WO2021037160A1 - Tumor enzyme-responsive recombinant pyroptosis protein delivery system and antitumor use thereof - Google Patents

Tumor enzyme-responsive recombinant pyroptosis protein delivery system and antitumor use thereof Download PDF

Info

Publication number
WO2021037160A1
WO2021037160A1 PCT/CN2020/111821 CN2020111821W WO2021037160A1 WO 2021037160 A1 WO2021037160 A1 WO 2021037160A1 CN 2020111821 W CN2020111821 W CN 2020111821W WO 2021037160 A1 WO2021037160 A1 WO 2021037160A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
fusion protein
gsdma3
tumor
cells
Prior art date
Application number
PCT/CN2020/111821
Other languages
French (fr)
Chinese (zh)
Inventor
黄永焯
黄晶晶
陈应之
Original Assignee
中国科学院上海药物研究所
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 中国科学院上海药物研究所 filed Critical 中国科学院上海药物研究所
Publication of WO2021037160A1 publication Critical patent/WO2021037160A1/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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N1/00Microorganisms, e.g. protozoa; Compositions thereof; Processes of propagating, maintaining or preserving microorganisms or compositions thereof; Processes of preparing or isolating a composition containing a microorganism; Culture media therefor
    • C12N1/20Bacteria; Culture media therefor
    • C12N1/205Bacterial isolates
    • 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
    • C12N15/70Vectors or expression systems specially adapted for E. coli
    • 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
    • C07K2319/10Fusion polypeptide containing a localisation/targetting motif containing a tag for extracellular membrane crossing, e.g. TAT or VP22
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/21Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a His-tag
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/50Fusion polypeptide containing protease site
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12RINDEXING SCHEME ASSOCIATED WITH SUBCLASSES C12C - C12Q, RELATING TO MICROORGANISMS
    • C12R2001/00Microorganisms ; Processes using microorganisms
    • C12R2001/01Bacteria or Actinomycetales ; using bacteria or Actinomycetales
    • C12R2001/185Escherichia
    • C12R2001/19Escherichia coli

Definitions

  • the present invention belongs to the field of biomedicine. More specifically, the present invention relates to a tumor enzyme-responsive recombinant pyroptosis protein delivery system and its anti-tumor use.
  • Pyrodepressor protein is a family of proteins that can mediate pyrodesis of cells (gasdermin protein family), with 45% sequence homology among members, including GSDMA(1-3), GSDMB, GSDMC(1-4), GSDMD, DFNA5 (GSDME), DFNB59 (GSDMF).
  • DFNB59 Except for DFNB59, other pyro-apoptotic proteins have similar structures: two domains including N-domain and C-domain, and a linker that connects these two domains. Under normal circumstances, the two domains are tightly combined in a state of self-inhibition.
  • the active N domain binds to phosphatidylinositol (PI) or phosphatidylserine (PS) in the inner leaf of the cell membrane, or directly binds to the cardiolipin on the outer side of the bacterial plasma membrane, and accumulates in the cell membrane to form holes, resulting in cell swelling and cell membrane Rupture, release of contents, and cause an inflammatory response and ultimately lead to cell clearance. This process is called scorch of the cell.
  • PI phosphatidylinositol
  • PS phosphatidylserine
  • Pyrolysis of cells is defined as a new, pro-inflammatory, programmed cell death method. Studies have found that pyrolysis and pyrolysis proteins are related to a variety of diseases. A large number of intracellular factors are released during the process of pyrolysis, including high mobility protein (HMGB1), lactate dehydrogenase (LDH), calreticulin (CRT), IL-1 ⁇ , etc., so the process of pyrolysis can also be defined as secondary The process of sexual death.
  • HMGB1 high mobility protein
  • LDH lactate dehydrogenase
  • CRT calreticulin
  • IL-1 ⁇ etc.
  • DCs dendritic cells
  • cytotoxic T cells cytotoxic T cells
  • T lymphocyte T lymphocyte (CTL) specifically kills tumors and reduces the level of intracellular ATP.
  • the purpose of the present invention is to provide a technical means capable of enabling pyrooptin to be activated in the tumor microenvironment with high specificity and to enter tumor cells efficiently.
  • a fusion protein is provided, and the fusion protein has the structure shown in formula I from the N-terminus to the C-terminus:
  • Z0 is an optional label element
  • Z1 is the N domain element of pyrolysis protein
  • Z2 is a penetrating peptide sequence element
  • Z3 is a peptide sequence element that can be specifically cleaved by a protease specifically expressed in the tumor microenvironment;
  • Z4 is the C domain element of pyroopterin
  • the Z4 element inhibits the activity of the Z1 element by specifically binding to the Z1 element.
  • the tag is selected from the group consisting of His tag, GST tag, HA tag, c-Myc tag, Flag tag, or a combination thereof.
  • the pyroapone protein has the function of inducing the rupture of the cell membrane and releasing a large amount of content to cause the body's inflammatory response.
  • the pyroptosis protein is selected from the group consisting of human pyroptosis protein or murine pyroptosis protein.
  • the human pyroptosis protein is selected from the group consisting of GSDMA, GSDMB, GSDMC, GSDMD, DFNA5 (GSDME), DFNB59 (GSDMF), or a combination thereof.
  • the murine pyroptosis protein is selected from the following group: GSDMA1, GSDMA2, GSDMA3, GSDMC1, GSDMC2, GSDMC3, GSDMC4, or a combination thereof.
  • the pyroptin protein is GSDMA3.
  • the N domain is an active protein domain that forms a hole in the cell membrane.
  • amino acid sequence of the N domain is selected from the following group:
  • amino acid residues On the basis of SEQ ID NO:1, one or more amino acid residues are replaced, deleted, changed or inserted, or 1 to 30 amino acid residues are added to the N-terminus or C-terminus, preferably 1 to 10 amino acid residues, more preferably 1 to 5 amino acid residues, to obtain an amino acid sequence.
  • the penetrating peptide has the function of carrying different components across the cell membrane.
  • the penetrating peptide is selected from the group consisting of cationic cell penetrating peptides (such as TAT), hydrophobic cell penetrating peptides, and amphiphilic cell penetrating peptides.
  • the penetrating peptide is TAT.
  • the amino acid sequence of the penetrating peptide is selected from the following group:
  • the protease specifically expressed in the tumor microenvironment is selected from the following group: asparagine endopeptidase (Legumain), matrix metalloprotease, or a combination thereof.
  • the matrix metalloproteinase is selected from the group consisting of MMP-2, MMP-7, MMP-9, MMP-12, or a combination thereof.
  • the protease specifically expressed in the tumor microenvironment is asparagine endopeptidase.
  • the peptide sequence is selected from: PTN, a substrate peptide sequence specifically recognized and cleaved by Legumain.
  • the amino acid sequence of the peptide sequence is selected from the following group:
  • amino acid sequence of the N domain is selected from the following group:
  • amino acid residues On the basis of SEQ ID NO: 2, one or more amino acid residues are replaced, deleted, changed or inserted, or 1 to 30 amino acid residues are added to its N-terminal or C-terminal, preferably 1 to 10 amino acid residues, more preferably 1 to 5 amino acid residues, to obtain an amino acid sequence.
  • amino acid sequence of the fusion protein is shown in SEQ ID NO: 4.
  • an isolated polynucleotide which encodes the fusion protein according to the first aspect of the present invention.
  • sequence of the polynucleotide is shown in SEQ ID NO: 5.
  • a vector which contains the polynucleotide according to the second aspect of the present invention.
  • the vector is selected from the group consisting of pET vector, pMAL vector, and pGEX vector.
  • the vector is selected from the group consisting of pET28a, pMAL-2c, pGEX-4T-2, or a combination thereof.
  • a host cell in the fourth aspect of the present invention, contains the vector as described in the third aspect of the present invention, or the polynucleotide as described in the second aspect of the present invention is integrated into the genome.
  • the host cell is Escherichia coli.
  • the host cell is selected from the group consisting of BL21(DE3), Rosetta, Origami, or a combination thereof.
  • a method for producing the fusion protein according to the first aspect of the present invention which includes the steps:
  • the host cell according to claim 4 is cultured, thereby expressing the fusion protein according to the first aspect of the present invention.
  • composition comprising:
  • the content of the component (a) is 0.1-99.9% by weight, preferably 10-99.9% by weight, more preferably 70%-99.9% by weight.
  • the pharmaceutical composition is liquid, solid, or semi-solid.
  • the dosage form of the pharmaceutical composition is an oral dosage form, an injection, or a topical pharmaceutical dosage form.
  • the dosage form of the pharmaceutical composition includes tablets, granules, capsules, oral liquids, or injections.
  • the pharmaceutical composition is a liquid composition.
  • the pharmaceutically acceptable carrier is selected from the following group: infusion carrier and/or injection carrier, preferably, the carrier is one or more carriers selected from the following group : Normal saline, dextrose saline, or a combination thereof.
  • the pharmaceutical composition can be used alone or in combination with other anti-tumor drugs.
  • a fusion protein as described in the first aspect of the present invention a polynucleotide as described in the second aspect of the present invention, a vector as described in the third aspect of the present invention, and a fusion protein as described in the present invention.
  • the use of the host cell according to the fourth aspect of the invention is for preparing a preparation or pharmaceutical composition, and the preparation or pharmaceutical composition is used for one or more selected from the following group:
  • the immunogenic cell death (ICD) related characteristic molecules are selected from the following group: ATP, HMGB1, CRT, or a combination thereof.
  • the tumor is selected from the group consisting of breast cancer, colon cancer, prostate cancer, ovarian tumor, or a combination thereof.
  • the tumor cells are 4T1 cells or CT26 colon cancer cells.
  • the anti-cancer factor is selected from the group consisting of TNF- ⁇ , IL-1 ⁇ , IL-2, IFN- ⁇ , or a combination thereof.
  • the antigen presenting molecule is MHC class I molecule or MHC class II molecule.
  • the effector T cells are selected from the group consisting of CD8 + T cells, CD4 + T cells, CD8 + &GranzymeB + T cells, CD8 + &IFN- ⁇ + T cells, or a combination thereof.
  • the tumor proliferation and metastasis-related protein is MR or Legumain.
  • the cancer-promoting factor is TGF- ⁇ .
  • a method for treating tumors comprising the steps of: administering the fusion protein according to the first aspect of the present invention and the polynucleoside according to the second aspect of the present invention to a subject in need Acid, the vector according to the third aspect of the present invention, the host cell according to the fourth aspect of the present invention, or the pharmaceutical composition according to the sixth aspect of the present invention.
  • the subject includes humans or non-human mammals.
  • the non-human mammals include rodents (such as rats and mice) and primates (such as monkeys).
  • Figure 1 shows the FPLC desalting column and molecular sieve purification diagram of recombinant pyopterin.
  • (A-C) are the FPLC chromatograms of GSDMA3, GSDMA3-PTN and GSDMA3-TAT-PTN after passing through the desalting column; (D-E) are the FPLC chromatograms of GSDMA3, GSDMA3-PTN and GSDMA3-TAT-PTN after molecular sieve purification.
  • Figure 2 shows the characterization diagram of the recombinant protein.
  • lane M is Marker; lanes 1-3, lanes 4-6, and lanes 7-9 are the protein samples with three absorption peaks after GSDMA3-TAT-PTN, GSDMA3-PTN, and GSDMA3 are purified by Superdex 75.
  • Figure 3 shows the level of Legumain enzyme expression in cell lines cultured in vitro.
  • Figure 4 shows the in vitro Legumain cleavage experiment of the fusion pyrolysis protein.
  • lane M is Marker; lane 1 is GSDMA3-TAT-PTN after Legumain digestion; lanes 2 and 3 are GSDMA3-PTN after Legumain digestion; lane 4 is GSDMA3 after Legumain digestion.
  • FIG. 5 shows the results of the cell uptake experiment.
  • (A) shows the results of the uptake of pyro-depth protein detected by flow cytometry after digestion; (B) shows the results of statistical analysis of the uptake results.
  • Figure 6 shows the results of the toxicity experiment of pyrooptin.
  • (A) shows the killing effect of pyrodepressin on 4T1 cells; (B) shows the killing effect of pyrodepressin on DC2.4 cells.
  • Figure 7 shows the changes in cell morphology after recombinant protein treatment.
  • (A-D) are the results of bright field photography of cells in the PBS, GSDMA3, GSDMA3-PTN, and GSDMA3-TAT-PTN administration treatment groups in order.
  • Figure 8 shows the experimental results of CRT eversion caused by the pyrooptin-mediated immunogenic death of tumor cells.
  • (A-D) are the results of PBS, GSDMA3, digested GSDMA3-PTN, and digested GSDMA3-TAT-PTN in order of treating tumor cells; (E) shows the statistical analysis of CRT.
  • Figure 9 shows the released amount of HMGB1; (B) shows the amount of ATP released outside the cell.
  • Figure 10 shows the results of the in vitro sensitization experiment of DC.
  • (A) shows the amount of CD80 + DC cells after pyrolyst protein treatment
  • (B) shows the amount of CD86 + DC cells after pyrolyst protein treatment
  • (C) shows CD80 + /CD86 after pyrolyst protein treatment + The amount of double positive DC cells.
  • Figure 11 shows the effect of pyroptin on antigen presentation.
  • Figure 12 shows the results of the pharmacodynamic experiment of recombinant pyrooptosis protein on murine 4T1 breast cancer in situ tumors.
  • (A) is the tumor volume change graph;
  • (B) is the drug effect flow chart;
  • (C) is the mouse body weight change curve;
  • (D) shows the weight graph of the tumor at the experimental end point;
  • (E) shows the experimental end point Tumor inhibition rate of each treatment group at time;
  • (F) is the photo of the tumor at the end of the experiment (*P ⁇ 0.05, **P ⁇ 0.01, ***P ⁇ 0.001 and ****P ⁇ 0.0001).
  • Figure 13 shows a schematic diagram of T cells and their granzymes in the spleen.
  • (AC) shows the spleen of the PBS group as an example, and the schematic diagram of the flow gate;
  • (DG) shows the CD8 + T cells in the PBS group, GSDMA3 group, GSDMA3-PTN group and GSDMA3-TAT-PTN group in sequence. Changes in granzyme secretion.
  • Figure 14 shows the changes of T cells and their secreted factors in the spleen of each treatment group.
  • (A) shows the change in the amount of CD4 + T cells in the spleen; (B) shows the change in the amount of CD8 + T cells in the spleen; (C) shows the CD8 + &GranzymeB + T in the spleen of each group after treatment Cell changes; (D) shows the changes of CD8 + &IFN- ⁇ + T cells in the spleen of each group after treatment.
  • FIG. 15 shows the changes of T cells and their secreted factors in tumors after drug treatment.
  • (A) shows the change in the amount of CD4 + T cells in the tumor tissue; (B) shows the change in the amount of CD8 + T cells in the tumor tissue; (C) shows the CD8 + in each group of tumors after treatment The changes of &Granzyme + T cells; (D) shows the changes of CD8 + &IFN- ⁇ + T cells in the tumor tissues of each group after treatment.
  • Figure 16 shows the changes of T cells and their secreted factors in tumors after drug treatment.
  • (A) shows the change in the amount of CD4 + T cells in the lymph nodes; (B) shows the change in the amount of CD8 + T cells in the lymph nodes; (C) shows the CD8 + &Granzyme + T in each group of lymph nodes after treatment Cell changes; (D) shows the changes of CD8 + &IFN- ⁇ + T cells in the lymph nodes of each group after treatment.
  • Figure 17 shows the changes of macrophages and NK cells in tumors after administration of treatment.
  • (A-B) shows the changes of M1 type macrophages
  • (C-D) shows the changes of M2 type macrophages
  • (E-F) shows the changes of NK cells.
  • (A, D, E) are the tumor cells in the GSDMA3-TAT-PTN treatment group as an example.
  • Figure 18 shows the changes in the expression of related proteins in tumor tissues in different treatment groups.
  • Figure 19 shows the changes of cytokines in tumor tissues.
  • A-B indicate the changes of IL-2 and TGF- ⁇ in the tumor tissue at the end of the experiment in turn.
  • Figure 20 shows the changes in the weight of the main organs.
  • Figure 21 shows a pathological section of the main organs.
  • the scale is 100 ⁇ m.
  • Figure 22 shows the test results of liver function and kidney function of mice at the end of the experiment.
  • (A-C) respectively represent the changes of alanine aminotransferase, aspartate aminotransferase and total bilirubin content related to liver function; (D-F) represent the changes of serum urea, serum creatinine and serum uric acid content related to renal function, respectively.
  • the present inventors took the cell pyrolysis and the pyrolysis protein Gasdermin A3 (GSDMA3) as the research object, and used genetic engineering technology to modify its structure, and introduced the Legumain substrate peptide sequence PTN between the two structural domains of the pyrolysis protein.
  • the arginine-rich cationic penetrating peptide TAT RKKRRQRRR was selected and constructed in the adjacent position of the PTN sequence and close to the N-terminal side of the pyrolysis protein (N-GSDMA3-TAT-PTN-GSDMA3-C, or Marked as GSDMA3-TAT-PTN).
  • the experimental results show that the GSDMA3-TAT-PTN protein of the present invention exhibits very good anti-tumor activity in in vivo and in vitro experiments.
  • the present invention has been completed on this basis.
  • fusion protein of the present invention refers to the fusion protein according to the first aspect of the present invention, which has the ability to induce cell membrane rupture and release A large number of inclusions cause the function of the body's inflammatory response.
  • the provided fusion protein has the structure shown in formula I from N-terminus to C-terminus:
  • Z0 is an optional tag element
  • Z1 is the N-domain element of pyrooptin
  • Z2 is a penetrating peptide sequence element
  • Z3 is a peptide sequence element that can be specifically cleaved by a protease specifically expressed in the tumor microenvironment
  • Z4 is The C domain element of pyrolysis protein
  • "-" represents the peptide bond connecting the above elements
  • the Z4 element inhibits the activity of the Z1 element by specifically binding to the Z1 element.
  • the pyrolyst protein can be selected from human pyrolyt protein (such as GSDMA, GSDMB, GSDMC, GSDMD, DFNA5 (GSDME), DFNB59 (GSDMF), etc.) or murine pyrolyt protein (such as GSDMA1, GSDMA2, GSDMA3, GSDMC1, GSDMC2, GSDMC3, GSDMC4, etc.).
  • human pyrolyt protein such as GSDMA, GSDMB, GSDMC, GSDMD, DFNA5 (GSDME), DFNB59 (GSDMF), etc.
  • murine pyrolyt protein such as GSDMA1, GSDMA2, GSDMA3, GSDMC1, GSDMC2, GSDMC3, GSDMC4, etc.
  • the pyroptin protein is GSDMA3.
  • the N domain is an active protein domain that forms holes in the cell membrane; in the Z2, the penetrating peptide has the function of carrying different components through the cell membrane .
  • the penetrating peptide is selected from the group consisting of cationic cell penetrating peptides (such as TAT), hydrophobic cell penetrating peptides, and amphiphilic cell penetrating peptides.
  • the penetrating peptide is TAT.
  • the protease specifically expressed in the tumor microenvironment is selected from the following group: asparagine endopeptidase (Legumain), matrix metalloproteinase (such as MMP-2, MMP- 7. MMP-9, MMP-12, etc.), or a combination thereof.
  • asparagine endopeptidase Legumain
  • matrix metalloproteinase such as MMP-2, MMP- 7. MMP-9, MMP-12, etc.
  • the protease specifically expressed in the tumor microenvironment is asparagine endopeptidase.
  • the peptide sequence is selected from: a substrate peptide sequence PTN specifically recognized and cleaved by Legumain.
  • genes provided in the examples of the present invention are of murine origin, they are derived from other similar species (especially mammals) and are compatible with the sequence of the present invention (preferably, the sequence is shown in SEQ ID NO: 5).
  • the gene sequence of recombinant pyroopterin protein with certain homology (conservation) is also included in the scope of the present invention, as long as those skilled in the art can easily obtain information from other sources after reading this application according to the information provided in this application.
  • the sequence is isolated from species (especially mammals).
  • the polynucleotide of the present invention may be in the form of DNA or RNA.
  • DNA forms include: DNA, genomic DNA or synthetic DNA.
  • DNA can be single-stranded or double-stranded.
  • DNA can be a coding strand or a non-coding strand.
  • the coding region sequence encoding the fusion protein may be the same as the coding region sequence shown in SEQ ID NO: 5 or a degenerate variant.
  • the polynucleotide encoding the fusion protein includes: only the coding sequence of the fusion protein; the coding sequence of the fusion protein and various additional coding sequences; the coding sequence (and optional additional coding sequence) of the fusion protein and non-coding sequences.
  • polynucleotide encoding a fusion protein may include a polynucleotide encoding the fusion protein, or a polynucleotide that also includes additional coding and/or non-coding sequences.
  • the present invention also relates to variants of the aforementioned polynucleotides, which encode fragments, analogs and derivatives of polyglycosides or polypeptides having the same amino acid sequence as the present invention.
  • the variants of this polynucleotide can 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. It may be a substitution, deletion or insertion of one or more nucleotides, but does not substantially change the fusion protein encoded by it.
  • the present invention also relates to polynucleotides that hybridize with the aforementioned sequences and have at least 50%, preferably at least 70%, and more preferably at least 80% identity between the two sequences.
  • the present invention particularly relates to polynucleotides that can hybridize with the polynucleotides of the present invention under stringent conditions.
  • stringent conditions refer to: (1) hybridization and elution at lower ionic strength and higher temperature, such as 0.2 ⁇ SSC, 0.1% SDS, 60°C; or (2) adding during hybridization There are denaturants, such as 50% (v/v) methylphthalamide, 0.1% calf serum/0.1% Ficoll, 42°C, etc.; or (3) only the identity between the two sequences is at least 90% or more, More preferably, the hybridization occurs when 95% or more occurs.
  • the full-length nucleotide sequence or fragments thereof encoding the fusion protein of the present invention can usually be obtained by PCR amplification method, recombination method or artificial synthesis method.
  • primers can be designed according to the relevant nucleotide sequence disclosed in the present invention, especially the open reading frame sequence, and a commercially available DNA library or a cDNA prepared by a conventional method known to those skilled in the art can be used.
  • the library is used as a template to amplify the relevant sequences. When the sequence is long, it is often necessary to perform two or more PCR amplifications, and then splice the amplified fragments together in the correct order.
  • the recombination method can be used to obtain the relevant sequence in large quantities. It is usually cloned into a vector, and then transferred into a cell, and then the relevant sequence is isolated from the proliferated host cell by conventional methods.
  • artificial synthesis methods can also be used to synthesize related sequences, especially when the fragment length is short. Usually, by first synthesizing multiple small fragments, and then ligating to obtain fragments with very long sequences. At present, the DNA sequence encoding the fusion protein (or fragment or derivative thereof) of the present invention can be obtained completely through chemical synthesis. The DNA sequence can then be introduced into various existing DNA molecules (or such as vectors) and cells known in the art. In addition, mutations can also be introduced into the fusion protein sequence of the present invention through chemical synthesis.
  • the present invention relates to a recombinant pyroptin fusion protein used in an anti-tumor drug delivery system.
  • the amino acid sequence of the fusion protein is shown in SEQ ID NO: 4.
  • the polypeptide of the present invention can effectively induce the rupture of the cell membrane and release a large amount of contents to cause inflammation in the body.
  • the present invention also includes 50% or more of the sequence shown in SEQ ID NO: 4 of the present invention (preferably 60% or more, 70% or more, 80% or more, more preferably 90% or more, more preferably 95% or more, most preferably 98 % Or more, such as 99%) homologous polypeptides or proteins with the same or similar functions.
  • the "same or similar function” mainly refers to: "effectively induce the rupture of the cell membrane and release a large amount of content to cause the body's inflammatory response".
  • the fusion protein of the present invention can be a recombinant polypeptide, a natural polypeptide, or a synthetic polypeptide.
  • the fusion protein of the present invention can be a natural purified product, or a chemically synthesized product, or produced from a prokaryotic or eukaryotic host (for example, bacteria, yeast, plant, insect, and mammalian cells) using recombinant technology.
  • a prokaryotic or eukaryotic host for example, bacteria, yeast, plant, insect, and mammalian cells
  • the fusion protein of the present invention may be glycosylated or non-glycosylated.
  • the fusion protein of the present invention may also include or not include the initial methionine residue.
  • the present invention also includes other polypeptide fragments and analogs having the activity of the fusion protein of the present invention.
  • fragment and “analog” refer to polypeptides that substantially retain the same biological function or activity as the fusion protein of the present invention.
  • polypeptide fragments, derivatives or analogues of the present invention may be: (i) polypeptides with one or more conservative or non-conservative amino acid residues (preferably conservative amino acid residues) substituted, and such substituted amino acid residues
  • the base may or may not be encoded by the genetic code; or (ii) a polypeptide with a substitution group in one or more amino acid residues; or (iii) the mature polypeptide and another compound (such as a compound that extends the half-life of the polypeptide, For example, polyethylene glycol) fused to form a polypeptide; or (iv) additional amino acid sequence is fused to the polypeptide sequence to form a polypeptide (such as a leader sequence or secretory sequence, or a sequence or proprotein sequence used to purify the polypeptide, or Fusion protein).
  • these fragments, derivatives and analogs belong to the scope well known to those skilled in the art.
  • the fusion protein variant is the amino acid sequence shown in SEQ ID NO: 4, after several (usually 1-10, preferably 1-8, more preferably 1-4 , Preferably 1-2) the derived sequence obtained by substituting, deleting or adding at least one amino acid, and adding one or several (usually within 10, preferably within 5) at the C-terminus and/or N-terminus , More preferably within 3) amino acids.
  • the protein when the protein is substituted with amino acids with similar or similar properties, it usually does not change the function of the protein. Adding one or several (such as 1-3) amino acids at the C-terminus and/or N-terminus is usually also Does not change the function of the protein.
  • the present invention also includes analogs of the claimed protein.
  • the difference between these analogs and the natural SEQ ID NO: 4 may be the difference in the amino acid sequence, the difference in the modified form that does not affect the sequence, or both.
  • Analogs of these proteins include natural or induced genetic variants. Induced variants can be obtained by various techniques, such as random mutagenesis by radiation or exposure to mutagens, site-directed mutagenesis or other known molecular biology techniques. Analogs also include analogs having residues different from natural L-amino acids (such as D-amino acids), and analogs having non-naturally occurring or synthetic amino acids (such as ⁇ , ⁇ -amino acids). It should be understood that the protein of the present invention is not limited to the representative proteins listed above.
  • Modified (usually not changing the primary structure) forms include: chemically derived forms of proteins in vivo or in vitro, such as acetate or carboxylation. Modifications also include glycosylation, such as those that undergo glycosylation modifications during protein synthesis and processing. This modification can be accomplished by exposing the protein to an enzyme that performs glycosylation (such as a mammalian glycosylase or deglycosylase). Modified forms also include sequences with phosphorylated amino acid residues (such as phosphotyrosine, phosphoserine, and phosphothreonine).
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier and an effective amount of the following active ingredients: the fusion protein or its coding gene as described in the first aspect of the present invention.
  • an effective amount or “effective dose” refers to an amount that can produce function or activity on humans and/or animals and can be accepted by humans and/or animals.
  • pharmaceutically acceptable ingredients are substances that are suitable for humans and/or mammals without excessive side effects (such as toxicity, irritation, and allergic reactions), that is, substances that have a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier refers to a carrier used for the administration of a therapeutic agent, and includes various excipients and diluents.
  • the pharmaceutical composition of the present invention contains a safe and effective amount of the active ingredient of the present invention and a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier include (but are not limited to): saline, buffer, glucose, water, glycerol, ethanol, and combinations thereof.
  • the pharmaceutical preparation should match the mode of administration. For example, it can be prepared by conventional methods with physiological saline or an aqueous solution containing glucose and other adjuvants.
  • the pharmaceutical composition should be manufactured under aseptic conditions.
  • the effective amount of the active ingredient of the present invention can vary with the mode of administration and the severity of the disease to be treated.
  • the selection of the preferred effective amount can be determined by a person of ordinary skill in the art according to various factors (for example, through clinical trials).
  • the factors include, but are not limited to: the pharmacokinetic parameters of the active ingredients such as bioavailability, metabolism, half-life, etc.; the severity of the disease to be treated by the patient, the patient's weight, the patient's immune status, and administration The way and so on. For example, due to the urgent requirement of treating the condition, several divided doses can be given every day, or the dose can be reduced proportionally.
  • the pharmaceutically acceptable carriers of the present invention include (but are not limited to): water, saline, liposomes, lipids, proteins, protein-antibody conjugates, peptides, cellulose, nanogels, or Its combination.
  • the choice of carrier should match the mode of administration, which are well known to those of ordinary skill in the art.
  • the present invention uses genetic engineering technology to transform the pyrooptosis protein, which can realize the multifunctional delivery of the pyroptosis protein.
  • the penetrating peptide sequence is introduced into the pyroptosis protein, which gives the pyroptosis protein the ability to enter the cell, thereby exerting the pyroptosis effect.
  • the present invention applies the pyroopterin to induce immunogenic cell death (ICD) and tumor immunotherapy.
  • the present invention uses tumor-related enzymes such as Legumain-PTN as a responsive design for activating pyroapone proteins, which has a wide range of applications, improves the selectivity of tumor killing effects, and reduces its toxic side effects.
  • tumor-related enzymes such as Legumain-PTN as a responsive design for activating pyroapone proteins, which has a wide range of applications, improves the selectivity of tumor killing effects, and reduces its toxic side effects.
  • the prokaryotic expression plasmid pET28a-GSDMA3 was used as a template for subcloning and construction. Part of the amino acid sequence was replaced with Legumain-specific digested substrate peptide sequence PTN, and incorporated into the cell-penetrating peptide sequence TAT. Constructed into pET28a-PTN-GSDMA3, pET28a-PTN-TAT-GSDMA3 plasmids. The recombinant plasmid is transformed in vitro and expressed in Escherichia coli with high efficiency. The protein’s His-tag is used for preliminary purification, and then further purified by molecular sieve, and finally the recombinant protein is obtained.
  • pET28a-GSDMA3 as a template for subcloning construction, construct pET28a-PTN-GSDMA3, pET28a-PTN-TAT-GSDMA3 plasmids.
  • the plasmid is transferred to competent cells for culture, and then the culture is expanded and the protein expression is induced, and the obtained protein is purified.
  • the His-tag that comes with the protein to purify the Ni binding column
  • Purifier 10 Purifies the preliminarily purified protein by molecular sieve (Superdex 75), and uses SDS-PAGE electrophoresis to detect the purity of the protein.
  • FITC dye to label the protein, and then use Legumain digestion medium prepared in vitro to digest the recombinant protein, and then perform the cell uptake experiment on the recombinant protein after digestion. Detect the killing ability of the recombinant protein after digestion on tumor cells and observe it with a microscope. Detection of changes in the amount of ICD-related molecules (including CRT and HMGB1, etc.) released during the process of cell pyrolysis caused by the recombinant protein. Observe the effect of recombinant protein on the sensitization and presentation of DC.
  • Construct 4T1 breast cancer subcutaneous tumor in situ model when the tumor volume reaches about 40mm 3 , use recombinant pyrooptosis protein for treatment, conduct pharmacodynamic research, and perform eyeball blood collection at the end of the experiment (for liver function and Renal function test), the mice were then euthanized, and the tumors and organs (heart, liver, spleen, lung, kidney) of the mice were dissected. They were weighed and photographed.
  • a part of the tumor was immersed in 4% paraformaldehyde and fixed for subsequent slice experiments and biosafety (pathological slices, etc.) testing experiments; the other part was used to study the spleen, lymph nodes, and tumor tissues in vivo
  • WB was used to detect the changes of legumain, MR, TGF- ⁇ , and TNF- ⁇ in tumor tissues after treatment, and ELISA kits were used to detect cytokines (TGF- ⁇ , TNF- ⁇ ) in tumor tissues of each group after treatment. The change situation.
  • the proteins that have been preliminarily purified by a nickel column are passed through a desalting column (HiTrap Desalting) ( Figure 1A-C) and a gel exclusion chromatography column (Superdex 75) ( Figure 1D-F) Purification.
  • the peak shapes and peak positions of the three proteins are basically the same, which preliminarily shows that the protein modified by genetic engineering in this experiment did not affect its structure and function.
  • Lanes 1-9 are the proteins of the three absorption peaks of GSDMA3-TAT-PTN, GSDMA3-PTN and GSDMA3, respectively.
  • the band of GSDMA3 conforms to the theoretical molecular weight of about 55kDa (lane 7); the band of GSDMA3-PTN (lane 4) basically maintains the same level as the band of GSDMA3, which shows that the molecular weight of the two is close to and theoretically consistent; the recombinant protein GSDMA3 -The TAT-PTN band (lane 1) has a significant upward shift; while the other lanes have mixed bands, which indicates that the protein purity of the first absorption peak is better, while the other two absorption peaks have a large amount of mixed protein , So all subsequent experiments use the protein with the first absorption peak.
  • Example 4 In vitro Legumain cleavage experiment of the fusion pyrolysis protein
  • Figure 4 shows the results of digestion of GSDMA3-TAT-PTN, GSDMA3-PTN and GSDMA3 in turn.
  • the substrate peptide PTN is cleaved by Legumain
  • the N-domain and C-domain of the recombinant pyrolysis protein are released.
  • the fragments produced by GSDMA3-PTN and GSDMA3-TAT-PTN proteins are similar in size, so the band positions are close, and the results show that the recombination
  • the protein can be cleaved by Legumain enzyme.
  • the FITC-labeled recombinant protein was cleaved and activated by Legumain enzyme in advance, and then the uptake experiment of 4T1 cells was performed.
  • the experimental results are shown in Figure 5, the digested GSDMA3-TAT-PTN has the best ingestion effect.
  • the average fluorescence intensity values of GSDMA3, digested GSDMA3-TAT-PTN and GSDMA3-PTN are about 51.8, 109 and 67.4 respectively.
  • the uptake efficiency of GSDMA3-TAT-PTN is 1.6 times that of GSDMA3-PTN and 2.1 that of GSDMA3. Times. This indicates that the active fragment produced by GSDMA3-TAT-PTN after being activated by Legumain enzyme cleavage has a higher cell entry efficiency under the action of the penetrating peptide TAT.
  • 4T1 and DC2.4 cells were used to detect the toxicity of pyroptin.
  • 4T1 cells with the increase of protein concentration in the experimental concentration range, the survival rate of the cells decreased.
  • the recombinant protein GSDMA3-TAT-PTN after restriction digestion was significantly better than the other two groups in killing 4T1 tumor cells ( Figure 6A) .
  • All of the pyrolyzed proteins had no killing effect on DC2.4 cells within the tested concentration range ( Figure 6B), and had good biocompatibility, so the pyrolyzed proteins would not affect the function of antigen presenting cells.
  • 4T1 cells were treated with three groups of pyroptin proteins for 48h, as shown in Figure 7: normal 4T1 cells would grow into a network in sheets (Figure 7A); after treatment with GSDMA3 and digested GSDMA3-PTN, the morphology of the cells changed. There are also a large number of cells that are absorbing water and swelling ( Figures 7B and 7C), which is consistent with their cell killing effect and the results of the uptake experiment. After the digested GSDMA3-TAT-PTN treats the cells, a large number of cells have died, and have been washed away during the washing process. In the remaining fixed cells, a large number of swollen cells can also be seen, and there are basically no complete cells. Exist ( Figure 7D).
  • Figure 8 shows the change in the amount of extracellular CRT.
  • the tumor cells in the control group will have a negative peak on the left and a positive peak on the right ( Figure 8A).
  • Figure 8A After 4T1 cells are treated with pyroptin, the positive peaks of all groups are significantly increased.
  • the negative peak of the cells treated with GSDMA3-TAT-PTN was significantly decreased, and the positive peak was significantly increased ( Figures 8B, 8C and 8D).
  • the average fluorescence intensities of GSDMA3-TAT-PTN, GSDMA3-PTN and GSDMA3 after digestion were 132, 87 and 80.7, respectively.
  • the intensities of GSDMA3-TAT-PTN were 1.5 times and 1.6 times that of the latter two, respectively.
  • the GSDMA3 and GSDMA3-PTN administration groups can increase the release of HMGB1, but there is no significant difference from the PBS group.
  • the GSDMA3-TAT-PTN administration group can greatly increase the release of HMGB1, and the release concentration of HMGB1 is 1.9 times and 2.4 times that of the GSDMA3 group and the GSDMA3-PTN group, respectively, with significant differences.
  • the extracellular ATP content of all experimental groups increased significantly.
  • the extracellular ATP content of the GSDMA3-TAT-PTN treatment group was significantly different from the other treatment groups, indicating that ATP was released from the inside of the cell to the outside of the cell, which is considered to be
  • the hole formed by the pyrolysis protein is released to the outside of the cell or the membrane channel activated by the pyrolysis is released to the outside of the cell.
  • the amount of CD80 + and CD86 + DC cells basically maintained an upward trend, but there was no significant difference in the amount of CD80 + DC cells between the groups (Figure 10A).
  • the number of CD86 + DC cells can reach the level of the positive control group ( Figure 10B), and the number of CD80 + /CD86 + double-positive DCs in the protein treatment group can also reach the level of the positive control group.
  • the DCs treated with GSDMA3-TAT-PTN The double positive amount of cells was the highest ( Figure 10C).
  • Example 11 The effect of recombinant pyroopterin on antigen presentation
  • Example 12 Pharmacodynamic Study of Recombinant Pyroopterin on Subcutaneous Xenograft Tumor Model
  • Figure 12B is a flowchart.
  • the tumor volume of each treatment group increased, but the pyrooptin treatment group could slow down the growth trend (Figure 12A), and the tumor inhibition rates of the three groups at the experimental end point could reach 21%, 34% and 62% respectively (Figure 12E).
  • Figure 12C The body weight of the mice did not change much during the protein treatment ( Figure 12C), indicating that there were no obvious side effects.
  • Figures 12D and 12F are after the tumor was dissected at the end of the experiment (day 31) and weighed and photographed. It was found that the tumors in the pyroopterin treatment group were reduced, and the tumors in the GSDMA3-TAT-PTN group were the smallest, which was similar to other groups. There is a significant difference than that.
  • Example 13 Changes of T cells and cytokines secreted by them in the body after treatment
  • Figure 13 (A-D) takes the mouse spleen cells in the PBS treatment group as an example, showing a schematic diagram of the entire flow cytometry gate.
  • Figures (13E-F) show the changes in the amount of granzyme secreted in the spleen of mice after GSDMA3, GSDMA3-PTN, and GSDMA3-TAT-PTN treatment. The results of the secretion of granzyme and its interferon in tumors and lymph nodes are also analyzed with reference to this section.
  • Figure 14 shows the statistical results of T cells, granzyme and interferon in the spleen. After treatment, the amount of CD4 + T cells in each group did not change significantly (Figure 14A and 14B).
  • the GSDMA3-TAT-PTN treatment group had the largest number of CD8 + T cells and secreted the most granzyme and interferon ( Figure 14B).
  • 14C and 14D which indicates that the treatment of tumor-bearing mice with GSDMA3-TAT-PTN in this experiment can effectively activate T cells in the spleen and release a large amount of cytokines to kill the tumor.
  • Figure 16 shows the statistical results of T cells, granzyme and interferon in the axillary lymph nodes around the tumor.
  • GSDMA3-TAT-PTN treatment group and the other groups as compared to CD4 + T no significant change, but CD4 + T cells and the PBS group as compared to significantly reduce GSDMA3 and GSDMA3-PTN treatment groups (FIG. 16A); and all Compared with the PBS group, CD8 + T cells in the protein treatment group increased significantly (Figure 16B).
  • the expression levels of granzyme B and interferon in CD8 + T cells of all pyrostatin treatment groups increased, and the expression levels of the GSDMA3-TAT-PTN treatment group increased significantly ( Figure 16C and 16D).
  • Example 14 Changes of macrophages and NK cells in tumors
  • FIGS 17A and 17B M1 type macrophages of all protein treatment groups increased, and M1 type macrophages of tumors in the GSDMA3-TAT-PTN group increased significantly.
  • all protein treatment groups down-regulated M2 type macrophages ( Figure 17C and 17D), and the M2 type macrophages of the GSDMA3-PTN and GSDMA3-TAT-PTN treatment groups decreased significantly.
  • Figure 17 (E-F) shows that NK cells in the tumor tissue of the GSDMA3-TAT-PTN treatment group increased significantly, while there was no significant change in the other groups.
  • Example 15 Changes of legumain, MR, TGF- ⁇ , and TNF- ⁇ in tumor tissues of each group
  • Example 16 Changes in cytokines in tumor tissues
  • the ELISA kit was used to check the secretion of cytokines in the tumor tissues of each group at the end of the experiment, as shown in Figure 19. It was found that the secretion of cancer cell cytokines (TGF- ⁇ ) decreased in each treatment group, combined with the result analysis in Figure 18: GSDMA3-TAT-PTN treatment group can enhance the body’s immunity and cell killing and reduce tumor metastasis, and has a good anti-tumor effect. effect.
  • mice bearing 4T1 breast cancer had obvious splenomegaly, while GSDMA3-
  • the spleen of the mice in the TAT-PTN treatment group was smaller than that of the PBS group at the end of treatment, and there was no significant difference in other organs in each group.
  • mice The main organs (heart, liver, spleen, lung, and kidney) of the mice at the end of the experiment were subjected to pathological analysis, as shown in Figure 21. It was found that the alveoli in the GSDMA3 treatment group were deformed, and the alveoli in the other groups were normal in shape without obvious cell shedding; while other organs in each group had no obvious lesions, indicating that the recombinant protein has good biological safety and no obvious damage to the organs.
  • AST in the GSDMA3-TAT-PTN treatment group was significantly lower than the other two groups, but there was no significant difference compared with the PBS group. Based on the analysis of the increased expression of AST in acute, chronic hepatitis and toxic hepatitis, GSDMA3-TAT-PTN treatment does not cause liver damage. There were no significant changes in other indicators in each group, which indicated that the use of pyroapone protein treatment did not cause liver and kidney damage and inflammation.
  • the pyrolysis protein does not have a toxic effect when it is not activated by cutting. Once it is activated by cutting, it will release the toxic end, leading to perforation of the cell membrane, release of the contents, and inflammatory reaction, thereby mediating cell death, that is, pyrolysis. It is a class of promising biological macromolecular prodrugs, but it is an intracellular protein that must overcome the cell membrane barrier to enter the cytoplasm to play a role. In addition, the role of pyrolysis protein lacks cell specificity and is likely to cause toxic side effects.
  • TME tumor microenvironment
  • the invention utilizes the specific and highly expressed protease Legumain enzyme in TME, and realizes the anti-tumor effect of the pyrostat protein mediated by tumor enzyme activation by introducing the Legumain substrate peptide sequence PTN between the two structural domains of the pyrostat protein.
  • GSDMA3 of the pyrodepressin family is selected for research, and its N-domain is the active domain, which can cause cell pyrolysis.
  • genetic engineering technology was used to insert PTN into the linker sequence between the C-domain and N-domain of GSDMA3, so that it can be cleaved in TME to release the active N-domain.
  • the efficiency of protein itself is not high, and the N-domain needs to enter the cell to play a role. Therefore, this study introduced a penetrating peptide sequence between the N-domain and PTN to promote the penetration of the cell through the penetrating peptide. After entering the cell, the N-domain forms a hole in the cell membrane, leading to cell death.
  • a large amount of intracellular ATP, HMGB1, LDH, IL-1 ⁇ , calreticulin, etc. are released outside the cell to activate immune cells and inhibit tumor proliferation.
  • the GSDMA3-TAT-PTN protein provided by the present invention exhibits very good anti-tumor activity in in vivo and in vitro experiments, which shows the effectiveness of the design of the pyrostat protein delivery strategy using tumor enzyme activation and fusion of penetrating peptides.

Landscapes

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

Abstract

Provided is a fusion protein, a tumor enzyme-responsive recombinant pyroptosis protein delivery system, and antitumor use thereof. The fusion protein has the structure shown in formula I from the N-terminus to the C-terminus: Z0-Z1-Z2-Z3-Z4 (formula I), Z0 being an optional tag element; Z1 being the N-domain element of the pyrolysis protein; Z2 being a cell-penetrating peptide sequence element; Z3 being a peptide sequence element that can be specifically cleaved by a protease specifically expressed in a tumor microenvironment; Z4 being the C-domain element of the pyrolysis protein; "-" representing a peptide bond linking the elements; in the fusion protein, element Z4 inhibiting the activity of element Z1 by specifically binding to element Z1.

Description

肿瘤酶响应型重组焦亡蛋白递药系统及其抗肿瘤用途Tumor enzyme-responsive recombinant pyrooptosis protein delivery system and its anti-tumor use 技术领域Technical field
本发明属于生物医药领域,更具体而言,本发明涉及一种肿瘤酶响应型重组焦亡蛋白递药系统及其抗肿瘤用途。The present invention belongs to the field of biomedicine. More specifically, the present invention relates to a tumor enzyme-responsive recombinant pyroptosis protein delivery system and its anti-tumor use.
背景技术Background technique
焦亡蛋白是一类能够介导细胞焦亡的蛋白家族(gasdermin protein family),成员之间具有45%的序列同源性,包括GSDMA(1-3)、GSDMB、GSDMC(1-4)、GSDMD、DFNA5(GSDME)、DFNB59(GSDMF)。Pyrodepressor protein is a family of proteins that can mediate pyrodesis of cells (gasdermin protein family), with 45% sequence homology among members, including GSDMA(1-3), GSDMB, GSDMC(1-4), GSDMD, DFNA5 (GSDME), DFNB59 (GSDMF).
除了DFNB59,其他的焦亡蛋白都有着相似的结构:包括N结构域(N-domain)和C结构域(C-domain)这两个结构域以及连接这两个结构域的接头(linker)。正常情况下,两个结构域紧密结合处于自抑制状态。Except for DFNB59, other pyro-apoptotic proteins have similar structures: two domains including N-domain and C-domain, and a linker that connects these two domains. Under normal circumstances, the two domains are tightly combined in a state of self-inhibition.
一旦linker被切割,则破坏C结构域对N结构域的抑制作用,释放出活性的N结构域。活性的N结构域和细胞膜内叶的磷脂酰肌醇(PI)或磷脂酰丝氨酸(PS)结合,或者直接和细菌质膜外侧的心磷脂结合,在细胞膜上聚集形成孔洞,导致细胞肿胀、细胞膜破裂、内含物释放,以及引起炎症反应并最终导致细胞清除。这个过程被称为细胞的焦亡。Once the linker is cleaved, the inhibitory effect of the C domain on the N domain is destroyed, and the active N domain is released. The active N domain binds to phosphatidylinositol (PI) or phosphatidylserine (PS) in the inner leaf of the cell membrane, or directly binds to the cardiolipin on the outer side of the bacterial plasma membrane, and accumulates in the cell membrane to form holes, resulting in cell swelling and cell membrane Rupture, release of contents, and cause an inflammatory response and ultimately lead to cell clearance. This process is called scorch of the cell.
细胞的焦亡被定义为一种新型的、促炎的细胞程序性死亡方式。研究发现,焦亡以及焦亡蛋白和多种疾病相关。焦亡过程中会释放大量胞内因子包括高迁移率蛋白(HMGB1)、乳酸脱氢酶(LDH)、钙网蛋白(CRT)、IL-1β等,因而焦亡的过程也可以定义成继发性死亡过程。Pyrolysis of cells is defined as a new, pro-inflammatory, programmed cell death method. Studies have found that pyrolysis and pyrolysis proteins are related to a variety of diseases. A large number of intracellular factors are released during the process of pyrolysis, including high mobility protein (HMGB1), lactate dehydrogenase (LDH), calreticulin (CRT), IL-1β, etc., so the process of pyrolysis can also be defined as secondary The process of sexual death.
肿瘤细胞的焦亡在引起炎症的同时,释放的免疫原性相关分子,能提高树突状细胞(dendritic cells,DC)对肿瘤的识别及其抗原提呈能力,DC能激活毒性T细胞(cytotoxic T lymphocyte,CTL)对肿瘤特异性的杀伤,并降低胞内ATP的水平。While the pyrolysis of tumor cells causes inflammation, the immunogenic related molecules released can improve the recognition of dendritic cells (DC) to tumors and their ability to present antigens. DCs can activate cytotoxic T cells (cytotoxic T cells). T lymphocyte (CTL) specifically kills tumors and reduces the level of intracellular ATP.
然而,目前在肿瘤细胞的焦亡研究中,仍然存在一些难以克服的难点。焦亡蛋白的N结构域必须要在细胞内才能发挥作用,而现有的技术中难以保证焦亡蛋白的入胞效率。此外,如何使有活性焦亡蛋白的高特异性地靶向肿瘤细胞,或如何使焦亡蛋白高特异性地在肿瘤微环境中被激活,也是本领域亟待解决的问题。However, there are still some insurmountable difficulties in the research of tumor cell pyrexia. The N-domain of pyrolysis protein must be in the cell to play a role, and it is difficult to ensure the efficiency of pyrolysis protein in the existing technology. In addition, how to target tumor cells with high specificity of the active pyroptosis protein, or how to enable the pyroptosis protein to be activated in the tumor microenvironment with high specificity, is also an urgent problem to be solved in this field.
因此,本领域迫切需要开发一种能够使焦亡蛋白高特异性地在肿瘤微环境中被激活,并且高效进入肿瘤细胞的技术手段。Therefore, there is an urgent need in the art to develop a technical means that can enable pyroopterin to be activated in the tumor microenvironment with high specificity and enter tumor cells efficiently.
发明内容Summary of the invention
本发明的目的就是提供一种能够使焦亡蛋白高特异性地在肿瘤微环境中被激活,并且高效进入肿瘤细胞的技术手段。The purpose of the present invention is to provide a technical means capable of enabling pyrooptin to be activated in the tumor microenvironment with high specificity and to enter tumor cells efficiently.
在本发明的第一方面,提供了一种融合蛋白,所述融合蛋白从N端到C端具有式I所示的结构:In the first aspect of the present invention, a fusion protein is provided, and the fusion protein has the structure shown in formula I from the N-terminus to the C-terminus:
Z0-Z1-Z2-Z3-Z4   (式I)Z0-Z1-Z2-Z3-Z4 (Formula I)
式中,Where
Z0为任选的标签元件;Z0 is an optional label element;
Z1为焦亡蛋白的N结构域元件;Z1 is the N domain element of pyrolysis protein;
Z2为穿膜肽序列元件;Z2 is a penetrating peptide sequence element;
Z3为能够被肿瘤微环境中特异性表达的蛋白酶特异性切割的肽序列元件;Z3 is a peptide sequence element that can be specifically cleaved by a protease specifically expressed in the tumor microenvironment;
Z4为焦亡蛋白的C结构域元件;Z4 is the C domain element of pyroopterin;
“-”表示连接上述元件的肽键;"-" indicates a peptide bond connecting the above-mentioned elements;
其中,在融合蛋白中,所述Z4元件通过特异性地结合Z1元件来抑制Z1元件的活性。Wherein, in the fusion protein, the Z4 element inhibits the activity of the Z1 element by specifically binding to the Z1 element.
在另一优选例中,所述Z0元件中,所述标签选自下组:His标签、GST标签、HA标签、c-Myc标签、Flag标签,或其组合。In another preferred embodiment, in the Z0 element, the tag is selected from the group consisting of His tag, GST tag, HA tag, c-Myc tag, Flag tag, or a combination thereof.
在另一优选例中,所述焦亡蛋白具有诱导细胞膜破裂并释放大量内含物引起机体炎症反应的功能。In another preferred embodiment, the pyroapone protein has the function of inducing the rupture of the cell membrane and releasing a large amount of content to cause the body's inflammatory response.
在另一优选例中,所述焦亡蛋白选自人源焦亡蛋白或鼠源焦亡蛋白。In another preferred embodiment, the pyroptosis protein is selected from the group consisting of human pyroptosis protein or murine pyroptosis protein.
在另一优选例中,所述人源焦亡蛋白选自下组:GSDMA、GSDMB、GSDMC、GSDMD、DFNA5(GSDME)、DFNB59(GSDMF),或其组合。In another preferred embodiment, the human pyroptosis protein is selected from the group consisting of GSDMA, GSDMB, GSDMC, GSDMD, DFNA5 (GSDME), DFNB59 (GSDMF), or a combination thereof.
在另一优选例中,所述鼠源焦亡蛋白选自下组:GSDMA1、GSDMA2、GSDMA3、GSDMC1、GSDMC2、GSDMC3、GSDMC4,或其组合。In another preferred embodiment, the murine pyroptosis protein is selected from the following group: GSDMA1, GSDMA2, GSDMA3, GSDMC1, GSDMC2, GSDMC3, GSDMC4, or a combination thereof.
在另一优选例中,所述焦亡蛋白为GSDMA3。In another preferred embodiment, the pyroptin protein is GSDMA3.
在另一优选例中,所述Z1中,所述N结构域为具有在细胞膜上形成孔洞的活性 蛋白结构域。In another preferred example, in the Z1, the N domain is an active protein domain that forms a hole in the cell membrane.
在另一优选例中,所述Z1中,所述N结构域的氨基酸序列选自下组:In another preferred example, in the Z1, the amino acid sequence of the N domain is selected from the following group:
(i)如SEQ ID NO:1所示的序列;(i) The sequence shown in SEQ ID NO:1;
(ii)在SEQ ID NO:1的基础上,进行一个或多个氨基酸残基的替换、缺失、改变或插入,或在其N端或C端添加1至30个氨基酸残基,较佳地1至10个氨基酸残基,更佳地1至5个氨基酸残基,从而获得的氨基酸序列。(ii) On the basis of SEQ ID NO:1, one or more amino acid residues are replaced, deleted, changed or inserted, or 1 to 30 amino acid residues are added to the N-terminus or C-terminus, preferably 1 to 10 amino acid residues, more preferably 1 to 5 amino acid residues, to obtain an amino acid sequence.
在另一优选例中,所述Z2中,所述穿膜肽具有携带不同成分穿过细胞膜的功能。In another preferred example, in the Z2, the penetrating peptide has the function of carrying different components across the cell membrane.
在另一优选例中,所述Z2中,所述穿膜肽选自下组:阳离子细胞穿膜肽(如TAT)、疏水性细胞穿膜肽、两亲性细胞穿膜肽。In another preferred embodiment, in the Z2, the penetrating peptide is selected from the group consisting of cationic cell penetrating peptides (such as TAT), hydrophobic cell penetrating peptides, and amphiphilic cell penetrating peptides.
在另一优选例中,所述Z2中,所述穿膜肽为TAT。In another preferred embodiment, in the Z2, the penetrating peptide is TAT.
在另一优选例中,所述Z2中,所述穿膜肽的氨基酸序列选自下组:In another preferred embodiment, in the Z2, the amino acid sequence of the penetrating peptide is selected from the following group:
(i)如SEQ ID NO:3所示的序列;(i) The sequence shown in SEQ ID NO: 3;
(ii)在SEQ ID NO:3的基础上,进行一个或多个氨基酸残基的替换、缺失、改变或插入,从而获得的氨基酸序列。(ii) On the basis of SEQ ID NO: 3, one or more amino acid residues are replaced, deleted, changed or inserted to obtain an amino acid sequence.
在另一优选例中,所述Z3中,所述肿瘤微环境中特异性表达的蛋白酶选自下组:天冬酰胺内肽酶(Legumain)、基质金属蛋白酶,或其组合。In another preferred example, in the Z3, the protease specifically expressed in the tumor microenvironment is selected from the following group: asparagine endopeptidase (Legumain), matrix metalloprotease, or a combination thereof.
在另一优选例中,所述基质金属蛋白酶选自下组:MMP-2、MMP-7、MMP-9、MMP-12,或其组合。In another preferred embodiment, the matrix metalloproteinase is selected from the group consisting of MMP-2, MMP-7, MMP-9, MMP-12, or a combination thereof.
在另一优选例中,所述Z3中,所述肿瘤微环境中特异性表达的蛋白酶为天冬酰胺内肽酶。In another preferred example, in the Z3, the protease specifically expressed in the tumor microenvironment is asparagine endopeptidase.
在另一优选例中,所述Z3中,所述肽序列选自:Legumain特异性识别并切割的底物肽序列PTN。In another preferred example, in the Z3, the peptide sequence is selected from: PTN, a substrate peptide sequence specifically recognized and cleaved by Legumain.
在另一优选例中,所述Z3中,所述肽序列的氨基酸序列选自下组:In another preferred example, in the Z3, the amino acid sequence of the peptide sequence is selected from the following group:
(i)序列PTN;(i) Sequence PTN;
(ii)在序列PTN的基础上,进行N端或C端一个或多个氨基酸残基的插入,从而获得的氨基酸序列。(ii) The amino acid sequence obtained by inserting one or more amino acid residues at the N-terminal or C-terminal on the basis of the sequence PTN.
在另一优选例中,所述Z4中,所述N结构域的氨基酸序列选自下组:In another preferred example, in the Z4, the amino acid sequence of the N domain is selected from the following group:
(i)如SEQ ID NO:2所示的序列;(i) The sequence shown in SEQ ID NO: 2;
(ii)在SEQ ID NO:2的基础上,进行一个或多个氨基酸残基的替换、缺失、改变或插入,或在其N端或C端添加1至30个氨基酸残基,较佳地1至10个氨基酸残基,更佳地1至5个氨基酸残基,从而获得的氨基酸序列。(ii) On the basis of SEQ ID NO: 2, one or more amino acid residues are replaced, deleted, changed or inserted, or 1 to 30 amino acid residues are added to its N-terminal or C-terminal, preferably 1 to 10 amino acid residues, more preferably 1 to 5 amino acid residues, to obtain an amino acid sequence.
在另一优选例中,所述融合蛋白的氨基酸序列如SEQ ID NO:4所示。In another preferred embodiment, the amino acid sequence of the fusion protein is shown in SEQ ID NO: 4.
在本发明的第二方面,提供了一种分离的多核苷酸,所述多核苷酸编码如本发明第一方面所述的融合蛋白。In the second aspect of the present invention, an isolated polynucleotide is provided, which encodes the fusion protein according to the first aspect of the present invention.
在另一优选例中,所述多核苷酸的序列如SEQ ID NO:5所示。In another preferred embodiment, the sequence of the polynucleotide is shown in SEQ ID NO: 5.
在本发明的第三方面,提供了一种载体,所述载体中含有如本发明第二方面所述的多核苷酸。In the third aspect of the present invention, a vector is provided, which contains the polynucleotide according to the second aspect of the present invention.
在另一优选例中,所述载体选自下组:pET载体、pMAL载体、pGEX载体。In another preferred embodiment, the vector is selected from the group consisting of pET vector, pMAL vector, and pGEX vector.
在另一优选例中,所述载体选自下组:pET28a、pMAL-2c、pGEX-4T-2,或其组合。In another preferred embodiment, the vector is selected from the group consisting of pET28a, pMAL-2c, pGEX-4T-2, or a combination thereof.
在本发明的第四方面,提供了一种宿主细胞,所述宿主细胞中含有如本发明第三方面所述的载体,或基因组中整合有如本发明第二方面所述的多核苷酸。In the fourth aspect of the present invention, a host cell is provided, the host cell contains the vector as described in the third aspect of the present invention, or the polynucleotide as described in the second aspect of the present invention is integrated into the genome.
在另一优选例中,所述宿主细胞为大肠杆菌。In another preferred embodiment, the host cell is Escherichia coli.
在另一优选例中,所述宿主细胞选自下组:BL21(DE3)、Rosetta、Origami,或其组合。In another preferred embodiment, the host cell is selected from the group consisting of BL21(DE3), Rosetta, Origami, or a combination thereof.
在本发明的第五方面,提供了一种生产如本发明第一方面所述的融合蛋白的方法,包括步骤:In the fifth aspect of the present invention, a method for producing the fusion protein according to the first aspect of the present invention is provided, which includes the steps:
在适合表达的条件下,培养如权利要求4所述的宿主细胞,从而表达出如本发明第一方面所述的融合蛋白。Under conditions suitable for expression, the host cell according to claim 4 is cultured, thereby expressing the fusion protein according to the first aspect of the present invention.
在本发明的第六方面,提供了一种药物组合物,包括:In the sixth aspect of the present invention, there is provided a pharmaceutical composition comprising:
(a)如本发明第一方面所述的融合蛋白或其编码基因;(a) The fusion protein or its encoding gene according to the first aspect of the present invention;
(b)药学上可接受的载体。(b) A pharmaceutically acceptable carrier.
在另一优选例中,所述组分(a)的含量为0.1-99.9wt%,较佳地10-99.9wt%,更佳地70%-99.9wt%。In another preferred example, the content of the component (a) is 0.1-99.9% by weight, preferably 10-99.9% by weight, more preferably 70%-99.9% by weight.
在另一优选例中,所述药物组合物为液态、固体、或半固体。In another preferred embodiment, the pharmaceutical composition is liquid, solid, or semi-solid.
在另一优选例中,所述的药物组合物的剂型为口服剂型、注射剂、或外用药物剂型。In another preferred embodiment, the dosage form of the pharmaceutical composition is an oral dosage form, an injection, or a topical pharmaceutical dosage form.
在另一优选例中,所述药物组合物的剂型包括片剂、颗粒剂、胶囊、口服液、或注射剂。In another preferred embodiment, the dosage form of the pharmaceutical composition includes tablets, granules, capsules, oral liquids, or injections.
在另一优选例中,所述药物组合物为液态组合物。In another preferred embodiment, the pharmaceutical composition is a liquid composition.
在另一优选例中,所述的药学上可接受的载体选自下组:输液剂载体和/或注射剂载体,较佳地,所述的载体是选自下组的一种或多种载体:生理盐水、葡萄糖盐水、或其组合。In another preferred embodiment, the pharmaceutically acceptable carrier is selected from the following group: infusion carrier and/or injection carrier, preferably, the carrier is one or more carriers selected from the following group : Normal saline, dextrose saline, or a combination thereof.
在另一优选例中,所述药物组合物可单独使用,或与其他抗肿瘤药物联合使用。In another preferred embodiment, the pharmaceutical composition can be used alone or in combination with other anti-tumor drugs.
在本发明的第七方面,提供了一种如本发明第一方面所述的融合蛋白、如本发明第二方面所述的多核苷酸、如本发明第三方面所述的载体和如本发明第四方面所述的宿主细胞的用途,用于制备一制剂或药物组合物,所述制剂或药物组合物用于选自下组的一种或多种:In the seventh aspect of the present invention, there is provided a fusion protein as described in the first aspect of the present invention, a polynucleotide as described in the second aspect of the present invention, a vector as described in the third aspect of the present invention, and a fusion protein as described in the present invention. The use of the host cell according to the fourth aspect of the invention is for preparing a preparation or pharmaceutical composition, and the preparation or pharmaceutical composition is used for one or more selected from the following group:
(a)杀死肿瘤微环境中的肿瘤细胞;(a) Kill tumor cells in the tumor microenvironment;
(b)提高肿瘤微环境中的M1型巨噬细胞的数量,并降低肿瘤微环境中M2型巨噬细胞的数量;(b) Increase the number of M1 macrophages in the tumor microenvironment, and reduce the number of M2 macrophages in the tumor microenvironment;
(c)提高肿瘤微环境中的抗癌细胞因子、抗原提呈分子、效应T细胞、免疫原性细胞死亡(ICD)相关特征分子(如ATP、HMGB1、CRT)、LDH等促炎因子的水平;(c) Increase the level of anti-cancer factors, antigen presenting molecules, effector T cells, immunogenic cell death (ICD) related characteristic molecules (such as ATP, HMGB1, CRT), LDH and other pro-inflammatory factors in the tumor microenvironment ;
(d)降低肿瘤增殖转移相关蛋白、促癌细胞因子的表达水平。(d) Reduce the expression level of tumor proliferation and metastasis-related proteins and cancer cell factors.
在另一优选例中所述免疫原性细胞死亡(ICD)相关特征分子选自下组:ATP、HMGB1、CRT,或其组合。In another preferred embodiment, the immunogenic cell death (ICD) related characteristic molecules are selected from the following group: ATP, HMGB1, CRT, or a combination thereof.
在另一优选例中,所述的肿瘤选自下组:乳腺癌、结肠癌、前列腺癌、卵巢肿瘤,或其组合。In another preferred embodiment, the tumor is selected from the group consisting of breast cancer, colon cancer, prostate cancer, ovarian tumor, or a combination thereof.
在另一优选例中,所述的肿瘤细胞为4T1细胞或CT26结肠癌细胞。In another preferred example, the tumor cells are 4T1 cells or CT26 colon cancer cells.
在另一优选例中,所述抗癌细胞因子选自下组:TNF-α、IL-1β、IL-2、IFN-γ,或其组合。In another preferred embodiment, the anti-cancer factor is selected from the group consisting of TNF-α, IL-1β, IL-2, IFN-γ, or a combination thereof.
在另一优选例中,所述抗原提呈分子为MHC I类分子或MHC II类分子。In another preferred embodiment, the antigen presenting molecule is MHC class I molecule or MHC class II molecule.
在另一优选例中,所述效应T细胞选自下组:CD8 +T细胞、CD4 +T细胞、CD8 +&GranzymeB +T细胞、CD8 +&IFN-γ +T细胞,或其组合。 In another preferred embodiment, the effector T cells are selected from the group consisting of CD8 + T cells, CD4 + T cells, CD8 + &GranzymeB + T cells, CD8 + &IFN-γ + T cells, or a combination thereof.
在另一优选例中,所述肿瘤增殖转移相关蛋白为MR或Legumain。In another preferred embodiment, the tumor proliferation and metastasis-related protein is MR or Legumain.
在另一优选例中,所述促癌细胞因子为TGF-β。In another preferred embodiment, the cancer-promoting factor is TGF-β.
在本发明的第八方面,提供了一种治疗肿瘤的方法,包括步骤:向有所需要的对象施用如本发明第一方面所述的融合蛋白、如本发明第二方面所述的多核苷酸、如本发明第三方面所述的载体、如本发明第四方面所述的宿主细胞,或如本发明第六方面所述的药物组合物。In the eighth aspect of the present invention, there is provided a method for treating tumors, comprising the steps of: administering the fusion protein according to the first aspect of the present invention and the polynucleoside according to the second aspect of the present invention to a subject in need Acid, the vector according to the third aspect of the present invention, the host cell according to the fourth aspect of the present invention, or the pharmaceutical composition according to the sixth aspect of the present invention.
在另一优选例中,所述对象包括人或非人哺乳动物。In another preferred embodiment, the subject includes humans or non-human mammals.
在另一优选例中,所述非人哺乳动物包括:啮齿动物(如大鼠、小鼠)、灵长动物(如猴)。In another preferred embodiment, the non-human mammals include rodents (such as rats and mice) and primates (such as monkeys).
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。It should be understood that within the scope of the present invention, the above-mentioned technical features of the present invention and the technical features specifically described in the following (such as the embodiments) can be combined with each other to form a new or preferred technical solution. Due to space limitations, I will not repeat them here.
附图说明Description of the drawings
图1显示了重组焦亡蛋白的FPLC脱盐柱及分子筛纯化图。Figure 1 shows the FPLC desalting column and molecular sieve purification diagram of recombinant pyopterin.
其中,(A-C)依次为GSDMA3、GSDMA3-PTN和GSDMA3-TAT-PTN经过脱盐柱的FPLC色谱图;(D-E)依次为GSDMA3、GSDMA3-PTN和GSDMA3-TAT-PTN经过分子筛纯化的FPLC色谱图。Among them, (A-C) are the FPLC chromatograms of GSDMA3, GSDMA3-PTN and GSDMA3-TAT-PTN after passing through the desalting column; (D-E) are the FPLC chromatograms of GSDMA3, GSDMA3-PTN and GSDMA3-TAT-PTN after molecular sieve purification.
图2显示了重组蛋白表征图。Figure 2 shows the characterization diagram of the recombinant protein.
其中,泳道M为Marker;泳道1-3、泳道4-6、泳道7-9依次为GSDMA3-TAT-PTN、GSDMA3-PTN、GSDMA3经过Superdex 75纯化后三个吸收峰的蛋白样品。Among them, lane M is Marker; lanes 1-3, lanes 4-6, and lanes 7-9 are the protein samples with three absorption peaks after GSDMA3-TAT-PTN, GSDMA3-PTN, and GSDMA3 are purified by Superdex 75.
图3显示了体外培养的细胞系表达Legumain酶的水平。Figure 3 shows the level of Legumain enzyme expression in cell lines cultured in vitro.
图4显示了融合焦亡蛋白体外Legumain酶切实验。Figure 4 shows the in vitro Legumain cleavage experiment of the fusion pyrolysis protein.
其中,泳道M是Marker;泳道1是Legumain酶切后的GSDMA3-TAT-PTN;泳道2和3是Legumain酶切后的GSDMA3-PTN;泳道4是Legumain酶切后的GSDMA3。Among them, lane M is Marker; lane 1 is GSDMA3-TAT-PTN after Legumain digestion; lanes 2 and 3 are GSDMA3-PTN after Legumain digestion; lane 4 is GSDMA3 after Legumain digestion.
图5显示了细胞摄取实验的结果。Figure 5 shows the results of the cell uptake experiment.
其中,(A)显示了流式细胞仪检测酶切后焦亡蛋白的摄取结果;(B)显示了摄取结果的统计分析结果。Among them, (A) shows the results of the uptake of pyro-depth protein detected by flow cytometry after digestion; (B) shows the results of statistical analysis of the uptake results.
图6显示了焦亡蛋白的毒性实验结果。Figure 6 shows the results of the toxicity experiment of pyrooptin.
其中,(A)显示了焦亡蛋白对4T1细胞的杀伤作用;(B)显示了焦亡蛋白对DC2.4细胞的杀伤作用。Among them, (A) shows the killing effect of pyrodepressin on 4T1 cells; (B) shows the killing effect of pyrodepressin on DC2.4 cells.
图7显示了重组蛋白处理后的细胞形态变化情况。Figure 7 shows the changes in cell morphology after recombinant protein treatment.
其中,(A-D)依次为PBS、GSDMA3、GSDMA3-PTN、GSDMA3-TAT-PTN给药处理组的细胞的明场拍摄结果。Among them, (A-D) are the results of bright field photography of cells in the PBS, GSDMA3, GSDMA3-PTN, and GSDMA3-TAT-PTN administration treatment groups in order.
图8显示了焦亡蛋白介导肿瘤细胞免疫原性死亡中引起CRT外翻实验结果。Figure 8 shows the experimental results of CRT eversion caused by the pyrooptin-mediated immunogenic death of tumor cells.
其中,(A-D)依次为PBS、GSDMA3、酶切后的GSDMA3-PTN、酶切后的GSDMA3-TAT-PTN处理肿瘤细胞的结果;(E)显示了CRT的统计分析。Among them, (A-D) are the results of PBS, GSDMA3, digested GSDMA3-PTN, and digested GSDMA3-TAT-PTN in order of treating tumor cells; (E) shows the statistical analysis of CRT.
图9(A)显示了HMGB1的释放量;(B)显示了释放到胞外的ATP的含量。Figure 9 (A) shows the released amount of HMGB1; (B) shows the amount of ATP released outside the cell.
图10显示了DC的体外致敏实验结果。Figure 10 shows the results of the in vitro sensitization experiment of DC.
其中,(A)显示了焦亡蛋白处理后CD80 +的DC细胞量;(B)显示了焦亡蛋白处理后CD86 +的DC细胞量;(C)显示了焦亡蛋白处理后CD80 +/CD86 +双阳性的DC细胞量。 Among them, (A) shows the amount of CD80 + DC cells after pyrolyst protein treatment; (B) shows the amount of CD86 + DC cells after pyrolyst protein treatment; (C) shows CD80 + /CD86 after pyrolyst protein treatment + The amount of double positive DC cells.
图11显示了焦亡蛋白对抗原呈递的作用。Figure 11 shows the effect of pyroptin on antigen presentation.
图12显示了重组焦亡蛋白在鼠源4T1乳腺癌原位瘤上的药效实验结果。Figure 12 shows the results of the pharmacodynamic experiment of recombinant pyrooptosis protein on murine 4T1 breast cancer in situ tumors.
其中,(A)为肿瘤体积变化图;(B)为药效流程图;(C)为小鼠体重变化曲线图;(D)显示了实验终点肿瘤的重量图;(E)显示了实验终点时各治疗组的抑瘤率;(F)为实验终点剖出的肿瘤照片(*P<0.05,**P<0.01,***P<0.001和****P<0.0001)。Among them, (A) is the tumor volume change graph; (B) is the drug effect flow chart; (C) is the mouse body weight change curve; (D) shows the weight graph of the tumor at the experimental end point; (E) shows the experimental end point Tumor inhibition rate of each treatment group at time; (F) is the photo of the tumor at the end of the experiment (*P<0.05, **P<0.01, ***P<0.001 and ****P<0.0001).
图13显示了脾脏中T细胞及其颗粒酶的示意图。Figure 13 shows a schematic diagram of T cells and their granzymes in the spleen.
其中,(A-C)显示了以PBS组脾脏为例,流式画门的示意图;(D-G)依次显示了PBS组、GSDMA3组、GSDMA3-PTN组和GSDMA3-TAT-PTN组的CD8 +T细胞的颗粒酶分泌量的变化情况。 Among them, (AC) shows the spleen of the PBS group as an example, and the schematic diagram of the flow gate; (DG) shows the CD8 + T cells in the PBS group, GSDMA3 group, GSDMA3-PTN group and GSDMA3-TAT-PTN group in sequence. Changes in granzyme secretion.
图14显示了各治疗组的脾脏中T细胞及其其分泌的因子的变化情况。Figure 14 shows the changes of T cells and their secreted factors in the spleen of each treatment group.
其中,(A)显示了脾脏中CD4 +T细胞量的变化情况;(B)显示了脾脏中CD8 +T细胞量的变化情况;(C)显示了治疗后各组脾脏中CD8 +&GranzymeB +T细胞的变化情况;(D)显示了治疗后各组脾脏中CD8 +&IFN-γ +T细胞的变化情况。 Among them, (A) shows the change in the amount of CD4 + T cells in the spleen; (B) shows the change in the amount of CD8 + T cells in the spleen; (C) shows the CD8 + &GranzymeB + T in the spleen of each group after treatment Cell changes; (D) shows the changes of CD8 + &IFN-γ + T cells in the spleen of each group after treatment.
图15显示了药物治疗后肿瘤内T细胞及其分泌的因子的变化情况。Figure 15 shows the changes of T cells and their secreted factors in tumors after drug treatment.
其中,(A)显示了肿瘤组织中CD4 +T细胞量的变化情况;(B)显示了肿瘤组织中CD8 +T细胞量的变化情况;(C)显示了治疗后各组肿瘤肿瘤中CD8 +&Granzyme +T细胞的变化情况;(D)显示了治疗后各组肿瘤组织中CD8 +&IFN-γ +T细胞的变化情况。 Among them, (A) shows the change in the amount of CD4 + T cells in the tumor tissue; (B) shows the change in the amount of CD8 + T cells in the tumor tissue; (C) shows the CD8 + in each group of tumors after treatment The changes of &Granzyme + T cells; (D) shows the changes of CD8 + &IFN-γ + T cells in the tumor tissues of each group after treatment.
图16显示了药物治疗后肿瘤内T细胞及其其分泌的因子的变化情况。Figure 16 shows the changes of T cells and their secreted factors in tumors after drug treatment.
其中,(A)显示了淋巴结中CD4 +T细胞量的变化情况;(B)显示了淋巴结中CD8 +T细胞量的变化情况;(C)显示了治疗后各组淋巴结中CD8 +&Granzyme +T细胞的变化情况;(D)显示了治疗后各组淋巴结中CD8 +&IFN-γ +T细胞的变化情况。 Among them, (A) shows the change in the amount of CD4 + T cells in the lymph nodes; (B) shows the change in the amount of CD8 + T cells in the lymph nodes; (C) shows the CD8 + &Granzyme + T in each group of lymph nodes after treatment Cell changes; (D) shows the changes of CD8 + &IFN-γ + T cells in the lymph nodes of each group after treatment.
图17显示了给药治疗后肿瘤内巨噬细胞及NK细胞的变化。Figure 17 shows the changes of macrophages and NK cells in tumors after administration of treatment.
其中,(A-B)显示了M1型巨噬细胞的变化;(C-D)显示了M2型巨噬细胞的变化;(E-F)显示了NK细胞的变化。其中(A、D、E)均是以GSDMA3-TAT-PTN治疗组的肿瘤细胞为例。Among them, (A-B) shows the changes of M1 type macrophages; (C-D) shows the changes of M2 type macrophages; (E-F) shows the changes of NK cells. Among them (A, D, E) are the tumor cells in the GSDMA3-TAT-PTN treatment group as an example.
图18显示了不同治疗组肿瘤组织中相关蛋白表达量的变化情况。Figure 18 shows the changes in the expression of related proteins in tumor tissues in different treatment groups.
图19显示了肿瘤组织中细胞因子的变化。Figure 19 shows the changes of cytokines in tumor tissues.
其中,A-B依次表示实验终点肿瘤组织中IL-2和TGF-β的变化情况。Among them, A-B indicate the changes of IL-2 and TGF-β in the tumor tissue at the end of the experiment in turn.
图20显示了主要脏器的重量改变。Figure 20 shows the changes in the weight of the main organs.
图21显示了主要脏器的病理切片。Figure 21 shows a pathological section of the main organs.
其中,标尺为100μm。Among them, the scale is 100μm.
图22显示了实验终点小鼠的肝功能和肾功能的检测结果。Figure 22 shows the test results of liver function and kidney function of mice at the end of the experiment.
其中,(A-C)分别表示肝功能相关的谷丙转氨酶、谷草转氨酶和总胆红素含量的变化;(D-F)分别表示和肾功能相关的血清尿素、血清肌酐和血清尿酸的含量变化。Among them, (A-C) respectively represent the changes of alanine aminotransferase, aspartate aminotransferase and total bilirubin content related to liver function; (D-F) represent the changes of serum urea, serum creatinine and serum uric acid content related to renal function, respectively.
具体实施方式detailed description
本发明人经过广泛而深入的研究,经过大量的筛选,首次开发了一种重组焦亡蛋白递药系统。After extensive and in-depth research and extensive screening, the inventors developed a recombinant pyroptosis protein delivery system for the first time.
本发明人以细胞焦亡作用及焦亡蛋白Gasdermin A3(GSDMA3)为研究对象,采用基因工程技术对其进行了结构改造,在焦亡蛋白两个结构域之间引入Legumain底物肽序列PTN。并且,选用富含精氨酸的阳离子穿膜肽TAT(RKKRRQRRR),将其构建在PTN序列相邻位置并靠近焦亡蛋白N端一侧(N-GSDMA3-TAT-PTN-GSDMA3-C,或记为记为GSDMA3-TAT-PTN)。实验结果表明,本发明中的GSDMA3-TAT-PTN蛋白在体内外实验中表现出非常好的抗肿瘤活性。The present inventors took the cell pyrolysis and the pyrolysis protein Gasdermin A3 (GSDMA3) as the research object, and used genetic engineering technology to modify its structure, and introduced the Legumain substrate peptide sequence PTN between the two structural domains of the pyrolysis protein. In addition, the arginine-rich cationic penetrating peptide TAT (RKKRRQRRR) was selected and constructed in the adjacent position of the PTN sequence and close to the N-terminal side of the pyrolysis protein (N-GSDMA3-TAT-PTN-GSDMA3-C, or Marked as GSDMA3-TAT-PTN). The experimental results show that the GSDMA3-TAT-PTN protein of the present invention exhibits very good anti-tumor activity in in vivo and in vitro experiments.
在此基础上完成了本发明。The present invention has been completed on this basis.
本发明融合蛋白及其编码序列Fusion protein of the present invention and its coding sequence
如本文所用,术语“本发明融合蛋白”、“重组焦亡蛋白”、“融合焦亡蛋白”可互换使用,是指本发明第一方面所述的融合蛋白,其具有诱导细胞膜破裂并释放大量内含物引起机体炎症反应的功能。As used herein, the terms "fusion protein of the present invention", "recombinant pyroptosis protein" and "fusion pyroptosis protein" are used interchangeably and refer to the fusion protein according to the first aspect of the present invention, which has the ability to induce cell membrane rupture and release A large number of inclusions cause the function of the body's inflammatory response.
在本发明中,提供的融合蛋白从N端到C端具有式I所示的结构:In the present invention, the provided fusion protein has the structure shown in formula I from N-terminus to C-terminus:
Z0-Z1-Z2-Z3-Z4   (式I)Z0-Z1-Z2-Z3-Z4 (Formula I)
式中,Where
Z0为任选的标签元件;Z1为焦亡蛋白的N结构域元件;Z2为穿膜肽序列元件;Z3为能够被肿瘤微环境中特异性表达的蛋白酶特异性切割的肽序列元件;Z4为焦亡蛋白的C结构域元件;“-”表示连接上述元件的肽键;Z0 is an optional tag element; Z1 is the N-domain element of pyrooptin; Z2 is a penetrating peptide sequence element; Z3 is a peptide sequence element that can be specifically cleaved by a protease specifically expressed in the tumor microenvironment; Z4 is The C domain element of pyrolysis protein; "-" represents the peptide bond connecting the above elements;
其中,在融合蛋白中,所述Z4元件通过特异性地结合Z1元件来抑制Z1元件的活性。Wherein, in the fusion protein, the Z4 element inhibits the activity of the Z1 element by specifically binding to the Z1 element.
在本发明中,所述的焦亡蛋白可选自人源焦亡蛋白(例如GSDMA、GSDMB、GSDMC、GSDMD、DFNA5(GSDME)、DFNB59(GSDMF)等)或鼠源焦亡蛋白(例如GSDMA1、GSDMA2、GSDMA3、GSDMC1、GSDMC2、GSDMC3、GSDMC4等)。In the present invention, the pyrolyst protein can be selected from human pyrolyt protein (such as GSDMA, GSDMB, GSDMC, GSDMD, DFNA5 (GSDME), DFNB59 (GSDMF), etc.) or murine pyrolyt protein (such as GSDMA1, GSDMA2, GSDMA3, GSDMC1, GSDMC2, GSDMC3, GSDMC4, etc.).
在一个优选的实施方式中,所述焦亡蛋白为GSDMA3。In a preferred embodiment, the pyroptin protein is GSDMA3.
在本发明的融合蛋白中,所述Z1中,所述N结构域为具有在细胞膜上形成孔洞的活性蛋白结构域;所述Z2中,所述穿膜肽具有携带不同成分穿过细胞膜的功能。In the fusion protein of the present invention, in the Z1, the N domain is an active protein domain that forms holes in the cell membrane; in the Z2, the penetrating peptide has the function of carrying different components through the cell membrane .
优选地,所述Z2中,所述穿膜肽选自下组:阳离子细胞穿膜肽(如TAT)、疏水性细胞穿膜肽、两亲性细胞穿膜肽。在一个优选的实施方式中,所述穿膜肽为 TAT。Preferably, in the Z2, the penetrating peptide is selected from the group consisting of cationic cell penetrating peptides (such as TAT), hydrophobic cell penetrating peptides, and amphiphilic cell penetrating peptides. In a preferred embodiment, the penetrating peptide is TAT.
在本发明的融合蛋白中,所述Z3中,所述肿瘤微环境中特异性表达的蛋白酶选自下组:天冬酰胺内肽酶(Legumain)、基质金属蛋白酶(例如MMP-2、MMP-7、MMP-9、MMP-12等),或其组合。In the fusion protein of the present invention, in the Z3, the protease specifically expressed in the tumor microenvironment is selected from the following group: asparagine endopeptidase (Legumain), matrix metalloproteinase (such as MMP-2, MMP- 7. MMP-9, MMP-12, etc.), or a combination thereof.
在一个优选的实施方式中,所述Z3中,所述肿瘤微环境中特异性表达的蛋白酶为天冬酰胺内肽酶。In a preferred embodiment, in the Z3, the protease specifically expressed in the tumor microenvironment is asparagine endopeptidase.
优选地,所述Z3中,所述肽序列选自:Legumain特异性识别并切割的底物肽序列PTN。Preferably, in the Z3, the peptide sequence is selected from: a substrate peptide sequence PTN specifically recognized and cleaved by Legumain.
应理解,尽管本发明的实例中提供的基因是鼠源的,但是来源于其它类似的物种(尤其是哺乳动物)的、与本发明的序列(优选地,序列如SEQ ID NO:5所示)具有一定同源性(保守性)的重组焦亡蛋白的基因序列,也包括在本发明的范围内,只要本领域技术人员在阅读了本申请后根据本申请提供的信息可以方便地从其它物种(尤其是哺乳动物)中分离得到该序列。It should be understood that although the genes provided in the examples of the present invention are of murine origin, they are derived from other similar species (especially mammals) and are compatible with the sequence of the present invention (preferably, the sequence is shown in SEQ ID NO: 5). ) The gene sequence of recombinant pyroopterin protein with certain homology (conservation) is also included in the scope of the present invention, as long as those skilled in the art can easily obtain information from other sources after reading this application according to the information provided in this application. The sequence is isolated from species (especially mammals).
本发明的多核苷酸可以是DNA形式或RNA形式。DNA形式包括:DNA、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。编码融合蛋白的编码区序列可以与SEQ ID NO:5所示的编码区序列相同或者是简并的变异体。The polynucleotide of the present invention may be in the form of DNA or RNA. DNA forms include: DNA, genomic DNA or synthetic DNA. DNA can be single-stranded or double-stranded. DNA can be a coding strand or a non-coding strand. The coding region sequence encoding the fusion protein may be the same as the coding region sequence shown in SEQ ID NO: 5 or a degenerate variant.
编码融合蛋白的多核苷酸包括:只编码融合蛋白的编码序列;融合蛋白的编码序列和各种附加编码序列;融合蛋白的编码序列(和任选的附加编码序列)以及非编码序列。The polynucleotide encoding the fusion protein includes: only the coding sequence of the fusion protein; the coding sequence of the fusion protein and various additional coding sequences; the coding sequence (and optional additional coding sequence) of the fusion protein and non-coding sequences.
术语“编码融合蛋白的多核苷酸”可以是包括编码此融合蛋白的多核苷酸,也可以是还包括附加编码和/或非编码序列的多核苷酸。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多苷或多肽的片段、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的融合蛋白的功能。The term "polynucleotide encoding a fusion protein" may include a polynucleotide encoding the fusion protein, or a polynucleotide that also includes additional coding and/or non-coding sequences. The present invention also relates to variants of the aforementioned polynucleotides, which encode fragments, analogs and derivatives of polyglycosides or polypeptides having the same amino acid sequence as the present invention. The variants of this polynucleotide can be naturally occurring allelic variants or non-naturally occurring variants. These nucleotide variants include substitution variants, deletion variants and insertion variants. As known in the art, an allelic variant is an alternative form of a polynucleotide. It may be a substitution, deletion or insertion of one or more nucleotides, but does not substantially change the fusion protein encoded by it. Features.
本发明还涉及与上述的序列杂交且两个序列之间具有至少50%,较佳地至 少70%,更佳地至少80%相同性的多核苷酸。本发明特别涉及在严格条件下与本发明所述多核苷酸可杂交的多核苷酸。在本发明中,“严格条件”是指:(1)在较低离子强度和较高温度下的杂交和洗脱,如0.2×SSC,0.1%SDS,60℃;或(2)杂交时加有变性剂,如50%(v/v)甲酞胺,0.1%小牛血清/0.1%Ficoll,42℃等;或(3)仅在两条序列之间的相同性至少在90%以上,更好是95%以上时才发生杂交。The present invention also relates to polynucleotides that hybridize with the aforementioned sequences and have at least 50%, preferably at least 70%, and more preferably at least 80% identity between the two sequences. The present invention particularly relates to polynucleotides that can hybridize with the polynucleotides of the present invention under stringent conditions. In the present invention, "stringent conditions" refer to: (1) hybridization and elution at lower ionic strength and higher temperature, such as 0.2×SSC, 0.1% SDS, 60°C; or (2) adding during hybridization There are denaturants, such as 50% (v/v) methylphthalamide, 0.1% calf serum/0.1% Ficoll, 42°C, etc.; or (3) only the identity between the two sequences is at least 90% or more, More preferably, the hybridization occurs when 95% or more occurs.
编码本发明的融合蛋白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,可根据本发明所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DNA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。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。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。The full-length nucleotide sequence or fragments thereof encoding the fusion protein of the present invention can usually be obtained by PCR amplification method, recombination method or artificial synthesis method. For the PCR amplification method, primers can be designed according to the relevant nucleotide sequence disclosed in the present invention, especially the open reading frame sequence, and a commercially available DNA library or a cDNA prepared by a conventional method known to those skilled in the art can be used. The library is used as a template to amplify the relevant sequences. When the sequence is long, it is often necessary to perform two or more PCR amplifications, and then splice the amplified fragments together in the correct order. Once the relevant sequence is obtained, the recombination method can be used to obtain the relevant sequence in large quantities. It is usually cloned into a vector, and then transferred into a cell, and then the relevant sequence is isolated 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。目前,已经可以完全通过化学合成来得到编码本发明融合蛋白(或其片段,或其衍生物)的DNA序列。然后可将该DNA序列引入本领域中已知的各种现有的DNA分子(或如载体)和细胞中。此外,还可通过化学合成将突变引入本发明融合蛋白序列中。In addition, artificial synthesis methods can also be used to synthesize related sequences, especially when the fragment length is short. Usually, by first synthesizing multiple small fragments, and then ligating to obtain fragments with very long sequences. At present, the DNA sequence encoding the fusion protein (or fragment or derivative thereof) of the present invention can be obtained completely through chemical synthesis. The DNA sequence can then be introduced into various existing DNA molecules (or such as vectors) and cells known in the art. In addition, mutations can also be introduced into the fusion protein sequence of the present invention through chemical synthesis.
本发明涉及一种用于抗肿瘤递药系统的重组焦亡蛋白融合蛋白,在本发明的一个优选例中,所述融合蛋白的氨基酸序列如SEQ ID NO:4所示。本发明的多肽能够有效诱导细胞膜破裂并释放大量内含物引起机体炎症反应。The present invention relates to a recombinant pyroptin fusion protein used in an anti-tumor drug delivery system. In a preferred embodiment of the present invention, the amino acid sequence of the fusion protein is shown in SEQ ID NO: 4. The polypeptide of the present invention can effectively induce the rupture of the cell membrane and release a large amount of contents to cause inflammation in the body.
本发明还包括与本发明的SEQ ID NO:4所示序列具有50%或以上(优选60%以上,70%以上,80%以上,更优选90%以上,更优选95%以上,最优选98%以上,如99%)同源性的具有相同或相似功能的多肽或蛋白。The present invention also includes 50% or more of the sequence shown in SEQ ID NO: 4 of the present invention (preferably 60% or more, 70% or more, 80% or more, more preferably 90% or more, more preferably 95% or more, most preferably 98 % Or more, such as 99%) homologous polypeptides or proteins with the same or similar functions.
所述“相同或相似功能”主要是指:“有效诱导细胞膜破裂并释放大量内含物引起机体炎症反应”。The "same or similar function" mainly refers to: "effectively induce the rupture of the cell membrane and release a large amount of content to cause the body's inflammatory response".
本发明的融合蛋白可以是重组多肽、天然多肽、合成多肽。本发明的融合蛋白可以是天然纯化的产物,或是化学合成的产物,或使用重组技术从原核或真核宿主(例如,细菌、酵母、植物、昆虫和哺乳动物细胞)中产生。根据重组生产方案所用的宿主,本发明的融合蛋白可以是糖基化的,或可以是非糖基化的。本发明的融合蛋白还可包括或不包括起始的甲硫氨酸残基。The fusion protein of the present invention can be a recombinant polypeptide, a natural polypeptide, or a synthetic polypeptide. The fusion protein of the present invention can be a natural purified product, or a chemically synthesized product, or produced from a prokaryotic or eukaryotic host (for example, bacteria, yeast, plant, insect, and mammalian cells) using recombinant technology. Depending on the host used in the recombinant production protocol, the fusion protein of the present invention may be glycosylated or non-glycosylated. The fusion protein of the present invention may also include or not include the initial methionine residue.
本发明还包括具有本发明融合蛋白的活性的其他多肽片段和类似物。如本文所用,术语“片段”和“类似物”是指基本上保持本发明的融合蛋白相同的生物学功能或活性的多肽。The present invention also includes other polypeptide fragments and analogs having the activity of the fusion protein of the present invention. As used herein, the terms "fragment" and "analog" refer to polypeptides that substantially retain the same biological function or activity as the fusion protein of the present invention.
本发明的多肽片段、衍生物或类似物可以是:(i)有一个或多个保守或非保守性氨基酸残基(优选保守性氨基酸残基)被取代的多肽,而这样的取代的氨基酸残基可以是也可以不是由遗传密码编码的;或(ii)在一个或多个氨基酸残基中具有取代基团的多肽;或(iii)成熟多肽与另一个化合物(比如延长多肽半衰期的化合物,例如聚乙二醇)融合所形成的多肽;或(iv)附加的氨基酸序列融合到此多肽序列而形成的多肽(如前导序列或分泌序列或用来纯化此多肽的序列或蛋白原序列,或融合蛋白)。根据本文的定义这些片段、衍生物和类似物属于本领域熟练技术人员公知的范围。The polypeptide fragments, derivatives or analogues of the present invention may be: (i) polypeptides with one or more conservative or non-conservative amino acid residues (preferably conservative amino acid residues) substituted, and such substituted amino acid residues The base may or may not be encoded by the genetic code; or (ii) a polypeptide with a substitution group in one or more amino acid residues; or (iii) the mature polypeptide and another compound (such as a compound that extends the half-life of the polypeptide, For example, polyethylene glycol) fused to form a polypeptide; or (iv) additional amino acid sequence is fused to the polypeptide sequence to form a polypeptide (such as a leader sequence or secretory sequence, or a sequence or proprotein sequence used to purify the polypeptide, or Fusion protein). According to the definition herein, these fragments, derivatives and analogs belong to the scope well known to those skilled in the art.
本发明中,所述的融合蛋白变体是如SEQ ID NO:4所示的氨基酸序列,经过若干个(通常为1-10个,较佳地1-8个,更佳地1-4个,最佳地1-2个)取代、缺失或添加至少一个氨基酸所得的衍生序列,以及在C末端和/或N末端添加一个或数个(通常为10个以内,较佳地为5个以内,更佳地为3个以内)氨基酸。例如,在所述蛋白中,用性能相近或相似的氨基酸进行取代时,通常不会改变蛋白质的功能,在C末端和/或N末端添加一个或数个(如1-3个)氨基酸通常也不会改变蛋白质的功能。这些保守性变异最好根据表1进行替换而产生。In the present invention, the fusion protein variant is the amino acid sequence shown in SEQ ID NO: 4, after several (usually 1-10, preferably 1-8, more preferably 1-4 , Preferably 1-2) the derived sequence obtained by substituting, deleting or adding at least one amino acid, and adding one or several (usually within 10, preferably within 5) at the C-terminus and/or N-terminus , More preferably within 3) amino acids. For example, when the protein is substituted with amino acids with similar or similar properties, it usually does not change the function of the protein. Adding one or several (such as 1-3) amino acids at the C-terminus and/or N-terminus is usually also Does not change the function of the protein. These conservative variants are best produced by substitutions according to Table 1.
表1Table 1
最初的残基Initial residue 代表性的取代Representative substitution 优选的取代Preferred substitution
Ala(A)Ala(A) Val;Leu;IleVal; Leu; Ile ValVal
Arg(R)Arg(R) Lys;Gln;AsnLys; Gln; Asn LysLys
Asn(N)Asn(N) Gln;His;Lys;ArgGln; His; Lys; Arg GlnGln
Asp(D)Asp(D) GluGlu GluGlu
Cys(C)Cys(C) SerSer SerSer
Gln(Q)Gln(Q) AsnAsn AsnAsn
Glu(E)Glu(E) AspAsp AspAsp
Gly(G)Gly(G) Pro;AlaPro; Ala AlaAla
His(H)His(H) Asn;Gln;Lys;ArgAsn; Gln; Lys; Arg ArgArg
Ile(I)Ile(I) Leu;Val;Met;Ala;PheLeu; Val; Met; Ala; Phe LeuLeu
Leu(L)Leu(L) Ile;Val;Met;Ala;PheIle; Val; Met; Ala; Phe IleIle
Lys(K)Lys(K) Arg;Gln;AsnArg; Gln; Asn ArgArg
Met(M)Met(M) Leu;Phe;IleLeu; Phe; Ile LeuLeu
Phe(F)Phe(F) Leu;Val;Ile;Ala;TyrLeu; Val; Ile; Ala; Tyr LeuLeu
Pro(P)Pro(P) AlaAla AlaAla
Ser(S)Ser(S) ThrThr ThrThr
Thr(T)Thr(T) SerSer SerSer
Trp(W)Trp(W) Tyr;PheTyr; Phe TyrTyr
Tyr(Y)Tyr(Y) Trp;Phe;Thr;SerTrp; Phe; Thr; Ser PhePhe
Val(V)Val(V) Ile;Leu;Met;Phe;AlaIle; Leu; Met; Phe; Ala LeuLeu
本发明还包括所要求保护的蛋白的类似物。这些类似物与天然SEQ ID NO:4差别可以是氨基酸序列上的差异,也可以是不影响序列的修饰形式上的差异,或者兼而有之。这些蛋白的类似物包括天然或诱导的遗传变异体。诱导变异体可以通过各种技术得到,如通过辐射或暴露于诱变剂而产生随机诱变,还可通过定点诱变法或其他已知分子生物学的技术。类似物还包括具有不同于天然L-氨基酸的残基(如D-氨基酸)的类似物,以及具有非天然存在的或合成的氨基酸(如β、γ-氨基酸)的类似物。应理解,本发明的蛋白并不限于上述列举的代表性的蛋白。The present invention also includes analogs of the claimed protein. The difference between these analogs and the natural SEQ ID NO: 4 may be the difference in the amino acid sequence, the difference in the modified form that does not affect the sequence, or both. Analogs of these proteins include natural or induced genetic variants. Induced variants can be obtained by various techniques, such as random mutagenesis by radiation or exposure to mutagens, site-directed mutagenesis or other known molecular biology techniques. Analogs also include analogs having residues different from natural L-amino acids (such as D-amino acids), and analogs having non-naturally occurring or synthetic amino acids (such as β, γ-amino acids). It should be understood that the protein of the present invention is not limited to the representative proteins listed above.
修饰(通常不改变一级结构)形式包括:体内或体外蛋白的化学衍生形式如乙酸化或羧基化。修饰还包括糖基化,如那些在蛋白质合成和加工中进行糖基化修饰。这种修饰可以通过将蛋白暴露于进行糖基化的酶(如哺乳动物的糖基化酶或去糖基化酶)而完成。修饰形式还包括具有磷酸化氨基酸残基(如磷酸酪氨酸,磷酸丝氨酸,磷酸苏氨酸)的序列。Modified (usually not changing the primary structure) forms include: chemically derived forms of proteins in vivo or in vitro, such as acetate or carboxylation. Modifications also include glycosylation, such as those that undergo glycosylation modifications during protein synthesis and processing. This modification can be accomplished by exposing the protein to an enzyme that performs glycosylation (such as a mammalian glycosylase or deglycosylase). Modified forms also include sequences with phosphorylated amino acid residues (such as phosphotyrosine, phosphoserine, and phosphothreonine).
药物组合物及其施用方法Pharmaceutical composition and its application method
本发明提供了一种药物组合物,包括药学上可接受的载体和有效量的以下活性成分:如本发明第一方面所述的融合蛋白或其编码基因。The present invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and an effective amount of the following active ingredients: the fusion protein or its coding gene as described in the first aspect of the present invention.
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。As used herein, the term "effective amount" or "effective dose" refers to an amount that can produce function or activity on humans and/or animals and can be accepted by humans and/or animals.
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, "pharmaceutically acceptable" ingredients are substances that are suitable for humans and/or mammals without excessive side effects (such as toxicity, irritation, and allergic reactions), that is, substances that have a reasonable benefit/risk ratio. The term "pharmaceutically acceptable carrier" refers to a carrier used for the administration of a therapeutic agent, and includes 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活性成分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。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。The pharmaceutical composition of the present invention contains a safe and effective amount of the active ingredient of the present invention and a pharmaceutically acceptable carrier. Such carriers include (but are not limited to): saline, buffer, glucose, water, glycerol, ethanol, and combinations thereof. Usually the pharmaceutical preparation should match the mode of administration. For example, it can be prepared by conventional methods with physiological saline or an aqueous solution containing glucose and other adjuvants. The pharmaceutical composition should be manufactured under aseptic conditions.
本发明所述的活性成分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:所述的活性成分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。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 active ingredient of the present invention can vary with the mode of administration and the severity of the disease to be treated. The selection of the preferred effective amount can be determined by a person of ordinary skill in the art according to various factors (for example, through clinical trials). The factors include, but are not limited to: the pharmacokinetic parameters of the active ingredients such as bioavailability, metabolism, half-life, etc.; the severity of the disease to be treated by the patient, the patient's weight, the patient's immune status, and administration The way and so on. For example, due to the urgent requirement of treating the condition, several divided doses can be given every day, or the dose can be reduced proportionally.
本发明所述的药学上可接受的载体包括(但不限于):水、盐水、脂质体、脂质、蛋白、蛋白-抗体缀合物、肽类物质、纤维素、纳米凝胶、或其组合。载体的选择应与给药方式相匹配,这些都是本领域的普通技术人员所熟知的。The pharmaceutically acceptable carriers of the present invention include (but are not limited to): water, saline, liposomes, lipids, proteins, protein-antibody conjugates, peptides, cellulose, nanogels, or Its combination. The choice of carrier should match the mode of administration, which are well known to those of ordinary skill in the art.
本发明的主要优点包括:The main advantages of the present invention include:
1)本发明使用基因工程技术对焦亡蛋白进行改造,可实现焦亡蛋白多功能递药,在焦亡蛋白里引入穿膜肽序列,赋予了焦亡蛋白入胞能力,从而发挥焦亡作用。1) The present invention uses genetic engineering technology to transform the pyrooptosis protein, which can realize the multifunctional delivery of the pyroptosis protein. The penetrating peptide sequence is introduced into the pyroptosis protein, which gives the pyroptosis protein the ability to enter the cell, thereby exerting the pyroptosis effect.
2)本发明将焦亡蛋白应用于诱导免疫原性细胞死亡(ICD)及肿瘤免疫治疗。2) The present invention applies the pyroopterin to induce immunogenic cell death (ICD) and tumor immunotherapy.
3)本发明使用Legumain-PTN等肿瘤相关酶作为激活焦亡蛋白的响应式设计,具有较广泛的应用范围,并提高了肿瘤杀伤作用的选择性,降低其毒副作 用。3) The present invention uses tumor-related enzymes such as Legumain-PTN as a responsive design for activating pyroapone proteins, which has a wide range of applications, improves the selectivity of tumor killing effects, and reduces its toxic side effects.
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 present invention will be further explained below in conjunction with specific embodiments. It should be understood that these embodiments are only used to illustrate the present invention and not to limit the scope of the present invention. The experimental methods without specific conditions in the following examples usually follow conventional conditions, such as the conditions described in Sambrook et al., Molecular Cloning: Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989), or according to manufacturing The conditions suggested by the manufacturer. Unless otherwise specified, percentages and parts are percentages by weight and parts by weight.
方法method
利用基因工程技术对原核表达质粒pET28a-GSDMA3为模板进行亚克隆构建,将部分氨基酸序列替换成Legumain特异性酶切的底物肽序列PTN,并融入细胞穿膜肽序列TAT。构建成pET28a-PTN-GSDMA3、pET28a-PTN-TAT-GSDMA3质粒。重组的质粒在体外进行转化、并在大肠杆菌中实现高效表达,利用蛋白的His-tag标签进行初步纯化,然后通过分子筛进一步纯化,最后得到重组蛋白。Using genetic engineering technology, the prokaryotic expression plasmid pET28a-GSDMA3 was used as a template for subcloning and construction. Part of the amino acid sequence was replaced with Legumain-specific digested substrate peptide sequence PTN, and incorporated into the cell-penetrating peptide sequence TAT. Constructed into pET28a-PTN-GSDMA3, pET28a-PTN-TAT-GSDMA3 plasmids. The recombinant plasmid is transformed in vitro and expressed in Escherichia coli with high efficiency. The protein’s His-tag is used for preliminary purification, and then further purified by molecular sieve, and finally the recombinant protein is obtained.
重组焦亡蛋白质粒的构建Construction of recombinant pyroptosis protein pellet
以pET28a-GSDMA3为模板进行亚克隆构建,构建成pET28a-PTN-GSDMA3、pET28a-PTN-TAT-GSDMA3质粒。Using pET28a-GSDMA3 as a template for subcloning construction, construct pET28a-PTN-GSDMA3, pET28a-PTN-TAT-GSDMA3 plasmids.
重组融合蛋白的表达及纯化Expression and purification of recombinant fusion protein
将质粒转入感受态细胞进行培养,然后扩大培养并诱导蛋白表达,将得到的蛋白进行纯化,首先利用蛋白自带的His-tag进行Ni结合柱纯化,使用
Figure PCTCN2020111821-appb-000001
purifier 10(FPLC)将初步纯化的蛋白进行分子筛(Superdex 75)纯化,采用SDS-PAGE电泳检测蛋白的纯度。
The plasmid is transferred to competent cells for culture, and then the culture is expanded and the protein expression is induced, and the obtained protein is purified. First, use the His-tag that comes with the protein to purify the Ni binding column, and use
Figure PCTCN2020111821-appb-000001
Purifier 10 (FPLC) purifies the preliminarily purified protein by molecular sieve (Superdex 75), and uses SDS-PAGE electrophoresis to detect the purity of the protein.
细胞水平实验Cell level experiment
使用FITC染料标记蛋白,然后使用体外制备的Legumain酶切介质进行重组蛋白的酶切,将酶切后的重组蛋白进行细胞摄取实验。检测酶切后的重组蛋白对肿瘤细胞的杀伤能力并使用显微镜进行观察。检测重组蛋白引起细胞焦亡的过程中释放的ICD相关分子(包括CRT和HMGB1等)的量变化。观察重组蛋白对DC的致敏及提呈的影响。Use FITC dye to label the protein, and then use Legumain digestion medium prepared in vitro to digest the recombinant protein, and then perform the cell uptake experiment on the recombinant protein after digestion. Detect the killing ability of the recombinant protein after digestion on tumor cells and observe it with a microscope. Detection of changes in the amount of ICD-related molecules (including CRT and HMGB1, etc.) released during the process of cell pyrolysis caused by the recombinant protein. Observe the effect of recombinant protein on the sensitization and presentation of DC.
动物水平实验Animal level experiment
构建4T1乳腺癌皮下瘤原位模型,当肿瘤体积达到约40mm 3时,使用重组焦亡蛋白进行治疗,进行药效学研究,并在实验终点的时进行摘眼球取血(用于肝功能和肾功能检测),然后将小鼠进行安乐死,解剖取出老鼠的肿瘤以及脏器(心、肝、脾、肺、肾)。对它们进行称重并拍照,一部分肿瘤浸泡于4%多聚甲醛中固定用于后续的切片实验和生物安全性(病理切片等)检测实验;另一部分用于研究脾脏、淋巴结、肿瘤组织中体内T细胞及分泌的因子的变化情况,并检测肿瘤组织巨噬细胞、NK细胞的变化情况。采用WB检测给药治疗后肿瘤组织中的legumain、MR、TGF-β、TNF-α变化,使用ELISA试剂盒检测给药治疗后各组肿瘤组织中的细胞因子(TGF-β、TNF-α)的变化情况。 Construct 4T1 breast cancer subcutaneous tumor in situ model, when the tumor volume reaches about 40mm 3 , use recombinant pyrooptosis protein for treatment, conduct pharmacodynamic research, and perform eyeball blood collection at the end of the experiment (for liver function and Renal function test), the mice were then euthanized, and the tumors and organs (heart, liver, spleen, lung, kidney) of the mice were dissected. They were weighed and photographed. A part of the tumor was immersed in 4% paraformaldehyde and fixed for subsequent slice experiments and biosafety (pathological slices, etc.) testing experiments; the other part was used to study the spleen, lymph nodes, and tumor tissues in vivo The changes of T cells and secreted factors, and the changes of tumor tissue macrophages and NK cells. WB was used to detect the changes of legumain, MR, TGF-β, and TNF-α in tumor tissues after treatment, and ELISA kits were used to detect cytokines (TGF-β, TNF-α) in tumor tissues of each group after treatment. The change situation.
实施例1:FPLC的纯化结果Example 1: Purification results of FPLC
将经过镍柱初步纯化的蛋白(GSDMA3、GSDMA3-PTN和GSDMA3-TAT-PTN),依次经过脱盐柱(HiTrap Desalting)(图1A-C)和凝胶排阻层析柱(Superdex 75)(图1D-F)进行纯化。三个蛋白的峰形和出峰位置基本是一致的,这初步表明本实验通过基因工程改造的蛋白并没有影响它的结构和功能。The proteins (GSDMA3, GSDMA3-PTN and GSDMA3-TAT-PTN) that have been preliminarily purified by a nickel column are passed through a desalting column (HiTrap Desalting) (Figure 1A-C) and a gel exclusion chromatography column (Superdex 75) (Figure 1D-F) Purification. The peak shapes and peak positions of the three proteins are basically the same, which preliminarily shows that the protein modified by genetic engineering in this experiment did not affect its structure and function.
实施例2:融合蛋白的纯化鉴定Example 2: Purification and identification of fusion protein
将Superdex 75的前三个吸收峰的蛋白样品制样,进行SDS-PAGE电泳进行验证,结果如图2所示。泳道1-9分别是GSDMA3-TAT-PTN、GSDMA3-PTN和GSDMA3的三个吸收峰的蛋白。GSDMA3的条带符合理论分子量约为55kDa(泳道7);GSDMA3-PTN的条带(泳道4)基本和GSDMA3的条带基本保持同一水平上,这表明两者分子量接近和理论一致;重组蛋白GSDMA3-TAT-PTN条带(泳道1)有明显上移;而其他泳道都存在着杂带,这表明:第一个吸收峰的蛋白纯度较好,而其他两个吸收峰都存在着大量杂蛋白,所以后续实验使用的都是第一个吸收峰的蛋白。The protein samples of the first three absorption peaks of Superdex 75 were prepared and verified by SDS-PAGE electrophoresis. The results are shown in Figure 2. Lanes 1-9 are the proteins of the three absorption peaks of GSDMA3-TAT-PTN, GSDMA3-PTN and GSDMA3, respectively. The band of GSDMA3 conforms to the theoretical molecular weight of about 55kDa (lane 7); the band of GSDMA3-PTN (lane 4) basically maintains the same level as the band of GSDMA3, which shows that the molecular weight of the two is close to and theoretically consistent; the recombinant protein GSDMA3 -The TAT-PTN band (lane 1) has a significant upward shift; while the other lanes have mixed bands, which indicates that the protein purity of the first absorption peak is better, while the other two absorption peaks have a large amount of mixed protein , So all subsequent experiments use the protein with the first absorption peak.
实施例3:WB检测细胞Legumain的表达水平Example 3: WB detects the expression level of Legumain in cells
利用Western Blot检测诱导成M2型巨噬细胞的RAW 264.7以及4T1细胞的Legumain表达水平。如图3所示,Legumain在肿瘤相关的巨噬细胞中高表达,本 研究在体外培养4T1细胞发现确实低表达Legumain酶。Western Blot was used to detect the expression level of Legumain in RAW 264.7 and 4T1 cells induced into M2 macrophages. As shown in Figure 3, Legumain is highly expressed in tumor-associated macrophages. In this study, 4T1 cells were cultured in vitro and it was found that Legumain was indeed low in expression.
实施例4:融合焦亡蛋白体外Legumain酶切实验Example 4: In vitro Legumain cleavage experiment of the fusion pyrolysis protein
图4依次展示的是GSDMA3-TAT-PTN、GSDMA3-PTN和GSDMA3被酶切的结果。底物肽PTN被Legumain酶切开后,释放重组焦亡蛋白的N-domain和C-domain,GSDMA3-PTN和GSDMA3-TAT-PTN蛋白产生的片段大小近似,所以条带位置接近,结果显示重组蛋白能被Legumain酶切割。Figure 4 shows the results of digestion of GSDMA3-TAT-PTN, GSDMA3-PTN and GSDMA3 in turn. After the substrate peptide PTN is cleaved by Legumain, the N-domain and C-domain of the recombinant pyrolysis protein are released. The fragments produced by GSDMA3-PTN and GSDMA3-TAT-PTN proteins are similar in size, so the band positions are close, and the results show that the recombination The protein can be cleaved by Legumain enzyme.
实施例5:细胞摄取Example 5: Cellular uptake
将标记了FITC的重组蛋白事先经Legumain酶切割激活后再进行4T1细胞的摄取实验。实验结果如图5所示,酶切后的GSDMA3-TAT-PTN的摄取效果最佳。其中GSDMA3、酶切后的GSDMA3-TAT-PTN和GSDMA3-PTN的平均荧光强度值分别约为51.8、109和67.4,GSDMA3-TAT-PTN的摄取效率是GSDMA3-PTN的1.6倍,是GSDMA3的2.1倍。这表明GSDMA3-TAT-PTN经过Legumain酶切割激活后产生的活性片段,在穿膜肽TAT作用下具有较高的入胞效率。The FITC-labeled recombinant protein was cleaved and activated by Legumain enzyme in advance, and then the uptake experiment of 4T1 cells was performed. The experimental results are shown in Figure 5, the digested GSDMA3-TAT-PTN has the best ingestion effect. The average fluorescence intensity values of GSDMA3, digested GSDMA3-TAT-PTN and GSDMA3-PTN are about 51.8, 109 and 67.4 respectively. The uptake efficiency of GSDMA3-TAT-PTN is 1.6 times that of GSDMA3-PTN and 2.1 that of GSDMA3. Times. This indicates that the active fragment produced by GSDMA3-TAT-PTN after being activated by Legumain enzyme cleavage has a higher cell entry efficiency under the action of the penetrating peptide TAT.
实施例6:细胞毒性实验Example 6: Cytotoxicity test
采用4T1和DC2.4细胞检测焦亡蛋白的毒性。对于4T1细胞:在实验浓度范围内随着蛋白浓度的增加,细胞的存活率下降,其中酶切后的重组蛋白GSDMA3-TAT-PTN杀伤4T1肿瘤细胞的效果明显好于其他两组(图6A)。所有的焦亡蛋白在测试浓度范围内对于DC2.4细胞基本没有杀伤作用(图6B),具有较好的生物相容性,所以焦亡蛋白不会影响抗原提呈细胞的功能。4T1 and DC2.4 cells were used to detect the toxicity of pyroptin. For 4T1 cells: with the increase of protein concentration in the experimental concentration range, the survival rate of the cells decreased. Among them, the recombinant protein GSDMA3-TAT-PTN after restriction digestion was significantly better than the other two groups in killing 4T1 tumor cells (Figure 6A) . All of the pyrolyzed proteins had no killing effect on DC2.4 cells within the tested concentration range (Figure 6B), and had good biocompatibility, so the pyrolyzed proteins would not affect the function of antigen presenting cells.
实施例7:明场显微镜观察(Bright field imaging)Example 7: Bright field imaging
使用三组焦亡蛋白处理4T1细胞48h后如图7所示:正常的4T1细胞会成片生长成网状(图7A);GSDMA3和酶切后的GSDMA3-PTN处理细胞后,形态发生变化,也有大量的细胞正在吸水肿胀(图7B和7C),这与它们的细胞杀伤效果是和摄取实验结果相一致。酶切后的GSDMA3-TAT-PTN处理细胞后大量的细胞已经死亡,在清洗的过程中已经被洗去,剩余被固定的细胞中也可以看到大量吸水胀破的细胞,基本没有完整的细胞存在(图7D)。4T1 cells were treated with three groups of pyroptin proteins for 48h, as shown in Figure 7: normal 4T1 cells would grow into a network in sheets (Figure 7A); after treatment with GSDMA3 and digested GSDMA3-PTN, the morphology of the cells changed. There are also a large number of cells that are absorbing water and swelling (Figures 7B and 7C), which is consistent with their cell killing effect and the results of the uptake experiment. After the digested GSDMA3-TAT-PTN treats the cells, a large number of cells have died, and have been washed away during the washing process. In the remaining fixed cells, a large number of swollen cells can also be seen, and there are basically no complete cells. Exist (Figure 7D).
实施例8:钙网蛋白(CRT)测定实验Example 8: Calreticulin (CRT) measurement experiment
如图8显示的是胞外CRT量的变化,对照组肿瘤细胞会有左边阴性峰和右边阳性峰(图8A),用焦亡蛋白处理4T1细胞后,所有组的阳性峰均明显上升,其中酶切后的GSDMA3-TAT-PTN处理的细胞的阴性峰明显下降、阳性峰显著升高(图8B、8C和8D)。酶切后的GSDMA3-TAT-PTN和GSDMA3-PTN以及GSDMA3的平均荧光强度分别是:132、87和80.7,GSDMA3-TAT-PTN的强度分别是后两者的1.5倍和1.6倍。Figure 8 shows the change in the amount of extracellular CRT. The tumor cells in the control group will have a negative peak on the left and a positive peak on the right (Figure 8A). After 4T1 cells are treated with pyroptin, the positive peaks of all groups are significantly increased. After digestion, the negative peak of the cells treated with GSDMA3-TAT-PTN was significantly decreased, and the positive peak was significantly increased (Figures 8B, 8C and 8D). The average fluorescence intensities of GSDMA3-TAT-PTN, GSDMA3-PTN and GSDMA3 after digestion were 132, 87 and 80.7, respectively. The intensities of GSDMA3-TAT-PTN were 1.5 times and 1.6 times that of the latter two, respectively.
实施例9:HMGB1的迁移及胞外ATP释放实验Example 9: HMGB1 migration and extracellular ATP release experiment
如图9所示,GSDMA3和GSDMA3-PTN给药组,能提高HMGB1的释放量,但和PBS组没有显著性差异。而GSDMA3-TAT-PTN给药组则能大幅度提高HMGB1的释放量,它的HMGB1释放的浓度分别是GSDMA3组和GSDMA3-PTN组的1.9倍和2.4倍,具有显著性差异。所有实验组的胞外ATP含量都显著性增加,其中GSDMA3-TAT-PTN处理组的胞外ATP含量和其他处理组相比均有显著性差异,表明ATP从细胞内释放到细胞外,认为是通过焦亡蛋白形成的孔洞释放到胞外或者是焦亡激活的膜通道释放到胞外。As shown in Figure 9, the GSDMA3 and GSDMA3-PTN administration groups can increase the release of HMGB1, but there is no significant difference from the PBS group. The GSDMA3-TAT-PTN administration group can greatly increase the release of HMGB1, and the release concentration of HMGB1 is 1.9 times and 2.4 times that of the GSDMA3 group and the GSDMA3-PTN group, respectively, with significant differences. The extracellular ATP content of all experimental groups increased significantly. Among them, the extracellular ATP content of the GSDMA3-TAT-PTN treatment group was significantly different from the other treatment groups, indicating that ATP was released from the inside of the cell to the outside of the cell, which is considered to be The hole formed by the pyrolysis protein is released to the outside of the cell or the membrane channel activated by the pyrolysis is released to the outside of the cell.
实施例10:树突状细胞体外致敏实验Example 10: Dendritic cell sensitization experiment in vitro
如图10所示,焦亡蛋白处理BMDC后,CD80 +和CD86 +的DC细胞量基本保持上升趋势,但各组间CD80 +DC细胞量没有显著性差异(图10A),各蛋白处理组的CD86 +DC细胞量能达到阳性对照组的水平(图10B),而蛋白处理组CD80 +/CD86 +双阳性的DC数量也能达到阳性对照组的水平,其中GSDMA3-TAT-PTN处理过的DC细胞双阳量最高(图10C)。 As shown in Figure 10, after treating BMDC with pyrolysis protein, the amount of CD80 + and CD86 + DC cells basically maintained an upward trend, but there was no significant difference in the amount of CD80 + DC cells between the groups (Figure 10A). The number of CD86 + DC cells can reach the level of the positive control group (Figure 10B), and the number of CD80 + /CD86 + double-positive DCs in the protein treatment group can also reach the level of the positive control group. Among them, the DCs treated with GSDMA3-TAT-PTN The double positive amount of cells was the highest (Figure 10C).
实施例11:重组焦亡蛋白对抗原提呈的作用Example 11: The effect of recombinant pyroopterin on antigen presentation
如图11所示,和PBS组相比,所有的蛋白处理组均能提高MHC-I类分子的表达强度(图11A),其中GSDMA3和GSDMA3-TAT-PTN处理DC后的量约是PBS组的两倍,形成了显著性差异。GSDMA3处理组MHC-II类分子的量比PBS组略微下降,但没有显著性差异(图11B)。而GSDMA3-PTN和GSDMA3-TAT-PTN处理组均能显著增加MHC-II的量,其中GSDMA3-TAT-PTN处理组增强的效果最佳。As shown in Figure 11, compared with the PBS group, all protein treatment groups can increase the expression intensity of MHC-I molecules (Figure 11A). Among them, the amount of GSDMA3 and GSDMA3-TAT-PTN treated DC is about the same as that of the PBS group. Twice, forming a significant difference. The amount of MHC-II molecules in the GSDMA3 treatment group was slightly lower than that in the PBS group, but there was no significant difference (Figure 11B). The GSDMA3-PTN and GSDMA3-TAT-PTN treatment groups can significantly increase the amount of MHC-II, and the GSDMA3-TAT-PTN treatment group has the best enhancement effect.
实施例12:重组焦亡蛋白在皮下移植瘤模型上的药效学研究Example 12: Pharmacodynamic Study of Recombinant Pyroopterin on Subcutaneous Xenograft Tumor Model
药效学结果如图12所示:其中图12B为流程图。各治疗组的瘤体积均增加,但焦亡蛋白治疗组均能减缓增长趋势(图12A),在实验终点三组的抑瘤率分别能达到21%、34%和62%(图12E),其中GSDMA3-TAT-PTN治疗组和对照组形成显著差异。蛋白治疗期间小鼠体重变化不大(图12C),表明无明显毒副作用。图12D和12F是在实验终点(第31天)解剖出肿瘤后并称重拍照,发现焦亡蛋白治疗组的肿瘤都有所减小,其中GSDMA3-TAT-PTN组肿瘤最小,和其它组相比有显著性差异。The pharmacodynamic results are shown in Figure 12: Figure 12B is a flowchart. The tumor volume of each treatment group increased, but the pyrooptin treatment group could slow down the growth trend (Figure 12A), and the tumor inhibition rates of the three groups at the experimental end point could reach 21%, 34% and 62% respectively (Figure 12E). Among them, there is a significant difference between the GSDMA3-TAT-PTN treatment group and the control group. The body weight of the mice did not change much during the protein treatment (Figure 12C), indicating that there were no obvious side effects. Figures 12D and 12F are after the tumor was dissected at the end of the experiment (day 31) and weighed and photographed. It was found that the tumors in the pyroopterin treatment group were reduced, and the tumors in the GSDMA3-TAT-PTN group were the smallest, which was similar to other groups. There is a significant difference than that.
实施例13:给药治疗后体内T细胞及其分泌的细胞因子的变化Example 13: Changes of T cells and cytokines secreted by them in the body after treatment
13.1脾脏13.1 Spleen
图13(A-D)以PBS治疗组的小鼠脾脏细胞为例,显示整个流式圈门的示意图。图(13E-F)依次为GSDMA3、GSDMA3-PTN、GSDMA3-TAT-PTN治疗小鼠后脾脏中分泌的颗粒酶量的变化情况。肿瘤和淋巴结的颗粒酶及其干扰素的分泌实验结果也参照此部分进行分析。Figure 13 (A-D) takes the mouse spleen cells in the PBS treatment group as an example, showing a schematic diagram of the entire flow cytometry gate. Figures (13E-F) show the changes in the amount of granzyme secreted in the spleen of mice after GSDMA3, GSDMA3-PTN, and GSDMA3-TAT-PTN treatment. The results of the secretion of granzyme and its interferon in tumors and lymph nodes are also analyzed with reference to this section.
图14是脾脏中T细胞、颗粒酶及干扰素的统计学结果。给药治疗后各组CD4 +T细胞量都没有显著性变化(图14A和14B),GSDMA3-TAT-PTN治疗组CD8 +T细胞数最多并且分泌的颗粒酶和干扰素也是最多的(图14B、14C和14D),这表明本实验采用GSDMA3-TAT-PTN治疗荷瘤小鼠后能有效激活脾脏中的T细胞并释放大量细胞因子对肿瘤进行杀伤。 Figure 14 shows the statistical results of T cells, granzyme and interferon in the spleen. After treatment, the amount of CD4 + T cells in each group did not change significantly (Figure 14A and 14B). The GSDMA3-TAT-PTN treatment group had the largest number of CD8 + T cells and secreted the most granzyme and interferon (Figure 14B). , 14C and 14D), which indicates that the treatment of tumor-bearing mice with GSDMA3-TAT-PTN in this experiment can effectively activate T cells in the spleen and release a large amount of cytokines to kill the tumor.
13.2肿瘤13.2 Tumor
实验终点肿瘤组织中T细胞及其分泌的因子变化情况如图15所示,蛋白药物治疗后各组CD4 +T细胞和CD8 +T细胞都没有显著性变化(图15A和15B)。但GSDMA3-TAT-PTN治疗组的小鼠肿瘤CD8 +T细胞中颗粒酶B的表达量显著性上升,而另外几个治疗组则没有明显变化(图15C),这表明本研究构建的GSDMA3-TAT-PTN具有非常好的肿瘤杀伤作用。所有焦亡蛋白治疗过的肿瘤的CD8 +T细胞分泌的干扰素都有明显增加,均和PBS组形成显著性对比,其中GSDMA3-TAT-PTN分泌的量最多(图15D)。 The changes of T cells and their secreted factors in the tumor tissue at the end of the experiment are shown in Figure 15. After protein drug treatment, there were no significant changes in CD4 + T cells and CD8 + T cells in each group (Figures 15A and 15B). However, the expression of granzyme B in mouse tumor CD8 + T cells in the GSDMA3-TAT-PTN treatment group increased significantly, while there was no significant change in the other treatment groups (Figure 15C), which indicates that the GSDMA3-constructed in this study TAT-PTN has a very good tumor killing effect. The interferon secreted by CD8 + T cells of all tumors treated with pyroostatin was significantly increased, which formed a significant contrast with the PBS group, in which GSDMA3-TAT-PTN secreted the most amount (Figure 15D).
13.3淋巴结13.3 Lymph nodes
如图16展示的肿瘤周边部位腋下淋巴结中T细胞、颗粒酶及干扰素的统计学 结果。GSDMA3-TAT-PTN治疗组和其他组相比CD4 +T并没有显著性变化,但是GSDMA3和GSDMA3-PTN治疗组的CD4 +T细胞和PBS组相比显著性减少(图16A);而所有的蛋白治疗组的CD8 +T细胞和PBS组相比均有显著性增加(图16B)。所有焦亡蛋白治疗组的CD8 +T细胞中颗粒酶B和干扰素表达量都增加,其中GSDMA3-TAT-PTN治疗组的表达量显著性上升(图16C和16D)。 Figure 16 shows the statistical results of T cells, granzyme and interferon in the axillary lymph nodes around the tumor. GSDMA3-TAT-PTN treatment group and the other groups as compared to CD4 + T no significant change, but CD4 + T cells and the PBS group as compared to significantly reduce GSDMA3 and GSDMA3-PTN treatment groups (FIG. 16A); and all Compared with the PBS group, CD8 + T cells in the protein treatment group increased significantly (Figure 16B). The expression levels of granzyme B and interferon in CD8 + T cells of all pyrostatin treatment groups increased, and the expression levels of the GSDMA3-TAT-PTN treatment group increased significantly (Figure 16C and 16D).
实施例14:肿瘤中巨噬细胞和NK细胞的变化Example 14: Changes of macrophages and NK cells in tumors
14.1巨噬细胞14.1 Macrophages
如图17A和17B所示:所有蛋白治疗组的M1型巨噬细胞均有所增加,其中GSDMA3-TAT-PTN组肿瘤的M1型巨噬细胞显著性提高。所有蛋白治疗组和PBS组相比均下调M2型巨噬细胞(图17C和17D),其中GSDMA3-PTN和GSDMA3-TAT-PTN治疗组的M2型巨噬细胞显著性下降。As shown in Figures 17A and 17B: M1 type macrophages of all protein treatment groups increased, and M1 type macrophages of tumors in the GSDMA3-TAT-PTN group increased significantly. Compared with the PBS group, all protein treatment groups down-regulated M2 type macrophages (Figure 17C and 17D), and the M2 type macrophages of the GSDMA3-PTN and GSDMA3-TAT-PTN treatment groups decreased significantly.
14.2 NK细胞14.2 NK cells
图17(E-F)表明:GSDMA3-TAT-PTN治疗组的肿瘤组织中NK细胞显著增加,而其他几组则没有明显变化。Figure 17 (E-F) shows that NK cells in the tumor tissue of the GSDMA3-TAT-PTN treatment group increased significantly, while there was no significant change in the other groups.
实施例15:各组肿瘤组织中的legumain、MR、TGF-β、TNF-α的变化情况Example 15: Changes of legumain, MR, TGF-β, and TNF-α in tumor tissues of each group
如图18所示,使用WB结果分析实验终点时肿瘤组织中相关蛋白的表达情况,GSDMA3-TAT-PTN治疗后瘤内促炎因子(TNF-α)显著上升,同时利于肿瘤增殖转移的相关蛋白(MR、Legumain)、因子(TGF-β)的表达水平均显著下降。这表明本研究构建的重组蛋白有非常好的抗肿瘤效果。As shown in Figure 18, using the WB results to analyze the expression of related proteins in tumor tissues at the end of the experiment, the intratumoral pro-inflammatory factor (TNF-α) increased significantly after GSDMA3-TAT-PTN treatment, and at the same time, related proteins that are conducive to tumor proliferation and metastasis The expression levels of (MR, Legumain) and factor (TGF-β) decreased significantly. This indicates that the recombinant protein constructed in this study has a very good anti-tumor effect.
实施例16:肿瘤组织中细胞因子的变化Example 16: Changes in cytokines in tumor tissues
使用ELISA试剂盒检实验终点时各组肿瘤组织中的细胞因子的分泌情况,如图19所示。发现各治疗组促癌细胞细胞因子(TGF-β)分泌下降,结合图18的结果分析:GSDMA3-TAT-PTN治疗组它能增强机体免疫和细胞杀伤并减低肿瘤的转移,有着良好的抗肿瘤效果。The ELISA kit was used to check the secretion of cytokines in the tumor tissues of each group at the end of the experiment, as shown in Figure 19. It was found that the secretion of cancer cell cytokines (TGF-β) decreased in each treatment group, combined with the result analysis in Figure 18: GSDMA3-TAT-PTN treatment group can enhance the body’s immunity and cell killing and reduce tumor metastasis, and has a good anti-tumor effect. effect.
实施例17:生物安全性评价Example 17: Biosafety Evaluation
17.1脏器系数17.1 Organ coefficient
将实验终点的老鼠的主要脏器(心、肝、肺、肾、脾)取出称重,如图20所示,荷载有4T1乳腺癌的小鼠有明显的脾脏肿大的现象,而GSDMA3-TAT-PTN治疗组在治疗终点时小鼠的脾脏较PBS组小,各组的其他脏器并没有明显区别。The main organs (heart, liver, lung, kidney, spleen) of the mice at the end of the experiment were taken out and weighed. As shown in Figure 20, mice bearing 4T1 breast cancer had obvious splenomegaly, while GSDMA3- The spleen of the mice in the TAT-PTN treatment group was smaller than that of the PBS group at the end of treatment, and there was no significant difference in other organs in each group.
17.2病理切片17.2 Pathological Sections
将实验终点的老鼠的主要脏器(心脏、肝脏、脾脏、肺、肾脏)进行病理分析,如图21所示。发现GSDMA3治疗组肺泡变形,其他组肺泡形状正常,无明显细胞脱落;而各组其他脏器均无明显病变,表明重组蛋白的生物安全较好,对脏器无明显损伤。The main organs (heart, liver, spleen, lung, and kidney) of the mice at the end of the experiment were subjected to pathological analysis, as shown in Figure 21. It was found that the alveoli in the GSDMA3 treatment group were deformed, and the alveoli in the other groups were normal in shape without obvious cell shedding; while other organs in each group had no obvious lesions, indicating that the recombinant protein has good biological safety and no obvious damage to the organs.
17.3生化指标17.3 Biochemical indicators
在实验终点取血检测肝功能和肾功能,结果如图22所示:GSDMA3-TAT-PTN治疗组的AST和另外两组相比显著性下降,但和PBS组相比并无显著性差异,结合AST在急慢性肝炎和中毒性肝炎中的表达会增加的情况来分析,GSDMA3-TAT-PTN治疗后并不会导致肝损伤。各组的其他指标均无显著性变化,这表明使用焦亡蛋白治疗后并没有引起肝脏和肾脏的损伤和炎症。Blood was taken at the end of the experiment to detect liver and kidney functions. The results are shown in Figure 22: AST in the GSDMA3-TAT-PTN treatment group was significantly lower than the other two groups, but there was no significant difference compared with the PBS group. Based on the analysis of the increased expression of AST in acute, chronic hepatitis and toxic hepatitis, GSDMA3-TAT-PTN treatment does not cause liver damage. There were no significant changes in other indicators in each group, which indicated that the use of pyroapone protein treatment did not cause liver and kidney damage and inflammation.
讨论:discuss:
焦亡蛋白在未被切割激活的情况下不显毒性作用,一旦被切割激活将释放毒性末端,导致细胞膜穿孔、内含物释放、引起炎性反应,从而介导细胞死亡即焦亡,所以它是一类具有前景的生物大分子前药,但它是胞内作用蛋白,必须要克服细胞膜屏障进入胞浆内发挥作用,而且焦亡蛋白的作用缺乏细胞特异性,易造成毒副作用。The pyrolysis protein does not have a toxic effect when it is not activated by cutting. Once it is activated by cutting, it will release the toxic end, leading to perforation of the cell membrane, release of the contents, and inflammatory reaction, thereby mediating cell death, that is, pyrolysis. It is a class of promising biological macromolecular prodrugs, but it is an intracellular protein that must overcome the cell membrane barrier to enter the cytoplasm to play a role. In addition, the role of pyrolysis protein lacks cell specificity and is likely to cause toxic side effects.
本发明人在设计重组焦亡蛋白递药系统时,考虑到肿瘤微环境(TME)内Legumain高表达的特征,选择了靶向TME激活的递药策略,通过基因工程重组技术,制备具有TME响应激活功能的焦亡蛋白,以增强肿瘤部位蛋白的作用并降低正常组织的毒副作用。When designing a recombinant pyroptosis protein delivery system, the inventors took into account the high expression of Legumain in the tumor microenvironment (TME), and chose a drug delivery strategy targeting TME activation. Through genetic engineering recombination technology, a TME-responsive drug delivery strategy was prepared. Activate functional pyro-apoptosis protein to enhance the effect of tumor site protein and reduce the toxic side effects of normal tissues.
本发明利用TME中特异性高表达的蛋白酶Legumain酶,通过在焦亡蛋白两个结构域之间引入Legumain底物肽序列PTN来实现肿瘤酶激活的焦亡蛋白介导抗肿瘤作用。The invention utilizes the specific and highly expressed protease Legumain enzyme in TME, and realizes the anti-tumor effect of the pyrostat protein mediated by tumor enzyme activation by introducing the Legumain substrate peptide sequence PTN between the two structural domains of the pyrostat protein.
在本发明中,选用了焦亡蛋白家族的GSDMA3进行研究,它的N-domain是 活性结构域,能够引起细胞焦亡。本研究采用基因工程技术将PTN插入至GSDMA3的C-domain和N-domain之间的linker序列,使它能在TME中被切割,释放出活性N-domain。但蛋白本身的入胞效率不高,而N-domain需要进入细胞内发挥作用,于是本研究在N-domain与PTN之间引入了一段穿膜肽序列,通过穿膜肽促进入胞发挥作用。入胞后的N-domain在细胞膜上形成孔洞,导致细胞死亡,同时胞内ATP、HMGB1、LDH、IL-1β、钙网蛋白等大量释放到胞外,激活免疫细胞,抑制肿瘤的增殖。In the present invention, GSDMA3 of the pyrodepressin family is selected for research, and its N-domain is the active domain, which can cause cell pyrolysis. In this study, genetic engineering technology was used to insert PTN into the linker sequence between the C-domain and N-domain of GSDMA3, so that it can be cleaved in TME to release the active N-domain. However, the efficiency of protein itself is not high, and the N-domain needs to enter the cell to play a role. Therefore, this study introduced a penetrating peptide sequence between the N-domain and PTN to promote the penetration of the cell through the penetrating peptide. After entering the cell, the N-domain forms a hole in the cell membrane, leading to cell death. At the same time, a large amount of intracellular ATP, HMGB1, LDH, IL-1β, calreticulin, etc. are released outside the cell to activate immune cells and inhibit tumor proliferation.
本发明提供的GSDMA3-TAT-PTN蛋白在体内外实验中表现出非常好的抗肿瘤活性,这表明利用肿瘤酶激活及融合穿膜肽的焦亡蛋白递药策略这一设计的有效性。The GSDMA3-TAT-PTN protein provided by the present invention exhibits very good anti-tumor activity in in vivo and in vitro experiments, which shows the effectiveness of the design of the pyrostat protein delivery strategy using tumor enzyme activation and fusion of penetrating peptides.
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 invention are cited as references in this application, as if each document was individually cited as a reference. In addition, it should be understood that after reading the above teaching content of the present invention, those skilled in the art can make various changes or modifications to the present invention, and these equivalent forms also fall within the scope defined by the appended claims of the present application.

Claims (10)

  1. 一种融合蛋白,其特征在于,所述融合蛋白从N端到C端具有式I所示的结构:A fusion protein, characterized in that the fusion protein has a structure shown in formula I from N-terminus to C-terminus:
    Z0-Z1-Z2-Z3-Z4  (式I)Z0-Z1-Z2-Z3-Z4 (Formula I)
    式中,Where
    Z0为任选的标签元件;Z0 is an optional label element;
    Z1为焦亡蛋白的N结构域元件;Z1 is the N domain element of pyrolysis protein;
    Z2为穿膜肽序列元件;Z2 is a penetrating peptide sequence element;
    Z3为能够被肿瘤微环境中特异性表达的蛋白酶特异性切割的肽序列元件;Z3 is a peptide sequence element that can be specifically cleaved by a protease specifically expressed in the tumor microenvironment;
    Z4为焦亡蛋白的C结构域元件;Z4 is the C domain element of pyroopterin;
    “-”表示连接上述元件的肽键;"-" indicates a peptide bond connecting the above-mentioned elements;
    其中,在融合蛋白中,所述Z4元件通过特异性地结合Z1元件来抑制Z1元件的活性。Wherein, in the fusion protein, the Z4 element inhibits the activity of the Z1 element by specifically binding to the Z1 element.
  2. 如权利要求1所述的融合蛋白,其特征在于,所述焦亡蛋白具有诱导细胞膜破裂并释放大量内含物引起机体炎症反应的功能。The fusion protein according to claim 1, wherein the pyroopterin protein has the function of inducing the rupture of the cell membrane and releasing a large amount of content to cause the body's inflammatory response.
  3. 如权利要求1所述的融合蛋白,其特征在于,所述焦亡蛋白为GSDMA3。The fusion protein according to claim 1, wherein the pyrolysis protein is GSDMA3.
  4. 如权利要求1所述的融合蛋白,其特征在于,所述融合蛋白的氨基酸序列如SEQ ID NO:4所示。The fusion protein of claim 1, wherein the amino acid sequence of the fusion protein is shown in SEQ ID NO: 4.
  5. 一种分离的多核苷酸,其特征在于,所述多核苷酸编码如权利要求1所述的融合蛋白。An isolated polynucleotide, characterized in that the polynucleotide encodes the fusion protein of claim 1.
  6. 一种载体,其特征在于,所述载体中含有如权利要求5所述的多核苷酸。A vector, characterized in that it contains the polynucleotide according to claim 5.
  7. 一种宿主细胞,其特征在于,所述宿主细胞中含有如权利要求6所述的载体,或基因组中整合有如权利要求5所述的多核苷酸。A host cell, characterized in that the host cell contains the vector according to claim 6, or the polynucleotide according to claim 5 is integrated into the genome.
  8. 一种生产如权利要求1所述的融合蛋白的方法,其特征在于,包括步骤:A method for producing the fusion protein according to claim 1, characterized in that it comprises the steps of:
    在适合表达的条件下,培养如权利要求7所述的宿主细胞,从而表达出如权利要求1所述的融合蛋白。Under conditions suitable for expression, the host cell according to claim 7 is cultured to express the fusion protein according to claim 1.
  9. 一种药物组合物,其特征在于,包括:A pharmaceutical composition, characterized in that it comprises:
    (a)如权利要求1所述的融合蛋白或其编码基因;(a) The fusion protein of claim 1 or its encoding gene;
    (b)药学上可接受的载体。(b) A pharmaceutically acceptable carrier.
  10. 一种如权利要求1所述的融合蛋白、如权利要求5所述的多核苷酸、如权利要求6所述的载体和如权利要求7所述的宿主细胞的用途,其特征在于,用于制备一制剂或药物组合物,所述制剂或药物组合物用于选自下组的一种或多种:A use of the fusion protein according to claim 1, the polynucleotide according to claim 5, the vector according to claim 6 and the host cell according to claim 7, characterized in that it is used for A preparation or pharmaceutical composition is prepared, and the preparation or pharmaceutical composition is used for one or more selected from the following group:
    (a)杀死肿瘤微环境中的肿瘤细胞;(a) Kill tumor cells in the tumor microenvironment;
    (b)提高肿瘤微环境中的M1型巨噬细胞的数量,并降低肿瘤微环境中M2型巨噬细胞的数量;(b) Increase the number of M1 macrophages in the tumor microenvironment, and reduce the number of M2 macrophages in the tumor microenvironment;
    (c)提高肿瘤微环境中的抗癌细胞因子、抗原提呈分子、效应T细胞、免疫原性细胞死亡(ICD)相关特征分子(如ATP、HMGB1、CRT)、LDH等促炎因子的水平;(c) Increase the level of anti-cancer factors, antigen presenting molecules, effector T cells, immunogenic cell death (ICD) related characteristic molecules (such as ATP, HMGB1, CRT), LDH and other pro-inflammatory factors in the tumor microenvironment ;
    (d)降低肿瘤增殖转移相关蛋白、促癌细胞因子的表达水平。(d) Reduce the expression level of tumor proliferation and metastasis-related proteins and cancer cell factors.
PCT/CN2020/111821 2019-08-28 2020-08-27 Tumor enzyme-responsive recombinant pyroptosis protein delivery system and antitumor use thereof WO2021037160A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910803462.X 2019-08-28
CN201910803462.XA CN112442129B (en) 2019-08-28 2019-08-28 Tumor enzyme response type recombinant pyroprotein drug delivery system and anti-tumor application thereof

Publications (1)

Publication Number Publication Date
WO2021037160A1 true WO2021037160A1 (en) 2021-03-04

Family

ID=74683588

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/111821 WO2021037160A1 (en) 2019-08-28 2020-08-27 Tumor enzyme-responsive recombinant pyroptosis protein delivery system and antitumor use thereof

Country Status (2)

Country Link
CN (1) CN112442129B (en)
WO (1) WO2021037160A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115838435A (en) * 2022-10-28 2023-03-24 中国人民解放军空军军医大学 Cell apoptosis-related molecule recombinant immune coupling protein and preparation method and application thereof

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113337569A (en) * 2021-05-26 2021-09-03 深圳市人民医院 Method for screening antitumor drugs based on induction of cell inflammatory death
WO2023108803A1 (en) * 2021-12-13 2023-06-22 中山大学肿瘤防治中心(中山大学附属肿瘤医院、中山大学肿瘤研究所) Fusion protein, and preparation method therefor and use thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104707150A (en) * 2013-12-11 2015-06-17 中国科学院上海药物研究所 Tumor enzyme activated quantum dot probe, preparation method and application thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006019153A1 (en) * 2004-08-20 2006-02-23 Inter University Research Institution Corporation, Research Organization Of Information And Systems NONHUMAN ANIMAL WITH INHIBITED GdsmA GENE FUNCTION AND PROMOTED OR INHIBITIED CANCERATION-ASSOCIATED GENE FUNCTION

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104707150A (en) * 2013-12-11 2015-06-17 中国科学院上海药物研究所 Tumor enzyme activated quantum dot probe, preparation method and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE PROTEIN 15 February 2019 (2019-02-15), ANONYMOUS: "gasdermin-A3 [Mus musculus]", XP055786904, retrieved from GENBANK Database accession no. NP_001007462 *
JINGJIN DING ET AL.: "Pore-forming activity and structural autoinhibition of the gasdermin family", NATURE, vol. 535, no. 7610, 7 July 2016 (2016-07-07), pages 111 - 116, XP055786908, ISSN: 1476-4687 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115838435A (en) * 2022-10-28 2023-03-24 中国人民解放军空军军医大学 Cell apoptosis-related molecule recombinant immune coupling protein and preparation method and application thereof
CN115838435B (en) * 2022-10-28 2023-10-03 中国人民解放军空军军医大学 Cell scorch related molecule recombinant immunoconjugate protein, and preparation method and application thereof

Also Published As

Publication number Publication date
CN112442129A (en) 2021-03-05
CN112442129B (en) 2023-05-12

Similar Documents

Publication Publication Date Title
WO2021037160A1 (en) Tumor enzyme-responsive recombinant pyroptosis protein delivery system and antitumor use thereof
CN104507489B (en) For preventing and treat the compositions of rheumatoid arthritis
EP1417228B1 (en) Peptides effective in the treatment of tumors and other conditions requiring the removal or destruction of cells
EP1994152B1 (en) Peptides effective in the treatment of tumors and other conditions requiring the removal or destruction of cells
EP3360893B1 (en) High-affinity and soluble pdl-1 molecule
BR112014016076B1 (en) Peptide, vector, chimeric peptide construct and pharmaceutical composition
TW200835513A (en) HLA-A* 1101-restricted WT1 peptide and pharmaceutical composition comprising the same
JP5966000B2 (en) Tumor targeting tumor necrosis factor-related apoptosis-inducing ligand mutant and its application
TW200821386A (en) Human GLP-1 mimetibodies, compositions, methods and uses
TWI395948B (en) A novel physiologically active compound, nesfatin, related compounds thereof and their use
CN107530400A (en) The method for the illness for destroying or removing cell is needed using the peptide treatment from NF-M
AU2005298245B2 (en) A thymus-specific protein
CN101400364A (en) Method of preventing or reducing the risk or incidence of cancer using neural thread protein based peptides
TW518340B (en) Tumor antigen peptide derivative
EP2589605B1 (en) Telomerase activity inhibiting peptide and manufacturing method and application thereof
WO2016179861A1 (en) Slit2d2-hsa fusion protein and use thereof against tumours
WO2019101110A1 (en) Use of trichosanthin for sensitizing and/or activating dendritic cells
WO2015055148A1 (en) Yap protein inhibiting polypeptide and application thereof
NZ547274A (en) A recombinant protein with cancer suppression action, its encoding gene and use
US9943567B2 (en) Method for treating arthritis using IK factor or nucleic acid encoding IK factor
KR102348838B1 (en) Active TRAIL trimer and tumor targeting peptide multi-displayed on ferritin nanocage and use in anti-cancer agent thereof
CN111909246B (en) AAV mutants highly efficient in infecting supporting cells
TW519549B (en) Tumor antigen protein, gene thereof, and utilization thereof
JP4544715B2 (en) Modified cDNA and modified protein of rat bcl-x gene
TWI419901B (en) Compositions and methods of using crmp-1 and its fragments for treating cancer

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: 20857186

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: 20857186

Country of ref document: EP

Kind code of ref document: A1