WO2014055668A1 - Compositions and methods for immunotherapy - Google Patents

Compositions and methods for immunotherapy Download PDF

Info

Publication number
WO2014055668A1
WO2014055668A1 PCT/US2013/063097 US2013063097W WO2014055668A1 WO 2014055668 A1 WO2014055668 A1 WO 2014055668A1 US 2013063097 W US2013063097 W US 2013063097W WO 2014055668 A1 WO2014055668 A1 WO 2014055668A1
Authority
WO
WIPO (PCT)
Prior art keywords
antigen
cell
receptor
immunoresponsive
tumor
Prior art date
Application number
PCT/US2013/063097
Other languages
French (fr)
Inventor
Christopher C. KLOSS
Michael Sadelain
Original Assignee
Memorial Sloan-Kettering Cancer Center
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=50435409&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2014055668(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to MX2015004287A priority Critical patent/MX370148B/en
Priority to EP19179456.9A priority patent/EP3597215A1/en
Priority to EP13844468.2A priority patent/EP2903637B1/en
Priority to CN201380063036.8A priority patent/CN104853766A/en
Priority to ES13844468T priority patent/ES2743738T3/en
Priority to JP2015534828A priority patent/JP6441802B2/en
Priority to NZ706541A priority patent/NZ706541A/en
Priority to RU2015116901A priority patent/RU2729401C2/en
Priority to SG11201502598SA priority patent/SG11201502598SA/en
Priority to KR1020157011160A priority patent/KR102198058B1/en
Priority to CA2886859A priority patent/CA2886859C/en
Application filed by Memorial Sloan-Kettering Cancer Center filed Critical Memorial Sloan-Kettering Cancer Center
Priority to AU2013327136A priority patent/AU2013327136A1/en
Publication of WO2014055668A1 publication Critical patent/WO2014055668A1/en
Priority to IL238047A priority patent/IL238047B/en
Priority to US14/676,255 priority patent/US10654928B2/en
Priority to PH12015500747A priority patent/PH12015500747A1/en
Priority to ZA201502880A priority patent/ZA201502880B/en
Priority to HK15109439.3A priority patent/HK1208631A1/en
Priority to AU2018204297A priority patent/AU2018204297B2/en
Priority to PH12019502424A priority patent/PH12019502424A1/en
Priority to IL272539A priority patent/IL272539B/en
Priority to US16/847,059 priority patent/US11712469B2/en
Priority to AU2020294287A priority patent/AU2020294287B2/en
Priority to US18/333,753 priority patent/US20240091353A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • 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
    • A61K35/15Cells of the myeloid line, e.g. granulocytes, basophils, eosinophils, neutrophils, leucocytes, monocytes, macrophages or mast cells; Myeloid precursor cells; Antigen-presenting cells, e.g. dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001103Receptors for growth factors
    • A61K39/001106Her-2/neu/ErbB2, Her-3/ErbB3 or Her 4/ErbB4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001103Receptors for growth factors
    • A61K39/001109Vascular endothelial growth factor receptors [VEGFR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001111Immunoglobulin superfamily
    • A61K39/001113CD22, BL-CAM, siglec-2 or sialic acid- binding Ig-related lectin 2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001111Immunoglobulin superfamily
    • A61K39/001114CD74, Ii, MHC class II invariant chain or MHC class II gamma chain
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001116Receptors for cytokines
    • A61K39/001117Receptors for tumor necrosis factors [TNF], e.g. lymphotoxin receptor [LTR] or CD30
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001116Receptors for cytokines
    • A61K39/001119Receptors for interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001124CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001128CD44 not IgG
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001129Molecules with a "CD" designation not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001152Transcription factors, e.g. SOX or c-MYC
    • A61K39/001153Wilms tumor 1 [WT1]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001154Enzymes
    • A61K39/001157Telomerase or TERT [telomerase reverse transcriptase]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001166Adhesion molecules, e.g. NRCAM, EpCAM or cadherins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001166Adhesion molecules, e.g. NRCAM, EpCAM or cadherins
    • A61K39/001168Mesothelin [MSLN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001169Tumor associated carbohydrates
    • A61K39/00117Mucins, e.g. MUC-1
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001169Tumor associated carbohydrates
    • A61K39/001171Gangliosides, e.g. GM2, GD2 or GD3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00118Cancer antigens from embryonic or fetal origin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00118Cancer antigens from embryonic or fetal origin
    • A61K39/001182Carcinoembryonic antigen [CEA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001184Cancer testis antigens, e.g. SSX, BAGE, GAGE or SAGE
    • A61K39/001186MAGE
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001184Cancer testis antigens, e.g. SSX, BAGE, GAGE or SAGE
    • A61K39/001188NY-ESO
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001193Prostate associated antigens e.g. Prostate stem cell antigen [PSCA]; Prostate carcinoma tumor antigen [PCTA]; PAP or PSGR
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001193Prostate associated antigens e.g. Prostate stem cell antigen [PSCA]; Prostate carcinoma tumor antigen [PCTA]; PAP or PSGR
    • A61K39/001195Prostate specific membrane antigen [PSMA]
    • 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
    • A61K39/44Antibodies bound to carriers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464493Prostate associated antigens e.g. Prostate stem cell antigen [PSCA]; Prostate carcinoma tumor antigen [PCTA]; Prostatic acid phosphatase [PAP]; Prostate-specific G-protein-coupled receptor [PSGR]
    • 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
    • A61K39/464493Prostate associated antigens e.g. Prostate stem cell antigen [PSCA]; Prostate carcinoma tumor antigen [PCTA]; Prostatic acid phosphatase [PAP]; Prostate-specific G-protein-coupled receptor [PSGR]
    • A61K39/464495Prostate specific membrane antigen [PSMA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/14Drugs for genital or sexual disorders; Contraceptives for lactation disorders, e.g. galactorrhoea
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70517CD8
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70521CD28, CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3069Reproductive system, e.g. ovaria, uterus, testes, prostate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • 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
    • C12N5/0638Cytotoxic T lymphocytes [CTL] or lymphokine activated killer cells [LAK]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/24Hydrolases (3) acting on glycosyl compounds (3.2)
    • C12N9/2402Hydrolases (3) acting on glycosyl compounds (3.2) hydrolysing O- and S- glycosyl compounds (3.2.1)
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/74Inducing cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • 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
    • C12N2999/00Further aspects of viruses or vectors not covered by groups C12N2710/00 - C12N2796/00 or C12N2800/00
    • C12N2999/002Adverse teaching

Definitions

  • Immunotherapy is a targeted therapy that in principle provides for the treatment of such cancers .
  • Targeted T ceil therapies utilizing genetically modified autologous T cells are beginning to show evidence of therapeutic efficacy in melanoma and indolent B cell malignancies.
  • Current T ceil engineering strategies retarget patient T cells to tumor antigens through a transduced T cell receptor (TCR) or a chimeric antigen receptor (CAR).
  • TCR transduced T cell receptor
  • CAR chimeric antigen receptor
  • the present invention generally provides immunoresponsive cells, including T ceils and Natural Killer (NK) cells, expressing an antigen binding receptor (e.g., CAR or TCR) having immune cell activating actvity and a chimeric co-stimulating receptor (CCR), and methods of use therefore for the treatment of neoplasia, infectious disease, and other pathologies.
  • an antigen binding receptor e.g., CAR or TCR
  • CCR chimeric co-stimulating receptor
  • the invention provides an isolated immunoresponsive cell having an antigen recognizing receptor that binds a first antigen with low affinity, where the binding activates the immunoresponsive ceil, and a chimeric co stimulating receptor (CCR) that binds a second antigen and stimulates the immunoresponsive ceil.
  • CCR chimeric co stimulating receptor
  • the invention provides a method of inducing tumor cell death in a subject, the method comprising administering an effective amount of an immunoresponsive cell comprising an antigen recognizing receptor that binds a first antigen with low affinity, where the binding activates the immunoresponsive cell, and a chimeric co-stimulating receptor (CCR) that binds a second antigen and stimulates the immunoresponsive cell, thereby inducing tumor cell death in the subject.
  • an immunoresponsive cell comprising an antigen recognizing receptor that binds a first antigen with low affinity, where the binding activates the immunoresponsive cell, and a chimeric co-stimulating receptor (CCR) that binds a second antigen and stimulates the immunoresponsive cell, thereby inducing tumor cell death in the subject.
  • CCR chimeric co-stimulating receptor
  • the invention provides a method of treating or preventing a neoplasia in a subject, the method comprising administering an effective amount of an immunoresponsive cell comprising an antigen recognizing receptor that binds a first antigen with low affinity, where the binding activates the immunoresponsive ceil, and a chimeric co-stimulating receptor (CCR) that binds a second antigen and stimulates the immunoresponsive cell, thereby treating or preventing a neoplasia in the subject.
  • an immunoresponsive cell comprising an antigen recognizing receptor that binds a first antigen with low affinity, where the binding activates the immunoresponsive ceil, and a chimeric co-stimulating receptor (CCR) that binds a second antigen and stimulates the immunoresponsive cell, thereby treating or preventing a neoplasia in the subject.
  • CCR chimeric co-stimulating receptor
  • the invention provides a method of treating prostate cancer in a subject in need thereof, the method comprising
  • a T cell comprising an antigen recognizing receptor that binds PSCA or CD19 with low affinity, where the binding activates the immunoresponsive cell, and a chimeric co-stimulating receptor (CCR) that binds PSMA and stimulates the
  • the invention provides a method for producing an antigen- specific immunoresponsive ceil, the method involving introducing into the immunoresponsive cell a nucleic acid sequence that encodes a chimeric co- stimulating receptor (CCR), where the chimeric co-stimulating receptor has an antigen-binding domain coupled to an intracellular signaling domain that stimulates an immunoresponsive cell, where the immunoresponsive cell has an antigen recognizing receptor that binds a first antigen with low affinity, wherein the binding activates the immunoresponsive cell.
  • CCR chimeric co- stimulating receptor
  • the invention provides a pharmaceutical composition comprising an effective amount of an immunoresponsive ceil of the invention (e.g., a tumor antigen-specific T cell in a pharmaceutical composition for the treatment of neoplasia) in a pharmaceutically acceptable excipient.
  • an immunoresponsive ceil of the invention e.g., a tumor antigen-specific T cell in a pharmaceutical composition for the treatment of neoplasia
  • the invention provides a kit for treatment of a neoplasia, pathogen infection, an autoimmune disorder, or an allogeneic transplant, the kit containing an immunoresponsive cell having an antigen recognizing receptor that binds a first antigen and activates the
  • kits may further comprise written instructions for using the immunoresponsive cell for the treatment of a subject having a neoplasia, a pathogen infection, an autoimmune disorder, or an allogeneic transplant.
  • the immunoresponsive cell is selected as having an antigen recognizing receptor with low affinity. This may involve selecting the immunoresponsive cell as having an antigen recognizing receptor that binds a first antigen with low affinity, in various embodiments of any of the aspects delineated herein, the antigen recognizing receptor is selected as having low affinity for expression in the cell. This may involve introducing a second nucleic acid sequence that encodes a chimeric antigen receptor, where the chimeric antigen receptor comprises a second antigen-binding domain coupled to a second intracellular signaling domain that activates an immunoresponsive cell.
  • the antigen recognizing receptor is a T cell receptor (TCR) or chimeric antigen receptor (CAR).
  • TCR T cell receptor
  • CAR chimeric antigen receptor
  • the intracellular signaling domain of said antigen recognizing receptor is the CD3-chain signaling domain.
  • the intracellular signaling domain of the chimeric co-stimulating receptor (CCR) is a CD97, CD1 1a-CD18, CD2, ICOS, CD27, CD154, CD5, OX40, 4-1 BB or CD28 signaling domain.
  • the antigen recognizing receptor is exogenous or endogenous.
  • the antigen recognizing receptor is recombinantly expressed.
  • the antigen recognizing receptor is expressed from a vector.
  • the chimeric co-stimulating receptor (CCR) is expressed from a vector.
  • the immunoresponsive cell expresses a recombinant or an
  • endogenous antigen receptor that is 19z1 or Pz1.
  • the immunoresponsive cell is a T cell, a Natural Killer (NK) cell, a cytotoxic T
  • lymphocyte CTL
  • regulatory T cell a human embryonic stem ceil
  • human embryonic stem ceil a human embryonic stem ceil
  • the antigen is a tumor or pathogen antigen.
  • one or more antigen-binding domains are tumor antigen-binding domains.
  • the antigens or tumor antigens are selected from CAIX, CEA, CD5, CD7, CD10, CD19, CD20, CD22, CD30, CD33, CD34, CD38, CD41 , CD44, CD49f, CD56, CD74, CD133, CD138, a cytomegalovirus (CMV) infected ceil antigen, EGP-2, EGP-40, EpCAM, erb-B2,3,4, FBP, Fetal acetylcholine receptor, folate receptor-a, GD2, GD3, HER-2, hTERT, IL13R-a2, x-iight chain, KDR, LeY, Li cell adhesion molecule, MAGE-AI , MUC1 , Mesothelin, NKG2D iigands, NY- ES0-1 , oncofetal antigen (h5T4), PSCA, PSMA, ROR1 , TAG-72, VEGF-
  • CMV cytomegalovirus
  • the first and second antigens are selected from CD133, a cytomegalovirus (CMV) infected cell antigen, erbB2, KDR Mesothelin, NKG2D Iigands, NY-ES0-1 , oncofetal antigen (h5T4), PSCA, PSMA, CD19, VEGF-R2, and WT-1.
  • the first and second antigens are selected from HER2, MUC1 , CD44, CD49f, EpCAM, CEA, CD133, a
  • cytomegalovirus (CMV) infected cell antigen EGP-2, EGP-40, EpCAM, erb-B2,3,4, FBP, KDR, Mesothelin, NKG2D Iigands, NY-ES0-1 , oncofetal antigen (h5T4), PSCA, PSMA, VEGF-R2, or WT-1.
  • the first and second antigens are selected from CD10 and CD19.
  • the first and second antigens are selected from CD56 and CD138.
  • the first and second antigens are selected from mesothelin, folate receptor-a, CD44, and CD133.
  • the neoplasia is selected from the group consisting of prostate cancer, breast cancer, B cell leukemia, multiple myeloma, and ovarian cancer.
  • the method reduces the number of tumor cells, reduces tumor size, and/or eradicates the tumor in the subject.
  • the neoplasia is prostate cancer and the first and second tumor antigens are distinct antigens selected from PSCA, PSMA, CD19, CD133, a cytomegalovirus (CMV) infected cell antigen, erb-B2, KDR
  • the neoplasia is breast cancer and the first and second tumor antigens are distinct antigens selected from HER2, MUC1 , CD44, CD49f, EpCAM, CEA, CD133, a cytomegalovirus (CMV) infected cell antigen, EGP-2, EGP-40, EpCAM, erb-B2,3,4, FBP, KDR, Mesothelin, NKG2D iigands, NY- ESO-1 , oncofetal antigen (h5T4), PSCA, PSMA, VEGF-R2, or WT-1 .
  • CMV cytomegalovirus
  • the neoplasia is B cell leukemia and the first and second tumor antigens are selected from CDIO and CD19.
  • the neoplasia is multiple myeloma and the first and second tumor antigens are selected from CD56 and CD138.
  • the neoplasia is ovarian cancer and the first and second tumor antigens are distinct antigens selected from mesothe!in, folate receptor-a, CD44, and CD133.
  • compositions and methods that provide for T ceil targeting of tumor cells.
  • Compositions and articles defined by the invention were isolated or otherwise manufactured in connection with the examples provided below. Other features and advantages of the invention will be apparent from the detailed description, and from the claims.
  • Figures 1A-C are graphics depicting chimeric antigen receptor (CAR) and chimeric costimulatory receptor (CCR) vector design and expression via transduction of primary human T cells.
  • CAR chimeric antigen receptor
  • CCR costimulatory receptor
  • CAR expression can be easily detected by correlation to dsRED fluorescence (data not shown).
  • the CCR was generated by fusing an scFv to a CD28 transmembrane and signaling domain 15 , fused to a 4-1 BB (aka CD137) cytosoiic signaling domain. 21 CCR expression can be correlated to the bicistronic expression of hrGFP (data not shown).
  • LTR Long Terminal Repeat
  • SD Splice Donor site
  • SA Splice Acceptor site
  • VH or V L -Variable Heavy or Light domains respectively
  • EC Extracellular domain
  • TM Transmembrane domain
  • C Cytosoiic domain
  • IRES Internal Ribosomal Entry Site
  • hrGFP Human
  • Recombinant Green Fluorescent Protein depicts representative transduction efficiencies of primary human T cells using these retroviral vectors.
  • C depicts transduction of CTLs with different and multiple CARs for the present studies.
  • Figures 2A-D show that dual-receptor, CAR/CCR-mediated activation of human T cells allowed for robust CTL function, long-term proliferation, and enhanced tumor eradication upon binding of two antigens.
  • A shows that T cells expressing chimeric receptors lysed cells positive for antigen when the
  • CAR specific to CD19 is expressed by T cells in CTL assays, compared to untransduced or P28BB transduced T cells.
  • Plots are representative of n > 4 experiments, with error bars representing standard deviation of the mean of 3 replicates.
  • (B) shows long-term proliferation of T cells by absolute T cell counts over 31 days of T cells expressing none, one, or both chimeric receptors that were co-cultured with human tumor cell lines expressing both or either antigen alone. Arrows indicate re-stimulation of T cells using freshly irradiated tumor cells. Only when dual-receptor expressing T ceils encounter both antigens is robust long-term proliferation observed.
  • Plots are representative of n > 4 experiments with error bars representing standard deviation of the mean of 3 replicates.
  • TTS tumor- sensing T
  • IV intravenously
  • (D) depicts selective eradication of DP tumors using a tri-tumor mouse model by subcutaneously injecting 1 x 10 6 PC3 tumors cells each of ceils positive for CD19 alone into the left flanks, cells positive for PSMA alone into the right flanks, and cells positive for both CD19 and PSMA into the backs of the mice.
  • T ceils expressing either 19z1 , P28BB, or both 19z1 + P28BB of the chimeric receptors were infused intravenously 7 days post tumor infusion. Representative images of 2 mice per group bearing these tumors are shown with luminescence of tumors represented in color. Tumors were quantitatively measured using calipers and tumor volumes were plotted versus time for each tumor. Error bars represent standard deviation from the mean of 6 mice.
  • Statistical significance was determined using two- tailed unpaired t tests to compare values obtained from 19z1 T cells and 19z1 + P28BB T cells and p values are represented as * for ⁇ 0.05 or ** for ⁇ 0.01.
  • FIGS 3A-E depict that tumor-sensing T (T TS ) ceils selectively eradicated human prostate tumors when targeting two prostate tumor antigens
  • T TS tumor-sensing T
  • A depicts the evaluation of three different scFvs specific to PSCA for their assembly into bispecific antibodies that contain specificity for CD3 as well. T ceils were co- cultured at ratio of 20:1 with PSCA + PC3 tumor cells and antibodies added at varying amounts and specific iysis was measured.
  • (B) depicts generation of CARs using the anti-PSCA scFvs that display varied efficacy in cytotoxicity assays.
  • Figures 4A-D depict enhanced cytokine secretion and Bd xL
  • TTS ceils when co-cultured on DP tumors.
  • A depicts multiplex cytokine analysis of untransduced T cells or T cells transduced with 19z1 , P28BB, or both 48 hours post first antigen stimulation using either untransduced PC3 cells (Empty) or CD19 + PSMA + PC3 ceils. Error bars represent standard deviation from the mean of 2 biological replicates.
  • B-D depict multiplex cytokine analysis of untransduced T cells or T cells transduced with Hz] (B), Mzl (C), and Lz1 (D) anti-PSCA CARs, P28BB CCRs, or both CAR + CCR is shown 48 hours post second antigen stimulation using either Empty or PSCA+PSMA+ PC3 cells.
  • E depicts Western blot analysis for BdxL performed using cellular lysates of untransduced T ceils or T ceils transduced with 19z1 , P28BB, or both after 24 hours post initial antigen stimulation. Total amount of Akt was used as a loading control.
  • FIG. 5 depicts generation of prostate tumor cells for the expression of fusion protein GFP-Firefiy Luciferase (GFP/Luc) and tumor antigens.
  • Untransduced PC3 cells (Empty) were transduced with GFP/Luc and either CD19, PSMA, PSCA, or a combination of two antigens using retroviral expression constructs. Cells were purified via double purity FACS for GFP/Luc, CD19, PSMA, and/or PSCA.
  • FIG. 6A-C illustrate the tumor-sensing T cell concept.
  • A depicts that TTS cells expressing an efficient CAR, become potently stimulated by A + 113 + cells to facilitate immune response against A + ceils.
  • CAR + CCR + cells can bind tumor antigen A + cells with a CAR that supplies CD3 activation signals. This can result in short-term cell lysis.
  • CAR + CCR + cells can bind tumor antigen B + cells with a CCR that supplies CD28 and CD137 signals. This signal alone is not sufficient to induce lysis or proliferation. Only when CAR + CCR + cells bind tumor antigen A + B + ceils with a CAR and CCR can both activation and stimulation be provided.
  • FIG. 6B depicts that by reducing the efficacy of the CAR, TTs celis can be functionally rescued by CCR binding when A + B + cells are encountered to selectively respond and eradicate A + 13 + ceils, while avoiding response to A+ cells.
  • (C) shows that by co-expressing one CAR that supplies a TCR activation signal upon binding a tumor antigen and a second CAR that supplies stimulation signals upon binding a different tumor antigen, T lymphocytes will only eradicate tumors expressing both antigens, but not tumors expressing either antigen alone.
  • an immunoresponsive ceil activates induction of signal transduction or changes in protein expression in the cell resulting in initiation of an immune response.
  • CD3 Chains cluster in response to ligand binding and immunoreceptor tyrosine-based inhibition motifs (ITAMs) a signal transduction cascade is produced.
  • ITAMs immunoreceptor tyrosine-based inhibition motifs
  • a formation of an immunologicai synapse occurs that includes clustering of many molecuies near the bound receptor (e.g. CD4 or CD8, CD3 / / / , etc.) This clustering of membrane bound signaling molecules allows for ITAM motifs contained within the CD3 chains to become phosphorylated.
  • T ceil activation pathway ultimately activating transcription factors, such as NF-KB and AP-1.
  • transcription factors induce global gene expression of the T cell to increase IL-2 production for proliferation and express master regulator T cell proteins in order to initiate a T ceil mediated immune response.
  • Stimulates an immunoresponsive cell is meant a signal that results in a robust and sustained immune response. In various embodiments, this occurs after immune cell (e.g., T-cell) activation or concomitantly mediated through receptors including, but not limited to, CD28, CD137 (4-1 BB), OX40, and ICOS.
  • receiving multiple stimulatory signals is important to mount a robust and long-term T cell mediated immune response.
  • T cells Without receiving these stimulatory signals, T cells quickly become inhibited and unresponsive to antigen. While the effects of these co-stimuiatory signals vary and remain partially understood, they generally result in increasing gene expression in order to generate long lived, proliferative, and anti-apoptotic T cells that robustly respond to antigen for complete and sustained eradication.
  • antigen recognizing receptor refers to a receptor that is capable of activating an immune cell (e.g., a T-cell) in response to antigen binding.
  • exemplary antigen recognizing receptors may be native or endogenous T cell receptors or chimeric antigen receptors in which a tumor antigen-binding domain is fused to an intracellular signaling domain capable of activating activating an immune cell (e.g., a T-cell).
  • an antigen recognizing receptor is selected to have low or minimal affinity or avidity for the antigen.
  • affinity is meant a measure of the binding strength between antibody and a simple hapten or antigen determinant. Without being bound to theory, affinity depends on the closeness of stereochemical fit between antibody combining sites and antigen determinants, on the size of the area of contact between them, and on the distribution of charged and hydrophobic groups. Affinity also includes the term “avidity,” which refers to the strength of the antigen-antibody bond after formation of reversible complexes. Methods for calculating the affinity of an antibody for an antigen are known in the art, including use of binding experiments to calculate affinity. In the case of an antibody (Ab) binding to an antigen (Ag), the affinity constant is used (expressed as inverted dissociation constant).
  • the chemical equilibrium of antibody binding is also the ratio of the on-rate (k forward) and off-rate (kback) constants.
  • Two antibodies can have the same affinity, but one may have both a high on- and off-rate constant, while the other may have both a low on- and off-rate constant.
  • Antibody activity in functionai assays is also reflective of antibody affinity.
  • the antigen recognizing receptor has low affinity.
  • Low affinity includes micromolar and nanomolar affinities (e.g. IfJ 5 , 50 "6 , I0 “6 , 5x 1fJ 7 , 10 "7 , 5x 10 "8 , 10 “8 , 5x1 fJ 9 , iO "9 M).
  • Anitbody and affinities can be phenotypically characterized and compared using functional assay (e.g., cell lysis assay).
  • chimeric co-stimulatory receptor refers to a specific type of chimeric antigen receptor (CAR) that mediates costimulation independently of activation.
  • CAR chimeric antigen receptor
  • the CCR When expressed on immunoresponsive cells in combination with an antigen recognizing receptor (e.g., CAR or TCR that activates the cell), the CCR is targeted to a second antigen.
  • the CCR has mid or high affinity for its target antigen.
  • chimeric antigen receptor refers to a tumor antigen-binding domain that is fused to an intracellular signaling domain capable of activating or stimulating T ceils. Most commonly, the CAR'S
  • extracellular binding domain is composed of a single chain variable fragment (scFv) derived from fusing the variable heavy and light regions of a murine or humanized monoclonal antibody.
  • scFv's may be used that are derived from Fab's (instead of from an antibody, e.g., obtained from Fab libraries).
  • this scFv is fused to a transmembrane domain and then to intracellular signaling domain.
  • “First-generation” CARs include those that solely provide CD3 signals upon antigen binding
  • “Second-generation” CARs include those that provide both costimulation (e.g. CD28 or CD137) and activation (CD3 ).
  • “Third-generation” CARs include those that provide multiple
  • CD3 polypeptide is meant a protein having at least 85, 90, 95, 96, 97, 98, 99 or 100% identity to NCBI Reference No: NP_932170 or a fragment thereof that has activating or stimulatory activity.
  • An exemplary CD3 is provided in Table 1 below.
  • CD3 nucleic acid molecule is meant a polynucleotide encoding a CD3 polypeptide.
  • CD8 polypeptide is meant a protein having at least 85, 90, 95, 96, 97, 98, 99or 100% identity to NCBI Reference No: NP_001759 or a fragment thereof that has stimulatory activity.
  • An exemplary CD8 is provided in Table 1 below.
  • CD8 nucleic acid molecule is meant a polynucleotide encoding a CD8 polypeptide.
  • CD28 polypeptide is meant a protein having at least 85, 90, 95, 96, 97, 98, 99 or 100% identity to NCBI Reference No: NP_006130 or a fragment thereof that has stimulatory activity.
  • An exemplary CD28 is provided in Table 1 below.
  • CD28 nucleic acid molecule is meant a polynucleotide encoding a CD28 polypeptide.
  • 4-1 BB polypeptide is meant a protein having at least 85, 90, 95,
  • 4-1 BBL nucleic acid molecule is meant a polynucleotide encoding a 4-1 BBL polypeptide.
  • CD80 polypeptide is meant a protein having at least 85, 90, 95, 96,
  • CD80 polypeptide is provided in Table 1 below.
  • CD80 nucleic acid molecule any polynucleotide encoding a CD80 polypeptide.
  • An exemplary CD80 nucleic acid molecule is NM_005191 .
  • OX40L polypeptide is meant a protein having at least 85, 90, 95, 96,
  • OX40L nucleic acid molecule is meant a polynucleotide encoding a OX40L polypeptide.
  • 19z1 polypeptide is meant a protein having at least 85, 90, 95, 96, 97, 98, 99 or 100% identity to the sequence provided below and having activating activity when bound to CD19.
  • P28z polypeptide is meant a protein having at least 85, 90, 95, 96, 97, 98, 99 or 100% identity to the sequence provided below.
  • CD19 is meant a protein having at least 85, 90, 95, 96, 97, 98, 99 or 100% identity to the sequence provided below and is able to bind CD19.
  • PSMA is meant a protein having at least 85, 90, 95, 96, 97, 98, 99 or 100% identity to the sequence provided below and is able to bind PSMA.
  • P28BB is meant a protein having at least 85, 90, 95, 96, 97, 98, 99 or 100% identity to the sequence provided below and having stimulatory activity when bound to PSMA.
  • CD3 ⁇ mkwkalftaa ilqaqlpite aqsfglldpk lcylldgilf iygviltalf Irvkfsrsad apayqqgqnq lynelnlgrr eeydvldkrr grdpemggkp qrrknpqegl ynelqkdkma eayseigmkg errrgkghdg lyqglsta tkdtydalhm qalppr
  • CD80 evkevatlsc ghnvsveela qtriywqkek kmvltmmsgd mni peyknr tifditnnls ivilalrpsd egtyecvvlk yekdafkreh laevtlsvka dfptpsisdf eiptsnirri icstsggfpe phlswlenge elnainttvs qdpetelyav sskldfnmtt nhsfmcliky ghlrvnqtfn wnttkqehfp dnllpswait lisvngif i ccltycfapr crerrrnerl rresvrov mervqpleen vgnaarprfe rnklllvasv iqglgl ll
  • GYAFSSYWMNWKQRPGQGLE IGQIYPGDGDTNYNGKFKGQA TLTADKSSSTAYMQLSGLTSEDSAVYECARKTISSWDFYFDY WGQGTTVTVSSGGGGSGGGGSGGGGSDIELTQSPKFMSTSVGD RVSVTCKASQ VGT VAWYQQKPGQSPKPLIYSATYRNSGVPD RFTGSGSGTDFTLTITNVQSKDLADYFCQQYNRYPYTSGGGTK LEIKRAAAPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGA VHTRGLDFACDIYI APLAGTCGVLLLSLVITLYCNHRVKFSR SAEPPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKP RRK PQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQG LSTATKDTYDALHMQALPPR
  • Nucleic acid molecules useful in the methods of the invention include any nucleic acid molecule that encodes a polypeptide of the invention or a fragment thereof. Such nucleic acid molecules need not be 100% identical with an endogenous nucleic acid sequence, but will typically exhibit substantial identity. Polynucleotides having "substantial identity" to an endogenous
  • hybridize is meant pair to form a double- stranded molecule between complementary polynucleotide sequences (e.g., a gene described herein), or portions thereof, under various conditions of stringency.
  • complementary polynucleotide sequences e.g., a gene described herein
  • stringent salt concentration will ordinarily be less than about 750 mM NaCI and 75 mM trisodium citrate, preferably less than about 500 mM NaC1 and 50 mM trisodium citrate, and more preferably less than about 250 mM NaC1 and 25 mM trisodium citrate.
  • Low stringency hybridization can be obtained in the absence of organic solvent, e.g., formamide, while high
  • stringency hybridization can be obtained in the presence of at least about 35% formamide, and more preferably at least about 50% formamide.
  • Stringent temperature conditions wiil ordinarily inciude temperatures of at least about 30° C, more preferably of at least about 37° C, and most preferably of at least about 42° C.
  • Varying additional parameters, such as hybridization time, the concentration of detergent, e.g., sodium dodecyl sulfate (SDS), and the inclusion or exclusion of carrier DNA are well known to those skilled in the art.
  • Various levels of stringency are accomplished by combining these various conditions as needed. In a preferred: embodiment, hybridization will occur at 30° C in 750 mM NaCI, 75 mM trisodium citrate, and 1% SDS.
  • hybridization will occur at 37° C in 500 mM NaCI, 50 mM trisodium citrate, 1 % SDS, 35% formamide, and 1001.1 g/m1 denatured salmon sperm DNA (ssDNA).
  • hybridization wiil occur at 42° C C. in 250 mM NaCI, 25 mM trisodium citrate, 1 % SDS, 50% formamide, and 200 p.g/ml ssDNA. Useful variations on these conditions will be readily apparent to those skilled in the art.
  • wash stringency conditions can be defined by salt concentration and by temperature. As above, wash stringency can be increased by decreasing salt concentration or by increasing temperature.
  • stringent salt concentration for the wash steps will preferably be less than about 30 mM NaC1 and 3 mM trisodium citrate, and most preferably less than about 15 mM NaCI and 1.5 mM trisodium citrate.
  • Stringent temperature conditions for the wash steps wiil ordinarily inciude a temperature of at least about 25° C, more preferably of at least about 42° C, and even more preferably of at least about 68° C.
  • wash steps wiil occur at 25°C in 30 mM NaCI, 3 mM trisodium citrate, and 0.1% SDS.
  • wash steps will occur at
  • wash steps wiil occur at 68° C in 15 mM NaC1 , 1.5 mM trisodium citrate, and 0.1% SDS. Additional variations on these conditions will be readily apparent to those skilled in the art.
  • Hybridization techniques are well known to those skilled in the art and are described, for example, in Benton and Davis (Science 196:180, 1977); Grunstein and Hogness (Proc. Natl. Acad.Sci., USA 72:3961 , 1975); Ausubel et al. (Current Protocols in Molecular Biology, Wiley Interscience, New York, 2001 ); Berger and Kimmei (Guide to Molecular Cloning Techniques, 1987, Academic Press, New York); and Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, New York.
  • substantially identical is meant a polypeptide or nucleic acid molecule exhibiting at least 50% identity to a reference amino acid sequence (for example, any one of the amino acid sequences described herein) or nucleic acid sequence (for example, any one of the nucleic acid sequences described herein).
  • a reference amino acid sequence for example, any one of the amino acid sequences described herein
  • nucleic acid sequence for example, any one of the nucleic acid sequences described herein.
  • such a sequence is at least 60%, more preferably 80% or 85%, and more preferably 90%, 95% or even 99% identical at the amino acid level or nucleic acid to the sequence used for comparison.
  • Sequence identity is typically measured using sequence analysis software (for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, BESTFIT, GAP, or PILEUP/PRETTYBOX programs). Such software matches identical or similar sequences by assigning degrees of homology to various substitutions, deletions, and/or other modifications.
  • sequence analysis software for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, BESTFIT, GAP, or PILEUP/PRETTYBOX programs.
  • Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparagine, g!utamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine.
  • a BLAST program may be used, with a probability score between e-3 and e-100 indicating a closely related sequence.
  • analog is meant a structurally related polypeptide or nucleic acid molecule having the function of a reference polypeptide or nucleic acid molecule.
  • iigand refers to a molecule that binds to a receptor.
  • the iigand binds a receptor on another cell, allowing for cell-to-cel! recognition.
  • disease is meant any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ.
  • diseases include neoplasia or pathogen infection of cell.
  • effective amount is meant an amount sufficient to arrest, ameliorate, or inhibit the continued proiiferation, growth, or metastasis (e.g., invasion, or migration) of a neoplasia.
  • enforcing tolerance is meant preventing the activity of self-reactive cells or immunoresponsive cells that target transplanted organs or tissues.
  • exogenous is meant a nucleic acid molecule or polypeptide that is not endogenously present in the cell, or not present at a level sufficient to achieve the functional effects obtained when over-expressed.
  • exogenous would therefore encompass any recombinant nucleic acid molecule or polypeptide expressed in a cell, such as foreign, heterologous, and over- expressed nucleic acid molecules and polypeptides.
  • nucleic acid molecule or polypeptide a nucleic acid molecule (e.g., a cDNA, DNA or RNA molecule) or polypeptide that is not normally present in a cell or sample obtained from a cell.
  • This nucleic acid may be from another organism, or it may be, for example, an mRNA molecule that is not normally expressed in a cell or sample.
  • immunoresponsive cell is meant a cell that functions in an immune response or a progenitor, or progeny thereof.
  • isolated cell is meant a ceil that is separated from the molecular and/or cellular components that naturally accompany the cell.
  • isolated refers to material that is free to varying degrees from components which normally accompany it as found in its native state.
  • Isolate denotes a degree of separation from original source or surroundings.
  • Purify denotes a degree of separation that is higher than isolation.
  • A"purified” or “biologically pure” protein is sufficiently free of other materials such that any impurities do not materially affect the biological properties of the protein or cause other adverse consequences. That is, a nucleic acid or peptide of this invention is purified if it is substantially free of cellular material, viral material, or culture medium when produced by
  • purified can denote that a nucleic acid or protein gives rise to essentially one band in an electrophoretic gel.
  • modifications for example, phosphorylation or glycosylation, different modifications may give rise to different isolated proteins, which can be separately purified.
  • tumor antigen-binding domain refers to a domain capable of specifically binding a particular antigenic determinant or set of antigenic determinants present on a tumor.
  • modulate is meant to alter positively or negatively.
  • exemplary modulations include a 1%, 2%, 5%, 10%, 25%, 50%, 75%, or 100% change.
  • Neoplasia is meant a disease characterized by the pathological proliferation of a cell or tissue and its subsequent migration to or invasion of other issues or organs. Neoplasia growth is typically uncontrolled and progressive, and occurs under conditions that would not elicit, or would cause cessation of, multiplication of normal cells.
  • Neoplasias can affect a variety of cell types, tissues, or organs, including but not limited to an organ selected from the group consisting of bladder, bone, brain, breast, cartilage, glia, esophagus, fallopian tube, gallbladder, heart, intestines, kidney, liver, lung, lymph node, nervous tissue, ovaries, pancreas, prostate, skeletal muscle, skin, spinal cord, spleen, stomach, testes, thymus, thyroid, trachea, urogenital tract, ureter, urethra, uterus, and vagina, or a tissue or cell type thereof.
  • an organ selected from the group consisting of bladder, bone, brain, breast, cartilage, glia, esophagus, fallopian tube, gallbladder, heart, intestines, kidney, liver, lung, lymph node, nervous tissue, ovaries, pancreas, prostate, skeletal muscle, skin, spinal cord, spleen, stomach, teste
  • Neoplasias include cancers, such as sarcomas, carcinomas, or plasmacytomas ⁇ malignant tumor of the plasma cells).
  • Illustrative neoplasms for which the invention can be used include, but are not limited to leukemias (e.g., acute Ieukemia, acute lymphocytic Ieukemia, acute myelocytic leukemi a, acute myeloblasts Ieukemia, acute promyelocytic ieukemia, acute myelomonocytic ieukemia, acute monocytic Ieukemia, acute erythroieukemia, chronic Ieukemia, chronic myelocytic Ieukemia, chronic lymphocytic Ieukemia), polycythemia vera, lymphoma (Hodgkin's disease, non-Hodgkin's disease),
  • leukemias e.g., acute Ieukemia, acute lymphocytic Ieukemia, acute myelocy
  • Waldenstrom's macroglobulinemia, heavy chain disease, and solid tumors such as sarcomas and carcinomas ⁇ e.g., fibrosarcoma, myxosarcoma,
  • liposarcoma chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma,iymphangiosarcoma,lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous giand carcinoma, papillary carcinoma, papillary
  • adenocarcinomas cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal ceil carcinoma, hepatoma, nile duct carcinoma,
  • choriocarcinoma seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, uterine cancer, testicular cancer, lung carcinoma, small ceil lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medullobiastoma, craniopharyngioma, ependymoma, pinealoma,
  • screening methods of the invention identify compositions that are useful for treating breast or lung cancer.
  • receptor is meant a polypeptide, or portion thereof, present on a cell membrane that selectively binds one or more ligands.
  • a T cell that recognizes a virus typically expresses a receptor that binds an antigen expressed by the virus.
  • Retroviridae e.g. human immunodeficiency viruses, such as HIV-1 (also referred to as HDTV-ill, LAVE or HTLV-III/LAV, or HIV-III; and other isolates, such as HIV- LP; Picornaviridae (e.g. polio viruses, hepatitis A virus; enteroviruses, human Coxsackie viruses, rhinoviruses, echoviruses); Calciviridae (e.g. strains that cause gastroenteritis); Togaviridae (e.g. equine encephalitis viruses, rubella viruses); Flaviridae (e.g. dengue viruses, encephalitis viruses, yellow fever viruses);
  • Retroviridae e.g. human immunodeficiency viruses, such as HIV-1 (also referred to as HDTV-ill, LAVE or HTLV-III/LAV, or HIV-III; and other isolates, such as HIV- LP; Picornaviridae (e.g.
  • Coronoviridae e.g. coronaviruses
  • Rhabdoviridae e.g. vesicular stomatitis viruses, rabies viruses
  • Filoviridae e.g. ebola viruses
  • Paramyxoviridae e.g. parainfluenza viruses, mumps virus, measles virus, respiratory syncytial virus
  • Orthomyxoviridae e.g . influenza viruses
  • Bungaviridae e.g. Hantaan viruses, bunga viruses, phieboviruses and Nairo viruses
  • Arena Viridae hemorrhagic fever viruses
  • Reoviridae e.g. reoviruses, orbiviurses and
  • rotaviruses rotaviruses
  • S/ ' ma r/ ' c/ae Hepadnaviridae (Hepatitis B virus); Parvovihda ⁇ parvov ⁇ ruses);Papovaviridae (papilloma viruses, polyoma viruses);
  • Adenoviridae most adenoviruses
  • Herpesviridae herpes simplex virus (HSV) 1 and 2, varicella zoster virus, cytomegalovirus (CMV), herpes virus
  • Poxviridae variola viruses, vaccinia viruses, pox viruses
  • Iridoviridae e.g. African swine fever virus
  • Exemplary bacteria include, but are not limited to, Pasteurella, Staphylococci, Streptococcus, Escherichia coli, Pseudomonas species, and Salmonella species.
  • Specific examples of infectious bacteria include but are not limited to, Helicobacter pyloris, Borelia burgdoiferi, Legionella
  • pneumophilia e.g. M. tuberculosis, M. avium, M.
  • M. kansaii M. gordonae
  • Staphylococcus aureus Staphylococcus aureus
  • Neisseria gonorrhoeae Neisseria meningitidis
  • Listeria monocytogenes Listeria monocytogenes
  • Streptococcus pyo genes (Group A Streptococcus), Streptococcus agalactiae (Group B Streptococcus), Streptococcus (viridans group), Streptococcus faecalis, Streptococcus bovis, Streptococcus (anaerobic sps.), Streptococcus pneumoniae, pathogen cCampylobacter sp.,
  • corynebacterium diphtheriae corynebacterium sp., Erysipelothrix rhusiopathiae, Clostridium perfringers, Clostridium tetani, Enterobacter aerogenes, Klebsiella pneumoniae, Pasturella multocida, Bacteroides sp., Fusobacterium nucleatum, Streptobacillus moniliformis, Treponema pallidium, Treponema peramba, Leptospira, Rickettsia, and Actinomyces israelii.
  • polypeptide or fragment thereof that recognizes and binds a polypeptide of interest, but which does not
  • a sample for example, a biological sample, which naturally includes a polypeptide of the invention.
  • tumor antigen refers to any polypeptide expressed by a tumor that is capable of inducing an immune response.
  • virus antigen is meant a polypeptide expressed by a virus that is capable of inducing an immune response.
  • treatment refers to clinical intervention in an attempt to alter the disease course of the individual or cell being treated, and can be performed either for prophylaxis or during the course of clinical pathology.
  • Therapeutic effects of treatment include, without limitation, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastases, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • a treatment can prevent deterioration due to a disorder in an affected or diagnosed subject or a subject suspected of having the disorder, but also a treatment may prevent the onset of the disorder or a symptom of the disorder in a subject at risk for the disorder or suspected of having the disorder.
  • subject refers to a vertebrate, preferably a mammal, more preferably a human.
  • immunocompromised refers to a subject who has an immunodeficiency.
  • the subject is very vulnerable to opportunistic infections, infections caused by organisms that usually do not cause disease in a person with a healthy immune system, but can affect people with a poorly functioning or suppressed immune system.
  • the present invention generally provides cells, including genetically modified immunoresponsive cells (e.g., T cells, Natural Killer (NK) cells, cytotoxic T lymphocytes (CTL) ceils) expressing at least a combination of an antigen- recognizing receptor (e.g., TCR or CAR) and a chimeric co-stimulating receptor (CCR), and methods of use therefore for the treatment of neoplasia and other pathologies where an increase in an antigen-specific immune response is desired.
  • genetically modified immunoresponsive cells e.g., T cells, Natural Killer (NK) cells, cytotoxic T lymphocytes (CTL) ceils
  • an antigen- recognizing receptor e.g., TCR or CAR
  • CTL chimeric co-stimulating receptor
  • the invention is based, at least in part, on the discovery that the simultaneous engagement of two antigens co-expressed by a tumor cell by an antigen-recognizing receptor and chimeric co-stimulating receptor is useful for activating and stimulating an immunoreactive cell without systemic effects.
  • the reactivity against tissues expressing either antigen alone is preferably minimal, inducing T cell activaiion in the presence of both antigens but not either one alone.
  • T ceil activation is mediated by a TCR or a CAR targeted to an antigen (e.g., CD19 or prostate stem cell antigen, PSCA).
  • Costimulation is independently mediated by a "chimeric costimulatory receptor" (CCR), 2 ' 3 which is targeted to a second antigen (e.g., prostate-specific membrane antigen, PSMA).
  • CCR chimeric costimulatory receptor
  • PSMA prostate-specific membrane antigen
  • this approach is not limited to the treatment of neoplasias, but is amenable to a wide range of applications where an increase in an antigen-specific immune response is desired, such applications include not only the treatment of neoplasias, but also for the enhancement of an immune response against a pathogen infection or an infectious disease and to reinforce immune tolerance in regulatory T ceils in the context of autoimmunity or allogeneic transplantation.
  • lymphoid lineage comprising B, T and natural killer (NK) cells, provides for the production of antibodies, regulation of the cellular immune system, detection of foreign agents in the blood, detection of cells foreign to the host, and the iike.
  • T cells refers to lymphocytes that mature in the thymus and are chiefly responsible for cell- mediated immunity. T cells are involved in the adaptive immune system.
  • NK ceils refers to lymphocytes that are part of cell-mediated immunity and act during the innate immune response. They do not require prior activation in order to perform their cytotoxic effect on target ceils. Cytotoxic T cells (CTL or killer T ceils) are a subset of T TTL or killer T ceils.
  • lymphocytes capable of inducing the death of infected somatic or tumor cells.
  • the present invention provides cells expressing a combination of an antigen-recognizing receptor that activates an immunoresponsive ceil (e.g., TCR, CAR) and a chimeric co-stimulating receptor (CCR), and methods of using such cells for the treaiment of a disease that requires an enhanced immune response.
  • an immunoresponsive ceil e.g., TCR, CAR
  • CCR chimeric co-stimulating receptor
  • tumor antigen-specific T cells, NK cells, CTL ceils or other immunoresponsive cells are used as shuttles for the selective enrichment of one or more co-stimulatory ligands for the treaiment or prevention of neoplasia.
  • a T cell expressing a chimeric antigen receptor 19z1 that recognizes CD19 is co-expressed in a T cell that expresses a chimeric co- stimuiatory receptor P28BB that recognizes and binds Prostate Specific
  • PSMA Membrane Antigen
  • T cells NK cells
  • CTL cells can be used for the treatment of viral diseases.
  • a chimeric co-stimuiatory antigen receptor that recognizes a first CMV antigen and a chimeric antigen receptor that recognizes and binds a second CMV antigen are co-expressed in cytotoxic T lymphocytes for the treatment of CMV.
  • Types of tumor antigen-specific human lymphocytes that can be used in the methods of the invention include, without limitation, peripheral donor lymphocytes genetically modified to express chimeric antigen receptors (CARs) (Sadelain, M., et al. 2003 Nat Rev Cancer 3:35-45), peripheral donor
  • CARs chimeric antigen receptors
  • lymphocytes genetically modified to express a full-length tumor antigen- recognizing T cell receptor complex comprising the a and p heterodimer (Morgan, R.A., et al. 2006 Science 314:126-129), lymphocyte cultures derived from tumor infiltrating lymphocytes (TILs) in tumor biopsies (Panelii, M.C., et al. 2000 J Immunol 164:495-504; Panelii, M.C., et al.
  • TILs tumor infiltrating lymphocytes
  • T ceils may be autologous, allogeneic, or derived in vitro from engineered progenitor or stem cells.
  • any suitable tumor antigen is suitable for use in the tumor-related embodiments described herein.
  • Sources of antigen include, but are not limited to cancer proteins.
  • the antigen can be expressed as a peptide or as an intact protein or portion thereof.
  • the intact protein or a portion thereof can be native or mutagenized.
  • Suitable antigens include prostate specific membrane antigen (PSMA) and prostate stem cell antigen (PCSA).
  • Viral antigen-specific T lymphocytes and NK cells
  • Suitable antigens for use in the treatment of pathogen infection or other infectious disease, for example, in an immunocompromised subject include, without limitation, viral antigens present in Cytomegalovirus (CMV), Epstein Barr Virus (EBV), Human Immunodeficiency Virus (HIV), and influenza virus.
  • CMV Cytomegalovirus
  • EBV Epstein Barr Virus
  • HAV Human Immunodeficiency Virus
  • the unpurified source of CTLs may be any known in the art, such as the bone marrow, fetal, neonate or adult or other hematopoietic ceil source, e.g. , fetal liver, peripheral blood or umbilical cord blood.
  • hematopoietic ceil source e.g. , fetal liver, peripheral blood or umbilical cord blood.
  • Various techniques can be employed to separate the cells. For instance, negative selection methods can remove non-CTLs initially. mAbs are particularly useful for identifying markers associated with particular cell lineages and/or stages of differentiation for both positive and negative selections.
  • a large proportion of terminally differentiated cells can be initially removed by a relatively crude separation.
  • magnetic bead separations can be used initially to remove large numbers of irrelevant ceils.
  • at least about 80%, usually at least 70% of the total hematopoietic cells will be removed prior to cell isolation.
  • Procedures for separation include, but are not limited to, density gradient centrifugation; resetting; coupling to particles that modify cell density; magnetic separation with antibody-coated magnetic beads; affinity
  • cytotoxic agents joined to or used in conjunction with a mAb including, but not limited to, complement and cytotoxins; and panning with antibody attached to a solid matrix, e.g. plate, chip, elutriation or any other convenient technique.
  • Techniques for separation and analysis include, but are not limited to, flow cytometry, which can have varying degrees of sophistication, e.g., a plurality of color channels, low angle and obtuse light scattering detecting channels, impedance channels.
  • the cells can be selected against dead cells, by employing dyes associated with dead cells such as propidium iodide (PI).
  • PI propidium iodide
  • the ceils are coi!ected in a medium comprising 2% fetal calf serum (FCS) or 0.2% bovine serum albumin (BSA) or any other suitable, preferably sterile, isotonic medium.
  • FCS fetal calf serum
  • BSA bovine serum albumin
  • the invention generally provides an immunoresponsive celi, such as a virus specific or tumor specific T cell comprising a receptor that binds a first antigen and activates the immunresponsive celi and a receptor that binds a second antigen and stimulates the immunresponsive cell.
  • an immunoresponsive celi such as a virus specific or tumor specific T cell comprising a receptor that binds a first antigen and activates the immunresponsive celi and a receptor that binds a second antigen and stimulates the immunresponsive cell.
  • immunoresponsive ceils e.g., T cells, CTL cells, NK cells
  • Genetic modification of immunoresponsive ceils can be accomplished by transducing a substantially homogeneous cell composition with a recombinant DNA construct.
  • a retroviral vector (either gamma-retroviral or lentiviral) is employed for the introduction of the DNA construct into the ceil.
  • a polynucleotide encoding a receptor that binds an antigen can be cloned into a retroviral vector and expression can be driven from its endogenous promoter, from the retroviral long terminal repeat, or from a promoter specific for a target cell type of interest.
  • an antigen e.g., a tumor antigen, or a variant, or a fragment thereof
  • Non-viral vectors may be used as well.
  • a retroviral vector is generally employed for transduction, however any other suitable viral vector or non-viral delivery system can be used.
  • retroviral gene transfer for subsequent genetic modification of the cells to provide ceils comprising an antigen presenting complex comprising at least two co- stimulatory ligands, retroviral gene transfer (transduction) likewise proves effective.
  • Combinations of retroviral vector and an appropriate packaging line are also suitable, where the capsid proteins will be functional for infecting human cells.
  • Various amphotropic virus-producing cell lines are known, including, but not limited to, PA12 (Miller, et ai. (1985) Mol. Cell. Biol.
  • Non- amphotropic particles are suitable too, e.g., particles pseudotyped with VSVG, RD114 or GALV envelope and any other known in the art.
  • Possible methods of transduction also include direct co-culture of the cells with producer cells, e.g., by the method of Bregni, et al. (1992) Blood 80:1418- 1422, or culturing with viral supernatant alone or concentrated vector stocks with or without appropriate growth factors and poiycations, e.g., by the method of Xu, et al. (1994) Exp. Hemat. 22:223-230; and Hughes, et al. (1992) J. Clin. Invest. 89:1817.
  • transducing viral vectors can be used to express a co-stimulatory ligand of the invention in an immunoresponsive ceil.
  • the chosen vector exhibits high efficiency of infection and stable integration and expression (see, e.g., Cayouette et ai., Human Gene Therapy 8:423-430, 1997; Kido et al., Current Eye Research 15:833-844, 1996; Bloomer et al., Journal of Virology 71 :6641-6649, 1997; Naldini et al., Science 272:263-267, 1996; and Miyoshi et al., Proc. Natl. Acad. Sci. U.S.A. 94:10319, 1997).
  • viral vectors that can be used include, for example, adenoviral, lentiviral, and adeno-associated viral vectors, vaccinia virus, a bovine papilloma virus, or a herpes virus, such as Epstein-Barr Virus (also see, for example, the vectors of Miller, Human Gene Therapy 15-14, 1990; Friedman, Science 244:1275-1281 , 1989; Eglitis et al., BioTechniques 6:608-614, 1988;
  • Epstein-Barr Virus also see, for example, the vectors of Miller, Human Gene Therapy 15-14, 1990; Friedman, Science 244:1275-1281 , 1989; Eglitis et al., BioTechniques 6:608-614, 1988;
  • Retroviral vectors are particularly well developed and have been used in clinical settings (Rosenberg et al., N. Engl. J . Med 323:370, 1990; Anderson et al., U.S. Pat. No. 5,399,346).
  • Non-viral approaches can also be employed for the expression of a protein in ceil.
  • a nucleic acid molecule can be introduced into a cell by administering the nucleic acid in the presence of lipofection (Feigner et al., Proc. Natl. Acad. Sci. U.S.A. 84:741 3, 1987; Ono et a!., Neuroscience Letters 17:259, 1990; Brigham et al., Am. J. Med. Sci. 298:278, 1989;
  • RNA molecules can also be potentially beneficial for delivery of DNA into a ceil.
  • Transplantation of normal genes into the affected tissues of a subject can also be accomplished by transferring a normal nucleic acid into a cuitivatable ceil type ex vivo (e.g., an autologous or heterologous primary cell or progeny thereof), after which the cell (or its descendants) are injected into a targeted tissue or are injected systemically.
  • Recombinant receptors can also be derived or obtained using transposases or targeted nucleases (e.g. Zinc finger nucleases, meganucleases, or TALE nucleases).
  • Transient expression may be obtained by RNA
  • cDNA expression for use in polynucleotide therapy methods can be directed from any suitable promoter (e.g., the human cytomegalovirus (CMV), simian virus 40 (SV40), or metailothionein promoters), and regulated by any appropriate mammalian regulatory element or intron (e.g. the elongation factor 1c enhancer/promoter/intron structure).
  • CMV human cytomegalovirus
  • SV40 simian virus 40
  • metailothionein promoters regulated by any appropriate mammalian regulatory element or intron (e.g. the elongation factor 1c enhancer/promoter/intron structure).
  • enhancers known to preferentially direct gene expression in specific cell types can be used to direct the expression of a nucleic acid.
  • the enhancers used can include, without limitation, those that are characterized as tissue- or cell-specific enhancers.
  • regulation can be mediated by the cognate regulatory sequences or, if desired, by regulatory sequences derived from a heterologous source, including any of the promoters or regulatory elements described above.
  • the resulting cells can then be grown under conditions similar to those for unmodified cells, whereby the modified ceils can be expanded and used for a variety of purposes.
  • Also included in the invention are 19z1 , CD19, CD8, CD3 , dsRed, P28BB, PSMA, CD28, 4-1 BB, GFP polypeptides or fragments thereof that are modified in ways that enhance their anti-neopiastic activity when expressed in an immunoresponsive cell.
  • the invention provides methods for optimizing an amino acid sequence or nucleic acid sequence by producing an alteration in the sequence. Such alterations may include certain mutations, deletions, insertions, or post-translational modifications.
  • the invention further includes analogs of any naturally-occurring polypeptide of the invention. Analogs can differ from a naturally-occurring polypeptide of the invention by amino acid sequence differences, by post-translational modifications, or by both.
  • Analogs of the invention will generally exhibit at least 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identity with all or part of a naturally-occurring amino, acid sequence of the invention.
  • the length of sequence comparison is at least 5, 10, 15 or 20 amino acid residues, preferably at least 25, 50, or 75 amino acid residues, and more preferably more than 100 amino acid residues.
  • a BLAST program may be used, with a probability score between e 3 and e " 00 indicating a closely related sequence.
  • Modifications include in vivo and in vitro chemical derivatization of polypeptides, e.g., acetylation, carboxyiation, phosphorylation, or giycosylation; such modifications may occur during polypeptide synthesis or processing or following treatment with isolated modifying enzymes.
  • Analogs can also differ from the naturally-occurring polypeptides of the invention by alterations in primary sequence.
  • the invention also provides fragments of any one of the polypeptides or peptide domains of the invention.
  • a fragment means at least 5, 10, 13, or 15 amino acids.
  • a fragment is at least 20 contiguous amino acids, at least 30 contiguous amino acids, or at least 50 contiguous amino acids, and in other embodiments at least 60 to 80, 100, 200, 300 or more contiguous amino acids.
  • Fragments of the invention can be generated by methods known to those skilled in the art or may result from normal protein processing (e.g., removal of amino acids from the nascent polypeptide that are not required for biological activity or removal of amino acids by alternative mRNA splicing or alternative protein processing events).
  • Non-protein analogs have a chemical structure designed to mimic the functional activity of a protein of the invention. Such analogs are administered according to methods of the invention. Such analogs may exceed the
  • Co- stimulatory ligands include, without limitation, tumor necrosis factor (TNF) ligands, cytokines (such as IL-2, IL-12, !L-15 or IL21 ), and immunoglobulin (ig) superfamily ligands.
  • TNF tumor necrosis factor
  • cytokines such as IL-2, IL-12, !L-15 or IL21
  • immunoglobulin (ig) superfamily ligands include, without limitation, tumor necrosis factor (TNF) ligands, cytokines (such as IL-2, IL-12, !L-15 or IL21 ), and immunoglobulin (ig) superfamily ligands.
  • ig immunoglobulin
  • Tumor necrosis factor is a cytokine involved in systemic inflammation and stimulates the acute phase reaction, its primary role is in the regulation of immune cells.
  • Tumor necrosis factor (TNF) ligands share a number of common features. The majority of the ligands are synthesized as type II transmembrane proteins (extracelluiarC-terminus) containing a short cytoplasmic segment and a relatively long extracellular region.
  • TNF ligands include, without limitation, nerve growth factor (NGF), CD40L (CD40L)/CD154, CD137L/4- 1 BBL, tumor necrosis factor alpha (TNFa), CD134L/OX40L/CD252, CD27L/CD70, Fas ligand (FasL), CD30L/CD153, tumor necrosis factor beta (TNF(3)/iymphotoxin- aipha (LTa), lymphotoxin-beta (ur(3), CD257/B cell-activating factor
  • immunoglobulin (Ig) superfamily is a large group of cell surface and soluble proteins that are involved in the recognition, binding, or adhesion processes of cells. These proteins share structural features with immunoglobulins - they possess an immunoglobulin domain (fold). Immunoglobulin superfamily ligands include, without limitation, CD80 and CD86, both ligands for CD28.
  • compositions comprising genetically modified immunoresponsive ceils of the invention can be provided systemically or directly to a subject for the treatment of a neoplasia, pathogen infection, or infectious disease.
  • ceils of the invention are directly injected into an organ of interest (e.g., an organ affected by a neoplasia).
  • compositions comprising genetically modified immunoresponsive cells are provided indirectly to the organ of interest, for example, by administration into the circulatory system (e.g. , the tumor vasculature).
  • Expansion and differentiation agents can be provided prior to, during or after administration of the cells to increase production of T cells, NK cells, or CTL cells in vitro or in vivo.
  • the modified cells can be administered in any physiologically acceptable vehicle, normaily intravascuiarly, although they may also be introduced into bone or other convenient site where the cells may find an appropriate site for regeneration and differentiation (e.g., thymus). Usuaily, at least 1x10 5 cells will be administered, eventually reaching 1x10 10 , or more.
  • Genetically modified immunoresponsive cells of the invention can comprise a purified population of celis. Those skilled in the art can readily determine the percentage of genetically modified immunoresponsive cells in a population using various well-known methods, such as fluorescence activated cell sorting (FACS). Preferable ranges of purity in populations comprising genetically modified immunoresponsive ceils are about 50 to about 55%, about 55 to about 60%, and about 65 to about 70%.
  • the purity is about 70 to about 75%, about 75 to about 80%, about 80 to about 85%; and still more preferably the purity is about 85 to about 90%, about 90 to about 95%, and about 95 to about 100%. Dosages can be readily adjusted by those skilled in the art (e.g., a decrease in purity may require an increase in dosage).
  • the cells can be introduced by injection, catheter, or the like, if desired, factors can also be included, including, but not limited to, interleukins, e.g. IL-2, IL- 3, IL-6, and IL-1 1 , as well as the other interleukins, the colony stimulating factors, such as G-, M- and GM-CSF, interferons, e.g. .gamma. -interferon and erythropoietin.
  • interleukins e.g. IL-2, IL- 3, IL-6, and IL-1 1
  • the colony stimulating factors such as G-, M- and GM-C
  • compositions of the invention include pharmaceutical compositions comprising genetically modified immunoresponsive celis or their progenitors and a pharmaceutically acceptable carrier.
  • Administration can be autologous or heterologous.
  • immunoresponsive ceils, or progenitors can be obtained from one subject, and administered to the same subject or a different, compatible subject.
  • Peripheral blood derived immunoresponsive cells of the invention or their progeny e.g., in vivo, ex vivo or in vitro derived
  • a therapeutic composition of the present invention e.g., a pharmaceutical composition containing a genetically modified immunoresponsive cell
  • it will generally be formulated in a unit dosage injectable form (solution, suspension, emulsion).
  • compositions of the invention comprising genetically modified immunoresponsive cells can be conveniently provided as sterile liquid preparations, e.g., isotonic aqueous solutions, suspensions, emulsions, dispersions, or viscous compositions, which may be buffered to a selected pH.
  • sterile liquid preparations e.g., isotonic aqueous solutions, suspensions, emulsions, dispersions, or viscous compositions, which may be buffered to a selected pH.
  • Liquid preparations are normally easier to prepare than gels, other viscous compositions, and solid compositions. Additionally, liquid compositions are somewhat more convenient to administer, especially by injection. Viscous compositions, on the other hand, can be formulated within the appropriate viscosity range to provide longer contact periods with specific tissues.
  • Liquid or viscous compositions can comprise carriers, which can be a solvent or dispersing medium containing, for example, water, saline, phosphate buffered saline, polyoi (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like) and suitable mixtures thereof.
  • carriers can be a solvent or dispersing medium containing, for example, water, saline, phosphate buffered saline, polyoi (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like) and suitable mixtures thereof.
  • Sterile injectable solutions can be prepared by incorporating the genetically modified immunoresponsive cells utilized in practicing the present invention in the required amount of the appropriate solvent with various amounts of the other ingredients, as desired.
  • Such compositions may be in admixture with a suitable carrier, diluent, or excipient such as sterile water, physioiogical saline, glucose, dextrose, or the like.
  • a suitable carrier diluent, or excipient
  • the compositions can also be lyophiiized.
  • compositions can contain auxiliary substances such as wetting, dispersing, or emulsifying agents (e.g., methylcelluiose), pH buffering agents, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired.
  • auxiliary substances such as wetting, dispersing, or emulsifying agents (e.g., methylcelluiose), pH buffering agents, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired.
  • Standard texts such as "REMINGTON'S PHARMACEUTICAL SCIENCE", 17th edition, 1985, incorporated herein by reference, may be consulted to prepare suitable preparations, without undue experimentation.
  • compositions including antimicrobial preservatives, antioxidants, chelating agents, and buffers, can be added.
  • Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like.
  • Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin. According to the present invention, however, any vehicle, diluent, or additive used would have to be compatible with the genetically modified
  • compositions can be isotonic, i.e., they can have the same osmotic pressure as blood and lacrimal fluid.
  • the desired isotonicity of the compositions of this invention may be accomplished using sodium chloride, or other
  • Sodium chloride is preferred particularly for buffers containing sodium ions.
  • Viscosity of the compositions can be maintained at the selected level using a pharmaceutically acceptable thickening agent.
  • Methyiceiluiose is preferred because it is readily and economically available and is easy to work with.
  • suitable thickening agents include, for example, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, carbomer, and the like.
  • concentration of the thickener will depend upon the agent selected. The important point is to use an amount that will achieve the selected viscosity.
  • suitable carriers and other additives will depend on the exact route of administration and the nature of the particular dosage form, e.g., liquid dosage form (e.g., whether the composition is to be formulated into a solution, a suspension, gel or another liquid form, such as a time release form or liquid-filled form).
  • compositions should be selected to be chemically inert and will not affect the viability or efficacy of the genetically modified immunoresponsive cells as described in the present invention. This will present no problem to those skilled in chemical and pharmaceutical principies,or problems can be readily avoided by reference to standard texts or by simple experiments (not involving undue experimentation), from this disclosure and the documents cited herein.
  • One consideration concerning the therapeutic use of genetically modified immunoresponsive cells of the invention is the quantity of ceils necessary to achieve an optimal effect. The quantity of ceils to be administered will vary for the subject being treated.
  • between 10 4 to 10 10 , between 10 5 to 10 9 , or between 10 6 and 10 s genetically modified immunoresponsive cells of the invention are administered to a human subject. More effective ceils may be administered in even smaller numbers. In some embodiments, at least about 1 x 10 8 , 2 x 10 8 , 3 x 10 8 , 4 x 10 8 , and 5 x 10 8 genetically modified immunoresponsive cells of the invention are administered to a human subject. The precise determination of what would be considered an effective dose may be based on factors individual to each subject, including their size, age, sex, weight, and condition of the particular subject. Dosages can be readily ascertained by those skilled in the art from this disclosure and the knowledge in the art.
  • any additives in addition to the active ceil(s) and/or agent(s) are present in an amount of 0.001 to 50 % (weight) solution in phosphate buffered saline, and the active ingredient is present in the order of micrograms to milligrams, such as about 0.0001 to about 5 wt %, preferably about 0.0001 to about 1 wt %, still more preferably about 0.0001 to about 0.05 wt % or about 0.001 to about 20 wt %, preferably about 0.01 to about 10 wt %, and still more preferably about 0.05 to about 5 wt %.
  • any composition to be administered to an animal or human, and for any particular method of
  • toxicity such as by determining the lethal dose (LD) and LD50 in a suitable animal model e.g., rodent such as mouse; and, the dosage of the composition(s), concentration of components therein and timing of administering the composition(s), which elicit a suitable response.
  • kits for treating neoplasia in a subject are also contemplated herein.
  • methods for treating a pathogen infection or other infectious disease in a subject comprise administering a T cell, NK cell, or CTL cell of the invention in an amount effective to achieve the desired effect, be it palliation of an existing condition or prevention of recurrence.
  • the amount administered is an amount effective in producing the desired effect.
  • An effective amount can be provided in one or a series of administrations.
  • An effective amount can be provided in a bolus or by continuous perfusion.
  • an “effective amount” is an amount sufficient to effect a beneficial or desired clinicai result upon treatment.
  • An effective amount can be administered to a subject in one or more doses.
  • an effective amount is an amount that is sufficient to palliate, ameliorate, stabilize, reverse or slow the progression of the disease, or otherwise reduce the pathological consequences of the disease.
  • the effective amount is generally determined by the physician on a case-by-case basis and is within the skill of one in the art. Several factors are typically taken into account when
  • determining an appropriate dosage to achieve an effective amount include age, sex and weight of the subject, the condition being treated, the severity of the condition and the form and effective concentration of the antigen-binding fragment administered.
  • ceil doses in the range of 10 6 — 10 10 are typically infused.
  • T cells are induced that are specifically directed against the specific antigen.
  • "Induction" of T cells can include inactivation of antigen-specific T ceils such as by deletion or anergy. inactivation is particularly useful to establish or reestablish tolerance such as in autoimmune disorders.
  • the modified cells can be administered by any method known in the art including, but not limited to, intravenous, subcutaneous, intranodal, intratumorai, intrathecal, intrapleural, intraperitoneal and directly to the thymus.
  • the invention provides methods for increasing an immune response in a subject in need thereof.
  • the invention provides methods for treating or preventing a neoplasia in a subject.
  • the invention provides therapies that are particularly useful for the treatment of subjects having prostate cancer, or metastatic prostate cancer that is not amenable to conventional therapeutic interventions.
  • Suitabiehuman subjects for therapy typically comprise two treatment groups that can be distinguished by clinical criteria. Subjects with "advanced disease” or "high tumor burden” are those who bear a clinically measurable tumor.
  • a clinically measurable tumor is one that can be detected on the basis of tumor mass (e.g., by palpation, CAT scan, sonogram, mammogram or X-ray; positive biochemical or histopathologic markers on their own are insufficient to identify this population).
  • composition embodied in this invention is administered to these subjects to elicit an anti-tumor response, with the objective of palliating their condition.
  • reduction in tumor mass occurs as a result, but any clinical improvement constitutes a benefit.
  • Clinical improvement includes decreased risk or rate of progression or reduction in pathological
  • adjuvant group are individuals who have had a history of neoplasia, but have been responsive to another mode of therapy.
  • the prior therapy can have included, but is not restricted to, surgical resection, radiotherapy, and traditional chemotherapy. As a result, these individuals have no clinically measurable tumor. However, they are suspected of being at risk for
  • This group can be further subdivided into high-risk and iow-risk individuals. The subdivision is made on the basis of features observed before or after the initial treatment. These features are known in the clinical arts, and are suitably defined for each different neoplasia. Features typical of high-risk subgroups are those in which the tumor has invaded neighboring tissues, or who show involvement of lymph nodes.
  • Another group have a genetic predisposition to neoplasia but have not yet evidenced clinical signs of neoplasia. For instance, women testing positive for a genetic mutation associated with breast cancer, but still of childbearing age, can wish to receive one or more of the antigen-binding fragments described herein in treatment prophylacticaliy to prevent the occurrence of neoplasia until it is suitable to perform preventive surgery.
  • glioblastoma melanoma
  • neuroblastoma adenocarcinoma
  • glioma soft tissue sarcoma
  • various carcinomas including prostate and small cell lung cancer
  • Suitable carcinomas further include any known in the field of oncology, including, but not limited to, astrocytoma,
  • fibrosarcoma myxosarcoma, liposarcoma, oligodendroglioma, ependymoma, medullobiastoma, primitive neural ectodermal tumor (PNET), chondrosarcoma, osteogenic sarcoma, pancreatic ductal adenocarcinoma, small and large cell lung adenocarcinomas, chordoma, angiosarcoma, endotheliosarcoma, squamous cell carcinoma, bronchoalveolarcarcinoma, epithelial adenocarcinoma, and liver metastases thereof, lymphangiosarcoma, iymphangioendotheliosarcoma, hepatoma, cholangiocarcinoma, synovioma, mesothelioma, Ewing's tumor, rhabdomyosarcoma, colon carcinoma, basal cell carcinoma, sweat gland carcinoma, papillar
  • craniopharyngioma ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, neuroblastoma, retinoblastoma, leukemia, multiple myeloma, Waldenstrom's macroglobuiinemia, and heavy chain disease
  • breast tumors such as ductal and lobular adenocarcinoma, squamous and adenocarcinomas of the uterine cervix, uterine and ovarian epithelial carcinomas, prostatic adenocarcinomas, transitional squamous cell carcinoma of the bladder, B and T cell lymphomas (nodular and diffuse) plasmacytoma, acute and chronic leukemias, malignant melanoma, soft tissue sarcomas and leiomyosarcomas.
  • the subjects can have an advanced form of disease, in which case the treatment objective can include mitigation or reversal of disease progression, and/or amelioration of side effects.
  • the subjects can have a history of the condition, for which they have already been treated, in which case the therapeutic objective will typically include a decrease or delay in the risk of recurrence.
  • the invention provides a method of treating or preventing a neoplasia in a subject, the method comprising administering an effective amount of an immunoresponsive cell comprising a receptor that binds a tumor antigen and activates the immunoresponsive ceil (e.g., TCR, CAR) and a vector encoding a receptor that binds another tumor antigen and stimulates the immunoresponsive cell, in one embodiment, the neoplasia is selected from the group consisting of prostate cancer, breast cancer, blood cancers (e.g.
  • leukemias lymphomas, and myelomas
  • ovarian cancer bladder cancer, brain cancer, colon cancer, intestinal cancer, liver cancer, lung cancer, pancreatic cancer, prostate cancer, skin cancer, stomach cancer, glioblastoma, and throat cancer.
  • the tumor antigen is one or more of carbonic anhydrase IX (CAIX), carcinoembryonic antigen (CEA), CD5, CD7, CD10, CD19, CD20, CD22, CD30, CD33, CD34, CD38, CD41 , CD44, CD49f, CD56, CD74, CD133, CD138, an antigen of a cytomegalovirus (CMV) infected cell (e.g., a cell surface antigen), epithelial giycoprotein-2 (EGP-2), epithelial glycoprotein-40 (EGP-40), epithelial cell adhesion molecule (EpCAM), receptor tyrosine-protein kinases erb-B2,3,4, foiate-binding protein (FBP), fetal acetylcholine receptor (AChR), folate receptor-a, Gangiioside G2 (GD2), Gangiioside G3 (GD3), human Epidermal Growth Factor Receptor 2 (HER-2
  • adoptively transferred human T or NK cells are endowed with augmented and selective cytolytic activity at the tumor site.
  • a receptor that binds a tumor antigen and activates the immunoresponsive ceil e.g., TCR, CAR
  • a vector encoding a receptor that binds another tumor antigen and stimulates the immunoresponsive cell e.g. CCR
  • adoptively transferred human T or NK cells are endowed with augmented and selective cytolytic activity at the tumor site.
  • co-stimulatory ligandexpressing T ceils turn the tumor or viral infection site into a highly conductive environment for a wide range of immune ceils involved in the physiological anti-tumor or antiviral response (tumor infiltrating lymphocytes, NK-, NKT- cells, dendritic cells, and macrophages).
  • the invention provides methods for treating subjects with a pathogen infection (e.g., viral infection, bacterial infection, fungal infection, parasite infection, or protozoa! infection).
  • a pathogen infection e.g., viral infection, bacterial infection, fungal infection, parasite infection, or protozoa! infection.
  • the invention is particularly useful for enhancing an immune response in an
  • Exemplary viral infections susceptible to treatment using a method of the invention include, but are not limited to, Cytomegalovirus (CMV), Epstein Barr Virus (EBV), Human Immunodeficiency Virus (HIV), and influenza virus infections.
  • CMV Cytomegalovirus
  • EBV Epstein Barr Virus
  • HAV Human Immunodeficiency Virus
  • influenza virus infections include, but are not limited to, influenza virus infections.
  • the invention provides a method of treating or preventing a pathogen infection in a subject, the method comprising administering an effective amount of an immunoresponsive cell as described herein.
  • kits for the treatment or prevention of a neoplasia, pathogen infection, immune disorder or allogeneic transplant includes a therapeutic or prophylactic composition containing an effective amount of an immunoresponsive cell comprising an activating antigen receptor and a co-stimulatory antigen receptor in unit dosage form.
  • the kit further comprise a co-stimulatory ligand.
  • the kit comprises a sterile container which contains a
  • Such containers can be boxes, ampules, bottles, vials, tubes, bags, pouches, blister-packs, or other suitable container forms known in the art.
  • Such containers can be made of plastic, glass, laminated paper, metal foil, or other materials suitable for holding
  • the immunoresponsive cell is provided together with instructions for administering the ceil to a subject having or at risk of developing a neoplasia, pathogen infection, immune disorder or allogeneic transplant.
  • the instructions will generally include information about the use of the composition for the treatment or prevention of neoplasia, pathogen infection, immune disorder or allogeneic transplant.
  • the instructions include at least one of the foliowing: description of the therapeutic agent; dosage schedule and administration for treatment or prevention of a neoplasia, pathogen infection, immune disorder or allogeneic transplant or symptoms thereof; precautions; warnings; indications; counter-indications; overdosage information; adverse reactions; animal pharmacology; clinical studies; and/or references.
  • the instructions may be printed directly on the container (when present), or as a label applied to the container, or as a separate sheet, pamphlet, card, or folder supplied in or with the container.
  • Example 1 T cells co-expressing a chimeric antigen receptor (CAR) and a chimeric co-stimulating receptor (CCR) eradicated established tumors.
  • CAR chimeric antigen receptor
  • CCR chimeric co-stimulating receptor
  • the invention provides "tumor-sensing T ceils" that simultaneously engage two antigens co-expressed by a tumor ceil. Importantly, it has been found that the reactivity against tissues expressing either antigen alone should be negligible, only unleashing T cell activation in the presence of both antigens but not either one alone.
  • the invention is at least based in part on the discoveries that in combination provide selective T ceil immunoreactivity, and, thus, make this approach clinically relevant.
  • the first is to assign T cell activation to one antigen (e.g., CD19 or prostate stem cell antigen, PSCA), which may be mediated by a T cell receptor (TCR) or a chimeric antigen receptor (CAR). Costimulation is independently mediated by a "chimeric costimuiatory receptor" (CCR ), 2,13 which is targeted to a second antigen (e.g., prostate-specific membrane antigen, PSMA).
  • TCR T cell receptor
  • CAR chimeric antigen receptor
  • mice were systemicaily engrafted with 2.0 x 10 6 firefly-!uciferase expressing PC3 tumor cells that expressed both CD19 and PSMA ( Figure 5) and treated 19 days later with a single intravenous infusion of 1 .0 x 10 6 19z1 , 19z1 +P28BB, P28BB or control T cells. Thirty-five days later, mice that received P28BB T cells or control T cells were sacrificed due to tumor burden.
  • mice treated with 19z1 T cells had a marked reduction of tumor burden.
  • mice treated with 19z1 +P28BB T cells had undetectable tumor burden (Figure 2C).
  • the CD19 + tumors eventually relapsed in mice that received 19z1 T cells, while complete remission persisted in ail mice that received 19z1 +P28BB T cells ( Figure 2C). This result strongly indicated that tumor eradication had been achieved.
  • mice were administered one of 19z1 , P28BB, or 19z1 +P28BB T cells (1 .0 x 10 6 cells) intravenously. Mice that received P28BB T cells had progression of all three tumors and needed to be sacrificed within 35 days ( Figure 2D).
  • the CD19 + PSMA ⁇ and CD19 + PSMA + tumors underwent a substantial reduction compared to their progression in recipients of P28BB T cells, before eventually progressing. Consistent with prior results, mice treated with 19z1 +P28BB T cells showed complete eradication of CD19 + PSMA + tumors.
  • mice were inoculated intravenously with 2 x 106 FFLuc-expressing PC3 cells positive for PSMA, PSCA, or both (Figure 5).
  • one set of mice received 1 x 106 Mzl+P28BB CAR+ T cells infused intravenously, and another set of received 1 x 106 Lz1 +P28BB CARP T cells.
  • Mice bearing PSCA+PSMA tumor cells that were treated with the more efficient Mzl+P28BB T cells exhibited greater tumor regression than mice treated with Lz1 +PBB T cells (Figure 3C). Similar to the CD19 experiment ( Figure 2C), these tumors eventually relapsed and progressed.
  • mice bearing PSCA+PSMA+ tumor celis Mzl+P28BB T ceils induced robust and long-term tumor eradication. Consistent with the lesser potency of Lzl+P28BB T ceils, tumor eradication in mice bearing PSCA+PSMA+ tumor cells treated with Lz1 +P28BB T cells was slower but nonetheless equally successful, resulting in strong tumor eradication and long-term survival of ail treated mice ( Figure 3C). Tumor eradication was not enhanced in control mice bearing either PSCAVSMA- or PSCA-PSMA+ tumors ( Figure 3C). A more stringent evaluation of background activity against PSCAVSMA.- tumors was tested in the context of animals also bearing PSCA+PSMA+ and PSCAPSMA+ tumors. Lz1 +P28BB T cells mediated eradication of PSCA+PSMA+ tumors without increasing eradication of PSCA+PSMA tumors ( Figure 3E), which was not different from that induced by Lzl T celis.
  • Targeted T ceil therapies have the potential to provide curative treatments but their applicability is limited by the paucity of validated tumor-specific targets. Extra- tumoral expression results indeed in "on-target, off-tumor” effects that 2-4" may be sometimes tolerable but are eventually lethal 11.
  • the method described herein provides improved targeting by supplying titrated activation and
  • T ceils are primed in lymph nodes by receiving activating and costimuiatory signals and migrate to peripheral sites, where effector functions of the T cells are not as dependent on costimuiation.
  • T cells engaged through an antigen receptor and a CCR may recirculate to other peripheral sites and display heightened cytolytic activity against tissues expressing only one of the targeted antigens (Figure 6A). Therefore, the present strategy was developed to address this problem of a potential systemic effect in order to spare cells singly positive for the antigen, including non-tumor cells (Figure 6B).
  • PSCA and PSMA are promising targets for the treatment of metastatic prostate cancer 26 ' 27 , although neither is absolutely prostate-specific.
  • PSCA expression is found in prostate cancer and within the renal pelvis, ureter, urinary bladder, and urethra.
  • Expression of PSMA strongly correlates with primary prostate cancer, metastases, as well as in astrocytes type II, the kidney proximal tubule and the intestinal brush border.
  • Dual PSCA/PSMA targeting is thus expected to increase prostate cancer targeting and reduce reactivity against these normal tissues. It is appreciated that this principle can be extended to other tumor types which express a pair of antigens, especially those that confer true tumor-specificity.
  • HER2, MUC1 , CD44, CD49f, and/or EpCAM could be used in this manner to treat breast cancer.
  • mesothelin, folate receptor-a, CD44, and/or CD133 could be used to treat ovarian cancer.
  • 32 ' 33 The targeting of tumor initiating cells or cancer stem cells, for which unique target antigens/structures have not yet been clearly identified34.3s would be particularly attractive using this approach.
  • An important aspect of this approach is to constrain and nearly abolish T cell activation in response to a single antigen. CARs with low affinity or low avidity that only provide a poor activation signal were found to be usefui for achieving this effect.
  • an endogenous TCR with low affinity or low avidity may be used in combination with a CCR to provide antigen-specific costimulation.
  • This backbone construct was used to exchange scFvs to generate SFG-Hzl, SFG-Mzl, and SFG-Lzl by directional cloning utilizing a Ncol site located 5' of the scFv and a Nod site located 3' of the scFv.
  • SFG- P28BB the fused CD28 and CD137 domains were PCR amplified from SFG- P28BBz1 and iigated 3' of the PSMA scFv using a 5' Ncol site and a 3' BamHI site to include a stop codon 3' of the BB domain, while the CD3 domain was removed.
  • VH variabie heavy
  • VL variabie light
  • Hzl, Mzl, and Lzl were generated byampiifying the variabie heavy (VH) and variabie light (VL) domains conferring PSCA antigen specificity of non-overlapping epitopes using degenerate primers from hybridomas as previously described. 36 These VH and V
  • Peripherai blood leukocytes were isolated using Ficoli gradients and transduced as previously described. Briefly, after 48-hour activation with 2 &g/ml_ phytohemagglutinin, cells were transduced twice via spinocuiation for 1 hour on retronectin coated plates over the next 48 hours and 20 U/mL of lL-2 was added. After allowing 3 days for vector expression, transduction efficiencies were determined via flow cytometry and bulk unsorted ceils were used for various assays or adoptive transfers.
  • PC3 human prostate tumor line was obtained from ATCC and retrovirally 16 transduced in order to generate PC3 -GFP/Luc, which was subsequently used to create 10 PC3-CD19, PC3-PSMA, PC3-CD19-PSMA, PC3- PSCA, and PC3-PSCA-PSMA via retroviral transduction.
  • Target cells expressing desired antigen were labeled with 5 Cr and co- cultured with T cells at decreasing effector : target ratio's. After 4 hours of culture, supernatant was removed and used to measure radioactivity released from chromium. Specific lysis was determined by subtracting background radioactivity of target celis not cultured with T cells and dividing by the radioactivity measured from target cells completely Iysed by using 0.2% Triton X-100.
  • Tumor celis expressing desired antigen were irradiated with 30 Gy prior to co-culture with 1.0 x 10 6 T cells at a 5:1 effector : target ratio. T cells were counted weekly using an Invitrogen Countess cell counter and then re-stimuiated with irradiated tumor celis. No exogenous cytokines were added to these co- cultures.
  • PC3 tumor cells were infused into NOD/SCID-IL2Ry mice obtained from either Jackson Laboratories or from in-house breeding under the protocol 04-10- 024 approved by the MSKCC Institutional Animal Care and Use Committee.
  • 1.0 x 10 6 tumor cells were injected per tumor site, established for 7 days upon which 1.0 x 10 6 chimeric positive T ceils were infused IV.
  • tumor burden quantitativeiy measure tumor burden by correlating the amount of tumor burden to luminescence using an S 100 system (Caliper Life Sciences) as previously described.
  • S 100 system Caliper Life Sciences
  • Tumor volume was calculated by multiplying the length, width, and height of each tumor.
  • Bispecific antibodies containing a PSCA specific scFv fused to a CD3 specific scFv were added at various amounts to untransduced T cells co-cultured with PSCA + PC3 at a 20:1 ratio, respectively in standard 4hr chromium release assay assays.
  • Cells were analyzed using an LSRII flow cytometer or sorted using a FACSAria cell sorter (BD Biosciences) as previously described. 16 Detection of chimeric receptor at the cell surface could be achieved directly by using AF647 conjugated goat-anti-mouse antibody (invitrogen). Antibodies for CD4-PE-Cy7, CD8-Pacific Blue, and CD19-APC were obtained from Invitrogen while PSCA antibodies were purified from hybridoma supernatants and PSMA antibodies were obtained from MBL Interantional.
  • Ciinicai cancer research an official journal of the American Association for Cancer Research 16, 3533-3538 (2010).
  • Ciinicai cancer research an official journal of the American Association for Cancer Research 11 , 2591-2596 (2005).
  • Ciinicai cancer research :

Abstract

The present invention provides immunoresponsive cells, including T cells, cytotoxic T cells, regulatory T cells, and Natural Killer (NK) cells, expressing at least one of an antigen recognizing receptor and one of a chimeric costimulatory receptor. Methods of using the immunoresponsive cell include those for the treatment of neoplasia and other pathologies where an increase in an antigen-specific immune response is desired.

Description

COMPOSITIONS AND METHODS FOR IMMUNOTHERAPY Cross-Reference to Related Applications
[0001] This application is a non-provisional application claiming priority to U.S.
provisional application serial no. 61/709,072 filed October 2, 2012, the contents of which are hereby incorporated by reference into the present application.
Sequence Listing
[0002] This application contains a Sequence Listing, created on September 30, 2013; the fiie, in ASCII format, is designated 3314040AWO_Sequence Listing_ST25.txt and is 27.5 kilobytes in size. The sequence listing file is hereby incorporated by reference in its entirety into the application.
Background of the Invention
[ 0003 ] Prostate cancer is the most frequent cancer in males in the United
States and the cause of nearly 31 ,000 deaths per year. When diagnosed early, cancer can be effectively treated by surgery or radiation. Postsurgical residual disease requires radiation and/or hormonal therapy, which may prevent tumor progression and metastasis. At present, there is no curative treatment for hormone refractory, metastatic prostate cancer. Immunotherapy is a targeted therapy that in principle provides for the treatment of such cancers .
[ 0004 ] Targeted T ceil therapies utilizing genetically modified autologous T cells are beginning to show evidence of therapeutic efficacy in melanoma and indolent B cell malignancies. Current T ceil engineering strategies retarget patient T cells to tumor antigens through a transduced T cell receptor (TCR) or a chimeric antigen receptor (CAR). The newfound ability to induce potent immune responses, however, commands the need to confine immune attacks to the tumor and avoid reactions against normal tissues that may express the targeted antigen. Alas, the limited availability of truly tumor-restricted antigens often precludes
achieving highly specific targeting. Among the limitations precluding achieving highly specific targeting is the limited availability of truly tumor-restricted antigens. Accordingly, new methods of treating neoplasia are urgently required.
Summary of the Invention
[ 0005 ] The present invention generally provides immunoresponsive cells, including T ceils and Natural Killer (NK) cells, expressing an antigen binding receptor (e.g., CAR or TCR) having immune cell activating actvity and a chimeric co-stimulating receptor (CCR), and methods of use therefore for the treatment of neoplasia, infectious disease, and other pathologies.
[ 0006] In one aspect, the invention provides an isolated immunoresponsive cell having an antigen recognizing receptor that binds a first antigen with low affinity, where the binding activates the immunoresponsive ceil, and a chimeric co stimulating receptor (CCR) that binds a second antigen and stimulates the immunoresponsive ceil.
[0007] In another aspect, the invention provides a method of inducing tumor cell death in a subject, the method comprising administering an effective amount of an immunoresponsive cell comprising an antigen recognizing receptor that binds a first antigen with low affinity, where the binding activates the immunoresponsive cell, and a chimeric co-stimulating receptor (CCR) that binds a second antigen and stimulates the immunoresponsive cell, thereby inducing tumor cell death in the subject.
[0008] In still another aspect, the invention provides a method of treating or preventing a neoplasia in a subject, the method comprising administering an effective amount of an immunoresponsive cell comprising an antigen recognizing receptor that binds a first antigen with low affinity, where the binding activates the immunoresponsive ceil, and a chimeric co-stimulating receptor (CCR) that binds a second antigen and stimulates the immunoresponsive cell, thereby treating or preventing a neoplasia in the subject.
[0009] In yet another aspect, the invention provides a method of treating prostate cancer in a subject in need thereof, the method comprising
administering to the subject a therapeutically effective amount of a T cell comprising an antigen recognizing receptor that binds PSCA or CD19 with low affinity, where the binding activates the immunoresponsive cell, and a chimeric co-stimulating receptor (CCR) that binds PSMA and stimulates the
immunoresponsive cell, thereby treating prostate cancer in the subject.
[0010] In still another, the invention provides a method for producing an antigen- specific immunoresponsive ceil, the method involving introducing into the immunoresponsive cell a nucleic acid sequence that encodes a chimeric co- stimulating receptor (CCR), where the chimeric co-stimulating receptor has an antigen-binding domain coupled to an intracellular signaling domain that stimulates an immunoresponsive cell, where the immunoresponsive cell has an antigen recognizing receptor that binds a first antigen with low affinity, wherein the binding activates the immunoresponsive cell.
[0011] In a related aspect, the invention provides a pharmaceutical composition comprising an effective amount of an immunoresponsive ceil of the invention (e.g., a tumor antigen-specific T cell in a pharmaceutical composition for the treatment of neoplasia) in a pharmaceutically acceptable excipient.
[0012] In an additional aspect, the invention provides a kit for treatment of a neoplasia, pathogen infection, an autoimmune disorder, or an allogeneic transplant, the kit containing an immunoresponsive cell having an antigen recognizing receptor that binds a first antigen and activates the
immunoresponsive cell, and a chimeric co-stimuiating receptor (CCR) that binds a second viral antigen and stimulates the immunoresponsive cell. The kit may further comprise written instructions for using the immunoresponsive cell for the treatment of a subject having a neoplasia, a pathogen infection, an autoimmune disorder, or an allogeneic transplant.
[0013] In various embodiments of any of the aspects delineated herein, the immunoresponsive cell is selected as having an antigen recognizing receptor with low affinity. This may involve selecting the immunoresponsive cell as having an antigen recognizing receptor that binds a first antigen with low affinity, in various embodiments of any of the aspects delineated herein, the antigen recognizing receptor is selected as having low affinity for expression in the cell. This may involve introducing a second nucleic acid sequence that encodes a chimeric antigen receptor, where the chimeric antigen receptor comprises a second antigen-binding domain coupled to a second intracellular signaling domain that activates an immunoresponsive cell. In various embodiments of any of the aspects delineated herein, the antigen recognizing receptor is a T cell receptor (TCR) or chimeric antigen receptor (CAR). In various embodiments, the intracellular signaling domain of said antigen recognizing receptor is the CD3-chain signaling domain. In various embodiments, the intracellular signaling domain of the chimeric co-stimulating receptor (CCR) is a CD97, CD1 1a-CD18, CD2, ICOS, CD27, CD154, CD5, OX40, 4-1 BB or CD28 signaling domain.
[0014] In various embodiments of any of the aspects delineated herein, the antigen recognizing receptor is exogenous or endogenous. In various
embodiments of any of the aspects delineated herein, the antigen recognizing receptor is recombinantly expressed. In various embodiments, the antigen recognizing receptor is expressed from a vector. In various embodiments, the chimeric co-stimulating receptor (CCR) is expressed from a vector. In particular embodiments, the immunoresponsive cell expresses a recombinant or an
endogenous antigen receptor that is 19z1 or Pz1.
[0015] In various embodiments of any of the aspects delineated herein, the immunoresponsive cell is a T cell, a Natural Killer (NK) cell, a cytotoxic T
lymphocyte (CTL), a regulatory T cell, a human embryonic stem ceil, or a
piuripotent stem ceil from which lymphoid cells may be differentiated. In various embodiments of any of the aspects delineated herein, the immunoresponsive cell of any one of claims 1-9, where said immunoresponsive cell is autologous.
[0016] In various embodiments of any of the aspects delineated herein, the antigen is a tumor or pathogen antigen. In various embodiments of any of the aspects delineated herein, one or more antigen-binding domains are tumor antigen-binding domains. In various embodiments of any of the aspects delineated herein, the antigens or tumor antigens are selected from CAIX, CEA, CD5, CD7, CD10, CD19, CD20, CD22, CD30, CD33, CD34, CD38, CD41 , CD44, CD49f, CD56, CD74, CD133, CD138, a cytomegalovirus (CMV) infected ceil antigen, EGP-2, EGP-40, EpCAM, erb-B2,3,4, FBP, Fetal acetylcholine receptor, folate receptor-a, GD2, GD3, HER-2, hTERT, IL13R-a2, x-iight chain, KDR, LeY, Li cell adhesion molecule, MAGE-AI , MUC1 , Mesothelin, NKG2D iigands, NY- ES0-1 , oncofetal antigen (h5T4), PSCA, PSMA, ROR1 , TAG-72, VEGF-R2, and WT-1. In various embodiments, the first and second antigens are selected from CD133, a cytomegalovirus (CMV) infected cell antigen, erbB2, KDR Mesothelin, NKG2D Iigands, NY-ES0-1 , oncofetal antigen (h5T4), PSCA, PSMA, CD19, VEGF-R2, and WT-1. In particular embodiments, the first and second antigens are selected from HER2, MUC1 , CD44, CD49f, EpCAM, CEA, CD133, a
cytomegalovirus (CMV) infected cell antigen, EGP-2, EGP-40, EpCAM, erb-B2,3,4, FBP, KDR, Mesothelin, NKG2D Iigands, NY-ES0-1 , oncofetal antigen (h5T4), PSCA, PSMA, VEGF-R2, or WT-1. In specific embodiments, the first and second antigens are selected from CD10 and CD19. In other embodiments, the first and second antigens are selected from CD56 and CD138. In certain embodiments, the first and second antigens are selected from mesothelin, folate receptor-a, CD44, and CD133.
[0017] In various embodiments of any of the aspects delineated herein, the neoplasia is selected from the group consisting of prostate cancer, breast cancer, B cell leukemia, multiple myeloma, and ovarian cancer. In various embodiments of any of the aspects delineated herein, the method reduces the number of tumor cells, reduces tumor size, and/or eradicates the tumor in the subject.
[0018] In various embodiments, the neoplasia is prostate cancer and the first and second tumor antigens are distinct antigens selected from PSCA, PSMA, CD19, CD133, a cytomegalovirus (CMV) infected cell antigen, erb-B2, KDR
Mesothelin, NKG2D Iigands, NY-ES0-1 , oncofetal antigen (h5T4), VEGF-R2, and WT-I. In various embodiments, the neoplasia is breast cancer and the first and second tumor antigens are distinct antigens selected from HER2, MUC1 , CD44, CD49f, EpCAM, CEA, CD133, a cytomegalovirus (CMV) infected cell antigen, EGP-2, EGP-40, EpCAM, erb-B2,3,4, FBP, KDR, Mesothelin, NKG2D iigands, NY- ESO-1 , oncofetal antigen (h5T4), PSCA, PSMA, VEGF-R2, or WT-1 . in particular embodiments, the neoplasia is B cell leukemia and the first and second tumor antigens are selected from CDIO and CD19. in certain embodiments, the neoplasia is multiple myeloma and the first and second tumor antigens are selected from CD56 and CD138. In various embodiments, the neoplasia is ovarian cancer and the first and second tumor antigens are distinct antigens selected from mesothe!in, folate receptor-a, CD44, and CD133.
[0019] The invention provides compositions and methods that provide for T ceil targeting of tumor cells. Compositions and articles defined by the invention were isolated or otherwise manufactured in connection with the examples provided below. Other features and advantages of the invention will be apparent from the detailed description, and from the claims.
Brief Description of the Drawings
[0020] Figures 1A-C are graphics depicting chimeric antigen receptor (CAR) and chimeric costimulatory receptor (CCR) vector design and expression via transduction of primary human T cells. (A) depicts generation of CARs by fusing heavy and light chains of immunoglobulin variable domains to the CD8
transmembrane domain, which is fused to the cytosoiic signaling domains of CD3 . By using an internal Ribosomal Entry Site (IRES) to enable bicistronic expression, CAR expression can be easily detected by correlation to dsRED fluorescence (data not shown). The CCR was generated by fusing an scFv to a CD28 transmembrane and signaling domain15, fused to a 4-1 BB (aka CD137) cytosoiic signaling domain.21 CCR expression can be correlated to the bicistronic expression of hrGFP (data not shown). Abbreviations: LTR— Long Terminal Repeat; SD— Splice Donor site; SA— Splice Acceptor site; VH or VL -Variable Heavy or Light domains, respectively; EC— Extracellular domain; TM— Transmembrane domain; C— Cytosoiic domain; IRES— Internal Ribosomal Entry Site; dsRED - Discosoma sp. Red fluorescent protein, hrGFP— Human
Recombinant Green Fluorescent Protein (B) depicts representative transduction efficiencies of primary human T cells using these retroviral vectors. (C) depicts transduction of CTLs with different and multiple CARs for the present studies.
[0021] Figures 2A-D show that dual-receptor, CAR/CCR-mediated activation of human T cells allowed for robust CTL function, long-term proliferation, and enhanced tumor eradication upon binding of two antigens. (A) shows that T cells expressing chimeric receptors lysed cells positive for antigen when the
CAR specific to CD19 is expressed by T cells in CTL assays, compared to untransduced or P28BB transduced T cells. Plots are representative of n > 4 experiments, with error bars representing standard deviation of the mean of 3 replicates. (B) shows long-term proliferation of T cells by absolute T cell counts over 31 days of T cells expressing none, one, or both chimeric receptors that were co-cultured with human tumor cell lines expressing both or either antigen alone. Arrows indicate re-stimulation of T cells using freshly irradiated tumor cells. Only when dual-receptor expressing T ceils encounter both antigens is robust long-term proliferation observed. Plots are representative of n > 4 experiments with error bars representing standard deviation of the mean of 3 replicates. (C) depicts the efficacy of systemic tumor eradication by tumor- sensing T (TTS) cells assessed by infusing 1 .0 x 106 T cells intravenously (IV) into NSG mice bearing luciferase expressing CD19+PSMA+ PC3 human prostate tumor. Tumor burden was quantitatively measured weekly by using BLI. Images of two representative mice from each group are shown with the pixel intensity from the luminescence of tumors represented in color. An average of tumor burden was plotted with error bars representing standard deviation from the mean of values from 6 mice per group. (D) depicts selective eradication of DP tumors using a tri-tumor mouse model by subcutaneously injecting 1 x 106 PC3 tumors cells each of ceils positive for CD19 alone into the left flanks, cells positive for PSMA alone into the right flanks, and cells positive for both CD19 and PSMA into the backs of the mice. T ceils expressing either 19z1 , P28BB, or both 19z1 + P28BB of the chimeric receptors were infused intravenously 7 days post tumor infusion. Representative images of 2 mice per group bearing these tumors are shown with luminescence of tumors represented in color. Tumors were quantitatively measured using calipers and tumor volumes were plotted versus time for each tumor. Error bars represent standard deviation from the mean of 6 mice. Statistical significance was determined using two- tailed unpaired t tests to compare values obtained from 19z1 T cells and 19z1 + P28BB T cells and p values are represented as * for <0.05 or ** for <0.01.
[0022] Figures 3A-E depict that tumor-sensing T (TTS) ceils selectively eradicated human prostate tumors when targeting two prostate tumor antigens (A) depicts the evaluation of three different scFvs specific to PSCA for their assembly into bispecific antibodies that contain specificity for CD3 as well. T ceils were co- cultured at ratio of 20:1 with PSCA+ PC3 tumor cells and antibodies added at varying amounts and specific iysis was measured. (B) depicts generation of CARs using the anti-PSCA scFvs that display varied efficacy in cytotoxicity assays. The CAR mediated specific Iysis of target cells expressing PSCA corroborated the reduced efficacy of the Lzl scFv by requiring a 50 fold high effector : target ratio to achieve the same level of Iysis of that for either Hzi or Mzl. (C) and (D) depict selective eradication of systemic prostate tumors expressing PSCA and PSMA was investigated by using these inefficient scFvs. Tumors (Figure 5) were established and treated as described in Figure 2. After 14 days, 1 .0 x 106 chimeric receptor positive T cells for Mzl +P28BB (Figure 3C) or Lzl + P28BB (Figure 3D) were infused intravenously. Images of two representative mice from each group are shown with luminescence from tumors represented in color (from Blue = 5 x 105 to Red = 2 x 107 photons). The average tumor burden was quantified by luminescence and plotted with error bars representing standard deviation from the mean of values from 5 mice per group. Two mice that received PSMA tumor (green line) died after day 49 and therefore the mean value for luminescence was averaged from 3 values for days 56 and 63. Figure 3E Selective antitumor responses to only PSCA+PSMA+ tumors was achieved by Lzi + P28BB T cells in mice that also had PSCA'PSMA* and PSCA+PSMA" tumors, similar to Figure 2D. Statistical significance was determined using two-tailed unpaired t tests to compare values obtained from Lzl T cells and Lzl + P28BB T cells and p values are represented as * for <0.05 or ** for <0.01.
[0023] Figures 4A-D depict enhanced cytokine secretion and Bd xL
expression is found by TTS ceils when co-cultured on DP tumors. (A) depicts multiplex cytokine analysis of untransduced T cells or T cells transduced with 19z1 , P28BB, or both 48 hours post first antigen stimulation using either untransduced PC3 cells (Empty) or CD19+PSMA+ PC3 ceils. Error bars represent standard deviation from the mean of 2 biological replicates. (B-D) depict multiplex cytokine analysis of untransduced T cells or T cells transduced with Hz] (B), Mzl (C), and Lz1 (D) anti-PSCA CARs, P28BB CCRs, or both CAR + CCR is shown 48 hours post second antigen stimulation using either Empty or PSCA+PSMA+ PC3 cells. (E) depicts Western blot analysis for BdxL performed using cellular lysates of untransduced T ceils or T ceils transduced with 19z1 , P28BB, or both after 24 hours post initial antigen stimulation. Total amount of Akt was used as a loading control.
[0024] Figure 5 depicts generation of prostate tumor cells for the expression of fusion protein GFP-Firefiy Luciferase (GFP/Luc) and tumor antigens. Untransduced PC3 cells (Empty) were transduced with GFP/Luc and either CD19, PSMA, PSCA, or a combination of two antigens using retroviral expression constructs. Cells were purified via double purity FACS for GFP/Luc, CD19, PSMA, and/or PSCA.
[0025] Figures 6A-C illustrate the tumor-sensing T cell concept. (A) depicts that TTS cells expressing an efficient CAR, become potently stimulated by A+113+ cells to facilitate immune response against A+ ceils. CAR+CCR+ cells can bind tumor antigen A+ cells with a CAR that supplies CD3 activation signals. This can result in short-term cell lysis. CAR+CCR+ cells can bind tumor antigen B+ cells with a CCR that supplies CD28 and CD137 signals. This signal alone is not sufficient to induce lysis or proliferation. Only when CAR+CCR+ cells bind tumor antigen A+B+ ceils with a CAR and CCR can both activation and stimulation be provided. This results in robust lysis, T cell proliferation, enhanced cytokine secretion, upregulation of BclxL, and the ability to selectively eradicate tumors in vivo. However, depending on the efficacy of the CAR, these CAR+CCR+ cells can potentially recirculate to lyse cells single positive for antigen specific to the CAR. Figure 6B depicts that by reducing the efficacy of the CAR, TTs celis can be functionally rescued by CCR binding when A+B+ cells are encountered to selectively respond and eradicate A+13+ ceils, while avoiding response to A+ cells. (C) shows that by co-expressing one CAR that supplies a TCR activation signal upon binding a tumor antigen and a second CAR that supplies stimulation signals upon binding a different tumor antigen, T lymphocytes will only eradicate tumors expressing both antigens, but not tumors expressing either antigen alone.
Detailed Description of the invention
[0026] All patents, published applications and other references mentioned in this specification are herein incorporated by reference into the present disclosure. [0027] Unless defined otherwise, all technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art to which this invention belongs. The following references provide one of skill with a general definition of many of the terms used in this invention: Singleton et al., Dictionary of Microbiology and Molecular Biology (2nd ed. 1994); The
Cambridge Dictionary of Science and Technology (Walker ed., 1988); The Glossary of Genetics, 5th Ed., R. Rieger et al. (eds.), Springer Verlag ( 1991 ); and Hale & Marham, The Harper Collins Dictionary of Biology (1991 ). As used herein, the following terms have the meanings ascribed to them below, unless specified otherwise.
[0028] By "activates an immunoresponsive ceil" is meant induction of signal transduction or changes in protein expression in the cell resulting in initiation of an immune response. For example, when CD3 Chains cluster in response to ligand binding and immunoreceptor tyrosine-based inhibition motifs (ITAMs) a signal transduction cascade is produced. In certain embodiments, when an endogenous TCR or an exogenous CAR binds antigen, a formation of an immunologicai synapse occurs that includes clustering of many molecuies near the bound receptor (e.g. CD4 or CD8, CD3 / / / , etc.) This clustering of membrane bound signaling molecules allows for ITAM motifs contained within the CD3 chains to become phosphorylated. This phosphorylation in turn initiates a T ceil activation pathway ultimately activating transcription factors, such as NF-KB and AP-1. These transcription factors induce global gene expression of the T cell to increase IL-2 production for proliferation and express master regulator T cell proteins in order to initiate a T ceil mediated immune response. By "stimulates an immunoresponsive cell" is meant a signal that results in a robust and sustained immune response. In various embodiments, this occurs after immune cell (e.g., T-cell) activation or concomitantly mediated through receptors including, but not limited to, CD28, CD137 (4-1 BB), OX40, and ICOS. Without being bound to a particular theory, receiving multiple stimulatory signals is important to mount a robust and long-term T cell mediated immune response. Without receiving these stimulatory signals, T cells quickly become inhibited and unresponsive to antigen. While the effects of these co-stimuiatory signals vary and remain partially understood, they generally result in increasing gene expression in order to generate long lived, proliferative, and anti-apoptotic T cells that robustly respond to antigen for complete and sustained eradication.
[0029] The term "antigen recognizing receptor" as used herein refers to a receptor that is capable of activating an immune cell (e.g., a T-cell) in response to antigen binding. Exemplary antigen recognizing receptors may be native or endogenous T cell receptors or chimeric antigen receptors in which a tumor antigen-binding domain is fused to an intracellular signaling domain capable of activating activating an immune cell (e.g., a T-cell). In various embodiments, an antigen recognizing receptor is selected to have low or minimal affinity or avidity for the antigen.
[0030] By "affinity" is meant a measure of the binding strength between antibody and a simple hapten or antigen determinant. Without being bound to theory, affinity depends on the closeness of stereochemical fit between antibody combining sites and antigen determinants, on the size of the area of contact between them, and on the distribution of charged and hydrophobic groups. Affinity also includes the term "avidity," which refers to the strength of the antigen-antibody bond after formation of reversible complexes. Methods for calculating the affinity of an antibody for an antigen are known in the art, including use of binding experiments to calculate affinity. In the case of an antibody (Ab) binding to an antigen (Ag), the affinity constant is used (expressed as inverted dissociation constant).
A b + A g = A b A g
Ka = iAbAql = 1
[Ab)] [Ag] Ka
The chemical equilibrium of antibody binding is also the ratio of the on-rate (k forward) and off-rate (kback) constants. Two antibodies can have the same affinity, but one may have both a high on- and off-rate constant, while the other may have both a low on- and off-rate constant.
Ka = ^forward = on-rate
¾ack off-rate Antibody activity in functionai assays (e.g., cell lysis assay) is also reflective of antibody affinity. In various embodiments of the invention, the antigen recognizing receptor has low affinity. Low affinity includes micromolar and nanomolar affinities (e.g. IfJ5, 50"6, I0"6, 5x 1fJ7, 10"7, 5x 10"8, 10"8, 5x1 fJ9, iO"9 M). Anitbody and affinities can be phenotypically characterized and compared using functional assay (e.g., cell lysis assay).
[0031] By "affinity" is meant a measure of the binding strength between antibody and a simple The term "chimeric co-stimulatory receptor" (CCR), as used herein refers to a specific type of chimeric antigen receptor (CAR) that mediates costimulation independently of activation. When expressed on immunoresponsive cells in combination with an antigen recognizing receptor (e.g., CAR or TCR that activates the cell), the CCR is targeted to a second antigen. In certain embodiments, the CCR has mid or high affinity for its target antigen.
[0032] The term "chimeric antigen receptor" (CAR) as used herein refers to a tumor antigen-binding domain that is fused to an intracellular signaling domain capable of activating or stimulating T ceils. Most commonly, the CAR'S
extracellular binding domain is composed of a single chain variable fragment (scFv) derived from fusing the variable heavy and light regions of a murine or humanized monoclonal antibody. Alternatively, scFv's may be used that are derived from Fab's (instead of from an antibody, e.g., obtained from Fab libraries). In various embodiments, this scFv is fused to a transmembrane domain and then to intracellular signaling domain. "First-generation" CARs include those that solely provide CD3 signals upon antigen binding, "Second-generation" CARs include those that provide both costimulation (e.g. CD28 or CD137) and activation (CD3 ). "Third-generation" CARs include those that provide multiple
costimulation (e.g. CD28 and CD137) and activation (CD3 ). in CAR
applications to date, the CAR is selected to have high affinity or avidity for the antigen, which is distinct and distinguishable from the invention described herein. [0033] By "CD3 polypeptide" is meant a protein having at least 85, 90, 95, 96, 97, 98, 99 or 100% identity to NCBI Reference No: NP_932170 or a fragment thereof that has activating or stimulatory activity. An exemplary CD3 is provided in Table 1 below. By "CD3 nucleic acid molecule" is meant a polynucleotide encoding a CD3 polypeptide.
[0034] By "CD8 polypeptide" is meant a protein having at least 85, 90, 95, 96, 97, 98, 99or 100% identity to NCBI Reference No: NP_001759 or a fragment thereof that has stimulatory activity. An exemplary CD8 is provided in Table 1 below. By "CD8 nucleic acid molecule" is meant a polynucleotide encoding a CD8 polypeptide.
[0035] By "CD28 polypeptide" is meant a protein having at least 85, 90, 95, 96, 97, 98, 99 or 100% identity to NCBI Reference No: NP_006130 or a fragment thereof that has stimulatory activity. An exemplary CD28 is provided in Table 1 below. By "CD28 nucleic acid molecule" is meant a polynucleotide encoding a CD28 polypeptide.
[0036] By "4-1 BB polypeptide" is meant a protein having at least 85, 90, 95,
96, 97, 98, 99 or 100% identity to NCBI Reference No: P41273 or NP_001552 or a fragment thereof that that acts as a tumor necrosis factor (TNF) iigand. An exemplary 4-1 BB is provided in Table 1 below. By "4-1 BBL nucleic acid molecule" is meant a polynucleotide encoding a 4-1 BBL polypeptide.
[0037] By "CD80 polypeptide" is meant a protein having at least 85, 90, 95, 96,
97, 98, 99 or 100% identity to NCBI Reference No: NP_005182 or a fragment thereof that acts as an ig superfamily Iigand. An exemplary CD80 polypeptide is provided in Table 1 below.
[0038] By "CD80 nucleic acid molecule" is meant any polynucleotide encoding a CD80 polypeptide. An exemplary CD80 nucleic acid molecule is NM_005191 .
[0039] By "OX40L polypeptide" is meant a protein having at least 85, 90, 95, 96,
97, 98, 99 or 100% identity to NCBI Reference No: BAB18304 or NP_003317 or a fragment thereof that is a tumor necrosis factor (TNF) ligand. By "OX40L nucleic acid molecule" is meant a polynucleotide encoding a OX40L polypeptide.
[0040] By "19z1 polypeptide" is meant a protein having at least 85, 90, 95, 96, 97, 98, 99 or 100% identity to the sequence provided below and having activating activity when bound to CD19.
[0041] By "P28z polypeptide" is meant a protein having at least 85, 90, 95, 96, 97, 98, 99 or 100% identity to the sequence provided below.
[0042] By " CD19" is meant a protein having at least 85, 90, 95, 96, 97, 98, 99 or 100% identity to the sequence provided below and is able to bind CD19.
[0043] By " PSMA" is meant a protein having at least 85, 90, 95, 96, 97, 98, 99 or 100% identity to the sequence provided below and is able to bind PSMA.
[0044] By "P28BB" is meant a protein having at least 85, 90, 95, 96, 97, 98, 99 or 100% identity to the sequence provided below and having stimulatory activity when bound to PSMA.
Table 1
SEQ ID Name
NO.
1
CD3 ζ mkwkalftaa ilqaqlpite aqsfglldpk lcylldgilf iygviltalf Irvkfsrsad apayqqgqnq lynelnlgrr eeydvldkrr grdpemggkp qrrknpqegl ynelqkdkma eayseigmkg errrgkghdg lyqglsta tkdtydalhm qalppr
2
CD8 malpvtalll plalllhaar psqfrvspld rtwnlgetve
Ikcqvllsnp tsgcs lfqp rgaaasptfl lylsqnkpka aegldtqrfs gkrlgdtfvl tlsdfrrene gyyfcsalsn simyfshfvp vflpakpttt paprpptpap tiasqplslr peacrpaagg avhtrgldfa cdiyiwapla gtcgvlllsl vitlycnhrn rrrvckcprp vvksgdkpsl saryv
3
CD28 mlrlllalnl fpsiqvtgnk ilvkqspmlv aydnavnlsc kysynlfsre fraslhkgld savevcvvyg nysqqlqvys ktgfncdgkl gnesvtfylq nlyvnqtdiy fckievmypp pyldneksng tiihvkgkhl cpsplfpgps kpf vlvwg gvlacysllv tvafiifwvr skrsrllhsd ymnmtprrpg ptrkhyqpya pprdfaayrs
mgnscyniva tlllvlnfer trslqdpcsn cpagtfcdnn
4 4-1 BB rnqicspcpp nsfssaggqr tcdicrqckg vfrtrkecss tsnaecdctp gfhclgagcs mceqdckqgq eltkkgckdc cfgtfndqkr gicrpwtncs Idgksvlvng tkerdvvcgp spadlspgas svtppapare pghspqiisf flaltstall fllffltlrf svvkrgrkkl lyifkqpfmr pvqttqeedg cscrfpeeee ggcel
raghtrrqgts pskcpylnff qllvlaglsh fcsgvihvtk
CD80 evkevatlsc ghnvsveela qtriywqkek kmvltmmsgd mni peyknr tifditnnls ivilalrpsd egtyecvvlk yekdafkreh laevtlsvka dfptpsisdf eiptsnirri icstsggfpe phlswlenge elnainttvs qdpetelyav sskldfnmtt nhsfmcliky ghlrvnqtfn wnttkqehfp dnllpswait lisvngif i ccltycfapr crerrrnerl rresvrov mervqpleen vgnaarprfe rnklllvasv iqglgl llcf
OX40L tyiclhfsal qvshrypriq sikvqfteyk kekgfiltsq kedeimkvqn nsviincdgf ylislkgyfs qevnislhyq kdeeplfqlk kvrsvnslmv asltykdkvy lnvttdntsl ddfhvngge1 ilihqnpgef cvl 9z1 MALPVTALLLPLALLLHAEVKLQQSGAELVRPGSSVKISCKAS
GYAFSSYWMNWKQRPGQGLE IGQIYPGDGDTNYNGKFKGQA TLTADKSSSTAYMQLSGLTSEDSAVYECARKTISSWDFYFDY WGQGTTVTVSSGGGGSGGGGSGGGGSDIELTQSPKFMSTSVGD RVSVTCKASQ VGT VAWYQQKPGQSPKPLIYSATYRNSGVPD RFTGSGSGTDFTLTITNVQSKDLADYFCQQYNRYPYTSGGGTK LEIKRAAAPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGA VHTRGLDFACDIYI APLAGTCGVLLLSLVITLYCNHRVKFSR SAEPPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKP RRK PQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQG LSTATKDTYDALHMQALPPR
MALPVTALLLPLALLLHAEVQLQQSGPELVKPGTSVRISCKTSGYTF
P28z TEYTIHWVKQSHGKSLE IGNINPNNGGTTYNQKFEDKATLTVDKSS
STAYMELRSLTSEDSAVYYCAAG NFDY GQGTTVTVSSGGGGSGGG GSGGGGSDIVM QSHKFMSTSVGDRVS11CKASQDVG A D YQQKP GQSPKLLIYWASTRHTGVPDRFTGSGSGTDFTLTITNVQSEDLADYF CQQY SYPLTFGAGTMLDLKRAAAIEVMYPPPYLDNEKSNGTIIHVK GKHLCPSPLFPGPSKPF VLVWGGVLACYSLLVTVAF11F VRSKR SRLLHSDYM MTPRRPGPTRKHYQPYAPPRDFAAYRSRVKFSRSADA PAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEG LYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDAL HMQALPPR
.VKLQQSGAELVRPGSSVKISCKASGYAFSSYWMNWVKQRPGQGLEW 9 CD19 IGQIYPGDGDTNYNGKFKGQATLTADKSSSTAYMQLSGLTSEDSAVY FCARKTISSVVDFYFDY GQGTTVTVSSGGGGSGGGGSGGGGSDIEL TQSPKFMSTSVGDRVSVTCKASQNVGTNVAWYQQKPGQSPKPLIYSA TYRNSGVPDRFTGSGSGTDFTLTITNVQSKDLADYFCQQYNRYPYTS GGGTKLEIKR
M ALPVTALLLPLALLLHAEVQLQQSGPELVKPGTSVRISCKTSGYT
10 PSMA FTEYTIHWVKQSHGKSLEWIGNINPNNGGTTYNQKFEDKATLTVDKS
SS AYMELRSLTSEDSAVYYCAAG NFDYWGQG VTVSSGGGGSGG GGSGGGGSDIVMTQSHKFMSTSVGDRVSI ICKASQDVGTAVDWYQQK PGQSPKLLIYWASTRHTGVPDRFTGSGSGTDFTL I VQSEDLADY FCQQYNSYPLTFGAGTMLDLKR
MALPVTALLLPLALLLHAEVQLQQSGPELVKPGTSVRISCKTSGYTF
11 P28BB TEYTIH VKQSHGKSLEWIGNINPNNGGTTYNQKFEDKATLTVDKSS
STAYMELRSLTSEDSAVYYCAAGWNFDYWGQGTTVTVSSGGGGSGGG GSGGGGSDIV TQSHKFMSTSVGDRVSIICKASQDVGTAVDWYQQKP GQSPKLLIY ASTRHTGVPDRFTGSGSGTDFTLTITNVQSEDLADYF CQQYNSYPLTFGAGTMLDLKRAAAIEVMYPPPYLD EKSNGTI IHVK GKHLCPSPLFPGPSKPF VLVWGGVLACYSLLVTVAF11FWVRSKR SRLLHSDYMNMTPRRPGPTRKHYQPYAPPRDFAAYRSRFSVVKRGRK KLLYIFKQPF RPVQTTQEEDGCSCRFPEEEEGGCE
[0045] Nucleic acid molecules useful in the methods of the invention include any nucleic acid molecule that encodes a polypeptide of the invention or a fragment thereof. Such nucleic acid molecules need not be 100% identical with an endogenous nucleic acid sequence, but will typically exhibit substantial identity. Polynucleotides having "substantial identity" to an endogenous
sequence are typically capable of hybridizing with at least one strand of a double- stranded nucleic acid molecule. By "hybridize" is meant pair to form a double- stranded molecule between complementary polynucleotide sequences (e.g., a gene described herein), or portions thereof, under various conditions of stringency. (See, e.g., Wahi, G. M. and S. L. Berger (1987) Methods Enzymol. 152:399; Kimmel, A. R. (1987) Methods Enzymol. 152:507).
[0046] For example, stringent salt concentration will ordinarily be less than about 750 mM NaCI and 75 mM trisodium citrate, preferably less than about 500 mM NaC1 and 50 mM trisodium citrate, and more preferably less than about 250 mM NaC1 and 25 mM trisodium citrate. Low stringency hybridization can be obtained in the absence of organic solvent, e.g., formamide, while high
stringency hybridization can be obtained in the presence of at least about 35% formamide, and more preferably at least about 50% formamide. Stringent temperature conditions wiil ordinarily inciude temperatures of at least about 30° C, more preferably of at least about 37° C, and most preferably of at least about 42° C. Varying additional parameters, such as hybridization time, the concentration of detergent, e.g., sodium dodecyl sulfate (SDS), and the inclusion or exclusion of carrier DNA, are well known to those skilled in the art. Various levels of stringency are accomplished by combining these various conditions as needed. In a preferred: embodiment, hybridization will occur at 30° C in 750 mM NaCI, 75 mM trisodium citrate, and 1% SDS. In a more preferred embodiment, hybridization will occur at 37° C in 500 mM NaCI, 50 mM trisodium citrate, 1 % SDS, 35% formamide, and 1001.1 g/m1 denatured salmon sperm DNA (ssDNA). In a most preferred embodiment, hybridization wiil occur at 42° C C. in 250 mM NaCI, 25 mM trisodium citrate, 1 % SDS, 50% formamide, and 200 p.g/ml ssDNA. Useful variations on these conditions will be readily apparent to those skilled in the art.
[0047] For most applications, washing steps that follow hybridization will also vary in stringency. Wash stringency conditions can be defined by salt concentration and by temperature. As above, wash stringency can be increased by decreasing salt concentration or by increasing temperature. For example, stringent salt concentration for the wash steps will preferably be less than about 30 mM NaC1 and 3 mM trisodium citrate, and most preferably less than about 15 mM NaCI and 1.5 mM trisodium citrate. Stringent temperature conditions for the wash steps wiil ordinarily inciude a temperature of at least about 25° C, more preferably of at least about 42° C, and even more preferably of at least about 68° C. In a preferred embodiment, wash steps wiil occur at 25°C in 30 mM NaCI, 3 mM trisodium citrate, and 0.1% SDS. In a more preferred embodiment, wash steps will occur at
42. degree C. in 15 mM NaC1 , 1.5 mM trisodium citrate, and 0.1 % SDS. In another embodiment, wash steps wiil occur at 68° C in 15 mM NaC1 , 1.5 mM trisodium citrate, and 0.1% SDS. Additional variations on these conditions will be readily apparent to those skilled in the art.
[0048] Hybridization techniques are well known to those skilled in the art and are described, for example, in Benton and Davis (Science 196:180, 1977); Grunstein and Hogness (Proc. Natl. Acad.Sci., USA 72:3961 , 1975); Ausubel et al. (Current Protocols in Molecular Biology, Wiley Interscience, New York, 2001 ); Berger and Kimmei (Guide to Molecular Cloning Techniques, 1987, Academic Press, New York); and Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, New York.
[0049] By "substantially identical" is meant a polypeptide or nucleic acid molecule exhibiting at least 50% identity to a reference amino acid sequence (for example, any one of the amino acid sequences described herein) or nucleic acid sequence (for example, any one of the nucleic acid sequences described herein). Preferably, such a sequence is at least 60%, more preferably 80% or 85%, and more preferably 90%, 95% or even 99% identical at the amino acid level or nucleic acid to the sequence used for comparison.
[0050] Sequence identity is typically measured using sequence analysis software (for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, BESTFIT, GAP, or PILEUP/PRETTYBOX programs). Such software matches identical or similar sequences by assigning degrees of homology to various substitutions, deletions, and/or other modifications.
Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparagine, g!utamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. In an exemplary approach to determining the degree of identity, a BLAST program may be used, with a probability score between e-3 and e-100 indicating a closely related sequence.
[0051] By "analog" is meant a structurally related polypeptide or nucleic acid molecule having the function of a reference polypeptide or nucleic acid molecule.
[0052] The term "iigand" as used herein refers to a molecule that binds to a receptor. In particular, the iigand binds a receptor on another cell, allowing for cell-to-cel! recognition.
[0053] The term "constitutive expression" as used herein refers to expression under all physiological conditions.
[0054] By "disease" is meant any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ. Examples of diseases include neoplasia or pathogen infection of cell. [0055] By "effective amount" is meant an amount sufficient to arrest, ameliorate, or inhibit the continued proiiferation, growth, or metastasis (e.g., invasion, or migration) of a neoplasia.
[0056] By "enforcing tolerance" is meant preventing the activity of self-reactive cells or immunoresponsive cells that target transplanted organs or tissues.
[0057] By "exogenous" is meant a nucleic acid molecule or polypeptide that is not endogenously present in the cell, or not present at a level sufficient to achieve the functional effects obtained when over-expressed. The term
"exogenous" would therefore encompass any recombinant nucleic acid molecule or polypeptide expressed in a cell, such as foreign, heterologous, and over- expressed nucleic acid molecules and polypeptides.
[0058] By a "heterologous nucleic acid molecule or polypeptide" is meant a nucleic acid molecule (e.g., a cDNA, DNA or RNA molecule) or polypeptide that is not normally present in a cell or sample obtained from a cell. This nucleic acid may be from another organism, or it may be, for example, an mRNA molecule that is not normally expressed in a cell or sample.
[0059] By "immunoresponsive cell" is meant a cell that functions in an immune response or a progenitor, or progeny thereof.
[0060] By "isolated cell" is meant a ceil that is separated from the molecular and/or cellular components that naturally accompany the cell.
[0061] The terms "isolated," "purified," or "biologically pure" refer to material that is free to varying degrees from components which normally accompany it as found in its native state. "Isolate" denotes a degree of separation from original source or surroundings. "Purify" denotes a degree of separation that is higher than isolation. A"purified" or "biologically pure" protein is sufficiently free of other materials such that any impurities do not materially affect the biological properties of the protein or cause other adverse consequences. That is, a nucleic acid or peptide of this invention is purified if it is substantially free of cellular material, viral material, or culture medium when produced by
recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized. Purity and homogeneity are typically determined using analytical chemistry techniques, for example, polyacryiamide gel
electrophoresis or high performance liquid chromatography. The term "purified" can denote that a nucleic acid or protein gives rise to essentially one band in an electrophoretic gel. For a protein that canbe subjected to modifications, for example, phosphorylation or glycosylation, different modifications may give rise to different isolated proteins, which can be separately purified.
[0062] The term "tumor antigen-binding domain" as used herein refers to a domain capable of specifically binding a particular antigenic determinant or set of antigenic determinants present on a tumor.
[0063] By "modulate" is meant to alter positively or negatively. Exemplary modulations include a 1%, 2%, 5%, 10%, 25%, 50%, 75%, or 100% change.
[0064] By "neoplasia" is meant a disease characterized by the pathological proliferation of a cell or tissue and its subsequent migration to or invasion of other issues or organs. Neoplasia growth is typically uncontrolled and progressive, and occurs under conditions that would not elicit, or would cause cessation of, multiplication of normal cells. Neoplasias can affect a variety of cell types, tissues, or organs, including but not limited to an organ selected from the group consisting of bladder, bone, brain, breast, cartilage, glia, esophagus, fallopian tube, gallbladder, heart, intestines, kidney, liver, lung, lymph node, nervous tissue, ovaries, pancreas, prostate, skeletal muscle, skin, spinal cord, spleen, stomach, testes, thymus, thyroid, trachea, urogenital tract, ureter, urethra, uterus, and vagina, or a tissue or cell type thereof. Neoplasias include cancers, such as sarcomas, carcinomas, or plasmacytomas {malignant tumor of the plasma cells). Illustrative neoplasms for which the invention can be used include, but are not limited to leukemias (e.g., acute Ieukemia, acute lymphocytic Ieukemia, acute myelocytic leukemi a, acute myeloblasts Ieukemia, acute promyelocytic ieukemia, acute myelomonocytic ieukemia, acute monocytic Ieukemia, acute erythroieukemia, chronic Ieukemia, chronic myelocytic Ieukemia, chronic lymphocytic Ieukemia), polycythemia vera, lymphoma (Hodgkin's disease, non-Hodgkin's disease),
Waldenstrom's macroglobulinemia, heavy chain disease, and solid tumors such as sarcomas and carcinomas {e.g., fibrosarcoma, myxosarcoma,
liposarcoma.chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma,iymphangiosarcoma,lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous giand carcinoma, papillary carcinoma, papillary
adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal ceil carcinoma, hepatoma, nile duct carcinoma,
choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, uterine cancer, testicular cancer, lung carcinoma, small ceil lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medullobiastoma, craniopharyngioma, ependymoma, pinealoma,
hemangioblastoma, acoustic neuroma, !igodenroglioma, schwannoma, meningioma, melanoma, neuroblastoma, and retinoblastoma). In one embodiment, screening methods of the invention identify compositions that are useful for treating breast or lung cancer.
[0065] By "receptor" is meant a polypeptide, or portion thereof, present on a cell membrane that selectively binds one or more ligands.
[0066] By "recognize" is meant selectively binds a target. A T cell that recognizes a virus typically expresses a receptor that binds an antigen expressed by the virus.
[0067] By "pathogen" is meant a virus, bacteria, fungi, parasite or protozoa capable of causing disease. Exemplary viruses include, but are not limited to, Retroviridae (e.g. human immunodeficiency viruses, such as HIV-1 (also referred to as HDTV-ill, LAVE or HTLV-III/LAV, or HIV-III; and other isolates, such as HIV- LP; Picornaviridae (e.g. polio viruses, hepatitis A virus; enteroviruses, human Coxsackie viruses, rhinoviruses, echoviruses); Calciviridae (e.g. strains that cause gastroenteritis); Togaviridae (e.g. equine encephalitis viruses, rubella viruses); Flaviridae (e.g. dengue viruses, encephalitis viruses, yellow fever viruses);
Coronoviridae (e.g. coronaviruses); Rhabdoviridae (e.g. vesicular stomatitis viruses, rabies viruses); Filoviridae (e.g. ebola viruses); Paramyxoviridae (e.g. parainfluenza viruses, mumps virus, measles virus, respiratory syncytial virus); Orthomyxoviridae (e.g . influenza viruses); Bungaviridae (e.g. Hantaan viruses, bunga viruses, phieboviruses and Nairo viruses); Arena Viridae (hemorrhagic fever viruses); Reoviridae (e.g. reoviruses, orbiviurses and
rotaviruses);S/'ma r/'c/ae; Hepadnaviridae (Hepatitis B virus); Parvovihda {parvov\ruses);Papovaviridae (papilloma viruses, polyoma viruses);
Adenoviridae (most adenoviruses); Herpesviridae (herpes simplex virus (HSV) 1 and 2, varicella zoster virus, cytomegalovirus (CMV), herpes virus; Poxviridae (variola viruses, vaccinia viruses, pox viruses); and Iridoviridae (e.g. African swine fever virus); and unclassified viruses (e.g. the agent of delta hepatitis (thought to be a defective satellite of hepatitis B virus), the agents of non-A, non-B hepatitis (class 1 = internally transmitted; class 2 = parenterally transmitted (i.e. Hepatitis C); Norwalk and related viruses, and astroviruses).
[0068] Exemplary bacteria include, but are not limited to, Pasteurella, Staphylococci, Streptococcus, Escherichia coli, Pseudomonas species, and Salmonella species. Specific examples of infectious bacteria include but are not limited to, Helicobacter pyloris, Borelia burgdoiferi, Legionella
pneumophilia, Mycobacteria sps (e.g. M. tuberculosis, M. avium, M.
intracellulare, M. kansaii, M. gordonae), Staphylococcus aureus, Neisseria gonorrhoeae, Neisseria meningitidis, Listeria monocytogenes,
Streptococcus pyo genes (Group A Streptococcus), Streptococcus agalactiae (Group B Streptococcus), Streptococcus (viridans group), Streptococcus faecalis, Streptococcus bovis, Streptococcus (anaerobic sps.), Streptococcus pneumoniae, pathogen cCampylobacter sp.,
Enterococcus sp,, Haemophilus influenzae, Bacillus antracis,
corynebacterium diphtheriae, corynebacterium sp., Erysipelothrix rhusiopathiae, Clostridium perfringers, Clostridium tetani, Enterobacter aerogenes, Klebsiella pneumoniae, Pasturella multocida, Bacteroides sp., Fusobacterium nucleatum, Streptobacillus moniliformis, Treponema pallidium, Treponema pertenue, Leptospira, Rickettsia, and Actinomyces israelii.
[0069] By "specifically binds" is meant a polypeptide or fragment thereof that recognizes and binds a polypeptide of interest, but which does not
substantially recognize and bind other molecules in a sample, for example, a biological sample, which naturally includes a polypeptide of the invention.
[0070] The term "tumor antigen" as used herein refers to any polypeptide expressed by a tumor that is capable of inducing an immune response.
[0071] By "virus antigen" is meant a polypeptide expressed by a virus that is capable of inducing an immune response. [0072] The terms "comprises", "comprising", and are intended to have the broad meaning ascribed to them in U.S. Patent Law and can mean "includes", "including" and the like.
[0073] As used herein, "treatment" refers to clinical intervention in an attempt to alter the disease course of the individual or cell being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Therapeutic effects of treatment include, without limitation, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastases, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. By preventing progression of a disease or disorder, a treatment can prevent deterioration due to a disorder in an affected or diagnosed subject or a subject suspected of having the disorder, but also a treatment may prevent the onset of the disorder or a symptom of the disorder in a subject at risk for the disorder or suspected of having the disorder.
[0074] The term "subject" as used herein refers to a vertebrate, preferably a mammal, more preferably a human.
[0075] The term "immunocompromised" as used herein refers to a subject who has an immunodeficiency. The subject is very vulnerable to opportunistic infections, infections caused by organisms that usually do not cause disease in a person with a healthy immune system, but can affect people with a poorly functioning or suppressed immune system.
[0076] Other aspects of the invention are described in the following disclosure and are within the ambit of the invention.
[0077] The present invention generally provides cells, including genetically modified immunoresponsive cells (e.g., T cells, Natural Killer (NK) cells, cytotoxic T lymphocytes (CTL) ceils) expressing at least a combination of an antigen- recognizing receptor (e.g., TCR or CAR) and a chimeric co-stimulating receptor (CCR), and methods of use therefore for the treatment of neoplasia and other pathologies where an increase in an antigen-specific immune response is desired. The invention is based, at least in part, on the discovery that the simultaneous engagement of two antigens co-expressed by a tumor cell by an antigen-recognizing receptor and chimeric co-stimulating receptor is useful for activating and stimulating an immunoreactive cell without systemic effects. In particular, the reactivity against tissues expressing either antigen alone is preferably minimal, inducing T cell activaiion in the presence of both antigens but not either one alone. T ceil activation is mediated by a TCR or a CAR targeted to an antigen (e.g., CD19 or prostate stem cell antigen, PSCA). Costimulation is independently mediated by a "chimeric costimulatory receptor" (CCR), 2' 3 which is targeted to a second antigen (e.g., prostate-specific membrane antigen, PSMA). Such an approach resulted in augmented reactivity against dual-antigen positive (DP) tumors, but failed to avert enhanced reactivity against single antigen positive (SP) tumors, it was found that tumor sensing T cells could be made to differentiate DP tumors from SP tumors by attenuating T ceil activation to a level where T cell activation is by itself ineffective, but functionally rescued at the tumor site by a CCR engaged by an independent, co-expressed antigen. This approach provides immunogenicity within the tumor microenvironment for tumor eradication while not affecting SP cells that are normal or non-neoplastic and represents a significant advance over conventional adoptive T cell therapy.
[0078] Furthermore, this approach is not limited to the treatment of neoplasias, but is amenable to a wide range of applications where an increase in an antigen-specific immune response is desired, such applications include not only the treatment of neoplasias, but also for the enhancement of an immune response against a pathogen infection or an infectious disease and to reinforce immune tolerance in regulatory T ceils in the context of autoimmunity or allogeneic transplantation.
Hematopoietic Cell Lineages
[0079] Mammalian hematopoietic (blood) cells provide a diverse range of physiologic activities. Hematopoietic ceils are divided into lymphoid, myeloid and erythroid lineages. The lymphoid lineage, comprising B, T and natural killer (NK) cells, provides for the production of antibodies, regulation of the cellular immune system, detection of foreign agents in the blood, detection of cells foreign to the host, and the iike. The term "T cells" as used herein refers to lymphocytes that mature in the thymus and are chiefly responsible for cell- mediated immunity. T cells are involved in the adaptive immune system. The term "natural killer (NK) ceils" as used herein refers to lymphocytes that are part of cell-mediated immunity and act during the innate immune response. They do not require prior activation in order to perform their cytotoxic effect on target ceils. Cytotoxic T cells (CTL or killer T ceils) are a subset of T
lymphocytes capable of inducing the death of infected somatic or tumor cells.
Cells for Use in the Methods of the Invention
[0080] The present invention provides cells expressing a combination of an antigen-recognizing receptor that activates an immunoresponsive ceil (e.g., TCR, CAR) and a chimeric co-stimulating receptor (CCR), and methods of using such cells for the treaiment of a disease that requires an enhanced immune response. In one approach, tumor antigen-specific T cells, NK cells, CTL ceils or other immunoresponsive cells are used as shuttles for the selective enrichment of one or more co-stimulatory ligands for the treaiment or prevention of neoplasia. For example, a T cell expressing a chimeric antigen receptor 19z1 that recognizes CD19 is co-expressed in a T cell that expresses a chimeric co- stimuiatory receptor P28BB that recognizes and binds Prostate Specific
Membrane Antigen (PSMA). Such cells are administered to a human subject in need thereof for the treatment or prevention of prostate cancer. In another approach, viral antigen-specific T cells, NK cells, CTL cells can be used for the treatment of viral diseases. For example, a chimeric co-stimuiatory antigen receptor that recognizes a first CMV antigen and a chimeric antigen receptor that recognizes and binds a second CMV antigen are co-expressed in cytotoxic T lymphocytes for the treatment of CMV. Tumor antigen-specific T lymphocytes (and NK cells)
[0081] Types of tumor antigen-specific human lymphocytes that can be used in the methods of the invention include, without limitation, peripheral donor lymphocytes genetically modified to express chimeric antigen receptors (CARs) (Sadelain, M., et al. 2003 Nat Rev Cancer 3:35-45), peripheral donor
lymphocytes genetically modified to express a full-length tumor antigen- recognizing T cell receptor complex comprising the a and p heterodimer (Morgan, R.A., et al. 2006 Science 314:126-129), lymphocyte cultures derived from tumor infiltrating lymphocytes (TILs) in tumor biopsies (Panelii, M.C., et al. 2000 J Immunol 164:495-504; Panelii, M.C., et al. 2000 J Immunol1] 64:4382-4392), and selectively in vitro-expanded antigen-specific peripheral blood leukocytes employing artificial antigen-presenting cells (AAPCs) or pulsed dendritic cells (Dupont, J., et al. 2005 Cancer Res 65:5417-5427; Papanicolaou, G.A., et al. 2003 Blood 102:2498-2505). The T ceils may be autologous, allogeneic, or derived in vitro from engineered progenitor or stem cells.
[0082] Any suitable tumor antigen (antigenic peptide) is suitable for use in the tumor-related embodiments described herein. Sources of antigen include, but are not limited to cancer proteins. The antigen can be expressed as a peptide or as an intact protein or portion thereof. The intact protein or a portion thereof can be native or mutagenized. Suitable antigens include prostate specific membrane antigen (PSMA) and prostate stem cell antigen (PCSA).
Viral antigen-specific T lymphocytes (and NK cells)
[0083] Suitable antigens for use in the treatment of pathogen infection or other infectious disease, for example, in an immunocompromised subject include, without limitation, viral antigens present in Cytomegalovirus (CMV), Epstein Barr Virus (EBV), Human Immunodeficiency Virus (HIV), and influenza virus.
[0084] The unpurified source of CTLs may be any known in the art, such as the bone marrow, fetal, neonate or adult or other hematopoietic ceil source, e.g. , fetal liver, peripheral blood or umbilical cord blood. Various techniques can be employed to separate the cells. For instance, negative selection methods can remove non-CTLs initially. mAbs are particularly useful for identifying markers associated with particular cell lineages and/or stages of differentiation for both positive and negative selections.
[0085] A large proportion of terminally differentiated cells can be initially removed by a relatively crude separation. For example, magnetic bead separations can be used initially to remove large numbers of irrelevant ceils. Preferably, at least about 80%, usually at least 70% of the total hematopoietic cells will be removed prior to cell isolation.
[0086] Procedures for separation include, but are not limited to, density gradient centrifugation; resetting; coupling to particles that modify cell density; magnetic separation with antibody-coated magnetic beads; affinity
chromatography; cytotoxic agents joined to or used in conjunction with a mAb, including, but not limited to, complement and cytotoxins; and panning with antibody attached to a solid matrix, e.g. plate, chip, elutriation or any other convenient technique.
[0087] Techniques for separation and analysis include, but are not limited to, flow cytometry, which can have varying degrees of sophistication, e.g., a plurality of color channels, low angle and obtuse light scattering detecting channels, impedance channels.
[0088] The cells can be selected against dead cells, by employing dyes associated with dead cells such as propidium iodide (PI). Preferably, the ceils are coi!ected in a medium comprising 2% fetal calf serum (FCS) or 0.2% bovine serum albumin (BSA) or any other suitable, preferably sterile, isotonic medium.
[0089] Accordingly, the invention generally provides an immunoresponsive celi, such as a virus specific or tumor specific T cell comprising a receptor that binds a first antigen and activates the immunresponsive celi and a receptor that binds a second antigen and stimulates the immunresponsive cell. Vectors
[0090] Genetic modification of immunoresponsive ceils (e.g., T cells, CTL cells, NK cells) can be accomplished by transducing a substantially homogeneous cell composition with a recombinant DNA construct. Preferably, a retroviral vector (either gamma-retroviral or lentiviral) is employed for the introduction of the DNA construct into the ceil. For example, a polynucleotide encoding a receptor that binds an antigen (e.g., a tumor antigen, or a variant, or a fragment thereof), can be cloned into a retroviral vector and expression can be driven from its endogenous promoter, from the retroviral long terminal repeat, or from a promoter specific for a target cell type of interest. Non-viral vectors may be used as well.
[0091] For initial genetic modification of the ceils to provide tumor or viral antigen-specific cells, a retroviral vector is generally employed for transduction, however any other suitable viral vector or non-viral delivery system can be used. For subsequent genetic modification of the cells to provide ceils comprising an antigen presenting complex comprising at least two co- stimulatory ligands, retroviral gene transfer (transduction) likewise proves effective. Combinations of retroviral vector and an appropriate packaging line are also suitable, where the capsid proteins will be functional for infecting human cells. Various amphotropic virus-producing cell lines are known, including, but not limited to, PA12 (Miller, et ai. (1985) Mol. Cell. Biol. 5:431- 437); PA317 (Miller, et al. (1986) Mol. Cell. Biol. 6:2895-2902); and CRIP (Danos, et al. (1988) Proc. Natl. Acad. Sci. USA 85:6460-6464). Non- amphotropic particles are suitable too, e.g., particles pseudotyped with VSVG, RD114 or GALV envelope and any other known in the art.
[0092] Possible methods of transduction also include direct co-culture of the cells with producer cells, e.g., by the method of Bregni, et al. (1992) Blood 80:1418- 1422, or culturing with viral supernatant alone or concentrated vector stocks with or without appropriate growth factors and poiycations, e.g., by the method of Xu, et al. (1994) Exp. Hemat. 22:223-230; and Hughes, et al. (1992) J. Clin. Invest. 89:1817.
[0093] Other transducing viral vectors can be used to express a co-stimulatory ligand of the invention in an immunoresponsive ceil. Preferably, the chosen vector exhibits high efficiency of infection and stable integration and expression (see, e.g., Cayouette et ai., Human Gene Therapy 8:423-430, 1997; Kido et al., Current Eye Research 15:833-844, 1996; Bloomer et al., Journal of Virology 71 :6641-6649, 1997; Naldini et al., Science 272:263-267, 1996; and Miyoshi et al., Proc. Natl. Acad. Sci. U.S.A. 94:10319, 1997). Other viral vectors that can be used include, for example, adenoviral, lentiviral, and adeno-associated viral vectors, vaccinia virus, a bovine papilloma virus, or a herpes virus, such as Epstein-Barr Virus (also see, for example, the vectors of Miller, Human Gene Therapy 15-14, 1990; Friedman, Science 244:1275-1281 , 1989; Eglitis et al., BioTechniques 6:608-614, 1988;
Tolstoshev et al., Current Opinion in Biotechnology 1 :55-61 , 1990; Sharp, The Lancet 337:1277-1278, 1991 ; Cornetta et al., Nucleic Acid Research and Molecular Biology 36:31 1-322, 1987; Anderson, Science 226:401-409,1984; Moen, Blood Ceils 17:407-416, 1991 ; Miller et al., Biotechnology 7:980-990, 1989; Le Gal La Salle et al., Science 259:988-990, 1993; and Johnson, Chest 107:77S-83S, 1995). Retroviral vectors are particularly well developed and have been used in clinical settings (Rosenberg et al., N. Engl. J . Med 323:370, 1990; Anderson et al., U.S. Pat. No. 5,399,346).
[0094] Non-viral approaches can also be employed for the expression of a protein in ceil. For example, a nucleic acid molecule can be introduced into a cell by administering the nucleic acid in the presence of lipofection (Feigner et al., Proc. Natl. Acad. Sci. U.S.A. 84:741 3, 1987; Ono et a!., Neuroscience Letters 17:259, 1990; Brigham et al., Am. J. Med. Sci. 298:278, 1989;
Staubinger et al., Methods in Enzymology 101 :512, 1983), asialoorosomucoid- polyiysine conjugation (Wu et al., Journal of Biological Chemistry 263: 14621 , 1988; Wu et al., Journal of Biological Chemistry 264: 16985, 1989), or by micro-injection under surgical conditions (Wolff et al., Science 247:1465, 1990).
[0095] Other non-viral means for gene transfer include transfection in vitro using calcium phosphate, DEAE dextran, electroporation, and protoplast fusion. Liposomes can also be potentially beneficial for delivery of DNA into a ceil. Transplantation of normal genes into the affected tissues of a subject can also be accomplished by transferring a normal nucleic acid into a cuitivatable ceil type ex vivo (e.g., an autologous or heterologous primary cell or progeny thereof), after which the cell (or its descendants) are injected into a targeted tissue or are injected systemically. Recombinant receptors can also be derived or obtained using transposases or targeted nucleases (e.g. Zinc finger nucleases, meganucleases, or TALE nucleases). Transient expression may be obtained by RNA
eiectroporation.cDNA expression for use in polynucleotide therapy methods can be directed from any suitable promoter (e.g., the human cytomegalovirus (CMV), simian virus 40 (SV40), or metailothionein promoters), and regulated by any appropriate mammalian regulatory element or intron (e.g. the elongation factor 1c enhancer/promoter/intron structure). For example, if desired, enhancers known to preferentially direct gene expression in specific cell types can be used to direct the expression of a nucleic acid. The enhancers used can include, without limitation, those that are characterized as tissue- or cell-specific enhancers. Alternatively, if a genomic clone is used as a therapeutic construct, regulation can be mediated by the cognate regulatory sequences or, if desired, by regulatory sequences derived from a heterologous source, including any of the promoters or regulatory elements described above.
[0096] The resulting cells can then be grown under conditions similar to those for unmodified cells, whereby the modified ceils can be expanded and used for a variety of purposes.
[0097] Also included in the invention are 19z1 , CD19, CD8, CD3 , dsRed, P28BB, PSMA, CD28, 4-1 BB, GFP polypeptides or fragments thereof that are modified in ways that enhance their anti-neopiastic activity when expressed in an immunoresponsive cell. The invention provides methods for optimizing an amino acid sequence or nucleic acid sequence by producing an alteration in the sequence. Such alterations may include certain mutations, deletions, insertions, or post-translational modifications. The invention further includes analogs of any naturally-occurring polypeptide of the invention. Analogs can differ from a naturally-occurring polypeptide of the invention by amino acid sequence differences, by post-translational modifications, or by both. Analogs of the invention will generally exhibit at least 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identity with all or part of a naturally-occurring amino, acid sequence of the invention. The length of sequence comparison is at least 5, 10, 15 or 20 amino acid residues, preferably at least 25, 50, or 75 amino acid residues, and more preferably more than 100 amino acid residues. Again, in an exemplary approach to determining the degree of identity, a BLAST program may be used, with a probability score between e3 and e" 00 indicating a closely related sequence. Modifications include in vivo and in vitro chemical derivatization of polypeptides, e.g., acetylation, carboxyiation, phosphorylation, or giycosylation; such modifications may occur during polypeptide synthesis or processing or following treatment with isolated modifying enzymes. Analogs can also differ from the naturally-occurring polypeptides of the invention by alterations in primary sequence. These include genetic variants, both natural and induced (for example, resulting from random mutagenesis by irradiation or exposure to ethanemethylsuifate or by site-specific mutagenesis as described in Sambrook, Fritsch and Maniatis, Molecular Cloning: A Laboratory Manual (2d ed.), CSH Press, 1989, or Ausubel et al., supra). Also included are cyclized peptides, molecules, and analogs which contain residues other than L-amino acids, e.g., D-amino acids or non-naturally occurring or synthetic amino acids, e.g., - or - amino acids.
[0098] In addition to full-length polypeptides, the invention also provides fragments of any one of the polypeptides or peptide domains of the invention. As used herein, the term "a fragment" means at least 5, 10, 13, or 15 amino acids. In other embodiments a fragment is at least 20 contiguous amino acids, at least 30 contiguous amino acids, or at least 50 contiguous amino acids, and in other embodiments at least 60 to 80, 100, 200, 300 or more contiguous amino acids. Fragments of the invention can be generated by methods known to those skilled in the art or may result from normal protein processing (e.g., removal of amino acids from the nascent polypeptide that are not required for biological activity or removal of amino acids by alternative mRNA splicing or alternative protein processing events).
[0099] Non-protein analogs have a chemical structure designed to mimic the functional activity of a protein of the invention. Such analogs are administered according to methods of the invention. Such analogs may exceed the
physiological activity of the original polypeptide. Methods of analog design are well known in the art, and synthesis of analogs can be carried out according to such methods by modifying the chemical structures such that the resultant analogs increase the anti-neoplastic activity of the original polypeptide when expressed in an immunoresponsive ceil. These chemical modifications include, but are not limited to, substituting alternative R groups and varying the degree of saturation at specific carbon atoms of a reference polypeptide. Preferably, the protein analogs are relatively resistant to in vivo degradation, resulting in a more prolonged therapeutic effect upon administration. Assays for measuring functional activity include, but are not limited to, those described in the
Examples below.
Co-stimulatory ligands
[00100] The interaction with at least one co-stimulatory ligand provides a non- antigenspecific signal important for full activation of an immune ceil (e.g., T cell). Co- stimulatory ligands include, without limitation, tumor necrosis factor (TNF) ligands, cytokines (such as IL-2, IL-12, !L-15 or IL21 ), and immunoglobulin (ig) superfamily ligands.
[00101] Tumor necrosis factor (TNF) is a cytokine involved in systemic inflammation and stimulates the acute phase reaction, its primary role is in the regulation of immune cells. Tumor necrosis factor (TNF) ligands share a number of common features. The majority of the ligands are synthesized as type II transmembrane proteins (extracelluiarC-terminus) containing a short cytoplasmic segment and a relatively long extracellular region. TNF ligands include, without limitation, nerve growth factor (NGF), CD40L (CD40L)/CD154, CD137L/4- 1 BBL, tumor necrosis factor alpha (TNFa), CD134L/OX40L/CD252, CD27L/CD70, Fas ligand (FasL), CD30L/CD153, tumor necrosis factor beta (TNF(3)/iymphotoxin- aipha (LTa), lymphotoxin-beta (ur(3), CD257/B cell-activating factor
(BAFF)/Blys/THANK/Ta11-1 , glucocorticoid-induced TNF Receptor ligand (GITRL), and TNF-re!ated apoptosis-inducing ligand (TRAIL), LIGHT (TNFSF14). The immunoglobulin (Ig) superfamily is a large group of cell surface and soluble proteins that are involved in the recognition, binding, or adhesion processes of cells. These proteins share structural features with immunoglobulins - they possess an immunoglobulin domain (fold). Immunoglobulin superfamily ligands include, without limitation, CD80 and CD86, both ligands for CD28.
[00102] Compositions comprising genetically modified immunoresponsive ceils of the invention (e.g., T cells, NK cells, CTL cells, or their progenitors) can be provided systemically or directly to a subject for the treatment of a neoplasia, pathogen infection, or infectious disease. In one embodiment, ceils of the invention are directly injected into an organ of interest (e.g., an organ affected by a neoplasia). Alternatively, compositions comprising genetically modified immunoresponsive cells are provided indirectly to the organ of interest, for example, by administration into the circulatory system (e.g. , the tumor vasculature). Expansion and differentiation agents can be provided prior to, during or after administration of the cells to increase production of T cells, NK cells, or CTL cells in vitro or in vivo.
[00103] The modified cells can be administered in any physiologically acceptable vehicle, normaily intravascuiarly, although they may also be introduced into bone or other convenient site where the cells may find an appropriate site for regeneration and differentiation (e.g., thymus). Usuaily, at least 1x105 cells will be administered, eventually reaching 1x1010, or more. Genetically modified immunoresponsive cells of the invention can comprise a purified population of celis. Those skilled in the art can readily determine the percentage of genetically modified immunoresponsive cells in a population using various well-known methods, such as fluorescence activated cell sorting (FACS). Preferable ranges of purity in populations comprising genetically modified immunoresponsive ceils are about 50 to about 55%, about 55 to about 60%, and about 65 to about 70%. More preferably the purity is about 70 to about 75%, about 75 to about 80%, about 80 to about 85%; and still more preferably the purity is about 85 to about 90%, about 90 to about 95%, and about 95 to about 100%. Dosages can be readily adjusted by those skilled in the art (e.g., a decrease in purity may require an increase in dosage). The cells can be introduced by injection, catheter, or the like, if desired, factors can also be included, including, but not limited to, interleukins, e.g. IL-2, IL- 3, IL-6, and IL-1 1 , as well as the other interleukins, the colony stimulating factors, such as G-, M- and GM-CSF, interferons, e.g. .gamma. -interferon and erythropoietin.
[00104] Compositions of the invention include pharmaceutical compositions comprising genetically modified immunoresponsive celis or their progenitors and a pharmaceutically acceptable carrier. Administration can be autologous or heterologous. For example, immunoresponsive ceils, or progenitors can be obtained from one subject, and administered to the same subject or a different, compatible subject. Peripheral blood derived immunoresponsive cells of the invention or their progeny (e.g., in vivo, ex vivo or in vitro derived) can be administered via localized injection, including catheter administration, systemic injection, localized injection, intravenous injection, or parenteral administration. When administering a therapeutic composition of the present invention (e.g., a pharmaceutical composition containing a genetically modified immunoresponsive cell), it will generally be formulated in a unit dosage injectable form (solution, suspension, emulsion).
Formulations
[00105] Compositions of the invention comprising genetically modified immunoresponsive cells can be conveniently provided as sterile liquid preparations, e.g., isotonic aqueous solutions, suspensions, emulsions, dispersions, or viscous compositions, which may be buffered to a selected pH. Liquid preparations are normally easier to prepare than gels, other viscous compositions, and solid compositions. Additionally, liquid compositions are somewhat more convenient to administer, especially by injection. Viscous compositions, on the other hand, can be formulated within the appropriate viscosity range to provide longer contact periods with specific tissues. Liquid or viscous compositions can comprise carriers, which can be a solvent or dispersing medium containing, for example, water, saline, phosphate buffered saline, polyoi (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like) and suitable mixtures thereof.
[00106] Sterile injectable solutions can be prepared by incorporating the genetically modified immunoresponsive cells utilized in practicing the present invention in the required amount of the appropriate solvent with various amounts of the other ingredients, as desired. Such compositions may be in admixture with a suitable carrier, diluent, or excipient such as sterile water, physioiogical saline, glucose, dextrose, or the like. The compositions can also be lyophiiized. The compositions can contain auxiliary substances such as wetting, dispersing, or emulsifying agents (e.g., methylcelluiose), pH buffering agents, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired. Standard texts, such as "REMINGTON'S PHARMACEUTICAL SCIENCE", 17th edition, 1985, incorporated herein by reference, may be consulted to prepare suitable preparations, without undue experimentation.
[00107] Various additives which enhance the stability and sterility of the
compositions, including antimicrobial preservatives, antioxidants, chelating agents, and buffers, can be added. Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin. According to the present invention, however, any vehicle, diluent, or additive used would have to be compatible with the genetically modified
immunoresponsive ceils or their progenitors.
[00108] The compositions can be isotonic, i.e., they can have the same osmotic pressure as blood and lacrimal fluid. The desired isotonicity of the compositions of this invention may be accomplished using sodium chloride, or other
pharmaceutically acceptable agents such as dextrose, boric acid, sodium tartrate, propylene glycol or other inorganic or organic solutes. Sodium chloride is preferred particularly for buffers containing sodium ions.
[00109] Viscosity of the compositions, if desired, can be maintained at the selected level using a pharmaceutically acceptable thickening agent.
Methyiceiluiose is preferred because it is readily and economically available and is easy to work with. Other suitable thickening agents include, for example, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, carbomer, and the like. The preferred concentration of the thickener will depend upon the agent selected. The important point is to use an amount that will achieve the selected viscosity. Obviously, the choice of suitable carriers and other additives will depend on the exact route of administration and the nature of the particular dosage form, e.g., liquid dosage form (e.g., whether the composition is to be formulated into a solution, a suspension, gel or another liquid form, such as a time release form or liquid-filled form).
[00110] Those skilled in the art will recognize that the components of the compositions should be selected to be chemically inert and will not affect the viability or efficacy of the genetically modified immunoresponsive cells as described in the present invention. This will present no problem to those skilled in chemical and pharmaceutical principies,or problems can be readily avoided by reference to standard texts or by simple experiments (not involving undue experimentation), from this disclosure and the documents cited herein. One consideration concerning the therapeutic use of genetically modified immunoresponsive cells of the invention is the quantity of ceils necessary to achieve an optimal effect. The quantity of ceils to be administered will vary for the subject being treated. In a one embodiment, between 104 to 1010, between 105 to 109, or between 106 and 10s genetically modified immunoresponsive cells of the invention are administered to a human subject. More effective ceils may be administered in even smaller numbers. In some embodiments, at least about 1 x 108, 2 x 108, 3 x 108, 4 x 108, and 5 x 108 genetically modified immunoresponsive cells of the invention are administered to a human subject. The precise determination of what would be considered an effective dose may be based on factors individual to each subject, including their size, age, sex, weight, and condition of the particular subject. Dosages can be readily ascertained by those skilled in the art from this disclosure and the knowledge in the art.
[00111] The skilled artisan can readily determine the amount of cells and optional additives, vehicles, and/or carrier in compositions and to be administered in methods of the invention. Typically, any additives (in addition to the active ceil(s) and/or agent(s)) are present in an amount of 0.001 to 50 % (weight) solution in phosphate buffered saline, and the active ingredient is present in the order of micrograms to milligrams, such as about 0.0001 to about 5 wt %, preferably about 0.0001 to about 1 wt %, still more preferably about 0.0001 to about 0.05 wt % or about 0.001 to about 20 wt %, preferably about 0.01 to about 10 wt %, and still more preferably about 0.05 to about 5 wt %. Of course, for any composition to be administered to an animal or human, and for any particular method of
administration, it is preferred to determine therefore: toxicity, such as by determining the lethal dose (LD) and LD50 in a suitable animal model e.g., rodent such as mouse; and, the dosage of the composition(s), concentration of components therein and timing of administering the composition(s), which elicit a suitable response. Such determinations do not require undue experimentation from the knowledge of the skilled artisan, this disclosure and the documents cited herein. And, the time for sequential administrations can be ascertained without undue experimentation.
Methods of Treatment
[00112] Provided herein are methods for treating neoplasia in a subject. Also contemplated herein are methods for treating a pathogen infection or other infectious disease in a subject, such as an immunocompromised human subject. The methods comprise administering a T cell, NK cell, or CTL cell of the invention in an amount effective to achieve the desired effect, be it palliation of an existing condition or prevention of recurrence. For treatment, the amount administered is an amount effective in producing the desired effect. An effective amount can be provided in one or a series of administrations. An effective amount can be provided in a bolus or by continuous perfusion.
[00113] An "effective amount" (or, "therapeutically effective amount") is an amount sufficient to effect a beneficial or desired clinicai result upon treatment. An effective amount can be administered to a subject in one or more doses. In terms of treatment, an effective amount is an amount that is sufficient to palliate, ameliorate, stabilize, reverse or slow the progression of the disease, or otherwise reduce the pathological consequences of the disease. The effective amount is generally determined by the physician on a case-by-case basis and is within the skill of one in the art. Several factors are typically taken into account when
determining an appropriate dosage to achieve an effective amount. These factors include age, sex and weight of the subject, the condition being treated, the severity of the condition and the form and effective concentration of the antigen-binding fragment administered.
[00114] For adoptive immunotherapy using antigen-specific T cells, ceil doses in the range of 106— 1010 (e.g., 109) are typically infused. Upon administration of the genetically modified cells into the host and subsequent differentiation, T cells are induced that are specifically directed against the specific antigen. "Induction" of T cells can include inactivation of antigen-specific T ceils such as by deletion or anergy. inactivation is particularly useful to establish or reestablish tolerance such as in autoimmune disorders. The modified cells can be administered by any method known in the art including, but not limited to, intravenous, subcutaneous, intranodal, intratumorai, intrathecal, intrapleural, intraperitoneal and directly to the thymus.
[00115] The invention provides methods for increasing an immune response in a subject in need thereof. In one embodiment, the invention provides methods for treating or preventing a neoplasia in a subject. The invention provides therapies that are particularly useful for the treatment of subjects having prostate cancer, or metastatic prostate cancer that is not amenable to conventional therapeutic interventions. Suitabiehuman subjects for therapy typically comprise two treatment groups that can be distinguished by clinical criteria. Subjects with "advanced disease" or "high tumor burden" are those who bear a clinically measurable tumor. A clinically measurable tumor is one that can be detected on the basis of tumor mass (e.g., by palpation, CAT scan, sonogram, mammogram or X-ray; positive biochemical or histopathologic markers on their own are insufficient to identify this population). A
pharmaceutical composition embodied in this invention is administered to these subjects to elicit an anti-tumor response, with the objective of palliating their condition. Ideally, reduction in tumor mass occurs as a result, but any clinical improvement constitutes a benefit. Clinical improvement includes decreased risk or rate of progression or reduction in pathological
consequences of the tumor.
[00116] A second group of suitable subjects is known in the art as the
"adjuvant group." These are individuals who have had a history of neoplasia, but have been responsive to another mode of therapy. The prior therapy can have included, but is not restricted to, surgical resection, radiotherapy, and traditional chemotherapy. As a result, these individuals have no clinically measurable tumor. However, they are suspected of being at risk for
progression of the disease, either near the original tumor site, or by metastases. This group can be further subdivided into high-risk and iow-risk individuals. The subdivision is made on the basis of features observed before or after the initial treatment. These features are known in the clinical arts, and are suitably defined for each different neoplasia. Features typical of high-risk subgroups are those in which the tumor has invaded neighboring tissues, or who show involvement of lymph nodes.
[00117] Another group have a genetic predisposition to neoplasia but have not yet evidenced clinical signs of neoplasia. For instance, women testing positive for a genetic mutation associated with breast cancer, but still of childbearing age, can wish to receive one or more of the antigen-binding fragments described herein in treatment prophylacticaliy to prevent the occurrence of neoplasia until it is suitable to perform preventive surgery.
[00118] Human neoplasia subjects having any of the following neoplasias:
glioblastoma, melanoma, neuroblastoma, adenocarcinoma, glioma, soft tissue sarcoma, and various carcinomas (including prostate and small cell lung cancer) are especially appropriate subjects. Suitable carcinomas further include any known in the field of oncology, including, but not limited to, astrocytoma,
fibrosarcoma, myxosarcoma, liposarcoma, oligodendroglioma, ependymoma, medullobiastoma, primitive neural ectodermal tumor (PNET), chondrosarcoma, osteogenic sarcoma, pancreatic ductal adenocarcinoma, small and large cell lung adenocarcinomas, chordoma, angiosarcoma, endotheliosarcoma, squamous cell carcinoma, bronchoalveolarcarcinoma, epithelial adenocarcinoma, and liver metastases thereof, lymphangiosarcoma, iymphangioendotheliosarcoma, hepatoma, cholangiocarcinoma, synovioma, mesothelioma, Ewing's tumor, rhabdomyosarcoma, colon carcinoma, basal cell carcinoma, sweat gland carcinoma, papillary carcinoma, sebaceous gland carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, testicular tumor, medullobiastoma,
craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, neuroblastoma, retinoblastoma, leukemia, multiple myeloma, Waldenstrom's macroglobuiinemia, and heavy chain disease, breast tumors such as ductal and lobular adenocarcinoma, squamous and adenocarcinomas of the uterine cervix, uterine and ovarian epithelial carcinomas, prostatic adenocarcinomas, transitional squamous cell carcinoma of the bladder, B and T cell lymphomas (nodular and diffuse) plasmacytoma, acute and chronic leukemias, malignant melanoma, soft tissue sarcomas and leiomyosarcomas.
[00119] The subjects can have an advanced form of disease, in which case the treatment objective can include mitigation or reversal of disease progression, and/or amelioration of side effects. The subjects can have a history of the condition, for which they have already been treated, in which case the therapeutic objective will typically include a decrease or delay in the risk of recurrence.
[00120] Accordingly, the invention provides a method of treating or preventing a neoplasia in a subject, the method comprising administering an effective amount of an immunoresponsive cell comprising a receptor that binds a tumor antigen and activates the immunoresponsive ceil (e.g., TCR, CAR) and a vector encoding a receptor that binds another tumor antigen and stimulates the immunoresponsive cell, in one embodiment, the neoplasia is selected from the group consisting of prostate cancer, breast cancer, blood cancers (e.g.
leukemias, lymphomas, and myelomas), ovarian cancer, bladder cancer, brain cancer, colon cancer, intestinal cancer, liver cancer, lung cancer, pancreatic cancer, prostate cancer, skin cancer, stomach cancer, glioblastoma, and throat cancer. In another embodiment, the tumor antigen is one or more of carbonic anhydrase IX (CAIX), carcinoembryonic antigen (CEA), CD5, CD7, CD10, CD19, CD20, CD22, CD30, CD33, CD34, CD38, CD41 , CD44, CD49f, CD56, CD74, CD133, CD138, an antigen of a cytomegalovirus (CMV) infected cell (e.g., a cell surface antigen), epithelial giycoprotein-2 (EGP-2), epithelial glycoprotein-40 (EGP-40), epithelial cell adhesion molecule (EpCAM), receptor tyrosine-protein kinases erb-B2,3,4, foiate-binding protein (FBP), fetal acetylcholine receptor (AChR), folate receptor-a, Gangiioside G2 (GD2), Gangiioside G3 (GD3), human Epidermal Growth Factor Receptor 2 (HER-2), human telomerase reverse transcriptase (hTERT), lnterleukin-13 receptor subunit alpha-2 (IL- 13Ra2), ic-light chain, kinase insert domain receptor (KDR), Lewis Y (LeY), LI cell adhesion molecule (L1 CAM), melanoma antigen family A, 1 (MAGE-A1 ), Mucin 1 (MUC1 ), Mesothelin (MSLN), NKG2D ligands, cancer-testis antigen NY-ES0-1 , oncofetal antigen (h5T4), prostate stem cell antigen (PSCA), prostate-specific membrane antigen (PSMA), tumor-associated glycoprotein 72 (TAG-72), vascular endothelial growth factor R2 (VEGF-R2), or Wilms tumor protein (WT-1 ).
[00121] As a consequence of surface expression of a receptor that binds a tumor antigen and activates the immunoresponsive ceil (e.g., TCR, CAR) and a vector encoding a receptor that binds another tumor antigen and stimulates the immunoresponsive cell (e.g. CCR), adoptively transferred human T or NK cells are endowed with augmented and selective cytolytic activity at the tumor site. Furthermore, subsequent to their localization to tumor or viral infection and their proliferation, co-stimulatory ligandexpressing T ceils turn the tumor or viral infection site into a highly conductive environment for a wide range of immune ceils involved in the physiological anti-tumor or antiviral response (tumor infiltrating lymphocytes, NK-, NKT- cells, dendritic cells, and macrophages).
[00122] In other embodiments, the invention provides methods for treating subjects with a pathogen infection (e.g., viral infection, bacterial infection, fungal infection, parasite infection, or protozoa! infection). The invention is particularly useful for enhancing an immune response in an
immunocompromised subject. Exemplary viral infections susceptible to treatment using a method of the invention include, but are not limited to, Cytomegalovirus (CMV), Epstein Barr Virus (EBV), Human Immunodeficiency Virus (HIV), and influenza virus infections.
[00123] Accordingly,the invention provides a method of treating or preventing a pathogen infection in a subject, the method comprising administering an effective amount of an immunoresponsive cell as described herein.
Kits
[00124] The invention provides kits for the treatment or prevention of a neoplasia, pathogen infection, immune disorder or allogeneic transplant. In one embodiment, the kit includes a therapeutic or prophylactic composition containing an effective amount of an immunoresponsive cell comprising an activating antigen receptor and a co-stimulatory antigen receptor in unit dosage form. In particular embodiments, the ceiis further comprise a co-stimulatory ligand. In some embodiments, the kit comprises a sterile container which contains a
therapeutic or prophylactic vaccine; such containers can be boxes, ampules, bottles, vials, tubes, bags, pouches, blister-packs, or other suitable container forms known in the art. Such containers can be made of plastic, glass, laminated paper, metal foil, or other materials suitable for holding
medicaments.
[00125] If desired the immunoresponsive cell is provided together with instructions for administering the ceil to a subject having or at risk of developing a neoplasia, pathogen infection, immune disorder or allogeneic transplant. The instructions will generally include information about the use of the composition for the treatment or prevention of neoplasia, pathogen infection, immune disorder or allogeneic transplant. In other embodiments, the instructions include at least one of the foliowing: description of the therapeutic agent; dosage schedule and administration for treatment or prevention of a neoplasia, pathogen infection, immune disorder or allogeneic transplant or symptoms thereof; precautions; warnings; indications; counter-indications; overdosage information; adverse reactions; animal pharmacology; clinical studies; and/or references. The instructions may be printed directly on the container (when present), or as a label applied to the container, or as a separate sheet, pamphlet, card, or folder supplied in or with the container.
EXAMPLES
[00126] The practice of the present invention employs, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are weli within the purview of the skilled artisan. Such techniques are explained fully in the literature, such as, "Molecular Cloning: A Laboratory Manual", second edition (Sambrook, 1989); "Oligonucleotide Synthesis" (Gait, 1984); "Animal Cell Culture" (Freshney, 1987); "Methods in Enzymology" "Handbook of Experimental
Immunology" (Weir, 1996); "Gene Transfer Vectors for Mammalian Cells" (Miller and Caios, 1987); "Current Protocols in Molecular Biology" (Ausubel, 1987); "PCR: The Polymerase Chain Reaction", (Muiiis, 1994); "Current Protocols in Immunology" (Coligan, 1991 ). These techniques are applicable to the production of the polynucleotides and polypeptides of the invention, and, as such, may be considered in making and practicing the invention. Particularly useful techniques for particular embodiments will be discussed in the sections that follow.
[00127] The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the assay, screening, and therapeutic methods of the invention, and are not intended to limit the scope of what the inventors regard as their invention.
Example 1. T cells co-expressing a chimeric antigen receptor (CAR) and a chimeric co-stimulating receptor (CCR) eradicated established tumors.
[00128] The invention provides "tumor-sensing T ceils" that simultaneously engage two antigens co-expressed by a tumor ceil. Importantly, it has been found that the reactivity against tissues expressing either antigen alone should be negligible, only unleashing T cell activation in the presence of both antigens but not either one alone. The invention is at least based in part on the discoveries that in combination provide selective T ceil immunoreactivity, and, thus, make this approach clinically relevant. The first is to assign T cell activation to one antigen (e.g., CD19 or prostate stem cell antigen, PSCA), which may be mediated by a T cell receptor (TCR) or a chimeric antigen receptor (CAR). Costimulation is independently mediated by a "chimeric costimuiatory receptor" (CCR ), 2,13 which is targeted to a second antigen (e.g., prostate-specific membrane antigen, PSMA). This approach resulted in increased
immunoreactivity against dual antigen positive (DP) tumors, but failed to avert enhanced immunoreactivity against single antigen positive (SP) tumors. The second principle important for tumor sensing T ceils to differentiate DP tumors from SP tumors, is to diminish T cell activation to a level where it is by itself ineffective, but functionally rescued at the tumor site by a CCR engaged by an independent, co-expressed antigen. As CARs and CCRs recognize cell surface antigens rather than HLA-peptide complexes, T cells engineered in this manner are directly targeted to the tumor and will not be costimulated by interacting with ceils cross-presenting the targeted antigens. As demonstrated herein, this approach resulted in selective tumor eradication in multiple tumor-bearing mice.
[00129] To demonstrate that both T ceil activation and costimulation signals can be supplied in vivo using two distinct antigen-specific receptors, the combination of a CAR, providing a CD3 activation signal upon recognition of the B cell marker CD1914 and a CCR specific for PSMA 2' 5 was evaluated. Because of the synergy between CD28 and 4-1 BB 6' 7, inciuding tandem cytoplasmic domains18"21 , 4-1 BB cytoplasmic domain was added to the PSMA CCR P2815 as described2' (Figure 1 A). Primary human peripheral blood T cells were transduced with the 19z1 and/or P28BB receptors and showed readily detectable expression of both receptors, with transduction efficiencies in the range of 4570% (Figure 1 B). Four groups of T ceils were analyzed in all subsequent studies, comprising anti-CD19 CAR (19z1 ), anti-PSMA CCR (P28BB), both anti-CD19 CAR and anti-PSMA CCR in combination
(19z1 +P28BB), and a mock-transduced control group (mock) (Figure 1 C). The in vitro cytotoxic and proliferative response upon exposure to CD19 and/or PSMA showed that cytotoxicity directed against CD19 was, as expected, imparted by 19z1 and unaltered in the presence of PSMA.
[00130] A quantitative comparison of the T cell groups, normalized to the fraction of 19z1 -transduced T cells for the 19z1 and 19z1 +P28BB groups and the P28BBtransduced fraction in the P28BB group showed that 19z1 and 19z1 +P28BB T ceils specificaiiy iysed 40-47% CD19+ targets at the 50: 1 E:T ratio while the P28BB-transduced T cells failed to lyse PSMA+ targets (Figure 2A). However, upon repeated exposure to these antigens in the absence of exogenous cyiokine, only the 19z1 +P28BB T celis exhibited robust proliferation with a 58-fold expansion for 31 days when co-cultured on artificial antigen presenting ceils (AAPCs) that expressed both antigens. By comparison, 19z1 or P28BB T celis only displayed modest expansion over the first 14 days, as did the 19z1 +P28BB T ceils on CD19+PSMA" APCs (Figure 2B). Further evidence of stronger T ceil activation in the presence of both antigens was provided by the quantitative assessment of cytokine production and the induction of the antiapoptotic molecule BclxL in 19z1 +P28BB T cells, which were distinctiy greater in the presence of CD19+PSMA+ APCs than in the presence of either antigen alone (Figures 4A, 4E).
[00131] Initially, the in vivo ability of these duai-receptor expressing T ceils to eradicate established systemic human prostate tumors in immunocompromised NOD/SCID-yC KO (NSG) mice bearing dual-positive (CD19 +PSMA +) tumor cells was tested. The NSG mice were systemicaily engrafted with 2.0 x 106 firefly-!uciferase expressing PC3 tumor cells that expressed both CD19 and PSMA (Figure 5) and treated 19 days later with a single intravenous infusion of 1 .0 x 106 19z1 , 19z1 +P28BB, P28BB or control T cells. Thirty-five days later, mice that received P28BB T cells or control T cells were sacrificed due to tumor burden. In contrast, mice treated with 19z1 T cells had a marked reduction of tumor burden. Strikingly, mice treated with 19z1 +P28BB T cells had undetectable tumor burden (Figure 2C). Over 70 days of post-infusion monitoring, the CD19+ tumors eventually relapsed in mice that received 19z1 T cells, while complete remission persisted in ail mice that received 19z1 +P28BB T cells (Figure 2C). This result strongly indicated that tumor eradication had been achieved.
[00132] These findings however posed a concern because of the potential base line eradication of CD19+PSMA" tumors by 19z1 +P28BB T cells. There was a likelihood that T ceil immunoreactivity would be undesirably enhanced in recipients bearing dual-positive CD19+PSMA+ tumors due to recirculation of T cells. To test this hypothesis, mice were subcutaneously infused with
CD19+PSMA" tumors into the left flanks, CD19" PSMA+ tumors into the right flanks, and CD19+PSMA+ tumors into their backs. One week later, mice were administered one of 19z1 , P28BB, or 19z1 +P28BB T cells (1 .0 x 106 cells) intravenously. Mice that received P28BB T cells had progression of all three tumors and needed to be sacrificed within 35 days (Figure 2D). In mice treated with 19z1 T cells, the CD19+PSMA~ and CD19+PSMA+ tumors underwent a substantial reduction compared to their progression in recipients of P28BB T cells, before eventually progressing. Consistent with prior results, mice treated with 19z1 +P28BB T cells showed complete eradication of CD19+PSMA+ tumors.
However, as hypothesized, rejection of CD19+PSMA" tumors was also substantially enhanced and superior to that observed in recipients of 19z1 T cells (Figure 2D, lower panels). Thus, a split signal approach targeting two antigens failed to restrict T cell reactivity and to protect single antigen tumors.
[00133] To address the problem of single antigen reactivity, it was proposed that T cell activation would have to be minimized, almost to the point of extinction, only to be rescued at the site of dual antigen expressing by an adequate CCR engagement. Thus, CARs with diminished activity were sought. For these experiments, a clinically relevant combination of antigens targeting PSCA and PSMA were used. Three PSCA-specific scFvs were evaluated with different binding affinities for PSCA (Figure 3A). While the Hzi scFv efficiently lysed tumor cells down to the picogram range, the Lzl scFv required 1 ,000— 10,000 fold more antibody to achieve similar efficiency of specific lysis. These scFvs were used to derive three CD3 -based CARs with different activities in cytotoxicity assays (Figure 3B). Two of the CARs, Hzl and Mzi, directed moderate lytic activity against PSCA+ targets (20% specific iysis at the 50: 1 E:T ratio). In contrast the third CAR, Lzi, only reached 10%, qualifying it as a inefficient antigen receptor. This hierarchy was further confirmed in cytokine release assays, which showed enhanced cytokine secretion by 19z1 +P28BB T cells (Figure 4A) and Hz1+P28BB T cells (Figure 4B) compared to ceils with either receptor alone. This
enhancement was less in Mzl+P28BB T cells (Figure 4C) and even further decreased in Lz1+P28BB T cells (Figure 4D).
[00134] In order to evaluate the therapeutic efficacy and targeting profile of PSCA+PSMA- reactive T cells, the anti-tumor activity of these T cells was tested in animals bearing PSCA+PSMA- and/or PSCA+PSMA+ tumors. First, to test the ability of Mzl+P28BB and Lz1 +P28BB T ceils to eradicate
PSCA+PSMA+ ceils selectively, mice were inoculated intravenously with 2 x 106 FFLuc-expressing PC3 cells positive for PSMA, PSCA, or both (Figure 5). Fourteen days later, one set of mice received 1 x 106 Mzl+P28BB CAR+ T cells infused intravenously, and another set of received 1 x 106 Lz1 +P28BB CARP T cells. Mice bearing PSCA+PSMA tumor cells that were treated with the more efficient Mzl+P28BB T cells exhibited greater tumor regression than mice treated with Lz1 +PBB T cells (Figure 3C). Similar to the CD19 experiment (Figure 2C), these tumors eventually relapsed and progressed. However, in mice bearing PSCA+PSMA+ tumor celis Mzl+P28BB T ceils induced robust and long-term tumor eradication. Consistent with the lesser potency of Lzl+P28BB T ceils, tumor eradication in mice bearing PSCA+PSMA+ tumor cells treated with Lz1 +P28BB T cells was slower but nonetheless equally successful, resulting in strong tumor eradication and long-term survival of ail treated mice (Figure 3C). Tumor eradication was not enhanced in control mice bearing either PSCAVSMA- or PSCA-PSMA+ tumors (Figure 3C). A more stringent evaluation of background activity against PSCAVSMA.- tumors was tested in the context of animals also bearing PSCA+PSMA+ and PSCAPSMA+ tumors. Lz1 +P28BB T cells mediated eradication of PSCA+PSMA+ tumors without increasing eradication of PSCA+PSMA tumors (Figure 3E), which was not different from that induced by Lzl T celis.
[00135] Thus, these results demonstrate the feasibility of decreasing T cell activation to the point of averting immune reactivity against tissues expressing one targeted antigen and rescuing T celi activation at the tumor site where two antigens are co-expressed, without running the risk of igniting reactivity against the single antigen-expressing tissues, in doing so, the results demonstrate proof-of- principle for achieving two complementary outcomes that determine specificity and safety: 1 ) the ability to create targeting specificity in the absence of a unique target antigen through combinatorial antigen recognition; and 2) the protection of cells expressing only one of the antigens by titrating activation and costimuiatory signals, so as to practically confine activation to sites of target antigen coexpression.
[00136] Targeted T ceil therapies have the potential to provide curative treatments but their applicability is limited by the paucity of validated tumor-specific targets. Extra- tumoral expression results indeed in "on-target, off-tumor" effects that 2-4" may be sometimes tolerable but are eventually lethal 11. The method described herein provides improved targeting by supplying titrated activation and
costimuiation signals through combinatorial antigen recognition (Figures 6A-6C).
[00137] In physiological antigen presentation,22 T ceils are primed in lymph nodes by receiving activating and costimuiatory signals and migrate to peripheral sites, where effector functions of the T cells are not as dependent on costimuiation. Simiiarly, T cells engaged through an antigen receptor and a CCR may recirculate to other peripheral sites and display heightened cytolytic activity against tissues expressing only one of the targeted antigens (Figure 6A). Therefore, the present strategy was developed to address this problem of a potential systemic effect in order to spare cells singly positive for the antigen, including non-tumor cells (Figure 6B). In a tri-tumor mouse model (PSCA+, PSMA\ and PSCA+PSMA+), eradication of PSCAVSMA" was accomplished, while sparing the PSCA+PSMA" and PSCA"PSMA+ tumors (Figure 3E).
[00138] As shown by the present studies, this selectivity for DP tumors can be achieved by reducing the efficacy of the CAR, which creates cells that are less cytotoxic (Figure 3B) and that have reduced levels of cytokine secretion (Figures 4A-4D). While the levels of both TH I and TH2 cytokines are relatively high for both 19z1 and Hzi CARs, using less efficient CARs Mzl and Lzl resulted in reducing these levels. The enhancement of cytokine levels in Lzl+P28BB T cells compared to Lzl T cells was minimal except for IL-2 and IL-13. While IL-2 induces proliferation and can promote either a TH 1 or TH2 response23, IL-13 is associated with a TH2 response specific to 4- 1 BB/CD1 37 signaling.24,25
[00139] PSCA and PSMA are promising targets for the treatment of metastatic prostate cancer 26'27, although neither is absolutely prostate-specific. In human subjects, PSCA expression is found in prostate cancer and within the renal pelvis, ureter, urinary bladder, and urethra.28 Expression of PSMA strongly correlates with primary prostate cancer, metastases, as well as in astrocytes type II, the kidney proximal tubule and the intestinal brush border.29 Dual PSCA/PSMA targeting is thus expected to increase prostate cancer targeting and reduce reactivity against these normal tissues. It is appreciated that this principle can be extended to other tumor types which express a pair of antigens, especially those that confer true tumor-specificity. For example, HER2, MUC1 , CD44, CD49f, and/or EpCAM could be used in this manner to treat breast cancer.3"1 Likewise, mesothelin, folate receptor-a, CD44, and/or CD133 could be used to treat ovarian cancer.32' 33 The targeting of tumor initiating cells or cancer stem cells, for which unique target antigens/structures have not yet been clearly identified34.3s would be particularly attractive using this approach. [00140] An important aspect of this approach is to constrain and nearly abolish T cell activation in response to a single antigen. CARs with low affinity or low avidity that only provide a poor activation signal were found to be usefui for achieving this effect. Alternatively, an endogenous TCR with low affinity or low avidity may be used in combination with a CCR to provide antigen-specific costimulation. Altogether, the results indicate the advantages of restricting the activity of engineered T cells, reconciling potency with safety through combinatorial antigen recognition by tumor-sensing T cells.
Gammaretroviral vector construction and viral production
[00141] The gammaretroviral vector SFG-19z1 has been extensively
described.14 This backbone construct was used to exchange scFvs to generate SFG-Hzl, SFG-Mzl, and SFG-Lzl by directional cloning utilizing a Ncol site located 5' of the scFv and a Nod site located 3' of the scFv. To generate SFG- P28BB, the fused CD28 and CD137 domains were PCR amplified from SFG- P28BBz1 and iigated 3' of the PSMA scFv using a 5' Ncol site and a 3' BamHI site to include a stop codon 3' of the BB domain, while the CD3 domain was removed.21 Bicistronic gene expression for CARs to be coexpressed with dsRED and CCRs to be coexpressed with hrGFP was achieved by using an Internal Ribosomal Entry Site as previously described. Vectors were used to transiently transfect cell lines to generate stable viral producing lines as previously described.
Generation of anti-PSCA scFvs
[00142] Three novel PSCA specific scFvs, termed Hzl, Mzl, and Lzl were generated byampiifying the variabie heavy (VH) and variabie light (VL) domains conferring PSCA antigen specificity of non-overlapping epitopes using degenerate primers from hybridomas as previously described.36 These VH and V|_ domains were fused together using a linker and were used to replace the CD19 scFv in the SFG-19z1 backbone using 5'Sphl to 3'Notl sites. Isolation, retroviral transduction, and culture of primary human T cells
[00143] Peripherai blood leukocytes were isolated using Ficoli gradients and transduced as previously described. Briefly, after 48-hour activation with 2 &g/ml_ phytohemagglutinin, cells were transduced twice via spinocuiation for 1 hour on retronectin coated plates over the next 48 hours and 20 U/mL of lL-2 was added. After allowing 3 days for vector expression, transduction efficiencies were determined via flow cytometry and bulk unsorted ceils were used for various assays or adoptive transfers.
Generation of antigen expressing tumor cell lines
[00144] The PC3 human prostate tumor line was obtained from ATCC and retrovirally16 transduced in order to generate PC3 -GFP/Luc, which was subsequently used to create 10 PC3-CD19, PC3-PSMA, PC3-CD19-PSMA, PC3- PSCA, and PC3-PSCA-PSMA via retroviral transduction.
CTL Chromium Release Killing Assays
[00145] Target cells expressing desired antigen were labeled with 5 Cr and co- cultured with T cells at decreasing effector : target ratio's. After 4 hours of culture, supernatant was removed and used to measure radioactivity released from chromium. Specific lysis was determined by subtracting background radioactivity of target celis not cultured with T cells and dividing by the radioactivity measured from target cells completely Iysed by using 0.2% Triton X-100.
Long-term T cell proliferation assays
[00146] Tumor celis expressing desired antigen were irradiated with 30 Gy prior to co-culture with 1.0 x 106 T cells at a 5:1 effector : target ratio. T cells were counted weekly using an Invitrogen Countess cell counter and then re-stimuiated with irradiated tumor celis. No exogenous cytokines were added to these co- cultures.
Generation of tumor models in mice
[00147] PC3 tumor cells were infused into NOD/SCID-IL2Ry mice obtained from either Jackson Laboratories or from in-house breeding under the protocol 04-10- 024 approved by the MSKCC Institutional Animal Care and Use Committee. For systemic tumor experiments, 2.0 x 106 tumor infused into mice with 1.0 x 106 chimeric receptor positive T ceils infused 14 days later. For subcutaneous tumor experiments, 1.0 x 106 tumor cells were injected per tumor site, established for 7 days upon which 1.0 x 106 chimeric positive T ceils were infused IV.
Quantification of tumor burden
[00148] For systemic tumor experiments, bioiiluminescent imaging (BLI) was used to
quantitativeiy measure tumor burden by correlating the amount of tumor burden to luminescence using an S 100 system (Caliper Life Sciences) as previously described. For subcutaneous tumors, calipers were used to measure tumor size. Tumor volume was calculated by multiplying the length, width, and height of each tumor.
Bispecific antibody mediated tumor lysis
[00149] Bispecific antibodies containing a PSCA specific scFv fused to a CD3 specific scFv were added at various amounts to untransduced T cells co-cultured with PSCA+ PC3 at a 20:1 ratio, respectively in standard 4hr chromium release assay assays.
Flow Cytometry
[00150] Cells were analyzed using an LSRII flow cytometer or sorted using a FACSAria cell sorter (BD Biosciences) as previously described.16 Detection of chimeric receptor at the cell surface could be achieved directly by using AF647 conjugated goat-anti-mouse antibody (invitrogen). Antibodies for CD4-PE-Cy7, CD8-Pacific Blue, and CD19-APC were obtained from Invitrogen while PSCA antibodies were purified from hybridoma supernatants and PSMA antibodies were obtained from MBL Interantional.
Cytokine Analysis
[00151] Supernatants harvested 48 hours after the second tumor stimulation from long- term T cell proliferation experiments and were used for cytokine analysis by using a custom multiplex system HCYTMAG-60K (Miilipore) and analyzed using a Luminex 100 instrument (Luminex) as previously described.21
Western Blot Analysis
[00152] Cells were harvested 24 hours after initial tumor stimulation from long-term T cell proliferation experiments to be used for western blot analysis of BclxL expression. Western blots were performed as previously described21 using BclxL and Akt primary antibodies (Ceil Signaling Technology).
[00153] From the foregoing description, it will be apparent that variations and modifications may be made to the invention described herein to adopt it to various usages and conditions. Such embodiments are also within the scope of the following claims.
[00154] The recitation of a listing of elements in any definition of a variable herein includes definitions of that variable as any single element or
combination (or subcombination) of listed elements. The recitation of an embodiment herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof.
[00155] This application may be related to U.S. Patent Application No.
12/593,751 , which is the U.S. national phase application, pursuant to 35 U.S.C. §371 , of International Patent Application No.: PCT/US2008/004251 , filed March 8, 2010, which claims the benefit of U.S. Provisional Application Ser. No.
60/921 , 144, filed March 30, 2007, the disclosures of which are hereby incorporated herein in their entireties by reference.
References
1. Robbins, P.F. et ai. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. Journal of clinical oncology: official journai of the American Society of Clinical Oncology 29, 917-924 (201 1 ).
2. Kaios, M. et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med 3, 95ra73 (201 1 ).
3. Brentjens, R.J. et al. Safety and persistence of adoptively transferred autologous CD19- ta rgeted T ceils in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 1 18, 4817-4828 (2011 ).
4 . Kochenderfer, J.N. et al. B-cell depletion and remissions of malignancy along with
cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen- receptor- transduced T cells. Blood 1 19, 2709-2720 (2012).
5. Sadelain, M., Riviere, I. & Brentjens, R. Targeting tumours with genetically enhanced T lymphocytes. Nat Rev Cancer 3, 35-45 (2003).
6. Ho, W.Y., Biattman, J.N., Dossett, M.L., Yee, C. & Greenberg, P.D. Adoptive immunotherapy: engineering T cell responses as biologic weapons for tumor mass destruction.
Cancer cell 3, 431-437 (2003).
7. Rosenberg, S.A., Restifo, N.P., Yang, J.C., Morgan, R.A. & Dudley, M.E. Adoptive cell transfer: a clinicai path to effective cancer immunotherapy. Nature reviews. Cancer 8, 299-308 (2008) .
8. Sadelain, M., Brentjens, R. & Riviere, I. The promise and potential pitfalls of chimeric
antigen receptors. Curr Opin Immunol 21 , 215-223 (2009). 9. Jorritsma, A., Schotte, R., Coccoris, M., de Witte, M.A. & Schumacher, T.N. Prospects and iimitations of T celi receptor gene therapy. Current gene therapy 1 1 , 276-287 (2011 ) .
10. Johnson, L.A. et al. Gene therapy with human and mouse T, cell receptors mediates
cancer regression and targets normal tissues expressing cognate antigen. Blood 114, 535-546
(2009) .
1 1. Morgan, R.A. et ai. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor
recognizing ERBB2. Molecular therapy: the journal of the American Society of Gene Therapy 18, 843-851 (2010).
1 2 .Krause, A. et al. Antigen-dependent CD28 signaling selectively enhances survival and
proliferation in genetically modified activated human primary T lymphocytes. J Exp Med 188, 619-626 (1998).
13. Wilkie, S. et al. Dual Targeting of ErbB2 and MUC1 in Breast Cancer Using Chimeric Antigen Receptors Engineered to Provide Complementary Signaling. Journal of clinical
immunology (2012).
14. Brentjens, R.J . et al. Eradication of systemic B-ceil tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nature medicine 9, 279-286 (2003).
is. Maher, J., Brentjens, R.J., Gunset, G., Riviere, I. & Sadelain, M. Human T- lymphocyte
cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor. Nat Bio
technoi. 20, 70-75 (2002).
16 . Stephan, M .T. et al. T cell-encoded CD80 and 4-1 BBL induce auto- and
transcostimuiation, resulting i n potent tumor rejection. Nature medicine 13, 1440-1449 (2007). 17. Watts, T.H. TNF/TNFR family members in costimulation of T cell responses. Annual
review of immunology 23, 23-68 (2005).
18. Wang, J. et al. Optimizing adoptive polyclonal T cell immunotherapy of lymphomas,
using a chimeric T cell receptor possessing CD28 and CD 137 costimulatory domains. Human gene therapy 18, 712-725 (2007).
19 . Ca rpe n ito, C . et al. Control of large, established tumor xenografts with genetically
retargeted human T cells containing CD28 and CD137 domains. Proceedings of the National
Academy of Sciences of the United States of America 106, 3360-3365 (2009).
20. Tammana, S. et a 1. 4-1 BB and CD28 signaling plays a synergistic role in redirecting umbilical cord blood T cells against B-cell malignancies. Hum Gene Ther 2 1 , 75 -86 ( 2 0 10) .
21. Zhong, X.S., Matsushita, M., Piotkin, J., Riviere, I. & Sadeiain, M. Chimeric antigen receptors combining 4-1 BB and CD28 signaling domains augment
Pi3kinase/AKT/Bc1-X L
activation and C D8+ T cell-mediated tumor eradication. Molecular therapy: the journal of the
American Society of Gene Therapy 18, 413-420 (2010).
22. Schwartz, R . H . T cell a n e rgy. Annual review of immunology 21 , 305-334 (2003).
23. Liao, W., Lin, J .X . & Leonard, W.J . I L-2 family cytokines: new insights into the complex roles of IL-2 as a broad regulator of T helper ceil differentiation.
Current opinion in
immunology 23, 598-604 (201 1 ).
24 . Nam, K. 0 ., Shin, S . M . & Lee, H.W. Cross-linking of 4-IBB up- regulates IL-13
expression in CD8(+) T lymphocytes. Cytokine 33, 87-94 (2006).
25. Shin, S M . et a 1. 4- IBB triggers IL-13 production from T cells to limit the polarized,
Thl-mediated inflammation. Journal of leukocyte biology 81 , 1455-1465 (2007). 26. Saeki, N.. Gu, J., Yoshida, T. & Wu, X. Prostate stem cell antigen: a Jekyil and Hyde
molecule? Ciinicai cancer research : an official journal of the American Association for Cancer Research 16, 3533-3538 (2010).
27. Olson, W.C., Heston, W.D. & Rajasekaran, A.K. Ciinicai trials of cancer therapies targeting prostate-specific membrane antigen. Reviews on recent ciinicai trials 2, 182-190 30 (2007).
28. Lam, J.S. et al. Prostate stem cell antigen is overexpressed in prostate cancer metastases.
Ciinicai cancer research : an official journal of the American Association for Cancer Research 11 , 2591-2596 (2005).
29. Silver, D.A., Pellicer, I., Fair, W.R., Heston, W.D. & Cordon-Cardo, C. Prostate-specific
membrane antigen expression in normal and malignant human tissues. Ciinicai cancer research :
an official journal of the American Association for Cancer Research 3, 81-85 (1997).
30. Liu, J.C. et al. Seventeen-gene signature from enriched Her2/Neu mammary tumor- initiating cells predicts clinical outcome for human HER2+:ERalpha- breast cancer. Proceedings of the National Academy of Sciences of the United States of America 109, 58325837 (2012).
31. Meyer, M.J. et al. CD44posCD49fhiCD133/2hi defines xenograft-initiating cells in
estrogen receptor-negative breast cancer. Cancer research 70, 4624-4633
(2010) .
32. Strauss, R. et al. Analysis of epitheiial and mesenchymai markers in ovarian cancer reveals phenotypic heterogeneity and plasticity. PioS one 6, ei 6186
(2011 ) .
33. Shihle, M . & Davidson, B. Pathogenesis of ovarian cancer: clues from selected
overexpressed genes. Future Oncol 5, 1641-1657 (2009). 34. Nguyen, L.V., Vanner, R., Dirks, P. & Eaves, C.J. Cancer stem cells: an evolving concept. Nature reviews. Cancer 12, 133-143 (2012).
5. Magee, J.A., Piskounova, E. & Morrison, S.J. Cancer stem cells: impact, heterogeneity, and uncertainty. Cancer cell 21 , 283-296 (2012).
36. Orlandi, R., Gussow, D.H., Jones, P.T. & Winter, G. Cloning immunoglobulin variable
domains for expression by the polymerase chain reaction. Proceedings of the National Academy of Sciences of the United States of America 86, 3833-3837 (1989).

Claims

What is claimed is:
1. An immunoresponsive cell comprising:
a. an antigen recognizing receptor that binds a first antigen with low affinity, wherein binding of the receptor to the first antigen activates the
immunoresponsive cell, and
b. a chimeric co-stimulating receptor (CCR) that binds a second antigen and stimulates the immunoresponsive cell.
2. The immunoresponsive cell of any one of claims 1 , wherein the cell is selected from the group consisting of a T cell, a Natural Killer (NK) cell, a cytotoxic T lymphocyte (CTL), a regulatory T cell, a human embryonic stem ceil, and a pluripotent stem ceil from which lymphoid cells may be differentiated.
3. The immunoresponsive cell of claim 1 or 2, wherein the antigen is a tumor or pathogen antigen.
4. The immunoresponsive ceil of any of claims 1 -3, wherein said
antigen recognizing receptor is a T cell receptor (TCR) or chimeric antigen receptor (CAR).
5. The immunoresponsive cell of any one of claims 1-4, wherein said antigen recognizing receptor is exogenous or endogenous.
6. The immunoresponsive cell of any one of claims 1-5, wherein said antigen recognizing receptor is recombinantiy expressed.
7. The immunoresponsive ceil of any one of claims 1-6, wherein the antigen recognizing receptor is expressed from a vector.
8. The immunoresponsive cell of any one of claims 1-7, wherein the
chimeric co-stimulating receptor (CCR) is expressed from a vector.
9. The immunoresponsive ceil of any one of claims 1-8, wherein said immunoresponsive ceil is autologous.
10. The immunoresponsive cell of any one of claims 1-9, wherein said first and second antigens are selected from the group consisting of CAIX, CEA, CD5, CD7, CD10, CD19, CD20, CD22, CD30, CD33, CD34, CD38, CD41, CD44, CD49f, CD56, CD74, CD133, CD138, a cytomegalovirus (CMV) infected cell antigen, EGP-2, EGP-40, EpCAM, erb-B2,3,4, FBP, Fetal acetylcholine receptor, folate receptor-a, GD2, GD3, HER-2, hTERT, IL-13R-a2, x-light chain, KDR, LeY, LI cell adhesion molecule, MAGE-AI , MUC1, Mesotheiin, NKG2D iigands, NY-ESO-1, oncofetal antigen (h5T4), PSCA, PSMA, ROR1, TAG-72, VEGF-R2, or WT-1.
11. The immunoresponsive cell of any one of claims 1-9,
wherein said first and second antigens are distinct antigens selected from the group consisting of CD133, a cytomegalovirus (CMV) infected cell antigen, erb-B2, KDR Mesotheiin, NKG2D iigands, NY-ESO-1,
oncofetal antigen (h5T4), PSCA, PSMA, CD19, VEGF-R2, and WT-1.
12. The immunoresponsive ceil of any one of claims 1-9, wherein said first and second antigens are selected from the group consisting of HER2, MUC1 , CD44, CD49f, EpCAM, CEA, CD133, a cytomegalovirus (CMV) infected cell antigen, EGP-2, EGP-40, EpCAM, erb-B2,3,4, FBP, KDR, Mesotheiin, NKG2D iigands, NY-ESO-1, oncofetal antigen (h5T4), PSCA, PSMA, VEGF-R2, or WT-1.
13. The immunoresponsive cell of any one of claims 1-11, wherein said first and second antigens are CD10 and CD19.
14. The immunoresponsive ceil of any one of claims 1-11, wherein said first and second antigens are CD56 and CD138.
15. The immunoresponsive cell of any one of claims 1-11, wherein said first and second antigens are distinct antigens selected from the group
consisting of mesotheiin, folate receptor-a, CD44, and CD133.
16. The immunoresponsive ceii of any one of ciaims 1 -15,
wherein the intracellular signaling domain of said antigen recognizing
receptor is the CD3 -chain signaling domain.
17. The immunoresponsive ceil of any one of claims 1-16,
wherein the intracellular signaling domain of the chimeric co-stimulating
receptor (CCR) is a CD97, CD1 ia-CD18, CD2, ICOS, CD27, CD154, CD5, OX40, 4-1 BB or CD28 signaling domain.
18. The immunoresponsive cell of any one of claims 1 -17, wherein the cell expresses an antigen receptor that is 19z1 or Pz1 .
19. A method of inducing tumor cell death in a tumor, the
method comprising contacting said tumor eel! with an immunoresponsive cell comprising an antigen recognizing receptor that binds a first tumor antigen with low affinity, wherein the binding activates the
immunoresponsive cell, and a chimeric co-stimulating receptor (CCR) that binds a second tumor antigen and stimulates the immunoresponsive cell, thereby inducing tumor eel! death in said tumor.
20. The method of claim 19, wherein the method reduces the number of tumor cells.
21. The method of claim 19, wherein the method reduces tumor size.
22. The method of claim 19, wherein the method eradicates the tumor.
23. A method of treating or preventing a neoplasia in a subject, the method comprising administering an effective amount of an
immunoresponsive ceii comprising an antigen recognizing receptor that binds a first antigen with low affinity, wherein the binding activates the
immunoresponsive ceii, and a chimeric co-stimulating receptor (CCR) that binds a second antigen and stimulates the immunoresponsive cell, thereby treating or preventing a neoplasia in the subject.
24. The method of claim 23, wherein the neoplasia is selected from the group consisting of prostate cancer, breast cancer, B ceil leukemia, multiple myeloma, and ovarian cancer.
25. The method of claim 23, wherein the neoplasia is breast cancer and the first and second tumor antigens are distinct antigens selected from the group consisting of HER2, MUC1 , CD44, CD49f, EpCAM, CEA, CD133, a
cytomegalovirus (CMV) infected cell antigen, EGP-2, EGP-40, EpCAM, erb- B2.3.4, FBP, KDR, Mesothelin, NKG2D ligands, NY-ESO-1 , oncofetal antigen (h5T4), PSCA, PSMA, VEGF-R2, or WT-1.
26. The method of claim 23, wherein the neoplasia is B cell leukemia and the first and second tumor antigens are CD10 and CD19.
27. The method of claim 24, wherein the neoplasia is multiple myeloma and the first and second tumor antigens are CD56 and CD138.
28. The method of claim 24, wherein the neoplasia is ovarian cancer and the first and second tumor antigens are distinct antigens selected from the group consisting of mesothelin, folate receptor-a, CD44, and CD133.
29. The method of any one of claims 23-28, wherein said immunoresponsive ceil is selected as having an antigen recognizing receptor with low affinity.
30. The method of any one of claims 23-28, wherein said antigen recognizing receptor is selected for expression in the cell as having low affinity.
31 . The method of any one of claims 23-28, wherein said antigen
recognizing receptor is a T cell receptor (TCR) or chimeric antigen receptor (CAR).
32. The method of any one of claims 23-28, wherein said antigen recognizing receptor is exogenous or endogenous.
33. The method of any one of claims 23-32, wherein the antigen
recognizing receptor is expressed from a vector.
34. The method of any one of claims 23-32, wherein the chimeric co- stimulating receptor (CCR) is expressed from a vector.
35. The method of any one of claims 23-34, wherein the cell is selected from the group consisting of a T cell, a Natural Killer (NK) cell, a cytotoxic T lymphocyte (CTL), a regulatory T ceil, a human embryonic stem cell, and a pluripotent stem ceil from which lymphoid cells may be differentiated.
36. The method of any one of claims 23-35, wherein the method reduces or eradicates the tumor burden in the subject.
37. A method of inducing tumor eel! death, the method comprising administering an effective amount of an immunoresponsive cell comprising an antigen recognizing receptor that binds a first antigen with low affinity, wherein the binding activates the immunoresponsive ceil, and a chimeric co-stimulating receptor (CCR) that binds a second antigen and stimulates the immunoresponsive cell, thereby inducing tumor cell death in the subject.
38. A method for producing an antigen-specific immunoresponsive cell, the method comprising introducing into the immunoresponsive cell a nucleic acid sequence that encodes a chimeric co-stimulating receptor (CCR), wherein the chimeric co-stimulating receptor comprises an
antigen-binding domain coupled to an intracellular signaling domain that stimulates an immunoresponsive ceil, wherein the immunoresponsive cell comprises an antigen recognizing receptor that binds a first antigen with low affinity, wherein the binding activates the immunoresponsive cell.
39. The method of claim 38, the method further comprising introducing a second nucleic acid sequence that encodes a chimeric antigen receptor, wherein the chimeric antigen receptor comprises a second antigen-binding domain coupled to a second intracellular signaling domain that activates an immunoresponsive celi.
40. The method of any one of claims 38-39, wherein the tumor antigens are distinct antigens selected from the list consisting of CAIX, CEA, CD5, CD7, CDIO, CD19, CD20, CD22, CD30, CD33, CD34, CD38, CD41 , CD44, CD49f, CD56, CD74, CD133, CD138, a cytomegalovirus (CMV) infected ceil antigen, EGP-2, EGP-40, EpCAM, erbB2,3,4, FBP, Fetal acetyichoiine receptor, folate receptor-a, GD2, GD3, HER-2,hTERT, IL-13R-a2, x-light chain, KDR, LeY, LI ceil adhesion molecule, MAGE-AI, MUC1 , Mesothelin, NKG2D iigands, NY-ES0-1 , oncofetal antigen (h5T4), PSCA, PSMA, ROR1 , TAG-72, VEGF-R2, or WT-1.
41. The method of any one of ciaims 38-40, wherein said antigen
recognizing receptor is a T ceil receptor (TCR) or chimeric antigen receptor (CAR).
42. The method of any one of ciaims 38-41 , wherein said antigen
recognizing receptor is exogenous or endogenous.
43. The method of any one of claims 38-42, wherein said antigen
recognizing receptor is recombinantly expressed.
44. The method of any one of ciaims 38-43, wherein the antigen
recognizing receptor is expressed from a vector.
45. The method of any one of ciaims 38-44, wherein the chimeric co-stimulating receptor (CCR) is expressed from a vector.
46. The method of any one of ciaims 38-45, wherein the cell is selected from the group consisting of a T cell, a Natural Killer (NK) cell, a cytotoxic T lymphocyte (CTL), a reguiatory T ceil, a human embryonic stem celi, and a pluripotent stem ceil from which lymphoid ceils may be differentiated.
47. The method of any one of claims 38-46, wherein the intracellular
signaling domain of said antigen recognizing receptor is the CD3 -chain
signaling domain.
48. The method of any one of claims 38-47, wherein the intracellular signaling domain of the chimeric co-stimulating receptor (CCR) is a CD97, CD11a-CD18, CD2, ICOS, CD27, CD154, CD5, OX40, 4-1 BB or CD28 signaling domain.
49. A method of treating prostate cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a T cell comprising an antigen recognizing receptor that binds PSCA or CD19 with low affinity, wherein the binding activates the immunoresponsive cell, and a chimeric co-stimulating receptor (CCR) that binds PSMA and stimulates the
immunoresponsive cell, thereby treating prostate cancer in the subject.
50. A pharmaceutical composition comprising an immunoresponsive cell of any one of claims 1-18 and a pharmaceutically acceptable excipient.
53. An immunoresponsive cell of any of claims 1-18 for use in the treatment of a neoplasia or tumor disease.
54. A T cell comprising an antigen recognizing receptor that binds PSCA or CD19 with low affinity, wherein the binding activates the immunoresponsive ceil, and a chimeric co-stimulating receptor (CCR) that binds PSMA and stimulates the immunoresponsive ceil for use in the treatment of prostate cancer.
55. A kit comprising an immunoresponsive cell comprising an antigen recognizing receptor that binds a first antigen and activates the
immunoresponsive cell, and a chimeric co-stimulating receptor (CCR) that binds a second viral antigen and stimulates the immunoresponsive cell.
PCT/US2013/063097 2012-10-02 2013-10-02 Compositions and methods for immunotherapy WO2014055668A1 (en)

Priority Applications (23)

Application Number Priority Date Filing Date Title
EP13844468.2A EP2903637B1 (en) 2012-10-02 2013-10-02 Compositions and methods for immunotherapy
RU2015116901A RU2729401C2 (en) 2012-10-02 2013-10-02 Compositions and methods for immunotherapy
AU2013327136A AU2013327136A1 (en) 2012-10-02 2013-10-02 Compositions and methods for immunotherapy
CN201380063036.8A CN104853766A (en) 2012-10-02 2013-10-02 Compositions and methods for immunotherapy
CA2886859A CA2886859C (en) 2012-10-02 2013-10-02 Compositions and methods for immunotherapy
JP2015534828A JP6441802B2 (en) 2012-10-02 2013-10-02 Compositions and methods for immunotherapy
NZ706541A NZ706541A (en) 2012-10-02 2013-10-02 Compositions and methods for immunotherapy
ES13844468T ES2743738T3 (en) 2012-10-02 2013-10-02 Compositions and methods for immunotherapy
SG11201502598SA SG11201502598SA (en) 2012-10-02 2013-10-02 Compositions and methods for immunotherapy
KR1020157011160A KR102198058B1 (en) 2012-10-02 2013-10-02 Compositions and methods for immunotherapy
EP19179456.9A EP3597215A1 (en) 2012-10-02 2013-10-02 Compositions and methods for immunotherapy
MX2015004287A MX370148B (en) 2012-10-02 2013-10-02 Compositions and methods for immunotherapy.
IL238047A IL238047B (en) 2012-10-02 2015-03-30 Compositions and methods for immunotherapy
US14/676,255 US10654928B2 (en) 2012-10-02 2015-04-01 Compositions and methods for immunotherapy
PH12015500747A PH12015500747A1 (en) 2012-10-02 2015-04-06 Compositions and methods for immunotherapy
ZA201502880A ZA201502880B (en) 2012-10-02 2015-04-28 Compositions and methods for immunotherapy
HK15109439.3A HK1208631A1 (en) 2012-10-02 2015-09-24 Compositions and methods for immunotherapy
AU2018204297A AU2018204297B2 (en) 2012-10-02 2018-06-15 Compositions and methods for immunotherapy
PH12019502424A PH12019502424A1 (en) 2012-10-02 2019-10-25 Compositions and methods for immunotherapy
IL272539A IL272539B (en) 2012-10-02 2020-02-07 Compositions and methods for immunotherapy
US16/847,059 US11712469B2 (en) 2012-10-02 2020-04-13 Compositions and methods for immunotherapy
AU2020294287A AU2020294287B2 (en) 2012-10-02 2020-12-24 Compositions and methods for immunotherapy
US18/333,753 US20240091353A1 (en) 2012-10-02 2023-06-13 Compositions and methods for immunotherapy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261709072P 2012-10-02 2012-10-02
US61/709,072 2012-10-02

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/676,255 Continuation US10654928B2 (en) 2012-10-02 2015-04-01 Compositions and methods for immunotherapy

Publications (1)

Publication Number Publication Date
WO2014055668A1 true WO2014055668A1 (en) 2014-04-10

Family

ID=50435409

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/063097 WO2014055668A1 (en) 2012-10-02 2013-10-02 Compositions and methods for immunotherapy

Country Status (17)

Country Link
US (3) US10654928B2 (en)
EP (2) EP3597215A1 (en)
JP (5) JP6441802B2 (en)
KR (1) KR102198058B1 (en)
CN (3) CN104853766A (en)
AU (3) AU2013327136A1 (en)
CA (1) CA2886859C (en)
ES (1) ES2743738T3 (en)
HK (1) HK1208631A1 (en)
IL (2) IL238047B (en)
MX (1) MX370148B (en)
NZ (2) NZ746914A (en)
PH (2) PH12015500747A1 (en)
RU (2) RU2020124583A (en)
SG (1) SG11201502598SA (en)
WO (1) WO2014055668A1 (en)
ZA (1) ZA201502880B (en)

Cited By (254)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014134165A1 (en) * 2013-02-26 2014-09-04 Memorial Sloan-Kettering Cancer Center Compositions and methods for immunotherapy
WO2015188141A2 (en) 2014-06-06 2015-12-10 Memorial Sloan-Kettering Cancer Ceneter Mesothelin-targeted chimeric antigen receptors and uses thereof
WO2015188119A1 (en) * 2014-06-06 2015-12-10 Bluebird Bio, Inc. Improved t cell compositions
WO2016033570A1 (en) 2014-08-28 2016-03-03 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for cd19
WO2016064929A1 (en) 2014-10-20 2016-04-28 Juno Therapeutics, Inc. Methods and compositions for dosing in adoptive cell therapy
WO2016073602A2 (en) 2014-11-05 2016-05-12 Juno Therapeutics, Inc. Methods for transduction and cell processing
WO2016090190A1 (en) 2014-12-03 2016-06-09 Juno Therapeutics, Inc. Methods and compositions for adoptive cell therapy
WO2016115177A1 (en) 2015-01-12 2016-07-21 Juno Therapeutics, Inc. Modified hepatitis post-transcriptional regulatory elements
WO2016115559A1 (en) 2015-01-16 2016-07-21 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ror1
WO2016138034A1 (en) * 2015-02-24 2016-09-01 The Regents Of The University Of California Binding-triggered transcriptional switches and methods of use thereof
WO2016166568A1 (en) 2015-04-16 2016-10-20 Juno Therapeutics Gmbh Methods, kits and apparatus for expanding a population of cells
WO2016168493A1 (en) * 2015-04-15 2016-10-20 Prospect CharterCare RWMC, LLC d/b/a Roger Williams Medical Center Hepatic arterial infusion of car-t cells
WO2016172606A1 (en) * 2015-04-23 2016-10-27 Baylor College Of Medicine Cd5 chimeric antigen receptor for adoptive t cell therapy
WO2016196388A1 (en) 2015-05-29 2016-12-08 Juno Therapeutics, Inc. Composition and methods for regulating inhibitory interactions in genetically engineered cells
US9546206B2 (en) 2014-08-08 2017-01-17 The Board Of Trustees Of The Leland Stanford Junior University High affinity PD-1 agents and methods of use
WO2017021701A1 (en) * 2015-07-31 2017-02-09 King's College London Therapeutic agents
WO2017040945A1 (en) 2015-09-04 2017-03-09 Memorial Sloan Kettering Cancer Center Immune cell compositions and methods of use
WO2017053906A1 (en) 2015-09-24 2017-03-30 Abvitro Llc Hiv antibody compositions and methods of use
CN106574272A (en) * 2014-08-29 2017-04-19 哥莫阿波单克隆有限责任公司 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
WO2017068421A1 (en) 2015-10-22 2017-04-27 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
WO2017068425A1 (en) 2015-10-22 2017-04-27 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
WO2017068419A2 (en) 2015-10-22 2017-04-27 Juno Therapeutics Gmbh Methods, kits, agents and apparatuses for transduction
WO2017079705A1 (en) 2015-11-05 2017-05-11 Juno Therapeutics, Inc. Chimeric receptors containing traf-inducing domains and related compositions and methods
WO2017079703A1 (en) 2015-11-05 2017-05-11 Juno Therapeutics, Inc. Vectors and genetically engineered immune cells expressing metabolic pathway modulators and uses in adoptive cell therapy
WO2017096327A2 (en) 2015-12-03 2017-06-08 Juno Therapeutics, Inc. Compositions and methods for reducing immune responses against cell therapies
WO2017096329A1 (en) 2015-12-03 2017-06-08 Juno Therapeutics, Inc. Modified chimeric receptors and related compositions and methods
WO2017120501A1 (en) 2016-01-07 2017-07-13 Mayo Foundation For Medical Education And Research Methods of treating cancer with interferon
CN107002045A (en) * 2014-12-24 2017-08-01 Ucl商务股份有限公司 Cell
WO2017161212A1 (en) 2016-03-16 2017-09-21 Juno Therapeutics, Inc. Methods for adaptive design of a treatment regimen and related treatments
WO2017161208A1 (en) 2016-03-16 2017-09-21 Juno Therapeutics, Inc. Methods for determining dosing of a therapeutic agent and related treatments
WO2017165571A1 (en) 2016-03-22 2017-09-28 Seattle Children's Hospital (dba Seattle Children's Research Institute) Early intervention methods to prevent or ameliorate toxicity
US9777061B2 (en) 2014-07-21 2017-10-03 Novartis Ag Treatment of cancer using a CD33 chimeric antigen receptor
WO2017173091A1 (en) 2016-03-30 2017-10-05 Musc Foundation For Research Development Methods for treatment and diagnosis of cancer by targeting glycoprotein a repetitions predominant (garp) and for providing effective immunotherapy alone or in combination
WO2017193107A2 (en) 2016-05-06 2017-11-09 Juno Therapeutics, Inc. Genetically engineered cells and methods of making the same
JP2017533706A (en) * 2014-10-31 2017-11-16 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Methods and compositions for modified T cells
WO2017205846A1 (en) 2016-05-27 2017-11-30 Aadigen, Llc Peptides and nanoparticles for intracellular delivery of genome-editing molecules
WO2017214207A2 (en) 2016-06-06 2017-12-14 Juno Therapeutics, Inc. Methods for the treatment of b cell malignancies using adoptive cell therapy
WO2018005556A1 (en) 2016-06-27 2018-01-04 Juno Therapeutics, Inc. Mhc-e restricted epitopes, binding molecules and related methods and uses
WO2018005559A1 (en) 2016-06-27 2018-01-04 Juno Therapeutics, Inc. Method of identifying peptide epitopes, molecules that bind such epitopes and related uses
WO2018023094A1 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Methods for assessing the presence or absence of replication competent virus
WO2018023100A2 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Anti-idiotypic antibodies and related methods
WO2018023093A1 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Immunomodulatory polypeptides and related compositions and methods
WO2018027036A1 (en) * 2016-08-03 2018-02-08 Dipersio John F Gene editing of car-t cells for the treatment of t cell malignancies with chimeric antigen receptors
WO2018049420A1 (en) 2016-09-12 2018-03-15 Juno Therapeutics, Inc. Perfusion bioreactor bag assemblies
WO2018063985A1 (en) 2016-09-28 2018-04-05 Atossa Genetics Inc. Methods of adoptive cell therapy
WO2018067618A1 (en) 2016-10-03 2018-04-12 Juno Therapeutics, Inc. Hpv-specific binding molecules
CN107922488A (en) * 2015-04-02 2018-04-17 纪念斯隆-凯特琳癌症中心 TNFRSF14/HVEM albumen and its application method
WO2018071873A2 (en) 2016-10-13 2018-04-19 Juno Therapeutics, Inc. Immunotherapy methods and compositions involving tryptophan metabolic pathway modulators
WO2018085731A2 (en) 2016-11-03 2018-05-11 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and a btk inhibitor
WO2018093591A1 (en) 2016-11-03 2018-05-24 Juno Therapeutics, Inc. Combination therapy of a cell based therapy and a microglia inhibitor
WO2018102787A1 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods for determining car-t cells dosing
WO2018102612A1 (en) 2016-12-02 2018-06-07 Juno Therapeutics, Inc. Engineered b cells and related compositions and methods
WO2018102785A2 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods and compositions for use of therapeutic t cells in combination with kinase inhibitors
WO2018102786A1 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods for modulation of car-t cells
WO2018106732A1 (en) 2016-12-05 2018-06-14 Juno Therapeutics, Inc. Production of engineered cells for adoptive cell therapy
AU2014214850B2 (en) * 2013-02-06 2018-06-14 Celgene Corporation Modified T lymphocytes having improved specificity
EP3336107A1 (en) 2016-12-15 2018-06-20 Miltenyi Biotec GmbH Immune cells expressing an antigen binding receptor and a chimeric costimulatory receptor
WO2018132518A1 (en) 2017-01-10 2018-07-19 Juno Therapeutics, Inc. Epigenetic analysis of cell therapy and related methods
WO2018134691A2 (en) 2017-01-20 2018-07-26 Juno Therapeutics Gmbh Cell surface conjugates and related cell compositions and methods
JP2018522567A (en) * 2015-08-07 2018-08-16 シアトル チルドレンズ ホスピタル, ディービーエー シアトル チルドレンズ リサーチ インスティテュート Bispecific CAR T cells targeting solid tumors
WO2018157171A2 (en) 2017-02-27 2018-08-30 Juno Therapeutics, Inc. Compositions, articles of manufacture and methods related to dosing in cell therapy
WO2018165228A1 (en) 2017-03-08 2018-09-13 Memorial Sloan Kettering Cancer Center Immune cell compositions and methods of use
WO2018170188A2 (en) 2017-03-14 2018-09-20 Juno Therapeutics, Inc. Methods for cryogenic storage
WO2018167486A1 (en) 2017-03-15 2018-09-20 Oxford Biomedica (Uk) Limited Method
WO2018187791A1 (en) 2017-04-07 2018-10-11 Juno Therapeutics, Inc Engineered cells expressing prostate-specific membrane antigen (psma) or a modified form thereof and related methods
WO2018191723A1 (en) 2017-04-14 2018-10-18 Juno Therapeutics, Inc. Methods for assessing cell surface glycosylation
WO2018195175A1 (en) 2017-04-18 2018-10-25 FUJIFILM Cellular Dynamics, Inc. Antigen-specific immune effector cells
WO2018197949A1 (en) 2017-04-27 2018-11-01 Juno Therapeutics Gmbh Oligomeric particle reagents and methods of use thereof
WO2018200583A1 (en) * 2017-04-26 2018-11-01 Eureka Therapeutics, Inc. Cells expressing chimeric activating receptors and chimeric stimulating receptors and uses thereof
US10117896B2 (en) 2012-10-05 2018-11-06 The Trustees Of The University Of Pennsylvania Use of a trans-signaling approach in chimeric antigen receptors
WO2018204427A1 (en) 2017-05-01 2018-11-08 Juno Therapeutics, Inc. Combination of a cell therapy and an immunomodulatory compound
US20180348227A1 (en) * 2016-08-04 2018-12-06 Memorial Sloan-Kettering Cancer Center Cancer antigen targets and uses thereof
WO2018223098A1 (en) 2017-06-02 2018-12-06 Juno Therapeutics, Inc. Articles of manufacture and methods related to toxicity associated with cell therapy
WO2018223101A1 (en) 2017-06-02 2018-12-06 Juno Therapeutics, Inc. Articles of manufacture and methods for treatment using adoptive cell therapy
WO2018234370A1 (en) 2017-06-20 2018-12-27 Institut Curie Immune cells defective for suv39h1
WO2019006427A1 (en) 2017-06-29 2019-01-03 Juno Therapeutics, Inc. Mouse model for assessing toxicities associated with immunotherapies
WO2019027850A1 (en) 2017-07-29 2019-02-07 Juno Therapeutics, Inc. Reagents for expanding cells expressing recombinant receptors
WO2019032927A1 (en) 2017-08-09 2019-02-14 Juno Therapeutics, Inc. Methods for producing genetically engineered cell compositions and related compositions
WO2019032929A1 (en) 2017-08-09 2019-02-14 Juno Therapeutics, Inc. Methods and compositions for preparing genetically engineered cells
WO2019046832A1 (en) 2017-09-01 2019-03-07 Juno Therapeutics, Inc. Gene expression and assessment of risk of developing toxicity following cell therapy
WO2019051335A1 (en) 2017-09-07 2019-03-14 Juno Therapeutics, Inc. Methods of identifying cellular attributes related to outcomes associated with cell therapy
WO2019057102A1 (en) 2017-09-20 2019-03-28 Tsinghua University A gRNA TARGETING HPK1 AND A METHOD FOR EDITING HPK1 GENE
EP3307876A4 (en) * 2015-06-10 2019-04-03 Nantkwest, Inc. Modified nk-92 cells for treating cancer
WO2019070541A1 (en) 2017-10-03 2019-04-11 Juno Therapeutics, Inc. Hpv-specific binding molecules
EP3328994A4 (en) * 2015-07-31 2019-04-17 Memorial Sloan-Kettering Cancer Center Antigen-binding proteins targeting cd56 and uses thereof
WO2019090202A1 (en) 2017-11-06 2019-05-09 Editas Medicine, Inc. Methods, compositions and components for crispr-cas9 editing of cblb in t cells for immunotherapy
WO2019089982A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Method of assessing activity of recombinant antigen receptors
WO2019089855A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Process for generating therapeutic compositions of engineered cells
WO2019090364A1 (en) 2017-11-06 2019-05-09 Juno Therapeutics, Inc. Combination of a cell therapy and a gamma secretase inhibitor
WO2019090003A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Chimeric antigen receptors specific for b-cell maturation antigen (bcma)
WO2019089848A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods associated with tumor burden for assessing response to a cell therapy
WO2019089884A2 (en) 2017-11-01 2019-05-09 Editas Medicine, Inc. Methods, compositions and components for crispr-cas9 editing of tgfbr2 in t cells for immunotherapy
WO2019087151A1 (en) 2017-11-03 2019-05-09 Sorrento Therapeutics, Inc. Cd38-directed chimeric antigen receptor constructs
WO2019090004A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Process for producing a t cell composition
WO2019089858A2 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods of assessing or monitoring a response to a cell therapy
WO2019094835A1 (en) 2017-11-10 2019-05-16 Juno Therapeutics, Inc. Closed-system cryogenic vessels
WO2019109053A1 (en) 2017-12-01 2019-06-06 Juno Therapeutics, Inc. Methods for dosing and for modulation of genetically engineered cells
WO2019113556A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Serum-free media formulation for culturing cells and methods of use thereof
WO2019113559A2 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Phenotypic markers for cell therapy and related methods
WO2019113557A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Process for producing a composition of engineered t cells
WO2019118937A1 (en) 2017-12-15 2019-06-20 Juno Therapeutics, Inc. Anti-cct5 binding molecules and methods of use thereof
WO2019141270A1 (en) * 2018-01-19 2019-07-25 科济生物医药(上海)有限公司 Synnotch receptor-regulated expression of il12
WO2019152743A1 (en) 2018-01-31 2019-08-08 Celgene Corporation Combination therapy using adoptive cell therapy and checkpoint inhibitor
WO2019152747A1 (en) 2018-01-31 2019-08-08 Juno Therapeutics, Inc. Methods and reagents for assessing the presence or absence of replication competent virus
WO2019148656A1 (en) * 2018-02-05 2019-08-08 深圳市默赛尔生物医学科技发展有限公司 Multi-signal chimeric antigen receptor and expression gene thereof, nk cells modified thereby and anti-tumor drug
US10383929B2 (en) 2014-12-12 2019-08-20 Bluebird Bio, Inc. BCMA chimeric antigen receptors
WO2019161133A1 (en) 2018-02-15 2019-08-22 Memorial Sloan Kettering Cancer Center Foxp3 targeting agent compositions and methods of use for adoptive cell therapy
WO2019170147A1 (en) 2018-03-09 2019-09-12 科济生物医药(上海)有限公司 Method and composition for treating tumors
WO2019170845A1 (en) 2018-03-09 2019-09-12 Ospedale San Raffaele S.R.L. Il-1 antagonist and toxicity induced by cell therapy
US10428305B2 (en) 2014-05-15 2019-10-01 National University Of Singapore Modified natural killer cells that express IL15 and uses thereof
WO2019195491A1 (en) 2018-04-05 2019-10-10 Juno Therapeutics, Inc. T cells expressing a recombinant receptor, related polynucleotides and methods
WO2019195486A1 (en) 2018-04-05 2019-10-10 Juno Therapeutics, Inc. T cell receptors and engineered cells expressing same
WO2019195492A1 (en) 2018-04-05 2019-10-10 Juno Therapeutics, Inc. Methods of producing cells expressing a recombinant receptor and related compositions
US10464988B2 (en) 2015-10-23 2019-11-05 Eureka Therapeutics, Inc. Antibody/T-cell receptor chimeric constructs and uses thereof
WO2019213184A1 (en) 2018-05-03 2019-11-07 Juno Therapeutics, Inc. Combination therapy of a chimeric antigen receptor (car) t cell therapy and a kinase inhibitor
US10538739B2 (en) 2013-01-28 2020-01-21 St. Jude Children's Research Hospital, Inc. Chimeric receptor with NKG2D specificity for use in cell therapy against cancer and infectious disease
WO2020020210A1 (en) 2018-07-24 2020-01-30 科济生物医药(上海)有限公司 Method for treating tumor using immune effector cell
WO2020033916A1 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Methods for assessing integrated nucleic acids
WO2020033927A2 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Processes for generating engineered cells and compositions thereof
WO2020056047A1 (en) 2018-09-11 2020-03-19 Juno Therapeutics, Inc. Methods for mass spectrometry analysis of engineered cell compositions
WO2020061256A1 (en) 2018-09-19 2020-03-26 FUJIFILM Cellular Dynamics, Inc. Protein l for activation and expansion of chimeric antigen receptor-modified immune cells
WO2020092854A2 (en) 2018-11-01 2020-05-07 Juno Therapeutics, Inc. Chimeric antigen receptors specific for g protein-coupled receptor class c group 5 member d (gprc5d)
WO2020089343A1 (en) 2018-10-31 2020-05-07 Juno Therapeutics Gmbh Methods for selection and stimulation of cells and apparatus for same
WO2020092848A2 (en) 2018-11-01 2020-05-07 Juno Therapeutics, Inc. Methods for treatment using chimeric antigen receptors specific for b-cell maturation antigen
WO2020097403A1 (en) 2018-11-08 2020-05-14 Juno Therapeutics, Inc. Methods and combinations for treatment and t cell modulation
WO2020097132A1 (en) 2018-11-06 2020-05-14 Juno Therapeutics, Inc. Process for producing genetically engineered t cells
WO2020102770A1 (en) 2018-11-16 2020-05-22 Juno Therapeutics, Inc. Methods of dosing engineered t cells for the treatment of b cell malignancies
WO2020106621A1 (en) 2018-11-19 2020-05-28 Board Of Regents, The University Of Texas System A modular, polycistronic vector for car and tcr transduction
WO2020113029A2 (en) 2018-11-28 2020-06-04 Board Of Regents, The University Of Texas System Multiplex genome editing of immune cells to enhance functionality and resistance to suppressive environment
WO2020112493A1 (en) 2018-11-29 2020-06-04 Board Of Regents, The University Of Texas System Methods for ex vivo expansion of natural killer cells and use thereof
WO2020113188A2 (en) 2018-11-30 2020-06-04 Juno Therapeutics, Inc. Methods for dosing and treatment of b cell malignancies in adoptive cell therapy
WO2020113194A2 (en) 2018-11-30 2020-06-04 Juno Therapeutics, Inc. Methods for treatment using adoptive cell therapy
WO2020160050A1 (en) 2019-01-29 2020-08-06 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for receptor tyrosine kinase like orphan receptor 1 (ror1)
US10738278B2 (en) 2014-07-15 2020-08-11 Juno Therapeutics, Inc. Engineered cells for adoptive cell therapy
WO2020169974A1 (en) 2019-02-19 2020-08-27 King's College London Hypoxia-responsive chimeric antigen receptors
US10774343B2 (en) 2014-04-25 2020-09-15 Bluebird Bio, Inc. MND promoter chimeric antigen receptors
US10786533B2 (en) 2015-07-15 2020-09-29 Juno Therapeutics, Inc. Engineered cells for adoptive cell therapy
WO2020223571A1 (en) 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cells expressing a chimeric receptor from a modified cd247 locus, related polynucleotides and methods
WO2020223535A1 (en) 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cells expressing a recombinant receptor from a modified tgfbr2 locus, related polynucleotides and methods
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
WO2020247832A1 (en) 2019-06-07 2020-12-10 Juno Therapeutics, Inc. Automated t cell culture
WO2020252218A1 (en) 2019-06-12 2020-12-17 Juno Therapeutics, Inc. Combination therapy of a cell-mediated cytotoxic therapy and an inhibitor of a prosurvival bcl2 family protein
WO2021013950A1 (en) 2019-07-23 2021-01-28 Mnemo Therapeutics Immune cells defective for suv39h1
WO2021035194A1 (en) 2019-08-22 2021-02-25 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and an enhancer of zeste homolog 2 (ezh2) inhibitor and related methods
WO2021041994A2 (en) 2019-08-30 2021-03-04 Juno Therapeutics, Inc. Machine learning methods for classifying cells
WO2021038036A1 (en) * 2019-08-28 2021-03-04 King's College London B CELL TARGETED PARALLEL CAR (pCAR) THERAPEUTIC AGENTS
WO2021043804A1 (en) 2019-09-02 2021-03-11 Institut Curie Immunotherapy targeting tumor neoantigenic peptides
US10967005B2 (en) 2013-03-15 2021-04-06 Celgene Corporation Modified T lymphocytes comprising a BAFF antibody-inducible caspase and methods of apoptosis
WO2021078910A1 (en) 2019-10-22 2021-04-29 Institut Curie Immunotherapy targeting tumor neoantigenic peptides
WO2021084050A1 (en) 2019-10-30 2021-05-06 Juno Therapeutics Gmbh Cell selection and/or stimulation devices and methods of use
WO2021092498A1 (en) 2019-11-07 2021-05-14 Juno Therapeutics, Inc. Combination of a t cell therapy and (s)-3-[4-(4-morpholin-4 ylmethyl-benzyloxy)-l-oxo-l,3-dihydro-isoindol-2-yl]- piperidine-2,6-dione
WO2021092097A1 (en) 2019-11-05 2021-05-14 Juno Therapeutics, Inc. Methods of determining attributes of therapeutic t cell compositions
WO2021113776A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to bcma-targeted binding domains and related compositions and methods
WO2021113780A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to gprc5d-targeted binding domains and related compositions and methods
WO2021113770A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Methods related to toxicity and response associated with cell therapy for treating b cell malignancies
WO2021151008A1 (en) 2020-01-24 2021-07-29 Juno Therapuetics, Inc. Methods for dosing and treatment of follicular lymphoma and marginal zone lymphoma in adoptive cell therapy
WO2021154887A1 (en) 2020-01-28 2021-08-05 Juno Therapeutics, Inc. Methods for t cell transduction
WO2021163389A1 (en) 2020-02-12 2021-08-19 Juno Therapeutics, Inc. Bcma-directed chimeric antigen receptor t cell compositions and methods and uses thereof
WO2021163391A1 (en) 2020-02-12 2021-08-19 Juno Therapeutics, Inc. Cd19-directed chimeric antigen receptor t cell compositions and methods and uses thereof
WO2021164959A1 (en) * 2020-02-17 2021-08-26 Miltenyi Biotec B.V. & Co. KG Method for providing personalized cells with chimeric antigen receptors (car) against tumor microenvironment cells
WO2021167908A1 (en) 2020-02-17 2021-08-26 Board Of Regents, The University Of Texas System Methods for expansion of tumor infiltrating lymphocytes and use thereof
US11130820B2 (en) 2012-12-20 2021-09-28 Celgene Corporation Chimeric antigen receptors
US11141436B2 (en) 2019-03-05 2021-10-12 Nkarta, Inc. Immune cells engineered to express CD19-directed chimeric antigen receptors and uses thereof in immunotherapy
WO2021207689A2 (en) 2020-04-10 2021-10-14 Juno Therapeutics, Inc. Methods and uses related to cell therapy engineered with a chimeric antigen receptor targeting b-cell maturation antigen
WO2021222330A2 (en) 2020-04-28 2021-11-04 Juno Therapeutics, Inc. Combination of bcma-directed t cell therapy and an immunomodulatory compound
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
WO2021231657A1 (en) 2020-05-13 2021-11-18 Juno Therapeutics, Inc. Methods of identifying features associated with clinical response and uses thereof
WO2021228999A1 (en) 2020-05-12 2021-11-18 Institut Curie Neoantigenic epitopes associated with sf3b1 mutations
WO2021231661A2 (en) 2020-05-13 2021-11-18 Juno Therapeutics, Inc. Process for producing donor-batched cells expressing a recombinant receptor
WO2021237068A2 (en) 2020-05-21 2021-11-25 Board Of Regents, The University Of Texas System T cell receptors with vgll1 specificity and uses thereof
WO2021260186A1 (en) 2020-06-26 2021-12-30 Juno Therapeutics Gmbh Engineered t cells conditionally expressing a recombinant receptor, related polynucleotides and methods
WO2022016119A1 (en) 2020-07-17 2022-01-20 Simurx, Inc. Chimeric myd88 receptors for redirecting immunosuppressive signaling and related compositions and methods
US11229669B2 (en) 2018-02-11 2022-01-25 Memorial Sloan-Kettering Cancer Center Cells comprising non-HLA restricted T cell receptors
WO2022023576A1 (en) 2020-07-30 2022-02-03 Institut Curie Immune cells defective for socs1
WO2022029660A1 (en) 2020-08-05 2022-02-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to ror1-targeted binding domains and related compositions and methods
US11305017B2 (en) 2015-04-25 2022-04-19 The General Hospital Corporation Anti-fugetactic agent and anti-cancer agent combination therapy and compositions for the treatment of cancer
WO2022098787A1 (en) 2020-11-04 2022-05-12 Juno Therapeutics, Inc. Cells expressing a chimeric receptor from a modified invariant cd3 immunoglobulin superfamily chain locus and related polynucleotides and methods
WO2022104109A1 (en) 2020-11-13 2022-05-19 Catamaran Bio, Inc. Genetically modified natural killer cells and methods of use thereof
WO2022115611A1 (en) 2020-11-25 2022-06-02 Catamaran Bio, Inc. Cellular therapeutics engineered with signal modulators and methods of use thereof
EP4011381A1 (en) 2016-06-03 2022-06-15 Memorial Sloan-Kettering Cancer Center Adoptive cell therapies as early treatment options
EP4012415A2 (en) 2015-12-04 2022-06-15 Juno Therapeutics, Inc. Methods and compositions related to toxicity associated with cell therapy
US11365236B2 (en) 2017-03-27 2022-06-21 Nkarta, Inc. Truncated NKG2D chimeric receptors and uses thereof in natural killer cell immunotherapy
WO2022133030A1 (en) 2020-12-16 2022-06-23 Juno Therapeutics, Inc. Combination therapy of a cell therapy and a bcl2 inhibitor
WO2022150731A1 (en) 2021-01-11 2022-07-14 Sana Biotechnology, Inc. Use of cd8-targeted viral vectors
US11400116B2 (en) 2016-05-06 2022-08-02 The Regents Of The University Of California Systems and methods for targeting cancer cells
US11401332B2 (en) 2016-08-23 2022-08-02 The Regents Of The University Of California Proteolytically cleavable chimeric polypeptides and methods of use thereof
US11408005B2 (en) 2016-12-12 2022-08-09 Seattle Children's Hospital Chimeric transcription factor variants with augmented sensitivity to drug ligand induction of transgene expression in mammalian cells
WO2022187406A1 (en) 2021-03-03 2022-09-09 Juno Therapeutics, Inc. Combination of a t cell therapy and a dgk inhibitor
WO2022189620A1 (en) 2021-03-11 2022-09-15 Institut Curie Transmembrane neoantigenic peptides
WO2022189626A2 (en) 2021-03-11 2022-09-15 Mnemo Therapeutics Tumor neoantigenic peptides
WO2022189639A1 (en) 2021-03-11 2022-09-15 Mnemo Therapeutics Tumor neoantigenic peptides and uses thereof
RU2780020C2 (en) * 2017-04-26 2022-09-19 Еурека Терапьютикс, Инк. Cells expressing chimeric activating receptors and chimeric stimulating receptors and their use
WO2022204070A1 (en) 2021-03-22 2022-09-29 Juno Therapeutics, Inc. Methods of determining potency of a therapeutic cell composition
WO2022204071A1 (en) 2021-03-22 2022-09-29 Juno Therapeutics, Inc. Method to assess potency of viral vector particles
WO2022212384A1 (en) 2021-03-29 2022-10-06 Juno Therapeutics, Inc. Combination of a car t cell therapy and an immunomodulatory compound for treatment of lymphoma
WO2022212400A1 (en) 2021-03-29 2022-10-06 Juno Therapeutics, Inc. Methods for dosing and treatment with a combination of a checkpoint inhibitor therapy and a car t cell therapy
WO2022221726A2 (en) 2021-04-16 2022-10-20 Juno Therapeutics, Inc. Combination therapies with bcma-directed t cell therapy
WO2022221737A1 (en) 2021-04-16 2022-10-20 Juno Therapeutics, Inc. T cell therapy in patients who have had prior stem cell transplant
US11479755B2 (en) 2015-12-07 2022-10-25 2Seventy Bio, Inc. T cell compositions
WO2022234009A2 (en) 2021-05-06 2022-11-10 Juno Therapeutics Gmbh Methods for stimulating and transducing t cells
WO2022238386A1 (en) 2021-05-10 2022-11-17 Institut Curie Methods for the treatment of cancer, inflammatory diseases and autoimmune diseases
WO2022248602A1 (en) 2021-05-25 2022-12-01 Institut Curie Myeloid cells overexpressing bcl2
US11517589B2 (en) 2015-02-19 2022-12-06 Myeloid Therapeutics, Inc. Chimeric antigen receptor dendritic cell (CAR-DC) for treatment of cancer
US11547694B2 (en) 2017-03-23 2023-01-10 The General Hospital Corporation CXCR4/CXCR7 blockade and treatment of human papilloma virus-associated disease
WO2023015217A1 (en) 2021-08-04 2023-02-09 Sana Biotechnology, Inc. Use of cd4-targeted viral vectors
WO2023014922A1 (en) 2021-08-04 2023-02-09 The Regents Of The University Of Colorado, A Body Corporate Lat activating chimeric antigen receptor t cells and methods of use thereof
US11623961B2 (en) 2017-11-01 2023-04-11 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for B-cell maturation antigen
US11628218B2 (en) 2020-11-04 2023-04-18 Myeloid Therapeutics, Inc. Engineered chimeric fusion protein compositions and methods of use thereof
WO2023081900A1 (en) 2021-11-08 2023-05-11 Juno Therapeutics, Inc. Engineered t cells expressing a recombinant t cell receptor (tcr) and related systems and methods
WO2023081735A1 (en) 2021-11-03 2023-05-11 Celgene Corporation Chimeric antigen receptors specific for b-cell maturation antigen for use in treating myeloma
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
US11655452B2 (en) 2015-06-25 2023-05-23 Icell Gene Therapeutics Inc. Chimeric antigen receptors (CARs), compositions and methods of use thereof
WO2023105000A1 (en) 2021-12-09 2023-06-15 Zygosity Limited Vector
WO2023115039A2 (en) 2021-12-17 2023-06-22 Sana Biotechnology, Inc. Modified paramyxoviridae fusion glycoproteins
WO2023115041A1 (en) 2021-12-17 2023-06-22 Sana Biotechnology, Inc. Modified paramyxoviridae attachment glycoproteins
WO2023126458A1 (en) 2021-12-28 2023-07-06 Mnemo Therapeutics Immune cells with inactivated suv39h1 and modified tcr
WO2023139269A1 (en) 2022-01-21 2023-07-27 Mnemo Therapeutics Modulation of suv39h1 expression by rnas
WO2023147515A1 (en) 2022-01-28 2023-08-03 Juno Therapeutics, Inc. Methods of manufacturing cellular compositions
WO2023150518A1 (en) 2022-02-01 2023-08-10 Sana Biotechnology, Inc. Cd3-targeted lentiviral vectors and uses thereof
US11738048B2 (en) 2016-08-30 2023-08-29 Memorial Sloan Kettering Cancer Center Immune cell compositions and methods of use for treating viral and other infections
WO2023164440A1 (en) 2022-02-22 2023-08-31 Juno Therapeutics, Inc. Proteinase 3 (pr3) chimeric autoantibody receptor t cells and related methods and uses
US11759480B2 (en) 2017-02-28 2023-09-19 Endocyte, Inc. Compositions and methods for CAR T cell therapy
WO2023178348A1 (en) 2022-03-18 2023-09-21 The Regents Of The University Of Colorado, A Body Corporate Genetically engineered t-cell co-receptors and methods of use thereof
WO2023180552A1 (en) 2022-03-24 2023-09-28 Institut Curie Immunotherapy targeting tumor transposable element derived neoantigenic peptides in glioblastoma
WO2023187024A1 (en) 2022-03-31 2023-10-05 Institut Curie Modified rela protein for inducing interferon expression and engineered immune cells with improved interferon expression
WO2023193015A1 (en) 2022-04-01 2023-10-05 Sana Biotechnology, Inc. Cytokine receptor agonist and viral vector combination therapies
US11779602B2 (en) 2018-01-22 2023-10-10 Endocyte, Inc. Methods of use for CAR T cells
WO2023196933A1 (en) 2022-04-06 2023-10-12 The Regents Of The University Of Colorado, A Body Corporate Chimeric antigen receptor t cells and methods of use thereof
WO2023196921A1 (en) 2022-04-06 2023-10-12 The Regents Of The University Of Colorado, A Body Corporate Granzyme expressing t cells and methods of use
WO2023211972A1 (en) 2022-04-28 2023-11-02 Medical University Of South Carolina Chimeric antigen receptor modified regulatory t cells for treating cancer
WO2023213969A1 (en) 2022-05-05 2023-11-09 Juno Therapeutics Gmbh Viral-binding protein and related reagents, articles, and methods of use
WO2023220655A1 (en) 2022-05-11 2023-11-16 Celgene Corporation Methods to overcome drug resistance by re-sensitizing cancer cells to treatment with a prior therapy via treatment with a t cell therapy
WO2023220641A2 (en) 2022-05-11 2023-11-16 Juno Therapeutics, Inc. Methods and uses related to t cell therapy and production of same
US11820819B2 (en) 2016-06-24 2023-11-21 Icell Gene Therapeutics Inc. Chimeric antigen receptors (CARs), compositions and methods thereof
EP4279085A1 (en) 2022-05-20 2023-11-22 Mnemo Therapeutics Compositions and methods for treating a refractory or relapsed cancer or a chronic infectious disease
WO2023222617A1 (en) 2022-05-16 2023-11-23 Miltenyi Biotec B.V. & Co. KG Endogenous signaling molecule activating chimeric antigen receptors and methods of generation thereof
WO2023230581A1 (en) 2022-05-25 2023-11-30 Celgene Corporation Methods of manufacturing t cell therapies
WO2023230548A1 (en) 2022-05-25 2023-11-30 Celgene Corporation Method for predicting response to a t cell therapy
US11845803B2 (en) 2017-02-17 2023-12-19 Fred Hutchinson Cancer Center Combination therapies for treatment of BCMA-related cancers and autoimmune disorders
US11851659B2 (en) 2017-03-22 2023-12-26 Novartis Ag Compositions and methods for immunooncology
WO2023250400A1 (en) 2022-06-22 2023-12-28 Juno Therapeutics, Inc. Treatment methods for second line therapy of cd19-targeted car t cells
WO2024006960A1 (en) 2022-06-29 2024-01-04 Juno Therapeutics, Inc. Lipid nanoparticles for delivery of nucleic acids
EP4302768A2 (en) 2017-06-22 2024-01-10 Board Of Regents, The University Of Texas System Methods for producing regulatory immune cells and uses thereof
US11890348B2 (en) 2015-09-18 2024-02-06 The General Hospital Corporation Localized delivery of anti-fugetactic agent for treatment of cancer
WO2024031091A2 (en) 2022-08-05 2024-02-08 Juno Therapeutics, Inc. Chimeric antigen receptors specific for gprc5d and bcma
US11896616B2 (en) 2017-03-27 2024-02-13 National University Of Singapore Stimulatory cell lines for ex vivo expansion and activation of natural killer cells
US11896614B2 (en) 2015-04-17 2024-02-13 Novartis Ag Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
WO2024044779A2 (en) 2022-08-26 2024-02-29 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for delta-like ligand 3 (dll3)
US11919937B2 (en) 2018-01-09 2024-03-05 Board Of Regents, The University Of Texas System T cell receptors for immunotherapy
WO2024054944A1 (en) 2022-09-08 2024-03-14 Juno Therapeutics, Inc. Combination of a t cell therapy and continuous or intermittent dgk inhibitor dosing
US11939389B2 (en) 2018-06-13 2024-03-26 Novartis Ag BCMA chimeric antigen receptors and uses thereof
WO2024062138A1 (en) 2022-09-23 2024-03-28 Mnemo Therapeutics Immune cells comprising a modified suv39h1 gene
US11944644B2 (en) 2017-12-05 2024-04-02 The Medical Research Infrastructure And Health Services Fund Of The Tel Aviv Medical Center T-cells comprising anti-CD38 and anti-CD138 chimeric antigen receptors and uses thereof
WO2024081820A1 (en) 2022-10-13 2024-04-18 Sana Biotechnology, Inc. Viral particles targeting hematopoietic stem cells

Families Citing this family (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015509716A (en) * 2012-02-22 2015-04-02 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 CD2 signaling domains in second generation chimeric antigen receptors
WO2014055668A1 (en) 2012-10-02 2014-04-10 Memorial Sloan-Kettering Cancer Center Compositions and methods for immunotherapy
AU2014248119B2 (en) * 2013-04-03 2019-06-20 Memorial Sloan-Kettering Cancer Center Effective generation of tumor-targeted T-cells derived from pluripotent stem cells
ES2792849T3 (en) * 2014-02-10 2020-11-12 Univ Emory Expression of chemical polypeptide with variable lymphocyte receptors in immune cells and uses to treat cancer
SG11201706041XA (en) 2015-01-26 2017-08-30 Fate Therapeutics Inc Methods and compositions for inducing hematopoietic cell differentiation
AU2016225012B2 (en) 2015-02-27 2020-09-03 Kevin Chen Chimeric antigen receptors (CARS) targeting hematologic malignancies, compositions and methods of use thereof
CN107995913B (en) 2015-05-18 2022-02-11 T细胞受体治疗公司 Compositions and methods for reprogramming TCRs using fusion proteins
CN105087495B (en) * 2015-08-21 2016-04-27 深圳市茵冠生物科技有限公司 T lymphocyte that two Chimeric antigen receptor is modified and preparation method thereof
CN105924529B (en) * 2015-10-13 2019-08-06 中国人民解放军总医院 Chimeric antigen receptor and its gene and recombinant expression carrier, CAR138-NKT cell and its preparation method and application
US10858628B2 (en) 2015-11-04 2020-12-08 Fate Therapeutics, Inc. Methods and compositions for inducing hematopoietic cell differentiation
AU2016349504B2 (en) 2015-11-04 2023-02-09 Fate Therapeutics, Inc. Genomic engineering of pluripotent cells
WO2017112877A1 (en) * 2015-12-22 2017-06-29 Icell Gene Therapeutics, Llc Chimeric antigen receptors and enhancement of anti-tumor activity
SG11201809542XA (en) * 2016-05-25 2018-12-28 Council Queensland Inst Medical Res Methods of immunotherapy
WO2018022646A1 (en) * 2016-07-25 2018-02-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods of producing modified natural killer cells and methods of use
CA3032498A1 (en) 2016-08-02 2018-02-08 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
IL302917A (en) 2016-10-07 2023-07-01 Tcr2 Therapeutics Inc Compositions and methods for t-cell receptors reprogramming using fusion proteins
EP3544996A2 (en) 2016-11-22 2019-10-02 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
CN106749677B (en) * 2017-01-04 2020-06-12 上海交通大学医学院附属瑞金医院 Bispecific chimeric antigen receptor gene targeting MLL leukemia and application thereof
CN110573529A (en) 2017-04-05 2019-12-13 韩国生命工学研究院 Fusion protein activating NK cells, NK cells and pharmaceutical composition comprising the same
CN107326014B (en) * 2017-07-31 2019-09-24 时力生物科技(北京)有限公司 A kind of T lymphocyte and its preparation method and application of bispecific chimeric antigen receptor modification
JP2021502829A (en) * 2017-11-14 2021-02-04 メモリアル スローン ケタリング キャンサー センター Immune-responsive cells secreting IL-36 and their use
US20210300986A1 (en) * 2017-12-05 2021-09-30 The Medical Research Infrastrutrure And Health Services Fund Of The Tel Aviv Medecal Center T-cells comprising two different chimeric antigen receptors and uses thereof
CN109988242A (en) * 2018-01-02 2019-07-09 武汉波睿达生物科技有限公司 Joint Chimeric antigen receptor, expression vector, slow virus and T cell
CA3095595A1 (en) * 2018-03-30 2019-10-03 Eureka Therapeutics, Inc. Constructs targeting cd22 and uses thereof
CN108864288A (en) * 2018-04-26 2018-11-23 上海怡豪生物科技有限公司 The double target spot CAR-T therapy vectors and its construction method of breast cancer and application
US20220185910A1 (en) * 2018-06-18 2022-06-16 Eureka Therapeutics, Inc. Constructs targeting prostate-specific membrane antigen (psma) and uses thereof
CN110615843B (en) * 2018-06-20 2023-05-09 上海隆耀生物科技有限公司 Chimeric antigen receptor containing third signal receptor and application thereof
WO2020049496A1 (en) * 2018-09-05 2020-03-12 Glaxosmithkline Intellectual Property Development Limited T cell modification
CA3134498A1 (en) * 2019-03-22 2020-10-01 The Regents Of The University Of California Switchable chimeric antigen receptor-engineered human natural killer cells
US11661459B2 (en) 2020-12-03 2023-05-30 Century Therapeutics, Inc. Artificial cell death polypeptide for chimeric antigen receptor and uses thereof
US11883432B2 (en) 2020-12-18 2024-01-30 Century Therapeutics, Inc. Chimeric antigen receptor system with adaptable receptor specificity
TW202306997A (en) 2021-06-16 2023-02-16 英商英斯特生物科技有限公司 Receptors providing targeted costimulation for adoptive cell therapy
WO2023288278A1 (en) 2021-07-16 2023-01-19 Instil Bio (Uk) Limited Chimeric molecules providing targeted costimulation for adoptive cell therapy
WO2023019213A1 (en) * 2021-08-13 2023-02-16 Simcere Innovation, Inc. Three-dimensional culturing of pluripotent stem cells to produce hematopoietic stem cells
CN115058387B (en) * 2022-06-11 2023-12-01 重庆医科大学 Method for preparing anti-prostate cancer medicine by co-culturing human embryonic stem cells and prostate cancer cells
CN117736335A (en) * 2022-09-20 2024-03-22 深圳先进技术研究院 Double-targeting CAR-T cell targeting mesothelin and NKG2D ligand and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
WO2008121420A1 (en) * 2007-03-30 2008-10-09 Memorial Sloan-Kettering Cancer Center Constitutive expression of costimulatory ligands on adoptively transferred t lymphocytes
WO2012109624A2 (en) * 2011-02-11 2012-08-16 Zyngenia, Inc. Monovalent and multivalent multispecific complexes and uses thereof

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5712149A (en) 1995-02-03 1998-01-27 Cell Genesys, Inc. Chimeric receptor molecules for delivery of co-stimulatory signals
GB9605515D0 (en) 1996-03-15 1996-05-15 Medical Res Council Improvements in or relating to cell stimulation
US7446190B2 (en) * 2002-05-28 2008-11-04 Sloan-Kettering Institute For Cancer Research Nucleic acids encoding chimeric T cell receptors
US7629440B2 (en) * 2002-08-20 2009-12-08 Genitrix, Llc Lectin compositions and methods for modulating an immune response to an antigen
US20050129671A1 (en) 2003-03-11 2005-06-16 City Of Hope Mammalian antigen-presenting T cells and bi-specific T cells
US20050113564A1 (en) * 2003-11-05 2005-05-26 St. Jude Children's Research Hospital Chimeric receptors with 4-1BB stimulatory signaling domain
US8315214B2 (en) 2007-05-18 2012-11-20 Research In Motion Limited Method and system for discontinuous reception de-synchronization detection
GB201108236D0 (en) * 2011-05-17 2011-06-29 Ucl Business Plc Method
ES2723181T3 (en) * 2011-07-29 2019-08-22 Univ Pennsylvania Costimulatory switching receivers
WO2014055668A1 (en) * 2012-10-02 2014-04-10 Memorial Sloan-Kettering Cancer Center Compositions and methods for immunotherapy
US10117896B2 (en) * 2012-10-05 2018-11-06 The Trustees Of The University Of Pennsylvania Use of a trans-signaling approach in chimeric antigen receptors
EP3569712A1 (en) 2012-11-09 2019-11-20 DSM IP Assets B.V. Process for enzymatic hydrolysis of lignocellulosic material and fermentation of sugars
NZ750426A (en) 2013-02-06 2020-06-26 Celgene Corp Modified t lymphocytes having improved specificity
EP2970426B1 (en) 2013-03-15 2019-08-28 Michael C. Milone Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
WO2008121420A1 (en) * 2007-03-30 2008-10-09 Memorial Sloan-Kettering Cancer Center Constitutive expression of costimulatory ligands on adoptively transferred t lymphocytes
WO2012109624A2 (en) * 2011-02-11 2012-08-16 Zyngenia, Inc. Monovalent and multivalent multispecific complexes and uses thereof

Non-Patent Citations (96)

* Cited by examiner, † Cited by third party
Title
"REMINGTON'S PHARMACEUTICAL SCIENCE", 1985
"Sequence Analysis Software Package of the Genetics Computer Group", UNIVERSITY OF WISCONSIN BIOTECHNOLOGY CENTER
"The Cambridge Dictionary of Science and Technology", 1988
"The Glossary of Genetics", 1991, SPRINGER VERLAG
ANDERSON, SCIENCE, vol. 226, 1984, pages 401 - 409
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 2001, WILEY INTERSCIENCE
AUSUBEL, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, 1987
BENTON; DAVIS, SCIENCE, vol. 196, 1977, pages 180
BERGER; KIMMEL: "Guide to Molecular Cloning Techniques", 1987, ACADEMIC PRESS
BLOOMER ET AL., JOURNAL OF VIROLOGY, vol. 71, 1997, pages 6641 - 6649
BREGNI ET AL., BLOOD, vol. 80, 1992, pages 1418 - 1422
BRENTJENS, R.J. ET AL.: "Eradication of systemic B-ce!l tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15", NATURE MEDICINE, vol. 9, 2003, pages 279 - 286, XP002389127, DOI: doi:10.1038/nm827
BRENTJENS, R.J. ET AL.: "Safety and persistence of adoptively transferred autologous CD19- ta rgeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias", BLOOD, vol. 118, 2011, pages 4817 - 4828, XP055169039, DOI: doi:10.1182/blood-2011-04-348540
BRIGHAM ET AL., AM. J. MED. SCI., vol. 298, 1989, pages 278
CA RPE N ITO, C . ET AL.: "Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 106, 2009, pages 3360 - 3365
CAYOUETTE ET AL., HUMAN GENE THERAPY, vol. 8, 1997, pages 423 - 430
COLIGAN, CURRENT PROTOCOLS IN IMMUNOLOGY, 1991
CORNETTA ET AL., NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY, vol. 36, 1987, pages 311 - 322
DANOS ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 6460 - 6464
DUPONT, J. ET AL., CANCERRES, vol. 65, 2005, pages 5417 - 5427
EGLITIS ET AL., BIOTECHNIQUES, vol. 6, 1988, pages 608 - 614
FEIGNER ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 84, 1987, pages 7413
FRESHNEY: "Animal Cell Culture", 1987
FRIEDMAN, SCIENCE, vol. 244, 1989, pages 1275 - 1281
GAIT: "Oligonucleotide Synthesis", 1984
GRUNSTEIN; HOGNESS, PROC. NATL. ACAD.SCI., USA, vol. 72, 1975, pages 3961
HALE; MARHAM, THE HARPER COLLINS DICTIONARY OF BIOLOGY, 1991
HO, W.Y.; BLATTMAN, J.N.; DOSSETT, M.L.; YEE, C.; GREENBERG, P.O.: "Adoptive immunotherapy: engineering T cell responses as biologic weapons for tumor mass destruction", CANCER CELL, vol. 3, 2003, pages 431 - 437, XP002409705, DOI: doi:10.1016/S1535-6108(03)00113-2
HUGHES ET AL., J. CLIN. INVEST., vol. 89, 1992, pages 1817
JOHNSON, CHEST, vol. 107, 1995, pages 77S - 83S
JOHNSON, L.A. ET AL.: "Gene therapy with human and mouse T, cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen", BLOOD, vol. 114, 2009, pages 535 - 546
JORRITSMA, A.; SCHOTTE, R.; COCCORIS, M.; DE WITTE, M.A.; SCHUMACHER, T.N.: "Prospects and limitations of T cell receptor gene therapy", CURRENT GENE THERAPY, vol. 11, 2011, pages 276 - 287
KALOS, M. ET AL.: "T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia", SCI TRANSL MED, vol. 3, 2011, pages 95ra73, XP002667262, DOI: doi:10.1126/scitranslmed.3002842
KIDO ET AL., CURRENT EYE RESEARCH, vol. 15, 1996, pages 833 - 844
KIMMEL, A. R., METHODS ENZYMOL., vol. 152, 1987, pages 507
KOCHENDERFER, J.N. ET AL.: "B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor- transduced T cells", BLOOD, vol. 119, 2012, pages 2709 - 2720, XP055145503, DOI: doi:10.1182/blood-2011-10-384388
KOWOLIK, C ET AL.: "CD28 Costimulation Provided Through A CD19-Specific Chimeric Antigen Receptor Enhances In Vivo Persistence And Antitumor Efficacy Of Adoptively Transferred T Cells.", CANCER RESEARCH., vol. 66, 15 November 2006 (2006-11-15), pages 10995 - 11004, XP055044266 *
KRAUSE, A. ET AL.: "Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes", J EXP MED, vol. 188, 1998, pages 619 - 626, XP001240431, DOI: doi:10.1084/jem.188.4.619
LAM, J.S. ET AL.: "Prostate stem cell antigen is overexpressed in prostate cancer metastases", CLINICAL CANCER RESEARCH : AN OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH, vol. 11, 2005, pages 2591 - 2596, XP002494615, DOI: doi:10.1158/1078-0432.CCR-04-1842
LE GAL LA SALLE ET AL., SCIENCE, vol. 259, 1993, pages 988 - 990
LIAO, W.; LIN, J .X; LEONARD, W.J: "I L-2 family cytokines: new insights into the complex roles of IL-2 as a broad regulator of T helper cell differentiation", CURRENT OPINION IN IMMUNOLOGY, vol. 23, 2011, pages 598 - 604, XP028309071, DOI: doi:10.1016/j.coi.2011.08.003
LIU, J.C. ET AL.: "Seventeen-gene signature from enriched Her2/Neu mammary tumor- initiating cells predicts clinical outcome for human HER2+:ERalpha- breast cancer", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 109, 2012, pages 58325837
MAGEE, J.A.; PISKOUNOVA, E.; MORRISON, S.J.: "Cancer stem cells: impact, heterogeneity, and uncertainty", CANCER CELL, vol. 21, 2012, pages 283 - 296, XP055023489, DOI: doi:10.1016/j.ccr.2012.03.003
MAHER, J.; BRENTJENS, R.J.; GUNSET, G.; RIVIERE, I.; SADELAIN, M.: "Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor", NAT BIO TECHNOL, vol. 20, 2002, pages 70 - 75, XP002588277
MATSUI, K ET AL.: "Low Affinity Interaction Of Peptide-MHC Complexes With T Cell Receptors.", SCIENCE., vol. 254, 20 December 1991 (1991-12-20), pages 1788 - 1791, XP001094133 *
MEYER, M.J. ET AL.: "CD44posCD49fhiCD133/2hi defines xenograft-initiating cells in estrogen receptor-negative breast cancer", CANCER RESEARCH, vol. 70, 2010, pages 4624 - 4633
MILLER ET AL., BIOTECHNOLOGY, vol. 7, 1989, pages 980 - 990
MILLER ET AL., MOL. CELL. BIOL., vol. 5, 1985, pages 431 - 437
MILLER ET AL., MOL. CELL. BIOL., vol. 6, 1986, pages 2895 - 2902
MILLER, HUMAN GENE THERAPY, 1990, pages 15 - 14
MILLER; CALES, GENE TRANSFER VECTORS FOR MAMMALIAN CELLS, 1987
MIYOSHI ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 94, 1997, pages 10319
MOEN, BLOOD CELLS, vol. 17, 1991, pages 407 - 416
MORGAN, R.A. ET AL., SCIENCE, vol. 314, 2006, pages 126 - 129
MORGAN, R.A. ET AL.: "Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2", MOLECULAR THERAPY: THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY, vol. 18, 2010, pages 843 - 851, XP055023624, DOI: doi:10.1038/mt.2010.24
MULLIS, PCR: THE POLYMERASE CHAIN REACTION, 1994
NALDINI ET AL., SCIENCE, vol. 272, 1996, pages 263 - 267
NAM, K. 0 .; SHIN, S . M .; LEE, H.W.: "Cross-linking of 4-IBB up-regulates IL-13 expression in CDS(+) T lymphocytes", CYTOKINE, vol. 33, 2006, pages 87 - 94
NGUYEN, L.V.; VANNER, R.; DIRKS, P.; EAVES, C.J.: "Cancer stem cells: an evolving concept. Nature reviews", CANCER, vol. 12, 2012, pages 133 - 143
OLSON, W.C.; HESTON, W.D.; RAJASEKARAN, A.K.: "Clinical trials of cancer therapies targeting prostate-specific membrane antigen", REVIEWS ON RECENT CLINICAL TRIALS, vol. 2, no. 30, 2007, pages 182 - 190, XP007922264, DOI: doi:10.2174/157488707781662724
ONO ET AL., NEUROSCIENCE LETTERS, vol. 17, 1990, pages 259
ORLANDI, R.; GUSSOW, D.H.; JONES, P.T.; WINTER, G.: "Cloning immunoglobulin variable domains for expression by the polymerase chain reaction", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 86, 1989, pages 3833 - 3837, XP000026475, DOI: doi:10.1073/pnas.86.10.3833
PANELLI, M.C. ET AL., J IMMUNO, vol. 164, 2000, pages 4382 - 4392
PANELLI, M.C. ET AL., J IMMUNOL, vol. 164, 2000, pages 495 - 504
PAPANICOLAOU, G.A. ET AL., BLOOD, vol. 102, 2003, pages 2498 - 2505
ROBBINS, P.F. ET AL.: "Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1", JOURNAL OF CLINICAL ONCOLOGY: OFFICIAL JOURNAL OF THE AMERICAN SOCIETY OF CLINICAL ONCOLOGY, vol. 29, 2011, pages 917 - 924, XP002705979, DOI: doi:10.1200/JCO.2010.32.2537
ROSENBERG ET AL., N. ENGL. J. MED, vol. 323, 1990, pages 370
ROSENBERG, S.A.; RESTIFO, N.P.; YANG, J.C.; MORGAN, R.A.; DUDLEY, M.E.: "Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nature reviews", CANCER, vol. 8, 2008, pages 299 - 308
SADELAIN, M. ET AL., NAT REV CANCER, vol. 3, 2003, pages 35 - 45
SADELAIN, M.; BRENTJENS, R.; RIVIERE, I.: "The promise and potential pitfalls of chimeric antigen receptors", CURR OPIN IMMUNOL, vol. 21, 2009, pages 215 - 223, XP026058399, DOI: doi:10.1016/j.coi.2009.02.009
SADELAIN, M.; RIVIERE, I.; BRENTJENS, R.: "Targeting tumours with genetically enhanced T lymphocytes", NAT REV CANCER, vol. 3, 2003, pages 35 - 45, XP009026020, DOI: doi:10.1038/nrc971
SAEKI, N.; GU, J.; YOSHIDA, T.; WU, X.: "Prostate stem cell antigen: a Jekyll and Hyde molecule?", CLINICAL CANCER RESEARCH :AN OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH, vol. 16, 2010, pages 3533 - 3538
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", COLD SPRING HARBOR LABORATORY PRESS
SAMBROOK: "Molecular Cloning: A Laboratory Manual", 1989
SAMBROOK; FRITSCH; MANIATIS: "Molecular Cloning: A Laboratory Manual", 1989, CSH PRESS
SCHWARTZ, R . H . T: "cell a n e rgy", ANNUAL REVIEW OF IMMUNOLOGY, vol. 21, 2003, pages 305 - 334
SHARP, THE LANCET, vol. 337, 1991, pages 1277 - 1278
SHIHLE, M .; DAVIDSON, B.: "Pathogenesis of ovarian cancer: clues from selected overexpressed genes", FUTURE ONCOL, vol. 5, 2009, pages 1641 - 1657
SHIN, S M: "1.4- IBB triggers IL-13 production from T cells to limit the polarized, Thl-mediated inflammation", JOURNAL OF LEUKOCYTE BIOLOGY, vol. 81, 2007, pages 1455 - 1465
SILVER, D.A.; PELLICER, I.; FAIR, W.R.; HESTON, W.D.; CORDON-CARDO, C.: "Prostate-specific membrane antigen expression in normal and malignant human tissues", CLINICAL CANCER RESEARCH : AN OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH, vol. 3, 1997, pages 81 - 85, XP000941663
SINGLETON ET AL.: "Dictionary of Microbiology and Molecular Biology", 1994
STAUBINGER ET AL., METHODS IN ENZYMOLOGY, vol. 101, 1983, pages 512
STEPHAN, M .T. ET AL.: "T cell-encoded CD80 and 4-1 BBL induce auto-and transcostimulation, resulting in potent tumor rejection", NATURE MEDICINE, vol. 13, 2007, pages 1440 - 1449, XP002571429, DOI: doi:10.1038/nm1676
STRAUSS, R. ET AL.: "Analysis of epithelial and mesenchymal markers in ovarian cancer reveals phenotypic heterogeneity and plasticity", PLOS ONE, vol. 6, 2011, pages el 6186
TAMMANA, S.: "1. 4-1 BB and CD28 signaling plays a synergistic role in redirecting umbilical cord blood T cells against B-cell malignancies", HUM GENE THER, vol. 2, no. 1, 2000, pages 75 - 86
TOLSTOSHEV ET AL., CURRENT OPINION IN BIOTECHNOLOGY, vol. 1, 1990, pages 55 - 61
WAHL, G. M.; S. L. BERGER, METHODS ENZYMOL., vol. 152, 1987, pages 399
WANG, J. ET AL.: "Optimizing adoptive polyclonal T cell immunotherapy of lymphomas, using a chimeric T cell receptor possessing CD28 and CD137 costimulatory domains", HUMAN GENE THERAPY, vol. 18, 2007, pages 712 - 725, XP002605938, DOI: doi:10.1089/HUM.2007.028
WATTS, T.H.: "TNF/TNFR family members in costimulation of T cell responses", ANNUAL REVIEW OF IMMUNOLOGY, vol. 23, 2005, pages 23 - 68, XP008051583, DOI: doi:10.1146/annurev.immunol.23.021704.115839
WEIR: "Handbook of Experimental Immunology", 1996, article "Methods in Enzymology"
WILKIE, S. ET AL.: "Dual Targeting of ErbB2 and MUC1 in Breast Cancer Using Chimeric Antigen Receptors Engineered to Provide Complementary Signaling", JOURNAL OF CLINICAL IMMUNOLOGY, 2012
WOLFF ET AL., SCIENCE, vol. 247, 1990, pages 1465
WU ET AL., JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 263, 1988, pages 14621
WU ET AL., JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 264, 1989, pages 16985
XU ET AL., EXP. HEMAT., vol. 22, 1994, pages 223 - 230
ZHONG, X.S.; MATSUSHITA, M.; PLOTKIN, J.; RIVIERE, I.; SADELAIN, M.: "Chimeric antigen receptors combining 4-1 BB and CD28 signaling domains augment PI3kinase/AKT/Bc1-X L activation and C D8+ T cell-mediated tumor eradication", MOLECULAR THERAPY: THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY, vol. 18, 2010, pages 413 - 420, XP055023623, DOI: doi:10.1038/mt.2009.210

Cited By (390)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11484552B2 (en) 2012-10-05 2022-11-01 The Trustees Of The University Of Pennsylvania Use of trans-signaling approach in chimeric antigen receptors
US10117896B2 (en) 2012-10-05 2018-11-06 The Trustees Of The University Of Pennsylvania Use of a trans-signaling approach in chimeric antigen receptors
US11130820B2 (en) 2012-12-20 2021-09-28 Celgene Corporation Chimeric antigen receptors
US10829737B2 (en) 2013-01-28 2020-11-10 St. Jude Children's Research Hospital, Inc. Chimeric receptor with NKG2D specificity for use in cell therapy against cancer and infectious disease
US10836999B2 (en) 2013-01-28 2020-11-17 St. Jude Children's Research Hospital, Inc. Chimeric receptor with NKG2D specificity for use in cell therapy against cancer and infectious disease
US10774309B2 (en) 2013-01-28 2020-09-15 St. Jude Children's Research Hospital, Inc. Natural killer cell immunotherapy for treating cancer
US10538739B2 (en) 2013-01-28 2020-01-21 St. Jude Children's Research Hospital, Inc. Chimeric receptor with NKG2D specificity for use in cell therapy against cancer and infectious disease
US11873512B2 (en) 2013-01-28 2024-01-16 St. Jude Children's Research Hospital, Inc. Chimeric receptor with NKG2D specificity for use in cell therapy against cancer and infectious disease
US10801012B2 (en) 2013-01-28 2020-10-13 St. Jude Children's Research Hospital, Inc. Chimeric receptor with NKG2D specificity for use in cell therapy against cancer and infectious disease
AU2014214850C1 (en) * 2013-02-06 2018-12-06 Celgene Corporation Modified T lymphocytes having improved specificity
AU2014214850B2 (en) * 2013-02-06 2018-06-14 Celgene Corporation Modified T lymphocytes having improved specificity
WO2014134165A1 (en) * 2013-02-26 2014-09-04 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
US11103531B2 (en) 2013-02-26 2021-08-31 Memorial Sloan-Kettering Cancer Center Compositions and methods for immunotherapy
US11806365B2 (en) 2013-03-15 2023-11-07 Celgene Corporation Modified T lymphocytes comprising a CD52 antibody-inducible caspase and methods of apoptosis
US10967005B2 (en) 2013-03-15 2021-04-06 Celgene Corporation Modified T lymphocytes comprising a BAFF antibody-inducible caspase and methods of apoptosis
US10774343B2 (en) 2014-04-25 2020-09-15 Bluebird Bio, Inc. MND promoter chimeric antigen receptors
US10428305B2 (en) 2014-05-15 2019-10-01 National University Of Singapore Modified natural killer cells that express IL15 and uses thereof
US10774311B2 (en) 2014-05-15 2020-09-15 National University Of Singapore Natural killer cells modified to express membrane-bound interleukin 15 and uses thereof
US11560548B2 (en) 2014-05-15 2023-01-24 National University Of Singapore Immune cells expressing membrane-bound interleukin 15 (mbIL15) and uses thereof
CN111394317A (en) * 2014-06-06 2020-07-10 蓝鸟生物公司 Improved T cell compositions
AU2015269197B2 (en) * 2014-06-06 2018-10-04 2Seventy Bio, Inc. Improved T cell compositions
US10479975B2 (en) 2014-06-06 2019-11-19 Bluebird Bio, Inc. Methods of making T cell compositions
JP2020072756A (en) * 2014-06-06 2020-05-14 ブルーバード バイオ, インコーポレイテッド Improved t-cell composition
WO2015188119A1 (en) * 2014-06-06 2015-12-10 Bluebird Bio, Inc. Improved t cell compositions
CN106793780A (en) * 2014-06-06 2017-05-31 蓝鸟生物公司 The T cell composition of improvement
EP4166148A1 (en) 2014-06-06 2023-04-19 Memorial Sloan-Kettering Cancer Center Mesothelin-targeted chimeric antigen receptors and uses thereof
WO2015188141A2 (en) 2014-06-06 2015-12-10 Memorial Sloan-Kettering Cancer Ceneter Mesothelin-targeted chimeric antigen receptors and uses thereof
US11560547B2 (en) 2014-06-06 2023-01-24 2Seventy Bio, Inc. Methods of making T cell compositions
JP2017518052A (en) * 2014-06-06 2017-07-06 ブルーバード バイオ, インコーポレイテッド Improved T cell composition
EP3685842A1 (en) 2014-06-06 2020-07-29 Memorial Sloan-Kettering Cancer Center Mesothelin-targeted chimeric antigen receptors and uses thereof
US10738278B2 (en) 2014-07-15 2020-08-11 Juno Therapeutics, Inc. Engineered cells for adoptive cell therapy
US10851166B2 (en) 2014-07-21 2020-12-01 Novartis Ag Treatment of cancer using a CD33 chimeric antigen receptor
US9777061B2 (en) 2014-07-21 2017-10-03 Novartis Ag Treatment of cancer using a CD33 chimeric antigen receptor
US9546206B2 (en) 2014-08-08 2017-01-17 The Board Of Trustees Of The Leland Stanford Junior University High affinity PD-1 agents and methods of use
US11814419B2 (en) 2014-08-08 2023-11-14 The Board Of Trustees Of The Leland Stanford Junior University High affinity PD-1 agents and methods of use
US9562087B2 (en) 2014-08-08 2017-02-07 The Board Of Trustees Of The Leland Stanford Junior University High affinity PD-1 agents and methods of use
US10533055B2 (en) 2014-08-28 2020-01-14 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for CD19
WO2016033570A1 (en) 2014-08-28 2016-03-03 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for cd19
EP3805267A1 (en) 2014-08-28 2021-04-14 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for cd19
US11827714B2 (en) 2014-08-28 2023-11-28 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for CD19
CN106574272A (en) * 2014-08-29 2017-04-19 哥莫阿波单克隆有限责任公司 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
CN106574272B (en) * 2014-08-29 2022-04-29 哥莫阿波有限责任公司 Universal chimeric antigen receptor expressing immune cells targeting diverse antigens, methods of making and use thereof in the treatment of cancer, infection and autoimmune disease
US11633426B2 (en) 2014-10-20 2023-04-25 Juno Therapeutics, Inc. Methods and compositions for dosing in adoptive cell therapy
WO2016064929A1 (en) 2014-10-20 2016-04-28 Juno Therapeutics, Inc. Methods and compositions for dosing in adoptive cell therapy
EP3932950A1 (en) 2014-10-20 2022-01-05 Juno Therapeutics, Inc. Methods and compositions for dosing in adoptive cell therapy
US10507219B2 (en) 2014-10-20 2019-12-17 Juno Therapeutics, Inc. Methods and compositions for dosing in adoptive cell therapy
JP7372728B2 (en) 2014-10-31 2023-11-01 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Methods and compositions related to modified T cells
JP2017533706A (en) * 2014-10-31 2017-11-16 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Methods and compositions for modified T cells
US10428351B2 (en) 2014-11-05 2019-10-01 Juno Therapeutics, Inc. Methods for transduction and cell processing
WO2016073602A2 (en) 2014-11-05 2016-05-12 Juno Therapeutics, Inc. Methods for transduction and cell processing
EP3757206A1 (en) 2014-11-05 2020-12-30 Juno Therapeutics, Inc. Methods for transduction and cell processing
US11802295B2 (en) 2014-11-05 2023-10-31 Juno Therapeutics, Inc. Methods for transduction and cell processing
US11266739B2 (en) 2014-12-03 2022-03-08 Juno Therapeutics, Inc. Methods and compositions for adoptive cell therapy
WO2016090190A1 (en) 2014-12-03 2016-06-09 Juno Therapeutics, Inc. Methods and compositions for adoptive cell therapy
EP3766895A1 (en) 2014-12-03 2021-01-20 Juno Therapeutics, Inc. Methods and compositions for adoptive cell therapy
US11382965B2 (en) 2014-12-12 2022-07-12 2Seventy Bio, Inc. BCMA chimeric antigen receptors
US10624960B2 (en) 2014-12-12 2020-04-21 Bluebird Bio, Inc. BCMA chimeric antigen receptors
US11351236B2 (en) 2014-12-12 2022-06-07 2Seventy Bio, Inc. BCMA chimeric antigen receptors
US11633463B2 (en) 2014-12-12 2023-04-25 2Seventy Bio, Inc. BCMA chimeric antigen receptors
US10646558B2 (en) 2014-12-12 2020-05-12 Bluebird Bio, Inc. BCMA chimeric antigen receptors
US10383929B2 (en) 2014-12-12 2019-08-20 Bluebird Bio, Inc. BCMA chimeric antigen receptors
US10639358B2 (en) 2014-12-12 2020-05-05 Bluebird Bio, Inc. BCMA chimeric antigen receptors
US11020466B2 (en) 2014-12-12 2021-06-01 Bluebird Bio, Inc. BCMA chimeric antigen receptors
US10639359B2 (en) 2014-12-12 2020-05-05 Bluebird Bio, Inc. BCMA chimeric antigen receptors
US11091532B2 (en) 2014-12-24 2021-08-17 Autolus Limited T cell which co-expresses a CD19 chimeric antigen receptor and a CD22 chimeric antigen receptor
RU2768019C2 (en) * 2014-12-24 2022-03-23 Отолус Лимитед Cell
US11034750B2 (en) 2014-12-24 2021-06-15 Autolus Limited Chimeric antigen receptor (CAR) comprising a CD19-binding domain
CN107002045A (en) * 2014-12-24 2017-08-01 Ucl商务股份有限公司 Cell
US10981970B2 (en) 2014-12-24 2021-04-20 Autolus Limited Chimeric antigen receptor (CAR) comprising a CD22-binding domain
EP3237442B1 (en) 2014-12-24 2019-07-10 UCL Business PLC Cell
WO2016115177A1 (en) 2015-01-12 2016-07-21 Juno Therapeutics, Inc. Modified hepatitis post-transcriptional regulatory elements
US10363269B2 (en) 2015-01-12 2019-07-30 Juno Therapeutics, Inc. Modified hepatitis post-transcriptional regulatory elements
EP3760644A1 (en) 2015-01-16 2021-01-06 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ror1
US11919970B2 (en) 2015-01-16 2024-03-05 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ROR1
US10889652B2 (en) 2015-01-16 2021-01-12 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ROR1
WO2016115559A1 (en) 2015-01-16 2016-07-21 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ror1
US11918604B2 (en) 2015-02-19 2024-03-05 Myeloid Therapeutics, Inc. Chimeric antigen receptor dendritic cell (CAR-DC) for treatment of cancer
US11517589B2 (en) 2015-02-19 2022-12-06 Myeloid Therapeutics, Inc. Chimeric antigen receptor dendritic cell (CAR-DC) for treatment of cancer
US11918605B1 (en) 2015-02-19 2024-03-05 Myeloid Therapeutics, Inc. Chimeric antigen receptor dendritic cell (CAR-DC) for treatment of cancer
WO2016138034A1 (en) * 2015-02-24 2016-09-01 The Regents Of The University Of California Binding-triggered transcriptional switches and methods of use thereof
US9834608B2 (en) 2015-02-24 2017-12-05 The Regents Of The University Of California Binding-triggered transcriptional switches and methods of use thereof
US10822387B2 (en) 2015-02-24 2020-11-03 The Regents Of The University Of California Binding-triggered transcriptional switches and methods of use thereof
US10590182B2 (en) 2015-02-24 2020-03-17 The Regents Of The University Of California Binding-triggered transcriptional switches and methods of use thereof
US9670281B2 (en) 2015-02-24 2017-06-06 The Regents Of The University Of California Binding-triggered transcriptional switches and methods of use thereof
US10836808B2 (en) 2015-02-24 2020-11-17 The Regents Of The University Of California Binding-triggered transcriptional switches and methods of use thereof
CN107922488A (en) * 2015-04-02 2018-04-17 纪念斯隆-凯特琳癌症中心 TNFRSF14/HVEM albumen and its application method
WO2016168493A1 (en) * 2015-04-15 2016-10-20 Prospect CharterCare RWMC, LLC d/b/a Roger Williams Medical Center Hepatic arterial infusion of car-t cells
US10471098B2 (en) 2015-04-15 2019-11-12 Prospect Chartercare Rwmc, Llc Hepatic arterial infusion of CAR-T cells
WO2016166568A1 (en) 2015-04-16 2016-10-20 Juno Therapeutics Gmbh Methods, kits and apparatus for expanding a population of cells
US11896614B2 (en) 2015-04-17 2024-02-13 Novartis Ag Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
US10786549B2 (en) 2015-04-23 2020-09-29 Baylor College Of Medicine CD5 chimeric antigen receptor for adoptive T cell therapy
WO2016172606A1 (en) * 2015-04-23 2016-10-27 Baylor College Of Medicine Cd5 chimeric antigen receptor for adoptive t cell therapy
US11305017B2 (en) 2015-04-25 2022-04-19 The General Hospital Corporation Anti-fugetactic agent and anti-cancer agent combination therapy and compositions for the treatment of cancer
WO2016196388A1 (en) 2015-05-29 2016-12-08 Juno Therapeutics, Inc. Composition and methods for regulating inhibitory interactions in genetically engineered cells
EP4272757A3 (en) * 2015-06-10 2023-12-27 ImmunityBio, Inc. Modified nk-92 cells for treating cancer
EP3307876A4 (en) * 2015-06-10 2019-04-03 Nantkwest, Inc. Modified nk-92 cells for treating cancer
US10738279B2 (en) 2015-06-10 2020-08-11 Nantkwest, Inc. Modified NK-92 cells for treating cancer
US11788059B2 (en) 2015-06-10 2023-10-17 Immunity Bio, Inc. Modified NK-92 cells for treating cancer
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
US11655452B2 (en) 2015-06-25 2023-05-23 Icell Gene Therapeutics Inc. Chimeric antigen receptors (CARs), compositions and methods of use thereof
US10786533B2 (en) 2015-07-15 2020-09-29 Juno Therapeutics, Inc. Engineered cells for adoptive cell therapy
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
US10865231B2 (en) 2015-07-31 2020-12-15 King's College London Therapeutic agents
RU2747733C1 (en) * 2015-07-31 2021-05-13 Кинг'С Колледж Лондон Therapeutic agents
WO2017021701A1 (en) * 2015-07-31 2017-02-09 King's College London Therapeutic agents
US11802143B2 (en) 2015-07-31 2023-10-31 King's College London Therapeutic agents
CN107735407B (en) * 2015-07-31 2022-08-16 伦敦国王学院 Therapeutic agents
JP7053037B2 (en) 2015-07-31 2022-04-12 キングス カレッジ ロンドン Therapeutic drug
JP2021168675A (en) * 2015-07-31 2021-10-28 キングス カレッジ ロンドンKings College London Therapeutic agent
US10703794B2 (en) 2015-07-31 2020-07-07 King's College London Therapeutic agents
CN107735407A (en) * 2015-07-31 2018-02-23 伦敦国王学院 therapeutic agent
US10899818B2 (en) 2015-07-31 2021-01-26 King's College London Therapeutic agents
US10730941B2 (en) 2015-07-31 2020-08-04 Memorial Sloan-Kettering Cancer Center Antigen-binding proteins targeting CD56 and uses thereof
EP3939992A1 (en) * 2015-07-31 2022-01-19 King's College London Therapeutic agents
JP2018521663A (en) * 2015-07-31 2018-08-09 キングス カレッジ ロンドンKings College London Remedy
EP3328994A4 (en) * 2015-07-31 2019-04-17 Memorial Sloan-Kettering Cancer Center Antigen-binding proteins targeting cd56 and uses thereof
JP7162530B2 (en) 2015-08-07 2022-10-28 シアトル チルドレンズ ホスピタル (ディービーエイ シアトル チルドレンズ リサーチ インスティテュート) Bispecific CAR T cells targeting solid tumors
US11458167B2 (en) 2015-08-07 2022-10-04 Seattle Children's Hospital Bispecific CAR T-cells for solid tumor targeting
US11123369B2 (en) 2015-08-07 2021-09-21 Seattle Children's Hospital Bispecific CAR T-cells for solid tumor targeting
JP2018522567A (en) * 2015-08-07 2018-08-16 シアトル チルドレンズ ホスピタル, ディービーエー シアトル チルドレンズ リサーチ インスティテュート Bispecific CAR T cells targeting solid tumors
US11242375B2 (en) 2015-09-04 2022-02-08 Memorial Sloan Kettering Cancer Center Immune cell compositions and methods of use
WO2017040945A1 (en) 2015-09-04 2017-03-09 Memorial Sloan Kettering Cancer Center Immune cell compositions and methods of use
US11890348B2 (en) 2015-09-18 2024-02-06 The General Hospital Corporation Localized delivery of anti-fugetactic agent for treatment of cancer
EP3662930A1 (en) 2015-09-24 2020-06-10 AbVitro LLC Hiv antibody compositions and methods of use
WO2017053906A1 (en) 2015-09-24 2017-03-30 Abvitro Llc Hiv antibody compositions and methods of use
WO2017068425A1 (en) 2015-10-22 2017-04-27 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
WO2017068419A2 (en) 2015-10-22 2017-04-27 Juno Therapeutics Gmbh Methods, kits, agents and apparatuses for transduction
US11466253B2 (en) 2015-10-22 2022-10-11 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
US11913024B2 (en) 2015-10-22 2024-02-27 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
WO2017068421A1 (en) 2015-10-22 2017-04-27 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
US11248238B2 (en) 2015-10-22 2022-02-15 Juno Therapeutics Gmbh Methods, kits, agents and apparatuses for transduction
US11421013B2 (en) 2015-10-23 2022-08-23 Eureka Therapeutics, Inc. Antibody/T-cell receptor chimeric constructs and uses thereof
US10822389B2 (en) 2015-10-23 2020-11-03 Eureka Therapeutics, Inc. Antibody/T-cell receptor chimeric constructs and uses thereof
US10464988B2 (en) 2015-10-23 2019-11-05 Eureka Therapeutics, Inc. Antibody/T-cell receptor chimeric constructs and uses thereof
WO2017079705A1 (en) 2015-11-05 2017-05-11 Juno Therapeutics, Inc. Chimeric receptors containing traf-inducing domains and related compositions and methods
WO2017079703A1 (en) 2015-11-05 2017-05-11 Juno Therapeutics, Inc. Vectors and genetically engineered immune cells expressing metabolic pathway modulators and uses in adoptive cell therapy
US11020429B2 (en) 2015-11-05 2021-06-01 Juno Therapeutics, Inc. Vectors and genetically engineered immune cells expressing metabolic pathway modulators and uses in adoptive cell therapy
EP4212547A1 (en) 2015-12-03 2023-07-19 Juno Therapeutics, Inc. Modified chimeric receptors and related compositions and methods
WO2017096329A1 (en) 2015-12-03 2017-06-08 Juno Therapeutics, Inc. Modified chimeric receptors and related compositions and methods
EP4212166A1 (en) 2015-12-03 2023-07-19 Juno Therapeutics, Inc. Compositions and methods for reducing immune responses against cell therapies
WO2017096327A2 (en) 2015-12-03 2017-06-08 Juno Therapeutics, Inc. Compositions and methods for reducing immune responses against cell therapies
US11815514B2 (en) 2015-12-04 2023-11-14 Juno Therapeutics, Inc. Methods and compositions related to toxicity associated with cell therapy
EP4012415A2 (en) 2015-12-04 2022-06-15 Juno Therapeutics, Inc. Methods and compositions related to toxicity associated with cell therapy
US11479755B2 (en) 2015-12-07 2022-10-25 2Seventy Bio, Inc. T cell compositions
WO2017120501A1 (en) 2016-01-07 2017-07-13 Mayo Foundation For Medical Education And Research Methods of treating cancer with interferon
WO2017161208A1 (en) 2016-03-16 2017-09-21 Juno Therapeutics, Inc. Methods for determining dosing of a therapeutic agent and related treatments
WO2017161212A1 (en) 2016-03-16 2017-09-21 Juno Therapeutics, Inc. Methods for adaptive design of a treatment regimen and related treatments
EP4015536A1 (en) 2016-03-22 2022-06-22 Seattle Children's Hospital (DBA Seattle Children's Research Institute) Early intervention methods to prevent or ameliorate toxicity
US11760804B2 (en) 2016-03-22 2023-09-19 Seattle Children's Hospital Early intervention methods to prevent or ameliorate toxicity
US11518814B2 (en) 2016-03-22 2022-12-06 Seattle Children's Hospital Early intervention methods to prevent or ameliorate toxicity
WO2017165571A1 (en) 2016-03-22 2017-09-28 Seattle Children's Hospital (dba Seattle Children's Research Institute) Early intervention methods to prevent or ameliorate toxicity
WO2017173091A1 (en) 2016-03-30 2017-10-05 Musc Foundation For Research Development Methods for treatment and diagnosis of cancer by targeting glycoprotein a repetitions predominant (garp) and for providing effective immunotherapy alone or in combination
US11400116B2 (en) 2016-05-06 2022-08-02 The Regents Of The University Of California Systems and methods for targeting cancer cells
WO2017193107A2 (en) 2016-05-06 2017-11-09 Juno Therapeutics, Inc. Genetically engineered cells and methods of making the same
WO2017205846A1 (en) 2016-05-27 2017-11-30 Aadigen, Llc Peptides and nanoparticles for intracellular delivery of genome-editing molecules
EP3910059A1 (en) 2016-05-27 2021-11-17 Aadigen, Llc Peptides and nanoparticles for intracellular delivery of genome-editing molecules
EP4011381A1 (en) 2016-06-03 2022-06-15 Memorial Sloan-Kettering Cancer Center Adoptive cell therapies as early treatment options
WO2017214207A2 (en) 2016-06-06 2017-12-14 Juno Therapeutics, Inc. Methods for the treatment of b cell malignancies using adoptive cell therapy
US11820819B2 (en) 2016-06-24 2023-11-21 Icell Gene Therapeutics Inc. Chimeric antigen receptors (CARs), compositions and methods thereof
EP3992632A1 (en) 2016-06-27 2022-05-04 Juno Therapeutics, Inc. Mhc-e restricted epitopes, binding molecules and related methods and uses
WO2018005559A1 (en) 2016-06-27 2018-01-04 Juno Therapeutics, Inc. Method of identifying peptide epitopes, molecules that bind such epitopes and related uses
WO2018005556A1 (en) 2016-06-27 2018-01-04 Juno Therapeutics, Inc. Mhc-e restricted epitopes, binding molecules and related methods and uses
US11421287B2 (en) 2016-07-29 2022-08-23 Juno Therapeutics, Inc. Methods for assessing the presence or absence of replication competent virus
WO2018023093A1 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Immunomodulatory polypeptides and related compositions and methods
WO2018023100A2 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Anti-idiotypic antibodies and related methods
WO2018023094A1 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Methods for assessing the presence or absence of replication competent virus
WO2018027036A1 (en) * 2016-08-03 2018-02-08 Dipersio John F Gene editing of car-t cells for the treatment of t cell malignancies with chimeric antigen receptors
US20180348227A1 (en) * 2016-08-04 2018-12-06 Memorial Sloan-Kettering Cancer Center Cancer antigen targets and uses thereof
US11193938B2 (en) * 2016-08-04 2021-12-07 Memorial Sloan-Kettering Cancer Center Cancer antigen targets and uses thereof
US11401332B2 (en) 2016-08-23 2022-08-02 The Regents Of The University Of California Proteolytically cleavable chimeric polypeptides and methods of use thereof
US11738048B2 (en) 2016-08-30 2023-08-29 Memorial Sloan Kettering Cancer Center Immune cell compositions and methods of use for treating viral and other infections
WO2018049420A1 (en) 2016-09-12 2018-03-15 Juno Therapeutics, Inc. Perfusion bioreactor bag assemblies
EP4353319A2 (en) 2016-09-28 2024-04-17 Atossa Therapeutics, Inc. Methods of adoptive cell therapy
WO2018063985A1 (en) 2016-09-28 2018-04-05 Atossa Genetics Inc. Methods of adoptive cell therapy
US11072660B2 (en) 2016-10-03 2021-07-27 Juno Therapeutics, Inc. HPV-specific binding molecules
WO2018067618A1 (en) 2016-10-03 2018-04-12 Juno Therapeutics, Inc. Hpv-specific binding molecules
EP4190335A1 (en) 2016-10-13 2023-06-07 Juno Therapeutics, Inc. Immunotherapy methods and compositions involving tryptophan metabolic pathway modulators
US11896615B2 (en) 2016-10-13 2024-02-13 Juno Therapeutics, Inc. Immunotherapy methods and compositions involving tryptophan metabolic pathway modulators
WO2018071873A2 (en) 2016-10-13 2018-04-19 Juno Therapeutics, Inc. Immunotherapy methods and compositions involving tryptophan metabolic pathway modulators
WO2018093591A1 (en) 2016-11-03 2018-05-24 Juno Therapeutics, Inc. Combination therapy of a cell based therapy and a microglia inhibitor
WO2018085731A2 (en) 2016-11-03 2018-05-11 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and a btk inhibitor
WO2018102612A1 (en) 2016-12-02 2018-06-07 Juno Therapeutics, Inc. Engineered b cells and related compositions and methods
US11793833B2 (en) 2016-12-02 2023-10-24 Juno Therapeutics, Inc. Engineered B cells and related compositions and methods
WO2018102787A1 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods for determining car-t cells dosing
EP4279136A2 (en) 2016-12-03 2023-11-22 Juno Therapeutics, Inc. Methods for determining car-t cells dosing
WO2018102785A2 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods and compositions for use of therapeutic t cells in combination with kinase inhibitors
WO2018102786A1 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods for modulation of car-t cells
US11590167B2 (en) 2016-12-03 2023-02-28 Juno Therapeutic, Inc. Methods and compositions for use of therapeutic T cells in combination with kinase inhibitors
WO2018106732A1 (en) 2016-12-05 2018-06-14 Juno Therapeutics, Inc. Production of engineered cells for adoptive cell therapy
US11408005B2 (en) 2016-12-12 2022-08-09 Seattle Children's Hospital Chimeric transcription factor variants with augmented sensitivity to drug ligand induction of transgene expression in mammalian cells
EP3336107A1 (en) 2016-12-15 2018-06-20 Miltenyi Biotec GmbH Immune cells expressing an antigen binding receptor and a chimeric costimulatory receptor
WO2018108766A1 (en) 2016-12-15 2018-06-21 Miltenyi Biotec Gmbh Immune cells expressing an antigen binding receptor and a chimeric costimulatory receptor
US11821027B2 (en) 2017-01-10 2023-11-21 Juno Therapeutics, Inc. Epigenetic analysis of cell therapy and related methods
WO2018132518A1 (en) 2017-01-10 2018-07-19 Juno Therapeutics, Inc. Epigenetic analysis of cell therapy and related methods
US11517627B2 (en) 2017-01-20 2022-12-06 Juno Therapeutics Gmbh Cell surface conjugates and related cell compositions and methods
WO2018134691A2 (en) 2017-01-20 2018-07-26 Juno Therapeutics Gmbh Cell surface conjugates and related cell compositions and methods
US11845803B2 (en) 2017-02-17 2023-12-19 Fred Hutchinson Cancer Center Combination therapies for treatment of BCMA-related cancers and autoimmune disorders
WO2018157171A2 (en) 2017-02-27 2018-08-30 Juno Therapeutics, Inc. Compositions, articles of manufacture and methods related to dosing in cell therapy
EP4353818A2 (en) 2017-02-27 2024-04-17 Juno Therapeutics, Inc. Compositions, articles of manufacture and methods related to dosing in cell therapy
US11850262B2 (en) 2017-02-28 2023-12-26 Purdue Research Foundation Compositions and methods for CAR T cell therapy
US11759480B2 (en) 2017-02-28 2023-09-19 Endocyte, Inc. Compositions and methods for CAR T cell therapy
WO2018165228A1 (en) 2017-03-08 2018-09-13 Memorial Sloan Kettering Cancer Center Immune cell compositions and methods of use
WO2018170188A2 (en) 2017-03-14 2018-09-20 Juno Therapeutics, Inc. Methods for cryogenic storage
WO2018167486A1 (en) 2017-03-15 2018-09-20 Oxford Biomedica (Uk) Limited Method
US11427645B2 (en) 2017-03-15 2022-08-30 Oxford Biomedica (Uk) Limited 5T4-targeting agents and methods
US11851659B2 (en) 2017-03-22 2023-12-26 Novartis Ag Compositions and methods for immunooncology
US11547694B2 (en) 2017-03-23 2023-01-10 The General Hospital Corporation CXCR4/CXCR7 blockade and treatment of human papilloma virus-associated disease
US11365236B2 (en) 2017-03-27 2022-06-21 Nkarta, Inc. Truncated NKG2D chimeric receptors and uses thereof in natural killer cell immunotherapy
US11896616B2 (en) 2017-03-27 2024-02-13 National University Of Singapore Stimulatory cell lines for ex vivo expansion and activation of natural killer cells
WO2018187791A1 (en) 2017-04-07 2018-10-11 Juno Therapeutics, Inc Engineered cells expressing prostate-specific membrane antigen (psma) or a modified form thereof and related methods
US11796534B2 (en) 2017-04-14 2023-10-24 Juno Therapeutics, Inc. Methods for assessing cell surface glycosylation
WO2018191723A1 (en) 2017-04-14 2018-10-18 Juno Therapeutics, Inc. Methods for assessing cell surface glycosylation
WO2018195175A1 (en) 2017-04-18 2018-10-25 FUJIFILM Cellular Dynamics, Inc. Antigen-specific immune effector cells
EP4083063A2 (en) 2017-04-18 2022-11-02 FUJIFILM Cellular Dynamics, Inc. Antigen-specific immune effector cells
RU2780020C2 (en) * 2017-04-26 2022-09-19 Еурека Терапьютикс, Инк. Cells expressing chimeric activating receptors and chimeric stimulating receptors and their use
US11613573B2 (en) 2017-04-26 2023-03-28 Eureka Therapeutics, Inc. Chimeric antibody/T-cell receptor constructs and uses thereof
US10822413B2 (en) 2017-04-26 2020-11-03 Eureka Therapeutics, Inc. Cells expressing chimeric activating receptors and chimeric stimulating receptors and uses thereof
US20200115448A1 (en) * 2017-04-26 2020-04-16 Eureka Therapeutics, Inc. Cells expressing chimeric activating receptors and chimeric stimulating receptors and uses thereof
US11965021B2 (en) 2017-04-26 2024-04-23 Eureka Therapeutics, Inc. Cells expressing chimeric activating receptors and chimeric stimulating receptors and uses thereof
WO2018200583A1 (en) * 2017-04-26 2018-11-01 Eureka Therapeutics, Inc. Cells expressing chimeric activating receptors and chimeric stimulating receptors and uses thereof
US11866465B2 (en) 2017-04-27 2024-01-09 Juno Therapeutics Gmbh Oligomeric particle reagents and methods of use thereof
WO2018197949A1 (en) 2017-04-27 2018-11-01 Juno Therapeutics Gmbh Oligomeric particle reagents and methods of use thereof
EP4327878A2 (en) 2017-05-01 2024-02-28 Juno Therapeutics, Inc. Combination of a cell therapy and an immunomodulatory compound
WO2018204427A1 (en) 2017-05-01 2018-11-08 Juno Therapeutics, Inc. Combination of a cell therapy and an immunomodulatory compound
US11944647B2 (en) 2017-06-02 2024-04-02 Juno Therapeutics, Inc. Articles of manufacture and methods for treatment using adoptive cell therapy
WO2018223101A1 (en) 2017-06-02 2018-12-06 Juno Therapeutics, Inc. Articles of manufacture and methods for treatment using adoptive cell therapy
US11413310B2 (en) 2017-06-02 2022-08-16 Juno Therapeutics, Inc. Articles of manufacture and methods for treatment using adoptive cell therapy
WO2018223098A1 (en) 2017-06-02 2018-12-06 Juno Therapeutics, Inc. Articles of manufacture and methods related to toxicity associated with cell therapy
US11740231B2 (en) 2017-06-02 2023-08-29 Juno Therapeutics, Inc. Articles of manufacture and methods related to toxicity associated with cell therapy
EP3828264A1 (en) 2017-06-20 2021-06-02 Institut Curie Immune cells defective for suv39h1
WO2018234370A1 (en) 2017-06-20 2018-12-27 Institut Curie Immune cells defective for suv39h1
EP4302768A2 (en) 2017-06-22 2024-01-10 Board Of Regents, The University Of Texas System Methods for producing regulatory immune cells and uses thereof
WO2019006427A1 (en) 2017-06-29 2019-01-03 Juno Therapeutics, Inc. Mouse model for assessing toxicities associated with immunotherapies
WO2019027850A1 (en) 2017-07-29 2019-02-07 Juno Therapeutics, Inc. Reagents for expanding cells expressing recombinant receptors
US11851678B2 (en) 2017-08-09 2023-12-26 Juno Therapeutics, Inc. Methods for producing genetically engineered cell compositions and related compositions
WO2019032929A1 (en) 2017-08-09 2019-02-14 Juno Therapeutics, Inc. Methods and compositions for preparing genetically engineered cells
WO2019032927A1 (en) 2017-08-09 2019-02-14 Juno Therapeutics, Inc. Methods for producing genetically engineered cell compositions and related compositions
WO2019046832A1 (en) 2017-09-01 2019-03-07 Juno Therapeutics, Inc. Gene expression and assessment of risk of developing toxicity following cell therapy
WO2019051335A1 (en) 2017-09-07 2019-03-14 Juno Therapeutics, Inc. Methods of identifying cellular attributes related to outcomes associated with cell therapy
WO2019057102A1 (en) 2017-09-20 2019-03-28 Tsinghua University A gRNA TARGETING HPK1 AND A METHOD FOR EDITING HPK1 GENE
WO2019070541A1 (en) 2017-10-03 2019-04-11 Juno Therapeutics, Inc. Hpv-specific binding molecules
EP4215543A2 (en) 2017-10-03 2023-07-26 Juno Therapeutics, Inc. Hpv-specific binding molecules
US11952408B2 (en) 2017-10-03 2024-04-09 Juno Therapeutics, Inc. HPV-specific binding molecules
WO2019089855A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Process for generating therapeutic compositions of engineered cells
WO2019090004A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Process for producing a t cell composition
US11066475B2 (en) 2017-11-01 2021-07-20 Juno Therapeutics, Inc. Chimeric antigen receptors specific for B-cell maturation antigen and encoding polynucleotides
US11623961B2 (en) 2017-11-01 2023-04-11 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for B-cell maturation antigen
WO2019090003A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Chimeric antigen receptors specific for b-cell maturation antigen (bcma)
WO2019089858A2 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods of assessing or monitoring a response to a cell therapy
WO2019089982A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Method of assessing activity of recombinant antigen receptors
WO2019089848A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods associated with tumor burden for assessing response to a cell therapy
WO2019089884A2 (en) 2017-11-01 2019-05-09 Editas Medicine, Inc. Methods, compositions and components for crispr-cas9 editing of tgfbr2 in t cells for immunotherapy
US11564946B2 (en) 2017-11-01 2023-01-31 Juno Therapeutics, Inc. Methods associated with tumor burden for assessing response to a cell therapy
US11851679B2 (en) 2017-11-01 2023-12-26 Juno Therapeutics, Inc. Method of assessing activity of recombinant antigen receptors
WO2019087151A1 (en) 2017-11-03 2019-05-09 Sorrento Therapeutics, Inc. Cd38-directed chimeric antigen receptor constructs
WO2019090202A1 (en) 2017-11-06 2019-05-09 Editas Medicine, Inc. Methods, compositions and components for crispr-cas9 editing of cblb in t cells for immunotherapy
WO2019090364A1 (en) 2017-11-06 2019-05-09 Juno Therapeutics, Inc. Combination of a cell therapy and a gamma secretase inhibitor
WO2019094835A1 (en) 2017-11-10 2019-05-16 Juno Therapeutics, Inc. Closed-system cryogenic vessels
WO2019109053A1 (en) 2017-12-01 2019-06-06 Juno Therapeutics, Inc. Methods for dosing and for modulation of genetically engineered cells
US11944644B2 (en) 2017-12-05 2024-04-02 The Medical Research Infrastructure And Health Services Fund Of The Tel Aviv Medical Center T-cells comprising anti-CD38 and anti-CD138 chimeric antigen receptors and uses thereof
WO2019113557A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Process for producing a composition of engineered t cells
WO2019113559A2 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Phenotypic markers for cell therapy and related methods
WO2019113556A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Serum-free media formulation for culturing cells and methods of use thereof
WO2019118937A1 (en) 2017-12-15 2019-06-20 Juno Therapeutics, Inc. Anti-cct5 binding molecules and methods of use thereof
US11919937B2 (en) 2018-01-09 2024-03-05 Board Of Regents, The University Of Texas System T cell receptors for immunotherapy
WO2019141270A1 (en) * 2018-01-19 2019-07-25 科济生物医药(上海)有限公司 Synnotch receptor-regulated expression of il12
US11779602B2 (en) 2018-01-22 2023-10-10 Endocyte, Inc. Methods of use for CAR T cells
WO2019152747A1 (en) 2018-01-31 2019-08-08 Juno Therapeutics, Inc. Methods and reagents for assessing the presence or absence of replication competent virus
US11535903B2 (en) 2018-01-31 2022-12-27 Juno Therapeutics, Inc. Methods and reagents for assessing the presence or absence of replication competent virus
WO2019152743A1 (en) 2018-01-31 2019-08-08 Celgene Corporation Combination therapy using adoptive cell therapy and checkpoint inhibitor
WO2019148656A1 (en) * 2018-02-05 2019-08-08 深圳市默赛尔生物医学科技发展有限公司 Multi-signal chimeric antigen receptor and expression gene thereof, nk cells modified thereby and anti-tumor drug
US11229669B2 (en) 2018-02-11 2022-01-25 Memorial Sloan-Kettering Cancer Center Cells comprising non-HLA restricted T cell receptors
WO2019161133A1 (en) 2018-02-15 2019-08-22 Memorial Sloan Kettering Cancer Center Foxp3 targeting agent compositions and methods of use for adoptive cell therapy
WO2019170147A1 (en) 2018-03-09 2019-09-12 科济生物医药(上海)有限公司 Method and composition for treating tumors
WO2019170845A1 (en) 2018-03-09 2019-09-12 Ospedale San Raffaele S.R.L. Il-1 antagonist and toxicity induced by cell therapy
US11471489B2 (en) 2018-04-05 2022-10-18 Juno Therapeutics, Inc. T cell receptors and engineered cells expressing same
WO2019195486A1 (en) 2018-04-05 2019-10-10 Juno Therapeutics, Inc. T cell receptors and engineered cells expressing same
WO2019195491A1 (en) 2018-04-05 2019-10-10 Juno Therapeutics, Inc. T cells expressing a recombinant receptor, related polynucleotides and methods
WO2019195492A1 (en) 2018-04-05 2019-10-10 Juno Therapeutics, Inc. Methods of producing cells expressing a recombinant receptor and related compositions
WO2019213184A1 (en) 2018-05-03 2019-11-07 Juno Therapeutics, Inc. Combination therapy of a chimeric antigen receptor (car) t cell therapy and a kinase inhibitor
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
WO2020020210A1 (en) 2018-07-24 2020-01-30 科济生物医药(上海)有限公司 Method for treating tumor using immune effector cell
WO2020033927A2 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Processes for generating engineered cells and compositions thereof
WO2020033916A1 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Methods for assessing integrated nucleic acids
WO2020056047A1 (en) 2018-09-11 2020-03-19 Juno Therapeutics, Inc. Methods for mass spectrometry analysis of engineered cell compositions
WO2020061256A1 (en) 2018-09-19 2020-03-26 FUJIFILM Cellular Dynamics, Inc. Protein l for activation and expansion of chimeric antigen receptor-modified immune cells
WO2020089343A1 (en) 2018-10-31 2020-05-07 Juno Therapeutics Gmbh Methods for selection and stimulation of cells and apparatus for same
WO2020092854A2 (en) 2018-11-01 2020-05-07 Juno Therapeutics, Inc. Chimeric antigen receptors specific for g protein-coupled receptor class c group 5 member d (gprc5d)
WO2020092848A2 (en) 2018-11-01 2020-05-07 Juno Therapeutics, Inc. Methods for treatment using chimeric antigen receptors specific for b-cell maturation antigen
WO2020097132A1 (en) 2018-11-06 2020-05-14 Juno Therapeutics, Inc. Process for producing genetically engineered t cells
WO2020097403A1 (en) 2018-11-08 2020-05-14 Juno Therapeutics, Inc. Methods and combinations for treatment and t cell modulation
WO2020102770A1 (en) 2018-11-16 2020-05-22 Juno Therapeutics, Inc. Methods of dosing engineered t cells for the treatment of b cell malignancies
WO2020106621A1 (en) 2018-11-19 2020-05-28 Board Of Regents, The University Of Texas System A modular, polycistronic vector for car and tcr transduction
WO2020113029A2 (en) 2018-11-28 2020-06-04 Board Of Regents, The University Of Texas System Multiplex genome editing of immune cells to enhance functionality and resistance to suppressive environment
WO2020112493A1 (en) 2018-11-29 2020-06-04 Board Of Regents, The University Of Texas System Methods for ex vivo expansion of natural killer cells and use thereof
WO2020113194A2 (en) 2018-11-30 2020-06-04 Juno Therapeutics, Inc. Methods for treatment using adoptive cell therapy
WO2020113188A2 (en) 2018-11-30 2020-06-04 Juno Therapeutics, Inc. Methods for dosing and treatment of b cell malignancies in adoptive cell therapy
WO2020160050A1 (en) 2019-01-29 2020-08-06 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for receptor tyrosine kinase like orphan receptor 1 (ror1)
WO2020169974A1 (en) 2019-02-19 2020-08-27 King's College London Hypoxia-responsive chimeric antigen receptors
US11253547B2 (en) 2019-03-05 2022-02-22 Nkarta, Inc. CD19-directed chimeric antigen receptors and uses thereof in immunotherapy
US11141436B2 (en) 2019-03-05 2021-10-12 Nkarta, Inc. Immune cells engineered to express CD19-directed chimeric antigen receptors and uses thereof in immunotherapy
US11154575B2 (en) 2019-03-05 2021-10-26 Nkarta, Inc. Cancer immunotherapy using CD19-directed chimeric antigen receptors
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
WO2020223571A1 (en) 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cells expressing a chimeric receptor from a modified cd247 locus, related polynucleotides and methods
WO2020247832A1 (en) 2019-06-07 2020-12-10 Juno Therapeutics, Inc. Automated t cell culture
WO2020252218A1 (en) 2019-06-12 2020-12-17 Juno Therapeutics, Inc. Combination therapy of a cell-mediated cytotoxic therapy and an inhibitor of a prosurvival bcl2 family protein
WO2021013950A1 (en) 2019-07-23 2021-01-28 Mnemo Therapeutics Immune cells defective for suv39h1
WO2021035194A1 (en) 2019-08-22 2021-02-25 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and an enhancer of zeste homolog 2 (ezh2) inhibitor and related methods
WO2021038036A1 (en) * 2019-08-28 2021-03-04 King's College London B CELL TARGETED PARALLEL CAR (pCAR) THERAPEUTIC AGENTS
WO2021041994A2 (en) 2019-08-30 2021-03-04 Juno Therapeutics, Inc. Machine learning methods for classifying cells
WO2021043804A1 (en) 2019-09-02 2021-03-11 Institut Curie Immunotherapy targeting tumor neoantigenic peptides
WO2021078910A1 (en) 2019-10-22 2021-04-29 Institut Curie Immunotherapy targeting tumor neoantigenic peptides
WO2021084050A1 (en) 2019-10-30 2021-05-06 Juno Therapeutics Gmbh Cell selection and/or stimulation devices and methods of use
WO2021092097A1 (en) 2019-11-05 2021-05-14 Juno Therapeutics, Inc. Methods of determining attributes of therapeutic t cell compositions
WO2021092498A1 (en) 2019-11-07 2021-05-14 Juno Therapeutics, Inc. Combination of a t cell therapy and (s)-3-[4-(4-morpholin-4 ylmethyl-benzyloxy)-l-oxo-l,3-dihydro-isoindol-2-yl]- piperidine-2,6-dione
WO2021113770A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Methods related to toxicity and response associated with cell therapy for treating b cell malignancies
WO2021113780A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to gprc5d-targeted binding domains and related compositions and methods
WO2021113776A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to bcma-targeted binding domains and related compositions and methods
WO2021151008A1 (en) 2020-01-24 2021-07-29 Juno Therapuetics, Inc. Methods for dosing and treatment of follicular lymphoma and marginal zone lymphoma in adoptive cell therapy
WO2021154887A1 (en) 2020-01-28 2021-08-05 Juno Therapeutics, Inc. Methods for t cell transduction
WO2021163389A1 (en) 2020-02-12 2021-08-19 Juno Therapeutics, Inc. Bcma-directed chimeric antigen receptor t cell compositions and methods and uses thereof
WO2021163391A1 (en) 2020-02-12 2021-08-19 Juno Therapeutics, Inc. Cd19-directed chimeric antigen receptor t cell compositions and methods and uses thereof
WO2021167908A1 (en) 2020-02-17 2021-08-26 Board Of Regents, The University Of Texas System Methods for expansion of tumor infiltrating lymphocytes and use thereof
WO2021164959A1 (en) * 2020-02-17 2021-08-26 Miltenyi Biotec B.V. & Co. KG Method for providing personalized cells with chimeric antigen receptors (car) against tumor microenvironment cells
WO2021207689A2 (en) 2020-04-10 2021-10-14 Juno Therapeutics, Inc. Methods and uses related to cell therapy engineered with a chimeric antigen receptor targeting b-cell maturation antigen
WO2021222330A2 (en) 2020-04-28 2021-11-04 Juno Therapeutics, Inc. Combination of bcma-directed t cell therapy and an immunomodulatory compound
WO2021228999A1 (en) 2020-05-12 2021-11-18 Institut Curie Neoantigenic epitopes associated with sf3b1 mutations
WO2021231661A2 (en) 2020-05-13 2021-11-18 Juno Therapeutics, Inc. Process for producing donor-batched cells expressing a recombinant receptor
WO2021231657A1 (en) 2020-05-13 2021-11-18 Juno Therapeutics, Inc. Methods of identifying features associated with clinical response and uses thereof
WO2021237068A2 (en) 2020-05-21 2021-11-25 Board Of Regents, The University Of Texas System T cell receptors with vgll1 specificity and uses thereof
WO2021260186A1 (en) 2020-06-26 2021-12-30 Juno Therapeutics Gmbh Engineered t cells conditionally expressing a recombinant receptor, related polynucleotides and methods
WO2022016119A1 (en) 2020-07-17 2022-01-20 Simurx, Inc. Chimeric myd88 receptors for redirecting immunosuppressive signaling and related compositions and methods
WO2022023576A1 (en) 2020-07-30 2022-02-03 Institut Curie Immune cells defective for socs1
WO2022029660A1 (en) 2020-08-05 2022-02-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to ror1-targeted binding domains and related compositions and methods
WO2022098787A1 (en) 2020-11-04 2022-05-12 Juno Therapeutics, Inc. Cells expressing a chimeric receptor from a modified invariant cd3 immunoglobulin superfamily chain locus and related polynucleotides and methods
US11628218B2 (en) 2020-11-04 2023-04-18 Myeloid Therapeutics, Inc. Engineered chimeric fusion protein compositions and methods of use thereof
WO2022104109A1 (en) 2020-11-13 2022-05-19 Catamaran Bio, Inc. Genetically modified natural killer cells and methods of use thereof
WO2022115611A1 (en) 2020-11-25 2022-06-02 Catamaran Bio, Inc. Cellular therapeutics engineered with signal modulators and methods of use thereof
WO2022133030A1 (en) 2020-12-16 2022-06-23 Juno Therapeutics, Inc. Combination therapy of a cell therapy and a bcl2 inhibitor
WO2022150731A1 (en) 2021-01-11 2022-07-14 Sana Biotechnology, Inc. Use of cd8-targeted viral vectors
WO2022187406A1 (en) 2021-03-03 2022-09-09 Juno Therapeutics, Inc. Combination of a t cell therapy and a dgk inhibitor
WO2022189639A1 (en) 2021-03-11 2022-09-15 Mnemo Therapeutics Tumor neoantigenic peptides and uses thereof
WO2022189626A2 (en) 2021-03-11 2022-09-15 Mnemo Therapeutics Tumor neoantigenic peptides
WO2022189620A1 (en) 2021-03-11 2022-09-15 Institut Curie Transmembrane neoantigenic peptides
WO2022204071A1 (en) 2021-03-22 2022-09-29 Juno Therapeutics, Inc. Method to assess potency of viral vector particles
WO2022204070A1 (en) 2021-03-22 2022-09-29 Juno Therapeutics, Inc. Methods of determining potency of a therapeutic cell composition
WO2022212384A1 (en) 2021-03-29 2022-10-06 Juno Therapeutics, Inc. Combination of a car t cell therapy and an immunomodulatory compound for treatment of lymphoma
WO2022212400A1 (en) 2021-03-29 2022-10-06 Juno Therapeutics, Inc. Methods for dosing and treatment with a combination of a checkpoint inhibitor therapy and a car t cell therapy
WO2022221726A2 (en) 2021-04-16 2022-10-20 Juno Therapeutics, Inc. Combination therapies with bcma-directed t cell therapy
WO2022221737A1 (en) 2021-04-16 2022-10-20 Juno Therapeutics, Inc. T cell therapy in patients who have had prior stem cell transplant
WO2022234009A2 (en) 2021-05-06 2022-11-10 Juno Therapeutics Gmbh Methods for stimulating and transducing t cells
WO2022238386A1 (en) 2021-05-10 2022-11-17 Institut Curie Methods for the treatment of cancer, inflammatory diseases and autoimmune diseases
WO2022248602A1 (en) 2021-05-25 2022-12-01 Institut Curie Myeloid cells overexpressing bcl2
WO2023014922A1 (en) 2021-08-04 2023-02-09 The Regents Of The University Of Colorado, A Body Corporate Lat activating chimeric antigen receptor t cells and methods of use thereof
WO2023015217A1 (en) 2021-08-04 2023-02-09 Sana Biotechnology, Inc. Use of cd4-targeted viral vectors
WO2023081735A1 (en) 2021-11-03 2023-05-11 Celgene Corporation Chimeric antigen receptors specific for b-cell maturation antigen for use in treating myeloma
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
WO2023081900A1 (en) 2021-11-08 2023-05-11 Juno Therapeutics, Inc. Engineered t cells expressing a recombinant t cell receptor (tcr) and related systems and methods
WO2023105000A1 (en) 2021-12-09 2023-06-15 Zygosity Limited Vector
WO2023115039A2 (en) 2021-12-17 2023-06-22 Sana Biotechnology, Inc. Modified paramyxoviridae fusion glycoproteins
WO2023115041A1 (en) 2021-12-17 2023-06-22 Sana Biotechnology, Inc. Modified paramyxoviridae attachment glycoproteins
WO2023126458A1 (en) 2021-12-28 2023-07-06 Mnemo Therapeutics Immune cells with inactivated suv39h1 and modified tcr
WO2023139269A1 (en) 2022-01-21 2023-07-27 Mnemo Therapeutics Modulation of suv39h1 expression by rnas
WO2023147515A1 (en) 2022-01-28 2023-08-03 Juno Therapeutics, Inc. Methods of manufacturing cellular compositions
WO2023150518A1 (en) 2022-02-01 2023-08-10 Sana Biotechnology, Inc. Cd3-targeted lentiviral vectors and uses thereof
WO2023164440A1 (en) 2022-02-22 2023-08-31 Juno Therapeutics, Inc. Proteinase 3 (pr3) chimeric autoantibody receptor t cells and related methods and uses
WO2023178348A1 (en) 2022-03-18 2023-09-21 The Regents Of The University Of Colorado, A Body Corporate Genetically engineered t-cell co-receptors and methods of use thereof
WO2023180552A1 (en) 2022-03-24 2023-09-28 Institut Curie Immunotherapy targeting tumor transposable element derived neoantigenic peptides in glioblastoma
WO2023187024A1 (en) 2022-03-31 2023-10-05 Institut Curie Modified rela protein for inducing interferon expression and engineered immune cells with improved interferon expression
WO2023193015A1 (en) 2022-04-01 2023-10-05 Sana Biotechnology, Inc. Cytokine receptor agonist and viral vector combination therapies
WO2023196921A1 (en) 2022-04-06 2023-10-12 The Regents Of The University Of Colorado, A Body Corporate Granzyme expressing t cells and methods of use
WO2023196933A1 (en) 2022-04-06 2023-10-12 The Regents Of The University Of Colorado, A Body Corporate Chimeric antigen receptor t cells and methods of use thereof
WO2023211972A1 (en) 2022-04-28 2023-11-02 Medical University Of South Carolina Chimeric antigen receptor modified regulatory t cells for treating cancer
WO2023213969A1 (en) 2022-05-05 2023-11-09 Juno Therapeutics Gmbh Viral-binding protein and related reagents, articles, and methods of use
WO2023220641A2 (en) 2022-05-11 2023-11-16 Juno Therapeutics, Inc. Methods and uses related to t cell therapy and production of same
WO2023220655A1 (en) 2022-05-11 2023-11-16 Celgene Corporation Methods to overcome drug resistance by re-sensitizing cancer cells to treatment with a prior therapy via treatment with a t cell therapy
WO2023222617A1 (en) 2022-05-16 2023-11-23 Miltenyi Biotec B.V. & Co. KG Endogenous signaling molecule activating chimeric antigen receptors and methods of generation thereof
WO2023222928A2 (en) 2022-05-20 2023-11-23 Mnemo Therapeutics Compositions and methods for treating a refractory or relapsed cancer or a chronic infectious disease
EP4279085A1 (en) 2022-05-20 2023-11-22 Mnemo Therapeutics Compositions and methods for treating a refractory or relapsed cancer or a chronic infectious disease
WO2023230548A1 (en) 2022-05-25 2023-11-30 Celgene Corporation Method for predicting response to a t cell therapy
WO2023230581A1 (en) 2022-05-25 2023-11-30 Celgene Corporation Methods of manufacturing t cell therapies
WO2023250400A1 (en) 2022-06-22 2023-12-28 Juno Therapeutics, Inc. Treatment methods for second line therapy of cd19-targeted car t cells
WO2024006960A1 (en) 2022-06-29 2024-01-04 Juno Therapeutics, Inc. Lipid nanoparticles for delivery of nucleic acids
WO2024031091A2 (en) 2022-08-05 2024-02-08 Juno Therapeutics, Inc. Chimeric antigen receptors specific for gprc5d and bcma
WO2024044779A2 (en) 2022-08-26 2024-02-29 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for delta-like ligand 3 (dll3)
WO2024054944A1 (en) 2022-09-08 2024-03-14 Juno Therapeutics, Inc. Combination of a t cell therapy and continuous or intermittent dgk inhibitor dosing
WO2024062138A1 (en) 2022-09-23 2024-03-28 Mnemo Therapeutics Immune cells comprising a modified suv39h1 gene
WO2024081820A1 (en) 2022-10-13 2024-04-18 Sana Biotechnology, Inc. Viral particles targeting hematopoietic stem cells

Also Published As

Publication number Publication date
CN112458057A (en) 2021-03-09
PH12015500747B1 (en) 2015-05-25
RU2020124583A (en) 2020-08-03
NZ746914A (en) 2020-03-27
JP2020096625A (en) 2020-06-25
JP6963051B2 (en) 2021-11-05
CN104853766A (en) 2015-08-19
JP2020078331A (en) 2020-05-28
JP2022024161A (en) 2022-02-08
EP3597215A1 (en) 2020-01-22
SG11201502598SA (en) 2015-05-28
US20200317781A1 (en) 2020-10-08
US10654928B2 (en) 2020-05-19
KR20150063145A (en) 2015-06-08
MX2015004287A (en) 2016-03-01
KR102198058B1 (en) 2021-01-06
AU2020294287B2 (en) 2023-06-08
AU2018204297A1 (en) 2018-07-05
JP6441802B2 (en) 2018-12-19
RU2015116901A (en) 2016-11-27
CN112430580A (en) 2021-03-02
IL238047A0 (en) 2015-05-31
CA2886859A1 (en) 2014-04-10
PH12019502424A1 (en) 2021-09-01
PH12015500747A1 (en) 2015-05-25
AU2018204297B2 (en) 2020-10-01
US11712469B2 (en) 2023-08-01
NZ706541A (en) 2019-07-26
ZA201502880B (en) 2020-11-25
IL272539B (en) 2021-02-28
EP2903637A4 (en) 2016-10-19
JP2015535689A (en) 2015-12-17
JP2018108111A (en) 2018-07-12
RU2729401C2 (en) 2020-08-06
IL238047B (en) 2020-02-27
AU2020294287A1 (en) 2021-01-28
US20240091353A1 (en) 2024-03-21
AU2013327136A1 (en) 2015-04-16
ES2743738T3 (en) 2020-02-20
EP2903637A1 (en) 2015-08-12
US20150342993A1 (en) 2015-12-03
HK1208631A1 (en) 2016-03-11
EP2903637B1 (en) 2019-06-12
CA2886859C (en) 2022-05-17
MX370148B (en) 2019-12-03
IL272539A (en) 2020-03-31

Similar Documents

Publication Publication Date Title
AU2020294287B2 (en) Compositions and methods for immunotherapy
US11103531B2 (en) Compositions and methods for immunotherapy
WO2015142314A1 (en) Compositions and methods for immunotherapy

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: 13844468

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 238047

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2886859

Country of ref document: CA

Ref document number: 2015534828

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2015/004287

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 12015500747

Country of ref document: PH

ENP Entry into the national phase

Ref document number: 2013327136

Country of ref document: AU

Date of ref document: 20131002

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2013844468

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: IDP00201502523

Country of ref document: ID

ENP Entry into the national phase

Ref document number: 20157011160

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2015116901

Country of ref document: RU

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015007432

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112015007432

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150401