WO2019196790A1 - Anti-tumor fusion protein, preparation method therefor and application thereof - Google Patents

Anti-tumor fusion protein, preparation method therefor and application thereof Download PDF

Info

Publication number
WO2019196790A1
WO2019196790A1 PCT/CN2019/081735 CN2019081735W WO2019196790A1 WO 2019196790 A1 WO2019196790 A1 WO 2019196790A1 CN 2019081735 W CN2019081735 W CN 2019081735W WO 2019196790 A1 WO2019196790 A1 WO 2019196790A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
fusion protein
gnrh
tumor
cell
Prior art date
Application number
PCT/CN2019/081735
Other languages
French (fr)
Chinese (zh)
Inventor
颜浩为
侯天全
Original Assignee
颜浩为
侯天全
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 颜浩为, 侯天全 filed Critical 颜浩为
Priority to CN201980020574.6A priority Critical patent/CN112292402B/en
Publication of WO2019196790A1 publication Critical patent/WO2019196790A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • A61K38/1758Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals p53
    • 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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides 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
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4746Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used p53
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/23Luteinising hormone-releasing hormone [LHRH]; Related peptides
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention relates to the field of biomedicine, and more particularly to an anti-tumor fusion protein and a preparation method and application thereof.
  • An immunotoxin is a group of artificially constructed hybrid molecules with specific cell killing ability, consisting of a toxic part, a carrier part and a targeting part.
  • the toxic portion may be a cytotoxin of plant, animal, or microbial origin, and the targeting moiety may be a monoclonal antibody or a cytokine.
  • immunotoxins Compared with other anti-tumor drugs, immunotoxins have the advantages of strong toxicity and high specificity, and have shown great application prospects in tumor treatment.
  • Lyophilized recombinant human luteinizing hormone releasing hormone-exotoxin of pseudomonas aeruginosa fusion protein (LHRH-PE40) is a pair of research and production of Changchun genetic engineering drugs Tumor cells have a specific killing effect of recombinant toxins.
  • the production method comprises the following steps: firstly, the receptor binding region (I region) encoding the Pseudomonas aeruginosa exotoxin A gene is excised by a genetic engineering method, and replaced with the LHRH gene, and then the LHRH-PE40 recombinant gene is cloned into a plasmid and passed through engineering fermentation. The LHRH-PE40 fusion protein was expressed.
  • LHRH-PE40 The cytotoxic effect of LHRH-PE40 is exotoxin A of pseudomonas aeruginosa (PEA).
  • Pseudomonas aeruginosa exotoxin A is the most potent toxin of Pseudomonas aeruginosa, which can block the synthesis of cellular proteins and cause apoptosis of cells.
  • the leader in LHRH-PE40 is human luteinizing hormone releasing hormone (LHRH), which is targeted to introduce PEA into tumor cells to exert anti-tumor by binding to its type I receptor on the surface of tumor cells. effect.
  • LHRH human luteinizing hormone releasing hormone
  • LHRH has a high affinity for type I receptors.
  • type I receptors mainly exist in the anterior pituitary, and tissues such as gonads, placenta and brain tissues also contain a certain amount of type I receptors, and other important organs do not express type I LHRH receptors.
  • LHRH type I receptors are distributed due to receptor alienation, including reproductive system malignant tumors, melanoma, gastric cancer, liver cancer, pancreatic cancer, intestinal cancer, and lung cancer. Since type I LHRH receptors are generally highly expressed in tumor cells and are only localized in normal tissues, LHRH is an ideal guide for designing recombinant targeting toxins.
  • Type II receptors of LHRH are widely present in various tissues of the human body. LHRH has a low affinity for type II receptors. However, at very high doses, LHRH can bind to the type II receptor with low affinity, resulting in extensive cytotoxicity of LHRH-PE40. This is the theoretical basis for the toxic effects of LHRH-PE40 recombinant toxins on animals. LHRH-PE40 does not penetrate the blood-brain barrier and therefore does not cause toxic effects on the pituitary gland.
  • the object of the present invention is to provide an anti-tumor fusion protein, a preparation method and application thereof.
  • an anti-tumor fusion protein having the structure of formula I:
  • A is a GnRH protein element
  • B is the transmembrane transport region of the PEA protein or is absent
  • C is a P53 protein element
  • E is a TAT protein element or is absent
  • D is an optional signal peptide and/or leader peptide sequence
  • B and E are not the same at the same time
  • the anti-tumor fusion protein has the structure of Formula II:
  • A is a GnRH protein element
  • C is a P53 protein element
  • E is a TAT protein element
  • D is an optional signal peptide and/or leader peptide sequence
  • the anti-tumor fusion protein in another preferred embodiment, is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-tumor fusion protein
  • the TAT is a short peptide rich in basic amino acids encoded by HIV-I.
  • the TAT protein is derived from a human or a non-human mammal.
  • the TAT protein comprises wild-type and mutant forms.
  • the TAT protein comprises a full length, mature form of P53, or an active fragment thereof.
  • sequence of the TAT protein is shown in positions 405-416 of SEQ ID NO.: 5.
  • the anti-tumor fusion protein is selected from the group consisting of:
  • (C) A derivative polypeptide obtained by subjecting the amino acid sequence shown by SEQ ID NO: 5 to substitution, deletion or addition of 1-10 amino acid residues, and retaining the activity of inhibiting tumor cell growth.
  • the anti-tumor fusion protein has the structure of Formula II:
  • A is a GnRH protein element
  • B is a transmembrane transport region of PEA protein
  • C is a P53 protein element
  • D is an optional signal peptide and/or leader peptide sequence
  • the GnRH protein is derived from a human or a non-human mammal.
  • the GnRH protein comprises wild type and mutant form.
  • the GnRH protein comprises a full length, mature form of GnRH, or an active fragment thereof.
  • the GnRH protein comprises a type II GnRH protein and a type I GnRH protein.
  • the GnRH protein is a type II GnRH protein.
  • sequence of the GnRH protein is as shown in 1-10 of SEQ ID NO.: 2.
  • the P53 protein is derived from a human or a non-human mammal.
  • the P53 protein comprises wild type and mutant form.
  • the P53 protein comprises a full length, mature form of P53, or an active fragment thereof.
  • sequence of the P53 protein is shown in positions 128-520 of SEQ ID NO.: 2.
  • the transmembrane transport region of the PEA protein is derived from Pseudomonas aeruginosa.
  • the transmembrane transport region of the PEA protein is from 100 to 120 amino acids in length, preferably from 100 to 115 amino acids.
  • sequence of the transmembrane trafficking region of the PEA protein is shown in positions 13-127 of SEQ ID NO.: 2.
  • the anti-tumor fusion protein is a recombinant protein expressed by bacteria, preferably E. coli.
  • the anti-tumor fusion protein is a protein that is not glycosylated.
  • the anti-tumor fusion protein is selected from the group consisting of:
  • (C) A derivative polypeptide obtained by subjecting the amino acid sequence of SEQ ID NO: 2 to substitution, deletion or addition of 1-10 amino acid residues, and retaining the activity of inhibiting tumor cell growth.
  • the tumor cell is a tumor cell expressing GnRHR, preferably a tumor cell expressing type I GnRHR.
  • the tumor cell is a tumor cell expressing a type II GnRHR.
  • amino acid sequence of the anti-tumor fusion protein is set forth in SEQ ID NO: 2.
  • the anti-tumor fusion protein contains a 6xHis purification tag.
  • the anti-tumor fusion protein is capable of inhibiting tumor cell growth and/or inducing apoptosis of tumor cells.
  • an isolated polynucleotide encoding the anti-tumor fusion protein of the first aspect of the invention is provided.
  • sequence of the polynucleotide is as shown in SEQ ID NO.: 1.
  • a vector comprising the polynucleotide of the second aspect of the invention is provided.
  • the vector comprises a plasmid, a viral vector.
  • the viral vector comprises a lentiviral vector, an adenoviral vector, and a yellow fever virus vector.
  • the vector comprises an expression vector.
  • a host cell comprising the vector of the third aspect of the invention or the polynucleotide of the second aspect of the invention integrated in the genome.
  • the host cell comprises a prokaryotic cell and a eukaryotic cell.
  • the host cell is a bacterium, preferably E. coli.
  • a method of producing an anti-tumor fusion protein comprising the steps of:
  • step (b) isolating and purifying the anti-tumor fusion protein expressed in the step (a).
  • a pharmaceutical composition comprising the anti-tumor fusion protein of the first aspect of the invention, and a pharmaceutically acceptable carrier or excipient is provided.
  • a use of the anti-tumor fusion protein of the first aspect of the invention for the preparation of a medicament for treating or preventing a tumor is provided.
  • the tumor is a tumor that expresses GnRH.
  • the tumor is selected from the group consisting of breast cancer, lung cancer, colon cancer, pancreatic cancer, ovarian cancer, prostate cancer, kidney cancer, liver cancer, brain cancer, melanoma, multiple myeloma, head and neck Tumor.
  • a method for non-therapeutic inhibition of tumor cells in vitro comprising the step of culturing said tumor cells in the presence of the anti-tumor fusion protein of the first aspect of the invention.
  • a method of treating a tumor comprising the step of administering the anti-tumor fusion protein of the first aspect of the invention to a subject in need thereof.
  • the anti-tumor fusion protein is administered as a monomer, a dimer and/or a tetramer, preferably the anti-tumor fusion protein is administered as a tetramer.
  • the object is a human.
  • Figure 1 shows an SDS-PAGE electropherogram of the fusion protein of the present invention which induces expression.
  • the first lane is Marker
  • the second lane is the sample before the inducer
  • the third lane is the whole bacterial protein sample after the inducer
  • the fourth lane is the soluble expression component after the inducer
  • fifth The lane is the inclusion body expression component after the inducer is added.
  • Figure 2 shows the killing effect of GnRH+PII+P53 on Ishikawa cells.
  • Figure 3 shows a schematic diagram of the molecular structure of PEA.
  • the present inventors have extensively and intensively studied, and for the first time, unexpectedly discovered a fusion protein which efficiently delivers p53 protein to the nucleus and efficiently induces apoptosis.
  • the fusion protein not only efficiently expresses, is not easily degraded, but also efficiently forms a polymer (especially a tetramer).
  • the experimental results also show that the fusion protein (monomer or multimer) of the present invention can enter the intracellular membrane extremely efficiently and efficiently, and enter the nucleus efficiently and rapidly, thereby extremely efficiently inducing abnormal cells (such as tumors). Apoptosis of cells).
  • the present invention has been completed on this basis.
  • active ingredient refers to an anti-tumor fusion protein of the invention.
  • fusion protein of the invention As used herein, the terms “fusion protein of the invention”, “fusion protein”, “anti-tumor fusion protein” are used interchangeably and refer to the fusion protein of the first aspect of the invention.
  • the fusion protein has a GnRH+PII+P53 structure, and the sequence is shown in SEQ ID NO.: 2.
  • the fusion protein having the structure of GnRH + P53 + TAT, the sequence shown in (QHWSYGLRPGHMEEPQSDPSVEPPLSQETFSDLWKLLPENNVLSPLPSQAMDDLMLSPDDIEQWFTEDPGPDEAPRMPEAAPRVAPAPAAPTPAAPAPAPSWPLSSSVPSQKTYQGSYGFRLGFLHSGTAKSVTCTYSPALNKMFCQLAKTCPVQLWVDSTPPPGTRVRAMAIYKQSQHMTEVVRRCPHHERCSDSDGLAPPQHLIRVEGNLRVEYLDDRNTFRHSVVVPYEPPEVGSDCTTIHYNYMCNSSCMGGMNRRPILTIITLEDSSGNLLGRNSFEVHVCACPGRDRRTEEENLRKKGEPHHELPPGSTKRALSNNTSSSPQPKKKPLDGEYFTLQIRGRERFEMFRELNEALELKDAQAGKEPGGSRAHSSHLKSKKGQSTSRHKKLMFKTEGPDSDGRKKRR
  • the two fusion proteins of the present invention can be used in combination after mixing.
  • the fusion protein of the present invention has excellent inhibitory activity against tumor cell growth
  • the fusion protein of the present invention and a pharmaceutical composition containing the fusion protein of the present invention as a main active ingredient can be used for (a) prevention Or inhibit tumor growth, metastasis or tumor cell growth, migration, or (b) induce apoptosis in human tumor cells.
  • compositions of the present invention comprise a fusion protein of the invention and a pharmaceutically acceptable excipient or carrier in a safe and effective amount.
  • safe and effective amount it is meant that the amount of fusion protein is sufficient to significantly improve the condition without causing serious side effects.
  • the pharmaceutical compositions contain from 1 to 2000 mg of the fusion protein per agent of the invention, more preferably from 10 to 200 mg of the compound of the invention per agent.
  • the "one dose" is a capsule or tablet.
  • “Pharmaceutically acceptable carrier” means: one or more compatible solid or liquid fillers or gel materials which are suitable for human use and which must be of sufficient purity and of sufficiently low toxicity.
  • “compatibility” it is meant herein that the components of the composition, as well as the components and the active ingredients of the present invention, can be incorporated into each other without significantly reducing the efficacy of the active ingredient.
  • pharmaceutically acceptable carriers are cellulose and its derivatives (such as sodium carboxymethylcellulose, sodium ethylcellulose, etc.), gelatin, talc, solid lubricants (such as stearic acid, magnesium stearate).
  • calcium sulfate such as soybean oil, sesame oil, olive oil, etc.
  • polyol such as propylene glycol, glycerin, sorbitol, etc.
  • emulsifier such as Tween
  • a wetting agent such as sodium lauryl sulfate
  • a coloring agent such as a flavoring agent, a stabilizer, an antioxidant, a preservative, a pyrogen-free water, and the like.
  • the administration mode of the fusion protein or the pharmaceutical composition thereof according to the present invention is not particularly limited, and representative administration methods include, but are not limited to, oral, intratumoral, rectal, and parenteral (intravenous, intramuscular or subcutaneous) ).
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active ingredient is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or mixed with: (a) a filler or compatibilizer, for example, Starch, lactose, sucrose, and silicic acid; (b) binders, for example, hydroxymethylcellulose, gelatin, sucrose, and acacia; (c) humectants, for example, glycerin; (d) disintegrants, for example , agar, calcium carbonate, potato starch or tapioca starch, and sodium carbonate; (e) a slow solvent such as paraffin; (f) an absorption accelerator, for example, a quaternary amine compound; (g) a wetting agent such as cetyl alcohol and a single Glyceryl stearate; (h) an adsorbent, for example, kaolin
  • Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other materials known in the art. They may contain opacifying agents and the release of the active ingredient in such compositions may be released in a portion of the digestive tract in a delayed manner. Examples of embedding components that can be employed are polymeric and waxy materials. If necessary, the active ingredient may also be in microencapsulated form with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or elixirs.
  • the liquid dosage form may contain inert diluents conventionally employed in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or a mixture of these substances.
  • inert diluents conventionally employed in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethyl
  • compositions may contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents and perfumes.
  • the suspension may contain suspending agents, for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar or mixtures of these and the like.
  • suspending agents for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar or mixtures of these and the like.
  • compositions for parenteral injection may comprise a physiologically acceptable sterile aqueous or nonaqueous solution, dispersion, suspension or emulsion, and a sterile powder for reconstitution into a sterile injectable solution or dispersion.
  • Suitable aqueous and nonaqueous vehicles, diluents, solvents or vehicles include water, ethanol, polyols, and suitable mixtures thereof.
  • the fusion proteins of the invention may be administered alone or in combination with other pharmaceutically acceptable compounds (or tumor suppressors).
  • the other pharmaceutically acceptable compound comprises an antitumor drug selected from the group consisting of an alkylating agent, an antimetabolite, a folic acid analog, a pyrimidine analog, a purine analog, a vinblastine, and epipodophyllotoxin. ), antibiotics, L-aspartate, topoisomerase inhibitor, interferon, platinum coordination complex, emodin-substituted urea, methyl hydrazine derivative, adrenocortical inhibitor, adrenal corticosteroid, pregnancy Hormones, estrogens, antiestrogens, androgens, antiandrogens, and or gonadotropin-releasing hormone analogs.
  • an antitumor drug selected from the group consisting of an alkylating agent, an antimetabolite, a folic acid analog, a pyrimidine analog, a purine analog, a vinblastine, and epipodophyllotoxin.
  • antibiotics L-aspartate, top
  • the anti-tumor drug is selected from the group consisting of 5-fluorouracil (5-FU), formyltetrahydrofolate, irinotecan, oxaliplatin, capecitabine, paclitaxel, docetaxel, Or a combination thereof.
  • 5-FU 5-fluorouracil
  • formyltetrahydrofolate irinotecan
  • oxaliplatin oxaliplatin
  • capecitabine paclitaxel
  • docetaxel docetaxel
  • a safe and effective amount of the fusion protein of the present invention is applied to a mammal (e.g., a human) in need of treatment, wherein the dose at the time of administration is a pharmaceutically effective effective dose for a person weighing 60 kg.
  • the daily dose is usually from 1 to 2000 mg, preferably from 20 to 500 mg.
  • specific doses should also consider factors such as the route of administration, the health of the patient, etc., which are within the skill of the skilled physician.
  • PEA is a single-stranded toxin protein consisting of 613 amino acids with a molecular weight of 66 kD. It consists of three structural functional zones: Zone I, Zone II and Zone III.
  • the I region is in the antiparallel beta structure at the N-terminus of the PEA molecule.
  • the I region is further divided into an Ia region and an Ib region, which are separated in the DNA sequence but close together in the three-dimensional structure.
  • the Ia region contains amino acid 1-252, which is responsible for binding to the target cell surface receptor-cell binding function; the Ib region contains amino acids 365-399, and the deletion of most of the region (amino acids 365-380) does not affect PEA.
  • Biological activity is a single-stranded toxin protein consisting of 613 amino acids with a molecular weight of 66 kD. It consists of three structural functional zones: Zone I, Zone II and Zone III.
  • the I region is in the antiparallel beta structure at the N
  • the region II is the central region, including amino acids 253-364, and has six consecutive alpha helices, which are responsible for transmembrane translocation function.
  • Region III includes amino acids 400-613, which have two functions: one is to catalyze ADP ribosylation of EF-2; the other is that its C-terminal specific amino acid sequence mediates toxin fragments into the endoplasmic reticulum.
  • the series consists of five amino acid residue fragments (Arg609Glu610Asp611Leu612Lys613 or REDLK), and its deletion causes the cytotoxicity of PE to be lost, and the sequence adjustment can significantly improve the ADP ribosylation efficiency of the toxin.
  • Table 1 shows the structure and function of PE molecules.
  • GnRH was isolated and purified from animals by Schally in 1971, and its structure was synthesized and then synthesized, and the Nobel Prize of 1976 was obtained.
  • GnRH is a decapeptide containing no free amino acid and carboxyl group, and its molecular structure is: P-Glu-His-Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly-NH 2 , of which amino acids 4-6 It forms a ⁇ -turn, which is hairpin-shaped and is suitable for binding to receptors. Positions 2 and 3 are important for biological activity, and position 6 plays an important role in maintaining hairpin conformation. The first and fourth amino acids are involved. Receptor binding, if substituted for the above amino acid residues, can result in loss of vigor or geometrical enhancement.
  • Natural GnRH is easily degraded by proteolytic enzymes in the body, so its half-life is only 4-8 min.
  • the main sites of action of the hydrolase peptidase are Gly6-Leu7 and Pro9-Gly10-NH2.
  • GnRH analogs more than 3,000 GnRH analogs have been synthesized by picking up or replacing amino acids in their peptide chain structures. Since the synthetic GnRH has a longer half-life and is more potent, it is more suitable for the treatment of patients than natural GnRH.
  • the requirement for the synthesis of long-acting GnRH agonists is to stabilize the molecular structure, making it less susceptible to enzymatic hydrolysis, increasing binding to circulating proteins and membranes, and increasing affinity for the GnRH receptor.
  • the 6 position is an analog of D-amino acid and the substituted Gly10 amide group.
  • This GnRH agonist is not only resistant to protease hydrolysis but also has a high affinity for receptors.
  • the introduction of a bulky hydrophobic group at position 6 further increases the affinity for the receptor. Such substitutions stabilize the "active" configuration of the release hormone analog, increasing binding to circulating proteins, thereby extending half-life.
  • the normal human gonadotropin-releasing hormone (GnRH) receptor is mainly present in the anterior pituitary, and there is a small amount of GnRH receptor distribution in the pituitary tissues such as the gonad placenta, although it can also be found in important organs such as liver, kidney, heart and skeletal muscle. A certain level of mRNA for the GnRH receptor was detected. However, using the method of quantitative analysis of receptors - radioligand analysis (RLA) to detect these organ tissues, only negative results can be obtained.
  • RLA radioligand analysis
  • GnRH can basically be divided into two types, namely
  • GnRH I the primary structure is as follows:
  • GnRH II the primary structure is as follows:
  • the corresponding receptors are also divided into two types, the type I GnRH receptor and the type II GnRH receptor.
  • the main differences between the two types of GnRH receptors are:
  • the transcription direction of the gene is different: the transcription direction of the type I GnRH receptor gene is in the sense direction, and the transcription direction of the type II GnRH receptor gene is in the antisense direction, so the difference between the RNA cleavage and the termination site is also large. the difference.
  • Amino acid composition is different: The amino acid homology of the amino acid expressed by exons 2 and 3 of the type II GnRH receptor gene and the same site of the type I receptor are 45% and 41%, respectively.
  • Type II GnRH receptor structure has a C-terminal cytoplasmic tail.
  • the type I GnRH receptor is absent.
  • the selectivity to ligand binding is different: ligand binding specificity tests performed on functional cells indicate that both receptors have clear ligand selectivity, and type II receptors have very high reactivity with type II GnRH, whereas The response capacity to type I GnRH is very low, and the difference between the two is 420 times.
  • GnRHRI mRNA is mainly distributed in pituitary and a few reproductive system tissues, while GnRHRII mRNA is widely distributed in almost all organ tissues. The significance of the widespread and abundant presence of type II GnRH receptors in human tissues is still unclear.
  • Wtp53 wild-type p53
  • Wtp53 is the most potent tumor suppressor found to date. Studying how to use wtp53 to treat tumors has become a hot spot in the field of cancer research. Because p53 protein has a short half-life and is a non-secretory protein, there is no p53 receptor or ligand on the cell membrane, making it difficult for p53 protein to break through the cell membrane and enter the cell. Play a role.
  • the p53 gene is located on human chromosome 17p13 and is about 20 kb in length. It consists of 11 exons and 10 introns and is transcribed into 2.5 kb mRNA, encoding a protein of 393 amino acids with a relative molecular mass of 53 ⁇ 10 3 .
  • the protein is a nuclear-binding protein with three major functional regions: 1) an N-terminal transcriptional activation region that activates transcription and mediates protein-protein interactions, which can also bind to p53 negative regulators; 2) Central DNA core binding region, which has specific binding to DNA function and is a hot spot for tumor cell mutation; 3) C-terminal non-specific DNA binding region, including nuclear localization signal region and nuclear export signal region
  • P53 protein is a phosphorylated protein that is easily hydrolyzed in cells and has a half-life of only about 30 minutes, which is almost undetectable in the nucleus of normal cells. However, when cells are stimulated by external factors, such as hypoxia, ultraviolet radiation or certain drugs, DNA damage, p53 protein acts as a transcription factor, inhibiting cell proliferation by transactivation; on the other hand, p53 protein It can directly interact with components in the DNA replication mechanism to inhibit DNA replication and ensure genetic stability. Specifically, P53 has the following functions:
  • P53 can effectively prevent malignant transformation of cells, monitor the integrity of the genome, and identify various abnormalities that may cause tumors. It is called “Geneguards.” Various damages in DNA can regulate p53 function through specific post-translational modifications, including phosphorylation and acetylation. Regulators such as viral-encoded proteins, intracellular proteins, and transcriptional repressors can also affect their function. By interacting with different synergistic molecules, P53 induces activation of different target genes, regulates the cell cycle, and stops the cell cycle at specific checkpoints.
  • P53 regulates the p21 gene and regulates it with p21, which plays an important regulatory role in the G1/S phase arrest caused by DNA damage.
  • CDK-activating kinase a cyclin-dependent protein kinase, binds to and activates cyclins, phosphorylates target proteins, and regulates cell cycle progression.
  • PCNA proliferating cell nuclear antigen
  • p21waf1 binds to PCNA, and the complex formed prevents the extension of DNA replication and affects the progression of the cell cycle.
  • PCNA and replication factor C recognize primer-template primers to promote polymerase ⁇ (pol ⁇ ) loading, and PCNA can also bind to polymerase ⁇ (pol ⁇ ).
  • the PCNA-RFC-pol ⁇ complex slides the DNA in the loop it forms, allowing the DNA leader strand to be continuously synthesized, accelerating the progress of the DNA replication elongation phase.
  • Direct binding of P21waf1 to PCNA results in rapid dissociation of the PCNA-RFC-pol ⁇ complex from the DNA replication fork, attenuating the initiation of PCNA cell proliferation, thereby preventing DNA replication synthesis.
  • p53 protein can directly interact with PCNA, inhibit DNA replication and prevent cell division.
  • p21waf1 disappeared in the nucleus, and in the late phase of the G2 phase, p21waf1 re-entered the nucleus and stayed for a short time. Similar to p53 blocking the G1 arrest by a non-transcriptional mechanism, p21waf1 also binds to the cyclin A and B complexes. P21waf1 prevents the protein substrate cdk2 from being activated by CAK, or directly inhibits cdk2 activity, preventing cells from passing through the checkpoint and interfering with cell cycle progression.
  • P21waf1 also forms a complex with cdk2/cyclinA, and the p21waf1-cdk2/cyclinA complex blocks the interaction of the substrate with cdk2/cyclinA.
  • the activation of cdk2 positively regulates the cdk1/cyclin B complex is weakened, making it impossible for cells to enter the mitosis phase.
  • P53 can induce tumor cell growth inhibition and promote apoptosis through two mechanisms.
  • p53 induces transcription-induced apoptosis such as Bax, Bcl-2, and p53-induced apoptosis-inducing proteins in the nucleus.
  • Bax is an essential signal for initiating apoptosis.
  • the p53 binding site is present on the Bax promoter.
  • the p53 recognition binding site acts directly on the Bax gene.
  • p53 induces apoptosis by inducing Bax transcription.
  • P53 promotes apoptosis
  • p53 mitochondrial anchoring mainly acts through the E3 ligase murine double minute 2 (mdm2).
  • mdm2 E3 ligase murine double minute 2
  • mdm2 E3 ligase murine double minute 2
  • mdm2 E3 ligase murine double minute 2
  • mdm2 E3 ligase murine double minute 2
  • mdm2 gene mediates the degradation of p53, inhibits the transcriptional activation of p53, and down-regulates its tumor growth inhibitory activity.
  • mdm2 induced by p53 can stabilize the function of p53 protein.
  • the cell-regulated autophagy modulator is mainly involved in the autophagy of cells.
  • the DRAM contains a p53-binding sequence and is a newly discovered p53 downstream target gene.
  • DRAM can activate the cell's self-phagocytic function and decompose long-acting proteins to stabilize the cell's morphology and maintain its basic life.
  • DRAM induces apoptosis when cells are stimulated.
  • the study found that when p53 is deleted and only DRAM is expressed, the killing effect of DRAM on cells is only 2% to 4% of the original killing effect, and when DRAM and p53 are co-transfected, the killing ability is greatly improved. Therefore, it is concluded that the role of DRAM in inducing programmed cell death is dependent on p53, which can induce autophagic death by p53.
  • TSP-1 thrombospondin-1
  • p53 can activate a (II) collagen prolyl-4-hydroxylase (a2PH) transcription, resulting in the synthesis and secretion of full-length collagen, and the production of anti-angiomotin, induced protein hydrolysis at the substrate level, increased Synthesis and decomposition of collagen-derived anti-angiogenic collagen.
  • P53 inhibits tumor angiogenesis by inhibiting the expression of angiogenic genes by stimulation.
  • VEGF vascular endothelial growth factor
  • P53 protein plays an important role in DNA repair process, mainly in the case of DNA damage, p53 protein prevents DNA replication, and gains time for DNA repair. If repair fails, p53 protein activates apoptosis-inducing mechanism and promotes programmed cell death. To maintain the stability of the body. P53 can also interact with DNA repair factors such as RPA, PCNA and other genes directly involved in DNA damage repair process. At the same time, p53 can interact with the components of Nucleotide Excision Repair (NER) to make NER also aggregate here for nucleotide repair.
  • NER Nucleotide Excision Repair
  • P53 has multiple abilities to interact with DNA, allowing p53 to be directly involved in DNA repair, such as binding to DNA repair factors, or directly involved in DNA repair through p53 protein-protein interactions, which bind p53 to damaged DNA. It is of great significance. Under the action of DNA damage factors, the damaged DNA is detected at the C-terminus of p53, and binds to form a p53-DNA complex. P53 binds to damaged DNA and acts as a transcription factor, binds to sequence-specific DNA, and participates in and enhances DNA repair by trans-activating target genes.
  • Ras (P21) protein is located inside the cell membrane and plays an important role in transmitting cell growth and differentiation signals. Ras regulates the role of Rasomolog gene fami ly member A (RhoA). On the one hand, Ras can anchor the membrane of RhoA; on the other hand, Ras promotes tyrosine phosphorylation of p190Rho GTPase activating protein (Rho GAP), which promotes the hydrolysis of RhoA-GTP to inactive RhoA-GDP, resulting in decreased RhoA activity.
  • Rho GAP Rasomolog gene fami ly member A
  • P53 is an essential factor for Ras to promote Rho GAP phosphorylation, independent of membrane anchoring of RhoA. After P53 deletion, the phosphorylation of p190Rho GAP is reduced, which reduces the loading of RhoA GTP, which greatly promotes the cell's ability to move.
  • HIV-I Human immunodeficiency virus type I
  • Tat is the causative agent of acquired immunodeficiency syndrome (AIDS).
  • Tat is a short peptide rich in basic amino acids encoded by HIV-I, and its sequence is YGRKKRRQRRR (SEQ ID NO.: 5, positions 405-416), which is one of the six regulatory proteins it encodes.
  • the regulatory protein, and the core region of the Tat transduction domain is composed of these 11 amino acid residues.
  • Tat protein can efficiently mediate DNA, peptide, protein and other molecules covalently linked to it into almost all tissues and cells, and even pass the blood-brain barrier, and the transduction efficiency is high. It has almost no damage to cells and maintains the biological activity of the protein.
  • the Tat fusion protein system is considered to be a promising and efficient delivery vehicle with broad application prospects in basic medical research and clinical treatment.
  • Tat protein can guide a variety of peptides and proteins into almost all target cells, which is the transduction of Tat protein, also known as internalization.
  • the transduction of Tat protein depends mainly on the concentration of the polypeptide or protein, and is different from the general channel, receptor, and endocytosis mode of entry. Because of this function, Tat can be used as a carrier for mediating foreign proteins through cell membranes, and is therefore receiving increasing attention.
  • Tat protein mediates foreign matter into cells by cavern pathway
  • Caveolae is a small depression on the cell membrane, about 50-70 nm.
  • Eguchi performed a bacteriomycin test on Tat-mediated exogenous gene transduction, which inhibited the entry of foreign substances into cells via a cavelet pathway, and found that Tat-mediated foreign protein transduction was inhibited. Therefore, it is concluded that Tat enters the cell in a small nest by destroying the plasma membrane.
  • This method is simple to operate, free from external factors such as temperature, and is not affected by other internal environment, does not produce toxicity, and can be directly applied to cells in the body.
  • the induction effect of this method is significantly higher than other transfection methods, the speed is fast, the transduction efficiency is high, and the biological activity of the protein is retained during the transduction process.
  • step (b) isolating and purifying the anti-tumor fusion protein expressed in the step (a).
  • the host cell is a bacterium, preferably E. coli.
  • the method for preparing GnRH+PII+P53 provided by the present invention includes the following aspects:
  • the engineered strain expresses the fusion protein 6xhis-SUMO-GnRH+PII+P53 under the induction of IPTG, and the fusion protein is expressed in the form of inclusion bodies;
  • the fusion protein is renatured by dropwise addition to the renaturation fluid
  • GnRH+PII+P53 crude product was purified by molecular sieve, and finally GnRH+PII+P53 with purity greater than 95% was obtained, which showed a single band on SDS-PAGE electrophoresis and tetramer protein on molecular sieve;
  • step 10 The pure GnRH+PII+P53 obtained in step 9) was tested for biological activity by ISHIKAWA cells, and >95% of the cells disintegrated through the apoptotic pathway at a final protein concentration of 0.5 ⁇ g/ml or more.
  • the fusion protein of the present invention is not easily degraded, and a tetramer can be formed naturally and efficiently.
  • the fusion protein (tetramer) of the present invention can efficiently enter target cells and rapidly enter the nuclear region to induce apoptosis of the mutant cells (such as tumor cells).
  • the fusion protein of the present invention has no toxicity in animal experiments due to the use of P53.
  • GnRH+PII+P53 According to the designed amino acid sequence of GnRH+PII+P53, through gene software analysis, the rare codon and the secondary structure of the gene, as well as the use of synonymous codon, were fully considered, and finally the preferred GnRH+PII+P53 encoding nucleic acid sequence was obtained. As shown in SEQ ID NO.: 1, the specific nucleic acid sequence is:
  • the GnRH+PII+P53 protein sequence is shown in SEQ ID NO.: 2, and the specific protein sequence is:
  • restriction endonuclease NdeI site and the sequence encoding the purified and purified sequence and the ulp1 recognition sequence sumo were added to the N-terminus of the optimized sequence, and the restriction endonuclease Hind III site was added at the C-terminus.
  • SEQ ID NO.: 3 the specific nucleic acid sequence is:
  • the sequence was synthesized by commercial gene synthesis company Shanghai Shenggong Bioengineering Co., Ltd. and cloned into PUC19 vector. After sequencing, it was named as PUC19-GnRH+PII+P53.
  • the preferred expression vector for expression of GnRH+PII+P53 in E. coli was PET21, and Pet21 and PUC19-GnRH+PII+P53 were digested with restriction endonucleases Nde I and Hind III, and the digested products were agarose-condensed. After gel electrophoresis separation, the gel was recovered, and then ligated with T4 ligase, and the ligated product was transformed into Escherichia coli DH5 ⁇ . A positive clone was picked and the plasmid was extracted, and after sequencing, it was named pET220-GnRH+PII+P53.
  • the pET220-GnRH+PII+P53 protein sequence is shown in SEQ ID NO.: 4, and the specific protein sequence is:
  • the plasmid pET220-GnRH+PII+P53 was transformed into E. coli host BL21 Codon Plus (DE3) RP. After centrifugation, the bacteria were coated on LB plates containing 100 ⁇ g/ml ampicillin and cultured overnight at 37 ° C on LB plates. Monoclonal culture was carried out and identified by colony PCR to obtain monoclonal transformant pET220-GnRH+PII+P53/BL21Codon Plus(DE3)RP inoculated with monoclonal transformant pET220-GnRH+PII+P53/BL21Codon Plus(DE3)RP.
  • the results are shown in Figure 1.
  • the fusion protein is expressed in the form of inclusion bodies, and the expression of the fusion protein accounts for about 25% of the total bacterial protein.
  • pET220-GnRH+PII+P53/BL21Codon Plus(DE3)RP was inoculated in LB medium and cultured overnight at 37 °C. The next day, 2% of the inoculum was used to access fresh TB (glycerol 5 g/L, peptone 12 g). / liter, yeast extract 24 g / liter, K2HPO4 12.54 g / liter, KH2PO42.31 g / liter) in the fermentation liquid culture medium, cultured at 37 ° C until the bacterial OD600 value reached 1, adding a final concentration of 0.5 mM IPTG induced fusion protein Expression, the cells were collected after 4 hours of incubation.
  • TB glycerol 5 g/L, peptone 12 g.
  • the fusion protein was expressed in the form of inclusion bodies.
  • the cells were resuspended in a ratio of 1:10 with PBS, and the homogenate pressure was set to 750 Pa, homogenized twice, and the precipitated fraction was collected by centrifugation at 15,000 g; Part of the inclusion body protein is in the precipitated fraction of the collection.
  • the inclusion bodies were fully dissolved in a ratio of 1:20 with solution 1 (20 Mm Tris-HCL, 500 mM NaCl, 20 mM Imidazole, 8 M Urea 20 mM 2-Mercaptoethanol pH 8.0), centrifuged at 15000 g for 20 minutes under centrifugation, and the supernatant fraction was taken. Further purification
  • the supernatant fraction was purified by denaturing conditions by NI 2+ metal chelating chromatography.
  • the fusion protein was placed in a denaturation solution containing 8 M Urea, and the fusion protein was treated with DTT at a final concentration of 5 mM, and stirred at room temperature overnight.
  • the treated denatured protein was added dropwise to the reconstitution solution (reconstitution solution was 100 mM Tris-HCL, 500 mM NaCl, 0.5 M Arginine, 1% triton X-100, 10% glycerol, 1 mM EDTA, 1 mM GSH, 0.5 mM). GSSG pH 8.0) After the addition is completed, place in the refrigerator at 4 ° C for 48-72 hours, fully renatured.
  • the fully refolded protein solution was placed in a dialysis bag and dialyzed against a buffer (20 Mm Tris. CL, Ph 8.5) for 4 hours each time for a total of 3 times of dialysis; after dialysis, the target protein was recovered by an anion column Q.
  • the ULP1 enzyme of 20IU/mg fusion protein was added to the fusion protein recovered by anion column Q, and stirred slowly by adding a magnetic stirrer to ensure sufficient digestion.
  • the enzyme digestion conditions were: 4 ° C, 16-24 hours.
  • the digested fusion protein solution is mainly composed of two components: fusion expression tag protein and GnRH+PII+P53, and a small amount of undigested intact fusion protein, wherein the fusion expression tag protein and the intact fusion protein both contain a 6xHis purification tag.
  • the two parts of the protein can be combined by NI 2+ metal chelating chromatography, and GnRH+PII+P53 cannot be bound to the NI 2+ metal chelating column and exists in the flow-through portion.
  • the column was filled with GnRH+PII+P53 in the flow-through portion (step 7) by affinity chromatography packed with heparin affinity packing purchased from GE Healthcare, and the concentrated GnRH+PII+P53 was refined by SUPERDEX 200.
  • the protein solution is sterile filtered and used for subsequent experiments.
  • the GnRH+PII+P53 fusion protein Slg001 prepared in Example 4 was used as an example to demonstrate its killing effect on Ishikawa cells. The detection results are shown in Figure 2. The high concentration of cells is lethal, and there are no normal cells. Particle fragments. details as follows:
  • the medium in the 20 ⁇ g/ml Slg001 treated plate was red, and a large number of granular cell debris were observed by microscopic observation.
  • the medium in the 10 ⁇ g/ml Slg001 treated plate was light red, microscopic observation Some cells and cell debris were observed, and the cells treated with low concentration of Slg001 were not significantly different from the control group;
  • the GnRH+PII+P53 fusion protein prepared in Example 4 was intravenously administered to three groups of CAL-27 tumor-bearing mice, and administered every other day for five times.
  • the doses were 250 ⁇ g/kg, 500 ⁇ g/kg, and 1000 ⁇ g, respectively.
  • /kg, 2000 ⁇ g/kg, 4000 ⁇ g/kg observe whether the tumor-bearing mice have toxic reaction and whether the drug is effective. As a result, no toxic reaction was observed, and the tumor was not enlarged in the first 4 needles, and the tumor inhibition rate was 72% after the fifth needle.

Landscapes

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

Abstract

The present invention provides an anti-tumor fusion protein, a preparation method therefor and an application thereof. Specifically, the present invention provides a fusion protein comprising a GnRH protein element, a transmembrane transport region of a PEA protein, and a P53 protein element. The fusion protein can efficiently transport a p53 protein to a cell nucleus and efficiently induce tumor cell apoptosis. The present invention also provides a nucleotide encoding the fusion protein, a method for producing the fusion protein, and a pharmaceutical composition comprising the fusion protein.

Description

抗肿瘤融合蛋白及其制法和应用Anti-tumor fusion protein and preparation method and application thereof 技术领域Technical field
本发明涉及生物医药领域,更具体地涉及一种抗肿瘤融合蛋白及其制法和应用。The invention relates to the field of biomedicine, and more particularly to an anti-tumor fusion protein and a preparation method and application thereof.
背景技术Background technique
免疫毒素是一组人工构建的具有特异性细胞杀伤能力的杂合分子,由毒性部分、载体部分和靶向部分三部分组成。毒性部分可以是植物、动物、微生物来源的细胞毒素,靶向部分可以是单克隆抗体或细胞因子。免疫毒素与其他抗肿瘤药物相比,具有毒性强和特异性高的优点,在肿瘤治疗中显示出巨大的应用前景。An immunotoxin is a group of artificially constructed hybrid molecules with specific cell killing ability, consisting of a toxic part, a carrier part and a targeting part. The toxic portion may be a cytotoxin of plant, animal, or microbial origin, and the targeting moiety may be a monoclonal antibody or a cytokine. Compared with other anti-tumor drugs, immunotoxins have the advantages of strong toxicity and high specificity, and have shown great application prospects in tumor treatment.
冻干重组人促黄体激素释放激素-绿脓杆菌外毒素A融合蛋白(lyophilized recombinant human luteinizing hormone releasing hormone-exotoxin of pseudomonas aeruginosa fusion protein,简称LHRH-PE40)是长春基因工程药物研究生产的一种对肿瘤细胞有特异性杀伤作用的重组毒素。其生产方法为首先采用基因工程的方法将编码绿脓杆菌外毒素A基因的受体结合区(I区)切除,置换成LHRH基因,再将LHRH-PE40重组基因克隆于质粒中,通过工程发酵表达出LHRH-PE40融合蛋白。Lyophilized recombinant human luteinizing hormone releasing hormone-exotoxin of pseudomonas aeruginosa fusion protein (LHRH-PE40) is a pair of research and production of Changchun genetic engineering drugs Tumor cells have a specific killing effect of recombinant toxins. The production method comprises the following steps: firstly, the receptor binding region (I region) encoding the Pseudomonas aeruginosa exotoxin A gene is excised by a genetic engineering method, and replaced with the LHRH gene, and then the LHRH-PE40 recombinant gene is cloned into a plasmid and passed through engineering fermentation. The LHRH-PE40 fusion protein was expressed.
LHRH-PE40中发挥细胞毒作用的是绿脓杆菌外毒素A(exotoxin A of pseudomonas aeruginosa,PEA)。绿脓杆菌外毒素A是绿脓杆菌中毒力最强的一种毒素,能阻断细胞蛋白质的合成并引起细胞的凋亡。LHRH-PE40中的导向物为人促黄体激素释放激素(human luteinizing hormone releasing hormone,LHRH),通过与其在肿瘤细胞表面的I型受体结合而靶向性地把PEA导入到肿瘤细胞中发挥抗肿瘤作用。The cytotoxic effect of LHRH-PE40 is exotoxin A of pseudomonas aeruginosa (PEA). Pseudomonas aeruginosa exotoxin A is the most potent toxin of Pseudomonas aeruginosa, which can block the synthesis of cellular proteins and cause apoptosis of cells. The leader in LHRH-PE40 is human luteinizing hormone releasing hormone (LHRH), which is targeted to introduce PEA into tumor cells to exert anti-tumor by binding to its type I receptor on the surface of tumor cells. effect.
LHRH的受体有两型,即I型和II型。LHRH与I型受体的亲和力很高。人体正常情况下,I型受体主要存在于垂体前叶,垂体外组织如性腺、胎盘和脑组织也含有一定量的I型受体,其它重要器官均不表达I型LHRH受体。然而某些肿瘤细胞表面由于受体异化则分布着大量的LHRH I型受体,包括生殖系统恶性肿瘤、黑色素瘤、胃癌、肝癌、胰腺癌、肠癌、肺癌等。由于I型LHRH受体在肿瘤细胞中普遍高表达,而在正常组织中只呈局限性分布,因此LHRH是设计重组靶向毒素所需的理想导向物。There are two types of receptors for LHRH, type I and type II. LHRH has a high affinity for type I receptors. Under normal conditions, type I receptors mainly exist in the anterior pituitary, and tissues such as gonads, placenta and brain tissues also contain a certain amount of type I receptors, and other important organs do not express type I LHRH receptors. However, on the surface of some tumor cells, a large number of LHRH type I receptors are distributed due to receptor alienation, including reproductive system malignant tumors, melanoma, gastric cancer, liver cancer, pancreatic cancer, intestinal cancer, and lung cancer. Since type I LHRH receptors are generally highly expressed in tumor cells and are only localized in normal tissues, LHRH is an ideal guide for designing recombinant targeting toxins.
LHRH的II型受体广泛存在于人体各组织中。LHRH与II型受体的亲和力很低。但在很高剂量的情况下,LHRH可以和II型受体发生低亲和力结合,从而使LHRH-PE40产生广泛的细胞毒作用。这是LHRH-PE40重组毒素对动物产生毒性作用的理论基础。LHRH-PE40不能透过血脑屏障,因此不会对垂体产生毒性作用。Type II receptors of LHRH are widely present in various tissues of the human body. LHRH has a low affinity for type II receptors. However, at very high doses, LHRH can bind to the type II receptor with low affinity, resulting in extensive cytotoxicity of LHRH-PE40. This is the theoretical basis for the toxic effects of LHRH-PE40 recombinant toxins on animals. LHRH-PE40 does not penetrate the blood-brain barrier and therefore does not cause toxic effects on the pituitary gland.
尽管以LHRH-PE40为代表的免疫毒素显示出良好的应用前景,但其仍存在免疫原性和非特异的细胞毒性等问题,阻碍了免疫毒素在临床上的应用。因此, 本领域迫切需要开发特异性好、细胞毒性低的新的免疫毒素。Although the immunotoxin represented by LHRH-PE40 shows a good application prospect, it still has problems such as immunogenicity and non-specific cytotoxicity, which hinders the clinical application of immunotoxin. Therefore, there is an urgent need in the art to develop new immunotoxins with good specificity and low cytotoxicity.
发明内容Summary of the invention
本发明的目的在于提供一种抗肿瘤融合蛋白及其制法和应用。The object of the present invention is to provide an anti-tumor fusion protein, a preparation method and application thereof.
在本发明的第一方面,提供了一种抗肿瘤融合蛋白,所述的抗肿瘤融合蛋白具有式I所述结构:In a first aspect of the invention, an anti-tumor fusion protein is provided, said anti-tumor fusion protein having the structure of formula I:
D-A-B-C-E     (I)D-A-B-C-E (I)
其中,among them,
A为GnRH蛋白元件;A is a GnRH protein element;
B为PEA蛋白的跨膜转运区或为无;B is the transmembrane transport region of the PEA protein or is absent;
C为P53蛋白元件;C is a P53 protein element;
E为TAT蛋白元件或为无;E is a TAT protein element or is absent;
D为任选的信号肽和/或前导肽序列;D is an optional signal peptide and/or leader peptide sequence;
并且,B和E不同时为无;Also, B and E are not the same at the same time;
其中,“-”表示连接上述各元件的肽键。Here, "-" means a peptide bond connecting the above elements.
在另一优选例中,所述的抗肿瘤融合蛋白具有式I I所述结构:In another preferred embodiment, the anti-tumor fusion protein has the structure of Formula II:
D-A-C-E     (II)D-A-C-E (II)
其中,among them,
A为GnRH蛋白元件;A is a GnRH protein element;
C为P53蛋白元件;C is a P53 protein element;
E为TAT蛋白元件;E is a TAT protein element;
D为任选的信号肽和/或前导肽序列;D is an optional signal peptide and/or leader peptide sequence;
其中,“-”表示连接上述各元件的肽键。Here, "-" means a peptide bond connecting the above elements.
在另一优选例中,所述的抗肿瘤融合蛋白In another preferred embodiment, the anti-tumor fusion protein
在另一优选例中,所述的TAT为HIV-I编码的一段富含碱性氨基酸的短肽。In another preferred embodiment, the TAT is a short peptide rich in basic amino acids encoded by HIV-I.
在另一优选例中,所述的TAT蛋白来源于人或非人哺乳动物。In another preferred embodiment, the TAT protein is derived from a human or a non-human mammal.
在另一优选例中,所述的TAT蛋白包括野生型和突变型。In another preferred embodiment, the TAT protein comprises wild-type and mutant forms.
在另一优选例中,所述的TAT蛋白包括全长的、成熟形式的P53,或其活性片段。In another preferred embodiment, the TAT protein comprises a full length, mature form of P53, or an active fragment thereof.
在另一优选例中,所述的TAT蛋白的序列如SEQ ID NO.:5中第405-416位所示。In another preferred embodiment, the sequence of the TAT protein is shown in positions 405-416 of SEQ ID NO.: 5.
在另一优选例中,所述的抗肿瘤融合蛋白选自下组:In another preferred embodiment, the anti-tumor fusion protein is selected from the group consisting of:
(A)具有SEQ ID NO.:5所示氨基酸序列的多肽;(A) a polypeptide having the amino acid sequence of SEQ ID NO.: 5;
(B)具有与SEQ ID NO.:5所示氨基酸序列≥80%同源性(优选地,≥90%的同源性;等优选地≥95%的同源性;最优选地,≥97%的同源性,如98%以上,99%以上)的多肽,且所述多肽具有抑制肿瘤细胞生长的活性;(B) having ≥80% homology to the amino acid sequence shown in SEQ ID NO.: 5 (preferably, ≥90% homology; etc. preferably ≥95% homology; most preferably, ≥97 % homology, such as 98% or more, 99% or more) of the polypeptide, and the polypeptide has an activity of inhibiting tumor cell growth;
(C)将SEQ ID NO:5所示氨基酸序列经过1-10个氨基酸残基的取代、缺失或 添加而形成的,且保留抑制肿瘤细胞生长活性的衍生多肽。(C) A derivative polypeptide obtained by subjecting the amino acid sequence shown by SEQ ID NO: 5 to substitution, deletion or addition of 1-10 amino acid residues, and retaining the activity of inhibiting tumor cell growth.
在另一优选例中,所述的抗肿瘤融合蛋白具有式I I所述结构:In another preferred embodiment, the anti-tumor fusion protein has the structure of Formula II:
D-A-B-C       (II)D-A-B-C (II)
其中,among them,
A为GnRH蛋白元件;A is a GnRH protein element;
B为PEA蛋白的跨膜转运区;B is a transmembrane transport region of PEA protein;
C为P53蛋白元件;C is a P53 protein element;
D为任选的信号肽和/或前导肽序列;D is an optional signal peptide and/or leader peptide sequence;
“-”表示连接上述各元件的肽键。"-" indicates a peptide bond connecting the above elements.
在另一优选例中,所述的GnRH蛋白来源于人或非人哺乳动物。In another preferred embodiment, the GnRH protein is derived from a human or a non-human mammal.
在另一优选例中,所述的GnRH蛋白包括野生型和突变型。In another preferred embodiment, the GnRH protein comprises wild type and mutant form.
在另一优选例中,所述的GnRH蛋白包括全长的、成熟形式的GnRH,或其活性片段。In another preferred embodiment, the GnRH protein comprises a full length, mature form of GnRH, or an active fragment thereof.
在另一优选例中,所述的GnRH蛋白包括II型GnRH蛋白和I型GnRH蛋白。In another preferred embodiment, the GnRH protein comprises a type II GnRH protein and a type I GnRH protein.
在另一优选例中,所述的GnRH蛋白为II型GnRH蛋白。In another preferred embodiment, the GnRH protein is a type II GnRH protein.
在另一优选例中,所述的GnRH蛋白的序列如SEQ ID NO.:2中第1-10位所示。In another preferred embodiment, the sequence of the GnRH protein is as shown in 1-10 of SEQ ID NO.: 2.
在另一优选例中,所述的P53蛋白来源于人或非人哺乳动物。In another preferred embodiment, the P53 protein is derived from a human or a non-human mammal.
在另一优选例中,所述的P53蛋白包括野生型和突变型。In another preferred embodiment, the P53 protein comprises wild type and mutant form.
在另一优选例中,所述的P53蛋白包括全长的、成熟形式的P53,或其活性片段。In another preferred embodiment, the P53 protein comprises a full length, mature form of P53, or an active fragment thereof.
在另一优选例中,所述的P53蛋白的序列如SEQ ID NO.:2中第128-520位所示。In another preferred embodiment, the sequence of the P53 protein is shown in positions 128-520 of SEQ ID NO.: 2.
在另一优选例中,所述PEA蛋白的跨膜转运区来源于绿脓杆菌。In another preferred embodiment, the transmembrane transport region of the PEA protein is derived from Pseudomonas aeruginosa.
在另一优选例中,所述PEA蛋白的跨膜转运区的长度为100-120氨基酸,较佳地为100-115个氨基酸。In another preferred embodiment, the transmembrane transport region of the PEA protein is from 100 to 120 amino acids in length, preferably from 100 to 115 amino acids.
在另一优选例中,所述PEA蛋白的跨膜转运区的序列如SEQ ID NO.:2中第13-127位所示。In another preferred embodiment, the sequence of the transmembrane trafficking region of the PEA protein is shown in positions 13-127 of SEQ ID NO.: 2.
在另一优选例中,所述的抗肿瘤融合蛋白是由细菌,较佳地由大肠杆菌表达的重组蛋白。In another preferred embodiment, the anti-tumor fusion protein is a recombinant protein expressed by bacteria, preferably E. coli.
在另一优选例中,所述的抗肿瘤融合蛋白是不经糖基化修饰的蛋白。In another preferred embodiment, the anti-tumor fusion protein is a protein that is not glycosylated.
在另一优选例中,所述的抗肿瘤融合蛋白选自下组:In another preferred embodiment, the anti-tumor fusion protein is selected from the group consisting of:
(A)具有SEQ ID NO.:2所示氨基酸序列的多肽;(A) a polypeptide having the amino acid sequence of SEQ ID NO.: 2;
(B)具有与SEQ ID NO.:2所示氨基酸序列≥80%同源性(优选地,≥90%的同源性;等优选地≥95%的同源性;最优选地,≥97%的同源性,如98%以上, 99%以上)的多肽,且所述多肽具有抑制肿瘤细胞生长的活性;(B) having ≥80% homology to the amino acid sequence shown in SEQ ID NO.: 2 (preferably, ≥90% homology; etc. preferably ≥95% homology; most preferably, ≥97 % homology, such as 98% or more, 99% or more) of the polypeptide, and the polypeptide has an activity of inhibiting tumor cell growth;
(C)将SEQ ID NO:2所示氨基酸序列经过1-10个氨基酸残基的取代、缺失或添加而形成的,且保留抑制肿瘤细胞生长活性的衍生多肽。(C) A derivative polypeptide obtained by subjecting the amino acid sequence of SEQ ID NO: 2 to substitution, deletion or addition of 1-10 amino acid residues, and retaining the activity of inhibiting tumor cell growth.
在另一优选例中,所述的肿瘤细胞为表达GnRHR的肿瘤细胞,较佳地为表达I型GnRHR的肿瘤细胞。In another preferred embodiment, the tumor cell is a tumor cell expressing GnRHR, preferably a tumor cell expressing type I GnRHR.
在另一优选例中,所述的肿瘤细胞为表达II型GnRHR的肿瘤细胞。In another preferred embodiment, the tumor cell is a tumor cell expressing a type II GnRHR.
在另一优选例中,所述抗肿瘤融合蛋白的氨基酸序列如SEQ ID NO:2所示。In another preferred embodiment, the amino acid sequence of the anti-tumor fusion protein is set forth in SEQ ID NO: 2.
在另一优选例中,所述抗肿瘤融合蛋白含有6xHis纯化标签。In another preferred embodiment, the anti-tumor fusion protein contains a 6xHis purification tag.
在另一优选例中,所述抗肿瘤融合蛋白能够抑制肿瘤细胞生长和/或诱导肿瘤细胞的凋亡。In another preferred embodiment, the anti-tumor fusion protein is capable of inhibiting tumor cell growth and/or inducing apoptosis of tumor cells.
在本发明的第二方面,提供了一种分离的多核苷酸,所述的多核苷酸编码本发明第一方面所述的抗肿瘤融合蛋白。In a second aspect of the invention, an isolated polynucleotide encoding the anti-tumor fusion protein of the first aspect of the invention is provided.
在另一优选例中,所述多核苷酸的序列如SEQ ID NO.:1所示。In another preferred embodiment, the sequence of the polynucleotide is as shown in SEQ ID NO.: 1.
在本发明的第三方面,提供了一种载体,所述载体含有本发明第二方面所述的多核苷酸。In a third aspect of the invention, a vector comprising the polynucleotide of the second aspect of the invention is provided.
在另一优选例中,所述的载体包括质粒、病毒载体。In another preferred embodiment, the vector comprises a plasmid, a viral vector.
在另一优选例中,所述的病毒载体包括:慢病毒载体、腺病毒载体、黄热病毒载体。In another preferred embodiment, the viral vector comprises a lentiviral vector, an adenoviral vector, and a yellow fever virus vector.
在另一优选例中,所述的载体包括表达载体。In another preferred embodiment, the vector comprises an expression vector.
在本发明的第四方面,提供了一种宿主细胞,所述宿主细胞含有本发明第三方面所述的载体或基因组中整合有本发明第二方面所述的多核苷酸。In a fourth aspect of the invention, there is provided a host cell comprising the vector of the third aspect of the invention or the polynucleotide of the second aspect of the invention integrated in the genome.
在另一优选例中,所述宿主细胞包括原核细胞和真核细胞。In another preferred embodiment, the host cell comprises a prokaryotic cell and a eukaryotic cell.
在另一优选例中,所述宿主细胞为细菌,较佳地为大肠杆菌。In another preferred embodiment, the host cell is a bacterium, preferably E. coli.
在本发明的第五方面,提供了一种生产抗肿瘤融合蛋白的方法,包括步骤:In a fifth aspect of the invention, a method of producing an anti-tumor fusion protein comprising the steps of:
(a)在适合表达条件下,培养本发明第四方面所述的宿主细胞,从而表达本发明第一方面所述的抗肿瘤融合蛋白;(a) cultivating the host cell of the fourth aspect of the invention under conditions suitable for expression, thereby expressing the anti-tumor fusion protein of the first aspect of the invention;
(b)分离纯化出步骤(a)中所表达的抗肿瘤融合蛋白。(b) isolating and purifying the anti-tumor fusion protein expressed in the step (a).
在本发明的第六方面,提供了一种药物组合物,所述药物组合物含有本发明第一方面所述的抗肿瘤融合蛋白,以及药学上可接受的载体或赋形剂。In a sixth aspect of the invention, a pharmaceutical composition comprising the anti-tumor fusion protein of the first aspect of the invention, and a pharmaceutically acceptable carrier or excipient is provided.
在本发明的第七方面,提供了一种本发明第一方面所述抗肿瘤融合蛋白的用途,用于制备治疗或预防肿瘤的药物。In a seventh aspect of the invention, there is provided a use of the anti-tumor fusion protein of the first aspect of the invention for the preparation of a medicament for treating or preventing a tumor.
在另一优选例中,所述的肿瘤为表达GnRH的肿瘤。In another preferred embodiment, the tumor is a tumor that expresses GnRH.
在另一优选例中,所述肿瘤选自下组:乳腺癌,肺癌,大肠癌,胰腺癌,卵巢癌,前列腺癌,肾癌,肝癌,脑癌,黑色素瘤,多发性骨髓瘤,头颈部肿瘤。In another preferred embodiment, the tumor is selected from the group consisting of breast cancer, lung cancer, colon cancer, pancreatic cancer, ovarian cancer, prostate cancer, kidney cancer, liver cancer, brain cancer, melanoma, multiple myeloma, head and neck Tumor.
在本发明的第八方面,提供了一种体外非治疗性的抑制肿瘤细胞的方法,包括步骤:在本发明第一方面所述的抗肿瘤融合蛋白存在的条件下,培养所述肿瘤细胞。In an eighth aspect of the invention, a method for non-therapeutic inhibition of tumor cells in vitro is provided, comprising the step of culturing said tumor cells in the presence of the anti-tumor fusion protein of the first aspect of the invention.
在本发明的第九方面,提供了一种治疗肿瘤的方法,包括步骤:给需要的对象施用本发明第一方面所述的抗肿瘤融合蛋白。In a ninth aspect of the invention, a method of treating a tumor comprising the step of administering the anti-tumor fusion protein of the first aspect of the invention to a subject in need thereof.
在另一优选例中,所述的抗肿瘤融合蛋白以单体、二聚体和/或四聚体形式施用,较佳地所述的抗肿瘤融合蛋白以四聚体形式施用。In another preferred embodiment, the anti-tumor fusion protein is administered as a monomer, a dimer and/or a tetramer, preferably the anti-tumor fusion protein is administered as a tetramer.
在另一优选例中,所述的对象是人。In another preferred embodiment, the object is a human.
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。It is to be understood that within the scope of the present invention, the various technical features of the present invention and the various technical features specifically described hereinafter (as in the embodiments) may be combined with each other to constitute a new or preferred technical solution. Due to space limitations, we will not repeat them here.
附图说明DRAWINGS
图1显示了诱导表达的本发明融合蛋白的SDS-PAGE电泳图。其中,第一泳道为Marker,第二泳道为加诱导剂前的样品,第三泳道为加诱导剂后的全菌蛋白样品,第四泳道为加诱导剂后的可溶表达组份,第五泳道为加诱导剂后的包涵体表达组份。Figure 1 shows an SDS-PAGE electropherogram of the fusion protein of the present invention which induces expression. Wherein, the first lane is Marker, the second lane is the sample before the inducer, the third lane is the whole bacterial protein sample after the inducer, and the fourth lane is the soluble expression component after the inducer, fifth The lane is the inclusion body expression component after the inducer is added.
图2显示了GnRH+PII+P53对Ishikawa细胞的杀伤效果。Figure 2 shows the killing effect of GnRH+PII+P53 on Ishikawa cells.
图3显示了PEA分子结构示意图。Figure 3 shows a schematic diagram of the molecular structure of PEA.
具体实施方式detailed description
本发明人经过广泛而深入地研究,首次意外地发现一种高效地将p53蛋白输送到细胞核并高效引发细胞凋亡的融合蛋白。该融合蛋白不仅可以高效地表达,不易降解,而且高效地形成多聚体(尤其是四聚体)。此外,实验结果还表明,本发明的融合蛋白(单体或多聚体)可以极其高效地穿过细胞膜进入到胞内,并高效快速地进入细胞核,从而极其高效地诱导非正常细胞(如肿瘤细胞)的凋亡。在此基础上完成了本发明。The present inventors have extensively and intensively studied, and for the first time, unexpectedly discovered a fusion protein which efficiently delivers p53 protein to the nucleus and efficiently induces apoptosis. The fusion protein not only efficiently expresses, is not easily degraded, but also efficiently forms a polymer (especially a tetramer). In addition, the experimental results also show that the fusion protein (monomer or multimer) of the present invention can enter the intracellular membrane extremely efficiently and efficiently, and enter the nucleus efficiently and rapidly, thereby extremely efficiently inducing abnormal cells (such as tumors). Apoptosis of cells). The present invention has been completed on this basis.
融合蛋白Fusion protein
如本文所用,术语“活性成分”是指本发明所述的抗肿瘤融合蛋白。The term "active ingredient" as used herein refers to an anti-tumor fusion protein of the invention.
如本文所用,术语“本发明的融合蛋白”、“融合蛋白”、“抗肿瘤融合蛋白”可互换使用,是指本发明第一方面所述的融合蛋白。As used herein, the terms "fusion protein of the invention", "fusion protein", "anti-tumor fusion protein" are used interchangeably and refer to the fusion protein of the first aspect of the invention.
在一优选例中,所述的融合蛋白具有GnRH+PII+P53结构,序列如SEQ ID NO.: 2所示。In a preferred embodiment, the fusion protein has a GnRH+PII+P53 structure, and the sequence is shown in SEQ ID NO.: 2.
在另一优选例中,所述的融合蛋白具有GnRH+P53+TAT结构,序列SEQ ID NO.:5所示(QHWSYGLRPGHMEEPQSDPSVEPPLSQETFSDLWKLLPENNVLSPLPSQAMDDLMLSPDDIEQWFTEDPGPDEAPRMPEAAPRVAPAPAAPTPAAPAPAPSWPLSSSVPSQKTYQGSYGFRLGFLHSGTAKSVTCTYSPALNKMFCQLAKTCPVQLWVDSTPPPGTRVRAMAIYKQSQHMTEVVRRCPHHERCSDSDGLAPPQHLIRVEGNLRVEYLDDRNTFRHSVVVPYEPPEVGSDCTTIHYNYMCNSSCMGGMNRRPILTIITLEDSSGNLLGRNSFEVHVCACPGRDRRTEEENLRKKGEPHHELPPGSTKRALSNNTSSSPQPKKKPLDGEYFTLQIRGRERFEMFRELNEALELKDAQAGKEPGGSRAHSSHLKSKKGQSTSRHKKLMFKTEGPDSDGRKKRRQRRRPQ),其中第1-10为GnRH,第12-404位为P53,第405-416位为TAT。In another preferred embodiment, the fusion protein having the structure of GnRH + P53 + TAT, the sequence shown in (QHWSYGLRPGHMEEPQSDPSVEPPLSQETFSDLWKLLPENNVLSPLPSQAMDDLMLSPDDIEQWFTEDPGPDEAPRMPEAAPRVAPAPAAPTPAAPAPAPSWPLSSSVPSQKTYQGSYGFRLGFLHSGTAKSVTCTYSPALNKMFCQLAKTCPVQLWVDSTPPPGTRVRAMAIYKQSQHMTEVVRRCPHHERCSDSDGLAPPQHLIRVEGNLRVEYLDDRNTFRHSVVVPYEPPEVGSDCTTIHYNYMCNSSCMGGMNRRPILTIITLEDSSGNLLGRNSFEVHVCACPGRDRRTEEENLRKKGEPHHELPPGSTKRALSNNTSSSPQPKKKPLDGEYFTLQIRGRERFEMFRELNEALELKDAQAGKEPGGSRAHSSHLKSKKGQSTSRHKKLMFKTEGPDSDGRKKRRQRRRPQ) SEQ ID NO.:5, which is of GnRH 1-10, 12-404 of bit P53, the first Bits 405-416 are TAT.
本发明的两种融合蛋白可以混合后共同使用。The two fusion proteins of the present invention can be used in combination after mixing.
药物组合物Pharmaceutical composition
由于本发明所述的融合蛋白具有优异的对肿瘤细胞生长的抑制活性,因此本发明所述的融合蛋白以及含有本发明所述的融合蛋白为主要活性成分的药物组合物可用于(a)预防或抑制肿瘤生长、转移或肿瘤细胞增长、迁移,或(b)诱导人肿瘤细胞凋亡。Since the fusion protein of the present invention has excellent inhibitory activity against tumor cell growth, the fusion protein of the present invention and a pharmaceutical composition containing the fusion protein of the present invention as a main active ingredient can be used for (a) prevention Or inhibit tumor growth, metastasis or tumor cell growth, migration, or (b) induce apoptosis in human tumor cells.
本发明的药物组合物包含安全有效量范围内的本发明所述的融合蛋白及药理上可以接受的赋形剂或载体。其中“安全有效量”指的是:融合蛋白的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有1-2000mg本发明所述的融合蛋白/剂,更佳地,含有10-200mg本发明化合物/剂。较佳地,所述的“一剂”为一个胶囊或药片。The pharmaceutical compositions of the present invention comprise a fusion protein of the invention and a pharmaceutically acceptable excipient or carrier in a safe and effective amount. By "safe and effective amount" it is meant that the amount of fusion protein is sufficient to significantly improve the condition without causing serious side effects. In general, the pharmaceutical compositions contain from 1 to 2000 mg of the fusion protein per agent of the invention, more preferably from 10 to 200 mg of the compound of the invention per agent. Preferably, the "one dose" is a capsule or tablet.
“药学上可以接受的载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组合物中各组份之间以及各组份和本发明所述活性成分能相互掺和,而不明显降低活性成分的药效。药学上可以接受的载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、橄榄油等)、多元醇(如丙二醇、甘油、山梨醇等)、乳化剂(如吐温
Figure PCTCN2019081735-appb-000001
)、润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。
"Pharmaceutically acceptable carrier" means: one or more compatible solid or liquid fillers or gel materials which are suitable for human use and which must be of sufficient purity and of sufficiently low toxicity. By "compatibility" it is meant herein that the components of the composition, as well as the components and the active ingredients of the present invention, can be incorporated into each other without significantly reducing the efficacy of the active ingredient. Examples of pharmaceutically acceptable carriers are cellulose and its derivatives (such as sodium carboxymethylcellulose, sodium ethylcellulose, etc.), gelatin, talc, solid lubricants (such as stearic acid, magnesium stearate). , calcium sulfate, vegetable oil (such as soybean oil, sesame oil, olive oil, etc.), polyol (such as propylene glycol, glycerin, sorbitol, etc.), emulsifier (such as Tween
Figure PCTCN2019081735-appb-000001
), a wetting agent (such as sodium lauryl sulfate), a coloring agent, a flavoring agent, a stabilizer, an antioxidant, a preservative, a pyrogen-free water, and the like.
施用方法Application method
本发明所述的融合蛋白或其药物组合物的施用方式没有特别限制,代表性的施用方式包括(但并不限于):口服、瘤内、直肠、和肠胃外(静脉内、肌肉内或皮下)。The administration mode of the fusion protein or the pharmaceutical composition thereof according to the present invention is not particularly limited, and representative administration methods include, but are not limited to, oral, intratumoral, rectal, and parenteral (intravenous, intramuscular or subcutaneous) ).
用于口服给药的固体剂型包括胶囊剂、片剂、丸剂、散剂和颗粒剂。在这些固体剂型中,活性成分与至少一种常规惰性赋形剂(或载体)混合,如柠檬酸钠或磷酸二钙,或与下述成分混合:(a)填料或增容剂,例如,淀粉、乳糖、蔗糖、和硅酸;(b)粘合剂,例如,羟甲基纤维素、明胶、蔗糖和阿拉伯胶; (c)保湿剂,例如,甘油;(d)崩解剂,例如,琼脂、碳酸钙、马铃薯淀粉或木薯淀粉、和碳酸钠;(e)缓溶剂,例如石蜡;(f)吸收加速剂,例如,季胺化合物;(g)润湿剂,例如鲸蜡醇和单硬脂酸甘油酯;(h)吸附剂,例如,高岭土;和(i)润滑剂,例如,滑石、硬脂酸钙、十二烷基硫酸钠,或其混合物。胶囊剂、片剂和丸剂中,剂型也可包含缓冲剂。Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In these solid dosage forms, the active ingredient is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or mixed with: (a) a filler or compatibilizer, for example, Starch, lactose, sucrose, and silicic acid; (b) binders, for example, hydroxymethylcellulose, gelatin, sucrose, and acacia; (c) humectants, for example, glycerin; (d) disintegrants, for example , agar, calcium carbonate, potato starch or tapioca starch, and sodium carbonate; (e) a slow solvent such as paraffin; (f) an absorption accelerator, for example, a quaternary amine compound; (g) a wetting agent such as cetyl alcohol and a single Glyceryl stearate; (h) an adsorbent, for example, kaolin; and (i) a lubricant, for example, talc, calcium stearate, sodium lauryl sulfate, or a mixture thereof. In capsules, tablets and pills, the dosage form may also contain a buffer.
固体剂型如片剂、糖丸、胶囊剂、丸剂和颗粒剂可采用包衣和壳材制备,如肠衣和其它本领域公知的材料。它们可包含不透明剂,并且,这种组合物中活性成分的释放可以延迟的方式在消化道内的某一部分中释放。可采用的包埋组分的实例是聚合物质和蜡类物质。必要时,活性成分也可与上述赋形剂中的一种或多种形成微胶囊形式。Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other materials known in the art. They may contain opacifying agents and the release of the active ingredient in such compositions may be released in a portion of the digestive tract in a delayed manner. Examples of embedding components that can be employed are polymeric and waxy materials. If necessary, the active ingredient may also be in microencapsulated form with one or more of the above-mentioned excipients.
用于口服给药的液体剂型包括药学上可接受的乳液、溶液、悬浮液、糖浆或酊剂。除了活性成分外,液体剂型可包含本领域中常规采用的惰性稀释剂,如水或其它溶剂,增溶剂和乳化剂,例知,乙醇、异丙醇、碳酸乙酯、乙酸乙酯、丙二醇、1,3-丁二醇、二甲基甲酰胺以及油,特别是棉籽油、花生油、玉米胚油、橄榄油、蓖麻油和芝麻油或这些物质的混合物等。Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or elixirs. In addition to the active ingredient, the liquid dosage form may contain inert diluents conventionally employed in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or a mixture of these substances.
除了这些惰性稀释剂外,组合物也可包含助剂,如润湿剂、乳化剂和悬浮剂、甜味剂、矫味剂和香料。In addition to these inert diluents, the compositions may contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents and perfumes.
除了活性成分外,悬浮液可包含悬浮剂,例如,乙氧基化异十八烷醇、聚氧乙烯山梨醇和脱水山梨醇酯、微晶纤维素、甲醇铝和琼脂或这些物质的混合物等。In addition to the active ingredient, the suspension may contain suspending agents, for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar or mixtures of these and the like.
用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、分散液、悬浮液或乳液,和用于重新溶解成无菌的可注射溶液或分散液的无菌粉末。适宜的含水和非水载体、稀释剂、溶剂或赋形剂包括水、乙醇、多元醇及其适宜的混合物。Compositions for parenteral injection may comprise a physiologically acceptable sterile aqueous or nonaqueous solution, dispersion, suspension or emulsion, and a sterile powder for reconstitution into a sterile injectable solution or dispersion. Suitable aqueous and nonaqueous vehicles, diluents, solvents or vehicles include water, ethanol, polyols, and suitable mixtures thereof.
本发明所述的融合蛋白可以单独给药,或者与其他药学上可接受的化合物(或肿瘤抑制剂)联合给药。The fusion proteins of the invention may be administered alone or in combination with other pharmaceutically acceptable compounds (or tumor suppressors).
所述其他药学上可接受的化合物包含选自下组的抗肿瘤药物:烷化剂、抗代谢物、叶酸类似物、嘧啶类似物、嘌呤类似物、长春花碱类、表鬼臼毒素(epipodophyllotoxin)、抗生素、L-天冬酞胺酶、拓扑异构酶抑制剂、干扰素、铂配位复合物、大黄素取代的脲、甲基肼衍生物、肾上腺皮质抑制剂、肾上腺皮质类固醇、孕激素、雌激素、抗雌激素、雄激素、抗雄激素以及或促性腺激素-释放激素类似物。The other pharmaceutically acceptable compound comprises an antitumor drug selected from the group consisting of an alkylating agent, an antimetabolite, a folic acid analog, a pyrimidine analog, a purine analog, a vinblastine, and epipodophyllotoxin. ), antibiotics, L-aspartate, topoisomerase inhibitor, interferon, platinum coordination complex, emodin-substituted urea, methyl hydrazine derivative, adrenocortical inhibitor, adrenal corticosteroid, pregnancy Hormones, estrogens, antiestrogens, androgens, antiandrogens, and or gonadotropin-releasing hormone analogs.
优选地,所述抗肿瘤药物选自下组的物质:5-氟尿嘧啶(5-FU)、甲酰四氢叶酸、伊立替康、奥沙利铂、卡培他滨、紫杉醇、多西紫杉醇、或其组合。Preferably, the anti-tumor drug is selected from the group consisting of 5-fluorouracil (5-FU), formyltetrahydrofolate, irinotecan, oxaliplatin, capecitabine, paclitaxel, docetaxel, Or a combination thereof.
使用药物组合物时,是将安全有效量的本发明所述的融合蛋白适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,日给药剂量通常为1~2000mg,优选20~500mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。When a pharmaceutical composition is used, a safe and effective amount of the fusion protein of the present invention is applied to a mammal (e.g., a human) in need of treatment, wherein the dose at the time of administration is a pharmaceutically effective effective dose for a person weighing 60 kg. In general, the daily dose is usually from 1 to 2000 mg, preferably from 20 to 500 mg. Of course, specific doses should also consider factors such as the route of administration, the health of the patient, etc., which are within the skill of the skilled physician.
PEAPEA
PEA是由613个氨基酸构成的单链毒素蛋白,分子量66kD。它由三个结构功能区构成:I区、II区和III区。I区在PEA分子的N端,呈反向平行的β结构。I区又分为Ia区和Ib区,这两部分在DNA序列上是分离的,但在三维结构中紧靠在一起。Ia区含第1-252位氨基酸,负责与靶细胞表面受体结合-细胞结合功能;Ib区含365-399位氨基酸,此区的大部(第365-380位氨基酸)缺失并不影响PEA的生物学活性。II区为中央区,包括第253-364位氨基酸,有6个连续的α螺旋,它负责跨膜转位功能,当II区缺失时,尽管其细胞结合功能和ADP核糖基化活性尚存,但细胞毒性丧失,说明II区为毒素转位功能所必需。III区包括400-613位氨基酸,它有两个功能:其一是催化EF-2的ADP核糖基化作用;其二是它的C末端特异氨基酸序列介导毒素片断进行入内质网,此特异系列是由(Arg609Glu610Asp611Leu612Lys613即REDLK)五个氨基酸残基片段构成,它的缺失,使PE的细胞毒性丧失,而将其进行序列调整,能够明显增进毒素的ADP核糖基化效率。下表1显示PE分子结构与功能关系。PEA is a single-stranded toxin protein consisting of 613 amino acids with a molecular weight of 66 kD. It consists of three structural functional zones: Zone I, Zone II and Zone III. The I region is in the antiparallel beta structure at the N-terminus of the PEA molecule. The I region is further divided into an Ia region and an Ib region, which are separated in the DNA sequence but close together in the three-dimensional structure. The Ia region contains amino acid 1-252, which is responsible for binding to the target cell surface receptor-cell binding function; the Ib region contains amino acids 365-399, and the deletion of most of the region (amino acids 365-380) does not affect PEA. Biological activity. The region II is the central region, including amino acids 253-364, and has six consecutive alpha helices, which are responsible for transmembrane translocation function. When the region II is deleted, although its cell-binding function and ADP ribosylation activity remain, However, loss of cytotoxicity indicates that zone II is required for toxin translocation. Region III includes amino acids 400-613, which have two functions: one is to catalyze ADP ribosylation of EF-2; the other is that its C-terminal specific amino acid sequence mediates toxin fragments into the endoplasmic reticulum. The series consists of five amino acid residue fragments (Arg609Glu610Asp611Leu612Lys613 or REDLK), and its deletion causes the cytotoxicity of PE to be lost, and the sequence adjustment can significantly improve the ADP ribosylation efficiency of the toxin. Table 1 below shows the structure and function of PE molecules.
表1:PE分子结构与功能关系Table 1: Relationship between structure and function of PE molecules
Figure PCTCN2019081735-appb-000002
Figure PCTCN2019081735-appb-000002
PEA分子的结构如图3所示。The structure of the PEA molecule is shown in Figure 3.
GnRHGnRH
GnRH是由Schally等于1971年从动物体内分离纯化,阐明其结构后又人工合成的,并以此获得了1976年的Nobel奖。GnRH为一个不含游离氨基酸与羧基的十肽,其分子结构为:P-Glu-His-Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly-NH 2,其中第4-6位氨基酸形成β转折,呈发夹形,适合与受体结合,第2和3位对生物活性很重要,第6位对维持发夹构象起重要作用,第1位与第4-10位氨基酸均参与受体结合,若置换以上氨基酸残基可导致活力丧失或呈几何级增强。 GnRH was isolated and purified from animals by Schally in 1971, and its structure was synthesized and then synthesized, and the Nobel Prize of 1976 was obtained. GnRH is a decapeptide containing no free amino acid and carboxyl group, and its molecular structure is: P-Glu-His-Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly-NH 2 , of which amino acids 4-6 It forms a β-turn, which is hairpin-shaped and is suitable for binding to receptors. Positions 2 and 3 are important for biological activity, and position 6 plays an important role in maintaining hairpin conformation. The first and fourth amino acids are involved. Receptor binding, if substituted for the above amino acid residues, can result in loss of vigor or geometrical enhancement.
天然GnRH在体内很容易被蛋白水解酶降解,故其半衰期仅4-8min。其水解酶肽酶的主要作用部位是Gly6-Leu7和Pro9-Gly10-NH2。为寻求高效且持久的GnRH类似物,通过对其肽链结构中氨基酸的拾取或替换,已合成3000多种GnRH类似物。由于合成的GnRH半衰期长,作用更强,所以比天然的GnRH更适于病人的治疗。合成长效的GnRH激动剂的要求是稳定分子结构,使之不易被酶水解, 增加与循环中的蛋白和胞膜的结合及提高对GnRH受体的亲和力。如6位为D-氨基酸的类似物和取代Gly10酰胺基。这种GnRH激动剂不仅耐蛋白酶水解作用较大,而且对受体有较高的亲和力。若在第6位引入庞大的疏水基团可进一步增加与受体的亲和力。这样的置换稳定了释放激素类似物的“活性”构型,提高了与循环中蛋白的结合,从而延长半衰期。Natural GnRH is easily degraded by proteolytic enzymes in the body, so its half-life is only 4-8 min. The main sites of action of the hydrolase peptidase are Gly6-Leu7 and Pro9-Gly10-NH2. To find efficient and long lasting GnRH analogs, more than 3,000 GnRH analogs have been synthesized by picking up or replacing amino acids in their peptide chain structures. Since the synthetic GnRH has a longer half-life and is more potent, it is more suitable for the treatment of patients than natural GnRH. The requirement for the synthesis of long-acting GnRH agonists is to stabilize the molecular structure, making it less susceptible to enzymatic hydrolysis, increasing binding to circulating proteins and membranes, and increasing affinity for the GnRH receptor. For example, the 6 position is an analog of D-amino acid and the substituted Gly10 amide group. This GnRH agonist is not only resistant to protease hydrolysis but also has a high affinity for receptors. The introduction of a bulky hydrophobic group at position 6 further increases the affinity for the receptor. Such substitutions stabilize the "active" configuration of the release hormone analog, increasing binding to circulating proteins, thereby extending half-life.
正常人的性腺激素释放激素(GnRH)受体主要存在于垂体前叶,垂体外组织如性腺胎盘组织中有少量GnRH受体分布,虽然在重要器官如肝、肾、心及骨骼肌中也可以检测出一定水平的GnRH受体的mRNA。但使用受体定量分析的方法—放射配体分析法(RLA)检测这些器官组织,则只能获得阴性结果。The normal human gonadotropin-releasing hormone (GnRH) receptor is mainly present in the anterior pituitary, and there is a small amount of GnRH receptor distribution in the pituitary tissues such as the gonad placenta, although it can also be found in important organs such as liver, kidney, heart and skeletal muscle. A certain level of mRNA for the GnRH receptor was detected. However, using the method of quantitative analysis of receptors - radioligand analysis (RLA) to detect these organ tissues, only negative results can be obtained.
目前的研究表明,GnRH基本上可以分成两种,即Current research shows that GnRH can basically be divided into two types, namely
GnRH Ⅰ,一级结构如下:GnRH I, the primary structure is as follows:
pGlu-His-Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly-NH 2 pGlu-His-Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly-NH 2
GnRH Ⅱ,一级结构如下:GnRH II, the primary structure is as follows:
pGlu-His-Trp-Ser-His-Gly-Trp-Tyr-Pro-Gly-NH 2 pGlu-His-Trp-Ser-His-Gly-Trp-Tyr-Pro-Gly-NH 2
其相应的受体也分为两种即Ⅰ型GnRH受体和Ⅱ型GnRH受体。两型GnRH受体的主要区别是:The corresponding receptors are also divided into two types, the type I GnRH receptor and the type II GnRH receptor. The main differences between the two types of GnRH receptors are:
1基因存在位置不同:Ⅰ型GnRH受体基因存在于8号染色体上,Ⅱ型GnRH受体基因存在于20号染色体上。1 The genes exist in different positions: the type I GnRH receptor gene is present on chromosome 8, and the type II GnRH receptor gene is present on chromosome 20.
2基因转录方向不同:Ⅰ型GnRH受体基因转录方向是正义方向进行的,而Ⅱ型GnRH受体基因转录方向是反义方向进行的,因此造成RNA剪切和终止位点的差异也有很大区别。2 The transcription direction of the gene is different: the transcription direction of the type I GnRH receptor gene is in the sense direction, and the transcription direction of the type II GnRH receptor gene is in the antisense direction, so the difference between the RNA cleavage and the termination site is also large. the difference.
3氨基酸组成不同:Ⅱ型GnRH受体基因外显子2和3所表达的氨基酸与Ⅰ型受体的相同部位氨基酸同源性分别为45%和41%。3 Amino acid composition is different: The amino acid homology of the amino acid expressed by exons 2 and 3 of the type II GnRH receptor gene and the same site of the type I receptor are 45% and 41%, respectively.
4分子结构不同:Ⅱ型GnRH受体结构上有一个C末端胞浆区尾部。而Ⅰ型GnRH受体则无。4 Molecular structure is different: Type II GnRH receptor structure has a C-terminal cytoplasmic tail. The type I GnRH receptor is absent.
5与配基结合的选择性不同:在功能细胞上进行的配基结合特异性试验表明两种受体有明确的配基选择性,Ⅱ型受体对Ⅱ型GnRH的反应能力非常高,而对I型GnRH的反应能力则很低,二者相差420余倍。5 The selectivity to ligand binding is different: ligand binding specificity tests performed on functional cells indicate that both receptors have clear ligand selectivity, and type II receptors have very high reactivity with type II GnRH, whereas The response capacity to type I GnRH is very low, and the difference between the two is 420 times.
6在组织中分布不同:利用RT-PCR检测,GnRHRⅠ的mRNA主要分布在垂体中和少数生殖系统组织中,而GnRHRⅡ的mRNA则广泛分布在几乎所有的器官组织中。Ⅱ型GnRH受体在人组织中广泛而丰富的存在的意义至今尚不十分清楚。6 Distribution in tissues: Using RT-PCR, GnRHRI mRNA is mainly distributed in pituitary and a few reproductive system tissues, while GnRHRII mRNA is widely distributed in almost all organ tissues. The significance of the widespread and abundant presence of type II GnRH receptors in human tissues is still unclear.
7不同配基与不同受体结合后所产生的信号不同。7 different ligands are different from the signals produced by binding to different receptors.
P53P53
Wtp53(wild-type p53)是迄今为止发现的最有效的肿瘤抑制因子。研究如何应用wtp53治疗肿瘤已成为肿瘤研究领域中的热点,由于p53蛋白半衰期短暂,且属于非分泌型蛋白,细胞膜上不存在p53受体或配体,使p53蛋白很难突破细胞膜而进入细胞内发挥作用。Wtp53 (wild-type p53) is the most potent tumor suppressor found to date. Studying how to use wtp53 to treat tumors has become a hot spot in the field of cancer research. Because p53 protein has a short half-life and is a non-secretory protein, there is no p53 receptor or ligand on the cell membrane, making it difficult for p53 protein to break through the cell membrane and enter the cell. Play a role.
目前,国内外的研究大多集中在通过载体(如病毒载体AAV)将p53基因导入细胞内,但导入的载体存在不安全性因素,因此如何使p53蛋白进入细胞内发挥作用,且既不影响p53蛋白的生物活性,又避免导入的载体的不安全性是应用p53治疗肿瘤必须解决的问题。At present, most studies at home and abroad focus on introducing p53 gene into cells through a vector (such as viral vector AAV), but the introduced vector has unsafe factors, so how to make p53 protein enter the cell, and it does not affect p53. The biological activity of the protein, and avoiding the unsafeness of the introduced vector, is a problem that must be solved in the treatment of tumors with p53.
p53基因定位于人染色体17p13,全长约20kb,由11个外显子和10个内含子组成,转录成2.5kbmRNA,编码393个氨基酸的蛋白质,相对分子质量为53×10 3。该蛋白是一种核结合蛋白,含有3个主要功能区:1)N-末端转录激活区,可激活转录,介导蛋白间相互作用,这一区域还可和p53负调控因子结合;2)中央DNA核心结合区,这一区域具有特异性结合DNA功能,并且是肿瘤细胞突变热点区域;3)C-末端非专一DNA结合区,包括核定位信号区和核输出信号区 The p53 gene is located on human chromosome 17p13 and is about 20 kb in length. It consists of 11 exons and 10 introns and is transcribed into 2.5 kb mRNA, encoding a protein of 393 amino acids with a relative molecular mass of 53×10 3 . The protein is a nuclear-binding protein with three major functional regions: 1) an N-terminal transcriptional activation region that activates transcription and mediates protein-protein interactions, which can also bind to p53 negative regulators; 2) Central DNA core binding region, which has specific binding to DNA function and is a hot spot for tumor cell mutation; 3) C-terminal non-specific DNA binding region, including nuclear localization signal region and nuclear export signal region
P53蛋白是一种磷酸化蛋白,在细胞中易水解,半衰期只有大约30分钟,在正常细胞的细胞核中几乎检测不到。但当细胞受到外界因素刺激,如低氧、紫外照射或某些药物作用而导致DNA损伤时,p53蛋白一方面作为转录因子,通过反式激活对细胞增殖起抑制作用;另一方面,p53蛋白可直接同DNA复制机制中的成分相互作用,抑制DNA的复制,保证了遗传的稳定性。具体地,P53具有以下功能:P53 protein is a phosphorylated protein that is easily hydrolyzed in cells and has a half-life of only about 30 minutes, which is almost undetectable in the nucleus of normal cells. However, when cells are stimulated by external factors, such as hypoxia, ultraviolet radiation or certain drugs, DNA damage, p53 protein acts as a transcription factor, inhibiting cell proliferation by transactivation; on the other hand, p53 protein It can directly interact with components in the DNA replication mechanism to inhibit DNA replication and ensure genetic stability. Specifically, P53 has the following functions:
P53对细胞周期调控P53 regulates cell cycle
P53能有效防止细胞恶性转化,监视基因组的完整性,识别各种可能引发肿瘤的异常情况,被称为“基因卫士”。DNA的各种损伤均可通过特异性的翻译后修饰来调节p53功能,主要包括磷酸化和乙酰化,病毒编码的蛋白、细胞内蛋白和转录抑制物等调节物也可影响其功能。P53通过与不同的协同分子相互作用,诱导激活不同的靶基因,对细胞周期进行调控,使细胞周期停顿于特定的检查点。P53 can effectively prevent malignant transformation of cells, monitor the integrity of the genome, and identify various abnormalities that may cause tumors. It is called "Geneguards." Various damages in DNA can regulate p53 function through specific post-translational modifications, including phosphorylation and acetylation. Regulators such as viral-encoded proteins, intracellular proteins, and transcriptional repressors can also affect their function. By interacting with different synergistic molecules, P53 induces activation of different target genes, regulates the cell cycle, and stops the cell cycle at specific checkpoints.
细胞周期G1期停顿Cell cycle G1 pause
P53通过上调p21基因,与p21相互调节,在DNA损伤所致的G1/S期停顿过程中起重要调控作用。P53 regulates the p21 gene and regulates it with p21, which plays an important regulatory role in the G1/S phase arrest caused by DNA damage.
P53的另一种阻滞细胞周期G1期停滞通过非转录机制进行。CDK激酶(CDK-activating kinase,CAK)即周期蛋白依赖性蛋白激酶,能与细胞周期蛋白结合并活化,使靶蛋白磷酸化、调控细胞周期进程。Another blockade of P53 in the G1 phase of cell cycle arrest is through a non-transcriptional mechanism. CDK-activating kinase (CAK), a cyclin-dependent protein kinase, binds to and activates cyclins, phosphorylates target proteins, and regulates cell cycle progression.
细胞周期S期停顿Cell cycle S phase pause
P21waf1可与增殖细胞核抗原(PCNA),一种参与真核细胞复制的蛋白质结合,而抑制PCNA的活性。PCNA只存在与增殖细胞及肿瘤细胞内,p21waf1与PCNA结合,形成的复合物阻止DNA复制的延伸,影响细胞周期的进展。在DNA复制过程中,PCNA与复制因子C(replication factor C,RFC)共同识别引物-模板连接(primer template junction),促进聚合酶δ(polδ)加载,PCNA也可与聚合酶ε(polε)结合,PCNA-RFC-polδ复合物使DNA在其形成的环中滑行,使DNA前导链连续合成,加快DNA复制延伸阶段进展。P21waf1与PCNA直接结合导致PCNA-RFC-polδ复合物快速从DNA复制叉上解离,使PCNA细胞增殖的启动作用减弱,从而阻止DNA的复制合成。此外,p53蛋白还可以直接与PCNA作用,抑制 DNA的复制,阻止细胞分裂。P21waf1 binds to proliferating cell nuclear antigen (PCNA), a protein involved in eukaryotic cell replication, and inhibits PCNA activity. PCNA only exists in proliferating cells and tumor cells, and p21waf1 binds to PCNA, and the complex formed prevents the extension of DNA replication and affects the progression of the cell cycle. During DNA replication, PCNA and replication factor C (RFC) recognize primer-template primers to promote polymerase δ (polδ) loading, and PCNA can also bind to polymerase ε (polε). The PCNA-RFC-polδ complex slides the DNA in the loop it forms, allowing the DNA leader strand to be continuously synthesized, accelerating the progress of the DNA replication elongation phase. Direct binding of P21waf1 to PCNA results in rapid dissociation of the PCNA-RFC-polδ complex from the DNA replication fork, attenuating the initiation of PCNA cell proliferation, thereby preventing DNA replication synthesis. In addition, p53 protein can directly interact with PCNA, inhibit DNA replication and prevent cell division.
细胞周期G2期停顿Cell cycle G2 pause
在细胞周期S期时,p21waf1在核内消失,而在细胞周期G2期的后期,p21waf1又再次进入核内,短暂停留。与p53通过非转录机制阻滞G1期停滞类似,p21waf1也能与cyclin A及B复合物结合。P21waf1可阻止蛋白底物cdk2被CAK活化,或直接抑制cdk2活性,使细胞无法度过检验点,干扰细胞周期的进展。P21waf1还能与cdk2/cyclinA形成复合物,p21waf1-cdk2/cyclinA复合物可阻断底物与cdk2/cyclinA的相互作用。另外,当cdk2与p21waf1形成复合物后,cdk2正向调节cdk1/cyclin B复合物的激活作用减弱,使细胞无法进入有丝分裂期。In the S phase of the cell cycle, p21waf1 disappeared in the nucleus, and in the late phase of the G2 phase, p21waf1 re-entered the nucleus and stayed for a short time. Similar to p53 blocking the G1 arrest by a non-transcriptional mechanism, p21waf1 also binds to the cyclin A and B complexes. P21waf1 prevents the protein substrate cdk2 from being activated by CAK, or directly inhibits cdk2 activity, preventing cells from passing through the checkpoint and interfering with cell cycle progression. P21waf1 also forms a complex with cdk2/cyclinA, and the p21waf1-cdk2/cyclinA complex blocks the interaction of the substrate with cdk2/cyclinA. In addition, when cdk2 forms a complex with p21waf1, the activation of cdk2 positively regulates the cdk1/cyclin B complex is weakened, making it impossible for cells to enter the mitosis phase.
P53与细胞凋亡P53 and apoptosis
P53可通过两种机制诱导肿瘤细胞生长抑制,促进细胞凋亡。一方面作为转录因子,p53在细胞核中诱导Bax、Bcl-2和p53调节凋亡的诱导蛋白等转录诱发凋亡。Bax是启动凋亡的必需信号,Bax启动子上存在p53的结合位点,p53识别结合位点直接作用于Bax基因,p53通过诱导Bax转录诱发细胞凋亡P53 can induce tumor cell growth inhibition and promote apoptosis through two mechanisms. On the one hand, as a transcription factor, p53 induces transcription-induced apoptosis such as Bax, Bcl-2, and p53-induced apoptosis-inducing proteins in the nucleus. Bax is an essential signal for initiating apoptosis. The p53 binding site is present on the Bax promoter. The p53 recognition binding site acts directly on the Bax gene. p53 induces apoptosis by inducing Bax transcription.
P53蛋白的另一种促进细胞凋亡的机制是直接在细胞质中锚定至线粒体,诱导线粒体依赖性细胞凋亡。研究表明,p53线粒体锚定作用主要通过E3连接酶鼠双微体2(murine double minute 2,mdm2)起作用。P53作为重要的抑癌基因可通过多种途径调控肿瘤的发生和发展,而mdm2基因在同p53的相互作用中,一方面介导了p53的降解,抑制p53转录激活,下调其肿瘤生长抑制活性;另一方面由p53诱导产生的mdm2又可以稳定p53蛋白的作用Another mechanism by which P53 promotes apoptosis is anchoring to the mitochondria directly in the cytoplasm, inducing mitochondria-dependent apoptosis. Studies have shown that p53 mitochondrial anchoring mainly acts through the E3 ligase murine double minute 2 (mdm2). As an important tumor suppressor gene, P53 can regulate the occurrence and development of tumor through various pathways. In the interaction with p53, mdm2 gene mediates the degradation of p53, inhibits the transcriptional activation of p53, and down-regulates its tumor growth inhibitory activity. On the other hand, mdm2 induced by p53 can stabilize the function of p53 protein.
促进细胞自我吞噬Promote cell self-phagocytosis
细胞调控损伤的自噬调节剂(damage-regulated autophagy modulator,DRAM)主要参与细胞的自我吞噬作用。DRAM内含有p53结合序列,是新发现的p53下游靶基因。DRAM在细胞营养缺乏时,能激活细胞自我吞噬功能,分解长效蛋白质,以稳定细胞的形态和维持细胞基本生命状态。DRAM在细胞受到刺激时,则诱导细胞凋亡。研究发现,当p53缺失而仅表达DRAM时,DRAM对细胞的杀伤作用仅为原有杀伤作用的2%~4%,而当DRAM与p53共转染时,其杀伤能力大大提高。因此得出结论,DRAM的诱导细胞程序性死亡的作用是依赖于p53发挥的,DRAM可依赖p53蛋白诱导细胞自噬性死亡。The cell-regulated autophagy modulator (DRAM) is mainly involved in the autophagy of cells. The DRAM contains a p53-binding sequence and is a newly discovered p53 downstream target gene. In the absence of cell nutrition, DRAM can activate the cell's self-phagocytic function and decompose long-acting proteins to stabilize the cell's morphology and maintain its basic life. DRAM induces apoptosis when cells are stimulated. The study found that when p53 is deleted and only DRAM is expressed, the killing effect of DRAM on cells is only 2% to 4% of the original killing effect, and when DRAM and p53 are co-transfected, the killing ability is greatly improved. Therefore, it is concluded that the role of DRAM in inducing programmed cell death is dependent on p53, which can induce autophagic death by p53.
血管生成抑制作用Angiogenesis inhibition
肿瘤发展到一定程度后,通过自分泌途径可以形成促血管生成因子,促进新生血管生成,有利于肿瘤快速生长,这种现象的产生是血小板凝集素-1(thrombospondin-1TSP-1)基因表达水平下降的结果。TSP-1是血管生成强有力的抑制因子,p53对TSP-1基因表达有上调作用,能激活内源性的TSP-1基因,正性调节TSP-1的启动子序列。研究还发现,p53可激活a(Ⅱ)胶原脯氨酰-4-羟化酶(a2PH)转录,致使全长胶原合成以及分泌,并产生抗血管生成抑素,在基质水平诱导蛋白质水解,增加胶原源性抗血管生成胶原的合成及分解。P53通过刺激抑制血管生成基因的表达,抑制肿瘤血管形成。同时,研究表明,p53 与血管内皮生长因子(VEGF)间也有明显的关联性,p53突变后能上调VEGF,增加微血管数量促使肿瘤血管生成。After the tumor develops to a certain extent, pro-angiogenic factors can be formed through the autocrine pathway, which promotes neovascularization and is conducive to rapid tumor growth. This phenomenon is caused by the expression level of thrombospondin-1 (TSP-1) gene. The result of the decline. TSP-1 is a potent inhibitor of angiogenesis. p53 up-regulates TSP-1 gene expression, activates the endogenous TSP-1 gene, and positively regulates the promoter sequence of TSP-1. The study also found that p53 can activate a (II) collagen prolyl-4-hydroxylase (a2PH) transcription, resulting in the synthesis and secretion of full-length collagen, and the production of anti-angiomotin, induced protein hydrolysis at the substrate level, increased Synthesis and decomposition of collagen-derived anti-angiogenic collagen. P53 inhibits tumor angiogenesis by inhibiting the expression of angiogenic genes by stimulation. At the same time, studies have shown that there is a significant correlation between p53 and vascular endothelial growth factor (VEGF). After p53 mutation, it can up-regulate VEGF and increase the number of microvessels to promote tumor angiogenesis.
DNA修复DNA repair
P53蛋白在DNA修复过程中起重要作用,主要表现在:若DNA损伤,p53蛋白阻止DNA复制,为DNA修复争取时间,若修复失败,p53蛋白则激活诱导细胞凋亡机制,促使细胞程序性死亡,以维持机体的稳定性。P53也可以于DNA修复因子如RPA、PCNA等基因相互作用,直接参与DNA的损伤修复过程。同时p53又可以和核酸切除修复基因(Nucleotide Excision Repair,NER)的组成成分相互作用,使NER也聚集于此,进行核苷酸修复。P53 protein plays an important role in DNA repair process, mainly in the case of DNA damage, p53 protein prevents DNA replication, and gains time for DNA repair. If repair fails, p53 protein activates apoptosis-inducing mechanism and promotes programmed cell death. To maintain the stability of the body. P53 can also interact with DNA repair factors such as RPA, PCNA and other genes directly involved in DNA damage repair process. At the same time, p53 can interact with the components of Nucleotide Excision Repair (NER) to make NER also aggregate here for nucleotide repair.
P53具有多种与DNA相互作用方式的能力,这使得p53可以直接参与DNA修复,如与DNA修复因子相结合,或通过p53蛋白-蛋白相互作用直接参与DNA修复,这些能力对p53与损伤DNA结合具有重要意义。在DNA损伤因子作用下,p53的C-端探测到损伤的DNA,与其结合形成p53-DNA复合物。P53与损伤DNA结合后起转录因子作用,与序列特异的DNA结合,通过反式激活靶基因,参与并增强DNA修复。P53 has multiple abilities to interact with DNA, allowing p53 to be directly involved in DNA repair, such as binding to DNA repair factors, or directly involved in DNA repair through p53 protein-protein interactions, which bind p53 to damaged DNA. It is of great significance. Under the action of DNA damage factors, the damaged DNA is detected at the C-terminus of p53, and binds to form a p53-DNA complex. P53 binds to damaged DNA and acts as a transcription factor, binds to sequence-specific DNA, and participates in and enhances DNA repair by trans-activating target genes.
抑制肿瘤细胞运动Inhibition of tumor cell movement
细胞迁移有赖于细胞丝状伪足的形成以及细胞外基质的完整性被破坏,细胞骨架和其结合蛋白是这一过程的物质基础。P53蛋白可负性调节细胞的铺展及纤维黏连蛋白的形成。Ras(P21)蛋白位于细胞膜内侧,它在传递细胞生长分化信号方面起重要作用。Ras可调节Ras同族基因家族成员A(Rashomolog gene fami ly member A,RhoA)的作用。一方面,Ras可使RhoA的膜锚定;另一方面,Ras促进p190Rho GTPase激活蛋白(Rho GAP)酪氨酸磷酸化,促进RhoA-GTP水解成无活性的RhoA-GDP,致使RhoA活性降低。P53是Ras促进Rho GAP磷酸化作用的必需因子,与RhoA的膜锚定作用无关。P53缺失后,由于p190Rho GAP磷酸化降低,降低了RhoA GTP载入,从而大大促进了细胞的运动能力。Cell migration depends on the formation of filopodia and the integrity of the extracellular matrix, and the cytoskeleton and its binding proteins are the material basis of this process. P53 protein negatively regulates cell spreading and fibronectin formation. The Ras (P21) protein is located inside the cell membrane and plays an important role in transmitting cell growth and differentiation signals. Ras regulates the role of Rasomolog gene fami ly member A (RhoA). On the one hand, Ras can anchor the membrane of RhoA; on the other hand, Ras promotes tyrosine phosphorylation of p190Rho GTPase activating protein (Rho GAP), which promotes the hydrolysis of RhoA-GTP to inactive RhoA-GDP, resulting in decreased RhoA activity. P53 is an essential factor for Ras to promote Rho GAP phosphorylation, independent of membrane anchoring of RhoA. After P53 deletion, the phosphorylation of p190Rho GAP is reduced, which reduces the loading of RhoA GTP, which greatly promotes the cell's ability to move.
TATTAT
人类免疫缺陷病毒Ⅰ型(HIV-Ⅰ)是获得性免疫缺陷综合征(AIDS)的病原体。Tat是由HIV-Ⅰ编码的一段富含碱性氨基酸的短肽,其序列为YGRKKRRQRRR(SEQ ID NO.:5第405-416位),是其编码的6个调控蛋白中的一个很重要的调控蛋白,而Tat转导结构域的核心区就是由这11个氨基酸残基所组成。作为新发现的一种蛋白转导结构域,Tat蛋白能高效地介导与其共价连接的DNA、多肽、蛋白质等分子进入几乎所有的组织和细胞,甚至可以通过血脑屏障,转导效率高且对细胞几乎没有损伤,并且能保持蛋白的生物活性。Tat融合蛋白系统被认为是一种很有前途的高效运载工具,在基础医学研究和临床治疗方面都有着非常广阔的应用前景。Human immunodeficiency virus type I (HIV-I) is the causative agent of acquired immunodeficiency syndrome (AIDS). Tat is a short peptide rich in basic amino acids encoded by HIV-I, and its sequence is YGRKKRRQRRR (SEQ ID NO.: 5, positions 405-416), which is one of the six regulatory proteins it encodes. The regulatory protein, and the core region of the Tat transduction domain is composed of these 11 amino acid residues. As a newly discovered protein transduction domain, Tat protein can efficiently mediate DNA, peptide, protein and other molecules covalently linked to it into almost all tissues and cells, and even pass the blood-brain barrier, and the transduction efficiency is high. It has almost no damage to cells and maintains the biological activity of the protein. The Tat fusion protein system is considered to be a promising and efficient delivery vehicle with broad application prospects in basic medical research and clinical treatment.
Tat蛋白的转导作用Transduction of Tat protein
Tat蛋白可以引导多种多肽和蛋白质进入几乎所有的靶细胞,这即是Tat蛋白的转导作用(transduction),又称为内化作用(internalization)。Tat蛋白的转导作用主要依赖于多肽或蛋白质的浓度,而不同于一般的通道、受体、内 吞作用的的入胞方式。由于具有这一功能,Tat可被用来作为介导外源蛋白通过细胞膜的运载工具,因此日益受到人们的重视。Tat protein can guide a variety of peptides and proteins into almost all target cells, which is the transduction of Tat protein, also known as internalization. The transduction of Tat protein depends mainly on the concentration of the polypeptide or protein, and is different from the general channel, receptor, and endocytosis mode of entry. Because of this function, Tat can be used as a carrier for mediating foreign proteins through cell membranes, and is therefore receiving increasing attention.
Tat蛋白以小窝途径介导外源物质进入细胞Tat protein mediates foreign matter into cells by cavern pathway
小窝(caveolae)是细胞膜上很小的凹陷,大约50-70nm。Eguchi对Tat蛋白介导的外源基因转导进行了制菌霉素试验,制菌霉素能够抑制外源物质以小窝途径进入细胞,研究发现Tat介导的外源蛋白转导受到了抑制,因此得出结论Tat通过破坏细胞质膜,以小窝的方式进入细胞。这种方式操作简单,不受外界因素如温度的影响,也不受其他体内环境的影响,不产生毒性,可直接应用与体内的细胞。这种方法的诱导效果明显高于其他转染方法,速度快,转导效率高,在转导过程中保留了蛋白的生物活性。用另一种抑制小窝途径的药物fillipin使小窝的一个必需成分小窝蛋白-1在细胞表面重新分布,影响小窝途径后,发现Tat介导的外源蛋白输送受到了抑制,证明Tat蛋白是通过小窝途径穿膜的。Caveolae is a small depression on the cell membrane, about 50-70 nm. Eguchi performed a bacteriomycin test on Tat-mediated exogenous gene transduction, which inhibited the entry of foreign substances into cells via a cavelet pathway, and found that Tat-mediated foreign protein transduction was inhibited. Therefore, it is concluded that Tat enters the cell in a small nest by destroying the plasma membrane. This method is simple to operate, free from external factors such as temperature, and is not affected by other internal environment, does not produce toxicity, and can be directly applied to cells in the body. The induction effect of this method is significantly higher than other transfection methods, the speed is fast, the transduction efficiency is high, and the biological activity of the protein is retained during the transduction process. Using another drug, fillipin, which inhibits the caveola pathway, redistributes a small fraction of caveolin-1 on the cell surface, affecting the cavelet pathway, and found that Tat-mediated foreign protein delivery is inhibited, proving Tat Protein is transmembrane through the cavern pathway.
融合蛋白的制备Preparation of fusion protein
生物技术的发展为重组人溶菌酶的生产提供了有效途径,一种典型的生产抗肿瘤融合蛋白的方法,包括步骤:The development of biotechnology provides an effective way to produce recombinant human lysozyme, a typical method for producing anti-tumor fusion proteins, including steps:
(a)在适合表达条件下,培养本发明第四方面所述的宿主细胞,从而表达本发明第一方面所述的抗肿瘤融合蛋白;(a) cultivating the host cell of the fourth aspect of the invention under conditions suitable for expression, thereby expressing the anti-tumor fusion protein of the first aspect of the invention;
(b)分离纯化出步骤(a)中所表达的抗肿瘤融合蛋白。(b) isolating and purifying the anti-tumor fusion protein expressed in the step (a).
在另一优选例中,所述宿主细胞为细菌,较佳地为大肠杆菌。In another preferred embodiment, the host cell is a bacterium, preferably E. coli.
在另一优选例中,本发明提供的GnRH+PII+P53的制备方法包括下列几个方面:In another preferred embodiment, the method for preparing GnRH+PII+P53 provided by the present invention includes the following aspects:
1)以GnRH+PII+P53氨基酸序列为基础,通过序列的结构优化以及密码子的简并性全基因合成了GnRH+PII+P53的编码基因,获得了该基因在大肠杆菌中的高表达;1) Based on the amino acid sequence of GnRH+PII+P53, the gene encoding GnRH+PII+P53 was synthesized by sequence optimization and codon degeneracy, and the high expression of this gene in E. coli was obtained.
2)在1)的基础上在GnRH+PII+P53编码序列前加入ULP1的识别位点98AA的SUMO,保证融合蛋白在经过ULP1的酶解后能够获得和GnRH+PII+P53理论设计一样的N端残基序列;2) Add the SUMO of the recognition site 98AA of ULP1 to the GnRH+PII+P53 coding sequence on the basis of 1) to ensure that the fusion protein can obtain the same theoretical design as GnRH+PII+P53 after ULP1 enzymatic hydrolysis. Terminal residue sequence;
3)在2)的基础上,加入了一段有利于表达和纯化的序列,最终通过NdeI和Hind III克隆到表达载体pET21中得到表达构建pET21-6xhis-SUMO-GnRH+PII+P53,命名为pET220-GnRH+PII+P53;3) On the basis of 2), a sequence which is favorable for expression and purification was added, and finally cloned into expression vector pET21 by NdeI and Hind III to obtain expression construct pET21-6xhis-SUMO-GnRH+PII+P53, named pET220 -GnRH+PII+P53;
4)表达载体pET220-GnRH+PII+P53转入大肠杆菌菌株BL21(DE3)RP plus中,得到工程菌;4) The expression vector pET220-GnRH+PII+P53 was transferred into E. coli strain BL21(DE3)RP plus to obtain an engineering strain;
5)工程菌在IPTG的诱导下表达融合蛋白6xhis-SUMO-GnRH+PII+P53,融合蛋白以包涵体形式表达;5) The engineered strain expresses the fusion protein 6xhis-SUMO-GnRH+PII+P53 under the induction of IPTG, and the fusion protein is expressed in the form of inclusion bodies;
6)融合表达的6xhis-SUMO-GnRH+PII+P53经过裂菌、包涵体溶解、金属螯合纯化等步骤得到纯化的融合蛋白;6) The fusion expression of 6xhis-SUMO-GnRH+PII+P53 is obtained by the steps of lysis, inclusion body dissolution, metal chelation purification and the like;
7)融合蛋白通过滴加到到复性液中进行复性;7) the fusion protein is renatured by dropwise addition to the renaturation fluid;
8)复性后的融合蛋白经过透析,酶切,亲和等层析步骤得到GnRH+PII+P53粗品;8) The fused protein fusion product is subjected to dialysis, enzyme digestion, affinity chromatography, etc. to obtain crude GnRH+PII+P53;
9)GnRH+PII+P53粗品通过分子筛精制纯化,最终得到纯度大于95%的GnRH+PII+P53,在SDS-PAGE电泳上显示为单一条带,在分子筛上表现为四聚体的蛋白;9) GnRH+PII+P53 crude product was purified by molecular sieve, and finally GnRH+PII+P53 with purity greater than 95% was obtained, which showed a single band on SDS-PAGE electrophoresis and tetramer protein on molecular sieve;
10)步骤9)中所获得GnRH+PII+P53纯品通过ISHIKAWA细胞检测生物学活性,在蛋白终浓度为0.5μg/ml以上时,>95%的细胞通过凋亡途径崩解。10) The pure GnRH+PII+P53 obtained in step 9) was tested for biological activity by ISHIKAWA cells, and >95% of the cells disintegrated through the apoptotic pathway at a final protein concentration of 0.5 μg/ml or more.
本发明的主要优点包括:The main advantages of the invention include:
(a)本发明融合蛋白不易降解,可以自然高效地形成四聚体。(a) The fusion protein of the present invention is not easily degraded, and a tetramer can be formed naturally and efficiently.
(b)本发明的融合蛋白(四聚体)可以高效的进入靶细胞,并快速地进入细胞核区域,诱导已突变细胞(如肿瘤细胞)凋亡。(b) The fusion protein (tetramer) of the present invention can efficiently enter target cells and rapidly enter the nuclear region to induce apoptosis of the mutant cells (such as tumor cells).
(c)本发明融合蛋白因P53的使用,动物实验未见毒性。(c) The fusion protein of the present invention has no toxicity in animal experiments due to the use of P53.
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,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。The invention is further illustrated below in conjunction with specific embodiments. It is to be understood that the examples are not intended to limit the scope of the invention. The experimental methods in the following examples which do not specify the specific conditions are usually in accordance with conventional conditions or according to the conditions recommended by the manufacturer. Percentages and parts are by weight unless otherwise stated.
实施例1Example 1
编码本发明GnRH+PII+P53基因序列的优化设计和全基因合成Optimization design and whole gene synthesis of the GnRH+PII+P53 gene sequence encoding the present invention
根据设计的GnRH+PII+P53氨基酸序列,通过基因软件分析,充分考虑了稀有密码子以及基因的二级结构,以及同义密码子的使用,最终得到优选的GnRH+PII+P53编码核酸序列如SEQ ID NO.:1所示,具体核酸序列为:According to the designed amino acid sequence of GnRH+PII+P53, through gene software analysis, the rare codon and the secondary structure of the gene, as well as the use of synonymous codon, were fully considered, and finally the preferred GnRH+PII+P53 encoding nucleic acid sequence was obtained. As shown in SEQ ID NO.: 1, the specific nucleic acid sequence is:
Figure PCTCN2019081735-appb-000003
Figure PCTCN2019081735-appb-000003
Figure PCTCN2019081735-appb-000004
Figure PCTCN2019081735-appb-000004
GnRH+PII+P53蛋白序列如SEQ ID NO.:2所示,具体蛋白序列为:The GnRH+PII+P53 protein sequence is shown in SEQ ID NO.: 2, and the specific protein sequence is:
Figure PCTCN2019081735-appb-000005
Figure PCTCN2019081735-appb-000005
在优化后的序列N端加入限制性内切酶NdeI位点和有利于表达和纯化的序列以及ulp1识别序列sumo的编码序列,在C端加入限制性内切酶Hind III位点,得到的序列如SEQ ID NO.:3所示,具体核酸序列为:The restriction endonuclease NdeI site and the sequence encoding the purified and purified sequence and the ulp1 recognition sequence sumo were added to the N-terminus of the optimized sequence, and the restriction endonuclease Hind III site was added at the C-terminus. As shown in SEQ ID NO.: 3, the specific nucleic acid sequence is:
Figure PCTCN2019081735-appb-000006
Figure PCTCN2019081735-appb-000006
Figure PCTCN2019081735-appb-000007
Figure PCTCN2019081735-appb-000007
通过商业化基因合成公司上海生工生物工程有限公司全基因合成该序列,并克隆到PUC19载体中,经测序验证后命名为PUC19-GnRH+PII+P53。The sequence was synthesized by commercial gene synthesis company Shanghai Shenggong Bioengineering Co., Ltd. and cloned into PUC19 vector. After sequencing, it was named as PUC19-GnRH+PII+P53.
实施例2Example 2
表达GnRH+PII+P53的载体构建Construction of vector expressing GnRH+PII+P53
GnRH+PII+P53在大肠杆菌中表达的优先表达载体为PET21,将Pet21和PUC19-GnRH+PII+P53用限制性内切酶Nde I和Hind III做双酶切,酶切产物用琼脂糖凝胶电泳分离后切胶回收,再用T4连接酶连接,连接产物转化到大肠杆菌DH5α中。挑出阳性克隆,抽出其中的质粒,经过测序验证无误后命名为pET220-GnRH+PII+P53。The preferred expression vector for expression of GnRH+PII+P53 in E. coli was PET21, and Pet21 and PUC19-GnRH+PII+P53 were digested with restriction endonucleases Nde I and Hind III, and the digested products were agarose-condensed. After gel electrophoresis separation, the gel was recovered, and then ligated with T4 ligase, and the ligated product was transformed into Escherichia coli DH5α. A positive clone was picked and the plasmid was extracted, and after sequencing, it was named pET220-GnRH+PII+P53.
pET220-GnRH+PII+P53蛋白序列如SEQ ID NO.:4所示,具体蛋白序列为:The pET220-GnRH+PII+P53 protein sequence is shown in SEQ ID NO.: 4, and the specific protein sequence is:
Figure PCTCN2019081735-appb-000008
Figure PCTCN2019081735-appb-000008
实施例3Example 3
表达GnRH+PII+P53蛋白的工程菌构建及诱导表达分析Construction of engineered bacteria expressing GnRH+PII+P53 protein and its induced expression analysis
将质粒pET220-GnRH+PII+P53转化到大肠杆菌宿主BL21Codon Plus(DE3)RP中,离心后取细菌涂布在含100μg/ml的氨苄青霉素的LB平板上, 37℃培养过夜,在LB平板上挑单克隆培养,经过菌落PCR鉴定,得到单克隆转化子pET220-GnRH+PII+P53/BL21Codon Plus(DE3)RP得到的单克隆转化子pET220-GnRH+PII+P53/BL21Codon Plus(DE3)RP接种于3ml的含100μg/ml的氨苄青霉素的LB培养液中,37℃培养过夜;按2%的接种量取过夜培养的大肠杆菌接种到新鲜的LB培养液中37℃培养,当细菌密度OD600值达到0.6-1.0之间时,加入终浓度为0.5mM的IPTG诱导融合蛋白表达,继续培养3.5小时后,收集菌体。通过SDS-PAGE分析蛋白表达情况。The plasmid pET220-GnRH+PII+P53 was transformed into E. coli host BL21 Codon Plus (DE3) RP. After centrifugation, the bacteria were coated on LB plates containing 100 μg/ml ampicillin and cultured overnight at 37 ° C on LB plates. Monoclonal culture was carried out and identified by colony PCR to obtain monoclonal transformant pET220-GnRH+PII+P53/BL21Codon Plus(DE3)RP inoculated with monoclonal transformant pET220-GnRH+PII+P53/BL21Codon Plus(DE3)RP. Incubate in 3 ml of LB medium containing 100 μg/ml ampicillin at 37 ° C overnight; inoculate E. coli in 2% inoculation amount into fresh LB medium and incubate at 37 ° C, when the bacteria density is OD600 When the ratio was between 0.6 and 1.0, the expression of the fusion protein was induced by adding IPTG at a final concentration of 0.5 mM, and after continuing to culture for 3.5 hours, the cells were collected. Protein expression was analyzed by SDS-PAGE.
结果如图1所示,融合蛋白以包涵体形式表达,融合蛋白表达量占细菌总蛋白的25%左右。The results are shown in Figure 1. The fusion protein is expressed in the form of inclusion bodies, and the expression of the fusion protein accounts for about 25% of the total bacterial protein.
实施例4Example 4
GnRH+PII+P53蛋白的发酵生产与分离纯化Fermentation, purification and purification of GnRH+PII+P53 protein
1、发酵生产1. Fermentation production
将pET220-GnRH+PII+P53/BL21Codon Plus(DE3)RP接种于LB培养基中37℃培养过夜,第二天按2%的接种量接入新鲜的TB(甘油5克/升,蛋白胨12克/升,酵母膏24克/升,K2HPO4 12.54克/升,KH2PO42.31克/升)发酵液体培养液中,37℃培养到细菌OD600值达到1,加入终浓度为0.5mM的IPTG诱导融合蛋白表达,继续培养4小时后收集菌体。pET220-GnRH+PII+P53/BL21Codon Plus(DE3)RP was inoculated in LB medium and cultured overnight at 37 °C. The next day, 2% of the inoculum was used to access fresh TB (glycerol 5 g/L, peptone 12 g). / liter, yeast extract 24 g / liter, K2HPO4 12.54 g / liter, KH2PO42.31 g / liter) in the fermentation liquid culture medium, cultured at 37 ° C until the bacterial OD600 value reached 1, adding a final concentration of 0.5 mM IPTG induced fusion protein Expression, the cells were collected after 4 hours of incubation.
2、包涵体获取2, inclusion body acquisition
融合蛋白以包涵体形式表达,为了获得包涵体,用PBS按1:10的比例重悬菌体,设置匀浆压力为750pa,匀浆两次,15000g的离心条件下离心收集沉淀部分;绝大部分包涵体蛋白在收集的沉淀部分中。The fusion protein was expressed in the form of inclusion bodies. In order to obtain inclusion bodies, the cells were resuspended in a ratio of 1:10 with PBS, and the homogenate pressure was set to 750 Pa, homogenized twice, and the precipitated fraction was collected by centrifugation at 15,000 g; Part of the inclusion body protein is in the precipitated fraction of the collection.
3、融合蛋白的溶解和纯化3. Dissolution and purification of fusion protein
包涵体按1:20的比例用溶液1(20Mm Tris-HCL,500mM NaCl,20mM Imidazole,8M Urea 20mM 2-Mercaptoethanol pH8.0)充分溶解,15000g的离心条件下离心20分钟,取上清组分做进一步的纯化The inclusion bodies were fully dissolved in a ratio of 1:20 with solution 1 (20 Mm Tris-HCL, 500 mM NaCl, 20 mM Imidazole, 8 M Urea 20 mM 2-Mercaptoethanol pH 8.0), centrifuged at 15000 g for 20 minutes under centrifugation, and the supernatant fraction was taken. Further purification
上清组分通过NI 2+metal chelating chromatography在变性条件下纯化得纯的融合蛋白,融合蛋白处在含8M Urea的变性溶液中,加入终浓度为5mM的DTT处理融合蛋白,室温下搅拌过夜 The supernatant fraction was purified by denaturing conditions by NI 2+ metal chelating chromatography. The fusion protein was placed in a denaturation solution containing 8 M Urea, and the fusion protein was treated with DTT at a final concentration of 5 mM, and stirred at room temperature overnight.
4、变性蛋白的重折叠(蛋白复性)4. Refolding of denatured proteins (protein renaturation)
经过处理的变性蛋白通过滴加到复性液中(复性液配方为100mM Tris-HCL,500mM NaCl,0.5M Arginine,1%triton X-100,10%glycerol,1mM EDTA,1mM GSH,0.5mM GSSG pH8.0)滴加完后4℃冰箱中放置48-72小时,充分复性The treated denatured protein was added dropwise to the reconstitution solution (reconstitution solution was 100 mM Tris-HCL, 500 mM NaCl, 0.5 M Arginine, 1% triton X-100, 10% glycerol, 1 mM EDTA, 1 mM GSH, 0.5 mM). GSSG pH 8.0) After the addition is completed, place in the refrigerator at 4 ° C for 48-72 hours, fully renatured.
5、复性后蛋白的回收5. Recovery of protein after renaturation
充分复性的蛋白溶液装入透析袋对缓冲液(20Mm Tris.CL,Ph8.5)透析,每次透析4小时,总共透析3次;透析后,用阴离子柱Q回收目的蛋白The fully refolded protein solution was placed in a dialysis bag and dialyzed against a buffer (20 Mm Tris. CL, Ph 8.5) for 4 hours each time for a total of 3 times of dialysis; after dialysis, the target protein was recovered by an anion column Q.
6、融合蛋白肠ULP1酶切6, fusion protein intestinal ULP1 digestion
阴离子柱Q回收的融合蛋白中加入20IU/mg融合蛋白的ULP1酶,加入磁力搅拌子缓慢搅拌,保证充分酶切,酶切条件:4℃,16-24小时The ULP1 enzyme of 20IU/mg fusion protein was added to the fusion protein recovered by anion column Q, and stirred slowly by adding a magnetic stirrer to ensure sufficient digestion. The enzyme digestion conditions were: 4 ° C, 16-24 hours.
7、酶切后纯化7. Purification after digestion
经过酶切后的融合蛋白溶液,主要为二种成分:融合表达标签蛋白和GnRH+PII+P53,以及少量未酶切的完整融合蛋白,其中融合表达标签蛋白和完整融合蛋白都含有6xHis纯化标签,通过NI 2+metal chelating chromatography可以结合这两部分蛋白,GnRH+PII+P53不能结合到NI 2+metal chelating柱而存在于流穿部分中 The digested fusion protein solution is mainly composed of two components: fusion expression tag protein and GnRH+PII+P53, and a small amount of undigested intact fusion protein, wherein the fusion expression tag protein and the intact fusion protein both contain a 6xHis purification tag. The two parts of the protein can be combined by NI 2+ metal chelating chromatography, and GnRH+PII+P53 cannot be bound to the NI 2+ metal chelating column and exists in the flow-through portion.
8、GnRH+PII+P53的肝素亲和层析浓缩、分子筛精制8. Heparin affinity chromatography of GnRH+PII+P53, molecular sieve refining
用购自GE healthcare的肝素亲和填料装填的层析柱亲和浓缩存在于(步骤7)流穿部分的GnRH+PII+P53,浓缩后的GnRH+PII+P53通过SUPERDEX 200精制,精制后的蛋白溶液无菌过滤后供后续实验使用。The column was filled with GnRH+PII+P53 in the flow-through portion (step 7) by affinity chromatography packed with heparin affinity packing purchased from GE Healthcare, and the concentrated GnRH+PII+P53 was refined by SUPERDEX 200. The protein solution is sterile filtered and used for subsequent experiments.
实施例5Example 5
GnRH+PII+P53对不同肿瘤细胞的杀伤效果测定Killing effect of GnRH+PII+P53 on different tumor cells
用实施例4制得的GnRH+PII+P53融合蛋白处理下表2中的各细胞,测定IC50值。Each of the cells in Table 2 below was treated with the GnRH+PII+P53 fusion protein prepared in Example 4, and the IC50 value was determined.
结果如表2所示,GnRH+PII+P53对正常细胞没有杀伤效果IC50>100μg/ml,GnRH+PII+P53对多种肿瘤细胞均具有较好的杀伤效果,IC50基本均小于5μg/ml。The results are shown in Table 2. GnRH+PII+P53 has no killing effect on normal cells with IC50>100μg/ml, and GnRH+PII+P53 has good killing effect on various tumor cells. The IC50 is less than 5μg/ml.
表2 GnRH+PII+P53对不同肿瘤细胞的杀伤效果Table 2 Killing effect of GnRH+PII+P53 on different tumor cells
细胞中文名称Cell Chinese name 细胞系Cell line IC50(μg/ml)IC50 (μg/ml)
血管内皮细胞(正常细胞)Vascular endothelial cells (normal cells) HAoSMCHAoSMC >100>100
阴道(正常细胞)Vagina (normal cells) VK2/E6E7VK2/E6E7 >100>100
人肺(正常细胞)Human lung (normal cell) WI-38WI-38 >100>100
人胚肾(正常细胞)Human embryonic kidney (normal cell)   >100>100
非洲狨猴肾(正常细胞)African colobus monkey kidney (normal cell) VeroVero >100>100
食管癌Esophageal cancer KYSE-150KYSE-150 2.60682.6068
乳腺癌Breast cancer MDA-MB-231MDA-MB-231 4.62184.6218
乳腺癌Breast cancer CAL120CAL120 1.013761.01376
乳腺癌Breast cancer HCCC1954HCCC1954 4.90324.9032
卵巢癌Ovarian cancer SK-OV-3SK-OV-3 23.21223.212
卵巢癌Ovarian cancer SW626SW626 3.1643.164
卵巢癌Ovarian cancer A2780A2780 1.86711.8671
宫颈癌Cervical cancer HeLaHeLa 4.91894.9189
宫颈癌Cervical cancer HeLa229HeLa229 3.52063.5206
结直肠癌Colorectal cancer SW480SW480 2.3732.373
结直肠癌Colorectal cancer NCI-H716NCI-H716 0.52620.5262
结直肠癌Colorectal cancer COLO205COLO205 0.74270.7427
肝癌Liver cancer HuCCT1HuCCT1 3.86093.8609
肝癌Liver cancer HepG2HepG2 2.73282.7328
子宫内膜癌Endometrial cancer IshikawaIshikawa 0.35440.3544
子宫内膜癌Endometrial cancer HEC-1BHEC-1B 0.1520.152
子宫平滑肌瘤Uterine leiomyoma SK-UT-1SK-UT-1 10.56210.562
子宫肉瘤Uterine sarcoma MES-SAMES-SA 2.4642.464
绒毛膜癌Choriocarcinoma JEG-3JEG-3 0.54630.5463
肾癌Kidney cancer A498A498 0.9380.938
肾上腺皮质癌Adrenal cortical carcinoma SW13SW13 0.20380.2038
肺鳞癌Squamous cell carcinoma NCI-H520NCI-H520 0.91050.9105
肺癌Lung cancer A549A549 5.4825.482
膀胱癌Bladder Cancer 56375637 3.8633.863
前列腺癌Prostate cancer DU145DU145 2.46732.4673
前列腺癌Prostate cancer RWPE-1RWPE-1 0.7220.722
舌癌Tongue cancer CAL-27CAL-27 0.15080.1508
舌癌Tongue cancer SCC-4SCC-4 1.36571.3657
舌癌Tongue cancer OSC-19OSC-19 0.74530.7453
舌癌Tongue cancer SCC-25SCC-25 0.38070.3807
舌癌Tongue cancer SCC-15SCC-15 0.84360.8436
实施例6Example 6
GnRH+PII+P53对Ishikawa细胞的杀伤效果测定Determination of the killing effect of GnRH+PII+P53 on Ishikawa cells
以实施例4制得的GnRH+PII+P53融合蛋白Slg001为例,说明其对Ishikawa细胞的杀伤效果,检测结果如图2所示,高浓度细胞致死,无正常细胞,仅可见培养基中大量颗粒碎片。具体如下:The GnRH+PII+P53 fusion protein Slg001 prepared in Example 4 was used as an example to demonstrate its killing effect on Ishikawa cells. The detection results are shown in Figure 2. The high concentration of cells is lethal, and there are no normal cells. Particle fragments. details as follows:
Ishikawa细胞Slg001处理72小时后,20μg/ml Slg001处理的孔板中培养基呈现红色,显微观察可见大量颗粒状细胞碎片,10μg/ml Slg001处理的孔板中培养基呈现浅红色,显微观察可见部分细胞和细胞碎片,低浓度Slg001处理的细胞则和对照组差异不大;After 72 hours of treatment with Igkawa cells Slg001, the medium in the 20μg/ml Slg001 treated plate was red, and a large number of granular cell debris were observed by microscopic observation. The medium in the 10μg/ml Slg001 treated plate was light red, microscopic observation Some cells and cell debris were observed, and the cells treated with low concentration of Slg001 were not significantly different from the control group;
随着处理时间延长至8天,不同与对照组细胞,0.4-20μg/ml Slg001处理的细胞都有不同程度的细胞碎片出现。空白对照组培养基在5%CO 2下呈黄色,pH为6.0,而20μg/ml Slg001处理的细胞在8天后培养基呈紫红色,pH在8.0左右。 As the treatment time was extended to 8 days, cells treated with 0.4-20 μg/ml Slg001 had different degrees of cell debris in different control cells. The blank control medium was yellow under 5% CO 2 and the pH was 6.0, while the cells treated with 20 μg/ml Slg001 were purple-red after 8 days and the pH was around 8.0.
实施例7Example 7
GnRH+PII+P53对接种舌鳞癌CAL-27的荷瘤鼠静脉给药有效性试验Effectiveness of GnRH+PII+P53 in intravenous administration of tumor-bearing mice inoculated with tongue squamous cell carcinoma CAL-27
对3组接种了CAL-27荷瘤鼠静脉施用实施例4制得的GnRH+PII+P53融合蛋白,隔天给药,共给药五次,剂量分别是250μg/kg、500μg/kg、1000μg/kg、2000μg/kg、4000μg/kg,观察荷瘤鼠是否有毒性反应,药物是否有效。结果是,未见毒性反应,前4针未见瘤体增大,第5针后测定抑瘤率达72%。The GnRH+PII+P53 fusion protein prepared in Example 4 was intravenously administered to three groups of CAL-27 tumor-bearing mice, and administered every other day for five times. The doses were 250 μg/kg, 500 μg/kg, and 1000 μg, respectively. /kg, 2000μg/kg, 4000μg/kg, observe whether the tumor-bearing mice have toxic reaction and whether the drug is effective. As a result, no toxic reaction was observed, and the tumor was not enlarged in the first 4 needles, and the tumor inhibition rate was 72% after the fifth needle.
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。All documents mentioned in the present application are hereby incorporated by reference in their entirety in their entireties in the the the the the the the the In addition, it should be understood that various modifications and changes may be made by those skilled in the art in the form of the appended claims.

Claims (10)

  1. 一种抗肿瘤融合蛋白,其特征在于,所述的融合蛋白具有式I所述结构:An anti-tumor fusion protein, characterized in that the fusion protein has the structure of formula I:
    D-A-B-C-E           (I)D-A-B-C-E (I)
    其中,among them,
    A为GnRH蛋白元件;A is a GnRH protein element;
    B为PEA蛋白的跨膜转运区或为无;B is the transmembrane transport region of the PEA protein or is absent;
    C为P53蛋白元件;C is a P53 protein element;
    E为TAT蛋白元件或为无;E is a TAT protein element or is absent;
    D为任选的信号肽和/或前导肽序列;D is an optional signal peptide and/or leader peptide sequence;
    并且,B和E不同时为无;Also, B and E are not the same at the same time;
    其中,“-”表示连接上述各元件的肽键。Here, "-" means a peptide bond connecting the above elements.
  2. 如权利要求1所述的融合蛋白,其特征在于,所述的融合蛋白具有式II所述结构:The fusion protein according to claim 1, wherein said fusion protein has the structure of formula II:
    D-A-B-C          (II)D-A-B-C (II)
    其中,among them,
    A为GnRH蛋白元件;A is a GnRH protein element;
    B为PEA蛋白的跨膜转运区;B is a transmembrane transport region of PEA protein;
    C为P53蛋白元件;C is a P53 protein element;
    D为任选的信号肽和/或前导肽序列;D is an optional signal peptide and/or leader peptide sequence;
    “-”表示连接上述各元件的肽键。"-" indicates a peptide bond connecting the above elements.
  3. 如权利要求1所述的融合蛋白,其特征在于,所述的GnRH蛋白包括II型GnRH蛋白和I型GnRH蛋白。The fusion protein according to claim 1, wherein said GnRH protein comprises a type II GnRH protein and a type I GnRH protein.
  4. 如权利要求1所述的融合蛋白,其特征在于,所述的融合蛋白是由细菌,较佳地由大肠杆菌表达的重组蛋白。The fusion protein according to claim 1, wherein the fusion protein is a recombinant protein expressed by bacteria, preferably E. coli.
  5. 一种分离的多核苷酸,其特征在于,所述的多核苷酸编码权利要求1所述的融合蛋白。An isolated polynucleotide, characterized in that the polynucleotide encodes the fusion protein of claim 1.
  6. 一种载体,其特征在于,所述载体含有权利要求5所述的多核苷酸。A vector comprising the polynucleotide of claim 5.
  7. 一种宿主细胞,其特征在于,所述宿主细胞含有权利要求6所述的载体或基因组中整合有权利要求5所述的多核苷酸。A host cell comprising the vector of claim 6 or a polynucleotide in which the polynucleotide of claim 5 is integrated.
  8. 一种生产抗肿瘤融合蛋白的方法,其特征在于,包括步骤:A method for producing an anti-tumor fusion protein, comprising the steps of:
    (a)在适合表达条件下,培养权利要求7所述的宿主细胞,从而表达权利要 求1所述的融合蛋白;(a) cultivating the host cell of claim 7 under conditions suitable for expression, thereby expressing the fusion protein of claim 1;
    (b)分离纯化出步骤(a)中所表达的抗肿瘤融合蛋白。(b) isolating and purifying the anti-tumor fusion protein expressed in the step (a).
  9. 一种药物组合物,其特征在于,所述药物组合物含有权利要求1所述的融合蛋白,以及药学上可接受的载体或赋形剂。A pharmaceutical composition comprising the fusion protein of claim 1 and a pharmaceutically acceptable carrier or excipient.
  10. 一种权利要求1所述的抗肿瘤融合蛋白的用途,其特征在于,用于制备治疗或预防肿瘤的药物。Use of an anti-tumor fusion protein according to claim 1, which is for use in the preparation of a medicament for treating or preventing a tumor.
PCT/CN2019/081735 2018-04-08 2019-04-08 Anti-tumor fusion protein, preparation method therefor and application thereof WO2019196790A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201980020574.6A CN112292402B (en) 2018-04-08 2019-04-08 Anti-tumor fusion protein and preparation method and application thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201810306907.9 2018-04-08
CN201810306907.9A CN110357968B (en) 2018-04-08 2018-04-08 Anti-tumor fusion protein and preparation method and application thereof

Publications (1)

Publication Number Publication Date
WO2019196790A1 true WO2019196790A1 (en) 2019-10-17

Family

ID=68163494

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/081735 WO2019196790A1 (en) 2018-04-08 2019-04-08 Anti-tumor fusion protein, preparation method therefor and application thereof

Country Status (2)

Country Link
CN (2) CN110357968B (en)
WO (1) WO2019196790A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116023512A (en) * 2023-02-16 2023-04-28 浙江大学 Polypeptide of target DNA damage repair protein and application thereof

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110357968B (en) * 2018-04-08 2023-08-25 吉林省汇融生物科技有限公司 Anti-tumor fusion protein and preparation method and application thereof
CN116735878B (en) * 2023-06-06 2024-03-19 北京三品医疗科技有限公司 Use of polypeptides in diagnosing cancer

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101684472A (en) * 2008-09-26 2010-03-31 中国人民解放军军事医学科学院毒物药物研究所 Recombinant fusion proteins containing p53 genes, recombinant and application thereof

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6258782B1 (en) * 1998-05-20 2001-07-10 Trimeris, Inc. Hybrid polypeptides with enhanced pharmacokinetic properties
WO2006029078A2 (en) * 2004-09-07 2006-03-16 The Regents Of The University Of California Targeting transducible molecules to specific cell types
CN1966529A (en) * 2006-06-21 2007-05-23 深圳市高科生物科技有限公司 Recombinant toxin agent containing gonadorelin and pyocyanin functional frament
CN101557817A (en) * 2006-09-14 2009-10-14 人类基因科学公司 Albumin fusion proteins
CN101070547B (en) * 2007-05-15 2010-12-22 中国人民解放军军事医学科学院军事兽医研究所 Recombined protein carrier PEA II-HphA of actively carrying exogenic gene, its preparing method and use
CN101343328B (en) * 2008-08-25 2011-06-01 北京博翱泰生物技术有限公司 Target-specificity dual-mutant amalgamation protein
CN101987873A (en) * 2009-08-07 2011-03-23 普罗特奥姆技术公司 P53 fusion protein and application thereof
CN101880327A (en) * 2010-02-09 2010-11-10 沈阳药科大学 Scorpion arialgesic anti-tumoral peptide fusion and acquisition method thereof
CN102603897B (en) * 2011-12-08 2014-10-08 山东省科学院生物研究所 Fusion protein containing guide peptide and GnRH-PE39KDEL as well as nucleic acid and application thereof
EP4052717A1 (en) * 2012-03-13 2022-09-07 Celularity Inc. Modified erythrocyte precursor cells and uses thereof
CN103805621B (en) * 2013-12-21 2016-07-06 中国人民解放军军事医学科学院军事兽医研究所 The novel preparation process of targeting antineoplastic amalgamation protein matter LPO
CN106117352B (en) * 2016-07-05 2020-10-09 中国人民解放军军事医学科学院军事兽医研究所 Human single-chain antibody E3-scFv resisting botulinum toxin type A and application thereof
CN110357968B (en) * 2018-04-08 2023-08-25 吉林省汇融生物科技有限公司 Anti-tumor fusion protein and preparation method and application thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101684472A (en) * 2008-09-26 2010-03-31 中国人民解放军军事医学科学院毒物药物研究所 Recombinant fusion proteins containing p53 genes, recombinant and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
RYU J., ET AL.: "Intracellular Delivery of p53 Fused to the Basic Domain of HIV-1 Tat", MOL. CELL, 31 December 2004 (2004-12-31), XP008110919 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116023512A (en) * 2023-02-16 2023-04-28 浙江大学 Polypeptide of target DNA damage repair protein and application thereof

Also Published As

Publication number Publication date
CN110357968B (en) 2023-08-25
CN112292402B (en) 2023-09-19
CN110357968A (en) 2019-10-22
CN112292402A (en) 2021-01-29

Similar Documents

Publication Publication Date Title
WO2019196790A1 (en) Anti-tumor fusion protein, preparation method therefor and application thereof
AU2018208708A1 (en) Delivery system for functional nucleases
US20080274956A1 (en) Fusion Protein Comprising a Bh3-Domain of a Bh3-Only Protein
JP2019147792A (en) Methods and compositions for treatment of cancer
JPH07503617A (en) Transport polypeptide derived from TAT
KR102050621B1 (en) Rtrail mutant and monomethyl auristatin e conjugate thereof
US9145446B2 (en) Conjugate comprising P21 protein for the treatment of cancer
CN106589131B (en) Fusion protein 4D5Fv-PE25, and preparation method and application thereof
US20160222362A1 (en) Target-Specific Double-Mutant Fusion Protein and Preparation Process Therefor
WO1998007858A1 (en) HUMAN POLYHOMEOTIC 1(hphl) ACTS AS A TUMOR SUPPRESSOR
CN102993314B (en) Anti-tumor fusion protein, as well as preparation method and application thereof
CN109400711B (en) PDGFR beta targeting tumor necrosis factor related apoptosis inducing ligand variant and preparation method and application thereof
US11566045B2 (en) Tumor targeting polypeptide and method of use thereof
WO2019011293A1 (en) Saponin compound targeting pd-1 and application thereof
US20230226157A1 (en) Anti-tumor fusion protein, preparation method therefor and application thereof
WO2022241167A1 (en) Delivery constructs derived from bacterial toxins and uses thereof
EP2708241A1 (en) Recombinant Fc-fusion protein of the two Immunoglobulin domains of UNC5
EP3762401A2 (en) Combination of depletes tregs and a checkpoint inhibitor
WO2002085305A2 (en) Compositions and methods for inducing cancer cell death
WO2015069044A1 (en) Fusion protein comprising p53 protein and p15 protein bound to each other, and composition containing the same for preventing or treating cancer
WO2011079431A1 (en) Fusion protein with telomerase inhibiting activity, preparation method and use thereof
US7838000B2 (en) Inhibition of pathogenic agents including α6β1 integrin receptor or α6β4 integrin receptor at a surface
WO2012112869A2 (en) Targeted cancer therapy using ets transcriptional inhibitors
CN116284321A (en) Cell penetrating peptide and application thereof
Villiers et al. 22. Engineering of an HB-EGF Inhibitor from the Diphtheria Toxin Receptor–Binding Domain for the Treatment of HB-EGF-Related Diseases

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: 19786100

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: 19786100

Country of ref document: EP

Kind code of ref document: A1