US20220348631A1 - Receptors providing targeted costimulation for adoptive cell therapy - Google Patents

Receptors providing targeted costimulation for adoptive cell therapy Download PDF

Info

Publication number
US20220348631A1
US20220348631A1 US17/843,480 US202217843480A US2022348631A1 US 20220348631 A1 US20220348631 A1 US 20220348631A1 US 202217843480 A US202217843480 A US 202217843480A US 2022348631 A1 US2022348631 A1 US 2022348631A1
Authority
US
United States
Prior art keywords
cells
costar
cell
seq
domain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/843,480
Other languages
English (en)
Inventor
John Bridgeman
Robert Hawkins
Ruben Rodriguez
Gray Kueberuwa
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Instil Bio UK Ltd
Instil Bio Inc
Original Assignee
Instil Bio UK Ltd
Instil Bio 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 Instil Bio UK Ltd, Instil Bio Inc filed Critical Instil Bio UK Ltd
Priority to US17/843,480 priority Critical patent/US20220348631A1/en
Priority to US17/823,223 priority patent/US20230002470A1/en
Assigned to INSTIL BIO INC. reassignment INSTIL BIO INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: RODRIGUEZ, RUBEN
Publication of US20220348631A1 publication Critical patent/US20220348631A1/en
Assigned to INSTIL BIO (UK) LIMITED reassignment INSTIL BIO (UK) LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KUEBERUWA, Gray, HAWKINS, ROBERT, BRIDGEMAN, JOHN
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • 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
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4636Immune checkpoint inhibitors
    • 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
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • 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
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464429Molecules with a "CD" designation not provided for elsewhere
    • 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
    • A61K39/46448Cancer antigens from embryonic or fetal origin
    • A61K39/464482Carcinoembryonic antigen [CEA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70521CD28, CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • C12N15/625DNA sequences coding for fusion proteins containing a sequence coding for a signal sequence
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/59Reproductive system, e.g. uterus, ovaries, cervix or testes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins 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
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the present invention relates to a chimeric costimulatory antigen receptor (CoStAR) useful in adoptive cell therapy (ACT), and cells comprising the CoStAR.
  • CoStAR can act as a modulator of cellular activity enhancing responses to defined antigens.
  • the present invention also provides CoStAR proteins, nucleic acids encoding the CoStAR and therapeutic uses thereof.
  • T-cells may be genetically modified to retarget them towards defined tumor antigens. This can be done via the gene transfer of peptide (p)-major histocompatibility complex (MHC) specific T-cell Receptors (TCRs) or synthetic fusions between tumor specific single chain antibody fragment (scFv) and T-cell signaling domains (e.g. CD3 ⁇ ), the latter being termed chimeric antigen receptors (CARs).
  • MHC peptide
  • TCRs tumor specific T-cell Receptors
  • scFv tumor specific single chain antibody fragment
  • CD3 ⁇ T-cell signaling domains
  • TIL and TCR transfer has proven particularly good when targeting melanoma (Rosenberg et al. 2011; Morgan 2006), whereas CAR therapy has shown much promise in the treatment of certain B-cell malignancies (Grupp et al. 2013).
  • Costimulatory signals are useful to achieve robust CAR T cell expansion, function, persistence and antitumor activity.
  • CAR therapy in leukemia has been partly attributed to the incorporation of costimulatory domains (e.g. CD28 or CD137) into the CAR construct, signals from which synergize with the signal provided by CD3 ⁇ to enhance anti-tumor activity.
  • costimulatory domains e.g. CD28 or CD137
  • signal 1 provided by the TCR complex
  • costimulatory receptors such as CD28, CD137 or CD134
  • costimulatory receptors such as CD28, CD137 or CD134
  • AICD activation induced cell death
  • Targeted costimulation will have beneficial effects for non-CAR-based T-cell therapies.
  • incorporating costimulatory domains into a chimeric TCR has been shown to enhance responses of T-cells towards pMHC (Govers 2014).
  • tumor infiltrating lymphocytes TILs
  • TILs utilize their endogenous TCRs to mediate tumor recognition, it has not been possible to engineer the endogenous TCR.
  • TIL are subject to substantial limitations as tumor cells express very few costimulatory ligands.
  • the ability to induce targeted costimulation of TIL, or indeed any other adoptive T-cell therapy product, would be beneficial.
  • the invention provides novel chimeric costimulatory antigen receptors (CoStARs) and cells comprising or expressing the CoStARs which are beneficial for CAR and non-CAR based T-cell therapies alike.
  • the present invention uses cells that express a novel chimeric costimulatory receptor to provides a costimulatory signal to T-cells upon engagement with a defined disease-associated, for example tumor-associated, antigen.
  • a CoStAR of the invention induces signal 2 upon engagement with a defined antigen such as a disease associated or tumor associated antigen.
  • a defined antigen such as a disease associated or tumor associated antigen.
  • a full length CD28 molecule contains motifs critical to its native function in binding members of the B7 family of receptors; although this is potentially dangerous from the perspective of CARs carrying CD28 and CD3 ⁇ receptors in tandem, wherein ligation of CAR by B7 could trigger T-cell activation, there are beneficial qualities for receptors harboring signal 2 receptors alone.
  • the invention provides a targeted chimeric costimulatory receptor (CoStAR) which comprises an extracellular antigen binding domain operatively linked to a transmembrane domain, a first signaling domain, and a CD40 signaling domain or a signaling fragment thereof.
  • costimulatory receptors comprising a CD40 signaling domain display novel and improved activity profiles.
  • the CD40 signaling domain comprises SEQ ID NO:23, SEQ ID NO:24, or SEQ ID NO:25.
  • the CD40 signaling fragment comprises, consists, or consists essentially of an SH3 motif (KPTNKAPH (SEQ ID NO:26), PTNKAPHP (SEQ ID NO:118) or PTNKAPH(SEQ ID NO:119), TRAF2 motif (PKQE (SEQ ID NO:27), PKQET (SEQ ID NO:120), PVQE (SEQ ID NO:28), PVQET (SEQ ID NO:121), SVQE (SEQ ID NO:29), SVQET (SEQ ID NO:122)), TRAF6 motif (QEPQEINFP (SEQ ID NO:30) or QEPQEINFP (SEQ ID NO:123)), PKA motif (KKPTNKA (SEQ ID NO:31), SRISVQE (SEQ ID NO:32), or a combination thereof, or is a full length CD
  • one or more of the SH3, TRAF2, TRAF6, or PKA motifs of the CD40 signaling domain is mutated. In certain embodiments, one or more of the SH3, TRAF2, TRAF6, or PKA motifs of the CD40 signaling domain is present in multiple copies.
  • the first signaling domain of the CoStAR comprises a signaling domain or signaling fragment of a receptor, such as, for example a tumor necrosis factor receptor superfamily (TNFRSF) receptor, including but not limited to CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6.
  • TNFRSF tumor necrosis factor receptor superfamily
  • the CoStAR comprises CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6.
  • the first signaling domain comprises a CD40 signaling domain
  • the CoStAR comprises elements of two CD40 signaling domains.
  • the CoStAR comprises a second signaling domain or signaling fragment of a receptor, such as, for example a tumor necrosis factor receptor superfamily (TNFRSF) receptor, including but not limited to CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6.
  • the first signaling domain or signaling fragment, the CD40 signaling domain or signaling fragment, and the second signaling domain or signaling fragment can be in any order.
  • Exemplary embodiments include, without limitation, CoStAR which comprise CD28, CD137, and CD40 signaling domains, CD28, CD134, and CD40 signaling domains, CD28, CD2, and CD40 signaling domains, CD28, GITR, and CD40 signaling domains, CD28, CD29, and CD40 signaling domains, or CD28, CD150, and CD40 signaling domains.
  • CoStAR which comprise CD28, CD137, and CD40 signaling domains, CD28, CD134, and CD40 signaling domains, CD28, CD2, and CD40 signaling domains, CD28, GITR, and CD40 signaling domains, CD28, CD29, and CD40 signaling domains, or CD28, CD150, and CD40 signaling domains.
  • a CoStAR of the invention is engineered not to provide signal 1. Accordingly, in certain embodiments, a CoStAR of the invention does not comprise a signal 1 signaling domain. In certain embodiments, a CoStAR of the invention does not comprise a CD3 ⁇ signaling domain.
  • a CoStAR of the invention is engineered to provide signal 2 in a cell that is capable of providing signal 1 upon antigen binding (e.g., a T cell receptor provides signal 1 upon antigen engagement).
  • a CoStAR of the invention is engineered to provide signal 2 in a cell in response to antigen-specific binding by the CoStAR when the antigen is on the surface of a target cell.
  • a CoStAR of the invention is engineered not to provide signal 2 in a cell in response to antigen-specific binding by the CoStAR when the antigen is soluble and not attached to the surface of a target cell.
  • the extracellular binding domain of a CoStAR of the invention is operatively linked to the transmembrane domain by a linker and/or a spacer.
  • the linker comprises from about 5 to about 20 amino acids.
  • the linker comprises AAAGSGGSG (SEQ ID NO:8).
  • a CoStAR of the invention comprises a spacer which operatively links the extracellular binding domain to the transmembrane domain and comprises from about 10 to about 250 amino acids.
  • the spacer comprises an extracellular sequence of CD8 or CD28 or a fragment thereof.
  • the CoStAR comprises a second extracellular binding domain.
  • the second binding domain comprises an extracellular ligand binding domain from CD8 or CD28.
  • the spacer comprises one or more immunoglobulin domains or an immunoglobulin constant region.
  • the spacer comprises one or more immunoglobulin domains or an immunoglobulin constant region of SEQ ID NO:13.
  • the transmembrane domain of a CoStAR of the invention comprises a transmembrane domain of a TNFRSF protein. In certain embodiments, a transmembrane domain of a CoStAR of the invention comprises a transmembrane domain of CD28 or CD8. In certain embodiments, a transmembrane domain of a CoStAR of the invention comprises a transmembrane sequence of SEQ ID NO:11 or SEQ ID NO:12.
  • a CoStAR of the invention is useful to stimulate immune an immune response against a selected target.
  • a CoStAR of the invention comprises an extracellular binding domain that binds to a tumor associated antigen.
  • a CoStAR of the invention comprises an extracellular binding domain that binds to a tumor microenvironment associated antigen.
  • the CoStAR comprises two or more extracellular binding domains.
  • the extracellular binding domain binds to CD70, CD146, FOLR1, carcinoembryonic antigen (CEA), 5T4, mellanotransferrin (CD228), Her2, EGFR, GPC3, melanoma-associated chondroitin sulphate proteoglycan (MCSP/CSPG4), CD71, EPCAM, SM5-1, folate receptor or CA125, PDL-1, CD155 PD-1, mesothelin, or a tumor specific peptide (p)-major histocompatibility (MHC) complex, or a tumor specific pMHC complex antigen specific single chain T-cell receptor (scTCR), or transferrin, or an antibody or antigen binding protein.
  • CCA carcinoembryonic antigen
  • 5T4 mellanotransferrin
  • CD228 Her2, EGFR, GPC3, melanoma-associated chondroitin sulphate proteoglycan
  • MCSP/CSPG4 melanom
  • the CoStAR comprises SEQ ID NO:6. In certain embodiments wherein the extracellular binding domain binds to CEA, the CoStAR comprises SEQ ID NO:5. In certain embodiments wherein the extracellular binding domain binds to FOLR1, the CoStAR comprises SEQ ID NO:4. In certain embodiments wherein the binding domain binds to CD155, CD112 or CD113, the CoStAR comprises SEQ ID NO:7.
  • an extracellular binding domain can comprise, without limit, an scFv, a peptide, an antigen binding portion of an antibody, an antibody heavy-chain, a ligand of a target receptor or a ligand binding portion of a receptor.
  • a CoStAR of the invention comprises a CD3 ⁇ signaling domain, for example located at the C-terminus.
  • a CoStAR of the invention comprises an N-terminal signal peptide.
  • nucleic acid which encodes a CoStAR of the invention.
  • the nucleic acid may be optimized, for example be codon optimized for expression in a host cell.
  • nucleic acid is codon optimized for expression in a human cell.
  • vector which encodes and is capable of expressing a CoStAR of the invention.
  • a cell which expresses a CoStAR of the invention.
  • the cell expresses a CoStAR that binds to FOLR1.
  • the cell expresses a CoStAR that binds to CA125.
  • the invention provides a cell which expresses a CoStAR that is specific for FOLR1 wherein the cell is activated when the CoStAR reacts with or binds to FOLR1 on the surface of a target cell but not when the CoStAR binds to or reacts with soluble FOLR1.
  • the cell is a T cell or a TIL that expresses a T cell receptor or other receptor specific for a tumor associated antigen expressed by the target cell.
  • the cell expresses a CoStAR specific for PDL1. In other embodiments, the cell expresses a CoStAR specific for CEA. In certain embodiments, the cell expresses two or more CoStARs of the invention. In a particular embodiment, the cell expresses a CoStAR that binds to FOLR1 and a CoStAR that binds to CA125, such as but not limited to anti-FOLR1.CD28.CD40 and anti-CA125.41BB.CD40.
  • the cell expresses a CoStAR which binds to FOLR1 and a CoStAR which binds to PDL1, such as but not limited to anti-FOLR1.CD28.CD40 and PD1.CD28.CD40.
  • a cell engineered to express a CoStAR of the invention comprises an alpha-beta T cell, gamma-delta T cell, T regulatory cell, TIL, NKT cell or NK cell.
  • a cell engineered to express a CoStAR of the invention coexpresses a chimeric antigen receptor (CAR) or a T cell receptor (TCR).
  • the invention provides a method of making the cell which expresses a CoStAR which comprises transducing or transfecting a cell with a vector which encodes and is capable of expressing a CoStAR of the invention.
  • the invention provides a method for preparing a population of cells that express a CoStAR of the invention by transducing or transfecting cells, detecting expression of the CoStAR and enriching, expanding, and/or selecting cells that express the CoStAR.
  • the invention provides a method of treating a disease in a subject by administering a population of cells which express a CoStAR of the invention.
  • the invention provides a method of preparing TIL comprising disaggregating a resected tumor to obtain a refined resected tumor product, performing a first expansion by culturing the refined resected tumor product in a cell culture medium comprising IL-2 to produce a first population of TILs, performing a second expansion by culturing the first population of TILs with additional IL-2, OKT-3, and antigen presenting cells (APCs), to produce a second population of TILs; and harvesting and/or cryopreserving the second population of TILs, wherein the method comprises transfecting or transducing the TILs to express a CoStAR of the invention.
  • the tumor comprises an ovarian tumor.
  • the tumor comprises a renal tumor.
  • the tumor comprises a lung tumor.
  • FIG. 1 Structure of single costimulatory and fusion costimulatory domain receptors.
  • a schematic representation of CoStAR receptors set out in the claims is shown.
  • FIGS. 2A-2G Genetic organisation of potential CoStAR configurations—The CoStAR consists of an antigen binding domain, an optional spacer domain and a costimulatory domain as shown in figure and described in claims.
  • the CoStAR may be expressed as shown; alone from a promoter with the CoStAR consisting of a single ( FIG. 2A ) or fusion ( FIG. 2B ) costimulatory receptor; ( FIG. 2C ) may be expressed with an epitope tag (e.g. His tag, DYKDDDDK (SEQ ID NO:14) etc) at the N or C-terminus to enable direct staining of the CoStAR; ( FIG.
  • FIG. 2D along with a marker gene separated using a 2A cleavage sequence or internal ribosomal entry site (IRES);
  • FIG. 2E along with a marker gene which is expressed from a second promoter;
  • FIG. 2F along with a protein of interest such as a chimeric antigen receptor or T-cell receptor separated using a 2A cleavage sequence or internal ribosomal entry site (IRES);
  • FIG. 2G along with a protein of interest such as a chimeric antigen receptor or T-cell receptor which is expressed from a second promoter.
  • FIGS. 3A-3E Fluorescenceal activity of CoStAR in T-cells in response to LS174T and LoVo tumor presented antigen.
  • Normal donor T-cell populations from donor 1 ( FIGS. 3A & 3D ), donor 2 ( FIG. 3B ) and donor 3 ( FIGS. 3C & 3E ) were lentivirally engineered to express a CoStAR which targets carcinoembryonic antigen and magnetically sorted to enrich for the transgene using CD34 magnetic selection.
  • T-cells were mixed with wild-type un-engineered CEA+ tumor cells (Non-activating tumor) or CEA+ tumor cells engineered to express a cell surface anchored anti-CD3 single chain antibody fragment (Activating tumor) at the indicated effector to target ratios and IL-2 measured in the supernatant by ELISA.
  • FIGS. 4A-4D Effect of CoStAR on T-cell proliferation. 5 ⁇ 10 5 transduced and non transduced T-cells were mixed with 6.25 ⁇ 10 3 wild-type LoVo or LoVo-OKT3 cells in the presence ( FIG. 4A ) or absence ( FIG. 4B ) of IL-2 and cell counts made after three days. In another assay under the same cell ratios T-cells from two donors ( FIGS. 4C and 4D ) were loaded with proliferation dye and the number of proliferation cycles the cells had gone through determined by dye dilution after six days using flow cytometry.
  • FIG. 5 IL-2 activity of CoStAR fusion receptors in primary human T-cells.
  • Normal donor CD8+ T-cells from seven donors (except control CoStAR is three donors) were lentivirally transduced with the indicated CEA-targeting CoStARs and IL-2 production assessed after an overnight stimulation in the presence of LoVo-OKT3 cells.
  • the proportion of IL-2 positive cells was determined using intracellular flow staining in both the CD34 negative (CoStAR non-transduced) and CD34+ (CoStAR transduced) populations.
  • Asterisks show significant differences between the transduced and non-transduced populations using paired Wilcoxon signed rank test with * p ⁇ 0.05
  • FIGS. 6A-6D Multi parameter analysis of CoStAR activity in primary human T-cells.
  • Normal donor CD8+ T-cells were lentivirally transduced with the indicated CEA-targeting CoStARs and IL-2 production assessed after an overnight stimulation in the presence of LoVo-OKT3 cells.
  • the proportion of IL-2 (seven donors) ( FIG. 6A ), IFN ⁇ (seven donors) ( FIG. 6B ), bcl-xL (five donors) ( FIG. 6C ) and CD107a (six donors) ( FIG. 6D ) positive cells was determined using intracellular flow staining in both the CD34 negative (CoStAR non-transduced) and CD34+ (CoStAR transduced) populations.
  • Control is an irrelevant CA125 targeting CoStAR and is from three donors in all instances.
  • Heat maps are averages of all donors with the intensity of colour related to the percentage of cells positive for a particular read out under the defined conditions.
  • FIG. 7 CD40 enhances IL-2 production from CD28-based CoStARs.
  • Primary human T-cells from three healthy donors were left non-transduced or transduced with either extracellular domain truncated CD28 (Tr CD28), full length CD28 (FL CD28), or CD28.CD40-based CoStARs harboring a CEA specific scFv (MFE23). Transduced cells were selected using a CD34 marker gene and expanded prior to analysis. T-cells were mixed at an 8:1 effector to target ratio with OKT3 expressing CEA+ LoVo cells for 20 hours before analysis of IL-2 production by ELISA.
  • FIG. 8 Effect of signalling domain and target antigen on CoStAR-mediated T-cell expansion.
  • T-cells were transduced with either DYKDDDDK (SEQ ID NO:14) epitope-tagged CD28 or CD28.CD40 based CoStARs harboring CA125, FOLR1 or CEA specific scFv, or FOLR1 specific binding peptide (C7).
  • T-cells were mixed with OKT3 expressing, CA125+/FOLR1+/CEA ⁇ cell line OVCAR3. The number of transduced cells were counted every 7 days up to 21 days, with fresh OVCAR3 cells added following each count.
  • FIG. 9 CD40 based CoStARs enhance costimulation of T-cells in a model of TCR-transfer.
  • Primary human T-cells from three healthy donors were transduced with a CEA specific TCR plus either a DYKDDDK-tagged CD28 or CD28.
  • CD40 based CoStAR harboring either an MFE23 (open or closed circles) or CA125 (open squares) specific scFv.
  • T-cells were mixed at a 1:1 effector:target ratio with CEA+/CA125 ⁇ H508 cells and intracellular cytokine staining performed to determine the number of responding CD4+ or CD8+ T-cells in the TCR+/CoStAR+, TCR+/CoStAR ⁇ , TCR ⁇ /CoStAR+ and TCR ⁇ /CoStAR ⁇ populations.
  • a 2-way ANOVA (Tukeys test) was performed to determine significant differences in activity: *p>0.05, ** p>0.01, *** p>0.001, **** p>0.0001.
  • FIGS. 10A-10B CoStAR dependent enhancement of activity in a model of TCR transfer.
  • Primary human T-cells from three healthy donors were transduced with a CEA specific TCR plus either a DYKDDDK-tagged CD28 or CD28.CD40 based CoStAR harboring either an MFE23 (open or closed circles) or CA125 (open squares) specific scFv.
  • T-cells were mixed at a 1:1 effector:target ratio with CEA+/CA125 ⁇ H508 cells and intracellular cytokine staining performed to determine the number of responding CD4+( FIG. 10A ) or CD8+( FIG. 10B ) T-cells in the TCR+/CoStAR+, populations.
  • a 2-way ANOVA (Tukeys test) was performed to determine significant differences in activity: ** p>0.01, **** p>0.0001.
  • FIG. 11 depicts enrichment and expansion of primary human T-cells transduced to express costimulatory molecules of the invention.
  • MFE23 is a single chain Fv antibody that has a high affinity for carcinoembryonic antigen (CEA).
  • Primary human T-cells were mock transduced or transduced with MFE23.CD28 or MFE23.CD28.CD40 CoStAR, each harboring a CD34 marker gene separated by a 2A cleavage peptide.
  • MACSTM paramagnetic selection reagents Miltenyi Biotech
  • FIGS. 12A-12D depict expansion of T-cells transduced with costimulatory molecules of the invention in response to stimulation and exogenous IL-2.
  • Cells were mock transduced or transduced with MFE23.CD28 or MFE23.CD28.CD40 CoStAR and cocultured with LoVo-OKT3 cells at an 8:1 effector:target ratio in the presence (200 IU/ml) or absence of exogenous IL-2.
  • days 1, 4, 7, 11 and 18 cells were taken and the number of viable T-cells enumerated by using anti-CD2 reagents on a MACSQuant flow cytometer.
  • FIG. 12A In the absence of stimulation by tumor and IL-2 cells declined in number as would be expected.
  • FIG. 12A In the absence of stimulation by tumor and IL-2 cells declined in number as would be expected.
  • FIG. 12B In the absence of stimulation but presence of IL-2 there was a more apparent survival of the cells, but no specific growth.
  • FIG. 12C In the presence of tumor, but absence of IL-2 mock cells did not show specific survival.
  • MFE23.CD28 CoStAR mediated an apparent doubling in expansion over the first four days followed by decline.
  • MFE23.CD28.CD40 mediated a greater expansion up to day 7 followed by a steady decline.
  • FIG. 12D Under the same conditions but in the presence of IL-2 both mock and MFE23.CD28 transduced cells demonstrated a 20-fold expansion over 18 days, whereas MFE23.CD28.CD40 cells expanded by over 60-fold.
  • CD28.CD40 based receptors demonstrate superior expansion and survival under conditions of stimulation both in the presence and absence of exogenous IL-2.
  • FIGS. 13A-13M depict cytokine production by mock, MFE23.CD28 or MFE23.CD28.CD40 engineered T-cells.
  • Bead array analysis was performed on supernatants obtained from T-cell/tumor cocultures.
  • Engineered T-cells were incubated at a 1:1 effector:target ratio with LoVo-OKT3 cells for 24 hours and supernatant collected.
  • Conditioned supernatant was also collected from an equal number of T-cells alone, or LoVo-OKT3 cells alone. Cytokine production was analysed using a LegendplexTM Human TH1/TH2 cytokine panel (Biolegend).
  • FIG. 13A IL-2
  • FIG. 13B IFN- ⁇
  • FIG. 13A IL-2
  • FIG. 13B IFN- ⁇
  • FIG. 13C TNF ⁇ ;
  • FIG. 13D IL-4;
  • FIG. 13E IL-5;
  • FIG. 13F IL-13;
  • FIG. 13G IL-17A;
  • FIG. 13H IL-17F;
  • FIG. 13I IL-22;
  • FIG. 13J IL-6;
  • FIG. 13K IL-10;
  • FIG. 13L IL-9;
  • FIG. 13M IL-21.
  • Cytokines were either very low or undetectable in media from T-cells or tumor alone. When cocultured with tumor, cytokine production was enhanced.
  • MFE23.CD28 enhanced production of IL-2, IL-5, IL-17A/17F, IL-10, IL-9 and IL-21 compared to mock.
  • MFE23.CD28.CD40 also enhanced production of TNF ⁇ , IL-13 and IL-22.
  • MFE23.CD28.CD40 and further enhanced the production of a number of cytokines greater than that provided by MFE23.CD28 (IL-2, IL-9 and IL-17F), as well as reducing the production of some cytokines below the levels seen with MFE23.CD28 (IL-5 and IL-10).
  • MFE23.CD28 enhanced production of IL-2, IL-5, IL-17A/17F, IL-10, IL-9 and IL-21 compared to mock.
  • MFE23.CD28.CD40 also enhanced production of TNF ⁇ , IL-13 and IL-22.
  • MFE23.CD28.CD40 and further enhanced the production of a number of cytokines greater than that provided by MFE23.CD
  • FIGS. 14A-14M depict an analysis of chemokines using a LegendplexTM Human Pro inflammatory chemokine panel.
  • FIG. 14A IL-8 (CXCL8);
  • FIG. 14B IP-10 (CSCL10);
  • FIG. 14C Eotaxin (CCL11);
  • FIG. 14D TARC (CCL17);
  • FIG. 14E MCP-1 (CCL2);
  • FIG. 14F RANTES (CCL5);
  • FIG. 14G MIP-1a (CCL3)
  • FIG. 14H MIG (CXCL9)
  • FIG. 14I ENA-78 (CXCL5)
  • FIG. 14J MIP-3a (CCL20) ( FIG.
  • CXCL1 GRO ⁇ (CXCL1) ( FIG. 14L ) I-TAC (CXCL11) ( FIG. 14M ) MEP-1 ⁇ (CCL4).
  • Chemokines were either very low or undetectable in media from T-cells alone. When cocultured with tumor, chemokine production was enhanced.
  • MFE23.CD28 enhanced production of CXCL5, CXCL10, CXCL11, CCL17 and CCL20 compared to mock.
  • MFE23.CD28.CD40 also enhanced production of CCL2, CXCL1 and CXCL9.
  • MFE23.CD28.CD40 further enhanced the production of a number of cytokines greater than that provided by MFE23.CD28 (CXCL1, CXCL9, CXCL10, CXCL11, CCL17, CCL2, CXCL9, CCL5 and CCL20), as well as reducing the production of some cytokines below the levels seen with MFE23.CD28 (CCL4).
  • FIGS. 15A-15H depict functional activity of ovarian CoStAR engineered cells using a CoStAR harboring a FolR or CA125 reactive scFv (MOV19 & 196-14 respectively).
  • Human folate receptor alpha represents a suitable target for a number of tumors including ovarian, head and neck, renal and lung and CA125 represents an alternative target for ovarian cancer.
  • Primary human T-cells from six healthy donors were engineered with either 196-14.CD28, 196-14.CD28.CD40, MOV19.CD28 or MOV19.CD28.CD40 receptors, all harboring a DYKDDDDK epitope tag for detection.
  • Transduced cells were mixed with FolR+/CA125+ OvCAR-OKT3 cells before analysis of effector activity using intracellular staining in the epitope tag positive and negative populations.
  • Specific enhancement of effector activity determined by production of IL-2 ( 15 A and 15 B), TNF ⁇ ( 15 C and 15 D), CD137 ( 15 E and 15 F), and BCL-xL ( 15 G and 15 H) was observed in CD28 and CD28.CD40 engineered cells in response to both CA125 and FolR, except for specific BCL-xL induction by MOV19.CD28 which was not observed compared to MOV19.CD28.CD40.
  • FIGS. 16A-16F depict three TIL populations mock transduced or engineered with MOV19.CD28.CD40 CoStAR and then mixed with patient matched tumor digest.
  • the donor tumors displayed varying levels of FolR on the digest, ranging from negative ( FIG. 16A ), low expression ( FIG. 16B ) to high expression ( FIG. 16C ).
  • Mock and CoStAR negative TIL in the CoStAR engineered populations of TIL matched for the FolR negative digest demonstrated similar levels of CD137 upregulation following tumor coculture which was not enhanced by the presence of CoStAR ( FIG. 16D ).
  • FIGS. 17A-17C depict enhancement of effector functions.
  • a FolR targeting CoStAR enhanced CD137 expression from ⁇ 20% to ⁇ 50% ( FIG. 17A ), TNF ⁇ production from 10% to 15% ( FIG. 17B ) and IL-2 production from 2% to 5%. ( FIG. 17C ) in response to FolR+ tumor digest.
  • FIGS. 18A-18F depict soluble ligand does not inhibit effector functions.
  • T-cells from three healthy donors were engineered with MOV19.CD28 or MOV19.CD28.CD40 CoStAR and activated with either immobilised OKT3, providing stimulation in the absence of FolR, or with OvCAR-OKT3, to provide TCR and CoStAR activity.
  • Bcl-XL activity was increased from between 10 and 20% across the three donors following OKT3 stimulation ( FIG. 18A ) whereas IL-2 was increased between 0 and 12% ( FIG. 18B ) and TNF ⁇ increased between 0 and 20% ( FIG. 18C ).
  • the presence of exogenous soluble FolR did not enhance any of these particular effector functions.
  • FIG. 19 depicts exemplary CoStAR constructs.
  • MFE23 scFv specific for carcinoembryonic antigen (CEA). Costimulatory domains are identified.
  • CTP188 SEQ ID NO:89; CTP189: SEQ ID NO:109; CTP190: SEQ ID NO:41; CTP191: SEQ ID NO:45; CTP192: SEQ ID NO:43; CTP193: SEQ ID NO:42; CTP194: SEQ ID NO: 33;
  • CTP196 SEQ ID NO:111; CTP197 SEQ ID NO:112;
  • CTP198 SEQ ID NO:113;
  • CTP199 SEQ ID NO:114;
  • CTP200 SEQ ID NO:115; CTP201: SEQ ID NO:116; CTP202: SEQ ID NO:117; CTP203: SEQ ID NO:49; CTP204: SEQ ID NO:59.
  • FIG. 20 depicts CD4+ and CD8+ subpopulations of CD40 CoStAR modified T cells.
  • T cells of four healthy donors were activated and transduced with various CD40 CoStARs with a CD34 marker or mock transduced. Cells were enriched for their CD34 expression and expanded following the rapid expansion protocol (REP).
  • FIGS. 21A-21C depict increased amount of IL-2 in PD-1 fusion CD40 CoStAR compared to mock transduced T cells.
  • Donor cells activated with Dynabeads and transduced with CTP188, CTP189, CTP194 ( FIG. 21A ) or mock-transduced were enriched for CD34 (transduction marker) expression ( FIG. 21B ), expanded following the rapid expansion protocol (REP) and frozen for subsequent experiments. After thaw, cells were rested for 3-4 days in complete RPMI supplemented with IL-2. The viability and absolute count were assessed after overnight IL-2 starvation using DRAQ-7 (1:200) by flow cytometry (Novocyte) and data were analysed using the NovoExpress 1.5.0 software.
  • Transduced T cells were cocultured in absence of IL-2 with LoVo (CCL-229TM from ATCC) or LoVo.OKT3.GFP tumor cells at 8:1 effector to target ratio. After 24 hours, supernatants were collected and frozen. LoVo and LoVo.OKT3.GFP naturally express CEA and PD-L1 on their surface, conferring signal 2 through the CoStAR alone (LoVo) or associated with signal 1 (LoVo.OKT3.GFP) to the transduced T cells. Cocultures were performed in triplicates and corresponding negative (T cells alone, tumor cells alone) and positive (PMA+ionomycin) controls were included in the experiment.
  • FIG. 22 depicts PD-1 extracellular domain conferring a slight proliferation advantage to CD40 CoStAR transduced T cells when cocultured with LoVo.OKT3.
  • Healthy donor T cells activated with Dynabeads and transduced with CTP188, CTP189, CTP194 or mock-transduced were enriched for CD34 expression, expanded following the rapid expansion protocol (REP) and frozen for subsequent experiments. After thaw, cells were rested for 3-4 days in complete RPMI supplemented with IL-2 and their transduction rate was determined looking at the CD34 marker gene expression.
  • FIGS. 23A-23C depict exhaustion profiles of PD-1 fusion CD40 CoStAR transduced T cells after tumor challenge.
  • Healthy donor T cells were activated with Dynabeads and transduced with CTP188, CTP189, CTP194 or mock transduced.
  • Cells were enriched for CD34 marker expression, expanded following the rapid expansion protocol (REP) and frozen for subsequent experiments. After thaw, cells were rested for 3-4 days in complete RPMI supplemented with IL-2. The viability and absolute count were assessed after overnight IL-2 starvation using DRAQ-7 (1:200) by flow cytometry (Novocyte) and data were analysed using the NovoExpress 1.5.0 software.
  • Transduced T cells were cocultured in absence of IL-2 for 6-8 days with LoVo.OKT3.GFP tumor cells at 8:1 effector to target ratio, changing half of the culture medium every 3-4 days.
  • LoVo.OKT3.GFP naturally expresses CEA and PD-L1 on their surface, conferring both signal 2 and signal 1 (OKT3) to the transduced T cells.
  • the viability and the absolute count were assessed, and live T cells were rechallenged for an additional week with fresh LoVo.OKT3.GFP tumor cells as described above.
  • Exhaustion profiles LAG-3 ( FIG. 23A ), PD-1 ( FIG. 23B ), TIM-3 ( FIG.
  • FIGS. 24A-24B depict T cells transduced with CD28, CD137 and CD40 alone CoStARs secrete higher amount of IL-2 following activation compared to mock transduced T cells.
  • FIG. 24A Healthy donor T cells were activated with Dynabeads and transduced with CTP190, CTP191, CTP192, CTP193, CTP194 or mock transduced.
  • REP rapid expansion protocol
  • FIG. 24B The viability and absolute count were assessed after overnight IL-2 starvation using DRAQ-7 (1:200) by flow cytometry (Novocyte) and data were analysed using the NovoExpress 1.5.0 software.
  • Transduced T cells were cocultured in absence of IL-2 with LoVo (CCL229TM from ATCC) or LoVo.OKT3.GFP tumor cells at 8:1 effector to target ratio. After 24 hours, supernatants were collected and frozen.
  • LoVo and LoVo.OKT3.GFP naturally express CEA and PD-L1 on their surface, conferring signal 2 through the CoStAR alone (LoVo) or associated with signal 1 (LoVo.OKT3.GFP) to the transduced T cells.
  • Cocultures were performed in triplicates and corresponding negative (T cells alone, tumor cells alone) and positive (PMA+ionomycin) controls were included in the experiment. After thaw, secreted IL-2 and IFN- ⁇ were detected by ELISA and the absorbance was measured using the FLUOstar Omega microplate reader and subsequently analysed with the Omega MARS 3.42 R5 software. Each dot represents the mean of triplicates for one donor. Note that negative controls (T cells alone, tumor cells alone) were all below the detection range (#)( FIG. 24B ).
  • FIG. 25 depicts CD28 and CD137 endodomains conferring a proliferation advantage to CD40 CoStAR transduced T cells when cocultured with LoVo.OKT3.
  • Healthy donor T cells were activated with Dynabeads and transduced with CTP190, CTP191, CTP192, CTP193, CTP194 or mock transduced.
  • Cells were enriched for CD34 marker expression, expanded following the rapid expansion protocol (REP) and frozen for subsequent experiments. After thaw, cells were rested for 3-4 days in complete RPMI supplemented with IL-2. The viability and absolute count were assessed after overnight IL-2 starvation using DRAQ-7 (1:200) by flow cytometry (Novocyte) and data were analysed using the NovoExpress 1.5.0 software.
  • Transduced T cells were cocultured in absence of IL-2 for 6-8 days with LoVo.OKT3.GFP tumor cells at 8:1 effector to target ratio, changing half of the culture medium every 3-4 days.
  • LoVo.OKT3.GFP naturally expresses CEA and PD-L1 on their surface, conferring both signal 2 and signal 1 (OKT3) to the transduced T cells.
  • the viability and absolute count were assessed, and live T cells were rechallenged for an additional week with fresh LoVo.OKT3.GFP tumor cells as described above.
  • the viability and absolute count were measured, and the fold expansion was calculated. Data shown as mean+/ ⁇ SEM of n ⁇ 3 donors analysed in triplicates.
  • FIGS. 26A-26C depict exhaustion profiles of transduced T cells with CD28, CD2, CD137 and CD40 alone CoStARs after tumor challenge.
  • Healthy donor T cells were activated with Dynabeads and transduced with CTP190, CTP191, CTP192, CTP193, CTP194 or mock transduced.
  • Cells were enriched for CD34 marker expression, expanded following the rapid expansion protocol (REP) and frozen for subsequent experiments. After thaw, cells were rested for 3-4 days in complete RPMI supplemented with IL-2. The viability and absolute count were assessed after overnight IL-2 starvation using DRAQ-7 (1:200) by flow cytometry (Novocyte) and data were analysed using the NovoExpress 1.5.0 software.
  • Transduced T cells were cocultured in absence of IL-2 for 6-8 days with LoVo.OKT3.GFP tumor cells at 8:1 effector to target ratio, changing half of the culture medium every 3-4 days.
  • LoVo.OKT3.GFP naturally expresses CEA and PD-L1 on their surface, conferring both signal 2 and signal 1 (OKT3) to the transduced T cells.
  • the viability and the absolute count were assessed, and live T cells were rechallenged for an additional week with fresh LoVo.OKT3.GFP tumor cells as described above.
  • Exhaustion profiles LAG-3 ( FIG. 26A ), PD-1 ( FIG. 26B ), TIM-3 ( FIG.
  • FIGS. 27A-27B depict CD40 CoStAR TRAF-binding site mutations have a direct impact on the secretion of IL-2 and IFN- ⁇ following activation.
  • FIG. 27A Cells of three donors were activated with Dynabeads and transduced with CTP194, CTP195, CTP196, CTP197, CTP198, CTP199, CTP200 or mock transduced. Cells were enriched for CD34 marker expression, expanded following the rapid expansion protocol (REP) and frozen for subsequent experiments. After thaw, cells were rested for 3-4 days in complete RPMI supplemented with IL-2 and their transduction rate was determined looking at the CD34 marker gene expression (A, lower panel).
  • FIG. 27A Cells of three donors were activated with Dynabeads and transduced with CTP194, CTP195, CTP196, CTP197, CTP198, CTP199, CTP200 or mock transduced. Cells were enriched for CD34 marker expression, expanded following the rapid expansion protocol (
  • FIG. 28 depicts the critical role of the PVQET TRAF-binding motif in long term survival and proliferation of CD28.CD40 CoStAR transduced T cells when cocultured with LoVo.OKT3.
  • Cells of three donors were activated with Dynabeads and transduced with CTP194, CTP195, CTP196, CTP197, CTP198, CTP199, CTP200 or mock transduced.
  • Cells were enriched for CD34 marker expression, expanded following the rapid expansion protocol (REP) and frozen for subsequent experiments. After thaw, cells were rested for 3-4 days in complete RPMI supplemented with IL-2.
  • REP rapid expansion protocol
  • FIGS. 29A-29C depict exhaustion profiles of transduced T cells with CD28.CD40 mutants CoStAR constructs after tumor challenge.
  • Cells of three donors were activated with Dynabeads and transduced with CTP194, CTP195, CTP196, CTP197, CTP198, CTP199, CTP200 or mock transduced.
  • Cells were enriched for CD34 marker expression, expanded following the rapid expansion protocol (REP) and frozen for subsequent experiments. After thaw, cells were rested for 3-4 days in complete RPMI supplemented with IL-2.
  • REP rapid expansion protocol
  • FIGS. 30A-30B depict CD28 mutants and IgG4 CD40 CoStAR transduced T cells secreting higher amount of IL-2 and IFN- ⁇ following activation compared to mock transduced T cells.
  • FIG. 30A Cells of three donors were activated with Dynabeads and transduced with CTP194, CTP201, CTP202, CTP203 or mock transduced. Cells were enriched for CD34 marker expression, expanded following the rapid expansion protocol (REP) and frozen for subsequent experiments. After thaw, cells were rested for 3-4 days in complete RPMI supplemented with IL-2 and their transduction rate was determined looking at the CD34 marker gene expression (A, lower panel).
  • FIG. 30A Cells of three donors were activated with Dynabeads and transduced with CTP194, CTP201, CTP202, CTP203 or mock transduced. Cells were enriched for CD34 marker expression, expanded following the rapid expansion protocol (REP) and frozen for subsequent experiments. After thaw, cells
  • FIG. 31 depicts the critical role of CD28 PYAP and YMNM motifs in the long term survival and proliferation of CD40 CoStAR transduced T cells when cocultured with LoVo.OKT3.
  • Cells of three donors were activated with Dynabeads and transduced with CTP194, CTP201, CTP202, CTP203 or mock transduced.
  • Cells were enriched for CD34 marker expression, expanded following the rapid expansion protocol (REP) and frozen for subsequent experiments. After thaw, cells were rested for 3-4 days in complete RPMI supplemented with IL-2.
  • REP rapid expansion protocol
  • FIGS. 32A-32C depict exhaustion profiles of transduced T cells with CD28 mutant CD40 CoStAR constructs after tumor challenge.
  • Cells of three donors were activated with Dynabeads and transduced (spinoculation, MOI 5) with CTP194, CTP201, CTP202, CTP203 or mock transduced.
  • Cells were enriched for CD34 marker expression, expanded following the rapid expansion protocol (REP) and frozen for subsequent experiments. After thaw, cells were rested for 3-4 days in complete RPMI supplemented with IL-2. The viability and absolute count were assessed after overnight IL-2 starvation using DRAQ-7 (1:200) by flow cytometry (Novocyte) and data were analysed using the NovoExpress 1.5.0 software.
  • Transduced T cells were cocultured in absence of IL-2 for 6-8 days with LoVo.OKT3.GFP tumor cells at 8:1 effector to target ratio, changing half of the culture medium every 3-4 days.
  • LoVo.OKT3.GFP naturally expresses CEA and PD-L1 on their surface, conferring both signal 2 and signal 1 (OKT3) to the transduced T cells.
  • the viability and the absolute count were assessed, and live T cells were rechallenged for an additional week with fresh LoVo.OKT3.GFP tumor cells as described above.
  • Exhaustion profiles LAG-3 ( FIG. 32A ), PD-1 ( FIG. 32B ), TIM-3 ( FIG.
  • FIG. 33 depicts generation of transduced T cells from four healthy donors following CD34 enrichment and expansion.
  • T cells of 4 healthy donors (NBC360, NBC362, NBC358, NBC361) were activated with Dynabeads and transduced with CTP188, CTP189, CTP190, CTP191, CTP192, CTP193, CTP194, CTP195, CTP196, CTP197, CTP198, CTP199, CTP200, CTP201, CTP202, CTP203, CTP204 or mock transduced.
  • Cells were then magnetically enriched for their CD34 expression and expanded following the rapid expansion protocol (REP). Viability of each sample 10-11 days after REP was assessed by flow cytometry (Novocyte). Data were analysed with NovoExpress 1.5.0 software. Each dot within the same donor represents a different construct.
  • FIGS. 34A-34D depict expression of activation markers and cytokine production of non-transduced (NTD) or anti-FOLR1 CoStAR modified T cells (CoStAR) from 3 healthy donors co-cultured overnight with Ba/F3 targets.
  • CoStAR engagement enhances cytokine secretion.
  • FIG. 34C Non-transduced and CoStAR cytotoxicity is comparable. Tumor counts of Ba/F3 targets were assessed by flow cytometry after overnight coculture with non-transduced (NTD) and CoStAR T cells.
  • FIG. 34D CoStAR engagement enhances both CD4 and CD8 T cell proliferation. NTD and CoStAR T cell counts as well as proliferation were assessed by flow cytometry after overnight or 5-day coculture with Ba/F3 targets.
  • FIGS. 35A-35D depicts expression of activation markers of anti-FOLR1 CoStAR modified T cells (CoStAR) from 3 healthy donors preincubated with soluble folate receptor (sFOLR1) followed by co-culture overnight with Ba/F3 targets.
  • X-axis shows sFOLR1 as ng/mL.
  • bars 1 ⁇ 4 are non-transduced
  • bars 5-8 are CoStAR transduced.
  • Soluble FOLR1 does not impact upregulation of activation markers on CoStAR T cells.
  • Expression of activation markers 4-1BB ( FIG. 35A ) and CD69 ( FIG. 35B ) was determined.
  • FIG. 35C sFOLR1 does not impact cytoxicity of CoStAR T cells.
  • sFOLR1 does not impact cytokine secretion. IL-2 production is shown in anti-FOLR1 CoStAR modified T cells preincubated with soluble folate receptor (sFOLR) followed by co-culture overnight with Ba/F3 targets.
  • FIGS. 36A-36D depict FOLR1 CoStAR requires signal 1 to function.
  • FIGS. 36A and 36B Expression of activation markers ( FIG. 36A ) and cytokine production ( FIG. 36B ) respectively, from non-transduced (NTD) and anti-FOLR1 CoStAR modified T cells (CoStAR) from 3 healthy donors co-cultured overnight with Ba/F3 targets.
  • FIG. 36A There is minimal upregulation of activation markers with signal 2 only.
  • FIG. 36B No cytokine secretion was observed with signal 2 only.
  • FIG. 36C Tumor counts of Ba/F3 targets assessed by flow cytometry after overnight coculture with NTD and CoStAR T cells. No cytoxicity was observed with signal 2 only.
  • FIG. 36D NTD and CoStAR T cell counts were assessed by flow cytometry after overnight or 5-day coculture with Ba/F3 targets. Proliferation was not observed with signal 2 only.
  • FIGS. 37A-37C depict TIL function with CoStAR in ovarian cancer.
  • FIG. 37A TIL from 6 ovarian tumors were liberated by digestion and cultured in 3000 U IL-2. Transduction with a 3 rd generation lentiviral vector encoding a CoStAR molecule with and scFv targeting human FOLR1, linker, full length CD28 fused to truncated CD40 cytoplasmic domain was carried out at an MOI of 5, both 48 h and 72 h after tumor digestion. Rapid expansion protocol was carried out on days 12-23. ( FIG.
  • FIG. 37B Flow cytometric analysis was used to determine the frequency of CD4 and CD8 T-cells expressing the CoStAR molecule using an anti-idiotype antibody for surface detection.
  • FIG. 37C Flow cytometric analysis was used to determine the frequency of cells expressing TCR ⁇ and TCR ⁇ by flow cytometric surface staining. Mock—untransduced cells.
  • CoStAR ⁇ /+ cells negative or positive for CoStAR molecule in the treated cell population as determined by flow cytometry gating.
  • FIGS. 38A-38B depict TIL function with CoStAR in ovarian cancer.
  • FIG. 38A CoStAR modified TIL from 5 ovarian tumors were co-cultured with autologous digest overnight in the presence of brefeldin A. The frequency of cells expressing IL-2 or TNF ⁇ was assessed the following day by flow cytometry. The frequency of TIL reacting to autologous digest is enhanced by the CoStAR molecule. NTD: untransduced cells.
  • CoStAR ⁇ /+ cells negative or positive for CoStAR molecule in the treated cell population as determined by flow cytometry gating.
  • CoStAR modified TIL from 5 ovarian tumors were co-cultured with autologous digest and supernatant assessed for cytokine release.
  • CoStAR modified cells had increased effector functions as demonstrated by increased IFN ⁇ , TNF ⁇ and IL-13 release. Maximal levels of these molecules was similar in response to stimulation with PMA (Phorbol 12-myristate 13-acetate) and ionomycin.
  • FIGS. 39A-39C depict CoStAR TIL retain robust effector functions and retain a requirement for signal 1 and 2.
  • FIG. 39A CoStAR modified TIL from 5 ovarian tumors were co-cultured with BA/F3 cells or BA/F3 cells engineered to express OKT3, FOLR1 or both. Cytokine secretion of non-modified and CoStAR modified TIL was equivalent when co-cultured with non-modified BA/F3 or BA/F3 expressing OKT3 alone or FOLR1 alone. CoStAR modified TIL secreted increased levels of cytokines when co-cultured with BA/F3 modified to express both FOLR1 and OKT3. ( FIG. 39A)
  • FIG. 39B CoStAR modified TIL from 5 ovarian tumors were co-cultured with BA/F3 cells or BA/F3 cells engineered to express OKT3, FOLR1 or both. Cytotoxicity towards BA/F3 target cells was assessed via cell counts, determined by flow cytometric analysis of mouse CD45. Non-modified and CoStAR modified cells kill cells expressing OKT3 equivalently. CoStAR modified TILs do not kill BA/F3 cells expressing FOLR1 alone.
  • FIG. 39C Mock or CoStAR modified TIL from 3 ovarian cancer patients were co-cultured with autologous tumor in the presence of no blocking, MHCI, MHC II or MHC I+MHC II blocking or antibodies or isotype control.
  • costimulatory receptors comprising a CD40 signaling domain display novel and improved activity profiles.
  • the activity profiles can be modulated by selecting an intracellular domain of a receptor protein for joining to the CD40 signaling domain and/or by selecting elements of the CD40 signaling domains to join to the intracellular domain of a receptor protein.
  • recombinant costimulatory antigen receptors comprising: (i) a disease- or tumor-associated antigen binding domain, (ii) a first intracellular segment comprising an intracellular signaling domain of a receptor protein, and (iii) a second intracellular signaling domain of a CD40 receptor protein or signal transducing fragment thereof.
  • the CoStAR comprises an extracellular segment of a stimulatory receptor protein.
  • the extracellular segment of the stimulatory receptor protein is capable of binding ligand.
  • the extracellular segment of a stimulatory receptor protein is truncated and does not bind ligand.
  • the extracellular segment of the stimulatory receptor protein operates as an adjustable length spacer allowing the disease- or tumor-associated antigen binding domain to be located away from the surface of the cell in which it is expressed for example to form a more optimal immune synapse.
  • the extracellular segment of a stimulatory receptor protein and the first intracellular segment comprise segments of the same receptor protein.
  • the extracellular segment and the first intracellular segment comprise segments of different receptor proteins.
  • the CoStARs comprise an intervening transmembrane domain between the disease or tumor antigen binding domain and the first intracellular domain.
  • the transmembrane domain is intervening between the extracellular segment and the first intracellular signaling domain.
  • full length protein or “full length receptor” refers to a receptor protein, such as, for example, a CD28 receptor protein.
  • full length encompasses receptor proteins lacking up to about 5 or up to 10 amino acids, for example 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids, at the N-terminal of the mature receptor protein once its signal peptide has been cleaved. For instance, while a specific cleavage site of a receptors N-terminal signal peptide may be defined, variability in exact point of cleavage has been observed.
  • the term “full length” does not imply presence or absence of amino acids of the receptors N-terminal signal peptide. In one embodiment, the term “full length” (e.g.
  • a full length CD28 or a full length CD40 intracellular domain encompasses mature receptor proteins (e.g. CD28 according to certain aspects of the invention) lacking the N terminal signal peptide lacking up to about 5, for example 1, 2, 3, 4, 5, or up to 10 amino acids at the N-terminal of the mature receptor protein once its signal peptide has been cleaved.
  • a “full length” CD28 receptor or other receptor or tumor antigen binding domain does not include the signal peptide and may lack up to about 5, for example 1, 2, 3, 4, 5, or up to 10 amino acids at the N-terminal of the mature receptor protein (e.g. N terminal residues N, K, I, L and/or V).
  • SEQ ID Nos. 4-12 This is shown in the exemplary fusions, e.g. SEQ ID Nos. 4-12 (note that these may lack up to about 5, for example 1, 2, 3, 4, 5, or up to 10 amino acids at the N-terminal of the mature receptor protein as shown in the boxed region).
  • CoStARs have modular form and can be constructed to comprise extracellular, transmembrane and intracellular domains obtained from a one or more proteins, along with the scFv obtained from an antibody that binds to a disease-associated antigen, for example, a tumor associated antigen.
  • a CoStAR comprises a disease-associated, for example a tumor-associated, antigen receptor, such as but not limited to a tumor-associated antigen specific scFv, and a primary costimulatory receptor protein that is capable of binding to its cognate ligand and providing an intracellular signal.
  • the primary costimulatory receptor can be less than a full length protein but is sufficient to bind cognate ligand and transduce a signal.
  • the primary costimulatory receptor domain is full length, such as but not limited to, full length CD28.
  • CoStAR constructs that comprise an antigen binding domain, an optional spacer, an optional costimulatory receptor protein comprising an extracellular ligand binding segment or fragment thereof and intracellular CD40 signaling domain.
  • a CoStAR comprises an antigen binding domain, an optional spacer, an extracellular ligand-binding portion of a costimulatory receptor protein, a transmembrane domain, and an intracellular signaling domain of a selected costimulatory receptor protein and intracellular CD40 signaling domain.
  • the extracellular ligand-binding portion comprises a CD28 truncation, for example, a C-terminal CD28 truncation after amino acids IEV, and is followed by an intracellular signaling domain.
  • the intracellular signaling domain is from CD40.
  • the transmembrane domain separating the extracellular ligand-binding and intracellular signaling domains can be from, with limitation, CD28, CD40.
  • CoStARs can comprise additional costimulatory domains, for example a third, intracellular costimulatory signaling domain and in this respect may be similar to certain chimeric antigen receptors (CARs), which have been classified into first (CD3 ⁇ only), second (one costimulatory domain+CD3 ⁇ ), or third generation (more than one costimulatory domain+CD3 ⁇ ).
  • CARs chimeric antigen receptors
  • Costimulatory receptor proteins useful in CoStARs of the invention include, without limitation, CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6, which in their natural form comprise extracellular ligand binding domains and intracellular signal transducing domains.
  • CD2 is characterized as a cell adhesion molecule found on the surface of T cells and is capable of initiating intracellular signals necessary for T cell activation.
  • CD27 is characterized as a type II transmembrane glycoprotein belonging to the TNFR superfamily (TNFRSF) whose expression on B cells is induced by antigen-receptor activation in B cells.
  • CD28 is one of the proteins on T cells and is the receptor for CD80 (B7.1) and CD86 (B7.2) ligands on antigen-presenting cells.
  • CD137 (4-1BB) ligand is found on most leukocytes and on some non-immune cells.
  • OX40 ligand is expressed on many antigen-presenting cells such as DC2s (dendritic cells), macrophages, and B lymphocytes.
  • the costimulatory receptor protein is full length CD28 as defined herein.
  • CD40 is a member of the tumor necrosis factor receptor (TNFR) superfamily and several isoforms are generated by alternative splicing. Its ligand, CD154 (also called CD40L) is a protein that is primarily expressed on activated T cells.
  • CD40 isoform 1 protein sequence is set forth in GenBank accession No. NP_001241.1, including signal peptide (amino acids 1-20), transmembrane domain (amino acids 194-215), and cytoplasmic domain (amino acids 216-277)(SEQ ID NO:22).
  • CD40 receptor signaling involves adaptor proteins including but not limited to TNF receptor-associated factors (TRAF), and the CD40 cytoplasmic domain comprises signaling components, including amino acid sequences fitting an SH3 motif (KPTNKAPH or PTNKAPHP or PTNKAPH), TRAF2 motif (PKQE, PKQET, PVQE, PVQET, SVQE, SVQET), TRAF6 motif (QEPQEINF or QEPQEINFP) and PKA motif (KKPTNKA, SRISVQE).
  • the invention further includes engineered signaling domains, such as engineered CD40 signaling domains, comprising TRAF-binding amino acid sequences.
  • Engineered signaling domains that bind to TRAF1, TRAF2, TRAF3, and TRAF5 may comprise the major consensus sequence (P/S/A/T)X(Q/E)E or minor consensus sequence PXQXXD and can be identified in or obtained from, without limitation, TNFR family members such as CD30, Ox40, 4-1BB, and the EBV oncoprotein LMP1.
  • TNFR family members such as CD30, Ox40, 4-1BB
  • EBV oncoprotein LMP1 EBV oncoprotein LMP1.
  • Examples disclosed herein demonstrate operation of CD40 as a costimulatory signaling domain in a CoStAR and further that cytokine and chemokine expression profiles are altered by signaling domain selection.
  • the costimulatory CD40 signaling domain of a CoStAR promotes pro-inflammatory cytokines (e.g., IL-2, TNF ⁇ ).
  • the costimulatory CD40 signaling domain of a CoStAR reduces immunosuppressive cytokines (e.g., IL-5, IL-10).
  • Costimulatory activity of a CD40 signaling domain or fragment can be observed in combination with a first receptor signaling domain such as but not limited to CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6, as compared to activity of the first receptor signaling domain without the CD40 signaling domain or fragment.
  • a first receptor signaling domain such as but not limited to CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357
  • the CD40 signaling domains of the invention including signaling fragments comprising particular factor binding sites or wherein particular factor binding sites are mutated, in combination with a costimulatory first signaling domain, are capable of promoting or suppressing relative expression of particular cytokines and/or chemokines as compared to the first signaling domain alone.
  • activity of a costimulatory signaling domain See, e.g., Ahonen, C L et al., The CD 40- TRAF 6 axis controls affinity maturation and the generation of long - lived plasma cells . Nat Immunol.
  • a CoStAR of the invention comprises substantially all of a CD40 costimulatory domain. In certain embodiments, a CoStAR of the invention comprises two or more CD40 costimulatory domains. In certain embodiments, a CoStAR of the invention comprises a CD40 costimulatory domain signaling component or motif, including but not limited to an SH3 motif (KPTNKAPH), TRAF2 motif (PKQE, PVQE, SVQE), TRAF3 motif, TRAF6 motif (QEPQEINFP) or PKA motif (KKPTNKA, SRISVQE) as well as two or more, or three or more, or four or more such components of motifs, which can be in multiple copies and arranged in any order.
  • KPTNKAPH SH3 motif
  • PQE TRAF2 motif
  • PVQE PVQE
  • SVQE TRAF3 motif
  • TRAF6 motif QEPQEINFP
  • PKA motif KKPTNKA, SRISVQE
  • a CoStAR of the invention comprises a CD40 costimulatory domain and a CD40 costimulatory domain signaling component or motif.
  • the SH3 motif, TRAF2 motif, and TRAF6 motif are sufficient to modulate pro-inflammatory and/or immunosuppressive cytokines.
  • adding tandem copies of those motifs and/or mutating certain motifs amplifies these effects.
  • selection of one or more costimulatory domain signaling component or motif is guided by the cell in which the CoStAR is to be expressed and/or a desired costimulatory activity more closely identified with a signaling component or motif, or avoidance of a costimulatory activity more closely identified with a signaling component or motif.
  • a CoStAR signaling domain comprises, in addition to a CD40 costimulatory domain or signaling component or motif thereof, or two or more such domains or components or motifs or combinations thereof, an additional full length costimulatory domain or signaling component thereof from, without limitation, CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6,
  • the human CD28 protein sequence is set forth in GenBank accession No. NP_006130.1, including signal peptide (amino acids 1-18), extracellular domain (amino acids 19-152), transmembrane domain (amino acids 153-179) and cytoplasmic domain (amino acids 180-200).
  • the extracellular domain includes an immunoglobulin type domain (amino acids 21-136) which contains amino acids with compose the antigen binding site and amino acids that form the homodimer interface.
  • the extracellular domain includes several asparagine residues which may be glycosylated, and the intracellular domain comprises serine and tyrosine residues, which may be phosphorylated.
  • the human CD8 alpha chain protein sequence is set forth by GenBank accession No. NP_001139345.1, including signal peptide (amino acids 1-21), extracellular domain (amino acids 22-182), transmembrane domain (amino acids 183-203), and cytoplasmic domain (amino acids 204-235).
  • the extracellular domain includes an immunoglobulin type domain (amino acids 28-128) which contains amino acids with compose the antigen binding site and amino acids that form the homodimer interface.
  • the extracellular domain includes several asparagine residues which may be glycosylated, and the intracellular domain comprises serine and tyrosine residues, which may be phosphorylated.
  • the human IgG4 constant region sequence is set forth in UniProtKB/Swiss-Prot: accession No. P01861.1, including CH1 (amino acids 1-98), hinge (amino acids 99-110), CH2 (amino acids 111-220), CH3 (amino acids 221-327).
  • the CH2 region includes asparagine at amino acid 177, which is the glycosylated and associated with Fc receptor and antibody-dependent cell-mediated cytotoxicity (ADCC).
  • CD137 human CD137
  • NP_001552.2 signal peptide (amino acids 1-23), extracellular domain (amino acids 24-186), transmembrane domain (amino acids 187-213), and cytoplasmic domain (amino acids 214-255).
  • binding of CD137L ligand trimers expressed on antigen presenting cells to CD137 leads to receptor trimerization and activation of signaling cascades involved in T cell reactivity and survival (Li et al., Limited Cross - Linking of 4-1 BB by 4-1 BB Ligand and the Agonist Monoclonal Antibody Utomilumab .
  • the human CD134 (OX40) protein sequence is set forth by GenBank accession No. NP_003318.1, including signal peptide (amino acids 1-28), extracellular domain (amino acids 29-214), transmembrane domain (amino acids 215-235), and cytoplasmic domain (amino acids 236-277).
  • This receptor has been shown to activate NF-kappaB through its interaction with adaptor proteins TRAF2 and TRAF5 and studies suggest that this receptor promotes expression of apoptosis inhibitors BCL2 and BCL21L1/BCL2-XL.
  • the human T-cell surface antigen CD2 has at least two isoforms.
  • the human CD2 isoform1 protein sequence is set forth by NP_001315538.1, including signal peptide (amino acids 1-24), extracellular domain (amino acids 25-235), transmembrane domain (amino acids 236-261), and cytoplasmic domain (amino acids 262-377).
  • the human CD2 isoform2 protein sequence is set forth by NP_001758.2
  • the human CD357 (GITR) isoform-1 protein sequence is set forth by GenBank accession No. NP_004186.1, including signal peptide (amino acids 1-25), extracellular domain (amino acids 26-162), transmembrane domain (amino acids 163-183), and cytoplasmic domain (amino acids 184-241).
  • the human CD29 (beta1 integrin) protein sequence is set forth by GenBank accession No. NP_596867, including signal peptide (amino acids 1-20), extracellular domain (amino acids 21-728), transmembrane domain (amino acids 729-751), and cytoplasmic domain (amino acids 752-798).
  • human CD150 (SLAM) protein sequence has at several isoforms.
  • mCD150 transmembrane form of CD150
  • sCD150 secreted form of CD150
  • human SLAM isoform b is set forth by GenBank accession No. NP_003028.1, including signal peptide (amino acids 1-20), extracellular domain (amino acids 21-237), transmembrane domain (amino acids 238-258), and cytoplasmic domain (amino acids 259-335).
  • Human SLAM isoform a is set forth by GenBank accession No. NP_001317683.1.
  • a CoStAR may be expressed alone under the control of a promoter in a therapeutic population of cells that have therapeutic activity, for example, Tumor Infiltrating Lymphocytes (TILs).
  • TILs Tumor Infiltrating Lymphocytes
  • the CoStAR may be expressed along with a therapeutic transgene such as a chimeric antigen receptor (CAR) and/or T-cell Receptor (TCR), for example as described in SEQ ID NOS:67-79 (note that may lack up to about 5, for example 1, 2, 3, 4, 5, or up to 10 amino acids at the N-terminal of the mature receptor protein).
  • CAR chimeric antigen receptor
  • TCR T-cell Receptor
  • the invention also relates to CoStAR constructs having a sequence as shown in any of SEQ ID NOS:67-79, including one of these sequences which lacks up to about 5, for example 1, 2, 3, 4, 5, or up to 10 amino acids at the N-terminal of the mature receptor protein).
  • Suitable TCRs and CARs are well known in the literature, for example HLA-A*02-NYESO-1 specific TCRs (Rapoport et al. Nat Med 2015) or anti-CD19scFv.CD3 ⁇ fusion CARs (Kochenderfer et al. J Clin Oncol 2015) which have been successfully used to treat Myeloma or B-cell malignancies respectively.
  • the CoStARs described herein may be expressed with any known CAR or TCR thus providing the cell with a regulatable growth switch to allow cell expansion in-vitro or in-vivo, and a conventional activation mechanism in the form of the TCR or CAR for anti-cancer activity.
  • the invention provides a cell for use in adoptive cell therapy comprising a CoStAR as described herein and a TCR and/or CAR that specifically binds to a tumor associated antigen.
  • An exemplary CoStAR comprising CD28 includes an extracellular antigen binding domain and an extracellular, transmembrane and intracellular signaling domain.
  • antigen binding domain refers to an antibody fragment including, but not limited to, a diabody, a Fab, a Fab′, a F(ab′) 2, an Fv fragment, a disulfide stabilized Fv fragment (dsFv), a (dsFv)2, a bispecific dsFv (dsFv-dsFv′), a disulfide stabilized diabody (ds diabody), a single-chain antibody molecule (scFv), an scFv dimer (bivalent diabody), a multispecific antibody formed from a portion of an antibody comprising one or more CDRs, a camelized single domain antibody, a nanobody, a domain antibody, a bivalent domain antibody, or any other antibody fragment that binds to an antigen but does not comprise a complete antibody structure.
  • an antigen binding domain is capable of binding to the same antigen to which the parent antibody or a parent antibody fragment (e.g., a parent scFv) binds.
  • an antigen-binding fragment may comprise one or more complementarity determining regions (CDRs) from a particular human antibody grafted to frameworks (FRs) from one or more different human antibodies.
  • the antigen binding domain can be made specific for any disease-associated antigen, including but not limited to tumor-associated antigens (TAAs) and infectious disease-associated antigens.
  • TAAs tumor-associated antigens
  • the ligand binding domain is bispecific. Antigens have been identified in most of the human cancers, including Burkitt lymphoma, neuroblastoma, melanoma, osteosarcoma, renal cell carcinoma, breast cancer, prostate cancer, lung carcinoma, and colon cancer.
  • TAA's include, without limitation, CD19, CD20, CD22, CD24, CD33, CD38, CD123, CD228, CD138, BCMA, GPC3, CEA, folate receptor (FR ⁇ ), mesothelin, CD276, gp100, 5T4, GD2, EGFR, MUC-1, PSMA, EpCAM, MCSP, SM5-1, MICA, MICB, ULBP and HER-2.
  • TAAs further include neoantigens, peptide/MHC complexes, and HSP/peptide complexes.
  • the antigen binding domain comprises a T-cell receptor or binding fragment thereof that binds to a defined tumor specific peptide-MHC complex.
  • T cell receptor refers to a heterodimeric receptor composed of ⁇ or ⁇ chains that pair on the surface of a T cell. Each ⁇ , ⁇ , ⁇ , and ⁇ chain is composed of two Ig-like domains: a variable domain (V) that confers antigen recognition through the complementarity determining regions (CDR), followed by a constant domain (C) that is anchored to cell membrane by a connecting peptide and a transmembrane (TM) region. The TM region associates with the invariant subunits of the CD3 signaling apparatus.
  • V variable domain
  • CDR complementarity determining regions
  • C constant domain
  • TM transmembrane
  • Each of the V domains has three CDRs. These CDRs interact with a complex between an antigenic peptide bound to a protein encoded by the major histocompatibility complex (pMHC) (Davis and Bjorkman (1988) Nature, 334, 395-402; Davis et al. (1998) Annu Rev Immunol, 16, 523-544; Murphy (2012), xix, 868 p.).
  • pMHC major histocompatibility complex
  • the antigen binding domain comprises a natural ligand of a tumor expressed protein or tumor-binding fragment thereof.
  • PD1 which binds to PDL1.
  • TfR1 transferrin receptor 1
  • CD71 transferrin receptor 1
  • TfR1 is expressed at a low level in a broad variety of cells, it is expressed at higher levels in rapidly proliferating cells, including malignant cells in which overexpression has been associated with poor prognosis.
  • the antigen binding domain comprises transferrin or a transferrin receptor-binding fragment thereof.
  • the antigen binding domain is specific to a defined tumor associated antigen, such as but not limited to FR ⁇ , CEA, 5T4, CA125, SM5-1 or CD71.
  • the tumor associated antigen can be a tumor-specific peptide-MHC complex.
  • the peptide is a neoantigen.
  • the tumor associated antigen it a peptide-heat shock protein complex.
  • the invention provides a CoStAR which comprises
  • CEA carcinoembryonic antigen
  • an scFv that binds to CEA ii. an scFv that binds to CEA, a spacer and transmembrane sequence of CD28, and a CD40 signaling domain.
  • scFv that binds to CEA, a spacer and transmembrane sequence of CD28, a CD137 signaling domain, and a CD40 signaling domain.
  • an scFv that binds to CEA iv. an scFv that binds to CEA, a spacer and transmembrane sequence of CD28, a CD134 signaling domain, and a CD40 signaling domain.
  • scFv that binds to CEA, a spacer and transmembrane sequence of CD28, a CD2 signaling domain, and a CD40 signaling domain.
  • an scFv that binds to CEA a spacer and transmembrane sequence of CD28, a GITR signaling domain, and a CD40 signaling domain.
  • an scFv that binds to CEA a spacer and transmembrane sequence of CD28, a CD29 signaling domain, and a CD40 signaling domain.
  • scFv that binds to CEA, a spacer and transmembrane sequence of CD28, a CD150 signaling domain, and a CD40 signaling domain.
  • scFv that binds to CEA, a spacer and transmembrane sequence of CD8, a CD28 signaling domain, and a CD40 signaling domain.
  • scFv that binds to CEA, a spacer and transmembrane sequence of CD8, a CD137 signaling domain, and a CD40 signaling domain.
  • xii an scFv that binds to CEA, a spacer and transmembrane sequence of CD8, a CD134 signaling domain, and a CD40 signaling domain.
  • xiii an scFv that binds to CEA, a spacer and transmembrane sequence of CD8, a CD2 signaling domain, and a CD40 signaling domain.
  • scFv that binds to CEA, a spacer and transmembrane sequence of CD8, a GITR signaling domain, and a CD40 signaling domain.
  • xv. an scFv that binds to CEA, a spacer and transmembrane sequence of CD8, a CD29 signaling domain, and a CD40 signaling domain.
  • an scFv that binds to CEA a spacer and transmembrane sequence of CD8, a CD150 signaling domain, and a CD40 signaling domain.
  • xvii. an scFv that binds to CEA, a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a CD28 signaling domain, and a CD40 signaling domain.
  • xviii an scFv that binds to CEA, a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, and a CD40 signaling domain.
  • xix an scFv that binds to CEA, a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a CD137 signaling domain, and a CD40 signaling domain.
  • an scFv that binds to CEA a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a CD134 signaling domain, and a CD40 signaling domain.
  • an scFv that binds to CEA a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a CD2 signaling domain, and a CD40 signaling domain.
  • an scFv that binds to CEA a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a GITR signaling domain, and a CD40 signaling domain.
  • scFv that binds to CEA
  • a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a CD29 signaling domain, and a CD40 signaling domain.
  • an scFv that binds to CEA a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a CD150 signaling domain, and a CD40 signaling domain.
  • an scFv that binds to CEA a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a first CD40 signaling domain and a second CD40 signaling domain
  • an scFv that binds to CEA a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a first CD40 signaling domain and a second mutated CD40 signaling domain
  • a binding domain that binds to PDL1 a short spacer and transmembrane sequence of CD28, a CD28 signaling domain, and a CD40 signaling domain.
  • a binding domain that binds to PDL1 a short spacer and transmembrane sequence of CD28, and a CD40 signaling domain.
  • xxix an binding domain that binds to CD155, CD112, or CD113, a CD28 transmembrane domain, a CD28 signaling domain, and a CD40 signaling domain.
  • xxx a binding domain that binds to CD155, CD112, or CD113, a CD28 transmembrane domain, and a CD40 signaling domain.
  • scFv that binds to CEA
  • a binding domain that binds to PDL1 a short spacer and transmembrane sequence of CD28
  • CD28 signaling domain a CD40 signaling domain
  • xxxii an scFv that binds to CEA, a binding domain that binds to PDL1, a short spacer and transmembrane sequence of CD28, and a CD40 signaling domain.
  • scFv that binds to CEA
  • a binding domain that binds to CD155, CD112, or CD113
  • a short spacer and transmembrane sequence of CD28 a CD28 signaling domain
  • a CD40 signaling domain a CD40 signaling domain
  • an scFv that binds to CEA a binding domain that binds to CD155, CD112, or CD113, a short spacer and transmembrane sequence of CD28, and a CD40 signaling domain.
  • the invention provides a CoStAR which comprises the spacer, transmembrane, and signaling domain structure of any one of i-xxxiv and binds to FOLR1.
  • the invention provides a CoStAR which comprises
  • scFv that binds to FOLR1
  • spacer and transmembrane sequence of CD28 a CD28 signaling domain
  • CD40 signaling domain a CD40 signaling domain
  • scFv that binds to FOLR1, a spacer and transmembrane sequence of CD28, and a CD40 signaling domain.
  • an scFv that binds to FOLR1 a spacer and transmembrane sequence of CD28, a CD134 signaling domain, and a CD40 signaling domain.
  • scFv that binds to FOLR1, a spacer and transmembrane sequence of CD28, a CD2 signaling domain, and a CD40 signaling domain.
  • scFv that binds to FOLR1, a spacer and transmembrane sequence of CD28, a GITR signaling domain, and a CD40 signaling domain.
  • scFv that binds to FOLR1, a spacer and transmembrane sequence of CD28, a CD150 signaling domain, and a CD40 signaling domain.
  • scFv that binds to FOLR1, a spacer and transmembrane sequence of CD8, a CD28 signaling domain, and a CD40 signaling domain.
  • scFv that binds to FOLR1, a spacer and transmembrane sequence of CD8, a CD137 signaling domain, and a CD40 signaling domain.
  • xii an scFv that binds to FOLR1, a spacer and transmembrane sequence of CD8, a CD134 signaling domain, and a CD40 signaling domain.
  • xiii an scFv that binds to FOLR1, a spacer and transmembrane sequence of CD8, a CD2 signaling domain, and a CD40 signaling domain.
  • scFv that binds to FOLR1, a spacer and transmembrane sequence of CD8, a GITR signaling domain, and a CD40 signaling domain.
  • xv. an scFv that binds to FOLR1, a spacer and transmembrane sequence of CD8, a CD29 signaling domain, and a CD40 signaling domain.
  • scFv that binds to FOLR1, a spacer and transmembrane sequence of CD8, a CD150 signaling domain, and a CD40 signaling domain.
  • xvii. an scFv that binds to FOLR1, a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a CD28 signaling domain, and a CD40 signaling domain.
  • xviii an scFv that binds to FOLR1, a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, and a CD40 signaling domain.
  • xix an scFv that binds to FOLR1, a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a CD137 signaling domain, and a CD40 signaling domain.
  • an scFv that binds to FOLR1 a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a CD134 signaling domain, and a CD40 signaling domain.
  • an scFv that binds to FOLR1 a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a CD2 signaling domain, and a CD40 signaling domain.
  • scFv that binds to FOLR1
  • a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a GITR signaling domain, and a CD40 signaling domain.
  • scFv that binds to FOLR1
  • a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a CD29 signaling domain, and a CD40 signaling domain.
  • an scFv that binds to FOLR1 a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a CD150 signaling domain, and a CD40 signaling domain.
  • scFv that binds to FOLR1
  • spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a first CD40 signaling domain and a second CD40 signaling domain
  • an scFv that binds to FOLR1 a spacer comprising an IgG4 constant region and CD28 transmembrane sequence, a first CD40 signaling domain and a second mutated CD40 signaling domain
  • the term “specifically binds” or “is specific for” refers to measurable and reproducible interactions, such as binding between a target and an antibody or antibody moiety that is determinative of the presence of the target in the presence of a heterogeneous population of molecules, including biological molecules.
  • an antibody moiety that specifically binds to a target is an antibody moiety that binds the target with greater affinity, avidity, more readily, and/or with greater duration than its bindings to other targets.
  • an antibody moiety that specifically binds to an antigen reacts with one or more antigenic determinants of the antigen (for example a cell surface antigen or a peptide/MHC protein complex) with a binding affinity that is at least about 10 times its binding affinity for other targets.
  • one or more antigenic determinants of the antigen for example a cell surface antigen or a peptide/MHC protein complex
  • a CoStAR of the invention optionally comprises a spacer region between the antigen binding domain and the costimulatory receptor.
  • the term “spacer” refers to the extracellular structural region of a CoStAR that separates the antigen binding domain from the external ligand binding domain of the costimulatory protein. The spacer provides flexibility to access the targeted antigen and receptor ligand. In certain embodiments long spacers are employed, for example to target membrane-proximal epitopes or glycosylated antigens (see Guest R. D. et al. The role of extracellular spacer regions in the optimal design of chimeric immune receptors: evaluation of four different scFvs and antigens. J. Immunother.
  • CoStARs bear short spacers, for example to target membrane distal epitopes (see Hudecek M. et al., Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells. Clin. Cancer Res. 2013; 19:3153-3164; Hudecek M. et al., The nonsignalling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol.
  • the spacer comprises all or part of or is derived from an IgG hinge, including but not limited to IgG1, IgG2, or IgG4.
  • a spacer can comprise all or part of one or more antibody constant domains, such as but not limited to CH2 and/or CH3 domains.
  • the CH2 domain in a spacer comprising all or part of a CH2 domain, is modified so as not to bind to an Fc receptor. For example, Fc receptor binding in myeloid cells has been found to impair CAR T cell functionality.
  • the spacer comprises all or part of an Ig-like hinge from CD28, CD8, or other protein comprising a hinge region. In certain embodiments of the invention that comprise a spacer, the spacer is from 1 and 50 amino acids in length.
  • the spacer comprises essentially all of an extracellular domain, for example a CD28 extracellular domain (i.e. from about amino acid 19, 20, 21, or 22 to about amino acid 152) or an extracellular domain of another protein, including but not limited to another TNFR superfamily member.
  • the spacer comprises a portion of an extracellular domain, for example a portion of a CD28 extracellular domain, and may lack all or most of the Ig domain.
  • the spacer includes amino acids of CD28 from about 141 to about 152 but not other portions of the CD28 extracellular domain.
  • the spacer includes amino acids of CD8 from about 128 to about 182 but not other portions of the CD8 extracellular domain.
  • the CoStAR extracellular domain comprises a linker.
  • Linkers comprise short runs of amino acids used to connect domains, for example a binding domain with a spacer or transmembrane domain.
  • a ligand binding domain will usually be connected to a spacer or a transmembrane domain by flexible linker comprising from about 5 to 25 amino acids, such as, for example, AAAGSGGSG (SEQ ID NO:7), GGGGSGGGGSGGGGS (SEQ ID NO:62).
  • a CoStAR comprises a binding domain joined directly to a transmembrane domain by a linker, and without a spacer.
  • a CoStAR comprises a binding domain joined directly to a transmembrane by a spacer and without a linker, exemplified by SEQ ID NOS:58 and 59.
  • a CoStAR comprises a full length primary costimulatory receptor which can comprise an extracellular ligand binding and intracellular signaling portion of, without limitation, CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6.
  • the costimulatory receptor comprises a chimeric protein, for instance comprising an extracellular ligand binding domain of one of the aforementioned proteins and an intracellular signaling domain of another of the aforementioned proteins.
  • the signaling portion of the CoStAR comprises a single signaling domain.
  • the signaling portion of the CoStAR comprises a second intracellular signaling domain such as but not limited to: CD2, CD27, CD28, CD40, CD134 (OX40), CD137 (4-1BB), CD150 (SLAM).
  • the first and second intracellular signaling domains are the same. In other embodiments, the first and second intracellular signaling domains are different.
  • the costimulatory receptor is capable of dimerization. Without being bound by theory, it is thought that CoStARs dimerize or associate with other accessory molecules for signal initiation. In certain embodiments, CoStARs dimerize or associate with accessory molecules through transmembrane domain interactions. In certain embodiments, dimerization or association with accessory molecules is assisted by costimulatory receptor interactions in the intracellular portion, and/or the extracellular portion of the costimulatory receptor.
  • the transmembrane domain influences CoStAR function.
  • the transmembrane domain is comprised by the full length primary costimulatory receptor domain.
  • the transmembrane domain can be that of the extracellular domain or the intracellular domain.
  • the transmembrane domain is from CD4, CD8a, CD28, or ICOS. Gueden et al.
  • the transmembrane domain comprises a hydrophobic a helix that spans the cell membrane.
  • the transmembrane domain comprises amino acids of the CD28 transmembrane domain from about amino acid 153 to about amino acid 179. In another embodiment, the transmembrane domain comprises amino acids of the CD8 transmembrane domain from about amino acid 183 to about amino acid 203. In certain embodiments, the CoStARs of the invention may include several amino acids between the transmembrane domain and signaling domain. For example, in one construct described herein the link from a CD8 transmembrane domain to a signaling domain comprises several amino acids of the CD8 cytoplasmic domain (e.g., amino acids 204-210 of CD8).
  • amino acid sequence variants of the antibody moieties or other moieties provided herein are contemplated.
  • Amino acid sequence variants of an antibody moiety may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody moiety, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody moiety. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., antigen-binding.
  • antibody binding domain moieties comprising one or more amino acid substitutions, deletions, or insertions are provided.
  • Sites of interest for mutational changes include the antibody binding domain heavy and light chain variable regions (VRs) and frameworks (FRs).
  • Amino acid substitutions may be introduced into a binding domain of interest and the products screened for a desired activity, e.g., retained/improved antigen binding or decreased immunogenicity.
  • amino acid substitutions may be introduced into one or more of the primary co-stimulatory receptor domain (extracellular or intracellular), secondary costimulatory receptor domain, or extracellular co-receptor domain. Accordingly, the invention encompasses CoStAR proteins and component parts particularly disclosed herein as well as variants thereof, i.e.
  • CoStAR proteins and component parts having at least 75%, at least 80%, at least 85%, at least 87%, 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% sequence identity to the amino acid sequences particularly disclosed herein.
  • the terms “percent similarity,” “percent identity,” and “percent homology” when referring to a particular sequence are used as set forth in the University of Wisconsin GCG software program BestFit. Other algorithms may be used, e.g. BLAST, psiBLAST or TBLASTN (which use the method of Altschul et al. (1990) J. Mol. Biol. 215: 405-410), FASTA (which uses the method of Pearson and Lipman (1988) PNAS USA 85: 2444-2448).
  • Particular amino acid sequence variants may differ from a reference sequence by insertion, addition, substitution or deletion of 1 amino acid, 2, 3, 4, 5-10, 10-20 or 20-30 amino acids.
  • a variant sequence may comprise the reference sequence with 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more residues inserted, deleted or substituted. For example, 5, 10, 15, up to 20, up to 30 or up to 40 residues may be inserted, deleted or substituted.
  • a variant may differ from a reference sequence by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more conservative substitutions.
  • Conservative substitutions involve the replacement of an amino acid with a different amino acid having similar properties. For example, an aliphatic residue may be replaced by another aliphatic residue, a non-polar residue may be replaced by another non-polar residue, an acidic residue may be replaced by another acidic residue, a basic residue may be replaced by another basic residue, a polar residue may be replaced by another polar residue or an aromatic residue may be replaced by another aromatic residue.
  • Conservative substitutions may, for example, be between amino acids within the following groups:
  • Amino acids may be grouped into different classes according to common side-chain properties: a. hydrophobic: Norleucine, Met, Ala, Val, Leu, Ile; b. neutral hydrophilic: Cys, Ser, Thr, Asn, Gln; c. acidic: Asp, Glu; d. basic: His, Lys, Arg; e. residues that influence chain orientation: Gly, Pro; aomatic: Trp, Tyr, Phe. Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • the cells used in the present invention may be any lymphocyte that is useful in adoptive cell therapy, such as a T-cell or a natural killer (NK) cell, an NKT cell, a gamma/delta T-cell or T regulatory cell.
  • the cells may be allogeneic or autologous to the patient.
  • T cells or T lymphocytes are a type of lymphocyte that have a central role in cell-mediated immunity. They can be distinguished from other lymphocytes, such as B cells and natural killer cells (NK cells), by the presence of a T-cell receptor (TCR) on the cell surface.
  • TCR T-cell receptor
  • TC cells Cytotoxic T cells
  • CTLs destroy virally infected cells and tumor cells, and are also implicated in transplant rejection.
  • CTLs express the CD8 molecule at their surface.
  • CD8+ cells recognize their targets by binding to antigen associated with MHC class I, which is present on the surface of all nucleated cells.
  • MHC class I MHC class I
  • IL-10 adenosine and other molecules secreted by regulatory T cells, the CD8+ cells can be inactivated to an anergic state, which prevent autoimmune diseases such as experimental autoimmune encephalomyelitis.
  • Memory T cells are a subset of antigen-specific T cells that persist long-term after an infection has resolved. They quickly expand to large numbers of effector T cells upon re-exposure to their cognate antigen, thus providing the immune system with “memory” against past infections.
  • Memory T cells comprise three subtypes: central memory T cells (TCM cells) and two types of effector memory T cells (TEM cells and TEMRA cells). Memory cells may be either CD4+ or CD8+.
  • Memory T cells typically express the cell surface protein CD45RO.
  • Regulatory T cells (Treg cells), formerly known as suppressor T cells, are crucial for the maintenance of immunological tolerance. Their major role is to shut down T cell-mediated immunity toward the end of an immune reaction and to suppress auto-reactive T cells that escaped the process of negative selection in the thymus.
  • Treg cells Two major classes of CD4+ Treg cells have been described—naturally occurring Treg cells and adaptive Treg cells.
  • Naturally occurring Treg cells also known as CD4 + CD25 + FoxP3 + Treg cells
  • Naturally occurring Treg cells can be distinguished from other T cells by the presence of an intracellular molecule called FoxP3.
  • Adaptive Treg cells also known as Tr1 cells or Th3 cells may originate during a normal immune response.
  • Natural Killer Cells are a type of cytolytic cell which form part of the innate immune system. NK cells provide rapid responses to innate signals from virally infected cells in an MHC independent manner. NK cells (belonging to the group of innate lymphoid cells) are defined as large granular lymphocytes (LGL) and constitute the third kind of cells differentiated from the common lymphoid progenitor generating B and T lymphocytes.
  • LGL large granular lymphocytes
  • therapeutic cells of the invention comprise autologous cells engineered to express a CoStAR.
  • therapeutic cells of the invention comprise allogeneic cells engineered to express a CoStAR.
  • Autologous cells expressing CoStARs may be advantageous in avoiding graft-versus-host disease (GVHD) due to TCR-mediated recognition of recipient alloantigens.
  • GVHD graft-versus-host disease
  • the immune system of a CoStAR recipient could attack the infused CoStAR cells, causing rejection.
  • endogenous TcR is removed from allogeneic CoStAR cells by genome editing.
  • An aspect of the invention provides a nucleic acid sequence of the invention, encoding any of the CoStARs, polypeptides, or proteins described herein (including functional portions and functional variants thereof).
  • polynucleotide “nucleotide”, and “nucleic acid” are intended to be synonymous with each other. It will be understood by a skilled person that numerous different polynucleotides and nucleic acids can encode the same polypeptide as a result of the degeneracy of the genetic code.
  • Nucleic acids according to the invention may comprise DNA or RNA. They may be single stranded or double-stranded. They may also be polynucleotides which include within them synthetic or modified nucleotides. A number of different types of modification to oligonucleotides are known in the art.
  • polynucleotides may be modified by any method available in the art. Such modifications may be carried out in order to enhance the in vivo activity or life span of polynucleotides of interest.
  • variant in relation to a nucleotide sequence include any substitution of, variation of, modification of, replacement of, deletion of or addition of one (or more) nucleic acid from or to the sequence.
  • the nucleic acid sequence may encode the protein sequence shown as SEQ ID NO:2 or a variant thereof.
  • the nucleotide sequence may comprise a codon optimised nucleic acid sequence shown engineered for expression in human cells.
  • the invention also provides a nucleic acid sequence which comprises a nucleic acid sequence encoding a CoStAR and a further nucleic acid sequence encoding a T-cell receptor (TCR) and/or chimeric antigen receptor (CAR).
  • TCR T-cell receptor
  • CAR chimeric antigen receptor
  • the nucleic acid sequences may be joined by a sequence allowing co-expression of the two or more nucleic acid sequences.
  • the construct may comprise an internal promoter, an internal ribosome entry sequence (IRES) sequence or a sequence encoding a cleavage site.
  • the cleavage site may be self-cleaving, such that when the polypeptide is produced, it is immediately cleaved into the discrete proteins without the need for any external cleavage activity.
  • Various self-cleaving sites are known, including the Foot- and Mouth disease virus (FMDV) and the 2A self-cleaving peptide.
  • the co-expressing sequence may be an internal ribosome entry sequence (IRES).
  • the co-expressing sequence may be an internal promoter.
  • the present invention provides a vector which comprises a nucleic acid sequence or nucleic acid construct of the invention.
  • Such a vector may be used to introduce the nucleic acid sequence(s) or nucleic acid construct(s) into a host cell so that it expresses one or more CoStAR(s) according to the first aspect of the invention and, optionally, one or more other proteins of interest (POI), for example a TCR or a CAR.
  • the vector may, for example, be a plasmid or a viral vector, such as a retroviral vector or a lentiviral vector, or a transposon-based vector or synthetic mRNA.
  • nucleic acids 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. See, e.g., U.S. Pat. Nos. 5,399,346, 5,580,859, 5,589,466, incorporated by reference herein in their entireties.
  • Vectors derived from retroviruses are suitable tools to achieve long-term gene transfer since they allow long-term, stable integration of a transgene or transgenes and its propagation in daughter cells.
  • the vector may be capable of transfecting or transducing a lymphocyte including a T cell or an NK cell.
  • the present invention also provides vectors in which a nucleic acid of the present invention is inserted.
  • the expression of natural or synthetic nucleic acids encoding a CoStAR, and optionally a TCR or CAR is typically achieved by operably linking a nucleic acid encoding the CoStAR and TCR/CAR polypeptide or portions thereof to one or more promoters, and incorporating the construct into an expression vector.
  • promoter elements e.g., enhancers
  • promoters regulate the frequency of transcriptional initiation.
  • 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.
  • 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.
  • CMV immediate early cytomegalovirus
  • EF-1a Elongation Growth Factor-1a
  • 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, MSCV promoter, MND 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.
  • SV40 simian virus 40
  • MMTV mouse mammary tumor virus
  • HSV human immunodeficiency virus
  • LTR long terminal repeat
  • MoMuLV promoter MoMuLV promoter
  • MSCV MND promoter
  • MND promoter an avian leukemia virus promoter
  • the vectors can be suitable for replication and integration in eukaryotic cells.
  • Typical cloning vectors contain transcription and translation terminators, initiation sequences, and promoters useful for regulation of the expression of the desired nucleic acid sequence.
  • 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, see also, WO 01/96584; WO 01/29058; and U.S. Pat. No. 6,326,193).
  • the constructs expressed are as shown in SEQ ID NOS:32-65 and 67-79.
  • the nucleic acids are multi-cistronic constructs that permit the expression of multiple transgenes (e.g., CoStAR and a TCR and/or CAR etc.) under the control of a single promoter.
  • the transgenes e.g., CoStAR and a TCR and/or CAR etc.
  • the transgenes are separated by a self-cleaving 2A peptide.
  • 2A peptides useful in the nucleic acid constructs of the invention include F2A, P2A, T2A and E2A.
  • the nucleic acid construct of the invention is a multi-cistronic construct comprising two promoters; one promoter driving the expression of CoStAR and the other promoter driving the expression of the TCR or CAR.
  • the dual promoter constructs of the invention are uni-directional. In other embodiments, the dual promoter constructs of the invention are bi-directional.
  • 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 transduced through viral vectors.
  • a source of cells e.g., immune effector cells, e.g., T cells or NK cells
  • T cells can be obtained from a number of sources, including 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.
  • T 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.
  • T cell may be collected at an apheresis center and cell storage facility where T cells can be harvested, maintained, and easily transferred.
  • the T cells can be cryopreserved and stored for later use. An acceptable duration of storage may be determined and validated and can be up to 6 months, up to a year, or longer.
  • Tumor infiltrating cells are isolated and/or expanded from a tumor, for example by a fragmented, dissected, or enzyme digested tumor biopsy or mass.
  • the TILs may be produced in a two-stage process using a tumor biopsy as the starting material: Stage 1 (generally performed over 2-3 hours) initial collection and processing of tumor material using dissection, enzymatic digestion and homogenization to produce a single cell suspension which can be directly cryopreserved to stabilize the starting material for subsequent manufacture and Stage 2 which can occur days or years later.
  • Stage 2 may be performed over 4 weeks, which may be a continuous process starting with thawing of the product of Stage 1 and growth of the TIL out of the tumor starting material (about 2 weeks) followed by a rapid expansion process of the TIL cells (about 2 weeks) to increase the amount of cells and therefore dose.
  • the TILs maybe concentrated and washed prior to formulation as a liquid suspension of cells.
  • the TIL population can be transduced at any point following collection.
  • a cryopreserved TIL population is transduced to express a CoStAR following thawing.
  • a TIL population is transduced to express a CoStAR during outgrowth or initial expansion from tumor starting material.
  • a TIL population is transduced to express a CoStAR during REP, for example but not limited to from about day 8 to about day 10 of REP.
  • An exemplary TIL preparation is described in Applicant's U.S. patent application Ser. No. 62/951,559, filed Dec. 20, 2019.
  • T cells can be further isolated by positive or negative selection techniques.
  • T cells are isolated by incubation with anti-CD3/anti-CD28-conjugated beads, such as DYNABEADS® M-450 CD3/CD28 T, for a time period sufficient for positive selection of the desired T cells.
  • the time period is about 30 minutes.
  • the time period ranges from 30 minutes to 36 hours or longer and all integer values there between.
  • the time period is at least 1, 2, 3, 4, 5, or 6 hours.
  • the time period is 10 to 24 hours.
  • the incubation time period is 24 hours. Longer incubation times may be used to isolate T cells in any situation where there are few T cells as compared to other cell types, such in isolating tumor infiltrating lymphocytes (TIL) from tumor tissue or from immunocompromised individuals. Further, use of longer incubation times can increase the efficiency of capture of CD8+ T cells. Thus, by simply shortening or lengthening the time T cells are allowed to bind to the CD3/CD28 beads and/or by increasing or decreasing the ratio of beads to T cells (as described further herein), subpopulations of T cells can be preferentially selected for or against at culture initiation or at other time points during the process.
  • TIL tumor infiltrating lymphocytes
  • subpopulations of T cells can be preferentially selected for or against at culture initiation or at other desired time points.
  • multiple rounds of selection can also be used in the context of this invention. In certain aspects, it may be desirable to perform the selection procedure and use the “unselected” cells in the activation and expansion process. “Unselected” cells can also be subjected to further rounds of selection.
  • Enrichment of a T cell population by negative selection can be accomplished with a combination of antibodies directed to surface markers unique to the negatively selected cells.
  • One method is cell sorting and/or selection via negative magnetic immunoadherence or flow cytometry that uses a cocktail of monoclonal antibodies directed to cell surface markers present on the cells negatively selected.
  • a monoclonal antibody cocktail typically includes antibodies to CD14, CD20, CD16, HLA-DR, and CD8.
  • T regulatory cells are depleted by anti-CD25 conjugated beads or other similar method of selection.
  • the methods described herein can include, e.g., selection of a specific subpopulation of immune effector cells, e.g., T cells, that are a T regulatory cell-depleted population, CD25+ depleted cells, using, e.g., a negative selection technique, e.g., described herein.
  • the population of T regulatory depleted cells contains less than 30%, 25%, 20%, 15%, 10%, 5%, 4%, 3%, 2%, 1% of CD25+ cells.
  • a specific subpopulation of CoStAR effector cells that specifically bind to a target antigen can be enriched for by positive selection techniques.
  • effector cells are enriched for by incubation with target antigen-conjugated beads for a time period sufficient for positive selection of the desired abTCR effector cells.
  • the time period is about 30 minutes.
  • the time period ranges from 30 minutes to 36 hours or longer (including all ranges between these values).
  • the time period is at least one, 2, 3, 4, 5, or 6 hours.
  • the time period is 10 to 24 hours.
  • the incubation time period is 24 hours.
  • T cells for stimulation can also be frozen after a washing step. After the washing step that removes plasma and platelets, the cells may be suspended in a freezing solution. While many freezing solutions and parameters are known in the art and will be useful in this context, one method involves using PBS containing 20% DMSO and 8% human serum albumin, or culture media containing 10% Dextran 40 and 5% Dextrose, 20% Human Serum Albumin and 7.5% DMSO, or 31.25% Plasmalyte-A, 31.25% Dextrose 5%, 0.45% NaCl, 10% Dextran 40 and 5% Dextrose, 20% Human Serum Albumin, and 7.5% DMSO or other suitable cell freezing media containing for example, Hespan and PlasmaLyte A, the cells then are frozen to ⁇ 80° C. at a rate of 1° per minute and stored in the vapor phase of a liquid nitrogen storage tank. Other methods of controlled freezing may be used as well as uncontrolled freezing immediately at ⁇ 20° C. or in liquid nitrogen.
  • the immune effector cell can be an allogeneic immune effector cell, e.g., T cell or NK cell.
  • the cell can be an allogeneic T cell, e.g., an allogeneic T cell lacking expression of endogenous T cell receptor (TCR) and/or human leukocyte antigen (HLA), e.g., HLA class I and/or HLA class II.
  • TCR endogenous T cell receptor
  • HLA human leukocyte antigen
  • a T cell lacking a functional endogenous TCR can be, e.g., engineered such that it does not express any functional TCR on its surface, engineered such that it does not express one or more subunits that comprise a functional TCR (e.g., engineered such that it does not express (or exhibits reduced expression) of TCR alpha, TCR beta, TCR gamma, TCR delta, TCR epsilon, and/or TCR zeta) or engineered such that it produces very little functional TCR on its surface.
  • the T cell can express a substantially impaired TCR, e.g., by expression of mutated or truncated forms of one or more of the subunits of the TCR.
  • substantially impaired TCR means that this TCR will not elicit an adverse immune reaction in a host.
  • a T cell described herein can be, e.g., engineered such that it does not express a functional HLA on its surface.
  • a T cell described herein can be engineered such that cell surface expression HLA, e.g., HLA class 1 and/or HLA class II, is downregulated.
  • HLA e.g., HLA class 1 and/or HLA class II
  • downregulation of HLA may be accomplished by reducing or eliminating expression of beta-2 microglobulin (B2M).
  • the T cell can lack a functional TCR and a functional HLA, e.g., HLA class I and/or HLA class II.
  • Modified T cells that lack expression of a functional TCR and/or HLA can be obtained by any suitable means, including a knock out or knock down of one or more subunit of TCR or HLA.
  • the T cell can include a knock down of TCR and/or HLA using siRNA, shRNA, clustered regularly interspaced short palindromic repeats (CRISPR) transcription-activator like effector nuclease (TALEN), or zinc finger endonuclease (ZFN).
  • siRNA siRNA
  • shRNA clustered regularly interspaced short palindromic repeats
  • TALEN clustered regularly interspaced short palindromic repeats
  • ZFN zinc finger endonuclease
  • the allogeneic cell can be a cell which does not expresses or expresses at low levels an inhibitory molecule, e.g. a cell engineered by any method described herein.
  • the cell can be a cell that does not express or expresses at low levels an inhibitory molecule, e.g., that can decrease the ability of a CoStAR-expressing cell to mount an immune effector response.
  • inhibitory molecules examples include PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, Ga19, adenosine, and TGFR beta.
  • Inhibition of an inhibitory molecule e.g., by inhibition at the DNA, RNA or protein level, can optimize a CAR-expressing cell performance.
  • an inhibitory nucleic acid e.g., an inhibitory nucleic acid, e.g., a dsRNA, e.g., an siRNA or shRNA, a clustered regularly interspaced short palindromic repeats (CRISPR), a transcription-activator like effector nuclease (TALEN), or a zinc finger endonuclease (ZFN), e.g., as described herein, can be used.
  • an inhibitory nucleic acid e.g., a dsRNA, e.g., an siRNA or shRNA, a clustered regularly interspaced short palindromic repeats (CRISPR), a transcription-activator like effector nuclease (TALEN), or a zinc finger endonuclease (ZFN), e.g., as described herein, can be used.
  • CRISPR clustered regularly interspaced short palindromic repeats
  • TALEN transcription-activator like effector nu
  • siRNA or shRNA to inhibit endogenous TCR or HLA
  • TCR expression and/or HLA expression can be inhibited using siRNA or shRNA that targets a nucleic acid encoding a TCR and/or HLA, and/or an inhibitory molecule described herein (e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, Ga19, adenosine, and TGFR beta), in a T cell.
  • siRNA or shRNA that targets a nucleic acid encoding a TCR and/or HLA, and/or an inhibitory molecule described herein (e.g., PD1,
  • siRNA and shRNAs in T cells can be achieved using any conventional expression system, e.g., such as a lentiviral expression system.
  • exemplary shRNAs that downregulate expression of components of the TCR are described, e.g., in US Publication No.: 2012/0321667.
  • Exemplary siRNA and shRNA that downregulate expression of HLA class I and/or HLA class II genes are described, e.g., in U.S. publication No.: US 2007/0036773.
  • CRISPR or “CRISPR to inhibit TCR and/or HLA” as used herein refers to a set of clustered regularly interspaced short palindromic repeats, or a system comprising such a set of repeats. “Cas”, as used herein, refers to a CRISPR-associated protein.
  • CRISPR/Cas refers to a system derived from CRISPR and Cas which can be used to silence or mutate a TCR and/or HLA gene, and/or an inhibitory molecule described herein (e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAGS, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GALS, adenosine, and TGFR beta).
  • an inhibitory molecule described herein e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3
  • CRISPR/Cas systems are found in approximately 40% of sequenced eubacteria genomes and 90% of sequenced archaea. Grissa et al. (2007) BMC Bioinformatics 8: 172. This system is a type of prokaryotic immune system that confers resistance to foreign genetic elements such as plasmids and phages and provides a form of acquired immunity. Barrangou et al. (2007) Science 315: 1709-1712; Marragini et al. (2008) Science 322: 1843-1845.
  • T cells may be activated and expanded generally using methods as described, for example, in U.S. Pat. Nos. 6,352,694; 6,534,055; 6,905,680; 6,692,964; 5,858,358; 6,887,466; 6,905,681; 7,144,575; 7,067,318; 7,172,869; 7,232,566; 7,175,843; 5,883,223; 6,905,874; 6,797,514; 6,867,041; and U.S. Patent Application Publication No. 20060121005.
  • the T cells of the invention may be expanded by contact with a surface having attached thereto an agent that stimulates a CD3/TCR complex associated signal and a ligand that stimulates a costimulatory molecule on the surface of the T cells.
  • T cell populations may be stimulated as described herein, such as by contact with an anti-CD3 antibody, or antigen-binding fragment thereof, or an anti-CD2 antibody immobilized on a surface, or by contact with a protein kinase C activator (e.g., bryostatin) in conjunction with a calcium ionophore.
  • a ligand that binds the accessory molecule is used for co-stimulation of an accessory molecule on the surface of the T cells.
  • a population of T cells can be contacted with an anti-CD3 antibody and an anti-CD28 antibody, under conditions appropriate for stimulating proliferation of the T cells.
  • an anti-CD3 antibody and an anti-CD28 antibody can be used.
  • an anti-CD28 antibody include 9.3, B-T3, XR-CD28 (Diaclone, Besancon, France) can be used as can other methods commonly known in the art (Berg et al., Transplant Proc. 30(8):3975-3977, 1998; Haanen et al., J. Exp. Med. 190(9):13191328, 1999; Garland et al., J. Immunol Meth. 227(1-2):53-63, 1999).
  • expansion can be performed using flasks or containers, or gas-permeable containers known by those of skill in the art and can proceed for 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days, about 7 days to about 14 days, about 8 days to about 14 days, about 9 days to about 14 days, about 10 days to about 14 days, about 11 days to about 14 days, about 12 days to about 14 days, or about 13 days to about 14 days.
  • the second TIL expansion can proceed for about 14 days.
  • the expansion can be performed using non-specific T-cell receptor stimulation in the presence of interleukin-2 (IL-2) or interleukin-15 (IL-15).
  • the non-specific T-cell receptor stimulus can include, for example, an anti-CD3 antibody, such as about 30 ng/ml of OKT3, a mouse monoclonal anti-CD3 antibody (commercially available from Ortho-McNeil, Raritan, N.J. or Miltenyi Biotech, Auburn, Calif.) or UHCT-1 (commercially available from BioLegend, San Diego, Calif., USA).
  • CoStAR cells can be expanded in vitro by including one or more antigens, including antigenic portions thereof, such as epitope(s), of a cancer, which can be optionally expressed from a vector, such as a human leukocyte antigen A2 (HLA-A2) binding peptide, e.g., 0.3.mu ⁇ M MART-1:26-35 (27L) or gp100:209-217 (210M), optionally in the presence of a T-cell growth factor, such as 300 IU/mL IL-2 or IL-15.
  • HLA-A2 human leukocyte antigen A2
  • T-cell growth factor such as 300 IU/mL IL-2 or IL-15.
  • CoStAR cells may also be rapidly expanded by restimulation with the same antigen(s) of the cancer pulsed onto HLA-A2-expressing antigen-presenting cells.
  • the CoStAR cells can be further stimulated with, e.g., example, irradiated, autologous lymphocytes or with irradiated HLA-A2+ allogeneic lymphocytes and IL-2.
  • the stimulation occurs as part of the expansion.
  • the expansion occurs in the presence of irradiated, autologous lymphocytes or with irradiated HLA-A2+ allogeneic lymphocytes and IL-2.
  • the cell culture medium comprises IL-2.
  • the cell culture medium comprises about 1000 IU/mL, about 1500 IU/mL, about 2000 IU/mL, about 2500 IU/mL, about 3000 IU/mL, about 3500 IU/mL, about 4000 IU/mL, about 4500 IU/mL, about 5000 IU/mL, about 5500 IU/mL, about 6000 IU/mL, about 6500 IU/mL, about 7000 IU/mL, about 7500 IU/mL, or about 8000 IU/mL, or between 1000 and 2000 IU/mL, between 2000 and 3000 IU/mL, between 3000 and 4000 IU/mL, between 4000 and 5000 IU/mL, between 5000 and 6000 IU/mL, between 6000 and 7000 IU/mL, between 7000 and 8000 IU/mL, or between 8000 IU/mL of IL-2.
  • the cell culture medium comprises OKT3 antibody.
  • the cell culture medium comprises about 0.1 ng/mL, about 0.5 ng/mL, about 1 ng/mL, about 2.5 ng/mL, about 5 ng/mL, about 7.5 ng/mL, about 10 ng/mL, about 15 ng/mL, about 20 ng/mL, about 25 ng/mL, about 30 ng/mL, about 35 ng/mL, about 40 ng/mL, about 50 ng/mL, about 60 ng/mL, about 70 ng/mL, about 80 ng/mL, about 90 ng/mL, about 100 ng/mL, about 200 ng/mL, about 500 ng/mL, about 1 ⁇ g/mL or between 0.1 ng/mL and 1 ng/mL, between 1 ng/mL and 5 ng/mL, between 5 ng/mL and 10 ng/mL, between 10 ng/mL
  • a combination of IL-2, IL-7, IL-15, and/or IL-21 are employed as a combination during the expansion.
  • IL-2, IL-7, IL-15, and/or IL-21 as well as any combinations thereof can be included during the expansion.
  • a combination of IL-2, IL-15, and IL-21 are employed as a combination during the expansion.
  • IL-2, IL-15, and IL-21 as well as any combinations thereof can be included.
  • the expansion can be conducted in a supplemented cell culture medium comprising IL-2, OKT-3, and antigen-presenting feeder cells.
  • the expansion culture media comprises about 500 IU/mL of IL-15, about 400 IU/mL of IL-15, about 300 IU/mL of IL-15, about 200 IU/mL of IL-15, about 180 IU/mL of IL-15, about 160 IU/mL of IL-15, about 140 IU/mL of IL-15, about 120 IU/mL of IL-15, or about 100 IU/mL of IL-15, or about 500 IU/mL of IL-15 to about 100 IU/mL of IL-15, or about 400 IU/mL of IL-15 to about 100 IU/mL of IL-15 or about 300 IU/mL of IL-15 to about 100 IU/mL of IL-15 or about 200 IU/mL of IL-15, or about 180 IU/mL of IL-15.
  • the expansion culture media comprises about 20 IU/mL of IL-21, about 15 IU/mL of IL-21, about 12 IU/mL of IL-21, about 10 IU/mL of IL-21, about 5 IU/mL of IL-21, about 4 IU/mL of IL-21, about 3 IU/mL of IL-21, about 2 IU/mL of IL-21, about 1 IU/mL of IL-21, or about 0.5 IU/mL of IL-21, or about 20 IU/mL of IL-21 to about 0.5 IU/mL of IL-21, or about 15 IU/mL of IL-21 to about 0.5 IU/mL of IL-21, or about 12 IU/mL of IL-21 to about 0.5 IU/mL of IL-21, or about 10 IU/mL of IL-21 to about 0.5 IU/mL of IL-21, or about 5 IU/mL of IL-21 to about 1 IU/m
  • the antigen-presenting feeder cells are PBMCs.
  • the ratio of CoStAR cells to PBMCs and/or antigen-presenting cells in the expansion is about 1 to 25, about 1 to 50, about 1 to 100, about 1 to 125, about 1 to 150, about 1 to 175, about 1 to 200, about 1 to 225, about 1 to 250, about 1 to 275, about 1 to 300, about 1 to 325, about 1 to 350, about 1 to 375, about 1 to 400, or about 1 to 500, or between 1 to 50 and 1 to 300, or between 1 to 100 and 1 to 200.
  • the primary stimulatory signal and the costimulatory signal for the T cell may be provided by different protocols.
  • the agents providing each signal may be in solution or coupled to a surface. When coupled to a surface, the agents may be coupled to the same surface (i.e., in “cis” formation) or to separate surfaces (i.e., in “trans” formation).
  • one agent may be coupled to a surface and the other agent in solution.
  • the agent providing the costimulatory signal is bound to a cell surface and the agent providing the primary activation signal is in solution or coupled to a surface. In certain aspects, both agents can be in solution.
  • the agents may be in soluble form, and then cross-linked to a surface, such as a cell expressing Fc receptors or an antibody or other binding agent which will bind to the agents.
  • a surface such as a cell expressing Fc receptors or an antibody or other binding agent which will bind to the agents.
  • aAPCs artificial antigen presenting cells
  • the two agents are immobilized on beads, either on the same bead, i.e., “cis,” or to separate beads, i.e., “trans.”
  • the agent providing the primary activation signal is an anti-CD3 antibody or an antigen-binding fragment thereof and the agent providing the costimulatory signal is an anti-CD28 antibody or antigen-binding fragment thereof; and both agents are co-immobilized to the same bead in equivalent molecular amounts.
  • a 1:1 ratio of each antibody bound to the beads for CD4+ T cell expansion and T cell growth is used.
  • a ratio of anti CD3:CD28 antibodies bound to the beads is used such that an increase in T cell expansion is observed as compared to the expansion observed using a ratio of 1:1. In one particular aspect an increase of from about 1 to about 3 fold is observed as compared to the expansion observed using a ratio of 1:1.
  • the ratio of CD3:CD28 antibody bound to the beads ranges from 100:1 to 1:100 and all integer values there between. In one aspect of the present invention, more anti-CD28 antibody is bound to the particles than anti-CD3 antibody, i.e., the ratio of CD3:CD28 is less than one. In certain aspects of the invention, the ratio of anti CD28 antibody to anti CD3 antibody bound to the beads is greater than 2:1.
  • a 1:100 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:75 CD3:CD28 ratio of antibody bound to beads is used. In a further aspect, a 1:50 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:30 CD3:CD28 ratio of antibody bound to beads is used. In one preferred aspect, a 1:10 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:3 CD3:CD28 ratio of antibody bound to the beads is used. In yet one aspect, a 3:1 CD3:CD28 ratio of antibody bound to the beads is used.
  • Ratios of particles to cells from 1:500 to 500:1 and any integer values in between may be used to stimulate T cells or other target cells.
  • the ratio of particles to cells may depend on particle size relative to the target cell. For example, small sized beads could only bind a few cells, while larger beads could bind many.
  • the ratio of cells to particles ranges from 1:100 to 100:1 and any integer values in-between and in further aspects the ratio comprises 1:9 to 9:1 and any integer values in between, can also be used to stimulate T cells.
  • the ratio of anti-CD3- and anti-CD28-coupled particles to T cells that result in T cell stimulation can vary as noted above, however certain preferred values include 1:100, 1:50, 1:40, 1:30, 1:20, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, and 15:1 with one preferred ratio being at least 1:1 particles per T cell.
  • a ratio of particles to cells of 1:1 or less is used.
  • a preferred particle:cell ratio is 1:5.
  • the ratio of particles to cells can be varied depending on the day of stimulation.
  • the ratio of particles to cells is from 1:1 to 10:1 on the first day and additional particles are added to the cells every day or every other day thereafter for up to 10 days, at final ratios of from 1:1 to 1:10 (based on cell counts on the day of addition).
  • the ratio of particles to cells is 1:1 on the first day of stimulation and adjusted to 1:5 on the third and fifth days of stimulation.
  • particles are added on a daily or every other day basis to a final ratio of 1:1 on the first day, and 1:5 on the third and fifth days of stimulation.
  • the ratio of particles to cells is 2:1 on the first day of stimulation and adjusted to 1:10 on the third and fifth days of stimulation.
  • particles are added on a daily or every other day basis to a final ratio of 1:1 on the first day, and 1:10 on the third and fifth days of stimulation.
  • ratios will vary depending on particle size and on cell size and type.
  • the most typical ratios for use are in the neighborhood of 1:1, 2:1 and 3:1 on the first day.
  • the cells such as T cells
  • the cells are combined with agent-coated beads, the beads and the cells are subsequently separated, and then the cells are cultured.
  • the agent-coated beads and cells prior to culture, are not separated but are cultured together.
  • the beads and cells are first concentrated by application of a force, such as a magnetic force, resulting in increased ligation of cell surface markers, thereby inducing cell stimulation.
  • Retrovirus-based gene delivery is a mature, well-characterized technology, which has been used to permanently integrate CARs into the host cell genome (Scholler J., e.g. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci. Transl. Med. 2012; 4:132ra53; Rosenberg S. A. et al., Gene transfer into humans—immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduction. N. Engl. J. Med. 1990; 323:570-578)
  • Non-viral DNA transfection methods can also be used.
  • Singh et al describes use of the Sleeping Beauty (SB) transposon system developed to engineer CAR T cells (Singh H., et al., Redirecting specificity of T-cell populations for CD19 using the Sleeping Beauty system. Cancer Res. 2008; 68:2961-2971) and is being used in clinical trials (see e.g., ClinicalTrials.gov: NCT00968760 and NCT01653717).
  • SB Sleeping Beauty
  • SB100X hyperactive transposase
  • MMs L. et al. Molecular evolution of a novel hyperactive Sleeping Beauty transposase enables robust stable gene transfer in vertebrates. Nat. Genet. 2009; 41:753-761
  • multiple transgenes can be delivered from multicistronic single plasmids (e.g., Thokala R.
  • Morita et al describes the piggyBac transposon system to integrate larger transgenes (Morita D. et al., Enhanced expression of anti-CD19 chimeric antigen receptor in piggyBac transposon-engineered T cells. Mol. Ther. Methods Clin. Dev. 2017; 8:131-140)
  • Nakazawa et al. describes use of the system to generate EBV-specific cytotoxic T-cells expressing HER2-specific chimeric antigen receptor (Nakazawa Y et al, PiggyBac-mediated cancer immunotherapy using EBV-specific cytotoxic T-cells expressing HER2-specific chimeric antigen receptor. Mol. Ther. 2011; 19:2133-2143).
  • Manuri et al used the system to generate CD-19 specific T cells (Manuri P. V. R. et al., piggyBac transposon/transposase system to generate CD19-specific T cells for the treatment of B-lineage malignancies. Hum. Gene Ther. 2010; 21:427-437).
  • Transposon technology is easy and economical.
  • One potential drawback is the longer expansion protocols currently employed may result in T cell differentiation, impaired activity and poor persistence of the infused cells.
  • Monjezi et al describe development minicircle vectors that minimize these difficulties through higher efficiency integrations (Monjezi R. et al., Enhanced CAR T-cell engineering using non-viral Sleeping Beauty transposition from minicircle vectors. Leukemia. 2017; 31:186-194). These transposon technologies can be used for CoStARs of the invention.
  • the present invention also relates to a pharmaceutical composition containing a vector or a CoStAR expressing cell of the invention together with a pharmaceutically acceptable carrier, diluent or excipient, and optionally one or more further pharmaceutically active polypeptides and/or compounds.
  • a pharmaceutical composition comprising a CoStAR described above and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition comprising a nucleic acid encoding a CoStAR according to any of the embodiments described above and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition is provided comprising an effector cell expressing a CoStAR described above and a pharmaceutically acceptable carrier.
  • Such a formulation may, for example, be in a form suitable for intravenous infusion.
  • pharmaceutically acceptable or “pharmacologically compatible” is meant a material that is not biologically or otherwise undesirable, e.g., the material may be incorporated into a pharmaceutical composition administered to a patient without causing any significant undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained.
  • Pharmaceutically acceptable carriers or excipients have preferably met the required standards of toxicological and manufacturing testing and/or are included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug administration.
  • An aspect of the invention provides a population of modified T cells expressing a recombinant CoStAR.
  • a suitable population may be produced by a method described above.
  • the population of modified T cells may be for use as a medicament.
  • a population of modified T cells as described herein may be used in cancer immunotherapy therapy, for example adoptive T cell therapy.
  • aspects of the invention provide the use of a population of modified T cells as described herein for the manufacture of a medicament for the treatment of cancer, a population of modified T cells as described herein for the treatment of cancer, and a method of treatment of cancer may comprise administering a population of modified T cells as described herein to an individual in need thereof.
  • the population of modified T cells may be autologous i.e. the modified T cells were originally obtained from the same individual to whom they are subsequently administered (i.e. the donor and recipient individual are the same).
  • a suitable population of modified T cells for administration to the individual may be produced by a method comprising providing an initial population of T cells obtained from the individual, modifying the T cells to express a cAMP PDE or fragment thereof and an antigen receptor which binds specifically to cancer cells in the individual, and culturing the modified T cells.
  • the population of modified T cells may be allogeneic i.e. the modified T cells were originally obtained from a different individual to the individual to whom they are subsequently administered (i.e. the donor and recipient individual are different).
  • the donor and recipient individuals may be HLA matched to avoid GVHD and other undesirable immune effects.
  • a suitable population of modified T cells for administration to a recipient individual may be produced by a method comprising providing an initial population of T cells obtained from a donor individual, modifying the T cells to express a CoStAR which binds specifically to cancer cells in the recipient individual, and culturing the modified T cells.
  • the recipient individual may exhibit a T cell mediated immune response against cancer cells in the recipient individual. This may have a beneficial effect on the cancer condition in the individual.
  • Cancer conditions may be characterised by the abnormal proliferation of malignant cancer cells and may include leukaemias, such as AML, CML, ALL and CLL, lymphomas, such as Hodgkin lymphoma, non-Hodgkin lymphoma and multiple myeloma, and solid cancers such as sarcomas, skin cancer, melanoma, bladder cancer, brain cancer, breast cancer, uterus cancer, ovary cancer, prostate cancer, lung cancer, colorectal cancer, cervical cancer, liver cancer, head and neck cancer, oesophageal cancer, pancreas cancer, renal cancer, adrenal cancer, stomach cancer, testicular cancer, cancer of the gall bladder and biliary tracts, thyroid cancer, thymus cancer, cancer of bone, and cerebral cancer, as well as cancer of unknown primary (CUP).
  • leukaemias such as AML, CML, ALL and CLL
  • lymphomas such as Hodgkin lymphoma, non-Hodgkin lymphoma and multiple mye
  • Cancer cells within an individual may be immunologically distinct from normal somatic cells in the individual (i.e. the cancerous tumor may be immunogenic).
  • the cancer cells may be capable of eliciting a systemic immune response in the individual against one or more antigens expressed by the cancer cells.
  • the tumor antigens that elicit the immune response may be specific to cancer cells or may be shared by one or more normal cells in the individual.
  • An individual suitable for treatment as described above may be a mammal, such as a rodent (e.g. a guinea pig, a hamster, a rat, a mouse), murine (e.g. a mouse), canine (e.g. a dog), feline (e.g. a cat), equine (e.g. a horse), a primate, simian (e.g. a monkey or ape), a monkey (e.g. marmoset, baboon), an ape (e.g. gorilla, chimpanzee, orang-utan, gibbon), or a human.
  • a rodent e.g. a guinea pig, a hamster, a rat, a mouse
  • murine e.g. a mouse
  • canine e.g. a dog
  • feline e.g. a cat
  • equine e.g. a horse
  • the individual is a human.
  • non-human mammals especially mammals that are conventionally used as models for demonstrating therapeutic efficacy in humans (e.g. murine, primate, porcine, canine, or rabbit animals) may be employed.
  • therapeutically effective amount refers to an amount of a CoStAR or composition comprising a CoStAR as disclosed herein, effective to “treat” a disease or disorder in an individual.
  • the therapeutically effective amount of a CoStAR or composition comprising a CoStAR as disclosed herein can reduce the number of cancer cells; reduce the tumor size or weight; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer.
  • a CoStAR or composition comprising a CoStAR as disclosed herein can prevent growth and/or kill existing cancer cells, it can be cytostatic and/or cytotoxic.
  • the therapeutically effective amount is a growth inhibitory amount. In some embodiments, the therapeutically effective amount is an amount that improves progression free survival of a patient.
  • the therapeutically effective amount of a CoStAR or composition comprising a CoStAR as disclosed herein can reduce the number of cells infected by the pathogen; reduce the production or release of pathogen-derived antigens; inhibit (i.e., slow to some extent and preferably stop) spread of the pathogen to uninfected cells; and/or relieve to some extent one or more symptoms associated with the infection.
  • the therapeutically effective amount is an amount that extends the survival of a patient.
  • T and NK cells expressing CoStARs for use in the methods of the present may either be created ex vivo either from a patient's own peripheral blood (autologous), or in the setting of a haematopoietic stem cell transplant from donor peripheral blood (allogenic), or peripheral blood from an unconnected donor (allogenic).
  • T-cells or NK cells may be derived from ex-vivo differentiation of inducible progenitor cells or embryonic progenitor cells to T-cells or NK cells.
  • T-cells expressing a CoStAR and, optionally, a CAR and/or TCR are generated by introducing DNA or RNA coding for the CoStAR and, optionally, a CAR and/or TCR, by one of many means including transduction with a viral vector, transfection with DNA or RNA.
  • T or NK cells expressing a CoStAR of the present invention and, optionally, expressing a TCR and/or CAR may be used for the treatment of haematological cancers or solid tumors.
  • a method for the treatment of disease relates to the therapeutic use of a vector or cell, including a T or NK cell, of the invention.
  • the vector, or T or NK cell may be administered to a subject having an existing disease or condition in order to lessen, reduce or improve at least one symptom associated with the disease and/or to slow down, reduce or block the progression of the disease.
  • the method of the invention may cause or promote T-cell mediated killing of cancer cells.
  • the vector, or T or NK cell according to the present invention may be administered to a patient with one or more additional therapeutic agents.
  • the one or more additional therapeutic agents can be co-administered to the patient.
  • co-administering is meant administering one or more additional therapeutic agents and the vector, or T or NK cell of the present invention sufficiently close in time such that the vector, or T or NK cell can enhance the effect of one or more additional therapeutic agents, or vice versa.
  • the vectors or cells can be administered first and the one or more additional therapeutic agents can be administered second, or vice versa.
  • the vectors or cells and the one or more additional therapeutic agents can be administered simultaneously.
  • One co-administered therapeutic agent that may be useful is IL-2, as this is currently used in existing cell therapies to boost the activity of administered cells.
  • IL-2 treatment is associated with toxicity and tolerability issues.
  • the CoStAR effector cells can be allogeneic or autologous to the patient.
  • allogeneic cells are further genetically modified, for example by gene editing, so as to minimize or prevent GVHD and/or a patient's immune response against the CoStAR cells.
  • the CoStAR effector cells are used to treat cancers and neoplastic diseases associated with a target antigen.
  • Cancers and neoplastic diseases that may be treated using any of the methods described herein include tumors that are not vascularized, or not yet substantially vascularized, as well as vascularized tumors.
  • the cancers may comprise non-solid tumors (such as hematological tumors, for example, leukemias and lymphomas) or may comprise solid tumors.
  • Types of cancers to be treated with the CoStAR effector cells of the invention include, but are not limited to, carcinoma, blastoma, and sarcoma, and certain leukemia or lymphoid malignancies, benign and malignant tumors, and malignancies e.g., sarcomas, carcinomas, and melanomas.
  • carcinoma a malignant neoplasm
  • blastoma a malignant na
  • sarcoma e.g., sarcomas, carcinomas, and melanomas.
  • malignancies e.g., sarcomas, carcinomas, and melanomas.
  • adult tumors/cancers and pediatric tumors/cancers are also included.
  • Hematologic cancers are cancers of the blood or bone marrow.
  • hematological (or hematogenous) cancers include leukemias, including acute leukemias (such as acute lymphocytic leukemia, acute myelocytic leukemia, acute myelogenous leukemia and myeloblastic, promyelocytic, myelomonocytic, monocytic and erythroleukemia), chronic leukemias (such as chronic myelocytic (granulocytic) leukemia, chronic myelogenous leukemia, and chronic lymphocytic leukemia), polycythemia vera, lymphoma, Hodgkin's disease, non-Hodgkin's lymphoma (indolent and high grade forms), multiple myeloma, plasmacytoma, Waldenstrom's macroglobulinemia, heavy chain disease, myelodysplastic syndrome, hairy cell leukemia and myelodysplasia.
  • Solid tumors are abnormal masses of tissue that usually do not contain cysts or liquid areas. Solid tumors can be benign or malignant. Different types of solid tumors are named for the type of cells that form them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumors, such as sarcomas and carcinomas, include adrenocortical carcinoma, cholangiocarcinoma, fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteosarcoma, and other sarcomas, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, stomach cancer, lymphoid malignancy, pancreatic cancer, breast cancer, lung cancers, ovarian cancer, prostate cancer, hepatocellular carcinoma, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma,
  • an immunologically effective amount When “an immunologically effective amount,” “an anti-tumor effective amount,” “a tumor-inhibiting effective amount,” or “therapeutic amount” is indicated, the precise amount of the compositions of the present invention to be administered can be determined by a physician with consideration of individual differences in age, weight, tumor size, extent of infection or metastasis, and condition of the patient (subject). It can generally be stated that a pharmaceutical composition comprising the T cells described herein may be administered at a dosage of 10 4 to 10 9 cells/kg body weight, in some instances 10 5 to 10 6 cells/kg body weight, including all integer values within those ranges. T cell compositions may also be administered multiple times at these dosages. The cells can be administered by using infusion techniques that are commonly known in immunotherapy (see, e.g., Rosenberg et al., New Eng. J. of Med. 319:1676, 1988).
  • a CoStAR-expressing cell described herein may be used in combination with other known agents and therapies.
  • Administered “in combination”, as used herein, means that two (or more) different treatments are delivered to the subject during the course of the subject's affliction with the disorder, e.g., the two or more treatments are delivered after the subject has been diagnosed with the disorder and before the disorder has been cured or eliminated or treatment has ceased for other reasons.
  • the delivery of one treatment is still occurring when the delivery of the second begins, so that there is overlap in terms of administration. This is sometimes referred to herein as “simultaneous” or “concurrent delivery”.
  • the delivery of one treatment ends before the delivery of the other treatment begins.
  • the treatment is more effective because of combined administration.
  • the second treatment is more effective, e.g., an equivalent effect is seen with less of the second treatment, or the second treatment reduces symptoms to a greater extent, than would be seen if the second treatment were administered in the absence of the first treatment, or the analogous situation is seen with the first treatment.
  • delivery is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one treatment delivered in the absence of the other.
  • the effect of the two treatments can be partially additive, wholly additive, or greater than additive.
  • the delivery can be such that an effect of the first treatment delivered is still detectable when the second is delivered.
  • a CoStAR-expressing cell described herein and the at least one additional therapeutic agent can be administered simultaneously, in the same or in separate compositions, or sequentially.
  • the CAR-expressing cell described herein can be administered first, and the additional agent can be administered second, or the order of administration can be reversed.
  • the CoStAR therapy and/or other therapeutic agents, procedures or modalities can be administered during periods of active disorder, or during a period of remission or less active disease.
  • the CoStAR therapy can be administered before the other treatment, concurrently with the treatment, post-treatment, or during remission of the disorder.
  • the therapy and the additional agent can be administered in an amount or dose that is higher, lower or the same than the amount or dosage of each agent used individually, e.g., as a monotherapy.
  • the administered amount or dosage of the CoStAR therapy, the additional agent (e.g., second or third agent), or all is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50%) than the amount or dosage of each agent used individually, e.g., as a monotherapy.
  • the amount or dosage of the CoStAR therapy, the additional agent (e.g., second or third agent), or all, that results in a desired effect is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50% lower) than the amount or dosage of each agent used individually, e.g., as a monotherapy, required to achieve the same therapeutic effect.
  • a CoStAR-expressing cell described herein may be used in a treatment regimen in combination with surgery, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies or other antibody therapies, cytoxin, fludarabine, cyclosporin, FK506, rapamycin, mycophenolic acid, steroids, FR901228, cytokines, and irradiation, peptide vaccine, such as that described in Izumoto et al. 2008 J Neurosurg 108:963-971.
  • immunosuppressive agents such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies
  • immunoablative agents such as CAMPATH, anti-CD3 antibodies or other antibody therapies, cytoxin, fludarabine, cyclosporin, FK506,
  • compounds of the present invention are combined with other therapeutic agents, such as other anti-cancer agents, anti-allergic agents, anti-nausea agents (or anti-emetics), pain relievers, cytoprotective agents, and combinations thereof.
  • other therapeutic agents such as other anti-cancer agents, anti-allergic agents, anti-nausea agents (or anti-emetics), pain relievers, cytoprotective agents, and combinations thereof.
  • a CoStAR-expressing cell described herein can be used in combination with a chemotherapeutic agent.
  • chemotherapeutic agents include an anthracycline (e.g., doxorubicin (e.g., liposomal doxorubicin)), a vinca alkaloid (e.g., vinblastine, vincristine, vindesine, vinorelbine), an alkylating agent (e.g., cyclophosphamide, decarbazine, melphalan, ifosfamide, temozolomide), an immune cell antibody (e.g., alemtuzamab, gemtuzumab, rituximab, ofatumumab, tositumomab, brentuximab), an antimetabolite (including, e.g., folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase
  • chemotherapeutic agents considered for use in combination therapies include busulfan (Myleran®), busulfan injection (Busulfex®), cladribine (Leustatin®), cyclophosphamide (Cytoxan® or Neosar®), cytarabine, cytosine arabinoside (Cytosar-U®), cytarabine liposome injection (DepoCyt®), daunorubicin hydrochloride (Cerubidine®), daunorubicin citrate liposome injection (DaunoXome®), dexamethasone, doxorubicin hydrochloride (Adriamycin®, Rubex®), etoposide (Vepesid®), fludarabine phosphate (Fludara®), hydroxyurea (Hydrea®), Idarubicin (Idamycin®), mitoxantrone (Novantrone®), Gemtuzumab Ozogamicin
  • general chemotherapeutic agents considered for use in combination therapies include anastrozole (Arimidex®), bicalutamide (Casodex®), bleomycin sulfate (Blenoxane®), busulfan (Myleran®), busulfan injection (Busulfex®), capecitabine (Xeloda®), N4-pentoxycarbonyl-5-deoxy-5-fluorocytidine, carboplatin (Paraplatin®), carmustine (BiCNU®), chlorambucil (Leukeran®), cisplatin (Platinol®), cladribine (Leustatin®), cyclophosphamide (Cytoxan® or Neosar®), cytarabine, cytosine arabinoside (Cytosar-U®), cytarabine liposome injection (DepoCyt®), dacarbazine (DTIC-Dome®), d
  • Treatments can be evaluated, for example, by tumor regression, tumor weight or size shrinkage, time to progression, duration of survival, progression free survival, overall response rate, duration of response, quality of life, protein expression and/or activity.
  • Approaches to determining efficacy of the therapy can be employed, including for example, measurement of response through radiological imaging.
  • sequences include complete CoStARs and CoStAR components and are non-limiting.
  • Components include signal peptides (SP), binding domains (BD), linkers, spacers and transmembrane domains (STM), a CD28 transmembrane fragment without extracellular or intracellular sequences (STM-CD28TM), intracellular signal domains (SD) and CD40 domains and motifs.
  • SEQ ID NOS:33-108 comprise CoStARs with N-terminal signal peptides.
  • Component locations within whole proteins can be confirmed from GenBank and other sources. The constructs and components are illustrative as to precise sizes and extents and components can be from more than one source.
  • the multiple domains can be in any order. It will be understood that whereas certain proteins may comprise N-terminal signal peptides when expressed, those signal peptides are cleaved and may be imprecisely cleaved when the proteins are expressed, and that the resulting proteins from which signal peptides are removed comprise binding domains having variation of up to about five amino acids in the location of the N-terminal amino acid.
  • Sequence 1 SP-OSM MGVLLTQRTL LSLVLALLFP SMASM 2 SP-PD1 MQIPQAPWPV VWAVLQLGWR PGW 3 SP-TGIT MRWCLLLIWA QGLRQAPLAS G 4 BD1-MOV19 QVQLQQSGAE LVKPGASVKI SCKASGYSFT GYFMNWVKQS HGKSLEWIGR IHPYDGDTFY NQNFKDKATL TVDKSSNTAH MELLSLTSED FAVYYCTRYD GSRAMDYWGQ GTTVTVSSGG GGSGGGGSGG GGSDIELTQS PASLAVSLGQ RAIISCKASQ SVSFAGTSLM HWYHQKPGQQ PKLLIYRASN LEAGVPTRFS GSGSKTDFTL NIHPVEEEDA ATYYCQQSRE YPYTFGGGTK LEIK 5 BD1-MFE23 QVQLQ
  • the MFE23 CoStAR consists of an MFE23 derived single chain antibody fragment nucleotide sequence with an oncostatin M1 leader sequence fused to the entire human CD28 nucleic acid sequence.
  • the CoStAR nucleotide sequence was codon optimised and gene synthesised by Genewiz Inc. The constructs were cloned into pSF.Lenti (Oxford Genetics) via an XbaI and NheI site.
  • Lentiviral Production was performed using a three-plasmid packaging system (Cell Biolabs, San Diego, USA) by mixing 10 ⁇ g of each plasmid, plus 10 ⁇ g of the pSF.Lenti lentiviral plasmid containing the transgene, together in serum free RPMI containing 50 mM CaCl 2 ). The mixture was added dropwise to a 50% confluent monolayer of 293T cells in 75 cm 2 flasks. The viral supernatants were collected at 48 and 72 h post transfection, pooled and concentrated using LentiPac lentiviral supernatant concentration (GeneCopoeia, Rockville, Md., USA) solution according to the manufacturer's instructions.
  • LentiPac lentiviral supernatant concentration GeneCopoeia, Rockville, Md., USA
  • Lentiviral supernatants were concentrated 10-fold and used to directly infect primary human T-cells in the presence of 4 ⁇ g/ml polybrene (Sigma-Aldrich, Dorset, UK). Peripheral blood mononuclear cells were isolated from normal healthy donors before activation for 24 hours with T-cell activation and expansion beads (Invitrogen) according to the manufacturer's instructions before addition of lentiviral supernatants.
  • Cell transduction was assessed 96 hours post infection using CEA.hFc protein and anti-hFc-PE secondary, plus anti-CD34-APC or by anti-CD34-PE antibodies alone. Cells were then expanded further using ⁇ 10 donor mismatched irradiated PB MC feeders at a 1:20-1:200 ratio in RPMI+10% FCS with the addition of 1 ⁇ g/ml PHA and 200 IU/ml IL-2. After 14 days the cells were stained as previous and stored ready for assay.
  • Proliferation assays were performed by first loading T-cells with 10 ⁇ M eFluor450 proliferation dye (eBioscience, UK) for 10 min at 37° C. at a concentration of 1 ⁇ 10 7 cells/ml before incubating the cells in 5 volumes of cold T-cell media for 5 min on ice. Cells were then washed excessively to remove unbound dye and added to cocultures containing tumor cells. Cells were removed at 2, 6 and 10 days, 1:200 dilution of DRAQ7 added and the cells analysed using a MACSQuant cytometer and MACSQuantify software.
  • Cell counts for proliferation assays were performed by taking cells from the wells and staining with anti-CD2 PerCP eFluor710 antibody (eBioscience, UK) for 20 min in the dark, followed by DRAQ7 staining and counts made using a MACSQuant analyser.
  • T-cells Primary human T-cells were isolated from Buffy coats obtained from commercial suppliers (Lonza or NHSBT). T-cells were isolated by Ficoll-mediated isolation and T-cell negative isolation kits (StemCell Technologies). The isolated T-cells were activated with human T-cell activation and expansion beads (Invitrogen, UK). Cells were incubated with concentrated lentiviral particles and expanded over a number of days.
  • the lentivirus contained the DNA sequence of the MFE.CoStAR.2A.tCD34 construct (MFE23.scFv fused to full length human CD28 co-expressed with truncated human CD34 via a 2A cleavage sequence).
  • Donor 1 transduction was measured at 22.69% (17.15 CD34+/CoStAR+ plus 5.53% CD34 ⁇ /CoStAR+), donor 2 was measured at 20.73%, and donor 3 at 13.34%.
  • Cells were enriched for CoStAR expression using anti-CD34 antibodies to obtain T-cell populations greater than 90% CoStAR positive.
  • the non-transduced and transduced cells were tested against the CEA+ tumor cell lines LoVo and LS174T.
  • the tumor cells To enable activation of the T-cells in response to the unmatched tumor lines we engineered the tumor cells to express an anti-CD3 single chain antibody fragment anchored to the cell membrane by way of a synthetic transmembrane domain and split from the GFP marker gene using an IRES element to visualise transduced cells using flow cytometry.
  • CoStAR enhanced IL-2 secretion towards OKT3 engineered tumor cells was found in all three donors tested. The effect was most evident at E:T ratios of 8:1 and 16:1 and at higher E:T ratios IL-2 secretion was too low to measure accurately. At lower effector to target ratios it appeared that IL-2 secretion was saturating from non-transduced cells. These observations were repeated in LoVo cells with two of the three donors tested against LS174T with similar results ( FIGS. 3D & E).
  • CoStAR engineered cells went through 5, 6 or 7 proliferation cycles over 6 days compared to non-engineered cells in response to LoVo-OKT3, whereas CoStAR transduced and non-transduced cells went through an average of approximately 2 cycles over the same duration in response to wild-type LoVo.
  • CD28(IEV) is truncated such that the C-terminal motif of CD28 is the amino acid triad ‘IEV’.
  • Sequences were generated de novo by Genewiz and cloned into a lentiviral vector under an EF1 ⁇ promoter along with a CD34 marker gene separated from the fusion CoStAR by a 2A self-cleaving peptide.
  • Primary CD8+ T-cells were isolated using EasySep beads (StemCell Technologies) and activated with anti-CD3/anti-CD28 activation/expansion Dynabeads before addition of lentiviral particles.
  • FIG. 5 shows the IL-2 response from CD34 ⁇ (CoStAR non-transduced) and CD34+(CoStAR transduced). Statistical analysis demonstrated that all receptors tested induced a significant increase in the proportion of cells producing IL-2 when harboring the variant CoStAR receptors.
  • IFN ⁇ a cytokine released under normal signal 1 conditions but enhanced by costimulation
  • CD107a a marker of degranulation
  • Bcl-xL an antiapoptotic protein upregulated by costimulation.
  • Engagement of CoStAR enhanced all the effector functions analysed to varying degrees.
  • CD28 and CD28.CD40 based CoStARs were transduced with either the CD28(IEV) truncated CoStAR, full length CD28 CoStAR or CD28.CD40 CoStAR (having the full length CD28 as shown in SEQ ID NO. 10, but lacking the N terminal N and K residues) or left non-transduced. T-cells were enriched for CoStAR expression using the CD34 marker gene, and following expansion cells were mixed with LoVo-OKT3 cells and IL-2 secretion analysed by ELISA (See FIG. 7 ).
  • Non-transduced cells on average produced 0.80 ng/ml IL-2, with CD28(IEV) and full length CD28 CoStAR producing 4.6 and 5.0 ng/ml IL-2 respectively.
  • CD28.CD40 induced 29.0 ng/ml IL-2 on average across three donors thus demonstrating a clear benefit to incorporating CD40 into the basic CD28-based CoStAR.
  • T-cells from seven donors were transduced with either CD28 or CD28.CD40 CoStARs with either an anti-CA125 (196-14) or anti-Folate receptor (MOV-19) scFv, or an anti-Folate receptor peptide (C7) antigen binding domain. Additional cells were transduced with a CD28 CoStAR harboring an anti-CEA scFv as a mismatched control. Cells were then mixed with CA125+/Folate receptor+/CEA ⁇ cell line OvCAR3 engineered to express a membrane bound OKT3 (OvCAR-OKT3).
  • OvCAR-OKT3 membrane bound OKT3
  • T-cells were engineered with a murine constant domain modified TCR which recognizes a CEA peptide (691-699) in the context of HLA-A*02 as well as the CD28 or CD28.CD40 CoStAR targeted towards cell surface CEA protein. As a control cells were also transduced with a CA125 specific CD28 CoStAR. The T-cells were mixed with HLA-A*02+/CEA+H508 cells and cytokine production analysed by intracellular flow cytometry staining.
  • Flow cytometric gating was performed using antibodies directed towards the murine TCR ⁇ constant domain (marks the TCR engineered cells) as well as the DYKDDDDK (SEQ ID NO:14) epitope tag (marks the CoStAR engineered cells).
  • TCR ⁇ /CoStAR ⁇ , TCR+/CoStAR ⁇ , TCR ⁇ /CoStAR+ and TCR+/CoStAR+ cells were analysed in each subpopulation in either the CD4+ or CD8+ T-cells ( FIG. 9 ).
  • CD28.CD40 CoStAR enhanced CD137 and TNF ⁇ production above TCR stimulation alone, however the TCR response in CD4+ cells was poor due to the dependency of the TCR on CD8.
  • CD8+ cells there was more robust effector activity with IL-2 and CD107a in particular showing a stronger induction in the CD28.CD40 CoStAR groups.
  • the effector activity in just the TCR+/CoStAR+ groups was plotted in CD4+ and CD8+ cells ( FIGS. 10A-10B ).
  • CD4+ cells induction of CD137 was significantly enhanced by CD28.CD40 compared to either CEA or mismatched targeting CD28 CoStAR.
  • CD137 induction was significantly increased compared to either CEA or mismatched targeting CD28 CoStAR, whereas CD107a induction was increased compared to the control CoStAR.
  • CD28.CD40 shows enhanced effector activity across a broad range of models and effector activities.
  • MFE23.CD28 or MFE23.CD28.CD40 CoStAR were mock transduced or transduced with MFE23.CD28 or MFE23.CD28.CD40 CoStAR, each harboring a CD34 marker gene separated by a 2A cleavage peptide.
  • MFE23 is a single chain Fv antibody that has a high affinity for carcinoembryonic antigen (CEA).
  • CEA carcinoembryonic antigen
  • MACSTM paramagnetic selection reagents Miltenyi Biotech
  • MFE23.CD28 CoStAR strongly mediated expansion of CD34 + T cells
  • MFE23.CD28.CD40 CoStAR further enhanced expansion ( FIG. 11 ).
  • T cells mock transduced or transfected with MFE23.CD28 or MFE23.CD28.CD40 were cocultured with LoVo-OKT3 cells at an 8:1 effector:target ratio in the presence (200 IU/ml) or absence of exogenous IL-2.
  • days 1, 4, 7, 11 and 18 cells were taken and the number of viable T-cells enumerated by using anti-CD2 reagents on a MACSQuant flow cytometer.
  • FIG. 12A In the absence of stimulation by tumor and IL-2, cells declined in number as would be expected
  • FIG. 12B In the absence of stimulation but presence of IL-2 there was a more apparent survival of the cells, but no specific growth.
  • Mock transduced and T cells transduced with MFE23.CD28 or MFE23.CD28.CD40 CoStARs were then tested for cytokine production.
  • Bead array analysis was performed on supernatants obtained from T-cell/tumor cocultures.
  • Engineered T-cells were incubated at a 1:1 effector:target ratio with LoVo-OKT3 cells for 24 hours and supernatant collected.
  • Conditioned supernatant was also collected from an equal number of T-cells alone, or LoVo-OKT3 cells alone.
  • IL-2 IFN- ⁇ , TNF ⁇ , IL-4, IL-5, IL-13, IL-17A, IL-17F, IL-22, IL-6, IL-10, IL-9, and IL-21 was analysed using a LegendplexTM Human TH1/TH2 cytokine panel (Biolegend) ( FIGS. 13A-13M ). Cytokines were either very low or undetectable in media from T-cells or tumor alone. However when cocultured with tumor cytokine production was enhanced. MFE23.CD28 enhanced production of IL-2, IL-5, IL-17A/17F, IL-10, IL-9 and IL-21 compared to mock.
  • MFE23.CD28.CD40 also enhanced production of TNF ⁇ , IL-13 and IL-22.
  • MFE23.CD28.CD40 also enhanced the production of a number of cytokines greater than that elicited by MFE23.CD28 (IL-2, IL-9 and IL-17F), but also reduced the production of some cytokines below the levels seen with MFE23.CD28 (IL-5 and IL-10).
  • Chemokines were either very low or undetectable in media from T-cells alone. When cocultured with tumor, chemokine production was enhanced. MFE23.CD28 enhanced production of CXCL5, CXCL10, CXCL11, CCL17 and CCL20 compared to mock. However, MFE23.CD28.CD40 enhanced production of CCL2, CXCL1 and CXCL9.
  • MFE23.CD28.CD40 also further enhanced the production of certain cytokines to a greater amount than that elicited by MFE23.CD28 (CXCL1, CXCL9, CXCL10, CXCL11, CCL17, CCL2, CXCL9, CCL5 and CCL20), while reducing the production of some cytokines below the levels seen with MFE23.CD28 (CCL4).
  • CoStARs were tested for functional activity against cancer targets.
  • Cells were transduced with CD28 or CD28.CD40 CoStARs engineered with an scFv binding domain specific for FolR or CA125 (scFv MOV19 and scFv 196-14 respectively).
  • Human folate receptor alpha represents a suitable target for a number of tumors including ovarian, head and neck, renal and lung and CA125 represents an alternative target for ovarian cancer.
  • Primary human T-cells from six healthy donors were engineered with either 196-14.CD28, 196-14.CD28.CD40, MOV19.CD28 or MOV19.CD28.CD40 receptors, all harboring a DYKDDDDK epitope tag for detection.
  • Transduced cells were mixed with FolR+/CA125+ OvCAR-OKT3 cells before analysis of effector activity using intracellular staining in the epitope tag positive and negative populations.
  • Specific enhancement of effector activity determined by production of IL-2 ( FIGS. 15A and 15B ), TNF ⁇ ( FIGS. 15C and 15D ), CD137 ( FIGS. 15E and 15F ), and BCL-xL ( FIGS. 15G and 15H ) was observed in CD28 and CD28.CD40 engineered cells compared to mock transduce cells in response to both CA125 and FolR, although specific BCL-xL induction by MOV19.CD28 was not substantial as compared to MOV19.CD28.CD40.
  • FIGS. 16A-16F Mock transduced TILs or TILs engineered with MOV19.CD28.CD40 CoStAR were evaluated for expansion and CD137 production stimulated by patient matched tumor digest.
  • FIGS. 16A-16F Three donor tumors were tested which displayed varying levels of FolR on the digest, ranging from negative ( FIG. 16A ), low expression ( FIG. 16B ) to high expression ( FIG. 16C ).
  • Mock and CoStAR negative TIL in the CoStAR engineered populations of TIL matched for the FolR negative digest demonstrated similar levels of CD137 upregulation following tumor coculture which was not enhanced by the presence of CoStAR ( FIG. 16D ).
  • FIG. 17A A FolR targeting CoStAR was examined for enhancement of effector functions.
  • MOV19.CD28.CD40 enhanced CD137 expression from ⁇ 20% to ⁇ 50% ( FIG. 17A ), TNF ⁇ production from 10% to 15% ( FIG. 17B ) and IL-2 production from 2% to 5% ( FIG. 17C ) in response to FolR+ tumor digest.
  • CoStAR mediated stimulation by soluble ligand was also examined.
  • T-cells from three healthy donors were engineered with MOV19.CD28 or MOV19.CD28.CD40 CoStAR and activated with either immobilised OKT3, providing stimulation in the absence of FolR, or with OvCAR-OKT3, to provide TCR and CoStAR activity.
  • Bcl-XL activity was increased from between 10 and 20% across the three donors following OKT3 stimulation ( FIG. 18A ) whereas IL-2 was increased between 0 and 12% ( FIG. 18B ) and TNF ⁇ increased between 0 and 20% (FIG. 18 C).
  • the presence of exogenous soluble FolR did not enhance any of these particular effector functions.
  • the MFE23, MOV19 and 196-14 CoStAR constructs include an MFE23 (CEA specific), MOV19 (Folate receptor a specific) or 196-14 (CA125 specific) derived single chain antibody fragment nucleotide sequence with an oncostatin M1 leader sequence fused to a costimulatory domain.
  • the costimulatory domains contain an extracellular spacer region and transmembrane domain derived from human CD8 or CD28 and a signalling domain of either CD28, CD2 or CD137 and/or wild-type or mutant CD40 variants.
  • Some CoStARs detailed herein comprise a human PD1 extracellular domain fused to CD28 and CD40.
  • Receptors were cloned with a P2A cleavage sequence and a truncated form of human CD34 to permit detection of transduced cells.
  • the CoStAR nucleotide sequence was codon optimised and gene synthesised by Genewiz Inc.
  • the constructs were cloned into a third generation lentiviral vector.
  • Peripheral blood mononuclear cells were isolated from normal healthy donors before activation for 24 hours with T-cell activation and expansion beads (Invitrogen) according to the manufacturer's instructions before addition of lentiviral supernatants.
  • Cell transduction was assessed 96 hours post infection using CEA.hFc protein (R&D Systems) and anti-hFc-PE secondary, plus anti-CD34-APC or by anti-CD34-PE antibodies alone. Cells were then expanded further using ⁇ 10 donor mismatched irradiated PBMC feeders at a 1:20-1:200 ratio in RPMI+10% FCS with the addition of 30 ng/ml OKT3 and 200 IU/ml IL-2. After 14 days the cells were stained as previous and stored ready for assay.
  • Functionality assays were performed by mixing CoStAR positive or negative cells with wild-type or OKT3 engineered CEA-Positive LoVo cells. Briefly, T-cells were mixed with LoVo cells at varying ratios in 96-well plates. For flow analysis cocultures were incubated with Brefeldin and monensin and anti-CD107a antibodies for 16 hours following which cells were stained with Fixable Viability Dye ef450 (eBiosciences), fixed with 4% paraformaldehyde and then permeabilised using Fix/Perm wash buffer (BD Biosciences).
  • Fixable Viability Dye ef450 eBiosciences
  • cytokine bead array LGENDPLEXTM Human Th Cytokine Panel (12-plex)
  • chemokine bead array LGENDPLEXTM Human Proinflammatory Chemokine Panel (13-plex).
  • Proliferation assays were performed by mixing T-cells and tumor cells at an 8:1 effector:target ratio in complete T-cell media (TCM: RPMI supplemented with 10% FCS, 0.01 M HEPES and 1% Penicillin/streptomycin, 50 mM 3-mercaptoethanol) in the presence or absence of IL-2. Cell counts were made at indicated time points and fresh tumor cells were added in restimulation assays at a final E:T of 8:1. Cell counts for proliferation assays were performed by taking cells from the wells and staining with anti-CD2 PerCP eFluor710 antibody (eBioscience, UK) for 20 min in the dark, followed by DRAQ7 staining and counts made using a MACSQuant analyser.
  • MOV19 is a single chain Fv antibody that has a high affinity for Folate Receptor alpha (FOLR1).
  • Immunocompromised mice are implanted with an established ovarian cancer cell line (A2870, OVCAR-5, OVCAR-8 or SK-OV-3), which is allowed to grow in the animal for few days.
  • mice are subsequently staged according to their tumor burden, and finally injected with the mock transduced T cells or MOV19.CD28.CD40 transduced T cells.
  • some of the mice are injected with intravenous IL-2 (5 ⁇ g IL-2, Q2Dx7) to support the engraftment and initial expansion of T cells.
  • the final study design contains 5 groups (each one containing 5 mice): PBS (no cells dosed), mock transduced T cells, mock transduced T cells with IL-2 supplementation, MOV19.CD28.CD40 transduced T cells and MOV19.CD28.CD40 transduced T cells with IL-2 supplementation. Tumor growth and mice survival is monitored on weekly basis for a total of 40 days.
  • mice administered with MOV19.CD28.CD40 transduced cells show better tumor control and prolonged survival compared to the mock transduced groups, whether or not supplemented with IL-2.
  • This data demonstrates the ability of the CoStAR platform to improve in vivo the T cell anti-tumor response and also illustrates how this improved response is independent of the presence of exogenous IL-2.
  • the example relates to identification of key components of CoStAR such as, but not limited to, PD-1, MFE23, CD40 combined with another component, a spacer, a CD40 mutant and/or a CD28 mutant.
  • key components of CoStAR such as, but not limited to, PD-1, MFE23, CD40 combined with another component, a spacer, a CD40 mutant and/or a CD28 mutant.
  • Virus production was carried out by CaCl 2 ) transfection of HEK293T cells.
  • CD34 a marker gene expression was determined by titration with JRT3 cells.
  • Day 0 was T cell isolation from frozen PBMCs. Day 0 was also activation with Dynabeads. Day 2 was transduction by spinoculation. Day 5 was bead removal. Day 8 was measuring viability and transduction rate. Day 8 was also post activation (before REP), Days 13-15 was freezing.
  • An experimental design in healthy donors included the outgrowth as described above as well as REP: Day ⁇ 2 was transduced T cells thawing, Day ⁇ 1 was magnetic CD34 enrichment, Day 0 was REP with G-Rex, Day 5-6 was changing medium, Day 11-12 was measuring viability and transduction rate and freezing.
  • CD40 CoStAR modified T cells were enriched in CD4 after CD34 enrichment and REP ( FIG. 20 ).
  • CD4 and CD8 T cell phenotypes were assessed 10-11 days after REP using anti-human CD4-PerCP-eF710, anti-human CD8-PE-Cy7, and anti-human CD3-FITC. Analysis was performed by flow cytometry (Novocyte) and data were analyzed using NovoExpress 1.5.0 software with the following gating strategy: live/dead exclusion, single cells, CD3+ cells, CD4+ cells or CD8+ cells.
  • CoStAR modified CD4 T cells were highly transduced compared to the CD8 population. Healthy donors were activated with Dynabeads and transduced (spinoculation, MOI 5) with CD40 CoStAR constructs or MOCK. Cells were then magnetically enriched for their CD34 expression and expanded following the rapid expansion protocol (REP). Surface expression of the marker gene CD34 on CD4 and CD8 T cells, was assessed 10-11 days after REP using anti-human CD34-PE associated with anti-human CD4-PerCP-eF710, anti-human CD8-PE-Cy7, and anti-human CD3-FITC.
  • CoStARs composed of an extracellular checkpoint binding domain fused to a CD40 costimulatory domain could convert an inhibitory signal into an activating signal upon engagement of the CoStAR.
  • PD1-fusion CoStARs based on the descriptions outlined in Ankri et al. J Immunol 2013; 191:4121-4129 and Prosser et al. Molecular Immunology 51 (2012) 263-272, but with the addition of CD40 to the signalling domain ( FIG. 21A ).
  • Transduced T-cells were enriched using CD34 microbeads and expanded via a rapid expansion protocol using irradiated feeder cells before banking. After thaw, cells were rested for 3-4 days in complete RPMI supplemented with IL-2. The viability and absolute count were assessed after overnight IL-2 starvation using DRAQ-7 (1:200) by flow cytometry (Novocyte) and data were analysed using the NovoExpress 1.5.0 software.
  • Transduced T cells were cocultured in the absence of IL-2 with LoVo (CCL-229TM) or LoVo.OKT3.GFP tumor cells at 8:1 effector to target ratio. After 24 hours, supernatants were collected and frozen. LoVo and LoVo.OKT3.GFP naturally express CEA and PD-L1 on their surface, conferring signal 2 through the CoStAR alone (LoVo) or associated with signal 1 (LoVo.OKT3.GFP) to the transduced T cells. Cocultures were performed in triplicate and corresponding negative (T cells alone, tumor cells alone) and positive (PMA+ionomycin) controls were included in the experiment.
  • FIG. 22 shows the ability of PD1-fusion CoStARs to mediate T-cell survival in the presence of repeated tumor challenge.
  • CoStAR or mock transduced T-cells were mixed at 8:1 E:T ratio with LoVo-OKT3 cells at day 0 and 7 and counts and checkpoint expression phenotyping made at day 6-8 and 14-15 ( FIGS. 23A-23C ).
  • FIG. 22 shows the fold expansion of cells over the duration of the experiment, with mock transduced cells dropping in number throughout the experiment; conversely MFE23.CD28.CD40 (CTP194) engineered cells expanded upon serial stimulation with tumor up to 12-fold by day 14.
  • TIM3 mirrored LAG3 expression in CD4+ and CD8+ cells at day 6-8, whereas at day 14-15 TIM3 expression was generally low, however we did observe a high expression of TIM3 in mock transduced cells at day 14-15 ( ⁇ 80% of cells), which was lower in PD1-fusion CoStAR cells, but ⁇ 20% in cells harboring MFE23.CD28.CD40.
  • CoStARs consisting of an antigen recognition domain which inverts signals, such as PD1, are functional but do not perform as well in cytokine release or expansion assays as cells harboring CoStAR with an scFv-based antigen recognition domain.
  • PD1-fusion receptors can also modulate checkpoint expression compared to mock engineered cells as well.
  • CD40 may operate as a single component of the CoStAR, or in combination with costimulatory domains other than CD28.
  • the first receptor consists of a CD28.CD40 signalling domain with CD8 extracellular and transmembrane domain (CTP190), CD2.CD40 signalling domain with CD8 extracellular and transmembrane domain (CTP191), or CD137.CD40 signalling domain with CD8 extracellular and transmembrane domain (CTP192).
  • FIG. 24A Flow cytometric analysis of transduced cells showed that expression of these receptors did not correlate well with CD34 marker gene expression, suggesting that the structural formats did not permit efficient surface expression ( FIG. 24A , lower left panel). Nonetheless we conducted functionality assays and showed that the CD28.CD40, CD137.CD40 and CD40 receptors could mediate enhanced IL-2 secretion compared to mock transduced cells in LoVo-OKT3 cocultures, whereas IFN ⁇ secretion was lower than from mock transduced cells, and MFE23.CD28.CD40 engineered cells ( FIG. 24B ). Analysis of expansion in the presence of LoVo-OKT3 cells ( FIG.
  • LAG3 was present on ⁇ 50% of CD4+ mock cells but ⁇ 20% of engineered cells (data unavailable for some receptors due to insufficient cell numbers to analyse).
  • PD1 expression was again ⁇ 20% on CD4+ and CD8+ cells at both time points analysed except for the CD2.CD40 CTP191 engineered cells at day 6-8 and mock transduced cells at the later time point.
  • TIM3 expression was generally low in CD4+ cells at both timepoints analysed, but higher in CD8+ cells, in particular in CD137.CD40 (CTP192) and CD40 (CTP193) engineered cells.
  • IL-2 production from mock transduced cells was below the level of detection.
  • IL-2 production from the control CTP194 receptor was approximately 4000 pg/ml, as was production from CTP195 harboring the SVQE-AVQA mutations, and cells harboring the CTP198 receptor with the P227A polymorphism.
  • FIGS. 29A-29B Phenotypic analysis of cell expressing these different mutations was also conducted ( FIGS. 29A-29B ). No clear differences were seen in the relative expression of LAG3 in CD4+ or CD8+ cells at days 6-8 between transduced and non-transduced cells, However mock transduced cells had higher LAG3 expression at days 14-15 compared to CD4+ cells expressing any of the CoStARs. No differences were observed with regards LAG3 expression in CD8+ cells at days 14-15. PD1 expression was found to be ⁇ 20% on average at days 6-8 for all receptor engineered CD4+ cells, with higher expression in mock engineered cells.
  • TIM3 expression was found to be lower than 20% on average in all CD4+ cell groups at both time points analysed. Expression was generally more variable in CD8+ cells at the first time point, with an average of approximately 30%, although slightly higher in cells expressing CTP196. At days 14-15 mock transduced cells had considerably higher TIM3 expression than transduced cells and cells expressing CTP196 had approximately twice as much TIM3 expression than cells from other groups.
  • CTP201 contains a PYAP-AYAA mutation
  • CTP202 contains a YMNM-FMNM mutation.
  • CTP203 has an extended IgG4 hinge into this cohort to establish whether CoStARs containing a longer linker domain maintain functionality (CTP203) ( FIG. 30A ).
  • CTP203 shows that CoStARs containing a longer linker domain maintain functionality.
  • IL-2 production from mock or transduced T-cells was assessed following coculture with LoVo-OKT3 cells ( FIG. 30B ).
  • IL-2 from CTP194 expressing cells was approximately 4000 pg/ml and lower for the CD28 mutant receptors, both being approximately 2500 pg/ml.
  • IL-2 from IgG4 hinge receptor expressing cells was lower at approximately 1000 pg/ml.
  • As a control IL-2 from mock transduced cells was below the lower level of detection.
  • IFN ⁇ from CTP194 was approximately 1000 pg/ml, as was IFN ⁇ from CTP201 cells harboring the PYAP-AYAA CD28 mutation.
  • IFN ⁇ secretion from cells expressing the YMNM-FMNM mutation.
  • the production of IFN ⁇ was highest from cells expressing the CTP203 IgG4 hinge receptor.
  • LAG3 expression was higher in CD4+ mock cells at days 14-15 at approximately 50%+, compared to an average of 10% or lower in transduced cells, and was also higher in CD8+ mock cells compared to transduced cells.
  • PD1 expression analysis revealed approximately 10% expression in MFE23.CD28.CD40, or CD28 mutant CD4+ cells at the first time point, whereas cells expressing the IgG4 receptor had >20% PD1 expression, as did mock transduced cells. At days 14-15 the difference was greater still with 100% of mock CD4+ cells being PD1+.
  • CD8+ cells demonstrated low PD1 positivity at both time points.
  • CD4 or CD8+ TIM3 expression at days 6-8, however at days 14-15 CD4+ cells expressing the IgG4 spacer domain receptor showed higher PD1 positivity compared to cells expressing the control or CD28 mutant receptors, with a similar effect observed in CD8+ cells.
  • T cells were thawed one day prior to coculture, resuspended at 1 ⁇ 10 6 cells/mL in TCM without IL-2, and incubated overnight at 37° C. with 5% CO 2 .
  • T cells and BA/F3 targets ie, WT, OKT3, FOLR1, and OKT3-FOLR1 were collected and counted using a ViCELL BLU per manufacturer's instructions.
  • Non-Td and Td T cells were preincubated for 30 minutes at room temperature with a range of solFOLR1 (ie, 0, 20, 60 and 200 ng/mL) concentrations that represent concentrations reported in ovarian cancer patient serum as well as supraphysiological levels. Following incubation, cells were cocultured with either BA/F3 WT, OKT3, FOLR1, or OKT3-FOLR1 targets at the following E:T ratios (3:1, 1:1, 1:3) overnight. Each condition was performed in duplicates. T cells stimulated with PMA/ionomycin as per manufacturer's instructions and unstimulated T cells served as positive and negative controls, respectively. Following overnight, plates were collected and centrifuged at 500 ⁇ g for 3 minutes. 100 ⁇ L of supernatant was collected from each well and stored at ⁇ 80° C. prior to analysis of cytokine content. The remaining cells in plates were then stained as described below.
  • solFOLR1 ie, 0, 20, 60 and 200 ng/mL
  • T cells were first labeled with CellTraceTM Violet Dye on the day of coculture setup, according to the manufacturer's instructions. The labeled cells were then cultured for 5 days with BA/F3 target cells (BA/F3, BA/F3-FOLR1, BA/F3-OKT3-FOLR1) at a E:T of 10:1.
  • BA/F3 target cells BA/F3, BA/F3-FOLR1, BA/F3-OKT3-FOLR1
  • Lymphocytes (forward scatter [FSC]—A vs side scatter [SSC]-A)
  • Tumor vs T cells (anti-mouse CD45 BV785 vs anti-human CD45 BV650)
  • Activation markers 4-1BB FSC-H vs anti-human 4-1BB BV421) and CD69 (FSC-H vs anti-human CD69 BV711) gated specifically from T cells
  • IL-2 cytokine production
  • IL-2 cytokine production
  • NTD non-transduced
  • CoStAR anti-FOLR1 CoStAR modified T cells
  • TIL from 6 ovarian tumors were liberated by digestion and cultured in 3000 U IL-2.
  • Transduction with a 3rd generation lentiviral vector encoding a CoStAR molecule with and scFv targeting human FOLR1, linker, full length CD28 fused to truncated CD40 cytoplasmic domain was carried out at an MOI of 5, both 48 h and 72 h after tumor digestion.
  • Flow cytometric analysis was used to determine the frequency of CD4 and CD8 T-cells expressing the CoStAR Molecule using an anti-idiotype antibody for surface detection. About 20% to 70% of CD4 and CD8 T-cells expressed the CoStAR molecule ( FIG. 37A ).
  • CoStAR modified TIL from 6 ovarian tumors were co-cultured with autologous digest overnight in the presence of brefeldin A.
  • the frequency of cells expressing IL-2 or TNF ⁇ was assessed the following day by flow cytometry.
  • the frequency of TIL reacting to autologous digest is enhanced by the CoStAR molecule ( FIG. 38A ).
  • CoStAR modified TIL from 6 ovarian tumors were co-cultured with autologous digest and supernatant assessed for cytokine release.
  • CoStAR modified cells had increased effector functions as demonstrated by increased IFN ⁇ , TNF ⁇ and IL-13 release. Maximal levels of these molecules was similar in response to stimulation with PMA (Phorbol 12-myristate 13-acetate) and ionomycin ( FIG. 38B ).
  • CoStAR modified TIL from 5 ovarian tumors were co-cultured with BA/F3 cells or BA/F3 cells engineered to express OKT3, FOLR or both. Cytokine secretion of non-modified and CoStAR modified TIL was equivalent when co-cultured with non-modified BA/F3 or BA/F3 expressing OKT3 alone or FOLR1 alone. CoStAR modified TIL secreted increased levels of cytokines IL-2 and IFN ⁇ when co-cultured with BA/F3 modified to express both FOLR1 and OKT3 ( FIG. 39A )
  • CoStAR modified TIL from 5 ovarian tumors were co-cultured with BA/F3 cells or BA/F3 cells engineered to express OKT3, FOLR or both. Cytotoxicity towards BA/F3 target cells was assessed via cell counts, determined by flow cytometric analysis of mouse CD45. Non-modified and CoStAR modified cells killed target cells expressing OKT3 equivalently. CoStAR modified TILs do not kill BA/F3 cells expressing FOLR1 alone ( FIG. 39B ).
  • Mock or CoStAR modified TIL from 3 ovarian cancer patients were co-cultured with autologous tumor in the presence of no blocking, MHCI, MHC II or MHC I+MHC II blocking or antibodies or isotype control. Supernatant was assessed for the level of IFN ⁇ release. Normalized to levels of release without antibody, IFN ⁇ levels are similarly reduced in mock and CoStAR modified TIL, showing that activity is led by endogenous TCR-MHC peptide interactions ( FIG. 39C ).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Oncology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Hematology (AREA)
  • Pregnancy & Childbirth (AREA)
  • Gynecology & Obstetrics (AREA)
  • Reproductive Health (AREA)
  • Virology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
US17/843,480 2020-07-17 2022-06-17 Receptors providing targeted costimulation for adoptive cell therapy Pending US20220348631A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US17/843,480 US20220348631A1 (en) 2020-07-17 2022-06-17 Receptors providing targeted costimulation for adoptive cell therapy
US17/823,223 US20230002470A1 (en) 2020-07-17 2022-08-30 Receptors providing targeted costimulation for adoptive cell therapy

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US202063053494P 2020-07-17 2020-07-17
PCT/US2021/042075 WO2022016112A1 (en) 2020-07-17 2021-07-16 Receptors providing targeted costimulation for adoptive cell therapy
US17/843,480 US20220348631A1 (en) 2020-07-17 2022-06-17 Receptors providing targeted costimulation for adoptive cell therapy

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/042075 Continuation WO2022016112A1 (en) 2020-07-17 2021-07-16 Receptors providing targeted costimulation for adoptive cell therapy

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/823,223 Continuation US20230002470A1 (en) 2020-07-17 2022-08-30 Receptors providing targeted costimulation for adoptive cell therapy

Publications (1)

Publication Number Publication Date
US20220348631A1 true US20220348631A1 (en) 2022-11-03

Family

ID=77301001

Family Applications (2)

Application Number Title Priority Date Filing Date
US17/843,480 Pending US20220348631A1 (en) 2020-07-17 2022-06-17 Receptors providing targeted costimulation for adoptive cell therapy
US17/823,223 Pending US20230002470A1 (en) 2020-07-17 2022-08-30 Receptors providing targeted costimulation for adoptive cell therapy

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/823,223 Pending US20230002470A1 (en) 2020-07-17 2022-08-30 Receptors providing targeted costimulation for adoptive cell therapy

Country Status (10)

Country Link
US (2) US20220348631A1 (zh)
EP (1) EP4182338A1 (zh)
JP (1) JP2023534808A (zh)
KR (1) KR20230088333A (zh)
CN (1) CN116348485A (zh)
AR (1) AR123004A1 (zh)
AU (1) AU2021308702A1 (zh)
CA (1) CA3189671A1 (zh)
IL (1) IL299886A (zh)
WO (1) WO2022016112A1 (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11697677B2 (en) 2021-07-16 2023-07-11 Instil Bio (Uk) Limited Chimeric molecules providing targeted costimulation for adoptive cell therapy
WO2023141530A3 (en) * 2022-01-20 2023-09-21 Instil Bio (Uk) Limited Receptors providing targeted costimulation for adoptive cell therapy
US11945876B2 (en) 2021-06-16 2024-04-02 Instil Bio (Uk) Limited Receptors providing targeted costimulation for adoptive cell therapy

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201700621D0 (en) 2017-01-13 2017-03-01 Guest Ryan Dominic Method,device and kit for the aseptic isolation,enrichment and stabilsation of cells from mammalian solid tissue
KR20220119439A (ko) 2019-12-20 2022-08-29 인스틸 바이오 유케이 리미티드 종양 침윤 림프구를 분리하기 위한 장치 및 방법 및 그것의 용도
US20240058447A1 (en) * 2022-05-25 2024-02-22 Instil Bio (Uk) Limited Use of fusion constructs for il-2 independent t cell therapy
WO2024030758A1 (en) * 2022-08-01 2024-02-08 Iovance Biotherapeutics, Inc. Chimeric costimulatory receptors, chemokine receptors, and the use of same in cellular immunotherapies

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7507412B2 (en) * 2001-07-18 2009-03-24 Merck Patent Gmbh Glycoprotein VI fusion proteins
WO2016126608A1 (en) * 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
US20180319862A1 (en) * 2015-11-05 2018-11-08 Juno Therapeutics, Inc. Chimeric receptors containing traf-inducing domains and related compositions and methods

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR901228A (fr) 1943-01-16 1945-07-20 Deutsche Edelstahlwerke Ag Système d'aimant à entrefer annulaire
US5858358A (en) 1992-04-07 1999-01-12 The United States Of America As Represented By The Secretary Of The Navy Methods for selectively stimulating proliferation of T cells
US6534055B1 (en) 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US6905680B2 (en) 1988-11-23 2005-06-14 Genetics Institute, Inc. Methods of treating HIV infected subjects
US6352694B1 (en) 1994-06-03 2002-03-05 Genetics Institute, Inc. Methods for inducing a population of T cells to proliferate using agents which recognize TCR/CD3 and ligands which stimulate an accessory molecule on the surface of the T cells
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
US7175843B2 (en) 1994-06-03 2007-02-13 Genetics Institute, Llc Methods for selectively stimulating proliferation of T cells
US7067318B2 (en) 1995-06-07 2006-06-27 The Regents Of The University Of Michigan Methods for transfecting T cells
US6692964B1 (en) 1995-05-04 2004-02-17 The United States Of America As Represented By The Secretary Of The Navy Methods for transfecting T cells
AU1086501A (en) 1999-10-15 2001-04-30 Carnegie Institution Of Washington Rna interference pathway genes as tools for targeted genetic interference
US6326193B1 (en) 1999-11-05 2001-12-04 Cambria Biosciences, Llc Insect control agent
DK1257632T3 (da) 2000-02-24 2008-01-28 Xcyte Therapies Inc Samtidig stimulering og opkoncentrering af celler
US7572631B2 (en) 2000-02-24 2009-08-11 Invitrogen Corporation Activation and expansion of T cells
US6797514B2 (en) 2000-02-24 2004-09-28 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US6867041B2 (en) 2000-02-24 2005-03-15 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
AU2001275474A1 (en) 2000-06-12 2001-12-24 Akkadix Corporation Materials and methods for the control of nematodes
US7745140B2 (en) 2002-01-03 2010-06-29 The Trustees Of The University Of Pennsylvania Activation and expansion of T-cells using an engineered multivalent signaling platform as a research tool
JP5070045B2 (ja) 2004-05-27 2012-11-07 ザ・トラステイーズ・オブ・ザ・ユニバーシテイ・オブ・ペンシルベニア 新規人工抗原提示細胞およびそれらの用途
US20070036773A1 (en) 2005-08-09 2007-02-15 City Of Hope Generation and application of universal T cells for B-ALL
WO2011059836A2 (en) 2009-10-29 2011-05-19 Trustees Of Dartmouth College T cell receptor-deficient t cell compositions
WO2015112626A1 (en) * 2014-01-21 2015-07-30 June Carl H Enhanced antigen presenting ability of car t cells by co-introduction of costimulatory molecules
WO2018156711A1 (en) * 2017-02-22 2018-08-30 H. Lee Moffitt Cancer Center And Research Institute, Inc. Il13ra2-binding chimeric antigen receptors
GB201900858D0 (en) * 2019-01-22 2019-03-13 Price Nicola Kaye Receptors providing targeted costimulation for adoptive cell therapy

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7507412B2 (en) * 2001-07-18 2009-03-24 Merck Patent Gmbh Glycoprotein VI fusion proteins
WO2016126608A1 (en) * 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
US20180319862A1 (en) * 2015-11-05 2018-11-08 Juno Therapeutics, Inc. Chimeric receptors containing traf-inducing domains and related compositions and methods

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Kandalaft et al .A phase I clinical trial of adoptive transfer of folate receptor-alpha redirected autologous T cells for recurrent ovarian cancer(Journal of Translational Medicine (2012 10:157). (Year: 2012) *
Lanitis et al. Chimeric Antigen Receptor T Cells with Dissociated Signaling Domains Exhibit Focused Antitumor Activity with Reduced Potential for Toxicity In Vivo(Cancer Immunology Research; 1(1) July 2013) (Year: 2013) *
Lee et al.A mechanical switch couples T cell receptor triggering to the cytoplasmic juxtamembrane regions of CD3ζζ.Immunity 2015 August 18; 43(2): 227-239. Doi:10.1016. (Year: 2015) *
Lee supplemental. (Year: 2015) *
Levin et al . Potent Activation of Human T Cells by mRNA Encoding Constitutively Active CD40 (Journal Immunology, (2018) 201 (10): 2959–2968) (Year: 2018) *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11945876B2 (en) 2021-06-16 2024-04-02 Instil Bio (Uk) Limited Receptors providing targeted costimulation for adoptive cell therapy
US11697677B2 (en) 2021-07-16 2023-07-11 Instil Bio (Uk) Limited Chimeric molecules providing targeted costimulation for adoptive cell therapy
WO2023141530A3 (en) * 2022-01-20 2023-09-21 Instil Bio (Uk) Limited Receptors providing targeted costimulation for adoptive cell therapy

Also Published As

Publication number Publication date
AU2021308702A1 (en) 2023-03-16
WO2022016112A1 (en) 2022-01-20
JP2023534808A (ja) 2023-08-14
IL299886A (en) 2023-03-01
KR20230088333A (ko) 2023-06-19
WO2022016112A8 (en) 2022-02-24
AR123004A1 (es) 2022-10-19
CA3189671A1 (en) 2022-01-20
CN116348485A (zh) 2023-06-27
EP4182338A1 (en) 2023-05-24
US20230002470A1 (en) 2023-01-05

Similar Documents

Publication Publication Date Title
US20220160760A1 (en) Receptors providing targeted costimulation for adoptive cell therapy
US20220348631A1 (en) Receptors providing targeted costimulation for adoptive cell therapy
KR102483822B1 (ko) 태그된 키메라 이펙터 분자 및 그의 리셉터
US20190375815A1 (en) Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
JP2021525509A (ja) 細胞療法のための多様な抗原結合ドメイン、新規プラットフォームおよびその他の強化
US20230055694A1 (en) Receptors providing targeted costimulation for adoptive cell therapy
US20230277670A1 (en) Chimeric molecules providing targeted costimulation for adoptive cell therapy
US20230348854A1 (en) Chimeric antigen receptors (cars) targeting natural killer cells
US20230227576A1 (en) Receptors providing targeted costimulation for adoptive cell therapy
US20230331808A1 (en) Chimeric molecules providing targeted costimulation for adoptive cell therapy
CN115485369A (zh) γδT细胞及其用途
TW202216751A (zh) 用於過繼細胞療法之提供靶向共刺激之受體
US20240058447A1 (en) Use of fusion constructs for il-2 independent t cell therapy
WO2023141530A2 (en) Receptors providing targeted costimulation for adoptive cell therapy
WO2024123835A1 (en) Engineered switches for immune cell activity and methods of use thereof
TW202216752A (zh) 用於過繼細胞療法之提供靶向共刺激之嵌合分子

Legal Events

Date Code Title Description
AS Assignment

Owner name: INSTIL BIO INC., TEXAS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:RODRIGUEZ, RUBEN;REEL/FRAME:061549/0659

Effective date: 20221003

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

Free format text: NON FINAL ACTION MAILED

AS Assignment

Owner name: INSTIL BIO (UK) LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BRIDGEMAN, JOHN;HAWKINS, ROBERT;KUEBERUWA, GRAY;SIGNING DATES FROM 20221004 TO 20221011;REEL/FRAME:061801/0061

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: FINAL REJECTION MAILED

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER