WO2023083192A1 - Engineered immune cell for combined expression of ccr2b and cd40l, and preparation and application thereof - Google Patents

Engineered immune cell for combined expression of ccr2b and cd40l, and preparation and application thereof Download PDF

Info

Publication number
WO2023083192A1
WO2023083192A1 PCT/CN2022/130696 CN2022130696W WO2023083192A1 WO 2023083192 A1 WO2023083192 A1 WO 2023083192A1 CN 2022130696 W CN2022130696 W CN 2022130696W WO 2023083192 A1 WO2023083192 A1 WO 2023083192A1
Authority
WO
WIPO (PCT)
Prior art keywords
car
cells
nkg2d
tumor
immune cell
Prior art date
Application number
PCT/CN2022/130696
Other languages
French (fr)
Chinese (zh)
Inventor
张曦
黄智宏
韩德平
燕妮
王调霞
朱晓娜
王宁
林伟雄
卢永
Original Assignee
广州百吉生物制药有限公司
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 广州百吉生物制药有限公司 filed Critical 广州百吉生物制药有限公司
Publication of WO2023083192A1 publication Critical patent/WO2023083192A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • 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
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7158Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for chemokines
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0646Natural killers cells [NK], NKT cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • C12N2510/02Cells for production

Definitions

  • the invention belongs to the field of tumor immunity and cell therapy, and in particular relates to an engineered immune cell jointly expressing CCR2b and CD40L.
  • Cellular immunotherapy is an emerging tumor treatment mode with significant curative effect, and it is a new type of autoimmune anti-cancer treatment. It is a method of using biotechnology and biological agents to culture, modify and amplify immune cells collected from patients in vitro, and then infuse them back into the patient's body to stimulate and enhance the body's autoimmune function, so as to achieve the purpose of treating tumors.
  • T cells are an important class of lymphocytes involved in cellular immunity, and can specifically recognize and kill tumor cells through the signal transmission of antigen-presenting cells.
  • tumor cells can also be affected by the reduction or loss of antigenic epitopes, immunosuppression, and tumor heterogeneity (that is, the same malignant tumor exists in different patients or between tumor cells in different parts of the same patient from genotype to phenotype). Differences) and other ways to hinder the specific recognition of T cells, thereby evading the body's immune response.
  • Chimeric antigen receptor T cell (chimeric antigen receptor T cell, CAR-T) therapy was developed to address this problem.
  • the CAR molecule is an artificially designed and constructed receptor molecule, which consists of a signal peptide, an extracellular antigen-binding domain, a hinge region, a transmembrane region, a co-stimulatory domain, and an intracellular signaling domain. Therefore, CAR molecules have the functions of specifically recognizing tumor surface antigens, activating T cell killing activity, and stimulating T cell proliferation.
  • the patient's own T cells can express CAR molecules. After reinfusion into the patient's body, T cells can efficiently and specifically recognize and kill tumor cells through CAR molecules, achieving the effect of cancer treatment.
  • the first-generation CAR-T only has a single-chain antibody as the extracellular antigen-binding domain and CD3 ⁇ as the intracellular signaling domain, which cannot fully activate the activity of T cells, and the therapeutic effect is not good.
  • the second-generation CAR-T introduced a co-stimulatory domain on the basis of the first-generation CAR-T, which improved the in vitro proliferation ability and cytokine release level of T cells.
  • the third-generation CAR-T adds a co-stimulatory domain to the second-generation CAR-T. Although it can improve the killing activity of T cells, it may induce excessive release of cytokines.
  • the new-generation CAR-T jointly expresses other auxiliary factors, such as the joint expression of IL-12 or the STAT3/5 binding domain in the IL-2R ⁇ cell, which helps to improve tumor killing Effects such as activity and safety.
  • CAR-T therapy has achieved satisfactory results in hematological tumors, there is still a lot of room for improvement in the therapeutic effect of CAR-T on solid tumors.
  • the reasons are as follows: (1) Many solid tumors are difficult to be detected in the early stage, and have the characteristics of high malignancy, high recurrence rate, and poor prognosis. For example, 83% of colorectal cancer patients were in the middle and late stage when they were first diagnosed, and 44% of the patients had metastases to the liver, lung and other parts, and nearly half of the patients survived for less than 5 years; about 70% of ovarian cancer When patients are diagnosed, the cancer cells have already metastasized, and it is difficult to be cured by surgery, chemotherapy, and radiotherapy.
  • the recurrence rate after treatment is still as high as 70%; 90% of pancreatic cancer patients are diagnosed at an advanced stage, and the 5-year survival rate is only 7%.
  • tumor tissues often have an immunosuppressive microenvironment, which can hinder the migration and infiltration of CAR-T cells.
  • Many malignant solid tumors are also characterized by high heterogeneity, and a single target antigen often cannot achieve the best therapeutic effect, and there is a risk of recurrence. Therefore, CAR-T cell therapy for patients with malignant solid tumors such as colorectal cancer, ovarian cancer, and pancreatic cancer needs to further improve its efficiency and effectiveness.
  • the purpose of the present invention is to provide an engineered immune cell (such as CAR-T cell) with higher efficiency and better therapeutic effect for malignant tumors (especially solid tumors).
  • an engineered immune cell such as CAR-T cell
  • Another object of the present invention is to provide an engineered immune cell (such as CAR-T cell) jointly expressing CCR2b and CD40L and its preparation method and application.
  • an engineered immune cell such as CAR-T cell
  • the first aspect of the present invention provides an engineered immune cell, the engineered immune cell is a T cell or NK cell, and the immune cell has the following characteristics:
  • the immune cell expresses a chimeric antigen receptor (chimeric antigen receptor, CAR), wherein the CAR targets surface markers of tumor cells, wherein the antigen-binding domain of the CAR includes the extracellular domain of NKG2D ;
  • CAR chimeric antigen receptor
  • said immune cell expresses an exogenous CCR2b protein
  • the immune cells express exogenous CD40L protein.
  • the T cells include ⁇ T, ⁇ T cells, NKT cells, MAIT cells, or combinations thereof.
  • the engineered immune cells are selected from the following group:
  • CAR-T cells chimeric antigen receptor T cells
  • CAR-NK cells Chimeric antigen receptor NK cells
  • the CCR2b protein and/or CD40L can be expressed constitutively or inducibly.
  • a chimeric antigen receptor T cell (CAR-T cell) is provided, and the CAR-T cell has one or more of the following characteristics:
  • the cell expresses a chimeric antigen receptor CAR that targets a surface marker of the tumor cell
  • CAR, CCR2b and CD40L proteins are expressed in series.
  • CAR, CCR2b and CD40L proteins are independently expressed.
  • the "activation" refers to the binding of the CAR to surface markers of tumor cells.
  • the "tumor surface marker” refers to a specific antigen on the surface of the tumor.
  • the chimeric antigen receptor CAR is located on the cell membrane of the engineered immune cells.
  • the chimeric antigen receptor CAR is located on the cell membrane of the CAR-T cell.
  • the CCR2b protein is located on the cell membrane of the CAR-T cells.
  • L is nothing or a signal peptide sequence
  • NKG2D is an NKG2D extracellular domain or an active fragment thereof
  • H is none or hinge region
  • TM is the transmembrane domain
  • C is costimulatory signal domain
  • CD3 ⁇ is a cytoplasmic signaling sequence derived from CD3 ⁇ (including wild type, or mutants/modifiers thereof);
  • the "-" is a connecting peptide or a peptide bond.
  • said L is respectively selected from signal peptides of proteins in the following group: CD8, GM-CSF, CD4, CD28, CD137, or mutants/modifications thereof, or combinations thereof.
  • the H is selected from the hinge region of proteins in the following group: CD8, CD28, CD137, IgG, or a combination thereof.
  • the TM is selected from the transmembrane region of the following group of proteins: CD28, CD3epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134 , CD137, CD154, CD278, CD152, CD279, CD233, CD314, or a mutation/modification thereof, or a combination thereof.
  • the C is selected from the co-stimulatory domains of proteins in the following group: OX40, CD2, CD7, CD27, CD28, CD30, CD40, CD70, CD134, 4-1BB (CD137), PD1, Dap10 , CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), NKG2D, GITR, OX40L, or mutants/modifications thereof, or combinations thereof.
  • C is a co-stimulatory domain derived from 4-1BB.
  • amino acid sequence of the extracellular domain of NKG2D is as shown in SEQ ID NO:1 73-216 or as shown in SEQ ID No:4.
  • the CAR cell in addition to the first CAR shown in formula I, also contains a second CAR for targeting the second antigen, and the structure of the second CAR is shown in formula II:
  • L is nothing or a signal peptide sequence
  • scFv is an antigen-binding domain
  • H is none or hinge region
  • TM is the transmembrane domain
  • C is costimulatory domain
  • CD3 ⁇ is a cytoplasmic signaling sequence derived from CD3 ⁇ or a mutation/modification thereof;
  • the "-" is a connecting peptide or a peptide bond.
  • the scFv is an antibody single-chain variable region sequence targeting a tumor antigen.
  • the scFv is an antibody single-chain variable region sequence targeting an antigen selected from the following group: CD19, CD20, CD22, CD123, CD47, CD138, CD33, CD30, CD271, GUCY2C, CD24, CD133 , CD44, CD166, CD133, CD276, ABCB5, ALDH1, mesothelin (MSLN), EGFR, GPC3, BCMA, ErbB2, NKG2D ligands (ligands), LMP1, EpCAM, VEGFR-1, Lewis-Y, ROR1 , Claudin18.2, CEA, TAG-72, TROP2, or combinations thereof.
  • an antigen selected from the following group: CD19, CD20, CD22, CD123, CD47, CD138, CD33, CD30, CD271, GUCY2C, CD24, CD133 , CD44, CD166, CD133, CD276, ABCB5, ALDH1, mesothelin (MSLN), EGFR, GPC3, BCMA, Erb
  • amino acid sequence of the NKG2D is shown in SEQ ID NO: 1, wherein the extracellular domain is 73-216.
  • the CCR2b protein includes a full-length CCR2b protein or an active fragment thereof (ie, an active fragment retaining the function of binding to a corresponding ligand).
  • the amino acid sequence of the CCR2b protein is shown in SEQ ID NO:2.
  • the CD40L protein includes full-length CD40L protein or its active fragment (ie, the active fragment retains the function of binding to CD40).
  • amino acid sequence of the CD40L protein is shown in SEQ ID NO:5.
  • first CAR shown in formula I and the second CAR shown in formula II can be combined into one to form a CAR shown in formula IIIa or IIIb:
  • L is nothing or a signal peptide sequence
  • NKG2D is an NKG2D extracellular domain or an active fragment thereof
  • scFv is an antigen-binding domain
  • H is none or hinge region
  • TM is the transmembrane domain
  • C is costimulatory domain
  • CD3 ⁇ is a cytoplasmic signaling sequence derived from CD3 ⁇ or a mutation/modification thereof;
  • the "-" is a connecting peptide or a peptide bond.
  • the second aspect of the present invention provides a method for preparing the engineered immune cells described in the first aspect of the present invention, comprising the following steps:
  • step (B) including:
  • the step (B1) can be performed before, after, simultaneously or alternately with the step (B2).
  • the step (B1) can be performed before, after, simultaneously, or alternately before the step (B3).
  • the step (B2) can be performed before, after, simultaneously or alternately with the step (B3).
  • steps (B1), (B2) and (B3) are performed simultaneously or alternately.
  • a method for preparing the CAR-T cells of the present invention comprising the following steps:
  • step (B) in step (B), it includes: introducing the first expression cassette expressing the CAR into the T cell; and introducing the second expression cassette expressing CCR2b and the third expression cassette expressing CD40L The T cells; wherein the introducing steps can be performed in any order.
  • any two expression cassettes in the first, second and third expression cassettes taking the first expression cassette and the second expression cassette as an example, their transcription directions are in the same direction ( ⁇ ), opposite ( ⁇ ) or opposite ( ⁇ ).
  • first expression cassette, the second expression cassette and the third expression cassette are located on the same or different vectors.
  • the first expression cassette, the second expression cassette and the third expression cassette are located in the same vector.
  • a connecting peptide is also included between the two proteins.
  • the connecting peptide is P2A or T2A.
  • the vector is a viral vector, preferably the viral vector contains the first and second expression cassettes in tandem form.
  • the vector is selected from the group consisting of DNA, RNA, plasmid, lentiviral vector, adenoviral vector, retroviral vector, transposon, other gene transfer systems, or combinations thereof.
  • the vector is a pCDH series lentiviral vector.
  • the third aspect of the present invention provides a preparation, which contains the engineered immune cells described in the first aspect of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient.
  • the preparation contains the CAR-T cells of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient.
  • the formulation is a liquid formulation.
  • the dosage form of the preparation includes injection.
  • the concentration of the engineered immune cells (such as CAR-T cells) in the preparation is 1 ⁇ 10 3 -1 ⁇ 10 8 cells/ml, preferably 1 ⁇ 10 4 -1 ⁇ 10 7 cells/ml.
  • the fourth aspect of the present invention provides the use of the engineered immune cell according to the first aspect of the present invention for preparing a drug or preparation for preventing and/or treating cancer.
  • the use of the CAR-T cells according to the first aspect of the present invention is provided for the preparation of drugs or preparations for preventing and/or treating cancer or tumors.
  • the preparation contains CAR-T cells, and a pharmaceutically acceptable carrier, diluent or excipient.
  • the tumor includes a solid tumor.
  • the tumor is selected from the group consisting of colon cancer, rectal cancer, ovarian cancer, or pancreatic cancer.
  • the tumor is a tumor with high expression of NKG2D ligand and/or high expression of chemokines and/or high expression of CD40.
  • the tumor is a tumor with high expression of NKG2D ligands and high expression of chemokines.
  • the tumor is a tumor with high expression of NKG2D ligands and high expression of chemokines.
  • the tumor is a tumor with high expression of NKG2D ligand and high expression of CD40.
  • the chemokine is selected from the group consisting of CCL2, CCL7, or a combination thereof.
  • the tumor is a tumor with high expression of NKG2D ligand, high expression of chemokines CCL2 and/or CCL7, and high expression of CD40.
  • the tumor is a tumor with high expression of NKG2D ligands (including any one of MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5, ULBP6, or a combination thereof).
  • NKG2D ligands including any one of MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5, ULBP6, or a combination thereof.
  • the tumor has high expression of NKG2D ligands (including any one of MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5, ULBP6, or a combination thereof) and/or chemokines (Including tumors with high expression of CCL2, CCL7, CCL8, CCL12, CCL13, CCL16, or their combination) and/or high expression of CD40.
  • NKG2D ligands including any one of MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5, ULBP6, or a combination thereof
  • chemokines Including tumors with high expression of CCL2, CCL7, CCL8, CCL12, CCL13, CCL16, or their combination
  • CD40 high expression of CD40.
  • the fifth aspect of the present invention provides a kit for preparing the engineered immune cell described in the first aspect of the present invention, the kit includes a container, and in the container:
  • kits for preparing the engineered immune cell according to the first aspect of the present invention includes a container, and in the container:
  • first, second and third nucleic acid sequences are independent or linked.
  • first, second and third nucleic acid sequences are located in the same or different containers.
  • first, second and third nucleic acid sequences are located on the same or different vectors.
  • first, second and third nucleic acid sequences are located in the same vector.
  • a connecting peptide expression for expressing the connecting peptide is also included between them. box.
  • the connecting peptide is P2A or T2A.
  • the vector is a viral vector, preferably the viral vector contains the first, second and third nucleic acid sequences in tandem form.
  • the sixth aspect of the present invention provides a method for treating tumors, comprising the step of: administering a safe and effective amount of the preparation described in the third aspect of the present invention to a subject in need, thereby treating the subject's tumor.
  • FIG 1 shows the structures of CAR molecules of different generations.
  • Figure 2 shows the structure of the CAR molecule.
  • Figure 3 shows the expression rate of NKG2D CAR molecules of each CAR-T cell detected by flow cytometry.
  • Figure 4 shows the expression rate of CCR2b of each CAR-T cell detected by flow cytometry.
  • Figure 5 shows the expression rate of CD40L of each CAR-T cell detected by flow cytometry.
  • Figure 6 shows the expression rate of NKG2D ligands (MICA/MICB) in target cells detected by flow cytometry.
  • Figure 7 shows the expression rate of NKG2D ligand (ULBP-1) in target cells detected by flow cytometry.
  • Figure 8 shows the expression rate of NKG2D ligands (ULBP-2/5/6) in target cells detected by flow cytometry.
  • Figure 9 shows the expression rate of NKG2D ligand (ULBP-3) in target cells detected by flow cytometry.
  • Figure 10 shows the expression rate of NKG2D ligand (ULBP-4) in target cells detected by flow cytometry.
  • Figure 11 shows the expression rate of CD40 in target cells detected by flow cytometry.
  • Figure 12 shows the results of Incucyte's real-time detection of the chemotaxis and migration ability of BN009.
  • Figure 13 shows the killing effect of each NKG2D CAR-T cell on tumor cells detected by EuTDA.
  • Figure 14 shows the IFN- ⁇ release level of each NKG2D CAR-T cell detected by ELISA.
  • the inventors co-expressed specific CAR, CCR2b and CD40L proteins for the first time in engineered immune cells, that is, combining CAR containing NKG2D extracellular domain (ED) with CCR2b and CD40L Expressed in CAR-T and other immune cells.
  • the CAR-T cells of the present invention simultaneously have the multi-target recognition ability of NKG2D, the enhanced chemotaxis and migration ability of CCR2b, and the immune activation ability of CD40L, and unexpectedly exhibit a synergistic killing effect on tumor cells in vitro.
  • the present invention has been accomplished on this basis.
  • the immune cells of the present invention can improve the sensitivity of CAR-T cells to chemokines such as CCL2 and CCL7 through CCR2b, and promote tumor lesions of solid tumors such as colorectal cancer, ovarian cancer, and pancreatic cancer.
  • NKG2D CAR molecules can recognize various target antigens on the surface of malignant tumor cells such as colorectal cancer, ovarian cancer, and pancreatic cancer (including MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5 , ULBP6), reducing the risk of decreased efficacy due to tumor heterogeneity or loss of target antigens.
  • co-expressed CD40L can effectively activate the body's endogenous natural and adaptive immune responses, thereby helping T cells overcome the immunosuppressive tumor microenvironment, improve tumor therapy, and reduce the risk of tumor recurrence.
  • the present invention takes CAR-T cells as an example to representatively describe the engineered immune cells of the present invention in detail.
  • the engineered immune cells of the present invention are not limited to the CAR-T cells described above, and the engineered immune cells of the present invention have the same or similar technical features and beneficial effects as the CAR-T cells described above.
  • immune cells express chimeric antigen receptor CAR
  • NK cells are equivalent to T cells (or T cells can be replaced by NK cells).
  • administration refers to the physical introduction of a product of the invention into a subject using any of a variety of methods and delivery systems known to those skilled in the art, including intravenous, intratumoral, intramuscular, subcutaneous, intraperitoneal , spinal or other parenteral routes of administration, for example by injection or infusion.
  • the term "antibody” shall include, but not be limited to, an immunoglobulin that specifically binds an antigen and comprises at least two heavy (H) chains and two light (L) chains interconnected by disulfide bonds , or an antigen-binding portion thereof.
  • Each H chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
  • the heavy chain constant region comprises three constant domains CH1, CH2 and CH3.
  • Each light chain comprises a light chain variable region (abbreviated herein as VL) and a light chain constant region.
  • the light chain constant region comprises one constant domain, CL.
  • VH and VL regions can be further subdivided into hypervariable regions called complementarity determining regions (CDRs), interspersed with more conserved regions called framework regions (FRs).
  • CDRs complementarity determining regions
  • FRs framework regions
  • Each VH and VL contains three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain binding domains that interact with the antigen.
  • antigen-binding domain and “single-chain antibody fragment” all refer to a Fab fragment, Fab' fragment, F(ab') 2 fragment, or a single Fv fragment having 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 the structures required for antigen binding.
  • the antigen binding domain is usually a scFv (single-chain variable fragment).
  • a single-chain antibody is preferably a sequence of one amino acid chain encoded by one nucleotide chain.
  • the scFv comprises an NKG2D extracellular domain or an active fragment thereof that specifically recognizes an antigen highly expressed by a tumor.
  • the immune cells of the present invention may also contain additional antibodies that specifically recognize antigens highly expressed in tumors, preferably single-chain antibodies or Fv antibodies.
  • a Chimeric antigen receptor includes an extracellular domain, an optional hinge region, a transmembrane domain, and an intracellular domain.
  • the extracellular domain includes an optional signal peptide and a target-specific binding domain (also known as an antigen binding domain).
  • the intracellular domain includes the co-stimulatory domain and the CD3 ⁇ chain portion.
  • the extracellular segment can recognize a specific antigen, and then transduce the signal through the intracellular domain, causing cell activation and proliferation, cytolytic toxicity and secretion of cytokines such as IL-2 and IFN- ⁇ etc., affecting tumor cells so that they do not grow, are induced to die, or are otherwise affected, and result 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 the co-stimulatory molecule and the CD3zeta chain.
  • the antigen binding domain is fused to the intracellular domain of the combination of the 4-1BB signaling domain and the CD3 ⁇ signaling domain.
  • the CAR of the present invention targets NKG2D ligands and can specifically bind to MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5, and ULBP6.
  • CAR-T cell As used herein, the terms “CAR-T cell”, “CAR-T” and “CAR-T cell of the present invention” all refer to the CAR-T cell described in the first aspect of the present invention.
  • the CAR-T cells of the present invention can be used to treat tumors with high expression of NKG2D ligands, such as colorectal cancer, ovarian cancer, pancreatic cancer and the like.
  • CAR-T cells have the following advantages over other T-cell-based therapies: (1) The action process of CAR-T cells is not restricted by MHC; (2) Since many tumor cells express the same tumor antigen, it can target a certain tumor Once the CAR gene construction of the antigen is completed, it can be widely used; (3) CAR can use both tumor protein antigens and glycolipid non-protein antigens, expanding the target range of tumor antigens; (4) using the patient's own The cells reduce the risk of rejection; (5) CAR-T cells have immune memory function and can survive in the body for a long time.
  • CAR-NK cells As used herein, the terms “CAR-NK cells”, “CAR-NK” and “CAR-NK cells of the present invention” all refer to the CAR-NK cells described in the first aspect of the present invention.
  • the CAR-NK cells of the present invention can be used to treat tumors with high expression of NKG2D ligands, such as colorectal cancer, ovarian cancer, pancreatic cancer and the like.
  • NK cells are a major type of immune effector cells, which protect the body from virus infection and tumor cell invasion through non-antigen-specific pathways. NK cells through engineering (gene modification) may obtain new functions, including the ability to specifically recognize tumor antigens and have enhanced anti-tumor cytotoxicity.
  • CAR-NK cells Compared with autologous CAR-T cells, CAR-NK cells also have the following advantages, for example: (1) directly kill tumor cells by releasing perforin and granzymes, but have no killing effect on normal cells of the body; (2) they release A very small amount of cytokines reduces the risk of cytokine storm; (3) It is very easy to expand in vitro and develop into "off-the-shelf" products. Other than that, it is similar to CAR-T cell therapy.
  • NKG2D includes wild type or its mutant or its derivative form or its active fragment.
  • Preferred NKG2Ds are NKG2Ds from mammals such as humans and non-human primates.
  • accession number of the amino acid sequence of the human NKG2D protein is NP_031386, and the accession number of the nucleotide amino acid sequence is NM_007360.
  • the full-length amino acid sequence of human NKG2D is as follows:
  • positions 1-51 are intracellular domains; positions 52-72 are transmembrane regions; positions 73-216 are NKG2D extracellular domains (underlined).
  • Chemokines are a special class of cytokines, including more than 50 members. According to the structure, it is divided into four types: CC, CXC, CX3C and XC; chemokine receptors are correspondingly divided into four types: CCR, CXCR, CX3CR and XCR, with about 20 members.
  • the expressed chemokine receptor is CCR2b protein
  • the chemokines that can be bound include CCL2, CCL7, CCL8, CCL12, CCL13, CCL16 and the like.
  • accession number of the amino acid sequence of the CCR2b protein is NP_001116868.1, and the accession number of the nucleotide amino acid sequence is NM_001123396.4.
  • the specific sequence is as follows:
  • Leukocyte differentiation antigen 40 ligand cluster of differentiation 40 ligand, CD40L
  • CD154 tumor necrosis factor-associated activation protein
  • TRAP tumor necrosis factor-associated activation protein
  • accession number of the amino acid sequence of human CD40L protein is NP_000065.1, and the accession number of the nucleotide amino acid sequence is NM_000074.3.
  • the full-length amino acid sequence of human CD40L is as follows:
  • suitable CD40L includes wild-type and mutant-type CD40L, as long as the mutant-type CD40L has basic functions of wild-type CD40L.
  • preferred CD40L is from mammals, such as humans and non-human mammals.
  • CD40L is mainly expressed in activated CD4+T lymphocytes, activated CD8 + T cells, basophils, mast cells and NK cells.
  • CD40L and its receptor CD40 are a pair of co-stimulatory molecules in the inflammatory and immune response systems in vivo.
  • the CD40L/CD40 co-stimulatory pathway is an important trigger for the maturation process of monocytes, which mainly drives the differentiation of monocytes into M1 lineage macrophages and DC cells.
  • this pathway can also promote the release of cytokines and chemokines from DC cells, induce the expression of other co-stimulatory molecules, and promote the cross-presentation of antigens.
  • this pathway In humoral immunity, this pathway is also involved in the T cell-dependent B lymphocyte response process, the formation of germinal centers, the generation of long-term memory B cells, antibody production, and antibody class switching. In cellular immunity, this pathway can promote T cell activation and amplify T cell-mediated immune response, play an important role in the process of CD4 + T cell differentiation, and can also promote the expansion and pluripotency of CD8 + T cells, It is the basis for generating memory CD8+ T cells.
  • the CD40L/CD40 co-stimulatory pathway also plays multiple roles, such as activating the proliferation of T cells and the release of cytokines, and inducing the M2 lineage macrophages to M1 lineage macrophages with anti-tumor activity. shift etc. In tumors with loss of some target antigens but high expression of CD40, this pathway can also mediate the killing effect of T cells on tumor cells.
  • CD40L when CD40L is co-expressed with a specific CAR molecule and CCR2b, CD40L, as an auxiliary factor of CAR-T cells, can activate the endogenous immune response of the body while CAR-T cells kill tumor cells, enhance and Prolongs the healing effect.
  • CD40L can also play a synergistic effect with CCR2b, thereby synergistically significantly improving the killing effect against tumor cells in vitro.
  • an "expression cassette” or “expression cassette of the invention” includes a first expression cassette, a second expression cassette and a third expression cassette.
  • the expression cassette of the present invention is as described in the fifth aspect of the present invention, the first expression cassette comprises the nucleic acid sequence encoding the CAR.
  • the second expression cassette expresses exogenous CCR2b protein.
  • the second expression cassette expresses exogenous CD40L protein.
  • CCR2b and CD40L proteins can be expressed constitutively or inducibly.
  • the second expression cassette expresses the CCR2b protein
  • the third expression cassette expresses the CD40L protein; thus, when the CAR-T cells of the present invention are not exposed to the corresponding induced
  • the second expression cassette does not express the CCR2b protein
  • the third expression cassette does not express the CD40L protein.
  • the first expression cassette, the second expression cassette and/or the third expression cassette further comprise a promoter and/or a terminator, respectively.
  • the promoters of the second and third expression cassettes may be constitutive or inducible promoters.
  • the present invention also provides a vector comprising the expression cassette of the present invention.
  • 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 in the genome of the cell and replication of the genome of the daughter cell.
  • Lentiviral vectors have advantages over vectors derived from oncogenic retroviruses such as murine leukemia virus in that they can transduce non-proliferating cells and have the advantage of low immunogenicity.
  • the expression cassette or nucleic acid sequence of the present invention can be connected downstream of the promoter through routine operations, and incorporated into an expression vector.
  • the vector can be integrated into the genome of eukaryotic cells and then replicated.
  • 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 vectors of the invention can also be used in standard gene delivery protocols for nucleic acid immunization and gene therapy. 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.
  • the expression cassette or nucleic acid sequence can be cloned into many types of vectors.
  • the expression cassette or nucleic acid sequence can be cloned into such vectors including, but not limited to, plasmids, phagemids, phage derivatives, animal viruses, and cosmids.
  • Particular vectors of interest include expression vectors, replication vectors, and the like.
  • expression vectors can be provided to cells in the form of viral vectors.
  • Viral vector technology is well known in the art and described, for example, in Molecular Cloning: A Laboratory Manual (Sambrook et al., Cold Spring Harbor Laboratory, New York, 2001) and other virology and molecular biology manuals.
  • Viruses that can be used as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpesviruses, and lentiviruses.
  • suitable vectors contain at least one origin of replication functional in the organism, a promoter sequence, convenient restriction enzyme sites, and one or more selectable markers (e.g., 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.
  • lentiviral vectors are used.
  • Many DNA viral systems are known in the art.
  • an adenoviral vector is used.
  • Many adenoviral vectors are known in the art.
  • promoter elements can regulate the frequency of transcription initiation.
  • these elements are located in a region of 30-110 bp 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 an element is inverted or moved relative to another element.
  • tk thymidine kinase
  • the spacing between promoter elements can be increased by 50 bp apart before activity begins to decline.
  • individual elements can act cooperatively or independently to initiate transcription.
  • a suitable promoter is the cytomegalovirus (CMV) promoter sequence.
  • the promoter sequence is a strong constitutive promoter sequence capable of driving high level expression of any polynucleotide sequence operably linked thereto.
  • Another example of a suitable promoter is elongation growth factor-1 alpha (EF-1 alpha).
  • constitutive promoter sequences can also be used, including but not limited to the simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, avian leukemia virus promoter, Epstein-Barr virus (Epstein-Barr virus, EBV) immediate early promoter, Ruth's sarcoma virus promoter, and human gene promoters, such as but not limited to protein 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 the polynucleotide sequence linked to the inducible promoter when desired, or turning off expression when 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 cells may also contain either or both of a selectable marker gene or a reporter gene to facilitate the identification and selection of expressing cells from the transfected or infected cell population by the viral vector.
  • selectable markers can be carried on a single piece of DNA and used in a co-transfection procedure. Both the selectable marker gene and the reporter gene may be flanked by appropriate regulatory sequences to enable expression in the host cell.
  • Useful selectable marker genes include, for example, antibiotic resistance genes such as neomycin and the like.
  • the vector can be easily introduced into host cells, eg, mammalian (eg, human T cells), bacterial, yeast or insect cells, by any method in the art.
  • expression vectors can be transferred into host cells by physical, chemical or biological means.
  • Physical methods for introducing polynucleotides into host cells include calcium phosphate precipitation, cationic complex transfection, lipofection, particle bombardment, microinjection, electroporation, and the like. Methods for producing cells comprising vectors and/or exogenous nucleic acids are well known in the art. See, e.g., Molecular Cloning: A Laboratory Manual (Sambrook et al., Cold Spring Harbor Laboratory, New York, 2001). Preferred methods for introducing polynucleotides into host cells are liposome transfection and cationic complex polyethylenimine transfection.
  • Biological methods for introducing polynucleotides into host cells include the use of DNA and RNA vectors.
  • Viral vectors especially retroviral vectors, have become the most widely used method of 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, US Patent Nos. 5,350,674 and 5,585,362.
  • 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.
  • lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and lipid-based systems.
  • An exemplary colloidal system for use as a delivery vehicle in vitro and in vivo is a liposome (eg, an artificial membrane vesicle).
  • an exemplary delivery vehicle is liposomes.
  • lipid formulations to introduce 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
  • linker molecules associated with both liposomes and oligonucleotides
  • entrapped in liposomes complexed with liposomes, dispersed in lipid-containing solutions, mixed with lipids, associated with lipids, contained in lipids as a suspension, contained in micelles or Complexes 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 lipid 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 hydrocarbons and their derivatives such as fatty acids, alcohols, amines, aminoalcohols, and aldehydes.
  • the vector is a lentiviral vector.
  • the present invention provides an engineered immune cell (such as CAR-T cell) according to the first aspect of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient.
  • the formulation is a liquid formulation.
  • the preparation is an injection.
  • the concentration of the CAR-T cells in the preparation is 1 ⁇ 10 3 -1 ⁇ 10 8 cells/ml, more preferably 1 ⁇ 10 4 -1 ⁇ 10 7 cells/ml.
  • the formulation 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; chelating agents such as EDTA or glutathione; adjuvants (eg, aluminum hydroxide); and preservatives.
  • buffers such as neutral buffered saline, sulfate buffered saline, etc.
  • carbohydrates such as glucose, mannose, sucrose or dextran, mannitol
  • proteins polypeptides or amino acids
  • antioxidants such as glycine
  • chelating agents such as EDTA or glutathione
  • adjuvants eg, aluminum hydroxide
  • preservatives e.g, aluminum hydroxide
  • the invention includes the therapeutic use of cells (eg, T cells) transduced with a vector (eg, a lentiviral vector) comprising an expression cassette of the invention.
  • a vector eg, a lentiviral vector
  • the transduced T cells can target the surface markers of tumor cells and express CCR2b protein, synergistically and significantly improving their killing efficiency against tumor cells.
  • the present invention also provides a method for stimulating an immune response mediated by T cells targeting mammalian tumor cell populations or tissues, comprising the following steps: administering the CAR-T cells of the present invention to mammals.
  • the present invention includes a type of cell therapy, in which a patient's own T cells (or a heterologous 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 heterologous donor
  • the probability of graft-versus-host reaction is extremely low, and the antigen is recognized by T cells without MHC restriction.
  • a single CAR-T can treat all cancers that express that antigen.
  • CAR-T cells are able to replicate in vivo, resulting in long-term persistence that can lead to sustained tumor control.
  • the CAR-T cells of the present invention can undergo stable in vivo expansion and last for several months to several years.
  • the CAR-mediated immune response can be part of an adoptive immunotherapy step in which CAR-T cells can induce a specific immune response to tumor cells that overexpress the antigen recognized by the CAR antigen-binding domain.
  • the CAR-T cells of the present invention elicit a specific immune response against tumor cells with high expression of NKG2D ligands.
  • Treatable cancers include tumors that are not or substantially not vascularized, as well as vascularized tumors.
  • Cancer types treated with the CAR of the present invention include, but are not limited to: colorectal cancer, ovarian cancer, and pancreatic cancer.
  • the present invention provides a method of treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of a CAR-T cell of the present invention.
  • the CAR-T cells of the present 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 compositions of the present invention may comprise a target cell population as described herein, in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients.
  • 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 factors such as the patient's condition, and the type and severity of the patient's disease, or may be determined by clinical trials.
  • compositions of the invention to be administered can be determined by a physician, taking into account the patient (subject ) with individual differences in age, weight, tumor size, degree of infection or metastasis, and disease.
  • Pharmaceutical compositions comprising T cells described herein may be administered at a dose of 10 4 to 10 9 cells/kg body weight, preferably at a dose of 10 5 to 10 7 cells/kg body weight (including all integer values within the range). 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.
  • compositions described herein can be administered to a patient subcutaneously, intradermally, intratumorally, intranodally, intraspinally, intramuscularly, by intravenous injection or intraperitoneally.
  • the 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 intravenous 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 (e.g., previously , simultaneously or subsequently) to the patient in a form of treatment including but not limited to treatment with agents such as antiviral therapy, cidofovir and interleukin-2, cytarabine (also known as ARA-C) or natalizumab treatment for MS patients or erfatizumab treatment for psoriasis patients or other treatments for PML patients.
  • agents such as antiviral therapy, cidofovir and interleukin-2, cytarabine (also known as ARA-C) or natalizumab treatment for MS patients or erfatizumab treatment for psoriasis patients or other treatments for PML patients.
  • the T cells of the invention may be used in combination with chemotherapy, radiation, immunosuppressants such as cyclosporine, azathioprine, methotrexate, 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 ⁇ 10 5 to 1 ⁇ 10 10 modified T cells of the present invention can be administered to the patient for each treatment or each course of treatment, for example, through intravenous infusion.
  • the present invention uses the CCR2b protein to enable the immune cells of the present invention to migrate to the tumor site more efficiently, thereby significantly improving the effect of inhibiting tumors and reducing toxic and side effects.
  • the present invention significantly improves the ability of CAR-T cells to migrate to places with high CCL2 concentration (such as lesion sites).
  • the antigen binding domain of the engineered immune cells of the present invention adopts the extracellular binding domain of NKG2D, which can recognize 8 kinds of targets on the cell surface of malignant tumors (such as colorectal cancer cells, ovarian cancer, pancreatic cancer, etc.) through NKG2D CAR molecules.
  • Spot antigens (MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5, ULBP6), reduce the risk of decreased efficacy due to tumor heterogeneity or loss of target antigens.
  • the present invention uses CD40L as the second cofactor to more effectively activate the body's endogenous natural and adaptive immune responses.
  • CD40L activates the proliferation of T cells and the release of cytokines, induces the transformation of M2 lineage macrophages into M1 lineage macrophages with anti-tumor activity, enhances the antigen presentation function of DC cells, etc., and at the same time improves the ability of T cells to certain Killing effect of tumor cells with antigen loss but high CD40 expression.
  • each NKG2D CAR comprises the following partial structures: human CD8 signal peptide [abbreviated as CD8 (SP)], human NKG2D extracellular domain [abbreviated as NKG2D (ED)], optimized human CD8 hinge region [abbreviated as CD8 ( hinge)], human CD8 transmembrane domain [abbreviated as CD8(TM)], human 4-1BB intracellular domain [abbreviated as 4-1BB(ID)], human CD3 ⁇ intracellular signal transduction domain [abbreviated as CD3 ⁇ (ID) )], self-cleaving peptide P2A, human CCR2b, self-cleaving peptide T2A, human CD40L.
  • CD8 signal peptide
  • SP human NKG2D extracellular domain
  • ED optimized human CD8 hinge region
  • CD8 (TM) human CD8 transmembrane domain
  • 4-1BB intracellular domain abbreviated as 4-1BB(ID)
  • CD3 ⁇ intracellular signal transduction domain abbreviated as CD
  • the second-generation NKG2D CAR molecule used as a control was named BN001
  • the new-generation NKG2D CAR molecule jointly expressing CCR2 was named BN003
  • the new-generation NKG2D CAR molecule jointly expressing CD40L was named BN004
  • the new-generation NKG2D CAR molecule jointly expressing CCR2 and CD40L was named BN004. Named BN009.
  • BN001 is composed of CD8 (SP), NKG2D (ED), CD8 (hinge), CD8 (TM), 4-1BB (ID) and CD3 ⁇ (ID) in series from its amino terminal to carboxyl terminal.
  • BN003 consists of CD8 (SP), NKG2D (ED), CD8 (hinge), CD8 (TM), 4-1BB (ID), CD3 ⁇ (ID), P2A, and CCR2b in series from its amino terminal to carboxyl terminal.
  • BN004 consists of CD8 (SP), NKG2D (ED), CD8 (hinge), CD8 (TM), 4-1BB (ID), CD3 ⁇ (ID), P2A, and CD40L in series from its amino terminal to carboxyl terminal.
  • CD8(SP), NKG2D(ED), CD8(hinge), CD8(TM), 4-1BB(ID), CD3 ⁇ (ID), P2A, CCR2b, T2A, CD40L are sequentially connected in series from the amino terminal to the carboxyl terminal of BN009 composition.
  • SEQ ID NO: 1 human NKG2D amino acid sequence
  • SEQ ID NO: 2 human CCR2b amino acid sequence
  • SEQ ID No: 3 amino acid sequence of human CD8 signal peptide
  • SEQ ID No: 4 amino acid sequence of human NKG2D extracellular domain
  • SEQ ID No: 8 amino acid sequence of human 4-1BB intracellular domain
  • SEQ ID No:9 amino acid sequence of human CD3 ⁇ intracellular signal transduction domain
  • SEQ ID No:10 amino acid sequence of self-cleaving peptide P2A
  • SEQ ID No: 11 amino acid sequence of self-cleaving peptide T2A
  • Example 1 lentivirus preparation
  • nucleotide sequences of BN001, BN003, BN004, and BN009 were synthesized from the whole gene, and then connected to the lentiviral vector pCDH-EF1-MCS-T2A-copGFP plasmid by molecular cloning, so that it was in the human EF1 ⁇ promoter and Kozak sequence expression under control.
  • lentiviral vector plasmids with lentiviral packaging plasmids pRSV-Rev, pMDLg/pRRE and pMD2.G with polyethyleneimine transfection reagent, and co-transfect 293T cells. After culturing for 48 hours, collect the virus supernatant, centrifuge at 4,500 rpm for 10-15 minutes at 4°C, filter through a filter membrane with a pore size of 0.5 ⁇ m, and then use a hollow fiber column ultrafiltration system to concentrate the lentivirus, and then use chromatography to extract the lentivirus. Purify, and finally filter and sterilize with a filter membrane with a pore size of 0.22 ⁇ m, and store in -80°C.
  • the cells in each well were gently mixed and transferred to a 1.5-ml centrifuge tube, washed twice with staining buffer (100 ml PBS+1% BSA), and centrifuged at 800 g for 3 min each time.
  • staining buffer 100 ml PBS+1% BSA
  • the above cells were stained with corresponding antibodies, and then the proportion of Jurkat cells successfully transduced by lentivirus was detected by flow cytometry.
  • the lentivirus infection rate of Jurkat cells is recorded as P (%), the volume of virus liquid is recorded as V (ml), and the dilution factor of virus liquid is recorded as N, and the lentivirus titer is calculated by the following formula:
  • the titer of BN001 was 5.46 ⁇ 10 8 TU/ml
  • the titer of BN003 was 2.46 ⁇ 10 8 TU/ml
  • the titer of BN004 was 6.31 ⁇ 10 8 TU/ml
  • the titer of BN009 was 9.51 ⁇ 10 8 TU/ml.
  • the obtained T cells were washed, and the cell density was adjusted to 2 ⁇ 10 6 /ml.
  • Add lentivirus at MOI 1 ⁇ 10TU/cell for transduction, supplement 50ng/ml anti-CD3 antibody, 50ng/ml anti-CD28 antibody, and 200IU/ml recombinant IL-2 at the same time, and culture in a cell culture incubator ( The culture temperature was 37°C, and the carbon dioxide concentration was 5%). After 24 hours, the cell density was adjusted to 1.5-2 ⁇ 10 6 /ml, and 300 IU/ml of IL-2 was supplemented.
  • the cells were washed to remove residual lentiviral particles in the supernatant, and continued to be cultured in a cell incubator for 5 days (the culture temperature was 37°C, and the carbon dioxide concentration was 5%), during which the cell density was maintained at 1 ⁇ 2 ⁇ 10 6 /ml.
  • the obtained CAR-T cells followed the nomenclature of the corresponding CAR molecules, namely BN001, BN003, BN004 and BN009, and the T cells not transduced with lentivirus were named Ctrl T.
  • the Ctrl T, BN001, BN003, BN004 and BN009 cells to be tested were washed twice with PBS, and resuspended with FACS buffer (PBS containing 0.1% sodium azide and 0.4% BSA).
  • FACS buffer PBS containing 0.1% sodium azide and 0.4% BSA.
  • APC-labeled anti-human NKG2D antibody and BV421-labeled anti-human CD3 antibody to the cell suspension to be tested according to the antibody instructions, and incubate at 4°C for 60 min.
  • Ctrl T cells as a negative control, the expression rates of NKG2D CAR molecules in BN001 and BN009 cells were detected by flow cytometry. Analyzed by CytExpert software.
  • the results are shown in Figure 3.
  • the gate was set according to the APC fluorescence signal level of Ctrl T cells, and the expression rate of CAR molecules in Ctrl T cells as a negative control was regarded as 0.64%, and the expression rate of CAR molecules in BN001 cells was about 92.17%.
  • the CAR molecule expression rate of BN003 cells is about 98.31%
  • the CAR molecule expression rate of BN004 cells is about 97.71%
  • the CAR molecule expression rate of BN009 cells is about 85.29%.
  • the Ctrl T, BN001, BN003, BN004 and BN009 cells to be tested were washed twice with PBS and resuspended with FACS buffer. According to the antibody instructions, PE-labeled anti-human CCR2b antibody and BV421-labeled anti-human CD3 antibody were added to the cell suspension to be detected, and incubated at 4°C for 60 min. Using Ctrl T cells not transfected with lentivirus as a negative control, the expression rate of CCR2b in the above CAR-T cells was detected by flow cytometry. Analyzed by CytExpert software.
  • Ctrl T cells can be divided into CCR2-negative and CCR2-positive cell populations.
  • the CCR2 expression rate of Ctrl T is about 44.20%
  • the CCR2b expression rate of BN001 is about 23.71%
  • the CCR2b expression rate of BN003 cells is about 99.37%
  • the BN004 cells The expression rate of CCR2b in BN009 cells is about 24.55%
  • the expression rate of CCR2b in BN009 cells is about 96.11%.
  • the Ctrl T, BN001, BN003, BN004 and BN009 cells to be tested were washed twice with PBS and resuspended with FACS buffer. According to the antibody instructions, PE-labeled anti-human CD40L antibody and BV421-labeled anti-human CD3 antibody were added to the cell suspension to be detected, and incubated at 4°C for 60 min. Using Ctrl T cells not transfected with lentivirus as a negative control, the CD expression rate of the above CAR-T cells was detected by flow cytometry. Analyzed by CytExpert software.
  • Ctrl T cells can be divided into CD40L-negative and CD40L-positive cell populations.
  • the CD40L expression rate of Ctrl T is about 2.75%
  • the CD40L expression rate of BN001 is about 21.43%
  • the CD40L expression rate of BN003 is about 41.85%
  • the CD40L expression rate of BN004 is about 2.75%.
  • the expression rate is about 86.74%
  • the expression rate of BN009 cells is about 71.86%.
  • Tested colorectal cancer cell lines also known as target cells or target cell lines: HCT 116 (McCoy's 5a medium + 10% fetal bovine serum + 100U/ml penicillin + 100 ⁇ g/ml streptomycin), LS174T (EMEM medium +10% fetal bovine serum+100U/ml penicillin+100 ⁇ g/ml streptomycin), LoVo (F-12K medium+10% fetal bovine serum+100U/ml penicillin+100 ⁇ g/ml streptomycin), SW480 (Leibovitz's L-15 Medium + 10% fetal bovine serum + 100 U/ml penicillin + 100 ⁇ g/ml streptomycin).
  • Tested ovarian cancer cell line SK-OV-3 (McCoy's 5a medium + 10% fetal bovine serum + 100 U/ml penicillin + 100 ⁇ g/ml streptomycin).
  • the above target cells were washed twice with PBS and resuspended with FACS buffer. Add APC-labeled anti-human MICA/MICB antibody to each target cell suspension according to the antibody instructions, and incubate at 4°C for 60 min. The target cells incubated without antibody were used as negative control, and the MICA/MICB expression rate of the target cells was detected by flow cytometry. Analyzed by CytExpert software.
  • the above target cells were washed twice with PBS and resuspended with FACS buffer. Add PC5.5-labeled anti-human ULBP-1 antibody to each target cell suspension according to the antibody instructions, and incubate at 4°C for 60 min. The target cells incubated without antibody were used as a negative control, and the expression rate of ULBP-2/5/6 in the target cells was detected by flow cytometry. Analyzed by CytExpert software.
  • the ULBP-1 expression rate of HCT116 is about 77.25%
  • the ULBP-1 expression rate of LoVo is about 9.09%
  • the ULBP-1 expression rate of other cells is lower than 3%.
  • the above target cells were washed twice with PBS and resuspended with FACS buffer. According to the antibody instructions, PE-labeled anti-human ULBP-2/5/6 antibodies were added to each target cell suspension, and incubated at 4°C for 60 min. The target cells incubated without antibody were used as a negative control, and the expression rate of ULBP-2/5/6 in the target cells was detected by flow cytometry. Analyzed by CytExpert software.
  • the results are shown in Figure 8.
  • the ULBP-2/5/6 expression rates of HCT116 and SW480 are both higher than 92%, the ULBP-2/5/6 expression rate of LoVo is about 86.82%, and the ULBP-2/5/6 expression rate of SK-OV-3 is about 86.82%.
  • the expression rate of 2/5/6 was about 56.90%, and the expression rate of ULBP-2/5/6 in LS174T was about 33.42%.
  • the above target cells were washed twice with PBS and resuspended with FACS buffer. According to the antibody instructions, PE-labeled anti-human ULBP-3 antibody was added to each target cell suspension, and incubated at 4°C for 60 min. The target cells incubated without antibody were used as a negative control, and the expression rate of ULBP-3 in the target cells was detected by flow cytometry. Analyzed by CytExpert software.
  • the above target cells were washed twice with PBS and resuspended with FACS buffer. According to the antibody instructions, PE-labeled anti-human ULBP-4 antibody was added to each target cell suspension, and incubated at 4°C for 60 min. The target cells incubated without antibody were used as a negative control, and the expression rate of ULBP-4 in the target cells was detected by flow cytometry. Analyzed by CytExpert software.
  • the above target cells were washed twice with PBS and resuspended with FACS buffer. Add APC-labeled anti-human CD40 antibody to each target cell suspension according to the antibody instructions, and incubate at 4°C for 60 min. The target cells incubated without antibody were used as a negative control, and the CD40 expression rate of the target cells was detected by flow cytometry. Analyzed by CytExpert software.
  • the Incucyte S3 Live Cell Dynamic Imaging Analyzer was used to detect the migration of T cells in real time.
  • Pretreatment of ClearView 96-well Chemotaxis Motility Microplate Add 20 ⁇ g/ml Protein-G protein to the upper chamber of the microplate, and place it at 37°C for 1 hour to coat the upper chamber microporous membrane; after washing with D-PBS, Add 5 ⁇ g/ml of ICAM-1 protein to the upper chamber of the microwell plate, place it at 37°C for 2 hours, and coat the upper chamber microporous membrane twice; finally use 1% BSA to coat the upper and lower surfaces of the upper chamber microporous membrane Carry out sealing for 30min.
  • the change of the total area of T cells in the upper chamber was analyzed by the Incucyte chemotaxis and migration analysis module software to reflect the chemotaxis and migration ability of T cells (the more cells migrated from the upper chamber to the lower chamber, the number of cells in the upper chamber and the corresponding The smaller the total cell area, the stronger the chemotaxis and migration ability).
  • Target cells once with AIM-V medium. Adjust the target cell density to 1 ⁇ 10 6 /ml, add DELFIA BATDA Reagent at 2 ⁇ l/ml, mix well, and incubate at 37°C for 30 minutes. After the target cells were washed three times with AIM-V medium, the target cells were seeded in a 96-well plate at a density of 1 ⁇ 10 4 /well. 100 ⁇ l of T cells were added, and cultured in a carbon dioxide incubator for 2 hours (the culture temperature was 37° C., and the carbon dioxide concentration was 5%).
  • Ctrl T, BN001, BN003, BN004, and BN009 were co-cultured with corresponding target cells in AIM-V medium without IL-2 (effect-to-target ratio 2.5:1). After 24 hours, dissolve the INF- ⁇ standard with ddH 2 O, place at room temperature for 15-20 minutes to ensure full dissolution, and dilute the standard according to the recommended gradient ratio. Aspirate the supernatant of the above co-cultured cells and dilute them 2-fold and 20-fold with ddH 2 O. The standard and experimental samples were added to the corresponding reaction wells, 100 ⁇ l per well.
  • mice Using LoVo as target cells and BN001 and BN009 as effector cells to test the inhibitory effect of subcutaneous tumor transplantation in mice.
  • Experiments were conducted with immunodeficient B-NDG mice to observe the effect of NKG2D CAR-T cells co-expressing CCR2 on tumor infiltration and inhibition.
  • B-NDG mice aged 6-8 weeks (Biocytogen Jiangsu Gene Biotechnology Co., Ltd.) were used for subcutaneous tumor efficacy experiments. A total of 24 mice were selected for the experiment, and were randomly divided into 4 groups, 6 mice in each group, which were vehicle control group, Ctrl T control group, BN001 control group, and BN009 experimental group.
  • the target cells in logarithmic growth phase and in good growth state were collected by trypsinization method, washed once with normal saline, and the cell density was adjusted to 2 ⁇ 10 7 /ml. 100 ⁇ l of cell suspension was subcutaneously injected into the right side of the B-NDG mice near the armpit, that is, each mouse was inoculated with 2 ⁇ 10 6 target cells, and the inoculation diary was regarded as day 0.
  • CAR-T cells (1 ⁇ 10 7 /monkey), Ctrl T cells (1 ⁇ 10 7 /monkey) and vehicle were respectively injected through the tail vein. (100 ⁇ l/only), the day of injection of the test substance was recorded as the 0th day of treatment. The tumor size and mouse weight were measured 2 to 3 times a week, and blood samples were collected on the 3rd, 10th, and 28th days. After adding EDTA for anticoagulation, qPCR was used to detect the retention of CAR-T cells in the blood cells in the mice. In this case, INF- ⁇ was detected by ELISA to monitor the release level of cytokines.
  • mice After 28 days of treatment, the mice were euthanized, and the tumor, heart, liver, spleen, lung, kidney, brain, ovary and other tissues were taken and weighed.
  • the tissues of 2 mice in each group were stored in a refrigerator at 80°C for DNA extraction , to detect the infiltration of CAR-T cells in tumor tissues and their distribution in various organs; the tissues of 2 mice in each group were fixed, and the morphology of tumor cells was detected by HE staining and the tissues were detected by immunohistochemistry.
  • mice in vehicle control group were about 947 mm 3 , and the average tumor burden was about 1.215 g; the average tumor volume of mice in Ctrl T control group was about
  • the average tumor volume of mice in the BN001 control group was about 1178mm 3 , and the average tumor burden was about 1.267g; the average tumor volume of mice in the BN009 experimental group was about 623mm 3 , and the average tumor The load is about 0.860g.
  • the average tumor burden of mice injected with BN009 cells was significantly reduced by about 32.1%.
  • LTR sequences in tumor tissue were detected by qPCR to measure the infiltration of CAR-T cells in tumor tissue.
  • the results showed that the average background signal level of LTR in the tumor tissue of the mice in the vehicle control group was 8.18 copies/ ⁇ g DNA; the average background signal level of LTR in the tumor tissue of the mice in the Ctrl T control group was 42.86 copies/ ⁇ g DNA;
  • the average LTR content in the tumor tissue of the mice in the BN001 control group was 2055.38 copies/ ⁇ g DNA; the average LTR content in the tumor tissue of the mice in the BN009 experimental group was 3872.70 copies/ ⁇ g DNA.
  • the homing ability of BN009 to subcutaneous transplanted tumor tissue in mice was significantly improved, with an increase rate of about 88.4%.
  • NKG2D ligands include MICA, MICB, ULBP-1, ULBP-2, ULBP-3, ULBP-4, ULBP-5 and ULBP-6, which are widely expressed in a variety of tumor cells.
  • NKG2D also known as CD314
  • CD314 is an important activating receptor in the innate immune system, mainly expressed on the surface of natural killer cells, ⁇ T cells and CD8+T cells.
  • NKG2D CAR-T does not require the antigen presentation process of MHC molecules to directly recognize tumor cells.
  • NKG2D ligands are expressed at high levels in epithelial tumor cells such as colorectal cancer, ovarian cancer, pancreatic cancer, and leukemia, but are not expressed or expressed at very low levels in normal cells. ideal target.
  • the surface of NKG2D CAR-T cells does not carry any foreign protein structures that may trigger the patient's immune response, thereby reducing the possibility of CAR-T cells being rejected by the patient's immune system.
  • Solid tumor cells can prevent the migration and infiltration of CAR-T cells into tumor tissue by secreting chemokines CXCL12 and CXCL5. In contrast, solid tumor cells secrete less chemokines that can promote CAR-T cell migration. These two factors make it difficult for CAR-T cells to reach solid tumor sites. Therefore, improving the specific recognition and sensitivity of CAR-T cells to tumor chemokines is one of the key factors affecting the therapeutic effect of CAR-T, and the combined expression of chemokine receptors in CAR-T cells is the solution to this problem. important method. Chemokines are a special class of cytokines, including more than 50 members.
  • chemokine receptors are correspondingly divided into four types: CCR, CXCR, CX3CR and XCR, with about 20 members.
  • One of the main mechanisms of action of chemokines is to induce directional migration of immune cells by forming concentration gradients soluble or immobilized in the matrix, thereby regulating the infiltration of immune cells in tissues.
  • some CAR-T technologies have used the combined expression of chemokine receptors to promote the rapid migration of CAR-T cells to cancer cells, thereby improving the tumor therapeutic effect of CAR-T cells.
  • Different types of solid tumors release different types and levels of chemokines, and have different immune escape mechanisms. Therefore, selecting the appropriate target antigen for a specific cancer type, combined with the expression of an appropriate chemokine receptor, is the key to improving the therapeutic effect of this type of CAR-T cells.
  • the present invention not only efficiently solves the problem of targeting malignant tumor lesions, but also effectively overcomes the problem of malignant tumor heterogeneity, and achieves synergistic and excellent treatment effect.
  • the present invention uses CCR2b as a co-expressed chemokine receptor to improve the ability of CAR-T cells to migrate to malignant tumor lesions, thereby improving the treatment efficiency.
  • CCL2 a chemokine mainly recognized by CCR2b, is abnormally highly expressed in various malignant tumors such as colorectal cancer, ovarian cancer, and pancreatic cancer, and its expression level is extremely low in normal tissue cells.
  • the affinity between CCR2b and CCL2 is extremely high (about 0.7nM, the smaller the IC 50 value, the higher the corresponding affinity and sensitivity), and the sensitivity is much higher than the combination of other chemokine receptors and chemokines (such as CCR2a/
  • the IC 50 value of the combination of CCL2 is about 5nM, and the C 50 value of the combination of CXCR3/CXCL11 is about 8.2nM).
  • the present invention uses NKG2D as the extracellular recognition domain of the CAR molecule, which can recognize various target antigens on the surface of tumor cells (including MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5, ULBP6), and can reduce The risk of reduced efficacy due to tumor heterogeneity or antigen loss is more conducive to improving the therapeutic effect on malignant tumors such as colorectal cancer, ovarian cancer, and pancreatic cancer. Therefore, the present invention further improves the effectiveness of treatment, and improves the ability of immune cells such as CAR-T cells to fight against the high heterogeneity of malignant tumors.
  • NKG2D CAR-T will also target immunosuppressive cells and new blood vessels in the tumor microenvironment, helping immune cells such as T cells overcome the immunosuppressive tumor microenvironment and improve the effect of tumor treatment.
  • the present invention uses CD40L as a second cofactor to more effectively activate the body's endogenous natural and adaptive immune responses. Activate the proliferation of T cells and the release of cytokines through CD40L, induce the transformation of M2 lineage macrophages into M1 lineage macrophages with anti-tumor activity, etc., and at the same time improve the ability of T cells to respond to tumors with certain antigen loss but high CD40 expression cell killing.
  • the NKG2D CAR molecule of the present invention when the NKG2D CAR molecule of the present invention, exogenous CCR2b protein and exogenous CD40L protein are jointly expressed, they can synergistically significantly improve the in vitro killing effect on tumor cells.
  • the present invention provides a novel and more efficient engineered immune cell (such as CAR-T cell), which can efficiently chemotaxis and migrate to malignant solid tumors such as colorectal cancer, ovarian cancer, and pancreatic cancer, and can effectively activate Endogenous immune system, and CAR-T cells that can effectively fight against tumor heterogeneity.
  • CAR-T cell engineered immune cell

Abstract

Provided are an engineered immune cell for combined expression of CCR2b and CD40L, and preparation thereof and an application thereof. By simultaneously expressing, in a CAR-T cell in a combined manner, a CAR molecule containing an NKG2D extracellular domain, and CCR2b and CD40L, the sensitivity of the CAR-T cell to a chemokine and the migration capability of the CAR-T cell to a solid tumor lesion can be improved by means of CCR2b; by means of CD40L, the endogenous natural and adaptive immune response of an organism can also be activated, the tumor treat effect is improved, and the relapse risk is reduced; and by means of an NKG2D CAR molecule, various target antigens on the surfaces of malignant solid tumor cells can further be identified, and the risk of reduction in therapeutic effect caused due to tumor heterogeneity or target antigen loss is reduced, thereby improving the tumor treatment effect. The engineered immune cell can synergistically and remarkably improve the in-vitro killing effect on tumor cells.

Description

联合表达CCR2b和CD40L的工程化免疫细胞及其制备和应用Engineered immune cells jointly expressing CCR2b and CD40L and its preparation and application 技术领域technical field
本发明属于肿瘤免疫和细胞治疗领域,具体地,涉及一种联合表达CCR2b和CD40L的工程化免疫细胞。The invention belongs to the field of tumor immunity and cell therapy, and in particular relates to an engineered immune cell jointly expressing CCR2b and CD40L.
背景技术Background technique
细胞免疫治疗是一种新兴的、具有显著疗效的肿瘤治疗模式,是一种自身免疫抗癌的新型治疗方法。它是运用生物技术和生物制剂对从病人体内采集的免疫细胞进行体外培养、修饰改造和扩增后回输到病人体内的方法,以激发、增强机体自身免疫功能,从而达到治疗肿瘤的目的。Cellular immunotherapy is an emerging tumor treatment mode with significant curative effect, and it is a new type of autoimmune anti-cancer treatment. It is a method of using biotechnology and biological agents to culture, modify and amplify immune cells collected from patients in vitro, and then infuse them back into the patient's body to stimulate and enhance the body's autoimmune function, so as to achieve the purpose of treating tumors.
T细胞是参与细胞免疫的一类重要的淋巴细胞,通过抗原呈递细胞的信号传递,可以特异性地识别并杀伤肿瘤细胞。然而,肿瘤细胞也会通过减少或丢失抗原表位、免疫抑制作用、肿瘤异质性(即同一种恶性肿瘤在不同患者个体间或者同一患者体内不同部位肿瘤细胞间从基因型到表型上存在的差异)等方式阻碍T细胞的特异性识别,从而逃避机体的免疫应答。T cells are an important class of lymphocytes involved in cellular immunity, and can specifically recognize and kill tumor cells through the signal transmission of antigen-presenting cells. However, tumor cells can also be affected by the reduction or loss of antigenic epitopes, immunosuppression, and tumor heterogeneity (that is, the same malignant tumor exists in different patients or between tumor cells in different parts of the same patient from genotype to phenotype). Differences) and other ways to hinder the specific recognition of T cells, thereby evading the body's immune response.
嵌合抗原受体T细胞(chimeric antigen receptor T cell,CAR-T)疗法正是针对该问题应运而生的。具体来说,CAR分子是一种人为设计和构建的受体分子,由信号肽、胞外抗原结合域、铰链区、跨膜区、共刺激结构域、胞内信号传导结构域等部分组成。因此,CAR分子具有特异性识别肿瘤表面抗原、激活T细胞杀伤活性和刺激T细胞增殖等功能。通过采集培养肿瘤患者的T细胞并以人工方法转导CAR分子的编码基因,使患者自体的T细胞表达CAR分子。回输至患者体内后,T细胞可以通过CAR分子高效且特异性地识别并杀伤肿瘤细胞,达到癌症治疗的效果。Chimeric antigen receptor T cell (chimeric antigen receptor T cell, CAR-T) therapy was developed to address this problem. Specifically, the CAR molecule is an artificially designed and constructed receptor molecule, which consists of a signal peptide, an extracellular antigen-binding domain, a hinge region, a transmembrane region, a co-stimulatory domain, and an intracellular signaling domain. Therefore, CAR molecules have the functions of specifically recognizing tumor surface antigens, activating T cell killing activity, and stimulating T cell proliferation. By collecting and culturing T cells from tumor patients and artificially transducing the gene encoding CAR molecules, the patient's own T cells can express CAR molecules. After reinfusion into the patient's body, T cells can efficiently and specifically recognize and kill tumor cells through CAR molecules, achieving the effect of cancer treatment.
CAR-T疗法的概念从1989年首次被首次提出以来,经历了三十年的发展及多轮的技术更迭(图1)。第一代CAR-T仅有作为胞外抗原结合域的单链抗体和作为胞内信号传导结构域的CD3ζ,无法完全激活T细胞的活性,治疗效果不佳。第二代CAR-T在第一代CAR-T的基础上引入了一个共刺激结构域,提高了T细胞的体外增殖能力和细胞因子释放水平。第三代CAR-T在第二代CAR-T的基础上,增加了一个共刺激结构域,虽然可以提高T细胞的杀伤活性,但有可能诱发细胞因子的过量释放。因此,新一代CAR-T在第二代CAR-T的基础上联合表达其他辅助因子,例如联合表达IL-12或IL-2Rβ胞内的STAT3/5结合结构域等,有助于提高肿瘤杀伤活性和安全性等效果。Since the concept of CAR-T therapy was first proposed in 1989, it has experienced 30 years of development and multiple rounds of technological changes (Figure 1). The first-generation CAR-T only has a single-chain antibody as the extracellular antigen-binding domain and CD3ζ as the intracellular signaling domain, which cannot fully activate the activity of T cells, and the therapeutic effect is not good. The second-generation CAR-T introduced a co-stimulatory domain on the basis of the first-generation CAR-T, which improved the in vitro proliferation ability and cytokine release level of T cells. The third-generation CAR-T adds a co-stimulatory domain to the second-generation CAR-T. Although it can improve the killing activity of T cells, it may induce excessive release of cytokines. Therefore, on the basis of the second-generation CAR-T, the new-generation CAR-T jointly expresses other auxiliary factors, such as the joint expression of IL-12 or the STAT3/5 binding domain in the IL-2Rβ cell, which helps to improve tumor killing Effects such as activity and safety.
虽然在血液瘤中CAR-T治疗获得令人满意的效果,但CAR-T对于实体瘤的治疗效果仍有很大的提升空间。原因在于:(1)很多实体肿瘤都难以在早期被发现,具有恶性程度高、复发率高、预后差等特点。例如,83%的结直肠癌患者在首次确诊 时已处于中晚期,且44%的患者已经出现了肝、肺等部位的转移,近半数患者生存期不到5年;约70%的卵巢癌患者在确诊时,癌细胞已经发生转移,难以通过手术、化疗、放疗手段治愈,治疗后的复发率仍高达70%以上;90%的胰腺癌患者被确诊时已属晚期,5年生存率仅7%。(2)在实体瘤治疗过程中,肿瘤组织往往具有免疫抑制性的微环境,可以阻碍CAR-T细胞的迁移和浸润。(3)很多恶性实体肿瘤还具有异质性高的特点,单一靶点抗原往往不能达到最佳的治疗效果,且有复发的风险。因此,用于结直肠癌、卵巢癌、胰腺癌等恶性实体肿瘤患者的CAR-T细胞疗法,需要更进一步提高其作用效率和有效性。Although CAR-T therapy has achieved satisfactory results in hematological tumors, there is still a lot of room for improvement in the therapeutic effect of CAR-T on solid tumors. The reasons are as follows: (1) Many solid tumors are difficult to be detected in the early stage, and have the characteristics of high malignancy, high recurrence rate, and poor prognosis. For example, 83% of colorectal cancer patients were in the middle and late stage when they were first diagnosed, and 44% of the patients had metastases to the liver, lung and other parts, and nearly half of the patients survived for less than 5 years; about 70% of ovarian cancer When patients are diagnosed, the cancer cells have already metastasized, and it is difficult to be cured by surgery, chemotherapy, and radiotherapy. The recurrence rate after treatment is still as high as 70%; 90% of pancreatic cancer patients are diagnosed at an advanced stage, and the 5-year survival rate is only 7%. (2) During the treatment of solid tumors, tumor tissues often have an immunosuppressive microenvironment, which can hinder the migration and infiltration of CAR-T cells. (3) Many malignant solid tumors are also characterized by high heterogeneity, and a single target antigen often cannot achieve the best therapeutic effect, and there is a risk of recurrence. Therefore, CAR-T cell therapy for patients with malignant solid tumors such as colorectal cancer, ovarian cancer, and pancreatic cancer needs to further improve its efficiency and effectiveness.
综上所述,本领域仍然需要进一步的研究,针对恶性肿瘤(尤其是实体瘤)开发一种能更高效、治疗效果更好的工程化免疫细胞。To sum up, further research is still needed in this field to develop an engineered immune cell with higher efficiency and better therapeutic effect for malignant tumors (especially solid tumors).
发明内容Contents of the invention
本发明的目的是针对恶性肿瘤(尤其是实体瘤)提供一种能更高效、治疗效果更好的工程化免疫细胞(如CAR-T细胞)。The purpose of the present invention is to provide an engineered immune cell (such as CAR-T cell) with higher efficiency and better therapeutic effect for malignant tumors (especially solid tumors).
本发明的又一目的是提供一种联合表达CCR2b和CD40L的工程化免疫细胞(如CAR-T细胞)及其制法和应用。Another object of the present invention is to provide an engineered immune cell (such as CAR-T cell) jointly expressing CCR2b and CD40L and its preparation method and application.
本发明的第一方面,提供了一种工程化免疫细胞,所述工程化免疫细胞为T细胞或NK细胞,并且所述的免疫细胞具有以下特征:The first aspect of the present invention provides an engineered immune cell, the engineered immune cell is a T cell or NK cell, and the immune cell has the following characteristics:
(a)所述免疫细胞表达嵌合抗原受体(chimeric antigen receptor,CAR),其中所述CAR靶向肿瘤细胞的表面标志物,其中所述CAR的抗原结合结构域包括NKG2D的胞外结构域;(a) the immune cell expresses a chimeric antigen receptor (chimeric antigen receptor, CAR), wherein the CAR targets surface markers of tumor cells, wherein the antigen-binding domain of the CAR includes the extracellular domain of NKG2D ;
(b)所述的免疫细胞表达外源的CCR2b蛋白;和(b) said immune cell expresses an exogenous CCR2b protein; and
(c)所述的免疫细胞表达外源的CD40L蛋白。(c) The immune cells express exogenous CD40L protein.
在另一优选例中,所述的T细胞包括αβT、γδT细胞、NKT细胞、MAIT细胞,或其组合。In another preferred embodiment, the T cells include αβT, γδT cells, NKT cells, MAIT cells, or combinations thereof.
在另一优选例中,所述的工程化免疫细胞选自下组:In another preferred example, the engineered immune cells are selected from the following group:
(i)嵌合抗原受体T细胞(CAR-T细胞);(i) chimeric antigen receptor T cells (CAR-T cells);
(ii)嵌合抗原受体NK细胞(CAR-NK细胞)。(ii) Chimeric antigen receptor NK cells (CAR-NK cells).
在另一优选例中,所述的CCR2b蛋白和/或CD40L可以是组成型表达或诱导型表达。In another preferred example, the CCR2b protein and/or CD40L can be expressed constitutively or inducibly.
在另一优选例中,提供了一种嵌合抗原受体T细胞(CAR-T细胞),所述CAR-T细胞具有以下一个或多个特征:In another preferred embodiment, a chimeric antigen receptor T cell (CAR-T cell) is provided, and the CAR-T cell has one or more of the following characteristics:
(a)所述细胞表达嵌合抗原受体CAR,所述CAR靶向肿瘤细胞的表面标志物;和(a) the cell expresses a chimeric antigen receptor CAR that targets a surface marker of the tumor cell; and
(b)当所述CAR-T细胞接触诱导剂时,所述CAR-T细胞诱导表达CCR2b和/或CD40L蛋白。(b) When the CAR-T cells are exposed to an inducer, the CAR-T cells are induced to express CCR2b and/or CD40L proteins.
在另一优选例中,在所述的CAR细胞中,CAR、CCR2b和CD40L蛋白是串联表达的。In another preferred example, in the CAR cells, CAR, CCR2b and CD40L proteins are expressed in series.
在另一优选例中,在所述的CAR细胞中,CAR、CCR2b和CD40L蛋白各自独立地表达的。In another preferred example, in the CAR cells, CAR, CCR2b and CD40L proteins are independently expressed.
在另一优选例中,所述“激活”指所述CAR与肿瘤细胞的表面标志物结合。In another preferred embodiment, the "activation" refers to the binding of the CAR to surface markers of tumor cells.
在另一优选例中,所述“肿瘤的表面标志物”指肿瘤表面的特异性抗原。In another preferred example, the "tumor surface marker" refers to a specific antigen on the surface of the tumor.
在另一优选例中,所述的嵌合抗原受体CAR定位于所述工程化免疫细胞的细胞膜。In another preferred example, the chimeric antigen receptor CAR is located on the cell membrane of the engineered immune cells.
在另一优选例中,所述的嵌合抗原受体CAR定位于所述CAR-T细胞的细胞膜。In another preferred example, the chimeric antigen receptor CAR is located on the cell membrane of the CAR-T cell.
在另一优选例中,所述的CCR2b蛋白定位于所述CAR-T细胞的细胞膜。In another preferred example, the CCR2b protein is located on the cell membrane of the CAR-T cells.
在另一优选例中,所述CAR的结构如式I所示:In another preferred example, the structure of the CAR is shown in Formula I:
L-NKG2D-H-TM-C-CD3ζ  (I)L-NKG2D-H-TM-C-CD3ζ (I)
式中,In the formula,
L为无或信号肽序列;L is nothing or a signal peptide sequence;
NKG2D为NKG2D胞外结构域或其活性片段;NKG2D is an NKG2D extracellular domain or an active fragment thereof;
H为无或铰链区;H is none or hinge region;
TM为跨膜结构域;TM is the transmembrane domain;
C为共刺激信号结构域;C is costimulatory signal domain;
CD3ζ为源于CD3ζ的胞浆信号传导序列(包括野生型、或其突变体/修饰体);CD3ζ is a cytoplasmic signaling sequence derived from CD3ζ (including wild type, or mutants/modifiers thereof);
所述“-”为连接肽或肽键。The "-" is a connecting peptide or a peptide bond.
在另一优选例中,所述L分别选自下组的蛋白的信号肽:CD8、GM-CSF、CD4、CD28、CD137、或其突变/修饰体、或其组合。In another preferred example, said L is respectively selected from signal peptides of proteins in the following group: CD8, GM-CSF, CD4, CD28, CD137, or mutants/modifications thereof, or combinations thereof.
在另一优选例中,所述H选自下组的蛋白的铰链区:CD8、CD28、CD137、IgG、或其组合。In another preferred example, the H is selected from the hinge region of proteins in the following group: CD8, CD28, CD137, IgG, or a combination thereof.
在另一优选例中,所述TM选自下组的蛋白的跨膜区:CD28、CD3epsilon、CD45、CD4、CD5、CD8、CD9、CD16、CD22、CD33、CD37、CD64、CD80、CD86、CD134、CD137、CD154、CD278、CD152、CD279、CD233、CD314、或其突变/修饰体、或其组合。In another preferred embodiment, the TM is selected from the transmembrane region of the following group of proteins: CD28, CD3epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134 , CD137, CD154, CD278, CD152, CD279, CD233, CD314, or a mutation/modification thereof, or a combination thereof.
在另一优选例中,所述C选自下组的蛋白的共刺激结构域:OX40、CD2、CD7、CD27、CD28、CD30、CD40、CD70、CD134、4-1BB(CD137)、PD1、Dap10、CDS、ICAM-1、LFA-1(CD11a/CD18)、ICOS(CD278)、NKG2D、GITR、OX40L、或其突变/修饰体、或其组合。In another preferred example, the C is selected from the co-stimulatory domains of proteins in the following group: OX40, CD2, CD7, CD27, CD28, CD30, CD40, CD70, CD134, 4-1BB (CD137), PD1, Dap10 , CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), NKG2D, GITR, OX40L, or mutants/modifications thereof, or combinations thereof.
在另一优选例中,C为4-1BB来源的共刺激结构域。In another preferred example, C is a co-stimulatory domain derived from 4-1BB.
在另一优选例中,所述NKG2D的胞外结构域的氨基酸序列如SEQ ID NO:1第73-216位所示或如SEQ ID No:4所示。In another preferred example, the amino acid sequence of the extracellular domain of NKG2D is as shown in SEQ ID NO:1 73-216 or as shown in SEQ ID No:4.
在另一优选例中,除了式I所示的第一CAR之外,所述CAR细胞还含有用于针对第二抗原的第二CAR,所述的第二CAR的结构如式II所示:In another preferred example, in addition to the first CAR shown in formula I, the CAR cell also contains a second CAR for targeting the second antigen, and the structure of the second CAR is shown in formula II:
L-scFv-H-TM-C-CD3ζ(II)L-scFv-H-TM-C-CD3ζ(II)
式中,In the formula,
L为无或信号肽序列;L is nothing or a signal peptide sequence;
scFv为抗原结合结构域;scFv is an antigen-binding domain;
H为无或铰链区;H is none or hinge region;
TM为跨膜结构域;TM is the transmembrane domain;
C为共刺激结构域;C is costimulatory domain;
CD3ζ为源于CD3ζ的胞浆信号传导序列或其突变/修饰体;CD3ζ is a cytoplasmic signaling sequence derived from CD3ζ or a mutation/modification thereof;
所述“-”为连接肽或肽键。The "-" is a connecting peptide or a peptide bond.
在另一优选例中,所述scFv为靶向肿瘤抗原的抗体单链可变区序列。In another preferred example, the scFv is an antibody single-chain variable region sequence targeting a tumor antigen.
在另一优选例中,所述scFv为靶向选自下组抗原的抗体单链可变区序列:CD19、CD20、CD22、CD123、CD47、CD138、CD33、CD30、CD271、GUCY2C、CD24、CD133、CD44、CD166、CD133、CD276、ABCB5、ALDH1、间皮素(mesothelin,MSLN)、EGFR、GPC3、BCMA、ErbB2、NKG2D配体(ligands)、LMP1、EpCAM、VEGFR-1、Lewis-Y、ROR1、Claudin18.2、CEA、TAG-72、TROP2或其组合。In another preferred example, the scFv is an antibody single-chain variable region sequence targeting an antigen selected from the following group: CD19, CD20, CD22, CD123, CD47, CD138, CD33, CD30, CD271, GUCY2C, CD24, CD133 , CD44, CD166, CD133, CD276, ABCB5, ALDH1, mesothelin (MSLN), EGFR, GPC3, BCMA, ErbB2, NKG2D ligands (ligands), LMP1, EpCAM, VEGFR-1, Lewis-Y, ROR1 , Claudin18.2, CEA, TAG-72, TROP2, or combinations thereof.
在另一优选例中,所述NKG2D的氨基酸序列如SEQ ID NO:1所示,其中胞外结构域为第73-216位。In another preferred example, the amino acid sequence of the NKG2D is shown in SEQ ID NO: 1, wherein the extracellular domain is 73-216.
在另一优选例中,所述的CCR2b蛋白包括全长CCR2b蛋白或其活性片段(即保留可与相应配体发生结合的功能的活性片段)。在另一优选例中,所述CCR2b蛋白的氨基酸序列如SEQ ID NO:2所示。In another preferred example, the CCR2b protein includes a full-length CCR2b protein or an active fragment thereof (ie, an active fragment retaining the function of binding to a corresponding ligand). In another preferred example, the amino acid sequence of the CCR2b protein is shown in SEQ ID NO:2.
在另一优选例中,所述的CD40L蛋白包括全长CD40L蛋白或其活性片段(即保留可与CD40发生结合的功能的活性片段)。In another preferred embodiment, the CD40L protein includes full-length CD40L protein or its active fragment (ie, the active fragment retains the function of binding to CD40).
在另一优选例中,所述CD40L蛋白的氨基酸序列如SEQ ID NO:5所示。In another preferred example, the amino acid sequence of the CD40L protein is shown in SEQ ID NO:5.
在另一优选例中,式I所示的第一CAR和式II所示的第二CAR可合而为一,从而构成如式IIIa或IIIb所示的CAR:In another preferred example, the first CAR shown in formula I and the second CAR shown in formula II can be combined into one to form a CAR shown in formula IIIa or IIIb:
L-NKG2D-scFv-H-TM-C-CD3ζ  (IIIa)L-NKG2D-scFv-H-TM-C-CD3ζ (IIIa)
L-scFv-NKG2D-H-TM-C-CD3ζ  (IIIb)L-scFv-NKG2D-H-TM-C-CD3ζ (IIIb)
式中,In the formula,
L为无或信号肽序列;L is nothing or a signal peptide sequence;
NKG2D为NKG2D胞外结构域或其活性片段;NKG2D is an NKG2D extracellular domain or an active fragment thereof;
scFv为抗原结合结构域;scFv is an antigen-binding domain;
H为无或铰链区;H is none or hinge region;
TM为跨膜结构域;TM is the transmembrane domain;
C为共刺激结构域;C is costimulatory domain;
CD3ζ为源于CD3ζ的胞浆信号传导序列或其突变/修饰体;CD3ζ is a cytoplasmic signaling sequence derived from CD3ζ or a mutation/modification thereof;
所述“-”为连接肽或肽键。The "-" is a connecting peptide or a peptide bond.
本发明的第二方面,提供了一种制备本发明第一方面所述的工程化免疫细胞的方法,包括以下步骤:The second aspect of the present invention provides a method for preparing the engineered immune cells described in the first aspect of the present invention, comprising the following steps:
(A)提供一待改造的免疫细胞;和(A) providing an immune cell to be modified; and
(B)对所述的免疫细胞进行改造,从而使得所述的免疫细胞表达CAR分子以及外源的CCR2b蛋白和外源的CD40L蛋白,从而获得本发明第一方面所述的工程化免疫细胞,其中所述CAR靶向肿瘤细胞的表面标志物,其中所述CAR的抗原结合结构域包括NKG2D的胞外结构域。(B) transforming the immune cells so that the immune cells express CAR molecules and exogenous CCR2b protein and exogenous CD40L protein, thereby obtaining the engineered immune cells described in the first aspect of the present invention, Wherein the CAR targets surface markers of tumor cells, wherein the antigen binding domain of the CAR includes the extracellular domain of NKG2D.
在另一优选例中,在步骤(B)中,包括:In another preference, in step (B), including:
(B1)将表达所述CAR的第一表达盒导入所述免疫细胞;(B2)将表达CCR2b的第二表达盒导入所述免疫细胞;(B3)将表达CD40L的第三表达盒导入所述免疫细胞;其中所述的步骤(B1)、(B2)和(B3)可以按任意次序进行。(B1) introducing the first expression cassette expressing the CAR into the immune cells; (B2) introducing the second expression cassette expressing CCR2b into the immune cells; (B3) introducing the third expression cassette expressing CD40L into the immune cells Immune cells; wherein the steps (B1), (B2) and (B3) can be performed in any order.
在另一优选例中,所述的步骤(B1)可在步骤(B2)之前、之后、同时、或交替进行。In another preferred example, the step (B1) can be performed before, after, simultaneously or alternately with the step (B2).
在另一优选例中,所述的步骤(B1)可在步骤(B3)之前、之后、同时、或交替进行。In another preferred example, the step (B1) can be performed before, after, simultaneously, or alternately before the step (B3).
在另一优选例中,所述的步骤(B2)可在步骤(B3)之前、之后、同时、或交替进行。In another preferred example, the step (B2) can be performed before, after, simultaneously or alternately with the step (B3).
在另一优选例中,所述的步骤(B1)、(B2)和(B3)同时或交替进行。In another preferred example, the steps (B1), (B2) and (B3) are performed simultaneously or alternately.
在另一优选例中,提供了一种制备本发明所述的CAR-T细胞的方法,包括以下步骤:In another preferred embodiment, a method for preparing the CAR-T cells of the present invention is provided, comprising the following steps:
(A)提供一种待改造的T细胞;(A) providing a T cell to be modified;
(B)对所述的T细胞进行改造,使得所述的T细胞表达所述的CAR分子以及外源的CCR2b蛋白和外源的CD40L蛋白,从而获得本发明第一方面所述的工程化免疫细胞。(B) transforming the T cells so that the T cells express the CAR molecule and exogenous CCR2b protein and exogenous CD40L protein, thereby obtaining the engineered immunity described in the first aspect of the present invention cell.
在另一优选例中,在步骤(B)中,包括:将表达所述CAR的第一表达盒导入所 述T细胞;和将表达CCR2b的第二表达盒和表达CD40L的第三表达盒导入所述T细胞;其中所述的导入步骤可以按任意次序进行。In another preferred embodiment, in step (B), it includes: introducing the first expression cassette expressing the CAR into the T cell; and introducing the second expression cassette expressing CCR2b and the third expression cassette expressing CD40L The T cells; wherein the introducing steps can be performed in any order.
在另一优选例中,对于所述的第一、第二和第三表达盒中任何两个表达盒(以第一表达盒和第二表达盒为例),其转录方向是同向的(→→)、相向的(→←)或相背的(←→)。In another preferred example, for any two expression cassettes in the first, second and third expression cassettes (taking the first expression cassette and the second expression cassette as an example), their transcription directions are in the same direction ( →→), opposite (→←) or opposite (←→).
在另一优选例中,所述的第一表达盒、第二表达盒和第三表达盒位于相同或不同的载体上。In another preferred example, the first expression cassette, the second expression cassette and the third expression cassette are located on the same or different vectors.
在另一优选例中,所述的第一表达盒、第二表达盒和第三表达盒位于同一载体。In another preferred example, the first expression cassette, the second expression cassette and the third expression cassette are located in the same vector.
在另一优选例中,当所述的CAR分子、外源的CCR2b蛋白和外源的CD40L蛋白中的两个或三个串联表达时,在两个蛋白之间,还包括连接肽。In another preferred example, when two or three of the CAR molecule, the exogenous CCR2b protein and the exogenous CD40L protein are expressed in series, a connecting peptide is also included between the two proteins.
在另一优选例中,所述连接肽为P2A或T2A。In another preferred example, the connecting peptide is P2A or T2A.
在另一优选例中,所述的载体为病毒载体,较佳地所述病毒载体含有串联形式的第一和第二表达盒。In another preferred example, the vector is a viral vector, preferably the viral vector contains the first and second expression cassettes in tandem form.
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、逆转录病毒载体、转座子、其他基因转移系统、或其组合。In another preferred embodiment, the vector is selected from the group consisting of DNA, RNA, plasmid, lentiviral vector, adenoviral vector, retroviral vector, transposon, other gene transfer systems, or combinations thereof.
在另一优选例中,所述的载体为pCDH系列慢病毒载体。In another preferred example, the vector is a pCDH series lentiviral vector.
本发明的第三方面,提供了一种制剂,所述制剂含有本发明第一方面所述的工程化免疫细胞,以及药学上可接受的载体、稀释剂或赋形剂。The third aspect of the present invention provides a preparation, which contains the engineered immune cells described in the first aspect of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient.
在另一优选例中,所述制剂含有本发明所述的CAR-T细胞,以及药学上可接受的载体、稀释剂或赋形剂。In another preferred embodiment, the preparation contains the CAR-T cells of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient.
在另一优选例中,所述制剂为液态制剂。In another preferred example, the formulation is a liquid formulation.
在另一优选例中,所述制剂的剂型包括注射剂。In another preferred example, the dosage form of the preparation includes injection.
在另一优选例中,所述制剂中所述工程化免疫细胞(如CAR-T细胞)的浓度为1×10 3-1×10 8个细胞/ml,较佳地1×10 4-1×10 7个细胞/ml。 In another preferred example, the concentration of the engineered immune cells (such as CAR-T cells) in the preparation is 1×10 3 -1×10 8 cells/ml, preferably 1×10 4 -1 ×10 7 cells/ml.
本发明的第四方面,提供了如本发明第一方面所述的工程化免疫细胞的用途,用于制备预防和/或治疗癌症的药物或制剂。The fourth aspect of the present invention provides the use of the engineered immune cell according to the first aspect of the present invention for preparing a drug or preparation for preventing and/or treating cancer.
在另一优选例中,提供了如本发明第一方面所述的CAR-T细胞的用途,用于制备预防和/或治疗癌症或肿瘤的药物或制剂。In another preferred embodiment, the use of the CAR-T cells according to the first aspect of the present invention is provided for the preparation of drugs or preparations for preventing and/or treating cancer or tumors.
在另一优选例中,所述制剂含有CAR-T细胞,以及药学上可接受的载体、稀释剂或赋形剂。In another preferred embodiment, the preparation contains CAR-T cells, and a pharmaceutically acceptable carrier, diluent or excipient.
在另一优选例中,所述肿瘤包括实体瘤。In another preferred example, the tumor includes a solid tumor.
在另一优选例中,所述肿瘤选自下组:结肠癌、直肠癌、卵巢癌、或胰腺癌。In another preferred example, the tumor is selected from the group consisting of colon cancer, rectal cancer, ovarian cancer, or pancreatic cancer.
在另一优选例中,所述肿瘤为NKG2D配体高表达和/或趋化因子高表达和/或 CD40高表达的肿瘤。In another preferred example, the tumor is a tumor with high expression of NKG2D ligand and/or high expression of chemokines and/or high expression of CD40.
在另一优选例中,所述肿瘤为NKG2D配体高表达和趋化因子高表达的肿瘤。In another preferred example, the tumor is a tumor with high expression of NKG2D ligands and high expression of chemokines.
在另一优选例中,所述肿瘤为NKG2D配体高表达和趋化因子高表达的肿瘤。In another preferred example, the tumor is a tumor with high expression of NKG2D ligands and high expression of chemokines.
在另一优选例中,所述肿瘤为NKG2D配体高表达和CD40高表达的肿瘤。In another preferred example, the tumor is a tumor with high expression of NKG2D ligand and high expression of CD40.
在另一优选例中,所述的趋化因子选自下组:CCL2、CCL7、或其组合。In another preferred embodiment, the chemokine is selected from the group consisting of CCL2, CCL7, or a combination thereof.
在另一优选例中,所述肿瘤为NKG2D配体高表达、趋化因子CCL2和/或CCL7高表达和CD40高表达的肿瘤。In another preferred example, the tumor is a tumor with high expression of NKG2D ligand, high expression of chemokines CCL2 and/or CCL7, and high expression of CD40.
在另一优选例中,所述肿瘤为NKG2D配体(包括MICA、MICB、ULBP1、ULBP2、ULBP3、ULBP4、ULBP5、ULBP6中的任意一种,或其组合)高表达的肿瘤。In another preferred example, the tumor is a tumor with high expression of NKG2D ligands (including any one of MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5, ULBP6, or a combination thereof).
在另一优选例中,所述肿瘤为NKG2D配体(包括MICA、MICB、ULBP1、ULBP2、ULBP3、ULBP4、ULBP5、ULBP6中的任意一种,或其组合)高表达和/或趋化因子(包括CCL2、CCL7、CCL8、CCL12、CCL13、CCL16中的任意一种,或其组合)高表达和/或CD40高表达的肿瘤。In another preferred example, the tumor has high expression of NKG2D ligands (including any one of MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5, ULBP6, or a combination thereof) and/or chemokines ( Including tumors with high expression of CCL2, CCL7, CCL8, CCL12, CCL13, CCL16, or their combination) and/or high expression of CD40.
本发明的第五方面,提供了一种用于制备本发明第一方面所述的工程化免疫细胞的试剂盒,所述试剂盒含有容器,以及位于容器内的:The fifth aspect of the present invention provides a kit for preparing the engineered immune cell described in the first aspect of the present invention, the kit includes a container, and in the container:
(1)第一核酸序列,所述第一核酸序列含有用于表达所述CAR的第一表达盒,其中所述CAR的抗原结合结构域为NKG2D的胞外结构域;(1) a first nucleic acid sequence containing a first expression cassette for expressing the CAR, wherein the antigen-binding domain of the CAR is the extracellular domain of NKG2D;
(2)第二核酸序列,所述第二核酸序列含有用于联合表达CCR2b的第二表达盒;和(2) a second nucleic acid sequence containing a second expression cassette for the combined expression of CCR2b; and
(3)第三核酸序列,所述第三核酸序列含有用于联合表达CD40L的第三表达盒。(3) A third nucleic acid sequence containing a third expression cassette for co-expressing CD40L.
在另一优选例中,提供了一种用于制备本发明第一方面所述的工程化免疫细胞的试剂盒,所述试剂盒含有容器,以及位于容器内的:In another preferred embodiment, a kit for preparing the engineered immune cell according to the first aspect of the present invention is provided, the kit includes a container, and in the container:
(1)第一核酸序列,所述第一核酸序列含有用于表达所述CAR的第一表达盒;(1) a first nucleic acid sequence containing a first expression cassette for expressing the CAR;
(2)第二核酸序列,所述第二核酸序列含有用于联合表达CCR2b的第二表达盒;和(2) a second nucleic acid sequence containing a second expression cassette for the combined expression of CCR2b; and
(3)第三核酸序列,所述第三核酸序列含有用于联合表达CD40L的第三表达盒。(3) A third nucleic acid sequence containing a third expression cassette for co-expressing CD40L.
在另一优选例中,所述的第一、第二和第三核酸序列为独立的或相连的。In another preferred example, the first, second and third nucleic acid sequences are independent or linked.
在另一优选例中,所述的第一、第二和第三核酸序列位于相同或不同的容器内。In another preferred example, the first, second and third nucleic acid sequences are located in the same or different containers.
在另一优选例中,所述的第一、第二和第三核酸序列位于相同或不同的载体上。In another preferred example, the first, second and third nucleic acid sequences are located on the same or different vectors.
在另一优选例中,所述的第一、第二和第三核酸序列位于同一载体。In another preferred example, the first, second and third nucleic acid sequences are located in the same vector.
在另一优选例中,当所述的第一、第二和第三核酸序列中两个或三个位于同一载体时,在所述其之间,还包括用于表达连接肽的连接肽表达盒。In another preferred example, when two or three of the first, second and third nucleic acid sequences are located on the same vector, a connecting peptide expression for expressing the connecting peptide is also included between them. box.
在另一优选例中,所述连接肽为P2A或T2A。In another preferred example, the connecting peptide is P2A or T2A.
在另一优选例中,所述的载体为病毒载体,较佳地所述病毒载体含有串联形式 的第一、第二和第三核酸序列。In another preferred embodiment, the vector is a viral vector, preferably the viral vector contains the first, second and third nucleic acid sequences in tandem form.
本发明的第六方面,提供了一种肿瘤的治疗方法,包括步骤:将安全有效量的本发明第三方面所述的制剂,施用于所需对象,从而治疗所述对象的肿瘤。The sixth aspect of the present invention provides a method for treating tumors, comprising the step of: administering a safe and effective amount of the preparation described in the third aspect of the present invention to a subject in need, thereby treating the subject's tumor.
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。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, we will not repeat them here.
附图说明Description of drawings
图1显示了历代CAR分子的结构。Figure 1 shows the structures of CAR molecules of different generations.
图2显示了CAR分子的结构。Figure 2 shows the structure of the CAR molecule.
图3显示了流式检测各CAR-T细胞的NKG2D CAR分子的表达率。Figure 3 shows the expression rate of NKG2D CAR molecules of each CAR-T cell detected by flow cytometry.
图4显示了流式检测各CAR-T细胞的CCR2b的表达率。Figure 4 shows the expression rate of CCR2b of each CAR-T cell detected by flow cytometry.
图5显示了流式检测各CAR-T细胞的CD40L的表达率。Figure 5 shows the expression rate of CD40L of each CAR-T cell detected by flow cytometry.
图6显示了流式检测靶细胞中NKG2D配体(MICA/MICB)的表达率。Figure 6 shows the expression rate of NKG2D ligands (MICA/MICB) in target cells detected by flow cytometry.
图7显示了流式检测靶细胞中NKG2D配体(ULBP-1)的表达率。Figure 7 shows the expression rate of NKG2D ligand (ULBP-1) in target cells detected by flow cytometry.
图8显示了流式检测靶细胞中NKG2D配体(ULBP-2/5/6)的表达率。Figure 8 shows the expression rate of NKG2D ligands (ULBP-2/5/6) in target cells detected by flow cytometry.
图9显示了流式检测靶细胞中NKG2D配体(ULBP-3)的表达率。Figure 9 shows the expression rate of NKG2D ligand (ULBP-3) in target cells detected by flow cytometry.
图10显示了流式检测靶细胞中NKG2D配体(ULBP-4)的表达率。Figure 10 shows the expression rate of NKG2D ligand (ULBP-4) in target cells detected by flow cytometry.
图11显示了流式检测靶细胞中CD40的表达率。Figure 11 shows the expression rate of CD40 in target cells detected by flow cytometry.
图12显示了Incucyte实时检测BN009的趋化迁移能力的结果。Figure 12 shows the results of Incucyte's real-time detection of the chemotaxis and migration ability of BN009.
图13显示了EuTDA检测各NKG2D CAR-T细胞对肿瘤细胞的杀伤效果。Figure 13 shows the killing effect of each NKG2D CAR-T cell on tumor cells detected by EuTDA.
图14显示了ELISA检测各NKG2D CAR-T细胞的IFN-γ释放水平。Figure 14 shows the IFN-γ release level of each NKG2D CAR-T cell detected by ELISA.
具体实施方式Detailed ways
本发明人经过广泛而深入地研究,经过大量的筛选,首次将特定的CAR和CCR2b以及CD40L蛋白共表达工程化的免疫细胞,即将含NKG2D胞外结构域(ED)的CAR与CCR2b和CD40L联合表达于CAR-T等免疫细胞中。本发明的CAR-T细胞同时具备NKG2D的多靶点识别能力、CCR2b的增强趋化迁移能力和CD40L的免疫激活能力,并且出乎意料地表现出协同的针对肿瘤细胞的体外杀伤作用。在此基础上完成了本发明。After extensive and in-depth research and a large number of screenings, the inventors co-expressed specific CAR, CCR2b and CD40L proteins for the first time in engineered immune cells, that is, combining CAR containing NKG2D extracellular domain (ED) with CCR2b and CD40L Expressed in CAR-T and other immune cells. The CAR-T cells of the present invention simultaneously have the multi-target recognition ability of NKG2D, the enhanced chemotaxis and migration ability of CCR2b, and the immune activation ability of CD40L, and unexpectedly exhibit a synergistic killing effect on tumor cells in vitro. The present invention has been accomplished on this basis.
实验提示,与现有技术相比,本发明的免疫细胞既能通过CCR2b提高CAR-T细胞对CCL2、CCL7等趋化因子的灵敏度,向结直肠癌、卵巢癌、胰腺癌等实体瘤的病灶高效迁移,提高治疗效率;同时又能通过NKG2D CAR分子识别结直肠癌、 卵巢癌、胰腺癌等恶性肿瘤细胞表面的多种靶点抗原(包括MICA、MICB、ULBP1、ULBP2、ULBP3、ULBP4、ULBP5、ULBP6),降低因肿瘤异质性或靶点抗原丢失而导致疗效下降的风险。此外,共表达的CD40L可有效激活机体内源的天然和适应性免疫应答,从而有助于T细胞克服免疫抑制的肿瘤微环境,提高肿瘤治疗,降低肿瘤复发风险。Experiments suggest that, compared with the prior art, the immune cells of the present invention can improve the sensitivity of CAR-T cells to chemokines such as CCL2 and CCL7 through CCR2b, and promote tumor lesions of solid tumors such as colorectal cancer, ovarian cancer, and pancreatic cancer. Efficient migration to improve treatment efficiency; at the same time, NKG2D CAR molecules can recognize various target antigens on the surface of malignant tumor cells such as colorectal cancer, ovarian cancer, and pancreatic cancer (including MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5 , ULBP6), reducing the risk of decreased efficacy due to tumor heterogeneity or loss of target antigens. In addition, co-expressed CD40L can effectively activate the body's endogenous natural and adaptive immune responses, thereby helping T cells overcome the immunosuppressive tumor microenvironment, improve tumor therapy, and reduce the risk of tumor recurrence.
本发明以CAR-T细胞为例,代表性地对本发明的工程化免疫细胞进行详细说明。本发明的工程化免疫细胞不限于上下文所述的CAR-T细胞,本发明的工程化免疫细胞具有与上下文所述的CAR-T细胞相同或类似的技术特征和有益效果。具体地,当免疫细胞表达嵌合抗原受体CAR时,NK细胞等同于T细胞(或T细胞可替换为NK细胞)。The present invention takes CAR-T cells as an example to representatively describe the engineered immune cells of the present invention in detail. The engineered immune cells of the present invention are not limited to the CAR-T cells described above, and the engineered immune cells of the present invention have the same or similar technical features and beneficial effects as the CAR-T cells described above. Specifically, when immune cells express chimeric antigen receptor CAR, NK cells are equivalent to T cells (or T cells can be replaced by NK cells).
术语the term
为了可以更容易地理解本公开,首先定义某些术语。如本申请中所使用的,除非本文另有明确规定,否则以下术语中的每一个应具有下面给出的含义。In order that the present disclosure may be more readily understood, certain terms are first defined. As used in this application, unless expressly stated otherwise herein, each of the following terms shall have the meaning given below.
术语“约”可以是指在本领域普通技术人员确定的特定值或组成的可接受误差范围内的值或组成,其将部分地取决于如何测量或测定值或组成。The term "about" can refer to a value or composition within an acceptable error range for a particular value or composition as determined by one of ordinary skill in the art, which will depend in part on how the value or composition is measured or determined.
术语“给予”是指使用本领域技术人员已知的各种方法和递送系统中的任一种将本发明的产品物理引入受试者,包括静脉内、瘤内、肌内、皮下、腹膜内、脊髓或其它肠胃外给药途径,例如通过注射或输注。The term "administration" refers to the physical introduction of a product of the invention into a subject using any of a variety of methods and delivery systems known to those skilled in the art, including intravenous, intratumoral, intramuscular, subcutaneous, intraperitoneal , spinal or other parenteral routes of administration, for example by injection or infusion.
抗体Antibody
如本文所用,术语“抗体”(Ab)应包括但不限于免疫球蛋白,其特异性结合抗原并包含通过二硫键互连的至少两条重(H)链和两条轻(L)链,或其抗原结合部分。每条H链包含重链可变区(本文缩写为VH)和重链恒定区。重链恒定区包含三个恒定结构域CH1、CH2和CH3。每条轻链包含轻链可变区(本文缩写为VL)和轻链恒定区。轻链恒定区包含一个恒定结构域CL。VH和VL区可以进一步细分为称为互补决定区(CDR)的高变区,其散布有更保守的称为框架区(FR)的区域。每个VH和VL包含三个CDR和四个FR,从氨基末端到羧基末端按照以下顺序排列:FR1,CDR1,FR2,CDR2,FR3,CDR3,FR4。重链和轻链的可变区含有与抗原相互作用的结合结构域。As used herein, the term "antibody" (Ab) shall include, but not be limited to, an immunoglobulin that specifically binds an antigen and comprises at least two heavy (H) chains and two light (L) chains interconnected by disulfide bonds , or an antigen-binding portion thereof. Each H chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region comprises three constant domains CH1, CH2 and CH3. Each light chain comprises a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region comprises one constant domain, CL. The VH and VL regions can be further subdivided into hypervariable regions called complementarity determining regions (CDRs), interspersed with more conserved regions called framework regions (FRs). Each VH and VL contains three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain binding domains that interact with the antigen.
抗原结合结构域antigen binding domain
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') 2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)。单链抗体优选是由一条核苷酸链编码的一条氨基酸链序列。 As used herein, "antigen-binding domain" and "single-chain antibody fragment" all refer to a Fab fragment, Fab' fragment, F(ab') 2 fragment, or a single Fv fragment having 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 the structures required for antigen binding. The antigen binding domain is usually a scFv (single-chain variable fragment). A single-chain antibody is preferably a sequence of one amino acid chain encoded by one nucleotide chain.
在本发明中,所述scFv包含特异性识别肿瘤高表达的抗原的NKG2D胞外结构域或其活性片段。In the present invention, the scFv comprises an NKG2D extracellular domain or an active fragment thereof that specifically recognizes an antigen highly expressed by a tumor.
此外,本发明的免疫细胞还可含有额外的特异性识别肿瘤高表达的抗原的抗体,较佳地为单链抗体或Fv抗体。In addition, the immune cells of the present invention may also contain additional antibodies that specifically recognize antigens highly expressed in tumors, preferably single-chain antibodies or Fv antibodies.
嵌合抗原受体(CAR)Chimeric Antigen Receptor (CAR)
如本文所用,嵌合免疫抗原受体(Chimeric antigen receptor,CAR)包括细胞外结构域、任选的铰链区、跨膜结构域、和细胞内结构域。胞外结构域包括任选的信号肽和靶点特异性结合结构域(也称为抗原结合结构域)。细胞内结构域包括共刺激结构域和CD3ζ链部分。CAR在T细胞中表达时,胞外段可识别一个特异的抗原,随后通过胞内结构域转导该信号,引起细胞的活化增殖、细胞溶解毒性和分泌细胞因子如IL-2和IFN-γ等,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CD3ζ链中的一个或多个的细胞内结构域融合。优选地,抗原结合结构域与4-1BB信号传导结构域和CD3ζ信号结构域组合的细胞内结构域融合。As used herein, a Chimeric antigen receptor (CAR) includes an extracellular domain, an optional hinge region, a transmembrane domain, and an intracellular domain. The extracellular domain includes an optional signal peptide and a target-specific binding domain (also known as an antigen binding domain). The intracellular domain includes the co-stimulatory domain and the CD3ζ chain portion. When CAR is expressed in T cells, the extracellular segment can recognize a specific antigen, and then transduce the signal through the intracellular domain, causing cell activation and proliferation, cytolytic toxicity and secretion of cytokines such as IL-2 and IFN-γ etc., affecting tumor cells so that they do not grow, are induced to die, or are otherwise affected, and result 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 the co-stimulatory molecule and the CD3zeta chain. Preferably, the antigen binding domain is fused to the intracellular domain of the combination of the 4-1BB signaling domain and the CD3ζ signaling domain.
在一个实施方式中,本发明CAR靶向NKG2D配体,能与MICA、MICB、ULBP1、ULBP2、ULBP3、ULBP4、ULBP5、ULBP6特异性结合。In one embodiment, the CAR of the present invention targets NKG2D ligands and can specifically bind to MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5, and ULBP6.
嵌合抗原受体T细胞(CAR-T细胞)Chimeric Antigen Receptor T Cells (CAR-T Cells)
如本文所用,术语“CAR-T细胞”、“CAR-T”、“本发明CAR-T细胞”均指本发明第一方面所述的CAR-T细胞。本发明CAR-T细胞可用于治疗NKG2D配体高表达的肿瘤,如结直肠癌、卵巢癌、胰腺癌等。As used herein, the terms "CAR-T cell", "CAR-T" and "CAR-T cell of the present invention" all refer to the CAR-T cell described in the first aspect of the present invention. The CAR-T cells of the present invention can be used to treat tumors with high expression of NKG2D ligands, such as colorectal cancer, ovarian cancer, pancreatic cancer and the like.
CAR-T细胞较其它基于T细胞的治疗方式存在以下优势:(1)CAR-T细胞的作用过程不受MHC的限制;(2)鉴于很多肿瘤细胞表达相同的肿瘤抗原,针对某一种肿瘤抗原的CAR基因构建一旦完成,便可以被广泛利用;(3)CAR既可以利用肿瘤蛋白质抗原,又可利用糖脂类非蛋白质抗原,扩大了肿瘤抗原的靶点范围;(4)使用患者自体细胞降低了排异反应的风险;(5)CAR-T细胞具有免疫记忆功能,可以长期在体内存活。CAR-T cells have the following advantages over other T-cell-based therapies: (1) The action process of CAR-T cells is not restricted by MHC; (2) Since many tumor cells express the same tumor antigen, it can target a certain tumor Once the CAR gene construction of the antigen is completed, it can be widely used; (3) CAR can use both tumor protein antigens and glycolipid non-protein antigens, expanding the target range of tumor antigens; (4) using the patient's own The cells reduce the risk of rejection; (5) CAR-T cells have immune memory function and can survive in the body for a long time.
嵌合抗原受体NK细胞(CAR-NK细胞)Chimeric Antigen Receptor NK Cells (CAR-NK Cells)
如本文所用,术语“CAR-NK细胞”、“CAR-NK”、“本发明CAR-NK细胞”均指本发明第一方面所述的CAR-NK细胞。本发明CAR-NK细胞可用于治疗NKG2D 配体高表达的肿瘤,如结直肠癌、卵巢癌、胰腺癌等。As used herein, the terms "CAR-NK cells", "CAR-NK" and "CAR-NK cells of the present invention" all refer to the CAR-NK cells described in the first aspect of the present invention. The CAR-NK cells of the present invention can be used to treat tumors with high expression of NKG2D ligands, such as colorectal cancer, ovarian cancer, pancreatic cancer and the like.
自然杀伤(NK)细胞是一类主要的免疫效应细胞,通过非抗原特异性途径去保护机体免受病毒感染和肿瘤细胞的侵袭。通过工程化(基因修饰)的NK细胞可能获得新的功能,包括特异性识别肿瘤抗原的能力及具有增强的抗肿瘤细胞毒作用。Natural killer (NK) cells are a major type of immune effector cells, which protect the body from virus infection and tumor cell invasion through non-antigen-specific pathways. NK cells through engineering (gene modification) may obtain new functions, including the ability to specifically recognize tumor antigens and have enhanced anti-tumor cytotoxicity.
与自体CAR-T细胞相比,CAR-NK细胞还具有以下优点,例如:(1)通过释放穿孔素和颗粒酶直接杀伤肿瘤细胞,而对机体正常的细胞没有杀伤作用;(2)它们释放很少量的细胞因子从而降低了细胞因子风暴的危险;(3)体外极易扩增及发展为“现成的”产品。除此之外,与CAR-T细胞治疗类似。Compared with autologous CAR-T cells, CAR-NK cells also have the following advantages, for example: (1) directly kill tumor cells by releasing perforin and granzymes, but have no killing effect on normal cells of the body; (2) they release A very small amount of cytokines reduces the risk of cytokine storm; (3) It is very easy to expand in vitro and develop into "off-the-shelf" products. Other than that, it is similar to CAR-T cell therapy.
蛋白NKG2Dprotein NKG2D
在本发明中,NKG2D包括野生型或其突变型或其衍生形式或其活性片段。优选的NKG2D来自于哺乳动物(如人和非人灵长动物)的NKG2D。In the present invention, NKG2D includes wild type or its mutant or its derivative form or its active fragment. Preferred NKG2Ds are NKG2Ds from mammals such as humans and non-human primates.
人NKG2D蛋白的氨基酸序列的登录号为NP_031386,核苷酸氨基酸序列的登录号为NM_007360。人NKG2D的全长氨基酸序列如下所示:The accession number of the amino acid sequence of the human NKG2D protein is NP_031386, and the accession number of the nucleotide amino acid sequence is NM_007360. The full-length amino acid sequence of human NKG2D is as follows:
Figure PCTCN2022130696-appb-000001
Figure PCTCN2022130696-appb-000001
其中,第1-51位是胞内结构域;第52-72位是跨膜区;第73-216位是NKG2D胞外结构域(下划线部分)。Among them, positions 1-51 are intracellular domains; positions 52-72 are transmembrane regions; positions 73-216 are NKG2D extracellular domains (underlined).
趋化因子Chemokine
趋化因子是一类特殊的细胞因子,包含50多个成员。根据结构分为CC、CXC、CX3C和XC四类;趋化因子受体则相应分为CCR、CXCR、CX3CR和XCR4种,约有20个成员。Chemokines are a special class of cytokines, including more than 50 members. According to the structure, it is divided into four types: CC, CXC, CX3C and XC; chemokine receptors are correspondingly divided into four types: CCR, CXCR, CX3CR and XCR, with about 20 members.
在本发明的工程化免疫细胞中,表达的趋化因子受体是CCR2b蛋白,可以结合的趋化因子包括CCL2、CCL7、CCL8、CCL12、CCL13、CCL16等。In the engineered immune cells of the present invention, the expressed chemokine receptor is CCR2b protein, and the chemokines that can be bound include CCL2, CCL7, CCL8, CCL12, CCL13, CCL16 and the like.
CCR2b蛋白的氨基酸序列的登录号为NP_001116868.1,核苷酸氨基酸序列的登录号为NM_001123396.4。具体序列如下所示:The accession number of the amino acid sequence of the CCR2b protein is NP_001116868.1, and the accession number of the nucleotide amino acid sequence is NM_001123396.4. The specific sequence is as follows:
氨基酸序列:Amino acid sequence:
Figure PCTCN2022130696-appb-000002
Figure PCTCN2022130696-appb-000002
Figure PCTCN2022130696-appb-000003
Figure PCTCN2022130696-appb-000003
蛋白CD40Lprotein CD40L
白细胞分化抗原40配体(cluster of differentiation 40 ligand,CD40L),也称CD154或肿瘤坏死因子相关激活蛋白(tumornecrosis factor-associated activationprotein,TRAP)。Leukocyte differentiation antigen 40 ligand (cluster of differentiation 40 ligand, CD40L), also known as CD154 or tumor necrosis factor-associated activation protein (tumor necrosis factor-associated activation protein, TRAP).
人CD40L蛋白的氨基酸序列的登录号为NP_000065.1,核苷酸氨基酸序列的登录号为NM_000074.3。人CD40L的全长氨基酸序列如下所示:The accession number of the amino acid sequence of human CD40L protein is NP_000065.1, and the accession number of the nucleotide amino acid sequence is NM_000074.3. The full-length amino acid sequence of human CD40L is as follows:
Figure PCTCN2022130696-appb-000004
Figure PCTCN2022130696-appb-000004
在本发明中,合适的CD40L包括野生型和突变型的CD40L,只要该突变型CD40L具有野生型CD40L的基本功能。此外,在本发明中,优选的CD40L来自哺乳动物,如人和非人哺乳动物。In the present invention, suitable CD40L includes wild-type and mutant-type CD40L, as long as the mutant-type CD40L has basic functions of wild-type CD40L. Furthermore, in the present invention, preferred CD40L is from mammals, such as humans and non-human mammals.
CD40L主要表达于活化的CD4+T淋巴细胞、活化的CD8 +T细胞、嗜碱性粒细胞、肥大细胞和NK细胞。CD40L与其受体CD40是体内炎症和免疫反应系统中的一对共同刺激分子。在天然免疫中,CD40L/CD40共刺激途径是单核细胞成熟过程的重要触发因素,主要驱动单核细胞分化为M1谱系的巨噬细胞和DC细胞。同时,该途径也可促进DC细胞释放细胞因子和趋化因子,诱导其他共刺激分子的表达,并促进抗原的交叉呈递。在体液免疫中,该途径也参与T细胞依赖性的B淋巴细胞应答过程、生发中心的形成、长期记忆性B细胞的产生、抗体的产生及抗体类别转换。在细胞免疫中,该途径能促进T细胞活化并放大T细胞介导的免疫应答,在CD4 +T细胞分化的过程中起重要作用,也能够促进CD8+T细胞的扩增和多能性,是产生记忆性CD8+T细胞的基础。 CD40L is mainly expressed in activated CD4+T lymphocytes, activated CD8 + T cells, basophils, mast cells and NK cells. CD40L and its receptor CD40 are a pair of co-stimulatory molecules in the inflammatory and immune response systems in vivo. In innate immunity, the CD40L/CD40 co-stimulatory pathway is an important trigger for the maturation process of monocytes, which mainly drives the differentiation of monocytes into M1 lineage macrophages and DC cells. At the same time, this pathway can also promote the release of cytokines and chemokines from DC cells, induce the expression of other co-stimulatory molecules, and promote the cross-presentation of antigens. In humoral immunity, this pathway is also involved in the T cell-dependent B lymphocyte response process, the formation of germinal centers, the generation of long-term memory B cells, antibody production, and antibody class switching. In cellular immunity, this pathway can promote T cell activation and amplify T cell-mediated immune response, play an important role in the process of CD4 + T cell differentiation, and can also promote the expansion and pluripotency of CD8 + T cells, It is the basis for generating memory CD8+ T cells.
在抗肿瘤免疫反应中,CD40L/CD40共刺激途径也发挥多种作用,如激活T细胞的增殖和细胞因子的释放,诱导M2谱系的巨噬细胞向具有抗肿瘤活性的M1谱系的巨噬细胞转变等。在某些靶点抗原丢失但CD40高表达的肿瘤中,该途径还可以介导T细胞对肿瘤细胞的杀伤作用。In the anti-tumor immune response, the CD40L/CD40 co-stimulatory pathway also plays multiple roles, such as activating the proliferation of T cells and the release of cytokines, and inducing the M2 lineage macrophages to M1 lineage macrophages with anti-tumor activity. shift etc. In tumors with loss of some target antigens but high expression of CD40, this pathway can also mediate the killing effect of T cells on tumor cells.
在本发明中,当与特定的CAR分子和CCR2b共表达CD40L时,CD40L作为CAR-T细胞的辅助因子,可以在CAR-T细胞杀伤肿瘤细胞的同时,激活机体内源的免疫应答,增强和延长治疗效果。此外,出乎意料的是,CD40L还可与CCR2b一起发挥协同作用,从而协同地显著提高针对肿瘤细胞的体外杀伤作用。In the present invention, when CD40L is co-expressed with a specific CAR molecule and CCR2b, CD40L, as an auxiliary factor of CAR-T cells, can activate the endogenous immune response of the body while CAR-T cells kill tumor cells, enhance and Prolongs the healing effect. In addition, unexpectedly, CD40L can also play a synergistic effect with CCR2b, thereby synergistically significantly improving the killing effect against tumor cells in vitro.
表达盒expression cassette
如本文所用,“表达盒”或“本发明表达盒”包括第一表达盒、第二表达盒和第三表达盒。本发明表达盒如本发明第五方面所述,第一表达盒包含编码所述CAR的核酸序列。所述第二表达盒表达外源的CCR2b蛋白。所述第二表达盒表达外源的CD40L蛋白。As used herein, an "expression cassette" or "expression cassette of the invention" includes a first expression cassette, a second expression cassette and a third expression cassette. The expression cassette of the present invention is as described in the fifth aspect of the present invention, the first expression cassette comprises the nucleic acid sequence encoding the CAR. The second expression cassette expresses exogenous CCR2b protein. The second expression cassette expresses exogenous CD40L protein.
在本发明中,CCR2b和CD40L蛋白可以是组成型表达或诱导型表达。In the present invention, CCR2b and CD40L proteins can be expressed constitutively or inducibly.
在诱导表达情况下,在所述CAR-T细胞被相应诱导剂激活时,第二表达盒表达CCR2b蛋白,第三表达盒表达CD40L蛋白;这样,在本发明CAR-T细胞在未接触相应诱导剂时,第二表达盒不表达CCR2b蛋白,第三表达盒不表达CD40L蛋白。In the case of induced expression, when the CAR-T cells are activated by the corresponding inducer, the second expression cassette expresses the CCR2b protein, and the third expression cassette expresses the CD40L protein; thus, when the CAR-T cells of the present invention are not exposed to the corresponding induced When the agent is used, the second expression cassette does not express the CCR2b protein, and the third expression cassette does not express the CD40L protein.
在一个实施方式中,所述第一表达盒、第二表达盒和/或第三表达盒分别还包括启动子和/或终止子。第二表达盒和第三表达盒的启动子可以为组成型或诱导型启动子。In one embodiment, the first expression cassette, the second expression cassette and/or the third expression cassette further comprise a promoter and/or a terminator, respectively. The promoters of the second and third expression cassettes may be constitutive or inducible promoters.
载体carrier
本发明还提供了含有本发明表达盒的载体。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、稳定地整合于细胞基因组中并随子细胞基因组的复制而复制。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,且具有低免疫原性的优点。The present invention also provides a vector comprising the expression cassette of the present invention. 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 in the genome of the cell and replication of the genome of the daughter cell. Lentiviral vectors have advantages over vectors derived from oncogenic retroviruses such as murine leukemia virus in that they can transduce non-proliferating cells and have the advantage of low immunogenicity.
通常,可通过常规操作将本发明的表达盒或核酸序列连接至启动子下游,并将其并入表达载体。该载体可整合至真核细胞基因组中并随之复制。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。Usually, the expression cassette or nucleic acid sequence of the present invention can be connected downstream of the promoter through routine operations, and incorporated into an expression vector. The vector can be integrated into the genome of eukaryotic cells and then replicated. 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.
本发明的表达载体也可用于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已知的。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。The expression vectors of the invention can also be used in standard gene delivery protocols for nucleic acid immunization and gene therapy. 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.
所述表达盒或核酸序列可被克隆入许多类型的载体。例如,该表达盒或核酸序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、复制载体等。The expression cassette or nucleic acid sequence can be cloned into many types of vectors. For example, the expression cassette or nucleic acid sequence can be cloned into such vectors including, but not limited to, plasmids, phagemids, phage derivatives, animal viruses, and cosmids. Particular vectors of interest include expression vectors, replication vectors, and the like.
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Molecular Cloning:A Laboratory Manual(Sambrook等,Cold Spring Harbor Laboratory,New York,2001)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、腺相关病毒、疱疹病毒和慢病毒。通常,合适的载体包含至少一种在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,WO01/96584;WO01/29058;和美国专利号6,326,193)。Furthermore, expression vectors can be provided to cells in the form of viral vectors. Viral vector technology is well known in the art and described, for example, in Molecular Cloning: A Laboratory Manual (Sambrook et al., Cold Spring Harbor Laboratory, New York, 2001) and other virology and molecular biology manuals. 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 at least one origin of replication functional in the organism, a promoter sequence, convenient restriction enzyme sites, and one or more selectable markers (e.g., WO01/96584; WO01/29058; and US Patent No. 6,326,193).
已经有许多基于病毒的系统被开发出来,并用于哺乳动物细胞的基因转导。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。该重组病毒随后可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。在一个实施方式中,使用慢病毒载体。许多DNA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。A number of virus-based systems have been developed and used for gene transduction in 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 one embodiment, lentiviral vectors are used. Many DNA viral systems are known in the art. In some embodiments, an adenoviral vector is used. Many adenoviral vectors are known in the art.
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,这些元件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元件被倒置或移动时,保持启动子功能。在胸苷激酶(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。Additional promoter elements, such as enhancers, can regulate the frequency of transcription initiation. Typically, these elements are located in a region of 30-110 bp 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 an element is inverted or moved relative to another element. In the thymidine kinase (tk) promoter, the spacing between promoter elements can be increased by 50 bp apart before activity begins to decline. Depending on the promoter, it appears that individual elements can act cooperatively or independently to initiate transcription.
合适的启动子的一个例子为巨细胞病毒(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苷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-1α(EF-1α)。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-巴尔病毒(Epstein-Barr virus,EBV)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,能够在需要时,启动连接诱导型启动子的多核苷酸序列的表达,或在不需要时关闭表达。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。An example of a suitable promoter is the cytomegalovirus (CMV) promoter sequence. The promoter sequence is a strong constitutive promoter sequence capable of driving high level expression of any polynucleotide sequence operably linked thereto. Another example of a suitable promoter is elongation growth factor-1 alpha (EF-1 alpha). However, other constitutive promoter sequences can also be used, including but not limited to the simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, avian leukemia virus promoter, Epstein-Barr virus (Epstein-Barr virus, EBV) immediate early promoter, Ruth's sarcoma virus promoter, and human gene promoters, such as but not limited to protein 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 the polynucleotide sequence linked to the inducible promoter when desired, or turning off expression when 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.
被引入细胞的表达载体也可包含可选择的标记基因或报告基因中的任一个或两者,以便于通过病毒载体从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。可选择的标记基因和报告基因两者的侧翼都可具有适当的调节序列,以便能够在宿主细胞中表达。有用的可选择标记基因包括例如抗生素抗性基因,诸如neomycin等等。The expression vector introduced into the cells may also contain either or both of a selectable marker gene or a reporter gene to facilitate the identification and selection of expressing cells from the transfected or infected cell population by the viral vector. In other aspects, selectable markers can be carried on a single piece of DNA and used in a co-transfection procedure. Both the selectable marker gene and the reporter gene may be flanked by appropriate regulatory sequences to enable expression in the host cell. Useful selectable marker genes include, for example, antibiotic resistance genes such as neomycin and the like.
将基因引入细胞和将基因表达入细胞的方法在本领域中是已知的。在表达载体的内容中,载体可通过在本领域中的任何方法容易地引入宿主细胞,例如,哺乳动物(如人T细胞)、细菌、酵母或昆虫细胞。例如,表达载体可通过物理、化学或生物学手段转移入宿主细胞。Methods of introducing genes into cells and expressing genes into cells are known in the art. In the context of an expression vector, the vector can be easily introduced into host cells, eg, mammalian (eg, human T cells), bacterial, yeast or insect cells, by any method in the art. For example, expression vectors can be transferred into host cells by physical, chemical or biological means.
将多核苷酸引入宿主细胞的物理方法包括磷酸钙沉淀、阳离子复合物转染法、脂质转染法、粒子轰击、微注射、电穿孔等等。生产包括载体和/或外源核酸的细胞的方法在本领域中是公知的。见例如Molecular Cloning:A Laboratory Manual(Sambrook等,Cold Spring Harbor Laboratory,New York,2001)。将多核苷酸引入宿主细胞的优选方法为脂质体法转染法和阳离子复合物聚乙烯亚胺转染法。Physical methods for introducing polynucleotides into host cells include calcium phosphate precipitation, cationic complex transfection, lipofection, particle bombardment, microinjection, electroporation, and the like. Methods for producing cells comprising vectors and/or exogenous nucleic acids are well known in the art. See, e.g., Molecular Cloning: A Laboratory Manual (Sambrook et al., Cold Spring Harbor Laboratory, New York, 2001). Preferred methods for introducing polynucleotides into host cells are liposome transfection and cationic complex polyethylenimine transfection.
将多核苷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,已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。例如见美国专利号5,350,674和5,585,362。Biological methods for introducing polynucleotides into host cells include the use of DNA and RNA vectors. Viral vectors, especially retroviral vectors, have become the most widely used method of 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, US Patent Nos. 5,350,674 and 5,585,362.
将多核苷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(delivery vehicle)的示例性胶体系统为脂质体(例如,人造膜囊)。Chemical means of introducing polynucleotides into host cells include colloidal dispersion systems, such as macromolecular complexes, nanocapsules, microspheres, beads; and lipid-based systems, including oil-in-water emulsions, micelles, mixed micelles, and lipid-based systems. plastid. An exemplary colloidal system for use as a delivery vehicle in vitro and in vivo is a liposome (eg, an artificial membrane vesicle).
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,经与脂质体和寡核苷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。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。脂质为脂类物质,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烃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。Where a non-viral delivery system is used, an exemplary delivery vehicle is liposomes. Consider the use of lipid formulations to introduce 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 To liposomes, entrapped in liposomes, complexed with liposomes, dispersed in lipid-containing solutions, mixed with lipids, associated with lipids, contained in lipids as a suspension, contained in micelles or Complexes 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. They may also simply be dispersed in solution, possibly forming aggregates of non-uniform size or shape. Lipids are lipid 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 hydrocarbons and their derivatives such as fatty acids, alcohols, amines, aminoalcohols, and aldehydes.
在本发明的一个优选的实施方式中,所述载体为慢病毒载体。In a preferred embodiment of the present invention, the vector is a lentiviral vector.
应理解,在本发明中,除了采用多个慢病毒进行转导,还用直接转染mRNA或质粒,或者通过表达人工转录因子等方法,从而在T细胞等免疫细胞中联合表达CCR2b、CD40L和NKG2D CAR分子。It should be understood that in the present invention, in addition to using multiple lentiviruses for transduction, direct transfection of mRNA or plasmids, or expression of artificial transcription factors, etc., can jointly express CCR2b, CD40L and T cells in immune cells such as T cells. NKG2D CAR molecule.
制剂preparation
本发明提供了一种含有本发明第一方面所述的工程化免疫细胞(如CAR-T细胞),以及药学上可接受的载体、稀释剂或赋形剂。在一个实施方式中,所述制剂为液态制剂。优选地,所述制剂为注射剂。优选地,所述制剂中所述CAR-T细胞的浓度为1×10 3-1×10 8个细胞/ml,更优地1×10 4-1×10 7个细胞/ml。 The present invention provides an engineered immune cell (such as CAR-T cell) according to the first aspect of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient. In one embodiment, the formulation is a liquid formulation. Preferably, the preparation is an injection. Preferably, the concentration of the CAR-T cells in the preparation is 1×10 3 -1×10 8 cells/ml, more preferably 1×10 4 -1×10 7 cells/ml.
在一个实施方式中,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。In one embodiment, the formulation 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; chelating agents such as EDTA or glutathione; adjuvants (eg, aluminum hydroxide); and preservatives. The formulations of the invention are preferably formulated for intravenous administration.
治疗性应用therapeutic application
本发明包括含本发明表达盒的载体(如慢病毒载体)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表面标志物并表达CCR2b蛋白,协同而显著地提高其对肿瘤细胞的杀伤效率。The invention includes the therapeutic use of cells (eg, T cells) transduced with a vector (eg, a lentiviral vector) comprising an expression cassette of the invention. The transduced T cells can target the surface markers of tumor cells and express CCR2b protein, synergistically and significantly improving their killing efficiency against tumor cells.
因此,本发明也提供了刺激靶向哺乳动物肿瘤细胞群或组织的T细胞所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CAR-T细胞。Therefore, the present invention also provides a method for stimulating an immune response mediated by T cells targeting mammalian tumor cell populations or tissues, comprising the following steps: administering the CAR-T cells of the present invention to mammals.
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。这种方式使移植物抗宿主反应的发生概率极低,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,产生可导致持续控制肿瘤的长期持久性。In one embodiment, the present invention includes a type of cell therapy, in which a patient's own T cells (or a heterologous 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 graft-versus-host reaction is extremely low, and the antigen is recognized by T cells without MHC restriction. Furthermore, a single CAR-T can treat all cancers that express that 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.
在一个实施方式中,本发明的CAR-T细胞可经历稳定的体内扩增并可持续数月至数年的时间。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-T细胞可诱导对CAR抗原结合结构域所识别的抗原的高表达肿瘤细胞的特异性免疫应答。例如,本发明的CAR-T细胞引起针对NKG2D配体高表达的肿瘤细胞的特异性免疫应答。In one embodiment, the CAR-T cells of the present invention can undergo stable in vivo expansion and last for several months to several years. Alternatively, the CAR-mediated immune response can be part of an adoptive immunotherapy step in which CAR-T cells can induce a specific immune response to tumor cells that overexpress the antigen recognized by the CAR antigen-binding domain. For example, the CAR-T cells of the present invention elicit a specific immune response against tumor cells with high expression of NKG2D ligands.
可治疗的癌症包括没有被血管化或基本上还没有被血管化的肿瘤,以及血管化的肿瘤。用本发明的CAR治疗的癌症类型包括但不限于:结直肠癌、卵巢癌和胰腺癌。Treatable cancers include tumors that are not or substantially not vascularized, as well as vascularized tumors. Cancer types treated with the CAR of the present invention include, but are not limited to: colorectal cancer, ovarian cancer, and pancreatic cancer.
通常地,如本文所述活化和扩增的细胞可用于治疗和预防肿瘤等疾病。因此,本发明提供了治疗癌症的方法,其包括施用给需要其的对象治疗有效量的本发明的CAR-T细胞。Generally, cells activated and expanded as described herein can be used for the treatment and prevention of diseases such as tumors. Accordingly, the present invention provides a method of treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of a CAR-T cell of the present invention.
本发明的CAR-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。The CAR-T cells of the present 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 compositions of the present invention may comprise a target cell population as described herein, in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients.
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如患者的病症、和患者疾病的类型和严重度等因素确定,或可由临床试验确定。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 factors such as the patient's condition, and the type and severity of the patient's disease, or may be determined by clinical trials.
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。包括本文描述的T细胞的药物组合物可以以10 4至10 9个细胞/kg体重的剂量,优选10 5至10 7个细胞/kg体重的剂量(包括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.ofMed.319:1676,1988)施用。对于具体患者的最佳剂量和治疗方案可由医学领域技术人员通过监测患者的疾病迹象容易地确定,并以此调整治疗。 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 (subject ) with individual differences in age, weight, tumor size, degree of infection or metastasis, and disease. Pharmaceutical compositions comprising T cells described herein may be administered at a dose of 10 4 to 10 9 cells/kg body weight, preferably at a dose of 10 5 to 10 7 cells/kg body weight (including all integer values within the range). 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.
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静脉内注射施用。T细胞的组合物可被直接注入肿瘤,淋巴结或感染位置。Administration of the compositions to a subject may be by any convenient means, 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 injection or intraperitoneally. In one embodiment, the T cell composition of the invention is administered to a patient by intradermal or subcutaneous injection. In another embodiment, the T cell composition of the invention is preferably administered by intravenous injection. Compositions of T cells can be injected directly into tumors, lymph nodes or sites of infection.
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、阿糖胞苷(也已知为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,环孢菌素、硫唑嘌呤、甲氨喋呤、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。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 (e.g., previously , simultaneously or subsequently) to the patient in a form of treatment including but not limited to treatment with agents such as antiviral therapy, cidofovir and interleukin-2, cytarabine (also known as ARA-C) or natalizumab treatment for MS patients or erfatizumab treatment for psoriasis patients or other treatments for PML patients. In a further embodiment, the T cells of the invention may be used in combination with chemotherapy, radiation, immunosuppressants such as cyclosporine, azathioprine, methotrexate, mycophenolate mofetil and FK506, antibodies or other immunotherapeutic agents. In a further embodiment, the cell composition of the invention is administered 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.
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1×10 5个至1×10 10个本发明经修饰的T细胞,通过例如静脉回输的方式,施用于患者。 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, 1×10 5 to 1×10 10 modified T cells of the present invention can be administered to the patient for each treatment or each course of treatment, for example, through intravenous infusion.
本发明的主要优点Main advantages of the invention
(1)本发明利用CCR2b蛋白使得本发明的免疫细胞更高效地迁移到肿瘤部位,从而显著提高抑制肿瘤的效果,并可减少毒副作用。实验表明,本发明显著提高了CAR-T细胞向CCL2浓度高处迁移(如病灶部位)的能力。(1) The present invention uses the CCR2b protein to enable the immune cells of the present invention to migrate to the tumor site more efficiently, thereby significantly improving the effect of inhibiting tumors and reducing toxic and side effects. Experiments show that the present invention significantly improves the ability of CAR-T cells to migrate to places with high CCL2 concentration (such as lesion sites).
(2)本发明的工程化免疫细胞的抗原结合域采用NKG2D的胞外结合域,可通 过NKG2D CAR分子识别恶性肿瘤(如结直肠癌细胞、卵巢癌、胰腺癌等)细胞表面的8种靶点抗原(MICA、MICB、ULBP1、ULBP2、ULBP3、ULBP4、ULBP5、ULBP6),降低因肿瘤异质性或靶点抗原丢失而导致疗效下降的风险。(2) The antigen binding domain of the engineered immune cells of the present invention adopts the extracellular binding domain of NKG2D, which can recognize 8 kinds of targets on the cell surface of malignant tumors (such as colorectal cancer cells, ovarian cancer, pancreatic cancer, etc.) through NKG2D CAR molecules. Spot antigens (MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5, ULBP6), reduce the risk of decreased efficacy due to tumor heterogeneity or loss of target antigens.
(3)本发明采用CD40L作为第二辅助因子,以便更有效地激活机体内源的天然和适应性免疫应答。通过CD40L激活T细胞的增殖和细胞因子的释放,诱导M2谱系的巨噬细胞向具有抗肿瘤活性的M1谱系的巨噬细胞转变,增强DC细胞的抗原呈递功能等,同时提高T细胞对某些抗原丢失但CD40高表达的肿瘤细胞的杀伤作用。(3) The present invention uses CD40L as the second cofactor to more effectively activate the body's endogenous natural and adaptive immune responses. CD40L activates the proliferation of T cells and the release of cytokines, induces the transformation of M2 lineage macrophages into M1 lineage macrophages with anti-tumor activity, enhances the antigen presentation function of DC cells, etc., and at the same time improves the ability of T cells to certain Killing effect of tumor cells with antigen loss but high CD40 expression.
(4)NKG2D胞外域的CAR分子与CCR2b及CD40L同时联合时,可出乎意料地显著提高了CAR-T细胞向高浓度CCL2的趋化迁移能力。(4) When the CAR molecule of the NKG2D extracellular domain is combined with CCR2b and CD40L at the same time, it can unexpectedly significantly improve the chemotactic migration ability of CAR-T cells to high concentrations of CCL2.
(5)出乎意料地,当本发明NKG2D CAR分子、外源CCR2b蛋白和外源CD40L蛋白的联合表达时,可协同地显著提高针对肿瘤细胞的体外杀伤作用。(5) Unexpectedly, when the NKG2D CAR molecule of the present invention, exogenous CCR2b protein and exogenous CD40L protein are jointly expressed, the in vitro killing effect on tumor cells can be synergistically and significantly improved.
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《分子克隆:实验室手册》(Sambrook等人,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。Below in conjunction with specific embodiment, further illustrate the present invention. It should be understood that these examples are only used to illustrate the present invention, not 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 the conditions described in "Molecular Cloning: A Laboratory Manual" (Sambrook et al., New York: Cold Spring Harbor Laboratory Press, 1989), or Follow the conditions recommended by the manufacturer. Percentages and parts are by weight unless otherwise indicated.
除非另外说明,实施例中采用的试剂和材料为市售获得。Reagents and materials used in the examples were obtained commercially unless otherwise stated.
材料与方法Materials and Methods
CAR分子及其结构CAR molecule and its structure
在实施例中,各NKG2D CAR包含以下部分结构:人CD8信号肽[简称CD8(SP)]、人NKG2D胞外结构域[简称NKG2D(ED)]、优化了的人CD8铰链区[简称CD8(hinge)]、人CD8跨膜结构域[简称CD8(TM)]、人4-1BB胞内结构域[简称4-1BB(ID)]、人CD3ζ胞内信号转导结构域[简称CD3ζ(ID)]、自剪切肽P2A、人CCR2b、自剪切肽T2A、人CD40L。In the embodiments, each NKG2D CAR comprises the following partial structures: human CD8 signal peptide [abbreviated as CD8 (SP)], human NKG2D extracellular domain [abbreviated as NKG2D (ED)], optimized human CD8 hinge region [abbreviated as CD8 ( hinge)], human CD8 transmembrane domain [abbreviated as CD8(TM)], human 4-1BB intracellular domain [abbreviated as 4-1BB(ID)], human CD3ζ intracellular signal transduction domain [abbreviated as CD3ζ(ID) )], self-cleaving peptide P2A, human CCR2b, self-cleaving peptide T2A, human CD40L.
作为对照的第二代NKG2D CAR分子命名为BN001,联合表达CCR2的新一代NKG2D CAR分子命名为BN003,联合表达CD40L的新一代NKG2D CAR分子命名为BN004,联合表达CCR2和CD40L的新一代NKG2D CAR分子命名为BN009。The second-generation NKG2D CAR molecule used as a control was named BN001, the new-generation NKG2D CAR molecule jointly expressing CCR2 was named BN003, the new-generation NKG2D CAR molecule jointly expressing CD40L was named BN004, and the new-generation NKG2D CAR molecule jointly expressing CCR2 and CD40L was named BN004. Named BN009.
所述CAR分子的具体结构如图2所示,具体如下:The specific structure of the CAR molecule is shown in Figure 2, specifically as follows:
BN001从其氨基端至羧基端由CD8(SP)、NKG2D(ED)、CD8(hinge)、CD8(TM)、4-1BB(ID)、CD3ζ(ID)依次串联组成。 BN001 is composed of CD8 (SP), NKG2D (ED), CD8 (hinge), CD8 (TM), 4-1BB (ID) and CD3ζ (ID) in series from its amino terminal to carboxyl terminal.
BN003从其氨基端至羧基端由CD8(SP)、NKG2D(ED)、CD8(hinge)、CD8(TM)、 4-1BB(ID)、CD3ζ(ID)、P2A、CCR2b依次串联组成。 BN003 consists of CD8 (SP), NKG2D (ED), CD8 (hinge), CD8 (TM), 4-1BB (ID), CD3ζ (ID), P2A, and CCR2b in series from its amino terminal to carboxyl terminal.
BN004从其氨基端至羧基端由CD8(SP)、NKG2D(ED)、CD8(hinge)、CD8(TM)、4-1BB(ID)、CD3ζ(ID)、P2A、CD40L依次串联组成。 BN004 consists of CD8 (SP), NKG2D (ED), CD8 (hinge), CD8 (TM), 4-1BB (ID), CD3ζ (ID), P2A, and CD40L in series from its amino terminal to carboxyl terminal.
BN009从其氨基端至羧基端由CD8(SP)、NKG2D(ED)、CD8(hinge)、CD8(TM)、4-1BB(ID)、CD3ζ(ID)、P2A、CCR2b、T2A、CD40L依次串联组成。 CD8(SP), NKG2D(ED), CD8(hinge), CD8(TM), 4-1BB(ID), CD3ζ(ID), P2A, CCR2b, T2A, CD40L are sequentially connected in series from the amino terminal to the carboxyl terminal of BN009 composition.
SEQ ID NO:1(人NKG2D氨基酸序列)SEQ ID NO: 1 (human NKG2D amino acid sequence)
Figure PCTCN2022130696-appb-000005
Figure PCTCN2022130696-appb-000005
SEQ ID NO:2(人CCR2b氨基酸序列)SEQ ID NO: 2 (human CCR2b amino acid sequence)
Figure PCTCN2022130696-appb-000006
Figure PCTCN2022130696-appb-000006
SEQ ID No:3(人CD8信号肽氨基酸序列)SEQ ID No: 3 (amino acid sequence of human CD8 signal peptide)
Figure PCTCN2022130696-appb-000007
Figure PCTCN2022130696-appb-000007
SEQ ID No:4(人NKG2D胞外结构域氨基酸序列)SEQ ID No: 4 (amino acid sequence of human NKG2D extracellular domain)
Figure PCTCN2022130696-appb-000008
Figure PCTCN2022130696-appb-000008
SEQ ID No:5(人CD40L氨基酸序列)SEQ ID No:5 (human CD40L amino acid sequence)
Figure PCTCN2022130696-appb-000009
Figure PCTCN2022130696-appb-000009
Figure PCTCN2022130696-appb-000010
Figure PCTCN2022130696-appb-000010
SEQ ID No:6(优化了的人CD8铰链区氨基酸序列)SEQ ID No:6 (optimized human CD8 hinge region amino acid sequence)
Figure PCTCN2022130696-appb-000011
Figure PCTCN2022130696-appb-000011
SEQ ID No:7(人CD8跨膜结构域氨基酸序列)SEQ ID No: 7 (human CD8 transmembrane domain amino acid sequence)
Figure PCTCN2022130696-appb-000012
Figure PCTCN2022130696-appb-000012
SEQ ID No:8(人4-1BB胞内结构域氨基酸序列)SEQ ID No: 8 (amino acid sequence of human 4-1BB intracellular domain)
Figure PCTCN2022130696-appb-000013
Figure PCTCN2022130696-appb-000013
SEQ ID No:9(人CD3ζ胞内信号转导结构域氨基酸序列)SEQ ID No:9 (amino acid sequence of human CD3ζ intracellular signal transduction domain)
Figure PCTCN2022130696-appb-000014
Figure PCTCN2022130696-appb-000014
SEQ ID No:10(自剪切肽P2A氨基酸序列)SEQ ID No:10 (amino acid sequence of self-cleaving peptide P2A)
Figure PCTCN2022130696-appb-000015
Figure PCTCN2022130696-appb-000015
SEQ ID No:11(自剪切肽T2A氨基酸序列)SEQ ID No: 11 (amino acid sequence of self-cleaving peptide T2A)
Figure PCTCN2022130696-appb-000016
Figure PCTCN2022130696-appb-000016
实施例1慢病毒制备Example 1 lentivirus preparation
1.1慢病毒载体质粒的获得1.1 Obtaining lentiviral vector plasmids
全基因合成BN001、BN003、BN004和BN009的核苷酸序列,再通过分子克隆的方式连接到慢病毒载体pCDH-EF1-MCS-T2A-copGFP质粒中,使之在人EF1α启动子和Kozak序列的调控下表达。The nucleotide sequences of BN001, BN003, BN004, and BN009 were synthesized from the whole gene, and then connected to the lentiviral vector pCDH-EF1-MCS-T2A-copGFP plasmid by molecular cloning, so that it was in the human EF1α promoter and Kozak sequence expression under control.
1.2慢病毒载体质粒转染293T细胞1.2 Transfection of lentiviral vector plasmid into 293T cells
将上述各慢病毒载体质粒与慢病毒包装质粒pRSV-Rev、pMDLg/pRRE和pMD2.G用聚乙烯亚胺转染试剂混合,共转染293T细胞。培养48h后,分别收集病毒上清液,于4℃下4500rpm离心10~15min,经0.5μm孔径的滤膜过滤后用中空纤维柱超滤系统进行慢病毒浓缩,再用层析法进行慢病毒纯化,最后用0.22μm孔径的滤膜过滤除菌后分装置于-80℃保存。Mix the above-mentioned lentiviral vector plasmids with lentiviral packaging plasmids pRSV-Rev, pMDLg/pRRE and pMD2.G with polyethyleneimine transfection reagent, and co-transfect 293T cells. After culturing for 48 hours, collect the virus supernatant, centrifuge at 4,500 rpm for 10-15 minutes at 4°C, filter through a filter membrane with a pore size of 0.5 μm, and then use a hollow fiber column ultrafiltration system to concentrate the lentivirus, and then use chromatography to extract the lentivirus. Purify, and finally filter and sterilize with a filter membrane with a pore size of 0.22 μm, and store in -80°C.
1.3慢病毒滴度测定1.3 Determination of lentivirus titer
将Jurkat细胞的浓度调整至1×10 5个/300μl,充分混匀后取300μl重悬的细胞至24孔板的每个孔中。取70μl慢病毒浓缩液用Opti-MEM培养基进行5倍梯度稀释。将各稀释梯度的慢病毒以200μl/孔的用量加入上述24孔板中,使慢病毒感染Jurkat细胞(阴性对照组的Jurkat细胞只加入Opti-MEM培养基),并置于细胞培养箱中培养(培养温度为37℃,二氧化碳浓度为5%)。培养3天后,将各孔内的细胞轻柔混匀并转移至1.5-ml离心管中,用染色缓冲液(100ml PBS+1%BSA)清洗两次,每次800g离心3min。用相应抗体对上述细胞进行染色,再用流式细胞仪进行检测成功被慢病毒转导的Jurkat细胞的比例。将Jurkat细胞的慢病毒感染率记为P(%),病毒液体积记为V(ml),病毒液稀释倍数记为N,通过以下公式计算慢病毒滴度: Adjust the concentration of Jurkat cells to 1×10 5 cells/300 μl, mix thoroughly and take 300 μl of resuspended cells to each well of a 24-well plate. Take 70 μl lentiviral concentrate and perform 5-fold serial dilution with Opti-MEM medium. Add 200 μl/well of each diluted lentivirus into the above 24-well plate to infect Jurkat cells with the lentivirus (the Jurkat cells in the negative control group are only added with Opti-MEM medium), and culture in a cell culture incubator (Cultivation temperature is 37°C, carbon dioxide concentration is 5%). After culturing for 3 days, the cells in each well were gently mixed and transferred to a 1.5-ml centrifuge tube, washed twice with staining buffer (100 ml PBS+1% BSA), and centrifuged at 800 g for 3 min each time. The above cells were stained with corresponding antibodies, and then the proportion of Jurkat cells successfully transduced by lentivirus was detected by flow cytometry. The lentivirus infection rate of Jurkat cells is recorded as P (%), the volume of virus liquid is recorded as V (ml), and the dilution factor of virus liquid is recorded as N, and the lentivirus titer is calculated by the following formula:
慢病毒滴度(TU/ml)=P/V×N×10 5 Lentivirus titer (TU/ml) = P/V×N×10 5
结果:BN001滴度为5.46×10 8TU/ml,BN003滴度为2.46×10 8TU/ml,BN004滴度为6.31×10 8TU/ml,BN009滴度为9.51×10 8TU/ml。 Results: The titer of BN001 was 5.46×10 8 TU/ml, the titer of BN003 was 2.46×10 8 TU/ml, the titer of BN004 was 6.31×10 8 TU/ml, and the titer of BN009 was 9.51×10 8 TU/ml.
实施例2 CAR-T细胞的制备和检测Example 2 Preparation and detection of CAR-T cells
(a)制备T细胞(a) Preparation of T cells
将健康供者的外周血单个核细胞密度调整至2×10 6/ml,加入50ng/ml抗CD3抗体、50ng/ml抗CD28抗体,以及200IU/ml重组IL-2,置于细胞培养箱中培养24h(培养温度为37℃,二氧化碳浓度为5%)。 Adjust the density of peripheral blood mononuclear cells from healthy donors to 2×10 6 /ml, add 50ng/ml anti-CD3 antibody, 50ng/ml anti-CD28 antibody, and 200IU/ml recombinant IL-2, and place in a cell culture incubator Cultivate for 24 hours (cultivation temperature is 37° C., carbon dioxide concentration is 5%).
(b)慢病毒转导T细胞(b) Lentiviral transduction of T cells
清洗获得的T细胞,并将细胞密度调整至2×10 6/ml。以MOI=1~10TU/cell的用量加入慢病毒进行转导,同时补充50ng/ml抗CD3抗体、50ng/ml抗CD28抗体,以及200IU/ml重组IL-2,置于细胞培养箱中培养(培养温度为37℃,二氧化碳浓度为5%)。24h后,将细胞密度调整至1.5~2×10 6/ml,并补充300IU/ml的IL-2。转导后第4天,清洗细胞以去除上清中残留的慢病毒粒子,并继续置于细胞培养箱中培养5天(培养温度为37℃,二氧化碳浓度为5%),期间保持细胞密度为1~2×10 6/ml。转导后第10天收取细胞,并用冻存液(含有5%人血清白蛋白的冻存培养基:生理盐水=1:1)冻存于液氮中备用。获得的CAR-T细胞沿用相应CAR分子的命名,分别为BN001、BN003、BN004和BN009,未用慢病毒转导的T细胞命名为Ctrl T。 The obtained T cells were washed, and the cell density was adjusted to 2×10 6 /ml. Add lentivirus at MOI=1~10TU/cell for transduction, supplement 50ng/ml anti-CD3 antibody, 50ng/ml anti-CD28 antibody, and 200IU/ml recombinant IL-2 at the same time, and culture in a cell culture incubator ( The culture temperature was 37°C, and the carbon dioxide concentration was 5%). After 24 hours, the cell density was adjusted to 1.5-2×10 6 /ml, and 300 IU/ml of IL-2 was supplemented. On the 4th day after transduction, the cells were washed to remove residual lentiviral particles in the supernatant, and continued to be cultured in a cell incubator for 5 days (the culture temperature was 37°C, and the carbon dioxide concentration was 5%), during which the cell density was maintained at 1~2×10 6 /ml. The cells were harvested on the 10th day after transduction, and frozen in liquid nitrogen with a freezing medium (freezing medium containing 5% human serum albumin: normal saline = 1:1) for future use. The obtained CAR-T cells followed the nomenclature of the corresponding CAR molecules, namely BN001, BN003, BN004 and BN009, and the T cells not transduced with lentivirus were named Ctrl T.
(c)检测CAR分子的表达(c) Detection of the expression of CAR molecules
用PBS清洗待检测的Ctrl T、BN001、BN003、BN004和BN009细胞两次,并用FACS缓冲液(含0.1%叠氮化钠和0.4%BSA的PBS)重悬。按照抗体说明书将APC标记的抗人NKG2D抗体和BV421标记的抗人CD3抗体加入待检测细胞悬液中,4℃孵育60min。以Ctrl T细胞为阴性对照,用流式细胞仪检测BN001和BN009 细胞的NKG2D CAR分子的表达率。采用CytExpert软件分析。The Ctrl T, BN001, BN003, BN004 and BN009 cells to be tested were washed twice with PBS, and resuspended with FACS buffer (PBS containing 0.1% sodium azide and 0.4% BSA). Add APC-labeled anti-human NKG2D antibody and BV421-labeled anti-human CD3 antibody to the cell suspension to be tested according to the antibody instructions, and incubate at 4°C for 60 min. Using Ctrl T cells as a negative control, the expression rates of NKG2D CAR molecules in BN001 and BN009 cells were detected by flow cytometry. Analyzed by CytExpert software.
结果如图3所示,根据Ctrl T细胞的APC荧光信号水平设门,将作为阴性对照的Ctrl T细胞中CAR分子的表达率视为0.64%,BN001细胞的CAR分子表达率约为92.17%,BN003细胞的CAR分子表达率约为98.31%,BN004细胞的CAR分子表达率约为97.71%,BN009细胞的CAR分子表达率约为85.29%。The results are shown in Figure 3. The gate was set according to the APC fluorescence signal level of Ctrl T cells, and the expression rate of CAR molecules in Ctrl T cells as a negative control was regarded as 0.64%, and the expression rate of CAR molecules in BN001 cells was about 92.17%. The CAR molecule expression rate of BN003 cells is about 98.31%, the CAR molecule expression rate of BN004 cells is about 97.71%, and the CAR molecule expression rate of BN009 cells is about 85.29%.
(d)检测CCR2b的表达(d) Detection of expression of CCR2b
用PBS清洗待检测的Ctrl T、BN001、BN003、BN004和BN009细胞两次,并用FACS缓冲液重悬。按照抗体说明书将PE标记的抗人CCR2b抗体和BV421标记的抗人CD3抗体加入待检测细胞悬液中,4℃孵育60min。以未用慢病毒转染的Ctrl T细胞为阴性对照,用流式细胞仪检测上述CAR-T细胞的CCR2b表达率。采用CytExpert软件分析。The Ctrl T, BN001, BN003, BN004 and BN009 cells to be tested were washed twice with PBS and resuspended with FACS buffer. According to the antibody instructions, PE-labeled anti-human CCR2b antibody and BV421-labeled anti-human CD3 antibody were added to the cell suspension to be detected, and incubated at 4°C for 60 min. Using Ctrl T cells not transfected with lentivirus as a negative control, the expression rate of CCR2b in the above CAR-T cells was detected by flow cytometry. Analyzed by CytExpert software.
结果如图4所示,由于T细胞有一定水平的内源性CCR2表达,Ctrl T细胞可分为CCR2阴性和CCR2阳性细胞群。根据CCR2阴性的Ctrl T细胞群的PE荧光信号水平设门,Ctrl T的CCR2表达率约为44.20%,BN001的CCR2b表达率约为23.71%,BN003细胞的CCR2b表达率约为99.37%,BN004细胞的CCR2b表达率约为24.55%,BN009细胞的CCR2b表达率约为96.11%。The results are shown in Figure 4. Since T cells have a certain level of endogenous CCR2 expression, Ctrl T cells can be divided into CCR2-negative and CCR2-positive cell populations. According to the PE fluorescence signal level of the CCR2-negative Ctrl T cell population, the CCR2 expression rate of Ctrl T is about 44.20%, the CCR2b expression rate of BN001 is about 23.71%, the CCR2b expression rate of BN003 cells is about 99.37%, and the BN004 cells The expression rate of CCR2b in BN009 cells is about 24.55%, and the expression rate of CCR2b in BN009 cells is about 96.11%.
(e)检测CD40L的表达(e) Detection of CD40L expression
用PBS清洗待检测的Ctrl T、BN001、BN003、BN004和BN009细胞两次,并用FACS缓冲液重悬。按照抗体说明书将PE标记的抗人CD40L抗体和BV421标记的抗人CD3抗体加入待检测细胞悬液中,4℃孵育60min。以未用慢病毒转染的Ctrl T细胞为阴性对照,用流式细胞仪检测上述CAR-T细胞的CD表达率。采用CytExpert软件分析。The Ctrl T, BN001, BN003, BN004 and BN009 cells to be tested were washed twice with PBS and resuspended with FACS buffer. According to the antibody instructions, PE-labeled anti-human CD40L antibody and BV421-labeled anti-human CD3 antibody were added to the cell suspension to be detected, and incubated at 4°C for 60 min. Using Ctrl T cells not transfected with lentivirus as a negative control, the CD expression rate of the above CAR-T cells was detected by flow cytometry. Analyzed by CytExpert software.
结果如图5所示,由于T细胞有一定水平的内源性CD40L表达,Ctrl T细胞可分为CD40L阴性和CD40L阳性细胞群。根据CD40L阴性的Ctrl T细胞群的PE荧光信号水平设门,Ctrl T的CD40L表达率约为2.75%,BN001的CD40L表达率约为21.43%,BN003的CD40L表达率约为41.85%,BN004的CD40L表达率约为86.74%,BN009细胞的表达率约为71.86%。The results are shown in Figure 5. Since T cells have a certain level of endogenous CD40L expression, Ctrl T cells can be divided into CD40L-negative and CD40L-positive cell populations. According to the PE fluorescence signal level of the CD40L-negative Ctrl T cell population, the CD40L expression rate of Ctrl T is about 2.75%, the CD40L expression rate of BN001 is about 21.43%, the CD40L expression rate of BN003 is about 41.85%, and the CD40L expression rate of BN004 is about 2.75%. The expression rate is about 86.74%, and the expression rate of BN009 cells is about 71.86%.
实施例3靶细胞检测Example 3 Target cell detection
(a)靶细胞培养条件(a) Target cell culture conditions
供试结直肠癌细胞系(又称靶细胞或靶细胞系):HCT 116(McCoy's 5a培养基+10%胎牛血清+100U/ml青霉素+100μg/ml链霉素),LS174T(EMEM培养基+10%胎牛血清+100U/ml青霉素+100μg/ml链霉素),LoVo(F-12K培养基+10% 胎牛血清+100U/ml青霉素+100μg/ml链霉素),SW480(Leibovitz's L-15Medium培养基+10%胎牛血清+100U/ml青霉素+100μg/ml链霉素)。供试卵巢癌细胞系:SK-OV-3(McCoy's 5a培养基+10%胎牛血清+100U/ml青霉素+100μg/ml链霉素)。Tested colorectal cancer cell lines (also known as target cells or target cell lines): HCT 116 (McCoy's 5a medium + 10% fetal bovine serum + 100U/ml penicillin + 100μg/ml streptomycin), LS174T (EMEM medium +10% fetal bovine serum+100U/ml penicillin+100μg/ml streptomycin), LoVo (F-12K medium+10% fetal bovine serum+100U/ml penicillin+100μg/ml streptomycin), SW480 (Leibovitz's L-15 Medium + 10% fetal bovine serum + 100 U/ml penicillin + 100 μg/ml streptomycin). Tested ovarian cancer cell line: SK-OV-3 (McCoy's 5a medium + 10% fetal bovine serum + 100 U/ml penicillin + 100 μg/ml streptomycin).
(b)检测NKG2D配体(MICA/MICB)的表达(b) Detection of expression of NKG2D ligands (MICA/MICB)
用PBS清洗上述靶细胞两次,并用FACS缓冲液重悬。按照抗体说明书将APC标记的抗人MICA/MICB抗体加入各靶细胞悬液中,4℃孵育60min。以不加抗体孵育的靶细胞作为阴性对照,用流式细胞仪检测靶细胞的MICA/MICB表达率。采用CytExpert软件分析。The above target cells were washed twice with PBS and resuspended with FACS buffer. Add APC-labeled anti-human MICA/MICB antibody to each target cell suspension according to the antibody instructions, and incubate at 4°C for 60 min. The target cells incubated without antibody were used as negative control, and the MICA/MICB expression rate of the target cells was detected by flow cytometry. Analyzed by CytExpert software.
结果如图6所示,HCT 116、LS174T和SW480的MICA/MICB表达率均高于96%以上,LoVo和SK-OV-3的MICA/MICB表达率低于8%。The results are shown in Figure 6, the MICA/MICB expression rates of HCT 116, LS174T and SW480 were all higher than 96%, and the MICA/MICB expression rates of LoVo and SK-OV-3 were lower than 8%.
(c)检测NKG2D配体(ULBP-1)的表达率(c) Detection of expression rate of NKG2D ligand (ULBP-1)
用PBS清洗上述靶细胞两次,并用FACS缓冲液重悬。按照抗体说明书将PC5.5标记的抗人ULBP-1抗体加入各靶细胞悬液中,4℃孵育60min。以不加抗体孵育的靶细胞作为阴性对照,用流式细胞仪检测靶细胞的ULBP-2/5/6表达率。采用CytExpert软件分析。The above target cells were washed twice with PBS and resuspended with FACS buffer. Add PC5.5-labeled anti-human ULBP-1 antibody to each target cell suspension according to the antibody instructions, and incubate at 4°C for 60 min. The target cells incubated without antibody were used as a negative control, and the expression rate of ULBP-2/5/6 in the target cells was detected by flow cytometry. Analyzed by CytExpert software.
结果如图7所示,HCT116的ULBP-1表达率约为77.25%,LoVo的ULBP-1表达率约为9.09%,其他细胞的ULBP-1表达率均低于3%。The results are shown in Figure 7, the ULBP-1 expression rate of HCT116 is about 77.25%, the ULBP-1 expression rate of LoVo is about 9.09%, and the ULBP-1 expression rate of other cells is lower than 3%.
(d)检测NKG2D配体(ULBP-2/5/6)的表达率(d) Detection of expression rate of NKG2D ligand (ULBP-2/5/6)
用PBS清洗上述靶细胞两次,并用FACS缓冲液重悬。按照抗体说明书将PE标记的抗人ULBP-2/5/6抗体加入各靶细胞悬液中,4℃孵育60min。以不加抗体孵育的靶细胞作为阴性对照,用流式细胞仪检测靶细胞的ULBP-2/5/6表达率。采用CytExpert软件分析。The above target cells were washed twice with PBS and resuspended with FACS buffer. According to the antibody instructions, PE-labeled anti-human ULBP-2/5/6 antibodies were added to each target cell suspension, and incubated at 4°C for 60 min. The target cells incubated without antibody were used as a negative control, and the expression rate of ULBP-2/5/6 in the target cells was detected by flow cytometry. Analyzed by CytExpert software.
结果如图8所示,HCT116和SW480的ULBP-2/5/6表达率均高于92%,LoVo的ULBP-2/5/6表达率约为86.82%,SK-OV-3的ULBP-2/5/6表达率约为56.90%,LS174T的ULBP-2/5/6表达率约为33.42%。The results are shown in Figure 8. The ULBP-2/5/6 expression rates of HCT116 and SW480 are both higher than 92%, the ULBP-2/5/6 expression rate of LoVo is about 86.82%, and the ULBP-2/5/6 expression rate of SK-OV-3 is about 86.82%. The expression rate of 2/5/6 was about 56.90%, and the expression rate of ULBP-2/5/6 in LS174T was about 33.42%.
(e)检测NKG2D配体(ULBP-3)的表达率(e) Detection of expression rate of NKG2D ligand (ULBP-3)
用PBS清洗上述靶细胞两次,并用FACS缓冲液重悬。按照抗体说明书将PE标记的抗人ULBP-3抗体加入各靶细胞悬液中,4℃孵育60min。以不加抗体孵育的靶细胞作为阴性对照,用流式细胞仪检测靶细胞的ULBP-3表达率。采用CytExpert软件分析。The above target cells were washed twice with PBS and resuspended with FACS buffer. According to the antibody instructions, PE-labeled anti-human ULBP-3 antibody was added to each target cell suspension, and incubated at 4°C for 60 min. The target cells incubated without antibody were used as a negative control, and the expression rate of ULBP-3 in the target cells was detected by flow cytometry. Analyzed by CytExpert software.
结果如图9所示,HCT116和SW480的ULBP-3表达率均高于92%,LS174T 的ULBP-3表达率约为46.57%,LoVo和SK-OV-3的ULBP-3表达率均低于34%。The results are shown in Figure 9, the ULBP-3 expression rates of HCT116 and SW480 are both higher than 92%, the ULBP-3 expression rates of LS174T are about 46.57%, and the ULBP-3 expression rates of LoVo and SK-OV-3 are lower than 34%.
(f)检测NKG2D配体(ULBP-4)的表达率(f) Detection of the expression rate of NKG2D ligand (ULBP-4)
用PBS清洗上述靶细胞两次,并用FACS缓冲液重悬。按照抗体说明书将PE标记的抗人ULBP-4抗体加入各靶细胞悬液中,4℃孵育60min。以不加抗体孵育的靶细胞作为阴性对照,用流式细胞仪检测靶细胞的ULBP-4表达率。采用CytExpert软件分析。The above target cells were washed twice with PBS and resuspended with FACS buffer. According to the antibody instructions, PE-labeled anti-human ULBP-4 antibody was added to each target cell suspension, and incubated at 4°C for 60 min. The target cells incubated without antibody were used as a negative control, and the expression rate of ULBP-4 in the target cells was detected by flow cytometry. Analyzed by CytExpert software.
结果如图10所示,HCT 116、LS174T、LoVo和SW480的ULBP-4表达率均在80%以上,SK-OV-3的ULBP-4表达率约为71.64%。The results are shown in Figure 10. The ULBP-4 expression rates of HCT 116, LS174T, LoVo and SW480 were all above 80%, and the ULBP-4 expression rate of SK-OV-3 was about 71.64%.
(g)检测CD40的表达率(g) Detect the expression rate of CD40
用PBS清洗上述靶细胞两次,并用FACS缓冲液重悬。按照抗体说明书将APC标记的抗人CD40抗体加入各靶细胞悬液中,4℃孵育60min。以不加抗体孵育的靶细胞作为阴性对照,用流式细胞仪检测靶细胞的CD40表达率。采用CytExpert软件分析。The above target cells were washed twice with PBS and resuspended with FACS buffer. Add APC-labeled anti-human CD40 antibody to each target cell suspension according to the antibody instructions, and incubate at 4°C for 60 min. The target cells incubated without antibody were used as a negative control, and the CD40 expression rate of the target cells was detected by flow cytometry. Analyzed by CytExpert software.
结果如图11所示,HCT 116的CD40表达率约为94.56%,SK-OV-3的表达率约为86.61%,其他细胞的表达率均低于3%。The results are shown in Figure 11, the expression rate of CD40 in HCT 116 is about 94.56%, the expression rate of SK-OV-3 is about 86.61%, and the expression rate of other cells is lower than 3%.
实施例4 CAR-T细胞的趋化迁移Example 4 Chemotaxis migration of CAR-T cells
在本实施例中,使用Incucyte S3活细胞动态成像分析仪,对T细胞迁移进行实时检测。ClearView 96孔趋化运动微孔板前处理:在微孔板的上室加入20μg/ml的Protein-G蛋白,37℃放置1h,以包被上室微孔滤膜;用D-PBS清洗后,在微孔板的上室加入5μg/ml的ICAM-1蛋白,37℃放置2h,对上室微孔滤膜进行二次包被;最后用1%BSA对上室微孔滤膜的上下表面进行封闭30min。用含0.5%FBS的RPMI 1640培养基清洗并重悬待检测的T细胞,将细胞密度调整至2~8×10 4个/ml。用D-PBS清洗后,在微孔板的上室加入60μl待检测的T细胞悬液(5000个/孔)。静置1h后,在下室加入含0.5%FBS的RPMI 1640培养基及相应浓度的CCL2,并置于Incucyte S3活细胞动态影像成像分析仪中,以30min为间隔,对上室的T细胞进行24h成像记录。通过Incucyte趋化迁移分析模块软件对上室T细胞总面积的变化情况进行分析,以反映T细胞的趋化迁移能力(从上室迁移至下室的细胞越多,上室的细胞数量和相应的细胞总面积越小,趋化迁移能力越强)。 In this example, the Incucyte S3 Live Cell Dynamic Imaging Analyzer was used to detect the migration of T cells in real time. Pretreatment of ClearView 96-well Chemotaxis Motility Microplate: Add 20 μg/ml Protein-G protein to the upper chamber of the microplate, and place it at 37°C for 1 hour to coat the upper chamber microporous membrane; after washing with D-PBS, Add 5 μg/ml of ICAM-1 protein to the upper chamber of the microwell plate, place it at 37°C for 2 hours, and coat the upper chamber microporous membrane twice; finally use 1% BSA to coat the upper and lower surfaces of the upper chamber microporous membrane Carry out sealing for 30min. Wash and resuspend the T cells to be tested with RPMI 1640 medium containing 0.5% FBS, and adjust the cell density to 2-8×10 4 cells/ml. After washing with D-PBS, 60 μl of T cell suspension to be detected (5000 cells/well) was added to the upper chamber of the microwell plate. After standing still for 1 hour, RPMI 1640 medium containing 0.5% FBS and corresponding concentration of CCL2 were added to the lower chamber, and placed in the Incucyte S3 live cell dynamic imaging analyzer, and the T cells in the upper chamber were tested for 24 hours at intervals of 30 minutes. Imaging records. The change of the total area of T cells in the upper chamber was analyzed by the Incucyte chemotaxis and migration analysis module software to reflect the chemotaxis and migration ability of T cells (the more cells migrated from the upper chamber to the lower chamber, the number of cells in the upper chamber and the corresponding The smaller the total cell area, the stronger the chemotaxis and migration ability).
结果如图12所示,在24h内,未加入CCL2诱导的BN001、BN003、BN004和BN009的迁移效率没有显著差异(图12中的A);当在下室培养基中加入100ng/ml CCL2后,BN004向下室的迁移效率与BN001没有显著差异,但BN003和BN009细胞的迁移效率都显著高于BN001(图12中的B),且BN009的迁移效率最高。从 该迁移曲线可知,加入CCL2后,BN003中趋化迁移的程度是逐渐缓慢提高的,而BN009中发生迁移的程度则是在约6h开始出现显著提高,说明BN009能以更灵敏的方式响应CCL2的诱导而进行更高效的趋化迁移,联合表达CCL2和CD40L可以有效提升BN009向高浓度CCL2的趋化迁移能力。The results are shown in Figure 12, within 24h, there was no significant difference in the migration efficiency of BN001, BN003, BN004 and BN009 induced by CCL2 (A in Figure 12); after adding 100ng/ml CCL2 in the lower chamber medium, The migration efficiency of BN004 to the lower chamber was not significantly different from that of BN001, but the migration efficiency of BN003 and BN009 cells was significantly higher than that of BN001 (B in Figure 12), and BN009 had the highest migration efficiency. From the migration curve, it can be seen that after adding CCL2, the degree of chemotactic migration in BN003 gradually increased slowly, while the degree of migration in BN009 began to increase significantly at about 6 hours, indicating that BN009 can respond to CCL2 in a more sensitive manner The combined expression of CCL2 and CD40L can effectively enhance the chemotactic migration ability of BN009 to high concentrations of CCL2.
此外,虽然BN009和BN003都表达CCR2b,而原本认为CD40L和NKG2D胞外域等结构对于趋化迁移能力无实质性影响(图12B中,BN001和BN004向高浓度CCL2的趋化迁移能力基本差别),图12B的结果出乎意料地显示:NKG2D胞外域的CAR分子与CCR2b及CD40L同时联合表达于CAR-T细胞所制备的BN009,居然具有显著优于BN003的高浓度CCL2的趋化迁移能力(如图12B中10-24小时的数据)。In addition, although both BN009 and BN003 express CCR2b, it was originally believed that the CD40L and NKG2D ectodomain and other structures had no substantial impact on the chemotactic migration ability (in Figure 12B, the chemotactic migration ability of BN001 and BN004 to high concentrations of CCL2 is basically different), The results in Figure 12B unexpectedly show that the CAR molecule of the NKG2D ectodomain is co-expressed with CCR2b and CD40L in CAR-T cells at the same time. Data from 10-24 hours in Figure 12B).
实施例5 CAR-T细胞的体外功能Example 5 In vitro function of CAR-T cells
(a)通过EuTDA法检测杀伤效果(a) Detection of killing effect by EuTDA method
用AIM-V培养基清洗靶细胞一次。将靶细胞密度调节至1×10 6/ml,并以2μl/ml的用量加入DELFIA BATDA Reagent混匀,37℃下孵育30min。用AIM-V培养基清洗靶细胞三次后,以1×10 4/孔的密度将靶细胞接种于96孔板中。加入100μl T细胞,并置于二氧化碳培养箱培养2h(培养温度为37℃,二氧化碳浓度为5%)。最后,以500×g离心5min,取20μl上清液转移至加有Europium solution(200μl/孔)的96孔板中。室温孵育15min后,在酶标仪中检测。 Wash target cells once with AIM-V medium. Adjust the target cell density to 1×10 6 /ml, add DELFIA BATDA Reagent at 2 μl/ml, mix well, and incubate at 37°C for 30 minutes. After the target cells were washed three times with AIM-V medium, the target cells were seeded in a 96-well plate at a density of 1×10 4 /well. 100 μl of T cells were added, and cultured in a carbon dioxide incubator for 2 hours (the culture temperature was 37° C., and the carbon dioxide concentration was 5%). Finally, centrifuge at 500×g for 5 min, and transfer 20 μl of the supernatant to a 96-well plate added with Europium solution (200 μl/well). After incubation at room temperature for 15 min, it was detected in a microplate reader.
结果如表1和图13所示,与Ctrl T相比,BN001、BN003、BN004和BN009对各靶细胞都有显著的杀伤效果。与BN001相比,BN003和BN004对各靶细胞的杀伤率没有显著提升,而BN009的杀伤率则显著高于BN001(P<0.05)。The results are shown in Table 1 and Figure 13. Compared with Ctrl T, BN001, BN003, BN004 and BN009 have significant killing effects on each target cell. Compared with BN001, BN003 and BN004 did not significantly increase the killing rate of each target cell, while the killing rate of BN009 was significantly higher than that of BN001 (P<0.05).
为分析在CAR-T细胞杀伤肿瘤细胞的过程中,同时联合表达CCR2和CD40L是否对杀伤效果有协同增效作用,统计BN003、BN004、BN009相对于BN001的杀伤率提升幅度(提升幅度=相应CAR-T细胞杀伤率-BN001杀伤率)。结果如表2所示,BN009对各靶细胞杀伤效果的提升幅度均高于BN003和BN004提升幅度之和,说明在CAR-T细胞中同时联合表达CCL2和CD40L对杀伤效果有协同增效作用,显著优于在CAR-T细胞中单独表达CCL2或CD40L的方案。In order to analyze whether the combined expression of CCR2 and CD40L has a synergistic effect on the killing effect in the process of CAR-T cells killing tumor cells, the increase in the killing rate of BN003, BN004, and BN009 relative to BN001 was calculated (increase = corresponding CAR - T cell killing rate - BN001 killing rate). The results are shown in Table 2. The increase in the killing effect of BN009 on each target cell is higher than the sum of the increase in BN003 and BN004, indicating that the combined expression of CCL2 and CD40L in CAR-T cells has a synergistic effect on the killing effect. Significantly better than the scheme of expressing CCL2 or CD40L alone in CAR-T cells.
表1 EuTDA检测各NKG2D CAR-T细胞对肿瘤细胞的杀伤率(n=3)Table 1 The killing rate of NKG2D CAR-T cells on tumor cells detected by EuTDA (n=3)
Figure PCTCN2022130696-appb-000017
Figure PCTCN2022130696-appb-000017
Figure PCTCN2022130696-appb-000018
Figure PCTCN2022130696-appb-000018
表2各NKG2D CAR-T细胞相对于BN001的杀伤率提升幅度(n=3)Table 2 The killing rate improvement of each NKG2D CAR-T cell relative to BN001 (n=3)
Figure PCTCN2022130696-appb-000019
Figure PCTCN2022130696-appb-000019
*:当BN009的相对于BN001的提升幅度(即CCR2b及CD40L的总体提升幅度)大于“BN003提升幅度(CCR2b的提升幅度)+BN004提升幅度(CD40L的提升幅度)”之和,则表明存在协同效应。*: When the improvement of BN009 relative to BN001 (that is, the overall improvement of CCR2b and CD40L) is greater than the sum of "the improvement of BN003 (the improvement of CCR2b) + the increase of BN004 (the improvement of CD40L)", it indicates that there is synergy effect.
(b)检测细胞因子分泌(b) Detection of cytokine secretion
将Ctrl T、BN001、BN003、BN004和BN009分别与相应的靶细胞共培养于不含IL-2的AIM-V培养基中(效靶比为2.5:1)。24h后,用ddH 2O溶解INF-γ标准品,室温放置15~20min保证充分溶解,将标准品按推荐梯度倍比稀释。吸取上述共培养的细胞上清,用ddH 2O进行2倍和20倍稀释。分别将标准品和实验样品加入相应反应孔中,每孔100μl。室温孵育1~3h后,配制1×清洗液,每孔用360μl清洗液清洗4次,并将孔中液体拍干,每孔加入200μl酶标检测抗体,室温孵育1~3h。每孔用360μl清洗液清洗4次,并将孔中液体拍干后,加入200μl显色底物。室温避光孵育30~60min后,每孔加入50μl终止液,用酶标仪测定450nm的吸光值。 Ctrl T, BN001, BN003, BN004, and BN009 were co-cultured with corresponding target cells in AIM-V medium without IL-2 (effect-to-target ratio 2.5:1). After 24 hours, dissolve the INF-γ standard with ddH 2 O, place at room temperature for 15-20 minutes to ensure full dissolution, and dilute the standard according to the recommended gradient ratio. Aspirate the supernatant of the above co-cultured cells and dilute them 2-fold and 20-fold with ddH 2 O. The standard and experimental samples were added to the corresponding reaction wells, 100 μl per well. After incubating at room temperature for 1-3 hours, prepare 1× cleaning solution, wash each well with 360 μl cleaning solution 4 times, pat dry the liquid in the well, add 200 μl enzyme-labeled detection antibody to each well, and incubate at room temperature for 1-3 hours. Each well was washed 4 times with 360 μl of washing solution, and after the liquid in the well was patted dry, 200 μl of chromogenic substrate was added. After incubation at room temperature in the dark for 30-60 min, 50 μl of stop solution was added to each well, and the absorbance at 450 nm was measured with a microplate reader.
结果如表3和图14所示,与Ctrl T细胞相比,BN001、BN003、BN004和BN009与各靶细胞共培养后都有显著的IFN-γ释放。对于SW480和SK-OV-3细胞,均未检测到BN003、BN004和BN009相对于BN001的IFN-γ释放水平的提高。对于LS174T细胞,BN004和BN009的IFN-γ显著高于BN001(P<0.05).对于HCT116 和SK-OV-3,只有BN009的IFN-γ释放水平显著高于BN001(P<0.05)。上述结果说明,对于部分靶细胞,在CAR-T细胞中同时联合表达CCL2和CD40L可以显著提升IFN-γ的释放水平。The results are shown in Table 3 and Figure 14. Compared with Ctrl T cells, BN001, BN003, BN004 and BN009 all had significant IFN-γ release after co-culture with each target cell. For both SW480 and SK-OV-3 cells, no increase in IFN-γ release levels was detected for BN003, BN004 and BN009 relative to BN001. For LS174T cells, the IFN-γ release level of BN004 and BN009 was significantly higher than that of BN001 (P<0.05). For HCT116 and SK-OV-3, only the IFN-γ release level of BN009 was significantly higher than that of BN001 (P<0.05). The above results indicated that, for some target cells, co-expression of CCL2 and CD40L in CAR-T cells can significantly increase the release level of IFN-γ.
表3ELISA检测各NKG2D CAR-T细胞的IFN-γ释放水平(pg/ml)Table 3 ELISA detection of IFN-γ release level of each NKG2D CAR-T cell (pg/ml)
Figure PCTCN2022130696-appb-000020
Figure PCTCN2022130696-appb-000020
*信号低于检测阈值。 * Signal below detection threshold.
实施例6 CAR-T细胞体内抑瘤功能Example 6 In vivo tumor suppression function of CAR-T cells
以LoVo为靶细胞,以BN001和BN009为效应细胞进行小鼠皮下移植瘤抑制效果测试。用免疫缺陷的B-NDG小鼠进行实验,以观察联合表达CCR2的NKG2D CAR-T细胞对肿瘤浸润和抑制效果。取6~8周龄的B-NDG小鼠(百奥赛图江苏基因生物技术有限公司)进行皮下瘤药效实验。共取24只小鼠进行实验,随机分为4组,每组6只,分别为溶媒对照组、Ctrl T对照组、BN001对照组、BN009实验组。Using LoVo as target cells and BN001 and BN009 as effector cells to test the inhibitory effect of subcutaneous tumor transplantation in mice. Experiments were conducted with immunodeficient B-NDG mice to observe the effect of NKG2D CAR-T cells co-expressing CCR2 on tumor infiltration and inhibition. B-NDG mice aged 6-8 weeks (Biocytogen Jiangsu Gene Biotechnology Co., Ltd.) were used for subcutaneous tumor efficacy experiments. A total of 24 mice were selected for the experiment, and were randomly divided into 4 groups, 6 mice in each group, which were vehicle control group, Ctrl T control group, BN001 control group, and BN009 experimental group.
用胰酶消化法收集处于对数生长期且生长状态良好的靶细胞,用生理盐水洗涤1次后,调整细胞密度为2×10 7/ml。在B-NDG小鼠右侧靠近腋下部位皮下注射100μl细胞悬液,即每只小鼠接种2×10 6的靶细胞,接种日记为第0天。 The target cells in logarithmic growth phase and in good growth state were collected by trypsinization method, washed once with normal saline, and the cell density was adjusted to 2×10 7 /ml. 100 μl of cell suspension was subcutaneously injected into the right side of the B-NDG mice near the armpit, that is, each mouse was inoculated with 2×10 6 target cells, and the inoculation diary was regarded as day 0.
接种靶细胞后第7天(或肿瘤平均体积约为100mm 3时),通过尾静脉分别注射CAR-T细胞(1×10 7/只)、Ctrl T细胞(1×10 7/只)和溶媒(100μl/只),注射受试物当天记为治疗的第0天。每周测量肿瘤大小和小鼠体重2~3次,在第3天、第10天、第28天采集血样,加入EDTA抗凝后,用qPCR检测血细胞中CAR-T细胞在小鼠体内的留存情况,通过ELISA检测INF-γ以监测细胞因子的释放水平。治疗28天后,对小鼠实施安乐死,取肿瘤、心、肝、脾、肺、肾、脑、卵巢等组织称重,每组取2只小鼠的组织保存在80℃冰箱,用于提取DNA,检测CAR-T细胞在肿瘤组织中 的浸润情况及其在各器官的分布情况;每组取2只小鼠的组织进行固定,用HE染色法检测肿瘤细胞的形态和用免疫组化检测组织中抗原的表达情况。 Seven days after inoculation of target cells (or when the average tumor volume is about 100 mm 3 ), CAR-T cells (1×10 7 /monkey), Ctrl T cells (1×10 7 /monkey) and vehicle were respectively injected through the tail vein. (100 μl/only), the day of injection of the test substance was recorded as the 0th day of treatment. The tumor size and mouse weight were measured 2 to 3 times a week, and blood samples were collected on the 3rd, 10th, and 28th days. After adding EDTA for anticoagulation, qPCR was used to detect the retention of CAR-T cells in the blood cells in the mice. In this case, INF-γ was detected by ELISA to monitor the release level of cytokines. After 28 days of treatment, the mice were euthanized, and the tumor, heart, liver, spleen, lung, kidney, brain, ovary and other tissues were taken and weighed. The tissues of 2 mice in each group were stored in a refrigerator at 80°C for DNA extraction , to detect the infiltration of CAR-T cells in tumor tissues and their distribution in various organs; the tissues of 2 mice in each group were fixed, and the morphology of tumor cells was detected by HE staining and the tissues were detected by immunohistochemistry. The expression of antigens in the
结果如表4所示,在注射CAR-T细胞后28天,溶媒对照组小鼠的平均肿瘤体积约为947mm 3,平均肿瘤负荷约为1.215g;Ctrl T对照组小鼠的平均肿瘤体积约为895mm 3,平均肿瘤负荷约为1.192g;BN001对照组小鼠的平均肿瘤体积约为1178mm 3,平均肿瘤负荷约为1.267g;BN009实验组小鼠的平均肿瘤体积约为623mm 3,平均肿瘤负荷约为0.860g。与BN001相比,注射BN009细胞的小鼠的平均肿瘤负荷显著下降,下降幅度约为32.1%。 The results are shown in Table 4. Twenty-eight days after injection of CAR-T cells, the average tumor volume of mice in vehicle control group was about 947 mm 3 , and the average tumor burden was about 1.215 g; the average tumor volume of mice in Ctrl T control group was about The average tumor volume of mice in the BN001 control group was about 1178mm 3 , and the average tumor burden was about 1.267g; the average tumor volume of mice in the BN009 experimental group was about 623mm 3 , and the average tumor The load is about 0.860g. Compared with BN001, the average tumor burden of mice injected with BN009 cells was significantly reduced by about 32.1%.
用qPCR检测肿瘤组织中的LTR序列,以衡量CAR-T细胞在肿瘤组织中的浸润情况。结果发现,溶媒对照组小鼠的肿瘤组织内LTR的平均本底信号水平为8.18copies/μg DNA;Ctrl T对照组小鼠的肿瘤组织内LTR的平均本底信号水平为42.86copies/μg DNA;BN001对照组小鼠的肿瘤组织内平均LTR含量为2055.38copies/μg DNA;BN009实验组小鼠的肿瘤组织内平均LTR含量为3872.70copies/μg DNA。与BN001相比,BN009向小鼠的皮下移植瘤组织的归巢能力获得显著提升,提升幅度约为88.4%。LTR sequences in tumor tissue were detected by qPCR to measure the infiltration of CAR-T cells in tumor tissue. The results showed that the average background signal level of LTR in the tumor tissue of the mice in the vehicle control group was 8.18 copies/μg DNA; the average background signal level of LTR in the tumor tissue of the mice in the Ctrl T control group was 42.86 copies/μg DNA; The average LTR content in the tumor tissue of the mice in the BN001 control group was 2055.38 copies/μg DNA; the average LTR content in the tumor tissue of the mice in the BN009 experimental group was 3872.70 copies/μg DNA. Compared with BN001, the homing ability of BN009 to subcutaneous transplanted tumor tissue in mice was significantly improved, with an increase rate of about 88.4%.
表4体内药效实验中CAR-T抑瘤效果与归巢情况Table 4 CAR-T antitumor effect and homing situation in the in vivo drug efficacy experiment
Figure PCTCN2022130696-appb-000021
Figure PCTCN2022130696-appb-000021
讨论discuss
在针对结直肠癌等实体瘤的CAR-T治疗中,主要识别的靶点有CD133、CEA、EGFR、HER-2和NKG2D配体等。其中,NKG2D配体包括MICA、MICB、ULBP-1、ULBP-2、ULBP-3、ULBP-4、ULBP-5和ULBP-6,广泛表达于多种肿瘤细胞中。NKG2D(又称CD314)是天然免疫系统中的一个重要的激活性受体,主要表达于自然杀伤细胞、γδT细胞和CD8+T细胞的表面。以NKG2D作为CAR分子的抗原识别结构域具有众多优点。与针对单一靶点的特异性抗体CAR分子不同,基于NKG2D改造获得的CAR分子可以识别上述8种不同的NKG2D配体,更有利于治疗于异质性高或容易丢失靶点抗原的肿瘤。另外,这些NKG2D配体都位于靶细胞表面。因此,与TCR-T相比,NKG2D CAR-T不需要MHC分子的抗原呈递过程即可直接识别肿瘤细胞。重要的是,NKG2D配体在结直肠癌、卵巢癌、胰腺癌、白血病等上 皮源性的肿瘤细胞上有高水平表达,但在正常细胞中不表达或者表达水平极低,是肿瘤特异性治疗的理想靶点。此外,NKG2D CAR-T细胞表面不携带任何外来的可能引发患者免疫反应的蛋白结构,从而降低了CAR-T细胞被患者免疫系统排斥的可能性。In CAR-T therapy for solid tumors such as colorectal cancer, the main targets identified are CD133, CEA, EGFR, HER-2, and NKG2D ligands. Among them, NKG2D ligands include MICA, MICB, ULBP-1, ULBP-2, ULBP-3, ULBP-4, ULBP-5 and ULBP-6, which are widely expressed in a variety of tumor cells. NKG2D (also known as CD314) is an important activating receptor in the innate immune system, mainly expressed on the surface of natural killer cells, γδT cells and CD8+T cells. Using NKG2D as the antigen recognition domain of CAR molecules has many advantages. Unlike specific antibody CAR molecules targeting a single target, CAR molecules engineered based on NKG2D can recognize the above-mentioned 8 different NKG2D ligands, which is more conducive to the treatment of tumors with high heterogeneity or easy loss of target antigens. Additionally, these NKG2D ligands are all located on the surface of target cells. Therefore, compared with TCR-T, NKG2D CAR-T does not require the antigen presentation process of MHC molecules to directly recognize tumor cells. Importantly, NKG2D ligands are expressed at high levels in epithelial tumor cells such as colorectal cancer, ovarian cancer, pancreatic cancer, and leukemia, but are not expressed or expressed at very low levels in normal cells. ideal target. In addition, the surface of NKG2D CAR-T cells does not carry any foreign protein structures that may trigger the patient's immune response, thereby reducing the possibility of CAR-T cells being rejected by the patient's immune system.
实体瘤细胞可以通过分泌趋化因子CXCL12和CXCL5,阻止CAR-T细胞向肿瘤组织的迁移和浸润。相反,实体瘤细胞较少分泌可促进CAR-T细胞迁移的趋化因子。这两个因素使CAR-T细胞难以到达实体瘤部位。因此,提高CAR-T细胞对肿瘤趋化因子的特异性识别和灵敏度是影响CAR-T治疗效果的关键因素之一,而在CAR-T细胞中联合表达趋化因子受体是解决改问题的重要方法。趋化因子是一类特殊的细胞因子,包含50多个成员。根据结构分为CC、CXC、CX3C和XC四类;趋化因子受体则相应分为CCR、CXCR、CX3CR和XCR4种,约有20个成员。趋化因子的一种主要作用机制是通过形成可溶性的或固定于基质中的浓度梯度来诱导免疫细胞的定向迁移,从而调免疫细胞在组织中的浸润。目前,有部分CAR-T技术已采用联合表达趋化因子受体的方式来促进CAR-T细胞快速迁移至癌细胞处,从而提高CAR-T细胞的肿瘤治疗效果。不同类型的实体瘤释放的趋化因子的种类和水平都不尽相同,也有不同的免疫逃逸机制。因此,针对特定癌种选择适合的靶点抗原,再联合表达合适的趋化因子受体,是提高这类型CAR-T细胞治疗效果的关键。Solid tumor cells can prevent the migration and infiltration of CAR-T cells into tumor tissue by secreting chemokines CXCL12 and CXCL5. In contrast, solid tumor cells secrete less chemokines that can promote CAR-T cell migration. These two factors make it difficult for CAR-T cells to reach solid tumor sites. Therefore, improving the specific recognition and sensitivity of CAR-T cells to tumor chemokines is one of the key factors affecting the therapeutic effect of CAR-T, and the combined expression of chemokine receptors in CAR-T cells is the solution to this problem. important method. Chemokines are a special class of cytokines, including more than 50 members. According to the structure, it is divided into four types: CC, CXC, CX3C and XC; chemokine receptors are correspondingly divided into four types: CCR, CXCR, CX3CR and XCR, with about 20 members. One of the main mechanisms of action of chemokines is to induce directional migration of immune cells by forming concentration gradients soluble or immobilized in the matrix, thereby regulating the infiltration of immune cells in tissues. At present, some CAR-T technologies have used the combined expression of chemokine receptors to promote the rapid migration of CAR-T cells to cancer cells, thereby improving the tumor therapeutic effect of CAR-T cells. Different types of solid tumors release different types and levels of chemokines, and have different immune escape mechanisms. Therefore, selecting the appropriate target antigen for a specific cancer type, combined with the expression of an appropriate chemokine receptor, is the key to improving the therapeutic effect of this type of CAR-T cells.
本发明通过特定CAR分子和特定趋化因子受体以及CD40L的特定组合,既高效解决了靶向恶性肿瘤病灶的问题,又有效克服了恶性肿瘤异质性的问题,并实现了协同的优异的治疗效果。Through the specific combination of specific CAR molecules, specific chemokine receptors and CD40L, the present invention not only efficiently solves the problem of targeting malignant tumor lesions, but also effectively overcomes the problem of malignant tumor heterogeneity, and achieves synergistic and excellent treatment effect.
一方面,本发明通过CCR2b作为联合表达的趋化因子受体,提高了CAR-T细胞向恶性肿瘤病灶的迁移能力,从而提升了治疗效率。CCR2b主要识别的趋化因子CCL2在结直肠癌、卵巢癌、胰腺癌等多种恶性肿瘤中有异常的高表达,且在正常组织细胞中表达水平极低。同时,CCR2b与CCL2的亲和力极高(约为0.7nM,IC 50数值越小,对应的亲和力和灵敏度越高),灵敏度远高于其他趋化因子受体和趋化因子的组合(如CCR2a/CCL2组合的IC 50数值约为5nM,CXCR3/CXCL11的组合C 50数值约为8.2nM)。 On the one hand, the present invention uses CCR2b as a co-expressed chemokine receptor to improve the ability of CAR-T cells to migrate to malignant tumor lesions, thereby improving the treatment efficiency. CCL2, a chemokine mainly recognized by CCR2b, is abnormally highly expressed in various malignant tumors such as colorectal cancer, ovarian cancer, and pancreatic cancer, and its expression level is extremely low in normal tissue cells. At the same time, the affinity between CCR2b and CCL2 is extremely high (about 0.7nM, the smaller the IC 50 value, the higher the corresponding affinity and sensitivity), and the sensitivity is much higher than the combination of other chemokine receptors and chemokines (such as CCR2a/ The IC 50 value of the combination of CCL2 is about 5nM, and the C 50 value of the combination of CXCR3/CXCL11 is about 8.2nM).
另一方面,本发明采用NKG2D作为CAR分子的胞外识别结构域,可以识别肿瘤细胞表面的多种靶点抗原(包括MICA、MICB、ULBP1、ULBP2、ULBP3、ULBP4、ULBP5、ULBP6),可降低因肿瘤异质性或抗原丢失而降低疗效的风险,更有利于提高对结直肠癌、卵巢癌、胰腺癌等恶性肿瘤的治疗效果。因此,本发明进一步提升了治疗的有效性,提高了CAR-T细胞等免疫细胞对抗恶性肿瘤的高度异质性的能力。同时,研究还表明NKG2D CAR-T也会靶向肿瘤微环境中的免疫抑制细胞和新生血管,有助于T细胞等免疫细胞克服免疫抑制的肿瘤微环境,提高肿瘤治疗的效 果。On the other hand, the present invention uses NKG2D as the extracellular recognition domain of the CAR molecule, which can recognize various target antigens on the surface of tumor cells (including MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5, ULBP6), and can reduce The risk of reduced efficacy due to tumor heterogeneity or antigen loss is more conducive to improving the therapeutic effect on malignant tumors such as colorectal cancer, ovarian cancer, and pancreatic cancer. Therefore, the present invention further improves the effectiveness of treatment, and improves the ability of immune cells such as CAR-T cells to fight against the high heterogeneity of malignant tumors. At the same time, studies have also shown that NKG2D CAR-T will also target immunosuppressive cells and new blood vessels in the tumor microenvironment, helping immune cells such as T cells overcome the immunosuppressive tumor microenvironment and improve the effect of tumor treatment.
再一方面,本发明采用CD40L作为第二辅助因子,以便更有效地激活机体内源的天然和适应性免疫应答。通过CD40L激活T细胞的增殖和细胞因子的释放,诱导M2谱系的巨噬细胞向具有抗肿瘤活性的M1谱系的巨噬细胞转变等,同时提高T细胞对某些抗原丢失但CD40高表达的肿瘤细胞的杀伤作用。此外,出乎意料的是,在本发明中,当本发明NKG2D CAR分子、外源CCR2b蛋白和外源CD40L蛋白的联合表达时,可协同地显著提高针对肿瘤细胞的体外杀伤作用。In yet another aspect, the present invention uses CD40L as a second cofactor to more effectively activate the body's endogenous natural and adaptive immune responses. Activate the proliferation of T cells and the release of cytokines through CD40L, induce the transformation of M2 lineage macrophages into M1 lineage macrophages with anti-tumor activity, etc., and at the same time improve the ability of T cells to respond to tumors with certain antigen loss but high CD40 expression cell killing. In addition, unexpectedly, in the present invention, when the NKG2D CAR molecule of the present invention, exogenous CCR2b protein and exogenous CD40L protein are jointly expressed, they can synergistically significantly improve the in vitro killing effect on tumor cells.
综上,本发明提供一种新颖的更高效的工程化免疫细胞(如CAR-T细胞),它可对结直肠癌、卵巢癌、胰腺癌等恶性实体肿瘤进行高效趋化迁移,可有效激活内源的免疫系统,并有效对抗肿瘤异质性的CAR-T细胞。In summary, the present invention provides a novel and more efficient engineered immune cell (such as CAR-T cell), which can efficiently chemotaxis and migrate to malignant solid tumors such as colorectal cancer, ovarian cancer, and pancreatic cancer, and can effectively activate Endogenous immune system, and CAR-T cells that can effectively fight against tumor heterogeneity.
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。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 teachings of the present invention, those skilled in the art can 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 (15)

  1. 一种工程化免疫细胞,其特征在于,所述工程化免疫细胞为T细胞或NK细胞,并且所述的免疫细胞具有以下特征:An engineered immune cell, characterized in that the engineered immune cell is a T cell or NK cell, and the immune cell has the following characteristics:
    (a)所述免疫细胞表达嵌合抗原受体(chimeric antigen receptor,CAR),其中所述CAR靶向肿瘤细胞的表面标志物,其中所述CAR的抗原结合结构域包括NKG2D的胞外结构域;(a) the immune cell expresses a chimeric antigen receptor (chimeric antigen receptor, CAR), wherein the CAR targets surface markers of tumor cells, wherein the antigen-binding domain of the CAR includes the extracellular domain of NKG2D ;
    (b)所述的免疫细胞表达外源的CCR2b蛋白;和(b) said immune cell expresses an exogenous CCR2b protein; and
    (c)所述的免疫细胞表达外源的CD40L蛋白。(c) The immune cells express exogenous CD40L protein.
  2. 如权利要求1所述的工程化免疫细胞,其特征在于,所述CAR的结构如式I所示:The engineered immune cell according to claim 1, wherein the structure of the CAR is as shown in formula I:
    L-NKG2D-H-TM-C-CD3ζ  (I)L-NKG2D-H-TM-C-CD3ζ (I)
    式中,In the formula,
    L为无或信号肽序列;L is nothing or a signal peptide sequence;
    NKG2D为NKG2D胞外结构域或其活性片段;NKG2D is an NKG2D extracellular domain or an active fragment thereof;
    H为无或铰链区;H is none or hinge region;
    TM为跨膜结构域;TM is the transmembrane domain;
    C为共刺激信号结构域;C is costimulatory signal domain;
    CD3ζ为源于CD3ζ的胞浆信号传导序列;CD3ζ is a cytoplasmic signaling sequence derived from CD3ζ;
    所述“-”为连接肽或肽键。The "-" is a connecting peptide or a peptide bond.
  3. 如权利要求1所述的工程化免疫细胞,其特征在于,除了式I所示的第一CAR之外,所述CAR细胞还含有用于针对第二抗原的第二CAR,所述的第二CAR的结构如式II所示:The engineered immune cell according to claim 1, characterized in that, in addition to the first CAR shown in formula I, the CAR cell also contains a second CAR for the second antigen, the second CAR The structure of CAR is shown in Formula II:
    L-scFv-H-TM-C-CD3ζ   (II)L-scFv-H-TM-C-CD3ζ (II)
    式中,In the formula,
    L为无或信号肽序列;L is nothing or a signal peptide sequence;
    scFv为抗原结合结构域;scFv is an antigen-binding domain;
    H为无或铰链区;H is none or hinge region;
    TM为跨膜结构域;TM is the transmembrane domain;
    C为共刺激结构域;C is costimulatory domain;
    CD3ζ为源于CD3ζ的胞浆信号传导序列或其突变/修饰体;CD3ζ is a cytoplasmic signaling sequence derived from CD3ζ or a mutation/modification thereof;
    所述“-”为连接肽或肽键。The "-" is a connecting peptide or a peptide bond.
  4. 如权利要求1所述的工程化免疫细胞,其特征在于,所述NKG2D的胞外结构域的氨基酸序列如SEQ ID NO:1第73-216位所示。The engineered immune cell according to claim 1, wherein the amino acid sequence of the extracellular domain of the NKG2D is shown in positions 73-216 of SEQ ID NO:1.
  5. 如权利要求1所述的工程化免疫细胞,其特征在于,所述CCR2b蛋白的氨 基酸序列如SEQ ID NO:2所示;和/或The engineered immune cell according to claim 1, wherein the amino acid sequence of the CCR2b protein is as shown in SEQ ID NO: 2; and/or
    所述CD40L蛋白的氨基酸序列如SEQ ID NO:5所示。The amino acid sequence of the CD40L protein is shown in SEQ ID NO:5.
  6. 如权利要求1所述的工程化免疫细胞,其特征在于,式I所示的第一CAR和式II所示的第二CAR可合而为一,从而构成如式IIIa或IIIb所示的CAR:The engineered immune cell according to claim 1, wherein the first CAR shown in formula I and the second CAR shown in formula II can be combined into one to form a CAR shown in formula IIIa or IIIb :
    L-NKG2D-scFv-H-TM-C-CD3ζ   (IIIa)L-NKG2D-scFv-H-TM-C-CD3ζ (IIIa)
    L-scFv-NKG2D-H-TM-C-CD3ζ   (IIIb)L-scFv-NKG2D-H-TM-C-CD3ζ (IIIb)
    式中,In the formula,
    L为无或信号肽序列;L is nothing or a signal peptide sequence;
    NKG2D为NKG2D胞外结构域或其活性片段;NKG2D is an NKG2D extracellular domain or an active fragment thereof;
    scFv为抗原结合结构域;scFv is an antigen-binding domain;
    H为无或铰链区;H is none or hinge region;
    TM为跨膜结构域;TM is the transmembrane domain;
    C为共刺激结构域;C is costimulatory domain;
    CD3ζ为源于CD3ζ的胞浆信号传导序列或其突变/修饰体;CD3ζ is a cytoplasmic signaling sequence derived from CD3ζ or a mutation/modification thereof;
    所述“-”为连接肽或肽键。The "-" is a connecting peptide or a peptide bond.
  7. 一种制备权利要求1所述的工程化免疫细胞的方法,其特征在于,包括以下步骤:A method for preparing the engineered immune cell according to claim 1, comprising the following steps:
    (A)提供一待改造的免疫细胞;和(A) providing an immune cell to be modified; and
    (B)对所述的免疫细胞进行改造,从而使得所述的免疫细胞表达所述的CAR分子以及外源的CCR2b蛋白和外源的CD40L蛋白,从而获得权利要求1所述的工程化免疫细胞。(B) transforming the immune cell so that the immune cell expresses the CAR molecule and exogenous CCR2b protein and exogenous CD40L protein, thereby obtaining the engineered immune cell according to claim 1 .
  8. 如权利要求7所述的方法,其特征在于,在步骤(B)中,包括:The method according to claim 7, characterized in that, in step (B), comprising:
    (B1)将表达所述CAR的第一表达盒导入所述免疫细胞;(B2)将表达CCR2b的第二表达盒导入所述免疫细胞;(B3)将表达CD40L的第三表达盒导入所述免疫细胞;其中所述的步骤(B1)、(B2)和(B3)可以按任意次序进行。(B1) introducing the first expression cassette expressing the CAR into the immune cells; (B2) introducing the second expression cassette expressing CCR2b into the immune cells; (B3) introducing the third expression cassette expressing CD40L into the immune cells Immune cells; wherein the steps (B1), (B2) and (B3) can be performed in any order.
  9. 一种制剂,其特征在于,所述制剂含有权利要求1所述的工程化免疫细胞,以及药学上可接受的载体、稀释剂或赋形剂。A preparation, characterized in that the preparation contains the engineered immune cell according to claim 1, and a pharmaceutically acceptable carrier, diluent or excipient.
  10. 如权利要求9所述的制剂,其特征在于,所述制剂含有本发明所述的CAR-T细胞,以及药学上可接受的载体、稀释剂或赋形剂。The preparation according to claim 9, wherein the preparation contains the CAR-T cells of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient.
  11. 一种如权利要求1所述的工程化免疫细胞的用途,其特征在于,用于制备可预防和/或治疗癌症的药物或制剂。A use of the engineered immune cell according to claim 1, characterized in that it is used to prepare a drug or preparation that can prevent and/or treat cancer.
  12. 如权利要求11所述的用途,其特征在于,所述肿瘤选自下组:结肠癌、直肠癌、卵巢癌、或胰腺癌。The use according to claim 11, wherein the tumor is selected from the group consisting of colon cancer, rectal cancer, ovarian cancer, or pancreatic cancer.
  13. 如权利要求11所述的用途,其特征在于,所述肿瘤为NKG2D配体高表达和/或趋化因子高表达和/或CD40高表达的肿瘤。The use according to claim 11, characterized in that the tumor is a tumor with high expression of NKG2D ligand and/or high expression of chemokines and/or high expression of CD40.
  14. 一种用于制备权利要求1所述的工程化免疫细胞的试剂盒,其特征在于,所述试剂盒含有容器,以及位于容器内的:A kit for preparing the engineered immune cell according to claim 1, characterized in that the kit contains a container, and in the container:
    (1)第一核酸序列,所述第一核酸序列含有用于表达所述CAR的第一表达盒,其中所述CAR的抗原结合结构域为NKG2D的胞外结构域;(1) a first nucleic acid sequence containing a first expression cassette for expressing the CAR, wherein the antigen-binding domain of the CAR is the extracellular domain of NKG2D;
    (2)第二核酸序列,所述第二核酸序列含有用于联合表达CCR2b的第二表达盒;和(2) a second nucleic acid sequence containing a second expression cassette for the combined expression of CCR2b; and
    (3)第三核酸序列,所述第三核酸序列含有用于联合表达CD40L的第三表达盒。(3) A third nucleic acid sequence containing a third expression cassette for co-expressing CD40L.
  15. 一种肿瘤的治疗方法,其特征在于,包括步骤:将安全有效量的如权利要求9所述的制剂,施用于所需对象,从而治疗所述对象的肿瘤。A method for treating tumors, comprising the step of: administering a safe and effective amount of the preparation according to claim 9 to a subject in need, thereby treating the subject's tumor.
PCT/CN2022/130696 2021-11-10 2022-11-08 Engineered immune cell for combined expression of ccr2b and cd40l, and preparation and application thereof WO2023083192A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111322947.0A CN113755448B (en) 2021-11-10 2021-11-10 Engineered immune cells jointly expressing CCR2b and CD40L, and preparation and application thereof
CN202111322947.0 2021-11-10

Publications (1)

Publication Number Publication Date
WO2023083192A1 true WO2023083192A1 (en) 2023-05-19

Family

ID=78784861

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/130696 WO2023083192A1 (en) 2021-11-10 2022-11-08 Engineered immune cell for combined expression of ccr2b and cd40l, and preparation and application thereof

Country Status (2)

Country Link
CN (1) CN113755448B (en)
WO (1) WO2023083192A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113755448B (en) * 2021-11-10 2022-02-18 广州百吉生物制药有限公司 Engineered immune cells jointly expressing CCR2b and CD40L, and preparation and application thereof
WO2024041618A1 (en) * 2022-08-25 2024-02-29 广州百吉生物制药有限公司 Engineered immune cell co-expressing cd40l, preparation therefor, and use thereof
CN117625547A (en) * 2022-08-25 2024-03-01 广州百吉生物制药有限公司 New generation chimeric antigen receptor for combined expression of GPX4 and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019062817A1 (en) * 2017-09-27 2019-04-04 亘喜生物科技(上海)有限公司 Engineered immune cell capable of inducing secretion of anti-cd47 antibody
CN113621582A (en) * 2021-09-23 2021-11-09 广州百吉生物制药有限公司 Engineered immune cell for combined expression of CCR2b, and preparation and application thereof
CN113755448A (en) * 2021-11-10 2021-12-07 广州百吉生物制药有限公司 Engineered immune cells jointly expressing CCR2b and CD40L, and preparation and application thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2019291627A1 (en) * 2018-06-19 2020-10-08 Nanjing Legend Biotech Co., Ltd. Engineered cells and uses thereof
CN111378625A (en) * 2020-03-18 2020-07-07 华东师范大学 Preparation and application of CXCL13 chemotactic CAR-T cell
CN112961248B (en) * 2021-02-22 2022-03-18 广州百暨基因科技有限公司 Chimeric antigen receptor fusion protein for coexpression of IL-7 and CCR2b and application thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019062817A1 (en) * 2017-09-27 2019-04-04 亘喜生物科技(上海)有限公司 Engineered immune cell capable of inducing secretion of anti-cd47 antibody
CN113621582A (en) * 2021-09-23 2021-11-09 广州百吉生物制药有限公司 Engineered immune cell for combined expression of CCR2b, and preparation and application thereof
CN113755448A (en) * 2021-11-10 2021-12-07 广州百吉生物制药有限公司 Engineered immune cells jointly expressing CCR2b and CD40L, and preparation and application thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
KEVIN J CURRAN: "Enhancing antitumor efficacy of chimeric antigen receptor T cells through constitutive CD40L expression", THE AMERICAN SOCIETY OF GENE & CELL THERAPY, vol. 23, no. 4, 10 February 2015 (2015-02-10), XP002789068, ISSN: 2626-7721, DOI: 10.1038/mt.2015.4 *
NICHOLAS F. KUHN ET AL.: "CD40 Ligand-Modified Chimeric Antigen Receptor T Cells Enhance CD40 Ligand-Modified Chimeric Antigen Receptor T Cells Enhance Antitumor Function by Eliciting an Endogenous Antitumor Response", CANCER CELL, vol. 35, no. 3, 18 March 2019 (2019-03-18), XP085638453, ISSN: 1535-6108, DOI: 10.1016/j.ccell.2019.02.006 *
PAN, ZEYAN ET AL.: "Current Status and Future Strategies of Chimeric Antigen Receptor T-cells in the Treatment of Pancreatic Cancer", CHINA ONCOLOGY, vol. 29, no. 7, 10 February 2015 (2015-02-10), XP093072457, ISSN: 1007-3639 *

Also Published As

Publication number Publication date
CN113755448B (en) 2022-02-18
CN113755448A (en) 2021-12-07

Similar Documents

Publication Publication Date Title
US10780120B2 (en) Prostate-specific membrane antigen cars and methods of use thereof
WO2023046110A1 (en) Engineered immune cell co-expressing ccr2b, preparation therefor and application thereof
CN110950953B (en) Monoclonal antibody against B7-H3 and application thereof in cell therapy
WO2021098882A1 (en) Cd7-car-t cell and preparation and application thereof
WO2019196713A1 (en) Bcma-targeted chimeric antigen receptor as well as preparation method therefor and application thereof
WO2023083192A1 (en) Engineered immune cell for combined expression of ccr2b and cd40l, and preparation and application thereof
WO2018145649A1 (en) Construction of chimeric antigen receptor targeting cd20 antigen and activity identification of engineered t cells thereof
US20240082401A1 (en) Bispecific cs1-bcma car-t cell and application thereof
EP3517125B1 (en) Chimeric antigen receptor for efficient targeted proliferation in vitro and uses thereof
CN109575143B (en) Bispecific CD20-CD19-CAR and application thereof
CN113087806B (en) Novel CAR-T cells targeting multiple tumors, and preparation and methods thereof
WO2018145648A1 (en) Cd20 targeting car construction and activity identification of engineered t cells thereof
CN113784732A (en) BCMA-targeted engineered immune cells and uses thereof
CN111378625A (en) Preparation and application of CXCL13 chemotactic CAR-T cell
CN107936120B (en) CD19 targeted chimeric antigen receptor and preparation method and application thereof
WO2023051735A1 (en) Chimeric antigen receptor immune cell, and preparation method therefor and application thereof
CN113330038A (en) CD20 combination targeted engineered immune cells
WO2021208750A1 (en) Cd22-targeted chimeric antigen receptor, preparation method therefor and application thereof
CN114853893A (en) Target GPC3 chimeric antigen receptor T cell and application thereof
JP2023522112A (en) Technology for producing versatile humanized CAR19-DNT cells and use thereof
WO2024041618A1 (en) Engineered immune cell co-expressing cd40l, preparation therefor, and use thereof
CN116496397B (en) Humanized antibodies targeting CD19CAR-T cells
CN115975056B (en) Construction of TSHR-targeted CAR-T cells using the native protein TSH as an antigen binding site
CN116444669B (en) Humanized antibodies targeting BCMA CAR-T cells
CN117940559A (en) Engineered immune cells for combined expression of CD40L, and preparation and application thereof

Legal Events

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

Ref document number: 22891986

Country of ref document: EP

Kind code of ref document: A1