WO2014127261A1 - Chimeric antigen receptor and methods of use thereof - Google Patents

Chimeric antigen receptor and methods of use thereof Download PDF

Info

Publication number
WO2014127261A1
WO2014127261A1 PCT/US2014/016527 US2014016527W WO2014127261A1 WO 2014127261 A1 WO2014127261 A1 WO 2014127261A1 US 2014016527 W US2014016527 W US 2014016527W WO 2014127261 A1 WO2014127261 A1 WO 2014127261A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
car
heterodimeric
conditionally active
domain
Prior art date
Application number
PCT/US2014/016527
Other languages
French (fr)
Inventor
Chia-Yung WU
James Onuffer
Wendell A. Lim
Original Assignee
The Regents Of The University Of California
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
Priority to KR1020237003986A priority Critical patent/KR20230022452A/en
Priority to EP21158396.8A priority patent/EP3881868B1/en
Priority to EP17187293.0A priority patent/EP3300745B9/en
Priority to LTEP14751227.1T priority patent/LT2956175T/en
Priority to EP19188398.2A priority patent/EP3613439B1/en
Priority to KR1020217038402A priority patent/KR20210147101A/en
Priority to JP2015558159A priority patent/JP6450690B2/en
Priority to CA2901115A priority patent/CA2901115A1/en
Priority to BR112015019640-3A priority patent/BR112015019640A2/en
Priority to IL272279A priority patent/IL272279B2/en
Priority to KR1020197034038A priority patent/KR102132246B1/en
Priority to EP14751227.1A priority patent/EP2956175B1/en
Application filed by The Regents Of The University Of California filed Critical The Regents Of The University Of California
Priority to CN201480016737.0A priority patent/CN105142677B/en
Priority to EP23198143.2A priority patent/EP4303232A3/en
Priority to KR1020157024731A priority patent/KR102064230B1/en
Priority to ES14751227T priority patent/ES2653487T3/en
Priority to DK14751227.1T priority patent/DK2956175T3/en
Priority to MX2015010522A priority patent/MX369545B/en
Priority to KR1020207019131A priority patent/KR102332790B1/en
Priority to CN201910722248.1A priority patent/CN110423282B/en
Priority to NZ710925A priority patent/NZ710925B2/en
Priority to IL305629A priority patent/IL305629A/en
Priority to PL14751227T priority patent/PL2956175T3/en
Priority to PL19188398T priority patent/PL3613439T3/en
Priority to PL17187293T priority patent/PL3300745T3/en
Priority to US14/766,105 priority patent/US20150368342A1/en
Priority to AU2014216130A priority patent/AU2014216130B2/en
Publication of WO2014127261A1 publication Critical patent/WO2014127261A1/en
Priority to IL240472A priority patent/IL240472B/en
Priority to US15/064,938 priority patent/US9587020B2/en
Priority to HK16106629.8A priority patent/HK1218625A1/en
Priority to US15/419,729 priority patent/US9821012B2/en
Priority to US15/669,707 priority patent/US10632152B2/en
Priority to US15/801,133 priority patent/US10105391B2/en
Priority to US15/835,329 priority patent/US10888581B2/en
Priority to AU2018201102A priority patent/AU2018201102A1/en
Priority to AU2019246785A priority patent/AU2019246785B2/en
Priority to CY20191101245T priority patent/CY1122386T1/en
Priority to US16/824,434 priority patent/US11478510B2/en
Priority to US17/115,565 priority patent/US20210196757A1/en
Priority to AU2021204054A priority patent/AU2021204054B2/en
Priority to US17/932,259 priority patent/US20230051989A1/en
Priority to AU2023204612A priority patent/AU2023204612A1/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
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464412CD19 or B4
    • 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
    • A61K39/464466Adhesion molecules, e.g. NRCAM, EpCAM or cadherins
    • A61K39/464468Mesothelin [MSLN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6891Pre-targeting systems involving an antibody for targeting specific cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70517CD8
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70521CD28, CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70575NGF/TNF-superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/90Isomerases (5.)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y502/00Cis-trans-isomerases (5.2)
    • C12Y502/01Cis-trans-Isomerases (5.2.1)
    • C12Y502/01008Peptidylprolyl isomerase (5.2.1.8), i.e. cyclophilin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • 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
    • A61K2239/10Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the structure of the chimeric antigen receptor [CAR]
    • A61K2239/23On/off switch
    • 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
    • A61K2239/10Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the structure of the chimeric antigen receptor [CAR]
    • A61K2239/23On/off switch
    • A61K2239/24Dimerizable CARs; CARs with adapter
    • 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
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • 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
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
    • 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

Definitions

  • immune cells isolated from a patient can be used.
  • CAR chimeric antigen receptor
  • An example of a currently used CAR is a fusion of an extracellular recognition domain (e.g., an antigen-binding domain), a transmembrane domain, and one or more intracellular signaling domains.
  • the intracellular signaling portion of the CAR can initiate an activation-related response in an immune cell, such are release of cytolytic molecules to induce tumor cell death, etc.
  • CARs are not capable of being pharmacologically controlled. There is a need in the art for a conditionally activatable CAR that can be controlled pharmacologically.
  • the present disclosure provides a heterodimeric, conditionally active chimeric antigen
  • CAR CAR receptor
  • a nucleic acid comprising a nucleotide sequence encoding the CAR.
  • the present disclosure provides cells genetically modified to produce the CAR.
  • a CAR of the present disclosure can be used in various methods, which are also provided.
  • Figures 1A and IB provide nucleotide and amino acid sequences of the domains of
  • Figures 2A and 2B provide nucleotide and amino acid sequences of the domains of
  • Figures 3A and 3B provide nucleotide and amino acid sequences of the domains of
  • Figure 4 provides nucleotide and amino acid sequences of the domains of construct #126.
  • Figures 5A and 5B provide nucleotide and amino acid sequences of the domains of
  • Figures 6A-C provide nucleotide and amino acid sequences of the domains of construct #169.
  • Figures 7A and 7B provide nucleotide and amino acid sequences of the domains of
  • Figures 8A and 8B provide nucleotide and amino acid sequences of the domains of
  • Figures 9A-C provide nucleotide and amino acid sequences of the domains of construct #206.
  • Figures 10A and 10B provide nucleotide and amino acid sequences of the domains of construct #207.
  • Figures 11A-C provide nucleotide and amino acid sequences of the domains of construct #199.
  • Figure 12 depicts IL-2 production triggered by five On-switch CAR variants.
  • Figure 13 depicts IL-2 production by control Jurkat lines.
  • Figure 14 depicts a comparison between CAR constructs "122 + 206" and "197 + 206".
  • Figure 15 depicts cytotoxicity data with the On-switch CAR "197+206.”
  • Figure 16 depicts T cell activation data using CAR constructs "122 + 199"; “197 + 199”; and "122 + 168.”
  • Figure 17 is a schematic representation of an exemplary On-switch CAR.
  • Figures 18A and 18B depict various exemplary On-switch CAR.
  • Figures 19A-G depict IL-2 production triggered by 3 different On-switch CAR variants recognizing human mesothelin.
  • Figures 20A-C depict IL-2 production triggered by an On-switch CAR variant with a
  • Figures 21A-D depict exemplary On-switch CARs and conventional CARs with various co- stimulatory domains.
  • Figures 22A and 22B provide nucleotide and amino acid sequences of the domains of
  • Figures 23A and 23B provide nucleotide and amino acid sequences of the domains of
  • Figures 24A and 24B provide nucleotide and amino acid sequences of the domains of
  • Figures 25A and 25B provide nucleotide and amino acid sequences of the domains of
  • Figures 26A and 26B provide nucleotide and amino acid sequences of the domains of
  • Figures 27A and 27B provide nucleotide and amino acid sequences of the domains of
  • Figures 28A and 28B provide nucleotide and amino acid sequences of the domains of
  • Figures 29A and 29B provide nucleotide and amino acid sequences of the domains of
  • Figures 30A and 30B provide nucleotide and amino acid sequences of the domains of
  • Figures 31A and 3 IB provide nucleotide and amino acid sequences of the domains of
  • Figures 32A and 32B provide nucleotide and amino acid sequences of the domains of
  • Figures 33A and 33B provide nucleotide and amino acid sequences of the domains of
  • polynucleotide and “nucleic acid,” used interchangeably herein, refer to a polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides.
  • this term includes, but is not limited to, single-, double-, or multi-stranded DNA or RNA, genomic DNA, cDNA, DNA-RNA hybrids, or a polymer comprising purine and pyrimidine bases or other natural, chemically or biochemically modified, non-natural, or derivatized nucleotide bases.
  • antibodies and immunoglobulin include antibodies or immunoglobulins of any isotype, fragments of antibodies which retain specific binding to antigen, including, but not limited to, Fab, Fv, scFv, and Fd fragments, chimeric antibodies, humanized antibodies, single-chain antibodies, and fusion proteins comprising an antigen-binding portion of an antibody and a non-antibody protein.
  • Antibody fragments comprise a portion of an intact antibody, for example, the antigen binding or variable region of the intact antibody.
  • antibody fragments include Fab, Fab', F(ab') 2 , and Fv fragments; diabodies; linear antibodies (Zapata et al., Protein Eng. 8(10): 1057-1062 (1995)); single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • Papain digestion of antibodies produces two identical antigen -binding fragments, called "Fab” fragments, each with a single antigen-binding site, and a residual "Fc” fragment, a designation reflecting the ability to crystallize readily.
  • Pepsin treatment yields an F(ab') 2 fragment that has two antigen combining sites and is still capable of cross-linking antigen.
  • Single-chain Fv or “sFv” antibody fragments comprise the V H and V L domains of
  • the Fv polypeptide further comprises a polypeptide linker between the V H and V L domains, which enables the sFv to form the desired structure for antigen binding.
  • affinity refers to the equilibrium constant for the reversible
  • Affinity can be at least 1-fold greater, at least 2-fold greater, at least 3-fold greater, at least 4-fold greater, at least 5-fold greater, at least 6-fold greater, at least 7-fold greater, at least 8-fold greater, at least 9-fold greater, at least 10-fold greater, at least 20-fold greater, at least 30-fold greater, at least 40-fold greater, at least 50-fold greater, at least 60-fold greater, at least 70-fold greater, at least 80-fold greater, at least 90-fold greater, at least 100-fold greater, or at least 1000-fold greater, or more, than the affinity of an antibody for unrelated amino acid sequences.
  • Affinity of an antibody to a target protein can be, for example, from about 100 nanomolar (nM) to about 0.1 nM, from about 100 nM to about 1 picomolar (pM), or from about 100 nM to about 1 femtomolar (fM) or more.
  • nM nanomolar
  • pM picomolar
  • fM femtomolar
  • the term “avidity” refers to the resistance of a complex of two or more agents to dissociation after dilution.
  • the terms “immunoreactive” and “preferentially binds” are used interchangeably herein with respect to antibodies and/or antigen-binding fragments.
  • binding refers to a direct association between two molecules, due to, for
  • covalent, electrostatic, hydrophobic, and ionic and/or hydrogen-bond interactions including interactions such as salt bridges and water bridges.
  • Non-specific binding would refer to binding with an affinity of less than about 10 " M, e.g., binding with an affinity of 10 "6 M, 10 "5 M, 10 "4 M, etc.
  • hinge region refers to a flexible polypeptide connector region (also referred to herein as “hinge” or “spacer”) providing structural flexibility and spacing to flanking polypeptide regions and can consist of natural or synthetic polypeptides.
  • a "hinge region” derived from an immunoglobulin e.g., IgGl
  • IgGl immunoglobulin
  • a "hinge region” derived from an immunoglobulin is generally defined as stretching from Glu 21 6 to Pro 230 of human IgGl (Burton (1985) Molec. Immunol., 22: 161- 206).
  • Hinge regions of other IgG isotypes may be aligned with the IgGl sequence by placing the first and last cysteine residues forming inter-heavy chain disulfide (S-S) bonds in the same positions.
  • the hinge region may be of natural occurrence or non-natural occurrence, including but not limited to an altered hinge region as described in U.S. Pat. No. 5,677,425.
  • the hinge region can include complete hinge region derived from an antibody of a different class or subclass from that of the CHI domain.
  • the term "hinge region" can also include regions derived from CD8 and other receptors that provide a similar function in providing flexibility and spacing to flanking regions.
  • An "isolated" polypeptide is one that has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or
  • the polypeptide will be purified (1) to greater than 90%, greater than 95%, or greater than 98%, by weight of antibody as determined by the Lowry method, for example, more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by sodium dodecyl sulfate- polyacrylamide gel electrophoresis (SDS-PAGE) under reducing or nonreducing conditions using Coomassie blue or silver stain.
  • Isolated polypeptide includes the polypeptide in situ within recombinant cells since at least one component of the polypeptide's natural environment will not be present. In some instances, isolated polypeptide will be prepared by at least one purification step.
  • immune cells generally includes white blood cells (leukocytes) which are derived from hematopoietic stem cells (HSC) produced in the bone marrow.
  • HSC hematopoietic stem cells
  • Immune cells includes, e.g., lymphocytes (T cells, B cells, natural killer (NK) cells) and myeloid-derived cells (neutrophil, eosinophil, basophil, monocyte, macrophage, dendritic cells).
  • T cell includes all types of immune cells expressing CD3 including T-helper cells (CD4 + cells), cytotoxic T-cells (CD8 + cells), T-regulatory cells (Treg) and gamma-delta T cells.
  • a "cytotoxic cell” includes CD8 + T cells, natural-killer (NK) cells, and neutrophils, which cells are capable of mediating cytotoxicity responses.
  • stem cell generally includes pluripotent or multipotent stem
  • stem cells includes, e.g., embryonic stem cells (ES); mesenchymal stem cells (MSC); induced-pluripotent stem cells (iPS); and committed progenitor cells (hematopoeitic stem cells (HSC); bone marrow derived cells, etc.).
  • ES embryonic stem cells
  • MSC mesenchymal stem cells
  • iPS induced-pluripotent stem cells
  • HSC hematopoeitic stem cells
  • HSC bone marrow derived cells, etc.
  • treatment refers to obtaining a desired pharmacologic and/or physiologic effect.
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease.
  • Treatment covers any treatment of a disease in a mammal, e.g., in a human, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; and (c) relieving the disease, i.e., causing regression of the disease.
  • the terms "individual,” “subject,” “host,” and “patient,” used interchangeably herein, refer to a mammal, including, but not limited to, murines (e.g., rats, mice), lagomorphs (e.g., rabbits), non-human primates, humans, canines, felines, ungulates (e.g., equines, bovines, ovines, porcines, caprines), etc.
  • murines e.g., rats, mice
  • lagomorphs e.g., rabbits
  • non-human primates humans
  • canines felines
  • ungulates e.g., equines, bovines, ovines, porcines, caprines
  • a “therapeutically effective amount” or “efficacious amount” refers to the amount of an agent, or combined amounts of two agents, that, when administered to a mammal or other subject for treating a disease, is sufficient to effect such treatment for the disease.
  • the “therapeutically effective amount” will vary depending on the agent(s), the disease and its severity and the age, weight, etc., of the subject to be treated.
  • chimeric antigen receptor includes a plurality of such chimeric antigen receptor and reference to “the dimerizer-binding pair” includes reference to one or more dimerizer-binding pairs and equivalents thereof known to those skilled in the art, and so forth.
  • the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as “solely,” “only” and the like in connection with the recitation of claim elements, or use of a “negative” limitation.
  • the present disclosure provides a heterodimeric, conditionally active chimeric antigen
  • CAR CAR receptor
  • a nucleic acid comprising a nucleotide sequence encoding the CAR.
  • the present disclosure provides cells genetically modified to produce the CAR.
  • a CAR of the present disclosure can be used in various methods, which are also provided.
  • the present disclosure provides a heterodimeric, conditionally active chimeric antigen
  • CAR CAR receptor
  • a CAR of the present disclosure comprises: a) a first polypeptide comprising: i) a member of a specific binding pair (e.g., an antigen-binding domain); ii) a first modulatory domain; iii) a first member of a dimerization pair; and iv) a transmembrane domain interposed between the member of a specific binding pair (e.g., an antigen-binding domain) and the first modulatory domain; and b) a second polypeptide comprising: i) a transmembrane domain; ii) a second modulatory domain; iii) a second member of the dimerization pair; and iv) an intracellular signaling domain.
  • the modulatory domain can be a co- stimulatory domain.
  • a CAR of the present disclosure comprises: a) a first polypeptide comprising: i) a member of a specific binding pair (e.g., an antigen-binding domain); ii) a first co-stimulatory domain; iii) a first member of a dimerization pair (e.g., a dimerizer- binding pair); and iv) a transmembrane domain interposed between the member of a specific binding pair (e.g., an antigen-binding domain) and the first co- stimulatory domain; and b) a second polypeptide comprising: i) a transmembrane domain; ii) a second co- stimulatory domain; iii) a second member of the dimerization pair (e.g., the dimerizer-binding pair); and iv) an intracellular signaling domain.
  • a first polypeptide comprising: i) a member of a specific binding pair (e.g., an antigen-bind
  • a CAR of the present disclosure comprises: a) a first polypeptide comprising: i) a member of a specific binding pair (e.g., an antigen-binding domain); ii) a modulatory domain; iii) a first member of a dimerization pair (e.g., a dimerizer-binding pair); iv) a transmembrane domain interposed between the member of a specific binding pair (e.g., an antigen-binding domain) and the modulatory domain; and b) a second polypeptide comprising: i) a second member of the dimerization pair (e.g., the dimerizer- binding pair); and ii) an intracellular signaling domain.
  • the modulatory domain can be a co- stimulatory domain.
  • a CAR of the present disclosure comprises: a) a first polypeptide comprising: i) a member of a specific binding pair (e.g., an antigen-binding domain); ii) a co-stimulatory domain; iii) a first member of a dimerization pair (e.g., a dimerizer-binding pair); iv) a transmembrane domain interposed between the member of a specific binding pair (e.g., an antigen-binding domain) and the co- stimulatory domain; and b) a second polypeptide comprising: i) a second member of the dimerization pair (e.g., the dimerizer- binding pair); and ii) an intracellular signaling domain.
  • a first polypeptide comprising: i) a member of a specific binding pair (e.g., an antigen-binding domain); ii) a co-stimulatory domain; iii) a first member of a dimer
  • a CAR of the present disclosure can be present in the plasma membrane of a eukaryotic cell, e.g., a mammalian cell, where suitable mammalian cells include, but are not limited to, a cytotoxic cell, a T lymphocyte, a stem cell, a progeny of a stem cell, a progenitor cell, a progeny of a progenitor cell, and an NK cell.
  • a eukaryotic cell e.g., a mammalian cell
  • suitable mammalian cells include, but are not limited to, a cytotoxic cell, a T lymphocyte, a stem cell, a progeny of a stem cell, a progenitor cell, a progeny of a progenitor cell, and an NK cell.
  • a CAR of the present disclosure When present in the plasma membrane of a eukaryotic cell, a CAR of the present disclosure is active in the presence of: 1) a dimerizing agent binds to the first and second members of the dimerizer-binding pair in the CAR, or otherwise induces dimerization of the first and second members of the dimer; and 2) a factor that binds the member of a specific binding pair (e.g., an antigen-binding domain), e.g., an antigen that binds the antigen-binding domain of the CAR.
  • the factor that binds the member of the specific binding pair is a second member of the specific binding pair.
  • the second member of the specific binding pair can be a soluble (e.g., not bound to a cell) factor; a factor present on the surface of a cell such as a target cell; a factor presented on a solid surface; a factor present in a lipid bilayer; and the like.
  • the member of a specific binding pair is an antibody
  • the second member of the specific binding pair is an antigen
  • the antigen can be a soluble (e.g., not bound to a cell) antigen; an antigen present on the surface of a cell such as a target cell; an antigen presented on a solid surface; an antigen present in a lipid bilayer; and the like.
  • a CAR of the present disclosure when present in the plasma membrane of a eukaryotic cell, and when activated by a second member of a specific binding pair that binds the member of the specific-binding pair of the CAR (e.g., an antigen that binds the antigen -binding domain of the CAR) and a dimerizing agent, increases expression of at least one nucleic acid in the cell.
  • a second member of a specific binding pair that binds the member of the specific-binding pair of the CAR e.g., an antigen that binds the antigen -binding domain of the CAR
  • a dimerizing agent increases expression of at least one nucleic acid in the cell.
  • a CAR of the present disclosure when present in the plasma membrane of a eukaryotic cell, and when activated by an antigen that binds the antigen-binding domain of the CAR and a dimerizing agent, increases expression of at least one nucleic acid in the cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared with the level of transcription of the nucleic acid in the absence of the antigen and/or the dimerizing agent.
  • the second polypeptide of a CAR of the present disclosure can include an immunoreceptor tyrosine -based activation motif (ITAM)-containing intracellular signaling polypeptide; in such cases, a CAR of the present disclosure, when present in the plasma membrane of a eukaryotic cell, and when activated by an antigen that binds the antigen- binding domain of the CAR and a dimerizing agent, increases nuclear factor of activated T cells (NFAT)-dependent transcription.
  • ITAM immunoreceptor tyrosine -based activation motif
  • NFAT-dependent transcription includes transcription induced by any member of the NFAT family, including, e.g., NFATcl, NFATc2, NFATc3, NFATc4, NFAT5; AP-1; Spl; ⁇ ; and the like.
  • a CAR of the present disclosure when present in the plasma membrane of a eukaryotic cell, and when activated by an antigen that binds the antigen-binding domain of the CAR and a dimerizing agent, can, in some instances, result in increased production of one or more cytokines by the cell.
  • a CAR of the present disclosure when present in the plasma membrane of a eukaryotic cell, and when activated by an antigen that binds the antigen-binding domain of the CAR and a dimerizing agent, can increase production of a cytokine by the cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared with the amount of cytokine produced by the cell in the absence of the antigen and/or the dimerizing agent.
  • Cytokines whose production can be increased include, but are not limited to, an interferon, e.g., IL-2, interferon gamma (IFN- ⁇ ), tumor necrosis factor-alpha (TNF-a), IL-15, IL-12, IL-4, IL-5, IL-10; a chemokine; a growth factor; and the like.
  • an interferon e.g., IL-2, interferon gamma (IFN- ⁇ ), tumor necrosis factor-alpha (TNF-a), IL-15, IL-12, IL-4, IL-5, IL-10; a chemokine; a growth factor; and the like.
  • a CAR of the present disclosure when present in the plasma membrane of a eukaryotic cell, and when activated by an antigen that binds the antigen-binding domain of the CAR and a dimerizing agent, can result in both an increase in transcription of a nucleic acid in the cell and an increase in production of a cytokine by the cell.
  • a CAR of the present disclosure when present in the plasma membrane of a eukaryotic cell, and when activated by a dimerizing agent, results in cytotoxic activity by the cell toward a target cell that expresses on its cell surface an antigen to which the antigen -binding domain of the first polypeptide of the CAR binds.
  • a CAR of the present disclosure when present in the plasma membrane of the cell, and when activated by a dimerizing agent, increases cytotoxic activity of the cell toward a target cell that expresses on its cell surface an antigen to which the antigen-binding domain of the first polypeptide of the CAR binds.
  • a CAR of the present disclosure when present in the plasma membrane of the cell, and when activated by a dimerizing agent, increases cytotoxic activity of the cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared to the cytotoxic activity of the cell in the absence of the dimerizing agent.
  • a CAR of the present disclosure when present in the plasma membrane of a eukaryotic cell, and when activated by an antigen that binds the antigen-binding domain of the CAR and a dimerizing agent, can result in other CAR activation related events such as proliferation and expansion (either due to increased cellular division or anti-apoptotic responses).
  • a CAR of the present disclosure when present in the plasma membrane of a eukaryotic cell, and when activated by an antigen that binds the antigen-binding domain of the CAR and a dimerizing agent, can result in other CAR activation related events such as intracellular signaling modulation, cellular differentiation, or cell death.
  • a CAR of the present disclosure can be present in a eukaryotic cell membrane, where the first and second polypeptides of the CAR are not covalently linked to one another.
  • a CAR of the present disclosure can be present in a eukaryotic cell membrane as a single heterodimer that is not covalently linked to any other polypeptide in the membrane.
  • a first CAR of the present disclosure can be present in a eukaryotic cell membrane as a heterodimer that is covalently or non-covalently linked to a second CAR of the present disclosure.
  • the first and the second CAR are covalently linked via a disulfide bond formed between cysteines present in a hinge region present in both the first polypeptide of the first CAR and the first polypeptide of the second CAR.
  • a CAR of the present disclosure can be present in a eukaryotic cell
  • first polypeptides of the CAR comprise an antibody fragment and the second polypeptides of the CAR comprise a signal transducing domain derived from a cytokine receptor, such that, upon dimerization, the CAR may represent a heterodimeric- signalobody CAR, e.g., a signalobody composed of at least two independent polypeptides.
  • a "signalobody”, as it is known in the art, is a single chimeric macromolecule composed of an antibody fragment and a signal transduction domain derived from a cytokine receptor.
  • a heterodimeric-signalobody CAR of the present disclosure when present in the cell membrane of a eukaryotic cell, dimerized by a dimerizer, and activated by an antigen, e.g., an oligomerized antigen, may induce the oligomerization of the heterodimeric- signalobody CAR.
  • an antigen e.g., an oligomerized antigen
  • Such ligand-induced oligomerization of a heterodimeric-signalobody CAR may activate, e.g., increase, or perpetuate, e.g., maintain, signal transduction, e.g., ligand-induced oligomerization of a heterodimeric-signalobody CAR may transmit a signal eliciting a cellular response.
  • a plurality of heterodimeric-signalobody CARs may be utilized combinatorially to elicit a desired cellular response.
  • a CAR of the present disclosure includes a member of a specific binding pair.
  • Specific binding pairs include, but are not limited to, antigen-antibody binding pairs; ligand-receptor binding pairs; and the like.
  • a member of a specific binding pair suitable for use in a CAR of the present disclosure includes an antigen; an antibody; a ligand; and a ligand- binding receptor.
  • an antigen-binding domain suitable for use in a CAR of the present disclosure can be any antigen-binding polypeptide, a wide variety of which are known in the art.
  • the antigen-binding domain is a single chain Fv (scFv).
  • Other antibody based recognition domains cAb VHH (camelid antibody variable domains) and humanized versions, IgNAR VH (shark antibody variable domains) and humanized versions, sdAb VH (single domain antibody variable domains) and "camelized" antibody variable domains are suitable for use.
  • T-cell receptor (TCR) based recognition domains such as single chain TCR (scTv, single chain two-domain TCR containing ⁇ ⁇ ) are also suitable for use.
  • an antigen-binding domain suitable for use in a CAR of the present disclosure can have a variety of antigen-binding specificities.
  • the antigen-binding domain is specific for an epitope present in an antigen that is expressed by (synthesized by) a cancer cell, i.e., a cancer cell associated antigen.
  • the cancer cell associated antigen can be an antigen associated with, e.g., a breast cancer cell, a B cell lymphoma, a Hodgkin lymphoma cell, an ovarian cancer cell, a prostate cancer cell, a mesothelioma, a lung cancer cell (e.g., a small cell lung cancer cell), a non-Hodgkin B-cell lymphoma (B-NHL) cell, an ovarian cancer cell, a prostate cancer cell, a mesothelioma cell, a lung cancer cell (e.g., a small cell lung cancer cell), a melanoma cell, a chronic lymphocytic leukemia cell, an acute lymphocytic leukemia cell, a neuroblastoma cell, a glioma, a glioblastoma, a
  • a cancer cell associated antigen may also be expressed by a non-cancerous cell.
  • Non-limiting examples of antigens to which an antigen-binding domain of a subject CAR can bind include, e.g., CD19, CD20, CD38, CD30, Her2/neu, ERBB2, CA125, MUC-1, prostate-specific membrane antigen (PSMA), CD44 surface adhesion molecule, mesothelin, carcinoembryonic antigen (CEA), epidermal growth factor receptor (EGFR), EGFRvIII, vascular endothelial growth factor receptor-2 (VEGFR2), high molecular weight-melanoma associated antigen (HMW-MAA), MAGE-A1, IL-13R-a2, GD2, and the like.
  • PSMA prostate-specific membrane antigen
  • CEA carcinoembryonic antigen
  • EGFR epidermal growth factor receptor
  • EGFRvIII vascular endothelial growth factor receptor-2
  • HMW-MAA high molecular weight-melanoma associated antigen
  • MAGE-A1 IL-13R-
  • a member of a specific binding pair suitable for use in a subject CAR is a ligand for a receptor.
  • Ligands include, but are not limited to, cytokines (e.g., IL-13, etc.); growth factors (e.g., heregulin; vascular endothelial growth factor (VEGF); and the like); an integrin-binding peptide (e.g., a peptide comprising the sequence Arg-Gly-Asp); and the like.
  • the CAR can be activated in the presence of both a dimerizer agent and a second member of the specific binding pair, where the second member of the specific binding pair is a receptor for the ligand.
  • the ligand is VEGF
  • the second member of the specific binding pair can be a VEGF receptor, including a soluble VEGF receptor.
  • the second member of the specific binding pair can be Her2. Receptors
  • the member of a specific binding pair that is included in a subject CAR is a receptor, e.g., a receptor for a ligand, a co-receptor, etc.
  • the receptor can be a ligand-binding fragment of a receptor.
  • Suitable receptors include, but are not limited to, a growth factor receptor (e.g., a VEGF receptor); a killer cell lectin-like receptor subfamily K, member 1 (NKG2D) polypeptide (receptor for MICA, MICB, and ULB6); a cytokine receptor (e.g., an IL-13 receptor; an IL-2 receptor; etc.); Her2; CD27; a natural cytotoxicity receptor (NCR) (e.g., NKP30 (NCR3/CD337) polypeptide (receptor for HLA-B-associated transcript 3 (BAT3) and B7-H6); etc.); etc.
  • a growth factor receptor e.g., a VEGF receptor
  • a killer cell lectin-like receptor subfamily K, member 1 (NKG2D) polypeptide receptor for MICA, MICB, and ULB6
  • a cytokine receptor e.g., an IL-13 receptor; an IL-2 receptor
  • the first polypeptide of a subject CAR comprises a hinge region (also
  • the hinge region is interposed between the antigen- binding domain and the transmembrane domain.
  • the hinge region is an immunoglobulin heavy chain hinge region.
  • the hinge region is a hinge region polypeptide derived from a receptor (e.g., a CD8-derived hinge region).
  • the hinge region can have a length of from about 4 amino acids to about 50 amino acids, e.g., from about 4 aa to about 10 aa, from about 10 aa to about 15 aa, from about 15 aa to about 20 aa, from about 20 aa to about 25 aa, from about 25 aa to about 30 aa, from about 30 aa to about 40 aa, or from about 40 aa to about 50 aa.
  • Suitable spacers can be readily selected and can be of any of a number of suitable lengths, such as from 1 amino acid (e.g., Gly) to 20 amino acids, from 2 amino acids to 15 amino acids, from 3 amino acids to 12 amino acids, including 4 amino acids to 10 amino acids, 5 amino acids to 9 amino acids, 6 amino acids to 8 amino acids, or 7 amino acids to 8 amino acids, and can be 1, 2, 3, 4, 5, 6, or 7 amino acids.
  • 1 amino acid e.g., Gly
  • suitable lengths such as from 1 amino acid (e.g., Gly) to 20 amino acids, from 2 amino acids to 15 amino acids, from 3 amino acids to 12 amino acids, including 4 amino acids to 10 amino acids, 5 amino acids to 9 amino acids, 6 amino acids to 8 amino acids, or 7 amino acids to 8 amino acids, and can be 1, 2, 3, 4, 5, 6, or 7 amino acids.
  • Exemplary spacers include glycine polymers (G) n , glycine- serine polymers (including, for example, (GS) n , (GSGGS) n (SEQ ID NO:37) and (GGGS) n (SEQ ID NO:38), where n is an integer of at least one), glycine-alanine polymers, alanine-serine polymers, and other flexible linkers known in the art. Glycine and glycine-serine polymers can be used; both Gly and Ser are relatively unstructured, and therefore can serve as a neutral tether between components.
  • Glycine polymers can be used; glycine accesses significantly more phi-psi space than even alanine, and is much less restricted than residues with longer side chains (see Scheraga, Rev. Computational Chem. 11173-142 (1992)).
  • Exemplary spacers can comprise amino acid sequences including, but not limited to, GGSG (SEQ ID NO:39), GGSGG (SEQ ID NO:40), GSGSG (SEQ ID NO:41), GSGGG (SEQ ID NO:42), GGGSG
  • the hinge region in the first polypeptide of a subject CAR includes at least one cysteine.
  • the hinge region can include the sequence Cys-
  • a cysteine in the hinge region of a first CAR can be available to form a disulfide bond with a hinge region in a second CAR.
  • Immunoglobulin hinge region amino acid sequences are known in the art; see, e.g., Tan et al. (1990) Proc. Natl. Acad. Sci. USA 87: 162; and Huck et al. (1986) Nucl. Acids Res.
  • an immunoglobulin hinge region can include one of the following amino acid sequences: DKTHT (SEQ ID NO:45); CPPC (SEQ ID NO:46);
  • KCCVDCP (SEQ ID NO:50); KYGPPCP (SEQ ID NO:51); EPKSCDKTHTCPPCP (SEQ ID NO:50);
  • ELKTPLGDTTHTCPRCP (SEQ ID NO:54) (human IgG3 hinge); SPNMVPHAHHAQ
  • the hinge region can comprise an amino acid sequence of a human IgGl, IgG2, IgG3, or
  • the hinge region can include one or more amino acid substitutions and/or insertions and/or deletions compared to a wild-type (naturally-occurring) hinge region.
  • His 2 29 of human IgGl hinge can be substituted with Tyr, so that the hinge region comprises the sequence EPKSCDKTYTCPPCP (SEQ ID NO:52); see, e.g.,
  • the hinge region can comprise an amino acid sequence derived from human CD8; e.g., the hinge region can comprise the amino acid sequence:
  • TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACD (SEQ ID NO:56), or a variant thereof.
  • the first and the second polypeptides of a CAR of the present disclosure include
  • transmembrane domains for insertion into a eukaryotic cell membrane.
  • the transmembrane domain of the first polypeptide is interposed between the antigen-binding domain and the co-stimulatory domain.
  • the first polypeptide includes a hinge region, the
  • transmembrane domain is interposed between the hinge region and the co- stimulatory domain, such that the first polypeptide comprises, in order from the amino terminus (N- terminus) to the carboxyl terminus (C-terminus): an antigen-binding domain; a hinge region; a transmembrane domain; a first co- stimulatory domain; and a first member of a dimerizer-binding pair.
  • the transmembrane domain of the second polypeptide is at or near the N-terminus of the polypeptide, such that the second polypeptide comprises, in order from N-terminus to C- terminus: a transmembrane domain; a second co-stimulatory domain; a second member of the dimerizer-binding pair; and an intracellular signaling domain.
  • TM domain transmembrane domain that provides for insertion of a polypeptide into the cell membrane of a eukaryotic (e.g., mammalian) cell is suitable for use.
  • TM sequence IYIWAPLAGTCGVLLLSLVITLYC SEQ ID NO:30
  • suitable TM sequences include: a) CD8 beta derived : LGLLVAGVLVLLVSLGVAIHLCC (SEQ ID NO:57); b) CD4 derived:
  • WVLVVVGGVLACYSLLVTVAFIIFWV (SEQ ID NO:60); e) CD134 (OX40) derived: VAAILGLGLVLGLLGPLAILLALYLL (SEQ ID NO:61); and f) CD7 derived:
  • a first polypeptide of a subject CAR includes a linker between any two
  • a linker can be disposed between the transmembrane domain and the first co- stimulatory domain of the first polypeptide.
  • a linker can be disposed between the first co-stimulatory domain and the first member of a dimerizer-binding pair of the first polypeptide.
  • a linker can be disposed between the transmembrane domain and the second co- stimulatory domain of the second polypeptide.
  • a linker can be disposed between the second co- stimulatory domain and the second member of the dimerizer-binding pair of the second polypeptide.
  • a linker can be disposed between the second member of the dimerizer-binding pair and the intracellular signaling domain of the second polypeptide.
  • the linker peptide may have any of a variety of amino acid sequences. Proteins can be joined by a spacer peptide, generally of a flexible nature, although other chemical linkages are not excluded.
  • a linker can be a peptide of between about 6 and about 40 amino acids in length, or between about 6 and about 25 amino acids in length. These linkers can be produced by using synthetic, linker-encoding oligonucleotides to couple the proteins.
  • Peptide linkers with a degree of flexibility can be used.
  • the linking peptides may have virtually any amino acid sequence, bearing in mind that suitable linkers will have a sequence that results in a generally flexible peptide.
  • suitable linkers will have a sequence that results in a generally flexible peptide.
  • small amino acids such as glycine and alanine, are of use in creating a flexible peptide. The creation of such sequences is routine to those of skill in the art.
  • Suitable linkers can be readily selected and can be of any of a suitable of different lengths, such as from 1 amino acid (e.g., Gly) to 20 amino acids, from 2 amino acids to 15 amino acids, from 3 amino acids to 12 amino acids, including 4 amino acids to 10 amino acids, 5 amino acids to 9 amino acids, 6 amino acids to 8 amino acids, or 7 amino acids to 8 amino acids, and may be 1, 2, 3, 4, 5, 6, or 7 amino acids.
  • Exemplary flexible linkers include glycine polymers (G) n , glycine- serine polymers
  • n is an integer of at least one
  • Glycine and glycine- serine polymers are of interest since both of these amino acids are relatively unstructured, and therefore may serve as a neutral tether between components.
  • Glycine polymers are of particular interest since glycine accesses significantly more phi-psi space than even alanine, and is much less restricted than residues with longer side chains (see Scheraga, Rev. Computational Chem.
  • Exemplary flexible linkers include, but are not limited GGSG (SEQ ID NO:39), GGSGG (SEQ ID NO:40), GSGSG (SEQ ID NO:41), GSGGG (SEQ ID NO:42), GGGSG (SEQ ID NO:43), GSSSG (SEQ ID NO:44), and the like.
  • GGSG SEQ ID NO:39
  • GGSGG SEQ ID NO:40
  • GSGSG SEQ ID NO:41
  • GSGGG SEQ ID NO:42
  • GGGSG SEQ ID NO:43
  • GSSSG SEQ ID NO:44
  • Modulatory domains suitable for use in a CAR of the present disclosure include co- stimulatory domains.
  • the modulatory domain on the first polypeptide of a subject CAR has
  • the modulatory domain on the first polypeptide of a CAR comprises an amino acid sequence that is at least about 90%, at least about 95%, at least about 98%, at least about 99%, or 100%, identical to the amino acid sequence of the modulatory domain on the second polypeptide of the CAR.
  • the modulatory domain of the first polypeptide of a subject CAR can have substantially the same length as the modulatory domain of the second polypeptide of a subject CAR; e.g., the first and second modulatory domains can differ in length from one another by fewer than 10 amino acids, or fewer than 5 amino acids. In some cases, the first and second modulatory domains have the same length.
  • a modulatory domain suitable for inclusion in the first and the second polypeptide of a subject CAR can have a length of from about 30 amino acids to about 70 amino acids (aa), e.g., a modulatory domain can have a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
  • modulatory domain can have a length of from about 70 aa to about 100 aa, from about 100 aa to about 200 aa, or greater than 200 aa.
  • Co- stimulatory domains suitable for use in a CAR of the present disclosure are
  • co- stimulatory domains homodimerize.
  • a subject co- stimulatory domain can be an intracellular portion of a transmembrane protein (i.e., the co- stimulatory domain can be derived from a
  • transmembrane protein transmembrane protein
  • suitable co-stimulatory polypeptides include, but are not limited to, 4-lBB (CD137), CD28, ICOS, OX-40, BTLA, CD27, CD30, GITR, and HVEM.
  • the co-stimulatory domain on the first polypeptide of a subject CAR has substantially the same amino acid sequence as the co-stimulatory domain on the second polypeptide of the CAR.
  • the co- stimulatory domain on the first polypeptide of a CAR comprises an amino acid sequence that is at least about 90%, at least about 95%, at least about 98%, at least about 99%, or 100%, identical to the amino acid sequence of the co- stimulatory domain on the second polypeptide of the CAR.
  • the co- stimulatory domain of the first polypeptide of a subject CAR can have substantially the same length as the co- stimulatory domain of the second polypeptide of a subject CAR; e.g., the first and second co- stimulatory domains can differ in length from one another by fewer than 10 amino acids, or fewer than 5 amino acids. In some cases, the first and second co- stimulatory domains have the same length.
  • polypeptide of a subject CAR can have a length of from about 30 amino acids to about 70 amino acids (aa), e.g., a co- stimulatory domain can have a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
  • the co- stimulatory domain can have a length of from about 70 aa to about 100 aa, from about 100 aa to about 200 aa, or greater than 200 aa.
  • the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein 4-1BB (also known as TNFRSF9; CD137; 4-1BB; CDwl37; ILA; etc.).
  • a suitable co-stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
  • the co- stimulatory domain of both the first and the second polypeptide has a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
  • the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein CD28 (also known as Tp44).
  • a suitable co- stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
  • the co- stimulatory domain of both the first and the second polypeptide has a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
  • the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein ICOS (also known as AILIM, CD278, and CVID1).
  • a suitable co-stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
  • the co- stimulatory domain of both the first and the second polypeptide has a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
  • the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein OX-40 (also known as TNFRSF4, RP5-902P8.3, ACT35, CD134, OX40, TXGP1L).
  • a suitable co- stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
  • the co- stimulatory domain of both the first and the second polypeptide has a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
  • the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein BTLA (also known as BTLA1 and CD272).
  • BTLA also known as BTLA1 and CD272.
  • a suitable co-stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence: CCLRRHQGKQNELSDTAGREINLVDAHLKSEQTEASTRQNSQVLLSETGIYDNDPD LCFRMQEGSEVYSNPCLEENKPGIVYASLNHSVIGPNSRLARNVKEAPTEYASICVR
  • the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein CD27 (also known as S 152, T14, TNFRSF7, and Tp55).
  • a suitable co-stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
  • the co- stimulatory domain of both the first and the second polypeptide has a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
  • the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein CD30 (also known as TNFRSF8, D1S166E, and Ki-1).
  • a suitable co-stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, from about 150 aa to about 160 aa, or from about 160 aa to about 185 aa of the following amino acid sequence:
  • the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein GITR (also known as TNFRSF18, RP5-902P8.2, AITR, CD357, and GITR-D).
  • GITR also known as TNFRSF18, RP5-902P8.2, AITR, CD357, and GITR-D.
  • a suitable co- stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
  • the co-stimulatory domain of both the first and the second polypeptide has a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
  • the co-stimulatory domain derived from an intracellular portion of the transmembrane protein HVEM (also known as TNFRSF14, RP3-395M20.6, ATAR, CD270, HVEA, HVEM, LIGHTR, and TR2).
  • a suitable co-stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
  • the co-stimulatory domain of both the first and the second polypeptide has a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
  • Dimer pairs suitable for use in a subject CAR include dimerizer-binding pairs.
  • Dimerizer-binding pairs suitable for use in a CAR of the present disclosure are in some embodiments polypeptides that bind to a different site of the same molecule (referred to herein as a "dimerizer").
  • a dimerizer polypeptides that bind to a different site of the same molecule
  • binding to the dimerizer is reversible.
  • binding to the dimerizer is irreversible.
  • binding to the dimerizer is non-covalent.
  • binding to the dimerizer is covalent.
  • dimer pairs suitable for use include dimerizer-binding pairs that dimerize upon binding of a first member of a dimer pair to a dimerizing agent, where the dimerizing agent induces a conformational change in the first member of the dimer pair, and where the conformational change allows the first member of the dimer pair to bind (covalently or non- covalently) to a second member of the dimer pair.
  • dimer pairs suitable for use include dimer pairs in which exposure to light
  • dimer pair will dimerize upon exposure to an agent that induces dimerization, where the agent is in some cases a small molecule, or, in other cases, light.
  • agent that induces dimerization
  • dimerizer-binding pairs includes dimer pairs that dimerize regardless of the mechanism.
  • dimers include, but are not limited to:
  • FKBP FK506 binding protein
  • DHFR dihydrofolate reductase
  • a first or a second member of a dimer e.g., a dimerizer-binding pair of a subject
  • CAR can have a length of from about 50 amino acids to about 300 amino acids or more; e.g., a first or a second member of a dimer (e.g., a dimerizer-binding pair) of a subject CAR can have a length of from about 50 aa to about 100 aa, from about 100 aa to about 150 aa, from about 150 aa to about 200 aa, from about 200 aa toa bout 250 aa, from about 250 aa to about 300 aa, or more than 300 aa.
  • a dimer e.g., a dimerizer-binding pair
  • a member of a dimer e.g., a dimerizer-binding pair of a subject
  • a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
  • a member of a dimerizer-binding pair of a subject CAR is derived from calcineurin catalytic subunit A (also known as PPP3CA; CALN; CALNA; CALNA1; CCN1; CNA1; PPP2B; CAM-PRP catalytic subunit; calcineurin A alpha; calmodulin- dependent calcineurin A subunit alpha isoform; protein phosphatase 2B, catalytic subunit, alpha isoform; etc.).
  • calcineurin catalytic subunit A also known as PPP3CA; CALN; CALNA; CALNA1; CCN1; CNA1; PPP2B; CAM-PRP catalytic subunit; calcineurin A alpha; calmodulin- dependent calcineurin A subunit alpha isoform; protein phosphatase 2B, catalytic subunit, alpha isoform; etc.
  • a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence (PP2Ac domain):
  • a member of a dimer is derived from cyclophilin (also known cyclophilin A, PPIA, CYPA, CYPH, PPIase A, etc.).
  • cyclophilin also known cyclophilin A, PPIA, CYPA, CYPH, PPIase A, etc.
  • a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
  • a member of a dimer (e.g., a dimerizer-binding pair) is derived from
  • MTOR also known as FKBP-rapamycin associated protein; FK506 binding protein 12- rapamycin associated protein 1; FK506 binding protein 12-rapamycin associated protein 2; FK506-binding protein 12-rapamycin complex-associated protein 1; FRAP; FRAP1;
  • a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence (also known as "Frb”: Fkbp- Rapamycin Binding Domain):
  • a member of a dimer (e.g., a dimerizer-binding pair) is derived from
  • GyrB also known as DNA gyrase subunit B.
  • a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 200 amino acids (aa), from about 200 aa to about 300 aa, from about 300 aa to about 400 aa, from about 400 aa to about 500 aa, from about 500 aa to about 600 aa, from about 600 aa to about 700 aa, or from about 700 aa to about 800 aa, of the following GyrB amino acid sequence from Escherichia coli (or to the DNA gyrase subunit B sequence from any organism):
  • a member of a dimerizer-binding pair comprises an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to amino acids 1-220 of the above-listed GyrB amino acid sequence from Escherichia coli.
  • a member of a dimer (e.g., a dimerizer-binding pair) is derived from
  • a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
  • a member of a dimer is derived from the DmrB binding domain (i.e., DmrB homodimerization domain).
  • a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence: MASRGVQVETISPGDGRTFPKRGQTCVVHYTGMLEDGKKVDSSRDRNKPFKFMLG KQEVIRGWEEGVAQMSVGQRAKLTISPDYAYGATGHPGIIPPHATLVFDVELLKLE
  • a member of a dimer (e.g., a dimerizer-binding pair) is derived from a
  • PYL protein also known as abscisic acid receptor and as RCAR.
  • a member of a subject dimerizer-binding pair can be derived from proteins such as those of Arabidopsis thaliana: PYR1, RCAR1(PYL9), PYL1, PYL2, PYL3, PYL4, PYL5, PYL6, PYL7, PYL8 (RCAR3), PYL10, PYL11, PYL12, PYL13.
  • a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to any of the following amino acid sequences:
  • a member of a dimer is derived from an ABI protein (also known as Abscisic Acid- Insensitive).
  • ABI protein also known as Abscisic Acid- Insensitive
  • a member of a subject dimerizer-binding pair can be derived from proteins such as those of Arabidopsis thaliana: ABI1 (Also known as ABSCISIC ACID-INSENSITIVE 1, Protein phosphatase 2C 56, AtPP2C56, P2C56, and PP2C ABI1) and/or ABI2(also known as P2C77, Protein phosphatase 2C 77, AtPP2C77, ABSCISIC ACID-INSENSITIVE 2, Protein phosphatase 2C ABI2, and PP2C ABI2).
  • a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, from about 150 aa to about 160 aa, from about 160 aa to about 170 aa, from about 170 aa to about 180 aa, from about 180 aa to about 190 aa, or from about 190 aa to about 200 aa of any of the following amino acid sequences:
  • a member of a dimer (e.g., a dimerizer-binding pair) is derived from a
  • Cry2 protein also known as cryptochrome 2.
  • a member of a subject dimer e.g., a dimerizer-binding pair
  • Cry2 proteins from any organism (e.g., a plant) such as, but not limited to, those of Arabidopsis thaliana.
  • a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, from about 150 aa to about 160 aa, from about 160 aa to about 170 aa, from about 170 aa to about 180 aa, from about 180 aa to about 190 aa, or from about 190 aa to about 200 aa of any of the following amino acid sequences:
  • a member of a dimer is derived from the CIB1 Arabidopsis thaliana protein (also known as transcription factor bHLH63).
  • a suitable dimer (e.g., a dimerizer-binding pair) member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, from about 150 aa to about 160 aa, from about 160 aa to about 170 aa, from about 170 aa to about 180 aa, from about 180 aa to about 190 aa, or from about 190 aa to about 200 aa of the following amino acid sequence:
  • a member of a dimer is derived from the G AI Arabidopsis thaliana protein (also known as Gibberellic Acid Insensitive, and DELLA protein GAI).
  • a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, from about 150 aa to about 160 aa, from about 160 aa to about 170 aa, from about 170 aa to about 180 aa, from about 180 aa to about 190 aa, or from about 190 aa to about 200 aa of the following amino acid sequence:
  • a member of a dimer (e.g., a dimerizer-binding pair) is derived from a
  • a suitable dimer member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, from about 150 aa to about 160 aa, from about 160 aa to about 170 aa, from about 170 aa to about 180 aa, from about 180 aa to about 190 aa, or from about 190 aa to about 200 aa of any of the following amino acid sequences:
  • GID1A [00149]
  • GID1B [00150]
  • GID1C [00151]
  • AFVNAECQ (SEQ ID NO:97).
  • Dimerizers (“dimerizing agents) that can provide for dimerization of a first member of a dimerizer-binding pair and a second member of a dimerizer-binding pair include, e.g. (where the dimerizer is in parentheses following the dimerizer-binding pair:
  • FKBP and FKBP rapamycin
  • GyrB and GyrB (coumermycin);
  • rapamycin can serve as a dimerizer.
  • a rapamycin derivative or analog can be used. See, e.g., W096/41865; WO 99/36553; WO 01/14387; and Ye et al (1999) Science 283:88-91.
  • analogs, homologs, derivatives and other compounds related structurally to rapamycin include, among others, variants of rapamycin having one or more of the following modifications relative to rapamycin: demethylation, elimination or replacement of the methoxy at C7, C42 and/or C29; elimination, derivatization or replacement of the hydroxy at CI 3, C43 and/or C28; reduction, elimination or derivatization of the ketone at C14, C24 and/or C30; replacement of the 6-membered pipecolate ring with a 5-membered prolyl ring; and alternative substitution on the cyclohexyl ring or replacement of the cyclohexyl ring with a substituted cyclopentyl ring.
  • Rapamycin has the structure:
  • rapalog Also suitable as a rapalog is a compound of the formula:
  • n is 1 or 2; R and R are independently H, or a substituted or unsubstituted aliphatic or acyl moiety; one of R 7a and R 7b is H and the other is halo, R A , OR A , SR A , - OC(0)R A , -OC(0)NR A R B , -NR A R B , -NR B C(OR)R A , NR B C(0)OR A , -NR B S0 2 R A , or NR B S0 2 NR A R B ; or R 7a and R 7b , taken together, are H in the tetraene moiety:
  • R is H or a substituted or unsubstituted aliphatic, hetero aliphatic, aryl, or heteroaryl moiety and where R B and R B' are independently H, OH, or a substituted or unsubstituted aliphatic, heteroaliphatic, aryl, or heteroaryl moiety.
  • coumermycin can serve as a dimerizing agent.
  • a dimerizing agent e.g., a dimerizing agent for a dimerizing agent for a dimerizing agent for a dimerizing agent for a dimerizing agent for a dimerizing agent for a dimerizing agent.
  • coumermycin analog can be used. See, e.g., Farrar et al. (1996) Nature 383: 178-181; and U.S. Pat. No. 6,916,846.
  • the dimerizing agent is methotrexate, e.g., a non- cytotoxic, homo-bifunctional methotrexate dimer. See, e.g., U.S. Patent No. 8,236,925. Intracellular signaling domain
  • Intracellular signaling domains suitable for use in a CAR of the present disclosure include any desired signaling domain that provides a distinct and detectable signal (e.g., increased production of one or more cytokines by the cell; change in transcription of a target gene; change in activity of a protein; change in cell behavior, e.g., cell death; cellular proliferation; cellular differentiation; cell survival; modulation of cellular signaling responses; etc.) in response to activation of the CAR (i.e., activated by antigen and dimerizing agent).
  • the intracellular signaling domain includes at least one (e.g., one, two, three, four, five, six, etc.) IT AM motifs as described below.
  • the intracellular signaling domain includes DAP10/CD28 type signaling chains.
  • the intracellular signaling domain is not covalently attached to the membrane bound CAR, but is instead diffused in the cytoplasm.
  • Intracellular signaling domains suitable for use in a CAR of the present disclosure include immunoreceptor tyrosine-based activation motif (ITAM)-containing intracellular signaling polypeptides.
  • ITAM immunoreceptor tyrosine-based activation motif
  • An ITAM motif is YXiX 2 L/I, where and X 2 are independently any amino acid (SEQ ID NO: 130).
  • the intracellular signaling domain of a subject CAR comprises 1, 2, 3, 4, or 5 ITAM motifs.
  • an ITAM motif is repeated twice in an intracellular signaling domain, where the first and second instances of the ITAM motif are separated from one another by 6 to 8 amino acids, e.g., (YX 1 X 2 L/I)(X 3 ) n (YX 1 X 2 L/I), where n is an integer from 6 to 8, and each of the 6-8 X 3 can be any amino acid (SEQ ID NO: 131).
  • the intracellular signaling domain of a subject CAR comprises 3 IT AM motifs.
  • a suitable intracellular signaling domain can be an ⁇ motif-containing portion that is derived from a polypeptide that contains an IT AM motif.
  • a suitable intracellular signaling domain can be an IT AM motif-containing domain from any IT AM motif-containing protein.
  • a suitable intracellular signaling domain need not contain the entire sequence of the entire protein from which it is derived.
  • IT AM motif-containing polypeptides include, but are not limited to: DAP12; FCER1G (Fc epsilon receptor I gamma chain); CD3D (CD3 delta); CD3E (CD3 epsilon); CD3G (CD3 gamma); CD3Z (CD3 zeta); and CD79A (antigen receptor complex-associated protein alpha chain).
  • the intracellular signaling domain is derived from DAP12 (also
  • a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to any of the following amino acid sequences (4 isoforms):
  • MGGLEPCSRLLLLPLLLAVSDCSCSTVSPGVLAGIVMGDLVLTVLIALAVYFLGRLV PRGRGAAEATRKORITETESPYOELOGORSDVYSDLNTQRPYYK (SEQ ID NO: 101), where the IT AM motifs are in bold and are underlined.
  • a suitable intracellular signaling domain polypeptide can comprise an
  • a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to the following amino acid sequence:
  • the intracellular signaling domain is derived from FCER1G (also known as FCRG; Fc epsilon receptor I gamma chain; Fc receptor gamma-chain; fc-epsilon Rl-gamma; fcRgamma; fceRI gamma; high affinity immunoglobulin epsilon receptor subunit gamma; immunoglobulin E receptor, high affinity, gamma chain; etc.).
  • FCER1G also known as FCRG
  • Fc epsilon receptor I gamma chain Fc receptor gamma-chain
  • fcRgamma fceRI gamma
  • high affinity immunoglobulin epsilon receptor subunit gamma immunoglobulin E receptor, high affinity, gamma chain; etc.
  • a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
  • MIPAVVLLLLLLVEQAAALGEPQLCYILDAILFLYGIVLTLLYCRLKIQVRKAAITSY EKSDGVYTGLSTRNQETYETLKHEKPPQ (SEQ ID NO: 103), where the ITAM motifs are in bold and are underlined.
  • a suitable intracellular signaling domain polypeptide can comprise an
  • a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to the following amino acid sequence:
  • DGVYTGLSTRNOETYETLKHE (SEQ ID NO: 104), where the ITAM motifs are in bold and are underlined.
  • the intracellular signaling domain is derived from T-cell surface glycoprotein CD3 delta chain (also known as CD3D; CD3-DELTA; T3D; CD3 antigen, delta subunit; CD3 delta; CD3d antigen, delta polypeptide (TiT3 complex); OKT3, delta chain; T-cell receptor T3 delta chain; T-cell surface glycoprotein CD3 delta chain; etc.).
  • T-cell surface glycoprotein CD3 delta chain also known as CD3D; CD3-DELTA; T3D; CD3 antigen, delta subunit; CD3 delta; CD3d antigen, delta polypeptide (TiT3 complex); OKT3, delta chain; T-cell receptor T3 delta chain; T-cell surface glycoprotein CD3 delta chain; etc.
  • a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, or from about 150 aa to about 170 aa, of either of the following amino acid sequences (2 isoforms):
  • a suitable intracellular signaling domain polypeptide can comprise an
  • a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to the following amino acid sequence:
  • DOVYOPLRDRDDAOYSHLGGN SEQ ID NO: 107
  • ITAM motifs are in bold and are underlined.
  • the intracellular signaling domain is derived from T-cell surface
  • glycoprotein CD3 epsilon chain also known as CD3e, T-cell surface antigen T3/Leu-4 epsilon chain, T-cell surface glycoprotein CD3 epsilon chain, AI504783, CD3, CD3epsilon, T3e, etc.
  • a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, or from about 150 aa to about 205 aa, of the following amino acid sequence:
  • a suitable intracellular signaling domain polypeptide can comprise an
  • a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to the following amino acid sequence:
  • NPDYEPIRKGQRDLYSGLNQR SEQ ID NO: 109
  • ITAM motifs are in bold and are underlined.
  • the intracellular signaling domain is derived from T-cell surface
  • a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, or from about 150 aa to about 180 aa, of the following amino acid sequence:
  • a suitable intracellular signaling domain polypeptide can comprise an
  • a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to the following amino acid sequence:
  • DOLYOPLKDREDDOYSHLOGN SEQ ID NO: 111
  • ITAM motifs are in bold and are underlined.
  • the intracellular signaling domain is derived from T-cell surface
  • a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, or from about 150 aa to about 160 aa, of either of the following amino acid sequences (2 isoforms):
  • a suitable intracellular signaling domain polypeptide can comprise an
  • a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to any of the following amino acid sequences:
  • DGLYOGLSTATKDTYDALHMO SEQ ID NO: 116
  • ITAM motifs are in bold and are underlined.
  • the intracellular signaling domain is derived from CD79A (also
  • B-cell antigen receptor complex-associated protein alpha chain CD79a antigen (immunoglobulin-associated alpha); MB-1 membrane glycoprotein; ig-alpha; membrane- bound immunoglobulin-associated protein; surface IgM-associated protein; etc.).
  • a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 150 aa, from about 150 aa to about 200 aa, or from about 200 aa to about 220 aa, of either of the following amino acid sequences (2 isoforms):
  • a suitable intracellular signaling domain polypeptide can comprise an
  • a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to the following amino acid sequence: ENLYEGLNLDDCSMYEDISRG (SEQ ID NO: 119), where the ITAM motifs are in bold and are underlined.
  • Intracellular signaling domains suitable for use in a CAR of the present disclosure include a DAP10/CD28 type signaling chain.
  • DAP 10 signaling chain is the amino acid sequence is:
  • a suitable intracellular signaling domain comprises an amino acid sequence having at least about 85%, at least about 90%, at least about 95%, at least about 98%, or at least about 99%, amino acid sequence identity to the entire length of the amino acid sequence
  • CD28 signaling chain is the amino acid sequence
  • a suitable intracellular signaling domain comprises an amino acid sequence having at least about 85%, at least about 90%, at least about 95%, at least about 98%, or at least about 99%, amino acid sequence identity to the entire length of the amino acid sequence
  • PYAPPRDFAAYRS (SEQ ID NO: 121).
  • Intracellular signaling domains suitable for use in a CAR of the present disclosure include a ZAP70 polypeptide, e.g., a polypeptide comprising an amino acid sequence having at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, or 100%, amino acid sequence identity to a contiguous stretch of from about 300 amino acids to about 400 amino acids, from about 400 amino acids to about 500 amino acids, or from about 500 amino acids to 619 amino acids, of the following amino acid sequence:
  • TQKAEAACA (SEQ ID NO:36).
  • the first and/or the second polypeptide of a subject CAR can further include one or more additional polypeptide domains, where such domains include, but are not limited to, a signal sequence; an epitope tag; an affinity domain; and a polypeptide that produces a detectable signal.
  • polypeptide of a subject CAR include any eukaryotic signal sequence, including a naturally-occurring signal sequence, a synthetic (e.g., man-made) signal sequence, etc.
  • Suitable epitope tags include, but are not limited to, hemagglutinin (HA; e.g., HA;
  • YPYDVPDYA (SEQ ID NO: 122); FLAG (e.g., DYKDDDDK (SEQ ID NO: 123); c-myc (e.g., EQKLISEEDL; SEQ ID NO:4), and the like.
  • Affinity domains include peptide sequences that can interact with a binding partner, e.g., such as one immobilized on a solid support, useful for identification or purification.
  • DNA sequences encoding multiple consecutive single amino acids, such as histidine, when fused to the expressed protein, may be used for one-step purification of the recombinant protein by high affinity binding to a resin column, such as nickel sepharose.
  • Exemplary affinity domains include His5 (HHHHH) (SEQ ID NO: 124), HisX6 (HHHHHH) (SEQ ID NO: 125), C-myc (EQKLISEEDL) (SEQ ID NO:4), Flag (DYKDDDDK) (SEQ ID NO:
  • HA Tag e.g., HA Tag
  • YPYDVPDYA (SEQ ID NO: 122), GST, thioredoxin, cellulose binding domain, RYIRS (SEQ ID NO: 127), Phe-His-His-Thr (SEQ ID NO: 128), chitin binding domain, S-peptide, T7 peptide, SH2 domain, C-end RNA tag, WEAAAREACCRECCARA (SEQ ID NO: 129), metal binding domains, e.g., zinc binding domains or calcium binding domains such as those from calcium-binding proteins, e.g., calmodulin, troponin C, calcineurin B, myosin light chain, recoverin, S-modulin, visinin, VILIP, neurocalcin, hippocalcin, frequenin, caltractin, calpain large- subunit, S100 proteins, parvalbumin, calbindin D9K, calbindin D28K, and calretinin, inteins, biotin
  • Suitable detectable signal-producing proteins include, e.g., fluorescent proteins; enzymes that catalyze a reaction that generates a detectable signal as a product; and the like.
  • Suitable fluorescent proteins include, but are not limited to, green fluorescent protein
  • GFP blue fluorescent variant of GFP
  • BFP blue fluorescent variant of GFP
  • CFP yellow fluorescent variant of GFP
  • YFP yellow fluorescent variant of GFP
  • EGFP enhanced GFP
  • ECFP enhanced CFP
  • EYFP enhanced YFP
  • GFPS65T Emerald, Topaz (TYFP)
  • Citrine mCitrine
  • GFPuv destabilised EGFP
  • dECFP destabilised ECFP
  • EYFP destabilised EYFP
  • mCFPm Cerulean, T-Sapphire, CyPet, YPet, mKO, HcRed, t-HcRed, DsRed, DsRed2, DsRed-monomer, J-Red, dimer2, t-dimer2(12), mRFPl, pocilloporin, Renilla GFP, Monster GFP, paGFP, Kaede protein and kindling protein, Phycobiliproteins and
  • Suitable enzymes include, but are not limited to, horse radish peroxidase (HRP), alkaline phosphatase (AP), beta-galactosidase (GAL), glucose-6-phosphate dehydrogenase, beta-N-acetylglucosaminidase, ⁇ -glucuronidase, invertase, Xanthine Oxidase, firefly luciferase, glucose oxidase (GO), and the like.
  • HRP horse radish peroxidase
  • AP alkaline phosphatase
  • GAL beta-galactosidase
  • glucose-6-phosphate dehydrogenase beta-N-acetylglucosaminidase
  • ⁇ -glucuronidase invertase
  • Xanthine Oxidase firefly luciferase
  • glucose oxidase GO
  • sequences of the polypeptides of a CAR may be rearranged or deleted in a cell through the use of site- specific recombination technology.
  • the cellular activation-related response to a particular CAR can be changed by site-specific recombination, e.g., a first intracellular signaling domain of a CAR eliciting a first activation-related response may be exchanged for a second intracellular signaling domain eliciting a second activation-related response.
  • the response to a particular dimerizer of a CAR can be changed by site- specific recombination, e.g., a first dimerizer-binding pair causing the dimerization of a CAR in the presence of a first dimerizer may be exchanged for a second dimerizer-binding pair causing the dimerization of the CAR in the presence of a second dimerizer.
  • site-specific recombination can be used in a cell to exchange any domain or sequence of a CAR with any other domain or sequence as disclosed herein.
  • site-specific recombination can be used in a cell to delete any domain or sequence of a CAR.
  • the present disclosure provides a nucleic acid that comprises a nucleotide sequence encoding the first and/or the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure.
  • a nucleic acid comprising a nucleotide sequence encoding the first and/or the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure will in some embodiments be DNA, including, e.g., a recombinant expression vector.
  • a nucleic acid comprising a nucleotide sequence encoding the first and/or the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure will in some embodiments be RNA, e.g., in vitro synthesized RNA.
  • a nucleic acid of the present disclosure comprises a nucleotide
  • nucleic acid of the present disclosure comprises a nucleotide sequence encoding only the second polypeptide (and not the first polypeptide) of a heterodimeric, conditionally active CAR of the present disclosure.
  • a nucleic acid of the present disclosure comprises a nucleotide sequence encoding both the first polypeptide and the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure.
  • a subject nucleic acid provides for production of a CAR of the
  • a subject nucleic acid provides for amplification of the CAR-encoding nucleic acid.
  • a nucleotide sequence encoding the first and/or the second polypeptide of a CAR of the present disclosure can be operably linked to a transcriptional control element, e.g., a promoter, and enhancer, etc.
  • Suitable promoter and enhancer elements are known in the art.
  • suitable promoters include, but are not limited to, lacl, lacZ, T3, T7, gpt, lambda P and trc.
  • suitable promoters include, but are not limited to, light and/or heavy chain immunoglobulin gene promoter and enhancer elements; cytomegalovirus immediate early promoter; herpes simplex virus thymidine kinase promoter; early and late SV40 promoters; promoter present in long terminal repeats from a retrovirus; mouse metallothionein-I promoter; and various art-known tissue specific promoters.
  • Suitable reversible promoters including reversible inducible promoters are known in the art. Such reversible promoters may be isolated and derived from many organisms, e.g., eukaryotes and prokaryotes. Modification of reversible promoters derived from a first organism for use in a second organism, e.g., a first prokaryote and a second a eukaryote, a first eukaryote and a second a prokaryote, etc., is well known in the art.
  • Such reversible promoters, and systems based on such reversible promoters but also comprising additional control proteins include, but are not limited to, alcohol regulated promoters (e.g., alcohol dehydrogenase I (alcA) gene promoter, promoters responsive to alcohol transactivator proteins (AlcR), etc.), tetracycline regulated promoters, (e.g., promoter systems including Tet Activators, TetON, TetOFF, etc.), steroid regulated promoters (e.g., rat glucocorticoid receptor promoter systems, human estrogen receptor promoter systems, retinoid promoter systems, thyroid promoter systems, ecdysone promoter systems, mifepristone promoter systems, etc.), metal regulated promoters (e.g., metallothionein promoter systems, etc.), pathogenesis-related regulated promoters (e.g., salicylic acid regulated promoters, ethylene regulated promoter
  • promoter is irreversibly switched through the induction of an inducible system.
  • Suitable systems for induction of an irreversible switch are well known in the art, e.g., induction of an irreversible switch may make use of a Cre-lox-mediated recombination (see, e.g., Fuhrmann-Benzakein, et al., PNAS (2000) 28:e99, the disclosure of which is incorporated herein by reference). Any suitable combination of recombinase, endonuclease, ligase, recombination sites, etc. known to the art may be used in generating an irreversibly switchable promoter.
  • the promoter is a CD8 cell-specific promoter, a CD4 cell-specific promoter, a neutrophil- specific promoter, or an NK-specific promoter.
  • a CD4 gene promoter can be used; see, e.g., Salmon et al. (1993) Proc. Natl. Acad. Sci. USA 90:7739; and Marodon et al. (2003) Blood 101:3416.
  • a CD8 gene promoter can be used.
  • NK cell-specific expression can be achieved by use of an Ncrl (p46) promoter; see, e.g., Eckelhart et al. (2011) Blood 117: 1565.
  • a suitable promoter is a constitutive promoter such as an ADH1 promoter, a PGK1 promoter, an ENO promoter, a PYKl promoter and the like; or a regulatable promoter such as a GALl promoter, a GAL10 promoter, an ADH2 promoter, a PH05 promoter, a CUP1 promoter, a GAL7 promoter, a MET25 promoter, a MET3 promoter, a CYC1 promoter, a HIS 3 promoter, an ADH1 promoter, a PGK promoter, a GAPDH promoter, an ADC1 promoter, a TRP1 promoter, a URA3 promoter, a LEU2 promoter, an ENO promoter, a TP1 promoter, and AOX1 (e.g., for use in Pichia). Selection of the appropriate vector and promoter is a constitutive promoter such as an ADH1 promoter, a PGK1 promote
  • Suitable promoters for use in prokaryotic host cells include, but are not limited to, a bacteriophage T7 RNA polymerase promoter; a trp promoter; a lac operon promoter; a hybrid promoter, e.g., a lac/tac hybrid promoter, a tac/trc hybrid promoter, a trp/lac promoter, a T7/lac promoter; a trc promoter; a tac promoter, and the like; an araBAD promoter; in vivo regulated promoters, such as an ssaG promoter or a related promoter ⁇ see, e.g., U.S. Patent Publication No.
  • a pagC promoter (Pulkkinen and Miller, J. Bacteriol., 1991: 173(1): 86-93; Alpuche-Aranda et al., PNAS, 1992; 89(21): 10079-83), a nirB promoter (Harborne et al. (1992) Mol. Micro. 6:2805-2813), and the like ⁇ see, e.g., Dunstan et al. (1999) Infect. Immun. 67:5133-5141; McKelvie et al. (2004) Vaccine
  • a sigma70 promoter e.g., a consensus sigma70 promoter (see, e.g., GenBank Accession Nos. AX798980, AX798961, and AX798183); a stationary phase promoter, e.g., a dps promoter, an spv promoter, and the like; a promoter derived from the pathogenicity island SPI-2 ⁇ see, e.g., W096/17951); an actA promoter ⁇ see, e.g., Shetron-Rama et al. (2002) Infect. Immun.
  • Suitable strong promoters for use in prokaryotes such as Escherichia coli include, but are not limited to Trc, Tac, T5, T7, and PLambda.
  • operators for use in bacterial host cells include a lactose promoter operator (Lacl repressor protein changes conformation when contacted with lactose, thereby preventing the Lacl repressor protein from binding to the operator), a tryptophan promoter operator (when complexed with tryptophan, TrpR repressor protein has a conformation that binds the operator; in the absence of tryptophan, the TrpR repressor protein has a conformation that does not bind to the operator), and a tac promoter operator (see, for example, deBoer et al. (1983) Proc. Natl. Acad. Sci. U.S.A. 80:21-25).
  • a nucleotide sequence encoding a subject CAR can be present in an expression vector and/or a cloning vector. Where a subject CAR comprises two separate polypeptides, nucleotide sequences encoding the two polypeptides can be cloned in the same or separate vectors.
  • An expression vector can include a selectable marker, an origin of replication, and other features that provide for replication and/or maintenance of the vector. Suitable expression vectors include, e.g., plasmids, viral vectors, and the like.
  • vectors and promoters are known to those of skill in the art; many are commercially available for generating a subject recombinant constructs.
  • the following vectors are provided by way of example.
  • Bacterial pBs, phagescript, PsiX174, pBluescript SK, pBs KS, pNH8a, pNH16a, pNH18a, pNH46a (Stratagene, La Jolla, Calif., USA); pTrc99A, pKK223-3, pKK233-3, pDR540, and pRIT5 (Pharmacia, Uppsala, Sweden).
  • Eukaryotic pWLneo, pSV2cat, pOG44, PXR1, pSG (Stratagene) pSVK3, pBPV, pMSG and pSVL (Pharmacia).
  • Expression vectors generally have convenient restriction sites located near the
  • Suitable expression vectors include, but are not limited to, viral vectors (e.g. viral vectors based on vaccinia virus; poliovirus; adenovirus (see, e.g., Li et al., Invest Opthalmol Vis Sci 35:2543 2549, 1994; Borras et al., Gene Ther 6:515 524, 1999; Li and Davidson, PNAS 92:7700 7704, 1995; Sakamoto et al., H Gene Ther 5: 1088 1097, 1999; WO 94/12649, WO 93/03769; WO 93/19191; WO 94/28938; WO 95/11984 and WO 95/00655); adeno- associated virus (see, e.g., Ali et al., Hum Gene Ther 9:81 86,
  • a retroviral vector e.g., Murine Leukemia Virus, s
  • a nucleic acid comprising a nucleotide
  • RNA sequence encoding the first and/or the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure will in some embodiments be RNA, e.g., in vitro synthesized RNA.
  • Methods for in vitro synthesis of RNA are known in the art; any known method can be used to synthesize RNA comprising a nucleotide sequence encoding the first and/or the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure.
  • Methods for introducing RNA into a host cell are known in the art. See, e.g., Zhao et al. (2010) Cancer Res. 15:9053.
  • RNA comprising a nucleotide sequence encoding the first and/or the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure into a host cell can be carried out in vitro or ex vivo or in vivo.
  • a host cell e.g., an NK cell, a cytotoxic T lymphocyte, etc.
  • RNA comprising a nucleotide sequence encoding the first and/or the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure.
  • the present disclosure provides a mammalian cell that is genetically modified to produce a heterodimeric, conditionally active CAR of the present disclosure.
  • Suitable mammalian cells include primary cells and immortalized cell lines.
  • Suitable mammalian cell lines include human cell lines, non-human primate cell lines, rodent (e.g., mouse, rat) cell lines, and the like.
  • Suitable mammalian cell lines include, but are not limited to, HeLa cells (e.g., American Type Culture Collection (ATCC) No. CCL-2), CHO cells (e.g., ATCC Nos. CRL9618, CCL61, CRL9096), 293 cells (e.g., ATCC No. CRL-1573), Vero cells, NIH 3T3 cells (e.g., ATCC No. CRL-1658), Huh-7 cells, BHK cells (e.g., ATCC No. CCL10), PC12 cells (ATCC No.
  • HeLa cells e.g., American Type Culture Collection (ATCC) No. CCL-2
  • CHO cells e.g., ATCC Nos. CRL9618, CCL61, CRL9096
  • 293 cells e.g., ATCC No. CRL
  • CRL1721) COS cells, COS-7 cells (ATCC No. CRL1651), RATI cells, mouse L cells (ATCC No. CCLI.3), human embryonic kidney (HEK) cells (ATCC No. CRL1573), HLHepG2 cells, Hut-78, Jurkat, HL-60, NK cell lines (e.g., NKL, NK92, and YTS), and the like.
  • the cell is not an immortalized cell line, but is instead a cell (e.g., a primary cell) obtained from an individual.
  • a cell e.g., a primary cell
  • the cell is an immune cell obtained from an individual.
  • the cell is a T lymphocyte obtained from an individual.
  • the cell is a cytotoxic cell obtained from an individual.
  • the cell is a stem cell or progenitor cell obtained from an individual.
  • the present disclosure provides methods of activating an immune cell in vitro, in vivo, or ex vivo.
  • the methods generally involve contacting an immune cell (in vitro, in vivo, or ex vivo) with a dimerizing agent and an antigen, where the immune cell is genetically modified to produce a heterodimeric, conditionally active CAR of the present disclosure.
  • the heterodimeric, conditionally active CAR dimerizes and activates the immune cell, thereby producing an activated immune cell.
  • Immune cells include, e.g., a cytotoxic T lymphocyte, an NK cell, a CD4 + T cell, a T regulatory (Treg) cell, etc.
  • a cytokine by the immune cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared with the amount of cytokine produced by the immune cell in the absence of the second member of a specific binding pair and/or the dimerizing agent.
  • Cytokines whose production can be increased include, but are not limited to, IL-2 and IFN- ⁇ .
  • a cytokine by the immune cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2- fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10- fold, compared with the amount of cytokine produced by the immune cell in the absence of the antigen and/or the dimerizing agent.
  • Cytokines whose production can be increased include, but are not limited to, IL-2 and IFN- ⁇ .
  • a genetically modified cytotoxic cell e.g., cytotoxic T lymphocyte
  • a dimerizing agent and a second member of a specific binding pair e.g., an antigen, a ligand, a receptor
  • a second member of a specific binding pair e.g., an antigen, a ligand, a receptor
  • cytotoxic activity of the cytotoxic cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared to the cytotoxic activity of the cytotoxic cell in the absence of the dimerizing agent.
  • a genetically modified cytotoxic cell e.g., cytotoxic T lymphocyte
  • a dimerizing agent and an antigen can increase cytotoxic activity of the cytotoxic cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared to the cytotoxic activity of the cytotoxic cell in the absence of the dimerizing agent.
  • contacting a genetically modified host cell with a dimerizing agent and an antigen can increase or decrease cell proliferation, cell survival, cell death, and the like.
  • the present disclosure provides a method of generating a conditionally activatable cell.
  • the method generally involves genetically modifying a mammalian cell with an expression vector, or an RNA (e.g., in vitro transcribed RNA), comprising nucleotide sequences encoding a heterodimeric, conditionally active CAR of the present disclosure.
  • the genetically modified cell is conditionally activatable in the presence of: a) an antigen to which the first polypeptide of the CAR binds; and b) a dimerizer (a dimerizing agent).
  • the genetic modification can be carried out in vivo, in vitro, or ex vivo.
  • the cell can be an immune cell (e.g., a T lymphocyte or NK cell), a stem cell, a progenitor cell, etc.
  • the genetic modification is carried out ex vivo.
  • a T cell for example, a TGF-binding protein
  • lymphocyte, a stem cell, or an NK cell is obtained from an individual; and the cell obtained from the individual is genetically modified to express a CAR of the present disclosure.
  • the genetically modified cell is conditionally activatable in the presence of: a) an antigen to which the first polypeptide of the CAR binds; and b) a dimerizer.
  • the genetically modified cell is activated ex vivo.
  • the genetically modified cell is introduced into an individual (e.g., the individual from whom the cell was obtained); and the genetically modified cell is activated in vivo, e.g., by administering to the individual a dimerizer.
  • the antigen is present on the surface of a cell in the individual, there is no need to administer the antigen.
  • the genetically modified cell comes into contact with the antigen present on the surface of a cell in the individual; and, upon administration to the individual of a dimerizer, the genetically modified cell is activated.
  • the genetically modified cell is a T lymphocyte, the genetically modified cell can exhibit cytotoxicity toward a cell that presents an antigen on its surface to which the CAR binds.
  • the present disclosure provides various treatment methods using a subject CAR.
  • a CAR of the present disclosure when present in a T lymphocyte or an NK cell, can mediate cytotoxicity toward a target cell.
  • a CAR of the present disclosure binds to an antigen present on a target cell, thereby mediating killing of a target cell by a T lymphocyte or an NK cell genetically modified to produce the CAR.
  • the antigen-binding domain of the CAR binds to an antigen present on the surface of a target cell.
  • Target cells include, but are not limited to, cancer cells.
  • a cytotoxic immune effector cell e.g., a cytotoxic T cell, or an NK cell
  • a subject CAR e.g., a cytotoxic T cell, or an NK cell
  • the present disclosure provides a method of treating cancer in an individual having a cancer, the method comprising: i) genetically modifying T lymphocytes obtained from the individual with an expression vector comprising nucleotide sequences encoding the heterodimeric, conditionally active CAR of the present disclosure, where the antigen- binding domain of the heterodimeric, conditionally active CAR is specific for an epitope on a cancer cell in the individual, and where the genetic modification is carried out ex vivo; ii) introducing the genetically modified T lymphocytes into the individual; and iii)
  • dimerizing agent induces dimerization of the heterodimeric, conditionally active CAR, wherein said dimerization provides for activation of the genetically modified T lymphocytes and killing of the cancer cell, thereby treating the cancer.
  • Carcinomas that can be amenable to therapy by a method disclosed herein include, but are not limited to, esophageal carcinoma, hepatocellular carcinoma, basal cell carcinoma (a form of skin cancer), squamous cell carcinoma (various tissues), bladder carcinoma, including transitional cell carcinoma (a malignant neoplasm of the bladder), bronchogenic carcinoma, colon carcinoma, colorectal carcinoma, gastric carcinoma, lung carcinoma, including small cell carcinoma and non-small cell carcinoma of the lung, adrenocortical carcinoma, thyroid carcinoma, pancreatic carcinoma, breast carcinoma, ovarian carcinoma, prostate carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, renal cell carcinoma, ductal carcinoma in situ or bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical carcinoma, uterine carcinoma
  • Sarcomas that can be amenable to therapy by a method disclosed herein include, but are not limited to, fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, chordoma, osteogenic sarcoma, osteosarcoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's sarcoma,
  • Other solid tumors that can be amenable to therapy by a method disclosed herein include, but are not limited to, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblastoma, and retinoblastoma.
  • Leukemias that can be amenable to therapy by a method disclosed herein include, but are not limited to, a) chronic myeloproliferative syndromes (neoplastic disorders of multipotential hematopoietic stem cells); b) acute myelogenous leukemias (neoplastic transformation of a multipotential hematopoietic stem cell or a hematopoietic cell of restricted lineage potential; c) chronic lymphocytic leukemias (CLL; clonal proliferation of immunologically immature and functionally incompetent small lymphocytes), including B- cell CLL, T-cell CLL prolymphocytic leukemia, and hairy cell leukemia; and d) acute lymphoblastic leukemias (characterized by accumulation of lymphoblasts).
  • CLL chronic lymphocytic leukemias
  • Lymphomas that can be treated using a subject method include, but are not limited to, B-cell lymphomas (e.g., Burkitt's lymphoma); Hodgkin's lymphoma; non-Hodgkin's lymphoma, and the like.
  • B-cell lymphomas e.g., Burkitt's lymphoma
  • Hodgkin's lymphoma e.g., Hodgkin's lymphoma
  • non-Hodgkin's lymphoma e.g., Hodgkin's lymphoma
  • cancers that can be amenable to treatment according to the methods disclosed herein include atypical meningioma (brain), islet cell carcinoma (pancreas), medullary carcinoma (thyroid), mesenchymoma (intestine), hepatocellular carcinoma (liver), hepatoblastoma (liver), clear cell carcinoma (kidney), and neurofibroma mediastinum.
  • a subject method can also be used to treat inflammatory conditions and autoimmune disease.
  • a subject CAR is expressed in a T-helper cell or a Tregs for use in an
  • Immunomodulatory methods include, e.g., enhancing an immune response in a mammalian subject toward a pathogen; enhancing an immune response in a subject who is immunocompromised; reducing an inflammatory response; reducing an immune response in a mammalian subject to an autoantigen, e.g., to treat an autoimmune disease; and reducing an immune response in a mammalian subject to a transplanted organ or tissue, to reduce organ or tissue rejection.
  • the antigen used to activate the CAR is an autoantigen.
  • the antigen used to activate the CAR is an antigen specific to the transplanted organ.
  • administration to an individual in need thereof of an effective amount of a dimerizer agent may also involve administration of an antigen.
  • an "effective amount" of a dimerizer agent is in some cases an amount that, when administered in one or more doses to an individual in need thereof, increases the level of cytotoxic activity of a T lymphocyte expressing a subject CAR by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared to the cytotoxic activity of the T lymphocyte in the absence of the dimerizing agent.
  • an "effective amount" of a dimerizer agent is in some cases an amount that, when administered in one or more doses to an individual in need thereof, increases the level of cytotoxic activity of an NK cell expressing a subject CAR by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared to the cytotoxic activity of the NK cell in the absence of the dimerizing agent.
  • an "effective amount" of a dimerizer agent is in some cases an amount that, when administered in one or more doses to an individual in need thereof, reduces the number of cancer cells in the individual and/or reduces tumor mass in the individual, by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, or more than 75%, compared to the number of cancer cells and/or tumor mass in the absence of the dimerizing agent.
  • an effective amount of a dimerizer is an amount that, when administered alone (e.g., in monotherapy) or in combination (e.g., in combination therapy) with one or more additional therapeutic agents, in one or more doses, is effective to reduce one or more of tumor growth rate, cancer cell number, and tumor mass, by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more, compared to the tumor growth rate, cancer cell number, or tumor mass in the absence of treatment with the dimerizer.
  • a dimerizer can be administered to the host using any combination of formula (1) and any combination of formula (1) and any combination of formula (2) and any combination of formula (3) and any combination of formula (3) and any combination of formula (3) and any combination of formula (3) and any combination of formula (3) and any combination of formula (3) and any combination of formula (3) and any combination of formula (3) and any combination of formula (3) and any combination of formula (3) and any combination of formula (3) and any combination of formula (3) and any combination of formula (3) and any combination of formula (Ca)-2-aminoethyl-N-(2-aminoethyl)-2-a dimerizer, or any combination of the following compounds described above-a dimerizers thereof.
  • the dimerizer can be incorporated into a variety of formulations for therapeutic administration. More particularly, a dimerizer can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants and aerosols.
  • a dimerizer in pharmaceutical dosage forms, can be administered in the form of their pharmaceutically acceptable salts, or they may also be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds.
  • the following methods and excipients are merely exemplary and are in no way limiting.
  • Suitable excipient vehicles are, for example, water, saline, dextrose, glycerol,
  • the vehicle may contain minor amounts of auxiliary substances such as wetting or emulsifying agents or pH buffering agents.
  • auxiliary substances such as wetting or emulsifying agents or pH buffering agents.
  • Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art. See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pennsylvania, 17th edition, 1985.
  • the composition or formulation to be administered will, in any event, contain a quantity of a dimerizer adequate to achieve the desired state in the subject being treated.
  • compositions such as vehicles, adjuvants, carriers or diluents
  • pharmaceutically acceptable auxiliary substances such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are readily available to the public.
  • a dimerizer for oral preparations, can be used alone or in combination with
  • appropriate additives to make tablets, powders, granules or capsules for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
  • conventional additives such as lactose, mannitol, corn starch or potato starch
  • binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins
  • disintegrators such as corn starch, potato starch or sodium carboxymethylcellulose
  • lubricants such as talc or magnesium stearate
  • diluents buffering agents, moistening agents
  • a dimerizer can be formulated into preparations for injection by dissolving
  • suspending or emulsifying them in an aqueous or nonaqueous solvent such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
  • compositions comprising a dimerizer are prepared by mixing the dimerizer having the desired degree of purity with optional physiologically acceptable carriers, excipients, stabilizers, surfactants, buffers and/or tonicity agents.
  • Acceptable carriers, excipients and/or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid, glutathione, cysteine, methionine and citric acid; preservatives (such as ethanol, benzyl alcohol, phenol, m-cresol, p-chlor-m-cresol, methyl or propyl parabens, benzalkonium chloride, or combinations thereof); amino acids such as arginine, glycine, ornithine, lysine, histidine, glutamic acid, aspartic acid, isoleucine, leucine, alanine, phenylalanine, tyros
  • the pharmaceutical composition may be in a liquid form, a lyophilized form or a liquid form reconstituted from a lyophilized form, wherein the lyophilized preparation is to be reconstituted with a sterile solution prior to administration.
  • the standard procedure for reconstituting a lyophilized composition is to add back a volume of pure water (typically equivalent to the volume removed during lyophilization); however solutions comprising antibacterial agents may be used for the production of pharmaceutical compositions for parenteral administration; see also Chen (1992) Drug Dev Ind Pharm 18, 1311-54.
  • unit dosage form refers to physically discrete units
  • a dimerizer is formulated in a controlled release formulation.
  • Sustained-release preparations may be prepared using methods well known in the art.
  • sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the dimerizer in which the matrices are in the form of shaped articles, e.g. films or microcapsules.
  • sustained-release matrices include polyesters, copolymers of L-glutamic acid and ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, hydrogels, polylactides, degradable lactic acid-glycolic acid copolymers and poly-D-(-)-3-hydroxybutyric acid. Possible loss of biological activity may be prevented by using appropriate additives, by controlling moisture content and by developing specific polymer matrix compositions.
  • a suitable dosage can be determined by an attending physician or other qualified medical personnel, based on various clinical factors. As is well known in the medical arts, dosages for any one patient depend upon many factors, including the patient's size, body surface area, age, the particular dimerizer to be administered, sex of the patient, time, and route of administration, general health, and other drugs being administered concurrently.
  • a dimerizer may be administered in amounts between 1 ng/kg body weight and 20 mg/kg body weight per dose, e.g. between 0.1 mg/kg body weight to 10 mg/kg body weight, e.g. between 0.5 mg/kg body weight to 5 mg/kg body weight; however, doses below or above this exemplary range are envisioned, especially considering the aforementioned factors. If the regimen is a continuous infusion, it can also be in the range of 1 ⁇ g to 10 mg per kilogram of body weight per minute.
  • dose levels can vary as a function of the specific dimerizer, the severity of the symptoms and the susceptibility of the subject to side effects. Preferred dosages for a given compound are readily determinable by those of skill in the art by a variety of means.
  • a dimerizer is administered to an individual using any available method and route suitable for drug delivery, including in vivo and ex vivo methods, as well as systemic and localized routes of administration.
  • routes of administration include intratumoral, peritumoral, intramuscular, intratracheal, intracranial, subcutaneous, intradermal, topical application, intravenous, intraarterial, rectal, nasal, oral, and other enteral and parenteral routes of administration. Routes of administration may be combined, if desired, or adjusted depending upon the dimerizer and/or the desired effect.
  • a dimerizer can be administered in a single dose or in multiple doses. In some embodiments, a dimerizer is administered orally. In some embodiments, a dimerizer is administered via an inhalational route. In some embodiments, a dimerizer is administered intranasally. In some
  • a dimerizer is administered locally. In some embodiments, a dimerizer is administered intratumorally. In some embodiments, a dimerizer is administered
  • a dimerizer is administered intracranially. In some embodiments, a dimerizer is administered intravenously.
  • the agent can be administered to a host using any available conventional methods and routes suitable for delivery of conventional drugs, including systemic or localized routes.
  • routes of administration contemplated by the invention include, but are not necessarily limited to, enteral, parenteral, or inhalational routes.
  • Parenteral routes of administration other than inhalation administration include, but are not necessarily limited to, topical, transdermal, subcutaneous, intramuscular,
  • intraorbital intracapsular, intraspinal, intrasternal, intratumoral, peritumoral, and
  • parenteral administration can be carried to effect systemic or local delivery of a dimerizer. Where systemic delivery is desired, administration typically involves invasive or systemically absorbed topical or mucosal administration of pharmaceutical preparations.
  • a dimerizer can also be delivered to the subject by enteral administration.
  • Enteral routes of administration include, but are not necessarily limited to, oral and rectal ⁇ e.g., using a suppository) delivery.
  • treatment is meant at least an amelioration of the symptoms associated with the pathological condition afflicting the host, where amelioration is used in a broad sense to refer to at least a reduction in the magnitude of a parameter, e.g. symptom, associated with the pathological condition being treated, such as cancer.
  • amelioration also includes situations where the pathological condition, or at least symptoms associated therewith, are completely inhibited, e.g. prevented from happening, or stopped, e.g. terminated, such that the host no longer suffers from the pathological condition, or at least the symptoms that characterize the pathological condition.
  • a dimerizer is administered by injection and/or delivery, e.g., to a site in a brain artery or directly into brain tissue.
  • a dimerizer can also be administered directly to a target site e.g., by direct injection, by implantation of a drug delivery device such as an osmotic pump or slow release particle, by biolistic delivery to the target site, etc.
  • a dimerizer is administered as an adjuvant therapy to a patient.
  • Standard cancer therapies include surgery (e.g., surgical removal of cancerous tissue), radiation therapy, bone marrow transplantation, chemotherapeutic treatment, antibody treatment, biological response modifier treatment, and certain combinations of the foregoing.
  • Radiation therapy includes, but is not limited to, x-rays or gamma rays that are
  • Suitable antibodies for use in cancer treatment include, but are not limited to, naked antibodies, e.g., trastuzumab (Herceptin) , bevacizumab (AvastinTM), cetuximab
  • Lambrolizumab (MK-3475), pertuzumab (PerjetaTM), ranibizumab (LucentisTM) etc., and conjugated antibodies, e.g., gemtuzumab ozogamicin (MylortargTM), Brentuximab vedotin (AdcetrisTM), 90 Y-labelled ibritumomab tiuxetan (ZevalinTM), 131 I-labelled tositumoma (BexxarTM), etc.
  • Suitable antibodies for use in cancer treatment include, but are not limited to, antibodies raised against tumor- associated antigens.
  • Such antigens include, but are not limited to, CD20, CD30, CD33, CD52, EpCAM, CEA, gpA33, Mucins, TAG-72, CAIX, PSMA, Folate-binding protein, Gangliosides (e.g., GD2, GD3, GM2, etc.), Le y , VEGF, VEGFR, Integrin alpha- V-beta-3, Integrin alpha-5-beta-l, EGFR, ERBB2, ERBB3, MET, IGF1R, EPHA3, TRAILR1, TRAILR2, RANKL, FAP, Tenascin, etc.
  • Biological response modifiers suitable for use in connection with the methods of the present disclosure include, but are not limited to, (1) inhibitors of tyrosine kinase (RTK) activity; (2) inhibitors of serine/threonine kinase activity; (3) tumor-associated antigen antagonists, such as antibodies that bind specifically to a tumor antigen; ( 4) apoptosis receptor agonists; (5) interleukin-2; (6) interferon-a.; (7) interferon - ⁇ ; (8) colony- stimulating factors; (9) inhibitors of angiogenesis; and (10) antagonists of tumor necrosis factor.
  • RTK tyrosine kinase
  • Chemotherapeutic agents are non-peptidic (i.e., non-proteinaceous) compounds that reduce proliferation of cancer cells, and encompass cytotoxic agents and cytostatic agents.
  • Non-limiting examples of chemotherapeutic agents include alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, plant (vinca) alkaloids, and steroid hormones.
  • Agents that act to reduce cellular proliferation are known in the art and widely used.
  • Such agents include alkylating agents, such as nitrogen mustards, nitrosoureas, ethylenimine derivatives, alkyl sulfonates, and triazenes, including, but not limited to, mechlorethamine, cyclophosphamide (CytoxanTM), melphalan (L-sarcolysin), carmustine (BCNU), lomustine (CCNU), semustine (methyl-CCNU), streptozocin, chlorozotocin, uracil mustard, chlormethine, ifosfamide, chlorambucil, pipobroman, triethylenemelamine,
  • alkylating agents such as nitrogen mustards, nitrosoureas, ethylenimine derivatives, alkyl sulfonates, and triazenes, including, but not limited to, mechlorethamine, cyclophosphamide (CytoxanTM), melphalan (L-sarcolysin),
  • triethylenethiophosphoramine busulfan, dacarbazine, and temozolomide.
  • Antimetabolite agents include folic acid analogs, pyrimidine analogs, purine
  • adenosine deaminase inhibitors including, but not limited to, cytarabine (CYTOSAR-U), cytosine arabinoside, fluorouracil (5-FU), floxuridine (FudR), 6- thioguanine, 6-mercaptopurine (6-MP), pentostatin, 5 -fluorouracil (5-FU), methotrexate, 10-propargyl-5,8-dideazafolate (PDDF, CB3717), 5,8-dideazatetrahydrofolic acid
  • Suitable natural products and their derivatives e.g., vinca alkaloids, antitumor
  • antibiotics include, but are not limited to, Ara-C, paclitaxel (Taxol®), docetaxel (Taxotere®), deoxycoformycin, mitomycin-C, L- asparaginase, azathioprine; brequinar; alkaloids, e.g. vincristine, vinblastine, vinorelbine, vindesine, etc.; podophyllotoxins, e.g. etoposide, teniposide, etc.; antibiotics, e.g.
  • anthracycline daunorubicin hydrochloride (daunomycin, rubidomycin, cerubidine), idarubicin, doxorubicin, epirubicin and morpholino derivatives, etc.; phenoxizone biscyclopeptides, e.g. dactinomycin; basic glycopeptides, e.g. bleomycin; anthraquinone glycosides, e.g. plicamycin (mithramycin); anthracenediones, e.g. mitoxantrone;
  • azirinopyrrolo indolediones e.g. mitomycin
  • macrocyclic immunosuppressants e.g.
  • anti-proliferative cytotoxic agents are navelbene, CPT-11, anastrazole, letrazole, capecitabine, reloxafine, cyclophosphamide, ifosamide, and droloxafine.
  • Microtubule affecting agents that have antiproliferative activity are also suitable for use and include, but are not limited to, allocolchicine (NSC 406042), Halichondrin B (NSC 609395), colchicine (NSC 757), colchicine derivatives (e.g., NSC 33410), dolstatin 10 (NSC 376128), maytansine (NSC 153858), rhizoxin (NSC 332598), paclitaxel (Taxol®), Taxol® derivatives, docetaxel (Taxotere®), thiocolchicine (NSC 361792), trityl cysterin, vinblastine sulfate, vincristine sulfate, natural and synthetic epothilones including but not limited to, eopthilone A, epothilone B, discodermolide; estramustine, nocodazole, and the like.
  • Hormone modulators and steroids include, but are not limited to, adrenocorticosteroids, e.g. prednisone, dexamethasone, etc.; estrogens and pregestins, e.g. hydroxyprogesterone caproate, medroxyprogesterone acetate, megestrol acetate, estradiol, clomiphene, tamoxifen; etc.; and adrenocortical suppressants, e.g.
  • adrenocorticosteroids e.g. prednisone, dexamethasone, etc.
  • estrogens and pregestins e.g. hydroxyprogesterone caproate, medroxyprogesterone acetate, megestrol acetate, estradiol, clomiphene, tamoxifen; etc.
  • adrenocortical suppressants e.g.
  • estradiosteroids may inhibit T cell proliferation.
  • chemotherapeutic agents include metal complexes, e.g. cisplatin (cis-DDP), carboplatin, etc.; ureas, e.g. hydroxyurea; and hydrazines, e.g. N-methylhydrazine;
  • epidophyllotoxin a topoisomerase inhibitor
  • procarbazine mitoxantrone
  • leucovorin a topoisomerase inhibitor
  • anti-proliferative agents of interest include immunosuppressants, e.g. mycophenolic acid, thalidomide, desoxyspergualin, azasporine, leflunomide, mizoribine, azaspirane (SKF 105685); Iressa® (ZD 1839, 4-(3-chloro-4-fluorophenylamino)-7- methoxy-6- (3- (4-morpholinyl)propoxy)quinazoline) ; etc .
  • immunosuppressants e.g. mycophenolic acid, thalidomide, desoxyspergualin, azasporine, leflunomide, mizoribine, azaspirane (SKF 105685); Iressa® (ZD 1839, 4-(3-chloro-4-fluorophenylamino)-7- methoxy-6- (3- (4-morpholinyl)propoxy)quinazoline) ; etc .
  • Taxanes include paclitaxel, as well as any active taxane derivative or pro-drug.
  • “Paclitaxel” (which should be understood herein to include analogues, formulations, and derivatives such as, for example, docetaxel, TAXOLTM, TAXOTERETM (a formulation of docetaxel), 10-desacetyl analogs of paclitaxel and 3'N-desbenzoyl-3'N-t-butoxycarbonyl analogs of paclitaxel) may be readily prepared utilizing techniques known to those skilled in the art (see also WO 94/07882, WO 94/07881, WO 94/07880, WO 94/07876, WO
  • Paclitaxel should be understood to refer to not only the common chemically
  • paclitaxel available form of paclitaxel, but analogs and derivatives (e.g., TaxotereTM docetaxel, as noted above) and paclitaxel conjugates (e.g., paclitaxel-PEG, paclitaxel-dextran, or paclitaxel-xylose).
  • analogs and derivatives e.g., TaxotereTM docetaxel, as noted above
  • paclitaxel conjugates e.g., paclitaxel-PEG, paclitaxel-dextran, or paclitaxel-xylose.
  • Taxane also included within the term “taxane” are a variety of known derivatives, including both hydrophilic derivatives, and hydrophobic derivatives. Taxane derivatives include, but not limited to, galactose and mannose derivatives described in International Patent Application No. WO 99/18113; piperazino and other derivatives described in WO
  • a variety of subjects are suitable for treatment with a subject method of treating cancer.
  • suitable subjects include any individual, e.g., a human or non-human animal who has cancer, who has been diagnosed with cancer, who is at risk for developing cancer, who has had cancer and is at risk for recurrence of the cancer, who has been treated with an agent other than a dimerizer for the cancer and failed to respond to such treatment, or who has been treated with an agent other than a dimerizer for the cancer but relapsed after initial response to such treatment.
  • Subjects suitable for treatment with a subject immunomodulatory method include individuals who have an autoimmune disorder; individuals who are organ or tissue transplant recipients; and the like; individuals who are immunocompromised; and individuals who are infected with a pathogen.
  • Standard abbreviations may be used, e.g., bp, base pair(s); kb, kilobase(s); pi, picoliter(s); s or sec, second(s); min, minute(s); h or hr, hour(s); aa, amino acid(s); kb, kilobase(s); bp, base pair(s); nt, nucleotide(s); i.m., intramuscular(ly); i.p., intraperitoneal (ly); s.c, subcutaneous (ly); i.v., intravenous (ly); and the like.
  • Example 1 Generation of CAR
  • FIGS. 18A and 18B summarize the molecular structure of each CAR consisting of two numerically identified polypeptides. All membrane- anchored polypeptides are di-sulfide bonded homo-dimers. The membrane- anchored polypeptides are depicted as monomers for graphical simplicity.
  • Sequence encoding the anti-human CD 19 scFv was cloned from a construct.
  • the human 4- IBB co- stimulation and CD3 zeta ⁇ signaling chains were cloned from cDNAs supplied by Open Biosystems.
  • FKBP- and FRB-encoding sequences were cloned from plasmids supplied by Addgene.
  • Standard molecular cloning techniques (polymerase chain reaction (PCR), restriction digestion, ligation, etc.) were applied to generate lentiviral expression plasmids.
  • telomeres were cultured in human T cell medium, consisting of X-VIV015 (Lonza #04-418Q), 5% human AB serum (Valley Biomedical Inc., #HP1022), lOmM N-acetyl L-Cysteine (Sigma- Aldrich #A9165) and 100 IU/mL recombinant human IL-2 (NCI/BRB Preclinical Repository).
  • a Jurkat cell line expressing the Green Fluorescent Protein (GFP) upon NFAT activation was maintained in RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS), penicillin and streptomycin.
  • K562 target cells from U. Penn were cultured in IMDM supplemented with 10% FBS.
  • Pantropic VSV-G pseudotyped lentivirus was produced from Lenti-X 293T cells
  • Heterodimerizer (Clontech Laboratories #635055) were serially diluted in medium and added to reaction mixtures. After 20-24 hours of incubation, medium supernatants were collected and analyzed with BD OptEIA Human IL-2 ELISA Set (BD Biosciences).
  • the cognate and non-cognate K562 target cells were engineered to express distinct fluorescent proteins so that both cell types in a mixture could be simultaneously quantified by flow cytometry.
  • the target cell types were mixed at a 1: 1 ratio and co-incubated with human primary CD8+ effector T cells at a 5:2 effector: target ratio.
  • 100 IU/mL human IL-2 and varying amounts of the rapalog were added to reaction mixtures. After 24 hours of incubation, samples were centrifuged at 400g for 5 minutes.
  • Pelleted cells were resuspended in wash buffer (PBS + 0.5% BSA + 0.1% sodium azide) and fixed with an equal volume of BD Cytofix (BD cat #554655) prior to flow cytometry. Ratios of the surviving cognate target cells to non-cognate target cells were calculated for each sample to enumerate re-directed cytotoxic activities of the effector cells.
  • Figure 13 IL-2 production by control Jurkat lines in the same experiment as that described in Figure 12. Construct "125" encodes a conventional control currently used in clinical trials.
  • Figure 15 demonstrates pharmacologically titratable cytoxicity conferred by the On- switch CAR "197+206"
  • the CAR effectively mediates re-directed cytotoxicity towards cognate target cells.
  • this On-switch CAR can signal as strongly as the "125" conventional CAR.
  • Effector human primary CD8+ T cells engineered with CARs or a control vector.
  • Target fluorescent derivatives of K562 cell lines expressing either the cognate human CD19 antigen or the non-cognate human mesothelin antigen.
  • Figure 16 depicts data for CARs constructed with the cytoplasmic tyrosine kinase
  • Figure 16 shows data from Jurkat cells engineered with several variants of On- switch CARs.
  • the engineered Jurkat cells were co-incubated with K562 target cells with or without the cognate antigen (CD 19) and the indicated concentrations of rapalog.
  • CD 19 cognate antigen
  • the Zap70 kinase first and second structures from left featuring "199" was as effective as the IT AM (third structure from left featuring "168") in activating NFAT function.
  • Addition of the 4- IBB signaling domain increased surface expression of the antigen recognition portion of the receptor and led to stronger signaling by "197+199".
  • a non-signaling CAR far-right was included as a negative control.
  • FIGS. 19A, 19B, and 19C summarize the molecular structure of each anti-human mesothelin CAR, with each CAR comprising two polypeptides.
  • each anti-human mesothelin CAR comprises two 4- IBB co- stimulatory domains, an FKBP and FRB dimerizer-binding pair, and an IT AM intracellular signaling domain.
  • the three different antigen recognition domains shown here are anti- mesothelin HN1 scFv, SSI scFv, and m912 scFv. All membrane- anchored polypeptides are di-sulfide bonded homo-dimers.
  • Sequences encoding the anti-mesothelin were cloned from constructs or synthesized via gene assembly by PCR.
  • the human 4- IBB co- stimulation and CD3 zeta ⁇ signaling chains were cloned from cDNAs supplied by Open Biosystems.
  • HN1 scFv-, SS I scFv-, and m912 scFv-encoding sequences were synthesized by PCR and, in some cases, codon optimized.
  • FKBP- and FRB -encoding sequences were cloned from Addgene plasmids.
  • Standard molecular cloning techniques (polymerase chain reaction (PCR), restriction digestion, ligation, etc.) were applied to generate lentiviral expression plasmids.
  • K562 target cells were cultured in IMDM supplemented with 10% fetal bovine serum (FBS).
  • FBS fetal bovine serum
  • Pantropic VSV-G pseudotyped lentivirus was produced from Lenti-X 293T cells
  • Jurkat and K562 cells were split 1-2 days in advance to ensure that cultures would be in log phase at the time of transduction. Transduced Jurkat and K562 cells were cultured for at least 7 days before experiments were conducted. Expression levels of CARs encoded in the lentiviral constructs were quantified by detecting either fluorophore-conjugated antibodies or fluorescent reporter proteins using a flow cytometer.
  • K562 target cells at a 1 :2 effectontarget ratio.
  • the rapalog A/C Heterodimerizer (Clontech Laboratories #635055) were serially diluted in medium and added to reaction mixtures. After 20-24 hours of incubation, medium supernatants were collected and analyzed with BD OptEIA Human IL-2 ELISA Set (BD Biosciences #555190).
  • IL-2 production elicited by the anti-mesothelin CAR constructs was assessed. The data are presented in Figure 19D-F.
  • Figure 19 IL-2 production triggered by HN1 scFv (Fig. 19D), SS I scFv (Fig. 19E), and m912 scFv (Fig. 19F) On-switch CAR variants.
  • IL-2 production by a conventional CAR Fig. 19G, construct #358
  • Effector human CD4+ Jurkat T cells engineered with CARs.
  • Target K562 cell lines with or without the cognate mesothelin antigen. Amounts of secreted IL-2 by effector cells were quantified by enzyme-linked immunosorbent assay (ELISA).
  • Figure 20A summarizes the molecular structure of the subject gibberellic acid
  • the antigen binding portion comprises the anti-human CD 19 scFv.
  • the intracellular portion comprises two 4- IBB co- stimulatory domains, a GID1 and GAI dimerizer-binding pair, and an IT AM intracellular signaling domain. All membrane- anchored polypeptides are di-sulfide bonded homo-dimers.
  • the anti-CD 19 scFv was cloned from a plasmid.
  • the human 4- IBB co- stimulation and CD3 zeta IT AM signaling chains were cloned from cDNAs supplied by Open Biosystems.
  • GID 1- and GAI-encoding sequences were cloned from Addgene plasmids. Standard molecular cloning techniques (polymerase chain reaction (PCR), restriction digestion, ligation, etc.) were applied to generate lentiviral expression plasmids. Effector and target cell culturing conditions
  • K562 target cells were cultured in IMDM supplemented with 10% fetal bovine serum (FBS).
  • FBS fetal bovine serum
  • Pantropic VSV-G pseudotyped lentivirus was produced from Lenti-X 293T cells
  • Jurkat and K562 cells were split 1-2 days in advance to ensure that cultures would be in log phase at the time of transduction.
  • Transduced Jurkat and K562 cells were cultured for at least 7 days before experiments were conducted.
  • Expression levels of CARs encoded in the lentiviral constructs were quantified by detecting either fluorophore-conjugated antibodies or fluorescent reporter proteins using a flow cytometer.
  • K562 target cells at a 1 :2 effectontarget ratio.
  • the gibberellic acid-3 acetoxymethyl ester (gibberrelic acid-3 AM) pre-dissolved in ethanol (Toronto Research Chemicals #G377500) was diluted in growth medium and added to reaction mixtures.
  • Gibberellic acid (gibberellic acid-3 AM) was used at 10 mM. After 20-24 hours of incubation, medium supernatants were collected and analyzed with BD OptEIA Human IL-2 ELISA Set (BD Biosciences #555190).
  • FIG. 20 IL-2 production triggered by gibberellic acid dimerizer CAR variant
  • IL-2 production by a conventional CAR (Fig. 20C, construct "125") was measured and included for comparison to On-switch CAR.
  • Effector human CD4+ Jurkat T cells engineered with CARs.
  • Target K562 cell lines with or without the cognate CD19 antigen. Amounts of secreted IL-2 by effector cells were quantified by enzyme-linked
  • FIG. 21 A and 2 IB summarize the molecular structure of the CARs described here.
  • OX-40 encoding sequences were cloned from cDNAs supplied by Open Biosystems.
  • FKBP- and FRB-encoding sequences were cloned from plasmids from Addgene.
  • Effector and target cells are cultured and transfected according to Example 1 using the on-switch CAR CD28 and OX-40 co- stimulatory domain containing constructs described (Fig. 21A-B, constructs “365+367” and “399+400", respectively) and corresponding conventional CAR controls (Fig. 21C-D, constructs "366” and “398", respectively).
  • IL-2 production, NFAT activity assays, and flow cytometry-based assays can also be performed with the CD28 co- stimulatory domain containing construct and OX-40 co-stimulatory domain containing construct as described for Example 1.
  • subunits of on-switch CAR CD28 and OX-40 co- stimulatory domain containing constructs can be paired with subunits of constructs from Example 1 (e.g., "197+367”, “ 365+206,” “197+400”, “399+206,” etc.).
  • An On-switch CAR can be assessed for its ability to mediate in vivo killing of a target tumor cell. In vivo tumor cell killing elicited by injection of T cells expressing the ON-switch CAR is assessed. Tumor cell lines that have been confirmed in vitro to express the cognate antigen and can be killed by CD8 + T cells expressing the corresponding CAR are used. Tumor cells engineered to express either the firefly or Renilla lucif erase to enable bio-luminescence imaging to quantify tumor burden in vivo can be used.
  • Tumor cells are injected into immunocompromised mice (e.g., 6-10 week old female NOD scid gamma (NSG) mice) either subcutaneously for subcutaneous tumor models or intravenously for systemic tumor models.
  • the method of tumor implantation and the optimal number of tumor cells to implant can be based on conditions optimal for the tumor cell line used. Tumor burden can be monitored twice a week by bio-luminescence imaging and by caliper measurement when applicable. As soon as tumor burden is detectable, 0.5-2.5 x 10 ⁇ 7 total T cells (1: 1 CD4 + :CD8 + ) expressing the ON-switch CAR are intravenously injected into mice to begin treatment.
  • a dimerizing small molecule drug (e.g., rapalog) is administered intraperitoneally in a vehicle formulation.
  • On-switch CAR-expressing T cells can be injected repeatedly during the experiment to enhance the anti-tumor effect.
  • Interleukin-2 (IL-2) can be administered to enhance the anti-tumor effect.

Abstract

The present disclosure provides a heterodimeric, conditionally active chimeric antigen receptor (CAR), and a nucleic acid comprising a nucleotide sequence encoding the CAR. The present disclosure provides cells genetically modified to produce the CAR. A CAR of the present disclosure can be used in various methods, which are also provided.

Description

CHIMERIC ANTIGEN RECEPTOR AND METHODS OF USE THEREOF
CROSS-REFERENCE
[0001] This application claims the benefit of U.S. Provisional Patent Application No. 61/765,585, filed February 15, 2013, which application is incorporated herein by reference in its entirety.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH
[0002] This invention was made with government support under Grant Nos. EY016546 and
GM 101782 awarded by the National Institutes of Health. The government has certain rights in the invention.
INCORPORATION BY REFERENCE OF SEQUENCE LISTING PROVIDED AS A TEXT FILE
[0003] A Sequence Listing is provided herewith as a text file, "UCSF-464WO SeqList_ST25.txt" created on February 13, 2014 and having a size of 153 KB. The contents of the text file are incorporated by reference herein in their entirety.
INTRODUCTION
[0004] In cell-based adoptive immunotherapy, immune cells isolated from a patient can be
modified to express synthetic proteins that enable the cells to perform new therapeutic functions after they are subsequently transferred back into the patient. An example of such a synthetic protein is a chimeric antigen receptor (CAR). An example of a currently used CAR is a fusion of an extracellular recognition domain (e.g., an antigen-binding domain), a transmembrane domain, and one or more intracellular signaling domains. Upon antigen engagement, the intracellular signaling portion of the CAR can initiate an activation-related response in an immune cell, such are release of cytolytic molecules to induce tumor cell death, etc. However, such CARs are not capable of being pharmacologically controlled. There is a need in the art for a conditionally activatable CAR that can be controlled pharmacologically.
SUMMARY
[0005] The present disclosure provides a heterodimeric, conditionally active chimeric antigen
receptor (CAR), and a nucleic acid comprising a nucleotide sequence encoding the CAR. The present disclosure provides cells genetically modified to produce the CAR. A CAR of the present disclosure can be used in various methods, which are also provided.
BRIEF DESCRIPTION OF THE DRAWINGS
[0006] Figures 1A and IB provide nucleotide and amino acid sequences of the domains of
construct #122.
[0007] Figures 2A and 2B provide nucleotide and amino acid sequences of the domains of
construct #123.
[0008] Figures 3A and 3B provide nucleotide and amino acid sequences of the domains of
construct #125.
[0009] Figure 4 provides nucleotide and amino acid sequences of the domains of construct #126.
[0010] Figures 5A and 5B provide nucleotide and amino acid sequences of the domains of
construct #168.
[0011] Figures 6A-C provide nucleotide and amino acid sequences of the domains of construct #169.
[0012] Figures 7A and 7B provide nucleotide and amino acid sequences of the domains of
construct #170.
[0013] Figures 8A and 8B provide nucleotide and amino acid sequences of the domains of
construct #197.
[0014] Figures 9A-C provide nucleotide and amino acid sequences of the domains of construct #206.
[0015] Figures 10A and 10B provide nucleotide and amino acid sequences of the domains of construct #207.
[0016] Figures 11A-C provide nucleotide and amino acid sequences of the domains of construct #199.
[0017] Figure 12 depicts IL-2 production triggered by five On-switch CAR variants.
[0018] Figure 13 depicts IL-2 production by control Jurkat lines.
[0019] Figure 14 depicts a comparison between CAR constructs "122 + 206" and "197 + 206".
[0020] Figure 15 depicts cytotoxicity data with the On-switch CAR "197+206."
[0021] Figure 16 depicts T cell activation data using CAR constructs "122 + 199"; "197 + 199"; and "122 + 168."
[0022] Figure 17 is a schematic representation of an exemplary On-switch CAR.
[0023] Figures 18A and 18B depict various exemplary On-switch CAR. [0024] Figures 19A-G depict IL-2 production triggered by 3 different On-switch CAR variants recognizing human mesothelin.
[0025] Figures 20A-C depict IL-2 production triggered by an On-switch CAR variant with a
gibberellic acid responsive dimerization pair.
[0026] Figures 21A-D depict exemplary On-switch CARs and conventional CARs with various co- stimulatory domains.
[0027] Figures 22A and 22B provide nucleotide and amino acid sequences of the domains of
construct #270.
[0028] Figures 23A and 23B provide nucleotide and amino acid sequences of the domains of
construct #300.
[0029] Figures 24A and 24B provide nucleotide and amino acid sequences of the domains of
construct #336.
[0030] Figures 25A and 25B provide nucleotide and amino acid sequences of the domains of
construct #337.
[0031] Figures 26A and 26B provide nucleotide and amino acid sequences of the domains of
construct #357.
[0032] Figures 27A and 27B provide nucleotide and amino acid sequences of the domains of
construct #365.
[0033] Figures 28A and 28B provide nucleotide and amino acid sequences of the domains of
construct #366.
[0034] Figures 29A and 29B provide nucleotide and amino acid sequences of the domains of
construct #367.
[0035] Figures 30A and 30B provide nucleotide and amino acid sequences of the domains of
construct #398.
[0036] Figures 31A and 3 IB provide nucleotide and amino acid sequences of the domains of
construct #399.
[0037] Figures 32A and 32B provide nucleotide and amino acid sequences of the domains of
construct #400.
[0038] Figures 33A and 33B provide nucleotide and amino acid sequences of the domains of
construct #358.
DEFINITIONS
[0039] The terms "polynucleotide" and "nucleic acid," used interchangeably herein, refer to a polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides. Thus, this term includes, but is not limited to, single-, double-, or multi-stranded DNA or RNA, genomic DNA, cDNA, DNA-RNA hybrids, or a polymer comprising purine and pyrimidine bases or other natural, chemically or biochemically modified, non-natural, or derivatized nucleotide bases.
[0040] The terms "antibodies" and "immunoglobulin" include antibodies or immunoglobulins of any isotype, fragments of antibodies which retain specific binding to antigen, including, but not limited to, Fab, Fv, scFv, and Fd fragments, chimeric antibodies, humanized antibodies, single-chain antibodies, and fusion proteins comprising an antigen-binding portion of an antibody and a non-antibody protein.
[0041] "Antibody fragments" comprise a portion of an intact antibody, for example, the antigen binding or variable region of the intact antibody. Examples of antibody fragments include Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies (Zapata et al., Protein Eng. 8(10): 1057-1062 (1995)); single-chain antibody molecules; and multispecific antibodies formed from antibody fragments. Papain digestion of antibodies produces two identical antigen -binding fragments, called "Fab" fragments, each with a single antigen-binding site, and a residual "Fc" fragment, a designation reflecting the ability to crystallize readily.
Pepsin treatment yields an F(ab')2 fragment that has two antigen combining sites and is still capable of cross-linking antigen.
[0042] "Single-chain Fv" or "sFv" antibody fragments comprise the VH and VL domains of
antibody, wherein these domains are present in a single polypeptide chain. In some embodiments, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains, which enables the sFv to form the desired structure for antigen binding. For a review of sFv, see Pluckthun in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315 (1994).
[0043] As used herein, the term "affinity" refers to the equilibrium constant for the reversible
binding of two agents and is expressed as a dissociation constant (Kd). Affinity can be at least 1-fold greater, at least 2-fold greater, at least 3-fold greater, at least 4-fold greater, at least 5-fold greater, at least 6-fold greater, at least 7-fold greater, at least 8-fold greater, at least 9-fold greater, at least 10-fold greater, at least 20-fold greater, at least 30-fold greater, at least 40-fold greater, at least 50-fold greater, at least 60-fold greater, at least 70-fold greater, at least 80-fold greater, at least 90-fold greater, at least 100-fold greater, or at least 1000-fold greater, or more, than the affinity of an antibody for unrelated amino acid sequences. Affinity of an antibody to a target protein can be, for example, from about 100 nanomolar (nM) to about 0.1 nM, from about 100 nM to about 1 picomolar (pM), or from about 100 nM to about 1 femtomolar (fM) or more. As used herein, the term "avidity" refers to the resistance of a complex of two or more agents to dissociation after dilution. The terms "immunoreactive" and "preferentially binds" are used interchangeably herein with respect to antibodies and/or antigen-binding fragments.
[0044] The term "binding" refers to a direct association between two molecules, due to, for
example, covalent, electrostatic, hydrophobic, and ionic and/or hydrogen-bond interactions, including interactions such as salt bridges and water bridges. Non-specific binding would refer to binding with an affinity of less than about 10" M, e.g., binding with an affinity of 10"6 M, 10"5 M, 10"4 M, etc.
[0045] As used herein, the term "hinge region" refers to a flexible polypeptide connector region (also referred to herein as "hinge" or "spacer") providing structural flexibility and spacing to flanking polypeptide regions and can consist of natural or synthetic polypeptides. A "hinge region" derived from an immunoglobulin (e.g., IgGl) is generally defined as stretching from Glu216 to Pro230 of human IgGl (Burton (1985) Molec. Immunol., 22: 161- 206). Hinge regions of other IgG isotypes may be aligned with the IgGl sequence by placing the first and last cysteine residues forming inter-heavy chain disulfide (S-S) bonds in the same positions. The hinge region may be of natural occurrence or non-natural occurrence, including but not limited to an altered hinge region as described in U.S. Pat. No. 5,677,425. The hinge region can include complete hinge region derived from an antibody of a different class or subclass from that of the CHI domain. The term "hinge region" can also include regions derived from CD8 and other receptors that provide a similar function in providing flexibility and spacing to flanking regions.
[0046] An "isolated" polypeptide is one that has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or
nonproteinaceous solutes. In some embodiments, the polypeptide will be purified (1) to greater than 90%, greater than 95%, or greater than 98%, by weight of antibody as determined by the Lowry method, for example, more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by sodium dodecyl sulfate- polyacrylamide gel electrophoresis (SDS-PAGE) under reducing or nonreducing conditions using Coomassie blue or silver stain. Isolated polypeptide includes the polypeptide in situ within recombinant cells since at least one component of the polypeptide's natural environment will not be present. In some instances, isolated polypeptide will be prepared by at least one purification step.
[0047] As used herein, the term "immune cells" generally includes white blood cells (leukocytes) which are derived from hematopoietic stem cells (HSC) produced in the bone marrow. "Immune cells" includes, e.g., lymphocytes (T cells, B cells, natural killer (NK) cells) and myeloid-derived cells (neutrophil, eosinophil, basophil, monocyte, macrophage, dendritic cells).
[0048] "T cell" includes all types of immune cells expressing CD3 including T-helper cells (CD4+ cells), cytotoxic T-cells (CD8+ cells), T-regulatory cells (Treg) and gamma-delta T cells.
[0049] A "cytotoxic cell" includes CD8+ T cells, natural-killer (NK) cells, and neutrophils, which cells are capable of mediating cytotoxicity responses.
[0050] As used herein, the term "stem cell" generally includes pluripotent or multipotent stem
cells. "Stem cells" includes, e.g., embryonic stem cells (ES); mesenchymal stem cells (MSC); induced-pluripotent stem cells (iPS); and committed progenitor cells (hematopoeitic stem cells (HSC); bone marrow derived cells, etc.).
[0051] As used herein, the terms "treatment," "treating," and the like, refer to obtaining a desired pharmacologic and/or physiologic effect. The effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease. "Treatment," as used herein, covers any treatment of a disease in a mammal, e.g., in a human, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; and (c) relieving the disease, i.e., causing regression of the disease.
[0052] The terms "individual," "subject," "host," and "patient," used interchangeably herein, refer to a mammal, including, but not limited to, murines (e.g., rats, mice), lagomorphs (e.g., rabbits), non-human primates, humans, canines, felines, ungulates (e.g., equines, bovines, ovines, porcines, caprines), etc.
[0053] A "therapeutically effective amount" or "efficacious amount" refers to the amount of an agent, or combined amounts of two agents, that, when administered to a mammal or other subject for treating a disease, is sufficient to effect such treatment for the disease. The "therapeutically effective amount" will vary depending on the agent(s), the disease and its severity and the age, weight, etc., of the subject to be treated. [0054] Before the present invention is further described, it is to be understood that this invention is not limited to particular embodiments described, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.
[0055] Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention.
[0056] Unless defined otherwise, all technical and scientific terms used herein have the same
meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited.
[0057] It must be noted that as used herein and in the appended claims, the singular forms "a,"
"an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a chimeric antigen receptor" includes a plurality of such chimeric antigen receptor and reference to "the dimerizer-binding pair" includes reference to one or more dimerizer-binding pairs and equivalents thereof known to those skilled in the art, and so forth. It is further noted that the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as "solely," "only" and the like in connection with the recitation of claim elements, or use of a "negative" limitation.
[0058] It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable sub-combination. All combinations of the embodiments pertaining to the invention are specifically embraced by the present invention and are disclosed herein just as if each and every combination was individually and explicitly disclosed. In addition, all subcombinations of the various embodiments and elements thereof are also specifically embraced by the present invention and are disclosed herein just as if each and every such sub-combination was individually and explicitly disclosed herein.
[0059] The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided may be different from the actual publication dates which may need to be independently confirmed.
DETAILED DESCRIPTION
[0060] The present disclosure provides a heterodimeric, conditionally active chimeric antigen
receptor (CAR), and a nucleic acid comprising a nucleotide sequence encoding the CAR. The present disclosure provides cells genetically modified to produce the CAR. A CAR of the present disclosure can be used in various methods, which are also provided.
HETERODIMERIC, CONDITIONALLY ACTIVE CHIMERIC ANTIGEN RECEPTOR.
[0061] The present disclosure provides a heterodimeric, conditionally active chimeric antigen
receptor, which, for simplicity, is referred to herein as "CAR."
[0062] In some embodiments, a CAR of the present disclosure comprises: a) a first polypeptide comprising: i) a member of a specific binding pair (e.g., an antigen-binding domain); ii) a first modulatory domain; iii) a first member of a dimerization pair; and iv) a transmembrane domain interposed between the member of a specific binding pair (e.g., an antigen-binding domain) and the first modulatory domain; and b) a second polypeptide comprising: i) a transmembrane domain; ii) a second modulatory domain; iii) a second member of the dimerization pair; and iv) an intracellular signaling domain. The modulatory domain can be a co- stimulatory domain.
[0063] In some embodiments, a CAR of the present disclosure comprises: a) a first polypeptide comprising: i) a member of a specific binding pair (e.g., an antigen-binding domain); ii) a first co-stimulatory domain; iii) a first member of a dimerization pair (e.g., a dimerizer- binding pair); and iv) a transmembrane domain interposed between the member of a specific binding pair (e.g., an antigen-binding domain) and the first co- stimulatory domain; and b) a second polypeptide comprising: i) a transmembrane domain; ii) a second co- stimulatory domain; iii) a second member of the dimerization pair (e.g., the dimerizer-binding pair); and iv) an intracellular signaling domain.
[0064] In some embodiments, a CAR of the present disclosure comprises: a) a first polypeptide comprising: i) a member of a specific binding pair (e.g., an antigen-binding domain); ii) a modulatory domain; iii) a first member of a dimerization pair (e.g., a dimerizer-binding pair); iv) a transmembrane domain interposed between the member of a specific binding pair (e.g., an antigen-binding domain) and the modulatory domain; and b) a second polypeptide comprising: i) a second member of the dimerization pair (e.g., the dimerizer- binding pair); and ii) an intracellular signaling domain. The modulatory domain can be a co- stimulatory domain.
[0065] In some embodiments, a CAR of the present disclosure comprises: a) a first polypeptide comprising: i) a member of a specific binding pair (e.g., an antigen-binding domain); ii) a co-stimulatory domain; iii) a first member of a dimerization pair (e.g., a dimerizer-binding pair); iv) a transmembrane domain interposed between the member of a specific binding pair (e.g., an antigen-binding domain) and the co- stimulatory domain; and b) a second polypeptide comprising: i) a second member of the dimerization pair (e.g., the dimerizer- binding pair); and ii) an intracellular signaling domain.
[0066] An example of a subject CAR is represented schematically in Figure 17. A CAR of the present disclosure can be present in the plasma membrane of a eukaryotic cell, e.g., a mammalian cell, where suitable mammalian cells include, but are not limited to, a cytotoxic cell, a T lymphocyte, a stem cell, a progeny of a stem cell, a progenitor cell, a progeny of a progenitor cell, and an NK cell. When present in the plasma membrane of a eukaryotic cell, a CAR of the present disclosure is active in the presence of: 1) a dimerizing agent binds to the first and second members of the dimerizer-binding pair in the CAR, or otherwise induces dimerization of the first and second members of the dimer; and 2) a factor that binds the member of a specific binding pair (e.g., an antigen-binding domain), e.g., an antigen that binds the antigen-binding domain of the CAR. The factor that binds the member of the specific binding pair is a second member of the specific binding pair. The second member of the specific binding pair can be a soluble (e.g., not bound to a cell) factor; a factor present on the surface of a cell such as a target cell; a factor presented on a solid surface; a factor present in a lipid bilayer; and the like. Where the member of a specific binding pair is an antibody, and the second member of the specific binding pair is an antigen, the antigen can be a soluble (e.g., not bound to a cell) antigen; an antigen present on the surface of a cell such as a target cell; an antigen presented on a solid surface; an antigen present in a lipid bilayer; and the like.
[0067] In some instances, a CAR of the present disclosure, when present in the plasma membrane of a eukaryotic cell, and when activated by a second member of a specific binding pair that binds the member of the specific-binding pair of the CAR (e.g., an antigen that binds the antigen -binding domain of the CAR) and a dimerizing agent, increases expression of at least one nucleic acid in the cell. For example, in some cases, a CAR of the present disclosure, when present in the plasma membrane of a eukaryotic cell, and when activated by an antigen that binds the antigen-binding domain of the CAR and a dimerizing agent, increases expression of at least one nucleic acid in the cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared with the level of transcription of the nucleic acid in the absence of the antigen and/or the dimerizing agent.
[0068] As an example, the second polypeptide of a CAR of the present disclosure can include an immunoreceptor tyrosine -based activation motif (ITAM)-containing intracellular signaling polypeptide; in such cases, a CAR of the present disclosure, when present in the plasma membrane of a eukaryotic cell, and when activated by an antigen that binds the antigen- binding domain of the CAR and a dimerizing agent, increases nuclear factor of activated T cells (NFAT)-dependent transcription. NFAT-dependent transcription includes transcription induced by any member of the NFAT family, including, e.g., NFATcl, NFATc2, NFATc3, NFATc4, NFAT5; AP-1; Spl; ΝΚκΒ; and the like.
[0069] A CAR of the present disclosure, when present in the plasma membrane of a eukaryotic cell, and when activated by an antigen that binds the antigen-binding domain of the CAR and a dimerizing agent, can, in some instances, result in increased production of one or more cytokines by the cell. For example, a CAR of the present disclosure, when present in the plasma membrane of a eukaryotic cell, and when activated by an antigen that binds the antigen-binding domain of the CAR and a dimerizing agent, can increase production of a cytokine by the cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared with the amount of cytokine produced by the cell in the absence of the antigen and/or the dimerizing agent. Cytokines whose production can be increased include, but are not limited to, an interferon, e.g., IL-2, interferon gamma (IFN-γ), tumor necrosis factor-alpha (TNF-a), IL-15, IL-12, IL-4, IL-5, IL-10; a chemokine; a growth factor; and the like.
[0070] In some cases, a CAR of the present disclosure, when present in the plasma membrane of a eukaryotic cell, and when activated by an antigen that binds the antigen-binding domain of the CAR and a dimerizing agent, can result in both an increase in transcription of a nucleic acid in the cell and an increase in production of a cytokine by the cell.
[0071] In some instances, a CAR of the present disclosure, when present in the plasma membrane of a eukaryotic cell, and when activated by a dimerizing agent, results in cytotoxic activity by the cell toward a target cell that expresses on its cell surface an antigen to which the antigen -binding domain of the first polypeptide of the CAR binds. For example, where the eukaryotic cell is a cytotoxic cell (e.g., an NK cell or a cytotoxic T lymphocyte), a CAR of the present disclosure, when present in the plasma membrane of the cell, and when activated by a dimerizing agent, increases cytotoxic activity of the cell toward a target cell that expresses on its cell surface an antigen to which the antigen-binding domain of the first polypeptide of the CAR binds. For example, where the eukaryotic cell is an NK cell or a T lymphocyte, a CAR of the present disclosure, when present in the plasma membrane of the cell, and when activated by a dimerizing agent, increases cytotoxic activity of the cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared to the cytotoxic activity of the cell in the absence of the dimerizing agent.
[0072] In some cases, a CAR of the present disclosure, when present in the plasma membrane of a eukaryotic cell, and when activated by an antigen that binds the antigen-binding domain of the CAR and a dimerizing agent, can result in other CAR activation related events such as proliferation and expansion (either due to increased cellular division or anti-apoptotic responses).
[0073] In some cases, a CAR of the present disclosure, when present in the plasma membrane of a eukaryotic cell, and when activated by an antigen that binds the antigen-binding domain of the CAR and a dimerizing agent, can result in other CAR activation related events such as intracellular signaling modulation, cellular differentiation, or cell death.
[0074] A CAR of the present disclosure can be present in a eukaryotic cell membrane, where the first and second polypeptides of the CAR are not covalently linked to one another. A CAR of the present disclosure can be present in a eukaryotic cell membrane as a single heterodimer that is not covalently linked to any other polypeptide in the membrane. Alternatively, a first CAR of the present disclosure can be present in a eukaryotic cell membrane as a heterodimer that is covalently or non-covalently linked to a second CAR of the present disclosure. In some cases, the first and the second CAR are covalently linked via a disulfide bond formed between cysteines present in a hinge region present in both the first polypeptide of the first CAR and the first polypeptide of the second CAR.
[0075] In some cases, a CAR of the present disclosure can be present in a eukaryotic cell
membrane, where the first polypeptides of the CAR comprise an antibody fragment and the second polypeptides of the CAR comprise a signal transducing domain derived from a cytokine receptor, such that, upon dimerization, the CAR may represent a heterodimeric- signalobody CAR, e.g., a signalobody composed of at least two independent polypeptides. A "signalobody", as it is known in the art, is a single chimeric macromolecule composed of an antibody fragment and a signal transduction domain derived from a cytokine receptor. In certain instances, a heterodimeric-signalobody CAR of the present disclosure, when present in the cell membrane of a eukaryotic cell, dimerized by a dimerizer, and activated by an antigen, e.g., an oligomerized antigen, may induce the oligomerization of the heterodimeric- signalobody CAR. Such ligand-induced oligomerization of a heterodimeric-signalobody CAR may activate, e.g., increase, or perpetuate, e.g., maintain, signal transduction, e.g., ligand-induced oligomerization of a heterodimeric-signalobody CAR may transmit a signal eliciting a cellular response. In some instances, a plurality of heterodimeric-signalobody CARs may be utilized combinatorially to elicit a desired cellular response.
Member of a specific binding pair
[0076] A CAR of the present disclosure includes a member of a specific binding pair. Specific binding pairs include, but are not limited to, antigen-antibody binding pairs; ligand-receptor binding pairs; and the like. Thus, a member of a specific binding pair suitable for use in a CAR of the present disclosure includes an antigen; an antibody; a ligand; and a ligand- binding receptor.
Antigen-binding domain
[0077] An antigen-binding domain suitable for use in a CAR of the present disclosure can be any antigen-binding polypeptide, a wide variety of which are known in the art. In some instances, the antigen-binding domain is a single chain Fv (scFv). Other antibody based recognition domains (cAb VHH (camelid antibody variable domains) and humanized versions, IgNAR VH (shark antibody variable domains) and humanized versions, sdAb VH (single domain antibody variable domains) and "camelized" antibody variable domains are suitable for use. In some instances, T-cell receptor (TCR) based recognition domains such as single chain TCR (scTv, single chain two-domain TCR containing ν νβ) are also suitable for use.
[0078] An antigen-binding domain suitable for use in a CAR of the present disclosure can have a variety of antigen-binding specificities. In some cases, the antigen-binding domain is specific for an epitope present in an antigen that is expressed by (synthesized by) a cancer cell, i.e., a cancer cell associated antigen. The cancer cell associated antigen can be an antigen associated with, e.g., a breast cancer cell, a B cell lymphoma, a Hodgkin lymphoma cell, an ovarian cancer cell, a prostate cancer cell, a mesothelioma, a lung cancer cell (e.g., a small cell lung cancer cell), a non-Hodgkin B-cell lymphoma (B-NHL) cell, an ovarian cancer cell, a prostate cancer cell, a mesothelioma cell, a lung cancer cell (e.g., a small cell lung cancer cell), a melanoma cell, a chronic lymphocytic leukemia cell, an acute lymphocytic leukemia cell, a neuroblastoma cell, a glioma, a glioblastoma, a
medulloblastoma, a colorectal cancer cell, etc. A cancer cell associated antigen may also be expressed by a non-cancerous cell.
[0079] Non-limiting examples of antigens to which an antigen-binding domain of a subject CAR can bind include, e.g., CD19, CD20, CD38, CD30, Her2/neu, ERBB2, CA125, MUC-1, prostate-specific membrane antigen (PSMA), CD44 surface adhesion molecule, mesothelin, carcinoembryonic antigen (CEA), epidermal growth factor receptor (EGFR), EGFRvIII, vascular endothelial growth factor receptor-2 (VEGFR2), high molecular weight-melanoma associated antigen (HMW-MAA), MAGE-A1, IL-13R-a2, GD2, and the like.
Ligand
[0080] In some cases, a member of a specific binding pair suitable for use in a subject CAR is a ligand for a receptor. Ligands include, but are not limited to, cytokines (e.g., IL-13, etc.); growth factors (e.g., heregulin; vascular endothelial growth factor (VEGF); and the like); an integrin-binding peptide (e.g., a peptide comprising the sequence Arg-Gly-Asp); and the like.
[0081] Where the member of a specific binding pair in a subject CAR is a ligand, the CAR can be activated in the presence of both a dimerizer agent and a second member of the specific binding pair, where the second member of the specific binding pair is a receptor for the ligand. For example, where the ligand is VEGF, the second member of the specific binding pair can be a VEGF receptor, including a soluble VEGF receptor. As another example, where the ligand is heregulin, the second member of the specific binding pair can be Her2. Receptors
[0082] As noted above, in some cases, the member of a specific binding pair that is included in a subject CAR is a receptor, e.g., a receptor for a ligand, a co-receptor, etc. The receptor can be a ligand-binding fragment of a receptor. Suitable receptors include, but are not limited to, a growth factor receptor (e.g., a VEGF receptor); a killer cell lectin-like receptor subfamily K, member 1 (NKG2D) polypeptide (receptor for MICA, MICB, and ULB6); a cytokine receptor (e.g., an IL-13 receptor; an IL-2 receptor; etc.); Her2; CD27; a natural cytotoxicity receptor (NCR) (e.g., NKP30 (NCR3/CD337) polypeptide (receptor for HLA-B-associated transcript 3 (BAT3) and B7-H6); etc.); etc.
Hinge region
[0083] In some cases, the first polypeptide of a subject CAR comprises a hinge region (also
referred to herein as a "spacer"), where the hinge region is interposed between the antigen- binding domain and the transmembrane domain. In some cases, the hinge region is an immunoglobulin heavy chain hinge region. In some cases, the hinge region is a hinge region polypeptide derived from a receptor (e.g., a CD8-derived hinge region).
[0084] The hinge region can have a length of from about 4 amino acids to about 50 amino acids, e.g., from about 4 aa to about 10 aa, from about 10 aa to about 15 aa, from about 15 aa to about 20 aa, from about 20 aa to about 25 aa, from about 25 aa to about 30 aa, from about 30 aa to about 40 aa, or from about 40 aa to about 50 aa.
[0085] Suitable spacers can be readily selected and can be of any of a number of suitable lengths, such as from 1 amino acid (e.g., Gly) to 20 amino acids, from 2 amino acids to 15 amino acids, from 3 amino acids to 12 amino acids, including 4 amino acids to 10 amino acids, 5 amino acids to 9 amino acids, 6 amino acids to 8 amino acids, or 7 amino acids to 8 amino acids, and can be 1, 2, 3, 4, 5, 6, or 7 amino acids.
[0086] Exemplary spacers include glycine polymers (G)n, glycine- serine polymers (including, for example, (GS)n, (GSGGS)n (SEQ ID NO:37) and (GGGS)n (SEQ ID NO:38), where n is an integer of at least one), glycine-alanine polymers, alanine-serine polymers, and other flexible linkers known in the art. Glycine and glycine-serine polymers can be used; both Gly and Ser are relatively unstructured, and therefore can serve as a neutral tether between components. Glycine polymers can be used; glycine accesses significantly more phi-psi space than even alanine, and is much less restricted than residues with longer side chains (see Scheraga, Rev. Computational Chem. 11173-142 (1992)). Exemplary spacers can comprise amino acid sequences including, but not limited to, GGSG (SEQ ID NO:39), GGSGG (SEQ ID NO:40), GSGSG (SEQ ID NO:41), GSGGG (SEQ ID NO:42), GGGSG
(SEQ ID NO:43), GSSSG (SEQ ID NO:44), and the like.
[0087] In some cases, the hinge region in the first polypeptide of a subject CAR includes at least one cysteine. For example, in some cases, the hinge region can include the sequence Cys-
Pro-Pro-Cys. If present, a cysteine in the hinge region of a first CAR can be available to form a disulfide bond with a hinge region in a second CAR.
[0088] Immunoglobulin hinge region amino acid sequences are known in the art; see, e.g., Tan et al. (1990) Proc. Natl. Acad. Sci. USA 87: 162; and Huck et al. (1986) Nucl. Acids Res.
14: 1779. As non-limiting examples, an immunoglobulin hinge region can include one of the following amino acid sequences: DKTHT (SEQ ID NO:45); CPPC (SEQ ID NO:46);
CPEPKSCDTPPPCPR (SEQ ID NO:47) (see, e.g., Glaser et al. (2005) J. Biol. Chem.
280:41494); ELKTPLGDTTHT (SEQ ID NO:48); KSCDKTHTCP (SEQ ID NO:49);
KCCVDCP (SEQ ID NO:50); KYGPPCP (SEQ ID NO:51); EPKSCDKTHTCPPCP (SEQ
ID NO:52) (human IgGl hinge); ERKCCVECPPCP (SEQ ID NO:53) (human IgG2 hinge);
ELKTPLGDTTHTCPRCP (SEQ ID NO:54) (human IgG3 hinge); SPNMVPHAHHAQ
(SEQ ID NO:55) (human IgG4 hinge); and the like.
[0089] The hinge region can comprise an amino acid sequence of a human IgGl, IgG2, IgG3, or
IgG4, hinge region. The hinge region can include one or more amino acid substitutions and/or insertions and/or deletions compared to a wild-type (naturally-occurring) hinge region. For example, His229 of human IgGl hinge can be substituted with Tyr, so that the hinge region comprises the sequence EPKSCDKTYTCPPCP (SEQ ID NO:52); see, e.g.,
Yan et al. (2012) J. Biol. Chem. 287:5891.
[0090] The hinge region can comprise an amino acid sequence derived from human CD8; e.g., the hinge region can comprise the amino acid sequence:
TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACD (SEQ ID NO:56), or a variant thereof.
Transmembrane domain
[0091] The first and the second polypeptides of a CAR of the present disclosure include
transmembrane domains for insertion into a eukaryotic cell membrane. The transmembrane domain of the first polypeptide is interposed between the antigen-binding domain and the co-stimulatory domain. Where the first polypeptide includes a hinge region, the
transmembrane domain is interposed between the hinge region and the co- stimulatory domain, such that the first polypeptide comprises, in order from the amino terminus (N- terminus) to the carboxyl terminus (C-terminus): an antigen-binding domain; a hinge region; a transmembrane domain; a first co- stimulatory domain; and a first member of a dimerizer-binding pair.
[0092] The transmembrane domain of the second polypeptide is at or near the N-terminus of the polypeptide, such that the second polypeptide comprises, in order from N-terminus to C- terminus: a transmembrane domain; a second co-stimulatory domain; a second member of the dimerizer-binding pair; and an intracellular signaling domain.
[0093] Any transmembrane (TM) domain that provides for insertion of a polypeptide into the cell membrane of a eukaryotic (e.g., mammalian) cell is suitable for use. As one non-limiting example, the TM sequence IYIWAPLAGTCGVLLLSLVITLYC (SEQ ID NO:30) can be used. Additional non-limiting examples of suitable TM sequences include: a) CD8 beta derived : LGLLVAGVLVLLVSLGVAIHLCC (SEQ ID NO:57); b) CD4 derived:
ALIVLGGVAGLLLFIGLGIFFCVRC (SEQ ID NO:58); c) CD3 zeta derived:
LCYLLDGILFIYGVILTALFLRV (SEQ ID NO:59); d) CD28 derived:
WVLVVVGGVLACYSLLVTVAFIIFWV (SEQ ID NO:60); e) CD134 (OX40) derived: VAAILGLGLVLGLLGPLAILLALYLL (SEQ ID NO:61); and f) CD7 derived:
ALPAALAVISFLLGLGLGVACVLA (SEQ ID NO:62).
Linkers
[0094] In some cases, a first polypeptide of a subject CAR includes a linker between any two
adjacent domains. For example, a linker can be disposed between the transmembrane domain and the first co- stimulatory domain of the first polypeptide. As another example, a linker can be disposed between the first co-stimulatory domain and the first member of a dimerizer-binding pair of the first polypeptide. As another example, a linker can be disposed between the transmembrane domain and the second co- stimulatory domain of the second polypeptide. As another example, a linker can be disposed between the second co- stimulatory domain and the second member of the dimerizer-binding pair of the second polypeptide. As another example, a linker can be disposed between the second member of the dimerizer-binding pair and the intracellular signaling domain of the second polypeptide.
[0095] The linker peptide may have any of a variety of amino acid sequences. Proteins can be joined by a spacer peptide, generally of a flexible nature, although other chemical linkages are not excluded. A linker can be a peptide of between about 6 and about 40 amino acids in length, or between about 6 and about 25 amino acids in length. These linkers can be produced by using synthetic, linker-encoding oligonucleotides to couple the proteins.
Peptide linkers with a degree of flexibility can be used. The linking peptides may have virtually any amino acid sequence, bearing in mind that suitable linkers will have a sequence that results in a generally flexible peptide. The use of small amino acids, such as glycine and alanine, are of use in creating a flexible peptide. The creation of such sequences is routine to those of skill in the art.
[0096] Suitable linkers can be readily selected and can be of any of a suitable of different lengths, such as from 1 amino acid (e.g., Gly) to 20 amino acids, from 2 amino acids to 15 amino acids, from 3 amino acids to 12 amino acids, including 4 amino acids to 10 amino acids, 5 amino acids to 9 amino acids, 6 amino acids to 8 amino acids, or 7 amino acids to 8 amino acids, and may be 1, 2, 3, 4, 5, 6, or 7 amino acids.
[0097] Exemplary flexible linkers include glycine polymers (G)n, glycine- serine polymers
(including, for example, (GS)n, GSGGSn (SEQ ID NO:37) and GGGSn (SEQ ID NO:38), where n is an integer of at least one), glycine- alanine polymers, alanine-serine polymers, and other flexible linkers known in the art. Glycine and glycine- serine polymers are of interest since both of these amino acids are relatively unstructured, and therefore may serve as a neutral tether between components. Glycine polymers are of particular interest since glycine accesses significantly more phi-psi space than even alanine, and is much less restricted than residues with longer side chains (see Scheraga, Rev. Computational Chem. 11173-142 (1992)). Exemplary flexible linkers include, but are not limited GGSG (SEQ ID NO:39), GGSGG (SEQ ID NO:40), GSGSG (SEQ ID NO:41), GSGGG (SEQ ID NO:42), GGGSG (SEQ ID NO:43), GSSSG (SEQ ID NO:44), and the like. The ordinarily skilled artisan will recognize that design of a peptide conjugated to any elements described above can include linkers that are all or partially flexible, such that the linker can include a flexible linker as well as one or more portions that confer less flexible structure.
Modulatory domains
[0098] Modulatory domains suitable for use in a CAR of the present disclosure include co- stimulatory domains.
[0099] In some cases, the modulatory domain on the first polypeptide of a subject CAR has
substantially the same amino acid sequence as the modulatory domain on the second polypeptide of the CAR. For example, in some cases, the modulatory domain on the first polypeptide of a CAR comprises an amino acid sequence that is at least about 90%, at least about 95%, at least about 98%, at least about 99%, or 100%, identical to the amino acid sequence of the modulatory domain on the second polypeptide of the CAR. The modulatory domain of the first polypeptide of a subject CAR can have substantially the same length as the modulatory domain of the second polypeptide of a subject CAR; e.g., the first and second modulatory domains can differ in length from one another by fewer than 10 amino acids, or fewer than 5 amino acids. In some cases, the first and second modulatory domains have the same length.
[00100] A modulatory domain suitable for inclusion in the first and the second polypeptide of a subject CAR can have a length of from about 30 amino acids to about 70 amino acids (aa), e.g., a modulatory domain can have a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa. In other cases, modulatory domain can have a length of from about 70 aa to about 100 aa, from about 100 aa to about 200 aa, or greater than 200 aa.
[00101] Co- stimulatory domains suitable for use in a CAR of the present disclosure are
generally polypeptides derived from receptors. In some embodiments, co- stimulatory domains homodimerize. A subject co- stimulatory domain can be an intracellular portion of a transmembrane protein (i.e., the co- stimulatory domain can be derived from a
transmembrane protein). Non-limiting examples of suitable co-stimulatory polypeptides include, but are not limited to, 4-lBB (CD137), CD28, ICOS, OX-40, BTLA, CD27, CD30, GITR, and HVEM.
[00102] In some cases, the co-stimulatory domain on the first polypeptide of a subject CAR has substantially the same amino acid sequence as the co-stimulatory domain on the second polypeptide of the CAR. For example, in some cases, the co- stimulatory domain on the first polypeptide of a CAR comprises an amino acid sequence that is at least about 90%, at least about 95%, at least about 98%, at least about 99%, or 100%, identical to the amino acid sequence of the co- stimulatory domain on the second polypeptide of the CAR. The co- stimulatory domain of the first polypeptide of a subject CAR can have substantially the same length as the co- stimulatory domain of the second polypeptide of a subject CAR; e.g., the first and second co- stimulatory domains can differ in length from one another by fewer than 10 amino acids, or fewer than 5 amino acids. In some cases, the first and second co- stimulatory domains have the same length.
[00103] A co- stimulatory domain suitable for inclusion in the first and the second
polypeptide of a subject CAR can have a length of from about 30 amino acids to about 70 amino acids (aa), e.g., a co- stimulatory domain can have a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa. In other cases, the co- stimulatory domain can have a length of from about 70 aa to about 100 aa, from about 100 aa to about 200 aa, or greater than 200 aa.
[00104] In some cases, the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein 4-1BB (also known as TNFRSF9; CD137; 4-1BB; CDwl37; ILA; etc.). For example, a suitable co-stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL (SEQ ID NO:24). In some of these embodiments, the co- stimulatory domain of both the first and the second polypeptide has a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
[00105] In some cases, the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein CD28 (also known as Tp44). For example, a suitable co- stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
FWVRSKRSRLLHSDYMNMTPRRPGPTRKHYQPYAPPRDFAAYRS (SEQ ID NO:63). In some of these embodiments, the co- stimulatory domain of both the first and the second polypeptide has a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
[00106] In some cases, the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein ICOS (also known as AILIM, CD278, and CVID1). For example, a suitable co-stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
TKKKYSSSVHDPNGEYMFMRAVNTAKKSRLTDVTL (SEQ ID NO:64). In some of these embodiments, the co- stimulatory domain of both the first and the second polypeptide has a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
[00107] In some cases, the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein OX-40 (also known as TNFRSF4, RP5-902P8.3, ACT35, CD134, OX40, TXGP1L). For example, a suitable co- stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
RRDQRLPPDAHKPPGGGSFRTPIQEEQADAHSTLAKI (SEQ ID NO:65). In some of these embodiments, the co- stimulatory domain of both the first and the second polypeptide has a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
[00108] In some cases, the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein BTLA (also known as BTLA1 and CD272). For example, a suitable co-stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence: CCLRRHQGKQNELSDTAGREINLVDAHLKSEQTEASTRQNSQVLLSETGIYDNDPD LCFRMQEGSEVYSNPCLEENKPGIVYASLNHSVIGPNSRLARNVKEAPTEYASICVR
S (SEQ ID NO:66).
[00109] In some cases, the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein CD27 (also known as S 152, T14, TNFRSF7, and Tp55). For example, a suitable co-stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
HQRRKYRSNKGESPVEPAEPCRYSCPREEEGSTIPIQEDYRKPEPACSP (SEQ ID NO:67). In some of these embodiments, the co- stimulatory domain of both the first and the second polypeptide has a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
[00110] In some cases, the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein CD30 (also known as TNFRSF8, D1S166E, and Ki-1). For example, a suitable co-stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, from about 150 aa to about 160 aa, or from about 160 aa to about 185 aa of the following amino acid sequence:
RRACRKRIRQKLHLCYPVQTSQPKLELVDSRPRRSSTQLRSGASVTEPVAEERGLMS QPLMETCHSVGAAYLESLPLQDASPAGGPSSPRDLPEPRVSTEHTNNKIEKIYIMKA DTVIVGTVKAELPEGRGLAGPAEPELEEELEADHTPHYPEQETEPPLGSCSDVMLSV EEEGKEDPLPTAASGK (SEQ ID NO:68).
[00111] In some cases, the co-stimulatory domain is derived from an intracellular portion of the transmembrane protein GITR (also known as TNFRSF18, RP5-902P8.2, AITR, CD357, and GITR-D). For example, a suitable co- stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
HIWQLRSQCMWPRETQLLLEVPPSTEDARSCQFPEEERGERSAEEKGRLGDLWV
(SEQ ID NO:69). In some of these embodiments, the co-stimulatory domain of both the first and the second polypeptide has a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
[00112] In some cases, the co-stimulatory domain derived from an intracellular portion of the transmembrane protein HVEM (also known as TNFRSF14, RP3-395M20.6, ATAR, CD270, HVEA, HVEM, LIGHTR, and TR2). For example, a suitable co-stimulatory domain can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
CVKRRKPRGDVVKVIVSVQRKRQEAEGEATVIEALQAPPDVTTVAVEETIPSFTGRS PNH (SEQ ID NO:70). In some of these embodiments, the co-stimulatory domain of both the first and the second polypeptide has a length of from about 30 aa to about 35 aa, from about 35 aa to about 40 aa, from about 40 aa to about 45 aa, from about 45 aa to about 50 aa, from about 50 aa to about 55 aa, from about 55 aa to about 60 aa, from about 60 aa to about 65 aa, or from about 65 aa to about 70 aa.
Dimer pairs
[00113] Dimer pairs suitable for use in a subject CAR include dimerizer-binding pairs.
Dimerizer-binding pairs suitable for use in a CAR of the present disclosure are in some embodiments polypeptides that bind to a different site of the same molecule (referred to herein as a "dimerizer"). In the presence of a dimerizer, both members of the dimerizer- binding pair bind to a different site of the dimerizer and are thus brought into proximity with one another. In some embodiments, binding to the dimerizer is reversible. In some embodiments, binding to the dimerizer is irreversible. In some embodiments, binding to the dimerizer is non-covalent. In some embodiments, binding to the dimerizer is covalent.
[00114] Other dimer pairs suitable for use include dimerizer-binding pairs that dimerize upon binding of a first member of a dimer pair to a dimerizing agent, where the dimerizing agent induces a conformational change in the first member of the dimer pair, and where the conformational change allows the first member of the dimer pair to bind (covalently or non- covalently) to a second member of the dimer pair.
[00115] Other dimer pairs suitable for use include dimer pairs in which exposure to light
(e.g., blue light) induces dimerization of the dimer pair.
[00116] Regardless of the mechanism, the dimer pair will dimerize upon exposure to an agent that induces dimerization, where the agent is in some cases a small molecule, or, in other cases, light. Thus, for simplicity, the discussion below referring to "dimerizer-binding pairs" includes dimer pairs that dimerize regardless of the mechanism.
[00117] Non-limiting examples of suitable dimers (e.g., dimerizer-binding pairs) include, but are not limited to:
a) FK506 binding protein (FKBP) and FKBP;
b) FKBP and calcineurin catalytic subunit A (CnA);
c) FKBP and cyclophilin;
d) FKBP and FKBP-rapamycin associated protein (FRB);
e) gyrase B (GyrB) and GyrB;
f) dihydrofolate reductase (DHFR) and DHFR;
g) DmrB and DmrB; h) PYL and ABI;
i) Cry2 and CIB1; and
j) GAI and GID1.
[00118] A first or a second member of a dimer (e.g., a dimerizer-binding pair) of a subject
CAR can have a length of from about 50 amino acids to about 300 amino acids or more; e.g., a first or a second member of a dimer (e.g., a dimerizer-binding pair) of a subject CAR can have a length of from about 50 aa to about 100 aa, from about 100 aa to about 150 aa, from about 150 aa to about 200 aa, from about 200 aa toa bout 250 aa, from about 250 aa to about 300 aa, or more than 300 aa.
[00119] In some cases, a member of a dimer (e.g., a dimerizer-binding pair) of a subject
CAR is derived from FKBP. For example, a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
MGVQVETISPGDGRTFPKRGQTCVVHYTGMLEDGKKFDSSRDRNKPFKFMLGKQE VIRGWEEGVAQMSVGQRAKLTISPDYAYGATGHPGIIPPHATLVFDVELLKLE (SEQ ID NO: 12).
[00120] In some cases, a member of a dimerizer-binding pair of a subject CAR is derived from calcineurin catalytic subunit A (also known as PPP3CA; CALN; CALNA; CALNA1; CCN1; CNA1; PPP2B; CAM-PRP catalytic subunit; calcineurin A alpha; calmodulin- dependent calcineurin A subunit alpha isoform; protein phosphatase 2B, catalytic subunit, alpha isoform; etc.). For example, a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence (PP2Ac domain):
LEESVALRIITEGASILRQEKNLLDIDAPVTVCGDIHGQFFDLMKLFEVGGSPANTRY LFLGDYVDRGYFSIECVLYLWALKILYPKTLFLLRGNHECRHLTEYFTFKQECKIKY SERVYDACMDAFDCLPLAALMNQQFLCVHGGLSPEINTLDDIRKLDRFKEPPAYGP MCDILWSDPLEDFGNEKTQEHFTHNTVRGCSYFYSYPAVCEFLQHNNLLSILRAHE AQDAGYRMYRKSQTTGFPSLITIFSAPNYLDVYNNKAAVLKYENNVMNIRQFNCSP HPYWLPNFM (SEQ ID NO:71).
[00121] In some cases, a member of a dimer (e.g., a dimerizer-binding pair) is derived from cyclophilin (also known cyclophilin A, PPIA, CYPA, CYPH, PPIase A, etc.). For example, a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
MVNPTVFFDIAVDGEPLGRVSFELFADKVPKTAENFRALSTGEKGFGYKGSCFHRII PGFMCQGGDFTRHNGTGGKSIYGEKFEDENFILKHTGPGILSMANAGPNTNGSQFFI CTAKTEWLDGKHVVFGKVKEGMNIVEAMERFGSRNGKTSKKITIADCGQLE (SEQ ID NO:72).
[00122] In some cases, a member of a dimer (e.g., a dimerizer-binding pair) is derived from
MTOR (also known as FKBP-rapamycin associated protein; FK506 binding protein 12- rapamycin associated protein 1; FK506 binding protein 12-rapamycin associated protein 2; FK506-binding protein 12-rapamycin complex-associated protein 1; FRAP; FRAP1;
FRAP2; RAFT1; and RAPT1). For example, a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence (also known as "Frb": Fkbp- Rapamycin Binding Domain):
MILWHEMWHEGLEEASRLYFGERNVKGMFEVLEPLHAMMERGPQTLKETSFNQA YGRDLMEAQEWCRKYMKSGNVKDLLQAWDLYYHVFRRISK (SEQ ID NO: 14).
[00123] In some cases, a member of a dimer (e.g., a dimerizer-binding pair) is derived from
GyrB (also known as DNA gyrase subunit B). For example, a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 200 amino acids (aa), from about 200 aa to about 300 aa, from about 300 aa to about 400 aa, from about 400 aa to about 500 aa, from about 500 aa to about 600 aa, from about 600 aa to about 700 aa, or from about 700 aa to about 800 aa, of the following GyrB amino acid sequence from Escherichia coli (or to the DNA gyrase subunit B sequence from any organism):
MSNSYDSSSIKVLKGLDAVRKRPGMYIGDTDDGTGLHHMVFEVVDNAIDEALAGH CKEIIVTIHADNSVSVQDDGRGIPTGIHPEEGVSAAEVIMTVLHAGGKFDDNSYKVS GGLHGVGVSVVNALSQKLELVIQREGKIHRQIYEHGVPQAPLAVTGETEKTGTMV RFWPSLETFTNVTEFEYEILAKRLRELSFLNSGVSIRLRDKRDGKEDHFHYEGGIKAF VEYLNKNKTPIHPNIFYFSTEKDGIGVEVALQWNDGFQENIYCFTNNIPQRDGGTHL AGFRAAMTRTLNAYMDKEGYSKKAKVSATGDDAREGLIAVVSVKVPDPKFSSQT KDKLVSSEVKSAVEQQMNELLAEYLLENPTDAKIVVGKIIDAARAREAARRAREM TRRKGALDLAGLPGKLADCQERDPALSELYLVEGDSAGGSAKQGRNRKNQAILPL KGKILNVEKARFDKMLSSQEVATLITALGCGIGRDEYNPDKLRYHSIIIMTDADVDG SHIRTLLLTFFYRQMPEIVERGHVYIAQPPLYKVKKGKQEQYIKDDEAMDQYQISIA LDGATLHTNASAPALAGEALEKLVSEYNATQKMINRMERRYPKAMLKELIYQPTL TEADLSDEQTVTRWVNALVSELNDKEQHGSQWKFDVHTNAEQNLFEPIVRVRTHG VDTDYPLDHEFITGGEYRRICTLGEKLRGLLEEDAFIERGERRQPVASFEQALDWLV KESRRGLSIQRYKGLGEMNPEQLWETTMDPESRRMLRVTVKDAIAADQLFTTLMG DAVEPRRAFIEENALKAANIDI (SEQ ID NO:73). In some cases, a member of a dimerizer-binding pair comprises an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to amino acids 1-220 of the above-listed GyrB amino acid sequence from Escherichia coli.
[00124] In some cases, a member of a dimer (e.g., a dimerizer-binding pair) is derived from
DHFR (also known as dihydrofolate reductase, DHFRP1, and DYR). For example, a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
M VGS LNCIV A VS QNMGIGKNGDLPWPPLRNEFR YFQRMTTTS S VEGKQNLVIMGK KTWFSIPEKNRPLKGRINLVLSRELKEPPQGAHFLSRSLDDALKLTEQPELANKVDM VWrVGGSSVYKEAMNHPGHLKLFVTRIMQDFESDTFFPEIDLEKYKLLPEYPGVLS DVQEEKGIKYKFEVYEKND (SEQ ID NO:74).
[00125] In some cases, a member of a dimer (e.g., a dimerizer-binding pair) is derived from the DmrB binding domain (i.e., DmrB homodimerization domain). For example, a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence: MASRGVQVETISPGDGRTFPKRGQTCVVHYTGMLEDGKKVDSSRDRNKPFKFMLG KQEVIRGWEEGVAQMSVGQRAKLTISPDYAYGATGHPGIIPPHATLVFDVELLKLE
(SEQ ID NO:75).
[00126] In some cases, a member of a dimer (e.g., a dimerizer-binding pair) is derived from a
PYL protein (also known as abscisic acid receptor and as RCAR). For example a member of a subject dimerizer-binding pair can be derived from proteins such as those of Arabidopsis thaliana: PYR1, RCAR1(PYL9), PYL1, PYL2, PYL3, PYL4, PYL5, PYL6, PYL7, PYL8 (RCAR3), PYL10, PYL11, PYL12, PYL13. For example, a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to any of the following amino acid sequences:
[00127] PYL10:
MNGDETKKVESEYIKKHHRHELVESQCSSTLVKHIKAPLHLVWSIVRRFDEPQKYK PFISRCVVQGKKLEVGSVREVDLKSGLPATKSTEVLEILDDNEHILGIRIVGGDHRLK NYSSTISLHSETIDGKTGTLAIESFVVDVPEGNTKEETCFFVEALIQCNLNSLADVTE RLQAESMEKKI (SEQ ID NO:76).
[00128] PYL11:
METSQKYHTCGSTLVQTIDAPLSLVWSILRRFDNPQAYKQFVKTCNLSSGDGGEGS VREVTVVSGLPAEFSRERLDELDDESHVMMISIIGGDHRLVNYRSKTMAFVAADTE EKTVVVESYVVDVPEGNSEEETTSFADTIVGFNLKSLAKLSERVAHLKL (SEQ ID NO:77)
[00129] PYL12:
MKTSQEQHVCGSTVVQTINAPLPLVWSILRRFDNPKTFKHFVKTCKLRSGDGGEGS VREVTVVSDLPASFSLERLDELDDESHVMVISIIGGDHRLVNYQSKTTVFVAAEEEK TVVVESYVVDVPEGNTEEETTLFADTIVGCNLRSLAKLSEKMMELT (SEQ ID NO:78).
[00130] PYL13:
MESSKQKRCRSSVVETIEAPLPLVWSILRSFDKPQAYQRFVKSCTMRSGGGGGKGG EGKGSVRDVTLVSGFPADFSTERLEELDDESHVMVVSIIGGNHRLVNYKSKTKVVA SPEDMAKKTVVVESYVVDVPEGTSEEDTIFFVDNIIRYNLTSLAKLTKKMMK (SEQ ID NO:79).
[00131] PYL1:
MANSES S S SP VNEEENS QRISTLHHQTMPSDLTQDEFTQLS QS IAEFHT YQLGNGRC SSLLAQRIHAPPETVWSVVRRFDRPQIYKHFIKSCNVSEDFEMRVGCTRDVNVISGL PANTSRERLDLLDDDRRVTGFSITGGEHRLRNYKSVTTVHRFEKEEEEERr TVVLE SYVVDVPEGNSEEDTRLFADTVIRLNLQKLASITEAMNRNNNNNNSSQVR (SEQ ID NO:80).
[00132] PYL2:
MSSSPAVKGLTDEEQKTLEPVIKTYHQFEPDPTTCTSLITQRIHAPASVVWPLIRRFD NPERYKHFVKRCRLISGDGDVGSVREVTVISGLPASTSTERLEFVDDDHRVLSFRVV GGEHRLKNYKSVTSVNEFLNQDSGKVYTVVLESYTVDIPEGNTEEDTKMFVDTVV KLNLQKLGVAATSAPMHDDE (SEQ ID NO:81).
[00133] PYL3:
MNLAPIHDPSSSSTTTTSSSTPYGLTKDEFSTLDSIIRTHHTFPRSPNTCTSLIAHRVDA PAHAr RFVRDFANPNKYKHFIKSCTIRVNGNGIKEIKVGTIREVSVVSGLPASTSVE ILEVLDEEKRILSFRVLGGEHRLNNYRSVTSVNEFVVLEKDKKKRVYSVVLESYIVD IPQGNTEEDTRMFVDTVVKSNLQNLAVISTASPT (SEQ ID NO:82).
[00134] PYL4:
MLAVHRPSSAVSDGDSVQIPMMIASFQKRFPSLSRDSTAARFHTHEVGPNQCCSAVI QEISAPISTVWSVVRRFDNPQAYKHFLKSCSVIGGDGDNVGSLRQVHVVSGLPAAS STERLDILDDERHVISFSVVGGDHRLSNYRSVTTLHPSPISGTVVVESYVVDVPPGNT KEETCDFVDVIVRCNLQSLAKIAENTAAESKKKMSL (SEQ ID NO:83).
[00135] PYL5:
MRSPVQLQHGSDATNGFHTLQPHDQTDGPIKRVCLTRGMHVPEHVAMHHTHDVG PDQCCSSVVQMIHAPPESVWALVRRFDNPKVYKNFIRQCRIVQGDGLHVGDLREV MVVSGLPAVSSTERLEILDEERHVISFSVVGGDHRLKNYRSVTTLHASDDEGTVVV ESYrVDVPPGNTEEETLSFVDTIVRCNLQSLARSTNRQ (SEQ ID NO:84).
[00136] PYL6:
MPTSIQFQRSSTAAEAANATVRNYPHHHQKQVQKVSLTRGMADVPEHVELSHTHV VGPSQCFSVVVQDVEAPVSTVWSILSRFEHPQAYKHFVKSCHVVIGDGREVGSVRE VRVVSGLPAAFSLERLEIMDDDRHVISFSVVGGDHRLMNYKSVTTVHESEEDSDGK KRTRVVESYVVDVPAGNDKEETCSFADTIVRCNLQSLAKLAENTSKFS (SEQ ID NO:85).
[00137] PYL7:
MEMIGGDDTDTEMYGALVTAQSLRLRHLHHCRENQCTSVLVKYIQAPVHLVWSL VRRFDQPQKYKPFISRCTVNGDPEIGCLREVNVKSGLPATTSTERLEQLDDEEHILGI NIIGGDHRLKNYSSILTVHPEMIDGRSGTMVMESFVVDVPQGNTKDDTCYFVESLIK CNLKSLACVSERLAAQDITNSIATFCNASNGYREKNHTETNL (SEQ ID NO:86).
[00138] PYL8:
MEANGIENLTNPNQEREFIRRHHKHELVDNQCSSTLVKHINAPVHIVWSLVRRFDQ PQKYKPFISRCVVKGNMEIGTVREVDVKSGLPATRSTERLELLDDNEHILSIRIVGGD HRLKNYSSIISLHPETIEGRIGTLVIESFVVDVPEGNTKDETCYFVEALIKCNLKSLAD ISERLAVQDTTESRV (SEQ ID NO: 87). [00139] PYL9:
MMDGVEGGTAMYGGLETVQYVRTHHQHLCRENQCTSALVKHIKAPLHLVWSLV RRFDQPQKYKPFVSRCTVIGDPEIGSLREVNVKSGLPATTSTERLELLDDEEHILGIKI IGGDHRLKNYSSILTVHPEIIEGRAGTMVIESFVVDVPQGNTKDETCYFVEALIRCNL KSLADVSERLASQDITQ (SEQ ID NO:88).
[00140] PYR1:
MPSELTPEERSELKNSIAEFHTYQLDPGSCSSLHAQRIHAPPELVWSIVRRFDKPQTY KHFIKSCSVEQNFEMRVGCTRDVIVISGLPANTSTERLDILDDERRVTGFSIIGGEHR LTNYKSVTTVHRFEKENRIWTVVLESYVVDMPEGNSEDDTRMFADTVVKLNLQKL ATVAEAMARNSGDGSGSQVT (SEQ ID NO: 89).
[00141] In some cases, a member of a dimer (e.g., a dimerizer-binding pair) is derived from an ABI protein (also known as Abscisic Acid- Insensitive). For example a member of a subject dimerizer-binding pair can be derived from proteins such as those of Arabidopsis thaliana: ABI1 (Also known as ABSCISIC ACID-INSENSITIVE 1, Protein phosphatase 2C 56, AtPP2C56, P2C56, and PP2C ABI1) and/or ABI2(also known as P2C77, Protein phosphatase 2C 77, AtPP2C77, ABSCISIC ACID-INSENSITIVE 2, Protein phosphatase 2C ABI2, and PP2C ABI2). For example, a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, from about 150 aa to about 160 aa, from about 160 aa to about 170 aa, from about 170 aa to about 180 aa, from about 180 aa to about 190 aa, or from about 190 aa to about 200 aa of any of the following amino acid sequences:
[00142] ABI1:
MEEVSPAIAGPFRPFSETQMDFTGIRLGKGYCNNQYSNQDSENGDLMVSLPETSSCS VSGSHGSESRKVLISRINSPNLNMKESAAADIVVVDISAGDEINGSDITSEKKMISRT ESRSLFEFKSVPLYGFTSICGRRPEMEDAVSTIPRFLQSSSGSMLDGRFDPQSAAHFF GVYDGHGGSQVANYCRERMHLALAEEIAKEKPMLCDGDTWLEKWKKALFNSFLR VDSEIESVAPETVGSTSVVAVVFPSHIFVANCGDSRAVLCRGKTALPLSVDHKPDRE DEAARIEAAGGKVIQWNGARVFGVLAMSRSIGDRYLKPSIIPDPEVTAVKRVKEDD CLILASDGVWDVMTDEEACEMARKRILLWHKKNAVAGDASLLADERRKEGKDPA AMS A AE YLS KLAIQRGS KDNIS V V V VDLKPRRKLKS KPLN (SEQ ID NO:90). [00143] ABI2:
MDEVSPAVAVPFRPFTDPHAGLRGYCNGESRVTLPESSCSGDGAMKDSSFEINTRQ
DSLTSSSSAMAGVDISAGDEINGSDEFDPRSMNQSEKKVLSRTESRSLFEFKCVPLY
GVTSICGRRPEMEDSVSTIPRFLQVSSSSLLDGRVTNGFNPHLSAHFFGVYDGHGGS
QVANYCRERMHLALTEEIVKEKPEFCDGDTWQEKWKKALFNSFMRVDSEIETVAH
APETVGSTSVVAVVFPTHIFVANCGDSRAVLCRGKTPLALSVDHKPDRDDEAARIE
AAGGKVIRWNGARVFGVLAMSRSIGDRYLKPSVIPDPEVTSVRRVKEDDCLILASD
GLWDVMTNEEVCDLARKRILLWHKKNAMAGEALLPAEKRGEGKDPAAMSAAEY
LSKMALQKGSKDNISVVVVDLKGIRKFKSKSLN (SEQ ID NO:91).
[00144] In some cases, a member of a dimer (e.g., a dimerizer-binding pair) is derived from a
Cry2 protein (also known as cryptochrome 2). For example a member of a subject dimer (e.g., a dimerizer-binding pair) can be derived from Cry2 proteins from any organism (e.g., a plant) such as, but not limited to, those of Arabidopsis thaliana. For example, a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, from about 150 aa to about 160 aa, from about 160 aa to about 170 aa, from about 170 aa to about 180 aa, from about 180 aa to about 190 aa, or from about 190 aa to about 200 aa of any of the following amino acid sequences:
[00145] Cry2 (Arabidopsis thaliana)
MKMDKKTIVWFRRDLRIEDNPALAAAAHEGSVFPVFr CPEEEGQFYPGRASRWW
MKQS LAHLS QSLK ALGSDLTLIKTHNTIS AILDCIRVTG ATKV VFNHLYDP VS LVRD
HTVKEKLVERGISVQSYNGDLLYEPWEIYCEKGKPFTSFNSYWKKCLDMSIESVML
PPPWRLMPITAAAEAr ACSIEELGLENEAEKPSNALLTRAWSPGWSNADKLLNEFI
EKQLIDYAKNSKKVVGNSTSLLSPYLHFGEISVRHVFQCARMKQIIWARDKNSEGE
ESADLFLRGIGLREYSRYICFNFPFTHEQSLLSHLRFFPWDADVDKFKAWRQGRTG
YPLVDAGMRELWATGWMHNRIRVIVSSFAVKFLLLPWKWGMKYFWDTLLDADL
ECDILGWQYISGSIPDGHELDRLDNPALQGAKYDPEGEYIRQWLPELARLPTEWIHH
PWDAPLTVLKASGVELGTNYAKPIVDIDTARELLAKAISRTREAQIMIGAAPDEIVA
DSFEALGANTIKEPGLCPSVSSNDQQVPSAVRYNGSKRVKPEEEEERDMKKSRGFD
ERELFSTAESSSSSSVFFVSQSCSLASEGKNLEGIQDSSDQITTSLGKNGCK (SEQ ID
NO:92). [00146] In some cases, a member of a dimer (e.g., a dimerizer-binding pair) is derived from the CIB1 Arabidopsis thaliana protein (also known as transcription factor bHLH63). For example, a suitable dimer (e.g., a dimerizer-binding pair) member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, from about 150 aa to about 160 aa, from about 160 aa to about 170 aa, from about 170 aa to about 180 aa, from about 180 aa to about 190 aa, or from about 190 aa to about 200 aa of the following amino acid sequence:
MNGAIGGDLLLNFPDMSVLERQRAHLKYLNPTFDSPLAGFFADSSMITGGEMDSYL
STAGLNLPMMYGETTVEGDSRLSISPETTLGTGNFKKRKFDTETKDCNEKKKKMT
MNRDDLVEEGEEEKSKITEQNNGSTKSIKKMKHKAKKEENNFSNDSSKVTKELEKT
DYIHVRARRGQATDSHSIAERVRREKISERMKFLQDLVPGCDKITGKAGMLDEIINY
VQSLQRQIEFLSMKLAIVNPRPDFDMDDIFAKEVASTPMTVVPSPEMVLSGYSHEM
VHSGYSSEMVNSGYLHVNPMQQVNTSSDPLSCFNNGEAPSMWDSHVQNLYGNLG
V (SEQ ID NO:93).
[00147] In some cases, a member of a dimer (e.g., a dimerizer-binding pair) is derived from the G AI Arabidopsis thaliana protein (also known as Gibberellic Acid Insensitive, and DELLA protein GAI). For example, a suitable dimerizer-binding pair member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, from about 150 aa to about 160 aa, from about 160 aa to about 170 aa, from about 170 aa to about 180 aa, from about 180 aa to about 190 aa, or from about 190 aa to about 200 aa of the following amino acid sequence:
MKRDHHHHHHQDKKTMMMNEEDDGNGMDELLAVLGYKVRSSEMADVAQKLEQ LEVMMSNVQEDDLSQLATETVHYNPAELYTWLDSMLTDLNPPSSNAEYDLKAIPG DAILNQFAIDSASSSNQGGGGDTYTTNKRLKCSNGVVETTTATAESTRHVVLVDSQ ENGVRLVHALLACAEAVQKENLTVAEALVKQIGFLAVSQIGAMRKVATYFAEALA RRIYRLSPSQSPIDHSLSDTLQMHFYETCPYLKFAHFTANQAILEAFQGKKRVHVIDF SMSQGLQWPALMQALALRPGGPPVFRLTGIGPPAPDNFDYLHEVGCKLAHLAEAIH VEFEYRGFVANTLADLDASMLELRPSEIESVAVNSVFELHKLLGRPGAIDKVLGVV NQIKPEIFTVVEQESNHNSPIFLDRFTESLHYYSTLFDSLEGVPSGQDKVMSEVYLGK QICNVVACDGPDRVERHETLSQWRNRFGSAGFAAAHIGSNAFKQASMLLALFNGG EGYRVEESDGCLMLGWHTRPLIATSAWKLSTN (SEQ ID NO:94).
[00148] In some cases, a member of a dimer (e.g., a dimerizer-binding pair) is derived from a
GID1 Arabidopsis thaliana protein (also known as Gibberellin receptor GID1). For example, a suitable dimer member can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, from about 150 aa to about 160 aa, from about 160 aa to about 170 aa, from about 170 aa to about 180 aa, from about 180 aa to about 190 aa, or from about 190 aa to about 200 aa of any of the following amino acid sequences:
[00149] GID1A:
MAASDEVNLIESRTVVPLNTWVLISNFKVAYNILRRPDGTFNRHLAEYLDRKVTAN ANPVDGVFSFDVLIDRRINLLSRVYRPAYADQEQPPSILDLEKPVDGDIVPVILFFHG GSFAHSSANSAIYDTLCRRLVGLCKCVVVSVNYRRAPENPYPCAYDDGWIALNWV NSRSWLKSKKDSKVHIFLAGDSSGGNIAHNVALRAGESGIDVLGNILLNPMFGGNE RTESEKSLDGKYFVTVRDRDWYWKAFLPEGEDREHPACNPFSPRGKSLEGVSFPKS LVVVAGLDLIRDWQLAYAEGLKKAGQEVKLMHLEKATVGFYLLPNNNHFHNVM DEISAFVNAEC (SEQ ID NO:95).
[00150] GID1B:
MAGGNEVNLNECKRIVPLNTWVLISNFKLAYKVLRRPDGSFNRDLAEFLDRKVPA NSFPLDGVFSFDHVDSTTNLLTRIYQPASLLHQTRHGTLELTKPLSTTEIVPVLIFFHG GSFTHSSANSAIYDTFCRRLVTICGVVVVSVDYRRSPEHRYPCAYDDGWNALNWV KSRVWLQSGKDSNVYVYLAGDSSGGNIAHNVAVRATNEGVKVLGNILLHPMFGG QERTQSEKTLDGKYFVTIQDRDWYWRAYLPEGEDRDHPACNPFGPRGQSLKGVNF PKSLVVVAGLDLVQDWQLAYVDGLKKTGLEVNLLYLKQATIGFYFLPNNDHFHCL MEELNKFVHSIEDSQSKSSPVLLTP (SEQ ID NO:96)
[00151] GID1C:
MAGSEEVNLIESKTVVPLNTWVLISNFKLAYNLLRRPDGTFNRHLAEFLDRKVPAN ANPVNGVFSFDVIIDRQTNLLSRVYRPADAGTSPSITDLQNPVDGEIVPVIVFFHGGS FAHSSANSAIYDTLCRRLVGLCGAVVVSVNYRRAPENRYPCAYDDGWAVLKWVN
SSSWLRSKKDSKVRIFLAGDSSGGNIVHNVAVRAVESRIDVLGNILLNPMFGGTERT
ESEKRLDGKYFVTVRDRDWYWRAFLPEGEDREHPACSPFGPRSKSLEGLSFPKSLV
VVAGLDLIQDWQLKYAEGLKKAGQEVKLLYLEQATIGFYLLPNNNHFHTVMDEIA
AFVNAECQ (SEQ ID NO:97).
Dimerizers
[00152] Dimerizers ("dimerizing agents) that can provide for dimerization of a first member of a dimerizer-binding pair and a second member of a dimerizer-binding pair include, e.g. (where the dimerizer is in parentheses following the dimerizer-binding pair:
a) FKBP and FKBP (rapamycin);
b) FKBP and CnA (rapamycin);
c) FKBP and cyclophilin (rapamycin);
d) FKBP and FRG (rapamycin);
e) GyrB and GyrB (coumermycin);
f) DHFR and DHFR (methotrexate);
g) DmrB and DmrB (AP20187);
h) PYL and ABI (abscisic acid);
i) Cry2 and CIB1 (blue light); and
j) GAI and GID1 (gibberellin).
[00153] As noted above, rapamycin can serve as a dimerizer. Alternatively, a rapamycin derivative or analog can be used. See, e.g., W096/41865; WO 99/36553; WO 01/14387; and Ye et al (1999) Science 283:88-91. For example, analogs, homologs, derivatives and other compounds related structurally to rapamycin ("rapalogs") include, among others, variants of rapamycin having one or more of the following modifications relative to rapamycin: demethylation, elimination or replacement of the methoxy at C7, C42 and/or C29; elimination, derivatization or replacement of the hydroxy at CI 3, C43 and/or C28; reduction, elimination or derivatization of the ketone at C14, C24 and/or C30; replacement of the 6-membered pipecolate ring with a 5-membered prolyl ring; and alternative substitution on the cyclohexyl ring or replacement of the cyclohexyl ring with a substituted cyclopentyl ring. Additional information is presented in, e.g., U.S. Pat. Nos. 5,525,610; 5,310,903 5,362,718; and 5,527,907. Selective epimerization of the C-28 hydroxyl group has been described; see, e.g., WO 01/14387. Additional synthetic dimerizing agents suitable for use as an alternative to rapamycin include those described in U.S. Patent Publication No. 2012/0130076. [00154] Rapamycin has the structure:
Figure imgf000034_0001
28-epi rapamycin
[00156] Also suitable as a rapalog is a compound of the formula:
Figure imgf000034_0002
[00157] where n is 1 or 2; R and R are independently H, or a substituted or unsubstituted aliphatic or acyl moiety; one of R7a and R7b is H and the other is halo, RA, ORA, SRA, - OC(0)RA, -OC(0)NRARB, -NRARB, -NRBC(OR)RA, NRBC(0)ORA, -NRBS02RA, or NRBS02NRARB ; or R7a and R7b, taken together, are H in the tetraene moiety:
Figure imgf000035_0001
[00158] where R is H or a substituted or unsubstituted aliphatic, hetero aliphatic, aryl, or heteroaryl moiety and where R B and R B' are independently H, OH, or a substituted or unsubstituted aliphatic, heteroaliphatic, aryl, or heteroaryl moiety.
[00159] As noted above, coumermycin can serve as a dimerizing agent. Alternatively, a
coumermycin analog can be used. See, e.g., Farrar et al. (1996) Nature 383: 178-181; and U.S. Pat. No. 6,916,846.
[00160] As noted above, in some cases, the dimerizing agent is methotrexate, e.g., a non- cytotoxic, homo-bifunctional methotrexate dimer. See, e.g., U.S. Patent No. 8,236,925. Intracellular signaling domain
[00161] Intracellular signaling domains suitable for use in a CAR of the present disclosure include any desired signaling domain that provides a distinct and detectable signal (e.g., increased production of one or more cytokines by the cell; change in transcription of a target gene; change in activity of a protein; change in cell behavior, e.g., cell death; cellular proliferation; cellular differentiation; cell survival; modulation of cellular signaling responses; etc.) in response to activation of the CAR (i.e., activated by antigen and dimerizing agent). In some embodiments, the intracellular signaling domain includes at least one (e.g., one, two, three, four, five, six, etc.) IT AM motifs as described below. In some embodiments, the intracellular signaling domain includes DAP10/CD28 type signaling chains. In some embodiments, the intracellular signaling domain is not covalently attached to the membrane bound CAR, but is instead diffused in the cytoplasm.
ITAM
[00162] Intracellular signaling domains suitable for use in a CAR of the present disclosure include immunoreceptor tyrosine-based activation motif (ITAM)-containing intracellular signaling polypeptides. An ITAM motif is YXiX2L/I, where and X2 are independently any amino acid (SEQ ID NO: 130). In some cases, the intracellular signaling domain of a subject CAR comprises 1, 2, 3, 4, or 5 ITAM motifs. In some cases, an ITAM motif is repeated twice in an intracellular signaling domain, where the first and second instances of the ITAM motif are separated from one another by 6 to 8 amino acids, e.g., (YX1X2L/I)(X3)n(YX1X2L/I), where n is an integer from 6 to 8, and each of the 6-8 X3 can be any amino acid (SEQ ID NO: 131). In some cases, the intracellular signaling domain of a subject CAR comprises 3 IT AM motifs.
[00163] A suitable intracellular signaling domain can be an ΓΓΑΜ motif-containing portion that is derived from a polypeptide that contains an IT AM motif. For example, a suitable intracellular signaling domain can be an IT AM motif-containing domain from any IT AM motif-containing protein. Thus, a suitable intracellular signaling domain need not contain the entire sequence of the entire protein from which it is derived. Examples of suitable IT AM motif-containing polypeptides include, but are not limited to: DAP12; FCER1G (Fc epsilon receptor I gamma chain); CD3D (CD3 delta); CD3E (CD3 epsilon); CD3G (CD3 gamma); CD3Z (CD3 zeta); and CD79A (antigen receptor complex-associated protein alpha chain).
[00164] In some cases, the intracellular signaling domain is derived from DAP12 (also
known as TYROBP; TYRO protein tyrosine kinase binding protein; KARAP; PLOSL; DN AX- activation protein 12; KAR-associated protein; TYRO protein tyrosine kinase- binding protein; killer activating receptor associated protein; killer- activating receptor- associated protein; etc.). For example, a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to any of the following amino acid sequences (4 isoforms):
MGGLEPCSRLLLLPLLLAVSGLRPVQAQAQSDCSCSTVSPGVLAGIVMGDLVLTVLI ALAVYFLGRLVPRGRGAAEAATRKORITETESPYOELOGORSDVYSDLNTQRPYY
K (SEQ ID NO:98);
MGGLEPCSRLLLLPLLLAVSGLRPVQAQAQSDCSCSTVSPGVLAGIVMGDLVLTVLI ALAVYFLGRLVPRGRGAAEATRKORITETESPYOELOGORSDVYSDLNTQRPYYK
(SEQ ID NO:99);
MGGLEPCSRLLLLPLLLAVSDCSCSTVSPGVLAGIVMGDLVLTVLIALAVYFLGRLV PRGRGAAEAATRKORITETESPYOELOGORSDVYSDLNTQRPYYK (SEQ ID
NO: 100); or
MGGLEPCSRLLLLPLLLAVSDCSCSTVSPGVLAGIVMGDLVLTVLIALAVYFLGRLV PRGRGAAEATRKORITETESPYOELOGORSDVYSDLNTQRPYYK (SEQ ID NO: 101), where the IT AM motifs are in bold and are underlined.
[00165] Likewise, a suitable intracellular signaling domain polypeptide can comprise an
IT AM motif-containing portion of the full length DAP12 amino acid sequence. Thus, a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to the following amino acid sequence:
ESPYOELOGORSDVYSDLNTO (SEQ ID NO: 102), where the ITAM motifs are in bold and are underlined.
[00166] In some cases, the intracellular signaling domain is derived from FCER1G (also known as FCRG; Fc epsilon receptor I gamma chain; Fc receptor gamma-chain; fc-epsilon Rl-gamma; fcRgamma; fceRI gamma; high affinity immunoglobulin epsilon receptor subunit gamma; immunoglobulin E receptor, high affinity, gamma chain; etc.). For example, a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100% amino acid sequence identity to the following amino acid sequence:
MIPAVVLLLLLLVEQAAALGEPQLCYILDAILFLYGIVLTLLYCRLKIQVRKAAITSY EKSDGVYTGLSTRNQETYETLKHEKPPQ (SEQ ID NO: 103), where the ITAM motifs are in bold and are underlined.
[00167] Likewise, a suitable intracellular signaling domain polypeptide can comprise an
ITAM motif-containing portion of the full length FCER1G amino acid sequence. Thus, a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to the following amino acid sequence:
DGVYTGLSTRNOETYETLKHE (SEQ ID NO: 104), where the ITAM motifs are in bold and are underlined.
[00168] In some cases, the intracellular signaling domain is derived from T-cell surface glycoprotein CD3 delta chain (also known as CD3D; CD3-DELTA; T3D; CD3 antigen, delta subunit; CD3 delta; CD3d antigen, delta polypeptide (TiT3 complex); OKT3, delta chain; T-cell receptor T3 delta chain; T-cell surface glycoprotein CD3 delta chain; etc.). For example, a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, or from about 150 aa to about 170 aa, of either of the following amino acid sequences (2 isoforms):
MEHSTFLSGLVLATLLSQVSPFKIPIEELEDRVFVNCNTSITWVEGTVGTLLSDITRL DLGKRILDPRGIYRCNGTDIYKDKESTVQVHYRMCQSCVELDPATVAGIIVTDVIAT LLLALGVFCFAGHETGRLSGAADTOALLRNDOVYOPLRDRDDAOYSHLGGNWAR NK (SEQ ID NO: 105) or
MEHSTFLSGLVLATLLSQVSPFKIPIEELEDRVFVNCNTSITWVEGTVGTLLSDITRL DLGKRILDPRGIYRCNGTDIYKDKESTVOVHYRTADTOALLRNDOVYOPLRDRDD AQYSHLGGNWARNK (SEQ ID NO: 106) , where the ITAM motifs are in bold and are underlined.
[00169] Likewise, a suitable intracellular signaling domain polypeptide can comprise an
ITAM motif-containing portion of the full length CD3 delta amino acid sequence. Thus, a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to the following amino acid sequence:
DOVYOPLRDRDDAOYSHLGGN (SEQ ID NO: 107), where the ITAM motifs are in bold and are underlined.
[00170] In some cases, the intracellular signaling domain is derived from T-cell surface
glycoprotein CD3 epsilon chain (also known as CD3e, T-cell surface antigen T3/Leu-4 epsilon chain, T-cell surface glycoprotein CD3 epsilon chain, AI504783, CD3, CD3epsilon, T3e, etc.). For example, a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, or from about 150 aa to about 205 aa, of the following amino acid sequence:
MQSGTHWRVLGLCLLSVGVWGQDGNEEMGGITQTPYKVSISGTTVILTCPQYPGSE ILWQHNDKNIGGDEDDKNIGSDEDHLSLKEFSELEQSGYYVCYPRGSKPEDANFYL YLRARVCENCMEMDVMSVATIVIVDICITGGLLLLVYYWSKNRKAKAKPVTRGAG AGGRQRGQNKERPPPVPNPDYEPIRKGQRDLYSGLNQRRI (SEQ ID NO: 108), where the ITAM motifs are in bold and are underlined. [00171] Likewise, a suitable intracellular signaling domain polypeptide can comprise an
IT AM motif-containing portion of the full length CD3 epsilon amino acid sequence. Thus, a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to the following amino acid sequence:
NPDYEPIRKGQRDLYSGLNQR (SEQ ID NO: 109), where the ITAM motifs are in bold and are underlined.
[00172] In some cases, the intracellular signaling domain is derived from T-cell surface
glycoprotein CD3 gamma chain (also known as CD3G, T-cell receptor T3 gamma chain, CD3-GAMMA, T3G, gamma polypeptide (TiT3 complex), etc.). For example, a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, or from about 150 aa to about 180 aa, of the following amino acid sequence:
MEQGKGLAVLILAIILLQGTLAQSIKGNHLVKVYDYQEDGSVLLTCDAEAKNITWF KDGKMIGFLTEDKKKWNLGSNAKDPRGMYQCKGSQNKSKPLQVYYRMCQNCIEL NAATISGFLFAEIVSIFVLAVGVYFIAGODGVROSRASDKOTLLPNDOLYOPLKDRE DDQYSHLQGNQLRRN (SEQ ID NO: 110), where the ITAM motifs are in bold and are underlined.
[00173] Likewise, a suitable intracellular signaling domain polypeptide can comprise an
ITAM motif-containing portion of the full length CD3 gamma amino acid sequence. Thus, a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to the following amino acid sequence:
DOLYOPLKDREDDOYSHLOGN (SEQ ID NO: 111), where the ITAM motifs are in bold and are underlined.
[00174] In some cases, the intracellular signaling domain is derived from T-cell surface
glycoprotein CD3 zeta chain (also known as CD3Z, T-cell receptor T3 zeta chain, CD247, CD3-ZETA, CD3H, CD3Q, T3Z, TCRZ, etc.). For example, a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 140 aa, from about 140 aa to about 150 aa, or from about 150 aa to about 160 aa, of either of the following amino acid sequences (2 isoforms):
MKWKALFTAAILQAQLPITEAQSFGLLDPKLCYLLDGILFIYGVILTALFLRVKFSRS ADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYN ELOKDKMAEAYSEIGMKGERRRGKGHDGLYOGLSTATKDTYDALHMQALPPR
(SEQ ID NO: 112) or
MKWKALFTAAILQAQLPITEAQSFGLLDPKLCYLLDGILFIYGVILTALFLRVKFSRS ADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPQRRKNPQEGLY NELOKDKMAEAYSEIGMKGERRRGKGHDGLYOGLSTATKDTYDALHMQALPPR( SEQ ID NO: 113) , where the ITAM motifs are in bold and are underlined.
[00175] Likewise, a suitable intracellular signaling domain polypeptide can comprise an
ITAM motif-containing portion of the full length CD3 zeta amino acid sequence. Thus, a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to any of the following amino acid sequences:
RVKFSRSADAPAYOQGONOLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNP OEGLYNELOKDKMAEAYSEIGMKGERRRGKGHDGLYOGLSTATKDTYDALHMQ ALPPR (SEQ ID NO: 18);
NOLYNELNLGRREEYDVLDKR (SEQ ID NO: 114);
EGLYNELQKDKMAEAYSEIGMK (SEQ ID NO: 115); or
DGLYOGLSTATKDTYDALHMO (SEQ ID NO: 116), where the ITAM motifs are in bold and are underlined.
[00176] In some cases, the intracellular signaling domain is derived from CD79A (also
known as B-cell antigen receptor complex-associated protein alpha chain; CD79a antigen (immunoglobulin-associated alpha); MB-1 membrane glycoprotein; ig-alpha; membrane- bound immunoglobulin-associated protein; surface IgM-associated protein; etc.). For example, a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to a contiguous stretch of from about 100 amino acids to about 110 amino acids (aa), from about 110 aa to about 115 aa, from about 115 aa to about 120 aa, from about 120 aa to about 130 aa, from about 130 aa to about 150 aa, from about 150 aa to about 200 aa, or from about 200 aa to about 220 aa, of either of the following amino acid sequences (2 isoforms):
[00177] MPGGPGVLQALPATIFLLFLLSAVYLGPGCQALWMHKVPASLMVSLGEDA HFQCPHNSSNNANVTWWRVLHGNYTWPPEFLGPGEDPNGTLIIQNVNKSHGGIYV CRVQEGNESYQQSCGTYLRVRQPPPRPFLDMGEGTKNRIITAEGIILLFCAVVPGTLL LFRKRWONEKLGLDAGDEYEDENLYEGLNLDDCSMYEDISRGLOGTYQDVGSLN IGDVQLEKP (SEQ ID NO: 117); or
[00178] MPGGPGVLQALPATIFLLFLLSAVYLGPGCQALWMHKVPASLMVSLGEDA HFQCPHNSSNNANVTWWRVLHGNYTWPPEFLGPGEDPNEPPPRPFLDMGEGTKNR IITAEGIILLFCAVVPGTLLLFRKRWQNEKLGLDAGDEYEDENLYEGLNLDDCSMYE DISRGLQGTYQDVGSLNIGDVQLEKP (SEQ ID NO: 118) , where the ITAM motifs are in bold and are underlined.
[00179] Likewise, a suitable intracellular signaling domain polypeptide can comprise an
ITAM motif-containing portion of the full length CD79A amino acid sequence. Thus, a suitable intracellular signaling domain polypeptide can comprise an amino acid sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, or 100%, amino acid sequence identity to the following amino acid sequence: ENLYEGLNLDDCSMYEDISRG (SEQ ID NO: 119), where the ITAM motifs are in bold and are underlined.
DAP10/CD28
[00180] Intracellular signaling domains suitable for use in a CAR of the present disclosure include a DAP10/CD28 type signaling chain.
[00181] An example of a DAP 10 signaling chain is the amino acid sequence is:
RPRRSPAQDGKVYINMPGRG (SEQ ID NO: 120). In some embodiments, a suitable intracellular signaling domain comprises an amino acid sequence having at least about 85%, at least about 90%, at least about 95%, at least about 98%, or at least about 99%, amino acid sequence identity to the entire length of the amino acid sequence
RPRRSPAQDGKVYINMPGRG (SEQ ID NO: 120).
[00182] An example of a CD28 signaling chain is the amino acid sequence is
FWVLVVVGGVLACYSLLVTVAFIIFWVRSKRSRLLHSDYMNMTPRRPGPTRKHYQ PYAPPRDFAAYRS (SEQ ID NO: 121). In some embodiments, a suitable intracellular signaling domain comprises an amino acid sequence having at least about 85%, at least about 90%, at least about 95%, at least about 98%, or at least about 99%, amino acid sequence identity to the entire length of the amino acid sequence
FWVLVVVGGVLACYSLLVTVAFIIFWVRSKRSRLLHSDYMNMTPRRPGPTRKHYQ
PYAPPRDFAAYRS (SEQ ID NO: 121).
ZAP70
[00183] Intracellular signaling domains suitable for use in a CAR of the present disclosure include a ZAP70 polypeptide, e.g., a polypeptide comprising an amino acid sequence having at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, or 100%, amino acid sequence identity to a contiguous stretch of from about 300 amino acids to about 400 amino acids, from about 400 amino acids to about 500 amino acids, or from about 500 amino acids to 619 amino acids, of the following amino acid sequence:
MPDP A AHLPFF YGSIS RAE AEEHLKLAGM ADGLFLLRQCLRS LGG Y VLS LVHD VRF
HHFPIERQLNGTYAIAGGKAHCGPAELCEFYSRDPDGLPCNLRKPCNRPSGLEPQPG
VFDCLRDAMVRDYVRQTWKLEGEALEQAIISQAPQVEKLIATTAHERMPWYHSSL
TREEAERKLYSGAQTDGKFLLRPRKEQGTYALSLIYGKTVYHYLISQDKAGKYCIP
EGTKFDTLWQLVEYLKLKADGLIYCLKEACPNSSASNASGAAAPTLPAHPSTLTHP
QRRIDTLNSDGYTPEPARITSPDKPRPMPMDTSVYESPYSDPEELKDKKLFLKRDNL
LIADIELGCGNFGSVRQGVYRMRKKQIDVAIKVLKQGTEKADTEEMMREAQIMHQ
LDNPYIVRLIGVCQAEALMLVMEMAGGGPLHKFLVGKREEIPVSNVAELLHQVSM
GMKYLEEKNFVHRDLAARNVLLVNRHYAKISDFGLSKALGADDSYYTARSAGKW
PLKWYAPECINFRKFSSRSDVWSYGVTMWEALSYGQKPYKKMKGPEVMAFIEQG
KRMECPPECPPELYALMSDCWIYKWEDRPDFLTVEQRMRACYYSLASKVEGPPGS
TQKAEAACA (SEQ ID NO:36).
Additional sequences
[00184] The first and/or the second polypeptide of a subject CAR can further include one or more additional polypeptide domains, where such domains include, but are not limited to, a signal sequence; an epitope tag; an affinity domain; and a polypeptide that produces a detectable signal.
Signal sequences
[00185] Signal sequences that are suitable for use in a subject CAR, e.g., in the first
polypeptide of a subject CAR, include any eukaryotic signal sequence, including a naturally-occurring signal sequence, a synthetic (e.g., man-made) signal sequence, etc. Epitope tag
[00186] Suitable epitope tags include, but are not limited to, hemagglutinin (HA; e.g.,
YPYDVPDYA (SEQ ID NO: 122); FLAG (e.g., DYKDDDDK (SEQ ID NO: 123); c-myc (e.g., EQKLISEEDL; SEQ ID NO:4), and the like.
Affinity domain
[00187] Affinity domains include peptide sequences that can interact with a binding partner, e.g., such as one immobilized on a solid support, useful for identification or purification. DNA sequences encoding multiple consecutive single amino acids, such as histidine, when fused to the expressed protein, may be used for one-step purification of the recombinant protein by high affinity binding to a resin column, such as nickel sepharose. Exemplary affinity domains include His5 (HHHHH) (SEQ ID NO: 124), HisX6 (HHHHHH) (SEQ ID NO: 125), C-myc (EQKLISEEDL) (SEQ ID NO:4), Flag (DYKDDDDK) (SEQ ID
NO: 123), StrepTag (WSHPQFEK) (SEQ ID NO: 126), hemagluttinin, e.g., HA Tag
(YPYDVPDYA) (SEQ ID NO: 122), GST, thioredoxin, cellulose binding domain, RYIRS (SEQ ID NO: 127), Phe-His-His-Thr (SEQ ID NO: 128), chitin binding domain, S-peptide, T7 peptide, SH2 domain, C-end RNA tag, WEAAAREACCRECCARA (SEQ ID NO: 129), metal binding domains, e.g., zinc binding domains or calcium binding domains such as those from calcium-binding proteins, e.g., calmodulin, troponin C, calcineurin B, myosin light chain, recoverin, S-modulin, visinin, VILIP, neurocalcin, hippocalcin, frequenin, caltractin, calpain large- subunit, S100 proteins, parvalbumin, calbindin D9K, calbindin D28K, and calretinin, inteins, biotin, streptavidin, MyoD, Id, leucine zipper sequences, and maltose binding protein.
Detectable signal-producing polypeptides
[00188] Suitable detectable signal-producing proteins include, e.g., fluorescent proteins; enzymes that catalyze a reaction that generates a detectable signal as a product; and the like.
[00189] Suitable fluorescent proteins include, but are not limited to, green fluorescent protein
(GFP) or variants thereof, blue fluorescent variant of GFP (BFP), cyan fluorescent variant of GFP (CFP), yellow fluorescent variant of GFP (YFP), enhanced GFP (EGFP), enhanced CFP (ECFP), enhanced YFP (EYFP), GFPS65T, Emerald, Topaz (TYFP), Venus, Citrine, mCitrine, GFPuv, destabilised EGFP (dEGFP), destabilised ECFP (dECFP), destabilised EYFP (dEYFP), mCFPm, Cerulean, T-Sapphire, CyPet, YPet, mKO, HcRed, t-HcRed, DsRed, DsRed2, DsRed-monomer, J-Red, dimer2, t-dimer2(12), mRFPl, pocilloporin, Renilla GFP, Monster GFP, paGFP, Kaede protein and kindling protein, Phycobiliproteins and Phycobiliprotein conjugates including B-Phycoerythrin, R-Phycoerythrin and Allophycocyanin. Other examples of fluorescent proteins include mHoneydew, mBanana, mOrange, dTomato, tdTomato, mTangerine, mStrawberry, mCherry, mGrapel,
mRaspberry, mGrape2, mPlum (Shaner et al. (2005) Nat. Methods 2:905-909), and the like. Any of a variety of fluorescent and colored proteins from Anthozoan species, as described in, e.g., Matz et al. (1999) Nature Biotechnol. 17:969-973, is suitable for use.
[00190] Suitable enzymes include, but are not limited to, horse radish peroxidase (HRP), alkaline phosphatase (AP), beta-galactosidase (GAL), glucose-6-phosphate dehydrogenase, beta-N-acetylglucosaminidase, β-glucuronidase, invertase, Xanthine Oxidase, firefly luciferase, glucose oxidase (GO), and the like.
Recombination of sequences
[00191] In certain instances, sequences of the polypeptides of a CAR, e.g., CAR domains, may be rearranged or deleted in a cell through the use of site- specific recombination technology. In certain embodiments, the cellular activation-related response to a particular CAR can be changed by site-specific recombination, e.g., a first intracellular signaling domain of a CAR eliciting a first activation-related response may be exchanged for a second intracellular signaling domain eliciting a second activation-related response. In certain instances, the response to a particular dimerizer of a CAR can be changed by site- specific recombination, e.g., a first dimerizer-binding pair causing the dimerization of a CAR in the presence of a first dimerizer may be exchanged for a second dimerizer-binding pair causing the dimerization of the CAR in the presence of a second dimerizer. As will be clear to one skilled in the art, site-specific recombination can be used in a cell to exchange any domain or sequence of a CAR with any other domain or sequence as disclosed herein. As will also be clear to one skilled in the art, site-specific recombination can be used in a cell to delete any domain or sequence of a CAR. Such exchange and excision of sequences and domains is known in the art, see, e.g., domain switching in signalobodies as described in Tone et al. (2013) Biotechnology and Bioengineering, 3219-3226, the disclosure of which is disclosed herein by reference. Mechanisms and requirements for performing site-specific
recombination in vivo are also well known in the art, see, e.g., Grindley et al. (2006) Annual Review of Biochemistry, 567-605 and Tropp (2012) Molecular Biology (Jones & Bartlett Publishers, Sudbury, MA), the disclosures of which are incorporated herein by reference. NUCLEIC ACIDS
[00192] The present disclosure provides a nucleic acid that comprises a nucleotide sequence encoding the first and/or the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure. A nucleic acid comprising a nucleotide sequence encoding the first and/or the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure will in some embodiments be DNA, including, e.g., a recombinant expression vector. A nucleic acid comprising a nucleotide sequence encoding the first and/or the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure will in some embodiments be RNA, e.g., in vitro synthesized RNA.
[00193] In some cases, a nucleic acid of the present disclosure comprises a nucleotide
sequence encoding only the first polypeptide (and not the second polypeptide) of a heterodimeric, conditionally active CAR of the present disclosure. In some cases, a nucleic acid of the present disclosure comprises a nucleotide sequence encoding only the second polypeptide (and not the first polypeptide) of a heterodimeric, conditionally active CAR of the present disclosure. In some cases, a nucleic acid of the present disclosure comprises a nucleotide sequence encoding both the first polypeptide and the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure.
[00194] In some cases, a subject nucleic acid provides for production of a CAR of the
present disclosure, e.g., in a mammalian cell. In other cases, a subject nucleic acid provides for amplification of the CAR-encoding nucleic acid.
[00195] A nucleotide sequence encoding the first and/or the second polypeptide of a CAR of the present disclosure can be operably linked to a transcriptional control element, e.g., a promoter, and enhancer, etc.
[00196] Suitable promoter and enhancer elements are known in the art. For expression in a bacterial cell, suitable promoters include, but are not limited to, lacl, lacZ, T3, T7, gpt, lambda P and trc. For expression in a eukaryotic cell, suitable promoters include, but are not limited to, light and/or heavy chain immunoglobulin gene promoter and enhancer elements; cytomegalovirus immediate early promoter; herpes simplex virus thymidine kinase promoter; early and late SV40 promoters; promoter present in long terminal repeats from a retrovirus; mouse metallothionein-I promoter; and various art-known tissue specific promoters.
[00197] Suitable reversible promoters, including reversible inducible promoters are known in the art. Such reversible promoters may be isolated and derived from many organisms, e.g., eukaryotes and prokaryotes. Modification of reversible promoters derived from a first organism for use in a second organism, e.g., a first prokaryote and a second a eukaryote, a first eukaryote and a second a prokaryote, etc., is well known in the art. Such reversible promoters, and systems based on such reversible promoters but also comprising additional control proteins, include, but are not limited to, alcohol regulated promoters (e.g., alcohol dehydrogenase I (alcA) gene promoter, promoters responsive to alcohol transactivator proteins (AlcR), etc.), tetracycline regulated promoters, (e.g., promoter systems including Tet Activators, TetON, TetOFF, etc.), steroid regulated promoters (e.g., rat glucocorticoid receptor promoter systems, human estrogen receptor promoter systems, retinoid promoter systems, thyroid promoter systems, ecdysone promoter systems, mifepristone promoter systems, etc.), metal regulated promoters (e.g., metallothionein promoter systems, etc.), pathogenesis-related regulated promoters (e.g., salicylic acid regulated promoters, ethylene regulated promoters, benzothiadiazole regulated promoters, etc.), temperature regulated promoters (e.g., heat shock inducible promoters (e.g., HSP-70, HSP-90, soybean heat shock promoter, etc.), light regulated promoters, synthetic inducible promoters, and the like.
[00198] In some instances, the locus or construct or transgene containing the suitable
promoter is irreversibly switched through the induction of an inducible system. Suitable systems for induction of an irreversible switch are well known in the art, e.g., induction of an irreversible switch may make use of a Cre-lox-mediated recombination (see, e.g., Fuhrmann-Benzakein, et al., PNAS (2000) 28:e99, the disclosure of which is incorporated herein by reference). Any suitable combination of recombinase, endonuclease, ligase, recombination sites, etc. known to the art may be used in generating an irreversibly switchable promoter. Methods, mechanisms, and requirements for performing site- specific recombination, described elsewhere herein, find use in generating irreversibly switched promoters and are well known in the art, see, e.g., Grindley et al. (2006) Annual Review of Biochemistry, 567-605 and Tropp (2012) Molecular Biology (Jones & Bartlett Publishers, Sudbury, MA), the disclosures of which are incorporated herein by reference.
[00199] In some cases, the promoter is a CD8 cell-specific promoter, a CD4 cell-specific promoter, a neutrophil- specific promoter, or an NK-specific promoter. For example, a CD4 gene promoter can be used; see, e.g., Salmon et al. (1993) Proc. Natl. Acad. Sci. USA 90:7739; and Marodon et al. (2003) Blood 101:3416. As another example, a CD8 gene promoter can be used. NK cell-specific expression can be achieved by use of an Ncrl (p46) promoter; see, e.g., Eckelhart et al. (2011) Blood 117: 1565.
[00200] In some embodiments, e.g., for expression in a yeast cell, a suitable promoter is a constitutive promoter such as an ADH1 promoter, a PGK1 promoter, an ENO promoter, a PYKl promoter and the like; or a regulatable promoter such as a GALl promoter, a GAL10 promoter, an ADH2 promoter, a PH05 promoter, a CUP1 promoter, a GAL7 promoter, a MET25 promoter, a MET3 promoter, a CYC1 promoter, a HIS 3 promoter, an ADH1 promoter, a PGK promoter, a GAPDH promoter, an ADC1 promoter, a TRP1 promoter, a URA3 promoter, a LEU2 promoter, an ENO promoter, a TP1 promoter, and AOX1 (e.g., for use in Pichia). Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art.
[00201] Suitable promoters for use in prokaryotic host cells include, but are not limited to, a bacteriophage T7 RNA polymerase promoter; a trp promoter; a lac operon promoter; a hybrid promoter, e.g., a lac/tac hybrid promoter, a tac/trc hybrid promoter, a trp/lac promoter, a T7/lac promoter; a trc promoter; a tac promoter, and the like; an araBAD promoter; in vivo regulated promoters, such as an ssaG promoter or a related promoter {see, e.g., U.S. Patent Publication No. 20040131637), a pagC promoter (Pulkkinen and Miller, J. Bacteriol., 1991: 173(1): 86-93; Alpuche-Aranda et al., PNAS, 1992; 89(21): 10079-83), a nirB promoter (Harborne et al. (1992) Mol. Micro. 6:2805-2813), and the like {see, e.g., Dunstan et al. (1999) Infect. Immun. 67:5133-5141; McKelvie et al. (2004) Vaccine
22:3243-3255; and Chatfield et al. (1992) Biotechnol. 10:888-892); a sigma70 promoter, e.g., a consensus sigma70 promoter (see, e.g., GenBank Accession Nos. AX798980, AX798961, and AX798183); a stationary phase promoter, e.g., a dps promoter, an spv promoter, and the like; a promoter derived from the pathogenicity island SPI-2 {see, e.g., W096/17951); an actA promoter {see, e.g., Shetron-Rama et al. (2002) Infect. Immun. 70: 1087-1096); an rpsM promoter {see, e.g., Valdivia and Falkow (1996). Mol. Microbiol. 22:367); a tet promoter {see, e.g., Hillen,W. and Wissmann,A. (1989) In Saenger,W. and Heinemann,U. (eds), Topics in Molecular and Structural Biology, Protein-Nucleic Acid Interaction. Macmillan, London, UK, Vol. 10, pp. 143-162); an SP6 promoter {see, e.g., Melton et al. (1984) Nucl. Acids Res. 12:7035); and the like. Suitable strong promoters for use in prokaryotes such as Escherichia coli include, but are not limited to Trc, Tac, T5, T7, and PLambda. Non-limiting examples of operators for use in bacterial host cells include a lactose promoter operator (Lacl repressor protein changes conformation when contacted with lactose, thereby preventing the Lacl repressor protein from binding to the operator), a tryptophan promoter operator (when complexed with tryptophan, TrpR repressor protein has a conformation that binds the operator; in the absence of tryptophan, the TrpR repressor protein has a conformation that does not bind to the operator), and a tac promoter operator (see, for example, deBoer et al. (1983) Proc. Natl. Acad. Sci. U.S.A. 80:21-25).
[00202] A nucleotide sequence encoding a subject CAR can be present in an expression vector and/or a cloning vector. Where a subject CAR comprises two separate polypeptides, nucleotide sequences encoding the two polypeptides can be cloned in the same or separate vectors. An expression vector can include a selectable marker, an origin of replication, and other features that provide for replication and/or maintenance of the vector. Suitable expression vectors include, e.g., plasmids, viral vectors, and the like.
[00203] Large numbers of suitable vectors and promoters are known to those of skill in the art; many are commercially available for generating a subject recombinant constructs. The following vectors are provided by way of example. Bacterial: pBs, phagescript, PsiX174, pBluescript SK, pBs KS, pNH8a, pNH16a, pNH18a, pNH46a (Stratagene, La Jolla, Calif., USA); pTrc99A, pKK223-3, pKK233-3, pDR540, and pRIT5 (Pharmacia, Uppsala, Sweden). Eukaryotic: pWLneo, pSV2cat, pOG44, PXR1, pSG (Stratagene) pSVK3, pBPV, pMSG and pSVL (Pharmacia).
[00204] Expression vectors generally have convenient restriction sites located near the
promoter sequence to provide for the insertion of nucleic acid sequences encoding heterologous proteins. A selectable marker operative in the expression host may be present. Suitable expression vectors include, but are not limited to, viral vectors (e.g. viral vectors based on vaccinia virus; poliovirus; adenovirus (see, e.g., Li et al., Invest Opthalmol Vis Sci 35:2543 2549, 1994; Borras et al., Gene Ther 6:515 524, 1999; Li and Davidson, PNAS 92:7700 7704, 1995; Sakamoto et al., H Gene Ther 5: 1088 1097, 1999; WO 94/12649, WO 93/03769; WO 93/19191; WO 94/28938; WO 95/11984 and WO 95/00655); adeno- associated virus (see, e.g., Ali et al., Hum Gene Ther 9:81 86, 1998, Flannery et al., PNAS 94:6916 6921, 1997; Bennett et al., Invest Opthalmol Vis Sci 38:2857 2863, 1997; Jomary et al., Gene Ther 4:683 690, 1997, Rolling et al., Hum Gene Ther 10:641 648, 1999; Ali et al., Hum Mol Genet 5:591 594, 1996; Srivastava in WO 93/09239, Samulski et al., J. Vir. (1989) 63:3822-3828; Mendelson et al., Virol. (1988) 166: 154-165; and Flotte et al., PNAS (1993) 90: 10613-10617); SV40; herpes simplex virus; human immunodeficiency virus (see, e.g., Miyoshi et al., PNAS 94: 10319 23, 1997; Takahashi et al., J Virol 73:7812 7816, 1999); a retroviral vector (e.g., Murine Leukemia Virus, spleen necrosis virus, and vectors derived from retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, human immunodeficiency virus, myeloproliferative sarcoma virus, and mammary tumor virus); and the like.
[00205] As noted above, in some embodiments, a nucleic acid comprising a nucleotide
sequence encoding the first and/or the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure will in some embodiments be RNA, e.g., in vitro synthesized RNA. Methods for in vitro synthesis of RNA are known in the art; any known method can be used to synthesize RNA comprising a nucleotide sequence encoding the first and/or the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure. Methods for introducing RNA into a host cell are known in the art. See, e.g., Zhao et al. (2010) Cancer Res. 15:9053. Introducing RNA comprising a nucleotide sequence encoding the first and/or the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure into a host cell can be carried out in vitro or ex vivo or in vivo. For example, a host cell (e.g., an NK cell, a cytotoxic T lymphocyte, etc.) can be electroporated in vitro or ex vivo with RNA comprising a nucleotide sequence encoding the first and/or the second polypeptide of a heterodimeric, conditionally active CAR of the present disclosure.
CELLS
[00206] The present disclosure provides a mammalian cell that is genetically modified to produce a heterodimeric, conditionally active CAR of the present disclosure.
[00207] Suitable mammalian cells include primary cells and immortalized cell lines.
Suitable mammalian cell lines include human cell lines, non-human primate cell lines, rodent (e.g., mouse, rat) cell lines, and the like. Suitable mammalian cell lines include, but are not limited to, HeLa cells (e.g., American Type Culture Collection (ATCC) No. CCL-2), CHO cells (e.g., ATCC Nos. CRL9618, CCL61, CRL9096), 293 cells (e.g., ATCC No. CRL-1573), Vero cells, NIH 3T3 cells (e.g., ATCC No. CRL-1658), Huh-7 cells, BHK cells (e.g., ATCC No. CCL10), PC12 cells (ATCC No. CRL1721), COS cells, COS-7 cells (ATCC No. CRL1651), RATI cells, mouse L cells (ATCC No. CCLI.3), human embryonic kidney (HEK) cells (ATCC No. CRL1573), HLHepG2 cells, Hut-78, Jurkat, HL-60, NK cell lines (e.g., NKL, NK92, and YTS), and the like.
[00208] In some instances, the cell is not an immortalized cell line, but is instead a cell (e.g., a primary cell) obtained from an individual. For example, in some cases, the cell is an immune cell obtained from an individual. As an example, the cell is a T lymphocyte obtained from an individual. As another example, the cell is a cytotoxic cell obtained from an individual. As another example, the cell is a stem cell or progenitor cell obtained from an individual.
METHODS OF ACTIVATING AN IMMUNE CELL
[00209] The present disclosure provides methods of activating an immune cell in vitro, in vivo, or ex vivo. The methods generally involve contacting an immune cell (in vitro, in vivo, or ex vivo) with a dimerizing agent and an antigen, where the immune cell is genetically modified to produce a heterodimeric, conditionally active CAR of the present disclosure. In the presence of the dimerizing agent and the antigen, the heterodimeric, conditionally active CAR dimerizes and activates the immune cell, thereby producing an activated immune cell. Immune cells include, e.g., a cytotoxic T lymphocyte, an NK cell, a CD4+ T cell, a T regulatory (Treg) cell, etc.
[00210] Contacting the genetically modified immune cell (e.g., a T lymphocyte, an NK cell) with a dimerizing agent and a second member of a specific binding pair (e.g., an antigen, a ligand, a receptor) can increase production of a cytokine by the immune cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared with the amount of cytokine produced by the immune cell in the absence of the second member of a specific binding pair and/or the dimerizing agent. Cytokines whose production can be increased include, but are not limited to, IL-2 and IFN-γ.
[00211] Contacting the genetically modified immune cell (e.g., a T lymphocyte, an NK cell) with a dimerizing agent and an antigen can increase production of a cytokine by the immune cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2- fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10- fold, compared with the amount of cytokine produced by the immune cell in the absence of the antigen and/or the dimerizing agent. Cytokines whose production can be increased include, but are not limited to, IL-2 and IFN-γ.
[00212] Contacting a genetically modified cytotoxic cell (e.g., cytotoxic T lymphocyte) with a dimerizing agent and a second member of a specific binding pair (e.g., an antigen, a ligand, a receptor) can increase cytotoxic activity of the cytotoxic cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared to the cytotoxic activity of the cytotoxic cell in the absence of the dimerizing agent.
[00213] Contacting a genetically modified cytotoxic cell (e.g., cytotoxic T lymphocyte) with a dimerizing agent and an antigen can increase cytotoxic activity of the cytotoxic cell by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared to the cytotoxic activity of the cytotoxic cell in the absence of the dimerizing agent. [00214] In other embodiments, e.g., depending on the host immune cell, contacting a genetically modified host cell with a dimerizing agent and an antigen can increase or decrease cell proliferation, cell survival, cell death, and the like.
METHODS OF GENERATING A CONDITIONALLY ACTIVATABLE CELL
[00215] The present disclosure provides a method of generating a conditionally activatable cell. The method generally involves genetically modifying a mammalian cell with an expression vector, or an RNA (e.g., in vitro transcribed RNA), comprising nucleotide sequences encoding a heterodimeric, conditionally active CAR of the present disclosure. The genetically modified cell is conditionally activatable in the presence of: a) an antigen to which the first polypeptide of the CAR binds; and b) a dimerizer (a dimerizing agent). The genetic modification can be carried out in vivo, in vitro, or ex vivo. The cell can be an immune cell (e.g., a T lymphocyte or NK cell), a stem cell, a progenitor cell, etc.
[00216] In some cases, the genetic modification is carried out ex vivo. For example, a T
lymphocyte, a stem cell, or an NK cell is obtained from an individual; and the cell obtained from the individual is genetically modified to express a CAR of the present disclosure. The genetically modified cell is conditionally activatable in the presence of: a) an antigen to which the first polypeptide of the CAR binds; and b) a dimerizer. In some cases, the genetically modified cell is activated ex vivo. In other cases, the genetically modified cell is introduced into an individual (e.g., the individual from whom the cell was obtained); and the genetically modified cell is activated in vivo, e.g., by administering to the individual a dimerizer. For example, where the antigen is present on the surface of a cell in the individual, there is no need to administer the antigen. The genetically modified cell comes into contact with the antigen present on the surface of a cell in the individual; and, upon administration to the individual of a dimerizer, the genetically modified cell is activated. For example, where the genetically modified cell is a T lymphocyte, the genetically modified cell can exhibit cytotoxicity toward a cell that presents an antigen on its surface to which the CAR binds.
TREATMENT METHODS
[00217] The present disclosure provides various treatment methods using a subject CAR.
Cytotoxicity methods
[00218] A CAR of the present disclosure, when present in a T lymphocyte or an NK cell, can mediate cytotoxicity toward a target cell. A CAR of the present disclosure binds to an antigen present on a target cell, thereby mediating killing of a target cell by a T lymphocyte or an NK cell genetically modified to produce the CAR. The antigen-binding domain of the CAR binds to an antigen present on the surface of a target cell.
[00219] Target cells include, but are not limited to, cancer cells. Thus, the present disclosure provides methods of killing, or inhibiting the growth of, a target cancer cell, the method involving contacting a cytotoxic immune effector cell (e.g., a cytotoxic T cell, or an NK cell) that is genetically modified to produce a subject CAR, such that the T lymphocyte or NK cell recognizes an antigen present on the surface of a target cancer cell, and mediates killing of the target cell.
[00220] The present disclosure provides a method of treating cancer in an individual having a cancer, the method comprising: i) genetically modifying T lymphocytes obtained from the individual with an expression vector comprising nucleotide sequences encoding the heterodimeric, conditionally active CAR of the present disclosure, where the antigen- binding domain of the heterodimeric, conditionally active CAR is specific for an epitope on a cancer cell in the individual, and where the genetic modification is carried out ex vivo; ii) introducing the genetically modified T lymphocytes into the individual; and iii)
administering to the individual an effective amount of a dimerizing agent, wherein the dimerizing agent induces dimerization of the heterodimeric, conditionally active CAR, wherein said dimerization provides for activation of the genetically modified T lymphocytes and killing of the cancer cell, thereby treating the cancer.
[00221] Carcinomas that can be amenable to therapy by a method disclosed herein include, but are not limited to, esophageal carcinoma, hepatocellular carcinoma, basal cell carcinoma (a form of skin cancer), squamous cell carcinoma (various tissues), bladder carcinoma, including transitional cell carcinoma (a malignant neoplasm of the bladder), bronchogenic carcinoma, colon carcinoma, colorectal carcinoma, gastric carcinoma, lung carcinoma, including small cell carcinoma and non-small cell carcinoma of the lung, adrenocortical carcinoma, thyroid carcinoma, pancreatic carcinoma, breast carcinoma, ovarian carcinoma, prostate carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, renal cell carcinoma, ductal carcinoma in situ or bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical carcinoma, uterine carcinoma, testicular carcinoma, osteogenic carcinoma, epithelial carcinoma, and nasopharyngeal carcinoma.
[00222] Sarcomas that can be amenable to therapy by a method disclosed herein include, but are not limited to, fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, chordoma, osteogenic sarcoma, osteosarcoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's sarcoma,
leiomyosarcoma, rhabdomyosarcoma, and other soft tissue sarcomas.
[00223] Other solid tumors that can be amenable to therapy by a method disclosed herein include, but are not limited to, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblastoma, and retinoblastoma.
[00224] Leukemias that can be amenable to therapy by a method disclosed herein include, but are not limited to, a) chronic myeloproliferative syndromes (neoplastic disorders of multipotential hematopoietic stem cells); b) acute myelogenous leukemias (neoplastic transformation of a multipotential hematopoietic stem cell or a hematopoietic cell of restricted lineage potential; c) chronic lymphocytic leukemias (CLL; clonal proliferation of immunologically immature and functionally incompetent small lymphocytes), including B- cell CLL, T-cell CLL prolymphocytic leukemia, and hairy cell leukemia; and d) acute lymphoblastic leukemias (characterized by accumulation of lymphoblasts). Lymphomas that can be treated using a subject method include, but are not limited to, B-cell lymphomas (e.g., Burkitt's lymphoma); Hodgkin's lymphoma; non-Hodgkin's lymphoma, and the like.
[00225] Other cancers that can be amenable to treatment according to the methods disclosed herein include atypical meningioma (brain), islet cell carcinoma (pancreas), medullary carcinoma (thyroid), mesenchymoma (intestine), hepatocellular carcinoma (liver), hepatoblastoma (liver), clear cell carcinoma (kidney), and neurofibroma mediastinum.
Immunomodulatory methods
[00226] A subject method can also be used to treat inflammatory conditions and autoimmune disease. A subject CAR is expressed in a T-helper cell or a Tregs for use in an
immunomodulatory method. Immunomodulatory methods include, e.g., enhancing an immune response in a mammalian subject toward a pathogen; enhancing an immune response in a subject who is immunocompromised; reducing an inflammatory response; reducing an immune response in a mammalian subject to an autoantigen, e.g., to treat an autoimmune disease; and reducing an immune response in a mammalian subject to a transplanted organ or tissue, to reduce organ or tissue rejection.
[00227] Where the method involves reducing an immune response to an autoantigen, the antigen used to activate the CAR is an autoantigen. Where the method involves reducing an immune response to a transplanted organ or tissue, the antigen used to activate the CAR is an antigen specific to the transplanted organ. Formulations, dosages, and routes of administration
[00228] As discussed above, a treatment method of the present disclosure involves
administration to an individual in need thereof of an effective amount of a dimerizer agent, and may also involve administration of an antigen.
[00229] An "effective amount" of a dimerizer agent is in some cases an amount that, when administered in one or more doses to an individual in need thereof, increases the level of cytotoxic activity of a T lymphocyte expressing a subject CAR by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared to the cytotoxic activity of the T lymphocyte in the absence of the dimerizing agent.
[00230] An "effective amount" of a dimerizer agent is in some cases an amount that, when administered in one or more doses to an individual in need thereof, increases the level of cytotoxic activity of an NK cell expressing a subject CAR by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, at least about 10-fold, or more than 10-fold, compared to the cytotoxic activity of the NK cell in the absence of the dimerizing agent.
[00231] An "effective amount" of a dimerizer agent is in some cases an amount that, when administered in one or more doses to an individual in need thereof, reduces the number of cancer cells in the individual and/or reduces tumor mass in the individual, by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 75%, or more than 75%, compared to the number of cancer cells and/or tumor mass in the absence of the dimerizing agent.
[00232] In some embodiments, an effective amount of a dimerizer is an amount that, when administered alone (e.g., in monotherapy) or in combination (e.g., in combination therapy) with one or more additional therapeutic agents, in one or more doses, is effective to reduce one or more of tumor growth rate, cancer cell number, and tumor mass, by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more, compared to the tumor growth rate, cancer cell number, or tumor mass in the absence of treatment with the dimerizer. Formulations
[00233] In the subject methods, a dimerizer can be administered to the host using any
convenient means capable of resulting in the desired therapeutic effect or diagnostic effect. Thus, the dimerizer can be incorporated into a variety of formulations for therapeutic administration. More particularly, a dimerizer can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants and aerosols.
[00234] In pharmaceutical dosage forms, a dimerizer can be administered in the form of their pharmaceutically acceptable salts, or they may also be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds. The following methods and excipients are merely exemplary and are in no way limiting.
[00235] Suitable excipient vehicles are, for example, water, saline, dextrose, glycerol,
ethanol, or the like, and combinations thereof. In addition, if desired, the vehicle may contain minor amounts of auxiliary substances such as wetting or emulsifying agents or pH buffering agents. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art. See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pennsylvania, 17th edition, 1985. The composition or formulation to be administered will, in any event, contain a quantity of a dimerizer adequate to achieve the desired state in the subject being treated.
[00236] The pharmaceutically acceptable excipients, such as vehicles, adjuvants, carriers or diluents, are readily available to the public. Moreover, pharmaceutically acceptable auxiliary substances, such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are readily available to the public.
[00237] For oral preparations, a dimerizer can be used alone or in combination with
appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
[00238] A dimerizer can be formulated into preparations for injection by dissolving,
suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
[00239] Pharmaceutical compositions comprising a dimerizer are prepared by mixing the dimerizer having the desired degree of purity with optional physiologically acceptable carriers, excipients, stabilizers, surfactants, buffers and/or tonicity agents. Acceptable carriers, excipients and/or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid, glutathione, cysteine, methionine and citric acid; preservatives (such as ethanol, benzyl alcohol, phenol, m-cresol, p-chlor-m-cresol, methyl or propyl parabens, benzalkonium chloride, or combinations thereof); amino acids such as arginine, glycine, ornithine, lysine, histidine, glutamic acid, aspartic acid, isoleucine, leucine, alanine, phenylalanine, tyrosine, tryptophan, methionine, serine, proline and combinations thereof; monosaccharides, disaccharides and other carbohydrates; low molecular weight (less than about 10 residues) polypeptides; proteins, such as gelatin or serum albumin; chelating agents such as EDTA; sugars such as trehalose, sucrose, lactose, glucose, mannose, maltose, galactose, fructose, sorbose, raffinose, glucosamine, N- methylglucosamine, galactosamine, and neuraminic acid; and/or non-ionic surfactants such as Tween, Brij Pluronics, Triton-X, or polyethylene glycol (PEG).
[00240] The pharmaceutical composition may be in a liquid form, a lyophilized form or a liquid form reconstituted from a lyophilized form, wherein the lyophilized preparation is to be reconstituted with a sterile solution prior to administration. The standard procedure for reconstituting a lyophilized composition is to add back a volume of pure water (typically equivalent to the volume removed during lyophilization); however solutions comprising antibacterial agents may be used for the production of pharmaceutical compositions for parenteral administration; see also Chen (1992) Drug Dev Ind Pharm 18, 1311-54.
[00241] 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 a dimerizer calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle. The specifications for a given dimerizer may depend on the particular dimerizer employed and the effect to be achieved, and the pharmacodynamics associated with each dimerizer in the host. [00242] In some embodiments, a dimerizer is formulated in a controlled release formulation.
Sustained-release preparations may be prepared using methods well known in the art.
Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the dimerizer in which the matrices are in the form of shaped articles, e.g. films or microcapsules. Examples of sustained-release matrices include polyesters, copolymers of L-glutamic acid and ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, hydrogels, polylactides, degradable lactic acid-glycolic acid copolymers and poly-D-(-)-3-hydroxybutyric acid. Possible loss of biological activity may be prevented by using appropriate additives, by controlling moisture content and by developing specific polymer matrix compositions.
Dosages
[00243] A suitable dosage can be determined by an attending physician or other qualified medical personnel, based on various clinical factors. As is well known in the medical arts, dosages for any one patient depend upon many factors, including the patient's size, body surface area, age, the particular dimerizer to be administered, sex of the patient, time, and route of administration, general health, and other drugs being administered concurrently. A dimerizer may be administered in amounts between 1 ng/kg body weight and 20 mg/kg body weight per dose, e.g. between 0.1 mg/kg body weight to 10 mg/kg body weight, e.g. between 0.5 mg/kg body weight to 5 mg/kg body weight; however, doses below or above this exemplary range are envisioned, especially considering the aforementioned factors. If the regimen is a continuous infusion, it can also be in the range of 1 μg to 10 mg per kilogram of body weight per minute.
[00244] Those of skill will readily appreciate that dose levels can vary as a function of the specific dimerizer, the severity of the symptoms and the susceptibility of the subject to side effects. Preferred dosages for a given compound are readily determinable by those of skill in the art by a variety of means.
Routes of administration
[00245] A dimerizer is administered to an individual using any available method and route suitable for drug delivery, including in vivo and ex vivo methods, as well as systemic and localized routes of administration.
[00246] Conventional and pharmaceutically acceptable routes of administration include intratumoral, peritumoral, intramuscular, intratracheal, intracranial, subcutaneous, intradermal, topical application, intravenous, intraarterial, rectal, nasal, oral, and other enteral and parenteral routes of administration. Routes of administration may be combined, if desired, or adjusted depending upon the dimerizer and/or the desired effect. A dimerizer can be administered in a single dose or in multiple doses. In some embodiments, a dimerizer is administered orally. In some embodiments, a dimerizer is administered via an inhalational route. In some embodiments, a dimerizer is administered intranasally. In some
embodiments, a dimerizer is administered locally. In some embodiments, a dimerizer is administered intratumorally. In some embodiments, a dimerizer is administered
peritumorally. In some embodiments, a dimerizer is administered intracranially. In some embodiments, a dimerizer is administered intravenously.
[00247] The agent can be administered to a host using any available conventional methods and routes suitable for delivery of conventional drugs, including systemic or localized routes. In general, routes of administration contemplated by the invention include, but are not necessarily limited to, enteral, parenteral, or inhalational routes.
[00248] Parenteral routes of administration other than inhalation administration include, but are not necessarily limited to, topical, transdermal, subcutaneous, intramuscular,
intraorbital, intracapsular, intraspinal, intrasternal, intratumoral, peritumoral, and
intravenous routes, i.e., any route of administration other than through the alimentary canal. Parenteral administration can be carried to effect systemic or local delivery of a dimerizer. Where systemic delivery is desired, administration typically involves invasive or systemically absorbed topical or mucosal administration of pharmaceutical preparations.
[00249] A dimerizer can also be delivered to the subject by enteral administration. Enteral routes of administration include, but are not necessarily limited to, oral and rectal {e.g., using a suppository) delivery.
[00250] By treatment is meant at least an amelioration of the symptoms associated with the pathological condition afflicting the host, where amelioration is used in a broad sense to refer to at least a reduction in the magnitude of a parameter, e.g. symptom, associated with the pathological condition being treated, such as cancer. As such, treatment also includes situations where the pathological condition, or at least symptoms associated therewith, are completely inhibited, e.g. prevented from happening, or stopped, e.g. terminated, such that the host no longer suffers from the pathological condition, or at least the symptoms that characterize the pathological condition.
[00251] In some embodiments, a dimerizer is administered by injection and/or delivery, e.g., to a site in a brain artery or directly into brain tissue. A dimerizer can also be administered directly to a target site e.g., by direct injection, by implantation of a drug delivery device such as an osmotic pump or slow release particle, by biolistic delivery to the target site, etc. Combination therapy
[00252] In some embodiments, a dimerizer is administered as an adjuvant therapy to a
standard cancer therapy. Standard cancer therapies include surgery (e.g., surgical removal of cancerous tissue), radiation therapy, bone marrow transplantation, chemotherapeutic treatment, antibody treatment, biological response modifier treatment, and certain combinations of the foregoing.
[00253] Radiation therapy includes, but is not limited to, x-rays or gamma rays that are
delivered from either an externally applied source such as a beam, or by implantation of small radioactive sources.
[00254] Suitable antibodies for use in cancer treatment include, but are not limited to, naked antibodies, e.g., trastuzumab (Herceptin) , bevacizumab (Avastin™), cetuximab
(Erbitux™), panitumumab (Vectibix™), Ipilimumab (Yervoy™), rituximab (Rituxan), alemtuzumab (Lemtrada™), Ofatumumab (Arzerra™), Oregovomab (OvaRex™),
Lambrolizumab (MK-3475), pertuzumab (Perjeta™), ranibizumab (Lucentis™) etc., and conjugated antibodies, e.g., gemtuzumab ozogamicin (Mylortarg™), Brentuximab vedotin (Adcetris™), 90Y-labelled ibritumomab tiuxetan (Zevalin™), 131I-labelled tositumoma (Bexxar™), etc. Suitable antibodies for use in cancer treatment include, but are not limited to, antibodies raised against tumor- associated antigens. Such antigens include, but are not limited to, CD20, CD30, CD33, CD52, EpCAM, CEA, gpA33, Mucins, TAG-72, CAIX, PSMA, Folate-binding protein, Gangliosides (e.g., GD2, GD3, GM2, etc.), Le y , VEGF, VEGFR, Integrin alpha- V-beta-3, Integrin alpha-5-beta-l, EGFR, ERBB2, ERBB3, MET, IGF1R, EPHA3, TRAILR1, TRAILR2, RANKL, FAP, Tenascin, etc.
[00255] Biological response modifiers suitable for use in connection with the methods of the present disclosure include, but are not limited to, (1) inhibitors of tyrosine kinase (RTK) activity; (2) inhibitors of serine/threonine kinase activity; (3) tumor-associated antigen antagonists, such as antibodies that bind specifically to a tumor antigen; ( 4) apoptosis receptor agonists; (5) interleukin-2; (6) interferon-a.; (7) interferon -γ; (8) colony- stimulating factors; (9) inhibitors of angiogenesis; and (10) antagonists of tumor necrosis factor.
[00256] Chemotherapeutic agents are non-peptidic (i.e., non-proteinaceous) compounds that reduce proliferation of cancer cells, and encompass cytotoxic agents and cytostatic agents. Non-limiting examples of chemotherapeutic agents include alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, plant (vinca) alkaloids, and steroid hormones. [00257] Agents that act to reduce cellular proliferation are known in the art and widely used.
Such agents include alkylating agents, such as nitrogen mustards, nitrosoureas, ethylenimine derivatives, alkyl sulfonates, and triazenes, including, but not limited to, mechlorethamine, cyclophosphamide (Cytoxan™), melphalan (L-sarcolysin), carmustine (BCNU), lomustine (CCNU), semustine (methyl-CCNU), streptozocin, chlorozotocin, uracil mustard, chlormethine, ifosfamide, chlorambucil, pipobroman, triethylenemelamine,
triethylenethiophosphoramine, busulfan, dacarbazine, and temozolomide.
[00258] Antimetabolite agents include folic acid analogs, pyrimidine analogs, purine
analogs, and adenosine deaminase inhibitors, including, but not limited to, cytarabine (CYTOSAR-U), cytosine arabinoside, fluorouracil (5-FU), floxuridine (FudR), 6- thioguanine, 6-mercaptopurine (6-MP), pentostatin, 5 -fluorouracil (5-FU), methotrexate, 10-propargyl-5,8-dideazafolate (PDDF, CB3717), 5,8-dideazatetrahydrofolic acid
(DDATHF), leucovorin, fludarabine phosphate, pentostatine, and gemcitabine.
[00259] Suitable natural products and their derivatives, (e.g., vinca alkaloids, antitumor
antibiotics, enzymes, lymphokines, and epipodophyllotoxins), include, but are not limited to, Ara-C, paclitaxel (Taxol®), docetaxel (Taxotere®), deoxycoformycin, mitomycin-C, L- asparaginase, azathioprine; brequinar; alkaloids, e.g. vincristine, vinblastine, vinorelbine, vindesine, etc.; podophyllotoxins, e.g. etoposide, teniposide, etc.; antibiotics, e.g.
anthracycline, daunorubicin hydrochloride (daunomycin, rubidomycin, cerubidine), idarubicin, doxorubicin, epirubicin and morpholino derivatives, etc.; phenoxizone biscyclopeptides, e.g. dactinomycin; basic glycopeptides, e.g. bleomycin; anthraquinone glycosides, e.g. plicamycin (mithramycin); anthracenediones, e.g. mitoxantrone;
azirinopyrrolo indolediones, e.g. mitomycin; macrocyclic immunosuppressants, e.g.
cyclosporine, FK-506 (tacrolimus, prograf), rapamycin, etc. ; and the like.
[00260] Other anti-proliferative cytotoxic agents are navelbene, CPT-11, anastrazole, letrazole, capecitabine, reloxafine, cyclophosphamide, ifosamide, and droloxafine.
[00261] Microtubule affecting agents that have antiproliferative activity are also suitable for use and include, but are not limited to, allocolchicine (NSC 406042), Halichondrin B (NSC 609395), colchicine (NSC 757), colchicine derivatives (e.g., NSC 33410), dolstatin 10 (NSC 376128), maytansine (NSC 153858), rhizoxin (NSC 332598), paclitaxel (Taxol®), Taxol® derivatives, docetaxel (Taxotere®), thiocolchicine (NSC 361792), trityl cysterin, vinblastine sulfate, vincristine sulfate, natural and synthetic epothilones including but not limited to, eopthilone A, epothilone B, discodermolide; estramustine, nocodazole, and the like. [00262] Hormone modulators and steroids (including synthetic analogs) that are suitable for use include, but are not limited to, adrenocorticosteroids, e.g. prednisone, dexamethasone, etc.; estrogens and pregestins, e.g. hydroxyprogesterone caproate, medroxyprogesterone acetate, megestrol acetate, estradiol, clomiphene, tamoxifen; etc.; and adrenocortical suppressants, e.g. aminoglutethimide; 17a-ethinylestradiol; diethylstilbestrol, testosterone, fluoxymesterone, dromostanolone propionate, testolactone, methylprednisolone, methyl- testosterone, prednisolone, triamcinolone, chlorotrianisene, hydroxyprogesterone, aminoglutethimide, estramustine, medroxyprogesterone acetate, leuprolide, Flutamide (Drogenil), Toremifene (Fareston), and Zoladex®. Estrogens stimulate proliferation and differentiation, therefore compounds that bind to the estrogen receptor are used to block this activity. Corticosteroids may inhibit T cell proliferation.
[00263] Other chemotherapeutic agents include metal complexes, e.g. cisplatin (cis-DDP), carboplatin, etc.; ureas, e.g. hydroxyurea; and hydrazines, e.g. N-methylhydrazine;
epidophyllotoxin; a topoisomerase inhibitor; procarbazine; mitoxantrone; leucovorin;
tegafur; etc.. Other anti-proliferative agents of interest include immunosuppressants, e.g. mycophenolic acid, thalidomide, desoxyspergualin, azasporine, leflunomide, mizoribine, azaspirane (SKF 105685); Iressa® (ZD 1839, 4-(3-chloro-4-fluorophenylamino)-7- methoxy-6- (3- (4-morpholinyl)propoxy)quinazoline) ; etc .
[00264] "Taxanes" include paclitaxel, as well as any active taxane derivative or pro-drug.
"Paclitaxel" (which should be understood herein to include analogues, formulations, and derivatives such as, for example, docetaxel, TAXOL™, TAXOTERE™ (a formulation of docetaxel), 10-desacetyl analogs of paclitaxel and 3'N-desbenzoyl-3'N-t-butoxycarbonyl analogs of paclitaxel) may be readily prepared utilizing techniques known to those skilled in the art (see also WO 94/07882, WO 94/07881, WO 94/07880, WO 94/07876, WO
93/23555, WO 93/10076; U.S. Pat. Nos. 5,294,637; 5,283,253; 5,279,949; 5,274,137;
5,202,448; 5,200,534; 5,229,529; and EP 590,267), or obtained from a variety of commercial sources, including for example, Sigma Chemical Co., St. Louis, Mo. (T7402 from Taxus brevifolia; or T-1912 from Taxus yannanensis).
[00265] Paclitaxel should be understood to refer to not only the common chemically
available form of paclitaxel, but analogs and derivatives (e.g., Taxotere™ docetaxel, as noted above) and paclitaxel conjugates (e.g., paclitaxel-PEG, paclitaxel-dextran, or paclitaxel-xylose).
[00266] Also included within the term "taxane" are a variety of known derivatives, including both hydrophilic derivatives, and hydrophobic derivatives. Taxane derivatives include, but not limited to, galactose and mannose derivatives described in International Patent Application No. WO 99/18113; piperazino and other derivatives described in WO
99/14209; taxane derivatives described in WO 99/09021, WO 98/22451, and U.S. Patent No. 5,869,680; 6-thio derivatives described in WO 98/28288; sulfenamide derivatives described in U.S. Patent No. 5,821,263; and taxol derivative described in U.S. Patent No. 5,415,869. It further includes prodrugs of paclitaxel including, but not limited to, those described in WO 98/58927; WO 98/13059; and U.S. Patent No. 5,824,701.
SUBJECTS SUITABLE FOR TREATMENT
[00267] A variety of subjects are suitable for treatment with a subject method of treating cancer. Suitable subjects include any individual, e.g., a human or non-human animal who has cancer, who has been diagnosed with cancer, who is at risk for developing cancer, who has had cancer and is at risk for recurrence of the cancer, who has been treated with an agent other than a dimerizer for the cancer and failed to respond to such treatment, or who has been treated with an agent other than a dimerizer for the cancer but relapsed after initial response to such treatment.
[00268] Subjects suitable for treatment with a subject immunomodulatory method include individuals who have an autoimmune disorder; individuals who are organ or tissue transplant recipients; and the like; individuals who are immunocompromised; and individuals who are infected with a pathogen.
EXAMPLES
[00269] The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the present invention, and are not intended to limit the scope of what the inventors regard as their invention nor are they intended to represent that the experiments below are all or the only experiments performed. Efforts have been made to ensure accuracy with respect to numbers used (e.g. amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees Celsius, and pressure is at or near atmospheric. Standard abbreviations may be used, e.g., bp, base pair(s); kb, kilobase(s); pi, picoliter(s); s or sec, second(s); min, minute(s); h or hr, hour(s); aa, amino acid(s); kb, kilobase(s); bp, base pair(s); nt, nucleotide(s); i.m., intramuscular(ly); i.p., intraperitoneal (ly); s.c, subcutaneous (ly); i.v., intravenous (ly); and the like. Example 1 : Generation of CAR
MATERIALS AND METHODS
[00270] The anti-human CD 19 scFv was selected as the antigen recognition domain in CARs throughout the design optimization process. Figures 18A and 18B summarize the molecular structure of each CAR consisting of two numerically identified polypeptides. All membrane- anchored polypeptides are di-sulfide bonded homo-dimers. The membrane- anchored polypeptides are depicted as monomers for graphical simplicity.
Generation of CAR constructs
[00271] Sequence encoding the anti-human CD 19 scFv was cloned from a construct. The human 4- IBB co- stimulation and CD3 zeta ΓΓΑΜ signaling chains were cloned from cDNAs supplied by Open Biosystems. FKBP- and FRB-encoding sequences were cloned from plasmids supplied by Addgene.
[00272] Standard molecular cloning techniques (polymerase chain reaction (PCR), restriction digestion, ligation, etc.) were applied to generate lentiviral expression plasmids.
Effector and target cell culturing conditions
[00273] Human primary CD8+ T cells were isolated from anonymous donor' s blood after apheresis (Trima residuals from Blood Centers of the Pacific, San Francisco, CA) by negative selection using RosetteSep Human CD8+ T Cell Enrichment Cocktail
(STEMCELL Technologies #15063) as approved by University Institutional Review Board. Cells were cultured in human T cell medium, consisting of X-VIV015 (Lonza #04-418Q), 5% human AB serum (Valley Biomedical Inc., #HP1022), lOmM N-acetyl L-Cysteine (Sigma- Aldrich #A9165) and 100 IU/mL recombinant human IL-2 (NCI/BRB Preclinical Repository). A Jurkat cell line expressing the Green Fluorescent Protein (GFP) upon NFAT activation was maintained in RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS), penicillin and streptomycin. K562 target cells from U. Penn were cultured in IMDM supplemented with 10% FBS.
Effector and target cell engineering with lentivirus
[00274] Pantropic VSV-G pseudotyped lentivirus was produced from Lenti-X 293T cells
(Clontech Laboratories #632180) co-transfected with a pHR'SIN:CSW transgene expression vector, viral packaging plasmids pCMVdR8.91 and pMD2.G using
Lipofectamine LTX (Life Technologies #15338). Infection medium supernatant was collected 48 hours after transfection and used directly for transduction.
[00275] Twenty four hours prior to viral transduction, primary human T cells were activated using the human T- Activator CD3/CD28 Dynabeads (Life Technologies #111-3 ID) at a 1 :3 celhbead ratio. Jurkat and K562 cells were split 1-2 days in advance to ensure that cultures would be in log phase at the time of transduction. Transduced Jurkat and K562 cells were cultured for at least 7 days before experiments were conducted. Primary T cells were maintained at ~10A6/mL in human T cell medium for about two weeks until cells returned to a resting state. Expression levels of CARs encoded in the lentiviral constructs were quantified by detecting either fluorophore-conjugated antibodies or fluorescent reporter proteins using a flow cytometer.
Quantitation of IL-2 production and NFAT activity
[00276] Jurkat CD4+ T cells expressing CARs were mixed with cognate or non-cognate
K562 target cells from U. Penn at a 1:2 effectontarget ratio. The rapalog A/C
Heterodimerizer (Clontech Laboratories #635055) were serially diluted in medium and added to reaction mixtures. After 20-24 hours of incubation, medium supernatants were collected and analyzed with BD OptEIA Human IL-2 ELISA Set (BD Biosciences
#555190). Flow cytometry was performed to quantify NFAT-dependent GFP reporter expression in Jurkat cells as a separate indicator for CAR activity.
Flow cytometry-based re-directed cytotoxicity assay
[00277] The cognate and non-cognate K562 target cells were engineered to express distinct fluorescent proteins so that both cell types in a mixture could be simultaneously quantified by flow cytometry. The target cell types were mixed at a 1: 1 ratio and co-incubated with human primary CD8+ effector T cells at a 5:2 effector: target ratio. 100 IU/mL human IL-2 and varying amounts of the rapalog (Clontech Laboratories #635055) were added to reaction mixtures. After 24 hours of incubation, samples were centrifuged at 400g for 5 minutes. Pelleted cells were resuspended in wash buffer (PBS + 0.5% BSA + 0.1% sodium azide) and fixed with an equal volume of BD Cytofix (BD cat #554655) prior to flow cytometry. Ratios of the surviving cognate target cells to non-cognate target cells were calculated for each sample to enumerate re-directed cytotoxic activities of the effector cells. RESULTS
[00278] IL-2 production elicited by the various CAR constructs was assessed. The data are presented in Figure 12.
[00279] Figure 12. IL-2 production triggered by five On-switch CAR variants. Effector = human CD4+ Jurkat T cells engineered with CARs. Target = K562 cell lines with or without the cognate CD 19 antigen. Amounts of secreted IL-2 by effector cells were quantified by enzyme-linked immunosorbent assay (ELISA). [00280] Figure 13. IL-2 production by control Jurkat lines in the same experiment as that described in Figure 12. Construct "125" encodes a conventional control currently used in clinical trials.
[00281] Figure 14. Comparison between "122 + 206" and "197 + 206" in a separate
experiment under conditions identical to those described in Figure 12.
[00282] Figure 15 demonstrates pharmacologically titratable cytoxicity conferred by the On- switch CAR "197+206" In the presence of the small molecule rapalog, the CAR effectively mediates re-directed cytotoxicity towards cognate target cells. At high dosages of rapalog, this On-switch CAR can signal as strongly as the "125" conventional CAR. Effector = human primary CD8+ T cells engineered with CARs or a control vector. Target = fluorescent derivatives of K562 cell lines expressing either the cognate human CD19 antigen or the non-cognate human mesothelin antigen.
[00283] Figure 16 depicts data for CARs constructed with the cytoplasmic tyrosine kinase
Zap70 from the T cell receptor pathway as the intracellular signaling domain.
[00284] Figure 16 shows data from Jurkat cells engineered with several variants of On- switch CARs. The engineered Jurkat cells were co-incubated with K562 target cells with or without the cognate antigen (CD 19) and the indicated concentrations of rapalog. As a CAR component, the Zap70 kinase (first and second structures from left featuring "199") was as effective as the IT AM (third structure from left featuring "168") in activating NFAT function. Addition of the 4- IBB signaling domain increased surface expression of the antigen recognition portion of the receptor and led to stronger signaling by "197+199". A non-signaling CAR (far-right) was included as a negative control.
Example 2: CARs targeting mesothelin
MATERIALS AND METHODS
[00285] A number of chimeric antigen receptor constructs were made and tested. The
constructs shown here encode three different anti-human mesothelin scFv as the antigen recognition domains. Figures 19A, 19B, and 19C summarize the molecular structure of each anti-human mesothelin CAR, with each CAR comprising two polypeptides. The
intercellular portion of each anti-human mesothelin CAR comprises two 4- IBB co- stimulatory domains, an FKBP and FRB dimerizer-binding pair, and an IT AM intracellular signaling domain. The three different antigen recognition domains shown here are anti- mesothelin HN1 scFv, SSI scFv, and m912 scFv. All membrane- anchored polypeptides are di-sulfide bonded homo-dimers. Generation of CAR constructs
[00286] Sequences encoding the anti-mesothelin were cloned from constructs or synthesized via gene assembly by PCR. The human 4- IBB co- stimulation and CD3 zeta ΓΓΑΜ signaling chains were cloned from cDNAs supplied by Open Biosystems. HN1 scFv-, SS I scFv-, and m912 scFv-encoding sequences were synthesized by PCR and, in some cases, codon optimized. FKBP- and FRB -encoding sequences were cloned from Addgene plasmids.
[00287] Standard molecular cloning techniques (polymerase chain reaction (PCR), restriction digestion, ligation, etc.) were applied to generate lentiviral expression plasmids.
Effector and target cell culturing conditions
[00288] A Jurkat cell line expressing GFP upon NFAT activation was maintained in RPMI-
1640 medium supplemented with 10% FBS, penicillin and streptomycin. K562 target cells were cultured in IMDM supplemented with 10% fetal bovine serum (FBS).
Effector and target cell engineering with lentivirus
[00289] Pantropic VSV-G pseudotyped lentivirus was produced from Lenti-X 293T cells
(Clontech Laboratories #632180) co-transfected with a pHR'SIN:CSW transgene expression vector, viral packaging plasmids pCMVdR8.91 and pMD2.G using
Lipofectamine LTX (Life Technologies #15338). Infection medium supernatant was collected 48 hours after transfection and used directly for transduction.
[00290] Jurkat and K562 cells were split 1-2 days in advance to ensure that cultures would be in log phase at the time of transduction. Transduced Jurkat and K562 cells were cultured for at least 7 days before experiments were conducted. Expression levels of CARs encoded in the lentiviral constructs were quantified by detecting either fluorophore-conjugated antibodies or fluorescent reporter proteins using a flow cytometer.
Quantitation of IL-2 production
[00291] Jurkat CD4+ T cells expressing CARs were mixed with cognate or non-cognate
K562 target cells at a 1 :2 effectontarget ratio. The rapalog A/C Heterodimerizer (Clontech Laboratories #635055) were serially diluted in medium and added to reaction mixtures. After 20-24 hours of incubation, medium supernatants were collected and analyzed with BD OptEIA Human IL-2 ELISA Set (BD Biosciences #555190).
RESULTS
[00292] IL-2 production elicited by the anti-mesothelin CAR constructs was assessed. The data are presented in Figure 19D-F. [00293] Figure 19. IL-2 production triggered by HN1 scFv (Fig. 19D), SS I scFv (Fig. 19E), and m912 scFv (Fig. 19F) On-switch CAR variants. IL-2 production by a conventional CAR (Fig. 19G, construct #358) was measured and included for comparison to On-switch CARs (Fig. 19D). Effector = human CD4+ Jurkat T cells engineered with CARs. Target = K562 cell lines with or without the cognate mesothelin antigen. Amounts of secreted IL-2 by effector cells were quantified by enzyme-linked immunosorbent assay (ELISA).
Example 3: Gibberellic acid as a dimerizer of On-switch CARs
MATERIALS AND METHODS
[00294] Figure 20A summarizes the molecular structure of the subject gibberellic acid
dimerizer CAR. The antigen binding portion comprises the anti-human CD 19 scFv. The intracellular portion comprises two 4- IBB co- stimulatory domains, a GID1 and GAI dimerizer-binding pair, and an IT AM intracellular signaling domain. All membrane- anchored polypeptides are di-sulfide bonded homo-dimers.
Generation of CAR constructs
[00295] Sequences encoding the gibberellic acid dimerizer CAR were cloned from
constructs. The anti-CD 19 scFv was cloned from a plasmid. The human 4- IBB co- stimulation and CD3 zeta IT AM signaling chains were cloned from cDNAs supplied by Open Biosystems. GID 1- and GAI-encoding sequences were cloned from Addgene plasmids. Standard molecular cloning techniques (polymerase chain reaction (PCR), restriction digestion, ligation, etc.) were applied to generate lentiviral expression plasmids. Effector and target cell culturing conditions
[00296] A Jurkat cell line expressing GFP upon NFAT activation was maintained in RPMI-
1640 medium supplemented with 10% FBS, penicillin and streptomycin. K562 target cells were cultured in IMDM supplemented with 10% fetal bovine serum (FBS).
Effector and target cell engineering with lentivirus
[00297] Pantropic VSV-G pseudotyped lentivirus was produced from Lenti-X 293T cells
(Clontech Laboratories #632180) co-transfected with a pHR'SIN:CSW transgene expression vector, viral packaging plasmids pCMVdR8.91 and pMD2.G using
Lipofectamine LTX (Life Technologies #15338). Infection medium supernatant was collected 48 hours after transfection and used directly for transduction.
[00298] Jurkat and K562 cells were split 1-2 days in advance to ensure that cultures would be in log phase at the time of transduction. Transduced Jurkat and K562 cells were cultured for at least 7 days before experiments were conducted. Expression levels of CARs encoded in the lentiviral constructs were quantified by detecting either fluorophore-conjugated antibodies or fluorescent reporter proteins using a flow cytometer.
Quantitation of IL-2 production
[00299] Jurkat CD4+ T cells expressing CARs were mixed with cognate or non-cognate
K562 target cells at a 1 :2 effectontarget ratio. The gibberellic acid-3 acetoxymethyl ester (gibberrelic acid-3 AM) pre-dissolved in ethanol (Toronto Research Chemicals #G377500) was diluted in growth medium and added to reaction mixtures. Gibberellic acid (gibberellic acid-3 AM) was used at 10 mM. After 20-24 hours of incubation, medium supernatants were collected and analyzed with BD OptEIA Human IL-2 ELISA Set (BD Biosciences #555190).
RESULTS
[00300] IL-2 production elicited by the gibberellic acid dimerizer CAR construct was
assessed. The data are presented in Figure 20.
[00301] Figure 20. IL-2 production triggered by gibberellic acid dimerizer CAR variant (Fig.
20B). IL-2 production by a conventional CAR (Fig. 20C, construct "125") was measured and included for comparison to On-switch CAR. Effector = human CD4+ Jurkat T cells engineered with CARs. Target = K562 cell lines with or without the cognate CD19 antigen. Amounts of secreted IL-2 by effector cells were quantified by enzyme-linked
immunosorbent assay (ELISA).
Example 4: On-switch CARs with various co- stimulatory domains
MATERIALS AND METHODS
[00302] A number of chimeric antigen receptor constructs were made essentially as
described for Example 1, except various other co-stimulatory domains were exchanged for the 4- IBB co- stimulatory domains. Figures 21 A and 2 IB summarize the molecular structure of the CARs described here.
Generation of CAR constructs
[00303] Sequences encoding the anti-human CD 19 scFv were cloned from a plasmid. The human CD3 zeta ΓΓΑΜ signaling chain and the human co- stimulatory domains CD28 and
OX-40 encoding sequences were cloned from cDNAs supplied by Open Biosystems.
FKBP- and FRB-encoding sequences were cloned from plasmids from Addgene.
[00304] Standard molecular cloning techniques (polymerase chain reaction (PCR), restriction digestion, ligation, etc.) were applied to generate lentiviral expression plasmids. Testing of CAR constructs
[00305] Effector and target cells are cultured and transfected according to Example 1 using the on-switch CAR CD28 and OX-40 co- stimulatory domain containing constructs described (Fig. 21A-B, constructs "365+367" and "399+400", respectively) and corresponding conventional CAR controls (Fig. 21C-D, constructs "366" and "398", respectively). IL-2 production, NFAT activity assays, and flow cytometry-based assays can also be performed with the CD28 co- stimulatory domain containing construct and OX-40 co-stimulatory domain containing construct as described for Example 1. Alternatively, subunits of on-switch CAR CD28 and OX-40 co- stimulatory domain containing constructs can be paired with subunits of constructs from Example 1 (e.g., "197+367", " 365+206," "197+400", "399+206," etc.).
Example 5: In vivo assessment of On-switch CAR
[00306] An On-switch CAR can be assessed for its ability to mediate in vivo killing of a target tumor cell. In vivo tumor cell killing elicited by injection of T cells expressing the ON-switch CAR is assessed. Tumor cell lines that have been confirmed in vitro to express the cognate antigen and can be killed by CD8+ T cells expressing the corresponding CAR are used. Tumor cells engineered to express either the firefly or Renilla lucif erase to enable bio-luminescence imaging to quantify tumor burden in vivo can be used. Tumor cells are injected into immunocompromised mice (e.g., 6-10 week old female NOD scid gamma (NSG) mice) either subcutaneously for subcutaneous tumor models or intravenously for systemic tumor models. The method of tumor implantation and the optimal number of tumor cells to implant can be based on conditions optimal for the tumor cell line used. Tumor burden can be monitored twice a week by bio-luminescence imaging and by caliper measurement when applicable. As soon as tumor burden is detectable, 0.5-2.5 x 10Λ7 total T cells (1: 1 CD4+:CD8+) expressing the ON-switch CAR are intravenously injected into mice to begin treatment. A dimerizing small molecule drug (e.g., rapalog) is administered intraperitoneally in a vehicle formulation. On-switch CAR-expressing T cells can be injected repeatedly during the experiment to enhance the anti-tumor effect. Interleukin-2 (IL-2) can be administered to enhance the anti-tumor effect.
[00307] While the present invention has been described with reference to the specific
embodiments thereof, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, many modifications may be made to adapt a particular situation, material, composition of matter, process, process step or steps, to the objective, spirit and scope of the present invention. All such modifications are intended to be within the scope of the claims appended hereto.

Claims

[00308] CLAIMS What is claimed is:
1. A heterodimenc, conditionally active chimeric antigen receptor (CAR) comprising:
a) a first polypeptide comprising:
i) a first member of a specific binding pair;
ii) a first modulatory domain;
iii) a first member of a dimerization pair; and
iv) a transmembrane domain interposed between the first member of a specific binding pair and the first modulatory domain; and
b) a second polypeptide comprising:
i) a transmembrane domain;
ii) a second modulatory domain;
iii) a second member of the dimerization pair; and
iv) an intracellular signaling domain;
or comprising:
a) a first polypeptide comprising:
i) a first member of a specific binding pair;
ii) a modulatory domain;
iii) a first member of a dimerization pair;
iv) a transmembrane domain interposed between the first member of a specific binding pair and the modulatory domain; and
b) a second polypeptide comprising:
i) a second member of the dimerization pair; and
ii) an intracellular signaling domain.
2. The heterodimeric, conditionally active CAR of claim 1, wherein the first polypeptide comprises a hinge region interposed between the first member of the specific binding pair and the transmembrane domain.
3. The heterodimeric, conditionally active CAR of claim 1, wherein the first member of the specific binding pair is an antibody or antibody fragment, a ligand, or a receptor.
4. The heterodimeric, conditionally active CAR of claim 2, wherein the hinge region is an immunoglobulin IgG hinge region or a hinge derived from CD 8.
5. The heterodimeric, conditionally active CAR of claim 1, wherein the first and second modulatory domains are selected from 4-lBB (CD137), CD28, ICOS, BTLA, OX- 40, CD27, CD30, GITR, HVEM, DAP10, DAP 12, and CD28.
6. The heterodimeric, conditionally active CAR of claim 1, wherein the intracellular signaling domain is selected from ZAP70 and CD3-zeta.
7. The heterodimeric, conditionally active CAR of claim 1, wherein the intracellular signaling domain comprises an immunoreceptor tyrosine-based activation motif (ITAM).
8. The heterodimeric, conditionally active CAR of claim 1, wherein the first and second members of the dimerization pair form a homodimer in the presence of a small molecule dimerizer.
9. The heterodimeric, conditionally active CAR of claim 1, wherein the first and second members of the dimerization pair form a heterodimer in the presence of a small molecule dimerizer.
10. The heterodimeric, conditionally active CAR of claim 1, wherein the first and second members of the dimerization pair are selected from:
a) FK506 binding protein (FKBP) and FKBP;
b) FKBP and calcineurin catalytic subunit A (CnA);
c) FKBP and cyclophilin;
d) FKBP and FKBP-rapamycin associated protein (FRB);
e) gyrase B (GyrB) and GyrB;
f) dihydrofolate reductase (DHFR) and DHFR; g) DmrB and DmrB;
h) PYL and ABI;
i) Cry2 and CIP;
j) GAI and GID1.
11. The heterodimeric, conditionally active CAR of claim 1, wherein:
i) the first and second modulatory domains are derived from 4- IBB;
ii) the first and second members of the dimerization pair are FKBP and FRB; and ii) the signaling domain comprises an ITAM.
12. The heterodimeric, conditionally active CAR of claim 1, wherein the first member of the specific binding pair is a single-chain Fv.
13. The heterodimeric, conditionally active CAR of claim 1, wherein the first member of the specific binding pair binds an epitope present on a cell, on a solid surface, or a lipid bilayer.
14. The heterodimeric, conditionally active CAR of claim 13, wherein the cell is a cancer cell.
15. A mammalian cell genetically modified to produce the heterodimeric, conditionally active CAR of claim 1.
16. The cell of claim 15, wherein the cell is a stem cell, a progenitor cell, or a cell derived from a stem cell or a progenitor cell.
17. The cell of claim 15, wherein the cell is a T lymphocyte or an NK cell.
18. A nucleic acid comprising nucleotide sequences encoding the heterodimeric, conditionally active CAR of claim 1.
19. The nucleic acid of claim 18, wherein the nucleotide sequences are operably linked to a T lymphocyte- specific promoter or an NK cell- specific promoter.
20. The nucleic acid of claim 18, wherein the nucleic acid is in vitro transcribed
RNA.
21. A recombinant expression vector comprising the nucleic acid of claim 18.
22. A method of activating a T lymphocyte, the method comprising contacting the T lymphocyte with a dimerizing agent and a second member of a specific binding pair, wherein the T lymphocyte is genetically modified to produce a heterodimeric, conditionally active CAR of claim 1, and wherein, in the presence of the dimerizing agent and the second member of a specific binding pair, the heterodimeric, conditionally active CAR dimerizes and activates the T lymphocyte, thereby producing an activated T lymphocyte.
23. The method of claim 22, wherein the second member of a specific binding pair is an antigen.
24. The method of claim 22, wherein said contacting occurs in vivo.
25. The method of claim 22, wherein the activated T lymphocyte mediates killing of a target cell.
26. The method of claim 22, wherein the activated T lymphocyte produces IL-2 and/or IFN-γ.
27. The method of claim 25, wherein the target cell is a cancer cell.
28. The method of claim 22, wherein the first member of the specific binding pair of the heterodimeric, conditionally active CAR is an antibody specific for an epitope on a cancer cell.
29. A method of making the cell of claim 15, the method comprising genetically modifying a mammalian cell with an expression vector comprising nucleotide sequences encoding the heterodimeric, conditionally active CAR of claim 1, or genetically modifying a mammalian cell with an RNA comprising nucleotide sequences encoding the heterodimeric, conditionally active CAR of claim 1.
30. The method of claim 29, wherein said genetic modification is carried out ex
VIVO .
31. The method of claim 29, wherein the cell is a T lymphocyte, a stem cell, an NK cell, a progenitor cell, a cell derived from a stem cell, or a cell derived from a progenitor cell.
32. A method of treating a cancer in an individual, the method comprising:
i) genetically modifying T lymphocytes obtained from the individual with an expression vector comprising nucleotide sequences encoding the heterodimeric,
conditionally active CAR of claim 1, wherein the antigen-binding domain of the
heterodimeric, conditionally active CAR is specific for an epitope on a cancer cell in the individual, and wherein said genetic modification is carried out ex vivo;
ii) introducing the genetically modified T lymphocytes into the individual; and iii) administering to the individual an effective amount of a dimerizing agent, wherein the dimerizing agent induces dimerization of the heterodimeric, conditionally active CAR, wherein said dimerization provides for activation of the genetically modified T lymphocytes and killing of the cancer cell, thereby treating the cancer.
33. The method of claim 32, wherein the dimerizing agent is a rapalog.
34. A method of modulating the activity of a host cell, the method comprising contacting the host cell with a dimerizing agent and a second member of a specific binding pair, wherein the T lymphocyte is genetically modified to produce a heterodimeric, conditionally active CAR of claim 1, and wherein, in the presence of the dimerizing agent and the second member of a specific binding pair, the heterodimeric, conditionally active CAR dimerizes and modulates at least one activity of the host cell.
35. The method of claim 34, wherein the activity is proliferation, cell survival, apoptosis, gene expression, or immune activation.
36. The method of claim 34, wherein the second member of a specific binding pair is an antigen.
PCT/US2014/016527 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof WO2014127261A1 (en)

Priority Applications (42)

Application Number Priority Date Filing Date Title
NZ710925A NZ710925B2 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
EP17187293.0A EP3300745B9 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
EP21158396.8A EP3881868B1 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
EP19188398.2A EP3613439B1 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
KR1020217038402A KR20210147101A (en) 2013-02-15 2014-02-14 Chimeric Antigen Receptor and Methods of Use Thereof
JP2015558159A JP6450690B2 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and method of use thereof
CA2901115A CA2901115A1 (en) 2013-02-15 2014-02-14 Heterodimeric conditionally active chimeric antigen receptor and methods of use thereof
BR112015019640-3A BR112015019640A2 (en) 2013-02-15 2014-02-14 heterodimeric conditionally active chimeric antigen (car) receptor, genetically modified in vitro or ex vivo mammalian cell, nucleic acid, method of activating a t-lymphocyte in vitro or ex vivo, method of producing a cell, and method of modulating the T lymphocyte host cell activity
IL272279A IL272279B2 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
CN201910722248.1A CN110423282B (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
EP14751227.1A EP2956175B1 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
IL305629A IL305629A (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
CN201480016737.0A CN105142677B (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and its application method
EP23198143.2A EP4303232A3 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
KR1020157024731A KR102064230B1 (en) 2013-02-15 2014-02-14 Chimeric Antigen Receptor and Methods of Use Thereof
ES14751227T ES2653487T3 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
DK14751227.1T DK2956175T3 (en) 2013-02-15 2014-02-14 CHEMICAL ANTIGEN RECEPTOR AND PROCEDURES FOR USE THEREOF
MX2015010522A MX369545B (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof.
KR1020207019131A KR102332790B1 (en) 2013-02-15 2014-02-14 Chimeric Antigen Receptor and Methods of Use Thereof
KR1020197034038A KR102132246B1 (en) 2013-02-15 2014-02-14 Chimeric Antigen Receptor and Methods of Use Thereof
LTEP14751227.1T LT2956175T (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
KR1020237003986A KR20230022452A (en) 2013-02-15 2014-02-14 Chimeric Antigen Receptor and Methods of Use Thereof
PL14751227T PL2956175T3 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
PL19188398T PL3613439T3 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
PL17187293T PL3300745T3 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
US14/766,105 US20150368342A1 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
AU2014216130A AU2014216130B2 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
IL240472A IL240472B (en) 2013-02-15 2015-08-10 Chimeric antigen receptor and methods of use thereof
US15/064,938 US9587020B2 (en) 2013-02-15 2016-03-09 Chimeric antigen receptor and methods of use thereof
HK16106629.8A HK1218625A1 (en) 2013-02-15 2016-06-08 Chimeric antigen receptor and methods of use thereof
US15/419,729 US9821012B2 (en) 2013-02-15 2017-01-30 Chimeric antigen receptor and methods of use thereof
US15/669,707 US10632152B2 (en) 2013-02-15 2017-08-04 Chimeric antigen receptor and methods of use thereof
US15/801,133 US10105391B2 (en) 2013-02-15 2017-11-01 Chimeric antigen receptor and methods of use thereof
US15/835,329 US10888581B2 (en) 2013-02-15 2017-12-07 Chimeric antigen receptor and methods of use thereof
AU2018201102A AU2018201102A1 (en) 2013-02-15 2018-02-15 Chimeric antigen receptor and methods of use thereof
AU2019246785A AU2019246785B2 (en) 2013-02-15 2019-10-08 Chimeric antigen receptor and methods of use thereof
CY20191101245T CY1122386T1 (en) 2013-02-15 2019-11-27 CHIMERAL ANTIGONE RECEPTOR AND METHODS OF USING THEREOF
US16/824,434 US11478510B2 (en) 2013-02-15 2020-03-19 Chimeric antigen receptor and methods of use thereof
US17/115,565 US20210196757A1 (en) 2013-02-15 2020-12-08 Chimeric antigen receptor and methods of use thereof
AU2021204054A AU2021204054B2 (en) 2013-02-15 2021-06-17 Chimeric antigen receptor and methods of use thereof
US17/932,259 US20230051989A1 (en) 2013-02-15 2022-09-14 Chimeric antigen receptor and methods of use thereof
AU2023204612A AU2023204612A1 (en) 2013-02-15 2023-07-12 Chimeric antigen receptor and methods of use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361765585P 2013-02-15 2013-02-15
US61/765,585 2013-02-15

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US14/766,105 A-371-Of-International US20150368342A1 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof
US15/064,938 Continuation US9587020B2 (en) 2013-02-15 2016-03-09 Chimeric antigen receptor and methods of use thereof
US15/669,707 Continuation US10632152B2 (en) 2013-02-15 2017-08-04 Chimeric antigen receptor and methods of use thereof

Publications (1)

Publication Number Publication Date
WO2014127261A1 true WO2014127261A1 (en) 2014-08-21

Family

ID=51354582

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/016527 WO2014127261A1 (en) 2013-02-15 2014-02-14 Chimeric antigen receptor and methods of use thereof

Country Status (22)

Country Link
US (9) US20150368342A1 (en)
EP (5) EP4303232A3 (en)
JP (5) JP6450690B2 (en)
KR (5) KR20230022452A (en)
CN (2) CN110423282B (en)
AU (5) AU2014216130B2 (en)
BR (1) BR112015019640A2 (en)
CA (1) CA2901115A1 (en)
CY (1) CY1122386T1 (en)
DK (2) DK2956175T3 (en)
ES (3) ES2758227T3 (en)
HK (2) HK1218625A1 (en)
HR (1) HRP20192123T1 (en)
HU (2) HUE047487T2 (en)
IL (3) IL272279B2 (en)
LT (2) LT2956175T (en)
MX (2) MX369545B (en)
NO (1) NO2929995T3 (en)
PL (3) PL3613439T3 (en)
PT (3) PT3300745T (en)
SI (2) SI3300745T1 (en)
WO (1) WO2014127261A1 (en)

Cited By (167)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014145252A2 (en) * 2013-03-15 2014-09-18 Milone Michael C Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2015017214A1 (en) 2013-07-29 2015-02-05 Bluebird Bio, Inc. Multipartite signaling proteins and uses thereof
WO2015090229A1 (en) 2013-12-20 2015-06-25 Novartis Ag Regulatable chimeric antigen receptor
WO2015142661A1 (en) 2014-03-15 2015-09-24 Novartis Ag Regulatable chimeric antigen receptor
WO2015150771A1 (en) * 2014-04-01 2015-10-08 Ucl Business Plc Chimeric antigen receptor (car) signalling system
WO2015157252A1 (en) 2014-04-07 2015-10-15 BROGDON, Jennifer Treatment of cancer using anti-cd19 chimeric antigen receptor
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
WO2016033331A1 (en) * 2014-08-28 2016-03-03 Bioatla, Llc Conditionally active chimeric antigen receptors for modified t-cells
WO2016030691A1 (en) * 2014-08-29 2016-03-03 Ucl Business Plc Signalling system
WO2016044605A1 (en) * 2014-09-17 2016-03-24 Beatty, Gregory Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2016054013A1 (en) * 2014-10-03 2016-04-07 Yale University Innate immune system modification for anticancer therapy
WO2016057705A1 (en) 2014-10-08 2016-04-14 Novartis Ag Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
EP3025719A1 (en) 2014-11-26 2016-06-01 Miltenyi Biotec GmbH Combination immunotherapy of antigen-recognizing receptors and hematopoietic cells for the treatment of diseases
WO2016090312A1 (en) * 2014-12-05 2016-06-09 Memorial Sloan-Kettering Cancer Center Chimeric antigen receptors targeting g-protein coupled receptor and uses thereof
WO2016097334A1 (en) * 2014-12-19 2016-06-23 ETH Zürich Chimeric antigen receptors and methods of use
WO2016124930A1 (en) * 2015-02-05 2016-08-11 Ucl Business Plc Signalling system
WO2016151315A1 (en) * 2015-03-23 2016-09-29 Ucl Business Plc Chimeric antigen receptor
WO2016164731A2 (en) 2015-04-08 2016-10-13 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
WO2016168595A1 (en) 2015-04-17 2016-10-20 Barrett David Maxwell Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
WO2016098078A3 (en) * 2014-12-19 2016-10-27 Novartis Ag Dimerization switches and uses thereof
WO2016174405A1 (en) * 2015-04-27 2016-11-03 Ucl Business Plc Nucleic acid construct for expressing more than one chimeric antigen receptor
WO2016174407A1 (en) * 2015-04-27 2016-11-03 Ucl Business Plc Nucleic acid construct for expressing more than one chimeric antigen receptor
WO2016174406A1 (en) * 2015-04-27 2016-11-03 Ucl Business Plc Nucleic acid construct for expressing more than one chimeric antigen receptor
WO2016161415A3 (en) * 2015-04-02 2016-12-01 Memorial Sloan Kettering Cancer Center Tnfrsf14/ hvem proteins and methods of use thereof
WO2016168773A3 (en) * 2015-04-15 2016-12-01 The California Institute For Biomedical Research Optimized pne-based chimeric receptor t cell switches and uses thereof
WO2016210293A1 (en) * 2015-06-25 2016-12-29 Icell Gene Therapeutics Llc CHIMERIC ANTIGEN RECEPTORS (CARs), COMPOSITIONS AND METHODS OF USE THEREOF
WO2017015427A1 (en) 2015-07-21 2017-01-26 Novartis Ag Methods for improving the efficacy and expansion of immune cells
US9587020B2 (en) 2013-02-15 2017-03-07 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof
WO2017040930A2 (en) 2015-09-03 2017-03-09 The Trustees Of The University Of Pennsylvania Biomarkers predictive of cytokine release syndrome
JP2017507917A (en) * 2014-01-14 2017-03-23 セレクティスCellectis Chimeric antigen receptor using antigen recognition domain from cartilaginous fish
WO2017087723A1 (en) * 2015-11-19 2017-05-26 The Regents Of The University Of California Conditionally repressible immune cell receptors and methods of use thereof
WO2017091546A1 (en) * 2015-11-23 2017-06-01 Trustees Of Boston University Methods and compositions relating to chimeric antigen receptors
EP3184548A1 (en) 2015-12-23 2017-06-28 Miltenyi Biotec GmbH Chimeric antigen receptor with cytokine receptor activating or blocking domain
WO2017120546A1 (en) * 2016-01-08 2017-07-13 The Regents Of The University Of California Conditionally active heterodimeric polypeptides and methods of use thereof
WO2017137759A1 (en) * 2016-02-12 2017-08-17 Autolus Limited Signalling system
WO2017137758A1 (en) * 2016-02-12 2017-08-17 Autolus Limited Signalling system
CN107109368A (en) * 2014-10-07 2017-08-29 塞勒克提斯公司 For the method for the immunologic cellular activity for adjusting CAR inductions
EP3211420A1 (en) 2016-02-29 2017-08-30 Miltenyi Biotec GmbH Assay for detection of chimeric antigen receptor t cells
WO2017173403A1 (en) * 2016-03-31 2017-10-05 University Of Southern California A highly sensitive and specific luciferase based reporter assay for antigen detection
WO2017173410A1 (en) 2016-04-01 2017-10-05 Amgen Inc. Chimeric receptors to flt3 and methods of use thereof
WO2017173256A1 (en) 2016-04-01 2017-10-05 Kite Pharma, Inc. Chimeric antigen and t cell receptors and methods of use
WO2017173384A1 (en) 2016-04-01 2017-10-05 Kite Pharma, Inc. Chimeric receptors and methods of use thereof
WO2017210617A2 (en) 2016-06-02 2017-12-07 Porter, David, L. Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
US20180072789A1 (en) * 2014-12-02 2018-03-15 Roger Williams Hospital Methods and compositions for treating cancer
WO2018067992A1 (en) 2016-10-07 2018-04-12 Novartis Ag Chimeric antigen receptors for the treatment of cancer
US9944690B2 (en) 2013-03-14 2018-04-17 Bellicum Pharmaceuticals, Inc. Methods for controlling T cell proliferation
CN108138362A (en) * 2015-09-01 2018-06-08 加利福尼亚大学董事会 Modularization polypeptide libraries and its preparation and application
WO2018111834A1 (en) * 2016-12-13 2018-06-21 Seattle Children's Hospital (dba Seattle Children's Research Institute) Methods of exogenous drug activation of chemical-induced signaling complexes expressed in engineered cells in vitro and in vivo
WO2018112266A1 (en) 2016-12-14 2018-06-21 The Board Of Trustees Of The Leland Stanford Junior University Il-13 superkine: immune cell targeting constructs and methods of use thereof
JP2018522904A (en) * 2015-08-05 2018-08-16 株式会社柳英製薬Yoo Young Pharm Co., Ltd. Chimeric antigen receptor and T cell expressing chimeric antigen receptor
WO2018169922A2 (en) 2017-03-13 2018-09-20 Kite Pharma, Inc. Chimeric antigen receptors for melanoma and uses thereof
WO2018200496A1 (en) 2017-04-24 2018-11-01 Kite Pharma, Inc. Humanized antigen-binding domains against cd19 and methods of use
WO2018211244A1 (en) * 2017-05-15 2018-11-22 Autolus Limited A cell comprising a chimeric antigen receptor (car)
EP3447075A2 (en) 2015-05-15 2019-02-27 The General Hospital Corporation Antagonistic anti-tumor necrosis factor receptor superfamily antibodies
WO2019051001A1 (en) * 2017-09-06 2019-03-14 California Institute Of Technology Signaling and antigen-presenting bifunctional receptors (sabr)
WO2019064030A1 (en) * 2017-09-29 2019-04-04 TC Biopharm Limited Modified car-t
WO2019062518A1 (en) * 2017-09-26 2019-04-04 南京安吉生物科技有限公司 New dual chimeric antigen receptor-t cell which can be regulated, construction method therefor and use thereof
US10273280B2 (en) 2015-02-27 2019-04-30 Icell Gene Therapeutics Llc Chimeric antigen receptors (CARs), targeting hematologic malignancies, compositions and methods of use thereof
WO2019084288A1 (en) 2017-10-25 2019-05-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2019099707A1 (en) 2017-11-16 2019-05-23 Kite Pharma, Inc Modified chimeric antigen receptors and methods of use
JPWO2018052142A1 (en) * 2016-09-16 2019-06-24 キッセイ薬品工業株式会社 Genetically modified cell and method for producing the same
EP3502130A1 (en) 2017-12-20 2019-06-26 St. Anna Kinderkrebsforschung Ligand regulated protein-protein interaction system
WO2019122188A1 (en) 2017-12-20 2019-06-27 St. Anna Kinderkrebsforschung Ligand regulated protein-protein interaction system
WO2019126358A1 (en) * 2017-12-22 2019-06-27 Cell Design Labs, Inc. Single- and multi-chain chimeric antigen receptors
WO2019134950A1 (en) 2018-01-04 2019-07-11 Miltenyi Biotec Gmbh Chimeric antigen receptor specific for bdca2 antigen
WO2019115818A3 (en) * 2017-12-14 2019-07-25 Celyad S.A. Pooling signaling and costimulatory domains in flexible car design
EP3517125A1 (en) * 2018-01-24 2019-07-31 Xuanwu Hospital of Capital Medical University Chimeric antigen receptor for efficient targeted proliferation in vitro and uses thereof
WO2019161271A1 (en) 2018-02-16 2019-08-22 Kite Pharma, Inc. Modified pluripotent stem cells and methods of making and use
US10428305B2 (en) 2014-05-15 2019-10-01 National University Of Singapore Modified natural killer cells that express IL15 and uses thereof
WO2019197676A1 (en) * 2018-04-13 2019-10-17 Ludwig Institute For Cancer Research Ltd Heterodimeric inactivatable chimeric antigen receptors
WO2019200007A1 (en) 2018-04-10 2019-10-17 Amgen Inc. Chimeric receptors to dll3 and methods of use thereof
EP3556768A1 (en) 2014-12-24 2019-10-23 Aadigen, LLC Peptides and nanoparticles for intracellular delivery of molecules
KR20190121716A (en) 2018-04-18 2019-10-28 앱클론(주) Switch Molecule And Switchable Chimeric Antigen Receptor
WO2019213282A1 (en) 2018-05-01 2019-11-07 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
US10478457B2 (en) 2015-02-24 2019-11-19 Ucl Business Ltd Chimeric protein
WO2019210207A3 (en) * 2018-04-27 2019-11-28 The Trustees Of The University Of Pennsylvania Chimeric antigen receptor t regulatory cells for the treatment of atherosclerosis
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
WO2019232523A1 (en) 2018-06-01 2019-12-05 The Board Of Trustees Of The Leland Stanford Junior University Il-13/il-4 superkines: immune cell targeting constructs and methods of use thereof
US10538739B2 (en) 2013-01-28 2020-01-21 St. Jude Children's Research Hospital, Inc. Chimeric receptor with NKG2D specificity for use in cell therapy against cancer and infectious disease
WO2020018695A1 (en) 2018-07-18 2020-01-23 Amgen Inc. Chimeric receptors to steap1 and methods of use thereof
WO2020033927A2 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Processes for generating engineered cells and compositions thereof
WO2020047452A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020047449A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
US10590196B2 (en) 2014-12-05 2020-03-17 Memorial Sloan-Kettering Cancer Center Antibodies targeting G-protein coupled receptor and methods of use
WO2020069409A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
WO2020069405A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd22 chimeric antigen receptor (car) therapies
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
EP3632460A1 (en) 2018-10-05 2020-04-08 St. Anna Kinderkrebsforschung A group of chimeric antigen receptors (cars)
EP3632461A1 (en) 2018-10-05 2020-04-08 St. Anna Kinderkrebsforschung A group of chimeric antigen receptors (cars)
WO2020070289A1 (en) 2018-10-05 2020-04-09 St. Anna Kinderkrebsforschung A group of chimeric antigen receptors (cars)
WO2020070290A1 (en) 2018-10-05 2020-04-09 St. Anna Kinderkrebsforschung A group of chimeric antigen receptors (cars)
WO2020050993A3 (en) * 2018-09-07 2020-04-16 Bioatla, Llc Conditionally active chimeric antigen receptors for modified t-cells
EP3660042A1 (en) 2014-07-31 2020-06-03 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
WO2020117952A2 (en) 2018-12-05 2020-06-11 Genentech, Inc. Diagnostic methods and compositions for cancer immunotherapy
WO2020123691A2 (en) 2018-12-12 2020-06-18 Kite Pharma, Inc Chimeric antigen and t cell receptors and methods of use
US10689450B2 (en) 2016-04-01 2020-06-23 Kite Pharma, Inc BCMA binding molecules and methods of use thereof
US10703794B2 (en) 2015-07-31 2020-07-07 King's College London Therapeutic agents
WO2020152197A1 (en) 2019-01-23 2020-07-30 Miltenyi Biotec B.V. & Co. KG A combination of compositions for elimination and enhanced engraftment of hematopoietic stem cells in the bone marrow of a subject
WO2020180591A1 (en) 2019-03-01 2020-09-10 Allogene Therapeutics, Inc. Dll3 targeting chimeric antigen receptors and binding agents
WO2020201527A1 (en) 2019-04-04 2020-10-08 Umc Utrecht Holding B.V. Modified immune receptor constructs
WO2020210678A1 (en) 2019-04-12 2020-10-15 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020219812A1 (en) 2019-04-26 2020-10-29 Allogene Therapeutics, Inc. Methods of manufacturing allogeneic car t cells
WO2020219848A1 (en) 2019-04-26 2020-10-29 Allogene Therapeutics, Inc. Rituximab-resistant chimeric antigen receptors and uses thereof
WO2020223571A1 (en) 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cells expressing a chimeric receptor from a modified cd247 locus, related polynucleotides and methods
WO2020223535A1 (en) 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cells expressing a recombinant receptor from a modified tgfbr2 locus, related polynucleotides and methods
WO2020239866A1 (en) 2019-05-28 2020-12-03 Miltenyi Biotec B.V. & Co. KG Method for generation of genetically modified t cells
US10888608B2 (en) 2014-09-02 2021-01-12 Bellicum Pharmaceuticals, Inc. Costimulation of chimeric antigen receptors by MyD88 and CD40 polypeptides
US10934346B2 (en) 2014-02-14 2021-03-02 Bellicum Pharmaceuticals, Inc. Modified T cell comprising a polynucleotide encoding an inducible stimulating molecule comprising MyD88, CD40 and FKBP12
EP3580212A4 (en) * 2017-02-08 2021-03-17 Dana Farber Cancer Institute, Inc. Regulating chimeric antigen receptors
WO2021047804A1 (en) 2019-09-11 2021-03-18 Miltenyi Biotec B.V. & Co. KG In vitro method for transduction of t cells in the presence of malignant cells
US10988542B2 (en) 2015-08-24 2021-04-27 Cellectis Chimeric antigen receptors with integrated controllable functions
US11046954B2 (en) 2015-08-06 2021-06-29 Dana-Farber Cancer Institute, Inc. Targeted protein degradation to attenuate adoptive T-cell therapy associated adverse inflammatory responses
CN113087805A (en) * 2019-12-31 2021-07-09 华东师范大学 Preparation and application of chimeric antigen receptor T cell of co-expression immune regulatory molecule
WO2021156277A1 (en) 2020-02-04 2021-08-12 Miltenyi Biotec B.V. & Co. KG Immune cell expressing adapter chimeric antigen receptor for sensing soluble antigens
WO2021163618A1 (en) 2020-02-14 2021-08-19 Novartis Ag Method of predicting response to chimeric antigen receptor therapy
EP3723787A4 (en) * 2017-12-14 2021-09-01 Bluebird Bio, Inc. Daric interleukin receptors
WO2021173995A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021173985A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
US11111310B2 (en) 2017-03-28 2021-09-07 Cell Design Labs, Inc. Chimeric polypeptides and methods of altering the membrane localization of the same
US11111288B2 (en) 2014-08-28 2021-09-07 Bioatla, Inc. Conditionally active chimeric antigen receptors for modified t-cells
EP3684424A4 (en) * 2017-09-22 2021-09-29 H. Lee Moffitt Cancer Center And Research Institute, Inc. Chimeric antigen receptors with enhanced nfkb signaling
US11141436B2 (en) 2019-03-05 2021-10-12 Nkarta, Inc. Immune cells engineered to express CD19-directed chimeric antigen receptors and uses thereof in immunotherapy
US11173179B2 (en) 2015-06-25 2021-11-16 Icell Gene Therapeutics Llc Chimeric antigen receptor (CAR) targeting multiple antigens, compositions and methods of use thereof
EP3916388A1 (en) 2020-05-27 2021-12-01 Miltenyi Biotec B.V. & Co. KG Reagents for detection of chimeric antigen receptor cells
EP3915578A1 (en) 2020-05-28 2021-12-01 Miltenyi Biotec B.V. & Co. KG Chimeric antigen receptor with a spacer comprising c2-set ig-like domains
WO2021260186A1 (en) 2020-06-26 2021-12-30 Juno Therapeutics Gmbh Engineered t cells conditionally expressing a recombinant receptor, related polynucleotides and methods
EP3964585A1 (en) 2020-09-03 2022-03-09 Miltenyi Biotec B.V. & Co. KG Cd62l specific lentiviral vector particle for targeted transduction of t cell subsets
WO2022049217A1 (en) 2020-09-04 2022-03-10 Miltenyi Biotec B.V. & Co. KG System for inducible expression of an adapter in immune cells
US11286291B2 (en) * 2016-10-04 2022-03-29 Precision Biosciences, Inc. Co-stimulatory domains for use in genetically-modified cells
US11311576B2 (en) 2018-01-22 2022-04-26 Seattle Children's Hospital Methods of use for CAR T cells
US11325957B2 (en) 2017-06-19 2022-05-10 Cell Design Labs, Inc. Methods and compositions for reducing the immunogenicity of chimeric notch receptors
WO2022096664A1 (en) 2020-11-09 2022-05-12 Miltenyi Biotec B.V. & Co. KG Methods and compositions for eliminating engineered immune cells
WO2022104061A1 (en) 2020-11-13 2022-05-19 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
US11365236B2 (en) 2017-03-27 2022-06-21 Nkarta, Inc. Truncated NKG2D chimeric receptors and uses thereof in natural killer cell immunotherapy
US20220195039A1 (en) * 2016-04-14 2022-06-23 2Seventy Bio, Inc. Salvage chimeric antigen receptor systems
WO2022140388A1 (en) 2020-12-21 2022-06-30 Allogene Therapeutics, Inc. Protease-activating cd45-gate car
US11377637B2 (en) 2016-04-15 2022-07-05 Memorial Sloan Kettering Cancer Center Transgenic T cell and chimeric antigen receptor T cell compositions and related methods
US11384131B2 (en) 2014-04-24 2022-07-12 The Board Of Trustees Of The Leland Stanford Junior University Superagonists, partial agonists and antagonists of interleukin-2
EP4036109A2 (en) 2014-12-29 2022-08-03 Novartis AG Methods of making chimeric antigen receptor-expressing cells
WO2022165378A1 (en) * 2021-01-29 2022-08-04 Outpace Bio, Inc. Small molecule-regulated cell signaling expression system
CN114929752A (en) * 2019-10-08 2022-08-19 省卫生服务机构 Chimeric cytokine receptors
WO2022258981A1 (en) 2021-06-11 2022-12-15 Quell Therapeutics Limited Multichain chimeric antigen receptor
US11542312B2 (en) 2017-06-19 2023-01-03 Medicenna Therapeutics, Inc. IL-2 superagonists in combination with anti-PD-1 antibodies
WO2023009989A1 (en) 2021-07-26 2023-02-02 Kite Pharma, Inc. Split chimeric antigen receptors and methods of use
WO2023044304A1 (en) * 2021-09-15 2023-03-23 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric adaptor and kinase signaling proteins and their use in immunotherapy
WO2023057285A1 (en) 2021-10-06 2023-04-13 Miltenyi Biotec B.V. & Co. KG Method for targeted gene insertion into immune cells
EP4166148A1 (en) * 2014-06-06 2023-04-19 Memorial Sloan-Kettering Cancer Center Mesothelin-targeted chimeric antigen receptors and uses thereof
WO2023062113A1 (en) 2021-10-15 2023-04-20 Miltenyi Biotec B.V. & Co. KG Method for the generation of genetically modified nk cells
US11649288B2 (en) 2017-02-07 2023-05-16 Seattle Children's Hospital Phospholipid ether (PLE) CAR T cell tumor targeting (CTCT) agents
US11667691B2 (en) * 2015-08-07 2023-06-06 Novartis Ag Treatment of cancer using chimeric CD3 receptor proteins
US11680090B2 (en) 2013-09-24 2023-06-20 Medicenna Therapeutics, Inc. Interleukin-2 fusion proteins and uses thereof
US11708401B2 (en) 2018-06-22 2023-07-25 Kite Pharma, Inc. Chimeric transmembrane proteins and uses thereof
US11759480B2 (en) 2017-02-28 2023-09-19 Endocyte, Inc. Compositions and methods for CAR T cell therapy
WO2023187031A1 (en) 2022-04-01 2023-10-05 Miltenyi Biotec B.V. & Co. KG A system for drug-inducible expression of a polynucleotide
WO2023217796A1 (en) 2022-05-10 2023-11-16 Miltenyi Biotec B.V. & Co. KG Compositions comprising il-15, il-15 receptor alpha and the intracellular signaling domain of cd2 for immune cell therapy
WO2023220459A1 (en) * 2022-05-13 2023-11-16 Northwestern University Active loading of cargo entity into lipid bilayer particles using dimerization domains
US11820819B2 (en) 2016-06-24 2023-11-21 Icell Gene Therapeutics Inc. Chimeric antigen receptors (CARs), compositions and methods thereof
WO2023222617A1 (en) 2022-05-16 2023-11-23 Miltenyi Biotec B.V. & Co. KG Endogenous signaling molecule activating chimeric antigen receptors and methods of generation thereof
EP4069728A4 (en) * 2019-12-03 2023-12-27 University of Massachusetts Compositions and methods for optogenetic immunotherapy
US11879011B2 (en) 2016-05-13 2024-01-23 Bioatla, Inc. Anti-ROR2 antibodies, antibody fragments, their immunoconjucates and uses thereof
US11896616B2 (en) 2017-03-27 2024-02-13 National University Of Singapore Stimulatory cell lines for ex vivo expansion and activation of natural killer cells
TWI835848B (en) 2018-09-07 2024-03-21 美商拜奧亞特拉公司 Conditionally active chimeric antigen receptors for modified t-cells
WO2024056809A1 (en) 2022-09-15 2024-03-21 Novartis Ag Treatment of autoimmune disorders using chimeric antigen receptor therapy
US11938153B2 (en) 2017-03-31 2024-03-26 The Board Of Trustees Of The Leland Stanford Junior University Methods of treating T cell exhaustion by inhibiting or modulating T cell receptor signaling
US11939389B2 (en) 2018-06-13 2024-03-26 Novartis Ag BCMA chimeric antigen receptors and uses thereof
EP4342488A1 (en) 2022-09-26 2024-03-27 Miltenyi Biotec B.V. & Co. KG Chimeric antigen receptor specific for folate receptor 1

Families Citing this family (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3279315A3 (en) * 2012-05-25 2018-02-28 Cellectis Use of pre t alpha or functional variant thereof for expanding tcr alpha deficient t cells
MX2016007958A (en) 2013-12-17 2016-08-03 Genentech Inc Anti-cd3 antibodies and methods of use.
US11697825B2 (en) 2014-12-12 2023-07-11 Voyager Therapeutics, Inc. Compositions and methods for the production of scAAV
US20160175359A1 (en) * 2014-12-15 2016-06-23 Bellicum Pharmaceuticals, Inc. Methods for controlled activation or elimination of therapeutic cells
US10195272B2 (en) 2015-03-02 2019-02-05 The Nemours Foundation Adoptive t-cell therapy using FcεRI-based chimeric antigen receptors for treating IgE-mediated allergic diseases
CN114634943A (en) 2015-05-18 2022-06-17 T细胞受体治疗公司 Compositions and methods for reprogramming TCRs using fusion proteins
EP3569244A1 (en) 2015-09-23 2019-11-20 CytoImmune Therapeutics, LLC Flt3 directed car cells for immunotherapy
US20190255107A1 (en) 2015-10-09 2019-08-22 The Brigham And Women's Hospital, Inc. Modulation of novel immune checkpoint targets
WO2017075451A1 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by detecting and targeting pou2af1
WO2017075465A1 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by detecting and targeting gata3
WO2017087708A1 (en) 2015-11-19 2017-05-26 The Brigham And Women's Hospital, Inc. Lymphocyte antigen cd5-like (cd5l)-interleukin 12b (p40) heterodimers in immunity
WO2017106638A1 (en) 2015-12-16 2017-06-22 Gritstone Oncology, Inc. Neoantigen identification, manufacture, and use
EA201891619A1 (en) 2016-01-11 2019-02-28 Те Борд Оф Трастиз Оф Те Лилэнд Стэнфорд Джуниор Юниверсити CHEMERAL PROTEINS AND METHODS OF REGULATING GENE EXPRESSION
EA201891614A1 (en) 2016-01-11 2019-02-28 Те Борд Оф Трастиз Оф Те Лилэнд Стэнфорд Джуниор Юниверсити CHEMICAL PROTEINS AND METHODS OF IMMUNOTHERAPY
CN105567640A (en) * 2016-01-27 2016-05-11 苏州佰通生物科技有限公司 Chimeric antigen receptor adipose-derived stem cell and preparation method thereof
WO2017172981A2 (en) * 2016-03-29 2017-10-05 University Of Southern California Chimeric antigen receptors targeting cancer
US11446398B2 (en) 2016-04-11 2022-09-20 Obsidian Therapeutics, Inc. Regulated biocircuit systems
US11384156B2 (en) 2016-07-25 2022-07-12 The Nemours Foundation Adoptive T-cell therapy using EMPD-specific chimeric antigen receptors for treating IgE-mediated allergic diseases
CA3032498A1 (en) 2016-08-02 2018-02-08 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
CA3000514A1 (en) * 2016-08-04 2018-02-08 Memorial Sloan-Kettering Cancer Center Cancer antigen targets and uses thereof
CN109789164B (en) * 2016-08-30 2023-04-28 亘喜生物科技(上海)有限公司 Chimeric antigen receptor with GITR intracellular domain as co-stimulatory domain
US20180064758A1 (en) * 2016-09-05 2018-03-08 Ucl Business Plc Chimeric antigen receptor
CN107793483B (en) * 2016-09-06 2019-08-23 伍志强 Chimeric antigen receptor and its gene and recombinant expression carrier, CARMSLN-NKT cell and its preparation method and application
WO2018049025A2 (en) 2016-09-07 2018-03-15 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses
CA3036745A1 (en) 2016-10-07 2018-04-12 TCR2 Therapeutics Inc. Compositions and methods for t-cell receptors reprogramming using fusion proteins
US20200016202A1 (en) 2016-10-07 2020-01-16 The Brigham And Women's Hospital, Inc. Modulation of novel immune checkpoint targets
CN109983027A (en) * 2016-10-20 2019-07-05 细胞基因公司 Based on CEREBLON can heterodimerization Chimeric antigen receptor
US11851491B2 (en) 2016-11-22 2023-12-26 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
CN107058315B (en) * 2016-12-08 2019-11-08 上海优卡迪生物医药科技有限公司 Strike the siRNA for subtracting people PD-1, recombinant expression CAR-T carrier and its construction method and application
EP3568406A4 (en) * 2017-01-10 2020-10-21 The General Hospital Corporation T cells expressing a chimeric antigen receptor
CN107573419A (en) * 2017-01-24 2018-01-12 深圳市体内生物医药科技有限公司 A kind of nucleic acid molecules for strengthening T cell antitumor activity
US11913075B2 (en) 2017-04-01 2024-02-27 The Broad Institute, Inc. Methods and compositions for detecting and modulating an immunotherapy resistance gene signature in cancer
US20200071773A1 (en) 2017-04-12 2020-03-05 Massachusetts Eye And Ear Infirmary Tumor signature for metastasis, compositions of matter methods of use thereof
EP3612210A4 (en) 2017-04-19 2021-01-27 Board Of Regents, The University Of Texas System Immune cells expressing engineered antigen receptors
CA3061676A1 (en) * 2017-05-12 2018-11-15 Cellectis Protease based switch chimeric antigen receptors for safer cell immunotherapy
CN107141356B (en) * 2017-06-07 2020-03-13 胜武(北京)生物科技有限公司 Photoinduced dimer type chimeric antigen receptor
EP3638218A4 (en) 2017-06-14 2021-06-09 The Broad Institute, Inc. Compositions and methods targeting complement component 3 for inhibiting tumor growth
CN107446937B (en) * 2017-09-05 2020-12-25 深圳华云生物技术有限公司 Chimeric antigen receptor, expression gene thereof, photo-controlled chimeric antigen receptor modified T cell and application thereof
CN109504697A (en) * 2017-09-15 2019-03-22 上海恒润达生生物科技有限公司 A kind of method and purposes of the Chimeric antigen receptor of targeting humanized mesothelin
WO2019075112A1 (en) 2017-10-10 2019-04-18 Gritstone Oncology, Inc. Neoantigen identification using hotspots
WO2019084055A1 (en) 2017-10-23 2019-05-02 Massachusetts Institute Of Technology Calling genetic variation from single-cell transcriptomes
WO2019094983A1 (en) 2017-11-13 2019-05-16 The Broad Institute, Inc. Methods and compositions for treating cancer by targeting the clec2d-klrb1 pathway
JP2021503897A (en) 2017-11-22 2021-02-15 グリットストーン オンコロジー インコーポレイテッド Reduced junction epitope presentation for nascent antigens
EP4083192A1 (en) 2017-12-22 2022-11-02 Fate Therapeutics, Inc. Enhanced immune effector cells and use thereof
PE20211116A1 (en) * 2018-02-08 2021-06-23 Genentech Inc BISPECIFIC ANTIGEN BINDING MOLECULES AND METHODS OF USE
WO2019157496A1 (en) * 2018-02-12 2019-08-15 University Of Florida Researchfoundation, Inc. Fviii chimeric antigen receptor tregs for tolerance induction in hemophilia a
US11813292B2 (en) * 2018-03-12 2023-11-14 Immunity Bio, Inc. Use of CD33CAR modified high affinity NK cells (t-haNK) to reduce myeloid-derived suppressor cells suppressor activity (or reduce negative impact on NK cell activity)
CN111954715A (en) 2018-03-29 2020-11-17 菲特治疗公司 Engineered immune effector cells and uses thereof
AU2019247830A1 (en) * 2018-04-06 2020-10-22 The Regents Of The University Of California Trans-antigen targeting in heterogeneous cancers and methods of use thereof
CN108410908B (en) * 2018-04-12 2021-08-03 吉林大学 Method for regulating cell pathway by using plant hormone GA and small molecular substance PAC
US11957695B2 (en) 2018-04-26 2024-04-16 The Broad Institute, Inc. Methods and compositions targeting glucocorticoid signaling for modulating immune responses
CN108715616A (en) * 2018-04-27 2018-10-30 上海恒润达生生物科技有限公司 The Chimeric antigen receptor method and purposes of targeting humanized mesothelin
WO2019232542A2 (en) 2018-06-01 2019-12-05 Massachusetts Institute Of Technology Methods and compositions for detecting and modulating microenvironment gene signatures from the csf of metastasis patients
CN110577604A (en) * 2018-06-07 2019-12-17 亘喜生物科技(上海)有限公司 Chimeric antigen receptor T cell carrying GITR costimulatory signal targeting EGFR
CN110669138A (en) * 2018-07-02 2020-01-10 中国药科大学 Double-chimeric antigen receptor, T cell, construction method and application thereof
EP3821012A4 (en) 2018-07-13 2022-04-20 The Regents of The University of California Retrotransposon-based delivery vehicle and methods of use thereof
US20210382068A1 (en) 2018-10-02 2021-12-09 Dana-Farber Cancer Institute, Inc. Hla single allele lines
GB201816839D0 (en) * 2018-10-16 2018-11-28 Glaxosmithkline Ip Dev Ltd Novel control switch
WO2020081730A2 (en) 2018-10-16 2020-04-23 Massachusetts Institute Of Technology Methods and compositions for modulating microenvironment
WO2020118305A1 (en) * 2018-12-07 2020-06-11 The Trustees Of Columbia University In The City Of New York Muc4 car-t cells for treating cancer
WO2020131586A2 (en) 2018-12-17 2020-06-25 The Broad Institute, Inc. Methods for identifying neoantigens
WO2020141106A1 (en) 2019-01-03 2020-07-09 Kemijski Institut Engineered externally regulated artificial transcription regulatory system based on engineered nfat
US11739156B2 (en) 2019-01-06 2023-08-29 The Broad Institute, Inc. Massachusetts Institute of Technology Methods and compositions for overcoming immunosuppression
US20220154282A1 (en) 2019-03-12 2022-05-19 The Broad Institute, Inc. Detection means, compositions and methods for modulating synovial sarcoma cells
EP3942023A1 (en) 2019-03-18 2022-01-26 The Broad Institute, Inc. Compositions and methods for modulating metabolic regulators of t cell pathogenicity
WO2020224605A1 (en) * 2019-05-07 2020-11-12 亘喜生物科技(上海)有限公司 Bcma-targeting engineered immune cell and use thereof
JP2022531911A (en) 2019-05-07 2022-07-12 グレイセル・バイオテクノロジーズ(シャンハイ)カンパニー・リミテッド Manipulated immune cells targeting BCMA and their use
US20220235340A1 (en) 2019-05-20 2022-07-28 The Broad Institute, Inc. Novel crispr-cas systems and uses thereof
WO2020243371A1 (en) 2019-05-28 2020-12-03 Massachusetts Institute Of Technology Methods and compositions for modulating immune responses
JP2022541320A (en) * 2019-07-24 2022-09-22 エウレカ セラピューティクス インコーポレイテッド Chimeric antigen receptor T cell and use thereof
US20220282333A1 (en) 2019-08-13 2022-09-08 The General Hospital Corporation Methods for predicting outcomes of checkpoint inhibition and treatment thereof
US20220298501A1 (en) 2019-08-30 2022-09-22 The Broad Institute, Inc. Crispr-associated mu transposase systems
US11793787B2 (en) 2019-10-07 2023-10-24 The Broad Institute, Inc. Methods and compositions for enhancing anti-tumor immunity by targeting steroidogenesis
US11844800B2 (en) 2019-10-30 2023-12-19 Massachusetts Institute Of Technology Methods and compositions for predicting and preventing relapse of acute lymphoblastic leukemia
JP2023512452A (en) * 2020-01-13 2023-03-27 ンカルタ・インコーポレイテッド BCMA-directed cellular immunotherapy compositions and methods
AU2021211713A1 (en) 2020-01-23 2022-08-25 The Children's Medical Center Corporation Stroma-free T cell differentiation from human pluripotent stem cells
JP2023517222A (en) * 2020-03-11 2023-04-24 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 Methods and compositions for gene delivery using engineered viral particles
CN113402612A (en) 2020-03-17 2021-09-17 西比曼生物科技(香港)有限公司 Combined chimeric antigen receptor targeting CD19 and CD20 and application thereof
IL297236A (en) 2020-04-17 2022-12-01 Hope City Flt3-targeted chimeric antigen receptor modified cells for treatment of flt3-positive malignancies
AU2021286655A1 (en) * 2020-06-12 2023-01-05 Nkarta, Inc. Genetically modified natural killer cells for CD70-directed cancer immunotherapy
WO2022173703A1 (en) * 2021-02-10 2022-08-18 The Regents Of The University Of California Immune receptors with synthetic co-stimulatory domains
GB202104030D0 (en) * 2021-03-23 2021-05-05 Autolus Ltd Signalling system
EP4313102A2 (en) * 2021-04-01 2024-02-07 A2 Biotherapeutics, Inc. Polypeptides targeting hla-a*11 and methods of use thereof
WO2023130462A1 (en) * 2022-01-10 2023-07-13 卡瑞济(北京)生命科技有限公司 CHIMERIC ANTIGEN RECEPTOR TARGETING IL13Rα2 AND USE THEREOF
WO2024077256A1 (en) 2022-10-07 2024-04-11 The General Hospital Corporation Methods and compositions for high-throughput discovery ofpeptide-mhc targeting binding proteins

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011119773A1 (en) * 2010-03-23 2011-09-29 Roeth Jeremiah F Vectors conditionally expressing therapeutic proteins, host cells comprising the vectors, and uses thereof
WO2012099973A2 (en) * 2011-01-18 2012-07-26 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer

Family Cites Families (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5677425A (en) 1987-09-04 1997-10-14 Celltech Therapeutics Limited Recombinant antibody
US5906936A (en) 1988-05-04 1999-05-25 Yeda Research And Development Co. Ltd. Endowing lymphocytes with antibody specificity
US5585362A (en) 1989-08-22 1996-12-17 The Regents Of The University Of Michigan Adenovirus vectors for gene therapy
CA2052928A1 (en) * 1990-10-15 1992-04-16 John J. Siekierka Genes encoding the human fk-506 binding protein and a.s. cerevisiae homolog, fkb1 and their expression
WO1992010591A1 (en) 1990-12-14 1992-06-25 Cell Genesys, Inc. Chimeric chains for receptor-associated signal transduction pathways
TW197439B (en) 1991-04-04 1993-01-01 Ueno Pharmaceutics Applic Res Co Ltd
WO1993003769A1 (en) 1991-08-20 1993-03-04 THE UNITED STATES OF AMERICA, represented by THE SECRETARY, DEPARTEMENT OF HEALTH AND HUMAN SERVICES Adenovirus mediated transfer of genes to the gastrointestinal tract
US5283253A (en) 1991-09-23 1994-02-01 Florida State University Furyl or thienyl carbonyl substituted taxanes and pharmaceutical compositions containing them
US5252479A (en) 1991-11-08 1993-10-12 Research Corporation Technologies, Inc. Safe vector for gene therapy
WO1993010076A1 (en) 1991-11-22 1993-05-27 The University Of Mississippi Synthesis and optical resolution of the taxol side chain and related compounds
US5200534A (en) 1992-03-13 1993-04-06 University Of Florida Process for the preparation of taxol and 10-deacetyltaxol
FR2688514A1 (en) 1992-03-16 1993-09-17 Centre Nat Rech Scient Defective recombinant adenoviruses expressing cytokines and antitumour drugs containing them
IL104570A0 (en) 1992-03-18 1993-05-13 Yeda Res & Dev Chimeric genes and cells transformed therewith
EP0642586A4 (en) 1992-05-21 1995-11-29 Penn State Res Found CULTURED -i(TAXUS) TISSUES AS A SOURCE OF TAXOL, RELATED TAXANES AND OTHER NOVEL ANTI-TUMOR/ANTI-VIRAL COMPOUNDS.
US5274137A (en) 1992-06-23 1993-12-28 Nicolaou K C Intermediates for preparation of taxols
US5294637A (en) 1992-07-01 1994-03-15 Bristol-Myers Squibb Company Fluoro taxols
US5202448A (en) 1992-08-14 1993-04-13 Napro Biotherapeutics, Inc. Processes of converting taxanes into baccatin III
CA2100808A1 (en) 1992-10-01 1994-04-02 Vittorio Farina Deoxy paclitaxels
FR2696464B1 (en) 1992-10-05 1994-11-10 Rhone Poulenc Rorer Sa New esterification process for baccatin III and 10-deacetyl baccatin III.
FR2696461B1 (en) 1992-10-05 1994-11-10 Rhone Poulenc Rorer Sa New derivatives of taxol analogs, their preparation and compositions containing them.
FR2696463B1 (en) 1992-10-05 1994-11-25 Rhone Poulenc Rorer Sa Process for obtaining 10-deacetyl baccatin III.
FR2696462B1 (en) 1992-10-05 1994-11-25 Rhone Poulenc Rorer Sa Process for obtaining 10-deacetyl baccatin III.
FR2696458B1 (en) 1992-10-05 1994-11-10 Rhone Poulenc Rorer Sa Process for the preparation of taxane derivatives.
EP1024198A3 (en) 1992-12-03 2002-05-29 Genzyme Corporation Pseudo-adenoviral vectors for the gene therapy of haemophiliae
US5279949A (en) 1992-12-07 1994-01-18 Board Of Trustees Operating Michigan State University Process for the isolation and purification of taxol and taxanes from Taxus spp
US5834266A (en) 1993-02-12 1998-11-10 President & Fellows Of Harvard College Regulated apoptosis
US5869337A (en) 1993-02-12 1999-02-09 President And Fellows Of Harvard College Regulated transcription of targeted genes and other biological events
US5830462A (en) 1993-02-12 1998-11-03 President & Fellows Of Harvard College Regulated transcription of targeted genes and other biological events
JP3824633B2 (en) * 1993-02-12 2006-09-20 ザ・ボード・オブ・トラスティーズ・オブ・ザ・リランド・スタンフォード・ジュニアー・ユニバーシティ Regulatory transcription of target genes and other biological consequences
US5310903A (en) 1993-03-05 1994-05-10 Merck & Co., Inc. Imidazolidyl rapamycin derivatives
AU687829B2 (en) 1993-06-24 1998-03-05 Advec, Inc. Adenovirus vectors for gene therapy
US5824701A (en) 1993-10-20 1998-10-20 Enzon, Inc. Taxane-based prodrugs
SK283703B6 (en) 1993-10-25 2003-12-02 Canji, Inc. Recombinant adenoviral vector and methods of use
US5415869A (en) 1993-11-12 1995-05-16 The Research Foundation Of State University Of New York Taxol formulation
US5527907A (en) 1993-11-19 1996-06-18 Abbott Laboratories Macrolide immunomodulators
US5525610A (en) 1994-03-31 1996-06-11 American Home Products Corporation 42-Epi-rapamycin and pharmaceutical compositions thereof
US5362718A (en) 1994-04-18 1994-11-08 American Home Products Corporation Rapamycin hydroxyesters
US6133456A (en) 1994-08-18 2000-10-17 Ariad Gene Therapeutics, Inc. Synthetic multimerizing agents
US20030036654A1 (en) 1994-08-18 2003-02-20 Holt Dennis A. Synthetic multimerizing agents
US6150527A (en) 1994-08-18 2000-11-21 Ariad Pharmaceuticals, Inc. Synthetic multimerizing agents
CZ296981B6 (en) 1994-12-09 2006-08-16 Imperial College Innovations Limited Method for identifying microorganisms, method for identifying a gene, microorganism per se, vaccine and process for its preparation, method of obtaining a mutant microorganism, process for preparing a pharmaceutical composition, method for identifyin
US5712149A (en) 1995-02-03 1998-01-27 Cell Genesys, Inc. Chimeric receptor molecules for delivery of co-stimulatory signals
US6187757B1 (en) 1995-06-07 2001-02-13 Ariad Pharmaceuticals, Inc. Regulation of biological events using novel compounds
WO1996041865A1 (en) 1995-06-07 1996-12-27 Ariad Gene Therapeutics, Inc. Rapamcycin-based regulation of biological events
GB9526131D0 (en) 1995-12-21 1996-02-21 Celltech Therapeutics Ltd Recombinant chimeric receptors
WO1998008833A1 (en) 1996-08-26 1998-03-05 Bristol-Myers Squibb Company Sulfenamide taxane derivatives
EP0961619A4 (en) 1996-09-27 2001-09-26 Bristol Myers Squibb Co Hydrolyzable prodrugs for delivery of anticancer drugs to metastatic cells
AU4966597A (en) 1996-11-19 1998-06-10 Daiichi Pharmaceutical Co., Ltd. Taxol derivatives
US5977386A (en) 1996-12-24 1999-11-02 Bristol-Myers Squibb Company 6-thio-substituted paclitaxels
IL133585A0 (en) 1997-06-20 2001-04-30 Baker Norton Pharma Soluble prodrugs of paclitaxel
US7288665B1 (en) 1997-08-18 2007-10-30 Florida State University Process for selective derivatization of taxanes
JPH1192468A (en) 1997-09-17 1999-04-06 Yakult Honsha Co Ltd New taxane derivative
AU741433B2 (en) 1997-10-08 2001-11-29 Bio Research Corporation Of Yokohama Taxoid derivatives and process for producing the same
JP2002508971A (en) 1998-01-15 2002-03-26 アリアド・ジーン・セラピューティクス・インコーポレーテッド Regulation of biological events using multimeric chimeric proteins
WO2000023573A2 (en) 1998-10-20 2000-04-27 City Of Hope Cd20-specific redirected t cells and their use in cellular immunotherapy of cd20+ malignancies
ATE264863T1 (en) 1999-08-24 2004-05-15 Ariad Gene Therapeutics Inc 28-EPIRAPALOGUE
GB9925854D0 (en) * 1999-11-01 1999-12-29 Celltech Therapeutics Ltd Biological products
US7514239B2 (en) 2000-03-28 2009-04-07 Amgen Inc. Nucleic acid molecules encoding beta-like glycoprotein hormone polypeptides and heterodimers thereof
US6916846B2 (en) 2000-05-12 2005-07-12 Merck & Co. Inc. Coumermycin analogs as chemical dimerizers of chimeric proteins
GB0025307D0 (en) 2000-10-16 2000-11-29 Celltech Chiroscience Ltd Biological products
ATE338124T1 (en) 2000-11-07 2006-09-15 Hope City CD19-SPECIFIC TARGETED IMMUNE CELLS
GB0105402D0 (en) 2001-03-05 2001-04-18 Inpharmatica Ltd Novel proteins
GB0105924D0 (en) 2001-03-09 2001-04-25 Microscience Ltd Promoter
US8492122B2 (en) 2002-03-11 2013-07-23 The Johns Hopkins University Molecular switches and methods for making and using the same
US7446190B2 (en) 2002-05-28 2008-11-04 Sloan-Kettering Institute For Cancer Research Nucleic acids encoding chimeric T cell receptors
EP1641927B1 (en) 2003-02-18 2015-07-08 Baylor College of Medicine Induced activation in dendritic cells
US7435596B2 (en) 2004-11-04 2008-10-14 St. Jude Children's Research Hospital, Inc. Modified cell line and method for expansion of NK cell
US20050113564A1 (en) 2003-11-05 2005-05-26 St. Jude Children's Research Hospital Chimeric receptors with 4-1BB stimulatory signaling domain
WO2006036445A2 (en) 2004-09-24 2006-04-06 Trustees Of Dartmouth College Chimeric nk receptor and methods for treating cancer
US8236925B1 (en) 2005-08-26 2012-08-07 University Of Minnesota Protein nanorings
AT503861B1 (en) 2006-07-05 2008-06-15 F Star Biotech Forsch & Entw METHOD FOR MANIPULATING T-CELL RECEPTORS
WO2011053825A2 (en) * 2009-10-30 2011-05-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Compositions and methods for the treatment or prevention of mitochondrial diseases
JP5856073B2 (en) 2009-12-29 2016-02-09 エマージェント プロダクト デベロップメント シアトル, エルエルシー RON binding construct and method of use thereof
WO2011130624A2 (en) 2010-04-16 2011-10-20 Immune Disease Institute, Inc. Sustained polypeptide expression from synthetic, modified rnas and uses thereof
WO2011146862A1 (en) 2010-05-21 2011-11-24 Bellicum Pharmaceuticals, Inc. Methods for inducing selective apoptosis
CN103201293B (en) 2010-09-08 2016-04-27 哈洛齐梅公司 The method of assessment and qualification or development condition active therapeutic protein
FR2968013A1 (en) * 2010-11-29 2012-06-01 Cis Bio Int METHOD OF CONDITIONALLY RETAINING A PROTEIN OF INTEREST IN THE ENDOPLASMIC RETICULUM
BR122021026173B1 (en) * 2010-12-09 2023-12-05 The Trustees Of The University Of Pennsylvania PHARMACEUTICAL COMPOSITION
EP3279315A3 (en) 2012-05-25 2018-02-28 Cellectis Use of pre t alpha or functional variant thereof for expanding tcr alpha deficient t cells
EP2893004B1 (en) 2012-09-04 2018-10-24 Cellectis Multi-chain chimeric antigen receptor and uses thereof
WO2014055657A1 (en) * 2012-10-05 2014-04-10 The Trustees Of The University Of Pennsylvania Use of a trans-signaling approach in chimeric antigen receptors
LT2956175T (en) 2013-02-15 2017-12-11 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof
US9944690B2 (en) 2013-03-14 2018-04-17 Bellicum Pharmaceuticals, Inc. Methods for controlling T cell proliferation
EP3623380A1 (en) 2013-03-15 2020-03-18 Michael C. Milone Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
ES2741308T3 (en) * 2013-10-15 2020-02-10 Scripps Research Inst T-cell switches with chimeric antigen receptors and uses thereof
JP6793902B2 (en) 2013-12-20 2020-12-02 ノバルティス アーゲー Adjustable chimeric antigen receptor
CA2937750A1 (en) 2014-02-14 2015-08-20 Bellicum Pharmaceuticals, Inc. Methods for activating t cells using an inducible chimeric polypeptide
WO2015142661A1 (en) * 2014-03-15 2015-09-24 Novartis Ag Regulatable chimeric antigen receptor
GB201405845D0 (en) 2014-04-01 2014-05-14 Ucl Business Plc Signalling system
US10472613B2 (en) 2014-10-07 2019-11-12 Cellectis Method for modulating car-induced immune cells activity
US20210206826A1 (en) 2015-11-19 2021-07-08 The Regents Of The University Of California Conditionally repressible immune cell receptors and methods of use thereof
EP3399991A4 (en) 2016-01-08 2019-08-07 The Regents of The University of California Conditionally active heterodimeric polypeptides and methods of use thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011119773A1 (en) * 2010-03-23 2011-09-29 Roeth Jeremiah F Vectors conditionally expressing therapeutic proteins, host cells comprising the vectors, and uses thereof
WO2012099973A2 (en) * 2011-01-18 2012-07-26 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
DEROSE ET AL.: "Manipulating signaling at will: chemically-inducible dimerization (CID) techniques resolve problems in cell biology.", PFLUGERS ARCH, vol. 465, no. 3, March 2013 (2013-03-01), pages 409 - 417, XP055221913 *
DI STASI ET AL.: "Inducible apoptosis as a safety switch for adoptive cell therapy.", N ENGL J MED, vol. 365, no. 18, 3 November 2011 (2011-11-03), pages 1673 - 83, XP055181696 *
FEGAN ET AL.: "Chemically controlled protein assembly: techniques and applications.", CHEM REV, vol. 110, no. 6, 9 June 2010 (2010-06-09), pages 3315 - 36, XP055221903 *
GRAEF ET AL.: "Proximity and orientation underlie signaling by the non-receptor tyrosine kinase ZAP70.", EMBO J, vol. 16, no. 18, 15 September 1997 (1997-09-15), pages 5618 - 2628, XP055263591 *
KLOSS ET AL.: "Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells", NAT BIOTECHNOL, vol. 31, no. 1, 16 December 2012 (2012-12-16), pages 71 - 75, XP055130697 *
PORTER ET AL.: "Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia.", N ENGL J MED, vol. 365, no. 8, 25 August 2011 (2011-08-25), pages 725 - 733, XP055052475 *

Cited By (322)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11873512B2 (en) 2013-01-28 2024-01-16 St. Jude Children's Research Hospital, Inc. Chimeric receptor with NKG2D specificity for use in cell therapy against cancer and infectious disease
US10538739B2 (en) 2013-01-28 2020-01-21 St. Jude Children's Research Hospital, Inc. Chimeric receptor with NKG2D specificity for use in cell therapy against cancer and infectious disease
US10774309B2 (en) 2013-01-28 2020-09-15 St. Jude Children's Research Hospital, Inc. Natural killer cell immunotherapy for treating cancer
US10801012B2 (en) 2013-01-28 2020-10-13 St. Jude Children's Research Hospital, Inc. Chimeric receptor with NKG2D specificity for use in cell therapy against cancer and infectious disease
US10836999B2 (en) 2013-01-28 2020-11-17 St. Jude Children's Research Hospital, Inc. Chimeric receptor with NKG2D specificity for use in cell therapy against cancer and infectious disease
US10829737B2 (en) 2013-01-28 2020-11-10 St. Jude Children's Research Hospital, Inc. Chimeric receptor with NKG2D specificity for use in cell therapy against cancer and infectious disease
US9587020B2 (en) 2013-02-15 2017-03-07 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof
US11478510B2 (en) 2013-02-15 2022-10-25 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof
US10105391B2 (en) 2013-02-15 2018-10-23 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof
US10888581B2 (en) 2013-02-15 2021-01-12 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof
US9821012B2 (en) 2013-02-15 2017-11-21 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof
US10632152B2 (en) 2013-02-15 2020-04-28 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof
US9944690B2 (en) 2013-03-14 2018-04-17 Bellicum Pharmaceuticals, Inc. Methods for controlling T cell proliferation
US11919946B2 (en) 2013-03-15 2024-03-05 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US9745368B2 (en) 2013-03-15 2017-08-29 The Trustees Of The University Of Pennsylvania Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2014145252A2 (en) * 2013-03-15 2014-09-18 Milone Michael C Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
EP3623380A1 (en) * 2013-03-15 2020-03-18 Michael C. Milone Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2014145252A3 (en) * 2013-03-15 2014-12-11 Milone Michael C Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US10640553B2 (en) 2013-03-15 2020-05-05 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US10457731B2 (en) 2013-07-29 2019-10-29 Bluebird Bio, Inc. Multipartite signaling proteins and uses thereof
US10196444B2 (en) 2013-07-29 2019-02-05 Bluebird Bio, Inc. Multipartite signaling proteins and uses thereof
WO2015017214A1 (en) 2013-07-29 2015-02-05 Bluebird Bio, Inc. Multipartite signaling proteins and uses thereof
JP2016531567A (en) * 2013-07-29 2016-10-13 ブルーバード バイオ, インコーポレイテッド Multipart signaling protein and uses thereof
JP2020178690A (en) * 2013-07-29 2020-11-05 ブルーバード バイオ, インコーポレイテッド Multipartite signaling proteins and uses thereof
AU2014296626B2 (en) * 2013-07-29 2019-03-07 2Seventy Bio, Inc. Multipartite signaling proteins and uses thereof
US11530265B2 (en) 2013-07-29 2022-12-20 2Seventy Bio, Inc. Multipartite signaling proteins and uses thereof
EP3027204A4 (en) * 2013-07-29 2017-03-15 Bluebird Bio, Inc. Multipartite signaling proteins and uses thereof
JP2019146598A (en) * 2013-07-29 2019-09-05 ブルーバード バイオ, インコーポレイテッド Multipartite signaling proteins and uses thereof
US10428142B2 (en) 2013-07-29 2019-10-01 Bluebird Bio, Inc. Multipartite signaling proteins and uses thereof
US11680090B2 (en) 2013-09-24 2023-06-20 Medicenna Therapeutics, Inc. Interleukin-2 fusion proteins and uses thereof
EP3087101A4 (en) * 2013-12-20 2017-12-06 Novartis AG Regulatable chimeric antigen receptor
US11578130B2 (en) 2013-12-20 2023-02-14 Novartis Ag Regulatable chimeric antigen receptor
US10287354B2 (en) 2013-12-20 2019-05-14 Novartis Ag Regulatable chimeric antigen receptor
WO2015090229A1 (en) 2013-12-20 2015-06-25 Novartis Ag Regulatable chimeric antigen receptor
US10526406B2 (en) 2014-01-14 2020-01-07 Cellectis Chimeric antigen receptor using antigen recognition domains derived from cartilaginous fish
JP2017507917A (en) * 2014-01-14 2017-03-23 セレクティスCellectis Chimeric antigen receptor using antigen recognition domain from cartilaginous fish
US10934346B2 (en) 2014-02-14 2021-03-02 Bellicum Pharmaceuticals, Inc. Modified T cell comprising a polynucleotide encoding an inducible stimulating molecule comprising MyD88, CD40 and FKBP12
EP3119425A4 (en) * 2014-03-15 2017-11-22 Novartis AG Regulatable chimeric antigen receptor
EP3811970A1 (en) * 2014-03-15 2021-04-28 Novartis AG Regulatable chimeric antigen receptor
WO2015142661A1 (en) 2014-03-15 2015-09-24 Novartis Ag Regulatable chimeric antigen receptor
WO2015150771A1 (en) * 2014-04-01 2015-10-08 Ucl Business Plc Chimeric antigen receptor (car) signalling system
US10588967B2 (en) 2014-04-01 2020-03-17 Ucl Business Ltd Chimeric antigen receptor (CAR) signalling system
EP3888674A1 (en) 2014-04-07 2021-10-06 Novartis AG Treatment of cancer using anti-cd19 chimeric antigen receptor
WO2015157252A1 (en) 2014-04-07 2015-10-15 BROGDON, Jennifer Treatment of cancer using anti-cd19 chimeric antigen receptor
US11384131B2 (en) 2014-04-24 2022-07-12 The Board Of Trustees Of The Leland Stanford Junior University Superagonists, partial agonists and antagonists of interleukin-2
US11560548B2 (en) 2014-05-15 2023-01-24 National University Of Singapore Immune cells expressing membrane-bound interleukin 15 (mbIL15) and uses thereof
US10428305B2 (en) 2014-05-15 2019-10-01 National University Of Singapore Modified natural killer cells that express IL15 and uses thereof
US10774311B2 (en) 2014-05-15 2020-09-15 National University Of Singapore Natural killer cells modified to express membrane-bound interleukin 15 and uses thereof
EP4166148A1 (en) * 2014-06-06 2023-04-19 Memorial Sloan-Kettering Cancer Center Mesothelin-targeted chimeric antigen receptors and uses thereof
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
EP4205749A1 (en) 2014-07-31 2023-07-05 Novartis AG Subset-optimized chimeric antigen receptor-containing cells
EP3660042A1 (en) 2014-07-31 2020-06-03 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
JP2021137013A (en) * 2014-08-28 2021-09-16 バイオアトラ、エルエルシー Conditionally active chimeric antigen receptors for modified t-cells
US11584927B2 (en) 2014-08-28 2023-02-21 Bioatla, Inc. Conditionally active chimeric antigen receptors for modified T-cells
WO2016033331A1 (en) * 2014-08-28 2016-03-03 Bioatla, Llc Conditionally active chimeric antigen receptors for modified t-cells
WO2017034615A1 (en) * 2014-08-28 2017-03-02 Bioatla, Llc Conditionally active chimeric antigen receptors for modified t-cells
RU2764074C2 (en) * 2014-08-28 2022-01-13 Байоатла, Ллк Conditionally active chimeric antigen receptors for modified t-cells
RU2759957C2 (en) * 2014-08-28 2021-11-19 Байоатла, Ллк Conditionally active chimeric antigenic receptors for modified t cells
US11111288B2 (en) 2014-08-28 2021-09-07 Bioatla, Inc. Conditionally active chimeric antigen receptors for modified t-cells
JP7286267B2 (en) 2014-08-28 2023-06-05 バイオアトラ インコーポレイテッド Conditionally active chimeric antigen receptor for modified T cells
CN107074975A (en) * 2014-08-28 2017-08-18 生物蛋白有限公司 Condition active chimeric antigen receptor for the T cell of modification
JP7405444B2 (en) 2014-08-28 2023-12-26 バイオアトラ インコーポレイテッド Conditionally activated chimeric antigen receptor for modified T cells
CN107922473A (en) * 2014-08-28 2018-04-17 生物蛋白有限公司 Condition active chimeric antigen receptor for the T cell of modification
US10654927B2 (en) 2014-08-29 2020-05-19 Ucl Business Ltd Signalling system
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
EP3981417A1 (en) * 2014-08-29 2022-04-13 Autolus Limited Signalling system
AU2015308243B2 (en) * 2014-08-29 2020-01-30 Autolus Limited Signalling system
CN106573989B (en) * 2014-08-29 2022-02-01 奥托路斯有限公司 Signal conduction system
KR20170045258A (en) * 2014-08-29 2017-04-26 유씨엘 비즈니스 피엘씨 Signalling system
JP2017526361A (en) * 2014-08-29 2017-09-14 ユーシーエル ビジネス ピーエルシー Signal transduction system
RU2731638C2 (en) * 2014-08-29 2020-09-07 ЮСиЭл БИЗНЕС ЛТД Signal system
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
KR102222615B1 (en) * 2014-08-29 2021-03-05 유씨엘 비즈니스 리미티드 Signalling system
WO2016030691A1 (en) * 2014-08-29 2016-03-03 Ucl Business Plc Signalling system
CN106573989A (en) * 2014-08-29 2017-04-19 Ucl商务股份有限公司 Signalling system
US10918705B2 (en) 2014-09-02 2021-02-16 Bellicum Pharmaceutics, Inc. Costimulation of chimeric antigen receptors by MYD88 and CD40 polypeptides
US10888608B2 (en) 2014-09-02 2021-01-12 Bellicum Pharmaceuticals, Inc. Costimulation of chimeric antigen receptors by MyD88 and CD40 polypeptides
KR102590396B1 (en) 2014-09-17 2023-10-19 노파르티스 아게 Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
JP2017534261A (en) * 2014-09-17 2017-11-24 ノバルティス アーゲー Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2016044605A1 (en) * 2014-09-17 2016-03-24 Beatty, Gregory Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
CN107580628A (en) * 2014-09-17 2018-01-12 诺华股份有限公司 The targeting cytotoxic cell with Chimerical receptor for adoptive immunotherapy
KR20170056622A (en) * 2014-09-17 2017-05-23 노파르티스 아게 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
WO2016054013A1 (en) * 2014-10-03 2016-04-07 Yale University Innate immune system modification for anticancer therapy
CN107109368A (en) * 2014-10-07 2017-08-29 塞勒克提斯公司 For the method for the immunologic cellular activity for adjusting CAR inductions
JP2017531430A (en) * 2014-10-07 2017-10-26 セレクティスCellectis Method for modulating the activity of immune cells induced by CAR
US10472613B2 (en) * 2014-10-07 2019-11-12 Cellectis Method for modulating car-induced immune cells activity
WO2016057705A1 (en) 2014-10-08 2016-04-14 Novartis Ag Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
EP3025719A1 (en) 2014-11-26 2016-06-01 Miltenyi Biotec GmbH Combination immunotherapy of antigen-recognizing receptors and hematopoietic cells for the treatment of diseases
US20180072789A1 (en) * 2014-12-02 2018-03-15 Roger Williams Hospital Methods and compositions for treating cancer
US11820806B2 (en) 2014-12-05 2023-11-21 Memorial Sloan-Kettering Cancer Center Chimeric antigen receptors targeting G-protein coupled receptor and uses thereof
US10590196B2 (en) 2014-12-05 2020-03-17 Memorial Sloan-Kettering Cancer Center Antibodies targeting G-protein coupled receptor and methods of use
US11566071B2 (en) 2014-12-05 2023-01-31 Memorial Sloan Kettering Cancer Center Nucleic acid molecules encoding anti-GPRC5D antibodies
US11866478B2 (en) 2014-12-05 2024-01-09 Memorial Sloan-Kettering Cancer Center Nucleic acid molecules encoding chimeric antigen receptors targeting G-protein coupled receptor
US10633426B2 (en) 2014-12-05 2020-04-28 Memorial Sloan Kettering Cancer Center Chimeric antigen receptors targeting G-protein coupled receptor and uses thereof
WO2016090312A1 (en) * 2014-12-05 2016-06-09 Memorial Sloan-Kettering Cancer Center Chimeric antigen receptors targeting g-protein coupled receptor and uses thereof
US10906956B2 (en) 2014-12-05 2021-02-02 Memorial Sloan Kettering Cancer Center Methods of treatments using chimeric antigen receptors targeting G-protein coupled receptor
EP3851450A1 (en) * 2014-12-19 2021-07-21 ETH Zürich Chimeric antigen receptors and methods of use
JP2017537642A (en) * 2014-12-19 2017-12-21 イーティーエッチ チューリッヒ Chimeric antigen receptor and method of use thereof
US11939572B2 (en) 2014-12-19 2024-03-26 ETH Zürich Chimeric antigen receptors and methods of use
WO2016097334A1 (en) * 2014-12-19 2016-06-23 ETH Zürich Chimeric antigen receptors and methods of use
WO2016098078A3 (en) * 2014-12-19 2016-10-27 Novartis Ag Dimerization switches and uses thereof
US10865408B2 (en) 2014-12-19 2020-12-15 Eth Zurich Chimeric antigen receptors and methods of use
CN107995926A (en) * 2014-12-19 2018-05-04 苏黎世联邦理工学院 Chimeric antigen receptor and application method
CN107995926B (en) * 2014-12-19 2021-07-06 苏黎世联邦理工学院 Chimeric antigen receptors and methods of use
EP3556768A1 (en) 2014-12-24 2019-10-23 Aadigen, LLC Peptides and nanoparticles for intracellular delivery of molecules
US11713336B2 (en) 2014-12-24 2023-08-01 Aadigen, Llc Peptides and nanoparticles for intracellular delivery of molecules
US10745440B2 (en) 2014-12-24 2020-08-18 Aadigen, Llc Peptides and nanoparticles for intracellular delivery of molecules
EP4036109A2 (en) 2014-12-29 2022-08-03 Novartis AG Methods of making chimeric antigen receptor-expressing cells
US10604570B2 (en) 2015-02-05 2020-03-31 Ucl Business Ltd Chimeric antigen receptor signalling system comprising heterodimerization domains
WO2016124930A1 (en) * 2015-02-05 2016-08-11 Ucl Business Plc Signalling system
US11103532B2 (en) 2015-02-24 2021-08-31 Autolus Limited Chimeric protein
US10478457B2 (en) 2015-02-24 2019-11-19 Ucl Business Ltd Chimeric protein
US10273280B2 (en) 2015-02-27 2019-04-30 Icell Gene Therapeutics Llc Chimeric antigen receptors (CARs), targeting hematologic malignancies, compositions and methods of use thereof
RU2752880C2 (en) * 2015-03-23 2021-08-11 Отолус Лимитед Chimeric antigen receptor
US11058722B2 (en) 2015-03-23 2021-07-13 Autolus Limited Chimeric antigen receptor comprising a cartilage-oligomeric matrix protein (comp) coiled-coil spacer domain
CN107406518B (en) * 2015-03-23 2022-02-01 奥托路斯有限公司 Chimeric antigen receptors
CN107406518A (en) * 2015-03-23 2017-11-28 Ucl商务股份有限公司 Chimeric antigen receptor
WO2016151315A1 (en) * 2015-03-23 2016-09-29 Ucl Business Plc Chimeric antigen receptor
JP2020048588A (en) * 2015-03-23 2020-04-02 ユーシーエル ビジネス リミテッド Chimeric antigen receptor
JP2018510639A (en) * 2015-03-23 2018-04-19 ユーシーエル ビジネス ピーエルシー Chimeric antigen receptor
JP2018515069A (en) * 2015-04-02 2018-06-14 メモリアル スローン ケタリング キャンサー センター TNFRSF14 / HVEM protein and method of use thereof
US11261232B2 (en) 2015-04-02 2022-03-01 Memorial Sloan Kettering Cancer Center TNFRSF14 / HVEM proteins and methods of use thereof
WO2016161415A3 (en) * 2015-04-02 2016-12-01 Memorial Sloan Kettering Cancer Center Tnfrsf14/ hvem proteins and methods of use thereof
EP4056588A1 (en) 2015-04-08 2022-09-14 Novartis AG Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car)- expressing cell
WO2016164731A2 (en) 2015-04-08 2016-10-13 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
WO2016168773A3 (en) * 2015-04-15 2016-12-01 The California Institute For Biomedical Research Optimized pne-based chimeric receptor t cell switches and uses thereof
EP4234685A2 (en) 2015-04-17 2023-08-30 Novartis AG Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
US11896614B2 (en) 2015-04-17 2024-02-13 Novartis Ag Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
WO2016168595A1 (en) 2015-04-17 2016-10-20 Barrett David Maxwell Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
WO2016174407A1 (en) * 2015-04-27 2016-11-03 Ucl Business Plc Nucleic acid construct for expressing more than one chimeric antigen receptor
WO2016174405A1 (en) * 2015-04-27 2016-11-03 Ucl Business Plc Nucleic acid construct for expressing more than one chimeric antigen receptor
WO2016174406A1 (en) * 2015-04-27 2016-11-03 Ucl Business Plc Nucleic acid construct for expressing more than one chimeric antigen receptor
EP3447075A2 (en) 2015-05-15 2019-02-27 The General Hospital Corporation Antagonistic anti-tumor necrosis factor receptor superfamily antibodies
EP4292664A2 (en) 2015-05-15 2023-12-20 The General Hospital Corporation Antagonistic anti-tumor necrosis factor receptor superfamily antibodies
US11655452B2 (en) 2015-06-25 2023-05-23 Icell Gene Therapeutics Inc. Chimeric antigen receptors (CARs), compositions and methods of use thereof
WO2016210293A1 (en) * 2015-06-25 2016-12-29 Icell Gene Therapeutics Llc CHIMERIC ANTIGEN RECEPTORS (CARs), COMPOSITIONS AND METHODS OF USE THEREOF
US11173179B2 (en) 2015-06-25 2021-11-16 Icell Gene Therapeutics Llc Chimeric antigen receptor (CAR) targeting multiple antigens, compositions and methods of use thereof
JP7146632B2 (en) 2015-07-21 2022-10-04 ノバルティス アーゲー Methods of Improving Immune Cell Efficacy and Expansion
JP2018519842A (en) * 2015-07-21 2018-07-26 ノバルティス アーゲー Methods for improving the effectiveness and expansion of immune cells
WO2017015427A1 (en) 2015-07-21 2017-01-26 Novartis Ag Methods for improving the efficacy and expansion of immune cells
US11802143B2 (en) 2015-07-31 2023-10-31 King's College London Therapeutic agents
US10703794B2 (en) 2015-07-31 2020-07-07 King's College London Therapeutic agents
US10865231B2 (en) 2015-07-31 2020-12-15 King's College London Therapeutic agents
US10899818B2 (en) 2015-07-31 2021-01-26 King's College London Therapeutic agents
US10975148B2 (en) 2015-08-05 2021-04-13 CellabMED Inc. Chimeric antigen receptors, and T cells in which chimeric antigen receptor is expressed
JP2018522904A (en) * 2015-08-05 2018-08-16 株式会社柳英製薬Yoo Young Pharm Co., Ltd. Chimeric antigen receptor and T cell expressing chimeric antigen receptor
US11293023B2 (en) 2015-08-06 2022-04-05 Dana-Farber Cancer Institute, Inc. Tunable endogenous protein degradation
US11046954B2 (en) 2015-08-06 2021-06-29 Dana-Farber Cancer Institute, Inc. Targeted protein degradation to attenuate adoptive T-cell therapy associated adverse inflammatory responses
US11667691B2 (en) * 2015-08-07 2023-06-06 Novartis Ag Treatment of cancer using chimeric CD3 receptor proteins
US10988542B2 (en) 2015-08-24 2021-04-27 Cellectis Chimeric antigen receptors with integrated controllable functions
CN108138362B (en) * 2015-09-01 2022-03-29 加利福尼亚大学董事会 Modular polypeptide libraries and methods of making and using the same
CN108138362A (en) * 2015-09-01 2018-06-08 加利福尼亚大学董事会 Modularization polypeptide libraries and its preparation and application
EP4282979A3 (en) * 2015-09-01 2024-02-28 The Regents of the University of California Modular polypeptide libraries and methods of making and using same
EP3344803A4 (en) * 2015-09-01 2019-06-12 The Regents of The University of California Modular polypeptide libraries and methods of making and using same
EP3954772A1 (en) 2015-09-01 2022-02-16 The Regents of The University of California Modular polypeptide libraries and methods of making and using same
JP2021119780A (en) * 2015-09-01 2021-08-19 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The Regents Of The University Of California Module polypeptide library, method for producing the same, and use of the same
JP2018525992A (en) * 2015-09-01 2018-09-13 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 Modular polypeptide library and method for producing the same and use thereof
JP7397821B2 (en) 2015-09-01 2023-12-13 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 Modular polypeptide library and its production method and use thereof
US11560561B2 (en) 2015-09-01 2023-01-24 The Regents Of The University Of California Modular polypeptide libraries and methods of making and using same
EP4282979A2 (en) 2015-09-01 2023-11-29 The Regents of the University of California Modular polypeptide libraries and methods of making and using same
WO2017040930A2 (en) 2015-09-03 2017-03-09 The Trustees Of The University Of Pennsylvania Biomarkers predictive of cytokine release syndrome
WO2017087723A1 (en) * 2015-11-19 2017-05-26 The Regents Of The University Of California Conditionally repressible immune cell receptors and methods of use thereof
EP3377523A4 (en) * 2015-11-19 2019-06-19 The Regents of The University of California Conditionally repressible immune cell receptors and methods of use thereof
JP2018538277A (en) * 2015-11-23 2018-12-27 トラスティーズ オブ ボストン ユニバーシティ Methods and compositions for chimeric antigen receptors
WO2017091546A1 (en) * 2015-11-23 2017-06-01 Trustees Of Boston University Methods and compositions relating to chimeric antigen receptors
CN108495927A (en) * 2015-11-23 2018-09-04 波士顿大学董事会 The relevant method and composition of Chimeric antigen receptor
US11530252B2 (en) 2015-11-23 2022-12-20 Trustees Of Boston University Methods and compositions relating to chimeric antigen receptors
EP3184548A1 (en) 2015-12-23 2017-06-28 Miltenyi Biotec GmbH Chimeric antigen receptor with cytokine receptor activating or blocking domain
US11512139B2 (en) 2015-12-23 2022-11-29 Miltenyi Biotec B.V. & Co. KG Chimeric antigen receptor with cytokine receptor activating or blocking domain
US11136562B2 (en) 2016-01-08 2021-10-05 The Regents Of The University Of California Conditionally active heterodimeric polypeptides and methods of use thereof
JP2022064949A (en) * 2016-01-08 2022-04-26 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 Conditionally active heterodimeric polypeptides and methods of use thereof
CN109310727B (en) * 2016-01-08 2022-09-27 加利福尼亚大学董事会 Conditionally active heterodimeric polypeptides and methods of use thereof
JP7068169B2 (en) 2016-01-08 2022-05-16 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 Conditionally active heterodimer polypeptide and how to use it
WO2017120546A1 (en) * 2016-01-08 2017-07-13 The Regents Of The University Of California Conditionally active heterodimeric polypeptides and methods of use thereof
JP2019507586A (en) * 2016-01-08 2019-03-22 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 Conditionally active heterodimeric polypeptides and methods of using the same
CN109310727A (en) * 2016-01-08 2019-02-05 加利福尼亚大学董事会 It has ready conditions active heterologous polypeptides and its application method
US11466070B2 (en) 2016-02-12 2022-10-11 Autolus Limited Signalling system
WO2017137759A1 (en) * 2016-02-12 2017-08-17 Autolus Limited Signalling system
EP4227317A1 (en) * 2016-02-12 2023-08-16 Autolus Limited Signalling system
WO2017137758A1 (en) * 2016-02-12 2017-08-17 Autolus Limited Signalling system
EP3211420A1 (en) 2016-02-29 2017-08-30 Miltenyi Biotec GmbH Assay for detection of chimeric antigen receptor t cells
WO2017173403A1 (en) * 2016-03-31 2017-10-05 University Of Southern California A highly sensitive and specific luciferase based reporter assay for antigen detection
US11768203B2 (en) 2016-03-31 2023-09-26 University Of Southern California Highly sensitive and specific luciferase based reporter assay for antigen detection
US10597456B2 (en) 2016-04-01 2020-03-24 Amgen Inc. Chimeric receptors and methods of use thereof
EP3984559A1 (en) 2016-04-01 2022-04-20 Kite Pharma, Inc. Chimeric antigen and t cell receptors and methods of use
WO2017173410A1 (en) 2016-04-01 2017-10-05 Amgen Inc. Chimeric receptors to flt3 and methods of use thereof
WO2017173256A1 (en) 2016-04-01 2017-10-05 Kite Pharma, Inc. Chimeric antigen and t cell receptors and methods of use
US10689450B2 (en) 2016-04-01 2020-06-23 Kite Pharma, Inc BCMA binding molecules and methods of use thereof
WO2017173384A1 (en) 2016-04-01 2017-10-05 Kite Pharma, Inc. Chimeric receptors and methods of use thereof
US11505613B2 (en) 2016-04-01 2022-11-22 Kite Pharma, Inc. BCMA binding molecules and methods of use thereof
US10603380B2 (en) 2016-04-01 2020-03-31 Kite Pharma, Inc. Chimeric antigen and T cell receptors and methods of use
EP4180449A1 (en) 2016-04-01 2023-05-17 Kite Pharma, Inc. Chimeric receptors and methods of use thereof
US20220195039A1 (en) * 2016-04-14 2022-06-23 2Seventy Bio, Inc. Salvage chimeric antigen receptor systems
US11377637B2 (en) 2016-04-15 2022-07-05 Memorial Sloan Kettering Cancer Center Transgenic T cell and chimeric antigen receptor T cell compositions and related methods
US11879011B2 (en) 2016-05-13 2024-01-23 Bioatla, Inc. Anti-ROR2 antibodies, antibody fragments, their immunoconjucates and uses thereof
WO2017210617A2 (en) 2016-06-02 2017-12-07 Porter, David, L. Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
US11820819B2 (en) 2016-06-24 2023-11-21 Icell Gene Therapeutics Inc. Chimeric antigen receptors (CARs), compositions and methods thereof
JPWO2018052142A1 (en) * 2016-09-16 2019-06-24 キッセイ薬品工業株式会社 Genetically modified cell and method for producing the same
US10683355B2 (en) 2016-09-16 2020-06-16 Kissei Pharmaceutical Co., Ltd. Genetically-modified cells and method for producing same
US11286291B2 (en) * 2016-10-04 2022-03-29 Precision Biosciences, Inc. Co-stimulatory domains for use in genetically-modified cells
WO2018067992A1 (en) 2016-10-07 2018-04-12 Novartis Ag Chimeric antigen receptors for the treatment of cancer
EP3555296A4 (en) * 2016-12-13 2020-07-29 Seattle Children's Hospital (DBA Seattle Children's Research Institute) Methods of exogenous drug activation of chemical-induced signaling complexes expressed in engineered cells in vitro and in vivo
WO2018111834A1 (en) * 2016-12-13 2018-06-21 Seattle Children's Hospital (dba Seattle Children's Research Institute) Methods of exogenous drug activation of chemical-induced signaling complexes expressed in engineered cells in vitro and in vivo
US11753460B2 (en) 2016-12-13 2023-09-12 Seattle Children's Hospital Methods of exogenous drug activation of chemical-induced signaling complexes expressed in engineered cells in vitro and in vivo
WO2018112266A1 (en) 2016-12-14 2018-06-21 The Board Of Trustees Of The Leland Stanford Junior University Il-13 superkine: immune cell targeting constructs and methods of use thereof
US11649288B2 (en) 2017-02-07 2023-05-16 Seattle Children's Hospital Phospholipid ether (PLE) CAR T cell tumor targeting (CTCT) agents
EP4119552A1 (en) * 2017-02-08 2023-01-18 Dana-Farber Cancer Institute, Inc. Regulating chimeric antigen receptors
EP3580212A4 (en) * 2017-02-08 2021-03-17 Dana Farber Cancer Institute, Inc. Regulating chimeric antigen receptors
US11311609B2 (en) 2017-02-08 2022-04-26 Dana-Farber Cancer Institute, Inc. Regulating chimeric antigen receptors
US11759480B2 (en) 2017-02-28 2023-09-19 Endocyte, Inc. Compositions and methods for CAR T cell therapy
US11850262B2 (en) 2017-02-28 2023-12-26 Purdue Research Foundation Compositions and methods for CAR T cell therapy
WO2018169922A2 (en) 2017-03-13 2018-09-20 Kite Pharma, Inc. Chimeric antigen receptors for melanoma and uses thereof
US11365236B2 (en) 2017-03-27 2022-06-21 Nkarta, Inc. Truncated NKG2D chimeric receptors and uses thereof in natural killer cell immunotherapy
US11896616B2 (en) 2017-03-27 2024-02-13 National University Of Singapore Stimulatory cell lines for ex vivo expansion and activation of natural killer cells
US11111310B2 (en) 2017-03-28 2021-09-07 Cell Design Labs, Inc. Chimeric polypeptides and methods of altering the membrane localization of the same
US11938153B2 (en) 2017-03-31 2024-03-26 The Board Of Trustees Of The Leland Stanford Junior University Methods of treating T cell exhaustion by inhibiting or modulating T cell receptor signaling
US10844120B2 (en) 2017-04-24 2020-11-24 Kite Pharma, Inc. Humanized antigen-binding domains and methods of use
EP4286415A2 (en) 2017-04-24 2023-12-06 Kite Pharma, Inc. Humanized antigen-binding domains against cd19 and methods of use
WO2018200496A1 (en) 2017-04-24 2018-11-01 Kite Pharma, Inc. Humanized antigen-binding domains against cd19 and methods of use
CN110621694A (en) * 2017-05-15 2019-12-27 奥托路斯有限公司 Cells comprising Chimeric Antigen Receptors (CAR)
WO2018211244A1 (en) * 2017-05-15 2018-11-22 Autolus Limited A cell comprising a chimeric antigen receptor (car)
US11542312B2 (en) 2017-06-19 2023-01-03 Medicenna Therapeutics, Inc. IL-2 superagonists in combination with anti-PD-1 antibodies
US11325957B2 (en) 2017-06-19 2022-05-10 Cell Design Labs, Inc. Methods and compositions for reducing the immunogenicity of chimeric notch receptors
WO2019051001A1 (en) * 2017-09-06 2019-03-14 California Institute Of Technology Signaling and antigen-presenting bifunctional receptors (sabr)
EP3684424A4 (en) * 2017-09-22 2021-09-29 H. Lee Moffitt Cancer Center And Research Institute, Inc. Chimeric antigen receptors with enhanced nfkb signaling
US11932872B2 (en) 2017-09-26 2024-03-19 Nanjing Anji Biological Technology Co., Ltd Dual chimeric antigen receptor-t cell which can be regulated, construction method therefor and use thereof
WO2019062518A1 (en) * 2017-09-26 2019-04-04 南京安吉生物科技有限公司 New dual chimeric antigen receptor-t cell which can be regulated, construction method therefor and use thereof
US11890302B2 (en) 2017-09-29 2024-02-06 TC Biopharm Limited Gamma delta CAR-T cells comprising Fc gamma intracellular signaling domains
WO2019064030A1 (en) * 2017-09-29 2019-04-04 TC Biopharm Limited Modified car-t
JP2021500866A (en) * 2017-09-29 2021-01-14 ティーシー バイオファーム リミテッド Modified CAR-T
CN111417650A (en) * 2017-09-29 2020-07-14 特希生物制药有限公司 Modified CAR-T
WO2019084288A1 (en) 2017-10-25 2019-05-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
US11390655B2 (en) 2017-11-16 2022-07-19 Kite Pharma, Inc. Modified chimeric antigen receptors and methods of use
WO2019099707A1 (en) 2017-11-16 2019-05-23 Kite Pharma, Inc Modified chimeric antigen receptors and methods of use
WO2019115818A3 (en) * 2017-12-14 2019-07-25 Celyad S.A. Pooling signaling and costimulatory domains in flexible car design
US11952413B2 (en) 2017-12-14 2024-04-09 2Seventy Bio, Inc. Dimerizing agent regulated immunoreceptor complexes comprising interleukin receptor signaling domains
EP3723787A4 (en) * 2017-12-14 2021-09-01 Bluebird Bio, Inc. Daric interleukin receptors
US11844813B2 (en) 2017-12-20 2023-12-19 St. Anna Kinderkrebsforschung Ligand regulated protein-protein interaction system
EP3502130A1 (en) 2017-12-20 2019-06-26 St. Anna Kinderkrebsforschung Ligand regulated protein-protein interaction system
WO2019122188A1 (en) 2017-12-20 2019-06-27 St. Anna Kinderkrebsforschung Ligand regulated protein-protein interaction system
WO2019126358A1 (en) * 2017-12-22 2019-06-27 Cell Design Labs, Inc. Single- and multi-chain chimeric antigen receptors
WO2019134950A1 (en) 2018-01-04 2019-07-11 Miltenyi Biotec Gmbh Chimeric antigen receptor specific for bdca2 antigen
US11779602B2 (en) 2018-01-22 2023-10-10 Endocyte, Inc. Methods of use for CAR T cells
US11311576B2 (en) 2018-01-22 2022-04-26 Seattle Children's Hospital Methods of use for CAR T cells
EP3517125A1 (en) * 2018-01-24 2019-07-31 Xuanwu Hospital of Capital Medical University Chimeric antigen receptor for efficient targeted proliferation in vitro and uses thereof
US11001639B2 (en) * 2018-01-24 2021-05-11 Xuanwu Hospital Capital Medical University Chimeric antigen receptor for efficient selective proliferation in vitro and uses thereof
WO2019161271A1 (en) 2018-02-16 2019-08-22 Kite Pharma, Inc. Modified pluripotent stem cells and methods of making and use
WO2019200007A1 (en) 2018-04-10 2019-10-17 Amgen Inc. Chimeric receptors to dll3 and methods of use thereof
WO2019197676A1 (en) * 2018-04-13 2019-10-17 Ludwig Institute For Cancer Research Ltd Heterodimeric inactivatable chimeric antigen receptors
KR20210058769A (en) 2018-04-18 2021-05-24 앱클론(주) Switch Molecule And Switchable Chimeric Antigen Receptor
KR20190121716A (en) 2018-04-18 2019-10-28 앱클론(주) Switch Molecule And Switchable Chimeric Antigen Receptor
WO2019210207A3 (en) * 2018-04-27 2019-11-28 The Trustees Of The University Of Pennsylvania Chimeric antigen receptor t regulatory cells for the treatment of atherosclerosis
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
WO2019232523A1 (en) 2018-06-01 2019-12-05 The Board Of Trustees Of The Leland Stanford Junior University Il-13/il-4 superkines: immune cell targeting constructs and methods of use thereof
US11952428B2 (en) 2018-06-13 2024-04-09 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11939389B2 (en) 2018-06-13 2024-03-26 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11708401B2 (en) 2018-06-22 2023-07-25 Kite Pharma, Inc. Chimeric transmembrane proteins and uses thereof
WO2020018695A1 (en) 2018-07-18 2020-01-23 Amgen Inc. Chimeric receptors to steap1 and methods of use thereof
WO2020033927A2 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Processes for generating engineered cells and compositions thereof
WO2020047449A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020047452A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020050993A3 (en) * 2018-09-07 2020-04-16 Bioatla, Llc Conditionally active chimeric antigen receptors for modified t-cells
TWI835848B (en) 2018-09-07 2024-03-21 美商拜奧亞特拉公司 Conditionally active chimeric antigen receptors for modified t-cells
WO2020069405A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd22 chimeric antigen receptor (car) therapies
WO2020069409A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
WO2020070290A1 (en) 2018-10-05 2020-04-09 St. Anna Kinderkrebsforschung A group of chimeric antigen receptors (cars)
WO2020070289A1 (en) 2018-10-05 2020-04-09 St. Anna Kinderkrebsforschung A group of chimeric antigen receptors (cars)
EP3632461A1 (en) 2018-10-05 2020-04-08 St. Anna Kinderkrebsforschung A group of chimeric antigen receptors (cars)
EP3632460A1 (en) 2018-10-05 2020-04-08 St. Anna Kinderkrebsforschung A group of chimeric antigen receptors (cars)
CN113286608A (en) * 2018-10-05 2021-08-20 圣安娜儿童癌症研究中心 Chimeric Antigen Receptor (CAR) groups
CN113164576A (en) * 2018-10-05 2021-07-23 圣安娜儿童癌症研究中心 Chimeric Antigen Receptor (CAR) groups
US20220041687A1 (en) * 2018-10-05 2022-02-10 St. Anna Kinderkrebsforschung A group of chimeric antigen receptors (cars)
WO2020117952A2 (en) 2018-12-05 2020-06-11 Genentech, Inc. Diagnostic methods and compositions for cancer immunotherapy
EP4198057A1 (en) 2018-12-05 2023-06-21 F. Hoffmann-La Roche AG Diagnostic methods and compositions for cancer immunotherapy
WO2020123691A2 (en) 2018-12-12 2020-06-18 Kite Pharma, Inc Chimeric antigen and t cell receptors and methods of use
US11793834B2 (en) 2018-12-12 2023-10-24 Kite Pharma, Inc. Chimeric antigen and T cell receptors and methods of use
WO2020152197A1 (en) 2019-01-23 2020-07-30 Miltenyi Biotec B.V. & Co. KG A combination of compositions for elimination and enhanced engraftment of hematopoietic stem cells in the bone marrow of a subject
WO2020180591A1 (en) 2019-03-01 2020-09-10 Allogene Therapeutics, Inc. Dll3 targeting chimeric antigen receptors and binding agents
US11673953B2 (en) 2019-03-01 2023-06-13 Allogene Therapeutics, Inc. DLL3 targeting chimeric antigen receptors and binding agents
US11141436B2 (en) 2019-03-05 2021-10-12 Nkarta, Inc. Immune cells engineered to express CD19-directed chimeric antigen receptors and uses thereof in immunotherapy
US11253547B2 (en) 2019-03-05 2022-02-22 Nkarta, Inc. CD19-directed chimeric antigen receptors and uses thereof in immunotherapy
US11154575B2 (en) 2019-03-05 2021-10-26 Nkarta, Inc. Cancer immunotherapy using CD19-directed chimeric antigen receptors
WO2020201527A1 (en) 2019-04-04 2020-10-08 Umc Utrecht Holding B.V. Modified immune receptor constructs
WO2020210678A1 (en) 2019-04-12 2020-10-15 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020219848A1 (en) 2019-04-26 2020-10-29 Allogene Therapeutics, Inc. Rituximab-resistant chimeric antigen receptors and uses thereof
US11896617B2 (en) 2019-04-26 2024-02-13 Allogene Therapeutics, Inc. Polynucleotides encoding rituximab-resistant chimeric antigen receptors
WO2020219812A1 (en) 2019-04-26 2020-10-29 Allogene Therapeutics, Inc. Methods of manufacturing allogeneic car t cells
EP4295851A2 (en) 2019-04-26 2023-12-27 Allogene Therapeutics, Inc. Rituximab-resistant chimeric antigen receptors and uses thereof
WO2020223571A1 (en) 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cells expressing a chimeric receptor from a modified cd247 locus, related polynucleotides and methods
WO2020223535A1 (en) 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cells expressing a recombinant receptor from a modified tgfbr2 locus, related polynucleotides and methods
WO2020239866A1 (en) 2019-05-28 2020-12-03 Miltenyi Biotec B.V. & Co. KG Method for generation of genetically modified t cells
WO2021047804A1 (en) 2019-09-11 2021-03-18 Miltenyi Biotec B.V. & Co. KG In vitro method for transduction of t cells in the presence of malignant cells
CN114929752A (en) * 2019-10-08 2022-08-19 省卫生服务机构 Chimeric cytokine receptors
EP4069728A4 (en) * 2019-12-03 2023-12-27 University of Massachusetts Compositions and methods for optogenetic immunotherapy
CN113087805A (en) * 2019-12-31 2021-07-09 华东师范大学 Preparation and application of chimeric antigen receptor T cell of co-expression immune regulatory molecule
WO2021156277A1 (en) 2020-02-04 2021-08-12 Miltenyi Biotec B.V. & Co. KG Immune cell expressing adapter chimeric antigen receptor for sensing soluble antigens
WO2021163618A1 (en) 2020-02-14 2021-08-19 Novartis Ag Method of predicting response to chimeric antigen receptor therapy
WO2021173995A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021173985A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
EP3916388A1 (en) 2020-05-27 2021-12-01 Miltenyi Biotec B.V. & Co. KG Reagents for detection of chimeric antigen receptor cells
EP3915578A1 (en) 2020-05-28 2021-12-01 Miltenyi Biotec B.V. & Co. KG Chimeric antigen receptor with a spacer comprising c2-set ig-like domains
WO2021260186A1 (en) 2020-06-26 2021-12-30 Juno Therapeutics Gmbh Engineered t cells conditionally expressing a recombinant receptor, related polynucleotides and methods
EP3964585A1 (en) 2020-09-03 2022-03-09 Miltenyi Biotec B.V. & Co. KG Cd62l specific lentiviral vector particle for targeted transduction of t cell subsets
WO2022049217A1 (en) 2020-09-04 2022-03-10 Miltenyi Biotec B.V. & Co. KG System for inducible expression of an adapter in immune cells
WO2022096664A1 (en) 2020-11-09 2022-05-12 Miltenyi Biotec B.V. & Co. KG Methods and compositions for eliminating engineered immune cells
WO2022104061A1 (en) 2020-11-13 2022-05-19 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
WO2022140388A1 (en) 2020-12-21 2022-06-30 Allogene Therapeutics, Inc. Protease-activating cd45-gate car
WO2022165378A1 (en) * 2021-01-29 2022-08-04 Outpace Bio, Inc. Small molecule-regulated cell signaling expression system
WO2022258981A1 (en) 2021-06-11 2022-12-15 Quell Therapeutics Limited Multichain chimeric antigen receptor
WO2023009989A1 (en) 2021-07-26 2023-02-02 Kite Pharma, Inc. Split chimeric antigen receptors and methods of use
WO2023044304A1 (en) * 2021-09-15 2023-03-23 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric adaptor and kinase signaling proteins and their use in immunotherapy
WO2023057285A1 (en) 2021-10-06 2023-04-13 Miltenyi Biotec B.V. & Co. KG Method for targeted gene insertion into immune cells
WO2023062113A1 (en) 2021-10-15 2023-04-20 Miltenyi Biotec B.V. & Co. KG Method for the generation of genetically modified nk cells
WO2023187031A1 (en) 2022-04-01 2023-10-05 Miltenyi Biotec B.V. & Co. KG A system for drug-inducible expression of a polynucleotide
WO2023217796A1 (en) 2022-05-10 2023-11-16 Miltenyi Biotec B.V. & Co. KG Compositions comprising il-15, il-15 receptor alpha and the intracellular signaling domain of cd2 for immune cell therapy
WO2023220459A1 (en) * 2022-05-13 2023-11-16 Northwestern University Active loading of cargo entity into lipid bilayer particles using dimerization domains
WO2023222617A1 (en) 2022-05-16 2023-11-23 Miltenyi Biotec B.V. & Co. KG Endogenous signaling molecule activating chimeric antigen receptors and methods of generation thereof
WO2024056809A1 (en) 2022-09-15 2024-03-21 Novartis Ag Treatment of autoimmune disorders using chimeric antigen receptor therapy
EP4342488A1 (en) 2022-09-26 2024-03-27 Miltenyi Biotec B.V. & Co. KG Chimeric antigen receptor specific for folate receptor 1

Also Published As

Publication number Publication date
KR20200083682A (en) 2020-07-08
US20210196757A1 (en) 2021-07-01
HK1253407A1 (en) 2019-06-14
EP2956175A1 (en) 2015-12-23
MX2015010522A (en) 2016-06-23
HRP20192123T1 (en) 2020-02-21
JP2019068822A (en) 2019-05-09
US11478510B2 (en) 2022-10-25
DK2956175T3 (en) 2017-11-27
EP4303232A2 (en) 2024-01-10
DK3300745T3 (en) 2019-11-04
IL240472A0 (en) 2015-09-24
US10105391B2 (en) 2018-10-23
EP3613439A1 (en) 2020-02-26
PT3300745T (en) 2019-11-27
JP2023145542A (en) 2023-10-11
US20230051989A1 (en) 2023-02-16
HUE036250T2 (en) 2018-06-28
PL3613439T3 (en) 2021-11-22
US20150368342A1 (en) 2015-12-24
KR20210147101A (en) 2021-12-06
EP3881868B1 (en) 2023-09-27
AU2019246785B2 (en) 2021-04-08
IL272279B1 (en) 2023-10-01
US20160185862A1 (en) 2016-06-30
JP6687712B2 (en) 2020-04-28
KR20150119134A (en) 2015-10-23
NZ710925A (en) 2021-03-26
EP3881868A1 (en) 2021-09-22
ES2653487T3 (en) 2018-02-07
ES2868247T3 (en) 2021-10-21
CY1122386T1 (en) 2021-01-27
US20170143765A1 (en) 2017-05-25
KR20230022452A (en) 2023-02-15
JP2022046809A (en) 2022-03-23
JP2016508518A (en) 2016-03-22
CN105142677A (en) 2015-12-09
EP4303232A3 (en) 2024-04-17
US20200237824A1 (en) 2020-07-30
AU2023204612A1 (en) 2023-08-03
EP3300745B9 (en) 2020-04-15
CN105142677B (en) 2019-08-30
AU2014216130B2 (en) 2017-11-16
PL3300745T3 (en) 2020-03-31
KR20190131152A (en) 2019-11-25
AU2019246785A1 (en) 2019-10-31
HK1218625A1 (en) 2017-03-03
EP3300745A1 (en) 2018-04-04
EP2956175A4 (en) 2016-08-24
PT3613439T (en) 2021-05-12
PT2956175T (en) 2017-12-26
AU2021204054B2 (en) 2023-04-20
US20180042963A1 (en) 2018-02-15
NZ750521A (en) 2021-03-26
ES2758227T9 (en) 2020-05-12
LT2956175T (en) 2017-12-11
IL305629A (en) 2023-11-01
AU2018201102A1 (en) 2018-03-08
AU2014216130A1 (en) 2015-08-27
US9821012B2 (en) 2017-11-21
EP2956175B1 (en) 2017-10-04
CN110423282B (en) 2023-09-08
HUE047487T2 (en) 2020-04-28
EP3881868C0 (en) 2023-09-27
US9587020B2 (en) 2017-03-07
ES2758227T3 (en) 2020-05-04
MX369545B (en) 2019-11-12
KR102064230B1 (en) 2020-01-13
US10632152B2 (en) 2020-04-28
MX2019013498A (en) 2020-01-20
IL240472B (en) 2020-02-27
LT3300745T (en) 2019-12-10
KR102132246B1 (en) 2020-07-09
EP3613439B1 (en) 2021-04-07
CA2901115A1 (en) 2014-08-21
IL272279A (en) 2020-02-27
IL272279B2 (en) 2024-02-01
JP2020121982A (en) 2020-08-13
KR102332790B1 (en) 2021-12-01
EP3300745B1 (en) 2019-09-11
US10888581B2 (en) 2021-01-12
US20170340672A1 (en) 2017-11-30
JP6450690B2 (en) 2019-01-09
US20180085401A1 (en) 2018-03-29
PL2956175T3 (en) 2018-05-30
SI3300745T1 (en) 2020-01-31
BR112015019640A2 (en) 2021-08-03
AU2021204054A1 (en) 2021-07-08
JP7317159B2 (en) 2023-07-28
JP7014843B2 (en) 2022-02-01
NO2929995T3 (en) 2018-02-03
CN110423282A (en) 2019-11-08
SI3613439T1 (en) 2021-11-30

Similar Documents

Publication Publication Date Title
US11478510B2 (en) Chimeric antigen receptor and methods of use thereof
NZ710925B2 (en) Chimeric antigen receptor and methods of use thereof

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201480016737.0

Country of ref document: CN

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

Ref document number: 14751227

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14766105

Country of ref document: US

REEP Request for entry into the european phase

Ref document number: 2014751227

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2014751227

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2015558159

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 240472

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2901115

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/A/2015/010522

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015019640

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2014216130

Country of ref document: AU

Date of ref document: 20140214

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20157024731

Country of ref document: KR

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01E

Ref document number: 112015019640

Country of ref document: BR

Free format text: - SOLICITA-SE APRESENTAR NOVAS FOLHAS REFERENTES AOS DESENHOS DO PEDIDO, UMA VEZ QUE O CONJUNTO APRESENTADO TEM INCORRECAO NA NUMERACAO DE SUAS PAGINAS OU PAGINA FALTANTE (PAG. 32); - SOLICITA-SE APRESENTAR NOVO ARQUIVO ELETRONICO DA LISTAGEM DE SEQUENCIAS BIOLOGICAS COM CORRECAO DO TITULO E NOME DO DEPOSITANTE DO PEDIDO NA FASE NACIONAL.

ENP Entry into the national phase

Ref document number: 112015019640

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150814