequences, and in some embodiments of the present invention, are operably linked to polynucleic acids encoding the polypeptides of this invention. In additional embodiments of the present invention, these promoters are either constitutive or inducible, and provide a means of high and low levels of expression of the polypeptides of this invention, and in some embodiments, for regulated expression of multiple polypeptides of the invention, which in some embodiments are expressed as a fusion protein. Page 64 of 203 1097657169\1\AMERICAS [00240] Many well-known bacterial promoters, including the T7 promoter system, the lactose promoter system, tryptophan (Trp) promoter system, Trc/Tac Promoter Systems, beta-lactamase promoter system, tetA Promoter systems, arabinose regulated promoter system, Phage T5 Promoter, or a promoter system from phage lambda, may be employed, and others, as well, and comprise embodiments of the present invention. The promoters will typically control expression, optionally with an operator sequence and may include ribosome binding site sequences for example, for initiating and completing transcription and translation. According to additional embodiments, the vector may also contain expression control sequences, enhancers that may regulate the transcriptional activity of the promoter, appropriate restriction sites to facilitate cloning of inserts adjacent to the promoter and other necessary information processing sites, such as RNA splice sites, polyadenylation sites and transcription termination sequences as well as any other sequence which may facilitate the expression of the inserted nucleic acid. C. Purification of Anti-B7H6 Antibody [00241] Forms of anti-B7H6 antibody may be recovered from culture medium or from host cell lysates. If membrane-bound, it can be released from the membrane using a suitable detergent solution (e.g., Triton-X 100) or by enzymatic cleavage. Cells employed in expression of anti-B7H6 antibody can be disrupted by various physical or chemical means, such as freeze-thaw cycling, sonication, mechanical disruption, or cell lysing agents. [00242] It may be desired to purify anti-B7H6 antibody from recombinant cell proteins or polypeptides. The following procedures are exemplary of suitable purification procedures: by fractionation on an ion-exchange column; ethanol precipitation; reverse phase HPLC; chromatography on silica or on a cation-exchange resin such as DEAE; chromatofocusing; SDS- PAGE; ammonium sulfate precipitation; gel filtration using, for example, Sephadex G-75; protein A Sepharose columns to remove contaminants such as IgG; and metal chelating columns to bind epitope-tagged forms of the anti-B7H6 antibody. Various methods of protein purification may be employed and such methods are known in the art and described for example in Deutscher, Methods in Enzymology, 182 (1990); Scopes, Protein Purification: Principles and Practice, Springer- Verlag, New York (1982). The purification step(s) selected will depend, for example, on the nature of the production process used and the particular anti-B7H6 antibody produced. Page 65 of 203 1097657169\1\AMERICAS [00243] When using recombinant techniques, the antibody can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. If the antibody is produced intracellularly, as a first step, the particulate debris, either host cells or lysed fragments, are removed, for example, by centrifugation or ultrafiltration. Carter et al., Bio/Technology 10: 163-7 (1992) describe a procedure for isolating antibodies which are secreted to the periplasmic space of E. coli. Briefly, cell paste is thawed in the presence of sodium acetate (pH 3.5), EDTA, and phenylmethylsulfonylfluoride (PMSF) over about 30 min. Cell debris can be removed by centrifugation. Where the antibody is secreted into the medium, supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit. A protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants. [00244] The antibody composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography, with affinity chromatography being the preferred purification technique. The suitability of protein A as an affinity ligand depends on the species and isotype of any immunoglobulin Fc domain that is present in the antibody. Protein A can be used to purify antibodies that are based on human γ1, γ2 or γ4 heavy chains (Lindmark et al., J. Immunol. Meth. 62: 1-13 (1983)). Protein G is recommended for all mouse isotypes and for human γ3 (Guss et al., EMBO J.5: 15671575 (1986)). The matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose. Where the antibody comprises a CH3 domain, the Bakerbond ABX™resin (J. T. Baker, Phillipsburg, NJ) is useful for purification. Other techniques for protein purification such as fractionation on an ion-exchange column, ethanol precipitation, Reverse Phase HPLC, chromatography on silica, chromatography on heparin SEPHAROSE™ chromatography on an anion or cation exchange resin (such as a polyaspartic acid column), chromatofocusing, SDS- PAGE, and ammonium sulfate precipitation are also available depending on the antibody to be recovered. [00245] Following any preliminary purification step(s), the mixture comprising the antibody of interest and contaminants may be subjected to low pH hydrophobic interaction chromatography Page 66 of 203 1097657169\1\AMERICAS using an elution buffer at a pH between about 2.5-4.5, and generally at low salt concentrations (e.g., from about 0-0.25M salt). D. Assays [00246] The antibody of the present invention may be employed in any known assay method, such as ELISA, competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays (Zola, (1987) Monoclonal Antibodies: A Manual of Techniques, pp.147-158, CRC Press, Inc.). [00247] A detection label may be useful for localizing, visualizing, and quantitating a binding or recognition event. The labelled antibodies of the invention can detect cell-surface receptors or antigens. Another use for detectably labelled antibodies is a method of bead-based immunocapture comprising conjugating a bead with a fluorescent labelled antibody and detecting a fluorescence signal upon binding of a ligand. Similar binding detection methodologies utilize the surface plasmon resonance (SPR) effect to measure and detect antibody-antigen interactions. [00248] Detection labels such as fluorescent dyes and chemiluminescent dyes (Briggs et al (1997) J. Chem. Soc., Perkin-Trans. 1: 1051-8) provide a detectable signal and are generally applicable for labelling antibodies, preferably with the following properties: (i) the labelled antibody should produce a very high signal with low background so that small quantities of antibodies can be sensitively detected in both cell-free and cell-based assays; and (ii) the labelled antibody should be photostable so that the fluorescent signal may be observed, monitored and recorded without significant photo bleaching. For applications involving cell surface binding of labelled antibody to membranes or cell surfaces, especially live cells, the labels preferably (iii) have good water-solubility to achieve effective conjugate concentration and detection sensitivity and (iv) are non-toxic to living cells so as not to disrupt the normal metabolic processes of the cells or cause premature cell death. [00249] Direct quantification of cellular fluorescence intensity and enumeration of fluorescently labelled events, e.g., cell surface binding of peptide-dye conjugates may be conducted on a system (FMAT® 8100 HTS System, Applied Biosystems, Foster City, Calif.) that automates mix-and-read, non-radioactive assays with live cells or beads (Miraglia, “Homogeneous cell- and bead-based assays for high throughput screening using fluorometric microvolume assay technology”, (1999) J. of Biomolecular Screening 4:193-204). Uses of labelled antibodies also Page 67 of 203 1097657169\1\AMERICAS include cell surface receptor binding assays, inmmunocapture assays, fluorescence linked immunosorbent assays (FLISA), caspase-cleavage (Zheng, “Caspase-3 controls both cytoplasmic and nuclear events associated with Fas-mediated apoptosis in vivo”, (1998) Proc. Natl. Acad. Sci. USA 95:618-23; US 6372907), apoptosis (Vermes, “A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V” (1995) J. Immunol. Methods 184:39-51) and cytotoxicity assays. Fluorometric microvolume assay technology can be used to identify the up or down regulation by a molecule that is targeted to the cell surface (Swartzman, “A homogeneous and multiplexed immunoassay for high-throughput screening using fluorometric microvolume assay technology”, (1999) Anal. Biochem.271:143-51). [00250] Labelled antibodies of the invention are useful as imaging biomarkers and probes by the various methods and techniques of biomedical and molecular imaging such as: (i) MRI (magnetic resonance imaging); (ii) MicroCT (computerized tomography); (iii) SPECT (single photon emission computed tomography); (iv) PET (positron emission tomography) Chen et al Bioconjugate Chem.15: 41-9 (2004); (v) bioluminescence; (vi) fluorescence; and (vii) ultrasound. Immunoscintigraphy is an imaging procedure in which antibodies labeled with radioactive substances are administered to an animal or human patient and a picture is taken of sites in the body where the antibody localizes (US 6528624). Imaging biomarkers may be objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacological responses to a therapeutic intervention. [00251] Peptide labelling methods are well known (e.g., Haugland, 2003, Molecular Probes Handbook of Fluorescent Probes and Research Chemicals, Molecular Probes, Inc.; Brinkley, 1992, Bioconjugate Chem. 3:2; Garman, (1997) Non-Radioactive Labelling: A Practical Approach, Academic Press, London; Means (1990) Bioconjugate Chem.1:2; Glazer et al. (1975) Chemical Modification of Proteins. Laboratory Techniques in Biochemistry and Molecular Biology (T. S. Work and E. Work, Eds.) American Elsevier Publishing Co., New York; Lundblad, R. L. and Noyes, C. M. (1984) Chemical Reagents for Protein Modification, Vols. I and II, CRC Press, New York; Pfleiderer, G. (1985) “Chemical Modification of Proteins”, Modern Methods in Protein Chemistry, H. Tschesche, Ed., Walter DeGryter, Berlin and New York; and Wong (1991) Chemistry of Protein Conjugation and Cross-linking, CRC Press, Boca Raton, Fla.); De Leon- Rodriguez et al. (2004) Chem.Eur. J. 10:1149-1155; Lewis et al. (2001) Bioconjugate Chem. Page 68 of 203 1097657169\1\AMERICAS 12:320-324; Li et al. (2002) Bioconjugate Chem. 13:110-115; Mier et al. (2005) Bioconjugate Chem.16:240-237). [00252] Peptides and proteins labelled with two moieties, a fluorescent reporter and quencher in sufficient proximity undergo fluorescence resonance energy transfer (FRET). Reporter groups are typically fluorescent dyes that are excited by light at a certain wavelength and transfer energy to an acceptor, or quencher, group, with the appropriate Stokes shift for emission at maximal brightness. Fluorescent dyes include molecules with extended aromaticity, such as fluorescein and rhodamine, and their derivatives. The fluorescent reporter may be partially or significantly quenched by the quencher moiety in an intact peptide. Upon cleavage of the peptide by a peptidase or protease, a detectable increase in fluorescence may be measured (Knight, C. (1995) “Fluorimetric Assays of Proteolytic Enzymes”, Methods in Enzymology, Academic Press, 248:18- 34). [00253] The labelled antibodies of the invention may also be used as an affinity purification agent. In this process, the labelled antibody is immobilized on a solid phase such a Sephadex resin or filter paper, using methods well known in the art. The immobilized antibody is contacted with a sample containing the antigen to be purified, and thereafter the support is washed with a suitable solvent that will remove substantially all the material in the sample except the antigen to be purified, which is bound to the immobilized polypeptide variant. Finally, the support is washed with another suitable solvent, such as glycine buffer, pH 5.0, that will release the antigen from the polypeptide variant. 1. Activity assays [00254] In one aspect, assays are provided for identifying anti-B7H6 antibodies thereof having biological activity. Biological activity may include, e.g., the ability to inhibit cell growth or proliferation (e.g., “cell killing” activity), or the ability to induce cell death, including programmed cell death (apoptosis). Antibodies having such biological activity in vivo and/or in vitro are also provided. [00255] In certain embodiments, an anti-B7H6 antibody is tested for its ability to inhibit cell growth or proliferation in vitro. Assays for inhibition of cell growth or proliferation are well known in the art. Certain assays for cell proliferation, for example “cell killing” assays, measure cell viability. One such assay is the CellTiter-GloTM Luminescent Cell Viability Assay, which is Page 69 of 203 1097657169\1\AMERICAS commercially available from Promega (Madison, WI). That assay determines the number of viable cells in culture based on quantitation of ATP present, which is an indication of metabolically active cells. See Crouch et al (1993) J. Immunol. Meth.160: 81-8, US Pat. No.6602677. The assay may be conducted in 96- or 384-well format, making it amenable to automated high-throughput screening (HTS) (see Cree et al (1995) AntiCancer Drugs 6: 398-404). The assay procedure involves adding a single reagent (CellTiter-Glo® Reagent) directly to cultured cells. This results in cell lysis and generation of a luminescent signal produced by a luciferase reaction. The luminescent signal is proportional to the amount of ATP present, which is directly proportional to the number of viable cells present in culture. Data can be recorded by luminometer or CCD camera imaging device. The luminescence output is expressed as relative light units (RLU). [00256] Another assay for cell proliferation is the “MTT” assay, a colorimetric assay that measures the oxidation of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide to formazan by mitochondrial reductase. Like the CellTiter-GloTM assay, this assay indicates the number of metabolically active cells present in a cell culture (see, e.g., Mosmann (1983) J. Immunol. Meth.65:55-63, and Zhang et al. (2005) Cancer Res.65: 3877-82). [00257] In one aspect, an anti-B7H6 antibody is tested for its ability to induce cell death in vitro. Assays for induction of cell death are well known in the art. In embodiments, such assays measure, e.g., loss of membrane integrity as indicated by uptake of propidium iodide (PI), trypan blue (see Moore et al. Cytotechnology, 17: 1-11 (1995)), or 7AAD. In an exemplary PI uptake assay, cells are cultured in Dulbecco’s Modified Eagle Medium (D-MEM):Ham’s F-12 (50:50) supplemented with 10% heat-inactivated FBS (Hyclone) and 2 mM L-glutamine. Thus, the assay is performed in the absence of complement and immune effector cells. Cells are seeded at a density of 3 x 106 per dish in 100 x 20 mm dishes and allowed to attach overnight. The medium is removed and replaced with fresh medium alone or medium containing various concentrations of the antibody. The cells are incubated for a 3-day time period. Following treatment, monolayers are washed with PBS and detached by trypsinization. Cells are then centrifuged at 1200 rpm for 5 minutes at 4 ºC, the pellet resuspended in 3 mL cold Ca2+ binding buffer (10 mM Hepes, pH 7.4, 140 mM NaCl, 2.5 mM CaCl2) and aliquoted into 35 mm strainer-capped 12 x 75 mm tubes (1 mL per tube, 3 tubes per treatment group) for removal of cell clumps. Tubes then receive PI (10 μg/mL). Samples are analyzed using a FACSCAN™ flow cytometer and FACSCONVERT™ CellQuest software Page 70 of 203 1097657169\1\AMERICAS (Becton Dickinson). Antibodies which induce statistically significant levels of cell death as determined by PI uptake are thus identified. [00258] In one aspect, an anti-B7H6 antibody is tested for its ability to induce apoptosis (programmed cell death) in vitro. An exemplary assay for antibodies that induce apoptosis is an annexin binding assay. In an exemplary annexin binding assay, cells are cultured and seeded in dishes as discussed in the preceding paragraph. The medium is removed and replaced with fresh medium alone or medium containing 0.001 to 10 µg/mL of the antibody. Following a three-day incubation period, monolayers are washed with PBS and detached by trypsinization. Cells are then centrifuged, resuspended in Ca2+ binding buffer, and aliquoted into tubes as discussed in the preceding paragraph. Tubes then receive labeled annexin (e.g., annexin V-FITC) (1 µg/mL). Samples are analyzed using a FACSCAN™ flow cytometer and FACSCONVERT™ CellQuest software (BD Biosciences). Antibodies that induce statistically significant levels of annexin binding relative to control are thus identified. Another exemplary assay for antibodies that induce apoptosis is a histone DNA ELISA colorimetric assay for detecting internucleosomal degradation of genomic DNA. Such an assay can be performed using, e.g., the Cell Death Detection ELISA kit (Roche, Palo Alto, CA). [00259] Cells for use in any of the above in vitro assays include cells or cell lines that naturally express B7H6 or that have been engineered to express B7H6. Such cells include tumor cells that overexpress B7H6 relative to normal cells of the same tissue origin. Such cells also include cell lines (including tumor cell lines) that express B7H6 and cell lines that do not normally express B7H6 but have been transfected with nucleic acid encoding B7H6. [00260] In one aspect, an anti-B7H6 antibody thereof is tested for its ability to inhibit cell growth or proliferation in vivo. In certain embodiments, an anti-B7H6 antibody thereof is tested for its ability to inhibit tumor growth in vivo. In vivo model systems, such as xenograft models, can be used for such testing. In an exemplary xenograft system, human tumor cells are introduced into a suitably immunocompromised non-human animal, e.g., a SCID mouse. An antibody of the invention is administered to the animal. The ability of the antibody to inhibit or decrease tumor growth is measured. In certain embodiments of the above xenograft system, the human tumor cells are tumor cells from a human patient. In certain embodiments, the human tumor cells are Page 71 of 203 1097657169\1\AMERICAS introduced into a suitably immunocompromised non-human animal by subcutaneous injection or by transplantation into a suitable site, such as a mammary fat pad. 2. Binding assays and other assays [00261] In one aspect, an anti-B7H6 antibody is tested for its antigen binding activity. For example, in certain embodiments, an anti-B7H6 antibody is tested for its ability to bind to B7H6 expressed on the surface of a cell. A FACS assay may be used for such testing. [00262] In one aspect, competition assays may be used to identify a monoclonal antibody that competes with a monoclonal antibody comprising a HCVR/LCVR sequence pair of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, 15/16, 17/18, 19/20, 21/22, 23/24, 25/26, 27/28, 29/30, 31/32, 33/34, 35/36, 37/38, or 39/40; or that competes with a monoclonal antibody comprising the six CDRs of a HCVR/LCVR sequence pair selected from SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, 15/16, 17/18, 19/20, 21/22, 23/24, 25/26, 27/28, 29/30, 31/32, 33/34, 35/36, 37/38, or 39/40. [00263] In certain embodiments, such a competing antibody binds to the same epitope (e.g., a linear or a conformational epitope) that is bound by a monoclonal antibody comprising a HCVR/LCVR sequence pair of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, 15/16, 17/18, 19/20, 21/22, 23/24, 25/26, 27/28, 29/30, 31/32, 33/34, 35/36, 37/38, or 39/40; or a monoclonal antibody comprising the six CDRs of a HCVR/LCVR sequence pair selected from SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, 15/16, 17/18, 19/20, 21/22, 23/24, 25/26, 27/28, 29/30, 31/32, 33/34, 35/36, 37/38, or 39/40. Exemplary competition assays include, but are not limited to, routine assays such as those provided in Harlow and Lane (1988) Antibodies: A Laboratory Manual ch.14 (Cold Spring Harbor Laboratory, Cold Spring Harbor, NY). Detailed exemplary methods for mapping an epitope to which an antibody binds are provided in Morris (1996) “Epitope Mapping Protocols,” in Methods in Molecular Biology vol.66 (Humana Press, Totowa, NJ). Two antibodies are said to bind to the same epitope if each blocks binding of the other by 50% or more. [00264] In an exemplary competition assay, immobilized B7H6 is incubated in a solution comprising a first labeled antibody that binds to B7H6 and a second unlabeled antibody that is being tested for its ability to compete with the first antibody for binding to B7H6. The second antibody may be present in a hybridoma supernatant. As a control, immobilized B7H6 is incubated in a solution comprising the first labeled antibody but not the second unlabeled antibody. After Page 72 of 203 1097657169\1\AMERICAS incubation under conditions permissive for binding of the first antibody to B7H6, excess unbound antibody is removed, and the amount of label associated with immobilized B7H6 is measured. If the amount of label associated with immobilized B7H6 is substantially reduced in the test sample relative to the control sample, then that indicates that the second antibody is competing with the first antibody for binding to B7H6. In certain embodiments, immobilized B7H6 is present on the surface of a cell or in a membrane preparation obtained from a cell expressing B7H6 on its surface. [00265] In one aspect, purified anti-B7H6 antibodies can be further characterized by a series of assays including, but not limited to, N-terminal sequencing, amino acid analysis, non-denaturing size exclusion high pressure liquid chromatography (HPLC), mass spectrometry, ion exchange chromatography and papain digestion. E. Methods of Epitope Identification [00266] In one aspect, the present disclosure provides a method for identifying the epitope of an anti-B7H6 antibody or antigen-binding fragment thereof. [00267] An epitope can include at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acids in a unique spatial conformation. Epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids can be typically retained on exposure to denaturing solvents whereas epitopes formed by tertiary folding can be typically lost on treatment with denaturing solvents. [00268] Epitope mapping can be performed to identify the linear or non-linear, discontinuous amino acid sequence(s), i.e. the epitope, that is (e.g., specifically) recognized by an anti-B7H6 antibody or antigen-binding fragment thereof. A general approach for epitope mapping can require the expression of the full-length polypeptide sequence that is recognized by an antibody or ligand of interest, as well as various fragments, i.e., truncated forms of the polypeptide sequence, generally in a heterologous expression system. These various recombinant polypeptide sequences or fragments thereof (e.g., fused with an N-terminal protein (e.g., GFP)) can then be used to determine if the antibody or ligand of interest is capable of binding to one or more of the truncated forms of the polypeptide sequence. Page 73 of 203 1097657169\1\AMERICAS [00269] Through the use of reiterative truncation and the generation of recombinant polypeptide sequences with overlapping amino acid regions, it is possible to identify the region of the polypeptide sequence that is recognized by the antibody of interest (see, e.g., Epitope Mapping Protocols in Methods in Molecular Biology, Vol.66, Glenn E. Morris, Ed (1996)). The methods rely on the ability of an agent such as an antibody of interest to bind to sequences that have been recreated from epitope libraries, such as epitope libraries derived from, synthetic peptide arrays on membrane supports, combinatorial phage display peptide libraries. The epitope libraries then provide a range of possibilities that are screened against an antibody. Additionally, site specific mutagenesis, or random Ala scan, targeting one or more residues of an epitope can be pursued to confirm the identity of an epitope. [00270] A library of epitopes can be created by synthetically designing various portions of B7H6 as constructs and expressing them in a suitable system. In other cases, portions of B7H6 can be amplified out of Total RNA extracted from B7H6-expressing-cells isolated from human normal and/or malignant tissue. [00271] The host system can be any suitable expression system such as 293 cells, insect cells, or a suitable in-vitro translation system. The binding of an anti-B7H6 antibody or antigen-binding fragment thereof to one of the epitopes in the above-described library can be detected by contacting a labeled B7H6 antibody of the present disclosure with an epitope of the library and detecting a signal from the label. [00272] For epitope mapping, computational algorithms have also been developed which have been shown to map conformational discontinuous epitopes. Conformational epitopes can be identified by determining spatial conformation of amino acids with methods that include, e.g., x- ray crystallography and 2-dimensional nuclear magnetic resonance. Some epitope mapping methods, such as, x-ray analyses of crystals of antigen: antibody complexes can provide atomic resolution of the epitope. In other cases, computational combinatorial methods for epitope mapping can be employed to model a potential epitope based on the sequence of the anti-B7H6 or antigen-binding fragment thereof. In such cases, the antigen binding portion of the antibody is sequenced, and computation models are used to reconstruct and predict a potential binding site of the antibody. Page 74 of 203 1097657169\1\AMERICAS [00273] In some cases the disclosure provides a method of determining a B7H6 epitope, comprising: (a) preparing a library of epitopes from B7H6; (b) contacting the library of epitopes with an anti-B7H6 antibody; and (b) identifying the amino acid sequence of at least one epitope in the library of epitopes that is bound by the antibody. In one instance, the antibody is attached to a solid support. The library of epitopes can comprise sequences that correspond to continuous and discontinuous epitopes of B7H6. In some cases, the library of epitopes comprises fragments from B7H6 ranging from about 10 amino acids to about 30 amino acids in length, from about 10 amino acids to about 20 amino acids in length, or from about 5 amino acids to about 12 amino acids in length. In some cases, the anti-B7H6 antibody or antigen-binding fragment thereof is labeled and the label is a radioactive molecule, a luminescent molecule, a fluorescent molecule, an enzyme, or biotin. [00274] Phage panning may be used to identify B7H6-binding molecules that bind to B7H6 or one or more epitopes on B7H6. In embodiments, phage panning using biotinylated recombinant human B7H6 protein bound to streptavidin beads is performed on highly diverse synthetic scFv phage display libraries using multiple (e.g., 5) rounds of selection, each with decreasing antigen concentration (e.g., starting at 100 pmol to 2 pmol) but increasing wash stringency. In embodiments, an alternate panning strategy with alternating rounds of selection with 1e8 B7H6+ cells or antigen (e.g., 100 pmol and 25 pmol, respectively) may be used. Antigen-bound phage may then be pulled down, eluted, amplified and screened using ELISAs for binder confirmation and selection. Following NGS and/or Sanger clone sequencing, the selected scFvs may be reformatted into IgGs, expressed, purified and characterized. F. Chimeric Antigen Receptor (CAR) Constructs [00275] Aspects of the invention include nucleic acids encoding CARs, and constructs and vectors containing such nucleic acids. In some cases, the nucleic acid is a, e.g., heterologous, component of an expression cassette. In embodiments, the nucleic acid is a, e.g., heterologous, component of a retroviral vector. In embodiments, the nucleic acid is a, e.g., heterologous, component of an ^ ^ or γδ T cell, and preferably a γδ T cell. In embodiments, the nucleic acid is a, e.g., heterologous, component of an γ+ T cell and/or a δ+ T cell. In embodiments, the nucleic acid is a, e.g., heterologous, component of an α- T cell and/or a β- T cell. Page 75 of 203 1097657169\1\AMERICAS [00276] A subject CAR of the invention comprises an antigen binding domain capable of specifically binding to B7H6. The antigen binding domain may be operably linked to another domain of the CAR, for example a transmembrane domain, a costimulatory domain and/or an intracellular signaling domain, as described herein. The antigen binding domains described herein can be combined with any of the transmembrane, costimulatory, and/or intracellular signaling domain(s) described herein, and/or any of the other domains described herein that may be included in a CAR of the present invention. A subject CAR of the present invention may also include a hinge domain as described herein. A subject CAR of the present invention may also include at least one spacer domain as described herein. 1. Antigen Binding Domain [00277] The antigen binding domain can include any domain that binds to B7H6 and may include, but is not limited to, a monoclonal antibody, a polyclonal antibody, a synthetic antibody, a human antibody, a humanized antibody, a non-human antibody, a bispecific antibody, and any fragment thereof. In embodiments, the antigen binding domain portion comprises a mammalian antibody or a fragment thereof. The choice of antigen binding domain may depend upon the type and number of antigens that are present on the surface of a target cell. [00278] In embodiments, the antigen binding domain is selected from the group consisting of an antibody, an antigen binding fragment (Fab), and a single-chain variable fragment (scFv). [00279] The present disclosure provides antibodies and CARs with “substantial identity” or “substantial similarity” to the sequences provided herein in the CDR or framework regions. The term "substantial identity" or "substantially identical," when referring to a nucleic acid or fragment thereof, indicates that, when optimally aligned with another nucleic acid (or the complementary strand of the other nucleic acid), there is nucleotide sequence identity in %, for example, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, or 100% of the nucleotide bases, as measured by any well-known algorithm of sequence identity, such as FASTA, BLAST or GAP, as discussed below. A nucleic acid molecule having substantial identity to a reference nucleic acid molecule may, in certain instances, encode a polypeptide having the same Page 76 of 203 1097657169\1\AMERICAS or substantially similar amino acid sequence as the polypeptide encoded by the reference nucleic acid molecule. [00280] As applied to polypeptides, the term "substantial similarity" or “substantially similar” means that two peptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, or 100% sequence identity. In some aspects, residue positions, which are not identical, differ by conservative amino acid substitutions. A "conservative amino acid substitution" is one in which an amino acid residue is substituted by another amino acid residue having a side chain (R group) with similar chemical properties (e.g., charge or hydrophobicity). In general, a conservative amino acid substitution will not substantially change the functional properties of a protein. In cases where two or more amino acid sequences differ from each other by conservative substitutions, the percent or degree of similarity may be adjusted upwards to correct for the conservative nature of the substitution. Means for making this adjustment are well known to those of skill in the art. See, e.g., Pearson (1994) Methods Mol. Biol.24: 307-331, which is herein incorporated by reference. Examples of groups of amino acids that have side chains with similar chemical properties include 1) aliphatic side chains: glycine, alanine, valine, leucine and isoleucine; 2) aliphatic-hydroxyl side chains: serine and threonine; 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; 5) basic side chains: lysine, arginine, and histidine; 6) acidic side chains: aspartate and glutamate, and 7) sulfur-containing side chains: cysteine and methionine. Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine- tyrosine, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-glutamine. Alternatively, a conservative replacement is any change having a positive value in the PAM250 log-likelihood matrix disclosed in Gonnet et al. (1992) Science 256: 144345, herein incorporated by reference. A "moderately conservative" replacement is any change having a nonnegative value in the PAM250 log-likelihood matrix. [00281] Sequence identity and/or similarity for polypeptides is typically measured using sequence analysis software. Protein analysis software matches similar sequences using measures of similarity assigned to various substitutions, deletions and other modifications, including Page 77 of 203 1097657169\1\AMERICAS conservative amino acid substitutions. For instance, GCG software contains programs such as GAP and BESTFIT which can be used with default parameters to determine sequence homology or sequence identity between closely related polypeptides, such as homologous polypeptides from different species of organisms or between a wild type protein and a mutein thereof. See, e.g., GCG Version 6.1. Polypeptide sequences also can be compared using FASTA with default or recommended parameters; a program in GCG Version 6.1. FASTA (e.g., FASTA2 and FASTA3) provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (Pearson (2000) supra). Sequences also can be compared using the Smith-Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 2, BLOSUM matrix of 62. Another preferred algorithm when comparing a sequence disclosed herein to a database containing a large number of sequences from different organisms is the computer program BLAST, especially BLASTP or TBLASTN, using default parameters. See, e.g., Altschul et al. (1990) J. Mol. Biol.215: 403-410 and (1997) Nucleic Acids Res.25:3389-3402, each of which is herein incorporated by reference. [00282] Provided herein are anti-B7H6 CARs comprising variants of any of the HCVR, LCVR, and/or CDR amino acid sequences disclosed herein having one or more substitutions (e.g., conservative substitutions). For example, the present disclosure includes anti-B7H6 CARs having HCVR, LCVR, and/or CDR amino acid sequences with, e.g., 20 or fewer, 19 or fewer, 18 or fewer, 17 or fewer, 16 or fewer, 15 or fewer, 14 or fewer, 13 or fewer, 12 or fewer, 11 or fewer, 10 or fewer, 9 or fewer, 8 or fewer, 7 or fewer, 6 or fewer, 5 or fewer, 4 or fewer, 3 or fewer, 2 or fewer, or 1 amino acid substitutions relative to any of the HCVR, LCVR, and/or CDR (e.g., HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, or LCDR3) amino acid sequences disclosed herein. For example, an anti-B7H6 CAR can comprise 20, 19, 18, 17, 16, 15, 1413, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid substitutions (e.g., conservative amino acid substitutions) relative to any of the HCVR, LCVR, and/or CDR (e.g., HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, or LCDR3) amino acid sequences disclosed herein. [00283] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising an amino acid sequence selected from the group consisting of any one of SEQ ID NOs: 1-40. In embodiments, the anti-B7H6 binding domain binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence selected from the group Page 78 of 203 1097657169\1\AMERICAS consisting of any one of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, and 39. In embodiments, the anti-B7H6 binding domain binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a LCVR amino acid sequence selected from the group consisting of any one of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, and 40. In embodiments, the anti-B7H6 binding domain binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence selected from the group consisting of any one of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, and 15. In embodiments, the anti-B7H6 binding domain binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a LCVR amino acid sequence selected from the group consisting of any one of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, and 16. [00284] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 amino acid sequence selected from the group consisting of SEQ ID NOs: 41- 60; a CDR2 amino acid sequence selected from the group consisting of SEQ ID NOs: 61-80; and a CDR3 amino acid sequence selected from the group consisting of SEQ ID NOs: 81-100. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a LCVR comprising a CDR1 amino acid sequence selected from the group consisting of SEQ ID NOs: 101-120; a CDR2 amino acid sequence selected from the group consisting of SEQ ID NOs: 121-140; and a CDR3 amino acid sequence selected from the group consisting of SEQ ID NOs: 141-160. [00285] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 amino acid sequence selected from the group consisting of SEQ ID NOs: 41- 48; a CDR2 amino acid sequence selected from the group consisting of SEQ ID NOs: 61-68; and a CDR3 amino acid sequence selected from the group consisting of SEQ ID NOs: 81-88. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a LCVR comprising a CDR1 amino acid sequence selected from the group consisting of SEQ ID NOs: 101-108; a CDR2 amino acid sequence selected from the group consisting of SEQ ID NOs: 121-128; and a CDR3 amino acid sequence selected from the group consisting of SEQ ID NOs: 141-148. Page 79 of 203 1097657169\1\AMERICAS [00286] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 1, and a LCVR amino acid sequence set forth at SEQ ID NO: 2. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 3, and a LCVR amino acid sequence set forth at SEQ ID NO: 4. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 5, and a LCVR amino acid sequence set forth at SEQ ID NO: 6. In embodiments, an isolated nucleic acid encodes an anti- B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 7, and a LCVR amino acid sequence set forth at SEQ ID NO: 8. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 9, and a LCVR amino acid sequence set forth at SEQ ID NO: 10. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti- B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 11, and a LCVR amino acid sequence set forth at SEQ ID NO: 12. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 13, and a LCVR amino acid sequence set forth at SEQ ID NO: 14. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 15, and a LCVR amino acid sequence set forth at SEQ ID NO: 16. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 17, and a LCVR amino acid sequence set forth at SEQ ID NO: 18. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 19, and a LCVR amino acid sequence set forth at SEQ ID NO: 20. In embodiments, an isolated nucleic Page 80 of 203 1097657169\1\AMERICAS acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 21, and a LCVR amino acid sequence set forth at SEQ ID NO: 22. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 23, and a LCVR amino acid sequence set forth at SEQ ID NO: 24. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 25, and a LCVR amino acid sequence set forth at SEQ ID NO: 26. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 27, and a LCVR amino acid sequence set forth at SEQ ID NO: 28. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 29, and a LCVR amino acid sequence set forth at SEQ ID NO: 30. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 31, and a LCVR amino acid sequence set forth at SEQ ID NO: 32. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 33, and a LCVR amino acid sequence set forth at SEQ ID NO: 34. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 35, and a LCVR amino acid sequence set forth at SEQ ID NO: 36. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 37, and a LCVR amino acid sequence set forth at SEQ ID NO: 38. In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 39, and a LCVR amino acid sequence set forth at SEQ ID NO: 40. Page 81 of 203 1097657169\1\AMERICAS [00287] Preferred embodiments include those in which an isolated nucleic acid encodes an anti- B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR amino acid sequence set forth as SEQ ID NO: 1, and a LCVR amino acid sequence set forth at SEQ ID NO: 2; a HCVR amino acid sequence set forth as SEQ ID NO: 3, and a LCVR amino acid sequence set forth at SEQ ID NO: 4; a HCVR amino acid sequence set forth as SEQ ID NO: 5, and a LCVR amino acid sequence set forth at SEQ ID NO: 6; a HCVR amino acid sequence set forth as SEQ ID NO: 7, and a LCVR amino acid sequence set forth at SEQ ID NO: 8; a HCVR amino acid sequence set forth as SEQ ID NO: 9, and a LCVR amino acid sequence set forth at SEQ ID NO: 10; a HCVR amino acid sequence set forth as SEQ ID NO: 11, and a LCVR amino acid sequence set forth at SEQ ID NO: 12; a HCVR amino acid sequence set forth as SEQ ID NO: 13, and a LCVR amino acid sequence set forth at SEQ ID NO: 14; or a HCVR amino acid sequence set forth as SEQ ID NO: 15, and a LCVR amino acid sequence set forth as SEQ ID NO: 16. [00288] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 41, a CDR2 sequence set forth as SEQ ID NO: 61, and a CDR3 sequence set forth as SEQ ID NO: 81; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 101, a CDR2 sequence set forth as SEQ ID NO: 121, and a CDR3 sequence set forth as SEQ ID NO: 141. [00289] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 42, a CDR2 sequence set forth as SEQ ID NO: 62, and a CDR3 sequence set forth as SEQ ID NO: 82; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 102, a CDR2 sequence set forth as SEQ ID NO: 122, and a CDR3 sequence set forth as SEQ ID NO: 142. [00290] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 43, a CDR2 sequence set forth as SEQ ID NO: 63, and a CDR3 sequence set forth as SEQ ID NO: 83; and/or comprising a LCVR comprising Page 82 of 203 1097657169\1\AMERICAS a CDR1 sequence set forth as SEQ ID NO: 103, a CDR2 sequence set forth as SEQ ID NO: 123, and a CDR3 sequence set forth as SEQ ID NO: 143. [00291] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 44, a CDR2 sequence set forth as SEQ ID NO: 64, and a CDR3 sequence set forth as SEQ ID NO: 84; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 104, a CDR2 sequence set forth as SEQ ID NO: 124, and a CDR3 sequence set forth as SEQ ID NO: 144. [00292] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 45, a CDR2 sequence set forth as SEQ ID NO: 65, and a CDR3 sequence set forth as SEQ ID NO: 85; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 105, a CDR2 sequence set forth as SEQ ID NO: 125, and a CDR3 sequence set forth as SEQ ID NO: 145. [00293] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 46, a CDR2 sequence set forth as SEQ ID NO: 66, and a CDR3 sequence set forth as SEQ ID NO: 86; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 106, a CDR2 sequence set forth as SEQ ID NO: 126, and a CDR3 sequence set forth as SEQ ID NO: 146. [00294] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 47, a CDR2 sequence set forth as SEQ ID NO: 67, and a CDR3 sequence set forth as SEQ ID NO: 87; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 107, a CDR2 sequence set forth as SEQ ID NO: 127, and a CDR3 sequence set forth as SEQ ID NO: 147. [00295] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 48, a CDR2 sequence set forth as SEQ ID NO: 68, and a CDR3 sequence set forth as SEQ ID NO: 88; and/or comprising a LCVR comprising Page 83 of 203 1097657169\1\AMERICAS a CDR1 sequence set forth as SEQ ID NO: 108, a CDR2 sequence set forth as SEQ ID NO: 128, and a CDR3 sequence set forth as SEQ ID NO: 148. [00296] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 49, a CDR2 sequence set forth as SEQ ID NO: 69, and a CDR3 sequence set forth as SEQ ID NO: 89; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 109, a CDR2 sequence set forth as SEQ ID NO: 129, and a CDR3 sequence set forth as SEQ ID NO: 149. [00297] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 50, a CDR2 sequence set forth as SEQ ID NO: 70, and a CDR3 sequence set forth as SEQ ID NO: 90; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 110, a CDR2 sequence set forth as SEQ ID NO: 130, and a CDR3 sequence set forth as SEQ ID NO: 150. [00298] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 51, a CDR2 sequence set forth as SEQ ID NO: 71, and a CDR3 sequence set forth as SEQ ID NO: 91; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 111, a CDR2 sequence set forth as SEQ ID NO: 131, and a CDR3 sequence set forth as SEQ ID NO: 151. [00299] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 52, a CDR2 sequence set forth as SEQ ID NO: 72, and a CDR3 sequence set forth as SEQ ID NO: 92; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 112, a CDR2 sequence set forth as SEQ ID NO: 132, and a CDR3 sequence set forth as SEQ ID NO: 152. [00300] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 53, a CDR2 sequence set forth as SEQ ID NO: 73, and a CDR3 sequence set forth as SEQ ID NO: 93; and/or comprising a LCVR comprising Page 84 of 203 1097657169\1\AMERICAS a CDR1 sequence set forth as SEQ ID NO: 113, a CDR2 sequence set forth as SEQ ID NO: 133, and a CDR3 sequence set forth as SEQ ID NO: 153. [00301] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 54, a CDR2 sequence set forth as SEQ ID NO: 74, and a CDR3 sequence set forth as SEQ ID NO: 94; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 114, a CDR2 sequence set forth as SEQ ID NO: 134, and a CDR3 sequence set forth as SEQ ID NO: 154. [00302] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 55, a CDR2 sequence set forth as SEQ ID NO: 75, and a CDR3 sequence set forth as SEQ ID NO: 95; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 115, a CDR2 sequence set forth as SEQ ID NO: 135, and a CDR3 sequence set forth as SEQ ID NO: 155. [00303] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 56, a CDR2 sequence set forth as SEQ ID NO: 76, and a CDR3 sequence set forth as SEQ ID NO: 96; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 116, a CDR2 sequence set forth as SEQ ID NO: 136, and a CDR3 sequence set forth as SEQ ID NO: 156. [00304] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 57, a CDR2 sequence set forth as SEQ ID NO: 77, and a CDR3 sequence set forth as SEQ ID NO: 97; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 117, a CDR2 sequence set forth as SEQ ID NO: 137, and a CDR3 sequence set forth as SEQ ID NO: 157. [00305] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 58, a CDR2 sequence set forth as SEQ ID NO: 78, and a CDR3 sequence set forth as SEQ ID NO: 98; and/or comprising a LCVR comprising Page 85 of 203 1097657169\1\AMERICAS a CDR1 sequence set forth as SEQ ID NO: 118, a CDR2 sequence set forth as SEQ ID NO: 138, and a CDR3 sequence set forth as SEQ ID NO: 158. [00306] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 59, a CDR2 sequence set forth as SEQ ID NO: 79, and a CDR3 sequence set forth as SEQ ID NO: 99; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 119, a CDR2 sequence set forth as SEQ ID NO: 139, and a CDR3 sequence set forth as SEQ ID NO: 159. [00307] In embodiments, an isolated nucleic acid encodes an anti-B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 60, a CDR2 sequence set forth as SEQ ID NO: 80, and a CDR3 sequence set forth as SEQ ID NO: 100; and/or comprising a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 120, a CDR2 sequence set forth as SEQ ID NO: 140, and a CDR3 sequence set forth as SEQ ID NO: 160. [00308] Preferred embodiments include those in which an isolated nucleic acid encodes an anti- B7H6 binding domain that binds the same epitope as, competes with, or is an anti-B7H6 binding domain comprising a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 41, a CDR2 sequence set forth as SEQ ID NO: 61, and a CDR3 sequence set forth as SEQ ID NO: 81, and a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 101, a CDR2 sequence set forth as SEQ ID NO: 121, and a CDR3 sequence set forth as SEQ ID NO: 141; a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 42, a CDR2 sequence set forth as SEQ ID NO: 62, and a CDR3 sequence set forth as SEQ ID NO: 82, and a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 102, a CDR2 sequence set forth as SEQ ID NO: 122, and a CDR3 sequence set forth as SEQ ID NO: 142; a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 43, a CDR2 sequence set forth as SEQ ID NO: 63, and a CDR3 sequence set forth as SEQ ID NO: 83, and a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 103, a CDR2 sequence set forth as SEQ ID NO: 123, and a CDR3 sequence set forth as SEQ ID NO: 143; a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 44, a CDR2 sequence set forth as SEQ ID NO: 64, and a CDR3 sequence set forth as SEQ ID NO: 84, and a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 104, a CDR2 sequence set forth as SEQ ID NO: 124, and a CDR3 sequence set Page 86 of 203 1097657169\1\AMERICAS forth as SEQ ID NO: 144; a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 45, a CDR2 sequence set forth as SEQ ID NO: 65, and a CDR3 sequence set forth as SEQ ID NO: 85, and a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 105, a CDR2 sequence set forth as SEQ ID NO: 125, and a CDR3 sequence set forth as SEQ ID NO: 145; a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 46, a CDR2 sequence set forth as SEQ ID NO: 66, and a CDR3 sequence set forth as SEQ ID NO: 86, and a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 106, a CDR2 sequence set forth as SEQ ID NO: 126, and a CDR3 sequence set forth as SEQ ID NO: 146; a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 47, a CDR2 sequence set forth as SEQ ID NO: 67, and a CDR3 sequence set forth as SEQ ID NO: 87, and a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 107, a CDR2 sequence set forth as SEQ ID NO: 127, and a CDR3 sequence set forth as SEQ ID NO: 147; or a HCVR comprising a CDR1 sequence set forth as SEQ ID NO: 48, a CDR2 sequence set forth as SEQ ID NO: 68, and a CDR3 sequence set forth as SEQ ID NO: 88, and a LCVR comprising a CDR1 sequence set forth as SEQ ID NO: 108, a CDR2 sequence set forth as SEQ ID NO: 128, and a CDR3 sequence set forth as SEQ ID NO: 148. 2. Transmembrane Domain [00309] CARs of the present disclosure may comprise a transmembrane domain that couples the antigen binding domain of the CAR to one or more intracellular domains of the CAR. The transmembrane domain of a CAR of the present disclosure is a region that is capable of spanning the plasma membrane of a cell (e.g., a γδ T cell). In embodiments, the transmembrane domain is interposed between the antigen binding domain and the one or more intracellular domains of a CAR. [00310] In embodiments, the transmembrane domain is naturally associated with one or more of the domains in the CAR. In embodiments, the transmembrane domain can e selected or modified by one or more amino acid substitutions to avoid binding of such domains to the transmembrane domains of the same or different surface membrane proteins, to minimize interactions with other members of the receptor complex. [00311] For example and without limitation, a transmembrane domain may be derived either from a natural or from a synthetic source. Where the source is natural, the domain may be derived from any membrane-bound or transmembrane protein. Transmembrane regions of particular use Page 87 of 203 1097657169\1\AMERICAS in this invention may be derived from (i.e. comprise at least the transmembrane region(s) of) 4- 1BB/CD137, activating NK cell receptors, an Immunoglobulin protein, B7-H3, BAFFR, BLAME (SLAMF8), BTLA, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, or CD154, CD100 (SEMA4D), CD103, CD160 (BY55), CD18, CD19, CD19a, CD2, CD247, CD27, CD276 (B7-H3), CD28, CD29, CD3 delta, CD3 epsilon, CD3 gamma, CD3 zeta, CD30, CD4, CD40, CD49a, CD49D, CD49f, CD69, CD7, CD84, CD8, CD8alpha, CD8beta, CD96 (Tactile), CD11a, CD11b, CD11c, CD11d, CDS, CEACAM1, CRT AM, cytokine receptor, DAP10, DNAM1 (CD226), Fc gamma receptor, GADS, GITR, HVEM (LIGHTR), IA4, ICAM-1, Ig alpha (CD79a), IL-2R beta, IL-2R gamma, IL-7R alpha, inducible T cell costimulator (ICOS), integrins, ITGA4, ITGA6, ITGAD, ITGAE, ITGAL, ITGAM, ITGAX, ITGB2, ITGB7, ITGBl, KIRDS2, LAT, LFA-1, a ligand that specifically binds with CD83, LIGHT, LTBR, Ly9 (CD229), lymphocyte function-associated antigen-1 (LFA-1; CD11a/CD18), MHC class 1 molecule, NKG2C, NKG2D, NKp30, NKp44, NKp46, NKp80 (KLRF1), OX-40, PAG/Cbp, programmed death-1 (PD-1), PSGL1, SELPLG (CD162), Signaling Lymphocytic Activation Molecules (SLAM proteins), SLAM (SLAMF1; CD150; IPO-3), SLAMF4 (CD244; 2B4), SLAMF6 (NTB-A; Lyl08), SLAMF7, SLP-76, TNF receptor proteins, TNFR2, TNFSF14, a Toll ligand receptor, TRANCE/RANKL, VLA1, or VLA-6, or a fragment, truncation, or a combination thereof. Alternatively, the transmembrane domain may be synthetic, in which case it will comprise predominantly hydrophobic residues such as leucine and valine. Preferably a triplet of phenylalanine, tryptophan and valine will be found at each end of a synthetic transmembrane domain. [00312] In certain embodiments, the transmembrane domain comprises a transmembrane domain of CD8. In certain embodiments, the transmembrane domain of CD8 is a transmembrane domain of CD8α. In certain embodiments, the transmembrane domain of CD8 comprises the amino acid sequence set forth in SEQ ID NO: 211. In certain embodiments, the transmembrane domain comprises a transmembrane domain of CD28. In certain embodiments, the transmembrane domain of CD28 comprises the amino acid sequence set forth in SEQ ID NO: 215. In certain embodiments, the transmembrane domain comprises a transmembrane domain of ICOS. In certain embodiments, the transmembrane domain of ICOS comprises the amino acid sequence set forth in SEQ ID NO: 216. Page 88 of 203 1097657169\1\AMERICAS [00313] The transmembrane domains described herein can be combined with any of the antigen binding domains described herein, any of the intracellular domains described herein, or any of the other domains described herein that may be included in a subject CAR. [00314] In embodiments, the transmembrane domain further comprises a hinge region. A subject CAR of the present invention may also include a hinge region. The hinge region of the CAR is a hydrophilic region which is located between the antigen binding domain and the transmembrane domain. In embodiments, this domain facilitates proper protein folding for the CAR. The hinge region is an optional component for the CAR. The hinge region may include a domain selected from Fc fragments of antibodies, hinge regions of antibodies, CH2 regions of antibodies, CH3 regions of antibodies, artificial hinge sequences or combinations thereof. Examples of hinge regions include, without limitation, a CD8α hinge, CD8β hinge, CD28 hinge, 4-1BB hinge, CD7 hinge, artificial hinges made of polypeptides which may be as small as, three glycines (Gly), as well as CHI and CH3 domains of IgGs (such as human IgG4). Naturally- occurring hinge domains may be used as wild-type hinge regions or the molecules may be altered. [00315] In embodiments, a subject CAR of the present disclosure includes a hinge region that couples the antigen binding domain with the transmembrane domain, which, in turn, couples to one or more intracellular domain(s). The hinge region is preferably capable of supporting the antigen binding domain to recognize and bind to the target antigen on the target cells (see, e.g., Hudecek et al., Cancer Immunol. Res. (2015) 3(2): 125-135). In embodiments, the hinge region is a flexible domain, thus allowing the antigen binding domain to have a structure to optimally recognize the specific structure and density of the target antigens on a cell such as tumor cell (Hudecek et al., supra). The flexibility of the hinge region permits the hinge region to adopt many different conformations. In embodiments, the hinge region is an immunoglobulin heavy chain hinge region. In embodiments, the hinge region is a hinge region polypeptide derived from a receptor (e.g., a CD8-derived hinge region). [00316] The hinge region can have a length of from about 4 amino acids to about 50 amino acids, e.g., from about 4 aa to about 10 aa, from about 10 aa to about 15 aa, from about 15 aa to about 20 aa, from about 20 aa to about 25 aa, from about 25 aa to about 30 aa, from about 30 aa to about 40 aa, or from about 40 aa to about 50 aa. In embodiments, the hinge region can have a length of greater than 5 aa, greater than 10 aa, greater than 15 aa, greater than 20 aa, greater than Page 89 of 203 1097657169\1\AMERICAS 25 aa, greater than 30 aa, greater than 35 aa, greater than 40 aa, greater than 45 aa, greater than 50 aa, greater than 55 aa, or more. [00317] Suitable hinge regions can be readily selected and can be of any of a number of suitable lengths, such as from 1 amino acid (e.g., Gly) to 20 amino acids, from 2 amino acids to 15 amino acids, from 3 amino acids to 12 amino acids, including 4 amino acids to 10 amino acids, 5 amino acids to 9 amino acids, 6 amino acids to 8 amino acids, or 7 amino acids to 8 amino acids, and can be 1, 2, 3, 4, 5, 6, or 7 amino acids. Suitable hinge regions can have a length of greater than 20 amino acids (e.g., 30, 40, 50, 60 or more amino acids). [00318] For example, hinge regions include glycine polymers (G)n, glycine-serine polymers (including, for example, (GS)n, (GSGGS)n (SEQ ID NO: 174) and (GGGS)n (SEQ ID NO: 175), where n is an integer of at least one), glycine-alanine polymers, alanine-serine polymers, and other flexible linkers known in the art. Glycine and glycine-serine polymers can be used; both Gly and Ser are relatively unstructured, and therefore can serve as a neutral tether between components. Glycine polymers can be used; glycine accesses significantly more phi-psi space than even alanine, and is much less restricted than residues with longer side chains (see, e.g., Scheraga, Rev. Computational. Chem. (1992) 2: 73-142). Exemplary hinge regions can comprise amino acid sequences including, but not limited to, GGSG (SEQ ID NO: 177), GGSGG (SEQ ID NO: 178), GSGSG (SEQ ID NO: 179), GSGGG (SEQ ID NO: 180), GGGSG (SEQ ID NO: 181), GSSSG (SEQ ID NO: 182), and the like. [00319] In embodiments, the hinge region is an immunoglobulin heavy chain hinge region. Immunoglobulin hinge region amino acid sequences are known in the art; see, e.g., Tan et ah, Proc. Natl. Acad. Sci. USA (1990) 87(1): 162-166; and Huck et ah, Nucleic Acids Res. (1986) 14(4): 1779-1789. As non-limiting examples, an immunoglobulin hinge region can include one of the following amino acid sequences: DKTHT (SEQ ID NO: 217); CPPC (SEQ ID NO: 218); CPEPKSCDTPPPCPR (SEQ ID NO: 219) (see, e.g., Glaser et al., J. Biol. Chem. (2005) 280:41494-41503); ELKTPLGDTTHT (SEQ ID NO: 220); KSCDKTHTCP (SEQ ID NO: 221); KCCVDCP (SEQ ID NO: 222); KYGPPCP (SEQ ID NO: 223); EPKSCDKTHTCPPCP (SEQ ID NO: 224) (human IgGl hinge); ERKCCVECPPCP (SEQ ID NO: 225) (human IgG2 hinge); ELKTPLGDTTHTCPRCP (SEQ ID NO: 226) (human IgG3 hinge); SPNMVPHAHHAQ (SEQ ID NO: 227) (human IgG4 hinge); and the like. Page 90 of 203 1097657169\1\AMERICAS [00320] The hinge region can comprise an amino acid sequence of a human IgGl, IgG2, IgG3, or IgG4, hinge region. In one embodiment, the hinge region can include one or more amino acid substitutions and/or insertions and/or deletions compared to a wild-type (naturally-occurring) hinge region. For example, His229 of human IgGl hinge can be substituted with Tyr, so that the hinge region comprises the sequence EPKSCDKTYTCPPCP (SEQ ID NO: 228); see, e.g., Yan et al., J. Biol. Chem. (2012) 287: 5891-5897. [00321] In certain embodiments, the hinge region can comprise an amino acid sequence derived from human CD8, or a variant thereof. In certain embodiments, the CAR comprises a CD8 alpha hinge sequence comprising the amino acid sequence set forth in SEQ ID NO: 209. In certain embodiments, the CAR comprises a hinge and transmembrane domain sequence comprising the amino acid sequence set forth in SEQ ID NO: 213. 3. Costimulatory Domain [00322] In embodiments, a CAR encoded by a nucleic acid may further comprise at least one costimulatory domain, wherein the costimulatory domain comprises functional costimulatory signaling domain derived from e.g., a MHC class I molecule, TNF receptor proteins, immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocytic activation molecules (SLAM proteins), activating NK cell receptors, BTLA, a Toll ligand receptor, and the like. For example, it is within the scope of this disclosure that the CAR can include 2, 3, 4 or more costimulatory domains. It is also within the scope of this disclosure that when more than one costimulatory domain is included, the costimulatory domains may be the same, or they may be different. In embodiments, the costimulatory domains are derived from one or more of TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, CARD11, B7-H3, CEACAM1, CRTAM, CD2, CD3C, CD4, CD7, CD8α, CD8β, CD11a, CD11b, CD11c, CD11d, IL2Rβ, IL2γ, IL7Rα, IL4R, IL7R, IL15R, IL21R, CD18, CD19, CD19a, CD27, CD28, CD29, CD30, CD40, CDS, CD49a, CD49D, CD49f, CD54 (ICAM), CD69, CD70, CD80, CD83, CD84, CD86, CD96 (Tactile), CD100 (SEMA4D), CD103, CD134 (OX40), CD137 (4-1BB), CD152 (CTLA-4), CD160 (BY55), CD162 (SELPLG), CD244 (2B4), CD270 (HVEM), CD226 (DNAM1), CD229 (Ly9), CD278 (ICOS), ICAM-1, LFA-1 (CD11a/CD18), FcR, FcγRI, FcγRII, FcγRIII, LAT, NKG2C, SLP76, TRIM, ZAP70, GITR, BAFFR, LTBR, LAT, GADS, LIGHT, HVEM (LIGHTR), KIRDS2, ITGA4, ITGA6, ITGAD, ITGAE, ITGAL, ITGAM, ITGAX, ITGB1, Page 91 of 203 1097657169\1\AMERICAS ITGB2, ITGB7, NKG2C, NKG2D, IA4, VLA-1, VLA-6, SLAM (SLAMF1, CD150, IPO-3), SLAMF4, SLAMF6 (NTB-A, Ly108), SLAMF7, SLAMF8 (BLAME), SLP-76, PAG/Cbp, NKp80 (KLRF1), NKp44, NKp30, NKp46, BTLA, JAML, CD150, PSGL1, TSLP, TNFR2, and TRANCE/RANKL, or a portion thereof, and combinations thereof. [00323] In embodiments, a nucleic acid encoding a CAR encodes at least one 4-1BB costimulatory domain, and optionally a second costimulatory domain selected from 4-1BB, 2B4, ICOS, CD28, OX40, and CD27 costimulatory domains, or any of the above-mentioned costimulatory domains. In embodiments, the nucleic acid encodes at least two 4-1BB costimulatory domains, or at least two 4-1BB costimulatory domains in combination with one, two, three, or four, or more, costimulatory domains selected from 4-1BB, ICOS, CD28, OX40, and CD27, or any of the above-mentioned costimulatory domains. In embodiments, the 4-1BB costimulatory domain comprises an amino acid sequence set forth as SEQ ID NO: 202. In embodiments, the 4-1BB costimulatory domain comprises an amino acid sequence having at least one, at least two, or at least three or more modifications of an amino acid sequence of SEQ ID NO: 202. In embodiments, the 4-1BB costimulatory domain is substantially similar to the 4-1BB costimulatory domain comprising SEQ ID NO: 202. [00324] In embodiments, a nucleic acid encoding a CAR encodes at least one CD27 costimulatory domain, and optionally at least one second costimulatory domain selected from 4- 1BB, ICOS, CD28, OX40, 2B4, and CD27 costimulatory domains, or any of the above-mentioned costimulatory domains. In embodiments, the nucleic acid encodes at least one CD27 costimulatory domain, and a 4-IBB costimulatory domain. In embodiments, the nucleic acid encodes two CD27 costimulatory domains, and at least one second costimulatory domain selected from a 4-1BB, ICOS, CD28, and CD27. In embodiments, the CD27 costimulatory domain comprises SEQ ID NO: 208. In embodiments, the CD27 costimulatory domain comprises an amino acid sequence having at least one, at least two, at least three or more modifications of an amino acid sequence of SEQ ID NO: 208. In embodiments, the CD27 costimulatory domain is substantially similar to the CD27 costimulatory domain comprising SEQ ID NO: 208. [00325] In embodiments, a nucleic acid encoding a CAR encodes at least one CD28 costimulatory domain, and optionally a second costimulatory domain selected from 4-1BB, 2B4, ICOS, CD28, OX40, and CD27 costimulatory domains, or any of the above-mentioned Page 92 of 203 1097657169\1\AMERICAS costimulatory domains. In embodiments, the nucleic acid encodes at least two CD28 costimulatory domains, or at least two CD28 costimulatory domains in combination with one, two, three, or four, or more, costimulatory domains selected from a 4-1BB, ICOS, CD28, OX40, and CD27, or any of the above-mentioned costimulatory domains. In embodiments, the CD28 costimulatory domain comprises SEQ ID NO: 204. In embodiments, the CD28 costimulatory domain comprises SEQ ID NO: 205. Included in SEQ ID NO: 204 and SEQ ID NO: 205 are three subdomains YMNM, PRRP, and PYAP, that are capable to regulate signaling pathways. In embodiments, a disclosed CAR comprises mutation or deletion of one or more of the subdomains (see e.g., WO2019010383). In embodiments, the CD28 costimulatory domain comprises an amino acid sequence having at least one, at least two, at least three or more modifications of an amino acid sequence of SEQ ID NO: 204, or an amino acid sequence of SEQ ID NO: 205. In embodiments, the CD28 costimulatory domain is substantially similar to the CD28 costimulatory domain comprising SEQ ID NO: 204. In embodiments, the CD28 costimulatory domain is substantially similar to the CD28 costimulatory domain comprising SEQ ID NO: 205. [00326] In embodiments, a nucleic acid encoding a CAR encodes at least one ICOS costimulatory domain, and optionally a second costimulatory domain selected from 4-1BB, 2B4, ICOS, CD28, OX40, and CD27 costimulatory domains, or any of the above-mentioned costimulatory domains. In embodiments, the nucleic acid encodes at least two ICOS costimulatory domains, or at least two ICOS costimulatory domains in combination with one, two, three, or four, or more, costimulatory domains selected from 4-1BB, ICOS, CD28, OX40, and CD27, or any of the above-mentioned costimulatory domains. In embodiments, the ICOS costimulatory domain comprises SEQ ID NO: 206. In embodiments, the ICOS costimulatory domain comprises an amino acid sequence having at least one, at least two, at least three or more modifications of an amino acid sequence of SEQ ID NO: 206 (see e.g., US20170209492). In embodiments, the ICOS costimulatory domain is substantially similar to the ICOS costimulatory domain comprising SEQ ID NO: 206. [00327] In embodiments, a nucleic acid encoding a CAR encodes at least one OX40 costimulatory domain, and optionally a second costimulatory domain selected from 4-1BB, 2B4, ICOS, CD28, OX40, and CD27 costimulatory domains, or any of the above-mentioned costimulatory domains. In embodiments, the nucleic acid encodes at least two OX40 costimulatory domains, or at least two OX40 costimulatory domains in combination with one, two, three, or four, Page 93 of 203 1097657169\1\AMERICAS or more, costimulatory domains selected from 4-1BB, ICOS, CD28, OX40, and CD27, or any of the above-mentioned costimulatory domains. In embodiments, the OX40 costimulatory domain comprises SEQ ID NO: 207. In embodiments, the OX40 costimulatory domain comprises an amino acid sequence having at least one, at least two, at least three or more modifications of an amino acid sequence of SEQ ID NO: 207. In embodiments, the OX40 costimulatory domain is substantially similar to the OX40 costimulatory domain comprising SEQ ID NO: 207. 4. Intracellular Signaling Domain [00328] In embodiments, a nucleic acid encoding a CAR encodes at least one intracellular signaling domain. In embodiments, the at least one intracellular signaling domain is additional to one or more costimulatory domains. In embodiments, the one or more intracellular signaling domains are included to increase proliferation, persistence, and/or cytotoxic activity of the host cell, preferably a γδ cell, harboring the CAR as herein disclosed. For example, in some embodiments, the intracellular signaling domain(s) comprise CD3ζ, repeat (e.g., 2-5) DAP10 YINM motifs, signaling domains derived from LFA-1, DAP12, FcRγ, FcRβ, CD3γ, CD3δ, CD3ε, CD79a, CD79b, CD5, CD22, FcεRI, CD66d, and the like. It is within the scope of this disclosure that the endodomain of a disclosed CAR can include a plurality (e.g., 2, 3, 4, or more) of intracellular signaling domains. In a case where more than one intracellular signaling domain is included, the intracellular signaling domains may be the same, or they may be different. [00329] In embodiments, an intracellular signaling domain of a disclosed CAR is or comprises a CD3ζ signaling domain. In embodiments, a CD3ζ signaling domain is or comprises the amino acid sequence set forth in SEQ ID NO: 229, 231, or 232. 5. Additional Polypeptides [00330] In embodiments, an isolated nucleic acid encoding a CAR of the subject invention can also encode for one or more multicistronic linker region(s) configured to facilitate translation of the CAR polypeptide and one or more additional polypeptides. In embodiments, nucleic acids encoding the one or more additional polypeptides and associated linker region can be positioned at the 3’ end of the isolated nucleic acid, or at the 5’ end of the isolated nucleic acid, or in some examples at both the 5’ end and the 3’ end of the isolated nucleic acid. In some examples, the linker region(s) can encode a self-cleavage and/or a cleavage polypeptide sequence. In some examples, the self-cleavage sequence is a 2A self-cleaving sequence (e.g., T2A, P2A, E2A, F2A) Page 94 of 203 1097657169\1\AMERICAS which can induce ribosomal skipping during translation of the CAR. In embodiments, the cleavage sequence is a furin sequence. In some examples, the cleavage sequence (e.g., furin cleavage sequence as set forth in SEQ ID NO: 190) is amino terminal to a self-cleavage sequence, for example furin-P2A (FP2A). In embodiments, the multicistronic linker region encodes an internal ribosome entry site. In embodiments, the addition of an optional linker “GSG” or “SGSG” and the like can improve cleavage efficiency. In this way, the one or more additional polypeptides can be release from the CAR and directed to the secretory pathway. [00331] In embodiments, the cleavage sequence is the FP2A amino acid sequence as set forth in SEQ ID NO: 184. In embodiments, the cleavage sequence is a P2A amino acid sequence as set forth in SEQ ID NO: 186, or SEQ ID NOs: 188-189. In embodiments, the cleavage sequence is a furin amino acid sequence as set forth in SEQ ID NO: 190. In embodiments, the cleavage sequence is a F2A amino acid sequence as set forth in SEQ ID NO: 191. In embodiments, the cleavage sequence is a E2A amino acid sequence as set forth in SEQ ID NO: 192. In embodiments, the cleavage sequence is a T2A amino acid sequence as set forth in SEQ ID NO: 193. In certain aspects, multiple cleavage and/or self-cleavage sequences can be encoded carboxy-terminal to signaling and/or costimulatory domain(s) and amino-terminal to an encoded one or more additional polypeptide. In certain aspects, one or more self-cleavage sequences and one or more sequences cleaved by an endogenous protease are encoded in a construct described herein. In certain embodiments, an endogenous protease recognition site is encoded amino terminal to a self- cleavage sequence. [00332] In embodiments, the multi-cistronic linker region encodes an internal ribosome entry site. An exemplary internal ribosome entry site is encoded by the nucleotide sequence set forth in SEQ ID NO: 194. Another exemplary internal ribosome entry site is encoded by the nucleotide sequence set forth in SEQ ID NO: 195. Further suitable internal ribosome entry sites include, but are not limited to, those disclosed e.g., in Nucleic Acids Res.2010 Jan;38(Database issue):D131- 6. doi: 10.1093/nar/gkp981. Epub 2009 Nov 16, those described at iresite.org, those described in WO 2018/215787, the sequence described in GenBank accession No. KP019382.1, and the IRES element disclosed in GenBank accession No. LT727339.1. Additional multi-cistronic linker regions, including cleavage self-cleavage, and IRES elements, are disclosed in US 2018/0360992 and U.S.8,865,467. Page 95 of 203 1097657169\1\AMERICAS [00333] In embodiments, the one or more additional polypeptides include one or more soluble gamma chain cytokines expressed as separate polypeptides from the CAR. The one or more soluble common gamma chain cytokines can include but are not limited to IL-2, IL-4, IL-7, IL-9, IL-15, IL-21, IL-23. In embodiments, the common gamma chain cytokine is selected from IL-2, IL-7, and IL-15. In embodiments, the common gamma chain cytokine is IL-15. IL-15 sequences, including codon optimized nucleic acid sequences encoding soluble IL-15 (sIL-15) are disclosed herein and in WO 2007/037780. [00334] In embodiments, the one or more additional polypeptides include one or more labels or markers, for example to facilitate an ability to monitor CAR expression level, serve as an internal control, and the like. In embodiments, an isolated nucleic acid encoding a CAR encodes for a fluorescent protein, examples of which include but are not limited to green fluorescent protein (GFP), red fluorescent protein (RFP), enhanced GFP (EGFP), enhanced cyan fluorescent protein (ECFP), enhanced yellow fluorescent protein (EYFP), and the like. Other examples can include but are not limited to chloramphenicol acetyltransferase, beta-galactosidase, beta-glucuronidase, beta-lactamase, luciferase, and the like. [00335] In embodiments, the one or more additional polypeptides include a protein that is expressed on a cell surface to facilitate detection and/or isolation of cells expressing the protein, e.g., via fluorescent activated cell sorting (FACS); or for enrichment through positive selection using an antibody specific to the encoded protein, e.g., use of an antibody to purify or enrich the cells product on a column or apparatus; or for in vivo binding of an antibody to the protein to enhance or eliminate activity, e.g., to facilitate removal of cells expressing the protein in patients as a safety consideration. Exemplary proteins useful for these purposes include, e.g., CD19, CD20 (Rituxumab recognition domain), RQR8, LNGFR, a truncated form of the human epidermal growth factor receptor (EGFRt), and the like. By way of example, EGFRt can be targeted by a clinical stage antibody, where such treatment of a patient with the antibody results in elimination of cells containing an isolated nucleic acid encoding a CAR and/or the CAR as disclosed herein. See, e.g., Wang et al. A transgene-encoded cell surface polypeptide for selection, in vivo tracking, and ablation of engineered cells; Blood 2011 118(5):1255-63; Philip et al., A highly compact epitope-based marker/suicide gene for easier and safer T-cell therapy; Blood 2014124(8):1277- 87; Smith J. et al., UCART19, an allogeneic “off-the-shelf” adoptive T-cell immunotherapy against CD19+ B-cell leukemias, DOI: 10.1200/jco.2015.33.15_suppl.3069 Journal of Clinical Page 96 of 203 1097657169\1\AMERICAS Oncology 33, no.15_suppl (May 20, 2015) 3069-3069; Gouble A. et al., In Vivo Proof of Concept of Activity and Safety of UCART19, an Allogeneic “Off-the-Shelf” Adoptive T-Cell Immunotherapy Against CD19+ B-Cell Leukemias, Blood (2014) 124 (21): 4689, doi.org/10.1182/blood.V124.21.4689.4689, each of which is incorporated by reference in its entirety. [00336] In embodiments, the one or more additional polypeptides include a protein that functions to increase resistance to exhaustion and activation-induced apoptosis and/or upregulate one or more proinflammatory cytokines, costimulatory molecules and/or antigen presentation machinery. A representative example includes but is not limited to lymphotoxin beta receptor (LTBR). LTBR is typically expressed in a subset of myeloid cells but is absent in lymphocytes. When expressed in T cells, LTBR may induce transcriptional remodeling that imparts the T cell with one or more of the above-mentioned advantageous functions (Legut et al., Blood. (2021); 138(1): 1726). [00337] In embodiments, the one or more additional polypeptides includes a polypeptide that imparts host cells with the capability to resist tumor antigen-specific cellular immunity, for example that mediated by transforming growth factor beta (TGF-β). For example, an isolated nucleic acid may encode a dominant negative receptor for TGF-beta (dnTGFβR2), e.g., as described in Foster et al., J Immunother. (2008); 31: 500-505, WO2019/173324A1, WO2020/183131A1, and WO2020042647A1. Incorporation of such a dominant negative receptor for TGF-beta may provide a functional advantage over control cells that lack such a dominant negative receptor for TGF-beta in the presence of a TGF-beta-secreting tumor, including enhanced anti-tumor activity. [00338] In embodiments, an isolated nucleic acid encodes a signal peptide operably linked to facilitate directing of the one or more additional polypeptides to the secretory pathway. Such one or more additional polypeptides can be those that reside inside certain organelles, are secreted from the host cell, or are inserted into cellular membranes. In embodiments, the signal peptide comprises or consists of the amino acid sequence set forth as SEQ ID NO: 170. In embodiments, the signal peptide comprises or consists of the amino acid sequence set forth as SEQ ID NO: 196. In embodiments, the signal peptide comprises or consists of the amino acid sequence set forth as SEQ ID NO: 235. In embodiments, the signal peptide comprises or consists of the amino acid Page 97 of 203 1097657169\1\AMERICAS sequence set forth as SEQ ID NO: 239. In embodiments, the signal peptide comprises or consists of the amino acid sequence set forth as SEQ ID NO: 243. In embodiments, the signal peptide comprises or consists of the amino acid sequence set forth as SEQ ID NO: 247. [00339] In embodiments, the one or more additional polypeptides comprise or consist of the EGFRt amino acid sequence as set forth in SEQ ID NO: 237. In embodiments, the one or more additional polypeptides comprise or consist of the dominant-negative TGFβ receptor II (dnTGFβR2) amino acid sequence as set forth in SEQ ID NO: 241. In embodiments, the signal peptide comprising or consisting of the amino acid sequence set forth as SEQ ID NO: 239 is operably linked to SEQ ID NO: 241. In embodiments, the one or more additional polypeptides includes the full-length LTBR amino acid sequence as set forth in SEQ ID NO: 245. In embodiments, the signal peptide comprising or consisting of the amino acid sequence set forth as SEQ ID NO: 243 is operably linked to SEQ ID NO: 245. In embodiments, the one or more additional polypeptides includes the LNGFR amino acid sequence as set forth in SEQ ID NO: 249. In embodiments, the signal peptide comprising or consisting of the amino acid sequence set forth as SEQ ID NO: 247 is operably linked to SEQ ID NO: 249. In embodiments, the one or more additional polypeptides includes the sIL-15 amino acid sequence as set forth in SEQ ID NO: 197. In embodiments, the signal peptide comprising or consisting of the amino acid sequence as set forth in SEQ ID NO: 196 is operably linked to SEQ ID NO: 197. [00340] In embodiments, the one or more additional polypeptides includes a chimeric switch receptor comprising an extracellular domain of a TGFβ receptor for binding to TGFβ (e.g., TGFβRI and/or TGFβRII), and an intracellular domain of a cytokine receptor. The chimeric switch receptors can convert a TGFβ signal into a cytokine signal that promotes cytotoxicity. Examples of such chimeric switch receptors include those descried in WO2012138858, WO2016122738, WO2018094244, WO2014172584, WO2019109980, and WO2022037562, each of which is incorporated by reference in its entirety. [00341] In embodiments, the one or more additional polypeptides includes a dominant negative Fas (dnFas). Incorporation of such a dominant negative Fas in a T cell may provide a functional advantage over control cells that lack such a dominant negative Fas in the prevention of Fas ligand- induced apoptosis and allowing for T cell persistence and antitumor efficacy. Examples of the dnFas include that described in Yamamoto TN et al., T cells genetically engineered to overcome Page 98 of 203 1097657169\1\AMERICAS death signaling enhance adoptive cancer immunotherapy, J Clin Invest.2019 Feb 25;129(4):1551- 1565, which is incorporated by reference herein in its entirety. [00342] In embodiments, the one or more additional polypeptides includes a membrane-bound IL-12 (mbIL-12). Incorporation of such a mbIL-12 in a T cell may provide a functional advantage over control cells that lack such a mbIL-12 in enhancing effector functions of the T cells and/or limiting the systemic toxicity associated with IL-12. Examples of the mbIL-12 include those described in Hu J. et al., Cell membrane-anchored and tumor-targeted IL-12 (attIL12)-T cell therapy for eliminating large and heterogeneous solid tumors, J Immunother Cancer. 2022 Jan;10(1):e003633; Hombach A. et al., IL12 integrated into the CAR exodomain converts CD8+ T cells to poly-functional NK-like cells with superior killing of antigen-loss tumors, Mol Ther. 2022 Feb 2;30(2):593-605; and Lee EH et al., Antigen-dependent IL-12 signaling in CAR T cells promotes regional to systemic disease targeting, bioRxiv.2023 Jan 7;2023.01.06.522784, each of which is incorporated by reference herein in its entirety. [00343] In embodiments, the one or more additional polypeptides includes an antibody or fragment thereof that bind to CD70, or a CAR comprising such antibody or fragment. Incorporation of such a CD70-binding molecule in a T cell may provide a functional advantage over control cells that lack the CD70-binding molecule in reducing HvG alloreactivity by targeting CD70+ activated T cells. Examples of such CD70-binding molecules include those described in PCT/US2023/29047, which is incorporated by reference herein in its entirety. 6. Exemplary CARs [00344] The present invention provides nucleic acid molecules encoding one or more CAR constructs described herein. In one aspect, the nucleic acid molecule is provided as a messenger RNA transcript. In one aspect, the nucleic acid molecule is provided as a DNA construct. [00345] In embodiments, an isolated nucleic acid encodes SEQ ID NO: 293, a CAR polypeptide PL754 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3ζ signaling domain. [00346] In embodiments, a nucleic acid encoding a PL754 CAR comprises the sequence of SEQ ID NO: 294. Table 2 below provides annotation of the nucleotide sequence of SEQ ID NO: 294. Page 99 of 203 1097657169\1\AMERICAS Table 2. PL754 Nucleotide Sequence Annotation Annotation Numbering
Figure imgf000101_0001
[00347] In embodiments, an isolated nucleic acid encodes SEQ ID NO: 297, a CAR polypeptide PL755 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3ζ signaling domain. [00348] In embodiments, a nucleic acid encoding a PL755 CAR comprises the sequence of SEQ ID NO: 298. Table 3 below provides annotation of the nucleotide sequence of SEQ ID NO: 298. Table 3. PL755 Nucleotide Sequence Annotation
Figure imgf000101_0002
[00349] In embodiments, an isolated nucleic acid encodes SEQ ID NO: 301, a CAR polypeptide PL758 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3ζ signaling domain. Page 100 of 203 1097657169\1\AMERICAS [00350] In embodiments, a nucleic acid encoding a PL758 CAR comprises the sequence of SEQ ID NO: 302. Table 4 below provides annotation of the nucleotide sequence of SEQ ID NO: 302. Table 4. PL758 Nucleotide Sequence Annotation Annotation Numbering
Figure imgf000102_0001
[00351] In embodiments, an isolated nucleic acid encodes SEQ ID NO: 305, a CAR polypeptide PL874 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3ζ signaling domain. [00352] In embodiments, a nucleic acid encoding a PL874 CAR comprises the sequence of SEQ ID NO: 306. Table 5 below provides annotation of the nucleotide sequence of SEQ ID NO: 306. Table 5. PL874 Nucleotide Sequence Annotation
Figure imgf000102_0002
[00353] In embodiments, an isolated nucleic acid encodes SEQ ID NO: 309, a CAR polypeptide PL875 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a Page 101 of 203 1097657169\1\AMERICAS CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3ζ signaling domain. [00354] In embodiments, a nucleic acid encoding a PL875 CAR comprises the sequence of SEQ ID NO: 310. Table 6 below provides annotation of the nucleotide sequence of SEQ ID NO: 310. Table 6. PL875 Nucleotide Sequence Annotation Annotation Numbering
Figure imgf000103_0001
[00355] In embodiments, an isolated nucleic acid encodes SEQ ID NO: 313, a CAR polypeptide PL876 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3ζ signaling domain. [00356] In embodiments, a nucleic acid encoding a PL876 CAR comprises the sequence of SEQ ID NO: 314. Table 7 below provides annotation of the nucleotide sequence of SEQ ID NO: 314. Table 7. PL876 Nucleotide Sequence Annotation
Figure imgf000103_0002
Page 102 of 203 1097657169\1\AMERICAS [00357] In embodiments, an isolated nucleic acid encodes SEQ ID NO: 317, a CAR polypeptide PL1007 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3ζ signaling domain. [00358] In embodiments, a nucleic acid encoding a PL1007 CAR comprises the sequence of SEQ ID NO: 318. Table 8 below provides annotation of the nucleotide sequence of SEQ ID NO: 318. Table 8. PL1007 Nucleotide Sequence Annotation Ann t ti n N mb rin
Figure imgf000104_0001
[00359] In embodiments, an isolated nucleic acid encodes SEQ ID NO: 321, a CAR polypeptide PL1012 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3ζ signaling domain. [00360] In embodiments, a nucleic acid encoding a PL1012 CAR comprises the sequence of SEQ ID NO: 322. Table 9 below provides annotation of the nucleotide sequence of SEQ ID NO: 322. Table 9. PL1012 Nucleotide Sequence Annotation
Figure imgf000104_0002
Page 103 of 203 1097657169\1\AMERICAS CD8 Hinge and Transmembrane 784-990 Domain [00361]
Figure imgf000105_0001
In embodiments, an isolated nucleic acid encodes SEQ ID NO: 325, a CAR polypeptide PL1019 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3ζ signaling domain. [00362] In embodiments, a nucleic acid encoding a PL1019 CAR comprises the sequence of SEQ ID NO: 326. Table 10 below provides annotation of the nucleotide sequence of SEQ ID NO: 326. Table 10. PL1019 Nucleotide Sequence Annotation
Figure imgf000105_0002
[00363] In embodiments, an isolated nucleic acid encodes SEQ ID NO: 329, a CAR polypeptide PL1025 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3ζ signaling domain. [00364] In embodiments, a nucleic acid encoding a PL1025 CAR comprises the sequence of SEQ ID NO: 330. Table 11 below provides annotation of the nucleotide sequence of SEQ ID NO: 330. Table 11. PL1025 Nucleotide Sequence Annotation
Figure imgf000105_0003
Page 104 of 203 1097657169\1\AMERICAS Annotation Numbering Signal Peptide 1-60
Figure imgf000106_0001
[00365] In embodiments, an isolated nucleic acid encodes SEQ ID NO: 333, a CAR polypeptide PL1026 comprising the following domains in order: a signal peptide, a B7H6-binding domain, a a CD8 hinge and transmembrane domain, a 4-1BB costimulatory domain, and a CD3ζ signaling domain. [00366] In embodiments, a nucleic acid encoding a PL1026 CAR comprises the sequence of SEQ ID NO: 334. Table 12 below provides annotation of the nucleotide sequence of SEQ ID NO: 334. Table 12. PL1026 Nucleotide Sequence Annotation
Figure imgf000106_0002
[00367] The above-mentioned CARs and nucleic acids encoding the same include specific signal peptides. In embodiments, it may be desirable to substitute one signal peptide for another in a CAR of the present disclosure. Accordingly, in embodiments, an isolated nucleic acid comprising SEQ ID NO: 296 encodes SEQ ID NO: 295 comprising PL754 minus signal peptide; Page 105 of 203 1097657169\1\AMERICAS an isolated nucleic acid comprising SEQ ID NO: 300 encodes SEQ ID NO: 299 comprising PL755 minus signal peptide; an isolated nucleic acid comprising SEQ ID NO: 304 encodes SEQ ID NO: 303 comprising PL758 minus signal peptide; an isolated nucleic acid comprising SEQ ID NO: 308 encodes SEQ ID NO: 307 comprising PL874 minus signal peptide; a nucleic acid comprising SEQ ID NO: 312 encodes SEQ ID NO: 311 comprising PL875 minus signal peptide; an isolated nucleic acid comprising SEQ ID NO: 316 encodes SEQ ID NO: 315 comprising PL876 minus signal peptide; a nucleic acid comprising SEQ ID NO: 320 encodes SEQ ID NO: 319 comprising PL1007 minus signal peptide; an isolated nucleic acid comprising SEQ ID NO: 324 encodes SEQ ID NO: 323 comprising PL1012 minus signal peptide; an isolated nucleic acid comprising SEQ ID NO: 328 encodes SEQ ID NO: 327 comprising PL1019 minus signal peptide; an isolated nucleic acid comprising SEQ ID NO: 332 encodes SEQ ID NO: 331 comprising PL1025 minus signal peptide; or an isolated nucleic acid comprising SEQ ID NO: 336 encodes SEQ ID NO: 335 comprising PL1026 minus signal peptide. It is to be understood that any nucleic acid sequence encoding a desired signal peptide can be substituted in any of the above-referenced nucleic acid sequences which lack signal peptides, to thereby encode a CAR comprising the desired signal peptide. [00368] Furthermore, any of the above-mentioned isolated nucleic acids encoding the particular CAR polypeptides can further encode one or more additional polypeptides, as discussed herein. For example and without limitation, any of the above-mentioned nucleic acids encoding the particular CARs can include at least one multicistronic linker and a polynucleic acid encoding a dnTGFβR2 polypeptide. 7. Vectors [00369] The present invention encompasses a DNA construct comprising sequences of a CAR. The nucleic acid sequences coding for the desired molecules can be obtained using recombinant methods known in the art, such as, for example by screening libraries from cells expressing the gene, by deriving the gene from a vector known to include the same, or by isolating directly from cells and tissues containing the same, using standard techniques. Alternatively, the gene of interest can be produced synthetically, rather than cloned. [00370] The present invention provides vectors in which a DNA of the present invention is inserted. Vectors derived from retroviruses such as the lentivirus are suitable tools to achieve long- term gene transfer since they allow long-term, stable integration of a transgene and its propagation Page 106 of 203 1097657169\1\AMERICAS in daughter cells. Lentiviral vectors have the added advantage over vectors derived from onco- retroviruses such as murine leukemia viruses in that they can transduce non-proliferating cells, such as hepatocytes. They also have the added advantage of low immunogenicity. [00371] In another embodiment, the vector comprising the nucleic acid encoding the desired CAR of the invention is an adenoviral vector (A5/35). In another embodiment, the expression of nucleic acids encoding CARs can be accomplished using of transposons such as sleeping beauty, crisper, CAS9, and zinc finger nucleases. [00372] In brief summary, the expression of natural or synthetic nucleic acids encoding CARs is typically achieved by operably linking a nucleic acid encoding the CAR polypeptide or portions thereof to a promoter and incorporating the construct into an expression vector. The vectors can be suitable for replication and integration eukaryotes. Typical cloning vectors contain transcription and translation terminators, initiation sequences, and promoters useful for regulation of the expression of the desired nucleic acid sequence. [00373] The expression constructs of the present invention may also be used for nucleic acid immunization and gene therapy, using standard gene delivery protocols. Methods for gene delivery are known in the art (e.g., U.S. Pat. Nos.5,399,346, 5,580,859, 5,589,466, incorporated by reference herein in their entireties). In another embodiment, the invention provides a gene therapy vector. [00374] The nucleic acid can be cloned into a number of types of vectors. For example, the nucleic acid can be cloned into a vector including, but not limited to a plasmid, a phagemid, a phage derivative, an animal virus, and a cosmid. Vectors of particular interest include expression vectors, replication vectors, probe generation vectors, and sequencing vectors. [00375] Further, the expression vector may be provided to a cell in the form of a viral vector. Viral vector technology is well known in the art and is described, for example, in Sambrook et al. (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York), and in other virology and molecular biology manuals. Viruses, which are useful as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpes viruses, and lentiviruses. In general, a suitable vector contains an origin of replication functional in at least one organism, a promoter sequence, convenient restriction endonuclease sites, and one or more selectable markers, (e.g., WO 01/96584; WO 01/29058; and U.S. Pat. No.6,326,193). A number Page 107 of 203 1097657169\1\AMERICAS of viral based systems have been developed for gene transfer into mammalian cells. For example, retroviruses provide a convenient platform for gene delivery systems. A selected gene can be inserted into a vector and packaged in retroviral particles using techniques known in the art. The recombinant virus can then be isolated and delivered to cells of the subject either in vivo or ex vivo. A number of retroviral systems are known in the art. In embodiments, adenovirus vectors are used. A number of adenovirus vectors are known in the art. In one embodiment, lentivirus vectors are used. [00376] Additional promoter elements, e.g., enhancers, regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upstream of the start site, although a number of promoters have recently been shown to contain functional elements downstream of the start site as well. The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the thymidine kinase (tk) promoter, the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline. Depending on the promoter, it appears that individual elements can function either cooperatively or independently to activate transcription. [00377] One example of a suitable promoter is the immediate early cytomegalovirus (CMV) promoter sequence. This promoter sequence is a strong constitutive promoter sequence capable of driving high levels of expression of any polynucleotide sequence operatively linked thereto. Another example of a suitable promoter is Elongation Growth Factor-1α (EF-1α). However, other constitutive promoter sequences may also be used, including, but not limited to the simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, an avian leukemia virus promoter, an Epstein-Barr virus immediate early promoter, a Rous sarcoma virus promoter, as well as human gene promoters such as, but not limited to, the actin promoter, the myosin promoter, the hemoglobin promoter, and the creatine kinase promoter. Further, the invention should not be limited to the use of constitutive promoters. Inducible promoters are also contemplated as part of the invention. The use of an inducible promoter provides a molecular switch capable of turning on expression of the polynucleotide sequence which it is operatively linked when such expression is desired or turning off the expression when expression is not desired. Examples of inducible promoters include, but are not limited to a metallothionine promoter, a glucocorticoid promoter, a progesterone promoter, and a tetracycline promoter. Page 108 of 203 1097657169\1\AMERICAS [00378] In order to assess the expression of a CAR polypeptide or portions thereof, the expression vector to be introduced into a cell can also contain either a selectable marker gene or a reporter gene or both to facilitate identification and selection of expressing cells from the population of cells sought to be transfected or infected through viral vectors. In other aspects, the selectable marker may be carried on a separate piece of DNA and used in a co-transfection procedure. Both selectable markers and reporter genes may be flanked with appropriate regulatory sequences to enable expression in the host cells. Useful selectable markers include, for example, antibiotic-resistance genes, such as neo and the like. [00379] Reporter genes are used for identifying potentially transfected cells and for evaluating the functionality of regulatory sequences. In general, a reporter gene is a gene that is not present in or expressed by the recipient organism or tissue and that encodes a polypeptide whose expression is manifested by some easily detectable property, e.g., enzymatic activity. Expression of the reporter gene is assayed at a suitable time after the DNA has been introduced into the recipient cells. Suitable reporter genes may include genes encoding luciferase, beta-galactosidase, chloramphenicol acetyl transferase, secreted alkaline phosphatase, or the green fluorescent protein gene (e.g., Ui-Tei et al., 2000 FEBS Letters 479: 79-82). Suitable expression systems are well known and may be prepared using known techniques or obtained commercially. In general, the construct with the minimal 5' flanking region showing the highest level of expression of reporter gene is identified as the promoter. Such promoter regions may be linked to a reporter gene and used to evaluate agents for the ability to modulate promoter-driven transcription. [00380] Methods of introducing and expressing genes into a cell are known in the art. In the context of an expression vector, the vector can be readily introduced into a host cell, e.g., mammalian, bacterial, yeast, or insect cell by any method in the art. For example, the expression vector can be transferred into a host cell by physical, chemical, or biological means. [00381] Physical methods for introducing a polynucleotide into a host cell include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and the like. Methods for producing cells comprising vectors and/or exogenous nucleic acids are well- known in the art. See, for example, Sambrook et al. (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York). One method for the introduction of a polynucleotide into a host cell is calcium phosphate transfection. Page 109 of 203 1097657169\1\AMERICAS [00382] Biological methods for introducing a polynucleotide of interest into a host cell include the use of DNA and RNA vectors. Viral vectors, and especially retroviral vectors, have become the most widely used method for inserting genes into mammalian, e.g., human cells. Other viral vectors can be derived from lentivirus, poxviruses, herpes simplex virus I, adenoviruses and adeno- associated viruses, and the like. See, for example, U.S. Pat. Nos.5,350,674 and 5,585,362. [00383] Chemical means for introducing a polynucleotide into a host cell include colloidal dispersion systems, such as macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. An exemplary colloidal system for use as a delivery vehicle in vitro and in vivo is a liposome (e.g., an artificial membrane vesicle). [00384] In the case where a non-viral delivery system is utilized, an exemplary delivery vehicle is a liposome. The use of lipid formulations is contemplated for the introduction of the nucleic acids into a host cell (in vitro, ex vivo or in vivo). In another aspect, the nucleic acid may be associated with a lipid. The nucleic acid associated with a lipid may be encapsulated in the aqueous interior of a liposome, interspersed within the lipid bilayer of a liposome, attached to a liposome via a linking molecule that is associated with both the liposome and the oligonucleotide, entrapped in a liposome, complexed with a liposome, dispersed in a solution containing a lipid, mixed with a lipid, combined with a lipid, contained as a suspension in a lipid, contained or complexed with a micelle, or otherwise associated with a lipid. Lipid, lipid/DNA or lipid/expression vector associated compositions are not limited to any particular structure in solution. For example, they may be present in a bilayer structure, as micelles, or with a “collapsed” structure. They may also simply be interspersed in a solution, possibly forming aggregates that are not uniform in size or shape. Lipids are fatty substances which may be naturally occurring or synthetic lipids. For example, lipids include the fatty droplets that naturally occur in the cytoplasm as well as the class of compounds which contain long-chain aliphatic hydrocarbons and their derivatives, such as fatty acids, alcohols, amines, amino alcohols, and aldehydes. [00385] Lipids suitable for use can be obtained from commercial sources. For example, dimyristyl phosphatidylcholine (“DMPC”) can be obtained from Sigma, St. Louis, Mo.; dicetyl phosphate (“DCP”) can be obtained from K & K Laboratories (Plainview, N.Y.); cholesterol (“Choi”) can be obtained from Calbiochem-Behring; dimyristyl phosphatidylglycerol (“DMPG”) Page 110 of 203 1097657169\1\AMERICAS and other lipids may be obtained from Avanti Polar Lipids, Inc. (Birmingham, AL). Stock solutions of lipids in chloroform or chloroform/methanol can be stored at about -20.degree. C. Chloroform is used as the only solvent since it is more readily evaporated than methanol. “Liposome” is a generic term encompassing a variety of single and multilamellar lipid vehicles formed by the generation of enclosed lipid bilayers or aggregates. Liposomes can be characterized as having vesicular structures with a phospholipid bilayer membrane and an inner aqueous medium. Multilamellar liposomes have multiple lipid layers separated by aqueous medium. They form spontaneously when phospholipids are suspended in an excess of aqueous solution. The lipid components undergo self-rearrangement before the formation of closed structures and entrap water and dissolved solutes between the lipid bilayers (Ghosh et al., 1991 Glycobiology 5: 505-10). However, compositions that have different structures in solution than the normal vesicular structure are also encompassed. For example, the lipids may assume a micellar structure or merely exist as nonuniform aggregates of lipid molecules. Also contemplated are lipofectamine-nucleic acid complexes. [00386] Regardless of the method used to introduce exogenous nucleic acids into a host cell or otherwise expose a cell to the inhibitor of the present invention, in order to confirm the presence of the recombinant DNA sequence in the host cell, a variety of assays may be performed. Such assays include, for example, “molecular biological” assays well known to those of skill in the art, such as Southern and Northern blotting, RT-PCR and PCR; “biochemical” assays, such as detecting the presence or absence of a particular peptide, e.g., by immunological means (ELISAs and Western blots) or by assays described herein to identify agents falling within the scope of the invention. 8. Host Cells [00387] CAR polypeptides of the present disclosure may be expressed via their corresponding nucleic acid constructs in a wide variety of host cells. In embodiments, the host cells are mammalian cells. Host cells, as described herein, can be stored, e.g., cryopreserved, for use in adoptive cell transfer. In embodiments, the host cells are stored prior to engineering the cells to express a CAR polypeptide. In embodiments, the cells are engineered to express a CAR polypeptide and then the cells are stored. Page 111 of 203 1097657169\1\AMERICAS [00388] Preferred host cells for use with the CARs of the present disclosure comprise immune cells. Such cells may be obtained from the subject to be treated (i.e. are autologous) or, alternatively, immune cell lines or donor immune cells (allogeneic, syngeneic) can be used. Immune cells can be obtained from a number of sources, including from peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors. Immune cells can be obtained from blood collected from a subject using any number of techniques known to the skilled artisan, such as Ficoll™ separation. For example, cells from the circulating blood of an individual may be obtained by apheresis. In embodiments, immune cells are isolated from peripheral blood lymphocytes by lysing the red blood cells and depleting the monocytes, for example, by centrifugation through a PERCOLL™ gradient or by counterflow centrifugal elutriation. A specific subpopulation of immune cells can be further isolated by positive or negative selection techniques. For example, immune cells can be isolated using a combination of antibodies directed to surface markers unique to the positively selected cells, e.g., by incubation with antibody- conjugated beads for a time period sufficient for positive selection of the desired immune cells. Alternatively, enrichment of immune cell populations can be accomplished by negative selection using a combination of antibodies directed to surface markers unique to the negatively selected cells. Other specific manners of isolation and/or enrichment are disclosed herein. [00389] In embodiments, the immune cells comprise any leukocyte involved in defending the body against infectious disease and foreign materials. For example, the immune cells can comprise lymphocytes, monocytes, macrophages, dendritic cells, mast cells, neutrophils, basophils, eosinophils, or any combinations thereof. For example, immune cells relevant to the present disclosure can include but are not limited to αβ T cells, ^ ^ ^T cells, NK cells, NKT cells, ^ ^ ^ ^ ^ ^ ^cells, B cells, innate lymphoid cells (ILCs), cytokine induced killer (CIK) cells, cytotoxic T lymphocytes (CTLs), lymphokine activated killer (LAK) cells, regulatory T cells, and the like. In embodiments, preferred immune cells comprise αβ T cells, ^ ^ ^T cells, NK cells, NKT cells, ^ ^ ^ ^ ^ ^ ^cells, and/or, in some examples, macrophages. In embodiments, preferred immune cells comprise ^ ^ ^T cells. In embodiments, the immune cells relevant to the present disclosure comprise allogeneic cells, autologous cells, or syngeneic cells. Page 112 of 203 1097657169\1\AMERICAS [00390] Accordingly, aspects of the invention include host cells, γδ T cells in some preferred embodiments, that functionally express an isolated nucleic acid described herein, and thereby express a CAR on the surface of the cell. [00391] Aspects of the invention can additionally or alternatively include host cells, preferably γδ T cells, having in vitro or in vivo cytotoxic activity against a tumor cell that exhibits cell surface expression of B7H6. [00392] In some cases, the cytotoxic activity is innate activity. In some cases, the cytotoxicity is at least in part, significantly (> about 25%), or entirely, due to the presence of a CAR construct having a binding domain that specifically binds B7H6 expressed on the surface of the tumor cell. In some cases, the host cells, preferably γδ T cells, exhibit tumor cell killing activity that is greater than an innate level of in vitro and/or in vivo tumor cell killing activity in a control cell of the same cell type. In some cases, the control cell does not comprise a CAR construct. In some cases, the control cell comprises a CAR construct lacking a binding domain described herein, a hinge region described herein, a transmembrane domain described herein, an intracellular signaling domain described herein, and/or a costimulatory domain described herein. [00393] In some cases, the cytotoxicity is at least in part, significantly (> about 25%), or entirely, due to the presence of a CAR construct having a binding domain that specifically binds B7H6 or an epitope within B7H6. In some cases, the host cells, preferably γδ T cells, functionally express a B7H6-specific CAR encoded by an isolated nucleic acid described herein. [00394] In embodiments where the host cells are γδ T cells, the γδ T cells can exhibit HLA- restricted (e.g., HLA class I restricted) cytotoxicity. In other embodiments, most (>50%), substantially all (>90%), or all of the cytotoxic activity is not HLA-restricted (e.g., HLA class I restricted). HLA-restricted cytotoxic activity can be assessed by comparing in vitro cytotoxicity against an HLA (e.g., HLA class I) (null) tumor cell line versus in vitro cytotoxicity against an HLA+ (e.g., HLA class I+) tumor cell line. In embodiments, the HLA-restricted cytotoxic activity is at least in part, significantly (>25%), or entirely, provided by the use of a T cell Receptor-like binding domain. T cell receptor like binding domains are binding domains that specifically recognize the antigen when presented on the surface of a cell in complex with an MHC molecule. T cell Receptor-like binding domains are further described, e.g., in WO 2016/199141. Page 113 of 203 1097657169\1\AMERICAS [00395] Host cells described herein, preferably γδ T cells, can exhibit robust and/or persistent tumor cell killing activity. In some cases, the tumor cell killing activity can persist for at least about 6 days to 120 days, or for at least about 6 days to 180 days, from first contact with a tumor cell. In some cases, the tumor cell killing activity of a host cell described herein, preferably a γδ T cell, or a progeny thereof, can persist for at least about 6 days to 120 days, or for at least about 6 days to 180 days, from first contact with a tumor cell, or from administration of the host cell. This persistent tumor cell killing activity can be exhibited in vitro, in vivo, or both in vitro and in vivo. [00396] Aspects of the invention can additionally or alternatively include host cells, preferably γδ T cells, that proliferate in response to contact with cells that exhibit cell surface expression, or overexpression, of B7H6. The cells that exhibit cell surface expression, or overexpression, of B7H6 can be tumor cells or can be non-tumor cells. In some cases, the proliferation is an innate activity. In some cases, the proliferation is at least in part, significantly (> about 20% or > about 25%), or entirely, due to the presence of a CAR construct having a binding domain that specifically binds B7H6 expressed on the surface of a tumor cell. In some cases, the host cells, preferably γδ T cells, exhibit a greater level of in vitro and/or in vivo proliferation as compared to a control cell of the same type. In some cases, the control cell does not comprise a CAR construct. In some cases, the control cell comprises a CAR construct lacking a binding domain described herein, a hinge region described herein, a transmembrane domain described herein, an intracellular signaling domain described herein, and/or a costimulatory domain described herein. [00397] Host cells as described herein, preferably γδ T cells, can exhibit robust and/or persistent proliferation in a host organism that comprises a cell, for example a tumor cell, that exhibits cell surface expression, or overexpression, of B7H6. In some cases, the proliferation can persist for at least about 6 days to 120 days, or for at least about 6 days to 180 days, from first contact with a tumor cell or from a date of administration of the host cell, preferably a γδ T cell, to the host organism. In some cases, the proliferation of a host cell, preferably a γδ T cell described herein, or a progeny thereof, in the host organism that comprises the cell that exhibits cell surface expression, or overexpression, of B7H6 can persist for at least about 6 days to 120 days, or for at least about 6 days to 180 days, from first contact with a B7H6-expressing cell or from the date of first administration of the host cell, preferably a γδ T cell, to the host organism. In some cases, the proliferation in the host organism is at least in part, significantly (> about 20% or > about 25%), Page 114 of 203 1097657169\1\AMERICAS or entirely, due to the presence of a CAR construct having a binding domain that specifically binds B7H6 or an epitope within B7H6. In some cases, host cells, preferably γδ T cells, exhibiting proliferation in the host organism comprising a cell that exhibits cell surface expression of B7H6 functionally express a B7H6 specific CAR encoded by an isolated nucleic acid described herein. [00398] In embodiments, the host cells, preferably γδ T cells described herein, express, or persistently express, pro-inflammatory cytokines such as tumor necrosis factor alpha or interferon gamma after contact with a B7H6-expressing cell. In embodiments, the host cells described herein, or progeny thereof, express, or persistently express, pro-inflammatory cytokines such as tumor necrosis factor alpha or interferon gamma after contact with the B7H6-expressing cell, e.g., in a host organism comprising the B7H6-expressing cell. [00399] In embodiments, a γδ T cell, or a pharmaceutical composition containing the γδ T cell, exhibits essentially no, or no graft versus host response when introduced into an allogeneic host. In embodiments, the γδ T cell, or a pharmaceutical composition containing the γδ T cell, exhibits a clinically acceptable level of graft versus host response when introduced into an allogeneic host. In embodiments, a clinically acceptable level is an amount of graft versus host response that does not require cessation of a γδ T cell treatment to achieve a therapeutically effective treatment. In embodiments, a clinically acceptable level of graft versus host response (GvHD) is an acute response that is less severe than Grade C according to an applicable IBMTR grading scale. The severity of acute graft versus host response is determined by an assessment of the degree of involvement of the skin, liver, and gastrointestinal tract. The stages of individual organ involvement are combined to produce an overall grade, which has prognostic significance. Grade I(A) GvHD is characterized as mild disease, grade II(B) GvHD as moderate, grade III(C) as severe, and grade IV(D) life-threatening. The IBMTR grading system defines the severity of acute GvHD as follows (Rowlings et al., Br J Haematol 1997; 97:855): ●Grade A – Stage 1 skin involvement alone (maculopapular rash over <25 percent of the body) with no liver or gastrointestinal involvement ●Grade B – Stage 2 skin involvement; Stage 1 to 2 gut or liver involvement ●Grade C – Stage 3 involvement of any organ system (generalized erythroderma; bilirubin 6.1 to 15.0 mg/dL; diarrhea 1500 to 2000 mL/day) Page 115 of 203 1097657169\1\AMERICAS ●Grade D – Stage 4 involvement of any organ system (generalized erythroderma with bullous formation; bilirubin >15 mg/dL; diarrhea >2000 mL/day OR pain OR ileus). See also, Schoemans et al., Bone Marrow Transplantation volume 53, pages1401–1415 (2018), e.g., at Tables 1 and 2, which discloses criteria for assessing and grading acute GvHD. [00400] In embodiments, a γδ T cell, or a pharmaceutical composition containing the γδ T cell, exhibits reduced or substantially reduced graft versus host response when introduced into an allogeneic host as compared to a graft versus host response exhibited by control αβ T cells, or a control pharmaceutical composition comprising the control αβ T cells, administered to an allogeneic host. In some cases, the control αβ T cell is an allogeneic non-engineered control αβ T cell. In some cases, the control αβ T cell does not comprise a CAR or does not comprise the same CAR as a reference γδ T cell. [00401] In embodiments, host cells, preferably γδ T cells, described herein can be modified to comprise one or more gene edits. Discussed herein, gene editing is a type of genetic engineering in which nucleotide(s)/nucleic acid(s) is/are inserted, deleted, and/or substituted in a DNA sequence, such as the genome of a γδ T cell. Targeted gene editing enables insertion, deletion, and/or substitution at pre-selected sites in the genome of a targeted cell. When a sequence of an endogenous gene is edited, for example by deletion, insertion, or substitution of nucleotide(s)/nucleic acid(s), the endogenous gene comprising the affected sequence may be knocked-out or knocked-down due to the sequence alteration. Therefore, targeted editing may be used to disrupt endogenous gene expression. Discussed herein, a “disrupted gene” refers to a gene comprising an insertion, deletion or substitution relative to an endogenous gene such that expression of a functional protein from the endogenous gene is reduced or inhibited. As used herein, “disrupting a gene” refers to a method of inserting, deleting, or substituting at least one nucleotide/nucleic acid in an endogenous gene such that expression of a functional protein from the endogenous gene is reduced or inhibited. Methods of disrupting a gene are known to those of skill in the art, and described, e.g., in United States Patent No.11254912, incorporated herein by reference in its entirety. [00402] In embodiments, a nuclease-dependent approach can be used to conduct targeted gene editing of a T cell. Such a nuclease-dependent approach can achieve targeted editing through the specific introduction of double strand breaks (DSBs) by specific endonucleases. Such nuclease- Page 116 of 203 1097657169\1\AMERICAS dependent targeted editing utilizes DNA repair mechanisms, for example, non-homologous end joining (NHEJ), which occurs in response to DSBs. DNA repair by NHEJ often leads to random insertions or deletions (indels) of a small number of endogenous nucleotides. In contrast to NHEJ mediated repair, repair can also occur by a homology directed repair (HDR). When a donor template containing exogenous genetic material flanked by a pair of homology arms is present, the exogenous genetic material can be introduced into the genome by HDR, which results in targeted integration of the exogenous genetic material. Available endonucleases capable of introducing specific and targeted DSBs include, but are not limited to, zinc-finger nucleases (ZFN), transcription activator-like effector nucleases (TALEN), and RNA-guided CRISPR-Cas9 nuclease (CRISPR/Cas9; Clustered Regular Interspaced Short Palindromic Repeats Associated 9). Discussed herein, a CRISPR system, or CRISPR nuclease system can include a non-coding RNA molecule (e.g., guide RNA) that binds DNA and Cas proteins (e.g., Cas9) with nuclease functionality (Sander et al., Nature Biotechnology (2014); 32:347-355; Hsu et al., Cell (2014); 157(6):1262-1278). [00403] In embodiments, a host cell, preferably a γδ T cell, comprises one or more disrupted genes. For example, one or more genes whose expression is disrupted can comprise adenosine A2a receptor (ADORA), CD276, V-set domain containing T cell activation inhibitor 1 (VTCN1), B and T lymphocyte associated (BTLA), cytotoxic T-lymphocyte-associated protein 4 (CTLA4), indoleamine 2,3-dioxygenase 1 (IDO1), killer cell immunoglobulin-like receptor, three domains, long cytoplasmic tail, 1 (KIR3DL1), lymphocyte-activation gene 3 (LAG3), programmed cell death 1 (PD-1), hepatitis A virus cellular receptor 2 (HAVCR2), V-domain immunoglobulin suppressor of T-cell activation (VISTA), natural killer cell receptor 2B4 (CD244), cytokine inducible SH2-containing protein (CISH), hypoxanthine phosphoribosyltransferase 1 (HPRT), adeno-associated virus integration site (AAVS SITE (E.G. AAVS1, AAVS2, ETC.)), or chemokine (C—C motif) receptor 5 (gene/pseudogene) (CCR5), CD160 molecule (CD160), T- cell immunoreceptor with Ig and ITIM domains (TIGIT), CD96 molecule (CD96), cytotoxic and regulatory T-cell molecule (CRTAM), leukocyte associated immunoglobulin like receptor 1(LAIR1), sialic acid binding Ig like lectin 7 (SIGLEC7), sialic acid binding Ig like lectin 9 (SIGLEC9), tumor necrosis factor receptor superfamily member 10b (TNFRSF10B), tumor necrosis factor receptor superfamily member 10a (TNFRSF10A), caspase 8 (CASP8), caspase 10 (CASP10), caspase 3 (CASP3), caspase 6 (CASP6), caspase 7 (CASP7), Fas associated via death Page 117 of 203 1097657169\1\AMERICAS domain (FADD), Fas cell surface death receptor (FAS), transforming growth factor beta receptor II (TGFBRII), transforming growth factor beta receptor I (TGFBR1), SMAD family member 2 (SMAD2), SMAD family member 3 (SMAD3), SMAD family member 4 (SMAD4), SKI proto- oncogene (SKI), SKI-like proto-oncogene (SKIL), TGFB induced factor homeobox 1 (TGIF1), interleukin 10 receptor subunit alpha (IL10RA), interleukin 10 receptor subunit beta (IL10RB), heme oxygenase 2 (HMOX2), interleukin 6 receptor (IL6R), interleukin 6 signal transducer (IL6ST), c-src tyrosine kinase (CSK), phosphoprotein membrane anchor with glycosphingolipid microdomains 1(PAG1), signaling threshold regulating transmembrane adaptor 1 (SIT1), forkhead box P3 (FOXP3), PR domain 1 (PRDM1), basic leucine zipper transcription factor, ATF-like (BATF), guanylate cyclase 1, soluble, alpha 2 (GUCY1A2), guanylate cyclase 1, soluble, alpha 3 (GUCY1A3), guanylate cyclase 1, soluble, beta 2 (GUCY1B2), guanylate cyclase 1, soluble, beta 3 (GUCY1B3), cytokine inducible SH2-containing protein (CISH), prolyl hydroxylase domain (PHD1, PHD2, PHD3) family of proteins, Cbl proto-oncogene B (CBL-B), Zinc Finger Protein 91 (ZFP91), Roquin, CD58, ICAM-1, or any combination thereof. [00404] In embodiments, a gene whose expression is disrupted is CISH, a negative regulator of TCR signaling. Disruption of the CISH gene may provide a functional advantage over control cells that have an intact CISH gene in improving the sensitivity to certain cytokines (e.g., IL-2/IL- 15), increasing T cell proliferation, and/or limiting T cell exhaustion. In some examples, the CISH gene may be disrupted by methods described in Daher M. et al, Targeting a cytokine checkpoint enhances the fitness of armored cord blood CAR-NK cells, Blood. 2021 Feb 4;137(5):624-636, which is incorporated by reference herein in its entirety. In some examples, the CISH gene is disrupted by gene editing using an RNA-guided nuclease system comprising one or more guide RNAs comprising the sequences of any one of SEQ ID NOs: 198-201 and 339-340. [00405] In embodiments, a gene whose expression is disrupted is CBL-B, a negative regulator of T cell activation. Disruption of the CBL-B gene may provide a functional advantage over control cells that have an intact CBL-B gene in enhancing T cell activation. In some examples, the CBL- B gene may be disrupted by methods described in Augustin R. et al., Targeting Cbl-b in cancer immunotherapy, J Immunother Cancer. 2023 Feb;11(2):e006007; Hooper K. et al., Knockout of CBLB Greatly Enhances Anti-Tumor Activity of CAR T Cells, Blood (2018) 132 (Supplement 1): 338; and Guo X. et al., CBLB ablation with CRISPR/Cas9 enhances cytotoxicity of human placental stem cell-derived NK cells for cancer immunotherapy, J Immunother Cancer. 2021 Page 118 of 203 1097657169\1\AMERICAS Mar;9(3):e001975, each of which is incorporated by reference herein in its entirety. In one example, the CBL-B gene is disrupted by gene editing using a CRISPR-Cas system comprising one or more guide RNAs comprising the sequence of any one of SEQ ID NOs: 341-344. [00406] In embodiments, a gene whose expression is disrupted is Roquin (e.g., Roquin-1). Disruption of the Roquin gene may provide a functional advantage over control cells that have an intact Roquin gene in increasing T cell proliferation and enhancing antitumor activity. In some examples, the Roquin gene may be disrupted by methods described in Mai D et al., Combined disruption of T cell inflammatory regulators Regnase-1 and Roquin-1 enhances antitumor activity of engineered human T cells, Proc Natl Acad Sci U S A. 2023 Mar 21;120(12):e2218632120, which is incorporated by reference herein in its entirety. [00407] In embodiments, a gene whose expression is disrupted is ZFP91. Disruption of the ZFP91 gene may provide a functional advantage over control cells that have an intact ZFP91 gene in improving T cell glycolytic fitness and effector function. In some examples, the ZFP91 gene may be disrupted by method described in Wang F. et al., J Clin Invest. 2021 Oct 1;131(19):e144318, which is incorporated by reference herein in its entirety. [00408] In embodiments, a gene whose expression is disrupted is CD58. In embodiments, a gene whose expression is disrupted is ICAM-1. In embodiments, both CD58 and ICAM-1 are disrupted. Disruption of the CD58 and/or ICAM-1 genes may provide a functional advantage over control cells that have an intact CD58 and/or ICAM-1 gene in disrupting T cell adhesion and co- stimulatory interactions to reduce Host vs Graft allocytotoxicity. In one example, the ICAM-1 gene may be disrupted as described in Teo HY et al. IL12/18/21 Preactivation Enhances the Antitumor Efficacy of Expanded γδT Cells and Overcomes Resistance to Anti-PD-L1 Treatment, Cancer Immunol Res.2023 Jul 5;11(7):978-999, which is incorporated by reference in its entirety. In one example, the ICAM-1 gene is disrupted by gene editing using an RNA-guided nuclease system (e.g., CRISPR-Cas or CRISPR-Mad7 system) comprising one or more guide RNAs comprising the sequences of any one of SEQ ID NOs: 349-350. In one example, the CD58 gene is disrupted by gene editing using an RNA-guided nuclease system (e.g., CRISPR-Cas or CRISPR- Mad7 system) comprising one or more guide RNAs comprising the sequences of any one of SEQ ID NOs: 351-352. Page 119 of 203 1097657169\1\AMERICAS [00409] In embodiments, host cells of the present disclosure, preferably γδ T cells, can be modified to include a nucleic acid construct that encodes a protein which imparts a desired functionality to the host cells. For example and without limitation, such a nucleic acid may encode for a chimeric DAP10 adaptor polypeptide, described in U.S. Provisional Application No. 63/272,613 and U.S. Provisional Application No. 63/347,194, the contents of each of which is hereby expressly incorporated herein by reference in their entirety. In embodiments, a chimeric DAP10 adaptor polypeptide is capable of associating with a chimeric antigen receptor of the present disclosure, for example a CAR of the present disclosure may comprise a DAP10- interacting domain. In such an example, the chimeric DAP10 adaptor polypeptide may also associate with one or more additional endogenous or exogenous polypeptides, for example endogenous or exogenous NKG2D. In embodiments, a chimeric DAP10 adaptor polypeptide may not associate with a CAR of the present disclosure, but instead may interact with an endogenous or exogenous polypeptide (e.g., NKG2D) that includes a DAP10-interacting domain. G. Diagnostic and Detection Methods and Compositions [00410] It is to be understood by a skilled artisan that the antibody, antigen-binding fragments, or compositions provided herein can be used in diagnostic or therapeutic procedures. [00411] One embodiment provided herein is directed to a method of diagnosing the presence of a tumor or a cancer growth in a subject. In an embodiment, a method includes determining the presence of B7H6 in a sample suspected of containing the B7H6 polypeptide. The sample may contain cells, which may be cancer cells, suspected of expressing B7H6. In embodiment, a method comprises exposing the sample to an antibody or antigen-binding fragment as herein disclosed, whereby specific binding of the antibody or antigen-binding fragment to the sample is indicative of the presence of a tumor or cancer growth in the subject. The antibody employed in the method may optionally be detectably labeled, attached to a solid support, or the like. Detectable labels can include but are not limited to a photoactivatable agent, a fluorophore, a radioisotope, a bioluminescent protein, a bioluminescent peptide, a fluorescent tag, a fluorescent protein, or a fluorescent peptide. Exemplary assays for achieving detection of a signal from a label includes assays routinely used in the art such as, but not limited to flow cytometry, ELISA, Western blotting, immunohistochemistry, membrane assays, and microscopic imaging. H. Imaging of conditions associated with abnormal B7H6 expression Page 120 of 203 1097657169\1\AMERICAS [00412] In one embodiment, the composition comprising an antibody or antigen-binding fragment thereof of the present invention is administered to a subject having a disease involving inappropriate expression of a target antigen, a protein or other molecule. For example, in one embodiment, the composition comprising an antibody or antibody fragment that binds to B7H6 is administered to detect the presence, abundance, location, or combination thereof of B7H6 in the subject. Within the scope of the present invention this is meant to include diseases and disorders characterized by aberrant proteins, due for example to alterations in the amount of a protein present, protein localization, posttranslational modification, conformational state, the presence of a mutant or pathogen protein, etc. Similarly, the disease or disorder may be characterized by alterations molecules including but not limited to polysaccharides and gangliosides. An overabundance may be due to any cause, including but not limited to overexpression at the molecular level, prolonged or accumulated appearance at the site of action, or increased activity of a protein relative to normal. Included within this definition are diseases and disorders characterized by a reduction of a protein. This reduction may be due to any cause, including but not limited to reduced expression at the molecular level, shortened or reduced appearance at the site of action, mutant forms of a protein, or decreased activity of a protein relative to normal. Such an overabundance or reduction of a protein can be measured relative to normal expression, appearance, or activity of a protein, and the measurement may play an important role in the development and/or clinical testing of the antibodies of the present invention. [00413] In one embodiment, antibody or antibody fragment of the present invention binds the B7H6 antigen expressed on tumor cells when administrated in a subject; in another embodiment, antibody or antibody fragment of the present invention administrated in a subject binds the antigen expressed on hematological and/or solid tumor cells, including but not limited to colon cancer, pancreatic cancer, Non-Hodgkin Lymphoma, cervical cancer, hepatic cancer, gastro-intestinal cancer, lung cancer, ovarian cancer, glioblastoma, prostate cancer, breast cancer, ovarian cancer, kidney cancer, bladder cancer, melanoma, and glioma. [00414] Detection methods for identification of binding can be direct or indirect and can include, for example, the measurement of light emission, radioisotopes, calorimetric dyes and fluorochromes. Direct detection includes methods that operate without intermediates or secondary measuring procedures to assess the amount of bound antigen or ligand. Such methods generally employ ligands that are themselves labeled by, for example, radioactive, light emitting or Page 121 of 203 1097657169\1\AMERICAS fluorescent moieties. In contrast, indirect detection includes methods that operate through an intermediate or secondary measuring procedure. These methods generally employ molecules that specifically react with the antigen or ligand and can themselves be directly labeled or detected by a secondary reagent. For example, an antibody specific for a ligand can be detected using a secondary antibody capable of interacting with the first antibody specific for the ligand, again using the detection methods described above for direct detection. Indirect methods can additionally employ detection by enzymatic labels. Moreover, for the specific example of screening for catalytic antibodies, the disappearance of a substrate or the appearance of a product can be used as an indirect measure of binding affinity or catalytic activity. I. Pharmaceutical Compositions [00415] The antibodies, antigen-binding fragments thereof, and/or engineered cells of the present disclosure can be administered to a subject per se, or in a pharmaceutical composition where it is mixed with suitable carriers or excipients. [00416] The antibodies and/or engineered cells of the invention may be administered by any route appropriate to the condition to be treated. Administration is typically parenterally, i.e. infusion, subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural. [00417] Therapeutic formulations comprising an anti-B7H6 antibody used in accordance with the present invention are prepared for storage by mixing the antibody having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington’s Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as acetate, Tris, phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents Page 122 of 203 1097657169\1\AMERICAS such as EDTA; tonicifiers such as trehalose and sodium chloride; sugars such as sucrose, mannitol, trehalose or sorbitol; surfactant such as polysorbate; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN®, PLURONICS® or polyethylene glycol (PEG). Pharmaceutical formulations to be used for in vivo administration are generally sterile. This is readily accomplished by filtration through sterile filtration membranes. [00418] The active ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano- particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington’s Pharmaceutical Sciences, 16th edition, Osol, A. Ed. (1980). [00419] Sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semi-permeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2- hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and γ ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT® (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly- D-(-)-3-hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid- glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods. When encapsulated immunoglobulins remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37ºC, resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S-S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions. Page 123 of 203 1097657169\1\AMERICAS [00420] An antibody may be formulated in any suitable form for delivery to a target cell/tissue. For example, antibodies may be formulated as immunoliposomes. A “liposome” is a small vesicle composed of various types of lipids, phospholipids and/or surfactant which is useful for delivery of a drug to a mammal. The components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes. Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA 82:3688 (1985); Hwang et al., Proc. Natl Acad. Sci. USA 77:4030 (1980); U.S. Pat. Nos. 4,485,045 and 4,544,545; and WO97/38731 published October 23, 1997. Liposomes with enhanced circulation time are disclosed in U.S. Patent No.5,013,556. [00421] Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG- derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter. Fab' fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et al., J. Biol. Chem. 257: 286-8 (1982) via a disulfide interchange reaction. A chemotherapeutic agent is optionally contained within the liposome (See Gabizon et al., J. National Cancer Inst. 81(19): 1484 (1989)). [00422] Additional pharmaceutical formulations appropriate for the compositions for administration in the methods of the invention are known in the art (see, e.g., Remington's Pharmaceutical Sciences (1990) 18th ed., Mack Publishing Co., Easton, Pa.; The Merck Index (1996) 12th ed., Merck Publishing Group, Whitehouse, N.J.; and Pharmaceutical Principles of Solid Dosage Forms, Technonic Publishing Co., Inc., Lancaster, Pa., (1993)). The pharmaceutical formulations can be packaged in dosage unit form for ease of administration and uniformity of dosage. "Dosage unit form" as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the pharmaceutical carrier or excipient. [00423] Pharmaceutical compositions suitable for use in context of some embodiments of the invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. More specifically, a therapeutically effective amount means an Page 124 of 203 1097657169\1\AMERICAS amount of active ingredients (anti-B7H6 antibody, antigen-binding fragment thereof, CAR- modified immune cells) effective to prevent, alleviate or ameliorate symptoms of a disorder (e.g., cancer) or prolong the survival of the subject being treated. [00424] Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. [00425] For any preparation used in the methods of the invention, the therapeutically effective amount or dose can be estimated initially from in vitro and cell culture assays. For example, a dose can be formulated in animal models to achieve a desired concentration or titer. Such information can be used to more accurately determine useful doses in humans. Toxicity and therapeutic efficacy of the active ingredients described herein can be determined by standard pharmaceutical procedures in vitro, in cell cultures or experimental animals. The data obtained from these in vitro and cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage may vary depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl, et al., 1975, in "The Pharmacological Basis of Therapeutics", Ch.1 p. l). [00426] Depending on the severity and responsiveness of the condition to be treated, dosing can be of a single or a plurality of administrations, with course of treatment lasting from several days to several weeks or until cure is effected or diminution of the disease state is achieved. [00427] The amount of a composition to be administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc. [00428] J. Methods of Treatment [00429] Pharmaceutical compositions comprising anti-B7H6 antibodies, antigen-binding fragments thereof, or CAR-modified immune cells comprising the same, as described herein, may be administered for prophylactic and/or therapeutic treatments. [00430] An embodiment of the present invention provides a method of treating a condition associated with abnormal B7H6 expression, for example a B7H6-expressing cancer, in a subject. Page 125 of 203 1097657169\1\AMERICAS The method may include targeting a B7H6-expressing cell, for example a B7H6-expressing tumor cell, with a therapeutically effective amount of the antibody or antigen-binding fragment. [00431] In certain embodiments, the method comprises administering to the subject a composition comprising a therapeutically effective amount of anti-B7H6 antibodies, antigen- binding fragments thereof, or CAR-modified immune cells comprising the same. Preferably, the CAR-modified immune cells are CAR-modified γδ T cells. In one embodiment, the anti-B7H6 antibody is used in the form of an ADC. For example, the anti-B7H6 antibody may be coupled to a biologically active agent or a combination of such agents including but not limited to a radioisotope, a toxin, and the like. [00432] In one embodiment, provided is a method of treating a hematological or solid tumor with abnormal B7H6 expression. Such a method may comprise the step of targeting B7H6- expressing cells with a therapeutically effective amount of an anti-B7H6 antibody, antigen-binding fragment thereof, or CAR-modified immune cell comprising the same. Preferably, the CAR- modified immune cells are CAR-modified γδ T cells. Accordingly, embodiments include administering to a subject having a tumor associated with abnormal B7H6 expression, an anti- B7H6 antibody, antigen-binding fragment thereof, or CAR-modified immune cell comprising the same. In embodiments, the anti-B7H6 antibody is used in the form of an ADC. [00433] In one embodiment, provided is a method of inhibiting cell growth or proliferation, the method comprising exposing a B7H6-expressing cell to an anti-B7H6 antibody or antigen-binding fragment, or a CAR-modified immune cell comprising the same, thereof under conditions permissive for binding of the antibody or antigen-binding fragment to B7H6. Preferably, the CAR- modified immune cells are CAR-modified γδ T cells. “Inhibiting cell growth or proliferation” refers to decreasing a cell’s growth or proliferation by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100%, and can include inducing cell death. In embodiments, the B7H6- expressing cell is a tumor cell. [00434] In one embodiment, an anti-B7H6 antibody or antigen-binding fragment, or a CAR- modified immune cell comprising the same, is used to treat or prevent a cell proliferative disorder. Preferably, the CAR-modified immune cells are CAR-modified γδ T cells. In certain embodiments, the cell proliferative disorder is associated with abnormal expression and/or activity of B7H6. For example, in certain embodiments, the cell proliferative disorder is associated with Page 126 of 203 1097657169\1\AMERICAS increased expression of B7H6 on the surface of a cell. In certain embodiments, the cell proliferative disorder is a tumor or a cancer. [00435] Pharmaceutical compositions comprising engineered host cells that express a CAR, and/or admixtures thereof, may be administered for prophylactic and/or therapeutic treatments. In preferred embodiments, a pharmaceutical composition comprises γδ T cells engineered to express a B7H6-targeting CAR. An admixture may comprise non-engineered cells of a same or different type. Additionally or alternatively, an admixture may comprise engineered cells of a same type that express a different B7H6-targeting CAR. Additionally or alternatively, an admixture may comprise engineered cells of a different type that express a same or different B7H6-targeting CAR. For example and without limitation, an admixture may comprise a population of γδ T cells engineered to express a B7H6-targeting CAR, and a population of non-engineered γδ T cells. As another example and without limitation, an admixture may comprise a population of γδ T cells engineered to express a B7H6-targeting CAR, and a population of engineered or non-engineered cells e.g., NK cells, NKT cells, γδ cells, αβ cells, etc. In therapeutic applications, the compositions can be administered to a subject already suffering from a disease or condition in an amount sufficient to decrease at least one sign or symptom associated with the disease or condition. In embodiments, the amount is sufficient to cure the disease or condition. [00436] An engineered host cell population and/or admixtures thereof can also be administered to lessen a likelihood of developing, contracting, or worsening a condition. Effective amount of a population of engineered host cells, non-engineered host cell, and/or admixtures thereof, for therapeutic use can vary based on the severity and course of the disease or condition, previous therapy, the subject’s health status, weight, and/or response to various drugs, and/or the judgement of a treating physician. [00437] In embodiments, the one or multiple engineered host cell populations, non-engineered cells and/or admixtures thereof, of the present disclosure can be used to treat a subject in need of treatment for a condition. Examples of such conditions include but are not limited to cancer. Subjects can be humans, non-human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the Page 127 of 203 1097657169\1\AMERICAS like. A subject can be of any age. Subjects can be, for example, elderly adults, adults, adolescents, pre-adolescents, children, toddlers, infants. [00438] A method of treating a condition (e.g., ailment) in a subject may comprise administering to the subject a therapeutically-effective amount of one or more engineered host cell populations, preferably engineered γδ T cells, non-engineered cells and/or admixtures thereof. The one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof can be administered at various regimens (e.g., timing, concentration, dosage, spacing between treatment, and/or formulation). A subject can also be preconditioned with, for example, chemotherapy, radiation, or a combination of both, prior to receiving the therapeutically-effective amount of one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof. As part of a treatment, one or multiple engineered host cell populations, non- engineered cells, and/or admixtures thereof may be administered to a subject at a first regimen and the subject may be monitored to determine whether the treatment at the first regimen meets a given level of therapeutic efficacy. In some cases, the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof may be administered to the subject at a second regimen, based on information gleaned from providing the subject with the first regimen. [00439] In embodiments, a pharmaceutical composition comprising at least one host cell engineered to express a CAR of the present disclosure, preferably a γδ T cell, may be administered in a first regimen. The subject may be monitored, for example by a healthcare provider (e.g., treating physician or nurse). In some examples, the subject is monitored to determine or gauge an efficacy of the engineered host cell in treating the condition of the subject. In some situations, the subject may also be monitored to determine the in vivo expansion of an engineered host cell population in the subject. Another pharmaceutical composition comprising at least one host cell engineered to express a CAR of the present disclosure may be administered to the subject in a second regimen. The pharmaceutical composition administered in the second regimen may comprise a same type of host cell expressing a same CAR as that administered to the subject in the first regimen. However, it is within the scope of this disclosure that the pharmaceutical composition administered in the second regimen may comprise a different type of host cell, optionally expressing a different CAR. In some examples, the second regimen is not performed, for example if the first regimen is found to be effective (e.g., a single round of administration may Page 128 of 203 1097657169\1\AMERICAS be sufficient to treat the condition). In embodiments, a population of engineered host cells can be administered to various subjects (e.g., where the host cell has universal donor characteristics). [00440] One or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof, having cytotoxic activity against a B7H6-expressing cell such as a tumor cell, can be administered to a subject in any order or simultaneously. If simultaneously, the engineered host cell(s), and/or admixtures thereof, of the disclosure can be provided in a single, unified form, such as an intravenous injection, or in multiple forms, for example, as multiple intravenous infusions, s.c, injections or pills. The one or multiple engineered host cell populations, non- engineered cells, and/or admixtures thereof of the disclosure can be packed together or separately, in a single package or in a plurality of packages. One or all of the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof of the disclosure can be given in multiple doses. If not simultaneous, the timing between the multiple doses may vary to as much as about a week, a month, two months, three months, four months, five months, six months, or about a year. In some cases, an engineered host cell, preferably an engineered γδ T cell, of the disclosure can proliferate within a subject's body, in vivo, after administration to a subject. The one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof of the present disclosure can be frozen to provide cells for multiple treatments with the same cell preparation. The one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof, and pharmaceutical compositions comprising the same, can be packaged as a kit. A kit may include instructions (e.g., written instructions) on the use of the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof, and compositions comprising the same. [00441] The one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof, described herein can be administered before, during, or after the occurrence of a disease or condition, and the timing of administering a pharmaceutical composition containing the engineered host cell population can vary. For example, the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof can be used as a prophylactic and can be administered continuously to subjects with a propensity to conditions or diseases in order to lessen a likelihood of the occurrence of the disease or condition. The initial administration can be via any route practical, such as by any route described herein using any formulation described herein. In some examples, the administration of the one or multiple engineered host cell Page 129 of 203 1097657169\1\AMERICAS populations, non-engineered cells, and/or admixtures thereof of the disclosure is an intravenous administration. One or multiple dosages of the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof can be administered as soon as is practicable after the onset of a particular condition (e.g., cancer) and for a length of time necessary for the treatment of the disease/condition, such as, for example, from about 24 hours to about 48 hours, from about 48 hours to about 1 week, from about 1 week to about 2 weeks, from about 2 weeks to about 1 month, from about 1 month to about 3 months. In embodiments, one or multiple dosages of the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof can be administered years after onset of the disease/condition (e.g., cancer) and before or after other treatments. [00442] In embodiments, the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof, of the disclosure, is administered simultaneously or sequentially with one or more methods to elevate common gamma chain cytokine(s). As used herein, "one or more methods to elevate common gamma chain cytokine(s): refers to a method, or combination of methods, that alters the physiological state of a subject, such that at least one common gamma chain cytokine level is elevated in the subject. In embodiments, the method elevates the level of one or more common gamma chain cytokine(s) selected from the group consisting of IL-2, IL-4, IL-7, IL-15, and IL-21 in the subject. In embodiments, the method comprises lymphodepletion. In embodiments, the method comprises administering one or more common gamma chain cytokine(s) to the subject. In some cases, IL-2, IL-4, IL-7, IL-15, and/or IL-21 are administered. In embodiments, the method comprises secreting common gamma chain cytokine(s) from an administered engineered host cell. In some cases, IL-2, IL-4, IL-7, IL-15, and/or IL-21 are secreted. [00443] In embodiments, the administering one or more methods to elevate common gamma chain cytokine(s) comprises lymphodepletion before introducing the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof, of the disclosure. In embodiments, the administering one or more methods to elevate common gamma chain cytokine(s) comprises administering simultaneously with introducing the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof, or sequentially an amount of common gamma chain cytokine(s) effective to increase proliferation, cytotoxic activity, persistence, or the combination thereof of the introduced one or multiple engineered host Page 130 of 203 1097657169\1\AMERICAS cell populations, non-engineered cells, and/or admixtures thereof. The amount of administered common gamma chain cytokine(s) can be an amount effective to increase proliferation, cytotoxic activity, persistence, or the combination thereof of the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof. Exemplary amounts of IL-15 include, without limitation between 0.01 — 10 µg/kg/dose every 24 hours for IL-15. Exemplary amounts of IL-2 include, without limitation, between about 3x106 and about 22x106 units every 8 - 48 hours. For example, the dosing regimen for IL2 in RCC is 600,000 International Units/kg (0.037 mg/kg) IV 48hr infused over 15 minutes for a maximum 14 doses. [00444] In embodiments, the administering one or more methods to elevate common gamma chain cytokine(s) comprises lymphodepletion before administering the one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof and administering simultaneously with introducing the one or multiple engineered host cell populations, non- engineered cells, and/or admixtures thereof, or sequentially, an amount of common gamma chain cytokine(s) effective to increase proliferation, cytotoxic activity, persistence, or the combination thereof of the introduced one or multiple engineered host cell populations, non-engineered cells, and/or admixtures thereof. [00445] In embodiments, elevating common gamma chain cytokine(s) is accomplished, at least in part, via the engineered host cell(s), where the common gamma chain cytokine(s) are expressed from CAR construct as disclosed herein. In such an example, it is within the scope of this disclosure that one or more additional gamma chain cytokine(s) are additionally administered in a manner to elevate the additional gamma chain cytokine(s). [00446] The γδ T cells of the subject invention can also be advantageously administered to a patient in conjunction with (e.g., before, simultaneously or following) any number of relevant treatment modalities (e.g., for treating cancer) including, e.g., chemotherapy, radiation therapy, or immunotherapy. A patient can also be preconditioned with a therapeutically effective amount of γδ T cells prior to receiving the chemotherapy, radiation therapy, or immunotherapy (e.g., cell therapy). Suitable immunotherapies for use in combination with γδ T cells include autologous and allogeneic cell therapies, engineered T and NK cells, immune engagers, fusion proteins, or other immune-oncology agents. Page 131 of 203 1097657169\1\AMERICAS [00447] In embodiments, the subject γδ T cells can be administered in conjunction with an appropriate cellular immunotherapy for treating a disease such as cancer (e.g. CAR T or CAR NK cells, or Treg therapy). For example, a method of treatment (e.g., for treating cancer) according to the present invention can include a conditioning step, e.g. a preconditioning step, of administering a therapeutically effective amount of γδ T cells to a subject simultaneously or sequentially with administration of a cellular immunotherapy directed against the disease (e.g., cancer). In embodiments, the cellular immunotherapy can comprise further administering an engineered T cell or NK cell therapy comprising a CAR binding to any tumor associated antigen of interest. In embodiments, the subject γδ T cells may comprise a dual CAR binding to CD70 and B7H6. [00448] In embodiments, the γδ T cells of the subject invention may be advantageously administered in conjunction with adoptive cell therapies (ACT) (for reviews of HSCT and adoptive cell therapy approaches, see, Rager & Porter, Ther Adv Hematol (2011) 2(6) 409-428; Roddie & Peggs, Expert Opin. Biol. Ther. (2011) 11(4):473-487; Wang et al. Int. J. Cancer: (2015)136, 1751-1768; and Chang, Y. J. and X. J. Huang, Blood Rev, 2013.27(1): 55-62), each of which is incorporated by reference herein in its entirety. Such adoptive cell therapies include, but are not limited to, allogeneic and autologous hematopoietic stem cell transplantation, donor leukocyte (or lymphocyte) infusion (DLI), adoptive transfer of tumor infiltrating lymphocytes, or adoptive transfer of T cells or NK cells (including recombinant cells, e.g., CAR T, CAR NK). Beyond the necessity for donor-derived cells to reconstitute hematopoiesis after radiation and chemotherapy, immunologic reconstitution from transferred cells is important for the elimination of residual tumor cells. The efficacy of ACT as a curative option for malignancies is influenced by a number of factors including the origin, composition and phenotype (lymphocyte subset, activation status) of the donor cells, the underlying disease, the pre-transplant conditioning regimen and post-transplant immune support (e.g., IL-2 therapy) and the graft-versus-tumor (GVT) effect mediated by donor cells within the graft. Additionally, these factors may be balanced against transplant-related mortality, typically arising from the conditioning regimen and/or excessive immune activity of donor cells within the host (i.e., graft-versus-host disease, cytokine release syndrome, etc.). [00449] K. Articles of Manufacture and Kits Page 132 of 203 1097657169\1\AMERICAS [00450] Another embodiment of the invention is an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of disease or condition associated with B7H6- expression. The disease associated with B7H6 expression is selected from a proliferative disease such as a cancer or malignancy or a precancerous condition, or is a non-cancer related indication associated with expression of B7H6. Examples of cancerous conditions relevant to the present disclosure include but are not limited to colon cancer, pancreatic cancer, Non-Hodgkin Lymphoma, cervical cancer, hepatic cancer, gastro-intestinal cancer, lung cancer, ovarian cancer, glioblastoma, prostate cancer, breast cancer, ovarian cancer, kidney cancer, bladder cancer, melanoma, and glioma. [00451] The article of manufacture comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, etc. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is effective for treating, preventing and/or diagnosing the condition associated with B7H6 expression and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is an anti-B7H6 antibody or antigen- binding fragment thereof of the invention, or a CAR-modified immune cell of the invention (preferably a CAR-modified γδ T cell, or a nucleic acid of the invention. Optionally, a composition further comprises a carrier, for example a pharmaceutically acceptable carrier. The label or package insert indicates that the composition is used for treating the condition associated with B7H6 expression, for example cancer. The label or package insert will further comprise instructions for administering the antibody composition to the patient. Additionally, the article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer’s solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes. [00452] Kits are also provided that are useful for various purposes, e.g., for B7H6- expressing cell killing assays, for purification or immunoprecipitation of B7H6 polypeptide from cells. For isolation and purification of B7H6 polypeptide, the kit can contain an anti-B7H6 antibody coupled to beads (e.g., sepharose beads). Kits can be provided which contain the antibodies for detection and quantitation of B7H6 polypeptide in vitro, e.g., in an ELISA or a Page 133 of 203 1097657169\1\AMERICAS Western blot. As with the article of manufacture, the kit comprises a container and a label or package insert on or associated with the container. The container holds a composition comprising at least one anti-B7H6 antibody or antigen-binding fragment thereof, of the invention. Additional containers may be included that contain, e.g., diluents and buffers, control antibodies. The label or package insert may provide a description of the composition as well as instructions for the intended in vitro or detection use. [00453] For example, a kit can comprise a first container comprising a composition comprising one or more B7H6 antibodies or CAR modified immune cells of the invention; and a second container comprising a buffer. The buffer may be pharmaceutically acceptable. EXAMPLES [00454] The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the methods and compositions of the invention, and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is average molecular weight, temperature is in degrees Centigrade, and pressure is at or near atmospheric. [00455] Example 1. Binding profiles of anti-B7H6 antibodies [00456] This Example demonstrates functionality of anti-B7H6 antibodies of the present disclosure. [00457] As shown at FIG.1, synthetic phage display libraries containing fully human antibody sequences were used to pan against recombinant human B7H6 (rhuB7H6) or B7H6+ cell lines. Anti-B7H6 scFv clones derived from phage display panning were reformatted into human IgG antibodies (Abs) and tested using flow cytometry against HeLa cells with B7H6 knock-out (KO) and HeLa cells with B7H6 KO HeLa cells re-engineered to express B7H6 at higher levels (HeLa B7H6high). The Ab clones only bound to HeLa B7H6high cells compared to B7H6 KO HeLa cells. [00458] Anti-B7H6 IgG1 Ab clones were evaluated against rhuB7H6 conjugated to MagPlex- C Microspheres. The Luminex FLEXMAP 3D® instrument (Austin, Texas) was used to detect the mean fluorescence intensity (MFI) at various Ab concentrations. EC50 values from each Ab clone Page 134 of 203 1097657169\1\AMERICAS were determined from the titrated Ab curves (FIG.2). The Ab clones showed a diverse repertoire of binding levels toward rhuB7-H6. [00459] Turning to FIG.3A, depicted is an illustration of membrane domains of B7H6. Shown is full length B7H6 comprising of V-distal domain and C-proximal domain, V-distal domain with B7H6 stump region, and C-proximal domain with B7H6 stump region. Binding of phage-display derived antibody IgG clones to the different membrane domains of B7H6 measured by flow cytometry are indicative of all antibody IgG clones binding to full-length B7H6 and majority binding to the distal region (FIG.3B). Linear epitope mapping of phage-display derived antibody IgG clones performed using a chemifluorescent-based assay suggests that majority of the antibody clones bind to the membrane distal region (FIG.3C). Each row indicates the IgG clone, and each column indicates an epitope’s sequence. The color gradient for each cell of the heatmap plot represents the relative fluorescence unit, indicative of relative binding to the epitope sequence. [00460] Target cell lines HeLa B7H6 KO (Synthego), HeLa, LoVo, Raji, HCT-15, and NCIH716 (ATCC) were stained with anti-B7H6 antibody (Biolegend) or isotype (R&D Systems) and were analyzed by flow cytometry. B7H6 expression in different cell lines was graphed as MFI (mean fluorescence intensity) ratio. The resulting ratio of antibody MFI to isotype MFI was graphed (FIG.4). Based on this data the cell lines could be classified as B7-H6 low expressing: HeLa; medium expressing: Lovo, Raji, HCT-15; and High expressing: NCIH716. [00461] Example 2. General methodology for identification of lead anti-B7H6 CARs [00462] Briefly, phage display technology was used to screen for binders as mentioned above. The screens incorporate recombinant proteins, cancer cell lines with variable target densities, recombinant lines expressing full-length and unique domains of interest (e.g., domains comprising post-translational modifications (PTMs) known to be relevant to a particular cancer type. To minimize “on-target, off-tumor” binding, target null cell lines, and cell lines expressing related family members were used for counter screens. [00463] Phage panning using biotinylated recombinant human B7H6 protein (Acro Biosciences) bound to streptavidin beads was performed on highly diverse synthetic scFv phage display libraries (Twist Bio) using 5 rounds of selection, each with decreasing antigen concentration (starting at 100 pmol to 2 pmol) but increasing wash stringency. An alternate panning strategy with alternating rounds of selection with 1e8 B7H6+ cells or antigen (100 pmol Page 135 of 203 1097657169\1\AMERICAS and 25 pmol, respectively) was also used. Antigen-bound phage was pulled down, eluted, amplified and screened using ELISAs for binder confirmation and selection. Following NGS and Sanger clone sequencing, scFvs were reformatted into IgGs, expressed, purified and characterized. [00464] Lead binders were reformatted into human immunoglobulins of appropriate isotype (e.g., IgG) or certain CAR surrogate formats (e.g., CAR diabodies) for further characterization and to assess a wide variety of potential liabilities. This is referred to as a “Molecular Assessment Approach,” which aims to optimize biophysical and chemical properties of molecules in early stages prior to CAR development. Criteria evaluated included propensity to aggregate, solubility level, propensity to fragment, deamidation, oxidation, isomerization, cyclization or other chemical modification of key residues, disulfide bond shuffling, and glycation, among others. Furthermore, some liabilities were assessed via the design of screening libraries selected for phage display. In some cases, use of protein microarray screens was adopted to assess cross-reactivity of the fully formatted IgGs. By use of curated phage display libraries and assessment of the formatted antibody, a wide variety of potential liabilities associated with particular binders were assessed early in the process, before CAR manufacture. [00465] Anti-target antibodies were further screened for/characterized by their physical/chemical properties and/or biological activities by various assays. This matrix-based, multiparametric approach was used to generate and rank order the final leads against the B7H6 target. For example, for biochemical/biophysical characterization, antibodies in a cohort were tested for their antigen binding activity by known methods including but not limited to biolayer inferometry (BLI) and surface plasmon resonance (SPR) on appropriate instruments, using a soluble preparation of recombinant full-length or tagged human proteins. In this way, kinetic constants (Kon & Koff) and the dissociation constant (KD) of reformatted antibodies and, where appropriate, benchmark antibody (47.39 as herein disclosed), were generated. Biochemical assays relied upon include but are not limited to ELISA, Western Blotting and/or flow cytometry assays, used to generate relative affinity data (e.g., EC50s) and maximum specific binding (Bmax) for the cohort and benchmark antibodies. [00466] Benchmark CAR PL741 in which a B7H6 binding domain is derived from the 47.39 antibody (see WO 2013/169691) as herein described was included as a comparative CAR control. Briefly, a nucleic acid encodes SEQ ID NO: 337, a PL741 CAR comprising the following domains Page 136 of 203 1097657169\1\AMERICAS in order: a signal peptide, a B7H6 binding domain, a CD8 hinge and transmembrane domain, a 4- 1BB costimulatory domain, and a CD3ζ signaling domain. A nucleic acid encoding the PL741 CAR comprises the sequence of SEQ ID NO: 338. Table 13 below provides annotation of the nucleotide sequence of SEQ ID NO: 338. Table 13. PL741 Nucleotide Sequence Annotation Annotation Numbering
Figure imgf000138_0001
[00467] In some cases, specific assays for mapping epitopes bound by the disclosed antibodies and benchmark antibodies were also used, for example, via the use of overlapping peptides spanning the target, in an ELISA format. Where appropriate, recombinant cell lines were made expressing full-length target or domains known to have specific epitopes, for screening and binning. To further characterize lead epitopes, hydrogen/deuterium exchange mass spectrometry (HDX-MS) was used. [00468] Lead binders were reformatted into CARs in both heavy-light (HL) and LH (light- heavy) orientations, and these CARs were further screened in Jurkat Lucia NFAT cells to assess CAR cell surface expression, tonic signaling, and activation in the context of target-based stimulation, in the form of recombinant protein or cancer cell lines expressing the B7H6 target. CARs that showed robust activation in Jurkat cells were transduced into Vδ1 γδ T cells, and potency against target-expressing cell lines was assessed using a stringent 120 hr in vitro cytotoxicity assay. [00469] Example 3. Functional assessment of anti-B7H6 CARs [00470] Anti-B7H6 scFvs were formatted into CARs, inserted into a gammaretroviral vector, and transduced into Jurkat-LuciaTM NFAT cells (Invivogen). CAR transgene activity in Jurkat- Page 137 of 203 1097657169\1\AMERICAS Lucia cells was evaluated in a co-culture assay with HeLa or HeLa B7-H6 KO cells. Supernatants were collected after 18-hrs, was mixed with QUANTI-LucTM reagent (InvivoGen), and Lucia luciferase activity (NFAT activity) was detected using the Cytation™ 5 imaging plate reader. Increased NFAT activity was associated with B7H6 scFv CARs against B7H6+ HeLa cells, compared to HLA B7H6 KO cells (FIGS.5A-5B). [00471] For a long-term cytotoxicity assay, B7H6 CAR Vδ1 γδ T cells were cocultured with Nuc-NIR expressing target cell lines at an effector to target ratio (E:T) of 1:1 for 120 hours. Viable Nuc-NIR expressing target cells were quantified every 4 hours by the Incucyte® SX5 (Sartorius). Cytotoxicity index was calculated by dividing the total NIR object area of each time point by the value of time 0 hours. Target cells were modified to express Nuc-NIR by transducing them with Incucyte® Nuclight NIR lentivirus (Sartorius). B7H6 CAR Vδ1 γδ T cells showed strong cytotoxic activity against different cancer cell lines such as Raji (Lymphoma) (FIG. 6A), HeLa (cervical) (FIG.6B), and HCT-15 (colon) (FIG.6C). The lower cytotoxicity index indicates better cytotoxic potential of B7H6 CAR Vδ1 γδ T cells versus target cells alone (black diamonds). PL741 (Benchmark, see Example 2 above)) is a B7H6 CAR previously validated and the lead B7H6 CAR Vδ1 γδ T cells outperform it. [00472] B7H6 CAR Vδ1 γδ T cells were evaluated for polyfunctionality by the Isolight single cell secretome assay (Isoplexis). Cells were stimulated with rhuB7-H6 (AcroBio) for 18 hrs prior to loading CAR T cells onto the IsoCode chip for single-cell multiplex cytokine analysis. Polyfunctional strength index (PSI) (FIG.7A), is defined by the percentage of polyfunctional cells with the intensity of the cytokines measured. Percent polyfunctionality (FIG.7B) is the percent of cells secreting 2 or more cytokines. Lead B7H6 CAR Vδ1 γδ T cells show a higher degree of polyfunctionality as compared to PL741 (Benchmark) B7H6 CAR Vδ1 γδ T cells. [00473] Example 4. B7H6 CAR γδ T cell Phenotype [00474] B7H6 CAR Vδ1 γδ T cells were stained for multiple phenotypic markers and were analyzed by flow cytometry. Expression profiles showed a predominant naïve-like T cell memory phenotype of the candidate B7H6 CAR+ Vδ1 γδ T cells (FIG.8A). Percentages of CAR+ Vδ1 γδ T cells in B7H6 CAR+ Vδ1 γδ T cell products that express multiple chemokine receptors, activation markers, and cells that co-express of PD-1 and another co-inhibitory receptor are shown in the heatmap (FIG.8B). Page 138 of 203 1097657169\1\AMERICAS [00475] Example 5. Proliferative Potential of B7H6 CAR γδ T cell [00476] Proliferative potential of B7-H6 CAR Vδ1 γδ T cells labeled with Cell Trace Violet (CTV) (Molecular Probes) following target antigen exposure with B7-H6+ cell lines in a 7-day culture period was assessed. Flow cytometry was used to determine the fold change of CTV dye dilution (CTV Geometric mean at day 0 / CTV Geometric mean at day 7). B7-H6 CAR Vδ1 γδ T cells demonstrated increased proliferation when stimulated with various B7-H6+ tumor cell lines (FIG.9). [00477] Example 6. B7H6 CAR γδ T cell Cytotoxic Potential [00478] Cytotoxic potentials of lead B7-H6 CAR Vδ1 γδ T cells and Benchmark B7-H6 CAR Vδ1 γδ T cells were evaluated against B7H6+ tumor cell line, HCT-15, in a 48-hour Incucyte Immune Cell Killing Assay in the presence and absence of exogenously added soluble B7-H6 (sB7-H6) (AcroBio). Low (FIG.10A) and high (FIG.10B) levels of spiked sB7H6 were used to simulate shed B7H6 levels in serum and tumor microenvironment, respectively. T cells were co- cultured with HCT-15 NucNIR-expressing target cells at a suboptimal E:T ratio of 1:1. The Cytotoxicity Index was calculated by dividing the total NIR object area (mm2/well) of all time points by the value at time = 0. No significant differences in cytotoxic activity were observed from the candidate B7H6 CAR Vδ1 γδ T cells between +/- sB7H6 conditions. This Example demonstrates that sB7H6 interfered with the activity of Benchmark B7-H6 CAR Vδ1 γδ T but did not impair the activity of the lead B7-H6 CAR Vδ1 γδ T cells. [00479] Example 7. In Vivo Efficacy of B7H6 CAR γδ T cells [00480] The in vivo efficacy of B7H6 CAR Vδ1 γδ T cells was assessed in a subcutaneous colon cancer model using HCT-15 cells. Female NSG mice (The Jackson Laboratory) were implanted subcutaneously with 2 x 106 HCT-15 cells (ATCC), mixed with Matrigel (1:1 ratio) (Corning). Animals were randomized into groups of 5 when tumor volume reached an average of 100 mm3 and were injected IV (tail vein) with 5 x 106 or 15 x 106 B7H6 CAR Vδ1 γδ cells. Negative control group was tumor alone group (no treatment). Human IL-2 (Proleukin) was administered intraperitoneally prior to treatment and then thrice a week. Tumor volume was measured twice a week with calipers. The study was terminated when mean tumor volumes reached ≥2000mm3. Tumor response was durable at both doses for PL755 and at the 15 x106 dose for PL754 B7-H6 CAR Vδ1 γδ T cells as compared to control groups (FIGS. 11A-11B). These Page 139 of 203 1097657169\1\AMERICAS data support the cytotoxic potential of B7-H6 CAR Vδ1 γδ T cells in a human colon cancer cell xenograft model. [00481] Example 8. Analysis of B7H6 CAR γδ T cells with dnTGFβRII Bolt-on [00482] CAR expression is not impacted by the expression of the dnTGFβRII Bolt-on in B7H6 CAR Vδ1 γδ T cells. B7-H6 CAR Vδ1 γδ T cells with or without the dnTGFβRII Bolt-on were stained for TGFβRII (R&D Systems) and was only detected on the cell surface of B7H6 CAR Vδ1 γδ T cells with the Bolt-on using flow cytometry (FIG.12). [00483] B7H6 CAR Vδ1 γδ T cells with the dnTGFβRII Bolt-on showed inhibition of pSMAD2/3 levels in the presence of 20ng/mL of TGF-β1 (R&D Systems) using intracellular flow cytometry, confirming the functionality of the dnTGFβRII by inhibiting TGF-β1 signaling. B7H6 CAR Vδ1 γδ T cells without the dnTGFβRII Bolt-on and untransduced Vδ1 γδ T cells showed an increase in pSMAD2/3 after exposure to TGF-β1 compared to no TGF-β1 treatment (FIG.13). [00484] Cytotoxic potentials of B7H6 CAR Vδ1 γδ T cells (red circles) and B7H6 CAR Vδ1 γδ T cells with dnTGFβRII Bolt-on (blue circles) were evaluated against B7H6+ tumor cell lines HCT-15 expressing Nuc-NIR in the 120-hour Incucyte Immune Cell Killing Assay at effector to target ratio 1:1 in the presence or absence of TGF-β1 (R&D Systems). The Cytotoxicity Index was calculated by dividing the total NIR object area (mm2/well) of all time points by the value at time = 0. B7H6 CAR Vδ1 γδ T cells with dnTGFβRII Bolt-on are associated with a cytotoxicity advantage over B7-H6 CAR Vδ1 γδ T cells without Bolt-on in the presence of exogenous TGFβ- 1 (FIG.14). [00485] Example 9. Gene editing of γδ T cells [00486] To knockout the CISH gene in Vδ1 T cells, a ribonucleoprotein (RNP) complex comprised of CRISPR/Cas9 (Integrated DNA Technologies) and CISH sgRNA’s (Synthego Corporation) were combined with activated Vδ1 T cells from PBMC’s (P3 Primary Cell 4D- Nucleofector X Kit L, Lonza). The RNP complex was delivered into Vδ1 T cells by electroporation (Lonza 4D-Nucleofector). Post electroporation, the cells were transferred to T flasks (Corning) containing X-Vivo15 (Lonza) for cell recovery and expansion in a 37°C CO2 incubator (ThermoFisher). To evaluate CISH KO efficiency, cell pellets were collected following αβ T cell depletion (StemCell Technologies) on day 7 post gene edit and genomic DNA was extracted using Page 140 of 203 1097657169\1\AMERICAS the NucleoSpin Tissue kit (MACHEREY-NAGEL). The edited region of the CISH gene was amplified by PCR and Sanger sequenced (Sequetech). Gene knockout efficiency was determined using the ICE Analysis tool (Synthego). As shown in FIG. 15, high efficiency knockout of the CISH gene was observed from Vδ1 T cells. [00487] Post CISH gene knockout, Vδ1 T cell enrichment in PBMCs was monitored over time by flow cytometry (anti-Vδ1, BioLegend). As shown in FIG.16A, CISH gene knockout did not affect Vδ1 T cell enrichment as compared to unedited Mock control cells. Similar purity of CISH gene edited Vδ1 T cells was achieved post TCRαβ depletion (StemCell Technologies) as compared to Mock control cells. Cell viability of Vδ1 T cell enriched PBMCs was monitored post CISH gene knockout (Countess II, ThermoFisher). As shown in FIG.16B, CISH KO cells maintained high cell viability similarly to unedited Mock control cells. [00488] Example 10: CISH KO enhances the function of Vδ1 T cells [00489] Healthy donor peripheral blood mononuclear cells (PBMCs) were used to activate, expand, CRISPR-gene edit, and engineer Vδ1 γδ T cells to express a CAR. To knockout the CISH gene in Vδ1 T cells, a ribonucleoprotein (RNP) complex comprising CRISPR-Cas9 (Integrated DNA Technologies) or -MAD7 (Aldevron) and CISH sgRNA were combined with activated Vδ1 T cells from PBMCs (P3 Primary Cell 4D-Nucleofector X Kit L, Lonza). The RNP complex was delivered into Vδ1 T cells by electroporation (Lonza 4D-Nucleofector). Post electroporation, the cells were transferred to T flasks (Corning) containing X-Vivo15 (Lonza) for cell recovery prior to transduction with gammaretroviral vector encoding the CAR and then were subsequently expanded. To evaluate CISH KO efficiency, cell pellets were collected, and genomic DNA was extracted using the NucleoSpin Tissue kit (MACHEREY-NAGEL). The edited region of the CISH gene was amplified by PCR and Sanger sequenced (Sequetech). Gene knockout efficiency was determined using the ICE Analysis tool (Synthego). To evaluate the KO of CISH protein in CAR Vδ1 T cells, WT (unedited) and CISH KO CAR Vδ1 T cells were incubated with and without IL- 2 (Peprotech) for 5 hours. Cell lysates were generated for western blot detection of CISH protein using anti-CISH Rabbit mAb (Cell Signaling). An IRDye 800CW goat anti-Rabbit IgG secondary antibody (LI-COR) was used for detection of the primary antibody and the presence of the IRDye was imaged with the Odyssey imaging system (LI-COR). The results are shown in FIGS. 17A- 17B. (FIG. 17A) WT CAR Vδ1 T cells showed an increase in levels of CISH protein when Page 141 of 203 1097657169\1\AMERICAS exposed to IL-2. In contrast, CISH KO CAR Vδ1 T cells had substantially reduced levels of CISH protein in the presence of IL-2 supporting the KO of CISH. (FIG.17B) To demonstrate enhanced function of CISH KO CAR-transduced and untransduced Vδ1 T cells, the cytolytic activity against the B7-H6+ HCT-15 cell line (ATCC) was evaluated using an in vitro repetitive stimulation assay. HCT-15 NIR fluorescently labeled cell line and effector cells were added into a 96-well plate at an E:T ratio of 5:1 with or without different concentrations of IL-2. Each stimulation was monitored for 3 days after which fresh target cells were plated in another 96-well plate. The effector cells from each stimulation were then transferred into the plate with fresh target cells. The co-cultures were monitored every 4 hours using Incucyte® SX5 (Satorius). The Cytotoxicity Index was calculated based on the remaining viable target tumor cells by dividing the total NIR object area of each time point by the value at time 0 for each stimulation. The lower the cytotoxicity index, the better the cytotoxic potential. After the second stimulation, CISH KO untransduced Vδ1 T cells were associated with greater tumor cell killing compared to WT untransduced Vδ1 T cells only in the presence of IL-2. Without IL-2, CISH KO CAR Vδ1 T cells had superior tumor control compared to WT CAR Vδ1 T cells. The results suggest CISH KO enhances the in vitro cytotoxicity of Vδ1 T cells. [00490] CAS9 CISH sgRNA: TGTACAGCAGTGGCTGGTGG (AGG) (SEQ ID NO: 339) MAD7 CISH sgRNA: (TTTA) GGTGTACAGCAGTGGCTGGTG (SEQ ID NO: 340). [00491] Example 11: Enhancement of CAR Vδ1 T cells effector function using membrane bound IL-12 (mbIL-12) [00492] CAR vector, consisting of the anti-CD19 (clone FMC63) scFv (as described in Kochendefer JN et al., Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells, Blood.2010 Nov 11;116(19):3875-86), CD8α hinge and transmembrane domains, and 4-1BB and CD3ζ signaling domains followed by a self-cleaving P2A sequence that is used to separate the mbIL-12 molecule (as described in Lee et al., Antigen-dependent IL-12 signaling in CAR T cells promotes regional to systemic disease targeting, bioRxiv.2023 Jan 7;2023.01.06.522784), was constructed by cloning a DNA fragment containing all CAR domains (Integrated DNA Technologies) into a self-inactivating (SIN) Moloney Murine Leukemia Virus (MMLV) gamma-retroviral plasmid. The Page 142 of 203 1097657169\1\AMERICAS constructs design is shown in FIG. 18 (construct A) CAR (FMC63) construct, (construct B) CAR-mbIL-12 construct, and (construct C) mbIL-12 construct. [00493] Healthy donor peripheral blood mononuclear cells (PBMCs) were used to activate, expand and engineer Vδ1 γδ T cells to express the CD19 CAR-mbIL-12 transgene. The results are shown in FIGS. 19A-19D. (FIG. 19A) Vδ1 γδ T cells (post-transduction) were sampled at different time points over the course of the expansion to determine cell counts. The total Vδ1 γδ T cell counts did not differ between CAR and CAR-mbIL-12, suggesting mbIL-12 has no negative impact on the expansion of Vδ1 γδ T cells. Cells were harvested, stained for CAR and mbIL-12 using anti-FMC63 antibody conjugated to PE (Acrobio Systems, Newark, DE) and anti-IL12 conjugated to APC (Miltenyi Biotec) respectively, and acquired on the Novocyte flow cytometer (Agilent Technologies) to determine the percentage of CAR and mbIL-12 positive Vδ1 γδ T cells. (FIG.19B) No difference in % CAR was observed between Vδ1 γδ T cells expressing CAR and CAR-mbIL-12 at the end of the process to further support mbIL-12 did not affect CAR surface expression. (FIG. 19C) Over the course of the expansion, the % mbIL-12 expressed on the cell surface of Vδ1 γδ T cells decreased over time. (FIG.19D) CAR-mbIL-12 Vδ1 γδ T cells were co- cultured with and without CD19+ Raji cells for 18 hrs. After stimulation with target cells, mbIL- 12 increased on the cell surface of the Vδ1 γδ T cells compared to unstimulated conditions suggesting the surface expression of mbIL-12 is dependent on CAR activation. [00494] To demonstrate the functionality of CAR-mbIL-12, cytolytic activity against CD19- positive cell lines using CAR, CAR-mbIL-12, and mbIL-12 transduced Vδ1 γδ T cells was evaluated in an in vitro repetitive stimulation assay. Raji-Nuc NIR fluorescently labeled cell line and effector cells were added into a 96-well plate at E:T ratios of 5:1, 2.5:1, and 1.25:1. The first stimulation was monitored for 3 days after which fresh target cells were plated in another 96-well plate. The effector cells from the first stimulation were then transferred into the plate with fresh target cells. The second stimulation was monitored for an additional 3 days, and the same procedure was performed for a third stimulation. The co-cultures were monitored every 4 hours using Incucyte® SX5 (Satorius). The Cytotoxicity Index was calculated based on the remaining viable target tumor cells by dividing the total NIR object area of each time point by the value at time 0 for each stimulation. The lower the cytotoxicity index, the better the cytotoxic potential. After the second and third stimulation, the CAR-mbIL-12 Vδ1 γδ T cells were associated with greater tumor cell killing compared to CAR and mbIL12 Vδ1 γδ T cells across the E:T ratios Page 143 of 203 1097657169\1\AMERICAS tested. Co-culture conditions with a Cytotoxicity Index > 1 (indication of incomplete tumor control), were not analyzed in the subsequent stimulation. The results are shown in FIG.20 and demonstrate the functionality of the CAR-mbIL-12 expressed by Vδ1 γδ T cells to sustain cytolytic killing of tumor cells. [00495] CAR-mbIL-12 Vδ1 γδ T cells were evaluated in Raji cell subcutaneous xenograft tumor model at a suboptimal dose of 2.5e6 CAR+ viable cells. Raji cell subcutaneous xenografts were established by inoculation of 1E6 cells/mouse on the right hind flank of female NSG mice. Mice were randomized into study cohorts when tumor volumes were approximately 200 mm3 (N=5 mice per group). Vδ1 γδ T cells expressing CAR and CAR-mbIL-12 were administered on Day 0 as a single bolus dose. Human IL-2 supplementation was given three times a week for the duration of the study. Tumor volume and mouse weight were monitored over the duration of the experiment. The results are shown in FIG. 21. The CAR-mbIL-12 Vδ1 γδ T cells were associated with a substantially greater inhibition of tumor cell growth compared to the CAR Vδ1 γδ T cell group at the suboptimal treatment dose. The data supports the enhanced functionality of Vδ1 γδ T cells expressing CAR-mbIL-12. [00496] Example 12: CBL-B KO enhances the function of Vδ1 T cells [00497] Healthy donor peripheral blood mononuclear cells (PBMCs) were used to activate, expand, CRISPR-gene edit, and engineer Vδ1 γδ T cells to express a CAR. To knockout the CBL- B gene in Vδ1 T cells, a ribonucleoprotein (RNP) complex comprising CRISPR-Cas9 (Integrated DNA Technologies) and CBL-B sgRNAs were combined with activated Vδ1 T cells from PBMCs (P3 Primary Cell 4D-Nucleofector X Kit L, Lonza). The RNP complex was delivered into Vδ1 T cells by electroporation (Lonza 4D-Nucleofector). Post electroporation, the cells were transferred to T flasks (Corning) containing X-Vivo15 (Lonza) for cell recovery prior to transduction with gammaretroviral vector encoding the CAR and then were subsequently expanded. The results are shown in FIGS.22A-22B. (FIG. 22A) To determine CBL-B KO efficiency, genomic DNA was extracted from cell pellets using the NucleoSpin Tissue kit (MACHARY-NAGEL). The edited region was amplified by PCR using previously in-house optimized primers. The PCR reaction was run on an agarose gel, and the prominent band excised, and gel extracted using NucleoSpin Gel and PCR Cleanup kit (MACHARY-NAGEL). The extracted DNA was sequenced by Sanger sequencing (Sequetech). % Indels were determined using the ICE Analysis tool (Synthego). (FIG. Page 144 of 203 1097657169\1\AMERICAS 22B) To demonstrate enhanced function of CBL-B KO CAR-transduced and untransduced Vδ1 T cells, the cytolytic activity against the PSMA+ PC3 cell line was evaluated using an in vitro repetitive stimulation assay (with PC3-PMSA targets in prostate adenocarcinoma). PC3-PSMA NIR fluorescently labeled cell line and effector cells were added into a 96-well plate at an E:T ratio of ~3:1 with or without IL-2. Each stimulation was monitored for 3 days after which fresh target cells were plated in another 96-well plate. The effector cells from each stimulation were then transferred into the plate with fresh target cells. The co-cultures were monitored every 4 hours using Incucyte® SX5 (Satorius). The Cytotoxicity Index was calculated based on the remaining viable target tumor cells by dividing the total NIR object area of each time point by the value at time 0 for each stimulation. [00498] Cas9 Multi-guide sgRNA sequences: [00499] AAGACUCUUUAAAGAAGGCA (SEQ ID NO: 341) [00500] AGUACUCAUUCUCACUGAGU (SEQ ID NO: 342) [00501] CGUAAAUGCUGAUAUGUAUC (SEQ ID NO: 343) [00502] Cas9 Single-guide: TAATCTGGTGGACCTCATGA (AGG) (SEQ ID NO: 344). [00503] Example 13: Roquin-1 KO enhances the function of Vδ1 T cells [00504] Healthy donor peripheral blood mononuclear cells (PBMCs) were used to activate, expand, CRISPR-gene edit, and engineer Vδ1 γδ T cells to express a CAR. To knockout the Roquin-1 gene in Vδ1 T cells, a ribonucleoprotein (RNP) complex comprising CRISPR-Cas9 (Integrated DNA Technologies) and Roquin-1 sgRNAs were combined with activated Vδ1 T cells from PBMCs (P3 Primary Cell 4D-Nucleofector X Kit L, Lonza). The RNP complex was delivered into Vδ1 T cells by electroporation (Lonza 4D-Nucleofector). Post electroporation, the cells were transferred to T flasks (Corning) containing X-Vivo15 (Lonza) for cell recovery prior to transduction with gammaretroviral vector encoding the CAR and then were subsequently expanded. The results are shown in FIGS. 23A-23B. (FIG.23A). To determine the Roquin KO efficiency genomic DNA was extracted from cell pellets using the NucleoSpin Tissue kit (MACHARY-NAGEL). The edited region was amplified by PCR using previously in-house optimized primers. The PCR reaction was run on an agarose gel, and the prominent band was excised, and gel extracted using NucleoSpin Gel and PCR Cleanup kit (MACHARY-NAGEL). Page 145 of 203 1097657169\1\AMERICAS The extracted DNA was sequenced by Sanger sequencing (Sequetech). % Indels were determined using the ICE Analysis tool (Synthego). Edits were first generated using a 1:1:1 ratio of 3 gRNA sequences to demonstrate the ability to edit the gene in Vd1 T cells, prior to single-guide optimization to identify a gRNA with the highest KO efficiency. (FIG. 23B) To demonstrate enhanced function of Roquin-1 KO CAR-transduced and untransduced Vδ1 T cells, the cytolytic activity against the PSMA+ PC3 cell line was evaluated using an in vitro repetitive stimulation assay (with PC3-PMSA targets in prostate adenocarcinoma). PC3-PSMA NIR fluorescently labeled cell line and effector cells were added into a 96-well plate at an E:T ratio of ~3:1 with or without IL-2. Each stimulation was monitored for 3 days after which fresh target cells were plated in another 96-well plate. The effector cells from each stimulation were then transferred into the plate with fresh target cells. The co-cultures were monitored every 4 hours using Incucyte® SX5 (Satorius). The Cytotoxicity Index was calculated based on the remaining viable target tumor cells by dividing the total NIR object area of each time point by the value at time 0 for each stimulation. [00505] Multi-guide sgRNA sequences: [00506] AAGCCCAUCAGUUUGGGUUG (SEQ ID NO: 345) [00507] UGUACAAGCUCCACAAUGGA (SEQ ID NO: 346) [00508] CAAAUGGGCAAGCCUUGCGG (SEQ ID NO: 347) [00509] In-house optimized single gRNA sequence: CCTGAATAAACTCCACCGCA (AGG) (SEQ ID NO: 348). [00510] Example 14: ICAM-1 and CD58 KO enhances the in vitro cell survival of CAR Vδ1 T cells in the presence of allogeneic PBMCs [00511] Healthy donor peripheral blood mononuclear cells (PBMCs) were used to activate, expand, CRISPR-gene edit, and engineer Vδ1 γδ T cells to express a CAR. To knockout the CISH gene in Vδ1 T cells, a ribonucleoprotein (RNP) complex comprised of CRISPR-Cas9 (Integrated DNA Technologies) or -MAD7 (Aldevron) and ICAM-1 or CD58 sgRNA were combined with activated Vδ1 T cells from PBMCs (P3 Primary Cell 4D-Nucleofector X Kit L, Lonza). The RNP complex was delivered into Vδ1 T cells by electroporation (Lonza 4D-Nucleofector). Post electroporation, the cells were transferred to T flasks (Corning) containing X-Vivo15 (Lonza) for cell recovery prior to transduction with gammaretroviral vector encoding the CAR and then were Page 146 of 203 1097657169\1\AMERICAS subsequently expanded. To determine the KO efficiency of ICAM-1 and CD58, WT (unedited), ICAM-1 KO, and CD58 KO CAR Vδ1 γδ T cells were stained with anti-ICAM-1, anti-CD58, or isotype control antibody conjugated to PE (BioLegend) and acquired using the Novocyte flow cytometer (Agilent Technologies). [00512] The results are shown in FIGS.24A-24B. (FIG.24A) Greater than 80% KO efficiency was observed for both ICAM-1 and CD58 KO in CAR Vδ1 γδ T cells. (FIG.24B) To evaluate if the of ICAM-1 and CD58 can enhance the survival of CAR Vδ1 T cells in the presence of allogeneic PBMCs, a 10-day mixed-lymphocyte reaction assay (MLR) was performed. WT (unedited), CISH KO, ICAM-1 KO, and CD58 KO CAR Vδ1 γδ T cells (target cells) were co- cultured with allogeneic PBMCs (effector cells) at an E:T ratio of 10:1 in the presence of IL-2. During the co-culture, half of the medium was replaced with fresh media containing IL-2 every 2- 3 days. At days 3, 6, and 10, cells were harvested from the co-culture and stained for CAR+ and Vδ1+ γδ T cells using B7-H6 conjugated to APC (Adicet Bio generated) for CAR detection and anti-Vδ1 antibody conjugated to PE-CY7 (Adicet Bio generated) for Vδ1 T cell detection. The cells were acquired using the Novocyte flow cytometer to obtain absolute counts of double positive CAR+ Vδ1+ T cells. To determine the % target cell change at each time point, the absolute count of WT CAR+ Vδ1+ γδ T cells was compared to the KO CAR+ Vδ1+ γδ T cell count conditions using the formula: [(WT CAR+ Vδ1+ γδ T cells count – KO CAR+ Vδ1+ γδ T cell count) ÷ (WT CAR+ Vδ1+ γδ T cells count)] x 100. Based on the % target cell change relative to the WT CAR+ Vδ1+ γδ T cells counts, only the ICAM-1 KO and CD58 KO CAR+ Vδ1+ γδ T cells had an increased % target change, suggesting the disruption of ICAM-1 and CD58 reduced the in vitro killing by allogeneic PBMCs for improved cell survival. [00513] CAS9 ICAM-1 sgRNA: AAAGUCAUCCUGCCCCGGGG (SEQ ID NO: 349) [00514] MAD7 ICAM-1 sgRNA: TTTG/AATAGCACATTGGTTGGCTAT (SEQ ID NO: 350) [00515] CAS9 CD58 sgRNA: AAAUAUAUGGUGUUGUGUAU (SEQ ID NO: 351) [00516] MAD7 CD58 sgRNA: TTTA/GACACTGTGTCAGGTAGCCTC (SEQ ID NO: 352). [00517] The embodiments and examples described above are intended to be merely illustrative and non-limiting. Those skilled in the art will recognize or will be able to ascertain using no more Page 147 of 203 1097657169\1\AMERICAS than routine experimentation, numerous equivalents of specific compounds, materials and procedures. All such equivalents are considered to be within the scope and are encompassed by the appended claims. Table 14. Illustrative Immunoglobulin Sequences – Heavy Chain Variable Region Name SEQ ID Heavy Chain Variable Region E T E T E A L T L L T L T L T T L
Figure imgf000149_0001
Page 148 of 203 1097657169\1\AMERICAS 96_021 25 EVQLLESGGGLVQPGGSLRLSCAASGFTFNAYPMTWVRQAPGKGL EWVSWIEGRGTETYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDT E T E A E V E A L T L T E A L AI
Figure imgf000150_0001
Table 15. Illustrative Immunoglobulin Sequences – Light Chain Variable Region T L L L P LI
Figure imgf000150_0002
Page 149 of 203 1097657169\1\AMERICAS 97_006 10 DIQMTQSPSSLSASVGDRVTITCRASHSISSYLNWYQQKPGKAPKLLI YGASTLQAGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYDTPL P L T LI P L LI L K Q L T LI L L P L A LI L L PI LI L L P
Figure imgf000151_0001
Page 150 of 203 1097657169\1\AMERICAS 104_005 40 DIQMTQSPSSLSASVGDRVTITCRASQSISGYLNWYQQKPGKAPKLL IYEDTKRPSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQTYDTP LI P
Figure imgf000152_0001
Table 16. Complementarity Determining Regions, Heavy Variable Region (Kabat) Heavy Chain Variable Region CDRs P S
Figure imgf000152_0002
Page 151 of 203 1097657169\1\AMERICAS 97_035 SQGMS AISGSGDYTYYADSVKG EGMATTGIPFDY (SEQ ID NO: 57) (SEQ ID NO: 77) (SEQ ID NO: 97)
Figure imgf000153_0001
Table 17. Complementarity Determining Regions, Light Variable Region (Kabat) Light Chain Variable Region CDRs
Figure imgf000153_0002
Page 152 of 203 1097657169\1\AMERICAS 97_017 RASQSISPYLN DASSLHS QQSYDTPLT (SEQ ID NO: 116) (SEQ ID NO: 136) (SEQ ID NO: 156)
Figure imgf000154_0001
Table 18: Sequence information (Amino acid/Nucleotide Sequence) SEQ ID Name Sequence
Figure imgf000154_0002
Page 153 of 203 1097657169\1\AMERICAS 184 Furin P2A RAKRSGSGATNFSLLKQAGDVEENPGP (amino acid) C A T A C C T C T G G G T
Figure imgf000155_0001
Page 154 of 203 1097657169\1\AMERICAS CCTTGGTAACAAGGACCCACGGGGCCAAAAGCCACGCCCACA CGGGCCCGTCATGTGTGCAACCCCAGCACGGCGACTTTACTGC CT A A G C K L G A N
Figure imgf000156_0001
Page 155 of 203 1097657169\1\AMERICAS 208 CD27 QRRKYRSNKGESPVEPAEPCHYSCPREEEGSTIPIQEDYRKPE costimulatory PACSP A T C T A T C C T
Figure imgf000157_0001
Page 156 of 203 1097657169\1\AMERICAS 222 Ig hinge region KCCVDCP (amino acid) E G A T R R G I A A G A C
Figure imgf000158_0001
Page 157 of 203 1097657169\1\AMERICAS GTCCAGGAGCGCACCATCTTCTTCAAGGACGACGGCAACT ACAAGACCCGCGCCGAGGTGAAGTTCGAGGGCGACACCC A C A C G A R F G H ta g g a g a a c ca g a a c g t
Figure imgf000159_0001
Page 158 of 203 1097657169\1\AMERICAS (amino acid) 240 dnTGFBR2 atgggtcgggggctgctcaggggcctgtggccgctgcacatcgtcctgtggacgcgtatcgcc K I g c g at g c g tt G C A L L P S G C A C G T G G A
Figure imgf000160_0001
Page 159 of 203 1097657169\1\AMERICAS GATGCGAGAATCAGGGCCTCGTGGAAGCTGCCCCTGGAAC TGCTCAGAGCGACACCACATGCAAGAACCCTCTGGAACCC G A C A G T A G G G D D G M G A C T G T T
Figure imgf000161_0001
Page 160 of 203 1097657169\1\AMERICAS CTCCAGAGGGCAGCGATAGCACAGCCCCTTCTACACAAGA GCCCGAGGCTCCTCCTGAGCAGGATCTGATTGCCTCTACA C T C T C T G C A T T A C C C A G A T C A G G C A C
Figure imgf000162_0001
Page 161 of 203 1097657169\1\AMERICAS GGTTTACAATATCACGCGACAATTCCAAGAATACGCTGTA TTTGCAAATGAACAGTCTGCGCGCGGAAGACACAGCTGTA A A G T C T C A G A A C C C G G C T C T T T T
Figure imgf000163_0001
Page 162 of 203 1097657169\1\AMERICAS AGGTACAGATTTCACCCTTACAATCAGTAGTCTTCAACCA GAGGATTTCGCAACCTATTACTGCCAGCAAACCTATGACA A C C G A C A G A T C C C T A G T A A C G A C T
Figure imgf000164_0001
Page 163 of 203 1097657169\1\AMERICAS CTACTATTGTGCGAGAGATTTGGGATATGACCATTTTGACT CATGGGGCCAAGGGACCCTCGTGACCGTCTCCTCA T A C C A C C C G A A C T C A C C T C G T T A A G T
Figure imgf000165_0001
Page 164 of 203 1097657169\1\AMERICAS 271 97_017 HCVR GAGGTACAACTTCTCGAAAGCGGTGGTGGATTGGTGCAAC (nucleotide) CAGGTGGAAGCCTTAGGCTCAGTTGTGCCGCAAGTGGTTT A A G T T A G G T T G T C A G C C T G T T C T T A
Figure imgf000166_0001
Page 165 of 203 1097657169\1\AMERICAS GGGAATTTCCACTTACTTGAATTGGTATCAACAGAAACCG GGAAAAGCACCTAAGCTTCTCATTTACGGTGCTTCAGCAC C A C T C G T C C A G C A C C G T A T A A C
Figure imgf000167_0001
Page 166 of 203 1097657169\1\AMERICAS GTGGGACATGCGACTTATTATGCGGATTCCGTGAAGGGAC GGTTTACAATTTCTCGCGACAATTCCAAGAATACTTTATAC T A A T C G A A C T A A G G A C A C C T T T T A T C C
Figure imgf000168_0001
Page 167 of 203 1097657169\1\AMERICAS GGCACTGACTTTACCCTGACTATTAGCAGCTTGCAACCAG AAGACTTTGCGACGTACTACTGTCAGCAGTCATATTCCAA A C T C C T A T G C C T A G T A A T A C G A C
Figure imgf000169_0001
Page 168 of 203 1097657169\1\AMERICAS 292 47.39 LCVR GACATCAAGATGACCCAGTCTCCATCTTCCATGTATGCAT (nucleotide) CTCTAGGAGAGAGAGTCACTATCACTTGCAAGGCGAGTCA A T C S A G S T T G E G C G G C A G A T A G T C A
Figure imgf000170_0001
Page 169 of 203 1097657169\1\AMERICAS ACGGGGCAGAAAGAAACTCCTGTATATATTCAAACAACCA TTTATGAGACCAGTACAAACTACTCAAGAGGAAGATGGCT G A G T C P Q G R A Y C C C T A C G G T A A C G C G C G
Figure imgf000171_0001
Page 170 of 203 1097657169\1\AMERICAS CCCTTGGCCGGGACTTGTGGGGTCCTTCTCCTGTCACTGGT TATCACCCTTTACTGCAAACGGGGCAGAAAGAAACTCCTG A A G G C C A C S A G S T T G E G C G G C A G A T A G
Figure imgf000172_0001
Page 171 of 203 1097657169\1\AMERICAS CCAGCGCCGCGACCACCAACACCGGCGCCCACCATCGCGT CGCAGCCCCTGTCCCTGCGCCCAGAGGCGTGCCGGCCAGC C A A T A G T C P Q G R A Y C C C T A C G G T A A C G C
Figure imgf000173_0001
Page 172 of 203 1097657169\1\AMERICAS AGCACCCCTCCTCTGACATTTGGACAGGGCACCAAGGTGG AAATCAAGACCACGACGCCAGCGCCGCGACCACCAACAC C C G T G A G G C C A C S W L N G K S G H C G T C A T G G T G
Figure imgf000174_0001
Page 173 of 203 1097657169\1\AMERICAS ATGCCAGCAATCTGCAGAGCGGCGTGCCCAGCAGATTTTC TGGCTCTGGCAGCGGCACCGACTTCACCCTGACCATATCT C A G A C T T G A C Q S L I P L C C A A C G T C
Figure imgf000175_0001
Page 174 of 203 1097657169\1\AMERICAS AACAACCTGAACTGGTATCAGCAGAAGCCCGGCAAGGCC CCTAAGCTGCTGATCTACGATGCCAGCAATCTGCAGAGCG A A C A C G T A G C T S G F F S P L E V E C G C G G G
Figure imgf000176_0001
Page 175 of 203 1097657169\1\AMERICAS CTGGAATCTGGCGGAGGACTGGTTCAACCTGGCGGCTCTC TGAGACTGTCTTGTGCCGCCAGCGGCTTCACCTTCAGCAG A A C C T A C G T C A G A T Y T F P R Y A C T T C
Figure imgf000177_0001
Page 176 of 203 1097657169\1\AMERICAS CCTCCACCTGATTTTGGCGGAGGCACCAAGGTGGAAATCA AGGGAGGCGGAGGATCTGGTGGTGGTGGATCTGGCGGCG T C T C C T G G T T C A G T A C C T EI S K L A G E G C G
Figure imgf000178_0001
Page 177 of 203 1097657169\1\AMERICAS AACTGGTATCAGCAGAAGCCCGGCAAGGCCCCTAAGCTGC TGATCTACAGAACCAGCTGGCTGCAGAGCGGCGTGCCAAG C C G C A T C C C G A T G T A T G K Y L L P P D
Figure imgf000179_0001
Page 178 of 203 1097657169\1\AMERICAS 312 PL875 (-signal GATATCCAGATGACACAGAGCCCCAGCAGCCTGTCTGCCT peptide) CTGTGGGAGACAGAGTGACCATCACCTGTCGGGCCAGCCA C C C A T G G T C G C A A G T T G A T C G T G G F R G K R G
Figure imgf000180_0001
Page 179 of 203 1097657169\1\AMERICAS GKPQRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKG HDGLYQGLSTATKDTYDALHMQALPPR C C G G G G A T T G G A G T C G T A C A K Y E W P
Figure imgf000181_0001
Page 180 of 203 1097657169\1\AMERICAS GTCGVLLLSLVITLYCKRGRKKLLYIFKQPFMRPVQTTQEED GCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNELN K L A C A T C G C C C T T T C C T C G A G G G C A S D D L
Figure imgf000182_0001
Page 181 of 203 1097657169\1\AMERICAS LQAGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYDTPL TFGGGTKVEIKTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGG K S G H C G T C A G G G C A C G G T A C T A G C G C C P M
Figure imgf000183_0001
Page 182 of 203 1097657169\1\AMERICAS (amino acid) NSLRAEDTAVYYCARDLGYDHFDSWGQGTLVTVSSGGGGS GGGGSGGGGSDIQMTQSPSSLSASVGDRVTITCRASHSISSYL I P L C T T G T A C C A A T T C G A G G G C A
Figure imgf000184_0001
Page 183 of 203 1097657169\1\AMERICAS 321 PL1012 MSVPTQVLGLLLLWLTDARCEVQLLESGGGLVQPGGSLRLS (amino acid) CAASGFTFGDYDMWWVRQAPGKGLEWVSVISSSGGETSYA G S T A R G C T T G A G T T A C T T T C T C G T A C
Figure imgf000185_0001
Page 184 of 203 1097657169\1\AMERICAS CGCCGGAGGGGCAAGGGGCACGATGGCCTTTACCAGGGT CTCAGTACAGCCACCAAGGACACCTACGACGCCCTTCACA A G P P D C T C C T C A A T G C T C C T C G A G G
Figure imgf000186_0001
Page 185 of 203 1097657169\1\AMERICAS GAACTGCAGAAAGATAAGATGGCGGAGGCCTACAGTGAG ATTGGGATGAAAGGCGAGCGCCGGAGGGGCAAGGGGCAC A T G Y G K R G G G T C T T C T C C G G C C G T G C A
Figure imgf000187_0001
Page 186 of 203 1097657169\1\AMERICAS CAGGGCCAGAACCAGCTCTATAACGAGCTCAATCTAGGAC GAAGAGAGGAGTACGATGTTTTGGACAAGAGACGTGGCC A A K E A P L A A C G A G A A T G G G G C A G
Figure imgf000188_0001
Page 187 of 203 1097657169\1\AMERICAS AAGGAGGATGTGAACTGAGAGTGAAGTTCAGCAGGAGCG CAGACGCCCCCGCGTACCAGCAGGGCCAGAACCAGCTCTA T A G G T K G D A R G G A G T G T A C G C A C T C T
Figure imgf000189_0001
Page 188 of 203 1097657169\1\AMERICAS CCTGTCACTGGTTATCACCCTTTACTGCAAACGGGGCAGA AAGAAACTCCTGTATATATTCAAACAACCATTTATGAGAC G T A C A K Y E E A T A L A T G C A A T C T C G G
Figure imgf000190_0001
Page 189 of 203 1097657169\1\AMERICAS CTGGACTTCGCCTGTGATATCTACATCTGGGCGCCCTTGGC CGGGACTTGTGGGGTCCTTCTCCTGTCACTGGTTATCACCC T G A T A A A C T G K G G G L A K D A T C T G G T A G G A
Figure imgf000191_0001
Page 190 of 203 1097657169\1\AMERICAS CCGTGAGCTCCACCACGACGCCAGCGCCGCGACCACCAAC ACCGGCGCCCACCATCGCGTCGCAGCCCCTGTCCCTGCGC C G G C A C C A G C G L L A T C K M A A T A G G G C G
Figure imgf000192_0001
Page 191 of 203 1097657169\1\AMERICAS CGGTCTATTATTGCGCCCGGGGTTGGGATCTGGATTATTG GGGACAAGGAACACTGGTTACCGTGAGCTCCACCACGAC G G T A A T A G T C E Y G T F P R Y C T A T A C A G G
Figure imgf000193_0001
Page 192 of 203 1097657169\1\AMERICAS CAAACAGATTGGTAGATGGGGTCCCATCAAGGTTCAGTGG CAGTGGATCTGGGCAAGATTATTCTCTCACCATCAGCAGC T T C C G G C A C C A G C G
Figure imgf000194_0001
Page 193 of 203 1097657169\1\AMERICAS

Claims

CLAIMS: What is claimed is: 1. An affinity binding entity comprising an antigen binding domain that specifically binds to B7 homolog 6 (B7H6), wherein the antigen binding domain comprises: a heavy chain variable region/light chain variable region (HCVR/LCVR) sequence pair selected from the group consisting of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, 15/16, 17/18, 19/20, 21/22, 23/24, 25/26, 27/28, 29/30, 31/32, 33/34, 35/36, 37/38, and 39/40; or the six CDRs of a HCVR/LCVR sequence pair selected from the group consisting of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, 15/16, 17/18, 19/20, 21/22, 23/24, 25/26, 27/28, 29/30, 31/32, 33/34, 35/36, 37/38, and 39/40. 2. The affinity binding entity of claim 1, wherein said antigen binding domain comprises: a HCVR/LCVR sequence pair selected from the group consisting of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, and 15/16; or the six CDRs of a HCVR/LCVR sequence pair selected from the group consisting of SEQ ID NOs: 1/2, 3/4, 5/6, 7/8, 9/10, 11/12, 13/14, and 15/16. 3. The affinity binding entity of claim 1, wherein said antigen binding domain comprises a HCVR/LCVR sequence pair of SEQ ID NOs: 1/2; or the six CDRs of a HCVR/LCVR sequence pair of SEQ ID NOs: 1/2. 4. The affinity binding entity of claim 1, wherein said antigen binding domain comprises a HCVR/LCVR sequence pair of SEQ ID NOs: 3/4; or the six CDRs of a HCVR/LCVR sequence pair of SEQ ID NOs: 3/4. 5. The affinity binding entity of any one of claims 1-4, wherein said affinity binding entity is an antibody, or an antibody fragment; optionally wherein said affinity binding entity is selected from the group consisting of scFv, Fab, Fab’, Fv, F(ab’)2, dsFv, dAb, and any combination or plurality thereof. Page 194 of 203 1097657169\1\AMERICAS (KLRF1), OX-40, PAG/Cbp, programmed death-1 (PD-1), PSGL1, SELPLG (CD162), Signaling Lymphocytic Activation Molecules (SLAM proteins), SLAM (SLAMF1; CD150; IPO-3), SLAMF4 (CD244; 2B4), SLAMF6 (NTB-A; Lyl08), SLAMF7, SLP-76, TNF receptor proteins, TNFR2, TNFSF14, a Toll ligand receptor, TRANCE/RANKL, VLA1, or VLA-6, or a fragment, truncation, or a combination thereof. 12. The CAR of claim 11, wherein said TM domain comprises a TM domain of CD8, preferably wherein said CD8 TM domain is a TM domain of CD8 alpha; optionally wherein said TM domain comprises the amino acid sequence set forth as SEQ ID NO: 211. 13. The CAR of any one of claims 8-12, further comprising at least one costimulatory domain; optionally wherein said costimulatory domain comprises a costimulatory domain of TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, CARD11, B7-H3, CEACAM1, CRTAM, CD2, CD3C, CD4, CD7, CD8α, CD8β, CD11a, CD11b, CD11c, CD11d, IL2Rβ, IL2γ, IL7Rα, IL4R, IL7R, IL15R, IL21R, CD18, CD19, CD19a, CD27, CD28, CD29, CD30, CD40, CDS, CD49a, CD49D, CD49f, CD54 (ICAM), CD69, CD70, CD80, CD83, CD84, CD86, CD96 (Tactile), CD100 (SEMA4D), CD103, CD134 (OX40), CD137 (4-1BB), CD152 (CTLA-4), CD160 (BY55), CD162 (SELPLG), CD244 (2B4), CD270 (HVEM), CD226 (DNAM1), CD229 (Ly9), CD278 (ICOS), ICAM-1, LFA-1 (CD11a/CD18), FcR, FcγRI, FcγRII, FcγRIII, LAT, NKG2C, SLP76, TRIM, ZAP70, GITR, BAFFR, LTBR, LAT, GADS, LIGHT, HVEM (LIGHTR), KIRDS2, ITGA4, ITGA6, ITGAD, ITGAE, ITGAL, ITGAM, ITGAX, ITGB1, ITGB2, ITGB7, NKG2C, NKG2D, IA4, VLA-1, VLA-6, SLAM (SLAMF1, CD150, IPO-3), SLAMF4, SLAMF6 (NTB-A, Ly108), SLAMF7, SLAMF8 (BLAME), SLP-76, PAG/Cbp, NKp80 (KLRF1), NKp44, NKp30, NKp46, BTLA, JAML, CD150, PSGL1, TSLP, TNFR2, or TRANCE/RANKL, or a portion thereof, or combinations thereof. 14. The CAR of claim 13, wherein said costimulatory domain is a 4-1BB costimulatory domain; optionally wherein said 4-1BB costimulatory domain comprises an amino acid sequence set forth as SEQ ID NO: 202. Page 196 of 203 1097657169\1\AMERICAS
15. The CAR of any one of claims 8-14, further comprising one or more intracellular signaling domains, preferably wherein said intracellular signaling domain is a CD3ζ intracellular signaling domain; optionally wherein the CD3ζ intracellular signaling domain comprises an amino acid sequence set forth as SEQ ID NO: 229, 231, or 232. 16. The CAR of any one of claims 8-15, further comprising a signal peptide; optionally wherein said signal peptide comprises an amino acid sequence set forth as SEQ ID NO: 170. 17. An isolated polynucleotide comprising a nucleic acid sequence encoding the affinity binding entity of any one of claims 1-7. 18. An expression vector comprising the isolated polynucleotide of claim 17, operably linked to a cis-acting regulatory element. 19. A cell comprising the affinity binding entity of any one of claims 1-7, the isolated polynucleotide of claim 17, and/or the expression vector of claim 18. 20. An isolated polynucleotide comprising a nucleic acid sequence encoding the CAR of any one of claims 8-16. 21. The isolated polynucleotide of claim 20, further comprising a nucleic acid sequence encoding at least one multicistronic linker region; optionally wherein the multicistronic region encodes a cleavage sequence and/or an internal ribosomal entry site (IRES). 22. The isolated polynucleotide of claim 21, wherein the cleavage sequence is selected from T2A, F2A, P2A, E2A, furin, and furin-P2A (FP2A). 23. The isolated polynucleotide of any one of claims 22-22, further comprising a nucleic acid sequence encoding one or more additional polypeptides. Page 197 of 203 1097657169\1\AMERICAS
24. The isolated polynucleotide of claim 23, wherein the one or more additional polypeptides is selected from the group consisting of lymphotoxin beta receptor (LTBR), low-affinity nerve growth factor receptor (LNGFR), a dominant negative (dn) receptor for TGF-beta or Fas, a truncated form of the human epidermal growth factor receptor (EGFRt), membrane-bound IL-12 (mbIL-12), a fluorescent protein, a gamma chain cytokine, CD19, CD20, dominant negative Fas, membrane-bound IL-12, CAR that binds to CD70, or any combination thereof. 25. The isolated polynucleotide of claim 24, wherein the one or more additional polypeptides is dnTGFβR2, optionally wherein said dnTGFβR2 comprises an amino acid sequence set forth as SEQ ID NO: 241. 26. The isolated polynucleotide of claim 24, wherein the one or more additional polypeptides is LTBR, optionally wherein said LTBR comprises an amino acid sequence set forth as SEQ ID NO: 245. 27. The isolated polynucleotide of claim 24, wherein the one or more additional polypeptides is EGFRt, optionally wherein said EGFRt comprises an amino acid sequence set forth as SEQ ID NO: 237. 28. The isolated polynucleotide of claim 24, wherein the one or more additional polypeptides is LNGFR, optionally wherein said LNGFR comprises an amino acid sequence set forth as SEQ ID NO: 249. 29. The isolated polynucleotide of any one of claims 23-28, wherein the one or more additional polypeptides are operably linked to a nucleic acid sequence encoding a signal peptide; optionally wherein the signal peptide comprises an amino acid sequence selected from SEQ ID NO: 235, SEQ ID NO: 239, SEQ ID NO: 243, SEQ ID NO: 247, and SEQ ID NO: 196. 30. The isolated polynucleotide of any one of claims 20-29, comprising a nucleic acid sequence of SEQ ID NO: 294, 298, 302, 306, 310, 314, 318, 322, 326, 330, or 334. Page 198 of 203 1097657169\1\AMERICAS
31. An expression vector comprising the isolated polynucleotide of any one of claims 20-30, operably linked to a cis-acting regulatory element. 32. A γδ T cell comprising: (a) a nucleic acid sequence encoding a chimeric antigen receptor (CAR), said CAR comprising an antigen binding domain that specifically binds to B7 homolog 6 (B7H6); and/or (b) a polypeptide comprising a CAR comprising an amino acid sequence encoded by the nucleic acid of (a); wherein the γδ T cell functionally expresses the binding domain of the polypeptide or nucleic acid sequence encoded CAR on the surface of the γδ T cell. 33. The γδ T cell of claim 32, wherein the CAR comprises the affinity binding entity according to any one of claims 2-7; or wherein the nucleic acid sequence comprises the isolated polynucleotide of any one of claims 20-30, or the expression vector of claim 31. 34. A modified immune cell, comprising the CAR of any one of claims 8-16, the isolated polynucleotide of any one of claims 20-30, or the expression vector of claim 31. 35. The modified immune cell of claim 34, wherein said modified immune cell is a ^ ^ ^T cell, a ^ ^ ^ ^ ^ ^ ^cell, an αβ T cell, a NK cell, a NKT cell, or a macrophage. 36. The modified immune cell of claim 35, wherein said modified immune cell is a ^ ^ ^T cell; optionally wherein said ^ ^ ^T cell is a δ1, a δ2, a δ3, or a δ4 γδ T cell, preferably a δ2- γδ T cell, more preferably a δ1 γδ T cell. 37. The modified immune cell of any one of claims 34-36, or the γδ T cell of claim 32 or claim 33, wherein said modified immune cell or said γδ T cell exhibits in vitro and/or in vivo cell killing activity against a tumor cell that exhibits cell surface expression of B7H6. 38. The modified immune cell of any one of claims 34-37, or the γδ T cell of claim 32 or claim 33, wherein said modified immune cell or γδ T cell proliferates in response to contact with the Page 199 of 203 1097657169\1\AMERICAS tumor cell that exhibits cell surface expression of B7H6; optionally wherein the modified immune cell or γδ T cell proliferates in a host organism that comprises a tumor cell that exhibits cell surface expression of B7H6. 39. The modified immune cell of any one of claims 34-38, or the γδ T cell of any one of claims 33-34, wherein the modified immune cell or the γδ T cell expresses pro-inflammatory cytokines after contact with a tumor cell that exhibits cell surface expression of B7H6. 40. The modified immune cell of any one of claims 34-39, or the γδ T cell of any one of claims 33-34, further comprising at least one disrupted gene. 41. The modified immune cell or the γδ T cell of claim 40, wherein said at least one disrupted gene is cytokine inducible SH2-containing protein (CISH), Cbl proto-oncogene B (CBL-B), Zinc Finger Protein 91 (ZFP91), CD58, ICAM-1, or any combination thereof. 42. A method of making the modified immune cell of any one of claims 34-41, the γδ T cell of any one of claims 32-33, wherein the method comprises transfecting immune cell(s) or γδ T cell(s) with a construct comprising the expression vector of claim 31, optionally wherein said cell(s) has/have at least one disrupted gene. 43. The method of claim 42, wherein the method comprises retroviral transduction. 44. The method of claim 42 or claim 43, wherein the method comprises ex vivo expansion of the immune cell(s) or γδ T cell(s), wherein the ex vivo expansion is performed before transfection and/or after transfection of the expression vector. 45. An antibody-drug conjugate (ADC), comprising the affinity binding entity of any one of claims 1-7. 46. A pharmaceutical composition comprising the affinity binding entity of any one of claims 1- 7, or an ADC of claim 45, and a pharmaceutically acceptable carrier. Page 200 of 203 1097657169\1\AMERICAS
47. A pharmaceutical composition comprising a plurality of modified immune cells according to any one of claims 34-41, or a plurality of γδ T cells according to any one of claims 32-33; optionally wherein the plurality comprises a composition that is at least 60%, 80%, or from about 60% or 80% to about 90% or 95% δ1, δ2, δ3, or δ4 γδ T cells, preferably δ1 or δ2 γδ T cells, more preferably δ2- γδ T cells, most preferably δ1 γδ T cells, and a pharmaceutically acceptable carrier. 48. The pharmaceutical composition of claim 47, wherein the plurality comprises at least about 107 modified immune cells or γδ T cells, respectively, preferably from about 108 modified immune cells or γδ T cells to about 1011 modified immune cells or γδ T cells, respectively. 49. A method of inhibiting the growth of a cell that exhibits cell surface expression of B7H6, comprising contacting said cell with the affinity binding entity of any one of claims 1-7, the modified immune cell(s) of any one of claims 34-41, the γδ T cell(s) of any one of claims 32-33, the ADC of claim 45, or the pharmaceutical composition of any one of claims 46-48. 50. A method of killing a tumor cell that exhibits cell surface expression of B7H6, the method comprising contacting the tumor cell with a therapeutically effective amount of the affinity binding entity of any one of claims 1-7, the modified immune cell(s) of any one of claims 34-41, the γδ T cell(s) of any one of claims 32-33, ADC of claim 45, or the pharmaceutical composition of any one of claims 46-48. 51. The method of claim 50, wherein the method comprises introducing into a host organism comprising the tumor cell the therapeutically affective amount of the affinity binding entity, the modified immune cell(s), the γδ T cell(s), the ADC, or the pharmaceutical composition. 52. The method of claim 51, further comprising simultaneously or sequentially administering one or more methods to elevate common gamma chain cytokine(s); optionally wherein the administering one or more methods to elevate common gamma chain cytokine(s) comprises simultaneously or sequentially administering an amount of common gamma chain cytokine(s), Page 201 of 203 1097657169\1\AMERICAS lymphodepletion before introducing the modified immune cell(s) or the γδ T cell(s), and/or secretion of one or more common gamma chain cytokine(s) from the introduced modified immune cell(s) or γδ T cell(s). 53. The method of any one of claims 51-52, wherein the host organism is human, and the method is a method of treating cancer in a subject in need thereof. 54. Use of the affinity binding entity of any one of claims 1-7, the modified immune cell(s) of any one of claims 34-41, the γδ T cell(s) of any one of claims 32-33, the ADC of claim 45, or the pharmaceutical composition of claim 46-48, in the preparation of a medicament for the treatment of cancer. Page 202 of 203 1097657169\1\AMERICAS lymphodepletion before introducing the modified immune cell(s) or the yS T cell(s), and/or secretion of one or more common gamma chain cytokine(s) from the introduced modified immune cell(s) or y5 T cell(s).
53. The method of any one of claims 51-52, wherein the host organism is human, and the method is a method of treating cancer in a subject in need thereof.
54. Use of the affinity binding entity of any one of claims 1-7, the modified immune cell(s) of any one of claims 34-41, the y5 T cell(s) of any one of claims 32-33, the ADC of claim 45, or the pharmaceutical composition of claim 46-48, in the preparation of a medicament for the treatment of cancer.
PCT/US2023/034227 2022-09-30 2023-09-29 Affinity binding entities directed to b7h6 and methods of use thereof WO2024073111A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263411988P 2022-09-30 2022-09-30
US63/411,988 2022-09-30

Publications (1)

Publication Number Publication Date
WO2024073111A2 true WO2024073111A2 (en) 2024-04-04

Family

ID=90479309

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/034227 WO2024073111A2 (en) 2022-09-30 2023-09-29 Affinity binding entities directed to b7h6 and methods of use thereof

Country Status (1)

Country Link
WO (1) WO2024073111A2 (en)

Similar Documents

Publication Publication Date Title
JP6704954B2 (en) Affinity matured anti-CCR4 humanized monoclonal antibodies and methods of use
JP7062640B2 (en) Anti-idiotype antibody against anti-CD19 antibody
JP6847037B2 (en) Concomitant therapeutic agents containing anti-CD73 antibody and A2A receptor inhibitor and their use
CN112789293B (en) Single domain antibodies to CLL1 and constructs thereof
US10533055B2 (en) Antibodies and chimeric antigen receptors specific for CD19
CN103408661B (en) The method of disease-specific binding molecules and target is provided
JP6060073B2 (en) Antibody against CD122
CA2718573C (en) Targeting abcb5 for cancer therapy
US20100136030A1 (en) Antagonist ox40 antibodies and their use in the treatment of inflammatory and autoimmune diseases
KR20170081699A (en) Cd47 antibodies, methods, and uses
TW201702272A (en) T cell receptor-like antibodies specific for a PRAME peptide
KR20150123825A (en) Non-platelet depleting and non-red blood cell depleting cd47 antibodies and methods of use thereof
KR20150008049A (en) Cd47 antibodies and methods of use thereof
CN113056487A (en) anti-TNFR 2 antibodies and uses thereof
JP2019512207A (en) T cell receptor-like antibody specific for FOXP3-derived peptides
US20220288203A1 (en) Anti-podocalyxin antibodies and methods of using the same
IL293430A (en) Anti-idiotypic antibodies to bcma-targeted binding domains and related compositions and methods
JP2008502368A (en) Diagnosis and treatment of SIGLEC-6 related diseases
WO2023133360A2 (en) Anti-collagen triple helix repeat containing 1 (cthrc1) antibodies and methods of using the same
WO2022140670A2 (en) Anti-activin antibodies and methods of using the same
WO2024073111A2 (en) Affinity binding entities directed to b7h6 and methods of use thereof
WO2024035953A9 (en) Affinity binding entities directed to psma and methods of use thereof
WO2023020471A1 (en) Cd123-targetting antibodies and uses thereof in cancer therapies
WO2023019393A1 (en) Cd123-targetting antibodies, chimeric antigen receptors, and uses thereof
WO2021010326A1 (en) Anti-mutation type fgfr3 antibody and use therefor