WO2017071173A1 - Agent thérapeutique antitumoral modifié par protéine de fusion il-12/cd62l, son procédé de préparation et son utilisation - Google Patents

Agent thérapeutique antitumoral modifié par protéine de fusion il-12/cd62l, son procédé de préparation et son utilisation Download PDF

Info

Publication number
WO2017071173A1
WO2017071173A1 PCT/CN2016/080420 CN2016080420W WO2017071173A1 WO 2017071173 A1 WO2017071173 A1 WO 2017071173A1 CN 2016080420 W CN2016080420 W CN 2016080420W WO 2017071173 A1 WO2017071173 A1 WO 2017071173A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
cells
cd62l
fusion protein
tumor
Prior art date
Application number
PCT/CN2016/080420
Other languages
English (en)
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 WO2017071173A1 publication Critical patent/WO2017071173A1/fr

Links

Images

Classifications

    • 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
    • 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
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/861Adenoviral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/867Retroviral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention relates to the field of medicine, in particular to a tumor therapeutic agent modified by an IL-12/CD62L fusion protein, a preparation method thereof and use thereof.
  • the present invention provides a class of IL-12/CD62L gene-modified T cells with enhanced anti-tumor effects.
  • 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 present inventors have also succeeded in the effective amplification of T cells in vitro by TCR activation of a gene modification scheme that specifically regulates the expression of IL-12, and have also achieved certain antitumor effects, but at the same time, IL exists.
  • the toxic side effects caused by -12 out of regulation cannot be effectively applied to clinical applications.
  • the object of the present invention is to provide a tumor therapeutic drug with high efficiency and low toxic and 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 CD62L.
  • operably linked means that the signal peptide can direct expression or transmembrane transfer (localization) of the fusion element.
  • the fusion protein has a structure selected from the group consisting of:
  • A is an IL-12 protein element
  • B is a CD62L protein element
  • C is an optional linker element
  • D is an optional signal peptide signal peptide and/or leader peptide sequence
  • 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 (G 6 S) (SEQ ID NO.:8)
  • sequence of the IL12 protein element is set forth in SEQ ID NO.: 4.
  • the CD62L protein element has a cleavage site for K283-S284.
  • the CD62L protein is derived from a human or non-human mammal.
  • the CD62L protein comprises wild type and mutant form.
  • the CD62L protein comprises a full length, mature form of CD62L, or an active fragment thereof.
  • sequence of the 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 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.: 7).
  • the fusion protein is in single-stranded IL-12 (first protein element) and CD62L (second protein)
  • a linker peptide is provided between the elements), preferably a peptide 218 (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 cleaved by the ADAM17 protein to release 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 is provided, the polynucleotide encoding the fusion protein of the first aspect.
  • 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 vector of the third aspect of the invention or the polynucleotide of the second aspect integrated in the genome.
  • 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,
  • a method of producing the protein of the first aspect comprising the steps of:
  • composition comprising:
  • the fusion protein of the first aspect and
  • a pharmaceutically acceptable carrier is selected from:
  • an immune cell carrying or present on the cell surface thereof) the fusion protein of the first aspect.
  • the immune cells of at least 10 3 (preferably 103 to 109, more preferably preferably 104-108 th) the cell population of immune cells.
  • 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.
  • the use of the fusion protein of the first aspect and/or the immune cell of the seventh aspect, for the preparation of a medicament for treating a tumor is provided.
  • 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 the fusion protein of the first aspect and/or the immune cell of the seventh aspect to a subject in need thereof.
  • the fusion protein is administered as a monomer and/or a dimer.
  • the object is a human.
  • Figure 1 shows membrane surface cleavage and release induced by antigen-specific activation by CD62L/L-selectin.
  • FIG. 1A and FIG. 1B show schematic diagrams of the molecular mechanism of membrane molecule CD62L cleavage and release.
  • CD62L on the surface of T cells undergoes directed aggregation of microfilaments (actin, etc.) under the action of specific activators such as antigens, resulting in specific cleavage membrane molecules of the cleavage enzyme (Adam17, etc.) on the surface of the cell membrane.
  • specific activators such as antigens
  • FIG. 1C shows tumor antigen-induced membrane molecule-specific cleavage and release.
  • Tumor cell 526 is a related antigen MART-1 expressing MHC I A2 molecule, which is a positive target cell; 938 tumor cell does not express MHC A2 molecule, and is a negative control cell.
  • PBMC was activated by anti-CD3/CD28 magnetic beads for 24 hours, T cells were transduced with lentiviral engineering supernatant expressing anti-tumor MART-1 TCR; 14 days later, T cells were associated with MART-1 antigen-expressing tumor cells 938 and 526 Co-culture. As shown in the figure, co-cultured T cells were tested for molecular CD45RO and CD62L on the surface of the membrane by flow cytometry.
  • the T cells of each group were sorted into CD8 cell subsets (column 1 and column 3) by FlowJo software analysis. And CD8/MART-1 cell subsets (column 2 and column 4).
  • the time points noted on the right indicate the time of collection and analysis of cells after co-culture, for a total of 4 time points: 1 h, 2 h, 4 h and 6 h.
  • the antibodies used in flow cytometry were CD62L FITC, CD45RO APC, MART-1PE and CD8 PercP.
  • Figure 2 shows that the cleavage and release of CD62L is dependent on tumor antigen stimulation.
  • PBMCs were transactivated by anti-CD3/CD28 magnetic beads for 24 hours, and T cells were transduced with lentiviral engineering supernatant expressing anti-tumor MART-1 TCR; 14 days later, T cells were associated with MART-1 antigen-expressing tumor cells 938 and 526 was co-cultured for 4 h.
  • the 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, CD8/MART- 1 cell subpopulation.
  • CD62L levels in free CD62L and co-cultured cells in the supernatant were detected by ELISA kit.
  • the amount of CD62L in the supernatant of the T+526 group was set to 100%, and the other groups calculated the percentage of the CD62L content by comparison.
  • Figure 3 shows that the cleavage of CD62L is a prerequisite for CD107a expression.
  • T cells were transduced by anti-tumor MART-1 TCR, co-cultured and analyzed and organized by FACS (as described above).
  • FACS The antibodies used for the assay were as follows, CD107a FITC, MART-1PE, CD8 PercP5.5, CD45RO PE Cy-7, CD62L APC, CD3APC Cy-7.
  • CD107a in each T cell subset is indicated by the arrow.
  • the lower panel EM represents the effector memory cells of CD62L-CD45RO+; CM represents the central memory cells of CD62L+CD45RO+.
  • Figure 4 shows the specific cleavage and release of IL-12 in the fusion protein CD62L/IL-12 induced by tumor antigen.
  • CD62L after CD62L is activated by tumor antigen, CD62L undergoes tumor antigen-induced cleavage and release.
  • the mechanism diagram shows how IL-12 in CD62L/IL-12 fusion protein is cleaved and released, ie, fusion protein IL-12 occurs. Partial release. According to this effector mechanism, the release of biologically active IL-12 can increase the recognition of tumor cells by anti-tumor T cells, thereby maximizing the anti-tumor effect.
  • TCR, CD107a, Perforin, Granzyme, CD62L and IL-12 are shown in the figure.
  • Figure 5 shows the construction of a lentiviral vector expressing a CD62L/IL-12 fusion protein and its expression in human T cells.
  • Figure 6 shows that co-culture of lentiviral vector hscIL-12/CD62L transduced T cells with tumors enhances IFNy expression and tumor antigen-dependent release of IL-12.
  • Figure 7 shows that lentiviral vector hscIL-12/CD62L-transduced murine T cell-mediated cell transfusion therapy significantly prolongs survival in tumor-bearing mice.
  • Figure 8 shows that the lentiviral vector hscIL-12/CD62L transduced T cells can effectively avoid the toxic side effects of in vitro T cell expansion caused by persistent secretion of IL-12.
  • the fusion protein of the invention is an IL-12/CD62L fusion protein.
  • IL-12/CD62L fusion protein when expressed in T cells, it is effectively displayed on the surface of T cells.
  • the IL-12/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 can be tumor antigen-dependent when approaching tumor cells.
  • the induced specific cleavage of CD62L is activated to release IL-12 at a site near the tumor cells, ie, release of IL-12 on the surface of the cell membrane that activates the tumor antigen.
  • IL-12 is activated to release IL-12 at a site near the tumor cells, ie, release of IL-12 on the surface of the cell membrane that activates the tumor antigen.
  • maximization of anti-tumor immunity effect can be achieved very effectively, and IL- is extremely significantly reduced. 12 toxic side effects.
  • the present invention has been completed on this basis.
  • the present inventors adopted a protocol for connecting IL-12 by expressing a cell membrane, and in view of the specific release of tumor antigen reactivity of IL-12, the present invention innovatively employs IL-12 and CD62L.
  • IL-12 and CD62L A strategy of fusion that regulates the specific release of tumor immunity by IL-12 by cleavage of CD62L tumor antigen reactivity.
  • IL-12/CD62L lentiviral gene-modified T cells are used to enhance the anti-tumor effect, and the tumor antigen-dependent cleavage and release of CD62L is demonstrated by in vitro experiments, further confirming lentiviral-mediated fusion.
  • the protein IL-12/CD62L can not only effectively modify the anti-tumor T cell expression fusion protein, but also can release the IL-12 on the membrane surface by CD62L cleavage.
  • T cell/tumor cell co-culture experiments demonstrated that IL-12/CD62L lentiviral gene-modified anti-tumor T cells can significantly increase the intensity of the immune response, which is shown to significantly enhance the secretion level of IFN ⁇ .
  • 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.
  • 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.
  • the peptide linker should be of sufficient length and flexibility to ensure that the two proteins attached have sufficient 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): human sources Signal peptide of the 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.
  • bifunctional fusion protein in the present invention, "recombinant bifunctional fusion protein", "protein of the present invention”, “fusion protein of the present invention”, “bifunctional fusion protein”, “IL-12-CD62L fusion protein”, “IL-12/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 an IL-12 protein element, CD62L, and a linker element.
  • a representative example is IL12-CD62L.
  • 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.
  • the DNA can be a coding strand or a non-coding strand.
  • 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 (such as IL12 or CD62L) or a fragment thereof can usually be amplified by PCR. Obtained by law, recombinant method or 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 such methods include, but are not limited to, conventional renaturation treatment, treatment with a protein precipitant (salting method), centrifugation, osmotic sterilizing, super treatment, ultracentrifugation, molecular sieve chromatography (gel filtration), adsorption layer Analysis, ion exchange chromatography, high performance liquid chromatography (HPLC) and various other liquid chromatography techniques and combinations of these methods.
  • conventional renaturation treatment treatment with a protein precipitant (salting method), centrifugation, osmotic sterilizing, super treatment, ultracentrifugation, molecular sieve chromatography (gel filtration), adsorption layer Analysis, ion exchange chromatography, high performance liquid chromatography (HPLC) and
  • 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 human T cells.
  • said T cell surface carries a MART-1 TCR.
  • a lentiviral expression system gene-modified T cell which expresses an anti-tumor TCR (T-cell receptor) and simultaneously expresses CD62L, hscIL-12 (human single-chain IL-12) And hscIL-12/CD62L fusion protein.
  • 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 may be formulated in a non-toxic, inert, and pharmaceutically acceptable aqueous carrier medium wherein the pH is usually from about 5 to about 8, preferably 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, 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 is not significantly toxic to T cells
  • the fusion protein of the present invention effectively inhibits the immune microenvironment of T cell-tumor tissue by locally and fixedly and periodically releasing IL-12 when T cells attack tumors, thereby inhibiting tumors very effectively and significantly reducing toxic side effect.
  • the primers and DNA sequences used were all synthesized by Invitrogen.
  • FIG. 5 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.
  • 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 and hscIL-12/CD62L and CD107a on the membrane surface were detected by flow cytometry.
  • the recombinant lentiviral vector carrying the human TCR ⁇ and ⁇ chain genes which specifically recognize the human melanoma-associated antigen MART-1 was transfected into autologous peripheral blood lymphocytes by molecular biological techniques, and the recombinant TCR was expressed in T lymphocytes.
  • 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.
  • CD62L lentiviral expression vector Construction of the CD62L lentiviral expression vector was based on published literature (Yang, Cohen et al. 2008, Yang, Liu et al. 2011), and the construction of hscIL-12 was referenced to published literature (Zhang, Kerkar et al. 2011).
  • the fusion protein of hscIL-12/CD62L is linked by the 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 the LVV-MSCV-MART-1 TCR, CD62L, hscIL-12L, and hscIL-12/CD62L fusion proteins were co-transfected into 293T cells together with three other packaging plasmids, using commercially available calcium phosphate as a vehicle. 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/CD62L on the membrane surface was manipulated by live cell staining of a conventional flow cytometer. CD62L in the supernatant and 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, diluted in 5000 cells and loaded into 250- ⁇ l syringe (Hamilton, Reno, NV) in 5 ⁇ l of liquid, using a 25-gauge needle.
  • mice received whole body 5 Gy radiation. On day 2, mice received a 0.5-1 ⁇ 10 6 DC vaccine subcutaneously, or a 1 ⁇ 10 7 trans-transfusion of anti-MART-1 TCR and IL-12/CD62L lentiviral gene-modified T cells via tail vein IV.
  • Mouse T cells were obtained from mouse spleen cells and activated with 10 ug/ml concanavalin (Con A) in the presence of IL-2 (5 IU/ml); on day 2, the lentiviral vector was shown T cells were transduced, and 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 groups of DCs and T cells are illustrated in the right side of Figure 7. 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 were confirmed by 1% agarose electrophoresis and sequencing.
  • the structure of the obtained 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.
  • mixture The composition is as follows, take 2ml Optimum I and add pLenti-MSCV (22.5ug), pMD2.G (7.5ug), gag/pol (15ug), pRev (10ug), mix; take 2ml 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.
  • 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.
  • the obtained lentiviral expression vectors were designated as LV-CD62L, LV-hscIL-12/CD62L, 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 A suitable amount of lentivirus was added in a ratio of 3:1, centrifuged at 2000 xg for 2 h, and after 6 h, culture was continued by adding 100 IU/ml IL-2; the second transduction was performed on days 5 to 10, 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.
  • Anti-tumor TCR can directly produce anti-tumor T cells in vitro by modifying the PBMC of tumor patients with lentiviral genes. This kind of T cells does not require DC induction and has the specific effect of killing tumors.
  • T cells cultured for 2-3 weeks in vitro mimics the immune cells produced by the in vivo immune response, and can be divided into CD45RO+/CD62L2 (Tem, effector memory T cells) by flow cytometry.
  • CD45RO+/CD62L+ Tcm, central memory T cells
  • CD45ROlow/CD62L+ Tn, initial T cells
  • the local release of the antigen-reactive tumor immune response of IL-12 can be achieved by genetically modifying anti-tumor T cells to express new IL-12/CD62L molecules and utilizing the characteristics of CD62L tumor antigen reactive cleavage and release. Enhance the local anti-tumor immune response.
  • the lentivirus used is a third generation lentiviral vector
  • the promoter is MSCV
  • the expression gene component can be replaced by a gene to be expressed.
  • the 5' and 3' LTR (long terminal repeat) of this vector was transformed into SIN-LTR (self-inactivating-LTR), aiming at reducing the probability of lentiviral recombination, enhancing safety performance, and combining WPRE to terminate RNA transcription. It is currently widely used in clinical carrier structures.
  • CD62L and hscIL-12 are linked by a linker peptide.
  • T cells were transduced by two transductions, namely MART-1 TCR (first day) and sequential transduction of the three vectors (Day 10). On day 14, the surface of CD cells was detected by flow cytometry. Expression and expression of hscIL-12 on the membrane surface.
  • the result is shown in Figure 5.
  • the CD62L/IL-12 fusion protein was expressed by the third generation lentiviral vector, and the selected promoter was MSCV, which has been widely used in tumor immunocytotherapy of human body, and has the stability and safety of expression, and the expression of gene components can be
  • the gene is replaced by a gene to be expressed, namely, CD62L, MART-1 TCR, and IL-12/CD62L fusion protein.
  • the 5' and 3' LTR (long terminal repeat) of the vector were modified into SIN-LTR (self-inactivating-LTR), The aim is to reduce the probability of lentiviral recombination, enhance safety performance, and combine WPRE to terminate RNA transcription. It is the latest generation of genetic engineering vector structure widely used in clinical practice.
  • hscIL-12 human single chain IL-12
  • CD62L/hscIL-12 hscIL-12 fused IL-12 secreting peptide (lead seq) and linked to lentivirus by gene cloning
  • CD62L and hscIL-12 are linked by the peptide SGSG.
  • T cells were transduced by two transductions, namely MART-1 TCR (first day) and sequential transduction of the three vectors (Day 10). On day 14, the surface of CD cells was detected by flow cytometry. Expression and expression of hscIL-12 on the membrane surface.
  • CD62L can be expressed in a large amount on the surface of anti-tumor T cells.
  • IL-12 on the surface of membrane cells can be detected only in the fusion protein group expressing IL-12/CD62L, and the surface of secreted IL-12 membrane is not expressed.
  • the mechanism shown in Figure 4 is fully verified.
  • the mechanism of the invention is as follows: IL-12 cleaves and releases from the membrane surface by CD62L, and indicates that this release is dependent on the tumor immune response. According to this effect mechanism, the release of biologically active IL-12 can increase the anti-tumor response of anti-tumor T cells, improve the microenvironment of local tumor immunity, and achieve an optimized anti-tumor immune response.
  • T cells were sequentially transduced with anti-tumor TCR and CD62L/hscIL-12 fusion protein for 20 days, and co-cultured with tumor cells 526 and 938. After 24 hours, the expression levels of IFN ⁇ and IL-12 in the supernatant were detected by ELISA kit.
  • Co-culture of lentiviral vector hscIL-12/CD62L transduced T cells with tumors can enhance the expression level of IFN ⁇ and tumor antigen-dependent release of IL-12.
  • T cells were sequentially transduced with anti-tumor TCR and CD62L/hscIL-12 fusion protein for 20 days, and co-cultured with tumor cells 526 and 938. After 24 h, the expression levels of IFN ⁇ and IL-12 in the supernatant were detected by ELISA kit.
  • anti-tumor T cells modified by IL-12 soluble gene like the IL-12/CD62L gene modified on the membrane surface, can significantly increase the secretion level of IFN ⁇ compared with the anti-tumor T cell group.
  • the present invention further confirmed that only the cleavage and release of IL-12 in the IL-12/CD62L group is associated with activation of tumor antigens, and the sustained secretion of soluble IL-12 is not regulated by tumor antigens and is maintained at a high level. At the same time, the present invention also observes that the level of IL-12 cleavage and release on the surface of the membrane is lower than that of the continuously secreted IL-12, and we predict that the invention has high safety and clinical operability.
  • mice Female pmel mice (7 mice per group) were implanted intracranial (IC) 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 with 10 ug/ml concanavalin (Con A) in the presence of IL-2 (5 IU/ml); on day 2, the lentiviral vector was shown T cells were transduced, and 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 groups of DC and T cells are illustrated in the right side of Figure 7. Asterisks indicate that the experimental group was compared with the other groups, p ⁇ 0.001.
  • the present inventors have shown that by genetically modifying anti-tumor T cells to express new IL-12/CD62L molecules, and utilizing the characteristics of CD62L tumor antigen reactive cleavage and release, it is possible to achieve a partial immunoreactive tumor immune response of IL-12. Release, enhances the local anti-tumor immune response, and significantly reduces the toxic side effects of IL-12.
  • IL-12/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.
  • 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 an important role in the treatment of tumor immune cells. effect.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Virology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Cell Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne une protéine de fusion IL-12/CD62L ainsi que son procédé de préparation et son utilisation. En particulier, la libération de l'IL-12 sur la surface d'une membrane cellulaire activée par l'antigène tumoral est obtenue par une modification des lymphocytes T antitumoraux avec un lentivirus contenant un gène de fusion IL-12/CD62L, ainsi qu'un clivage spécifique et une libération induite par une activation dépendante de l'antigène tumoral CD62L.
PCT/CN2016/080420 2015-10-26 2016-04-27 Agent thérapeutique antitumoral modifié par protéine de fusion il-12/cd62l, son procédé de préparation et son utilisation WO2017071173A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201510702467.5A CN105218682B (zh) 2015-10-26 2015-10-26 经il-12/cd62l融合蛋白改造的肿瘤治疗剂及其制法和用途
CN201510702467.5 2015-10-26

Publications (1)

Publication Number Publication Date
WO2017071173A1 true WO2017071173A1 (fr) 2017-05-04

Family

ID=54988006

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/080420 WO2017071173A1 (fr) 2015-10-26 2016-04-27 Agent thérapeutique antitumoral modifié par protéine de fusion il-12/cd62l, son procédé de préparation et son utilisation

Country Status (2)

Country Link
CN (1) CN105218682B (fr)
WO (1) WO2017071173A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109306340A (zh) * 2017-07-27 2019-02-05 上海细胞治疗研究院 一种高效扩增全t细胞的人工抗原递呈细胞及其用途
CN109337872A (zh) * 2017-07-27 2019-02-15 上海细胞治疗研究院 高效扩增car-t细胞的人工抗原递呈细胞及其用途
CN110396133A (zh) * 2018-04-25 2019-11-01 中国科学院生物物理研究所 一种以白介素12为活性成分的融合蛋白型药物前体

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105218682B (zh) * 2015-10-26 2019-05-07 杨晶 经il-12/cd62l融合蛋白改造的肿瘤治疗剂及其制法和用途
CN105585637B (zh) * 2015-12-23 2020-04-03 杨晶 基于il-12稳定膜表达的肿瘤治疗剂及其制法和用途

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1318644A (zh) * 2000-04-18 2001-10-24 西安医科大学 一种重组人单链白细胞介素-12的构建方法
CN1432064A (zh) * 2000-03-23 2003-07-23 格林维尔医院系统公司 双功能癌症治疗剂
WO2003092737A1 (fr) * 2002-04-30 2003-11-13 Molmed Spa Fusions de cytokines et de proteines de ciblage tumoral
CN105218682A (zh) * 2015-10-26 2016-01-06 深圳精准医疗科技有限公司 经il-12/cd62l融合蛋白改造的肿瘤治疗剂及其制法和用途

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1432064A (zh) * 2000-03-23 2003-07-23 格林维尔医院系统公司 双功能癌症治疗剂
CN1318644A (zh) * 2000-04-18 2001-10-24 西安医科大学 一种重组人单链白细胞介素-12的构建方法
WO2003092737A1 (fr) * 2002-04-30 2003-11-13 Molmed Spa Fusions de cytokines et de proteines de ciblage tumoral
CN105218682A (zh) * 2015-10-26 2016-01-06 深圳精准医疗科技有限公司 经il-12/cd62l融合蛋白改造的肿瘤治疗剂及其制法和用途

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109306340A (zh) * 2017-07-27 2019-02-05 上海细胞治疗研究院 一种高效扩增全t细胞的人工抗原递呈细胞及其用途
CN109337872A (zh) * 2017-07-27 2019-02-15 上海细胞治疗研究院 高效扩增car-t细胞的人工抗原递呈细胞及其用途
CN109306340B (zh) * 2017-07-27 2023-06-06 上海细胞治疗研究院 一种高效扩增全t细胞的人工抗原递呈细胞及其用途
CN109337872B (zh) * 2017-07-27 2023-06-23 上海细胞治疗研究院 高效扩增car-t细胞的人工抗原递呈细胞及其用途
CN110396133A (zh) * 2018-04-25 2019-11-01 中国科学院生物物理研究所 一种以白介素12为活性成分的融合蛋白型药物前体
CN110396133B (zh) * 2018-04-25 2021-07-23 免疫靶向有限公司 一种以白介素12为活性成分的融合蛋白型药物前体

Also Published As

Publication number Publication date
CN105218682B (zh) 2019-05-07
CN105218682A (zh) 2016-01-06

Similar Documents

Publication Publication Date Title
CN108018299B (zh) 靶向bcma的嵌合抗原受体及其用途
US20210177902A1 (en) Ror-1 specific chimeric antigen receptors and uses thereof
CN109306016B (zh) 共表达细胞因子il-7的nkg2d-car-t细胞及其用途
CN108728459B (zh) 靶向cd19的嵌合抗原受体并联合表达il-15的方法和用途
CN108004259B (zh) 靶向b细胞成熟抗原的嵌合抗原受体及其用途
CN109320615B (zh) 靶向新型bcma的嵌合抗原受体及其用途
WO2017071173A1 (fr) Agent thérapeutique antitumoral modifié par protéine de fusion il-12/cd62l, son procédé de préparation et son utilisation
MX2014003176A (es) Celulas t diseñadas mediante arn para el tratamiento de cancer.
CN113913379B (zh) T淋巴细胞及其应用
US11976134B2 (en) MUC16 specific chimeric antigen receptors and uses thereof
CN108441505B (zh) 一种靶向ror1的嵌合抗原受体及其用途
CN113416260B (zh) 靶向Claudin18.2的特异性嵌合抗原受体细胞及其制备方法和应用
CN113896801B (zh) 靶向人Claudin18.2和NKG2DL的嵌合抗原受体细胞及其制备方法和应用
CN111378625A (zh) 一种cxcl13趋化型car-t细胞的制备和应用
CN110923255A (zh) 靶向bcma和cd19嵌合抗原受体及其用途
WO2018103734A1 (fr) Récepteur d'antigène chimère, son utilisation et son procédé de préparation
CN108707619B (zh) 靶向ror1的嵌合抗原受体及其用途
CN111978412B (zh) 武装靶向TGF-β的特异性嵌合抗原受体细胞及其制备方法和应用
CN111378046B (zh) 一种免疫效应细胞转换受体
WO2024055339A1 (fr) Procédé de préparation et d'amplification d'une cellule car-nk anti-cd19 humanisée universelle et son utilisation
CN110699371A (zh) 一种基于FcγRⅢa的嵌合基因及其用途
WO2017107353A1 (fr) Agent de traitement anticancéreux, son procédé de préparation et son utilisation employant il-12 avec une expression de membrane stable
CN111499766B (zh) 针对慢性淋巴细胞白血病的免疫效应细胞、其制备方法和应用
CN108165568B (zh) 一种培养CD19CAR-iNKT细胞方法及用途
CN110139875B (zh) Col14a1衍生的肿瘤抗原多肽及其应用

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: 16858621

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: 16858621

Country of ref document: EP

Kind code of ref document: A1