WO2012082841A2 - Universal anti-tag chimeric antigen receptor-expressing t cells and methods of treating cancer - Google Patents

Universal anti-tag chimeric antigen receptor-expressing t cells and methods of treating cancer Download PDF

Info

Publication number
WO2012082841A2
WO2012082841A2 PCT/US2011/064808 US2011064808W WO2012082841A2 WO 2012082841 A2 WO2012082841 A2 WO 2012082841A2 US 2011064808 W US2011064808 W US 2011064808W WO 2012082841 A2 WO2012082841 A2 WO 2012082841A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
car
subject
expressing
fitc
Prior art date
Application number
PCT/US2011/064808
Other languages
French (fr)
Other versions
WO2012082841A9 (en
Inventor
Eduardo Davila
Koji Tamada
Original Assignee
University Of Maryland, Baltimore
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=46245339&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2012082841(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to AU2011343887A priority Critical patent/AU2011343887B2/en
Priority to EP11849221.4A priority patent/EP2651442B1/en
Priority to ES11849221T priority patent/ES2791716T3/en
Priority to EP20161627.3A priority patent/EP3693017A1/en
Priority to JP2013544725A priority patent/JP5897035B2/en
Application filed by University Of Maryland, Baltimore filed Critical University Of Maryland, Baltimore
Priority to CA2821080A priority patent/CA2821080C/en
Priority to US13/993,396 priority patent/US9233125B2/en
Publication of WO2012082841A2 publication Critical patent/WO2012082841A2/en
Publication of WO2012082841A9 publication Critical patent/WO2012082841A9/en
Priority to US14/990,514 priority patent/US10973893B2/en
Priority to US17/228,001 priority patent/US20210330769A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464403Receptors for growth factors
    • A61K39/464404Epidermal growth factor receptors [EGFR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular 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
    • A61K39/464403Receptors for growth factors
    • A61K39/464406Her-2/neu/ErbB2, Her-3/ErbB3 or Her 4/ ErbB4
    • 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
    • A61K47/54Medicinal 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 organic compound
    • A61K47/555Medicinal 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 organic compound pre-targeting systems involving an organic compound, other than a peptide, protein or antibody, for targeting specific cells
    • 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
    • A61K47/6835Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6855Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from breast cancer cell
    • 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
    • A61K47/6835Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6859Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from liver or pancreas cancer cell
    • 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
    • A61K47/6835Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6863Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from stomach or intestines cancer cell
    • 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
    • A61K47/6897Pre-targeting systems with two or three steps using antibody conjugates; Ligand-antiligand therapies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • 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
    • C07K16/2863Immunoglobulins [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 growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/572Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 cytotoxic response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • A61K2039/585Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/50Colon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/54Pancreas
    • 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
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/515CD3, T-cell receptor complex
    • 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

  • the invention relates to T cell-based anti-cancer therapeutics and methods of using the therapeutics in the treatment of cancer.
  • a chimeric antigen receptor CAR
  • the extracellular domain of a typical CAR consists of the V H and V L domains -single-chain fragment variable (scFv)- from the antigen binding sites of a monoclonal antibody.
  • the scFv is linked to a flexible transmembrane domain followed by a tyrosine-based activation motif such as that from CD3 ⁇ (Sadelain et al. Curr. Opin. Immunol.
  • CAR T cells offer the opportunity to seek out and destroy cancer cells by recognizing tumor-associated antigens (TAA) expressed on their surface (Sadelain et al. Curr. Opin. Immunol. 21, 215-223 (2009)). As such, the recognition of a tumor cells occurs via an MHC-independent mechanism.
  • TAA tumor-associated antigens
  • the present invention provides a universal, yet adaptable, anti-tag chimeric antigen receptor (AT-CAR)-expressing T cell system that fully addresses the deficiencies of current systems.
  • the system uses a gene therapy platform to generate immune cells capable of recognizing various cancers types and that have broad and valuable clinical implications for the use of T cell-based therapies.
  • a versatile AT-CAR system which grants T cells specificity to recognize and bind tagged proteins, such as antibodies, has been developed.
  • ccFITC-CAR-expressing human T cells have been developed that specifically recognize various human cancer cells when those cells are bound by cancer-reactive FITC-labeled antibodies.
  • the activation of FITC-CAR-expressing T cells is shown to induce efficient target lysis, T cell proliferation, and cytokine/chemokine production in vitro and ex vivo.
  • aFITC-CAR-expressing T cells plus FITC-cetuximab (Ctx) are shown to delay colon cancer tumor establishment but lead to the selection of tumor- associated antigen (TAA)-negative cancer cells.
  • ccFITC-CAR-expressing T cells were observed to eradicate an established tumor and prevent tumor growth.
  • This 'off-the-shelf system advances existing CAR technology through its potential to target various tagged proteins in the treatment of cancer patients.
  • the invention is drawn to a method of treating cancer in a subject, comprising: (a) administering a formulation of tagged proteins to a subject in need of treatment, wherein the tagged proteins bind a cancer cell in the subject, and (b) administering a therapeutically-effective population of anti-tag chimeric antigen receptor (AT-CAR)-expressing T cells to the subject, wherein the AT-CAR-expressing T cells bind the tagged proteins and induce cancer cell death, thereby treating cancer in a subject.
  • AT-CAR anti-tag chimeric antigen receptor
  • the invention is drawn to a method of treating cancer in a subject, comprising: (a) administering one or more formulations of tagged proteins to a subject in need of treatment, wherein the tagged proteins bind a cancer cell in the subject, and (b) administering one or more therapeutically-effective populations of AT-CAR-expressing T cells to the subject, wherein the AT-CAR-expressing T cells bind the tagged proteins and induce cancer cell death, thereby treating cancer in a subject.
  • the invention is drawn to a method of treating cancer in a subject, comprising: (a) administering at least two formulations of tagged proteins to a subject in need of treatment, wherein the tagged proteins bind a cancer cell in the subject, and (b) administering at least two therapeutically-effective populations of AT-CAR-expressing T cells to the subject, wherein the AT-CAR-expressing T cells bind the tagged proteins and induce cancer cell death, thereby treating cancer in a subject.
  • the tag of each formulation of tagged proteins is the same or different and the tag is selected from the group consisting of fluorescein isothiocyanate (FITC), streptavidin, biotin, histidine, dinitrophenol, peridinin chlorophyll protein complex, green fluorescent protein, phycoerythrin (PE), horse radish peroxidase, palmitoylation, nitrosylation, alkalanine phosphatase, glucose oxidase, and maltose binding protein.
  • FITC fluorescein isothiocyanate
  • streptavidin biotin
  • biotin histidine
  • dinitrophenol dinitrophenol
  • peridinin chlorophyll protein complex dinitrophenol
  • peridinin chlorophyll protein complex green fluorescent protein
  • PE phycoerythrin
  • horse radish peroxidase palmitoylation
  • nitrosylation alkalanine phosphatase
  • glucose oxidase glucose oxidas
  • the protein of each formulation of tagged proteins is the same or different and the protein is an antibody or an antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof is cetuximab, nimotuzumab, panitumumab, retuximab, omalizumab, tositumomab, trastuzumab, gemtuzumab, alemtuzumab, bevacuzimab or an antigen-binding fragment of any one thereof.
  • the AT-CAR of each population of AT-CAR-expressing T cells is the same or different and the AT-CAR comprises a tag-binding domain, a transmembrane domain, and an activation domain.
  • the tag-binding domain is an antibody or an antigen-binding fragment thereof.
  • the tag-binding domain specifically binds FITC, biotin, PE, histidine or streptavidin.
  • the antigen-binding domain is a single chain variable fragment (scFv), such as a scFv that specifically binds FITC, biotin, PE, histidine or streptavidin.
  • scFv single chain variable fragment
  • the transmembrane domain is the hinge and transmembrane regions of the human CD8a chain.
  • the activation domain comprises one or more of the cytoplasmic region of CD28, the cytoplasmic region of CD137 (41BB), OX40, HVEM, CD3 ⁇ and FcRs.
  • the T cells of each population of AT-CAR-expressing T cells are the same or different and wherein the T cells are selected from the group consisting of T cells of any HLA -background from peripheral blood mononuclear cells (PBMC), T cells isolated from a tumor explant of the subject, and
  • PBMC peripheral blood mononuclear cells
  • the T cells of each population of AT-CAR-expressing T cells consist of HLA-A2+ peripheral blood mononuclear cells (PBMC).
  • PBMC peripheral blood mononuclear cells
  • the formulation(s) of tagged proteins are administered to the subject prior to administration of the therapeutically- effective population(s) of AT-CAR-expressing T cells.
  • the formulation(s) of tagged proteins are administered to the subject concurrently with administration of the therapeutically-effective population(s) of AT-CAR-expressing T cells.
  • the formulation(s) of tagged proteins are administered to the subject after administration of the therapeutically- effective population(s) of AT-CAR-expressing T cells.
  • the formulation(s) of tagged proteins and the therapeutically-effective population(s) of AT-CAR-expressing T cells are administered to the subject in any order.
  • AT-CAR-expressing T cell binding to the tagged proteins induces cytolytic activation of the T cells.
  • the subject is a human.
  • FIG. 1 Anti-FITC-CAR expression, characterization and in vitro functionality.
  • A Diagram of the anti-FITC-CAR (ccFITC-CAR). Boxed diagram on left shows the genetic elements included in the polynucleotide engineered to express ccFITC-CAR, beginning with a 5'- long terminal repeat, and followed by the coding region for anti-FITC scFv, CD28
  • transmembrane domain CD28, 4 IBB, zeta chain ( ⁇ ) and a 3 '-long terminal repeat.
  • Figure on right shows the ccFITC-CAR polypeptide traversing the T cell membrane and position to bind a tumor-reactive antibody labeled with FITC.
  • HLA-A2 + PBMCs were activated with anti-CD3 antibodies in the presence of IL-2 followed by transduction with ccFITC-CAR retrovirus.
  • ccFITC- CAR expression was determined by staining cells with CD3 and FITC-conjugated cetuximab (FITC-Ctx) or FITC-conjugated dextran beads (FITC-Dex).
  • Cetuximab is an antibody with binding specificity for EGFR-expressing tumor cells.
  • C ccFITC-CAR functionality on transduced and control T cells was examined in proliferation assays using plate-bound FITC-Ctx, FITC-Dex or Ctx.
  • D The proliferation of FrfC-CAR T cells and control T cells was measured in response to stimulation with SW480 colon cancer cells stained with titrating concentrations of FITC-Ctx.
  • E T cell cytotoxicity by ccFITC-CAR and control T cells was measured against SW480 colon cancer cells stained with FITC-Ctx or FITC-mouse IgG.
  • F ccFITC-CAR
  • cytotoxicity was measured against Pane 6.03 stained with unlabeled Ctx and Her2 antibodies or stained with FITC-Ctx or FITC-Her2 at various effector to target ratios. Alternatively AU565 breast cancer cells were stained with FITC-Her2 or unlabled Her2 mAb. All data are
  • FIG. 1 ccFITC-CAR T cells delay tumor establishment but promote the growth of antigen-negative tumor cells.
  • SW480 human colon cancer cells were injected subcutaneously into NSG mice followed by injection with FITC-Ctx (i.p.) one day later. Twenty four hours after, 5xl0 6 ccFITC-CAR T cells were injected into the tail vein. FITC-Ctx was injected (i.p.) weekly for three weeks. Data are representative of two or three experiments (three or more mice per group). *, P ⁇ 0.02, ANOVA; n.s. not significant.
  • B The percentage of CD3 + ccFITC-CAR positive and negative T cells was determined by flow cytometry. The average percentage of
  • FITC-CAR + T cells prior to injection or from the tumor explant of four mice is shown.
  • C Tumor explants were finely minced and cultured in trypsin for 2 hours at 37°C followed by T cell enrichment using a negative selection kit. T cells were co-cultured with SW480 colon cancer cells which had been pulsed with FITC-Ctx or Ctx (O ⁇ g/lxlO 6 cells). Three days later proliferation was determined by [ H]thymidine uptake (+SD).
  • D Alternatively, cytokine and chemokine production was measured using a Milliplex array 72 hours after stimulation.
  • E EGFR expression on SW480 tumor explants or SW480 cells taken from tissue culture was examined by flow cytometry using FITC-Ctx.
  • FIG. 3 ccFITC-CAR T cells prevent tumor growth and eradicate established tumors.
  • A EGFR expression on the pancreatic human cancer cells (Pane 6.03) was determined by staining cells with FITC-Ctx or with control antibody FITC-mlgG and analyzed by flow cytometry.
  • “about” refers to a numeric value, including, for example, whole numbers, fractions, and percentages, whether or not explicitly indicated.
  • the term “about” generally refers to a range of numerical values (e.g., +/- 5-10% of the recited value) that one of ordinary skill in the art would consider equivalent to the recited value (e.g., having the same function or result). In some instances, the term “about” may include numerical values that are rounded to the nearest significant figure.
  • ccFITC-CAR anti-FITC CAR-CD28-41BB-CD3
  • This platform takes advantage of the high affinity interaction between the anti-FITC scFv (on the cell's surface) and FITC as well as the ability to cheaply and easily conjugated FITC molecules (or other tags) to any anti-cancer-based monoclonal antibody such as cetuximab (anti-EGFR), retuximab (anti-CD20) and herceptin (anti-Her2) used to treat patients with various types of cancers.
  • This system allows for extreme specificity to the antigen and is accompanied by robust effector function and proliferative capacity with no cross -reactivity to 'self-antigens' .
  • effector cells may be autologous, syngeneic or allogeneic, with the selection dependent on the disease to be treated and the means available to do so.
  • Suitable populations of effector cells that may be used in the methods include any immune cells with cytolytic activity, such as T cells.
  • Exemplary sub- populations of T cells include, but are not limited to those expressing CD3 + including CD3 + CD8 + T cells, CD3 + CD4+ T cells, and NKT cells.
  • the T cells are HLA-A2+ peripheral blood mononuclear cells (PBMC) but the T cells can be of any HLA background from PBMCs and utilized in an autologous, syngeneic or allogeneic system.
  • PBMC peripheral blood mononuclear cells
  • T cells may also be isolated from any source, including from a tumor explant of the subject being treated or intratumoral T cells of the subject being treated.
  • the effector cells are commonly referred to herein as T cells, but it should be understood that any reference to T cells, unless otherwise indicated, is a reference to all effector cell types as defined herein.
  • Anti-tag chimeric antigen receptor (AT-CAR).
  • AT-CAR anti-tag chimeric antigen receptor
  • the skilled artisan will appreciate that the anti-tag chimeric antigen receptor (AT-CAR) expressed by the T cells used in the methods of the present invention allow for great flexibility.
  • the sole requirements for the AT-CARs used in the methods are (i) that the AT-CAR has binding specificity for a particular tag that can be conjugated to a protein (such as an antibody) that binds to a tumor- associated antigen (TAA), and (ii) that T cells can be engineered to express the AT-CAR.
  • Additional features include (i) that the AT-CAR includes an activation domain that induces efficient target lysis upon T cell binding and activation, and (ii) the ability to replace the scFv portion of the AT-CAR with one of specificity to other tags, such as biotin or phycoerythrin, which can be conjugated to target (i.e., tumor) -reactive proteins such as antibodies to be used in vivo.
  • target i.e., tumor
  • the AT-CAR comprises three domains.
  • the first domain is the tag-binding domain. This domain is typically present at the amino terminal end of the
  • the tag-binding domain is typically an antibody or an antigen-binding fragment thereof.
  • the identity of the antibody or fragment is only limited by the identity of the tag of the tagged protein.
  • the antibodies may be obtained from any species of animal, though preferably from a mammal such as a human, simian, mouse, rat, rabbit, guinea pig, horse, cow, sheep, goat, pig, dog or cat.
  • the antibodies are human or humanized antibodies.
  • Antibody fragments include single-chain variable fragment (scFv), single chain antibodies, F(ab') 2 fragments, Fab fragments, and fragments produced by an Fab expression library, with the only limitation being that the antibody fragments retain the ability to bind the selected tag.
  • the antibodies may also be polyclonal, monoclonal, or chimeric antibodies, such as where an antigen binding region (e.g., F(ab')2 or hypervariable region) of a non-human antibody is transferred into the framework of a human antibody by recombinant DNA techniques to produce a substantially human molecule.
  • Antigen-binding fragments such as scFv, may be prepared therefrom.
  • AT-CARs of the present invention can be produced using commercially-available antibodies.
  • antibodies to a selected tag may be produced.
  • various hosts including, but not limited to, goats, rabbits, rats, mice, humans, etc., can be immunized by injection with a particular protein or any portion, fragment, or oligopeptide that retains immunogenic properties of the protein.
  • adjuvants can be used to increase the immunological response.
  • adjuvants include, but are not limited to, detoxified heat labile toxin from E. coli, Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol.
  • BCG Bacillus Calmette-Guerin
  • Corynebacterium parvum are also potentially useful adjuvants.
  • Antibodies and fragments thereof can be prepared using any technique that provides for the production of antibody molecules, such as by continuous cell lines in culture for monoclonal antibody production.
  • Such techniques include, but are not limited to, the hybridoma technique originally described by Koehler and Milstein (Nature 256:495-497 (1975)), the human B-cell hybridoma technique (Kosbor et al., Immunol Today 4:72 (1983); Cote et al., Proc Natl. Acad. Sci 80:2026-2030 (1983)), and the EBV-hybridoma technique (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss Inc, New York N.Y., pp 77-96 (1985)).
  • chimeric antibodies i.e., the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can also be used (Morrison et al., Proc Natl. Acad. Sci 81:6851-6855 (1984); Neuberger et al., Nature 312:604-608(1984); Takeda et al., Nature 314:452-454(1985)).
  • techniques described for the production of single chain antibodies such as disclosed in U.S. Patent No. 4,946,778, incorporated herein by reference in its entirety, can be adapted to produce tag- specific single chain antibodies.
  • the tag-binding domain is a single-chain variable fragment (scFv).
  • a scFv comprises the variable regions of the heavy (VH) and light chains (VL) of an antibody, typically linked via a short peptide of ten to about 25 amino acids.
  • the linker can either connect the N-terminus of the VH with the C-terminus of the VL, or vice versa.
  • the binding specificity of the tag-binding domain will depend on the identity of the tag that is conjugated to the protein that is used to bind target cells.
  • the tag when the tag is FITC (Fluorescein isothiocyanate), the tag-binding domain may constitute an anti-FITC scFv.
  • the tag when the tag is biotin or PE (phycoerythrin), the tag- binding domain may constitute an anti-biotin scFv or an anti-PE scFv.
  • the second domain is a transmembrane (TM) domain.
  • TM domain allows the CAR to be anchored into the cell membrane of the T cell.
  • TM domains include, but are not limited to, the hinge and transmembrane regions of the human CD8a chain.
  • the third domain when present, is the T cell activation domain.
  • This domain aids in T cell activation upon binding of the CAR to the tagged protein that is bound to the target cell.
  • T cell activation includes induction of cytokine and chemokine production, as well as activation of the cytolytic activity of the cells.
  • Exemplary T cell activation domains include, but are not limited to, the cytoplasmic regions of CD28, CD137 (41BB), OX40 and HVEM which serve to enhance T cell survival and proliferation; and CD3 ⁇ and FcRs which induce T cell activation.
  • One or more than one T cell activation domain may be included in the CAR, such as two, three, four or more T cell activation domains.
  • T cells may be engineered to express AT-CARs by means readily known to the skilled artisan.
  • a polynucleotide vector is constructed that encodes the AT-CAR and the vector is transfected into a population of T cells. The cells are then grown under conditions promoting expression of the AT-CAR by the T cells.
  • Successful transfection (or transduction which refers to viral-mediated gene integration) and display of AT- CARs by T cells is conducted via conventional means, some of which are disclosed in the Examples herein.
  • T cells may be engineered to produce AT-CARs by first constructing a retroviral vector encoding a selected AT-CAR.
  • An exemplary retroviral vector includes, but is not limited to, the vector backbone pMSGVl-CD8-28BBZ, which is derived from pMSGV (murine stem cell virus-based splice-gag vector). DNA sequencing can be used to confirm proper construction of the vector before transfection of T cells. Retroviral transduction may be performed using known techniques, such as that of Johnson et al. ⁇ Blood 114, 535-546 (2009)).
  • AT-CAR on transduced T cells may be determined, for example, by flow cytometry after staining cells with tag-conjugated protein or tag-conjugated dextran beads.
  • the tag portion of the protein or beads will be bound by the tag-binding domain of the CAR expressed by the cells.
  • AT-CAR T cell administration Populations of AT-CAR-expressing T cells may be formulated for administered to a subject using techniques known to the skilled artisan.
  • Formulations comprising populations of AT-CAR-expressing T cells may include
  • Excipients included in the formulations will have different purposes depending, for example, on the nature of the tag-binding domain comprising the AT-CAR, the subpopulation of T cells used, and the mode of administration.
  • examples of generally used excipients include, without limitation: saline, buffered saline, dextrose, water-for- infection, glycerol, ethanol, and combinations thereof, stabilizing agents, solubilizing agents and surfactants, buffers and preservatives, tonicity agents, bulking agents, and lubricating agents.
  • the formulations comprising populations of AT-CAR-expressing T cells will typically have been prepared and cultured in the absence of any non-human components, such as animal serum (e.g., bovine serum albumin).
  • a formulation may include one population of AT-CAR-expressing T cells, or more than one, such as two, three, four, five, six or more populations of AT-CAR-expressing T cells.
  • the different populations of AT-CAR T cells can vary based on the identity of the tag-binding domain, the identity of the activation domain, the identity of the subpopulation of T cells, or a combination thereof.
  • a formulation may comprise populations of AT-CAR-expressing T cells that recognize and bind to one, or more than one, such as two, three, four, five, six or more different tagged proteins.
  • a formulation may comprise populations of AT-CAR-expressing T cells that recognize and bind FITC, biotin and PE.
  • the formulation comprises three different populations of AT-CAR-expressing T cells that recognize and bind cells tagged by FITC-conjugated antibodies, biotin-conjugated antibodies, and PE-conjugated antibodies.
  • This formulation would therefore comprise aFrfC-CAR- expressing T cells, abiotin-CAR-expressing T cells and ⁇ -CAR-expressing T cells.
  • compositions comprising population(s) of AT-CAR T cells may be administered to a subject using modes and techniques known to the skilled artisan.
  • Exemplary modes include, but are not limited to, intravenous injection.
  • Other modes include, without limitation,
  • intratumoral intradermal, subcutaneous (s.c, s.q., sub-Q, Hypo), intramuscular (i.m.), intraperitoneal (i.p.), intra-arterial, intramedulary, intracardiac, intra- articular (joint),
  • intrasynovial joint fluid area
  • intracranial intracranial
  • intraspinal intrathecal
  • Any known device useful for parenteral injection or infusion of the formulations can be used to effect such administration.
  • compositions comprising population(s) of AT-CAR-expressing T cells that are administered to a subject comprise a number of AT-CAR-expressing T cells that is effective for the treatment and/or prophylaxis of the specific indication or disease.
  • therapeutically- effective populations of AT-CAR-expressing T cells are administered to subjects when the methods of the present invention are practiced.
  • formulations are administered that comprise between about 1 x 10 4 and about 1 x 10 10 AT-CAR-expressing T cells.
  • the formulation will comprise between about 1 x 10 5 and about 1 x 10 9 AT-CAR-expressing T cells, from about 5 x 10 5 to about 5 x 10 8 AT-CAR-expressing T cells, or from about 1 x 10 6 to about 1 x 10' AT-CAR -expressing T cells.
  • the number of AT-CAR-expressing T cells administered to a subject will vary between wide limits, depending upon the location, source, identity, extent and severity of the cancer, the age and condition of the individual to be treated, etc. A physician will ultimately determine appropriate dosages to be used.
  • Tagged proteins are administered to a subject prior to, or concurrent with, or after administration of the AT-CAR-expressing T cells.
  • the tagged proteins bind to target cells in the subject.
  • the "protein" portion of the tagged protein is the portion of the molecule that binds to the target cell.
  • the protein may be an antibody that binds to a tumor- associated antigen (TAA) or a tumor specific antigen (TSA) expressed by the target cell.
  • TAA tumor- associated antigen
  • TSA tumor specific antigen
  • the "protein” may be any molecule that binds to a target cell.
  • Exemplary proteins include, but are not limited to, anti-cancer-based monoclonal antibodies such as cetuximab (anti- EGFR), nimotuzumab (anti-EGFR), panitumumab (anti-EGFR), retuximab (anti-CD20), omalizumab (anti-CD20), tositumomab (anti-CD20), trastuzumab (anti-Her2), gemtuzumab (anti-CD33), alemtuzumab (anti-CD52), and bevacuzimab (anti-VEGF).
  • anti-cancer-based monoclonal antibodies such as cetuximab (anti- EGFR), nimotuzumab (anti-EGFR), panitumumab (anti-EGFR), retuximab (anti-CD20), omalizumab (anti-CD20), tositumomab (anti-CD20), trastuzumab (anti-
  • the "tag" portion of the tagged protein is only constrained by being a molecular that can be recognized and specifically bound by the AT-CAR, specifically, the tag-binding domain of the AT-CAR.
  • Exemplary tags include, but are not limited to, fluorescein isothiocyanate (FITC), dinitrophenol, peridinin chlorophyll protein complex, green fluorescent protein, biotin, phycoerythrin (PE), histidine, streptavidin, horse radish peroxidase, palmitoylation, nitrosylation, alkalanine phosphatase, glucose oxidase, Glutathione S -transferase, maltose binding protein, and any types of fluorescent materials including quantum dot nanocrystals.
  • FITC fluorescein isothiocyanate
  • PE peridinin chlorophyll protein complex
  • green fluorescent protein biotin
  • PE phycoerythrin
  • histidine streptavidin
  • the tagged proteins include FITC-conjugated antibodies, biotin-conjugated antibodies, PE-conjugated antibodies, histidine-conjugated antibodies and streptavidin-conjugated antibodies, where the antibody binds to a TAA or a TSA expressed by the target cells.
  • the tagged proteins of the present invention include, but are not limited to, FITC-conjugated cetuximab, FITC-conjugated retuximab, FITC-conjugated herceptin, biotin-conjugated cetuximab, biotin-conjugated retuximab, biotin-conjugated herceptin, PE- conjugated cetuximab, PE-conjugated retuximab, PE-conjugated herceptin, histidine-conjugated cetuximab, histidine-conjugated retuximab, histidine-conjugated herceptin, streptavidin- conjugated cetuximab, streptavidin-conjugated retuximab, and streptavidin-conjugated herceptin.
  • the AT-CAR cells can be redirected to target and/or destroy vascular cells feeding the tumor.
  • T cells expressing aFITC-VEGF as the AT-CAR can target endothelial vascular cells to which FITC-tagged VEGF is bound, where the FITC-tagged VEGF is bound by the VEGF receptor.
  • a protein lacking a tag may be used as the tagged protein.
  • a naked (tagless) protein such as an antibody
  • the tag-binding domain may be an antibody or antigen-binding fragment thereof that recognizes and binds a second antibody, where the second antibody functions as the tagged protein and where the second antibody lacks a tag.
  • the tag may be conjugated to the proteins using techniques such as chemical coupling and chemical cross -linkers.
  • polynucleotide vectors can be prepared that encode the tagged proteins as fusion proteins.
  • Cell lines can then be engineered to express the tagged proteins, and the tagged proteins can be isolated from culture media, purified and used in the methods disclosed herein.
  • the tagged proteins may be formulated for administered to a subject using techniques known to the skilled artisan.
  • Formulations of the tagged proteins may include pharmaceutically acceptable excipient(s).
  • Excipients included in the formulations will have different purposes depending, for example, on the nature of the tag, the protein, and the mode of administration. Examples of generally used excipients include, without limitation: saline, buffered saline, dextrose, water-for-infection, glycerol, ethanol, and combinations thereof, stabilizing agents, solubilizing agents and surfactants, buffers and preservatives, tonicity agents, bulking agents, and lubricating agents.
  • a formulation of tagged proteins may include one type of tagged protein, or more than one, such as two, three, four, five, six or more types of tagged proteins.
  • the different types of tagged proteins can vary based on the identity of the tag, the identity of the protein, or both.
  • a formulation comprising three types of tagged protein might include FITC- conjugated cetuximab, FITC-conjugated rituximab and FITC-conjugated herceptin, or FITC- conjugated cetuximab, biotin-conjugated cetuximab, and PE-conjugated cetuximab.
  • the tagged proteins may be administered to a subject using modes and techniques known to the skilled artisan.
  • Exemplary modes include, but are not limited to, intravenous, intraperitoneal, and intratumoral injection.
  • Other modes include, without limitation, intradermal, subcutaneous (s.c, s.q., sub-Q, Hypo), intramuscular (i.m.), intra-arterial, intramedulary, intracardiac, intra-articular (joint), intrasynovial (joint fluid area), intracranial, intraspinal, and intrathecal (spinal fluids). Any known device useful for parenteral injection or infusion of the formulations can be used to effect such administration.
  • Formulations comprising the tagged proteins are administered to a subject in an amount which is effective for treating and/or prophylaxis of the specific indication or disease.
  • formulations comprising at least about 0.1 mg/kg to about 100 mg/kg body weight of the tagged proteins are administered to a subject in need of treatment.
  • the dosage is from about 1 mg/kg to about 10 mg/kg body weight of the tagged proteins daily, taking into account the routes of administration, symptoms, etc.
  • tagged-bevacizumab is administered in a dosage of from about 2.5 to about 5 mg kg.
  • tagged- cetuximab is administered in a dosage ranging from about 100 to about 400mg/m .
  • the amount of tagged protein in formulations administered to a subject will vary between wide limits, depending upon the location, source, identity, extent and severity of the cancer, the age and condition of the individual to be treated, etc. A physician will ultimately determine appropriate dosages to be used.
  • the present invention relates to methods of treating a subject having cancer, comprising administering to a subject in need of treatment one or more formulations of tagged proteins, wherein the tagged proteins bind a cancer cell, and administering one or more therapeutically-effective populations of AT-CAR-expressing T cells, wherein the AT-CAR- expressing T cells bind the tagged proteins and induce cancer cell death.
  • cancer is intended to be broadly interpreted and it encompasses all aspects of abnormal cell growth and/or cell division.
  • carcinoma including but not limited to adenocarcinoma, squamous cell carcinoma, adenosquamous carcinoma, anaplastic carcinoma, large cell carcinoma, small cell carcinoma, and cancer of the skin, breast, prostate, bladder, vagina, cervix, uterus, liver, kidney, pancreas, spleen, lung, trachea, bronchi, colon, small intestine, stomach, esophagus, gall bladder; sarcoma, including but not limited to chondrosarcoma, Ewing's sarcoma, malignant hemangioendothelioma, malignant schwannoma, osteosarcoma, soft tissue sarcoma, and cancers of bone, cartilage, fat, muscle, vascular, and hematopoietic tissues;
  • lymphoma and leukemia including but not limited to mature B cell neoplasms, such as chronic lymphocytic leukemia/small lymphocytic lymphoma, B-cell prolymphocytic leukemia, lymphomas, and plasma cell neoplasms, mature T cell and natural killer (NK) cell neoplasms, such as T cell prolymphocytic leukemia, T cell large granular lymphocytic leukemia, aggressive NK cell leukemia, and adult T cell leukemia/lymphoma, Hodgkin lymphomas, and
  • lymphoproliferative disorders germ cell tumors, including but not limited to testicular and ovarian cancer; blastoma, including but not limited to hepatoblastoma, medulloblastoma, nephroblastoma, neuroblastoma, pancreatoblastoma, leuropulmonary blastoma and retinoblastoma.
  • germ cell tumors including but not limited to testicular and ovarian cancer
  • blastoma including but not limited to hepatoblastoma, medulloblastoma, nephroblastoma, neuroblastoma, pancreatoblastoma, leuropulmonary blastoma and retinoblastoma.
  • blastoma including but not limited to hepatoblastoma, medulloblastoma, nephroblastoma, neuroblastoma, pancreatoblastoma, leuropulmonary blastoma and retinoblastoma.
  • the terms “treat”, “treating”, and “treatment” have their ordinary and customary meanings, and include one or more of: blocking, ameliorating, or decreasing in severity and/or frequency a symptom of cancer in a subject, and/or inhibiting the growth, division, spread, or proliferation of cancer cells, or progression of cancer (e.g., emergence of new tumors) in a subject.
  • Treatment means blocking, ameliorating, decreasing, or inhibiting by about 1% to about 100% versus a subject in which the methods of the present invention have not been practiced.
  • the blocking, ameliorating, decreasing, or inhibiting is about 100%, 99%, 98%, 97%, 96%, 95%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 5% or 1% versus a subject in which the methods of the present invention have not been practiced.
  • Administration frequencies of both formulations comprising populations of AT-CAR- expressing T cells and formulations of tagged proteins will vary depending on factors that include the disease being treated, the elements comprising the AT-CAR-expressing T cells and the tagged proteins, and the modes of administration.
  • Each formulation may be independently administered 4, 3, 2 or once daily, every other day, every third day, every fourth day, every fifth day, every sixth day, once weekly, every eight days, every nine days, every ten days, bi-weekly, monthly and bi-monthly.
  • the formulation(s) of tagged proteins and the population(s) of AT-CAR-expressing T cells may be administered to a subject in any order.
  • the formulation(s) of tagged proteins may be administered to a subject before, after or concurrently with the population(s) of AT-CAR- expressing T cells.
  • the administration can be staggered. For example, a first formulation of tagged proteins can be administered, followed by a first population of AT-CAR-expressing T cells, which is then followed by a second formulation of tagged proteins and then a second population of AT-CAR- expressing T cells.
  • the present invention also includes methods whereby a population of AT-CAR- expressing T cells is coated with tagged proteins prior to administration of the AT-CAR- expressing T cells to the subject.
  • the subject receiving treatment is a human or non-human animal, e.g., a non-human primate, bird, horse, cow, goat, sheep, a companion animal, such as a dog, cat or rodent, or other mammal.
  • a human or non-human animal e.g., a non-human primate, bird, horse, cow, goat, sheep, a companion animal, such as a dog, cat or rodent, or other mammal.
  • the invention also provides a kit comprising one or more containers filled with one or more populations of AT-CAR-expressing T cells and one or more formulations of tagged proteins.
  • the kit may also include instructions for use.
  • Associated with the kit may further be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • mice were purchased from the Jackson Laboratory (Bar Harbor, ME, USA), housed in the University of Maryland, Baltimore, specific pathogen-free animal facility and used as recipients for adoptive immunotherapy.
  • the human EGFR+ colon adenocarcinoma cell line SW480 (ATCC, Manassas, VA) was maintained in Dulbecco's modified Eagle's medium (DMEM) (GIBCO brand; Invitrogen, Carlsbad, CA, USA) supplemented with 10% heat inactivated fetal bovine serum (Gemini Bio-Products, West Sacramento, CA, USA), 2 mM L-glutamine (GIBCO brand; Invitrogen) and 1% penicillin- streptomycin (GIBCO brand; Invitrogen).
  • DMEM Dulbecco's modified Eagle's medium
  • the EGFR+ HER2+ pancreatic adenocarcinoma cell line Pane 6.03 was kindly provided by Dr. Elizabeth Jaffee (Sidney Kimmel Cancer Center at Johns Hopkins), and cultured in RPMI 1640 medium (GIBCO brand; Invitrogen) supplemented with 20% FBS, 1% MEM non-essential amino acids (GIBCO brand; Invitrogen), 1% sodium pyruvate (GIBCO brand; Invitrogen), 2 mM L- glutamine, 1% penicillin-streptomycin and 100 IU/ml insulin.
  • RPMI 1640 medium Gibcorice, fetal bovine serum
  • FBS 1% MEM non-essential amino acids
  • 1% sodium pyruvate GBCO brand
  • 2 mM L- glutamine 1% penicillin-streptomycin
  • 100 IU/ml insulin The HER-2+ breast
  • adenocarcinoma cell line AU565 was cultured in RPMI 1640 medium supplemented with 10% FBS, 2 mM L-glutamine and 1% penicillin- streptomycin.
  • the Phoenix Ampho packaging cell line was purchased from Orbigen (San Diego, CA, USA) and maintained in D10 medium containing DMEM, 10% FBS, 1% sodium pyruvate, 2 mM L-glutamine and 1% penicillin-streptomycin.
  • the surface expression of EGFR or HER2 on SW480, Pane 6.03 and AU565 cell lines was determined by flow cytometry using FITC-conjugated Cetuximab (Ctx) or FITC-conjugated Herceptin (Her2).
  • Retroviral vector backbone pMSGVl-CD8- 28BBZ (Hughes M.S. et al., Transfer of a TCR gene derived from a patient with a marked antitumor response conveys highly active T-cell effector functions.
  • Hum Gene Ther 2005 Apr;16(4):457-72) was a kind gift from Dr. Richard Morgan (National Cancer Institute) and is derived from pMSGV (murine stem cell virus-based splice-gag vector).
  • Figure 1A shows schematically representation of the vector construct, and the order of placement of components in-frame from the 5' to the 3' ends.
  • the mouse scFv against FITC is referred to aFrfC-CAR and is linked to the hinge and transmembrane regions of the human CD8a chain (nucleotide sequence 1271-1519, Genbank NM 001768.6), and the cytoplasmic regions of the human CD28
  • aFITC-CAR sequence was synthesized by BlueHeron (Bothell, WA).
  • PBMC peripheral blood mononuclear cells
  • Isolated PBMC were cultured at 3 x 10 6 per well in 24- well tissue culture plates in AIM V medium (GIBCO brand; Invitrogen) supplemented with 5% human AB serum (Sigma- Aldrich), 1% MEM non-essential amino acids, 1% penicillin-streptomycin and 100 U/ml recombinant human IL-2 (BioLegend, San Diego, CA, USA), and activated with 50 ng/ml OKT3 (eBioscience, San Diego, CA, USA). Two days later, cells were collected for retroviral transduction.
  • Transductions were conducted as previously described (Johnson et al. Blood 114, 535-546 (2009)). Briefly, approximately 2.5 ml of retroviral supernatant were added to each coated well followed by centrifugation at 2000g for 2 h at 32 °C. 1.5 ml of viral supernatant was removed and 1 x 10 6 (0.5 ml) activated PBMC were added to each well in the presence of 100 U/ml IL-2. Plates were centrifugated at lOOOg for 10 min, and then incubated overnight at 37 °C. After transduction, cells were washed and maintained in the presence of IL-2 (lOOU/ml) and used in experiments five days after transduction.
  • IL-2 lOOU/ml
  • aFrfC-CAR transduced human T cells The surface expression of aFrfC-CAR on transduced human T cells was determined by flow cytometry after staining cells with CD3 or CD8 and FITC-conjugated cetuximab. In some experiments, aFrfC-CAR transduced T cells were stained with FITC-conjugated dextran beads. Cells stained with FITC-conjugated purified human IgG (Invitrogen) were used as an isotype control. [0068] T cell proliferation assay, cytokine and chemokine production assay.
  • 1 x 10 5 T cells were cultured in 96-well round-bottom plates coated with cetuximab, FITC-conjugated cetuximab, or FITC-Dextran for 72 h.
  • SW480 cells were pulsed with the indicated concentrations of antibodies for 1 h at 37 °C, washed 3 times.
  • 1 x 10 5 effector T cells and 1 x 10 5 tumor cells were co-cultured in 200 ⁇ of culture volume in 96-well round-bottom plates for 72 h.
  • 200,000 T cells were co-cultured with SW480 (50,000) colon cancer cells which had been pulsed with FITC-Ctx or Ctx (0.5 ⁇ g/l x 10 6 cells). Three days later proliferation was determined by [ H]thymidine uptake ( ⁇ SD) or cytokine and chemokine production was measured using a Milliplex array.
  • Cytotoxicity assay Cytotoxic activity against tumor target cells was measured using a standard 51 Cr release assay. 1 x 10 6 target cells were labeled with 200 ⁇ of 51 Cr for 2 h at 37°C, washed 3 times, and pulsed with anti-human antibodies for 1 h at 37 °C. 1 x 10 4 labeled target cells were then co-cultured with decreasing numbers of effector T cells at the indicated effector to target (E:T) ratios in 200 ⁇ of culture volume in 96-well round-bottom plates. Target cells incubated in medium alone were used to determined spontaneous 51 Cr release, and maximal release was determined by incubating labeled target cells in 10% Triton X-100.
  • % specific lysis (test release - spontaneous release) / (maximal release - spontaneous release) x 100. All tests were performed in triplicate wells and results are shown as mean +SD.
  • mice were injected intraperitoneally (i.p.) with FITC-Ctx or Ctx (25 ⁇ g/mouse).
  • mice were injected intravenous (i.v.) with 5 x 10 6 aFITC-CAR transduced human T cells.
  • mice were injected i.p. with antibodies (25 ⁇ g/mouse) weekly for three weeks.
  • Tumor area was measured with digital calipers in a blinded manner two to three times per week, and tumor sizes (mm") were calculated by perpendicular measurement by longitudinal diameter. Mice were euthanized when tumor sizes reach 200 mm or if mice became moribund or had trouble ambulating. All experiments were performed independently at least twice with similar results. Survival data were analyzed with the exact long-rank test.
  • aFITC-CAR or control (mock transduced) T cells were activated using titrating concentrations of plate-bound FITC-Ctx, unbound cetuximab, or FITC-Dex beads.
  • aFITC-CAR T cells proliferated vigorously and in a dose dependent manner following stimulation with FrfC- Ctx and FITC-Dex but did not divide in response to stimulation with Ctx alone ( Figure 1C, left panel).
  • control T cells did not proliferate to FITC-Ctx or FITC-Dex ( Figure 1C, right panel).
  • T cells were also co-cultured with EGFR + colon cancer cells (SW480) which had been stained with titrating concentrations of FITC-Ctx. FrfC reactivity by aFITC-CAR T cells was demonstrated by their ability to divide following activation with FITC-Ctx-stained cancer cells, Figure ID. However, proliferation was similar to that of control at the lowest
  • aFITC-CAR T cells were cultured together with FITC-Ctx-stained SW480 colon cancer cells at various effector to target ratios.
  • aFITC- CAR T cells lysed SW480 colon cancer cells at effector to target ratios as low as one T cell to twenty target cells ( Figure IE, left panel) but did not lyse cancer cells labeled with FITC-mouse IgG ( Figure IE, right panel).
  • control T cells did not show an appreciable level of cytolytic activity against FITC-Ctx-labeled or FITC-IgG-labeled SW480 cancer cells ( Figure IE).
  • CcFITC-CAR T cells were co-cultured with pancreatic cancer cells (Pane 6.03) stained with FITC-Ctx or stained with FITC-Herceptin (anti-Her-2 mAb; FITC-Her2) and breast cancer cells (AU565) stained with FITC-Her2.
  • ccFITC-CAR T cells efficiently and specifically lysed cancer cells stained with FITC-Ctx or FITC-Her2 ( Figure IF).
  • ccFITC-CAR T cells produced a wide array of cytokines beneficial to T cell survival, expansion and chemotaxis (Table 1).
  • the background cytokine levels produced by control T cells activated with FITC-Ctx- or Ctx-stained cancer cells were identical to one another.
  • Cytokines and chemokine production by aFrfC-CAR T cells HLA-A2 + PBMCs were activated with anti-CD3 antibodies in the presence of IL-2 followed by transduction with aFFTC- CAR retrovirus.
  • aFITC-CAR functionality on transduced was determined by their ability to produce cytokines and chemokines following activation with SW480colon cancer cells that were stained with FITC-Ctx or Ctx.
  • the levels of various cytokines and chemokines produced by a- FITC-CAR T cells were measured 72 hours after stimulation using a Milliplex
  • mice receiving aFITC-CAR T cells plus FITC-Ctx succumbed to tumor challenge within 55 days of tumor implantation.
  • aFITC-CAR T cells were activated with CD3 mAb and IL-2, CAR T cells can display a shortened life despite receiving prosurvival signals from CD28 or 41BB (Sadelain et al. Curr. Opin. Immunol. 21, 215-223 (2009); Brentjens et al. Nat. Med. 9, 279-286 (2003)).
  • the presence of aFITC-CAR T cells in various tissues was assayed, including the spleen, liver, bone marrow, circulation and in tumor explants. aFITC-CAR T cells were found in all tissues analyzed.
  • aFITC-CAR T cells Approximately 10% of all human T cells detected in tumor explants were aFITC-CAR T cells, Figure 2B. Similar percentages were found in the other tissues (data not shown). However, the overall percentage of aFITC-CAR T cells at the time of organ collection (between days 38 and 45) was significantly lower than the starting percentage of 60% at infusion. It is also worth noting that 60% to 90% of the T cells recovered from mice were CD8 + , as compared with the starting percentage of 40-50%. These data suggest a potential survival advantage of CD8 + subset over CD4 + T cells in vivo. The frequency or role for CD4 T Regs was not examined but remains a viable option that merits further investigation.
  • aFrfC-CAR T cells might not have become sufficiently activated by antigen (FITC) perhaps due to anergy or other suppressive mechanisms.
  • Total T cells were enriched from the tumor explant and immediately reactivated using SW480 cells from tissue culture that were stained with FITC-Ctx or Ctx alone.
  • aFITC-CAR T cells proliferated ( Figure 2C) and produced various effector molecules and chemokines ( Figure 2D) following stimulation with FITC-Ctx-stained SW480 cells but did not respond to SW480 cells stained with Ctx.
  • aFITC-CAR T cells isolated from tumor explants were not sufficient to test their killing aptitude.
  • -FI C-CAR T cells are clearly capable of responding to FITC stimulation.
  • One advantage to the use of an anti-tag CAR T cells is the potential to use several FITC 'tagged' tumor-reactive antibodies simultaneously.
  • the use of CARs expressing scFvs specific for biotin or PE-conjugated antibodies would add to the diversity of anti-tag CARs.
  • mice receiving aFITC-CAR T cells plus Ctx succumbed to tumor challenge within 35 days of tumor implantation Figure 3B right panel. It is worth noting that the administration of FITC-Ctx via i.v., i.p., or intratumoral injection all resulted in antibody localization to the tumor (data not shown).
  • An alternative method to redirecting T cells to the tumor is to coat ccFITC-CAR T cells with FITC-Ctx prior to adaptive transfer.

Abstract

The present invention provides a universal, yet adaptable, anti-tag chimeric antigen receptor (AT-CAR) system which provides T cells with the ability and specificity to recognize and kill target cells, such as tumor cells, that have been marked by tagged antibodies. As an example, FITC-CAR-expressing T cells have been developed that specifically recognize various human cancer cells when those cells are bound by cancer-reactive FITC-labeled antibodies. The activation of FITC-CAR-expressing T cells is shown to induce efficient target lysis, T cell proliferation, and cytokine/chemokine production. The system can be used to treating subjects having cancer.

Description

UNIVERSAL ANTI-TAG CHIMERIC ANTIGEN RECEPTOR-EXPRESSING T CELLS
AND METHODS OF TREATING CANCER
STATEMENT OF FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT
[0001] This invention was made with government support under Grant No. R01CA140917- 01 and R01HL088954 awarded by the National Cancer Institute. The government has certain rights in the invention.
TECHNICAL FIELD
[0002] The invention relates to T cell-based anti-cancer therapeutics and methods of using the therapeutics in the treatment of cancer.
BACKGROUND OF INVENTION
[0003] The ability to make a universal yet versatile system to generate T cells that are capable of recognizing various types of cancers has important clinical implications for the use of T cell-based therapies. One current strategy incorporates the use of genetic engineering to express a chimeric antigen receptor (CAR) on T cells. The extracellular domain of a typical CAR consists of the VH and VL domains -single-chain fragment variable (scFv)- from the antigen binding sites of a monoclonal antibody. The scFv is linked to a flexible transmembrane domain followed by a tyrosine-based activation motif such as that from CD3ζ (Sadelain et al. Curr. Opin. Immunol. 21, 215-223 (2009); Gross et al. Proc. Natl. Acad. Sci. USA 86, 10024-10028 (1989); Ertl et al. Cancer Res. 71, 3175-3181 (2011). The so-called second and third generation CARs include additional activation domains from co- stimulatory molecules such as CD28 and CD137 (41BB) which serve to enhance T cell survival and proliferation. CAR T cells offer the opportunity to seek out and destroy cancer cells by recognizing tumor-associated antigens (TAA) expressed on their surface (Sadelain et al. Curr. Opin. Immunol. 21, 215-223 (2009)). As such, the recognition of a tumor cells occurs via an MHC-independent mechanism. Various preclinical and early-phase clinical trials highlight the efficacy of CAR T cells to treat cancer patients with solid tumors and hematopoietic malignancies (Kershaw et al. Clin. Cancer Res. 12, 6106-6115 (2006); Lamers et al. J. Clin. Oncol. 24, e20-e22 (2006); Morgan et al. Mol. Ther. 18, 843-851 (2010); Pule et al. Nat. Med. 14, 1264-1270 (2008); Till et al. Blood 112, 2261-2271 (2008)). [0004] Despite of the promise that CAR T cells might have in treating cancer patients there are several limitations to the generalized clinical application of CAR T cells. First, since no single tumor antigen is universally expressed by all cancer types, scFv in CAR needs to be constructed for each tumor antigen to be targeted. Second, the financial cost and labor-intensive tasks associated with identifying and engineering scFvs against a variety of tumor antigens poses a major challenge. Third, tumor antigens targeted by CAR could be down-regulated or mutated in response to treatment resulting in tumor evasion. Since current CAR T cells recognize only one target antigen, such changes in tumors negate the therapeutic effects. Therefore, the generation of CAR T cells capable of recognizing multiple tumor antigens is highly desired. Finally, CAR T cells react with target antigen weakly expressed on non-tumor cells, potentially causing severe adverse effects (Morgan et al. Mol. Ther. 18, 843-851 (2010)). To avoid such "on-target off-tumor" reaction, use of scFvs with higher specificity to tumor antigen is required. And although ongoing studies are focused on generating methods to shut off CAR T cells in vivo this system has yet to be developed and might pose additional inherent challenges.
[0005] Modifications to existing CAR T cell systems that address and overcome the hurdles currently preventing development of the systems into effective means of in vivo treatment are therefore needed.
BRIEF SUMMARY OF INVENTION
[0006] The present invention provides a universal, yet adaptable, anti-tag chimeric antigen receptor (AT-CAR)-expressing T cell system that fully addresses the deficiencies of current systems. The system uses a gene therapy platform to generate immune cells capable of recognizing various cancers types and that have broad and valuable clinical implications for the use of T cell-based therapies. As disclosed herein, a versatile AT-CAR system which grants T cells specificity to recognize and bind tagged proteins, such as antibodies, has been developed.
[0007] For example, and as further described herein, ccFITC-CAR-expressing human T cells have been developed that specifically recognize various human cancer cells when those cells are bound by cancer-reactive FITC-labeled antibodies. The activation of FITC-CAR-expressing T cells is shown to induce efficient target lysis, T cell proliferation, and cytokine/chemokine production in vitro and ex vivo. In vivo, aFITC-CAR-expressing T cells plus FITC-cetuximab (Ctx) are shown to delay colon cancer tumor establishment but lead to the selection of tumor- associated antigen (TAA)-negative cancer cells. Using a pancreatic tumor model with uniform TAA expression, ccFITC-CAR-expressing T cells were observed to eradicate an established tumor and prevent tumor growth. This 'off-the-shelf system advances existing CAR technology through its potential to target various tagged proteins in the treatment of cancer patients.
[0008] In certain embodiments, the invention is drawn to a method of treating cancer in a subject, comprising: (a) administering a formulation of tagged proteins to a subject in need of treatment, wherein the tagged proteins bind a cancer cell in the subject, and (b) administering a therapeutically-effective population of anti-tag chimeric antigen receptor (AT-CAR)-expressing T cells to the subject, wherein the AT-CAR-expressing T cells bind the tagged proteins and induce cancer cell death, thereby treating cancer in a subject.
[0009] In a related embodiment, the invention is drawn to a method of treating cancer in a subject, comprising: (a) administering one or more formulations of tagged proteins to a subject in need of treatment, wherein the tagged proteins bind a cancer cell in the subject, and (b) administering one or more therapeutically-effective populations of AT-CAR-expressing T cells to the subject, wherein the AT-CAR-expressing T cells bind the tagged proteins and induce cancer cell death, thereby treating cancer in a subject.
[0010] In a further related embodiment, the invention is drawn to a method of treating cancer in a subject, comprising: (a) administering at least two formulations of tagged proteins to a subject in need of treatment, wherein the tagged proteins bind a cancer cell in the subject, and (b) administering at least two therapeutically-effective populations of AT-CAR-expressing T cells to the subject, wherein the AT-CAR-expressing T cells bind the tagged proteins and induce cancer cell death, thereby treating cancer in a subject.
[0011] In particular aspects of the embodiments of the invention, the tag of each formulation of tagged proteins is the same or different and the tag is selected from the group consisting of fluorescein isothiocyanate (FITC), streptavidin, biotin, histidine, dinitrophenol, peridinin chlorophyll protein complex, green fluorescent protein, phycoerythrin (PE), horse radish peroxidase, palmitoylation, nitrosylation, alkalanine phosphatase, glucose oxidase, and maltose binding protein.
[0012] In particular aspects of the embodiments of the invention, the protein of each formulation of tagged proteins is the same or different and the protein is an antibody or an antigen-binding fragment thereof. In preferred aspects, the antibody or antigen-binding fragment thereof is cetuximab, nimotuzumab, panitumumab, retuximab, omalizumab, tositumomab, trastuzumab, gemtuzumab, alemtuzumab, bevacuzimab or an antigen-binding fragment of any one thereof.
[0013] In particular aspects of the embodiments of the invention, the AT-CAR of each population of AT-CAR-expressing T cells is the same or different and the AT-CAR comprises a tag-binding domain, a transmembrane domain, and an activation domain. In preferred aspects, the tag-binding domain is an antibody or an antigen-binding fragment thereof. In preferred aspects, the tag-binding domain specifically binds FITC, biotin, PE, histidine or streptavidin. In preferred aspects where the tag-binding domain is antigen-binding fragment, the antigen-binding fragment is a single chain variable fragment (scFv), such as a scFv that specifically binds FITC, biotin, PE, histidine or streptavidin. In preferred aspects the transmembrane domain is the hinge and transmembrane regions of the human CD8a chain. In preferred aspects, the activation domain comprises one or more of the cytoplasmic region of CD28, the cytoplasmic region of CD137 (41BB), OX40, HVEM, CD3^ and FcRs.
[0014] In particular aspects of the embodiments of the invention, the T cells of each population of AT-CAR-expressing T cells are the same or different and wherein the T cells are selected from the group consisting of T cells of any HLA -background from peripheral blood mononuclear cells (PBMC), T cells isolated from a tumor explant of the subject, and
intratumoral T cells of the subject.
[0015] In particular aspects of the embodiments of the invention, the T cells of each population of AT-CAR-expressing T cells consist of HLA-A2+ peripheral blood mononuclear cells (PBMC).
[0016] In particular aspects of the embodiments of the invention, the formulation(s) of tagged proteins are administered to the subject prior to administration of the therapeutically- effective population(s) of AT-CAR-expressing T cells.
[0017] In particular aspects of the embodiments of the invention, the formulation(s) of tagged proteins are administered to the subject concurrently with administration of the therapeutically-effective population(s) of AT-CAR-expressing T cells.
[0018] In particular aspects of the embodiments of the invention, the formulation(s) of tagged proteins are administered to the subject after administration of the therapeutically- effective population(s) of AT-CAR-expressing T cells. [0019] In particular aspects of the embodiments of the invention, the formulation(s) of tagged proteins and the therapeutically-effective population(s) of AT-CAR-expressing T cells are administered to the subject in any order.
[0020] In particular aspects of the embodiments of the invention, AT-CAR-expressing T cell binding to the tagged proteins induces cytolytic activation of the T cells.
[0021] In particular aspects of the embodiments of the invention, the subject is a human.
[0022] The foregoing has outlined rather broadly the features and technical advantages of the present invention in order that the detailed description of the invention that follows may be better understood. Additional features and advantages of the invention will be described herein, which form the subject of the claims of the invention. It should be appreciated by those skilled in the art that any conception and specific embodiment disclosed herein may be readily utilized as a basis for modifying or designing other formulations for carrying out the same purposes of the present invention. It should also be realized by those skilled in the art that such equivalent formulations do not depart from the spirit and scope of the invention as set forth in the appended claims. The novel features which are believed to be characteristic of the invention, both as to its organization and method of operation, together with further objects and advantages will be better understood from the following description when considered in connection with the accompanying figures. It is to be expressly understood, however, that any description, figure, example, etc. is provided for the purpose of illustration and description only and is by no means intended to define the limits the invention.
BRIEF DESCRIPTION OF DRAWINGS
[0023] Figure 1. Anti-FITC-CAR expression, characterization and in vitro functionality. (A) Diagram of the anti-FITC-CAR (ccFITC-CAR). Boxed diagram on left shows the genetic elements included in the polynucleotide engineered to express ccFITC-CAR, beginning with a 5'- long terminal repeat, and followed by the coding region for anti-FITC scFv, CD28
transmembrane domain, CD28, 4 IBB, zeta chain (ζ) and a 3 '-long terminal repeat. Figure on right shows the ccFITC-CAR polypeptide traversing the T cell membrane and position to bind a tumor-reactive antibody labeled with FITC. (B) HLA-A2+ PBMCs were activated with anti-CD3 antibodies in the presence of IL-2 followed by transduction with ccFITC-CAR retrovirus. ccFITC- CAR expression was determined by staining cells with CD3 and FITC-conjugated cetuximab (FITC-Ctx) or FITC-conjugated dextran beads (FITC-Dex). Cetuximab is an antibody with binding specificity for EGFR-expressing tumor cells. (C) ccFITC-CAR functionality on transduced and control T cells was examined in proliferation assays using plate-bound FITC-Ctx, FITC-Dex or Ctx. (D) The proliferation of FrfC-CAR T cells and control T cells was measured in response to stimulation with SW480 colon cancer cells stained with titrating concentrations of FITC-Ctx. (E) T cell cytotoxicity by ccFITC-CAR and control T cells was measured against SW480 colon cancer cells stained with FITC-Ctx or FITC-mouse IgG. (F) ccFITC-CAR
cytotoxicity was measured against Pane 6.03 stained with unlabeled Ctx and Her2 antibodies or stained with FITC-Ctx or FITC-Her2 at various effector to target ratios. Alternatively AU565 breast cancer cells were stained with FITC-Her2 or unlabled Her2 mAb. All data are
representative of three or more independent experiments each yielding identical trends.
[0024] Figure 2. ccFITC-CAR T cells delay tumor establishment but promote the growth of antigen-negative tumor cells. (A) SW480 human colon cancer cells were injected subcutaneously into NSG mice followed by injection with FITC-Ctx (i.p.) one day later. Twenty four hours after, 5xl06 ccFITC-CAR T cells were injected into the tail vein. FITC-Ctx was injected (i.p.) weekly for three weeks. Data are representative of two or three experiments (three or more mice per group). *, P < 0.02, ANOVA; n.s. not significant. (B) The percentage of CD3+ ccFITC-CAR positive and negative T cells was determined by flow cytometry. The average percentage of
FITC-CAR+ T cells prior to injection or from the tumor explant of four mice is shown. (C) Tumor explants were finely minced and cultured in trypsin for 2 hours at 37°C followed by T cell enrichment using a negative selection kit. T cells were co-cultured with SW480 colon cancer cells which had been pulsed with FITC-Ctx or Ctx (O^g/lxlO6 cells). Three days later proliferation was determined by [ H]thymidine uptake (+SD). (D) Alternatively, cytokine and chemokine production was measured using a Milliplex array 72 hours after stimulation. (E) EGFR expression on SW480 tumor explants or SW480 cells taken from tissue culture was examined by flow cytometry using FITC-Ctx.
[0025] Figure 3. ccFITC-CAR T cells prevent tumor growth and eradicate established tumors. (A) EGFR expression on the pancreatic human cancer cells (Pane 6.03) was determined by staining cells with FITC-Ctx or with control antibody FITC-mlgG and analyzed by flow cytometry. (B) In the prophylactic tumor model, Pane 6.03 cells were injected subcutaneously into NSG mice (n=5) followed by injection with FITC-Ctx (i.p.) one day later. Twenty four hours later 5xl06 aFITC-CAR T cells were injected into the tail vein. 25μg of FITC-Ctx or Ctx was injected (i.p.) weekly for three weeks. Tumor growth and mouse survival is shown. (C) In the therapeutic model Pane 6.03 tumors were grown to sizes between 3-10 mm and then injected (i.p.) with 25 μg of FITC-Ctx or Ctx every week for three weeks. One day after the first injection of FITC-Ctx, mice were administered 5xl06 aFrfC-CAR T cells via tail vein injection. *, P < 0.02, ANOVA; n.s. not significant.
DETAILED DESCRIPTION OF THE INVENTION
/. Definitions
[0026] Unless otherwise noted, technical terms are used according to conventional usage. Definitions of common terms in molecular biology may be found, for example, in Benjamin Lewin, Genes VII, published by Oxford University Press, 2000 (ISBN 019879276X); Kendrew et al. (eds.); The Encyclopedia of Molecular Biology, published by Blackwell Publishers, 1994 (ISBN 0632021829); and Robert A. Meyers (ed.), Molecular Biology and Biotechnology: a Comprehensive Desk Reference, published by Wiley, John & Sons, Inc., 1995 (ISBN
0471186341); and other similar technical references.
[0027] As used herein, "a" or "an" may mean one or more. As used herein when used in conjunction with the word "comprising," the words "a" or "an" may mean one or more than one. As used herein "another" may mean at least a second or more. Furthermore, unless otherwise required by context, singular terms include pluralities and plural terms include the singular.
[0028] As used herein, "about" refers to a numeric value, including, for example, whole numbers, fractions, and percentages, whether or not explicitly indicated. The term "about" generally refers to a range of numerical values (e.g., +/- 5-10% of the recited value) that one of ordinary skill in the art would consider equivalent to the recited value (e.g., having the same function or result). In some instances, the term "about" may include numerical values that are rounded to the nearest significant figure.
//. The Present Invention
[0029] An adaptable generation of CARs has been developed which allows for the widespread use of personalized T cell-based immunotherapy. Human T cells have been engineered to express an anti-FITC CAR-CD28-41BB-CD3 (referred to ccFITC-CAR). This platform takes advantage of the high affinity interaction between the anti-FITC scFv (on the cell's surface) and FITC as well as the ability to cheaply and easily conjugated FITC molecules (or other tags) to any anti-cancer-based monoclonal antibody such as cetuximab (anti-EGFR), retuximab (anti-CD20) and herceptin (anti-Her2) used to treat patients with various types of cancers. This system allows for extreme specificity to the antigen and is accompanied by robust effector function and proliferative capacity with no cross -reactivity to 'self-antigens' .
[0030] Effector cells. The effector cells used in the methods of the present invention may be autologous, syngeneic or allogeneic, with the selection dependent on the disease to be treated and the means available to do so. Suitable populations of effector cells that may be used in the methods include any immune cells with cytolytic activity, such as T cells. Exemplary sub- populations of T cells include, but are not limited to those expressing CD3+ including CD3+CD8+ T cells, CD3+CD4+ T cells, and NKT cells. In one aspect, the T cells are HLA-A2+ peripheral blood mononuclear cells (PBMC) but the T cells can be of any HLA background from PBMCs and utilized in an autologous, syngeneic or allogeneic system. T cells may also be isolated from any source, including from a tumor explant of the subject being treated or intratumoral T cells of the subject being treated. For the sake of convenience, the effector cells are commonly referred to herein as T cells, but it should be understood that any reference to T cells, unless otherwise indicated, is a reference to all effector cell types as defined herein.
[0031] Anti-tag chimeric antigen receptor (AT-CAR). The skilled artisan will appreciate that the anti-tag chimeric antigen receptor (AT-CAR) expressed by the T cells used in the methods of the present invention allow for great flexibility. The sole requirements for the AT-CARs used in the methods are (i) that the AT-CAR has binding specificity for a particular tag that can be conjugated to a protein (such as an antibody) that binds to a tumor- associated antigen (TAA), and (ii) that T cells can be engineered to express the AT-CAR. Additional features that are preferred, but not required, include (i) that the AT-CAR includes an activation domain that induces efficient target lysis upon T cell binding and activation, and (ii) the ability to replace the scFv portion of the AT-CAR with one of specificity to other tags, such as biotin or phycoerythrin, which can be conjugated to target (i.e., tumor) -reactive proteins such as antibodies to be used in vivo.
[0032] In particular aspects, the AT-CAR comprises three domains. The first domain is the tag-binding domain. This domain is typically present at the amino terminal end of the
polypeptide that comprises the AT-CAR. Locating the tag-binding domain at the amino terminus permits the tag-binding domain unfettered access to the tagged protein that is bound to the target cell. As used herein, the tag-binding domain is typically an antibody or an antigen-binding fragment thereof. The identity of the antibody or fragment is only limited by the identity of the tag of the tagged protein. For example, the antibodies may be obtained from any species of animal, though preferably from a mammal such as a human, simian, mouse, rat, rabbit, guinea pig, horse, cow, sheep, goat, pig, dog or cat. Preferably the antibodies are human or humanized antibodies. Nor is there a limitation on the particular class of antibody that may be used, including IgGl, IgG2, IgG3, IgG4, IgM, IgAl, IgA2, IgD and IgE antibodies. Antibody fragments include single-chain variable fragment (scFv), single chain antibodies, F(ab')2 fragments, Fab fragments, and fragments produced by an Fab expression library, with the only limitation being that the antibody fragments retain the ability to bind the selected tag.
[0033] The antibodies may also be polyclonal, monoclonal, or chimeric antibodies, such as where an antigen binding region (e.g., F(ab')2 or hypervariable region) of a non-human antibody is transferred into the framework of a human antibody by recombinant DNA techniques to produce a substantially human molecule. Antigen-binding fragments, such as scFv, may be prepared therefrom.
[0034] One advantage of the AT-CARs of the present invention is that they can be produced using commercially-available antibodies. Alternatively, antibodies to a selected tag may be produced. For the production of antibodies, various hosts including, but not limited to, goats, rabbits, rats, mice, humans, etc., can be immunized by injection with a particular protein or any portion, fragment, or oligopeptide that retains immunogenic properties of the protein.
Depending on the host species, various adjuvants can be used to increase the immunological response. Such adjuvants include, but are not limited to, detoxified heat labile toxin from E. coli, Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol. BCG (Bacillus Calmette-Guerin) and Corynebacterium parvum are also potentially useful adjuvants.
[0035] Antibodies and fragments thereof can be prepared using any technique that provides for the production of antibody molecules, such as by continuous cell lines in culture for monoclonal antibody production. Such techniques include, but are not limited to, the hybridoma technique originally described by Koehler and Milstein (Nature 256:495-497 (1975)), the human B-cell hybridoma technique (Kosbor et al., Immunol Today 4:72 (1983); Cote et al., Proc Natl. Acad. Sci 80:2026-2030 (1983)), and the EBV-hybridoma technique (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss Inc, New York N.Y., pp 77-96 (1985)).
[0036] Techniques developed for the production of "chimeric antibodies," i.e., the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can also be used (Morrison et al., Proc Natl. Acad. Sci 81:6851-6855 (1984); Neuberger et al., Nature 312:604-608(1984); Takeda et al., Nature 314:452-454(1985)). Alternatively, techniques described for the production of single chain antibodies, such as disclosed in U.S. Patent No. 4,946,778, incorporated herein by reference in its entirety, can be adapted to produce tag- specific single chain antibodies.
[0037] In one aspect, the tag-binding domain is a single-chain variable fragment (scFv). A scFv comprises the variable regions of the heavy (VH) and light chains (VL) of an antibody, typically linked via a short peptide of ten to about 25 amino acids. The linker can either connect the N-terminus of the VH with the C-terminus of the VL, or vice versa.
[0038] As indicated above, the binding specificity of the tag-binding domain will depend on the identity of the tag that is conjugated to the protein that is used to bind target cells. For example, when the tag is FITC (Fluorescein isothiocyanate), the tag-binding domain may constitute an anti-FITC scFv. Alternatively, when the tag is biotin or PE (phycoerythrin), the tag- binding domain may constitute an anti-biotin scFv or an anti-PE scFv.
[0039] The second domain is a transmembrane (TM) domain. The TM domain allows the CAR to be anchored into the cell membrane of the T cell. Exemplary TM domains include, but are not limited to, the hinge and transmembrane regions of the human CD8a chain.
[0040] The third domain, when present, is the T cell activation domain. This domain aids in T cell activation upon binding of the CAR to the tagged protein that is bound to the target cell. T cell activation includes induction of cytokine and chemokine production, as well as activation of the cytolytic activity of the cells. Exemplary T cell activation domains include, but are not limited to, the cytoplasmic regions of CD28, CD137 (41BB), OX40 and HVEM which serve to enhance T cell survival and proliferation; and CD3ζ and FcRs which induce T cell activation. One or more than one T cell activation domain may be included in the CAR, such as two, three, four or more T cell activation domains.
[0041] AT-CAR T cell production. T cells may be engineered to express AT-CARs by means readily known to the skilled artisan. Generally, a polynucleotide vector is constructed that encodes the AT-CAR and the vector is transfected into a population of T cells. The cells are then grown under conditions promoting expression of the AT-CAR by the T cells. Successful transfection (or transduction which refers to viral-mediated gene integration) and display of AT- CARs by T cells is conducted via conventional means, some of which are disclosed in the Examples herein.
[0042] In one aspect, T cells may be engineered to produce AT-CARs by first constructing a retroviral vector encoding a selected AT-CAR. An exemplary retroviral vector includes, but is not limited to, the vector backbone pMSGVl-CD8-28BBZ, which is derived from pMSGV (murine stem cell virus-based splice-gag vector). DNA sequencing can be used to confirm proper construction of the vector before transfection of T cells. Retroviral transduction may be performed using known techniques, such as that of Johnson et al. {Blood 114, 535-546 (2009)). The surface expression of AT-CAR on transduced T cells may be determined, for example, by flow cytometry after staining cells with tag-conjugated protein or tag-conjugated dextran beads. The tag portion of the protein or beads will be bound by the tag-binding domain of the CAR expressed by the cells.
[0043] AT-CAR T cell administration. Populations of AT-CAR-expressing T cells may be formulated for administered to a subject using techniques known to the skilled artisan.
Formulations comprising populations of AT-CAR-expressing T cells may include
pharmaceutically acceptable excipient(s). Excipients included in the formulations will have different purposes depending, for example, on the nature of the tag-binding domain comprising the AT-CAR, the subpopulation of T cells used, and the mode of administration. Examples of generally used excipients include, without limitation: saline, buffered saline, dextrose, water-for- infection, glycerol, ethanol, and combinations thereof, stabilizing agents, solubilizing agents and surfactants, buffers and preservatives, tonicity agents, bulking agents, and lubricating agents. The formulations comprising populations of AT-CAR-expressing T cells will typically have been prepared and cultured in the absence of any non-human components, such as animal serum (e.g., bovine serum albumin).
[0044] A formulation may include one population of AT-CAR-expressing T cells, or more than one, such as two, three, four, five, six or more populations of AT-CAR-expressing T cells. The different populations of AT-CAR T cells can vary based on the identity of the tag-binding domain, the identity of the activation domain, the identity of the subpopulation of T cells, or a combination thereof. For example, a formulation may comprise populations of AT-CAR- expressing T cells that recognize and bind to one, or more than one, such as two, three, four, five, six or more different tagged proteins. As an example, a formulation may comprise populations of AT-CAR-expressing T cells that recognize and bind FITC, biotin and PE. Thus, in this example, the formulation comprises three different populations of AT-CAR-expressing T cells that recognize and bind cells tagged by FITC-conjugated antibodies, biotin-conjugated antibodies, and PE-conjugated antibodies. This formulation would therefore comprise aFrfC-CAR- expressing T cells, abiotin-CAR-expressing T cells and αΡΕ-CAR-expressing T cells.
[0045] The formulations comprising population(s) of AT-CAR T cells may be administered to a subject using modes and techniques known to the skilled artisan. Exemplary modes include, but are not limited to, intravenous injection. Other modes include, without limitation,
intratumoral, intradermal, subcutaneous (s.c, s.q., sub-Q, Hypo), intramuscular (i.m.), intraperitoneal (i.p.), intra-arterial, intramedulary, intracardiac, intra- articular (joint),
intrasynovial (joint fluid area), intracranial, intraspinal, and intrathecal (spinal fluids). Any known device useful for parenteral injection or infusion of the formulations can be used to effect such administration.
[0046] The formulations comprising population(s) of AT-CAR-expressing T cells that are administered to a subject comprise a number of AT-CAR-expressing T cells that is effective for the treatment and/or prophylaxis of the specific indication or disease. Thus, therapeutically- effective populations of AT-CAR-expressing T cells are administered to subjects when the methods of the present invention are practiced. In general, formulations are administered that comprise between about 1 x 104 and about 1 x 1010 AT-CAR-expressing T cells. In most cases, the formulation will comprise between about 1 x 105 and about 1 x 109 AT-CAR-expressing T cells, from about 5 x 105 to about 5 x 108 AT-CAR-expressing T cells, or from about 1 x 106 to about 1 x 10' AT-CAR -expressing T cells. However, the number of AT-CAR-expressing T cells administered to a subject will vary between wide limits, depending upon the location, source, identity, extent and severity of the cancer, the age and condition of the individual to be treated, etc. A physician will ultimately determine appropriate dosages to be used.
[0047] Tagged proteins. Tagged proteins are administered to a subject prior to, or concurrent with, or after administration of the AT-CAR-expressing T cells. The tagged proteins bind to target cells in the subject. In general, the "protein" portion of the tagged protein is the portion of the molecule that binds to the target cell. For example, the protein may be an antibody that binds to a tumor- associated antigen (TAA) or a tumor specific antigen (TSA) expressed by the target cell. However, the "protein" may be any molecule that binds to a target cell. Exemplary proteins include, but are not limited to, anti-cancer-based monoclonal antibodies such as cetuximab (anti- EGFR), nimotuzumab (anti-EGFR), panitumumab (anti-EGFR), retuximab (anti-CD20), omalizumab (anti-CD20), tositumomab (anti-CD20), trastuzumab (anti-Her2), gemtuzumab (anti-CD33), alemtuzumab (anti-CD52), and bevacuzimab (anti-VEGF).
[0048] The "tag" portion of the tagged protein is only constrained by being a molecular that can be recognized and specifically bound by the AT-CAR, specifically, the tag-binding domain of the AT-CAR. Exemplary tags include, but are not limited to, fluorescein isothiocyanate (FITC), dinitrophenol, peridinin chlorophyll protein complex, green fluorescent protein, biotin, phycoerythrin (PE), histidine, streptavidin, horse radish peroxidase, palmitoylation, nitrosylation, alkalanine phosphatase, glucose oxidase, Glutathione S -transferase, maltose binding protein, and any types of fluorescent materials including quantum dot nanocrystals.
[0049] Thus, in some aspects, the tagged proteins include FITC-conjugated antibodies, biotin-conjugated antibodies, PE-conjugated antibodies, histidine-conjugated antibodies and streptavidin-conjugated antibodies, where the antibody binds to a TAA or a TSA expressed by the target cells. For example, the tagged proteins of the present invention include, but are not limited to, FITC-conjugated cetuximab, FITC-conjugated retuximab, FITC-conjugated herceptin, biotin-conjugated cetuximab, biotin-conjugated retuximab, biotin-conjugated herceptin, PE- conjugated cetuximab, PE-conjugated retuximab, PE-conjugated herceptin, histidine-conjugated cetuximab, histidine-conjugated retuximab, histidine-conjugated herceptin, streptavidin- conjugated cetuximab, streptavidin-conjugated retuximab, and streptavidin-conjugated herceptin. Alternatively, the AT-CAR cells can be redirected to target and/or destroy vascular cells feeding the tumor. For example, T cells expressing aFITC-VEGF as the AT-CAR can target endothelial vascular cells to which FITC-tagged VEGF is bound, where the FITC-tagged VEGF is bound by the VEGF receptor.
[0050] In some aspects, a protein lacking a tag may be used as the tagged protein. For example, a naked (tagless) protein (such as an antibody) that binds to a TAA or a TSA on a target cell may be used as the tagged protein. Under such circumstances, the AT-CAR will recognize and specifically bind the protein. As an example, the tag-binding domain may be an antibody or antigen-binding fragment thereof that recognizes and binds a second antibody, where the second antibody functions as the tagged protein and where the second antibody lacks a tag.
[0051] The tag may be conjugated to the proteins using techniques such as chemical coupling and chemical cross -linkers. Alternatively, polynucleotide vectors can be prepared that encode the tagged proteins as fusion proteins. Cell lines can then be engineered to express the tagged proteins, and the tagged proteins can be isolated from culture media, purified and used in the methods disclosed herein.
[0052] The tagged proteins may be formulated for administered to a subject using techniques known to the skilled artisan. Formulations of the tagged proteins may include pharmaceutically acceptable excipient(s). Excipients included in the formulations will have different purposes depending, for example, on the nature of the tag, the protein, and the mode of administration. Examples of generally used excipients include, without limitation: saline, buffered saline, dextrose, water-for-infection, glycerol, ethanol, and combinations thereof, stabilizing agents, solubilizing agents and surfactants, buffers and preservatives, tonicity agents, bulking agents, and lubricating agents.
[0053] A formulation of tagged proteins may include one type of tagged protein, or more than one, such as two, three, four, five, six or more types of tagged proteins. The different types of tagged proteins can vary based on the identity of the tag, the identity of the protein, or both. For example, a formulation comprising three types of tagged protein might include FITC- conjugated cetuximab, FITC-conjugated rituximab and FITC-conjugated herceptin, or FITC- conjugated cetuximab, biotin-conjugated cetuximab, and PE-conjugated cetuximab. [0054] The tagged proteins may be administered to a subject using modes and techniques known to the skilled artisan. Exemplary modes include, but are not limited to, intravenous, intraperitoneal, and intratumoral injection. Other modes include, without limitation, intradermal, subcutaneous (s.c, s.q., sub-Q, Hypo), intramuscular (i.m.), intra-arterial, intramedulary, intracardiac, intra-articular (joint), intrasynovial (joint fluid area), intracranial, intraspinal, and intrathecal (spinal fluids). Any known device useful for parenteral injection or infusion of the formulations can be used to effect such administration.
[0055] Formulations comprising the tagged proteins are administered to a subject in an amount which is effective for treating and/or prophylaxis of the specific indication or disease. In general, formulations comprising at least about 0.1 mg/kg to about 100 mg/kg body weight of the tagged proteins are administered to a subject in need of treatment. In most cases, the dosage is from about 1 mg/kg to about 10 mg/kg body weight of the tagged proteins daily, taking into account the routes of administration, symptoms, etc. As an example, tagged-bevacizumab is administered in a dosage of from about 2.5 to about 5 mg kg. As a further example, tagged- cetuximab is administered in a dosage ranging from about 100 to about 400mg/m . However, the amount of tagged protein in formulations administered to a subject will vary between wide limits, depending upon the location, source, identity, extent and severity of the cancer, the age and condition of the individual to be treated, etc. A physician will ultimately determine appropriate dosages to be used.
[0056] Cancer. The present invention relates to methods of treating a subject having cancer, comprising administering to a subject in need of treatment one or more formulations of tagged proteins, wherein the tagged proteins bind a cancer cell, and administering one or more therapeutically-effective populations of AT-CAR-expressing T cells, wherein the AT-CAR- expressing T cells bind the tagged proteins and induce cancer cell death. The term "cancer" is intended to be broadly interpreted and it encompasses all aspects of abnormal cell growth and/or cell division. Examples include: carcinoma, including but not limited to adenocarcinoma, squamous cell carcinoma, adenosquamous carcinoma, anaplastic carcinoma, large cell carcinoma, small cell carcinoma, and cancer of the skin, breast, prostate, bladder, vagina, cervix, uterus, liver, kidney, pancreas, spleen, lung, trachea, bronchi, colon, small intestine, stomach, esophagus, gall bladder; sarcoma, including but not limited to chondrosarcoma, Ewing's sarcoma, malignant hemangioendothelioma, malignant schwannoma, osteosarcoma, soft tissue sarcoma, and cancers of bone, cartilage, fat, muscle, vascular, and hematopoietic tissues;
lymphoma and leukemia, including but not limited to mature B cell neoplasms, such as chronic lymphocytic leukemia/small lymphocytic lymphoma, B-cell prolymphocytic leukemia, lymphomas, and plasma cell neoplasms, mature T cell and natural killer (NK) cell neoplasms, such as T cell prolymphocytic leukemia, T cell large granular lymphocytic leukemia, aggressive NK cell leukemia, and adult T cell leukemia/lymphoma, Hodgkin lymphomas, and
immunodeficiency-associated lymphoproliferative disorders; germ cell tumors, including but not limited to testicular and ovarian cancer; blastoma, including but not limited to hepatoblastoma, medulloblastoma, nephroblastoma, neuroblastoma, pancreatoblastoma, leuropulmonary blastoma and retinoblastoma. The term also encompasses benign tumors.
[0057] As used herein, the terms "treat", "treating", and "treatment" have their ordinary and customary meanings, and include one or more of: blocking, ameliorating, or decreasing in severity and/or frequency a symptom of cancer in a subject, and/or inhibiting the growth, division, spread, or proliferation of cancer cells, or progression of cancer (e.g., emergence of new tumors) in a subject. Treatment means blocking, ameliorating, decreasing, or inhibiting by about 1% to about 100% versus a subject in which the methods of the present invention have not been practiced. Preferably, the blocking, ameliorating, decreasing, or inhibiting is about 100%, 99%, 98%, 97%, 96%, 95%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 5% or 1% versus a subject in which the methods of the present invention have not been practiced.
[0058] Administration frequencies of both formulations comprising populations of AT-CAR- expressing T cells and formulations of tagged proteins will vary depending on factors that include the disease being treated, the elements comprising the AT-CAR-expressing T cells and the tagged proteins, and the modes of administration. Each formulation may be independently administered 4, 3, 2 or once daily, every other day, every third day, every fourth day, every fifth day, every sixth day, once weekly, every eight days, every nine days, every ten days, bi-weekly, monthly and bi-monthly.
[0059] The duration of treatment will be based on the disease being treated and will be best determined by the attending physician. However, continuation of treatment is contemplated to last for a number of days, weeks, or months. [0060] The present invention offers flexibility in the methods of treatment, and as a result, the formulation(s) of tagged proteins and the population(s) of AT-CAR-expressing T cells may be administered to a subject in any order. Thus, the formulation(s) of tagged proteins may be administered to a subject before, after or concurrently with the population(s) of AT-CAR- expressing T cells. Alternatively, where more than one formulation of tagged proteins and/or more than one population of AT-CAR-expressing T cells are administered to a subject, the administration can be staggered. For example, a first formulation of tagged proteins can be administered, followed by a first population of AT-CAR-expressing T cells, which is then followed by a second formulation of tagged proteins and then a second population of AT-CAR- expressing T cells.
[0061] The present invention also includes methods whereby a population of AT-CAR- expressing T cells is coated with tagged proteins prior to administration of the AT-CAR- expressing T cells to the subject.
[0062] In each of the embodiments of the present invention the subject receiving treatment is a human or non-human animal, e.g., a non-human primate, bird, horse, cow, goat, sheep, a companion animal, such as a dog, cat or rodent, or other mammal.
[0063] The invention also provides a kit comprising one or more containers filled with one or more populations of AT-CAR-expressing T cells and one or more formulations of tagged proteins. The kit may also include instructions for use. Associated with the kit may further be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
IV. Examples
[0064] Mice and cell lines. NOD-scid IL2RgammanuU (NSG) mice were purchased from the Jackson Laboratory (Bar Harbor, ME, USA), housed in the University of Maryland, Baltimore, specific pathogen-free animal facility and used as recipients for adoptive immunotherapy.
Experiments were reviewed and approved by the University of Maryland Baltimore Institutional Animal Care and Use Committee. The human EGFR+ colon adenocarcinoma cell line SW480 (ATCC, Manassas, VA) was maintained in Dulbecco's modified Eagle's medium (DMEM) (GIBCO brand; Invitrogen, Carlsbad, CA, USA) supplemented with 10% heat inactivated fetal bovine serum (Gemini Bio-Products, West Sacramento, CA, USA), 2 mM L-glutamine (GIBCO brand; Invitrogen) and 1% penicillin- streptomycin (GIBCO brand; Invitrogen). The EGFR+ HER2+ pancreatic adenocarcinoma cell line Pane 6.03 was kindly provided by Dr. Elizabeth Jaffee (Sidney Kimmel Cancer Center at Johns Hopkins), and cultured in RPMI 1640 medium (GIBCO brand; Invitrogen) supplemented with 20% FBS, 1% MEM non-essential amino acids (GIBCO brand; Invitrogen), 1% sodium pyruvate (GIBCO brand; Invitrogen), 2 mM L- glutamine, 1% penicillin-streptomycin and 100 IU/ml insulin. The HER-2+ breast
adenocarcinoma cell line AU565 (ATCC) was cultured in RPMI 1640 medium supplemented with 10% FBS, 2 mM L-glutamine and 1% penicillin- streptomycin. The Phoenix Ampho packaging cell line was purchased from Orbigen (San Diego, CA, USA) and maintained in D10 medium containing DMEM, 10% FBS, 1% sodium pyruvate, 2 mM L-glutamine and 1% penicillin-streptomycin. The surface expression of EGFR or HER2 on SW480, Pane 6.03 and AU565 cell lines was determined by flow cytometry using FITC-conjugated Cetuximab (Ctx) or FITC-conjugated Herceptin (Her2).
[0065] Construction of retroviral vector. The retroviral vector backbone pMSGVl-CD8- 28BBZ (Hughes M.S. et al., Transfer of a TCR gene derived from a patient with a marked antitumor response conveys highly active T-cell effector functions. Hum Gene Ther 2005 Apr;16(4):457-72) was a kind gift from Dr. Richard Morgan (National Cancer Institute) and is derived from pMSGV (murine stem cell virus-based splice-gag vector). Figure 1A shows schematically representation of the vector construct, and the order of placement of components in-frame from the 5' to the 3' ends. The mouse scFv against FITC is referred to aFrfC-CAR and is linked to the hinge and transmembrane regions of the human CD8a chain (nucleotide sequence 1271-1519, Genbank NM 001768.6), and the cytoplasmic regions of the human CD28
(nucleotide sequence 760-882, Genbank NM 006139.2), 4-1BB (nucleotide sequence 886-1026, Genbank NM 001561.5) and CD3^ (nucleotide sequence 299-637, Genbank NM 000734.3) molecules. The aFITC-CAR sequence was synthesized by BlueHeron (Bothell, WA).
[0066] The sequence was confirmed by DNA sequencing and has the following sequence : agttgcctgttaggttgttggtgctgatgttctggattcctgcttccagcagtgatgtcgtgatgacccaaactccactctccctgcctgtcagtct tggagatcaagcctccatctcttgcagatctagtcagagccttgtacacagtaatggaaacacctatttacgttggtacctgcagaagccagg ccagtctccaaaggtcctgatctacaaagtttccaaccgattttctggggtcccagacaggttcagtggcagtggatcagggacagatttcac actcaagatcagcagagtggaggctgaggatctgggagtttatttctgctctcaaagtacacatgttccgtggacgttcggtggaggcacca agctggaaatcaaaagtagtgctgatgatgctaagaaggatgctgctaagaaggatgatgctaagaaggatgatgctaagaaggatggtga ggtgaagctggatgagactggaggaggcttggtgcaacctgggaggcccatgaaactctcctgtgttgcctctggattcacttttagtgacta ctggatgaactgggtccgccagtctccagagaaaggactggagtgggtagcacaaattagaaacaaaccttataattatgaaacatattattc agattctgtgaaaggcagattcaccatctcaagagatgattccaaaagtagtgtctacctgcaaatgaacaacttaagagttgaagacatggg tatctattactgtacgggttcttactatggtatggactactggggtcaaggaacctcagtcaccgtctcc (SEQ ID N0: 1). The sequence was ligated into pMSGVl to generate the aFITC-CAR retroviral vector.
[0067] Retroviral transduction of human T cells. HLA-A2+ peripheral blood mononuclear cells (PBMC) from healthy donors were purchased from Biological Specialty Corp (Colmar, PA, USA), and isolated by Ficoll-Pague (GE Healthcare, Piscataway, NJ, USA) density gradient centrifugation. Isolated PBMC were cultured at 3 x 106 per well in 24- well tissue culture plates in AIM V medium (GIBCO brand; Invitrogen) supplemented with 5% human AB serum (Sigma- Aldrich), 1% MEM non-essential amino acids, 1% penicillin-streptomycin and 100 U/ml recombinant human IL-2 (BioLegend, San Diego, CA, USA), and activated with 50 ng/ml OKT3 (eBioscience, San Diego, CA, USA). Two days later, cells were collected for retroviral transduction. For transduction, 24-well non-tissue culture treated plates (BD Biosciences, Franklin Lakes, NJ, USA) were coated with 0.5 ml per well of 10 μg/ml recombinant human fibronectin fragment (RetroNectin; Takara, Otsu, Shiga, Japan) overnight at 4 °C. After incubation, wells were blocked with 1 ml of Hanks' s balanced salt solution (GIBCO brand; Invitrogen) plus 2.5% human AB serum for 30 min at RT, and washed with Hanks' s balanced salt solution plus 2.5% N-2-hydroxyethylpiperazine-N'-2-ethanesulfonic acid (HEPES) (GIBCO brand; Invitrogen). Transductions were conducted as previously described (Johnson et al. Blood 114, 535-546 (2009)). Briefly, approximately 2.5 ml of retroviral supernatant were added to each coated well followed by centrifugation at 2000g for 2 h at 32 °C. 1.5 ml of viral supernatant was removed and 1 x 106 (0.5 ml) activated PBMC were added to each well in the presence of 100 U/ml IL-2. Plates were centrifugated at lOOOg for 10 min, and then incubated overnight at 37 °C. After transduction, cells were washed and maintained in the presence of IL-2 (lOOU/ml) and used in experiments five days after transduction. The surface expression of aFrfC-CAR on transduced human T cells was determined by flow cytometry after staining cells with CD3 or CD8 and FITC-conjugated cetuximab. In some experiments, aFrfC-CAR transduced T cells were stained with FITC-conjugated dextran beads. Cells stained with FITC-conjugated purified human IgG (Invitrogen) were used as an isotype control. [0068] T cell proliferation assay, cytokine and chemokine production assay. Three to five days after transduction, 1 x 105 T cells were cultured in 96-well round-bottom plates coated with cetuximab, FITC-conjugated cetuximab, or FITC-Dextran for 72 h. For T cell specific reactivity against tumor cells, SW480 cells were pulsed with the indicated concentrations of antibodies for 1 h at 37 °C, washed 3 times. 1 x 105 effector T cells and 1 x 105 tumor cells were co-cultured in 200 μΐ of culture volume in 96-well round-bottom plates for 72 h. Sixteen hours before harvesting, 0.5 μθ of 3H-thymidine was added to each well prior to measuring thymidine uptake using a 1450 LSC & luminescence counter (PerkinElmer, Waltham, MA, USA). Cytokine and chemokine production levels were measured from culture supernatants collected 48 hours after stimulation using a Cytokine/Chemokine kit (Millipore, Billerica, MA, USA) according to manufacturer's instructions. In some experiments, tumor explants were finely minced and cultured in trypsin for 2 hours at 37°C followed by T cell enrichment using a negative selection kit (In vitro gen/Life Technologies, Grand Island, NY). 200,000 T cells were co-cultured with SW480 (50,000) colon cancer cells which had been pulsed with FITC-Ctx or Ctx (0.5 μg/l x 106 cells). Three days later proliferation was determined by [ H]thymidine uptake (±SD) or cytokine and chemokine production was measured using a Milliplex array.
[0069] Cytotoxicity assay. Cytotoxic activity against tumor target cells was measured using a standard 51Cr release assay. 1 x 106 target cells were labeled with 200 μα of 51Cr for 2 h at 37°C, washed 3 times, and pulsed with anti-human antibodies for 1 h at 37 °C. 1 x 104 labeled target cells were then co-cultured with decreasing numbers of effector T cells at the indicated effector to target (E:T) ratios in 200 μΐ of culture volume in 96-well round-bottom plates. Target cells incubated in medium alone were used to determined spontaneous 51Cr release, and maximal release was determined by incubating labeled target cells in 10% Triton X-100. After 5 hours at 37°C, 50 μΐ of supernatant was collected and 51Cr radioactivity was measured in a 1450 LSC & luminescence counter. The mean percentage of specific lysis was calculated according to the following equation: % specific lysis = (test release - spontaneous release) / (maximal release - spontaneous release) x 100. All tests were performed in triplicate wells and results are shown as mean +SD.
[0070] Tumor models and adoptive immunotherapy. In prophylactic tumor models 6- to 8- wk-old male NSG mice (n = 5 for each group) were injected subcutaneously (s.c.) in the rear leg flank with 1 - 2 x 106 SW480 or Pane 6.03 tumor cells. One day later mice were injected intraperitoneally (i.p.) with FITC-Ctx or Ctx (25 μg/mouse). One day after Ctx injection, mice were injected intravenous (i.v.) with 5 x 106 aFITC-CAR transduced human T cells. After adoptive T cells transfer, mice were injected i.p. with antibodies (25 μg/mouse) weekly for three weeks. Tumor area was measured with digital calipers in a blinded manner two to three times per week, and tumor sizes (mm") were calculated by perpendicular measurement by longitudinal diameter. Mice were euthanized when tumor sizes reach 200 mm or if mice became moribund or had trouble ambulating. All experiments were performed independently at least twice with similar results. Survival data were analyzed with the exact long-rank test.
[0071] Experiment 1. Peripheral blood mononuclear cells were activated with anti-CD3 mAb in the presence of IL-2 followed by transduction with the -FITC-CD28-41BB-CD3 -CAR vector (referred to as aFITC-CAR) as shown in Figure 1A and as described above. The expression of the ccFITC scFv on T cells was analyzed by staining cells with anti-CD8 (or anti- CD3) and FITC-labeled cetuximab (FITC-Ctx) or FITC-labeled dextran (FITC-Dex) beads. On average, 60% of total T cells expressed aFITC-CAR (Figure IB). To confirm their functionality and specificity, aFITC-CAR or control (mock transduced) T cells were activated using titrating concentrations of plate-bound FITC-Ctx, unbound cetuximab, or FITC-Dex beads. aFITC-CAR T cells proliferated vigorously and in a dose dependent manner following stimulation with FrfC- Ctx and FITC-Dex but did not divide in response to stimulation with Ctx alone (Figure 1C, left panel). In contrast, control T cells did not proliferate to FITC-Ctx or FITC-Dex (Figure 1C, right panel). T cells were also co-cultured with EGFR+ colon cancer cells (SW480) which had been stained with titrating concentrations of FITC-Ctx. FrfC reactivity by aFITC-CAR T cells was demonstrated by their ability to divide following activation with FITC-Ctx-stained cancer cells, Figure ID. However, proliferation was similar to that of control at the lowest
concentrations of FITC-Ctx. Control T cells did not proliferative in response to any
concentration of FITC-Ctx-stained cancer cells, Figure ID.
[0072] To determine their cytolytic capacity, aFITC-CAR T cells were cultured together with FITC-Ctx-stained SW480 colon cancer cells at various effector to target ratios. aFITC- CAR T cells lysed SW480 colon cancer cells at effector to target ratios as low as one T cell to twenty target cells (Figure IE, left panel) but did not lyse cancer cells labeled with FITC-mouse IgG (Figure IE, right panel). Similarly, control T cells did not show an appreciable level of cytolytic activity against FITC-Ctx-labeled or FITC-IgG-labeled SW480 cancer cells (Figure IE). To confirm their ability to recognize a variety of target cells expressing different antigens CcFITC-CAR T cells were co-cultured with pancreatic cancer cells (Pane 6.03) stained with FITC-Ctx or stained with FITC-Herceptin (anti-Her-2 mAb; FITC-Her2) and breast cancer cells (AU565) stained with FITC-Her2. ccFITC-CAR T cells efficiently and specifically lysed cancer cells stained with FITC-Ctx or FITC-Her2 (Figure IF). It is worth noting that the increased cytolytic activity against FITC-Her2-stained pancreatic cancer cells above cells stained with FITC-Ctx is likely associated with the higher expression level Her-2 as compared with EGFR (data not shown). Furthermore, ccFITC-CAR T cells produced a wide array of cytokines beneficial to T cell survival, expansion and chemotaxis (Table 1). The background cytokine levels produced by control T cells activated with FITC-Ctx- or Ctx-stained cancer cells were identical to one another.
Figure imgf000025_0001
Table 1. Cytokines and chemokine production by aFrfC-CAR T cells. HLA-A2+ PBMCs were activated with anti-CD3 antibodies in the presence of IL-2 followed by transduction with aFFTC- CAR retrovirus. aFITC-CAR functionality on transduced was determined by their ability to produce cytokines and chemokines following activation with SW480colon cancer cells that were stained with FITC-Ctx or Ctx. The levels of various cytokines and chemokines produced by a- FITC-CAR T cells were measured 72 hours after stimulation using a Milliplex
cytokine/chemokine array. These data are representative of three independent experiments (three different donors) with each experiment yielding the same trends.
[0073] Collectively, these data demonstrate: 1) the functionality of aFITC-CAR T cells, 2) their specificity against FITC-Ab-stained cells but not soluble FITC, 3) their ability to lyse a diverse set tumor cell types, and 4) the use of various FITC-tagged antibodies.
[0074] Experiment 2. The ability to re-direct aFITC-CAR T cells to eliminate tumor cells in vivo was examined. Mice were injected s.c. with SW840 colon cancer cells followed by the administration of FITC-Ctx or Ctx via i.p. injection. One day later ccFITC-CAR T cells were administered via i.v. injection into the tail vein. Tumor growth kinetics was similar between mice receiving FITC-CAR T cells plus Ctx and those receiving Ctx alone, Figure 2A left panel. In sharp contrast, tumor growth was greatly suppressed in mice receiving aFITC-CAR T cells plus FITC-Ctx (Figure 2A left panel). Likewise, the tumor-free occurrence (Figure 2A middle panel) and overall survival (Figure 2A right panel) was significantly improved in mice receiving aFITC-CAR T cells plus FITC-Ctx as compared with control groups. In spite of this survival advantage however, mice receiving a-ITC-CAR T cells plus FITC-Ctx succumbed to tumor challenge within 55 days of tumor implantation.
[0075] The mechanisms contributing to the failure of aFITC-CAR T cells in long-term treatment were further investigated. Because T cells were activated with CD3 mAb and IL-2, CAR T cells can display a shortened life despite receiving prosurvival signals from CD28 or 41BB (Sadelain et al. Curr. Opin. Immunol. 21, 215-223 (2009); Brentjens et al. Nat. Med. 9, 279-286 (2003)). The presence of aFITC-CAR T cells in various tissues was assayed, including the spleen, liver, bone marrow, circulation and in tumor explants. aFITC-CAR T cells were found in all tissues analyzed. Approximately 10% of all human T cells detected in tumor explants were aFITC-CAR T cells, Figure 2B. Similar percentages were found in the other tissues (data not shown). However, the overall percentage of aFITC-CAR T cells at the time of organ collection (between days 38 and 45) was significantly lower than the starting percentage of 60% at infusion. It is also worth noting that 60% to 90% of the T cells recovered from mice were CD8+, as compared with the starting percentage of 40-50%. These data suggest a potential survival advantage of CD8+ subset over CD4+ T cells in vivo. The frequency or role for CD4 TRegs was not examined but remains a viable option that merits further investigation.
[0076] Alternatively, aFrfC-CAR T cells might not have become sufficiently activated by antigen (FITC) perhaps due to anergy or other suppressive mechanisms. Total T cells were enriched from the tumor explant and immediately reactivated using SW480 cells from tissue culture that were stained with FITC-Ctx or Ctx alone. aFITC-CAR T cells proliferated (Figure 2C) and produced various effector molecules and chemokines (Figure 2D) following stimulation with FITC-Ctx-stained SW480 cells but did not respond to SW480 cells stained with Ctx.
Unfortunately, the numbers of aFITC-CAR T cells isolated from tumor explants were not sufficient to test their killing aptitude. However, based on their ability to divide and produce effector molecules ex vivo, -FI C-CAR T cells are clearly capable of responding to FITC stimulation.
[0077] Based on the observations that α-FITC-CAR were present in mice and were responsive to FITC-CAR stimulation, EGFR expression on tumor explants was examined. As examined by flow cytometry, all tumor explants were completely devoid of EGFR expression as compared with isotype control and with SW480 cells taken from tissue culture (Figure 2E). In addition, although the majority of SW480 cells taken from tissue culture expressed EGFR, its expression was heterogeneous with some cells lacking EGFR.
[0078] Taken together, these data support the contention that aFITC-CAR killed EGFR+ cells but in time allowed the growth of EGFR- cells which were no longer a target for aFITC- CAR T cells. The lack of EGFR expression, and therefore lack of FITC-mediated stimulation, might have also contributed to the lower frequency of aFITC-CAR T cells observed at later time points in tumor-bearing mice as compared with the percentage of aFITC-CAR T cells prior to injection. Additionally, these data highlight the potential for tumor escape occurring in patients in whose TAA is heterogeneously expressed. These studies also emphasize the potential need to use CAR T cells with specificity to several TAAs. One advantage to the use of an anti-tag CAR T cells is the potential to use several FITC 'tagged' tumor-reactive antibodies simultaneously. Alternatively, the use of CARs expressing scFvs specific for biotin or PE-conjugated antibodies would add to the diversity of anti-tag CARs.
[0079] Experiment 3. Given the heterogeneity of EGFR expression on SW480 cancer cells, the ability of aFITC-CAR T cells to destroy a population of cancer cells in which all of the cells expressed the antigen was examined. The pancreatic cancer cell line Pane 6.03 was selected as the cells uniformly express EGFR (Figure 3A). The cytolytic activity of aFITC-CAR T cells was examined in a prophylactic tumor model using the same procedure described in Experiment 2. Tumor growth was clearly suppressed in mice receiving aFITC-CAR T cells plus FITC-Ctx whereas mice receiving α-FITC-CAR T cells plus Ctx demonstrated rapid tumor growth, Figure 3B left panel. All mice receiving aFITC-CAR T cells plus Ctx succumbed to tumor challenge within 35 days of tumor implantation, Figure 3B right panel. It is worth noting that the administration of FITC-Ctx via i.v., i.p., or intratumoral injection all resulted in antibody localization to the tumor (data not shown). An alternative method to redirecting T cells to the tumor is to coat ccFITC-CAR T cells with FITC-Ctx prior to adaptive transfer.
[0080] Experiment 4. The ability of FITC-CAR T cells to destroy an established pancreatic tumor was examined. Pancreatic tumors were grown to sizes between 3-10 mm when tumors were well-vascularized and then injected (i.p.) with FITC-Ctx or Ctx. One day later mice were administered aFITC-CAR T cells via tail vein injection. T cells with redirected specificity for EGFR eradicated established pancreatic tumors in all mice (Figure 3C left panel) and improved survival as compared with mice treated with Ctx and aFITC-CAR T cells (Figure 3C right panel). No tumor relapse occurred during the time of observation.
[0081] In summary, these studies are the first to describe the generation of a universal and adaptable CAR system which confers T cells specificity to FITC-tagged antibodies which when bound to various cancer types mediate tumor destruction. This report is also the first to emphasize the importance of using CAR T cells to target more than one TAA as TAA-negative tumor variants can arise and eventually kill the host. The platform is considered an 'off-the- shelf system that considerably advances the existing CAR technology through its potential to target an assortment of tagged proteins (i.e., antibodies) in order to target various cancer types.
* * * *
[0082] It will be apparent to those skilled in the art that various modifications and variations can be made in the present invention without departing from the scope or spirit of the invention. Other embodiments of the invention will be apparent to those skilled in the art from
consideration of the specification and practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with a true scope and spirit of the invention being indicated by the following claims.
[0083] Throughout this application, various publications, patents, and/or patent applications are referenced in order to more fully describe the state of the art to which this invention pertains. The disclosures of these publications, patents, and/or patent applications are herein incorporated by reference in their entireties to the same extent as if each independent publication, patent, and/or patent application was specifically and individually indicated to be incorporated by reference.

Claims

WHAT IS CLAIMED IS:
1. A method of treating cancer in a subject, comprising:
(a) administering a formulation of tagged proteins to a subject in need of treatment, wherein the tagged proteins bind a cancer cell in the subject, and
(b) administering a therapeutically-effective population of anti-tag chimeric antigen receptor (AT-CAR)-expressing T cells to the subject, wherein the AT-CAR-expressing T cells bind the tagged proteins and induce cancer cell death, thereby treating cancer in a subject.
2. A method of treating cancer in a subject, comprising:
(a) administering one or more formulations of tagged proteins to a subject in need of treatment, wherein the tagged proteins bind a cancer cell in the subject, and
(b) administering one or more therapeutically-effective populations of AT-CAR-expressing T cells to the subject, wherein the AT-CAR-expressing T cells bind the tagged proteins and induce cancer cell death, thereby treating cancer in a subject.
3. A method of treating cancer in a subject, comprising:
(a) administering at least two formulations of tagged proteins to a subject in need of treatment, wherein the tagged proteins bind a cancer cell in the subject, and
(b) administering at least two therapeutically-effective populations of AT-CAR-expressing T cells to the subject, wherein the AT-CAR-expressing T cells bind the tagged proteins and induce cancer cell death, thereby treating cancer in a subject.
4. The method of any one of claims 1-3, wherein the tag of each formulation of tagged proteins is the same or different and the tag is selected from the group consisting of fluorescein isothiocyanate (FITC), streptavidin, biotin, histidine, dinitrophenol, peridinin chlorophyll protein complex, green fluorescent protein, phycoerythrin (PE), horse radish peroxidase, palmitoylation, nitrosylation, alkalanine phosphatase, glucose oxidase, and maltose binding protein.
5. The method of any one of claims 1-3, wherein the protein of each formulation of tagged proteins is the same or different and the protein is an antibody or an antigen-binding fragment thereof.
6. The method of claim 5, wherein the antibody or antigen-binding fragment thereof is cetuximab, nimotuzumab, panitumumab, retuximab, omalizumab, tositumomab, trastuzumab, gemtuzumab, alemtuzumab, bevacuzimab or an antigen-binding fragment of any one thereof.
7. The method of any one of claims 1-3, wherein the AT-CAR of each population of AT-CAR-expressing T cells is the same or different and the AT-CAR comprises a tag-binding domain, a transmembrane domain, and an activation domain.
8. The method of claim 7, wherein the tag-binding domain is an antibody or an antigen-binding fragment thereof.
9. The method of claim 7, wherein the tag-binding domain specifically binds FITC, biotin, PE, histidine or streptavidin.
10. The method of claim 8, wherein the antigen-binding fragment is a single chain variable fragment (scFv).
11. The method of claim 8, wherein the antigen-binding fragment is a single chain variable fragment (scFv) that specifically binds FITC, biotin, PE, histidine or streptavidin.
12. The method of claim 7, wherein the transmembrane domain is the hinge and transmembrane regions of the human CD8a chain.
13. The method of claim 7, wherein the activation domain comprises one or more of the cytoplasmic region of CD28, the cytoplasmic region of CD137 (41BB), OX40, HVEM, CD3C and FcRs.
14. The method of any one of claims 1-3, wherein the T cells of each population of AT-CAR-expressing T cells are the same or different and wherein the T cells are selected from the group consisting of T cells of any HLA-background from peripheral blood mononuclear cells (PBMC), T cells isolated from a tumor explant of the subject, and intratumoral T cells of the subject.
15. The method of any one of claims 1-3, wherein the T cells of each population of AT-CAR-expressing T cells consist of HLA-A2+ peripheral blood mononuclear cells (PBMC).
16. The method of any one of claims 1-3, wherein the formulation(s) of tagged proteins are administered to the subject prior to administration of the therapeutically-effective population(s) of AT-CAR-expressing T cells.
17. The method of any one of claims 1-3, wherein the formulation(s) of tagged proteins are administered to the subject concurrently with administration of the therapeutically- effective population(s) of AT-CAR-expressing T cells.
18. The method of any one of claims 1-3, wherein the formulation(s) of tagged proteins are administered to the subject after administration of the therapeutically-effective population(s) of AT-CAR-expressing T cells.
19. The method of any one of claims 1-3, wherein the formulation(s) of tagged proteins and the therapeutically-effective population(s) of AT-CAR-expressing T cells are administered to the subject in any order.
20. The method of any one of claims 1-3, wherein AT-CAR-expressing T cell binding to the tagged proteins induces cytolytic activation of the T cells.
21. The method of any one of claims 1-3, wherein the subject is a human.
PCT/US2011/064808 2010-12-14 2011-12-14 Universal anti-tag chimeric antigen receptor-expressing t cells and methods of treating cancer WO2012082841A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
US13/993,396 US9233125B2 (en) 2010-12-14 2011-12-14 Universal anti-tag chimeric antigen receptor-expressing T cells and methods of treating cancer
EP11849221.4A EP2651442B1 (en) 2010-12-14 2011-12-14 Universal anti-tag chimeric antigen receptor-expressing t cells and methods of treating cancer
ES11849221T ES2791716T3 (en) 2010-12-14 2011-12-14 T cells expressing the universal anti-label chimeric antigen receptor and methods for the treatment of cancer
EP20161627.3A EP3693017A1 (en) 2010-12-14 2011-12-14 Universal anti-tag chimeric antigen receptor-expressing t cells and methods of treating cancer
JP2013544725A JP5897035B2 (en) 2010-12-14 2011-12-14 General-purpose anti-tag chimeric antigen receptor-expressing T cells and methods for treating cancer
AU2011343887A AU2011343887B2 (en) 2010-12-14 2011-12-14 Universal anti-tag chimeric antigen receptor-expressing T cells and methods of treating cancer
CA2821080A CA2821080C (en) 2010-12-14 2011-12-14 Universal anti-tag chimeric antigen receptor-expressing t cells and methods of treating cancer
US14/990,514 US10973893B2 (en) 2010-12-14 2016-01-07 Universal anti-tag chimeric antigen receptor-expressing T cells and methods of treating cancer
US17/228,001 US20210330769A1 (en) 2010-12-14 2021-04-12 Universal anti-tag chimeric antigen receptor-expressing t cells and methods of treating cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US42268110P 2010-12-14 2010-12-14
US61/422,681 2010-12-14

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/993,396 A-371-Of-International US9233125B2 (en) 2010-12-14 2011-12-14 Universal anti-tag chimeric antigen receptor-expressing T cells and methods of treating cancer
US14/990,514 Continuation US10973893B2 (en) 2010-12-14 2016-01-07 Universal anti-tag chimeric antigen receptor-expressing T cells and methods of treating cancer

Publications (2)

Publication Number Publication Date
WO2012082841A2 true WO2012082841A2 (en) 2012-06-21
WO2012082841A9 WO2012082841A9 (en) 2012-08-09

Family

ID=46245339

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/064808 WO2012082841A2 (en) 2010-12-14 2011-12-14 Universal anti-tag chimeric antigen receptor-expressing t cells and methods of treating cancer

Country Status (7)

Country Link
US (3) US9233125B2 (en)
EP (2) EP3693017A1 (en)
JP (1) JP5897035B2 (en)
AU (1) AU2011343887B2 (en)
CA (2) CA3102782A1 (en)
ES (1) ES2791716T3 (en)
WO (1) WO2012082841A2 (en)

Cited By (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013044225A1 (en) * 2011-09-22 2013-03-28 The Trustees Of The University Of Pennsylvania A universal immune receptor expressed by t cells for the targeting of diverse and multiple antigens
WO2014011987A1 (en) * 2012-07-13 2014-01-16 The Trustees Of The University Of Pennsylvania Compositions and methods for regulating car t cells
WO2014100615A1 (en) * 2012-12-20 2014-06-26 Purdue Research Foundation Chimeric antigen receptor-expressing t cells as anti-cancer therapeutics
WO2014134165A1 (en) 2013-02-26 2014-09-04 Memorial Sloan-Kettering Cancer Center Compositions and methods for immunotherapy
US8906682B2 (en) 2010-12-09 2014-12-09 The Trustees Of The University Of Pennsylvania Methods for treatment of cancer
WO2015017214A1 (en) 2013-07-29 2015-02-05 Bluebird Bio, Inc. Multipartite signaling proteins and uses thereof
WO2015057852A1 (en) * 2013-10-15 2015-04-23 The California Institute For Biomedical Research Chimeric antigen receptor t cell switches and uses thereof
JP2015523386A (en) * 2012-07-13 2015-08-13 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 Use of CART19 to deplete normal B cells to induce tolerance
US20150307564A1 (en) * 2013-10-15 2015-10-29 The California Institute For Biomedical Research Peptidic chimeric antigen receptor t cell switches and uses thereof
CN105194661A (en) * 2014-06-26 2015-12-30 科济生物医药(上海)有限公司 System for inhibiting pathological target cells in space-time adjustable manner
EP2990416A1 (en) 2014-08-29 2016-03-02 GEMoaB Monoclonals GmbH Universal chimeric antigen receptor expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
US9394368B2 (en) 2013-02-20 2016-07-19 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
JP2017501702A (en) * 2013-12-20 2017-01-19 フレッド ハッチンソン キャンサー リサーチ センター Tagged chimeric effector molecule and its receptor
US9573988B2 (en) 2013-02-20 2017-02-21 Novartis Ag Effective targeting of primary human leukemia using anti-CD123 chimeric antigen receptor engineered T cells
WO2017041143A1 (en) * 2015-09-11 2017-03-16 Ctm@Crc Ltd. Chimeric antigen receptors and uses thereof
US9745368B2 (en) 2013-03-15 2017-08-29 The Trustees Of The University Of Pennsylvania Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US9777061B2 (en) 2014-07-21 2017-10-03 Novartis Ag Treatment of cancer using a CD33 chimeric antigen receptor
US9815901B2 (en) 2014-08-19 2017-11-14 Novartis Ag Treatment of cancer using a CD123 chimeric antigen receptor
EP3315511A1 (en) 2016-10-29 2018-05-02 Miltenyi Biotec GmbH Adapter chimeric antigen receptor expressing cells for targeting of multiple antigens
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
EP3283113A4 (en) * 2015-04-15 2018-12-05 The California Institute for Biomedical Research Optimized pne-based chimeric receptor t cell switches and uses thereof
WO2018224660A1 (en) 2017-06-09 2018-12-13 Armin Ehninger Targeting modules for universal chimeric antigen receptor expressing immune cells and use in the treatment of cancer infections and autoimmune disorders
US10174095B2 (en) 2014-07-21 2019-01-08 Novartis Ag Nucleic acid encoding a humanized anti-BCMA chimeric antigen receptor
US10221245B2 (en) 2013-03-16 2019-03-05 Novartis Ag Treatment of cancer using humanized anti-CD19 chimeric antigen receptor
WO2019060425A1 (en) 2017-09-19 2019-03-28 Massachusetts Institute Of Technology Compositions for chimeric antigen receptor t cell therapy and uses thereof
US10253086B2 (en) 2015-04-08 2019-04-09 Novartis Ag CD20 therapies, CD22 therapies, and combination therapies with a CD19 chimeric antigen receptor (CAR)-expressing cell
US10273300B2 (en) 2014-12-29 2019-04-30 The Trustees Of The University Of Pennsylvania Methods of making chimeric antigen receptor-expressing cells
US10287354B2 (en) 2013-12-20 2019-05-14 Novartis Ag Regulatable chimeric antigen receptor
WO2019104562A1 (en) * 2017-11-29 2019-06-06 广州中科蓝华生物科技有限公司 Chimeric antigen receptor and application thereof
US10357514B2 (en) 2014-04-07 2019-07-23 The Trustees Of The University Of Pennsylvania Treatment of cancer using anti-CD19 Chimeric Antigen Receptor
EP3581200A1 (en) 2018-06-13 2019-12-18 GEMoaB Monoclonals GmbH Reversed universal chimeric antigen receptor expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
US20190388468A1 (en) * 2016-12-15 2019-12-26 Miltenyi Biotec Gmbh Immune cells expressing an antigen binding receptor and a chimeric costimulatory receptor
US10525083B2 (en) 2016-10-07 2020-01-07 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
US10568947B2 (en) 2014-07-21 2020-02-25 Novartis Ag Treatment of cancer using a CLL-1 chimeric antigen receptor
US10577417B2 (en) 2014-09-17 2020-03-03 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
EP3620464A1 (en) 2018-09-10 2020-03-11 Miltenyi Biotec GmbH Car cell having crosslinked disulfide bridge on antigen recognizing moiety
WO2020068261A1 (en) 2018-09-28 2020-04-02 Massachusetts Institute Of Technology Collagen-localized immunomodulatory molecules and methods thereof
EP2651442B1 (en) 2010-12-14 2020-04-22 University of Maryland, Baltimore Universal anti-tag chimeric antigen receptor-expressing t cells and methods of treating cancer
US10640569B2 (en) 2013-12-19 2020-05-05 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
CN111454909A (en) * 2019-01-22 2020-07-28 北京大学深圳研究生院 Preparation method of natural ligand-mediated multi-target recognition controllable genetically engineered immune cells
US10774388B2 (en) 2014-10-08 2020-09-15 Novartis Ag Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
US10800828B2 (en) 2015-03-26 2020-10-13 The Scripps Research Institute Switchable non-scFv chimeric receptors, switches, and methods of use thereof to treat cancer
WO2020213724A1 (en) 2019-04-19 2020-10-22 中外製薬株式会社 Chimeric receptor recognizing modification site of antibody
US10829735B2 (en) 2015-07-21 2020-11-10 The Trustees Of The University Of Pennsylvania Methods for improving the efficacy and expansion of immune cells
WO2020263399A1 (en) 2019-06-26 2020-12-30 Massachusetts Institute Of Technology Immunomodulatory fusion protein-metal hydroxide complexes and methods thereof
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
US11028143B2 (en) 2014-01-21 2021-06-08 Novartis Ag Enhanced antigen presenting ability of RNA CAR T cells by co-introduction of costimulatory molecules
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
EP3878464A1 (en) 2020-03-09 2021-09-15 Miltenyi Biotec B.V. & Co. KG Use of a car cell having crosslinked disulfide bridge on antigen recognizing moiety for targeting cancer cells
EP3881866A1 (en) 2020-03-16 2021-09-22 GEMoaB GmbH A targeting module comprising pd-l1 and/or pd-l2 for use in a method for stimulating a chimeric antigen receptor mediated response in a mammal
US11161907B2 (en) 2015-02-02 2021-11-02 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
US11174306B2 (en) 2016-10-19 2021-11-16 The Scripps Research Institute Chimeric antigen receptor effector cell switches with humanized targeting moieties and/or optimized chimeric antigen receptor interacting domains and uses thereof
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
CN113846062A (en) * 2014-07-25 2021-12-28 赛拉福柯蒂斯公司 Lentiviral vectors for regulated expression of chimeric antigen receptor molecules
US11225520B2 (en) 2016-02-16 2022-01-18 Dana-Farber Cancer Institute, Inc. Immunotherapy compositions and methods
WO2022025220A1 (en) 2020-07-31 2022-02-03 中外製薬株式会社 Pharmaceutical composition including cell expressing chimeric receptor
WO2022038411A1 (en) 2020-08-19 2022-02-24 Astellas Pharma, Inc. Human non-naturally occurring modified fc region of igg specifically binding to non-naturally occurring modified fc receptor
WO2022049217A1 (en) 2020-09-04 2022-03-10 Miltenyi Biotec B.V. & Co. KG System for inducible expression of an adapter in immune cells
US11311576B2 (en) 2018-01-22 2022-04-26 Seattle Children's Hospital Methods of use for CAR T cells
WO2022133169A1 (en) * 2020-12-18 2022-06-23 Century Therapeutics, Inc. Chimeric antigen receptor systems with adaptable receptor specificity
US11413340B2 (en) 2015-12-22 2022-08-16 Novartis Ag Mesothelin chimeric antigen receptor (CAR) and antibody against PD-L1 inhibitor for combined use in anticancer therapy
US11459390B2 (en) 2015-01-16 2022-10-04 Novartis Ag Phosphoglycerate kinase 1 (PGK) promoters and methods of use for expressing chimeric antigen receptor
WO2022258681A1 (en) * 2021-06-08 2022-12-15 Biontech Cell & Gene Therapies Gmbh Agents and methods for activation and targeting of immune effector cells
US11535662B2 (en) 2017-01-26 2022-12-27 Novartis Ag CD28 compositions and methods for chimeric antigen receptor therapy
US11542488B2 (en) 2014-07-21 2023-01-03 Novartis Ag Sortase synthesized chimeric antigen receptors
US11549099B2 (en) 2016-03-23 2023-01-10 Novartis Ag Cell secreted minibodies and uses thereof
WO2023004461A1 (en) * 2021-07-28 2023-02-02 Prescient Therapeutics Ltd Universal receptor immune cell therapy
US11608382B2 (en) 2018-06-13 2023-03-21 Novartis Ag BCMA chimeric antigen receptors and uses thereof
EP4176895A1 (en) 2021-11-08 2023-05-10 AvenCell Europe GmbH Targeting modules against il13ra2 or her2 for use in combination with a chimeric antigen receptor
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
US11747346B2 (en) 2015-09-03 2023-09-05 Novartis Ag Biomarkers predictive of cytokine release syndrome
US11759480B2 (en) 2017-02-28 2023-09-19 Endocyte, Inc. Compositions and methods for CAR T cell therapy
EP4093768A4 (en) * 2019-10-18 2023-11-15 Trustees of Boston University Cal-t constructs and uses thereof
US11851659B2 (en) 2017-03-22 2023-12-26 Novartis Ag Compositions and methods for immunooncology
US11883494B2 (en) 2014-11-26 2024-01-30 Miltenyi Biotec B.V. & Co. KG Combination of immune effector cells specific for a target antigen and hematopoietic calls that express the target antigen in an altered form
US11896614B2 (en) 2015-04-17 2024-02-13 Novartis Ag Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
US11952413B2 (en) 2017-12-14 2024-04-09 2Seventy Bio, Inc. Dimerizing agent regulated immunoreceptor complexes comprising interleukin receptor signaling domains
WO2024078995A1 (en) 2022-10-15 2024-04-18 Miltenyi Biotec B.V. & Co. KG Transduction of gammadelta t cells with pseudotyped retroviral vectors

Families Citing this family (117)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100908114B1 (en) * 2002-03-09 2009-07-16 삼성전자주식회사 Scalable lossless audio encoding / decoding apparatus and method thereof
ES2625288T3 (en) 2011-04-15 2017-07-19 The Johns Hopkins University Secure Sequencing System
ES2701742T3 (en) 2012-10-29 2019-02-25 Univ Johns Hopkins Papanicolaou test for ovarian and endometrial cancers
JP6315848B2 (en) * 2013-03-13 2018-04-25 ザ リージェンツ オブ ザ ユニバーシティー オブ ミシガン MEK / PI3K dual inhibitor and therapeutic method using the inhibitor
US9587237B2 (en) 2013-03-14 2017-03-07 Elwha Llc Compositions, methods, and computer systems related to making and administering modified T cells
SG11201506974XA (en) * 2013-03-14 2015-10-29 Bellicum Pharmaceuticals Inc Methods for controlling t cell proliferation
US9499855B2 (en) 2013-03-14 2016-11-22 Elwha Llc Compositions, methods, and computer systems related to making and administering modified T cells
US10208125B2 (en) 2013-07-15 2019-02-19 University of Pittsburgh—of the Commonwealth System of Higher Education Anti-mucin 1 binding agents and uses thereof
CN111849912B (en) 2014-02-14 2024-03-15 贝里坤制药股份有限公司 Method for activating T cells using inducible chimeric polypeptides
WO2015143224A1 (en) * 2014-03-19 2015-09-24 Ambrx, Inc. Chimeric antigen receptor-modified t-cells
WO2015188191A1 (en) * 2014-06-06 2015-12-10 Wong Wilson W Dna recombinase circuits for logical control of gene expression
AU2015312117A1 (en) 2014-09-02 2017-03-02 Bellicum Pharmaceuticals, Inc. Costimulation of chimeric antigen receptors by Myd88 and CD40 polypeptides
PL3597742T3 (en) * 2014-10-09 2022-11-14 Yamaguchi University Car expression vector and car-expressing t cells
WO2016138491A1 (en) 2015-02-27 2016-09-01 Icell Gene Therapeutics Llc Chimeric antigen receptors (cars) targeting hematologic malignancies, compositions and methods of use thereof
JP2018518152A (en) * 2015-03-27 2018-07-12 ユニバーシティ オブ サザン カリフォルニア CAR T cell therapy directed to LHR for treating solid tumors
EP3288570A4 (en) 2015-04-29 2018-11-21 Fred Hutchinson Cancer Research Center Modified stem cells and uses thereof
KR20180021137A (en) 2015-06-25 2018-02-28 아이셀 진 테라퓨틱스 엘엘씨 Chimeric antigen receptor (CAR), compositions and methods for their use
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
US11286531B2 (en) 2015-08-11 2022-03-29 The Johns Hopkins University Assaying ovarian cyst fluid
WO2017030370A1 (en) * 2015-08-17 2017-02-23 서울대학교산학협력단 Chimeric antibody receptor to which anti-cotinine antibody is linked, and use thereof
US20180243340A1 (en) * 2015-08-24 2018-08-30 University Of Houston System Combination therapy combining car + t cells with appropriately timed immunodulatory antibodies
EP3569244A1 (en) 2015-09-23 2019-11-20 CytoImmune Therapeutics, LLC Flt3 directed car cells for immunotherapy
WO2017069958A2 (en) 2015-10-09 2017-04-27 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
US11001622B2 (en) 2015-11-19 2021-05-11 The Brigham And Women's Hospital, Inc. Method of treating autoimmune disease with lymphocyte antigen CD5-like (CD5L) protein
WO2017112877A1 (en) * 2015-12-22 2017-06-29 Icell Gene Therapeutics, Llc Chimeric antigen receptors and enhancement of anti-tumor activity
WO2017112784A1 (en) * 2015-12-22 2017-06-29 The Trustees Of The University Of Pennsylvania Spycatcher and spytag: universal immune receptors for t cells
MY190565A (en) 2016-03-17 2022-04-27 Univ Yamaguchi Immunocompetent cell and expression vector expressing regulatory factors of immune function
CA3019835A1 (en) * 2016-04-08 2017-10-12 Purdue Research Foundation Methods and compositions for car t cell therapy
WO2017192536A1 (en) 2016-05-02 2017-11-09 University Of Kansas Eliminating mhc restriction from the t cell receptor as a strategy for immunotherapy
CN109562126A (en) 2016-06-24 2019-04-02 美商生物细胞基因治疗有限公司 Chimeric antigen receptor (CAR), composition and its application method
WO2018049025A2 (en) 2016-09-07 2018-03-15 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses
WO2018067991A1 (en) 2016-10-07 2018-04-12 The Brigham And Women's Hospital, Inc. Modulation of novel immune checkpoint targets
US10617720B2 (en) 2016-10-20 2020-04-14 Miltenyi Biotech, GmbH Chimeric antigen receptor specific for tumor cells
US11332713B2 (en) 2016-11-16 2022-05-17 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
KR102122546B1 (en) * 2017-01-05 2020-06-15 한국생명공학연구원 Natural killer cell expressing anti-cotinine chimeric antigen receptor specifically binding to cotinine
EP3567101A4 (en) 2017-01-05 2020-12-02 Korea Research Institute of Bioscience and Biotechnology Natural killer cell expressing anti-cotinine chimeric antigen receptor
EP4353818A2 (en) 2017-02-27 2024-04-17 Juno Therapeutics, Inc. Compositions, articles of manufacture and methods related to dosing in cell therapy
US11963966B2 (en) 2017-03-31 2024-04-23 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating ovarian tumors
EP3606518A4 (en) 2017-04-01 2021-04-07 The Broad Institute, Inc. Methods and compositions for detecting and modulating an immunotherapy resistance gene signature in cancer
JP7284707B2 (en) 2017-04-07 2023-05-31 ジュノー セラピューティクス インコーポレイテッド Engineered Cells Expressing Prostate-Specific Membrane Antigen (PSMA) or Modified Forms Thereof and Related Methods
US20200071773A1 (en) 2017-04-12 2020-03-05 Massachusetts Eye And Ear Infirmary Tumor signature for metastasis, compositions of matter methods of use thereof
JP7355650B2 (en) 2017-04-14 2023-10-03 ジュノー セラピューティクス インコーポレイテッド Methods for assessing cell surface glycosylation
US11344578B2 (en) 2017-04-19 2022-05-31 Board Of Regents, The University Of Texas System Immune cells expressing engineered antigen receptors
EP3638218A4 (en) 2017-06-14 2021-06-09 The Broad Institute, Inc. Compositions and methods targeting complement component 3 for inhibiting tumor growth
SG11202001010UA (en) 2017-08-07 2020-03-30 Univ Johns Hopkins Methods and materials for assessing and treating cancer
US11732257B2 (en) 2017-10-23 2023-08-22 Massachusetts Institute Of Technology Single cell sequencing libraries of genomic transcript regions of interest in proximity to barcodes, and genotyping of said libraries
US11117936B2 (en) 2017-11-10 2021-09-14 University of Pittsburg—Of the Commonwealth System of Higher Education Affinity-enhanced monomeric streptavidin chimeric antigen receptor (CAR)
US20210363260A1 (en) 2017-11-13 2021-11-25 The Broad Institute, Inc. Methods and compositions for treating cancer by targeting the clec2d-klrb1 pathway
WO2019134950A1 (en) 2018-01-04 2019-07-11 Miltenyi Biotec Gmbh Chimeric antigen receptor specific for bdca2 antigen
CA3088832A1 (en) 2018-01-19 2019-07-25 Miltenyi Biotec B.V. & Co. KG Regulatory t cell expressing a chimeric antigen receptor
EP3749695A4 (en) * 2018-02-06 2021-12-29 Seattle Children's Hospital (DBA Seattle Children's Research Institute) Fluorescein-specific cars exhibiting optimal t cell function against fl-ple labelled tumors
EP3759130A1 (en) 2018-02-26 2021-01-06 Fred Hutchinson Cancer Research Center Compositions and methods for cellular immunotherapy
US20190284553A1 (en) 2018-03-15 2019-09-19 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
JP2021518160A (en) 2018-03-15 2021-08-02 ケーエスキュー セラピューティクス, インコーポレイテッド Gene Regulatory Compositions and Methods for Improving Immunotherapy
US20210155986A1 (en) 2018-04-13 2021-05-27 The Johns Hopkins University Non-invasive detection of response to immunotherapy
US20210079384A1 (en) 2018-04-13 2021-03-18 The Johns Hopkins University Non-invasive detection of response to a targeted therapy
US11957695B2 (en) 2018-04-26 2024-04-16 The Broad Institute, Inc. Methods and compositions targeting glucocorticoid signaling for modulating immune responses
WO2019217913A1 (en) 2018-05-10 2019-11-14 Sensei Biotherapeutics, Inc. Aspartate beta-hydroxylase chimeric antigen receptors and uses thereof
WO2019222642A1 (en) * 2018-05-18 2019-11-21 Senti Biosciences, Inc. Engineered immune cells and methods of use
KR20210045953A (en) 2018-05-18 2021-04-27 더 존스 홉킨스 유니버시티 Cell-free DNA for the evaluation and/or treatment of cancer
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
WO2019241315A1 (en) 2018-06-12 2019-12-19 Obsidian Therapeutics, Inc. Pde5 derived regulatory constructs and methods of use in immunotherapy
US20210283180A1 (en) * 2018-07-06 2021-09-16 The Arizona Board Of Regents On Behalf Of The University Of Arizona Car t cell therapy to target t cell specific cancers
JP2021533746A (en) 2018-08-09 2021-12-09 ジュノー セラピューティクス インコーポレイテッド Methods for producing engineered cells and their compositions
CN110856724B (en) * 2018-08-24 2022-05-27 杭州康万达医药科技有限公司 Therapeutic agents comprising nucleic acids and CAR-modified immune cells and uses thereof
MA53612A (en) 2018-09-11 2021-09-15 Juno Therapeutics Inc METHODS FOR ANALYZING MODIFIED CELLULAR COMPOSITIONS BY MASS SPECTROMETRY
WO2020072700A1 (en) 2018-10-02 2020-04-09 Dana-Farber Cancer Institute, Inc. Hla single allele lines
US20220267435A1 (en) * 2018-10-03 2022-08-25 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Covalent adaptor synnotch and chimeric antigen receptors (cars) for programmable antigen-targeting
US20210379057A1 (en) 2018-10-16 2021-12-09 Massachusetts Institute Of Technology Nutlin-3a for use in treating a mycobacterium tuberculosis infection
EP3870600A1 (en) 2018-10-24 2021-09-01 Obsidian Therapeutics, Inc. Er tunable protein regulation
US20220002669A1 (en) 2018-10-31 2022-01-06 Juno Therapeutics Gmbh Methods for selection and stimulation of cells and apparatus for same
WO2020131586A2 (en) 2018-12-17 2020-06-25 The Broad Institute, Inc. Methods for identifying neoantigens
US11739156B2 (en) 2019-01-06 2023-08-29 The Broad Institute, Inc. Massachusetts Institute of Technology Methods and compositions for overcoming immunosuppression
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
WO2020186101A1 (en) 2019-03-12 2020-09-17 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
EP3946382A1 (en) 2019-04-04 2022-02-09 UMC Utrecht Holding B.V. Modified immune receptor constructs
JP2022531577A (en) 2019-05-01 2022-07-07 ジュノー セラピューティクス インコーポレイテッド Cells expressing chimeric receptors from the 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
WO2020236967A1 (en) 2019-05-20 2020-11-26 The Broad Institute, Inc. Random crispr-cas deletion mutant
JP2022534921A (en) 2019-05-28 2022-08-04 ミルテニイ ビオテック ベー.ファー. ウント コー.カーゲー Methods for generating genetically modified T cells
US20220226464A1 (en) 2019-05-28 2022-07-21 Massachusetts Institute Of Technology Methods and compositions for modulating immune responses
WO2021011781A1 (en) * 2019-07-16 2021-01-21 The Regents Of The University Of California Universal chimeric antigen receptors and methods for making and using them
US20220282333A1 (en) 2019-08-13 2022-09-08 The General Hospital Corporation Methods for predicting outcomes of checkpoint inhibition and treatment thereof
WO2021041922A1 (en) 2019-08-30 2021-03-04 The Broad Institute, Inc. Crispr-associated mu transposase systems
WO2021046451A1 (en) 2019-09-06 2021-03-11 Obsidian Therapeutics, Inc. Compositions and methods for dhfr tunable protein regulation
WO2021061648A1 (en) 2019-09-23 2021-04-01 Massachusetts Institute Of Technology Methods and compositions for stimulation of endogenous t cell responses
US11793787B2 (en) 2019-10-07 2023-10-24 The Broad Institute, Inc. Methods and compositions for enhancing anti-tumor immunity by targeting steroidogenesis
KR20220143634A (en) 2019-10-16 2022-10-25 우모자 바이오파마 인코포레이티드 Retroviral Vectors for Universal Receptor Therapy
US11844800B2 (en) 2019-10-30 2023-12-19 Massachusetts Institute Of Technology Methods and compositions for predicting and preventing relapse of acute lymphoblastic leukemia
CN115427550A (en) 2020-01-28 2022-12-02 朱诺治疗学股份有限公司 T cell transduction methods
CN115485295A (en) 2020-03-10 2022-12-16 麻省理工学院 Compositions and methods for immunotherapy of NPM1 c-positive cancers
US20230149460A1 (en) 2020-03-10 2023-05-18 Massachusetts Institute Of Technology Methods for generating engineered memory-like nk cells and compositions thereof
KR20230004680A (en) 2020-04-17 2023-01-06 시티 오브 호프 FLT3-targeted chimeric antigen receptor modified cells for the treatment of FLT3-positive malignancies
US20210338833A1 (en) 2020-05-01 2021-11-04 Massachusetts Institute Of Technology Chimeric antigen receptor-targeting ligands and uses thereof
WO2021221783A1 (en) 2020-05-01 2021-11-04 Massachusetts Institute Of Technology Methods for identifying chimeric antigen receptor-targeting ligands and uses thereof
CN115916250A (en) 2020-05-20 2023-04-04 美天施生物科技有限两合公司 Compositions and methods for treating CD90 and CD326 expressing cancers
WO2021260186A1 (en) 2020-06-26 2021-12-30 Juno Therapeutics Gmbh Engineered t cells conditionally expressing a recombinant receptor, related polynucleotides and methods
EP4182339A1 (en) 2020-07-16 2023-05-24 Umoja Biopharma, Inc. Gated adapter targeting receptor
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
EP4228660A1 (en) * 2020-10-14 2023-08-23 Georgia Tech Research Corporation Synthetic antigens as chimeric antigen receptor (car) ligands and uses thereof
EP4240403A2 (en) 2020-11-04 2023-09-13 Miltenyi Biotec B.V. & Co. KG Chimeric antigen receptor comprising an antigen binding domain specific for msln having a specificity for tumor cells
CN116802203A (en) 2020-11-04 2023-09-22 朱诺治疗学股份有限公司 Cells expressing chimeric receptors from modified constant CD3 immunoglobulin superfamily chain loci, related polynucleotides and methods
WO2022096664A1 (en) 2020-11-09 2022-05-12 Miltenyi Biotec B.V. & Co. KG Methods and compositions for eliminating engineered immune cells
KR20230159851A (en) 2021-03-22 2023-11-22 주노 쎄러퓨티크스 인코퍼레이티드 How to Determine the Potency of a Therapeutic Cell Composition
KR20230158573A (en) 2021-03-22 2023-11-20 주노 쎄러퓨티크스 인코퍼레이티드 Methods for Assessing Potency of Viral Vector Particles
JP2024517287A (en) * 2021-05-07 2024-04-19 ユニバーシティ オブ フロリダ リサーチ ファンデーション インコーポレーティッド CAR T-cell therapy method
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
WO2023081715A1 (en) 2021-11-03 2023-05-11 Viracta Therapeutics, Inc. Combination of car t-cell therapy with btk inhibitors and methods of use thereof
WO2023107825A1 (en) * 2021-12-07 2023-06-15 The Regents Of The University Of California Synthetic cytokine circuits to promote infiltration and clearance of immune excluded solid tumors by engineered immune 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
US20230372395A1 (en) 2022-05-19 2023-11-23 Massachusetts Institute Of Technology Car cells targeting an inserted ligand
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 (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5906936A (en) 1988-05-04 1999-05-25 Yeda Research And Development Co. Ltd. Endowing lymphocytes with antibody specificity
US5861156A (en) 1993-01-08 1999-01-19 Creative Biomolecules Methods of delivering agents to target cells
US20090011984A1 (en) 1998-02-23 2009-01-08 Seppo Yla-Herttuala Biotin-binding receptor molecules
EP1064026A4 (en) 1998-03-10 2003-04-09 Univ Pennsylvania Enhancement of intracellular delivery and tissue targeting of drugs and genes
JP2001213804A (en) 2000-01-31 2001-08-07 Chugai Pharmaceut Co Ltd Antitissue factor antibody complex
AU2001271273A1 (en) 2000-05-24 2001-12-03 Ludwig Institute For Cancer Research Multicomponent conjugates which bind to target molecules and stimulate cell lysis
DE60133287T2 (en) 2000-07-31 2009-03-26 Green Peptide Co., Ltd., Kurume TUMOR ANTIGEN
US7514537B2 (en) * 2001-04-30 2009-04-07 City Of Hope Chimeric immunoreceptor useful in treating human gliomas
US7446190B2 (en) * 2002-05-28 2008-11-04 Sloan-Kettering Institute For Cancer Research Nucleic acids encoding chimeric T cell receptors
US20050129671A1 (en) * 2003-03-11 2005-06-16 City Of Hope Mammalian antigen-presenting T cells and bi-specific T cells
TWI342312B (en) 2003-10-22 2011-05-21 Theravance Inc Hydrochloride salts of a glycopeptide phosphonate derivative
US20050113564A1 (en) * 2003-11-05 2005-05-26 St. Jude Children's Research Hospital Chimeric receptors with 4-1BB stimulatory signaling domain
CN100376599C (en) * 2004-04-01 2008-03-26 北京安波特基因工程技术有限公司 Recombining single chained three specific antibodies of anti CCA, anti CD 3, anti CD 28 through genetic engineering
JP5564181B2 (en) 2005-12-08 2014-07-30 シルワン,ハヴァル Immune stimulating compositions and methods
US20080003225A1 (en) 2006-06-29 2008-01-03 Henri Vie Method for enhancing the antibody-dependent cellular cytotoxicity (ADCC) and uses of T cells expressing CD16 receptors
US8865169B2 (en) 2007-02-20 2014-10-21 Tufts University Methods and systems for multi-antibody therapies
PL3006459T3 (en) * 2008-08-26 2022-01-17 City Of Hope Method and compositions for enhanced anti-tumor effector functioning of t cells
CA2742777A1 (en) 2008-11-06 2010-05-14 University Of Guelph Methods of improving the therapeutic efficacy and utility of antibody fragments
WO2011041093A1 (en) 2009-10-01 2011-04-07 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-vascular endothelial growth factor receptor-2 chimeric antigen receptors and use of same for the treatment of cancer
ES2791716T3 (en) 2010-12-14 2020-11-05 Univ Maryland T cells expressing the universal anti-label chimeric antigen receptor and methods for the treatment of cancer
WO2013044225A1 (en) 2011-09-22 2013-03-28 The Trustees Of The University Of Pennsylvania A universal immune receptor expressed by t cells for the targeting of diverse and multiple antigens

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules

Non-Patent Citations (25)

* Cited by examiner, † Cited by third party
Title
"Genbank", Database accession no. NM 000734.3
"Genbank", Database accession no. NM 001561.5
"Genbank", Database accession no. NM 001768.6
"Genbank", Database accession no. NM 006139.2
"Molecular Biology and Biotechnology: a Comprehensive Desk Reference", 1995, WILEY, JOHN & SONS, INC.
"The Encyclopedia of Molecular Biology", 1994, BLACKWELL PUBLISHERS
BENJAMIN LEWIN: "Genes VII", 2000, OXFORD UNIVERSITY PRESS
BRENTJENS ET AL., NAT. MED., vol. 9, 2003, pages 279 - 286
COLE ET AL.: "Monoclonal Antibodies and Cancer Therapy", 1985, ALAN R. LISS INC, pages: 77 - 96
COTE ET AL., PROC NATL. ACAD. SCI, vol. 80, 1983, pages 2026 - 2030
ERTL ET AL., CANCER RES., vol. 71, 2011, pages 3175 - 3181
GROSS ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 10024 - 10028
HUGHES M.S. ET AL.: "Transfer of a TCR gene derived from a patient with a marked antitumor response conveys highly active T-cell effector functions", HUM GENE THER, vol. 16, no. 4, April 2005 (2005-04-01), pages 457 - 72, XP055509137, DOI: doi:10.1089/hum.2005.16.457
JOHNSON ET AL., BLOOD, vol. 114, 2009, pages 535 - 546
KERSHAW ET AL., CLIN. CANCER RES., vol. 12, 2006, pages 6106 - 6115
KOEHLERMILSTEIN, NATURE, vol. 256, 1975, pages 495 - 497
KOSBOR ET AL., IMMUNOL TODAY, vol. 4, 1983, pages 72
LAMERS ET AL., J. CLIN. ONCOL., vol. 24, 2006, pages e20 - e22
MORGAN ET AL., MOL. THER., vol. 18, 2010, pages 843 - 851
MORRISON ET AL., PROC NATL. ACAD. SCI, vol. 81, 1984, pages 6851 - 6855
NEUBERGER ET AL., NATURE, vol. 312, 1984, pages 604 - 608
PULE ET AL., NAT. MED., vol. 14, 2008, pages 1264 - 1270
SADELAIN ET AL., CURR. OPIN. IMMUNOL., vol. 21, 2009, pages 215 - 223
TAKEDA ET AL., NATURE, vol. 314, 1985, pages 452 - 454
TILL ET AL., BLOOD, vol. 112, 2008, pages 2261 - 2271

Cited By (165)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8975071B1 (en) 2010-12-09 2015-03-10 The Trustees Of The University Of Pennsylvania Compositions for treatment of cancer
US9102760B2 (en) 2010-12-09 2015-08-11 The Trustees Of The University Of Pennsylvania Compositions for treatment of cancer
US9518123B2 (en) 2010-12-09 2016-12-13 The Trustees Of The University Of Pennsylvania Compositions and methods for treatment of cancer
US9499629B2 (en) 2010-12-09 2016-11-22 The Trustees Of The University Of Pennsylvania Use of chimeric antigen receptor-modified T-cells to treat cancer
US8906682B2 (en) 2010-12-09 2014-12-09 The Trustees Of The University Of Pennsylvania Methods for treatment of cancer
US8911993B2 (en) 2010-12-09 2014-12-16 The Trustees Of The University Of Pennsylvania Compositions for treatment of cancer
US8916381B1 (en) 2010-12-09 2014-12-23 The Trustees Of The University Of Pennyslvania Methods for treatment of cancer
US9540445B2 (en) 2010-12-09 2017-01-10 The Trustees Of The University Of Pennsylvania Compositions and methods for treatment of cancer
US9328156B2 (en) 2010-12-09 2016-05-03 The Trustees Of The University Of Pennsylvania Use of chimeric antigen receptor-modified T cells to treat cancer
US9464140B2 (en) 2010-12-09 2016-10-11 The Trustees Of The University Of Pennsylvania Compositions and methods for treatment of cancer
US9481728B2 (en) 2010-12-09 2016-11-01 The Trustees Of The University Of Pennsylvania Compositions and methods for treatment of cancer
US9102761B2 (en) 2010-12-09 2015-08-11 The Trustees Of The University Of Pennsylvania Compositions for treatment of cancer
US9101584B2 (en) 2010-12-09 2015-08-11 The Trustees Of The University Of Pennsylvania Methods for treatment of cancer
EP2651442B1 (en) 2010-12-14 2020-04-22 University of Maryland, Baltimore Universal anti-tag chimeric antigen receptor-expressing t cells and methods of treating cancer
US10266580B2 (en) 2011-09-22 2019-04-23 The Trustees Of The University Of Pennsylvania Universal immune receptor expressed by T cells for the targeting of diverse and multiple antigens
WO2013044225A1 (en) * 2011-09-22 2013-03-28 The Trustees Of The University Of Pennsylvania A universal immune receptor expressed by t cells for the targeting of diverse and multiple antigens
US11912753B2 (en) 2011-09-22 2024-02-27 The Trustees Of The University Of Pennsylvania Universal immune receptor expressed by T cells for the targeting of diverse and multiple antigens
US11041012B2 (en) 2011-09-22 2021-06-22 The Trustees Of The University Of Pennsylvania Universal immune receptor expressed by T cells for the targeting of diverse and multiple antigens
EP2872218A4 (en) * 2012-07-13 2016-07-06 Univ Pennsylvania Compositions and methods for regulating car t cells
AU2013289984B2 (en) * 2012-07-13 2018-03-08 The Trustees Of The University Of Pennsylvania Use of CART19 to deplete normal B cells to induce tolerance
EP2872184A4 (en) * 2012-07-13 2016-03-30 Univ Pennsylvania Use of cart19 to deplete normal b cells to induce tolerance
WO2014011987A1 (en) * 2012-07-13 2014-01-16 The Trustees Of The University Of Pennsylvania Compositions and methods for regulating car t cells
JP2018135363A (en) * 2012-07-13 2018-08-30 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Use of cart19 to deplete normal b cells to induce tolerance
CN104507537A (en) * 2012-07-13 2015-04-08 宾夕法尼亚大学董事会 Compositions and methods for regulating car t cells
JP2015523386A (en) * 2012-07-13 2015-08-13 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 Use of CART19 to deplete normal B cells to induce tolerance
AU2018203756B2 (en) * 2012-07-13 2019-10-24 The Trustees Of The University Of Pennsylvania Use of cart19 to deplete normal b cells to induce tolerance
EP3653212A1 (en) * 2012-12-20 2020-05-20 Purdue Research Foundation Chimeric antigen receptor-expressing t cells as anti-cancer therapeutics
EP4282419A1 (en) * 2012-12-20 2023-11-29 Purdue Research Foundation Chimeric antigen receptor-expressing t cells as anti-cancer therapeutics
WO2014100615A1 (en) * 2012-12-20 2014-06-26 Purdue Research Foundation Chimeric antigen receptor-expressing t cells as anti-cancer therapeutics
EP2934532A4 (en) * 2012-12-20 2016-12-07 Purdue Research Foundation Chimeric antigen receptor-expressing t cells as anti-cancer therapeutics
US11028177B2 (en) 2013-02-20 2021-06-08 Novartis Ag Effective targeting of primary human leukemia using anti-CD123 chimeric antigen receptor engineered T cells
US11865167B2 (en) 2013-02-20 2024-01-09 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
US9394368B2 (en) 2013-02-20 2016-07-19 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
US9573988B2 (en) 2013-02-20 2017-02-21 Novartis Ag Effective targeting of primary human leukemia using anti-CD123 chimeric antigen receptor engineered T cells
US10308717B2 (en) 2013-02-20 2019-06-04 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
EP2961831A4 (en) * 2013-02-26 2016-10-05 Sloan Kettering Inst Cancer Compositions and methods for immunotherapy
US11103531B2 (en) 2013-02-26 2021-08-31 Memorial Sloan-Kettering Cancer Center Compositions and methods for immunotherapy
US10124023B2 (en) 2013-02-26 2018-11-13 Memorial Sloan-Kettering Cancer Center Compositions and methods for immunotherapy
WO2014134165A1 (en) 2013-02-26 2014-09-04 Memorial Sloan-Kettering Cancer Center Compositions and methods for immunotherapy
CN113684185A (en) * 2013-02-26 2021-11-23 纪念斯隆-凯特琳癌症中心 Compositions and methods for immunotherapy
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
US10640553B2 (en) 2013-03-15 2020-05-05 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US10221245B2 (en) 2013-03-16 2019-03-05 Novartis Ag Treatment of cancer using humanized anti-CD19 chimeric antigen receptor
US10927184B2 (en) 2013-03-16 2021-02-23 Novartis Ag Treatment of cancer using humanized anti-CD19 chimeric antigen receptor
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
US10428142B2 (en) 2013-07-29 2019-10-01 Bluebird 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
WO2015017214A1 (en) 2013-07-29 2015-02-05 Bluebird Bio, Inc. Multipartite signaling proteins and uses thereof
AU2014337385B2 (en) * 2013-10-15 2020-04-30 The Scripps Research Institute Chimeric antigen receptor T cell switches and uses thereof
CN105814083A (en) * 2013-10-15 2016-07-27 加州生物医学研究所 Chimeric antigen receptor T cell switches and uses thereof
WO2015057852A1 (en) * 2013-10-15 2015-04-23 The California Institute For Biomedical Research Chimeric antigen receptor t cell switches and uses thereof
US20150307564A1 (en) * 2013-10-15 2015-10-29 The California Institute For Biomedical Research Peptidic chimeric antigen receptor t cell switches and uses thereof
CN105829349B (en) * 2013-10-15 2023-02-03 斯克利普斯研究所 Peptide chimeric antigen receptor T cell switches and uses thereof
KR102357968B1 (en) * 2013-10-15 2022-02-03 더 스크립스 리서치 인스티튜트 Chimeric antigen receptor t cell switches and uses thereof
KR20160062761A (en) * 2013-10-15 2016-06-02 더 캘리포니아 인스티튜트 포 바이오메디칼 리써치 Chimeric antigen receptor t cell switches and uses thereof
US10391155B2 (en) 2013-10-15 2019-08-27 The Scripps Research Institute Peptidic chimeric antigen receptor T cell switches and uses thereof
CN105829349A (en) * 2013-10-15 2016-08-03 加州生物医学研究所 Peptidic chimeric antigen receptor t cell switches and uses thereof
JP2016533174A (en) * 2013-10-15 2016-10-27 ザ カリフォルニア インスティテュート フォー バイオメディカル リサーチ Peptide chimeric antigen receptor T cell switch and use thereof
JP2016534995A (en) * 2013-10-15 2016-11-10 ザ カリフォルニア インスティテュート フォー バイオメディカル リサーチ Chimeric antigen receptor T cell switch and use thereof
US9624276B2 (en) 2013-10-15 2017-04-18 The California Institute For Biomedical Research Peptidic chimeric antigen receptor T cell switches and uses thereof
AU2014337367B2 (en) * 2013-10-15 2020-04-30 The Scripps Research Institute Peptidic chimeric antigen receptor T cell switches and uses thereof
US10640569B2 (en) 2013-12-19 2020-05-05 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
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
JP2017501702A (en) * 2013-12-20 2017-01-19 フレッド ハッチンソン キャンサー リサーチ センター Tagged chimeric effector molecule and its receptor
US11028143B2 (en) 2014-01-21 2021-06-08 Novartis Ag Enhanced antigen presenting ability of RNA CAR T cells by co-introduction of costimulatory molecules
US10357514B2 (en) 2014-04-07 2019-07-23 The Trustees Of The University Of Pennsylvania Treatment of cancer using anti-CD19 Chimeric Antigen Receptor
CN105194661B (en) * 2014-06-26 2019-05-03 科济生物医药(上海)有限公司 The system of space-time adjustable inhibition pathologic target cell
CN105194661A (en) * 2014-06-26 2015-12-30 科济生物医药(上海)有限公司 System for inhibiting pathological target cells in space-time adjustable manner
WO2015197016A1 (en) * 2014-06-26 2015-12-30 科济生物医药(上海)有限公司 Time and space adjustable system for inhibiting pathological target cells
US11084880B2 (en) 2014-07-21 2021-08-10 Novartis Ag Anti-BCMA chimeric antigen receptor
US10568947B2 (en) 2014-07-21 2020-02-25 Novartis Ag Treatment of cancer using a CLL-1 chimeric antigen receptor
US10851166B2 (en) 2014-07-21 2020-12-01 Novartis Ag Treatment of cancer using a CD33 chimeric antigen receptor
US11542488B2 (en) 2014-07-21 2023-01-03 Novartis Ag Sortase synthesized chimeric antigen receptors
US10174095B2 (en) 2014-07-21 2019-01-08 Novartis Ag Nucleic acid encoding a humanized anti-BCMA chimeric antigen receptor
US9777061B2 (en) 2014-07-21 2017-10-03 Novartis Ag Treatment of cancer using a CD33 chimeric antigen receptor
CN113846062A (en) * 2014-07-25 2021-12-28 赛拉福柯蒂斯公司 Lentiviral vectors for regulated expression of chimeric antigen receptor molecules
US10703819B2 (en) 2014-08-09 2020-07-07 The Trustees Of The University Of Pennsylvania Treatment of cancer using a CD123 chimeric antigen receptor
US9815901B2 (en) 2014-08-19 2017-11-14 Novartis Ag Treatment of cancer using a CD123 chimeric antigen receptor
US11591404B2 (en) 2014-08-19 2023-02-28 Novartis Ag Treatment of cancer using a CD123 chimeric antigen receptor
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
EP2990416A1 (en) 2014-08-29 2016-03-02 GEMoaB Monoclonals GmbH Universal chimeric antigen receptor expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
AU2015308499B2 (en) * 2014-08-29 2020-01-02 Avencell Europe Gmbh Universal chimeric antigen receptor expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
US10766943B2 (en) 2014-08-29 2020-09-08 Gemoab Monoclonals Gmbh Universal chimeric antigen expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
WO2016030414A1 (en) * 2014-08-29 2016-03-03 Gemoab Monoclonals Gmbh Universal chimeric antigen receptor expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
US10577417B2 (en) 2014-09-17 2020-03-03 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US10774388B2 (en) 2014-10-08 2020-09-15 Novartis Ag Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
US11883494B2 (en) 2014-11-26 2024-01-30 Miltenyi Biotec B.V. & Co. KG Combination of immune effector cells specific for a target antigen and hematopoietic calls that express the target antigen in an altered form
US10273300B2 (en) 2014-12-29 2019-04-30 The Trustees Of The University Of Pennsylvania Methods of making chimeric antigen receptor-expressing cells
US11459390B2 (en) 2015-01-16 2022-10-04 Novartis Ag Phosphoglycerate kinase 1 (PGK) promoters and methods of use for expressing chimeric antigen receptor
US11161907B2 (en) 2015-02-02 2021-11-02 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
US10800828B2 (en) 2015-03-26 2020-10-13 The Scripps Research Institute Switchable non-scFv chimeric receptors, switches, and methods of use thereof to treat cancer
US11149076B2 (en) 2015-04-08 2021-10-19 Novartis Ag CD20 therapies, CD22 therapies, and combination therapies with a CD19 chimeric antigen receptor (CAR)-expressing cell
US10253086B2 (en) 2015-04-08 2019-04-09 Novartis Ag CD20 therapies, CD22 therapies, and combination therapies with a CD19 chimeric antigen receptor (CAR)-expressing cell
US11091546B2 (en) 2015-04-15 2021-08-17 The Scripps Research Institute Optimized PNE-based chimeric receptor T cell switches and uses thereof
EP3283113A4 (en) * 2015-04-15 2018-12-05 The California Institute for Biomedical Research Optimized pne-based chimeric receptor t cell switches and uses thereof
US20190169289A1 (en) * 2015-04-15 2019-06-06 The California Institute For Biomedical Research Optimized pne-based chimeric receptor t cell switches and uses thereof
US11896614B2 (en) 2015-04-17 2024-02-13 Novartis Ag Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
US10829735B2 (en) 2015-07-21 2020-11-10 The Trustees Of The University Of Pennsylvania Methods for improving the efficacy and expansion of immune cells
US11667691B2 (en) 2015-08-07 2023-06-06 Novartis Ag Treatment of cancer using chimeric CD3 receptor proteins
US11747346B2 (en) 2015-09-03 2023-09-05 Novartis Ag Biomarkers predictive of cytokine release syndrome
EP3816294A1 (en) * 2015-09-11 2021-05-05 Biosceptre (UK) Limited Chimeric antigen receptors and uses thereof
WO2017041143A1 (en) * 2015-09-11 2017-03-16 Ctm@Crc Ltd. Chimeric antigen receptors and uses thereof
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
EP3380602A4 (en) * 2015-11-23 2019-09-25 Trustees of Boston University Methods and compositions relating to chimeric antigen receptors
US11413340B2 (en) 2015-12-22 2022-08-16 Novartis Ag Mesothelin chimeric antigen receptor (CAR) and antibody against PD-L1 inhibitor for combined use in anticancer therapy
US11225520B2 (en) 2016-02-16 2022-01-18 Dana-Farber Cancer Institute, Inc. Immunotherapy compositions and methods
US11549099B2 (en) 2016-03-23 2023-01-10 Novartis Ag Cell secreted minibodies and uses thereof
US10525083B2 (en) 2016-10-07 2020-01-07 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
USRE49847E1 (en) 2016-10-07 2024-02-27 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
US11872249B2 (en) 2016-10-07 2024-01-16 Novartis Ag Method of treating cancer by administering immune effector cells expressing a chimeric antigen receptor comprising a CD20 binding domain
US11026976B2 (en) 2016-10-07 2021-06-08 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
US11174306B2 (en) 2016-10-19 2021-11-16 The Scripps Research Institute Chimeric antigen receptor effector cell switches with humanized targeting moieties and/or optimized chimeric antigen receptor interacting domains and uses thereof
WO2018078066A1 (en) 2016-10-29 2018-05-03 Miltenyi Biotec Gmbh Adapter chimeric antigen receptor expressing cells for targeting of multiple antigens
EP3315511A1 (en) 2016-10-29 2018-05-02 Miltenyi Biotec GmbH Adapter chimeric antigen receptor expressing cells for targeting of multiple antigens
US20190388468A1 (en) * 2016-12-15 2019-12-26 Miltenyi Biotec Gmbh Immune cells expressing an antigen binding receptor and a chimeric costimulatory receptor
US11890300B2 (en) * 2016-12-15 2024-02-06 Miltenyi Biotec B.V. & Co. KG Immune cells expressing an antigen binding receptor and a chimeric costimulatory receptor
US11535662B2 (en) 2017-01-26 2022-12-27 Novartis Ag CD28 compositions and methods for chimeric antigen receptor therapy
US11649288B2 (en) 2017-02-07 2023-05-16 Seattle Children's Hospital Phospholipid ether (PLE) CAR T cell tumor targeting (CTCT) agents
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
US11851659B2 (en) 2017-03-22 2023-12-26 Novartis Ag Compositions and methods for immunooncology
WO2018224660A1 (en) 2017-06-09 2018-12-13 Armin Ehninger Targeting modules for universal chimeric antigen receptor expressing immune cells and use in the treatment of cancer infections and autoimmune disorders
US11560426B2 (en) 2017-06-09 2023-01-24 Avencell Europe Gmbh Targeting modules for universal chimeric antigen receptor expressing immune cells and use in the treatment of cancer infections and autoimmune disorders
WO2019060425A1 (en) 2017-09-19 2019-03-28 Massachusetts Institute Of Technology Compositions for chimeric antigen receptor t cell therapy and uses thereof
GB2585607A (en) * 2017-11-29 2021-01-13 Guangzhou Cas Lamvac Biotech Co Ltd Chimeric antigen receptor and application thereof
GB2585607B (en) * 2017-11-29 2022-09-07 Cas Lamvac Guangzhou Biomedical Tech Co Ltd Chimeric antigen receptor and application thereof
WO2019104562A1 (en) * 2017-11-29 2019-06-06 广州中科蓝华生物科技有限公司 Chimeric antigen receptor and application thereof
US11952413B2 (en) 2017-12-14 2024-04-09 2Seventy Bio, Inc. Dimerizing agent regulated immunoreceptor complexes comprising interleukin receptor signaling domains
US11311576B2 (en) 2018-01-22 2022-04-26 Seattle Children's Hospital Methods of use for CAR T cells
US11779602B2 (en) 2018-01-22 2023-10-10 Endocyte, Inc. Methods of use for CAR T cells
US11939389B2 (en) 2018-06-13 2024-03-26 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11952428B2 (en) 2018-06-13 2024-04-09 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11608382B2 (en) 2018-06-13 2023-03-21 Novartis Ag BCMA chimeric antigen receptors and uses thereof
EP3581200A1 (en) 2018-06-13 2019-12-18 GEMoaB Monoclonals GmbH Reversed universal chimeric antigen receptor expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
WO2019238722A1 (en) 2018-06-13 2019-12-19 Gemoab Monoclonals Gmbh Reversed universal chimeric antigen receptor expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
EP3620464A1 (en) 2018-09-10 2020-03-11 Miltenyi Biotec GmbH Car cell having crosslinked disulfide bridge on antigen recognizing moiety
WO2020068261A1 (en) 2018-09-28 2020-04-02 Massachusetts Institute Of Technology Collagen-localized immunomodulatory molecules and methods thereof
CN111454909A (en) * 2019-01-22 2020-07-28 北京大学深圳研究生院 Preparation method of natural ligand-mediated multi-target recognition controllable genetically engineered immune cells
KR20220004087A (en) 2019-04-19 2022-01-11 추가이 세이야쿠 가부시키가이샤 Antibody modification site recognition chimeric receptor
WO2020213724A1 (en) 2019-04-19 2020-10-22 中外製薬株式会社 Chimeric receptor recognizing modification site of antibody
WO2020263399A1 (en) 2019-06-26 2020-12-30 Massachusetts Institute Of Technology Immunomodulatory fusion protein-metal hydroxide complexes and methods thereof
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
EP4093768A4 (en) * 2019-10-18 2023-11-15 Trustees of Boston University Cal-t constructs and uses thereof
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
EP3878464A1 (en) 2020-03-09 2021-09-15 Miltenyi Biotec B.V. & Co. KG Use of a car cell having crosslinked disulfide bridge on antigen recognizing moiety for targeting cancer cells
WO2021185807A1 (en) 2020-03-16 2021-09-23 GEMoaB GmbH A targeting module comprising pd-l1 and/or pd-l2 for use in a method for stimulating a chimeric antigen receptor mediated immune response in a mammal
EP3881866A1 (en) 2020-03-16 2021-09-22 GEMoaB GmbH A targeting module comprising pd-l1 and/or pd-l2 for use in a method for stimulating a chimeric antigen receptor mediated response in a mammal
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
KR20230048059A (en) 2020-07-31 2023-04-10 추가이 세이야쿠 가부시키가이샤 Pharmaceutical composition comprising cells expressing chimeric receptors
WO2022025220A1 (en) 2020-07-31 2022-02-03 中外製薬株式会社 Pharmaceutical composition including cell expressing chimeric receptor
WO2022038411A1 (en) 2020-08-19 2022-02-24 Astellas Pharma, Inc. Human non-naturally occurring modified fc region of igg specifically binding to non-naturally occurring modified fc receptor
WO2022049217A1 (en) 2020-09-04 2022-03-10 Miltenyi Biotec B.V. & Co. KG System for inducible expression of an adapter in immune cells
US11883432B2 (en) 2020-12-18 2024-01-30 Century Therapeutics, Inc. Chimeric antigen receptor system with adaptable receptor specificity
WO2022133169A1 (en) * 2020-12-18 2022-06-23 Century Therapeutics, Inc. Chimeric antigen receptor systems with adaptable receptor specificity
WO2022258711A1 (en) * 2021-06-08 2022-12-15 Biontech Cell & Gene Therapies Gmbh Agents and methods for activation and targeting of immune effector cells
WO2022258681A1 (en) * 2021-06-08 2022-12-15 Biontech Cell & Gene Therapies Gmbh Agents and methods for activation and targeting of immune effector cells
WO2023004461A1 (en) * 2021-07-28 2023-02-02 Prescient Therapeutics Ltd Universal receptor immune cell therapy
WO2023079135A1 (en) 2021-11-08 2023-05-11 Avencell Europe Gmbh TARGETING MODULES AGAINST IL13Rα2 AND/OR HER2 FOR USE IN A METHOD FOR STIMULATING A CHIMERIC ANTIGEN RECEPTOR-MEDIATED IMMUNE RESPONSE IN A MAMMAL
EP4176895A1 (en) 2021-11-08 2023-05-10 AvenCell Europe GmbH Targeting modules against il13ra2 or her2 for use in combination with a chimeric antigen receptor
WO2024078995A1 (en) 2022-10-15 2024-04-18 Miltenyi Biotec B.V. & Co. KG Transduction of gammadelta t cells with pseudotyped retroviral vectors

Also Published As

Publication number Publication date
JP5897035B2 (en) 2016-03-30
AU2011343887A1 (en) 2013-07-04
US20160129109A1 (en) 2016-05-12
AU2011343887B2 (en) 2016-06-16
WO2012082841A9 (en) 2012-08-09
ES2791716T3 (en) 2020-11-05
US10973893B2 (en) 2021-04-13
CA3102782A1 (en) 2012-06-21
US20210330769A1 (en) 2021-10-28
US20130287752A1 (en) 2013-10-31
EP2651442A4 (en) 2016-06-01
CA2821080A1 (en) 2012-06-21
CA2821080C (en) 2021-02-02
EP2651442B1 (en) 2020-04-22
JP2014504294A (en) 2014-02-20
EP3693017A1 (en) 2020-08-12
EP2651442A2 (en) 2013-10-23
US9233125B2 (en) 2016-01-12

Similar Documents

Publication Publication Date Title
US20210330769A1 (en) Universal anti-tag chimeric antigen receptor-expressing t cells and methods of treating cancer
US20220204619A1 (en) Chimeric antigen receptors targeting cd-19
US20210309740A1 (en) Bispecific chimeric antigen receptors and therapeutic uses thereof
US20210206828A9 (en) 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
US10822419B2 (en) Masking chimeric antigen receptor T cells for tumor-specific activation
WO2017219934A1 (en) Killer cell capable of efficiently and stably expressing antibody, and uses thereof
WO2017219936A1 (en) Car-t cell capable of efficiently and stably expressing activated antibody, and uses thereof
Wu et al. HER2-specific chimeric antigen receptor-engineered natural killer cells combined with apatinib for the treatment of gastric cancer
EP4190804A1 (en) Chimeric antigen receptor (car) t cell stabilizing immune synapse
JP2022514477A (en) PSCA CAR-T cells

Legal Events

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

Ref document number: 11849221

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2821080

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2013544725

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13993396

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2011343887

Country of ref document: AU

Date of ref document: 20111214

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2011849221

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2011849221

Country of ref document: EP