WO2023161845A1 - Gpc3-targeting chimeric antigen receptor and use thereof - Google Patents

Gpc3-targeting chimeric antigen receptor and use thereof Download PDF

Info

Publication number
WO2023161845A1
WO2023161845A1 PCT/IB2023/051678 IB2023051678W WO2023161845A1 WO 2023161845 A1 WO2023161845 A1 WO 2023161845A1 IB 2023051678 W IB2023051678 W IB 2023051678W WO 2023161845 A1 WO2023161845 A1 WO 2023161845A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
car
nucleic acid
cell
present
Prior art date
Application number
PCT/IB2023/051678
Other languages
French (fr)
Chinese (zh)
Inventor
许元剑
郭志刚
Original Assignee
南京蓝盾生物科技有限公司
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 南京蓝盾生物科技有限公司 filed Critical 南京蓝盾生物科技有限公司
Publication of WO2023161845A1 publication Critical patent/WO2023161845A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material

Definitions

  • the present invention relates to the field of cellular immunotherapy, in particular, to a chimeric antigen receptor targeting Glypican 3 (GPC3) and its application.
  • GPC3 Glypican 3
  • CAR-T Chimeric Antigen Receptor T cell
  • Glypican 3 (GPC3) protein is a heparan sulfate glycoprotein on the surface of the cell membrane, expressed in some tissues during infancy, especially the liver and kidney, and is a cell related to organ formation. Extracellular matrix protein; The expression of GPC3 is hardly observed in adult tissues, but it is highly expressed in various cancer tissues such as hepatocellular carcinoma, melanoma, ovarian clear cell carcinoma, and lung squamous cell carcinoma.
  • the first aspect of the present invention provides a chimeric antigen receptor (CAR) construct targeting GPC3, the antigen-binding domain of the chimeric antigen receptor comprises a heavy chain variable region and a light chain variable region, wherein, the heavy chain variable region includes the following CDRs:
  • the CDR3 shown in SEQ ID NO: 21; and the light chain variable region includes the following complementarity determining region CDR:
  • the heavy chain variable region and the light chain variable region of the antigen-binding domain are derived from murine, human, or humanized antibodies.
  • said H is a hairpin chain region derived from CD8.
  • the amino acid sequence of H is shown in SEQ ID NO: 9.
  • the TM is a transmembrane region of a protein selected from the group consisting of ICOS, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, GD2, CD33, CD37, CD64, CD80,
  • the second aspect of the present invention provides an isolated nucleic acid molecule encoding the chimeric antigen receptor (CAR) construct described in the first aspect of the present invention.
  • the nucleic acid molecule is shown in SEQ ID NO: 18.
  • a nucleotide sequence encoding a chimeric antigen receptor is provided, the main part of the chimeric antigen receptor includes an antigen binding domain, a transmembrane domain and a costimulatory signal transduction region .
  • the GPC3-specific antibody coding gene is connected to the modified cloning site region of the CAR backbone containing the transmembrane structural gene and costimulatory signal gene to obtain the CAR gene.
  • the antigen-binding domain can specifically bind the tumor-specific antigen GPC3, and activate the NK cell through the transmembrane domain and costimulatory signal transduction region.
  • the antigen-binding domain is an antibody that specifically binds GPC3 or an antigen-binding fragment thereof, and the antigen-binding fragment is Fab or scFv o
  • the original binding domain It is scFv
  • the nucleic acid sequence of its heavy chain variable region is shown in SEQ ID NO: 4
  • the nucleic acid sequence of its light chain variable region is shown in SEQ ID NO: 8.
  • the vector is selected from the group consisting of DNA, RNA, plasmid, lentiviral vector, adenoviral vector, adeno-associated viral vector (AAV), retroviral vector, transposon, or a combination thereof .
  • the vector is selected from the group consisting of plasmids and viral vectors.
  • the vector is in the form of virus particles.
  • the vector is a lentiviral vector.
  • the fourth aspect of the present invention provides a host cell containing the vector according to the third aspect of the present invention, or an exogenous nucleic acid molecule as described in the second aspect of the present invention integrated in the chromosome, Or express the CAR construct as described in the first aspect of the present invention.
  • the host cells include eukaryotic cells and prokaryotic cells.
  • the host cells include Escherichia coli.
  • the sixth aspect of the present invention provides a preparation comprising the CAR construct according to the first aspect of the present invention, the isolated nucleic acid molecule according to the second aspect of the present invention, the third aspect of the present invention
  • the carrier or the immune cell according to the fifth aspect of the present invention, and a pharmaceutically acceptable carrier is a liquid formulation.
  • the dosage form of the preparation is injection.
  • the concentration of immune cells (GPC3 CAR-T cells) in the preparation is IX
  • the preparation may include a buffer such as neutral buffered saline , sulfate buffered saline, etc.; carbohydrates such as glucose, mannose, sucrose or dextran, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (eg eg, aluminum hydroxide); and preservatives.
  • the formulations of the invention are preferably formulated for intravenous administration.
  • the preparation further includes a second anti-tumor active ingredient, preferably a second antibody or a chemotherapeutic agent.
  • the chemotherapeutic agent is selected from the group consisting of docetaxel, calipers, or a combination thereof.
  • the preparation is a pharmaceutical composition.
  • the effect-to-target ratio of GPC3 CAR-T cells to tumor cells is (0.1-20): 1, preferably, the effect-to-target ratio is (1-4): 1.
  • the seventh aspect of the present invention provides the CAR construct according to the first aspect of the present invention, the nucleic acid molecule according to the second aspect of the present invention, the vector according to the third aspect of the present invention, the fifth aspect of the present invention
  • the use of the immune cells according to the aspect, or the preparation according to the sixth aspect of the present invention is used to prepare a drug for preventing and/or treating tumor or cancer.
  • the tumor or cancer highly (over) expresses GPC3.
  • the tumor or cancer is a solid tumor.
  • the solid tumor is selected from the group consisting of hepatocellular carcinoma, melanoma, ovarian clear cell carcinoma, lung squamous cell carcinoma, breast cancer, renal carcinoma, cholangiocarcinoma or a combination thereof.
  • the tumor or cancer is hepatocellular carcinoma.
  • the tumor or cancer is a liver cell that highly (over)expresses GPC3.
  • the engineered immune cells are CART cells or CAR-NK cells.
  • the method further includes the step of testing the function and effectiveness of the obtained engineered immune cells.
  • the ninth aspect of the present invention provides a method for treating tumor or cancer, comprising administering to a subject in need an effective amount of the immune cells described in the fifth aspect of the present invention, or the preparation described in the sixth aspect of the present invention .
  • the tumor or cancer highly (over) expresses GPC3.
  • the tumor or cancer is a solid tumor.
  • the solid tumor is selected from the group consisting of hepatocellular carcinoma, melanoma, ovarian clear cell carcinoma, lung squamous cell carcinoma, breast cancer, kidney cancer, cholangiocarcinoma or a combination thereof
  • the tumor or cancer is hepatocellular carcinoma.
  • FIG. 1 shows a schematic diagram of the structure of the GPC3-targeting chimeric antigen receptor GC52BB& of the present invention.
  • Figure 2 shows the construction and detection of HuH7-Luc>HepG2-Luc cells.
  • Figure 3 shows the detection of GC52BB C positive rate.
  • Figure 4 shows that GC52BB V kills HuH7-luc cells in vitro; the left panel shows the killing ability of GC52BB E on HuH7Tuc cells when E/T (effect-to-target ratio) is 4; Target ratio) is 1, the killing ability of GC52BB V on HuH7-luc cells.
  • Figure 5 shows that GC52BB C kills HepG2-luc cells in vitro; the left picture shows the killing ability of GC52BB to HepG2Tuc cells when E/T (effect-to-target ratio) is 4; Target ratio) is 1, GC52BB has the ability to kill HepG2-luc cells.
  • Figure 6 shows the anti-tumor effect of GC52BB V in vivo. DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS After extensive and in-depth research, the present inventors unexpectedly discovered for the first time a chimeric antigen receptor that targets and recognizes GPC3, its encoding nucleotides, and GPC3 CAR-T cells expressing the CAR and its preparation method and applications.
  • the CART cells of the present invention are specific to GPC3, and can specifically recognize and kill tumors, especially solid tumors (such as hepatocellular carcinoma) that highly express or overexpress GPC3, and have more efficient tumor killing activity, at low doses (approximately It is only 1/5 of the usual dose in the prior art to achieve a good anti-tumor effect, and its properties are stable, and it can be mass-produced and prepared. On this basis, the present invention has been accomplished. In order to better understand the present invention, certain terms are defined below. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
  • antigen-binding domain and “single-chain antibody fragment” all refer to a Fab fragment, Fab' fragment, F(ab 7 ) 2 fragment, or a single Fv fragment with antigen-binding activity.
  • Fv antibodies contain antibody heavy chain variable regions, light chain variable regions, but no constant region, and the smallest antibody fragment with all antigen-binding sites.
  • Fv antibodies also contain a polypeptide linker between the VH and VL domains and are capable of forming antigenic Combine the desired structure.
  • the antigen-binding domain is usually scFv (single-chain variable fragment) o
  • the size of scFv is generally 1/6 of a complete antibody.
  • a single chain antibody is preferably a sequence of one amino acid chain encoded by one nucleotide chain.
  • the scFv comprises an antibody that specifically recognizes GPC3, preferably a humanized single-chain antibody.
  • CAR can be designed to include the transmembrane domain fused to the extracellular domain of the CAR.
  • a transmembrane domain naturally associated with one of the domains in the CAR is used.
  • transmembrane domains may be selected, or modified by amino acid substitutions, to avoid binding such domains to transmembrane domains of the same or different surface membrane proteins, thereby minimizing interaction with the receptor complex.
  • antibody refers to an immunoglobulin molecule that specifically binds to an antigen.
  • Antibodies can be intact immunoglobulins, derived from natural sources or from recombinant sources, and can be immunoreactive portions of intact immunoglobulins.
  • Antibodies are typically tetramers of immunoglobulin molecules.
  • Antibodies in the present invention can exist in various forms, including, for example, polyclonal antibodies, monoclonal antibodies, Fv, Fab, and F (ab) 2 , as well as single-chain antibodies and humanized antibodies, etc.
  • antibody fragment refers to a portion of an intact antibody and refers to the antigenically determining variable regions of an intact antibody.
  • antibody fragments include, but are not limited to, Fab, Fab', F(ab')2 and Fv fragments, linear antibodies, scFv antibodies and multispecific antibodies formed from antibody fragments.
  • encoding nucleotides or “encoding nucleic acids” include all nucleotide sequences that are degenerate versions of each other and encode the same amino acid sequence.
  • a nucleotide sequence encoding a protein may include introns.
  • lentivirus refers to a genus of the Retroviridae family that is capable of efficiently infecting aperiodic and post-mitotic cells; they can transfer significant amounts of genetic information into the host cell's DNA, so that they are the most suitable vectors for gene delivery. one of the effective methods.
  • promoter is defined as a DNA sequence that is recognized by or directs the synthetic machinery of a cell, required to initiate specific transcription of a polynucleotide sequence.
  • specifically binds refers to recognition of a specific antigen but substantially no recognition or binding of other molecules in the sample.
  • vector is a composition of matter that includes an isolated nucleic acid and that can be used to deliver the isolated nucleic acid to the interior of a cell.
  • Many vectors are known in the art, including but not limited to linear polynucleotides, polynucleotides associated with ionic or amphiphilic compounds, plasmids and viruses.
  • vector includes autonomously replicating plasmids or viruses.
  • the term should also be construed to include the facilitation of transfer of nucleic acid into Cellular non-plasmid and non-viral compounds such as, for example, polylysine compounds, liposomes and the like.
  • viral vectors include, but are not limited to, adenoviral vectors, adeno-associated viral vectors, retroviral vectors, and the like.
  • cancer is defined as a disease characterized by the rapid and uncontrolled growth of abnormal cells. Cancer cells can spread locally or to other parts of the body through the bloodstream and lymphatic system. Examples of various cancers include, but are not limited to, breast cancer, prostate cancer, ovarian cancer, cervical cancer, skin cancer, pancreatic cancer, colorectal cancer, kidney cancer, liver cancer, brain cancer, lymphoma, leukemia, lung cancer, and the like.
  • the design of CARs has gone through the following process:
  • the first-generation CAR has only one intracellular signaling component CD3 & or Fc ⁇ RI molecule, because there is only one activation domain in the cell, so it can only cause transient T cell proliferation and less
  • the secretion of cytokines does not provide long-term T cell proliferation signals and sustained anti-tumor effects in vivo, so good clinical efficacy has not been achieved.
  • the second-generation CARs introduce a co-stimulatory molecule based on the original structure, such as CD28, 4-1BB, OX40, and ICOS. Compared with the first-generation CARs, the function is greatly improved, and the persistence of CAR-T cells and the ability to protect tumor cells are further enhanced. lethality.
  • the chimeric antigen receptor (CAR) of the present invention is a second-generation CAR, including an extracellular domain, a transmembrane domain, and an intracellular domain.
  • the extracellular domain includes target-specific binding elements (also known as antigen-binding structures area).
  • the intracellular domain includes the co-stimulatory signaling domain and the chain portion.
  • a co-stimulatory signaling region refers to a portion of an intracellular domain that includes co-stimulatory molecules.
  • Costimulatory molecules are cell surface molecules other than antigen receptors or their ligands that are required for an efficient response of lymphocytes to antigens.
  • 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.
  • the term "linker” generally refers to any oligopeptide or polypeptide that functions to link a transmembrane domain to the extracellular or cytoplasmic domain of a polypeptide chain.
  • Linkers may comprise 0-300 amino acids, preferably 2 to 100 amino acids and most preferably 3 to 50 amino acids.
  • the extracellular domain of the CAR provided by the present invention includes an antigen-binding domain targeting GPC3.
  • the CAR of the present invention can perform antigen recognition based on antigen binding specificity.
  • it binds to its cognate antigen, it affects tumor cells, causing tumor cells not to grow, being induced to die, or otherwise affected, and resulting in a reduction or elimination of the patient's tumor burden.
  • the antigen binding domain is preferably fused to an intracellular domain from one or more of a co-stimulatory molecule and a chain.
  • the nucleic acid sequence of the vector encoding the desired molecule can be obtained using recombinant methods known in the art, such as, for example, by screening a library from cells expressing the gene, by obtaining the gene from a vector known to include the gene, or by using standard technology, directly isolated from cells and tissues containing the gene. Alternatively, the gene of interest can be produced synthetically.
  • the present invention also provides a vector into which an expression cassette of the present invention is inserted.
  • Vectors derived from retroviruses such as lentiviruses are suitable tools for long-term gene transfer because they allow long-term, stable integration of the transgene and its propagation in daughter cells.
  • Lentiviral vectors have an advantage over vectors derived from oncogenic retroviruses, such as murine leukemia virus, because they can transduce non-proliferating cells, such as hepatocytes. They also have the advantage of low immunogenicity.
  • the expression cassette or nucleic acid sequence of the present invention is usually operably linked to a promoter and incorporated into an expression vector. This vector is suitable for replication and integration in eukaryotic cells.
  • a typical cloning vector contains transcriptional and translational terminators, an initial sequence and a promoter useful for regulating the expression of the desired nucleic acid sequence.
  • the expression constructs of the invention can also be used in nucleic acid immunization and gene therapy using standard gene delivery protocols.
  • the present invention provides gene therapy vectors.
  • the nucleic acid can be cloned into many types of vectors.
  • the nucleic acid can be cloned into vectors including, but not limited to, plasmids, phagemids, phage derivatives, animal viruses, and cosmids.
  • Particular vectors of interest include expression vectors, replication vectors, probe generation vectors and sequencing vectors.
  • expression vectors can be provided to cells in the form of viral vectors.
  • Viruses that can be used as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpesviruses, and lentiviruses.
  • suitable vectors contain an origin of replication functional in at least one organism, a promoter sequence, convenient restriction enzyme sites, and one or more selectable markers (eg, WO01/96584; WO01/29058; and US Patent No. 6,326,193).
  • retroviruses provide a convenient platform for gene delivery systems.
  • the gene of choice can be inserted into a vector and packaged into retroviral particles using techniques known in the art.
  • the recombinant virus can then be isolated and delivered to subject cells in vivo or ex vivo.
  • retroviral systems are known in the art.
  • an adenoviral vector is used.
  • Many adenoviral vectors are known in the art.
  • lentiviral vectors are used. Additional promoter elements, such as enhancers, can regulate the frequency of transcription initiation.
  • promoters typically these are located in the 30-110 bp region upstream of the initiation site, although it has recently been shown that many promoters also contain functional elements downstream of the initiation site.
  • the spacing between promoter elements is often flexible in order to preserve promoter function when elements are inverted or moved relative to one another.
  • tk thymidine kinase
  • the spacing between promoter elements can be increased by 50 bp before activity begins to decline.
  • individual elements can act cooperatively or independently to initiate transcription.
  • An example of a suitable promoter is the immediate early cytomegalovirus (CMV) promoter sequence.
  • CMV immediate early cytomegalovirus
  • the promoter sequence is a strong constitutive promoter sequence capable of driving high-level expression of any polynucleotide sequence operably linked thereto.
  • a suitable promoter is elongation growth factor-1 ⁇ (EF-1 ⁇ ) .
  • EF-1 ⁇ elongation growth factor-1 ⁇
  • other constitutive promoter sequences can also be used, including but not limited to the simian virus 40 (SV40) early promoter, mouse Mammary cancer virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, avian leukemia virus promoter, Epstein-Barr virus immediate early promoter, Lu Steiner's sarcoma virus promoter, and human gene promoters, such as but not limited to actin promoter, myosin promoter, heme promoter and creatine kinase promoter.
  • SV40 simian virus 40
  • MMTV mouse Mammary cancer virus
  • HMV human immunodeficiency virus
  • inducible promoters are also contemplated as part of the invention.
  • the use of an inducible promoter provides a molecular switch capable of turning on expression of a polynucleotide sequence operably linked to an inducible promoter when such expression is desired, or turning off expression when expression is not desired.
  • inducible promoters include, but are not limited to, the metallothionein promoter, the glucocorticoid promoter, the progesterone promoter, and the tetracycline promoter.
  • the expression vector introduced into the cell may also contain either or both of a selectable marker gene or a reporter gene, so as to seek transfected or infected cell populations from viral vectors Identification and selection of expressing cells.
  • selectable markers can be carried on a single piece of DNA and used in a co-transfection procedure. Flanking both selectable marker and reporter gene is well known in the field. See, eg, Sambrook et al. (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York) . o A preferred method for introducing polynucleotides into host cells is calcium phosphate transfection.
  • Biological methods for introducing polynucleotides of interest into host cells include the use of DNA and RNA vectors.
  • Viral vectors especially retroviral vectors, have become the most widely used method for inserting genes into mammalian, eg human, cells.
  • Other viral vectors can be derived from lentiviruses, poxviruses, herpes simplex virus I, adenoviruses, and adeno-associated viruses, among others. See, eg, U.S. Patent Nos. 5,350,674 and 5,585,362.
  • Chemical means for 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 lipid-based systems.
  • colloidal dispersion systems such as macromolecular complexes, nanocapsules, microspheres, beads
  • lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and lipid-based systems.
  • plastid An exemplary colloidal system for use as an in vitro and in vivo delivery vehicle is a liposome (eg, an artificial membrane vesicle). Where a non-viral delivery system is used, an exemplary delivery vehicle is liposomes.
  • the use of lipid formulations is contemplated for introducing nucleic acids into host cells (in vitro, ex vivo, or in vivo).
  • the nucleic acid can be associated with a lipid.
  • Lipid-associated nucleic acids can be encapsulated into the aqueous interior of liposomes, interspersed within the lipid bilayer of liposomes, attached via linker molecules associated with both liposomes and oligonucleotides into liposomes, entrapped in liposomes, complexed with liposomes, dispersed in a solution containing lipids, mixed with lipids, associated with lipids, contained in lipids as a suspension, contained in micelles or complexed with micelles, Or otherwise associated with lipids.
  • the lipid, lipid/DNA or lipid/expression vector associated with the composition is not limited to any particular structure in solution.
  • lipids are fatty substances, which may be naturally occurring or synthetic lipids.
  • lipids include fat droplets, which occur naturally in the cytoplasm as well as compounds comprising long-chain aliphatic cores and their derivatives such as fatty acids, alcohols, amines, aminoalcohols, and aldehydes.
  • the vector is a lentiviral vector.
  • the present invention includes therapeutic applications of cells (eg T cells) transduced with a lentiviral vector (LV) encoding a CAR of the present invention.
  • the transduced T cells can target the tumor cell marker GPC3, activate T cells cooperatively, and cause T cell immune responses, thereby significantly improving their killing efficiency against tumor cells. Therefore, the present invention also provides a method for stimulating a T cell-mediated immune response to a target cell population or tissue in a mammal, comprising the following steps: administering the CART cells of the present invention to the mammal.
  • the present invention includes a type of cell therapy, in which a patient's own T cells (or a heterogeneous donor) are isolated, activated and genetically modified to produce CAR-T cells, and then injected into the same patient.
  • a patient's own T cells or a heterogeneous donor
  • the probability of suffering from graft-versus-host disease is extremely low, and the antigen is recognized by T cells in an MHC-free manner.
  • one CAR-T can treat all cancers expressing the antigen.
  • CAR-T cells are able to replicate in vivo, resulting in long-term persistence that can lead to sustained tumor control.
  • CAR-T cells of the invention can undergo robust in vivo NK cell expansion for extended amounts of time.
  • the CAR-mediated immune response can be part of an adoptive immunotherapy step in which CAR-modified T cells induce an immune response specific for the antigen-binding domain in the CAR.
  • CAR-modified T cells induce an immune response specific for the antigen-binding domain in the CAR.
  • anti-GPC3 CART cells elicit a specific immune response against GPC3-positive cells.
  • the data disclosed herein specifically discloses lentiviral vectors comprising anti-GPC3 scFv, CD8 chain and CD8 transmembrane and intracellular regions, 4-1BB intracellular region and CD3 & signaling domain, the present invention should be considered Interpretation is to include any number of variations to each of the construct components.
  • Treatable cancers include non-solid tumors (such as hematological tumors, eg leukemias and lymphomas) or solid tumors, especially solid tumors.
  • the types of cancer treated with the CAR of the present invention include, but are not limited to, carcinoma, blastoma and sarcoma, and certain leukemia or lymphoid malignancies, benign and malignant tumors, and malignant tumors, such as sarcoma, carcinoma and melanoma. Also includes adult tumors/cancers and childhood tumors/cancers.
  • Hematological cancers are cancers of the blood or bone marrow.
  • hematological (or hematogenous) cancers include leukemias, including acute leukemias such as acute lymphoblastic leukemia, acute myeloid leukemia, acute myelogenous leukemia, and myeloblastic, promyelocytic, myelomonocytic , monocytic and erythroleukemia), chronic leukemias such as chronic myeloid (granulocytic) leukemia, chronic myelogenous leukemia and chronic lymphocytic leukemia), polycythemia vera, lymphoma, Hodgkin's disease, non-Hodgkin's lymphoma (indolent and high-grade forms), multiple myeloma, Waldenstrom's macroglobulin leukemia, heavy chain disease, myelodysplastic syndrome, hairy cell leukemia, and myelodysplasia.
  • acute leukemias such as acute lymphoblastic leukemia, acute myeloid leukemia,
  • Solid tumors are abnormal masses of tissue that usually do not contain cysts or areas of fluid. Solid tumors can be benign or malignant. The different types of solid tumors are named for the type of cells that form them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumors such as sarcomas and carcinomas include bladder cancer, brain cancer, head and neck cancer, pancreatic cancer, lung cancer, breast cancer, ovarian cancer, colon cancer, prostate cancer, and kidney cancer. In a preferred embodiment, the treatable cancer is a GPC3-positive tumor, such as hepatocellular carcinoma.
  • the CAR-modified T cells of the present invention can also be used as a type of vaccine for ex vivo immunization and/or in vivo therapy of mammals.
  • the mammal is a human.
  • at least one of the following occurs in vitro prior to administering the cells into the mammal: i) expanding the cells, ii) introducing a nucleic acid encoding a CAR into the cells, and/or iii) cryopreserving the cells.
  • 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 (ie, transduced or transfected in vitro) with a vector expressing a CAR disclosed herein. CAR-modified cells can be administered to mammalian recipients to provide therapeutic benefit.
  • the mammalian recipient can be a human, and the CAR-modified cells can be autologous to the recipient. Alternatively, cells may be allogeneic, syngeneic or xenogeneic with respect to the recipient.
  • the invention also provides compositions and methods for in vivo immunization to elicit an immune response against an antigen in a patient.
  • the present invention provides a method of treating a tumor comprising administering to a subject in need thereof a therapeutically effective amount of the CAR-modified T cells of the present invention.
  • the CAR-modified T cells of the invention can be administered alone or as a pharmaceutical composition with a diluent and/or in combination with other components such as IL-2, IL-17 or other cytokines or cell populations.
  • the pharmaceutical composition of the present invention may include the target cell population as described herein combined with one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients.
  • compositions may include buffers such as neutral buffered saline, sulfate buffered saline, etc.; carbohydrates such as glucose, mannose, sucrose or dextran, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; agents such as EDTA or glutathione; adjuvants (eg, aluminum hydroxide); and preservatives.
  • the compositions of the invention are preferably formulated for intravenous administration.
  • the pharmaceutical composition of the present invention can be administered in a manner suitable for the disease to be treated (or prevented). The amount and frequency of administration will be determined by such factors as the patient's condition, and the type and severity of the patient's disease, although appropriate dosages can be determined by clinical trials.
  • compositions comprising T cells described herein may be dosed at 1.4 to 1.9 cells/kg body weight, preferably at doses of 10, to 1013 cells/kg body weight (including those within the range All integer values) apply. T cell compositions can also be administered multiple times at these doses.
  • Cells can be administered using infusion techniques well known in immunotherapy (see, eg, Rosenberg et al., New Eng. J. of Med. 319: 1676, 1988).
  • the optimal dosage and treatment regimen for a particular patient can be readily determined by one skilled in the medical art by monitoring the patient for signs of disease and adjusting treatment accordingly.
  • Administration of the composition to a subject can be performed in any convenient manner, including by spraying, injection, swallowing, infusion, implantation or implantation.
  • the compositions described herein can be administered to a patient subcutaneously, intradermally, intratumorally, intranodally, intraspinally, intramuscularly, by intravenous (iv) injection or intraperitoneally.
  • a T cell composition of the invention is administered to a patient by intradermal or subcutaneous injection.
  • the T cell composition of the invention is preferably administered by iv injection.
  • Compositions of T cells can be injected directly into tumors, lymph nodes or sites of infection.
  • cells activated and expanded using the methods described herein or other methods known in the art to expand T cells to therapeutic levels are combined with any number of relevant treatment modalities (eg, previously , concurrently or subsequently) to the patient in the form of treatment including, but not limited to, treatment with agents such as antiviral therapy, cidofovir and interleukin-2, arabinocytosomes (also known ARA-C) or natalizumab in MS patients or erfatizumab in psoriasis or other treatments in PML.
  • agents such as antiviral therapy, cidofovir and interleukin-2, arabinocytosomes (also known ARA-C) or natalizumab in MS patients or erfatizumab in psoriasis or other treatments in PML.
  • the T cells of the present invention can be used in combination with: chemotherapy, radiation, immunosuppressants, such as cyclosporine, thiamin, methotrein, mycophenolate mofetil and FK506, antibodies or other immunotherapeutic agents.
  • the cell composition of the invention is administered in conjunction with (eg, before, simultaneously with or after) bone marrow transplantation, the use of chemotherapeutic agents such as fludarabine, external beam radiation therapy (XRT), cyclophosphamide patient.
  • chemotherapeutic agents such as fludarabine, external beam radiation therapy (XRT), cyclophosphamide patient.
  • a subject may undergo standard treatment with high-dose chemotherapy followed by peripheral blood stem cell transplantation.
  • the subject receives an infusion of expanded immune cells of the invention.
  • the expanded cells are administered before or after surgery. Dosages administered to a patient for the above treatments will vary with the precise nature of the condition being treated and the recipient of the treatment. Dosage ratios for human administration can be implemented according to practice accepted in the art. Usually, 1 x 1.6 to 1 x 10 "modified T cells (for example, CAR-T cells) of the present invention can be administered to Patients. According to the present invention, the pharmaceutical product (drug, medicament) or pharmaceutical composition of the present invention can be administered to a subject with any effective dose.
  • the pharmaceutical product (drug, medicament) or pharmaceutical composition of the present invention can be administered in multiple doses, for example, from about 2 to about 20 doses, more preferably from about 4 to 0 doses.
  • the frequency of administration about once every three weeks will be
  • the pharmaceutical product (drug, medicament) or pharmaceutical composition of the present invention is administered to the subject, such as injection, infusion or orally.
  • the administration is by injection into the tumor-bearing site.
  • the pharmaceutical product (drug, medicament) or pharmaceutical composition of the present invention may be formulated in any suitable manner for administration by any suitable route.
  • the dosage unit of the pharmaceutical product (drug, medicament) or pharmaceutical composition of the present invention is administered to a subject on a routine basis.
  • dosage units may be administered more than once daily, weekly, monthly, etc.
  • the dosage unit may be administered on a twice/week basis, that is, twice a week, for example, once every three days.
  • the instructions related to the pharmaceutical product contained in the pharmaceutical product of the present invention may contain the following contents: indications (eg, glioblastoma), dosage (eg, as exemplified above), and possible side effects, etc. . Sequences of the invention
  • MALPVTALLLPLALLLLHAARPGS (SEQ ID NO: 1) signal peptide SP nucleic acid:
  • the CAR-T cells of the present invention can efficiently target GPC3, and when the dose is 1 X 10 6 /mouse or lower, CAR-T can have a good tumor inhibitory effect; while the current solid tumor CAR-T
  • the dosage of T is generally 5 X 10&/mouse or above to achieve a good tumor inhibitory effect;
  • Example 2 HuH7-Luc, HepG2-Luc cell construction and detection
  • the luciferase gene GenBank: ACF93193.1
  • the luciferase signal value of the qualified cell line should be more than 100 times that of the original cells not transfected with lentivirus.
  • the qualified cells They were named HuH7-Luc and HepG2-Luc respectively; the results of luciferase signal detection are shown in Figure 2.
  • the detection method of CAR-T cell positive rate is as follows:
  • the BM positive control CAR-T has almost no killing effect (Figure 4); for HepG2-luc cells, When the killing activity of GC52BBC in vitro was E/T (effect-to-target ratio) of 4, both GC52BB £ and BM control CAR-T could It can kill nearly 100% of tumor cells, but when the E/T (effect-to-target ratio) is 1, GC52BB E can still have 80% killing ability, and at this time, the killing effect of BM positive control CAR-T has dropped to 50% following (Figure 5).
  • the dose was 1x107 mice, and the Mock T group only set up a high-dose 3x10° group; the day of grouping was defined as D1 day, and the drug was administered on the day of grouping.

Landscapes

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

Abstract

Provided in the invention are a GPC3-targeting chimeric antigen receptor (CAR) construct, and a CAR-T cell expressing the CAR construct. The antigen binding domain of the CAR construct comprises an antibody heavy chain variable region as shown in SEQ ID NO: 3, and an antibody light chain variable region as shown in SEQ ID NO: 7. The GPC3 CAR-T cell of the present invention can efficiently target GPC3, and an excellent anti-tumor effect can be obtained at a low dose.

Description

一种靶向 GPC3的嵌合抗原受体及其应用 技术领域 本发明涉及细 胞免疫治疗领域 , 具体地, 涉及一种靶向磷脂酰肌醇蛋白聚 糖 3 (GPC3)的嵌合抗原受体及其应用。 背景技术 A Chimeric Antigen Receptor Targeting GPC3 and Its Application Technical Field The present invention relates to the field of cellular immunotherapy, in particular, to a chimeric antigen receptor targeting Glypican 3 (GPC3) and its application. Background technique
CAR-T (Chimeric Antigen Receptor T cell)的概念最早由以色列科学家 Zelig Eshhar在 1989发表的 PNAS文章中提出, 包括抗原结合区、 皎链区、 跨膜 区和胞 内信号组成, 经历了几代结构 的发展和优化 。 2017年全球首款 CART产 品 Kymriah获得 FDA批准震撼上市, 用于儿童和年轻成年患者急性淋巴细胞性白 血病 (ALL)。 截止目前有多款靶向 CD19或 BCMA的 CAR-T产品已在多个国家或地区 获得上 市, 而且都达到了惊人的治疗效果; 然而, 这些产品都是针对血液肿瘤 的, 目前针对实体肿瘤还未有一款 CAR-T产品获批准上市, 大多数的实体肿瘤 CAR-T临床结果, 也没有血液肿瘤那 么突出。 因此, 本领域亟待开发能够有效 治疗实体 肿瘤的 CAR-T产品。 磷脂酰肌醇 蛋白聚糖 3 (Glypican 3, GPC3) 蛋白是一种细胞膜表面的硫 酸乙酰肝 素糖蛋白, 在婴儿期的一些组织中 , 特别是肝脏和肾脏中表达, 是与 器官形成 相关的胞外基质 蛋白; 在成人组织中几乎观察不到 GPC3的表达, 但是 在肝细胞 癌、黑素瘤、卵巢透明细胞癌、肺鳞状细胞癌等各 种癌组织中高表达 。 因此 GPC3是一种很好的肿瘤治疗靶点 , 可用于肿瘤靶向治疗药物的研制与开 发。
Figure imgf000002_0001
本发明的第 一方面, 提供了一种靶向 GPC3嵌合抗原受体 (CAR)构建物, 所 述嵌合抗 原受体的抗原结合 结构域包含重链可 变区和轻链可变 区, 其中, 所述的重链可变区包括 以下互补决定区 CDR:
The concept of CAR-T (Chimeric Antigen Receptor T cell) was first proposed by Israeli scientist Zelig Eshhar in a PNAS article published in 1989. It consists of antigen-binding region, chain region, transmembrane region and intracellular signal. It has gone through several generations of structure development and optimization. In 2017, the world's first CART product, Kymriah, was approved by the FDA for use in acute lymphoblastic leukemia (ALL) in children and young adults. So far, a number of CAR-T products targeting CD19 or BCMA have been launched in many countries or regions, and all of them have achieved amazing therapeutic effects; however, these products are all aimed at hematological tumors. No CAR-T product has been approved for marketing, and the clinical results of CAR-T in most solid tumors are not as prominent as in hematological tumors. Therefore, there is an urgent need in this field to develop CAR-T products that can effectively treat solid tumors. Glypican 3 (GPC3) protein is a heparan sulfate glycoprotein on the surface of the cell membrane, expressed in some tissues during infancy, especially the liver and kidney, and is a cell related to organ formation. Extracellular matrix protein; The expression of GPC3 is hardly observed in adult tissues, but it is highly expressed in various cancer tissues such as hepatocellular carcinoma, melanoma, ovarian clear cell carcinoma, and lung squamous cell carcinoma. Therefore, GPC3 is a good target for tumor therapy and can be used in the research and development of tumor-targeted therapy drugs.
Figure imgf000002_0001
The first aspect of the present invention provides a chimeric antigen receptor (CAR) construct targeting GPC3, the antigen-binding domain of the chimeric antigen receptor comprises a heavy chain variable region and a light chain variable region, Wherein, the heavy chain variable region includes the following CDRs:
SEQ ID N0:19所示的 CDR1, CDR1 shown in SEQ ID NO: 19,
SEQ ID N0:20所示的 CDR2, 和 CDR2 shown in SEQ ID NO: 20, and
SEQ ID NO: 21所示的 CDR3; 且所述的轻链 可变区包括以下 互补决定区 CDR: The CDR3 shown in SEQ ID NO: 21; and the light chain variable region includes the following complementarity determining region CDR:
SEQ ID N0:22所示的 CDR1' , CDR1' shown in SEQ ID NO:22,
SEQ ID N0:23所示的 CDR2? , 和
Figure imgf000003_0001
(GPC3) 。 在另一优选例 中, 所述抗原结合结构域的重链可变区 和轻链可变区来源 于 鼠源、 人源、 或人源化抗体。
Figure imgf000003_0002
在另一优选例 中, 所述的 H为 CD8来源的钗链区。 在另一优选例 中, H的氨基酸序列如 SEQ ID NO : 9所示。 在另一优选例 中, 所述的 TM为选自下组的蛋白的跨膜区: ICOS、 CD28. CD3 eps i lon、 CD45、 CD4、 CD5、 CD8、 CD9、 CD16、 GD2、 CD33、 CD37、 CD64、 CD80、
Figure imgf000004_0001
本发明的第 二方面, 提供了一种分离的核酸分子 , 所述核酸分子编码本发 明第一 方面所述的嵌合抗 原受体 (CAR)构建物。 在另一优选例 中, 所述核酸分子如 SEQ ID NO : 18所示。 在本发明 的另一方面, 提供了一种编码嵌合抗原受 体的核昔酸序列 , 所述 嵌合抗 原受体主要部分组 成包括抗原结合结 构域、 跨膜结构域和共刺激信号传 导区 。 将 GPC3特异性抗体编码基因连接到经改造的含有跨膜结构基因与共刺 激 信号基 因的 CAR骨架的克隆位点区中获得 CAR基因。 所述的抗原结合结构域能够 特异性 结合肿瘤特异性抗 原 GPC3 , 并通过跨膜结构域和共刺激信号传导区激活 该 NK细胞。 在另一优选例 中, 所述的抗原结合结构域为特异性结 合 GPC3的抗体或其抗 原结合 片段, 所述抗原结合片段为 Fab或 scFv o 在另一优选例 中, 所述原结合结构域为 scFv , 其重链可变区的核酸序列如 SEQ ID NO : 4所示, 以及其轻链可变区的核酸序列如 SEQ ID NO : 8所示。 本发明的第 三方面, 提供了一种载体, 所述的载体含有本发明第二方 面所 述的核 酸分子。 在另一优选 例中, 所述的载体选自下组 : DNA、 RNA、 质粒、 慢病毒载体、 腺病毒 载体、 腺相关病毒载体 (AAV)、 逆转录病毒载体、 转座子、 或其组合。 在另一优选例 中, 所述的载体选自下组: 质粒、 病毒载体。 在另一优选例 中, 所述载体为病毒颗粒的形式 。 在另一优选例 中, 所述载体为慢病毒载体。 本发明的第 四方面, 提供了一种宿主细胞, 所述宿主细胞中含有本发 明第 三方面所 述的载体, 或染色体中整合有外源 的如本发明第二方面 所述的核酸分 子, 或表达如本发明第一方 面所述的 CAR构建物。 在另一优选例 中, 所述的宿主细胞包括真核细胞 和原核细胞。 在另一优选例 中, 所述的宿主细胞包括大肠杆菌 。
Figure imgf000005_0001
本发明的第六 方面, 提供了一种制剂, 所述制剂包含如本发明第一方 面所 述的 CAR构建物、 如本发明第二方面所述的分 离的核酸分子 、 如本发明第三方 面所述 的载体或如本发 明第五方面所述的免 疫细胞, 以及药学上可接受的载体 。 在另一优选例 中, 所述制剂为液态制剂。 在另一优选例 中, 所述制剂的剂型为注射剂。 在另一优选 例中, 所述制剂中免疫细胞 (GPC3 CAR-T细胞) 的浓度为 I X
CDR2 shown in SEQ ID NO: 23 ? , and
Figure imgf000003_0001
(GPC3). In another preferred example, the heavy chain variable region and the light chain variable region of the antigen-binding domain are derived from murine, human, or humanized antibodies.
Figure imgf000003_0002
In another preferred example, said H is a hairpin chain region derived from CD8. In another preferred example, the amino acid sequence of H is shown in SEQ ID NO: 9. In another preferred example, the TM is a transmembrane region of a protein selected from the group consisting of ICOS, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, GD2, CD33, CD37, CD64, CD80,
Figure imgf000004_0001
The second aspect of the present invention provides an isolated nucleic acid molecule encoding the chimeric antigen receptor (CAR) construct described in the first aspect of the present invention. In another preferred example, the nucleic acid molecule is shown in SEQ ID NO: 18. In another aspect of the present invention, a nucleotide sequence encoding a chimeric antigen receptor is provided, the main part of the chimeric antigen receptor includes an antigen binding domain, a transmembrane domain and a costimulatory signal transduction region . The GPC3-specific antibody coding gene is connected to the modified cloning site region of the CAR backbone containing the transmembrane structural gene and costimulatory signal gene to obtain the CAR gene. The antigen-binding domain can specifically bind the tumor-specific antigen GPC3, and activate the NK cell through the transmembrane domain and costimulatory signal transduction region. In another preferred example, the antigen-binding domain is an antibody that specifically binds GPC3 or an antigen-binding fragment thereof, and the antigen-binding fragment is Fab or scFv o In another preferred example, the original binding domain It is scFv, the nucleic acid sequence of its heavy chain variable region is shown in SEQ ID NO: 4, and the nucleic acid sequence of its light chain variable region is shown in SEQ ID NO: 8. The third aspect of the present invention provides a vector containing the nucleic acid molecule described in the second aspect of the present invention. In another preferred embodiment, the vector is selected from the group consisting of DNA, RNA, plasmid, lentiviral vector, adenoviral vector, adeno-associated viral vector (AAV), retroviral vector, transposon, or a combination thereof . In another preferred example, the vector is selected from the group consisting of plasmids and viral vectors. In another preferred embodiment, the vector is in the form of virus particles. In another preferred example, the vector is a lentiviral vector. The fourth aspect of the present invention provides a host cell containing the vector according to the third aspect of the present invention, or an exogenous nucleic acid molecule as described in the second aspect of the present invention integrated in the chromosome, Or express the CAR construct as described in the first aspect of the present invention. In another preferred example, the host cells include eukaryotic cells and prokaryotic cells. In another preferred example, the host cells include Escherichia coli.
Figure imgf000005_0001
The sixth aspect of the present invention provides a preparation comprising the CAR construct according to the first aspect of the present invention, the isolated nucleic acid molecule according to the second aspect of the present invention, the third aspect of the present invention The carrier or the immune cell according to the fifth aspect of the present invention, and a pharmaceutically acceptable carrier. In another preferred example, the formulation is a liquid formulation. In another preferred example, the dosage form of the preparation is injection. In another preferred example, the concentration of immune cells (GPC3 CAR-T cells) in the preparation is IX
1O3-1 X 1 O8个细胞 /ml , 较佳地 1 X 104-5 X l(f个细胞 /ml。 在另一优选例 中, 所述制剂可包括缓冲液诸如中性 缓冲盐水、 硫酸盐缓冲 盐水等等 ; 碳水化合物诸如葡萄糖、甘露糖、蔗糖或葡聚糖、甘露醇 ; 蛋白质; 多肽或 氨基酸诸如甘氨 酸; 抗氧化剂; 螯合剂诸如 EDTA或谷胱甘肽; 佐剂(例 如, 氢氧化铝); 和防腐剂。 本发明的制剂优选配制用于静脉内施用。 在另一优选例 中, 所述的制剂还包含抗 肿瘤的第二活性成 分, 较佳地包括 第二抗体 、 或化疗剂。 在另一优选例 中, 所述的化疗剂选自下组: 多西他赛、 卡钳、 或其组合。 在另一优选例 中, 所述制剂为药物组 合物。 在另一优选例 中, 所述制剂施用时 GPC3 CAR-T细胞与肿瘤细胞的效靶比为 (0. 1-20) : 1 , 优选地, 所述效靶比为(1 - 4) : 1。 本发明的第 七方面, 提供了如本发明第一 方面所述的 CAR构建物、 如本发 明第二方 面所述的核酸分子 、 如本发明第三方面所述的载体、 如本发明第五方 面所述 的免疫细胞、 或如本发明第六方面所述 的制剂的用途 , 用于制备预防和 /或治疗肿瘤 或癌症的药物 。 在另一优选例 中, 所述肿瘤或癌症高 (过) 表达 GPC3。 在另一优选例 中, 所述肿瘤或癌症为实体瘤。 在另一优选例 中, 所述实体瘤选自下组: 肝细胞癌、 黑素瘤、 卵巢透明细 胞癌、 肺鳞状细胞癌、 乳腺癌、 肾癌、 胆管癌或其组合。 在另一优选例 中, 所述肿瘤或癌症为肝细胞癌 。 在另一优选例 中, 所述肿瘤或癌症为高 (过) 表达 GPC3的肝细胞。 本发明的第八 方面, 提供了一种制备如本发明第五方 面所述的工程化 的免 疫细胞 的方法, 所述方法包括以下步骤: 1O 3 -1 X 1 O 8 cells/ml, preferably 1 X 10 4 -5 X l (f cells/ml. In another preferred embodiment, the preparation may include a buffer such as neutral buffered saline , sulfate buffered saline, etc.; carbohydrates such as glucose, mannose, sucrose or dextran, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (eg eg, aluminum hydroxide); and preservatives. The formulations of the invention are preferably formulated for intravenous administration. In another preferred example, the preparation further includes a second anti-tumor active ingredient, preferably a second antibody or a chemotherapeutic agent. In another preferred example, the chemotherapeutic agent is selected from the group consisting of docetaxel, calipers, or a combination thereof. In another preferred example, the preparation is a pharmaceutical composition. In another preferred example, when the preparation is administered, the effect-to-target ratio of GPC3 CAR-T cells to tumor cells is (0.1-20): 1, preferably, the effect-to-target ratio is (1-4): 1. The seventh aspect of the present invention provides the CAR construct according to the first aspect of the present invention, the nucleic acid molecule according to the second aspect of the present invention, the vector according to the third aspect of the present invention, the fifth aspect of the present invention The use of the immune cells according to the aspect, or the preparation according to the sixth aspect of the present invention is used to prepare a drug for preventing and/or treating tumor or cancer. In another preferred example, the tumor or cancer highly (over) expresses GPC3. In another preferred example, the tumor or cancer is a solid tumor. In another preferred example, the solid tumor is selected from the group consisting of hepatocellular carcinoma, melanoma, ovarian clear cell carcinoma, lung squamous cell carcinoma, breast cancer, renal carcinoma, cholangiocarcinoma or a combination thereof. In another preferred example, the tumor or cancer is hepatocellular carcinoma. In another preferred example, the tumor or cancer is a liver cell that highly (over)expresses GPC3. The eighth aspect of the present invention provides a method for preparing the engineered immune cells according to the fifth aspect of the present invention, the method comprising the following steps:
(a)提供待改造的免疫细胞; 和 (a) providing immune cells to be engineered; and
(b)将如权利要求 2所述的核酸分子或如权利要求 3所述的载体转导入所述 免疫细胞 内, 从而获得所述工程化的免疫细 胞。 在另一优选例 中, 所述工程化的免疫细胞为 CART细胞或 CAR-NK细胞。 在另一优选例 中, 所述的方法还包括对获得的工程化 免疫细胞进行功 能和 有效性检 测的步骤。 本发明的第 九方面, 提供了一种治疗肿瘤或癌症 的方法, 包括向需要的对 象施用有 效量的如本发 明第五方面所述的免 疫细胞, 或如本发明第六方面所述 的制剂 。 在另一优选例 中, 所述肿瘤或癌症高 (过) 表达 GPC3。 在另一优选例 中, 所述肿瘤或癌症为实体瘤。 在另一优选例 中, 所述实体瘤选自下组: 肝细胞癌、 黑素瘤、 卵巢透明细 胞癌、 肺鳞状细胞癌、 乳腺癌、 肾癌、 胆管癌或其组合 在另一优选例 中, 所述肿瘤或癌症为肝细胞癌 。 应理解, 在本发明范围 内中, 本发明的上述各技术特征和 在下文(如实施 例)中具体描述的各 技术特征之间 都可以互相组合 , 从而构成新的或优选的技 术方案 。 限于篇幅, 在此不再一一累述。 附图说明 图 1显示了本发明的靶向 GPC3的嵌合抗原受体 GC52BB &的结构示意图。 图 2显示了 HuH7-Luc> HepG2-Luc细胞构建与检测。 图 3显示了 GC52BB C阳性率检测。 图 4显示了 GC52BB V体外杀伤 HuH7-luc细胞; 其中左图显示了在 E/T(效靶 比)为 4时, GC52BB E对 HuH7Tuc细胞的杀伤能力;右图显示了在 E/T(效靶比) 为 1时, GC52BB V对 HuH7-luc细胞的杀伤能力。 图 5显示了 GC52BB C体外杀伤 HepG2-luc细胞; 其中左图显示了在 E/T (效 靶比) 为 4时, GC52BB,对 HepG2Tuc细胞的杀伤能力; 右图显示了在 E/T (效 靶比) 为 1时, GC52BB,对 HepG2-luc细胞的杀伤能力。 图 6显示了 GC52BB V体内抑瘤效果。 具体实施方 式 本发明人经过 广泛而深入地研 究, 首次意外地发现一种靶向识别 GPC3的嵌 合抗原 受体、 其编码核昔酸, 以及表达该 CAR的 GPC3 CAR-T细胞及其制备方法 及应用 。本发明的 CART细胞对 GPC3具有特异性,能够特异性识别和杀伤肿瘤, 尤其是 高表达或过表达 GPC3的实体瘤 (例如肝细胞癌) 具有更高效的肿瘤杀伤 活性, 在低剂量下(约为现有技术中常用剂 量的 1/5)就可达到很好的抑瘤效果, 并且性状 稳定, 可大批量生产和制备。 在此基础上 , 完成了本发明。 为了更好地理 解本发明, 以下定义了某些术语。 除非另有定义, 本发明中 使用 的所有技术和 科学术语具有 与本发明所 述技术领域 的普通技术人 员通常 理解的相 同含义。 如本文所用 , “抗原结合结构域” “单链抗体片段” 均指 具有抗原结合活 性的 Fab片段, Fab ' 片段, F(ab 72片段, 或单一 Fv片段。 Fv抗体含有抗体重 链可变 区、 轻链可变区, 但没有恒定区, 并具有全部抗原结合位点的最小抗体 片段。 一般的, Fv抗体还包含 VH和 VL结构域之间的多肽接头, 且能够形成抗原 结合 所需 的结 构。 抗原结 合结构 域通 常是 scFv (single-chain variable fragment) o scFv的大小一般是一个完整抗体的 1/6。 单链抗体优选是由一条核 昔酸链 编码的一条氨基酸链 序列。 作为本发明的优选方式, 所述 scFv包含特异 性识别 GPC3的抗体, 较佳地为人源化的单链抗体。 对于绞链 区和跨膜区 (跨膜结构域) , CAR可被设计以包括融合至 CAR的胞外 结构域 的跨膜结构域 。 在一个实施方式中, 使用天然与 CAR中的结构域之一相 关联 的跨膜结构域。 在一些例子中, 可选择跨膜结构域, 或通过氨基酸置换进 行修饰 , 以避免将这样的结构域结合至相同或 不同的表面膜蛋 白的跨膜结构域, 从而最 小化与受体复合物 的其他成员的相互 作用。 本发明中 , 术语 “抗体 ”指的是与抗原特异性结合的免疫球 蛋白分子。 抗 体可为源 于自然源或源于 重组源的完整的免 疫球蛋白, 并可为完整免疫球蛋 白 的免疫反 应部分。 抗体通常为免疫球蛋 白分子的四聚物。 本发明中的抗体可以 以多种 形式存在, 包括例如, 多克隆抗体、 单克隆抗体、 Fv、 Fab和 F (ab) 2 , 以及单 链抗体和人源 化抗体等 ( Harlow等, 1999, In: Using Antibodies : A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY; Harlow等, 1989, In: Antibodies : A Laboratory Manual , Cold Spring Harbor , New York; Houston等, 1988, Proc. Natl. Acad. Sci. USA 85 : 5879-5883; Bird等, 1988, Science 242 : 423-426) 。 术语 “抗体片段”指 的是完整抗体的一部 分, 并指的是完整抗体的抗原决 定可变 区。 抗体片段的例子包括但不限于 Fab、 Fab'、 F (ab‘ ) 2和 Fv片段, 由抗 体片段 形成的线性抗体、 scFv抗体和多特异性抗体。 除非另有规定 , “编码核昔酸” 或 “编码核酸”包括为彼 此简并版本并编 码相 同的氨基酸序列的所有 核昔酸序列。 编码蛋白质的核昔酸序 列可包括内含 子。 术语 “慢病毒” 指的是逆转录病毒科的属 , 其能够有效感染非周期性和有 丝分裂 后的细胞; 它们可传递显著量 的遗传信息进入 宿主细胞的 DNA, 以便它 们是基 因传递载体的最有 效的方法之一。 术语 “启动子”被定义为 开始多核昔酸序 列的特异性转录需 要的, 由细胞 的合成机 器识别或引导合 成机器的 DNA序列。 术语 “特异性结合” 指识别特异性抗原但 基本上不识别 或结合样本 中的 其他分 子。 术语 “载体”为物质组 合物, 其包括分离的核酸, 并且其可用于传递分离 的核酸 至细胞内部。 很多载体在本领域中是 已知的, 包括但不限于线性多核昔 酸、 与离子或两性分子化合 物相关的多核昔 酸、 质粒和病毒。 因此, 术语 “载 体” 包括 自主复制的质粒或病毒 。 该术语也应被解释为包括便于将核 酸转移入 细胞 的非质粒和非病毒化合 物, 诸如例如聚赖氨酸化合物、 脂质体等等。 病毒 载体 的例子包括但不限于 , 腺病毒载体、 腺伴随病毒载体、 逆转录病毒载体等 等。 术语 “癌症”被定义为 以畸变细胞的快速 和失控生长为特 征的疾病。 癌症 细胞可 局部蔓延或通过血 流和淋巴系统蔓延 至身体的其他部分 。 各种癌症的例 子包括但 不限于乳腺癌、 前列腺癌、 卵巢癌、 子宫颈癌、 皮肤癌、 胰腺癌、 结 肠直肠癌 、 肾癌、 肝癌、 脑癌、 淋巴瘤、 白血病、 肺癌等等。 如本文所使 用的, “包含 ”与 “包括” 、 “含有 ”或 “特征在于” 同义, 并且是 包括在内的或 开放性的, 并且不排除另外的未 陈述的元件或 方法步骤。 术语 “包含 ”在本文中的任何表述 , 特别是在描述本发明的方法、 用途或产品 时, 应理解为包括基本上 由所述组分或元件 或步骤组成和由所述 组分或元件或 步骤组 成的那些产品、 方法和用途。 本文示例性描述的本发 明适当地可以在不 存在本 文未具体公开的任 何一种或多种元件 、 一种或多种限制的情况下进行实 践。 本文已采用 的术语和表述用作 描述性而不是限制 性术语, 并且在此种术语 和表述 的使用中不预期排 除所示和所述特征 或其部分的任何等 价物, 但应认识 到各种修 饰在请求保护 的本发明的范围内是 可能的。 因此, 应当理解尽管本发 明己通 过优选实施方案和任 选特征具体公开 , 但本领域技术人员可以采用本文 公开 的概念的修饰和变化 , 并且此类修饰和变化被视为在如 由附加权利要求定 义的本 发明的范围内。 本文中出现 的英文名称不区分 大小写,其表示的含义相同 ;例如 scFv、 ScFv、 SCFV等表示的含 义相同; GPC3 CAR-T与 Anti GPC3-CAR T表示相同的含义, 均 表示抗 GPC3分子的 CAR-T细胞。 嵌合抗原受体 (CAR) (b) Transducing the nucleic acid molecule according to claim 2 or the vector according to claim 3 into the immune cells, thereby obtaining the engineered immune cells. In another preferred example, the engineered immune cells are CART cells or CAR-NK cells. In another preferred example, the method further includes the step of testing the function and effectiveness of the obtained engineered immune cells. The ninth aspect of the present invention provides a method for treating tumor or cancer, comprising administering to a subject in need an effective amount of the immune cells described in the fifth aspect of the present invention, or the preparation described in the sixth aspect of the present invention . In another preferred example, the tumor or cancer highly (over) expresses GPC3. In another preferred example, the tumor or cancer is a solid tumor. In another preferred example, the solid tumor is selected from the group consisting of hepatocellular carcinoma, melanoma, ovarian clear cell carcinoma, lung squamous cell carcinoma, breast cancer, kidney cancer, cholangiocarcinoma or a combination thereof In another preferred example, the tumor or cancer is hepatocellular carcinoma. It should be understood that within the scope of the present invention, the above-mentioned technical features of the present invention and the technical features specifically described in the following (such as embodiments) can be combined with each other to form new or preferred technical solutions. Due to space limitations, they will not be repeated here. BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 shows a schematic diagram of the structure of the GPC3-targeting chimeric antigen receptor GC52BB& of the present invention. Figure 2 shows the construction and detection of HuH7-Luc>HepG2-Luc cells. Figure 3 shows the detection of GC52BB C positive rate. Figure 4 shows that GC52BB V kills HuH7-luc cells in vitro; the left panel shows the killing ability of GC52BB E on HuH7Tuc cells when E/T (effect-to-target ratio) is 4; Target ratio) is 1, the killing ability of GC52BB V on HuH7-luc cells. Figure 5 shows that GC52BB C kills HepG2-luc cells in vitro; the left picture shows the killing ability of GC52BB to HepG2Tuc cells when E/T (effect-to-target ratio) is 4; Target ratio) is 1, GC52BB has the ability to kill HepG2-luc cells. Figure 6 shows the anti-tumor effect of GC52BB V in vivo. DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS After extensive and in-depth research, the present inventors unexpectedly discovered for the first time a chimeric antigen receptor that targets and recognizes GPC3, its encoding nucleotides, and GPC3 CAR-T cells expressing the CAR and its preparation method and applications. The CART cells of the present invention are specific to GPC3, and can specifically recognize and kill tumors, especially solid tumors (such as hepatocellular carcinoma) that highly express or overexpress GPC3, and have more efficient tumor killing activity, at low doses (approximately It is only 1/5 of the usual dose in the prior art to achieve a good anti-tumor effect, and its properties are stable, and it can be mass-produced and prepared. On this basis, the present invention has been accomplished. In order to better understand the present invention, certain terms are defined below. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. As used herein, "antigen-binding domain" and "single-chain antibody fragment" all refer to a Fab fragment, Fab' fragment, F(ab 7 ) 2 fragment, or a single Fv fragment with antigen-binding activity. Fv antibodies contain antibody heavy chain variable regions, light chain variable regions, but no constant region, and the smallest antibody fragment with all antigen-binding sites. Typically, Fv antibodies also contain a polypeptide linker between the VH and VL domains and are capable of forming antigenic Combine the desired structure. The antigen-binding domain is usually scFv (single-chain variable fragment) o The size of scFv is generally 1/6 of a complete antibody. A single chain antibody is preferably a sequence of one amino acid chain encoded by one nucleotide chain. As a preferred mode of the present invention, the scFv comprises an antibody that specifically recognizes GPC3, preferably a humanized single-chain antibody. For the hinge region and transmembrane region (transmembrane domain), CAR can be designed to include the transmembrane domain fused to the extracellular domain of the CAR. In one embodiment, a transmembrane domain naturally associated with one of the domains in the CAR is used. In some instances, transmembrane domains may be selected, or modified by amino acid substitutions, to avoid binding such domains to transmembrane domains of the same or different surface membrane proteins, thereby minimizing interaction with the receptor complex. interactions with other members. In the present invention, the term "antibody" refers to an immunoglobulin molecule that specifically binds to an antigen. Antibodies can be intact immunoglobulins, derived from natural sources or from recombinant sources, and can be immunoreactive portions of intact immunoglobulins. Antibodies are typically tetramers of immunoglobulin molecules. Antibodies in the present invention can exist in various forms, including, for example, polyclonal antibodies, monoclonal antibodies, Fv, Fab, and F (ab) 2 , as well as single-chain antibodies and humanized antibodies, etc. (Harlow et al., 1999, In : Using Antibodies : A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY; Harlow et al., 1989, In: Antibodies : A Laboratory Manual , Cold Spring Harbor , New York; Houston et al., 1988, Proc. Natl. Acad. Sci. USA 85: 5879-5883; Bird et al., 1988, Science 242: 423-426). The term "antibody fragment" refers to a portion of an intact antibody and refers to the antigenically determining variable regions of an intact antibody. Examples of antibody fragments include, but are not limited to, Fab, Fab', F(ab')2 and Fv fragments, linear antibodies, scFv antibodies and multispecific antibodies formed from antibody fragments. Unless otherwise specified, "encoding nucleotides" or "encoding nucleic acids" include all nucleotide sequences that are degenerate versions of each other and encode the same amino acid sequence. A nucleotide sequence encoding a protein may include introns. The term "lentivirus" refers to a genus of the Retroviridae family that is capable of efficiently infecting aperiodic and post-mitotic cells; they can transfer significant amounts of genetic information into the host cell's DNA, so that they are the most suitable vectors for gene delivery. one of the effective methods. The term "promoter" is defined as a DNA sequence that is recognized by or directs the synthetic machinery of a cell, required to initiate specific transcription of a polynucleotide sequence. The term "specifically binds" refers to recognition of a specific antigen but substantially no recognition or binding of other molecules in the sample. The term "vector" is a composition of matter that includes an isolated nucleic acid and that can be used to deliver the isolated nucleic acid to the interior of a cell. Many vectors are known in the art, including but not limited to linear polynucleotides, polynucleotides associated with ionic or amphiphilic compounds, plasmids and viruses. Thus, the term "vector" includes autonomously replicating plasmids or viruses. The term should also be construed to include the facilitation of transfer of nucleic acid into Cellular non-plasmid and non-viral compounds such as, for example, polylysine compounds, liposomes and the like. Examples of viral vectors include, but are not limited to, adenoviral vectors, adeno-associated viral vectors, retroviral vectors, and the like. The term "cancer" is defined as a disease characterized by the rapid and uncontrolled growth of abnormal cells. Cancer cells can spread locally or to other parts of the body through the bloodstream and lymphatic system. Examples of various cancers include, but are not limited to, breast cancer, prostate cancer, ovarian cancer, cervical cancer, skin cancer, pancreatic cancer, colorectal cancer, kidney cancer, liver cancer, brain cancer, lymphoma, leukemia, lung cancer, and the like. As used herein, "comprising" is synonymous with "including", "comprising" or "characterized by" and is inclusive or open-ended and does not exclude additional unstated elements or method steps. Any expression of the term "comprising" herein, especially when describing the methods, uses or products of the present invention, should be understood as including consisting essentially of the components or elements or steps and consisting of the components or elements or steps consisting of those products, methods and uses. The invention exemplified herein suitably may be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein. The terms and expressions which have been employed herein are used as terms of description rather than limitation, and in the use of such terms and expressions it is not intended to exclude any equivalents of the features shown and described or parts thereof, but various modifications are recognized It is possible within the scope of the claimed invention. Accordingly, it should be understood that although the invention has been specifically disclosed by preferred embodiments and optional features, modifications and variations of the concepts disclosed herein may be employed by those skilled in the art and that such modifications and variations are considered to be defined within the scope of the invention. The English names appearing in this article are not case-sensitive, and they have the same meaning; for example, scFv, ScFv, SCFV, etc. have the same meaning; GPC3 CAR-T and Anti GPC3-CAR T have the same meaning, and both represent the anti-GPC3 molecule CAR-T cells. Chimeric Antigen Receptor (CAR)
CARs的设计经历了以下 过程: 第一代 CAR只有一个胞内信号组份 CD3 &或者 Fc y RI分子, 由于胞内只有一个活化结构域, 因此它只能引起短暂 的 T细胞增 殖和较 少的细胞因子 分泌, 而并不能提供长时间的 T细胞增殖信号和 持续的体 内抗肿瘤 效应, 所以并没有取得很好地临床 疗效。 第二代 CARs在原有结构基础 上引入 一个共刺激分 子, 如 CD28、 4-1BB、 0X40、 ICOS , 与一代 CARs相比功能 有很大 提高, 进一步加强 CAR-T细胞的持续性和对肿瘤细胞的杀 伤能力。 在二 代 CARs基础上串联一些新的免 疫共刺激分子如 CD27、 CD134 , 发展成为三代和 四代 CARs。 本发明的嵌合 抗原受体(CAR)为二代 CAR,包括细胞外结构域、跨膜结构域、 和细胞 内结构域。 胞外结构域包 括靶-特异性结 合元件(也称为抗原结合 结构 域)。 细胞内结构域包括共刺激信号 传导区和 匕链部分。 共刺激信号传导区指 包括共刺 激分子的细胞 内结构域的一部分。 共刺激分子为淋巴细 胞对抗原的有 效应答所 需要的细胞表面分 子, 而不是抗原受体或它们的配体 。 在 CAR的胞外结构域和跨膜结构域之间 , 或在 CAR的胞浆结构域和跨膜结构 域之间 , 可并入接头。 如本文所用的, 术语“接头”通常 指起到将跨膜结 构域 连接至 多肽链的胞外结构域 或胞浆结构域作用 的任何寡肽或多肽 。 接头可包括 0-300个氨基酸, 优选地 2至 100个氨基酸和最优选地 3至 50个氨基酸。 在本发明 的一个较佳的实施 方式中, 本发明提供的 CAR的胞外结构域包括 靶向 GPC3的抗原结合结构域。 本发明的 CAR当在 T细胞中表达时, 能够基于抗原 结合特异 性进行抗原识别 。 当其结合其关联抗原时, 影响肿瘤细胞, 导致肿瘤 细胞不生 长、 被促使死亡或以其他方式被影 响, 并导致患者的肿瘤负荷缩小或 消除。 抗原结合结构域优选 与来自共刺激分子 和匕链中的一个或 多个的细胞内 结构域融合 。 载体 编码期望分子 的核酸序列可利 用在本领域中已知 的重组方法获得 , 诸如例 如通过从 表达基因的细胞 中筛选文库, 通过从已知包括该基因 的载体中得到该 基因, 或通过利用标准的技 术, 从包含该基因的细胞和组织 中直接分离。 可选 地, 感兴趣的基因可被合成 生产。 本发明也提供 了其中插入本发 明的表达盒的载体 。 源于逆转录病毒诸如慢 病毒的载 体是实现长期基 因转移的合适工具 , 因为它们允许转基因长期、 稳定 的整合 并且其在子细胞 中增殖。 慢病毒载体具有超过源自致癌逆 转录病毒诸如 鼠科 白血病病毒的载体 的优点, 因为它们可转导非增 殖的细胞, 诸如肝细胞。 它们也具 有低免疫原性的优 点。 简单概括 , 通常可操作地连接本发明的表达盒或核 酸序列至启动子 , 并将 其并入表 达载体。 该载体适合于复制和整合真 核细胞。 典型的克隆载体包含可 用于调节 期望核酸序列表达 的转录和翻译终 止子、 初始序列和启动子。 本发明的表达 构建体也可利用标 准的基因传递方 案, 用于核酸免疫和基因 疗法 。 基因传递的方法在本领域中是 己知的。 见例如美国专利号 5, 399, 346、 5, 580, 859、 5, 589, 466, 在此通过引用全文并入。 在另一个实施方式中, 本发 明提供 了基因疗法载体。 该核酸可被 克隆入许多类 型的载体。 例如, 该核酸可被克隆入如此载体, 其包括但 不限于质粒、 噬菌粒、 噬菌体衍生物、 动物病毒和粘粒。 特定的感兴 趣载体包 括表达载体、 复制载体、 探针产生载体和测序载体 。 进一步地 , 表达载体可以以病毒载体形式提供给细 胞。 病毒载体技术在本 领域 中是公知的并在例如 Sambrook等 (2001, Molecular Cloning : A Laboratory Manual, Cold Spring Harbor Laboratory, New York)和其他病毒学和分子生物 学手册 中进行了描述 。 可用作载体的病毒包括但不 限于逆转录病毒 、 腺病毒、 腺伴随病 毒、 疱疹病毒和慢病毒。 通常, 合适的载体包含在至少一种有机体中 起作用 的复制起点、 启动子序列、 方便的限制酶位点和一个或 多个可选择的标 记 (例如, W001/96584; W001/29058; 和美国专利号 6, 326, 193)。 己经开发 许多基于病毒 的系统, 用于将基因转移入哺乳 动物细胞。 例如, 逆转录病 毒提供了用于基 因传递系统的方便 的平台。 可利用在本领域中已知的 技术将选 择的基因插入载体 并包装入逆转录 病毒颗粒。 该重组病毒可随后被分 离和传 递至体内或离体 的对象细胞。 许多逆转录病毒系统在本领 域中是已知的 。 在一些 实施方式中, 使用腺病毒载体 。 许多腺病毒载体在本领域 中是已知的。 在一个 实施方式中, 使用慢病毒载体。 额外的启动子 元件, 例如增强子, 可以调节转录开始的频率。 通常地, 这 些位于 起始位点上游的 30-110bp区域中, 尽管最近已经显示许多启动子也包含 起始位 点下游的功能元件 。 启动子元件之间的间隔经常是柔性 的, 以便当元件 相对于 另一个被倒置或 移动时, 保持启动子功能。 在胸昔激酶 (tk)启动子中, 启动子 元件之间的间隔 可被增加隔开 50bp, 活性才开始下降。 取决于启动子, 表现 出单个元件可合作或独 立地起作用, 以起动转录。 合适的启动 子的一个例子为 即时早期巨细 胞病毒 (CMV)启动子序列。 该启 动子序 列为能够驱 动可操作地连 接至其上 的任何多核昔 酸序列高水平 表达的 强 组成型 启动 子序 列。 合适的启动 子的 另一个 例子 为延 伸生 长因 子 -1 a (EF-1 a ) o 然而, 也可使用其他组成型启动子序列, 包括但不限于类人猿 病毒 40 (SV40)早期启动子、 小鼠乳癌病毒 (MMTV)、 人免疫缺陷病毒 (HIV)长末 端重复 (LTR)启动子、 MoMuLV启动子、 鸟类白血病病毒启动子、 艾伯斯坦 -巴尔 (Epstein-Barr)病毒即时早期启动子、 鲁斯氏肉瘤病毒启动子、 以及人基因启 动子 , 诸如但不限于肌动蛋白启动子、 肌球蛋白启动子、 血红素启动子和肌酸 激酶启动 子。 进一步地, 本发明不应被限于组成型启动子的应用 。 诱导型启动 子也被 考虑为本发明的一 部分。 诱导型启动子的使用提供了分 子开关, 其能够 当这样 的表达是期望的时 , 打开可操作地连接诱导型启动子 的多核昔酸序列的 表达 , 或当表达是不期望的时关闭表达。 诱导型启动子的例子包 括但不限于金 属硫蛋 白启动子、 糖皮质激素启动子、 孕酮启动子和四环素启动子 。 为了评估 CAR多肽或其部分的表达 , 被引入细胞的表达载体也可 包含可选 择的标记 基因或报道基 因中的任一个或两者 , 以便于从通过病毒载体寻求被转 染或感 染的细胞群中鉴定 和选择表达细胞 。 在其他方面, 可选择的标记可被携 带在单 独一段 DNA上并用于共转染程序。 可选择的标记和报 道基因两者的侧 翼
Figure imgf000012_0001
领域 中是公知的。 见例如 Sambrook等 (2001, Molecular Cloning : A Laboratory Manual, Cold Spring Harbor Laboratory, New York) o 将多核昔酸引入宿主细 胞的优选 方法为磷酸钙转染 。 将感兴趣 的多核昔酸引入 宿主细胞的生物 学方法包括使 用 DNA和 RNA载体。 病毒载体 , 特别是逆转录病毒载体, 己经成为最广泛使用的将基 因插入哺乳动 物例如 人细胞的方法。其他病毒 载体可源自慢病毒 、痘病毒、单纯疱疹病毒 I、 腺病毒 和腺伴随病毒等等 。 见例如美国专利号 5, 350, 674和 5, 585, 362。 将多核昔酸 引入宿主细胞的化 学手段包括胶体分 散系统, 诸如大分子复合 物、 纳米胶囊、 微球、 珠; 和基于脂质的系统, 包括水包油乳剂、 胶束、 混合 胶束和 脂质体。 用作体外和体内传递工具 (del ivery vehicle)的示例性胶体系 统为脂质 体 (例如, 人造膜囊)。 在使用非病 毒传递系统的情况 下, 示例性传递工具为脂质体。 考虑使用脂 质制剂 , 以将核酸引入宿主细胞 (体外、 离体 (ex vivo)或体内)。 在另一方面, 该核酸 可与脂质相关联 。 与脂质相关联的核酸可被封装入脂质体 的水性内部中, 散布在 脂质体的脂双层 内, 经与脂质体和寡核昔酸两者都相关联 的连接分子附 接至脂质 体, 陷入脂质体, 与脂质体复合, 分散在包含脂质的溶液中, 与脂质 混合 , 与脂质联合, 作为悬浮液包含在脂质中, 包含在胶束中或与 胶束复合, 或以其 他方式与脂质相 关联。 与组合物相关联的脂质 、 脂质 /DNA或脂质 /表达 载体不 限于溶液中的任何具 体结构。 例如, 它们可存在于双分子层结构中, 作 为胶束或 具有 “坍缩的(col lapsed) ” 结构。 它们也可简单地被散布在溶液中, 可能形成 大小或形状不均一 的聚集体。 脂质为脂肪物质, 其可为天然发生或合 成的脂质 。 例如, 脂质包括脂肪小滴, 其天然发生在细胞质以及包含长链脂肪 族炷和它们 的衍生物诸如脂 肪酸、 醇类、 胺类、 氨基醇类和醛类的该类化合物 中。 在本发明 的一个优选地实施方 式中, 所述载体为慢病毒载体。 治疗性应用 本发明包 括用编码本发 明 CAR的慢病毒载体(LV)转导的细胞(例如 T细胞) 进行的 治疗性应用。 转导的 T细胞可靶向肿瘤细胞的标志物 GPC3 , 协同激活 T细 胞, 引起 T细胞免疫应答, 从而显著提高其对肿瘤细胞的杀伤效率 。 因此, 本发明也提供了刺激 对哺乳动物 的靶细胞群或组织 的 T细胞介导的 免疫应答 的方法, 其包括以下步骤: 给哺乳动物施用本发明 的 CART细胞。 在一个实施 方式中, 本发明包括一类细胞 疗法, 分离病人自体 T细胞 (或 者异源供 体), 激活并进行基因改造产生 CAR-T细胞, 随后注入同一病人体内。 这种方式 患移植物抗宿主病 概率极低, 抗原被 T细胞以无 MHC限制方式识别。 此 外, 一种 CAR-T就可以治疗表达该抗原的所有癌症。 不像抗体疗法, CAR-T细胞 能够体 内复制, 产生可导致持续肿瘤控制 的长期持久性。 在一个实施 方式中, 本发明的 CAR-T细胞可经历稳固的体内 NK细胞扩展并 可持续 延长的时间量 。 另外, CAR介导的免疫应答可为过继免疫疗法步骤 的一 部分 , 其中 CAR -修饰 T细胞诱导对 CAR中的抗原结合结构域特异性的免疫应答。 例如, 抗 GPC3的 CART细胞引起抗 GPC3阳性的细胞的特异性免疫应答。 尽管本文公开 的数据具体公开 了包括抗 -GPC3 scFv、 CD8皎链和 CD8跨膜区 和胞 内区、 4-1BB胞内区和 CD3 &信号传导结构域的慢病毒载体, 但本发明应被 解释为包 括对构建体组成部 分中的每一个的任 何数量的变化。 可治疗的癌症 包括非实体瘤 (诸如血液学肿瘤, 例如白血病和淋巴瘤)或实 体瘤,尤其是实体瘤。 用本发明的 CAR治疗的癌症类型包括但不限于癌 、 胚细胞 瘤和肉瘤 , 和某些白血病或淋巴恶性肿瘤、 良性和恶性肿瘤、 和恶性瘤, 例如 肉瘤、 癌和黑素瘤。 也包括成人肿瘤 /癌症和儿童肿瘤/癌症。 血液学癌症为 血液或骨髓的癌症 。 血液学(或血原性)癌症的例子包括白血 病, 包括急性白血病 (诸如急性淋巴细胞白血病、 急性髓细胞白血病、 急性骨 髓性 白血病和成髓细胞 性、 前髓细胞性、 粒-单核细胞型、 单核细胞性和红白 血病)、 慢性白血病(诸如慢性髓细胞(粒细胞性)白血病、 慢性骨髓性白血病和 慢性淋 巴细胞白血病)、 真性红细胞增多症、 淋巴瘤、 霍奇金氏疾病、 非霍奇 金氏淋 巴瘤(无痛和高等级形式)、 多发性骨髓瘤、 瓦尔登斯特伦氏巨球蛋白血 症、 重链疾病、 骨髓增生异常综合征、 多毛细胞白血病和脊髓发 育不良。 实体瘤为通 常不包含囊肿或液体 区的组织的异常肿 块。 实体瘤可为良性或 恶性 的。 不同类型的实体瘤以形成它们 的细胞类型命 名(诸如肉瘤、 癌和淋巴 瘤)。 实体瘤诸如肉瘤和癌的例子包括膀胱癌 、 脑癌、 头颈癌、 胰腺癌、 肺癌、 乳腺癌 、 卵巢癌、 结肠癌、 前列腺癌和肾癌。 在优选的实施 方式中, 可治疗的癌症为 GPC3阳性肿瘤, 如肝细胞癌等。 本发明 的 CAR -修饰 T细胞也可用作对哺乳动物离体免疫和/或体内疗法 的 疫苗类 型。 优选地, 哺乳动物为人。 对于离体免疫 , 以下中的至少一项在将细胞施用进入 哺乳动物前在体 外发 生: i)扩增细胞, i i)将编码 CAR的核酸引入细胞, 和/或 i i i)冷冻保存细胞。 离体程序在 本领域中是公 知的, 并在以下更完全地进行 讨论。 简单地说, 细胞从 哺乳动物(优选人)中分离并用表达本文 公开的 CAR的载体进行基因修饰 (即, 体外转导或转染)。 CAR-修饰的细胞可被施用给哺乳动物接受者, 以提供 治疗益处 。 哺乳动物接受者可为人, 和 CAR-修饰的细胞可相对于接受者为自体 的。 可选地, 细胞可相对于接受者为 同种异基因 的、 同基因的(syngeneic)或 异种的 。 除了就离体 免疫而言使用基于 细胞的疫苗之外 , 本发明也提供了体内免疫 以引起针 对患者中抗原 的免疫应答的组合物 和方法。 本发明提供 了治疗肿瘤的方法 , 其包括施用给需要其的对象治疗有效 量的 本发明 的 CAR-修饰的 T细胞。 本发明 的 CAR-修饰的 T细胞可被单独施用或作为药物组合物 与稀释剂和/ 或与其他 组分诸如 IL-2、 IL-17或其他细胞因子或细胞群结合施用。简单地说, 本发明 的药物组合物可包括 如本文所述的靶细 胞群, 与一种或多种药学或生理 学上可接 受载体、 稀释剂或赋形剂结合。 这样的组合物可包括缓 冲液诸如中性 缓冲盐水 、 硫酸盐缓冲盐水等等; 碳水化合物诸如葡萄糖、 甘露糖、 蔗糖或葡 聚糖、甘露醇 ; 蛋白质; 多肽或氨基酸诸如甘氨酸; 抗氧化剂; 螯合剂诸如 EDTA 或谷胱甘 肽; 佐剂(例如, 氢氧化铝); 和防腐剂。 本发明的组合物优选配制用 于静脉 内施用。 本发明的药物 组合物可以以适 于待治疗(或预防)的疾病的方式施用。 施用 的数量和 频率将由这样 的因素确定, 如患者的病症、 和患者疾病的类型和严重 度 尽管适当的剂 量可由临床试验确 定。 当指出 “免疫学上有效量” 、 “抗肿瘤有效量” 、 “肿瘤-抑制有效量” 或“ 治疗量 ” 时, 待施用的本发明组合物的精确量可由医师确定, 其考虑患者 (对象)的年龄、 重量、 肿瘤大小、 感染或转移程度和病症的个体差异。 可通常 指出 : 包括本文描述的 T细胞的药物组合物可以以 1。4至 1。9个细胞 /kg体重的剂 量, 优选 10,至 io13个细胞 /kg体重的剂量(包括那些范围内的所有整数值)施用。 T细胞组合 物也可以以这 些剂量多次施用 。 细胞可通过使用免疫疗法 中公知的 注入技术 (见例如 Rosenberg等, NewEng. J. of Med. 319 : 1676, 1988)施用。 对 于具体 患者的最佳 剂量和治疗方 案可通过监 测患者的疾病 迹象并因此 调节治 疗由医学 领域技术人员容 易地确定。 对象组合物 的施用可以 以任何方便的方式 进行, 包括通过喷雾法、 注射、 吞咽、 输液、 植入或移植。 本文描述的组合物可被皮下、 皮内、 瘤内、 结内、 脊髓内 、 肌肉内、 通过静脉内(i. v.)注射或腹膜内施用给患者。 在一个实施方 式中 , 本发明的 T细胞组合物通过皮内或皮下注射被施 用给患者。 在另一个实 施方式 中, 本发明的 T细胞组合物优选通过 i. v.注射施用。 T细胞的组合物可被 直接注入 肿瘤, 淋巴结或感染位置。 在本发明 的某些实施方式中 , 利用本文描述的方法或本领域已知的其 他将 T细胞扩展 至治疗性水平 的方法活化和扩 展的细胞, 与任何数量的有 关治疗形 式结合 (例如, 之前、 同时或之后)施用给患者, 所述治疗形式包括但不限于用 以下试剂 进行治疗: 所述试剂诸如抗病毒疗 法、 西多福韦和白细胞介素- 2、 阿 糖胞背 (也己知为 ARA-C)或对 MS患者的那他珠单抗治疗或对牛皮癣患者的厄法 珠单抗 治疗或对 PML患者的其他治疗。 在进一步的实施方式中, 本发明的 T细胞 可与 以下结合使用: 化疗、 辐射、 免疫抑制剂, 诸如, 环抱菌素、 硫哩喋吟、 甲氨喋 吟、 麦考酚酯和 FK506, 抗体或其他免疫治疗剂。 在进一步的实施方式 中, 本发明的细胞组合物与 骨髓移植、 利用化疗剂诸如氟达拉滨 、 外部光束放 射疗法 (XRT)、环磷酰胺结合(例如, 之前、 同时或之后)而施用给患者。 例如, 在一个 实施方式中, 对象可经历高剂量化疗 的标准治疗, 之后进行外周血干细 胞移植 。 在一些实施方式中, 在移植后, 对象接受本发明的扩展的免疫细胞的 注入。在一个额 外的实施方式中 ,扩展的细胞在外科手术前或 外科手术后施用 。 施用给患 者的以上治疗 的剂量将随 着治疗病症 的精确属性和 治疗的接受 者而变化 。 人施用的剂量比例可根据本领域接 受的实践实施 。 通常, 每次治疗 或每个疗 程,可将 1 x 1。6个至 1 X 10"个本发明经修饰的 T细胞(如 , CAR-T细胞), 通过例如 静脉回输的方式 , 施用于患者。 根据本发 明, 本发明的药学产品 (药物、 药剂) 或药物组合物可以以任意 有效剂量 数施用给药受试者 。 优选地, 本发明的药学产品 (药物、 药剂) 或药 物组合 物可以以多次剂 量给药, 例如从约 2至约 20次剂量, 更优选从约 4T0次 剂量。 在特别优选的实施方 案, 在给药过程中, 以每三周给药约一次的频率将 本发明 的药学产品(药物、 药剂)或药物组合物给药至受试者, 例如注射、 输注 或口服 。 在特别优选的实施方案, 给药为通过注射施用至荷瘤 部位。 应当理解本 发明的药学产品 (药物、 药剂) 或药物组合物可以按用于通过 任意适宜 的途径给药的任意 适宜的方式配制 。 本发明的药 学产品 (药物、 药剂) 或药物组合物的剂量单位是基于常规进 行给药 受试者。 例如, 剂量单位可以给药多于每日一次、 每周一次、 每月一次 等。 剂量单位可以是以两次/ 周为基础给药, 即每周两次, 例如每三天一次。 本发明 的药学产品中所 包含的涉及 该药学产 品的说明书可 以含有如下 内 容: 适应症 (例如脑胶质母细胞瘤) 、 施用剂量 (例如上述所示例性说明的) 以及可 能产生的副作用等 等。 本发明的序 列
The design of CARs has gone through the following process: The first-generation CAR has only one intracellular signaling component CD3 & or Fc γ RI molecule, because there is only one activation domain in the cell, so it can only cause transient T cell proliferation and less The secretion of cytokines does not provide long-term T cell proliferation signals and sustained anti-tumor effects in vivo, so good clinical efficacy has not been achieved. The second-generation CARs introduce a co-stimulatory molecule based on the original structure, such as CD28, 4-1BB, OX40, and ICOS. Compared with the first-generation CARs, the function is greatly improved, and the persistence of CAR-T cells and the ability to protect tumor cells are further enhanced. lethality. On the basis of the second-generation CARs, some new immune co-stimulatory molecules such as CD27 and CD134 are connected in series to develop into the third-generation and fourth-generation CARs. The chimeric antigen receptor (CAR) of the present invention is a second-generation CAR, including an extracellular domain, a transmembrane domain, and an intracellular domain. The extracellular domain includes target-specific binding elements (also known as antigen-binding structures area). The intracellular domain includes the co-stimulatory signaling domain and the chain portion. A co-stimulatory signaling region refers to a portion of an intracellular domain that includes co-stimulatory molecules. Costimulatory molecules are cell surface molecules other than antigen receptors or their ligands that are required for an efficient response of lymphocytes to antigens. 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 the extracellular or cytoplasmic domain of a polypeptide chain. Linkers may comprise 0-300 amino acids, preferably 2 to 100 amino acids and most preferably 3 to 50 amino acids. In a preferred embodiment of the present invention, the extracellular domain of the CAR provided by the present invention includes an antigen-binding domain targeting GPC3. When the CAR of the present invention is expressed in T cells, it can perform antigen recognition based on antigen binding specificity. When it binds to its cognate antigen, it affects tumor cells, causing tumor cells not to grow, being induced to die, or otherwise affected, and resulting in a reduction or elimination of the patient's tumor burden. The antigen binding domain is preferably fused to an intracellular domain from one or more of a co-stimulatory molecule and a chain. The nucleic acid sequence of the vector encoding the desired molecule can be obtained using recombinant methods known in the art, such as, for example, by screening a library from cells expressing the gene, by obtaining the gene from a vector known to include the gene, or by using standard technology, directly isolated from cells and tissues containing the gene. Alternatively, the gene of interest can be produced synthetically. The present invention also provides a vector into which an expression cassette of the present invention is inserted. Vectors derived from retroviruses such as lentiviruses are suitable tools for long-term gene transfer because they allow long-term, stable integration of the transgene and its propagation in daughter cells. Lentiviral vectors have an advantage over vectors derived from oncogenic retroviruses, such as murine leukemia virus, because they can transduce non-proliferating cells, such as hepatocytes. They also have the advantage of low immunogenicity. Briefly summarized, the expression cassette or nucleic acid sequence of the present invention is usually operably linked to a promoter and incorporated into an expression vector. This vector is suitable for replication and integration in eukaryotic cells. A typical cloning vector contains transcriptional and translational terminators, an initial sequence and a promoter useful for regulating the expression of the desired nucleic acid sequence. The expression constructs of the invention can also be used in nucleic acid immunization and gene therapy using standard gene delivery protocols. Methods of gene delivery are known in the art. See, eg, US Patent Nos. 5,399,346, 5,580,859, 5,589,466, which are hereby incorporated by reference in their entirety. In another embodiment, the present invention provides gene therapy vectors. 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. Particular vectors of interest include expression vectors, replication vectors, probe generation vectors and sequencing vectors. Furthermore, expression vectors can be provided to cells in the form of viral vectors. Viral vector technology in this It is well known in the art and described, for example, in Sambrook et al. (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York) and other handbooks of virology and molecular biology. Viruses that can be used as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpesviruses, and lentiviruses. Generally, suitable vectors contain an origin of replication functional in at least one organism, a promoter sequence, convenient restriction enzyme sites, and one or more selectable markers (eg, WO01/96584; WO01/29058; and US Patent No. 6,326,193). A number of virus-based systems have been developed for gene transfer into mammalian cells. For example, retroviruses provide a convenient platform for gene delivery systems. The gene of choice can be inserted into a vector and packaged into retroviral particles using techniques known in the art. The recombinant virus can then be isolated and delivered to 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, lentiviral vectors are used. Additional promoter elements, such as enhancers, can regulate the frequency of transcription initiation. Typically these are located in the 30-110 bp region upstream of the initiation site, although it has recently been shown that many promoters also contain functional elements downstream of the initiation site. The spacing between promoter elements is often flexible in order to preserve promoter function when elements are inverted or moved relative to one another. In the thymidine kinase (tk) promoter, the spacing between promoter elements can be increased by 50 bp before activity begins to decline. Depending on the promoter, it appears that individual elements can act 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-1α (EF-1α) . However, other constitutive promoter sequences can also be used, including but not limited to the simian virus 40 (SV40) early promoter, mouse Mammary cancer virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, avian leukemia virus promoter, Epstein-Barr virus immediate early promoter, Lu Steiner's sarcoma virus promoter, and human gene promoters, such as but not limited to 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 capable of turning on expression of a polynucleotide sequence operably linked to an 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. In order to evaluate the expression of the CAR polypeptide or a part thereof, the expression vector introduced into the cell may also contain either or both of a selectable marker gene or a reporter gene, so as to seek transfected or infected cell populations from viral vectors Identification and selection of expressing cells. In other aspects, selectable markers can be carried on a single piece of DNA and used in a co-transfection procedure. Flanking both selectable marker and reporter gene
Figure imgf000012_0001
is well known in the field. See, eg, Sambrook et al. (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York) . o A preferred method for introducing polynucleotides into host cells is calcium phosphate transfection. Biological methods for introducing polynucleotides of interest into host cells include the use of DNA and RNA vectors. Viral vectors, especially retroviral vectors, have become the most widely used method for inserting genes into mammalian, eg human, cells. Other viral vectors can be derived from lentiviruses, poxviruses, herpes simplex virus I, adenoviruses, and adeno-associated viruses, among others. See, eg, U.S. Patent Nos. 5,350,674 and 5,585,362. Chemical means for 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 lipid-based systems. plastid. An exemplary colloidal system for use as an in vitro and in vivo delivery vehicle is a liposome (eg, an artificial membrane vesicle). Where a non-viral delivery system is used, an exemplary delivery vehicle is liposomes. The use of lipid formulations is contemplated for introducing nucleic acids into host cells (in vitro, ex vivo, or in vivo). In another aspect, the nucleic acid can be associated with a lipid. Lipid-associated nucleic acids can be encapsulated into the aqueous interior of liposomes, interspersed within the lipid bilayer of liposomes, attached via linker molecules associated with both liposomes and oligonucleotides into liposomes, entrapped in liposomes, complexed with liposomes, dispersed in a solution containing lipids, mixed with lipids, associated with lipids, contained in lipids as a suspension, contained in micelles or complexed with micelles, Or otherwise associated with lipids. 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 exist in bilayer structures, as micelles or have a "col lapsed" structure. They may also simply be dispersed in solution, possibly forming aggregates of non-uniform size or shape. Lipids are fatty substances, which may be naturally occurring or synthetic lipids. For example, lipids include fat droplets, which occur naturally in the cytoplasm as well as compounds comprising long-chain aliphatic cores and their derivatives such as fatty acids, alcohols, amines, aminoalcohols, and aldehydes. In a preferred embodiment of the present invention, the vector is a lentiviral vector. Therapeutic Applications The present invention includes therapeutic applications of cells (eg T cells) transduced with a lentiviral vector (LV) encoding a CAR of the present invention. The transduced T cells can target the tumor cell marker GPC3, activate T cells cooperatively, and cause T cell immune responses, thereby significantly improving their killing efficiency against tumor cells. Therefore, the present invention also provides a method for stimulating a T cell-mediated immune response to a target cell population or tissue in a mammal, comprising the following steps: administering the CART cells of the present invention to the mammal. In one embodiment, the present invention includes a type of cell therapy, in which a patient's own T cells (or a heterogeneous donor) are isolated, activated and genetically modified to produce CAR-T cells, and then injected into the same patient. In this way, the probability of suffering from graft-versus-host disease is extremely low, and the antigen is recognized by T cells in an MHC-free manner. In addition, one CAR-T can treat all cancers expressing the antigen. Unlike antibody therapies, CAR-T cells are able to replicate in vivo, resulting in long-term persistence that can lead to sustained tumor control. In one embodiment, CAR-T cells of the invention can undergo robust in vivo NK cell expansion for extended amounts of time. Additionally, the CAR-mediated immune response can be part of an adoptive immunotherapy step in which CAR-modified T cells induce an immune response specific for the antigen-binding domain in the CAR. For example, anti-GPC3 CART cells elicit a specific immune response against GPC3-positive cells. Although the data disclosed herein specifically discloses lentiviral vectors comprising anti-GPC3 scFv, CD8 chain and CD8 transmembrane and intracellular regions, 4-1BB intracellular region and CD3 & signaling domain, the present invention should be considered Interpretation is to include any number of variations to each of the construct components. Treatable cancers include non-solid tumors (such as hematological tumors, eg leukemias and lymphomas) or solid tumors, especially solid tumors. The types of cancer treated with the CAR of the present invention include, but are not limited to, carcinoma, blastoma and sarcoma, and certain leukemia or lymphoid malignancies, benign and malignant tumors, and malignant tumors, such as sarcoma, carcinoma and melanoma. Also includes adult tumors/cancers and childhood tumors/cancers. Hematological cancers are cancers of the blood or bone marrow. Examples of hematological (or hematogenous) cancers include leukemias, including acute leukemias such as acute lymphoblastic leukemia, acute myeloid leukemia, acute myelogenous leukemia, and myeloblastic, promyelocytic, myelomonocytic , monocytic and erythroleukemia), chronic leukemias such as chronic myeloid (granulocytic) leukemia, chronic myelogenous leukemia and chronic lymphocytic leukemia), polycythemia vera, lymphoma, Hodgkin's disease, non-Hodgkin's lymphoma (indolent and high-grade forms), multiple myeloma, Waldenstrom's macroglobulin leukemia, heavy chain disease, myelodysplastic syndrome, hairy cell leukemia, and myelodysplasia. Solid tumors are abnormal masses of tissue that usually do not contain cysts or areas of fluid. Solid tumors can be benign or malignant. The different types of solid tumors are named for the type of cells that form them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumors such as sarcomas and carcinomas include bladder cancer, brain cancer, head and neck cancer, pancreatic cancer, lung cancer, breast cancer, ovarian cancer, colon cancer, prostate cancer, and kidney cancer. In a preferred embodiment, the treatable cancer is a GPC3-positive tumor, such as hepatocellular carcinoma. The CAR-modified T cells of the present invention can 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 administering the cells into the mammal: i) expanding the cells, ii) introducing a nucleic acid encoding a CAR into the cells, and/or iii) cryopreserving the cells. 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 (ie, transduced or transfected in vitro) with a vector expressing a CAR disclosed herein. CAR-modified cells can be administered to mammalian recipients to provide therapeutic benefit. The mammalian recipient can be a human, and the CAR-modified cells can be autologous to the recipient. Alternatively, cells may be allogeneic, syngeneic or xenogeneic with respect to the recipient. In addition to the use of cell-based vaccines for ex vivo immunization, the invention also provides compositions and methods for in vivo immunization to elicit an immune response against an antigen in a patient. The present invention provides a method of treating a tumor comprising administering to a subject in need thereof a therapeutically effective amount of the CAR-modified T cells of the present invention. The CAR-modified T cells of the invention can be administered alone or as a pharmaceutical composition with a diluent and/or in combination with other components such as IL-2, IL-17 or other cytokines or cell populations. Briefly, the pharmaceutical composition of the present invention may include the target cell population as described herein combined 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, etc.; carbohydrates such as glucose, mannose, sucrose or dextran, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; agents such as EDTA or glutathione; adjuvants (eg, aluminum hydroxide); and preservatives. The compositions of the invention are preferably formulated for intravenous administration. The pharmaceutical composition of the present invention can be administered in a manner suitable for the disease to be treated (or prevented). The amount and frequency of administration will be determined by such factors as the patient's condition, and the type and severity of the patient's disease, although appropriate dosages can be determined by clinical trials. When an "immunologically effective amount", "antitumor effective amount", "tumor-suppressive effective amount" or "therapeutic amount" is indicated, the precise amount of a composition of the invention to be administered can be determined by a physician, taking into account the patient's Individual differences in (subjects') age, weight, tumor size, degree of infection or metastasis, and disease. It may generally be noted that: Pharmaceutical compositions comprising T cells described herein may be dosed at 1.4 to 1.9 cells/kg body weight, preferably at doses of 10, to 1013 cells/kg body weight (including those within the range All integer values) apply. T cell compositions can also be administered multiple times at these doses. Cells can be administered using infusion techniques well known in immunotherapy (see, eg, Rosenberg et al., New Eng. J. of Med. 319: 1676, 1988). The optimal dosage and treatment regimen for a particular patient can be readily determined by one skilled in the medical art by monitoring the patient for signs of disease and adjusting treatment accordingly. Administration of the composition to a subject can be performed in any convenient manner, including by spraying, injection, swallowing, infusion, implantation or implantation. The compositions described herein can be administered to a patient subcutaneously, intradermally, intratumorally, intranodally, intraspinally, intramuscularly, by intravenous (iv) injection or intraperitoneally. In one embodiment, a 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 iv injection. Compositions of T cells can be injected directly into tumors, lymph nodes or sites of infection. In certain embodiments of the invention, cells activated and expanded using the methods described herein or other methods known in the art to expand T cells to therapeutic levels are combined with any number of relevant treatment modalities (eg, previously , concurrently or subsequently) to the patient in the form of treatment including, but not limited to, treatment with agents such as antiviral therapy, cidofovir and interleukin-2, arabinocytosomes (also known ARA-C) or natalizumab in MS patients or erfatizumab in psoriasis or other treatments in PML. In a further embodiment, the T cells of the present invention can be used in combination with: chemotherapy, radiation, immunosuppressants, such as cyclosporine, thiamin, methotrein, mycophenolate mofetil and FK506, antibodies or other immunotherapeutic agents. In a further embodiment, the cell composition of the invention is administered in conjunction with (eg, before, simultaneously with or after) bone marrow transplantation, the use of chemotherapeutic agents such as fludarabine, external beam radiation therapy (XRT), cyclophosphamide patient. For example, in one embodiment, a subject may undergo standard treatment with high-dose chemotherapy followed by peripheral blood stem cell transplantation. In some embodiments, following transplantation, the subject receives an infusion of expanded immune cells of the invention. In an additional embodiment, the expanded cells are administered before or after surgery. Dosages administered to a patient for the above treatments will vary with the precise nature of the condition being treated and the recipient of the treatment. Dosage ratios for human administration can be implemented according to practice accepted in the art. Usually, 1 x 1.6 to 1 x 10 "modified T cells (for example, CAR-T cells) of the present invention can be administered to Patients. According to the present invention, the pharmaceutical product (drug, medicament) or pharmaceutical composition of the present invention can be administered to a subject with any effective dose. Preferably, the pharmaceutical product (drug, medicament) or pharmaceutical composition of the present invention It can be administered in multiple doses, for example, from about 2 to about 20 doses, more preferably from about 4 to 0 doses.In a particularly preferred embodiment, during the administration, the frequency of administration about once every three weeks will be The pharmaceutical product (drug, medicament) or pharmaceutical composition of the present invention is administered to the subject, such as injection, infusion or orally. In a particularly preferred embodiment, the administration is by injection into the tumor-bearing site. It should be understood that the pharmaceutical product (drug, medicament) or pharmaceutical composition of the present invention may be formulated in any suitable manner for administration by any suitable route. The dosage unit of the pharmaceutical product (drug, medicament) or pharmaceutical composition of the present invention is administered to a subject on a routine basis. For example, dosage units may be administered more than once daily, weekly, monthly, etc. The dosage unit may be administered on a twice/week basis, that is, twice a week, for example, once every three days. The instructions related to the pharmaceutical product contained in the pharmaceutical product of the present invention may contain the following contents: indications (eg, glioblastoma), dosage (eg, as exemplified above), and possible side effects, etc. . Sequences of the invention
GC52BB V序列: 信号肽 SP氨基酸: GC52BB V sequence: signal peptide SP amino acid:
MALPVTALLLPLALLLHAARPGS ( SEQ ID NO : 1 ) 信号肽 SP核酸:MALPVTALLLPLALLLLHAARPGS (SEQ ID NO: 1) signal peptide SP nucleic acid:
ATGGCCCTGCCTGTGACAGCCCTGCTGCTGCCTCTGGCCCTGCTGCTCCACGCCGCTAGACCCGGAAATGGCCCTGCCTGTGACAGCCCTGCTGCTGCCTCTGGCCCTGCTGCTCCACGCCGCTAGACCCGGAA
GC ( SEQ ID NO : 2 ) 重链 VH氨基酸:GC (SEQ ID NO: 2) heavy chain VH amino acid:
QIQLQQSGAELVRPGASVTLSCKASGYTFTDYEMHWVKQTPVHGLEWIGAIDPKTGGTAYNQKFKDQIQLQQSGAELVRPGASVTLSCKASGYTFTDYEMHWVKQTPVHGLEWIGAIDPKTGGTAYNQKFKD
KAILTADKSSSTAYMELRSLTSEDSAVYYCTRYYSYAYWGQGTLVTVSA ( SEQ ID NO : 3 ) 重链 VH核酸:KAILTADKSSSTAYMELRSLTSEDSAVYYCTRYYSYAYWGQGTLVTVSA (SEQ ID NO: 3) heavy chain VH nucleic acid:
CAGATCCAGCTGCAGCAGAGCGGCGCCGAGCTGGTGAGACCTGGCGCTTCCGTGACACTGTCCCAGATCCAGCTGCAGCAGAGCGGCGCCGAGCTGGTGAGACCTGGCGCTTCCGTGACACTGTCC
TGTAAGGCCAGCGGCTACACATTCACCGATTACGAGATGCACTGGGTGAAGCAGACACCCGTGCACG GCCTGGAGTGGATCGGCGCTATCGACCCTAAGACAGGCGGCACCGCCTACAATCAGAAGTTCAAGGA TAAGGCCATCCTGACCGCCGACAAGAGCTCCTCCACCGCCTACATGGAGCTGAGGTCCCTGACCTCC GAGGATTCCGCCGTGTACTACTGTACAAGATACTACAGCTACGCCTACTGGGGCCAGGGCACACTGGTGTAAGGCCAGCGGCTACACATTCACCGATTACGAGATGCACTGGGTGAAGCAGACACCCGTGCACGGCCTGGAGTGGATCGGCGCTATCGACCCTAAGACAGGCGGCACCGCCTACAATCAGAAGTTCAAGGATAAGGCCATCCTGACCGCCGACAAGAGCTCCTCCACCGCCTACATGGAGCTGAGGTCCCTGACCTCCGAGGATTCCGCCGTGT ACTACTGTACAAGATACTACAGCTACGCCTACTGGGGCCAGGGCACACTGG
TGACAGTGTCCGCC ( SEQ ID NO : 4) 接头 (Linker)氨基酸: TGACAGTGTCCGCC (SEQ ID NO: 4) linker (Linker) amino acid:
GGGGSGGGGSGGGGS ( SEQ ID NO : 5 ) 接头 (Linker)核酸:GGGGSGGGGSGGGGS (SEQ ID NO: 5) linker (Linker) nucleic acid:
GGCGGAGGCGGAAGCGGAGGAGGAGGAAGCGGCGGAGGCGGTAGC ( SEQ ID NO : 6) 轻链 VL氨基酸: GGCGGAGGCGGAAGCGGAGGAGGAGGAAGCGGCGGAGGCGGTAGC (SEQ ID NO: 6) Light chain VL amino acids:
DVVMTQTPLSLPVSLGDQAS ISCRSSQSPVHSNGNTYLHWYLQKPGQSPKLLIYKVSNRFSGVPDRDVVMTQTPLSLPVSLGDQAS ISCRSSQSPVHSNGNTYLHWYLQKPGQSPKLLIYKVSNRFSGVPDR
FSGSGSGTDFTLKISRVEAEDLGVYFCSQSTHVPYTFGGGTKLEIK ( SEQ ID NO : 7 ) 轻链 VL核酸: GATGTGGTGATGACCCAGACACCTCTGAGCCTGCCTGTGTCCCTGGGCGATCAGGCCTCCATC TCCTGCAGATCCAGCCAGAGCCCCGTGCACAGCAATGGCAATACATACCTGCACTGGTACCTGCAGA AGCCTGGCCAGAGCCCTAAGCTGCTGATCTACAAGGTGAGCAACAGATTCAGCGGCGTGCCCGACAG ATTCTCCGGCAGCGGCTCCGGCACAGACTTCACACTGAAGATCAGCAGGGTGGAGGCCGAGGACCTG GGCGTGTACTTCTGCAGCCAGAGCACCCACGTGCCCTACACCTTCGGCGGCGGCACCAAGCTGGAGA TCAAG ( SEQ ID NO : 8 ) 钗链区 Hinge氨基酸:FSGSGSGTDFTLKISRVEAEDLGVYFCSQSTHVPYTFGGGTKLEIK (SEQ ID NO: 7) light chain VL nucleic acid: GATGTGGTGATGACCCAGACACCTCTGAGCCTGCCTGTGTCCCTGGGCGATCAGGCCTCCATCTCCTGCAGATCCAGCCAGAGCCCCGTGCACAGCAATGGCAATACATACCTGCACTGGTACCTGCAGA AGCCTGGCCAGAGCCCTAAGCTGCTGATCTACAAGGTGAGCAACAGATTCAGCGGCGTGCCCGACAGATTCTCCGGCAGCGGCTCCGGC ACAGACTTCACACTGAAGATCAGCAGGGTGGAGGCCGAGGACCTGGGCGTGTACTTCTGCAGCCAGAGCACCCACGTGCCCTACACCTTCGGCGGCGGCACCAAGCTGGAGA TCAAG (SEQ ID NO: 8) Hinge amino acid in the hairpin chain region:
SGTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIY ( SEQ ID NO : 9 ) 钗链区 Hinge核酸:SGTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIY (SEQ ID NO: 9) Hinge nucleic acid of chain region:
TCCGGCACAACCACCCCTGCCCCCAGACCCCCTACCCCAGCTCCTACAATCGCCAGCCAGCCC CTGAGCCTCAGGCCTGAGGCCTGCAGGCCCGCTGCTGGAGGAGCTGTGCACACCAGGGGCCTGGACT TCGCCTGTGACATCTAC ( SEQ ID NO : 10) 跨膜区 TM氨基酸: TCCGGCACAACCACCCCTGCCCCCAGACCCCCTACCCCAGCTCCTACAATCGCCAGCCAGCCCCTGAGCCTCAGGCCTGAGGCCTGCAGGCCCGCTGCTGGAGGAGCTGTGCACACCAGGGGCCTGGACT TCGCCTGTGACATCTAC (SEQ ID NO: 10) Transmembrane region TM amino acids:
IWAPLAGTCGVLLLSLVITLYC ( SEQ ID NO : 11 ) 跨膜区 TM核酸:IWAPLAGTCGVLLLSLVITLYC (SEQ ID NO: 11) Transmembrane Region TM Nucleic Acid:
ATCTGGGCCCCCCTGGCCGGCACCTGTGGAGTTCTGCTGCTGTCCCTGGTGATCACACTGTAC TGC ( SEQ ID NO : 12 ) 共刺激结构域 4-1BB氨基酸:ATCTGGGCCCCCCTGGCCGGCACCTGTGGAGTTCTGCTGCTGTCCCTGGTGATCACACTGTAC TGC (SEQ ID NO: 12) co-stimulatory domain 4-1BB amino acids:
RFSVVKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL ( SEQ ID NO : 13 ) 共刺激结构域 4- IBB核酸:RFSVVKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL (SEQ ID NO: 13) costimulatory domain 4-IBB nucleic acid:
AGATTCTCCGTGGTGAAGAGAGGCAGAAAGAAGCTGCTGTACATCTTCAAGCAGCCCTTCATG AGGCCTGTGCAGACAACACAGGAGGAGGATGGCTGTTCCTGCAGATTCCCCGAGGAGGAGGAGGGCG GCTGTGAGCTG ( SEQ ID NO : 14) 胞内信号 CD3 V氨基酸:AGATTCTCCGTGGTGAAGAGAGGCAGAAAGAAGCTGCTGTACATCTTCAAGCAGCCCTTCATG AGGCCTGTGCAGACAACACAGGAGGAGGATGGCTGTTCCTGCAGATTCCCCGAGGAGGAGGAGGGCG GCTGTGAGCTG (SEQ ID NO: 14) Intracellular signal CD3 V amino acid:
RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKM AEAYSE I GMKGERRRGKGHDGLYQGLS TATKDTYDALHMQALPPR ( SEQ ID NO : 15 ) 胞内信号 CD3,核酸:RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKM AEAYSE I GMKGERRRGKGHDGLYQGLS TATKDTYDALHMQALPPR (SEQ ID NO: 15) intracellular signal CD3, nucleic acid:
AGAGTGAAGTTCAGCAGATCCGCCGATGCCCCCGCCTACCAGCAGGGACAGAATCAGCTGTAC AACGAGCTGAACCTGGGCAGAAGAGAGGAGTACGATGTGCTGGATAAGAGGAGGGGCAGGGACCCTG AGATGGGCGGCAAGCCCAGGAGGAAGAACCCCCAGGAGGGCCTGTACAACGAACTGCAGAAGGACAA GATGGCCGAGGCCTACAGCGAGATCGGCATGAAGGGCGAGAGAAGAAGAGGCAAGGGCCACGACGGC CTGTACCAGGGCCTGTCCACAGCCACCAAGGACACATACGACGCCCTGCACATGCAGGCCCTGCCTC CCAGATGA ( SEQ ID NO : 16) AGAGTGAAGTTCAGCAGATCCGCCGATGCCCCCGCCTACCAGCAGGGACAGAATCAGCTGTAC AACGAGCTGAACCTGGGCAGAAGAGAGGAGTACGATGTGCTGGATAAGAGGAGGGGCAGGGACCCTGAGATGGGCGGCAAGCCCAGGAGGAAGAACCCCCAGGAGGGCCTGTACAACGAACTGCAGAAGGACAA GATGGCCGAGG CCTACAGCGAGATCGGCATGAAGGGCGAGAGAAGAAGAGGCAAGGGCCACGACGGCCTGTACCAGGGCCTGTCCACAGCCACCAAGGACACATACGACGCCCTGCACATGCAGGCCCTGCCTCCCAGATGA (SEQ ID NO: 16)
GC52BB V CAR的氨基酸序列: Amino acid sequence of GC52BB V CAR:
MALPVTALLLPLALLLHAARPGSQIQLQQSGAELVRPGASVTLSCKASGYTFTDYEMHWVKQTPV HGLEWIGAIDPKTGGTAYNQKFKDKAILTADKSSSTAYMELRSLTSEDSAVYYCTRYYSYAYWGQGTLVTVMALPVTALLLPLALLLLHAARPGSQIQLQQSGAELVRPGASVTLSCKASGYTFTDYEMHWVKQTPV HGLEWIGAIDPKTGGTAYNQKFKDKAILTADKSSSTAYMELRSLTSEDSAVYYCTRYYSYAYWGQGTLVTV
SAGGGGSGGGGSGGGGSDVVMTQTPLSLPVSLGDQASISCRSSQSPVHSNGNTYLHWYLQKPGQSPKLLISAGGGGSGGGGSGGGGSDVVMTQTPLSLPVSLGDQASISCRSSQSPVHSNGNTYLHWYLQKPGQSPKLLI
YKVSNRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYFCSQSTHVPYTFGGGTKLEIKSGTTTPAPRPPYKVSNRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYFCSQSTHVPYTFGGGTKLEIKSGTTTPAPPRPP
TPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCRFSVVKRTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCRFSVVKR
GRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNELNLGGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNELNLG
RREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLS
TATKDTYDALHMQALPPR ( SEQ ID NO : 17 ) TATKDTYDALHMQALPPR (SEQ ID NO: 17)
GC52BB, CAR的核酸序列:GC52BB, the nucleic acid sequence of CAR:
ATGGCCCTGCCTGTGACAGCCCTGCTGCTGCCTCTGGCCCTGCTGCTCCACGCCGCTAGACCCGGAAATGGCCCTGCCTGTGACAGCCCTGCTGCTGCCTCTGGCCCTGCTGCTCCACGCCGCTAGACCCGGAA
GCCAGATCCAGCTGCAGCAGAGCGGCGCCGAGCTGGTGAGACCTGGCGCTTCCGTGACACTGTCCTGGCCAGATCCAGCTGCAGCAGAGCGGCGCCGAGCTGGTGAGACCTGGCGCTTCCGTGACACTGTCCTG
TAAGGCCAGCGGCTACACATTCACCGATTACGAGATGCACTGGGTGAAGCAGACACCCGTGCACGGCTAAGGCCAGCGGCTACACATTCACCGATTACGAGATGCACTGGGTGAAGCAGACACCCGTGCACGGC
CTGGAGTGGATCGGCGCTATCGACCCTAAGACAGGCGGCACCGCCTACAATCAGAAGTTCAAGGATACTGGAGTGGATCGGCGCTATCGACCCTAAGACAGGCGGCACCGCCTACAATCAGAAGTTCAAGGATA
AGGCCATCCTGACCGCCGACAAGAGCTCCTCCACCGCCTACATGGAGCTGAGGTCCCTGACCTCCGAAGGCCATCCTGACCGCCGACAAGAGCTCCTCCACCGCCTACATGGAGCTGAGGTCCCTGACCTCCGA
GGATTCCGCCGTGTACTACTGTACAAGATACTACAGCTACGCCTACTGGGGCCAGGGCACACTGGTGGGATTCCGCCGTGTACTACTGTACAAGATACTACAGCTACGCCTACTGGGGCCAGGGCACACTGGTG
ACAGTGTCCGCCGGCGGAGGCGGAAGCGGAGGAGGAGGAAGCGGCGGAGGCGGTAGCGATGTGGTGAACAGTGTCCGCCGGCGGAGGCGGAAGCGGAGGAGGAGGAAGCGGCGGAGGCGGTAGCGATGTGGTGA
TGACCCAGACACCTCTGAGCCTGCCTGTGTCCCTGGGCGATCAGGCCTCCATCTCCTGCAGATCCAGTGACCCAGACACCTCTGAGCCTGCCTGTGTCCCTGGGCGATCAGGCCTCCATCTCCTGCAGATCCAG
CCAGAGCCCCGTGCACAGCAATGGCAATACATACCTGCACTGGTACCTGCAGAAGCCTGGCCAGAGCCCAGAGCCCCGTGCACAGCAATGGCAATACATACCTGCACTGGTACCTGCAGAAGCCTGGCCAGAGC
CCTAAGCTGCTGATCTACAAGGTGAGCAACAGATTCAGCGGCGTGCCCGACAGATTCTCCGGCAGCGCCTAAGCTGCTGATCTACAAGGTGAGCAACAGATTCAGCGGCGTGCCCGACAGATTCTCCGGCAGCG
GCTCCGGCACAGACTTCACACTGAAGATCAGCAGGGTGGAGGCCGAGGACCTGGGCGTGTACTTCTGGCTCCGGCACAGACTTCACACTGAAGATCAGCAGGGTGGAGGCCGAGGACCTGGGCGTGTACTTCTG
CAGCCAGAGCACCCACGTGCCCTACACCTTCGGCGGCGGCACCAAGCTGGAGATCAAGTCCGGCACACAGCCAGAGCACCCACGTGCCCTACACCTTCGGCGGCGGCACCAAGCTGGAGATCAAGTCCGGCACA
ACCACCCCTGCCCCCAGACCCCCTACCCCAGCTCCTACAATCGCCAGCCAGCCCCTGAGCCTCAGGCACCACCCCTGCCCCCAGACCCCCTACCCCAGCTCCTACAATCGCCAGCCAGCCCCTGAGCCTCAGGC
CTGAGGCCTGCAGGCCCGCTGCTGGAGGAGCTGTGCACACCAGGGGCCTGGACTTCGCCTGTGACATCTGAGGCCTGCAGGCCCGCTGCTGGAGGAGCTGTGCACACCAGGGGCCTGGACTTCGCCTGTGACAT
CTACATCTGGGCCCCCCTGGCCGGCACCTGTGGAGTTCTGCTGCTGTCCCTGGTGATCACACTGTACCTACATCTGGGCCCCCCTGGCCGGCACCTGTGGAGTTCTGCTGCTGTCCCTGGTGATCACACTGTAC
TGCAGATTCTCCGTGGTGAAGAGAGGCAGAAAGAAGCTGCTGTACATCTTCAAGCAGCCCTTCATGATGCAGATTCTCCGTGGTGAAGAGAGGCAGAAAGAAGCTGCTGTACATCTTCAAGCAGCCCTTCATGA
GGCCTGTGCAGACAACACAGGAGGAGGATGGCTGTTCCTGCAGATTCCCCGAGGAGGAGGAGGGCGGGGCCTGTGCAGACAACACAGGAGGAGGATGGCTGTTCCTGCAGATTCCCCGAGGAGGAGGAGGGCGG
CTGTGAGCTGAGAGTGAAGTTCAGCAGATCCGCCGATGCCCCCGCCTACCAGCAGGGACAGAATCAGCTGTGAGCTGAGAGTGAAGTTCAGCAGATCCGCCGATGCCCCCGCCTACCAGCAGGGACAGAATCAG
CTGTACAACGAGCTGAACCTGGGCAGAAGAGAGGAGTACGATGTGCTGGATAAGAGGAGGGGCAGGGCTGTACAACGAGCTGAACCTGGGCAGAAGAGAGGAGTACGATGTGCTGGATAAGAGGAGGGGCAGGG
ACCCTGAGATGGGCGGCAAGCCCAGGAGGAAGAACCCCCAGGAGGGCCTGTACAACGAACTGCAGAAACCCTGAGATGGGCGGCAAGCCCAGGAGGAAGAACCCCCAGGAGGGCCTGTACAACGAACTGCAGAA
GGACAAGATGGCCGAGGCCTACAGCGAGATCGGCATGAAGGGCGAGAGAAGAAGAGGCAAGGGCCACGGACAAGATGGCCGAGGCCTACAGCGAGATCGGCATGAAGGGCGAGAGAAGAAGAGGCAAGGGCCAC
GACGGCCTGTACCAGGGCCTGTCCACAGCCACCAAGGACACATACGACGCCCTGCACATGCAGGCCCGACGGCCTGTACCAGGGCCTGTCCACAGCCACCAAGGACACATACGACGCCCTGCACATGCAGGCCC
TGCCTCCCAGATGA ( SEQ ID NO : 18 )
Figure imgf000018_0001
(2) 本发明的 CAR-T细胞能够高效靶向 GPC3 , 施用剂量在 1 X 106/只老鼠或 更低时 , CAR-T就能有很好的抑瘤效果; 而目前实体瘤 CAR-T的施用剂量一般都 在 5 X 10&/只老鼠或以上才能达到很好抑瘤效果;
TGCCTCCCAGATGA (SEQ ID NO: 18)
Figure imgf000018_0001
(2) The CAR-T cells of the present invention can efficiently target GPC3, and when the dose is 1 X 10 6 /mouse or lower, CAR-T can have a good tumor inhibitory effect; while the current solid tumor CAR-T The dosage of T is generally 5 X 10&/mouse or above to achieve a good tumor inhibitory effect;
(3) 使用比较低的剂量就能达到良好的 治疗效果, 为后期工艺和产业化提 供了更 佳的可能性和 便捷性, 也使得 CAR- T产品更加安全, 更少的发生由于大 量细胞 回输带来的毒副反应 。 下面结合具体 实施例, 进一步阐述本发明。 应理解, 这些实施例仅用于说 明本发 明而不用于限制本 发明的范围。 下列实施例中未注明具体 条件的实验方 法,通常按照常规 条件,例如 Sambrook等人, 分子克隆:实验室手册 (New York : Cold Spring Harbor Laboratory Press, 1989)中所述的条件, 或按照制造厂 商所建 议的条件。除非另外说明,否则百分 比和份数是重量百 分比和重量份数 。
Figure imgf000019_0001
实施例 2 HuH7-Luc、 HepG2-Luc细胞构建与检测 通过查询 luc iferase基因 ( GenBank : ACF93193. 1 ) 序列, 在 CR0公司合成 并构建至 PLVX-Puro载体中, 构建好的载体命名为 PLVX-Luc-Puroo 通过慢病毒 转染 的方式 构建稳 转细胞 系, 即使用 pGPol、 pVSVG包装质粒和目的质 粒 PLVX-Luc-Puro, 在公司自有慢病毒包装平 台, 生产出高滴度慢病毒, 然后按 照 M0I = l分别感染靶细胞 HuH7, HepG2 o 通过几轮 Purmycin的加压筛选后进行检 测, 合格的细胞系 luciferase信号值应为未转染慢病毒的原 始细胞的 100倍以 上, 将合格的细胞分 别命名为 HuH7-Luc. HepG2-Luc; luciferase信号检测结 果如 图 2所示。
Figure imgf000019_0002
GC76BB E、 4 u g pGPol和 2 u g pVSVG质粒, 移液枪上下吹打充分混匀后, 加入 18 u L PEL 立即用移液器上下吹打混 匀, 室温下静置 15分钟;
(3) A good therapeutic effect can be achieved with a relatively low dose, which provides better possibility and convenience for later-stage technology and industrialization, and also makes CAR-T products safer, and less likely to occur due to a large number of cell regeneration. Toxic and side effects caused by infusion. The present invention will be further described below in conjunction with specific examples. It should be understood that these examples are only used to illustrate the present invention and are not intended to limit the scope of the present invention. The experimental method that does not indicate specific conditions in the following examples is usually according to conventional conditions, such as Sambrook et al., Molecular cloning: the conditions described in the laboratory manual (New York: Cold Spring Harbor Laboratory Press, 1989), or according to the manufacturing conditions recommended by the manufacturer. Percentages and parts are by weight unless otherwise indicated.
Figure imgf000019_0001
Example 2 HuH7-Luc, HepG2-Luc cell construction and detection By querying the luciferase gene (GenBank: ACF93193.1) sequence, it was synthesized in CRO company and constructed into the PLVX-Puro vector, and the constructed vector was named PLVX-Luc -Puro o Construct a stable transfected cell line by lentivirus transfection, that is, use the pGPol, pVSVG packaging plasmid and the target plasmid PLVX-Luc-Puro to produce high-titer lentivirus on the company's own lentivirus packaging platform, and then Infect the target cells HuH7 and HepG2 respectively according to MOI = 1. After several rounds of Purmycin pressurized screening, the luciferase signal value of the qualified cell line should be more than 100 times that of the original cells not transfected with lentivirus. The qualified cells They were named HuH7-Luc and HepG2-Luc respectively; the results of luciferase signal detection are shown in Figure 2.
Figure imgf000019_0002
GC76BB E, 4 ug pGPol and 2 ug pVSVG plasmids, pipette up and down to mix thoroughly, add 18 uL PEL, immediately pipette up and down to mix, and let stand at room temperature for 15 minutes;
(4) 将上述 DNA/PEI复合物逐滴加入到 15cm培养皿中, 轻轻晃动培养皿, 充分混 匀。 将培养皿置于 37°C、 5 % CO?培养箱, 培养 6〜 8小时后, 将含有转染 试剂的培 养基去掉, 更换为新鲜的完全培养 基; (4) Add the above-mentioned DNA/PEI complex dropwise into a 15cm petri dish, shake the petri dish gently, and mix well. Place the culture dish in a 37°C, 5% CO incubator, and after culturing for 6 to 8 hours, remove the medium containing the transfection reagent and replace it with fresh complete medium;
(5) 连续培养 48小时后,收集培养皿中含有病毒的培养基上清,用 0. 45 u m 的滤膜过 滤, 然后 4P条件 20000g离心 2小时; 离心结束后, 小心将离心管中的 液体吸 去, 加入 400 u L PBS缓冲液将沉淀重悬, 分装病毒至 1. 5ml离心管中, 每管 100 H 1 , 将病毒置于- 80°C条件保存。 实施例 4 T细胞的分选与激活 按照以下步骤 进行 T细胞分选与激活: (5) After 48 hours of continuous culture, collect the medium supernatant containing the virus in the culture dish, filter it with a 0.45 μm filter membrane, and then centrifuge at 20000g for 2 hours under 4P conditions; after the centrifugation, carefully remove the liquid in the centrifuge tube Aspirate, add 400 uL of PBS buffer to resuspend the pellet, aliquot the virus into 1.5ml centrifuge tubes, each tube is 100 H 1 , and store the virus at -80°C. Example 4 Sorting and activation of T cells Follow the steps below to sort and activate T cells:
(1) 取 1只冻存 PBMC细胞, 37°C水浴复苏; 将复苏的 PBMC加入到己预先加 AlOml 预热培养基的 15ml离心管中; 然后 500g离心 5分钟; (1) Take 1 frozen PBMC cell and resuscitate in a 37°C water bath; add the resuscitated PBMC to a 15ml centrifuge tube that has been pre-added with AlOml preheated medium; then centrifuge at 500g for 5 minutes;
(2) 用分选缓冲液重悬细胞后, 进行细胞计数; (2) After resuspending cells with sorting buffer, perform cell counting;
(3) 500g离心 5分钟, 调整细胞密度为 1. 0*107/ml; (3) Centrifuge at 500g for 5 minutes, and adjust the cell density to 1.0*10 7 /ml;
(4) 按照 1. 0*10’的细胞加入 20 Li 1体积的分选磁珠量, 加入分选磁珠;(4) According to the cells of 1.0*10', add 20 Li 1 volume of sorting magnetic beads, and add sorting magnetic beads;
(5) 4°C条件下, 孵育 15分钟; (5) Incubate for 15 minutes at 4°C;
(6) 将孵育好的细胞加入分选柱中执行分 选操作; (6) Add the incubated cells into the sorting column to perform the sorting operation;
(7) 收集分选柱中流出液, 500g离心 5分钟; (7) Collect the effluent in the sorting column, and centrifuge at 500g for 5 minutes;
(8) 将细胞重悬于 X-vivo 15培养基 (含 300U/mL IL - 2 ) 中, 计数, 并调 整细胞密 度为 1. O^lOVml; (8) resuspend the cells in X-vivo 15 medium (containing 300U/mL IL-2), count, and adjust the cell density to 1.0^lOVml;
(9) 按照 1. 0*106细胞加入 10 11 1激活磁珠的比例, 加入激活磁珠, 然后将 细胞置于 37 °C . 5 % CO?培养箱培养 48小时。 从而获得激活 后的 T细胞。
Figure imgf000020_0001
而获得 CAR-T细胞。 实施例 6 CAR-T细胞阳性率检测
(9) According to the ratio of 1.0*10 6 cells to 10 11 1 activated magnetic beads, add activated magnetic beads, and then place the cells in a 37°C. 5% CO? incubator for 48 hours. Thus, activated T cells are obtained.
Figure imgf000020_0001
And obtain CAR-T cells. Example 6 Detection of positive rate of CAR-T cells
CAR-T细胞阳性率的检测方 法如下所述: The detection method of CAR-T cell positive rate is as follows:
(1) 500g离心 5分钟, 收集实施例 5中感染慢病毒第 7天后的 CAR- T细胞;(1) Centrifuge at 500g for 5 minutes, and collect the CAR-T cells infected with lentivirus on the 7th day in Example 5;
(2) 500 uL PBS重悬 CAR-T细胞, 500g离心 5分钟洗涤细胞一次; (2) Resuspend CAR-T cells in 500 uL PBS, centrifuge at 500g for 5 minutes to wash the cells once;
(3) 1 mL PBS重悬细胞, 取 2011 L重悬后细胞于细胞计数仪上计数;(3) Cells were resuspended in 1 mL of PBS, and 2011 L of resuspended cells were counted on a cell counter;
(4) 按照细胞计数结果, 取 5*107反应细胞进行 CART阳性率检测;(4) According to the cell counting results, 5*107 reaction cells were taken to detect the positive rate of CART;
(5) 按照 2 u g/ml浓度加检测抗体, 反应体积为 lOOuL, 然后 4°C条件下, 孵育 30分钟; (5) Add the detection antibody at a concentration of 2 μg/ml, the reaction volume is 100uL, and then incubate at 4°C for 30 minutes;
(6) 孵育结束后, 500g离心 5分钟; (6) After the incubation, centrifuge at 500g for 5 minutes;
(7) 500 uL PBS重悬细胞, 500g离心 5分钟洗涤细胞一次; (7) Resuspend the cells in 500 uL PBS, centrifuge at 500g for 5 minutes to wash the cells once;
(8) 重复步骤 7操作; (8) Repeat step 7;
(9) 200 uL PBS重悬细胞, 进行上机检测。 结果如图 3所示, CAR-T细胞阳性率为 68.79%。 实施例 7 CAR-T细胞对靶细胞杀伤 (9) Cells were resuspended in 200 uL PBS for testing on the machine. The results are shown in Figure 3, the positive rate of CAR-T cells was 68.79%. Example 7 CAR-T cells kill target cells
(1) 收集对数生长期靶细胞 (HuH7Tuc或 HepG2-luc) , 调整靶细胞密度 至 2*107ml, 取一块新的 96孔板, 按照 50UL/孔的量接种靶细胞。 96孔板四周 未用的孔 , 每孔加入 100P L培养基, 以防中间的实验孔水分蒸发。 (1) Collect logarithmic growth phase target cells (HuH7Tuc or HepG2-luc), adjust the target cell density to 2*107ml, take a new 96-well plate, and inoculate the target cells according to the amount of 50UL/well. To the unused wells around the 96-well plate, 100 PL medium was added to each well to prevent water evaporation in the middle experimental wells.
(2) 离心收集上述制备的 CAR- T细胞, 用 X-vivo 15培养基 (含 300U/mL IL -2) ; 然后按照设计的效靶比 (E/T) , 每孔加入 50U 1 CAR-T细胞至 96孔板。 设置阳 性对照 BM (GPC3抗体 GC33的 scFv构建的 CAR-T) ; UN-T为未转导慢病毒 的 T细胞, 所有细胞通过相同制备操作。 (2) Collect the above-prepared CAR-T cells by centrifugation, use X-vivo 15 medium (containing 300U/mL IL-2); then add 50U 1 CAR-T cells to each well according to the designed effect-to-target ratio (E/T). T cells to 96-well plate. Positive control BM (CAR-T constructed by scFv of GPC3 antibody GC33) was set; UN-T was T cells not transduced with lentivirus, and all cells were prepared through the same operation.
(3) 将孔板置于 5% C02 37°C培养箱中, 培养 14-18小时。 (3) Place the well plate in a 5% CO 2 37°C incubator and incubate for 14-18 hours.
(4) 培养结束后,将孔板从培养箱中取出,加入 50u 1 One-Gio luciferase 检测底物 300g室温离心 96孔板 3分钟, 轻轻的将板取出, 在酶标仪上读数。 (4) After the incubation, take the well plate out of the incubator, add 50u 1 One-Gio luciferase detection substrate, centrifuge the 96-well plate at room temperature for 3 minutes at 300g, gently take the plate out, and read it on a microplate reader.
(5) 通过酶标仪数据, 计算 CAR- T细胞对靶细胞杀伤效率; 计算公式: 杀 伤% =1-(样品读值 -Min) / (Max-Min)。 (5) Calculate the killing efficiency of CAR-T cells on target cells by using the microplate reader data; Calculation formula: killing % =1-(sample reading value-Min)/(Max-Min).
GC52BB V体外杀伤 HuH7-luc细胞、 HepG2-luc细胞的结果如图 4、图 5所示。 结果表明 , 对于 HuH7-luc细胞, GC52BBV体外杀伤活性在 E/T (效靶比) 为 4时, GC52BBC能够杀伤将近 100%的肿瘤细胞, 而 BM阳性对照 CAR-T只有 60% 的杀伤 效果; 即使在 E/T (效靶比)为 1时, GC52BBV也有将近 40%的杀伤能力, 此时的 BM阳性对照 CAR-T几乎看不出无杀伤效果 (图 4); 对于 HepG2-luc细胞, GC52BBC体 外杀伤活性在 E/T (效靶比) 为 4时, GC52BB £和 BM对照 CAR-T都能 够 杀伤将近 100%的肿瘤细胞, 但在 E/T (效靶比) 为 1时, GC52BB E还能有 80% 的杀伤能力, 此时的 BM阳性对照 CAR-T杀伤效果已经降至 50%以下 (图 5) 。
Figure imgf000022_0001
剂 量 1x107只小鼠,其中 Mock T组只设置高剂量 3x10°组;分组当天定义为 D1天, 并 在分组当天给药处 理。 实验观 察和数据采集:给药后,每周常规监测 肿瘤对动物正常 行为的影响。 具 体内容有实验动物 的活动性,摄食和饮水情况 ,体重增加或降低情况,眼睛、 被 毛及其它异常情况 。 测量并记录肿瘤大小和小鼠体重 。 肿瘤体 积计算方式为 : 肿瘤体积(mm3) = 0. 5 X (肿瘤长径 X 肿瘤短径 ?)。 结果如 图 6所示, 本发明的 CART细胞在较低的给药剂量下(1x10° /只小鼠), 就 具有优异的抑瘤效 果。 在本发 明提及的所有文献 都在本申请中 引用作为参考, 就如同每一篇文献 被 单独引用作为参考 那样。此外应理解,在阅读了本发 明的上述讲授 内容之后, 本 领域技术人员可 以对本发明作各种 改动或修改, 这些等价形式同样落于本 申 请 所附权利要求书所 限定的范围。
The results of GC52BB V killing HuH7-luc cells and HepG2-luc cells in vitro are shown in Figure 4 and Figure 5 . The results showed that for HuH7-luc cells, when the in vitro killing activity of GC52BBV was 4 when the E/T (effect-to-target ratio) was 4, GC52BBC could kill nearly 100% of tumor cells, while the BM positive control CAR-T had only 60% killing effect; Even when the E/T (effect-to-target ratio) is 1, GC52BBV has a killing ability of nearly 40%. At this time, the BM positive control CAR-T has almost no killing effect (Figure 4); for HepG2-luc cells, When the killing activity of GC52BBC in vitro was E/T (effect-to-target ratio) of 4, both GC52BB £ and BM control CAR-T could It can kill nearly 100% of tumor cells, but when the E/T (effect-to-target ratio) is 1, GC52BB E can still have 80% killing ability, and at this time, the killing effect of BM positive control CAR-T has dropped to 50% following (Figure 5).
Figure imgf000022_0001
The dose was 1x107 mice, and the Mock T group only set up a high-dose 3x10° group; the day of grouping was defined as D1 day, and the drug was administered on the day of grouping. Experimental observation and data collection: After administration, the effect of tumor on the normal behavior of animals was routinely monitored weekly. The specific content includes the activity of experimental animals, food intake and drinking conditions, weight gain or loss, eyes, coat and other abnormal conditions. Measure and record tumor size and mouse body weight. The calculation method of tumor volume is: tumor volume (mm 3 ) = 0.5 X (tumor long diameter X tumor short diameter?). The results are shown in Figure 6, the CART cells of the present invention have an excellent tumor inhibitory effect at a lower dosage (1x10°/mouse). All documents mentioned in this application are incorporated by reference in this application as if each were individually incorporated by reference. In addition, it should be understood that after reading the above teaching content of the present invention, those skilled in the art may make various changes or modifications to the present invention, and these equivalent forms also fall within the scope defined by the appended claims of the present application.

Claims

22 权 利 要 求
Figure imgf000023_0001
白的跨膜 区: ICOS、 CD28、 CD3 eps i lon. CD45、 CD4、 CD5、 CD8、 CD9、 CD16、 GD2、 CD33、 CD37、 CD64、 CD80、 CD86、 CD134、 CD137、 CD154、 或其组合。
22 claims
Figure imgf000023_0001
White transmembrane region: ICOS, CD28, CD3 eps i lon. CD45, CD4, CD5, CD8, CD9, CD16, GD2, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154, or a combination thereof.
7. 如权利要求 4所述的 CAR构建物,其特征在于, 所述的 C为选自下组的蛋 白的共刺 激信号分子: ICOS、 0X40、 CD2、 CD7、 CD27、 CD28、 CD30、 CD40、 CD70、 CD134、 4-lBB (CD137)、 PD1、 Dapl O、 CDS、 ICAMT、 LFA-1 (CD1 la/CD18)、 ICOS (CD278)、 NKG2D、 GITR、 TLR2、 或其组合。 7. The CAR construct according to claim 4, wherein said C is a co-stimulatory signal molecule of a protein selected from the group consisting of ICOS, OX40, CD2, CD7, CD27, CD28, CD30, CD40, CD70, CD134, 4-1BB (CD137), PD1, Dapl 0, CDS, ICAMT, LFA-1 (CD11a/CD18), ICOS (CD278), NKG2D, GITR, TLR2, or a combination thereof.
8. 如权利要求 4所述的 CAR构建物, 其特征在于, 所述 CAR的氨基酸序列如 SEQ ID NO : 17所示。 8. The CAR construct according to claim 4, wherein the amino acid sequence of the CAR is as shown in SEQ ID NO: 17.
9. 一种分离的核酸分子, 其特征在于, 所述核酸分子编码权利要求 1所述 的嵌合抗 原受体 (CAR)构建物。 9. An isolated nucleic acid molecule, characterized in that, the nucleic acid molecule encodes the chimeric antigen receptor (CAR) construct of claim 1.
10. 一种载体,其特征在于,所述的载体含有权利要求 9所述的核酸分子。10. a carrier, is characterized in that, described carrier contains nucleic acid molecule described in claim 9.
11. 一种宿主细胞, 其特征在于, 所述宿主细胞中含有如权利要求 10所述 的载体 , 或染色体中整合有外源的如权 利要求 9所述的核酸分子, 或表达如权 利要求 1所述的 CAR构建物。 11. A host cell, characterized in that, the host cell contains the vector according to claim 10, or the exogenous nucleic acid molecule according to claim 9 is integrated into the chromosome, or expresses the vector according to claim 1 The CAR construct.
12. 一种工程化的免疫细胞, 其特征在于, 所述免疫细胞表达有如权利要 求 1所述的 CAR构建物。 12. An engineered immune cell, characterized in that, the immune cell expresses the CAR construct according to claim 1.
13. 一种制剂,其特征在于,所述制剂包含如权利要求 1所述的 CAR构建物、 如权利 要求 9所述的分离的核酸分子、 如权利要求 10所述的载体或如权利 要求 12所述的免疫细 胞, 以及药学上可接受的载体。 13. A preparation, characterized in that, the preparation comprises the CAR construct as claimed in claim 1, the isolated nucleic acid molecule as claimed in claim 9, the carrier as claimed in claim 10 or the vector as claimed in claim 12 The immune cells, and a pharmaceutically acceptable carrier.
14. 如权利要求 1所述的 CAR构建物、 如权利要求 9所述的核酸分子、 如权 利要求 10所述的载体、 如权利要求 12所述的免疫细胞、 或如权利要求 9所述的 制剂的用 途, 用于制备预防和/或治疗肿瘤或癌 症的药物。 14. The CAR construct as claimed in claim 1, the nucleic acid molecule as claimed in claim 9, the carrier as claimed in claim 10, the immune cell as claimed in claim 12, or the The use of the preparation is used for preparing a medicine for preventing and/or treating tumor or cancer.
15. 一种治疗肿瘤或癌症的方法, 包括向需要的对象施用有效 量的如权利 要求 12所述的免疫细胞, 或如权利要求 13所述的制剂。 15. A method for treating tumor or cancer, comprising administering an effective amount of the immune cell according to claim 12, or the preparation according to claim 13 to a subject in need.
PCT/IB2023/051678 2022-02-23 2023-02-23 Gpc3-targeting chimeric antigen receptor and use thereof WO2023161845A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210167614.3 2022-02-23
CN202210167614 2022-02-23

Publications (1)

Publication Number Publication Date
WO2023161845A1 true WO2023161845A1 (en) 2023-08-31

Family

ID=87764944

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2023/051678 WO2023161845A1 (en) 2022-02-23 2023-02-23 Gpc3-targeting chimeric antigen receptor and use thereof

Country Status (1)

Country Link
WO (1) WO2023161845A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104140974A (en) * 2013-05-08 2014-11-12 上海益杰生物技术有限公司 Nucleic acid for coding GPC-3 (glypican-3) chimeric antigen receptor protein and T lymphocytes for expression of GPC-3 chimeric antigen receptor protein
CN105949324A (en) * 2016-06-30 2016-09-21 上海恒润达生生物科技有限公司 Chimeric antigen receptor of targeted GPC3 (Glypican 3) and application thereof
CN107384949A (en) * 2017-06-27 2017-11-24 闾民 The constant natural killer T cells of expression targeting GPC3 Chimeric antigen receptors(iNKT)And its prepare and apply
CN111349178A (en) * 2020-05-25 2020-06-30 北京广未生物科技有限公司 GPC 3-targeted Chimeric Antigen Receptor (CAR) and anticancer application thereof
CN112079932A (en) * 2020-09-22 2020-12-15 浙江康佰裕生物科技有限公司 Chimeric antigen receptor for treating liver cancer and application thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104140974A (en) * 2013-05-08 2014-11-12 上海益杰生物技术有限公司 Nucleic acid for coding GPC-3 (glypican-3) chimeric antigen receptor protein and T lymphocytes for expression of GPC-3 chimeric antigen receptor protein
CN105949324A (en) * 2016-06-30 2016-09-21 上海恒润达生生物科技有限公司 Chimeric antigen receptor of targeted GPC3 (Glypican 3) and application thereof
CN107384949A (en) * 2017-06-27 2017-11-24 闾民 The constant natural killer T cells of expression targeting GPC3 Chimeric antigen receptors(iNKT)And its prepare and apply
CN111349178A (en) * 2020-05-25 2020-06-30 北京广未生物科技有限公司 GPC 3-targeted Chimeric Antigen Receptor (CAR) and anticancer application thereof
CN112079932A (en) * 2020-09-22 2020-12-15 浙江康佰裕生物科技有限公司 Chimeric antigen receptor for treating liver cancer and application thereof

Similar Documents

Publication Publication Date Title
CN110760007B (en) CD7-CAR-T cell and preparation and application thereof
JP7148611B2 (en) CHIMERIC ANTIGEN RECEPTOR TARGETING BCMA AND METHOD FOR PRODUCING AND USE THEREOF
CN111051502B (en) Preparation technology of universal chimeric antigen receptor T cell
CN109575143B (en) Bispecific CD20-CD19-CAR and application thereof
CN113087806B (en) Novel CAR-T cells targeting multiple tumors, and preparation and methods thereof
CN112778427B (en) Bispecific CS1-BCMA CAR-T cells and uses thereof
WO2023046110A1 (en) Engineered immune cell co-expressing ccr2b, preparation therefor and application thereof
JP2023027314A (en) Combined chimeric antigen receptors targeting cd19 and cd20 and applications thereof
WO2021136040A1 (en) Preparation and applications of chimeric antigen receptor t-cell co-expressing immunomodulatory molecule
WO2023083192A1 (en) Engineered immune cell for combined expression of ccr2b and cd40l, and preparation and application thereof
WO2023161846A1 (en) Gpc3-targeting chimeric antigen receptor t cell and use thereof
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
WO2022151959A1 (en) Car-t cell targeting b7-h3 and application thereof in treatment of acute myeloid leukemia
WO2021208750A1 (en) Cd22-targeted chimeric antigen receptor, preparation method therefor and application thereof
WO2023161845A1 (en) Gpc3-targeting chimeric antigen receptor and use thereof
CN114907486A (en) EGFR targeting chimeric antigen receptor and preparation method and application thereof
CN116496397B (en) Humanized antibodies targeting CD19CAR-T cells
CN116444669B (en) Humanized antibodies targeting BCMA CAR-T cells
CN116462770B (en) Humanized antibody of CD19, CAR-T cell expressing bispecific chimeric antigen receptor and application thereof
WO2022262765A1 (en) Preparation and application of chimeric antigen receptor immune cell constructed on basis of granzyme b
CN110218702B (en) Immune cell combination targeting CD138 and CD19 and application thereof
WO2023093888A1 (en) Preparation and use of immune cells of chimeric antigen receptor constructed on the basis of efna1
WO2022105893A1 (en) Preparation method and application of cd7-car-t cells
CN109593137B (en) Construction and application of novel CD20-CAR vector of anti-CD 20 antibody

Legal Events

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

Ref document number: 23759417

Country of ref document: EP

Kind code of ref document: A1