CN115491358A - Preparation and application of targeting B7-H3 and FOLR1 double targeting CAR T - Google Patents

Preparation and application of targeting B7-H3 and FOLR1 double targeting CAR T Download PDF

Info

Publication number
CN115491358A
CN115491358A CN202110674194.3A CN202110674194A CN115491358A CN 115491358 A CN115491358 A CN 115491358A CN 202110674194 A CN202110674194 A CN 202110674194A CN 115491358 A CN115491358 A CN 115491358A
Authority
CN
China
Prior art keywords
car
ser
gly
leu
thr
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
CN202110674194.3A
Other languages
Chinese (zh)
Inventor
周春燕
徐敏
刘秀云
张宇
蔡丰灿
龚剑
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.)
Fosun Kaite Biotechnology Co ltd
Original Assignee
Fosun Kaite Biotechnology Co 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
Application filed by Fosun Kaite Biotechnology Co ltd filed Critical Fosun Kaite Biotechnology Co ltd
Priority to CN202110674194.3A priority Critical patent/CN115491358A/en
Publication of CN115491358A publication Critical patent/CN115491358A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0646Natural killers cells [NK], NKT cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Landscapes

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

Abstract

The invention provides a chimeric antigen receptor CAR-immune cell capable of targeting FOLR1 and B7-H3 double targets and application thereof. The application of the CAR-T cells of the invention can cover a wider range of ovarian cancer tumor cells. The CAR-T cell is structurally optimized on the basis of the traditional CAR-T cell, so that the CAR-T cell secretes BiTE, and double-target killing can be realized; meanwhile, the returned T cells are mobilized to a greater extent, so that the tumor cells are killed more efficiently.

Description

Preparation and application of targeting B7-H3 and FOLR1 double targeting CAR T
Technical Field
The invention belongs to the technical field of biology, and particularly relates to preparation and application of a targeting B7-H3 and FOLR1 double targeting target CAR T.
Background
Globally, ovarian cancer is the seventh most common cancer of women, and is also the eighth most lethal cancer. Despite advances in surgical methods and chemotherapy, 5-year survival rates remain below 45%. More importantly, the number of cases diagnosed per year is also increasing, which poses a serious threat to female life.
Cellular immunotherapy is a novel precise targeted therapy, and has a tumor treatment mode with a remarkable curative effect, especially CAR-T. CAR-T cell therapy first collects cells from a patient and then modifies them by activation and genetic engineering so that the T cell surface expresses a Chimeric Antigen Receptor (CAR). The chimeric antigen receptor is composed of an extracellular antigen binding region, namely scFv for recognizing tumor-associated antigen, a transmembrane region, an intracellular signal region and the like. These CAR-T cells are returned to the patient where they can recognize and destroy tumor cells expressing the indicated antigen.
Folate receptor1 (FOLR 1), also known as Folate receptor alpha or Folate binding protein, is expressed less in normal tissues than in tumor tissues. It is expressed on the plasma membrane of cell and can mediate the cell to take up folic acid and influence the downstream path to promote the growth of tumor. Studies have shown that FOLR1 is highly expressed in most ovarian cancers (76% of malignant ovarian cancers) as well as uterine, endometrial, pancreatic, renal, lung, and breast cancers, and it mediates folate uptake or production of regulatory signals to promote tumor cell growth. This expression pattern of FOLR1 makes it a good target for CAR-T cell therapy in ovarian cancer.
B7-H3 (also known as CD 276) is a type I transmembrane protein and belongs to the B7 immunoglobulin superfamily. In humans, mRNA of B7-H3 is widely expressed in various normal tissues, but protein is not expressed or is under-expressed in normal tissues. The B7-H3 protein is widely expressed in various cancer tissues, such as lung cancer, prostatic cancer, breast cancer, colorectal cancer, kidney cancer, ovarian cancer, liver cancer and the like, and the B7-H3 expression frequency of the ovarian cancer is the highest (88.3%). And high expression of B7-H3 protein is associated with disease progression and poor prognosis in a variety of cancers. In addition, in the tumor microenvironment, B7-H3 is also overexpressed on tumor-associated vascular endothelium and fibroblasts, but not on normal vascular endothelium as well as on physiological angiogenic endothelial cells. Given that B7-H3 is not or is poorly expressed in normal tissues and highly expressed in various cancer tissues, B7-H3 is yet another good target for CAR-T cells to treat ovarian cancer.
However, due to the heterogeneity of tumors, ovarian cancer treatment cannot be well achieved using the single targets. In addition, because of the limitations of current genetic modification methods, it is difficult to achieve 100% modification of the reinfused T cells.
However, if the FOLR1 and the B7-H3 are combined, the current method for killing the two targets simultaneously mainly comprises dual specificity CAR-T. However, there are still some drawbacks, and because of the limitations of the current genetic modification methods, it is difficult to achieve 100% modification of the returned T cells, and it is not possible to mobilize T cells in patients.
Therefore, there is an urgent need in the art to develop a more broad-spectrum bispecific CAR-T cell that can be efficiently modified, with a greater extent of mobilization back-transfusion.
Disclosure of Invention
The object of the present invention is to provide a more broad-spectrum bispecific CAR-T cell that can be efficiently modified, with a greater extent of mobilization back-transfusion.
In a first aspect of the invention, there is provided a chimeric antigen receptor CAR-immune cell, said CAR-immune cell comprising the following elements:
(a) A first CAR whose extracellular binding domain contains a first binding element that targets a first target protein and whose extracellular binding domain contains or does not contain a second binding element that targets a second target protein;
(b1) Optionally a second CAR whose extracellular binding domain contains a second binding element that targets a second target protein;
(b2) Optionally a nucleic acid sequence encoding a BiTE, wherein said BiTE targets a second target protein;
wherein, when the extracellular binding domain of the first CAR does not contain a second binding element that targets a second target protein, the CAR-immune cell contains the (b 1) and/or (b 2) elements;
and, the first target protein is folate receptor1 (FOLR 1), and the second target protein is B7-H3; alternatively, the first target protein is B7-H3, and the second target protein is folate receptor1 (FOLR 1).
In another preferred embodiment, the immune cell comprises: t cells, NK cells, or a combination thereof.
In another preferred embodiment, the extracellular binding domain of the first CAR comprises, in order from N-terminus to C-terminus: optionally a signal peptide, a first binding element targeting a first target protein, optionally a linker sequence, and a second binding element targeting a second target protein; the extracellular binding domain of the first CAR comprises from N-terminus to C-terminus: an optional signal peptide, a second binding element targeting a second target protein, an optional linker sequence, and a first binding element targeting a first target protein.
In another preferred embodiment, the linker sequence is a flexible peptide.
In another preferred embodiment, the connection sequence is (G) 4 S) n Wherein n is a positive integer (e.g., 1, 2, 3, 4, 5, or 6).
In another preferred embodiment, the CAR-immune cell contains the following elements:
(a) The extracellular binding domain of the first CAR contains a first binding element targeting a first target protein and does not contain a second binding element targeting a second target protein; and
(b1) A second CAR whose extracellular binding domain comprises a second binding element that targets a second target protein.
In another preferred embodiment, the CAR-immune cell contains the following elements:
(a) The extracellular binding domain of the first CAR contains a first binding element targeting a first target protein and does not contain a second binding element targeting a second target protein; and
(b2) A nucleic acid sequence encoding a BiTE, wherein said BiTE targets a second target protein.
In another preferred embodiment, the first CAR or the second CAR has the structure shown in formula I below:
L-EB-H-TM-C-CD3ζ (I)
in the formula (I), the compound is shown in the specification,
each "-" is independently a linker peptide or a peptide bond;
l is an optional signal peptide sequence;
EB is the extracellular binding domain;
h is an optional hinge region;
TM is a transmembrane domain;
c is a costimulatory signal molecule;
CD3 ζ is a cytoplasmic signaling sequence derived from CD3 ζ.
In another preferred embodiment, the sequence of "H-TM-C" in formula I can be the full-length sequence consisting of the extracellular domain of CD28, the transmembrane region of CD28 and the intracellular domain of CD28 (i.e., extra _ CD 28. TM. ICD).
In another preferred example, the amino acid sequence of the Extra _ CD28 TM _ ICD is shown as SEQ ID NO. 1.
In another preferred embodiment, the signal peptide is a signal peptide of an immune cell surface molecule commonly used in the art, preferably a signal peptide of a protein selected from the group consisting of: CD8, CD28, GM-CSF, CD4, CD137, NKG2D, or a combination thereof.
In another preferred embodiment, the signal peptide is the CD8 leader sequence (signal peptide), and the amino acid sequence thereof is shown in SEQ ID NO: 2.
In another preferred embodiment, the hinge region may be a hinge region of a protein selected from the group consisting of: CD8, CD28, CD137, NKG2D, or a combination thereof.
In another preferred embodiment, the hinge region is that of CD8 and its amino acid sequence is shown in SEQ ID NO 3.
In another preferred embodiment, the transmembrane domain may be a transmembrane region of an immune cell surface molecule commonly used in the art, preferably a transmembrane region of a protein selected from the group consisting of: CD28, CD3epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154, NKG2D, DAP10, DAP12, or a combination thereof.
In another preferred embodiment, the transmembrane domain comprises a CD 8-derived transmembrane region having the amino acid sequence shown in SEQ ID NO. 4.
In another preferred embodiment, the costimulatory signal molecule is a costimulatory signal molecule for a protein selected from the group consisting of: OX40, CD2, CD7, CD27, CD28, CD30, CD40, CD70, CD134, 4-1BB (CD 137), PD1, dap10, CDS, ICAM-1, LFA-1 (CD 11a/CD 18), ICOS (CD 278), NKG2D, GITR, TLR2, DAP10, DAP12, or a combination thereof.
In another preferred embodiment, the costimulatory signal molecule is a costimulatory signal molecule from 4-1BB source, and the amino acid sequence of the costimulatory signal molecule is shown in SEQ ID NO. 5.
In another preferred embodiment, the amino acid sequence of CD3 ζ is as shown in SEQ ID NO 6.
In another preferred embodiment, the first binding element or the second binding element targeting the first or second target protein may be an antigen binding domain ScFv of an antibody targeting said target protein, the ScFv having the structure of formula a or formula B:
V H1 -V L1 (A)
V L1 -V H1 (B)
in the formula, V H1 Is an antibody heavy chain variable region;
V L1 is an antibody light chain variable region;
"-" is a linker peptide or peptide bond.
In another preferred embodiment, the target protein is FOLR1; wherein, V H1 The amino acid sequence of (1) is shown as SEQ ID NO. 10, and V L1 The amino acid sequence of (A) is shown in SEQ ID NO. 11.
In another preferred embodiment, the amino acid sequence of the connecting peptide is shown in SEQ ID NO 12.
In another preferred embodiment, the target protein is B7-H3; wherein, V H1 The amino acid sequence of (1) is shown as SEQ ID NO. 7, and V L1 The amino acid sequence of (A) is shown in SEQ ID NO. 8.
In another preferred embodiment, the first binding element or the second binding element targeting the first or second target protein may be a native sequence that specifically binds to the target protein.
In another preferred embodiment, when said second target protein is B7-H3, said BiTE is a CD3 ScFv.
In another preferred example, the BiTE may be the Fc region of an antibody and the immune cell is an NK cell.
In another preferred embodiment, in the CD 3ScFv, the amino acid sequence of the heavy chain variable region is shown as SEQ ID NO. 13, and the amino acid sequence of the light chain variable region is shown as SEQ ID NO. 14.
In another preferred embodiment, when the CAR-immune cell comprises element (a) and element (b 2), the element (a) and element (b 2) can be derived from a peptide chain expressed from the same nucleic acid construct, the peptide chain having the structure of formula II from N-terminus to C-terminus:
SP1-B1-H-TM-C-CD3 zeta-CS-SP 2-B2-I-BiTE-M (formula II)
In the formula (I), the compound is shown in the specification,
SP1 and SP2 are each independently an optional signal peptide sequence;
b1 and B2 are a first binding element targeting a first target protein, and a second binding element targeting a second target protein, respectively;
H. TM, C and CD3 ζ are as defined for formula I;
CS is a site sequence which can be specifically recognized and cleaved by protease;
i is a null or a connecting sequence;
BiTE is a sequence targeting a second target protein;
m is an optional tag sequence.
In another preferred embodiment, the amino acid sequence of the peptide chain with the structure of formula II is shown in SEQ ID NO. 15.
In another preferred embodiment, the encoding nucleotide sequence of the peptide chain with the structure of the formula II is shown as SEQ ID NO. 16.
In another preferred embodiment, the SP1 is a signal peptide derived from a membrane protein, which may be an immune cell surface molecule commonly used in the art; preferably, said membrane protein-derived signal peptide is a signal peptide of a protein selected from the group consisting of: OX40, CD2, CD7, CD27, CD28, CD30, CD40, CD70, CD134, 4-1BB (CD 137), PD1, dap10, CDS, ICAM-1, LFA-1 (CD 11a/CD 18), ICOS (CD 278), GITR, TLR2, CD3epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD154, NKG2D, DAP10, DAP12, or a combination thereof.
In another preferred embodiment, the SP1 is a signal peptide sequence of CD8, and the amino acid sequence of the signal peptide sequence is shown as SEQ ID NO. 2.
In another preferred embodiment, the SP2 is a signal peptide sequence derived from a secretory protein, which may be a protein secreted by immune cells as is conventional in the art; preferably, the signal peptide of the secretory protein is a signal peptide of a protein selected from the group consisting of: immunoglobulin constitutive subunit, cytokine (IL-2, IL-12, IL-7, IFN-. Gamma.) and the like.
In another preferred embodiment, SP2 is a signal peptide of Ig kappa chain, and its amino acid sequence is shown in SEQ ID NO. 9.
In another preferred embodiment, when element (a) and element (b 2) are derived from the same peptide chain expressed by the nucleic acid construct, and the nucleic acid construct has two different promoters for promoting element (a) and element (b 2), respectively.
In a second aspect of the invention there is provided a nucleic acid molecule encoding the elements (a), (b 1) and/or (b 2) of the CAR-immune cell according to the first aspect of the invention.
In a third aspect of the invention, there is provided a vector comprising a nucleic acid molecule according to the second aspect of the invention.
In another preferred embodiment, the vector comprises DNA and RNA.
In another preferred embodiment, the carrier is selected from the group consisting of: a plasmid, a viral vector, a transposon, or a combination thereof.
In another preferred embodiment, the vector comprises a DNA virus or a retroviral vector.
In another preferred embodiment, the carrier is selected from the group consisting of: a lentiviral vector, an adenoviral vector, an adeno-associated viral vector, or a combination thereof.
In another preferred embodiment, the vector is a lentiviral vector.
In a fourth aspect of the invention, there is provided a host cell comprising a nucleic acid molecule according to the second aspect of the invention integrated exogenously into a vector or chromosome according to the third aspect of the invention.
In another preferred embodiment, the cell is an isolated cell, and/or the cell is a genetically engineered cell.
In another preferred embodiment, the cell is a mammalian cell.
In a fifth aspect of the invention, there is provided a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a CAR-immune cell according to the first aspect of the invention, a nucleic acid molecule according to the second aspect of the invention, a vector according to the third aspect of the invention, or a host cell according to the fourth aspect of the invention.
In a sixth aspect of the invention, there is provided the use of a CAR-immune cell according to the first aspect of the invention, a nucleic acid molecule according to the second aspect of the invention, a vector according to the third aspect of the invention, or a host cell according to the fourth aspect of the invention, for the manufacture of a medicament or formulation for the treatment of a tumour.
In another preferred embodiment, the tumor comprises a B7-H3 positive tumor.
In another preferred embodiment, the tumor comprises a FOLR1 positive tumor.
In another preferred embodiment, the tumor includes any tumor that is B7-H3 positive and FOLR1 positive.
In another preferred embodiment, the tumor comprises: ovarian cancer, breast cancer, gastric cancer, prostate cancer, colon cancer, lung cancer, or a combination thereof.
In another preferred embodiment, the tumor is ovarian cancer.
In a seventh aspect of the invention, there is provided a method of treating a disease comprising administering to a subject in need thereof an amount of a chimeric antigen receptor according to the first aspect of the invention, a nucleic acid molecule according to the second aspect of the invention, a vector according to the third aspect of the invention, or a cell according to the fourth aspect of the invention, or a pharmaceutical composition according to the fifth aspect of the invention.
In an eighth aspect of the invention there is provided a method of making a CAR-immune cell according to the first aspect of the invention, the method comprising the steps of: transducing a nucleic acid molecule according to the second aspect of the invention or a vector according to the third aspect of the invention into an immune cell, thereby obtaining the CAR-immune cell.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be repeated herein, depending on the space.
Drawings
FIG. 1 shows the structural schematic diagram of FOLR1-BB, FOLR1-B7H3-BB and B7H3-FOLR 1-BB. Wherein Hinge TM represents the Hinge and transmembrane regions of CD 8; 4-1BB represents the intracellular signaling region of 4-1 BB.
FIG. 2 shows a comparison of the cell killing effect of Tandem-CAR (FOLR 1-B7H3-BB, B7H3-FOLR 1-BB) and single-target secondary CAR (FOLR 1-BB).
FIG. 3 shows a comparison of the persistence of the cell killing effect of Tandem-CAR (FOLR 1-B7H3-BB, B7H3-FOLR 1-BB) and a single-target secondary CAR (FOLR 1-BB).
FIG. 4 shows a schematic diagram of the structures of FOLR1-BB-B7H3 BITE, B7H3 BITE only, and second generation CAR (FOLR 1-BB).
FIG. 5 shows a comparison of the cell killing effect of OLR1-BB-B7H3 BITE, B7H3 BITE only, and the second generation CAR (FOLR 1-BB).
FIG. 6 shows the detection of killing of target cells in the lower chamber of the Transwell.
Figure 7 shows the results of the BiTE secreted-CAR supernatant mediated cell killing assay.
Detailed Description
The inventor develops a targeting B7-H3 and FOLR1 double targeting target CAR-T cell for the first time through extensive and intensive research and a large amount of screening. In the present invention, the inventors combined the FOLR1 antibody with B7-H3 to construct bispecific CAR-T cells. The application of the CAR-T cells of the invention can cover a wider range of ovarian cancer tumor cells. The CAR-T cell is structurally optimized on the basis of the traditional CAR-T cell, so that the CAR-T cell secretes BiTE, and double-target killing can be realized; meanwhile, the returned T cells are mobilized to a greater extent, so that the tumor cells are killed more efficiently. The present invention has been completed on the basis of this finding.
Term(s)
In order that the disclosure may be more readily understood, certain terms are first defined. As used in this application, each of the following terms shall have the meaning given below, unless explicitly specified otherwise herein. Other definitions are set forth throughout the application.
The term "about" can refer to a value or composition that is within an acceptable error range for the particular value or composition, as determined by one of ordinary skill in the art, which will depend in part on how the value or composition is measured or determined.
The term "administering" refers to physically introducing the product of the invention into a subject using any of a variety of methods and delivery systems known to those skilled in the art, including intravenous, intramuscular, subcutaneous, intraperitoneal, spinal cord or other parenteral routes of administration, e.g., by injection or infusion.
The term "antibody" (Ab) shall include, but is not limited to, an immunoglobulin that specifically binds an antigen and comprises at least two heavy (H) chains and two light (L) chains, or antigen-binding portions thereof, interconnected by disulfide bonds. Each H chain comprises a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region comprises three constant domains, CH1, CH2 and CH3. Each light chain comprises a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region comprises one constant domain CL. The VH and VL regions may be further subdivided into regions of hypervariability, termed Complementarity Determining Regions (CDRs), interspersed with regions that are more conserved, termed Framework Regions (FRs). Each VH and VL comprises three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain binding domains that interact with antigens.
It should be understood that the amino acid names herein are given by the international single english letter designation, and the three english letters abbreviation corresponding to the amino acid names are: ala (A), arg (R), asn (N), asp (D), cys (C), gln (Q), glu (E), gly (G), his (H), I1E (I), leu (L), lys (K), met (M), phe (F), pro (P), ser (S), thr (T), trp (W), tyr (Y) and Val (V).
Chimeric Antigen Receptor (CAR) -immune cells
As used herein, the terms "Chimeric Antigen Receptor (CAR) -immune cell", "CAR-immune cell", "immune cell of the invention" are used interchangeably and all refer to a CAR-immune cell having the elements (a), (b 1) and/or (b 2) according to the first aspect of the invention, with dual target specificity.
The CAR-immune cells of the invention express a first CAR and optionally a second CAR. Wherein the first CAR and the second CAR, except for the specific extracellular binding domain, each have the structure of a chimeric antigen receptor as is conventional in the art.
In one embodiment of the invention, the CAR-immune cell is capable of secreting a BiTE sequence that targets a corresponding second CAR, which is capable of drawing the CAR-immune cell and unmodified immune cell into proximity with a target cell expressing the second CAR target protein, thereby further promoting killing of the target cell by the CAR-immune cell.
As used herein, the term "chimeric antigen receptor of the invention" refers to a first CAR and/or a second CAR in the invention.
The Chimeric Antigen Receptors (CARs) of the invention include an extracellular domain, a transmembrane domain, and an intracellular domain. The extracellular domain includes a target-specific binding member (also referred to as an antigen-binding domain). The intracellular domain includes a costimulatory signaling region and a zeta chain moiety. The costimulatory signaling region refers to a portion of the intracellular domain that includes the costimulatory molecule. Costimulatory molecules are cell surface molecules required for efficient response of lymphocytes to antigens, rather than antigen receptors or their ligands.
A linker may 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. As used herein, the term "linker" generally refers to any oligopeptide or polypeptide that functions to link a transmembrane domain to an extracellular domain or a cytoplasmic domain of a polypeptide chain. The linker may comprise 0-300 amino acids, preferably 2 to 100 amino acids and most preferably 3 to 50 amino acids.
As used herein, "antigen binding domain" and "single chain antibody fragment" each refers to a Fab fragment, fab 'fragment, F (ab') 2 A fragment, or a single Fv fragment. Fv antibodies contain the variable regions of the antibody heavy chain, the variable regions of the light chain, but no constant regions, and have the smallest antibody fragment of the entire antigen binding site. Generally, fv antibodies also comprise a polypeptide linker between the VH and VL domains, and are capable of forming the structure required for antigen binding. The antigen binding domain is typically a scFv (single-chain variable fragment). The size of scFv is typically 1/6 of that of an intact antibody. Single chain antibodies are preferably a sequence of amino acids encoded by a single nucleotide chain. In a preferred embodiment of the present invention, the scFv comprises an antibody, preferably a single chain antibody, that specifically recognizes FOLR 1.
For the hinge region and transmembrane region (transmembrane domain), the CAR can be designed to include a transmembrane domain fused to the extracellular domain of the CAR. In one embodiment, a transmembrane domain that is naturally associated with one of the domains in the CAR is used. In some examples, the transmembrane domains may be selected, or modified by amino acid substitutions, to avoid binding such domains to the transmembrane domains of the same or different surface membrane proteins, thereby minimizing interaction with other members of the receptor complex.
Carrier
Nucleic acid sequences encoding the desired molecule can be obtained using recombinant methods known in the art, such as, for example, by screening libraries from cells expressing the gene, by obtaining the gene from vectors known to include the gene, or by direct isolation from cells and tissues containing the gene using standard techniques. Alternatively, the gene of interest may be produced synthetically.
The present invention also provides a vector into which the expression cassette of the present invention is inserted. Vectors derived from retroviruses such as lentiviruses are suitable tools for achieving long-term gene transfer, as they allow long-term, stable integration of the transgene and its propagation in daughter cells. Lentiviral vectors have advantages over vectors derived from oncogenic retroviruses such as murine leukemia virus, in that they can transduce non-proliferating cells such as hepatocytes. They also have the advantage of low immunogenicity.
In brief summary, an expression cassette or nucleic acid sequence of the invention is typically operably linked to a promoter and incorporated into an expression vector. The vector is suitable for replication and integration into eukaryotic cells. Typical cloning vectors contain transcriptional and translational terminators, initial sequences, and promoters that may be used to regulate the expression of the desired nucleic acid sequence.
The expression constructs of the invention may also be used for nucleic acid immunization and gene therapy using standard gene delivery protocols. Methods of gene delivery are known in the art. See, for example, U.S. Pat. nos. 5,399,346, 5,580,859, 5,589,466, which are incorporated herein by reference in their entirety. In another embodiment, the invention provides a gene therapy vector.
The nucleic acid can be cloned into many types of vectors. For example, the nucleic acid can be cloned into vectors including, but not limited to, plasmids, phagemids, phage derivatives, animal viruses, and cosmids. Specific vectors of interest include expression vectors, replication vectors, probe generation vectors, and sequencing vectors.
Further, the expression vector may be provided to the 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. Viruses that can be used as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpes viruses, and lentiviruses. Generally, suitable vectors comprise an origin of replication, a promoter sequence, a convenient restriction enzyme site, and one or more selectable markers that function in at least one organism (e.g., WO01/96584, WO01/29058; and U.S. Pat. No. 6,326,193).
Many virus-based systems have been developed for gene transfer into mammalian cells. For example, retroviruses provide a convenient platform for gene delivery systems. The selected gene can be inserted into a vector and packaged into a retroviral particle using techniques known in the art. The recombinant virus can then be isolated and delivered to the subject cells in vivo or ex vivo. Many retroviral systems are known in the art. In some embodiments, an adenoviral vector is used. Many adenoviral vectors are known in the art. In one embodiment, a lentiviral vector is used.
Additional promoter elements, such as enhancers, may regulate the frequency of transcription initiation. Typically, these are located in the 30-110bp region upstream of the start site, although many promoters have recently been shown to also contain functional elements downstream of the start site. The spacing between promoter elements is often flexible so that promoter function is maintained when the elements are inverted or moved relative to one another. In the thymidine kinase (tk) promoter, the spacing between promoter elements can be increased by 50bp apart, and activity begins to decline. Depending on the promoter, it appears that the individual elements may function cooperatively or independently to initiate transcription.
An example of a suitable promoter is the immediate early Cytomegalovirus (CMV) promoter sequence. The promoter sequence is a strong constitutive promoter sequence capable of driving high level expression of any polynucleotide sequence operably linked thereto. Another example of a suitable promoter is elongation growth factor-1 α (EF-1 α). However, other constitutive promoter sequences may also be used, including, but not limited to, the simian virus 40 (SV 40) early promoter, the mouse mammary cancer virus (MMTV), the Human Immunodeficiency Virus (HIV) Long Terminal Repeat (LTR) promoter, the MoMuLV promoter, the avian leukemia virus promoter, the Epstein-Barr (Epstein-Barr) virus immediate early promoter, the rous sarcoma virus promoter, and human gene promoters, such as, but not limited to, the actin promoter, myosin promoter, heme promoter, and creatine kinase promoter. Further, the present invention should not be limited to the use of constitutive promoters. Inducible promoters are also contemplated as part of the invention. The use of an inducible promoter provides a molecular switch that is capable of turning on expression of a polynucleotide sequence operably linked to the inducible promoter when such expression is desired, or turning off expression when expression is not desired. Examples of inducible promoters include, but are not limited to, the metallothionein promoter, the glucocorticoid promoter, the progesterone promoter, and the tetracycline promoter.
To assess the expression of the CAR polypeptide or portion thereof, the expression vector introduced into the cells can also comprise either or both of a selectable marker gene or a reporter gene to facilitate identification and selection of expressing cells from a population of cells sought to be transfected or infected by the viral vector. In other aspects, the selectable marker may be carried on a single piece of DNA and used in a co-transfection procedure. Both the selectable marker and the reporter gene may be flanked by appropriate regulatory sequences to enable expression in a host cell. Useful selectable markers include, for example, antibiotic resistance genes, such as neo and the like.
The reporter gene is used to identify potentially transfected cells and to evaluate the functionality of the regulatory sequences. Typically, the reporter gene is the following: which is not present in or expressed by the recipient organism or tissue and which encodes a polypeptide whose expression is clearly indicated by some readily detectable property, such as enzymatic activity. After the DNA has been introduced into the recipient cell, the expression of the reporter gene is assayed at an appropriate time. Suitable reporter genes may include genes encoding luciferase, β -galactosidase, chloramphenicol acetyltransferase, secreted alkaline phosphatase, or green fluorescent protein (e.g., ui-Tei et al, 2000febs letters 479. Suitable expression systems are well known and can be prepared using known techniques or obtained commercially. Generally, the construct with the minimum of 5 flanking regions that showed the highest level of reporter gene expression was identified as the promoter. Such promoter regions can be linked to reporter genes and used to evaluate the ability of an agent to modulate promoter-driven transcription.
Methods for introducing and expressing genes into cells are known in the art. In the context of expression vectors, the vector may be readily introduced into a host cell by any method known in the art, e.g., mammalian, bacterial, yeast or insect cells. For example, the expression vector may be transferred into a host cell by physical, chemical or biological means.
Physical methods for introducing polynucleotides into host cells 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, e.g., sambrook et al (2001, molecular cloning. A preferred method for introducing the 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, particularly retroviral vectors, have become the most widely used method for inserting genes into mammalian, e.g., human, cells. Other viral vectors may be derived from lentiviruses, poxviruses, herpes simplex virus I, adenoviruses, adeno-associated viruses, and the like. See, for example, U.S. Pat. nos. 5,350,674 and 5,585,362.
Chemical means of introducing polynucleotides into host cells include colloidal dispersion systems such as macromolecular complexes, nanocapsules, microspheres, beads; and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. Exemplary colloidal systems for use as delivery vehicles in vitro and in vivo are liposomes (e.g., artificial membrane vesicles).
In the case of non-viral delivery systems, an exemplary delivery vehicle is a liposome. Lipid formulations are contemplated for use to introduce nucleic acids into host cells (ex vivo or in vivo). In another aspect, the nucleic acid can be associated with a lipid. The nucleic acid associated with the lipid may be encapsulated in the aqueous interior of the liposome, dispersed within the lipid bilayer of the liposome, attached to the liposome via a linker molecule associated with both the liposome and the oligonucleotide, entrapped in the liposome, complexed with the liposome, dispersed in a solution comprising the lipid, mixed with the lipid, associated with the lipid, contained as a suspension in the lipid, contained in or complexed with a micelle, or otherwise associated with the lipid. The lipid, lipid/DNA, or lipid/expression vector associated with the composition is not limited to any particular structure in solution. For example, they may be present in bilayer structures, either as micelles or with a "collapsed" structure. They may also simply be dispersed in a solution, possibly forming aggregates that are not uniform in size or shape. Lipids are fatty substances, which may be naturally occurring or synthetic lipids. For example, lipids include fatty droplets that occur naturally in the cytoplasm as well as such compounds that contain long-chain aliphatic hydrocarbons and their derivatives such as fatty acids, alcohols, amines, amino alcohols, and aldehydes.
In a preferred embodiment of the invention, the vector is a lentiviral vector.
Preparation
The invention provides a composition comprising a CAR-T cell according to the first aspect of the invention, and a pharmaceutically acceptable carrier, diluent or excipient. In one embodiment, the formulation is a liquid formulation. Preferably, the formulation is an injection. Preferably, the CAR-T cells are present in the formulation at a concentration of 1X 10 3 -1×10 8 Individual cells/ml, more preferably 1X 10 4 -1×10 7 Individual cells/ml.
In one embodiment, the formulation may include buffers such as neutral buffered saline, sulfate buffered saline, and the like; carbohydrates such as glucose, mannose, sucrose or dextran, mannitol; a protein; polypeptides or amino acids such as glycine; an antioxidant; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and a preservative. The formulations of the present invention are preferably formulated for intravenous administration.
Therapeutic applications
The invention includes therapeutic applications of cells (e.g., T cells) transduced with Lentiviral Vectors (LV) encoding expression cassettes of the invention. The transduced T cells can target markers B7-H3 and FOLR1 of tumor cells, can be used for autologous and allogeneic tumor treatment, can be prepared in a large scale, have uniform and stable quality, and can be used for any patient at any time.
Accordingly, the present invention also provides a method of stimulating a T cell-mediated immune response to a target cell population or tissue of a mammal comprising the steps of: administering to the mammal the CAR-T cells of the invention.
In one embodiment, the invention includes a class of cell therapies in which T cells are genetically modified to express a CAR of the invention, and the CAR-T cells are injected into a recipient in need thereof. The injected cells are capable of killing tumor cells of the recipient. Unlike antibody therapy, CAR-T cells are able to replicate in vivo, resulting in long-term persistence that can lead to sustained tumor control.
In one embodiment, the CAR-T cells of the invention can undergo robust in vivo T cell expansion and can last for an extended amount of time. In addition, the CAR-mediated immune response can be part of an adoptive immunotherapy step, wherein the CAR-modified T cell induces an immune response specific to the antigen binding domain in the CAR. For example, anti-B7-H3 CAR-T cells elicit a specific immune response to B7-H3 expressing cells.
Although the data disclosed herein specifically disclose lentiviral vectors comprising an anti-FOLR 1-B7-H3scFv, a human Fc hinge region, an ICOS transmembrane and intracellular region, and 4-1BB and CD3 zeta signaling domains, the invention should be construed to include any number of variations on each of the construct components.
Treatable cancers include tumors that are not vascularized or have not been substantially vascularized, as well as vascularized tumors. The cancer may comprise a non-solid tumor (such as a hematological tumor, e.g., leukemia and lymphoma) or may comprise a solid tumor. The types of cancer treated with the CARs of the invention include, but are not limited to, carcinomas, blastomas and sarcomas, and certain leukemias or lymphoid malignancies, benign and malignant tumors, such as sarcomas, carcinomas and melanomas. Also included are adult tumors/cancers and pediatric tumors/cancers.
A solid tumor is an abnormal mass of tissue that generally does not contain cysts or fluid areas. Solid tumors can be benign or malignant. Different types of solid tumors are named for the cell types that form them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumors such as sarcomas and carcinomas include fibrosarcoma, myxosarcoma, liposarcoma mesothelioma, lymphoid malignancies, pancreatic cancer of the ovary.
The CAR-immune cells of the invention may also be used as a type of vaccine for ex vivo immunization and/or in vivo therapy of mammals. Preferably, the mammal is a human.
For ex vivo immunization, at least one of the following occurs in vitro prior to administration of the cells into a mammal: i) Expanding the cell, ii) introducing a nucleic acid encoding the CAR into the cell, and/or iii) cryopreserving the cell.
Ex vivo procedures are well known in the art and are discussed more fully below. Briefly, cells are isolated from a mammal (preferably a human) and genetically modified (i.e., transduced or transfected in vitro) with a vector expressing a CAR disclosed herein. The CAR-modified cells can be administered to a mammalian recipient to provide a therapeutic benefit. The mammalian recipient can be a human, and the CAR-modified cells can be autologous with respect to the recipient. Alternatively, the cells may be allogeneic, syngeneic (syngeneic) or xenogeneic with respect to the recipient.
In addition to using cell-based vaccines for ex vivo immunization, the present invention also provides compositions and methods for in vivo immunization to elicit an immune response against an antigen in a patient.
The invention provides a method of treating a tumor comprising administering to a subject in need thereof a therapeutically effective amount of a CAR-immune cell of the invention.
The CAR-immune cells of the invention can be administered alone or as a pharmaceutical composition in combination with diluents and/or with other components such as IL-2, IL-17 or other cytokines or cell populations. Briefly, a pharmaceutical composition of the invention may comprise a target cell population as described herein, in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents, or excipients. Such compositions may include buffers such as neutral buffered saline, sulfate buffered saline, and the like; carbohydrates such as glucose, mannose, sucrose or dextran, mannitol; a protein; polypeptides or amino acids such as glycine; an antioxidant; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and a preservative. The compositions of the present invention are preferably formulated for intravenous administration.
The pharmaceutical compositions of the present invention may be administered in a manner suitable for the disease to be treated (or prevented). The number 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 the appropriate dosage may be determined by clinical trials.
When referring to an "immunologically effective amount", "an anti-tumor effective amount", "a tumor-inhibiting effective amount", or a "therapeutic amount", the precise amount of the composition of the invention to be administered can be determined by a physician, taking into account the age, weight, tumor size, extent of infection or metastasis, and individual differences in the condition of the patient (subject). It can be generally pointed out that: pharmaceutical compositions comprising T cells described herein can be in the range of 10 4 To 10 9 Dosage of individual cells/kg body weight, preferably 10 5 To 10 6 Doses of individual cells per kg body weight (including all integer values within those ranges) were administered. The T cell composition may also be administered multiple times at these doses. Cells can be administered by using infusion techniques well known in immunotherapy (see, e.g., rosenberg et al, new Eng.J.of Med.319:1676, 1988). Optimal dosages and treatment regimens for a particular patient can be readily determined by those skilled in the medical arts by monitoring the patient for signs of disease and adjusting the treatment accordingly.
Administration of the subject composition may be carried out in any convenient manner, including by spraying, injection, swallowing, infusion, implantation or transplantation. The compositions described herein can be administered to a patient subcutaneously, intradermally, intratumorally, intranodal, intraspinally, intramuscularly, by intravenous (i.v.) injection, or intraperitoneally. In one embodiment, the T cell composition of the invention is administered to a patient by intradermal or subcutaneous injection. In another embodiment, the T cell composition of the invention is preferably administered by i.v. injection. The composition of T cells can be injected directly into the tumor, lymph node or site of infection.
In certain embodiments of the invention, cells activated and expanded using the methods described herein or other methods known in the art for expanding T cells to therapeutic levels are administered to a patient in conjunction with (e.g., prior to, concurrently with, or subsequent to) any number of relevant treatment modalities, including but not limited to treatment with: such as bevacizumab, megestrol acetate dispersible tablet, paclitaxel injection, ifosfamide, treatment of ovarian cancer patients with ifosfamide for injection. In further embodiments, the CAR-immune cells of the invention can be used in combination with: chemotherapy, radiation, immunosuppressive agents such as cyclosporine, azathioprine, methotrexate, mycophenolate mofetil, and FK506, antibodies, or other immunotherapeutic agents. In a further embodiment, the cell composition of the invention is administered to the patient in conjunction with (e.g., prior to, concurrently with, or subsequent to) bone marrow transplantation with a chemotherapeutic agent such as fludarabine, external beam radiation therapy (XRT), cyclophosphamide. For example, in one embodiment, the subject may undergo standard treatment with high dose chemotherapy followed by peripheral blood stem cell transplantation. In some embodiments, after transplantation, the subject receives an injection of the expanded immune cells of the invention. In an additional embodiment, the expanded cells are administered pre-or post-surgery.
The dosage of the above treatments administered to a patient will vary with the precise nature of the condition being treated and the recipient of the treatment. The proportion of doses administered to a human can be effected in accordance with accepted practice in the art. Typically, 1X 10 may be administered per treatment or per course of treatment 6 To 1 × 10 10 The CAR-immune cells of the invention are administered to a patient, for example, by intravenous infusion.
The main advantages of the present invention include:
1) Because of tumor heterogeneity, ovarian cancer treatment may not be well achieved using a single target, in this patent we constructed CAR-T cells using FOLR1 antibody in combination with B7-H3. A wider range of ovarian cancer tumor cells can be covered.
2) Because of the limitations of current genetic modification methods, it is difficult to achieve 100% modification of the reinfused T cells. The invention carries out structure optimization on the basis of CAR-T, ensures that the CAR-T secretes BiTE, can kill double targets, simultaneously mobilizes the returned T cells to a greater extent, and realizes the killing of tumor cells.
The invention is further illustrated by the following examples: the following examples are illustrative and are intended to be purely exemplary of the invention and are not intended to limit the scope of the invention. It is expressly intended that all such similar substitutes and modifications apparent to those skilled in the art are deemed to be included in the invention. The methods used in the present invention are, unless otherwise specified, conventional in the art; the reagents of the present invention are conventional in the art unless otherwise specified.
TABLE 1 sequences of the invention
Figure BDA0003120393180000171
Figure BDA0003120393180000181
Figure BDA0003120393180000191
Example 1: functional testing of Tandem-CAR
1.1 viral packaging
According to the structure shown in figure 1, the gene is synthesized and cloned to a virus transferred to a lentiviral vector pWPXld, and the vector can be used after being sequenced without errors. 293T cells (DMEM containing 10% FBS) cultured in 10cm dishes were controlled to a density of 70% -80%. Plasmids pWPXld, pMD2G, psPAX2 were added to 2mL of Opti-Mem at a ratio of 10ug. This was then mixed with X-tremeGENE HP DNA Transfection Reagent. In general, the ratio of DNA to Transfection Reagent was 1. Incubate at room temperature for 15min.
The mixture was added dropwise to the 293 dish already prepared. After culturing for 6h in a cell culture incubator, the medium was washed and replaced with 15ml of MEM complete medium. After further culturing for 72h, the virus was harvested.
The virus supernatant was centrifuged at about 500g for 5 minutes to remove cell debris, and the supernatant was collected. The supernatant recovered above was then filtered through a 0.45 μm filter. Then, the mixture was concentrated 15 times for use. If not used for a while, they should be dispensed in time and frozen in liquid nitrogen and transferred to a-80 ℃ freezer as soon as possible.
1.2 CAR-T cell preparation
Commercial PBMC cells were cultured with X-VIVO 15 (LONZA, 04-418Q) containing 5 ‰ human serum albumin at an initial cell density of 1 × 10 6 Individual cells/mL.
CD3 antibody was added to a final concentration of 50ng/mL and IL-2 was added at 300 IU/mL to activate T cell expansion. After 48 hours of cell activation, appropriate amounts of virus and polybrene were added to infect T cells. Centrifuging for 90min at 800g, and culturing in a cell culture box after centrifugation.
After 24h of lentivirus infection, the cell suspension was aspirated and expressed at 1X 10 6 The cells/mL concentration was supplemented with completely fresh X-VIVO 15 medium. Observing cell density every day, and timely supplementing T cell culture solution to maintain the density of T cells at 1 × 10 6 About one cell/mL. And continuing to expand for 5-10 days to complete the preparation of the CAR-T cells.
1.3 cell killingBy authentication
And (3) constructing the target cell SKOV3-Hibit expressed by the Hibit +. For 96-well plate plating, 4E 3/well target cells were added to CAR-T cells at different E: T ratios (1.5, 1, 0.5, 1, 0.15). The culture was carried out at 37 ℃ in a 5% CO2-containing culture phase. When the preset incubation time is reached for 20h, the method is followed
Figure BDA0003120393180000201
The kit specification of the HiBiT excel cellular Detection System is used for detecting and detecting the killing activity of CAR-T cells, and the method refers to the kit specification. Cytotoxicity was calculated as follows:
cytotoxicity% = (experimental-spontaneous)/(maximum release-spontaneous) × 100%
The results of the cell killing assay are detailed in FIG. 2. The results show that: the killing of Tandem-CAR FOLR1-B7H3-BB modified CAR-T cells was not as different from that of secondary CAR modified CAR-T cells, but its persistence was superior to that of secondary CARs.
1.4 verification of cell killing Endurance
Day D0, SKOV3 was plated into 96-well plates with 5E4 cells per well. Effector cells were then added as per E: T of 2. CAR + cells were then counted every 2 days using Counting Beads and target cells were then replenished according to the CAR + numbers obtained (E: T is 2. Proliferation of CAR + was then counted.
The results of the cell killing assay are detailed in FIG. 3. The results show that: the persistence of Tandem CAR killing on target cells was superior to that of the second generation CARs.
Example 2: BITE-secreting-CAR functional assays
2.1 viral packaging
According to the structure shown in FIG. 4, the gene is synthesized and cloned to a lentivirus vector pWPXld, and the vector can be used after being sequenced without errors. 293T cells (DMEM containing 10% FBS) cultured in 10cm dishes were controlled to a density of 70% -80%. Plasmid pwxld, pMD2G, psPAX2 were added to 2mL of Opti-Mem at a ratio of 10ug. This was then mixed with X-tremeGENE HP DNA Transfection Reagent. In general, the ratio of DNA to Transfection Reagent was 1. Incubate at room temperature for 15min.
The mixture was added dropwise to the prepared 293 dish. After culturing for 6h in a cell culture incubator, the medium was washed and replaced with 15ml of MEM complete medium. And after further culturing for 72h, collecting the virus.
The virus supernatant was centrifuged at about 500g for 5 minutes to remove cell debris, and the supernatant was collected. The supernatant recovered above was then filtered through a 0.45 μm filter. Then, the mixture was concentrated 15 times for use. If not used for a while, they should be dispensed in time and frozen in liquid nitrogen and transferred to a-80 ℃ freezer as soon as possible.
2.2 CAR-T cell preparation
Commercial PBMC cells were cultured with X-VIVO 15 (LONZA, 04-418Q) containing 5 ‰ human serum albumin at an initial cell density of 1 × 10 6 Individual cells/mL.
CD3 antibody was added to a final concentration of 50ng/mL and IL-2 was added at 300 IU/mL to activate T cell expansion. After 48 hours of cell activation, appropriate amounts of virus and polybrene were added to infect T cells. Centrifuging for 90min at 800g, and culturing in a cell culture box after centrifugation.
After 24h of lentivirus infection, the cell suspension was aspirated and expressed at 1X 10 6 The cells/mL concentration was supplemented with completely fresh X-VIVO 15 medium. Observing cell density every day, and timely supplementing T cell culture solution to maintain the density of T cells at 1 × 10 6 About one cell/mL. And continuing to expand for 5-10 days to complete the preparation of the CAR-T cells.
2.3 verification of cell killing
And (3) constructing the target cell SKOV3-Hibit expressed by the Hibit +. For plating in 96-well plates, 4E 3/well target cells were added to CAR-T cells at different E: T ratios (0.01, 1, 0.03, 0.1, 1, 0.3. 5% at 37 ℃Culturing in an incubator containing CO 2. When the preset incubation time is reached for 20h, the method is followed
Figure BDA0003120393180000222
The kit specification of the HiBiT excellular Detection System is used for detecting the killing activity of CAR-T cells, and the method refers to the kit specification. Cytotoxicity was calculated as follows:
cytotoxicity% = (experimental-spontaneous)/(maximum release-spontaneous) × 100%
The results are shown in FIG. 5: when the E: T is 3.
2.4 Functional validation of BiTE secreted CAR
To further verify that BITE in FOLR1-BB- (B7-H3-BITE) plays a role, we performed a transwell experiment: FOLR1-BB- (B7-H3-BITE) CAR-T cells 1X 10 6 The cell number was resuspended in 200. Mu.L of X-VIVO 15 medium, respectively, and then added to the upper chamber of a 0.4- μm transwell (Coning, 3413); unedited T cells and target cells (SKOV 3-Hibit) were resuspended in 500. Mu.L of X-VIVO-15 medium according to the E: T ratio (10) and added to the Transwell lower chamber, the number of target cells was 2E 5; carefully placing the upper chamber into the lower chamber, and culturing in an incubator for 18 hours; the plates were removed from the co-culture overnight and 500. Mu.L of the lower chamber cells and supernatant were removed. 300g,5min centrifugation, transfer of 100. Mu.L of supernatant/well to a new 96 well cell culture plate using a multi-channel pipette, and then following
Figure BDA0003120393180000221
The kit instruction of the HiBiTextracellular Detection System is used for detecting the killing activity of the CAR-T cells, and the method refers to the kit instruction. Cytotoxicity was calculated as follows:
cytotoxicity% = (experimental-spontaneous)/(maximum release-spontaneous) × 100%
The results are shown in FIG. 6: the cells of FOLR1-BB-B7H3-BiTE and B7H3-BiTE only indeed secrete BiTE protein and can successfully mediate the killing of SKOV3-Hibit of MOCK-T target cells.
2.5 Detection of BiTE-sectioning-CAR-T culture supernatant mediated killing effect
We collected the Supernatant (SP) of FOLR1-BB- (B7H 3-BiTE) CAR-T at day 7 for killing experiments. Plating SKOV3-Hibit cells, plating 4E3 cells in each hole of a 96-well plate, removing supernatant after the cells are adhered to the wall, and then, according to the E: T ratio of 10:1 untransduced MOCK-T cells (10 uL/well) and finally 90uL of the supernatant collected above per well, while setting the maximum release group (target cells only, but lysis treatment) and the spontaneous group (target cells only). 5% of CO2 at 37 ℃ for about 20 hours. Cell killing was tested as described above.
The results are shown in FIG. 7. FOLR1-BB-B7H3-BiTE SP contains a BITE protein, and the protein has activity and can mediate Mock-T to kill tumor cells.
The comprehensive results show that: killing was stronger with dual-target CAR-T than with single-target CAR. Namely, the CAR-T with double targets has better anti-tumor effect.
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.
Sequence listing
<110> Compound Star Kate Biotechnology Ltd
<120> preparation and application of targeted B7-H3 and FOLR1 double targeting CAR T
<130> P2021-0824
<160> 16
<170> SIPOSequenceListing 1.0
<210> 1
<211> 107
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 1
Ile Glu Val Met Tyr Pro Pro Pro Tyr Leu Asp Asn Glu Lys Ser Asn
1 5 10 15
Gly Thr Ile Ile His Val Lys Gly Lys His Leu Cys Pro Ser Pro Leu
20 25 30
Phe Pro Gly Pro Ser Lys Pro Phe Trp Val Leu Val Val Val Gly Gly
35 40 45
Val Leu Ala Cys Tyr Ser Leu Leu Val Thr Val Ala Phe Ile Ile Phe
50 55 60
Trp Val Arg Ser Lys Arg Ser Arg Leu Leu His Ser Asp Tyr Met Asn
65 70 75 80
Met Thr Pro Arg Arg Pro Gly Pro Thr Arg Lys His Tyr Gln Pro Tyr
85 90 95
Ala Pro Pro Arg Asp Phe Ala Ala Tyr Arg Ser
100 105
<210> 2
<211> 21
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 2
Met Ala Leu Pro Val Thr Ala Leu Leu Leu Pro Leu Ala Leu Leu Leu
1 5 10 15
His Ala Ala Arg Pro
20
<210> 3
<211> 45
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 3
Thr Thr Thr Pro Ala Pro Arg Pro Pro Thr Pro Ala Pro Thr Ile Ala
1 5 10 15
Ser Gln Pro Leu Ser Leu Arg Pro Glu Ala Cys Arg Pro Ala Ala Gly
20 25 30
Gly Ala Val His Thr Arg Gly Leu Asp Phe Ala Cys Asp
35 40 45
<210> 4
<211> 24
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 4
Ile Tyr Ile Trp Ala Pro Leu Ala Gly Thr Cys Gly Val Leu Leu Leu
1 5 10 15
Ser Leu Val Ile Thr Leu Tyr Cys
20
<210> 5
<211> 42
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 5
Lys Arg Gly Arg Lys Lys Leu Leu Tyr Ile Phe Lys Gln Pro Phe Met
1 5 10 15
Arg Pro Val Gln Thr Thr Gln Glu Glu Asp Gly Cys Ser Cys Arg Phe
20 25 30
Pro Glu Glu Glu Glu Gly Gly Cys Glu Leu
35 40
<210> 6
<211> 112
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 6
Arg Val Lys Phe Ser Arg Ser Ala Asp Ala Pro Ala Tyr Gln Gln Gly
1 5 10 15
Gln Asn Gln Leu Tyr Asn Glu Leu Asn Leu Gly Arg Arg Glu Glu Tyr
20 25 30
Asp Val Leu Asp Lys Arg Arg Gly Arg Asp Pro Glu Met Gly Gly Lys
35 40 45
Pro Arg Arg Lys Asn Pro Gln Glu Gly Leu Tyr Asn Glu Leu Gln Lys
50 55 60
Asp Lys Met Ala Glu Ala Tyr Ser Glu Ile Gly Met Lys Gly Glu Arg
65 70 75 80
Arg Arg Gly Lys Gly His Asp Gly Leu Tyr Gln Gly Leu Ser Thr Ala
85 90 95
Thr Lys Asp Thr Tyr Asp Ala Leu His Met Gln Ala Leu Pro Pro Arg
100 105 110
<210> 7
<211> 118
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 7
Glu Val Gln Leu Leu Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Tyr
20 25 30
Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ser Ala Ile Ser Gly Ser Gly Gly Ser Thr Tyr Tyr Ala Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr
65 70 75 80
Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Lys Arg Gly Gly Gly Gly Gln Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 8
<211> 108
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 8
Asp Ile Val Met Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Ser Ile Ser Thr Tyr
20 25 30
Leu Asn Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile
35 40 45
Tyr Ala Ala Ser Ser Leu Gln Ser Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Asp Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Leu Ala Thr Tyr Tyr Cys Gln Gln Ser Tyr Ser Thr Pro Pro
85 90 95
Trp Thr Phe Gly Gln Gly Thr Lys Val Glu Ile Lys
100 105
<210> 9
<211> 21
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 9
Met Glu Thr Asp Thr Leu Leu Leu Trp Val Leu Leu Leu Trp Val Pro
1 5 10 15
Gly Ser Thr Gly Asp
20
<210> 10
<211> 118
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 10
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Val Ser Gly Phe Thr Phe Ser Asn Tyr
20 25 30
Gly Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Thr Ile Ser Ser Gly Gly Ser Tyr Thr Tyr Tyr Pro Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr
65 70 75 80
Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ser Thr Gln Gly Ser Ser Gly Tyr Val Gly Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 11
<211> 106
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 11
Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Val Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Lys Ala Ser Gln Asp Ile Thr Asn Phe
20 25 30
Ile Gly Trp Tyr Gln His Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile
35 40 45
Ser Tyr Thr Ser Ile Leu Glu Ser Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Tyr Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Phe Ala Thr Tyr Tyr Cys Leu Gln Tyr Tyr Asn Leu Trp Thr
85 90 95
Phe Gly Gly Gly Thr Lys Val Glu Ile Lys
100 105
<210> 12
<211> 15
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 12
Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser
1 5 10 15
<210> 13
<211> 121
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 13
Asp Ile Lys Leu Gln Gln Ser Gly Ala Glu Leu Ala Arg Pro Gly Ala
1 5 10 15
Ser Val Lys Met Ser Cys Lys Thr Ser Gly Tyr Thr Phe Thr Arg Tyr
20 25 30
Thr Met His Trp Val Lys Gln Arg Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Tyr Ile Asn Pro Ser Arg Gly Tyr Thr Asn Tyr Asn Gln Lys Phe
50 55 60
Lys Asp Lys Ala Thr Leu Thr Thr Asp Lys Ser Ser Ser Thr Ala Tyr
65 70 75 80
Met Gln Leu Ser Ser Leu Thr Ser Glu Asp Ser Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Tyr Tyr Asp Asp His Tyr Cys Leu Asp Tyr Trp Gly Gln Gly
100 105 110
Thr Thr Leu Thr Val Ser Ser Val Glu
115 120
<210> 14
<211> 108
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 14
Val Asp Asp Ile Gln Leu Thr Gln Ser Pro Ala Ile Met Ser Ala Ser
1 5 10 15
Pro Gly Glu Lys Val Thr Met Thr Cys Arg Ala Ser Ser Ser Val Ser
20 25 30
Tyr Met Asn Trp Tyr Gln Gln Lys Ser Gly Thr Ser Pro Lys Arg Trp
35 40 45
Ile Tyr Asp Thr Ser Lys Val Ala Ser Gly Val Pro Tyr Arg Phe Ser
50 55 60
Gly Ser Gly Ser Gly Thr Ser Tyr Ser Leu Thr Ile Ser Ser Met Glu
65 70 75 80
Ala Glu Asp Ala Ala Thr Tyr Tyr Cys Gln Gln Trp Ser Ser Asn Pro
85 90 95
Leu Thr Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys
100 105
<210> 15
<211> 1015
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 15
Met Ala Leu Pro Val Thr Ala Leu Leu Leu Pro Leu Ala Leu Leu Leu
1 5 10 15
His Ala Ala Arg Pro Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu
20 25 30
Val Gln Pro Gly Gly Ser Leu Arg Leu Ser Cys Ala Val Ser Gly Phe
35 40 45
Thr Phe Ser Asn Tyr Gly Met Ser Trp Val Arg Gln Ala Pro Gly Lys
50 55 60
Gly Leu Glu Trp Val Ala Thr Ile Ser Ser Gly Gly Ser Tyr Thr Tyr
65 70 75 80
Tyr Pro Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser
85 90 95
Lys Asn Thr Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr
100 105 110
Ala Val Tyr Tyr Cys Ser Thr Gln Gly Ser Ser Gly Tyr Val Gly Tyr
115 120 125
Trp Gly Gln Gly Thr Leu Val Thr Val Ser Ser Gly Gly Gly Gly Ser
130 135 140
Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Asp Ile Gln Met Thr Gln
145 150 155 160
Ser Pro Ser Ser Val Ser Ala Ser Val Gly Asp Arg Val Thr Ile Thr
165 170 175
Cys Lys Ala Ser Gln Asp Ile Thr Asn Phe Ile Gly Trp Tyr Gln His
180 185 190
Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile Ser Tyr Thr Ser Ile Leu
195 200 205
Glu Ser Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp
210 215 220
Tyr Thr Leu Thr Ile Ser Ser Leu Gln Pro Glu Asp Phe Ala Thr Tyr
225 230 235 240
Tyr Cys Leu Gln Tyr Tyr Asn Leu Trp Thr Phe Gly Gly Gly Thr Lys
245 250 255
Val Glu Ile Lys Thr Thr Thr Pro Ala Pro Arg Pro Pro Thr Pro Ala
260 265 270
Pro Thr Ile Ala Ser Gln Pro Leu Ser Leu Arg Pro Glu Ala Cys Arg
275 280 285
Pro Ala Ala Gly Gly Ala Val His Thr Arg Gly Leu Asp Phe Ala Cys
290 295 300
Asp Ile Tyr Ile Trp Ala Pro Leu Ala Gly Thr Cys Gly Val Leu Leu
305 310 315 320
Leu Ser Leu Val Ile Thr Leu Tyr Cys Lys Arg Gly Arg Lys Lys Leu
325 330 335
Leu Tyr Ile Phe Lys Gln Pro Phe Met Arg Pro Val Gln Thr Thr Gln
340 345 350
Glu Glu Asp Gly Cys Ser Cys Arg Phe Pro Glu Glu Glu Glu Gly Gly
355 360 365
Cys Glu Leu Arg Val Lys Phe Ser Arg Ser Ala Asp Ala Pro Ala Tyr
370 375 380
Gln Gln Gly Gln Asn Gln Leu Tyr Asn Glu Leu Asn Leu Gly Arg Arg
385 390 395 400
Glu Glu Tyr Asp Val Leu Asp Lys Arg Arg Gly Arg Asp Pro Glu Met
405 410 415
Gly Gly Lys Pro Arg Arg Lys Asn Pro Gln Glu Gly Leu Tyr Asn Glu
420 425 430
Leu Gln Lys Asp Lys Met Ala Glu Ala Tyr Ser Glu Ile Gly Met Lys
435 440 445
Gly Glu Arg Arg Arg Gly Lys Gly His Asp Gly Leu Tyr Gln Gly Leu
450 455 460
Ser Thr Ala Thr Lys Asp Thr Tyr Asp Ala Leu His Met Gln Ala Leu
465 470 475 480
Pro Pro Arg Gly Ser Gly Ala Thr Asn Phe Ser Leu Leu Lys Gln Ala
485 490 495
Gly Asp Val Glu Glu Asn Pro Gly Pro Met Glu Thr Asp Thr Leu Leu
500 505 510
Leu Trp Val Leu Leu Leu Trp Val Pro Gly Ser Thr Gly Asp Asp Ile
515 520 525
Val Met Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly Asp Arg
530 535 540
Val Thr Ile Thr Cys Arg Ala Ser Gln Ser Ile Ser Thr Tyr Leu Asn
545 550 555 560
Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile Tyr Ala
565 570 575
Ala Ser Ser Leu Gln Ser Gly Val Pro Ser Arg Phe Ser Gly Asp Gly
580 585 590
Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro Glu Asp
595 600 605
Leu Ala Thr Tyr Tyr Cys Gln Gln Ser Tyr Ser Thr Pro Pro Trp Thr
610 615 620
Phe Gly Gln Gly Thr Lys Val Glu Ile Lys Gly Gly Gly Gly Ser Gly
625 630 635 640
Gly Gly Gly Ser Gly Gly Gly Gly Ser Glu Val Gln Leu Leu Glu Ser
645 650 655
Gly Gly Gly Leu Val Gln Pro Gly Gly Ser Leu Arg Leu Ser Cys Ala
660 665 670
Ala Ser Gly Phe Thr Phe Ser Ser Tyr Ala Met Ser Trp Val Arg Gln
675 680 685
Ala Pro Gly Lys Gly Leu Glu Trp Val Ser Ala Ile Ser Gly Ser Gly
690 695 700
Gly Ser Thr Tyr Tyr Ala Asp Ser Val Lys Gly Arg Phe Thr Ile Ser
705 710 715 720
Arg Asp Asn Ser Lys Asn Thr Leu Tyr Leu Gln Met Asn Ser Leu Arg
725 730 735
Ala Glu Asp Thr Ala Val Tyr Tyr Cys Ala Lys Arg Gly Gly Gly Gly
740 745 750
Gln Phe Asp Tyr Trp Gly Gln Gly Thr Leu Val Thr Val Ser Ser Gly
755 760 765
Gly Gly Gly Ser Asp Ile Lys Leu Gln Gln Ser Gly Ala Glu Leu Ala
770 775 780
Arg Pro Gly Ala Ser Val Lys Met Ser Cys Lys Thr Ser Gly Tyr Thr
785 790 795 800
Phe Thr Arg Tyr Thr Met His Trp Val Lys Gln Arg Pro Gly Gln Gly
805 810 815
Leu Glu Trp Ile Gly Tyr Ile Asn Pro Ser Arg Gly Tyr Thr Asn Tyr
820 825 830
Asn Gln Lys Phe Lys Asp Lys Ala Thr Leu Thr Thr Asp Lys Ser Ser
835 840 845
Ser Thr Ala Tyr Met Gln Leu Ser Ser Leu Thr Ser Glu Asp Ser Ala
850 855 860
Val Tyr Tyr Cys Ala Arg Tyr Tyr Asp Asp His Tyr Cys Leu Asp Tyr
865 870 875 880
Trp Gly Gln Gly Thr Thr Leu Thr Val Ser Ser Val Glu Gly Gly Ser
885 890 895
Gly Gly Ser Gly Gly Ser Gly Gly Ser Gly Gly Val Asp Asp Ile Gln
900 905 910
Leu Thr Gln Ser Pro Ala Ile Met Ser Ala Ser Pro Gly Glu Lys Val
915 920 925
Thr Met Thr Cys Arg Ala Ser Ser Ser Val Ser Tyr Met Asn Trp Tyr
930 935 940
Gln Gln Lys Ser Gly Thr Ser Pro Lys Arg Trp Ile Tyr Asp Thr Ser
945 950 955 960
Lys Val Ala Ser Gly Val Pro Tyr Arg Phe Ser Gly Ser Gly Ser Gly
965 970 975
Thr Ser Tyr Ser Leu Thr Ile Ser Ser Met Glu Ala Glu Asp Ala Ala
980 985 990
Thr Tyr Tyr Cys Gln Gln Trp Ser Ser Asn Pro Leu Thr Phe Gly Ala
995 1000 1005
Gly Thr Lys Leu Glu Leu Lys
1010 1015
<210> 16
<211> 3048
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 16
atggccttac cagtgaccgc cttgctcctg ccgctggcct tgctgctcca cgccgccagg 60
ccggaagtgc agctggtgga gagcggcggc ggcctggtgc agcctggcgg cagcctgaga 120
ctgagctgcg ccgtgagcgg cttcaccttc agcaattacg gcatgagctg ggtgagacag 180
gcccctggca agggcctgga gtgggtggcc acaatcagca gcggcggcag ctacacctac 240
taccctgact ccgtgaaggg cagattcacc atctccagag acaatagcaa gaataccctg 300
tacctgcaga tgaatagcct gagagccgag gacaccgccg tgtactactg cagcacccag 360
ggcagcagcg gctacgtggg ctactggggc cagggcaccc tggtgaccgt gagcagcggc 420
ggaggcggaa gtggaggcgg aggatctggc ggcggaggct ctgacatcca gatgacccag 480
agccctagca gcgtgagcgc cagcgtgggc gacagagtga ccatcacctg caaggccagc 540
caggacatca ccaatttcat cggctggtac cagcacaagc ctggcaaggc ccctaagctg 600
ctgatcagct acaccagcat cctggagagc ggcgtgccta gcagattctc cggcagcggc 660
tccggcaccg actacaccct gaccatcagc agcctgcagc ctgaggactt cgccacctac 720
tactgcctgc agtactacaa tctgtggacc ttcggcggcg gcaccaaggt ggagatcaag 780
accacgacgc cagcgccgcg accaccaaca ccggcgccca ccatcgcgtc gcagcccctg 840
tccctgcgcc cagaggcgtg ccggccagcg gcggggggcg cagtgcacac gagggggctg 900
gacttcgcct gtgatatcta catctgggcg cccttggccg ggacttgtgg ggtccttctc 960
ctgtcactgg ttatcaccct ttactgcaaa cggggcagaa agaaactcct gtatatattc 1020
aaacaaccat ttatgagacc agtacaaact actcaagagg aagatggctg tagctgccga 1080
tttccagaag aagaagaagg aggatgtgaa ctgagagtga agttcagcag gagcgcagac 1140
gcccccgcgt accagcaggg ccagaaccag ctctataacg agctcaatct aggacgaaga 1200
gaggagtacg atgttttgga caagagacgt ggccgggacc ctgagatggg gggaaagccg 1260
agaaggaaga accctcagga aggcctgtac aatgaactgc agaaagataa gatggcggag 1320
gcctacagtg agattgggat gaaaggcgag cgccggaggg gcaaggggca cgatggcctt 1380
taccagggtc tcagtacagc caccaaggac acctacgacg cccttcacat gcaggccctg 1440
ccccctcgcg gatcaggagc aacaaacttc tccttgctta aacaagcagg agatgtggaa 1500
gagaatccgg gacctatgga aacagataca ttactcttat gggtactact gttatgggta 1560
ccgggttcaa caggagatga catcgtgatg acccagtctc catcctccct gtctgcgtct 1620
gtaggggaca gagtcaccat cacttgccgg gcaagtcaga gcattagcac ctatttaaat 1680
tggtatcagc agaaaccagg gaaagcccct aaactcctga tctatgctgc atccagtttg 1740
caaagtgggg tcccatcaag gttcagtggc gatggatctg ggacagattt cactctcacc 1800
atcagcagtc tgcaacctga agatcttgca acttactact gtcaacagag ttacagcacc 1860
cctccgtgga cgttcggcca agggaccaag gtggaaatca aaggcggagg cggaagtgga 1920
ggcggaggat ctggcggcgg aggctctgag gtgcagctgt tggagtctgg gggaggcttg 1980
gtacagcctg gggggtccct gagactctcc tgtgcagcct ctggattcac ctttagcagc 2040
tatgccatga gctgggtccg ccaggctcca gggaaggggc tggagtgggt ctcagctatt 2100
agtggtagtg gtggtagcac atactacgca gactccgtga agggccggtt caccatctcc 2160
agagacaatt ccaagaacac gctgtatctg caaatgaaca gcctgagagc cgaggacacg 2220
gccgtatatt actgtgcgaa acgaggaggg gggggacaat ttgactactg gggccagggc 2280
accctggtca ccgtctcatc aggaggagga ggatcagata tcaaacttca acaatcagga 2340
gcagaacttg caagacctgg agcatcagtg aagatgtctt gcaagacgtc cggatacaca 2400
tttacaagat acacaatgca ctgggtgaaa caaagacctg gacaaggact tgaatggatc 2460
ggatacatca acccttcaag aggatacaca aactacaacc agaagttcaa ggataaagca 2520
acacttacaa cagataaatc atcatcaaca gcatacatgc aactttcatc acttacatca 2580
gaagattcag cagtgtacta ctgcgcaaga tactacgatg atcactactg ccttgattac 2640
tgggggcagg gcacaacact tacagtgtca tcagtggaag gtggctcggg tggctccgga 2700
ggaagcggag ggtcaggagg cgtcgacgat atccaactta cacaatcacc tgcaatcatg 2760
tcagcatcac ctggagagaa ggttactatg acatgcagag catcatcatc agtgtcatac 2820
atgaactggt accaacagaa gtccggaacg tcgcctaagc ggtggatcta cgatacatca 2880
aaggtagcct caggagtgcc ttacagattc tccggctcag gaagtggtac tagctattcg 2940
cttacaatct catcaatgga agcagaagat gcagcaacat actactgcca acaatggtca 3000
tcaaaccctc ttacatttgg agcaggaaca aagttagagc ttaaataa 3048

Claims (10)

1. A chimeric antigen receptor CAR-immune cell, wherein said CAR-immune cell comprises the following elements:
(a) A first CAR whose extracellular binding domain contains a first binding element that targets a first target protein and whose extracellular binding domain contains or does not contain a second binding element that targets a second target protein;
(b1) Optionally a second CAR whose extracellular binding domain comprises a second binding element that targets a second target protein;
(b2) Optionally a nucleic acid sequence encoding a BiTE, wherein the BiTE targets a second target protein;
wherein, when the extracellular binding domain of the first CAR does not contain a second binding element that targets a second target protein, the CAR-immune cell contains the (b 1) and/or (b 2) elements;
and, the first target protein is folate receptor1 (FOLR 1) and the second target protein is B7-H3; alternatively, the first target protein is B7-H3, and the second target protein is folate receptor1 (FOLR 1).
2. The CAR-immune cell of claim 1, wherein the first CAR or the second CAR has the structure of formula I:
L-EB-H-TM-C-CD3ζ (I)
in the formula (I), the compound is shown in the specification,
each "-" is independently a linker peptide or a peptide bond;
l is an optional signal peptide sequence;
EB is the extracellular binding domain;
h is an optional hinge region;
TM is a transmembrane domain;
c is a costimulatory signal molecule;
CD3 ζ is a cytoplasmic signaling sequence derived from CD3 ζ.
3. The CAR-immune cell of claim 1, wherein when the second target protein is B7-H3, the BiTE is a CD3 ScFv.
4. A nucleic acid molecule encoding element (a), (b 1) and/or (b 2) in a CAR-immune cell of claim 1.
5. A vector comprising the nucleic acid molecule of claim 4.
6. A host cell comprising the vector or chromosome of claim 5 into which has been integrated an exogenous nucleic acid molecule of claim 4.
7. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and the CAR-immune cell of claim 1, the nucleic acid molecule of claim 4, the vector of claim 5, or the host cell of claim 6.
8. Use of the CAR-immune cell of claim 1, the nucleic acid molecule of claim 4, the vector of claim 5, or the host cell of claim 6, for the preparation of a medicament or formulation for treating a tumor.
9. The use according to claim 8, wherein the tumour is ovarian cancer.
10. A method of making the CAR-immune cell of claim 1, comprising the steps of: transferring the nucleic acid molecule of claim 4 or the vector of claim 5 into an immune cell, thereby obtaining the CAR-immune cell.
CN202110674194.3A 2021-06-17 2021-06-17 Preparation and application of targeting B7-H3 and FOLR1 double targeting CAR T Pending CN115491358A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110674194.3A CN115491358A (en) 2021-06-17 2021-06-17 Preparation and application of targeting B7-H3 and FOLR1 double targeting CAR T

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110674194.3A CN115491358A (en) 2021-06-17 2021-06-17 Preparation and application of targeting B7-H3 and FOLR1 double targeting CAR T

Publications (1)

Publication Number Publication Date
CN115491358A true CN115491358A (en) 2022-12-20

Family

ID=84465384

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110674194.3A Pending CN115491358A (en) 2021-06-17 2021-06-17 Preparation and application of targeting B7-H3 and FOLR1 double targeting CAR T

Country Status (1)

Country Link
CN (1) CN115491358A (en)

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109680004A (en) * 2019-01-09 2019-04-26 上海美丽人生医疗科技有限公司 The double target spot CAR carriers and its construction method of joint EpCAM and MSLN single-chain antibody and the application in oophoroma
CN109777784A (en) * 2019-02-22 2019-05-21 上海尚泰生物技术有限公司 It is a kind of to enhance the Chimeric antigen receptor carrier construction method migrated to tumor locus and application
CN110520524A (en) * 2017-04-14 2019-11-29 综合医院公司 The Chimeric antigen receptor T cell of target tumor microenvironment
CN111234032A (en) * 2019-12-12 2020-06-05 首都医科大学附属北京妇产医院 Double-target chimeric antigen receptor for treating ovarian cancer and preparation method and application thereof
CN111777686A (en) * 2020-07-13 2020-10-16 中国人民解放军陆军军医大学第一附属医院 FOLR1-MSLN dual targeting CAR-T cells, chimeric antigen receptors, and vectors for treating ovarian cancer
CN111954681A (en) * 2018-03-13 2020-11-17 东莞凡恩世生物医药有限公司 Anti-folate receptor 1 antibodies and uses thereof
CN111971053A (en) * 2018-02-12 2020-11-20 综合医院公司 Chimeric antigen receptors targeting tumor microenvironment
CN111995688A (en) * 2020-08-13 2020-11-27 金鑫 Bispecific chimeric antigen receptor targeting CD123 and NKG2D ligands and application thereof
WO2021027687A1 (en) * 2019-08-14 2021-02-18 博源润生医药(杭州)有限公司 Chimeric antigen receptor and immune effector cell expressing chimeric antigen receptor
CN112778427A (en) * 2021-01-29 2021-05-11 武汉思安医疗技术有限公司 Bispecific CS1-BCMA CAR-T cells and uses thereof
US20210137977A1 (en) * 2018-06-01 2021-05-13 University Of Southern California Diverse antigen binding domains, novel platforms and other enhancements for cellular therapy
CN113461818A (en) * 2021-06-30 2021-10-01 徐州医科大学 CD 276-targeted fully human antibody scFv, chimeric antigen receptor, engineered immune cell and preparation method thereof
WO2021213478A1 (en) * 2020-04-22 2021-10-28 复星凯特生物科技有限公司 Anti-human b7-h3 monoclonal antibody and application thereof
CN114686436A (en) * 2020-12-31 2022-07-01 复星凯特生物科技有限公司 Preparation and application of FSHR and FOLR1 targeting double targeting target point CAR T
WO2023129995A2 (en) * 2021-12-30 2023-07-06 ST. JUDE CHILDREN'S RESEARCH HOSPITAL,Inc. Chimeric antigen receptors comprising a pdz binding motif
WO2023164698A2 (en) * 2022-02-27 2023-08-31 Health Research, Inc. FOLATE RECEPTOR ALPHA -TARGETING BI-SPECIFIC T CELL ENGAGERS (BiTEs) AND USES THEREOF

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110520524A (en) * 2017-04-14 2019-11-29 综合医院公司 The Chimeric antigen receptor T cell of target tumor microenvironment
CN111971053A (en) * 2018-02-12 2020-11-20 综合医院公司 Chimeric antigen receptors targeting tumor microenvironment
CN111954681A (en) * 2018-03-13 2020-11-17 东莞凡恩世生物医药有限公司 Anti-folate receptor 1 antibodies and uses thereof
US20210137977A1 (en) * 2018-06-01 2021-05-13 University Of Southern California Diverse antigen binding domains, novel platforms and other enhancements for cellular therapy
CN109680004A (en) * 2019-01-09 2019-04-26 上海美丽人生医疗科技有限公司 The double target spot CAR carriers and its construction method of joint EpCAM and MSLN single-chain antibody and the application in oophoroma
CN109777784A (en) * 2019-02-22 2019-05-21 上海尚泰生物技术有限公司 It is a kind of to enhance the Chimeric antigen receptor carrier construction method migrated to tumor locus and application
WO2021027687A1 (en) * 2019-08-14 2021-02-18 博源润生医药(杭州)有限公司 Chimeric antigen receptor and immune effector cell expressing chimeric antigen receptor
CN111234032A (en) * 2019-12-12 2020-06-05 首都医科大学附属北京妇产医院 Double-target chimeric antigen receptor for treating ovarian cancer and preparation method and application thereof
WO2021213478A1 (en) * 2020-04-22 2021-10-28 复星凯特生物科技有限公司 Anti-human b7-h3 monoclonal antibody and application thereof
CN111777686A (en) * 2020-07-13 2020-10-16 中国人民解放军陆军军医大学第一附属医院 FOLR1-MSLN dual targeting CAR-T cells, chimeric antigen receptors, and vectors for treating ovarian cancer
CN111995688A (en) * 2020-08-13 2020-11-27 金鑫 Bispecific chimeric antigen receptor targeting CD123 and NKG2D ligands and application thereof
CN114686436A (en) * 2020-12-31 2022-07-01 复星凯特生物科技有限公司 Preparation and application of FSHR and FOLR1 targeting double targeting target point CAR T
CN112778427A (en) * 2021-01-29 2021-05-11 武汉思安医疗技术有限公司 Bispecific CS1-BCMA CAR-T cells and uses thereof
CN113461818A (en) * 2021-06-30 2021-10-01 徐州医科大学 CD 276-targeted fully human antibody scFv, chimeric antigen receptor, engineered immune cell and preparation method thereof
WO2023129995A2 (en) * 2021-12-30 2023-07-06 ST. JUDE CHILDREN'S RESEARCH HOSPITAL,Inc. Chimeric antigen receptors comprising a pdz binding motif
WO2023164698A2 (en) * 2022-02-27 2023-08-31 Health Research, Inc. FOLATE RECEPTOR ALPHA -TARGETING BI-SPECIFIC T CELL ENGAGERS (BiTEs) AND USES THEREOF

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BRYAN D. CHOI等: "CAR-T cells secreting BiTEs circumvent antigen escape without detectable toxicity", NATURE BIOTECHNOLOGY, vol. 37, 22 July 2019 (2019-07-22), pages 1049 - 1058, XP036878166, DOI: 10.1038/s41587-019-0192-1 *
JULIE DAIGRE等: "Preclinical Evaluation of Novel Folate Receptor 1-Directed CAR T Cells for Ovarian Cancer", CANCERS, vol. 16, 12 January 2024 (2024-01-12), pages 1 - 21 *
ZHEN LIANG等: "Tandem CAR-T cells targeting FOLR1 and MSLN enhance the antitumor effects in ovarian cancer", INT. J. BIOL. SCI., vol. 17, 22 October 2021 (2021-10-22), pages 4365 - 4376 *
梁振: "靶向叶酸受体α和间皮素串联型CAR-T的构建及其在上皮性卵巢癌的应用", 中国优秀硕士学位论文全文数据库(电子期刊)医药卫生科技辑, no. 01, 15 January 2021 (2021-01-15), pages 1 - 82 *

Similar Documents

Publication Publication Date Title
US11142581B2 (en) BCMA-targeted chimeric antigen receptor as well as preparation method therefor and application thereof
CN110760007B (en) CD7-CAR-T cell and preparation and application thereof
CN109593721B (en) Engineered immune cells targeting human mesothelin with suicide gene switch
CN112778427B (en) Bispecific CS1-BCMA CAR-T cells and uses thereof
CN109306016B (en) NKG2D-CAR-T cells co-expressing cytokine IL-7 and uses thereof
CN109575143B (en) Bispecific CD20-CD19-CAR and application thereof
CN114957475A (en) Monoclonal antibody against B7-H3 and application thereof in cell therapy
CN113784733A (en) BCMA-targeted engineered immune cells and uses thereof
CN112500492B (en) Chimeric antigen receptor and application thereof
CN113784732A (en) BCMA-targeted engineered immune cells and uses thereof
CN113980137A (en) Chimeric antigen receptor containing CD3 epsilon intracellular basic amino acid enrichment region motif and application thereof
CN113755448A (en) Engineered immune cells jointly expressing CCR2b and CD40L, and preparation and application thereof
CN112522208A (en) Transgenic tumor infiltrating lymphocyte and application thereof
CN114686436A (en) Preparation and application of FSHR and FOLR1 targeting double targeting target point CAR T
WO2023016524A1 (en) Combined her2 and meso dual-target car-t vector, construction method therefor and application thereof in cancer
CN109897114B (en) CD 47-targeted engineered immune cells with suicide gene switch
CN117024598A (en) Long-acting Meso-B7H3 double-target chimeric antigen receptor and application thereof
CN114853893A (en) Target GPC3 chimeric antigen receptor T cell and application thereof
JP2023521218A (en) CD22-targeted chimeric antigen receptor, its preparation method, and its application
CN114685683A (en) GD 2-targeted CAR-T cells and preparation and application thereof
CN115491358A (en) Preparation and application of targeting B7-H3 and FOLR1 double targeting CAR T
CN115572715A (en) Preparation and application of FOLR1 and HER2 targeting double targeting CAR T
CN115491360A (en) Preparation and application of targeted NKG2D ligand and FOLR1 double targeting target point CAR T
CN116444669B (en) Humanized antibodies targeting BCMA CAR-T cells
CN115975056B (en) Construction of TSHR-targeted CAR-T cells using the native protein TSH as an antigen binding site

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
CB03 Change of inventor or designer information
CB03 Change of inventor or designer information

Inventor after: Zhou Chunyan

Inventor after: Xu Min

Inventor after: Li Xiuyun

Inventor after: Zhang Yu

Inventor after: Cai Fengcan

Inventor after: Gong Jian

Inventor before: Zhou Chunyan

Inventor before: Xu Min

Inventor before: Liu Xiuyun

Inventor before: Zhang Yu

Inventor before: Cai Fengcan

Inventor before: Gong Jian