US20210292429A1 - Anti-liv1 immune cell cancer therapy - Google Patents

Anti-liv1 immune cell cancer therapy Download PDF

Info

Publication number
US20210292429A1
US20210292429A1 US17/291,198 US201917291198A US2021292429A1 US 20210292429 A1 US20210292429 A1 US 20210292429A1 US 201917291198 A US201917291198 A US 201917291198A US 2021292429 A1 US2021292429 A1 US 2021292429A1
Authority
US
United States
Prior art keywords
cells
engineered
cell
population
car
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
US17/291,198
Inventor
Jonathan Alexander Terrett
Jason Sagert
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.)
CRISPR Therapeutics AG
Original Assignee
CRISPR Therapeutics AG
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 CRISPR Therapeutics AG filed Critical CRISPR Therapeutics AG
Priority to US17/291,198 priority Critical patent/US20210292429A1/en
Assigned to CRISPR THERAPEUTICS AG reassignment CRISPR THERAPEUTICS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SAGERT, JASON, TERRETT, JONATHAN ALEXANDER
Publication of US20210292429A1 publication Critical patent/US20210292429A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • 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
    • C07K16/3015Breast
    • 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
    • A61K2239/49Breast
    • 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
    • A61K2239/56Kidney
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • A61K39/4632T-cell receptors [TCR]; antibody T-cell receptor constructs
    • 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
    • A61K39/464411Immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4717Plasma globulins, lactoglobulin
    • 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
    • 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
    • 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
    • C07K14/70517CD8
    • 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
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • 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
    • 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
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • A61K2039/812Breast
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • A61K2039/868Vaccine for a specifically defined cancer kidney
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2121/00Preparations for use in therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • 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
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/30Coculture with; Conditioned medium produced by 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
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/99Coculture with; Conditioned medium produced by genetically 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
    • 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/80Vectors containing sites for inducing double-stranded breaks, e.g. meganuclease restriction sites

Definitions

  • Chimeric antigen receptor (CAR) T-cell therapy uses genetically-modified T cells to more specifically and efficiently target and kill cancer cells. After T cells have been collected from the blood, the cells are engineered to include CARs on their surface. The CARs may be introduced into the T cells using CRISPR/Cas9 gene editing technology. When these allogeneic CAR T cells are injected into a patient, the receptors enable the T cells to kill cancer cells.
  • CAR Chimeric antigen receptor
  • LIV1 a member of the ZIP family of highly conserved transmembrane zinc transporter proteins, is expressed at elevated levels in estrogen receptor-positive breast cancer and tumors of the lymph nodes. Further aberrant expression of zinc transporters such as LIV1 is known to lead to deregulated Zn intake or deficiency, leading to uncontrolled growth such that occur in cancer. Thus, LIV1 is a desirable transmembrane protein for targeting cancer. In fact, the LIV-1 protein has been implicated in breast cancer, prostate cancer, squamous tumors, and neuronal tumors.
  • an engineered T cell comprising a nucleic acid encoding a chimeric antigen receptor (CAR), wherein the CAR comprise an ectodomain that binds specifically to LIV1.
  • the engineered T cell further comprises a disrupted T cell receptor alpha chain constant region (TRAC) gene.
  • TRAC T cell receptor alpha chain constant region
  • the TRAC gene may be disrupted by insertion of the nucleic acid encoding a CAR.
  • the engineered T cell further comprises a disrupted beta-2-microglobulin ( ⁇ 2M) gene.
  • the ectodomain of the CAR comprises an anti-LIV1 antibody.
  • the anti-LIV1 antibody is an anti-LIV1 single-chain variable fragment (scFv).
  • the anti-LIV1 scFv in some embodiments, comprises an amino acid sequence of any one of SEQ ID NO: 54, 70, 83 or 86.
  • the anti-LIV1 scFv comprises a heavy chain variable region (VH) comprising an amino acid sequence of any one of SEQ ID NO: 55 or 90 and/or a light chain variable region (VL) comprising an amino acid sequence of any one of SEQ ID NO: 56 or 88.
  • the anti-LIV1 scFv comprises a VH comprising CDR amino acid sequences of SEQ ID NO: 57, SEQ ID NO: 58, and/or SEQ ID NO: 59; and/or the anti-LIV1 scFv comprises a VL sequence comprising CDR amino acid sequences of SEQ ID NO: 60, SEQ ID NO: 61, and/or SEQ ID NO: 62.
  • the CAR in some embodiments, comprises a CD3 ⁇ cytoplasmic signaling domain. In some embodiments, the CAR comprises a CD28 co-stimulatory domain or a 41BB co-stimulatory domain.
  • the TRAC gene comprises the nucleotide sequence of any one of SEQ ID NOs: 63, 64, 107, or 111, and/or wherein the CAR comprises the nucleotide sequence of any one of SEQ ID NOs: 49, 51, 104, or 108.
  • the disrupted ⁇ 2M gene comprises at least one nucleotide sequence selected from any one of SEQ ID NOs: 9-14.
  • a population of engineered T cells e.g., comprising a nucleic acid encoding an anti-LIV1 CAR
  • at least 25% or at least 50% of engineered T cells of the population express the CAR.
  • at least 15% or at least 50% of engineered T cells of the population express the CAR.
  • at least 70% of engineered T cells of the population express the CAR.
  • at least 30% of engineered T cells of the population express the CAR.
  • At least 25% of engineered T cells of the population express the CAR following at least 7 or at least 14 days of in vitro proliferation.
  • At least 50% of engineered T cells of the population do not express a detectable level of T cell receptor (TCR) protein.
  • TCR T cell receptor
  • at least 90% of engineered T cells of the population may not express a detectable level of TCR protein.
  • At least 50% of engineered T cells of the population do not express a detectable level of ⁇ 2M protein.
  • at least 70% of engineered T cells of the population may not express a detectable level of ⁇ 2M protein.
  • engineered T cells of the population when co-cultured in vitro with a population of cancer cells that express LIV1, induce cell lysis of at least 50% of the cancer cells of the population.
  • engineered T cells of the population may induce cell lysis of at least 70%, at least 80%, or at least 90% of the cancer cells of the population.
  • engineered T cells of the population when co-cultured in vitro with a population of cancer cells, secrete IFN ⁇ .
  • the ratio of engineered T cells to cancer cells is 1:1 to 2:1.
  • the cancer cells may be, for example, sarcoma cells or breast cancer cells. Other cancer cells may be targeted.
  • proliferative capacity of engineered T cells of the population is within 10% of proliferative capacity of control cells.
  • percent body weight of the subject is within 10% of initial body weight of the subject, wherein initial body weight of the subject is body weight of the subject at the time of administration.
  • the subject is a human subject.
  • the subject has a cancer.
  • the cancer may express LIV1, for example.
  • the method further comprises delivering to the T cell a gRNA targeting the ⁇ 2M gene.
  • the gRNA targeting the ⁇ 2M gene comprises the nucleotide sequence of SEQ ID NO: 20 or 21, or targets the nucleotide sequence of SEQ ID NO: 41.
  • the RNA-guided nuclease is a Cas9 nuclease, optionally a S. pyogenes Cas9 nuclease.
  • the donor template comprises the nucleotide sequence of any one of SEQ ID NOs: 63, 64, 107, or 111.
  • the CAR comprises the nucleotide sequence of any one of SEQ ID NOs: 49, 51, 104, or 108.
  • FIG. 1 shows flow cytometry results to assess TRAC, ⁇ 2M, and anti-Livia CAR expression levels at the cell surface of the edited cell population.
  • FIGS. 2A-2B show that anti-Livia CAR T cells, particularly those expressing the CTX971, CTX975 and CTX976 constructs, exhibited potent cytotoxicity towards the A498 ( FIG. 2A ) and ZR-75-1 ( FIG. 2B ) cell lines.
  • FIGS. 3A-3D show cytokine secretion of anti-Liv1a CAR T cells when co-cultured with target cell lines A498 and ZR-75-1.
  • FIGS. 3A and 3B show secretion of the effector cytokine interferon- ⁇ (IFN- ⁇ ) in A498 ( FIG. 3A ) and ZR-75-1 ( FIG. 3B ) cell lines.
  • FIGS. 3C and 3D show secretion of the effector cytokine interleukin-2 (IL-2) in A498 ( FIG. 3C ) and ZR-75-1 ( FIG. 3D ) cell lines.
  • IFN- ⁇ effector cytokine interferon- ⁇
  • FIGS. 3C and 3D show secretion of the effector cytokine interleukin-2 (IL-2) in A498 ( FIG. 3C ) and ZR-75-1 ( FIG. 3D ) cell lines.
  • IL-2 effector cytokine interleukin
  • FIG. 4 shows an alignment of scFV constructs (VL and VH)—971 (SEQ ID NO: 54); 973 (SEQ ID NO: 82); 975 (SEQ ID NO: 83); 977 (SEQ ID NO: 84)
  • FIG. 5 shows an alignment of scFV constructs (VH and VL)—979 (SEQ ID NO: 70); 974 (SEQ ID NO: 85); 976 (SEQ ID NO: 86); 978 (SEQ ID NO: 87), 972 (SEQ ID NO: 127)
  • the T cells of the present disclosure are engineered with a chimeric antigen receptor (CAR) designed to target LIV1.
  • CAR chimeric antigen receptor
  • LIV1 also known as Solute Carrier Family 39 Member 6, SLC39A6, ZIP6, and LIV-1, is a member of the ZIP family of highly conserved transmembrane zinc transporter proteins.
  • LIV1 is expressed at elevated levels in breast cancer, e.g., estrogen receptor-positive breast cancer, prostate cancer, squamous tumors, e.g., of the skin, bladder, lung, cervix, endometrium, head neck, and biliary tract, and neuronal tumors.
  • LIV1 has a restricted expression in normal tissues, e.g., non-cancerous breast, prostate, and testis.
  • LIV1 is a desirable transmembrane protein for targeting cancer.
  • the LIV-1 protein has been implicated in breast cancer, prostate cancer, squamous tumors, and neuronal tumors.
  • T cells of the present disclosure are engineered to express a CAR comprising an anti-LIV1 antibody (e.g., anti-LIV1 scFv).
  • the anti-LIV1 antibody is an anti-LIV1 scFv encoded by the sequence of any one of SEQ ID NOs: 53, 69, 97, 102, 106, 110, 114, or 118.
  • the anti-LIV1 antibody is an anti-LIV1 scFv comprising the sequence of any one of SEQ ID NOs: 54, 70, 82, 83, 84, 85, 86, or 87.
  • the anti-LIV1 antibody is an anti-LIV1 scFv comprising a VH comprising an amino acid sequence of any one of SEQ ID NO: 55, 90 or 98. In some embodiments, the anti-LIV1 antibody is an anti-LIV1 scFv comprising a VL comprising an amino acid sequence of any one of SEQ ID NO: 56, 88 or 128. In some embodiments, a CAR comprising an anti-LIV1 antibody is encoded by the sequence of any one of SEQ ID NOs: 49, 51, 65, 67, 95, 100, 104, 108, 112, or 116.
  • a CAR comprising an anti-LIV7 antibody is encoded by a sequence comprising a nucleic acid that is at least 90% identical to SEQ ID NOs: 49, 51, 65, 67, 95, 100, 104, 108, 112, or 116.
  • a CAR comprising an anti-LIV1 antibody comprises the sequence of any one of SEQ ID NOs: 49, 51, 65, 67, 95, 100, 104, 108, 112, or 116.
  • a CAR comprising an anti-LIV1 antibody comprises an anti-LIV1 antibody as described in U.S. Pat. No. 9,228,026.
  • an engineered T cell may comprise a disrupted T cell receptor alpha chain constant region (TRAC) gene, a disrupted beta-2-microglobulin ( ⁇ 2M) gene, a disrupted programmed cell death-1 (PD-1 or PDCD1) gene, a disrupted CD70 gene, or any combination of two or more of the foregoing disrupted genes.
  • an engineered T cell comprises a disrupted TRAC gene, a disrupted ⁇ 2M gene, and a disrupted CD70 gene.
  • an engineered T cell comprises a disrupted TRAC gene, a disrupted ⁇ 2M gene, and a disrupted PD-1 gene. In some embodiments, an engineered T cell comprises a disrupted TRAC gene, a disrupted ⁇ 2M gene, a disrupted CD70 gene and a disrupted PD-1 gene.
  • a disrupted gene is a gene that does not encode functional protein.
  • a cell that comprises a disrupted gene does not express (e.g., at the cell surface) a detectable level (e.g. by antibody, e.g., by flow cytometry) of the protein encoded by the gene.
  • a cell that does not express a detectable level of the protein may be referred to as a knockout cell.
  • a cell having ⁇ 2M gene edit may be considered a ⁇ 2M knockout cell if ⁇ 2M protein cannot be detected at the cell surface using an antibody that specifically binds ⁇ 2M protein.
  • populations of cells in which a certain percentage of the cells has been edited e.g., ⁇ 2M gene edited
  • at least 50% e.g., 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 85%
  • at least 50% of the cells (e.g. T cells) of the population do not express detectable levels of ⁇ 2M protein.
  • At least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% of the cells of a gene-edited population of cells may be ⁇ 2M knockout cells.
  • an engineered T cell comprises a disrupted TRAC gene. This disruption leads to loss of function of the TCR and renders the engineered T cell non-alloreactive and suitable for allogeneic transplantation, minimizing the risk of graft versus host disease.
  • expression of the endogenous TRAC gene is eliminated to prevent a graft-versus-host response.
  • a disruption in the TRAC gene expression is created by knocking a chimeric antigen receptor (CAR) into the TRAC gene (e.g., using an adeno-associated viral (AAV) vector and donor template).
  • a disruption in the TRAC gene expression is created by gRNAs targeting the TRAC genomic region.
  • a genomic deletion in the TRAC gene is created by knocking a chimeric antigen receptor (CAR) into the TRAC gene (e.g., using an AAV vector and donor template).
  • a disruption in the TRAC gene expression is created by gRNAs targeting the TRAC genomic region and knocking a chimeric antigen receptor (CAR) into the TRAC gene.
  • Non-limiting examples of modified and unmodified TRAC gRNA sequences that may be used as provided herein to create a genomic disruption in the TRAC gene are listed in Table 4 (e.g., SEQ ID NOs: 18 and 19). See also International Application No. PCT/US2018/032334, filed May 11, 2018, incorporated herein by reference.
  • Other gRNA sequences may be designed using the TRAC gene sequence located on chromosome 14 (GRCh38: chromosome 14: 22,547,506-22,552,154; Ensembl; ENSG00000277734).
  • gRNAs targeting the TRAC genomic region create Indels in the TRAC gene disrupting expression of the mRNA or protein.
  • At least 50% of a population of engineered T cells do not express a detectable level of T cell receptor (TCR) surface protein.
  • TCR T cell receptor
  • at least 55%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% of a population may not express a detectable level of TCR surface protein.
  • 50%-100%, 50%-90%, 50%-80%, 50%-70%, 50%-60%, 60%-100%, 60%-90%, 60%-80%, 60%-70%, 70%-100%, 70%-90%, 70%-80%, 80%-100%, 80%-90%, or 90%-100% of the population of engineered T cells do not express a detectable level of TCR surface protein.
  • gRNAs targeting the TRAC genomic region create Indels in the TRAC gene comprising at least one nucleotide sequence selected from the following sequences in Table 1:
  • an engineered T cell comprises a deletion in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion of 15-30 base pairs in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion of 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 base pairs in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion of more than 30 base pairs in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion of 20 base pairs in the TRAC gene relative to unmodified T cells.
  • an engineered T cell comprises a deletion of SEQ ID NO: 92 (AGAGCAACAGTGCTGTGGCC) in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion comprising SEQ ID NO: 92 (AGAGCAACAGTGCTGTGGCC) in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion of SEQ ID NO: 40 in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion comprising SEQ ID NO: 40 in the TRAC gene relative to unmodified T cells.
  • an engineered T cell comprises a disrupted ⁇ 2M gene.
  • ⁇ 2M is a common (invariant) component of MHC I complexes. Disrupting its expression by gene editing will prevent host versus therapeutic allogeneic T cells responses leading to increased allogeneic T cell persistence. In some embodiments, expression of the endogenous ⁇ 2M gene is eliminated to prevent a host-versus-graft response.
  • Non-limiting examples of modified and unmodified ⁇ 2M gRNA sequences that may Non-limiting examples of modified and unmodified ⁇ 2M gRNA sequences that may be used as provided herein to create a genomic disruption in the ⁇ 2M gene are listed in Table 4 (e.g., SEQ ID NOs: 20 and 21). See also International Application No. PCT/US2018/032334, filed May 11, 2018, incorporated herein by reference. Other gRNA sequences may be designed using the ⁇ 2M gene sequence located on Chromosome 15 (GRCh38 coordinates: Chromosome 15: 44,711,477-44,718,877; Ensembl: ENSG00000166710).
  • gRNAs targeting the ⁇ 2M genomic region create Indels in the ⁇ 2M gene disrupting expression of the mRNA or protein.
  • At least 50% of the engineered T cells of a population of engineered T cells does not express a detectable level of ⁇ 2M surface protein.
  • at least 55%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% of the engineered T cells of a population may not express a detectable level of ⁇ 2M surface protein.
  • 50%-100%, 50%-90%, 50%-80%, 50%-70%, 50%-60%, 60%-100%, 60%-90%, 60%-80%, 60%-70%, 70%-100%, 70%-90%, 70%-80%, 80%-100%, 80%-90%, or 90%-100% of the engineered T cells of a population does not express a detectable level of ⁇ 2M surface protein.
  • an edited ⁇ 2M gene comprises at least one nucleotide sequence selected from the following sequences in Table 2.
  • PD-1 is an immune checkpoint molecule that is upregulated in activated T cells and serves to dampen or stop T cell responses. Disrupting PD-1 by gene editing could lead to more persistent and/or potent therapeutic T cell responses and/or reduce immune suppression in a subject.
  • an engineered T cell comprises a disrupted PD-1 gene. In some embodiments, expression of the endogenous PD-1 gene is eliminated to enhance anti-tumor efficacy of the CAR T cells of the present disclosure.
  • Non-limiting examples of modified and unmodified PD-1 gRNA sequences that may be used as provided herein to create a genomic deletion in the PD-1 gene are listed in Table 4 (e.g., SEQ ID NOs: 22 and 23). See also International Application No. PCT/US2018/032334, filed May 11, 2018, incorporated herein by reference.
  • Other gRNA sequences may be designed using the PD-1 gene sequence located on Chromosome 2 (GRCh38 coordinates: Chromosome 2: 241,849,881-241,858,908; Ensembl: ENSG00000188389).
  • gRNAs targeting the PD-1 genomic region create Indels in the PD-1 gene disrupting expression of the PD-1 mRNA or protein.
  • an engineered T cell comprises a disrupted PD-1 gene.
  • at least 50% of the engineered T cells of a population of engineered T cells does not express a detectable level of PD-1 surface protein.
  • at least 55%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% of the engineered T cells of a population may not express a detectable level of PD-1 surface protein.
  • 50%-100%, 50%-90%, 50%-80%, 50%-70%, 50%-60%, 60%-100%, 60%-90%, 60%-80%, 60%-70%, 70%-100%, 70%-90%, 70%-80%, 80%-100%, 80%-90%, or 90%-100% of the engineered T cells of a population does not express a detectable level of PD-1 surface protein.
  • CD70 Cluster of Differentiation 70
  • CD70 Cluster of Differentiation 70
  • an engineered T cell comprises a disrupted CD70 gene.
  • expression of the endogenous CD70 gene is eliminated to enhance anti-tumor efficacy of the CAR T cells of the present disclosure.
  • gRNAs targeting the CD70 genomic region create Indels in, or around, the CD70 gene disrupting expression of the CD70 mRNA and/or protein.
  • Non-limiting examples of modified and unmodified CD70 gRNA sequences that may be used as provided herein to create a genomic disruption in the CD70 gene are listed in Table 4 (e.g., SEQ ID NOs: 24-27).
  • Other gRNA sequences may be designed using the CD70 gene sequence located on Chromosome 19 (GRCh38 coordinates: Chromosome 19: 6,583,183-6,604,103; Ensembl: ENSG00000125726).
  • an engineered T cell comprises a disrupted CD70 gene.
  • at least 50% of the engineered T cells of a population of engineered T cells does not express a detectable level of CD70 surface protein.
  • at least 55%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% of the engineered T cells of a population may not express a detectable level of CD70 surface protein.
  • 50%-100%, 50%-90%, 50%-80%, 50%-70%, 50%-60%, 60%-100%, 60%-90%, 60%-80%, 60%-70%, 70%-100%, 70%-90%, 70%-80%, 80%-100%, 80%-90%, or 90%-100% of the engineered T cells of a population does not express a detectable level of CD70 surface protein.
  • one or more gene edits within a population of cells results in a phenotype associated with changes in cellular proliferative capacity, cellular exhaustion, cellular viability, cellular lysis capability (e.g., increase cytokine production and/or release), or any combination thereof.
  • engineered T cells of the present disclosure exhibit at least 20% greater cellular proliferative capacity, relative to control T cells.
  • engineered T cells may exhibit at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, or at least 90% greater cellular proliferative capacity, relative to control T cells.
  • engineered T cells of the present disclosure exhibit 20%-100%, 20%-90%, 20%-80%, 20%-70%, 20%-60%, 20%-50%, 30%-100%, 30%-90%, 30%-80%, 30%-70%, 30%-60%, 30%-50%, 40%-100%, 40%-90%, 40%-80%, 40%-70%, 40%-60%, 40%-50%, 50%-100%, 50%-90%, 50%-80%, 50%-70%, or 50%-60% greater cellular proliferative capacity, relative to control T cells.
  • engineered T cells of the present disclosure exhibit an at least 20% increase in cellular viability, relative to control cells.
  • engineered T cells of the present disclosure may exhibit at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, or at least 90% increase in cellular viability, relative to control cells.
  • engineered T cells of the present disclosure exhibit a 20%-100%, 20%-90%, 20%-80%, 20%-70%, 20%-60%, 20%-50%, 30%-100%, 30%-90%, 30%-80%, 30%-70%, 30%-60%, 30%-50%, 40%-100%, 40%-90%, 40%-80%, 40%-70%, 40%-60%, 40%-50%, 50%-100%, 50%-90%, 50%-80%, 50%-70%, or 50%-60% increase in cellular viability, relative to control cells.
  • engineered T cells of the present disclosure exhibit an at least 20% increase in cellular lysis capability (kill at least 20% more target cells), relative to control cells.
  • engineered T cells of the present disclosure may exhibit an at least at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, or at least 90% increase in cellular lysis capability, relative to control cells.
  • engineered T cells of the present disclosure exhibit a 20%-100%, 20%-90%, 20%-80%, 20%-70%, 20%-60%, 20%-50%, 30%-100%, 30%-90%, 30%-80%, 30%-70%, 30%-60%, 30%-50%, 40%-100%, 40%-90%, 40%-80%, 40%-70%, 40%-60%, 40%-50%, 50%-100%, 50%-90%, 50%-80%, 50%-70%, or 50%-60% increase in cellular lysis capability, relative to control cells.
  • the level of cytokines (e.g., IL-2 and/or IFN-gamma) secreted by the engineered T cells may at least 2-fold (e.g., at least 3-fold, at least 4-fold, or at least 5-fold) greater than the level of cytokines secreted by control T cells.
  • Control T cells are engineered T cells (e.g., gene edited T cells).
  • control T cells are engineered T cells that comprise a disrupted TRAC gene, a nucleic acid encoding a CAR (e.g., an anti-LIV1 CAR) inserted into the TRAC gene, and/or a disrupted ⁇ 2M gene.
  • control T cells are unedited T cells.
  • Gene editing is a type of genetic engineering in which nucleotide(s)/nucleic acid(s) is/are inserted, deleted, and/or substituted in a DNA sequence, such as in the genome of a targeted cell.
  • Targeted gene editing enables insertion, deletion, and/or substitution at pre-selected sites in the genome of a targeted cell (e.g., in a targeted gene or targeted DNA sequence).
  • an sequence of an endogenous gene is edited, for example by deletion, insertion or substitution of nucleotide(s)/nucleic acid(s)
  • the endogenous gene comprising the affected sequence may be knocked-out or knocked-down due to the sequence alteration.
  • Targeted editing may be used to disrupt endogenous gene expression.
  • “Targeted integration” refers to a process involving insertion of one or more exogenous sequences, with or without deletion of an endogenous sequence at the insertion site. Targeted integration can result from targeted gene editing when a donor template containing an exogenous sequence is present.
  • Targeted editing can be achieved either through a nuclease-independent approach, or through a nuclease-dependent approach.
  • nuclease-independent targeted editing approach homologous recombination is guided by homologous sequences flanking an exogenous polynucleotide to be introduced into an endogenous sequence through the enzymatic machinery of the host cell.
  • the exogenous polynucleotide may introduce deletions, insertions or replacement of nucleotides in the endogenous sequence.
  • nuclease-dependent approach can achieve targeted editing with higher frequency through the specific introduction of double strand breaks (DSBs) by specific rare-cutting nucleases (e.g., endonucleases).
  • DSBs double strand breaks
  • nuclease-dependent targeted editing also utilizes DNA repair mechanisms, for example, non-homologous end joining (NHEJ), which occurs in response to DSBs.
  • NHEJ non-homologous end joining
  • DNA repair by NHEJ often leads to random insertions or deletions (indels) of a small number of endogenous nucleotides.
  • repair can also occur by a homology directed repair (HDR).
  • HDR homology directed repair
  • Available endonucleases capable of introducing specific and targeted DSBs include, but not limited to, zinc-finger nucleases (ZFN), transcription activator-like effector nucleases (TALEN), and RNA-guided CRISPR-Cas9 nuclease (CRISPR/Cas9; Clustered Regular Interspaced Short Palindromic Repeats Associated 9). Additionally, DICE (dual integrase cassette exchange) system utilizing phiC31 and Bxb1 integrases may also be used for targeted integration.
  • ZFN zinc-finger nucleases
  • TALEN transcription activator-like effector nucleases
  • CRISPR/Cas9 Clustered Regular Interspaced Short Palindromic Repeats Associated 9
  • DICE dual integrase cassette exchange
  • ZFNs are targeted nucleases comprising a nuclease fused to a zinc finger DNA binding domain (ZFBD), which is a polypeptide domain that binds DNA in a sequence-specific manner through one or more zinc fingers.
  • ZFBD zinc finger DNA binding domain
  • a zinc finger is a domain of about 30 amino acids within the zinc finger binding domain whose structure is stabilized through coordination of a zinc ion. Examples of zinc fingers include, but not limited to, C2H2 zinc fingers, C3H zinc fingers, and C4 zinc fingers.
  • a designed zinc finger domain is a domain not occurring in nature whose design/composition results principally from rational criteria, e.g., application of substitution rules and computerized algorithms for processing information in a database storing information of existing ZFP designs and binding data. See, for example, U.S.
  • a selected zinc finger domain is a domain not found in nature whose production results primarily from an empirical process such as phage display, interaction trap or hybrid selection.
  • ZFNs are described in greater detail in U.S. Pat. Nos. 7,888,121 and 7,972,854. The most recognized example of a ZFN is a fusion of the FokI nuclease with a zinc finger DNA binding domain.
  • a TALEN is a targeted nuclease comprising a nuclease fused to a TAL effector DNA binding domain.
  • a “transcription activator-like effector DNA binding domain”, “TAL effector DNA binding domain”, or “TALE DNA binding domain” is a polypeptide domain of TAL effector proteins that is responsible for binding of the TAL effector protein to DNA. TAL effector proteins are secreted by plant pathogens of the genus Xanthomonas during infection. These proteins enter the nucleus of the plant cell, bind effector-specific DNA sequences via their DNA binding domain, and activate gene transcription at these sequences via their transactivation domains.
  • TAL effector DNA binding domain specificity depends on an effector-variable number of imperfect 34 amino acid repeats, which comprise polymorphisms at select repeat positions called repeat variable-diresidues (RVD).
  • RVD repeat variable-diresidues
  • TALENs are described in greater detail in US Patent Application No. 2011/0145940. The most recognized example of a TALEN in the art is a fusion polypeptide of the FokI nuclease to a TAL effector DNA binding domain.
  • targeted nucleases suitable for use as provided herein include, but are not limited to, Bxb1, phiC31, R4, PhiBT1, and W ⁇ /SPBc/TP901-1, whether used individually or in combination.
  • targeted nucleases include naturally-occurring and recombinant nucleases, e.g., CRISPR/Cas9, restriction endonucleases, meganucleases homing endonucleases, and the like.
  • the CRISPR-Cas9 system is a naturally-occurring defense mechanism in prokaryotes that has been repurposed as a RNA-guided DNA-targeting platform used for gene editing. It relies on the DNA nuclease Cas9, and two noncoding RNAs-crisprRNA (crRNA) and trans-activating RNA (tracrRNA)—to target the cleavage of DNA.
  • crRNA noncoding RNAs-crisprRNA
  • tracrRNA trans-activating RNA
  • crRNA drives sequence recognition and specificity of the CRISPR-Cas9 complex through Watson-Crick base pairing typically with a 20 nucleotide (nt) sequence in the target DNA. Changing the sequence of the 5′ 20 nt in the crRNA allows targeting of the CRISPR-Cas9 complex to specific loci.
  • the CRISPR-Cas9 complex only binds DNA sequences that contain a sequence match to the first 20 nt of the crRNA, single-guide RNA (sgRNA), if the target sequence is followed by a specific short DNA motif (with the sequence NGG) referred to as a protospacer adjacent motif (PAM).
  • sgRNA single-guide RNA
  • PAM protospacer adjacent motif
  • TracrRNA hybridizes with the 3′ end of crRNA to form an RNA-duplex structure that is bound by the Cas9 endonuclease to form the catalytically active CRISPR-Cas9 complex, which can then cleave the target DNA.
  • NHEJ non-homologous end-joining
  • HDR homology-directed repair
  • NHEJ is a robust repair mechanism that appears highly active in the majority of cell types, including non-dividing cells. NHEJ is error-prone and can often result in the removal or addition of between one and several hundred nucleotides at the site of the DSB, though such modifications are typically ⁇ 20 nt. The resulting insertions and deletions (indels) can disrupt coding or noncoding regions of genes.
  • HDR uses a long stretch of homologous donor DNA, provided endogenously or exogenously, to repair the DSB with high fidelity. HDR is active only in dividing cells, and occurs at a relatively low frequency in most cell types. In many embodiments of the present disclosure, NHEJ is utilized as the repair operant.
  • the Cas9 (CRISPR associated protein 9) endonuclease is from Streptococcus pyogenes , although other Cas9 homologs may be used. It should be understood, that wild-type Cas9 may be used or modified versions of Cas9 may be used (e.g., evolved versions of Cas9, or Cas9 orthologues or variants), as provided herein. In some embodiments, Cas9 may be substituted with another RNA-guided endonuclease, such as Cpf1 (of a class II CRISPR/Cas system).
  • Cpf1 of a class II CRISPR/Cas system
  • the present disclosure provides a genome-targeting nucleic acid that can direct the activities of an associated polypeptide (e.g., a site-directed polypeptide) to a specific target sequence within a target nucleic acid.
  • the genome-targeting nucleic acid can be an RNA.
  • a genome-targeting RNA is referred to as a “guide RNA” or “gRNA” herein.
  • a guide RNA comprises at least a spacer sequence that hybridizes to a target nucleic acid sequence of interest, and a CRISPR repeat sequence.
  • the gRNA also comprises a second RNA called the tracrRNA sequence.
  • the CRISPR repeat sequence and tracrRNA sequence hybridize to each other to form a duplex.
  • the crRNA forms a duplex.
  • the duplex binds a site-directed polypeptide, such that the guide RNA and site-direct polypeptide form a complex.
  • the genome-targeting nucleic acid provides target specificity to the complex by virtue of its association with the site-directed polypeptide. The genome-targeting nucleic acid thus directs the activity of the site-directed polypeptide.
  • each guide RNA is designed to include a spacer sequence complementary to its genomic target sequence. See Jinek et al., Science, 337, 816-821 (2012) and Deltcheva et al., Nature, 471, 602-607 (2011).
  • the genome-targeting nucleic acid is a double-molecule guide RNA. In some embodiments, the genome-targeting nucleic acid is a single-molecule guide RNA.
  • a double-molecule guide RNA comprises two strands of RNA.
  • the first strand comprises in the 5′ to 3′ direction, an optional spacer extension sequence, a spacer sequence and a minimum CRISPR repeat sequence.
  • the second strand comprises a minimum tracrRNA sequence (complementary to the minimum CRISPR repeat sequence), a 3′ tracrRNA sequence and an optional tracrRNA extension sequence.
  • a single-molecule guide RNA (sgRNA) in a Type II system comprises, in the 5′ to 3′ direction, an optional spacer extension sequence, a spacer sequence, a minimum CRISPR repeat sequence, a single-molecule guide linker, a minimum tracrRNA sequence, a 3′ tracrRNA sequence and an optional tracrRNA extension sequence.
  • the optional tracrRNA extension may comprise elements that contribute additional functionality (e.g., stability) to the guide RNA.
  • the single-molecule guide linker links the minimum CRISPR repeat and the minimum tracrRNA sequence to form a hairpin structure.
  • the optional tracrRNA extension comprises one or more hairpins.
  • a single-molecule guide RNA (referred to as a “sgRNA” or “gRNA”) in a Type V system comprises, in the 5′ to 3′ direction, a minimum CRISPR repeat sequence and a spacer sequence.
  • the sgRNA can comprise a 20 nucleotide spacer sequence at the 5′ end of the sgRNA sequence.
  • the sgRNA can comprise a less than 20 nucleotide spacer sequence at the 5′ end of the sgRNA sequence.
  • the sgRNA can comprise a more than 20 nucleotide spacer sequence at the 5′ end of the sgRNA sequence.
  • the sgRNA can comprise a variable length spacer sequence with 17-30 nucleotides at the 5′ end of the sgRNA sequence (see Table 3).
  • the sgRNA can comprise no uracil at the 3′ end of the sgRNA sequence.
  • the sgRNA can comprise one or more uracil at the 3′ end of the sgRNA sequence.
  • the sgRNA can comprise 1 uracil (U) at the 3′ end of the sgRNA sequence.
  • the sgRNA can comprise 2 uracil (UU) at the 3′ end of the sgRNA sequence.
  • the sgRNA can comprise 3 uracil (UUU) at the 3′ end of the sgRNA sequence.
  • the sgRNA can comprise 4 uracil (UUUU) at the 3′ end of the sgRNA sequence.
  • the sgRNA can comprise 5 uracil (UUUU) at the 3′ end of the sgRNA sequence.
  • the sgRNA can comprise 6 uracil (UUUUU) at the 3′ end of the sgRNA sequence.
  • the sgRNA can comprise 7 uracil (U) at the 3′ end of the sgRNA sequence.
  • the sgRNA can comprise 8 uracil (UU at the 3′ end of the sgRNA sequence.
  • modified sgRNAs can comprise one or more 2′-O-methyl phosphorothioate nucleotides.
  • guide RNAs used in the CRISPR/Cas/Cpf1 system can be readily synthesized by chemical means, as illustrated below and described in the art. While chemical synthetic procedures are continually expanding, purifications of such RNAs by procedures such as high performance liquid chromatography (HPLC, which avoids the use of gels such as PAGE) tends to become more challenging as polynucleotide lengths increase significantly beyond a hundred or so nucleotides.
  • HPLC high performance liquid chromatography
  • One approach used for generating RNAs of greater length is to produce two or more molecules that are ligated together. Much longer RNAs, such as those encoding a Cas9 or Cpf1 endonuclease, are more readily generated enzymatically.
  • RNA modifications can be introduced during or after chemical synthesis and/or enzymatic generation of RNAs, e.g., modifications that enhance stability, reduce the likelihood or degree of innate immune response, and/or enhance other attributes, as described in the art.
  • a gRNA comprises a spacer sequence.
  • a spacer sequence is a sequence (e.g., a 20 nucleotide sequence) that defines the target sequence (e.g., a DNA target sequences, such as a genomic target sequence) of a target nucleic acid of interest.
  • the spacer sequence is 15 to 30 nucleotides.
  • the spacer sequence is 15, 16, 17, 18, 19, 29, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides.
  • a spacer sequence is 20 nucleotides.
  • the “target sequence” is adjacent to a PAM sequence and is the sequence modified by an RNA-guided nuclease (e.g., Cas9).
  • the “target nucleic acid” is a double-stranded molecule: one strand comprises the target sequence and is referred to as the “PAM strand,” and the other complementary strand is referred to as the “non-PAM strand.”
  • PAM strand the target sequence
  • non-PAM strand the other complementary strand
  • the gRNA spacer sequence hybridizes to the reverse complement of the target sequence, which is located in the non-PAM strand of the target nucleic acid of interest.
  • the gRNA spacer sequence is the RNA equivalent of the target sequence.
  • the gRNA spacer sequence is 5′-AGAGCAACAGUGCUGUGGCC-3′ (SEQ ID NO: 93).
  • the spacer of a gRNA interacts with a target nucleic acid of interest in a sequence-specific manner via hybridization (i.e., base pairing).
  • the nucleotide sequence of the spacer thus varies depending on the target sequence of the target nucleic acid of interest.
  • the spacer sequence is designed to hybridize to a region of the target nucleic acid that is located 5′ of a PAM of the Cas9 enzyme used in the system.
  • the spacer may perfectly match the target sequence or may have mismatches.
  • Each Cas9 enzyme has a particular PAM sequence that it recognizes in a target DNA.
  • S. pyogenes recognizes in a target nucleic acid a PAM that comprises the sequence 5′-NRG-3′, where R comprises either A or G, where N is any nucleotide and N is immediately 3′ of the target nucleic acid sequence targeted by the spacer sequence.
  • the target nucleic acid sequence comprises 20 nucleotides. In some embodiments, the target nucleic acid comprises less than 20 nucleotides. In some embodiments, the target nucleic acid comprises more than 20 nucleotides. In some embodiments, the target nucleic acid comprises at least: 5, 10, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides. In some embodiments, the target nucleic acid comprises at most: 5, 10, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides. In some embodiments, the target nucleic acid sequence comprises 20 bases immediately 5′ of the first nucleotide of the PAM.
  • the target nucleic acid comprises the sequence that corresponds to the Ns, wherein N is any nucleotide, and the underlined NRG sequence is the S. pyogenes PAM.
  • Non-limiting examples of gRNAs that may be used as provided herein are provided in Table 4 and PCT/US2018/032334, filed May 11, 2018.
  • a chimeric antigen receptor refers to an artificial immune cell receptor that is engineered to recognize and bind to an antigen expressed by tumor cells.
  • a CAR is designed for a T cell and is a chimera of a signaling domain of the T-cell receptor (TCR) complex and an antigen-recognizing domain (e.g., a single chain fragment (scFv) of an antibody or other antibody fragment) (Enblad et al., Human Gene Therapy. 2015; 26(8):498-505).
  • TCR T-cell receptor
  • scFv single chain fragment
  • a T cell that expresses a CAR is referred to as a CAR T cell.
  • CARs have the ability to redirect T-cell specificity and reactivity toward a selected target in a non-MHC-restricted manner.
  • T-cells expressing CARs the ability to recognize an antigen independent of antigen processing, thus bypassing a major mechanism of tumor escape.
  • CARs when expressed in T-cells, CARs advantageously do not dimerize with endogenous T-cell receptor (TCR) alpha and beta chains.
  • First generation CARs join an antibody-derived scFv to the CD3zeta ( ⁇ or z) intracellular signaling domain of the T-cell receptor through hinge and transmembrane domains.
  • Second generation CARs incorporate an additional domain, e.g., CD28, 4-1BB (41BB), or ICOS, to supply a costimulatory signal.
  • Third-generation CARs contain two costimulatory domains fused with the TCR CD3 ⁇ chain.
  • Third-generation costimulatory domains may include, e.g., a combination of CD3 ⁇ , CD27, CD28, 4-1BB, ICOS, or OX40.
  • CARs in some embodiments, contain an ectodomain (e.g., CD3), commonly derived from a single chain variable fragment (scFv), a hinge, a transmembrane domain, and an endodomain with one (first generation), two (second generation), or three (third generation) signaling domains derived from CD3Z and/or co-stimulatory molecules (Maude et al., Blood. 2015; 125(26):4017-4023; Kakarla and Gottschalk, Cancer J. 2014; 20(2):151-155).
  • scFv single chain variable fragment
  • CARs typically differ in their functional properties.
  • the CD3 ⁇ signaling domain of the T-cell receptor when engaged, will activate and induce proliferation of T-cells but can lead to anergy (a lack of reaction by the body's defense mechanisms, resulting in direct induction of peripheral lymphocyte tolerance). Lymphocytes are considered anergic when they fail to respond to a specific antigen.
  • the addition of a costimulatory domain in second-generation CARs improved replicative capacity and persistence of modified T-cells. Similar antitumor effects are observed in vitro with CD28 or 4-1BB CARs, but preclinical in vivo studies suggest that 4-1BB CARs may produce superior proliferation and/or persistence.
  • a chimeric antigen receptor is a first generation CAR. In other embodiments, a chimeric antigen receptor is a second generation CAR. In yet other embodiments, a chimeric antigen receptor is a third generation CAR.
  • a CAR in some embodiments, comprises an extracellular (ecto) domain comprising an antigen binding domain (e.g., an antibody, such as an scFv), a transmembrane domain, and a cytoplasmic (endo) domain.
  • an antigen binding domain e.g., an antibody, such as an scFv
  • a transmembrane domain e.g., a transmembrane domain
  • endo cytoplasmic domain
  • the ectodomain is the region of the CAR that is exposed to the extracellular fluid and, in some embodiments, includes an antigen binding domain, and optionally a signal peptide, a spacer domain, and/or a hinge domain.
  • the antigen binding domain is a single-chain variable fragment (scFv) that include the light and heavy chains of immunoglobulins connected with a short linker peptide.
  • the linker in some embodiments, includes hydrophilic residues with stretches of glycine and serine for flexibility as well as stretches of glutamate and lysine for added solubility.
  • a single-chain variable fragment is not actually a fragment of an antibody, but instead is a fusion protein of the variable regions of the heavy (VH) and light chains (VL) of immunoglobulins, connected with a short linker peptide of ten to about 25 amino acids.
  • the linker is usually rich in glycine for flexibility, as well as serine or threonine for solubility, and can either connect the N-terminus of the VH with the C-terminus of the VL, or vice versa. This protein retains the specificity of the original immunoglobulin, despite removal of the constant regions and the introduction of the linker.
  • Non-limiting examples of VH and VL protein sequences that may be used to create an anti-LIV1 scFv may include the amino acid sequence of SEQ ID NOs: 55, 90 or 98 (VH) and SEQ ID NOs: 56, 88 or 128 (VL).
  • the scFv of the present disclosure is humanized.
  • the scFv is fully human.
  • the scFv is a chimera (e.g., of mouse and human sequence).
  • the scFv is an anti-LIV1 scFv (binds specifically to LIV1).
  • Non-limiting examples of anti-LIV1 scFv proteins may include the amino acid sequence of any one of SEQ ID NOs: 54, 70, 82, 83, 84, 85, 86, or 87. Other scFv proteins may be used.
  • the signal peptide can enhance the antigen specificity of CAR binding.
  • Signal peptides can be derived from antibodies, such as, but not limited to, CD8, as well as epitope tags such as, but not limited to, GST or FLAG. Examples of signal peptides include MLLLVTSLLLCELPHPAFLLIP (SEQ ID NO: 94) and MALPVTALLLPLALLLHAARP (SEQ ID NO: 73). Other signal peptides may be used.
  • a spacer domain or hinge domain is located between an extracellular domain (comprising the antigen binding domain) and a transmembrane domain of a CAR, or between a cytoplasmic domain and a transmembrane domain of the CAR.
  • a spacer domain is any oligopeptide or polypeptide that functions to link the transmembrane domain to the extracellular domain and/or the cytoplasmic domain in the polypeptide chain.
  • a hinge domain is any oligopeptide or polypeptide that functions to provide flexibility to the CAR, or domains thereof, or to prevent steric hindrance of the CAR, or domains thereof.
  • a spacer domain or a hinge domain may comprise up to 300 amino acids (e.g., 10 to 100 amino acids, or 5 to 20 amino acids). In some embodiments, one or more spacer domain(s) may be included in other regions of a CAR. In some embodiments, the hinge domain is a CD8 hinge domain. Other hinge domains may be used.
  • the transmembrane domain is a hydrophobic alpha helix that spans the membrane.
  • the transmembrane domain provides stability of the CAR.
  • the transmembrane domain of a CAR as provided herein is a CD8 transmembrane domain.
  • the transmembrane domain is a CD28 transmembrane domain.
  • the transmembrane domain is a chimera of a CD8 and CD28 transmembrane domain.
  • Other transmembrane domains may be used as provided herein.
  • the transmembrane domain is a CD8a transmembrane domain: FVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGG AVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCNHRNR (SEQ ID NO: 129).
  • Other transmembrane domains may be used.
  • Endodomain The endodomain is the functional end of the receptor. Following antigen recognition, receptors cluster and a signal is transmitted to the cell.
  • the most commonly used endodomain component is CD3-zeta, which contains three (3) immunoreceptor tyrosine-based activation motif (ITAM)s. This transmits an activation signal to the T cell after the antigen is bound.
  • CD3-zeta may not provide a fully competent activation signal and, thus, a co-stimulatory signaling is used.
  • CD28 and/or 4-1BB may be used with CD3-zeta (CD3) to transmit a proliferative/survival signal.
  • the co-stimulatory molecule of a CAR as provided herein is a CD28 co-stimulatory molecule.
  • the co-stimulatory molecule is a 4-1BB co-stimulatory molecule.
  • a CAR includes CD3 ⁇ and CD28.
  • a CAR includes CD3-zeta and 4-1BB.
  • An antibody is an immunoglobulin molecule capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, polypeptide, etc., through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule.
  • antibody encompasses not only intact (i.e., full-length) monoclonal antibodies, but also antigen-binding fragments (such as Fab, Fab′, F(ab′)2, Fv), single chain variable fragment (scFv), mutants thereof, fusion proteins comprising an antibody portion, humanized antibodies, chimeric antibodies, diabodies, linear antibodies, single chain antibodies, single domain antibodies (e.g., camel or llama VHH antibodies), multispecific antibodies (e.g., bispecific antibodies) and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site of the required specificity, including glycosylation variants of antibodies, amino acid sequence variants of antibodies, and covalently modified antibodies.
  • antigen-binding fragments such as Fab, Fab′, F(ab′)2, Fv
  • scFv single chain variable fragment
  • mutants thereof fusion proteins comprising an antibody portion, humanized antibodies, chimeric antibodies, diabodies, linear antibodies, single chain antibodies, single domain antibodies
  • a typical antibody molecule comprises a heavy chain variable region (VH) and a light chain variable region (VL), which are usually involved in antigen binding. These regions/residues that are responsible for antigen-binding can be identified from amino acid sequences of the VH/VL sequences of a reference antibody (e.g., an anti-LIV1 antibody as described herein) by methods known in the art.
  • the VH and VL regions can be further subdivided into regions of hypervariability, also known as “complementarity determining regions” (“CDR”), interspersed with regions that are more conserved, which are known as “framework regions” (“FR”).
  • CDR complementarity determining regions
  • Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the extent of the framework region and CDRs can be precisely identified using methodology known in the art, for example, by the Kabat definition, the Chothia definition, the AbM definition, and/or the contact definition, all of which are well known in the art.
  • a CDR may refer to the CDR defined by any method known in the art.
  • Two antibodies having the same CDR means that the two antibodies have the same amino acid sequence of that CDR as determined by the same method. See, e.g., Kabat, E. A., et al.
  • an antibody is an scFv, such as an anti-LIV1 scFv.
  • An antibody includes an antibody of any class, such as IgD, IgE, IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class.
  • immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • the antibodies to be used as provided herein can be murine, rat, human, or any other origin (including chimeric or humanized antibodies).
  • the antibody comprises a modified constant region, such as a constant region that is immunologically inert, e.g., does not trigger complement mediated lysis, or does not stimulate antibody-dependent cell mediated cytotoxicity (ADCC).
  • a modified constant region such as a constant region that is immunologically inert, e.g., does not trigger complement mediated lysis, or does not stimulate antibody-dependent cell mediated cytotoxicity (ADCC).
  • an antibody of the present disclosure is a humanized antibody.
  • Humanized antibodies refer to forms of non-human (e.g., murine) antibodies that are specific chimeric immunoglobulins, immunoglobulin chains, or antigen-binding fragments thereof that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat, or rabbit having the desired specificity, affinity, and capacity.
  • CDR complementary determining region
  • donor antibody such as mouse, rat, or rabbit having the desired specificity, affinity, and capacity.
  • Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • the humanized antibody may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences, but are included to further refine and optimize antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • a humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin.
  • humanized antibodies have one or more CDRs (one, two, three, four, five, six) which are altered with respect to the original antibody, which are also termed one or more CDRs “derived from” one or more CDRs from the original antibody. Humanized antibodies may also involve affinity maturation.
  • an antibody of the present disclosure is a chimeric antibody, which can include a heavy constant region and a light constant region from a human antibody.
  • Chimeric antibodies refer to antibodies having a variable region or part of variable region from a first species and a constant region from a second species.
  • the variable region of both light and heavy chains mimics the variable regions of antibodies derived from one species of mammals (e.g., a non-human mammal such as mouse, rabbit, and rat), while the constant portions are homologous to the sequences in antibodies derived from another mammal such as human.
  • amino acid modifications can be made in the variable region and/or the constant region.
  • an antibody of the present disclosure specifically binds a target antigen, such as human LIV1.
  • a target antigen such as human LIV1.
  • An antibody that “specifically binds” (used interchangeably herein) to a target or an epitope is a term well understood in the art, and methods to determine such specific binding are also well known in the art.
  • a molecule is said to exhibit “specific binding” if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular target antigen than it does with alternative targets.
  • An antibody “specifically binds” to a target antigen if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other substances.
  • an antibody that specifically (or preferentially) binds to a LIV1 epitope is an antibody that binds this LIV1 epitope with greater affinity, avidity, more readily, and/or with greater duration than it binds to other LIV1 epitopes or non-LIV1 epitopes. It is also understood by reading this definition that, for example, an antibody that specifically binds to a first target antigen may or may not specifically or preferentially bind to a second target antigen. As such, “specific binding” or “preferential binding” does not necessarily require (although it can include) exclusive binding. Generally, but not necessarily, reference to binding means preferential binding.
  • the equilibrium dissociation constant (K D ) between the antibody and LIV1 is 100 pM to 1 ⁇ M. In some embodiments, the K D between the antibody and LIV1 is 1 nM to 100 nM.
  • a functional variant may contain one or more amino acid residue variations in the VH and/or VL, or in one or more of the VH CDRs and/or one or more of the VL CDRs as relative to a reference antibody, while retaining substantially similar binding and biological activities (e.g., substantially similar binding affinity, binding specificity, inhibitory activity, anti-tumor activity, or a combination thereof) as the reference antibody.
  • an antibody disclosed herein comprises a VH CDR1, a VH CDR2, and a VH CDR3, which collectively contains no more than 10 amino acid variations (e.g., no more than 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid variation) as compared with the VH CDR1, VH CDR2, and VH CDR3 of a reference antibody such as in VH: SEQ ID NO: 55 or 90 or 98; VL: SEQ ID NO: 56 or 88 or 128. “Collectively” means that the total number of amino acid variations in all of the three VH CDRs is within the defined range.
  • antibody may comprise a VL CDR1, a VL CDR2, and a VL CDR3, which collectively contains no more than 10 amino acid variations (e.g., no more than 9, 8, 7, 6, 5, 4, 3, 2 or 1 amino acid variation) as compared with the VL CDR1, VL CDR2, and VL CDR3 of the reference antibody.
  • an antibody disclosed herein may comprise a VH CDR1, a VH CDR2, and a VH CDR3, at least one of which contains no more than 5 amino acid variations (e.g., no more than 4, 3, 2, or 1 amino acid variation) as the counterpart VH CDR of a reference antibody such as in VH: SEQ ID NO: 55 or 90 or 98; VL: SEQ ID NO: 56 or 88 or 128.
  • VH SEQ ID NO: SEQ ID NO: 55 or 90 or 98
  • VL SEQ ID NO: 56 or 88 or 128.
  • the antibody comprises a VH CDR3, which contains no more than 5 amino acid variations (e.g., no more than 4, 3, 2, or 1 amino acid variation) as the VH CDR3 of a reference antibody such as in VH: SEQ ID NO: 55 or 90 or 98; VL: SEQ ID NO: 56 or 88 or 128.
  • an antibody may comprise a VL CDR1, a VL CDR2, and a VL CDR3, at least one of which contains no more than 5 amino acid variations (e.g., no more than 4, 3, 2, or 1 amino acid variation) as the counterpart VL CDR of the reference antibody.
  • the antibody comprises a VL CDR3, which contains no more than 5 amino acid variations (e.g., no more than 4, 3, 2, or 1 amino acid variation) as the LC CDR3 of the reference antibody.
  • amino acid residue variations can be conservative amino acid residue substitutions.
  • a “conservative amino acid substitution” refers to an amino acid substitution that does not alter the relative charge or size characteristics of the protein in which the amino acid substitution is made.
  • Variants can be prepared according to methods for altering polypeptide sequence known to one of ordinary skill in the art such as are found in references which compile such methods, e.g. Molecular Cloning: A Laboratory Manual, J. Sambrook, et al., eds., Second Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989, or Current Protocols in Molecular Biology, F. M.
  • Conservative substitutions of amino acids include substitutions made amongst amino acids within the following groups: (a) A ⁇ G, S; (b) R ⁇ K, H; (c) N ⁇ Q, H; (d) D ⁇ E, N; (e) C ⁇ S, A; (f) Q ⁇ N; (g) E ⁇ D, Q; (h) G ⁇ A; (i) H ⁇ N, Q; (j) I ⁇ L, V; (k) L ⁇ I, V; (l) K ⁇ R, H; (m) M ⁇ L, I, Y; (n) F ⁇ Y, M, L; (o) P ⁇ A; (p) S ⁇ T; (q) T ⁇ S; (r) W ⁇ Y, F; (s) Y ⁇ W, F; and (t) V ⁇ I, L.
  • an antibody disclosed herein may comprise VH CDRs that collectively are at least 80% (e.g., 85%, 90%, 95%, or 98%) identical to the VH CDRs of a reference antibody such as Antibody A (VH: SEQ ID NO: 55; VL: SEQ ID NO: 56) or Antibody B (VH: SEQ ID NO: 90; VL: SEQ ID NO: 88).
  • the antibody may comprise VL CDRs that collectively are at least 80% (e.g., 85%, 90%, 95%, or 98%) identical to the VL CDRs of the reference antibody.
  • an antibody may comprise a VH that is at least 80% (e.g., 85%, 90%, 95%, or 98%) identical to the VH of a reference antibody such as in VH: SEQ ID NO: 55 or 90 or 98; VL: SEQ ID NO: 56 or 88 or 128 and/or a VL variable region that is at least 80% (e.g., 85%, 90%, 95%, or 98%) identical to the VL variable region of the reference antibody.
  • the nucleic acid encoding a CAR may be delivered to a T cell that comprises what is referred to herein as a donor template (also referred to as a donor polynucleotide).
  • a donor template can contain a non-homologous sequence, such as the nucleic acid encoding a CAR, flanked by two regions of homology to allow for efficient HDR at a genomic location of interest.
  • a donor template may have no regions of homology to the targeted location in the DNA and may be integrated by NHEJ-dependent end joining following cleavage at the target site.
  • a donor template can be DNA or RNA, single-stranded and/or double-stranded, and can be introduced into a cell in linear or circular form. If introduced in linear form, the ends of the donor sequence can be protected (e.g., from exonucleolytic degradation) by methods known to those of skill in the art. For example, one or more dideoxynucleotide residues are added to the 3′ terminus of a linear molecule and/or self-complementary oligonucleotides are ligated to one or both ends. See, for example, Chang et al., (1987) Proc. Natl. Acad. Sci.
  • Additional methods for protecting exogenous polynucleotides from degradation include, but are not limited to, addition of terminal amino group(s) and the use of modified internucleotide linkages such as, for example, phosphorothioates, phosphoramidates, and O-methyl ribose or deoxyribose residues.
  • a donor template can be introduced into a cell as part of a vector molecule having additional sequences such as, for example, replication origins, promoters and genes encoding antibiotic resistance.
  • a donor template can be introduced as naked nucleic acid, as nucleic acid complexed with an agent such as a liposome or poloxamer, or can be delivered by viruses (e.g., adenovirus, AAV, herpesvirus, retrovirus, lentivirus and integrase defective lentivirus (IDLV)).
  • viruses e.g., adenovirus, AAV, herpesvirus, retrovirus, lentivirus and integrase defective lentivirus (IDLV)
  • a donor template in some embodiments, is inserted so that its expression is driven by the endogenous promoter at the integration site, namely the promoter that drives expression of the endogenous gene into which the donor is inserted.
  • the donor template comprises an exogenous promoter and/or enhancer, for example a constitutive promoter, an inducible promoter, or tissue-specific promoter.
  • the exogenous promoter is an EF1 ⁇ promoter comprising a sequence of SEQ ID NO: 79. Other promoters may be used.
  • exogenous sequences may also include transcriptional or translational regulatory sequences, for example, promoters, enhancers, insulators, internal ribosome entry sites, sequences encoding 2A peptides and/or polyadenylation signals.
  • Nucleases and/or donor templates may be delivered using a vector system, including, but not limited to, plasmid vectors, DNA minicircles, retroviral vectors, lentiviral vectors, adenovirus vectors, poxvirus vectors; herpesvirus vectors and adeno-associated virus vectors, and combinations thereof.
  • a vector system including, but not limited to, plasmid vectors, DNA minicircles, retroviral vectors, lentiviral vectors, adenovirus vectors, poxvirus vectors; herpesvirus vectors and adeno-associated virus vectors, and combinations thereof.
  • Non-viral vector delivery systems include DNA plasmids, DNA minicircles, naked nucleic acid, and nucleic acid complexed with a delivery vehicle such as a liposome or poloxamer.
  • Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes after delivery to the cell.
  • Methods of non-viral delivery of nucleic acids include electroporation, lipofection, microinjection, biolistics, virosomes, liposomes, immunoliposomes, polycation or lipid:nucleic acid conjugates, naked DNA, naked RNA, capped RNA, artificial virions, and agent-enhanced uptake of DNA. Sonoporation using, e.g., the Sonitron 2000 system (Rich-Mar) can also be used for delivery of nucleic acids.
  • the donor nucleic acid encoding a CAR construct can be delivered to a cell using an adeno-associated virus (AAV).
  • AAVs are small viruses which integrate site-specifically into the host genome and can therefore deliver a transgene, such as CAR.
  • ITRs Inverted terminal repeats
  • capsids are present flanking the AAV genome and/or the transgene of interest and serve as origins of replication.
  • rep and cap proteins which, when transcribed, form capsids which encapsulate the AAV genome for delivery into target cells.
  • Surface receptors on these capsids which confer AAV serotype, which determines which target organs the capsids will primarily bind and thus what cells the AAV will most efficiently infect.
  • the AAV is AAV serotype 6 (AAV6).
  • Adeno-associated viruses are among the most frequently used viruses for gene therapy for several reasons. First, AAVs do not provoke an immune response upon administration to mammals, including humans. Second, AAVs are effectively delivered to target cells, particularly when consideration is given to selecting the appropriate AAV serotype. Finally, AAVs have the ability to infect both dividing and non-dividing cells because the genome can persist in the host cell without integration. This trait makes them an ideal candidate for gene therapy.
  • HDR Homology-Directed Repair
  • the donor nucleic acid encoding a CAR is inserted by homology directed repair (HDR) into the target gene locus. Both strands of the DNA at the target locus are cut by a CRISPR Cas9 enzyme. HDR then occurs to repair the double-strand break (DSB) and insert the donor DNA. For this to occur correctly, the donor sequence is designed with flanking residues which are complementary to the sequence surrounding the DSB site in the target gene (hereinafter “homology arms”). These homology arms serve as the template for DSB repair and allow HDR to be an essentially error-free mechanism.
  • the rate of homology directed repair (HDR) is a function of the distance between the mutation and the cut site so choosing overlapping or nearby target sites is important. Templates can include extra sequences flanked by the homologous regions or can contain a sequence that differs from the genomic sequence, thus allowing sequence editing.
  • the target gene can be associated with an immune response in a subject, wherein permanently deleting at least a portion of the target gene will modulate the immune response.
  • the target gene can be the TCR ⁇ constant region (TRAC). Disruption of TRAC leads to loss of function of the endogenous TCR.
  • the target gene is in a safe harbor locus.
  • Engineered (gene edited) CAR T cells of the present disclosure may be autologous (“self”) or non-autologous (“non-self,” e.g., allogeneic, syngeneic or xenogeneic). “Autologous” refers to cells from the same subject. “Allogeneic” refers to cells of the same species as a subject, but that differ genetically to the cells in the subject.
  • the T cells are obtained from a mammalian subject. In some embodiments, the T cells are obtained from a human subject.
  • T cells can be obtained from a number of sources including, but not limited to, peripheral blood mononuclear cells, bone marrow, lymph nodes tissue, cord blood, thymus issue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors.
  • T cells can be obtained from a unit of blood collected from a subject using any number of techniques known to the skilled person, such as sedimentation, e.g., FICOLLTM separation.
  • an isolated population of T cells is used.
  • both cytotoxic and helper T lymphocytes can be sorted into naive, memory, and effector T cell subpopulations either before or after activation, expansion, and/or genetic modification.
  • a specific subpopulation of T cells expressing one or more of the following cell surface markers: TCRab, CD3, CD4, CD8, CD27 CD28, CD38 CD45RA, CD45RO, CD62L, CD127, CD122, CD95, CD197, CCR7, KLRG1, MCH-I proteins and/or MCH-II proteins, can be further isolated by positive or negative selection techniques.
  • a specific subpopulation of T cells, expressing one or more of the markers selected from the group consisting of TCRab, CD4 and/or CD8, is further isolated by positive or negative selection techniques.
  • the engineered T cell populations do not express or do not substantially express one or more of the following markers: CD70, CD57, CD244, CD160, PD-1, CTLA4, HM3, and LAG3.
  • subpopulations of T cells may be isolated by positive or negative selection prior to genetic engineering and/or post genetic engineering.
  • an isolated population of T cells expresses one or more of the markers including, but not limited to a CD3+, CD4+, CD8+, or a combination thereof.
  • the T cells are isolated from a subject and first activated and stimulated to proliferate in vitro prior to undergoing gene editing.
  • T cells are often subjected to one or more rounds of stimulation, activation and/or expansion.
  • T cells can 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; and 6,867,041.
  • T cells are activated and expanded for about 1 day to about 4 days, about 1 day to about 3 days, about 1 day to about 2 days, about 2 days to about 3 days, about 2 days to about 4 days, about 3 days to about 4 days, or about 1 day, about 2 days, about 3 days, or about 4 days prior to introduction of the genome editing compositions into the T cells.
  • T cells are activated and expanded for about 4 hours, about 6 hours, about 12 hours, about 18 hours, about 24 hours, about 36 hours, about 48 hours, about 60 hours, or about 72 hours prior to introduction of the gene editing compositions into the T cells.
  • T cells are activated at the same time that genome editing compositions are introduced into the T cells.
  • cancers e.g.: breast cancer
  • cancers that may be treated as provided herein include: breast cancer, e.g., estrogen receptor-positive breast cancer, prostate cancer, squamous tumors, e.g., of the skin, bladder, lung, cervix, endometrium, head neck, and biliary tract, and neuronal tumors.
  • the methods comprise delivering the CAR T cells (e.g., anti-LIV1 CAR T cells) of the present disclosure to a subject having cancer, including, breast cancer, e.g., estrogen receptor-positive breast cancer, prostate cancer, squamous tumors, e.g., of the skin, bladder, lung, cervix, endometrium, head neck, and biliary tract, and/or neuronal tumors.
  • cancer including, breast cancer, e.g., estrogen receptor-positive breast cancer, prostate cancer, squamous tumors, e.g., of the skin, bladder, lung, cervix, endometrium, head neck, and biliary tract, and/or neuronal tumors.
  • the step of administering may include the placement (e.g., transplantation) of cells, e.g., engineered T cells, into a subject, by a method or route that results in at least partial localization of the introduced cells at a desired site, such as tumor, such that a desired effect(s) is produced.
  • Engineered T cells can be administered by any appropriate route that results in delivery to a desired location in the subject where at least a portion of the implanted cells or components of the cells remain viable.
  • the period of viability of the cells after administration to a subject can be as short as a few hours, e.g., twenty-four hours, to a few days, to as long as several years, or even the life time of the subject, i.e., long-term engraftment.
  • an effective amount of engineered T cells is administered via a systemic route of administration, such as an intraperitoneal or intravenous route.
  • a subject may be any subject for whom diagnosis, treatment, or therapy is desired.
  • the subject is a mammal. In some embodiments, the subject is a human.
  • a donor is an individual who is not the subject being treated.
  • a donor is an individual who is not the patient.
  • a donor is an individual who does not have or is not suspected of having the cancer being treated.
  • multiple donors e.g., two or more donors, are used.
  • an engineered T cell population being administered according to the methods described herein comprises allogeneic T cells obtained from one or more donors.
  • Allogeneic refers to a cell, cell population, or biological samples comprising cells, obtained from one or more different donors of the same species, where the genes at one or more loci are not identical to the recipient.
  • an engineered T cell population, being administered to a subject can be derived from one or more unrelated donors, or from one or more non-identical siblings.
  • syngeneic cell populations may be used, such as those obtained from genetically identical donors, (e.g., identical twins).
  • the cells are autologous cells; that is, the engineered T cells are obtained or isolated from a subject and administered to the same subject, i.e., the donor and recipient are the same.
  • an engineered T cell population being administered according to the methods described herein does not induce toxicity in the subject, e.g., the engineered T cells do not induce toxicity in non-cancer cells.
  • an engineered T cell population being administered does not trigger complement mediated lysis, or does not stimulate antibody-dependent cell mediated cytotoxicity (ADCC).
  • ADCC antibody-dependent cell mediated cytotoxicity
  • An effective amount refers to the amount of a population of engineered T cells needed to prevent or alleviate at least one or more signs or symptoms of a medical condition (e.g., cancer), and relates to a sufficient amount of a composition to provide the desired effect, e.g., to treat a subject having a medical condition.
  • An effective amount also includes an amount sufficient to prevent or delay the development of a symptom of the disease, alter the course of a symptom of the disease (for example but not limited to, slow the progression of a symptom of the disease), or reverse a symptom of the disease. It is understood that for any given case, an appropriate effective amount can be determined by one of ordinary skill in the art using routine experimentation.
  • an effective amount of cells comprises at least 10 2 cells, at least 5 ⁇ 10 2 cells, at least 10 3 cells, at least 5 ⁇ 10 3 cells, at least 10 4 cells, at least 5 ⁇ 10 4 cells, at least 10 5 cells, at least 2 ⁇ 10 5 cells, at least 3 ⁇ 10 5 cells, at least 4 ⁇ 10 5 cells, at least 5 ⁇ 10 5 cells, at least 6 ⁇ 10 5 cells, at least 7 ⁇ 10 5 cells, at least 8 ⁇ 10 5 cells, at least 9 ⁇ 10 5 cells, at least 1 ⁇ 10 6 cells, at least 2 ⁇ 10 6 cells, at least 3 ⁇ 10 6 cells, at least 4 ⁇ 10 6 cells, at least 5 ⁇ 10 6 cells, at least 6 ⁇ 10 6 cells, at least 7 ⁇ 10 6 cells, at least 8 ⁇ 10 6 cells, at least 9 ⁇ 10 6 cells, or multiples thereof.
  • the cells are derived from one or more donors, or are obtained from an autologous source. In some examples described herein, the cells are expanded in culture prior to administration to
  • Modes of administration include injection, infusion, instillation, or ingestion.
  • Injection includes, without limitation, intravenous, intramuscular, intra-arterial, intrathecal, intraventricular, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, sub capsular, subarachnoid, intraspinal, intracerebro spinal, and intrasternal injection and infusion.
  • the route is intravenous.
  • engineered T cells are administered systemically, which refers to the administration of a population of cells other than directly into a target site, tissue, or organ, such that it enters, instead, the subject's circulatory system and, thus, is subject to metabolism and other like processes.
  • the efficacy of a treatment comprising a composition for the treatment of a medical condition can be determined by the skilled clinician.
  • a treatment is considered “effective treatment,” if any one or all of the signs or symptoms of, as but one example, levels of functional target are altered in a beneficial manner (e.g., increased by at least 10%), or other clinically accepted symptoms or markers of disease (e.g., cancer) are improved or ameliorated.
  • Efficacy can also be measured by failure of a subject to worsen as assessed by hospitalization or need for medical interventions (e.g., progression of the disease is halted or at least slowed). Methods of measuring these indicators are known to those of skill in the art and/or described herein.
  • Treatment includes any treatment of a disease in subject and includes: (1) inhibiting the disease, e.g., arresting, or slowing the progression of symptoms; or (2) relieving the disease, e.g., causing regression of symptoms; and (3) preventing or reducing the likelihood of the development of symptoms.
  • Embodiment 1 An engineered T cell comprising a nucleic acid encoding a chimeric antigen receptor (CAR), wherein the CAR comprise an ectodomain that binds specifically to LIV1.
  • CAR chimeric antigen receptor
  • Embodiment 2 The engineered T cell of embodiment 1 further comprising a disrupted T cell receptor alpha chain constant region (TRAC) gene.
  • T cell receptor alpha chain constant region TAC
  • Embodiment 3 The engineered T cell of embodiment 2, wherein the nucleic acid encoding the CAR is inserted into the TRAC gene.
  • Embodiment 4 The engineered T cell of any one of embodiments 1-3 further comprising a disrupted beta-2-microglobulin (P211 gene.
  • Embodiment 5 The engineered T cell of any one of embodiments 1-4, wherein the ectodomain of the CAR comprises an anti-LIV1 antibody.
  • Embodiment 6 The engineered T cell of embodiment 5, wherein the anti-LIV1 antibody is an anti-LIV1 single-chain variable fragment (scFv).
  • scFv anti-LIV1 single-chain variable fragment
  • Embodiment 7 The engineered T cell of embodiment 6, wherein the anti-LIV1 scFv comprises the same heavy chain variable region (VH) complementarity determining regions (CDRs) and the same light chain variable region (VL) CDRs as a reference antibody, wherein the reference antibody comprises (i) a VH set forth as SEQ ID NO: 55 and a VL set forth as SEQ ID NO: 56, (ii) a VH set forth as SEQ ID NO: 69 and a VL set forth as SEQ ID NO: 70, (iii) a VH set forth as SEQ ID NO: 76 and a VL set forth as SEQ ID NO: 77, or (iv) a VH set forth as SEQ ID NO: 83 and a VL set forth as SEQ ID NO: 84.
  • VH variable region
  • CDRs complementarity determining regions
  • VL light chain variable region
  • Embodiment 8 The engineered T cell of embodiment 7, wherein the anti-LIV1 scFv comprises the same VH and VL chains as the reference antibody.
  • Embodiment 8.1 The engineered T cell of embodiment 7, wherein the anti-LIV1 scFv comprises the amino acid sequence of any one of SEQ ID NOs: 54 or 70.
  • Embodiment 9 The engineered T cell of any one of embodiments 1-8.1, wherein the CAR comprises a CD28 co-stimulatory domain or a 41BB co-stimulatory domain.
  • Embodiment 10 The engineered T cell of embodiment 9, wherein the CAR further comprises a CD3z cytoplasmic signaling domain.
  • Embodiment 11 The engineered T cell of any one of embodiments 3-10, wherein the TRAC gene comprises the nucleotide sequence of any one of SEQ ID NOS: 63, 64, 71, or 72, and/or wherein the CAR is encoded by the nucleotide sequence of any one of SEQ ID NOS: 49, 51, 65, or 67.
  • Embodiment 12 The engineered T cell of any one of embodiments 4-11, wherein the disrupted ⁇ 2M gene comprises at least one nucleotide sequence selected from any one of SEQ ID NOS: 9-14.
  • Embodiment 13 A population of the engineered T cell of any one of embodiments 1-12, wherein at least 25% or at least 50% of engineered T cells of the population express the CAR.
  • Embodiment 14 The population of embodiment 14, wherein at least 70% of engineered T cells of the population express the CAR.
  • Embodiment 15 The population of embodiment 13, wherein at least 25% of engineered T cells of the population express the CAR following at least 7 or at least 14 days of in vitro proliferation.
  • Embodiment 16 The population of any one of embodiments 13-15, wherein at least 50% of engineered T cells of the population do not express a detectable level of T cell receptor (TCR) protein.
  • TCR T cell receptor
  • Embodiment 17 The population of embodiment 16, wherein at least 90% of engineered T cells of the population do not express a detectable level of TCR protein.
  • Embodiment 18 The population of any one of embodiments 13-17, wherein at least 50% of engineered T cells of the population do not express a detectable level of ⁇ 2M protein.
  • Embodiment 19 The population of embodiment 18, wherein at least 70% of engineered T cells of the population do not express a detectable level of ⁇ 2M protein.
  • Embodiment 20 The population of any one of embodiments 13-19, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells that express LIV1, induce cell lysis of at least 10%, at least 25%, or at least 50% of the cancer cells of the population.
  • Embodiment 21 The population of embodiment 20, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells that express LIV1, induce cell lysis of at least 70%, at least 80%, or at least 90% of the population of cancer cells.
  • Embodiment 22 The population of embodiments 20 or 21, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells, secrete IFN ⁇ .
  • Embodiment 23 The population of any one of embodiments 20-22, wherein the ratio of engineered T cells to cancer cells is 1:1 to 2:1.
  • Embodiment 24 The population of any one of embodiments 20-23, wherein the cancer cells comprise sarcoma cells.
  • Embodiment 25 The population of any one of embodiments 20-23, wherein the cancer cells comprise breast cancer cells.
  • Embodiment 27 The population of any one of embodiments 13-26, when administered in vivo to a subject, does not induce toxicity in the subject.
  • Embodiment 26 A method comprising administering the population of engineered T cells any one of embodiments 13-27 to a subject.
  • Embodiment 27 The method of embodiment 26, wherein the subject is a human subject.
  • Embodiment 28 The method of embodiment 27, wherein the subject has a cancer.
  • Embodiment 29 The method of embodiment 28, wherein the cancer is selected from the group consisting of: breast cancer, e.g., estrogen receptor-positive breast cancer, prostate cancer, squamous tumors, e.g., of the skin, bladder, lung, cervix, endometrium, head neck, and biliary tract, and/or neuronal tumors.
  • breast cancer e.g., estrogen receptor-positive breast cancer
  • prostate cancer e.g., squamous tumors, e.g., of the skin, bladder, lung, cervix, endometrium, head neck, and biliary tract, and/or neuronal tumors.
  • Embodiment 30 The method of embodiments 28 or 31, wherein the cancer comprises cancer cells expressing LIV1.
  • Embodiment 31 A method for producing an engineered T cell, the method comprising (a) delivering to a T cell, a RNA-guided nuclease, a gRNA targeting a TRAC gene, and a vector comprising a donor template that comprises a nucleic acid encoding a CAR that comprise an ectodomain that binds specifically to LIV1; and (b) producing an engineered T cell having a disrupted TRAC gene and expressing the CAR.
  • Embodiment 32 The method of embodiment 31, wherein the gRNA targeting the TRAC gene comprises the nucleotide sequence of SEQ ID NO: 18 or 19, or targets the nucleotide sequence of SEQ ID NO: 40.
  • Embodiment 33 The method of embodiments 31 or 32, wherein the nucleic acid encoding the CAR is flanked by left and right homology arms to the TRAC gene.
  • Embodiment 34 The method of any one of embodiments 31-33 further comprising delivering to the T cell a gRNA targeting the ⁇ 2M gene.
  • Embodiment 35 The method of embodiment 34, wherein the gRNA targeting the ⁇ 2M gene comprises the nucleotide sequence of SEQ ID NO: 20 or 21, or targets the nucleotide sequence of SEQ ID NO: 41.
  • Embodiment 36 The method of any one of embodiments 31-35, wherein the RNA-guided nuclease is a Cas9 nuclease, optionally a S. pyogenes Cas9 nuclease.
  • Embodiment 37 The method of any one of embodiments 31-38, wherein the ectodomain of the CAR is an anti-LIV1 antibody.
  • Embodiment 38 The method of embodiment 37, wherein the anti-LIV1 antibody is an anti-LIV1 single-chain variable fragment (scFv).
  • scFv anti-LIV1 single-chain variable fragment
  • Embodiment 39 The method of embodiment 38, wherein the anti-LIV1 scFv comprises the same VH complementarity determining regions (CDRs) and the same VL CDRs as a reference antibody, wherein the reference antibody comprises (i) a VH set forth as SEQ ID NO: 55 and a VL set forth as SEQ ID NO: 56.
  • CDRs VH complementarity determining regions
  • Embodiment 40 The method of embodiment 39, wherein the anti-LIV1 scFv comprises the same VH and VL chains as the reference antibody.
  • Embodiment 41 The method of embodiment 39, wherein the anti-LIV1 scFv comprises the amino acid sequence of any one of SEQ ID NOs: 54 or 70.
  • Embodiment 42 The method of any one of embodiments 31-41, wherein the CAR comprises a CD28 co-stimulatory domain or a 41BB co-stimulatory domain.
  • Embodiment 43 The method of embodiment 42, wherein the CAR further comprises a CD3z cytoplasmic signaling domain.
  • Embodiment 44 The method of any one of embodiments 31-43, wherein the donor template comprises the nucleotide sequence of any one of SEQ ID NOS: 63, 64, 71, or 72.
  • Embodiment 45 The method of any one of embodiments 31-44, wherein the CAR is encoded by a nucleotide sequence of any one of SEQ ID NOS: 49, 51, 65, or 67.
  • Embodiment A1 An engineered T cell comprising a nucleic acid encoding a chimeric antigen receptor (CAR), wherein the CAR comprises an ectodomain that binds specifically to LIV1.
  • CAR chimeric antigen receptor
  • Embodiment A2 The engineered T cell of embodiment A1 further comprising a disrupted T cell receptor alpha chain constant region (TRAC) gene.
  • T cell receptor alpha chain constant region TAC
  • Embodiment A3 The engineered T cell of embodiment A1 or A2 further comprising a disrupted beta-2-microglobulin ( ⁇ 2M) gene.
  • ⁇ 2M beta-2-microglobulin
  • Embodiment A4 The engineered T cell of any one of embodiments A1-3, wherein the ectodomain of the CAR comprises an anti-LIV1 antibody.
  • Embodiment A5 The engineered T cell of embodiment A4, wherein the anti-LIV1 antibody is an anti-LIV1 single-chain variable fragment (scFv).
  • scFv anti-LIV1 single-chain variable fragment
  • Embodiment A6 The engineered T cell of embodiment A5, wherein the anti-LIV1 scFv comprises the same heavy chain variable domain (VH) complementarity determining regions (CDRs) and the same light chain variable domain (VL) CDRs as a reference antibody, wherein the reference antibody comprises (i) a VH set forth as SEQ ID NO: 55 and a VL set forth as SEQ ID NO: 56 or (ii) a VH set forth as SEQ ID NO: 90 and a VL set forth as SEQ ID NO: 88.
  • VH heavy chain variable domain
  • CDRs complementarity determining regions
  • VL light chain variable domain
  • Embodiment A7 The engineered T cell of embodiment A6, wherein the anti-LIV1 scFv comprises the same VH and VL chains as the reference antibody.
  • Embodiment A8 The engineered T cell of embodiment A6, wherein the anti-LIV1 scFv comprises the amino acid sequence of any one of SEQ ID NOs: 54, 70, 83, or 86.
  • Embodiment A9 The engineered T cell of any one of embodiments A1-A8, wherein the CAR further comprises a CD28 co-stimulatory domain or a 41BB co-stimulatory domain.
  • Embodiment A10 The engineered T cell of embodiment A9, wherein the CAR further comprises a CD3 cytoplasmic signaling domain.
  • Embodiment A11 The engineered T cell of any one of embodiments A1-A10, wherein the CAR is encoded by the nucleotide sequence of any one of SEQ ID NOs: 49, 51, 104, or 108 or a nucleotide sequence comprising a nucleic acid sequence that is at least 90% identical to SEQ ID NOs: 49, 51, 104, or 108.
  • Embodiment A12 The engineered T cell of any one of embodiments A1-A11, wherein the nucleic acid encoding the CAR is inserted into the disrupted TRAC gene.
  • Embodiment A13 The engineered T cell of any one of embodiments A2-A12, wherein the disrupted TRAC gene comprises the nucleotide sequence of any one of SEQ ID NOs: 63, 64, 107, or 111, and/or the nucleotide sequence of any one of SEQ ID NOs: 49, 51, 104, or 108.
  • Embodiment A14 The engineered T cell of any one of embodiments A4-A13, wherein the disrupted ⁇ 2M gene comprises at least one nucleotide sequence selected from any one of SEQ ID NOs: 9-14.
  • Embodiment A15 An engineered T cell comprising: (i) a disrupted TRAC gene; (ii) a disrupted ⁇ 2M gene; and (iii) a nucleic acid encoding a CAR comprising an anti-LIV1 antigen-binding fragment.
  • Embodiment A16 The engineered T cell of embodiment A15, wherein the CAR comprises (a) an ectodomain that comprises an anti-LIV1 antigen-binding fragment, (b) a CD8 transmembrane domain, and (c) an endodomain that comprises a 41BB co-stimulatory domain and a CD3 ⁇ cytoplasmic signaling domain.
  • Embodiment A17 The engineered T cell of embodiments A15 or A16, wherein the disrupted TRAC gene comprises the nucleic acid encoding the CAR.
  • Embodiment A18 An engineered T cell comprising: (i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR comprising (a) an ectodomain that comprises an anti-LIV1 antigen-binding fragment, (b) a CD8 transmembrane domain, and (c) an endodomain that comprises a 41BB co-stimulatory domain and a CD3 ⁇ cytoplasmic signaling domain; and (ii) a disrupted ⁇ 2M gene.
  • a disrupted TRAC gene comprises a nucleic acid encoding a CAR comprising (a) an ectodomain that comprises an anti-LIV1 antigen-binding fragment, (b) a CD8 transmembrane domain, and (c) an endodomain that comprises a 41BB co-stimulatory domain and a CD3 ⁇ cytoplasmic signaling domain; and (ii) a disrupted ⁇ 2M gene.
  • Embodiment A19 An engineered T cell comprising: (i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR comprising an amino acid sequence of any one of SEQ ID NOs: 50, 52, 105, 109, 68 or 66; and (ii) a disrupted ⁇ 2M gene.
  • Embodiment A20 An engineered T cell comprising: (i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR, wherein the nucleic acid sequence is at least 90% identical to SEQ ID NOs: 49, 51, 104, or 108 and/or encodes a CAR comprising an amino acid sequence of any one of SEQ ID NOs: 50, 52, 105, 109, 68 or 66; and (ii) a disrupted ⁇ 2M gene.
  • Embodiment A21 The engineered T cell of any one of embodiments A1-A20, wherein the T cell is a human T cell.
  • Embodiment A22 A population of cells comprising the engineered T cell of any one of embodiments A1-A21, wherein at least 15% or at least 50% of engineered T cells of the population express the CAR.
  • Embodiment A23 The population of embodiment A22, wherein at least 30% of engineered T cells of the population express the CAR.
  • Embodiment A24 The population of embodiment A22, wherein at least 70% of engineered T cells of the population express the CAR.
  • Embodiment A25 The population of embodiment A22, wherein at least 25% of engineered T cells of the population express the CAR following at least 7 days or at least 14 days of in vitro proliferation.
  • Embodiment A26 The population of any one of embodiments A22-A25, wherein at least 50% of engineered T cells of the population do not express a detectable level of T cell receptor (TCR) protein.
  • TCR T cell receptor
  • Embodiment A27 The population of embodiments A26, wherein at least 90% of engineered T cells of the population do not express a detectable level of TCR protein.
  • Embodiment A28 The population of any one of embodiments A22-A27, wherein at least 50% of engineered T cells of the population do not express a detectable level of ⁇ 2M protein.
  • Embodiment A29 The population of embodiment A28, wherein at least 70% of engineered T cells of the population do not express a detectable level of ⁇ 2M protein.
  • Embodiment A30 The population of any one of embodiments A22-A29, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells that express LIV1, induce cell lysis of at least 10%, at least 25%, or at least 50% of the cancer cells of the population.
  • Embodiment A31 The population of embodiment A30, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells that express LIV1, induce cell lysis of at least 70%, at least 80%, or at least 90% of the population of cancer cells.
  • Embodiment A32 The population of embodiments A30 or A31, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells, secrete IFN ⁇ .
  • Embodiment A33 The population of any one of embodiments A30-A32, wherein the ratio of engineered T cells to cancer cells is 1:1 to 2:1.
  • Embodiment A34 The population of any one of embodiments A30-A33, wherein the cancer cells comprise sarcoma cells.
  • Embodiment A35 The population of any one of embodiments A30-A33, wherein the cancer cells comprise breast cancer cells.
  • Embodiment A36 The population of any one of embodiments A22-A35, when administered in vivo to a subject, does not induce toxicity in the subject.
  • Embodiment A37 A population of cells comprising engineered T cells, wherein the engineered T cells comprise: (i) a disrupted TRAC gene; (ii) a disrupted ⁇ 2M gene; and (iii) a nucleic acid encoding a CAR comprising an anti-LIV1 antigen-binding fragment.
  • Embodiment A38 The population of cells of embodiment A37, wherein the CAR comprises (a) an ectodomain that comprises an anti-LIV1 antigen-binding fragment, (b) a CD8 transmembrane domain, and (c) an endodomain that comprises a 41BB co-stimulatory domain and a CD3 ⁇ cytoplasmic signaling domain.
  • Embodiment A39 The population of cells of embodiments A37 or A38, wherein the disrupted TRAC gene comprises the nucleic acid encoding the CAR.
  • Embodiment A40 A population of cells comprising engineered T cells, wherein the engineered T cells comprise: (i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR comprising (a) an ectodomain that comprises an anti-LIV1 antigen-binding fragment, (b) a CD8 transmembrane domain, and (c) an endodomain that comprises a 41BB co-stimulatory domain and a CD3 ⁇ cytoplasmic signaling domain; and (ii) a disrupted ⁇ 2M gene.
  • a disrupted TRAC gene comprises a nucleic acid encoding a CAR comprising (a) an ectodomain that comprises an anti-LIV1 antigen-binding fragment, (b) a CD8 transmembrane domain, and (c) an endodomain that comprises a 41BB co-stimulatory domain and a CD3 ⁇ cytoplasmic signaling domain; and (ii)
  • Embodiment A41 A population of cells comprising engineered T cells, wherein the engineered T cells comprise: (i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR, wherein the nucleic acid sequence is at least 90% identical to SEQ ID NOs: 49, 51, 104, or 108 and/or encodes the CAR of SEQ ID NOs: 50, 52, 105, 109, 68 or 66; and (ii) a disrupted ⁇ 2M gene.
  • Embodiment A42 A method comprising administering the population of engineered T cells any one of embodiments A22-A41 to a subject.
  • Embodiment A43 The method of embodiment A42, wherein the subject is a human subject.
  • Embodiment A44 The method of embodiment A43, wherein the subject has a cancer.
  • Embodiment A45 The method of embodiment A44, wherein the cancer is selected from the group consisting of: breast cancer, e.g., estrogen receptor-positive breast cancer, prostate cancer, squamous tumors, e.g., of the skin, bladder, lung, cervix, endometrium, head neck, and biliary tract, and/or neuronal tumors.
  • breast cancer e.g., estrogen receptor-positive breast cancer
  • prostate cancer e.g., squamous tumors, e.g., of the skin, bladder, lung, cervix, endometrium, head neck, and biliary tract, and/or neuronal tumors.
  • Embodiment A46 The method of embodiments A44 or A45, wherein the cancer comprises cancer cells expressing LIV1.
  • Embodiment A47 A method for producing an engineered T cell, the method comprising (a) delivering to a T cell (i) a RNA-guided nuclease, (ii) a gRNA targeting a TRAC gene, and (iii) a vector comprising a donor template that comprises a nucleic acid encoding a CAR that comprise an ectodomain that binds specifically to LIV1; and (b) producing an engineered T cell having a disrupted TRAC gene and expressing the CAR.
  • Embodiment A48 The method of embodiments A47, wherein the gRNA targeting the TRAC gene comprises the nucleotide sequence of SEQ ID NO: 18 or 19, or targets the nucleotide sequence of SEQ ID NO: 40.
  • Embodiment A49 The method of embodiments A47 or A48 further comprising delivering to the T cell a gRNA targeting the ⁇ 2M gene.
  • Embodiment A50 The method of embodiments A49, wherein the gRNA targeting the ⁇ 2M gene comprises the nucleotide sequence of SEQ ID NO: 20 or 21, or targets the nucleotide sequence of SEQ ID NO: 41.
  • Embodiment A51 The method of any one of embodiments A47-A50, wherein the ectodomain of the CAR comprises an anti-LIV1 antibody.
  • Embodiment A52 The method of embodiment A51, wherein the anti-LIV1 antibody is an anti-LIV1 single-chain variable fragment (scFv).
  • scFv anti-LIV1 single-chain variable fragment
  • Embodiment A53 The method of embodiment A52, wherein the anti-LIV1 scFv comprises the same heavy chain variable domain (VH) complementarity determining regions (CDRs) and the same light chain variable domain (VL) CDRs as a reference antibody, wherein the reference antibody comprises (i) a VH set forth as SEQ ID NO: 55 and a VL set forth as SEQ ID NO: 56, or (ii) a VH set forth as SEQ ID NO: 90 and a VL set forth as SEQ ID NO: 88.
  • VH heavy chain variable domain
  • CDRs complementarity determining regions
  • VL light chain variable domain
  • Embodiment A54 The method of embodiment A53, wherein the anti-LIV1 scFv comprises the same VH and VL chains as the reference antibody.
  • Embodiment A55 The method of embodiment A53, wherein the anti-LIV1 scFv comprises the amino acid sequence of any one of SEQ ID NOs: 54, 83, 86 or 70.
  • Embodiment A56 The method of any one of embodiments A47-A56, wherein the CAR further comprises a CD28 co-stimulatory domain or a 41BB co-stimulatory domain.
  • Embodiment A57 The method of embodiment A56, wherein the CAR further comprises a CD3 ⁇ cytoplasmic signaling domain.
  • Embodiment A58 The method of any one of embodiments A47-A57, wherein the CAR is encoded by a nucleotide sequence of any one of SEQ ID NOs: 49, 51, 104, or 108 or a nucleotide sequence comprising a nucleic acid sequence that is at least 90% identical to SEQ ID NOs: 49, 51, 104, or 108.
  • Embodiment A59 The method of any one of embodiments A47-A58, wherein the nucleic acid encoding the CAR is flanked by left and right homology arms to the TRAC gene.
  • Embodiment A60 The method of any one of embodiments A47-A59, wherein the donor template comprises the nucleotide sequence of any one of SEQ ID NOs: 63, 64, 107, or 111.
  • Embodiment A61 The method of any one of embodiments A47-A60, wherein the RNA-guided nuclease is a Cas9 nuclease, optionally a S. pyogenes Cas9 nuclease.
  • Embodiment A62 An engineered T cell produced by the method of any one of embodiments A47-A61.
  • Embodiment A63 A population of cells comprising the engineered T cell of embodiment A62.
  • Embodiment A64 A method of treating cancer in a subject, comprising administering to the subject the population of cells of any one of embodiments A22-A41 or A63.
  • Embodiment A65 The method of embodiment A64, wherein the cancer is selected from the group consisting of: pancreatic cancer, gastric cancer, ovarian cancer, uterine cancer, breast cancer, prostate cancer, testicular cancer, thyroid cancer, nasopharyngeal cancer, non-small cell lung (NSCLC), glioblastoma, neuronal, soft tissue sarcomas, leukemia, lymphoma, melanoma, colon cancer, colon adenocarcinoma, brain glioblastoma, hepatocellular carcinoma, liver hepatocholangiocarcinoma, osteosarcoma, gastric cancer, esophagus squamous cell carcinoma, advanced stage pancreas cancer, lung adenocarcinoma, lung squamous cell carcinoma, lung small cell cancer, renal carcinoma, and intrahepatic biliary cancer.
  • NSCLC non-small cell lung
  • glioblastoma neuronal
  • Embodiment A66 The method of embodiments A64 or A65, wherein the cancer comprises cancer cells expressing LIV1.
  • Activated primary human T cells were electroporated with Cas9:gRNA RNP complexes and adeno-associated adenoviral vectors (AAVs) to generate TRAC ⁇ / ⁇ 2M ⁇ /anti-Liv1a CAR + T cells.
  • Cas9:gRNA RNP complexes and adeno-associated adenoviral vectors (AAVs) to generate TRAC ⁇ / ⁇ 2M ⁇ /anti-Liv1a CAR + T cells.
  • AAVs adeno-associated adenoviral vectors
  • AAV serotype 6 comprising one of the nucleotide sequences encoding an anti-Livia CAR (971 (SEQ ID NO:49), 972 (SEQ ID NO: 65), 972b (SEQ ID NO: 67), 973 (SEQ ID NO: 95), 974 (SEQ ID NO: 100), 975 (SEQ ID NO: 104), and 976 (SEQ ID NO: 108), were delivered with Cas9:sgRNA RNPs (1 ⁇ M Cas9, 5 ⁇ M gRNA) to activated allogeneic human T cells.
  • the following sgRNAs were used: TRAC (SEQ ID NO: 28) and ⁇ 2M (SEQ ID NO: 30).
  • the unmodified versions (or other modified versions) of the gRNAs may also be used (e.g., SEQ ID NO: 18 or 20).
  • a cell killing (cytotoxicity) assay was used to assess the ability of the TRAC-/ ⁇ 2M-/anti-Livia CAR+ T cells to cause cellular lysis in adherent kidney carcinoma and breast cancer cell lines (A498 and ZR-75-1, respectively).
  • Adherent cells were seeded in 96-well plates at 50,000 cells per well and left overnight at 37° C. During the following day, T cells were added to the wells containing target cells at ratios of 8:1, 4:1, 2:1 or 1:1 T cell:target cell.
  • TRAC-/ ⁇ 2M-T cells were used as a negative control.
  • MILLIPLEX® standard and quality control (QC) samples were reconstituted, and serial dilutions of the working standards from 10,000 pg/mL to 3.2 pg/mL were prepared.
  • MILLIPLEX® standards, QCs and cell supernatants were added to each plate, and assay media was used to dilute the supernatants. All samples were incubated with anti-human IFN ⁇ and anti-human IL-2 beads for 2 hours. After incubation, the plate was washed using an automated magnetic plate washer. Human cytokine/chemokine detection antibody solution was added to each well and incubated for 1 hour followed by incubation with Streptavidin-Phycoerythrin for 30 minutes. The plate was subsequently washed, samples were resuspended with 150 ⁇ L Sheath Fluid, and agitated on a plate shaker for 5 minutes.
  • the samples were read using the Luminex® 100/200TM instrument with xPONENT® software and data acquisition and analysis was completed using MILLIPLEX® Analyst software.
  • the Median Fluorescent Intensity (MFI) data was automatically analyzed using a 5-parameter logistic curve-fitting method for calculating the cytokine concentration measured in the unknown samples.
  • allogeneic T cells containing the CTX971, 975, or 976 CARs secreted the effector cytokines interferon- ⁇ ( 3 A, B) and interleukin-2 ( 3 C, D) when co-cultured with the target cells lines A498 and ZR-75-1 at levels significantly above background (2KO/AAV neg T cells co-cultured with the target cell lines).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Cell Biology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biomedical Technology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Mycology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hospice & Palliative Care (AREA)
  • Virology (AREA)
  • Urology & Nephrology (AREA)
  • Endocrinology (AREA)

Abstract

Provided herein, in some embodiments, are methods and compositions (e.g., cell compositions) for the treatment of cancer, such as LIV1+ malignancies.

Description

    RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Application No. 62/756,723, filed Nov. 7, 2018, the disclosure of which is hereby incorporated by reference in its entirety.
  • INCORPORATION BY REFERENCE OF MATERIAL SUBMITTED ELECTRONICALLY
  • The Sequence Listing, which is a part of the present disclosure, is submitted concurrently with the specification as a text file. The name of the text file containing the Sequence Listing is “CT113_Seqlisting.txt”, which was created on Nov. 7, 2019 and is 191,166 bytes in size. The subject matter of the Sequence Listing is incorporated herein in its entirety by reference.
  • BACKGROUND
  • Chimeric antigen receptor (CAR) T-cell therapy uses genetically-modified T cells to more specifically and efficiently target and kill cancer cells. After T cells have been collected from the blood, the cells are engineered to include CARs on their surface. The CARs may be introduced into the T cells using CRISPR/Cas9 gene editing technology. When these allogeneic CAR T cells are injected into a patient, the receptors enable the T cells to kill cancer cells.
  • SUMMARY
  • LIV1, a member of the ZIP family of highly conserved transmembrane zinc transporter proteins, is expressed at elevated levels in estrogen receptor-positive breast cancer and tumors of the lymph nodes. Further aberrant expression of zinc transporters such as LIV1 is known to lead to deregulated Zn intake or deficiency, leading to uncontrolled growth such that occur in cancer. Thus, LIV1 is a desirable transmembrane protein for targeting cancer. In fact, the LIV-1 protein has been implicated in breast cancer, prostate cancer, squamous tumors, and neuronal tumors.
  • Some aspects of the present disclosure provide an engineered T cell comprising a nucleic acid encoding a chimeric antigen receptor (CAR), wherein the CAR comprise an ectodomain that binds specifically to LIV1. In some embodiments, the engineered T cell further comprises a disrupted T cell receptor alpha chain constant region (TRAC) gene. For example, the TRAC gene may be disrupted by insertion of the nucleic acid encoding a CAR. In some embodiments, the engineered T cell further comprises a disrupted beta-2-microglobulin (β2M) gene.
  • The ectodomain of the CAR, in some embodiments, comprises an anti-LIV1 antibody. In some embodiments, the anti-LIV1 antibody is an anti-LIV1 single-chain variable fragment (scFv). The anti-LIV1 scFv, in some embodiments, comprises an amino acid sequence of any one of SEQ ID NO: 54, 70, 83 or 86. In some embodiments, the anti-LIV1 scFv comprises a heavy chain variable region (VH) comprising an amino acid sequence of any one of SEQ ID NO: 55 or 90 and/or a light chain variable region (VL) comprising an amino acid sequence of any one of SEQ ID NO: 56 or 88. In some embodiments, the anti-LIV1 scFv comprises a VH comprising CDR amino acid sequences of SEQ ID NO: 57, SEQ ID NO: 58, and/or SEQ ID NO: 59; and/or the anti-LIV1 scFv comprises a VL sequence comprising CDR amino acid sequences of SEQ ID NO: 60, SEQ ID NO: 61, and/or SEQ ID NO: 62.
  • The CAR, in some embodiments, comprises a CD3ζ cytoplasmic signaling domain. In some embodiments, the CAR comprises a CD28 co-stimulatory domain or a 41BB co-stimulatory domain.
  • In some embodiments, the TRAC gene comprises the nucleotide sequence of any one of SEQ ID NOs: 63, 64, 107, or 111, and/or wherein the CAR comprises the nucleotide sequence of any one of SEQ ID NOs: 49, 51, 104, or 108. In some embodiments, the disrupted β2M gene comprises at least one nucleotide sequence selected from any one of SEQ ID NOs: 9-14.
  • Also provided herein, in some aspects, is a population of engineered T cells (e.g., comprising a nucleic acid encoding an anti-LIV1 CAR), wherein at least 25% or at least 50% of engineered T cells of the population express the CAR. In some aspects, at least 15% or at least 50% of engineered T cells of the population express the CAR. For example, at least 70% of engineered T cells of the population express the CAR. In another example, at least 30% of engineered T cells of the population express the CAR.
  • In some embodiments, at least 25% of engineered T cells of the population express the CAR following at least 7 or at least 14 days of in vitro proliferation.
  • In some embodiments, at least 50% of engineered T cells of the population do not express a detectable level of T cell receptor (TCR) protein. For example, at least 90% of engineered T cells of the population may not express a detectable level of TCR protein.
  • In some embodiments, at least 50% of engineered T cells of the population do not express a detectable level of β2M protein. For example, at least 70% of engineered T cells of the population may not express a detectable level of β2M protein.
  • In some embodiments, engineered T cells of the population, when co-cultured in vitro with a population of cancer cells that express LIV1, induce cell lysis of at least 50% of the cancer cells of the population. For example, engineered T cells of the population may induce cell lysis of at least 70%, at least 80%, or at least 90% of the cancer cells of the population. In some embodiments, engineered T cells of the population, when co-cultured in vitro with a population of cancer cells, secrete IFNγ. In some embodiments, the ratio of engineered T cells to cancer cells is 1:1 to 2:1. The cancer cells may be, for example, sarcoma cells or breast cancer cells. Other cancer cells may be targeted.
  • In some embodiments, proliferative capacity of engineered T cells of the population is within 10% of proliferative capacity of control cells.
  • Other aspects of the present disclosure provide a method that comprises administering the population of engineered T cells as described herein. In some embodiments, percent body weight of the subject, following 5-10 days of administration, is within 10% of initial body weight of the subject, wherein initial body weight of the subject is body weight of the subject at the time of administration. In some embodiments, the subject is a human subject. In some embodiments, the subject has a cancer. The cancer may express LIV1, for example.
  • Further aspects of the present disclosure provide a method for producing an engineered T cell, the method comprising (a) delivering to a T cell a RNA-guided nuclease, a gRNA targeting a TRAC gene, and a vector comprising a donor template that comprises a nucleic acid encoding a CAR that comprise an ectodomain that binds specifically to LIV1, wherein the nucleic acid encoding the CAR is flanked by left and right homology arms to the TRAC gene, and (b) producing an engineered T cell. In some embodiments, the gRNA targeting the TRAC gene comprises the nucleotide sequence of SEQ ID NO: 18 or 19, or targets the nucleotide sequence of SEQ ID NO: 40.
  • In some embodiments, the method further comprises delivering to the T cell a gRNA targeting the β2M gene. In some embodiments, the gRNA targeting the β2M gene comprises the nucleotide sequence of SEQ ID NO: 20 or 21, or targets the nucleotide sequence of SEQ ID NO: 41.
  • In some embodiments, the RNA-guided nuclease is a Cas9 nuclease, optionally a S. pyogenes Cas9 nuclease.
  • In some embodiments, the donor template comprises the nucleotide sequence of any one of SEQ ID NOs: 63, 64, 107, or 111.
  • In some embodiments, the CAR comprises the nucleotide sequence of any one of SEQ ID NOs: 49, 51, 104, or 108.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows flow cytometry results to assess TRAC, β2M, and anti-Livia CAR expression levels at the cell surface of the edited cell population.
  • FIGS. 2A-2B show that anti-Livia CAR T cells, particularly those expressing the CTX971, CTX975 and CTX976 constructs, exhibited potent cytotoxicity towards the A498 (FIG. 2A) and ZR-75-1 (FIG. 2B) cell lines.
  • FIGS. 3A-3D show cytokine secretion of anti-Liv1a CAR T cells when co-cultured with target cell lines A498 and ZR-75-1. FIGS. 3A and 3B show secretion of the effector cytokine interferon-γ (IFN-γ) in A498 (FIG. 3A) and ZR-75-1 (FIG. 3B) cell lines. FIGS. 3C and 3D show secretion of the effector cytokine interleukin-2 (IL-2) in A498 (FIG. 3C) and ZR-75-1 (FIG. 3D) cell lines.
  • FIG. 4 shows an alignment of scFV constructs (VL and VH)—971 (SEQ ID NO: 54); 973 (SEQ ID NO: 82); 975 (SEQ ID NO: 83); 977 (SEQ ID NO: 84)
  • FIG. 5 shows an alignment of scFV constructs (VH and VL)—979 (SEQ ID NO: 70); 974 (SEQ ID NO: 85); 976 (SEQ ID NO: 86); 978 (SEQ ID NO: 87), 972 (SEQ ID NO: 127)
  • DETAILED DESCRIPTION LIV1 Cancer Antigen
  • In some embodiments, the T cells of the present disclosure are engineered with a chimeric antigen receptor (CAR) designed to target LIV1. LIV1, also known as Solute Carrier Family 39 Member 6, SLC39A6, ZIP6, and LIV-1, is a member of the ZIP family of highly conserved transmembrane zinc transporter proteins. LIV1 is expressed at elevated levels in breast cancer, e.g., estrogen receptor-positive breast cancer, prostate cancer, squamous tumors, e.g., of the skin, bladder, lung, cervix, endometrium, head neck, and biliary tract, and neuronal tumors. Notably, LIV1 has a restricted expression in normal tissues, e.g., non-cancerous breast, prostate, and testis. Thus, LIV1 is a desirable transmembrane protein for targeting cancer. In fact, the LIV-1 protein has been implicated in breast cancer, prostate cancer, squamous tumors, and neuronal tumors.
  • Thus, in some embodiments, T cells of the present disclosure are engineered to express a CAR comprising an anti-LIV1 antibody (e.g., anti-LIV1 scFv). In some embodiments, the anti-LIV1 antibody is an anti-LIV1 scFv encoded by the sequence of any one of SEQ ID NOs: 53, 69, 97, 102, 106, 110, 114, or 118. In some embodiments, the anti-LIV1 antibody is an anti-LIV1 scFv comprising the sequence of any one of SEQ ID NOs: 54, 70, 82, 83, 84, 85, 86, or 87. In some embodiments, the anti-LIV1 antibody is an anti-LIV1 scFv comprising a VH comprising an amino acid sequence of any one of SEQ ID NO: 55, 90 or 98. In some embodiments, the anti-LIV1 antibody is an anti-LIV1 scFv comprising a VL comprising an amino acid sequence of any one of SEQ ID NO: 56, 88 or 128. In some embodiments, a CAR comprising an anti-LIV1 antibody is encoded by the sequence of any one of SEQ ID NOs: 49, 51, 65, 67, 95, 100, 104, 108, 112, or 116. In some embodiments, a CAR comprising an anti-LIV7 antibody is encoded by a sequence comprising a nucleic acid that is at least 90% identical to SEQ ID NOs: 49, 51, 65, 67, 95, 100, 104, 108, 112, or 116. In some embodiments, a CAR comprising an anti-LIV1 antibody comprises the sequence of any one of SEQ ID NOs: 49, 51, 65, 67, 95, 100, 104, 108, 112, or 116. In some embodiments, a CAR comprising an anti-LIV1 antibody comprises an anti-LIV1 antibody as described in U.S. Pat. No. 9,228,026.
  • Multi-Gene Editing
  • The engineered T cells of the present disclosure, in some embodiments, include more than one gene edit, for example, in more than one gene. For example, an engineered T cell may comprise a disrupted T cell receptor alpha chain constant region (TRAC) gene, a disrupted beta-2-microglobulin (β2M) gene, a disrupted programmed cell death-1 (PD-1 or PDCD1) gene, a disrupted CD70 gene, or any combination of two or more of the foregoing disrupted genes. In some embodiments, an engineered T cell comprises a disrupted TRAC gene, a disrupted β2M gene, and a disrupted CD70 gene. In some embodiments, an engineered T cell comprises a disrupted TRAC gene, a disrupted β2M gene, and a disrupted PD-1 gene. In some embodiments, an engineered T cell comprises a disrupted TRAC gene, a disrupted β2M gene, a disrupted CD70 gene and a disrupted PD-1 gene.
  • It should be understood that gene disruption encompasses gene modification through gene editing (e.g., using CRISPR/Cas gene editing to insert or delete one or more nucleotides). In some embodiments, a disrupted gene is a gene that does not encode functional protein. In some embodiments, a cell that comprises a disrupted gene does not express (e.g., at the cell surface) a detectable level (e.g. by antibody, e.g., by flow cytometry) of the protein encoded by the gene. A cell that does not express a detectable level of the protein may be referred to as a knockout cell. For example, a cell having β2M gene edit may be considered a β2M knockout cell if β2M protein cannot be detected at the cell surface using an antibody that specifically binds β2M protein.
  • Provided herein, in some embodiments, are populations of cells in which a certain percentage of the cells has been edited (e.g., β2M gene edited), resulting in a certain percentage of cells not expressing a particular gene and/or protein. In some embodiments, at least 50% (e.g., 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 85%) of the cells of a gene-edited population of cells are β2M knockout cells. In some embodiments, at least 50% of the cells (e.g. T cells) of the population do not express detectable levels of β2M protein. In some embodiments, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% of the cells of a gene-edited population of cells may be β2M knockout cells.
  • Methods of using CRISPR-Cas gene editing technology to create a genomic deletion in a cell (e.g., to knock out a gene in a cell) are known (Bauer D E et al. Vis. Exp. 2015; 95; e52118).
  • TRAC Gene Edit
  • In some embodiments, an engineered T cell comprises a disrupted TRAC gene. This disruption leads to loss of function of the TCR and renders the engineered T cell non-alloreactive and suitable for allogeneic transplantation, minimizing the risk of graft versus host disease. In some embodiments, expression of the endogenous TRAC gene is eliminated to prevent a graft-versus-host response. In some embodiments, a disruption in the TRAC gene expression is created by knocking a chimeric antigen receptor (CAR) into the TRAC gene (e.g., using an adeno-associated viral (AAV) vector and donor template). In some embodiments, a disruption in the TRAC gene expression is created by gRNAs targeting the TRAC genomic region. In some embodiments, a genomic deletion in the TRAC gene is created by knocking a chimeric antigen receptor (CAR) into the TRAC gene (e.g., using an AAV vector and donor template). In some embodiments, a disruption in the TRAC gene expression is created by gRNAs targeting the TRAC genomic region and knocking a chimeric antigen receptor (CAR) into the TRAC gene.
  • Non-limiting examples of modified and unmodified TRAC gRNA sequences that may be used as provided herein to create a genomic disruption in the TRAC gene are listed in Table 4 (e.g., SEQ ID NOs: 18 and 19). See also International Application No. PCT/US2018/032334, filed May 11, 2018, incorporated herein by reference. Other gRNA sequences may be designed using the TRAC gene sequence located on chromosome 14 (GRCh38: chromosome 14: 22,547,506-22,552,154; Ensembl; ENSG00000277734). In some embodiments, gRNAs targeting the TRAC genomic region create Indels in the TRAC gene disrupting expression of the mRNA or protein.
  • In some embodiments, at least 50% of a population of engineered T cells do not express a detectable level of T cell receptor (TCR) surface protein. For example, at least 55%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% of a population may not express a detectable level of TCR surface protein. In some embodiments, 50%-100%, 50%-90%, 50%-80%, 50%-70%, 50%-60%, 60%-100%, 60%-90%, 60%-80%, 60%-70%, 70%-100%, 70%-90%, 70%-80%, 80%-100%, 80%-90%, or 90%-100% of the population of engineered T cells do not express a detectable level of TCR surface protein.
  • In some embodiments, gRNAs targeting the TRAC genomic region create Indels in the TRAC gene comprising at least one nucleotide sequence selected from the following sequences in Table 1:
  • TABLE 1
    SEQ
    ID
    Sequence NO:
    AAGAGCAACAAATCTGACT 1
    AAGAGCAACAGTGCTGTGCCTGGAGCAACAAATCTGACT 2
    AAGAGCAACAAATCTGACT
    AAGAGCAACAGTGCTGGAGCAACAAATCTGACT 3
    AAGAGCAACAAATCTGACT
    AAGAGCAACAGTGCCTGGAGCAACAAATCTGACT
    4
    AAGAGCAACAAATCTGACT
    AAGAGCAACAGTGCTGACTAAGAGCAACAAATCTGACT
    5
    AAGAGCAACAGTGCTGTGGGCCTGGAGCAACAAATCTGA 6
    CTAAGAGCAACAAATCTGACT
    AAGAGCAACAGTGCTGGCCTGGAGCAACAAATCTGACT 7
    AAGAGCAACAAATCTGACT
    AAGAGCAACAGTGCTGTGTGCCTGGAGCAACAAATCTGA
    8
    CTAAGAGCAACAAATCTGACT
  • In some embodiments, an engineered T cell comprises a deletion in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion of 15-30 base pairs in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion of 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 base pairs in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion of more than 30 base pairs in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion of 20 base pairs in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion of SEQ ID NO: 92 (AGAGCAACAGTGCTGTGGCC) in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion comprising SEQ ID NO: 92 (AGAGCAACAGTGCTGTGGCC) in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion of SEQ ID NO: 40 in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion comprising SEQ ID NO: 40 in the TRAC gene relative to unmodified T cells.
  • β2M Gene Edit
  • In some embodiments, an engineered T cell comprises a disrupted β2M gene. β2M is a common (invariant) component of MHC I complexes. Disrupting its expression by gene editing will prevent host versus therapeutic allogeneic T cells responses leading to increased allogeneic T cell persistence. In some embodiments, expression of the endogenous β2M gene is eliminated to prevent a host-versus-graft response.
  • Non-limiting examples of modified and unmodified β2M gRNA sequences that may Non-limiting examples of modified and unmodified β2M gRNA sequences that may be used as provided herein to create a genomic disruption in the β2M gene are listed in Table 4 (e.g., SEQ ID NOs: 20 and 21). See also International Application No. PCT/US2018/032334, filed May 11, 2018, incorporated herein by reference. Other gRNA sequences may be designed using the β2M gene sequence located on Chromosome 15 (GRCh38 coordinates: Chromosome 15: 44,711,477-44,718,877; Ensembl: ENSG00000166710).
  • In some embodiments, gRNAs targeting the β2M genomic region create Indels in the β2M gene disrupting expression of the mRNA or protein.
  • In some embodiments, at least 50% of the engineered T cells of a population of engineered T cells does not express a detectable level of β2M surface protein. For example, at least 55%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% of the engineered T cells of a population may not express a detectable level of β2M surface protein. In some embodiments, 50%-100%, 50%-90%, 50%-80%, 50%-70%, 50%-60%, 60%-100%, 60%-90%, 60%-80%, 60%-70%, 70%-100%, 70%-90%, 70%-80%, 80%-100%, 80%-90%, or 90%-100% of the engineered T cells of a population does not express a detectable level of β2M surface protein.
  • In some embodiments, an edited β2M gene comprises at least one nucleotide sequence selected from the following sequences in Table 2.
  • TABLE 2
    SEQ
    ID
    Sequences NO:
    CGTGGCCTTAGCTGTGCTCGCGCTACTCTCTCTTTCTGCCTGGA  9
    GGCTATCCAGCGTGAGTCTCTCCTACCCTCCCGCT
    CGTGGCCTTAGCTGTGCTCGCGCTACTCTCTCTTTCGCCTGGAG 10
    GCTATCCAGCGTGAGTCTCTCCTACCCTCCCGCT
    CGTGGCCTTAGCTGTGCTCGCGCTACTCTCTCTTTCTGGAGGCT
    11
    ATCCAGCGTGAGTCTCTCCTACCCTCCCGCT
    CGTGGCCTTAGCTGTGCTCGCGCTACTCTCTCTTTCTGGATAGC 12
    CTGGAGGCTATCCAGCGTGAGTCTCTCCTACCCTCCCGCT
    CGTGGCCTTAGCTGTGCTCGCGCTATCCAGCGTGAGTCTCTCCT 13
    ACCCTCCCGCT
    CGTGGCCTTAGCTGTGCTCGCGCTACTCTCTCTTTCTGTGGCCT 14
    GGAGGCTATCCAGCGTGAGTCTCTCCTACCCTCCCGCT
  • PD-1 Gene Edit
  • PD-1 is an immune checkpoint molecule that is upregulated in activated T cells and serves to dampen or stop T cell responses. Disrupting PD-1 by gene editing could lead to more persistent and/or potent therapeutic T cell responses and/or reduce immune suppression in a subject. In some embodiments, an engineered T cell comprises a disrupted PD-1 gene. In some embodiments, expression of the endogenous PD-1 gene is eliminated to enhance anti-tumor efficacy of the CAR T cells of the present disclosure.
  • Non-limiting examples of modified and unmodified PD-1 gRNA sequences that may be used as provided herein to create a genomic deletion in the PD-1 gene are listed in Table 4 (e.g., SEQ ID NOs: 22 and 23). See also International Application No. PCT/US2018/032334, filed May 11, 2018, incorporated herein by reference. Other gRNA sequences may be designed using the PD-1 gene sequence located on Chromosome 2 (GRCh38 coordinates: Chromosome 2: 241,849,881-241,858,908; Ensembl: ENSG00000188389).
  • In some embodiments, gRNAs targeting the PD-1 genomic region create Indels in the PD-1 gene disrupting expression of the PD-1 mRNA or protein.
  • In some embodiments, an engineered T cell comprises a disrupted PD-1 gene. In some embodiments, at least 50% of the engineered T cells of a population of engineered T cells does not express a detectable level of PD-1 surface protein. For example, at least 55%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% of the engineered T cells of a population may not express a detectable level of PD-1 surface protein. In some embodiments, 50%-100%, 50%-90%, 50%-80%, 50%-70%, 50%-60%, 60%-100%, 60%-90%, 60%-80%, 60%-70%, 70%-100%, 70%-90%, 70%-80%, 80%-100%, 80%-90%, or 90%-100% of the engineered T cells of a population does not express a detectable level of PD-1 surface protein.
  • CD70 Gene Edit
  • Cluster of Differentiation 70 (CD70) is a member of the tumor necrosis factor superfamily and its expression is restricted to activated T and B lymphocytes and mature dendritic cells. CD70 has also been detected on hematological tumors and on carcinomas. CD70 is implicated in tumor cell and regulatory T cell survival through interaction with its ligand, CD27. Disrupting CD70 by gene editing increases cell expansion and reduces cell exhaustion. In some embodiments, an engineered T cell comprises a disrupted CD70 gene. In some embodiments, expression of the endogenous CD70 gene is eliminated to enhance anti-tumor efficacy of the CAR T cells of the present disclosure. In some embodiments, gRNAs targeting the CD70 genomic region create Indels in, or around, the CD70 gene disrupting expression of the CD70 mRNA and/or protein.
  • Non-limiting examples of modified and unmodified CD70 gRNA sequences that may be used as provided herein to create a genomic disruption in the CD70 gene are listed in Table 4 (e.g., SEQ ID NOs: 24-27). Other gRNA sequences may be designed using the CD70 gene sequence located on Chromosome 19 (GRCh38 coordinates: Chromosome 19: 6,583,183-6,604,103; Ensembl: ENSG00000125726).
  • In some embodiments, an engineered T cell comprises a disrupted CD70 gene. In some embodiments, at least 50% of the engineered T cells of a population of engineered T cells does not express a detectable level of CD70 surface protein. For example, at least 55%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% of the engineered T cells of a population may not express a detectable level of CD70 surface protein. In some embodiments, 50%-100%, 50%-90%, 50%-80%, 50%-70%, 50%-60%, 60%-100%, 60%-90%, 60%-80%, 60%-70%, 70%-100%, 70%-90%, 70%-80%, 80%-100%, 80%-90%, or 90%-100% of the engineered T cells of a population does not express a detectable level of CD70 surface protein.
  • Cellular Phenotypes
  • In some embodiments, one or more gene edits within a population of cells results in a phenotype associated with changes in cellular proliferative capacity, cellular exhaustion, cellular viability, cellular lysis capability (e.g., increase cytokine production and/or release), or any combination thereof.
  • In some embodiments, engineered T cells of the present disclosure exhibit at least 20% greater cellular proliferative capacity, relative to control T cells. For example, engineered T cells may exhibit at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, or at least 90% greater cellular proliferative capacity, relative to control T cells. In some embodiments, engineered T cells of the present disclosure exhibit 20%-100%, 20%-90%, 20%-80%, 20%-70%, 20%-60%, 20%-50%, 30%-100%, 30%-90%, 30%-80%, 30%-70%, 30%-60%, 30%-50%, 40%-100%, 40%-90%, 40%-80%, 40%-70%, 40%-60%, 40%-50%, 50%-100%, 50%-90%, 50%-80%, 50%-70%, or 50%-60% greater cellular proliferative capacity, relative to control T cells.
  • In some embodiments, engineered T cells of the present disclosure exhibit an at least 20% increase in cellular viability, relative to control cells. For example, engineered T cells of the present disclosure may exhibit at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, or at least 90% increase in cellular viability, relative to control cells. In some embodiments, engineered T cells of the present disclosure exhibit a 20%-100%, 20%-90%, 20%-80%, 20%-70%, 20%-60%, 20%-50%, 30%-100%, 30%-90%, 30%-80%, 30%-70%, 30%-60%, 30%-50%, 40%-100%, 40%-90%, 40%-80%, 40%-70%, 40%-60%, 40%-50%, 50%-100%, 50%-90%, 50%-80%, 50%-70%, or 50%-60% increase in cellular viability, relative to control cells.
  • In some embodiments, engineered T cells of the present disclosure exhibit an at least 20% increase in cellular lysis capability (kill at least 20% more target cells), relative to control cells. For example, engineered T cells of the present disclosure may exhibit an at least at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, or at least 90% increase in cellular lysis capability, relative to control cells. In some embodiments, engineered T cells of the present disclosure exhibit a 20%-100%, 20%-90%, 20%-80%, 20%-70%, 20%-60%, 20%-50%, 30%-100%, 30%-90%, 30%-80%, 30%-70%, 30%-60%, 30%-50%, 40%-100%, 40%-90%, 40%-80%, 40%-70%, 40%-60%, 40%-50%, 50%-100%, 50%-90%, 50%-80%, 50%-70%, or 50%-60% increase in cellular lysis capability, relative to control cells. For example, the level of cytokines (e.g., IL-2 and/or IFN-gamma) secreted by the engineered T cells may at least 2-fold (e.g., at least 3-fold, at least 4-fold, or at least 5-fold) greater than the level of cytokines secreted by control T cells.
  • Control T cells, in some embodiments, are engineered T cells (e.g., gene edited T cells). In some embodiments, control T cells are engineered T cells that comprise a disrupted TRAC gene, a nucleic acid encoding a CAR (e.g., an anti-LIV1 CAR) inserted into the TRAC gene, and/or a disrupted β2M gene. In some embodiments, control T cells are unedited T cells.
  • Gene Editing Methods
  • Gene editing (including genomic editing) is a type of genetic engineering in which nucleotide(s)/nucleic acid(s) is/are inserted, deleted, and/or substituted in a DNA sequence, such as in the genome of a targeted cell. Targeted gene editing enables insertion, deletion, and/or substitution at pre-selected sites in the genome of a targeted cell (e.g., in a targeted gene or targeted DNA sequence). When an sequence of an endogenous gene is edited, for example by deletion, insertion or substitution of nucleotide(s)/nucleic acid(s), the endogenous gene comprising the affected sequence may be knocked-out or knocked-down due to the sequence alteration. Therefore, targeted editing may be used to disrupt endogenous gene expression. “Targeted integration” refers to a process involving insertion of one or more exogenous sequences, with or without deletion of an endogenous sequence at the insertion site. Targeted integration can result from targeted gene editing when a donor template containing an exogenous sequence is present.
  • Targeted editing can be achieved either through a nuclease-independent approach, or through a nuclease-dependent approach. In the nuclease-independent targeted editing approach, homologous recombination is guided by homologous sequences flanking an exogenous polynucleotide to be introduced into an endogenous sequence through the enzymatic machinery of the host cell. The exogenous polynucleotide may introduce deletions, insertions or replacement of nucleotides in the endogenous sequence.
  • Alternatively, the nuclease-dependent approach can achieve targeted editing with higher frequency through the specific introduction of double strand breaks (DSBs) by specific rare-cutting nucleases (e.g., endonucleases). Such nuclease-dependent targeted editing also utilizes DNA repair mechanisms, for example, non-homologous end joining (NHEJ), which occurs in response to DSBs. DNA repair by NHEJ often leads to random insertions or deletions (indels) of a small number of endogenous nucleotides. In contrast to NHEJ mediated repair, repair can also occur by a homology directed repair (HDR). When a donor template containing exogenous genetic material flanked by a pair of homology arms is present, the exogenous genetic material can be introduced into the genome by HDR, which results in targeted integration of the exogenous genetic material.
  • Available endonucleases capable of introducing specific and targeted DSBs include, but not limited to, zinc-finger nucleases (ZFN), transcription activator-like effector nucleases (TALEN), and RNA-guided CRISPR-Cas9 nuclease (CRISPR/Cas9; Clustered Regular Interspaced Short Palindromic Repeats Associated 9). Additionally, DICE (dual integrase cassette exchange) system utilizing phiC31 and Bxb1 integrases may also be used for targeted integration.
  • ZFNs are targeted nucleases comprising a nuclease fused to a zinc finger DNA binding domain (ZFBD), which is a polypeptide domain that binds DNA in a sequence-specific manner through one or more zinc fingers. A zinc finger is a domain of about 30 amino acids within the zinc finger binding domain whose structure is stabilized through coordination of a zinc ion. Examples of zinc fingers include, but not limited to, C2H2 zinc fingers, C3H zinc fingers, and C4 zinc fingers. A designed zinc finger domain is a domain not occurring in nature whose design/composition results principally from rational criteria, e.g., application of substitution rules and computerized algorithms for processing information in a database storing information of existing ZFP designs and binding data. See, for example, U.S. Pat. Nos. 6,140,081; 6,453,242; and 6,534,261; see also WO 98/53058; WO 98/53059; WO 98/53060; WO 02/016536 and WO 03/016496. A selected zinc finger domain is a domain not found in nature whose production results primarily from an empirical process such as phage display, interaction trap or hybrid selection. ZFNs are described in greater detail in U.S. Pat. Nos. 7,888,121 and 7,972,854. The most recognized example of a ZFN is a fusion of the FokI nuclease with a zinc finger DNA binding domain.
  • A TALEN is a targeted nuclease comprising a nuclease fused to a TAL effector DNA binding domain. A “transcription activator-like effector DNA binding domain”, “TAL effector DNA binding domain”, or “TALE DNA binding domain” is a polypeptide domain of TAL effector proteins that is responsible for binding of the TAL effector protein to DNA. TAL effector proteins are secreted by plant pathogens of the genus Xanthomonas during infection. These proteins enter the nucleus of the plant cell, bind effector-specific DNA sequences via their DNA binding domain, and activate gene transcription at these sequences via their transactivation domains. TAL effector DNA binding domain specificity depends on an effector-variable number of imperfect 34 amino acid repeats, which comprise polymorphisms at select repeat positions called repeat variable-diresidues (RVD). TALENs are described in greater detail in US Patent Application No. 2011/0145940. The most recognized example of a TALEN in the art is a fusion polypeptide of the FokI nuclease to a TAL effector DNA binding domain.
  • Additional examples of targeted nucleases suitable for use as provided herein include, but are not limited to, Bxb1, phiC31, R4, PhiBT1, and Wβ/SPBc/TP901-1, whether used individually or in combination.
  • Other non-limiting examples of targeted nucleases include naturally-occurring and recombinant nucleases, e.g., CRISPR/Cas9, restriction endonucleases, meganucleases homing endonucleases, and the like.
  • CRISPR-Cas9 Gene Editing
  • The CRISPR-Cas9 system is a naturally-occurring defense mechanism in prokaryotes that has been repurposed as a RNA-guided DNA-targeting platform used for gene editing. It relies on the DNA nuclease Cas9, and two noncoding RNAs-crisprRNA (crRNA) and trans-activating RNA (tracrRNA)—to target the cleavage of DNA.
  • crRNA drives sequence recognition and specificity of the CRISPR-Cas9 complex through Watson-Crick base pairing typically with a 20 nucleotide (nt) sequence in the target DNA. Changing the sequence of the 5′ 20 nt in the crRNA allows targeting of the CRISPR-Cas9 complex to specific loci. The CRISPR-Cas9 complex only binds DNA sequences that contain a sequence match to the first 20 nt of the crRNA, single-guide RNA (sgRNA), if the target sequence is followed by a specific short DNA motif (with the sequence NGG) referred to as a protospacer adjacent motif (PAM).
  • TracrRNA hybridizes with the 3′ end of crRNA to form an RNA-duplex structure that is bound by the Cas9 endonuclease to form the catalytically active CRISPR-Cas9 complex, which can then cleave the target DNA.
  • Once the CRISPR-Cas9 complex is bound to DNA at a target site, two independent nuclease domains within the Cas9 enzyme each cleave one of the DNA strands upstream of the PAM site, leaving a double-strand break (DSB) where both strands of the DNA terminate in a base pair (a blunt end).
  • After binding of CRISPR-Cas9 complex to DNA at a specific target site and formation of the site-specific DSB, the next key step is repair of the DSB. Cells use two main DNA repair pathways to repair the DSB: non-homologous end-joining (NHEJ) and homology-directed repair (HDR).
  • NHEJ is a robust repair mechanism that appears highly active in the majority of cell types, including non-dividing cells. NHEJ is error-prone and can often result in the removal or addition of between one and several hundred nucleotides at the site of the DSB, though such modifications are typically <20 nt. The resulting insertions and deletions (indels) can disrupt coding or noncoding regions of genes. Alternatively, HDR uses a long stretch of homologous donor DNA, provided endogenously or exogenously, to repair the DSB with high fidelity. HDR is active only in dividing cells, and occurs at a relatively low frequency in most cell types. In many embodiments of the present disclosure, NHEJ is utilized as the repair operant.
  • In some embodiments, the Cas9 (CRISPR associated protein 9) endonuclease is from Streptococcus pyogenes, although other Cas9 homologs may be used. It should be understood, that wild-type Cas9 may be used or modified versions of Cas9 may be used (e.g., evolved versions of Cas9, or Cas9 orthologues or variants), as provided herein. In some embodiments, Cas9 may be substituted with another RNA-guided endonuclease, such as Cpf1 (of a class II CRISPR/Cas system).
  • Guide RNAs
  • The present disclosure provides a genome-targeting nucleic acid that can direct the activities of an associated polypeptide (e.g., a site-directed polypeptide) to a specific target sequence within a target nucleic acid. The genome-targeting nucleic acid can be an RNA. A genome-targeting RNA is referred to as a “guide RNA” or “gRNA” herein. A guide RNA comprises at least a spacer sequence that hybridizes to a target nucleic acid sequence of interest, and a CRISPR repeat sequence. In Type II systems, the gRNA also comprises a second RNA called the tracrRNA sequence. In the Type II guide RNA (gRNA), the CRISPR repeat sequence and tracrRNA sequence hybridize to each other to form a duplex. In the Type V guide RNA (gRNA), the crRNA forms a duplex. In both systems, the duplex binds a site-directed polypeptide, such that the guide RNA and site-direct polypeptide form a complex. In some embodiments, the genome-targeting nucleic acid provides target specificity to the complex by virtue of its association with the site-directed polypeptide. The genome-targeting nucleic acid thus directs the activity of the site-directed polypeptide.
  • As is understood by the person of ordinary skill in the art, each guide RNA is designed to include a spacer sequence complementary to its genomic target sequence. See Jinek et al., Science, 337, 816-821 (2012) and Deltcheva et al., Nature, 471, 602-607 (2011).
  • In some embodiments, the genome-targeting nucleic acid is a double-molecule guide RNA. In some embodiments, the genome-targeting nucleic acid is a single-molecule guide RNA.
  • A double-molecule guide RNA comprises two strands of RNA. The first strand comprises in the 5′ to 3′ direction, an optional spacer extension sequence, a spacer sequence and a minimum CRISPR repeat sequence. The second strand comprises a minimum tracrRNA sequence (complementary to the minimum CRISPR repeat sequence), a 3′ tracrRNA sequence and an optional tracrRNA extension sequence.
  • A single-molecule guide RNA (sgRNA) in a Type II system comprises, in the 5′ to 3′ direction, an optional spacer extension sequence, a spacer sequence, a minimum CRISPR repeat sequence, a single-molecule guide linker, a minimum tracrRNA sequence, a 3′ tracrRNA sequence and an optional tracrRNA extension sequence. The optional tracrRNA extension may comprise elements that contribute additional functionality (e.g., stability) to the guide RNA. The single-molecule guide linker links the minimum CRISPR repeat and the minimum tracrRNA sequence to form a hairpin structure. The optional tracrRNA extension comprises one or more hairpins.
  • A single-molecule guide RNA (referred to as a “sgRNA” or “gRNA”) in a Type V system comprises, in the 5′ to 3′ direction, a minimum CRISPR repeat sequence and a spacer sequence.
  • The sgRNA can comprise a 20 nucleotide spacer sequence at the 5′ end of the sgRNA sequence. The sgRNA can comprise a less than 20 nucleotide spacer sequence at the 5′ end of the sgRNA sequence. The sgRNA can comprise a more than 20 nucleotide spacer sequence at the 5′ end of the sgRNA sequence. The sgRNA can comprise a variable length spacer sequence with 17-30 nucleotides at the 5′ end of the sgRNA sequence (see Table 3).
  • The sgRNA can comprise no uracil at the 3′ end of the sgRNA sequence. The sgRNA can comprise one or more uracil at the 3′ end of the sgRNA sequence. For example, the sgRNA can comprise 1 uracil (U) at the 3′ end of the sgRNA sequence. The sgRNA can comprise 2 uracil (UU) at the 3′ end of the sgRNA sequence. The sgRNA can comprise 3 uracil (UUU) at the 3′ end of the sgRNA sequence. The sgRNA can comprise 4 uracil (UUUU) at the 3′ end of the sgRNA sequence. The sgRNA can comprise 5 uracil (UUUUU) at the 3′ end of the sgRNA sequence. The sgRNA can comprise 6 uracil (UUUUUU) at the 3′ end of the sgRNA sequence. The sgRNA can comprise 7 uracil (U) at the 3′ end of the sgRNA sequence. The sgRNA can comprise 8 uracil (UU at the 3′ end of the sgRNA sequence.
  • The sgRNA can be unmodified or modified. For example, modified sgRNAs can comprise one or more 2′-O-methyl phosphorothioate nucleotides.
  • TABLE 3
    SEQ
    ID
    NO. sgRNA sequence
    15 nnnnnnnnnnnnnnnnnnnnguuuuagagcuagaaauag
    caaguuaaaauaaggcuaguccguuaucaacuugaaaaa
    guggcaccgagucggugcuuuu
    16 nnnnnnnnnnnnnnnnnnnnguuuuagagcuagaaauag
    caaguuaaaauaaggcuaguccguuaucaacuugaaaaa
    guggcaccgagucggugc
    17 n(17-30)guuuuagagcuagaaauagcaaguuaaaaua
    aggcuaguccguuaucaacuugaaaaaguggcaccgagu
    cggugcu(1-8)
  • By way of illustration, guide RNAs used in the CRISPR/Cas/Cpf1 system, or other smaller RNAs can be readily synthesized by chemical means, as illustrated below and described in the art. While chemical synthetic procedures are continually expanding, purifications of such RNAs by procedures such as high performance liquid chromatography (HPLC, which avoids the use of gels such as PAGE) tends to become more challenging as polynucleotide lengths increase significantly beyond a hundred or so nucleotides. One approach used for generating RNAs of greater length is to produce two or more molecules that are ligated together. Much longer RNAs, such as those encoding a Cas9 or Cpf1 endonuclease, are more readily generated enzymatically. Various types of RNA modifications can be introduced during or after chemical synthesis and/or enzymatic generation of RNAs, e.g., modifications that enhance stability, reduce the likelihood or degree of innate immune response, and/or enhance other attributes, as described in the art.
  • Spacer Sequence
  • A gRNA comprises a spacer sequence. A spacer sequence is a sequence (e.g., a 20 nucleotide sequence) that defines the target sequence (e.g., a DNA target sequences, such as a genomic target sequence) of a target nucleic acid of interest. In some embodiments, the spacer sequence is 15 to 30 nucleotides. In some embodiments, the spacer sequence is 15, 16, 17, 18, 19, 29, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides. In some embodiments, a spacer sequence is 20 nucleotides.
  • The “target sequence” is adjacent to a PAM sequence and is the sequence modified by an RNA-guided nuclease (e.g., Cas9). The “target nucleic acid” is a double-stranded molecule: one strand comprises the target sequence and is referred to as the “PAM strand,” and the other complementary strand is referred to as the “non-PAM strand.” One of skill in the art recognizes that the gRNA spacer sequence hybridizes to the reverse complement of the target sequence, which is located in the non-PAM strand of the target nucleic acid of interest. Thus, the gRNA spacer sequence is the RNA equivalent of the target sequence. For example, if the target sequence is 5′-AGAGCAACAGTGCTGTGGCC-3′ (SEQ ID NO: 92), then the gRNA spacer sequence is 5′-AGAGCAACAGUGCUGUGGCC-3′ (SEQ ID NO: 93). The spacer of a gRNA interacts with a target nucleic acid of interest in a sequence-specific manner via hybridization (i.e., base pairing). The nucleotide sequence of the spacer thus varies depending on the target sequence of the target nucleic acid of interest.
  • In a CRISPR/Cas system herein, the spacer sequence is designed to hybridize to a region of the target nucleic acid that is located 5′ of a PAM of the Cas9 enzyme used in the system. The spacer may perfectly match the target sequence or may have mismatches. Each Cas9 enzyme has a particular PAM sequence that it recognizes in a target DNA. For example, S. pyogenes recognizes in a target nucleic acid a PAM that comprises the sequence 5′-NRG-3′, where R comprises either A or G, where N is any nucleotide and N is immediately 3′ of the target nucleic acid sequence targeted by the spacer sequence.
  • In some embodiments, the target nucleic acid sequence comprises 20 nucleotides. In some embodiments, the target nucleic acid comprises less than 20 nucleotides. In some embodiments, the target nucleic acid comprises more than 20 nucleotides. In some embodiments, the target nucleic acid comprises at least: 5, 10, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides. In some embodiments, the target nucleic acid comprises at most: 5, 10, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides. In some embodiments, the target nucleic acid sequence comprises 20 bases immediately 5′ of the first nucleotide of the PAM. For example, in a sequence comprising 5′-NNNNNNNNNNNNNNNNNNNNNRG-3′ (SEQ ID NO: 130), the target nucleic acid comprises the sequence that corresponds to the Ns, wherein N is any nucleotide, and the underlined NRG sequence is the S. pyogenes PAM.
  • Non-limiting examples of gRNAs that may be used as provided herein are provided in Table 4 and PCT/US2018/032334, filed May 11, 2018.
  • TABLE 4
    gRNA Sequences/Target Sequences
    gRNA Sequences
    Unmodified Modified
    Name Sequence Sequence
    TRAC  AGAGCAACAGUG A*G*A*GCAACA
    sgRNA CUGUGGCCguuu GUGCUGUGGCCg
    uagagcuagaaa uuuuagagcuag
    uagcaaguuaaa aaauagcaaguu
    auaaggcuaguc aaaauaaggcua
    cguuaucaacuu guccguuaucaa
    gaaaaaguggca cuugaaaaagug
    ccgagucggugc gcaccgagucgg
    UUUU ugcU*U*U*U
    (SEQ ID NO: 18) (SEQ ID NO: 28)
    TRAC   AGAGCAACAGUG A*G*A*GCAACA
    sgRNA CUGUGGCC GUGCUGUGGCC
    spacer (SEQ ID NO: 19) (SEQ ID NO: 29)
    β2M  GCUACUCUCUCU G*C*U*ACUCUC
    sgRNA UUCUGGCCguuu UCUUUCUGGCCg
    uagagcuagaaa uuuuagagcuag
    uagcaaguuaaa aaauagcaaguu
    auaaggcuaguc aaaauaaggcua
    cguuaucaacuu guccguuaucaa
    gaaaaaguggca cuugaaaaagug
    ccgagucggugc gcaccgagucgg
    UUUU ugcU*U*U*U
    (SEQ ID NO: 20) (SEQ ID NO: 30)
    β2M   GCUACUCUCUCU G*C*U*ACUCUC
    sgRNA UUCUGGCC UCUUUCUGGCC
    spacer (SEQ ID NO: 21) (SEQ ID NO: 31)
    PD-1  CUGCAGCUUCUC C*U*G*CAGCUU
    sgRNA CAACACAUguuu CUCCAACACAUg
    uagagcuagaaa uuuuagagcuag
    uagcaaguuaaa aaauagcaaguu
    auaaggcuaguc aaaauaaggcua
    cguuaucaacuu guccguuaucaa
    gaaaaaguggca cuugaaaaagug
    ccgagucggugc gcaccgagucgg
    UUUU ugcU*U*U*U
    (SEQ ID NO: 22) (SEQ ID NO: 32)
    PD-1   CUGCAGCUUCUC C*U*G*CAGCUU
    sgRNA CAACACAU CUCCAACACAU
    spacer (SEQ ID NO: 23) (SEQ ID NO: 33)
    CD70   GCUUUGGUCCCA G*C*U*UUGGUC
    sgRNA UUGGUCGCguuu CCAUUGGUCGCg
    (E1_T7) uagagcuagaaa uuuuagagcuag
    uagcaaguuaaa aaauagcaaguu
    auaaggcuaguc aaaauaaggcua
    cguuaucaacuu guccguuaucaa
    gaaaaaguggca cuugaaaaagug
    ccgagucggugc gcaccgagucgg
    UUUU ugcU*U*U*U
    (SEQ ID NO: 24) (SEQ ID NO: 34), 
    T7
    CD70   GCUUUGGUCCCA G*C*U*UUGGUC
     sgRNA UUGGUCGC CCAUUGGUCGC
    (E1_T7) (SEQ ID NO: 25) (SEQ ID NO: 35)
    spacer
    CD70   GCCCGCAGGACG G*C*C*CGCAGG
    sgRNA CACCCAUA ACGCACCCAUAg
    (E1_T8) guuuuagagcua uuuuagagcuag
    gaaauagcaagu aaauagcaaguu
    uaaaauaaggcu aaaauaaggcua
    aguccguuauca guccguuaucaa
    acuugaaaaagu cuugaaaaagug
    ggcaccgagucg gcaccgagucgg
    gugcUUUU ugcU*U*U*U
    (SEQ ID NO: 26) (SEQ ID NO: 36), 
    T8
    CD70   GCCCGCAGGACG G*C*C*CGCAGG
    sgRNA  CACCCAUA ACGCACCCAUA
    (E1_T8) (SEQ ID NO: 27) (SEQ ID NO: 37)
    spacer
    Target Sequences
    Guide
    Name Target Sequence (PAM)
    CD70   GCTTTGGTCCCATTGGTCGC (GGG)
    sgRNA (SEQ ID NO: 38)
    (E1_T7)
    CD70   GCCCGCAGGACGCACCCATA (GGG)
    sgRNA (SEQ ID NO: 39)
    (E1_T8)
    TRAC  AGAGCAACAGTGCTGTGGCC (TGG)
    sgRNA (SEQ ID NO: 40)
    β2M  GCTACTCTCTCTTTCTGGCC (TGG)
    sgRNA (SEQ ID NO: 41)
    PD-1  CTGCAGCTTCTCCAACACAT (CGG)
    sgRNA (SEQ ID NO: 42)
    *: 2′-O-methyl phosphorothioate residue
  • Chimeric Antigen Receptor (CAR) T Cells
  • A chimeric antigen receptor refers to an artificial immune cell receptor that is engineered to recognize and bind to an antigen expressed by tumor cells. Generally, a CAR is designed for a T cell and is a chimera of a signaling domain of the T-cell receptor (TCR) complex and an antigen-recognizing domain (e.g., a single chain fragment (scFv) of an antibody or other antibody fragment) (Enblad et al., Human Gene Therapy. 2015; 26(8):498-505). A T cell that expresses a CAR is referred to as a CAR T cell. CARs have the ability to redirect T-cell specificity and reactivity toward a selected target in a non-MHC-restricted manner. The non-MHC-restricted antigen recognition gives T-cells expressing CARs the ability to recognize an antigen independent of antigen processing, thus bypassing a major mechanism of tumor escape. Moreover, when expressed in T-cells, CARs advantageously do not dimerize with endogenous T-cell receptor (TCR) alpha and beta chains.
  • There are four generations of CARs, each of which contains different components. First generation CARs join an antibody-derived scFv to the CD3zeta (ζ or z) intracellular signaling domain of the T-cell receptor through hinge and transmembrane domains. Second generation CARs incorporate an additional domain, e.g., CD28, 4-1BB (41BB), or ICOS, to supply a costimulatory signal. Third-generation CARs contain two costimulatory domains fused with the TCR CD3ζ chain. Third-generation costimulatory domains may include, e.g., a combination of CD3ζ, CD27, CD28, 4-1BB, ICOS, or OX40. CARs, in some embodiments, contain an ectodomain (e.g., CD3), commonly derived from a single chain variable fragment (scFv), a hinge, a transmembrane domain, and an endodomain with one (first generation), two (second generation), or three (third generation) signaling domains derived from CD3Z and/or co-stimulatory molecules (Maude et al., Blood. 2015; 125(26):4017-4023; Kakarla and Gottschalk, Cancer J. 2014; 20(2):151-155).
  • CARs typically differ in their functional properties. The CD3ζ signaling domain of the T-cell receptor, when engaged, will activate and induce proliferation of T-cells but can lead to anergy (a lack of reaction by the body's defense mechanisms, resulting in direct induction of peripheral lymphocyte tolerance). Lymphocytes are considered anergic when they fail to respond to a specific antigen. The addition of a costimulatory domain in second-generation CARs improved replicative capacity and persistence of modified T-cells. Similar antitumor effects are observed in vitro with CD28 or 4-1BB CARs, but preclinical in vivo studies suggest that 4-1BB CARs may produce superior proliferation and/or persistence. Clinical trials suggest that both of these second-generation CARs are capable of inducing substantial T-cell proliferation in vivo, but CARs containing the 4-1BB costimulatory domain appear to persist longer. Third generation CARs combine multiple signaling domains (costimulatory) to augment potency.
  • In some embodiments, a chimeric antigen receptor is a first generation CAR. In other embodiments, a chimeric antigen receptor is a second generation CAR. In yet other embodiments, a chimeric antigen receptor is a third generation CAR.
  • A CAR, in some embodiments, comprises an extracellular (ecto) domain comprising an antigen binding domain (e.g., an antibody, such as an scFv), a transmembrane domain, and a cytoplasmic (endo) domain.
  • Ectodomain. The ectodomain is the region of the CAR that is exposed to the extracellular fluid and, in some embodiments, includes an antigen binding domain, and optionally a signal peptide, a spacer domain, and/or a hinge domain. In some embodiments, the antigen binding domain is a single-chain variable fragment (scFv) that include the light and heavy chains of immunoglobulins connected with a short linker peptide. The linker, in some embodiments, includes hydrophilic residues with stretches of glycine and serine for flexibility as well as stretches of glutamate and lysine for added solubility. A single-chain variable fragment (scFv) is not actually a fragment of an antibody, but instead is a fusion protein of the variable regions of the heavy (VH) and light chains (VL) of immunoglobulins, connected with a short linker peptide of ten to about 25 amino acids. The linker is usually rich in glycine for flexibility, as well as serine or threonine for solubility, and can either connect the N-terminus of the VH with the C-terminus of the VL, or vice versa. This protein retains the specificity of the original immunoglobulin, despite removal of the constant regions and the introduction of the linker. Non-limiting examples of VH and VL protein sequences that may be used to create an anti-LIV1 scFv may include the amino acid sequence of SEQ ID NOs: 55, 90 or 98 (VH) and SEQ ID NOs: 56, 88 or 128 (VL). In some embodiments, the scFv of the present disclosure is humanized. In other embodiments, the scFv is fully human. In yet other embodiments, the scFv is a chimera (e.g., of mouse and human sequence). In some embodiments, the scFv is an anti-LIV1 scFv (binds specifically to LIV1). Non-limiting examples of anti-LIV1 scFv proteins that may be used as provided herein may include the amino acid sequence of any one of SEQ ID NOs: 54, 70, 82, 83, 84, 85, 86, or 87. Other scFv proteins may be used.
  • The signal peptide can enhance the antigen specificity of CAR binding. Signal peptides can be derived from antibodies, such as, but not limited to, CD8, as well as epitope tags such as, but not limited to, GST or FLAG. Examples of signal peptides include MLLLVTSLLLCELPHPAFLLIP (SEQ ID NO: 94) and MALPVTALLLPLALLLHAARP (SEQ ID NO: 73). Other signal peptides may be used.
  • In some embodiments, a spacer domain or hinge domain is located between an extracellular domain (comprising the antigen binding domain) and a transmembrane domain of a CAR, or between a cytoplasmic domain and a transmembrane domain of the CAR. A spacer domain is any oligopeptide or polypeptide that functions to link the transmembrane domain to the extracellular domain and/or the cytoplasmic domain in the polypeptide chain. A hinge domain is any oligopeptide or polypeptide that functions to provide flexibility to the CAR, or domains thereof, or to prevent steric hindrance of the CAR, or domains thereof. In some embodiments, a spacer domain or a hinge domain may comprise up to 300 amino acids (e.g., 10 to 100 amino acids, or 5 to 20 amino acids). In some embodiments, one or more spacer domain(s) may be included in other regions of a CAR. In some embodiments, the hinge domain is a CD8 hinge domain. Other hinge domains may be used.
  • Transmembrane Domain. The transmembrane domain is a hydrophobic alpha helix that spans the membrane. The transmembrane domain provides stability of the CAR. In some embodiments, the transmembrane domain of a CAR as provided herein is a CD8 transmembrane domain. In other embodiments, the transmembrane domain is a CD28 transmembrane domain. In yet other embodiments, the transmembrane domain is a chimera of a CD8 and CD28 transmembrane domain. Other transmembrane domains may be used as provided herein. In some embodiments, the transmembrane domain is a CD8a transmembrane domain: FVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGG AVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCNHRNR (SEQ ID NO: 129). Other transmembrane domains may be used.
  • Endodomain. The endodomain is the functional end of the receptor. Following antigen recognition, receptors cluster and a signal is transmitted to the cell. The most commonly used endodomain component is CD3-zeta, which contains three (3) immunoreceptor tyrosine-based activation motif (ITAM)s. This transmits an activation signal to the T cell after the antigen is bound. In many cases, CD3-zeta may not provide a fully competent activation signal and, thus, a co-stimulatory signaling is used. For example, CD28 and/or 4-1BB may be used with CD3-zeta (CD3) to transmit a proliferative/survival signal. Thus, in some embodiments, the co-stimulatory molecule of a CAR as provided herein is a CD28 co-stimulatory molecule. In other embodiments, the co-stimulatory molecule is a 4-1BB co-stimulatory molecule. In some embodiments, a CAR includes CD3ζ and CD28. In other embodiments, a CAR includes CD3-zeta and 4-1BB. In still other embodiments, a CAR includes CD3, CD28, and 4-1BB. Table 5 provides examples of signaling molecules that may be used as provided herein.
  • TABLE 5
    SEQ
    ID
    Name Sequence NO:
    4-1BB AAACGGGGCAGAAAGAAACTCCTGTATATATT 43
    CAAACAACCATTTATGAGACCAGTACAAACTA
    CTCAAGAGGAAGATGGCTGTAGCTGCCGATTT
    CCAGAAGAAGAAGAAGGAGGATGTGAACTG
    KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRF 44
    PEEEEGGCEL
    CD28 TCAAAGCGGAGTAGGTTGTTGCATTCCGATTA 45
    CATGAATATGACTCCTCGCCGGCCTGGGCCGA
    CAAGAAAACATTACCAACCCTATGCCCCCCCA
    CGAGACTTCGCTGCGTACAGGTCC
    SKRSRLLHSDYMNMTPRRPGPTRKHYQPYAPP 46
    RDFAAYRS
    CD3-zeta CGAGTGAAGTTTTCCCGAAGCGCAGACGCTCC 47
    GGCATATCAGCAAGGACAGAATCAGCTGTATA
    ACGAACTGAATTTGGGACGCCGCGAGGAGTAT
    GACGTGCTTGATAAACGCCGGGGGAGAGACCC
    GGAAATGGGGGGTAAACCCCGAAGAAAGAATC
    CCCAAGAAGGACTCTACAATGAACTCCAGAAG
    GATAAGATGGCGGAGGCCTACTCAGAAATAGG
    TATGAAGGGCGAACGACGACGGGGAAAAGGTC
    ACGATGGCCTCTACCAAGGGTTGAGTACGGCA
    ACCAAAGATACGTACGATGCACTGCATATGCA
    GGCCCTGCCTCCCAGA
    RVKFSRSADAPAYQQGQNQLYNELNLGRREEY 48
    DVLDKRRGRDPEMGGKPRRKNPQEGLYNELQK
    DKMAEAYSEIGMKGERRRGKGHDGLYQGLSTA
    TKDTYDALHMQALPPR
  • Antibodies
  • An antibody (interchangeably used in plural form) is an immunoglobulin molecule capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, polypeptide, etc., through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule. As used herein, the term “antibody” encompasses not only intact (i.e., full-length) monoclonal antibodies, but also antigen-binding fragments (such as Fab, Fab′, F(ab′)2, Fv), single chain variable fragment (scFv), mutants thereof, fusion proteins comprising an antibody portion, humanized antibodies, chimeric antibodies, diabodies, linear antibodies, single chain antibodies, single domain antibodies (e.g., camel or llama VHH antibodies), multispecific antibodies (e.g., bispecific antibodies) and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site of the required specificity, including glycosylation variants of antibodies, amino acid sequence variants of antibodies, and covalently modified antibodies.
  • A typical antibody molecule comprises a heavy chain variable region (VH) and a light chain variable region (VL), which are usually involved in antigen binding. These regions/residues that are responsible for antigen-binding can be identified from amino acid sequences of the VH/VL sequences of a reference antibody (e.g., an anti-LIV1 antibody as described herein) by methods known in the art. The VH and VL regions can be further subdivided into regions of hypervariability, also known as “complementarity determining regions” (“CDR”), interspersed with regions that are more conserved, which are known as “framework regions” (“FR”). Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The extent of the framework region and CDRs can be precisely identified using methodology known in the art, for example, by the Kabat definition, the Chothia definition, the AbM definition, and/or the contact definition, all of which are well known in the art. As used herein, a CDR may refer to the CDR defined by any method known in the art. Two antibodies having the same CDR means that the two antibodies have the same amino acid sequence of that CDR as determined by the same method. See, e.g., Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242, Chothia et al., (1989) Nature 342:877; Chothia, C. et al. (1987) J. Mol. Biol. 196:901-917, Al-lazikani et al (1997) J. Molec. Biol. 273:927-948; and Almagro, J. Mol. Recognit. 17:132-143 (2004). See also hgmp.mrc.ac.uk and bioinf.org.uk/abs.
  • In some embodiments, an antibody is an scFv, such as an anti-LIV1 scFv. An antibody includes an antibody of any class, such as IgD, IgE, IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class. Depending on the antibody amino acid sequence of the constant domain of its heavy chains, immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2. The heavy-chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • The antibodies to be used as provided herein can be murine, rat, human, or any other origin (including chimeric or humanized antibodies). In some examples, the antibody comprises a modified constant region, such as a constant region that is immunologically inert, e.g., does not trigger complement mediated lysis, or does not stimulate antibody-dependent cell mediated cytotoxicity (ADCC).
  • In some embodiments, an antibody of the present disclosure is a humanized antibody. Humanized antibodies refer to forms of non-human (e.g., murine) antibodies that are specific chimeric immunoglobulins, immunoglobulin chains, or antigen-binding fragments thereof that contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat, or rabbit having the desired specificity, affinity, and capacity. In some instances, Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, the humanized antibody may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences, but are included to further refine and optimize antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. A humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin. Other forms of humanized antibodies have one or more CDRs (one, two, three, four, five, six) which are altered with respect to the original antibody, which are also termed one or more CDRs “derived from” one or more CDRs from the original antibody. Humanized antibodies may also involve affinity maturation.
  • In some embodiments, an antibody of the present disclosure is a chimeric antibody, which can include a heavy constant region and a light constant region from a human antibody. Chimeric antibodies refer to antibodies having a variable region or part of variable region from a first species and a constant region from a second species. Typically, in these chimeric antibodies, the variable region of both light and heavy chains mimics the variable regions of antibodies derived from one species of mammals (e.g., a non-human mammal such as mouse, rabbit, and rat), while the constant portions are homologous to the sequences in antibodies derived from another mammal such as human. In some embodiments, amino acid modifications can be made in the variable region and/or the constant region.
  • In some embodiments, an antibody of the present disclosure specifically binds a target antigen, such as human LIV1. An antibody that “specifically binds” (used interchangeably herein) to a target or an epitope is a term well understood in the art, and methods to determine such specific binding are also well known in the art. A molecule is said to exhibit “specific binding” if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular target antigen than it does with alternative targets. An antibody “specifically binds” to a target antigen if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other substances. For example, an antibody that specifically (or preferentially) binds to a LIV1 epitope is an antibody that binds this LIV1 epitope with greater affinity, avidity, more readily, and/or with greater duration than it binds to other LIV1 epitopes or non-LIV1 epitopes. It is also understood by reading this definition that, for example, an antibody that specifically binds to a first target antigen may or may not specifically or preferentially bind to a second target antigen. As such, “specific binding” or “preferential binding” does not necessarily require (although it can include) exclusive binding. Generally, but not necessarily, reference to binding means preferential binding.
  • In some embodiments, the equilibrium dissociation constant (KD) between the antibody and LIV1 is 100 pM to 1 μM. In some embodiments, the KD between the antibody and LIV1 is 1 nM to 100 nM.
  • Also within the scope of the present disclosure are functional variants of any of the exemplary antibodies as disclosed herein. A functional variant may contain one or more amino acid residue variations in the VH and/or VL, or in one or more of the VH CDRs and/or one or more of the VL CDRs as relative to a reference antibody, while retaining substantially similar binding and biological activities (e.g., substantially similar binding affinity, binding specificity, inhibitory activity, anti-tumor activity, or a combination thereof) as the reference antibody.
  • In some examples, an antibody disclosed herein comprises a VH CDR1, a VH CDR2, and a VH CDR3, which collectively contains no more than 10 amino acid variations (e.g., no more than 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid variation) as compared with the VH CDR1, VH CDR2, and VH CDR3 of a reference antibody such as in VH: SEQ ID NO: 55 or 90 or 98; VL: SEQ ID NO: 56 or 88 or 128. “Collectively” means that the total number of amino acid variations in all of the three VH CDRs is within the defined range. Alternatively or in addition, antibody may comprise a VL CDR1, a VL CDR2, and a VL CDR3, which collectively contains no more than 10 amino acid variations (e.g., no more than 9, 8, 7, 6, 5, 4, 3, 2 or 1 amino acid variation) as compared with the VL CDR1, VL CDR2, and VL CDR3 of the reference antibody.
  • In some examples, an antibody disclosed herein may comprise a VH CDR1, a VH CDR2, and a VH CDR3, at least one of which contains no more than 5 amino acid variations (e.g., no more than 4, 3, 2, or 1 amino acid variation) as the counterpart VH CDR of a reference antibody such as in VH: SEQ ID NO: 55 or 90 or 98; VL: SEQ ID NO: 56 or 88 or 128. In specific examples, the antibody comprises a VH CDR3, which contains no more than 5 amino acid variations (e.g., no more than 4, 3, 2, or 1 amino acid variation) as the VH CDR3 of a reference antibody such as in VH: SEQ ID NO: 55 or 90 or 98; VL: SEQ ID NO: 56 or 88 or 128. Alternatively or in addition, an antibody may comprise a VL CDR1, a VL CDR2, and a VL CDR3, at least one of which contains no more than 5 amino acid variations (e.g., no more than 4, 3, 2, or 1 amino acid variation) as the counterpart VL CDR of the reference antibody. In specific examples, the antibody comprises a VL CDR3, which contains no more than 5 amino acid variations (e.g., no more than 4, 3, 2, or 1 amino acid variation) as the LC CDR3 of the reference antibody.
  • In some instances, the amino acid residue variations can be conservative amino acid residue substitutions. As used herein, a “conservative amino acid substitution” refers to an amino acid substitution that does not alter the relative charge or size characteristics of the protein in which the amino acid substitution is made. Variants can be prepared according to methods for altering polypeptide sequence known to one of ordinary skill in the art such as are found in references which compile such methods, e.g. Molecular Cloning: A Laboratory Manual, J. Sambrook, et al., eds., Second Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989, or Current Protocols in Molecular Biology, F. M. Ausubel, et al., eds., John Wiley & Sons, Inc., New York. Conservative substitutions of amino acids include substitutions made amongst amino acids within the following groups: (a) A→G, S; (b) R→K, H; (c) N→Q, H; (d) D→E, N; (e) C→S, A; (f) Q→N; (g) E→D, Q; (h) G→A; (i) H→N, Q; (j) I→L, V; (k) L→I, V; (l) K→R, H; (m) M→L, I, Y; (n) F→Y, M, L; (o) P→A; (p) S→T; (q) T→S; (r) W→Y, F; (s) Y→W, F; and (t) V→I, L.
  • In some embodiments, an antibody disclosed herein may comprise VH CDRs that collectively are at least 80% (e.g., 85%, 90%, 95%, or 98%) identical to the VH CDRs of a reference antibody such as Antibody A (VH: SEQ ID NO: 55; VL: SEQ ID NO: 56) or Antibody B (VH: SEQ ID NO: 90; VL: SEQ ID NO: 88). Alternatively or in addition, the antibody may comprise VL CDRs that collectively are at least 80% (e.g., 85%, 90%, 95%, or 98%) identical to the VL CDRs of the reference antibody. In some embodiments, an antibody may comprise a VH that is at least 80% (e.g., 85%, 90%, 95%, or 98%) identical to the VH of a reference antibody such as in VH: SEQ ID NO: 55 or 90 or 98; VL: SEQ ID NO: 56 or 88 or 128 and/or a VL variable region that is at least 80% (e.g., 85%, 90%, 95%, or 98%) identical to the VL variable region of the reference antibody.
  • Donor Template
  • The nucleic acid encoding a CAR may be delivered to a T cell that comprises what is referred to herein as a donor template (also referred to as a donor polynucleotide). A donor template can contain a non-homologous sequence, such as the nucleic acid encoding a CAR, flanked by two regions of homology to allow for efficient HDR at a genomic location of interest. Alternatively, a donor template may have no regions of homology to the targeted location in the DNA and may be integrated by NHEJ-dependent end joining following cleavage at the target site.
  • A donor template can be DNA or RNA, single-stranded and/or double-stranded, and can be introduced into a cell in linear or circular form. If introduced in linear form, the ends of the donor sequence can be protected (e.g., from exonucleolytic degradation) by methods known to those of skill in the art. For example, one or more dideoxynucleotide residues are added to the 3′ terminus of a linear molecule and/or self-complementary oligonucleotides are ligated to one or both ends. See, for example, Chang et al., (1987) Proc. Natl. Acad. Sci. USA 84:4959-4963; Nehls et al., (1996) Science 272:886-889. Additional methods for protecting exogenous polynucleotides from degradation include, but are not limited to, addition of terminal amino group(s) and the use of modified internucleotide linkages such as, for example, phosphorothioates, phosphoramidates, and O-methyl ribose or deoxyribose residues.
  • A donor template can be introduced into a cell as part of a vector molecule having additional sequences such as, for example, replication origins, promoters and genes encoding antibiotic resistance. Moreover, a donor template can be introduced as naked nucleic acid, as nucleic acid complexed with an agent such as a liposome or poloxamer, or can be delivered by viruses (e.g., adenovirus, AAV, herpesvirus, retrovirus, lentivirus and integrase defective lentivirus (IDLV)).
  • A donor template, in some embodiments, is inserted so that its expression is driven by the endogenous promoter at the integration site, namely the promoter that drives expression of the endogenous gene into which the donor is inserted. However, in some embodiments, the donor template comprises an exogenous promoter and/or enhancer, for example a constitutive promoter, an inducible promoter, or tissue-specific promoter. In some embodiments, the exogenous promoter is an EF1α promoter comprising a sequence of SEQ ID NO: 79. Other promoters may be used.
  • Furthermore, exogenous sequences may also include transcriptional or translational regulatory sequences, for example, promoters, enhancers, insulators, internal ribosome entry sites, sequences encoding 2A peptides and/or polyadenylation signals.
  • Delivery Methods and Constructs
  • Nucleases and/or donor templates may be delivered using a vector system, including, but not limited to, plasmid vectors, DNA minicircles, retroviral vectors, lentiviral vectors, adenovirus vectors, poxvirus vectors; herpesvirus vectors and adeno-associated virus vectors, and combinations thereof.
  • Conventional viral and non-viral based gene transfer methods can be used to introduce nucleic acids encoding nucleases and donor templates in cells (e.g., T cells). Non-viral vector delivery systems include DNA plasmids, DNA minicircles, naked nucleic acid, and nucleic acid complexed with a delivery vehicle such as a liposome or poloxamer. Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes after delivery to the cell.
  • Methods of non-viral delivery of nucleic acids include electroporation, lipofection, microinjection, biolistics, virosomes, liposomes, immunoliposomes, polycation or lipid:nucleic acid conjugates, naked DNA, naked RNA, capped RNA, artificial virions, and agent-enhanced uptake of DNA. Sonoporation using, e.g., the Sonitron 2000 system (Rich-Mar) can also be used for delivery of nucleic acids.
  • Adeno-Associated Viral Delivery
  • The donor nucleic acid encoding a CAR construct can be delivered to a cell using an adeno-associated virus (AAV). AAVs are small viruses which integrate site-specifically into the host genome and can therefore deliver a transgene, such as CAR. Inverted terminal repeats (ITRs) are present flanking the AAV genome and/or the transgene of interest and serve as origins of replication. Also present in the AAV genome are rep and cap proteins which, when transcribed, form capsids which encapsulate the AAV genome for delivery into target cells. Surface receptors on these capsids which confer AAV serotype, which determines which target organs the capsids will primarily bind and thus what cells the AAV will most efficiently infect. There are twelve currently known human AAV serotypes. In some embodiments, the AAV is AAV serotype 6 (AAV6).
  • Adeno-associated viruses are among the most frequently used viruses for gene therapy for several reasons. First, AAVs do not provoke an immune response upon administration to mammals, including humans. Second, AAVs are effectively delivered to target cells, particularly when consideration is given to selecting the appropriate AAV serotype. Finally, AAVs have the ability to infect both dividing and non-dividing cells because the genome can persist in the host cell without integration. This trait makes them an ideal candidate for gene therapy.
  • Homology-Directed Repair (HDR)
  • The donor nucleic acid encoding a CAR is inserted by homology directed repair (HDR) into the target gene locus. Both strands of the DNA at the target locus are cut by a CRISPR Cas9 enzyme. HDR then occurs to repair the double-strand break (DSB) and insert the donor DNA. For this to occur correctly, the donor sequence is designed with flanking residues which are complementary to the sequence surrounding the DSB site in the target gene (hereinafter “homology arms”). These homology arms serve as the template for DSB repair and allow HDR to be an essentially error-free mechanism. The rate of homology directed repair (HDR) is a function of the distance between the mutation and the cut site so choosing overlapping or nearby target sites is important. Templates can include extra sequences flanked by the homologous regions or can contain a sequence that differs from the genomic sequence, thus allowing sequence editing.
  • The target gene can be associated with an immune response in a subject, wherein permanently deleting at least a portion of the target gene will modulate the immune response. For example, to generate a CAR T cell, the target gene can be the TCRα constant region (TRAC). Disruption of TRAC leads to loss of function of the endogenous TCR.
  • In some embodiments, the target gene is in a safe harbor locus.
  • Engineered T Cells
  • Engineered (gene edited) CAR T cells of the present disclosure may be autologous (“self”) or non-autologous (“non-self,” e.g., allogeneic, syngeneic or xenogeneic). “Autologous” refers to cells from the same subject. “Allogeneic” refers to cells of the same species as a subject, but that differ genetically to the cells in the subject. In some embodiments, the T cells are obtained from a mammalian subject. In some embodiments, the T cells are obtained from a human subject.
  • T cells can be obtained from a number of sources including, but not limited to, peripheral blood mononuclear cells, bone marrow, lymph nodes tissue, cord blood, thymus issue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors. In certain embodiments, T cells can be obtained from a unit of blood collected from a subject using any number of techniques known to the skilled person, such as sedimentation, e.g., FICOLL™ separation.
  • In some embodiments, an isolated population of T cells is used. In some embodiments, after isolation of peripheral blood mononuclear cells (PBMC), both cytotoxic and helper T lymphocytes can be sorted into naive, memory, and effector T cell subpopulations either before or after activation, expansion, and/or genetic modification.
  • A specific subpopulation of T cells, expressing one or more of the following cell surface markers: TCRab, CD3, CD4, CD8, CD27 CD28, CD38 CD45RA, CD45RO, CD62L, CD127, CD122, CD95, CD197, CCR7, KLRG1, MCH-I proteins and/or MCH-II proteins, can be further isolated by positive or negative selection techniques. In some embodiments, a specific subpopulation of T cells, expressing one or more of the markers selected from the group consisting of TCRab, CD4 and/or CD8, is further isolated by positive or negative selection techniques. In some embodiments, the engineered T cell populations do not express or do not substantially express one or more of the following markers: CD70, CD57, CD244, CD160, PD-1, CTLA4, HM3, and LAG3. In some embodiments, subpopulations of T cells may be isolated by positive or negative selection prior to genetic engineering and/or post genetic engineering.
  • In some embodiments, an isolated population of T cells expresses one or more of the markers including, but not limited to a CD3+, CD4+, CD8+, or a combination thereof. In some embodiments, the T cells are isolated from a subject and first activated and stimulated to proliferate in vitro prior to undergoing gene editing.
  • To achieve sufficient therapeutic doses of T cell compositions, T cells are often subjected to one or more rounds of stimulation, activation and/or expansion. T cells can 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; and 6,867,041. In some embodiments, T cells are activated and expanded for about 1 day to about 4 days, about 1 day to about 3 days, about 1 day to about 2 days, about 2 days to about 3 days, about 2 days to about 4 days, about 3 days to about 4 days, or about 1 day, about 2 days, about 3 days, or about 4 days prior to introduction of the genome editing compositions into the T cells.
  • In some embodiments, T cells are activated and expanded for about 4 hours, about 6 hours, about 12 hours, about 18 hours, about 24 hours, about 36 hours, about 48 hours, about 60 hours, or about 72 hours prior to introduction of the gene editing compositions into the T cells.
  • In some embodiments, T cells are activated at the same time that genome editing compositions are introduced into the T cells.
  • Treatment Methods and Compositions
  • Provided herein, in some embodiments, are methods for treating cancer (e.g.: breast cancer). Non-limiting examples of cancers that may be treated as provided herein include: breast cancer, e.g., estrogen receptor-positive breast cancer, prostate cancer, squamous tumors, e.g., of the skin, bladder, lung, cervix, endometrium, head neck, and biliary tract, and neuronal tumors. In some embodiments, the methods comprise delivering the CAR T cells (e.g., anti-LIV1 CAR T cells) of the present disclosure to a subject having cancer, including, breast cancer, e.g., estrogen receptor-positive breast cancer, prostate cancer, squamous tumors, e.g., of the skin, bladder, lung, cervix, endometrium, head neck, and biliary tract, and/or neuronal tumors.
  • The step of administering may include the placement (e.g., transplantation) of cells, e.g., engineered T cells, into a subject, by a method or route that results in at least partial localization of the introduced cells at a desired site, such as tumor, such that a desired effect(s) is produced. Engineered T cells can be administered by any appropriate route that results in delivery to a desired location in the subject where at least a portion of the implanted cells or components of the cells remain viable. The period of viability of the cells after administration to a subject can be as short as a few hours, e.g., twenty-four hours, to a few days, to as long as several years, or even the life time of the subject, i.e., long-term engraftment. For example, in some aspects described herein, an effective amount of engineered T cells is administered via a systemic route of administration, such as an intraperitoneal or intravenous route.
  • A subject may be any subject for whom diagnosis, treatment, or therapy is desired. In some embodiments, the subject is a mammal. In some embodiments, the subject is a human.
  • A donor is an individual who is not the subject being treated. A donor is an individual who is not the patient. In some embodiments, a donor is an individual who does not have or is not suspected of having the cancer being treated. In some embodiments, multiple donors, e.g., two or more donors, are used.
  • In some embodiments, an engineered T cell population being administered according to the methods described herein comprises allogeneic T cells obtained from one or more donors. Allogeneic refers to a cell, cell population, or biological samples comprising cells, obtained from one or more different donors of the same species, where the genes at one or more loci are not identical to the recipient. For example, an engineered T cell population, being administered to a subject can be derived from one or more unrelated donors, or from one or more non-identical siblings. In some embodiments, syngeneic cell populations may be used, such as those obtained from genetically identical donors, (e.g., identical twins). In some embodiments, the cells are autologous cells; that is, the engineered T cells are obtained or isolated from a subject and administered to the same subject, i.e., the donor and recipient are the same.
  • In some embodiments, an engineered T cell population being administered according to the methods described herein does not induce toxicity in the subject, e.g., the engineered T cells do not induce toxicity in non-cancer cells. In some embodiments, an engineered T cell population being administered does not trigger complement mediated lysis, or does not stimulate antibody-dependent cell mediated cytotoxicity (ADCC).
  • An effective amount refers to the amount of a population of engineered T cells needed to prevent or alleviate at least one or more signs or symptoms of a medical condition (e.g., cancer), and relates to a sufficient amount of a composition to provide the desired effect, e.g., to treat a subject having a medical condition. An effective amount also includes an amount sufficient to prevent or delay the development of a symptom of the disease, alter the course of a symptom of the disease (for example but not limited to, slow the progression of a symptom of the disease), or reverse a symptom of the disease. It is understood that for any given case, an appropriate effective amount can be determined by one of ordinary skill in the art using routine experimentation.
  • For use in the various aspects described herein, an effective amount of cells (e.g., engineered T cells) comprises at least 102 cells, at least 5×102 cells, at least 103 cells, at least 5×103 cells, at least 104 cells, at least 5×104 cells, at least 105 cells, at least 2×105 cells, at least 3×105 cells, at least 4×105 cells, at least 5×105 cells, at least 6×105 cells, at least 7×105 cells, at least 8×105 cells, at least 9×105 cells, at least 1×106 cells, at least 2×106 cells, at least 3×106 cells, at least 4×106 cells, at least 5×106 cells, at least 6×106 cells, at least 7×106 cells, at least 8×106 cells, at least 9×106 cells, or multiples thereof. The cells are derived from one or more donors, or are obtained from an autologous source. In some examples described herein, the cells are expanded in culture prior to administration to a subject in need thereof.
  • Modes of administration include injection, infusion, instillation, or ingestion. Injection includes, without limitation, intravenous, intramuscular, intra-arterial, intrathecal, intraventricular, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, sub capsular, subarachnoid, intraspinal, intracerebro spinal, and intrasternal injection and infusion. In some embodiments, the route is intravenous.
  • In some embodiments, engineered T cells are administered systemically, which refers to the administration of a population of cells other than directly into a target site, tissue, or organ, such that it enters, instead, the subject's circulatory system and, thus, is subject to metabolism and other like processes.
  • The efficacy of a treatment comprising a composition for the treatment of a medical condition can be determined by the skilled clinician. A treatment is considered “effective treatment,” if any one or all of the signs or symptoms of, as but one example, levels of functional target are altered in a beneficial manner (e.g., increased by at least 10%), or other clinically accepted symptoms or markers of disease (e.g., cancer) are improved or ameliorated. Efficacy can also be measured by failure of a subject to worsen as assessed by hospitalization or need for medical interventions (e.g., progression of the disease is halted or at least slowed). Methods of measuring these indicators are known to those of skill in the art and/or described herein. Treatment includes any treatment of a disease in subject and includes: (1) inhibiting the disease, e.g., arresting, or slowing the progression of symptoms; or (2) relieving the disease, e.g., causing regression of symptoms; and (3) preventing or reducing the likelihood of the development of symptoms.
  • The present disclosure is exemplified by the following embodiments:
  • Embodiment 1. An engineered T cell comprising a nucleic acid encoding a chimeric antigen receptor (CAR), wherein the CAR comprise an ectodomain that binds specifically to LIV1.
  • Embodiment 2. The engineered T cell of embodiment 1 further comprising a disrupted T cell receptor alpha chain constant region (TRAC) gene.
  • Embodiment 3. The engineered T cell of embodiment 2, wherein the nucleic acid encoding the CAR is inserted into the TRAC gene.
  • Embodiment 4. The engineered T cell of any one of embodiments 1-3 further comprising a disrupted beta-2-microglobulin (P211 gene.
  • Embodiment 5. The engineered T cell of any one of embodiments 1-4, wherein the ectodomain of the CAR comprises an anti-LIV1 antibody.
  • Embodiment 6. The engineered T cell of embodiment 5, wherein the anti-LIV1 antibody is an anti-LIV1 single-chain variable fragment (scFv).
  • Embodiment 7. The engineered T cell of embodiment 6, wherein the anti-LIV1 scFv comprises the same heavy chain variable region (VH) complementarity determining regions (CDRs) and the same light chain variable region (VL) CDRs as a reference antibody, wherein the reference antibody comprises (i) a VH set forth as SEQ ID NO: 55 and a VL set forth as SEQ ID NO: 56, (ii) a VH set forth as SEQ ID NO: 69 and a VL set forth as SEQ ID NO: 70, (iii) a VH set forth as SEQ ID NO: 76 and a VL set forth as SEQ ID NO: 77, or (iv) a VH set forth as SEQ ID NO: 83 and a VL set forth as SEQ ID NO: 84.
  • Embodiment 8. The engineered T cell of embodiment 7, wherein the anti-LIV1 scFv comprises the same VH and VL chains as the reference antibody.
  • Embodiment 8.1. The engineered T cell of embodiment 7, wherein the anti-LIV1 scFv comprises the amino acid sequence of any one of SEQ ID NOs: 54 or 70.
  • Embodiment 9. The engineered T cell of any one of embodiments 1-8.1, wherein the CAR comprises a CD28 co-stimulatory domain or a 41BB co-stimulatory domain.
  • Embodiment 10. The engineered T cell of embodiment 9, wherein the CAR further comprises a CD3z cytoplasmic signaling domain.
  • Embodiment 11. The engineered T cell of any one of embodiments 3-10, wherein the TRAC gene comprises the nucleotide sequence of any one of SEQ ID NOS: 63, 64, 71, or 72, and/or wherein the CAR is encoded by the nucleotide sequence of any one of SEQ ID NOS: 49, 51, 65, or 67.
  • Embodiment 12. The engineered T cell of any one of embodiments 4-11, wherein the disrupted β2M gene comprises at least one nucleotide sequence selected from any one of SEQ ID NOS: 9-14.
  • Embodiment 13. A population of the engineered T cell of any one of embodiments 1-12, wherein at least 25% or at least 50% of engineered T cells of the population express the CAR.
  • Embodiment 14. The population of embodiment 14, wherein at least 70% of engineered T cells of the population express the CAR.
  • Embodiment 15. The population of embodiment 13, wherein at least 25% of engineered T cells of the population express the CAR following at least 7 or at least 14 days of in vitro proliferation.
  • Embodiment 16. The population of any one of embodiments 13-15, wherein at least 50% of engineered T cells of the population do not express a detectable level of T cell receptor (TCR) protein.
  • Embodiment 17. The population of embodiment 16, wherein at least 90% of engineered T cells of the population do not express a detectable level of TCR protein.
  • Embodiment 18. The population of any one of embodiments 13-17, wherein at least 50% of engineered T cells of the population do not express a detectable level of β2M protein.
  • Embodiment 19. The population of embodiment 18, wherein at least 70% of engineered T cells of the population do not express a detectable level of β2M protein.
  • Embodiment 20. The population of any one of embodiments 13-19, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells that express LIV1, induce cell lysis of at least 10%, at least 25%, or at least 50% of the cancer cells of the population.
  • Embodiment 21. The population of embodiment 20, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells that express LIV1, induce cell lysis of at least 70%, at least 80%, or at least 90% of the population of cancer cells.
  • Embodiment 22. The population of embodiments 20 or 21, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells, secrete IFNγ.
  • Embodiment 23. The population of any one of embodiments 20-22, wherein the ratio of engineered T cells to cancer cells is 1:1 to 2:1.
  • Embodiment 24. The population of any one of embodiments 20-23, wherein the cancer cells comprise sarcoma cells.
  • Embodiment 25. The population of any one of embodiments 20-23, wherein the cancer cells comprise breast cancer cells.
  • Embodiment 27. The population of any one of embodiments 13-26, when administered in vivo to a subject, does not induce toxicity in the subject.
  • Embodiment 26. A method comprising administering the population of engineered T cells any one of embodiments 13-27 to a subject.
  • Embodiment 27. The method of embodiment 26, wherein the subject is a human subject.
  • Embodiment 28. The method of embodiment 27, wherein the subject has a cancer.
  • Embodiment 29. The method of embodiment 28, wherein the cancer is selected from the group consisting of: breast cancer, e.g., estrogen receptor-positive breast cancer, prostate cancer, squamous tumors, e.g., of the skin, bladder, lung, cervix, endometrium, head neck, and biliary tract, and/or neuronal tumors.
  • Embodiment 30. The method of embodiments 28 or 31, wherein the cancer comprises cancer cells expressing LIV1.
  • Embodiment 31. A method for producing an engineered T cell, the method comprising (a) delivering to a T cell, a RNA-guided nuclease, a gRNA targeting a TRAC gene, and a vector comprising a donor template that comprises a nucleic acid encoding a CAR that comprise an ectodomain that binds specifically to LIV1; and (b) producing an engineered T cell having a disrupted TRAC gene and expressing the CAR.
  • Embodiment 32. The method of embodiment 31, wherein the gRNA targeting the TRAC gene comprises the nucleotide sequence of SEQ ID NO: 18 or 19, or targets the nucleotide sequence of SEQ ID NO: 40.
  • Embodiment 33. The method of embodiments 31 or 32, wherein the nucleic acid encoding the CAR is flanked by left and right homology arms to the TRAC gene.
  • Embodiment 34. The method of any one of embodiments 31-33 further comprising delivering to the T cell a gRNA targeting the β2M gene.
  • Embodiment 35. The method of embodiment 34, wherein the gRNA targeting the β2M gene comprises the nucleotide sequence of SEQ ID NO: 20 or 21, or targets the nucleotide sequence of SEQ ID NO: 41.
  • Embodiment 36. The method of any one of embodiments 31-35, wherein the RNA-guided nuclease is a Cas9 nuclease, optionally a S. pyogenes Cas9 nuclease.
  • Embodiment 37. The method of any one of embodiments 31-38, wherein the ectodomain of the CAR is an anti-LIV1 antibody.
  • Embodiment 38. The method of embodiment 37, wherein the anti-LIV1 antibody is an anti-LIV1 single-chain variable fragment (scFv).
  • Embodiment 39. The method of embodiment 38, wherein the anti-LIV1 scFv comprises the same VH complementarity determining regions (CDRs) and the same VL CDRs as a reference antibody, wherein the reference antibody comprises (i) a VH set forth as SEQ ID NO: 55 and a VL set forth as SEQ ID NO: 56.
  • Embodiment 40. The method of embodiment 39, wherein the anti-LIV1 scFv comprises the same VH and VL chains as the reference antibody.
  • Embodiment 41. The method of embodiment 39, wherein the anti-LIV1 scFv comprises the amino acid sequence of any one of SEQ ID NOs: 54 or 70.
  • Embodiment 42. The method of any one of embodiments 31-41, wherein the CAR comprises a CD28 co-stimulatory domain or a 41BB co-stimulatory domain.
  • Embodiment 43. The method of embodiment 42, wherein the CAR further comprises a CD3z cytoplasmic signaling domain.
  • Embodiment 44. The method of any one of embodiments 31-43, wherein the donor template comprises the nucleotide sequence of any one of SEQ ID NOS: 63, 64, 71, or 72.
  • Embodiment 45. The method of any one of embodiments 31-44, wherein the CAR is encoded by a nucleotide sequence of any one of SEQ ID NOS: 49, 51, 65, or 67.
  • The present disclosure is further exemplified by the following embodiments:
  • Embodiment A1. An engineered T cell comprising a nucleic acid encoding a chimeric antigen receptor (CAR), wherein the CAR comprises an ectodomain that binds specifically to LIV1.
  • Embodiment A2. The engineered T cell of embodiment A1 further comprising a disrupted T cell receptor alpha chain constant region (TRAC) gene.
  • Embodiment A3. The engineered T cell of embodiment A1 or A2 further comprising a disrupted beta-2-microglobulin (β2M) gene.
  • Embodiment A4. The engineered T cell of any one of embodiments A1-3, wherein the ectodomain of the CAR comprises an anti-LIV1 antibody.
  • Embodiment A5. The engineered T cell of embodiment A4, wherein the anti-LIV1 antibody is an anti-LIV1 single-chain variable fragment (scFv).
  • Embodiment A6. The engineered T cell of embodiment A5, wherein the anti-LIV1 scFv comprises the same heavy chain variable domain (VH) complementarity determining regions (CDRs) and the same light chain variable domain (VL) CDRs as a reference antibody, wherein the reference antibody comprises (i) a VH set forth as SEQ ID NO: 55 and a VL set forth as SEQ ID NO: 56 or (ii) a VH set forth as SEQ ID NO: 90 and a VL set forth as SEQ ID NO: 88.
  • Embodiment A7. The engineered T cell of embodiment A6, wherein the anti-LIV1 scFv comprises the same VH and VL chains as the reference antibody.
  • Embodiment A8. The engineered T cell of embodiment A6, wherein the anti-LIV1 scFv comprises the amino acid sequence of any one of SEQ ID NOs: 54, 70, 83, or 86.
  • Embodiment A9. The engineered T cell of any one of embodiments A1-A8, wherein the CAR further comprises a CD28 co-stimulatory domain or a 41BB co-stimulatory domain.
  • Embodiment A10. The engineered T cell of embodiment A9, wherein the CAR further comprises a CD3 cytoplasmic signaling domain.
  • Embodiment A11. The engineered T cell of any one of embodiments A1-A10, wherein the CAR is encoded by the nucleotide sequence of any one of SEQ ID NOs: 49, 51, 104, or 108 or a nucleotide sequence comprising a nucleic acid sequence that is at least 90% identical to SEQ ID NOs: 49, 51, 104, or 108.
  • Embodiment A12. The engineered T cell of any one of embodiments A1-A11, wherein the nucleic acid encoding the CAR is inserted into the disrupted TRAC gene.
  • Embodiment A13. The engineered T cell of any one of embodiments A2-A12, wherein the disrupted TRAC gene comprises the nucleotide sequence of any one of SEQ ID NOs: 63, 64, 107, or 111, and/or the nucleotide sequence of any one of SEQ ID NOs: 49, 51, 104, or 108.
  • Embodiment A14. The engineered T cell of any one of embodiments A4-A13, wherein the disrupted β2M gene comprises at least one nucleotide sequence selected from any one of SEQ ID NOs: 9-14.
  • Embodiment A15. An engineered T cell comprising: (i) a disrupted TRAC gene; (ii) a disrupted β2M gene; and (iii) a nucleic acid encoding a CAR comprising an anti-LIV1 antigen-binding fragment.
  • Embodiment A16. The engineered T cell of embodiment A15, wherein the CAR comprises (a) an ectodomain that comprises an anti-LIV1 antigen-binding fragment, (b) a CD8 transmembrane domain, and (c) an endodomain that comprises a 41BB co-stimulatory domain and a CD3ζ cytoplasmic signaling domain.
  • Embodiment A17. The engineered T cell of embodiments A15 or A16, wherein the disrupted TRAC gene comprises the nucleic acid encoding the CAR.
  • Embodiment A18. An engineered T cell comprising: (i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR comprising (a) an ectodomain that comprises an anti-LIV1 antigen-binding fragment, (b) a CD8 transmembrane domain, and (c) an endodomain that comprises a 41BB co-stimulatory domain and a CD3ζ cytoplasmic signaling domain; and (ii) a disrupted β2M gene.
  • Embodiment A19. An engineered T cell comprising: (i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR comprising an amino acid sequence of any one of SEQ ID NOs: 50, 52, 105, 109, 68 or 66; and (ii) a disrupted β2M gene.
  • Embodiment A20. An engineered T cell comprising: (i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR, wherein the nucleic acid sequence is at least 90% identical to SEQ ID NOs: 49, 51, 104, or 108 and/or encodes a CAR comprising an amino acid sequence of any one of SEQ ID NOs: 50, 52, 105, 109, 68 or 66; and (ii) a disrupted β2M gene.
  • Embodiment A21. The engineered T cell of any one of embodiments A1-A20, wherein the T cell is a human T cell.
  • Embodiment A22. A population of cells comprising the engineered T cell of any one of embodiments A1-A21, wherein at least 15% or at least 50% of engineered T cells of the population express the CAR.
  • Embodiment A23. The population of embodiment A22, wherein at least 30% of engineered T cells of the population express the CAR.
  • Embodiment A24. The population of embodiment A22, wherein at least 70% of engineered T cells of the population express the CAR.
  • Embodiment A25. The population of embodiment A22, wherein at least 25% of engineered T cells of the population express the CAR following at least 7 days or at least 14 days of in vitro proliferation.
  • Embodiment A26. The population of any one of embodiments A22-A25, wherein at least 50% of engineered T cells of the population do not express a detectable level of T cell receptor (TCR) protein.
  • Embodiment A27. The population of embodiments A26, wherein at least 90% of engineered T cells of the population do not express a detectable level of TCR protein.
  • Embodiment A28. The population of any one of embodiments A22-A27, wherein at least 50% of engineered T cells of the population do not express a detectable level of β2M protein.
  • Embodiment A29. The population of embodiment A28, wherein at least 70% of engineered T cells of the population do not express a detectable level of β2M protein.
  • Embodiment A30. The population of any one of embodiments A22-A29, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells that express LIV1, induce cell lysis of at least 10%, at least 25%, or at least 50% of the cancer cells of the population.
  • Embodiment A31. The population of embodiment A30, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells that express LIV1, induce cell lysis of at least 70%, at least 80%, or at least 90% of the population of cancer cells.
  • Embodiment A32. The population of embodiments A30 or A31, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells, secrete IFNγ.
  • Embodiment A33. The population of any one of embodiments A30-A32, wherein the ratio of engineered T cells to cancer cells is 1:1 to 2:1.
  • Embodiment A34. The population of any one of embodiments A30-A33, wherein the cancer cells comprise sarcoma cells.
  • Embodiment A35. The population of any one of embodiments A30-A33, wherein the cancer cells comprise breast cancer cells.
  • Embodiment A36. The population of any one of embodiments A22-A35, when administered in vivo to a subject, does not induce toxicity in the subject.
  • Embodiment A37. A population of cells comprising engineered T cells, wherein the engineered T cells comprise: (i) a disrupted TRAC gene; (ii) a disrupted β2M gene; and (iii) a nucleic acid encoding a CAR comprising an anti-LIV1 antigen-binding fragment.
  • Embodiment A38. The population of cells of embodiment A37, wherein the CAR comprises (a) an ectodomain that comprises an anti-LIV1 antigen-binding fragment, (b) a CD8 transmembrane domain, and (c) an endodomain that comprises a 41BB co-stimulatory domain and a CD3ζ cytoplasmic signaling domain.
  • Embodiment A39. The population of cells of embodiments A37 or A38, wherein the disrupted TRAC gene comprises the nucleic acid encoding the CAR.
  • Embodiment A40. A population of cells comprising engineered T cells, wherein the engineered T cells comprise: (i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR comprising (a) an ectodomain that comprises an anti-LIV1 antigen-binding fragment, (b) a CD8 transmembrane domain, and (c) an endodomain that comprises a 41BB co-stimulatory domain and a CD3ζ cytoplasmic signaling domain; and (ii) a disrupted β2M gene.
  • Embodiment A41. A population of cells comprising engineered T cells, wherein the engineered T cells comprise: (i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR, wherein the nucleic acid sequence is at least 90% identical to SEQ ID NOs: 49, 51, 104, or 108 and/or encodes the CAR of SEQ ID NOs: 50, 52, 105, 109, 68 or 66; and (ii) a disrupted β2M gene.
  • Embodiment A42. A method comprising administering the population of engineered T cells any one of embodiments A22-A41 to a subject.
  • Embodiment A43. The method of embodiment A42, wherein the subject is a human subject.
  • Embodiment A44. The method of embodiment A43, wherein the subject has a cancer.
  • Embodiment A45. The method of embodiment A44, wherein the cancer is selected from the group consisting of: breast cancer, e.g., estrogen receptor-positive breast cancer, prostate cancer, squamous tumors, e.g., of the skin, bladder, lung, cervix, endometrium, head neck, and biliary tract, and/or neuronal tumors.
  • Embodiment A46. The method of embodiments A44 or A45, wherein the cancer comprises cancer cells expressing LIV1.
  • Embodiment A47. A method for producing an engineered T cell, the method comprising (a) delivering to a T cell (i) a RNA-guided nuclease, (ii) a gRNA targeting a TRAC gene, and (iii) a vector comprising a donor template that comprises a nucleic acid encoding a CAR that comprise an ectodomain that binds specifically to LIV1; and (b) producing an engineered T cell having a disrupted TRAC gene and expressing the CAR.
  • Embodiment A48. The method of embodiments A47, wherein the gRNA targeting the TRAC gene comprises the nucleotide sequence of SEQ ID NO: 18 or 19, or targets the nucleotide sequence of SEQ ID NO: 40.
  • Embodiment A49. The method of embodiments A47 or A48 further comprising delivering to the T cell a gRNA targeting the β2M gene.
  • Embodiment A50. The method of embodiments A49, wherein the gRNA targeting the β2M gene comprises the nucleotide sequence of SEQ ID NO: 20 or 21, or targets the nucleotide sequence of SEQ ID NO: 41.
  • Embodiment A51. The method of any one of embodiments A47-A50, wherein the ectodomain of the CAR comprises an anti-LIV1 antibody.
  • Embodiment A52. The method of embodiment A51, wherein the anti-LIV1 antibody is an anti-LIV1 single-chain variable fragment (scFv).
  • Embodiment A53. The method of embodiment A52, wherein the anti-LIV1 scFv comprises the same heavy chain variable domain (VH) complementarity determining regions (CDRs) and the same light chain variable domain (VL) CDRs as a reference antibody, wherein the reference antibody comprises (i) a VH set forth as SEQ ID NO: 55 and a VL set forth as SEQ ID NO: 56, or (ii) a VH set forth as SEQ ID NO: 90 and a VL set forth as SEQ ID NO: 88.
  • Embodiment A54. The method of embodiment A53, wherein the anti-LIV1 scFv comprises the same VH and VL chains as the reference antibody.
  • Embodiment A55. The method of embodiment A53, wherein the anti-LIV1 scFv comprises the amino acid sequence of any one of SEQ ID NOs: 54, 83, 86 or 70.
  • Embodiment A56. The method of any one of embodiments A47-A56, wherein the CAR further comprises a CD28 co-stimulatory domain or a 41BB co-stimulatory domain.
  • Embodiment A57. The method of embodiment A56, wherein the CAR further comprises a CD3ζ cytoplasmic signaling domain.
  • Embodiment A58. The method of any one of embodiments A47-A57, wherein the CAR is encoded by a nucleotide sequence of any one of SEQ ID NOs: 49, 51, 104, or 108 or a nucleotide sequence comprising a nucleic acid sequence that is at least 90% identical to SEQ ID NOs: 49, 51, 104, or 108.
  • Embodiment A59. The method of any one of embodiments A47-A58, wherein the nucleic acid encoding the CAR is flanked by left and right homology arms to the TRAC gene.
  • Embodiment A60. The method of any one of embodiments A47-A59, wherein the donor template comprises the nucleotide sequence of any one of SEQ ID NOs: 63, 64, 107, or 111.
  • Embodiment A61. The method of any one of embodiments A47-A60, wherein the RNA-guided nuclease is a Cas9 nuclease, optionally a S. pyogenes Cas9 nuclease.
  • Embodiment A62. An engineered T cell produced by the method of any one of embodiments A47-A61.
  • Embodiment A63. A population of cells comprising the engineered T cell of embodiment A62.
  • Embodiment A64. A method of treating cancer in a subject, comprising administering to the subject the population of cells of any one of embodiments A22-A41 or A63.
  • Embodiment A65. The method of embodiment A64, wherein the cancer is selected from the group consisting of: pancreatic cancer, gastric cancer, ovarian cancer, uterine cancer, breast cancer, prostate cancer, testicular cancer, thyroid cancer, nasopharyngeal cancer, non-small cell lung (NSCLC), glioblastoma, neuronal, soft tissue sarcomas, leukemia, lymphoma, melanoma, colon cancer, colon adenocarcinoma, brain glioblastoma, hepatocellular carcinoma, liver hepatocholangiocarcinoma, osteosarcoma, gastric cancer, esophagus squamous cell carcinoma, advanced stage pancreas cancer, lung adenocarcinoma, lung squamous cell carcinoma, lung small cell cancer, renal carcinoma, and intrahepatic biliary cancer.
  • Embodiment A66. The method of embodiments A64 or A65, wherein the cancer comprises cancer cells expressing LIV1.
  • EXAMPLES Example 1. CAR T Cell Generation and CAR Expression
  • Activated primary human T cells were electroporated with Cas9:gRNA RNP complexes and adeno-associated adenoviral vectors (AAVs) to generate TRAC/β2M/anti-Liv1a CAR+ T cells. Recombinant AAV serotype 6 (AAV6) comprising one of the nucleotide sequences encoding an anti-Livia CAR (971 (SEQ ID NO:49), 972 (SEQ ID NO: 65), 972b (SEQ ID NO: 67), 973 (SEQ ID NO: 95), 974 (SEQ ID NO: 100), 975 (SEQ ID NO: 104), and 976 (SEQ ID NO: 108), were delivered with Cas9:sgRNA RNPs (1 μM Cas9, 5 μM gRNA) to activated allogeneic human T cells. The following sgRNAs were used: TRAC (SEQ ID NO: 28) and β2M (SEQ ID NO: 30). The unmodified versions (or other modified versions) of the gRNAs may also be used (e.g., SEQ ID NO: 18 or 20).
  • About one (1) week post electroporation, cells were processed for flow cytometry to assess TRAC, β2M, and anti-Livia CAR expression levels at the cell surface of the edited cell population (FIG. 1). For all anti-Livia CAR T cells and TRAC/β2M control cells, >90% of viable cells lacked expression of TCR and >60% lacked expression of β2M. The cells treated with the construct encoding the 975 and 976 Livia CAR had the highest percentage of viable cells expressing an anti-Livia CAR+ (>30%).
  • Example 2. Cytotoxicity
  • Cell Kill Assay. A cell killing (cytotoxicity) assay was used to assess the ability of the TRAC-/β2M-/anti-Livia CAR+ T cells to cause cellular lysis in adherent kidney carcinoma and breast cancer cell lines (A498 and ZR-75-1, respectively). Adherent cells were seeded in 96-well plates at 50,000 cells per well and left overnight at 37° C. During the following day, T cells were added to the wells containing target cells at ratios of 8:1, 4:1, 2:1 or 1:1 T cell:target cell. TRAC-/β2M-T cells were used as a negative control. After approximately 24 hours, 100 μLs of supernatant was removed for cytokine quantification (see below) and T cells were removed from the culture by aspiration and 100 μL CellTiter-Glo® (Promega) was added to each well of the plate to assess the number of remaining viable cells. The amount of light emitted from each well was then quantified using a plate reader. The anti-Livia CAR T cells, particularly those expressing the CTX971, CTX975 and CTX976 constructs, exhibited potent cytotoxicity towards the A498 (FIG. 2A) and ZR-75-1 (FIG. 2B) cell lines.
  • Example 3. Effector Cytokine Secretion
  • The MILLIPLEX MAP Human Cytokine/Chemokine Magnetic Bead Panel—Immunology Multiplex Assay kit (Millipore, catalog #HCYTOMAG-60K) using magnetic microspheres, anti-human IFNγ bead (Millipore, catalog #HCYIFNG-MAG) and anti-human IL-2 bead (Millipore, catalog #HIL2-MAG), respectively, was used to quantify IFN-γ and IL-2 secretion in samples from the cytotoxicity assay. The assay was conducted following manufacturer's protocol. MILLIPLEX® standard and quality control (QC) samples were reconstituted, and serial dilutions of the working standards from 10,000 pg/mL to 3.2 pg/mL were prepared. MILLIPLEX® standards, QCs and cell supernatants were added to each plate, and assay media was used to dilute the supernatants. All samples were incubated with anti-human IFNγ and anti-human IL-2 beads for 2 hours. After incubation, the plate was washed using an automated magnetic plate washer. Human cytokine/chemokine detection antibody solution was added to each well and incubated for 1 hour followed by incubation with Streptavidin-Phycoerythrin for 30 minutes. The plate was subsequently washed, samples were resuspended with 150 μL Sheath Fluid, and agitated on a plate shaker for 5 minutes. The samples were read using the Luminex® 100/200™ instrument with xPONENT® software and data acquisition and analysis was completed using MILLIPLEX® Analyst software. The Median Fluorescent Intensity (MFI) data was automatically analyzed using a 5-parameter logistic curve-fitting method for calculating the cytokine concentration measured in the unknown samples.
  • As shown in FIGS. 3A-3D, allogeneic T cells containing the CTX971, 975, or 976 CARs secreted the effector cytokines interferon-γ (3A, B) and interleukin-2 (3C, D) when co-cultured with the target cells lines A498 and ZR-75-1 at levels significantly above background (2KO/AAV neg T cells co-cultured with the target cell lines).
  • TABLE 6
    SEQ ID
    CAR CAR structure NO:
    CTX-971 CD8[signal peptide]-VL-linker-VH-CD8[tm]- 49, 50
    CAR CD28[co-stimulatory domain]-CD3ζ
    CTX-971b CD8[signal peptide]-VL-linker-VH-CD8[tm]- 51, 52
    CAR 41BB[co-stimulatory domain]-CD3ζ
    CTX-972 CD8[signal peptide]-VH-linker-VL-CD8[tm]- 65, 125
    CAR CD28[co-stimulatory domain]-CD3ζ
    CTX-972b CD8[signal peptide]-VH-linker-VL-CD8[tm]- 67, 126
    CAR 41BB[co-stimulatory domain]-CD3ζ
  • TABLE 7
    CAR Components
    CAR Structure:
    CD8[signal peptide]-anti-LIV1[scFV]-CD8[tm]-CD28[co-stimulatory
    domain]-CD3ζ; or
    CD8[signal peptide]-anti-LIV1[scFV]-CD8[tm]-41BB[co-stimulatory
    domain]-CD3ζ
    SEQ
    ID
    Name Sequence NO:
    CTX-971 CCACCATGGCGCTTCCGGTGACAGCACTGCTCCTCCCCTTGGCGCTG  49
    CAR TTGCTCCACGCAGCAAGGCCGGACGTGGTCATGACTCAAAGCCCAC
    CD28  TTTCCTTGCCCGTGACTCTCGGACAACCGGCTTCAATATCTTGCCGC
    co-stim TCATCACAGTCCCTGCTGCATAGCAGTGGTAACACTTATCTTGAGTG
    GTACCAACAGCGGCCCGGCCAATCTCCTAGGCCCCTGATATATAAG
    ATAAGTACTCGCTTTTCCGGGGTCCCGGACCGGTTCAGCGGGTCTGG
    GAGTGGTACAGACTTCACATTGAAGATTTCACGAGTAGAAGCCGAA
    GACGTGGGTGTTTATTACTGCTTCCAAGGATCTCACGTGCCATATAC
    GTTTGGTGGGGGCACAAAAGTCGAGATTAAGGGAGGCGGAGGATC
    AGGAGGTGGGGGAAGTGGAGGTGGTGGGTCACAAGTACAGCTCGT
    GCAATCAGGGGCGGAGGTGAAGAAACCAGGGGCGTCTGTGAAGGT
    AAGCTGTAAGGCATCCGGATTGACAATCGAGGATTATTACATGCAT
    TGGGTCCGCCAGGCACCAGGGCAGGGATTGGAGTGGATGGGGTGGA
    TAGATCCTGAAAATGGGGATACAGAGTATGGCCCTAAGTTCCAGGG
    CAGAGTTACGATGACTCGAGATACTAGCATTAATACGGCCTACATG
    GAGCTTAGCCGCCTGCGGTCCGATGACACGGCCGTTTATTATTGCGC
    CGTACACAATGCGCACTACGGGACATGGTTCGCGTATTGGGGTCAA
    GGAACGCTCGTTACTGTCTCAAGTAGTGCTGCTGCCTTTGTCCCGGT
    ATTTCTCCCAGCCAAACCGACCACGACTCCCGCCCCGCGCCCTCCGA
    CACCCGCTCCCACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAG
    GCATGCCGACCCGCCGCCGGGGGTGCTGTTCATACGAGGGGCTTGG
    ACTTCGCTTGTGATATTTACATTTGGGCTCCGTTGGCGGGTACGTGC
    GGCGTCCTTTTGTTGTCACTCGTTATTACTTTGTATTGTAATCACAGG
    AATCGCTCAAAGCGGAGTAGGTTGTTGCATTCCGATTACATGAATAT
    GACTCCTCGCCGGCCTGGGCCGACAAGAAAACATTACCAACCCTAT
    GCCCCCCCACGAGACTTCGCTGCGTACAGGTCCCGAGTGAAGTTTTC
    CCGAAGCGCAGACGCTCCGGCATATCAGCAAGGACAGAATCAGCTG
    TATAACGAACTGAATTTGGGACGCCGCGAGGAGTATGACGTGCTTG
    ATAAACGCCGGGGGAGAGACCCGGAAATGGGGGGTAAACCCCGAA
    GAAAGAATCCCCAAGAAGGACTCTACAATGAACTCCAGAAGGATAA
    GATGGCGGAGGCCTACTCAGAAATAGGTATGAAGGGCGAACGACG
    ACGGGGAAAAGGTCACGATGGCCTCTACCAAGGGTTGAGTACGGCA
    ACCAAAGATACGTACGATGCACTGCATATGCAGGCCCTGCCTCCCA
    GATAAT
    CTX 971 MALPVTALLLPLALLLHAARPDVVMTQSPLSLPVTLGQPASISCRSSQS  50
    CAR LLHSSGNTYLEWYQQRPGQSPRPLIYKISTRFSGVPDRFSGSGSGTDFTL
    CD28  KISRVEAEDVGVYYCFQGSHVPYTFGGGTKVEIKGGGGSGGGGSGGG
    co-stim GSQVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMHWVRQAPGQGL
    EWMGWIDPENGDTEYGPKFQGRVTMTRDTSINTAYMELSRLRSDDTA
    VYYCAVHNAHYGTWFAYWGQGTLVTVSSSAAAFVPVFLPAKPTTTPA
    PRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGT
    CGVLLLSLVITLYCNHRNRSKRSRLLHSDYMNMTPRRPGPTRKHYQPY
    APPRDFAAYRSRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLD
    KRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRR
    GKGHDGLYQGLSTATKDTYDALHMQALPPR
    CTX-971b CCACCATGGCGCTTCCGGTGACAGCACTGCTCCTCCCCTTGGCGCTG  51
    CAR TTGCTCCACGCAGCAAGGCCGGACGTGGTCATGACTCAAAGCCCAC
    41BB  TTTCCTTGCCCGTGACTCTCGGACAACCGGCTTCAATATCTTGCCGC
    co-stim TCATCACAGTCCCTGCTGCATAGCAGTGGTAACACTTATCTTGAGTG
    GTACCAACAGCGGCCCGGCCAATCTCCTAGGCCCCTGATATATAAG
    ATAAGTACTCGCTTTTCCGGGGTCCCGGACCGGTTCAGCGGGTCTGG
    GAGTGGTACAGACTTCACATTGAAGATTTCACGAGTAGAAGCCGAA
    GACGTGGGTGTTTATTACTGCTTCCAAGGATCTCACGTGCCATATAC
    GTTTGGTGGGGGCACAAAAGTCGAGATTAAGGGAGGCGGAGGATC
    AGGAGGTGGGGGAAGTGGAGGTGGTGGGTCACAAGTACAGCTCGT
    GCAATCAGGGGCGGAGGTGAAGAAACCAGGGGCGTCTGTGAAGGT
    AAGCTGTAAGGCATCCGGATTGACAATCGAGGATTATTACATGCAT
    TGGGTCCGCCAGGCACCAGGGCAGGGATTGGAGTGGATGGGGTGGA
    TAGATCCTGAAAATGGGGATACAGAGTATGGCCCTAAGTTCCAGGG
    CAGAGTTACGATGACTCGAGATACTAGCATTAATACGGCCTACATG
    GAGCTTAGCCGCCTGCGGTCCGATGACACGGCCGTTTATTATTGCGC
    CGTACACAATGCGCACTACGGGACATGGTTCGCGTATTGGGGTCAA
    GGAACGCTCGTTACTGTCTCAAGTAGTGCTGCTGCCTTTGTCCCGGT
    ATTTCTCCCAGCCAAACCGACCACGACTCCCGCCCCGCGCCCTCCGA
    CACCCGCTCCCACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAG
    GCATGCCGACCCGCCGCCGGGGGTGCTGTTCATACGAGGGGCTTGG
    ACTTCGCTTGTGATATTTACATTTGGGCTCCGTTGGCGGGTACGTGC
    GGCGTCCTTTTGTTGTCACTCGTTATTACTTTGTATTGTAATCACAGG
    AATCGCAAACGGGGCAGAAAGAAACTCCTGTATATATTCAAACAAC
    CATTTATGAGACCAGTACAAACTACTCAAGAGGAAGATGGCTGTAG
    CTGCCGATTTCCAGAAGAAGAAGAAGGAGGATGTGAACTGCGAGTG
    AAGTTTTCCCGAAGCGCAGACGCTCCGGCATATCAGCAAGGACAGA
    ATCAGCTGTATAACGAACTGAATTTGGGACGCCGCGAGGAGTATGA
    CGTGCTTGATAAACGCCGGGGGAGAGACCCGGAAATGGGGGGTAA
    ACCCCGAAGAAAGAATCCCCAAGAAGGACTCTACAATGAACTCCAG
    AAGGATAAGATGGCGGAGGCCTACTCAGAAATAGGTATGAAGGGC
    GAACGACGACGGGGAAAAGGTCACGATGGCCTCTACCAAGGGTTGA
    GTACGGCAACCAAAGATACGTACGATGCACTGCATATGCAGGCCCT
    GCCTCCCAGATAAT
    CTX-971b MALPVTALLLPLALLLHAARPDVVMTQSPLSLPVTLGQPASISCRSSQS  52
    CAR LLHSSGNTYLEWYQQRPGQSPRPLIYKISTRFSGVPDRFSGSGSGTDFTL
    41BB co-stim KISRVEAEDVGVYYCFQGSHVPYTFGGGTKVEIKGGGGSGGGGSGGG
    GSQVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMHWVRQAPGQGL
    EWMGWIDPENGDTEYGPKFQGRVTMTRDTSINTAYMELSRLRSDDTA
    VYYCAVHNAHYGTWFAYWGQGTLVTVSSSAAAFVPVFLPAKPTTTPA
    PRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGT
    CGVLLLSLVITLYCNHRNRKRGRKKLLYIFKQPFMRPVQTTQEEDGCSC
    RFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLD
    KRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRR
    GKGHDGLYQGLSTATKDTYDALHMQALPPR
    CTX-971 and GACGTGGTCATGACTCAAAGCCCACTTTCCTTGCCCGTGACTCTCGG  53
    CTX-971b ACAACCGGCTTCAATATCTTGCCGCTCATCACAGTCCCTGCTGCATA
    scFv GCAGTGGTAACACTTATCTTGAGTGGTACCAACAGCGGCCCGGCCA
    ATCTCCTAGGCCCCTGATATATAAGATAAGTACTCGCTTTTCCGGGG
    TCCCGGACCGGTTCAGCGGGTCTGGGAGTGGTACAGACTTCACATT
    GAAGATTTCACGAGTAGAAGCCGAAGACGTGGGTGTTTATTACTGC
    TTCCAAGGATCTCACGTGCCATATACGTTTGGTGGGGGCACAAAAG
    TCGAGATTAAGGGAGGCGGAGGATCAGGAGGTGGGGGAAGTGGAG
    GTGGTGGGTCACAAGTACAGCTCGTGCAATCAGGGGCGGAGGTGAA
    GAAACCAGGGGCGTCTGTGAAGGTAAGCTGTAAGGCATCCGGATTG
    ACAATCGAGGATTATTACATGCATTGGGTCCGCCAGGCACCAGGGC
    AGGGATTGGAGTGGATGGGGTGGATAGATCCTGAAAATGGGGATAC
    AGAGTATGGCCCTAAGTTCCAGGGCAGAGTTACGATGACTCGAGAT
    ACTAGCATTAATACGGCCTACATGGAGCTTAGCCGCCTGCGGTCCG
    ATGACACGGCCGTTTATTATTGCGCCGTACACAATGCGCACTACGGG
    ACATGGTTCGCGTATTGGGGTCAAGGAACGCTCGTTACTGTCTCAAG
    T
    CTX-971 and DVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEWYQQRPGQSP  54
    CTX-971b RPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHV
    scFv PYTFGGGTKVEIK GGGGSGGGGSGGGGS QVQLVQSGAEVKKPGASV
    (linker KVSCKASGLTIEDYYMHWVRQAPGQGLEWMGWIDPENGDTEYGPKF
    underlined) QGRVTMTRDTSINTAYMELSRLRSDDTAVYYCAVHNAHYGTWFAYW
    GQGTLVTVSS
    CTX-971 and QVQLVQSGAEVKKPGASVKVSCKASGLTIE DYYMH WVRQAPGQGLE  55
    CTX-971b WMG WIDPENGDTEYGPKFQG RVTMTRDTSINTAYMELSRLRSDDTA
    scFvVH VYYCAV HNAHYGTWFAY WGQGTLVTVSS
    CDRs- in bold
    CTX-971 and DVVMTQSPLSLPVTLGQPASISC RSSQSLLHSSGNTYLE WYQQRPGQS  56
    CTX-971b PRPLIY KISTRFS GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC FQGS
    scFv VL HVPYT FGGGTKVEIK
    CDRs- in
    bold
    CTX-971 and DYYMH  57
    CTX-971b
    VH CDR1
    CTX-971 and WIDPENGDTEYGPKFQG  58
    CTX-971b
    VH CDR2
    CTX-971 and HNAHYGTWFAY  59
    CTX-971b
    VH CDR3
    CTX-971 and RSSQSLLHSSGNTYLE  60
    CTX-971b
    VL CDR1
    CTX-971 and KISTRFS  61
    CTX-971b
    VL CDR2
    CTX-971 and FQGSHVPYT  62
    CTX-971b
    VL CDR3
    CTX-971 GAGATGTAAGGAGCTGCTGTGACTTGCTCAAGGCCTTATATCGAGT  63
    Donor AAACGGTAGTGCTGGGGCTTAGACGCAGGTGTTCTGATTTATAGTTC
    LHA to RHA AAAACCTCTATCAATGAGAGAGCAATCTCCTGGTAATGTGATAGAT
    TTCCCAACTTAATGCCAACATACCATAAACCTCCCATTCTGCTAATG
    CCCAGCCTAAGTTGGGGAGACCACTCCAGATTCCAAGATGTACAGT
    TTGCTTTGCTGGGCCTTTTTCCCATGCCTGCCTTTACTCTGCCAGAGT
    TATATTGCTGGGGTTTTGAAGAAGATCCTATTAAATAAAAGAATAA
    GCAGTATTATTAAGTAGCCCTGCATTTCAGGTTTCCTTGAGTGGCAG
    GCCAGGCCTGGCCGTGAACGTTCACTGAAATCATGGCCTCTTGGCCA
    AGATTGATAGCTTGTGCCTGTCCCTGAGTCCCAGTCCATCACGAGCA
    GCTGGTTTCTAAGATGCTATTTCCCGTATAAAGCATGAGACCGTGAC
    TTGCCAGCCCCACAGAGCCCCGCCCTTGTCCATCACTGGCATCTGGA
    CTCCAGCCTGGGTTGGGGCAAAGAGGGAAATGAGATCATGTCCTAA
    CCCTGATCCTCTTGTCCCACAGATATCCAGAACCCTGACCCTGCCGT
    GTACCAGCTGAGAGACTCTAAATCCAGTGACAAGTCTGTCTGCCTAT
    TCACCGATTTTGATTCTCAAACAAATGTGTCACAAAGTAAGGATTCT
    GATGTGTATATCACAGACAAAACTGTGCTAGACATGAGGTCTATGG
    ACTTCAggctccggtgcccgtcagtgggcagagcgcacatcgcccac
    agtccccgagaagttggggggaggggtcggcaattgaaccggtgcct
    agagaaggtggcgcggggtaaactgggaaagtgatgtcgtgtactgg
    ctccgcctttttcccgagggtgggggagaaccgtatataagtgcagt
    agtcgccgtgaacgttctttttcgcaacgggtttgccgccagaacac
    aggtaagtgccgtgtgtggttcccgcgggcctggcctctttacgggt
    tatggcccttgcgtgccttgaattacttccactggctgcagtacgat
    tcttgatcccgagcttcgggttggaagtgggtgggagagttgtcgag
    gccttgcgcttaaggagccccttcgcctcgtgcttgagttgaggcct
    ggcctgggcgctggggccgccgcgtgcgaatctggtggcaccttcgc
    gcctgtctcgctgctttcgataagtctctagccatttaaaatttttg
    atgacctgctgcgacgctttttttctggcaagatagtcttgtaaatg
    cgggccaagatctgcacactggtatttcggtttttggggccgcgggc
    ggcgacggggcccgtgcgtcccagcgcacatgttcggcgaggcgggg
    cctgcgagcgcggccaccgagaatcggacgggggtagtctcaagctg
    gccggcctgctctggtgcctggcctcgcgccgccgtgtatcgccccg
    ccctgggcggcaaggctggcccggtcggcaccagttgcgtgagcgga
    aagatggccgcttcccggccctgctgcagggagctcaaaatggagga
    cgcggcgctcgggagagcgggcgggtgagtcacccacacaaaggaaa
    agggcctttccgtcctcagccgtcgcttcatgtgactccacggagtac
    cgggcgccgtccaggcacctcgattagttctcgagcttttggagtacg
    tcgtctttaggttggggggaggggttttatgcgatggagtttccccac
    actgagtgggtggagactgaagttaggccagcttggcacttgatgtaa
    ttctccttggaatttgccctttttgagtttggatcttggttcattctc
    aagcctcagacagtggttcaaagtttttttcttccatttcaggtgtcg
    tgaCCACCATGGCGCTTCCGGTGACAGCACTGCTCCTCCCCTTGGCGC
    TGTTGCTCCAC
    GCAGCAAGGCCGGACGTGGTCATGACTCAAAGCCCACTTTCCTTGC
    CCGTGACTCTCGGACAACCGGCTTCAATATCTTGCCGCTCATCACAG
    TCCCTGCTGCATAGCAGTGGTAACACTTATCTTGAGTGGTACCAACA
    GCGGCCCGGCCAATCTCCTAGGCCCCTGATATATAAGATAAGTACTC
    GCTTTTCCGGGGTCCCGGACCGGTTCAGCGGGTCTGGGAGTGGTAC
    AGACTTCACATTGAAGATTTCACGAGTAGAAGCCGAAGACGTGGGT
    GTTTATTACTGCTTCCAAGGATCTCACGTGCCATATACGTTTGGTGG
    GGGCACAAAAGTCGAGATTAAGGGAGGCGGAGGATCAGGAGGTGG
    GGGAAGTGGAGGTGGTGGGTCACAAGTACAGCTCGTGCAATCAGGG
    GCGGAGGTGAAGAAACCAGGGGCGTCTGTGAAGGTAAGCTGTAAG
    GCATCCGGATTGACAATCGAGGATTATTACATGCATTGGGTCCGCCA
    GGCACCAGGGCAGGGATTGGAGTGGATGGGGTGGATAGATCCTGAA
    AATGGGGATACAGAGTATGGCCCTAAGTTCCAGGGCAGAGTTACGA
    TGACTCGAGATACTAGCATTAATACGGCCTACATGGAGCTTAGCCG
    CCTGCGGTCCGATGACACGGCCGTTTATTATTGCGCCGTACACAATG
    CGCACTACGGGACATGGTTCGCGTATTGGGGTCAAGGAACGCTCGT
    TACTGTCTCAAGTAGTGCTGCTGCCTTTGTCCCGGTATTTCTCCCAGC
    CAAACCGACCACGACTCCCGCCCCGCGCCCTCCGACACCCGCTCCC
    ACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAGGCATGCCGACC
    CGCCGCCGGGGGTGCTGTTCATACGAGGGGCTTGGACTTCGCTTGTG
    ATATTTACATTTGGGCTCCGTTGGCGGGTACGTGCGGCGTCCTTTTG
    TTGTCACTCGTTATTACTTTGTATTGTAATCACAGGAATCGCTCAAA
    GCGGAGTAGGTTGTTGCATTCCGATTACATGAATATGACTCCTCGCC
    GGCCTGGGCCGACAAGAAAACATTACCAACCCTATGCCCCCCCACG
    AGACTTCGCTGCGTACAGGTCCCGAGTGAAGTTTTCCCGAAGCGCA
    GACGCTCCGGCATATCAGCAAGGACAGAATCAGCTGTATAACGAAC
    TGAATTTGGGACGCCGCGAGGAGTATGACGTGCTTGATAAACGCCG
    GGGGAGAGACCCGGAAATGGGGGGTAAACCCCGAAGAAAGAATCC
    CCAAGAAGGACTCTACAATGAACTCCAGAAGGATAAGATGGCGGA
    GGCCTACTCAGAAATAGGTATGAAGGGCGAACGACGACGGGGAAA
    AGGTCACGATGGCCTCTACCAAGGGTTGAGTACGGCAACCAAAGAT
    ACGTACGATGCACTGCATATGCAGGCCCTGCCTCCCAGATAATAAT
    AAAATCGCTATCCATCGAAGATGGATGTGTGTTGGTTTTTTGTGTGT
    GGAGCAACAAATCTGACTTTGCATGTGCAAACGCCTTCAACAACAG
    CATTATTCCAGAAGACACCTTCTTCCCCAGCCCAGGTAAGGGCAGCT
    TTGGTGCCTTCGCAGGCTGTTTCCTTGCTTCAGGAATGGCCAGGTTC
    TGCCCAGAGCTCTGGTCAATGATGTCTAAAACTCCTCTGATTGGTGG
    TCTCGGCCTTATCCATTGCCACCAAAACCCTCTTTTTACTAAGAAAC
    AGTGAGCCTTGTTCTGGCAGTCCAGAGAATGACACGGGAAAAAAGC
    AGATGAAGAGAAGGTGGCAGGAGAGGGCACGTGGCCCAGCCTCAG
    TCTCTCCAACTGAGTTCCTGCCTGCCTGCCTTTGCTCAGACTGTTTGC
    CCCTTACTGCTCTTCTAGGCCTCATTCTAAGCCCCTTCTCCAAGTTGC
    CTCTCCTTATTTCTCCCTGTCTGCCAAAAAATCTTTCCCAGCTCACTA
    AGTCAGTCTCACGCAGTCACTCATTAACCCACCAATCACTGATTGTG
    CCGGCACATGAATGCACCAGGTGTTGAAGTGGAGGAATTAAAAAGT
    CAGATGAGGGGTGTGCCCAGAGGAAGCACCATTCTAGTTGGGGGAG
    CCCATCTGTCAGCTGGGAAAAGTCCAAATAACTTCAGATTGGAATG
    TGTTTTAACTCAGGGTTGAGAAAACAGCTACCTTCAGGACAAAAGT
    CAGGGAAGGGCTCTCTGAAGAAATGCTACTTGAAGATACCAGCCCT
    ACCAAGGGCAGGGAGAGGACCCTATAGAGGCCTGGGACAGGAGCT
    CAATGAGAAAGG
    CTX-971b GAGATGTAAGGAGCTGCTGTGACTTGCTCAAGGCCTTATATCGAGT  64
    Donor AAACGGTAGTGCTGGGGCTTAGACGCAGGTGTTCTGATTTATAGTTC
    LHA to RHA AAAACCTCTATCAATGAGAGAGCAATCTCCTGGTAATGTGATAGAT
    TTCCCAACTTAATGCCAACATACCATAAACCTCCCATTCTGCTAATG
    CCCAGCCTAAGTTGGGGAGACCACTCCAGATTCCAAGATGTACAGT
    TTGCTTTGCTGGGCCTTTTTCCCATGCCTGCCTTTACTCTGCCAGAGT
    TATATTGCTGGGGTTTTGAAGAAGATCCTATTAAATAAAAGAATAA
    GCAGTATTATTAAGTAGCCCTGCATTTCAGGTTTCCTTGAGTGGCAG
    GCCAGGCCTGGCCGTGAACGTTCACTGAAATCATGGCCTCTTGGCCA
    AGATTGATAGCTTGTGCCTGTCCCTGAGTCCCAGTCCATCACGAGCA
    GCTGGTTTCTAAGATGCTATTTCCCGTATAAAGCATGAGACCGTGAC
    TTGCCAGCCCCACAGAGCCCCGCCCTTGTCCATCACTGGCATCTGGA
    CTCCAGCCTGGGTTGGGGCAAAGAGGGAAATGAGATCATGTCCTAA
    CCCTGATCCTCTTGTCCCACAGATATCCAGAACCCTGACCCTGCCGT
    GTACCAGCTGAGAGACTCTAAATCCAGTGACAAGTCTGTCTGCCTAT
    TCACCGATTTTGATTCTCAAACAAATGTGTCACAAAGTAAGGATTCT
    GATGTGTATATCACAGACAAAACTGTGCTAGACATGAGGTCTATGG
    ACTTCAggctccggtgcccgtcagtgggcagagcgcacatcgcccacag
    tccccgagaagttggggggaggggtcggcaattgaaccggtgcctagag
    aaggtggcgcggggtaaactgggaaagtgatgtcgtgtactggctccgc
    ctttttcccgagggtgggggagaaccgtatataagtgcagtagtcgccg
    tgaacgttctttttcgcaacgggtttgccgccagaacacaggtaagtgc
    cgtgtgtggttcccgcgggcctggcctctttacgggttatggcccttgc
    gtgccttgaattacttccactggctgcagtacgtgattcttgatcccga
    gcttcgggttggaagtgggtgggagagttcgaggccttgcgcttaagga
    gccccttcgcctcgtgcttgagttgaggcctggcctgggcgctggggcc
    gccgcgtgcgaatctggtggcaccttcgcgcctgtctcgctgctttcga
    taagtctctagccatttaaaatttttgatgacctgctgcgacgctttt
    tttctggcaagatagtcttgtaaatgcgggccaagatctgcacactggt
    atttcggtttttggggccgcgggcggcgacggggcccgtgcgtcccag
    cgcacatgttcggcgaggcggggcctgcgagcgcggccaccgagaatc
    ggacgggggtagtctcaagctggccggcctgctctggtgcctggcctc
    gcgccgccgtgtatcgccccgccctgggcggcaaggctggcccggtcg
    gcaccagttgcgtgagcggaaagatggccgcttcccggccctgctgca
    gggagctcaaaatggaggacgcggcgctcgggagagcgggcgggtgag
    tcacccacacaaaggaaaagggcctttccgtcctcagccgtcgcttca
    tgtgactccacggagtaccgggcgccgtccaggcacctcgattagttc
    tcgagcttttggagtacgtcgtctttaggttggggggaggggttttat
    gcgatggagtttccccacactgagtgggtggagactgaagttaggcca
    gcttggcacttgatgtaattctccttggaatttgccctttttgagttt
    ggatcttggttcattctcaagcctcagacagtggttcaaagttttt
    ttcttccatttcaggtgtcgtgaCCACCATGG
    CGCTTCCGGTGACAGCACTGCTCCTCCCCTTGGCGCTGTTGCTCCAC
    GCAGCAAGGCCGGACGTGGTCATGACTCAAAGCCCACTTTCCTTGC
    CCGTGACTCTCGGACAACCGGCTTCAATATCTTGCCGCTCATCACAG
    TCCCTGCTGCATAGCAGTGGTAACACTTATCTTGAGTGGTACCAACA
    GCGGCCCGGCCAATCTCCTAGGCCCCTGATATATAAGATAAGTACTC
    GCTTTTCCGGGGTCCCGGACCGGTTCAGCGGGTCTGGGAGTGGTAC
    AGACTTCACATTGAAGATTTCACGAGTAGAAGCCGAAGACGTGGGT
    GTTTATTACTGCTTCCAAGGATCTCACGTGCCATATACGTTTGGTGG
    GGGCACAAAAGTCGAGATTAAGGGAGGCGGAGGATCAGGAGGTGG
    GGGAAGTGGAGGTGGTGGGTCACAAGTACAGCTCGTGCAATCAGGG
    GCGGAGGTGAAGAAACCAGGGGCGTCTGTGAAGGTAAGCTGTAAG
    GCATCCGGATTGACAATCGAGGATTATTACATGCATTGGGTCCGCCA
    GGCACCAGGGCAGGGATTGGAGTGGATGGGGTGGATAGATCCTGAA
    AATGGGGATACAGAGTATGGCCCTAAGTTCCAGGGCAGAGTTACGA
    TGACTCGAGATACTAGCATTAATACGGCCTACATGGAGCTTAGCCG
    CCTGCGGTCCGATGACACGGCCGTTTATTATTGCGCCGTACACAATG
    CGCACTACGGGACATGGTTCGCGTATTGGGGTCAAGGAACGCTCGT
    TACTGTCTCAAGTAGTGCTGCTGCCTTTGTCCCGGTATTTCTCCCAGC
    CAAACCGACCACGACTCCCGCCCCGCGCCCTCCGACACCCGCTCCC
    ACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAGGCATGCCGACC
    CGCCGCCGGGGGTGCTGTTCATACGAGGGGCTTGGACTTCGCTTGTG
    ATATTTACATTTGGGCTCCGTTGGCGGGTACGTGCGGCGTCCTTTTG
    TTGTCACTCGTTATTACTTTGTATTGTAATCACAGGAATCGCAAACG
    GGGCAGAAAGAAACTCCTGTATATATTCAAACAACCATTTATGAGA
    CCAGTACAAACTACTCAAGAGGAAGATGGCTGTAGCTGCCGATTTC
    CAGAAGAAGAAGAAGGAGGATGTGAACTGCGAGTGAAGTTTTCCCG
    AAGCGCAGACGCTCCGGCATATCAGCAAGGACAGAATCAGCTGTAT
    AACGAACTGAATTTGGGACGCCGCGAGGAGTATGACGTGCTTGATA
    AACGCCGGGGGAGAGACCCGGAAATGGGGGGTAAACCCCGAAGAA
    AGAATCCCCAAGAAGGACTCTACAATGAACTCCAGAAGGATAAGAT
    GGCGGAGGCCTACTCAGAAATAGGTATGAAGGGCGAACGACGACG
    GGGAAAAGGTCACGATGGCCTCTACCAAGGGTTGAGTACGGCAACC
    AAAGATACGTACGATGCACTGCATATGCAGGCCCTGCCTCCCAGAT
    AATAATAAAATCGCTATCCATCGAAGATGGATGTGTGTTGGTTTTTT
    GTGTGTGGAGCAACAAATCTGACTTTGCATGTGCAAACGCCTTCAAC
    AACAGCATTATTCCAGAAGACACCTTCTTCCCCAGCCCAGGTAAGG
    GCAGCTTTGGTGCCTTCGCAGGCTGTTTCCTTGCTTCAGGAATGGCC
    AGGTTCTGCCCAGAGCTCTGGTCAATGATGTCTAAAACTCCTCTGAT
    TGGTGGTCTCGGCCTTATCCATTGCCACCAAAACCCTCTTTTTACTA
    AGAAACAGTGAGCCTTGTTCTGGCAGTCCAGAGAATGACACGGGAA
    AAAAGCAGATGAAGAGAAGGTGGCAGGAGAGGGCACGTGGCCCAG
    CCTCAGTCTCTCCAACTGAGTTCCTGCCTGCCTGCCTTTGCTCAGACT
    GTTTGCCCCTTACTGCTCTTCTAGGCCTCATTCTAAGCCCCTTCTCCA
    AGTTGCCTCTCCTTATTTCTCCCTGTCTGCCAAAAAATCTTTCCCAGC
    TCACTAAGTCAGTCTCACGCAGTCACTCATTAACCCACCAATCACTG
    ATTGTGCCGGCACATGAATGCACCAGGTGTTGAAGTGGAGGAATTA
    AAAAGTCAGATGAGGGGTGTGCCCAGAGGAAGCACCATTCTAGTTG
    GGGGAGCCCATCTGTCAGCTGGGAAAAGTCCAAATAACTTCAGATT
    GGAATGTGTTTTAACTCAGGGTTGAGAAAACAGCTACCTTCAGGAC
    AAAAGTCAGGGAAGGGCTCTCTGAAGAAATGCTACTTGAAGATACC
    AGCCCTACCAAGGGCAGGGAGAGGACCCTATAGAGGCCTGGGACA
    GGAGCTCAATGAGAAAGG
    CTX-972 CCACCATGGCGCTTCCGGTGACAGCACTGCTCCTCCCCTTGGCGCTG  65
    CAR TTGCTCCACGCAGCAAGGCCGCAAGTTCAACTGGTCCAGTCAGGCG
    CD28 co-stim CTGAGGTCAAAAAGCCCGGCGCGAGCGTAAAAGTCTCCTGCAAGGC
    GTCAGGGTTGACGATAGAAGATTATTACATGCATTGGGTCAGACAG
    GCACCCGGACAGGGATTGGAGTGGATGGGTTGGATCGACCCGGAAA
    ACGGTGACACGGAGTATGGGCCGAAGTTTCAGGGGAGGGTCACAAT
    GACACGAGATACGTCCATAAATACCGCTTACATGGAACTTTCTCGGC
    TTCGCTCTGATGATACAGCAGTTTACTACTGCGCTGTTCATAATGCC
    CATTACGGAACCTGGTTCGCGTACTGGGGCCAAGGGACCCTGGTTA
    CGGTTAGCTCTGGTGGGGGTGGAAGCGGGGGAGGGGGTAGCGGAG
    GTGGCGGAAGTGATGTTGTTATGACACAGAGTCCCCTGTCATTGCCC
    GTCACCCTCGGACAACCAGCTAGCATTTCATGCAGGTCTAGTCAAA
    GCCTCCTTCACAGTAGCGGCAACACCTACCTCGAATGGTATCAACA
    ACGGCCAGGGCAATCTCCTCGCCCACTCATATACAAAATCTCTACAC
    GCTTCTCAGGTGTTCCCGACCGCTTCAGCGGTTCCGGCTCTGGGACA
    GACTTTACCTTGAAAATAAGCAGGGTTGAAGCTGAGGACGTAGGGG
    TATATTATTGTTTTCAGGGCAGTCACGTGCCGTACACTGGGGGCGGA
    ACCAAAGTCGAGATAAAGAGTGCTGCTGCCTTTGTCCCGGTATTTCT
    CCCAGCCAAACCGACCACGACTCCCGCCCCGCGCCCTCCGACACCC
    GCTCCCACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAGGCATG
    CCGACCCGCCGCCGGGGGTGCTGTTCATACGAGGGGCTTGGACTTC
    GCTTGTGATATTTACATTTGGGCTCCGTTGGCGGGTACGTGCGGCGT
    CCTTTTGTTGTCACTCGTTATTACTTTGTATTGTAATCACAGGAATCG
    CTCAAAGCGGAGTAGGTTGTTGCATTCCGATTACATGAATATGACTC
    CTCGCCGGCCTGGGCCGACAAGAAAACATTACCAACCCTATGCCCC
    CCCACGAGACTTCGCTGCGTACAGGTCCCGAGTGAAGTTTTCCCGAA
    GCGCAGACGCTCCGGCATATCAGCAAGGACAGAATCAGCTGTATAA
    CGAACTGAATTTGGGACGCCGCGAGGAGTATGACGTGCTTGATAAA
    CGCCGGGGGAGAGACCCGGAAATGGGGGGTAAACCCCGAAGAAAG
    AATCCCCAAGAAGGACTCTACAATGAACTCCAGAAGGATAAGATGG
    CGGAGGCCTACTCAGAAATAGGTATGAAGGGCGAACGACGACGGG
    GAAAAGGTCACGATGGCCTCTACCAAGGGTTGAGTACGGCAACCAA
    AGATACGTACGATGCACTGCATATGCAGGCCCTGCCTCCCAGATAA
    T
    CTX-972 MALPVTALLLPLALLLHAARPQVQLVQSGAEVKKPGASVKVSCKASG 125
    CAR LTIEDYYMHWVRQAPGQGLEWMGWIDPENGDTEYGPKFQGRVTMTR
    CD28 co-stim DTSINTAYMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQGTLVTV
    SSGGGGSGGGGSGGGGSDVVMTQSPLSLPVTLGQPASISCRSSQSLLHS
    SGNTYLEWYQQRPGQSPRPLIYKISTRFSGVPDRFSGSGSGTDFTLKISR
    VEAEDVGVYYCFQGSHVPYTGGGTKVEIKSAAAFVPVFLPAKPTTTPA
    PRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGT
    CGVLLLSLVITLYCNHRNRSKRSRLLHSDYMNMTPRRPGPTRKHYQPY
    APPRDFAAYRSRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLD
    KRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRR
    GKGHDGLYQGLSTATKDTYDALHMQALPPR
    CTX-972b CCACCATGGCGCTTCCGGTGACAGCACTGCTCCTCCCCTTGGCGCTG  67
    CAR TTGCTCCACGCAGCAAGGCCGCAAGTTCAACTGGTCCAGTCAGGCG
    41BBco-stim CTGAGGTCAAAAAGCCCGGCGCGAGCGTAAAAGTCTCCTGCAAGGC
    GTCAGGGTTGACGATAGAAGATTATTACATGCATTGGGTCAGACAG
    GCACCCGGACAGGGATTGGAGTGGATGGGTTGGATCGACCCGGAAA
    ACGGTGACACGGAGTATGGGCCGAAGTTTCAGGGGAGGGTCACAAT
    GACACGAGATACGTCCATAAATACCGCTTACATGGAACTTTCTCGGC
    TTCGCTCTGATGATACAGCAGTTTACTACTGCGCTGTTCATAATGCC
    CATTACGGAACCTGGTTCGCGTACTGGGGCCAAGGGACCCTGGTTA
    CGGTTAGCTCTGGTGGGGGTGGAAGCGGGGGAGGGGGTAGCGGAG
    GTGGCGGAAGTGATGTTGTTATGACACAGAGTCCCCTGTCATTGCCC
    GTCACCCTCGGACAACCAGCTAGCATTTCATGCAGGTCTAGTCAAA
    GCCTCCTTCACAGTAGCGGCAACACCTACCTCGAATGGTATCAACA
    ACGGCCAGGGCAATCTCCTCGCCCACTCATATACAAAATCTCTACAC
    GCTTCTCAGGTGTTCCCGACCGCTTCAGCGGTTCCGGCTCTGGGACA
    GACTTTACCTTGAAAATAAGCAGGGTTGAAGCTGAGGACGTAGGGG
    TATATTATTGTTTTCAGGGCAGTCACGTGCCGTACACTGGGGGCGGA
    ACCAAAGTCGAGATAAAGAGTGCTGCTGCCTTTGTCCCGGTATTTCT
    CCCAGCCAAACCGACCACGACTCCCGCCCCGCGCCCTCCGACACCC
    GCTCCCACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAGGCATG
    CCGACCCGCCGCCGGGGGTGCTGTTCATACGAGGGGCTTGGACTTC
    GCTTGTGATATTTACATTTGGGCTCCGTTGGCGGGTACGTGCGGCGT
    CCTTTTGTTGTCACTCGTTATTACTTTGTATTGTAATCACAGGAATCG
    CAAACGGGGCAGAAAGAAACTCCTGTATATATTCAAACAACCATTT
    ATGAGACCAGTACAAACTACTCAAGAGGAAGATGGCTGTAGCTGCC
    GATTTCCAGAAGAAGAAGAAGGAGGATGTGAACTGCGAGTGAAGTT
    TTCCCGAAGCGCAGACGCTCCGGCATATCAGCAAGGACAGAATCAG
    CTGTATAACGAACTGAATTTGGGACGCCGCGAGGAGTATGACGTGC
    TTGATAAACGCCGGGGGAGAGACCCGGAAATGGGGGGTAAACCCC
    GAAGAAAGAATCCCCAAGAAGGACTCTACAATGAACTCCAGAAGG
    ATAAGATGGCGGAGGCCTACTCAGAAATAGGTATGAAGGGCGAAC
    GACGACGGGGAAAAGGTCACGATGGCCTCTACCAAGGGTTGAGTAC
    GGCAACCAAAGATACGTACGATGCACTGCATATGCAGGCCCTGCCT
    CCCAGATAAT
    CTX-972b MALPVTALLLPLALLLHAARPQVQLVQSGAEVKKPGASVKVSCKASG 126
    CAR LTIEDYYMHWVRQAPGQGLEWMGWIDPENGDTEYGPKFQGRVTMTR
    41BB co-stim DTSINTAYMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQGTLVTV
    SSGGGGSGGGGSGGGGSDVVMTQSPLSLPVTLGQPASISCRSSQSLLHS
    SGNTYLEWYQQRPGQSPRPLIYKISTRFSGVPDRFSGSGSGTDFTLKISR
    VEAEDVGVYYCFQGSHVPYTGGGTKVEIKSAAAFVPVFLPAKPTTTPA
    PRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGT
    CGVLLLSLVITLYCNHRNRKRGRKKLLYIFKQPFMRPVQTTQEEDGCSC
    RFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLD
    KRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRR
    GKGHDGLYQGLSTATKDTYDALHMQALPPR
    CTX-972 and CAAGTTCAACTGGTCCAGTCAGGCGCTGAGGTCAAAAAGCCCGGCG  69
    CTX-972b CGAGCGTAAAAGTCTCCTGCAAGGCGTCAGGGTTGACGATAGAAGA
    scFv TTATTACATGCATTGGGTCAGACAGGCACCCGGACAGGGATTGGAG
    TGGATGGGTTGGATCGACCCGGAAAACGGTGACACGGAGTATGGGC
    CGAAGTTTCAGGGGAGGGTCACAATGACACGAGATACGTCCATAAA
    TACCGCTTACATGGAACTTTCTCGGCTTCGCTCTGATGATACAGCAG
    TTTACTACTGCGCTGTTCATAATGCCCATTACGGAACCTGGTTCGCG
    TACTGGGGCCAAGGGACCCTGGTTACGGTTAGCTCTGGTGGGGGTG
    GAAGCGGGGGAGGGGGTAGCGGAGGTGGCGGAAGTGATGTTGTTA
    TGACACAGAGTCCCCTGTCATTGCCCGTCACCCTCGGACAACCAGCT
    AGCATTTCATGCAGGTCTAGTCAAAGCCTCCTTCACAGTAGCGGCAA
    CACCTACCTCGAATGGTATCAACAACGGCCAGGGCAATCTCCTCGC
    CCACTCATATACAAAATCTCTACACGCTTCTCAGGTGTTCCCGACCG
    CTTCAGCGGTTCCGGCTCTGGGACAGACTTTACCTTGAAAATAAGCA
    GGGTTGAAGCTGAGGACGTAGGGGTATATTATTGTTTTCAGGGCAG
    TCACGTGCCGTACACTGGGGGCGGAACCAAAGTCGAGATAAAG
    CTX-972 and QVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMHWVRQAPGQGLE 127
    CTX-972b WMGWIDPENGDTEYGPKFQGRVTMTRDTSINTAYMELSRLRSDDTAV
    scFv YYCAVHNAHYGTWFAYWGQGTLVTVSSGGGGSGGGGSGGGGSDVV
    (linker MTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEWYQQRPGQSPRPLI
    underlined) YKISTRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHVPYT
    GGGTKVEIK
    CTX-972 and QVQLVQSGAEVKKPGASVKVSCKASGLTIE DYYMH WVRQAPGQGLE  55
    CTX-972b WMG WIDPENGDTEYGPKFQG RVTMTRDTSINTAYMELSRLRSDDTA
    scFv VH VYYCAV HNAHYGTWFAY WGQGTLVTVSS
    CDRs- in bold
    CTX-972 and DVVMTQSPLSLPVTLGQPASISC RSSQSLLHSSGNTYLE WYQQRPGQS 128
    CTX-972b PRPLIY KISTRFS GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC FQGS
    scFv VL HVPYT GGGTKVEIK
    CDRs- in
    bold
    CTX-972 and DYYMH  57
    CTX-972b
    VH CDR1
    CTX-972 and WIDPENGDTEYGPKFQG  58
    CTX-972b
    VH CDR2
    CTX-972 and HNAHYGTWFAY  59
    CTX-972b
    VH CDR3
    CTX-972 and RSSQSLLHSSGNTYLE  60
    CTX-972b
    VL CDR1
    CTX-972 and KISTRFS  61
    CTX-972b
    VL CDR2
    CTX-972 and
    CTX-972b FQGSHVPYT  62
    VL CDR3
    CTX-972 GAGATGTAAGGAGCTGCTGTGACTTGCTCAAGGCCTTATATCGAGT  71
    Donor AAACGGTAGTGCTGGGGCTTAGACGCAGGTGTTCTGATTTATAGTTC
    LHA to RHA AAAACCTCTATCAATGAGAGAGCAATCTCCTGGTAATGTGATAGAT
    TTCCCAACTTAATGCCAACATACCATAAACCTCCCATTCTGCTAATG
    CCCAGCCTAAGTTGGGGAGACCACTCCAGATTCCAAGATGTACAGT
    TTGCTTTGCTGGGCCTTTTTCCCATGCCTGCCTTTACTCTGCCAGAGT
    TATATTGCTGGGGTTTTGAAGAAGATCCTATTAAATAAAAGAATAA
    GCAGTATTATTAAGTAGCCCTGCATTTCAGGTTTCCTTGAGTGGCAG
    GCCAGGCCTGGCCGTGAACGTTCACTGAAATCATGGCCTCTTGGCCA
    AGATTGATAGCTTGTGCCTGTCCCTGAGTCCCAGTCCATCACGAGCA
    GCTGGTTTCTAAGATGCTATTTCCCGTATAAAGCATGAGACCGTGAC
    TTGCCAGCCCCACAGAGCCCCGCCCTTGTCCATCACTGGCATCTGGA
    CTCCAGCCTGGGTTGGGGCAAAGAGGGAAATGAGATCATGTCCTAA
    CCCTGATCCTCTTGTCCCACAGATATCCAGAACCCTGACCCTGCCGT
    GTACCAGCTGAGAGACTCTAAATCCAGTGACAAGTCTGTCTGCCTAT
    TCACCGATTTTGATTCTCAAACAAATGTGTCACAAAGTAAGGATTCT
    GATGTGTATATCACAGACAAAACTGTGCTAGACATGAGGTCTATGG
    ACTTCAggctccggtgcccgtcagtgggcagagcgcacatcgcccac
    agtccccgagaagttggggggaggggtcggcaattgaaccggtgcct
    agagaaggtggcgcggggtaaactgggaaagtgatgtcgtgtactggc
    tccgcctttttcccgagggtgggggagaaccgtatataagtgcagtag
    tcgccgtgaacgttctttttcgcaacgggtttgccgccagaacacagg
    taagtgccgtgtgtggttcccgcgggcctggcctctttacgggttatg
    gcccttgcgtgccttgaattacttccactggctgcagtacgtgattct
    tgatcccgagcttcgggttggaagtgggtgggagagttcgaggccttgc
    gcttaaggagccccttcgcctcgtgcttgagttgaggcctggcctgggc
    gctggggccgccgcgtgcgaatctggtggcaccttcgcgcctgtctc
    gctgctttcgataagtctctagccatttaaaatttttgatgacct
    gctgcgacgctttttttctggcaagatagtcttgtaaatgcgggccaa
    gatctgcacactggtatttcggtttttggggccgcgggcggcgacggg
    gcccgtgcgtcccagcgcacatgttcggcgaggcggggcctgcgagcg
    cggccaccgagaatcggacgggggtagtctcaagctggccggc
    ctgctctggtgcctggcctcgcgccgccgtgtatcgccccgccctg
    ggcggcaaggctggcccggtcggcaccagttgcgtgagcggaaagat
    ggccgcttcccggccctgctgcagggagctcaaaatggaggacgcgg
    cgctcgggagagcgggcgggtgagtcacccacacaaaggaaaagg
    gcctttccgtcctcagccgtcgcttcatgtgactccacggagtaccg
    ggcgccgtccaggcacctcgattagttctcgagcttttggagtacgtc
    gtctttaggttggggggaggggttttatgcgatggagtttccccacact
    gagtgggtggagactgaagttaggccagcttggcacttgatgtaattct
    ccttggaatttgccctttttgagtttggatcttggttcattctcaagcc
    tcagacagtggttcaaagtttttttcttccatttcaggtgtcgtga
    CCACCATGG
    CGCTTCCGGTGACAGCACTGCTCCTCCCCTTGGCGCTGTTGCTCCAC
    GCAGCAAGGCCGCAAGTTCAACTGGTCCAGTCAGGCGCTGAGGTCA
    AAAAGCCCGGCGCGAGCGTAAAAGTCTCCTGCAAGGCGTCAGGGTT
    GACGATAGAAGATTATTACATGCATTGGGTCAGACAGGCACCCGGA
    CAGGGATTGGAGTGGATGGGTTGGATCGACCCGGAAAACGGTGACA
    CGGAGTATGGGCCGAAGTTTCAGGGGAGGGTCACAATGACACGAGA
    TACGTCCATAAATACCGCTTACATGGAACTTTCTCGGCTTCGCTCTG
    ATGATACAGCAGTTTACTACTGCGCTGTTCATAATGCCCATTACGGA
    ACCTGGTTCGCGTACTGGGGCCAAGGGACCCTGGTTACGGTTAGCTC
    TGGTGGGGGTGGAAGCGGGGGAGGGGGTAGCGGAGGTGGCGGAAG
    TGATGTTGTTATGACACAGAGTCCCCTGTCATTGCCCGTCACCCTCG
    GACAACCAGCTAGCATTTCATGCAGGTCTAGTCAAAGCCTCCTTCAC
    AGTAGCGGCAACACCTACCTCGAATGGTATCAACAACGGCCAGGGC
    AATCTCCTCGCCCACTCATATACAAAATCTCTACACGCTTCTCAGGT
    GTTCCCGACCGCTTCAGCGGTTCCGGCTCTGGGACAGACTTTACCTT
    GAAAATAAGCAGGGTTGAAGCTGAGGACGTAGGGGTATATTATTGT
    TTTCAGGGCAGTCACGTGCCGTACACTGGGGGCGGAACCAAAGTCG
    AGATAAAGAGTGCTGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAAA
    CCGACCACGACTCCCGCCCCGCGCCCTCCGACACCCGCTCCCACCAT
    CGCCTCTCAACCTCTTAGTCTTCGCCCCGAGGCATGCCGACCCGCCG
    CCGGGGGTGCTGTTCATACGAGGGGCTTGGACTTCGCTTGTGATATT
    TACATTTGGGCTCCGTTGGCGGGTACGTGCGGCGTCCTTTTGTTGTC
    ACTCGTTATTACTTTGTATTGTAATCACAGGAATCGCTCAAAGCGGA
    GTAGGTTGTTGCATTCCGATTACATGAATATGACTCCTCGCCGGCCT
    GGGCCGACAAGAAAACATTACCAACCCTATGCCCCCCCACGAGACT
    TCGCTGCGTACAGGTCCCGAGTGAAGTTTTCCCGAAGCGCAGACGC
    TCCGGCATATCAGCAAGGACAGAATCAGCTGTATAACGAACTGAAT
    TTGGGACGCCGCGAGGAGTATGACGTGCTTGATAAACGCCGGGGGA
    GAGACCCGGAAATGGGGGGTAAACCCCGAAGAAAGAATCCCCAAG
    AAGGACTCTACAATGAACTCCAGAAGGATAAGATGGCGGAGGCCTA
    CTCAGAAATAGGTATGAAGGGCGAACGACGACGGGGAAAAGGTCA
    CGATGGCCTCTACCAAGGGTTGAGTACGGCAACCAAAGATACGTAC
    GATGCACTGCATATGCAGGCCCTGCCTCCCAGATAATAATAAAATC
    GCTATCCATCGAAGATGGATGTGTGTTGGTTTTTTGTGTGTGGAGCA
    ACAAATCTGACTTTGCATGTGCAAACGCCTTCAACAACAGCATTATT
    CCAGAAGACACCTTCTTCCCCAGCCCAGGTAAGGGCAGCTTTGGTG
    CCTTCGCAGGCTGTTTCCTTGCTTCAGGAATGGCCAGGTTCTGCCCA
    GAGCTCTGGTCAATGATGTCTAAAACTCCTCTGATTGGTGGTCTCGG
    CCTTATCCATTGCCACCAAAACCCTCTTTTTACTAAGAAACAGTGAG
    CCTTGTTCTGGCAGTCCAGAGAATGACACGGGAAAAAAGCAGATGA
    AGAGAAGGTGGCAGGAGAGGGCACGTGGCCCAGCCTCAGTCTCTCC
    AACTGAGTTCCTGCCTGCCTGCCTTTGCTCAGACTGTTTGCCCCTTAC
    TGCTCTTCTAGGCCTCATTCTAAGCCCCTTCTCCAAGTTGCCTCTCCT
    TATTTCTCCCTGTCTGCCAAAAAATCTTTCCCAGCTCACTAAGTCAG
    TCTCACGCAGTCACTCATTAACCCACCAATCACTGATTGTGCCGGCA
    CATGAATGCACCAGGTGTTGAAGTGGAGGAATTAAAAAGTCAGATG
    AGGGGTGTGCCCAGAGGAAGCACCATTCTAGTTGGGGGAGCCCATC
    TGTCAGCTGGGAAAAGTCCAAATAACTTCAGATTGGAATGTGTTTTA
    ACTCAGGGTTGAGAAAACAGCTACCTTCAGGACAAAAGTCAGGGAA
    GGGCTCTCTGAAGAAATGCTACTTGAAGATACCAGCCCTACCAAGG
    GCAGGGAGAGGACCCTATAGAGGCCTGGGACAGGAGCTCAATGAG
    AAAGG
    CTX-972b GAGATGTAAGGAGCTGCTGTGACTTGCTCAAGGCCTTATATCGAGT  72
    Donor AAACGGTAGTGCTGGGGCTTAGACGCAGGTGTTCTGATTTATAGTTC
    LHA to RHA AAAACCTCTATCAATGAGAGAGCAATCTCCTGGTAATGTGATAGAT
    TTCCCAACTTAATGCCAACATACCATAAACCTCCCATTCTGCTAATG
    CCCAGCCTAAGTTGGGGAGACCACTCCAGATTCCAAGATGTACAGT
    TTGCTTTGCTGGGCCTTTTTCCCATGCCTGCCTTTACTCTGCCAGAGT
    TATATTGCTGGGGTTTTGAAGAAGATCCTATTAAATAAAAGAATAA
    GCAGTATTATTAAGTAGCCCTGCATTTCAGGTTTCCTTGAGTGGCAG
    GCCAGGCCTGGCCGTGAACGTTCACTGAAATCATGGCCTCTTGGCCA
    AGATTGATAGCTTGTGCCTGTCCCTGAGTCCCAGTCCATCACGAGCA
    GCTGGTTTCTAAGATGCTATTTCCCGTATAAAGCATGAGACCGTGAC
    TTGCCAGCCCCACAGAGCCCCGCCCTTGTCCATCACTGGCATCTGGA
    CTCCAGCCTGGGTTGGGGCAAAGAGGGAAATGAGATCATGTCCTAA
    CCCTGATCCTCTTGTCCCACAGATATCCAGAACCCTGACCCTGCCGT
    GTACCAGCTGAGAGACTCTAAATCCAGTGACAAGTCTGTCTGCCTAT
    TCACCGATTTTGATTCTCAAACAAATGTGTCACAAAGTAAGGATTCT
    GATGTGTATATCACAGACAAAACTGTGCTAGACATGAGGTCTATGG
    ACTTCAggctccggtgcccgtcagtgggcagagcgcacatcgcccacag
    tccccgagaagttggggggaggggtcggcaattgaaccggtgccta
    gagaaggtggcgcggggtaaactgggaaagtgatgtcgtgtactggc
    tccgcctttttcccgagggtgggggagaaccgtatataagtgcagtagt
    cgccgtgaacgttctttttcgcaacgggtttgccgccagaacacaggt
    aagtgccgtgtgtggttcccgcgggcctggcctctttacgggttatgg
    cccttgcgtgccttgaattacttccactggctgcagtacgtgattcttga
    tcccgagcttcgggttggaagtgggtgggagagttcgaggccttgcgct
    taaggagccccttcgcctcgtgcttgagttgaggcctggcctgggcgct
    ggggccgccgcgtgcgaatctggtggcaccttcgcgcctgtctcgctgct
    ttcgataagtctctagccatttaaaatttttgatgacctgctgcgacgct
    ttttttctggcaagatagtcttgtaaatgcgggccaagatctgcacactg
    gtatttcggtttttggggccgcgggcggcgacggggcccgtgcgtccca
    gcgcacatgttcggcgaggcggggcctgcgagcgcggccaccgagaatcg
    gacgggggtagtctcaagctggccggcctgctctggtgcctggcctcgcg
    ccgccgtgtatcgccccgccctgggcggcaaggctggcccggtcggc
    accagttgcgtgagcggaaagatggccgcttcccggccctgctgcag
    ggagctcaaaatggaggacgcggcgctcgggagagcgggcgggtgagtca
    cccacacaaaggaaaagggcctttccgtcctcagccgtcgcttcatgtgac
    tccacggagtaccgggcgccgtccaggcacctcgattagttctcgagc
    ttttggagtacgtcgtctttaggttggggggaggggttttatgcgatgg
    agtttccccacactgagtgggtggagactgaagttaggccagct
    tggcacttgatgtaattctccttggaatttgccctttttgagtttggat
    cttggttcattctcaagcctcagacagtggttcaaagtttttttcttcc
    atttcaggtgtcgtgaCCACCATGG
    CGCTTCCGGTGACAGCACTGCTCCTCCCCTTGGCGCTGTTGCTCCAC
    GCAGCAAGGCCGCAAGTTCAACTGGTCCAGTCAGGCGCTGAGGTCA
    AAAAGCCCGGCGCGAGCGTAAAAGTCTCCTGCAAGGCGTCAGGGTT
    GACGATAGAAGATTATTACATGCATTGGGTCAGACAGGCACCCGGA
    CAGGGATTGGAGTGGATGGGTTGGATCGACCCGGAAAACGGTGACA
    CGGAGTATGGGCCGAAGTTTCAGGGGAGGGTCACAATGACACGAGA
    TACGTCCATAAATACCGCTTACATGGAACTTTCTCGGCTTCGCTCTG
    ATGATACAGCAGTTTACTACTGCGCTGTTCATAATGCCCATTACGGA
    ACCTGGTTCGCGTACTGGGGCCAAGGGACCCTGGTTACGGTTAGCTC
    TGGTGGGGGTGGAAGCGGGGGAGGGGGTAGCGGAGGTGGCGGAAG
    TGATGTTGTTATGACACAGAGTCCCCTGTCATTGCCCGTCACCCTCG
    GACAACCAGCTAGCATTTCATGCAGGTCTAGTCAAAGCCTCCTTCAC
    AGTAGCGGCAACACCTACCTCGAATGGTATCAACAACGGCCAGGGC
    AATCTCCTCGCCCACTCATATACAAAATCTCTACACGCTTCTCAGGT
    GTTCCCGACCGCTTCAGCGGTTCCGGCTCTGGGACAGACTTTACCTT
    GAAAATAAGCAGGGTTGAAGCTGAGGACGTAGGGGTATATTATTGT
    TTTCAGGGCAGTCACGTGCCGTACACTGGGGGCGGAACCAAAGTCG
    AGATAAAGAGTGCTGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAAA
    CCGACCACGACTCCCGCCCCGCGCCCTCCGACACCCGCTCCCACCAT
    CGCCTCTCAACCTCTTAGTCTTCGCCCCGAGGCATGCCGACCCGCCG
    CCGGGGGTGCTGTTCATACGAGGGGCTTGGACTTCGCTTGTGATATT
    TACATTTGGGCTCCGTTGGCGGGTACGTGCGGCGTCCTTTTGTTGTC
    ACTCGTTATTACTTTGTATTGTAATCACAGGAATCGCAAACGGGGCA
    GAAAGAAACTCCTGTATATATTCAAACAACCATTTATGAGACCAGT
    ACAAACTACTCAAGAGGAAGATGGCTGTAGCTGCCGATTTCCAGAA
    GAAGAAGAAGGAGGATGTGAACTGCGAGTGAAGTTTTCCCGAAGCG
    CAGACGCTCCGGCATATCAGCAAGGACAGAATCAGCTGTATAACGA
    ACTGAATTTGGGACGCCGCGAGGAGTATGACGTGCTTGATAAACGC
    CGGGGGAGAGACCCGGAAATGGGGGGTAAACCCCGAAGAAAGAAT
    CCCCAAGAAGGACTCTACAATGAACTCCAGAAGGATAAGATGGCGG
    AGGCCTACTCAGAAATAGGTATGAAGGGCGAACGACGACGGGGAA
    AAGGTCACGATGGCCTCTACCAAGGGTTGAGTACGGCAACCAAAGA
    TACGTACGATGCACTGCATATGCAGGCCCTGCCTCCCAGATAATAAT
    AAAATCGCTATCCATCGAAGATGGATGTGTGTTGGTTTTTTGTGTGT
    GGAGCAACAAATCTGACTTTGCATGTGCAAACGCCTTCAACAACAG
    CATTATTCCAGAAGACACCTTCTTCCCCAGCCCAGGTAAGGGCAGCT
    TTGGTGCCTTCGCAGGCTGTTTCCTTGCTTCAGGAATGGCCAGGTTC
    TGCCCAGAGCTCTGGTCAATGATGTCTAAAACTCCTCTGATTGGTGG
    TCTCGGCCTTATCCATTGCCACCAAAACCCTCTTTTTACTAAGAAAC
    AGTGAGCCTTGTTCTGGCAGTCCAGAGAATGACACGGGAAAAAAGC
    AGATGAAGAGAAGGTGGCAGGAGAGGGCACGTGGCCCAGCCTCAG
    TCTCTCCAACTGAGTTCCTGCCTGCCTGCCTTTGCTCAGACTGTTTGC
    CCCTTACTGCTCTTCTAGGCCTCATTCTAAGCCCCTTCTCCAAGTTGC
    CTCTCCTTATTTCTCCCTGTCTGCCAAAAAATCTTTCCCAGCTCACTA
    AGTCAGTCTCACGCAGTCACTCATTAACCCACCAATCACTGATTGTG
    CCGGCACATGAATGCACCAGGTGTTGAAGTGGAGGAATTAAAAAGT
    CAGATGAGGGGTGTGCCCAGAGGAAGCACCATTCTAGTTGGGGGAG
    CCCATCTGTCAGCTGGGAAAAGTCCAAATAACTTCAGATTGGAATG
    TGTTTTAACTCAGGGTTGAGAAAACAGCTACCTTCAGGACAAAAGT
    CAGGGAAGGGCTCTCTGAAGAAATGCTACTTGAAGATACCAGCCCT
    ACCAAGGGCAGGGAGAGGACCCTATAGAGGCCTGGGACAGGAGCT
    CAATGAGAAAGG
    CD8 signal MALPVTALLLPLALLLHAARP  73
    peptide
    CD8a GCTGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAAACCGACCACGAC  74
    transmem- TCCCGCCCCGCGCCCTCCGACACCCGCTCCCACCATCGCCTCTCAAC
    brane + 5′ CTCTTAGTCTTCGCCCCGAGGCATGCCGACCCGCCGCCGGGGGTGCT
    Linker GTTCATACGAGGGGCTTGGACTTCGCTTGTGATATTTACATTTGGGC
    (underlined) TCCGTTGGCGGGTACGTGCGGCGTCCTTTTGTTGTCACTCGTTATTAC
    TTTGTATTGTAATCACAGGAATCGC
    CD8a SAAAFVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVH  75
    transmem- TRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCNHRNR
    brane + 5′
    Linker
    (underlined)
    CD8a TTTGTCCCGGTATTTCTCCCAGCCAAACCGACCACGACTCCCGCCCC  76
    transmem- GCGCCCTCCGACACCCGCTCCCACCATCGCCTCTCAACCTCTTAGTC
    brane TTCGCCCCGAGGCATGCCGACCCGCCGCCGGGGGTGCTGTTCATAC
    (without GAGGGGCTTGGACTTCGCTTGTGATATTTACATTTGGGCTCCGTTGG
    linker) CGGGTACGTGCGGCGTCCTTTTGTTGTCACTCGTTATTACTTTGTATT
    GTAATCACAGGAATCGC
    CD8a FVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGL  77
    transmem- DFACDIYIWAPLAGTCGVLLLSLVITLYCNHRNR
    brane
    (without
    linker)
    CD28 co- TCAAAGCGGAGTAGGTTGTTGCATTCCGATTACATGAATATGACTCC  45
    stimulatory TCGCCGGCCTGGGCCGACAAGAAAACATTACCAACCCTATGCCCCC
    CCACGAGACTTCGCTGCGTACAGGTCC
    CD28 co- SKRSRLLHSDYMNMTPRRPGPTRKHYQPYAPPRDFAAYRS  46
    stimulatory
    41BB co- AAACGGGGCAGAAAGAAACTCCTGTATATATTCAAACAACCATTTA  43
    stimulatory TGAGACCAGTACAAACTACTCAAGAGGAAGATGGCTGTAGCTGCCG
    ATTTCCAGAAGAAGAAGAAGGAGGATGTGAACTG
    41BB co- KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL  44
    stimulatory
    CD3ζ CGAGTGAAGTTTTCCCGAAGCGCAGACGCTCCGGCATATCAGCAAG  47
    GACAGAATCAGCTGTATAACGAACTGAATTTGGGACGCCGCGAGGA
    GTATGACGTGCTTGATAAACGCCGGGGGAGAGACCCGGAAATGGGG
    GGTAAACCCCGAAGAAAGAATCCCCAAGAAGGACTCTACAATGAAC
    TCCAGAAGGATAAGATGGCGGAGGCCTACTCAGAAATAGGTATGAA
    GGGCGAACGACGACGGGGAAAAGGTCACGATGGCCTCTACCAAGG
    GTTGAGTACGGCAACCAAAGATACGTACGATGCACTGCATATGCAG
    GCCCTGCCTCCCAGA
    CD3ζ RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGG  48
    KPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLS
    TATKDTYDALHMQALPPR
  • TABLE 8
    Donor Components
    Donor structure: TRAC[LHA]-EF1a[promoter]-CAR-
    polyA-TRAC[RHA]
    SEQ
    ID
    Name Sequence NO:
    TRAC- GAGATGTAAGGAGCTGCTGTGACTTGCTCAAGGCCTTATATCGAGT 78
    LHA AAACGGTAGTGCTGGGGCTTAGACGCAGGTGTTCTGATTTATAGTT
    CAAAACCTCTATCAATGAGAGAGCAATCTCCTGGTAATGTGATAGA
    TTTCCCAACTTAATGCCAACATACCATAAACCTCCCATTCTGCTAAT
    GCCCAGCCTAAGTTGGGGAGACCACTCCAGATTCCAAGATGTACAG
    TTTGCTTTGCTGGGCCTTTTTCCCATGCCTGCCTTTACTCTGCCAGA
    GTTATATTGCTGGGGTTTTGAAGAAGATCCTATTAAATAAAAGAAT
    AAGCAGTATTATTAAGTAGCCCTGCATTTCAGGTTTCCTTGAGTGG
    CAGGCCAGGCCTGGCCGTGAACGTTCACTGAAATCATGGCCTCTTG
    GCCAAGATTGATAGCTTGTGCCTGTCCCTGAGTCCCAGTCCATCAC
    GAGCAGCTGGTTTCTAAGATGCTATTTCCCGTATAAAGCATGAGAC
    CGTGACTTGCCAGCCCCACAGAGCCCCGCCCTTGTCCATCACTGGC
    ATCTGGACTCCAGCCTGGGTTGGGGCAAAGAGGGAAATGAGATCA
    TGTCCTAACCCTGATCCTCTTGTCCCACAGATATCCAGAACCCTGAC
    CCTGCCGTGTACCAGCTGAGAGACTCTAAATCCAGTGACAAGTCTG
    TCTGCCTATTCACCGATTTTGATTCTCAAACAAATGTGTCACAAAGT
    AAGGATTCTGATGTGTATATCACAGACAAAACTGTGCTAGACATGA
    GGTCTATGGACTTCA
    EF1α GGCTCCGGTGCCCGTCAGTGGGCAGAGCGCACATCGCCCACAGTCC
    79
    promoter CCGAGAAGTTGGGGGGAGGGGTCGGCAATTGAACCGGTGCCTAGA
    GAAGGTGGCGCGGGGTAAACTGGGAAAGTGATGTCGTGTACTGGC
    TCCGCCTTTTTCCCGAGGGTGGGGGAGAACCGTATATAAGTGCAGT
    AGTCGCCGTGAACGTTCTTTTTCGCAACGGGTTTGCCGCCAGAACA
    CAGGTAAGTGCCGTGTGTGGTTCCCGCGGGCCTGGCCTCTTTACGG
    GTTATGGCCCTTGCGTGCCTTGAATTACTTCCACTGGCTGCAGTACG
    TGATTCTTGATCCCGAGCTTCGGGTTGGAAGTGGGTGGGAGAGTTC
    GAGGCCTTGCGCTTAAGGAGCCCCTTCGCCTCGTGCTTGAGTTGAG
    GCCTGGCCTGGGCGCTGGGGCCGCCGCGTGCGAATCTGGTGGCACC
    TTCGCGCCTGTCTCGCTGCTTTCGATAAGTCTCTAGCCATTTAAAAT
    TTTTGATGACCTGCTGCGACGCTTTTTTTCTGGCAAGATAGTCTTGT
    AAATGCGGGCCAAGATCTGCACACTGGTATTTCGGTTTTTGGGGCC
    GCGGGCGGCGACGGGGCCCGTGCGTCCCAGCGCACATGTTCGGCG
    AGGCGGGGCCTGCGAGCGCGGCCACCGAGAATCGGACGGGGGTAG
    TCTCAAGCTGGCCGGCCTGCTCTGGTGCCTGGCCTCGCGCCGCCGT
    GTATCGCCCCGCCCTGGGCGGCAAGGCTGGCCCGGTCGGCACCAGT
    TGCGTGAGCGGAAAGATGGCCGCTTCCCGGCCCTGCTGCAGGGAG
    CTCAAAATGGAGGACGCGGCGCTCGGGAGAGCGGGCGGGTGAGTC
    ACCCACACAAAGGAAAAGGGCCTTTCCGTCCTCAGCCGTCGCTTCA
    TGTGACTCCACGGAGTACCGGGCGCCGTCCAGGCACCTCGATTAGT
    TCTCGAGCTTTTGGAGTACGTCGTCTTTAGGTTGGGGGGAGGGGTT
    TTATGCGATGGAGTTTCCCCACACTGAGTGGGTGGAGACTGAAGTT
    AGGCCAGCTTGGCACTTGATGTAATTCTCCTTGGAATTTGCCCTTTT
    TGAGTTTGGATCTTGGTTCATTCTCAAGCCTCAGACAGTGGTTCAA
    AGTTTTTTTCTTCCATTTCAGGTGTCGTGA
    Synthetic AATAAAATCGCTATCCATCGAAGATGGATGTGTGTTGGTTTTTTGT 80
    poly(A) GTG
    signal
    TRAC- TGGAGCAACAAATCTGACTTTGCATGTGCAAACGCCTTCAACAACA 81
    RHA GCATTATTCCAGAAGACACCTTCTTCCCCAGCCCAGGTAAGGGCAG
    CTTTGGTGCCTTCGCAGGCTGTTTCCTTGCTTCAGGAATGGCCAGGT
    TCTGCCCAGAGCTCTGGTCAATGATGTCTAAAACTCCTCTGATTGGT
    GGTCTCGGCCTTATCCATTGCCACCAAAACCCTCTTTTTACTAAGAA
    ACAGTGAGCCTTGTTCTGGCAGTCCAGAGAATGACACGGGAAAAA
    AGCAGATGAAGAGAAGGTGGCAGGAGAGGGCACGTGGCCCAGCCT
    CAGTCTCTCCAACTGAGTTCCTGCCTGCCTGCCTTTGCTCAGACTGT
    TTGCCCCTTACTGCTCTTCTAGGCCTCATTCTAAGCCCCTTCTCCAA
    GTTGCCTCTCCTTATTTCTCCCTGTCTGCCAAAAAATCTTTCCCAGC
    TCACTAAGTCAGTCTCACGCAGTCACTCATTAACCCACCAATCACT
    GATTGTGCCGGCACATGAATGCACCAGGTGTTGAAGTGGAGGAATT
    AAAAAGTCAGATGAGGGGTGTGCCCAGAGGAAGCACCATTCTAGT
    TGGGGGAGCCCATCTGTCAGCTGGGAAAAGTCCAAATAACTTCAG
    ATTGGAATGTGTTTTAACTCAGGGTTGAGAAAACAGCTACCTTCAG
    GACAAAAGTCAGGGAAGGGCTCTCTGAAGAAATGCTACTTGAAGA
    TACCAGCCCTACCAAGGGCAGGGAGAGGACCCTATAGAGGCCTGG
    GACAGGAGCTCAATGAGAAAGG
  • TABLE 9
    SEQ ID
    CAR CAR structure NO:
    CTX-973 CD8[signal peptide]-VL-linker-VH-CD8[tm]- 95, 96
    CAR 41BB[co-stimulatory domain]-CD3ζ
    CTX-974 CD8[signal peptide]-VL-linker-VH-CD8[tm]- 100, 101
    CAR 41BB[co-stimulatory domain]-CD3ζ
    CTX-975 CD8[signal peptide]-VH-linker-VL-CD8[tm]- 104, 105
    CAR 41BB[co-stimulatory domain]-CD3ζ
    CTX-976 CD8[signal peptide]-VH-linker-VL-CD8[tm]- 108, 109
    CAR 41BB[co-stimulatory domain]-CD3ζ
    CTX-977 CD8[signal peptide]-VL-linker-VH-CD8[tm]- 112, 113
    CAR 41BB[co-stimulatory domain]-CD3ζ
    CTX-978 CD8[signal peptide]-VH-linker-VL-CD8[tm]- 116, 117
    CAR 41BB[co-stimulatory domain]-CD3ζ
    CTX-979 CD8[signal peptide]-VL-linker-VH-CD8[tm]- 68
    CAR 41BB[co-stimulatory domain]-CD3ζ
    CTX-979b CD8[signal peptide]-VH-linker-VL-CD8[tm]- 66
    CAR CD28[co-stimulatory domain]-CD3ζ
  • TABLE 10
    CAR Components
    SEQ
    ID
    Name Sequence NO:
    CTX-973 CAR CCACCATGGCGCTTCCGGTGACAGCACTGCTCCTCCCCT  95
    41BB co-stim TGGCGCTGTTGCTCCACGCAGCAAGGCCGGATGTCGTTA
    (nt) TGACACAATCTCCCTTGAGTTTGCCGGTTACCTTGGGAC
    AACCTGCTAGTATTTCATGTAGGAGTTCTCAAAGTCTCTT
    GCACTCCTCAGGGAACACCTACCTCGAATGGTACCAACA
    ACGCCCTGGCCAAAGCCCGCGGCCCTTGATATACAAAAT
    ATCAACAAGATTTAGCGGGGTACCCGATAGATTCAGCGG
    CTCTGGCAGCGGGACGGATTTTACCCTGAAAATTAGTCG
    CGTAGAAGCTGAAGACGTTGGTGTGTATTACTGCTTTCA
    AGGGAGCCATGTGCCTTACACATTTGGAGGAGGCACCA
    AGGTCGAGATTAAGGGAGGGGGTGGATCAGGTGGGGGT
    GGGTCCGGAGGCGGCGGCAGTCAAGTGCAGTTGGTTCA
    ATCAGGAGCTGAAGTTAAAAAGCCAGGAGCTTCAGTCA
    AGGTTTCATGCAAGGCGTCCGGTCTCACTATAGAGGATT
    ACTACATGCACTGGGTGCGGCAAGCTCCAGGCCAGGGG
    CTGGAGTGGATGGGATGGATTGATCCGGAAAACGGGGA
    CACAGAGTATGGGCCCAAATTCCAAGGCCGGGTGACAA
    TGACCAGAGATACTAGTATTTCAACAGCATACATGGAGC
    TGTCACGGCTGAGGTCAGACGATACGGCAGTCTACTATT
    GTGCAGTACATAACGCACATTATGGTACGTGGTTCGCTT
    ATTGGGGTCAAGGTACCCTGGTCACGGTAAGTTCAAGTG
    CTGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAAACCGAC
    CACGACTCCCGCCCCGCGCCCTCCGACACCCGCTCCCAC
    CATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAGGCATGC
    CGACCCGCCGCCGGGGGTGCTGTTCATACGAGGGGCTTG
    GACTTCGCTTGTGATATTTACATTTGGGCTCCGTTGGCGG
    GTACGTGCGGCGTCCTTTTGTTGTCACTCGTTATTACTTT
    AACTCCTGTATATATTCAAACAACCATTTATGAGACCAG
    TACAAACTACTCAAGAGGAAGATGGCTGTAGCTGCCGAT
    TTCCAGAAGAAGAAGAAGGAGGATGTGAACTGCGAGTG
    AAGTTTTCCCGAAGCGCAGACGCTCCGGCATATCAGCAA
    GGACAGAATCAGCTGTATAACGAACTGAATTTGGGACG
    CCGCGAGGAGTATGACGTGCTTGATAAACGCCGGGGGA
    GAGACCCGGAAATGGGGGGTAAACCCCGAAGAAAGAAT
    CCCCAAGAAGGACTCTACAATGAACTCCAGAAGGATAA
    GATGGCGGAGGCCTACTCAGAAATAGGTATGAAGGGCG
    AACGACGACGGGGAAAAGGTCACGATGGCCTCTACCAA
    GGGTTGAGTACGGCAACCAAAGATACGTACGATGCACT
    GCATATGCAGGCCCTGCCTCCCAGATAAT
    CTX-973 CAR ALPVTALLLPLALLLHAARPDVVMTQSPLSLPVTLGQPASIS  96
    41BB co-stim CRSSQSLLHSSGNTYLEWYQQRPGQSPRPLIYKISTRFSGVP
    (aa) DRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHVPYTFGG
    GTKVEIKGGGGSGGGGSGGGGSQVQLVQSGAEVKKPGAS
    VKVSCKASGLTIEDYYMHWVRQAPGQGLEWMGWIDPEN
    GDTEYGPKFQGRVTMTRDTSTSTAYMELSRLRSDDTAVYY
    CAVHNAHYGTWFAYWGGGTLVTVSSSAAAFVPVFLPAKP
    TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDF
    ACDIYIWAPLAGTCGVLLLSLVITLYCNHRNRKRGRKKLLY
    IFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSA
    DAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGG
    KPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGH
    DGLYQGLSTATKDTYDALHMQALPPR
    CTX-973 GATGTCGTTATGACACAATCTCCCTTGAGTTTGCCGGTTA  97
    scFv (nt) CCTTGGGACAACCTGCTAGTATTTCATGTAGGAGTTCTC
    AAAGTCTCTTGCACTCCTCAGGGAACACCTACCTCGAAT
    GGTACCAACAACGCCCTGGCCAAAGCCCGCGGCCCTTGA
    TATACAAAATATCAACAAGATTTAGCGGGGTACCCGATA
    GATTCAGCGGCTCTGGCAGCGGGACGGATTTTACCCTGA
    AAATTAGTCGCGTAGAAGCTGAAGACGTTGGTGTGTATT
    ACTGCTTTCAAGGGAGCCATGTGCCTTACACATTTGGAG
    GAGGCACCAAGGTCGAGATTAAGGGAGGGGGTGGATCA
    GGTGGGGGTGGGTCCGGAGGCGGCGGCAGTCAAGTGCA
    GTTGGTTCAATCAGGAGCTGAAGTTAAAAAGCCAGGAG
    CTTCAGTCAAGGTTTCATGCAAGGCGTCCGGTCTCACTA
    TAGAGGATTACTACATGCACTGGGTGCGGCAAGCTCCAG
    GCCAGGGGCTGGAGTGGATGGGATGGATTGATCCGGAA
    AACGGGGACACAGAGTATGGGCCCAAATTCCAAGGCCG
    GGTGACAATGACCAGAGATACTAGTATTTCAACAGCATA
    CATGGAGCTGTCACGGCTGAGGTCAGACGATACGGCAG
    TCTACTATTGTGCAGTACATAACGCACATTATGGTACGT
    GGTTCGCTTATTGGGGTCAAGGTACCCTGGTCACGGTAA
    GTTCA
    CTX-973 DVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEWY  82
    scFv (aa) QQRPGQSPRPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRVE
    (linker AEDVGVYYCFQGSHVPYTFGGGTKVEIKGGGGSGGGGSG
    underlined) GGGSQVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMH
    WVRQAPGQGLEWMGWIDPENGDTEYGPKFQGRVTMTRD
    TSISTAYMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQ
    GTLVTVSS
    CTX-973 QVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMHWVRQ  98
    scFv VH (aa) APGQGLEWMGWIDPENGDTEYGPKFQGRVTMTRDTSIST
    AYMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQGTLV
    TVSS
    CTX-973 DVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEWY  56
    scFv VL (aa) QQRPGQSPRPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRVE
    AEDVGVYYCFQGSHVPYTFGGGTKVEIK
    CTX-973 GAGATGTAAGGAGCTGCTGTGACTTGCTCAAGGCCTTAT  99
    Donor (nt) ATCGAGTAAACGGTAGTGCTGGGGCTTAGACGCAGGTGT
    LHA to RHA TCTGATTTATAGTTCAAAACCTCTATCAATGAGAGAGCA
    ATCTCCTGGTAATGTGATAGATTTCCCAACTTAATGCCA
    ACATACCATAAACCTCCCATTCTGCTAATGCCCAGCCTA
    AGTTGGGGAGACCACTCCAGATTCCAAGATGTACAGTTT
    GCTTTGCTGGGCCTTTTTCCCATGCCTGCCTTTACTCTGC
    CAGAGTTATATTGCTGGGGTTTTGAAGAAGATCCTATTA
    AATAAAAGAATAAGCAGTATTATTAAGTAGCCCTGCATT
    TCAGGTTTCCTTGAGTGGCAGGCCAGGCCTGGCCGTGAA
    CGTTCACTGAAATCATGGCCTCTTGGCCAAGATTGATAG
    CTTGTGCCTGTCCCTGAGTCCCAGTCCATCACGAGCAGC
    TGGTTTCTAAGATGCTATTTCCCGTATAAAGCATGAGAC
    CGTGACTTGCCAGCCCCACAGAGCCCCGCCCTTGTCCAT
    CACTGGCATCTGGACTCCAGCCTGGGTTGGGGCAAAGAG
    GGAAATGAGATCATGTCCTAACCCTGATCCTCTTGTCCC
    ACAGATATCCAGAACCCTGACCCTGCCGTGTACCAGCTG
    AGAGACTCTAAATCCAGTGACAAGTCTGTCTGCCTATTC
    ACCGATTTTGATTCTCAAACAAATGTGTCACAAAGTAAG
    GATTCTGATGTGTATATCACAGACAAAACTGTGCTAGAC
    ATGAGGTCTATGGACTTCAggctccggtgcccgtcagtgggcagagcgca
    catcgcccacagtccccgagaagttggggggaggggtcggcaattgaaccggtgcctaga
    gaaggtggcgcggggtaaactgggaaagtgatgtcgtgtactggctccgcctttttcccgagg
    gtgggggagaaccgtatataagtgcagtagtcgccgtgaacgttctttttcgcaacgggtttgc
    cgccagaacacaggtaagtgccgtgtgtggttcccgcgggcctggcctctttacgggttatgg
    cccttgcgtgccttgaattacttccactggctgcagtacgtgattcttgatcccgagcttcgggtt
    ggaagtgggtgggagagttcgaggccttgcgcttaaggagccccttcgcctcgtgcttgagtt
    gaggcctggcctgggcgctggggccgccgcgtgrgaatctggtggcaccttcgcgcctgtc
    tcgctgctttcgataagtctctagccatttaaaatttttgatgacctgctgcgacgctttttttctggc
    aagatagtcttgtaaatgcgggccaagatctgcacactggtatttcggtttttggggccgcggg
    cggcgacggggcccgtgcgtcccagcgcacatgttcggcgaggcggggcctgcgagcgc
    ggccaccgagaatcggacgggggtagtctcaagctggccggcctgctctggtgcctggcct
    cgcgccgccgtgtatcgccccgccctgggcggcaaggctggcccggtcggcaccagttgc
    gtgagcggaaagatggccgcttcccggccctgctgcagggagctcaaaatggaggacgcg
    gcgctcgggagagcgggcgggtgagtcacccacacaaaggaaaagggcctttccgtcctc
    agccgtcgcttcatgtgactccacggagtaccgggcgccgtccaggcacctcgattagttctc
    gagcttttggagtacgtcgtctttaggttggggggaggggttttatgcgatggagtttccccaca
    ctgagtgggtggagactgaagttaggccagcttggcacttgatgtaattctccttggaatttgcc
    ctttttgagtttgtatcttggttcattctcaagcctcagacagtggttcaaagtttttttcttccatttc
    aggtgtcgtgaCCACCATGGCGCTTCCGGTGACAGCACTGCTC
    CTCCCCTTGGCGCTGTTGCTCCACGCAGCAAGGCCGGAT
    GTCGTTATGACACAATCTCCCTTGAGTTTGCCGGTTACCT
    TGGGACAACCTGCTAGTATTTCATGTAGGAGTTCTCAAA
    GTCTCTTGCACTCCTCAGGGAACACCTACCTCGAATGGT
    ACCAACAACGCCCTGGCCAAAGCCCGCGGCCCTTGATAT
    ACAAAATATCAACAAGATTTAGCGGGGTACCCGATAGA
    TTCAGCGGCTCTGGCAGCGGGACGGATTTTACCCTGAAA
    ATTAGTCGCGTAGAAGCTGAAGACGTTGGTGTGTATTAC
    TGCTTTCAAGGGAGCCATGTGCCTTACACATTTGGAGGA
    GGCACCAAGGTCGAGATTAAGGGAGGGGGTGGATCAGG
    TGGGGGTGGGTCCGGAGGCGGCGGCAGTCAAGTGCAGT
    TGGTTCAATCAGGAGCTGAAGTTAAAAAGCCAGGAGCTT
    CAGTCAAGGTTTCATGCAAGGCGTCCGGTCTCACTATAG
    AGGATTACTACATGCACTGGGTGCGGCAAGCTCCAGGCC
    AGGGGCTGGAGTGGATGGGATGGATTGATCCGGAAAAC
    GGGGACACAGAGTATGGGCCCAAATTCCAAGGCCGGGT
    GACAATGACCAGAGATACTAGTATTTCAACAGCATACAT
    GGAGCTGTCACGGCTGAGGTCAGACGATACGGCAGTCT
    ACTATTGTGCAGTACATAACGCACATTATGGTACGTGGT
    TCGCTTATTGGGGTCAAGGTACCCTGGTCACGGTAAGTT
    CAAGTGCTGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAA
    ACCGACCACGACTCCCGCCCCGCGCCCTCCGACACCCGC
    TCCCACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAG
    GCATGCCGACCCGCCGCCGGGGGTGCTGTTCATACGAGG
    GGCTTGGACTTCGCTTGTGATATTTACATTTGGGCTCCGT
    TGGCGGGTACGTGCGGCGTCCTTTTGTTGTCACTCGTTAT
    TACTTTGTATTGTAATCACAGGAATCGCAAACGGGGCAG
    AAAGAAACTCCTGTATATATTCAAACAACCATTTATGAG
    ACCAGTACAAACTACTCAAGAGGAAGATGGCTGTAGCT
    GCCGATTTCCAGAAGAAGAAGAAGGAGGATGTGAACTG
    CGAGTGAAGTTTTCCCGAAGCGCAGACGCTCCGGCATAT
    CAGCAAGGACAGAATCAGCTGTATAACGAACTGAATTT
    GGGACGCCGCGAGGAGTATGACGTGCTTGATAAACGCC
    GGGGGAGAGACCCGGAAATGGGGGGTAAACCCCGAAGA
    AAGAATCCCCAAGAAGGACTCTACAATGAACTCCAGAA
    GGATAAGATGGCGGAGGCCTACTCAGAAATAGGTATGA
    AGGGCGAACGACGACGGGGAAAAGGTCACGATGGCCTC
    TACCAAGGGTTGAGTACGGCAACCAAAGATACGTACGA
    TGCACTGCATATGCAGGCCCTGCCTCCCAGATAATAATA
    AAATCGCTATCCATCGAAGATGGATGTGTGTTGGTTTTTT
    GTGTGTGGAGCAACAAATCTGACTTTGCATGTGCAAACG
    CCTTCAACAACAGCATTATTCCAGAAGACACCTTCTTCC
    CCAGCCCAGGTAAGGGCAGCTTTGGTGCCTTCGCAGGCT
    GTTTCCTTGCTTCAGGAATGGCCAGGTTCTGCCCAGAGC
    TCTGGTCAATGATGTCTAAAACTCCTCTGATTGGTGGTCT
    CGGCCTTATCCATTGCCACCAAAACCCTCTTTTTACTAAG
    AAACAGTGAGCCTTGTTCTGGCAGTCCAGAGAATGACAC
    GGGAAAAAAGCAGATGAAGAGAAGGTGGCAGGAGAGG
    GCACGTGGCCCAGCCTCAGTCTCTCCAACTGAGTTCCTG
    CCTGCCTGCCTTTGCTCAGACTGTTTGCCCCTTACTGCTC
    TTCTAGGCCTCATTCTAAGCCCCTTCTCCAAGTTGCCTCT
    CCTTATTTCTCCCTGTCTGCCAAAAAATCTTTCCCAGCTC
    ACTAAGTCAGTCTCACGCAGTCACTCATTAACCCACCAA
    TCACTGATTGTGCCGGCACATGAATGCACCAGGTGTTGA
    AGTGGAGGAATTAAAAAGTCAGATGAGGGGTGTGCCCA
    GAGGAAGCACCATTCTAGTTGGGGGAGCCCATCTGTCAG
    CTGGGAAAAGTCCAAATAACTTCAGATTGGAATGTGTTT
    TAACTCAGGGTTGAGAAAACAGCTACCITCAGGACAAA
    AGTCAGGGAAGGGCTCTCTGAAGAAATGCTACTTGAAG
    ATACCAGCCCTACCAAGGGCAGGGAGAGGACCCTATAG
    AGGCCTGGGACAGGAGCTCAATGAGAAAGG
    CTX-974 CAR CCACCATGGCGCTTCCGGTGACAGCACTGCTCCTCCCCT 100
    41BB co-stim TGGCGCTGTTGCTCCACGCAGCAAGGCCGCAGGTGCAGC
    (nt) TGGTCCAAAGCGGCGCCGAGGTTAAGAAACCAGGCGCA
    TCCGTCAAGGTTTCATGTAAAGCAAGTGGCTTGACTATA
    GAAGACTACTACATGCATTGGGTACGGCAAGCCCCTGGG
    CAGGGGCTGGAATGGATGGGGTGGATCGACCCGGAGAA
    TGGTGATACAGAGTACGGACCTAAGTTCCAGGGACGAG
    TTACCATGACGCGAGATACATCCATCTCCACGGCATACA
    TGGAGCTGAGTCGACTGCGGAGCGATGATACAGCTGTCT
    ATTATTGTGCTCTTCCACAATGCGCACTACGGCACCTGGT
    TCGCTTATTGGGGACAAGGTACCCTGGTCACAGTCAGCT
    CTGGGGGTGGCGGCAGTGGAGGGGGTGGTTCTGGTGGC
    GGGGGGTTCCGATGTTGTAATGACTCAAAGCCCTCTTTCTT
    TGCCAGTCACTCTCGGACAACCCGCGAGCATATCTTGCA
    GGTCTTCACAATCACTCCTTCACAGTAGCGGGAATACTT
    ACTTGGAGTGGTATCAGCAGCGGCCTGGTCAGTCCCCTA
    GACCGCTTATATATAAGATCTCCACTAGGTTCAGTGGAG
    TGCCGGACCGCTTTTCAGGCTCAGGTTCCGGGACGGACT
    TTACATTGAAAATATCCAGGGTGGAGGCGGAGGACGTC
    GGAGTCTACTATTGCTTCCAAGGCTCCCACGTCCCATAC
    ACTTTCGGTGGCGGTACAAAAGTGGAAATAAAAAGTGC
    TGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAAACCGACC
    ACGACTCCCGCCCCGCGCCCTCCGACACCCGCTCCCACC
    ATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAGGCATGCC
    GACCCGCCGCCGGGGGTGCTGTTCATACGAGGGGCTTGG
    ACTTCGCTTGTGATATTTACATTTGGGCTCCGTTGGCGGG
    TACGTGCGGCGTCCTTTTGTTGTCACTCGTTATTACTTTG
    TATTGTAATCACAGGAATCGCAAACGGGGCAGAAAGAA
    ACTCCTGTATATATTCAAACAACCATTTATGAGACCAGT
    ACAAACTACTCAAGAGGAAGATGGCTGTAGCTGCCGATT
    TCCAGAAGAAGAAGAAGGAGGATGTGAACTGCGAGTGA
    AGTTTTCCCGAAGCGCAGACGCTCCGGCATATCAGCAAG
    GACAGAATCAGCTGTATAACGAACTGAATTTGGGACGCC
    GCGAGGAGTATGACGTGCTTGATAAACGCCGGGGGAGA
    GACCCGGAAATGGGGGGTAAACCCCGAAGAAAGAATCC
    CCAAGAAGGACTCTACAATGAACTCCAGAAGGATAAGA
    TGGCGGAGGCCTACTCAGAAATAGGTATGAAGGGCGAA
    CGACGACGGGGAAAAGGTCACGATGGCCTCTACCAAGG
    GTTGAGTACGGCAACCAAAGATACGTACGATGCACTGC
    ATATGCAGGCCCTGCCTCCCAGATAAT
    CTX-974 CAR MALPVTALLLPLALLLHAARPQVQLVQSGAEVKKPGASV 101
    41BB co-stim KVSCKASGLTIEDYYMHWVRQAPGQGLEWMGWIDPENG
    (aa) DTEYGPKFQGRVTMTRDTSISTAYMELSRLRSDDTAVYYC
    AVHNAHYGTWFAYWGQGTLVTVSSGGGGSGGGGSGGGG
    SDVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEW
    YQQRPGQSPRPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRV
    EAEDVGVYYCFQGSHVPYTFGGGTKVEIKSAAAFVPVFLP
    AKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRG
    LDFACDIYIWAPLAGTCGVLLLSLVITLYCNHRNRKRGRK
    KLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKF
    SRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDP
    EMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRR
    GKGHDGLYQGLSTATKDTYDALHMQALPPR
    CTX-974 CAGGTGCAGCTGGTCCAAAGCGGCGCCGAGGTTAAGAA 102
    scFv (nt) ACCAGGCGCATCCGTCAAGGTTTCATGTAAAGCAAGTGG
    CTTGACTATAGAAGACTACTACATGCATTGGGTACGGCA
    AGCCCCTGGGCAGGGGCTGGAATGGATGGGGTGGATCG
    ACCCGGAGAATGGTGATACAGAGTACGGACCTAAGTTC
    CAGGGACGAGTTACCATGACGCGAGATACATCCATCTCC
    ACGGCATACATGGAGCTGAGTCGACTGCGGAGCGATGA
    TACAGCTGTCTATTATTGTGCTGTCCACAATGCGCACTAC
    GGCACCTGGTTCGCTTATTGGGGACAAGGTACCCTGGTTC
    ACAGTCAGCTCTGGGGGTGGCGGCAGTGGAGGGGGTGG
    TTCTGGTGGCGGGGGTTCCGATGTTGTAATGACTCAAAG
    CCCTCTTTCTTTGCCAGTCACTCTCGGACAACCCGCGAGC
    ATATCTTGCAGGTCTTCACAATCACTCCTTCACAGTAGC
    GGGAATACTTACTTGGAGTGGTATCAGCAGCGGCCTGGT
    CAGTCCCCTAGACCGCTTATATATAAGATCTCCACTAGG
    TTCAGTGGAGTGCCGGACCGCTTTTCAGGCTCAGGTTCC
    GGGACGGACTTTACATTGAAAATATCCAGGGTGGAGGC
    GGAGGACGTCGGAGTCTACTATTGCTTCCAAGGCTCCCA
    CGTCCCATACACTTTCGGTGGCGGTACAAAAGTGGAAAT
    AAAA
    CTX-974 QVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMHWVRQ  85
    scFv (aa) APGQGLEWMGWIDPENGDTEYGPKFQGRVTMTRDTSISTA
    (linker YMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQGTLVT
    underlined) VSSGGGGSGGGGSGGGGSDVVMTQSPLSLPVTLGQPASISC
    RSSQSLLHSSGNTYLEWYQQRPGQSPRPLIYKISTRFSGVPD
    RFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHVPYTFGGG
    TKVEIK
    CTX-974 QVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMHWVRQ  98
    scFv VH (aa) APGQGLEWMGWIDPENGDTEYGPKFQGRVTMTRDTSIST
    AYMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQGTLV
    TVSS
    CTX-974 DVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEWY  56
    scFv VL (aa) QQRPGQSPRPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRVE
    AEDVGVYYCFQGSHVPYTFGGGTKVEIK
    CTX-974 GAGATGTAAGGAGCTGCTGTGACTTGCTCAAGGCCTTAT 103
    Donor (nt) ATCGAGTAAACGGTAGTGCTGGGGCTTAGACGCAGGTGT
    LHA to RHA TCTGATTTATAGTTCAAAACCTCTATCAATGAGAGAGCA
    ATCTCCTGGTAATGTGATAGATTTCCCAACTTAATGCCA
    ACATACCATAAACCTCCCATTCTGCTAATGCCCAGCCTA
    AGTTGGGGAGACCACTCCAGATTCCAAGATGTACAGTTT
    GCTTTGCTGGGCCTTTTTCCCATGCCTGCCTTTACTCTGC
    CAGAGTTATATTGCTGGGGTTTTGAAGAAGATCCTATTA
    AATAAAAGAATAAGCAGTATTATTAAGTAGCCCTGCATT
    TCAGGTTTCCTTGAGTGGCAGGCCAGGCCTGGCCGTGAA
    CGTTCACTGAAATCATGGCCTCTTGGCCAAGATTGATAG
    CTTGTGCCTGTCCCTGAGTCCCAGTCCATCACGAGCAGC
    TGGTTTCTAAGATGCTATTTCCCGTATAAAGCATGAGAC
    CGTGACTTGCCAGCCCCACAGAGCCCCGCCCTTGTCCAT
    CACTGGCATCTGGACTCCAGCCTGGGTTGGGGCAAAGAG
    GGAAATGAGATCATGTCCTAACCCTGATCCTCTTGTCCC
    ACAGATATCCAGAACCCTGACCCTGCCGTGTACCAGCTG
    AGAGACTCTAAATCCAGTGACAAGTCTGTCTGCCTATTC
    ACCGATTTTGATTCTCAAACAAATGTGTCACAAAGTAAG
    GATTCTGATGTGTATATCACAGACAAAACTGTGCTAGAC
    ATGAGGTCTATGGACTTCAggctccggtgcccgtcagtgggcagagcgca
    catcgcccacagtccccgagaagttggggggaggggtcggcaattgaaccggtgcctaga
    gaaggtggcgcggggtaaactgggaaagtgatgtcgtgtactggctccgcctttttcccgagg
    gtgggggagaaccgtatataagtgcagtagtcgccgtgaacgttctttttcgcaacgggtttgc
    cgccagaacacaggtaagtgccgtgtgtggttcccgcgggcctggcctctttacgggttatgg
    cccttgcgtgccttgaattacttccactggctgcagtacgtgattcttgatcccgagcttcgggtt
    ggaagtgggtgggagagttcgaggccttgcgcttaaggagccccttcgcctcgtgcttgagtt
    gaggcctggcctgggcgctggggccgccgcgtgcgaatctggtggcaccttcgcgcctgtc
    tcgctgctttcgataagtctctagccatttaaaatttttgatgacctgctgcgacgctttttttctggc
    aagatagtcttgtaaatgcgggccaagatctgcacactggtatttcggtttttggggccgcggg
    cggcgacggggcccgtgcgtcccagcgcacatgttcggcgaggcggggcctgcgagcgc
    ggccaccgagaatcggacgggggtagtctcaagctggccggcctgctctggtgcctggcct
    cgcgccgccgtgtatcgccccgccctgggcggcaaggctggcccggtcggcaccagttgc
    gtgagcggaaagatggccgcttcccggccctgctgcagggagctcaaaatggaggacgcg
    gcgctcgggagagcgggcgggtgagtcacccacacaaaggaaaagggcctttccgtcctc
    agccgtcgcttcatgtgactccacggagtaccgggcgccgtccaggcacctcgattagttctc
    gagcttttggagtacgtcgtctttaggttggggggaggggttttatgcgatggagtttccccaca
    ctgagtgggtggagactgaagttaggccagcttggcacttgatgtaattctccttggaatttgcc
    ctttttgagtttggatcttggttcattctcaagcctcagacagtggttcaaagtttttttcttccatttc
    aggtgtcgtgaCCACCATGGCGCTTCCGGTGACAGCACTGCTC
    CTCCCCTTGGCGCTGTTGCTCCACGCAGCAAGGCCGCAG
    GTGCAGCTGGTCCAAAGCGGCGCCGAGGTTAAGAAACC
    AGGCGCATCCGTCAAGGTTTCATGTAAAGCAAGTGGCTT
    GACTATAGAAGACTACTACATGCATTGGGTACGGCAAGC
    CCCTGGGCAGGGGCTGGAATGGATGGGGTGGATCGACC
    CGGAGAATGGTGATACAGAGTACGGACCTAAGTTCCAG
    GGACGAGTTACCATGACGCGAGATACATCCATCTCCACG
    GCATACATGGAGCTGAGTCGACTGCGGAGCGATGATAC
    AGCTGTCTATTATTGTGCTGTCCACAATGCGCACTACGG
    CACCTGGTTCGCTTATTGGGGACAAGGTACCCTGGTCAC
    AGTCAGCTCTGGGGGTGGCGGCAGTGGAGGGGGTGGTT
    CTGGTGGCGGGGGTTCCGATGTTGTAATGACTCAAAGCC
    CTCTTTCTTTGCCAGTCACTCTCGGACAACCCGCGAGCAT
    ATCTTGCAGGTCTTCACAATCACTCCTTCACAGTAGCGG
    GAATACTTACTTGGAGTGGTATCAGCAGCGGCCTGGTCA
    GTCCCCTAGACCGCTTATATATAAGATCTCCACTAGGTT
    CAGTGGAGTGCCGGACCGCTTTTCAGGCTCAGGTTCCGG
    GACGGACTTTACATTGAAAATATCCAGGGTGGAGGCGG
    AGGACGTCGGAGTCTACTATTGCTTCCAAGGCTCCCACG
    TCCCATACACTTTCGGTGGCGGTACAAAAGTGGAAATAA
    AAAGTGCTGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAA
    ACCGACCACGACTCCCGCCCCGCGCCCTCCGACACCCGC
    TCCCACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAG
    GCATGCCGACCCGCCGCCGGGGGTGCTGTTCATACGAGG
    GGCTTGGACTTCGCTTGTGATATTTACATTTGGGCTCCGT
    TGGCGGGTACGTGCGGCGTCCTTTTGTTGTCACTCGTTAT
    TACTTTGTATTGTAATCACAGGAATCGCAAACGGGGCAG
    AAAGAAACTCCTGTATATATTCAAACAACCATTTATGAG
    ACCAGTACAAACTACTCAAGAGGAAGATGGCTGTAGCT
    GCCGATTTCCAGAAGAAGAAGAAGGAGGATGTGAACTG
    CGAGTGAAGTTTTCCCGAAGCGCAGACGCTCCGGCATAT
    CAGCAAGGACAGAATCAGCTGTATAACGAACTGAATTT
    GGGACGCCGCGAGGAGTATGACGTGCTTGATAAACGCC
    GGGGGAGAGACCCGGAAATGGGGGGTAAACCCCGAAGA
    AAGAATCCCCAAGAAGGACTCTACAATGAACTCCAGAA
    GGATAAGATGGCGGAGGCCTACTCAGAAATAGGTATGA
    AGGGCGAACGACGACGGGGAAAAGGTCACGATGGCCTC
    TACCAAGGGTTGAGTACGGCAACCAAAGATACGTACGA
    TGCACTGCATATGCAGGCCCTGCCTCCCAGATAATAATA
    AAATCGCTATCCATCGAAGATGGATGTGTGTTGGTTTTTT
    GTGTGTGGAGCAACAAATCTGACTTTGCATGTGCAAACG
    CCTTCAACAACAGCATTATTCCAGAAGACACCTTCTTCC
    CCAGCCCAGGTAAGGGCAGCTTTGGTGCCTTCGCAGGCT
    GTTTCCTTGCTTCAGGAATGGCCAGGTTCTGCCCAGAGC
    TCTGGTCAATGATGTCTAAAACTCCTCTGATTGGTGGTCT
    CGGCCTTATCCATTGCCACCAAAACCCTCTTTTTACTAAG
    AAACAGTGAGCCTTGTTCTGGCAGTCCAGAGAATGACAC
    GGGAAAAAAGCAGATGAAGAGAAGGTGGCAGGAGAGG
    GCACGTGGCCCAGCCTCAGTCTCTCCAACTGAGTTCCTG
    CCTGCCTGCCTTTGCTCAGACTGTTTGCCCCTTACTGCTC
    TTCTAGGCCTCATTCTAAGCCCCTTCTCCAAGTTGCCTCT
    CCTTATTTCTCCCTGTCTGCCAAAAAATCTTTCCCAGCTC
    ACTAAGTCAGTCTCACGCAGTCACTCATTAACCCACCAA
    TCACTGATTGTGCCGGCACATGAATGCACCAGGTGTTGA
    AGTGGAGGAATTAAAAAGTCAGATGAGGGGTGTGCCCA
    GAGGAAGCACCATTCTAGTTGGGGGAGCCCATCTGTCAG
    CTGGGAAAAGTCCAAATAACTTCAGATTGGAATGTGTTT
    TAACTCAGGGTTGAGAAAACAGCTACCTTCAGGACAAA
    AGTCAGGGAAGGGCTCTCTGAAGAAATGCTACTTGAAG
    ATACCAGCCCTACCAAGGGCAGGGAGAGGACCCTATAG
    AGGCCTGGGACAGGAGCTCAATGAGAAAGG
    CTX-975 CAR CCACCATGGCGCTTCCGGTGACAGCACTGCTCCTCCCCT 104
    41BB co-stim TGGCGCTGTTGCTCCACGCAGCAAGGCCGGATGTAGTTA
    (nt) TGACCCAGAGTCCGCTCTCTTTGCCGGTGACGCTCGGCC
    AACCGGCGTCTATTTCTTGCAGAAGTAGTCAATCACTTC
    TGCACTCTAGCGGTAACACTTATTTGGAGTGGTATCTCC
    AACGACCAGGGCAAAGCCCCAAGCCGTTGATTTATAAG
    ATCTCTACAAGATTCAGCGGAGTGCCCGACAGATTTTCC
    GGGAGTGGGTCCGGTACTGATTTCACTTTGAAAATTTCC
    CGCGTCGAGGCTGAAGATGTTGGTGTCTACTACTGCTTT
    CAGGGGAGCCATGTTCCATATACCTTTGGAGGTGGGACT
    AAGGTAGAAATTAAAGGTGGGGGTGGATCAGGGGGTGG
    CGGCAGCGGGGGAGGGGGCTCACAAGTGCAACTTGTGC
    AAAGTGGGGCCGAGGTGAAAAAACCCGGTGCAAGTGTA
    AAGGTCTCATGCAAAGCGTCTGGTTTGACAATTGAAGAC
    TATTATATGCATTGGGTGAGACAGGCCCCGGGCCAAGG
    CTTGGAATGGATGGGATGGATAGACCCCGAAAACGGTG
    ACACGGAGTACGGACCTAAATTTCAAGGAAGAGTGACA
    ATGACACGCGATACATCTATTAACACGGCTTATATGGAA
    CTGAGCCGACTTCGGAGTGATGACACTGCTGTATATTAT
    TGCGCCGTCCACAACGCACATTATGGCACCTGGTTTGCG
    TACTGGGGACAGGGAACTTTGGTTACAGTATCAAGCAGT
    GCTGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAAACCG
    ACCACGACTCCCGCCCCGCGCCCTCCGACACCCGCTCCC
    ACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAGGCA
    TGCCGACCCGCCGCCGGGGGTGCTGTTCATACGAGGGG
    CTTGGACTTCGCTTGTGATATTTACATTTGGGCTCCGTTG
    GCGGGTACGTGCGGCGTCCTTTTGTTGTCACTCGTTATTA
    CTTTGTATTGTAATCACAGGAATCGCAAACGGGGCAGA
    AAGAAACTCCTGTATATATTCAAACAACCATTTATGAGA
    CCAGTACAAACTACTCAAGAGGAAGATGGCTGTAGCTG
    CCGATTTCCAGAAGAAGAAGAAGGAGGATGTGAACTGC
    GAGTGAAGTTTTCCCGAAGCGCAGACGCTCCGGCATATC
    AGCAAGGACAGAATCAGCTGTATAACGAACTGAATTTG
    GGACGCCGCGAGGAGTATGACGTGCTTGATAAACGCCG
    GGGGAGAGACCCGGAAATGGGGGGTAAACCCCGAAGA
    AAGAATCCCCAAGAAGGACTCTACAATGAACTCCAGAA
    GGATAAGATGGCGGAGGCCTACTCAGAAATAGGTATGA
    AGGGCGAACGACGACGGGGAAAAGGTCACGATGGCCTC
    TACCAAGGGTTGAGTACGGCAACCAAAGATACGTACGA
    TGCACTGCATATGCAGGCCCTGCCTCCCAGATAAT
    CTX-975 CAR MALPVTALLLPLALLLHAARPDVVMTQSPLSLPVTLGQPA 105
    41BB co-stim SISCRSSQSLLHSSGNTYLEWYLQRPGQSPKPLIYKISTRFSG
    (aa) VPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHVPYTF
    GGGTKVEIKGGGGSGGGGSGGGGSQVQLVQSGAEVKKPG
    ASVKVSCKASGLTIEDYYMHWVRQAPGQGLEWMGWIDP
    ENGDTEYGPKFQGRVTMTRDTSINTAYMELSRLRSDDTAV
    YYCAVHNAHYGTWFAYWGQGTLVTVSSSAAAFVPVFLPA
    KPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGL
    DFACDIYIWAPLAGTCGVLLLSLVITLYCNHRNRKRGRKK
    LLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFS
    RSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPE
    MGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRG
    KGHDGLYQGLSTATKDTYDALHMQALPPR
    CTX-975 GATGTAGTTATGACCCAGAGTCCGCTCTCTTTGCCGGTG 106
    scFv (nt) ACGCTCGGCCAACCGGCGTCTATTTCTTGCAGAAGTAGT
    CAATCACTTCTGCACTCTAGCGGTAACACTTATTTGGAG
    TGGTATCTCCAACGACCAGGGCAAAGCCCCAAGCCGTTG
    ATTTATAAGATCTCTACAAGATTCAGCGGAGTGCCCGAC
    AGATTTTCCGGGAGTGGGTCCGGTACTGATTTCACTTTG
    AAAATTTCCCGCGTCGAGGCTGAAGATGTTGGTGTCTAC
    TACTGCTTTCAGGGGAGCCATGTTCCATATACCTTTGGA
    GGTGGGACTAAGGTAGAAATTAAAGGTGGGGGTGGATC
    AGGGGGTGGCGGCAGCGGGGGAGGGGGCTCACAAGTGC
    AACTTGTGCAAAGTGGGGCCGAGGTGAAAAAACCCGGT
    GCAAGTGTAAAGGTCTCATGCAAAGCGTCTGGTTTGACA
    ATTGAAGACTATTATATGCATTGGGTGAGACAGGCCCCG
    GGCCAAGGCTTGGAATGGATGGGATGGATAGACCCCGA
    AAACGGTGACACGGAGTACGGACCTAAATTTCAAGGAA
    GAGTGACAATGACACGCGATACATCTATTAACACGGCTT
    ATATGGAACTGAGCCGACTTCGGAGTGATGACACTGCTG
    TATATTATTGCGCCGTCCACAACGCACATTATGGCACCT
    GGTTTGCGTACTGGGGACAGGGAACTTTGGTTACAGTAT
    CAAGC
    CTX-975 DVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEWY  83
    scFv (aa) LQRPGQSPKPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRVE
    (linker AEDVGVYYCFQGSHVPYTFGGGTKVEIKGGGGSGGGGSG
    underlined) GGGSQVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMH
    WVRQAPGQGLEWMGWIDPENGDTEYGPKFQGRVTMTRD
    TSINTAYMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQ
    GTLVTVSS
    CTX-975 QVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMHWVRQ  90
    scFv VH (aa) APGQGLEWMGWIDPENGDTEYGPKFQGRVTMTRDTSINT
    AYMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQGTLV
    TVSS
    CTX-975 DVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEWY  88
    scFv VL (aa) LQRPGQSPKPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRVE
    AEDVGVYYCFQGSHVPYTFGGGTKVEIK
    CTX-975 GAGATGTAAGGAGCTGCTGTGACTTGCTCAAGGCCTTAT 107
    Donor (nt) ATCGAGTAAACGGTAGTGCTGGGGCTTAGACGCAGGTGT
    LHA to RHA TCTGATTTATAGTTCAAAACCTCTATCAATGAGAGAGCA
    ATCTCCTGGTAATGTGATAGATTTCCCAACTTAATGCCA
    ACATACCATAAACCTCCCATTCTGCTAATGCCCAGCCTA
    AGTTGGGGAGACCACTCCAGATTCCAAGATGTACAGTTT
    GCTTTGCTGGGCCTTTTTCCCATGCCTGCCTTTACTCTGC
    CAGAGTTATATTGCTGGGGTTTTGAAGAAGATCCTATTA
    AATAAAAGAATAAGCAGTATTATTAAGTAGCCCTGCATT
    TCAGGTTTCCTTGAGTGGCAGGCCAGGCCTGGCCGTGAA
    CGTTCACTGAAATCATGGCCTCTTGGCCAAGATTGATAG
    CTTGTGCCTGTCCCTGAGTCCCAGTCCATCACGAGCAGC
    TGGTTTCTAAGATGCTATTTCCCGTATAAAGCATGAGAC
    CGTGACTTGCCAGCCCCACAGAGCCCCGCCCTTGTCCAT
    CACTGGCATCTGGACTCCAGCCTGGGTTGGGGCAAAGAG
    GGAAATGAGATCATGTCCTAACCCTGATCCTCTTGTCCC
    ACAGATATCCAGAACCCTGACCCTGCCGTGTACCAGCTG
    AGAGACTCTAAATCCAGTGACAAGTCTGTCTGCCTATTC
    ACCGATTTTGATTCTCAAACAAATGTGTCACAAAGTAAG
    GATTCTGATGTGTATATCACAGACAAAACTGTGCTAGAC
    ATGAGGTCTATGGACTTCAggctccggtgcccgtcagtgggcagagcgca
    catcgcccacagtccccgagaagttggggggaggggtcggcaattgaaccggtgcctaga
    gaaggtggcgcggggtaaactgggaaagtgatgtcgtgtactggctccgcctttttcccgagg
    gtgggggagaaccgtatataagtgcagtagtcgccgtgaacgttctttttcgcaacgggtttgc
    cgccagaacacaggtaagtgccgtgtgtggttcccgcgggcctggcctctttacgggttatgg
    cccttgcgtgccttgaattacttccactggctgcagtacgtgattcttgatcccgagcttcgggtt
    ggaagtgggtgggagagttcgaggccttgcgcttaaggagccccttcgcctcgtgcttgagtt
    gaggcctggcctgggcgctggggccgccgcgtgcgaatctggtggcaccttcgcgcctgtc
    tcgctgctttcgataagtctctagccatttaaaatttttgatgacctgctgcgacgctttttttctggc
    aagatagtcttgtaaatgcgggccaagatctgcacactggtatttcggtttttggggccgcggg
    cggcgacggggcccgtgcgtcccagcgcacatgttcggcgaggcggggcctgcgagcgc
    ggccaccgagaatcggacgggggtagtctcaagctggccggcctgctctggtgcctggcct
    cgcgccgccgtgtatcgccccgccctgggcggcaaggctggcccggtcggcaccagttgc
    gtgagcggaaagatggccgcttcccggccctgctgcagggagctcaaaatggaggacgcg
    gcgctcgggagagcgggcgggtgagtcacccacacaaaggaaaagggcctttccgtcctc
    agccgtcgcttcatgtgactccacggagtaccgggcgccgtccaggcacctcgattagttctc
    gagcttttggagtacgtcgtctttaggttggggggaggggttttatgcgatggagtttccccaca
    ctgagtgggtggagactgaagttaggccagcttggcacttgatgtaattctccttggaatttgcc
    ctttttgagtttggatcttggttcattctcaagcctcagacagtggttcaaagtttttttcttccatttc
    aggtgtcgtgaCCACCATGGCGCTTCCGGTGACAGCACTGCTC
    CTCCCCTTGGCGCTGTTGCTCCACGCAGCAAGGCCGGAT
    GTAGTTATGACCCAGAGTCCGCTCTCTTTGCCGGTGACG
    CTCGGCCAACCGGCGTCTATTTCTTGCAGAAGTAGTCAA
    TCACTTCTGCACTCTAGCGGTAACACTTATTTGGAGTGGT
    ATCTCCAACGACCAGGGCAAAGCCCCAAGCCGTTGATTT
    ATAAGATCTCTACAAGATTCAGCGGAGTGCCCGACAGAT
    TTTCCGGGAGTGGGTCCGGTACTGATTTCACTTTGAAAA
    TTTCCCGCGTCGAGGCTGAAGATGTTGGTGTCTACTACT
    GCTTTCAGGGGAGCCATGTTCCATATACCTTTGGAGGTG
    GGACTAAGGTAGAAATTAAAGGTGGGGGTGGATCAGGG
    GGTGGCGGCAGCGGGGGAGGGGGCTCACAAGTGCAACT
    TGTGCAAAGTGGGGCCGAGGTGAAAAAACCCGGTGCAA
    GTGTAAAGGTCTCATGCAAAGCGTCTGGTTTGACAATTG
    AAGACTATTATATGCATTGGGTGAGACAGGCCCCGGGCC
    AAGGCTTGGAATGGATGGGATGGATAGACCCCGAAAAC
    GGTGACACGGAGTACGGACCTAAATTTCAAGGAAGAGT
    GACAATGACACGCGATACATCTATTAACACGGCTTATAT
    GGAACTGAGCCGACTTCGGAGTGATGACACTGCTGTATA
    TTATTGCGCCGTCCACAACGCACATTATGGCACCTGGTT
    TGCGTACTGGGGACAGGGAACTTTGGTTACAGTATCAAG
    CAGTGCTGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAAA
    CCGACCACGACTCCCGCCCCGCGCCCTCCGACACCCGCT
    CCCACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAGG
    CATGCCGACCCGCCGCCGGGGGTGCTGTTCATACGAGGG
    GCTTGGACTTCGCTTGTGATATTTACATTTGGGCTCCGTT
    GGCGGGTACGTGCGGCGTCCTTTTGTTGTCACTCGTTATT
    ACTTTGTATTGTAATCACAGGAATCGCAAACGGGGCAGA
    AAGAAACTCCTGTATATATTCAAACAACCATTTATGAGA
    CCAGTACAAACTACTCAAGAGGAAGATGGCTGTAGCTG
    CCGATTTCCAGAAGAAGAAGAAGGAGGATGTGAACTGC
    GAGTGAAGTTTTCCCGAAGCGCAGACGCTCCGGCATATC
    AGCAAGGACAGAATCAGCTGTATAACGAACTGAATTTG
    GGACGCCGCGAGGAGTATGACGTGCTTGATAAACGCCG
    GGGGAGAGACCCGGAAATGGGGGGTAAACCCCGAAGAA
    AGAATCCCCAAGAAGGACTCTACAATGAACTCCAGAAG
    GATAAGATGGCGGAGGCCTACTCAGAAATAGGTATGAA
    GGGCGAACGACGACGGGGAAAAGGTCACGATGGCCTCT
    ACCAAGGGTTGAGTACGGCAACCAAAGATACGTACGAT
    GCACTGCATATGCAGGCCCTGCCTCCCAGATAATAATAA
    AATCGCTATCCATCGAAGATGGATGTGTGTTGGTTTTTTG
    TGTGTGGAGCAACAAATCTGACTTTGCATGTGCAAACGC
    CTTCAACAACAGCATTATTCCAGAAGACACCTTCTTCCC
    CAGCCCAGGTAAGGGCAGCTTTGGTGCCTTCGCAGGCTG
    TTTCCTTGCTTCAGGAATGGCCAGGTTCTGCCCAGAGCT
    CTGGTCAATGATGTCTAAAACTCCTCTGATTGGTGGTCTC
    GGCCTTATCCATTGCCACCAAAACCCTCTTTTTACTAAGA
    AACAGTGAGCCTTGTTCTGGCAGTCCAGAGAATGACACG
    GGAAAAAAGCAGATGAAGAGAAGGTGGCAGGAGAGGG
    CACGTGGCCCAGCCTCAGTCTCTCCAACTGAGTTCCTGC
    CTGCCTGCCTTTGCTCAGACTGTTTGCCCCTTACTGCTCT
    TCTAGGCCTCATTCTAAGCCCCTTCTCCAAGTTGCCTCTC
    CTTATTTCTCCCTGTCTGCCAAAAAATCTTTCCCAGCTCA
    CTAAGTCAGTCTCACGCAGTCACTCATTAACCCACCAAT
    CACTGATTGTGCCGGCACATGAATGCACCAGGTGTTGAA
    GTGGAGGAATTAAAAAGTCAGATGAGGGGTGTGCCCAG
    AGGAAGCACCATTCTAGTTGGGGGAGCCCATCTGTCAGC
    TGGGAAAAGTCCAAATAACTTCAGATTGGAATGTGTTTT
    AACTCAGGGTTGAGAAAACAGCTACCTTCAGGACAAAA
    GTCAGGGAAGGGCTCTCTGAAGAAATGCTACTTGAAGAT
    ACCAGCCCTACCAAGGGCAGGGAGAGGACCCTATAGAG
    GCCTGGGACAGGAGCTCAATGAGAAAGG
    CTX-976 CAR CCACCATGGCGCTTCCGGTGACAGCACTGCTCCTCCCCT 108
    41BB co-stim TGGCGCTGTTGCTCCACGCAGCAAGGCCGCAGGTTCAAC
    (nt) TGGTTCAGAGTGGAGCAGAGGTAAAAAAGCCCGGAGCG
    TCCGTCAAAGTGTCATGTAAAGCCTCTGGACTTACTATC
    GAAGACTACTACATGCACTGGGTGAGGCAGGCGCCTGG
    CCAAGGTCTCGAGTGGATGGGTTGGATTGACCCTGAAA
    ATGGAGATACAGAATACGGCCCTAAGTTTCAAGGGCGA
    GTAACTATGACTCGAGATACGTCAATTAATACGGCATAC
    ATGGAGTTGTCTCGGCTCCGATCTGATGACACTGCAGTT
    TACTATTGTGCCGTCCACAATGCTCATTACGGGACATGG
    TTCGCTTACTGGGGGCAAGGGACACTCGTAACGGTTAGC
    TCTGGGGGAGGAGGGTCTGGTGGAGGGGGCTCAGGAGG
    GGGTGGTAGCGACGTAGTAATGACCCAGTCACCTCTGTC
    TTTGCCGGTCACGTTGGGCCAGCCTGCATCCATATCCTG
    CAGATCCAGCCAGAGCCTCCTGCACAGTAGTGGCAACA
    CGTATTTGGAATGGTACCTGCAGAGGCCGGGTCAAAGTC
    CAAAACCGCTGATCTATAAGATATCTACGCGATTTTCAG
    GGGTGCCGGACCGATTTAGCGGATCAGGAAGTGGAACC
    GACTTTACGCTCAAGATCAGCCGGGTTGAAGCCGAAGA
    TGTCGGCGTTTACTACTGTTTCCAAGGAAGCCACGTACC
    CTATACGTTTGGTGGCGGCACGAAGGTCGAGATAAAGA
    GTGCTGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAAACC
    GACCACGACTCCCGCCCCGCGCCCTCCGACACCCGCTCC
    CACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAGGC
    ATGCCGACCCGCCGCCGGGGGTGCTGTTCATACGAGGG
    GCTTGGACTTCGCTTGTGATATTTACATTTGGGCTCCGTT
    GGCGGGTACGTGCGGCGTCCTTTTGTTGTCACTCGTTATT
    ACTTTGTATTGTAATCACAGGAATCGCAAACGGGGCAG
    AAAGAAACTCCTGTATATATTCAAACAACCATTTATGAG
    ACCAGTACAAACTACTCAAGAGGAAGATGGCTGTAGCT
    GCCGATTTCCAGAAGAAGAAGAAGGAGGATGTGAACTG
    CGAGTGAAGTTTTCCCGAAGCGCAGACGCTCCGGCATAT
    CAGCAAGGACAGAATCAGCTGTATAACGAACTGAATTT
    GGGACGCCGCGAGGAGTATGACGTGCTTGATAAACGCC
    GGGGGAGAGACCCGGAAATGGGGGGTAAACCCCGAAG
    AAAGAATCCCCAAGAAGGACTCTACAATGAACTCCAGA
    AGGATAAGATGGCGGAGGCCTACTCAGAAATAGGTATG
    AAGGGCGAACGACGACGGGGAAAAGGTCACGATGGCCT
    CTACCAAGGGTTGAGTACGGCAACCAAAGATACGTACG
    ATGCACTGCATATGCAGGCCCTGCCTCCCAGATAAT
    CTX-976 CAR MALPVTALLLPLALLLHAARPQVQLVQSGAEVKKPGASV 109
    41BB co-stim KVSCKASGLTIEDYYMHWVRQAPGQGLEWMGWIDPENG
    (aa) DTEYGPKFQGRVTMTRDTSINTAYMELSRLRSDDTAVYYC
    AVHNAHYGTWFAYWGQGTLVTVSSGGGGSGGGGSGGGG
    SDVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEW
    YLQRPGQSPKPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRV
    EAEDVGVYYCFQGSHVPYTFGGGTKVEIKSAAAFVPVFLP
    AKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRG
    LDFACDIYIWAPLAGTCGVLLLSLVITLYCNHRNRKRGRK
    KLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKF
    SRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDP
    EMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRR
    GKGHDGLYQGLSTATKDTYDALHMQALPPR
    CTX-976 CAGGTTCAACTGGTTCAGAGTGGAGCAGAGGTAAAAAA 110
    scFv (nt) GCCCGGAGCGTCCGTCAAAGTGTCATGTAAAGCCTCTGG
    ACTTACTATCGAAGACTACTACATGCACTGGGTGAGGCA
    GGCGCCTGGCCAAGGTCTCGAGTGGATGGGTTGGATTGA
    CCCTGAAAATGGAGATACAGAATACGGCCCTAAGTTTCA
    AGGGCGAGTAACTATGACTCGAGATACGTCAATTAATAC
    GGCATACATGGAGTTGTCTCGGCTCCGATCTGATGACAC
    TGCAGTTTACTATTGTGCCGTCCACAATGCTCATTACGG
    GACATGGTTCGCTTACTGGGGGCAAGGGACACTCGTAAC
    GGTTAGCTCTGGGGGAGGAGGGTCTGGTGGAGGGGGCT
    CAGGAGGGGGTGGTAGCGACGTAGTAATGACCCAGTCA
    CCTCTGTCTTTGCCGGTCCGTTGGGCCGCCTGCATCCA
    TATCCTGCAGATCCAGCCAGAGCCTCCTGCACAGTAGTG
    GCAACACGTATTTGGAATGGTACCTGCAGAGGCCGGGTC
    AAAGTCCAAAACCGCTGATCTATAAGATATCTACGCGAT
    TTTCAGGGGTGCCGGACCGATTTAGCGGATCAGGAAGTG
    GAACCGACTTTACGCTCAAGATCAGCCGGGTTGAAGCCG
    AAGATGTCGGCGTTTACTACTGTTTCCAAGGAAGCCACG
    TACCCTATACGTTTGGTGGCGGCACGAAGGTCGAGATAA
    AG
    CTX-976 QVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMHWVRQ  86
    scFv (aa) APGQGLEWMGWIDPENGDTEYGPKFQGRVTMTRDTSINT
    (linker AYMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQGTLV
    underlined) TVSSGGGGSGGGGSGGGGSDVVMTQSPLSLPVTLGQPASIS
    CRSSQSLLHSSGNTYLEWYLQRPGQSPKPLIYKISTRFSGVP
    DRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHVPYTFGG
    GTKVEIK
    CTX-976 QVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMHWVRQ  90
    scFv VH (aa) APGQGLEWMGWIDPENGDTEYGPKFQGRVTMTRDTSINT
    AYMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQGTLV
    TVSS
    CTX-976 DVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEWY  88
    scFv VL (aa) LQRPGQSPKPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRVE
    AEDVGVYYCFQGSHVPYTFGGGTKVEIK
    CTX-976 GAGATGTAAGGAGCTGCTGTGACTTGCTCAAGGCCTTAT 111
    Donor (nt) ATCGAGTAAACGGTAGTGCTGGGGCTTAGACGCAGGTGT
    LHA to RHA TCTGATTTATAGTTCAAAACCTCTATCAATGAGAGAGCA
    ATCTCCTGGTAATGTGATAGATTTCCCAACTTAATGCCA
    ACATACCATAAACCTCCCATTCTGCTAATGCCCAGCCTA
    AGTTGGGGAGACCACTCCAGATTCCAAGATGTACAGTTT
    GCTTTGCTGGGCCTTTTTCCCATGCCTGCCTTTACTCTGC
    CAGAGTTATATTGCTGGGGTTTTGAAGAAGATCCTATTA
    AATAAAAGAATAAGCAGTATTATTAAGTAGCCCTGCATT
    TCAGGTTTCCTTGAGTGGCAGGCCAGGCCTGGCCGTGAA
    CGTTCACTGAAATCATGGCCTCTTGGCCAAGATTGATAG
    CTTGTGCCTGTCCCTGAGTCCCAGTCCATCACGAGCAGC
    TGGTTTCTAAGATGCTATTTCCCGTATAAAGCATGAGAC
    CGTGACTTGCCAGCCCCACAGAGCCCCGCCCTTGTCCAT
    CACTGGCATCTGGACTCCAGCCTGGGTTGGGGCAAAGAG
    GGAAATGAGATCATGTCCTAACCCTGATCCTCTTGTCCC
    ACAGATATCCAGAACCCTGACCCTGCCGTGTACCAGCTG
    AGAGACTCTAAATCCAGTGACAAGTCTGTCTGCCTATTC
    ACCGATTTTGATTCTCAAACAAATGTGTCACAAAGTAAG
    GATTCTGATGTGTATATCACAGACAAAACTGTGCTAGAC
    ATGAGGTCTATGGACTTCAggctccggtgcccgtcagtgggcagagcgca
    catcgcccacagtccccgagaagttggggggaggggtcggcaattgaaccggtgcctaga
    gaaggtggcgcggggtaaactgggaaagtgatgtcgtgtactggctccgcctttttcccgagg
    gtgggggagaaccgtatataagtgcagtagtcgccgtgaacgttctttttcgcaacgggtttgc
    cgccagaacacaggtaagtgccgtgtgtggttcccgcgggcctggcctctttacgggttatgg
    cccttgcgtgccttgaattacttccactggctgcagtacgtgattcttgatcccgagcttcgggtt
    ggaagtgggtgggagagttcgaggccttgcgcttaaggagccccttcgcctcgtgcttgagtt
    gaggcctggcctgggcgctggggccgccgcgtgcgaatctggtggcaccttcgcgcctgtc
    tcgctgattcgataagtctctagccatttaaaatttttgatgacctgctgcgacgctttttttctggc
    aagatagtcttgtaaatgcgggccaagatctgcacactggtatttcggtttttggggccgcggg
    cggcgacggggcccgtgcgtcccagcgcacatgttcggcgaggcggggcctgcgagcgc
    ggccaccgagaatcggacgggggtagtctcaagctggccggcctgctctggtgcctggcct
    cgcgccgccgtgtatcgccccgccctgggcggcaaggctggcccggtcggcaccagttgc
    gtgagcggaaagatggccgcttcccggccctgctgcagggagctcaaaatggaggacgcg
    gcgctcgggagagcgggcgggtgagtcacccacacaaaggaaaagggcctttccgtcctc
    agccgtcgcttcatgtgactccacggagtaccgggcgccgtccaggcacctcgattagttctc
    gagcttttggagtacgtcgtctttaggttggggggaggggttttatgcgatggagtttccccaca
    ctgagtgggtggagactgaagttaggccagcttggcacttgatgtaattctccttggaatttgcc
    ctttttgagtttggatcttggttcattctcaagcctcagacagtggttcaaagtttttttcttccatttc
    aggtgtcgtgaCCACCATGGCGCTTCCGGTGACAGCACTGCTC
    CTCCCCTTGGCGCTGTTGCTCCACGCAGCAAGGCCGCAG
    GTTCAACTGGTTCAGAGTGGAGCAGAGGTAAAAAAGCC
    CGGAGCGTCCGTCAAAGTGTCATGTAAAGCCTCTGGACT
    TACTATCGAAGACTACTACATGCACTGGGTGAGGCAGGC
    GCCTGGCCAAGGTCTCGAGTGGATGGGTTGGATTGACCC
    TGAAAATGGAGATACAGAATACGGCCCTAAGTTTCAAG
    GGCGAGTAACTATGACTCGAGATACGTCAATTAATACGG
    CATACATGGAGTTGTCTCGGCTCCGATCTGATGACACTG
    CAGTTTACTATTGTGCCGTCCACAATGCTCATTACGGGA
    CATGGTTCGCTTACTGGGGGCAAGGGACACTCGTAACGG
    TTAGCTCTGGGGGAGGAGGGTCTGGTGGAGGGGGCTCA
    GGAGGGGGTGGTAGCGACGTAGTAATGACCCAGTCACC
    TCTGTCTTTGCCGGTCACGTTGGGCCAGCCTGCATCCATA
    TCCTGCAGATCCAGCCAGAGCCTCCTGCACAGTAGTGGC
    AACACGTATTTGGAATGGTACCTGCAGAGGCCGGGTCAA
    AGTCCAAAACCGCTGATCTATAAGATATCTACGCGATTT
    TCAGGGGTGCCGGACCGATTTAGCGGATCAGGAAGTGG
    AACCGACTTTACGCTCAAGATCAGCCGGGTTGAAGCCGA
    AGATGTCGGCGTTTACTACTGTTTCCAAGGAAGCCACGT
    ACCCTATACGTTTGGTGGCGGCACGAAGGTCGAGATAAA
    GAGTGCTGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAAA
    CCGACCACGACTCCCGCCCCGCGCCCTCCGACACCCGCT
    CCCACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAGG
    CATGCCGACCCGCCGCCGGGGGTGCTGTTCATACGAGGG
    GCTTGGACTTCGCTTGTGATATTTACATTTGGGCTCCGTT
    GGCGGGTACGTGCGGCGTCCTTTTGTTGTCACTCGTTATT
    ACTTTGTATTGTAATCACAGGAATCGCAAACGGGGCAGA
    AAGAAACTCCTGTATATATTCAAACAACCATTTATGAGA
    CCAGTACAAACTACTCAAGAGGAAGATGGCTGTAGCTG
    CCGATTTCCAGAAGAAGAAGAAGGAGGATGTGAACTGC
    GAGTGAAGTTTTCCCGAAGCGCAGACGCTCCGGCATATC
    AGCAAGGACAGAATCAGCTGTATAACGAACTGAATTTG
    GGACGCCGCGAGGAGTATGACGTGCTTGATAAACGCCG
    GGGGAGAGACCCGGAAATGGGGGGTAAACCCCGAAGAA
    AGAATCCCCAAGAAGGACTCTACAATGAACTCCAGAAG
    GATAAGATGGCGGAGGCCTACTCAGAAATAGGTATGAA
    GGGCGAACGACGACGGGGAAAAGGTCACGATGGCCTCT
    ACCAAGGGTTGAGTACGGCAACCAAAGATACGTACGAT
    GCACTGCATATGCAGGCCCTGCCTCCCAGATAATAATAA
    AATCGCTATCCATCGAAGATGGATGTGTGTTGGTTTTTTG
    TGTGTGGAGCAACAAATCTGACTTTGCATGTGCAAACGC
    CTTCAACAACAGCATTATTCCAGAAGACACCTTCTTCCC
    CAGCCCAGGTAAGGGCAGCTTTGGTGCCTTCGCAGGCTG
    TTTCCTTGCTTCAGGAATGGCCAGGTTCTGCCCAGAGCT
    CTGGTCAATGATGTCTAAAACTCCTCTGATTGGTGGTCTC
    GGCCTTATCCATTGCCACCAAAACCCTCTTTTTACTAAGA
    AACAGTGAGCCTTGTTCTGGCAGTCCAGAGAATGACACG
    GGAAAAAAGCAGATGAAGAGAAGGTGGCAGGAGAGGG
    CACGTGGCCCAGCCTCAGTCTCTCCAACTGAGTTCCTGC
    CTGCCTGCCTTTGCTCAGACTGTTTGCCCCTTACTGCTCT
    TCTAGGCCTCATTCTAAGCCCCTTCTCCAAGTTGCCTCTC
    CTTATTTCTCCCTGTCTGCCAAAAAATCTTTCCCAGCTCA
    CTAAGTCAGTCTCACGCAGTCACTCATTAACCCACCAAT
    CACTGATTGTGCCGGCACATGAATGCACCAGGTGTTGAA
    GTGGAGGAATTAAAAAGTCAGATGAGGGGTGTGCCCAG
    AGGAAGCACCATTCTAGTTGGGGGAGCCCATCTGTCAGC
    TGGGAAAAGTCCAAATAACTTCAGATTGGAATGTGTTTT
    AACTCAGGGTTGAGAAAACAGCTACCTTCAGGACAAAA
    GTCAGGGAAGGGCTCTCTGAAGAAATGCTACTTGAAGAT
    ACCAGCCCTACCAAGGGCAGGGAGAGGACCCTATAGAG
    GCCTGGGACAGGAGCTCAATGAGAAAGG
    CTX-977 CAR CCACCATGGCGCTTCCGGTGACAGCACTGCTCCTCCCCT 112
    41BB co-stim TGGCGCTGTTGCTCCACGCAGCAAGGCCGGACGTTGTGA
    (nt) TGACGCAGTCTCCTCTGAGCCTGCCAGTTACGTTGGGGC
    AACCCGCATCAATATCTTGTAGGTCCAGTCAGAGCCTGC
    TTCACAGCTCTGGCAACACTTACTTGGAATGGTACCTCC
    AGAGACCTGGACAGAGTCCCAAGCCATTGATTTACAAG
    ATTTCAACGCGATTTAGTGGAGTGCCCGATCGATTCTCT
    GGGAGTGGCTCTGGGACTGATTTCACACTTAAAATAAGT
    AGGGTGGAGGCTGAAGATGTGGGTGTATATTATTGTTTT
    CAAGGGTCCCATGTCCCTTACACTTTCGGCGGCGGCACC
    AAAGTTGAGATCAAAGGTGGTGGTGGGTCCGGCGGTGG
    AGGCAGTGGGGGTGGCGGGTCACAAGTTCAACTTGTCC
    AGTCAGGGGCTGAAGTAAAAAAGCCTGGTGCATCAGTT
    AAAGTTTCATGTAAGGCTTCCGGCCTTACCATTGAAGAT
    TACTATATGCACTGGGTTAGACAAGCTCCTGGACAAGGT
    CTGGAGTGGATGGGCTGGATAGACCCCGAGAATGGTGA
    CACAGAATACGGGCCTAAGTTCCAGGGTAGGGTAACAA
    TGACGCGGGATACATCCATTTCCACAGCTTACATGGAAC
    TGAGTAGACTCAGATCTGACGACACTGCTGTCTACTATT
    GTGCCGTCCATAACGCGCATTATGGCACTTGGTTCGCAT
    ATTGGGGGCAAGGCACTCTTGTTACAGTGTCCTCAAGTG
    CTGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAAACCGA
    CCACGACTCCCGCCCCGCGCCCTCCGACACCCGCTCCCA
    CCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAGGCAT
    GCCGACCCGCCGCCGGGGGTGCTGTTCATACGAGGGGC
    TTGGACTTCGCTTGTGATATTTACATTTGGGCTCCGTTGG
    CGGGTACGTGCGGCGTCCTTTTGTTGTCACTCGTTATTAC
    TTTGTATTGTAATCACAGGAATCGCAAACGGGGCAGAA
    AGAAACTCCTGTATATATTCAAACAACCATTTATGAGAC
    CAGTACAAACTACTCAAGAGGAAGATGGCTGTAGCTGC
    CGATTTCCAGAAGAAGAAGAAGGAGGATGTGAACTGCG
    AGTGAAGTTTTCCCGAAGCGCAGACGCTCCGGCATATCA
    GCAAGGACAGAATCAGCTGTATAACGAACTGAATTTGG
    GACGCCGCGAGGAGTATGACGTGCTTGATAAACGCCGG
    GGGAGAGACCCGGAAATGGGGGGTAAACCCCGAAGAA
    AGAATCCCCAAGAAGGACTCTACAATGAACTCCAGAAG
    GATAAGATGGCGGAGGCCTACTCAGAAATAGGTATGAA
    GGGCGAACGACGACGGGGAAAAGGTCACGATGGCCTCT
    ACCAAGGGTTGAGTACGGCAACCAAAGATACGTACGAT
    GCACTGCATATGCAGGCCCTGCCTCCCAGATAAT
    CTX-977 CAR MALPVTALLLPLALLLHAARPDVVMTQSPLSLPVTLGQPA 113
    41BB co-stim SISCRSSQSLLHSSGNTYLEWYLQRPGQSPKPLIYKISTRFSG
    (aa) VPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHVPYTF
    GGGTKVEIKGGGGSGGGGSGGGGSQVQLVQSGAEVKKPG
    ASVKVSCKASGLTIEDYYMHWVRQAPGQGLEWMGWIDP
    ENGDTEYGPKFQGRVTMTRDTSISTAYMELSRLRSDDTAV
    YYCAVHNAHYGTWFAYWGQGTLVTVSSSAAAFVPVFLPA
    KPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGL
    DFACDIYIWAPLAGTCGVLLLSLVITLYCNHRNRKRGRKK
    LLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFS
    RSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPE
    MGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRG
    KGHDGLYQGLSTATKDTYDALHMQALPPR
    CTX-977 GACGTTGTGATGACGCAGTCTCCTCTGAGCCTGCCAGTT 114
    scFv (nt) ACGTTGGGGCAACCCGCATCAATATCTTGTAGGTCCAGT
    CAGAGCCTGCTTCACAGCTCTGGCAACACTTACTTGGAA
    TGGTACCTCCAGAGACCTGGACAGAGTCCCAAGCCATTG
    ATTTACAAGATTTCAACGCGATTTAGTGGAGTGCCCGAT
    CGATTCTCTGGGAGIGGCTCTGGGACTGATTTCACACTT
    AAAATAAGTAGGGTGGAGGCTGAAGATGTGGGTGTATA
    TTATTGTTTTCAAGGGTCCCATGTCCCTTACACTTTCGGC
    GGCGGCACCAAAGTTGAGATCAAAGGTGGTGGTGGGTC
    CGGCGGTGGAGGCAGTGGGGGTGGCGGGTCACAAGTTC
    AACTTGTCCAGTCAGGGGCTGAAGTAAAAAAGCCTGGT
    GCATCAGTTAAAGTTTCATGTAAGGCTTCCGGCCTTACC
    ATTGAAGATTACTATATGCACTGGGTTAGACAAGCTCCT
    GGACAAGGTCTGGAGTGGATGGGCTGGATAGACCCCGA
    GAATGGTGACACAGAATACGGGCCTAAGTTCCAGGGTA
    GGGTAACAATGACGCGGGATACATCCATTTCCACAGCTT
    ACATGGAACTGAGTAGACTCAGATCTGACGACACTGCTG
    TCTACTATTGTGCCGTCCATAACGCGCATTATGGCACTTG
    GTTCGCATATTGGGGGCAAGGCACTCTTGTTACAGTGTC
    CTCA
    CTX-977 DVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEWY  84
    scFv (aa) LQRPGQSPKPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRVE
    (linker AEDVGVYYCFQGSHVPYTFGGGTKVEIKGGGGSGGGGSG
    underlined) GGGSQVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMH
    WVRQAPGQGLEWMGWIDPENGDTEYGPKFQGRVTMTRD
    TSISTAYMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQ
    GTLVTVSS
    CTX-977 QVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMEIWVRQ  98
    scFv VH (aa) APGQGLEWMGWIDPENGDTEYGPKFQGRVTMTRDTSIST
    AYMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQGTLV
    TVSS
    CTX-977 DVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEWY  88
    scFv VL (aa) LQRPGQSPKPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRVE
    AEDVGVYYCFQGSHVPYTFGGGTKVEIK
    CTX-977 GAGATGTAAGGAGCTGCTGTGACTTGCTCAAGGCCTTAT 115
    Donor (nt) ATCGAGTAAACGGTAGTGCTGGGGCTTAGACGCAGGTGT
    LHA to RHA TCTGATTTATAGTTCAAAACCTCTATCAATGAGAGAGCA
    ATCTCCTGGTAATGTGATAGATTTCCCAACTTAATGCCA
    ACATACCATAAACCTCCCATTCTGCTAATGCCCAGCCTA
    AGTTGGGGAGACCACTCCAGATTCCAAGATGTACAGTTT
    GCTTTGCTGGGCCTTTTTCCCATGCCTGCCTTTACTCTGC
    CAGAGTTATATTGCTGGGGTTTTGAAGAAGATCCTATTA
    AATAAAAGAATAAGCAGTATTATTAAGTAGCCCTGCATT
    TCAGGTTTCCTTGAGTGGCAGGCCAGGCCTGGCCGTGAA
    CGTTCACTGAAATCATGGCCTCTTGGCCAAGATTGATAG
    CTTGTGCCTGTCCCTGAGTCCCAGTCCATCACGAGCAGC
    TGGTTTCTAAGATGCTATTTCCCGTATAAAGCATGAGAC
    CGTGACTTGCCAGCCCCACAGAGCCCCGCCCTTGTCCAT
    CACTGGCATCTGGACTCCAGCCTGGGTTGGGGCAAAGAG
    GGAAATGAGATCATGTCCTAACCCTGATCCTCTTGTCCC
    ACAGATATCCAGAACCCTGACCCTGCCGTGTACCAGCTG
    AGAGACTCTAAATCCAGTGACAAGTCTGTCTGCCTATTC
    ACCGATTTTGATTCTCAAACAAATGTGTCACAAAGTAAG
    GATTCTGATGTGTATATCACAGACAAAACTGTGCTAGAC
    ATGAGGTCTATGGACTTCAggctccggtgcccgtcagtgggcagagcgca
    catcgcccacagtccccgagaagttggggggaggggtcggcaattgaaccggtgcctaga
    gaaggtggcgcggggtaaactgggaaagtgatgtcgtgtactggctccgcctttttcccgagg
    gtgggggagaaccgtatataagtgcagtagtcgccgtgaacgttctttttcgcaacgggtttgc
    cgccagaacacaggtaagtgccgtgtgtggttcccgcgggcctggcctctttacgggttatgg
    cccttgcgtgccttgaattacttccactggctgcagtacgtgattcttgatcccgagcttcgggtt
    ggaagtgggtgggagagttcgaggccttgcgcttaaggagcccatcgcctcgtgatgagtt
    gaggcctggcctgggcgctggggccgccgcgtgcgaatctggtggcaccttcgcgcctgtc
    tcgctgattcgataagtctctagccatttaaaatttttgatgacctgctgcgacgctttttttctggc
    aagatagtcttgtaaatgcgggccaagatctgcacactggtatttcggtttttggggccgcggg
    cggcgacggggcccgtgcgtcccagcgcacatgttcggcgaggcggggcctgcgagcgc
    ggccaccgagaatcggacgggggtagtctcaagctggccggcctgctctggtgcctggcct
    cgcgccgccgtgtatcgccccgccctgggcggcaaggctggcccggtcggcaccagttgc
    gtgagcggaaagatggccgcttcccggccctgctgcagggagctcaaaatggaggacgcg
    gcgctcgggagagcgggcgggtgagtcacccacacaaaggaaaagggcctttccgtcctc
    agccgtcgcttcatgtgactccacggagtaccgggcgccgtccaggcacctcgattagttctc
    gagcttttggagtacgtcgtattaggttggggggaggggttttatgcgatggagtttccccaca
    ctgagtgggtggagactgaagttaggccagcttggcacttgatgtaattctccttggaatttgcc
    ctttttgagtttggatcttggttcattctcaagcctcagacagtggttcaaagtttttttcttccatttc
    aggtgtcgtgaCCACCATGGCGCTTCCGGTGACAGCACTGCTC
    CTCCCCTTGGCGCTGTTGCTCCACGCAGCAAGGCCGGAC
    GTTGTGATGACGCAGTCTCCTCTGAGCCTGCCAGTTACG
    TTGGGGCAACCCGCATCAATATCTTGTAGGTCCAGTCAG
    AGCCTGCTTCACAGCTCTGGCAACACTTACTTGGAATGG
    TACCTCCAGAGACCTGGACAGAGTCCCAAGCCATTGATT
    TACAAGATTTCAACGCGATTTAGTGGAGTGCCCGATCGA
    TTCTCTGGGAGTGGCTCTGGGACTGATTTCACACTTAAA
    ATAAGTAGGGTGGAGGCTGAAGATGTGGGTGTATATTAT
    TGTTTTCAAGGGTCCCATGTCCCTTACACTTTCGGCGGCG
    GCACCAAAGTTGAGATCAAAGGTGGTGGTGGGTCCGGC
    GGTGGAGGCAGTGGGGGTGGCGGGTCACAAGTTCAACT
    TGTCCAGTCAGGGGCTGAAGTAAAAAAGCCTGGTGCATC
    AGTTAAAGTTTCATGTAAGGCTTCCGGCCTTACCATTGA
    AGATTACTATATGCACTGGGTTAGACAAGCTCCTGGACA
    AGGTCTGGAGTGGATGGGCTGGATAGACCCCGAGAATG
    GTGACACAGAATACGGGCCTAAGTTCCAGGGTAGGGTA
    ACAATGACGCGGGATACATCCATTTCCACAGCTTACATG
    GAACTGAGTAGACTCAGATCTGACGACACTGCTGTCTAC
    TATTGTGCCGTCCATAACGCGCATTATGGCACTTGGTTC
    GCATATTGGGGGCAAGGCACTCTTGTTACAGTGTCCTCA
    AGTGCTGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAAAC
    CGACCACGACTCCCGCCCCGCGCCCTCCGACACCCGCTC
    CCACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAGGC
    ATGCCGACCCGCCGCCGGGGGTGCTGTTCATACGAGGGG
    CTTGGACTTCGCTTGTGATATTTACATTTGGGCTCCGTTG
    GCGGGTACGTGCGGCGTCCTTTTGTTGTCACTCGTTATTA
    CTTTGTATTGTAATCACAGGAATCGCAAACGGGGCAGAA
    AGAAACTCCTGTATATATTCAAACAACCATTTATGAGAC
    CAGTACAAACTACTCAAGAGGAAGATGGCTGTAGCTGC
    CGATTTCCAGAAGAAGAAGAAGGAGGATGTGAACTGCG
    AGTGAAGTTTTCCCGAAGCGCAGACGCTCCGGCATATCA
    GCAAGGACAGAATCAGCTGTATAACGAACTGAATTTGG
    GACGCCGCGAGGAGTATGACGTGCTTGATAAACGCCGG
    GGGAGAGACCCGGAAATGGGGGGTAAACCCCGAAGAAA
    GAATCCCCAAGAAGGACTCTACAATGAACTCCAGAAGG
    ATAAGATGGCGGAGGCCTACTCAGAAATAGGTATGAAG
    GGCGAACGACGACGGGGAAAAGGTCACGATGGCCTCTA
    CCAAGGGTTGAGTACGGCAACCAAAGATACGTACGATG
    CACTGCATATGCAGGCCCTGCCTCCCAGATAATAATAAA
    ATCGCTATCCATCGAAGATGGATGTGTGTTGGTTTTTTGT
    GTGTGGAGCAACAAATCTGACTTTGCATGTGCAAACGCC
    TTCAACAACAGCATTATTCCAGAAGACACCTTCTTCCCC
    AGCCCAGGTAAGGGCAGCTTTGGTGCCTTCGCAGGCTGT
    TTCCTTGCTTCAGGAATGGCCAGGTTCTGCCCAGAGCTC
    TGGTCAATGATGTCTAAAACTCCTCTGATTGGTGGTCTC
    GGCCTTATCCATTGCCACCAAAACCCTCTTTTTACTAAGA
    AACAGTGAGCCTTGTTCTGGCAGTCCAGAGAATGACACG
    GGAAAAAAGCAGATGAAGAGAAGGTGGCAGGAGAGGG
    CACGTGGCCCAGCCTCAGTCTCTCCAACTGAGTTCCTGC
    CTGCCTGCCTTTGCTCAGACTGTTTGCCCCTTACTGCTCT
    TCTAGGCCTCATTCTAAGCCCCTTCTCCAAGTTGCCTCTC
    CTTATTTCTCCCTGTCTGCCAAAAAATCTTTCCCAGCTCA
    CTAAGTCAGTCTCACGCAGTCACTCATTAACCCACCAAT
    CACTGATTGTGCCGGCACATGAATGCACCAGGTGTTGAA
    GTGGAGGAATTAAAAAGTCAGATGAGGGGTGTGCCCAG
    AGGAAGCACCATTCTAGTTGGGGGAGCCCATCTGTCAGC
    TGGGAAAAGTCCAAATAACTTCAGATTGGAATGTGTTTT
    AACTCAGGGTTGAGAAAACAGCTACCTTCAGGACAAAA
    GTCAGGGAAGGGCTCTCTGAAGAAATGCTACTTGAAGAT
    ACCAGCCCTACCAAGGGCAGGGAGAGGACCCTATAGAG
    GCCTGGGACAGGAGCTCAATGAGAAAGG
    CTX-978 CAR CCACCATGGCGCTTCCGGTGACAGCACTGCTCCTCCCCT 116
    41BB co-stim TGGCGCTGTTGCTCCACGCAGCAAGGCCGCAGGTACAA
    (nt) CTCGTTCAGAGCGGTGCAGAGGTTAAGAAACCGGGCGC
    CAGTGTCAAAGTATCATGCAAGGCGAGTGGTCTGACCAT
    CGAAGATTATTATATGCATTGGGTGAGACAAGCACCGG
    GGCAGGGGCTCGAATGGATGGGTTGGATCGACCCCGAA
    AATGGTGATACGGAGTATGGCCCGAAATTTCAGGGTCG
    GGTCACGATGACCCGCGATACAAGCATCAGTACTGCAT
    ACATGGAGCTCTCTCGCTTGCGGAGTGATGATACCGCCG
    TTTATTATTGCGCGGTTCACAACGCTCATTATGGCACTTG
    GTTCGCGTATTGGGGCCAAGGAACACTGGTTACAGTGA
    GCAGTGGAGGGGGTGGCTCTGGTGGCGGCGGGAGCGGC
    GGAGGGGGCAGTGATGTTGTGATGACACAGTCACCCCT
    GAGTCTCCCGGTCACTCTTGGGCAACCAGCCAGCATAAG
    CTGTCGCAGTTCTCAGAGCTTGCTCCATAGCTCCGGGAA
    TACCTACCTCGAATGGTATCTCCAAAGACCCGGTCAATC
    TCCAAAGCCTTTGATTTACAAGATTAGTACACGATTTAG
    TGGGGTCCCAGATAGATTTTCAGGTAGTGGATCTGGTAC
    AGATTTCACATTGAAAATATCACGCGTCGAGGCGGAGG
    ATGTCGGGGTCTACTATTGCTTTCAAGGTAGTCACGTGC
    CCTACACGTTTGGTGGCGGTACGAAGGTCGAAATCAAG
    AGTGCTGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAAA
    CCGACCACGACTCCCGCCCCGCGCCCTCCGACACCCGCT
    CCCACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAG
    GCATGCCGACCCGCCGCCGGGGGTGCTGTTCATACGAG
    GGGCTTGGACTTCGCTTGTGATATTTACATTTGGGCTCC
    GTTGGCGGGTACGTGCGGCGTCCTTTTGTTGTCACTCGTT
    ATTACTTTGTATTGTAATCACAGGAATCGCAAACGGGGC
    AGAAAGAAACTCCTGTATATATTCAAACAACCATTTATG
    AGACCAGTACAAACTACTCAAGAGGAAGATGGCTGTAG
    CTGCCGATTTCCAGAAGAAGAAGAAGGAGGATGTGAAC
    TGCGAGTGAAGTTTTCCCGAAGCGCAGACGCTCCGGCAT
    ATCAGCAAGGACAGAATCAGCTGTATAACGAACTGAAT
    TTGGGACGCCGCGAGGAGTATGACGTGCTTGATAAACG
    CCGGGGGAGAGACCCGGAAATGGGGGGTAAACCCCGAA
    GAAAGAATCCCCAAGAAGGACTCTACAATGAACTCCAG
    AAGGATAAGATGGCGGAGGCCTACTCAGAAATAGGTAT
    GAAGGGCGAACGACGACGGGGAAAAGGTCACGATGGC
    CTCTACCAAGGGTTGAGTACGGCAACCAAAGATACGTA
    CGATGCACTGCATATGCAGGCCCTGCCTCCCAGATAAT
    CTX-978 CAR MALPVTALLLPLALLLHAARPQVQLVQSGAEVKKPGASV 117
    41BB co-stim KVSCKASGLTIEDYYMHWVRQAPGQGLEWMGWIDPENG
    (aa) DTEYGPKFQGRVTMTRDTSISTAYMELSRLRSDDTAVYYC
    AVHNAHYGTWFAYWGQGTLVTVSSGGGGSGGGGSGGGG
    SDVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEW
    YLQRPGQSPKPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRV
    EAEDVGVYYCFQGSHVPYTFGGGTKVEIKSAAAFVPVFLP
    AKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRG
    LDFACDIYIWAPLAGTCGVLLLSLVITLYCNHRNRKRGRK
    KLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKF
    SRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDP
    EMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRR
    GKGHDGLYQGLSTATKDTYDALHMQALPPR
    CTX-978 CAGGTACAACTCGTTCAGAGCGGTGCAGAGGTTAAGAA 118
    scFv (nt) ACCGGGCGCCAGTGTCAAAGTATCATGCAAGGCGAGTG
    GTCTGACCATCGAAGATTATTATATGCATTGGGTGAGAC
    AAGCACCGGGGCAGGGGCTCGAATGGATGGGTTGGATC
    GACCCCGAAAATGGTGATACGGAGTATGGCCCGAAATTT
    CAGGGTCGGGTCACGATGACCCGCGATACAAGCATCAG
    TACTGCATACATGGAGCTCTCTCGCTTGCGGAGTGATGA
    TACCGCCGTTTATTATTGCGCGGTTCACAACGCTCATTAT
    GGCACTTGGTTCGCGTATTGGGGCCAAGGAACACTGGTT
    ACAGTGAGCAGTGGAGGGGGTGGCTCTGGTGGCGGCGG
    GAGCGGCGGAGGGGGCAGTGATGTTGTGATGACACAGT
    CACCCCTGAGTCTCCCGGTCACTCTTGGGCAACCAGCCA
    GCATAAGCTGTCGCAGTTCTCAGAGCTTGCTCCATAGCT
    CCGGGAATACCTACCTCGAATGGTATCTCCAAAGACCCG
    GTCAATCTCCAAAGCCTTTGATTTACAAGATTAGTACAC
    GATTTAGTGGGGTCCCAGATAGATTTTCAGGTAGTGGAT
    CTGGTACAGATTTCACATTGAAAATATCACGCGTCGAGG
    CGGAGGATGTCGGGGTCTACTATTGCTTTCAAGGTAGTC
    ACGTGCCCTACACGTTTGGTGGCGGTACGAAGGTCGAAA
    TCAAG
    CTX-978 QVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMHWVRQ  87
    scFv (aa) APGQGLEWMGWIDPENGDTEYGPKFQGRVTMTRDTSISTA
    (linker YMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQGTLVT
    underlined) VSSGGGGSGGGGSGGGGSDVVMTQSPLSLPVTLGQPASISC
    RSSQSLLHSSGNTYLEWYLQRPGQSPKPLIYKISTRFSGVPD
    RFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHVPYTFGGG
    TKVEIK
    CTX-978 QVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMHWVRQ  98
    scFv VH (aa) APGQGLEWMGWIDPENGDTEYGPKFQGRVTMTRDTSIST
    AYMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQGTLV
    TVSS
    CTX-978 DVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEWY  88
    scFv VL (aa) LQRPGQSPKPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRVE
    AEDVGVYYCFQGSHVPYTFGGGTKVEIK
    CTX-978 GAGATGTAAGGAGCTGCTGTGACTTGCTCAAGGCCTTAT 119
    Donor (nt) ATCGAGTAAACGGTAGTGCTGGGGCTTAGACGCAGGTGT
    LHA to RHA TCTGATTTATAGTTCAAAACCTCTATCAATGAGAGAGCA
    ATCTCCTGGTAATGTGATAGATTTCCCAACTTAATGCCA
    ACATACCATAAACCTCCCATTCTGCTAATGCCCAGCCTA
    AGTTGGGGAGACCACTCCAGATTCCAAGATGTACAGTTT
    GCTTTGCTGGGCCTTTTTCCCATGCCTGCCTTTACTCTGC
    CAGAGTTATATTGCTGGGGTTTTGAAGAAGATCCTATTA
    AATAAAAGAATAAGCAGTATTATTAAGTAGCCCTGCATT
    TCAGGTTTCCTTGAGTGGCAGGCCAGGCCTGGCCGTGAA
    CGTTCACTGAAATCATGGCCTCTTGGCCAAGATTGATAG
    CTTGTGCCTGTCCCTGAGTCCCAGTCCATCACGAGCAGC
    TGGTTTCTAAGATGCTATTTCCCGTATAAAGCATGAGAC
    CGTGACTTGCCAGCCCCACAGAGCCCCGCCCTTGTCCAT
    CACTGGCATCTGGACTCCAGCCTGGGTTGGGGCAAAGAG
    GGAAATGAGATCATGTCCTAACCCTGATCCTCTTGTCCC
    ACAGATATCCAGAACCCTGACCCTGCCGTGTACCAGCTG
    AGAGACTCTAAATCCAGTGACAAGTCTGTCTGCCTATTC
    ACCGATTTTGATTCTCAAACAAATGTGTCACAAAGTAAG
    GATTCTGATGTGTATATCACAGACAAAACTGTGCTAGAC
    ATGAGGTCTATGGACTTCAggctccggtgcccgtcagtgggcagagcgca
    catcgcccacagtccccgagaagttggggggaggggtcggcaattgaaccggtgcctaga
    gaaggtggcgcggggtaaactgggaaagtgatgtcgtgtactggctccgcctttttcccgagg
    gtgggggagaaccgtatataagtgcagtagtcgccgtgaacgttctttttcgcaacgggtttgc
    cgccagaacacaggtaagtgccgtgtgtggttcccgcgggcctggcctctttacgggttatgg
    cccttgcgtgccttgaattacttccactggctgcagtacgtgattcttgatcccgagatcgggtt
    ggaagtgggtgggagagttcgaggccttgcgcttaaggagccccttcgcctcgtgcttgagtt
    gaggcctggcctgggcgctggggccgccgcgtgcgaatctggtggcaccttcgcgcctgtc
    tcgctgctttcgataagtctctagccatttaaaatttttgatgacctgctgcgacgctttttttctggc
    aagatagtcttgtaaatgcgggccaagatctgcacactggtatttcggtttttggggccgcggg
    cggcgacggggcccgtgcgtcccagcgcacatgttcggcgaggcggggcctgcgagcgc
    ggccaccgagaatcggacgggggtagtctcaagctggccggcctgctctggtgcctggcct
    cgcgccgccgtgtatcgccccgccctgggcggcaaggctggcccggtcggcaccagttgc
    gtgagcggaaagatggccgcttcccggccctgctgcagggagctcaaaatggaggacgcg
    gcgctcgggagagcgggcgggtgagtcacccacacaaaggaaaagggcctttccgtcctc
    agccgtcgcttcatgtgactccacggagtaccgggcgccgtccaggcacctcgattagttctc
    gagcttttggagtacgtcgtctttaggttggggggaggggttttatgcgatggagtttccccaca
    ctgagtgggtggagactgaagttaggccagcttggcacttgatgtaattctccttggaatttgcc
    ctttttgagtttggatcttggttcattctcaagcctcagacagtggttcaaagtttttttcttccatttc
    aggtgtcgtgaCCACCATGGCGCTTCCGGTGACAGCACTGCTC
    CTCCCCTTGGCGCTGTTGCTCCACGCAGCAAGGCCGCAG
    GTACAACTCGTTCAGAGCGGTGCAGAGGTTAAGAAACC
    GGGCGCCAGTGTCAAAGTATCATGCAAGGCGAGTGGTCT
    GACCATCGAAGATTATTATATGCATTGGGTGAGACAAGC
    ACCGGGGCAGGGGCTCGAATGGATGGGTTGGATCGACC
    CCGAAAATGGTGATACGGAGTATGGCCCGAAATTTCAG
    GGTCGGGTCACGATGACCCGCGATACAAGCATCAGTACT
    GCATACATGGAGCTCTCTCGCTTGCGGAGTGATGATACC
    GCCGTTTATTATTGCGCGGTTCACAACGCTCATTATGGC
    ACTTGGTTCGCGTATTGGGGCCAAGGAACACTGGTTACA
    GTGAGCAGTGGAGGGGGTGGCTCTGGTGGCGGCGGGAG
    CGGCGGAGGGGGCAGTGATGTTGTGATGACACAGTCAC
    CCCTGAGTCTCCCGGTCACTCTTGGGCAACCAGCCAGCA
    TAAGCTGTCGCAGTTCTCAGAGCTTGCTCCATAGCTCCG
    GGAATACCTACCTCGAATGGTATCTCCAAAGACCCGGTC
    AATCTCCAAAGCCTTTGATTTACAAGATTAGTACACGAT
    TTAGTGGGGTCCCAGATAGATTTTCAGGTAGTGGATCTG
    GTACAGATTTCACATTGAAAATATCACGCGTCGAGGCGG
    AGGATGTCGGGGTCTACTATTGCTTTCAAGGTAGTCACG
    TGCCCTACACGTTTGGTGGCGGTACGAAGGTCGAAATCA
    AGAGTGCTGCTGCCTTTGTCCCGGTATTTCTCCCAGCCAA
    ACCGACCACGACTCCCGCCCCGCGCCCTCCGACACCCGC
    TCCCACCATCGCCTCTCAACCTCTTAGTCTTCGCCCCGAG
    GCATGCCGACCCGCCGCCGGGGGTGCTGTTCATACGAGG
    GGCTTGGACTTCGCTTGTGATATTTACATTTGGGCTCCGT
    TGGCGGGTACGTGCGGCGTCCTTTTGTTGTCACTCGTTAT
    TACTTTGTATTGTAATCACAGGAATCGCAAACGGGGCAG
    AAAGAAACTCCTGTATATATTCAAACAACCATTTATGAG
    ACCAGTACAAACTACTCAAGAGGAAGATGGCTGTAGCT
    GCCGATTTCCAGAAGAAGAAGAAGGAGGATGTGAACTG
    CGAGTGAAGTTTTCCCGAAGCGCAGACGCTCCGGCATAT
    CAGCAAGGACAGAATCAGCTGTATAACGAACTGAATTT
    GGGACGCCGCGAGGAGTATGACGTGCTTGATAAACGCC
    GGGGGAGAGACCCGGAAATGGGGGGTAAACCCCGAAGA
    AAGAATCCCCAAGAAGGACTCTACAATGAACTCCAGAA
    GGATAAGATGGCGGAGGCCTACTCAGAAATAGGTATGA
    AGGGCGAACGACGACGGGGAAAAGGTCACGATGGCCTC
    TACCAAGGGTTGAGTACGGCAACCAAAGATACGTACGA
    TGCACTGCATATGCAGGCCCTGCCTCCCAGATAATAATA
    AAATCGCTATCCATCGAAGATGGATGTGTGTTGGTTTTTT
    GTGTGTGGAGCAACAAATCTGACTTTGCATGTGCAAACG
    CCTTCAACAACAGCATTATTCCAGAAGACACCTTCTTCC
    CCAGCCCAGGTAAGGGCAGCTTTGGTGCCTTCGCAGGCT
    GTTTCCTTGCTTCAGGAATGGCCAGGTTCTGCCCAGAGC
    TCTGGTCAATGATGTCTAAAACTCCTCTGATTGGTGGTCT
    CGGCCTTATCCATTGCCACCAAAACCCTCTTTTTACTAAG
    AAACAGTGAGCCTTGTTCTGGCAGTCCAGAGAATGACAC
    GGGAAAAAAGCAGATGAAGAGAAGGTGGCAGGAGAGG
    GCACGTGGCCCAGCCTCAGTCTCTCCAACTGAGTTCCTG
    CCTGCCTGCCTTTGCTCAGACTGTTTGCCCCTTACTGCTC
    TTCTAGGCCTCATTCTAAGCCCCTTCTCCAAGTTGCCTCT
    CCTTATTTCTCCCTGTCTGCCAAAAAATCTTTCCCAGCTC
    ACTAAGTCAGTCTCACGCAGTCACTCATTAACCCACCAA
    TCACTGATTGTGCCGGCACATGAATGCACCAGGTGTTGA
    AGTGGAGGAATTAAAAAGTCAGATGAGGGGTGTGCCCA
    GAGGAAGCACCATTCTAGTTGGGGGAGCCCATCTGTCAG
    CTGGGAAAAGTCCAAATAACTTCAGATTGGAATGTGTTT
    TAACTCAGGGTTGAGAAAACAGCTACCTTCAGGAAAA
    AGTCAGGGAAGGGCTCTCTGAAGAAATGCTACTTGAAG
    ATACCAGCCCTACCAAGGGCAGGGAGAGGACCCTATAG
    AGGCCTGGGACAGGAGCTCAATGAGAAAGG
    CTX-979 CAR MALPVTALLLPLALLLHAARPQVQLVQSGAEVKKPGASV  68
    41BB co-stim KVSCKASGLTIEDYYMHWVRQAPGQGLEWMGWIDPENG
    (aa) DTEYGPKFQGRVTMTRDTSINTAYMELSRLRSDDTAVYYC
    AVHNAHYGTWFAYWGQGTLVTVSSGGGGSGGGGSGGGG
    SDVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEW
    YQQRPGQSPRPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRV
    EAEDVGVYYCFQGSHVPYTFGGGTKVEIKSAAAFVPVFLP
    AKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRG
    LDFACDIYIWAPLAGTCGVLLLSLVITLYCNHRNRKRGRK
    KLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKF
    SRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDP
    EMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRR
    GKGHDGLYQGLSTATKDTYDALHMQALPPR
    CTX-979b MALPVTALLLPLALLLHAARPQVQLVQSGAEVKKPGASV  66
    CAR KVSCKASGLTIEDYYMHWVRQAPGQGLEWMGWIDPENG
    CD28 co-stim DTEYGPKFQGRVTMTRDTSINTAYMELSRLRSDDTAVYYC
    (aa) AVHNAHYGTWFAYWGQGTLVTVSSGGGGSGGGGSGGGG
    SDVVMTQSPLSLPVTLGQPASISCRSSQSLLHSSGNTYLEW
    YQQRPGQSPRPLIYKISTRFSGVPDRFSGSGSGTDFTLKISRV
    EAEDVGVYYCFQGSHVPYTFGGGTKVEIKSAAAFVPVFLP
    AKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRG
    LDFACDIYIWAPLAGTCGVLLLSLVITLYCNHRNRSKRSRL
    LHSDYMNMTPRRPGPTRKHYQPYAPPRDFAAYRSRVKFSR
    SADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEM
    GGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGK
    GHDGLYQGLSTATKDTYDALHMQALPPR
    CTX-979 and QVQLVQSGAEVKKPGASVKVSCKASGLTIEDYYMHWVRQ  70
    CTX-979b APGQGLEWMGWIDPENGDTEYGPKFQGRVTMTRDTSINT
    scFv (aa) AYMELSRLRSDDTAVYYCAVHNAHYGTWFAYWGQGTLV
    (linker TVSSGGGGSGGGGSGGGGSDVVMTQSPLSLPVTLGQPASIS
    underlined) CRSSQSLLHSSGNTYLEWYQQRPGQSPRPLIYKISTRFSGVP
    DRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHVPYTFGG
    GTKVEIK
    CTX-979 and QVQLVQSGAEVKKPGASVKVSCKASGLTIE DYYMH WVRQAPG  55
    CTX-979b QGLEWMG WIDPENGDTEYGPKFQG RVTMTRDTSINTAYMELS
    scFv VH (aa)  RLRSDDTAVYYCAV HNAHYGTWFAY WGQGTLVTVSS
    CDRs- in bold
    CTX-979 and DVVMTQSPLSLPVTLGQPASISC RSSQSLLHSSGNTYLE WYQQR  56
    CTX-979b PGQSPRPLIY KISTRFS GVPDRFSGSGSGTDFTLKISRVEAEDVGV
    scFv VL (aa)  YYC FQGSHVPYT FGGGTKVEIK
    CDRs- in bold
  • All references, patents and patent applications disclosed herein are incorporated by reference with respect to the subject matter for which each is cited, which in some cases may encompass the entirety of the document.
  • The indefinite articles “a” and “an,” as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to mean “at least one.”
  • It should also be understood that, unless clearly indicated to the contrary, in any methods claimed herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited.
  • In the claims, as well as in the specification above, all transitional phrases such as “comprising,” “including,” “carrying,” “having,” “containing,” “involving,” “holding,” “composed of,” and the like are to be understood to be open-ended, i.e., to mean including but not limited to. Only the transitional phrases “consisting of” and “consisting essentially of” shall be closed or semi-closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03.
  • The terms “about” and “substantially” preceding a numerical value mean±10% of the recited numerical value.
  • Where a range of values is provided, each value between the upper and lower ends of the range are specifically contemplated and described herein.

Claims (66)

1. An engineered T cell comprising a nucleic acid encoding a chimeric antigen receptor (CAR), wherein the CAR comprises an ectodomain that binds specifically to LIV1.
2. The engineered T cell of claim 1 further comprising a disrupted T cell receptor alpha chain constant region (TRAC) gene.
3. The engineered T cell of claim 1 further comprising a disrupted beta-2-microglobulin (β2M) gene.
4. The engineered T cell of claim 1, wherein the ectodomain of the CAR comprises an anti-LIV1 antibody.
5. The engineered T cell of claim 4, wherein the anti-LIV1 antibody is an anti-LIV1 single-chain variable fragment (scFv).
6. The engineered T cell of claim 5, wherein the anti-LIV1 scFv comprises the same heavy chain variable domain (VH) complementarity determining regions (CDRs) and the same light chain variable domain (VL) CDRs as a reference antibody, wherein the reference antibody comprises (i) a VH set forth as SEQ ID NO: 55 and a VL set forth as SEQ ID NO: 56 or (ii) a VH set forth as SEQ ID NO: 90 and a VL set forth as SEQ ID NO: 88.
7. The engineered T cell of claim 6, wherein the anti-LIV1 scFv comprises the same VH and VL chains as the reference antibody.
8. The engineered T cell of claim 6, wherein the anti-LIV1 scFv comprises the amino acid sequence of any one of SEQ ID NOs: 54, 70, 83, or 86.
9. The engineered T cell of claim 1, wherein the CAR further comprises a CD28 co-stimulatory domain or a 41BB co-stimulatory domain.
10. The engineered T cell of claim 9, wherein the CAR further comprises a CD3 cytoplasmic signaling domain.
11. The engineered T cell of claim 1, wherein the CAR is encoded by the nucleotide sequence of any one of SEQ ID NOs: 49, 51, 104, or 108 or a nucleotide sequence comprising a nucleic acid sequence that is at least 90% identical to SEQ ID NOs: 49, 51, 104, or 108.
12. The engineered T cell of claim 1, wherein the nucleic acid encoding the CAR is inserted into the disrupted TRAC gene.
13. The engineered T cell of claim 2, wherein the disrupted TRAC gene comprises the nucleotide sequence of any one of SEQ ID NOs: 63, 64, 107, or 111, and/or the nucleotide sequence of any one of SEQ ID NOs: 49, 51, 104, or 108.
14. The engineered T cell of claim 4, wherein the disrupted/32M gene comprises at least one nucleotide sequence selected from any one of SEQ ID NOs: 9-14.
15. An engineered T cell comprising:
(i) a disrupted TRAC gene;
(ii) a disrupted β2M gene; and
(iii) a nucleic acid encoding a CAR comprising an anti-LIV1 antigen-binding fragment.
16. The engineered T cell of claim 15, wherein the CAR comprises (a) an ectodomain that comprises an anti-LIV1 antigen-binding fragment, (b) a CD8 transmembrane domain, and (c) an endodomain that comprises a 41BB co-stimulatory domain and a CD3ζ cytoplasmic signaling domain.
17. The engineered T cell of claim 15, wherein the disrupted TRAC gene comprises the nucleic acid encoding the CAR.
18. An engineered T cell comprising:
(i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR comprising (a) an ectodomain that comprises an anti-LIV1 antigen-binding fragment, (b) a CD8 transmembrane domain, and (c) an endodomain that comprises a 41BB co-stimulatory domain and a CD3ζ cytoplasmic signaling domain; and
(ii) a disrupted β2M gene.
19. An engineered T cell comprising:
(i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR comprising an amino acid sequence of any one of SEQ ID NOs: 50, 52, 105, 109, 68 or 66; and
(ii) a disrupted β2M gene.
20. An engineered T cell comprising:
(i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR, wherein the nucleic acid sequence is at least 90% identical to SEQ ID NOs: 49, 51, 104, or 108 and/or encodes a CAR comprising an amino acid sequence of any one of SEQ ID NOs: 50, 52, 105, 109, 68 or 66; and
(ii) a disrupted β2M gene.
21. The engineered T cell of claim 1, wherein the T cell is a human T cell.
22. A population of cells comprising the engineered T cell of claim 1, wherein at least 15% or at least 50% of engineered T cells of the population express the CAR.
23. The population of claim 22, wherein at least 30% of engineered T cells of the population express the CAR.
24. The population of claim 22, wherein at least 70% of engineered T cells of the population express the CAR.
25. The population of claim 22, wherein at least 25% of engineered T cells of the population express the CAR following at least 7 days or at least 14 days of in vitro proliferation.
26. The population of claim 22, wherein at least 50% of engineered T cells of the population do not express a detectable level of T cell receptor (TCR) protein.
27. The population of claim 26, wherein at least 90% of engineered T cells of the population do not express a detectable level of TCR protein.
28. The population of claim 22, wherein at least 50% of engineered T cells of the population do not express a detectable level of β2M protein.
29. The population of claim 28, wherein at least 70% of engineered T cells of the population do not express a detectable level of β2M protein.
30. The population of claim 22, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells that express LIV1, induce cell lysis of at least 10%, at least 25%, or at least 50% of the cancer cells of the population.
31. The population of claim 30, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells that express LIV1, induce cell lysis of at least 70%, at least 80%, or at least 90% of the population of cancer cells.
32. The population of claim 30, wherein engineered T cells of the population, when co-cultured in vitro with a population of cancer cells, secrete IFNγ.
33. The population of claim 30, wherein the ratio of engineered T cells to cancer cells is 1:1 to 2:1.
34. The population of claim 30, wherein the cancer cells comprise sarcoma cells.
35. The population of claim 30, wherein the cancer cells comprise breast cancer cells.
36. The population of claim 22, when administered in vivo to a subject, does not induce toxicity in the subject.
37. A population of cells comprising engineered T cells, wherein the engineered T cells comprise:
(i) a disrupted TRAC gene;
(ii) a disrupted β2M gene; and
(iii) a nucleic acid encoding a CAR comprising an anti-LIV1 antigen-binding fragment.
38. The population of cells of claim 37, wherein the CAR comprises (a) an ectodomain that comprises an anti-LIV1 antigen-binding fragment, (b) a CD8 transmembrane domain, and (c) an endodomain that comprises a 41BB co-stimulatory domain and a CD3ζ cytoplasmic signaling domain.
39. The population of cells of claim 37, wherein the disrupted TRAC gene comprises the nucleic acid encoding the CAR.
40. A population of cells comprising engineered T cells, wherein the engineered T cells comprise:
(i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR comprising (a) an ectodomain that comprises an anti-LIV1 antigen-binding fragment, (b) a CD8 transmembrane domain, and (c) an endodomain that comprises a 41BB co-stimulatory domain and a CD3ζ cytoplasmic signaling domain; and
(ii) a disrupted β2M gene.
41. A population of cells comprising engineered T cells, wherein the engineered T cells comprise:
(i) a disrupted TRAC gene, wherein the disrupted TRAC gene comprises a nucleic acid encoding a CAR, wherein the nucleic acid sequence is at least 90% identical to SEQ ID NOs: 49, 51, 104, or 108 and/or encodes the CAR of SEQ ID NOs: 50, 52, 105, 109, 68 or 66; and
(ii) a disrupted β2M gene.
42. A method comprising administering the population of engineered T cells of claim 22 to a subject.
43. The method of claim 42, wherein the subject is a human subject.
44. The method of claim 43, wherein the subject has a cancer.
45. The method of claim 44, wherein the cancer is selected from the group consisting of: breast cancer, e.g., estrogen receptor-positive breast cancer, prostate cancer, squamous tumors, e.g., of the skin, bladder, lung, cervix, endometrium, head neck, and biliary tract, and/or neuronal tumors.
46. The method of claim 44, wherein the cancer comprises cancer cells expressing LIV1.
47. A method for producing an engineered T cell, the method comprising
(a) delivering to a T cell
(i) a RNA-guided nuclease,
(ii) a gRNA targeting a TRAC gene, and
(iii) a vector comprising a donor template that comprises a nucleic acid encoding a CAR that comprise an ectodomain that binds specifically to LIV1; and
(b) producing an engineered T cell having a disrupted TRAC gene and expressing the CAR.
48. The method of claim 47, wherein the gRNA targeting the TRAC gene comprises the nucleotide sequence of SEQ ID NO: 18 or 19, or targets the nucleotide sequence of SEQ ID NO: 40.
49. The method of claim 47 further comprising delivering to the T cell a gRNA targeting the β2M gene.
50. The method of claim 49, wherein the gRNA targeting the β2M gene comprises the nucleotide sequence of SEQ ID NO: 20 or 21, or targets the nucleotide sequence of SEQ ID NO: 41.
51. The method of claim 47, wherein the ectodomain of the CAR comprises an anti-LIV1 antibody.
52. The method of claim 51, wherein the anti-LIV1 antibody is an anti-LIV1 single-chain variable fragment (scFv).
53. The method of claim 52, wherein the anti-LIV1 scFv comprises the same heavy chain variable domain (VH) complementarity determining regions (CDRs) and the same light chain variable domain (VL) CDRs as a reference antibody, wherein the reference antibody comprises (i) a VH set forth as SEQ ID NO: 55 and a VL set forth as SEQ ID NO: 56, or (ii) a VH set forth as SEQ ID NO: 90 and a VL set forth as SEQ ID NO: 88.
54. The method of claim 53, wherein the anti-LIV1 scFv comprises the same VH and VL chains as the reference antibody.
55. The method of claim 53, wherein the anti-LIV1 scFv comprises the amino acid sequence of any one of SEQ ID NOs: 54, 83, 86 or 70.
56. The method of claim 47, wherein the CAR further comprises a CD28 co-stimulatory domain or a 41BB co-stimulatory domain.
57. The method of claim 56, wherein the CAR further comprises a CD3ζ cytoplasmic signaling domain.
58. The method of claim 47, wherein the CAR is encoded by a nucleotide sequence of any one of SEQ ID NOs: 49, 51, 104, or 108 or a nucleotide sequence comprising a nucleic acid sequence that is at least 90% identical to SEQ ID NOs: 49, 51, 104, or 108.
59. The method of claim 47, wherein the nucleic acid encoding the CAR is flanked by left and right homology arms to the TRAC gene.
60. The method of claim 47, wherein the donor template comprises the nucleotide sequence of any one of SEQ ID NOs: 63, 64, 107, or 111.
61. The method of claim 47, wherein the RNA-guided nuclease is a Cas9 nuclease, optionally a S. pyogenes Cas9 nuclease.
62. An engineered T cell produced by the method of claim 47.
63. A population of cells comprising the engineered T cell of claim 62.
64. A method of treating cancer in a subject, comprising administering to the subject the population of cells of claim 22.
65. The method of claim 64, wherein the cancer is selected from the group consisting of: pancreatic cancer, gastric cancer, ovarian cancer, uterine cancer, breast cancer, prostate cancer, testicular cancer, thyroid cancer, nasopharyngeal cancer, non-small cell lung (NSCLC), glioblastoma, neuronal, soft tissue sarcomas, leukemia, lymphoma, melanoma, colon cancer, colon adenocarcinoma, brain glioblastoma, hepatocellular carcinoma, liver hepatocholangiocarcinoma, osteosarcoma, gastric cancer, esophagus squamous cell carcinoma, advanced stage pancreas cancer, lung adenocarcinoma, lung squamous cell carcinoma, lung small cell cancer, renal carcinoma, and intrahepatic biliary cancer.
66. The method of claim 64, wherein the cancer comprises cancer cells expressing LIV1.
US17/291,198 2018-11-07 2019-11-07 Anti-liv1 immune cell cancer therapy Pending US20210292429A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/291,198 US20210292429A1 (en) 2018-11-07 2019-11-07 Anti-liv1 immune cell cancer therapy

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201862756723P 2018-11-07 2018-11-07
PCT/IB2019/059586 WO2020095249A1 (en) 2018-11-07 2019-11-07 Anti-liv1 immune cell cancer therapy
US17/291,198 US20210292429A1 (en) 2018-11-07 2019-11-07 Anti-liv1 immune cell cancer therapy

Publications (1)

Publication Number Publication Date
US20210292429A1 true US20210292429A1 (en) 2021-09-23

Family

ID=68582073

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/677,267 Pending US20200231699A1 (en) 2018-11-07 2019-11-07 Anti-liv1 immune cell cancer therapy
US17/291,198 Pending US20210292429A1 (en) 2018-11-07 2019-11-07 Anti-liv1 immune cell cancer therapy

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/677,267 Pending US20200231699A1 (en) 2018-11-07 2019-11-07 Anti-liv1 immune cell cancer therapy

Country Status (16)

Country Link
US (2) US20200231699A1 (en)
EP (1) EP3877419A1 (en)
JP (1) JP7471289B2 (en)
KR (1) KR20210089707A (en)
CN (1) CN113015750A (en)
AU (1) AU2019376903A1 (en)
BR (1) BR112021008082A2 (en)
CA (1) CA3118824A1 (en)
CO (1) CO2021006493A2 (en)
EA (1) EA202191257A1 (en)
IL (1) IL282531A (en)
MX (1) MX2021005395A (en)
PH (1) PH12021551036A1 (en)
SG (1) SG11202104523PA (en)
WO (1) WO2020095249A1 (en)
ZA (1) ZA202102740B (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2842895T3 (en) 2010-12-06 2021-07-15 Seagen Inc Humanized LIV-1 Antibodies and Their Use to Treat Cancer
KR102668549B1 (en) * 2014-06-02 2024-05-22 더 유나이티드 스테이츠 오브 어메리카, 애즈 리프리젠티드 바이 더 세크러테리, 디파트먼트 오브 헬쓰 앤드 휴먼 서비씨즈 Chimeric antigen receptors targeting cd-19
MA45324A (en) 2016-03-15 2019-01-23 Seattle Genetics Inc POLYTHERAPY USING ADC-LIV1 AND CHEMOTHERAPEUTIC AGENT
EP4263590A1 (en) 2020-12-21 2023-10-25 Allogene Therapeutics, Inc. Protease-activating cd45-gate car
EP4284918A1 (en) 2021-01-29 2023-12-06 Allogene Therapeutics, Inc. Knockdown or knockout of one or more of tap2, nlrc5, beta2m, trac, rfx5, rfxap and rfxank to mitigate t cell recognition of allogeneic cell products
WO2023111913A1 (en) 2021-12-15 2023-06-22 Crispr Therapeutics Ag Engineered anti-liv1 cell with regnase-1 and/or tgfbrii disruption
US20240042030A1 (en) 2022-07-29 2024-02-08 Allogene Therapeutics, Inc. Engineered cells with reduced gene expression to mitigate immune cell recognition
WO2024165056A1 (en) * 2023-02-07 2024-08-15 LaNova Medicines Limited Antibodies targeting liv-1 and uses thereof

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
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
US6534055B1 (en) 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US7175843B2 (en) 1994-06-03 2007-02-13 Genetics Institute, Llc Methods for selectively stimulating proliferation of 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
US7067318B2 (en) 1995-06-07 2006-06-27 The Regents Of The University Of Michigan Methods for transfecting T cells
GB9710809D0 (en) 1997-05-23 1997-07-23 Medical Res Council Nucleic acid binding proteins
GB9710807D0 (en) 1997-05-23 1997-07-23 Medical Res Council Nucleic acid binding proteins
US6140081A (en) 1998-10-16 2000-10-31 The Scripps Research Institute Zinc finger binding domains for GNN
US6453242B1 (en) 1999-01-12 2002-09-17 Sangamo Biosciences, Inc. Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites
US6534261B1 (en) 1999-01-12 2003-03-18 Sangamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
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
AU2001243288B2 (en) 2000-02-24 2005-11-24 Life Technologies Corporation Simultaneous stimulation and concentration of cells
JP2002060786A (en) 2000-08-23 2002-02-26 Kao Corp Germicidal stainproofing agent for hard surface
AU2002336373A1 (en) 2001-08-20 2003-03-03 The Scripps Research Institute Zinc finger binding domains for cnn
US7888121B2 (en) 2003-08-08 2011-02-15 Sangamo Biosciences, Inc. Methods and compositions for targeted cleavage and recombination
US7972854B2 (en) 2004-02-05 2011-07-05 Sangamo Biosciences, Inc. Methods and compositions for targeted cleavage and recombination
JP2013513389A (en) 2009-12-10 2013-04-22 リージェンツ オブ ザ ユニバーシティ オブ ミネソタ DNA modification mediated by TAL effectors
ES2842895T3 (en) 2010-12-06 2021-07-15 Seagen Inc Humanized LIV-1 Antibodies and Their Use to Treat Cancer
DK3116902T3 (en) * 2014-03-11 2020-04-06 Cellectis Method for generating T cells compatible for allogeneic transplantation
ES2983094T3 (en) * 2014-10-31 2024-10-21 Univ Pennsylvania Alteration of gene expression in CAR-T cells and their uses
JP7128741B2 (en) * 2015-12-18 2022-08-31 サンガモ セラピューティクス, インコーポレイテッド Targeted disruption of T-cell receptors
MA45324A (en) 2016-03-15 2019-01-23 Seattle Genetics Inc POLYTHERAPY USING ADC-LIV1 AND CHEMOTHERAPEUTIC AGENT
SG11201900772YA (en) * 2016-08-03 2019-02-27 John Dipersio Gene editing of car-t cells for the treatment of t cell malignancies with chimeric antigen receptors
WO2018073393A2 (en) 2016-10-19 2018-04-26 Cellectis Tal-effector nuclease (talen) -modified allogenic cells suitable for therapy
US20230138915A1 (en) * 2016-10-19 2023-05-04 Cellectis TAL- effector nuclease (TALEN) -MODIFIED ALLOGENIC CELLS SUITABLE FOR THERAPY
JP2020500530A (en) * 2016-12-02 2020-01-16 ユニバーシティ オブ サザン カリフォルニア Synthetic immune receptors and methods of use

Also Published As

Publication number Publication date
MX2021005395A (en) 2021-09-14
EA202191257A1 (en) 2021-12-06
CO2021006493A2 (en) 2021-05-31
JP2022513586A (en) 2022-02-09
CA3118824A1 (en) 2020-05-14
AU2019376903A1 (en) 2021-05-20
SG11202104523PA (en) 2021-05-28
KR20210089707A (en) 2021-07-16
EP3877419A1 (en) 2021-09-15
IL282531A (en) 2021-06-30
PH12021551036A1 (en) 2022-01-03
WO2020095249A1 (en) 2020-05-14
CN113015750A (en) 2021-06-22
BR112021008082A2 (en) 2021-08-10
ZA202102740B (en) 2022-10-26
US20200231699A1 (en) 2020-07-23
JP7471289B2 (en) 2024-04-19

Similar Documents

Publication Publication Date Title
US11649438B2 (en) Methods and compositions for treating cancer
US20210292429A1 (en) Anti-liv1 immune cell cancer therapy
US20220226375A1 (en) Anti-cd33 immune cell cancer therapy
JP2020537515A (en) HPV-specific binding molecule
US20200140815A1 (en) Anti-ptk7 immune cell cancer therapy
US11926676B2 (en) Masked chimeric antigen receptor specific to tyrosine-protein kinase like 7 (PTK7) and immune cells expressing such
CN116802203A (en) Cells expressing chimeric receptors from modified constant CD3 immunoglobulin superfamily chain loci, related polynucleotides and methods
WO2023111913A1 (en) Engineered anti-liv1 cell with regnase-1 and/or tgfbrii disruption
WO2024062388A2 (en) Genetically engineered immune cells expressing chimeric antigen receptor targeting cd20
WO2024192108A1 (en) Genetically modified car t cells and methods of making and using the same
WO2023084399A1 (en) Genetically engineered immune cells expressing masked chimeric antigen receptors specific to protein tyrosine kinase 7
WO2024003786A1 (en) Chimeric antigen receptor targeting gpc-3 and immune cells expressing such for therapeutic uses

Legal Events

Date Code Title Description
AS Assignment

Owner name: CRISPR THERAPEUTICS AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TERRETT, JONATHAN ALEXANDER;SAGERT, JASON;SIGNING DATES FROM 20200304 TO 20210416;REEL/FRAME:057217/0553

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

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

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