EP4255936A1 - Récepteurs antigéniques chimériques ciblant bcma - Google Patents

Récepteurs antigéniques chimériques ciblant bcma

Info

Publication number
EP4255936A1
EP4255936A1 EP21901383.6A EP21901383A EP4255936A1 EP 4255936 A1 EP4255936 A1 EP 4255936A1 EP 21901383 A EP21901383 A EP 21901383A EP 4255936 A1 EP4255936 A1 EP 4255936A1
Authority
EP
European Patent Office
Prior art keywords
seq
bcma
copies
car
gdna
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
EP21901383.6A
Other languages
German (de)
English (en)
Inventor
Yihong Yao
Jiaqi Huang
Xin Yao
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Cellular Biopharmaceutical Group Ltd
Original Assignee
Shanghai Cellular Biopharmaceutical Group Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US17/476,661 external-priority patent/US11498973B2/en
Application filed by Shanghai Cellular Biopharmaceutical Group Ltd filed Critical Shanghai Cellular Biopharmaceutical Group Ltd
Publication of EP4255936A1 publication Critical patent/EP4255936A1/fr
Pending legal-status Critical Current

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464416Receptors for cytokines
    • A61K39/464417Receptors for tumor necrosis factors [TNF], e.g. lymphotoxin receptor [LTR], CD30
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/27Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by targeting or presenting multiple antigens
    • A61K2239/28Expressing multiple CARs, TCRs or antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/48Blood cells, e.g. leukemia or lymphoma
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • 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
    • 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
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Definitions

  • the invention relates to the field of biomedicine, and more particularly to chimeric antigen receptors targeting BCMA, as well as preparation methods and applications thereof.
  • BCMA is a B cell maturation antigen, also known as CD269 or TNFRSF17, and is a member of the tumor necrosis factor receptor superfamily. Its ligands are B cell activating factor (BAFF) and a proliferation-induced ligand (APRIL).
  • BAFF B cell activating factor
  • APRIL proliferation-induced ligand
  • BCMA Binding of BCMA to BAFF and APRIL activates NF-kB and induces up-regulation of anti- apoptotic Bcl-2 members such as Bcl-xL or Bcl-2 and Mcl-1.
  • Bcl-xL anti- apoptotic Bcl-2 members
  • Mcl-1 anti- apoptotic Bcl-2 members
  • BCMA BCMA-deficient mice
  • Mature B cells can normally differentiate into plasma cells in BCMA knockout mice. The BCMA knockout mice looked normal and seemed healthy, and the number of B cells was normal, but the plasma cells could not survive for a long time.
  • BCMA is also highly expressed in malignant plasma cells, such as multiple myeloma and plasma cell leukemia. BCMA is also detected in HRS cells of patients with Hodgkin's lymphoma. In America, malignant tumors of blood system account for about 10% of all malignant tumors, and myeloma accounts for 15% of all malignant hematological tumors. According to the literature, the expression of BCMA is associated with progression of multiple myeloma disease. The BCMA gene is highly expressed in myeloma samples, but is low expressed in chronic lymphocytic leukemia, acute lymphocytic leukemia, and acute T-cell lymphocytic leukemia.
  • B cell lymphomas were significantly increased in a mouse model overexpressing BCMA ligands BAFF and APRIL.
  • Ligands that bind to BCMA have been shown to regulate the growth and survival of multiple myeloma cells expressing BCMA.
  • the combination of BCMA with BAFF and APRIL can make malignant plasma cells survive. Therefore, loss of tumor cells expressing BCMA and distribution of the interaction between BCMA ligand and receptor can improve outcome in the treatment of multiple myeloma or other BCMA positive B cell lines malignant lymphoma.
  • Plasmacytoma also known as plasmacytoma or Keller's disease
  • Plasma cells are a type of leukocyte that is responsible for production of antibodies.
  • myeloma accounts for 1.8% of all tumor cases, with a mortality rate of 2.1%.
  • the statistical results of 2010-2014 show that the incidence rate is about 6.6 in 100,000 per year and the mortality rate is about 50%.
  • Multiple myeloma is a middle-aged disease. The median age of onset in Europe and the United States is 68 years old. There are more males than females.
  • the drug was also marketed in China in 2005 and has become one of the options for the treatment of multiple myeloma with thalidomide and dexamethasone.
  • the treatment of multiple myeloma is usually combined.
  • multiple drugs are used at the same time, there are also negative effects of costly and cumulative side effects.
  • T cells can be genetically modified to express a chimeric antigen receptor (CAR), which includes an antigen recognition portion and a T cell activation region.
  • CAR chimeric antigen receptor
  • This non-MHC restricted antigen recognition allows CAR- expressing T cells to recognize antigen without antigen processing, thus avoiding a major mechanism of tumor escape.
  • CAR does not produce dimers with alpha chain and beta chain of the endogenous TCR.
  • CAR-T chimeric antigen receptor T cell therapy
  • the present disclosure provides for a chimeric antigen receptor (CAR).
  • the CAR may comprise: an anti-BCMA antigen-binding region which comprises a light chain variable region (VL) and a heavy chain variable region (VH).
  • VL light chain variable region
  • VH heavy chain variable region
  • VL may comprise three complementarity determining regions (CDRs), LCDR1, LCDR2 and LCDR3; VH may comprise three CDRs, HCDR1, HCDR2 and HCDR3.
  • CDRs complementarity determining regions
  • LCDR1, LCDR2 and LCDR3 complementarity determining regions
  • VH may comprise three CDRs, HCDR1, HCDR2 and HCDR3.
  • LCDR1, LCDR2 and LCDR3 may have amino acid sequences about 80% to about 100% identical to the amino acid sequences set forth in SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, respectively.
  • HCDR1, HCDR2 and HCDR3 may have amino acid sequences about 80% to about 100% identical to the amino acid sequences set forth in SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, respectively.
  • LCDR1, LCDR2 and LCDR3 may have amino acid sequences about 80% to about 100% identical to the amino acid sequences set forth in SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, respectively.
  • HCDR1, HCDR2 and HCDR3 may have amino acid sequences about 80% to about 100% identical to the amino acid sequences set forth in SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, respectively.
  • LCDR1, LCDR2 and LCDR3 may have amino acid sequences about 80% to about 100% identical to the amino acid sequences set forth in SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, respectively.
  • HCDR1, HCDR2 and HCDR3 may have amino acid sequences about 80% to about 100% identical to the amino acid sequences set forth in SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 56, respectively.
  • VL and VH of the CAR may have amino acid sequences about 80% to about 100% identical to amino acid sequences set forth in (a) SEQ ID NO: 1 and SEQ ID NO: 2, respectively; (a) SEQ ID NO: 3 and SEQ ID NO: 4, respectively; or (a) SEQ ID NO: 5 and SEQ ID NO: 6, respectively.
  • VL is located at the N-terminus of VH.
  • the anti-BCMA antigen-binding region may be a single-chain variable fragment (scFv) that specifically binds BCMA.
  • the CAR may further comprise one or more of the following: (a) a signal peptide, (b) a hinge region, (c) a transmembrane domain, (d) a co-stimulatory region, and (e) a cytoplasmic signaling domain.
  • the co-stimulatory region may comprise a co-stimulatory region of (or may be derived from) 4-1BB (CD137), CD28, 0X40, CD2, CD7, CD27, CD30, CD40, CD70, CD134, PD1, DaplO, CDS, ICAM-1, LFA-1 (CD1 la/CD18), ICOS (CD278), NKG2D, GITR, TLR2, or combinations thereof.
  • 4-1BB CD137
  • CD28 0X40, CD2, CD7, CD27, CD30, CD40, CD70, CD134, PD1, DaplO, CDS, ICAM-1, LFA-1 (CD1 la/CD18), ICOS (CD278), NKG2D, GITR, TLR2, or combinations thereof.
  • the cytoplasmic signaling domain may comprise a cytoplasmic signaling domain of (or may be derived from) CD3 ⁇ .
  • the hinge region may comprise a hinge region of (or may be derived from) CD8, CD28, CD137, Ig4, or combinations thereof.
  • the transmembrane domain may comprise a transmembrane domain of (or may be derived from) CD8, CD28, CD3 ⁇ , CD45, CD4, CD5, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154, or combinations thereof.
  • the present disclosure provides for an immune cell expressing the present CAR.
  • the immune cell may be a T cell or a natural killer (NK) cell.
  • the immune cell may be allogeneic or autologous.
  • nucleic acid encoding the present CAR.
  • the present disclosure further provides for a vector comprising the present nucleic acid.
  • the present disclosure provides for a method of treating cancer.
  • the method may comprise administering the immune cell to a subject in need thereof.
  • the cancer may be a hematologic cancer.
  • the cancer may be a plasma-cell malignancy.
  • the cancer may be a BCMA-positive malignancy.
  • the cancer may be multiple myeloma (MM), or plasma cell leukemia.
  • the immune cell may be administered by infusion, injection, transfusion, implantation, and/or transplantation.
  • the immune cell may be administered intravenously, subcutaneously, intradermally, intranodally, intratumorally, intramedullary, intramuscularly, or intraperitoneally.
  • the immune cell may be administered via intravenous infusion.
  • the subject may be a human.
  • the present disclosure provides for a method for treating cancer.
  • the method may comprise administering the present immune cell to a subject in need of.
  • the chimeric antigen receptor (CAR) may generate an area under the curve (AUC) ranging from about 5.0e+05 copies/ ⁇ g genomic DNA (copies/gDNA) to about 1.3e+07 copies/gDNA, from about 5.0e+06 copies/ ⁇ g genomic DNA (copies/gDNA) to about 1.0e+07 copies/gDNA, from about 5.0e+06 copies/ ⁇ g genomic DNA (copies/gDNA) to about 1.3e+07 copies/gDNA, or from about 7.0e+06 copies/ ⁇ g genomic DNA (copies/gDNA) to about 1.0e+07 copies/gDNA, in the blood of the subject in about 28 days after administration.
  • AUC area under the curve
  • the chimeric antigen receptor (CAR) may generate a maximum plasma concentration (Cmax) ranging from about 5x10 4 copies/ ⁇ g genomic DNA (copies/gDNA) to about 1.3x10 6 copies/gDNA, from about 5x10 5 copies/ ⁇ g genomic DNA (copies/gDNA) to about 1.3x10 6 copies/gDNA, or from about 7.5x10 5 copies/ ⁇ g genomic DNA (copies/gDNA) to about IxlO 6 copies/gDNA, in the blood of the subject.
  • Cmax maximum plasma concentration
  • the CAR may have a T m ax ranging from about 12 days to about 25 days, from about 14 days to about 20 days, or from about 6 days to about 22 days.
  • the anti-BCMA antigen-binding region includes a light chain variable region (VL) comprising an amino acid sequence at least or about 70%, at least or about 75%, at least or about 80%, at least or about 85%, at least or about 90%, at least or about 95%, at least or about 99%, at least or about 81%, at least or about 82%, at least or about 83%, at least or about 84%, at least or about 85%, at least or about 86%, at least or about 87%, at least or about 88%, at least or about 89%, at least or about 90%, at least or about 91%, at least or about 92%, at least or about 93%, at least or about 94%, at least or about 95%, at least or about 96%, at least or about 97%, at least or about 98%, at least or about 99%, or about 100% identical to the amino acid sequence set forth in SEQ ID NO: 1, SEQ ID NO: 3, or SEQ ID NO: 5.
  • VL light chain variable region
  • the anti-BCMA antigen-binding region includes a heavy chain variable region (VH) comprising an amino acid sequence at least or about 70%, at least or about 75%, at least or about 80%, at least or about 85%, at least or about 90%, at least or about 95%, at least or about 99%, at least or about 81%, at least or about 82%, at least or about 83%, at least or about 84%, at least or about 85%, at least or about 86%, at least or about 87%, at least or about 88%, at least or about 89%, at least or about 90%, at least or about 91%, at least or about 92%, at least or about 93%, at least or about 94%, at least or about 95%, at least or about 96%, at least or about 97%, at least or about 98%, at least or about 99%, or about 100% identical to the amino acid sequence set forth in SEQ ID NO: 2, SEQ ID NO: 4, or SEQ ID NO: 6.
  • VH heavy chain variable region
  • a light chain variable region of the anti-BCMA antigen-binding region can comprise one, two, or three complementarity determining regions (CDRs) that are at least or about 70%, at least or about 75%, at least or about 80%, at least or about 85%, at least or about 90%, at least or about 95%, at least or about 99%, at least or about 81%, at least or about 82%, at least or about 83%, at least or about 84%, at least or about 85%, at least or about 86%, at least or about 87%, at least or about 88%, at least or about 89%, at least or about 90%, at least or about 91%, at least or about 92%, at least or about 93%, at least or about 94%, at least or about 95%, at least or about 96%, at least or about 97%, at least or about 98%, at least or about 99%, or about 100%, identical to the CDRs of a light chain variable region of the BCMA-20 antibody (CDR1, CDR2 and CDR3 as set forth
  • a heavy chain variable region of the anti-BCMA antigen-binding region can comprise one, two, or three complementarity determining regions (CDRs) that are at least or about 70%, at least or about 75%, at least or about 80%, at least or about 85%, at least or about 90%, at least or about 95%, at least or about 99%, at least or about 81%, at least or about 82%, at least or about 83%, at least or about 84%, at least or about 85%, at least or about 86%, at least or about 87%, at least or about 88%, at least or about 89%, at least or about 90%, at least or about 91%, at least or about 92%, at least or about 93%, at least or about 94%, at least or about 95%, at least or about 96%, at least or about 97%, at least or about 98%, at least or about 99%, or about 100%, identical to the CDRs of a heavy chain variable region of the BCMA-20 antibody (CDR1, CDR2 and CDR3 as set forth
  • a light chain variable region of the anti-BCMA antigen-binding region can comprise one, two, or three complementarity determining regions (CDRs) that are at least or about 70%, at least or about 75%, at least or about 80%, at least or about 85%, at least or about 90%, at least or about 95%, at least or about 99%, at least or about 81%, at least or about 82%, at least or about 83%, at least or about 84%, at least or about 85%, at least or about 86%, at least or about 87%, at least or about 88%, at least or about 89%, at least or about 90%, at least or about 91%, at least or about 92%, at least or about 93%, at least or about 94%, at least or about 95%, at least or about 96%, at least or about 97%, at least or about 98%, at least or about 99%, or about 100%, identical to the CDRs of a light chain variable region of the BCMA-20 antibody (CDR1, CDR2 and CDR3 as set forth
  • a heavy chain variable region of the anti-BCMA antigen-binding region can comprise one, two, or three complementarity determining regions (CDRs) that are at least or about 70%, at least or about 75%, at least or about 80%, at least or about 85%, at least or about 90%, at least or about 95%, at least or about 99%, at least or about 81%, at least or about 82%, at least or about 83%, at least or about 84%, at least or about 85%, at least or about 86%, at least or about 87%, at least or about 88%, at least or about 89%, at least or about 90%, at least or about 91%, at least or about 92%, at least or about 93%, at least or about 94%, at least or about 95%, at least or about 96%, at least or about 97%, at least or about 98%, at least or about 99%, or about 100%, identical to the CDRs of a heavy chain variable region of the BCMA-20 antibody (CDR1, CDR2 and CDR3 as set forth
  • a light chain variable region of the anti-BCMA antigen-binding region includes three CDRs that are identical (e.g., 80% - 100% identical) to CDRs of a heavy chain variable region of the BCMA-20 antibody (CDR1, CDR2 and CDR3 as set forth in position 24-34, position 50-56, position 89-97 of SEQ ID NO: 1), and a heavy chain variable region of the anti-BCMA antigen-binding region includes three CDRs that are identical (e.g., 80% - 100% identical) to CDRs of a heavy chain variable region of the BCMA-20 antibody (CDR1, CDR2 and CDR3 as set forth in position 31-35, position 50-66, position 99-110 of SEQ ID NO: 2, respectively).
  • a light chain variable region of the anti-BCMA antigen-binding region includes three CDRs that are identical (e.g., 80% - 100% identical) to the CDRs of a light chain variable region of the BCMA-20 antibody (CDR1, CDR2 and CDR3 as set forth in SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, respectively), and a heavy chain variable region of the anti-BCMA antigen-binding region includes three CDRs that are identical (e.g., 80% - 100% identical) to the CDRs of a heavy chain variable region of the BCMA-20 antibody (CDR1, CDR2 and CDR3 as set forth in SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, respectively).
  • a light chain variable region of the anti-BCMA antigen-binding region includes three CDRs that are identical (e.g., 80% - 100% identical) to CDRs of a light chain variable region of the BCMA-CA8 antibody (CDR1, CDR2 and CDR3 as set forth in position 24- 34, position 50-56, position 89-97 of SEQ ID NO: 3, respectively), and a heavy chain variable region of the anti-BCMA antigen-binding region includes three CDRs that are identical (e.g., 80% - 100% identical) to CDRs of a heavy chain variable region of the BCMA-CA8 antibody (CDR1, CDR2 and CDR3 as set forth in position 31-35, position 50-66, position 99-110 of SEQ ID NO: 4, respectively).
  • a light chain variable region of the anti-BCMA antigen-binding region includes three CDRs that are identical (e.g., 80% - 100% identical) to the CDRs of a light chain variable region of the BCMA-CA8 antibody (CDR1, CDR2 and CDR3 as set forth in SEQ ID NO: 31, SEQ ID NO: 33, SEQ ID NO: 35, respectively), and a heavy chain variable region of the anti-BCMA antigen-binding region includes three CDRs that are identical (e.g., 80% - 100% identical) to the CDRs of a heavy chain variable region of the BCMA-CA8 antibody (CDR1, CDR2 and CDR3 as set forth in SEQ ID NO: 38, SEQ ID NO: 40, SEQ ID NO: 42, respectively).
  • a light chain variable region of the anti-BCMA antigen-binding region includes three CDRs that are identical to CDRs of a light chain variable region of the BCMA-M06 antibody (CDR1, CDR2 and CDR3 as set forth in position 24-34, position 50-56, position 89-97 of SEQ ID NO: 5, respectively), and a heavy chain variable region of the anti- BCMA antigen-binding region includes three CDRs that are identical to CDRs of a heavy chain variable region of the BCMA-MO6 antibody (CDR1, CDR2 and CDR3 as set forth in position 31- 35, position 50-66, position 99-110 of SEQ ID NO: 6, respectively).
  • a light chain variable region of the anti-BCMA antigen-binding region includes three CDRs that are identical (e.g., 80% - 100% identical) to the CDRs of a light chain variable region of the BCMA-M06 antibody (CDR1, CDR2 and CDR3 as set forth in SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, respectively), and a heavy chain variable region of the anti-BCMA antigen-binding region includes three CDRs that are identical (e.g., 80% - 100% identical) to the CDRs of a heavy chain variable region of the BCMA-M06 antibody (CDR1, CDR2 and CDR3 as set forth in SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 56, respectively).
  • the CAR may comprise an amino acid sequence about 80% to about 100%, about 85% to about 100%, about 90% to about 100%, about 95% to about 100%, at least or about 70%, at least or about 75%, at least or about 80%, at least or about 85%, at least or about 90%, at least or about 95%, at least or about 99%, at least or about 81%, at least or about 82%, at least or about 83%, at least or about 84%, at least or about 85%, at least or about 86%, at least or about 87%, at least or about 88%, at least or about 89%, at least or about 90%, at least or about 91%, at least or about 92%, at least or about 93%, at least or about 94%, at least or about 95%, at least or about 96%, at least or about 97%, at least or about 98%, at least or about 99%, or about 100%, identical to the amino acid sequence set forth in SEQ ID NO: 59, SEQ ID NO: 61, or SEQ ID NO:
  • the nucleic acid encoding the CAR may comprise a nucleic acid sequence about 80% to about 100%, about 85% to about 100%, about 90% to about 100%, about 95% to about 100%, at least or about 70%, at least or about 75%, at least or about 80%, at least or about 85%, at least or about 90%, at least or about 95%, at least or about 99%, at least or about 81%, at least or about 82%, at least or about 83%, at least or about 84%, at least or about 85%, at least or about 86%, at least or about 87%, at least or about 88%, at least or about 89%, at least or about 90%, at least or about 91%, at least or about 92%, at least or about 93%, at least or about 94%, at least or about 95%, at least or about 96%, at least or about 97%, at least or about 98%, at least or about 99%, or about 100%, identical to the amino acid sequence set forth in SEQ ID NO: 58, SEQ ID NO: 58
  • the CAR may generate an area under the curve (AUC) ranging from about 0.5e+06 copies/ ⁇ g genomic DNA (copies/gDNA) to about 2e+07 copies/gDNA, from about 5.0e+05 copies/ ⁇ g genomic DNA (copies/gDNA) to about 1.3e+07 copies/gDNA, from about 5.0e+05 copies/ ⁇ g genomic DNA (copies/gDNA) to about 2e+07 copies/gDNA, from about
  • AUC area under the curve
  • the CAR generates a maximum plasma concentration (Cmax) ranging from about 5x10 4 copies/ ⁇ g genomic DNA (copies/gDNA) to about 1.3x10 6 copies/gDNA, from about 5x10 4 copies/ ⁇ g genomic DNA (copies/gDNA) to about 1.5x10 6 copies/gDNA, from about 5x10 1 copies/ ⁇ g genomic DNA (copies/gDNA) to about 1.3x10 6 copies/gDNA, from about 7.5x10 5 copies/ ⁇ g genomic DNA (copies/gDNA) to about IxlO 6 copies/gDNA, from about 7x10 5 copies/ ⁇ g genomic DNA (copies/gDNA) to about IxlO 6 copies/gDNA, from about 8x10 5 copies/ ⁇ g genomic DNA (copies/gDNA) to about IxlO 6 copies/gDNA, from about 7.5x10 5 copies/pg genomic DNA (copies/gDNA) to about 1.5x10 6 copies/gDNA, from about 7x10 5 copies/pg genomic DNA (copies/gDNA) to about
  • the CAR has a T m ax (the time it takes the CAR to reach Cmax) ranging from about 12 days to about 25 days, from about 14 days to about 20 days, from about 6 days to about 22 days, from about 3 days to about 20 days, from about 4 days to about 18 days, from about 5 days to about 17 days, from about 6 days to about 16 days, from about 7 days to about 15 days, from about 9 days to about 15 days, from about 10 days to about 15 days, from about 10 days to about 14 days, from about 8 days to about 12 days, from about 6 days to about 14 days, from about 12 days to about 14 days, from about 8 days to about 11 days, from about 8 days to about 15 days, or from about 10 days to about 14 days.
  • the T m ax may be a median T m ax.
  • the CAR has a Tiast (the time corresponding to the last quantifiable CAR level) ranging from about 30 days to about 200 days, from about 50 days to about 150 days, from about 50 days to about 100 days, from about 60 days to about 80 days, from about 60 days to about 150 days, from about 80 days to about 150 days, from about 50 days to about 200 days, from about 50 days to about 60 days, from about 50 days to about 80 days, from about 50 days to about 100 days, from about 60 days to about 100 days, from about 80 days to about 100 days, from about 60 days to about 200 days, from about 80 days to about 200 days, from about 50 days to about 140 days, from about 60 days to about 140 days, or from about 80 days to about 140 days.
  • the Tiast may be a median Tiast.
  • BCMA targeted chimeric antigen receptor as well as preparation method and activity identification of a modified T cell (CART-BCMA) thereof.
  • the present disclosure provides a chimeric antigen receptor structure for use in the treatment of BCMA positive B cell lymphoma.
  • a chimeric antigen receptor (CAR) sequence
  • its antigen binding domain is an antibody single chain variable region sequence that targets extracellular region of BCMA.
  • the antigen binding domain is an antibody single chain variable region sequence that targets amino acid residues at positions 24 to 41 of the BCMA sequence.
  • NCBI accession number of the BCMA sequence is AY684975.1.
  • the structure of the antigen binding domain is shown in formula I as below: wherein VH is an antibody heavy chain variable region; VL is an antibody light chain variable region; and is a linker peptide or a peptide bond; and, the amino acid sequence of VL is as shown in SEQ ID NO: 1, and the amino acid sequence of V H is as shown in SEQ ID NO: 2; or, the amino acid sequence of VL is as shown in SEQ ID NO: 3, and the amino acid sequence of V H is as shown in SEQ ID NO: 4; or, the amino acid sequence of VL is as shown in SEQ ID NO: 5, and the amino acid sequence of V H is as shown in SEQ ID NO: 6.
  • amino acid sequence of the linker peptide is as shown in SEQ ID NO: 10 or SEQ ID NO: 11.
  • the antibody single chain variable region comprises a human, mouse, human-mouse chimeric antibody single chain variable region.
  • S is an optional signal peptide
  • H is a hinge region
  • TM is a transmembrane domain
  • C is a co- stimulatory signaling molecule
  • CD3 ⁇ is a cytoplasmic signaling sequence derived from CD3 ⁇ ;
  • V H and V L are as described above.
  • the S is a signal peptide of a protein selected from the group consisting of CD8, CD28, GM-CSF, CD4, CD137, or a combination thereof.
  • the S is a signal peptide derived from CD8.
  • amino acid sequence of S is as shown in SEQ ID NO: 9.
  • the H is a hinge region of a protein selected from the group consisting of CD8, CD28, CD137, or a combination thereof.
  • the H is a hinge region derived from CD8.
  • amino acid sequence of H is as shown in SEQ ID NO: 12.
  • the TM is a transmembrane region of a protein selected from the group consisting of CD28, CD3 ⁇ , CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154, or a combination thereof.
  • the TM is a transmembrane region derived from CD8.
  • sequence of TM is as shown in SEQ ID NO: 13.
  • the C is a co-stimulatory signaling molecule of a protein selected from the group consisting of 0X40, CD2, CD7, CD27, CD28, CD30, CD40, CD70, CD134, 4- 1BB (CD137), PD1, DaplO, CDS, ICAM-1, LFA-1 (CDl la/CD18), ICOS (CD278), NKG2D, GITR, TLR2, or a combination thereof.
  • a protein selected from the group consisting of 0X40, CD2, CD7, CD27, CD28, CD30, CD40, CD70, CD134, 4- 1BB (CD137), PD1, DaplO, CDS, ICAM-1, LFA-1 (CDl la/CD18), ICOS (CD278), NKG2D, GITR, TLR2, or a combination thereof.
  • C is a co-stimulatory signaling molecule derived from 4- IBB.
  • amino acid sequence of C is as shown in SEQ ID NO: 14.
  • amino acid sequence of CD3 ⁇ is as shown in SEQ ID NO: 15.
  • a nucleic acid molecule encoding the chimeric antigen receptor (CAR) of the first aspect of.
  • the nucleic acid molecule is isolated. In a third aspect, it provides a vector, comprising the nucleic acid molecule of the second aspect.
  • the vector is selected from the group consisting of DNA, RNA, plasmid, lentiviral vector, adenoviral vector, retroviral vector, transposon, or a combination thereof.
  • the vector is a lentiviral vector.
  • a host cell comprising the vector of the third aspect or having the exogenous nucleic acid molecule of the second aspect integrated into the chromosome or expressing the CAR of the first aspect.
  • the cell is an isolated cell, and/or the cell is a genetically engineered cell.
  • the cell is a mammalian cell.
  • the cell is a T cell.
  • a fifth aspect it provides a method for preparing a CAR-T cell expressing the CAR of the first aspect, and the method comprises the steps of: transducing the nucleic acid molecule of the second aspect or the vector of the third aspect into a T cell, thereby obtaining the CAR-T cell.
  • a preparation comprising the chimeric antigen receptor of the first aspect, the nucleic acid molecule of the second aspect, the vector of the third aspect, or the cell of the fourth aspect, and a pharmaceutically acceptable carrier, diluent or excipient.
  • the preparation is a liquid preparation.
  • the dosage form of the preparation is injection.
  • the concentration of the CAR-T cells in the preparation is 1 x 10 3 - IxlO 8 cells / ml, or 1 x 10 4 -lx10 7 cells / ml.
  • a seventh aspect it provides the use of the chimeric antigen receptor of the first aspect, the nucleic acid molecule of the second aspect, the vector of the third aspect, or the cell of the fourth aspect, for the preparation of a medicine or a preparation for preventing and/or treating tumor or cancer.
  • the tumor is selected from the group consisting of a hematological tumor, a solid tumor, or a combination thereof.
  • the cancer is B cell lymphoma.
  • the blood tumor is selected from the group consisting of acute myeloid leukemia (AML), multiple myeloma (MM), chronic lymphocytic leukemia (CLL), acute lymphoblastic leukemia (ALL), diffuse large B cell lymphoma (DLBCL), or a combination thereof.
  • AML acute myeloid leukemia
  • MM multiple myeloma
  • CLL chronic lymphocytic leukemia
  • ALL acute lymphoblastic leukemia
  • DLBCL diffuse large B cell lymphoma
  • the solid tumor is selected from the group consisting of gastric cancer, peritoneal metastasis of gastric cancer, liver cancer, leukemia, renal cancer, lung cancer, small intestine cancer, bone cancer, prostate cancer, colorectal cancer, breast cancer, large intestine cancer, cervical cancer, ovarian cancer, lymphoma, nasopharyngeal carcinoma, adrenal tumor, bladder tumor, non-small cell lung cancer (NSCLC), glioma, endometrial cancer, or a combination thereof.
  • gastric cancer peritoneal metastasis of gastric cancer
  • liver cancer leukemia, renal cancer, lung cancer, small intestine cancer, bone cancer, prostate cancer, colorectal cancer, breast cancer, large intestine cancer, cervical cancer, ovarian cancer, lymphoma, nasopharyngeal carcinoma, adrenal tumor, bladder tumor, non-small cell lung cancer (NSCLC), glioma, endometrial cancer, or a combination thereof.
  • NSCLC non-small cell lung
  • the tumor is a BCMA positive tumor, such as a BCMA positive B cell lymphoma, multiple myeloma, or plasma cell leukemia.
  • kits for the preparation of the cell of the fourth aspect comprises a container, and the nucleic acid molecule of the second aspect or the vector of the third aspect is located in the container.
  • a ninth aspect it provides a use of the cell of the fourth aspect, or the preparation of the sixth aspect for the prevention and/or treatment of cancer or tumor.
  • a tenth aspect provides a method of treating a disease comprising administering an appropriate amount of the cell of the fourth aspect, or the preparation of the sixth aspect, to a subject in need of treatment.
  • the disease is cancer or tumor.
  • FIG. 1A shows detection of transfection efficiency of engineered T cell with chimeric antigen receptors targeting BCMA.
  • the expression level of the CAR gene-encoded protein on the surface of the T cell membrane in CAR-BCMA cells cultured on day 6 was identified by Fc fragment staining method of recombinant human BCMA protein.
  • l* 10 5 of CART-BCMA cells cultured on day 10 were cultured respectively with BCMA-positive K562-BCMA-B9 tumor cell line, MM.
  • FIG. 2 shows structure of chimeric antigen receptor targeting BCMA.
  • the structure of CAR includes a leader sequence, an antigen recognition sequence, a linker region, a transmembrane region, a co-stimulatory factor signal region, and a CD3 ⁇ signaling region.
  • Figures 3 A, 3B, and 3C show detection of transfection efficiency of engineered T cell with chimeric antigen receptors targeting BCMA.
  • the expression levels of the CAR gene-encoded protein on the surface of the T cell membrane in CAR-BCMA cells cultured on day 7 (FIG. 3A) day 21 (FIG. 3B) and day 29 (FIG. 3C) were identified by Fc fragment staining method of recombinant human BCMA protein.
  • Figures 4A and 4B show the expression level of CD137 on the surface of T cell membrane (FIG. 4A) and the secretion level of IFNY i n the culture supernatant (FIG. 4B).
  • IxlO 5 of CART-BCMA cells cultured on day 7 were cultured respectively with BCMA-positive K562- BCMA-E7 tumor cell line and BCMA-negative K562 tumor cell line, or without tumor cells, in 200 pl GT-551 medium for 18h at a ratio of 1:1. Then the expression level of CD 137 on the surface of T cell membrane and the secretion level of IFNy in the culture supernatant was detected respectively.
  • Figure 5 shows detection of advanced apoptosis level of tumor cells induced by CART- BCMAs.
  • IxlO 4 CFSE-labeled BCMA-negative (NH929) or BCMA-positive (NH929-BCMA) tumor cell lines were co-cultured respectively with corresponding T cells in 100 pl of GT-551 medium for 4 h at a ratio as shown in the figure.
  • the proportion of Pl-positive cells in CFSE-positive cells was analyzed by flow cytometry after staining with 100 pl of 25% PI dye for 15 min.
  • the figure shows the statistical analysis of PI positive cells in the corresponding coculture samples.
  • FIG. 6A and 6B show detection of advanced apoptosis level of tumor cells induced by CART-BCMAs.
  • FIG. 6A shows ratio of CART positive cells in the analyzed samples, wherein NT and BCMA-M06 were calculated at 60%.
  • 1 x 10 4 CFSE-labeled BCMA-negative (NH929) or BCMA-positive (NH929-BCMA, MM. IS) tumor cell lines were co-cultured respectively with corresponding T cells in 100 pl of GT-551 medium for 4 h at a ratio as shown in the figure.
  • the proportion of Pl-positive cells in CFSE-positive cells was analyzed by flow cytometry after staining with 100 pl of 25% PI dye for 15 min.
  • FIG. 6B shows the statistical analysis of PI positive cells in the corresponding co-culture samples.
  • Figures 7A and 7B show the inhibitory effect of CART-BCMAs on the proliferation in vivo of myeloma cell line RPMI-8226 in B-NDG mice.
  • RPMI-8226 cells in logarithmic growth phase were collected, and 4.0x10 6 tumor cells were inoculated subcutaneously in the right back of mice.
  • the tumor volume reached about 120 mm 3
  • the animals were randomly divided into 4 groups according to tumor volume. Then solvent control, 7.5x10 6 NT (T cells only) and 7.5x10 6 CART- BCMAs cells were injected through tail vein respectively.
  • FIG. 7A shows that single injection of CART-BCMA-1 and CART-BCMA-20 through tail vein can effectively inhibit the growth of human myeloma RPMI-8226 cells.
  • FIG. 7B shows that CART-BCMA-1 and CART-BCMA-20 can significantly prolong the survival of tumor-bearing mice with human myeloma RPMI-8226 cells.
  • Figure 8 shows results the expression of BCMA, CR2, CXADR, DDR2, and MAG in 293T cells after transfection of relevant plasmids by flow cytometry.
  • Figures 9 A, 9B, and 9C show the cytokine detection results.
  • Figure 10 shows the killing ability of each cell to the target cell.
  • Figure 11 shows the effect of soluble BCMA on cell killing activity.
  • Figure 12 shows the survival rate of the mice in each group.
  • Figure 13 shows the experimental process of phase I clinical study.
  • Figure 14 shows the clinical response of phase I clinical study.
  • Figure 15 shows the treatment condition of the patient of ID Z0203-00801C008.
  • Figures 16A and 16B show the treatment condition of patient of ID Z0203 -00701 COO 1.
  • Figure 17 shows the expansion of C-CARO88 and the decrease of M-protein/sFLC levels in the blood.
  • Figures 18A-18C show the results of evaluating the binding specificity of the scFv of C- CARO88.
  • Figure 18A shows the experimental scheme.
  • Figure 18B shows the structure of the anti- BCMA CAR and scFv rabFc.
  • Figure 18C shows the experimental results.
  • Figures 19A-19B show tissue cross reactivity IHC GLP study and validation.
  • HAdCC human primary adrenal cortical cells.
  • HPTEC human primary thyroid epithelial cells.
  • A549 BCMA OE a stable strain overexpressing BCMA.
  • Figures 20 shows an example of the CAR (C-CARO88) manufacture process.
  • the process includes the usage of serum free media, a closed, modular integrated, semi-automated system, and digital monitoring. Stars indicate improved processes.
  • Median vein to vein time is about 17 days (range: 13 to 83 days).
  • Median manufacturing time is about 7 days (range: 5 to 10 days).
  • FIG. 21 shows C-CARO88 phase I clinical study design (in treating relapsed or refractory multiple myeloma). Phase I, open-label, dose escalation and expansion studies were conducted at four medical centers.
  • C-CARO88 is an embodiment of the present CAR which is based on the BCMA-20 antibody.
  • Figure 22 shows the C-CARO88 clinical response, including SD, MR, PR, CR, VGPR, MRD, and PD.
  • SD stable disease
  • PR partial response
  • CR complete response
  • PD progressive disease
  • MR minimal response
  • VGPR very good partial response
  • MRD minimal residual disease.
  • Figure 23 shows the C-CARO88 clinical response, including CR/sCR, VGPR and PR.
  • Figure 24 shows the Kaplan Meyer estimation of progression-free survival (PFS) for mid and high dose group.
  • Figure 25A shows the expansion of C-CARO88 in the blood of the patients after CAR administration.
  • Figure 25B shows the expansion of C-CARO88 in the blood of the patients up to the most recent visit.
  • Figure 25C shows Cmax levels in the blood of the patients after CAR administration.
  • Figure 25D shows AUC levels in the blood of the patients after CAR administration.
  • Figure 25E shows T m ax levels in the blood of the patients after CAR administration.
  • Figure 25F shows Tiast levels in the blood of the patients after CAR administration.
  • the present disclosure provides for chimeric antigen receptors (CARs) targeting BCMA.
  • the CARs are based on four monoclonal antibodies: BCMA-1, BCMA-20, BCMA-CA8, and BCMA-MO6.
  • the present disclosure also provides for the analysis and identification of the expression levels of the CARs in primary T cells, in vitro activation ability and tumor cell killing efficacy of these chimeric antigen receptors. Studies have shown that the chimeric antigen receptors of the present disclosure target BCMA positive cells and can be used to treat BCMA positive B cell lymphoma, multiple myeloma, plasma cell leukemia or other diseases.
  • the present disclosure identifies the correlation between the expression time and the expression intensity of different CAR structures on the surface of the cell membrane after virus infection, and further identifies the difference in expression of different CAR structural proteins. This finding suggests that different CAR structures exist a difference in the expression level of CAR protein on the membrane surface and the persistence of CART in vivo activity under same infection condition. After extensive screening, the CAR structure of the present disclosure was obtained. The results show that the protein encoded by the CAR structure of the present disclosure can be fully expressed and membrane-localized.
  • the preparation process of CAR-modified T cell targeting BCMA antigen is improved.
  • GT-551 serum-free medium supplemented with 1% human albumin is used to culture lymphocytes in vitro.
  • the term “about” may refer to a value or composition within an acceptable error range for a particular value or composition as determined by those skilled in the art, which will depend in part on how the value or composition is measured or determined.
  • the term “about” in reference to a numeric value may refer to ⁇ 10% of the stated numeric value. In other words, the numeric value can be in a range of 90% of the stated value to 110% of the stated value.
  • administering refers to the physical introduction of a product of the disclosure into a subject using any one of various methods and delivery systems known to those skilled in the art, including, but not limited to, intravenous, intramuscular, subcutaneous, intraperitoneal, spinal or other parenteral administration, such as by injection or infusion.
  • antibody may include, but is not limited to, an immunoglobulin that specifically binds an antigen and contains at least two heavy (H) chains and two light (L) chains linked by disulfide bonds, or an antigen binding parts thereof.
  • H chain contains a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
  • the heavy chain constant region contains three constant domains, CHI, CH2, and CH3.
  • Each light chain contains a light chain variable region (abbreviated herein as VL) and a light chain constant region.
  • the light chain constant region contains a constant domain CL.
  • VH and VL regions can be further subdivided into hypervariable regions called complementarity determining regions (CDR), which are interspersed within more conservative regions called framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each VH and VL contains three CDRs and four FRs, which are arranged from amino terminal to carboxy terminal in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the chimeric antigen receptors (CARs) of the present disclosure may comprise an extracellular domain, a transmembrane domain, and an intracellular domain.
  • the extracellular domain comprises a target- specific binding element (also known as an antigen binding domain).
  • the intracellular domain includes a co-stimulatory signaling region and a C, chain.
  • the costimulatory signaling region refers to a part of the intracellular domain that includes a costimulatory molecule.
  • the co-stimulatory molecule is a cell surface molecule required for efficient response of lymphocytes to antigens, rather than an antigen receptor or its ligand.
  • a linker can be incorporated between the extracellular domain and the transmembrane domain of the CAR, or between the cytoplasmic domain and the transmembrane domain of the CAR.
  • the term "linker” generally refers to any oligopeptide or polypeptide that plays a role of linking the transmembrane domain to the extracellular domain or the cytoplasmic domain in a polypeptide chain.
  • the linker may comprise 0-300 amino acids, 2-100 amino acids, or 3-50 amino acids.
  • the extracellular domain of the CAR comprises an antigen binding domain targeting BCMA.
  • antigen recognition can be performed based on antigen binding specificity.
  • the antigen binding domain may be fused to the intracellular domain from one or more of the co-stimulatory molecules and the C, chain.
  • the antigen binding domain may be fused with an intracellular domain of a combination of a 4-1BB signaling domain and a CD3 ⁇ signaling domain.
  • the “antigen binding domain” and “single-chain antibody fragment” may refer to a Fab fragment, a Fab’ fragment, a F (ab’) 2 fragment, or a single Fv fragment that has antigenbinding activity.
  • the Fv antibody contains the heavy chain variable region and the light chain variable region of the antibody, but has no constant region.
  • the Fv antibody has the smallest antibody fragment with all antigen-binding sites.
  • Fv antibodies also include a polypeptide linker between the VH and VL domains, and can form the structure required for antigen binding.
  • the antigen binding domain is usually a scFv (single-chain variable fragment). The size of scFv is typically 1/6 of a complete antibody.
  • the single-chain antibody may be an amino acid chain sequence encoded by a nucleotide chain.
  • the scFv may comprise an antibody which specifically recognizes the extracellular region of BCMA, such as amino acid residues at positions 24 to 41 of the BCMA sequence.
  • the antibody may be a single chain antibody.
  • the CAR can be designed to comprise a transmembrane domain fused to the extracellular domain of the CAR.
  • a transmembrane domain that is naturally associated with one of the domains in the CAR is used.
  • transmembrane domains may be selected or modified by amino acid substitutions to avoid binding such domains to the transmembrane domain of the same or different surface membrane proteins, thereby minimizing the interaction with other members of the receptor complexes.
  • the intracellular domain in the CAR comprises the signaling domain of 4- IBB and the signaling domain of CD3 ⁇ .
  • the CAR structure of the present disclosure comprises a signal peptide, an antigen recognition sequence (antigen-binding domain), a linker region, a transmembrane region, a co-stimulatory factor signal region, and a CD3zeta signaling region (£ chain portion).
  • the order of connection is as follows:
  • the signal peptide is a signal peptide sequence derived from CD8:
  • MALPVTALLLPLALLLHAARP (SEQ ID NO: 9)
  • VL light chain sequence of single-chain variable region derived from BCMA-1 antibody: NO: 7
  • VH heavy chain sequence of single-chain variable region derived from BCMA-1 antibody
  • BCMA-1 is an antibody sequence contained in a published Car-T sequence, and is used as a control in the present application.
  • VL light chain
  • VH heavy chain sequence of single-chain variable region derived from BCMA-20 antibody
  • VL light chain sequence of single-chain variable region derived from BCMA-CA8 antibody
  • VH heavy chain sequence of single-chain variable region derived from BCMA-CA8 antibody
  • VL light chain sequence of single-chain variable region derived from BCMA-MO6 antibody: NO: 5
  • the transmembrane region is a transmembrane region sequence of CD8 (CD8TM) antigen:
  • the co- stimulatory factor signal region is derived from the sequence of 4- IBB cytoplasmic signaling motif:
  • the signaling region of CD3 zeta is derived from the sequence of immunorecceptor tyrosine-based activation motif (FT AM) of CD3zeta in the TCR complex:
  • the nucleic acid encoding the CAR (derived from the BCMA-20 antibody) may have the following sequence (SEQ ID NO: 58):
  • the CAR (derived from the BCMA-20 antibody) may have the following amino acid sequence (SEQ ID NO: 59):
  • the nucleic acid encoding the CAR may have the following sequence (SEQ ID NO: 60):
  • the CAR (derived from the BCMA-CA8 antibody) may have the following amino acid sequence (SEQ ID NO: 61): P
  • the nucleic acid encoding the CAR may have the following sequence (SEQ ID NO: 62):
  • the CAR (derived from the BCMA-BCMA-MO6 antibody) may have the following amino acid sequence (SEQ ID NO: 63):
  • CAR-T cell As used herein, the terms “CAR-T cell”, “CAR-T”, and “CART”, may be used interchangeably.
  • the present disclosure relates to the construction of a chimeric antigen receptor structure targeting BCMA, a preparation method of a chimeric antigen receptor engineered T cell targeting BCMA, and activity identification thereof.
  • nucleic acid sequences coding for the desired molecules can be obtained using recombinant methods known in the art, such as, for example by screening libraries from cells expressing the gene, by deriving the gene from a vector known to include the same, or by isolating directly from cells and tissues containing the same, using standard techniques.
  • the gene of interest can be produced synthetically.
  • the present disclosure also provides vectors in which the expression cassette of the present disclosure is inserted.
  • Vectors derived from retroviruses such as the lentivirus are suitable tools to achieve long-term gene transfer since they allow long-term, stable integration of a transgene and its propagation in daughter cells.
  • Lentiviral vectors have the advantage over vectors derived from onco-retroviruses such as murine leukemia viruses in that they can transduce non-proliferating cells, such as hepatocytes. They also have the advantage of low immunogenicity.
  • the expression cassette or nucleic acid sequence is typically and operably linked to a promoter, and incorporated into an expression vector.
  • the vectors can be suitable for replication and integration in eukaryotes.
  • Typical cloning vectors contain transcription and translation terminators, initiation sequences, and promoters useful for regulation of the expression of the desired nucleic acid sequence.
  • the expression constructs of the present disclosure may also be used for nucleic acid immune and gene therapy, using standard gene delivery protocols. Methods for gene delivery are known in the art. See, e.g., U.S, Pat. Nos. 5,399,346, 5,580,859, 5,589,466, incorporated by reference herein in their entireties.
  • the disclosure provides a gene therapy vector.
  • the nucleic acid can be cloned into a number of types of vectors.
  • the nucleic acid can be cloned into a vector including, but not limited to a plasmid, a phagemid, a phage derivative, an animal virus, and a cosmid.
  • Vectors of particular interest include expression vectors, replication vectors, probe generation vectors, and sequencing vectors.
  • the expression vector may be provided to a cell in the form of a viral vector.
  • Viral vector technology is well known in the art and is described, for example, in Sambrook et al, (2001 , Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York), and in other virology and molecular biology manuals.
  • Viruses, which are useful as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpes viruses, and lentiviruses.
  • a suitable vector contains an origin of replication functional in at least one organism, a promoter sequence, convenient restriction endonuclease sites, and one or more selectable markers, (e.g., WO 01/96584; WO 01/29058; and U.S, Pat. No. 6,326, 193).
  • retroviruses provide a convenient platform for gene delivery systems.
  • a selected gene can be inserted into a vector and packaged in retroviral particles using techniques known in the art.
  • the recombinant virus can then be isolated and delivered to cells of the subject either in vivo or ex vivo.
  • retroviral systems are known in the art.
  • adenovirus vectors are used.
  • a number of adenovirus vectors are known in the art.
  • lentivirus vectors are used.
  • promoter elements e.g., enhancers
  • promoters regulate the frequency of transcriptional initiation.
  • these are located in the region 30-110 bp upstream of the start site, although a number of promoters have recently been shown to contain functional elements downstream of the start site as well.
  • the spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another.
  • tk thymidine kinase
  • the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline.
  • individual elements can function either cooperatively or independently to activate transcription.
  • a suitable promoter is the immediate early cytomegalovirus (CMV) promoter sequence.
  • CMV immediate early cytomegalovirus
  • This promoter sequence is a strong constitutive promoter sequence capable of driving high levels of expression of any polynucleotide sequence operatively linked thereto.
  • Another example of a suitable promoter is Elongation Growth Factor- la (EF- la).
  • constitutive promoter sequences may also be used, including, but not limited to the simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, an avian leukemia virus promoter, an Epstein-Barr virus immediate early promoter, a Rous sarcoma virus promoter, as well as human gene promoters such as, but not limited to, the actin promoter, the myosin promoter, the hemoglobin promoter, and the creatine kinase promoter. Further, either constitutive promoters or inducible promoters may be used.
  • SV40 simian virus 40
  • MMTV mouse mammary tumor virus
  • HSV human immunodeficiency virus
  • LTR long terminal repeat
  • MoMuLV promoter MoMuLV promoter
  • an avian leukemia virus promoter an Epstein-Barr virus immediate early promoter
  • an inducible promoter provides a molecular switch capable of turning on expression of the polynucleotide sequence which it is operatively linked when such expression is desired, or turning off the expression when expression is not desired.
  • inducible promoters include, but are not limited to a metallothionein promoter, a glucocorticoid promoter, a progesterone promoter, and a tetracycline promoter.
  • the expression vector to be introduced into a ceil can also contain either a selectable marker gene or a reporter gene or both to facilitate identification and selection of expressing cells from the population of cells sought to be transfected or infected through viral vectors.
  • the selectable marker may be carried on a separate piece of DNA and used in a co- transfection procedure. Both selectable markers and reporter genes may be flanked with appropriate regulatory sequences to enable expression in the host cells.
  • Useful selectable markers include, for example, antibioticresistance genes, such as neo and the like.
  • Reporter genes are used for identifying potentially transfected cells and for evaluating the functionality of regulatory sequences.
  • a reporter gene is a gene that is not present in or expressed by the recipient organism or tissue and that encodes a polypeptide whose expression is manifested by some easily detectable property, e.g., enzymatic activity. Expression of the reporter gene is assayed at a suitable time after the DNA has been introduced into the recipient cells.
  • Suitable reporter genes may include genes encoding luciferase, beta-galactosidase, chloramphenicol acetyl transferase, secreted alkaline phosphatase, or the green fluorescent protein gene (e.g., Ui-Tei et al., 2000 FEBS Letters 479: 79-82). Suitable expression systems are well known and may be prepared using known techniques or obtained commercially. In general, the construct with the minimal 5’ flanking region showing the highest level of expression of reporter gene is identified as the promoter. Such promoter regions may be linked to a reporter gene and used to evaluate agents for the ability to modulate promoter- driven transcription.
  • the vector can be readily introduced into a host cell, e.g., mammalian, bacterial, yeast, or insect cell by any method in the art.
  • the expression vector can be transferred into a host cell by physical, chemical, or biological means.
  • Physical methods for introducing a polynucleotide into a host cell include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and the like. Methods for producing cells comprising vectors and/or exogenous nucleic acids are well-known in the art. See, for example, Sambrook et al. (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York). A method for the introduction of a polynucleotide into a host cell is calcium phosphate transfection.
  • Biological methods for introducing a polynucleotide of interest into a host cell include the use of DNA and RNA vectors.
  • Viral vectors, and especially retroviral vectors have become the most widely used method for inserting genes into mammalian, e.g., human cells.
  • Other viral vectors can be derived from lentivirus, poxviruses, herpes simplex virus I, adenoviruses and adeno- associated viruses, and the like. See, for example, U.S. Pat, Nos. 5,350,674 and 5,585,362.
  • Chemical means for introducing a polynucleotide into a host cell include colloidal dispersion systems, such as macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes.
  • colloidal dispersion systems such as macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes.
  • An exemplary colloidal system for use as a delivery vehicle in vitro and in vivo is a liposome (e.g., an artificial membrane vesicle).
  • an exemplary delivery vehicle is a liposome.
  • lipid formulations is contemplated for the introduction of the nucleic acids into a host cell (in vitro, ex vivo or in vivo).
  • the nucleic acid may be associated with a lipid.
  • the nucleic acid associated with a lipid may be encapsulated in the aqueous interior of a liposome, interspersed within the lipid bilayer of a liposome, attached to a liposome via a linking molecule that is associated with both the liposome and the oligonucleotide, entrapped in a liposome, complexed with a liposome, dispersed in a solution containing a lipid, mixed with a lipid, combined with a lipid, contained as a suspension in a lipid, contained or complexed with a micelle, or otherwise associated with a lipid.
  • Lipid, lipid/DNA or lipid/expression vector associated compositions are not limited to any particular structure in solution.
  • Lipids are fatty substances which may be naturally occurring or synthetic lipids.
  • lipids include the fatty droplets that naturally occur in the cytoplasm as well as the class of compounds which contain long-chain aliphatic hydrocarbons and their derivatives, such as fatty acids, alcohols, amines, amino alcohols, and aldehydes.
  • the vector is a lentiviral vector.
  • the disclosure provides a composition comprising the immune cell (e.g., CAR-T cell), and a pharmaceutically acceptable carrier, diluent and/or excipient.
  • the composition is a liquid composition.
  • the composition is an injectable composition.
  • the concentration of the CAR-T cells in the composition is 1 x 10 3 -lx10 8 cells/ml, or 1 x 10 4 -lx10 7 cells/ml.
  • the composition may comprise buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives.
  • buffers such as neutral buffered saline, phosphate buffered saline and the like
  • carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol
  • proteins polypeptides or amino acids
  • antioxidants such as glycine
  • chelating agents such as EDTA or glutathione
  • adjuvants e.g., aluminum hydroxide
  • preservatives e.g., aluminum hydroxide
  • the disclosure comprises therapeutic applications using cells (e.g., T cells) transduced with a lentiviral vector (LV) encoding the expression cassette of the disclosure.
  • the transduced T cells can target the tumor cell marker BCMA, synergistically activate T cells, and cause T cell immune responses, thereby significantly increasing the killing efficiency against tumor cells.
  • the present disclosure also provides a method for stimulating a T cell-mediated immune response to a target cell population or tissue in a mammal comprising the step of administering to the mammal a CAR-T cell of the disclosure.
  • the present disclosure comprises a class of cell therapies, wherein T cells from autologous patient (or heterologous donor) are isolated, activated and genetically modified to generate CAR-T cells, and then injected into the same patient.
  • T cells from autologous patient or heterologous donor
  • the probability of graft versus host disease in the way is extremely low, and antigens are recognized by T cells in a non- MHC-restricted manner.
  • one CAR-T can treat all cancers that express the antigen.
  • CAR-T cells are able to replicate in vivo resulting in long-term persistence that can lead to sustained tumor control
  • the CAR-T cells of the disclosure can undergo robust in vivo T cell expansion and can persist for an extended amount of time.
  • the CAR mediated immune response may be part of an adoptive immunotherapy approach in which CAR-modified T cells induce an immune response specific to the antigen binding moiety in the CAR.
  • an anti-BCMA CAR-T cell elicits an immune response specific against cells expressing BCMA.
  • Cancers that may be treated include tumors that are unvascularized or largely unvascularized, and tumors that are vascularized. Cancers may include non-solid tumors (such as hematological tumors, for example, leukemias and lymphomas) or solid tumors. Types of cancers to be treated with the CARs include, but are not limited to, carcinoma, blastoma, and sarcoma, and certain leukemia or lymphoid malignancies, benign and malignant tumors, and malignancies e.g., sarcomas, carcinomas, and melanomas. Adult tumors/cancers and pediatric tumors/cancers are also included.
  • Hematologic cancers are cancers of the blood or bone marrow.
  • hematological (or hematogenous) cancers include leukemias, including acute leukemias (such as acute lymphocytic leukemia, acute myelocytic leukemia, acute myelogenous leukemia and myeloblasts, promyeiocytic, myelomonocytic, monocytic and erythroleukemia), chronic leukemias (such as chronic myelocytic (granulocytic) leukemia, chronic myelogenous leukemia, and chronic lymphocytic leukemia), polycythemia vera, lymphoma, Hodgkin’s disease, non-Hodgkin’s lymphoma (indolent and high grade forms), multiple myeloma, Waldenstrom’s macroglobulinemia, heavy chain disease, myelodysplastic syndrome, hairy cell leukemia and myelodysplasia.
  • Solid tumors are abnormal masses of tissue that usually do not contain cysts or liquid areas. Solid tumors can be benign or malignant. Different types of solid tumors are named for the type of cells that form them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumors, such as sarcomas and carcinomas, include fibrosarcoma, myxosarcoma, liposarcoma, mesothelioma, malignant lymphoma, pancreatic cancer and ovarian cancer.
  • the CAR-modified T cells may also serve as a type of vaccine for ex vivo immunization and/or in vivo therapy in a mammal.
  • the mammal is a human.
  • cells are isolated from a mammal (such as a human) and genetically modified (i.e., transduced or transfected in vitro) with a vector expressing a CAR disclosed herein.
  • the CAR-modified cell can be administered to a mammalian recipient to provide a therapeutic benefit.
  • the mammalian recipient may be a human and the CAR-modified cell can be autologous with respect to the recipient.
  • the cells can be allogeneic, syngeneic or xenogeneic with respect to the recipient.
  • compositions and methods for in vivo immunization to elicit an immune response directed against an antigen in a patient In addition to using a cell-based vaccine in terms of ex vivo immunization, the present disclosure also provides compositions and methods for in vivo immunization to elicit an immune response directed against an antigen in a patient.
  • the present disclosure provides methods for treating tumors comprising administering to a subject in need thereof, a therapeutically effective amount of the CAR-modified T cells.
  • compositions of the present disclosure may comprise a target cell population as described herein, in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients.
  • compositions may comprise buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives.
  • buffers such as neutral buffered saline, phosphate buffered saline and the like
  • carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol
  • proteins polypeptides or amino acids
  • antioxidants such as glycine
  • chelating agents such as EDTA or glutathione
  • adjuvants e.g., aluminum hydroxide
  • preservatives e.g., aluminum hydroxide
  • compositions of the present disclosure may be administered in a manner appropriate to the disease to be treated (or prevented).
  • the quantity and frequency of administration will be determined by such factors as the condition of the patient, and the type and severity of the patient’s disease, although appropriate dosages may be determined by clinical trials.
  • an immunologically effective amount When “an immunologically effective amount”, “an anti-tumor effective amount”, “an tumorinhibiting effective amount”, or “therapeutic amount” is indicated, the precise amount of the compositions of the present disclosure to be administered can be determined by a physician with consideration of individual differences in age, weight, tumor size, extent of infection or metastasis, and condition of the patient (subject). It can generally be stated that a pharmaceutical composition comprising the T cells described herein may be administered at a dosage of 10 4 to 10 9 cells/kg body weight, or 10 5 to 10 6 cells/kg body weight, including all integer values within those ranges. T cell compositions may also be administered multiple times at these dosages.
  • the cells can be administered by using infusion techniques that are commonly known in immunotherapy (see, e.g., Rosenberg et al uncomfortable Eng. J. of Med. 319: 1676, 1988).
  • the optimal dosage and treatment regime for a particular patient can readily be determined by one skilled in the art of medicine by monitoring the patient for signs of disease and adjusting the treatment accordingly.
  • the administration of the subject compositions may be carried out in any convenient manner, including by aerosol inhalation, injection, ingestion, transfusion, implantation or transplantation.
  • the compositions described herein may be administered to a patient subcutaneously, intradermaliy, intratumorally, intranodally, intramedullary, intramuscularly, by intravenous injection, or intraperitoneally.
  • the T cell compositions of the present disclosure are administered to a patient by intradermal or subcutaneous injection. In another embodiment, the T cell compositions of the present disclosure may be administered by intravenous injection. The compositions of T cells may be injected directly into a tumor, lymph node, or site of infection.
  • cells activated and expanded using the methods described herein, or other methods known in the art where T cells are expanded to therapeutic levels are administered to a patient in conjunction with (e.g., before, simultaneously or following) any number of relevant treatment modalities, including but not limited to treatment with agents such as antiviral therapy, cidofovir and interleukin-2, Cytarabine (also known as ARA- C) or natalizumab treatment for MS patients or efalizumab treatment for psoriasis patients or other treatments for PML patients.
  • agents such as antiviral therapy, cidofovir and interleukin-2, Cytarabine (also known as ARA- C) or natalizumab treatment for MS patients or efalizumab treatment for psoriasis patients or other treatments for PML patients.
  • the T cells of the disclosure may be used in combination with chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunotherapeutic agents.
  • the cell compositions of the present disclosure are administered to a patient in conjunction with (e.g., before, simultaneously or following) bone marrow transplantation, or the use of chemotherapy agents such as, fludarabine, external-beam radiation therapy (XRT), cyclophosphamide.
  • chemotherapy agents such as, fludarabine, external-beam radiation therapy (XRT), cyclophosphamide.
  • subjects may undergo standard treatment with high dose chemotherapy followed by peripheral blood stem cell transplantation.
  • subjects receive an infusion of the expanded immune cells of the present disclosure.
  • expanded cells are administered before or following surgery.
  • IxlO 6 to IxlO 10 of the modified T cells of the disclosure can be applied to patients by means of, for example, intravenous infusion each treatment or each course of treatment.
  • the main advantages of the present disclosure include:
  • the extracellular antigen binding domain is a specific anti-BCMA scFv; the CARs have shown a great ability of killing tumor cells with low cytotoxicity and low side effects.
  • the chimeric antigen receptor provided by the disclosure can achieve stable expression and membrane localization of CAR protein after T cells are infected by viral vectors (e.g., lentiviruses) carrying the CAR gene.
  • viral vectors e.g., lentiviruses
  • the CAR-modified T cells of the present disclosure have a longer survival time in vivo and strong anti-tumor effect.
  • the scFv used in the present disclosure may be a humanized or human-derived antibody, and is less likely to produce specific immunological rejection.
  • the full-length DNA synthesis and cloning were commissioned by Shanghai Boyi Biotechnology Co., Ltd to achieve the construction of coding plasmids.
  • the pWPT lentiviral vector was selected as a cloning vector, and the cloning sites were BamH I and Sal I sites.
  • the specific sequence is as described above.
  • PBMCs Mononuclear cells
  • PBMCs were seeded in a cell culture flask previously coated with CD3 monoclonal antibody (OKT3) at a final concentration of 5 pg/mL and Retronectin (purchased from TAKARA) at a final concentration of 10 pg/mL.
  • the medium was GT-551 cell culture medium containing 1% human albumin.
  • Recombinant human interleukin 2 (IL-2) was added to the medium at a final concentration of 1000 U/mL.
  • the cells were cultured in an CO 2 incubator with a saturated humidity of 5% at 37 °C.
  • CAR-BCMAs cells can be taken for the corresponding activity assay.
  • Example 3 Detection of the integration rate of the CAR gene in the T cell genome and the expression level of the encoded protein thereof on the membrane surface
  • Cell activation level indicator proteins CD137 and IFNy was detected using CART-BCMAs cells cultured on day 7 in Example 2. IxlO 5 of CART-BCMA cells cultured on day 7 were cultured respectively with BCMA-positive K562-BCMA+E7 tumor cell line and BCMA-negative K562 tumor cell line, or without tumor cells, in 200 pl GT-551 medium for 18h at a ratio of 1:1. Then the expression level of CD 137 on the surface of T cell membrane was detected by flow cytometry and the secretion level of IFNy in the culture supernatant was detected by ELISA.
  • Fig. 4A and Fig. 4B The results are shown in Fig. 4A and Fig. 4B, the expression of CD137 was detected on the surface of four CART cells, and the expression of IFNy was detected in the culture supernatant.
  • CAR-BCMA-20 shows best CD137 activation level and IFNy release level.
  • CART- BCMA-M06 which is constructed based on humanized MO6 antibody sequence shows a weaker level of CD 137 activation but a higher level of IFNy release than CART-BCMA-CA8 which is constructed based on mouse antibody sequence.
  • C-CARO88 induced higher levels of IFN-y release (Figure 4B) and CD137 expression ( Figure 4A) in BCMA-positive tumor cells, compared to CARs based on BCMA-1, BCMA-CA8 and BCMA-M06.
  • CART-BCMA-20 induced greater apoptosis of BCMA-positive tumor cells than CART-BCMA-1 (positive control), CART-BCMA-MO6 and CART-BCMA-CA8 (Figure 5).
  • the CART-BCMAs cells cultured on day 17 in Example 2 were mixed respectively with lxlO 4 CFSE-labeled BCMA-negative cells (NH929) or BCMA-positive self-constructed cells (NH929-BCMA overexpressing tumor cell line) at a ratio of 1:1, 2.5:1, 5:1, 10:1, 20:1 (as shown in Fig. 5).
  • the mixed cells were co-cultured in 100 pl GT-551 medium for 4 h, and then stained with 100 pl 25% PI dye for 15 min.
  • the proportion of PI positive cells in CFSE positive cells was analyzed by flow cytometry.
  • Example 2 The CART-BCMAs cells cultured on day 22 in Example 2 were mixed respectively with IxlO 4 CFSE-labeled BCMA-negative cells (NH929), BCMA-positive self-constructed cells (NH929-BCMA overexpressing tumor cell line) or MM. IS cell line naturally expressed BCMA at a ratio of 1:1, 5:1, 10:1, 10:1, 40:1 (as shown in Fig. 6B). The mixed cells were co-cultured in 100 pl GT-551 medium for 4 h, and then stained with 100 pl 25% PI dye for 15 min. The proportion of PI positive cells in CFSE positive cells was analyzed by flow cytometry.
  • RPMI-8226 cells in logarithmic growth phase were collected, and 4.0x10 6 tumor cells were inoculated subcutaneously in the right back of 6-8 week old B-NDG mice.
  • the tumor volume reached about 120 mm 3
  • the animals were randomly divided into 4 groups according to tumor volume, so that the tumor volume difference of each group was less than 10% of the mean value.
  • the solvent control, 7.5x10 6 NT and 7.5x10 6 CART-BCMAs cells were injected through tail vein respectively.
  • B-NDG mice were xenografted with human myeloma RPMI- 8226 cells.
  • the solvent control, nontransfected (NT) T cells (a negative control) or CART-BCMAs cells were injected through the tail vein of the mice.
  • the antibodies to be screened include: BCMA-1, BCMA-2, BCMA-69, BCMA-72, BCMA- 2A1, BCMA-1E1, BCMA-J22.9, BCMA-20, BCMA-CA8, and BCMA-M06.
  • Structures of chimeric antigen receptor targeting BCMA were constructed on the basis of the above antibodies.
  • BCMA- 1 and BCMA-2 are published Car-T sequences and used as a positive control for screening.
  • CAR-T cells were prepared in the same way as in Example 2, and detected in the same way as in Examples 3 and 4.
  • FIGs 1A, IB and 1C are the experimental results of two batches of CAR-T cells.
  • the expression of Car-T was detected with BCMA-Fc fusion protein. It can be seen that there is a high expression in primary T cells, as seen in Fig. 1A.
  • Fig. IB it can be seen that BCMA-1, BCMA-20, BCMA- IE 1, BCMA-CA8, BCMA-M06, and BCMA-J22.9 can be activated by the BCMA antigen.
  • Fig. 1C shows that the activated BCMA-1, BCMA-20, BCMA- IE 1, BCMA-CA8, BCMA-M06, and BCMA-J22.9 CAR-Ts can produce higher levels of IFN-y.
  • the results show that the CAR-T functions obtained by BCMA-1, BCMA-20, CA8 and MO6 are similar, so BCMA-20, CA8 and MO6CAR-T were further analyzed and studied.
  • the light chain variable region of SEQ ID NO: 1 and the heavy chain variable region of SEQ ID NO: 2 were used to prepare a single chain antibody B20-scFv-rabFc, and membrane protein array experiment was performed.
  • B20-scFv-rabFc 20 ug/mL of B20-scFv-rabFc was added to HEK293T cell array transiently transfected with 5344 membrane proteins, respectively.
  • Flow cytometry found that, under this test condition, B20- scFv-rabFc may cross -recognize with TNFRSF17 (Q02223), MAG (P20916), CR2 (P20023), CXADR (P78310) and DDR2 (Q16832), wherein TNFRSF17, i.e., BCMA is the specific target of B20-scFv, and MAG, CR2, CXADR and DDR2 are suspected non-specific targets.
  • CBM.BCMA CAR-T was co-cultured with 293T cells transfected with BCMA, CR2, CXADR, DDR2, and MAG, respectively.
  • the IFNy, TNF, IL-2 and other cytokines in co-culture supernatant were detected.
  • 293T cells transfected with empty vector were used as a negative control
  • 293T cells transfected with BCMA were used as a positive control.
  • Figure 8 shows results the expression of BCMA, CR2, CXADR, DDR2, and MAG in 293T cells after transfection of relevant plasmids by flow cytometry.
  • cytokine detection results are shown in Figures 9A, 9B and 9C.
  • Only the BCMA expressed on 293T cells can induce CBM.
  • BCMA CAR-T cells to produce large amounts of IFNy/TNF/IL-2, while the other four non-specific surface markers cannot activate CBM.
  • BCMA CAR-T cells are shown in Figures 9A, 9B and 9C.
  • Figure 8 and Figures 9A-9C show whole genome membrane proteome array and validation.
  • TNFRSF17, MAG, CR2, CXADR and DDR2 were identified with moderate binding with B-20 scFv rabFc at the concentrations of 20.0 pg/mL.
  • BCMA-CAR-T cells Only cells expressing TNFRSF17(BCMA) can induce BCMA-CAR-T cells producing a large number of cytokines (IFNy/TNF/IL-2).
  • Specific membrane staining was observed on human lymphocytes in thymus, spleen, lymph nodes, bone marrow and scattered lymphocytes in thyroid gland, adrenal gland at the concentrations of 20.0 pg/mL (Table 1 and Figures 19A-19B).
  • Table 1 a) number of positive staining in 3 donors. b) staining intensity of positive cells and percentage of positive cells in the cells of the same type.
  • the chimeric antigen receptor BCMA-20 (hereafter named C-CARO88) was selected for subsequent experiments.
  • T cells with C-CARO88, non-transfected (NT) T cells (a negative control) and positive control cells (Bluebird bb2121) were co-cultured with BCMA negative cells (NH929) or BCMA positive cells (NH929-BCMA) at different effect/target ratios. Then the cytotoxicity was assayed.
  • Figure 10 shows that C-CARO88 T cells and the positive control demonstrated strong cytotoxicity on NH929-BCMA cells, but not NH929 cells.
  • Example 10 Dose dependent effect of C-CAR088 cells 6 week old B-NDG mice (half male and half female) were selected and intraperitoneally injected with 2.5x10 6 human multiple myeloma cells RPMI-8226. Mice with similar tumor burden were selected and divided into 5 groups, and were injected with 2.5x10 6 C-CARO88 cells (low- dose group), 5x10 6 C-CARO88 cells (medium-dose group), IxlO 7 C-CARO88 cells (high-dose group), T cells and vehicle (with cryoprotectant (CBMG C-CFMC) as vehicle), respectively. The experiment lasted 54 days. During the experiment, the tumor burden of the mice was evaluated every 5 days. At the end of the experiment, the survival rate of the mice was calculated.
  • the treatment-emerging adverse events are shown in Table 3. Only one patient occurred grade 3 cytokine release syndrome in 15 patients. No neurotoxicity events and no dose-limiting toxicity (DLTs) were observed in the dose escalation. The cytopenias is mostly related to Cy/Flu lymphodepletion. It should be noted that the occurrence of a certain degree of cytokine release syndrome after treatment also illustrates the effectiveness of CART treatment from the side. None of the 15 patients had particularly serious cytokines, and C-CARO88 has better safety.
  • Figure 15 shows the treatment condition of the patient of ID Z0203-00801C008.
  • Figure 16 shows the treatment condition of patient of ID Z0203-00701C001.
  • the PET-CT images of the cancer lesions for one patient show that abnormal plasma cells in the bone marrow were significantly decreased after the C-CARO88 treatment (Figure 15).
  • Figures 16A and 16B show that majority of the PCs were abnormal (>90% were CD451o/-), were BCMA+ and clonal for Kappa light chain at the beginning of the experiment (Baseline).
  • Figure 16B shows that after 14 or 28 days of BCMA CAR-T treatment, abnormal PC in BM were significantly decreased, especially Kappa + PC, which decreased from the baseline level of 85.2% to 0%.
  • FIG 17 shows the expansion of C-CARO88 and the decrease of M-protein/sFLC levels in the blood. The results showed that C-CARO88 cells expanded effectively after injection, and the level of M-protein/sFLC markers continued to decrease. The M-marker level of one patient was dropped to 0 on day 14.
  • Example 12 An Anti-BCMA CAR T-Cell Therapy (C-CAR088) Showed Promising Safety and Efficacy Profile in Treating Relapsed or Refractory Multiple Myeloma (r/r MM)
  • BCMA-20 also termed “C- CARO88”
  • R/R MM relapsed/refractory multiple myeloma
  • ORR 95.7%, with 43.5% CR rate at median time to CR: 1.6 months (range: 0.5-9.5). Median time to response: 0.5 month.
  • C-CARO88 showed very good in vitro and in vivo anti-tumor activity and target specificity.
  • Clinical trial results in 24 patients with r/r MM showed strong therapeutic index with promising efficacy and manageable safety profile.
  • BCMA-20 also termed “C- CARO88”
  • R/R MM relapsed/refractory multiple myeloma
  • the median age of the patients dosed was 60 years (range: 45-74 years).
  • the median number of prior lines of therapy was 4 (ranging from 2-12 prior therapies). All patients had received prior treatment with IMiDs (immunomodulatory drug) and proteasome inhibitors.
  • IMiDs immunomodulatory drug
  • proteasome inhibitors 25% patents were previously treated with anti-CD38 monoclonal antibody, while 25% patents had received autologous hematopoietic stem cell transplant.
  • the clinical protocol, as well as the key inclusion criteria, is shown in Figure 21. Specifically, qualified subjects were enrolled, and the collected peripheral blood leukocytes were used to produce the CAR-T cells (C-CARO88). The CAR-T cells were then frozen and stored below -135°C until use. For CAR-T treatment, the CAR-T cells were thawed, and administration completed within 30-45 minutes.
  • the patients received lymphodepletion pretreatment, including fludarabine (30 mg/m 2 /d, intravenous, once per day for three days), and cyclophosphamide (300 mg/m 2 /d, intravenous, once per day for three days).
  • the patients were administered 1.0-6.0 x 10 6 CAR-T cells/kg on day 0 as 3 + 3 dose escalation.
  • follow-ups with the patients were carried out after the infusion starting on day 1 through month 24.
  • the primary objectives included safety: rated of dose limiting toxicities; incidence and severity of treatment-emergent adverse events (CTCAEV5.0). Secondary objectives included efficacy: IMWG 2016 ORR; DOR; PFS; OS. Exploratory objectives included CAR-T expansion and persistence.
  • an overall response rate (ORR, including CR and PR) of our anti- BCMA CAR-T trial is 96%.
  • the best overall response (BOR) included 12 stringent complete responses (sCRs), 2 complete responses (CR), 8 very good partial responses (VGPRs) and 1 partial response (PR).
  • the complete response (CR) is 67%.
  • SD stable disease
  • PR partial response
  • CR complete response
  • PD progressive disease
  • MR minimal response
  • VGPR very good partial response
  • MRD minimal residual disease.
  • the CR/sCR, VGPR and PR for overall and each dose group are shown in Table 5 and Figure 23.
  • 14/21 (66.7%) patients achieved a CR/sCR and all (14/14) patients achieved MRD negative by flow cytometry at 10 -4 -10 -6 .
  • ICANS immune effector cell-associated neurotoxicity syndrome.
  • CRS Cytokine release syndrome

Landscapes

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

Abstract

La présente divulgation concerne des récepteurs antigéniques chimériques (CAR) ciblant BCMA ainsi que des procédés de préparation et des utilisations associés. Les CAR selon la présente divulgation ciblent des cellules positives à BCMA, et peuvent être utilisés pour traiter le lymphome à cellules B positif à BCMA, le myélome multiple et la leucémie à cellules plasmatiques.
EP21901383.6A 2020-12-02 2021-12-01 Récepteurs antigéniques chimériques ciblant bcma Pending EP4255936A1 (fr)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US202063120692P 2020-12-02 2020-12-02
US202163153666P 2021-02-25 2021-02-25
US202163212289P 2021-06-18 2021-06-18
US17/476,661 US11498973B2 (en) 2018-04-12 2021-09-16 BCMA-targeted chimeric antigen receptor as well as preparation method therefor and application thereof
PCT/US2021/061410 WO2022119923A1 (fr) 2020-12-02 2021-12-01 Récepteurs antigéniques chimériques ciblant bcma

Publications (1)

Publication Number Publication Date
EP4255936A1 true EP4255936A1 (fr) 2023-10-11

Family

ID=81853652

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21901383.6A Pending EP4255936A1 (fr) 2020-12-02 2021-12-01 Récepteurs antigéniques chimériques ciblant bcma

Country Status (7)

Country Link
EP (1) EP4255936A1 (fr)
JP (1) JP2023553390A (fr)
KR (1) KR20230117367A (fr)
CN (1) CN116997345A (fr)
AU (1) AU2021393434A1 (fr)
CA (1) CA3201008A1 (fr)
WO (1) WO2022119923A1 (fr)

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116836297A (zh) * 2018-04-12 2023-10-03 上海赛比曼生物科技有限公司 靶向bcma的嵌合抗原受体及其制法和应用
BR112020024246A2 (pt) * 2018-06-01 2021-03-02 University Of Southern California pelo menos um polinucleotídeo recombinante, célula recombinante, receptor de antígeno quimérico, polinucleotídeo que codifica o receptor de antígeno quimérico, vetor, vírus, composição farmacêutica, e, método para tratar câncer

Also Published As

Publication number Publication date
CN116997345A (zh) 2023-11-03
JP2023553390A (ja) 2023-12-21
CA3201008A1 (fr) 2022-06-09
AU2021393434A1 (en) 2023-06-22
WO2022119923A1 (fr) 2022-06-09
KR20230117367A (ko) 2023-08-08

Similar Documents

Publication Publication Date Title
US11142581B2 (en) BCMA-targeted chimeric antigen receptor as well as preparation method therefor and application thereof
US11439665B2 (en) Combined chimeric antigen receptor targeting CD19 and CD20 and application thereof
CN111212903A (zh) 具有自杀基因开关的靶向人间皮素的工程化免疫细胞
AU2020270298A1 (en) Engineered immune cell targeting BCMA and use thereof
CN113087806B (zh) 靶向多种肿瘤的新型car-t细胞及其制备和方法
US20240000839A1 (en) Bcma-targeted chimeric antigen receptors
JP2023521218A (ja) Cd22標的キメラ抗原受容体、その調製方法、及びその適用
EP4255936A1 (fr) Récepteurs antigéniques chimériques ciblant bcma
CN114685683A (zh) 靶向gd2的car-t细胞及其制备和应用
CN114763388A (zh) 靶向b7-h3的car-t细胞及其在急性髓系白血病治疗中的应用
CN110218702B (zh) 靶向cd138和cd19的免疫细胞组合及其应用
US20230104705A1 (en) Combined chimeric antigen receptor targeting cd19 and cd20 and application thereof
EP4284824A2 (fr) Récepteurs antigéniques chimériques ciblant cd20

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20230630

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20240322