WO2023111913A1 - Cellule anti-liv1 modifiée avec lyse de régnase-1 et/ou tgfbrii - Google Patents

Cellule anti-liv1 modifiée avec lyse de régnase-1 et/ou tgfbrii Download PDF

Info

Publication number
WO2023111913A1
WO2023111913A1 PCT/IB2022/062244 IB2022062244W WO2023111913A1 WO 2023111913 A1 WO2023111913 A1 WO 2023111913A1 IB 2022062244 W IB2022062244 W IB 2022062244W WO 2023111913 A1 WO2023111913 A1 WO 2023111913A1
Authority
WO
WIPO (PCT)
Prior art keywords
gene
cells
engineered
car
cell
Prior art date
Application number
PCT/IB2022/062244
Other languages
English (en)
Inventor
Demetrios Kalaitzidis
Lauren Cari ZAKAS
Jonathan Alexander Terrett
Jason Sagert
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
Publication of WO2023111913A1 publication Critical patent/WO2023111913A1/fr

Links

Classifications

    • 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
    • A61K39/461Cellular 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
    • A61K39/464Cellular 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
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • 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]

Definitions

  • the present disclosure relates generally to the field of engineered cells, for example, engineered immune cells.
  • Chimeric antigen receptor (CAR) T-cell therapy uses genetically engineered T cells to specifically and efficiently target and kill cancer cells.
  • T cells can be engineered to express CARs on their surface by introducing CARs into the T cells using a gene editing technology such as CRISPR/Cas9 gene editing technology.
  • CRISPR/Cas9 gene editing technology such as CRISPR/Cas9 gene editing technology.
  • Described herein includes an engineered T cell, where the engineered T cell comprises, in some embodiments, a nucleic acid encoding a chimeric antigen receptor (CAR), and at least one of (i) a disrupted Regnase-1 (Reg ) gene, and (ii) a disrupted Transforming Growth Factor Beta Receptor II (TGFBRII) gene.
  • CAR anti-LIVl CAR
  • the CAR comprises (1) an ectodomain that binds specifically to LIV1, which may be an anti-LIVl antigen-binding fragment.
  • TGFBRII Transforming Growth Factor Beta Receptor II
  • the engineered T cell(s) can comprise both (i) and (ii).
  • the engineered T cell(s) may further comprise a disrupted T cell receptor alpha chain constant region (TRAC) gene, a disrupted beta-2-microglobulin (J32M) gene, or both.
  • the engineered T cell(s) further comprise the disrupted TRAC gene.
  • the engineered T cell(s) further comprise both the disrupted TRAC gene and the disrupted /32M gene.
  • the ectodomain of the CAR comprises an anti-LIVl antigenbinding fragment, which may comprise an anti-LIVl antibody.
  • the anti-LIVl antibody can be, for example, an anti-LIVl single-chain variable fragment (scFv).
  • the anti-LIVl 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.
  • the reference antibody comprises (a) a VH comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence of SEQ ID NO: 533 and a VL comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence of SEQ ID NO: 534.
  • the reference antibody comprises (a) a VH comprising the amino acid sequence of SEQ ID NO: 533 and (b) a VL comprising the amino acid sequence of SEQ ID NO: 534.
  • the reference antibody comprises (a) a VH comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence of SEQ ID NO: 568 and a VL comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence of SEQ ID NO: 566.
  • the reference antibody comprises (a) a VH comprising the amino acid sequence of SEQ ID NO: 568 and (b) a VL comprising the amino acid sequence of SEQ ID NO: 566.
  • the anti-LIVl scFv may comprise any of the scFv sequences provide in Sequence Tables 29 and 32 below.
  • the anti-LIVl scFv comprises an amino acid sequence having at least 90% sequence identity to the amino acid sequence of any one of SEQ ID NOs: 532, 548, 561, and 564.
  • the anti-LIVl scFv comprises an amino acid sequence having the amino acid sequence of any one of SEQ ID NOs: 532, 548, 561, and 564.
  • Any of the anti-LIVl CARs disclosed herein may further comprise a CD28 costimulatory domain or a 4 IBB co-stimulatory domain.
  • the anti-LIVl CAR may further comprise a CD3( ⁇ cytoplasmic signaling domain, a CD8 transmembrane domain, or both.
  • the anti-LIVl CAR may be any CAR constructs provided in Tables 28, 29, 31, and 32 below.
  • the anti-LIVl CAR may comprise the amino acid sequence of SEQ ID NO: 528 (with signal peptide) or SEQ ID NO: 600 (with no signal peptide).
  • the anti-LIVl CAR may comprise the amino acid sequence of SEQ ID NO: 530 (with signal peptide) or SEQ ID NO: 601 (with no signal peptide).
  • the anti-LIVl CAR may comprise the amino acid sequence of SEQ ID NO: 583 (with signal peptide) or SEQ ID NO: 570 (with no signal peptide).
  • the anti-LIVl CAR may comprise the amino acid sequence of SEQ ID NO: 587 (with signal peptide) or SEQ ID NO: 571 (with no signal peptide). In some embodiments, the anti-LIVl CAR may be encoded by a nucleic acid sequence that is at least 90% identical to SEQ ID NOs: 527, 529, 582, or 586. In some examples, the anti-LIVl CAR may be encoded by a nucleic acid comprising the nucleotide sequence of SEQ ID NOs: 527, 529, 582, or 586.
  • the nucleic acid encoding the anti-LIVl CAR is inserted into a genomic site of interest, e.g., the disrupted Regl gene, the disrupted TGFBRJI gene, the disrupted TRAC gene, or the disrupted /32M gene.
  • the nucleic acid encoding the CAR is inserted into the disrupted TRAC gene.
  • the nucleic acid encoding the CAR may replace the fragment of SEQ ID NO: 69 in the TRAC gene.
  • the disrupted TRAC gene may comprise the nucleotide sequence of any one of SEQ ID NOs: 541, 542, 585, and 589.
  • the disrupted Regl gene comprises a nucleotide sequence listed in Sequence Table 10, 12, 13, or 17.
  • the disrupted /32M comprises a nucleotide sequence listed in Sequence Table 4.
  • the disrupted TRAC gene may comprise a nucleotide sequence listed in Sequence Table 3.
  • the disrupted Regl gene is genetically edited in exon 2 and/or exon 4.
  • the disrupted TGFBRTI gene is genetically edited in exon 1, exon 2, exon 3, exon 4, or exon 5.
  • the disrupted TGFBRII gene is genetically edited in exon 4 or exon 5.
  • the disrupted Regl gene, the disrupted TGFBRII gene, the disrupted TRAC gene, and/or the disrupted /32M gene are genetically edited by a CRISPR/Cas-mediated gene editing system.
  • the CRISPR/Cas- mediated gene editing system comprises a guide RNA (gRNA) targeting a site in the TRAC gene that comprises SEQ ID NO: 69.
  • the CRISPR/Cas-mediated gene editing system comprises a guide RNA (gRNA) targeting a site in the Regl gene that comprises a nucleotide sequence selected from the group consisting of SEQ ID NOs: 320, 322, 323, and 327.
  • the CRISPR/Cas-mediated gene editing system comprises a guide RNA (gRNA) targeting a site in the TGFBRTI gene that comprises a nucleotide sequence of SEQ ID NOs: 269, 275, 281, 287, 293, 299, 305, 311, or 317.
  • the gRNA targeting the Regl gene comprises a nucleotide sequence of SEQ ID NOs: 22, 30, 34, or 50.
  • the gRNA targeting the TGFBRTI gene comprises a nucleotide sequence of SEQ ID NOs: 270, 300, 306, or 312.
  • the gRNA targeting the TRAC gene comprises the nucleotide sequence of SEQ ID NO: 61, for example, comprising the nucleotide sequence of SEQ ID NO: 59 (unmodified or modified such as that set forth in SEQ ID NO: 58).
  • the gRNA targeting the 02M gene comprises the nucleotide sequence of SEQ ID NO: 65, for example, comprising the nucleotide sequence of SEQ ID NO: 63 (unmodified or modified such as that set forth in SEQ ID NO: 62).
  • the engineered T cell or the population of engineered T cells may comprise: (i) a disrupted TRAC gene comprising a nucleic acid encoding a chimeric antigen receptor (CAR) that binds LIV1 (anti-LIVl CAR), (ii) a disrupted Regnase-1 (Regl gene; and (iii) a disrupted Transforming Growth Factor Beta Receptor II (TGFBRTI) gene.
  • the LIV1 CAR comprises an anti-LIVl scFv that comprises the VH of SEQ ID NO: 568 and the VL of SEQ ID NO: 566.
  • the anti-LIVl scFv comprises the amino acid sequence of SEQ ID NO: 561.
  • the anti-LIVl CAR comprises the amino acid sequence of SEQ ID NO: 587 (with signal peptide) or 571 (with no signal peptide) (CTX-975 shown in Table 32 below).
  • the disrupted TRAC gene in the engineered T cell pr the population of engineered T cells may comprise the nucleotide sequence of SEQ ID NO: 585.
  • the engineered T cells described herein can be, for example, a mammalian cell (e.g., a human T cell).
  • the engineered T cells can be derived from one or more healthy human donors.
  • the population of engineered T cells as described herein may contain at least 15%, 30%, 50% or 70% of the engineered T cells that express the anti-LIVl CAR.
  • at least 50% (e.g., 60%, 70%, 80%, 90% or above) of the engineered T cells in the population as described herein do not express a detectable level of T cell receptor (TCR) protein.
  • TCR T cell receptor
  • At least 30% (e.g., 40%, 50%, 60%, 70%, 80%, or above) of the engineered T cells in the population as described herein do not express a detectable level of the Regl protein. In some examples, at least 30% (e.g., 40%, 50%, 60%, 70%, 80%, or above) of the engineered T cells in the population as described herein do not express a detectable level of the TGFBRII protein. In some examples, at least 30% (e.g., 40%, 50%, 60%, or above) of the engineered T cells in the population as described herein do not express a detectable level of the B2M protein.
  • the 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%, 25%, 50%, 70%, 80% or 90% of the cancer cells of the population. In some embodiments, the engineered T cells of the population of cells, when co-cultured in vitro with a population of cancer cells that express LIV1, reduces at least 10%, 25%, 50%, 70%, 80% or 90% of the population of cancer cells (e.g., breast cancer cells). In some embodiments, at least 50% of the engineered T cells of the population do not express a detectable level of T cell receptor (TCR) protein.
  • TCR T cell receptor
  • the present disclosure provides a method of producing a population of engineered T cells as disclosed herein.
  • the method can, in some embodiments, comprises: (a) providing a plurality of cells, wherein the plurality of cells are T cells or precursor cells thereof; (b) delivering to the plurality of cells a nucleic acid encoding a chimeric antigen receptor (CAR) that comprise (1) an ectodomain that binds specifically to LIV1, e.g., an anti- LIVl antigen-binding fragment; (c) genetically editing the Regl gene, the TGFBRII gene, or both; and (d) producing a population of engineered T cells expressing the CAR and having a disrupted Regl gene and/or a disrupted TGFBRII gene.
  • CAR chimeric antigen receptor
  • the anti-LIVl CAR can be any of those disclosed herein, e.g., those described in Tables 28, 29, 31, and 32 below.
  • the anti-LIVl CAR can be CTX-975 or a functional variant thereof, which may have the same VH/VL sequences or the same anti-LIVl scFv sequence.
  • the anti-LIVl CAR can be CTX-971 or a functional variant thereof, which may have the same VH/VL sequences or the same anti-LIVl scFv sequence.
  • step (c) comprises genetically editing both the Regl gene and the TGFBRII gene.
  • step (b) and/or step (c) is performed by one or more CRISPR/Cas-mediated gene editing systems.
  • step (c) is performed by delivering to the plurality of cells an RNA-guided nuclease and a gRNA targeting the Regl gene.
  • the gRNA targeting the Regl gene is specific to an exon of the Regl gene, which may be exon 2 or exon 4.
  • step (c) is performed by delivering to the plurality of cells an RNA-guided nuclease and a gRNA targeting the TGFBRJI gene.
  • the gRNA targeting the TGFBRJI gene is specific to an exon of the TGFBRII gene, which may be exon 1, exon 2, exon 3, exon 4, or exon 5.
  • the gRNA targeting the TGFBRII gene is specific to exon 4.
  • the gRNA targeting the TGFBRJI gene is specific to exon 5.
  • the nucleic acid encoding the anti-LIVl CAR is in an AAV vector.
  • the nucleic acid encoding the CAR comprises a left homology arm and a right homology arm flanking the nucleotide sequence encoding the CAR.
  • the left homology arm and the right homology arm are homologous to a genomic locus of interest in the T cells, allowing for insertion of the nucleic acid into the genomic locus.
  • the genomic locus of interest can be, for example, in the Regl gene, the TGFBRII gene, the TRAC gene, or the /32M gene.
  • the genomic locus is in the TRAC gene, e.g., the site targeted by the gRNA for editing the TRAC gene.
  • step (b) comprising disrupting the TRAC gene by a CRISPR/Cas-mediated gene editing system comprising an RNA-guided nuclease and a gRNA targeting a TRAC gene, and the nucleic acid encoding the CAR is inserted at the site targeted by the gRNA.
  • the gRNA targeting a TRAC gene comprises a nucleotide sequence of SEQ ID NO: 61, for example, comprising the nucleotide sequence of SEQ ID NO: 59 (unmodified or a modified version thereof, such as that set forth in SEQ ID NO: 58).
  • the nucleic acid encoding the CAR is flanked by left and right homology arms to the TRAC gene.
  • the nucleic acid comprises the nucleotide sequence of any one of SEQ ID NOs: 541, 542, 585, and 589.
  • the method comprises genetically editing the /32M gene.
  • genetically editing the /32M gene comprises delivering to plurality of cells a gRNA targeting the /32M gene.
  • the gRNA targeting the /32M gene comprises the nucleotide sequence of SEQ ID NO: 65, for example, comprising the nucleotide sequence of SEQ ID NO: 63 (unmodified or a modified version thereof such as that set forth in SEQ ID NO: 62).
  • the RNA-guided nuclease can, for example, be a Cas9 nuclease (e.g., a S. pyogenes Cas9 nuclease).
  • the present disclosure provides a method, comprising administering to a subject any of the engineered T cells disclosed herein or any population of engineered T cells disclosed herein.
  • the subject can be, for example, a mammal (e.g., a human subject).
  • Also disclosed herein includes an engineered T cell or a population of engineered T cells for use in the treatment of cancer (e.g., a LIV1+ cancer).
  • the engineered T cell or the population of engineered T cells can be any of those as disclosed herein.
  • the subject e.g., a human patient
  • the cancer can be, but is not limited to, pancreatic cancer, gastric cancer, ovarian cancer, uterine cancer, breast cancer, prostate cancer, testicular cancer, thyroid cancer, nasopharyngeal cancer, nonsmall 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, intrahepatic biliary cancer, and a combination thereof.
  • the cancer is a solid tumor cancer, for
  • FIG. 1A shows an amino acid sequence alignment of scFV constructs (VL and VH) - 971 (SEQ ID NO: 532), 973 (SEQ ID NO: 560), 975 (SEQ ID NO: 561), and 977 (SEQ ID NO: 562).
  • FIG. IB shows an amino acid sequence alignment of scFV constructs (VH and VL) - 979 (SEQ ID NO: 548), 974 (SEQ ID NO: 563), 976 (SEQ ID NO: 564), 978 (SEQ ID NO: 565), and 972 (SEQ ID NO: 605).
  • FIG. 2 includes a diagram showing anti-LIVl CAR expression levels measured by flow cytometry and ddPCR.
  • FIG. 3 includes a diagram showing T cell expansion of exemplary engineered anti- LIVl CAR T cell variants over time.
  • FIG. 4A-FIG. 4B show the degree of cytotoxicity against MCF7 (FIG. 4A) and ZR- 75-1 (FIG. 4B) cells that were exhibited by exemplary engineered anti-LIVl CAR T cells and exemplary engineered anti-LIVl CAR TGFBR2/Reg knockout (KO) T cells.
  • FIG. 5A-FIG. 5D show cytokine secretion of exemplary engineered anti-LIVl CAR T cells and exemplary engineered anti-LIVl CAR TGFBR2/Reg KO T cells when cocultured with target MCF7 cells (FIG. 5A, FIG. 5C) and target ZR-75-1 cells (FIG. 5B, FIG. 5D).
  • FIG. 6 includes a diagram showing tumor volume control by exemplary engineered anti-LIVl CAR T cells and exemplary engineered anti-LIVl CAR TGFBR2/Reg KO T cells when co-cultured with MCF7 cell lines.
  • FIG. 7A-FIG. 7B include diagrams showing tumor volume control by exemplary engineered anti-LIVl CAR T cells and exemplary engineered anti-LIVl CAR TGFBR2/Reg KO T cells when co-cultured with MCF7 cell lines.
  • FIG. 8A-FIG. 8D include diagrams showing tumor volume control by exemplary engineered anti-LIVl CAR TGFBR2/Reg KO T cells when co-cultured with MCF7 cell lines.
  • FIG. 9 includes an exemplary diagram showing the LIV1 editing efficiency in the generation of two LIV1 knockout breast cancer cell lines MCF7 and ZR751.
  • FIG. 10A-FIG. 10B include exemplary diagrams showing the degree of cytotoxicity against native MCF7, Livl-2 knockout MCF7 and Livl-4 knockout MCF7 cells that were exhibited by exemplary engineered anti-LIVl CAR T cells (FIG. 10A) and exemplary engineered anti-LIVl CAR TGFBR2/Reg KO T cells (FIG. 10B).
  • FIG. 11A-FIG. 11B include exemplary diagrams showing the degree of cytotoxicity against native ZR751, Livl-2 knockout ZR751 and Livl-4 knockout ZR751 cells that were exhibited by exemplary engineered anti-LIVl CAR T cells (FIG. 11 A) and exemplary engineered anti-LIVl CAR TGFBR2/Reg KO T cells (FIG. 11B).
  • FIG. 12 is a diagram showing anti-tumor activity of various anti-LIVl CAR-T cells as indicated in a mouse model.
  • FIGs. 13A-13B include diagrams showing anti -tumor activity of various anti-LIVl CAR-T cells as indicated in a mouse model at different doses.
  • FIG. 13A at the dose of IxlO 7 CAR-T cells.
  • FIG. 13B at the dose of 2xl0 7 CAR-T cells.
  • 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. LIV-1 protein has been implicated in breast cancer, prostate cancer, squamous tumors, and neuronal tumors.
  • an engineered T cell e.g., anti-LIVl CAR-T cell
  • a method of producing the engineered T cell and a method of treating a subject (e.g., a subject having a cancer) using the engineered T cell or a population of the engineered T cells.
  • the engineered T cell can comprise a nucleic acid encoding a chimeric antigen receptor (CAR), wherein the CAR comprises (1) an ectodomain that binds specifically to LIV1, or (2) an anti-LIVl antigenbinding fragment.
  • CAR chimeric antigen receptor
  • the engineered T cell can also comprise at least one of (i) a disrupted Regnase-1 (Regl) gene, and (ii) a disrupted Transforming Growth Factor Beta Receptor II (TGFBRII) gene.
  • the engineered T cells comprise both a disrupted Regl gene and a disrupted TGFBRII gene.
  • Disclosed herein also includes a population of cells comprising the engineered T cell disclosed herein.
  • Disclosed herein also includes a method of producing an engineered T cell.
  • the method can comprise providing a plurality of cells, wherein the plurality of cells are T cells or precursor cells thereof, delivering to the plurality of cells a nucleic acid encoding a chimeric antigen receptor (CAR) that comprise (1) an ectodomain that binds specifically to LIV1, or (2) an anti-LIVl antigen-binding fragment, genetically editing the /e / gene, the TGFBRII gene, or both; and producing one or more engineered T cells expressing the CAR and having a disrupted Regl gene and/or a disrupted TGFBRII gene.
  • CAR chimeric antigen receptor
  • Disclosed herein also includes a method for the therapeutic uses of the engineered T cell or a population of the engineered T cells herein described.
  • the method can comprise administering to a subject the engineered T cell described herein or the population of engineered T cells described herein.
  • engineered T cells expressing an anti-LIVl CAR and having one or both of disrupted Regl gene and disrupted TGFBRII gene can provide synergistically and/or advantageous enhanced anti -tumor effects as compared with non-engineered T cells or engineered T cells expressing anti-LIVl CAR without having one or both of disrupted Regl gene and disrupted TGFBRII gene.
  • nucleic acid and “polynucleotide” are interchangeable and refer to any nucleic acid, whether composed of phosphodiester linkages or modified linkages such as phosphotriester, phosphoramidate, siloxane, carbonate, carboxymethylester, acetamidate, carbamate, thioether, bridged phosphoramidate, bridged methylene phosphonate, bridged phosphoramidate, bridged phosphoramidate, bridged methylene phosphonate, phosphorothioate, methylphosphonate, phosphorodithioate, bridged phosphorothioate or sultone linkages, and combinations of such linkages.
  • the terms “nucleic acid” and “polynucleotide” also specifically include nucleic acids composed of bases other than the five biologically occurring bases (adenine, guanine, thymine, cytosine and uracil).
  • naturally occurring and “biologically occurring” as used herein refer to materials which are found in nature or a form of the materials that is found in nature.
  • sequence identity or “identity” in the context of two nucleic acid or polypeptide sequences makes reference to the nucleotide bases or residues in the two sequences that are the same when aligned for maximum correspondence over a specified comparison window.
  • Methods of alignment of sequences for comparison are well known in the art. Various programs and alignment algorithms are described in: Smith & Waterman, Adv. Appl. Math. 2:482, 1981; Needleman & Wunsch, J. Mol. Biol. 48:443, 1970; Pearson & Lipman, Proc. Natl. Acad. Sci.
  • vector can refer to a vehicle for carrying or transferring a nucleic acid.
  • vectors include viral vectors (for example, adenovirus vectors, adeno-associated virus (AAV) vectors, retrovirus vectors, lentiviral vectors, herpes virus vectors, phages, and poxvirus vectors); non-viral vectors such as liposomes, naked DNA, plasmids, cosmids; and the like.
  • viral vectors for example, adenovirus vectors, adeno-associated virus (AAV) vectors, retrovirus vectors, lentiviral vectors, herpes virus vectors, phages, and poxvirus vectors
  • non-viral vectors such as liposomes, naked DNA, plasmids, cosmids; and the like.
  • a “donor” refers 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.
  • 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.
  • Conservative substitutions of amino acids include substitutions made amongst amino acids within the following groups: (a) A - G, S; (b) R H -> N, Q; (j) I -> L, V; (k) L -> I, V; (1) 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.
  • Variants can be prepared according to methods for altering polypeptide sequence known to one of 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, New York, 1989, or Current Protocols in Molecular Biology, F.M. Ausubel, et al., eds., John Wiley & Sons, Inc., New York.
  • a CDR can 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.
  • antibody encompasses intact (i.e., full-length) monoclonal antibodies, as well as 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 “disrupted gene” refers to a gene comprising an insertion, deletion or substitution relative to an endogenous gene such that expression of a functional protein from the endogenous gene is reduced or inhibited.
  • “disrupting a gene” refers to a method of inserting, deleting or substituting at least one nucleotide/nucleic acid in an endogenous gene such that expression of a functional protein from the endogenous gene is reduced or inhibited.
  • a “transmembrane domain” refers to a protein structure that is thermodynamically stable in a cell membrane, preferably a eukaryotic cell membrane.
  • Standard techniques can be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection).
  • Enzymatic reactions and purification techniques can be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein.
  • the foregoing techniques and procedures can be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), which is incorporated herein by reference for any purpose.
  • Anti-LIVl Chimeric Antigen Receptors (Anti-LIVl CARs) and T Cells Expressing Such
  • the T cells disclosed herein can be engineered to express 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, which makes it a desirable transmembrane protein for targeting cancer.
  • LIV-1 protein has been implicated in breast cancer, prostate cancer, squamous tumors, and neuronal tumors.
  • An engineered T cell disclosed herein can have nucleic acid encoding a chimeric antigen receptor (CAR).
  • the CAR comprises an ectodomain that binds specifically to LIV1 or an anti-LIVl antigen-binding fragment.
  • the anti-LIVl antigen-binding fragment comprises an anti-LIVl antibody.
  • a chimeric antigen receptor refers to an artificial immune cell receptor that is engineered to recognize and bind to an antigen expressed by undesired cells, for example, disease cells such as cancer cells.
  • a T cell that expresses a CAR polypeptide is referred to as a CAR T cell.
  • a CAR designed for a T cell 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
  • an antigen-recognizing domain e.g., a single chain fragment (scFv) of an antibody or other antibody fragment
  • 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 CAR-T cells the ability to recognize an antigen independent of antigen processing, thus bypassing a major mechanism of tumor escape.
  • CARs when expressed on T-cells, CARs advantageously do not dimerize with endogenous T-cell receptor (TCR) alpha and beta chains.
  • TCR T-cell receptor
  • 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 co-stimulatory domain, e.g., CD28, 4-1BB (41BB), or ICOS, to supply a costimulatory signal.
  • Third-generation CARs contain two costimulatory domains (e.g., a combination of CD27, CD28, 4- IBB, ICOS, or 0X40) fused with the TCR CD3( chain. Maude et al., Blood. 2015; 125(26):4017-4023; Kakarla and Gottschalk, Cancer J. 2014; 20(2): 151 - 155). Any of the various generations of CAR constructs is within the scope of the present disclosure.
  • CARs typically differ in their functional properties.
  • 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- IBB CARs may produce superior proliferation and/or persistence.
  • the CAR can be a first generation CAR. In some embodiments, a CAR is a second generation CAR. In yet some other embodiments, the CAR is a third generation CAR.
  • a CAR is a fusion polypeptide comprising an extracellular domain (ectodomain) that recognizes a target antigen (e.g., a single chain fragment (scFv) of an antibody or other antibody fragment) and an intracellular domain (endodomain) comprising a signaling domain of the T-cell receptor (TCR) complex (e.g., CD3Q and, in most cases, a co-stimulatory domain (Enblad et al., Human Gene Therapy. 2015; 26(8):498- 505).
  • a CAR construct may further comprise a hinge and transmembrane domain between the extracellular domain and the intracellular domain, as well as a signal peptide at the N- terminus for surface expression. Examples of signal peptides include SEQ ID NO: 95 and SEQ ID NO: 96 as provided in Sequence Table 6 below. Other signal peptides may be used.
  • 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.
  • 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.
  • 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.
  • VH heavy chain variable region
  • VL light chain variable region
  • 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-LIVl 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.
  • An antibody can, for example, 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-LIVl 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 (KD) between the antibody and LIV1 is 100 pM to 1 pM, for example, 1 nM to 100 nM.
  • the antigen binding domain specific to LIV1 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.
  • the anti-LIVl antibody for use in constructing the anti-LIVl CAR disclosed herein may comprise VH CDRs that collectively are at least 80% (e.g., about, at least, or at least about 80%, 85%, 90%, 95%, or 98%) identical to the VH CDRs of a reference antibody having a VH of SEQ ID NO: 533 and a VL of SEQ ID NO: 534, or a reference antibody having a VH of SEQ ID NO: 568 and a VL of SEQ ID NO: 566.
  • the antibody can comprise VL CDRs that collectively are at least 80% (e.g., about, at least, or at least about 80%, 85%, 90%, 95%, or 98%) identical to the VL CDRs of the reference antibody.
  • the antibody for use in constructing the anti-LIVl CAR comprises a VH that is at least 80% (e.g., about, at least, or at least about 80%, 85%, 90%, 95%, or 98%) identical to the VH of a reference antibody such as in VH: SEQ ID NO: 533 or 568 or 576; VL: SEQ ID NO: 534 or 566 or 606 and/or a VL variable region that is at least 80% (e.g., about, at least, or at least about 80%, 85%, 90%, 95%, or 98%) identical to the VL variable region of the reference antibody.
  • the antibody for use in constructing the anti-LIVl CAR 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: 533 or 568 or 576; VL: SEQ ID NO: 534 or 566 or 606. “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.
  • the antibody for use in constructing the anti-LIVl CAR comprises 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: 533 or 568 or 576; VL: SEQ ID NO: 534 or 566 or 606.
  • 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: 533 or 568 or 576; VL: SEQ ID NO: 534 or 566 or 606.
  • an antibody can 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 or comprise conservative amino acid residue substitutions.
  • 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, New York, 1989, or Current Protocols in Molecular Biology, F.M. Ausubel, et al., eds., John Wiley & Sons, Inc., New York.
  • VH and VL protein sequences that can be used to create an anti-LIVl scFv include those provided in sequence Tables 29 and 32, all of which are within the scope of the present disclosure.
  • the VH may comprise the amino acid sequence of SEQ ID NO: 533 and the VL may comprise the amino acid sequence of SEQ ID NO: 534 or 606.
  • An anti-LIVl scFv having such VH/VL may comprise the amino acid sequence of SEQ ID NO: 532, 548, or 605.
  • the VH may comprise the amino acid sequence of SEQ ID NO: 576 and the VL may comprise the amino acid sequence of SEQ ID NO: 534.
  • An anti-LIVl scFv having such VH/VL may comprise the amino acid sequence of SEQ ID NO: 560 or 563.
  • the VH may comprise the amino acid sequence of SEQ ID NO: 568 and the VL may comprise the amino acid sequence of SEQ ID NO: 566.
  • An anti-LIVl scFv having such VH/VL may comprise the amino acid sequence of SEQ ID NO: 561 or 564.
  • the VH may comprise the amino acid sequence of SEQ ID NO: 576 and the VL may comprise the amino acid sequence of SEQ ID NO: 566.
  • An anti-LIVl scFv having such VH/VL may comprise the amino acid sequence of SEQ ID NO: 562 or 565.
  • the scFv of the present disclosure is humanized. In some embodiments, the scFv is fully human. In some embodiments, the scFv is a chimera (e.g., of mouse and human sequence). In some embodiments, the scFv is an anti-LIVl scFv (binds specifically to LIV1).
  • anti-LIVl scFv proteins include the amino acid sequence of any one of SEQ ID NOs: 532, 548, 560, 561, 562, 563, 564, and 565. Other scFv proteins can also 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 SEQ ID NO: 95 and SEQ ID NO: 96. Other signal peptides may be used.
  • a spacer domain or hinge domain can be 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 can also be used.
  • the CAR polypeptide disclosed herein can contain a transmembrane domain, which can be a hydrophobic alpha helix that spans the membrane.
  • a “transmembrane domain” can be thermodynamically stable in a cell membrane, preferably a eukaryotic cell membrane. The transmembrane domain can provide stability of the CAR containing such.
  • the transmembrane domain of a CAR can be, or comprise, a CD8 transmembrane domain.
  • the transmembrane domain can be, or comprise, a CD28 transmembrane domain.
  • the transmembrane domain is, or comprise, 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 containing the sequence of SEQ ID NO: 97 as provided below in Sequence Table 6 or the sequence of SEQ ID NO: 553 or SEQ ID NO: 555 of Sequence Table 29.
  • Other transmembrane domains can also be used.
  • the endodomain (intracellular signaling domain, e.g., CD3 ⁇ , and optionally one or more co-stimulatory domains) is the functional end of the receptor. Following antigen recognition, receptors cluster and a signal is transmitted to the cell.
  • an endodomain comprises CD3 ⁇ , a cytoplasmic signaling domain of the T cell receptor complex.
  • CD3 ⁇ contains three (3) immunoreceptor tyrosinebased activation motif (ITAM)s, which transmit an activation signal to the T cell after the T cell is engaged with a cognate antigen.
  • ITAM immunoreceptor tyrosinebased activation motif
  • CD3 ⁇ provides a primary T cell activation signal but not a fully competent activation signal, which requires a co-stimulatory signaling.
  • CD28 and/or 4-1BB may be used with CD3-zeta (CD3Q to transmit a proliferative/ survival signal.
  • the CAR polypeptides disclosed herein may further comprise one or more co-stimulatory signaling domains.
  • the co-stimulatory domains of CD28 and/or 4-1BB may be used to transmit a full proliferative/survival signal, together with the primary signaling mediated by CD3 ⁇ .
  • the CAR disclosed herein comprises a CD28 co- stimulatory molecule.
  • the CAR disclosed herein comprises a 4-1BB co-stimulatory molecule.
  • a CAR includes a CD3( ⁇ signaling domain and a CD28 co-stimulatory domain.
  • a CAR includes a CD3 ⁇ signaling domain and 4- IBB co-stimulatory domain.
  • a CAR includes a CD3( ⁇ signaling domain, a CD28 co-stimulatory domain, and a 4-1BB co- stimulatory domain.
  • Sequence Table 6 provides examples of signaling domains derived from 4-1BB, CD28 and CD3-zeta that can be used herein.
  • engineered cells e.g., engineered cells in a population of cells
  • CAR engineered cells
  • at least 15%, 30%, 50% or 70% of the engineered T cells of the population express the CAR.
  • Immune cells e.g., T cells
  • the antigen-binding extracellular domain comprises an anti-LIVl antigenbinding fragment which can, for example, comprise an anti-LIVl antibody (e.g., anti-LIVl scFv).
  • anti-LIVl antigenbinding fragment which can, for example, comprise an anti-LIVl antibody (e.g., anti-LIVl scFv).
  • anti-LIVl antibody e.g., anti-LIVl scFv
  • Exemplary anti-LIVl CAR constructs, and functional elements thereof are provided in Tables 29 and 32 below. Any of such constructs, as well as functional elements thereof, is within the scope of the present disclosure.
  • the anti-LIVl antibody is an anti-LIVl scFv encoded by a sequence comprising or consisting of any one of SEQ ID NOs: 531, 547, 575, 580, 584, 588, 592, and 596, or an anti-LIVl scFv encoded by a nucleic acid sequence comprising or consisting of a sequence about, at least or at least about 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to one of SEQ ID NOs: 531,
  • the anti-LIVl antibody is an anti-LIVl scFv having a sequence comprising or consisting of any one of SEQ ID NOs: 532,
  • an anti-LIVl scFv comprising or consisting of a sequence having 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to one of SEQ ID NOs: 532, 548, 560, 561, 562, 563, 564, and 565.
  • the anti-LIVl antibody is an anti-LIVl scFv comprising a VH comprising or consisting of an amino acid sequence of any one of SEQ ID NO: 533, 568 and 576, or an anti-LIVl scFv comprising a VH having an amino acid sequence comprising or consisting of a sequence having 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to one of SEQ ID NOs: 533, 568 and 576.
  • the anti-LIVl antibody is an anti-LIVl scFv comprising a VL comprising or consisting the amino acid sequence of any one of SEQ ID NO: 534, 566 and 606, or an anti- LIVl scFv comprising a VL having an amino acid sequence comprising or consisting of a sequence having 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to any one of SEQ ID NOs: 534, 566 and 606.
  • a CAR comprising an anti-LIVl antibody is encoded by the sequence of any one of SEQ ID NOs: 527, 529, 543, 545, 573, 578, 582, 586, 590, and 594.
  • a CAR comprising an anti-LIV7 antibody is encoded by a sequence comprising a nucleic acid that is about, at least or at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NOs: 527, 529, 543, 545, 573, 578, 582, 586, 590, or 594.
  • a CAR comprising an anti-LIVl antibody comprises or consists of the sequence of any one of SEQ ID NOs: 527, 529, 543, 545, 573, 578, 582, 586, 590, or 594.
  • an anti-LIVl CAR comprise a CAR construct selected from CTX-971 CAR, CTX-971b CAR, CTX-972 CAR, and CTX-972b CAR listed in Sequence Table 28 with the sequences provided in Sequence Table 29.
  • an anti- LIVl CAR comprise a CAR construct selected from CTX-973 CAR, CTX-974 CAR, CTX- 975 CAR, CTX-976 CAR, CTX-977 CAR, CTX-978 CAR, CTX-979 CAR, or CTX-979b CAR listed in Sequence Table 31 with the sequences provided in Sequence Table 32.
  • the nucleic acid encoding the anti-LIVl CARs are inserted in the TRAC gene.
  • the disrupted TRAC gene in the anti-LIVl CAR cells comprises, or consists of, a nucleotide sequence of SEQ ID NO: 527, 529, 543, 545, 573, 578, 582, 586, 590, or 594.
  • the disrupted TRAC gene in the anti-LIVl CAR cells comprises, or consists of, a nucleotide sequence that is about, at least or at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 527, 529, 543, 545, 573, 578, 582, 586, 590, or 594.
  • the disrupted TRAC gene in the anti-LIVl CAR cells comprises, or consists of, a nucleotide sequence of SEQ ID NO: 527, 573, or 582.
  • the disrupted TRAC gene in the anti- LIV1 CAR cells comprises, or consists of, a sequence that is about, at least or at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 527, 573, or 582.
  • the disrupted TRAC gene in the anti-LIVl CAR cells comprises, or consists of, the nucleotide sequence of SEQ ID NO: 582.
  • the disrupted TRAC gene in the anti-LIVl CAR cells comprises, or consists of, a sequence that is about, at least or at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the nucleotide sequence of SEQ ID NO: 582.
  • the disrupted TRAC gene in the anti-LIVl CAR cells comprises, or consists of, a nucleotide sequence of SEQ ID NO: 541, 542, 549, 550, 577, 581, 585, 589, 593 or 597.
  • the disrupted TRAC gene in the anti-LIVl CAR cells comprises, or consists of, a sequence that is about, at least, or at least about, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 541, 542, 549, 550, 577, 581, 585, 589, 593 or 597.
  • the disrupted TRAC gene in the anti-LIVl CAR cells can comprise a nucleotide sequence of SEQ ID NO: 541, 542, 577, 585 or 589.
  • the disrupted TRAC gene in the anti-LIVl CAR cells comprises, or consists of, a sequence that is about, at least or at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 541, 542, 577, 585 or 589.
  • the disrupted TRAC gene in the anti-LIVl CAR cells comprises, or consists of, the nucleotide sequence of SEQ ID NO: 585.
  • the disrupted TRAC gene in the anti-LIVl CAR cells comprises, or consists of, a sequence that is about, at least or at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 585.
  • a CAR comprising an anti-LIVl antibody comprises an anti- LIVl antibody as described in US Patent No. 9,228,026, and W02020/095249, the relevant disclosures of each of which are incorporated by reference for the subject matter and purpose referenced herein.
  • the engineered T cells can comprise one or more gene edit(s), for example, in one or more gene(s).
  • an engineered T cell can comprise a disrupted T cell receptor alpha chain constant region (TRAC) gene, a disrupted TGFBRTI gene, a disrupted Regl gene, or a combination thereof.
  • an engineered T cell comprises a disrupted TRAC gene and at least one of a disrupted Regl gene and a disrupted TGFBRII gene.
  • an engineered T cell comprises a disrupted TRAC gene, a disrupted Regl gene, and a disrupted TGFBRII gene.
  • an engineered T cell can further comprise one or more of a disrupted /32M gene, a disrupted CD70 gene, and a disrupted PD-1 gene.
  • An engineered T cell can also comprise a disrupted beta-2-microglobulin (J32M) gene, a disrupted programmed cell death-1 (PD-1 or PDCDT) gene, a disrupted CD70 gene, or any combination of two or more of the foregoing disrupted genes.
  • J32M disrupted beta-2-microglobulin
  • PD-1 or PDCDT disrupted programmed cell death-1
  • a cell that comprises a disrupted gene does not express (e.g, at the cell surface) a detectable level (e.g, in an immune assay using an antibody binding to the encoded protein or by flow cytometry) of the protein encoded by the gene.
  • a detectable level e.g, in an immune assay using an antibody binding to the encoded protein or by flow cytometry
  • a cell that does not express a detectable level of the protein can be referred to as a knockout cell.
  • populations of cells in which a certain percentage of the cells has been edited e.g., TRAC, Regl and/or TGFBRTI gene
  • a certain percentage of the cells has been edited
  • at least 50% e.g, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or a number or a range between any two of these values
  • TRAC, Regl and/or TGFBRII knockout cells are TRAC, Regl and/or TGFBRII knockout cells.
  • At least 50% of the cells (e.g., T cells) of the population do not express detectable levels of T cell receptor (TCR) surface protein.
  • TCR T cell receptor
  • 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 can be TRAC knockout cells.
  • at least 50% of the cells (e.g. T cells) of the population do not express detectable levels of Regl and/or TGFBRII 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 can be Regl and/or TGFBRII knockout cells.
  • an engineered T cell herein disclosed 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.
  • an edited TRAC gene can comprise a nucleotide sequence selected from the following sequences in Sequence Table 3. It is known to those skilled in the art that different nucleotide sequences in an edited gene such as an edited TRAC gene (e.g., those in Sequence Table 3) may be generated by a single gRNA such as the one listed in Sequence Table 2 (TA-1). Non-limiting examples of modified and unmodified TRAC gRNA sequences that can be used to create a genomic disruption in the TRAC gene are listed in Sequence Table 2 (e.g., SEQ ID NOs: 58 and 59). See also International Application published as W02019215500, which is incorporated herein by reference.
  • gRNA sequences can 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.
  • 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: 69 (AGAGCAACAGTGCTGTGGCC) in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion comprising SEQ ID NO: 69 (AGAGCAACAGTGCTGTGGCC) in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion of SEQ ID NO: 68 in the TRAC gene relative to unmodified T cells. In some embodiments, an engineered T cell comprises a deletion comprising SEQ ID NO: 68 in the TRAC gene relative to unmodified T cells.
  • the TRAC gene editing is also described in International Patent Application published as W02020095249, the relevant disclosures of which are incorporated by reference for the subject matter and purpose referenced herein.
  • the nucleic acid encoding any of the anti-LIVl CAR may be inserted into the disrupted TRAC gene.
  • Such a disrupted TRAC gene may comprise the coding sequence of the anti-LIVl CAR.
  • the engineered T cells can comprise a disrupted gene involved in mRNA decay such as Regl.
  • Regl contains a zinc finger motif, binds RNA and exhibits ribonuclease activity. Regl plays roles in both immune and non-immune cells and its expression can be rapidly induced under diverse conditions including microbial infections, treatment with inflammatory cytokines and chemical or mechanical stimulation.
  • Human Regl gene is located on chromosome lp34.3. Additional information can be found in GenBank under Gene ID: 80149.
  • the engineered T cells can comprise a disrupted Regl gene such that the expression of Regl in the T cells is substantially reduced or eliminated completely.
  • the disrupted Regl gene can comprise one or more genetic edits at one or more suitable target sites (e.g., in coding regions or in non-coding regulatory regions such as promoter regions) that disrupt expression of the Regl gene.
  • suitable target sites e.g., in coding regions or in non-coding regulatory regions such as promoter regions
  • target sites can be identified based on the gene editing approach for use in making the genetically engineered T cells.
  • Exemplary target sites for the genetic edits may include exon 1, exon 2, exon 3, exon 4, exon 5, exon 6, or a combination thereof.
  • one or more genetic editing may occur in exon 2 or exon 4.
  • Such genetic editing can be induced by the CRISPR/Cas technology using a suitable guide RNA, for example, those listed in Sequence Table 1.
  • a suitable guide RNA for example, those listed in Sequence Table 1.
  • the resultant edited Regl gene using a gRNA listed in Sequence Table 1 can comprise one or more edited sequences provided in Sequence Tables 8-17 below.
  • the Regl gene editing is also described in WO2022/064428, the relevant disclosures of which are incorporated by reference for the subject matter and purpose referenced herein.
  • the engineered T cells described herein can comprise a disrupted TGFBRII gene, which encodes Transforming Growth Factor Receptor Type II (TGFBRII).
  • TGFBRII receptors are a family of serine/threonine kinase receptors involved in the TGF0 signaling pathway. These receptors bind growth factor and cytokine signaling proteins in the TGF0 family, for example, TGFPs (TGFpi, TGFP2, and TGFP3), bone morphogenetic proteins (BMPs), growth differentiation factors (GDFs), activin and inhibin, myostatin, anti -Mullerian hormone (AMH), and NODAL.
  • TGFBRII receptors are a family of serine/threonine kinase receptors involved in the TGF0 signaling pathway. These receptors bind growth factor and cytokine signaling proteins in the TGF0 family, for example, TGFPs (TGFpi, TGFP2, and TGFP3)
  • the engineered T cells can comprise a disrupted TGFBRII gene such that the expression of TGFBRII in the T cells is substantially reduced or eliminated completely.
  • the disrupted TGFBRII gene can comprise one or more genetic edits at one or more suitable target sites (e.g., in coding regions or in non-coding regulatory regions such as promoter regions) that disrupt expression of the TGFBRTI gene.
  • suitable target sites e.g., in coding regions or in non-coding regulatory regions such as promoter regions
  • target sites can be identified based on the gene editing approach for use in making the genetically engineered T cells.
  • Exemplary target sites for the genetic edits include exon 1, exon 2, exon 3, exon 4, exon 5, or a combination thereof.
  • one or more genetic editing can occur in exon 4 and/or exon 5.
  • Such genetic editing can be induced by a gene editing technology, (e.g., the CRISPR/Cas technology) using a suitable guide RNA, for example, those listed in Sequence Table 18.
  • a gene editing technology e.g., the CRISPR/Cas technology
  • the resultant edited TGFBRTI gene using a gRNA listed in Sequence Table 18 can comprise one or more edited sequences provided in Sequence Tables 19-27 below.
  • the TGFBRII gene editing is also described in WO2022/064428, the relevant disclosures of which are incorporated by reference for the subject matter and purpose referenced herein.
  • the genetically engineered T cells disclosed herein can comprise a disrupted /32M gene.
  • 02M is a common (invariant) component of MHC I complexes. Disrupting its expression by gene editing can prevent host versus therapeutic allogeneic T cells responses leading to increased allogeneic T cell persistence. In some embodiments, expression of the endogenous /32M gene is eliminated to prevent a host-versus- graft response.
  • an edited /32M gene can comprise a nucleotide sequence selected from the following sequences in Sequence Table 4. It is known to those skilled in the art that different nucleotide sequences in an edited gene such as an edited /32M gene (e.g., those in Sequence Table 4) can be generated by a single gRNA such as the one listed in Sequence Table 2 (02M-1). Non-limiting examples of modified and unmodified 02M gRNA sequences that can be used herein to create a genomic disruption in the /32M gene are listed include, for example, SEQ ID NOs: 62 and 63). See also International Application published as W02019215500, the relevant disclosures of which are incorporated by reference for the subject matter and purposes referenced herein.
  • gRNA sequences can be designed using the ?2A7 gene sequence located on Chromosome 15 (GRCh38 coordinates: Chromosome 15: 44,711,477-44,718,877 ; Ensembl: ENSG00000166710).
  • gRNAs targeting the 02M genomic region create Indels in the /32M 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 02M 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 02M 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 02M surface protein.
  • the genetically engineered T cells disclosed herein can comprise one or more additional gene edits (e.g., gene knock-in or knock-out) to improve T cell function.
  • additional gene edits e.g., gene knock-in or knock-out
  • knock-in or knock-out genes to improve target cell lysis
  • knock-in or knock-out genes to enhance performance of therapeutic T cells such as CAR-T cells prepared from the genetically engineered T cells.
  • the engineered T cell herein describe can further comprise a disrupted PD-1 gene.
  • 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 can lead to more persistent and/or potent therapeutic T cell responses and/or reduce immune suppression in a subject.
  • 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 Sequence Table 2 (e.g., SEQ ID NOs: 500 and 501). See also International Application published as W02019215500, the relevant disclosures of which are incorporated by reference for the subject matter and purposes referenced herein.
  • 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.
  • An engineered T cell disclosed herein can, in some embodiments, comprise a disrupted gene involved in cell exhaustion (e.g., T cell exhaustion). Genes involved in cell exhaustion refer to those that either positively regulate or negatively regulate this biological process. In some embodiments, an engineered T cell comprises a disrupted Cluster of Differentiation 70 (CD70) gene.
  • CD70 Cluster of Differentiation 70
  • 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 antitumor 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.
  • the gRNA targeting CD70 listed in Sequence Table 2 can be used for disrupting the CD70 gene via CRISPR/Cas9 gene editing.
  • an edited CD70 gene can comprise a nucleotide sequence selected from the following sequences in Sequence Table 5.
  • Other gRNA sequences can be designed using the CD70 gene sequence located on Chromosome 19 (GRCh38 coordinates: Chromosome 19: 6,583,183-6,604,103; Ensembl: ENSG00000125726).
  • the engineered T cells can comprise a disrupted CD70 gene such that the expression of CD70 in the T cells is substantially reduced or eliminated completely.
  • the disrupted CD70 gene can comprise one or more genetic edits at one or more suitable target sites (e.g., in coding regions or in non-coding regulatory regions such as promoter regions) that disrupt expression of the CD70 gene.
  • suitable target sites e.g., in coding regions or in non-coding regulatory regions such as promoter regions
  • target sites can be identified based on the gene editing approach for use in making the genetically engineered T cells.
  • 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.
  • Provided herein also includes a population of engineered immune cells (e.g., T cells such as human T cells) comprising a disrupted Regl gene, a disrupted TGFBRII gene, or a combination thereof, and expressing an anti-LIVl CAR.
  • the anti-LIVl CAR can comprise an ectodomain that binds specifically to LIV1 or an anti-LIVl antigen-binding fragment (e.g., an anti-LIVl antibody or a fragment thereof).
  • the population of engineered immune cells (e.g., T cells such as human T cells) comprise both a disrupted Regl gene and a disrupted TGFBRII gene, and express an anti-LIVl CAR, e.g., those disclosed herein.
  • the anti-LIVl CAR-T cells disclosed herein which express any of the anti-LIVl CAR disclosed herein (e.g., the anti-LIVl CAR comprising the amino acid sequence provided in Tables 29 and 32), can also comprise a disrupted TRAC gene and/or a disrupted /32M gene as also disclosed herein.
  • the engineered T cell is a human T cell.
  • the anti-LIVl CAR T cell comprise a construct of CTX-971 CAR, CTX-971b CAR, CTX-972 CAR, or CTX-972b CAR of Sequence Table 28 with the sequences provided in Sequence Table 29.
  • the anti-LIVl CAR T cell comprise a construct of CTX-973 CAR, CTX-974 CAR, CTX-975 CAR, CTX-976 CAR, CTX-977 CAR, CTX-978 CAR, CTX-979 CAR, or CTX-979b CAR of Sequence Table 31 with the sequences provided in Sequence Table 32.
  • the nucleic acid encoding the anti-LIVl CAR is inserted in the TRAC gene.
  • the disrupted TRAC gene in the anti-LIVl CAR cells can comprise a nucleotide sequence of SEQ ID NO: 527, 529, 543, 545, 573, 578, 582, 586, 590, or 594. In some embodiments, the disrupted TRAC gene in the anti-LIVl CAR cells can comprise a nucleotide sequence of SEQ ID NO: 527, 573, or 582. In some embodiments, the disrupted TRAC gene in the anti-LIVl CAR cells can comprise the nucleotide sequence of SEQ ID NO: 582.
  • the population of engineered T cells are anti-LIVl CAR cells that further comprise a disrupted Reg-1 gene.
  • anti-LIVl CAR cells are LIV1 directed T cells having disrupted TRAC gene and /32M gene.
  • the nucleic acid encoding the anti-LIVl CAR can be inserted in the disrupted TRAC gene at the site of SEQ ID NO: 69, which is replaced by the nucleic acid encoding the anti-LIVl CAR, thereby disrupting expression of the TRAC gene.
  • the disrupted TRAC gene in the anti-LIVl CAR cells can comprise the nucleotide sequence of SEQ ID NO: 582.
  • Anti-LIVl CAR T cells that comprise a disrupted Regl gene can be produced via ex vivo genetic modification using the CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR associated protein 9) technology to disrupt targeted genes (Regl, optionally TRAC and/or /32M genes), and adeno-associated virus (AAV) transduction to deliver the anti-LIVl CAR construct.
  • CRISPR/Cas9 Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR associated protein 9
  • CRISPR-Cas9-mediated gene editing involves at least a sgRNA targeting Regl (e.g., REG1-Z03 (SEQ ID NO: 22), REG1-Z05 (SEQ ID NO: 30), REG1-Z06 (SEQ ID NO: 34) or REG1-Z10 (SEQ ID NO: 50)), and optionally TA-1 sgRNA (SEQ ID NO: 59), which targets the TRAC locus, and 02M-1 sgRNA (SEQ ID NO: 63), which targets the f>2M locus.
  • a sgRNA targeting Regl e.g., REG1-Z03 (SEQ ID NO: 22), REG1-Z05 (SEQ ID NO: 30), REG1-Z06 (SEQ ID NO: 34) or REG1-Z10 (SEQ ID NO: 50)
  • TA-1 sgRNA SEQ ID NO: 59
  • 02M-1 sgRNA SEQ ID NO: 63
  • Anti-LIVl CAR T cells that comprise a disrupted TGFBRTI gene can be produced via ex vivo genetic modification using the CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR associated protein 9) technology to disrupt targeted genes TGFBRTI, optionally TRAC and/or /32M genes), and adeno-associated virus (AAV) transduction to deliver the anti-LIVl CAR construct.
  • CRISPR/Cas9 Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR associated protein 9
  • AAV adeno-associated virus
  • CRISPR-Cas9-mediated gene editing involves at least a sgRNA targeting TGFBRTI e.g., those listed in Sequence Table 18, e.g., TGFBRII EX1 T2, TGFBRII EX4 T1, TGFBRII EX4 T2, TGFBRII EX5 T1), and optionally TA-1 sgRNA (SEQ ID NO: 59), which targets the TRAC locus, and 02M-1 sgRNA (SEQ ID NO: 63), which targets the [>2M locus.
  • TGFBRTI e.g., those listed in Sequence Table 18, e.g., TGFBRII EX1 T2, TGFBRII EX4 T1, TGFBRII EX4 T2, TGFBRII EX5 T1
  • TA-1 sgRNA SEQ ID NO: 59
  • 02M-1 sgRNA SEQ ID NO: 63
  • Anti-LIVl CAR T cells that comprise both a disrupted TGFBRTI gene and a disrupted Regl gene can be produced via ex vivo genetic modification using the CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR associated protein 9) technology to disrupt targeted genes TGFBRTI and Regl, optionally TRAC and/or /32M genes), and adeno-associated virus (AAV) transduction to deliver the anti-LIVl CAR construct.
  • CRISPR/Cas9 Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR associated protein 9
  • AAV adeno-associated virus
  • CRISPR-Cas9-mediated gene editing involves at least a sgRNA targeting TGFBRTI (e.g, those listed in Sequence Table 18) and a sgRNA targeting Regl (e.g, those listed in Sequence Table 1), optionally TA-1 sgRNA (SEQ ID NO: 59), which targets the TRAC locus, and 02M-1 sgRNA (SEQ ID NO: 63), which targets the 2M locus.
  • TGFBRTI e.g, those listed in Sequence Table 18
  • a sgRNA targeting Regl e.g, those listed in Sequence Table 1
  • TA-1 sgRNA SEQ ID NO: 59
  • 02M-1 sgRNA SEQ ID NO: 63
  • the anti-LIVl CAR T cells are composed of an anti-LIVl single-chain antibody fragment (scFv, which can comprise the amino acid sequence of SEQ ID NO: 532, 548, 561, or 564), followed by a CD8 hinge and transmembrane domain (e.g., comprising the amino acid sequence of SEQ ID NO: 575) that is fused to an intracellular co-signaling domain of CD28 (e.g., SEQ ID NO: 579) and a CD3i signaling domain (e.g, SEQ ID NO: 581).
  • the anti-LIVl CAR disclosed herein may comprise the amino acid sequence of SEQ ID NO: 528, 574, or 583, or the counterpart thereof without the N-terminus signal peptide (see Table 29 and Table 32 below).
  • At least 30% of a population of anti-LIVl CAR T cells express a detectable level of the anti-LIVl CAR.
  • at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% of the anti-LIVl CAR T cells express a detectable level of the anti-LIVl CAR.
  • At least 50% of a population of anti-LIVl CAR T cells may not express a detectable level of 02M 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 anti- LIVl CAR T cells may not express a detectable level of 02M 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 02M surface protein.
  • At least 50% of a population of anti- LIVl CAR T cells do not express a detectable level of TRAC 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 anti-LIVl CAR T cells may not express a detectable level of TRAC 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 TRAC surface protein.
  • more than 90% (e.g., more than 99.5%) of the anti-LIVl CAR T cells do not express a detectable TRAC surface protein.
  • a substantial percentage of the population of anti-LIVl CAR T cells described herein can, in some embodiments, comprise more than one gene edit, which results in a certain percentage of cells not expressing more than one gene and/or protein. For example, in some embodiments, at least 50% of a population of anti-LIVl CAR T cells do not express a detectable level of two surface proteins, e.g, does not express a detectable level of P2M and TRAC proteins.
  • 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 anti-LIVl CAR T cells do not express a detectable level of TRAC and 02M surface proteins.
  • at least 50% of a population of the anti-LIVl CAR T cells do not express a detectable level of TRAC and 02M surface proteins.
  • the population of anti-LIVl CAR T cells described herein can, in some embodiments, comprise more than one gene edit (e.g., in more than one gene), which may be an edit described herein.
  • the population of anti-LIVl CAR T cells may comprise a disrupted TRAC gene via the CRISPR/Cas technology using the TA-1 TRAC gRNA.
  • the anti-LIVl CAR T cells can comprise a deletion in the TRAC gene relative to unmodified T cells.
  • the anti-LIVl CAR T cells can comprise a deletion of the fragment AGAGCAACAGTGCTGTGGCC (SEQ ID NO: 69) in the TRAC gene.
  • anti-LIVl CAR T cells can comprise a disrupted /32M gene via CRISPR/Cas9 technology using the gRNA of 02M-1.
  • anti-LIVl CAR T cells can comprise Indels in the /32M gene, which comprise one or more of the nucleotide sequences of SEQ ID NOs: 83-88.
  • anti-LIVl CAR T cells comprise > 30% CAR + T cells, ⁇ 50% 02M + cells, and ⁇ 30% TCRaP + cells.
  • anti- LIVl CAR T cells comprise > 30% CAR + T cells, ⁇ 30% p2M + cells, and ⁇ 0.5% TCRap + cells. See also WO 2019/097305A2, and W02019215500, the relevant disclosures of each of which are incorporated by reference for the subject matter and purpose referenced herein.
  • the engineered T cell population can be the anti-LIVl CAR T cells disclosed herein that further comprise a disrupted Regl gene.
  • the disrupted Reg 1 gene can comprise any of the sequences provided in Sequence Tables 29-38 below.
  • the anti-LIVl CAR T cells can comprise at least 80% Regl" cells, for example, at least 85%, at least 90%, at least 95%, at least 98% or above Regl" cells.
  • the engineered T cell population can be the anti-LIVl CAR T cells disclosed herein that further comprise a disrupted TGFBRTI gene.
  • the disrupted TGFBRTI gene can comprise a nucleotide sequence selected from those listed in Sequence Tables 40-48 below.
  • the anti-LIVl CAR T cells can comprise at least 80% TGFBRH' cells, for example, at least 85%, at least 90%, at least 95%, at least 98% or above TGFBRII ' cells.
  • the genetically engineered T cell population can be the anti- LIV1 CAR T cells disclosed herein that further comprise a disrupted TGFBRII gene and a disrupted Regl gene.
  • the disrupted Reg 1 gene can comprise any of the sequences provided in Sequence Tables 29-38 below.
  • the disrupted TGFBRII gene may comprise a nucleotide sequence selected from those listed in Sequence Tables 40-48 below.
  • the anti-LIVl CAR T cells can comprise at least 80% TGFBRII' cells, for example, at least 85%, at least 90%, at least 95%, at least 98% or above TGFBRII ' cells.
  • the anti-LIVl CAR T cells can comprise at least 80% Regl' cells, for example, at least 85%, at least 90%, at least 95%, at least 98% or above Reg ' cells.
  • the anti-LIVl CAR T cells can comprise at least 60% Regl' /TGFBRII' cells, for example, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98% or above Regl'/TGFBRII' cells.
  • such a population of genetically engineered T cells can comprise about 90-97% Regl' cells, about 80-89% TGFBRII' cells, about 90-99% TCR' cells, and/or about 60-82% 02M' cells.
  • the cell population can also contain at least 50% (e.g., at least 60%) cells expressing the anti-LIVl CAR.
  • 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), anti-tumor effects, or any combination thereof.
  • engineered T cells expressing a CAR e.g., anti-LIVl CAR
  • having one or both of Regl gene and TGFBRII gene can provide synergistic effects as compared with non-engineered T cells or engineered T cells expressing a CAR without having one or both of disrupted Regl gene and disrupted TGFBRII gene.
  • the engineered T cells disclosed herein exhibit an at least 20% increase in cellular lysis capability (i.e., kill at least 20% more target cells), relative to engineered T cells not having one or both of Regl gene and TGFBRII gene.
  • the engineered T cells can exhibit about, at least, or at least about 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 90%, 95% or a number between any two of the values, increase in cellular lysis capability, relative to non-engineered T cells or engineered T cells without having one or both of Regl gene and TGFBRII gene.
  • the engineered T cells exhibit about, at least or at least about 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, or a number or a range between any of these values, increase in cellular lysis capability, relative to non-engineered T cells or engineered T cells without having one or both of Regl gene and TGFBRII gene.
  • the level of cytokines (e.g., IL-2 and/or IFN-gamma) secreted by the engineered T cells can be 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 engineered T cells not having one or both of Regl gene and TGFBRII gene.
  • the engineered T cells can exhibit enhanced anti-tumor effects such as reduction of tumor size and/or elongated survival rates.
  • a combination of an anti- LIV1 gene and one or both of a disrupted Regl gene and a disrupted TGFBRII gene (e.g., a disrupted Regl gene and a disrupted TGFBRII gene) in an engineered T cell can result in significantly enhanced efficacy against cancer (e.g., breast cancer), causing tumor regression and cancer survival.
  • the resulted tumor regression and cancer survival rate/duration by the combination is more than additive, i.e., superior to the cumulated antitumor efficacy caused by T cells expressing an anti-LIVl CAR and T cells having one or both of a disrupted Regl gene and a disrupted TGFBRII gene, separately.
  • the inhibition of tumor progression is enhanced or synergistic, that is, the inhibition is greater than the combined inhibition of progression caused by T cells expressing anti-LIVl alone plus T cells having one or both disrupted Regl gene and disrupted TGFBRII gene alone.
  • anti-LIVl CAR T cells having a disrupted Regl gene and a disrupted TGFBRII gene show significantly enhanced anti-tumor activities relative to control T cells (e.g., anti-LIVl CAR T Cells without a disrupted Regl gene and/or a disrupted TGFBRII gene).
  • the engineered T cells exhibit an about, at least, at least about, at most, or at most about, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 210%, 220%, 230%, 240%, 250%, 260%, 270%, 280%, 290%, 300%, or a combination thereof, increase in anti-tumor activities, relative to control T cells (e.g., engineered T cells without one or both of Regl gene and TGFBRII gene).
  • control T cells e.g., engineered T cells without one or both of Regl gene and TGFBRII gene.
  • engineered T cells of the present disclosure exhibit an at least about 2-fold (e.g., 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10- fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, or a number or a range between any of these values) increase in anti-tumor activities, relative to control T cells (e.g., engineered T cells without one or both of Regl gene and TGFBRII gene).
  • the engineered T cells disclosed herein exhibit at least 20% greater cellular proliferative capacity, relative to control T cells.
  • engineered T cells can exhibit about, at least, or at least about 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, or 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.
  • the engineered T cells disclosed herein exhibit an at least 20% increase in cellular viability, relative to control cells.
  • engineered T cells can exhibit about, at least, or at least about 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 90% or a number between any two of the values, increase in cellular viability, relative to control cells.
  • engineered T cells 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.
  • Control T cells can be 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-LIVl CAR) inserted into the TRAC gene, and/or a disrupted /32M gene.
  • control T cells are unedited T cells.
  • control T cells do not comprise both a disrupted Regl gene and a disrupted TGFBRTI gene.
  • the method can comprise providing a plurality of cells, wherein the plurality of cells are T cells or precursor cells thereof.
  • the method can also comprise delivering to the plurality of cells a nucleic acid encoding a chimeric antigen receptor (CAR) that comprise (1) an ectodomain that binds specifically to LIV1, or (2) an anti-LIVl antigen-binding fragment, genetically editing the Regl gene, the TGFBRTI gene, or both, and producing one or more engineered T cells expressing the CAR and having a disrupted Regl gene and/or a disrupted TGFBRJI gene.
  • CAR chimeric antigen receptor
  • the plurality of cells can be derived from parent T cells (e.g., nonedited wild-type T cells) obtained from a suitable source, for example, one or more mammal donors.
  • parent T cells e.g., nonedited wild-type T cells
  • the parent T cells are primary T cells (e.g., non-transformed and terminally differentiated T cells) obtained from one or more human donors.
  • the parent T cells can be differentiated from precursor T cells obtained from one or more suitable donor or stem cells such as hematopoietic stem cells or inducible pluripotent stem cells (iPSC), which may be cultured in vitro.
  • suitable donor or stem cells such as hematopoietic stem cells or inducible pluripotent stem cells (iPSC), which may be cultured in vitro.
  • iPSC inducible pluripotent stem cells
  • 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 are 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.
  • T cells can be isolated from a mixture of immune cells (e.g., those described herein) to produce an isolated T cell population.
  • 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.
  • PBMC peripheral blood mononuclear cells
  • 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 can be isolated by positive or negative selection prior to genetic engineering and/or post genetic engineering.
  • an isolated population of T cells can express one or more of the T cell markers, including, but not limited to a CD3+, CD4+, CD8+, or a combination thereof.
  • the T cells are isolated from a donor, or subject, and first activated and stimulated to proliferate in vitro prior to undergoing gene editing.
  • the T cell population comprises primary T cells isolated from one or more human donors. Such T cells are terminally differentiated, not transformed, depend on cytokines and/or growth factors for growth, and/or have stable genomes.
  • the T cells can be derived from stem cells (e.g., HSCs or iPSCs) via in vitro differentiation.
  • T cells from a suitable source can be 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. Patent 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 can be activated and expanded for about, at least, at least about, at most, or at most about 4 hours, 6 hours, 12 hours, 24 hours, 1 day to 4 days, 1 day to 3 days, 1 day to 2 days, 2 days to 3 days, 2 days to 4 days, 3 days to 4 days, or 2 days, 3 days, or 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.
  • the T cell population can be expanded and/or activated after the genetic editing. T cell populations or isolated T cells generated by any of the gene editing methods described herein are also within the scope of the present disclosure.
  • the method herein described can comprise delivering to the plurality of cells (e.g., T cells or precursor cells thereof described above) a nucleic acid encoding a CAR.
  • the nucleic acid encoding a CAR can comprise an ectodomain that binds specifically to LIV1 or an anti- LIV1 antigen-binding fragment.
  • the ectodomain that binds specifically to LIV1 comprises an anti-LIVl antigen-binding fragment, and optionally the anti-LIVl antigen-binding fragment comprises an anti-LIVl antibody.
  • the nucleic acid encoding a CAR can be delivered to the cells via conventional viral and non-viral based gene transfer methods known to a skilled person.
  • a nucleic acid encoding a CAR construct can be delivered to a cell using an AAV such as AAV6.
  • a nucleic acid encoding a CAR can be designed to insert into a genomic site of interest in the host T cells via a donor template.
  • a nucleic acid encoding a CAR e.g., via a donor template, which can be carried by a viral vector such as an AAV vector
  • a viral vector such as an AAV vector
  • a nucleic acid encoding a CAR (e.g., via a donor template, which can be carried by a viral vector such as an AAV vector) can be designed such that it can insert into a location within a Regl gene, a TGFBRTI gene, or a /32M gene.
  • the method can comprise genetically editing one or more genes herein described using gene editing methods known in the art.
  • the method can comprise genetically editing one or both of the Regl gene and/or the TGFBRTI gene.
  • the method comprises genetically editing both .Reg/ gene and/or TGFBRTI gene.
  • genetically editing one or more genes herein described is performed by one or more CRISPR/Cas-mediated gene editing systems described below in details, which involves the use of an RNA-guided nuclease and one or more guide RNA targeting the one or more genes to be edited.
  • the engineered T cells having a disrupted Regl gene and/or a disrupted TGFBRTI gene and further expressing a chimeric antigen receptor (CAR), optionally having additional disrupted genes, e.g., /32M, CD70, or PD-1 can be produced by sequential targeting of the genes of interest.
  • CAR chimeric antigen receptor
  • the Rcq7 gene can be disrupted first, followed by disruption of TRAC and /32M genes and CAR insertion.
  • TGFBRTI and/or /32M genes can be disrupted first, followed by CAR insertion and disruption of the Regl or other target gene.
  • the genetically engineered T cells can be produced by multiple, sequential electroporation events with multiple ribonucleoproteins (RNPs, formed by guide RNs and the Cas protein, such as a CRISPR/Cas complex) targeting the genes of interest, including but not limited to, Regl, /32M, TRAC, and CD70.
  • RNPs ribonucleoproteins
  • the engineered CAR T cells can be produced by a single electroporation event with an RNP complex comprising an RNA-guided nuclease and multiple gRNAs targeting the genes of interest, including but not limited to, Regl, TGFBRTI, /32M, TRAC, CD70, and PD-1.
  • 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).
  • the endogenous gene comprising the affected sequence can be knocked-out or knocked-down due to the sequence alteration.
  • 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 can 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 Bxbl 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 sequencespecific 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. Patent 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 Publication US2011/0145940. 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, Bxbl, phiC31, R4, PhiBTl, and Wp/SPBc/TP901-l, 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.
  • CRISPR-Cas9 Gene Editing System e.g., CRISPR/Cas9, restriction endonucleases, meganucleases homing endonucleases, and the like.
  • one or more CRISPR-Cas9 gene editing systems are used to genetically edit one or more target genes herein disclosed.
  • 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
  • CRISPR is an abbreviation for Clustered Regularly Interspaced Short Palindromic Repeats, a family of DNA sequences found in the genomes of bacteria and archaea that contain fragments of DNA (spacer DNA) with similarity to foreign DNA previously exposed to the cell, for example, by viruses that have infected or attacked the prokaryote. These fragments of DNA are used by the prokaryote to detect and destroy similar foreign DNA upon re-introduction, for example, from similar viruses during subsequent attacks. Transcription of the CRISPR locus results in the formation of an RNA molecule comprising the spacer sequence, which associates with and targets Cas (CRISPR-associated) proteins able to recognize and cut the foreign, exogenous DNA.
  • spacer DNA fragments 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’ 20nt 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 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.
  • CRISPR-Cas9 gene editing system comprises an RNA-guided nuclease and one or more guide RNAs targeting one or more target genes.
  • the Cas9 (CRISPR associated protein 9) endonuclease can be used in a CRISPR method for genetically editing the one or more genes disclosed herein.
  • the Cas9 enzyme can be one from Streptococcus pyogenes, although other Cas9 homologs may be used. It should be understood, that wild-type Cas9 can be used or modified versions of Cas9 can be used (e.g., evolved versions of Cas9, or Cas9 orthologues or variants), as provided herein.
  • Cas9 can be substituted with another RNA-guided endonuclease, such as Cpfl (of a class II CRISPR/Cas system).
  • the CRISPR/Cas system comprises components derived from a Type-I, Type-II, or Type-III system.
  • Updated classification schemes for CRISPR/Cas loci define Class 1 and Class 2 CRISPR/Cas systems, having Types I to V or VI (Makarova et al., (2015) Nat Rev Microbiol, 13(11):722-36; Shmakov et al., (2015) Mol Cell, 60:385-397).
  • Class 2 CRISPR/Cas systems have single protein effectors.
  • Class 2 Cas nucleases include, for example, Cas9, Cpfl, C2cl, C2c2, and C2c3 proteins.
  • the Cpfl nuclease (Zetsche et al., (2015) Cell 163: 1-13) is homologous to Cas9, and contains a RuvC-like nuclease domain.
  • the Cas nuclease can be from a Type-II CRISPR/Cas system (e.g., a Cas9 protein from a CRISPR/Cas9 system).
  • the Cas nuclease is from a Class 2 CRISPR/Cas system (a single-protein Cas nuclease such as a Cas9 protein or a Cpfl protein).
  • the Cas9 and Cpfl family of proteins are enzymes with DNA endonuclease activity, and they can be directed to cleave a desired nucleic acid target by designing an appropriate guide RNA, as described further herein.
  • the Cas nuclease can comprise more than one nuclease domain.
  • a Cas9 nuclease can comprise at least one RuvC-like nuclease domain (e.g., Cpfl) and at least one HNH-like nuclease domain e.g., Cas9).
  • the Cas9 nuclease can, for example, introduce a DSB in the target sequence.
  • the Cas9 nuclease is modified to contain only one functional nuclease domain.
  • the Cas9 nuclease is modified such that one of the nuclease domains is mutated or fully or partially deleted to reduce its nucleic acid cleavage activity.
  • the Cas9 nuclease is modified to contain no functional RuvC-like nuclease domain. In some embodiments, the Cas9 nuclease is modified to contain no functional HNH-like nuclease domain. In some embodiments in which only one of the nuclease domains is functional, the Cas9 nuclease is a nickase that is capable of introducing a single-stranded break (a “nick”) into the target sequence. In some embodiments, a conserved amino acid within a Cas9 nuclease domain is substituted to reduce or alter a nuclease activity.
  • the Cas nuclease nickase comprises an amino acid substitution in the RuvC-like nuclease domain.
  • Exemplary amino acid substitutions in the RuvC-like nuclease domain include D10A (based on the S. pyogenes Cas9 nuclease).
  • the nickase comprises an amino acid substitution in the HNH-like nuclease domain.
  • Exemplary amino acid substitutions in the HNH-like nuclease domain include E762A, H840A, N863 A, H983 A, and D986A (based on the S. pyogenes Cas9 nuclease).
  • a Cas9 nuclease has an amino acid sequence of SEQ ID NO: 1 or a sequence having about, at least or at least about 85%, 90%, or 95% sequence identity to SEQ ID NO: 1.
  • Cas9 nuclease (SEQ ID NO: 1): MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFDSGETAEATRLKRTAR RRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPI FGNIVDEVAYHEKYPTIYHL RKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVD AKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLD NLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPE KYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRK
  • the Cas nuclease is from a Type-I CRISPR/Cas system. In some embodiments, the Cas nuclease is a component of the Cascade complex of a Type-I CRISPR/Cas system. For example, the Cas nuclease is a Cas3 nuclease. In some embodiments, the Cas nuclease is derived from a Type-III CRISPR/Cas system. In some embodiments, the Cas nuclease is derived from Type-IV CRISPR/Cas system. In some embodiments, the Cas nuclease is derived from a Type-V CRISPR/Cas system. In some embodiments, the Cas nuclease is derived from a Type- VI CRISPR/Cas system.
  • gRNAs Guide RNAs
  • the CRISPR/Cas-mediated gene editing system comprises 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 can be a double-molecule guide RNA, or 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 (referred to as “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 in a Type V system comprises, in the 5' to 3' direction, a minimum CRISPR repeat sequence and a spacer sequence.
  • a spacer sequence in a gRNA 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 gene of interest.
  • the spacer sequence range from 15 to 30 nucleotides.
  • the spacer sequence can contain 15, 16, 17, 18, 19, 29, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides.
  • a spacer sequence contains 20 nucleotides.
  • the “target sequence” is in a target gene that is adjacent to a PAM sequence and is the sequence to be modified by an RNA-guided nuclease (e.g., Cas9).
  • the “target sequence” is on the so-called PAM-strand in a “target nucleic acid,” which is a double-stranded molecule containing the PAM-strand and a complementary non-PAM strand.
  • target nucleic acid which is a double-stranded molecule containing the PAM-strand and a complementary non-PAM strand.
  • the gRNA spacer sequence hybridizes to the complementary sequence 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: 61).
  • the spacer of a gRNA interacts with a target nucleic acid of interest in a sequence-specific manner via hybridization (/.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 can be designed to hybridize to a region of the target nucleic acid that is located 5' of a PAM recognizable by a Cas9 enzyme used in the system.
  • the spacer can perfectly match the target sequence or can 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 can vary in length, for example, 20 nucleotides in length, less than 20 nucleotides in length, or more than 20 nucleotides in length. In some embodiments, the target nucleic acid has at least: 5, 10, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides in length. In some embodiments, the target nucleic acid has at most: 5, 10, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides in length. In some embodiments, the target nucleic acid sequence has 20 bases immediately 5' of the first nucleotide of the PAM.
  • the target nucleic acid in a sequence comprising 5'- NNNNNNNNNNNNNNNNNNNNNNNNNNNNNRG-3', can be the sequence that corresponds to the Ns, wherein N can be any nucleotide, and the underlined NRG sequence is the S. pyogenes PAM.
  • the guide RNA disclosed herein can target any sequence of interest via the spacer sequence in the crRNA.
  • the degree of complementarity between the spacer sequence of the guide RNA and the target sequence in the target gene can be about, at least, at least about, at most or at most about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99%, 100% or a number between any two of the values.
  • the spacer sequence of the guide RNA and the target sequence in the target gene is 100% complementary.
  • the spacer sequence of the guide RNA and the target sequence in the target gene may contain up to 10 mismatches, e.g., up to 9, up to 8, up to 7, up to 6, up to 5, up to 4, up to 3, up to 2, or up to 1 mismatch.
  • the length of the spacer sequence in any of the gRNAs disclosed herein can depend on the CRISPR/Cas9 system and components used for editing any of the target genes also disclosed herein.
  • the spacer sequence can have 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, or more than 50 nucleotides in length.
  • the spacer sequence can have 18-24 nucleotides in length.
  • the targeting sequence can have 19- 21 nucleotides in length.
  • the spacer sequence can comprise 20 nucleotides in length.
  • the gRNA can be an sgRNA, which can comprise a 20-nucleotide spacer sequence at the 5’ end of the sgRNA sequence. In some embodiments, the sgRNA can comprise a less than 20 nucleotide spacer sequence at the 5’ end of the sgRNA sequence. In some embodiments, the sgRNA can comprise a more than 20 nucleotide spacer sequence at the 5’ end of the sgRNA sequence. In some embodiments, the sgRNA comprises a variable length spacer sequence with 17-30 nucleotides at the 5’ end of the sgRNA sequence. Examples are provided in Sequence Table 2 below. In these exemplary sequences, the fragment of “n” refers to the spacer sequence at the 5’ end.
  • the sgRNA comprises 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 1-8 uracil residues, at the 3’ end of the sgRNA sequence, e.g., 1, 2, 3, 4, 5, 6, 7, or 8 uracil residues at the 3’ end of the sgRNA sequence.
  • any of the gRNA sequences provided herein those that do not explicitly indicate modifications are meant to encompass both unmodified sequences and sequences having any suitable modifications.
  • Any of the gRNAs disclosed herein, including any of the sgRNAs can be unmodified. Alternatively, it can contain one or more modified nucleotides and/or modified backbones.
  • a modified gRNA such as an sgRNA can comprise one or more 2'-O-m ethyl phosphorothioate nucleotides, which can be located at either the 5’ end, the 3’ end, or both.
  • More than one guide RNAs can be used with a CRISPR/Cas nuclease system.
  • Each guide RNA can contain a different targeting sequence, such that the CRISPR/Cas system cleaves more than one target nucleic acid.
  • one or more guide RNAs can have the same or differing properties such as activity or stability within the Cas9 RNP complex.
  • each guide RNA can be encoded on the same or on different vectors.
  • the promoters used to drive expression of the more than one guide RNA is the same or different.
  • the gRNAs disclosed herein target a TRAC gene.
  • gRNA sequences can 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 and RNA-guided nuclease create breaks in the TRAC genomic region resulting Indels in the TRAC gene disrupting expression of the mRNA or protein.
  • Exemplary spacer sequences and gRNAs targeting a TRAC gene are provided in Sequence Table 2 below.
  • the gRNAs disclosed herein target a Regl gene, for example, target a site within exon 1, exon 2, exon 3, exon 4, exon 5, or exon 6 of the Regl gene.
  • a gRNA may comprise a spacer sequence complementary (complete or partially) to the target sequences in exon 2 or exon 4 of a Regl gene, or a fragment thereof.
  • Exemplary target sequences of Regl and exemplary gRNA sequences are provided in Sequence Table 1 below.
  • the gRNAs disclosed herein target a TGFBRTI gene, for example, target a site within exon 1, exon 2, exon 3, exon 4, exon 5, or exon 6 of the TGFBRTI gene.
  • a gRNA may comprise a spacer sequence complementary (complete or partially) to the target sequences in exon 4 or exon 5 of a TGFBRTI gene, or a fragment thereof.
  • Exemplary target sequences of TGFBRII and exemplary gRNA sequences are provided in Sequence Table 18 below.
  • the gRNAs disclosed herein target a CD70 gene, for example, target a site within exon 1 or exon 3 of a CD70 gene.
  • a gRNA may comprise a spacer sequence complementary (complete or partially) to the target sequences in exon 1 or exon 3 of a CD70 gene, or a fragment thereof.
  • Exemplary target sequences in a CD70 gene and exemplary gRNAs specific to the CD70 gene are provided in Sequence Table 2 below.
  • the gRNAs disclosed herein target a /32M gene, for example, target a suitable site within a /32M gene. See also W02019097305 and W02020/095249, the relevant disclosures of which are incorporated by reference herein for the purpose and subject matter referenced herein.
  • Other gRNA sequences can be designed using the /32M gene sequence located on Chromosome 15 (GRCh38 coordinates: Chromosome 15: 44,711,477- 44,718,877; Ensembl: ENSG00000166710).
  • gRNAs targeting the /32M genomic region and RNA-guided nuclease create breaks in the /32M genomic region resulting in Indels in the (32M gene disrupting expression of the mRNA or protein.
  • Exemplary spacer sequences and gRNAs targeting a /32M gene are provided in Sequence Table 2 below.
  • RNAs used in the CRISPR/Cas/Cpfl 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 Cpfl 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.
  • the gRNAs can be produced in vitro transcription (IVT), synthetic and/or chemical synthesis methods, or a combination thereof. Enzymatic (IVT), solid-phase, liquid-phase, combined synthetic methods, small region synthesis, and ligation methods are utilized. In some embodiments, the gRNAs are made using IVT enzymatic synthesis methods. Methods of making polynucleotides by IVT are known in the art and are described in WO2013/151666. Accordingly, the present disclosure also includes polynucleotides, e.g., DNA, constructs and vectors are used to in vitro transcribe a gRNA described herein.
  • 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.
  • non-natural modified nucleobases can be introduced into any of the gRNAs disclosed herein during synthesis or post-synthesis.
  • the modifications can be on intemucleoside linkages, purine or pyrimidine bases, or sugar.
  • the modification can be introduced at the terminal of a gRNA with chemical synthesis or with a polymerase enzyme. Examples of modified nucleic acids and their synthesis are disclosed in WO2013/052523. Synthesis of modified polynucleotides is also described in Verma and Eckstein, Annual Review of Biochemistry, vol. 76, 99-134 (1998).
  • enzymatic or chemical ligation methods can be used to conjugate polynucleotides or their regions with different functional moieties, including but not limited to targeting or delivery agents, fluorescent labels, liquids, and nanoparticles.
  • Conjugates of polynucleotides and modified polynucleotides are reviewed in Goodchild, Bioconjugate Chemistry, vol. 1(3), 165-187 (1990).
  • a CRISPR/Cas nuclease system for use in genetically editing any of the target genes disclosed here can include at least one guide RNA.
  • the CRISPR/Cas nuclease system can contain multiple gRNAs, for example, 2, 3, or 4 gRNAs. Such multiple gRNAs can target different sites in a same target gene. Alternatively, the multiple gRNAs can target different genes.
  • the guide RNA(s) and the Cas protein can form a ribonucleoprotein (RNP), e.g., a CRISPR/Cas complex.
  • RNP ribonucleoprotein
  • the guide RNA(s) can guide the Cas protein to a target sequence(s) on one or more target genes as those disclosed herein, where the Cas protein cleaves the target gene at the target site.
  • the CRISPR/Cas complex is a Cpfl/guide RNA complex.
  • the CRISPR complex is a Type-II CRISPR/Cas9 complex.
  • the Cas protein is a Cas9 protein.
  • the CRISPR/Cas9 complex is a Cas9/guide RNA complex.
  • the indel frequency (editing frequency) of a particular CRISPR/Cas nuclease system, comprising one or more specific gRNAs can be determined using a TIDE analysis, which can be used to identify highly efficient gRNA molecules for editing a target gene.
  • a highly efficient gRNA yields a gene editing frequency of higher than 80%.
  • a gRNA is considered to be highly efficient if it yields a gene editing frequency of at least 80%, at least 85%, at least 90%, at least 95%, or 100%.
  • the CRISPR/Cas nuclease system disclosed herein which comprise one or more gRNAs and at least one RNA-guided nuclease, optionally a donor template as disclosed below, can be delivered to a target cell (e.g., a T cell) for genetic editing of a target gene, via any conventional method known to a skilled person.
  • a target cell e.g., a T cell
  • components of a CRISPR/Cas nuclease system can be delivered to a target cell separately, either simultaneously or sequentially.
  • the components of the CRISPR/Cas nuclease system can be delivered into a target together (e.g., as a complex).
  • gRNA and a RNA-guided nuclease can be pre-complexed together to form a ribonucleoprotein (RNP), which can then be delivered into a target cell.
  • RNPs can be used for the delivery of guide RNAs and nuclease to T cells.
  • RNPs are useful for gene editing, at least because they minimize the risk of promiscuous interactions in a nucleic acid-rich cellular environment and protect the RNA from degradation. Methods for forming RNPs are known in the art.
  • an RNP containing an RNA- guided nuclease e.g., a Cas nuclease, such as a Cas9 nuclease
  • an RNP can be delivered to a T cell by electroporation.
  • an RNA-guided nuclease can be delivered to a cell in a DNA vector that expresses the RNA-guided nuclease in the cell.
  • an RNA- guided nuclease can be delivered to a cell in an RNA that encodes the RNA-guided nuclease and expresses the nuclease in the cell.
  • a gRNA targeting a gene can be delivered to a cell as a RNA, or a DNA vector that expresses the gRNA in the cell.
  • RNA-guided nuclease gRNA, and/or an RNP can be through direct injection or cell transfection using known methods, for example, electroporation or chemical transfection. Other cell transfection methods may be used.
  • Nucleases and/or a nucleic acid can 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 lipidmucleic 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.
  • a nucleic acid encoding a CAR can be introduced into any of the engineered T cells disclosed herein by methods known to those of skill in the art.
  • a coding sequence of the CAR can be cloned into a vector, which can be introduced into the genetically engineered T cells for expression of the CAR.
  • a variety of different methods known in the art can be used to introduce any of the nucleic acids or expression vectors disclosed herein into an immune effector cell.
  • Non-limiting examples of methods for introducing nucleic acid into a cell include: lipofection, transfection (e.g., calcium phosphate transfection, transfection using highly branched organic compounds, transfection using cationic polymers, dendrimer-based transfection, optical transfection, particle-based transfection (e.g., nanoparticle transfection), or transfection using liposomes (e.g., cationic liposomes)), microinjection, electroporation, cell squeezing, sonoporation, protoplast fusion, impalefection, hydrodynamic delivery, gene gun, magnetofection, viral transfection, and nucleofection.
  • transfection e.g., calcium phosphate transfection, transfection using highly branched organic compounds, transfection using cationic polymers, dendrimer-based transfection, optical transfection, particle-based transfection (e.g., nanoparticle transfection), or transfection using liposomes (e.g., cationic liposomes)
  • a 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 for use in delivering the CAR-coding nucleic acid is AAV serotype 6 (AAV6).
  • Adeno-associated viruses are among the most frequently used viruses for gene therapy. AAVs do not provoke an immune response upon administration to mammals, including humans. AAVs are effectively delivered to target cells, particularly when consideration is given to selecting the appropriate AAV serotype. AAVs have the ability to infect both dividing and non-dividing cells because the genome can persist in the host cell without integration.
  • a nucleic acid encoding a CAR can be designed to insert into a genomic site of interest in the host T cells.
  • the target genomic site can be in a safe harbor locus.
  • a nucleic acid encoding a CAR can be designed to insert into a genomic site of interest in the host T cells via a donor template.
  • a donor template as disclosed herein can contain a coding sequence for a CAR.
  • the CAR-coding sequence is flanked by two regions of homology to allow for efficient HDR at a genomic location of interest, for example, at a TRAC gene using a gene editing method known in the art and described herein.
  • a CRISPR-based method can be used. In this case, both strands of the DNA at the target locus can be cut by a CRISPR Cas9 enzyme guided by gRNAs specific to the target locus.
  • HDR then occurs to repair the double-strand break (DSB) and insert the donor DNA coding for the CAR.
  • 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”), such as the TRAC gene.
  • 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.
  • a donor template has no regions of homology to the targeted location in the DNA and can 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.
  • 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 into a cell 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, can be inserted at a site nearby an endogenous prompter (e.g., downstream or upstream) so that its expression can be driven by the endogenous promoter.
  • the donor template may comprise an exogenous promoter and/or enhancer, for example, a constitutive promoter, an inducible promoter, or tissue-specific promoter to control the expression of the CAR gene.
  • the exogenous promoter is an EFla promoter, see, e.g., SEQ ID NO: 167 provided in Sequence Table 7 and SEQ ID NO: 557 provided in Sequence Table 30 below.
  • exogenous sequences can also include transcriptional or translational regulatory sequences, for example, promoters, enhancers, insulators, internal ribosome entry sites, sequences encoding 2A peptides and/or polyadenylation signals.
  • transcriptional or translational regulatory sequences for example, promoters, enhancers, insulators, internal ribosome entry sites, sequences encoding 2A peptides and/or polyadenylation signals.
  • a nucleic acid encoding a CAR (e.g., via a donor template) can be designed such that it can insert into the disrupted Regl gene, the disrupted TGFBRTI gene, the disrupted TRAC gene, or the disrupted /32M.
  • a nucleic acid encoding a CAR (e.g., via a donor template, which can be carried by a viral vector such as an adeno-associated viral (AAV) vector) can be designed such that it can insert into a location within a TRAC gene to disrupt the TRAC gene in the genetically engineered T cells and express the CAR polypeptide. Disruption of TRAC leads to loss of function of the endogenous TCR.
  • a disruption in the TRAC gene can be created with an endonuclease such as those described herein and one or more gRNAs targeting one or more TRAC genomic regions. Any of the gRNAs specific to a TRAC gene and the target regions disclosed herein can be used for this purpose.
  • a genomic deletion in the TRAC gene and replacement by a CAR coding segment can be created by homology directed repair or HDR (e.g., using a donor template, which can be part of a viral vector such as an adeno-associated viral (AAV) vector).
  • a disruption in the TRAC gene can be created with an endonuclease as those disclosed herein and one or more gRNAs targeting one or more TRAC genomic regions, and inserting a CAR coding segment into the TRAC gene.
  • a nucleic acid encoding a CAR (e.g., via a donor template, which can be carried by a viral vector such as an adeno-associated viral (AAV) vector) can be designed such that it can insert into a location within a /32M gene to disrupt the /32M gene in the genetically engineered T cells and express the CAR polypeptide. Disruption of /32M leads to loss of function of the endogenous MHC Class I complexes. For example, a disruption in the /32M gene can be created with an endonuclease such as those described herein and one or more gRNAs targeting one or more /32M genomic regions. Any of the gRNAs specific to a /32M gene and the target regions disclosed herein can be used for this purpose.
  • a genomic deletion in the /32M gene and replacement by a CAR coding segment can be created by homology directed repair or HDR (e.g., using a donor template, which may be part of a viral vector such as an adeno-associated viral (AAV) vector).
  • a disruption in the /32M gene can be created with an endonuclease as those disclosed herein and one or more gRNAs targeting one or more /32M genomic regions, and inserting a CAR coding segment into the /32M gene.
  • a nucleic acid encoding a CAR (e.g., via a donor template, which can be carried by a viral vector such as an adeno-associated viral (AAV) vector) can be designed such that it can insert into a location within a Regl gene to disrupt the Regl gene in the genetically engineered T cells and express the CAR polypeptide. Disruption of Regl leads to loss of function of the endogenous Regl protein.
  • a disruption in the Regl gene can be created with an endonuclease such as those described herein and one or more gRNAs targeting one or more Regl genomic regions. Any of the gRNAs specific to a Regl gene and the target regions disclosed herein can be used for this purpose.
  • a genomic deletion in the Regl gene and replacement by a CAR coding segment can be created by homology directed repair or HDR (e.g., using a donor template, which may be part of a viral vector such as an adeno-associated viral (AAV) vector).
  • a disruption in the Regl gene can be created with an endonuclease as those disclosed herein and one or more gRNAs targeting one or more Regl genomic regions, and inserting a CAR coding segment into the Regl gene.
  • a nucleic acid encoding a CAR (e.g., via a donor template, which can be carried by a viral vector such as an adeno-associated viral (AAV) vector) can be designed such that it can insert into a location within a TGFBRII gene to disrupt the TGFBRII gene in the genetically engineered T cells and express the CAR polypeptide. Disruption of Regl leads to loss of function of the endogenous TGFBRII receptor.
  • a disruption in the TGFBRII gene can be created with an endonuclease such as those described herein and one or more gRNAs targeting one or more TGFBRII genomic regions. Any of the gRNAs specific to a TGFBRII gene and the target regions disclosed herein can be used for this purpose.
  • a genomic deletion in the TGFBRII gene and replacement by a CAR coding segment can be created by homology directed repair or HDR (e.g., using a donor template, which may be part of a viral vector such as an adeno-associated viral (AAV) vector).
  • a disruption in the TGFBRII gene can be created with an endonuclease as those disclosed herein and one or more gRNAs targeting one or more TGFBRII genomic regions, and inserting a CAR coding segment into the TGFBRII gene.
  • one or more nucleic acid encoding one or more CARs can be designed such that it can insert into one or more target locations, such as CD70, to disrupt one or more target genes in the engineered T cells.
  • a donor template for delivering an anti-LIVl CAR can be an AAV vector inserted with a nucleic acid fragment comprising the coding sequence of the anti-LIVl CAR, and optionally regulatory sequences for expression of the anti-LIVl CAR (e.g., a promoter such as the EFla promoter provided in the sequence Table provided herein), which can be flanked by homologous arms for inserting the coding sequence and the regulatory sequences into a genomic locus of interest.
  • the nucleic acid fragment is inserted in the endogenous TRAC gene locus, thereby disrupting expression of the TRAC gene.
  • the nucleic acid can replace a fragment in the TRAC gene, for example, a fragment comprising the nucleotide sequence of SEQ ID NO: 69.
  • the donor template for delivering the anti-LIVl CAR can comprise a nucleotide sequence of SEQ ID NO: 528, 574, or 583, which can be inserted into a disrupted TRAC gene, for example, replacing the fragment of SEQ ID NO: 69.
  • the donor template for delivering the anti-LIVl CAR can comprise a nucleotide sequence of SEQ ID NO: 583, which can be inserted into a disrupted TRAC gene, for example, replacing the fragment of SEQ ID NO: 69.
  • Provided herein also include a method for treating cancer.
  • the method can comprise administering to a subject an engineered T cell herein described or a population of the engineered T cells.
  • Non-limiting examples of cancers that can 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.
  • 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
  • neuronal tumors e.g., neuronal tumors.
  • the methods comprise delivering the CAR T cells (e.g., anti-LIVl 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 engineered T cells, methods and kits disclosed herein can be used to various types of cancer, including but are not limited to, melanoma (e.g., metastatic malignant melanoma), renal cancer (e.g., clear cell carcinoma), prostate cancer (e.g., hormone refractory prostate adenocarcinoma), pancreatic adenocarcinoma, breast cancer, colon cancer, lung cancer (e.g., non-small cell lung cancer (NSCLC) and small-cell lung cancer (SCLC)), esophageal cancer, squamous cell carcinoma of the head and neck, liver cancer, ovarian cancer, cervical cancer, thyroid cancer, glioblastoma, glioma, leukemia, lymphoma, and other neoplastic malignancies.
  • melanoma e.g., metastatic malignant melanoma
  • renal cancer e.g., clear cell carcinoma
  • prostate cancer e.g., hormone refractory prostate a
  • the disease or condition provided herein includes refractory or recurrent malignancies whose growth may be inhibited using the methods and compositions disclosed herein.
  • the cancer is carcinoma, squamous carcinoma, adenocarcinoma, sarcomata, endometrial cancer, breast cancer, ovarian cancer, cervical cancer, fallopian tube cancer, primary peritoneal cancer, colon cancer, colorectal cancer, squamous cell carcinoma of the anogenital region, melanoma, renal cell carcinoma, lung cancer, non-small cell lung cancer, squamous cell carcinoma of the lung, stomach cancer, bladder cancer, gall bladder cancer, liver cancer, thyroid cancer, laryngeal cancer, salivary gland cancer, esophageal cancer, head and neck cancer, glioblastoma, glioma, squamous cell carcinoma of the head and neck, prostate cancer, pancreatic cancer, mesothelioma, sarcoma, hematological cancer, leuk
  • the cancer is carcinoma, squamous carcinoma (e.g., cervical canal, eyelid, tunica conjunctiva, vagina, lung, oral cavity, skin, urinary bladder, tongue, larynx, and gullet), and adenocarcinoma (for example, prostate, small intestine, endometrium, cervical canal, large intestine, lung, pancreas, gullet, rectum, uterus, stomach, mammary gland, and ovary).
  • the cancer is sarcomata (e.g., myogenic sarcoma), leukosis, neuroma, melanoma, and lymphoma.
  • the cancer can include 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, intrahepatic biliary cancer, and a combination thereof.
  • NSCLC non-small cell lung
  • the cancer can be a solid tumor, a liquid tumor, or a combination thereof.
  • the cancer is a solid tumor, including but are not limited to, melanoma, renal cell carcinoma, lung cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, gall bladder cancer, laryngeal cancer, liver cancer, thyroid cancer, stomach cancer, salivary gland cancer, prostate cancer, pancreatic cancer, Merkel cell carcinoma, brain and central nervous system cancers, and any combination thereof.
  • the cancer is a liquid tumor.
  • the cancer is a hematological cancer, including but not limited to, Diffuse large B cell lymphoma (“DLBCL”), Hodgkin's lymphoma (“HL”), Non-Hodgkin's lymphoma (“NHL”), Follicular lymphoma (“FL”), acute myeloid leukemia (“AML”), and Multiple myeloma (“MM”).
  • DBCL Diffuse large B cell lymphoma
  • HL Hodgkin's lymphoma
  • NHL Non-Hodgkin's lymphoma
  • FL Follicular lymphoma
  • AML acute myeloid leukemia
  • MM Multiple myeloma
  • the cancer is breast cancer, prostate cancer, squamous tumor cancer, neuronal tumor cancer, or a combination thereof. In some embodiments, the cancer comprises cancer cells expressing LIV1.
  • the step of administering can include introducing (e.g., transplantation) the cells, e.g., an engineered T cell or a population thereof described herein, 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, z.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 can be any subject for whom diagnosis, treatment, or therapy is desired.
  • the subject can be a mammal. In some embodiments, the subject is a human.
  • the engineered T cell population being administered according to the methods described herein can 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 can used, such as those obtained from genetically identical donors (e.g., identical twins).
  • the cells can be autologous cells; that is, the engineered T cells are obtained or isolated from a subject and administered to the same subject, z.e., the donor and recipient are the same.
  • a donor can be 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 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.
  • the effective amount of cells can comprise about, at least, or at least about, 10 2 cells, 5 x io 2 cells, 10 3 cells, 5 x io 3 cells, 10 4 cells, 5 x 10 4 cells, 10 5 cells, 2 x io 5 cells, 3 x io 5 cells, 4 x io 5 cells, 5 x io 5 cells, 6 x io 5 cells, 7 x io 5 cells, 8 x io 5 cells, 9 x io 5 cells, 1 x io 6 cells, 2 x io 6 cells, 3 x io 6 cells, 4 x io 6 cells, 5 x 10 6 cells, 6 x 10 6 cells, 7 x io 6 cells, 8 x io 6 cells, 9 x io 6 cells, 10 7 cells, 1.2 x io 7 cells, 1.4 x io 7 cells, 1.6 x io 7 cells, 1.8
  • the cells are derived from one or more donors, or are obtained from an autologous source.
  • 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 intrastemal injection and infusion.
  • the route is intravenous.
  • engineered T cells are administered systemically.
  • Systemic administration refers to the administration of a population of cells other than directly into a target site, tissue, or organ, such that it enters 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 engineered T cells, methods, and kits disclosed herein can be used with additional cancer therapeutics or therapy to treat cancer.
  • the treatment can comprise administration of at least one additional cancer therapeutics or cancer therapy.
  • the treatment can comprise administration a therapeutically effective amount of at least one additional cancer therapeutics or cancer therapy.
  • the engineered T cells herein described and the cancer therapeutics or cancer therapy can, for example, co-administered simultaneously or sequentially.
  • the cancer therapies include, but are not limited to, surgery, chemotherapy, radiation therapy, hormonal therapy, immunotherapy, complementary or alternative therapy, and any combination thereof. Kits
  • kits for use in producing the engineered T cells and for therapeutic uses can, for example, comprise components for performing genetic edit of one or more of TRAC gene, Regl gene, TGFBRII gene, /32M gene, CD90 gene and/or PD-1 gene.
  • a kit also comprises a nucleic acid encoding a CAR, wherein the CAR comprises an ectodomain that binds specifically to LIV1 or an anti-LIVl antigenbinding fragment, and components for delivery of the nucleic acid.
  • the nucleic acid encoding an anti-LIVl CAR is part of a donor template as disclosed herein, which can contain homologous arms flanking the anti-LIVl CAR coding sequence.
  • the donor template is carried by a viral vector such as an AAV vector.
  • the components for genetically editing one or more of the target genes can comprise a suitable endonuclease such as an RNA-guided endonuclease and one or more nucleic acid guides, which direct cleavage of one or more suitable genomic sites by the endonuclease.
  • the kit can comprise a Cas enzyme such as Cas 9 and one or more gRNAs targeting a TRAC gene, a. Regl gene, and/or a TGFBRTI gene.
  • the kit comprises gRNAs specific to TRAC gene for inserting the anti-LIVl CAR sequence into the TRAC gene.
  • kits can further comprise components for additional gene editing, for example, gRNAs and optionally additional endonucleases for editing other target genes such as /32M, CD90 and/or PD-1.
  • a kit can, for example, comprise a population of immune cells to which the genetic editing will be performed (e.g., a leukopak).
  • a leukopak sample can be an enriched leukapheresis product collected from peripheral blood, which typically contains a variety of blood cells including monocytes, lymphocytes, platelets, plasma, and red cells.
  • a kit disclosed herein can comprise a population of therapeutic T cells as disclosed for the intended therapeutic purposes.
  • Kit disclosed herein can further comprise instructions for making the engineered T cells, or therapeutic applications of the therapeutic T cells.
  • the included instructions can comprise a description of using the gene editing components to genetically engineer one or more of the target genes (e.g., TRAC, Regl, TGFBRII, (32M, PD-1, CD70, or a combination thereof).
  • the included instructions can comprise a description of how to introduce a nucleic acid encoding a CAR construction into the T cells for making therapeutic T cells.
  • the kit can comprise instructions for administration of the engineered T cells as disclosed herein to achieve the intended activity, e.g., eliminating disease cells targeted by the anti-LIVl CAR expressed on the therapeutic T cells.
  • the kit can further comprise a description of selecting a subject suitable for treatment based on identifying whether the subject is in need of the treatment.
  • the instructions relating to the use of the therapeutic T cells described herein generally include information as to dosage, dosing schedule, and route of administration for the intended treatment.
  • the containers can be unit doses, bulk packages (e.g., multi-dose packages) or sub-unit doses. Instructions supplied in the kits of the disclosure are typically written instructions on a label or package insert.
  • the label or package insert indicates that the therapeutic T cells are used for treating, delaying the onset, and/or alleviating a disease or disorder in a subject.
  • kits provided herein are in suitable packaging.
  • suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging, and the like.
  • packages for use in combination with a specific device such as an infusion device for administration of the therapeutic T cells.
  • a kit can have a sterile access port (for example, the container can be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the container can also have a sterile access port.
  • Allogeneic human T cells that lack expression of the TRAC gene, 02M gene, TGFBRII gene and Regnase-1 gene, and express a chimeric antigen receptor (CAR) targeting Livl were produced.
  • Activated primary human T cells were electroporated with Cas9:sgRNA RNP complexes and adeno-associated adenoviral vectors (AAVs) to generate TRAC-/P2M- /TGFBRII-/REG-1- anti-Livl CAR+ T cells.
  • the sgRNAs which form RNPs with the Cas9 enzyme, can be introduced into the T cells in a single electroporation event. After the electroporation, the cells were transduced with the recombinant AAVs to introduce the donor template encoding for the anti- Livl CAR.
  • AAV serotype 6 comprising one of the nucleotide sequences encoding an anti-Livl CAR (971 (SEQ ID NO: 527), 975 (SEQ ID NO: 582), and 976 (SEQ ID NO: 586), were delivered with Cas9:sgRNA RNPs (1 pM Cas9, 5 pM gRNA) to activated allogeneic human T cells.
  • Cas9:sgRNA RNPs (1 pM Cas9, 5 pM gRNA
  • the following sgRNAs were used: TRAC (SEQ ID NO: 61), p2M (SEQ ID NO: 64), TGFBRII (SEQ ID NO: 10) and REG-1 (SEQ ID NO: 327).
  • the unmodified versions (or other modified versions) of the sgRNAs may also be used (e.g., SEQ ID NOS: 14-53 and 264-315).
  • Table 33 presents in different edited T cells that were produced. ‘+’ indicates an intact gene; indicates gene disruption.
  • CTX971, CTX975 and CTX976 are previously described in W02020/095249, which is incorporated herein by reference.
  • FIG. 2 also includes diagrams showing the expressions of CAR in edited cells via ddPCR and flow cytometry.
  • FIG. 3 also includes diagrams showing the T cell numbers of the engineered CAR T cells. Table 35: T-cell Numbers (x 10 8 )
  • a cell killing (cytotoxicity) assay was used to assess the ability of the anti-Livl CAR + T cells to cause cellular lysis in breast cancer cell lines (MCF7 and ZR-75-1).
  • MCF7 cells were cultured in Eagle’s Minimum Essential Media (EMEM) with 10% fetal bovine serum (FBS).
  • ZR-75-1 cells were cultured in RPMI-1640 Medium (ATCC Modification) with 10% FBS. All cells were cultured at 37°C with 5% CO2.
  • T cells were added to the wells containing target cancer cells at ratios of 0.5: 1, 1 : 1, 2: 1, and 4: 1 T celktarget cell.
  • CAR- T cells that were electroporated with Cas9:sgRNA RNP complexes but were not transduced with AAV, and thus do not express an anti-Livl CAR, served as negative controls.
  • CTX975 and TGFBRII/Reg 975 cells exhibited the highest degree of cytotoxicity against the MCF7 and ZR-75-1 cell lines.
  • the EP only control does not have any CAR transduction but has TRAC/P2M edits.
  • the TGFBRII/Reg EP only control does not have any CAR transduction but has TRAC/ p2M/TGFBRII/Regnase edits.
  • the MILLIPLEX MAP Human Cytokine/Chemokine Magnetic Bead Panel - Immunology Multiplex Assay kit (Millipore, catalog # HCYTOMAG-60K) using magnetic microspheres, anti-human IFNy bead (Millipore, catalog # HCYIFNG-MAG) and anti-human IL-2 bead (Millipore, catalog # EHL2-MAG), respectively, was used to quantify IFN-y 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 IFNy 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 pL Sheath Fluid, and agitated on a plate shaker for 5 minutes.
  • TGFBRII/Reg 975 T cells secreted the effector cytokines interferon-y and interleukin-2 when co-cultured with the target cells lines MCF7 and ZR-75-1 at levels significantly higher than the other CAR T cells. See also FIGs. 5A-5D.
  • Table 37 IFN- y (pg/mL) after co-culture with different ratios of T cell: MCF7
  • MCF-7 cells or ZR751 cells were electroporated with Cas9:sgRNA RNP complexes using sgRNA targeting the Livl gene. After the electroporation, the cells were expanded and Livl editing was confirmed to be 75% by sequencing and subsequent TIDE. The cells were seeded at 10 cells per well in a 96-well plate and allowed to expand. Upon expansion of the individual wells, genomic DNA was isolated and subsequent sequencing and TIDE confirmed 99% editing in a population of cells. These cells were further expanded and utilized in a cytotoxicity assay.
  • the MCF7 cells were seeded in a 96 well plate in Eagle’s Minimum Essential Media (EMEM) with 10% fetal bovine serum (FBS) at 37°C with 5% CO2. The following day, CTX975 TGFBRII/Reg KO T cells were added to the wells containing target cancer cells at ratios of 1 : 1 and 2: 1 T cell: target cell. After approximately 24 hours, the T cells were removed from the culture by aspiration. PBS washes ensured the removal of all T cells. 100 pL CellTiter-Glo® (Promega) was added to each well of the plate to assess the number of remaining viable cells and incubated for 10 minutes at 37°C. The luminescence from each well was then quantified using a plate reader.
  • EMEM Eagle’s Minimum Essential Media
  • FBS fetal bovine serum
  • Table 41 % lysis of MCF7 cells versus Livl-edited MCF7 cells by TGFBRII/Reg 975 CAR T cells
  • mice Female NSG mice aged 5-8 weeks were implanted with one 90-day 0.72mg P-estradiol pellet three days prior to MCF-7 cell inoculation. After three days, le7 MCF-7 cells per mouse were inoculated into the mammary fat pad of each mouse. Tumors grew until they reached ⁇ 50mm 3 . At that point, mice were randomized and injected intravenously with CAR T cells at le7 or 2e7 CAR+ cells per mouse in a total volume of 200 uL. Tumor volumes and body weight were evaluated every few days. Tumor volumes are presented in Table 42. See also FIGs. 6, 7A-7B, and 8A-8D.
  • mice Female NSG mice were subcutaneously inoculated with le7 MDA-MB-231 cells per mouse into the right flank. Tumors were allowed to grow until they reached a mean tumor size between 25-75 mm3 with a target of 50 mm3. Upon reaching the target tumor volume, mice were randomized into 9 groups. Group 1 received no treatment. Groups 2-9 were given a single dose of CAR T cells on Day 1 by intravenous administration. CAR T cell dose was based off the percentage of CAR+ cells as measured by flow cytometry. Each CAR product was dosed at both le7 and 2e7 CAR+ cells per mouse in a total volume of 200 pL. Tumor volumes and body weights were measured twice a week and gross observations were made daily. Treatment details are provided in Tables 42-43 below.
  • FIG. 12 and FIG. 13A-13B Tumor volumes in mice treated by anti-Livl CAR-T cells, with or without TGFBRII and/or Regnase-1 edits are shown in FIG. 12 and FIG. 13A-13B. The results indicate that the additional gene editing of the TGFBRII gene and the Regnase-1 gene, either alone or in combination, improves the potency of the anti-Livl CAR-T cells.
  • * indicates a nucleotide with a 2'-O-methyl phosphorothioate modification.
  • n refers to the spacer sequence at the 5' end.
  • Sequence Table 7 Exemplary AAV Donor Template Sequences.
  • Sequence Table 21 On-Target Gene Edited Sequences > 1% Frequency in At Least One Gene Edited T Cell Donor for the TGFBRII-Exl-T3 gRNA.
  • On-target sequence centered on cleavage site, with 10 bp in either direction.
  • the portion of the gRNA target sequence aligning with the Reference on-target sequence is underlined and the PAM is indicated by parenthesis.
  • a On-target sequence centered on cleavage site, with 10 bp in either direction.
  • the portion of the gRNA target sequence aligning with the Reference on-target sequence is underlined and the PAM is indicated by parenthesis.
  • CAR and CTX-972b CAR Sequence Table 29.
  • Sequence Table 31 Exemplary CAR structures of CTX-973 CAR, CTX-974 CAR, CTX-975 CAR, CTX-976 CAR, CTX-977 CAR, CTX-978 CAR, CTX-979 CAR and CTX- 979b CAR.
  • a range includes each individual member.
  • a group having 1-3 articles refers to groups having 1, 2, or 3 articles.
  • a group having 1-5 articles refers to groups having 1, 2, 3, 4, or 5 articles, and so forth.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Mycology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Veterinary Medicine (AREA)
  • Cell Biology (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Oncology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

L'invention concerne des cellules T modifiées, ainsi que des procédés et des compositions associés pour produire les cellules T modifiées. L'invention concerne également des utilisations thérapeutiques des cellules T modifiées. Les cellules T modifiées peuvent exprimer un récepteur antigénique chimérique (CAR) qui se lie spécifiquement à LIV1 et présentent au moins l'un parmi un gène de Régnase-1 (Reg1) lysé et un gène de récepteur de facteur de croissance transformant bêta II (TGFBRII) lysé.
PCT/IB2022/062244 2021-12-15 2022-12-14 Cellule anti-liv1 modifiée avec lyse de régnase-1 et/ou tgfbrii WO2023111913A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202163290059P 2021-12-15 2021-12-15
US63/290,059 2021-12-15
US202163290550P 2021-12-16 2021-12-16
US63/290,550 2021-12-16

Publications (1)

Publication Number Publication Date
WO2023111913A1 true WO2023111913A1 (fr) 2023-06-22

Family

ID=84923108

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2022/062244 WO2023111913A1 (fr) 2021-12-15 2022-12-14 Cellule anti-liv1 modifiée avec lyse de régnase-1 et/ou tgfbrii

Country Status (1)

Country Link
WO (1) WO2023111913A1 (fr)

Citations (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998053060A1 (fr) 1997-05-23 1998-11-26 Gendaq Limited Proteines de liaison d'acide nucleique
WO1998053059A1 (fr) 1997-05-23 1998-11-26 Medical Research Council Proteines de liaison d'acide nucleique
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
US6140081A (en) 1998-10-16 2000-10-31 The Scripps Research Institute Zinc finger binding domains for GNN
WO2002016536A1 (fr) 2000-08-23 2002-02-28 Kao Corporation Detergent bactericide antisalissures, destine aux surfaces dures
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
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
WO2003016496A2 (fr) 2001-08-20 2003-02-27 The Scripps Research Institute Domaines de fixation en doigt de zinc pour cnn
US6534261B1 (en) 1999-01-12 2003-03-18 Sangamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
US6534055B1 (en) 1988-11-23 2003-03-18 Genetics Institute, Inc. 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
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
US6905874B2 (en) 2000-02-24 2005-06-14 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US6905680B2 (en) 1988-11-23 2005-06-14 Genetics Institute, Inc. Methods of treating HIV infected subjects
US7067318B2 (en) 1995-06-07 2006-06-27 The Regents Of The University Of Michigan Methods for transfecting T cells
US7175843B2 (en) 1994-06-03 2007-02-13 Genetics Institute, Llc Methods for selectively stimulating proliferation of T cells
US7888121B2 (en) 2003-08-08 2011-02-15 Sangamo Biosciences, Inc. Methods and compositions for targeted cleavage and recombination
US20110145940A1 (en) 2009-12-10 2011-06-16 Voytas Daniel F Tal effector-mediated dna modification
US7972854B2 (en) 2004-02-05 2011-07-05 Sangamo Biosciences, Inc. Methods and compositions for targeted cleavage and recombination
WO2013052523A1 (fr) 2011-10-03 2013-04-11 modeRNA Therapeutics Nucléosides, nucléotides et acides nucléiques modifiés, et leurs utilisations
WO2013151666A2 (fr) 2012-04-02 2013-10-10 modeRNA Therapeutics Polynucléotides modifiés destinés à la production de produits biologiques et de protéines associées à une maladie humaine
US9228026B2 (en) 2010-12-06 2016-01-05 Seattle Genetics, Inc. Humanized antibodies to LIV-1 and use of same to treat cancer
WO2019097305A2 (fr) 2017-05-12 2019-05-23 Crispr Therapeutics Ag Matériaux et procédés de génie cellulaire et leurs utilisations en immuno-oncologie
WO2019215500A1 (fr) 2018-05-11 2019-11-14 Crispr Therapeutics Ag Procédés et compositions pour le traitement du cancer
WO2020095249A1 (fr) 2018-11-07 2020-05-14 Crispr Therapeutics Ag Thérapie contre le cancer des cellules immunitaires anti-liv1
WO2020206248A1 (fr) * 2019-04-03 2020-10-08 Precision Biosciences, Inc. Cellules immunitaires génétiquement modifiées comprenant un arnsh adapté au microarn (shrnamir)
WO2020219682A2 (fr) * 2019-04-24 2020-10-29 St. Jude Children's Research Hospital, Inc. Knock-out de gènes pour améliorer la fonction des lymphocytes t
WO2020223535A1 (fr) * 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cellules exprimant un récepteur recombinant à base d'un locus modifié du tgfbr2, et polynucléotides et méthodes associés
WO2022064428A1 (fr) 2020-09-23 2022-03-31 Crispr Therapeutics Ag Lymphocytes t génétiquement modifiés à interruption de regnase-1 et/ou de tgfbrii ayant une fonctionnalité et une persistance améliorées

Patent Citations (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7144575B2 (en) 1988-11-23 2006-12-05 The Regents Of The University Of Michigan Methods for selectively stimulating proliferation of T cells
US6887466B2 (en) 1988-11-23 2005-05-03 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US6905680B2 (en) 1988-11-23 2005-06-14 Genetics Institute, Inc. Methods of treating HIV infected subjects
US5883223A (en) 1988-11-23 1999-03-16 Gray; Gary S. CD9 antigen peptides and antibodies thereto
US7232566B2 (en) 1988-11-23 2007-06-19 The United States As Represented By The Secretary Of The Navy Methods for treating HIV infected subjects
US6534055B1 (en) 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
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
US7175843B2 (en) 1994-06-03 2007-02-13 Genetics Institute, Llc Methods for selectively stimulating proliferation of T cells
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
US6905681B1 (en) 1994-06-03 2005-06-14 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US7172869B2 (en) 1995-05-04 2007-02-06 The United States Of America As Represented By The Secretary Of The Navy Methods for transfecting T cells
US6692964B1 (en) 1995-05-04 2004-02-17 The United States Of America As Represented By The Secretary Of The Navy Methods for transfecting T cells
US7067318B2 (en) 1995-06-07 2006-06-27 The Regents Of The University Of Michigan Methods for transfecting T cells
WO1998053060A1 (fr) 1997-05-23 1998-11-26 Gendaq Limited Proteines de liaison d'acide nucleique
WO1998053059A1 (fr) 1997-05-23 1998-11-26 Medical Research Council Proteines de liaison d'acide nucleique
WO1998053058A1 (fr) 1997-05-23 1998-11-26 Gendaq Limited Proteines de liaison d'acide nucleique
US6140081A (en) 1998-10-16 2000-10-31 The Scripps Research Institute Zinc finger binding domains for GNN
US6534261B1 (en) 1999-01-12 2003-03-18 Sangamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
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
US6905874B2 (en) 2000-02-24 2005-06-14 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
US6797514B2 (en) 2000-02-24 2004-09-28 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
WO2002016536A1 (fr) 2000-08-23 2002-02-28 Kao Corporation Detergent bactericide antisalissures, destine aux surfaces dures
WO2003016496A2 (fr) 2001-08-20 2003-02-27 The Scripps Research Institute Domaines de fixation en doigt de zinc pour 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
US20110145940A1 (en) 2009-12-10 2011-06-16 Voytas Daniel F Tal effector-mediated dna modification
US9228026B2 (en) 2010-12-06 2016-01-05 Seattle Genetics, Inc. Humanized antibodies to LIV-1 and use of same to treat cancer
WO2013052523A1 (fr) 2011-10-03 2013-04-11 modeRNA Therapeutics Nucléosides, nucléotides et acides nucléiques modifiés, et leurs utilisations
WO2013151666A2 (fr) 2012-04-02 2013-10-10 modeRNA Therapeutics Polynucléotides modifiés destinés à la production de produits biologiques et de protéines associées à une maladie humaine
WO2019097305A2 (fr) 2017-05-12 2019-05-23 Crispr Therapeutics Ag Matériaux et procédés de génie cellulaire et leurs utilisations en immuno-oncologie
WO2019215500A1 (fr) 2018-05-11 2019-11-14 Crispr Therapeutics Ag Procédés et compositions pour le traitement du cancer
WO2020095249A1 (fr) 2018-11-07 2020-05-14 Crispr Therapeutics Ag Thérapie contre le cancer des cellules immunitaires anti-liv1
WO2020206248A1 (fr) * 2019-04-03 2020-10-08 Precision Biosciences, Inc. Cellules immunitaires génétiquement modifiées comprenant un arnsh adapté au microarn (shrnamir)
WO2020219682A2 (fr) * 2019-04-24 2020-10-29 St. Jude Children's Research Hospital, Inc. Knock-out de gènes pour améliorer la fonction des lymphocytes t
WO2020223535A1 (fr) * 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cellules exprimant un récepteur recombinant à base d'un locus modifié du tgfbr2, et polynucléotides et méthodes associés
WO2022064428A1 (fr) 2020-09-23 2022-03-31 Crispr Therapeutics Ag Lymphocytes t génétiquement modifiés à interruption de regnase-1 et/ou de tgfbrii ayant une fonctionnalité et une persistance améliorées

Non-Patent Citations (29)

* Cited by examiner, † Cited by third party
Title
"Sequences of Proteins of Immunological Interest", 1991, U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES
AL-LAZIKANI ET AL., J. MOLEC. BIOL., vol. 273, 1997, pages 927 - 948
ALMAGRO, J., MOL. RECOGNIT., vol. 17, 2004, pages 132 - 143
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 10
CHANG ET AL., PROC. NATL. ACAD. SCI. USA, vol. 84, 1987, pages 4959 - 4963
CHOTHIA ET AL., NATURE, vol. 342, 1989, pages 877
CHOTHIA, C. ET AL., J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CORPET ET AL., NUC. ACIDS RES., vol. 16, 1988, pages 10881 - 90
DELTCHEVA ET AL., NATURE, vol. 471, 2011, pages 602 - 607
ENBLAD ET AL., HUMAN GENE THERAPY., vol. 26, no. 8, 2015, pages 498 - 505
GOODCHILD, BIOCONJUGATE CHEMISTRY, vol. 1, no. 3, 1990, pages 165 - 187
HIGGINSSHARP, CABIOS, vol. 5, 1989, pages 151 - 3
HIGGINSSHARP, GENE, vol. 73, 1988, pages 237 - 44
HUANG ET AL., COMPUTER APPLS. IN THE BIOSCIENCES, vol. 8, 1992, pages 155 - 65
JINEK ET AL., SCIENCE, vol. 337, 2012, pages 816 - 821
KAKARLAGOTTSCHALK, CANCER J., vol. 20, no. 2, 2014, pages 151 - 155
KOONIN ET AL., CURR OPIN MICROBIOL, vol. 37, 2017, pages 67 - 78
MAKAROVA ET AL., NAT REV MICROBIOL, vol. 13, no. 11, 2015, pages 722 - 36
MAUDE ET AL., BLOOD, vol. 125, no. 26, 2015, pages 4017 - 4023
NEEDLEMANWUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443
NEHLS ET AL., SCIENCE, vol. 272, 1996, pages 886 - 889
PEARSON ET AL., METH. MOL. BIO., vol. 24, 1994, pages 307 - 31
PEARSONLIPMAN, PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 2444
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS
SHMAKOV ET AL., MOL CELL, vol. 60, 2015, pages 385 - 397
SMITHWATERMAN, ADV. APPL. MATH, vol. 2, 1981, pages 482
VERMAECKSTEIN, ANNUAL REVIEW OF BIOCHEMISTRY, vol. 76, 1998, pages 99 - 134
WEI JUN ET AL: "Targeting REGNASE-1 programs long-lived effector T cells for cancer therapy", NATURE, NATURE PUBLISHING GROUP UK, LONDON, vol. 576, no. 7787, 1 December 2019 (2019-12-01), pages 471 - 476, XP036974188, ISSN: 0028-0836, [retrieved on 20191211], DOI: 10.1038/S41586-019-1821-Z *
ZETSCHE ET AL., CELL, vol. 163, 2015, pages 1 - 13

Similar Documents

Publication Publication Date Title
US11497773B2 (en) Genetically engineered t cells with regnase-1 and/or TGFBRII disruption have improved functionality and persistence
EP3790629A1 (fr) Procédés et compositions pour le traitement du cancer
JP7471289B2 (ja) 抗liv1免疫細胞癌療法
US20230346836A1 (en) Genetically engineered t cells with disrupted casitas b-lineage lymphoma proto-oncogene-b (cblb) and uses thereof
US20200140815A1 (en) Anti-ptk7 immune cell cancer therapy
US20220193134A1 (en) Co-use of lenalidomide with car-t cells
US11926676B2 (en) Masked chimeric antigen receptor specific to tyrosine-protein kinase like 7 (PTK7) and immune cells expressing such
CN116802203A (zh) 从经修饰的恒定cd3免疫球蛋白超家族链基因座表达嵌合受体的细胞、相关多核苷酸和方法
US20230303713A1 (en) Anti-cd19 car-t cells with multiple gene edits and therapeutic uses thereof
WO2023111913A1 (fr) Cellule anti-liv1 modifiée avec lyse de régnase-1 et/ou tgfbrii
US20220288122A1 (en) Genetically engineered t cells with ptpn2 knockout have improved functionality and anti-tumor activity
US20230128917A1 (en) Genetically engineered immune cells having a disrupted cd83 gene
WO2024062388A2 (fr) Cellules immunitaires génétiquement modifiées exprimant un récepteur antigénique chimérique ciblant cd20
US20230331841A1 (en) Anti-cd83 car-t cells with regnase-1 and/or tgfbrii disruption
WO2024023801A2 (fr) Cellules immunitaires génétiquement modifiées ayant un transporteur interrompu associé à un gène de traitement d'antigène -1 (tap -1)
WO2024023802A2 (fr) Cellules immunitaires génétiquement modifiées ayant un gène transporteur associé au traitement des antigènes-2 (tap-2) interrompu
WO2024023804A2 (fr) Cellules immunitaires génétiquement modifiées ayant un gène protéine de liaison transporteur associé au traitement des antigènes (tapbp) interrompu
WO2024003786A1 (fr) Récepteur antigénique chimérique ciblant gpc-3 et cellules immunitaires exprimant celui-ci pour des utilisations thérapeutiques
WO2023248126A1 (fr) Récepteur antigénique chimérique spécifique de cd117
WO2023084399A1 (fr) Cellules immunitaires génétiquement modifiées exprimant des récepteurs antigéniques chimériques masqués spécifiques de la protéine tyrosine kinase 7

Legal Events

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

Ref document number: 22840819

Country of ref document: EP

Kind code of ref document: A1