WO2013074916A1 - Car+ t cells genetically modified to eliminate expression of t- cell receptor and/or hla - Google Patents

Car+ t cells genetically modified to eliminate expression of t- cell receptor and/or hla Download PDF

Info

Publication number
WO2013074916A1
WO2013074916A1 PCT/US2012/065506 US2012065506W WO2013074916A1 WO 2013074916 A1 WO2013074916 A1 WO 2013074916A1 US 2012065506 W US2012065506 W US 2012065506W WO 2013074916 A1 WO2013074916 A1 WO 2013074916A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
antigen
receptor
tcr
Prior art date
Application number
PCT/US2012/065506
Other languages
French (fr)
Inventor
Laurence J. Neil COOPER
Hiroki TORIKAI
Original Assignee
Board Of Regents, The University Of Texas System
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 Board Of Regents, The University Of Texas System filed Critical Board Of Regents, The University Of Texas System
Priority to US14/358,828 priority Critical patent/US10391126B2/en
Publication of WO2013074916A1 publication Critical patent/WO2013074916A1/en
Priority to US16/504,427 priority patent/US11344577B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001103Receptors for growth factors
    • A61K39/001104Epidermal growth factor receptors [EGFR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001103Receptors for growth factors
    • A61K39/001106Her-2/neu/ErbB2, Her-3/ErbB3 or Her 4/ErbB4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001103Receptors for growth factors
    • A61K39/001109Vascular endothelial growth factor receptors [VEGFR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001103Receptors for growth factors
    • A61K39/00111Hepatocyte growth factor receptor [HGFR or c-met]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001111Immunoglobulin superfamily
    • A61K39/001112CD19 or B4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001116Receptors for cytokines
    • A61K39/001117Receptors for tumor necrosis factors [TNF], e.g. lymphotoxin receptor [LTR] or CD30
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001116Receptors for cytokines
    • A61K39/001119Receptors for interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001124CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001129Molecules with a "CD" designation not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001148Regulators of development
    • A61K39/00115Apoptosis related proteins, e.g. survivin or livin
    • A61K39/001151Apoptosis related proteins, e.g. survivin or livin p53
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001154Enzymes
    • A61K39/001164GTPases, e.g. Ras or Rho
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001169Tumor associated carbohydrates
    • A61K39/00117Mucins, e.g. MUC-1
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001169Tumor associated carbohydrates
    • A61K39/001171Gangliosides, e.g. GM2, GD2 or GD3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001174Proteoglycans, e.g. glypican, brevican or CSPG4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00118Cancer antigens from embryonic or fetal origin
    • A61K39/001181Alpha-feto protein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00118Cancer antigens from embryonic or fetal origin
    • A61K39/001182Carcinoembryonic antigen [CEA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00119Melanoma antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4632T-cell receptors [TCR]; antibody T-cell receptor constructs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464412CD19 or B4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • 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
    • A61K2239/26Universal/off- the- shelf cellular immunotherapy; Allogenic cells or means to avoid rejection
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the field of the invention includes at least medicine, immunology, ceil biology, and molecular biology.
  • the fseld of the invention includes immunotherapy .
  • HSCT hematopoietic stem-cell transplantation
  • donor-derived T cells can be genetically modified to express a tumor-specific chimeric antigen receptor (CAR) with specificity derived, for example, from the variable domains of a monoclonal antibody, thus focusing immimoreactivity towards the tumor in an MHC non-restricted manner (Cooper el al, 2004).
  • CAR tumor-specific chimeric antigen receptor
  • TCR ⁇ T-cell receptor
  • GVHD graft- versus-host-disease
  • the Sleeping Beauty (SB) transposon/transposase system was adapted for human application, e.g., to stably express a CD19-specific CAR (designated CD19RCD28) (Kohn et al, 201 1 ; Tzsvak et al, 2010;98ett et al, 2010; Williams, 2008; see U.S. Patent No. 6,489,458, incorporated by reference herein in its entirety).
  • CD19RCD28 Designated CD19RCD28
  • SB modified CAR 1" T cells can be numerically expanded to clinically-sufficient numbers by the recursive addition of ⁇ -irradiated designer artificial antigen presenting cells (aAPC) that co-express CD 19 and desired T-cell co-stimulatory molecules (Singh et al, 2008; Davies et al, 2010). This has been adapted for clinical translation at M.D.
  • aAPC ⁇ -irradiated designer artificial antigen presenting cells
  • the present invention is directed to methods and/or compositions for use in medicine, for example, for use in immunotherapy, including immunotherapy for infection or cancer, for example.
  • universal T cell-based immunotherapy including T cells engineered to express an antigen-specific chimeric antigen receptor (CAR) and to eliminate expression of endogenous alpha/beta T-cell receptor (TCR).
  • CAR antigen-specific chimeric antigen receptor
  • TCR endogenous alpha/beta T-cell receptor
  • CAR-'- T cells genetically modified to eliminate expression of TCR.
  • ZFNs zinc finger nucleases
  • there is knocking out of the T-cell receptor ⁇ -chain in CAR-expressing T cells for example using zinc finger nucleases.
  • T cells there are universal CAR-expressing T cells, such as from a healthy donor, that may be suitably stored, for example, in the freezer, and then infused into allogeneic individuals on demand. This allows interested parties to prepare and validate genetically modified antigen-specific T cells before individuals need them. These T cell products significantly broaden the application of antigen- specific T-eeil therapy, such as for cancer and infection, for example.
  • the invention provides off- the-shelf universal CAR " T cells from allogeneic healthy donors that can be administered to any patient without causing GVHD.
  • An advantage to the methods and compositions of the present invention is that ihe modified cells allow interrogation of ihe antigen independence of MHC and therefore the cells are suitable for any genetic background in the recipient.
  • the CAR h TCR uSg cells are autologous.
  • any kind of autoimmune disease, infection, and cancer can be treated with methods and/or compositions of the invention, including primary, metastatic, recurrent, sensitive-to-therapy, refractor -to -therapy, and so forth.
  • the methods and/or compositions are employed for chemo-refractory cancer.
  • the cancer may be of the blood, lung, brain, colon, prostate, breast, liver, kidney, stomach, cervix, ovary, testes, pituitary gland, esophagus, spleen, skin, bone, and so forth.
  • T ceils that continue to express TCR by using, for example, clinical-grade CDS -specific monoclonal antibody such that a T-cell product can be generated in which at least the majority have lost expression of endogenous TCR.
  • Other ways include the use of fluorescence-activated ceil sorting (FACS) and TCR- specific monoclonal antibodies or aptamers to reduce and possibly eliminate the presence of T cells that continue to express TCR.
  • FACS fluorescence-activated ceil sorting
  • TCR-specific monoclonal antibodies or aptamers to reduce and possibly eliminate the presence of T cells that continue to express TCR.
  • TCR-specific antibodies can be combined with other purification schemes, such as addition of complement, toxins, or resetting to reduce/eliminate T cells that continue to express TCR.
  • compositions encompassed by the invention there are methods of treating an individual in need of treatment using compositions encompassed by the invention.
  • the treatment includes employing particular T cells having a CAR and also having genetic modifications to exclude functional TCR.
  • modifications of the cells exclude functional TCR by any suitable means in certain aspects the modifications include knock out (or in alternative embodiments, knock down, such as by siRNA and TALENs, for example) of the a and/or ⁇ chains or the gamma and/or delta chains of the TCR.
  • a method of making a cell expressing a CAR comprising introducing an expression cassette in to the ceil, wherein the expression cassette encodes a polypeptide comprising a heterologous human extracellular antigen binding domain, a transmembrane domain, and one or more an intracellular signaling domain(s).
  • the methods further comprise stimulating the cells with antigen presenting cells, recombinant antigen, or an antibody to the receptor to cause the cells to proliferate, kill, and/or make cytokines.
  • the method further comprises stimulating the cells with antigen presenting cells to cause the cells to proliferate.
  • antigen-specific TCR eg cells expressing and bearing on the cell surface membrane an antigen-specific CAR human polypeptide comprising an intracellular signaling domain, a transmembrane domain and an extracellular domain, the extracellular domain comprising a human anti-antigen monoclonal antibody or antigen binding fragment thereof.
  • a method of treating a human disease condition associated with a cell expressing endogenous CD 19 comprising infusing a patient with an amount of a recombinant TCR ' * 8 ceil expressing a human antigen-specific CAR (such as CD19-specific) sufficient to treat the condition, wherein the human antigen-specific CAR comprises a heterologous human CD 19 extracellular binding domain, a transmembrane domain, and an intracellular signaling domain.
  • more than one pair of zinc finger nucleases can be used to modify a cell. For example, both zinc finger nucleases targeting the alpha chain and the beta chain can be used to eliminate T-cell receptor expression.
  • the zinc finger nucleases targeting the T-cell receptor can be used (for example, sequentially) with zinc finger nucleases to target one or more human leukocyte antigen(s) (HLA).
  • HLA human leukocyte antigen
  • T-cell population wherein cells of the population comprise an endogenous T-cell receptor coding sequence that is either not expressed or which encodes a nonfunctional T-cell receptor; and a recombinant chimeric antigen receptor comprising an intracellular signaling domain, a transmembrane domain, and an extracellular domain comprising an antigen binding region.
  • the T-ceil receptor is nonfunctional by virtue of one or more disruptions in the coding sequence of a chain, ⁇ chain, or both.
  • the endogenous T-celi receptor is knocked out.
  • the antigen binding region is an F(ab')2, Fab', Fab, Fv, or scFv and/or the antigen binding region binds a tumor associated antigen, such as CD 19, CD20, ROR] , carcinoembryonic antigen, alphafetoprotein, CA- 125, MUC- 1 , epithelial tumor antigen, melanoma-associated antigen, mutated p53, mutated ras, HER2/Neu, ERBB2, FTER3, folate binding protein, FlTV- l envelope glycoprotein gpl20, HIV- 1 envelope glycoprotein gp41 , GD2, CD 123, CD23.CD30 , CD56, c-Mef mesothelin, GD3, HERV-K, IL- 1 IRalpha, IL- 13Ralpha2, kappa chain, or lambda chain, CSPG4 (also known as, high molecular weight melanoma
  • the antigen binding region binds a pathogen antigen, such as a fungal, viral, or bacterial antigen.
  • a pathogen antigen such as a fungal, viral, or bacterial antigen.
  • the fungal antigen is from Aspergillus or Candida.
  • the viral antigen is from HSV, RSV, EBV, CMV, JC virus, BK virus, or Ebola.
  • the intracellular signaling domain comprises CD3 , CD28, OX40/CD134, 4- 1BB/CD137, FceRIy, ICOS/CD278, ILRB/CD 122, IL--2RG/CD 132, DAP molecules, CD70, cytokine receptor, CD40, or a combination thereof.
  • the transmembrane domain comprises IgG4Fc hinge, Fc regions, CD4 transmembrane domain, CD28 transmembrane domain, the CD3 transmembrane domain, cysteine mutated hitman € ⁇ 3 ⁇ domain, CD 16 transmembrane domain, CDS transmembrane domain, or erythropoietin receptor transmembrane domain.
  • there is an exemplary chimeric antigen receptor that comprises: a) an antigen binding domain from a variable region of an antigen-specific monoclonal antibody; b) a signaling domain of 033 ⁇ ; and c) CD28, CD 137, CD 134, or combinations thereof.
  • step b) occurs before step c) or step c) occurs before step b).
  • the T cell is provided from an umbilical cord blood bank, is provided from a peripheral blood bank, is an induced pluripotent stem cell, or is a human embryonic stem ceil. In some cases, the T cell is allogeneic in reference to one or more intended recipients.
  • the chimeric antigen receptor is stably introduced into the cell, and in certain embodiments, the polynucleotide that encodes the chimeric antigen receptor is introduced into the cell by a transposon/transposase system or a viral-based gene transfer system, such as by recombinant retrovirus or lentivirus.
  • the T cells following modification of the T cell(s), they are propagated by exposing the T cells to artificial antigen presenting cells, by using OK ' T ' 3 (or equivalent to cross-fink CDS) optionally with other co-stimulatory antibodies (e.g., anti- CD2S) on beads, or by using OKT3 (or equivalent to cross-link CD3) optionally with other co-stimulatory antibodies (e.g., anti-CD28) mixed with peripheral blood mononuclear cells.
  • OK ' T ' 3 or equivalent to cross-fink CDS
  • other co-stimulatory antibodies e.g., anti- CD2S
  • OKT3 or equivalent to cross-link CD3
  • other co-stimulatory antibodies e.g., anti-CD28
  • a polynucleotide that encodes the recombinant chimeric antigen receptor is electroporated into the T cell, in some embodiments, a polynucleotide that encodes the chimeric antigen receptor is present on a plasmid or viral vector.
  • the T cell can be genetically modified with zinc finger nuclease or TLAE nuclease to eliminate HLA expression.
  • the T cell may express a CAR and/or have been modified to eliminate TCR expression.
  • the endogenous T-cell receptor and/or HLA is disrupted by nonhomologous end joining repair, such as is generated by zinc finger nuclease, TALE nuclease, introduced into the cell by physical means, electro-transfer of mRNA species, viral vector, or non-viral vector,
  • nonhomologous end joining repair such as is generated by zinc finger nuclease, TALE nuclease
  • a medical condition such as autoimmune disease, cancer, or infection, including Aspergillus or Candida
  • a medical condition such as autoimmune disease, cancer, or infection, including Aspergillus or Candida
  • the cancer is lymphoma, leukemia, non-Hodgkin's lymphoma, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, chronic lymphocytic leukemia, or B cell-associated autoimmune diseases.
  • FIG. 1 ZFN pairs targeting sites within genomic iocs of TCR- and ⁇ constant region. Each exon is shown by a block. Black blocks represent coding regions. Grey columns represent non-coding regions.
  • One ZFN pair was designed to bind exon 1 of the TCR a constant region (IRAQ and another ZFN pair binds a conserved sequence on exon 1 of the TCR ⁇ constant regions 1 (TRBC1) and 2 (TRBC2). Underlined nucleotide sequences represent the intended binding sequence of each ZFN.
  • FIG. 2 Disruption of the TCR ⁇ -CDS complex in primary T ceils.
  • [0030] a. Schematic presentation of ZFN transfer. [0031] A pair of ZFN-encoding mRNA was electro-transferred into T cells six days after stimulation of CAR neg T cells, T cells were then cultured with 50 lU/mL of IL-2 and some cells were incubated at 30°C-5% C0 2 . CD3s expression was analyzed on days 7-9 after electroporation by flow cytometry. [0832] b. Down regulation of CDS after electro-transfer of ZFN targeting TCR ⁇ from mRNA.
  • Top panels shows CD3s expression in T cells cultured at 37°C after ZFN transfer and bottom panels shows CD3e expression in T cells transiently cultured at 30°C from days 2-3 after ZFN transfer.
  • c Surveyor Nuclease assay to detect ZFN-mediated modification of TCR target sites in T-cells.
  • TCR eg T ceils can be enriched by depletion of CD3s T cells.
  • the ⁇ usage clonogram was analyzed by a panel of TCR-specific mAbs, co- stained with CD4 and CDS. Percentage of specific Vp ' T-cell fractions within CD4 and CDS gating is shown. The nomenclatures of ⁇ repertoire shown are based on Arden et at (1995).
  • FIG. 4 EUmination of TCR fi-CBS complex from CD19-specific CAR '1 T cells. [ ⁇ 43] . Schematic of electro-transfer of tnRNA coding for ZFN pairs in CAR ' T cells.
  • mRNAs coding for ZFN pairs were electro-transferred into CAR 1 T cells, A pair of ZFN was electro-transfer to T cells two days after stimulation with CD19 h aAPC. After eiectroporation, ceils were maintained wiih 50 IU/inL of IL-2 and incubated for two days at 30°C-5% C0 2 . CD3s expression was analyzed nine days after eiectroporation by- flow cytometry.
  • FIG. 5 Functional consequences of TCR knockout in CA * ' T cells.
  • CAR "1" T cells Dilution of PKH26 was measured 10 days after stimulation with aAPC loaded with OKT3 (upper panel) or expressing CD 19 (lower panel). Flow cytometry data was gated on CAR "1" T cells.
  • CAR " T cells of an EL4 mouse T-ce!l line modified to express a truncated version of human CD 19 was measured by standard four hour J I Cr release assay. Specificity is shown by lack of lysis of CD19 eg (parental) EL4 cells (open symbols).
  • CAR " T cells were modified by ZFNs (TRAC and TRBC) or not modified by ZFNs (parental and no mRNA).
  • FIG. 6 Sustained proliferation of TCR neg CAR ⁇ T cells.
  • CAR + T cells with (TRAC and TRBC) or without (parental and no mRNA) TCR modification by ZFNs were stimulated with ⁇ -irradiated CD19 aAPC every two weeks. Viable T cells were enumerated every seven days and inferred total numbers were calculated.
  • FIG. 7 Schematic of the approach to genetically modify and propagate T cells from. PBMC.
  • T cells were propagated by stimulation with OKT3 loaded ⁇ - irradiated aAPC (clone #4) in the presence of soluble IL-2.
  • DNA plasmids coding for SB transposon (CD19RCD28) and SB iransposase (SB l l) were electro-transferred into primary human T cells.
  • CAR " T cells were selectively propagated by repeated additions of ⁇ - irradiated aAPC (clone #4) in the presence of rhIL-2.
  • Flow cytometry was used to compare expression of CD 19, CD64, CD86, CD137L, and MBIL-15 between parental K562 (grey shaded histogram) and K562-aAPC clone #4 (black open histogram).
  • MBIL-15 is composed of human IL- 15 peptide fused to modified human IgG4 Fc region and CD4 transmembrane domain and was detected by presence of EGFP (co-expressed with MBIL-15 after IRES element).
  • aAPC were used to co-culture T cells if expression of introduced iransgenes were 80%.
  • OKT3 was loaded onto aAPC clone #4 at 1 ⁇ tg per 10 h cells.
  • Flow cytometry data before and after OKT3 loading detected by antibody specific for mouse Fab region.
  • No-staining control grey shaded histogram; stain with anti-mouse Fab: black open histogram.
  • FIG. 9 Depletion of a CDSs-positive population. Representative results from multiple experiments using in vitro expanded T cells. Flow cytometry revealing expression of CD3s in CD4 and CD8 T cells. Numbers in the lower right quadrant represent the percentage of CD3s-negative ceils in CD4 1" and CD8 1" T-cell populations. [0059] FIG. 10. An exemplary illustration of interactions between donor and recipient cells in normal eases and in embodiments of the invention.
  • FIGS. 11a and lib Illustrations of use of particular zinc finger nucleases (ZF s) to achieve elimination of HLA in the genome.
  • FIGS. 12a, 12b, and 12c HLA- A targeted ZFNs are able to disrupt the HLA-
  • FIGS. 13a and 13b HLA-A targeting of ZFNs disrupting HLA-A. when they were expressed from in vitro transcribed mRNA (FIG. 13a).
  • the HLA-A ""1 ' T-cell population can be enriched (FIG. 13b).
  • FIGS. 14a, 14b, and 14c Disruption of HLA-A from exemplary CD19RCD28CA + T cells (FIG. 14a) including evasion of HLA-A restricted CTL attack (FIG. 14b) but maintenance of CD 19 specificity (FIG. 14c).
  • FIG. 15a TCR. expression can be disrupted from CD19RCD28CAR + T cells by TCR a or ⁇ constant region-targeted ZFN pairs.
  • FIGS. 15b and 15c TCR nuil CD19RCD28CAR i T cells maintain CD 19 specificity (FIG. 15b) and they can be propagated on CD 19-expressing K562-based artificial antigen presenting cells (FIG. 15c).
  • FIG. 16 Simultaneous knock out of HLA-A and TCR ⁇ .
  • Peripheral blood mononuclear cells from HLA-A2 -positive healthy donor were stimulated with OKT3 loaded artificial antigen presenting cells and cultured with 50 lU/mL of rh IL-2.
  • expanded T cells were electroporated with HLA-A- targeted or TCR ⁇ constant region (TRBC)-targeted ZFNs from mRNAs. Both TCR expression and HLA-A expression were evaluated by anti-CD38 monoclonal antibody (mAb) and anti-HLA-A.2 mAb four days after electroporation.
  • PI negative cells were positively gated according to CD4 and/or CDS. Numbers in the figure represent percentage of population in each quadrant.
  • aspects of the invention may "consist essentially of or “consist of” one or more elements or steps of the invention, for example. Some embodiments of the invention may consist of or consist essentially of one or more elements, method steps, and/or methods of the invention. It is contemplated that any method or composition described herein can be implemented with respect to any other method or composition described herein,
  • chimeric antigen receptors may be referred to as artificial T-ceil receptors, chimeric T-cell receptors, or chimeric immune-receptors, for example, and encompass engineered receptors that graft an artificial specificity onto a particular immune effector cell.
  • the CARs may be employed to impart the specificity of a monoclonal antibody onto a T cell, thereby allowing a large number of specific T cells to be generated, for example, in use for adoptive cell therapy.
  • the CARs direct specificity of the ceil to a tumor associated antigen, for example.
  • the CARs comprise an intracellular activation domain, a transmembrane domain and an extracellular domain comprising a tumor associated antigen binding region.
  • CARs comprise fusions of single-chain variable fragments (scFv) derived fro monoclonal antibodies, fused to CD3-zeta transmembrane and endodomain.
  • the specificity of other CARs designs may be derived from ligands of receptors (e.g., peptides) or from Dectins.
  • one can target malignant B cells by redirecting the specificity of T cells using a chimeric immunoreeeptor specific for the B- lineage molecule, CD 19.
  • the CARs comprise domains for additional co- stimulatory signaling, such as CD3-zeta, FcR, CD27, CD28, CD137, DAP 10, and/or OX40.
  • molecules can be co-expressed with the CAR. These include co-stimulatory molecules, reporter genes for imaging (e.g., for positron emission tomography), gene products that conditionally ablate the T cells upon addition of a pro-drug, homing receptors, cytokines, and cytokine receptors.
  • T-cell receptor refers to a protein receptor on T cells that is composed of a heterodimer of an alpha (a) and beta ( ⁇ ) chain, although in some cells the TCR consists of gamma and delta ( ⁇ / ⁇ ) chains.
  • the TCR may be modified on any cell comprising a TCR, including a helper T cell, a cytotoxic T cell, a memory T cell, regulatory T cell, natural killer T cell, and gamma delta T cell, for example.
  • Chimeric antigen receptors are designed for adoptive immunotherapy by connecting an extracellular antigen-binding domain to a transmembrane domain and an intracellular signaling domain (endodomain). It is a useful anti-tumor approach to eradicate tumor cells by adoptive transfer of T ceils expressing chimeric antigen receptors to recognize specific antigens presented on tumor cells and activate T cells to specifically lyse these tumor cells.
  • a critical aspect of this CAR strategy is the selection of target epitopes that are specifically or selectively expressed on tumors, are present on all tumor cells, and are membrane epitopes not prone to shed or modulate from the cell surface.
  • the CART T cells would be able to be used as a universal reagent or drug suitable for any mammalian (such as human) recipient.
  • any mammalian (such as human) recipient To employ the cells in such a manner, one must prevent their rejection in a graft -versus-host response without compromising CAR-dependeni effector functions.
  • T-cell receptor (TCR) ⁇ disruption from chimeric antigen receptor (CAR)-expressing T cells (CAR T cells) to establish "universal" T cell-based immu otherapy is provided.
  • Redirecting T-cell specificity to desired antigen can be achieved through CAR.
  • ex vivo generation of CAR T cells from patient is limited by time and expense.
  • T cells derived from patients are sometimes functionally flawed because of the multiple rounds of lymphotoxic (lympliodepleting) chemotherapy.
  • embodiments of the present invention concern the generation of CAR' T cells from healthy volunteer donor that can serve as "off-the-shelf reagents.
  • genetically modified T ceils from one donor can be pre -prepared and then infused into multiple recipients.
  • This will facilitate "centralized” manufacturing of the universal T cells and subsequent pre -positioning of the T cells at regional facilities for infusion on demand, enable clinical trials to be undertaken that are powered for efficacy, and facilitate combination therapies in which the universal T ceils can be administered with other biologies and therapeutics.
  • one can eliminate endogenous TCR ⁇ and ⁇ expression, which causes unwanted allogeneic immune reaction.
  • Such steps can occur by any suitable manner, including by introducing zinc finger nucleases (ZFN), for example, targeting TCR a constant region or ⁇ constant region.
  • ZFN zinc finger nucleases
  • Embodiments of the invention are unique as they combine (i) redirecting the specificity of T cells by introducing a CAR and (ii) eliminating expression of endogenous TCR to generate a desired T-cell product.
  • the introduction of CAR and elimination of TCR are accomplished by electroporation using the Sleeping Beauty (SB) system to stably express CAR and desired transient transfection of in vv ' iro-transcribed mRNA coding for ZFNs.
  • SB Sleeping Beauty
  • infusing specific CAR T TCR ne T cells are pre-prepared from healthy donors and thawed to be infused on demand as an off-the-shelf reagent.
  • the inventors demonstrate that ZFNs targeting either the a or ⁇ chains of endogenous TCRs in T cells resulted in the desired loss of TCR expression. As expected, these modified T cells did not respond to TCR stimulation, but maintained their CAR mediated re-directed specificity for the exemplary antigen, CD 19.
  • clinical-grade T-cells are genetically modified ex vivo to express a chimeric antigen receptor (CAR) to redirect specificity to a tumor associated antigen (TAA) thereby conferring anti-tumor activity in vivo.
  • CAR chimeric antigen receptor
  • TAA tumor associated antigen
  • T cells expressing a CDI9-specific CAR recognize B-cell malignancies in multiple recipients independent of MHC because the specificity domains are cloned from the variable chains of a CD 19 monoclonal antibody.
  • the present invention encompasses a major step towards eliminating the need to generate patient-specific T cells by generating "universal" allogeneic TAA-specific T cells from one donor that might be administered to multiple recipients.
  • CD 19-specific CAR 1" T cells to eliminate expression of the endogenous ⁇ T-cell receptor (TCR) to prevent a graft-versus-host response without compromising CAR-dependent effector functions.
  • TCR endogenous ⁇ T-cell receptor
  • Genetically modified T cells were generated using the Sleeping Beauty system to stably introduce the CD 19-specific CAR with subsequent permanent deletion of a or ⁇ TCR chains with designer zinc finger nucleases. The inventors show that these engineered T cells display the expected property of having redirected specificity for CD 19 without responding to TCR stimulation.
  • These CAR CR eg T cells may be used as off-the-shelf therapy for investigational treatment of B-lineage malignancies.
  • ZFNs zinc finger nucleases
  • NHEJ non-homologous end joining
  • an antigen is a molecule capable of being bound by an antibody or T-cell receptor.
  • An antigen is additionally capable of inducing a humoral immune response and/or cellular immune response leading to the production of B and/or T lymphocytes.
  • the present invention involves nucleic acids, including nucleic acids encoding an antigen-specific chimeric antigen receptor (CAR), including a CAR that has been humanized to reduce immunogenicity (hCAR), polypeptide comprising an intracellular signaling domain, a transmembrane domain, and an extracellular domain comprising one or more signaling motifs.
  • CAR antigen-specific chimeric antigen receptor
  • the CAR may recognize an epitope comprised of the shared space between one or more antigens.
  • the binding region can comprise complementary determining regions of a monoclonal antibody, variable regions of a monoclonal antibody, and/or antigen binding fragment thereof.
  • that specificity is derived from a peptide (e.g., cytokine) that binds to a receptor.
  • a complementarity determining region is a short amino acid sequence found in the variable domains of antigen receptor ⁇ e.g., immunoglobulin and T-cell receptor) proteins that complements an antigen and therefore provides the receptor with its specificity for that particular antigen.
  • Each polypeptide chain of an antigen receptor contains three CDRs (CDR1, CDR2, and CDR3). Since the antigen receptors are typically composed of two polypeptide chains, there are six CDRs for each antigen receptor that can come into contact with the antigen— each heavy and light chain contains three CDRs.
  • CDR3 shows the greatest variability as it is encoded by a recombination of the VJ (VDJ in the case of heavy chain and TCR ⁇ chain) regions.
  • VJ VDJ in the case of heavy chain and TCR ⁇ chain
  • the invention includes a full length CAR cDNA or coding region.
  • the antigen binding regions or domain can comprise a fragment of the VH and VL chains of a single-chain variable fragment (scFv) derived from a particular human monoclonal antibody, such as those described in U.S. Patent 7, 109,304.
  • the fragment can also be any number of different antigen binding domains of a human antigen-specific antibody.
  • the fragment is an antigen- specific scFv encoded by a sequence that is optimized for human codon usage for expression in human cells.
  • the arrangement could be multimeric, such as a diabody or multimers.
  • the multimers are most likely formed by cross pairing of the variable portion of the light and heavy chains into what has been referred to by Winters as a diabody.
  • the hinge portion of the construct can have multiple alternatives from being totally deleted, to having the first cysteine maintained, to a proline rather than a serine substitution, to being truncated up to the first cysteine.
  • the Fc portion can be deleted, although there is data to suggest that the receptor preferably extends from the membrane. Any protein that is stable and dimerizes can serve this purpose.
  • the intracellular signaling domain of the chimeric receptor of the invention is responsible for activation of at least one of the normal effector functions of the immune ceil in which the chimeric receptor has been placed.
  • effector function refers to a specialized function of a differentiated cell. Effector function of a T cell, for example, may be cytolytic activity or helper activity including the secretion of cytokines. Effector function in a memory or memory-type T cell includes antigen-dependent proliferation.
  • intracellular signalmg domain refers to the portion of a protein that transduces the effector function signal and directs the cell to perform a specialized function.
  • intracellular signaling domain While usually the entire intracellular signaling domain will be employed, in many eases it will not be necessary to use the entire intracellular polypeptide. To the extent that a trancated portion of the intracellular signaling domain may find use, such truncated portion may be used in place of the intact chain as long as it still transduces the effector function signal.
  • intracellular signaling domain is thus meant to include any trancated portion of the intracellular signaling domain sufficient to transduce the effector function signal.
  • Examples include the zeta chain of the T-cell receptor or any of its homo logs (e.g., eta, delta, gamma, or epsilon), MB1 chain, B29, Fc RUT, Fc R , and combinations of signaling molecules, such as ⁇ 3 ⁇ and CD2.8, 4- 1BB, OX40, and combination thereof, as well as other similar molecules and fragments.
  • Intracellular signaling portions of other members of the families of activating proteins can be used, such as FcyRIII and FcsRL See Gross el al. ( 1992), Stancovski el al. (1993), Moritz et al. (1994), Hwu el al. (1995), Weijtens et al. ( 1996), and Hekele et al (1996) for disclosures of cTCR's using these alternative transmembrane and intracellular domains.
  • the human CD3 ⁇ intracellular domain was taken for activation.
  • the antigen-specific extracellular domain and the intracellular signaling- domain may be linked by a transmembrane domain, such as the human IgG 4 Fe hinge and Fc regions.
  • a transmembrane domain such as the human IgG 4 Fe hinge and Fc regions.
  • Alternatives include the human CD4 transmembrane domain, the human CD28 transmembrane domain, the transmembrane human CD3 domain, or a cysteine mutated human € ⁇ 3 ⁇ domain, or other transmembrane domains from other human transmembrane signaling proteins, such as CD 16 and CD8 and erythropoietin receptor.
  • the CAR nucleic acid comprises a sequence encoding other costimulatory receptors, such as a transmembrane domain and a modified CD28 intracellular signaling domain.
  • costimulatory receptors include, but are not limited to one or more of CD28, OX-40 (CD 134), DAP 10, and 4- IBB (CD137).
  • CD28 CD28
  • OX-40 CD 134
  • DAP DAP
  • 4- IBB CD137
  • an additional signal provided by a human costimulatory receptor inserted in a human CAR is important for full activation of T cells and could help improve in vivo persistence and the therapeutic success of the adoptive immunotherapy.
  • the invention concerns isolated nucleic acid segments and expression cassettes incorporating DNA sequences that encode the CAR.
  • Vectors of the present invention are designed, primarily, to deliver desired genes to immune cells, preferably T cells under the control of regulated eukaryotic promoters, for example, MNDU3 promoter or EFlapha promoter, or Ubiquitin promoter. Also, the vectors may contain a selectable marker if. for no other reason, to facilitate their manipulation in vitro.
  • regulated eukaryotic promoters for example, MNDU3 promoter or EFlapha promoter, or Ubiquitin promoter.
  • the vectors may contain a selectable marker if. for no other reason, to facilitate their manipulation in vitro.
  • Chimeric antigen receptor molecules are recombinant and are distinguished by their ability to both bind antigen and transduce activation signals via immunoreeeptor activation motifs (ITAM's) present in their cytoplasmic tails.
  • Receptor constructs utilizing an antigen-binding moiety afford the additional advantage of being "universal” in that they bind native antigen on the target cell surface in an HLA-mdependent fashion.
  • non-human antigen binding regions are typically used in constructing a chimeric antigen receptor.
  • a potential problem with using non-human antigen binding regions, such as murine monoclonal antibodies, is the lack of human effector functionality and inability to penetrate into tumor masses. In other words, such antibodies may be unable to mediate complement-dependent lysis or iyse human target ceils through antibody- dependent cellular toxicity or Fc-receptor mediated phagocytosis to destroy cells expressing CAR.
  • non-human monoclonal antibodies can be recognized by the human host as a foreign protein, and therefore, repeated injections of such foreign antibodies can lead to the induction of immune responses leading to harmful hypersensitivity reactions.
  • the chimeric antigen receptor comprises: a) an intracellular signaling domain, b) a transmembrane domain, and c) an extracellular domain comprising an antigen binding region,
  • intracellular receptor signaling domains in the CAR include those of the T cell antigen receptor complex, such as the zeta chain of CD3, also Fey RIII costimulatory signaling domains, CD28, DAP 10, CD2, alone or in a series with CD3zeta, for example.
  • the intracellular domain (which may be referred to as the cytoplasmic domain) comprises part or all of one or more of TCR zeta chain, CD28, OX40/CD134, 4-1BB/CD137, FcsRTy, ICOS/CD278, ILRB/CD122, IL- 2RG/CD132, DAP molecule, CD27, DAP 10, DAP 12, and CD40.
  • one employs any part of the endogenous T cell recepior complex in the intracellular domain.
  • One or multiple cytoplasmic domains may be employed, as so-called third generation CARs have at least two or three signaling domains fused together for additive or synergistic effect, for example.
  • the antigen-specific portion of the recepior (which may be referred to as an extracellular domain comprising an antigen binding region) comprises a tumor associated antigen or a pathogen-specific antigen.
  • a tumor associated antigen may be of any kind so long as it is expressed on the cell surface of tumor cells.
  • Exemplary embodiments of tumor associated antigens include CD 19, CD20, carcinoembryonic antigen, alphafetoprotein, CA- 125, MUC-1, epithelial tumor antigen, melanoma- associated antigen, mutated p53, mutated ras, and so forth.
  • intracellular tumor associated antigens may be targeted, such as HA- 1, surviving, WT1, p53. This can be achieved by a CAR. expressed on a universal T cell that recognizes the processed peptide described from the intracellular tumor associated antigen in the context of HLA.
  • the universal T cell may be genetically modified to express a T-cell receptor pairing that recognizes the intracellular processed tumor associated antigen in the context of HL A.
  • the pathogen may be of any kind, but in specific embodiments the pathogen is a fungus, bacteria, or vims, for example.
  • exemplary viral pathogens include those of the families of Adenoviridae, Epstein-Barr virus (EBV), Cytomegalovirus (CMV), Respiratory Syncytial Virus (RSV), JC virus, BK virus, HSV, HHV family of viruses, Picornaviridae, Herpesviridae, Hepadnaviridae, Flaviviridae, Retroviridae, Orthomyxoviridae, Parainyxoviridae, Papovaviridae, Polyomavirus, Rhabdoviridae, and Togavkidae.
  • Exemplary pathogenic viruses cause smallpox, influenza, mumps, measles, chickenpox, ebola, and rubella.
  • Exemplary pathogenic fungi include Candida, Aspergillus, Cryplococcus , Histoplasma, Pneumocystis, and Stachybotrys .
  • Exemplary pathogenic bacteria include Streptococcus, Pseudomonas, Shigella, Campylobacter, Staphylococcus, Helicobacter, E, coli, Rickettsia, Bacillus, Bordetella, Chlamydia, Spirochetes, and Salmonella.
  • the pathogen receptor Dectin-1 can be used to generate a CAR that recognizes the carbohydrate structure on the ceil wall of fungi.
  • T ceils genetically modified to express the CAR based on the specificity of Dectin- 1 can recognize Aspergillus and target hyphal growth.
  • CARs can be made based on an antibody recognizing viral determinants ⁇ e.g., the glycoproteins from CMV and Ebola) to interrupt viral infections and pathology.
  • the pathogenic antigen is an Aspergillus carbohydrate antigen for which the extracellular domain in the CAR recognizes patterns of carbohydrates of the fungal cell wall.
  • A. chimeric immunoreceptor according to the present invention can be produced by any means known in the art, though preferably it is produced using recombinant DMA techniques.
  • a nucleic acid sequence encoding the several regions of the chimeric receptor can be prepared and assembled into a complete coding sequence by standard techniques of molecular cloning (genomic library screening, PCR, primer-assisted ligation, scFv libraries from yeast and bacteria, site-directed mutagenesis, etc. ).
  • the resulting coding region can be inserted into an expression vector and used to transform a suitable expression host allogeneic TCR !,es cell line.
  • nucleic acid construct or nucleic acid sequence or polynucleotide is intended to mean a DNA molecule that can be transformed or introduced into a T cell and be transcribed and translated to produce a product (e.g., a chimeric receptor).
  • the promoter is operablv linked to the nucleic acid sequence encoding the chimeric receptor of the present invention, i.e., they are positioned so as to promote transcription of the messenger R A from the DNA encoding the chimeric receptor.
  • the promoter can be of genomic origin or synthetically generated.
  • a variety of promoters for use in T cells are well-known in the art (e.g., the CD4 promoter disclosed by Marodon et a I. (2003)).
  • the promoter can be constitutive or inducible, where induction is associated with the specific cell type or a specific level of maturation, for example.
  • Promoters of interest include the ⁇ -actin promoter, SV40 early and late promoters, immunoglobulin promoter, human cytomegalovirus promoter, retrovirus promoter, and the Friend spleen focus-forming virus promoter.
  • the promoters may or may not be associated with enhancers, wherein the enhancers may be naturally associated with the particular promoter or associated with a different promoter.
  • the sequence of the open reading frame encoding the chimeric receptor can be obtained from a. genomic DNA source, a cDNA source, or can be synthesized ⁇ e.g., via PCR), or combinations thereof.
  • cDNA or a combination thereof it may be desirable to use cDNA or a combination thereof as it is found that introns stabilize the mRNA or provide T cell-specific expression (Barthel and Goldfeki, 2003). Also, it may be further advantageous to use endogenous or exogenous non-coding regions to stabilize the mRNA.
  • a chimeric receptor of the present invention For expression of a chimeric receptor of the present invention, the naturally occurring or endogenous transcriptional initiation region of the nucleic acid sequence encoding N-terminai components of the chimeric receptor can be used to generate the chimeric receptor in the target host.
  • an exogenous transcriptional initiation region can be used that allows for constitutive or inducible expression, wherein expression can be controlled depending upon the target host, the level of expression desired, the nature of the t arget host, and the like.
  • a signal sequence directing the chimeric receptor to the surface membrane can be the endogenous signal sequence of N -terminal component of the chimeric receptor.
  • the signal sequence selected should be compatible with the secretory pathway of T ceils so that the chimeric receptor is presented on the surface of the T cell.
  • a termination region may be provided by the naturally occurring or endogenous transcriptional termination region of the nucleic acid sequence encoding the C- terminal component of the chimeric receptor. Alternatively, the termination region may be derived from a different source.
  • the source of the termination region is generally not considered to be critical to the expression of a recombinant protein and a wide variety of termination regions can be employed without adversely affecting expression.
  • a few amino acids at the ends of the antigen binding domain in the CAR can be deleted, usually not more than 10, more usually not more than 5 residues, for example.
  • the deletion or insertion of amino acids may be as a result of the needs of the construction, providing for convenient restriction sites, ease of manipulation, improvement in levels of expression, or the like.
  • the substitute of one or more amino acids with a different amino acid can occur for similar reasons, usually not substituting more than about five amino acids in any one domain.
  • the chimeric construct that encodes the chimeric receptor according to the invention can be prepared in conventional ways. Because, for the most part, natural sequences may be employed, the natural genes may be isolated and manipulated, as appropriate, so as to allow for the proper joining of the various components. Thus, the nucleic acid sequences encoding for the N-terminal and C-terminal proteins of the chimeric receptor can be isolated by employing the polymerase chain reaction (PGR), using appropriate primers that result in deletion of the undesired portions of the gene. Alternatively, restriction digests of cloned genes can be used to generate the chimeric construct. In either case, the sequences can be selected to provide for restriction sites that are blunt-ended, or have complementar overlaps.
  • PGR polymerase chain reaction
  • the various manipulations for preparing the chimeric construct can be carried out in vitro and in particular embodiments the chimeric construct is introduced into vectors for cloning and expression in an appropriate host using standard transformation or transfection methods.
  • the resulting construct from joining of the DNA sequences is cloned, the vector isolated, and the sequence screened to ensure that the sequence encodes the desired chimeric receptor.
  • the sequence can be screened by restriction analysis, sequencing, or the like.
  • the chimeric constructs of the present invention find application in subjects having or suspected of having cancer by reducing the size of a tumor or preventing the growth or re-growth of a tumor in these subjects.
  • the present invention further relates to a method for reducing growth or preventing tumor formation in a subject by introducing a chimeric construct of the present invention into an isolated T cell of the subject and reintroducing into the subject the transformed T ceil, thereby effecting anti-tumor responses to reduce or eliminate tumors in the subject.
  • Suitable T cells include cytotoxic lymphocytes (CTL) or any cell having a T cell receptor in need of disruption.
  • CTL cytotoxic lymphocytes
  • various methods are readily available for isolating these cells from a subject. For example, using cell surface marker expression or using commercially available kits (e.g., ISOCELLTM from Pierce, Rockford, ill.).
  • the chimeric construct can be introduced into the subject's own T cells as naked DNA or in a suitable vector.
  • Methods of stably iransfeciing T cells by eieciroporation using naked DNA are known in the art. See, e.g., U.S. Pat. No. 6,410,319.
  • naked DNA generally refers to the DNA encoding a chimeric receptor of the present invention contained in a plasmid expression vector in proper orientation for expression.
  • the use of naked DNA reduces the time required to produce T cells expressing the chimeric receptor of the present invention.
  • a viral vector e.g., a retroviral vector, adenoviral vector, adeno-associated viral vector, or lentiviral vector
  • Suitable vectors for use in accordance with the method of the present invention are non-replicating in the subject's T cells.
  • a large number of vectors are known that are based on viruses, where the copy number of the virus maintained in the cell is low- enough to maintain the viability of the cell.
  • Illustrative vectors include the pFB-neo vectors (STRAT G ENE® ) disclosed herein as well as vectors based on HIV, SV40, EBV, HSV, or BPV.
  • the transfected or transduced T cell is capable of expressing the chimeric receptor as a surface membrane protein with the desired regulation and at a desired level, it can be determined whether the chimeric receptor is functional in the host cell to provide for the desired signal induction. Subsequently, the transduced T cells are reintroduced or administered to the subject to activate anti-tumor responses in the subject.
  • the transduced T cells according to the invention can be made into a pharmaceutical composition or made into an implant appropriate for administration in vivo, with appropriate carriers or diluents, which further can be pharmaceutically acceptable. The means of making such a composition or an implant have been described in the art (see, for instance.
  • the transduced T cells can be formulated into a preparation in semisolid or liquid form, such as a capsule, solution, injection, inhalant, or aerosol, in the usual ways for their respective route of administrat on. Means known in the art can be utilized to prevent or minimize release and absorption of the composition until it reaches the target tissue or organ, or to ensure timed-release of the composition. Desirably, however, a pharmaceutically acceptable form is employed that does not ineffectuate the cells expressing the chimeric receptor.
  • the transduced T cells can be made into a pharmaceutical composition containing a balanced salt solution, preferably Hanks' balanced salt solution, or normal saline.
  • CD19 a cell surface glycoprotein of the immunoglobulin superfamily, is a potentially attractive target for antibody therapy of B cell-associated malignancies.
  • This antigen is absent from hematopoietic stem cells, and in healthy individuals its presence is exclusively restricted to the B-lmeage and possibly some follicular dendritic cells (Scheuermann et a!.., 1995). In fact, it is present on B cells from the earliest recognizable B- lineage cells during development to B-cell blasts but is lost on maturation to plasma cells.
  • CD 19 is not shed from the ceil surface and rarely lost during neoplastic transformation (Scheuermann et ⁇ , 1995).
  • CD 19 primarily acts as a B cell co-receptor in conjunction with CD21 and CD81. Upon activation, the cytoplasmic tail of CD 19 becomes phosphorylated, which leads to binding by Src-famify kinases and recruitment of PI-3 kinase.
  • compositions of the invention include a human CD 19- specific chimeric T cell receptor (or chimeric antigen receptor, CAR) polypeptide (designated hCD 19CAR) comprising an intracellular signaling domain, a transmembrane domain, and an extracellular domain, the extracellular domain comprising a human CD 19 binding region.
  • the CD 19 binding region is an F(ab')2, Fab', Fab, Fv, or scFv.
  • the binding region may comprise an amino acid sequence that is at least, at most or about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the wild-type amino acid sequence.
  • the intracellular domain may comprise an intracellular signaling domain of human CO3 , and ma further comprise human CD28 intracellular segment.
  • the transmembrane domain is a CD28 transmembrane domain.
  • compositions of the invention include a nucleic acid encoding the polypeptide described above.
  • nucleic acid sequence is optimized for human codon usage.
  • compositions of the invention include cells expressing the polypeptide described herein, '
  • the T cell may comprise an expression cassette encoding hCD 19CAR polypeptide.
  • the expression cassette can be comprised in a non- viral vector, such as a transposon, or a human transposon, or recombinant variant thereof.
  • the expression cassette can be comprised in a viral vector or recombinant variant thereof.
  • the expression cassette can be genomically integrated or episomally maintained or expressed from mRNA.
  • the invention includes a method of making a T cell expressing a human CD 19 -specific CAR comprising introducing an expression cassette into the cell, wherein the expression cassette encodes a polypeptide comprising a human extracellular CD 19 binding domain, a transmembrane domain, and one or more intracellular signaling domain(s).
  • the method may further comprise stimulating the cells with CD 19 cells, recombinant CD 19, or an antibody to the receptor to cause ihe cells to proliferate, kill, and/or make cytokines; for example, the cells may be stimulated to proliferate or expand with CD 19 f artificial antigen presenting cells.
  • the invention includes methods of treating a human disease condition associated with a TCR neg cell expressing endogenous CD 19 comprising infusing a patient with an amount of a recombinant cell expressing a human CD 19--speciflc CAR sufficient to treat the condition, wherein the human CD19-specific CAR comprises a human CD 19 extracellular binding domain, a transmembrane domain, and an intracellular signaling domain.
  • the condition can be lymphoma, leukemia, Non-Hodgkin's lymphoma, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, chronic lymphocytic leukemia, or B cell-associated autoimmune diseases, for example.
  • the invention relates to the generation of a human CD 19-specific chimeric antigen receptor (hCD19RCD28 or hCAR).
  • hCD19RCD28 or hCAR chimeric antigen receptor
  • recombinant cells expressing hCAR have improved in vivo persistence and anti- tumor efficacy.
  • the human hCAR has a reduced immunogenicity compared to murine hCAR, which comprises a scFv segment derived from a murine CD19-specific monoclonal antibody (mAb),
  • mAb monoclonal antibody
  • Anti-tumor effects can be augmented by genetically modified cells, such as TCR rje8 cells rendered specific for CD 19.
  • T cell specificity is achieved by electrotransfer of an expression cassette encoding hC AR.
  • the hCAR may be a chimeric receptor comprising one or more activation endodomain(s), such as a CD3 ⁇ -derived activation domain. Additional T-cell activation motifs include, but are not limited to, CD28, OX-40, and 4-1 BB. In certain aspects the activation domain can also include a CD28 transmembrane and/or activation domain. In a further aspect the hCAR encoding region and/or expression cassette codon optimized for expression in human cells and subjects, e.g., in one embodiment the scFv region obtained from VH and VL sequences of a CD 19-specific human antibodies are incorporated into the CD19 binding segment of the hCAR (for example see U.S. Patent No.
  • the hCAR expression cassette is episomally maintained or integrated into the genome of the recombinant cell.
  • the expression cassette is comprised in a nucleic acid capable of integration by using an integrase mechanism, a viral vector, such as a retroviral vector, or a nonviral vector, such as a transposon mechanism.
  • the expression cassette is included in a transposon based nucleic acid.
  • the expression cassette is part of a two component Sleeping Beauty (SB) or piggyBac system that utilizes a transposon and transposase for enhanced non-viral gene transfer.
  • Recombinant hCAR expressing cells can be numerically expanded to clinically-meaningful numbers.
  • One example of such expansion uses artificial antigen presenting cells (aAPC).
  • Recombinant hCAR expressing cells can be verified and identified by flow cytometry and western blot analyses.
  • Recombinant hCAR expressing T cells, expressing a CD19-specific CAR can recognize and kill CD 19 expressing target cells.
  • hCAR can be expressed into Universal cells that can be infused across transplantation barriers to help prevent immunogenicity.
  • the hCAR can be used along with human genes for imaging (such as by positron emission tomography, PET) and conditional ablation of T cells, in the event of cytotoxicity.
  • the recombinant cells of the invention can be used in CD 19-specific cellular therapies.
  • the T ceils employed in methods and compositions lack functional T cell receptor. Although in some cases one could employ knockdown measures to impact TCR levels post-transcriptionally, such as with siRNA, in particular cases one utilizes knockout means to edit genomic DNA. In certain embodiments, one eliminates expression of the endogenous TCR. In particular embodiments, the system used to genetically modify the T cells employs stably introducing an expression construct that permits permanent deletion of a and/or ⁇ cha ins of TCR.
  • TCR recombinant restriction enzymes
  • TALENs are employed that include the TAL effector DNA -binding domain fused to a DNA cleavage domain.
  • the DNA -binding domain of the fusion protein are proteins secreted by Xanthomonas bacteria that comprise a highly conserved 33-34 amino acid sequence. The exception therein is at the 12th and 1 th amino acids that are highly variable (Repeat Variable Diresidue) and correlate with specific nucleotide recognition in a particular code. This correlation between amino acid sequence and DNA recognition provides the generation of specific DNA binding domains by choosing a particular combination of repeat segments containing the proper RVDs, for example,
  • TALENs can be used to alter the genome of T cells by inducing double-strand breaks (DSB) that the T cells respond to with repair mechanisms, Non- homologous end joining (NHEJ) rejoins DNA from both sides of a double-strand break in which there is little or no sequence overlap for annealing.
  • this repair mechanism beneficially induces errors in the genome via insertion, deletion, or chromosomal rearrangement such that the T-cell receptor encoded thereby is nonfunctional.
  • the ability to prevent T-cell receptor expression is dependent on the ability to disrupt the endogenous expression cassette.
  • T cell can be activated by as few as one T-celi receptor molecule, efforts to disrupt T-cell recepior expression at the level of post-transcriptional (or post-translational) modification (such as using interfering RNA species) are unlikely to be completely efficient leading to leakage and expression of the endogenous T-cell receptor.
  • post-transcriptional (or post-translational) modification such as using interfering RNA species
  • the only available method to prevent T-cell receptor expression is based on genetic disruption of the alpha and/or beta expression loci as can be achieved using designer zinc finger nucleases,
  • the invention includes a method of making and/or expanding the antigen-specific redirected TCR" 68 cells that comprises transfecting TCR eg cells with an expression vector containing a DNA constract encoding the hCAR, then stimulating the cells with CD 19+ cells, recombinant CD 19, or an antibody to the receptor to cause the cells to proliferate.
  • this invention is a method of stably transfecting and re-directing T ceils by electroporation, or other non-viral gene transfer (such as, but not limited to sonoporation) using naked DNA.
  • Most investigators have used viral vectors to carry heterologous genes into T cells.
  • naked DNA By using naked DNA, the time required to produce redirected T cells can be reduced.
  • naked DNA means DNA encoding a chimeric T-cell receptor (cTCR) contained in an expression cassette or vector in proper orientation for expression.
  • the electroporation method of this invention produces stable trans fectants that express and carry on their surfaces the chimeric TCR (cTCR).
  • Chimeric TCR means a receptor that is expressed by T cells and that comprises intracellular signaling, transmembrane, and extracellular domains, where ihe extracellular domain is capable of specifically binding in an MHC unrestricted manner an antigen that is not normally hound by a T-cell receptor in that manner. Stimulation of the T cells by the antigen under proper conditions results in proliferation (expansion) of the cells and/or production of U. ⁇ ?..
  • the exemplary CD19-specific chimeric receptor of this invention is an example of a chimeric TCR..
  • the method is applicable to transfection with chimeric TCRs that are specific for other target antigens, such as chimeric TCRs that are specific for HER2/Neu (Stance vski et al, 1993), ERBB2 (Moritz et at, 1994), folate binding protein (Hwu et al, 1995), renal cell carcinoma (Weitjens et al, 1996), and HIV-1 envelope glycoproteins gp! 20 and gp41 (Roberts et al, 1994).
  • chimeric TCRs that are specific for HER2/Neu (Stance vski et al, 1993), ERBB2 (Moritz et at, 1994), folate binding protein (Hwu et al, 1995), renal cell carcinoma (Weitjens et al, 1996), and HIV-1 envelope glycoproteins gp! 20 and gp41 (Roberts et al, 1994).
  • cell-surface target antigens include, but are not limited to, CD20, carcinoembryonic antigen, mesotheiin, c-Met, CD56, HERV-K, GD2, GD3, aiphafetoprotein, CD23, CD30, CD123, IL- l lRalpha, kappa chain, lambda chain, CD70, CA-125, MUC-1, EGFR and variants, epithelial tumor antigen, and so forth.
  • the T cells are primary human T cells, such as T cells derived from human peripheral blood mononuclear cells (PBMC), PBMC collected after stimulation with G-CSF, bone marrow, or umbilical cord blood. Conditions include the use of mRNA and DNA and eiectroporation. Following transfection the cells may be immediately infused or may be stored. In certain aspects, following transfection, the cells may be propagated for day s, weeks, or months ex vivo as a bulk population within about 1 , 2, 3, 4, 5 days or more following gene transfer into cells.
  • PBMC peripheral blood mononuclear cells
  • the transfectants are cloned and a clone demonstrating presence of a single integrated or episomally maintained expression cassette or plasmid, and expression of the chimeric receptor is expanded ex vivo.
  • the clone selected for expansion demonstrates the capacity to specifically recognize and lyse CD 19 expressing target cells.
  • the recombinant T cells may be expanded by stimulation with IL-2, or other cytokines that bind the common gamma-chain (e.g., IL-7, IL- 15, IL-21, and others).
  • the recombinant T cells may be expanded by stimulation with artificial antigen presenting cells.
  • the recombinant T cells may be expanded on artificial antigen presenting cell or with an antibody, such as OKT3, which cross links CD3 on the T cell surface. Subsets of the recombinant T cells may be deleted on artificial antigen presenting ceil or with an antibody, such as Campath, which binds CD52 on the T cell surface.
  • the genetically modified ceils may be cryopreserved.
  • T-cell propagation (survival) after infusion may be assessed by: (i) q-
  • This invention also represents the targeting of a B cell malignancy or disorder including B cells, with the cell-surface epitope being CD 19 ⁇ specific using a redirected immune T cell that is TCR n g .
  • Malignant B cells are an excellent target for redirected T cells, as B ceils can serve as imniunostimuiatory antigen-presenting cells for T cells.
  • CARTCR nes cells are delivered to an individual in need thereof, such as an individual that has cancer or an infection. The cells then enhance the individual's immune system to attack the respective cancer or pathogenic cells.
  • the individual is provided with one or more doses of the antigen-specific CAR " CR eg cells.
  • the duration between the admimstrations should be sufficient to allow time for propagation in the individual, and in specific embodiments the duration between doses is 1, 2, 3, 4, 5, 6, 7, or more days.
  • the source of the allogeneic T cells that are modified to include both a chimeric antigen receptor and that lack functional TCR may be of any kind, but in specific embodiments the cells are obtained from a bank of umbilical cord blood, peripheral blood, human embryonic stem ceils, or induced pluripotent stem ceils, for example.
  • the different banks will not share the same HLAs, so multiple banks may be employed.
  • Suitable doses for a therapeutic effect would be at least ⁇ or between about 10 s and about 10 1 cells per dose, for example, preferably in a series of dosing cycles.
  • An exemplary dosing regimen consists of four one-week dosing cycles of escalating doses, starting at least at about 10° ceils on Day 0, for example increasing incrementally up to a target dose of about 10'° cells within several weeks of initiating an intra-patient dose escalation scheme.
  • Suitable modes of administration include intravenous, subcutaneous, intracavitary (for example by reservoir-access device), intraperitoneal, and direct injection into a tumor mass, [08130]
  • a pharmaceutical composition of the present invention can be used alone or in combination with other well-established agents useful for treating cancer.
  • the pharmaceutical composition of the present invention can be delivered via various routes and to various sites in a mammalian, particularly human, body to achieve a particular effect.
  • a particular route can provide a more immediate and more effective reaction than another route.
  • intradermal delivery may be advantageously used over inhalation for the treatment of melanoma.
  • Local or systemic delivery can be accomplished by administration comprising application or instillation of the formulation into body cavities, inhalation or insufflation of an aerosol, or by parenteral introduction, comprising intramuscular, intravenous, intraportal, intrahepatic, peritoneal, subcutaneous, or intradermal administration.
  • a composition of the present invention can be provided in unit dosage form wherein each dosage unit, e.g. , an injection, contains a predetermined amount of the composition, alone or in appropriate combination with other active agents.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of the composition of the present invention, alone or in combination with other active agents, calculated in an amount sufficient to produce the desired effect, in association with a pharmaceutically acceptable diluent, carrier, or vehicle, where appropriate.
  • the specifications for the novel unit dosage forms of the present invention depend on the particular pharmacodynamics associated with the pharmaceutical composition in the particular subject.
  • an effective amount or sufficient number of the isolated transduced T ceils is present in the composition and introduced into the subject such that long-term, specific, anti-tumor responses are established to reduce the size of a tumor or eliminate tumor growth or regrowth than would otherwise result in the absence of such treatment.
  • the amount of transduced T cells reintroduced into the subject causes a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, or 100% decrease in tumor size when compared to otherwise same conditions wherein the transduced T cells are not present.
  • the amount of transduced T cells administered should take into account the route of administration and should be such that a sufficient number of the transduced T cells will be introduced so as to achieve the desired therapeutic response.
  • the amounts of each active agent included in the compositions described herein e.g., the amount per each cell to be contacted or the amount per certain body weight
  • the concentration of transduced T cells desirably should be sufficient to provide in the subject being treated at least from about 1 10" to about 1 x 10 9 transduced T cells, even more desirably, from about 1 x 10 ' ' to about 5 x 10 8 transduced T cells, although any suitable amount can be utilized either above, e.g., greater than 5 x 10 3 ⁇ 4 cells, or below, e.g., less than 1 x 10' cells.
  • the dosing schedule can be based on well-established cell-based therapies (see, e.g., Topalian and Rosenberg, 1987; U.S. Pat. No. 4,690,915), or an alternate continuous infusion strategy can be employed.
  • a medical disorder is treated by transfer of a redirected T cell that elicits a specific immune response.
  • B cell lineage malignancy or disorder is treated by transfer of a redirected T ceil that elicits a specific immune response.
  • the cells of the adaptive immune system are a type of leukocyte, called a lymphocyte.
  • B cells and T cells are the major types of lymphocytes.
  • B cells and T cells are derived from the same pl ripotent hematopoietic stem ceils, and are indistinguishable from one another until after they are activaied.
  • B cells play a large role in the humoral immune response, whereas T cells are intimately involved in cell-mediated immune responses. They can be distinguished from other lymphocyte types, such as B cells and NK cells by the presence of a special receptor on their cell surface called the T-celi receptor (TCR).
  • TCR T-celi receptor
  • B cells and T ceils are produced by stem cells in the bone marrow. T cells travel to and develop in the thymus, from which they derive their name. In humans, approximately l%-2% of the lymphocyte pool recirculates each hour to optimize the opportunities for antigen-specific lymphocytes to find their specific antigen within the secondary lymphoid tissues.
  • T lymphocytes arise from hematopoietic stem cells in the bone marrow, and migrate to the thymus gland to mature. T cells express a unique antigen binding receptor on their membrane (T-cell receptor), which can only recognize antigen in association with major histocompatibility complex (MHC) molecules on the surface of other ceils.
  • T-cell receptor a unique antigen binding receptor on their membrane
  • MHC major histocompatibility complex
  • T helper cells There are at least two populations of T cells, known as T helper cells, known as T helper cells, known as T helper cells, known as T helper cells and T cytotoxic cells. T helper cells and T cytotoxic cells are primarily distinguished by their display of the membrane bound glycoproteins CD4 and CDS, respectively. T helper cells secret various lymphokines that are crucial for the activation of B cells, T cytotoxic ceils, macrophages, and other cells of the immune system.
  • T helper cells secret various lymphokines that are crucial for the activation of B cells, T
  • CTLs cytotoxic T lymphocytes
  • Natural killer cells or NK cells are a type of cytotoxic lymphocyte that constitutes a major component of the innate immune system. NK cells play a major role in the rejection of tumors and cells infected by viruses. The cells kill by releasing small cytoplasmic granules of proteins called perforin and granzyme that cause the target cell to die by apoptosis.
  • a B cell identifies pathogens when antibodies on its surface bind to a specific foreign antigen.
  • This antigen/antibody complex is taken up by the B cell and processed by proteolysis into peptides.
  • the B cell displays these antigenic peptides on its surface MHC class II molecules.
  • This combination of MHC and antigen attracts a matching helper T cell, which releases lymphokines and activates the B cell.
  • These antibodies circulate in blood plasma and lymph, bind to pathogens expressing the antigen and mark (hem for destruction by complement activation or for uptake and destruction by phagocytes.
  • Antibodies can also neutralize challenges directly, by binding to bacterial toxins or by interfering with the receptors used by viruses and bacteria to infect cells.
  • NK cells or natural killer cells are defined as large granular lymphocytes that do not express T-eeil antigen receptors (TCR) or Pan T marker CDS or surface immunoglobulins (Ig) B cell receptor but that usually express the surface markers CD 16 (FcyRITT) and CD56 in humans, and NKl . l/ K1.2 in certain strains of mice.
  • TCR T-eeil antigen receptors
  • Ig surface immunoglobulins
  • Antigen-presenting cells which include macrophages, B lymphocytes, and dendritic ceils, are distinguished by their expression of a particular MHC molecule. APCs internalize antigen and re-express a part of that antigen, together with the MHC molecule on their outer cell membrane.
  • MHC The major histocompatibility complex
  • the MHC foci encode two major classes of MHC membrane molecules, referred to as class I and class II MHCs.
  • T helper lymphocytes generally recognize antigen associated with MHC class II molecules, and T cytotoxic lymphocytes recognize antigen associated with MHC class I molecules.
  • HLA complex In humans the MHC is referred to as the HLA complex and in mice the H-2 complex.
  • T-cell receptor is a molecule found on the surface of T lymphocytes (or T cells) that is generally responsible for recognizing antigens bound to major histocompatibility complex (MHC) molecules. It is a heterodimer consisting of an alpha and beta chain in 95% of T cells, while 5% of T cells have TCRs consisting of gamma and delta chains. Engagement of the TCR with antigen and MHC results in activation of its T lymphocyte through a series of biochemical events mediated by associated enzymes, co- receptors, and specialized accessory molecules.
  • MHC major histocompatibility complex
  • the CDS antigen (CD stands for cluster of differentiation) is a protein complex composed of four distinct chains (CDSv, CD35, and two times CDSe) in mammals, that associate with molecules known as the T-cell receptor (TCR) and the ⁇ -chain to generate an activation signal in T lymphocytes.
  • TCR T-cell receptor
  • the TCR, ⁇ -chain, and CDS molecules together comprise the TCR complex.
  • the CD3y, CD38, and CD3s chains are highly related cell surface proteins of the immunoglobulin superfamily containing a single extracellular immunoglobulin domain.
  • the transmembrane region of the CDS chains is negatively charged, a characteristic that allows these chains to associate with the positively charged TCR chains (TCRa and TCRfi).
  • the intracellular tails of the CDS molecules contain a single conserved motif known as an immunoreceptor tyrosine-based activation motif or IT AM for short, which is essential for the signaling capacity of the ' T ' CR.
  • CD28 is one of the molecules expressed on T cells that provide co- stimulatory signals, which are required for T cell activation.
  • CD28 is the receptor for B7.1 (CD80) and B7.2 (CD86). When activated by Toil-like receptor ligands, the B7.1 expression is upregulated in antigen presenting ceils (APCs). The B7.2 expression on antigen presenting cells is constitutive.
  • CD28 is the only B7 receptor co stitutively expressed on naive T cells. Stimulation through CD28 in addition to the TCR can provide a potent co-stimulatory signal to T ceils for the production of various interleukins (IL-2 and IL-6 in particular).
  • Malignant B cells appear to be an excellent targets for redirected T cells, as B cells can serve as immunostimuiatory antigen-presenting cells for T cells (Glimcher et al, 1982). Lymphoma, by virtue of its lymph node tropism, is anatomically ideally situated for T cell-mediated recognition and elimination. The localization of infused T cells to lymph node in large numbers has been documented in HIV patients receiving infusions of HIV-specific CDS " CTL clones. In these patients, evaluation of lymph node biopsy material revealed that infused clones constituted approximately 2%-8% of CDS' cells of lymph nodes.
  • Lymph node homing might be further improved by co-transfecting T cells with a cD A construct encoding the L-selection molecule under a constitutive promoter since this adhesion molecule directs circulating T cells back to lymph nodes and is down- regulated by in vitro expansion (Chao et al., 1997).
  • the present invention may provide a method of treating a human disease condition associated with a cell expressing endogenous CD 19 comprising infusing a patient with a therapeutically effective dose of the recombinant human CD19-specific CAR expressing cell as described above.
  • the human disease condition associated with a cell expressing endogenous CD 19 may be selected from the group consisting of lymphoma, leukemia, non-Hodgkiris lymphoma, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, chronic lymphocytic leukemia, and B cell-associated autoimmune diseases.
  • Leukemia is a cancer of the blood or bone marrow and is characterized by an abnormal proliferation (production by multiplication) of blood cells, usually white blood cells (leukocytes). t is part of the broad group of diseases called hematological neoplasms.
  • Leukemia is a broad term covering a spectrum of diseases. Leukemia is clinically and pathologically split into its acute and chronic forms.
  • Acute leukemia is characterized by the rapid proliferation of immature blood cells. This crowding makes the bone marrow unable to produce healthy blood cells. Acute forms of leukemia can occur in children and young adults. In fact, it is a more common cause of death for children in the U.S. than any other type of malignant disease. Immediate treatment is required in acute leukemia due to the rapid progression and accumulation of the malignant cells, which then spill over into the bloodstream and spread to other organs of the body. Central nervous system (CNS) involvement is uncommon, although the disease can occasionally cause cranial nerve palsies. Chronic leukemia is distinguished by the excessive build up of relatively mature, but still abnormal, blood cells.
  • Chronic leukemia mostly occurs in older people, but can theoretically occur in any age group. Whereas acute leukemia must be treated immediately, chronic forms are sometimes monitored for some time before treatment to ensure maximum effectiveness of therapy.
  • lymphocytic or lymphoblastic which indicate that the cancerous change took place in a type of marrow cell that normally goes on to form lymphocytes
  • myelogenous or myeloid which indicate that the cancerous change took place in a type of marrow ceil that normally goes on to form red cells, some types of white cells, and platelets (see lymphoid cells vs. myeloid cells).
  • Acute lymphocytic leukemia also known as acute lymphoblastic leukemia, or ALL
  • ALL acute lymphoblastic leukemia
  • Chronic lymphocytic leukemia most often affects adults over the age of 55. It sometimes occurs in younger adults, but it almost never affects children. Acute myelogenous leukemia (also known as acute myeloid leukemia, or AML) occurs more commonly in adults than in children. This type of leukemia was previously called “acute nonlymphocytic leukemia.” Chronic myelogenous leukemia (CML) occurs mainly in adults, A very small number of children also develop this disease. [00149] Lymphoma is a type of cancer that originates in lymphocytes (a type of white blood cell in the vertebrate immune system). There are many types of lymphoma. According to the U.S.
  • lymphomas account for about five percent of all cases of cancer in the United States, and Hodgkin's lymphoma in particular accounts for less than one percent of all cases of cancer in the United States. Because the lymphatic system is pari of the body's immune system, patients with a weakened immune system, such as from HIV infection or from certain drags or medication, also have a higher incidence of lymphoma.
  • lymphoma is broadly categorized as Hodgkin's lymphoma and non-Hodgkin lymphoma (all other types of lymphoma). Scientific classification of the types of lymphoma is more detailed. Although older classifications referred to histiocytic lymphomas, these are recognized in newer classifications as of B, T, or NK cell lineage.
  • Autoimmune disease or autoimmunity, is the failure of an organism to recognize its own constituent parts (down to the sub-molecular levels) as "self,” which results in an immune response against its own cells and tissues. Any disease that results from such an aberrant immune response is termed an autoimmune disease.
  • Prominent examples include Coeliac disease, diabetes mellitus type 1 (IDDM), systemic lupus erythematosus (SLE), Sjogren's syndrome, multiple sclerosis (MS), Hashimoto's thyroiditis, Graves' disease, idiopathic thrombocytopenic purpura, and rheumatoid arthritis (EA).
  • Inflammatory diseases including autoimmune diseases are also a class of diseases associated with B-cell disorders.
  • autoimmune diseases include, but are not limited to, acute idiopathic thrombocytopenic purpura, chronic idiopathic thrombocytopenic purpura, dermatomyositis, Sydenham's chorea, myasthenia gravis, systemic lupus erythematosus, lupus nephritis, rheumatic fever, polyglandular syndromes, bullous pemphigoid, diabetes mellitus, Henoch-Schonlein purpura, post- streptococcalnephritis, erythema nodosum, Takayasu's arteritis, Addison's disease, rheumatoid arthritis, multiple sclerosis, sarcoidosis, ulcerative colitis, erythema multiforme, IgA nephropathy, polyarte
  • compositions described herein may be comprised in a kit.
  • allogeneic CAR TCR e8 cells are provided in the kit, which also may include reagents suitable for expanding the cells, such as media.
  • a chimeric receptor expression construct In a non-limiting example, a chimeric receptor expression construct, one or more reagents to generate a chimeric receptor expression construct, cells for transfection of the expression constract, and/or one or more instruments to obtain allogeneic cells for transfection of the expression construct (such an instrument may be a syringe, pipette, forceps, and'Or any such medically approved apparatus).
  • an instrument may be a syringe, pipette, forceps, and'Or any such medically approved apparatus.
  • an expression constract for eliminating endogenous TCR ⁇ / ⁇ expression, one or more reagents to generate the construct, and/or CAR+ T cells are provided in the kit.
  • the kit comprises reagents or apparatuses for electroporation of cells. [00157] In some embodiments, the kit comprises artificial antigen presenting cells.
  • kits may comprise one or more suitably aliquoted compositions of the present invention or reagents to generate compositions of the invention.
  • the components of the kits may be packaged either in aqueous media or in lyophilized form.
  • the container means of the kits may include at least one vial, test tube, flask, bottle, syringe, or other container means, into which a component may he placed, and preferably, suitably aiiquoted. Where there is more than one component in the kit, the kit also will generally contain a second, third, or other additional container into which the additional components may be separately placed. However, various combinations of components may be comprised in a vial.
  • the kits of the present mvention also will typically include a means for containing the chimeric receptor construct and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow molded plastic containers into which the desired vials are retained, for example.
  • PBMC Peripheral blood mononuclear cells
  • ZFNs containing five or six fingers were assembled from an established archive of pre-validated 2-fmger and 1 -finger modules as described (Doyon et ⁇ ,, 2008; Isalan et al, 2001 ).
  • ZFN pairs were designed to bind either a sequence within exon 1 of the TCR a constant region (TRAC: NG 001332.2; ZFNs designated as TRAC-ZFN-1 and TRAC-ZFN-2) or a consensus sequence common to exon 1 of both TCR ⁇ constant regions 1 and 2 (TRBC1 and TRBC2: NG 001333.2; ZFNs designated as TRBC-ZFN-1 andTRBC- ZF -2).
  • poly-adenines were added using a poly A tailing kit (Ambion), the integrity and size of the mRNA species was validated on a denaturing 1 % agarose gel with 3-(N-morpholino) propanesulphonic acid (MOPS) buffer, and concentration was measured using a spectrophotometer (BioRad, Hercules, CA) at OD 2 6o.
  • the mRNA was stored at -80°C in nuclease-free vials for single use.
  • An Alexa 488-conjugated CAR-specific antibody (clone 136-20- 1) that recognizes an epitope within scFv region of CD19RCD28 was generated in the laboratory. TCR ⁇ usage was analyzed by a panel of anti-Vp monoclonal antibodies (IQTest® Beta Mark; Beckman Coulter, Brea, CA). The inventors added propidium iodide (Sigma-Aldrich, St. Louis, MO) just before collecting ceils on a flow-cytometer to exclude dead cells from analysis.
  • K562-derived aAPC were previously modified by lentiviral transduction to constttutively co-express CD 19, CD64, CD86, CD137L, membrane-bound (MB) IL- 15, and EGFP (the latter encoded following the emcv IRES element).
  • a clone (#4) was obtained by limiting dilution and numerically expanded for use (Manuri et at, 2010).
  • CD3-specific antibody (OKT3; eBioscience, San Diego, CA) was used to activate T cells by pulsing the mAb onto the CD64 " (FcR) clone #4 (FIG. 8). Expression of desired transgenes and bound OKT3 was validated weekly by How cytometry before use in co-culture with T cells.
  • T ceils derived from human PBMC isolated by density gradient separation using Ficoli-Paque Pius (GE Healthcare, Pittsburgh, PA), were numerically expanded i the presence of 50 lU/mL of recombinant human interieukin-2 ([rhIL-2] added three times a week; Chiron, Emeryville, CA) on ⁇ - irradiated (100 Gy) aAPC (clone #4, 1 :2 T cell : aAPC ratio) that had been pre-loaded with OKT3.
  • T cells with aAPC were cultured in complete medium (CM) defined as Hyclone-RPMI 1640 (Thermo Fisher Scientific, Waltham, MA) supplemented with 2 mnioi/L L-glutamine (Glutamax- l ; mvitrogen) and 10% heat-inactivated Hyclone-fetal bovine serum (Thermo Fisher Scientific).
  • CM complete medium
  • Hyclone-RPMI 1640 Thermo Fisher Scientific, Waltham, MA
  • 2 mnioi/L L-glutamine Glutamax- l ; mvitrogen
  • Hyclone-fetal bovine serum Thermo Fisher Scientific
  • TRBC1 forward 5'-CTGAACAAGGTGTTCCCACCC-3' (SEQ ID NO:
  • TRBC1 reverse 5'-GTGTGCGCTGGTTCCTTTCTT-3' (SEQ ID NO:
  • Target cells were labeled with 0.1 mCi of D l Cr (Perkm Elmer, Boston, MA) for 2 hours. After washing thrice with ice-cold CM, labeled ceils were diluted and plated at 10 cells/well in 100 uL CM in 96-weJl v-bottomed plates, T cells were added in 100 uLAvell at indicated effector target ratios and the plate was spun (i SOg for 3minutes without brake) to facilitate cell-to-cell contact. After a four hour incubation at 37°C, 5%C0 2 , 50 ⁇ , of supernatants were counted on TopCouiit (Perkin Elmer, Shelton, CT). All assays were performed in triplicate. The percent specific lysis was calculated as follows: ((experimental cpm - spontaneous cpm) / (maximum cpm - spontaneous cpm)) x 100.
  • T cells were incubated with 2.0 ⁇ of the red- fluorescent lipophilic dye PKH-26 (Cat # PKH26GL, Sigma-Aldrich) for five minutes at room temperature according to the manufacturer's instructions.
  • Cells, 100% labeled with PKH-26 were stimulated with either OKT3 loaded aAPC or CD 19 " aAPC in CM supplemented with 50 TU/niL rhIL-2 (added every-other-day).
  • PKH-26-derived fluorescence was measured by flow cytometry 10 days after stimulation and CD 19RCD28 r T cells were revealed using anti-CAR mAb clone 136-20-1.
  • TRAC-ZFN-2) or TCR ⁇ TCR ⁇ (TRBC-ZFN-1 and TRBC-ZFN-2) (FIG, 1) were developed and tested in primary human T cells propagated ex vivo for six days on OKT3-loaded aAPC (clone #4), Since transient expression of ZFNs is sufficient to mediate gene knockout, the inventors developed a "hit-and-run" delivery strategy to transiently express the ZFNs utilizing electro-transfer of in vitro transcribed mRNA species coding for the ZFN pairs (FIG. 2a). To measure TCR expression the inventors used a mAb specific for CD3E, which is only present on the ceil surface when TCRap is expressed.
  • TCR a or ⁇ loci To confirm that electroporated T cells had been genetically modified at the intended ZFN target sites (TCR a or ⁇ loci), a Surveyor Nuclease assay was performed using specific oligonucleotide primers flanking target sites within TRAC, TRBC I, or TRBC2. Cei-i nuclease digestion products, representative of genetic changes induced by the ZFNs, were present only after electro -transfer of ZFN pairs and the percent disruption assessed by densitometry correlated with loss of cell surface CD3s expression (FIG. 2c). These experiments in primary T cells confirmed that ZFNs designed to target TRAC or TRBC lead to permanent disruption of apTCR expression, as assessed by the Surveyor Nuclease assay and confirmed by flow cytometric analysis of CD3s.
  • TRBCl and TRBC2 disruption at the DNA level were approximately 20%-25% and that of TRAC disruption was approximately 60%. These numbers fit with the observed frequencies of CD3s ne8 CDl9RCD28 1 T cells because in each cell only one oui of four TRBC alleles (two TRBC l and two TRBC2) is expressed. Similarly one of two TRAC alleles is expressed in each T cell. Therefore, disruption of the expressed allele is sufficient to achieve the CD3-negative phenorype.
  • TCR ⁇ CAR T cells could not respond to TCR stimulation. To test this, they measured the proliferative response of these cells after stimulation by cross-linking CD3 with OKT3 in comparison to activating CAR for sustained proliferation upon docking with CD 19. TCR "eg CD19RCD28 f T cells proliferated in response to CD 19, but not OKT3 (FIG. 5a). Next, the inventors assessed the ability of TCR neg CD19RCD28 + T cells to lyse 0 19 " target cells in a standard 4-hour 5l Cr release assay (FIG. 5b).
  • TCR ueg CAR 1" T cells to specifically lyse CD 19 target cells was similar to that observed for TCR ' CD 19RCD28 f T cells. Together, these data confirmed ihat ihe absence of a measurable TCR on TCR eg CD19RCD28 " T ceils corresponds with abrogation of TCR. activity, but does not impact the CAR to activate genetically modified T cells for proliferation and killing.
  • Embodiments of the invention show that T cells and indeed CAR " T cells can be genetically edited by ZFNs to eliminate expression of the endogenous ⁇ TCR. This has therapeutic implications where donor-derived T cells are infused to achieve an anti- rumor effect. Therapeutic success after allogeneic HSCT is defined as achieving a GVL- effect without causing clinically-significant GVHD (Bleakley et al, 2004). Thus, separation of GVL and GVHD is the crucial issue following engraftment of allogeneic hematopoietic stem ceils and strategies to accomplish this are based on infusing desired T-cell effector populations predicted to reduce unwanted allogeneic effects.
  • CAR + T cells expressing alloreactive TCRs can be rendered anergic to disparate HLA while maintaining specificity for CD 19 (Davies et al, 2010) by blockade of co- stimulatory molecules upon co-culture of genetically modified T cells with stimulator cells expressing disparate HLA.
  • An alternative to pre-selection includes conditional ablation of infused allogeneic CAR 1 T cells in the event that serious adverse events occur. This has been accomplished by genetic modification of allogeneic T cells to express "suicide genes" such as HSVtk (Bonini et al, 1997), iCasp9 (Straathof et ai., 2005), CD2Q (mtrona et al , 2000), TMPK (Sato et al, 2007), and a modified Fas (Berger et al, 2004) that can be triggered for conditional ablation via the administration of specific molecules ⁇ e.g., ganciclovir to HSVtk expressing ceils).
  • suicide genes such as HSVtk (Bonini et al, 1997), iCasp9 (Straathof et ai., 2005), CD2Q (mtrona et al , 2000), TMPK (Sato et al
  • ZFNs target and thus disrupt gene expression at the genomic level, which is an advantage over techniques that involve transcriptional repression and require sustained expression of the inhibiting factor (e.g., enforced expression of shRNA to mediate TCR down regulation (Okamoto et al, 2009)).
  • Thai ZFNs can permanently disrupt gene expression after transient expression (without the inherent dangers of genomic integration) enabled the use of in vitro transcribed mRNA species in a "hit-and-run" manner for electro-transfer of ZFNs into T cells.
  • Previous reports suggest that T-celi activation mediated through an endogenous TCR is required to obtain a fully fimctional CAR in a model sysiem using Jurkat cell lines (Bridgeman et at, 2.010).
  • CD19RCD28 '1' T cells Additional inclusion of a CD 137 domain, also demonstrated to enhance the function and survival of CD19RCD28 '1' T cells (Carpenito et al., 2009; Wang et al, 2007), may be tested (Milone et al., 2009).
  • a benefit to expressing TCR with known specificity is that activation through the endogenous immune-receptor can be used to propagate T cells to achieve an anti-tumor effect mediated by the CAR (Cooper et al., 2005; Pule et al, 2008). It remains to be tested in humans whether coordinated co- stimulation achieved through multiple CAR signaling endodomains will be sufficient to sustain persistence in vivo or if triggering of T ceils through TCR is needed.
  • s can 3 ⁇ 4 6 generated using a genetic approach to remove (a) endogenous undesired TCR with ZFNs and (b) introduce a desired CAR with the SB system, for example.
  • the genetic approaches to introduce mRNA and DNA use a common electro-transfer platform approved for clinical trials.
  • the inventive approach abolishes the danger of GVHD posed by adoptive transfer of large numbers of allogeneic T cells while maintaining desired efiecior functions mediated by CD19RCD28 CAR io target malignant B cells.
  • This strategy provides an important step to developing a "universal" CAR " T cell that can be manufactured from one donor and administered on demand to multiple patients.
  • T cells therapy by infusion of T cells can reconstitute immunity to combat pathogens and malignancies.
  • the time required to manufacture T cells with ihe desired properties and in sufficient numbers ex vivo is often incompatible with ihe treatment window for patients.
  • autologous T cells from patients with advanced disease may have compromised function and be tolerant to desired antigens.
  • ZFNs designer zinc-finger nucleases
  • ZFNs comprise a zinc finger DNA-binding domain designed to bind a specific DNA sequence fused to the cleavage domain of Fokl endonuclease. Since Fokl dimerization is required to introduce a double strand break (DSB), the inventors generated ZFN pairs that flank the intended DNA target sequences in the required spatial conformation. Cellular repair of the DSB by error-prone nonhomologous end joining allows disruption of HLA gene expression. Transfection of ZFN pairs designed to target exon 3 of the HLA-A locus into the human kidney cell line HEK293 resulted in 10% genetic modification of the HLA-A loci.
  • DSB double strand break
  • the inventors generated clones of ⁇ 293 cells that showed deletion or insertion mutations within the ZFN binding site of one or both HLA-A alleles leading to early termination of translation. These HLA-A nuI1 HEK293 clones evaded HLA-A-restricted lysis by T cell clones, even after interferon- ⁇ and TNF-ct treatment was used to upreguiate HLA expression. Because only transient expression of ZF s is needed to disrupt a target gene, the inventors tested the ability to disrupt HLA-A gene expression by electro-transfer of in vr ' /ro-transcribed ZFN niRNA into primary T cells.
  • a single administration of the niRNA encoding the ZFNs targeting HLA-A could render over 40% of primary T cells HLA-A negative.
  • the inventors enriched the HLA-A 11" " population by paramagnetic bead separation to obtain a pool of T cells >90% of which lack HLA-A expression.
  • a useful clinical application for HLATM' 1 allogeneic T ceils is to redirect their specificity independeni of HLA via expression of a chimeric antigen receptor (CAR) targeting CD 19.
  • CAR chimeric antigen receptor
  • the inventors used ZFN pairs targeting the TCR a or the TCR ⁇ locus. Transient expression of these ZFNs resulted in permanent disruption of endogenous TCR expression and a highly enriched ⁇ TCRTM 11 cell population could be generated by paramagnetic bead selection. Thus, it is useful to provide allogeneic T cells as "off-the-shelf biologies that can be infused on demand as "drugs.”
  • CD 19 is a lineage-specific cell-surface antigen expressed on B-cell malignancies.
  • T cells can be manufactured to target CD 19 independent of HLA molecules by electro-transfer of CD19-specific chimeric antigen receptor (CD19RCD28CAR). This can be useful, given that T cells from patients can be functionally flawed and/or the time to manufacture a T-cell dose may not match the time windo for infusion, given the pace of rumor progression, [00202]
  • the inventors generate CD 19RCD28CAR+ T cells from third party healthy donors that can avoid rejection. In doing so, there is avoidance of unwanted immune reactions mediated by the interaction between HLA and TCR.
  • ZFNs zinc finger nucleases
  • FIG. 1 la A pair of ZFN can induce DNA double strand break (DSB) at a target nucleotide sequence in the genome. Error-prone non-homologous end joining repair of ZFN-mediated DNA DSB leads to complete elimination of target gene expression (FIG. l ib).
  • DSB DNA double strand break
  • HEK 293 cells (karyotyping analysis showed thai this ceil line has four copies of chromosome 6, wherein the HLA-A gene is located) were transduced with a pair of ZFN and HLA-A disruption was evaluated by Surveyor Nuclease (CEL-I) assay.
  • CEL-I Surveyor Nuclease
  • HLA-A-targeted ZFNs disrupt the HLA-A locus in up to 10% of HEK293 cells (FIG. 12a).
  • the inventors isolated ZFN-transduced HEK293 clones, and they could not detect HLA-A expression in these clones even after IFNy and TNF treatment (FIG. 12b). These HEK293 clones evade HLA-A restricted CTL clone attack, when they were pulsed with high dose cognate peptide (FIG, 12c),
  • HLA-A-targeted ZFNs can also disrupt HLA-A from T cells over 40% when they were expressed from in vitro transcribed mRNA (FIG. 13a).
  • HLA-A nu11 T-cell population can be enriched by simple paramagnetic beads-based sorting (FIG. 13b).
  • HLA-A-targeted ZFNs also disrupt HLA-A expression from
  • CD19RCD28CAR + T cells (FIG. 14A). HLA-A nali CD 19RCD28CAR + T cells evade HLA-A restricted CTL attack (FIG. 14b) but maintain CD 19 specificity (FIG. 14c). [08207] TCR expression can also be disrupted from CD 19RCD28CAR : T cells by TCR a or ⁇ constant region-targeted ZFN pairs (FIG. 15a). TCR nu!! CD 19RCD28CAR ⁇ T cells maintain CD 19 specificity (FIG. 6b) and also can be propagated on CD 19 -expressing K562-based artificial antigen presenting cells (FIG. 15c). [ ⁇ 8208] TCR and HLA-A expression can be simultaneously disrupted in
  • CD19RCD28CAR T cells by treating the cells with both TCR a or ⁇ constant region- targeted ZFN pairs and HLA-A-targeted ZFN pairs (FIG. 16),
  • Barker et ai Successful treatment of EBV-associated posttransplantation lymphoma after cord blood transplantation using third-party EBV-specific cytotoxic T lymphocytes, Blood, 1 6:5045-5049, 2010.

Abstract

The present invention concerns methods and compositions for immunotherapy employing a modified T cell comprising disrupted T cell receptor and/or HLA and comprising a chimeric antigen receptor. In certain embodiments, the compositions are employed allogeneically as universal reagents for "off-the-shelf treatment of medical conditions such as cancer, autoimmunity, and infection. In particular embodiments, the T cell receptor-negative and/or HLA-negaiive T cells are generated using zinc finger nucleases, for example.

Description

DESCRIPTION
CAR+ T CELLS GENETICALLY MODIFIED TO ELIMINATE EXPRESSION OF T- CELL RECEPTOR AND/OR HLA
[0001] The present application claims the priority benefit of United States provisional application number 61/561 ,364, filed November 18, 201 1, the entire contents of which are incorporated herein by reference. [0002] This invention was made with government support under Grant No.
PR064229, awarded by the Department of Defense, and Grant Nos. CA 124782, CA120956, CA141303, and CA1 16127, awarded by the National Institutes of Health. The government has certain rights to the invention.
BACKGROUND OF THE INVENTION 1. Field of the Invention
[08(53] The field of the invention includes at least medicine, immunology, ceil biology, and molecular biology. In certain aspects the fseld of the invention includes immunotherapy .
2. Description of Related Art
[0004] Allogeneic hematopoietic stem-cell transplantation (HSCT) can cure some patients with high risk B-eeii leukemia/lymphoma, but relapse remains a major cause of death. To improve the graft-versus-ieukemia/lymphoma (GVL)-effect, donor-derived T cells can be genetically modified to express a tumor-specific chimeric antigen receptor (CAR) with specificity derived, for example, from the variable domains of a monoclonal antibody, thus focusing immimoreactivity towards the tumor in an MHC non-restricted manner (Cooper el al, 2004). However, the endogenous αβ T-cell receptor (TCR) on infused allogeneic T cells may recognize major and minor histocompatibility antigens in the recipient, leading to graft- versus-host-disease (GVHD). As a result, the majority of current clinical trials infuse autologous CAR' T cells relying on immune tolerance to prevent TCR-mediated deleterious recognition of normal tissues after adoptive transfer (Jena et al, 2010). This approach has achieved early clinical successes (Kochenderfer et al, 2010), but is limited by the time and expense to manufacture patient-specific T-cell products. [Θ8Θ5] CD 19 is constitutive ly expressed on most acute and chronic B-ceil malignancies. Therefore, to target malignant B cells, the Sleeping Beauty (SB) transposon/transposase system was adapted for human application, e.g., to stably express a CD19-specific CAR (designated CD19RCD28) (Kohn et al, 201 1 ; Tzsvak et al, 2010; Hackett et al, 2010; Williams, 2008; see U.S. Patent No. 6,489,458, incorporated by reference herein in its entirety). SB modified CAR1" T cells can be numerically expanded to clinically-sufficient numbers by the recursive addition of γ-irradiated designer artificial antigen presenting cells (aAPC) that co-express CD 19 and desired T-cell co-stimulatory molecules (Singh et al, 2008; Davies et al, 2010). This has been adapted for clinical translation at M.D. Anderson Cancer Center, as four clinical trials based on the electroporation and propagation of CAR+ T cells have achieved institutional and federal regulatory approvals for the adoptive transfer of CD19RCD281 T cells after autologous and allogeneic hematopoietic stem-cell transplantation (e.g., IND #14193, ClinicalTrials.gov Identifier: NCT00968760) (Jena et al, 2010; Kohn et al, 201 1 : Hackett et al, 2010; Williams, 2008). However, a need in the art remains to develop methods and reagents that circumvent the time and expense to manufacture patient-specific 'T-cell products.
SUMMARY OF THE INVENTION
The present invention is directed to methods and/or compositions for use in medicine, for example, for use in immunotherapy, including immunotherapy for infection or cancer, for example. In some embodiments, there is provided universal T cell-based immunotherapy including T cells engineered to express an antigen-specific chimeric antigen receptor (CAR) and to eliminate expression of endogenous alpha/beta T-cell receptor (TCR). In specific embodiments, there are CAR-'- T cells genetically modified to eliminate expression of TCR. [Θ807] In some embodiments, there is disruption of the T-cell receptor α/β in CAR- expressing T cells using zinc finger nucleases (ZFNs) for generating universal T cells for immunotherapy. In embodiments of the invention, there is knocking out of the T-cell receptor αβ-chain in CAR-expressing T cells, for example using zinc finger nucleases.
[©008] In some embodiments, there are universal CAR-expressing T cells, such as from a healthy donor, that may be suitably stored, for example, in the freezer, and then infused into allogeneic individuals on demand. This allows interested parties to prepare and validate genetically modified antigen-specific T cells before individuals need them. These T cell products significantly broaden the application of antigen- specific T-eeil therapy, such as for cancer and infection, for example. In certain embodiments, the invention provides off- the-shelf universal CAR" T cells from allogeneic healthy donors that can be administered to any patient without causing GVHD. An advantage to the methods and compositions of the present invention is that ihe modified cells allow interrogation of ihe antigen independence of MHC and therefore the cells are suitable for any genetic background in the recipient. However, in some embodiments, the CARhTCRuSg cells are autologous.
[000.9] Any kind of autoimmune disease, infection, and cancer can be treated with methods and/or compositions of the invention, including primary, metastatic, recurrent, sensitive-to-therapy, refractor -to -therapy, and so forth. In some embodiments, the methods and/or compositions are employed for chemo-refractory cancer. The cancer may be of the blood, lung, brain, colon, prostate, breast, liver, kidney, stomach, cervix, ovary, testes, pituitary gland, esophagus, spleen, skin, bone, and so forth. [0010] The skilled artisan recognizes that in some methods there is simple depletion of T ceils that continue to express TCR by using, for example, clinical-grade CDS -specific monoclonal antibody such that a T-cell product can be generated in which at least the majority have lost expression of endogenous TCR. Other ways include the use of fluorescence-activated ceil sorting (FACS) and TCR- specific monoclonal antibodies or aptamers to reduce and possibly eliminate the presence of T cells that continue to express TCR. In addition, TCR-specific antibodies can be combined with other purification schemes, such as addition of complement, toxins, or resetting to reduce/eliminate T cells that continue to express TCR.
[0011] In certain embodiments there are methods of treating an individual in need of treatment using compositions encompassed by the invention. The treatment includes employing particular T cells having a CAR and also having genetic modifications to exclude functional TCR. Although in specific embodiments the modifications of the cells exclude functional TCR by any suitable means, in certain aspects the modifications include knock out (or in alternative embodiments, knock down, such as by siRNA and TALENs, for example) of the a and/or β chains or the gamma and/or delta chains of the TCR. [0012] In some embodiments, there is a method of making a cell expressing a CAR comprising introducing an expression cassette in to the ceil, wherein the expression cassette encodes a polypeptide comprising a heterologous human extracellular antigen binding domain, a transmembrane domain, and one or more an intracellular signaling domain(s). In some embodiments, the methods further comprise stimulating the cells with antigen presenting cells, recombinant antigen, or an antibody to the receptor to cause the cells to proliferate, kill, and/or make cytokines. In specific embodiments, the method further comprises stimulating the cells with antigen presenting cells to cause the cells to proliferate.
[0013] In some embodiments, there are recombinant antigen-specific TCR eg cells expressing and bearing on the cell surface membrane an antigen-specific CAR human polypeptide comprising an intracellular signaling domain, a transmembrane domain and an extracellular domain, the extracellular domain comprising a human anti-antigen monoclonal antibody or antigen binding fragment thereof.
[1)814] In some embodiments, there is a method of treating a human disease condition associated with a cell expressing endogenous CD 19 comprising infusing a patient with an amount of a recombinant TCR'*8 ceil expressing a human antigen-specific CAR (such as CD19-specific) sufficient to treat the condition, wherein the human antigen-specific CAR comprises a heterologous human CD 19 extracellular binding domain, a transmembrane domain, and an intracellular signaling domain. [Θ815] In some embodiments more than one pair of zinc finger nucleases can be used to modify a cell. For example, both zinc finger nucleases targeting the alpha chain and the beta chain can be used to eliminate T-cell receptor expression. In another instance, the zinc finger nucleases targeting the T-cell receptor can be used (for example, sequentially) with zinc finger nucleases to target one or more human leukocyte antigen(s) (HLA). This is desirable to generate universal T ceils that have lost not only T-cell receptor expression, but also HLA expression, and thus will be less susceptible to immune-mediated recognition from the allogeneic recipient and thus targeted for destruction.
[081 ] In one embodiment there is an isolated T-cell population wherein cells of the population comprise an endogenous T-cell receptor coding sequence that is either not expressed or which encodes a nonfunctional T-cell receptor; and a recombinant chimeric antigen receptor comprising an intracellular signaling domain, a transmembrane domain, and an extracellular domain comprising an antigen binding region. In specific embodiments, the T-ceil receptor is nonfunctional by virtue of one or more disruptions in the coding sequence of a chain, β chain, or both. In some embodiments, the endogenous T-celi receptor is knocked out. In specific embodiments, the antigen binding region is an F(ab')2, Fab', Fab, Fv, or scFv and/or the antigen binding region binds a tumor associated antigen, such as CD 19, CD20, ROR] , carcinoembryonic antigen, alphafetoprotein, CA- 125, MUC- 1 , epithelial tumor antigen, melanoma-associated antigen, mutated p53, mutated ras, HER2/Neu, ERBB2, FTER3, folate binding protein, FlTV- l envelope glycoprotein gpl20, HIV- 1 envelope glycoprotein gp41 , GD2, CD 123, CD23.CD30 , CD56, c-Mef mesothelin, GD3, HERV-K, IL- 1 IRalpha, IL- 13Ralpha2, kappa chain, or lambda chain, CSPG4 (also known as, high molecular weight melanoma associated antigen), EGFRvIII, and VEGFR2. In specific embodiments, the antigen binding region binds a pathogen antigen, such as a fungal, viral, or bacterial antigen. In some cases, the fungal antigen is from Aspergillus or Candida. In certain cases, the viral antigen is from HSV, RSV, EBV, CMV, JC virus, BK virus, or Ebola.
[00171 In embodiments of the invention, there is an intracellular signaling domain that is a T-lyrnphocyte activation domain. In some embodiments, the intracellular signaling domain comprises CD3 , CD28, OX40/CD134, 4- 1BB/CD137, FceRIy, ICOS/CD278, ILRB/CD 122, IL--2RG/CD 132, DAP molecules, CD70, cytokine receptor, CD40, or a combination thereof.
[00181 In certain embodiments, the transmembrane domain comprises IgG4Fc hinge, Fc regions, CD4 transmembrane domain, CD28 transmembrane domain, the CD3 transmembrane domain, cysteine mutated hitman €Β3ζ domain, CD 16 transmembrane domain, CDS transmembrane domain, or erythropoietin receptor transmembrane domain. 0019J In some embodiments, there is an exemplary chimeric antigen receptor that comprises: a) an antigen binding domain from a variable region of an antigen-specific monoclonal antibody; b) a signaling domain of 033ζ; and c) CD28, CD 137, CD 134, or combinations thereof.
[Θ820] In some cases, there are methods of generating the ceils of the invention, comprising the steps of: a) providing one or more T cells; b) modifying the T cell(s) to express a recombinant chimeric antigen receptor comprising an intracellular signaling domain, a transmembrane domain, and an extracellular domain comprising an antigen binding region; and c) modifying the T cell(s) to harbor an endogenous T-eell receptor coding sequence that is not expressed or encodes a nonfunctional T-cell receptor. In specific embodiments, step b) occurs before step c) or step c) occurs before step b). in certain aspects, the T cell is provided from an umbilical cord blood bank, is provided from a peripheral blood bank, is an induced pluripotent stem cell, or is a human embryonic stem ceil. In some cases, the T cell is allogeneic in reference to one or more intended recipients.
[0021J In some embodiments, the chimeric antigen receptor is stably introduced into the cell, and in certain embodiments, the polynucleotide that encodes the chimeric antigen receptor is introduced into the cell by a transposon/transposase system or a viral-based gene transfer system, such as by recombinant retrovirus or lentivirus.
[Θ022] In some embodiments, following modification of the T cell(s), they are propagated by exposing the T cells to artificial antigen presenting cells, by using OK'T'3 (or equivalent to cross-fink CDS) optionally with other co-stimulatory antibodies (e.g., anti- CD2S) on beads, or by using OKT3 (or equivalent to cross-link CD3) optionally with other co-stimulatory antibodies (e.g., anti-CD28) mixed with peripheral blood mononuclear cells. In specific aspects, a polynucleotide that encodes the recombinant chimeric antigen receptor is electroporated into the T cell, in some embodiments, a polynucleotide that encodes the chimeric antigen receptor is present on a plasmid or viral vector. [Θ823] In some embodiments, the T cell can be genetically modified with zinc finger nuclease or TLAE nuclease to eliminate HLA expression. The T cell may express a CAR and/or have been modified to eliminate TCR expression.
[0024J In some embodiments, the endogenous T-cell receptor and/or HLA is disrupted by nonhomologous end joining repair, such as is generated by zinc finger nuclease, TALE nuclease, introduced into the cell by physical means, electro-transfer of mRNA species, viral vector, or non-viral vector,
[Θ825] In some embodiments, there are methods of treating an individual with a medical condition (such as autoimmune disease, cancer, or infection, including Aspergillus or Candida), comprising the step of providing an effective amount of cells from the population of cells described herein, including more than once in some aspects, such as at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, or more days apart. In specific embodiments, the cancer is lymphoma, leukemia, non-Hodgkin's lymphoma, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, chronic lymphocytic leukemia, or B cell-associated autoimmune diseases.
[0026] The foregoing has outlined rather broadly the features and technical advantages of the present invention in order that the detailed description of the invention that follows may be better understood. Additional features and advantages of the invention will be described hereinafter, which form the subject of the claims of the invention. It should be appreciated by those skilled in the art that the conception and specific embodiment disclosed may be readily utilized as a basis for modifying or designing other structures for carrying out the same purposes of the present invention. It shoidd also be realized by those skilled in the art that such equivalent constructions do not depart from the spirit and scope of the invention as set forth in the appended claims. The novel features which are believed to be characteristic of the invention, both as to its organization and method of operation, together with further objects and advantages will be better understood from the following description when considered in connection with the accompanying figures. It is to be expressly understood, however, that each of the figures is provided for the puipose of illustration and description only and is not intended as a definition of the limits of the present invention.
BRIEF DESCRIPTION OF THE DRAWINGS
[0827] For a more complete understanding of the present invention, reference is now made to the following descriptions taken in conjunction with the accompanying drawing, in which:
[0028] FIG, 1. ZFN pairs targeting sites within genomic iocs of TCR- and β constant region. Each exon is shown by a block. Black blocks represent coding regions. Grey columns represent non-coding regions. One ZFN pair was designed to bind exon 1 of the TCR a constant region (IRAQ and another ZFN pair binds a conserved sequence on exon 1 of the TCR β constant regions 1 (TRBC1) and 2 (TRBC2). Underlined nucleotide sequences represent the intended binding sequence of each ZFN.
[0029] FIG, 2. Disruption of the TCR αβ-CDS complex in primary T ceils.
[0030] a. Schematic presentation of ZFN transfer. [0031] A pair of ZFN-encoding mRNA was electro-transferred into T cells six days after stimulation of CARneg T cells, T cells were then cultured with 50 lU/mL of IL-2 and some cells were incubated at 30°C-5% C02. CD3s expression was analyzed on days 7-9 after electroporation by flow cytometry. [0832] b. Down regulation of CDS after electro-transfer of ZFN targeting TCR αβ from mRNA.
[0833] Day 9 after electro-transfer of graded doses of mRNA coding for pairs of ZFN, TCR a. -CD3 expression was analyzed by co-staining for CD4, CDS, and CD3s. The representative flow data at day nine after ZFN electro-transfer is shown. Flow cytometry data are gated on cells excluding propidium iodide. Numbers in the lower right quadrant represent the percentage of CD3s negative cells in T-ceil populations.
[0834] Top panels shows CD3s expression in T cells cultured at 37°C after ZFN transfer and bottom panels shows CD3e expression in T cells transiently cultured at 30°C from days 2-3 after ZFN transfer. [8835] c. Surveyor Nuclease assay to detect ZFN-mediated modification of TCR target sites in T-cells.
[00361 Arrows indicate the Cel-l digested products after electro-transfer of graded doses of mRNA species. 'The numbers below the lanes represent percentages of target disruption in each sample. [0837] FIG. 3. TCR"eg T ceils can be enriched by depletion of CD3s T cells.
[0838] «, CDS expression before and after depletion.
[0039] Representative results using in vitro expanded T cells. Flow cytometry revealing expression of CD3s in CD4f and CDS " T cells. Numbers in the lower right quadrant represent the percentage of CD3e-negative cells in T-cell populations. [0040] b. Υβ repertoire analysis in T cells modified with ZFN.
[0041] The νβ usage clonogram was analyzed by a panel of TCR-specific mAbs, co- stained with CD4 and CDS. Percentage of specific Vp ' T-cell fractions within CD4 and CDS gating is shown. The nomenclatures of Υβ repertoire shown are based on Arden et at (1995).
[8842] FIG, 4. EUmination of TCR fi-CBS complex from CD19-specific CAR'1 T cells. [ΘΘ43] . Schematic of electro-transfer of tnRNA coding for ZFN pairs in CAR ' T cells.
[0844] mRNAs coding for ZFN pairs were electro-transferred into CAR1 T cells, A pair of ZFN was electro-transfer to T cells two days after stimulation with CD19 h aAPC. After eiectroporation, ceils were maintained wiih 50 IU/inL of IL-2 and incubated for two days at 30°C-5% C02. CD3s expression was analyzed nine days after eiectroporation by- flow cytometry.
[00451 b. Disruption of TCR αβ-CDS complex expression after electro-transfer of mRNA coding for ZFN.
[00461 Flow cytometry analysis of CD3s expression in T cells nine days after electro- transfer of mRNA species coding for ZFN is gated on propidium iodide negative.
[0047] c. Surveyor Nuclease assay.
[0848] Arrows indicate the Cei- 1 digested products analyzed nine days after eiectroporation. The numbers at the bottom represent percentages of target disruption.
[0049] FIG. 5. Functional consequences of TCR knockout in CA *' T cells. [0050] a. Loss of responsiveness of'TCR"eg CAR1' T cells to TCR stimulation.
[0051] Dilution of PKH26 was measured 10 days after stimulation with aAPC loaded with OKT3 (upper panel) or expressing CD 19 (lower panel). Flow cytometry data was gated on CAR"1" T cells. Parental: CAR+ T cells without modification; no mRNA: mock electroporated CAR+ T cells; TRAC CD3nsg: CAR1 T cells electroporated with mRNA coding for ZFN pairs specific for TRAC, and depleted CD3pos population; TRBC CD3"eg: CAR" T ceils electroporated with mRNA coding for ZFN pairs specific for TRBC, and depleted for (T) ""' population. [Θ852] b. Redirected specificity o/'TCR"eg CAR " T cells.
[0053] Specific lysis by CAR" T cells of an EL4 mouse T-ce!l line modified to express a truncated version of human CD 19 (closed symbols) was measured by standard four hour J ICr release assay. Specificity is shown by lack of lysis of CD19 eg (parental) EL4 cells (open symbols). CAR" T cells were modified by ZFNs (TRAC and TRBC) or not modified by ZFNs (parental and no mRNA).
[0054] FIG. 6. Sustained proliferation of TCRneg CAR÷ T cells.
[0055] CAR+ T cells with (TRAC and TRBC) or without (parental and no mRNA) TCR modification by ZFNs were stimulated with γ-irradiated CD19 aAPC every two weeks. Viable T cells were enumerated every seven days and inferred total numbers were calculated.
[0856] FIG. 7. Schematic of the approach to genetically modify and propagate T cells from. PBMC. (a)T cells were propagated by stimulation with OKT3 loaded γ- irradiated aAPC (clone #4) in the presence of soluble IL-2. (b) DNA plasmids coding for SB transposon (CD19RCD28) and SB iransposase (SB l l) were electro-transferred into primary human T cells. CAR" T cells were selectively propagated by repeated additions of γ- irradiated aAPC (clone #4) in the presence of rhIL-2.
[0057] FIG. 8. Characterization of aAPC clone #4. (a) Flow cytometry was used to compare expression of CD 19, CD64, CD86, CD137L, and MBIL-15 between parental K562 (grey shaded histogram) and K562-aAPC clone #4 (black open histogram). MBIL-15 is composed of human IL- 15 peptide fused to modified human IgG4 Fc region and CD4 transmembrane domain and was detected by presence of EGFP (co-expressed with MBIL-15 after IRES element). aAPC were used to co-culture T cells if expression of introduced iransgenes were 80%. (b) OKT3 was loaded onto aAPC clone #4 at 1 ^tg per 10h cells. Flow cytometry data before and after OKT3 loading detected by antibody specific for mouse Fab region. No-staining control: grey shaded histogram; stain with anti-mouse Fab: black open histogram.
[00S8] FIG. 9. Depletion of a CDSs-positive population. Representative results from multiple experiments using in vitro expanded T cells. Flow cytometry revealing expression of CD3s in CD4 and CD8 T cells. Numbers in the lower right quadrant represent the percentage of CD3s-negative ceils in CD41" and CD81" T-cell populations. [0059] FIG. 10. An exemplary illustration of interactions between donor and recipient cells in normal eases and in embodiments of the invention.
[Θ860] FIGS. 11a and lib. Illustrations of use of particular zinc finger nucleases (ZF s) to achieve elimination of HLA in the genome. [0061] FIGS. 12a, 12b, and 12c. HLA- A targeted ZFNs are able to disrupt the HLA-
A locus in HEK293 cells (FIG. 12a) to an undetectable level (FIG. 12b) and that the HEK293 cells can evade HLA-A restricted CTL clone attack. ΘΘ62] FIGS. 13a and 13b. HLA-A targeting of ZFNs disrupting HLA-A. when they were expressed from in vitro transcribed mRNA (FIG. 13a). The HLA-A""1' T-cell population can be enriched (FIG. 13b).
[8863] FIGS. 14a, 14b, and 14c. Disruption of HLA-A from exemplary CD19RCD28CA + T cells (FIG. 14a) including evasion of HLA-A restricted CTL attack (FIG. 14b) but maintenance of CD 19 specificity (FIG. 14c).
[0064] FIG. 15a. TCR. expression can be disrupted from CD19RCD28CAR+ T cells by TCR a or β constant region-targeted ZFN pairs. FIGS. 15b and 15c. TCRnuilCD19RCD28CARi T cells maintain CD 19 specificity (FIG. 15b) and they can be propagated on CD 19-expressing K562-based artificial antigen presenting cells (FIG. 15c).
[0065] FIG. 16. Simultaneous knock out of HLA-A and TCR β. Peripheral blood mononuclear cells from HLA-A2 -positive healthy donor were stimulated with OKT3 loaded artificial antigen presenting cells and cultured with 50 lU/mL of rh IL-2. Six days after stimulation, expanded T cells were electroporated with HLA-A- targeted or TCR β constant region (TRBC)-targeted ZFNs from mRNAs. Both TCR expression and HLA-A expression were evaluated by anti-CD38 monoclonal antibody (mAb) and anti-HLA-A.2 mAb four days after electroporation. PI negative cells were positively gated according to CD4 and/or CDS. Numbers in the figure represent percentage of population in each quadrant.
DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
[8866] As used herein the specification, "a" or "an" may mean one or more. As used herein in the cJaim(s), when used in conjunction with the word "comprising," the words "a" or "an" may mean one or more than one. As used herein "another" may mean at least a
I I second or more. In specific embodiments, aspects of the invention may "consist essentially of or "consist of" one or more elements or steps of the invention, for example. Some embodiments of the invention may consist of or consist essentially of one or more elements, method steps, and/or methods of the invention. It is contemplated that any method or composition described herein can be implemented with respect to any other method or composition described herein,
I. Definitions
[0867] The term "chimeric antigen receptors (CARs)" as used herein may be referred to as artificial T-ceil receptors, chimeric T-cell receptors, or chimeric immune-receptors, for example, and encompass engineered receptors that graft an artificial specificity onto a particular immune effector cell. The CARs may be employed to impart the specificity of a monoclonal antibody onto a T cell, thereby allowing a large number of specific T cells to be generated, for example, in use for adoptive cell therapy. In specific embodiments, the CARs direct specificity of the ceil to a tumor associated antigen, for example. In some embodiments, the CARs comprise an intracellular activation domain, a transmembrane domain and an extracellular domain comprising a tumor associated antigen binding region. In particular aspects, CARs comprise fusions of single-chain variable fragments (scFv) derived fro monoclonal antibodies, fused to CD3-zeta transmembrane and endodomain. The specificity of other CARs designs may be derived from ligands of receptors (e.g., peptides) or from Dectins. In particular embodiments, one can target malignant B cells by redirecting the specificity of T cells using a chimeric immunoreeeptor specific for the B- lineage molecule, CD 19. In certain cases, the CARs comprise domains for additional co- stimulatory signaling, such as CD3-zeta, FcR, CD27, CD28, CD137, DAP 10, and/or OX40. In some cases molecules can be co-expressed with the CAR. These include co-stimulatory molecules, reporter genes for imaging (e.g., for positron emission tomography), gene products that conditionally ablate the T cells upon addition of a pro-drug, homing receptors, cytokines, and cytokine receptors.
[8868] The tenn "T-cell receptor (TCR)" as used herein refers to a protein receptor on T cells that is composed of a heterodimer of an alpha (a) and beta (β) chain, although in some cells the TCR consists of gamma and delta (γ/δ) chains. In embodiments of the invention, the TCR may be modified on any cell comprising a TCR, including a helper T cell, a cytotoxic T cell, a memory T cell, regulatory T cell, natural killer T cell, and gamma delta T cell, for example.
II, General Embodiments of the Invention
[0069] Chimeric antigen receptors (CARs) are designed for adoptive immunotherapy by connecting an extracellular antigen-binding domain to a transmembrane domain and an intracellular signaling domain (endodomain). It is a useful anti-tumor approach to eradicate tumor cells by adoptive transfer of T ceils expressing chimeric antigen receptors to recognize specific antigens presented on tumor cells and activate T cells to specifically lyse these tumor cells. A critical aspect of this CAR strategy is the selection of target epitopes that are specifically or selectively expressed on tumors, are present on all tumor cells, and are membrane epitopes not prone to shed or modulate from the cell surface. However, ideally the CART T cells would be able to be used as a universal reagent or drug suitable for any mammalian (such as human) recipient. To employ the cells in such a manner, one must prevent their rejection in a graft -versus-host response without compromising CAR-dependeni effector functions.
[0070] In embodiments of this invention, T-cell receptor (TCR) β disruption from chimeric antigen receptor (CAR)-expressing T cells (CAR T cells) to establish "universal" T cell-based immu otherapy is provided. Redirecting T-cell specificity to desired antigen can be achieved through CAR. However, ex vivo generation of CAR T cells from patient is limited by time and expense. Moreover, T cells derived from patients are sometimes functionally flawed because of the multiple rounds of lymphotoxic (lympliodepleting) chemotherapy. To this end, embodiments of the present invention concern the generation of CAR' T cells from healthy volunteer donor that can serve as "off-the-shelf reagents. In other words, genetically modified T ceils from one donor can be pre -prepared and then infused into multiple recipients. This will facilitate "centralized" manufacturing of the universal T cells and subsequent pre -positioning of the T cells at regional facilities for infusion on demand, enable clinical trials to be undertaken that are powered for efficacy, and facilitate combination therapies in which the universal T ceils can be administered with other biologies and therapeutics. To achieve this, one can eliminate endogenous TCR αβ and γδ expression, which causes unwanted allogeneic immune reaction. Such steps can occur by any suitable manner, including by introducing zinc finger nucleases (ZFN), for example, targeting TCR a constant region or β constant region. Single ZFN transfection from in vitro transcribed mRNA generates up to 40% TCR αβ dismpted CAR+ T cells and the TCRap™'8 population is readily enriched by clinically compatible paramagnetic beads based sorting. The CAR TCR eg T cells can be propagated to clinically-appealing numbers by recursive additions of irradiated artificial antigen presenting cells (aAPC), Importantly TCRaP"egCAR+ T cells actually do not respond to TCR αβ-CDS stimulation while preserving target antigen specific function and proliferation through CAR. Embodiments of the invention are unique as they combine (i) redirecting the specificity of T cells by introducing a CAR and (ii) eliminating expression of endogenous TCR to generate a desired T-cell product. In certain embodiments, the introduction of CAR and elimination of TCR are accomplished by electroporation using the Sleeping Beauty (SB) system to stably express CAR and desired transient transfection of in vv'iro-transcribed mRNA coding for ZFNs. In embodiments of the invention, infusing specific CARTTCRne T cells are pre-prepared from healthy donors and thawed to be infused on demand as an off-the-shelf reagent.
[0071] The inventors demonstrate that ZFNs targeting either the a or β chains of endogenous TCRs in T cells resulted in the desired loss of TCR expression. As expected, these modified T cells did not respond to TCR stimulation, but maintained their CAR mediated re-directed specificity for the exemplary antigen, CD 19.
[0072] In certain embodiments of the invention, clinical-grade T-cells are genetically modified ex vivo to express a chimeric antigen receptor (CAR) to redirect specificity to a tumor associated antigen (TAA) thereby conferring anti-tumor activity in vivo. T cells expressing a CDI9-specific CAR recognize B-cell malignancies in multiple recipients independent of MHC because the specificity domains are cloned from the variable chains of a CD 19 monoclonal antibody. The present invention encompasses a major step towards eliminating the need to generate patient-specific T cells by generating "universal" allogeneic TAA-specific T cells from one donor that might be administered to multiple recipients. This was achieved by genetically editing CD 19-specific CAR1" T cells to eliminate expression of the endogenous αβ T-cell receptor (TCR) to prevent a graft-versus-host response without compromising CAR-dependent effector functions. Genetically modified T cells were generated using the Sleeping Beauty system to stably introduce the CD 19-specific CAR with subsequent permanent deletion of a or β TCR chains with designer zinc finger nucleases. The inventors show that these engineered T cells display the expected property of having redirected specificity for CD 19 without responding to TCR stimulation. These CAR CR eg T cells may be used as off-the-shelf therapy for investigational treatment of B-lineage malignancies.
[0873] In particular, to test the feasibility of using allogeneic CAR"1" T cells the inventors modified the culturing process for generating CAR T cells (FIG. 7) to include the editing of the genome of CARnes and CAR1 T cells to irreversibly eliminate expression of the αβ TCR. To knockout the β TCR loci the inventors developed zinc finger nucleases (ZFNs) (Urnov et a!., 2010), comprised of zinc finger protein DNA-binding domains fused to the DNA cleavage domain from the Fokl endonuclease, targeting genomic sequences in the constant regions of the endogenous a or β subunits of the TCR, ZFNs mediate genome editing by catalyzing the formation of a DNA double strand break (DSB) in the genome. Targeting a DSB to a predetermined site within the coding sequence of a gene has been previously shown to lead to permanent loss of functional target gene expression via repair by non-homologous end joining (NHEJ), an error-prone cellular repair pathway that results in the insertion or deletion of nucleotides at the cleaved site (Santiago et a!., 2008; Perez et ai , 2008).
IB. Chimeric Antigen Receptors
[0074] As used herein, the term "antigen" is a molecule capable of being bound by an antibody or T-cell receptor. An antigen is additionally capable of inducing a humoral immune response and/or cellular immune response leading to the production of B and/or T lymphocytes.
[0075] The present invention involves nucleic acids, including nucleic acids encoding an antigen-specific chimeric antigen receptor (CAR), including a CAR that has been humanized to reduce immunogenicity (hCAR), polypeptide comprising an intracellular signaling domain, a transmembrane domain, and an extracellular domain comprising one or more signaling motifs. In certain embodiments, the CAR may recognize an epitope comprised of the shared space between one or more antigens. In certain embodiments, the binding region can comprise complementary determining regions of a monoclonal antibody, variable regions of a monoclonal antibody, and/or antigen binding fragment thereof. In other embodiment, that specificity is derived from a peptide (e.g., cytokine) that binds to a receptor. A complementarity determining region (CDR) is a short amino acid sequence found in the variable domains of antigen receptor {e.g., immunoglobulin and T-cell receptor) proteins that complements an antigen and therefore provides the receptor with its specificity for that particular antigen. Each polypeptide chain of an antigen receptor contains three CDRs (CDR1, CDR2, and CDR3). Since the antigen receptors are typically composed of two polypeptide chains, there are six CDRs for each antigen receptor that can come into contact with the antigen— each heavy and light chain contains three CDRs. Because most sequence variation associated with immunoglobulins and T-cell receptors are found in the CDRs, these regions are sometimes referred to as hypervariable domains. Among these, CDR3 shows the greatest variability as it is encoded by a recombination of the VJ (VDJ in the case of heavy chain and TCR αβ chain) regions. [0876] it is contemplated that the human CAR nucleic acids are human genes to enhance cellular immunotherapy for human patients.
[0077] In a specific embodiment, the invention includes a full length CAR cDNA or coding region. The antigen binding regions or domain can comprise a fragment of the VH and VL chains of a single-chain variable fragment (scFv) derived from a particular human monoclonal antibody, such as those described in U.S. Patent 7, 109,304. The fragment can also be any number of different antigen binding domains of a human antigen-specific antibody. In a more specific embodimeni, the fragment is an antigen- specific scFv encoded by a sequence that is optimized for human codon usage for expression in human cells.
[0078] The arrangement could be multimeric, such as a diabody or multimers. The multimers are most likely formed by cross pairing of the variable portion of the light and heavy chains into what has been referred to by Winters as a diabody. The hinge portion of the construct can have multiple alternatives from being totally deleted, to having the first cysteine maintained, to a proline rather than a serine substitution, to being truncated up to the first cysteine. The Fc portion can be deleted, although there is data to suggest that the receptor preferably extends from the membrane. Any protein that is stable and dimerizes can serve this purpose. One could use just one of the Fc domains, e.g., either the CH2 or CHS domain from human immunoglobulin. One could also use the hinge, CH2 and CH3 region of a human immunoglobulin that has been modified to improve dimerization.
[0079] The intracellular signaling domain of the chimeric receptor of the invention is responsible for activation of at least one of the normal effector functions of the immune ceil in which the chimeric receptor has been placed. The term "effector function" refers to a specialized function of a differentiated cell. Effector function of a T cell, for example, may be cytolytic activity or helper activity including the secretion of cytokines. Effector function in a memory or memory-type T cell includes antigen-dependent proliferation. Thus the term "intracellular signalmg domain" refers to the portion of a protein that transduces the effector function signal and directs the cell to perform a specialized function. While usually the entire intracellular signaling domain will be employed, in many eases it will not be necessary to use the entire intracellular polypeptide. To the extent that a trancated portion of the intracellular signaling domain may find use, such truncated portion may be used in place of the intact chain as long as it still transduces the effector function signal. The term intracellular signaling domain is thus meant to include any trancated portion of the intracellular signaling domain sufficient to transduce the effector function signal. Examples include the zeta chain of the T-cell receptor or any of its homo logs (e.g., eta, delta, gamma, or epsilon), MB1 chain, B29, Fc RUT, Fc R , and combinations of signaling molecules, such as ΟΌ3ζ and CD2.8, 4- 1BB, OX40, and combination thereof, as well as other similar molecules and fragments. Intracellular signaling portions of other members of the families of activating proteins can be used, such as FcyRIII and FcsRL See Gross el al. ( 1992), Stancovski el al. (1993), Moritz et al. (1994), Hwu el al. (1995), Weijtens et al. ( 1996), and Hekele et al (1996) for disclosures of cTCR's using these alternative transmembrane and intracellular domains. In a preferred embodiment, the human CD3 ζ intracellular domain was taken for activation.
[Θ880] The antigen-specific extracellular domain and the intracellular signaling- domain may be linked by a transmembrane domain, such as the human IgG4Fe hinge and Fc regions. Alternatives include the human CD4 transmembrane domain, the human CD28 transmembrane domain, the transmembrane human CD3 domain, or a cysteine mutated human€Ό3ζ domain, or other transmembrane domains from other human transmembrane signaling proteins, such as CD 16 and CD8 and erythropoietin receptor.
[0081] In some embodiments, the CAR nucleic acid comprises a sequence encoding other costimulatory receptors, such as a transmembrane domain and a modified CD28 intracellular signaling domain. Other costimulatory receptors include, but are not limited to one or more of CD28, OX-40 (CD 134), DAP 10, and 4- IBB (CD137). In addition to a primary signal initiated by CD3 , an additional signal provided by a human costimulatory receptor inserted in a human CAR is important for full activation of T cells and could help improve in vivo persistence and the therapeutic success of the adoptive immunotherapy. [Θ882] In particular embodiments, the invention concerns isolated nucleic acid segments and expression cassettes incorporating DNA sequences that encode the CAR. Vectors of the present invention are designed, primarily, to deliver desired genes to immune cells, preferably T cells under the control of regulated eukaryotic promoters, for example, MNDU3 promoter or EFlapha promoter, or Ubiquitin promoter. Also, the vectors may contain a selectable marker if. for no other reason, to facilitate their manipulation in vitro.
[Θ883] Chimeric antigen receptor molecules are recombinant and are distinguished by their ability to both bind antigen and transduce activation signals via immunoreeeptor activation motifs (ITAM's) present in their cytoplasmic tails. Receptor constructs utilizing an antigen-binding moiety (for example, generated from single chain antibodies (scFv)) afford the additional advantage of being "universal" in that they bind native antigen on the target cell surface in an HLA-mdependent fashion. For example, several laboratories have reported on scFv constructs fused to sequences coding for the intracellular portion of the CD3 complex's zeta chain (ζ), the Fc receptor gamma chain, and sky tyrosine kinase (Eshhar et ai, 1993; Fitzer-Attas et ai , 1998). Re-directed T cell effector mechanisms including tumor recognition and lysis by CTL have been documented in several murine and human antigen- scFv: ζ systems (Eshhar, 1997; Altenschmidt ei ai, 1997; Brocker et a!., 1998).
[Θ884] To date non-human antigen binding regions are typically used in constructing a chimeric antigen receptor. A potential problem with using non-human antigen binding regions, such as murine monoclonal antibodies, is the lack of human effector functionality and inability to penetrate into tumor masses. In other words, such antibodies may be unable to mediate complement-dependent lysis or iyse human target ceils through antibody- dependent cellular toxicity or Fc-receptor mediated phagocytosis to destroy cells expressing CAR. Furthermore, non-human monoclonal antibodies can be recognized by the human host as a foreign protein, and therefore, repeated injections of such foreign antibodies can lead to the induction of immune responses leading to harmful hypersensitivity reactions. For murine-based monoclonal antibodies, this is often referred to as a Human Anti-Mouse Antibody (HAMA) response. Therefore, the use of human antibodies is more preferred because they do not elicit as strong a HAMA response as murine antibodies. Similarly, the use of human sequences in the CAR can avoid immune-mediated recognition and therefore elimination by endogenous T ceils that reside in the recipient and recognize processed antigen in the context of HLA. [0085] In some embodiments, the chimeric antigen receptor comprises: a) an intracellular signaling domain, b) a transmembrane domain, and c) an extracellular domain comprising an antigen binding region,
[0086] In specific embodiments, intracellular receptor signaling domains in the CAR include those of the T cell antigen receptor complex, such as the zeta chain of CD3, also Fey RIII costimulatory signaling domains, CD28, DAP 10, CD2, alone or in a series with CD3zeta, for example. In specific embodiments, the intracellular domain (which may be referred to as the cytoplasmic domain) comprises part or all of one or more of TCR zeta chain, CD28, OX40/CD134, 4-1BB/CD137, FcsRTy, ICOS/CD278, ILRB/CD122, IL- 2RG/CD132, DAP molecule, CD27, DAP 10, DAP 12, and CD40. In some embodiments, one employs any part of the endogenous T cell recepior complex in the intracellular domain. One or multiple cytoplasmic domains may be employed, as so-called third generation CARs have at least two or three signaling domains fused together for additive or synergistic effect, for example. [Θ887] In certain embodiments of the chimeric antigen receptor, the antigen-specific portion of the recepior (which may be referred to as an extracellular domain comprising an antigen binding region) comprises a tumor associated antigen or a pathogen-specific antigen.
[0088] A tumor associated antigen may be of any kind so long as it is expressed on the cell surface of tumor cells. Exemplary embodiments of tumor associated antigens include CD 19, CD20, carcinoembryonic antigen, alphafetoprotein, CA- 125, MUC-1, epithelial tumor antigen, melanoma- associated antigen, mutated p53, mutated ras, and so forth.
[0089] In certain embodiments intracellular tumor associated antigens may be targeted, such as HA- 1, surviving, WT1, p53. This can be achieved by a CAR. expressed on a universal T cell that recognizes the processed peptide described from the intracellular tumor associated antigen in the context of HLA. In addition, the universal T cell may be genetically modified to express a T-cell receptor pairing that recognizes the intracellular processed tumor associated antigen in the context of HL A.
[0090] The pathogen may be of any kind, but in specific embodiments the pathogen is a fungus, bacteria, or vims, for example. Exemplary viral pathogens include those of the families of Adenoviridae, Epstein-Barr virus (EBV), Cytomegalovirus (CMV), Respiratory Syncytial Virus (RSV), JC virus, BK virus, HSV, HHV family of viruses, Picornaviridae, Herpesviridae, Hepadnaviridae, Flaviviridae, Retroviridae, Orthomyxoviridae, Parainyxoviridae, Papovaviridae, Polyomavirus, Rhabdoviridae, and Togavkidae. Exemplary pathogenic viruses cause smallpox, influenza, mumps, measles, chickenpox, ebola, and rubella. Exemplary pathogenic fungi include Candida, Aspergillus, Cryplococcus , Histoplasma, Pneumocystis, and Stachybotrys . Exemplary pathogenic bacteria include Streptococcus, Pseudomonas, Shigella, Campylobacter, Staphylococcus, Helicobacter, E, coli, Rickettsia, Bacillus, Bordetella, Chlamydia, Spirochetes, and Salmonella. In one embodiment the pathogen receptor Dectin-1 can be used to generate a CAR that recognizes the carbohydrate structure on the ceil wall of fungi. T ceils genetically modified to express the CAR based on the specificity of Dectin- 1 can recognize Aspergillus and target hyphal growth. In another embodiment, CARs can be made based on an antibody recognizing viral determinants {e.g., the glycoproteins from CMV and Ebola) to interrupt viral infections and pathology.
[0891] In some embodimenis, the pathogenic antigen is an Aspergillus carbohydrate antigen for which the extracellular domain in the CAR recognizes patterns of carbohydrates of the fungal cell wall.
[8892] A. chimeric immunoreceptor according to the present invention can be produced by any means known in the art, though preferably it is produced using recombinant DMA techniques. A nucleic acid sequence encoding the several regions of the chimeric receptor can be prepared and assembled into a complete coding sequence by standard techniques of molecular cloning (genomic library screening, PCR, primer-assisted ligation, scFv libraries from yeast and bacteria, site-directed mutagenesis, etc. ). The resulting coding region can be inserted into an expression vector and used to transform a suitable expression host allogeneic TCR!,es cell line. [0893] As used herein, a nucleic acid construct or nucleic acid sequence or polynucleotide is intended to mean a DNA molecule that can be transformed or introduced into a T cell and be transcribed and translated to produce a product (e.g., a chimeric receptor).
[8894] In an exemplary nucleic acid construct (polynucleotide) employed in the present invention, the promoter is operablv linked to the nucleic acid sequence encoding the chimeric receptor of the present invention, i.e., they are positioned so as to promote transcription of the messenger R A from the DNA encoding the chimeric receptor. The promoter can be of genomic origin or synthetically generated. A variety of promoters for use in T cells are well-known in the art (e.g., the CD4 promoter disclosed by Marodon et a I. (2003)). The promoter can be constitutive or inducible, where induction is associated with the specific cell type or a specific level of maturation, for example. Alternatively, a number of well-known viral promoters are also suitable. Promoters of interest include the β-actin promoter, SV40 early and late promoters, immunoglobulin promoter, human cytomegalovirus promoter, retrovirus promoter, and the Friend spleen focus-forming virus promoter. The promoters may or may not be associated with enhancers, wherein the enhancers may be naturally associated with the particular promoter or associated with a different promoter. [0095] The sequence of the open reading frame encoding the chimeric receptor can be obtained from a. genomic DNA source, a cDNA source, or can be synthesized {e.g., via PCR), or combinations thereof. Depending upon the size of the genomic DN A and the number of introns, it may be desirable to use cDNA or a combination thereof as it is found that introns stabilize the mRNA or provide T cell-specific expression (Barthel and Goldfeki, 2003). Also, it may be further advantageous to use endogenous or exogenous non-coding regions to stabilize the mRNA.
[0096] For expression of a chimeric receptor of the present invention, the naturally occurring or endogenous transcriptional initiation region of the nucleic acid sequence encoding N-terminai components of the chimeric receptor can be used to generate the chimeric receptor in the target host. Alternatively, an exogenous transcriptional initiation region can be used that allows for constitutive or inducible expression, wherein expression can be controlled depending upon the target host, the level of expression desired, the nature of the t arget host, and the like.
[0097] Likewise, a signal sequence directing the chimeric receptor to the surface membrane can be the endogenous signal sequence of N -terminal component of the chimeric receptor. Optionally, in some instances, it may be desirable to exchange this sequence for a different signal sequence. However, the signal sequence selected should be compatible with the secretory pathway of T ceils so that the chimeric receptor is presented on the surface of the T cell. [0898] Similarly, a termination region may be provided by the naturally occurring or endogenous transcriptional termination region of the nucleic acid sequence encoding the C- terminal component of the chimeric receptor. Alternatively, the termination region may be derived from a different source. For the most part, the source of the termination region is generally not considered to be critical to the expression of a recombinant protein and a wide variety of termination regions can be employed without adversely affecting expression. [0899] As will be appreciated by one of skill in the art that, in some instances, a few amino acids at the ends of the antigen binding domain in the CAR can be deleted, usually not more than 10, more usually not more than 5 residues, for example. Also, it may be desirable to introduce a small number of amino acids at the borders, usually not more than 10, more usually not more than 5 residues. The deletion or insertion of amino acids may be as a result of the needs of the construction, providing for convenient restriction sites, ease of manipulation, improvement in levels of expression, or the like. In addition, the substitute of one or more amino acids with a different amino acid can occur for similar reasons, usually not substituting more than about five amino acids in any one domain.
The chimeric construct that encodes the chimeric receptor according to the invention can be prepared in conventional ways. Because, for the most part, natural sequences may be employed, the natural genes may be isolated and manipulated, as appropriate, so as to allow for the proper joining of the various components. Thus, the nucleic acid sequences encoding for the N-terminal and C-terminal proteins of the chimeric receptor can be isolated by employing the polymerase chain reaction (PGR), using appropriate primers that result in deletion of the undesired portions of the gene. Alternatively, restriction digests of cloned genes can be used to generate the chimeric construct. In either case, the sequences can be selected to provide for restriction sites that are blunt-ended, or have complementar overlaps.
[00101] The various manipulations for preparing the chimeric construct can be carried out in vitro and in particular embodiments the chimeric construct is introduced into vectors for cloning and expression in an appropriate host using standard transformation or transfection methods. Thus, after each manipulation, the resulting construct from joining of the DNA sequences is cloned, the vector isolated, and the sequence screened to ensure that the sequence encodes the desired chimeric receptor. The sequence can be screened by restriction analysis, sequencing, or the like. [Θ8102] The chimeric constructs of the present invention find application in subjects having or suspected of having cancer by reducing the size of a tumor or preventing the growth or re-growth of a tumor in these subjects. Accordingly, the present invention further relates to a method for reducing growth or preventing tumor formation in a subject by introducing a chimeric construct of the present invention into an isolated T cell of the subject and reintroducing into the subject the transformed T ceil, thereby effecting anti-tumor responses to reduce or eliminate tumors in the subject. Suitable T cells that can be used include cytotoxic lymphocytes (CTL) or any cell having a T cell receptor in need of disruption. As is well-known to one of skill in the art, various methods are readily available for isolating these cells from a subject. For example, using cell surface marker expression or using commercially available kits (e.g., ISOCELL™ from Pierce, Rockford, ill.).
[00103] It is contemplated that the chimeric construct can be introduced into the subject's own T cells as naked DNA or in a suitable vector. Methods of stably iransfeciing T cells by eieciroporation using naked DNA are known in the art. See, e.g., U.S. Pat. No. 6,410,319. Naked DNA generally refers to the DNA encoding a chimeric receptor of the present invention contained in a plasmid expression vector in proper orientation for expression. Advantageously, the use of naked DNA reduces the time required to produce T cells expressing the chimeric receptor of the present invention.
[Θ8104] Alternatively, a viral vector (e.g., a retroviral vector, adenoviral vector, adeno-associated viral vector, or lentiviral vector) can be used to introduce the chimeric construct into T cells. Suitable vectors for use in accordance with the method of the present invention are non-replicating in the subject's T cells. A large number of vectors are known that are based on viruses, where the copy number of the virus maintained in the cell is low- enough to maintain the viability of the cell. Illustrative vectors include the pFB-neo vectors (STRAT G ENE® ) disclosed herein as well as vectors based on HIV, SV40, EBV, HSV, or BPV.
[ΘΘ105] Once it is established that the transfected or transduced T cell is capable of expressing the chimeric receptor as a surface membrane protein with the desired regulation and at a desired level, it can be determined whether the chimeric receptor is functional in the host cell to provide for the desired signal induction. Subsequently, the transduced T cells are reintroduced or administered to the subject to activate anti-tumor responses in the subject. To facilitate administration, the transduced T cells according to the invention can be made into a pharmaceutical composition or made into an implant appropriate for administration in vivo, with appropriate carriers or diluents, which further can be pharmaceutically acceptable. The means of making such a composition or an implant have been described in the art (see, for instance. Remington's Pharmaceutical Sciences, 16th Ed., Mack, ed. (1980)). Where appropriate, the transduced T cells can be formulated into a preparation in semisolid or liquid form, such as a capsule, solution, injection, inhalant, or aerosol, in the usual ways for their respective route of administrat on. Means known in the art can be utilized to prevent or minimize release and absorption of the composition until it reaches the target tissue or organ, or to ensure timed-release of the composition. Desirably, however, a pharmaceutically acceptable form is employed that does not ineffectuate the cells expressing the chimeric receptor. Thus, desirably the transduced T cells can be made into a pharmaceutical composition containing a balanced salt solution, preferably Hanks' balanced salt solution, or normal saline.
IV. Exemplary Human CDI9-specifie Chimeric T-ccll Receptor (or chimeric antigen receptor, CAR)
[00106] CD19, a cell surface glycoprotein of the immunoglobulin superfamily, is a potentially attractive target for antibody therapy of B cell-associated malignancies. This antigen is absent from hematopoietic stem cells, and in healthy individuals its presence is exclusively restricted to the B-lmeage and possibly some follicular dendritic cells (Scheuermann et a!.., 1995). In fact, it is present on B cells from the earliest recognizable B- lineage cells during development to B-cell blasts but is lost on maturation to plasma cells. Furthermore, CD 19 is not shed from the ceil surface and rarely lost during neoplastic transformation (Scheuermann et αί, 1995). The protein is expressed on most malignant B- lineage cells, including cells from patients with chronic lymphocytic leukemia (CLL), non- Hodgkin lymphoma (NHL), and acute lymphoblastic leukemia (ALL) (Uckun et ah, 1988). CD 19 primarily acts as a B cell co-receptor in conjunction with CD21 and CD81. Upon activation, the cytoplasmic tail of CD 19 becomes phosphorylated, which leads to binding by Src-famify kinases and recruitment of PI-3 kinase.
[00107] In one aspect compositions of the invention include a human CD 19- specific chimeric T cell receptor (or chimeric antigen receptor, CAR) polypeptide (designated hCD 19CAR) comprising an intracellular signaling domain, a transmembrane domain, and an extracellular domain, the extracellular domain comprising a human CD 19 binding region. In another aspect, the CD 19 binding region is an F(ab')2, Fab', Fab, Fv, or scFv. The binding region may comprise an amino acid sequence that is at least, at most or about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the wild-type amino acid sequence. The intracellular domain may comprise an intracellular signaling domain of human CO3 , and ma further comprise human CD28 intracellular segment. In certain aspects the transmembrane domain is a CD28 transmembrane domain.
[Θ8108] In a further aspect compositions of the invention include a nucleic acid encoding the polypeptide described above. In certain aspects ihe nucleic acid sequence is optimized for human codon usage.
[Θ81Θ9] In still a further aspect compositions of the invention include cells expressing the polypeptide described herein, 'The T cell may comprise an expression cassette encoding hCD 19CAR polypeptide. The expression cassette can be comprised in a non- viral vector, such as a transposon, or a human transposon, or recombinant variant thereof. The expression cassette can be comprised in a viral vector or recombinant variant thereof. The expression cassette can be genomically integrated or episomally maintained or expressed from mRNA.
[Θ811Θ] In yet a further aspect the invention includes a method of making a T cell expressing a human CD 19 -specific CAR comprising introducing an expression cassette into the cell, wherein the expression cassette encodes a polypeptide comprising a human extracellular CD 19 binding domain, a transmembrane domain, and one or more intracellular signaling domain(s). The method may further comprise stimulating the cells with CD 19 cells, recombinant CD 19, or an antibody to the receptor to cause ihe cells to proliferate, kill, and/or make cytokines; for example, the cells may be stimulated to proliferate or expand with CD 19 f artificial antigen presenting cells. ΘΘΠ 1] In certain aspects the invention includes methods of treating a human disease condition associated with a TCRneg cell expressing endogenous CD 19 comprising infusing a patient with an amount of a recombinant cell expressing a human CD 19--speciflc CAR sufficient to treat the condition, wherein the human CD19-specific CAR comprises a human CD 19 extracellular binding domain, a transmembrane domain, and an intracellular signaling domain. The condition can be lymphoma, leukemia, Non-Hodgkin's lymphoma, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, chronic lymphocytic leukemia, or B cell-associated autoimmune diseases, for example.
[3)8112] The invention relates to the generation of a human CD 19-specific chimeric antigen receptor (hCD19RCD28 or hCAR). in certain aspects recombinant cells expressing hCAR have improved in vivo persistence and anti- tumor efficacy. The human hCAR has a reduced immunogenicity compared to murine hCAR, which comprises a scFv segment derived from a murine CD19-specific monoclonal antibody (mAb), Anti-tumor effects can be augmented by genetically modified cells, such as TCRrje8 cells rendered specific for CD 19. Typically, T cell specificity is achieved by electrotransfer of an expression cassette encoding hC AR.
[00113] The hCAR may be a chimeric receptor comprising one or more activation endodomain(s), such as a CD3 ^-derived activation domain. Additional T-cell activation motifs include, but are not limited to, CD28, OX-40, and 4-1 BB. In certain aspects the activation domain can also include a CD28 transmembrane and/or activation domain. In a further aspect the hCAR encoding region and/or expression cassette codon optimized for expression in human cells and subjects, e.g., in one embodiment the scFv region obtained from VH and VL sequences of a CD 19-specific human antibodies are incorporated into the CD19 binding segment of the hCAR (for example see U.S. Patent No. 7, 109,304, which is incorporated herein by reference in its entirety). In another embodiment, the hCAR expression cassette is episomally maintained or integrated into the genome of the recombinant cell. In certain aspects the expression cassette is comprised in a nucleic acid capable of integration by using an integrase mechanism, a viral vector, such as a retroviral vector, or a nonviral vector, such as a transposon mechanism. In a further embodiment the expression cassette is included in a transposon based nucleic acid. In a particular embodiment, the expression cassette is part of a two component Sleeping Beauty (SB) or piggyBac system that utilizes a transposon and transposase for enhanced non-viral gene transfer.
[88114] Recombinant hCAR expressing cells can be numerically expanded to clinically-meaningful numbers. One example of such expansion uses artificial antigen presenting cells (aAPC). Recombinant hCAR expressing cells can be verified and identified by flow cytometry and western blot analyses. Recombinant hCAR expressing T cells, expressing a CD19-specific CAR can recognize and kill CD 19 expressing target cells. In a further aspect, hCAR can be expressed into Universal cells that can be infused across transplantation barriers to help prevent immunogenicity. The hCAR can be used along with human genes for imaging (such as by positron emission tomography, PET) and conditional ablation of T cells, in the event of cytotoxicity. The recombinant cells of the invention can be used in CD 19-specific cellular therapies.
V. Exemplary Methods and Compositions for Disrupting TCR α/β Chains
[3)8115] In the present invention, the T ceils employed in methods and compositions lack functional T cell receptor. Although in some cases one could employ knockdown measures to impact TCR levels post-transcriptionally, such as with siRNA, in particular cases one utilizes knockout means to edit genomic DNA. In certain embodiments, one eliminates expression of the endogenous TCR. In particular embodiments, the system used to genetically modify the T cells employs stably introducing an expression construct that permits permanent deletion of a and/or β cha ins of TCR.
[00116] Although in some cases one could employ homologous recombination, in particular cases one utilizes non-homologous end joining to edit the genome. Any suitable protocol to modify the genome of a particular T cell is useful, although in specific embodiments one can employ zinc finger nucleases or TALE endonucleases.
[0ΘΠ 7] In specific embodiments, one targets disruption of a and/or β chains of
TCR. Although in some eases the TCR in the cell is disrupted prior to modification of the cell with a chimeric receptor, in particular cases the TCR in the cell is disrupted following modification of the cell with the chimeric receptor. The skilled artisan recognizes that this sequence of events is beneficial, given that knockout of the TCR results in loss of the ability to trigger the T cell to activate for proliferation; in the instance where the TCR is disrupted following insertion of the CAR, the cell can then propagate via the CAR. [ΘΘΠ8] In some embodiments, recombinant restriction enzymes called
TALENs are employed that include the TAL effector DNA -binding domain fused to a DNA cleavage domain. The DNA -binding domain of the fusion protein are proteins secreted by Xanthomonas bacteria that comprise a highly conserved 33-34 amino acid sequence. The exception therein is at the 12th and 1 th amino acids that are highly variable (Repeat Variable Diresidue) and correlate with specific nucleotide recognition in a particular code. This correlation between amino acid sequence and DNA recognition provides the generation of specific DNA binding domains by choosing a particular combination of repeat segments containing the proper RVDs, for example,
[08119] TALENs can be used to alter the genome of T cells by inducing double-strand breaks (DSB) that the T cells respond to with repair mechanisms, Non- homologous end joining (NHEJ) rejoins DNA from both sides of a double-strand break in which there is little or no sequence overlap for annealing. In the case of the present invention, this repair mechanism beneficially induces errors in the genome via insertion, deletion, or chromosomal rearrangement such that the T-cell receptor encoded thereby is nonfunctional. [00120] The ability to prevent T-cell receptor expression is dependent on the ability to disrupt the endogenous expression cassette. Since a T cell can be activated by as few as one T-celi receptor molecule, efforts to disrupt T-cell recepior expression at the level of post-transcriptional (or post-translational) modification (such as using interfering RNA species) are unlikely to be completely efficient leading to leakage and expression of the endogenous T-cell receptor. Thus, the only available method to prevent T-cell receptor expression is based on genetic disruption of the alpha and/or beta expression loci as can be achieved using designer zinc finger nucleases,
VI. Methods and Compositions Related to Embodiments of the Invention
[00121] In certain aspects, the invention includes a method of making and/or expanding the antigen-specific redirected TCR"68 cells that comprises transfecting TCR eg cells with an expression vector containing a DNA constract encoding the hCAR, then stimulating the cells with CD 19+ cells, recombinant CD 19, or an antibody to the receptor to cause the cells to proliferate.
[00122] In another aspect, this invention is a method of stably transfecting and re-directing T ceils by electroporation, or other non-viral gene transfer (such as, but not limited to sonoporation) using naked DNA. Most investigators have used viral vectors to carry heterologous genes into T cells. By using naked DNA, the time required to produce redirected T cells can be reduced. "Naked DNA" means DNA encoding a chimeric T-cell receptor (cTCR) contained in an expression cassette or vector in proper orientation for expression. The electroporation method of this invention produces stable trans fectants that express and carry on their surfaces the chimeric TCR (cTCR). [00123] "Chimeric TCR" means a receptor that is expressed by T cells and that comprises intracellular signaling, transmembrane, and extracellular domains, where ihe extracellular domain is capable of specifically binding in an MHC unrestricted manner an antigen that is not normally hound by a T-cell receptor in that manner. Stimulation of the T cells by the antigen under proper conditions results in proliferation (expansion) of the cells and/or production of U. ?.. The exemplary CD19-specific chimeric receptor of this invention is an example of a chimeric TCR.. However, the method is applicable to transfection with chimeric TCRs that are specific for other target antigens, such as chimeric TCRs that are specific for HER2/Neu (Stance vski et al, 1993), ERBB2 (Moritz et at, 1994), folate binding protein (Hwu et al, 1995), renal cell carcinoma (Weitjens et al, 1996), and HIV-1 envelope glycoproteins gp! 20 and gp41 (Roberts et al, 1994). Other cell-surface target antigens include, but are not limited to, CD20, carcinoembryonic antigen, mesotheiin, c-Met, CD56, HERV-K, GD2, GD3, aiphafetoprotein, CD23, CD30, CD123, IL- l lRalpha, kappa chain, lambda chain, CD70, CA-125, MUC-1, EGFR and variants, epithelial tumor antigen, and so forth.
[00124] In certain aspects, the T cells are primary human T cells, such as T cells derived from human peripheral blood mononuclear cells (PBMC), PBMC collected after stimulation with G-CSF, bone marrow, or umbilical cord blood. Conditions include the use of mRNA and DNA and eiectroporation. Following transfection the cells may be immediately infused or may be stored. In certain aspects, following transfection, the cells may be propagated for day s, weeks, or months ex vivo as a bulk population within about 1 , 2, 3, 4, 5 days or more following gene transfer into cells. In a further aspect, following transfection, the transfectants are cloned and a clone demonstrating presence of a single integrated or episomally maintained expression cassette or plasmid, and expression of the chimeric receptor is expanded ex vivo. The clone selected for expansion demonstrates the capacity to specifically recognize and lyse CD 19 expressing target cells. The recombinant T cells may be expanded by stimulation with IL-2, or other cytokines that bind the common gamma-chain (e.g., IL-7, IL- 15, IL-21, and others). The recombinant T cells may be expanded by stimulation with artificial antigen presenting cells. The recombinant T cells may be expanded on artificial antigen presenting cell or with an antibody, such as OKT3, which cross links CD3 on the T cell surface. Subsets of the recombinant T cells may be deleted on artificial antigen presenting ceil or with an antibody, such as Campath, which binds CD52 on the T cell surface. In a further aspect, the genetically modified ceils may be cryopreserved.
[00125] T-cell propagation (survival) after infusion may be assessed by: (i) q-
PCR using primers specific for the CAR; and/or (ii) flow cytometry using an antibody specific for the CAR.
[ΘΘ126] This invention also represents the targeting of a B cell malignancy or disorder including B cells, with the cell-surface epitope being CD 19~specific using a redirected immune T cell that is TCRn g. Malignant B cells are an excellent target for redirected T cells, as B ceils can serve as imniunostimuiatory antigen-presenting cells for T cells. Preclinical studies that support the anti-tumor activity of adoptive therapy with donor- derived C 19-specific TCR" 8 cells bearing a human or humanized CAR include (i) redirected killing of CD 19+ targets, (ii) redirected secretion/expression of cytokines after incubation with CD 19+ targets/stimulator cells, and (iii) sustained proliferation after incubation with CD 19+ targets/stimulator cells. [00127] In certain embodiments of the invention, the CARTCRnes cells are delivered to an individual in need thereof, such as an individual that has cancer or an infection. The cells then enhance the individual's immune system to attack the respective cancer or pathogenic cells. In some cases, the individual is provided with one or more doses of the antigen-specific CAR" CR eg cells. In cases where the individual is provided with two or more doses of the antigen-specific CARfTCR"eg ceils, the duration between the admimstrations should be sufficient to allow time for propagation in the individual, and in specific embodiments the duration between doses is 1, 2, 3, 4, 5, 6, 7, or more days.
[Θ8128] The source of the allogeneic T cells that are modified to include both a chimeric antigen receptor and that lack functional TCR may be of any kind, but in specific embodiments the cells are obtained from a bank of umbilical cord blood, peripheral blood, human embryonic stem ceils, or induced pluripotent stem ceils, for example. The different banks will not share the same HLAs, so multiple banks may be employed.
[Θ8129] Suitable doses for a therapeutic effect would be at least ΗΫ or between about 10s and about 101 cells per dose, for example, preferably in a series of dosing cycles. An exemplary dosing regimen consists of four one-week dosing cycles of escalating doses, starting at least at about 10° ceils on Day 0, for example increasing incrementally up to a target dose of about 10'° cells within several weeks of initiating an intra-patient dose escalation scheme. Suitable modes of administration include intravenous, subcutaneous, intracavitary (for example by reservoir-access device), intraperitoneal, and direct injection into a tumor mass, [08130] A pharmaceutical composition of the present invention can be used alone or in combination with other well-established agents useful for treating cancer. Whether delivered alone or in combination with other agents, the pharmaceutical composition of the present invention can be delivered via various routes and to various sites in a mammalian, particularly human, body to achieve a particular effect. One skilled in the art will recognize that, although more than one route can be used for administration, a particular route can provide a more immediate and more effective reaction than another route. For example, intradermal delivery may be advantageously used over inhalation for the treatment of melanoma. Local or systemic delivery can be accomplished by administration comprising application or instillation of the formulation into body cavities, inhalation or insufflation of an aerosol, or by parenteral introduction, comprising intramuscular, intravenous, intraportal, intrahepatic, peritoneal, subcutaneous, or intradermal administration.
[00131] A composition of the present invention can be provided in unit dosage form wherein each dosage unit, e.g. , an injection, contains a predetermined amount of the composition, alone or in appropriate combination with other active agents. The term unit dosage form as used herein refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of the composition of the present invention, alone or in combination with other active agents, calculated in an amount sufficient to produce the desired effect, in association with a pharmaceutically acceptable diluent, carrier, or vehicle, where appropriate. The specifications for the novel unit dosage forms of the present invention depend on the particular pharmacodynamics associated with the pharmaceutical composition in the particular subject.
[Θ8132] Desirably an effective amount or sufficient number of the isolated transduced T ceils is present in the composition and introduced into the subject such that long-term, specific, anti-tumor responses are established to reduce the size of a tumor or eliminate tumor growth or regrowth than would otherwise result in the absence of such treatment. Desirably, the amount of transduced T cells reintroduced into the subject causes a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, or 100% decrease in tumor size when compared to otherwise same conditions wherein the transduced T cells are not present.
[08133] Accordingly, the amount of transduced T cells administered should take into account the route of administration and should be such that a sufficient number of the transduced T cells will be introduced so as to achieve the desired therapeutic response. Furihermore, the amounts of each active agent included in the compositions described herein (e.g., the amount per each cell to be contacted or the amount per certain body weight) can vary in different applications. In general, the concentration of transduced T cells desirably should be sufficient to provide in the subject being treated at least from about 1 10" to about 1 x 109 transduced T cells, even more desirably, from about 1 x 10'' to about 5 x 108 transduced T cells, although any suitable amount can be utilized either above, e.g., greater than 5 x 10¾ cells, or below, e.g., less than 1 x 10' cells. The dosing schedule can be based on well-established cell-based therapies (see, e.g., Topalian and Rosenberg, 1987; U.S. Pat. No. 4,690,915), or an alternate continuous infusion strategy can be employed. [00134] These values provide general guidance of the range of transduced T cells to be utilized by the practitioner upon optimizing the method of the present invention for practice of the invention. The recitation herein of such ranges by no means precludes the use of a higher or lower amount of a component, as might be warranted in a particular application. For example, the actual dose and schedule can vary depending on whether the compositions are administered in combination with other pharmaceutical compositions, or depending on interindi vidua! differences in pharmacokinetics, drug disposition, and metabolism. One skilled in the art readily can make any necessary adjustments in accordance with the exigencies of the particular situation.
VII. Immune System and Immunotherapy
[00135] In some embodiments, a medical disorder is treated by transfer of a redirected T cell that elicits a specific immune response. In one embodiment of the present invention, B cell lineage malignancy or disorder is treated by transfer of a redirected T ceil that elicits a specific immune response. Thus, a basic understanding of the immunologic responses is necessaiy. [00136] The cells of the adaptive immune system are a type of leukocyte, called a lymphocyte. B cells and T cells are the major types of lymphocytes. B cells and T cells are derived from the same pl ripotent hematopoietic stem ceils, and are indistinguishable from one another until after they are activaied. B cells play a large role in the humoral immune response, whereas T cells are intimately involved in cell-mediated immune responses. They can be distinguished from other lymphocyte types, such as B cells and NK cells by the presence of a special receptor on their cell surface called the T-celi receptor (TCR). In nearly all other vertebrates, B cells and T ceils are produced by stem cells in the bone marrow. T cells travel to and develop in the thymus, from which they derive their name. In humans, approximately l%-2% of the lymphocyte pool recirculates each hour to optimize the opportunities for antigen-specific lymphocytes to find their specific antigen within the secondary lymphoid tissues.
[Θ8137] T lymphocytes arise from hematopoietic stem cells in the bone marrow, and migrate to the thymus gland to mature. T cells express a unique antigen binding receptor on their membrane (T-cell receptor), which can only recognize antigen in association with major histocompatibility complex (MHC) molecules on the surface of other ceils. There are at least two populations of T cells, known as T helper cells and T cytotoxic cells. T helper cells and T cytotoxic cells are primarily distinguished by their display of the membrane bound glycoproteins CD4 and CDS, respectively. T helper cells secret various lymphokines that are crucial for the activation of B cells, T cytotoxic ceils, macrophages, and other cells of the immune system. In contrast, T cytotoxic cells that recognize an anligen- MHC complex proliferate and differentiate into effector ceil called cytotoxic T lymphocytes (CTLs), CTLs eliminate cells of the body displaying antigen, such as virus infected ceils and tumor cells, by producing substances that result in cell lysis. Natural killer cells (or NK cells) are a type of cytotoxic lymphocyte that constitutes a major component of the innate immune system. NK cells play a major role in the rejection of tumors and cells infected by viruses. The cells kill by releasing small cytoplasmic granules of proteins called perforin and granzyme that cause the target cell to die by apoptosis.
[Θ8138] A B cell identifies pathogens when antibodies on its surface bind to a specific foreign antigen. This antigen/antibody complex is taken up by the B cell and processed by proteolysis into peptides. The B cell then displays these antigenic peptides on its surface MHC class II molecules. This combination of MHC and antigen attracts a matching helper T cell, which releases lymphokines and activates the B cell. As the activated B cell then begins to divide, its offspring (plasma cells) secrete millions of copies of the antibody that recognizes this antigen. These antibodies circulate in blood plasma and lymph, bind to pathogens expressing the antigen and mark (hem for destruction by complement activation or for uptake and destruction by phagocytes. Antibodies can also neutralize challenges directly, by binding to bacterial toxins or by interfering with the receptors used by viruses and bacteria to infect cells.
[00139] NK cells or natural killer cells are defined as large granular lymphocytes that do not express T-eeil antigen receptors (TCR) or Pan T marker CDS or surface immunoglobulins (Ig) B cell receptor but that usually express the surface markers CD 16 (FcyRITT) and CD56 in humans, and NKl . l/ K1.2 in certain strains of mice. [00140] Antigen-presenting cells, which include macrophages, B lymphocytes, and dendritic ceils, are distinguished by their expression of a particular MHC molecule. APCs internalize antigen and re-express a part of that antigen, together with the MHC molecule on their outer cell membrane. The major histocompatibility complex (MHC) is a large genetic complex with multiple loci. The MHC foci encode two major classes of MHC membrane molecules, referred to as class I and class II MHCs. T helper lymphocytes generally recognize antigen associated with MHC class II molecules, and T cytotoxic lymphocytes recognize antigen associated with MHC class I molecules. In humans the MHC is referred to as the HLA complex and in mice the H-2 complex.
[00141] The T-cell receptor, or TCR, is a molecule found on the surface of T lymphocytes (or T cells) that is generally responsible for recognizing antigens bound to major histocompatibility complex (MHC) molecules. It is a heterodimer consisting of an alpha and beta chain in 95% of T cells, while 5% of T cells have TCRs consisting of gamma and delta chains. Engagement of the TCR with antigen and MHC results in activation of its T lymphocyte through a series of biochemical events mediated by associated enzymes, co- receptors, and specialized accessory molecules. In immunology, the CDS antigen (CD stands for cluster of differentiation) is a protein complex composed of four distinct chains (CDSv, CD35, and two times CDSe) in mammals, that associate with molecules known as the T-cell receptor (TCR) and the ζ-chain to generate an activation signal in T lymphocytes. The TCR, ζ-chain, and CDS molecules together comprise the TCR complex. The CD3y, CD38, and CD3s chains are highly related cell surface proteins of the immunoglobulin superfamily containing a single extracellular immunoglobulin domain. The transmembrane region of the CDS chains is negatively charged, a characteristic that allows these chains to associate with the positively charged TCR chains (TCRa and TCRfi). The intracellular tails of the CDS molecules contain a single conserved motif known as an immunoreceptor tyrosine-based activation motif or IT AM for short, which is essential for the signaling capacity of the 'T'CR.
[00142] CD28 is one of the molecules expressed on T cells that provide co- stimulatory signals, which are required for T cell activation. CD28 is the receptor for B7.1 (CD80) and B7.2 (CD86). When activated by Toil-like receptor ligands, the B7.1 expression is upregulated in antigen presenting ceils (APCs). The B7.2 expression on antigen presenting cells is constitutive. CD28 is the only B7 receptor co stitutively expressed on naive T cells. Stimulation through CD28 in addition to the TCR can provide a potent co-stimulatory signal to T ceils for the production of various interleukins (IL-2 and IL-6 in particular).
[00143] The strategy of isolating and expanding antigen-specific T ceils as a therapeutic intervention for human disease has been validated in clinical trials (Riddell et al., 1992; Walter ei al., 1995; Heslop et al, 1996).
[08144] Malignant B cells appear to be an excellent targets for redirected T cells, as B cells can serve as immunostimuiatory antigen-presenting cells for T cells (Glimcher et al, 1982). Lymphoma, by virtue of its lymph node tropism, is anatomically ideally situated for T cell-mediated recognition and elimination. The localization of infused T cells to lymph node in large numbers has been documented in HIV patients receiving infusions of HIV-specific CDS" CTL clones. In these patients, evaluation of lymph node biopsy material revealed that infused clones constituted approximately 2%-8% of CDS' cells of lymph nodes. Lymph node homing might be further improved by co-transfecting T cells with a cD A construct encoding the L-selection molecule under a constitutive promoter since this adhesion molecule directs circulating T cells back to lymph nodes and is down- regulated by in vitro expansion (Chao et al., 1997). The present invention may provide a method of treating a human disease condition associated with a cell expressing endogenous CD 19 comprising infusing a patient with a therapeutically effective dose of the recombinant human CD19-specific CAR expressing cell as described above. The human disease condition associated with a cell expressing endogenous CD 19 may be selected from the group consisting of lymphoma, leukemia, non-Hodgkiris lymphoma, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, chronic lymphocytic leukemia, and B cell-associated autoimmune diseases. [00145] Leukemia is a cancer of the blood or bone marrow and is characterized by an abnormal proliferation (production by multiplication) of blood cells, usually white blood cells (leukocytes). t is part of the broad group of diseases called hematological neoplasms. Leukemia is a broad term covering a spectrum of diseases. Leukemia is clinically and pathologically split into its acute and chronic forms.
[00146] Acute leukemia is characterized by the rapid proliferation of immature blood cells. This crowding makes the bone marrow unable to produce healthy blood cells. Acute forms of leukemia can occur in children and young adults. In fact, it is a more common cause of death for children in the U.S. than any other type of malignant disease. Immediate treatment is required in acute leukemia due to the rapid progression and accumulation of the malignant cells, which then spill over into the bloodstream and spread to other organs of the body. Central nervous system (CNS) involvement is uncommon, although the disease can occasionally cause cranial nerve palsies. Chronic leukemia is distinguished by the excessive build up of relatively mature, but still abnormal, blood cells. Typically taking months to years to progress, the cells are produced at a much higher rate than normal cells, resulting in many abnormal white blood cells in the blood. Chronic leukemia mostly occurs in older people, but can theoretically occur in any age group. Whereas acute leukemia must be treated immediately, chronic forms are sometimes monitored for some time before treatment to ensure maximum effectiveness of therapy. [00147] Furthermore, the diseases are classified into lymphocytic or lymphoblastic, which indicate that the cancerous change took place in a type of marrow cell that normally goes on to form lymphocytes, and myelogenous or myeloid, which indicate that the cancerous change took place in a type of marrow ceil that normally goes on to form red cells, some types of white cells, and platelets (see lymphoid cells vs. myeloid cells). [00148] Acute lymphocytic leukemia (also known as acute lymphoblastic leukemia, or ALL) is the most common type of leukemia in young children. This disease also affects adults, especially those aged 65 and older. Chronic lymphocytic leukemia (CLL) most often affects adults over the age of 55. It sometimes occurs in younger adults, but it almost never affects children. Acute myelogenous leukemia (also known as acute myeloid leukemia, or AML) occurs more commonly in adults than in children. This type of leukemia was previously called "acute nonlymphocytic leukemia." Chronic myelogenous leukemia (CML) occurs mainly in adults, A very small number of children also develop this disease. [00149] Lymphoma is a type of cancer that originates in lymphocytes (a type of white blood cell in the vertebrate immune system). There are many types of lymphoma. According to the U.S. National Institutes of Health, lymphomas account for about five percent of all cases of cancer in the United States, and Hodgkin's lymphoma in particular accounts for less than one percent of all cases of cancer in the United States. Because the lymphatic system is pari of the body's immune system, patients with a weakened immune system, such as from HIV infection or from certain drags or medication, also have a higher incidence of lymphoma.
[00150] In the 19th and 20th centuries the affliction was called Hodgkin's Disease, as it was discovered by Thomas Hodgkin in 1832. Colloquially, lymphoma is broadly categorized as Hodgkin's lymphoma and non-Hodgkin lymphoma (all other types of lymphoma). Scientific classification of the types of lymphoma is more detailed. Although older classifications referred to histiocytic lymphomas, these are recognized in newer classifications as of B, T, or NK cell lineage. [0Q1S1] Autoimmune disease, or autoimmunity, is the failure of an organism to recognize its own constituent parts (down to the sub-molecular levels) as "self," which results in an immune response against its own cells and tissues. Any disease that results from such an aberrant immune response is termed an autoimmune disease. Prominent examples include Coeliac disease, diabetes mellitus type 1 (IDDM), systemic lupus erythematosus (SLE), Sjogren's syndrome, multiple sclerosis (MS), Hashimoto's thyroiditis, Graves' disease, idiopathic thrombocytopenic purpura, and rheumatoid arthritis (EA).
[00152] Inflammatory diseases, including autoimmune diseases are also a class of diseases associated with B-cell disorders. Examples of autoimmune diseases include, but are not limited to, acute idiopathic thrombocytopenic purpura, chronic idiopathic thrombocytopenic purpura, dermatomyositis, Sydenham's chorea, myasthenia gravis, systemic lupus erythematosus, lupus nephritis, rheumatic fever, polyglandular syndromes, bullous pemphigoid, diabetes mellitus, Henoch-Schonlein purpura, post- streptococcalnephritis, erythema nodosum, Takayasu's arteritis, Addison's disease, rheumatoid arthritis, multiple sclerosis, sarcoidosis, ulcerative colitis, erythema multiforme, IgA nephropathy, polyarteritis nodosa, ankylosing spondylitis, Goodpasture's syndrome, thromboangitisubiterans, Sjogren's syndrome, primary biliary cirrhosis, Hashimoto's thyroiditis, thyrotoxicosis, scleroderma, chronic active hepatitis, polymyositis/dermatomyositis, polychondritis, pamphigus vulgaris, Wegener's granulomatosis, membranous nephropathy, amyotrophic lateral sclerosis, tabes dorsalis, giant cell arteritis/polymyalgia, peraiciousanemia, rapidly progressive glomerulonephritis, psoriasis, and fibrosing alveolitis. The most common treatments are corticosteroids and cytotoxic drugs, which can be very toxic. These drugs also suppress the entire immune system, can result in serious infection, and have adverse affects on the bone marrow, liver, and kidneys. Other therapeutics that has been used to treat Class III autoimmune diseases to date have been directed against T cells and macrophages. There is a need for more effective methods of treating autoimmune diseases, particularly Class III autoimmune diseases. VIIL Embodiments of Kits of the invention
[ΘΘ153] Any of the compositions described herein may be comprised in a kit. in some embodiments, allogeneic CAR TCR e8 cells are provided in the kit, which also may include reagents suitable for expanding the cells, such as media.
[08154] In a non-limiting example, a chimeric receptor expression construct, one or more reagents to generate a chimeric receptor expression construct, cells for transfection of the expression constract, and/or one or more instruments to obtain allogeneic cells for transfection of the expression construct (such an instrument may be a syringe, pipette, forceps, and'Or any such medically approved apparatus).
[08155] In some embodiments, an expression constract for eliminating endogenous TCR α/β expression, one or more reagents to generate the construct, and/or CAR+ T cells are provided in the kit. In some embodiments, there includes expression constructs thai encode zinc finger nuclease(s).
[00156] In some aspects, the kit comprises reagents or apparatuses for electroporation of cells. [00157] In some embodiments, the kit comprises artificial antigen presenting cells.
[00158] The kits may comprise one or more suitably aliquoted compositions of the present invention or reagents to generate compositions of the invention. The components of the kits may be packaged either in aqueous media or in lyophilized form. The container means of the kits may include at least one vial, test tube, flask, bottle, syringe, or other container means, into which a component may he placed, and preferably, suitably aiiquoted. Where there is more than one component in the kit, the kit also will generally contain a second, third, or other additional container into which the additional components may be separately placed. However, various combinations of components may be comprised in a vial. The kits of the present mvention also will typically include a means for containing the chimeric receptor construct and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow molded plastic containers into which the desired vials are retained, for example.
EXAMPLES
[06159] The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples that follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments that are disclosed and still obtain a like or similar result without departing from the spirit and scope of the mvention.
EXAMPLE 1 EXEMPLARY MATERIALS AND METHODS [00160] Human subjects
[08161] Peripheral blood mononuclear cells (PBMC) were obtained from healthy adult volunteer donors who had provided informed consent from Gulf Coast Regional Center (Houston, TX) in accordance with the Declaration of Helsinki and participated in research approved by MD Anderson Cancer Center (MDACC).
[001 2] ZFNs targeting constant regions of a and β TCR
[00163] ZFNs containing five or six fingers were assembled from an established archive of pre-validated 2-fmger and 1 -finger modules as described (Doyon et ί,, 2008; Isalan et al, 2001 ). ZFN pairs were designed to bind either a sequence within exon 1 of the TCR a constant region (TRAC: NG 001332.2; ZFNs designated as TRAC-ZFN-1 and TRAC-ZFN-2) or a consensus sequence common to exon 1 of both TCR β constant regions 1 and 2 (TRBC1 and TRBC2: NG 001333.2; ZFNs designated as TRBC-ZFN-1 andTRBC- ZF -2). Genes encoding the ZFNs were assembled using PCR-based methodology and cloned into a DNA expression plasmid (pVAX; I dtrogen, Carlsbad, CA). These plasmids were linearized with Xhol and the RiboMAX Large Scale RNA Production System-T7 (Proinega, Madison, WI) with ARCA cap analog (Ambion, Austin, TX) was used to produce and cap mRNA. After in vitro transcription, poly-adenines were added using a poly A tailing kit (Ambion), the integrity and size of the mRNA species was validated on a denaturing 1 % agarose gel with 3-(N-morpholino) propanesulphonic acid (MOPS) buffer, and concentration was measured using a spectrophotometer (BioRad, Hercules, CA) at OD26o. The mRNA was stored at -80°C in nuclease-free vials for single use.
[Θ8164] Flow cytometry
[Θ8165] The following monoclonal antibodies (inAbs) and reagents were used with indicated specificity and the appropriate isotype controls. From BD Biosciences (San Jose, CA): phycoerythrin (PE)-conjugated anti-CD3e (cat # 347347, clone SK7), PE-anti- CD19 (cat # 555413, clone HIB-19), PE-CD64 (cat # 558592, clone 10.1 ), PE-CD86 (cat # 555658, clone 2331 ), PE-CD137L (cat # 559446, clone C65-485), FITC-conjugated anti-CD4 (cat # 555346, clone RPA-T4), FITC-anti-CD8 (cat #555634, clone HIT8a), PE-mouse IgG2bK (cat # 555058), and FITC-mouse IgGl (cat # 349041), From Jackson ImmunoResearch (West Grove, PA): PE-anti-mouse Fab (H+L) (cat # 1 15-1 16-146). An Alexa 488-conjugated CAR-specific antibody (clone 136-20- 1) that recognizes an epitope within scFv region of CD19RCD28 was generated in the laboratory. TCR Ύβ usage was analyzed by a panel of anti-Vp monoclonal antibodies (IQTest® Beta Mark; Beckman Coulter, Brea, CA). The inventors added propidium iodide (Sigma-Aldrich, St. Louis, MO) just before collecting ceils on a flow-cytometer to exclude dead cells from analysis. Data was acquired on a FACS Calibur (BD Biosciences) using CellQuest version 3.3 (BD Biosciences) and analyzed by FCS Express version 3.00 (De Novo Software, Los Angeles, CA) or FlowJo version 7.6.1 (Tree Star, Inc. Ashland, OR).
[88166] Artificial antigen presenting ceils [081 7] K562-derived aAPC were previously modified by lentiviral transduction to constttutively co-express CD 19, CD64, CD86, CD137L, membrane-bound (MB) IL- 15, and EGFP (the latter encoded following the emcv IRES element). A clone (#4) was obtained by limiting dilution and numerically expanded for use (Manuri et at, 2010). For some experiments CD3-specific antibody (OKT3; eBioscience, San Diego, CA) was used to activate T cells by pulsing the mAb onto the CD64" (FcR) clone #4 (FIG. 8). Expression of desired transgenes and bound OKT3 was validated weekly by How cytometry before use in co-culture with T cells.
[00168] Propagation of primary T cells
[08169] T ceils derived from human PBMC, isolated by density gradient separation using Ficoli-Paque Pius (GE Healthcare, Pittsburgh, PA), were numerically expanded i the presence of 50 lU/mL of recombinant human interieukin-2 ([rhIL-2] added three times a week; Chiron, Emeryville, CA) on γ- irradiated (100 Gy) aAPC (clone #4, 1 :2 T cell : aAPC ratio) that had been pre-loaded with OKT3. T cells with aAPC were cultured in complete medium (CM) defined as Hyclone-RPMI 1640 (Thermo Fisher Scientific, Waltham, MA) supplemented with 2 mnioi/L L-glutamine (Glutamax- l ; mvitrogen) and 10% heat-inactivated Hyclone-fetal bovine serum (Thermo Fisher Scientific).
[00170 J Generation and propagation of CAR+ T cells
[00171] DNA. supercoiled plasmids (15 g of CD19RCD28/pSBSO and 5 jig of pKan-CMV-SBl 1) (Davies et at, 2010) encoding the SB transposon (to stably express CD19RCD28) and the SB transposase (to transiently express SB 1 1 ) were electro-transferred using an Amaxa Nucleofector II device (Lonza, Basel, Switzerland) at 2 χ 107 PBMC/cuvette as previously described (FIG, 7) (Singh et at, 2008), T cells expressing CD19RCD28 were preferentially propagated in CM by recursive addition every 7 or 14 days of clone #4 (not loaded with OKT3) at 1 :2 T cell : aAPC (γ- irradiated to 100 Gy) ratio in the presence of rhlL- 2 50 lU/mL, added three times a week. [00172] Electro-transfer of messenger RNA species into primary or CAR+ 7 cells
[00173] Six days after stimulation of unmodified T cells with OKT3-loaded clone #4 or 2 to 4 days after the last stimulation of CD19RCD281 T cells with clone #4, 5 x 106 T cells were mixed with 2.5 to 10.0 ,g of each ZFN mRNA in 100 μΕ of Human T-Cell Nucleofector solution (Cat #VPA- 1002, Lonza) and electroporated using the Nucleofector II device with program T-20. Following electroporation, cells were immediately placed in 2 mL of pre-warmed CM and cultured at 37°C, 5% CQ2 for 4 to 6 hours and then 50 IU/mL of rhIL-2 was added with 2 mL of 20% FBS-RPMI. In some experiments, to enhance ZFN- mediated enzymatic activity, after overnight culture, cells were transferred to 30°C, 5% C02 and cultured for 2 days then returned to 37°C, 5% C02.
[00174] Enrichment ofCD3nes T cells
[08175] Cells washed with PBS supplemented with 2% FBS and 2 mM EDTA, were incubated for 10 minutes with CDS microbeads (Cat # 130-050-101, MilteneyiBiotec, Auburn, CA) at 4°C, After washing twice, cells were passed through an LD column (Cat # 130-042-901, MilteneyiBiotec), and ihe flow-through fraction was collected for further use.
[08176] Surveyor Nuclease assay
[00177] The levels of genomic disruption of TRAC, TRBC1, and TRBC2 in T cells were deiermined by Surveyor Nuclease assay (Transgenornics, Omaha, NE) (Guschm et ai , 2010). The percent target disruption was quantified by densitometry. The PCR primers used for the amplification of target locus are:
[00178] TRAC forward, 5'-GGGCAAAGAGGGAAATGAGA-3' (SEQ ID
NO: ! )
[08179] TRAC reverse, 5'-CAATGGATAAGGCCGAGACC-3' (SEQ TD
NO:2)
[00180] TRBC1 forward, 5'-CTGAACAAGGTGTTCCCACCC-3' (SEQ ID
NO:3)
[08181] TRBC1 reverse 5'-GTGTGCGCTGGTTCCTTTCTT-3' (SEQ ID
NO:4)
[00182] TRBC2 forward, 5'-CCTGGCCACAGGCTTCTACC-3' (SEQ ID
NO:5)
TRBC2 reverse 5'-CCACCTTGTCCACTCTGGCTT- [00184] 5i Chromium- release assay
[08185] Target cells were labeled with 0.1 mCi of D lCr (Perkm Elmer, Boston, MA) for 2 hours. After washing thrice with ice-cold CM, labeled ceils were diluted and plated at 10 cells/well in 100 uL CM in 96-weJl v-bottomed plates, T cells were added in 100 uLAvell at indicated effector target ratios and the plate was spun (i SOg for 3minutes without brake) to facilitate cell-to-cell contact. After a four hour incubation at 37°C, 5%C02, 50 μΐ, of supernatants were counted on TopCouiit (Perkin Elmer, Shelton, CT). All assays were performed in triplicate. The percent specific lysis was calculated as follows: ((experimental cpm - spontaneous cpm) / (maximum cpm - spontaneous cpm)) x 100.
[00186] PKH-26 dilution assay
[00187] T cells were incubated with 2.0 μΜ of the red- fluorescent lipophilic dye PKH-26 (Cat # PKH26GL, Sigma-Aldrich) for five minutes at room temperature according to the manufacturer's instructions. Cells, 100% labeled with PKH-26, were stimulated with either OKT3 loaded aAPC or CD 19" aAPC in CM supplemented with 50 TU/niL rhIL-2 (added every-other-day). PKH-26-derived fluorescence was measured by flow cytometry 10 days after stimulation and CD 19RCD28 r T cells were revealed using anti-CAR mAb clone 136-20-1.
EXAMPLE 2
DISRUPTION OF THE «p TCR-CD3 COMPLEX ON T CELLS USING ZFNS
[08188] Two ZFN pairs targeting the constant regions of TCR a (TRAC-ZFN-
1 and TRAC-ZFN-2) or TCR β (TRBC-ZFN-1 and TRBC-ZFN-2) (FIG, 1) were developed and tested in primary human T cells propagated ex vivo for six days on OKT3-loaded aAPC (clone #4), Since transient expression of ZFNs is sufficient to mediate gene knockout, the inventors developed a "hit-and-run" delivery strategy to transiently express the ZFNs utilizing electro-transfer of in vitro transcribed mRNA species coding for the ZFN pairs (FIG. 2a). To measure TCR expression the inventors used a mAb specific for CD3E, which is only present on the ceil surface when TCRap is expressed. Nine days after electro-transfer, flow cytometric analysis revealed that ZFN pairs targeting TRAC or TRBC eliminated CD3e expression on primary T cells at levels reaching 19.4% and 5.2%, respectively. The efficiency of TCR knockout correlated with the amount of electro-transferred mRNA (FIG. 2 b, upper panel). Although the electro-transfer of mRNA in primary T cells was well- tolerated, they did observe a slight reduction in cell viability that correlated with increasing the amount of introduced mRNA, ZF -mediated gene disruption has been reported to be more efficient when cells are transiently exposed to mild hypothermia (Doyon et αί, 2010). Thus, the inventors cultured T cells for 2 days at 30°C after electro-transfer. ZFN-mediaied disruption of CD3s was up to 2.4-fold better when electroporated T cells were cultured at 3()°C versus 37°C. Using this approach, 37% and 15% of electroporated T-cells lost expression of CD3s using the ZFN pair targeting TRAC and TRBC, respectively, (FIG. 2b, lower panel) with no change in the levels of CD3 negative ceils in the untransfecied samples and without an appreciable decrease in viability (measured by Trypan blue),
[00189] To confirm that electroporated T cells had been genetically modified at the intended ZFN target sites (TCR a or β loci), a Surveyor Nuclease assay was performed using specific oligonucleotide primers flanking target sites within TRAC, TRBC I, or TRBC2. Cei-i nuclease digestion products, representative of genetic changes induced by the ZFNs, were present only after electro -transfer of ZFN pairs and the percent disruption assessed by densitometry correlated with loss of cell surface CD3s expression (FIG. 2c). These experiments in primary T cells confirmed that ZFNs designed to target TRAC or TRBC lead to permanent disruption of apTCR expression, as assessed by the Surveyor Nuclease assay and confirmed by flow cytometric analysis of CD3s.
EXAMPLE 3
ENRICHMENT OF TCR αβ NEGATIVE T CELLS
[001 0] For future clinical applications, rapid and robust methods for isolating sources of TCR disrupted population will be needed. To begin to address this issue, the inventors enriched the TCR/CD3Beg population by negative selection using clinically- approved paramagnetic beads and a depletion column. With a single depletion step, the CD3sues population was enhanced to over 93% (FIG. 3a). A CD3sne* population could not be enriched from untransfected control cells. Back-to-back depletion steps resulted in >99% enrichment without skewing the CD4 or CD8 T cell subsets (FIG. 9). An analysis of TCR VP repertoire in enriched TCR'¾'S T cells via flow cytometry validated the elimination of TCR-β expression from the cell surface (FIG. 3b), EXAMPLE 4
GENERATION OF TCRmG CAR+ T CELLS BY ZFNS
[06191] To test the ability of ZFN pairs to knock out TCR αβ expression from allogeneic CD 19RCD281 T cells, the inventors initially genetically modified PBMC to stably express the CD19RCD28 CAR using the SB transposon/transposase system. The CD19RCD28" T-cell population was specifically propagated by stimulating with γ- irradiated CD 19" aAPC (clone #4) every seven days (FIG. 7b). After four rounds of stimulation, the inventors observed over 90% CAR expression in T cells similar to previously published results (Singh et ai, 2008). Within 2 to 4 days after the fifth stimulation with CD 19" aAPC, when T cells were activated, the inventors re-electroporated the cells with mRNA encoding the TRAC or TRBC ZFNs (FIG. 4a). Flow cytometry analysis revealed that up to 30% and 26% of CD I9RCD281 T cells lost CD3s expression after transection of the TRAC or TRBC ZFNs, respectively (FIG. 4b). The CD3s eg population was again readily enriched by paramagnetic beads, and the Surveyor Nuclease assay confirmed that the CD3ene8 population contained a high percentage of modified alleles at the intended ZFN target sites within the TRAC and TRBC loci (FIG. 4c). The frequency of TRBCl and TRBC2 disruption at the DNA level was approximately 20%-25% and that of TRAC disruption was approximately 60%. These numbers fit with the observed frequencies of CD3sne8CDl9RCD281 T cells because in each cell only one oui of four TRBC alleles (two TRBC l and two TRBC2) is expressed. Similarly one of two TRAC alleles is expressed in each T cell. Therefore, disruption of the expressed allele is sufficient to achieve the CD3-negative phenorype.
EXAMPLE 5
TCR EGCAR+ T CELLS DO NOT RESPOND TO TCR STIMULATION, BUT DO
MAINTAIN CD 19 SPECIFICITY [Θ8192] The inventors anticipated that TCR^CAR" T cells could not respond to TCR stimulation. To test this, they measured the proliferative response of these cells after stimulation by cross-linking CD3 with OKT3 in comparison to activating CAR for sustained proliferation upon docking with CD 19. TCR"egCD19RCD28 f T cells proliferated in response to CD 19, but not OKT3 (FIG. 5a). Next, the inventors assessed the ability of TCRnegCD19RCD28+ T cells to lyse 0 19 " target cells in a standard 4-hour 5lCr release assay (FIG. 5b). The capacity of TCRuegCAR1" T cells to specifically lyse CD 19 target cells was similar to that observed for TCR ' CD 19RCD28 f T cells. Together, these data confirmed ihat ihe absence of a measurable TCR on TCR egCD19RCD28" T ceils corresponds with abrogation of TCR. activity, but does not impact the CAR to activate genetically modified T cells for proliferation and killing. EXAMPLE 6
TCRNEGCD19RCD28+ T-CELLS CAN BE PROPAGATED ON CD19-EXPRESS1NG
AAPCS
[00193] The inventors confirmed that CD19.RCD28" T cells sustain their proliferative capacity to expand to the cell numbers required for clinical applications. Both ihe i'C -*('i) ! vR( D S and parental TCR¾D19RCD28+ T ceils exhibited similar growth kinetics in response to stimulation with the CD 19 aAPC (FIG. 6). There were no changes in CD3s expression on TCRnegCD19RCD28 T cells before and after propagation. These data confirm that TCR e8CAR f T cells can be propagated to achieve high enough cell numbers for a single donor-derived, modified T cell pool to be sufficient for subsequent infusion into multiple recipients, as the need arises.
EXAMPLE 7
SIGNIFICANCE OF CERTAIN EMBODIMENTS OF THE INVENTION
[00194] Embodiments of the invention show that T cells and indeed CAR" T cells can be genetically edited by ZFNs to eliminate expression of the endogenous αβ TCR. This has therapeutic implications where donor-derived T cells are infused to achieve an anti- rumor effect. Therapeutic success after allogeneic HSCT is defined as achieving a GVL- effect without causing clinically-significant GVHD (Bleakley et al, 2004). Thus, separation of GVL and GVHD is the crucial issue following engraftment of allogeneic hematopoietic stem ceils and strategies to accomplish this are based on infusing desired T-cell effector populations predicted to reduce unwanted allogeneic effects. This includes the adoptive transfer of donor-derived memory T cells employing a narrowed TCR νβ repertoire compared with naive T cells (Chen et al, 2004; Foster et al, 2004) or in vitro depletion of T cells activated through allo-antigens (Amrolia et al, 2003; Hartwig et al, 2006; Wehler el al, 2007). CAR+ T cells expressing alloreactive TCRs can be rendered anergic to disparate HLA while maintaining specificity for CD 19 (Davies et al, 2010) by blockade of co- stimulatory molecules upon co-culture of genetically modified T cells with stimulator cells expressing disparate HLA. This approach has clinical significance as it can be undertaken in compliance with current good manufacturing practice. An alternative to pre-selection includes conditional ablation of infused allogeneic CAR1 T cells in the event that serious adverse events occur. This has been accomplished by genetic modification of allogeneic T cells to express "suicide genes" such as HSVtk (Bonini et al, 1997), iCasp9 (Straathof et ai., 2005), CD2Q (mtrona et al , 2000), TMPK (Sato et al, 2007), and a modified Fas (Berger et al, 2004) that can be triggered for conditional ablation via the administration of specific molecules {e.g., ganciclovir to HSVtk expressing ceils). [Θ8195] The inventors recognized that approaches to selectively deplete T cells expressing undesired αβ TCR may be incomplete and that complete knockout of the endogenous TCR might be advantageous not only to prevent GVHD, but also to prevent (he endogenous TCR from adversely affecting CAR function (e.g. , through competition for transcription factors). Therefore, the inventors undertook a genetic approach using designer ZFNs to permanently disrupt the a and β constant region sequences in T cells, thereby eliminating TCR expression. Since TCR αβ receptors need to form heterodimers to express a functional cell surface molecule, knocking out either TRAC or TRBC was sufficient to eliminate TCR αβ expression. This is supported by a recent publication showing that a mutation in TRAC gene leads to the Joss of TCR. αβ expression (Morgan et al, 201 1). [Θ8196] ZFNs have been demonstrated to disrupt target gene expression as a consequence of error-prone DNA DSB repair by NHEJ, which in most cases results in a frame shift mutation leading to a premature stop of translation (Santiago et al , 2008). This technology is being evaluated in a pilot clinical trial infusing HIV -resistant T cells generated by ZFN-mediated disruption of the CCRS co-receptor for HIV-1 (Perez et ai, 2008; Holt et al, 2010). ZFNs target and thus disrupt gene expression at the genomic level, which is an advantage over techniques that involve transcriptional repression and require sustained expression of the inhibiting factor (e.g., enforced expression of shRNA to mediate TCR down regulation (Okamoto et al, 2009)). Thai ZFNs can permanently disrupt gene expression after transient expression (without the inherent dangers of genomic integration) enabled the use of in vitro transcribed mRNA species in a "hit-and-run" manner for electro-transfer of ZFNs into T cells. [Θ8197] Previous reports suggest that T-celi activation mediated through an endogenous TCR is required to obtain a fully fimctional CAR in a model sysiem using Jurkat cell lines (Bridgeman et at, 2.010). In contrast, the inventors observed that knocking out TCR β expression from CD19RCD28 T ceils did not appreciably alter the ability of these cells to specifically kill CD19' targets or proliferate in response to CD 19. One reason for this discrepancy other than the difference in host cells may be the use of a 2nd generation CAR, which includes signaling not only through CD3C (signal 1 ) but also CD28 (signal 2; co- stimulation) (Maher et al. , 2002; Kowolik et al., 2006; Savoldo et al., 201 1). Additional inclusion of a CD 137 domain, also demonstrated to enhance the function and survival of CD19RCD28'1' T cells (Carpenito et al., 2009; Wang et al, 2007), may be tested (Milone et al., 2009). A benefit to expressing TCR with known specificity is that activation through the endogenous immune-receptor can be used to propagate T cells to achieve an anti-tumor effect mediated by the CAR (Cooper et al., 2005; Pule et al, 2008). It remains to be tested in humans whether coordinated co- stimulation achieved through multiple CAR signaling endodomains will be sufficient to sustain persistence in vivo or if triggering of T ceils through TCR is needed. However, the propagation of CD19R.CD28" T cells on aAPC modified to co- express CD 19 along with co-stimulatory molecules results in the significant expansion of CAR memory T-celi subsets predicted for prolonged in vivo survival (Numbenjapon et al, 2007; Butler et al , 201 1 ). Therefore, any loss of persistence of TCRBe8CD 19RCD28÷ T cells may be off-set by co-stimulatory properties of aAPC and the encoded CD28 intra-cellular domain within the CAR.
[081 8] Preparing antigen-specific T cells from a third part}/ donor is clinically appealing as these products can be generated, stored, and validated before use and infused to multiple patients immediately as needed (Cooper, 2010). Indeed, third par y T cells have been successfully infused into patients with post-transplantation lymphoproliferative diseases (Barker et al, 2010; Plaque el al., 2002). Despite the fact that a majority of the viral-antigen specific TCR αβ chains demonstrate cross-reactivity to allo-HLA in vivo (Amir et al., 2.010), clinically significant GVHD was not observed. This may be in part due to the ex vivo repetitive antigen stimulation resulting in the emergence of either an oligoclonal or monoclonal TCR β repertoire that decreases the chance of T-ceil alloreactivity. On the contrary, when the inventors numerically expand CD 19RCD28 f T cells through in vitro CD 19 stimulation on aAPC independent of TCR stimulation they did not observe skewing of the TCR νβ usage when measured by a panel of νβ-specific antibodies. Therefore, elimination TCR αβ expression is needed in a manufacturing process to avoid GVHD upon infusion of allogeneic CAR1 T cells.
[081 9] TCRD^CAIT J ce||s can ¾6 generated using a genetic approach to remove (a) endogenous undesired TCR with ZFNs and (b) introduce a desired CAR with the SB system, for example. The genetic approaches to introduce mRNA and DNA use a common electro-transfer platform approved for clinical trials. The inventive approach abolishes the danger of GVHD posed by adoptive transfer of large numbers of allogeneic T cells while maintaining desired efiecior functions mediated by CD19RCD28 CAR io target malignant B cells. This strategy provides an important step to developing a "universal" CAR" T cell that can be manufactured from one donor and administered on demand to multiple patients. One can also focus on inhibiting rejection of the graft by "hiding" the TCRnegCAR1 T cells from ihe recipient's immune system. This may be accomplished, for example, by genetic modifications including knockout of T-celi MHC and over-expression of MHC homologues known to inhibit NK-cefl activity.
EXAMPLE 8
HLA AND TCR KNOCKOUT BY ZINC FINGER NUCLEASES: TOWARD "OFF- THE-SHELF" ALLOGENEIC T-CELL THERAPY FOR CD19+ MALIGNANCIES
[00200] Cell therapy by infusion of T cells can reconstitute immunity to combat pathogens and malignancies. However, the time required to manufacture T cells with ihe desired properties and in sufficient numbers ex vivo is often incompatible with ihe treatment window for patients. Furthermore, autologous T cells from patients with advanced disease may have compromised function and be tolerant to desired antigens. In some embodiments, one can infuse allogeneic T cells that avoid immune-mediated rejection caused by host T cells recognizing disparate major or minor histocompatibility antigens on the infused cells. To broaden the application of T cell therapy, it was characterized whether HLA gene expression can be disrupted by designer zinc-finger nucleases (ZFNs), for example. ZFNs comprise a zinc finger DNA-binding domain designed to bind a specific DNA sequence fused to the cleavage domain of Fokl endonuclease. Since Fokl dimerization is required to introduce a double strand break (DSB), the inventors generated ZFN pairs that flank the intended DNA target sequences in the required spatial conformation. Cellular repair of the DSB by error-prone nonhomologous end joining allows disruption of HLA gene expression. Transfection of ZFN pairs designed to target exon 3 of the HLA-A locus into the human kidney cell line HEK293 resulted in 10% genetic modification of the HLA-A loci. The inventors generated clones of ΗΈΚ293 cells that showed deletion or insertion mutations within the ZFN binding site of one or both HLA-A alleles leading to early termination of translation. These HLA-AnuI1 HEK293 clones evaded HLA-A-restricted lysis by T cell clones, even after interferon-γ and TNF-ct treatment was used to upreguiate HLA expression. Because only transient expression of ZF s is needed to disrupt a target gene, the inventors tested the ability to disrupt HLA-A gene expression by electro-transfer of in vr'/ro-transcribed ZFN niRNA into primary T cells. A single administration of the niRNA encoding the ZFNs targeting HLA-A could render over 40% of primary T cells HLA-A negative. The inventors enriched the HLA-A11"" population by paramagnetic bead separation to obtain a pool of T cells >90% of which lack HLA-A expression. A useful clinical application for HLA™'1 allogeneic T ceils is to redirect their specificity independeni of HLA via expression of a chimeric antigen receptor (CAR) targeting CD 19. Thus, the inventors eliminated HLA-A expression from CD19-specific CAR ' T cells and demonstrated that they (i) evade HLA-A- restricted lysis by T ceil clones and (ii) specifically lysed CD19' tumor targets. Finally, to further improve this T cell product and eliminate potential deleterious immune mediated recognition by the endogenous T cell receptor (TCR) on allogeneic CAR" T cells, the inventors used ZFN pairs targeting the TCR a or the TCR β locus. Transient expression of these ZFNs resulted in permanent disruption of endogenous TCR expression and a highly enriched αβ TCR™11 cell population could be generated by paramagnetic bead selection. Thus, it is useful to provide allogeneic T cells as "off-the-shelf biologies that can be infused on demand as "drugs."
EXAMPLE 9 HLA AND TCR KNOCKOUT BY ZING FINGER NUCLEASES: TOWARD "OFF- THE-SHELF" ALLOGENEIC T-CELL THERAPY FOR CD19+ MALIGNANCIES
[00201] CD 19 is a lineage-specific cell-surface antigen expressed on B-cell malignancies. T cells can be manufactured to target CD 19 independent of HLA molecules by electro-transfer of CD19-specific chimeric antigen receptor (CD19RCD28CAR). This can be useful, given that T cells from patients can be functionally flawed and/or the time to manufacture a T-cell dose may not match the time windo for infusion, given the pace of rumor progression, [00202] In FIG. 10, the inventors generate CD 19RCD28CAR+ T cells from third party healthy donors that can avoid rejection. In doing so, there is avoidance of unwanted immune reactions mediated by the interaction between HLA and TCR.
[00203] The inventors have also used zinc finger nucleases (ZFNs) to achieve elimination of HLA in the genome as well as elimination of the TCR. ZFN comprises two domains: a nuclease domain of Fold restriction enzyme and a designed zinc finger protein (FIG. 1 la). A pair of ZFN can induce DNA double strand break (DSB) at a target nucleotide sequence in the genome. Error-prone non-homologous end joining repair of ZFN-mediated DNA DSB leads to complete elimination of target gene expression (FIG. l ib). [00204] The inventors designed a pair of ZFN (ZFN-R and ZFN-L) to induce
DSB at a coding sequence of the HLA-A gene. HEK 293 cells (karyotyping analysis showed thai this ceil line has four copies of chromosome 6, wherein the HLA-A gene is located) were transduced with a pair of ZFN and HLA-A disruption was evaluated by Surveyor Nuclease (CEL-I) assay. HLA-A-targeted ZFNs disrupt the HLA-A locus in up to 10% of HEK293 cells (FIG. 12a). For detailed analysis, the inventors isolated ZFN-transduced HEK293 clones, and they could not detect HLA-A expression in these clones even after IFNy and TNF treatment (FIG. 12b). These HEK293 clones evade HLA-A restricted CTL clone attack, when they were pulsed with high dose cognate peptide (FIG, 12c),
[00205] HLA-A-targeted ZFNs can also disrupt HLA-A from T cells over 40% when they were expressed from in vitro transcribed mRNA (FIG. 13a). HLA-Anu11 T-cell population can be enriched by simple paramagnetic beads-based sorting (FIG. 13b).
[00206] HLA-A-targeted ZFNs also disrupt HLA-A expression from
CD19RCD28CAR+ T cells (FIG. 14A). HLA-AnaliCD 19RCD28CAR+ T cells evade HLA-A restricted CTL attack (FIG. 14b) but maintain CD 19 specificity (FIG. 14c). [08207] TCR expression can also be disrupted from CD 19RCD28CAR: T cells by TCR a or β constant region-targeted ZFN pairs (FIG. 15a). TCRnu!!CD 19RCD28CAR÷ T cells maintain CD 19 specificity (FIG. 6b) and also can be propagated on CD 19 -expressing K562-based artificial antigen presenting cells (FIG. 15c). [Θ8208] TCR and HLA-A expression can be simultaneously disrupted in
CD19RCD28CAR" T cells by treating the cells with both TCR a or β constant region- targeted ZFN pairs and HLA-A-targeted ZFN pairs (FIG. 16),
[3)8209] One can eiiminaie HLA-A and/or TCR αβ expression from primary T cells and genetically modified T cells by designer ZFNs expressed from electro-transferred mRNA that is amenable to clinical translation. One can enrich HLA"u1 , and/or TCR11"" populations by a single magnetic-beads based selection amenable to clinical translation. The inventors did not observe any adverse effects in ZFN-transduced T cells in terms of ceil growth or function.
REFERENCES
[00210] All patents and publications mentioned in the specification are indicative of the level of those skilled in the art to which the invention pertams. Ail patents and publications are herein incorporated by reference in their entirety to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference,
PATENTS AND PATENT APPLICATIONS [00211] U.S. Patent No. 4,690,915
[00212] U.S. Patent No. 6,410,319
[00213] U.S. Patent No. 6,489,458 [00214] U.S. Patent No. 7, 109,304
PUBLICATIONS [08215] Altenschmidt et al, J. Mol Med , 75:259, 1997.
[00216] Amir et ah , Allo-HLA reactivity of vims-specific memory T cells is common, Blood, 1 15:3146-3157, 2010.
[08217] Amrolia et ah . Selective depletion of donor alloreactive T cells withoui loss of antiviral or antileukemic responses, Blood, 102:2292-2299, 2003.
[00218] Arden et ah , Human T-cell receptor variable gene segment families.
Immunogenetics, 42:455-500, 1 995.
[08219] Barker et ai. Successful treatment of EBV-associated posttransplantation lymphoma after cord blood transplantation using third-party EBV-specific cytotoxic T lymphocytes, Blood, 1 6:5045-5049, 2010.
[08220] Barthel and Goidfeld, /. Immunol , 171 :3612-3619, 2003. [00221] Berger el al , Pharmacologically regulated Fas-mediated death of adoptively transferred T cells in a nonhuman primate model, Blood, 103 : 1261 - 1269, 2004,
[00222] Bleakley and Riddell, Molecules and mechanisms of the graft-versus - leukaemia effect, Nat. Rev. Cancer, 4:371 -380, 2004. [00223] Bonini et al , HSV-TK gene transfer into donor lymphocytes for control of allogeneic graft-versus-leukemia, Science, 276: 1719- 1 72.4, 1997.
[ΘΘ224] Bridgeman et al, The optimal antigen response of chimeric antigen receptors harboring the CDSzeta transmembrane domain is dependent upon incorporation of the receptor into the endogenous TCR/CD3 complex, J. Immunol , 184:6938-6949, 2010. [00225] Blocker et al, Adv. Immunol , 68:257, 1998.
[00226] Butler et al, Establishment of Antitumor Memory in Humans Using in
Vitro-Educated CD8+ T Cells, Sci. Transl. Med. , 3:80ra34, 2010.
[00227] Carpenito et al, Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD 137 domains, Proc. Natl. Acad. Sci. U. S. A., 106:3360-3365, 2009.
[00228] Chao et al, J. Immunol , 159: 1686, 1997.
[00229] Chen et al. Transfer of allogeneic CD62L- memory T cells without graft- versus -host disease, Blood, 103 : 1534- 1541 , 2004.
[00230] Cooper et al, Development and application of CD 19-specific T cells for adoptive immunotherapy of B cell malignancies, Blood Cells Mol Dis., 33 :83-89, 2004.
[00231] Cooper el al. Enhanced antilymphoma efficacy of CD 19 -redirected influenza MP 1 -specific CTLs by cotransfer of T cells modified to present influenza MP 1 , Blood, 105 : 1622- 1 631 , 2005.
[00232] Cooper, Off-the-shelf T-cell therapy, Blood, 1 16:4741-4743, 2010. [00233] Davies et al, Combining CD 19 redirection and alloanergizatton to generate tumor-specific human T cells for allogeneic cell therapy of B-cell malignancies, Cancer Res., 70:3915-3924, 2010. [ΘΘ234] Doyon et al, Transient cold shock enhances zinc-finger nuc lease- mediated gene disruption, Nat. Methods, 7:459-460, 2010.
[08235] Doyon et at., Heritable targeted gene disruption in zebrafish using designed zinc-finger nucleases, Nat. BiotechnoL, 26:702-708, 2008.
[00236] Eshhar et al, Proc, Natl Acad. Sci. U. S. A., 90:720, 1993.
[08237] Eshhar, Cancer Immunol. Immunother.. 45: 131, 997.
[00238] Fitzer-Attas et al, J. Immunol, 160: 145, 1998.
Foster el al, Human CD62L- memory T cells are less responsive to alloantigen stimulation than CD62L+ naive T cells: potential for adoptive immunotherapy and allodepleiion, Blood, 104:2403-2409, 2004.
ΗΗ)24«! Glimcher et al, J. Exp. Med, 155:445, 1982.
Gross et al, FASEB J., 6:3370, 1992.
[00242] Guschin el al, A rapid and general assay for monitoring endogenous gene modification, Methods Mol Biol, 649:247-256, 2010.
[ΘΘ243] Hackett el al, A transposon and transposase system for human application, Mol Ther., 18:674-683, 2010.
[00244] Haque et al, Treatment of Epstein-Barr-virus-positive posttransplantation lymphoproliferative disease with partly HLA-matched allogeneic cytotoxic T cells, Lancet, 360:436-442, 2002.
[00245] Hartwig et al, Depletion of alloreactive T cells via CD69: implications on antiviral, antileukemic and immunoregulatory T lymphocytes, Bone Marrow Transplant., 37:297-305, 2006.
Hekele el al, Int. J. Cancer, 68:232, 1996.
Heslop et αΙ. , ΝαΙ. Med., 2:551 , 1996. [00248] Holt et al. Human hematopoietic stem/progenitor cells modified by zinc-finger nucleases targeted to CCR5 control HTV-1 in vivo, Nat. BiolechnoL, 28:839-847, 2010.
[00249] Hwu et al. , Cancer Res. , 55 : 3369, 1995. [00250] Introna et al. Genetic modification of human T cells with CD20: a strategy to purify and lyse transduced cells with anti-CD20 antibodies, Hum. Gene. Ther., 1 1 :61 1-620, 2000.
[00251] Isaian and Choo, Rapid, high-throughput engineering of sequence- specific zinc finger DNA-binding proteins, Methods Enzymol, 340:593-609, 2001. [08252] Izsvak et al, Translating Sleeping Beaut}/ transposition into cellular therapies: victories and challenges, Bioessays, 32:756-767, 2010.
[00253] Jena et al.. Redirecting T-cell specificity by introducing a tumor- specific chimeric antigen receptor, Blood, 1 16: 1035- 1044, 2010.
[08254] Kochenderfer et al, Eradication of B-iineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD 19, Blood, 1 16:4099-4102, 2010.
[00255] Kohn et al, CARs on Track in the Clinic, Mol. Ther., 19:432-438,
201 1 .
[00256] Kowolik et al, CD2.8 costiniulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells, Cancer Res., 66: 10995-1 1004, 2006.
[08257] Maher et al.. Human T-lyrnphocyie cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor, Nat. Bioiechnol, 20:70-75, 2002.
[00258] Manuri et al, pigg Bac transposon/rransposase system to generate CD 19-specifie T ceils for the treatment of B-iineage malignancies, Hum. Gene Ther., 21 :427- 437, 2010.
[00259] Marodon et al., Blood, 101 :3416-3423, 2003. [ΘΘ26Θ] Milone el al , Chimeric receptors containing CD 137 signal transduction domains mediate enhanced survival of T ceils and increased antileukemic efficacy in vivo, Mol. Ther. , 17: 1453- 1464, 2009.
[00261] Morgan et al . Mutation in the TCRalpha subunit constant gene (TRAC) leads to a human immunodeficiency disorder characterized by a lack of TCRalphabeta T cells, J. Clin. Invest. , 121 :695-702, 201 1. 082 2] Moritz et al. , Proc. Nail. Acad. Sci. U. S. A. , 91 :4318, 1994.
[00263] Numbenjapon et al., Antigen-independent and antigen-dependent methods to numerically expand CD 19-specific CD8+ T cells, Exp. Hematol., 35 : 1083- 1090, 2007.
[88264] Okamoto el al. Improved expression and reactivity of transduced tumor-specific TCRs in human lymphocytes by specific silencing of endogenous TCR, Cancer Res., 69:9003-901 1 , 2009.
[ΘΘ265] Perez et al.. Establishment of HIV- 1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases, Nai. Biotechnol, 26:808-816, 2008.
[00266] Pule et al, Virus-specific T cells engineered to coexpress tumor- specific receptors: persistence and antitumor activity in individuals with neuroblastoma, Nat. Med., 14: 1264- 1270, 2008.
[88267] Remington's Pharmaceutical Sciences, 16th ed.. Mack, ed., 1980. [00268] Riddell et al, Science, 257:238, 1992.
[88269] Roberts et al. , Blood, 84:2878, 1994.
[00278] Santiago et al. , 'T argeted gene knockout in mammalian cells by using engineered zinc-finger nucleases, Proc. Natl. Acad. Sci. U. S. A. , 105 :5809-5814, 2008.
[88271] Sato et al., Engineered human tmpk/AZT as a novel enzyme/prodrug axis for suicide gene therapy, Mol. Ther. , 15:962-970, 2007. [00272] Savoldo et al, CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T ceils in lymphoma patients, J. Clin. Invest. , 121 : 1822- 1826, 201 1.
[00273] Scheuermann et al., Lent Lymphoma, 18:385-397, 1995. [00274] Singh et al.. Redirecting specificity of T-cell populations for CD 19 using the Sleeping Beauty system, Cancer Res., 68:2961-2971 , 2008.
[08275] Siancovski et al., J. Immunol., 151 :6577, 1993.
[00276] Stra.atb.of et al, An inducible caspa.se 9 safety switch for T-eeii therapy, Blood, 105:4247-4254, 2005. [08277] Topalian and Rosenberg, Ada Haematol, 78 Suppl 1 :75-76, 1987.
[00278] Uckun et al , Blood, 71 : 13 -29, 1988.
[88279] Urnov et al, Genome editing with engineered zinc finger nucleases,
Nat. Rev. Genet, 1 1 :636-646, 2010.
[00288] Walter et al, N. Engl. J. Med., 333: 1038, 1995. [88281] Wang et al, Optimizing adoptive polyclonal T ceil immunotherapy of lymphomas, using a chimeric T cell receptor possessing CD28 and CD 137 costimulatory domains, Hum. Gene. Ther., 18:712-725, 2007.
[00282] Wehler et al, Targeting (he activation- induced antigen CD 137 can selectively deplete alloreactive T cells from antileukemic and antitumor donor T-cell lines, Blood, 109:365-373, 2007.
[00283] Weijtens et al., ,1. Immunol, 157:836, 1996.
[88284] Williams, Sleeping beauty vector system moves toward human trials in the United States, Mol. Ther., 16: 1515-1516, 2008.
[00285] Although the present invention and its advantages have been described in detail, it should be understood that various changes, substitutions and alterations can be made herein without departing from the spirit and scope of the invention as defined by the appended claims. Moreover, the scope of the present application is not intended to be limited to the particular embodiments of the process, machine, manufacture, composition of matter, means, methods and steps described in the specification. As one of ordinary skill in the art will readily appreciate from the disclosure of the present invention, processes, machines, manufacture, compositions of matter, means, methods, or steps, presently existing or later to be developed that perform subsianiialiy the same function or achieve substantially the same result as the corresponding embodiments described herein may be utilized according to the present invention. Accordingly, the appended claims are intended to include within their scope such processes, machines, manufacture, compositions of matter, means, methods, or

Claims

1. An isolated in vitro T ceil population wherein ceils of the population comprise an endogenous T cell receptor coding sequence that is either not expressed or which encodes a nonfunctional T cell receptor and/or an endogenous HLA coding sequence that is either not expressed or which encodes a nonfunctional HLA; and a recombinant chimeric antigen receptor comprising an intracellular signaling domain, a transmembrane domain, and an extracellular domain comprising an antigen binding region.
2. The ceils of claim 1 , wherein the T cell receptor is nonfunctional by virtue of one or more disruptions in the coding sequence of a chain, β chain, or both.
3. The cells of claim 1 , wherein the endogenous T cell receptor is knocked out.
4. The cells of claim 1 , wherein the HLA is nonfunctional by virtue of one or more disruptions in the coding sequence.
5. The cells of clai 1 , wherein the endogenous HLA is knocked out.
6. The ceils of claim 1 , wherein the antigen binding region is an F(ab')2, Fab', Fab, Fv, or scFv.
7. The cells of claim 1 , wherein the antigen binding region binds a tumor associated antigen.
8. The cells of claim 7, wherein the tumor associated antigen is CD 19, CD20, carcinoembryonic antigen, alphafetoprotein, CA- 125, MUC- 1 , epithelial tumor antigen, melanoma-associated antigen, mutated p53, mutated ras, HER2/Neu, ERBB2, folate binding protein, HIV- l envelope glycoprotein gp !20, FfiV- 1 envelope glycoprotein gp41 , GD2, CD 123, CD23,CD30 , CD56, c-Met, meothelin, GD3, HERV-K, IL-l l Ralpha, kappa chain, lambda chain, CSPG4, ERBB2, EGFRvTTI, or VEGFR2.
9. The cells of claim 1 , wherein the antigen binding region binds a pathogen antigen.
10. The cells of claim 9, wherein the pathogen antigen is fungal, viral, or bacterial,
1 1. The cells of claim 10, wherein the fungal antigen is from Aspergillus or Candida.
12. The cells of claim 10, wherein the viral antigen is from HSV, RSV, EBV, CMV, JC virus, BK virus, or Ebola.
13. The ceils of claim 1, wherein the intracellular signaling domain is a T-lymphocyte activation domain.
14. The cells of claim 1 , wherein the intracellular signaling domain comprises CD3ζ, CD28, OX40/CD 134, 4- 1BB/CD 137, FceRly, ICOS/CD278, ILRB/CD 122, IL- 2RG/CD 132, DAP molecules, CD70, cytokine receptor, CD40, or a combination thereof.
15. The cells of claim 1 , wherein the transmembrane domain comprises lgG4Fc hinge, Fc regions, CD4 transmembrane domain, CD28 transmembrane domain, the CD3C transmembrane domain, cysteine mutated human CD3Q domain, CD 16 transmembrane domain, CDS transmembrane domain, or erythropoietin receptor transmembrane domain.
16. The cells of claim 1 , wherein the chimeric antigen receptor comprises:
a) an antigen binding domain from a variable region of antigen-specific monoclonal antibody;
b) a signaling domain of C T.)3..': and
c) CD28, CD 137, or both.
17. An in vitro method of generating the cells of claim 1, comprising the steps of:
a) providing a T cell;
b) modifying the T cell to express a recombinant chimeric antigen receptor comprising an intracellular signaling domain, a transmembrane domain, and an extracellular domain comprising an antigen binding region; and
c) modifying the T cell to harbor endogenous T cell receptor and/or HLA coding sequences that are not expressed or encode a nonfunctional T cell receptor and/or HLA.
1 8. The method of claim 17, wherein step b) occurs before step c).
19. The method of claim 17, wherein step c) occurs before step b).
20. The method of claim 17, wherein the T ceil is provided from an umbilical cord blood bank.
21. The method of claim 17, wherein the T cell is provided from a peripheral blood bank, is provided from an umbilical cord blood bank, is an induced pluripotent stem cell, or is a human embryonic stem cell.
22. The method of claim 17, wherein the T cell is allogeneic in reference to one or more intended recipients.
23. The method of claim 17, wherein step b) is further defined as stably introducing the chimeric antigen receptor into the cell.
24. The method of claim 17, wherein step b) is further defined as introducing a polynucleotide that encodes the chimeric antigen receptor to the cell by a transposon/iransposase system or a viral-based gene transfer system.
25. The method of claim 24, wherein the viral-based gene transfer system comprises recombinant retrovirus or lend virus.
26. The method of claim 17, wherein the T cell following modification in steps b), c), or both is propagated by exposing the T cells to artificial antigen presenting cells, by using OKT3 (or equivalent to cross-link CD3) optionally with other co-stimulatory antibodies (e.g., anti-CD28) on beads, or by using OKT3 (or equivalent to cross-link CD3) optionally with other co-stimulatory antibodies (e.g., anti-CD28) mixed with peripheral blood mononuclear cells.
27. The method of claim 17, wherein in step b) a polynucleotide that encodes the recombinant chimeric antigen receptor is electroporated into the T cell.
28. The method of claim 17, wrherein in step b) a polynucleotide that encodes the chimeric antigen receptor is present on a plasmid or viral vector.
29. The method of claim 17, wherein in step c) the endogenous T cell receptor and/or HLA are disrupted by nonhomologous end joining repair.
30. The method of claim 29, wherein the nonhomologous end joining repair is generated by zinc finger nuclease, introduced into the cell by physical means, viral vector, or non-viral vector.
31. The method of claim 29, wherein the nonhomologous end joining repair is generated by TALE nuclease, introduced into the cell by physical means, viral vector, or non- viral vector,
32. A composition comprising cells of the population of claim 1, for use in treating a medical condition.
33. The composition of claim 32, wherein the medical condition is autoimmune disease, cancer, or infection.
34. The composition of claim 32, wrherein the cells are to be provided more than once.
35. The composition of claim 32, wherein the cells are to be provided to the individual at least 1, 2, 3, 4, 5, 6, 7, or more days apart.
36. The composition of claim 33, wherein the medical condition is a cancer, and the cancer is lymphoma, leukemia, non-Hodgkin's lymphoma, acute lympho blastic leukemia, chronic lymphoblastic leukemia, chronic lymphocytic leukemia, or B cell- associated autoimmune diseases,
37. The composition of claim 33, wherein the medical condition is an infection and the infection is an Aspergillus or Candida infection.
PCT/US2012/065506 2011-11-18 2012-11-16 Car+ t cells genetically modified to eliminate expression of t- cell receptor and/or hla WO2013074916A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/358,828 US10391126B2 (en) 2011-11-18 2012-11-16 CAR+ T cells genetically modified to eliminate expression of T-cell receptor and/or HLA
US16/504,427 US11344577B2 (en) 2011-11-18 2019-07-08 Car+ T cells genetically modified to eliminate expression of T-cell receptor and/or HLA

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161561364P 2011-11-18 2011-11-18
US61/561,364 2011-11-18

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/358,828 A-371-Of-International US10391126B2 (en) 2011-11-18 2012-11-16 CAR+ T cells genetically modified to eliminate expression of T-cell receptor and/or HLA
US16/504,427 Division US11344577B2 (en) 2011-11-18 2019-07-08 Car+ T cells genetically modified to eliminate expression of T-cell receptor and/or HLA

Publications (1)

Publication Number Publication Date
WO2013074916A1 true WO2013074916A1 (en) 2013-05-23

Family

ID=48430186

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/065506 WO2013074916A1 (en) 2011-11-18 2012-11-16 Car+ t cells genetically modified to eliminate expression of t- cell receptor and/or hla

Country Status (2)

Country Link
US (2) US10391126B2 (en)
WO (1) WO2013074916A1 (en)

Cited By (152)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103409373A (en) * 2013-08-26 2013-11-27 上海易美生物技术有限公司 Preparation method for T cells modified by MDM2 (murine double minute 2) genetic recipient
WO2014191128A1 (en) * 2013-05-29 2014-12-04 Cellectis Methods for engineering t cells for immunotherapy by using rna-guided cas nuclease system
WO2015075175A1 (en) * 2013-11-22 2015-05-28 Cellectis Method for generating batches of allogeneic t-cells with averaged potency
WO2015090230A1 (en) * 2013-12-19 2015-06-25 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
WO2015121454A1 (en) * 2014-02-14 2015-08-20 Cellectis Cells for immunotherapy engineered for targeting antigen present both on immune cells and pathological cells
WO2015132598A1 (en) * 2014-03-05 2015-09-11 Ucl Business Plc Chimeric antigen receptor (car) with antigen binding domains to the t cell receptor beta constant region
WO2015142675A2 (en) 2014-03-15 2015-09-24 Novartis Ag Treatment of cancer using chimeric antigen receptor
WO2015157384A1 (en) * 2014-04-10 2015-10-15 Seattle Children's Hospital (dba Seattle Children's Research Institute) Defined composition gene modified t-cell products
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
EP2990416A1 (en) * 2014-08-29 2016-03-02 GEMoaB Monoclonals GmbH Universal chimeric antigen receptor expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
WO2016044605A1 (en) 2014-09-17 2016-03-24 Beatty, Gregory Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2016069283A1 (en) 2014-10-31 2016-05-06 The Trustees Of The University Of Pennsylvania Altering gene expression in cart cells and uses thereof
WO2016077526A1 (en) 2014-11-12 2016-05-19 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
WO2016083811A1 (en) * 2014-11-27 2016-06-02 Imperial Innovations Limited Genome editing methods
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
US9394368B2 (en) 2013-02-20 2016-07-19 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
WO2016120216A1 (en) 2015-01-26 2016-08-04 Cellectis mAb-DRIVEN CHIMERIC ANTIGEN RECEPTOR SYSTEMS FOR SORTING/DEPLETING ENGINEERED IMMUNE CELLS
CN105848484A (en) * 2013-10-25 2016-08-10 得克萨斯州大学系统董事会 Polyclonal [Gamma] [delta] T cells for immunotherapy
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
EP2970886A4 (en) * 2013-03-12 2016-08-17 Sangamo Biosciences Inc Methods and compositions for modification of hla
WO2016081518A3 (en) * 2014-11-17 2016-08-18 Adicet Bio, Inc. Engineered gamma delta t-cells
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
CN106222201A (en) * 2016-08-27 2016-12-14 北京艺妙神州医疗科技有限公司 A kind of method preparing CAR T cell and prepared CAR T cell and application thereof
WO2017027392A1 (en) 2015-08-07 2017-02-16 Novartis Ag Treatment of cancer using chimeric cd3 receptor proteins
US9629877B2 (en) 2013-05-14 2017-04-25 Board Of Regents, The University Of Texas System Human application of engineered chimeric antigen receptor (CAR) T-cells
WO2017083582A1 (en) 2015-11-12 2017-05-18 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
US20170137783A1 (en) * 2015-07-21 2017-05-18 Felipe Bedoya Methods for improving the efficacy and expansion of immune cells
WO2017091786A1 (en) 2015-11-23 2017-06-01 Novartis Ag Optimized lentiviral transfer vectors and uses thereof
WO2017114497A1 (en) 2015-12-30 2017-07-06 Novartis Ag Immune effector cell therapies with enhanced efficacy
US9701758B2 (en) 2013-05-24 2017-07-11 Board Of Regents, The University Of Texas System Anti-CD19 scFv (FMC63) polypeptide
WO2017149515A1 (en) 2016-03-04 2017-09-08 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
WO2017165683A1 (en) 2016-03-23 2017-09-28 Novartis Ag Cell secreted minibodies and uses thereof
WO2017181119A2 (en) 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective protein expression
US9855297B2 (en) 2013-05-29 2018-01-02 Cellectis Methods for engineering T cells for immunotherapy by using RNA-guided CAS nuclease system
WO2018007263A1 (en) * 2016-07-06 2018-01-11 Cellectis Sequential gene editing in primary immune cells
US9879087B2 (en) 2014-11-12 2018-01-30 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
WO2018026819A2 (en) 2016-08-01 2018-02-08 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
US9889160B2 (en) 2015-10-05 2018-02-13 Precision Biosciences, Inc. Genetically-modified cells comprising a modified human T cell receptor alpha constant region gene
US9896441B2 (en) 2014-05-05 2018-02-20 Lycera Corporation Tetrahydroquinoline sulfonamide and related compounds for use as agonists of RORγ and the treatment of disease
WO2018058002A1 (en) 2016-09-23 2018-03-29 Fred Hutchinson Cancer Research Center Tcrs specific for minor histocompatibility (h) antigen ha-1 and uses thereof
WO2018064076A1 (en) 2016-09-27 2018-04-05 Cero Therapeutics, Inc. Chimeric engulfment receptor molecules
US9937207B2 (en) 2013-03-21 2018-04-10 Sangamo Therapeutics, Inc. Targeted disruption of T cell receptor genes using talens
WO2018111340A1 (en) 2016-12-16 2018-06-21 Novartis Ag Methods for determining potency and proliferative function of chimeric antigen receptor (car)-t cells
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
US10040846B2 (en) 2012-02-22 2018-08-07 The Trustees Of The University Of Pennsylvania Compositions and methods for generating a persisting population of T cells useful for the treatment of cancer
WO2018144535A1 (en) 2017-01-31 2018-08-09 Novartis Ag Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
WO2018160731A1 (en) 2017-02-28 2018-09-07 Novartis Ag Shp inhibitor compositions and uses for chimeric antigen receptor therapy
JP2018531612A (en) * 2015-11-04 2018-11-01 フェイト セラピューティクス,インコーポレイテッド Genomic modification of universal cells
US10125193B2 (en) 2014-02-14 2018-11-13 Board Of Regents, The University Of Texas System Chimeric antigen receptors and methods of making
WO2018218038A1 (en) 2017-05-24 2018-11-29 Effector Therapeutics, Inc. Methods and compositions for cellular immunotherapy
WO2018219278A1 (en) 2017-06-01 2018-12-06 Innovative Cellular Therapeutics CO., LTD. Chimeric antigen receptor cell preparation and uses thereof
JP2018536390A (en) * 2015-10-05 2018-12-13 プレシジョン バイオサイエンシズ,インク. Meganucleases engineered to recognition sequences found in the human T cell receptor alpha constant region gene
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
US10166255B2 (en) 2015-07-31 2019-01-01 Regents Of The University Of Minnesota Intracellular genomic transplant and methods of therapy
WO2019002633A1 (en) 2017-06-30 2019-01-03 Cellectis Cellular immunotherapy for repetitive administration
US10174095B2 (en) 2014-07-21 2019-01-08 Novartis Ag Nucleic acid encoding a humanized anti-BCMA chimeric antigen receptor
US10189777B2 (en) 2014-05-05 2019-01-29 Lycera Corporation Benzenesulfonamido and related compounds for use as agonists of RORγ and the treatment of disease
CN109312315A (en) * 2015-12-30 2019-02-05 阿维塔斯有限公司 Gene editing albumen and composition are delivered to the carrier-free of cell and tissue
WO2019047899A1 (en) * 2017-09-06 2019-03-14 亘喜生物科技(上海)有限公司 Universal chimeric antigen receptor t-cell preparation technique
WO2019060425A1 (en) 2017-09-19 2019-03-28 Massachusetts Institute Of Technology Compositions for chimeric antigen receptor t cell therapy and uses thereof
WO2019067328A1 (en) 2017-09-26 2019-04-04 Cero Therapeutics, Inc. Chimeric engulfment receptor molecules and methods of use
WO2019079569A1 (en) 2017-10-18 2019-04-25 Novartis Ag Compositions and methods for selective protein degradation
US10273300B2 (en) 2014-12-29 2019-04-30 The Trustees Of The University Of Pennsylvania Methods of making chimeric antigen receptor-expressing cells
WO2019081983A1 (en) 2017-10-25 2019-05-02 Novartis Ag Antibodies targeting cd32b and methods of use thereof
WO2019084288A1 (en) 2017-10-25 2019-05-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
US10421751B2 (en) 2015-05-05 2019-09-24 Lycera Corporation Dihydro-2H-benzo[b][1,4]oxazine sulfonamide and related compounds for use as agonists of RORγ and the treatment of disease
WO2019191339A1 (en) 2018-03-28 2019-10-03 Cero Therapeutics, Inc. Expression vectors for chimeric engulfment receptors, genetically modified host cells, and uses thereof
WO2019191332A1 (en) 2018-03-28 2019-10-03 Cero Therapeutics, Inc. Chimeric engulfment receptors and uses thereof for neurodegenerative diseases
WO2019191334A1 (en) 2018-03-28 2019-10-03 Cero Therapeutics, Inc. Chimeric tim4 receptors and uses thereof
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
WO2019213282A1 (en) 2018-05-01 2019-11-07 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
WO2019237035A1 (en) 2018-06-08 2019-12-12 Intellia Therapeutics, Inc. Compositions and methods for immunooncology
EP3116902B1 (en) 2014-03-11 2020-01-01 Cellectis Method for generating t-cells compatible for allogenic transplantation
US10532088B2 (en) 2014-02-27 2020-01-14 Lycera Corporation Adoptive cellular therapy using an agonist of retinoic acid receptor-related orphan receptor gamma and related therapeutic methods
WO2020012337A1 (en) 2018-07-10 2020-01-16 Novartis Ag 3-(5-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of i karos family zinc finger 2 (ikzf2)-dependent diseases
US10568947B2 (en) 2014-07-21 2020-02-25 Novartis Ag Treatment of cancer using a CLL-1 chimeric antigen receptor
US10570186B2 (en) 2014-11-05 2020-02-25 Board Of Regents, The University Of Texas System Chimeric antigen receptors (CAR) to selectively target protein complexes
US10570382B2 (en) 2015-03-11 2020-02-25 Board Of Regents, The University Of Texas System Transposase polypeptides and uses thereof
WO2020068261A1 (en) 2018-09-28 2020-04-02 Massachusetts Institute Of Technology Collagen-localized immunomodulatory molecules and methods thereof
US10611740B2 (en) 2015-06-11 2020-04-07 Lycera Corporation Aryl dihydro-2H-benzo[b][1,4]oxazine sulfonamide and related compounds for use as agonists of RORγ and the treatment of disease
US10633445B2 (en) 2015-09-15 2020-04-28 Board Of Regents The University Of Texas System T-cell receptor (TCR)-binding antibodies and uses thereof
US20200200756A1 (en) * 2015-03-05 2020-06-25 Ucl Business Plc. Methods
WO2020128972A1 (en) 2018-12-20 2020-06-25 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
US10709775B2 (en) 2015-08-11 2020-07-14 Cellectis Cells for immunotherapy engineered for targeting CD38 antigen and for CD38 gene inactivation
US10729725B2 (en) 2017-05-12 2020-08-04 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US10730942B2 (en) 2016-07-25 2020-08-04 Ucl Business Ltd Protein-based T-cell receptor knockdown
WO2020165834A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
US20200263133A1 (en) * 2015-09-11 2020-08-20 Agenus Inc. Engineered host cells and methods of use thereof
WO2020191378A1 (en) * 2019-03-21 2020-09-24 Allogene Therapeutics, Inc. METHODS FOR ENHANCING TCRαβ+ CELL DEPLETION EFFICIENCY
US10808230B2 (en) 2015-02-24 2020-10-20 Board Of Regents, The University Of Texas System Selection methods for genetically-modified T cells
WO2020219742A1 (en) 2019-04-24 2020-10-29 Novartis Ag Compositions and methods for selective protein degradation
RU2739770C2 (en) * 2014-06-11 2020-12-28 полибайосепт ГмбХ Expansion of lymphocyte with a cytokine composition for active cellular immunotherapy
WO2020263399A1 (en) 2019-06-26 2020-12-30 Massachusetts Institute Of Technology Immunomodulatory fusion protein-metal hydroxide complexes and methods thereof
US10912797B2 (en) 2016-10-18 2021-02-09 Intima Bioscience, Inc. Tumor infiltrating lymphocytes and methods of therapy
US10947505B2 (en) 2015-11-04 2021-03-16 Fate Therapeutics, Inc. Methods and compositions for inducing hematopoietic cell differentiation
WO2021061648A1 (en) 2019-09-23 2021-04-01 Massachusetts Institute Of Technology Methods and compositions for stimulation of endogenous t cell responses
WO2021067875A1 (en) 2019-10-03 2021-04-08 Cero Therapeutics, Inc. Chimeric tim4 receptors and uses thereof
US11008548B2 (en) 2019-04-03 2021-05-18 Precision Biosciences, Inc. Genetically-modified immune cells comprising a microRNA-adapted shRNA (shRNAmiR)
CN112830974A (en) * 2021-01-08 2021-05-25 深圳市珈钰生物科技有限公司 Chimeric antigen receptor, carrier, human dendritic cell, cell line, solid tumor treatment drug, preparation method and application
EP3680338A4 (en) * 2017-09-08 2021-06-02 Carsgen Therapeutics Co., Ltd. Genetically engineered t cell and application thereof
WO2021106832A1 (en) * 2019-11-25 2021-06-03 国立大学法人京都大学 T-cell master cell bank
WO2021113759A1 (en) * 2019-12-04 2021-06-10 Celularity Inc. Placenta-derived allogeneic car-t cells and uses thereof
US11034748B2 (en) 2017-03-15 2021-06-15 Fred Hutchinson Cancer Research Center High affinity MAGE-A1-specific TCRs and uses thereof
US20210177953A1 (en) * 2017-11-14 2021-06-17 Asclepius Therapy Llc Engineered Non-Human Derived Immune Cells for Universal Adoptive Antigen Cellular Immunotherapy
US11041021B2 (en) * 2014-05-23 2021-06-22 University Of Florida Research Foundation, Incorporated Car based immunotherapy
WO2021123996A1 (en) 2019-12-20 2021-06-24 Novartis Ag Uses of anti-tgf-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
CN113061580A (en) * 2020-01-02 2021-07-02 江苏茂行科技有限公司 Modified immune effector cell and preparation method thereof
US11053484B2 (en) 2017-06-30 2021-07-06 Precision Biosciences, Inc. Genetically-modified T cells comprising a modified intron in the T cell receptor alpha gene
US11059879B2 (en) 2015-10-27 2021-07-13 Board Of Regents, The University Of Texas System Chimeric antigen receptor molecules and uses thereof
WO2021146487A2 (en) 2020-01-14 2021-07-22 Synthekine, Inc. Il2 orthologs and methods of use
EP3699268A4 (en) * 2017-10-20 2021-08-04 Chongqing Precision Biotech Company Limited Universal car-t cell, preparation method therefor and application thereof
US11098325B2 (en) 2017-06-30 2021-08-24 Intima Bioscience, Inc. Adeno-associated viral vectors for gene therapy
US11123369B2 (en) 2015-08-07 2021-09-21 Seattle Children's Hospital Bispecific CAR T-cells for solid tumor targeting
US11166985B2 (en) 2017-05-12 2021-11-09 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
WO2021252920A1 (en) 2020-06-11 2021-12-16 Novartis Ag Zbtb32 inhibitors and uses thereof
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
US11235004B2 (en) 2017-06-30 2022-02-01 Innovative Cellular Therapeutics Holdings, Ltd. Lymphocyte cell lines and uses thereof
US11242377B2 (en) 2015-10-23 2022-02-08 Autolus Limited Chimeric receptor with ligand binding exodomain and cytokine and T cell signalling endodomains
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
US11248058B2 (en) 2015-02-19 2022-02-15 University Of Florida Research Foundation, Inc. Chimeric antigen receptors and uses thereof
WO2022036285A1 (en) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Compositions and methods for treating cancer with chimeric tim receptors in combination with inhibitors of poly (adp-ribose) polymerase
WO2022036287A1 (en) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Anti-cd72 chimeric receptors and uses thereof
WO2022036265A1 (en) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Chimeric tim receptors and uses thereof
US11253609B2 (en) 2017-03-03 2022-02-22 Seagen Inc. Glycan-interacting compounds and methods of use
US11254912B2 (en) 2018-05-11 2022-02-22 Crispr Therapeutics Ag Methods and compositions for treating cancer
US11299708B2 (en) 2016-05-12 2022-04-12 Adicet Bio, Inc. Methods for selective expansion of γδ T-cell populations and compositions thereof
WO2022096664A1 (en) 2020-11-09 2022-05-12 Miltenyi Biotec B.V. & Co. KG Methods and compositions for eliminating engineered immune cells
US11389481B2 (en) 2019-04-30 2022-07-19 Crispr Therapeutics Ag Allogeneic cell therapy of B cell malignancies using genetically engineered T cells targeting CD19
US11401330B2 (en) 2016-11-17 2022-08-02 Seagen Inc. Glycan-interacting compounds and methods of use
US11413340B2 (en) 2015-12-22 2022-08-16 Novartis Ag Mesothelin chimeric antigen receptor (CAR) and antibody against PD-L1 inhibitor for combined use in anticancer therapy
US11434293B2 (en) 2017-06-09 2022-09-06 Autolus Limited Anti TRBC1 antigen binding domains
WO2022215011A1 (en) 2021-04-07 2022-10-13 Novartis Ag USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
WO2022229853A1 (en) 2021-04-27 2022-11-03 Novartis Ag Viral vector production system
WO2023274386A1 (en) * 2021-07-01 2023-01-05 宁波茂行生物医药科技有限公司 Universal car-t cell targeting egfr and preparation method therefor
WO2023010097A1 (en) 2021-07-28 2023-02-02 Cero Therapeutics, Inc. Chimeric tim4 receptors and uses thereof
RU2791327C2 (en) * 2017-06-09 2023-03-07 Отолус Лимитед Anti-trbc1 antigen-binding domain
US11634498B2 (en) 2016-04-29 2023-04-25 University Of Florida Research Foundation, Incorporated Chimeric antigen receptors and uses thereof
US11634688B2 (en) 2015-01-26 2023-04-25 Fate Therapeutics, Inc. Methods and compositions for inducing hematopoietic cell differentiation
US11661459B2 (en) 2020-12-03 2023-05-30 Century Therapeutics, Inc. Artificial cell death polypeptide for chimeric antigen receptor and uses thereof
WO2023122337A1 (en) * 2021-12-23 2023-06-29 Sana Biotechnology, Inc. Chimeric antigen receptor (car) t cells for treating autoimmune disease and associated methods
US11725061B2 (en) 2013-11-27 2023-08-15 The General Hospital Corporation CSGP4—specific chimeric antigen receptor for cancer
US11786554B2 (en) 2018-04-12 2023-10-17 Precision Biosciences, Inc. Optimized engineered nucleases having specificity for the human T cell receptor alpha constant region gene
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors
US11851659B2 (en) 2017-03-22 2023-12-26 Novartis Ag Compositions and methods for immunooncology
US11885806B2 (en) 2014-03-05 2024-01-30 Autolus Limited Method for depleting malignant T-cells
US11896614B2 (en) 2015-04-17 2024-02-13 Novartis Ag Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
US11903967B2 (en) 2018-01-09 2024-02-20 Autolus Limited Method of preparing T cells with increased activity
US11939389B2 (en) 2018-06-13 2024-03-26 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11952408B2 (en) 2017-10-03 2024-04-09 Juno Therapeutics, Inc. HPV-specific binding molecules

Families Citing this family (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9273283B2 (en) * 2009-10-29 2016-03-01 The Trustees Of Dartmouth College Method of producing T cell receptor-deficient T cells expressing a chimeric receptor
US9181527B2 (en) 2009-10-29 2015-11-10 The Trustees Of Dartmouth College T cell receptor-deficient T cell compositions
SG11201407802WA (en) 2012-05-25 2015-01-29 Cellectis Methods for engineering allogeneic and immunosuppressive resistant t cell for immunotherapy
US20150017136A1 (en) * 2013-07-15 2015-01-15 Cellectis Methods for engineering allogeneic and highly active t cell for immunotherapy
DK2981607T3 (en) * 2013-04-03 2020-11-16 Memorial Sloan Kettering Cancer Center EFFICIENT GENERATION OF TUMOR-TARGETED T-CELLS DERIVED FROM PLURIPOTENT STEM CELLS
CN105209054A (en) 2013-04-18 2015-12-30 阿尔莫生物科技股份有限公司 Methods of using interleukin-10 for treating diseases and disorders
US11311575B2 (en) * 2013-05-13 2022-04-26 Cellectis Methods for engineering highly active T cell for immunotherapy
US11077144B2 (en) 2013-05-13 2021-08-03 Cellectis CD19 specific chimeric antigen receptor and uses thereof
EP3434277A1 (en) 2013-06-17 2019-01-30 Armo Biosciences, Inc. Method for assessing protein identity and stability
AU2014311432A1 (en) 2013-08-30 2016-03-03 Armo Biosciences, Inc. Methods of using interleukin-10 for treating diseases and disorders
RU2016122957A (en) 2013-11-11 2017-12-19 Армо Байосайенсиз, Инк. Methods of using interleukin-10 for the treatment of diseases and disorders
WO2015187295A2 (en) 2014-06-02 2015-12-10 Armo Biosciences, Inc. Methods of lowering serum cholesterol
ES2861901T3 (en) * 2014-10-03 2021-10-06 Univ Oxford Innovation Ltd T-cell monotype analysis
EP3209320B1 (en) 2014-10-22 2023-03-08 Armo Biosciences, Inc. Methods of using interleukin-10 for treating diseases and disorders
CA2965509C (en) 2014-10-24 2023-03-14 Avectas Limited Delivery across cell plasma membranes
CN107708710A (en) 2015-03-17 2018-02-16 嵌合体生物工程公司 Smart CAR devices, DE CAR polypeptides, Side CAR and its use
KR20180020141A (en) 2015-05-28 2018-02-27 아르모 바이오사이언시스 인코포레이티드 PEGylated interleukin-10 for cancer treatment
CN108025040A (en) 2015-08-25 2018-05-11 阿尔莫生物科技股份有限公司 The method that disease and illness are treated using interleukin-10
KR20180049080A (en) * 2015-09-11 2018-05-10 더 보드 오브 트러스티스 오브 더 리랜드 스탠포드 쥬니어 유니버시티 Biologically related orthogonal cytokine / receptor pairs
MA44909A (en) 2015-09-15 2018-07-25 Acerta Pharma Bv THERAPEUTIC ASSOCIATION OF A CD19 INHIBITOR AND A BTK INHIBITOR
US11052111B2 (en) 2015-12-08 2021-07-06 Chimera Bioengineering, Inc. Smart CAR devices and DE CAR polypeptides for treating disease and methods for enhancing immune responses
JP7082051B2 (en) * 2016-01-11 2022-06-07 アルモ・バイオサイエンシーズ・インコーポレイテッド Interleukin-10 and its use in the production of antigen-specific CD8 + T cells
WO2017189705A1 (en) * 2016-04-26 2017-11-02 Temple University-Of The Commonwealth System Of Higher Education Dll4-expressing cells and vaccine using the same
WO2017192536A1 (en) 2016-05-02 2017-11-09 University Of Kansas Eliminating mhc restriction from the t cell receptor as a strategy for immunotherapy
US11214789B2 (en) 2016-05-03 2022-01-04 Flodesign Sonics, Inc. Concentration and washing of particles with acoustics
WO2017201019A1 (en) 2016-05-17 2017-11-23 Chimera Bioengineering, Inc. Methods for making novel antigen binding domains
CN110121352B (en) 2016-09-01 2020-12-11 嵌合体生物工程公司 GOLD-optimized CAR T-cells
WO2018132479A1 (en) * 2017-01-10 2018-07-19 The General Hospital Corporation Modified t cells and methods of their use
BR112019016672A8 (en) * 2017-02-13 2022-11-08 Hopitaux Paris Assist Publique METHOD TO GENERATE T CELL PROGENITORS
US11851649B2 (en) 2017-05-17 2023-12-26 Seattle Children's Hospital Generating mammalian T cell activation inducible synthetic promoters (SYN+PRO) to improve T cell therapy
JP7275054B2 (en) 2017-06-15 2023-05-17 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Targeted non-viral DNA insertion
KR20230034416A (en) 2017-10-27 2023-03-09 더 리전트 오브 더 유니버시티 오브 캘리포니아 Targeted replacement of endogenous t cell receptors
US11738047B2 (en) * 2017-12-12 2023-08-29 The Trustees Of The University Of Pennsylvania Genetically modified immune cells targeting NY-ESO-1 and methods of use thereof
WO2019160815A1 (en) 2018-02-13 2019-08-22 Chimera Bioengineering, Inc. Coordinating gene expression using rna destabilizing elements
WO2020018691A1 (en) * 2018-07-18 2020-01-23 The General Hospital Corporation Modified t cells and methods of their use
US20210277120A1 (en) * 2018-07-18 2021-09-09 The General Hospital Corporation Compositions and methods for treatment of t cell malignancies
EP4013431A4 (en) 2019-08-18 2024-05-01 Chimera Bioengineering Inc Combination therapy with gold controlled transgenes
CA3164233A1 (en) 2019-12-13 2021-06-17 Synthekine, Inc. Il-2 orthologs and methods of use
AU2021236145A1 (en) 2020-03-10 2022-09-22 Dana-Farber Cancer Institute, Inc. Methods for generating engineered memory-like NK cells and compositions thereof
MX2022011178A (en) 2020-03-10 2023-01-04 Massachusetts Inst Technology COMPOSITIONS AND METHODS FOR IMMUNOTHERAPY OF NPM1c-POSITIVE CANCER.
WO2021221782A1 (en) 2020-05-01 2021-11-04 Massachusetts Institute Of Technology Chimeric antigen receptor-targeting ligands and uses thereof
US20210340524A1 (en) 2020-05-01 2021-11-04 Massachusetts Institute Of Technology Methods for identifying chimeric antigen receptor-targeting ligands and uses thereof
TW202241935A (en) 2020-12-18 2022-11-01 美商世紀治療股份有限公司 Chimeric antigen receptor system with adaptable receptor specificity
WO2023274387A1 (en) * 2021-07-01 2023-01-05 宁波茂行生物医药科技有限公司 Universal car-t cell targeting gd2, preparation method therefor, and application thereof
CN115768445A (en) * 2021-07-01 2023-03-07 宁波茂行生物医药科技有限公司 IL13R alpha 2-targeted universal CAR-T cell and preparation method and application thereof
KR20240040770A (en) 2021-08-03 2024-03-28 제니시티 리미티드 Engineered TCR complexes and methods of using the same
EP4130028A1 (en) 2021-08-03 2023-02-08 Rhazes Therapeutics Ltd Engineered tcr complex and methods of using same
WO2023081715A1 (en) 2021-11-03 2023-05-11 Viracta Therapeutics, Inc. Combination of car t-cell therapy with btk inhibitors and methods of use thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011059836A2 (en) * 2009-10-29 2011-05-19 Trustees Of Dartmouth College T cell receptor-deficient t cell compositions

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU6869198A (en) * 1997-03-25 1998-10-20 Morphogenesis, Inc. Universal stem cells
US20030194704A1 (en) 2002-04-03 2003-10-16 Penn Sharron Gaynor Human genome-derived single exon nucleic acid probes useful for gene expression analysis two
WO2004067779A2 (en) 2003-01-30 2004-08-12 Applera Corporation Genetic polymorphisms associated with rheumatoid arthritis, methods of detection and uses thereof
US7888121B2 (en) 2003-08-08 2011-02-15 Sangamo Biosciences, Inc. Methods and compositions for targeted cleavage and recombination
KR101419729B1 (en) 2005-07-26 2014-07-17 상가모 바이오사이언스 인코포레이티드 Targeted integration and expression of exogenous nucleic acid sequences
US20070036773A1 (en) * 2005-08-09 2007-02-15 City Of Hope Generation and application of universal T cells for B-ALL
EP2027256A2 (en) 2006-05-31 2009-02-25 Projech Science to Technology, S.L. Animal models of tumour metastasis and toxicity
RU2355763C2 (en) 2006-09-13 2009-05-20 Закрытое акционерное общество "Научно-исследовательский институт Аджиномото-Генетика" (ЗАО АГРИ) Mutant acetolactase synthase and method of producing branched l-amino acids
WO2008108257A1 (en) 2007-03-05 2008-09-12 International Institute Of Cancer Immunology, Inc. Cancer antigen-specific t-cell receptor gene, peptide encoded by the gene, and use of them
WO2010048567A1 (en) 2008-10-24 2010-04-29 Wisconsin Alumni Research Foundation Pluripotent stem cells obtained by non-viral reprogramming
JP2010229050A (en) * 2009-03-26 2010-10-14 Shiseido Co Ltd Solubilized composition
AU2010279913B2 (en) 2009-08-07 2016-04-28 Kyoto University Method of efficiently establishing induced pluripotent stem cells
US8956828B2 (en) * 2009-11-10 2015-02-17 Sangamo Biosciences, Inc. Targeted disruption of T cell receptor genes using engineered zinc finger protein nucleases
US8945868B2 (en) 2010-07-21 2015-02-03 Sangamo Biosciences, Inc. Methods and compositions for modification of a HLA locus
WO2012149470A1 (en) * 2011-04-27 2012-11-01 Amyris, Inc. Methods for genomic modification
US9719066B2 (en) 2012-09-07 2017-08-01 Biolamina Ab Stem cell bank
CN103045591B (en) 2013-01-05 2014-03-12 上海荻硕贝肯生物科技有限公司 HLA gene specific PCR amplification primer, HLA typing method and kit
CA2904210C (en) 2013-03-12 2022-07-19 Sangamo Biosciences, Inc. Methods and compositions for modification of hla

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011059836A2 (en) * 2009-10-29 2011-05-19 Trustees Of Dartmouth College T cell receptor-deficient t cell compositions

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
EHLERS, STEFAN. ET AL.: "Alphabeta T Cell receptor-positive cells and interferon-gamma, but not inducible nitric oxide synthase, are critical for granuloma necrosis in a mouse model of mycobacteria-induced pulmonary immunopathology", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 194, no. 12, 17 December 2001 (2001-12-17), pages 1847 - 1859 *
GASCOIGNET, NICHOLAS R. J.: "Transport and secretion of truncated T cell receptor & chain occurs in the absence of association with CD3", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 265, no. 16, 5 June 1990 (1990-06-05), pages 9296 - 9301 *
TORIKAI, HIROKI. ET AL.: "A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric- antigen-receptor and eliminate expression of endogenous TCR", BLOOD, vol. 119, no. 24, 14 June 2012 (2012-06-14), pages 5697 - 5705 *

Cited By (308)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11299536B2 (en) 2012-02-22 2022-04-12 The Trustees Of The University Of Pennsylvania Compositions and methods for generating a persisting population of T cells useful for the treatment of cancer
US10800840B2 (en) 2012-02-22 2020-10-13 The Trustees Of The University Of Pennsylvania Compositions and methods for generating a persisting population of T cells useful for the treatment of cancer
US10040846B2 (en) 2012-02-22 2018-08-07 The Trustees Of The University Of Pennsylvania Compositions and methods for generating a persisting population of T cells useful for the treatment of cancer
US10308717B2 (en) 2013-02-20 2019-06-04 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
US11865167B2 (en) 2013-02-20 2024-01-09 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
US9394368B2 (en) 2013-02-20 2016-07-19 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
EP2970886A4 (en) * 2013-03-12 2016-08-17 Sangamo Biosciences Inc Methods and compositions for modification of hla
US9937207B2 (en) 2013-03-21 2018-04-10 Sangamo Therapeutics, Inc. Targeted disruption of T cell receptor genes using talens
EP2975942B1 (en) * 2013-03-21 2018-08-08 Sangamo Therapeutics, Inc. Targeted disruption of t cell receptor genes using engineered zinc finger protein nucleases
US10918668B2 (en) 2013-03-21 2021-02-16 Sangamo Therapeutics, Inc. Targeted disruption of T cell receptor genes using engineered zinc finger protein nucleases
US9629877B2 (en) 2013-05-14 2017-04-25 Board Of Regents, The University Of Texas System Human application of engineered chimeric antigen receptor (CAR) T-cells
US11786582B2 (en) 2013-05-14 2023-10-17 Board Of Regents, The University Of Texas System Human application of engineered chimeric antigen receptor (CAR) T-cells
US9701758B2 (en) 2013-05-24 2017-07-11 Board Of Regents, The University Of Texas System Anti-CD19 scFv (FMC63) polypeptide
US9855297B2 (en) 2013-05-29 2018-01-02 Cellectis Methods for engineering T cells for immunotherapy by using RNA-guided CAS nuclease system
US9890393B2 (en) 2013-05-29 2018-02-13 Cellectis Methods for engineering T cells for immunotherapy by using RNA-guided CAS nuclease system
WO2014191128A1 (en) * 2013-05-29 2014-12-04 Cellectis Methods for engineering t cells for immunotherapy by using rna-guided cas nuclease system
EP3004337B1 (en) 2013-05-29 2017-08-02 Cellectis Methods for engineering t cells for immunotherapy by using rna-guided cas nuclease system
AU2014273490B2 (en) * 2013-05-29 2019-05-09 Cellectis Methods for engineering T cells for immunotherapy by using RNA-guided Cas nuclease system
US10870864B2 (en) 2013-05-29 2020-12-22 Cellectis Methods for engineering T cells for immunotherapy by using RNA-guided Cas nuclease system
US11365430B2 (en) 2013-05-29 2022-06-21 Cellectis Methods for engineering T cells for immunotherapy by using RNA-guided Cas nuclease system
US10584352B2 (en) 2013-05-29 2020-03-10 Cellectis Methods for engineering T cells for immunotherapy by using RNA-guided Cas nuclease system
EP3309248A1 (en) * 2013-05-29 2018-04-18 Cellectis Methods for engineering t cells for immunotherapy by using rna-guided cas nuclease system
US11959091B2 (en) 2013-05-29 2024-04-16 Cellectis Methods for engineering T cells for immunotherapy by using RNA-guided Cas nuclease system
CN103409373A (en) * 2013-08-26 2013-11-27 上海易美生物技术有限公司 Preparation method for T cells modified by MDM2 (murine double minute 2) genetic recipient
CN105848484A (en) * 2013-10-25 2016-08-10 得克萨斯州大学系统董事会 Polyclonal [Gamma] [delta] T cells for immunotherapy
US9907820B2 (en) 2013-10-25 2018-03-06 Board Of Regents, The University Of Texas System Polyclonal gamma delta T cells for immunotherapy
EP3060059A4 (en) * 2013-10-25 2017-11-01 Board of Regents, The University of Texas System Polyclonal gamma delta t cells for immunotherapy
JP2016539117A (en) * 2013-11-22 2016-12-15 セレクティスCellectis Method for generating a batch of allogeneic T cells with averaged potency
AU2014351871B2 (en) * 2013-11-22 2020-02-13 Cellectis Method for generating batches of allogeneic T-cells with averaged potency
WO2015075175A1 (en) * 2013-11-22 2015-05-28 Cellectis Method for generating batches of allogeneic t-cells with averaged potency
US10357515B2 (en) 2013-11-22 2019-07-23 Cellectis Method for generating batches of allogeneic T-cells with averaged potency
US11725061B2 (en) 2013-11-27 2023-08-15 The General Hospital Corporation CSGP4—specific chimeric antigen receptor for cancer
WO2015090230A1 (en) * 2013-12-19 2015-06-25 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
JP2021035948A (en) * 2013-12-19 2021-03-04 ノバルティス アーゲー Human mesothelin chimeric antigen receptor and use thereof
US10640569B2 (en) 2013-12-19 2020-05-05 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
JP2017500869A (en) * 2013-12-19 2017-01-12 ノバルティス アーゲー Human mesothelin chimeric antigen receptor and use thereof
CN106459989A (en) * 2013-12-19 2017-02-22 诺华股份有限公司 Human mesothelin chimeric antigen receptors and uses thereof
CN106459989B (en) * 2013-12-19 2023-02-17 诺华股份有限公司 Human mesothelin chimeric antigen receptor and uses thereof
CN113234757A (en) * 2014-02-14 2021-08-10 得克萨斯州大学系统董事会 Chimeric antigen receptor and method of preparation
WO2015121454A1 (en) * 2014-02-14 2015-08-20 Cellectis Cells for immunotherapy engineered for targeting antigen present both on immune cells and pathological cells
EP3505623A1 (en) * 2014-02-14 2019-07-03 Cellectis Cells for immunotherapy engineered for targeting antigen present both on immune cells and pathological cells
US10125193B2 (en) 2014-02-14 2018-11-13 Board Of Regents, The University Of Texas System Chimeric antigen receptors and methods of making
EP3105317B1 (en) 2014-02-14 2018-09-19 Cellectis Cells for immunotherapy engineered for targeting antigen present both on immune cells and pathological cells
US11692169B2 (en) 2014-02-14 2023-07-04 Cellectis Cells for immunotherapy engineered for targeting antigen present both on immune cells and pathological cells
US10836998B2 (en) 2014-02-14 2020-11-17 Cellectis Cells for immunotherapy engineered for targeting antigen present both on immune cells and pathological cells
US10532088B2 (en) 2014-02-27 2020-01-14 Lycera Corporation Adoptive cellular therapy using an agonist of retinoic acid receptor-related orphan receptor gamma and related therapeutic methods
EP3241561A1 (en) * 2014-03-05 2017-11-08 Ucl Business Plc Chimeric antigen receptor (car) with antigen binding domains to the t cell receptor beta constant region
KR102235202B1 (en) 2014-03-05 2021-04-05 유씨엘 비즈니스 리미티드 Chimeric antigen receptor(car) with antigen binding domains to the t cell receptor beta constant region
RU2744046C2 (en) * 2014-03-05 2021-03-02 Отолус Лимитед CHIMERIC ANTIGEN RECEPTOR (CAR) WITH ANTIGEN-BINDING DOMAINS TO THE T-CELL RECEPTOR β- CONSTANT REGION
KR102235201B1 (en) * 2014-03-05 2021-04-05 유씨엘 비즈니스 리미티드 Chimeric antigen receptor(car) with antigen binding domains to the t cell receptor beta constant region
WO2015132598A1 (en) * 2014-03-05 2015-09-11 Ucl Business Plc Chimeric antigen receptor (car) with antigen binding domains to the t cell receptor beta constant region
CN106068276B (en) * 2014-03-05 2020-09-18 Ucl商务有限公司 Chimeric Antigen Receptor (CAR) with antigen binding domain to T cell receptor beta constant region
CN106068276A (en) * 2014-03-05 2016-11-02 Ucl商务股份有限公司 There is the Chimeric antigen receptor (CAR) of the antigen binding domain for φt cell receptor beta constant region
US11885806B2 (en) 2014-03-05 2024-01-30 Autolus Limited Method for depleting malignant T-cells
US11885807B2 (en) 2014-03-05 2024-01-30 Autolus Limited Method for depleting malignant T-cells
KR102088082B1 (en) * 2014-03-05 2020-03-11 유씨엘 비즈니스 리미티드 Chimeric antigen receptor(car) with antigen binding domains to the t cell receptor beta constant region
KR20160127130A (en) * 2014-03-05 2016-11-02 유씨엘 비즈니스 피엘씨 Chimeric antigen receptor(car) with antigen binding domains to the t cell receptor beta constant region
KR20200027577A (en) * 2014-03-05 2020-03-12 유씨엘 비즈니스 리미티드 Chimeric antigen receptor(car) with antigen binding domains to the t cell receptor beta constant region
KR20200027576A (en) * 2014-03-05 2020-03-12 유씨엘 비즈니스 리미티드 Chimeric antigen receptor(car) with antigen binding domains to the t cell receptor beta constant region
US11820996B2 (en) 2014-03-11 2023-11-21 Cellectis Method for generating T-cells compatible for allogenic transplantation
EP3116902B1 (en) 2014-03-11 2020-01-01 Cellectis Method for generating t-cells compatible for allogenic transplantation
EP3693384B1 (en) * 2014-03-11 2024-01-24 Cellectis Method for generating t-cells compatible for allogenic transplantation
EP3693384A1 (en) 2014-03-11 2020-08-12 Cellectis Method for generating t-cells compatible for allogenic transplantation
WO2015142675A2 (en) 2014-03-15 2015-09-24 Novartis Ag Treatment of cancer using chimeric antigen receptor
JP7106610B2 (en) 2014-04-10 2022-07-26 シアトル チルドレンズ ホスピタル (ディービーエイ シアトル チルドレンズ リサーチ インスティテュート) Gene tags for transgenes and methods of their use
US11414486B2 (en) 2014-04-10 2022-08-16 Seattle Children's Hospital Transgene genetic tags and methods of use
US10611837B2 (en) 2014-04-10 2020-04-07 Seattle Children's Hospital Transgene genetic tags and methods of use
US11155616B2 (en) 2014-04-10 2021-10-26 Seattle Children's Hospital Drug regulated transgene expression
US10865242B2 (en) 2014-04-10 2020-12-15 Seattle Children's Hospital Method and compositions for cellular immunotherapy
JP2021036867A (en) * 2014-04-10 2021-03-11 シアトル チルドレンズ ホスピタル, ディービーエー シアトル チルドレンズ リサーチ インスティテュート Transgene genetic tags and methods of using the same
US10266592B2 (en) 2014-04-10 2019-04-23 Seattle Children's Hospital Drug regulated transgene expression
JP2017518037A (en) * 2014-04-10 2017-07-06 シアトル チルドレンズ ホスピタル, ディービーエー シアトル チルドレンズ リサーチ インスティテュート Gene tags for transgenes and methods of use
WO2015157384A1 (en) * 2014-04-10 2015-10-15 Seattle Children's Hospital (dba Seattle Children's Research Institute) Defined composition gene modified t-cell products
JP2022153456A (en) * 2014-04-10 2022-10-12 シアトル チルドレンズ ホスピタル (ディービーエイ シアトル チルドレンズ リサーチ インスティテュート) Transgene genetic tags and method of use the same
JP7402933B2 (en) 2014-04-10 2023-12-21 シアトル チルドレンズ ホスピタル (ディービーエイ シアトル チルドレンズ リサーチ インスティテュート) Genetic tags for transgenes and their use
US10442798B2 (en) 2014-05-05 2019-10-15 Lycera Corporation Tetrahydroquinoline sulfonamide and related compounds for use as agonists of RORγ and the treatment of disease
US10189777B2 (en) 2014-05-05 2019-01-29 Lycera Corporation Benzenesulfonamido and related compounds for use as agonists of RORγ and the treatment of disease
US9896441B2 (en) 2014-05-05 2018-02-20 Lycera Corporation Tetrahydroquinoline sulfonamide and related compounds for use as agonists of RORγ and the treatment of disease
US10364237B2 (en) 2014-05-05 2019-07-30 Lycera Corporation Tetrahydroquinoline sulfonamide and related compounds for use as agonists of RORγ and the treatment of disease
US11041021B2 (en) * 2014-05-23 2021-06-22 University Of Florida Research Foundation, Incorporated Car based immunotherapy
RU2739770C2 (en) * 2014-06-11 2020-12-28 полибайосепт ГмбХ Expansion of lymphocyte with a cytokine composition for active cellular immunotherapy
US11084880B2 (en) 2014-07-21 2021-08-10 Novartis Ag Anti-BCMA chimeric antigen receptor
US10174095B2 (en) 2014-07-21 2019-01-08 Novartis Ag Nucleic acid encoding a humanized anti-BCMA chimeric antigen receptor
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
US10568947B2 (en) 2014-07-21 2020-02-25 Novartis Ag Treatment of cancer using a CLL-1 chimeric antigen receptor
WO2016030414A1 (en) * 2014-08-29 2016-03-03 Gemoab Monoclonals Gmbh Universal chimeric antigen receptor expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
EP2990416A1 (en) * 2014-08-29 2016-03-02 GEMoaB Monoclonals GmbH Universal chimeric antigen receptor expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
AU2015308499B2 (en) * 2014-08-29 2020-01-02 Avencell Europe Gmbh Universal chimeric antigen receptor expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
US10766943B2 (en) 2014-08-29 2020-09-08 Gemoab Monoclonals Gmbh Universal chimeric antigen expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
US10611814B2 (en) 2014-08-29 2020-04-07 Gemoab Monoclonals Gmbh Universal chimeric antigen expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
WO2016044605A1 (en) 2014-09-17 2016-03-24 Beatty, Gregory Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
EP3967709A1 (en) 2014-09-17 2022-03-16 Novartis AG Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US10577417B2 (en) 2014-09-17 2020-03-03 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
JP2017535261A (en) * 2014-10-31 2017-11-30 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Modification of gene expression in CART cells and use thereof
EP3215168A4 (en) * 2014-10-31 2018-08-15 The Trustees of the University of Pennsylvania Altering gene expression in modified t cells and uses thereof
WO2016069283A1 (en) 2014-10-31 2016-05-06 The Trustees Of The University Of Pennsylvania Altering gene expression in cart cells and uses thereof
US11203758B2 (en) 2014-10-31 2021-12-21 The Trustees Of The University Of Pennsylvania Altering gene expression in modified T cells and uses thereof
JP2018500006A (en) * 2014-10-31 2018-01-11 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Modification of gene expression in modified T cells and use thereof
US11208661B2 (en) 2014-10-31 2021-12-28 The Trustees Of The University Of Pennsylvania Altering gene expression in modified T cells and uses thereof
CN107206024A (en) * 2014-10-31 2017-09-26 宾夕法尼亚大学董事会 Gene expression in change CART cells and application thereof
JP2021058196A (en) * 2014-10-31 2021-04-15 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Altering gene expression in cart cells and uses thereof
CN114836385A (en) * 2014-10-31 2022-08-02 宾夕法尼亚大学董事会 Altering gene expression in CART cells and uses thereof
CN107249606A (en) * 2014-10-31 2017-10-13 宾夕法尼亚大学董事会 Gene expression in the T cell of change modification and application thereof
AU2015339744B2 (en) * 2014-10-31 2021-03-25 The Trustees Of The University Of Pennsylvania Altering gene expression in CART cells and uses thereof
US10570186B2 (en) 2014-11-05 2020-02-25 Board Of Regents, The University Of Texas System Chimeric antigen receptors (CAR) to selectively target protein complexes
USRE49435E1 (en) 2014-11-12 2023-02-28 Seagen Inc. Glycan-interacting compounds and methods of use
WO2016077526A1 (en) 2014-11-12 2016-05-19 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
EP4183806A2 (en) 2014-11-12 2023-05-24 Seagen Inc. Glycan-interacting compounds and methods of use
US9879087B2 (en) 2014-11-12 2018-01-30 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
RU2756247C2 (en) * 2014-11-17 2021-09-28 Эдисет Био, Инк. Genetically modified gamma delta t cells
WO2016081518A3 (en) * 2014-11-17 2016-08-18 Adicet Bio, Inc. Engineered gamma delta t-cells
US11135245B2 (en) 2014-11-17 2021-10-05 Adicet Bio, Inc. Engineered γδ T-cells
WO2016083811A1 (en) * 2014-11-27 2016-06-02 Imperial Innovations Limited Genome editing methods
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
US10273300B2 (en) 2014-12-29 2019-04-30 The Trustees Of The University Of Pennsylvania Methods of making chimeric antigen receptor-expressing cells
CN107438618A (en) * 2015-01-26 2017-12-05 塞勒克提斯公司 Assign the engineering immunocyte that the Chimeric antigen receptor for being bound to CD123 knocks out for treating the φt cell receptor of recurrent/intractable acute myeloid lymthoma or mother cell plasmacytoid dendritic cellss tumour
US11014989B2 (en) 2015-01-26 2021-05-25 Cellectis Anti-CLL1 specific single-chain chimeric antigen receptors (scCARs) for cancer immunotherapy
WO2016120216A1 (en) 2015-01-26 2016-08-04 Cellectis mAb-DRIVEN CHIMERIC ANTIGEN RECEPTOR SYSTEMS FOR SORTING/DEPLETING ENGINEERED IMMUNE CELLS
US11634688B2 (en) 2015-01-26 2023-04-25 Fate Therapeutics, Inc. Methods and compositions for inducing hematopoietic cell differentiation
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
US11248058B2 (en) 2015-02-19 2022-02-15 University Of Florida Research Foundation, Inc. Chimeric antigen receptors and uses thereof
US10808230B2 (en) 2015-02-24 2020-10-20 Board Of Regents, The University Of Texas System Selection methods for genetically-modified T cells
US11385233B2 (en) 2015-03-05 2022-07-12 Autolus Limited Methods of depleting malignant T-cells
US20200200756A1 (en) * 2015-03-05 2020-06-25 Ucl Business Plc. Methods
US10883094B2 (en) 2015-03-11 2021-01-05 Board Of Regents, The University Of Texas System Transposase polypeptides and uses thereof
US10570382B2 (en) 2015-03-11 2020-02-25 Board Of Regents, The University Of Texas System Transposase polypeptides and uses thereof
US11492604B2 (en) 2015-03-11 2022-11-08 Board Of Regents, The University Of Texas System Transposase polypeptides and uses thereof
US11896614B2 (en) 2015-04-17 2024-02-13 Novartis Ag Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
US10421751B2 (en) 2015-05-05 2019-09-24 Lycera Corporation Dihydro-2H-benzo[b][1,4]oxazine sulfonamide and related compounds for use as agonists of RORγ and the treatment of disease
US10611740B2 (en) 2015-06-11 2020-04-07 Lycera Corporation Aryl dihydro-2H-benzo[b][1,4]oxazine sulfonamide and related compounds for use as agonists of RORγ and the treatment of disease
US11059796B2 (en) 2015-06-11 2021-07-13 The Regents Of The University Of Michigan Aryl dihydro-2H benzo[b][1,4]oxazine sulfonamide and related compounds for use as agonists of RORγ and the treatment of disease
US20170137783A1 (en) * 2015-07-21 2017-05-18 Felipe Bedoya Methods for improving the efficacy and expansion of immune cells
US10829735B2 (en) * 2015-07-21 2020-11-10 The Trustees Of The University Of Pennsylvania Methods for improving the efficacy and expansion of immune cells
US11147837B2 (en) 2015-07-31 2021-10-19 Regents Of The University Of Minnesota Modified cells and methods of therapy
US11266692B2 (en) 2015-07-31 2022-03-08 Regents Of The University Of Minnesota Intracellular genomic transplant and methods of therapy
US11925664B2 (en) 2015-07-31 2024-03-12 Intima Bioscience, Inc. Intracellular genomic transplant and methods of therapy
US11642375B2 (en) 2015-07-31 2023-05-09 Intima Bioscience, Inc. Intracellular genomic transplant and methods of therapy
US10166255B2 (en) 2015-07-31 2019-01-01 Regents Of The University Of Minnesota Intracellular genomic transplant and methods of therapy
US11642374B2 (en) 2015-07-31 2023-05-09 Intima Bioscience, Inc. Intracellular genomic transplant and methods of therapy
US11583556B2 (en) 2015-07-31 2023-02-21 Regents Of The University Of Minnesota Modified cells and methods of therapy
US11903966B2 (en) 2015-07-31 2024-02-20 Regents Of The University Of Minnesota Intracellular genomic transplant and methods of therapy
US10406177B2 (en) 2015-07-31 2019-09-10 Regents Of The University Of Minnesota Modified cells and methods of therapy
US11458167B2 (en) 2015-08-07 2022-10-04 Seattle Children's Hospital Bispecific CAR T-cells for solid tumor targeting
WO2017027392A1 (en) 2015-08-07 2017-02-16 Novartis Ag Treatment of cancer using chimeric cd3 receptor proteins
US11123369B2 (en) 2015-08-07 2021-09-21 Seattle Children's Hospital Bispecific CAR T-cells for solid tumor targeting
US10709775B2 (en) 2015-08-11 2020-07-14 Cellectis Cells for immunotherapy engineered for targeting CD38 antigen and for CD38 gene inactivation
US11898165B2 (en) * 2015-09-11 2024-02-13 Mink Therapeutics, Inc. Engineered host cells and methods of use thereof
US20200263133A1 (en) * 2015-09-11 2020-08-20 Agenus Inc. Engineered host cells and methods of use thereof
US11548945B2 (en) 2015-09-15 2023-01-10 Board Of Regents, The University Of Texas System T-cell receptor (TCR)-binding antibodies and uses thereof
US10633445B2 (en) 2015-09-15 2020-04-28 Board Of Regents The University Of Texas System T-cell receptor (TCR)-binding antibodies and uses thereof
US9969975B1 (en) 2015-10-05 2018-05-15 Precision Biosciences, Inc. Genetically-modified cells comprising a modified human T cell receptor alpha constant region gene
US10093899B1 (en) 2015-10-05 2018-10-09 Precision Biosciences, Inc. Genetically-modified cells comprising a modified human T cell receptor alpha constant region gene
US10093900B2 (en) 2015-10-05 2018-10-09 Precision Biosciences, Inc. Genetically-modified cells comprising a modified human T cell receptor alpha constant region gene
US9889161B2 (en) 2015-10-05 2018-02-13 Precision Biosciences, Inc. Genetically-modified cells comprising a modified human T cell receptor alpha constant region gene
JP2018536390A (en) * 2015-10-05 2018-12-13 プレシジョン バイオサイエンシズ,インク. Meganucleases engineered to recognition sequences found in the human T cell receptor alpha constant region gene
US9950011B1 (en) 2015-10-05 2018-04-24 Precision Biosciences, Inc. Genetically-modified cells comprising a modified human T cell receptor alpha constant region gene
US11268065B2 (en) 2015-10-05 2022-03-08 Precision Biosciences, Inc. Genetically-modified cells comprising a modified human T cell receptor alpha constant region gene
US9950010B1 (en) 2015-10-05 2018-04-24 Precision Biosciences, Inc. Genetically-modified cells comprising a modified human T cell receptor alpha constant region gene
US10799535B2 (en) 2015-10-05 2020-10-13 Precision Biosciences, Inc. Engineered meganucleases with recognition sequences found in the human T cell receptor alpha constant region gene
US9993502B1 (en) 2015-10-05 2018-06-12 Precision Biosciences, Inc. Genetically-modified cells comprising a modified human T cell receptor alpha constant region gene
US11266693B2 (en) 2015-10-05 2022-03-08 Precision Biosciences, Inc. Nucleic acids encoding engineered meganucleases with recognition sequences found in the human T cell receptor alpha constant region gene
US9889160B2 (en) 2015-10-05 2018-02-13 Precision Biosciences, Inc. Genetically-modified cells comprising a modified human T cell receptor alpha constant region gene
US9993501B2 (en) 2015-10-05 2018-06-12 Precision Biosciences, Inc. Genetically-modified cells comprising a modified human T cell receptor alpha constant region gene
US11242377B2 (en) 2015-10-23 2022-02-08 Autolus Limited Chimeric receptor with ligand binding exodomain and cytokine and T cell signalling endodomains
US11059879B2 (en) 2015-10-27 2021-07-13 Board Of Regents, The University Of Texas System Chimeric antigen receptor molecules and uses thereof
US10947505B2 (en) 2015-11-04 2021-03-16 Fate Therapeutics, Inc. Methods and compositions for inducing hematopoietic cell differentiation
US11162076B2 (en) 2015-11-04 2021-11-02 Fate Therapeutics, Inc. Methods and compositions for inducing hematopoietic cell differentiation
JP2021191272A (en) * 2015-11-04 2021-12-16 フェイト セラピューティクス,インコーポレイテッド Genome modification of universality cell
JP2018531612A (en) * 2015-11-04 2018-11-01 フェイト セラピューティクス,インコーポレイテッド Genomic modification of universal cells
US11352607B2 (en) 2015-11-04 2022-06-07 Fate Therapeutics, Inc. Genomic engineering of pluripotent cells
US11162075B2 (en) 2015-11-04 2021-11-02 Fate Therapeutics, Inc. Methods and compositions for inducing hematopoietic cell differentiation
US11072781B2 (en) 2015-11-04 2021-07-27 Fate Therapeutics, Inc. Genomic engineering of pluripotent cells
WO2017083582A1 (en) 2015-11-12 2017-05-18 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
US11028181B2 (en) 2015-11-12 2021-06-08 Seagen Inc. Glycan-interacting compounds and methods of use
WO2017091786A1 (en) 2015-11-23 2017-06-01 Novartis Ag Optimized lentiviral transfer vectors and uses thereof
US11413340B2 (en) 2015-12-22 2022-08-16 Novartis Ag Mesothelin chimeric antigen receptor (CAR) and antibody against PD-L1 inhibitor for combined use in anticancer therapy
WO2017114497A1 (en) 2015-12-30 2017-07-06 Novartis Ag Immune effector cell therapies with enhanced efficacy
EP4219689A2 (en) 2015-12-30 2023-08-02 Novartis AG Immune effector cell therapies with enhanced efficacy
CN109312315A (en) * 2015-12-30 2019-02-05 阿维塔斯有限公司 Gene editing albumen and composition are delivered to the carrier-free of cell and tissue
WO2017149515A1 (en) 2016-03-04 2017-09-08 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
WO2017165683A1 (en) 2016-03-23 2017-09-28 Novartis Ag Cell secreted minibodies and uses thereof
EP4219721A2 (en) 2016-04-15 2023-08-02 Novartis AG Compositions and methods for selective protein expression
WO2017181119A2 (en) 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective protein expression
US11634498B2 (en) 2016-04-29 2023-04-25 University Of Florida Research Foundation, Incorporated Chimeric antigen receptors and uses thereof
US11299708B2 (en) 2016-05-12 2022-04-12 Adicet Bio, Inc. Methods for selective expansion of γδ T-cell populations and compositions thereof
US11466291B2 (en) 2016-07-06 2022-10-11 Cellectis Sequential gene editing in primary immune cells
WO2018007263A1 (en) * 2016-07-06 2018-01-11 Cellectis Sequential gene editing in primary immune cells
US11674155B2 (en) 2016-07-06 2023-06-13 Cellectis Sequential gene editing in primary immune cells
AU2017291851B2 (en) * 2016-07-06 2022-10-13 Cellectis Sequential gene editing in primary immune cells
US10730942B2 (en) 2016-07-25 2020-08-04 Ucl Business Ltd Protein-based T-cell receptor knockdown
WO2018026819A2 (en) 2016-08-01 2018-02-08 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
CN106222201A (en) * 2016-08-27 2016-12-14 北京艺妙神州医疗科技有限公司 A kind of method preparing CAR T cell and prepared CAR T cell and application thereof
US10538574B2 (en) 2016-09-23 2020-01-21 Fred Hutchinson Cancer Research Center TCRS specific for minor histocompatibility (H) antigen HA-1 and uses thereof
WO2018058002A1 (en) 2016-09-23 2018-03-29 Fred Hutchinson Cancer Research Center Tcrs specific for minor histocompatibility (h) antigen ha-1 and uses thereof
EP3519441A4 (en) * 2016-09-27 2020-05-27 Cero Therapeutics, Inc. Chimeric engulfment receptor molecules
WO2018064076A1 (en) 2016-09-27 2018-04-05 Cero Therapeutics, Inc. Chimeric engulfment receptor molecules
EP4089116A1 (en) 2016-09-27 2022-11-16 Cero Therapeutics, Inc. Chimeric engulfment receptor molecules
US11655282B2 (en) 2016-09-27 2023-05-23 Cero Therapeutics, Inc. Chimeric engulfment receptor molecules
US10912797B2 (en) 2016-10-18 2021-02-09 Intima Bioscience, Inc. Tumor infiltrating lymphocytes and methods of therapy
US11154574B2 (en) 2016-10-18 2021-10-26 Regents Of The University Of Minnesota Tumor infiltrating lymphocytes and methods of therapy
US11401330B2 (en) 2016-11-17 2022-08-02 Seagen Inc. Glycan-interacting compounds and methods of use
WO2018111340A1 (en) 2016-12-16 2018-06-21 Novartis Ag Methods for determining potency and proliferative function of chimeric antigen receptor (car)-t cells
EP4043485A1 (en) 2017-01-26 2022-08-17 Novartis AG Cd28 compositions and methods for chimeric antigen receptor therapy
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
WO2018144535A1 (en) 2017-01-31 2018-08-09 Novartis Ag Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
WO2018160731A1 (en) 2017-02-28 2018-09-07 Novartis Ag Shp inhibitor compositions and uses for chimeric antigen receptor therapy
US11253609B2 (en) 2017-03-03 2022-02-22 Seagen Inc. Glycan-interacting compounds and methods of use
US11034748B2 (en) 2017-03-15 2021-06-15 Fred Hutchinson Cancer Research Center High affinity MAGE-A1-specific TCRs and uses thereof
US11851659B2 (en) 2017-03-22 2023-12-26 Novartis Ag Compositions and methods for immunooncology
US10857184B2 (en) 2017-05-12 2020-12-08 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US10729725B2 (en) 2017-05-12 2020-08-04 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US11071755B1 (en) 2017-05-12 2021-07-27 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US11202802B2 (en) 2017-05-12 2021-12-21 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US11135247B2 (en) 2017-05-12 2021-10-05 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US11207351B2 (en) 2017-05-12 2021-12-28 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US11191783B2 (en) 2017-05-12 2021-12-07 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US11471491B1 (en) 2017-05-12 2022-10-18 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US10881689B2 (en) 2017-05-12 2021-01-05 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US11622977B2 (en) 2017-05-12 2023-04-11 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US11298378B2 (en) 2017-05-12 2022-04-12 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US11166985B2 (en) 2017-05-12 2021-11-09 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US10736919B2 (en) 2017-05-12 2020-08-11 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US11013767B2 (en) 2017-05-12 2021-05-25 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
WO2018218038A1 (en) 2017-05-24 2018-11-29 Effector Therapeutics, Inc. Methods and compositions for cellular immunotherapy
US11564947B2 (en) 2017-05-24 2023-01-31 Effector Therapeutics Inc. Methods and compositions for cellular immunotherapy
US10780119B2 (en) 2017-05-24 2020-09-22 Effector Therapeutics Inc. Methods and compositions for cellular immunotherapy
US11566223B2 (en) 2017-06-01 2023-01-31 Innovative Cellular Therapeutics Holdings, Ltd. Chimeric antigen receptor cell preparation and uses thereof
WO2018219278A1 (en) 2017-06-01 2018-12-06 Innovative Cellular Therapeutics CO., LTD. Chimeric antigen receptor cell preparation and uses thereof
EP3630980A4 (en) * 2017-06-01 2021-03-10 Innovative Cellular Therapeutics Co., Ltd. Chimeric antigen receptor cell preparation and uses thereof
US11434293B2 (en) 2017-06-09 2022-09-06 Autolus Limited Anti TRBC1 antigen binding domains
US11440961B2 (en) 2017-06-09 2022-09-13 Autolus Limited Anti TRBC1 antigen binding domains
RU2791327C2 (en) * 2017-06-09 2023-03-07 Отолус Лимитед Anti-trbc1 antigen-binding domain
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
US11235004B2 (en) 2017-06-30 2022-02-01 Innovative Cellular Therapeutics Holdings, Ltd. Lymphocyte cell lines and uses thereof
WO2019002633A1 (en) 2017-06-30 2019-01-03 Cellectis Cellular immunotherapy for repetitive administration
US11053484B2 (en) 2017-06-30 2021-07-06 Precision Biosciences, Inc. Genetically-modified T cells comprising a modified intron in the T cell receptor alpha gene
US11098325B2 (en) 2017-06-30 2021-08-24 Intima Bioscience, Inc. Adeno-associated viral vectors for gene therapy
EP3680328A4 (en) * 2017-09-06 2020-11-25 Gracell Biotechnologies (Shanghai) Co., Ltd. Universal chimeric antigen receptor t-cell preparation technique
WO2019047899A1 (en) * 2017-09-06 2019-03-14 亘喜生物科技(上海)有限公司 Universal chimeric antigen receptor t-cell preparation technique
CN111051502A (en) * 2017-09-06 2020-04-21 亘喜生物科技(上海)有限公司 Preparation technology of universal chimeric antigen receptor T cell
CN111051502B (en) * 2017-09-06 2022-08-02 亘喜生物科技(上海)有限公司 Preparation technology of universal chimeric antigen receptor T cell
EP3680338A4 (en) * 2017-09-08 2021-06-02 Carsgen Therapeutics Co., Ltd. Genetically engineered t cell and application thereof
WO2019060425A1 (en) 2017-09-19 2019-03-28 Massachusetts Institute Of Technology Compositions for chimeric antigen receptor t cell therapy and uses thereof
WO2019067328A1 (en) 2017-09-26 2019-04-04 Cero Therapeutics, Inc. Chimeric engulfment receptor molecules and methods of use
US11708423B2 (en) 2017-09-26 2023-07-25 Cero Therapeutics, Inc. Chimeric engulfment receptor molecules and methods of use
US11952408B2 (en) 2017-10-03 2024-04-09 Juno Therapeutics, Inc. HPV-specific binding molecules
WO2019079569A1 (en) 2017-10-18 2019-04-25 Novartis Ag Compositions and methods for selective protein degradation
EP3699268A4 (en) * 2017-10-20 2021-08-04 Chongqing Precision Biotech Company Limited Universal car-t cell, preparation method therefor and application thereof
WO2019084288A1 (en) 2017-10-25 2019-05-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2019081983A1 (en) 2017-10-25 2019-05-02 Novartis Ag Antibodies targeting cd32b and methods of use thereof
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
US20210177953A1 (en) * 2017-11-14 2021-06-17 Asclepius Therapy Llc Engineered Non-Human Derived Immune Cells for Universal Adoptive Antigen Cellular Immunotherapy
US11903967B2 (en) 2018-01-09 2024-02-20 Autolus Limited Method of preparing T cells with increased activity
WO2019191339A1 (en) 2018-03-28 2019-10-03 Cero Therapeutics, Inc. Expression vectors for chimeric engulfment receptors, genetically modified host cells, and uses thereof
WO2019191334A1 (en) 2018-03-28 2019-10-03 Cero Therapeutics, Inc. Chimeric tim4 receptors and uses thereof
WO2019191332A1 (en) 2018-03-28 2019-10-03 Cero Therapeutics, Inc. Chimeric engulfment receptors and uses thereof for neurodegenerative diseases
US11786554B2 (en) 2018-04-12 2023-10-17 Precision Biosciences, Inc. Optimized engineered nucleases having specificity for the human T cell receptor alpha constant region gene
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
WO2019213282A1 (en) 2018-05-01 2019-11-07 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
US11254912B2 (en) 2018-05-11 2022-02-22 Crispr Therapeutics Ag Methods and compositions for treating cancer
US11649438B2 (en) 2018-05-11 2023-05-16 Crispr Therapeutics Ag Methods and compositions for treating cancer
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
WO2019237035A1 (en) 2018-06-08 2019-12-12 Intellia Therapeutics, Inc. Compositions and methods for immunooncology
US11939389B2 (en) 2018-06-13 2024-03-26 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11952428B2 (en) 2018-06-13 2024-04-09 Novartis Ag BCMA chimeric antigen receptors and uses thereof
WO2020012337A1 (en) 2018-07-10 2020-01-16 Novartis Ag 3-(5-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of i karos family zinc finger 2 (ikzf2)-dependent diseases
WO2020068261A1 (en) 2018-09-28 2020-04-02 Massachusetts Institute Of Technology Collagen-localized immunomodulatory molecules and methods thereof
WO2020128972A1 (en) 2018-12-20 2020-06-25 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165834A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020191378A1 (en) * 2019-03-21 2020-09-24 Allogene Therapeutics, Inc. METHODS FOR ENHANCING TCRαβ+ CELL DEPLETION EFFICIENCY
US11453861B2 (en) 2019-04-03 2022-09-27 Precision Biosciences, Inc. Genetically-modified immune cells comprising a microRNA-adapted shRNA (shRNAmiR)
US11008548B2 (en) 2019-04-03 2021-05-18 Precision Biosciences, Inc. Genetically-modified immune cells comprising a microRNA-adapted shRNA (shRNAmiR)
US11851680B2 (en) 2019-04-03 2023-12-26 Precision Biosciences, Inc. Genetically-modified immune cells comprising a microRNA-adapted shRNA (shRNAmiR)
US11186822B2 (en) 2019-04-03 2021-11-30 Precision Biosciences, Inc. Genetically-modified immune cells comprising a microRNA-adapted shRNA (shRNAmiR)
US11384335B2 (en) 2019-04-03 2022-07-12 Precision Biosciences, Inc. Genetically-modified immune cells comprising a microRNA-adapted shRNA (shRNAmiR)
WO2020219742A1 (en) 2019-04-24 2020-10-29 Novartis Ag Compositions and methods for selective protein degradation
US11389481B2 (en) 2019-04-30 2022-07-19 Crispr Therapeutics Ag Allogeneic cell therapy of B cell malignancies using genetically engineered T cells targeting CD19
WO2020263399A1 (en) 2019-06-26 2020-12-30 Massachusetts Institute Of Technology Immunomodulatory fusion protein-metal hydroxide complexes and methods thereof
WO2021061648A1 (en) 2019-09-23 2021-04-01 Massachusetts Institute Of Technology Methods and compositions for stimulation of endogenous t cell responses
WO2021067875A1 (en) 2019-10-03 2021-04-08 Cero Therapeutics, Inc. Chimeric tim4 receptors and uses thereof
WO2021106832A1 (en) * 2019-11-25 2021-06-03 国立大学法人京都大学 T-cell master cell bank
WO2021113759A1 (en) * 2019-12-04 2021-06-10 Celularity Inc. Placenta-derived allogeneic car-t cells and uses thereof
CN115397442A (en) * 2019-12-04 2022-11-25 细胞结构公司 Placenta-derived allogeneic CAR-T cells and uses thereof
WO2021123996A1 (en) 2019-12-20 2021-06-24 Novartis Ag Uses of anti-tgf-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
EP4086342A1 (en) * 2020-01-02 2022-11-09 Ningbo T-MAXIMUM Biopharmaceuticals Co., Ltd. Modified immune effector cell and preparation method therefor
WO2021136263A1 (en) * 2020-01-02 2021-07-08 江苏茂行科技有限公司 Modified immune effector cell and preparation method therefor
CN115003802A (en) * 2020-01-02 2022-09-02 宁波茂行生物医药科技有限公司 Modified immune effector cell and preparation method thereof
CN113061580B (en) * 2020-01-02 2022-09-06 宁波茂行生物医药科技有限公司 Modified immune effector cell and preparation method thereof
CN113061580A (en) * 2020-01-02 2021-07-02 江苏茂行科技有限公司 Modified immune effector cell and preparation method thereof
WO2021146487A2 (en) 2020-01-14 2021-07-22 Synthekine, Inc. Il2 orthologs and methods of use
WO2021252920A1 (en) 2020-06-11 2021-12-16 Novartis Ag Zbtb32 inhibitors and uses thereof
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2022036285A1 (en) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Compositions and methods for treating cancer with chimeric tim receptors in combination with inhibitors of poly (adp-ribose) polymerase
WO2022036287A1 (en) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Anti-cd72 chimeric receptors and uses thereof
WO2022036265A1 (en) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Chimeric tim receptors and uses thereof
WO2022096664A1 (en) 2020-11-09 2022-05-12 Miltenyi Biotec B.V. & Co. KG Methods and compositions for eliminating engineered immune cells
US11661459B2 (en) 2020-12-03 2023-05-30 Century Therapeutics, Inc. Artificial cell death polypeptide for chimeric antigen receptor and uses thereof
CN112830974A (en) * 2021-01-08 2021-05-25 深圳市珈钰生物科技有限公司 Chimeric antigen receptor, carrier, human dendritic cell, cell line, solid tumor treatment drug, preparation method and application
CN112830974B (en) * 2021-01-08 2022-07-26 深圳市珈钰生物科技有限公司 Chimeric antigen receptor, carrier, human dendritic cell, cell line, solid tumor treatment drug, preparation method and application
WO2022215011A1 (en) 2021-04-07 2022-10-13 Novartis Ag USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
WO2022229853A1 (en) 2021-04-27 2022-11-03 Novartis Ag Viral vector production system
WO2023274386A1 (en) * 2021-07-01 2023-01-05 宁波茂行生物医药科技有限公司 Universal car-t cell targeting egfr and preparation method therefor
WO2023010097A1 (en) 2021-07-28 2023-02-02 Cero Therapeutics, Inc. Chimeric tim4 receptors and uses thereof
WO2023122337A1 (en) * 2021-12-23 2023-06-29 Sana Biotechnology, Inc. Chimeric antigen receptor (car) t cells for treating autoimmune disease and associated methods
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors

Also Published As

Publication number Publication date
US10391126B2 (en) 2019-08-27
US20140349402A1 (en) 2014-11-27
US20190388472A1 (en) 2019-12-26
US11344577B2 (en) 2022-05-31

Similar Documents

Publication Publication Date Title
US11344577B2 (en) Car+ T cells genetically modified to eliminate expression of T-cell receptor and/or HLA
US11786582B2 (en) Human application of engineered chimeric antigen receptor (CAR) T-cells
Weber et al. The emerging landscape of immune cell therapies
US20190175651A1 (en) Immortalized car-t cells genetically modified to elminate t-cell receptor and beta 2-microglobulin expression
US20220025001A1 (en) Nucleic acid constructs for co-expression of chimeric antigen receptor and transcription factor, cells containing and therapeutic use thereof
KR20160068960A (en) Polyclonal gamma delta t cells for immunotherapy
CN115335087A (en) Compositions and methods for reducing transplant rejection in allogeneic cell therapy
US20220387493A1 (en) Method for producing cytotoxic effector memory t-cells for car t-cell treatment of cancer
JP2023543556A (en) Chimeric antigen receptor (CAR) with CD28 transmembrane domain
AU2022312508A1 (en) Polycistronic vectors for cell-based therapies

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: 12849570

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12849570

Country of ref document: EP

Kind code of ref document: A1