WO2017107353A1 - Cancer treatment agent, preparation method and use thereof employing il-12 with stable membrane expression - Google Patents

Cancer treatment agent, preparation method and use thereof employing il-12 with stable membrane expression Download PDF

Info

Publication number
WO2017107353A1
WO2017107353A1 PCT/CN2016/080418 CN2016080418W WO2017107353A1 WO 2017107353 A1 WO2017107353 A1 WO 2017107353A1 CN 2016080418 W CN2016080418 W CN 2016080418W WO 2017107353 A1 WO2017107353 A1 WO 2017107353A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
protein
cd62l
fusion protein
cell
Prior art date
Application number
PCT/CN2016/080418
Other languages
French (fr)
Chinese (zh)
Inventor
杨世成
Original Assignee
杨世成
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 杨世成 filed Critical 杨世成
Publication of WO2017107353A1 publication Critical patent/WO2017107353A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5434IL-12
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • 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
    • C07K14/7056Lectin superfamily, e.g. CD23, CD72
    • C07K14/70564Selectins, e.g. CD62
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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

Definitions

  • the invention relates to the field of medicine, in particular to a high-efficiency and low-toxic tumor therapeutic agent based on IL-12 stable membrane expression, a preparation method thereof and use thereof.
  • IL-12 is a very important immune stimulating factor.
  • IL-12 is a heterodimeric cytokine linked by a covalent bond, consisting of the p35 and p40 subunits, secreted by activated immune cells in vivo.
  • IL-12 is an important cellular immune regulatory factor that acts through NK cells and CTLs in immunity against infection and malignancy.
  • IL-12 has a significant antitumor effect, it is extremely limited due to systemic systemic administration which causes systemic toxic side effects.
  • Prior to the present invention the clinical application of IL-12 was greatly limited in clinical application due to the systemic toxic side effects induced by it and the accidental death of two patients caused by systemic administration in early clinical trials.
  • the present inventors have attempted to maximize the antitumor effect by genetically modifying anti-tumor T cells to continuously secrete IL-12, but for unknown reasons (a possible cause is the side effects of IL-12) T cells that have been genetically modified by sustained secretion of the IL-12 gene are not efficiently amplified in vitro and cause a large number of T cell apoptosis.
  • the object of the present invention is to provide a tumor therapeutic drug with high efficiency and low toxic side effects, and a preparation method and application thereof.
  • a fusion protein comprising the following elements fused together:
  • signal peptide is operably linked to the fusion element consisting of (ii), (iii) and (iv);
  • the first protein element is an IL-12 protein element
  • the second protein element is a protein element of the mutant CD62L
  • the protein element of the mutant CD62L lacks a cleavage site of ADAM17.
  • the fusion protein has a structure selected from the group consisting of:
  • A is an IL-12 protein element
  • B is a mutant CD62L protein element
  • C is an optional linker element
  • D is an optional signal peptide signal peptide and/or leader peptide sequence
  • Each "-" independently indicates a peptide bond or a peptide linker to which the above elements are attached.
  • the protein element of the mutant CD62L lacks a partial or total sequence in the KLDKSFS sequence, resulting in the inability to be cleaved by ADAM17.
  • operably linked means that the signal peptide can direct expression or transmembrane transfer (localization) of the fusion element.
  • the linker peptide element comprises a linker peptide having the sequence set forth in SEQ ID NO.: 7.
  • the IL-12 protein is derived from a human or a non-human mammal.
  • the IL-12 protein comprises wild type and mutant form.
  • the IL-12 protein comprises a full length, mature form of IL-12, or an active fragment thereof.
  • the first protein element comprises one or two subunits of an IL-12 protein.
  • the subunit of the IL-12 protein is selected from the group consisting of the P40 and P35 subunits.
  • the first protein element comprises the IL-12 protein P40 and P35 subunits joined together.
  • the P40 and P35 subunits are "head-to-head”, “head-to-tail”, and “tail-tail”.
  • a linker is present or absent between the P40 and P35 subunits.
  • the linker is a flexible linker of 4-20 amino acids, more preferably, the linker is GGGGGGS (i.e., G 6 S) (SEQ ID NO.:8 ).
  • sequence of the IL12 protein element is set forth in SEQ ID NO.: 4.
  • the K283-S284 cleavage site is absent from the mutant CD62L protein element.
  • the mutant CD62L protein is derived from a human or non-human mammal.
  • the mutant CD62L protein comprises a full length, mature form of CD62L, or an active fragment thereof.
  • sequence of the mutant CD62L protein element is set forth in SEQ ID NO.: 6.
  • the peptide linker is 0-15 amino acids in length, preferably 1-10 amino acids.
  • the fusion protein further comprises a signal peptide element D.
  • the fusion protein is provided with a linker peptide, preferably a 218 peptide segment, between the single-stranded IL-12 (first protein element) and the mutant CD62L (second protein element) (SEQ ID NO.: 7).
  • a linker peptide preferably a 218 peptide segment, between the single-stranded IL-12 (first protein element) and the mutant CD62L (second protein element) (SEQ ID NO.: 7).
  • the first protein element in the fusion protein, is a single-stranded IL-12, and in the single-stranded IL-12, a linker peptide G 6 S is provided in the P40 subunit and the P35 subunit. (SEQ ID NO.: 8).
  • amino acid sequence of the fusion protein is shown in SEQ ID NO.: 2.
  • the fusion protein has the following characteristics:
  • the fusion protein is not cleaved by the ADAM17 protein, thereby not releasing IL-12;
  • the fusion protein comprises two subunits of IL-12, namely the P40 and P35 subunits, and is joined by GGGGGGS (G6S).
  • the fusion protein is a monomer, or a dimer.
  • an isolated polynucleotide encoding the fusion protein of the first aspect of the invention.
  • sequence of the polynucleotide is set forth in SEQ ID NO.: 1.
  • a vector comprising the polynucleotide of the second aspect of the invention is provided.
  • the vector comprises a plasmid, a viral vector.
  • the viral vector comprises a lentiviral vector, an adenoviral vector, and a yellow fever virus vector.
  • the vector comprises an expression vector.
  • a host cell comprising the polynucleotide of the second aspect of the invention, or a polynucleotide of the second aspect of the invention, is provided.
  • the host cell comprises a prokaryotic cell and a eukaryotic cell.
  • the host cell comprises a mammalian cell.
  • the host cell comprises an immune cell, preferably a T cell,
  • an immune cell which carries the fusion protein of the first aspect of the invention on the surface of the membrane.
  • the immune cells of at least 10 3 (preferably 103 to 109, more preferably preferably 104-108 th) the cell population of immune cells.
  • all or most (> 80%, preferably > 90%) of the cells of the immune cells or population of immune cells are viable.
  • At least a portion or all of said fusion protein is located on the cell membrane of said immune cell, and said first protein element, i.e., the IL-12 protein element, is located extracellularly.
  • the immune cells comprise T cells.
  • the T cell surface carries a MART-1 TCR.
  • composition comprising:
  • a pharmaceutically acceptable carrier is selected from:
  • the pharmaceutical composition is in a liquid state.
  • the pharmaceutical composition contains 1 x 10 3 - 1 x 10 7 of said immune cells/ml.
  • fusion protein of the first aspect of the invention and/or the immune cell of the sixth aspect of the invention which is used for the preparation of a medicament for treating a tumor.
  • the tumor comprises: a brain tumor, a colorectal cancer tumor, a lung cancer tumor, a liver cancer tumor, a breast cancer tumor, a gastric cancer tumor, and a pancreatic cancer tumor.
  • a method of treating a tumor comprising the step of administering to a subject in need thereof the fusion protein of the first aspect of the invention and/or the immune cell of the sixth aspect of the invention.
  • the fusion protein is administered as a monomer and/or a dimer.
  • the object is a human.
  • a non-therapeutic or therapeutic method for killing tumor cells in vitro comprising the steps of: contacting the immune cells of the sixth aspect of the invention with tumor cells, thereby killing The tumor cells are destroyed.
  • Figure 1 shows the amino acid composition of the CD62L transmembrane region and the cleavage site of ADAM17.
  • 1A and 1B show that the dKSCD62L mutant (with KLDKSFS deleted) cannot undergo cleavage and release based on a cleavage enzyme (such as Adam17) after T cell activation, resulting in dKSCD62L remaining on the surface of the T cell membrane.
  • WT represents the wild type sequence
  • dKS represents the CD62L mutant knocked out of the ADAM17 sequence.
  • Figure 2 shows that dKSCD62L lentiviral gene-modified anti-tumor T cells result in stable expression of dKSCD62L.
  • the anti-tumor T cell line JKF6 (an anti-tumor T cell line isolated and cultured from melanoma tissue can directly recognize the tumor) can be detected by lentiviral gene, and the expression of WTCD62L and dKSCD62L can be detected.
  • CD62L can undergo tumor antigen-specific cleavage and release after T (wild-type) CD62L is activated by tumor antigen 526.
  • T wild-type
  • dKSCD62L membrane after expression of dKSCD62L membrane, it loses the cleavage and release of tumor antigen activation.
  • the phenotype of expression of the stable membrane surface dKSCD62L after expression of dKSCD62L membrane, it loses the cleavage and release of tumor antigen activation.
  • JKF6T cells were co-cultured with tumor cells 938 and 526 expressing the MART-1 antigen. Among them, co-cultured T cells were detected by flow cytometry on the surface of the membrane CD45RO and CD62L, and analyzed by FlowJo software, each group of T cells were sorted into CD8 cell subsets.
  • the antibodies used for flow cytometry were CD62L FITC, CD45RO APC, MART-1PE and CD8PerCP.
  • Figure 3 shows that dKSCD62L lentiviral gene-modified anti-tumor JFK6 cells do not affect the anti-tumor activity of T cells.
  • FIG. 3A shows the statistical analysis treatment of IFNy and CD62L expression.
  • the data are analytical results of 3 independent experiments.
  • the expression level of dKSCD62L is consistently stable, and IFN ⁇ is similar to other groups, showing similar high levels. Results are expressed as mean ⁇ standard deviation, and T-test was used for statistical processing. * indicates that dKSCD62L is compared with the other two groups of CD62L expression levels, P ⁇ 0.01.
  • Figure 4 shows the construction of a lentiviral vector expressing dKSCD62L fusion protein and its expression in human T cells.
  • the lentivirus is a third generation lentiviral vector and the promoter is MSCV.
  • the 5' and 3' LTR (long terminal repeat) of this vector was engineered into SIN-LTR (self-inactivating-LTR) in order to reduce the probability of lentiviral recombination and enhance safety performance.
  • the above figure shows the structural diagram of the LVV plasmid and the gene expression constructs of human IL-12/WTCD62L (WT, wild type), IL-12/dKSCD62L and IL-12 (human single chain IL-12), wherein hscIL-12
  • WT wild type
  • IL-12/dKSCD62L human single chain IL-12
  • hscIL-12 human single chain IL-12
  • the LE-12 seq is fused to a lentiviral expression vector by gene cloning, and CD62L or dKSCD62L and hscIL-12 are linked by a peptide G 6 S.
  • T cells pass two transductions, namely MART-1 TCR (first day) and three in the illustration.
  • the cells were sequentially transduced (Day 5). On the 14th day, they were co-cultured with 526 and 938 in a 1:1 ratio for 4 hours, and then the expression of hscIL-12 on the surface of T cell membrane was detected by flow cytometry.
  • Figure 5 shows that co-culture of lentiviral vector IL-12/mutant CD62L-transduced T cells with tumor cells enhances IFNy expression and tumor antigen-dependent release of IL-12.
  • the mock is a blank control group and the tumor is a tumor group.
  • T cells were sequentially transduced with anti-tumor TCR and IL-12/mutant CD62L series fusion proteins for 14 days, and co-cultured with tumor cells 526 and 938. After 16 hours, the expression levels of IFN ⁇ and IL-12 in the supernatant were passed through ELISA kit. Detection. The results showed that the sustained IL-12 secretion group, IL-12/WTCD62L group and IL-12/dKSCD62L group could significantly enhance the reactivity of anti-tumor T cells with tumors compared with the T cell group, which showed a significant enhancement of IFN ⁇ secretion. Level, P ⁇ 0.001. The stable expression on the surface of dKSCD62L membrane was consistent.
  • IL-12/dKSCD62L also lost the cleavage and release of tumor antigen reactivity. Compared with IL-12/WTCD62L group and persistent secretion IL-12 group, the secreted IL- could be detected. The level of 12 was significantly reduced, P ⁇ 0.001.
  • Figure 6 shows that the lentiviral vector IL-12/dKSCD62L series transduced T cells can effectively avoid the toxic side effects of in vitro T cell expansion caused by persistent secretion of IL-12.
  • T cells were transduced with anti-tumor TCR and IL-12/CD62L series fusion proteins for 14 days, the expansion ratios of the cells in each group were compared with the control transduction group (T-cell), and the transduction of IL-12 group was compared with other groups.
  • the amplification factor of the group was significantly reduced, p ⁇ 0.001.
  • the transduction IL-12/dKSCD62L fusion protein group was the same as the other groups, and no significant difference was observed.
  • the data analysis was performed by setting the amplification factor of the T-cell group to 100%, and the values of the other groups were compared with the T-cell group.
  • Figure 7 shows that lentiviral vector IL-12/dLSCD62L series transduced murine T cell-mediated cell transfusion therapy significantly prolonged survival of tumor-bearing mice.
  • mice Female pmel mice (7 mice per group) were implanted intracranial (IC) for 5 days by B16F10 cells (5000 cells/only), and the mice received 5 Gy whole body radiotherapy 1 day before cell return.
  • Mouse T cells were obtained from mouse spleen cells and activated by 10 ug/ml concanavalin (Con A) in the presence of IL-2 (5 IU/ml); on day 2, transduction of T with lentiviral vector The cells were then cultured for 6 days, cells were collected, and (X) 5 ⁇ 10 6 T cells were injected through the tail vein of the mice.
  • the DC cells of the DC group were obtained from bone marrow cells, and induced to differentiate and mature for 8 days in vitro, and 1 ⁇ 10 6 cells were inoculated intraperitoneally. The group description of DC and T cells is shown on the right. An asterisk (*) indicates that the experimental group was compared with the other groups, p ⁇ 0.001.
  • the fusion protein of the present invention is a fusion protein of IL-12/mutant CD62L, wherein the mutant CD62L lacks the cleavage site of ADAM17.
  • the fusion protein of the present invention is expressed in T cells, it is effectively displayed in T The surface of the cell.
  • the IL-12/mutant CD62L fusion protein has little effect on the viability of T cells, and the anti-tumor T cells carrying the fusion protein of the present invention are unable to pass specificity when approaching tumor cells.
  • CD62L is cleaved to release IL-12, resulting in IL-12 on the cell membrane surface acting more efficiently and safely on tumor cells.
  • the T cell-targeted tumor is localized and the immune micro-environment of the T cell-tumor tissue is changed by the IL-12 on the cell surface, thereby synergistically and effectively achieving the maximization of the anti-tumor immunity effect, and extremely significantly reducing The toxic side effects of IL-12.
  • the present invention has been completed on this basis.
  • the present inventors developed a novel fusion protein of IL-12/dKSCD62L using a mutant CD62L (dKSCD62L) lacking the cleavage site of ADAM17, and modified anti-tumor T cells by a lentiviral gene.
  • the experimental data show that IL-12/dKSCD62L lentiviral gene-modified anti-tumor T cells, the expression of membrane surface is not regulated by T cell activation, can maintain high membrane surface expression, and enhance the response to tumor antigens. The effect of the immune response.
  • the preclinical tumor-bearing mouse model showed that the modified T cells could significantly prolong the survival of tumor-bearing mice, and no obvious cytotoxic effects were found.
  • the T cells expressing the fusion protein by the membrane are effective in killing tumor cells on the one hand, and have been confirmed to have safety in vitro and in vivo on the other hand.
  • the modified T cells themselves appear to be only slightly adversely affected or substantially unaffected by the expressed IL-12 (in vitro, the fusion protein of the invention is directed against T cells compared to T cells that continue to secrete IL-12) Amplification has no significant effect).
  • head refers to the N-terminus of a polypeptide or fragment thereof, particularly the N-terminus of a wild-type polypeptide or fragment thereof.
  • tail refers to the C-terminus of a polypeptide or fragment thereof, particularly the C-terminus of a wild-type polypeptide or fragment thereof.
  • containing includes “including”, “consisting essentially of”, “consisting essentially of”, and “consisting of”;
  • the subordinate concepts of “consisting of”, “consisting essentially of” and “consisting of” are “contained,” “having,” or “including.”
  • CD62L is widely expressed on the surface of T cells and is an important immunoregulatory factor. It can regulate the migration of T cells to the lymph nodes of the whole body and is an important T cell homing factor.
  • CD62L+T cell migrates to the lymph node and is exposed to the tumor antigen, CD62L can be tested for tumor antigen reactivity by ADAM17 on the peptide K283-S284 at the transmembrane region.
  • CD62L cleavage has the specificity of a tumor antigen reaction accompanied by CD107a (the molecule is a molecular protein on the lysosome, which is normally present in the cell)
  • the molecule migrates to the cell surface accompanied by degranulation of T cells; therefore, membrane surface detection of this molecule is an important indicator of membrane migration of T cell killing.
  • mutant CD62L refers to a CD62L polypeptide or active fragment thereof or a polypeptide derived therefrom which lacks a cleavage site for the cleavage enzyme ADAM17, i.e., a partial or total sequence of the KLDKSFS sequence is deleted, such that the CD62L polypeptide or The active fragment or derivative thereof cannot be cleaved by the cleavage enzyme ADAM17.
  • a preferred mutant CD62L is a CD62L polypeptide lacking the entire KLDKSFS sequence.
  • the CD62L may be from a mammal (human or non-human mammal) or may be derived from other eukaryotic species.
  • CD62L is wild type CD62L from human.
  • a particularly preferred mutant CD62L is dKSCD62L, a human CD62L polypeptide lacking the entire KLDKSFS sequence.
  • IL-12 is a very important immune stimulating factor.
  • IL-12 is a heterodimeric cytokine linked by a covalent bond, consisting of the p35 and p40 subunits, secreted by activated immune cells in vivo.
  • the IL-12 may be derived from a human or non-human mammal, and may be a full-length, mature form of IL-12, or an active fragment thereof.
  • the IL-12 (or IL-12 protein element) can be a single subunit or multiple subunits.
  • the first protein element may comprise one or more (e.g., two) subunits of an IL-12 protein.
  • the first protein element comprises the IL-12 protein P40 and P35 subunits joined together.
  • the manner of connecting the P40 and P35 subunits is not particularly limited, and includes "head-to-head”, “head-to-tail”, “tail-head”, and “tail-tail”, wherein “ “Head” refers to the N-terminus of a polypeptide, and “tail” refers to the C-terminus of a polypeptide.
  • a linker may or may not be present between the P40 and P35 subunits.
  • the linker is a flexible linker of 4-20 amino acids, more preferably, the linker is GGGGGGS (G 6 S) (SEQ ID NO.:8)
  • the bifunctional fusion proteins of the invention may optionally contain a peptide linker.
  • the size and complexity of the peptide linker may affect the activity of the protein.
  • peptide linkers should be of sufficient length and flexibility to ensure connectivity. The two proteins have enough freedom in space to perform their function. At the same time, the effect of the formation of an alpha helix or a beta sheet in the peptide linker on the stability of the fusion protein is avoided.
  • the length of the linker peptide is generally from 0 to 15 amino acids, preferably from 1 to 15 amino acids.
  • linker peptides include, but are not limited to, the linker peptide set forth in SEQ ID NO.: 7 or 8.
  • the fusion protein of the present invention may also contain other elements including, but not limited to, signal peptides, leader peptides and the like.
  • the fusion protein contains a signal peptide.
  • Representative examples include, but are not limited to, a signal peptide of the human IL-12P40 subunit.
  • the fusion protein is an isolated protein that is not associated with other proteins, polypeptides or molecules and is a product expressed by recombinant host cells, or isolated or purified.
  • “recombinant bifunctional fusion protein”, “protein of the present invention”, “fusion protein of the present invention”, “bifunctional fusion protein”, “IL-12-mutant CD62L fusion protein”, “IL-12/” “Mutant CD62L fusion protein” is used interchangeably and refers to a structure having the structure of Formula Ia, or the structure described in IIa, ie, a fusion protein comprising a protein element comprising a IL-12 protein element, a mutant CD62L, and a linker element.
  • a representative example is IL12-dKSCD62L.
  • the protein of the invention may be a monomer or a multimer (e.g., a dimer) formed from a monomer.
  • the term also encompasses active fragments and derivatives of fusion proteins.
  • isolated means that the substance is separated from its original environment (if it is a natural substance, the original environment is the natural environment).
  • the polynucleotides and polypeptides in the natural state in living cells are not isolated and purified, but the same polynucleotide or polypeptide is isolated and purified, as separated from other substances present in the natural state.
  • isolated recombinant fusion protein means that the recombinant fusion protein is substantially free of other proteins, lipids, carbohydrates or other materials with which it is naturally associated.
  • One skilled in the art can purify recombinant fusion proteins using standard protein purification techniques. Substantially pure proteins produce a single major band on a non-reducing polyacrylamide gel.
  • the polynucleotide of the present invention may be in the form of DNA or RNA.
  • DNA forms include cDNA, genomic DNA or synthetic DNA.
  • DNA can be single-stranded or double-stranded.
  • DNA can be a coding strand or Non-coding chain.
  • the present invention also relates to variants of the above polynucleotides which encode protein fragments, analogs and derivatives having the same amino acid sequence as the present invention.
  • Variants of this polynucleotide may be naturally occurring allelic variants or non-naturally occurring variants. These nucleotide variants include substitution variants, deletion variants, and insertion variants.
  • an allelic variant is an alternative form of a polynucleotide that may be a substitution, deletion or insertion of one or more nucleotides, but does not substantially alter the function of the polypeptide encoded thereby.
  • the term "primer” refers to a generic term for a oligodeoxynucleotide that, in pairing with a template, can be used to synthesize a DNA strand complementary to a template under the action of a DNA polymerase.
  • the primer may be native RNA, DNA, or any form of natural nucleotide.
  • the primer may even be a non-natural nucleotide such as LNA or ZNA.
  • the primer is “substantially” (or “substantially") complementary to a particular sequence on a strand on the template.
  • the primer must be sufficiently complementary to a strand on the template to initiate extension, but the sequence of the primer need not be fully complementary to the sequence of the template.
  • a sequence that is not complementary to the template is added to the 5' end of a primer complementary to the template at the 3' end, such primers are still substantially complementary to the template.
  • the non-fully complementary primers can also form a primer-template complex with the template for amplification.
  • the fusion protein of the present invention can conveniently prepare by various known methods. These methods are, for example but not limited to, recombinant DNA methods, artificial synthesis, and the like.
  • the full length nucleotide sequence of the element of the fusion protein of the present invention (e.g., IL12 or mutant CD62L) or a fragment thereof can be generally obtained by a PCR amplification method, a recombinant method or a synthetic method.
  • primers can be designed according to published nucleotide sequences, particularly open reading frame sequences, and used as commercially available cDNA libraries or cDNA libraries prepared by conventional methods known to those skilled in the art.
  • the template is amplified to obtain the relevant sequence. When the sequence is long, it is often necessary to perform two or more PCR amplifications, and then the amplified fragments are spliced together in the correct order.
  • the recombinant sequence can be used to obtain the relevant sequences in large quantities. This is usually done by cloning it into a vector, transferring it to a cell, and then isolating the relevant sequence from the proliferated host cell by conventional methods.
  • synthetic sequences can be used to synthesize related sequences, especially when the fragment length is short.
  • a long sequence of fragments can be obtained by first synthesizing a plurality of small fragments and then performing the ligation.
  • a method of amplifying DNA/RNA using PCR technology is preferably used to obtain the gene of the present invention.
  • the primers for PCR can be appropriately selected according to the sequence information of the present invention disclosed herein, and can be synthesized by a conventional method.
  • the amplified DNA/RNA fragment can be isolated and purified by conventional methods such as by gel electrophoresis.
  • the invention also relates to vectors comprising the polynucleotides of the invention, as well as host cells genetically engineered using the vector or fusion protein coding sequences of the invention, and methods of producing the proteins of the invention by recombinant techniques.
  • polynucleotide sequences of the present invention can be utilized to express or produce recombinant proteins by conventional recombinant DNA techniques. Generally there are the following steps:
  • Methods well known to those skilled in the art can be used to construct expression vectors containing the DNA sequences of the proteins of the invention and suitable transcription/translation control signals. These methods include in vitro recombinant DNA techniques, DNA synthesis techniques, in vivo recombinant techniques, and the like.
  • the DNA sequence can be operably linked to an appropriate promoter in an expression vector to direct mRNA synthesis.
  • the expression vector also includes a ribosome binding site for translation initiation and a transcription terminator.
  • the expression vector preferably comprises one or more selectable marker genes to provide phenotypic traits for selection of transformed host cells, such as dihydrofolate reductase for eukaryotic cell culture, neomycin resistance, and green Fluorescent protein (GFP), or tetracycline or ampicillin resistance for E. coli.
  • selectable marker genes to provide phenotypic traits for selection of transformed host cells, such as dihydrofolate reductase for eukaryotic cell culture, neomycin resistance, and green Fluorescent protein (GFP), or tetracycline or ampicillin resistance for E. coli.
  • Vectors comprising the appropriate DNA sequences described above, as well as appropriate promoters or control sequences, can be used to transform appropriate host cells to enable expression of the protein.
  • the host cell can be a prokaryotic cell, such as a bacterial cell; or a lower eukaryotic cell, such as a yeast cell; or a higher eukaryotic cell, such as a mammalian cell.
  • a prokaryotic cell such as a bacterial cell
  • a lower eukaryotic cell such as a yeast cell
  • a higher eukaryotic cell such as a mammalian cell.
  • Representative examples are: Escherichia coli, bacterial cells of the genus Streptomyces; fungal cells such as yeast; plant cells; insect cells of Drosophila S2 or Sf9; animal cells of CHO, COS, or 293 cells, and the like.
  • a particularly preferred cell is a cell of a human and a non-human mammal, especially an immune cell, including T cells, NK cells.
  • Transformation of host cells with recombinant DNA can be carried out using conventional techniques well known to those skilled in the art.
  • the host is a prokaryote such as E. coli
  • competent cells capable of absorbing DNA can be harvested after the exponential growth phase and treated by the CaCl 2 method, and the procedures used are well known in the art.
  • Another method is to use MgCl 2 .
  • Conversion can also be carried out by electroporation if desired.
  • the host is a eukaryote, the following DNA transfection methods can be used: calcium phosphate coprecipitation, conventional mechanical methods such as microinjection, electroporation, liposome packaging, and the like.
  • the obtained transformant can be cultured by a conventional method to express the polypeptide encoded by the gene of the present invention.
  • the medium used in the culture may be selected from various conventional media depending on the host cell used.
  • the cultivation is carried out under conditions suitable for the growth of the host cell.
  • the selected promoter is induced by a suitable method (such as temperature conversion or chemical induction) and the cells are cultured for a further period of time.
  • the protein in the above method can be expressed intracellularly, or on the cell membrane, or secreted outside the cell. If desired, the protein can be isolated and purified by various separation methods using its physical, chemical, and other properties. These methods are well known to those skilled in the art. Examples of these methods include but not Limited to: conventional renaturation treatment, treatment with protein precipitant (salting method), centrifugation, osmotic bacteria, super treatment, ultracentrifugation, molecular sieve chromatography (gel filtration), adsorption chromatography, ion exchange chromatography, high efficiency Liquid chromatography (HPLC) and various other liquid chromatography techniques and combinations of these methods.
  • conventional renaturation treatment treatment with protein precipitant (salting method), centrifugation, osmotic bacteria, super treatment, ultracentrifugation, molecular sieve chromatography (gel filtration), adsorption chromatography, ion exchange chromatography, high efficiency Liquid chromatography (HPLC) and various other
  • the present invention also provides an immune cell (abbreviated as "the immune cell of the present invention”) expressing the fusion protein of the present invention, which carries the fusion protein on the cell surface.
  • the immune cell of the present invention expresses the fusion protein of the present invention, which carries the fusion protein on the cell surface.
  • the fusion protein is located on the cell membrane of the immune cell, and the first protein element, i.e., the IL-12 protein element, is located extracellularly.
  • a preferred class of immune cells includes T cells, particularly human T cells.
  • said T cell surface carries a MART-1 TCR.
  • a lentiviral expression system genetically modified T cell which expresses an anti-tumor TCR (T-cell receptor) and simultaneously expresses an hscIL-12/mutant CD62L fusion protein.
  • T-cell receptor anti-tumor TCR
  • the T cells may optionally also express wild type CD62L, hscIL-12 (human single chain IL-12) or a combination thereof.
  • the mechanism of action of the membrane expressing dKSCD62L mutant is shown in Figure 1.
  • the CD62L transmembrane region contains the cleavage site of ADAM17, and in the case of T cell activation, ADAM7 can cleave and release CD62L.
  • the KLDKSFS sequence i.e., the cleavage site of ADAM17
  • the KLDKSFS sequence is knocked out by genetic engineering, thereby realizing the stable membrane expression of exogenous CD62L from a molecular mechanism.
  • a typical mutant CD62L is dKSCD62L.
  • T cells were activated (including tumor antigen-induced T cell activation), ADAM17 was unable to cleave dKSCD62L on the membrane surface.
  • the dKSCD62L mutant is not cleaved by the ADAM17 enzyme and cannot be released, resulting in the retention of dK-SCD62L on the surface of the T cell membrane, which in turn causes the IL-12 element in the fusion protein to remain on the surface of the T cell membrane.
  • the fusion protein retains both the biological activity of Il-12 and the function of T cells.
  • the invention also provides a composition
  • a composition comprising (a) an effective amount of a fusion protein of the invention and/or an effective amount of an immune cell of the invention, and a pharmaceutically acceptable carrier.
  • the fusion proteins of the invention can be formulated in non-toxic, inert and pharmaceutically acceptable water In a carrier medium, wherein the pH is usually from about 5 to about 8, preferably, the pH is from about 6 to about 8.
  • the term "effective amount” or “effective amount” refers to an amount that is functional or active to a human and/or animal and that is acceptable to humans and/or animals, such as from 0.001 to 99% by weight; preferably 0.01-95 wt%; more preferably, 0.1-90 wt%.
  • an effective amount or “effective amount” means 1 x 10 3 - 1 x 10 7 of said immune cells/ml.
  • a "pharmaceutically acceptable” ingredient is one that is suitable for use in humans and/or mammals without excessive adverse side effects (eg, toxicity, irritation, and allergies), ie, having a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier refers to a carrier for the administration of a therapeutic agent, including various excipients and diluents.
  • compositions of the present invention comprise a safe and effective amount of a fusion protein of the invention and a pharmaceutically acceptable carrier.
  • Such carriers include, but are not limited to, saline, buffer, dextrose, water, glycerol, ethanol, and combinations thereof.
  • the pharmaceutical preparation should be matched to the mode of administration, and the pharmaceutical composition of the present invention can be prepared into an injection form, for example, by a conventional method using physiological saline or an aqueous solution containing glucose and other adjuvants.
  • the pharmaceutical composition is preferably manufactured under sterile conditions.
  • the amount of active ingredient administered is a therapeutically effective amount.
  • the pharmaceutical preparation of the present invention can also be formulated into a sustained release preparation.
  • the effective amount of the fusion protein of the present invention may vary depending on the mode of administration and the severity of the disease to be treated and the like. The selection of a preferred effective amount can be determined by one of ordinary skill in the art based on various factors (e.g., by clinical trials). The factors include, but are not limited to, pharmacokinetic parameters of the fusion protein of the invention such as bioavailability, metabolism, half-life, etc.; severity of the disease to be treated by the patient, body weight of the patient, immune status of the patient, administration Ways, etc.
  • pharmacokinetic parameters of the fusion protein of the invention such as bioavailability, metabolism, half-life, etc.
  • severity of the disease to be treated by the patient body weight of the patient, immune status of the patient, administration Ways, etc.
  • a satisfactory effect can be obtained.
  • several separate doses may be administered per day, or the dose may be proportionally reduced, as is critical to the condition of the treatment.
  • the fusion proteins of the invention are particularly suitable for use in the treatment of diseases such as tumors.
  • Representative tumors include, but are not limited to, brain tumors, colorectal cancer tumors, lung cancer tumors, liver cancer tumors, breast cancer tumors, gastric cancer tumors, and pancreatic cancer tumors.
  • the fusion protein of the present invention has no significant toxicity to T cells. This may be due to the fact that IL-12 is prominent outside the T cell membrane and cannot be effectively contacted or acted on T cells, or it may be because the interstitial effect of CD62L reduces the toxicity of IL-12 to T cells.
  • the primers and DNA sequences used were all synthesized by Invitrogen.
  • Fig. 4 is an engineering vector pLenti-MSCV containing a gene of interest, which is a promoter which is optimized for transduction of T cells by using MSCV as a promoter.
  • the envelope protein particle pMD2.G contains VSV-G; gag/pol helper plasmid; and pRev plasmid system.
  • the plasmids pRRLSIN.cPPT.MSCV/GFP and 293FT cells used were commercially available, and the reagents used were also commercially available.
  • mutant CD62L refers to dKSCD62L.
  • hscIL-12/CD62L or IL-12/WTCD62L refers to hscIL-12/wild type CD62L.
  • Wild type CD62L is expressed as WTCD62L.
  • Tumor cells 938 and 526 are conventional melanoma cell lines (supplied by Dr. Rosenberg of the National Cancer Institute) and subcultured in vitro with 10% FCS RPMI medium and 0.25% pancreas every 2-3 days. Enzyme subculture. Both tumors express MART-1 antigen, of which 938 is MHC I A2-(negative) and 526 is MHC I A2+ (positive), and the genetically modified anti-tumor MART-1 T cells recognize only A2+ cell line, ie 526 cells.
  • PBMC peripheral blood cells were derived from healthy human peripheral blood, and T cells were stimulated for growth with CD3/CD28 magnetic beads or CD3 antibody for 1 day, and cultured in IL-2 (100 IU/ml) X-VIVO medium. After recombinant lentivirus transduction of T cells, the fluorescence intensity of MART-1 in T cells was detected by flow cytometry through the MART-1 tetramer peptide. At the same time, the expression of CD62L on the cell surface, hscIL-12/mutant CD62L and CD107a on the membrane surface were detected by flow cytometry.
  • the recombinant lentiviral vector of human TCR ⁇ and ⁇ chain genes is transfected into autologous lymphocytes of peripheral blood, so that recombinant TCR is expressed in T lymphocytes to achieve the purpose of effectively killing tumors.
  • a TCR expression vector containing a self-cleaving 2A peptide, furin (Furin) and a spacer sequence was constructed using a lentiviral vector with an optimized promoter.
  • WTCD62L wild-type CD62L
  • dKSCD62L mutant CD62L
  • WTCD62L wild-type CD62L
  • dKSCD62L mutant CD62L
  • hscIL-12 based on published literature (Zhang, Kerkar et al. 2011).
  • the fusion proteins of hscIL-12/wild type CD62L and hscIL-12/mutant CD62L were ligated with peptide G 6 S and 218 peptides, respectively.
  • 293T cells were cultured, and the cell density was adjusted in DMEM medium containing 10% fetal bovine serum one day before transfection. Then, 25 ⁇ 10 6 293T cells were inoculated per 15 cm cell culture dish, and cultured at 37 ° C, 5% CO 2 . The culture in the box can be used for transfection after the cell density is increased to 80% to 90% after 16h to 24h. On the day of transfection, the medium was changed to complete medium without antibiotics (P/S) (DMEM + 10% FBS).
  • P/S DMEM + 10% FBS
  • the lentiviral backbones of LVV-MSCV-MART-1TCR, CD62L, hscIL-12L, and hscIL-12/mutant CD62L fusion proteins were co-transfected into 293T cells together with three other packaging plasmids, using commercially available calcium phosphate. medium. After 6 hours of culture, the medium was discarded, washed 3 times with PBS and replaced with 20 ml of fresh complete medium (DMEM + 10% FBS + P / S). The culture supernatant of 30-72 h after transfection was collected, centrifuged at 6000 rpm for 10 min, and the cell debris was discarded.
  • the supernatant was filtered through a 0.45 ⁇ m PVDF filter into a 50 ml round bottom centrifuge tube, centrifuged at 50,000 g for 2 h at 4 ° C, and carefully discarded. Clear, DMEM (free of serum, double antibody) resuspended virus precipitate, according to the amount of virus used each time into a clean 15ml centrifuge tube, stored in a -80 ° C refrigerator, used to infect T cells.
  • the titer of the virus detected by the Lentivirus-Associated p24 ELISA Kit was 5 ⁇ 10 7 -1.5 ⁇ 10 8 IFU. For details, see the instructions of the Lentivirus-Associated p24 ELISA Kit. (Yang, Cohen et al. 2008).
  • T cells modified by anti-MART-1 TCR gene were co-cultured with 526, 938 cell lines, placed in a 1:1 ratio, ie 1 ⁇ 10 6 per cell, in a 14 ml round bottom polypropylene culture tube, total volume 1 ml, transfer The mixture was incubated in a CO 2 incubator at 37 ° C for 4 h. After 4 h, the cells were centrifuged at 800 x g for 10 minutes, the supernatant was collected, and the cells were lysed by RIPA lysate. The content of IFN ⁇ and IL-12 in the supernatant was detected by an ELISA reagent.
  • IL-12/wild-type CD62L or IL-12/mutant CD62L on the membrane surface was manipulated by live cell staining of a conventional flow cytometer. Wild-type or mutant CD62L in the supernatant and wild-type or mutant CD62L in the cells were detected by ELISA kit (R&D Systems, Minneapolis, MN).
  • Cell surface CD3, CD8, CD62L, CD107a, IL-12 and CD45RO are detected by fluorescently labeled corresponding antibodies, including isothiocyanate (FITC), allophycocyanin (APC), phycoerythrin (PE), PE-Cy7. , and APC-Cy7 (BD Biosciences, San Jose, CA).
  • FITC isothiocyanate
  • APC allophycocyanin
  • PE phycoerythrin
  • PE-Cy7 BD Biosciences, San Jose, CA
  • APC-Cy7 BD Biosciences, San Jose, CA.
  • the MART-1:27-35 tetramer was designed by the company (iTAg MHC Tetramer, Beckman Coulter, Fullerton, CA) to detect gene-modified TCR expression levels. The specific procedure is as follows.
  • the cells were washed twice with FACS staining solution (PBS containing 2% FBS), then 0.2 ml (10 6 /ml) was added to the flow tube, incubated at 4 ° C for 30 minutes, and then washed twice.
  • the dead cells were separated by adding 20 ⁇ l PI (l5 ⁇ g/ml propidium iodide) (Sigma-Aldrich, Saint Louis, MO) and cell subpopulation before the sample was taken to achieve the purpose of separation.
  • the streaming data is analyzed by FlowJo 8.1.1 for post-processing (FlowJo, Ashland, OR).
  • the Pmel experimental mouse model uses conventional methods, for example, see the literature (Overwijk, Tsung et al. 1998).
  • the method involved in this experiment was as follows: Female pmel mice (6-8 weeks, 7 mice per group) were selected for intracranial tumor inoculation (IC). B16F10-MART-1 tumor cells were stopped by 0.25% trypsinization with 0.02% EDTA and washed once with serum-containing medium to wash the trypsin reaction, followed by washing twice with PBS.
  • Tumor cells were finally mixed with methylcellulose in zinc option medium in a volume of 1:1, and 5000 cells were diluted in a 5 ⁇ l liquid and loaded onto a 250- ⁇ l syringe (Hamilton, Reno, NV) using a 25-gauge needle.
  • the Quinesential Stereotaxic Injector System (Stoelting Co. Wood Dale, IL) was injected into the right brain caudate nucleus of the mice. Five days after inoculation of the tumor cells, the mice received whole body 5 Gy radiation.
  • mice received a 0.5-1 ⁇ 10 6 DC vaccine subcutaneously, or a 1X 10 7 retransmission of anti-MART-1 TCR, IL-12/wild-type CD62L or IL-12/mutation by tail vein IV.
  • Type CD62L lentiviral gene-modified T cells Mouse T cells were obtained from mouse spleen cells, activated with 10 ug/ml concanavalin (Con A) in the presence of IL-2 (5 IU/ml); day 2, transduced with Lentiviral vector T The cells were then cultured for 6 days, cells were collected, and T cells were injected through the tail vein of the mice.
  • Con A concanavalin
  • IL-2 5 IU/ml
  • DC cells of DC group mice were obtained from mouse bone marrow cells, induced differentiation and maturation in vitro for 8 days, and inoculated intraperitoneally. The deaths of the mice were then recorded daily, growth curves were recorded and plotted by Prism mapping software. Asterisks indicate that the experimental group was compared with the other groups, p ⁇ 0.001.
  • the fusion gene was synthesized by Invitrogen, and the length and sequence of the fusion gene was passed through 1% agar. Glycoelectrophoresis and sequencing confirmed.
  • the structure of the obtained IL-12/mutant CD62L fusion gene was constructed as shown in SEQ ID NO.: 1, and the amino acid sequence of the encoded fusion protein is shown in SEQ ID NO.: 2.
  • the obtained IL-12/wild type CD62L fusion gene was constructed at the same time, and the IL-12/wild type CD62L fusion gene was very similar to SEQ ID NO.: 1, except that the nucleotide encoding the cleavage site was retained. The sequence, so the encoded fusion protein also retains the ADAM17 cleavage site (ie KLDKSFS).
  • the obtained lentiviral expression vectors were named LV-hscIL-12/wild type CD62L (expressing IL-12/CD62L), LV-hscIL-12/mutant CD62L (expressing IL-12/dKSCD62L), LV-hscIL-12, respectively. .
  • the method is as follows: CD3/CD28 magnetic beads or anti-CD3 antibody activates PBMC, and on day 2, the T cells are modified by lentiviral gene.
  • the brief method is as follows: Wash T cells three times in PBS buffer, according to virus titer and T cell The appropriate amount of lentivirus was added in a ratio of 3:1, centrifuged at 2000 ⁇ g for 2 h, and after 6 h, the culture was continued by adding 100 IU/ml IL-2; the second transduction was performed on the 5th day, or combined co-transduction. The flask was divided according to the growth of the cells, and two weeks later, the cells modified by the T cells were examined by flow cytometry.
  • dKSCD62L lentiviral gene-modified anti-tumor T cells achieve stable expression of dKSCD62L
  • a tumor infiltrating T cell line JKF6 isolated and cultured in melanoma tissue was selected.
  • the JKF6 cell line can be subcultured in vitro for a long time, and the membrane surface loses the CD62L membrane molecule, and JKF6 can effectively recognize melanoma cells.
  • JKF6 can detect high levels of exogenous WTCD62L and dKSCD62L expression by lentiviral gene modification. Wild-type CD62L and mutant dKSCD62L gene-modified JKF6 obtained stable membrane surface CD62L series expression.
  • wild-type CD62L After activation with tumor antigen 526, wild-type CD62L can undergo tumor antigen-specific cleavage and release; however, after expression of dKSCD62L membrane, it loses the cleavage and release of tumor antigen activation, which is expressed as stable membrane surface dKSCD62L expression.
  • Phenotype as shown in Fig. 2 and Fig. 3, the co-cultured T cells were detected by flow cytometry on the surface of the membrane CD45RO and CD62L, and the membrane stability of the CD62L mutant was fully verified by FlowJo software analysis.
  • dKSCD62L lentiviral gene modification against tumor JFK6 cells does not affect the antitumor activity of T cells
  • PMA/Ionomycin A broad-spectrum T cell activator PMA/Ionomycin (abbreviated as "PMA/Ion", a T cell activator) was used, and after activation of T cells with the activator for 4 hours, we detected CD62L on the cell membrane ( Expression of wild type or mutant type). In addition, it was also found that for the mutant dKSCD62L, activation-induced membrane cleavage disappeared, that is, stable membrane surface expression of dKSCD62L was obtained; for wild-type CD62L, T cell activation cleavage remained.
  • a lentiviral vector (structure shown in Figure 4) of IL-12/CD62L, IL-12/dKSCD62L was constructed, in which the IL-12 gene and the mutant or wild type CD62L gene pass the amino acid peptide G 6 S link, the lentivirus is a third generation lentiviral vector, and the promoter is MSCV.
  • the 5' and 3' LTR (long terminal repeat) of the vector was transformed into SIN-LTR (self-inactivating-LTR) to reduce the probability of lentiviral recombination and enhance safety performance.
  • IL-12/dKSCD62L fusion protein in anti-tumor T cells, was transduced, and the anti-tumor T cells were co-cultured with tumor cells 526 and 938 after 14 days of in vitro culture.
  • Lentiviral vector IL-12/CD62L transduced T cells co-culture with tumor can enhance the expression of IFN ⁇
  • a fusion protein vector stably expressing IL-12/dKSCD62L is constructed.
  • human T cells were sequentially transduced with anti-tumor TCR and IL-12/CD62L series fusion proteins for 14 days, and co-cultured with tumor cells 526 and 938. After 24 hours, the supernatants were IFN ⁇ and IL-12. Expression levels were detected by ELISA kit.
  • IL-12/dKSCD62L Consistent with the stable expression of the surface of dKSCD62L membrane, IL-12/dKSCD62L also lost the cleavage and release of tumor antigen reactivity, which was detectable compared with IL-12/wild type CD62L group and persistent secretion IL-12 group. The level of secreted IL-12 was significantly reduced, P ⁇ 0.001.
  • Lentiviral vector IL-12/dKSCD62L transduced T cells can effectively avoid the side effects of in vitro T cell expansion caused by persistent secretion of IL-12.
  • this example observes the effect of transduction of the IL-12/dKSCD62L fusion protein on T cell expansion in vitro.
  • the transduced hscIL-12/dKSCD62L fusion protein group was the same as the other groups, and no significant difference was observed.
  • the amplification fold of the hscIL-12/dKSCD62L fusion protein group was slightly higher than that of the hscIL-12/wild type CD62L fusion protein group. This suggests that the transduction is due to the release of free IL-12. This resulted in almost no significant toxicity of IL-12 to T cells.
  • mice Female pmel mice (7 mice per group) were selected for intracranial (IC) implantation by B16F10 cells for 5 days, and mice received 5 Gy whole body radiotherapy 1 day before cell return.
  • Mouse T cells were obtained from mouse spleen cells and activated by 10 ug/ml concanavalin (Con A) in the presence of IL-2 (5 IU/ml); on day 2, transduction of T with lentiviral vector The cells were then cultured for 6 days, cells were collected, and (X) 5 ⁇ 10 6 T cells were injected through the tail vein of the mice.
  • the DC cells of the DC group were obtained from bone marrow cells, and induced to differentiate and mature for 8 days in vitro, and 1 ⁇ 10 6 cells were inoculated intraperitoneally. The group description of DC and T cells is shown on the right. An asterisk (*) indicates that the experimental group was compared with the other groups, p ⁇ 0.001.
  • CD62L is a homing factor for T cell surface expression, which can achieve adhesion and release of T cells along the vessel wall by activation-induced cleavage of T cells. Studies have found that T cells lacking the tumor necrosis factor-converting enzyme 17 cause CD62L to be cleavable and released.
  • the present inventors knocked out the cleavage point of ADAM17 of CD62L (designated as dKSCD62L in the present invention), and the experimental results indicate that the expression of a novel IL-12/mutant CD62L molecule by genetically modifying anti-tumor T cells is not only Efficient killing of tumor cells can significantly reduce the side effects of IL-12.
  • IL-12/mutant CD62L fusion protein gene-modified anti-tumor T cells can not only prolong the survival of tumor-bearing mice, but also avoid the sustained secretion of IL-12.
  • Induced systemic cytotoxicity is a brand new immune cell therapy strategy that expresses IL-12 through cell membrane and cleaves and releases tumor antigen reactivity through CD62L. It will play a role in the treatment of tumor immune cells. Important role.

Abstract

Provided are an IL-12-mutant CD62L fusion protein, preparation method and use thereof, wherein the mutant CD62L lacks an ADAM17 cleavage site.

Description

基于IL-12稳定膜表达的肿瘤治疗剂及其制法和用途Tumor therapeutic agent based on IL-12 stable membrane expression, preparation method and use thereof 技术领域Technical field
本发明涉及医药领域,具体地涉及一种基于IL-12稳定膜表达的高效低毒的肿瘤治疗剂及其制法和用途。The invention relates to the field of medicine, in particular to a high-efficiency and low-toxic tumor therapeutic agent based on IL-12 stable membrane expression, a preparation method thereof and use thereof.
背景技术Background technique
IL-12是一种非常重要的免疫刺激因子。IL-12是由共价键链接的异质性二聚体细胞因子,由p35和p40亚基组成,在体内由激活的免疫细胞分泌。IL-12是重要的细胞免疫调控因子,在抗感染免疫及恶性肿瘤的免疫中通过NK细胞以及CTL发挥作用。IL-12 is a very important immune stimulating factor. IL-12 is a heterodimeric cytokine linked by a covalent bond, consisting of the p35 and p40 subunits, secreted by activated immune cells in vivo. IL-12 is an important cellular immune regulatory factor that acts through NK cells and CTLs in immunity against infection and malignancy.
虽然IL-12具有显著的抑瘤效果,但是由于其系统性全身给药会引起全身性毒副反应,因而受到极大的限制。在本发明之前,IL-12的临床应用由于其所诱导的全身性毒副作用,以及早期临床试验通过全身系统性给药引起的两例患者的意外死亡,使其临床应用受到大大的限制。Although IL-12 has a significant antitumor effect, it is extremely limited due to systemic systemic administration which causes systemic toxic side effects. Prior to the present invention, the clinical application of IL-12 was greatly limited in clinical application due to the systemic toxic side effects induced by it and the accidental death of two patients caused by systemic administration in early clinical trials.
为了避免全身性给药的毒副作用从而实现临床应用的目的,临床科研工作者们通过肿瘤局部注射、瞬时表达及多点注射等途径应用于肿瘤的临床试验,但是由于IL-12的治疗效果同回输的IL-12的剂量直接相关,上述局部的临床方案并未有取得显著的抑瘤效果。In order to avoid the toxic side effects of systemic administration and achieve clinical application, clinical researchers have applied to clinical trials of tumors through local injection of tumors, transient expression and multiple injections, but the therapeutic effects of IL-12 are the same. The dose of IL-12 returned was directly related, and the above-mentioned local clinical protocol did not achieve significant tumor inhibition.
为了解决这一问题,本发明人曾经试图通过基因修饰抗肿瘤T细胞持续分泌IL-12来实现抗肿瘤效果的最大化,但是由于未知的原因(一种可能的原因是IL-12的毒副作用)通过持续分泌IL-12方案基因修饰的T细胞在体外不能有效扩增,并引起大量的T细胞凋亡。In order to solve this problem, the present inventors have attempted to maximize the antitumor effect by genetically modifying anti-tumor T cells to continuously secrete IL-12, but for unknown reasons (a possible cause is the side effects of IL-12) T cells that have been genetically modified by sustained secretion of the IL-12 gene are not efficiently amplified in vitro and cause a large number of T cell apoptosis.
综上所述,本领域尚缺乏高效且低毒副作用的基于IL-12的抗肿瘤药物。因此,本领域迫切需要开发高效且低毒副作用的肿瘤治疗药物。In summary, there is a lack of IL-12-based antitumor drugs that are highly effective and have low toxic side effects. Therefore, there is an urgent need in the art to develop a tumor therapeutic drug that is highly effective and has low toxic side effects.
发明内容Summary of the invention
本发明的目的就是提供了一种高效且毒副作用低的肿瘤治疗药物及其制法和应用。The object of the present invention is to provide a tumor therapeutic drug with high efficiency and low toxic side effects, and a preparation method and application thereof.
在本发明的第一方面,提供了一种融合蛋白,所述融合蛋白包括融合在一起的以下元件:In a first aspect of the invention, there is provided a fusion protein comprising the following elements fused together:
(i)任选的位于N端的信号肽和/或前导肽; (i) an optional signal peptide and/or leader peptide at the N-terminus;
(ii)第一蛋白元件;(ii) a first protein component;
(iii)第二蛋白元件;以及(iii) a second protein component;
(iv)任选的位于第一蛋白元件和第二蛋白元件之间的连接肽元件;(iv) an optional linker element located between the first protein element and the second protein element;
其中,所述信号肽可操作地连于由(ii)、(iii)和(iv)所构成的融合元件;Wherein the signal peptide is operably linked to the fusion element consisting of (ii), (iii) and (iv);
并且第一蛋白元件为IL-12蛋白元件;第二蛋白元件为突变型CD62L的蛋白元件,所述突变型CD62L的蛋白元件缺失了ADAM17的切割位点。And the first protein element is an IL-12 protein element; the second protein element is a protein element of the mutant CD62L, and the protein element of the mutant CD62L lacks a cleavage site of ADAM17.
在另一优选例中,所述的融合蛋白具有选自下组的结构:In another preferred embodiment, the fusion protein has a structure selected from the group consisting of:
(1)式Ia所述结构:(1) Structure described by Formula Ia:
D-A-B       (Ia),或D-A-B (Ia), or
(2)式Ⅱa所述结构:(2) Structure described in Formula IIa:
D-A-C-B     (Ⅱa),D-A-C-B (IIa),
其中,among them,
A为IL-12蛋白元件;A is an IL-12 protein element;
B为突变型CD62L蛋白元件;B is a mutant CD62L protein element;
C为任选的连接肽元件;C is an optional linker element;
D为任选的信号肽信号肽和/或前导肽序列;D is an optional signal peptide signal peptide and/or leader peptide sequence;
各“-”独立地表示连接上述元件的肽键或肽接头。Each "-" independently indicates a peptide bond or a peptide linker to which the above elements are attached.
在另一优选例中,所述的突变型CD62L的蛋白元件缺失了KLDKSFS序列中的部分序列或全部序列,从而导致无法被ADAM17切割。In another preferred embodiment, the protein element of the mutant CD62L lacks a partial or total sequence in the KLDKSFS sequence, resulting in the inability to be cleaved by ADAM17.
在另一优选例中,所述的“可操作地连于”指所述信号肽可引导所述融合元件的表达或跨膜转移(定位)。In another preferred embodiment, "operably linked" means that the signal peptide can direct expression or transmembrane transfer (localization) of the fusion element.
在另一优选例中,所述的连接肽元件包括序列如SEQ ID NO.:7所示的连接肽。In another preferred embodiment, the linker peptide element comprises a linker peptide having the sequence set forth in SEQ ID NO.: 7.
在另一优选例中,所述的IL-12蛋白来源于人或非人哺乳动物。In another preferred embodiment, the IL-12 protein is derived from a human or a non-human mammal.
在另一优选例中,所述的IL-12蛋白包括野生型和突变型。In another preferred embodiment, the IL-12 protein comprises wild type and mutant form.
在另一优选例中,所述的IL-12蛋白包括全长的、成熟形式的IL-12,或其活性片段。In another preferred embodiment, the IL-12 protein comprises a full length, mature form of IL-12, or an active fragment thereof.
在另一优选例中,所述的第一蛋白元件包括IL-12蛋白的一个或两个亚基。In another preferred embodiment, the first protein element comprises one or two subunits of an IL-12 protein.
在另一优选例中,所述的IL-12蛋白的亚基选自下组:P40和P35亚基。In another preferred embodiment, the subunit of the IL-12 protein is selected from the group consisting of the P40 and P35 subunits.
在另一优选例中,所述的第一蛋白元件包括连接在一起的IL-12蛋白P40和P35亚基。In another preferred embodiment, the first protein element comprises the IL-12 protein P40 and P35 subunits joined together.
在另一优选例中,所述的P40和P35亚基为“头-头”、“头-尾”、“尾-尾”相连。In another preferred embodiment, the P40 and P35 subunits are "head-to-head", "head-to-tail", and "tail-tail".
在另一优选例中,所述的P40和P35亚基之间存在或不存在接头(linker)。较佳地,所述的接头为柔性的4-20个氨基酸的接头,更佳地,所述的接头为GGGGGGS(即G6S)(SEQ ID NO.:8)。 In another preferred embodiment, a linker is present or absent between the P40 and P35 subunits. Preferably, the linker is a flexible linker of 4-20 amino acids, more preferably, the linker is GGGGGGS (i.e., G 6 S) (SEQ ID NO.:8 ).
在另一优选例中,所述的IL12蛋白元件的序列如SEQ ID NO.:4所示。In another preferred embodiment, the sequence of the IL12 protein element is set forth in SEQ ID NO.: 4.
在另一优选例中,所述的突变型CD62L蛋白元件中缺少了K283-S284切割位点。In another preferred embodiment, the K283-S284 cleavage site is absent from the mutant CD62L protein element.
在另一优选例中,所述的突变型CD62L蛋白来源于人或非人哺乳动物。In another preferred embodiment, the mutant CD62L protein is derived from a human or non-human mammal.
在另一优选例中,所述的突变型CD62L蛋白包括全长的、成熟形式的CD62L,或其活性片段。In another preferred embodiment, the mutant CD62L protein comprises a full length, mature form of CD62L, or an active fragment thereof.
在另一优选例中,所述的突变型CD62L蛋白元件的序列如SEQ ID NO.:6所示。In another preferred embodiment, the sequence of the mutant CD62L protein element is set forth in SEQ ID NO.: 6.
在另一优选例中,所述的肽接头的长度为0-15个氨基酸,较佳地1-10个氨基酸。In another preferred embodiment, the peptide linker is 0-15 amino acids in length, preferably 1-10 amino acids.
在另一优选例中,所述融合蛋白还包括信号肽元件D。In another preferred embodiment, the fusion protein further comprises a signal peptide element D.
在另一优选例中,所述的融合蛋白中,在单链IL-12(第一蛋白元件)和突变型CD62L(第二蛋白元件)之间设有连接肽,优选218肽段(SEQ ID NO.:7)。In another preferred embodiment, the fusion protein is provided with a linker peptide, preferably a 218 peptide segment, between the single-stranded IL-12 (first protein element) and the mutant CD62L (second protein element) (SEQ ID NO.: 7).
在另一优选例中,所述的融合蛋白中,第一蛋白元件为单链IL-12,在所述单链IL-12中,在P40亚基及P35亚基设有连接肽G6S(SEQ ID NO.:8)。In another preferred embodiment, in the fusion protein, the first protein element is a single-stranded IL-12, and in the single-stranded IL-12, a linker peptide G 6 S is provided in the P40 subunit and the P35 subunit. (SEQ ID NO.: 8).
在另一优选例中,所述融合蛋白的氨基酸序列如SEQ ID NO.:2所示。In another preferred embodiment, the amino acid sequence of the fusion protein is shown in SEQ ID NO.: 2.
在另一优选例中,所述融合蛋白具有以下多种特征:In another preferred embodiment, the fusion protein has the following characteristics:
a)所述融合蛋白不能被ADAM17蛋白切割,从而不释放IL-12;a) the fusion protein is not cleaved by the ADAM17 protein, thereby not releasing IL-12;
b)所述融合蛋白包含IL-12的两条亚基,即P40和P35亚基,并由GGGGGGS(G6S)连接。b) The fusion protein comprises two subunits of IL-12, namely the P40 and P35 subunits, and is joined by GGGGGGS (G6S).
在另一优选例中,所述的融合蛋白为单体、或二聚体。In another preferred embodiment, the fusion protein is a monomer, or a dimer.
在本发明的第二方面,提供了一种分离的多核苷酸,所述的多核苷酸编码本发明第一方面所述的融合蛋白。In a second aspect of the invention, there is provided an isolated polynucleotide encoding the fusion protein of the first aspect of the invention.
在另一优选例中,所述的多核苷酸的序列如SEQ ID NO.:1所示。In another preferred embodiment, the sequence of the polynucleotide is set forth in SEQ ID NO.: 1.
在本发明的第三方面,提供了一种载体,它含有本发明第二方面所述的多核苷酸。In a third aspect of the invention, a vector comprising the polynucleotide of the second aspect of the invention is provided.
在另一优选例中,所述的载体包括质粒、病毒载体。In another preferred embodiment, the vector comprises a plasmid, a viral vector.
在另一优选例中,所述的病毒载体包括:慢病毒载体、腺病毒载体、黄热病毒载体。In another preferred embodiment, the viral vector comprises a lentiviral vector, an adenoviral vector, and a yellow fever virus vector.
在另一优选例中,所述的载体包括表达载体。In another preferred embodiment, the vector comprises an expression vector.
在本发明的第四方面,提供了一种宿主细胞,其特征在于,它含有本发明第三方面所述的载体或基因组中整合有本发明第二方面所述的多核苷酸。In a fourth aspect of the invention, a host cell comprising the polynucleotide of the second aspect of the invention, or a polynucleotide of the second aspect of the invention, is provided.
在另一优选例中,所述的宿主细胞包括原核细胞和真核细胞。In another preferred embodiment, the host cell comprises a prokaryotic cell and a eukaryotic cell.
在另一优选例中,所述的宿主细胞包括哺乳动物细胞。In another preferred embodiment, the host cell comprises a mammalian cell.
在另一优选例中,所述的宿主细胞包括免疫细胞,较佳地T细胞、In another preferred embodiment, the host cell comprises an immune cell, preferably a T cell,
在本发明的第五方面,提供了一种产生本发明第一方面所述的蛋白的方法,它 包括步骤:In a fifth aspect of the invention, there is provided a method of producing the protein of the first aspect of the invention, Including steps:
(1)在适合表达的条件下,培养本发明第四方面所述的宿主细胞,从而表达出本发明第一方面所述的融合蛋白;和(1) cultivating the host cell of the fourth aspect of the invention under conditions suitable for expression, thereby expressing the fusion protein of the first aspect of the invention;
(2)任选地分离所述融合蛋白。(2) Optionally isolating the fusion protein.
在本发明的第六方面,提供了一种免疫细胞,所述的免疫细胞在膜表面上携带本发明第一方面所述的融合蛋白。In a sixth aspect of the invention, there is provided an immune cell which carries the fusion protein of the first aspect of the invention on the surface of the membrane.
在另一优选例中,所述的免疫细胞为至少103个(较佳地103-109个,更佳地较佳地104-108个)所述免疫细胞的细胞群。In another preferred embodiment, the immune cells of at least 10 3 (preferably 103 to 109, more preferably preferably 104-108 th) the cell population of immune cells.
在另一优选例中,所述的免疫细胞或免疫细胞群中的全部或大多数(≥80%,较佳地≥90%)的细胞是活的。In another preferred embodiment, all or most (> 80%, preferably > 90%) of the cells of the immune cells or population of immune cells are viable.
在另一优选例中,至少一部分或全部所述的融合蛋白位于所述免疫细胞的细胞膜上,并且所述的第一蛋白元件即IL-12蛋白元件位于胞外。In another preferred embodiment, at least a portion or all of said fusion protein is located on the cell membrane of said immune cell, and said first protein element, i.e., the IL-12 protein element, is located extracellularly.
在另一优选例中,所述的免疫细胞包括T细胞。In another preferred embodiment, the immune cells comprise T cells.
在另一优选例中,所述的T细胞表面携带MART-1TCR。In another preferred embodiment, the T cell surface carries a MART-1 TCR.
在本发明的第七方面,提供了一种药物组合物,所述的组合物包含:In a seventh aspect of the invention, a pharmaceutical composition is provided, the composition comprising:
本发明第六方面所述的的免疫细胞,以及The immune cell of the sixth aspect of the invention, and
药学上可接受的载体。A pharmaceutically acceptable carrier.
在另一优选例中,所述的药物组合物为液态。In another preferred embodiment, the pharmaceutical composition is in a liquid state.
在另一优选例中,所述的药物组合物含有1×103-1×107个所述的免疫细胞/ml。In another preferred embodiment, the pharmaceutical composition contains 1 x 10 3 - 1 x 10 7 of said immune cells/ml.
在本发明的第八方面,提供了一种如本发明第一方面所述的融合蛋白和/或本发明第六方面所述的的免疫细胞的用途,它们被用于制备治疗肿瘤的药物。In an eighth aspect of the invention, there is provided a use of the fusion protein of the first aspect of the invention and/or the immune cell of the sixth aspect of the invention, which is used for the preparation of a medicament for treating a tumor.
在另一优选例中,所述肿瘤包括:脑肿瘤、大肠癌肿瘤、肺癌肿瘤、肝癌肿瘤、乳腺癌肿瘤、胃癌肿瘤、胰腺癌肿瘤。In another preferred embodiment, the tumor comprises: a brain tumor, a colorectal cancer tumor, a lung cancer tumor, a liver cancer tumor, a breast cancer tumor, a gastric cancer tumor, and a pancreatic cancer tumor.
在本发明第九方面,提供了一种治疗肿瘤的方法,包括步骤:给需要的对象施用本发明第一方面所述的融合蛋白和/或本发明第六方面所述的的免疫细胞。In a ninth aspect of the invention, there is provided a method of treating a tumor comprising the step of administering to a subject in need thereof the fusion protein of the first aspect of the invention and/or the immune cell of the sixth aspect of the invention.
在另一优选例中,所述的融合蛋白以单体和/或二聚体形式施用。In another preferred embodiment, the fusion protein is administered as a monomer and/or a dimer.
在另一优选例中,所述的对象是人。In another preferred embodiment, the object is a human.
在本发明的第十方面,提供了一种非治疗性或治疗性的体外杀灭肿瘤细胞的方法,包括步骤:将本发明第六方面所述的的免疫细胞与肿瘤细胞进行接触,从而杀灭所述肿瘤细胞。In a tenth aspect of the invention, a non-therapeutic or therapeutic method for killing tumor cells in vitro is provided, comprising the steps of: contacting the immune cells of the sixth aspect of the invention with tumor cells, thereby killing The tumor cells are destroyed.
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 is to be understood that within the scope of the present invention, the various technical features of the present invention and the various technical features specifically described hereinafter (as in the embodiments) may be combined with each other to constitute a new or preferred technical solution. Due to space limitations, we will not repeat them here.
附图说明DRAWINGS
图1显示了CD62L跨膜区的氨基酸组成及ADAM17的切割位点。图1A和1B表示dKSCD62L突变体(缺失了KLDKSFS)在T细胞活化后,不能发生基于切割酶(如Adam17等)的切割及释放,导致dKSCD62L还留在T细胞膜表面。图1C中,WT代表野生型的序列,dKS代表敲除了ADAM17序列的CD62L突变体。Figure 1 shows the amino acid composition of the CD62L transmembrane region and the cleavage site of ADAM17. 1A and 1B show that the dKSCD62L mutant (with KLDKSFS deleted) cannot undergo cleavage and release based on a cleavage enzyme (such as Adam17) after T cell activation, resulting in dKSCD62L remaining on the surface of the T cell membrane. In Figure 1C, WT represents the wild type sequence and dKS represents the CD62L mutant knocked out of the ADAM17 sequence.
图2显示了dKSCD62L慢病毒基因修饰的抗肿瘤T细胞导致dKSCD62L稳定表达。Figure 2 shows that dKSCD62L lentiviral gene-modified anti-tumor T cells result in stable expression of dKSCD62L.
其中,如上图所示,抗肿瘤T细胞系JKF6(由黑色素瘤组织中分离培养的抗肿瘤T细胞系,可以直接识别肿瘤)通过慢病毒基因修饰后,可以检测到WTCD62L及dKSCD62L的表达。Among them, as shown in the above figure, the anti-tumor T cell line JKF6 (an anti-tumor T cell line isolated and cultured from melanoma tissue can directly recognize the tumor) can be detected by lentiviral gene, and the expression of WTCD62L and dKSCD62L can be detected.
如下图所示,T(野生型)CD62L经过肿瘤抗原526活化后,CD62L可以发生肿瘤抗原特异性的切割及释放;但是,dKSCD62L的膜表达后,却丧失肿瘤抗原活化的切割及释放,表现为稳定的膜表面dKSCD62L的表达的表型。As shown in the figure below, CD62L can undergo tumor antigen-specific cleavage and release after T (wild-type) CD62L is activated by tumor antigen 526. However, after expression of dKSCD62L membrane, it loses the cleavage and release of tumor antigen activation. The phenotype of expression of the stable membrane surface dKSCD62L.
JKF6T细胞与表达MART-1抗原的肿瘤细胞938和526共同培养。其中,共培养的T细胞通过流式细胞仪检测膜表面的分子CD45RO和CD62L,通过FlowJo软件的分析处理,每组的T细胞被整理成CD8细胞亚群。流式细胞检测所用到的抗体为CD62L FITC,CD45RO APC,MART-1PE和CD8PerCP。JKF6T cells were co-cultured with tumor cells 938 and 526 expressing the MART-1 antigen. Among them, co-cultured T cells were detected by flow cytometry on the surface of the membrane CD45RO and CD62L, and analyzed by FlowJo software, each group of T cells were sorted into CD8 cell subsets. The antibodies used for flow cytometry were CD62L FITC, CD45RO APC, MART-1PE and CD8PerCP.
图3显示了dKSCD62L慢病毒基因修饰抗肿瘤JFK6细胞不影响T细胞的抗肿瘤活性。Figure 3 shows that dKSCD62L lentiviral gene-modified anti-tumor JFK6 cells do not affect the anti-tumor activity of T cells.
其中,图3A中,慢病毒基因修饰JKF6细胞后,通过PMA/Ionomycin(PMA/Ion,一种T细胞活化剂)处理激活T细胞4h,然后对细胞膜表面的CD62L系列及IFNγ通过常规固定及染色,并检测膜表面CD62L及胞浆内IFNγ的表达水平(上图)。图3B表示IFNγ及CD62L表达的统计学分析处理。数据为3次独立实验的分析统计结果,dKSCD62L的表达水平持续稳定,IFNγ也同其它组一样,表现为相似的高水平。结果表示为平均值±标准差,选用T-test进行统计学处理。*表示dKSCD62L与其它两组CD62L表达水平比较,P<0.01。Among them, in Figure 3A, after Lentiviral gene modification of JKF6 cells, T cells were activated by PMA/Ionomycin (PMA/Ion, a T cell activator) for 4 h, and then the CD62L series and IFNγ on the cell membrane surface were fixed and stained by routine. And the expression level of CD62L and intracytoplasmic IFNγ on the membrane surface was detected (top panel). Figure 3B shows the statistical analysis treatment of IFNy and CD62L expression. The data are analytical results of 3 independent experiments. The expression level of dKSCD62L is consistently stable, and IFNγ is similar to other groups, showing similar high levels. Results are expressed as mean ± standard deviation, and T-test was used for statistical processing. * indicates that dKSCD62L is compared with the other two groups of CD62L expression levels, P < 0.01.
图4显示了慢病毒载体表达dKSCD62L融合蛋白的构建及在人T细胞中的表达。所述慢病毒为第三代慢病毒载体,启动子为MSCV。该载体的5’及3’LTR(long terminal repeat)被改造成SIN-LTR(self-inactivating-LTR),目的是降低慢病毒重组的机率,增强安全性能。Figure 4 shows the construction of a lentiviral vector expressing dKSCD62L fusion protein and its expression in human T cells. The lentivirus is a third generation lentiviral vector and the promoter is MSCV. The 5' and 3' LTR (long terminal repeat) of this vector was engineered into SIN-LTR (self-inactivating-LTR) in order to reduce the probability of lentiviral recombination and enhance safety performance.
其中上图显示了LVV质粒的结构示意图以及人IL-12/WTCD62L(WT,野生型)、IL-12/dKSCD62L及IL-12(human single chain IL-12)的基因表达构件,其中hscIL-12融合了IL-12的分泌肽(lead seq),通过基因克隆方法连接到慢病毒表达载体中,CD62L或dKSCD62L和hscIL-12通过肽段G6S链接。The above figure shows the structural diagram of the LVV plasmid and the gene expression constructs of human IL-12/WTCD62L (WT, wild type), IL-12/dKSCD62L and IL-12 (human single chain IL-12), wherein hscIL-12 The LE-12 seq is fused to a lentiviral expression vector by gene cloning, and CD62L or dKSCD62L and hscIL-12 are linked by a peptide G 6 S.
如图5方案,T细胞通过两次转导即MART-1TCR(第一天)及图示中三个载 体的依次转导(第5天),于第14天,将其与526及938以1:1比例共培养4h后,通过流式细胞仪检测T细胞膜表面hscIL-12的表达。As shown in Figure 5, T cells pass two transductions, namely MART-1 TCR (first day) and three in the illustration. The cells were sequentially transduced (Day 5). On the 14th day, they were co-cultured with 526 and 938 in a 1:1 ratio for 4 hours, and then the expression of hscIL-12 on the surface of T cell membrane was detected by flow cytometry.
图5显示了慢病毒载体IL-12/突变型CD62L系列转导的T细胞与肿瘤细胞共培养可以增强IFNγ的表达及肿瘤抗原依赖的释放IL-12。图中,mock为空白对照组,tumor为肿瘤组。Figure 5 shows that co-culture of lentiviral vector IL-12/mutant CD62L-transduced T cells with tumor cells enhances IFNy expression and tumor antigen-dependent release of IL-12. In the figure, the mock is a blank control group and the tumor is a tumor group.
T细胞序贯转导抗肿瘤TCR及IL-12/突变型CD62L系列融合蛋白14天后,与肿瘤细胞526及938共培养,16小时后,上清中IFNγ及IL-12表达水平通过ELISA试剂盒检测。结果显示,持续性IL-12分泌组、IL-12/WTCD62L组及IL-12/dKSCD62L组较T细胞组比较,可以显著增强抗肿瘤T细胞与肿瘤的反应性,表现为显著增强IFNγ分泌的水平,P<0.001。dKSCD62L膜表面的稳定表达一致,IL-12/dKSCD62L也丧失了肿瘤抗原反应性的切割及释放,与IL-12/WTCD62L组、持续性分泌IL-12组比较,可以检测到的分泌的IL-12的水平显著降低,P<0.001。T cells were sequentially transduced with anti-tumor TCR and IL-12/mutant CD62L series fusion proteins for 14 days, and co-cultured with tumor cells 526 and 938. After 16 hours, the expression levels of IFNγ and IL-12 in the supernatant were passed through ELISA kit. Detection. The results showed that the sustained IL-12 secretion group, IL-12/WTCD62L group and IL-12/dKSCD62L group could significantly enhance the reactivity of anti-tumor T cells with tumors compared with the T cell group, which showed a significant enhancement of IFNγ secretion. Level, P < 0.001. The stable expression on the surface of dKSCD62L membrane was consistent. IL-12/dKSCD62L also lost the cleavage and release of tumor antigen reactivity. Compared with IL-12/WTCD62L group and persistent secretion IL-12 group, the secreted IL- could be detected. The level of 12 was significantly reduced, P < 0.001.
图6显示了慢病毒载体IL-12/dKSCD62L系列转导的T细胞可以有效避免持续性分泌IL-12造成的体外T细胞扩增的毒副作用。Figure 6 shows that the lentiviral vector IL-12/dKSCD62L series transduced T cells can effectively avoid the toxic side effects of in vitro T cell expansion caused by persistent secretion of IL-12.
T细胞依次转导抗肿瘤TCR及IL-12/CD62L系列融合蛋白14天后,各组细胞与对照转导组(T-cell)的扩增倍数比较,转导持续分泌IL-12组较其它各组的扩增倍数显著降低,p<0.001。其中,转导IL-12/dKSCD62L融合蛋白组同其它组比较扩增倍数相同,未见显著差异。数据分析采用设定T-cell组的扩增倍数为100%,其它各组的值系与T-cell组的比值。After T cells were transduced with anti-tumor TCR and IL-12/CD62L series fusion proteins for 14 days, the expansion ratios of the cells in each group were compared with the control transduction group (T-cell), and the transduction of IL-12 group was compared with other groups. The amplification factor of the group was significantly reduced, p < 0.001. Among them, the transduction IL-12/dKSCD62L fusion protein group was the same as the other groups, and no significant difference was observed. The data analysis was performed by setting the amplification factor of the T-cell group to 100%, and the values of the other groups were compared with the T-cell group.
图7显示了慢病毒载体IL-12/dLSCD62L系列转导鼠T细胞介导的细胞回输治疗显著延长荷瘤小鼠的生存。Figure 7 shows that lentiviral vector IL-12/dLSCD62L series transduced murine T cell-mediated cell transfusion therapy significantly prolonged survival of tumor-bearing mice.
雌性pmel小鼠(每组7只小鼠)通过B16F10细胞(5000个细胞/只)植入颅内(IC)5天,细胞回输前1天小鼠接受5Gy全身放疗。鼠的T细胞取自小鼠的脾脏细胞,通过10ug/ml的刀豆蛋白(Con A)在IL-2(5IU/ml)存在的条件下活化;第2天,用慢病毒载体转导T细胞,然后继续培养6天,收集细胞,通过小鼠尾静脉注射(IV)5X 106个T细胞。DC组小鼠的DC细胞取自骨髓细胞,经过体外诱导分化成熟8天,通过腹腔接种1X 106细胞。DC和T细胞的各组说明见右边标识。星号(*)表示该实验组与其它组比较,p<0.001。Female pmel mice (7 mice per group) were implanted intracranial (IC) for 5 days by B16F10 cells (5000 cells/only), and the mice received 5 Gy whole body radiotherapy 1 day before cell return. Mouse T cells were obtained from mouse spleen cells and activated by 10 ug/ml concanavalin (Con A) in the presence of IL-2 (5 IU/ml); on day 2, transduction of T with lentiviral vector The cells were then cultured for 6 days, cells were collected, and (X) 5 ×10 6 T cells were injected through the tail vein of the mice. The DC cells of the DC group were obtained from bone marrow cells, and induced to differentiate and mature for 8 days in vitro, and 1×10 6 cells were inoculated intraperitoneally. The group description of DC and T cells is shown on the right. An asterisk (*) indicates that the experimental group was compared with the other groups, p < 0.001.
具体实施方式detailed description
本发明人经过广泛而深入的研究,首次开发了一种结构新颖的、具有高效杀伤肿瘤细胞活性且毒副作用很小的IL-12融合蛋白。本发明的融合蛋白为IL-12/突变型CD62L的融合蛋白,其中,所述的突变型CD62L缺失了ADAM17的切割位点。实验表明,当本发明融合蛋白在T细胞中表达后,会有效展示在T 细胞的表面。出乎意料的是,该IL-12/突变型CD62L融合蛋白对T细胞的活力(viability)基本无影响,并且携带本发明融合蛋白的抗肿瘤T细胞在接近肿瘤细胞时,由于无法通过特异性切割CD62L而释放IL-12,导致位于细胞膜表面IL-12更有效而安全地作用于肿瘤细胞。在本发明中,通过T细胞攻击肿瘤的局部并通过细胞表面的IL-12改变T细胞-肿瘤组织的免疫微环境,从而协同而有效地实现抗肿瘤免疫效果的最大化,同时极其显著地降低IL-12的毒副作用。在此基础上完成了本发明。Through extensive and intensive research, the present inventors have for the first time developed a novel IL-12 fusion protein with high activity to kill tumor cells with little toxic side effects. The fusion protein of the present invention is a fusion protein of IL-12/mutant CD62L, wherein the mutant CD62L lacks the cleavage site of ADAM17. Experiments show that when the fusion protein of the present invention is expressed in T cells, it is effectively displayed in T The surface of the cell. Unexpectedly, the IL-12/mutant CD62L fusion protein has little effect on the viability of T cells, and the anti-tumor T cells carrying the fusion protein of the present invention are unable to pass specificity when approaching tumor cells. CD62L is cleaved to release IL-12, resulting in IL-12 on the cell membrane surface acting more efficiently and safely on tumor cells. In the present invention, the T cell-targeted tumor is localized and the immune micro-environment of the T cell-tumor tissue is changed by the IL-12 on the cell surface, thereby synergistically and effectively achieving the maximization of the anti-tumor immunity effect, and extremely significantly reducing The toxic side effects of IL-12. The present invention has been completed on this basis.
具体地,本发明人利用缺失了ADAM17的切割位点的突变型CD62L(dKSCD62L),开发了一种IL-12/dKSCD62L的全新的融合蛋白,并通过慢病毒基因修饰抗肿瘤T细胞。实验数据,显示IL-12/dKSCD62L慢病毒基因修饰抗肿瘤T细胞后,其膜表面的表达不受T细胞活化的调控,可以维持较高的膜表面的表达,对肿瘤抗原的反应表现出增强免疫反应的效应。临床前荷瘤鼠模型显示,该方案修饰的T细胞可以显著延长荷瘤小鼠的生存期,没有发现明显的细胞毒效应。换言之,膜表达所述融合蛋白的T细胞一方面可有效地杀灭肿瘤细胞,另一方面经证实具有体内外的安全性。此外,所述经修饰的T细胞自身似乎仅受到所表达IL-12轻微的不利影响或基本上不受其影响(与持续分泌IL-12的T细胞比较,本发明融合蛋白对T细胞的体外扩增无明显影响)。Specifically, the present inventors developed a novel fusion protein of IL-12/dKSCD62L using a mutant CD62L (dKSCD62L) lacking the cleavage site of ADAM17, and modified anti-tumor T cells by a lentiviral gene. The experimental data show that IL-12/dKSCD62L lentiviral gene-modified anti-tumor T cells, the expression of membrane surface is not regulated by T cell activation, can maintain high membrane surface expression, and enhance the response to tumor antigens. The effect of the immune response. The preclinical tumor-bearing mouse model showed that the modified T cells could significantly prolong the survival of tumor-bearing mice, and no obvious cytotoxic effects were found. In other words, the T cells expressing the fusion protein by the membrane are effective in killing tumor cells on the one hand, and have been confirmed to have safety in vitro and in vivo on the other hand. Furthermore, the modified T cells themselves appear to be only slightly adversely affected or substantially unaffected by the expressed IL-12 (in vitro, the fusion protein of the invention is directed against T cells compared to T cells that continue to secrete IL-12) Amplification has no significant effect).
术语the term
如本文所用,术语“头部”指多肽或其片段的N端,尤其是野生型多肽或其片段的N端。As used herein, the term "head" refers to the N-terminus of a polypeptide or fragment thereof, particularly the N-terminus of a wild-type polypeptide or fragment thereof.
如本文所用,术语“尾部”指多肽或其片段的C端,尤其是野生型多肽或其片段的C端。As used herein, the term "tail" refers to the C-terminus of a polypeptide or fragment thereof, particularly the C-terminus of a wild-type polypeptide or fragment thereof.
如本文所用,所述的“含有”,“具有”或“包括”包括了“包含”、“主要由……构成”、“基本上由……构成”、和“由……构成”;“主要由……构成”、“基本上由……构成”和“由……构成”属于“含有”、“具有”或“包括”的下位概念。As used herein, "containing", "having" or "including" includes "including", "consisting essentially of", "consisting essentially of", and "consisting of"; The subordinate concepts of "consisting of", "consisting essentially of" and "consisting of" are "contained," "having," or "including."
CD62LCD62L
CD62L广泛地表达于T细胞表面,是重要的免疫调节因子,可以调控T细胞迁移到全身的淋巴结,是重要的T细胞归巢因子。当CD62L+T cell迁移到淋巴结并接触到肿瘤抗原的情形下,CD62L可以在跨膜区的位点K283-S284的肽段上通过ADAM17实验肿瘤抗原反应性的切割。(Yang,Liu et al.2011)本发明中,本发明人证实了CD62L切割具有肿瘤抗原反应的特异性,同时伴随着CD107a(该分子是溶酶体上的分子蛋白,正常情况下存在于细胞浆内,在T细 胞激活状态下,该分子伴随着T细胞的脱颗粒迁移到细胞表面;因此,该分子的膜表面检测是T细胞杀伤的一个重要指标)的膜迁移。CD62L is widely expressed on the surface of T cells and is an important immunoregulatory factor. It can regulate the migration of T cells to the lymph nodes of the whole body and is an important T cell homing factor. When CD62L+T cell migrates to the lymph node and is exposed to the tumor antigen, CD62L can be tested for tumor antigen reactivity by ADAM17 on the peptide K283-S284 at the transmembrane region. (Yang, Liu et al. 2011) In the present invention, the inventors confirmed that CD62L cleavage has the specificity of a tumor antigen reaction accompanied by CD107a (the molecule is a molecular protein on the lysosome, which is normally present in the cell) In the pulp, in the T In the cell-activated state, the molecule migrates to the cell surface accompanied by degranulation of T cells; therefore, membrane surface detection of this molecule is an important indicator of membrane migration of T cell killing.
突变型CD62LMutant CD62L
如本文所用,术语“突变型CD62L”、“CD62L突变体”、“缺失ADAM17切割位点的CD62L”、“缺失ADAM17切割位点的CD62L突变体”或“缺失切割位点的CD62L突变体”等可互换使用,指这样的CD62L多肽或其活性片段或其衍生多肽,它缺失了切割酶ADAM17的切割位点,即缺失了KLDKSFS序列中的部分序列或全部序列,从而使得所述CD62L多肽或其活性片段或其衍生多肽不能被切割酶ADAM17所切割。As used herein, the terms "mutant CD62L", "CD62L mutant", "CD62L lacking the ADAM17 cleavage site", "CD62L mutant lacking the ADAM17 cleavage site" or "CD62L mutant lacking the cleavage site", etc., etc. &quot;Interchangeable use&quot; refers to a CD62L polypeptide or active fragment thereof or a polypeptide derived therefrom which lacks a cleavage site for the cleavage enzyme ADAM17, i.e., a partial or total sequence of the KLDKSFS sequence is deleted, such that the CD62L polypeptide or The active fragment or derivative thereof cannot be cleaved by the cleavage enzyme ADAM17.
一种优选的突变型CD62L是缺失了整个KLDKSFS序列的CD62L多肽。A preferred mutant CD62L is a CD62L polypeptide lacking the entire KLDKSFS sequence.
应理解,在本发明中,所述的CD62L可以是来自哺乳动物(人或非人哺乳动物),也可以来自其他真核物种。优选地,CD62L是来自人的野生型CD62L。It should be understood that in the present invention, the CD62L may be from a mammal (human or non-human mammal) or may be derived from other eukaryotic species. Preferably, CD62L is wild type CD62L from human.
在本发明中,一种特别优选的突变型CD62L是dKSCD62L,即缺失了整个KLDKSFS序列的人CD62L多肽。In the present invention, a particularly preferred mutant CD62L is dKSCD62L, a human CD62L polypeptide lacking the entire KLDKSFS sequence.
IL-12IL-12
IL-12是一种非常重要的免疫刺激因子。IL-12是由共价键链接的异质性二聚体细胞因子,由p35和p40亚基组成,在体内由激活的免疫细胞分泌。IL-12 is a very important immune stimulating factor. IL-12 is a heterodimeric cytokine linked by a covalent bond, consisting of the p35 and p40 subunits, secreted by activated immune cells in vivo.
应理解,在本发明中,所述的IL-12可以来自人或非人哺乳动物,可以是全长的、成熟形式的IL-12,或其活性片段。此外,所述的IL-12(或IL-12蛋白元件)可以是单个亚基或多个亚基。例如,在本发明中,所述的第一蛋白元件可包括IL-12蛋白的一个或多个(如两个)亚基。It will be understood that in the present invention, the IL-12 may be derived from a human or non-human mammal, and may be a full-length, mature form of IL-12, or an active fragment thereof. Furthermore, the IL-12 (or IL-12 protein element) can be a single subunit or multiple subunits. For example, in the present invention, the first protein element may comprise one or more (e.g., two) subunits of an IL-12 protein.
在另一优选例中,所述的第一蛋白元件包括连接在一起的IL-12蛋白P40和P35亚基。In another preferred embodiment, the first protein element comprises the IL-12 protein P40 and P35 subunits joined together.
在另一优选例中,所述的P40和P35亚基的连接方式没有特别限制,包括“头-头”、“头-尾”、“尾-头”、“尾-尾”相连,其中“头”指多肽的N端,“尾”指多肽的C端。In another preferred embodiment, the manner of connecting the P40 and P35 subunits is not particularly limited, and includes "head-to-head", "head-to-tail", "tail-head", and "tail-tail", wherein " "Head" refers to the N-terminus of a polypeptide, and "tail" refers to the C-terminus of a polypeptide.
此外,在P40和P35亚基之间可以存在或不存在接头(linker)。较佳地,所述的接头为柔性的4-20个氨基酸的接头,更佳地,所述的接头为GGGGGGS(G6S)(SEQ ID NO.:8)In addition, a linker may or may not be present between the P40 and P35 subunits. Preferably, the linker is a flexible linker of 4-20 amino acids, more preferably, the linker is GGGGGGS (G 6 S) (SEQ ID NO.:8)
肽接头Peptide linker
本发明的双功能融合蛋白可任选地含有肽接头。肽接头大小和复杂性可能会影响蛋白的活性。通常,肽接头应当具有足够的长度和柔韧性,以保证连接 的两个蛋白在空间上有足够的自由度以发挥其功能。同时避免肽接头中形成α螺旋或β折叠等对融合蛋白的稳定性的影响。The bifunctional fusion proteins of the invention may optionally contain a peptide linker. The size and complexity of the peptide linker may affect the activity of the protein. In general, peptide linkers should be of sufficient length and flexibility to ensure connectivity. The two proteins have enough freedom in space to perform their function. At the same time, the effect of the formation of an alpha helix or a beta sheet in the peptide linker on the stability of the fusion protein is avoided.
连接肽的长度一般为0-15个氨基酸,较佳地1-15个氨基酸。The length of the linker peptide is generally from 0 to 15 amino acids, preferably from 1 to 15 amino acids.
优选的连接肽例子包括(但并不限于):SEQ ID NO.:7或8所示的连接肽。Examples of preferred linker peptides include, but are not limited to, the linker peptide set forth in SEQ ID NO.: 7 or 8.
信号肽和前导肽Signal peptide and leader peptide
本发明中融合蛋白还可含有其他元件,代表性的元件包括(但并不限于):信号肽、前导肽等。The fusion protein of the present invention may also contain other elements including, but not limited to, signal peptides, leader peptides and the like.
在本发明的一个实例中,融合蛋白含有信号肽。代表性的例子包括(但并不限于):人源的IL-12P40亚基的信号肽。In one embodiment of the invention, the fusion protein contains a signal peptide. Representative examples include, but are not limited to, a signal peptide of the human IL-12P40 subunit.
双功能融合蛋白及其制备Bifunctional fusion protein and preparation thereof
如本文所用,除非另外说明,所述的融合蛋白是一种分离的蛋白,与其它蛋白、多肽或分子无联系,是重组宿主细胞所表达的,或经分离或纯化的产物。As used herein, unless otherwise stated, the fusion protein is an isolated protein that is not associated with other proteins, polypeptides or molecules and is a product expressed by recombinant host cells, or isolated or purified.
在本发明中,“重组双功能融合蛋白”、“本发明蛋白”、“本发明融合蛋白”、“双功能融合蛋白”、“IL-12-突变型CD62L融合蛋白”、“IL-12/突变型CD62L融合蛋白”可互换使用,指具有式Ia所述结构,或者IIa所述结构,即含有包括IL-12蛋白元件,突变型CD62L的蛋白元件和连接肽元件的融合蛋白。一个代表性的例子是IL12-dKSCD62L。本发明蛋白可以是单体或由单体形成的多聚体(如二聚体)。此外,应理解,所述术语还包括融合蛋白的活性片段和衍生物。In the present invention, "recombinant bifunctional fusion protein", "protein of the present invention", "fusion protein of the present invention", "bifunctional fusion protein", "IL-12-mutant CD62L fusion protein", "IL-12/" "Mutant CD62L fusion protein" is used interchangeably and refers to a structure having the structure of Formula Ia, or the structure described in IIa, ie, a fusion protein comprising a protein element comprising a IL-12 protein element, a mutant CD62L, and a linker element. A representative example is IL12-dKSCD62L. The protein of the invention may be a monomer or a multimer (e.g., a dimer) formed from a monomer. Furthermore, it is to be understood that the term also encompasses active fragments and derivatives of fusion proteins.
一种优选的融合蛋白的序列如SEQ ID NO.:2所示,其中第1-328位为IL-12的P40亚基;第329-335位为G6S连接肽;第336-532位为IL-12的P35亚基;第533-547位为218连接肽(SEQ ID NO.:7);第548-913位为dKSCD62L氨基酸序列。A preferred sequence of the fusion protein as shown in SEQ ID NO.:2, wherein the first bit is 1-328 of IL-12 P40 subunit; 329-335 G 6 S bit linker peptide; position 336-532 of It is the P35 subunit of IL-12; the 218-linker peptide (SEQ ID NO.: 7) at positions 533-547; and the dKSCD62L amino acid sequence at positions 548-913.
如本文所用,“分离的”是指物质从其原始环境中分离出来(如果是天然的物质,原始环境即是天然环境)。如活体细胞内的天然状态下的多核苷酸和多肽是没有分离纯化的,但同样的多核苷酸或多肽如从天然状态中同存在的其他物质中分开,则为分离纯化的。As used herein, "isolated" means that the substance is separated from its original environment (if it is a natural substance, the original environment is the natural environment). For example, the polynucleotides and polypeptides in the natural state in living cells are not isolated and purified, but the same polynucleotide or polypeptide is isolated and purified, as separated from other substances present in the natural state.
如本文所用,“分离的重组融合蛋白”是指重组融合蛋白基本上不含天然与其相关的其它蛋白、脂类、糖类或其它物质。本领域的技术人员能用标准的蛋白质纯化技术纯化重组融合蛋白。基本上纯的蛋白在非还原聚丙烯酰胺凝胶上能产生单一的主带。As used herein, "isolated recombinant fusion protein" means that the recombinant fusion protein is substantially free of other proteins, lipids, carbohydrates or other materials with which it is naturally associated. One skilled in the art can purify recombinant fusion proteins using standard protein purification techniques. Substantially pure proteins produce a single major band on a non-reducing polyacrylamide gel.
本发明的多核苷酸可以是DNA形式或RNA形式。DNA形式包括cDNA、基因组DNA或人工合成的DNA。DNA可以是单链的或是双链的。DNA可以是编码链或 非编码链。The polynucleotide of the present invention may be in the form of DNA or RNA. DNA forms include cDNA, genomic DNA or synthetic DNA. DNA can be single-stranded or double-stranded. DNA can be a coding strand or Non-coding chain.
本发明还涉及上述多核苷酸的变异体,其编码与本发明有相同的氨基酸序列的蛋白质片段、类似物和衍生物。此多核苷酸的变异体可以是天然发生的等位变异体或非天然发生的变异体。这些核苷酸变异体包括取代变异体、缺失变异体和插入变异体。如本领域所知的,等位变异体是一个多核苷酸的替换形式,它可能是一个或多个核苷酸的取代、缺失或插入,但不会从实质上改变其编码多肽的功能。The present invention also relates to variants of the above polynucleotides which encode protein fragments, analogs and derivatives having the same amino acid sequence as the present invention. Variants of this polynucleotide may be naturally occurring allelic variants or non-naturally occurring variants. These nucleotide variants include substitution variants, deletion variants, and insertion variants. As is known in the art, an allelic variant is an alternative form of a polynucleotide that may be a substitution, deletion or insertion of one or more nucleotides, but does not substantially alter the function of the polypeptide encoded thereby.
如本文所用,术语“引物”指的是在与模板配对,在DNA聚合酶的作用下能以其为起点进行合成与模板互补的DNA链的寡居核苷酸的总称。引物可以是天然的RNA、DNA,也可以是任何形式的天然核苷酸。引物甚至可以是非天然的核苷酸如LNA或ZNA等。引物“大致上”(或“基本上”)与模板上一条链上的一个特殊的序列互补。引物必须与模板上的一条链充分互补才能开始延伸,但引物的序列不必与模板的序列完全互补。比如,在一个3’端与模板互补的引物的5’端加上一段与模板不互补的序列,这样的引物仍大致上与模板互补。只要有足够长的引物能与模板充分的结合,非完全互补的引物也可以与模板形成引物-模板复合物,从而进行扩增。As used herein, the term "primer" refers to a generic term for a oligodeoxynucleotide that, in pairing with a template, can be used to synthesize a DNA strand complementary to a template under the action of a DNA polymerase. The primer may be native RNA, DNA, or any form of natural nucleotide. The primer may even be a non-natural nucleotide such as LNA or ZNA. The primer is "substantially" (or "substantially") complementary to a particular sequence on a strand on the template. The primer must be sufficiently complementary to a strand on the template to initiate extension, but the sequence of the primer need not be fully complementary to the sequence of the template. For example, a sequence that is not complementary to the template is added to the 5&apos; end of a primer complementary to the template at the 3&apos; end, such primers are still substantially complementary to the template. As long as there are sufficiently long primers to bind well to the template, the non-fully complementary primers can also form a primer-template complex with the template for amplification.
根据本发明提供的氨基酸序列,本技术领域人员可方便地用各种已知方法制得本发明的融合蛋白。这些方法例如但不限于:重组DNA法,人工合成等。According to the amino acid sequence provided by the present invention, one skilled in the art can conveniently prepare the fusion protein of the present invention by various known methods. These methods are, for example but not limited to, recombinant DNA methods, artificial synthesis, and the like.
本发明融合蛋白的元件(如IL12或突变型CD62L)的核苷酸全长序列或其片段通常可以用PCR扩增法、重组法或人工合成的方法获得。对于PCR扩增法,可根据已公开的有关核苷酸序列,尤其是开放阅读框序列来设计引物,并用市售的cDNA库或按本领域技术人员已知的常规方法所制备的cDNA库作为模板,扩增而得有关序列。当序列较长时,常常需要进行两次或多次PCR扩增,然后再将各次扩增出的片段按正确次序拼接在一起。The full length nucleotide sequence of the element of the fusion protein of the present invention (e.g., IL12 or mutant CD62L) or a fragment thereof can be generally obtained by a PCR amplification method, a recombinant method or a synthetic method. For PCR amplification, primers can be designed according to published nucleotide sequences, particularly open reading frame sequences, and used as commercially available cDNA libraries or cDNA libraries prepared by conventional methods known to those skilled in the art. The template is amplified to obtain the relevant sequence. When the sequence is long, it is often necessary to perform two or more PCR amplifications, and then the amplified fragments are spliced together in the correct order.
一旦获得了有关的序列,就可以用重组法来大批量地获得有关序列。这通常是将其克隆入载体,再转入细胞,然后通过常规方法从增殖后的宿主细胞中分离得到有关序列。Once the relevant sequences are obtained, the recombinant sequence can be used to obtain the relevant sequences in large quantities. This is usually done by cloning it into a vector, transferring it to a cell, and then isolating the relevant sequence from the proliferated host cell by conventional methods.
此外,还可用人工合成的方法来合成有关序列,尤其是片段长度较短时。通常,通过先合成多个小片段,然后再进行连接可获得序列很长的片段。In addition, synthetic sequences can be used to synthesize related sequences, especially when the fragment length is short. Usually, a long sequence of fragments can be obtained by first synthesizing a plurality of small fragments and then performing the ligation.
应用PCR技术扩增DNA/RNA的方法被优选用于获得本发明的基因。用于PCR的引物可根据本文所公开的本发明的序列信息适当地选择,并可用常规方法合成。可用常规方法如通过凝胶电泳分离和纯化扩增的DNA/RNA片段。A method of amplifying DNA/RNA using PCR technology is preferably used to obtain the gene of the present invention. The primers for PCR can be appropriately selected according to the sequence information of the present invention disclosed herein, and can be synthesized by a conventional method. The amplified DNA/RNA fragment can be isolated and purified by conventional methods such as by gel electrophoresis.
本发明也涉及包含本发明的多核苷酸的载体,以及用本发明的载体或融合蛋白编码序列经基因工程产生的宿主细胞,以及经重组技术产生本发明所述蛋白质的方法。 The invention also relates to vectors comprising the polynucleotides of the invention, as well as host cells genetically engineered using the vector or fusion protein coding sequences of the invention, and methods of producing the proteins of the invention by recombinant techniques.
通过常规的重组DNA技术,可利用本发明的多核苷酸序列可用来表达或生产重组蛋白。一般来说有以下步骤:The polynucleotide sequences of the present invention can be utilized to express or produce recombinant proteins by conventional recombinant DNA techniques. Generally there are the following steps:
(1).用本发明的编码本发明蛋白的多核苷酸(或变异体),或用含有该多核苷酸的重组表达载体转化或转导合适的宿主细胞;(1) using a polynucleotide (or variant) of the present invention encoding a protein of the present invention, or transforming or transducing a suitable host cell with a recombinant expression vector containing the polynucleotide;
(2).在合适的培养基中培养的宿主细胞;(2) a host cell cultured in a suitable medium;
(3).从培养基或细胞中分离、纯化蛋白质。(3). Separating and purifying the protein from the culture medium or the cells.
本领域的技术人员熟知的方法能用于构建含本发明蛋白的编码DNA序列和合适的转录/翻译控制信号的表达载体。这些方法包括体外重组DNA技术、DNA合成技术、体内重组技术等。所述的DNA序列可有效连接到表达载体中的适当启动子上,以指导mRNA合成。表达载体还包括翻译起始用的核糖体结合位点和转录终止子。Methods well known to those skilled in the art can be used to construct expression vectors containing the DNA sequences of the proteins of the invention and suitable transcription/translation control signals. These methods include in vitro recombinant DNA techniques, DNA synthesis techniques, in vivo recombinant techniques, and the like. The DNA sequence can be operably linked to an appropriate promoter in an expression vector to direct mRNA synthesis. The expression vector also includes a ribosome binding site for translation initiation and a transcription terminator.
此外,表达载体优选地包含一个或多个选择性标记基因,以提供用于选择转化的宿主细胞的表型性状,如真核细胞培养用的二氢叶酸还原酶、新霉素抗性以及绿色荧光蛋白(GFP),或用于大肠杆菌的四环素或氨苄青霉素抗性。Furthermore, the expression vector preferably comprises one or more selectable marker genes to provide phenotypic traits for selection of transformed host cells, such as dihydrofolate reductase for eukaryotic cell culture, neomycin resistance, and green Fluorescent protein (GFP), or tetracycline or ampicillin resistance for E. coli.
包含上述的适当DNA序列以及适当启动子或者控制序列的载体,可以用于转化适当的宿主细胞,以使其能够表达蛋白质。Vectors comprising the appropriate DNA sequences described above, as well as appropriate promoters or control sequences, can be used to transform appropriate host cells to enable expression of the protein.
宿主细胞可以是原核细胞,如细菌细胞;或是低等真核细胞,如酵母细胞;或是高等真核细胞,如哺乳动物细胞。代表性例子有:大肠杆菌,链霉菌属的细菌细胞;真菌细胞如酵母;植物细胞;果蝇S2或Sf9的昆虫细胞;CHO、COS、或293细胞的动物细胞等。The host cell can be a prokaryotic cell, such as a bacterial cell; or a lower eukaryotic cell, such as a yeast cell; or a higher eukaryotic cell, such as a mammalian cell. Representative examples are: Escherichia coli, bacterial cells of the genus Streptomyces; fungal cells such as yeast; plant cells; insect cells of Drosophila S2 or Sf9; animal cells of CHO, COS, or 293 cells, and the like.
一种特别优选的细胞为人和非人哺乳动物的细胞,尤其是免疫细胞,包括T细胞、NK细胞。A particularly preferred cell is a cell of a human and a non-human mammal, especially an immune cell, including T cells, NK cells.
用重组DNA转化宿主细胞可用本领域技术人员熟知的常规技术进行。当宿主为原核生物如大肠杆菌时,能吸收DNA的感受态细胞可在指数生长期后收获,用CaCl2法处理,所用的步骤在本领域众所周知。另一种方法是使用MgCl2。如果需要,转化也可用电穿孔的方法进行。当宿主是真核生物,可选用如下的DNA转染方法:磷酸钙共沉淀法,常规机械方法如显微注射、电穿孔、脂质体包装等。Transformation of host cells with recombinant DNA can be carried out using conventional techniques well known to those skilled in the art. When the host is a prokaryote such as E. coli, competent cells capable of absorbing DNA can be harvested after the exponential growth phase and treated by the CaCl 2 method, and the procedures used are well known in the art. Another method is to use MgCl 2 . Conversion can also be carried out by electroporation if desired. When the host is a eukaryote, the following DNA transfection methods can be used: calcium phosphate coprecipitation, conventional mechanical methods such as microinjection, electroporation, liposome packaging, and the like.
获得的转化子可以用常规方法培养,表达本发明的基因所编码的多肽。根据所用的宿主细胞,培养中所用的培养基可选自各种常规培养基。在适于宿主细胞生长的条件下进行培养。当宿主细胞生长到适当的细胞密度后,用合适的方法(如温度转换或化学诱导)诱导选择的启动子,将细胞再培养一段时间。The obtained transformant can be cultured by a conventional method to express the polypeptide encoded by the gene of the present invention. The medium used in the culture may be selected from various conventional media depending on the host cell used. The cultivation is carried out under conditions suitable for the growth of the host cell. After the host cell has grown to the appropriate cell density, the selected promoter is induced by a suitable method (such as temperature conversion or chemical induction) and the cells are cultured for a further period of time.
在上面的方法中的蛋白质可在细胞内、或在细胞膜上表达、或分泌到细胞外。如果需要,可利用其物理的、化学的和其它特性通过各种分离方法分离和纯化蛋白。这些方法是本领域技术人员所熟知的。这些方法的例子包括但并不 限于:常规的复性处理、用蛋白沉淀剂处理(盐析方法)、离心、渗透破菌、超处理、超离心、分子筛层析(凝胶过滤)、吸附层析、离子交换层析、高效液相层析(HPLC)和其它各种液相层析技术及这些方法的结合。The protein in the above method can be expressed intracellularly, or on the cell membrane, or secreted outside the cell. If desired, the protein can be isolated and purified by various separation methods using its physical, chemical, and other properties. These methods are well known to those skilled in the art. Examples of these methods include but not Limited to: conventional renaturation treatment, treatment with protein precipitant (salting method), centrifugation, osmotic bacteria, super treatment, ultracentrifugation, molecular sieve chromatography (gel filtration), adsorption chromatography, ion exchange chromatography, high efficiency Liquid chromatography (HPLC) and various other liquid chromatography techniques and combinations of these methods.
经修饰的免疫细胞Modified immune cell
本发明还提供一种表达本发明所述融合蛋白的免疫细胞(简称为“本发明免疫细胞”),所述免疫细胞在细胞表面携带所述的融合蛋白。The present invention also provides an immune cell (abbreviated as "the immune cell of the present invention") expressing the fusion protein of the present invention, which carries the fusion protein on the cell surface.
在本发明中,至少一部分或全部所述的融合蛋白位于所述免疫细胞的细胞膜上,并且所述的第一蛋白元件即IL-12蛋白元件位于胞外。In the present invention, at least a part or all of the fusion protein is located on the cell membrane of the immune cell, and the first protein element, i.e., the IL-12 protein element, is located extracellularly.
一类优选的免疫细胞包括T细胞,尤其是人的T细胞。优选地,所述的T细胞表面携带MART-1TCR。A preferred class of immune cells includes T cells, particularly human T cells. Preferably, said T cell surface carries a MART-1 TCR.
例如,在一个优选例中,提供了一种慢病毒表达体系基因修饰的T细胞,该T细胞表达抗肿瘤TCR(T-cell receptor)同时表达hscIL-12/突变型CD62L融合蛋白。For example, in a preferred embodiment, a lentiviral expression system genetically modified T cell is provided which expresses an anti-tumor TCR (T-cell receptor) and simultaneously expresses an hscIL-12/mutant CD62L fusion protein.
此外,所述的T细胞还可任选地同时表达野生型CD62L、hscIL-12(人源单链IL-12)或其组合。Furthermore, the T cells may optionally also express wild type CD62L, hscIL-12 (human single chain IL-12) or a combination thereof.
膜表达dKSCD62L突变体的机理Mechanism of membrane expression of dKSCD62L mutant
为了便于理解,本发明人提供以下机理供参考。应理解,本发明的保护范围并不受所述机理的限制。For ease of understanding, the inventors provide the following mechanism for reference. It should be understood that the scope of protection of the present invention is not limited by the mechanism.
膜表达dKSCD62L突变体的作用机制如图1所示。CD62L跨膜区中含有ADAM17的切割位点,在T细胞活化的情况下,ADAM7可以切割及释放CD62L。在本发明中,通过基因工程改造,敲除了KLDKSFS序列(即ADAM17的切割位点),从而从分子机制上实现了外源性CD62L的稳定膜表达。The mechanism of action of the membrane expressing dKSCD62L mutant is shown in Figure 1. The CD62L transmembrane region contains the cleavage site of ADAM17, and in the case of T cell activation, ADAM7 can cleave and release CD62L. In the present invention, the KLDKSFS sequence (i.e., the cleavage site of ADAM17) is knocked out by genetic engineering, thereby realizing the stable membrane expression of exogenous CD62L from a molecular mechanism.
一种典型的突变型CD62L是dKSCD62L。当T细胞被活化(包括肿瘤抗原诱导的T细胞活化)后,ADAM17不能够切割膜表面的dKSCD62L。换言之,dKSCD62L突变体在T细胞活化后,不会被ADAM17酶切割,不能释放,从而导致dK-SCD62L保留在T细胞膜表面,进而导致融合蛋白中的IL-12元件也同时保留在T细胞膜表面,最为关键的是该融合蛋白既保留了Il-12的生物学活性又不影响T细胞的功能。A typical mutant CD62L is dKSCD62L. After T cells were activated (including tumor antigen-induced T cell activation), ADAM17 was unable to cleave dKSCD62L on the membrane surface. In other words, after activation of T cells, the dKSCD62L mutant is not cleaved by the ADAM17 enzyme and cannot be released, resulting in the retention of dK-SCD62L on the surface of the T cell membrane, which in turn causes the IL-12 element in the fusion protein to remain on the surface of the T cell membrane. Most importantly, the fusion protein retains both the biological activity of Il-12 and the function of T cells.
药物组合物及施用方法Pharmaceutical composition and method of administration
本发明还提供了一种组合物,它含有(a)有效量的本发明融合蛋白和/或有效量的本发明的免疫细胞,以及药学上可接受的载体。The invention also provides a composition comprising (a) an effective amount of a fusion protein of the invention and/or an effective amount of an immune cell of the invention, and a pharmaceutically acceptable carrier.
通常,可将本发明的融合蛋白配制于无毒的、惰性的和药学上可接受的水 性载体介质中,其中pH通常约为5-8,较佳地,pH约为6-8。In general, the fusion proteins of the invention can be formulated in non-toxic, inert and pharmaceutically acceptable water In a carrier medium, wherein the pH is usually from about 5 to about 8, preferably, the pH is from about 6 to about 8.
如本文所用,术语“有效量”或“有效剂量”是指可对人和/或动物产生功能或活性的且可被人和/或动物所接受的量,如0.001-99wt%;较佳的0.01-95wt%;更佳的,0.1-90wt%。As used herein, the term "effective amount" or "effective amount" refers to an amount that is functional or active to a human and/or animal and that is acceptable to humans and/or animals, such as from 0.001 to 99% by weight; preferably 0.01-95 wt%; more preferably, 0.1-90 wt%.
当本发明的药物组合物含有免疫细胞时,“有效量”或“有效剂量”是指1×103-1×107个所述的免疫细胞/ml。When the pharmaceutical composition of the present invention contains immune cells, "effective amount" or "effective amount" means 1 x 10 3 - 1 x 10 7 of said immune cells/ml.
如本文所用,“药学上可接受的”的成分是适用于人和/或哺乳动物而无过度不良副反应(如毒性、刺激和变态反应)的,即具有合理的效益/风险比的物质。术语“药学上可接受的载体”指用于治疗剂给药的载体,包括各种赋形剂和稀释剂。As used herein, a "pharmaceutically acceptable" ingredient is one that is suitable for use in humans and/or mammals without excessive adverse side effects (eg, toxicity, irritation, and allergies), ie, having a reasonable benefit/risk ratio. The term "pharmaceutically acceptable carrier" refers to a carrier for the administration of a therapeutic agent, including various excipients and diluents.
本发明的药物组合物含有安全有效量的本发明的融合蛋白以及药学上可接受的载体。这类载体包括(但并不限于):盐水、缓冲液、葡萄糖、水、甘油、乙醇、及其组合。通常药物制剂应与给药方式相匹配,本发明的药物组合物可以被制成针剂形式,例如用生理盐水或含有葡萄糖和其他辅剂的水溶液通过常规方法进行制备。所述的药物组合物宜在无菌条件下制造。活性成分的给药量是治疗有效量。本发明的药物制剂还可制成缓释制剂。The pharmaceutical compositions of the present invention comprise a safe and effective amount of a fusion protein of the invention and a pharmaceutically acceptable carrier. Such carriers include, but are not limited to, saline, buffer, dextrose, water, glycerol, ethanol, and combinations thereof. Usually, the pharmaceutical preparation should be matched to the mode of administration, and the pharmaceutical composition of the present invention can be prepared into an injection form, for example, by a conventional method using physiological saline or an aqueous solution containing glucose and other adjuvants. The pharmaceutical composition is preferably manufactured under sterile conditions. The amount of active ingredient administered is a therapeutically effective amount. The pharmaceutical preparation of the present invention can also be formulated into a sustained release preparation.
本发明融合蛋白的有效量可随给药的模式和待治疗的疾病的严重程度等而变化。优选的有效量的选择可以由本领域普通技术人员根据各种因素来确定(例如通过临床试验)。所述的因素包括但不限于:本发明融合蛋白的药代动力学参数例如生物利用率、代谢、半衰期等;患者所要治疗的疾病的严重程度、患者的体重、患者的免疫状况、给药的途径等。通常,当本发明的融合蛋白每天以约5mg-20mg/kg动物体重(较佳的5mg-10mg/kg动物体重)的剂量给予,能得到令人满意的效果。例如,由治疗状况的迫切要求,可每天给予若干次分开的剂量,或将剂量按比例地减少。The effective amount of the fusion protein of the present invention may vary depending on the mode of administration and the severity of the disease to be treated and the like. The selection of a preferred effective amount can be determined by one of ordinary skill in the art based on various factors (e.g., by clinical trials). The factors include, but are not limited to, pharmacokinetic parameters of the fusion protein of the invention such as bioavailability, metabolism, half-life, etc.; severity of the disease to be treated by the patient, body weight of the patient, immune status of the patient, administration Ways, etc. In general, when the fusion protein of the present invention is administered at a dose of about 5 mg to 20 mg/kg of animal body weight per day (preferably 5 mg to 10 mg/kg of animal body weight), a satisfactory effect can be obtained. For example, several separate doses may be administered per day, or the dose may be proportionally reduced, as is critical to the condition of the treatment.
本发明融合蛋白特别适合用于治疗肿瘤等疾病。代表性的肿瘤包括(但并不限于):脑肿瘤、大肠癌肿瘤、肺癌肿瘤、肝癌肿瘤、乳腺癌肿瘤、胃癌肿瘤、胰腺癌肿瘤。The fusion proteins of the invention are particularly suitable for use in the treatment of diseases such as tumors. Representative tumors include, but are not limited to, brain tumors, colorectal cancer tumors, lung cancer tumors, liver cancer tumors, breast cancer tumors, gastric cancer tumors, and pancreatic cancer tumors.
本发明的主要优点包括:The main advantages of the invention include:
(a)本发明的融合蛋白对于T细胞无明显毒性。这可能是由于IL-12突出于T细胞膜外,无法有效接触或作用于T细胞,也可能是因为CD62L的间隔作用降低了IL-12对T细胞的毒性。(a) The fusion protein of the present invention has no significant toxicity to T cells. This may be due to the fact that IL-12 is prominent outside the T cell membrane and cannot be effectively contacted or acted on T cells, or it may be because the interstitial effect of CD62L reduces the toxicity of IL-12 to T cells.
(b)当携带本发明融合蛋白的抗肿瘤T细胞在接近肿瘤细胞时,由于无法通过特异性切割CD62L,从而不会释放出游离的IL-12(即IL-12保留在细胞膜表面)。这使得膜表面的IL-12能够更有效且只能作用于近距离的肿瘤细胞,从 而显著提高了安全性。(b) When the anti-tumor T cells carrying the fusion protein of the present invention are close to the tumor cells, since the CD62L cannot be specifically cleaved, free IL-12 is not released (i.e., IL-12 remains on the cell membrane surface). This makes IL-12 on the surface of the membrane more effective and can only act on close-range tumor cells, from And significantly improved security.
(c)通过T细胞攻击肿瘤的局部并通过细胞表面的IL-12改变T细胞-肿瘤组织的免疫微环境,从而协同而有效地实现抗肿瘤免疫效果的最大化。(c) Maximizing the anti-tumor immune effect synergistically and effectively by attacking the local part of the tumor by T cells and changing the immune microenvironment of the T cell-tumor tissue by IL-12 on the cell surface.
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,例如Sambrook等人,分子克隆:实验室手册(New York:Cold Spring Harbor Laboratory Press,1989)中所述的条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数是重量百分比和重量份数。The invention is further illustrated below in conjunction with specific embodiments. It is to be understood that the examples are not intended to limit the scope of the invention. Experimental methods in which the specific conditions are not indicated in the following examples are generally carried out according to the conditions described in conventional conditions, for example, Sambrook et al., Molecular Cloning: Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989), or according to the manufacturing conditions. The conditions recommended by the manufacturer. Unless otherwise stated, percentages and parts are by weight and parts by weight.
材料和通用方法Materials and general methods
所用引物及DNA序列均由Invitrogen公司合成。The primers and DNA sequences used were all synthesized by Invitrogen.
本发明实施例所使用的质粒LVV的图谱如图4所示,为含有目的基因的工程载体pLenti-MSCV,该载体由MSCV做为启动子,是最优化的可以有效转导T细胞的启动子;包膜蛋白质粒pMD2.G含有VSV-G;gag/pol辅助质粒;及pRev质粒系统。所使用的质粒pRRLSIN.cPPT.MSCV/GFP、293FT细胞为市售商品,所使用的试剂亦为市售商品。The map of the plasmid LVV used in the examples of the present invention is shown in Fig. 4, which is an engineering vector pLenti-MSCV containing a gene of interest, which is a promoter which is optimized for transduction of T cells by using MSCV as a promoter. The envelope protein particle pMD2.G contains VSV-G; gag/pol helper plasmid; and pRev plasmid system. The plasmids pRRLSIN.cPPT.MSCV/GFP and 293FT cells used were commercially available, and the reagents used were also commercially available.
实施例中,突变型CD62L指dKSCD62L。In the examples, the mutant CD62L refers to dKSCD62L.
hscIL-12/CD62L或IL-12/WTCD62L指hscIL-12/野生型CD62L。hscIL-12/CD62L or IL-12/WTCD62L refers to hscIL-12/wild type CD62L.
野生型CD62L表述为WTCD62L。Wild type CD62L is expressed as WTCD62L.
肿瘤细胞及T细胞的培养Tumor cell and T cell culture
肿瘤细胞938和526为常规的黑色素瘤细胞株(由美国国立癌症研究院的Dr.Rosenberg所赠),体外传代培养条件为含10%FCS RPMI培养基,每2-3天通过0.25%的胰酶传代培养。两种肿瘤均表达MART-1抗原,其中938为MHC I A2-(阴性),526为MHC I A2+(阳性),基因修饰的抗肿瘤MART-1T细胞只识别A2+细胞株,即526细胞。 Tumor cells 938 and 526 are conventional melanoma cell lines (supplied by Dr. Rosenberg of the National Cancer Institute) and subcultured in vitro with 10% FCS RPMI medium and 0.25% pancreas every 2-3 days. Enzyme subculture. Both tumors express MART-1 antigen, of which 938 is MHC I A2-(negative) and 526 is MHC I A2+ (positive), and the genetically modified anti-tumor MART-1 T cells recognize only A2+ cell line, ie 526 cells.
PBMC来自健康人外周血,用CD3/CD28磁珠或CD3抗体刺激T细胞生长1天,并在IL-2(100IU/ml)的X-VIVO培养基中维持培养。重组慢病毒转导T细胞后利用流式细胞仪通过MART-1四聚体肽段检测MART-1在T细胞的荧光强度。同时应用流式检测细胞表面CD62L、膜表面hscIL-12/突变型CD62L及CD107a的表达。PBMC were derived from healthy human peripheral blood, and T cells were stimulated for growth with CD3/CD28 magnetic beads or CD3 antibody for 1 day, and cultured in IL-2 (100 IU/ml) X-VIVO medium. After recombinant lentivirus transduction of T cells, the fluorescence intensity of MART-1 in T cells was detected by flow cytometry through the MART-1 tetramer peptide. At the same time, the expression of CD62L on the cell surface, hscIL-12/mutant CD62L and CD107a on the membrane surface were detected by flow cytometry.
慢病毒载体的构建Construction of lentiviral vector
采用分子生物学的技术方法将携带特异性识别人黑色素瘤相关抗原MART-1 的人源TCRα和β链基因的重组慢病毒载体转染外周血自体淋巴细胞,使重组TCR表达在T淋巴细胞中以达到高效杀伤肿瘤的目的。利用已优化启动子的慢病毒载体构建含自我剪切2A肽、弗林蛋白酶(Furin)及间隔序列的TCR表达载体。Using molecular biology techniques to carry a specific recognition of human melanoma-associated antigen MART-1 The recombinant lentiviral vector of human TCRα and β chain genes is transfected into autologous lymphocytes of peripheral blood, so that recombinant TCR is expressed in T lymphocytes to achieve the purpose of effectively killing tumors. A TCR expression vector containing a self-cleaving 2A peptide, furin (Furin) and a spacer sequence was constructed using a lentiviral vector with an optimized promoter.
表达野生型CD62L(简称为“WTCD62L”)和突变型CD62L(简称为“dKSCD62L”)的慢病毒表达载体的构建参照发表的文献(Yang,Cohen et al.2008,Yang,Liu et al.2011),hscIL-12的构建参考发表的文献(Zhang,Kerkar et al.2011)。其中hscIL-12/野生型CD62L和hscIL-12/突变型CD62L的融合蛋白均分别用肽段G6S及218肽段进行连接。Construction of a lentiviral expression vector expressing wild-type CD62L (abbreviated as "WTCD62L") and mutant CD62L (abbreviated as "dKSCD62L") is based on published literature (Yang, Cohen et al. 2008, Yang, Liu et al. 2011). The construction of hscIL-12 is based on published literature (Zhang, Kerkar et al. 2011). The fusion proteins of hscIL-12/wild type CD62L and hscIL-12/mutant CD62L were ligated with peptide G 6 S and 218 peptides, respectively.
慢病毒表达体系表达融合蛋白的制备Preparation of fusion protein expression in lentiviral expression system
培养293T细胞,转染前一天,以含10%胎牛血清的DMEM培养基调整细胞密度后,按照每15cm细胞培养皿接种25×106个293T细胞,置于37℃,5%CO2培养箱中培养,16h~24h后待细胞密度生长到80%~90%时即可用于转染。转染当天更换培养基为不含抗生素(P/S)的完全培养基(DMEM+10%FBS)。分别将LVV-MSCV-MART-1TCR、CD62L、hscIL-12L以及hscIL-12/突变型CD62L融合蛋白的慢病毒骨架与另外三种包装质粒一起共转染293T细胞,利用可以商业化的磷酸钙为媒介。培养6h后弃去培养基,用PBS洗3次后更换为20ml新鲜的完全培养基(DMEM+10%FBS+P/S)。收集转染后30-72h的培养上清,6000rpm离心10min,弃去细胞碎片,上清液以0.45μm PVDF滤器过滤至50ml圆底离心管中,4℃,50000g高速离心2h,小心弃去上清,DMEM(不含血清、双抗)重悬病毒沉淀,按每次使用的病毒量分装到洁净的15ml离心管中,-80℃冰箱保存,用于感染T细胞。Lentivirus-Associated p24ELISA Kit检测病毒的滴度为5×107-1.5×108IFU,具体步骤参见Lentivirus-Associated p24ELISA Kit的说明书。(Yang,Cohen et al.2008)。293T cells were cultured, and the cell density was adjusted in DMEM medium containing 10% fetal bovine serum one day before transfection. Then, 25×10 6 293T cells were inoculated per 15 cm cell culture dish, and cultured at 37 ° C, 5% CO 2 . The culture in the box can be used for transfection after the cell density is increased to 80% to 90% after 16h to 24h. On the day of transfection, the medium was changed to complete medium without antibiotics (P/S) (DMEM + 10% FBS). The lentiviral backbones of LVV-MSCV-MART-1TCR, CD62L, hscIL-12L, and hscIL-12/mutant CD62L fusion proteins were co-transfected into 293T cells together with three other packaging plasmids, using commercially available calcium phosphate. medium. After 6 hours of culture, the medium was discarded, washed 3 times with PBS and replaced with 20 ml of fresh complete medium (DMEM + 10% FBS + P / S). The culture supernatant of 30-72 h after transfection was collected, centrifuged at 6000 rpm for 10 min, and the cell debris was discarded. The supernatant was filtered through a 0.45 μm PVDF filter into a 50 ml round bottom centrifuge tube, centrifuged at 50,000 g for 2 h at 4 ° C, and carefully discarded. Clear, DMEM (free of serum, double antibody) resuspended virus precipitate, according to the amount of virus used each time into a clean 15ml centrifuge tube, stored in a -80 ° C refrigerator, used to infect T cells. The titer of the virus detected by the Lentivirus-Associated p24 ELISA Kit was 5×10 7 -1.5×10 8 IFU. For details, see the instructions of the Lentivirus-Associated p24 ELISA Kit. (Yang, Cohen et al. 2008).
肿瘤细胞/T细胞的共培养体系的建立Establishment of co-culture system of tumor cells/T cells
通过anti-MART-1TCR基因修饰的T细胞与526、938细胞系共培养,按1:1比例,即每种细胞1×106放置在14ml圆底的polypropylene培养管中,总体积1ml,转移至37℃的CO2培养箱中温浴4h。4h后,800×g离心细胞10分钟,收集上清,细胞通过RIPA裂解液裂解。上清中的IFNγ、IL-12的含量通过ELISA试剂检测。膜表面的IL-12/野生型CD62L或IL-12/突变型CD62L通过常规的流式细胞仪的活细胞染色进行操作。上清中的野生型或突变型CD62L及细胞内的野生型或突变型CD62L通过ELISA试剂盒检测(R&D Systems,Minneapolis,MN)。 T cells modified by anti-MART-1 TCR gene were co-cultured with 526, 938 cell lines, placed in a 1:1 ratio, ie 1×10 6 per cell, in a 14 ml round bottom polypropylene culture tube, total volume 1 ml, transfer The mixture was incubated in a CO 2 incubator at 37 ° C for 4 h. After 4 h, the cells were centrifuged at 800 x g for 10 minutes, the supernatant was collected, and the cells were lysed by RIPA lysate. The content of IFNγ and IL-12 in the supernatant was detected by an ELISA reagent. IL-12/wild-type CD62L or IL-12/mutant CD62L on the membrane surface was manipulated by live cell staining of a conventional flow cytometer. Wild-type or mutant CD62L in the supernatant and wild-type or mutant CD62L in the cells were detected by ELISA kit (R&D Systems, Minneapolis, MN).
流式细胞仪检测的分析Analysis by flow cytometry
细胞表面的CD3、CD8、CD62L、CD107a、IL-12和CD45RO通过荧光标记的对应的抗体进行检测,所应用的荧光抗体包括isothiocyanate(FITC),allophycocyanin(APC),phycoerythrin(PE),PE-Cy7,and APC-Cy7(BD Biosciences,San Jose,CA)。MART-1:27–35四聚体通过设计由公司合成(iTAg MHC Tetramer,Beckman Coulter,Fullerton,CA),用来检测基因修饰的TCR表达水平。具体流程如下,首先细胞经过FACS染色液(PBS containing2%FBS)洗涤两次,然后加入0.2ml(106/ml)于流式细胞管中,于4℃孵育30分钟,然后洗涤两次。死细胞通过样本上机前加入20μl PI(l5μg/ml propidium iodide)(Sigma-Aldrich,Saint Louis,MO)以及细胞亚群的划分来实现分离的目的。流式数据通过FlowJo 8.1.1进行上机后处理分析(FlowJo,Ashland,OR)。Cell surface CD3, CD8, CD62L, CD107a, IL-12 and CD45RO are detected by fluorescently labeled corresponding antibodies, including isothiocyanate (FITC), allophycocyanin (APC), phycoerythrin (PE), PE-Cy7. , and APC-Cy7 (BD Biosciences, San Jose, CA). The MART-1:27-35 tetramer was designed by the company (iTAg MHC Tetramer, Beckman Coulter, Fullerton, CA) to detect gene-modified TCR expression levels. The specific procedure is as follows. First, the cells were washed twice with FACS staining solution (PBS containing 2% FBS), then 0.2 ml (10 6 /ml) was added to the flow tube, incubated at 4 ° C for 30 minutes, and then washed twice. The dead cells were separated by adding 20 μl PI (l5 μg/ml propidium iodide) (Sigma-Aldrich, Saint Louis, MO) and cell subpopulation before the sample was taken to achieve the purpose of separation. The streaming data is analyzed by FlowJo 8.1.1 for post-processing (FlowJo, Ashland, OR).
荷瘤鼠模型的建立Establishment of tumor-bearing mouse model
Pmel实验小鼠模型采用常规方法,例如可参见文献(Overwijk,Tsung et al.1998)。本实验所涉及的方法如下:选择雌性pmel小鼠(6-8周,每组7只小鼠)进行颅内肿瘤接种(IC)。B16F10-MART-1肿瘤细胞通过含有0.02%EDTA的0.25%胰酶消化,并用含有血清的培养液洗涤一次来终止胰酶的反应,然后用PBS洗涤两次。肿瘤细胞最终以1:1的体积与methylcellulose in zinc option medium混合,5000个细胞稀释在5μl液体中上样到250-μl syringe(Hamilton,Reno,NV),选用25-gauge针头。应用Quitessential Stereotaxic Injector System(Stoelting Co.Wood Dale,IL)注射到小鼠的右侧脑caudate核中。接种肿瘤细胞5天后,小鼠接受全身5Gy的射线照射。第2天,小鼠皮下接受0.5-1X 106DC疫苗,或者通过尾静脉IV回输1X 107经过序贯转导anti-MART-1TCR、IL-12/野生型CD62L或IL-12/突变型CD62L慢病毒基因修饰的T细胞。鼠的T细胞取自小鼠的脾脏细胞,利用10ug/ml的刀豆蛋白(Con A)在IL-2(5IU/ml)存在的条件下活化;第2天,用慢病毒载体转导T细胞,然后继续培养6天,收集细胞,通过小鼠尾静脉注射T细胞。DC组小鼠的DC细胞取自小鼠的骨髓细胞,经过体外诱导分化成熟8天,通过腹腔接种。然后每天记录小鼠的死亡情况,记录生长曲线并通过Prism绘图软件制图。星号表示该实验组与其它组比较,p<0.001。The Pmel experimental mouse model uses conventional methods, for example, see the literature (Overwijk, Tsung et al. 1998). The method involved in this experiment was as follows: Female pmel mice (6-8 weeks, 7 mice per group) were selected for intracranial tumor inoculation (IC). B16F10-MART-1 tumor cells were stopped by 0.25% trypsinization with 0.02% EDTA and washed once with serum-containing medium to wash the trypsin reaction, followed by washing twice with PBS. Tumor cells were finally mixed with methylcellulose in zinc option medium in a volume of 1:1, and 5000 cells were diluted in a 5 μl liquid and loaded onto a 250-μl syringe (Hamilton, Reno, NV) using a 25-gauge needle. The Quinesential Stereotaxic Injector System (Stoelting Co. Wood Dale, IL) was injected into the right brain caudate nucleus of the mice. Five days after inoculation of the tumor cells, the mice received whole body 5 Gy radiation. On day 2, the mice received a 0.5-1×10 6 DC vaccine subcutaneously, or a 1X 10 7 retransmission of anti-MART-1 TCR, IL-12/wild-type CD62L or IL-12/mutation by tail vein IV. Type CD62L lentiviral gene-modified T cells. Mouse T cells were obtained from mouse spleen cells, activated with 10 ug/ml concanavalin (Con A) in the presence of IL-2 (5 IU/ml); day 2, transduced with Lentiviral vector T The cells were then cultured for 6 days, cells were collected, and T cells were injected through the tail vein of the mice. DC cells of DC group mice were obtained from mouse bone marrow cells, induced differentiation and maturation in vitro for 8 days, and inoculated intraperitoneally. The deaths of the mice were then recorded daily, growth curves were recorded and plotted by Prism mapping software. Asterisks indicate that the experimental group was compared with the other groups, p < 0.001.
实施例1Example 1
融合基因的构建Fusion gene construction
融合基因由Invitrogen公司合成,融合基因的长度及序列通过1%的琼脂 糖电泳及测序证实。The fusion gene was synthesized by Invitrogen, and the length and sequence of the fusion gene was passed through 1% agar. Glycoelectrophoresis and sequencing confirmed.
构建获得的IL-12/突变型CD62L融合基因的结构如SEQ ID NO.:1所示,所编码的融合蛋白的氨基酸序列如SEQ ID NO.:2所示。The structure of the obtained IL-12/mutant CD62L fusion gene was constructed as shown in SEQ ID NO.: 1, and the amino acid sequence of the encoded fusion protein is shown in SEQ ID NO.: 2.
同时构建获得的IL-12/野生型CD62L融合基因,所述IL-12/野生型CD62L融合基因与SEQ ID NO.:1非常相似,不同点仅在于:保留了编码切割位点的核苷酸序列,故所编码的融合蛋白也保留了ADAM17切割位点(即KLDKSFS)。The obtained IL-12/wild type CD62L fusion gene was constructed at the same time, and the IL-12/wild type CD62L fusion gene was very similar to SEQ ID NO.: 1, except that the nucleotide encoding the cleavage site was retained. The sequence, so the encoded fusion protein also retains the ADAM17 cleavage site (ie KLDKSFS).
实施例2Example 2
慢病毒表达载体的构建Construction of lentiviral expression vector
采用通用方法中“慢病毒载体的构建及T细胞的基因修饰”所述的方法:体外低代DMEM(含10%的FBS)培养基培养的人源化293T细胞经计数后,传到15CM的培养皿中,培养皿的底部经过poly-D-Lysine处理,每个培养皿中铺20X 106细胞,第二天,每个转染培养皿中加入由DNA混合液和Lipofectamine的混合液:混合液的组成如下,取2ml Optimum I并加入pLenti-MSCV(22.5ug),pMD2.G(7.5ug),gag/pol(15ug),pRev(10ug),混匀;同时取2ml Optimum I并加入Lipofectamine 160ul(Invitrogen),混匀。把两种混悬液混合,放置室温孵育5分钟,然后均匀地滴加到培养皿中。48-72小时后,收获含有基因工程载体的上清,2000g离心去除细胞碎片,收集上清,并用0.45uM的滤膜过滤去除可能的污染,分装并存放于负80冰箱。根据不同的需要,收集的病毒上清可以进行50000g超速离心,得到更高浓度的病毒载体。The method described in "General Construction of Lentiviral Vector and Gene Modification of T Cells" in a general method: humanized 293T cells cultured in a low-generation DMEM (containing 10% FBS) medium were counted and passed to 15 CM. In the culture dish, the bottom of the culture dish was treated with poly-D-Lysine, and 20×10 6 cells were plated in each dish. The next day, a mixture of DNA mixture and Lipofectamine was added to each transfection dish: mixing The composition of the solution was as follows. Take 2 ml of Optimum I and add pLenti-MSCV (22.5 ug), pMD2.G (7.5 ug), gag/pol (15 ug), pRev (10 ug), and mix; take 2 ml of Optimum I and add Lipofectamine. 160ul (Invitrogen), mix. The two suspensions were mixed, incubated at room temperature for 5 minutes, and then uniformly added dropwise to the Petri dish. After 48-72 hours, the supernatant containing the genetically engineered vector was harvested, centrifuged to remove cell debris at 2000 g, and the supernatant was collected and filtered through a 0.45 uM filter to remove possible contamination, dispensed and stored in a negative 80 refrigerator. According to different needs, the collected virus supernatant can be subjected to ultracentrifugation at 50,000 g to obtain a higher concentration of the viral vector.
获得的慢病毒表达载体分别命名为LV-hscIL-12/野生型CD62L(表达IL-12/CD62L)、LV-hscIL-12/突变型CD62L(表达IL-12/dKSCD62L)、LV-hscIL-12。The obtained lentiviral expression vectors were named LV-hscIL-12/wild type CD62L (expressing IL-12/CD62L), LV-hscIL-12/mutant CD62L (expressing IL-12/dKSCD62L), LV-hscIL-12, respectively. .
实施例3Example 3
表达IL-12/突变型CD62L融合蛋白的T细胞的制备Preparation of T cells expressing IL-12/mutant CD62L fusion protein
方法如下:CD3/CD28磁珠或抗-CD3抗体激活PBMC,第2天,利用慢病毒基因修饰T细胞,简要的方法如下:PBS缓冲液洗涤T细胞3次,按病毒滴度与T细胞的比例3:1加入适量的慢病毒,2000X g离心2h,6h后,加入100IU/ml IL-2继续培养;于第5天进行第2次转导,或联合共转导。根据细胞生长情况进行分瓶,二周后,根据T细胞所修饰的基因通过流式细胞仪进行检查。The method is as follows: CD3/CD28 magnetic beads or anti-CD3 antibody activates PBMC, and on day 2, the T cells are modified by lentiviral gene. The brief method is as follows: Wash T cells three times in PBS buffer, according to virus titer and T cell The appropriate amount of lentivirus was added in a ratio of 3:1, centrifuged at 2000×g for 2 h, and after 6 h, the culture was continued by adding 100 IU/ml IL-2; the second transduction was performed on the 5th day, or combined co-transduction. The flask was divided according to the growth of the cells, and two weeks later, the cells modified by the T cells were examined by flow cytometry.
实施例4Example 4
dKSCD62L慢病毒基因修饰的抗肿瘤T细胞实现dKSCD62L稳定表达dKSCD62L lentiviral gene-modified anti-tumor T cells achieve stable expression of dKSCD62L
为了更好地观察慢病毒基因介导的外源性CD62L、dKSCD62L蛋白的表达,从 而区别内源性生理性的CD62L的表达,选用了一种黑色素瘤组织中分离培养的肿瘤浸润T细胞系JKF6。JKF6细胞系可以长期体外传代培养,并且膜表面丧失了CD62L膜分子,JKF6可以有效地特异性识别黑色素瘤细胞。In order to better observe the expression of exogenous CD62L and dKSCD62L proteins mediated by lentiviral genes, To distinguish the expression of endogenous physiological CD62L, a tumor infiltrating T cell line JKF6 isolated and cultured in melanoma tissue was selected. The JKF6 cell line can be subcultured in vitro for a long time, and the membrane surface loses the CD62L membrane molecule, and JKF6 can effectively recognize melanoma cells.
JKF6通过慢病毒基因修饰后,可以检测到高水平的外源性WTCD62L及dKSCD62L的表达。野生型CD62L及突变型dKSCD62L基因修饰的JKF6获得了稳定的膜表面CD62L系列表达。JKF6 can detect high levels of exogenous WTCD62L and dKSCD62L expression by lentiviral gene modification. Wild-type CD62L and mutant dKSCD62L gene-modified JKF6 obtained stable membrane surface CD62L series expression.
通过与肿瘤抗原526活化后,野生型CD62L可以发生肿瘤抗原特异性的切割及释放;但是,dKSCD62L的膜表达后,却丧失肿瘤抗原活化的切割及释放,表现为稳定的膜表面dKSCD62L的表达的表型,如图2、图3所示,共培养的T细胞通过流式细胞仪检测膜表面的分子CD45RO和CD62L,通过FlowJo软件的分析处理,充分验证了CD62L突变体的膜稳定性。After activation with tumor antigen 526, wild-type CD62L can undergo tumor antigen-specific cleavage and release; however, after expression of dKSCD62L membrane, it loses the cleavage and release of tumor antigen activation, which is expressed as stable membrane surface dKSCD62L expression. Phenotype, as shown in Fig. 2 and Fig. 3, the co-cultured T cells were detected by flow cytometry on the surface of the membrane CD45RO and CD62L, and the membrane stability of the CD62L mutant was fully verified by FlowJo software analysis.
实施例5Example 5
dKSCD62L慢病毒基因修饰抗肿瘤JFK6细胞不影响T细胞的抗肿瘤活性dKSCD62L lentiviral gene modification against tumor JFK6 cells does not affect the antitumor activity of T cells
为了验证经过dKSCD62L慢病毒基因修饰JKF6的反应性,也考虑到CD62L体内的切割可能存在其它途径,在本实施例中,进一步验证T细胞的抗肿瘤活性。方法如下:In order to verify the reactivity of the dKSCD62L lentiviral gene-modified JKF6, it is also considered that there may be other pathways for cleavage in CD62L in vivo, and in this example, the anti-tumor activity of T cells is further verified. Methods as below:
选用了广谱的T细胞激活剂PMA/Ionomycin(简称为“PMA/Ion”,它是一种T细胞活化剂),用所述激活剂激活T细胞4小时后,我们检测到了细胞膜上CD62L(野生型或突变型)的表达。此外,还发现,对于突变型dKSCD62L而言,活化诱导的膜切割消失,即获得了稳定的膜表面表达dKSCD62L;对于野生型CD62L而言,T细胞活化切割依然存在。A broad-spectrum T cell activator PMA/Ionomycin (abbreviated as "PMA/Ion", a T cell activator) was used, and after activation of T cells with the activator for 4 hours, we detected CD62L on the cell membrane ( Expression of wild type or mutant type). In addition, it was also found that for the mutant dKSCD62L, activation-induced membrane cleavage disappeared, that is, stable membrane surface expression of dKSCD62L was obtained; for wild-type CD62L, T cell activation cleavage remained.
为了验证T细胞的反应性,测定经CD62L基因修饰后的反应性。结果如图3所示,经突变型CD62L基因(或融合蛋白基因)修饰的JKF6活化后可以表达高水平的IFNγ。该实验结果提示,经过融合蛋白基因修饰后,抗肿瘤JKF6的反应性正常,没有受到基因修饰的影响。To verify the reactivity of T cells, the reactivity after modification with the CD62L gene was determined. As a result, as shown in Fig. 3, JKF6 modified by the mutant CD62L gene (or fusion protein gene) can express high levels of IFNγ after activation. The results of this experiment suggest that after modification of the fusion protein gene, the anti-tumor JKF6 has normal reactivity and is not affected by genetic modification.
实施例6Example 6
慢病毒表达dKSCD62L融合蛋白载体的构建及在人T细胞中的表达Construction of lentiviral expression dKSCD62L fusion protein vector and its expression in human T cells
在本实施例中,构建了IL-12/CD62L、IL-12/dKSCD62L的慢病毒载体(结构如图4所示),其中IL-12基因与突变型或野生型CD62L基因通过氨基酸肽段G6S链接,所述慢病毒为第三代慢病毒载体,启动子为MSCV。该载体的5’及3’LTR(long terminal repeat)被改造成SIN-LTR(self-inactivating-LTR),以降低慢病毒重组的机率,增强安全性能。In this example, a lentiviral vector (structure shown in Figure 4) of IL-12/CD62L, IL-12/dKSCD62L was constructed, in which the IL-12 gene and the mutant or wild type CD62L gene pass the amino acid peptide G 6 S link, the lentivirus is a third generation lentiviral vector, and the promoter is MSCV. The 5' and 3' LTR (long terminal repeat) of the vector was transformed into SIN-LTR (self-inactivating-LTR) to reduce the probability of lentiviral recombination and enhance safety performance.
为了明确IL-12/dKSCD62L融合蛋白在抗肿瘤T细胞的表达情况,依次对T细 胞转导了抗肿瘤TCR及IL-12/dKSCD62L融合蛋白,抗肿瘤T细胞体外培养14天后与肿瘤细胞526、938共培养。In order to clarify the expression of IL-12/dKSCD62L fusion protein in anti-tumor T cells, The anti-tumor TCR and IL-12/dKSCD62L fusion proteins were transduced, and the anti-tumor T cells were co-cultured with tumor cells 526 and 938 after 14 days of in vitro culture.
结果观察到了IL-12/dKSCD62L的稳定膜表达的表型,同时也进一步证实了IL-12/CD62L的肿瘤抗原依赖性的特异性切割及释放。As a result, the phenotype of stable membrane expression of IL-12/dKSCD62L was observed, and the tumor antigen-dependent specific cleavage and release of IL-12/CD62L was further confirmed.
实施例7Example 7
慢病毒载体IL-12/CD62L系列转导的T细胞与肿瘤共培养可以增强IFNγ的表达Lentiviral vector IL-12/CD62L transduced T cells co-culture with tumor can enhance the expression of IFNγ
在本发明中,为了降低IL-12释放诱导产生的系统性细胞毒性,构建了稳定膜表达IL-12/dKSCD62L的融合蛋白载体。In the present invention, in order to reduce systemic cytotoxicity induced by IL-12 release, a fusion protein vector stably expressing IL-12/dKSCD62L is constructed.
如图5所示,对人的T细胞依次转导抗肿瘤TCR及IL-12/CD62L系列融合蛋白14天后,与肿瘤细胞526及938共培养,24小时后,上清中IFNγ及IL-12表达水平通过ELISA试剂盒检测。As shown in Fig. 5, human T cells were sequentially transduced with anti-tumor TCR and IL-12/CD62L series fusion proteins for 14 days, and co-cultured with tumor cells 526 and 938. After 24 hours, the supernatants were IFNγ and IL-12. Expression levels were detected by ELISA kit.
结果显示,持续性IL-12分泌组、IL-12/野生型CD62L组及IL-12/dKSCD62L组较T细胞组比较,可以显著增强抗肿瘤T细胞与肿瘤的反应性,表现为显著增强IFNγ分泌的水平,P<0.001。The results showed that the sustained IL-12 secretion group, IL-12/wild type CD62L group and IL-12/dKSCD62L group could significantly enhance the reactivity of anti-tumor T cells with tumors compared with the T cell group, which showed a significant enhancement of IFNγ. The level of secretion, P < 0.001.
同dKSCD62L膜表面的稳定表达情况一致,IL-12/dKSCD62L也丧失了肿瘤抗原反应性的切割及释放,与IL-12/野生型CD62L组、持续性分泌IL-12组比较,可以检测到的分泌的IL-12的水平显著降低,P<0.001。Consistent with the stable expression of the surface of dKSCD62L membrane, IL-12/dKSCD62L also lost the cleavage and release of tumor antigen reactivity, which was detectable compared with IL-12/wild type CD62L group and persistent secretion IL-12 group. The level of secreted IL-12 was significantly reduced, P < 0.001.
因此,这证实了稳定膜表达IL-12/dKSCD62L是可行的,更进一步证实该稳定膜表达IL-12/dKSCD62L的抗肿瘤T细胞与肿瘤细胞共培养后,T细胞的肿瘤抗原反应性没有发生改变,有助于实现IL-12抗肿瘤效果最大化。Therefore, this confirmed that it is feasible to express IL-12/dKSCD62L in a stable membrane, and further confirmed that the tumor antigen reactivity of T cells did not occur after co-culture of anti-tumor T cells expressing IL-12/dKSCD62L with the tumor cells. Changes can help maximize the anti-tumor effect of IL-12.
实施例8Example 8
慢病毒载体IL-12/dKSCD62L转导的T细胞可以有效避免持续性分泌IL-12造成的体外T细胞扩增的毒副作用Lentiviral vector IL-12/dKSCD62L transduced T cells can effectively avoid the side effects of in vitro T cell expansion caused by persistent secretion of IL-12.
在解决了稳定的膜表达及增强抗肿瘤反应性的问题后,本实施例观察转导该IL-12/dKSCD62L融合蛋白对T细胞体外扩增的影响。After solving the problem of stable membrane expression and enhancing anti-tumor reactivity, this example observes the effect of transduction of the IL-12/dKSCD62L fusion protein on T cell expansion in vitro.
如图6所示,对T细胞依次转导抗肿瘤TCR及野生型或突变型CD62L/hscIL-12融合蛋白14天后,各组细胞与对照转导组(T-cell)的扩增倍数比较,数据分析采用设定T-cell组的扩增倍数为100%,其它各组的值系与T-cell组的比值。As shown in Figure 6, after T cells were sequentially transduced with anti-tumor TCR and wild-type or mutant CD62L/hscIL-12 fusion protein for 14 days, the expansion ratios of the cells in each group were compared with the control transduction group (T-cell). The data analysis was performed by setting the amplification factor of the T-cell group to 100%, and the values of the other groups were compared with the T-cell group.
结果表明,转导持续分泌IL-12组较其它各组的扩增倍数显著降低,p<0.001。其中,转导hscIL-12/dKSCD62L融合蛋白组同其它组比较扩增倍数相同,未见显著差异。另外,转导hscIL-12/dKSCD62L融合蛋白组的扩增倍数略高于转导了hscIL-12/野生型CD62L融合蛋白组。这提示转导了,由于不释放游离的IL-12, 导致IL-12对T细胞几乎无明显毒性。The results showed that the number of amplifications in the transduced IL-12 group was significantly lower than that in the other groups, p < 0.001. Among them, the transduced hscIL-12/dKSCD62L fusion protein group was the same as the other groups, and no significant difference was observed. In addition, the amplification fold of the hscIL-12/dKSCD62L fusion protein group was slightly higher than that of the hscIL-12/wild type CD62L fusion protein group. This suggests that the transduction is due to the release of free IL-12. This resulted in almost no significant toxicity of IL-12 to T cells.
实施例9Example 9
慢病毒载体hscIL-12/dLSCD62L系列转导鼠T细胞介导的细胞回输治疗显著延长荷瘤小鼠的生存Lentiviral vector hscIL-12/dLSCD62L series transduced mouse T cell-mediated cell transfusion therapy significantly prolongs survival of tumor-bearing mice
在清楚了IL-12/dKSCD62L的表达、膜稳定性、肿瘤抗原反应的增效性及体外扩增的基础上,在本实施例中,进一步采用了临床前荷瘤小鼠模型,观测体内抑瘤效应。方法如下:Based on the clear expression of IL-12/dKSCD62L, membrane stability, synergistic effect of tumor antigen response and in vitro expansion, in this example, a pre-clinical tumor-bearing mouse model was further used to observe the inhibition in vivo. Tumor effect. Methods as below:
选择雌性pmel小鼠(每组7只小鼠)通过B16F10细胞植入颅内(IC)5天,细胞回输前1天小鼠接受5Gy全身放疗。鼠的T细胞取自小鼠的脾脏细胞,通过10ug/ml的刀豆蛋白(Con A)在IL-2(5IU/ml)存在的条件下活化;第2天,用慢病毒载体转导T细胞,然后继续培养6天,收集细胞,通过小鼠尾静脉注射(IV)5X 106个T细胞。DC组小鼠的DC细胞取自骨髓细胞,经过体外诱导分化成熟8天,通过腹腔接种1X 106细胞。DC和T细胞的各组说明见右边标识。星号(*)表示该实验组与其它组比较,p<0.001。Female pmel mice (7 mice per group) were selected for intracranial (IC) implantation by B16F10 cells for 5 days, and mice received 5 Gy whole body radiotherapy 1 day before cell return. Mouse T cells were obtained from mouse spleen cells and activated by 10 ug/ml concanavalin (Con A) in the presence of IL-2 (5 IU/ml); on day 2, transduction of T with lentiviral vector The cells were then cultured for 6 days, cells were collected, and (X) 5 ×10 6 T cells were injected through the tail vein of the mice. The DC cells of the DC group were obtained from bone marrow cells, and induced to differentiate and mature for 8 days in vitro, and 1×10 6 cells were inoculated intraperitoneally. The group description of DC and T cells is shown on the right. An asterisk (*) indicates that the experimental group was compared with the other groups, p < 0.001.
如图7所示。结果表明,与其它对照组相比,IL-12/dKSCD62L组显著地延长了荷瘤小鼠的生存期;持续分泌IL-12组表现为显著的系统性细胞毒性,小鼠于细胞回输后4-7天全部死亡。As shown in Figure 7. The results showed that compared with the other control groups, the IL-12/dKSCD62L group significantly prolonged the survival of tumor-bearing mice; the sustained secretion of IL-12 group showed significant systemic cytotoxicity, after the mice returned to the cells. All died in 4-7 days.
讨论discuss
CD62L是T细胞表面表达的归巢因子,可以通过T细胞的活化诱导的切割来实现T细胞沿着血管壁的黏附和释放。研究发现,缺乏ADAM17酶(tumor Necrosis factor-converting enzyme 17)的T细胞导致CD62L不能进行切割及释放。CD62L is a homing factor for T cell surface expression, which can achieve adhesion and release of T cells along the vessel wall by activation-induced cleavage of T cells. Studies have found that T cells lacking the tumor necrosis factor-converting enzyme 17 cause CD62L to be cleavable and released.
在本发明中,本发明人敲除了CD62L的ADAM17的切割点(本发明中命名为dKSCD62L),实验结果表明,通过基因修饰抗肿瘤T细胞表达新的IL-12/突变型CD62L分子,不仅可高效地杀灭肿瘤细胞,更可显著降低IL-12的毒副作用。In the present invention, the present inventors knocked out the cleavage point of ADAM17 of CD62L (designated as dKSCD62L in the present invention), and the experimental results indicate that the expression of a novel IL-12/mutant CD62L molecule by genetically modifying anti-tumor T cells is not only Efficient killing of tumor cells can significantly reduce the side effects of IL-12.
与持续分泌IL-12基因修饰的抗肿瘤T细胞相比较,IL-12/突变型CD62L融合蛋白基因修饰的抗肿瘤T细胞不但可以显著延长荷瘤鼠的生存,还可以避免持续分泌IL-12诱导的系统性细胞毒性,是一种全新的通过细胞膜表达IL-12,并通过CD62L来实现肿瘤抗原反应性的切割及释放的全新的免疫细胞治疗策略,在肿瘤的免疫细胞的治疗中将发挥重要的作用。Compared with anti-tumor T cells that continuously secrete IL-12 gene, IL-12/mutant CD62L fusion protein gene-modified anti-tumor T cells can not only prolong the survival of tumor-bearing mice, but also avoid the sustained secretion of IL-12. Induced systemic cytotoxicity is a brand new immune cell therapy strategy that expresses IL-12 through cell membrane and cleaves and releases tumor antigen reactivity through CD62L. It will play a role in the treatment of tumor immune cells. Important role.
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 the present application are hereby incorporated by reference in their entirety in their entireties in the the the the the the the the In addition, it should be understood that after reading the above teachings of the present invention, A person skilled in the art can make various changes or modifications to the invention, and such equivalents are also within the scope defined by the appended claims.
参考文献references
1.Zhang,L.等人(2011)."Improving adoptive T cell therapy by targeting and controlling IL-12expression to the tumor environment."Mol Ther 19(4):751-759.1. Zhang, L. et al. (2011). "Improving adoptive T cell therapy by targeting and controlling IL-12 expression to the tumor environment." Mol Ther 19(4): 751-759.
2.Overwijk,W.W.等人(1998)."gp100/pmel 17is a murine tumor rejection antigen:induction of"self"-reactive,tumoricidal T cells using high-affinity,altered peptide ligand."J Exp Med 188(2):277-286.2. Overwijk, WW et al. (1998). "gp100/pmel 17is a murine tumor rejection antigen: induction of "self"-reactive,tumoricidal T cells using high-affinity,altered peptide ligand."J Exp Med 188(2) :277-286.
3.Yang,S.等人(2008)."Development of optimal bicistronic lentiviral vectors facilitates high-level TCR gene expression and robust tumor cell recognition."Gene Ther 15(21):1411-1423.3. Yang, S. et al. (2008). "Development of optimal bicistronic lentiviral vectors facilitats high-level TCR gene expression and robust tumor cell recognition." Gene Ther 15(21): 1411-1423.
4.Yang,S.,等人(2011)."The shedding of CD62L(L-selectin)regulates the acquisition of lytic activity in human tumor reactive T lymphocytes."PLoS One 6(7):e22560. 4.Yang, S., et al. (2011). "The shedding of CD62L(L-selectin)regulates the acquisition of lytic activity in human tumor reactive T lymphocytes."PLoS One 6(7):e22560.
Figure PCTCN2016080418-appb-000001
Figure PCTCN2016080418-appb-000001
Figure PCTCN2016080418-appb-000002
Figure PCTCN2016080418-appb-000002
Figure PCTCN2016080418-appb-000003
Figure PCTCN2016080418-appb-000003
Figure PCTCN2016080418-appb-000004
Figure PCTCN2016080418-appb-000004
Figure PCTCN2016080418-appb-000005
Figure PCTCN2016080418-appb-000005
Figure PCTCN2016080418-appb-000006
Figure PCTCN2016080418-appb-000006

Claims (10)

  1. 一种融合蛋白,其特征在于,所述融合蛋白包括融合在一起的以下元件:A fusion protein characterized in that the fusion protein comprises the following elements fused together:
    (i)任选的位于N端的信号肽和/或前导肽;(i) an optional signal peptide and/or leader peptide at the N-terminus;
    (ii)第一蛋白元件;(ii) a first protein component;
    (iii)第二蛋白元件;以及(iii) a second protein component;
    (iv)任选的位于第一蛋白元件和第二蛋白元件之间的连接肽元件;(iv) an optional linker element located between the first protein element and the second protein element;
    其中,所述信号肽可操作地连于由(ii)、(iii)和(iv)所构成的融合元件;Wherein the signal peptide is operably linked to the fusion element consisting of (ii), (iii) and (iv);
    并且第一蛋白元件为IL-12蛋白元件;第二蛋白元件为突变型CD62L的蛋白元件,所述突变型CD62L的蛋白元件缺失了ADAM17的切割位点。And the first protein element is an IL-12 protein element; the second protein element is a protein element of the mutant CD62L, and the protein element of the mutant CD62L lacks a cleavage site of ADAM17.
  2. 如权利要求1所述的融合蛋白,其特征在于,所述的融合蛋白具有选自下组的结构:The fusion protein according to claim 1, wherein said fusion protein has a structure selected from the group consisting of:
    (1)式Ia所述结构:(1) Structure described by Formula Ia:
    D-A-B   (Ia),或D-A-B (Ia), or
    (2)式Ⅱa所述结构:(2) Structure described in Formula IIa:
    D-A-C-B   (Ⅱa),D-A-C-B (IIa),
    其中,among them,
    A为IL-12蛋白元件;A is an IL-12 protein element;
    B为突变型CD62L蛋白元件;B is a mutant CD62L protein element;
    C为任选的连接肽元件;C is an optional linker element;
    D为任选的信号肽信号肽和/或前导肽序列;D is an optional signal peptide signal peptide and/or leader peptide sequence;
    各“-”独立地表示连接上述元件的肽键或肽接头。Each "-" independently indicates a peptide bond or a peptide linker to which the above elements are attached.
  3. 一种分离的多核苷酸,其特征在于,所述的多核苷酸编码权利要求1所述的融合蛋白。An isolated polynucleotide, characterized in that the polynucleotide encodes the fusion protein of claim 1.
  4. 一种载体,其特征在于,它含有权利要求3所述的多核苷酸。A vector comprising the polynucleotide of claim 3.
  5. 一种宿主细胞,其特征在于,它含有权利要求4所述的载体或基因组中整合有权利要求3所述的多核苷酸。A host cell comprising the vector of claim 4 or a polynucleotide in which the polynucleotide of claim 3 is integrated.
  6. 一种产生权利要求1所述的蛋白的方法,它包括步骤:A method of producing the protein of claim 1 comprising the steps of:
    (1)在适合表达的条件下,培养权利要求5所述的宿主细胞,从而表达出权利要求1所述的融合蛋白;和 (1) cultivating the host cell of claim 5 under conditions suitable for expression, thereby expressing the fusion protein of claim 1;
    (2)任选地分离所述融合蛋白。(2) Optionally isolating the fusion protein.
  7. 一种免疫细胞,其特征在于,所述的免疫细胞在膜表面上携带权利要求1所述的融合蛋白。An immune cell characterized in that said immune cell carries the fusion protein of claim 1 on the surface of the membrane.
  8. 一种药物组合物,其特征在于,所述的组合物包含:A pharmaceutical composition, characterized in that the composition comprises:
    权利要求7所述的免疫细胞,以及The immune cell of claim 7;
    药学上可接受的载体。A pharmaceutically acceptable carrier.
  9. 一种如权利要求1所述的融合蛋白和/或权利要求7所述的免疫细胞的用途,其特征在于,用于制备治疗肿瘤的药物。Use of the fusion protein of claim 1 and/or the immune cell of claim 7 for the preparation of a medicament for treating a tumor.
  10. 一种非治疗性的体外杀灭肿瘤细胞的方法,其特征在于,包括步骤:将权利要求7所述的免疫细胞与肿瘤细胞进行接触,从而杀灭所述肿瘤细胞。 A non-therapeutic method for killing tumor cells in vitro, comprising the steps of: contacting the immune cells of claim 7 with tumor cells to kill the tumor cells.
PCT/CN2016/080418 2015-12-23 2016-04-27 Cancer treatment agent, preparation method and use thereof employing il-12 with stable membrane expression WO2017107353A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201510979423.7A CN105585637B (en) 2015-12-23 2015-12-23 Tumor therapeutic agent based on IL-12 stable membrane expression and preparation method and application thereof
CN201510979423.7 2015-12-23

Publications (1)

Publication Number Publication Date
WO2017107353A1 true WO2017107353A1 (en) 2017-06-29

Family

ID=55925568

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/080418 WO2017107353A1 (en) 2015-12-23 2016-04-27 Cancer treatment agent, preparation method and use thereof employing il-12 with stable membrane expression

Country Status (2)

Country Link
CN (1) CN105585637B (en)
WO (1) WO2017107353A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115505600B (en) * 2022-09-30 2024-01-30 北京奇迈永华生物科技有限公司 Method for efficiently infecting human NK cells by slow viruses

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1318644A (en) * 2000-04-18 2001-10-24 西安医科大学 Construction process of recombined human single-chain interleukin-12
CN1432064A (en) * 2000-03-23 2003-07-23 格林维尔医院系统公司 Bi-functional cancer treatment agents
WO2003092737A1 (en) * 2002-04-30 2003-11-13 Molmed Spa Fusions of cytokines and tumor targeting proteins
CN105218682A (en) * 2015-10-26 2016-01-06 深圳精准医疗科技有限公司 Through tumor therapeutic agent and method for making and the purposes of the transformation of IL-12/CD62L fusion rotein

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050180951A1 (en) * 2004-02-12 2005-08-18 Tsuneya Ohno Combined immunotherapy of fusion cells and interleukin-12 for treatment of cancer

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1432064A (en) * 2000-03-23 2003-07-23 格林维尔医院系统公司 Bi-functional cancer treatment agents
CN1318644A (en) * 2000-04-18 2001-10-24 西安医科大学 Construction process of recombined human single-chain interleukin-12
WO2003092737A1 (en) * 2002-04-30 2003-11-13 Molmed Spa Fusions of cytokines and tumor targeting proteins
CN105218682A (en) * 2015-10-26 2016-01-06 深圳精准医疗科技有限公司 Through tumor therapeutic agent and method for making and the purposes of the transformation of IL-12/CD62L fusion rotein

Also Published As

Publication number Publication date
CN105585637A (en) 2016-05-18
CN105585637B (en) 2020-04-03

Similar Documents

Publication Publication Date Title
CN108018299B (en) Chimeric antigen receptor targeting BCMA and uses thereof
US20210177902A1 (en) Ror-1 specific chimeric antigen receptors and uses thereof
CN109306016B (en) NKG2D-CAR-T cells co-expressing cytokine IL-7 and uses thereof
CN108004259B (en) Chimeric antigen receptor targeting B cell maturation antigen and uses thereof
CN109320615B (en) Chimeric antigen receptor targeting novel BCMA and uses thereof
BR112016028644B1 (en) IN VITRO OR EX VIVO METHOD FOR MANUFACTURING T CELLS, COMPOSITION UNDERSTANDING A CRYOPROTECTION AGENT AND A POPULATION OF IMMUNE EFFECTIVE CELLS AND THERAPEUTIC USES DADITA COMPOSITION
WO2017071173A1 (en) Tumor therapeutic agent modified by il-12/cd62l fusion protein and preparation method and use thereof
CN113913379B (en) T lymphocyte and application thereof
MX2014003176A (en) Rna engineered t cells for the treatment of cancer.
US20220153867A1 (en) Muc16 specific chimeric antigen receptors and uses thereof
WO2018006881A1 (en) Recombinant immune-checkpoint receptor and application thereof
CN113416260B (en) Claudin18.2-targeted specific chimeric antigen receptor cell and preparation method and application thereof
JP2023535264A (en) Chimeric antigen receptor and use thereof
CN113896801B (en) Chimeric antigen receptor cell targeting human Claudin18.2 and NKG2DL, and preparation method and application thereof
CN111378625A (en) Preparation and application of CXCL13 chemotactic CAR-T cell
CN110923255A (en) Chimeric antigen receptor targeting BCMA and CD19 and uses thereof
JP2023548957A (en) New PiggyBac transposon system and its application
WO2020211330A1 (en) Ubiquitination-deficiency chimeric antigen receptor and uses thereof
WO2017107353A1 (en) Cancer treatment agent, preparation method and use thereof employing il-12 with stable membrane expression
CN110699371A (en) Fc gamma RIIa-based chimeric gene and application thereof
CN111499766B (en) Immune effector cell aiming at chronic lymphocytic leukemia, preparation method and application thereof
WO2017117890A1 (en) Il-12/cd107a fusion protein and preparation method and use thereof
CN108165568B (en) Method for culturing CD19CAR-iNKT cells and application
CN115975056B (en) Construction of TSHR-targeted CAR-T cells using the native protein TSH as an antigen binding site
CN110938642B (en) Chimeric antigen receptor targeting CD123-BBz-IL1RN

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: 16877167

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16877167

Country of ref document: EP

Kind code of ref document: A1