WO2015150771A1 - Chimeric antigen receptor (car) signalling system - Google Patents

Chimeric antigen receptor (car) signalling system Download PDF

Info

Publication number
WO2015150771A1
WO2015150771A1 PCT/GB2015/050974 GB2015050974W WO2015150771A1 WO 2015150771 A1 WO2015150771 A1 WO 2015150771A1 GB 2015050974 W GB2015050974 W GB 2015050974W WO 2015150771 A1 WO2015150771 A1 WO 2015150771A1
Authority
WO
WIPO (PCT)
Prior art keywords
signalling
cid
component
car
domain
Prior art date
Application number
PCT/GB2015/050974
Other languages
French (fr)
Inventor
Martin PULÉ
Shaun CORDOBA
Khai KONG
Original Assignee
Ucl Business Plc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ucl Business Plc filed Critical Ucl Business Plc
Priority to DK15714272.0T priority Critical patent/DK3126380T3/en
Priority to PL15714272T priority patent/PL3126380T3/en
Priority to ES15714272T priority patent/ES2877129T3/en
Priority to EP15714272.0A priority patent/EP3126380B1/en
Priority to US15/301,148 priority patent/US10588967B2/en
Publication of WO2015150771A1 publication Critical patent/WO2015150771A1/en
Priority to US16/775,257 priority patent/US20200338194A1/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
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/166Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the carbon of a carboxamide group directly attached to the aromatic ring, e.g. procainamide, procarbazine, metoclopramide, labetalol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1774Immunoglobulin superfamily (e.g. CD2, CD4, CD8, ICAM molecules, B7 molecules, Fc-receptors, MHC-molecules)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/52Isomerases (5)
    • 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
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464412CD19 or B4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • 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
    • 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
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N5/0646Natural killers cells [NK], NKT cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • 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
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/90Isomerases (5.)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/11Protein-serine/threonine kinases (2.7.11)
    • C12Y207/11001Non-specific serine/threonine protein kinase (2.7.11.1), i.e. casein kinase or checkpoint kinase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y502/00Cis-trans-isomerases (5.2)
    • C12Y502/01Cis-trans-Isomerases (5.2.1)
    • C12Y502/01008Peptidylprolyl isomerase (5.2.1.8), i.e. cyclophilin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • 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
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor

Definitions

  • the present invention relates to an antigen receptor signalling system. BACKGROUND TO THE INVENTION
  • Chimeric antigen receptors are proteins which graft the specificity of a monoclonal antibody (mAb) to the effector function of a T-cell. Their usual form is that of a type I transmembrane domain protein with an antigen recognizing amino terminus, a spacer, a transmembrane domain all connected to a compound endodomain which transmits T-cell survival and activation signals (see Figure 1A).
  • scFv single-chain variable fragments
  • toxicities include immunological toxicity caused by sustained intense activation of the CAR T-cells resulting in a macrophage activation syndrome (MAS) and "On-target off-tumour" toxicity i.e. recognition of the target antigen on normal tissues.
  • MAS macrophage activation syndrome
  • MAS is presumed to be caused by persistent antigen-driven activation and proliferation of T-cells which in turn release copious inflammatory cytokines leading to hyper-activation of macrophages and a feed-forward cycle of immune activation.
  • a large spike in serum IL-6 is characteristic and the syndrome can result in a severe systemic illness requiring ICU admission.
  • On-target off-tumour toxicity has been reported with other CARs, for example a group of patients treated with a CAR against the renal cell carcinoma antigen CAIX developed unexpected and treatment limiting biliary toxicity.
  • Two fatalities have been reported with CAR studies: one patient died of a respiratory distress syndrome which occurred immediately post-infusion of a large dose of 3rd generation anti-ERBB2 CAR T-cells; a further patient died in a different study after a possible cytokine storm following treatment of CLL with a second generation anti-CD19 CAR.
  • CAR T-cells do not have a half-life and one cannot cease administration and wait for the agent to breakdown/become excreted. CAR T-cells are autonomous and can engraft and proliferate. Toxicity can therefore be progressive and fulminant.
  • Suicide genes are genetically expressed elements which can conditionally destroy cells which express them. Examples include Herpes-simplex virus thymidine kinase, which renders cells susceptible to Ganciclovir; inducible Caspase 9, which renders cells susceptible to a small molecular homodimerizer and CD20 and RQR8, which renders cells susceptible to Rituximab.
  • This technology adds a certain amount of safety to CAR T-cell therapy, however there are limitations. Firstly, it is a binary approach wherein all the CAR T-cells are destroyed upon addition of the suicide entity. In addition, medicinal therapeutics often have a therapeutic window. With a suicide gene the potency of the product cannot be tuned such that efficacy with tolerable toxicity can be achieved. Secondly, it is not clear whether a suicide gene would help with some of the immune-toxicities described above: for instance by the time a macrophage activation syndrome had been triggered, it may well no longer need the CAR T-cells to perpetuate and the suicide gene would no longer be helpful. The more acute cytokine release syndromes probably occur too quickly for the suicide gene to work.
  • suicide genes are not "fail-safe", i.e. the default status is for the CAR T-cells to be active. There is thus a need for alternative methods for controlling CAR T-cells that are not associated with the disadvantages and problems mentioned above.
  • Figure 1 - a Schematic diagram illustrating a classical CAR.
  • FIG. 3 Illustration of an example of an inducible CAR signalling system in its simplest incarnation,
  • (a) Receptor component comprising of an extracellular scFv, a spacer, a trans-membrane domain and an intracellular FKBP12.
  • CID chemical inducer of dimerization
  • the receptor component and the signalling components dimerize allowing signalling in the presence of cognate antigen.
  • FIG 4 Function of an inducible CAR system.
  • T-cells were transduced with a CAR system as illustrated in figure 3 and with SEQ ID No. 19. This CAR recognizes CD19.
  • CAR expressing T-cells were challenged with CD19+ targets at 1 : 1 and 2: 1 effector to target ratios in the presence of increasing concentration of CID.
  • T-cells respond to target cells only in the presence of CID. Stimulation with PMA/lonomycin is also show.
  • Response to CD19+ targets of T-cells expressing a standard CD19 CAR (MP15577) to different effector target ratios is also shown.
  • FIG. 5 Illustration of an inducible CAR system with two receptor components,
  • the two receptor components are co-expressed along with a single signalling component.
  • the two receptor components each have a different single chain which recognizes a different target.
  • the receptor components have endodomains comprising of the FRB fragment of mTOR.
  • a single signalling component comprises of FKBP12 and a fusion of CD28, OX40 and CD3 Zeta endodomains.
  • the FRB sequences are identical and this CAR will signal equally in response to either target.
  • the FRB sequences differ such that each has a different affinity for the CID. With this implementation, the CAR will signal with different strength in response to each cognate antigen in the presence of CID.
  • FIG. 6 Illustration of an example of a multiple transmembrane receptor domain receptor component for use in an inducible CAR signalling system.
  • the receptor component is a chimera of a type I CAR linked to the multi-span protein.
  • a scFv is connected to an Fc spacer domain and a TM domain. This in turn is connected to the CD20 molecule with truncated amino and carboxy termini. Each intracellular entry is attached to an FRB domain.
  • the signalling component comprises of FKBP12 and a fusion of CD28, OX40 and CD3 Zeta endodomains. Addition of the CID allows the stochastic ligation of multiple signalling domains to the multiple endodomains which recognize a single antigen receptor component, resulting in amplified signalling in response to antigen.
  • FIG. 7 Illustration of an inducible CAR system with multiple signalling component inducible CAR.
  • This CAR system has a single receptor component comprising of a scFv, an Fc spacer, a TM domain and a FKBP12 endodomain.
  • the three different signalling domains comprise of a fusion between the FRB fragment of mTOR and the CD28 endodomain, a fusion between the FRB fragment of mTOR and the 41 BB endodomain and a fusion between the FRB fragment of mTOR and the CD3 Zeta endodomain.
  • the FRB domains are identical so that each signalling component is recruited equally to the receptor component and the CAR transmits an equal CD28, 41 BB and CD3 Zeta signal in the presence of CID and upon recognition of the cognate antigen.
  • the FRB domains are different so that each signalling component can be recruited differently to the receptor component so that the CAR transmits more or less of each of CD28, 41 BB and CD3 Zeta signals depending on the optimum needed for the application.
  • Figure 8 Illustration of an inducible CAR with signalling component which comprises of two CID binding domains.
  • the signalling domain comprises of the FRB domain of mTOR, a fusion of CD28, OX40 and Zeta endodomains and an FKBP12 domain.
  • the signalling components multimerize with each other and with the receptor component. Upon antigen recognition, a signal equivalent to the sum of the signalling component multimer is transmitted.
  • RSL1 is one of a family of compounds that have been found to act as non-steroidal ecdysone agonists and can function as gene inducers.
  • FIG 10 - RSL1 inducible CAR (a) an RSL1 inducible CAR was constructed by using retinoic acid receptor domains and ecodysone receptor domains to constitute the heterodimerization motifs for the antigen recognition component and the signalling component respectively. The antigen recognition component recognizes CD19. (b) T- cells expressing this CAR along with non-transduced T-cells were stained with anti-Fc which recognizes the CAR spacer. The transduced T-cells expressed the CAR well, (c) Transduced T-cells were next challenged with increasing concentration of RSL1 and challenged with either CD19 negative or CD19 positive target cells. T-cells challenged with CD19 positive targets signal only in the presence of RSL1.
  • FIG 11 AP1903 homodimerizer inducible CAR.
  • AP1903 homodimerizer inducible CAR was constructed by using modified FKBP12 domains to constitute the heterodimerization motifs for both the antigen recognition component and the signalling component. The antigen recognition component recognizes CD19.
  • T- cells expressing this CAR along with non-transduced T-cells were stained with anti-Fc which recognizes the CAR spacer. The transduced T-cells expressed the CAR well,
  • Transduced T-cells were next challenged with increasing concentration of CID dimerizer AP1903 and challenged with either CD19 negative or CD19 positive target cells. T-cells challenged with CD19 positive targets signal only in the presence of CID. At high CID concentrations, signalling is reduced likely to saturation of heterodimerization domains with an excess of CID reducing the formation of heterodimers.
  • the present inventors have found that it is possible to separate the antigen- recognition and signalling components of a CAR to produce an inducible system, where signalling only takes place in the presence of an agent such as a small molecule which can induce dimerization (henceforth referred to as chemical inducer of dimerization or CID).
  • CID binds two protein domains simultaneously.
  • Functional signalling by the CAR is hence dependent on the presence of the CID.
  • This provides a mechanism for controlling the CAR T-cell after it has been administered to a patient:
  • the CAR T-cell can be remotely switched on by administering the CID to the patient, and in case of toxicity switched off by not administering further CID. If activity of the CAR is proportional to local concentration of the agent (for instance when CID concentration is not saturating) activity of the CAR T-cells can be "tuned” by altering CID dose administered to the patient.
  • the present relates to a chimeric antigen receptor (CAR) signalling system comprising of two separate protein components:
  • a membrane spanning receptor component comprising an extracellular antigen-binding domain, a transmembrane domain and a first intracellular CID binding domain (referred henceforth as CBD1);
  • CBD2 An intracellular signalling component comprising at a minimum a signalling domain and a second CID binding domain (henceforth referred to as CBD2);
  • CBD1 and CBD2 are capable of simultaneously and specifically binding to the CID.
  • the present invention provides a chimeric antigen receptor (CAR) signalling system comprising;
  • a receptor component comprising an extracellular antigen-binding domain, a transmembrane domain and a intracellular first chemical inducer of dimerization (CID) binding domain (CBD1);
  • an intracellular signalling component comprising a signalling domain and a second CID binding domain (CBD2);
  • CBD1 and CBD2 are capable of simultaneously binding to a CID
  • CBD1 and CBD2 of the CAR signalling system according to the first aspect may comprise different CID binding domains and the CID may comprise two different binding moieties.
  • CDB1 may comprise the rapamycin binding domain of FK-binding protein 12 (FKBP12), CDB2 may comprise the FKBP12-Rapamycin Binding (FRB) domain of mTOR, or visa-versa.
  • the CID may comprise rapamycin or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
  • CBD1 may comprise the FK506 (Tacrolimus) binding domain of FK-binding protein 12 (FKBP12), CBD2 may comprise the cyclosporin binding domain of cylcophilin A (or visa versa).
  • the CID may comprise an FK506/cyclosporin fusion protein or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
  • CBD1 may comprise an oestrogen-binding domain (EBD)
  • CBD2 may comprise a streptavidin binding domain, or visa-versa.
  • the CID may comprise an estrone/biotin fusion protein or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
  • CBD1 may comprise a glucocorticoid-binding domain (GBD)
  • CBD2 may comprise a dihydrofolate reductase (DHFR) binding domain, or visa-versa.
  • the CID may comprise a dexamethasone/methotrexate fusion molecule or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
  • CBD1 may comprise an 0 6 -alkylguanine-DNA alkyltransferase (AGT) binding domain
  • CBD2 may comprise a dihydrofolate reductase (DHFR) binding domain, or vice- versa.
  • the CID may comprise an 0 6 -benzylguanine derivative/methotrexate fusion molecule or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
  • CBD1 may comprise a retinoic acid receptor domain
  • CBD2 may comprise an ecodysone receptor domain, or vice-versa.
  • the CID may comprise RSL1 or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
  • CBD1 and CBD2 of the CAR signalling system according to the first aspect of the invention may comprise the same CID binding domain and the CID may comprise two identical binding moieties.
  • CBD1 and CBD2 of the CAR signalling system may comprise FK506 binding protein (FKBP12) binding domains comprising a F36V mutation and the CID may comprise AP1903 or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
  • FKBP12 FK506 binding protein
  • the CAR may comprise a receptor component which spans the membrane multiple times with each intracellular portion constituting a CBD1. Hence a single receptor component may recruit multiple signalling components hence amplifying signal.
  • the CAR may comprise a set of separate receptor components each with an intracellular CBD1 such that in the presence of CID, signalling components connect to each receptor protein component so that the CAR can signal in response to a multiplicity of cognate antigens.
  • the CID binding domains may be altered such that affinity to the CID differs. For instance altering amino acids at Positions 2095, 2098, and 2101 of FRB can alter binding to Rapamycin: KTW has high, KHF intermediate and PLW is low (Bayle et al, Chemistry & Biology 13, 99-107, January 2006)
  • the CAR may comprise multiple receptor components such that each one has a CBD1 domain of different affinity to the CID.
  • signalling protein components connect to each receptor protein component differentially and hence the CAR signal strengths differentially in response to each cognate antigen.
  • the signalling domain of the signalling protein component may comprise of a single endodomain selected from activating or co-stimulating T-cell signalling receptor such as CD3 zeta, CD28, 41 BB or OX40.
  • the signalling domain of the signalling protein component may comprise of a multiplicity of endodomains selected from activating and co- stimulating T-cell signalling receptor such as CD3 Zeta, CD28, 41 BB and OX40
  • the signalling domain of the signalling protein may comprise the endodomain of an inhibitory T-cell signalling receptor or a phosphatase.
  • the signalling component according to the above embodiments of the invention may comprise a plurality of signalling components, each comprising a different signalling domain and a similar CBD2, wherein the CID agent binding domains each recognise the same CID, but the signalling domains comprise different endodomains.
  • the CAR can transmit multiple signals but the signalling components being in trans rather than cis are sterically unencumbered from each other's signalling transmission molecules unlike a single signalling component containing a multiplicity of endodomains.
  • the CBD2 of the multiple signalling domains in the aspect described above may bind to the CID molecule with different affinities such that the relative contribution of each signalling component can be tuned.
  • the CAR can transmit a more complex signal rather than the fixed stoichiometry of a single signalling component containing a multiplicity of endodomains.
  • the CAR systems described above may comprise signalling domains which contains a CBD1 , single or multiple endodomains as well as CBD2. Hence, in the presence of CID, concatenation of multiple signalling domains together to amplify signals.
  • the present invention provides a receptor component suitable for use in the CAR signalling system of the first aspect of the invention which comprises an antigen-binding domain, a transmembrane domain and a CID binding domain.
  • the present invention provides a signalling component suitable for use in the CAR signalling system according to the first aspect of the invention which comprises a signalling domain and a CID binding domain.
  • the present invention provides a nucleic acid encoding the receptor component according to the second aspect of the invention.
  • the present invention provides a nucleic acid encoding the signalling component according to the third aspect of the invention.
  • the present invention provides a nucleic acid sequence encoding the receptor component according to the second aspect of the invention and the signalling component according to the third aspect of the invention, wherein the expressed molecule is a self-cleaving peptide which is cleaved between the receptor component and the signalling component.
  • the present invention provides a vector comprising a nucleic acid sequence according to any of the fourth to sixth aspects of the invention.
  • the present invention provides a retroviral vector or a lentiviral vector or a transposon comprising a vector according to the seventh aspect of the invention.
  • the invention provides a T cell or NK cell which expresses a receptor component according to the second aspect of the invention and at least one signalling component according to the third aspect of the invention.
  • the T cell or NK cell according to the ninth aspect of the invention may comprise the nucleic acid according to any of the fourth to sixth aspects of the invention or the vector according to the seventh or eighth aspects of the invention.
  • the present invention provides a pharmaceutical composition comprising a plurality of T cells or NK cells according to the ninth aspect of the invention.
  • the present invention provides a pharmaceutical composition according to the tenth13 th aspect of the invention for use in treating and/or preventing a disease.
  • the invention provides a method for treating and/or preventing a disease, which comprises the step of administering a pharmaceutical composition according to the tenth aspect of the invention to a subject.
  • the method according to the twelfth aspect of the invention may comprise the following steps:
  • the method according to the twelfth aspect of the invention may further comprise the step of administering a CID suitable for use in the CAR signalling system according to the first aspect to the subject.
  • the present invention provides a method for treating and/or preventing a disease in a subject which subject comprises T cells according to the ninth aspect of the invention, which method comprises the step of administering a CIFD suitable for use in the CAR signalling system according to the first aspect of the invention to the subject.
  • the method according to the twelfth or thirteenth aspect of the invention may involve monitoring the progression of disease and/or monitoring toxic activity in the subject and adjusting the dose of the CID to provide acceptable levels of disease progression and/or toxic activity.
  • the disease to be prevented and/or treated according to the eleventh, twelfth or thirteenth aspect of the invention may be cancer.
  • the present invention provides the use of a pharmaceutical composition according to the tenth aspect of the invention in the manufacture of a medicament for the treatment and/or prevention of a disease.
  • the present invention provides a kit which comprises a nucleic acid according to any of the fourth to sixth aspects of the invention or a vector according to the seventh or eighth aspects of the invention.
  • the kit may also comprise a CID suitable for use in the CAR signalling system according to the first aspect of the invention.
  • the invention provides a kit which comprises a T cell or NK cell according to the ninth aspect of the invention and a CID suitable for use in the CAR signalling system according to the first aspect of the invention.
  • the present invention provides a method for making a T cell or NK cell according to claim ninth aspect of the invention, which comprises the step of introducing: a nucleic acid sequence according to any of the fourth to sixth aspects of the invention or a vector according to the seventh or eighth aspects of the invention, into a T or NK cell.
  • the T or NK cell may be from a sample isolated from a subject.
  • the invention provides a method for activating the CAR signalling system according to the first aspect of the invention in a subject which comprises a T-cell according to the ninth aspect of the invention, which method comprises the step of administering the CID to the subject.
  • the invention provides a method for reducing the activity of the CAR signalling system according to the first aspect of the invention, in a subject which comprises a T-cell or NK cell according to the ninth aspect of the invention, which method comprises reducing or stopping administration of the CID to the subject.
  • the present invention provides a method for activating signalling in a CAR signalling system according to the first aspect of the invention in a T cell or NK cell according to the ninth aspect of the invention in culture, which comprises the step of introducing the CID agent to the T or NK cell culture.
  • the present invention provides a method for reducing or stopping signalling via a CAR signalling system according to the first aspect of the invention in a T or NK cell according to the ninth aspect of the invention in culture, which comprises the step of removing the CID from the T or NK cell culture.
  • a method for inducing dimerization in vivo between a receptor component according to the second aspect of the invention and a signalling component according to the third aspect of the invention which comprises the step of administering a chemical inducer of dimerization (CID) to a subject comprising a cell according to the ninth aspect of the invention.
  • CID chemical inducer of dimerization
  • the present invention therefore provides a means of tuning the CAR activity to the presence of an agent, such as a small molecule. This allows the potency of CAR cells to be controlled pharmacologically and tuned to an acceptable balance between achieving the desired therapeutic effect, while avoiding CAR-associated toxicities.
  • Classical CARs which are shown schematically in Figure 1 , are chimeric type I transmembrane proteins which connect an extracellular antigen-recognizing domain (binder) to an intracellular signalling domain (endodomain).
  • the binder is typically a single-chain variable fragment (scFv) derived from a monoclonal antibody (mAb), but it can be based on other formats which comprise an antibody-like antigen binding site.
  • scFv single-chain variable fragment
  • mAb monoclonal antibody
  • a spacer domain may be necessary to isolate the binder from the membrane and to allow it a suitable orientation.
  • a common spacer domain used is the Fc of lgG1. More compact spacers can suffice e.g. the stalk from CD8a and even just the lgG1 hinge alone, depending on the antigen.
  • a trans-membrane domain anchors the protein in the cell membrane and connects the spacer to the endodomain.
  • CAR-encoding nucleic acids may be transferred to T cells using, for example, retroviral vectors.
  • retroviral vectors In this way, a large number of antigen-specific T cells can be generated for adoptive cell transfer.
  • the CAR binds the target-antigen, this results in the transmission of an activating signal to the T-cell it is expressed on.
  • the CAR directs the specificity and cytotoxicity of the T cell towards cells expressing the targeted antigen.
  • the present invention relates to a CAR signalling system in which the antigen-recognizing/antigen binding domain and transmembrane domain are provided on a first molecule (termed herein 'receptor component'), which localizes to the cell membrane.
  • the intracellular signalling domain is provided on a second, intracellular molecule (termed herein 'signaling component').
  • Certain small molecules have been described which bind simultaneously to two protein domains. Such small molecules can hence act to dimerize proteins (termed herein "chemical inducer of dimerizeration" or "CID").
  • Both the receptor component and signaling components comprise a CID binding domain (termed herein 'CID Binding Domain 1 ', or 'CBD1 ' and 'CID Binding Domain 2' or 'CBD2' respectively) which allows each receptor component and signalling component to bind simultaneously to the same CID molecule (figure 3).
  • the receptor component and signalling component are located in a stochastically dispersed manner and binding of antigen by the antigen-binding domain of the receptor component does not result in signalling through the signaling component.
  • both the receptor and signalling component of the CAR bind the CID via their CID binding domains, resulting in co-localization of the receptor component and signaling components (heterodimerization).
  • This co-localisation allows signalling through the signalling domain of the signalling component when antigen is bound by the antigen-binding domain of the receptor component.
  • 'co-localization' or 'heterodimerization' of the receptor and signalling components is analogous to ligation/recruitment of the signalling component to the receptor component via the CID agent.
  • Antigen binding by the receptor component in the absence of the CID may be termed as resulting in 'non-productive' signaling through the signaling component. Such signaling does not result in cell activation, for example T cell activation. Antigen binding by the receptor component in the presence of CID may be termed as resulting in 'productive' signaling through the signaling component. This signaling results in T- cell activation, triggering for example target cell killing and T cell activation.
  • Antigen binding by the receptor component in the presence of CID may result in signalling through the signalling component which is 2, 5, 10, 50, 100, 1 ,000 or 10,000-fold higher than the signalling which occurs when antigen is bound by the receptor domain in the absence of CID.
  • Signaling through the signaling component may be determined by a variety of methods known in the art. Such methods include assaying signal transduction, for example assaying levels of specific protein tyrosine kinases (PTKs), breakdown of phosphatidylinositol 4,5-biphosphate (PIP 2 ), activation of protein kinase C (PKC) and elevation of intracellular calcium ion concentration.
  • PTKs protein tyrosine kinases
  • PIP 2 protein kinase C
  • Functional readouts such as clonal expansion of T cells, upregulation of activation markers on the cell surface, differentiation into effector cells and induction of cytotoxicity or cytokine secretion may also be utilised.
  • the inventors determined levels of interleukin-2 (IL-2) produced by T-cells expressing a receptor component and signalling component of the CAR signalling system according to the present invention upon binding of antigen to the receptor component in the presence of varying concentrations of CID.
  • IL-2 interleukin-2
  • the CID and CID binding domains of the CAR signalling system of the first aspect of the invention may be any combination of molecules/peptides/domains which enables the selective co-localisation and dimerization of the receptor component and signalling component in the presence of the CID.
  • the CID agent is a molecule which is able to simultaneously bind to the receptor component and the signalling component.
  • the CID agent therefore comprises at least two binding moieties.
  • the CID may be any pharmaceutically acceptable molecule which can simultaneously be bound by at least two binding domains.
  • the CID is capable being delivered to the cytoplasm of a target cell and being available for intracellular binding.
  • the binding moieties of the CID may interact with identical binding domains present on the receptor component and the signalling component. That is, the CID may comprise two identical binding moieties such that it can simultaneously interact with a binding domain on the receptor component and an identical binding domain on the signalling component.
  • the CID and CID binding domains may be the FK506 binding protein (FKBP) ligand dimerization system described by Clackson et at. (PNAS; 1998; 95; 10437-10442).
  • FKBP FK506 binding protein
  • This dimerization system comprises two FKBP-like binding domains with a F36V mutation in the FKBP binding domain and a dimerization agent (AP1903) with complementary amino acid substitutions.
  • Exposing cells engineered to express FKBP-like binding domain fusion proteins to AP103 results in the dimerization of the proteins comprising the FKBP-like binding domains but no interactions involving endogenous FKBP.
  • the dimerization system described by Farrar et a/. which utilises bacterial DNA gyrase B (GyrB) binding domains and the antibiotic coumermycin as CID may also be used in the signalling system of the present invention (Methods Enzymol; 2000; 327; 421-419 and Nature; 1996; 383; 178-181 ).
  • the binding moieties of the CID may interact with different binding domains on the receptor component and the signalling component. That is, the CID may comprise two different binding moieties which can simultaneously interact with a binding domain on the receptor component and a different binding domain on the signalling component.
  • the CID and CID binding domains may comprise the dimerization system described by Belshaw et al. (Nature; 1996; 93; 4604-4607), which utilises a FK506 (Tacrolimus)/cyclosporin fusion molecule as the CID agent with FK-binding protein 12 (FKBP12) and cylcophilin A as the binding domains.
  • the CID / CID binding domain may also be the rapamycin and FKBP12/FKBP12- Rapamycin Binding (FRB) domain of mTOR system described by Rivera et al. (Nature Med; 1996; 2; 1028-1032) or the non-immunosupressive rapamycin analogs (rapalogs) and FKBP12/FRB system described by Bayle et al. (Chem Bio; 2006; 13; 99-107).
  • the CID may be C-20-methyllyrlrapamycin (MaRap) or C16(S)- Butylsulfonamidorapamycin (C16-BS-Rap), as described by Bayle et al.
  • the CID may be C16-(S)-3-methylindolerapamycin (C16-iRap) or C16-(S)-7- methylindolerapamycin (AP21976/C16-AiRap) as described by Bayle et al., in combination with the respective complementary binding domains for each.
  • dimerization systems suitable for use in the signalling system of the present invention include an estrone/biotin CID in combination with an oestrogen-binding domain (EBD) and a streptavidin binding domain (Muddana & Peterson; Org. Lett; 2004; 6; 1409-1412; Hussey ef a/.; J. Am. Chem. Soc; 125; 3692-3693); a dexamethasone/methotrexate CID in combination with a glucocorticoid-binding domain (GBD) and a dihydrofolate reductase (DHFR) binding domain (Lin ef a/.; J. Am. Chem.
  • RSL1 is a synthetic non-steroidal analogue of 20-hydroxyecdysone (see Figure 9). It is a member of a class of insecticides known as diacylhydrazines and can function to act as a non-steroidal ecdysone agonist. These molecules induce premature moulting and larvae death but are well tolerated in vertebrates.
  • RSL1 has been used in artificial transcription switches in which a two-protein transcription switch is used consisting of a fusion between the ecdysone receptor (EcR) and GAL4, and the retinoid X receptor (RXR) and VP16.
  • EcR is modified to interact specifically with RSL13
  • RXR is chimeric comprising of helices 1-8 replaced with helices 1-8 of human RXRp, and helices 9-12 from Locusta migratoria RXR.
  • the signalling system of the present invention may use EcR and RXR domains for heterdimerisation in the presence of RSL1 or a derivative thereof.
  • CID binding domains may be or comprise the sequences shown as SEQ ID NO: 1 to SEQ ID NO: 5, SEQ ID No. 25 or 26.
  • Variant sequences may have at least 80%, 85%, 90%, 95%, 98% or 99% sequence identity to SEQ ID No. 1 to 5, 25 or 26 provided that the sequences provide an effective dimerization system. That is, provided that the sequences facilitate sufficient co-localisation of the antigen-binding and intracellular signalling components, in the presence of the CID, for successful signaling to occur upon binding of the antigen- binding domain to antigen.
  • the signalling system according to the present invention is not limited by the arrangement of a specific dimerization system.
  • the receptor component may comprise either binding domain of a given dimerization system so long as the signalling component comprises the corresponding, complementary binding domain which enables the receptor component and signalling component to co-localize in the presence of the CID.
  • the present invention also relates to a method for activating the CAR signalling system of the first aspect which comprises the step of administering the CID.
  • administration of the CID results in co-localization of the receptor component and signalling component, such that signalling through the signalling domain occurs upon binding of the antigen-binding domain to antigen.
  • the CID and CID binding domains may facilitate signalling through the CAR signalling system which is proportional to the concentration of CID present.
  • the present invention further provides a method for reducing the activity of the CAR system of the first aspect which comprises reducing or stopping the administration of the CID. Reducing the level of the CID results in fewer receptor : signalling component dimers and thus the total level of signalling through the CAR signaling system is decreased in the presence of the antigen. Stopping the administration of the CID results in the absence of receptor : signalling component dimers and termination of signalling despite the presence of antigen binding to the antigen- binding domain.
  • Another possibility of this system is to "tune" the signal strength the receptor transmits. If the affinity between one or both of the CID binding domains is lessened, less CAR systems will be active and hence signal propagation upon antigen binding is lessened. This may avoid toxicity.
  • the present invention provides a receptor component comprising an antigen-binding domain, an optional spacer domain, a transmembrane domain and a CID biding domain (CBD1).
  • CBD1 CID biding domain
  • the receptor component When expressed in a cell, the receptor component localises to the cell membrane.
  • the antigen-binding domain of the molecule is orientated on the extracellular side of the membrane and the CBD is localised to the intracellular side of the membrane.
  • the receptor component therefore provides the antigen-binding function of the CAR signalling system of the present invention.
  • the antigen-binding domain is the portion of a classical CAR which recognizes antigen.
  • the antigen-binding is located within the receptor component.
  • the antigen-binding domain may comprise: a single-chain variable fragment (scFv) derived from a monoclonal antibody; a natural ligand of the target antigen; a peptide with sufficient affinity for the target; a single domain binder such as a camelid; an artificial binder single as a Darpin; or a single-chain derived from a T-cell receptor.
  • scFv single-chain variable fragment
  • tumour associated antigens are known, as shown in the following Table 1.
  • the antigen-binding domain used in the present invention may be a domain which is capable of binding a TAA as indicated therein.
  • the transmembrane domain is the sequence of a classical CAR that spans the membrane.
  • the transmembrane domain is located in the receptor component. It may comprise a hydrophobic alpha helix.
  • the transmembrane domain may be derived from CD28, which gives good receptor stability.
  • the transmembrane domain may be from CD 148, as shown in SEQ ID No. 23.
  • the receptor component of the CAR signalling system of the present invention may comprise a signal peptide so that when the receptor component is expressed in a cell, such as a T-cell, the nascent protein is directed to the endoplasmic reticulum and subsequently to the cell surface, where it is displayed.
  • the core of the signal peptide may contain a long stretch of hydrophobic amino acids that has a tendency to form a single alpha-helix.
  • the signal peptide may begin with a short positively charged stretch of amino acids, which helps to enforce proper topology of the polypeptide during translocation.
  • W the end of the signal peptide
  • signal peptidase 22 there is typically a stretch of amino acids that is recognized and cleaved by signal peptidase.
  • Signal peptidase may cleave either during or after completion of translocation to generate a free signal peptide and a mature protein.
  • the free signal peptides are then digested by specific proteases.
  • the signal peptide may be at the amino terminus of the molecule.
  • the signal peptide may comprise the SEQ ID No. 6, 7 or 8 or a variant thereof having 5, 4, 3, 2 or 1 amino acid mutations (insertions, substitutions or additions) provided that the signal peptide still functions to cause cell surface expression of the CAR.
  • the signal peptide of SEQ ID No. 6 is compact and highly efficient. It is predicted to give about 95% cleavage after the terminal glycine, giving efficient removal by signal peptidase.
  • the signal peptide of SEQ ID No. 7 is derived from lgG1.
  • the signal peptide of SEQ ID No. 8 is derived from CD8.
  • the CAR signalling system described herein may comprise a spacer sequence to connect the antigen-binding domain with the transmembrane domain in the receptor component.
  • a flexible spacer allows the antigen-binding domain to orient in different directions to facilitate binding.
  • the spacer sequence may, for example, comprise an lgG1 Fc region, an lgG1 hinge or a human CD8 stalk or the mouse CD8 stalk.
  • the spacer may alternatively comprise an alternative linker sequence which has similar length and/or domain spacing properties as an lgG1 Fc region, an lgG1 hinge or a CD8 stalk.
  • a human lgG1 spacer may be altered to remove Fc binding motifs. Examples of amino acid sequences for these spacers are given below:
  • the receptor component may comprise an appropriate number of transmembrane domains such that each CID binding domain is orientated on the intracellular side of the cell membrane ( Figure 6).
  • the receptor component may comprise 3, 5, 7, 9, 1 1 , or more transmembrane domains.
  • a single receptor component may recruit multiple signalling components amplifying signalling in response to antigen.
  • the signalling system encoded by SEQ ID No. 22 is a multi- spanning receptor with 3 FRB domains.
  • the CAR signalling system may comprise of two or more receptor components each recognizing different antigens but comprising of the same intracellular CID binding domain. Such a CAR system would be capable of recognizing multiple antigens (Figure 5a). This might be useful for instance in avoiding tumour escape.
  • the CBD1 of the receptor components differ in residues which dictate their affinity to the CID. In this way, a CAR system can be tuned such that signalling in response to one antigen is greater or lesser than response to another (figure 5b). This might be useful for instance when targeting two tumour antigens simultaneously but one is expressed at a higher density than the other. Response to this antigen could be tuned down to avoid toxicity caused by over-stimulation.
  • Methods suitable for altering the amino acid residues of the CID binding domain such that its binding affinity for CID is altered are known in the art and include substitution, addition and removal of amino acids using both targeted and random mutagenesis.
  • Methods for determining the affinity of a CID for the CID binding domain are also well known in the art and include bioinformatics prediction of protein- protein interactions, affinity electrophoresis, surface plasma resonance, bio-layer interferometry, dual polarisation interferometry, static light scattering and dynamic light scattering.
  • the signalling system encoded by SEQ ID No. 20 comprises two receptor components and one signalling component.
  • the present invention also provides a signalling component comprising a signalling domain and a CID binding domain (CBD2).
  • the signalling component is a soluble molecule and thus localises to the cytoplasm when it is expressed in a cell, for example a T cell.
  • the intracellular signalling domain is the signal-transmission portion of a classical CAR.
  • the intracellular signalling domain (signalling domain) is located in the signalling component.
  • the membrane-bound, receptor component and the intracellular signalling component are brought into proximity. After antigen recognition, receptors cluster, native CD45 and CD148 are excluded from the synapse and a signal is transmitted to the cell.
  • the signalling domain of the signalling component is analogous to the endodomain of a classical CAR molecule.
  • CD3-zeta endodomain which contains 3 ITAMs. This transmits an activation signal to the T cell after antigen is bound.
  • CD3-zeta may not provide a fully competent activation signal and additional co-stimulatory signalling may be needed.
  • chimeric CD28 and OX40 can be used with CD3-Zeta to transmit a proliferative / survival signal, or all three can be used together (illustrated in Figure 1 B).
  • the signalling component described herein comprises a signalling domain, it may comprise the CD3-Zeta endodomain alone, the CD3-Zeta endodomain with that of either CD28 or OX40 or the CD28 endodomain and OX40 and CD3-Zeta endodomain ( Figure 3A).
  • the signalling component of a CAR signalling system may comprise the sequence shown as SEQ ID No. 14, 15 or 16 or a variant thereof.
  • a variant sequence may have at least 80%, 85%, 90%, 95%, 98% or 99% sequence identity to SEQ ID No. 14, 15 or 16, provided that the sequence provides an effective intracellular signalling domain.
  • the signalling system according to the first aspect of the present invention may comprise a plurality of signalling components, each comprising a signalling domain and a CIDB2, wherein each signalling domain is bound by the same CID but the signalling domains comprise different endodomains ( Figure 7a). In this way, multiple different endodomains can be activated simultaneously. This is advantageous over a compound signalling domain since each signalling domain remains unencumbered from other signalling domains.
  • each signalling component comprises of a CBD2 domains which differ in residues which alter their affinity to CID
  • the CID enables signalling components comprising different signalling domains to ligate to the CID with differing kinetics (Figure 7b). This allows greater control over the signalling in response to antigen-binding by the receptor component as different signalling components are recruited to the receptor component in varying kinetics/dynamics. This is advantageous since rather than a fixed equal ratio of signal transmitted by a compound endodomain, an optimal T-cell activation signal may require different proportions of different immunological signals.
  • the signalling system encoded by SEQ ID No. 21 has one receptor components and three signalling components.
  • SIGNALLING COMPONENTS WITH AN ADDITIONAL CID BINDING DOMAIN In the case of very low density of target antigen expression, signalling strength may have to be amplified. Take for instance, the native T-cell receptor which is associated with multiple ITAM containing CD3 components. In such circumstances, signalling components can be modified to contain CBD2, a signalling domain and also a CBD1 ( Figure 8). In this way, addition of CID would lead to concatenation of multiple signalling components. The average size of the concatenation can be tuned by varying the amount of signalling components which have a CBD2. In the event of antigen recognition and in the presence of CID, signalling through the concatenation of multiple signalling components would be more potent than through individual signalling components.
  • the third aspect of the invention relates to a nucleic acid encoding the receptor component of the second aspect and a nucleic acid encoding a signalling component of the third aspect.
  • polynucleotide As used herein, the terms “polynucleotide”, “nucleotide”, and “nucleic acid” are intended to be synonymous with each other.
  • Nucleic acids according to the second aspect of the invention may comprise DNA or RNA. They may be single-stranded or double-stranded. They may also be polynucleotides which include within them synthetic or modified nucleotides. A number of different types of modification to oligonucleotides are known in the art. These include methylphosphonate and phosphorothioate backbones, addition of acridine or polylysine chains at the 3' and/or 5' ends of the molecule. For the purposes of the use as described herein, it is to be understood that the polynucleotides may be modified by any method available in the art. Such modifications may be carried out in order to enhance the in vivo activity or life span of polynucleotides of interest.
  • variant in relation to a nucleotide sequence include any substitution of, variation of, modification of, replacement of, deletion of or addition of one (or more) nucleic acid from or to the sequence.
  • the nucleic acid according to the third aspect of the invention may be a nucleic acid which encodes both the receptor component and the signalling component.
  • the nucleic acid may produce a polypeptide which comprises the receptor component and the signalling component joined by a cleavage site.
  • the cleavage site may be self-cleaving, such that when the polypeptide is produced, it is immediately cleaved into the receptor component and the signalling component without the need for any external cleavage activity.
  • FMDV Foot-and- outh disease virus
  • the nucleic acid may have the general formula:
  • RC is a nucleic acid sequence encoding the receptor component
  • Coexprl and coexpr2 which may be the same or different, are nucleic acid sequences allowing co-expression of the two flanking polypeptides (e.g. a sequence which encodes a cleavage site)
  • SC1 and SC2 are nucleic acid sequence encoding signalling components.
  • the signalling components encoded by SC1 and SC2 may bind the same CID, but have different endodomains.
  • the nucleic acid construct may comprise nucleic acid sequences encoding more than two signalling components e.g. SC3, SC4 ... up to SCx, each separated by a coexpression sequence, coexpr3, coexpr4....up to coexprX.
  • each component may be in any order, for example: RC-coexpr1-SC1-coexpr2-SC2; SC1 -coexprl -SC2-coexpr2-RC; or SC1 - coexprl- RC-coexpr2-SC2.
  • the nucleic acid may have the general formula:
  • RC1 and RC2 are nucleic acid sequences encoding receptor components
  • Coexprl and coexpr2 which may be the same or different, are nucleic acid sequences allowing co-expression of the two flanking polypeptides (e.g. a sequence which encodes a cleavage site)
  • SC is a nucleic acid sequence encoding the signalling component.
  • the receptor components may bind to different antigens but the same CID. They may have the same CIDB1.
  • the nucleic acid construct may comprise nucleic acid sequences encoding more than two receptor components e.g. RC3, RC4 ... up to RCx, each separated by a coexpression sequence, coexpr3, coexpr4....up to coexprX.
  • the nucleic acid sequences encoding each component may be in any order, for example: RC1 -coexprl -RC2-coexpr2-SC; RC1 -coexprl -SC-coexpr2-RC2; or SC- coexprl -RC1 -coexpr2-RC2.
  • the co-expressing sequence may be an internal ribosome entry sequence (IRES).
  • the co-expressing sequence may be an internal promoter.
  • the present invention also provides a kit comprising a nucleic acid encoding the receptor component of the second aspect and/or a nucleic acid encoding a signalling component of the third aspect.
  • the kit may also comprise a heterodimerization agent which is suitable for use in the signalling system of the first aspect.
  • the present invention also provides a vector, or kit of vectors which comprises one or more nucleic acid sequence(s) encoding a receptor component of the second aspect and/or signalling component of the third aspect of the invention.
  • a vector may be used to introduce the nucleic acid sequence(s) into a host cell so that it expresses the receptor component and signalling component of the CAR signalling system according to the first aspect of the invention.
  • the kit may also comprise a CID which is suitable for use in the signalling system of the first aspect.
  • the vector may, for example, be a plasmid or a viral vector, such as a retroviral vector or a lentiviral vector, or a transposon based vector or synthetic mRNA.
  • the vector may be capable of transfecting or transducing a T cell.
  • the present invention also relates to an immune cell comprising the CAR signalling system according to the first aspect of the invention.
  • Cytolytic immune cells can be T cells or T lymphocytes which are a type of lymphocyte that play a central role in cell-mediated immunity. They can be distinguished from other lymphocytes, such as B cells and natural killer cells (NK cells), by the presence of a T-cell receptor (TCR) on the cell surface.
  • TCR T-cell receptor
  • Helper T helper cells assist other white blood cells in immunologic processes, including maturation of B cells into plasma cells and memory B cells, and activation of cytotoxic T cells and macrophages.
  • TH cells express CD4 on their surface.
  • TH cells become activated when they are presented with peptide antigens by MHC class II molecules on the surface of antigen presenting cells (APCs).
  • APCs antigen presenting cells
  • These cells can differentiate into one of several subtypes, including TH1 , TH2, TH3, TH17, Th9, or TFH, which secrete different cytokines to facilitate different types of immune responses.
  • Cytolytic T cells destroy virally infected cells and tumor cells, and are also implicated in transplant rejection.
  • CTLs express the CD8 at their surface. These cells recognize their targets by binding to antigen associated with MHC class I, which is present on the surface of all nucleated cells.
  • MHC class I MHC class I
  • IL-10 adenosine and other molecules secreted by regulatory T cells, the CD8+ cells can be inactivated to an anergic state, which prevent autoimmune diseases such as experimental autoimmune encephalomyelitis.
  • Memory T cells are a subset of antigen-specific T cells that persist long-term after an infection has resolved. They quickly expand to large numbers of effector T cells upon re-exposure to their cognate antigen, thus providing the immune system with "memory" against past infections.
  • Memory T cells comprise three subtypes: central memory T cells (TCM cells) and two types of effector memory T cells (TEM cells and TEMRA cells). Memory cells may be either CD4+ or CD8+. Memory T cells typically express the cell surface protein CD45RO.
  • Treg cells Regulatory T cells
  • suppressor T cells are crucial for the maintenance of immunological tolerance. Their major role is to shut down T cell- mediated immunity toward the end of an immune reaction and to suppress autoreactive T cells that escaped the process of negative selection in the thymus.
  • Treg cells Two major classes of CD4+ Treg cells have been described—natural occurring Treg cells and adaptive Treg cells.
  • Naturally occurring Treg cells arise in the thymus and have been linked to interactions between developing T cells with both myeloid (CD1 1 c+) and plasmacytoid (CD123+) dendritic cells that have been activated with TSLP.
  • Naturally occurring Treg cells can be distinguished from other T cells by the presence of an intracellular molecule called FoxP3. Mutations of the FOXP3 gene can prevent regulatory T cell development, causing the fatal autoimmune disease IPEX.
  • Adaptive Treg cells also known as Tr1 cells or Th3 cells may originate during a normal immune response.
  • Natural Killer Cells are a type of cytolytic cell which form part of the innate immune system. NK cells provide rapid responses to innate signals from virally infected cells in an HC independent manner
  • NK cells (belonging to the group of innate lymphoid cells) are defined as large granular lymphocytes (LGL) and constitute the third kind of cells differentiated from the common lymphoid progenitor generating B and T lymphocytes. NK cells are known to differentiate and mature in the bone marrow, lymph node, spleen, tonsils and thymus where they then enter into the circulation.
  • LGL large granular lymphocytes
  • the CAR cells of the invention may be any of the cell types mentioned above.
  • T or NK cells expressing the molecules of the CAR signalling system according to the first aspect of the invention may either be created ex vivo either from a patient's own peripheral blood (1 st party), or in the setting of a haematopoietic stem cell transplant from donor peripheral blood (2 nd party), or peripheral blood from an unconnected donor (3 rd party).
  • T or NK cells expressing the molecules of the CAR signalling system according to the first aspect of the invention may be derived from ex vivo differentiation of inducible progenitor cells or embryonic progenitor cells to T cells.
  • an immortalized T-cell line which retains its lytic function and could act as a therapeutic may be used.
  • CAR cells are generated by introducing DNA or RNA coding for the receptor component and signalling component by one of many means including transduction with a viral vector, transfection with DNA or RNA.
  • the CAR cell of the invention may be an ex vivo T or NK cell from a subject.
  • the T or NK cell may be from a peripheral blood mononuclear cell (PBMC) sample.
  • PBMC peripheral blood mononuclear cell
  • T or NK cells may be activated and/or expanded prior to being transduced with nucleic acid encoding the molecules providing the CAR signalling system according to the first aspect of the invention, for example by treatment with an anti-CD3 monoclonal antibody.
  • the T or NK cell of the invention may be made by:
  • transduction or transfection of the T or NK cells with one or more a nucleic acid sequence(s) encoding the receptor component and/or signalling component of the CAR signalling system according to the second and third aspects of the invention.
  • the T or NK cells may then by purified, for example, selected on the basis of expression of the antigen-binding domain of the antigen-binding polypeptide.
  • the present invention also provides a kit which comprises a T or NK cell comprising the CAR signalling system according to the first aspect of the invention and a heterodimerization agent suitable for use in the signalling system.
  • the present invention also relates to a pharmaceutical composition containing a plurality of cytolytic immune cells expressing the molecules of the CAR signalling system of the first aspect of the invention.
  • the pharmaceutical composition may additionally comprise a pharmaceutically acceptable carrier, diluent or excipient.
  • the pharmaceutical composition may optionally comprise one or more further pharmaceutically active polypeptides and/or compounds.
  • Such a formulation may, for example, be in a form suitable for intravenous infusion.
  • the present invention provides a method for treating and/or preventing a disease which comprises the step of administering the cytolytic immune cells of the present invention (for example in a pharmaceutical composition as described above) and/or a CID suitable for use in a signalling system according to the first aspect of the invention to a subject.
  • a method for treating a disease relates to the therapeutic use of the cytolytic immune cells of the present invention.
  • the cells may be administered to a subject having an existing disease or condition in order to lessen, reduce or improve at least one symptom associated with the disease and/or to slow down, reduce or block the progression of the disease.
  • the method for preventing a disease relates to the prophylactic use of the cytolytic immune cells of the present invention.
  • such cells may be administered to a subject who has not yet contracted the disease and/or who is not showing any symptoms of the disease to prevent or impair the cause of the disease or to reduce or prevent development of at least one symptom associated with the disease.
  • the subject may have a predisposition for, or be thought to be at risk of developing, the disease.
  • the method may involve the steps of:
  • the methods for treating a disease may further comprise the step of administering a CID suitable for use in the signalling system of the first aspect of the invention to the subject.
  • the present invention also provides a method for treating and/or preventing a disease in a subject which subject comprises cells of the invention, which method comprises the step of administering a CID agent suitable for use in the CAR signalling system according to the first aspect to the subject.
  • this method involves administering a suitable CID agent to a subject which already comprises CAR cells of the present invention.
  • the CID may be administered in the form of a pharmaceutical composition.
  • the pharmaceutical composition may additionally comprise a pharmaceutically acceptable carrier, diluent or excipient.
  • the pharmaceutical composition may optionally comprise one or more further pharmaceutically active polypeptides and/or compounds.
  • Such a formulation may, for example, be in a form suitable for intravenous infusion.
  • the methods for treating a disease provided by the present invention may involve monitoring the progression of the disease and monitoring any toxic activity and adjusting the dose of the CID administered to the subject to provide acceptable levels of disease progression and toxic activity.
  • Monitoring the progression of the disease means to assess the symptoms associated with the disease over time to determine if they are reducing/improving or increasing/worsening.
  • Toxic activities relate to adverse effects caused by the CAR cells of the invention following their administration to a subject.
  • Toxic activities may include, for example, immunological toxicity, biliary toxicity and respiratory distress syndrome.
  • the level of signalling through the signalling system of the first aspect of the invention can be adjusted by altering the amount of CID present.
  • the level of CAR cell activation may be augmented by increasing the dose of CID administered to the subject.
  • the level of CAR cell activation may be reduced by decreasing the dose of CID administered to the subject.
  • the dose of CID administered to a subject may be altered in order to provide an acceptable level of both disease progression and toxic activity.
  • the specific level of disease progression and toxic activities determined to be 'acceptable' will vary according to the specific circumstances and should be assessed on such a basis.
  • the present invention provides a method for altering the activation level of the CAR cells in order to achieve this appropriate level.
  • the present invention provides a CAR cell of the present invention for use in treating and/or preventing a disease.
  • the invention also relates to the use of a CAR cell of the present invention in the manufacture of a medicament for the treatment and/or prevention of a disease.
  • the present invention also provides a CID agent suitable for activating a CAR signalling system according to the first aspect of the invention for use in treating and/or preventing a disease.
  • the present invention also provides a CID agent for use in activating a CAR signalling system according to the first aspect of the invention in a CAR cell.
  • the invention also provides the use of a CID suitable for activating a CAR signalling system according to the first aspect of the invention in the manufacture of a medicament for the treatment and/or prevention of a disease.
  • the invention also provides the use of a CID in the manufacture of a medicament for in activating a CAR signalling system according to the first aspect of the invention in a CAR cell.
  • the disease to be treated and/or prevented by the methods of the present invention may be an infection, such as a viral infection.
  • the methods of the invention may also be for the control of pathogenic immune responses, for example in autoimmune diseases, allergies and graft-vs-host rejection.
  • the methods may be for the treatment of a cancerous disease, such as bladder cancer, breast cancer, colon cancer, endometrial cancer, kidney cancer (renal cell), leukemia, lung cancer, melanoma, non-Hodgkin lymphoma, pancreatic cancer, prostate cancer and thyroid cancer.
  • a cancerous disease such as bladder cancer, breast cancer, colon cancer, endometrial cancer, kidney cancer (renal cell), leukemia, lung cancer, melanoma, non-Hodgkin lymphoma, pancreatic cancer, prostate cancer and thyroid cancer.
  • the CAR cells of the present invention may be capable of killing target cells, such as cancer cells.
  • the target cell may be recognisable by expression of a TAA, for example the expression of a TAA provided above in Table 1.
  • the CAR cells and pharmaceutical compositions of present invention may be for use in the treatment and/or prevention of the diseases described above.
  • the CAR cells and pharmaceutical compositions of present invention may be for use in any of the methods described above.
  • the present invention also provides a method of administering a cell according to the ninth aspect of the invention to a subject in need of same.
  • the present invention also provides a method of administering a CID as defined herein to a subject who comprises a cell according to the ninth aspect of the invention.
  • T-cells were transduced with a retroviral vector coding for an inducible CAR detailed in figure 3 and the sequence of which is shown as SEQ ID 19.
  • This inducible CAR has a single receptor component and a single signalling component.
  • the receptor component comprises of an anti-CD19 scFv, a spacer domain derived from human lgG1 , a transmembrane-domain derived from CD28 and FKBP12 as endodomain.
  • the signalling component comprises of the FRB domain from mTOR and a signalling domain derived from endodomains of CD28, OX40 and CD3-Zeta.
  • Expression of the CAR was determined by staining the transduced cells with a conjugated antibody recognizing the spacer domain and analysing by flow cytometry.
  • the T-cells were incubated with either target cells expressing the cognate target CD19 in ratio of 2:1 , 1 : 1 and 0:1 (targeteffector) in the absence of rapamycin or at increasing concentrations. Signalling only occurred in the presence of both the cognate antigen and rapamycin. A dose response to rapamycin and target antigen was observed.
  • RSL1 is a synthetic non-steroidal analogue of 20-hydroxyecdysone ( Figure 9). It is a member of a class of insecticides known as diacylhydrazines and can function to act as a non-steroidal ecdysone agonist. These molecules induce premature moulting and larvae death but are well tolerated in vertebrates (Dhadialla et al (1998) Annu. Rev. Entomol. 43, 545-569).
  • RSL1 has been used in artificial transcription switches (Lessard et al (2007) The Prostate 67, 808-819) whereby a two-protein transcription switch consisting of a fusion between the ecdysone receptor (EcR) and GAL4, and the retinoid X receptor (RXR) and VP16.
  • EcR is modified to interact specifically with RSL1 (Kumar et al (2004) J. Biol. Chem. 279, 2721 1-27218), and RXR is chimeric comprising of helices 1-8 replaced with helices 1-8 of human RXR , and helices 9-12 from Locusta migratoria RXR.
  • the present inventors have constructed a split CD19 CAR based on the CD19 system described in Example 1 with the heterodimerization motif on the antigen recognition component and signalling components being EcR and RXR respectively ( Figure 10a and SEQ ID No. 23).
  • This CAR was transduced into T-cells and found to express well ( Figure 10b).
  • These T-cells were challenged with targets which were CD19 negative and CD19 positive.
  • Example 3 An AP1903-inducible CAR system
  • AP1903 is a synthetic chemical inducer of dimerization which binds to two mutated FKB12 domains (Clackson et al (1998). Proc. Natl. Acad. Sci. U. S. A. 95, 10437- 10442).
  • An inducible CAR was developed based on this system, using the modified FKBP12 domains to constitute the heterodimerization domain of both the antigen recognition component and signalling component in the framework as described in Example 1 with the antigen recognition domain and the signalling domain co- expressed with a FMD-2A like sequence ( Figure 1 1 a and SEQ ID No. 24).
  • the FKBP12 domains were codon-wobbled to prevent homologous recombination.
  • T-cells were transduced with this construct and challenged with target cells which were CD19 negative or positive.
  • T-cells responded only to CD19 positive targets in the presence of AP1903 ( Figure 1 1 b).
  • AP1903 At very high concentration of dimerizer, signalling was inhibited likely due to saturating conditions where the stoichiometry favours one FKBP12 binding a single dimerizer molecule rather than one dimerizer molecule binding two FKBP12 domains ( Figure 11 c).
  • Simple inducible CAR comprised of one receptor component and one signalling component.
  • the receptor component comprises of a signal peptide, an anti- CD19 scFv, a spacer from the Fc domain of human lgG1 , a CD28 transmembrane domain and FKB12 as endodomain.
  • a FMD-2A peptide separates this receptor component from the signalling component.
  • the signalling component comprises of FRB and a compound of endodomains from CD28, OX40 and CD3-Zeta.
  • aCD19 heavy chain > caccagcagc ctgagcgcca gcctgggcga ccgggtgacc atcagctgca gagccagcca ggacatcagc aagtacctg > aCD19 heavy chain
  • Inducible CAR comprised of two receptor component and one signalling component.
  • the first receptor component comprises of a signal peptide, an anti-CD19 scFv, a spacer from the stalk region of CD8, a transmembrane domain from CD28, and FKB12 as an endodomain.
  • the second signalling component comprises of an anti-CD29 scFv, spacer derived from the Fc domain of human lgG1 , a CD28 transmembrane domain and FKB12 as endodomain.
  • the signalling component comprises of FRB and a compound of endodomains from CD28, OX40 and CD3-Zeta.
  • a foot-and-mouth disease 2A peptide separates the components.
  • cagcagcccc aagcctgga tctacgccac cagcaacctg gccagcggcg tgcccgtgcg gttcagcggc agcggcagcg > aCDZO scFv >
  • Inducible CAR comprised of one receptor component and three signalling components.
  • the receptor component comprises of a signal peptide, an anti-CD19 scFv, a spacer derived from the Fc domain of human lgG1 , a CD28 transmembrane domain and FKB12 as endodomain.
  • the first signalling component comprises of FRB and endodomain from CD3 Zeta, the second comprises FRB and CD28 endodomain, the third FRB and 41 BB endodomain.
  • a foot-and-mouth disease 2A peptide separates the components.
  • scFv anti-CD19 721 atctactact gcgccaagca ctactactat ggcggcagct acgctatgga ctactggggc cagggcacca gcgtgaccgt >. scFv anti-CD19 > 721 atctactact gcgccaagca ctactactat ggcggcagct acgctatgga ctactggggc cagggcacca gcgtgaccgt >. scFv anti-CD19 > 721 atctactact gcgccaagca ctactactat ggcggcagct acgctatgga ctactggggc cagggcacca gcgtgaccgt >. scFv anti-CD19 > 721 atctactact gcgccaagca ctactactat ggcggcagct ac
  • the receptor component comprises of a signal peptide, an anti-CD19 scFv, a spacer from the Fc domain of human lgG1 , a CD28 transmembrane domain, a first FRB, then the first trans-membrane, minor extracellular loop and second transmembrane domain of CD20, then a second FRB, then the third transmembrane domain, the major extracellular loop and the fourth transmembrane domain of CD20, and a third FRB.
  • the signalling component comprises of FRB and a compound of endodomains from CD28, OX40 and CD3-Zeta. A foot-and-mouth disease 2A peptide separates the two components.
  • HCH2CH3pvaa 1041 gcgggaggag cagtacaaca gcacgtaccg tgtggtcagc gtcctcaccg tcctgcacca ggactggctg aatggcaagg > HCH2CH3pvaa >
  • 3601 cggaccccca tccaagagga gcaggccgac gcccactcca ccctggccaa gatcagagtg aagttcagca ggagegcaga > 280XZ > 3681 cgcccccgcg taccagcagg gccagaacca gctctataac gagctcaatc taggacgaag agaggagtac gatgttttgg > 280XZ >
  • SEQ ID No. 23 Simple inducible CAR comprised of one receptor component and one signalling component.
  • the signalling component comprises an ecdysone receptor and the CD3-Zeta endodomain (italic).
  • a FMD-2A peptide (shown in bold) separates this signalling component from the receptor component.
  • the receptor component comprises an anti-CD19 scFv, a HCH2CH3 spacer (underlined), a CD148 transmembrane domain (highlighted in grey) and the retinoid X receptor as endodomain.
  • Simple inducible CAR comprised of one receptor component and one signalling component.
  • the signalling component comprises an FKBP12 domain (underlined) and the CD3-Zeta endodomain (italic).
  • a FMD-2A peptide (shown in bold) separates this signalling component from the receptor component.
  • the receptor component comprises an anti-CD19 scFv, a HCH2CH3 spacer (highlighted in grey), a CD148 transmembrane domain and the and FKBP12 domain (underlined).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Wood Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Toxicology (AREA)
  • Mycology (AREA)
  • Hematology (AREA)
  • Endocrinology (AREA)
  • Oncology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention relates to a chimeric antigen receptor (CAR) signalling system comprising; (i) a receptor component comprising an extracellular antigen-binding domain, a transmembrane domain and a intracellular first chemical inducer of dimerization binding domain 1 (CBD1 ); and (ii) an intracellular signalling component comprising a signalling domain and a second chemical inducer of dimerization binding domain 2 (CBD2); wherein CBD1 and CBD2 are capable of simultaneously binding to a chemical inducer of dimerization (CID); wherein, in the absence of the CID, binding of the antigen-binding component to antigen does not result in signalling through the signalling component; whilst, in the presence of the CID, the receptor component and the signalling component heterodimerize and binding of the antigen-binding domain to antigen results in signalling through the signalling domain.

Description

CHIMERIC ANTIGEN RECEPTOR (CAR) SIGNALLING SYSTEM
FIELD OF THE INVENTION
The present invention relates to an antigen receptor signalling system. BACKGROUND TO THE INVENTION
Traditionally, antigen-specific T-cells have been generated by selective expansion of peripheral blood T-cells natively specific for the target antigen. However, it is difficult and quite often impossible to select and expand large numbers of T-cells specific for most cancer antigens. Gene-therapy with integrating vectors affords a solution to this problem as transgenic expression of Chimeric Antigen Receptor (CAR) allows generation of large numbers of T-cells specific to any surface antigen by ex vivo viral vector transduction of a bulk population of peripheral blood T-cells.
Chimeric antigen receptors are proteins which graft the specificity of a monoclonal antibody (mAb) to the effector function of a T-cell. Their usual form is that of a type I transmembrane domain protein with an antigen recognizing amino terminus, a spacer, a transmembrane domain all connected to a compound endodomain which transmits T-cell survival and activation signals (see Figure 1A).
The most common forms of these molecules are fusions of single-chain variable fragments (scFv) derived from monoclonal antibodies which recognize a target antigen, fused via a spacer and a trans-membrane domain to a signalling endodomain. Such molecules result in activation of the T-cell in response to recognition by the scFv of its target. When T cells express such a CAR, they recognize and kill target cells that express the target antigen. Several CARs have been developed against tumour associated antigens, and adoptive transfer approaches using such CAR-expressing T cells are currently in clinical trial for the treatment of various cancers.
A number of toxicities have been reported from CAR studies, and additional theoretical toxicities exist. Such toxicities include immunological toxicity caused by sustained intense activation of the CAR T-cells resulting in a macrophage activation syndrome (MAS) and "On-target off-tumour" toxicity i.e. recognition of the target antigen on normal tissues.
MAS is presumed to be caused by persistent antigen-driven activation and proliferation of T-cells which in turn release copious inflammatory cytokines leading to hyper-activation of macrophages and a feed-forward cycle of immune activation. A large spike in serum IL-6 is characteristic and the syndrome can result in a severe systemic illness requiring ICU admission.
On-target off-tumour toxicity has been reported with other CARs, for example a group of patients treated with a CAR against the renal cell carcinoma antigen CAIX developed unexpected and treatment limiting biliary toxicity. Two fatalities have been reported with CAR studies: one patient died of a respiratory distress syndrome which occurred immediately post-infusion of a large dose of 3rd generation anti-ERBB2 CAR T-cells; a further patient died in a different study after a possible cytokine storm following treatment of CLL with a second generation anti-CD19 CAR.
These toxicities are very difficult to predict even with detailed animal studies or non- human primate work. Crucially, unlike small molecules and biologies, CAR T-cells do not have a half-life and one cannot cease administration and wait for the agent to breakdown/become excreted. CAR T-cells are autonomous and can engraft and proliferate. Toxicity can therefore be progressive and fulminant.
Suicide genes are genetically expressed elements which can conditionally destroy cells which express them. Examples include Herpes-simplex virus thymidine kinase, which renders cells susceptible to Ganciclovir; inducible Caspase 9, which renders cells susceptible to a small molecular homodimerizer and CD20 and RQR8, which renders cells susceptible to Rituximab.
This technology adds a certain amount of safety to CAR T-cell therapy, however there are limitations. Firstly, it is a binary approach wherein all the CAR T-cells are destroyed upon addition of the suicide entity. In addition, medicinal therapeutics often have a therapeutic window. With a suicide gene the potency of the product cannot be tuned such that efficacy with tolerable toxicity can be achieved. Secondly, it is not clear whether a suicide gene would help with some of the immune-toxicities described above: for instance by the time a macrophage activation syndrome had been triggered, it may well no longer need the CAR T-cells to perpetuate and the suicide gene would no longer be helpful. The more acute cytokine release syndromes probably occur too quickly for the suicide gene to work. Finally, suicide genes are not "fail-safe", i.e. the default status is for the CAR T-cells to be active. There is thus a need for alternative methods for controlling CAR T-cells that are not associated with the disadvantages and problems mentioned above.
DESCRIPTION OF THE FIGURES
Figure 1 - a) Schematic diagram illustrating a classical CAR. (b) to (d): Different generations and permutations of CAR endodomains: (b) initial designs transmitted ITAM signals alone through FceRI -γ or ΟΌ3ζ endodomain, while later designs transmitted additional (c) one or (d) two co-stimulatory signals in the same compound endodomain.
Figure 2 - Structures of Rapamycin analogues.
Figure 3 - Illustration of an example of an inducible CAR signalling system in its simplest incarnation, (a) Receptor component comprising of an extracellular scFv, a spacer, a trans-membrane domain and an intracellular FKBP12. (b) Signalling component: intracellular fusion between FRB fragment of mTOR and the endodomains of CD28, OX40 and CD3 Zeta. (c) A chemical inducer of dimerization (CID), in this case is Rapamycin or an analogue, (d) Receptor component. In the presence of CID, the receptor component and the signalling components dimerize allowing signalling in the presence of cognate antigen.
Figure 4 - Function of an inducible CAR system. T-cells were transduced with a CAR system as illustrated in figure 3 and with SEQ ID No. 19. This CAR recognizes CD19. CAR expressing T-cells were challenged with CD19+ targets at 1 : 1 and 2: 1 effector to target ratios in the presence of increasing concentration of CID. T-cells respond to target cells only in the presence of CID. Stimulation with PMA/lonomycin is also show. Response to CD19+ targets of T-cells expressing a standard CD19 CAR (MP15577) to different effector target ratios is also shown.
Figure 5 - Illustration of an inducible CAR system with two receptor components, (a) Two receptor components are co-expressed along with a single signalling component. The two receptor components each have a different single chain which recognizes a different target. The receptor components have endodomains comprising of the FRB fragment of mTOR. A single signalling component comprises of FKBP12 and a fusion of CD28, OX40 and CD3 Zeta endodomains. In (a), the FRB sequences are identical and this CAR will signal equally in response to either target. In (b), the FRB sequences differ such that each has a different affinity for the CID. With this implementation, the CAR will signal with different strength in response to each cognate antigen in the presence of CID.
Figure 6 - Illustration of an example of a multiple transmembrane receptor domain receptor component for use in an inducible CAR signalling system. The receptor component is a chimera of a type I CAR linked to the multi-span protein. A scFv is connected to an Fc spacer domain and a TM domain. This in turn is connected to the CD20 molecule with truncated amino and carboxy termini. Each intracellular entry is attached to an FRB domain. The signalling component comprises of FKBP12 and a fusion of CD28, OX40 and CD3 Zeta endodomains. Addition of the CID allows the stochastic ligation of multiple signalling domains to the multiple endodomains which recognize a single antigen receptor component, resulting in amplified signalling in response to antigen.
Figure 7 - Illustration of an inducible CAR system with multiple signalling component inducible CAR. This CAR system has a single receptor component comprising of a scFv, an Fc spacer, a TM domain and a FKBP12 endodomain. The three different signalling domains comprise of a fusion between the FRB fragment of mTOR and the CD28 endodomain, a fusion between the FRB fragment of mTOR and the 41 BB endodomain and a fusion between the FRB fragment of mTOR and the CD3 Zeta endodomain. In (a), the FRB domains are identical so that each signalling component is recruited equally to the receptor component and the CAR transmits an equal CD28, 41 BB and CD3 Zeta signal in the presence of CID and upon recognition of the cognate antigen. In (b), the FRB domains are different so that each signalling component can be recruited differently to the receptor component so that the CAR transmits more or less of each of CD28, 41 BB and CD3 Zeta signals depending on the optimum needed for the application.
Figure 8 - Illustration of an inducible CAR with signalling component which comprises of two CID binding domains. In this example, there is one receptor component which comprises of a scFv, and Fc spacer, a transmembrane domain and an FKBP12 endodomain. The signalling domain comprises of the FRB domain of mTOR, a fusion of CD28, OX40 and Zeta endodomains and an FKBP12 domain. In the presence of CID, the signalling components multimerize with each other and with the receptor component. Upon antigen recognition, a signal equivalent to the sum of the signalling component multimer is transmitted. Figure 9 - Chemical structure of RSL1 , the synthetic diacylhydrazine (N-(2-ethyl-3- methoxybenzoyl)-N'-(3,5-dimethylbenzoyl)-N'-tert-butylhydrazine. RSL1 is one of a family of compounds that have been found to act as non-steroidal ecdysone agonists and can function as gene inducers.
Figure 10 - RSL1 inducible CAR (a) an RSL1 inducible CAR was constructed by using retinoic acid receptor domains and ecodysone receptor domains to constitute the heterodimerization motifs for the antigen recognition component and the signalling component respectively. The antigen recognition component recognizes CD19. (b) T- cells expressing this CAR along with non-transduced T-cells were stained with anti-Fc which recognizes the CAR spacer. The transduced T-cells expressed the CAR well, (c) Transduced T-cells were next challenged with increasing concentration of RSL1 and challenged with either CD19 negative or CD19 positive target cells. T-cells challenged with CD19 positive targets signal only in the presence of RSL1.
Figure 11 - AP1903 homodimerizer inducible CAR. (a) AP1903 homodimerizer inducible CAR was constructed by using modified FKBP12 domains to constitute the heterodimerization motifs for both the antigen recognition component and the signalling component. The antigen recognition component recognizes CD19. (b) T- cells expressing this CAR along with non-transduced T-cells were stained with anti-Fc which recognizes the CAR spacer. The transduced T-cells expressed the CAR well, (c) Transduced T-cells were next challenged with increasing concentration of CID dimerizer AP1903 and challenged with either CD19 negative or CD19 positive target cells. T-cells challenged with CD19 positive targets signal only in the presence of CID. At high CID concentrations, signalling is reduced likely to saturation of heterodimerization domains with an excess of CID reducing the formation of heterodimers.
SUMMARY OF ASPECTS OF THE INVENTION
The present inventors have found that it is possible to separate the antigen- recognition and signalling components of a CAR to produce an inducible system, where signalling only takes place in the presence of an agent such as a small molecule which can induce dimerization (henceforth referred to as chemical inducer of dimerization or CID). The CID binds two protein domains simultaneously. By incorporating a CID binding domain into each of two non-functional halves of a CAR, the presence of CID will reunite the halves constituting a functional whole.
Functional signalling by the CAR is hence dependent on the presence of the CID. This provides a mechanism for controlling the CAR T-cell after it has been administered to a patient: The CAR T-cell can be remotely switched on by administering the CID to the patient, and in case of toxicity switched off by not administering further CID. If activity of the CAR is proportional to local concentration of the agent (for instance when CID concentration is not saturating) activity of the CAR T-cells can be "tuned" by altering CID dose administered to the patient.
In a first aspect, the present relates to a chimeric antigen receptor (CAR) signalling system comprising of two separate protein components:
(i) A membrane spanning receptor component comprising an extracellular antigen-binding domain, a transmembrane domain and a first intracellular CID binding domain (referred henceforth as CBD1); and
(ii) An intracellular signalling component comprising at a minimum a signalling domain and a second CID binding domain (henceforth referred to as CBD2);
wherein CBD1 and CBD2 are capable of simultaneously and specifically binding to the CID.
Thus, in a first aspect, the present invention provides a chimeric antigen receptor (CAR) signalling system comprising;
(i) a receptor component comprising an extracellular antigen-binding domain, a transmembrane domain and a intracellular first chemical inducer of dimerization (CID) binding domain (CBD1); and
(ii) an intracellular signalling component comprising a signalling domain and a second CID binding domain (CBD2);
wherein CBD1 and CBD2 are capable of simultaneously binding to a CID;
wherein, in the absence of CID, binding of the antigen-binding domain to antigen does not result in signalling through the signalling domain; whilst, in the presence of CID, the receptor component and the signalling component heterodimerize and binding of the antigen-binding domain to antigen results in signalling through the signalling domain. CBD1 and CBD2 of the CAR signalling system according to the first aspect may comprise different CID binding domains and the CID may comprise two different binding moieties.
CDB1 may comprise the rapamycin binding domain of FK-binding protein 12 (FKBP12), CDB2 may comprise the FKBP12-Rapamycin Binding (FRB) domain of mTOR, or visa-versa. In this case, the CID may comprise rapamycin or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
CBD1 may comprise the FK506 (Tacrolimus) binding domain of FK-binding protein 12 (FKBP12), CBD2 may comprise the cyclosporin binding domain of cylcophilin A (or visa versa). In this case, the CID may comprise an FK506/cyclosporin fusion protein or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
CBD1 may comprise an oestrogen-binding domain (EBD), CBD2 may comprise a streptavidin binding domain, or visa-versa. In this case, the CID may comprise an estrone/biotin fusion protein or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
CBD1 may comprise a glucocorticoid-binding domain (GBD), CBD2 may comprise a dihydrofolate reductase (DHFR) binding domain, or visa-versa. In this case, the CID may comprise a dexamethasone/methotrexate fusion molecule or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
CBD1 may comprise an 06-alkylguanine-DNA alkyltransferase (AGT) binding domain, CBD2 may comprise a dihydrofolate reductase (DHFR) binding domain, or vice- versa. In this case, the CID may comprise an 06-benzylguanine derivative/methotrexate fusion molecule or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
CBD1 may comprise a retinoic acid receptor domain, CBD2 may comprise an ecodysone receptor domain, or vice-versa. In this case, the CID may comprise RSL1 or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize. CBD1 and CBD2 of the CAR signalling system according to the first aspect of the invention may comprise the same CID binding domain and the CID may comprise two identical binding moieties.
CBD1 and CBD2 of the CAR signalling system according to the first aspect of the invention may comprise FK506 binding protein (FKBP12) binding domains comprising a F36V mutation and the CID may comprise AP1903 or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
In a second embodiment, the CAR may comprise a receptor component which spans the membrane multiple times with each intracellular portion constituting a CBD1. Hence a single receptor component may recruit multiple signalling components hence amplifying signal.
In a third embodiment, the CAR may comprise a set of separate receptor components each with an intracellular CBD1 such that in the presence of CID, signalling components connect to each receptor protein component so that the CAR can signal in response to a multiplicity of cognate antigens.
The CID binding domains may be altered such that affinity to the CID differs. For instance altering amino acids at Positions 2095, 2098, and 2101 of FRB can alter binding to Rapamycin: KTW has high, KHF intermediate and PLW is low (Bayle et al, Chemistry & Biology 13, 99-107, January 2006)
Hence, in the fourth embodiment, the CAR may comprise multiple receptor components such that each one has a CBD1 domain of different affinity to the CID. In this way, in the presence of CID, signalling protein components connect to each receptor protein component differentially and hence the CAR signal strengths differentially in response to each cognate antigen.
In the above embodiment, the signalling domain of the signalling protein component may comprise of a single endodomain selected from activating or co-stimulating T-cell signalling receptor such as CD3 zeta, CD28, 41 BB or OX40. In the above embodiments, the signalling domain of the signalling protein component may comprise of a multiplicity of endodomains selected from activating and co- stimulating T-cell signalling receptor such as CD3 Zeta, CD28, 41 BB and OX40
If an inhibitory CAR is desired, in the above aspects, the signalling domain of the signalling protein may comprise the endodomain of an inhibitory T-cell signalling receptor or a phosphatase.
In a fifth embodiment, the signalling component according to the above embodiments of the invention may comprise a plurality of signalling components, each comprising a different signalling domain and a similar CBD2, wherein the CID agent binding domains each recognise the same CID, but the signalling domains comprise different endodomains. In this way, the CAR can transmit multiple signals but the signalling components being in trans rather than cis are sterically unencumbered from each other's signalling transmission molecules unlike a single signalling component containing a multiplicity of endodomains.
In a sixth embodiment, the CBD2 of the multiple signalling domains in the aspect described above may bind to the CID molecule with different affinities such that the relative contribution of each signalling component can be tuned. Hence the CAR can transmit a more complex signal rather than the fixed stoichiometry of a single signalling component containing a multiplicity of endodomains.
In a seventh embodiment, the CAR systems described above may comprise signalling domains which contains a CBD1 , single or multiple endodomains as well as CBD2. Hence, in the presence of CID, concatenation of multiple signalling domains together to amplify signals.
In a second aspect, the present invention provides a receptor component suitable for use in the CAR signalling system of the first aspect of the invention which comprises an antigen-binding domain, a transmembrane domain and a CID binding domain.
In a third aspect, the present invention provides a signalling component suitable for use in the CAR signalling system according to the first aspect of the invention which comprises a signalling domain and a CID binding domain. In a fourth aspect, the present invention provides a nucleic acid encoding the receptor component according to the second aspect of the invention.
In a fifth aspect, the present invention provides a nucleic acid encoding the signalling component according to the third aspect of the invention.
In a sixth aspect, the present invention provides a nucleic acid sequence encoding the receptor component according to the second aspect of the invention and the signalling component according to the third aspect of the invention, wherein the expressed molecule is a self-cleaving peptide which is cleaved between the receptor component and the signalling component.
In a seventh aspect, the present invention provides a vector comprising a nucleic acid sequence according to any of the fourth to sixth aspects of the invention.
In an eighth aspect, the present invention provides a retroviral vector or a lentiviral vector or a transposon comprising a vector according to the seventh aspect of the invention.
In a ninth aspect, the invention provides a T cell or NK cell which expresses a receptor component according to the second aspect of the invention and at least one signalling component according to the third aspect of the invention.
The T cell or NK cell according to the ninth aspect of the invention may comprise the nucleic acid according to any of the fourth to sixth aspects of the invention or the vector according to the seventh or eighth aspects of the invention.
In a tenth aspect, the present invention provides a pharmaceutical composition comprising a plurality of T cells or NK cells according to the ninth aspect of the invention.
In an eleventh aspect, the present invention provides a pharmaceutical composition according to the tenth13th aspect of the invention for use in treating and/or preventing a disease. In a twelfth aspect, the invention provides a method for treating and/or preventing a disease, which comprises the step of administering a pharmaceutical composition according to the tenth aspect of the invention to a subject.
The method according to the twelfth aspect of the invention may comprise the following steps:
(i) isolation of a T or NK cell-containing sample from a subject;
(ii) transduction or transfection of the T or NK cells with a nucleic acid sequence according to any the fourth to sixth aspects or a vector according to the seventh or eighth aspects; and
(iii) administering the cells from (ii) to the subject.
The method according to the twelfth aspect of the invention may further comprise the step of administering a CID suitable for use in the CAR signalling system according to the first aspect to the subject.
In a thirteenth, the present invention provides a method for treating and/or preventing a disease in a subject which subject comprises T cells according to the ninth aspect of the invention, which method comprises the step of administering a CIFD suitable for use in the CAR signalling system according to the first aspect of the invention to the subject.
The method according to the twelfth or thirteenth aspect of the invention may involve monitoring the progression of disease and/or monitoring toxic activity in the subject and adjusting the dose of the CID to provide acceptable levels of disease progression and/or toxic activity.
The disease to be prevented and/or treated according to the eleventh, twelfth or thirteenth aspect of the invention may be cancer.
In a fourteenth aspect, the present invention provides the use of a pharmaceutical composition according to the tenth aspect of the invention in the manufacture of a medicament for the treatment and/or prevention of a disease.
In a fifteenth aspect, the present invention provides a kit which comprises a nucleic acid according to any of the fourth to sixth aspects of the invention or a vector according to the seventh or eighth aspects of the invention. The kit may also comprise a CID suitable for use in the CAR signalling system according to the first aspect of the invention.
In a sixteenth aspect, the invention provides a kit which comprises a T cell or NK cell according to the ninth aspect of the invention and a CID suitable for use in the CAR signalling system according to the first aspect of the invention.
In a seventeenth aspect, the present invention provides a method for making a T cell or NK cell according to claim ninth aspect of the invention, which comprises the step of introducing: a nucleic acid sequence according to any of the fourth to sixth aspects of the invention or a vector according to the seventh or eighth aspects of the invention, into a T or NK cell.
The T or NK cell may be from a sample isolated from a subject.
In an eighteenth aspect, the invention provides a method for activating the CAR signalling system according to the first aspect of the invention in a subject which comprises a T-cell according to the ninth aspect of the invention, which method comprises the step of administering the CID to the subject.
In a nineteenth aspect the invention provides a method for reducing the activity of the CAR signalling system according to the first aspect of the invention, in a subject which comprises a T-cell or NK cell according to the ninth aspect of the invention, which method comprises reducing or stopping administration of the CID to the subject.
In a twentieth aspect the present invention provides a method for activating signalling in a CAR signalling system according to the first aspect of the invention in a T cell or NK cell according to the ninth aspect of the invention in culture, which comprises the step of introducing the CID agent to the T or NK cell culture.
In a twenty-first aspect the present invention provides a method for reducing or stopping signalling via a CAR signalling system according to the first aspect of the invention in a T or NK cell according to the ninth aspect of the invention in culture, which comprises the step of removing the CID from the T or NK cell culture. In a twenty-second aspect, there is provided a method for inducing dimerization in vivo between a receptor component according to the second aspect of the invention and a signalling component according to the third aspect of the invention, which comprises the step of administering a chemical inducer of dimerization (CID) to a subject comprising a cell according to the ninth aspect of the invention.
The present invention therefore provides a means of tuning the CAR activity to the presence of an agent, such as a small molecule. This allows the potency of CAR cells to be controlled pharmacologically and tuned to an acceptable balance between achieving the desired therapeutic effect, while avoiding CAR-associated toxicities.
DETAILED DESCRIPTION
CHIMERIC ANTIGEN RECEPTORS (CARs)
Classical CARs, which are shown schematically in Figure 1 , are chimeric type I transmembrane proteins which connect an extracellular antigen-recognizing domain (binder) to an intracellular signalling domain (endodomain). The binder is typically a single-chain variable fragment (scFv) derived from a monoclonal antibody (mAb), but it can be based on other formats which comprise an antibody-like antigen binding site. A spacer domain may be necessary to isolate the binder from the membrane and to allow it a suitable orientation. A common spacer domain used is the Fc of lgG1. More compact spacers can suffice e.g. the stalk from CD8a and even just the lgG1 hinge alone, depending on the antigen. A trans-membrane domain anchors the protein in the cell membrane and connects the spacer to the endodomain.
Early CAR designs had endodomains derived from the intracellular parts of either the γ chain of the FceR1 or ΟΌ3ζ. Consequently, these first generation receptors transmitted immunological signal 1 , which was sufficient to trigger T-cell killing of cognate target cells but failed to fully activate the T-cell to proliferate and survive. To overcome this limitation, compound endodomains have been constructed: fusion of the intracellular part of a T-cell co-stimulatory molecule to that of CD3 results in second generation receptors which can transmit an activating and co-stimulatory signal simultaneously after antigen recognition. The co-stimulatory domain most commonly used is that of CD28. This supplies the most potent co-stimulatory signal - namely immunological signal 2, which triggers T-cell proliferation. Some receptors have also been described which include TNF receptor family endodomains, such as the closely related OX40 and 41 BB which transmit survival signals. Even more W
14 potent third generation CARs have now been described which have endodomains capable of transmitting activation, proliferation and survival signals.
CAR-encoding nucleic acids may be transferred to T cells using, for example, retroviral vectors. In this way, a large number of antigen-specific T cells can be generated for adoptive cell transfer. When the CAR binds the target-antigen, this results in the transmission of an activating signal to the T-cell it is expressed on. Thus the CAR directs the specificity and cytotoxicity of the T cell towards cells expressing the targeted antigen.
In a first aspect, the present invention relates to a CAR signalling system in which the antigen-recognizing/antigen binding domain and transmembrane domain are provided on a first molecule (termed herein 'receptor component'), which localizes to the cell membrane. The intracellular signalling domain is provided on a second, intracellular molecule (termed herein 'signaling component'). Certain small molecules have been described which bind simultaneously to two protein domains. Such small molecules can hence act to dimerize proteins (termed herein "chemical inducer of dimerizeration" or "CID"). Both the receptor component and signaling components comprise a CID binding domain (termed herein 'CID Binding Domain 1 ', or 'CBD1 ' and 'CID Binding Domain 2' or 'CBD2' respectively) which allows each receptor component and signalling component to bind simultaneously to the same CID molecule (figure 3).
When antigen binds to the antigen-binding domain of the receptor component in the absence of the CID, there is no signalling through the signaling component. When the CID is present, receptor binding of antigen to the antigen-binding domain of the receptor component results in signaling through the signalling component.
Specifically, in the absence of CID, the receptor component and signalling component are located in a stochastically dispersed manner and binding of antigen by the antigen-binding domain of the receptor component does not result in signalling through the signaling component. In the presence of the CID, both the receptor and signalling component of the CAR bind the CID via their CID binding domains, resulting in co-localization of the receptor component and signaling components (heterodimerization). This co-localisation allows signalling through the signalling domain of the signalling component when antigen is bound by the antigen-binding domain of the receptor component. Herein 'co-localization' or 'heterodimerization' of the receptor and signalling components is analogous to ligation/recruitment of the signalling component to the receptor component via the CID agent.
Antigen binding by the receptor component in the absence of the CID may be termed as resulting in 'non-productive' signaling through the signaling component. Such signaling does not result in cell activation, for example T cell activation. Antigen binding by the receptor component in the presence of CID may be termed as resulting in 'productive' signaling through the signaling component. This signaling results in T- cell activation, triggering for example target cell killing and T cell activation.
Antigen binding by the receptor component in the presence of CID may result in signalling through the signalling component which is 2, 5, 10, 50, 100, 1 ,000 or 10,000-fold higher than the signalling which occurs when antigen is bound by the receptor domain in the absence of CID.
Signaling through the signaling component may be determined by a variety of methods known in the art. Such methods include assaying signal transduction, for example assaying levels of specific protein tyrosine kinases (PTKs), breakdown of phosphatidylinositol 4,5-biphosphate (PIP2), activation of protein kinase C (PKC) and elevation of intracellular calcium ion concentration. Functional readouts, such as clonal expansion of T cells, upregulation of activation markers on the cell surface, differentiation into effector cells and induction of cytotoxicity or cytokine secretion may also be utilised. As an illustration, in the present examples the inventors determined levels of interleukin-2 (IL-2) produced by T-cells expressing a receptor component and signalling component of the CAR signalling system according to the present invention upon binding of antigen to the receptor component in the presence of varying concentrations of CID.
CHEMICAL INDUCER OF DIMERIZATION & BINDING DOMAINS
The CID and CID binding domains of the CAR signalling system of the first aspect of the invention may be any combination of molecules/peptides/domains which enables the selective co-localisation and dimerization of the receptor component and signalling component in the presence of the CID. As such, the CID agent is a molecule which is able to simultaneously bind to the receptor component and the signalling component. The CID agent therefore comprises at least two binding moieties.
The CID may be any pharmaceutically acceptable molecule which can simultaneously be bound by at least two binding domains.
The CID is capable being delivered to the cytoplasm of a target cell and being available for intracellular binding.
Small molecule dimerization systems for facilitating the co-localization of peptides are known in the art (Corson et a/.; 2008; ACS Chemical Biology; 3(1 1); 667).
The binding moieties of the CID may interact with identical binding domains present on the receptor component and the signalling component. That is, the CID may comprise two identical binding moieties such that it can simultaneously interact with a binding domain on the receptor component and an identical binding domain on the signalling component.
The CID and CID binding domains may be the FK506 binding protein (FKBP) ligand dimerization system described by Clackson et at. (PNAS; 1998; 95; 10437-10442). This dimerization system comprises two FKBP-like binding domains with a F36V mutation in the FKBP binding domain and a dimerization agent (AP1903) with complementary amino acid substitutions. Exposing cells engineered to express FKBP-like binding domain fusion proteins to AP103 results in the dimerization of the proteins comprising the FKBP-like binding domains but no interactions involving endogenous FKBP.
The dimerization system described by Farrar et a/., which utilises bacterial DNA gyrase B (GyrB) binding domains and the antibiotic coumermycin as CID may also be used in the signalling system of the present invention (Methods Enzymol; 2000; 327; 421-419 and Nature; 1996; 383; 178-181 ).
The binding moieties of the CID may interact with different binding domains on the receptor component and the signalling component. That is, the CID may comprise two different binding moieties which can simultaneously interact with a binding domain on the receptor component and a different binding domain on the signalling component.
The CID and CID binding domains may comprise the dimerization system described by Belshaw et al. (Nature; 1996; 93; 4604-4607), which utilises a FK506 (Tacrolimus)/cyclosporin fusion molecule as the CID agent with FK-binding protein 12 (FKBP12) and cylcophilin A as the binding domains.
The CID / CID binding domain may also be the rapamycin and FKBP12/FKBP12- Rapamycin Binding (FRB) domain of mTOR system described by Rivera et al. (Nature Med; 1996; 2; 1028-1032) or the non-immunosupressive rapamycin analogs (rapalogs) and FKBP12/FRB system described by Bayle et al. (Chem Bio; 2006; 13; 99-107). For example the CID may be C-20-methyllyrlrapamycin (MaRap) or C16(S)- Butylsulfonamidorapamycin (C16-BS-Rap), as described by Bayle et al. and illustrated in Figure 2 in combination with the corresponding binding domains. The CID may be C16-(S)-3-methylindolerapamycin (C16-iRap) or C16-(S)-7- methylindolerapamycin (AP21976/C16-AiRap) as described by Bayle et al., in combination with the respective complementary binding domains for each.
Other dimerization systems suitable for use in the signalling system of the present invention include an estrone/biotin CID in combination with an oestrogen-binding domain (EBD) and a streptavidin binding domain (Muddana & Peterson; Org. Lett; 2004; 6; 1409-1412; Hussey ef a/.; J. Am. Chem. Soc; 125; 3692-3693); a dexamethasone/methotrexate CID in combination with a glucocorticoid-binding domain (GBD) and a dihydrofolate reductase (DHFR) binding domain (Lin ef a/.; J. Am. Chem. Soc; 2000; 122; 4247-4248); a similar system in which the methotrexate portion of the CID is replaced with the bacterial specific DHFR inhibitor trimethoprim (Gallagher et al.; Anal. Biochem.; 2007; 363; 160-162) and an Oe-benzylguanine derivative/methotrexate CID in combination with an 06-alkylguanine-DNA alkyltransferase (AGT) binding domain and a DHFR binding domain (Gendreizig et a/.; J. Am. Chem. Soc; 125; 14970-14971).
RSL1 is a synthetic non-steroidal analogue of 20-hydroxyecdysone (see Figure 9). It is a member of a class of insecticides known as diacylhydrazines and can function to act as a non-steroidal ecdysone agonist. These molecules induce premature moulting and larvae death but are well tolerated in vertebrates. RSL1 has been used in artificial transcription switches in which a two-protein transcription switch is used consisting of a fusion between the ecdysone receptor (EcR) and GAL4, and the retinoid X receptor (RXR) and VP16. In such systems, EcR is modified to interact specifically with RSL13, and RXR is chimeric comprising of helices 1-8 replaced with helices 1-8 of human RXRp, and helices 9-12 from Locusta migratoria RXR. The signalling system of the present invention may use EcR and RXR domains for heterdimerisation in the presence of RSL1 or a derivative thereof.
For example the CID binding domains may be or comprise the sequences shown as SEQ ID NO: 1 to SEQ ID NO: 5, SEQ ID No. 25 or 26.
SEQ ID No 1 - FKBP12 domain
MGVQVETISPGDGRTFPKRGQTCWHYTGMLEDGKKFDSSRDRN PFKFMLGKQE VIRGWEEGVAQMSVGQRAKLTISPDYAYGATGHPGIIPPHATLVFDVELLKLE
SEQ ID No 2 - wild-type FRB segment of mTOR
MASRILWHEMWHEGLEEASRLYFGERNVKGMFEVLEPLHAMMERGPQTLKETSFN QAYGRDLMEAQEWCRKYMKSGNVKDLTQAWDLYYHVFRRISKLES
SEQ ID No 3 - FRB with T to L substitution at 2098 which allows binding to AP21967
MASRILWHEMWHEGLEEASRLYFGERNVKGMFEVLEPLHAMMERGPQTLKETSFN QAYGRDLMEAQEWCRKYMKSGNVKDLLQAWDLYYHVFRRISKL.ES
SEQ ID No 4 - FRB segment of mTOR with T to H substitution at 2098 and to W at F at residue 2101 of the full mTOR which binds Rapamycin with reduced affinity to wild type
MASRILWHEMWHEGLEEASRLYFGERNVKGMFEVLEPLHAMMERGPQTLKETSFN QAYGRDLMEAQEWCRKYMKSGNVKDLHQAFDLYYHVFRRISKLES
SEQ ID No 5 - FRB segment of mTOR with K to P substitution at residue 2095 of the full mTOR which binds Rapamycin with reduced affinity
MASRILWHEMWHEGLEEASRLYFGERNVKGMFEVLEPLHAMMERGPQTLKETSFN QAYGRDLMEAQEWCRKYMKSGNVPDLTQAWDLYYHVFRRISKLES
SEQ ID No. 25 - EcR
MPVDRILEAELAVEQKSDQGVEGPGGTGGSGSSPNDPVTNICQAADKQLFTLVEW AKRIPHFSSLPLDDQVILLRAGWNELLIASFSHRSIDVRDGILLATGLHVHRNSAHSAG VGAIFDRVLTELVSKMRDMRMDKTELGCLRAIILFNPEVRGLKSAQEVELLREKVYAA
LEEYTRTTHPDEPGRFAKLLLRLPSLRSIGLKCLEHLFFFRLIGDVPIDTFLMEMLESP
SDS
SEQ ID No. 26 - RXR
RPECWPETQCAMKRKEKKAQKEKDKLPVSTTTVDDHMPPIMQCEPPPPEAARIHE
WPRFLSDKLLETNRQKNIPQLTANQQFLIARLIWYQDGYEQPSDEDLKRITQTWQQ
ADDENEESDTPFRQITE TILTVQLIVEFAKGLPGFAKISQPDQITLLKACSSEVMMLR
VARRYDAASDSILFANNQAYTRDNYRKAGMAEVIEDLLHFCRCMYSMALDNIHYALL
TAWIFSDRPGLEQPQLVEEIQRYYLNTLRIYILNQLSGSARSSVIYGKILSILSELRTLG
MQNSNMCISLKLKNRKLPPFLEEIWDVADMSHTQPPPILESPTNL
Variant sequences may have at least 80%, 85%, 90%, 95%, 98% or 99% sequence identity to SEQ ID No. 1 to 5, 25 or 26 provided that the sequences provide an effective dimerization system. That is, provided that the sequences facilitate sufficient co-localisation of the antigen-binding and intracellular signalling components, in the presence of the CID, for successful signaling to occur upon binding of the antigen- binding domain to antigen.
The signalling system according to the present invention is not limited by the arrangement of a specific dimerization system. The receptor componentmay comprise either binding domain of a given dimerization system so long as the signalling component comprises the corresponding, complementary binding domain which enables the receptor component and signalling component to co-localize in the presence of the CID.
The present invention also relates to a method for activating the CAR signalling system of the first aspect which comprises the step of administering the CID. As described above, administration of the CID results in co-localization of the receptor component and signalling component, such that signalling through the signalling domain occurs upon binding of the antigen-binding domain to antigen.
The CID and CID binding domains may facilitate signalling through the CAR signalling system which is proportional to the concentration of CID present.
The present invention further provides a method for reducing the activity of the CAR system of the first aspect which comprises reducing or stopping the administration of the CID. Reducing the level of the CID results in fewer receptor : signalling component dimers and thus the total level of signalling through the CAR signaling system is decreased in the presence of the antigen. Stopping the administration of the CID results in the absence of receptor : signalling component dimers and termination of signalling despite the presence of antigen binding to the antigen- binding domain.
Another possibility of this system is to "tune" the signal strength the receptor transmits. If the affinity between one or both of the CID binding domains is lessened, less CAR systems will be active and hence signal propagation upon antigen binding is lessened. This may avoid toxicity.
RECEPTOR COMPONENT
The present invention provides a receptor component comprising an antigen-binding domain, an optional spacer domain, a transmembrane domain and a CID biding domain (CBD1). When expressed in a cell, the receptor component localises to the cell membrane. Here, the antigen-binding domain of the molecule is orientated on the extracellular side of the membrane and the CBD is localised to the intracellular side of the membrane.
The receptor component therefore provides the antigen-binding function of the CAR signalling system of the present invention.
ANTIGEN BINDING DOMAIN
The antigen-binding domain is the portion of a classical CAR which recognizes antigen. In the signalling system of the present invention the antigen-binding is located within the receptor component.
Numerous antigen-binding domains are known in the art, including those based on the antigen binding site of an antibody, antibody mimetics, and T-cell receptors. For example, the antigen-binding domain may comprise: a single-chain variable fragment (scFv) derived from a monoclonal antibody; a natural ligand of the target antigen; a peptide with sufficient affinity for the target; a single domain binder such as a camelid; an artificial binder single as a Darpin; or a single-chain derived from a T-cell receptor. W
21
Various tumour associated antigens (TAA) are known, as shown in the following Table 1. The antigen-binding domain used in the present invention may be a domain which is capable of binding a TAA as indicated therein.
Table 1
Figure imgf000022_0001
TRANSMEMBRANE DOMAIN
The transmembrane domain is the sequence of a classical CAR that spans the membrane. In the signalling system of the present invention the transmembrane domain is located in the receptor component. It may comprise a hydrophobic alpha helix. The transmembrane domain may be derived from CD28, which gives good receptor stability. The transmembrane domain may be from CD 148, as shown in SEQ ID No. 23.
SIGNAL PEPTIDE
The receptor component of the CAR signalling system of the present invention may comprise a signal peptide so that when the receptor component is expressed in a cell, such as a T-cell, the nascent protein is directed to the endoplasmic reticulum and subsequently to the cell surface, where it is displayed.
The core of the signal peptide may contain a long stretch of hydrophobic amino acids that has a tendency to form a single alpha-helix. The signal peptide may begin with a short positively charged stretch of amino acids, which helps to enforce proper topology of the polypeptide during translocation. At the end of the signal peptide W
22 there is typically a stretch of amino acids that is recognized and cleaved by signal peptidase. Signal peptidase may cleave either during or after completion of translocation to generate a free signal peptide and a mature protein. The free signal peptides are then digested by specific proteases.
The signal peptide may be at the amino terminus of the molecule.
The signal peptide may comprise the SEQ ID No. 6, 7 or 8 or a variant thereof having 5, 4, 3, 2 or 1 amino acid mutations (insertions, substitutions or additions) provided that the signal peptide still functions to cause cell surface expression of the CAR.
SEQ ID No. 6: MGTSLLCWMALCLLGADHADG
The signal peptide of SEQ ID No. 6 is compact and highly efficient. It is predicted to give about 95% cleavage after the terminal glycine, giving efficient removal by signal peptidase.
SEQ ID No. 7: MSLPVTALLLPLALLLHAARP
The signal peptide of SEQ ID No. 7 is derived from lgG1.
SEQ ID No. 8: MAVPTQVLGLLLLWLTDARC
The signal peptide of SEQ ID No. 8 is derived from CD8.
SPACER DOMAIN
The CAR signalling system described herein may comprise a spacer sequence to connect the antigen-binding domain with the transmembrane domain in the receptor component. A flexible spacer allows the antigen-binding domain to orient in different directions to facilitate binding.
The spacer sequence may, for example, comprise an lgG1 Fc region, an lgG1 hinge or a human CD8 stalk or the mouse CD8 stalk. The spacer may alternatively comprise an alternative linker sequence which has similar length and/or domain spacing properties as an lgG1 Fc region, an lgG1 hinge or a CD8 stalk. A human lgG1 spacer may be altered to remove Fc binding motifs. Examples of amino acid sequences for these spacers are given below:
SEQ ID No. 9 (hinge-CH2CH3 of human lgG1)
AEPKSPDKTHTCPPCPAPPVAGPSVFLFPPKPKDTLMIARTPEVTCVWDVSHEDPE
VKFNWYVDGVEVHNAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSN A
LPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESN
GQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK
SLSLSPGKKD
SEQ ID No. 10 (human CD8 stalk):
TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDI
SEQ ID No. 11 (human lgG1 hinge):
AEPKSPDKTHTCPPCPKDPK
SEQ ID No. 12 (CD2 ectodomain)
KEITNALETWGALGQDINLDIPSFQMSDDIDDIKWEKTSDKKKIAQFRKEKETFKEKD TYKLFKNGTLKIKHLKTDDQDIYKVSIYDTKGKNVLEKIFDLKIQERVSKPKISWTCINT TLTCEVMNGTDPELNLYQDGKHLKLSQRVITHKWTTSLSAKFKCTAGNKVSKESSV EPVSCPEKGLD
SEQ ID no. 13 (CD34 ectodomain)
SLDNNGTATPELPTQGTFSNVSTNVSYQETTTPSTLGSTSLHPVSQHGNEATTNITE
TTVKFTSTSVITSVYGNTNSSVQSQTSVISTVFTTPANVSTPETTLKPSLSPGNVSDL
STTSTSLATSPTKPYTSSSPILSDIKAEIKCSGIREVKLTQGICLEQNKTSSCAEFKKD
RGEGLARVLCGEEQADADAGAQVCSLLLAQSEVRPQCLLLVLANRTEISSKLQLMK
KHQSDLKKLGILDFTEQDVASHQSYSQKT
RECEPTOR COMPONENT COMPRISING MULTIPLE TRANSMEMBRANE DOMAINS
The receptor component may comprise an appropriate number of transmembrane domains such that each CID binding domain is orientated on the intracellular side of the cell membrane (Figure 6). For example the receptor component may comprise 3, 5, 7, 9, 1 1 , or more transmembrane domains. In this way, a single receptor component may recruit multiple signalling components amplifying signalling in response to antigen. The signalling system encoded by SEQ ID No. 22 is a multi- spanning receptor with 3 FRB domains. MULTIPLE RECEPTOR COMPONENTS
In another embodiment of the invention, the CAR signalling system may comprise of two or more receptor components each recognizing different antigens but comprising of the same intracellular CID binding domain. Such a CAR system would be capable of recognizing multiple antigens (Figure 5a). This might be useful for instance in avoiding tumour escape. In a further related aspect of the invention, the CBD1 of the receptor components differ in residues which dictate their affinity to the CID. In this way, a CAR system can be tuned such that signalling in response to one antigen is greater or lesser than response to another (figure 5b). This might be useful for instance when targeting two tumour antigens simultaneously but one is expressed at a higher density than the other. Response to this antigen could be tuned down to avoid toxicity caused by over-stimulation.
Methods suitable for altering the amino acid residues of the CID binding domain such that its binding affinity for CID is altered are known in the art and include substitution, addition and removal of amino acids using both targeted and random mutagenesis. For example, in the case of CID binding to FRB, a set of mutations have been published with a range of affinity (Bayle et al, Chemistry & Biology 13, 99-107, January 2006). Methods for determining the affinity of a CID for the CID binding domain are also well known in the art and include bioinformatics prediction of protein- protein interactions, affinity electrophoresis, surface plasma resonance, bio-layer interferometry, dual polarisation interferometry, static light scattering and dynamic light scattering.
The signalling system encoded by SEQ ID No. 20 comprises two receptor components and one signalling component.
SIGNALLING COMPONENT
The present invention also provides a signalling component comprising a signalling domain and a CID binding domain (CBD2). The signalling component is a soluble molecule and thus localises to the cytoplasm when it is expressed in a cell, for example a T cell.
No signalling occurs through the signalling domain of the signalling component unless it is co-localised with the receptor component provided by the present invention. Such co-localisation occurs only in the presence of the CID, as described above. INTRACELLULAR SIGNALLING DOMAIN
The intracellular signalling domain is the signal-transmission portion of a classical CAR. In the signalling system of the present invention the intracellular signalling domain (signalling domain) is located in the signalling component. In the presence of the CID, the membrane-bound, receptor component and the intracellular signalling component are brought into proximity. After antigen recognition, receptors cluster, native CD45 and CD148 are excluded from the synapse and a signal is transmitted to the cell.
As such the signalling domain of the signalling component is analogous to the endodomain of a classical CAR molecule.
The most commonly used signalling domain component is that of CD3-zeta endodomain, which contains 3 ITAMs. This transmits an activation signal to the T cell after antigen is bound. CD3-zeta may not provide a fully competent activation signal and additional co-stimulatory signalling may be needed. For example, chimeric CD28 and OX40 can be used with CD3-Zeta to transmit a proliferative / survival signal, or all three can be used together (illustrated in Figure 1 B).
The signalling component described herein comprises a signalling domain, it may comprise the CD3-Zeta endodomain alone, the CD3-Zeta endodomain with that of either CD28 or OX40 or the CD28 endodomain and OX40 and CD3-Zeta endodomain (Figure 3A).
The signalling component of a CAR signalling system according to the present invention may comprise the sequence shown as SEQ ID No. 14, 15 or 16 or a variant thereof.
SEQ ID No. 14 - CD3 Z endodomain
RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNP QEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQAL PPR
SEQ ID No. 15 - CD28 and CD3 Zeta endodomains SKRSRLLHSDYMNMTPRRPGPTRKHYQPYAPPRDFAAYRSRVKFSRSADAPAYQQ GQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAE AYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR
SEQ ID No. 16 - CD28, OX40 and CD3 Zeta endodomains.
SKRSRLLHSDYMNMTPRRPGPTRKHYQPYAPPRDFAAYRSRDQRLPPDAHKPPG GGSFRTPIQEEQADAHSTLAKIRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDV LDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDG LYQGLSTATKDTYDALHMQALPPR
A variant sequence may have at least 80%, 85%, 90%, 95%, 98% or 99% sequence identity to SEQ ID No. 14, 15 or 16, provided that the sequence provides an effective intracellular signalling domain.
MULTIPLE SIGNALLING COMPONENTS
The signalling system according to the first aspect of the present invention may comprise a plurality of signalling components, each comprising a signalling domain and a CIDB2, wherein each signalling domain is bound by the same CID but the signalling domains comprise different endodomains (Figure 7a). In this way, multiple different endodomains can be activated simultaneously. This is advantageous over a compound signalling domain since each signalling domain remains unencumbered from other signalling domains.
If each signalling component comprises of a CBD2 domains which differ in residues which alter their affinity to CID, the CID enables signalling components comprising different signalling domains to ligate to the CID with differing kinetics (Figure 7b). This allows greater control over the signalling in response to antigen-binding by the receptor component as different signalling components are recruited to the receptor component in varying kinetics/dynamics. This is advantageous since rather than a fixed equal ratio of signal transmitted by a compound endodomain, an optimal T-cell activation signal may require different proportions of different immunological signals.
The signalling system encoded by SEQ ID No. 21 has one receptor components and three signalling components.
SIGNALLING COMPONENTS WITH AN ADDITIONAL CID BINDING DOMAIN In the case of very low density of target antigen expression, signalling strength may have to be amplified. Take for instance, the native T-cell receptor which is associated with multiple ITAM containing CD3 components. In such circumstances, signalling components can be modified to contain CBD2, a signalling domain and also a CBD1 (Figure 8). In this way, addition of CID would lead to concatenation of multiple signalling components. The average size of the concatenation can be tuned by varying the amount of signalling components which have a CBD2. In the event of antigen recognition and in the presence of CID, signalling through the concatenation of multiple signalling components would be more potent than through individual signalling components.
NUCLEIC ACID
The third aspect of the invention relates to a nucleic acid encoding the receptor component of the second aspect and a nucleic acid encoding a signalling component of the third aspect.
The specific encoding nucleic acid sequences for four inducible CARs are given later in description, following the Examples section.
As used herein, the terms "polynucleotide", "nucleotide", and "nucleic acid" are intended to be synonymous with each other.
It will be understood by a skilled person that numerous different polynucleotides and nucleic acids can encode the same polypeptide as a result of the degeneracy of the genetic code. In addition, it is to be understood that skilled persons may, using routine techniques, make nucleotide substitutions that do not affect the polypeptide sequence encoded by the polynucleotides described here to reflect the codon usage of any particular host organism in which the polypeptides are to be expressed.
Nucleic acids according to the second aspect of the invention may comprise DNA or RNA. They may be single-stranded or double-stranded. They may also be polynucleotides which include within them synthetic or modified nucleotides. A number of different types of modification to oligonucleotides are known in the art. These include methylphosphonate and phosphorothioate backbones, addition of acridine or polylysine chains at the 3' and/or 5' ends of the molecule. For the purposes of the use as described herein, it is to be understood that the polynucleotides may be modified by any method available in the art. Such modifications may be carried out in order to enhance the in vivo activity or life span of polynucleotides of interest.
The terms "variant", "homologue" or "derivative" in relation to a nucleotide sequence include any substitution of, variation of, modification of, replacement of, deletion of or addition of one (or more) nucleic acid from or to the sequence.
The nucleic acid according to the third aspect of the invention may be a nucleic acid which encodes both the receptor component and the signalling component.
The nucleic acid may produce a polypeptide which comprises the receptor component and the signalling component joined by a cleavage site. The cleavage site may be self-cleaving, such that when the polypeptide is produced, it is immediately cleaved into the receptor component and the signalling component without the need for any external cleavage activity.
Various self-cleaving sites are known, including the Foot-and- outh disease virus (FMDV) 2a self-cleaving peptide, which has the sequence shown as SEQ ID No. 17 or SEQ ID No. 18:
SEQ ID No. 17
RAEGRGSLLTCGDVEENPGP. or
SEQ ID No. 18
QCTNYALLKLAGDVESNPGP
Where the signalling system comprises a plurality of signalling components, each comprising a signalling domain and a CIDB2, the nucleic acid may have the general formula:
RC-coexpr1 -SC 1 -coexpr2-SC2 wherein:
RC is a nucleic acid sequence encoding the receptor component; Coexprl and coexpr2, which may be the same or different, are nucleic acid sequences allowing co-expression of the two flanking polypeptides (e.g. a sequence which encodes a cleavage site)
SC1 and SC2 are nucleic acid sequence encoding signalling components.
The signalling components encoded by SC1 and SC2 may bind the same CID, but have different endodomains. The nucleic acid construct may comprise nucleic acid sequences encoding more than two signalling components e.g. SC3, SC4 ... up to SCx, each separated by a coexpression sequence, coexpr3, coexpr4....up to coexprX.
The nucleic acid sequences encoding each component may be in any order, for example: RC-coexpr1-SC1-coexpr2-SC2; SC1 -coexprl -SC2-coexpr2-RC; or SC1 - coexprl- RC-coexpr2-SC2.
Where the signalling system comprises a plurality of receptor components, the nucleic acid may have the general formula:
RC 1 -coexprl -RC2-coexpr2-SC wherein:
RC1 and RC2 are nucleic acid sequences encoding receptor components;
Coexprl and coexpr2, which may be the same or different, are nucleic acid sequences allowing co-expression of the two flanking polypeptides (e.g. a sequence which encodes a cleavage site)
SC is a nucleic acid sequence encoding the signalling component.
The receptor components may bind to different antigens but the same CID. They may have the same CIDB1. The nucleic acid construct may comprise nucleic acid sequences encoding more than two receptor components e.g. RC3, RC4 ... up to RCx, each separated by a coexpression sequence, coexpr3, coexpr4....up to coexprX.
The nucleic acid sequences encoding each component may be in any order, for example: RC1 -coexprl -RC2-coexpr2-SC; RC1 -coexprl -SC-coexpr2-RC2; or SC- coexprl -RC1 -coexpr2-RC2. The co-expressing sequence may be an internal ribosome entry sequence (IRES). The co-expressing sequence may be an internal promoter.
The present invention also provides a kit comprising a nucleic acid encoding the receptor component of the second aspect and/or a nucleic acid encoding a signalling component of the third aspect. The kit may also comprise a heterodimerization agent which is suitable for use in the signalling system of the first aspect.
VECTOR
The present invention also provides a vector, or kit of vectors which comprises one or more nucleic acid sequence(s) encoding a receptor component of the second aspect and/or signalling component of the third aspect of the invention. Such a vector may be used to introduce the nucleic acid sequence(s) into a host cell so that it expresses the receptor component and signalling component of the CAR signalling system according to the first aspect of the invention. The kit may also comprise a CID which is suitable for use in the signalling system of the first aspect.
The vector may, for example, be a plasmid or a viral vector, such as a retroviral vector or a lentiviral vector, or a transposon based vector or synthetic mRNA.
The vector may be capable of transfecting or transducing a T cell.
CYTOLYTIC IMMUNE CELL
The present invention also relates to an immune cell comprising the CAR signalling system according to the first aspect of the invention.
Cytolytic immune cells can be T cells or T lymphocytes which are a type of lymphocyte that play a central role in cell-mediated immunity. They can be distinguished from other lymphocytes, such as B cells and natural killer cells (NK cells), by the presence of a T-cell receptor (TCR) on the cell surface. There are various types of T cell, as summarised below.
Helper T helper cells (TH cells) assist other white blood cells in immunologic processes, including maturation of B cells into plasma cells and memory B cells, and activation of cytotoxic T cells and macrophages. TH cells express CD4 on their surface. TH cells become activated when they are presented with peptide antigens by MHC class II molecules on the surface of antigen presenting cells (APCs). These cells can differentiate into one of several subtypes, including TH1 , TH2, TH3, TH17, Th9, or TFH, which secrete different cytokines to facilitate different types of immune responses.
Cytolytic T cells (TC cells, or CTLs) destroy virally infected cells and tumor cells, and are also implicated in transplant rejection. CTLs express the CD8 at their surface. These cells recognize their targets by binding to antigen associated with MHC class I, which is present on the surface of all nucleated cells. Through IL-10, adenosine and other molecules secreted by regulatory T cells, the CD8+ cells can be inactivated to an anergic state, which prevent autoimmune diseases such as experimental autoimmune encephalomyelitis.
Memory T cells are a subset of antigen-specific T cells that persist long-term after an infection has resolved. They quickly expand to large numbers of effector T cells upon re-exposure to their cognate antigen, thus providing the immune system with "memory" against past infections. Memory T cells comprise three subtypes: central memory T cells (TCM cells) and two types of effector memory T cells (TEM cells and TEMRA cells). Memory cells may be either CD4+ or CD8+. Memory T cells typically express the cell surface protein CD45RO.
Regulatory T cells (Treg cells), formerly known as suppressor T cells, are crucial for the maintenance of immunological tolerance. Their major role is to shut down T cell- mediated immunity toward the end of an immune reaction and to suppress autoreactive T cells that escaped the process of negative selection in the thymus.
Two major classes of CD4+ Treg cells have been described— naturally occurring Treg cells and adaptive Treg cells.
Naturally occurring Treg cells (also known as CD4+CD25+FoxP3+ Treg cells) arise in the thymus and have been linked to interactions between developing T cells with both myeloid (CD1 1 c+) and plasmacytoid (CD123+) dendritic cells that have been activated with TSLP. Naturally occurring Treg cells can be distinguished from other T cells by the presence of an intracellular molecule called FoxP3. Mutations of the FOXP3 gene can prevent regulatory T cell development, causing the fatal autoimmune disease IPEX. Adaptive Treg cells (also known as Tr1 cells or Th3 cells) may originate during a normal immune response.
Natural Killer Cells (or NK cells) are a type of cytolytic cell which form part of the innate immune system. NK cells provide rapid responses to innate signals from virally infected cells in an HC independent manner
NK cells (belonging to the group of innate lymphoid cells) are defined as large granular lymphocytes (LGL) and constitute the third kind of cells differentiated from the common lymphoid progenitor generating B and T lymphocytes. NK cells are known to differentiate and mature in the bone marrow, lymph node, spleen, tonsils and thymus where they then enter into the circulation.
The CAR cells of the invention may be any of the cell types mentioned above.
T or NK cells expressing the molecules of the CAR signalling system according to the first aspect of the invention may either be created ex vivo either from a patient's own peripheral blood (1st party), or in the setting of a haematopoietic stem cell transplant from donor peripheral blood (2nd party), or peripheral blood from an unconnected donor (3rd party).
Alternatively, T or NK cells expressing the molecules of the CAR signalling system according to the first aspect of the invention may be derived from ex vivo differentiation of inducible progenitor cells or embryonic progenitor cells to T cells. Alternatively, an immortalized T-cell line which retains its lytic function and could act as a therapeutic may be used.
In all these embodiments, CAR cells are generated by introducing DNA or RNA coding for the receptor component and signalling component by one of many means including transduction with a viral vector, transfection with DNA or RNA.
The CAR cell of the invention may be an ex vivo T or NK cell from a subject. The T or NK cell may be from a peripheral blood mononuclear cell (PBMC) sample. T or NK cells may be activated and/or expanded prior to being transduced with nucleic acid encoding the molecules providing the CAR signalling system according to the first aspect of the invention, for example by treatment with an anti-CD3 monoclonal antibody. The T or NK cell of the invention may be made by:
(i) isolation of a T or NK cell-containing sample from a subject or other sources listed above; and
(ii) transduction or transfection of the T or NK cells with one or more a nucleic acid sequence(s) encoding the receptor component and/or signalling component of the CAR signalling system according to the second and third aspects of the invention.
The T or NK cells may then by purified, for example, selected on the basis of expression of the antigen-binding domain of the antigen-binding polypeptide.
The present invention also provides a kit which comprises a T or NK cell comprising the CAR signalling system according to the first aspect of the invention and a heterodimerization agent suitable for use in the signalling system.
PHARMACEUTICAL COMPOSITION
The present invention also relates to a pharmaceutical composition containing a plurality of cytolytic immune cells expressing the molecules of the CAR signalling system of the first aspect of the invention. The pharmaceutical composition may additionally comprise a pharmaceutically acceptable carrier, diluent or excipient. The pharmaceutical composition may optionally comprise one or more further pharmaceutically active polypeptides and/or compounds. Such a formulation may, for example, be in a form suitable for intravenous infusion.
METHODS
The present invention provides a method for treating and/or preventing a disease which comprises the step of administering the cytolytic immune cells of the present invention (for example in a pharmaceutical composition as described above) and/or a CID suitable for use in a signalling system according to the first aspect of the invention to a subject.
A method for treating a disease relates to the therapeutic use of the cytolytic immune cells of the present invention. Herein the cells may be administered to a subject having an existing disease or condition in order to lessen, reduce or improve at least one symptom associated with the disease and/or to slow down, reduce or block the progression of the disease. The method for preventing a disease relates to the prophylactic use of the cytolytic immune cells of the present invention. Herein such cells may be administered to a subject who has not yet contracted the disease and/or who is not showing any symptoms of the disease to prevent or impair the cause of the disease or to reduce or prevent development of at least one symptom associated with the disease. The subject may have a predisposition for, or be thought to be at risk of developing, the disease.
The method may involve the steps of:
(i) isolating a T or NK cell-containing sample from a subject;
(ii) transducing or transfecting such cells with a nucleic acid sequence or vector provided by the present invention;
(iii) administering the cells from (ii) to the subject.
The methods for treating a disease may further comprise the step of administering a CID suitable for use in the signalling system of the first aspect of the invention to the subject.
The present invention also provides a method for treating and/or preventing a disease in a subject which subject comprises cells of the invention, which method comprises the step of administering a CID agent suitable for use in the CAR signalling system according to the first aspect to the subject. As such, this method involves administering a suitable CID agent to a subject which already comprises CAR cells of the present invention.
The CID may be administered in the form of a pharmaceutical composition. The pharmaceutical composition may additionally comprise a pharmaceutically acceptable carrier, diluent or excipient. The pharmaceutical composition may optionally comprise one or more further pharmaceutically active polypeptides and/or compounds. Such a formulation may, for example, be in a form suitable for intravenous infusion.
The methods for treating a disease provided by the present invention may involve monitoring the progression of the disease and monitoring any toxic activity and adjusting the dose of the CID administered to the subject to provide acceptable levels of disease progression and toxic activity. Monitoring the progression of the disease means to assess the symptoms associated with the disease over time to determine if they are reducing/improving or increasing/worsening.
Toxic activities relate to adverse effects caused by the CAR cells of the invention following their administration to a subject. Toxic activities may include, for example, immunological toxicity, biliary toxicity and respiratory distress syndrome.
The level of signalling through the signalling system of the first aspect of the invention, and therefore the level of activation CAR cells expressing the signalling system, can be adjusted by altering the amount of CID present. In the present method the level of CAR cell activation may be augmented by increasing the dose of CID administered to the subject. Conversely, the level of CAR cell activation may be reduced by decreasing the dose of CID administered to the subject.
Higher levels of CAR cell activation are likely to be associated with reduced disease progression but increased toxic activities, whilst lower levels of CAR cell activation are likely to be associated with increased disease progression but reduced toxic activities.
As such the dose of CID administered to a subject may be altered in order to provide an acceptable level of both disease progression and toxic activity. The specific level of disease progression and toxic activities determined to be 'acceptable' will vary according to the specific circumstances and should be assessed on such a basis. The present invention provides a method for altering the activation level of the CAR cells in order to achieve this appropriate level.
The present invention provides a CAR cell of the present invention for use in treating and/or preventing a disease.
The invention also relates to the use of a CAR cell of the present invention in the manufacture of a medicament for the treatment and/or prevention of a disease.
The present invention also provides a CID agent suitable for activating a CAR signalling system according to the first aspect of the invention for use in treating and/or preventing a disease. The present invention also provides a CID agent for use in activating a CAR signalling system according to the first aspect of the invention in a CAR cell.
The invention also provides the use of a CID suitable for activating a CAR signalling system according to the first aspect of the invention in the manufacture of a medicament for the treatment and/or prevention of a disease.
The invention also provides the use of a CID in the manufacture of a medicament for in activating a CAR signalling system according to the first aspect of the invention in a CAR cell.
The disease to be treated and/or prevented by the methods of the present invention may be an infection, such as a viral infection.
The methods of the invention may also be for the control of pathogenic immune responses, for example in autoimmune diseases, allergies and graft-vs-host rejection.
The methods may be for the treatment of a cancerous disease, such as bladder cancer, breast cancer, colon cancer, endometrial cancer, kidney cancer (renal cell), leukemia, lung cancer, melanoma, non-Hodgkin lymphoma, pancreatic cancer, prostate cancer and thyroid cancer.
The CAR cells of the present invention may be capable of killing target cells, such as cancer cells. The target cell may be recognisable by expression of a TAA, for example the expression of a TAA provided above in Table 1.
The CAR cells and pharmaceutical compositions of present invention may be for use in the treatment and/or prevention of the diseases described above.
The CAR cells and pharmaceutical compositions of present invention may be for use in any of the methods described above.
The present invention also provides a method of administering a cell according to the ninth aspect of the invention to a subject in need of same. The present invention also provides a method of administering a CID as defined herein to a subject who comprises a cell according to the ninth aspect of the invention.
The invention will now be further described by way of Examples, which are meant to serve to assist one of ordinary skill in the art in carrying out the invention and are not intended in any way to limit the scope of the invention.
EXAMPLES
Example 1 - A rapamvcin-inducible CAR system
T-cells were transduced with a retroviral vector coding for an inducible CAR detailed in figure 3 and the sequence of which is shown as SEQ ID 19. This inducible CAR has a single receptor component and a single signalling component. The receptor component comprises of an anti-CD19 scFv, a spacer domain derived from human lgG1 , a transmembrane-domain derived from CD28 and FKBP12 as endodomain. The signalling component comprises of the FRB domain from mTOR and a signalling domain derived from endodomains of CD28, OX40 and CD3-Zeta. Expression of the CAR was determined by staining the transduced cells with a conjugated antibody recognizing the spacer domain and analysing by flow cytometry. The T-cells were incubated with either target cells expressing the cognate target CD19 in ratio of 2:1 , 1 : 1 and 0:1 (targeteffector) in the absence of rapamycin or at increasing concentrations. Signalling only occurred in the presence of both the cognate antigen and rapamycin. A dose response to rapamycin and target antigen was observed.
Example 2 - An RSL1 -inducible CAR system
RSL1 is a synthetic non-steroidal analogue of 20-hydroxyecdysone (Figure 9). It is a member of a class of insecticides known as diacylhydrazines and can function to act as a non-steroidal ecdysone agonist. These molecules induce premature moulting and larvae death but are well tolerated in vertebrates (Dhadialla et al (1998) Annu. Rev. Entomol. 43, 545-569). RSL1 has been used in artificial transcription switches (Lessard et al (2007) The Prostate 67, 808-819) whereby a two-protein transcription switch consisting of a fusion between the ecdysone receptor (EcR) and GAL4, and the retinoid X receptor (RXR) and VP16. In such systems, EcR is modified to interact specifically with RSL1 (Kumar et al (2004) J. Biol. Chem. 279, 2721 1-27218), and RXR is chimeric comprising of helices 1-8 replaced with helices 1-8 of human RXR , and helices 9-12 from Locusta migratoria RXR.
The present inventors have constructed a split CD19 CAR based on the CD19 system described in Example 1 with the heterodimerization motif on the antigen recognition component and signalling components being EcR and RXR respectively (Figure 10a and SEQ ID No. 23). This CAR was transduced into T-cells and found to express well (Figure 10b). These T-cells were challenged with targets which were CD19 negative and CD19 positive. Only CAR T-cells challenged with SupT1 targets in the presence of RSL1 released IL-2 (Figure 10c).
Example 3 - An AP1903-inducible CAR system
AP1903 is a synthetic chemical inducer of dimerization which binds to two mutated FKB12 domains (Clackson et al (1998). Proc. Natl. Acad. Sci. U. S. A. 95, 10437- 10442). An inducible CAR was developed based on this system, using the modified FKBP12 domains to constitute the heterodimerization domain of both the antigen recognition component and signalling component in the framework as described in Example 1 with the antigen recognition domain and the signalling domain co- expressed with a FMD-2A like sequence (Figure 1 1 a and SEQ ID No. 24). The FKBP12 domains were codon-wobbled to prevent homologous recombination. T-cells were transduced with this construct and challenged with target cells which were CD19 negative or positive.
T-cells responded only to CD19 positive targets in the presence of AP1903 (Figure 1 1 b). At very high concentration of dimerizer, signalling was inhibited likely due to saturating conditions where the stoichiometry favours one FKBP12 binding a single dimerizer molecule rather than one dimerizer molecule binding two FKBP12 domains (Figure 11 c).
SPECIFIC INDUCIBLE CAR-ENCODING SEQUENCES
SEQ ID No. 19. Simple inducible CAR comprised of one receptor component and one signalling component. The receptor component comprises of a signal peptide, an anti- CD19 scFv, a spacer from the Fc domain of human lgG1 , a CD28 transmembrane domain and FKB12 as endodomain. A FMD-2A peptide separates this receptor component from the signalling component. The signalling component comprises of FRB and a compound of endodomains from CD28, OX40 and CD3-Zeta. SgrAI
1 atggagaccg acaccctgct gctgtgggtg ctgctgctgt gggtgccagg cagcaccggc gacatccaga tgacccagac » signal peptide >>
» aCD19 heavy chain > caccagcagc ctgagcgcca gcctgggcga ccgggtgacc atcagctgca gagccagcca ggacatcagc aagtacctg > aCD19 heavy chain
161 actggtacca gcagaagccc gacggcaccg tgaagctgct gatctaccac accagccggc tgcacagcgg cgtgcccagc >.. aCD19 heavy chain >
241 cggttcagcg gcagcggcag cggcaccgac tacagcctga ccatcagcaa cctggagcag gaggacatcg ccacctactt > aCD1 heavy chain >
321 ctgccagcag ggcaacaccc tgccctacac cttcggaggc ggcaccaagc tggagatcac caaggccgga ggcggaggct > aCD19 heavy chain »
>> .. linker ... >
401 ctggcggagg cggctctggc ggaggcggct ctggcggagg cggcagcgag gtgaagctgc aggagtctgg cccaggcctg > linker >>
» aCD19 light chain >
481 gtggccccaa gccagagcct gagcgtgacc tgcaccgtga gcggcgtgag cctgcccgac tacggcgtga gctggatcag > aCD19 light chain >
561 gcagccccca cggaagggcc tggagtggct gggcgtgatc tggggcagcg agaccaccta ctacaacagc gccctgaaga > aCD19 light chain >
641 gccggctgac catcatcaag gacaacagca agagccaggt gttcctgaag atgaacagcc tgcagaccga cgacaccgcc > aCD19 light chain >
721 atctactact gcgccaagca ctactactat ggcggcagct acgctatgga ctactggggc cagggcacca gcgtgaccgt > aCD19 light chain >
BamHI Fsel
801 gagctcggat cccgccgagc ccaaatctcc tgacaaaact cacacatgcc caccgtgccc agcacctccc gtggccggcc > . >>
» HCH2CH3pvaa >
881 cgtcagtctt cctcttcccc ccaaaaccca aggacaccct catgatcgcc cggacccctg aggtcacatg cgtggtggtg > HCH2CH3pvaa >
961 gacgtgagcc acgaagaccc tgaggtcaag ttcaactggt acgtggacgg cgtggaggtg cataatgcca agacaaagcc > HCH2CH3pvaa >
SacIT
1041 gcgggaggag cagtacaaca gcacgtaccg tgtggtcagc gtcctcaccg tcctgcacca ggactggctg aatggcaagg > HCH2CH3pvaa >
1121 agtacaagtg caaggtctcc aacaaagccc tcccagcccc catcgagaaa accatctcca aagccaaagg gcagccccga > HCH2CH3pvaa >
1201 gaaccacagg tgtacaccct gcccccatcc cgggatgagc tgaccaagaa ccaggtcagc ctgacctgcc tggtcaaagg > HCH2CH3pvaa >
1281 cttctatccc agcgacatcg ccgtggagtg ggagagcaat gggcaaccgg agaacaacta caagaccacg cctcccgtgc > HCH2CH3pvaa >
1361 tggactccga cggctccttc ttcctctaca gcaagctcac cgtggacaag agcaggtggc agcaggggaa cgtcttctca > HCH2CH3pvaa >
PpulOI
Nsil
BfrBI 1441 tgctccgrga tgcatgaggc cctgcacaat cactataccc agaaatct t gagtctgagc ccaggcaaga aggaccccaa
HCH?nH3rwaa
1521 gttctgggtc ctggtggtgg tqggaggcgt gctggcctgt tact tctcc tggtgaccgt gg cttcatc atcttctggg
CD28tm ... >
1601 tgggagtgca ggtggaaa.cc a ctccccag gagacgggcg ca cttcccc aagcgcggcc agacctgcgt ggtgcactac
»
.... , FKBP12
1681 accgggatgc ttgaagatgg aaagaaattc gattcctccc gggacagaaa caagcccttt aagtttatgc taggcaagca
F BP12 >
1761 ggaggtgatc cgaggctggg aagaaggggt tgcccagatg agtgtgggtc agagagccaa actgactata tctccagatt
. FKBP12 >
1841 atgcctatgg tgccactggg cacccaggca tcatcccacc acatgccact ctcgtcttcg atgtggagct tctaaaactg
FKBP12 >
1921 gaacgcgcag agggccgggg ctcattgctg acctgtggag atgtcgagga aaatcccggc ccaatggctt ctagaatcct »>
» ...2A >>
>> .. >
2001 ctggcatgag atgtggcatg aaggcctgga agaggcatct cgtttgtact utggggaaag gaacgtgaaa ggcatgtttg
FRB
Sp l
2081 aggtgctgga gcccttgcat gctatgatgg aacggggccc ccagactctg aaggaaacat ccttt aatca ggcctatggt > FRB ,
2161 cgagatttaa tggaggccoa agagtggtgc aggaagta a tgaaatcagg gaatgtcaag gacctcctcc aagcctggga
FRB
X oT
2241 cctctattat catgtgttcc gacgaatctc aaagctcgag agtggcggag gaggcagttc aaggtccgcc gacgcgcctg
FRB ... »
m2fif)X7, , >
2321 cataccagca ggggcagaat cagctgtaca acgagctcaa cctcggtagg cgcgaggaat acgatgtgct cgataagaga
CD280XZ
Nrul
2401 agaggtcgcg atcccgagat gggaggaaaa cctcagcgcc gcaagaaccc tcaggagggg ctgtataacg aactgcagaa
CD280XZ , >
Sfil
2481 ggataagatg gcagaggcct actccgagat tggcatgaag ggtgagagga gaagaggtaa aggccatgac ggcctctaoc
CD280XZ >
2561 aaggcctctc taccgcaacg aaagacactt atgacgctct gcatatgcag gctctccccc ctaggcagtg cactaattac
, CD280X7,
2641 gcccttctga aacttgccgg cgatgtggag tctaaccctg gccctatcct gtggcacgag atgtggcatg agggtctgga
CD280X
2721 agaggccagc cggctgtact tcggagagag aaatgttaag ggtatgttcg aggtgctgga gccgcttcac gctatgatgg
....... CD280XZ
2801 agaggggccc ccagaccctc aaagaaacca gcttcaatca agcctacggg agggatctta tggaggcaca ggaatggtgt
2881 cggaagtaca tgaagagcgg gaacgtcaag gacctgctcc aggcttggga tttgtattat cacgtcttta ggcggatcag > CD280XZ
2961 caagtctgga ggcggaggaa gtctgcacag cgattacatg aacatgaccc cccgaaggcc cggacccaca cgcaaacact
CD280XZ
3041 atcaacccta tgctccccca cgcgacttcg ccgcctaccg gtcacgcgcc gaggggcgcg gctctttgtt gacttgcggg 3121 gaogttgaag agaatcctgg ccccatcctt tggcacgaga tgtggcacga gggcc ggag gaagcctccc ggctgtattt
CD280XZ >
3201 cggagagcgc aacgtcaaag gaatgtttga ggtgctggag cctctccatg caatgatgga gagggggcct cagactctta
CD280XZ
3281 aagaaacatc ctttaatcag gcttacggta gagatttgat ggaggctcaa gaatggtgcc ggaaatacat gaagagtgga
CD280XZ
3361 aacgttaaag acctgctgca ggcatgggac ctgtactatc acgtattcag acggatctca aagtcagggg gcggtggctc
CD280XZ
3441 cctttatatc ttcaagcagc ctttcatgag gccggtgcag accacacaag aagaggatgg gtgctcttgc cggttccccg
, CD?80XZ
3521 aggaggagga gggcggatgc gagctc ga
> , CD280X
SEQ ID No. 20. Inducible CAR comprised of two receptor component and one signalling component. The first receptor component comprises of a signal peptide, an anti-CD19 scFv, a spacer from the stalk region of CD8, a transmembrane domain from CD28, and FKB12 as an endodomain. The second signalling component comprises of an anti-CD29 scFv, spacer derived from the Fc domain of human lgG1 , a CD28 transmembrane domain and FKB12 as endodomain. The signalling component comprises of FRB and a compound of endodomains from CD28, OX40 and CD3-Zeta. A foot-and-mouth disease 2A peptide separates the components.
1 atgagcctgc ccgtgaccgc cctgctgctg cccctggccc tgctgctgca cgccgccaga ccagacatcc agatgaccca »
»
81 gaccaccagc agcctgagcg ccagcctggg cgaccgggtg accatcagct gcagagccag ccaggacatc agcaagtacc aCDl9
161 tgaactggta ccagcagaag cccgacggca ccgtgaagct gctgatctac cacaccagcc ggctgcacag cggcgtgccc
......aCDl9 scFv >
241 agccggttca gcggcagcgg cagcggcacc gactacagcc tgaocatcag caacctggag caggaggaca tcgccaccta
aCDl9 scFv , >
321 cttctgccag cagggcaaca ccctgcccta caccttcgga ggcggcacca agctggagat caccaaggcc ggaggcggag
aCDl9 scFv ,
401 gctctggcgg aggcggctct ggcggaggcg gctctggcgg aggcggcagc gaggtgaagc tgcaggagtc tggcccaggc
, aCDl9 >
481 ctggtggccc caagccagag cctgagcgtg acctgcaccg tgagcggcgt gagcctgccc gactacggcg tgagctggat
aCD19 , >
561 caggcagccc ccacggaagg gcctggagtg gctgggcgtg atctggggca gcgagaccac ctactacaac agcgccctga
641 agagccggct gaccatcatc aaggacaaca gcaagagcca ggtgttcctg aagatgaaca gcotgcagac cgacgacacc
aCD19
721 gccatctact actgcgccaa gcactactac tatggcggca gctacgctat ggactactgg ggccagggca ccagcgtgac
> aCDl9 scFv > SgrAI
801 cgtgagctca gatcccacca cgacgccagc gccgcgacca ccaacaccgg cgcccaccat cgcgtcgcag cccctgtccc >....»
» CD8STK >
881 tgcgcccaga ggcgtgccgg ccagcggcgg ggggcgcagt gcacacgagg gggctggact tcgcctgtga tatcttttgg > CD8STK »
»...>
S61 gtgctggtgg tggttggtgg agtcctggct tgctatagct tgctagtaac agtggccttc atcatcttct gggtgggagt > CD28tmZ »
»..>
1041 gcaggtggaa accatctccc caggagacgg gcgcaccttc cccaagcgcg gccagacctg cgtggtgcac tacaccggga > FKBP12 >
1121 tgcttgaaga tggaaagaaa ttcgattcct cccgggacag aaacaagccc tttaagttta tgctaggcaa gcaggaggtg > FKBP12 >
1201 atccgaggct gggaagaagg ggttgcccag atgagtgtgg gtcagagagc caaactgact atatctccag attatgccta > FKBP12 >
1281 tggtgccact gggcacccag gcatcatccc accacatgcc actctcgtct tcgatgtgga gcttctaaaa ctggaacgag > E'KBP12 »
>> . >
Ncol
1361 ccgagggcag gggaagtctt ctaacatgcg gggacgtgga ggaaaatccc gggcccatgg agaccgacac cctgctgctg > 2A »
>>... signal peptide 2....>
1441 tgggtgctgc tgctgtgggt gcccggcagc accggccagg tgcagctgca gcagcccggc gccgagctgg tgaagcccgg > signal peptide 2 »
» aCD20_scFv > lb21 cgccagcgtg aagatgagct gcaaggccag cggctacacc ttcaccagct acaacatgca ctgggtgaag cagacccccg > aCD20 scFv >
1601 gccggggcct ggagtggatc ggcgccatct accccggcaa cggcgacacc agctacaacc agaagttcaa gggcaaggcc > aCD20 scFv >
1681 accctgaccg ccgacaagag cagcagcacc gcctacatgc agctgagcag cctgaccagc gaggacagcg ccgtgtacta > aCD20_scFv >
1761 ctgcgcccgg agcacctact acggcggcga ctggtacttc aacgtgtggg gcgccggcac caccgtgacc gtgagcggag > aCD20_sc.Fv >
1841 gcggcggcag cggaggaggc ggctctgggg gaggcggatc tcagatcgtg ctgagccaga gccccgccat cctgagcgcc > aCD20 scFv >
1921 agccccggcg agaaggtgac catgacctgc cgggccagca gcagcgtgag ctacatccac tggttccagc agaagcccgg > aCD20 scFv >
2001 cagcagcccc aagccctgga tctacgccac cagcaacctg gccagcggcg tgcccgtgcg gttcagcggc agcggcagcg > aCDZO scFv >
2081 gcaccagcta cagcctgacc atcagccggg tggaggccga ggacgccgcc acctactact gccagcagtg gaccagcaac > aCD20 scFv "....>
BairiHI
2161 ccacccacct tcggcggcgg caccaagctg gagatcaagc ggtcggatcc cgccgagccc aaatctcctg acaaaactca > aCD20_scFV »
» HCH2CH3pvaa' >
Fsel
2241 cacatgccca ccgtgcccag cacctcccgt ggccggcccg tcagtcttcc tcttcccccc aaaacccaag gacaccctca > HCH2CH3pvaa' >
2321 tgatcgcccg gacccctgag gtcacatgcg tggtggtgga cgtgagccac gaagaccctg aggtcaagtt caactggtac > HCH2CH3pvaa' > SacII
2401 gtggacggcg tggaggtgca taatgccaag acaaagccgc gggaggagca gtacaacagc acgtaccgtg tggtcagcgt > HCH2CH3pvaa' >
2461 cctcaccgtc ctgcaccagg actggctgaa tggcaaggag tacaagtgca aggtctccaa caaagccctc ccagccccca > HCH2CH3pvaa' >
2561 tcgagaaaac catctccaaa gccaaagggc agccccgaga accacaggtg tacaccctgc ccccatcccg ggatgagctg > HCH2CH3pvaa' >
2641 accaagaacc aggtcagcct gacctgcctg gtcaaaggct tctatcccag cgacatcgcc gtggagtggg agagcaatgg > HCH2CH3pvaa' >
2721 gcaaccggag aacaactaca agaccacgcc tcccgtgctg gactccgacg gctccttctt cctctacagc aagctcaccg > HCH2CH3pvaa' >
PpulOI
Nsil
2801 tggacaagag caggtggcag caggggaacg tcttctcatg ctccgtgatg catgaggccc tgcacaatca ctatacccag > HCH2CH3pvaa' >
2881 aaatctctga gtctgagccc aggcaagaag gaccccaagt tctgggtcct ggtggtggtg ggaggcgtgc tggcctgtta > HCH2CH3pvaa' »
» CD28tm >
2961 ctctctcctg gtgaccgtgg ccttcatcat cttctgggtg ggagtgcagg tggaaaccat ctccccagga gacgggcgca > CD28tm »
» FKBP12 >
3041 ccttccccaa gcgcggccag acctgcgtgg tgcactacac cgggatgctt gaagatggaa agaaattcga ttcctcccgg
3121 gacagaaaca agccctttaa gtttatgcta ggcaagcagg aggtgatccg aggctgggaa gaaggggttg
> , FKBP12
3201 tgtgggtcag agagccaaac tgactatatc tccagattat gcctatggtg ccactgggca cccaggcatc
> FKBP12
3281 atgccactct cgtcttcgat gtggagcttc taaaactgga acgcgcagag ggccggggct cattgctgac
> FKBP »
.2A.
3361 gtcgaggaaa atcccggccc aatggcttct agaatcctct ggcatgagat gtggcatgaa ggcctggaag aggcatctcg > 2A »
» FRB >
Sphl
3441 tttgtacttt ggggaaagga acgtgaaagg catgtttgag gtgctggagc ccttgcatgc tatgatggaa cggggccccc > FRB >
3521 agactctgaa ggaaacatcc tttaatcagg cctatggtcg agatttaatg gaggcccaag agtggtgcag gaagtacatg > FRB >
Xhol
3601 aaatcaggga atgtcaagga cctcctccaa gcctgggacc tctattatca tgtgttccga cgaatctcaa agctcgagta > FRB »
»
3681 tagcggcggc ggcagcagga gtaagaggag caggctcctg cacagtgact acatgaacat gactccccgc cgccccgggc >....linker .... »
» CD280XZ >
3761 ccacccgcaa gcattaccag ccctatgccc caccacgcga cttcgcagcc tatcgctcca gggaccagag gctgcccccc > CD280XZ >
3841 gatgcccaca agccccctgg gggaggcagt ttccggaccc ccatccaaga ggagcaggcc gacgcccact ccaccctggc > CD280XZ > 3921 caagatcaga gtgaagttca gcaggagcgc agacgccccc gcgtaccagc agggccagaa ccagctctat aacgagctca > CD280XZ >
4001 atctaggacg aagagaggag tacgatgttt tggacaagag acgtggccgg gaccctgaga tggggggaaa gccgagaagg > CD280XZ >
4081 aagaaccctc aggaaggcct gtacaatgaa ctgcagaaag ataagatggc ggaggcctac agtgagattg ggatgaaagg > CD280XZ >
4161 cgagcgccgg aggggcaagg ggcacgatgg cctttaccag ggtctcagta cagccaccaa ggacacctac gacgcccttc > CD280XZ >
4241 acatgcaggc cctgcctcct cgctaa
> CD280XZ »
SEQ ID No. 21 . Inducible CAR comprised of one receptor component and three signalling components. The receptor component comprises of a signal peptide, an anti-CD19 scFv, a spacer derived from the Fc domain of human lgG1 , a CD28 transmembrane domain and FKB12 as endodomain. The first signalling component comprises of FRB and endodomain from CD3 Zeta, the second comprises FRB and CD28 endodomain, the third FRB and 41 BB endodomain. A foot-and-mouth disease 2A peptide separates the components.
SgrAI
1 atggagaccg acaccctgct gctgtgggtg ctgctgctgt gggtgccagg cagcaccggc gacatccaga tgacccagac
>> signal peptide »
» scFv anti-CD19 >
81 caccagcagc ctgagcgcca gcctgggcga ccgggtgacc atcagctgca gagccagcca ggacatcagc aagtacctga
> scFv anti-CD19 >
161 actggtacca gcagaagccc gaoggcaccg tgaagctgct gatotaccac accagccggc tgcacagcgg cgtgcccagc
> scFv anti-CD19 >
241 cggttcagcg gcagcggcag cggcaccgac tacagcctga ccatcagcaa cctggagcag gaggacatcg ccacctactt
> scFv anti-CD19 >
321 ctgccagcag ggcaacaccc tgccctacac cttcggaggc ggcaccaagc tggagatcac caaggccgga ggcggaggct
> scFv anti-CD19 >
401 ctggcggagg cggctctggc ggaggcggct ctggcggagg cggcagcgag gtgaagctgc aggagtctgg cccaggcctg
> scFv anti-CD19 >
481 gtggccccaa gccagagcct gagcgtgacc tgcaccgtga gcggcgtgag cctgcccgac tacggcgtga gctggatcag
> scFv anti-CD19 >
561 gcagccccca cggaagggcc tggagtggct gggcgtgatc tggggcagcg agaccaccta ctacaacagc gccctgaaga
> scFv anti-CD19 >
641 gccggctgac catcatcaag gacaacagca agagccaggt gttcctgaag atgaacagcc tgcagaccga cgacaccgcc
> scFv anti-CD19 > 721 atctactact gcgccaagca ctactactat ggcggcagct acgctatgga ctactggggc cagggcacca gcgtgaccgt >. scFv anti-CD19 >
BamHI Fsel
801 gagctcggat cccgccgagc ccaaatctcc tgacaaaact cacacatgcc caccgtgccc agcacctccc gtggccggcc >.»
» HCH2CH3pvaa >
881 cgtcagtctt cctcttcccc ccaaaaccca aggacaccct catgatcgcc cggacccctg aggtcacatg cgtggtggtg > HCH2CH3pvaa >
961 gacgtgagcc acgaagaccc tgaggtcaag ttcaactggt acgtggacgg cgtggaggtg cataatgcca agacaaagcc > HCH2CH3pvaa >
SacII
1041 gcgggaggag cagtacaaca gcacgtaccg tgtggtcagc gtcctcaccg tcctgcacca ggactggctg aatggcaagg > HCH2CH3pvaa >
1121 agtacaagtg caaggtctcc aacaaagccc tcccagcccc catcgagaaa accatctcca aagccaaagg gcagccccga > HCH2CH3pvaa >
1201 gaaccacagg tgtacaccct gcccccatcc cgggatgagc tgaccaagaa ccaggtcagc ctgacctgcc tggtcaaagg > HCH2CH3pvaa >
1281 cttctatccc agcgacatcg ccgtggagtg ggagagcaat gggcaaccgg agaacaacta caagaccacg cctcccgtgc > HCH2CH3pvaa >
1361 tggactccga cggctccttc ttcctctaca gcaagctcac cgtggacaag agcaggtggc agcaggggaa cgtcttctca > HCH2CH3pvaa >
PpulOI
NslI
BfrBI
1441 tgctccgtga tgcatgaggo cctgcacaat cactataccc agaaatctct gagtctgagc ccaggcaaga aggaccccaa > HCH2CH3pvaa »
1521 gttctgggtc ctggtggtgg tgggaggcgt gctggcctgt tactctctcc tggtgaccgt ggccttcatc atcttctggg ».. CD28tm >
1601 tgggagtgca ggtggaaacc atctccccag gagacgggcg caccttcccc aagcgcggcc agacctgcgt ggtgcactac »
» FKBP12 >
1681 accgggatgc ttgaagatgg aaagaaattc gattcctccc gggacagaaa caagcccttt aagtttatgc taggcaagca > FKBP12 >
1761 ggaggtgatc cgaggctggg aagaaggggt tgcccagatg agtgtgggtc agagagccaa actgactata tctccagatt > FKBP12 >
1841 atgcctatgg tgccactggg cacccaggca tcatcccacc acatgccact ctcgtcttcg atgtggagct tctaaaactg > FKBP12 >
1921 gaacgcgcag agggccgggg ctcattgctg acctgtggag atgtcgagga aaatcccggc ccaatggctt ctagaatcct »>
» FMD-2A »
»..>
2001 ctggcatgag atgtggcatg aaggcctgga agaggcatct cgtttgtact ttggggaaag gaacgtgaaa ggcatgtttg >.. FRB >
Sphl
2081 aggtgctgga gcccttgcat gctatgatgg aacggggccc ccagactctg aaggaaacat cctttaatca ggcctatggt > FRB >
2161 cgagatttaa tggaggccca agagtggtgc aggaagtaca tgaaatcagg gaatgtcaag gacctcctcc aagcctggga > FRB >
Xhol 2241 cctctattat catgtgttcc gacgaatctc aaagctcgag agtggcggag gaggcagttc aaggtccgcc gacgcgcctg > FRB »
».... linker >>
» CD3-Zeta. >
2321 cataccagca ggggcagaat cagctgtaca acgagctcaa ecteggtagg cg gaggaat acgatgtgct cgataagaga
CD3-Zeta
2401 agaggtcgcg atcccgagat gggaggaaaa cct agcgcc gcaagaaccc tcaggagggg ctgtataacg aactgeagaa
, CD3-Zeta
Sfil
2481 ggataagatg gcagaggcct actccgagat tggcatgaag ggtgagagga gaagaggtaa aggccatgac ggcctctacc
)3-7. 1-3
2561 aaggcctctc taccgcaacg aaagacactt atgacgctct gcatatgcag gctctccccc ctaggcagtg cactaattac
.m
.. FMD-2A.. >
2641 gcccttctga aacttgccgg cgatgtggag tctaaccctg gccctatcct gtggcacgag atgtggcatg agggtctgga
.. FRB
2721 agaggccagc cggctgtact tcggagagag aaatgttaag ggtatgttcg aggtgctgga gccgcttcac gctatgatgg
FRB
2801 agaggggccc ccagaccctc aaagaaacca gcttcaatca agectaeggg agggatctta tggaggcaca ggaatggtgt
FRB
2881 cggaagtaca tgaagagcgg gaacgtcaag gacctgctcc aggcttggga tttgtattat cacgtcttta ggcggatcag
.... FRB
2961 caagtctgga ggcggaggaa gtctgcacag cgattacatg aacatgaccc cccgaaggcc cggacccaca cgcaaacact n
3041 atcaacccta tgctccccca cgcgacttcg ccgcctaccg gtcacgcgcc gaggggegeg gc ctttgtt gaettgeggg nn?R
3121 gacgttgaag agaatcctgg ccccatcctt tggcacgaga tgtggcacga gggcctggag gaagcctccc ggctgtattt
, FRB
3201 cggagagcgc aacgtcaaag gaatgtttga ggtgctggag cctctccatg caatgatgga gagggggect cagactctta
3281 aagaaacatc ctttaatcag get acggta gagatttgat ggaggctcaa gaatggtgcc ggaaatacat gaagagtgga
3361 aacgttaaag acctgctgca ggcatgggac ctgtactatc aegtattcag aeggatctea aagtcagggg gcggtggctc
3441 cctttatatc ttcaagcagc ctttcatgag g cggtgcag accacacaag aagaggatgg gtgctcttgc cggttccccg
41BB
3521 aggaggagga gggcggatgc gagctctga
...41BB SEQ ID No. 22. Inducible CAR with multi-spanning receptor component. The receptor component comprises of a signal peptide, an anti-CD19 scFv, a spacer from the Fc domain of human lgG1 , a CD28 transmembrane domain, a first FRB, then the first trans-membrane, minor extracellular loop and second transmembrane domain of CD20, then a second FRB, then the third transmembrane domain, the major extracellular loop and the fourth transmembrane domain of CD20, and a third FRB. The signalling component comprises of FRB and a compound of endodomains from CD28, OX40 and CD3-Zeta. A foot-and-mouth disease 2A peptide separates the two components.
SgrAI
1 atggagaccg acaccctgct gctgtgggtg ctgctgctgt gggtgccagg cagcaccggc gacatocaga tgacccagac >> signal peptide »
» scFv anti-CD19 >
81 caccagcagc ctgagcgcca gcctgggcga ccgggtgacc atcagctgca gagccagcca ggacatcagc aagtacctga > .. .... scFv anti-CD19 >
161 actggtacca gcagaagccc gacggcaccg tgaagctgct gatctaccac accagccggc tgcacagcgg cgtgcccagc > .... scFv anti-CDl9 , >
Xcml
241 cggttcagcg gcagcggcag cggcaccgac tacagcctga ccatcagcaa cctggagcag gaggacatcg ccacctactt > .... scFv anti-CD19 >
321 ctgccagcag ggcaacaccc tgccctacac cttcggaggc ggcaccaagc tggagatcac caaggccgga ggcggaggct > . .. , >
Stul
401 ctggcggagg cggctctggc ggaggcggct ctggcggagg cggcagcgag gtgaagctgc aggagtctgg cccaggcctg > .. ....scFv anti-CDl9 >
481 gtggccccaa gccagagcct gagcgtgacc tgcaccgtga gcggcgtgag cctgcccgac tacggcgtga gctggatcag > .... scFv anti-CDl9 , >
561 gcagccccca cggaagggcc tggagtggct gggcgtgatc tggggcagcg agaccaccta ctacaacagc gccctgaaga > . , ....scFv anti-CDl9 >
641 gccggctgac catcatcaag gacaacagca agagccaggt gttcctgaag atgaacagcc tgcagaccga cgacaccgcc > >
721 atctactact gcgccaagca ctactactat ggcggcagct acgctatgga ctactggggc cagggcacca gcgtgaccgt
>
BamHI Fsel
801 gagctcggat cccgccgagc ccaaatctcc tgacaaaact cacacatgcc caccgtgccc agcacctccc gtggccggcc > . >>
>> HCHPHHlnvaa >
881 cgtcagtctt cctcttcccc ccaaaaccca aggacaccct catgatcgcc cggacccctg aggtcacatg cgtggtggtg > , HCH2CH3pvaa , >
961 gacgtgagcc acgaagaccc tgaggtcaag ttcaactggt acgtggacgg cgtggaggtg cataatgcca agacaaagcc
HCH2CH3pvaa 1041 gcgggaggag cagtacaaca gcacgtaccg tgtggtcagc gtcctcaccg tcctgcacca ggactggctg aatggcaagg > HCH2CH3pvaa >
1121 agtacaagtg caaggtctcc aacaaagccc tcccagcccc catcgagaaa accatctcca aagccaaagg gcagccccga > HCH2CH3pvaa >
1201 gaaccacagg tgtacaccct gcccccatcc cgggatgagc tgaccaagaa ccaggtcagc ctgacctgcc tggtcaaagg > HCH2CH3pvaa >
1281 cttctatccc agcgacatcg ccgtggagtg ggagagcaat gggcaaccgg agaacaacta caagaccacg cctcccgtgc > HCH2CH3pvaa >
1361 tggactccga cggctccttc ttcctctaca gcaagctcac cgtggacaag agcaggtggc agcaggggaa cgtcttctca >. HCH2CH3pvaa >
PpulOI
Nsil
BfrBI
tgctccgtga tgcatgaggc cctgcacaat cactataccc agaaatctct gagtctgagc ccaggcaaga aggaccccaa > HCH2CH3pvaa »
1521 gttctgggtc ctggtggtgg tgggaggcgt gctggcctgt tactctctcc tggtgaccgt ggccttcatc atcttctggg » CD28tm > stui
1601 tgatcctctg gcatgagatg tggcatgaag gcctggaaga ggcatctcgt ttgtactttg gggaaaggaa cgtgaaaggc »
» FRB >
Stui tgtttgagg tgctggagcc cttgcatgct atgatggaac ggggccccca gactctgaag gaaacatcct ttaatcaggc
FRB >
1761 ctatggtcga gatttaatgg aggcccaaga gtggtgcagg aagtacatga aatcagggaa tgtcaaggac ctcctccaag > FRB >
1841 cctgggacct ctattatcat gtgttccgac gaatctcaaa gagcagcctg gtgggaccca cccagagctt cttcatgcgg > FRB »
» CD20 >
Stui
1921 gagagcaaga ccctgggagc cgtgcagatc atgaacggcc tgttccacat cgccctggga ggcctgctga tgatccctgc > CD20 >
2001 cggcatctac gccccaatct gcgtgaccgt gtggtaccca ctgtggggag gcatcatgta catcatcagc ggcagcctgc > CD20 >
2081 tggccgccac cgagaagaac agcggagggg gaagcatcct gtggcacgag atgtggcatg agggtctgga agaggccagc > CD20 »
»...linker...»
» FRB >
2161 cggctgtact tcggagagag aaatgttaag ggtatgttcg aggtgctgga gccgcttcac gctatgatgg agaggggccc > . FRB >
2241 ccagaccctc aaagaaacca gcttcaatca agcctacggg agggatctta tggaggcaca ggaatggtgt cggaagtaca > FRB > 2321 tgaagagcgg gaacgtcaag gacctgctcc aggcttggga tt gtattat cacgtcttta ggcggatcag caagtctgga > FRB »
» ... >
Bell
ggcggaggaa gtcggaagtg cctggtgaag ggcaagatga tcatgaacag cctgagcctg ttcgccgcca teageggcat >..linker..»
» CD20 >
2481 gatcctgagc atcatggaca tcctgaacat caagatcagc cacttcctga agatggagag cctgaacttc atccgggccc > CD20 >
2561 acaccccata catcaacatc tacaactgcg agcctgccaa ccccagcgag aagaacagcc ccagcaccca gtactgctac > CD20 >
2641 agcatccaga gcctgttcct gggcatcctg agcgtgatgc tgatcttege cttcttccag gagctggtga tcgccggcat > CD20 >
2.721 cgtggagaac gagtggaagc ggacctgcag ccggcccaag agcatcctgt ggcacgagat gtggcatgag ggtctggaag > CD20 »
» FRB >
2801 aggccagccg gctgtacttc ggagagagaa atgttaaggg tatgttcgag gtgctggagc cgcttcacgc tatgatggag > FRB >
2881 aggggccccc agaccctcaa agaaaccagc ttcaatcaag ectaegggag ggatcttatg gaggcacagg aatggtgtcg > FRB >
2961 gaagtacatg aagagcggga aegtcaagga cctgctccag gcttgggatt tgtattatca cgtctttagg eggatcagea > FRB >
3041 agegegcaga gggcegggge teattgetga cctgtggaga tgtcgaggaa aatcccggcc caggagtgea ggtggaaacc »>
» FMD-2A »
» FKBP12 >
3121 atctccccag gagacgggcg caccttcccc aagcgcggcc agacctgcgt ggtgcactac accgggatgc ttgaagatgg > F BP12 >
3201 aaagaaattc gattcctccc gggacagaaa caagcccttt aagtttatgc taggcaagca ggaggtgatc cgaggctggg > FKBP12 >
3281 aagaaggggt tgcccagatg agtgtgggtc agagagecaa actgactata tctccagatt atgcctatgg tgccactggg > FKBP12 >
3361 cacccaggca tcatcccacc acatgccact ctcgtcttcg atgtggagct tctaaaactg gaaaggagta agaggagcag > FKBP12 »
» 280XZ > gctcctgcac agtgactaca tgaacatgac tccccgccgc cccgggccca cccgcaagca ttaccagccc tatgccccac > 280XZ >
3521 cacgcgactt cgcagcctat cgctccaggg accagaggct gccccccgat gcccacaagc cccctggggg aggcagtttc > 280XZ >
3601 cggaccccca tccaagagga gcaggccgac gcccactcca ccctggccaa gatcagagtg aagttcagca ggagegcaga > 280XZ > 3681 cgcccccgcg taccagcagg gccagaacca gctctataac gagctcaatc taggacgaag agaggagtac gatgttttgg > 280XZ >
3761 acaagagacg tggccgggac octgagatgg ggggaaagcc gagaaggaag aaccctcagg aaggcctgta caatgaactg > 280XZ >
3841 cagaaagata agatggcgga ggcctacagt gagattggga tgaaaggcga gcgccggagg ggcaaggggc acgatggcct > 280XZ >
3921 ttaccagggt ctcagtacag ccaccaagga cacctacgac gcccttcaca tgcaggccct gcctcctcgc tga
> 280XZ »
SEQ ID No. 23. Simple inducible CAR comprised of one receptor component and one signalling component. The signalling component comprises an ecdysone receptor and the CD3-Zeta endodomain (italic). A FMD-2A peptide (shown in bold) separates this signalling component from the receptor component. The receptor component comprises an anti-CD19 scFv, a HCH2CH3 spacer (underlined), a CD148 transmembrane domain (highlighted in grey) and the retinoid X receptor as endodomain.
SEQ ID No. 23
MPVDRILEAELAVEQKSDQGVEGPGGTGGSGSSPNDPVTNICQAADKQLFTLVEWAKRIP
HFSSLPLDDQVILLRAGWNELLIASFSHRSIDVRDGILLATGLHVHRNSAHSAGVGAIFD
RVLTELVSKMRDMRMDKTELGCLRAIILFNPEVRGLKSAQEVELLREKVYAALEEYTRTT
HPDEPGRFAKLLLRLPSLRSIGLKCLEHLFFFRLIGDVPIDTFLMEMLESPSDSSGGGSA
DAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMA
EAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPRRREGRGSItliTCG VEE
NPGPMETDTLLLWVLLLWVPGSTGDIQMTQTTSSLSASLGDRVTISCRASQDISKYLNWY
QQKPDGTVKLLIYHTSRLHSGVPSRFSGSGSGTDYSLTISNLEQEDIATYFCQQGNTLPY
TFGGGTKLEITKAGGGGSGGGGSGGGGSGGGGSEVKLQESGPGLVAPSQSLSVTCTVSGV
SLPDYGVSWIRQPPRKGLEWLGVIWGSETTYYNSALKSRLTIIKDNSKSQVFLKMNSLQT
DDTAIYYCAKHYYYGG5YAMDYWGQGTSVTVSSDPAEPKSPDKTHTCPPCPAPPVAGPSV
FLFPPKPKDTLMIARTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTY
RVVSVLTVLHQD LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTK
NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQG
NVFSCSVMHEALHNHYTQKSLSLSPGKKDPM^
RQAI'^iAQTKIWVSEKKTAQA
EKDKLPVSTTTVDDHMPPIMQCEPPPPEAARIHEVVPRFLSDKLLETNRQKNIPQLTANQ QFLIARLIWYQDGYEQPSDEDLKRITQTWQQADDENEESDTPFRQITEMTILTVQLIVEF AKGLPGFAKISQPDQITLLKACSSEVMMLRVARRYDAASDSILFANNQAYTRDNYRKAGM AEVIEDLLHFCRCMYSMALDNIHYALLTAVVIFSDRPGLEQPQLVEEIQRYYLNTLRIYI LNQLSGSARSSVIYGKILSILSELRTLGMQNSNMCISLKLKNRKLPPFLEEIWDVADMSH TQPPPILESPTNL SEQ ID No. 24. Simple inducible CAR comprised of one receptor component and one signalling component. The signalling component comprises an FKBP12 domain (underlined) and the CD3-Zeta endodomain (italic). A FMD-2A peptide (shown in bold) separates this signalling component from the receptor component. The receptor component comprises an anti-CD19 scFv, a HCH2CH3 spacer (highlighted in grey), a CD148 transmembrane domain and the and FKBP12 domain (underlined).
SEQ ID No. 24
MGVQVETISPGDGRTFPKRGQTCVVHYTGMLEDGKKVDSSRDRNKPFKFMLGKQEVIRGW
EEGVAQMSVGQRAKLTISPDYAYGATGHPGIIPPHATLVFDVELLKLESGGGSAPAPArQ
QGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEG YNELQKDKMAEAYSEI
GMKGERRRGKGHDGLYQGLSTATKDTYDALM^QALPPRBI&GRGSI LTCGOVEENPGPME
TDTLLL VLLLWVPGSTGDIQMTQTTSSLSASLGDRVTISCRASQDISKYLNWYQQKPDG
TVKLLIYHTSRLHSGVPSRFSGSGSGTDYSLTISNLEQEDIATYFCQQGNTLPYTFGGGT
KLEITKAGGGGSGGGGSGGGGSGGGGSEVKLQESGPGLVAPSQSLSVTCTVSGVSLPDYG
VSWIRQPPRKGLEWLGVIWGSETTYYNSALKSRLTIIKDNSKSQVFLKMNSLQTDDTAIY
YCAKHYYYGGSYAMDYWGQGTSVTVSSDPAEPKSPDKTHTCPPC.PAPPVAGPSVTLFPPK
PKDTEMIARTPEVTG VDVSHEDPEVKFN YVDGVEVHNAKTKPREEQYNSTYRWSVL
TVIJ-IQEi LNGKEYKCKVS KAI.PAPI !!n iSKAKGQPREPQVyi\L PSil) !J'KNQVSI!'F
CLVK F P^
¼HE LMH¾£¾
AQIKRVVSEKKTAQAPHRFQKTCSPISGGGGSGVQVETISPGDGRTFPKRGQTCVVHYTG MLEDGKKVDSSRDRNKPFKFMLGKQEVIRG EEGVAQMSVGQRA LTISPDYAYGATGHP GIIPPHATLVFDVELLKLE
All publications mentioned in the above specification are herein incorporated by reference in their entirety, with special attention to the subject matter for which they are referred. Various modifications and variations of the described methods and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention which are obvious to those skilled in molecular biology or related fields are intended to be within the scope of the following claims.

Claims

1. A chimeric antigen receptor (CAR) signalling system comprising;
(i) a receptor component comprising an extracellular antigen-binding domain, a transmembrane domain and a intracellular first chemical inducer of dimerization (CID) binding domain (CBD1 ); and
(ii) an intracellular signalling component comprising a signalling domain and a second CID binding domain (CBD2);
wherein CBD1 and CBD2 are capable of simultaneously binding to a CID;
wherein, in the absence of CID, binding of the antigen-binding domain to antigen does not result in signalling through the signalling domain; whilst, in the presence of CID, the receptor component and the signalling component heterodimerize and binding of the antigen-binding domain to antigen results in signalling through the signalling domain.
2. The CAR signalling system according to claim 1 wherein CBD1 and CBD2 comprise different CID binding domains and the CID comprises two different binding moieties.
3. The CAR signalling system according to claim 2, wherein:
(i) CBD1 comprises the rapamycin binding domain of FK-binding protein 12 (FKBP12), and CBD2 comprises the FKBP12-Rapamycin Binding (FRB) domain of mTOR; or
(ii) CBD1 comprises the rapamycin binding domain of FK-binding protein 12 (FKBP12), and CBD2 comprises the FKBP12-Rapamycin Binding (FRB) domain of mTOR
and the CID is rapamycin or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
4. The CAR signalling system according to claim 2 wherein:
(i) CBD1 comprises the FK506 (Tacrolimus) binding domain of FK-binding protein 12 (FKBP12) and CBD2 comprises the cyclosporin binding domain of cylcophilin A; or
(ii) CBD1 comprises the cyclosporin binding domain of cylcophilin A and CBD2 comprises the FK506 (Tacrolimus) binding domain of FK-binding protein 12 (FKBP12), and the CID is an FK506/cyclosporin fusion or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
5. The CAR signalling system according to claim 2 wherein:
(i) CBD1 comprises an oestrogen-binding domain (EBD) and CBD2 comprises a streptavidin binding domain; or
(ii) CBD1 (A) comprises a streptavidin binding domain and, CBD2(B) comprises an oestrogen-binding domain (EBD),
and the CID is an estrone/biotin fusion protein or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
6. The CAR signalling system according to claim 2 wherein:
(i) CBD1 comprises a glucocorticoid-binding domain (GBD) and CBD2 comprises a dihydrofolate reductase (DHFR) binding domain; or
(ii) CBD1 (A) comprises a dihydrofolate reductase (DHFR) binding domain and CBD2(B) comprises a glucocorticoid-binding domain (GBD),
and the CID is a dexamethasone/methotrexate fusion protein or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
7. The CAR signalling system according to claim 2 wherein:
(i) CBD1 comprises an Oe-alkylguanine-DNA alkyltransferase (AGT) binding domain and CBD2 comprises a dihydrofolate reductase (DHFR) binding domain; or
(ii) CBD1 comprises a dihydrofolate reductase (DHFR) binding domain and CBD2 comprises an 06-alkylguanine-DNA alkyltransferase (AGT) binding domain and the CID is an 06-benzylguanine derivative/methotrexate fusion protein or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
8. The CAR signalling system according to claim 2, wherein:
(i) CBD1 comprises a retinoic acid receptor domains and CBD2 comprises an ecodysone receptor domain; or
(ii) CBD1 comprises an ecodysone receptor domain and CBD2 comprises a retinoic acid receptor domains and the CID is RSL1 or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
9. The CAR signalling system according to claim 1 wherein CBD1 and CBD2 are similar and the CID comprises two identical binding moieties.
10. The CAR signalling system according to claim 8 wherein CBD1 and CBD2 comprise FK506 binding protein (FKBP) binding domains comprising a F36V mutation and the CID is AP1903 or a derivative thereof which is capable of causing the receptor component and the signalling component to heterodimerize.
1 1. The CAR signalling system according to claims 1 to 10, wherein there are multiple receptor components, each recognizing a different antigen.
12. The CAR signalling system according to claim 1 1 , wherein the CBD1 of the multiple receptor components differ in binding to CID such that each antigen propagates different signalling strengths.
13. The CAR signalling system according to any of claims 1 to 10, wherein the signalling domain of the signalling component comprises a single endodomain selected from CD3 zeta endodomain, CD28 endodomain, 4 BB endodomain and OX40 endodomain.
14. The CAR signalling system according to any preceding claim, wherein the signalling domain of the signalling component comprises at least one of CD3 zeta endodomain, CD28 endodomain, 41 BB endodomain and OX40 endodomain.
15. The CAR signalling system according to any preceding claim which comprises a plurality of signalling components, each comprising a signalling domain and a CBD2, wherein the CBD2 each recognise the same CID but the signalling domains comprise different endodomains.
16. The CAR signalling system according to claim 15 wherein the plurality of signalling components comprise a plurality of CBD2s, each of which independently recognise the CID with different affinities.
17. A receptor component suitable for use in the CAR signalling system according to any of claims 1 to 16 which comprises an antigen-binding domain, a transmembrane domain and a CID binding domain.
18. A signalling component suitable for use in the CAR signalling system according to any one of claims 1 to 16 which comprises a signalling domain and a CID binding domain.
19. A nucleic acid encoding the receptor component according to claim 17.
20. A nucleic acid encoding the signalling component according to claim 18.
21. A nucleic acid sequence encoding a CAR signalling system according to any of claims 1 to 16, wherein the receptor component and signalling component are co- expressed by means of a self-cleaving peptide which is cleaved between the receptor component and the signalling component after translation.
22. A nucleic acid sequence according to claim 21 encoding the CAR signalling system according to claim 11 , 12, 5 or 16, wherein the multiple receptor components or multiple signalling components are co-expressed by means of a self-cleaving peptide which is cleaved between the receptor component(s) and the signalling component(s).
23. A vector comprising a nucleic acid sequence according to any of claims 19 to 22.
24. A retroviral vector or a lentiviral vector or a transposon comprising a vector according to claim 23.
25. A T cell or NK cell which expresses a receptor component according to claim 17 and at least one signalling component according to claim 8.
26. A T cell or NK cell according to claim 25 which comprises the nucleic acid according to any of claims 19 to 22 or the vector according to claim 23 or 24.
27. A pharmaceutical composition comprising a plurality of T cells or NK cells according to claim 25 or 26.
28. A pharmaceutical composition according to claim 27 for use in treating and/or preventing a disease.
29. A method for treating and/or preventing a disease, which comprises the step of administering a pharmaceutical composition according to claim 27 to a subject.
30. A method according to claim 29, which comprises the following steps:
(i) isolation of a T cell or NK containing sample from a subject;
(ii) transduction or transfection of the T or NK cells with a nucleic acid sequence according to any of claims 18 to 20 or a vector according to claim 21 or 22; and
(iii) administering the T cells or NK cells from (ii) to the subject.
31. A method according to claim 29 or 30, which further comprises the step of administering a CID for use in the CAR signalling system according to any of claims 1 to 16 to the subject.
32. A method for treating and/or preventing a disease in a subject which subject comprises T cells or NK cells according to claim 25 or 26, which comprises the step of administering a CID suitable for use in the CAR signalling system according to any of claims 1 to 16 to the subject.
33. A method according to any of claims 29 to 32, which involves monitoring the progression of disease and/or monitoring toxic activity in the subject and adjusting the dose of the CID to provide acceptable levels of disease progression and/or toxic activity.
34. A pharmaceutical composition for use according to claim 28 or a method according to any of claims 29 to 33, wherein the disease is a cancer.
35. The use of a pharmaceutical composition according to claim 27 in the manufacture of a medicament for the treatment and/or prevention of a disease.
36. A kit which comprises a nucleic acid according to any of claims 19 to 22 or the vector according to claim 23 or 24.
37. A kit according to claim 36 which also comprises a CID suitable for use in the CAR signalling system according to any of claims 1 to 16.
38. A kit which comprises a T or NK cell according to claim 25 or 26 and a CID suitable for use in the CAR signalling system according to any of claims 1 to 16.
39. A method for making a T or NK cell according to claim 25 or 26, which comprises the step of introducing: a nucleic acid sequence according to any of claims 19 to 22 or the vector according to claim 23 or 24, into a T cell.
40. A method according to claim 39, wherein the T or NK cell is from a sample isolated from a subject.
41. A method for activating the CAR signalling system according to any of claims 1 to 16 in a subject which comprises a T or NK cell according to claim 25, which method comprises the step of administering the CID to the subject.
42. A method for reducing the activity of the CAR signalling system according to any of claims 1 to 16, in a subject which comprises a T or NK cell according to claim 25, which method comprises reducing or stopping administration of the CID to the subject.
43. A method for inducing dimerization in vivo between a receptor component according to claim 17 and a signalling component according to claim 18, which comprises the step of administering a chemical inducer of dimerization (CID) to a subject comprising a cell according to claim 25 or 26.
PCT/GB2015/050974 2014-04-01 2015-03-31 Chimeric antigen receptor (car) signalling system WO2015150771A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
DK15714272.0T DK3126380T3 (en) 2014-04-01 2015-03-31 CHIMERIC ANTIGEN RECEPTOR SIGNAL SYSTEM (CAR)
PL15714272T PL3126380T3 (en) 2014-04-01 2015-03-31 Chimeric antigen receptor (car) signalling system
ES15714272T ES2877129T3 (en) 2014-04-01 2015-03-31 Chimeric antigen receptor (CAR) signaling system
EP15714272.0A EP3126380B1 (en) 2014-04-01 2015-03-31 Chimeric antigen receptor (car) signalling system
US15/301,148 US10588967B2 (en) 2014-04-01 2015-03-31 Chimeric antigen receptor (CAR) signalling system
US16/775,257 US20200338194A1 (en) 2014-04-01 2020-01-28 Chimeric antigen receptor (car) signalling system

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB201405845A GB201405845D0 (en) 2014-04-01 2014-04-01 Signalling system
GB1405845.7 2014-04-01

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/301,148 A-371-Of-International US10588967B2 (en) 2014-04-01 2015-03-31 Chimeric antigen receptor (CAR) signalling system
US16/775,257 Continuation US20200338194A1 (en) 2014-04-01 2020-01-28 Chimeric antigen receptor (car) signalling system

Publications (1)

Publication Number Publication Date
WO2015150771A1 true WO2015150771A1 (en) 2015-10-08

Family

ID=50737808

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2015/050974 WO2015150771A1 (en) 2014-04-01 2015-03-31 Chimeric antigen receptor (car) signalling system

Country Status (9)

Country Link
US (2) US10588967B2 (en)
EP (1) EP3126380B1 (en)
DK (1) DK3126380T3 (en)
ES (1) ES2877129T3 (en)
GB (1) GB201405845D0 (en)
HU (1) HUE054892T2 (en)
PL (1) PL3126380T3 (en)
PT (1) PT3126380T (en)
WO (1) WO2015150771A1 (en)

Cited By (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3025719A1 (en) 2014-11-26 2016-06-01 Miltenyi Biotec GmbH Combination immunotherapy of antigen-recognizing receptors and hematopoietic cells for the treatment of diseases
WO2016124930A1 (en) * 2015-02-05 2016-08-11 Ucl Business Plc Signalling system
WO2017070554A1 (en) 2015-10-23 2017-04-27 Fred Hutchinson Cancer Research Center Methods to create chemically-induced dimerizing protein systems for regulation of cellular events
WO2017081479A1 (en) * 2015-11-11 2017-05-18 Ucl Business Plc Multi-span chimeric antigen receptor
EP3184548A1 (en) 2015-12-23 2017-06-28 Miltenyi Biotec GmbH Chimeric antigen receptor with cytokine receptor activating or blocking domain
WO2017137758A1 (en) * 2016-02-12 2017-08-17 Autolus Limited Signalling system
WO2017137759A1 (en) * 2016-02-12 2017-08-17 Autolus Limited Signalling system
WO2017123559A3 (en) * 2016-01-11 2017-08-24 The Board Of Trustees Of The Leland Stanford Junior University Chimeric proteins and methods of regulating gene expression
EP3211420A1 (en) 2016-02-29 2017-08-30 Miltenyi Biotec GmbH Assay for detection of chimeric antigen receptor t cells
WO2017180993A1 (en) * 2016-04-14 2017-10-19 Bluebird Bio, Inc. Salvage chimeric antigen receptor systems
WO2017180587A3 (en) * 2016-04-11 2017-12-14 Obsidian Therapeutics, Inc. Regulated biocircuit systems
WO2017216562A1 (en) * 2016-06-16 2017-12-21 Autolus Limited Tunable chimeric antigen receptors
WO2018087557A1 (en) 2016-11-11 2018-05-17 Autolus Limited Chimeric antigen receptor
CN108138201A (en) * 2015-09-04 2018-06-08 托卡根公司 Include the recombinant vector of 2A peptides
WO2018150187A1 (en) 2017-02-17 2018-08-23 Autolus Limited Chimeric cytokines receptors
WO2018183551A1 (en) * 2017-03-28 2018-10-04 Cell Design Labs, Inc. Chimeric polypeptides and methods of altering their localisation in the cell membrane
US10105391B2 (en) 2013-02-15 2018-10-23 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof
EP3253865A4 (en) * 2015-02-06 2018-10-24 National University of Singapore Methods for enhancing efficacy of therapeutic immune cells
WO2018211244A1 (en) * 2017-05-15 2018-11-22 Autolus Limited A cell comprising a chimeric antigen receptor (car)
KR20180130534A (en) * 2016-03-29 2018-12-07 유니버시티 오브 써던 캘리포니아 Chimeric antigen receptor targeting cancer
WO2018224844A1 (en) 2017-06-09 2018-12-13 Autolus Limited Anti trbc1 antigen binding domains
WO2018229492A1 (en) 2017-06-15 2018-12-20 Autolus Limited Chimeric antigen receptor
EP3438123A1 (en) * 2015-08-20 2019-02-06 Autolus Limited Chimeric cytokine receptor
WO2019053420A1 (en) * 2017-09-13 2019-03-21 Autolus Limited Cell
GB201903237D0 (en) 2019-03-08 2019-04-24 Autolus Ltd Method
WO2019122875A1 (en) * 2017-12-20 2019-06-27 Ucl Business Plc Chimeric antigen receptor
US10336807B2 (en) 2016-01-11 2019-07-02 The Board Of Trustees Of The Leland Stanford Junior University Chimeric proteins and methods of immunotherapy
GB201910185D0 (en) 2019-07-16 2019-08-28 Autolus Ltd Method
WO2019162695A1 (en) * 2018-02-26 2019-08-29 Autolus Limited Cell
US10428305B2 (en) 2014-05-15 2019-10-01 National University Of Singapore Modified natural killer cells that express IL15 and uses thereof
WO2019215453A1 (en) 2018-05-11 2019-11-14 Autolus Limited Car-t cells expressing an inhibitory anti-tgf-beta-receptor ii single domain antibody
US10478457B2 (en) 2015-02-24 2019-11-19 Ucl Business Ltd Chimeric protein
WO2019220109A1 (en) 2018-05-15 2019-11-21 Autolus Limited Chimeric antigen receptor
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
US10550183B2 (en) 2016-11-22 2020-02-04 National University Of Singapore Blockade of CD7 expression and chimeric antigen receptors for immunotherapy of T-cell malignancies
GB201919017D0 (en) 2019-12-20 2020-02-05 Autolus Ltd Cell
WO2020035676A1 (en) 2018-08-13 2020-02-20 Autolus Limited Car t-cells comprising an anti cd33, an anti cll1 and at least one further car anti cd123 and/or ftl3
US10588967B2 (en) 2014-04-01 2020-03-17 Ucl Business Ltd Chimeric antigen receptor (CAR) signalling system
WO2020065330A2 (en) 2018-09-27 2020-04-02 Autolus Limited Chimeric antigen receptor
US10654927B2 (en) 2014-08-29 2020-05-19 Ucl Business Ltd Signalling system
WO2020183131A1 (en) 2019-03-08 2020-09-17 Autolus Limited Compositions and methods comprising engineered chimeric antigen receptor and modulator of car
WO2020222021A1 (en) 2019-05-02 2020-11-05 Autolus Limited Engineered t-cells co-expressing an anti-bcma car and an anti-ectoenzyme antibody and their use in the treatment of cancer
GB202017343D0 (en) 2020-11-02 2020-12-16 Autolus Ltd Cell
WO2021009510A1 (en) 2019-07-16 2021-01-21 Autolus Limited Method for preconditioning a subject who is about to receive a t-cell therapy
WO2021023987A1 (en) 2019-08-05 2021-02-11 Autolus Limited Chimeric cytokine receptor
GB202101491D0 (en) 2021-02-03 2021-03-17 Autolus Ltd Molecule
WO2021048564A2 (en) 2019-09-13 2021-03-18 Autolus Limited Antigen-binding domain
WO2021123810A1 (en) 2019-12-20 2021-06-24 Autolus Limited Antigen-binding domain binding to psma
US11058725B2 (en) 2019-09-10 2021-07-13 Obsidian Therapeutics, Inc. CA2 compositions and methods for tunable regulation
US11136562B2 (en) 2016-01-08 2021-10-05 The Regents Of The University Of California Conditionally active heterodimeric polypeptides and methods of use thereof
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
WO2021205175A1 (en) 2020-04-09 2021-10-14 Autolus Limited Molecule
WO2021205172A1 (en) 2020-04-08 2021-10-14 Autolus Limited Cell
WO2021229218A1 (en) 2020-05-13 2021-11-18 Autolus Limited Method
EP3916388A1 (en) 2020-05-27 2021-12-01 Miltenyi Biotec B.V. & Co. KG Reagents for detection of chimeric antigen receptor cells
EP3925618A1 (en) * 2013-07-29 2021-12-22 2seventy bio, Inc. Multipartite signaling proteins and uses thereof
US11365236B2 (en) 2017-03-27 2022-06-21 Nkarta, Inc. Truncated NKG2D chimeric receptors and uses thereof in natural killer cell immunotherapy
US11472860B2 (en) * 2016-09-08 2022-10-18 Westfaelisch-Wilhelms-Universitaet Chimeric antigen receptors
WO2023073352A1 (en) 2021-10-25 2023-05-04 Autolus Limited Chimeric cytokine receptor
US11648269B2 (en) 2017-08-10 2023-05-16 National University Of Singapore T cell receptor-deficient chimeric antigen receptor T-cells and methods of use thereof
US11851672B2 (en) 2017-10-25 2023-12-26 Autolus Limited CAR T-cells comprising transcriptional circuits
WO2024009093A1 (en) 2022-07-06 2024-01-11 Autolus Limited Engineered trispecific tancar expressing cells targeting cd33, cd123 and cll1 and uses thereof in cancer therapy
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
US11993794B2 (en) 2021-03-16 2024-05-28 Fred Hutchinson Cancer Center Methods to create chemically-induced dimerizing protein systems for regulation of cellular events

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018144535A1 (en) * 2017-01-31 2018-08-09 Novartis Ag Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
EP3662055A1 (en) * 2017-08-02 2020-06-10 Autolus Limited Cells expressing a chimeric antigen receptor or engineered tcr and comprising a nucleotide sequence which is selectively expressed
CN109836497A (en) * 2017-11-25 2019-06-04 深圳宾德生物技术有限公司 A kind of single-chain antibody of targeting EGFR, Chimeric antigen receptor T cell and its preparation method and application
WO2019246593A2 (en) * 2018-06-22 2019-12-26 Fred Hutchinson Cancer Research Center Compositions and methods to target cll-1 and cd123 for the treatment of acute myeloid leukemia and related disorders

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012099973A2 (en) * 2011-01-18 2012-07-26 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer
WO2014127261A1 (en) * 2013-02-15 2014-08-21 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4208479A (en) * 1977-07-14 1980-06-17 Syva Company Label modified immunoassays
US7229960B2 (en) * 2000-11-03 2007-06-12 University Of Vermont And State Agricultural College Methods and compositions for inhibiting GRB7
EP2534257B1 (en) 2010-02-12 2017-09-27 Oncomed Pharmaceuticals, Inc. Methods for identifying and isolating cells expressing a polypeptide
EP3012268B1 (en) 2010-09-08 2017-11-15 Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus Chimeric antigen receptors with an optimized hinge region
FI2997141T3 (en) 2013-05-13 2022-12-15 Cd19 specific chimeric antigen receptor and uses thereof
US10287354B2 (en) * 2013-12-20 2019-05-14 Novartis Ag Regulatable chimeric antigen receptor
US20170081411A1 (en) 2014-03-15 2017-03-23 Novartis Ag Regulatable chimeric antigen receptor
GB201405845D0 (en) 2014-04-01 2014-05-14 Ucl Business Plc Signalling system
GB201415347D0 (en) 2014-08-29 2014-10-15 Ucl Business Plc Signalling system
GB201501936D0 (en) 2015-02-05 2015-03-25 Ucl Business Plc Signalling system
GB201504840D0 (en) 2015-03-23 2015-05-06 Ucl Business Plc Chimeric antigen receptor
GB201602571D0 (en) 2016-02-12 2016-03-30 Autolus Ltd Signalling system
GB201602563D0 (en) 2016-02-12 2016-03-30 Autolus Ltd Signalling system
GB201610515D0 (en) 2016-06-16 2016-08-03 Autolus Ltd Cell

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012099973A2 (en) * 2011-01-18 2012-07-26 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer
WO2014127261A1 (en) * 2013-02-15 2014-08-21 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LESLIE E HUYE ET AL: "Combining mTor Inhibitors With Rapamycin-resistant T Cells: A Two-pronged Approach to Tumor Elimination", MOLECULAR THERAPY, vol. 19, no. 12, 30 August 2011 (2011-08-30), GB, pages 2239 - 2248, XP055191016, ISSN: 1525-0016, DOI: 10.1038/mt.2011.179 *
MICHAEL C JENSEN ET AL: "Design and implementation of adoptive therapy with chimeric antigen receptor-modified T cells Authors' addresses", IMMUNOLOGICAL REVIEWS., vol. 257, no. 1, Special Issue, 13 December 2013 (2013-12-13), MUNKSGAARD., pages 127 - 144, XP055156740, ISSN: 0105-2896 *

Cited By (112)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US10836999B2 (en) 2013-01-28 2020-11-17 St. Jude Children's Research Hospital, Inc. Chimeric receptor with NKG2D specificity for use in cell therapy against cancer and infectious disease
US10829737B2 (en) 2013-01-28 2020-11-10 St. Jude Children's Research Hospital, Inc. Chimeric receptor with NKG2D specificity for use in cell therapy against cancer and infectious disease
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
US11478510B2 (en) 2013-02-15 2022-10-25 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof
US10632152B2 (en) 2013-02-15 2020-04-28 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof
US10888581B2 (en) 2013-02-15 2021-01-12 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof
US10105391B2 (en) 2013-02-15 2018-10-23 The Regents Of The University Of California Chimeric antigen receptor and methods of use thereof
US11530265B2 (en) 2013-07-29 2022-12-20 2Seventy Bio, Inc. Multipartite signaling proteins and uses thereof
EP3925618A1 (en) * 2013-07-29 2021-12-22 2seventy bio, Inc. Multipartite signaling proteins and uses thereof
US10588967B2 (en) 2014-04-01 2020-03-17 Ucl Business Ltd Chimeric antigen receptor (CAR) signalling system
US11560548B2 (en) 2014-05-15 2023-01-24 National University Of Singapore Immune cells expressing membrane-bound interleukin 15 (mbIL15) 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
US10428305B2 (en) 2014-05-15 2019-10-01 National University Of Singapore Modified natural killer cells that express IL15 and uses thereof
US10654927B2 (en) 2014-08-29 2020-05-19 Ucl Business Ltd Signalling system
EP3025719A1 (en) 2014-11-26 2016-06-01 Miltenyi Biotec GmbH Combination immunotherapy of antigen-recognizing receptors and hematopoietic cells for the treatment of diseases
WO2016124930A1 (en) * 2015-02-05 2016-08-11 Ucl Business Plc Signalling system
US10604570B2 (en) 2015-02-05 2020-03-31 Ucl Business Ltd Chimeric antigen receptor signalling system comprising heterodimerization domains
EP3253865A4 (en) * 2015-02-06 2018-10-24 National University of Singapore Methods for enhancing efficacy of therapeutic immune cells
US10765699B2 (en) 2015-02-06 2020-09-08 National University Of Singapore Methods for enhancing efficacy of therapeutic immune cells
US11679132B2 (en) 2015-02-06 2023-06-20 National University Of Singapore Methods for enhancing efficacy of therapeutic immune cells
US11103532B2 (en) 2015-02-24 2021-08-31 Autolus Limited Chimeric protein
US10478457B2 (en) 2015-02-24 2019-11-19 Ucl Business Ltd Chimeric protein
EP3438123A1 (en) * 2015-08-20 2019-02-06 Autolus Limited Chimeric cytokine receptor
US10800855B2 (en) 2015-08-20 2020-10-13 Autolus Limited Chimeric cytokine receptors and encoding nucleic acids
US11479613B2 (en) 2015-08-20 2022-10-25 Autolus Limited Methods for treating cancer or modulating T cells or NK cells in a subject by administering cells comprising chimeric transmembrane proteins
US10800854B2 (en) 2015-08-20 2020-10-13 Autolus Limited Chimeric cytokine receptors and encoding nucleic acids
US11479614B2 (en) 2015-08-20 2022-10-25 Autolus Limited Methods for treating cancer or modulating T cells or NK cells in a subject with cancer by administering cells comprising chimeric cytokine receptors
EP4338756A2 (en) 2015-08-20 2024-03-20 Autolus Limited Chimeric cytokine receptor
CN108138201A (en) * 2015-09-04 2018-06-08 托卡根公司 Include the recombinant vector of 2A peptides
EP3365442A4 (en) * 2015-10-23 2019-05-01 Fred Hutchinson Cancer Research Center Methods to create chemically-induced dimerizing protein systems for regulation of cellular events
WO2017070554A1 (en) 2015-10-23 2017-04-27 Fred Hutchinson Cancer Research Center Methods to create chemically-induced dimerizing protein systems for regulation of cellular events
US10954500B2 (en) 2015-10-23 2021-03-23 Fred Hutchinson Cancer Research Center Methods to create chemically-induced dimerizing protein systems for regulation of cellular events
WO2017081479A1 (en) * 2015-11-11 2017-05-18 Ucl Business Plc Multi-span chimeric antigen receptor
US11512139B2 (en) 2015-12-23 2022-11-29 Miltenyi Biotec B.V. & Co. KG Chimeric antigen receptor with cytokine receptor activating or blocking domain
EP3184548A1 (en) 2015-12-23 2017-06-28 Miltenyi Biotec GmbH Chimeric antigen receptor with cytokine receptor activating or blocking domain
US11136562B2 (en) 2016-01-08 2021-10-05 The Regents Of The University Of California Conditionally active heterodimeric polypeptides and methods of use thereof
WO2017123559A3 (en) * 2016-01-11 2017-08-24 The Board Of Trustees Of The Leland Stanford Junior University Chimeric proteins and methods of regulating gene expression
US11111287B2 (en) 2016-01-11 2021-09-07 The Board Of Trustees Of The Leland Stanford Junior University Chimeric proteins and methods of immunotherapy
US10457961B2 (en) 2016-01-11 2019-10-29 The Board Of Trustees Of The Leland Stanford Junior University Chimeric proteins and methods of regulating gene expression
US10336807B2 (en) 2016-01-11 2019-07-02 The Board Of Trustees Of The Leland Stanford Junior University Chimeric proteins and methods of immunotherapy
US11773411B2 (en) 2016-01-11 2023-10-03 The Board Of Trustees Of The Leland Stanford Junior University Chimeric proteins and methods of regulating gene expression
US9856497B2 (en) 2016-01-11 2018-01-02 The Board Of Trustee Of The Leland Stanford Junior University Chimeric proteins and methods of regulating gene expression
WO2017137759A1 (en) * 2016-02-12 2017-08-17 Autolus Limited Signalling system
WO2017137758A1 (en) * 2016-02-12 2017-08-17 Autolus Limited Signalling system
US11466070B2 (en) 2016-02-12 2022-10-11 Autolus Limited Signalling system
EP3211420A1 (en) 2016-02-29 2017-08-30 Miltenyi Biotec GmbH Assay for detection of chimeric antigen receptor t cells
KR20180130534A (en) * 2016-03-29 2018-12-07 유니버시티 오브 써던 캘리포니아 Chimeric antigen receptor targeting cancer
KR102584300B1 (en) 2016-03-29 2023-10-05 유니버시티 오브 써던 캘리포니아 Chimeric antigen receptor targets cancer
EP3436070A4 (en) * 2016-03-29 2019-11-27 University of Southern California Chimeric antigen receptors targeting cancer
US11446398B2 (en) 2016-04-11 2022-09-20 Obsidian Therapeutics, Inc. Regulated biocircuit systems
WO2017180587A3 (en) * 2016-04-11 2017-12-14 Obsidian Therapeutics, Inc. Regulated biocircuit systems
WO2017180993A1 (en) * 2016-04-14 2017-10-19 Bluebird Bio, Inc. Salvage chimeric antigen receptor systems
US11976116B2 (en) 2016-04-14 2024-05-07 2Seventy Bio, Inc. Salvage chimeric antigen receptor systems
WO2017216562A1 (en) * 2016-06-16 2017-12-21 Autolus Limited Tunable chimeric antigen receptors
US11472860B2 (en) * 2016-09-08 2022-10-18 Westfaelisch-Wilhelms-Universitaet Chimeric antigen receptors
WO2018087557A1 (en) 2016-11-11 2018-05-17 Autolus Limited Chimeric antigen receptor
US11440958B2 (en) 2016-11-22 2022-09-13 National University Of Singapore Blockade of CD7 expression and chimeric antigen receptors for immunotherapy of T-cell malignancies
US11945865B2 (en) 2016-11-22 2024-04-02 National University Of Singapore Blockade of CD7 expression and chimeric antigen receptors for immunotherapy of T-cell malignancies
US10550183B2 (en) 2016-11-22 2020-02-04 National University Of Singapore Blockade of CD7 expression and chimeric antigen receptors for immunotherapy of T-cell malignancies
WO2018150187A1 (en) 2017-02-17 2018-08-23 Autolus Limited Chimeric cytokines receptors
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
CN110678480A (en) * 2017-03-28 2020-01-10 细胞设计实验室股份有限公司 Chimeric polypeptides and methods for altering their localization in cell membranes
TWI811212B (en) * 2017-03-28 2023-08-11 美商細胞設計實驗公司 Chimeric polypeptides and methods of altering the membrane localization of the same
US11111310B2 (en) 2017-03-28 2021-09-07 Cell Design Labs, Inc. Chimeric polypeptides and methods of altering the membrane localization of the same
WO2018183551A1 (en) * 2017-03-28 2018-10-04 Cell Design Labs, Inc. Chimeric polypeptides and methods of altering their localisation in the cell membrane
WO2018211244A1 (en) * 2017-05-15 2018-11-22 Autolus Limited A cell comprising a chimeric antigen receptor (car)
WO2018224844A1 (en) 2017-06-09 2018-12-13 Autolus Limited Anti trbc1 antigen binding domains
US11440961B2 (en) 2017-06-09 2022-09-13 Autolus Limited Anti TRBC1 antigen binding domains
US11434293B2 (en) 2017-06-09 2022-09-06 Autolus Limited Anti TRBC1 antigen binding domains
WO2018229492A1 (en) 2017-06-15 2018-12-20 Autolus Limited Chimeric antigen receptor
US11648269B2 (en) 2017-08-10 2023-05-16 National University Of Singapore T cell receptor-deficient chimeric antigen receptor T-cells and methods of use thereof
US11701386B2 (en) 2017-09-13 2023-07-18 Autolus Limited Cell
WO2019053420A1 (en) * 2017-09-13 2019-03-21 Autolus Limited Cell
US11851672B2 (en) 2017-10-25 2023-12-26 Autolus Limited CAR T-cells comprising transcriptional circuits
WO2019122875A1 (en) * 2017-12-20 2019-06-27 Ucl Business Plc Chimeric antigen receptor
WO2019162695A1 (en) * 2018-02-26 2019-08-29 Autolus Limited Cell
WO2019215453A1 (en) 2018-05-11 2019-11-14 Autolus Limited Car-t cells expressing an inhibitory anti-tgf-beta-receptor ii single domain antibody
WO2019220109A1 (en) 2018-05-15 2019-11-21 Autolus Limited Chimeric antigen receptor
US11963981B2 (en) 2018-05-15 2024-04-23 Autolus Limited Chimeric antigen receptor
US11590170B2 (en) 2018-05-15 2023-02-28 Autolus Limited Chimeric antigen receptor
WO2020035676A1 (en) 2018-08-13 2020-02-20 Autolus Limited Car t-cells comprising an anti cd33, an anti cll1 and at least one further car anti cd123 and/or ftl3
WO2020065330A2 (en) 2018-09-27 2020-04-02 Autolus Limited Chimeric antigen receptor
US11141436B2 (en) 2019-03-05 2021-10-12 Nkarta, Inc. Immune cells engineered to express CD19-directed chimeric antigen receptors and uses thereof in immunotherapy
US11253547B2 (en) 2019-03-05 2022-02-22 Nkarta, Inc. CD19-directed chimeric antigen receptors and uses thereof in immunotherapy
US11154575B2 (en) 2019-03-05 2021-10-26 Nkarta, Inc. Cancer immunotherapy using CD19-directed chimeric antigen receptors
WO2020183131A1 (en) 2019-03-08 2020-09-17 Autolus Limited Compositions and methods comprising engineered chimeric antigen receptor and modulator of car
GB201903237D0 (en) 2019-03-08 2019-04-24 Autolus Ltd Method
WO2020222021A1 (en) 2019-05-02 2020-11-05 Autolus Limited Engineered t-cells co-expressing an anti-bcma car and an anti-ectoenzyme antibody and their use in the treatment of cancer
GB201910185D0 (en) 2019-07-16 2019-08-28 Autolus Ltd Method
WO2021009510A1 (en) 2019-07-16 2021-01-21 Autolus Limited Method for preconditioning a subject who is about to receive a t-cell therapy
WO2021023987A1 (en) 2019-08-05 2021-02-11 Autolus Limited Chimeric cytokine receptor
US11058725B2 (en) 2019-09-10 2021-07-13 Obsidian Therapeutics, Inc. CA2 compositions and methods for tunable regulation
WO2021048564A2 (en) 2019-09-13 2021-03-18 Autolus Limited Antigen-binding domain
WO2021123810A1 (en) 2019-12-20 2021-06-24 Autolus Limited Antigen-binding domain binding to psma
WO2021123800A1 (en) 2019-12-20 2021-06-24 Autolus Limited Cell
GB201919017D0 (en) 2019-12-20 2020-02-05 Autolus Ltd Cell
WO2021205173A1 (en) 2020-04-08 2021-10-14 Autolus Limited Method
WO2021205172A1 (en) 2020-04-08 2021-10-14 Autolus Limited Cell
WO2021205175A1 (en) 2020-04-09 2021-10-14 Autolus Limited Molecule
WO2021229218A1 (en) 2020-05-13 2021-11-18 Autolus Limited Method
EP3916388A1 (en) 2020-05-27 2021-12-01 Miltenyi Biotec B.V. & Co. KG Reagents for detection of chimeric antigen receptor cells
GB202017343D0 (en) 2020-11-02 2020-12-16 Autolus Ltd Cell
WO2022167798A1 (en) 2021-02-03 2022-08-11 Autolus Limited Molecule
GB202101491D0 (en) 2021-02-03 2021-03-17 Autolus Ltd Molecule
US11993794B2 (en) 2021-03-16 2024-05-28 Fred Hutchinson Cancer Center Methods to create chemically-induced dimerizing protein systems for regulation of cellular events
US11993660B2 (en) 2021-08-13 2024-05-28 Cell Design Labs, Inc. Chimeric polypeptides and methods of altering the membrane localization of the same
WO2023073352A1 (en) 2021-10-25 2023-05-04 Autolus Limited Chimeric cytokine receptor
WO2024009093A1 (en) 2022-07-06 2024-01-11 Autolus Limited Engineered trispecific tancar expressing cells targeting cd33, cd123 and cll1 and uses thereof in cancer therapy

Also Published As

Publication number Publication date
PT3126380T (en) 2021-06-30
US20200338194A1 (en) 2020-10-29
US20170014508A1 (en) 2017-01-19
PL3126380T3 (en) 2021-12-06
EP3126380B1 (en) 2021-05-05
DK3126380T3 (en) 2021-06-28
US10588967B2 (en) 2020-03-17
ES2877129T3 (en) 2021-11-16
HUE054892T2 (en) 2021-10-28
GB201405845D0 (en) 2014-05-14
EP3126380A1 (en) 2017-02-08

Similar Documents

Publication Publication Date Title
US20200338194A1 (en) Chimeric antigen receptor (car) signalling system
US20240141041A1 (en) CHIMERIC ANTIGEN RECEPTORS (CARs), COMPOSITIONS AND METHODS THEREOF
JP6821568B2 (en) Controlled elimination method of therapeutic cells
US20200223918A1 (en) CHIMERIC ANTIGEN RECEPTORS (CARs), COMPOSITIONS AND METHODS THEREOF
JP2020191870A (en) Methods for controlled activation or elimination of therapeutic cells
JP2021078514A (en) COMPOSITIONS OF CHIMERIC ANTIBODY RECEPTORS (CARs) TARGETING HEMATOLOGIC MALIGNANCIES, AND METHODS OF USE THEREOF
JP2022506598A (en) G protein-coupled receptor class C group 5 member D (GPRC5D) -specific chimeric antigen receptor
US20240165158A1 (en) Major histocompatibility complex-based chimeric receptors and uses thereof for treating autoimmune diseases
CA3111384A1 (en) Allogeneic cell compositions and methods of use
KR20190015733A (en) Chimeric antigen receptors and CAR-T cells that bind to BCMA
JP2019536480A (en) Blockade of CD7 expression and chimeric antigen receptor for immunotherapy of T cell malignancies
JP2018522907A (en) Cells for immunotherapy engineered to target the CD38 antigen and to inactivate the CD38 gene
BR112019018288A2 (en) METHODS AND COMPOSITIONS FOR TRANSDUCING AND EXPANDING LYMPHOCYTES AND REGULATING THE SAME ACTIVITY
CN116425882A (en) Chimeric proteins
JP2021515598A (en) Compositions and Methods for TCR Reprogramming with Fusion Proteins
SK15702002A3 (en) Soluble CTLA4 mutant molecules and uses thereof
US20210214439A1 (en) Chimeric antigen receptors and gene editing of cd2 for immunotherapy of t-cell malignancies
KR20210008408A (en) Blocking of CD2 surface expression and expression of chimeric antigen receptors for immunotherapy of T-cell malignancies
CN115551893A (en) Chimeric Antigen Receptors (CAR) targeting natural killer cells
US20230277622A1 (en) CHIMERIC ANTIGEN RECEPTORS (CARs) COMPOSITIONS AND METHODS THEREOF
TW202413633A (en) Engineered immune cells expressing a car and a plurality of protein expression blockers and uses thereof
CN117460522A (en) TM4SF19 inhibitor and use thereof

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: 15714272

Country of ref document: EP

Kind code of ref document: A1

REEP Request for entry into the european phase

Ref document number: 2015714272

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015714272

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 15301148

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE