WO2012122942A1 - 抗乙型肝炎病毒x蛋白多肽药物 - Google Patents

抗乙型肝炎病毒x蛋白多肽药物 Download PDF

Info

Publication number
WO2012122942A1
WO2012122942A1 PCT/CN2012/072383 CN2012072383W WO2012122942A1 WO 2012122942 A1 WO2012122942 A1 WO 2012122942A1 CN 2012072383 W CN2012072383 W CN 2012072383W WO 2012122942 A1 WO2012122942 A1 WO 2012122942A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
polypeptide
cells
hepatitis
acid sequence
Prior art date
Application number
PCT/CN2012/072383
Other languages
English (en)
French (fr)
Inventor
张晓东
叶丽虹
Original Assignee
天津托普泰克生物科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 天津托普泰克生物科技有限公司 filed Critical 天津托普泰克生物科技有限公司
Publication of WO2012122942A1 publication Critical patent/WO2012122942A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • 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
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • 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
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing 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
    • C12N2730/00Reverse transcribing DNA viruses
    • C12N2730/00011Details
    • C12N2730/10011Hepadnaviridae
    • C12N2730/10111Orthohepadnavirus, e.g. hepatitis B virus
    • C12N2730/10122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • the present invention relates to the field of polypeptide medicines, and in particular to polypeptides against hepatitis B virus X protein and polynucleotides encoding the same, and their use.
  • Liver cancer is one of the malignant tumors that cause death of a patient, and its malignancy is high. According to statistics, in China, the mortality rate of liver cancer is second only to gastric cancer, ranking second in the mortality rate of malignant tumors. The number of new liver cancers in China is 300,000 per year, and the number of deaths from liver cancer is 110,000 per year.
  • Hepatitis B virus (HBV) infection can lead to hepatitis, cirrhosis and primary liver cancer.
  • HBV Hepatitis B virus
  • HBV is a DNA virus of about 3.2 kb in length, and its open reading frame expresses hepatitis B virus surface antigen (HBsAg), hepatitis B virus core antigen (HBcAg), hepatitis B virus polymerase and hepatitis B virus X antigen (HBxAg).
  • HBx hepatitis B virus X protein
  • HBx is a factor necessary for HBV DNA replication. Therefore, inhibition of the function of HBx means inhibition of HBV infection, and thus hepatitis and cirrhosis.
  • HBx acts as a trans-acting factor to promote the growth and proliferation of liver cancer, which is called oncoprotein.
  • HBx has a strong role in promoting proliferation and migration of liver cancer cells.
  • Transgenic mouse experiments have also demonstrated that HBx has a significant role in promoting the development of liver cancer.
  • a large number of studies have shown that the persistence of HBV infection can lead to chronic liver disease, including chronic hepatitis, chronic cirrhosis caused by the proliferation of fibrous connective tissue in liver tissue caused by recurrent episodes of chronic hepatitis, and liver cancer that occurs on the basis of cirrhosis.
  • HBx plays an important role in the development of chronic liver disease, including hepatitis, cirrhosis and liver cancer. Therefore, HBx is an important target for the prevention and treatment of liver diseases.
  • liver cancer is mainly based on surgery, supplemented by interventional therapy, and the chemotherapy effect is not satisfactory.
  • the positive detection rate of HBsAg and HBxAg in clinically operated liver cancer tissues is as high as 80% or even 90%. Therefore, since HBx is an important pathogenic factor for the development of liver cancer, the discovery and development of its specific inhibitors has important theoretical significance and practical clinical application value.
  • the three-dimensional conformational analysis of HBx is not yet completed, it is difficult to design its chemical inhibitors by the stereoscopic three-dimensional conformation of HBx.
  • Polypeptide fragments are useful as drugs and are widely used clinically. For example, thymosin
  • thymopeptide is a thymopentin extracted from the calf thymus. It has the function of promoting lymphocyte transformation and enhancing macrophage phagocytic activity, and can be used for treating various immunodeficiency diseases.
  • Peptide drugs are characterized by clear pharmacological effects, high safety, and ease of production. However, the discovery of multi-peptide drugs is difficult, and most of the polypeptide fragments have a short half-life in vivo, which directly affects the pharmacodynamic effect. Summary of the invention
  • the present invention relates to a polypeptide having a functional activity for inhibiting hepatitis B virus X protein (HBX), which inhibits HBx activity at a molecular level, a cellular level and an overall level, thereby inhibiting hepatitis B Hepatitis caused by viral infection, cirrhosis caused by recurrent hepatitis, and liver cancer that occurs on the basis of cirrhosis.
  • HBX hepatitis B virus X protein
  • the polypeptide and its peptidomimetics, including functional fragments and functional variants thereof, and genes encoding these polypeptides, peptidomimetics or functional fragments thereof, functional variants are widely used for the prevention and treatment of hepatitis B infection.
  • the invention provides an isolated polypeptide or peptidomimetic comprising the amino acid sequence set forth in SEQ ID NO: 1, or a functional fragment or functional variant thereof, the polypeptide or peptidomimetic having inhibition of hepatitis B
  • the function of the virus X protein and can inhibit the occurrence and development of chronic liver disease after hepatitis B virus infection.
  • the chronic liver disease after the hepatitis B virus infection may include hepatitis, cirrhosis caused by recurrent hepatitis, and liver cancer which occurs on the basis of cirrhosis.
  • the polypeptide or peptidomimetic comprising an amino acid sequence, a functional fragment or a functional variant thereof, may have at least 70%, or 80%, of the amino acid sequence set forth in SEQ ID NO: 1. , or 90%, and even higher identity.
  • these polypeptides or peptidomimetics comprise any of the amino acid sequences set forth in SEQ ID NOs: 1-6.
  • the invention provides an isolated polynucleotide comprising a polynucleotide encoding an amino acid sequence set forth in SEQ ID NO: 1, a functional fragment or a functional variant thereof; A polynucleotide in which the amino acid sequence shown in SEQ ID NO: 1, a functional fragment or a functional variant thereof is complementary or strictly hybridized.
  • these polynucleotides are polynucleotides encoding the amino acid sequence set forth in SEQ ID NOs: 1-6, or are complementary or strictly hybridized to a polynucleotide encoding the amino acid sequence set forth in SEQ ID NOs: 1-6. Polynucleotide.
  • the present invention provides a recombinant expression vector comprising an exogenous polynucleotide comprising a polynucleotide encoding the amino acid sequence set forth in SEQ ID NO: 1, a functional fragment or a functional variant thereof; or comprising and encoding A polynucleotide which is complementary or strictly hybridized to the amino acid sequence of SEQ ID NO: 1, a functional fragment thereof or a functional variant.
  • these recombinant expression vector rights comprise a polynucleotide encoding the amino acid sequence set forth in SEQ ID NOs: 1-6, or comprise a polynucleotide complementary or rigorous to the amino acid sequence set forth in SEQ ID NOs: 1-6.
  • Hybridized polynucleotide The host cell further comprising the above recombinant expression vector is also provided. Furthermore, the present invention also provides the use of the above polypeptide or peptidomimetic, nucleotide and recombinant expression vector for the manufacture of a medicament for chronic liver disease after infection with hepatitis B virus.
  • the drug can be a therapeutic vaccine for hepatitis B.
  • the medicament may comprise a pharmaceutical composition, which may comprise an optional pharmaceutical carrier.
  • the polypeptide (including its functional fragment or variant) and its peptidomimetic, as an effective inhibitor of HBx, have the ability to inhibit the biological activity of HBx and inhibit Hepatitis caused by hepatitis B virus infection, resulting in cirrhosis and liver cancer that occurs on the basis of cirrhosis. It is worth emphasizing that the above-mentioned polypeptide provided by the present invention has an obvious pharmacodynamic effect and thus can be an effective drug for treating liver diseases caused by hepatitis B virus infection.
  • FIG. 2 Application of reporter gene assay The effect of the polypeptide gene plasmid of the present invention on liver cancer cells expressing HBx protein.
  • the result shows - 1 ⁇ -13 ⁇ 4 ? 2# inhibited the activity of NF- ⁇ promoter in HepG2-X cells, L-O2-X cells and HepG2.2.15 cells in a dose-dependent manner, while HepG2 hepatoma cells without HBx and L- without HBx O2 liver cells have no effect.
  • the five functional variant genes (0.15 ⁇ g/well) of -1 ⁇ -13 ⁇ 4 ⁇ 2# have a certain inhibitory effect on the activity of NF- ⁇ promoter in HepG2-X cells and L-O2-X cells. .
  • Figure 3 Effect of detecting the polypeptide of the present invention on the activity of the expressed NF- ⁇ promoter by the expressed HBx protein at the cellular level.
  • the results showed that the synthetic peptide Anti-HBxP2# inhibited the activity of the NF- ⁇ promoter in HepG2-X cells, L-O2-X cells and HepG2.2.15 cells expressing HBx protein, and the inhibitory effect was observed.
  • Figure 4 Effect of transfection of the polypeptide gene plasmid of the present invention on liver cancer cells expressing HBx protein using MTT assay.
  • the result shows - 1 ⁇ -13 ⁇ 4 ? 2# inhibited the growth and proliferation of HepG2-X cells, L-O2-X cells and HepG2.2.15 cells in a dose-dependent manner, but for HepG2 hepatoma cells and L-O2 hepatocytes without HBx expression. no effect.
  • ⁇ - ⁇ 1 ⁇ - ⁇ 2# ⁇ 3 ⁇ 4 Five functional variant genes (0.15 ⁇ g/well) also inhibited the growth and proliferation of HepG2-X cells and L-O2-X cells.
  • Figure 5 Effect of the polypeptide of the invention on the growth of liver cancer cells using MTT. The results showed that the artificially synthesized peptide 1 ⁇ -13 ⁇ 4 was used. 2# pairs 11 ⁇ 2 02- cells, L-O2-X Cells and HepG2.2.15 cells have inhibitory effects on growth and proliferation, and their effects are dose-dependent. There was no effect on HepG2 liver cancer cells and L-O2 liver cells without HBx expression. *P ⁇ 0.05, **P ⁇ 0.01, Student's t test statistical analysis.
  • Figure 7 Effect of synthetic peptide VIII 1 ⁇ -13 ⁇ 4 ?2# on HepG2.2.15 cells.
  • the results of nude mice inoculation experiments showed that the artificially synthesized peptide 1 ⁇ -118 ⁇ 2# had a significant inhibitory effect on the growth and proliferation of HepG2.2.15 cells. ** P ⁇ 0.01, Student's t test statistical analysis
  • Figure 8 Schematic diagram of the construction of a polypeptide eukaryotic expression vector. detailed description
  • the term "separated” refers to the separation of a substance from its original environment (for example, if it is naturally produced, its natural environment).
  • a naturally occurring polynucleotide or polypeptide that is present in a living organism is not isolated, and the same polynucleotide or polypeptide is separated from some or all of the material with which it coexists in the natural system.
  • Such polynucleotides or polypeptides may be part of a vector or part of a composition. Since the carriers and compositions are not components of their natural environment, they are still separated.
  • purified means that it has been improved in purity.
  • Purity is a relative term in this and is unnecessarily interpreted as absolute purity.
  • the purity can be at least about 50%, or can be greater than 60%, 70%, 80%, 90%, or can be 100%.
  • the separated material is separated from its original environment. Living body The polynucleotides and polypeptides in the natural state of the cells are not isolated, but the same polynucleotides and polypeptides are separated from other substances existing in the natural state, and are separated, and the purity is improved. It is therefore also purified.
  • Nucleic acid refers to a nucleotide, oligonucleotide or polynucleotide and fragments or portions thereof.
  • the nucleic acid of the present invention can exist in the form of RNA (e.g., mRNA), or in the form of DNA (e.g., cDNA or genomic DNA).
  • DNA can be double-stranded or single-stranded.
  • the single-stranded DNA or RNA may be either a coding strand (sense strand) or a non-coding strand (antisense strand).
  • polynucleotide of the present invention may also be fused at its 5' or 3' end to a polynucleotide encoding a tag tag (tag sequence or tag sequence).
  • tag tag tag sequence or tag sequence
  • They may be synthetic or obtained from natural sources (eg, isolated and/or purified), which may comprise natural, non-natural or modified nucleotides, and which may comprise natural, non-natural or A bond between the altered nucleotides, such as a phosphoramidate linkage or a phosphorothioate linkage, is used in place of the phosphodiester linkage present between the nucleotides in the unmodified oligonucleotide.
  • amino acid sequence or “polypeptide” is meant an amino acid in which a peptide, oligopeptide, polypeptide or protein and a partial fragment thereof are linked by a peptide bond.
  • amino acid sequence ⁇ IJ in the present invention relates to the amino acid sequence of a naturally occurring protein molecule, such "polypeptide” or “protein” does not mean limiting the amino acid sequence to the integrity associated with the protein molecule.
  • the amino acid sequences of the invention may contain additional peptides. As an additional peptide, a peptide such as a histidine tag (His-tag) or an epitope such as Myc or FLAG is exemplified.
  • “Deletion” refers to the deletion of one or more amino acids or nucleotides in an amino acid sequence or nucleotide sequence.
  • “Insertion” or “addition” refers to an alteration in an amino acid sequence or nucleotide sequence that results in an increase in one or more amino acids or nucleotides compared to a molecule that is naturally occurring or altered. "Replacement” refers to the replacement of one or more amino acids or nucleotides by a different amino acid or nucleotide. "Deleting, replacing or adding one or more amino acids or nucleotides” means deleting, replacing or adding one or more numbers of amino acids or nucleosides by a known mutant nucleic acid or polypeptide such as site-directed mutagenesis. The acid, or the above mutation of the naturally occurring nucleic acid or polypeptide, is isolated and purified.
  • the amino acid mutation may comprise an amino acid having one or more amino acid residues in a D-type conformation, a rare amino acid present in nature, or an artificially modified amino acid, which may or may not be encoded by a genetic code.
  • mutations in the induced nucleic acid may include naturally occurring nucleotides in nature, and may also include modified nucleotides.
  • a "functional fragment" of a polypeptide refers to any portion of a polypeptide of the invention that retains substantially similar or identical biological activities and functions of the polypeptide (i.e., "parent" polypeptide) as part of it.
  • a “functional variant” of a polypeptide refers to an amino acid sequence that substantially retains a biological function or activity that is identical or similar to the polypeptide or amino acid sequence, and which may include, for example, 1) one or more amino acid residues in the original amino acid sequence.
  • a deletion and/or one or more amino acid residues are added; or 2) one or more amino acid residues in the original amino acid sequence are replaced by conservative or non-conservative amino acid residues; or 3) one or more of the original amino acid sequences a group on an amino acid residue is substituted by another group; or 4) a fusion of the original amino acid sequence with another molecule or compound (such as sugar, lipid, polyethylene glycol, etc.); or 5) the original a polypeptide sequence obtained by fusing an amino acid sequence with an added amino acid sequence (such as a leader sequence or a secretory sequence or a sequence for purifying the polypeptide, etc.); or 6) a reverse amino acid sequence of the original amino acid sequence; or 7) mixing.
  • a functional variant of the amino acid sequence may comprise an amino acid having one or more amino acid residue conformations as a D-type, a rare amino acid present in nature, or an artificially modified amino acid, which may or may not be genetically Codon encoded.
  • a "reverse analog" of a polypeptide refers to a polypeptide comprising an inverted amino acid sequence of the parent polypeptide such that the amino acid sequence of the inverse analog (when read from the N-terminus to the C-terminus) and when reading from the C-terminus to the N-terminus
  • the amino acid sequence of the parent polypeptide is the same; in addition, each amino acid in the reverse analog is the D isomer of the amino acid, and the D isomer is opposite to the L isomer.
  • the inverse analog of the tripeptide Val-Ala-Gly has the amino acid sequence Gly-Ala-Val, wherein each amino acid is the D isomer.
  • peptidomimetic means a compound having substantially the same general structure as the corresponding polypeptide and having, for example, a modification capable of increasing its stability or biological function.
  • Peptidomimetics include, for example, those compounds comprising the same amino acid sequence as the corresponding polypeptide, but between two or more of these amino acids, the peptidomimetic has an altered backbone.
  • the peptidomimetic can include synthetic or non-naturally occurring amino acids in place of naturally occurring amino acids.
  • a "degenerate variant" of a nucleotide sequence is a polynucleotide sequence which differs from the parent nucleotide sequence but which encodes a protein or polypeptide or a protein encoded by a parent nucleotide sequence or The same as the peptide.
  • "Nucleic acid hybridization” is well known in the art (see, for example, Sambrook et al.
  • hybridization temperature varies depending on the length of the base sequence or its base sequence.
  • stringent conditions refer to hybridization and elution at lower ionic strengths and higher temperatures.
  • This inhibition can be detected by performing hybridization (Southern blotting, Northern blotting, etc.) under conditions of reduced stringency.
  • a substantially homologous sequence or hybridization probe can compete for and inhibit binding of a fully complementary sequence to a target sequence under conditions of reduced stringency.
  • the condition of reduced stringency does not allow for non-specific binding, and the two sequences bind to each other and still require specific or selective interactions.
  • the "identity” or “identity” percentage of an amino acid sequence or nucleotide sequence refers to the percentage of sequences that are identical or similar in the comparison of two or more amino acid or nucleotide sequences.
  • MEGALIGN program Lasergene software package, DNASTA, Inc., Madison, WI.
  • 0 MEGALIGN program can compare two or more sequences according to different methods such as the Cluster method. (See Higgins & Sharp, Gene 73:237-244, (1988)).
  • the Cluster method arranges the sets of sequences into clusters by examining the distance between all pairs, and then assigns the clusters in pairs or groups.
  • the percent identity between two amino acid sequences, such as sequence A and sequence B can be calculated by:
  • nucleic acid sequences are determined by the Cluster method or by methods known in the art, such as the method of Jotun Hein (see Hein J., Methods in Emzumology 183: 625-645, 1990).
  • Recombinant expression vector refers to a genetically modified oligonucleotide or polynucleotide recombinant having a nucleotide sequence encoding an mRNA, a protein, a polypeptide, or a peptide, which can be introduced into a host cell. Expression of the corresponding mRNA, protein, polypeptide, or peptide.
  • the recombinant expression vector may comprise any type of nucleotide sequence, but is not limited to DNA or RNA, may be single-stranded or double-stranded, artificially synthesized or derived from nature, or may be non-natural or altered nucleoside. acid.
  • chronic liver disease after hepatitis B virus infection refers to liver disease caused by hepatitis B virus infection, including liver cirrhosis which occurs after fibrotic connective tissue proliferation in liver tissue caused by recurrent chronic hepatitis and chronic hepatitis, and Most of the liver cancer that occurs on the basis of cirrhosis.
  • liver cancer is the result of persistent infection of hepatitis B virus, which can be called liver cancer after hepatitis B.
  • treating and “preventing” and words derived therefrom may be considered to be a degree of treatment or prevention recognized by those skilled in the art.
  • “Prophylaxis” in the present invention is understood to delay the onset of a disease, or a symptom or condition thereof.
  • the invention relates to isolated or purified polypeptides.
  • the polypeptide may comprise, consist essentially of, or consists of the amino acid sequence bad 1 J Pro-Asp-Leu- His-Lys-Asn-Glu-Leu-Lys-His-Val-Lys -Tyr (SEQ ID NO: 1) consisting of Peptide.
  • SEQ ID NO: 1 J Pro-Asp-Leu- His-Lys-Asn-Glu-Leu-Lys-His-Val-Lys -Tyr SEQ ID NO: 1 J Pro-Asp-Leu- His-Lys-Asn-Glu-Leu-Lys-His-Val-Lys -Tyr
  • the polypeptide can inhibit the promoter activity of nuclear factor kappa (NF- ⁇ ) activated by HBx at a molecular level, thereby embodying the effective inhibition of HBx by the polypeptide, and the inhibitory effect thereof is in a dose-effect relationship;
  • the polypeptide also showed effective inhibition of the growth and proliferation of hepatoma cells containing HBx gene, and its inhibitory effect was also dose-effect relationship; the inoculation method of nude mice showed that the polypeptide could effectively inhibit expression at the overall level.
  • the tumorigenic ability of HBx liver cancer cells are examples of tumorigenic ability of HBx liver cancer cells.
  • the invention also provides various functional fragments of the polypeptides of the invention.
  • the functional fragment may be any fragment of a contiguous amino acid sequence of a polypeptide of the invention, provided that it retains the biological activity of the parent polypeptide to a similar extent, to the same extent, or to a greater extent than the parent polypeptide, eg, inhibits HBx Activity.
  • the functional fragment may comprise, for example, about 10%, 20%, 30%, 40%, 50%, 60%, 70% of the parent polypeptide,
  • the functional fragment may further comprise an additional amino acid, for example, an amino acid different from the amino acid sequence of the parent polypeptide, at the amino or carboxy terminus of the contiguous amino acid sequence fragment, or both of its amino and carboxyl groups.
  • the additional amino acid does not interfere with the biological function of the functional fragment, for example, inhibits the activity of HBx, and effectively inhibits the growth of cancer cells, preferably liver cancer cells, particularly liver cancer cells infected with hepatitis B virus. proliferation.
  • the additional amino acid can result in enhanced biological activity when compared to the biological activity of the parent polypeptide.
  • a functional fragment of a polypeptide of the invention comprising an amino acid sequence having at least 70% sequence identity to the parent polypeptide is a preferred sequence.
  • a functional fragment of the invention comprises the addition of an amino acid, such as one or two amino acids, at the amino terminus (N-terminus) and/or carboxy terminus (C-terminus) of the parent polypeptide.
  • functional variants of the polypeptides of the invention and functional fragments thereof are also included within the scope of the invention.
  • the functional variants of the polypeptides of the invention and functional fragments thereof retain substantially similar or identical biological activities as the parent polypeptide or the parental functional fragment, for example, inhibiting the activity of HBx, and effectively inhibiting liver cancer cells, particularly infections.
  • the functional variant may be at least about 50%, 60%, 70%, 80%, 90%, 95% or 100 with the amino acid sequence of the parent polypeptide or the parent functional fragment. % identity.
  • a functional variant of a polypeptide of the invention and a functional fragment thereof comprises at least 70% sequence identity to said parent polypeptide or a parental functional fragment. More preferably, a functional variant of a polypeptide of the invention and a functional fragment thereof comprises only 1-3 amino acids different from the parent polypeptide or the parental functional fragment.
  • a functional variant of a polypeptide of the invention and a functional fragment thereof differs from the parent polypeptide or a parental functional fragment by only one amino acid.
  • Functional variants of the polypeptides of the invention and functional fragments thereof can include a parent polypeptide or a parental functional fragment amino acid sequence having at least one conservative amino acid substitution.
  • the functional variants of the polypeptides of the invention and functional fragments thereof may comprise an amino acid sequence of a parent polypeptide or a parental functional fragment having 2, 3, 4, 5, or more conservative amino acid substitutions. .
  • functional variants of the polypeptides of the invention and functional fragments thereof may also comprise an amino acid sequence of a parent polypeptide or a parental functional fragment having at least one non-conservative amino acid substitution.
  • an amino acid sequence of a parent polypeptide or a parental functional fragment having 2, 3, 4, 5, or more non-conservative amino acid substitutions can be included.
  • the amino acid substitution increases the biological function or activity of a functional variant of a polypeptide of the invention and a functional fragment thereof such that when compared to the parent polypeptide or a parental functional fragment, the functional variant organism Learning function or activity promotion.
  • a functional variant of a polypeptide of the invention and functional fragments thereof comprises one or more conservative amino acid substitutions.
  • Conservative amino acid substitutions are well known in the art and refer to amino acid substitutions in which one amino acid having a certain physical and/or chemical property is exchanged for another amino acid having the same chemical or physical properties. Those skilled in the art understand that conservative amino acid substitutions may not result in significant changes in the structure or function of the protein.
  • Typical conservative substitutions include, for example, replacing another acidic amino acid (eg, AsP or Glu) with an acidic amino acid, and replacing another amino acid having a non-polar side chain with an amino acid having a non-polar side chain (eg, Ala, Gly, Val, He, Leu, Met, Phe, Pro, Trp, Val, etc.), replacing another basic amino acid with a basic amino acid (Lys, Arg, etc.), replacing the other with a polar side chain Amino acids of the side chain (Asn, Cy S ,
  • polypeptides of the invention may also include retro-inverse analogs of any of the polypeptides or functional fragments of the invention.
  • "reverse anti-analog” is Refers to a polypeptide comprising an inverted amino acid sequence of a parent polypeptide such that the amino acid sequence of the inverse analog (when read from the N-terminus to the C-terminus) and the amino acid sequence of the parent polypeptide when read from the C-terminus to the N-terminus the same.
  • each amino acid is the D isomer of the amino acid, and the D isomer is opposite to the L isomer.
  • the inverse analog of the tripeptide Val-Ala-Gly has the amino acid sequence Gly-Ala-Val, wherein each amino acid is the D isomer.
  • the functional variant of such a molecule preferably comprises the inverse analog of SEQ ID NO: 1.
  • the polypeptides (including functional fragments) of the invention and functional variants thereof can be of any length, i.e., can include any number of amino acids, provided that the polypeptide (including functional fragments) and functional variants retain the necessary biological activity.
  • a polypeptide of the invention can be from 4 to 2000 amino acids in length, such as 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 18, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, 125, 150, 175, 200, 300, 400,
  • the polypeptide of the present invention is 6-20 amino acids in length and meets the requirements for pharmacodynamics and half-life of the polypeptide drug.
  • a functional variant of a polypeptide of the invention and a functional fragment thereof has an amino acid difference from the parent polypeptide SEQ ID NO: 1 having a SEQ ID with the parent polypeptide
  • polypeptides of the invention Similar biological activity and function, for example, inhibition of HBx activity, and effective inhibition of cancer cells, preferably liver cancer cells, particularly liver cancer cells expressing HBx.
  • functional variants of the polypeptides of the invention and functional fragments thereof include SEQ ID NOs: 2-6.
  • the invention also provides peptidomimetics of the polypeptides, including functional fragments and functional variants.
  • the peptidomimetic is a peptidomimetic.
  • Peptidomimetics refers to peptide mimetics in which the side chain of each amino acid is attached to the nitrogen atom of the amino acid rather than to the alpha carbon.
  • a peptidomimetic can be considered a purine-substituted glycine having a repeating unit of the NRCH 2 CO general structure and having the same or substantially the same amino acid sequence as the corresponding polypeptide.
  • the peptidomimetic comprises an altered backbone, Wherein the bond between each amino acid is methylated.
  • the peptidomimetic can include a methylated peptide backbone of the structure:
  • Polypeptides (including functional fragments and functional variants) of the invention, as well as peptidomimetics, may include the replacement of naturally occurring amino acids with synthetic amino acids.
  • synthetic amino acids include, for example, aminocyclohexanecarboxylic acid, norleucine, ot-amino-n-decanoic acid, homoserine, S-acetylaminomethyl-cysteine Acid, trans-3-hydroxyproline, trans-4-hydroxyproline, 4-aminophenylalanine, 4-benzoylphenylalanine, 4-nitrophenylalanine, 4 -Chlorophenylalanine, 4-carboxyphenylalanine, ⁇ -phenylserine, ⁇ -hydroxyphenylalanine, phenylglycine, a-naphthylalanine, cyclohexylalanine, cyclohexylglycine , indoline-2-carboxylic acid, 1,2,3,
  • the polypeptides of the invention may also comprise a cell-penetrating peptide (CPP).
  • CPP cell-penetrating peptide
  • the CPP promotes passage of a polypeptide of the invention across a cell membrane and into a cell.
  • CPPs are known in the art. See, for example, Deshayes et al., Cell. Mol. Life Sci. 62: 1839-1849 (2005); EI-Andaloussi et al., Curr. Pharm. Design. 11 : 3597-3611 (2005).
  • the CPP can be any of those known in the art.
  • Polypeptides (including functional fragments and functional variants) and peptidomimetics of the invention may also, for example, be lipidated (eg, fatty acidated), glycosylated, amidated, carboxylated, phosphorylated, esterified, N- Acylation, cyclization by disulfide bonds, conversion to acid addition salts, dimerization or multimerization, and/or conjugation.
  • lipidated eg, fatty acidated
  • glycosylated glycosylated, amidated, carboxylated, phosphorylated, esterified, N- Acylation, cyclization by disulfide bonds, conversion to acid addition salts, dimerization or multimerization, and/or conjugation.
  • the polypeptides of the invention (including functional fragments and functional variants) as well as peptidomimetics can be lipidated derivatives.
  • the lipid molecules contained may include any lipid known in the art, for example, fatty acids, phospholipid groups, glycosylphosphatidylinositol, phosphatidylserine, phosphatidylethanolamine, sphingomyelin, phosphatidylcholine, cardiolipin, Phosphatidylinositol, phosphatidic acid, lysophosphoglyceride, and cholesterol groups.
  • fatty acids for example, fatty acids, phospholipid groups, glycosylphosphatidylinositol, phosphatidylserine, phosphatidylethanolamine, sphingomyelin, phosphatidylcholine, cardiolipin, Phosphatidylinositol, phosphatidic acid, lysophosphoglyceride, and cholesterol groups.
  • the lipidated derivative is a fatty acid derivative
  • the fatty acid molecule may be any C8-C20 fatty acid, for example, lauric acid, palmitic acid, myristic acid, stearic acid, oleic acid, linoleic acid, flax. Acid, arachidonic acid, eicosapentaenoic acid, erucic acid, or arachidic acid.
  • the fatty acid may also optionally contain other functional groups on any carbon atom, for example, one or more amino groups.
  • the fatty acid molecule can be attached to any of the appropriate portions of the polypeptides of the invention, including functional fragments and functional variants, as well as peptide mimetics.
  • a fatty acid molecule is included at the amino terminus, the carboxy terminus, or both the amino and carboxy termini of the polypeptide of the present invention.
  • the fatty acid molecules can be attached to the polypeptides of the invention (including functional fragments and functional variants) as well as to peptide mimetics, either directly or through a linker.
  • Polypeptides (including functional fragments and functional variants) of the invention, as well as peptidomimetics, including derivatives thereof, such as fatty acid derivatives may also be monomeric peptides, dimeric peptides or multimeric peptides.
  • the polypeptides of the invention (including functional fragments and functional variants) as well as peptidomimetics can be obtained by methods known to those skilled in the art (see, for example, Chan et al, Fmoc Solid
  • polypeptides of the invention can be isolated and/or purified from, for example, plants, bacteria, insects, mammals such as rats, humans, and the like. Methods of separation and purification are also well known in the art.
  • polypeptides described herein, including functional fragments thereof as well as functional variants can be purchased commercially from commercial companies.
  • the invention also provides isolated polynucleotides encoding any of the polypeptides of the invention (including functional fragments and functional variants) as described.
  • the polynucleotide comprises a coding sequence encoding any of the polypeptides of the invention (including functional fragments and functional variants), for example, the nucleotide sequence of SEQ ID NO: 1 encoding SEQ ID NO: 7, and any of these coding sequences Degenerate variants; alternatively, the polynucleotide may also comprise additional coding sequences and/or non-coding sequences.
  • the present invention provides a polynucleotide comprising the isolated nucleotide or a fragment thereof, which comprises a nucleotide sequence complementary to the nucleotide sequence of any of the nucleic acids of the present invention or under stringent conditions
  • the nucleotide sequence of any of the polynucleotides of the present invention hybridizes.
  • the invention also provides variants of the above polynucleotides which encode functional fragments or functional variants of polypeptides or polypeptides having the same amino acid sequence as the polypeptides of the invention, including functional fragments and functional variants. Variants of this polynucleotide may be naturally occurring allelic variants or non-naturally occurring variants.
  • nucleotide variants include substitution variants, deletion variants, and insertion variants.
  • an allelic variant is an alternative form of a polynucleotide which may be a substitution, deletion or insertion of one or more nucleotides, but does not substantially alter the function of the polypeptide encoded thereby.
  • the polynucleotide of the present invention may be obtained by purification in a natural form, may also be produced by recombination, or may be constructed based on a chemical synthesis/enzyme ligation reaction using methods known in the art.
  • the polynucleotide of the present invention may contain naturally occurring nucleotides, and may also contain modified nucleotides.
  • the modified nucleotide is designed to increase the biological stability of the molecule or to increase the physical stability of the duplex formed upon hybridization (eg, a phosphorothioate derivative and an acridine-substituted nucleotide), including , for example, 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxymethyl)uracil, 5 -Methoxyaminomethyl-2-thiouracil, uracil-5-glycolic acid, 5-methyl-2-thiouracil, 2-thiocytosine, 2-thiouracil, 4-thiouracil , 5-methyluracil, uracil-5-hydroxyacetic acid methyl ester, 3-(3-amino-3-N-2-carboxypropyl) uracil, and 2,6-diaminopurine.
  • nucleic acids of the invention are recombinantly produced.
  • the invention also provides a recombinant expression vector comprising any of the polynucleotides of the invention.
  • the recombinant expression vector can be any suitable recombinant expression vector and can be used to transform or transfect any suitable host cell.
  • Suitable vectors include those designed for propagation and amplification or for expression, or both, such as plasmids or viruses.
  • the vector can be freely combined by the following: pUC series, pcDNA series, pBluescript, pET series, pGEX series and pEX series. It is also possible to use a phage vector such as GT10, GTI K ⁇ . EMBL4 and the like.
  • Plant expression vectors can include pBI01, pBI101.2. pBI101.3.
  • Animal expression vectors can include pEUK-Cl, pMAM, and pMAMneo.
  • the invention relates to a plasmid using the pcDNA series.
  • the recombinant expression vector of the present invention can be prepared using standard recombinant DNA techniques.
  • a circular or linear expression vector construct can be made to comprise a replication system that functions in a prokaryotic or eukaryotic host cell.
  • the recombinant expression vector includes regulatory sequences, such as transcriptional and translational initiation and termination codons. The regulatory sequences are specific for the type of host (eg, bacteria, fungus, plant, or animal) into which the vector is introduced.
  • the recombinant expression vector can include one or more marker genes that allow for selection of transformed or transfected hosts.
  • Marker genes include resistance to biocides, for example, resistance to antibiotics, heavy metals, and the like, complementation in auxotrophic hosts to provide prototrophy, and catalytic biofluorescence, and the like.
  • Suitable marker genes for the expression vector of the present invention include, for example, neomycin/G418 resistance gene, luciferase reporter gene, hygromycin resistance gene, histidine resistance gene, tetracycline resistance gene, and ampicillin Resistance gene.
  • the recombinant expression vector can include a native or non-native promoter.
  • Promoter selection for example, strong, weak, inducible, tissue-specific, and development-specific, is within the skill of the skilled artisan. Similarly, the combination of a nucleotide sequence and a promoter is also in the art. Within the skills of the personnel.
  • the promoter may be a non-viral promoter or a viral promoter, for example, a cytomegalovirus (CMV) promoter, an SV40 promoter, an RSV promoter, and a promoter present in the long terminal repeat of the murine stem cell virus.
  • CMV cytomegalovirus
  • the recombinant expression vectors of the invention can be designed for transient expression, for stable expression, or for both. Furthermore, the recombinant expression vector can be prepared for constitutive expression or for inducible expression.
  • the recombinant expression vector can include a suicide gene.
  • suicide gene refers to a gene that causes cell death after expression in a cell. Suicide gene expression can affect the sensitivity of cells to certain agents, such as drugs, leading to cell death. Suicide genes are known in the art (see, for example, suicide gene therapy: methods and reviews (Suicide Gene)
  • HSV herpes simplex virus
  • TK thymidine Kinase gene and purine nucleoside phosphatase, as well as nitroreductase and the like.
  • the invention also provides host cells comprising any of the recombinant expression vectors described herein.
  • the host cell is any type of cell comprising a recombinant expression vector of the invention.
  • the host cell may be a eukaryotic cell, for example, a plant, an animal, a fungus, or an alga, or may be a prokaryotic cell, for example, a bacterium or a protozoan.
  • the host cell can be a cultured cell or a primary cell, i.e., a primary cell isolated directly from an organism such as a human.
  • the host cell can be an adherent cell or a suspended cell, i.e., a cell grown in a suspension.
  • Suitable host cells are known in the art and include, for example, DH5a E. coli cells, Chinese hamster ovary cells, monkey VERO cells, COS cells, HEK293 cells, and the like.
  • the host cell is preferably a prokaryotic cell.
  • the host cell is preferably a mammalian cell. Human cells are preferred host cells.
  • the host cell can be of any cell type, can be from any type of tissue, and can be of any developmental stage.
  • the invention also provides a population of cells comprising at least one host cell as described herein.
  • the cell population can be a heterogeneous population, including a host cell comprising any of the recombinant expression vectors, and at least one other cell, eg, a host cell (eg, a T cell) that does not comprise any of the recombinant expression vectors, or Cells other than T cells, for example, B cells, macrophages, neutrophils, erythrocytes, hepatocytes, endothelial cells, epithelial cells, myocytes, brain cells, and the like.
  • a host cell eg, a T cell
  • B cells macrophages, neutrophils, erythrocytes, hepatocytes, endothelial cells, epithelial cells, myocytes, brain cells, and the like.
  • the population of cells can be a substantially homogeneous population, wherein the population primarily comprises a host cell comprising the recombinant expression vector (e.g., consisting essentially of a host cell comprising the recombinant expression vector).
  • the population can also be a clonal population of cells, wherein all cells of the population are clones of a single host cell comprising a recombinant expression vector such that all cells of the population comprise the recombinant expression vector.
  • the population of cells is a clonal population comprising a host cell comprising a recombinant expression vector as described herein. Conjugate
  • the invention also includes conjugates, eg, biological conjugates, including the polypeptides of the invention (including functional fragments and functional variants), as well as any of peptidomimetics, polynucleotides, recombinant expression vectors, or host cells.
  • conjugates eg, biological conjugates, including the polypeptides of the invention (including functional fragments and functional variants), as well as any of peptidomimetics, polynucleotides, recombinant expression vectors, or host cells.
  • conjugates eg, biological conjugates, including the polypeptides of the invention (including functional fragments and functional variants), as well as any of peptidomimetics, polynucleotides, recombinant expression vectors, or host cells.
  • Conjugates as well as methods for the usual synthesis of conjugates, are also known in the art (see, for example, Hudecz, F., Methods Mol. Biol 298: 209-223 (2005) and Kirin Et.
  • the above various substances provided by the present invention include polypeptides (including functional fragments and functional variants) as well as peptidomimetics, fatty acid derivatives, polynucleotides, recombinant expression vectors, and host cells (including populations thereof), conjugates Etc. (collectively referred to as "the substance of the invention") can be isolated, purified, synthetic, and/or recombinant.
  • the substances of the invention may also be formulated into compositions such as pharmaceutical compositions.
  • the invention provides any of the polypeptides (including functional fragments and functional variants) as well as peptidomimetics, fatty acid derivatives, conjugates, nucleic acids, recombinant expression vectors, and host cells (including populations thereof), and A pharmaceutical composition of a pharmaceutically acceptable carrier.
  • the hair containing any of the substances of the present invention may comprise more than one substance of the invention, for example: a polypeptide and a nucleic acid, or two or more different polypeptides.
  • the pharmaceutical composition may include a combination with another pharmaceutically active agent or drug.
  • the other or more pharmaceutically active agents or drugs may preferably include, for example, a chemotherapeutic agent, for example, asparaginase, busulfan, carboplatin, cisplatin, daunorubicin, doxorubicin, fluorouracil, gemcitabine , hydroxyurea, methotrexate, paclitaxel, rituximab, vinblastine, vincristine, and the like.
  • the pharmaceutical composition comprises a substance of the invention in combination with a lipid.
  • the lipid may be any lipid, including fatty acids, phospholipids, sterols, sphingolipids, guanidines, glycerides, glycerophosphates, prenol lipids, glycolipids, and polyketides. Such lipids are known in the art.
  • the pharmaceutically acceptable carrier may be any of those conventionally used, and they may be defined only by chemical physical factors such as solubility and lack of reactivity with the active compound, and by route of administration.
  • the pharmaceutical carriers of the present invention for example, vehicles, adjuvants, excipients, diluents and the like, are well known to those skilled in the art and are readily available to the public.
  • the pharmaceutically acceptable carrier is chemically inert to the active agent and does not have deleterious side effects or toxicity under the conditions of use.
  • the choice of carrier should be determined by the particular material of the invention, as well as by the particular method used to administer the materials of the invention.
  • suitable formulations of the pharmaceutical compositions of the invention are exemplary and are not intended to be limiting in any way.
  • the agents of the invention may be administered using more than one route, and in certain instances, a particular route may provide a more direct and effective method than the other route.
  • the pharmaceutical composition is a topical formulation, an intravenous formulation, or a subcutaneous formulation.
  • the pharmaceutical combination is a topical formulation.
  • Topical formulations are well known to those skilled in the art. In the case where the invention is applied to the skin, the formulation is particularly suitable.
  • the topical preparation of the present invention may be, For example, ointments, lotions, ointments, patches, oils, pastes, sprays, such as aerosol sprays, gels, roll-on liquids, solid sticks, and the like.
  • the topical formulation of the invention is a cream, lotion, ointment, or patch.
  • Formulations suitable for oral administration may consist of: a) a liquid solution, such as an effective amount of a substance of the invention dissolved in a diluent such as water, saline, or orange juice; b) capsules, tablets, lozenges, and the like, each An active ingredient comprising a predetermined amount, solid or granules; c) a powder; d) a suspension in a suitable liquid; and e) a suitable emulsion.
  • Liquid preparations may include diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene glycol, with or without the addition of a pharmaceutically acceptable surfactant.
  • the capsule form can be a conventional hard or soft shell gelatin type comprising, for example, a surfactant, a lubricant, and an inert filler such as lactose, sucrose, calcium phosphate, and corn starch.
  • Tablet form may include one or more of the following: lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, gum arabic, gelatin, guar gum, gelatinous dioxide Silicon, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffers, disintegrants, wetting agents, preservatives, flavoring Agent, and other drug compatible excipients.
  • Tablet forms may include the materials of the invention in which the flavoring agent is typically in sucrose or gum arabic, and soft lozenges comprising the materials of the invention in an inert matrix such as gelatin and glycerin, or sucrose and gum arabic, additionally comprising Emulsions, gels and the like of excipients known in the art.
  • the agents of the invention alone or in combination with other suitable ingredients, can be formulated as an aerosol formulation for administration by inhalation. These aerosol formulations can be placed in a pressurized, usable medium such as dichlorodifluoromethane, acetonide, nitrogen, and the like. They can also be formulated as non-pressurized formulations, such as in nebulizers or nebulizers.
  • the spray formulation can also be used to spray into the mucosa.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injections, which may contain an antioxidant, a buffer, a bacteriostatic agent, and a solute which renders the formulation isotonic with the blood of the intended recipient, and Aqueous and non-aqueous sterile suspensions, which may include suspensions, solubilizers, thickeners, stabilizers, and preservatives.
  • the substance of the invention can be used in a pharmaceutically acceptable carrier Used in physiological diluents, such as sterile liquid or liquid mixtures, including water, saline, aqueous dextrose and related sugar solutions, alcohols such as ethanol or cetyl alcohol, glycols such as propylene glycol or polyethylene glycol, dimethyl Alum, glycerin, ketal such as 2,2-dimethyl-1,3-dioxolan-4-methanol, ether, oil, fatty acid, fatty acid ester or glyceride, or acetylated fatty acid glyceride, With or without the addition of pharmaceutically acceptable surfactants such as soaps or detergents, suspensions such as gums, methylcellulose, hydroxypropylmethylcellulose, or nitromethylcellulose, or emulsifiers and other pharmaceuticals Agent.
  • physiological diluents such as sterile liquid or liquid mixtures, including water, saline, aqueous dextrose and related
  • Oils that can be used in parenteral formulations include petroleum, animal oils, vegetable oils, or synthetic oils. Specific examples of the oil include peanut oil, soybean oil, sesame oil, cottonseed oil, corn oil, olive oil, petroleum, and mineral oil. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid.
  • Suitable soaps for use in parenteral formulations include fatty alkali metals and triethanolamine salts and the like, and contain suitable soil release agents, for example a) cationic detergents: dimethyldiindenyl ammonium halides and mercapto halogenated pyridines; b) anionic detergents: mercapto, aryl, olefin sulfonate, sulfhydryl, olefin, ether, monoglycerol sulfate and sulfosuccinate; c) nonionic detergent: fatty amine oxide , fatty acid chain sterol amides and polyoxyethylene polypropylene copolymers, etc.: d) Amphoteric detergents: mercapto- ⁇ -aminopropionate and 2-mercapto-imidazoline quaternary ammonium salt; e) mixtures thereof.
  • suitable soil release agents for example a) cationic detergents: dimethyldiin
  • the parenteral preparation may contain from about 0.5% to about 25% by weight of the substance of the invention in solution. Preservatives and buffers can be used. To minimize or eliminate irritation at the site of injection, the composition may comprise one or more hydrophilic-lipophilic balance (HLB) nonionic surfactants.
  • HLB hydrophilic-lipophilic balance
  • the surfactant is used in the formulation in an amount of from about 5 to 15% by weight. Suitable surfactants include polyethylene glycol sorbitan fatty acid esters and high molecular weight adducts of epoxy oxime and hydrophobic groups formed by condensation of glycidin and propylene glycol. .
  • the parenteral formulation can be presented in unit or multi-dose sealed containers and can be stored under lyophilized (lyophilized) conditions, requiring only the addition of sterile liquid excipients such as water for injection prior to use. Temporary preparation of sterile powders, granules, and tablets of the type previously described Injection solutions and suspensions.
  • the substance of the invention or a composition comprising the substance of the invention may also be formulated as an injectable preparation.
  • the need for injectable compositions for effective pharmaceutical carriers is well known to those of ordinary skill in the art.
  • the cell when administered to a cell, such as a dendritic cell, the cell is administered by injection.
  • inventive substance of the present invention can be prepared as a suppository by mixing with various bases such as an emulsifying base or a water-soluble base.
  • bases such as an emulsifying base or a water-soluble base.
  • Formulations suitable for, for example, vaginal administration may be presented as pessaries, tampons, ointments, gels, pastes, foams, or spray formulations, in addition to the active ingredients thereof, those which are known in the art .
  • inventive materials of the present invention may be formulated to comprise a complex, such as a cyclodextrin comprising a complex, or a liposome.
  • the amount or dose of a substance of the invention to be administered should produce, for example, a therapeutic or prophylactic response in a subject or animal within a reasonable time frame.
  • the dose of the substance of the present invention should be sufficient to inhibit the proliferation of diseased cells during a period of about 2 hours or more, such as 12-24 hours or longer from the time of administration, to treat or prevent diseases (e.g., tumors, cancers). The role of etc.). In certain embodiments, the time period may even be longer.
  • the dosage should be determined by the efficacy of the particular substance of the invention and the condition of the animal (e.g., human) to be treated, as well as the body weight of the animal (e.g., human).
  • the dosage of the substance of the invention will also be determined by the presence, nature and extent of any side effects that may accompany the administration of a particular subject of the invention.
  • the materials of the present invention may be modified in any manner to enhance the therapeutic or prophylactic efficacy of the agents of the present invention.
  • the substance of the present invention can Conjugated to the targeted moiety either directly or indirectly via a linker.
  • the practice of conjugating a compound, such as a substance of the invention, to a targeting moiety is well known in the art (see, Wadwa et al, J. Drug Targeting, 3: 1 11, (1995) and U.S. Patent. No.
  • the agents of the invention may be modified in the form of a depot such that the manner in which the substance of the invention is released into the body to which it is administered is controlled in terms of time and body parts (see, e.g., U.S. Patent No. 4, 450,150).
  • the depot form of the substance of the invention may be, for example, an implantable composition comprising a substance of the invention and a porous or non-porous material, such as a polymer, wherein the substance of the invention is degraded by said substance and/or said non-porous substance And spread.
  • the depot is then implanted into the desired site in the body and the material of the invention is released from the implant at a predetermined rate.
  • the methods of the present invention for preventing and inhibiting the growth and proliferation of cancer cells include contacting the diseased cells with any of the pharmaceutical compositions of the present invention having an effective inhibitory amount.
  • the pharmaceutical composition of the present invention can be delivered to cancer cells by any conventional method.
  • the pharmaceutical composition is administered topically to the host.
  • the pharmaceutical composition is administered directly to cancer cells, for example, intratumoral delivery.
  • the pharmaceutical composition of the present invention comprising a polypeptide (including functional fragments and functional variants) and a peptidomimetic, a nucleic acid, a recombinant expression vector, and/or a host cell, can be used for preventing and inhibiting chronic liver disease induced by hepatitis B virus infection. , including hepatitis, and the resulting methods of cirrhosis and liver cancer.
  • chronic liver disease induced by the hepatitis B virus of the present invention may be present in any host.
  • the host is a mammal.
  • the host is a human.
  • the invention will be further illustrated below in conjunction with specific embodiments.
  • the present invention synthesizes a polypeptide having the amino acid sequence Pro-Asp-Leu-His-Lys-Asn-Glu-Leu-Lys-His-Val-Lys-Tyr (SEQ ID NO: 1) by artificial synthesis (hereinafter referred to as Anti -HBxP2#) .
  • the preparation of the polypeptide is carried out by a solid phase synthesis method, such as the use of AAPPTEC's Apex 396 polypeptide synthesis instrument, in the closed explosion-proof glass reactor according to the sequence shown in SEQ ID NO: 1 from the C-terminal to the N-terminal-amino
  • the amino acid is synthesized at the end, which means that the first amino acid monomer added to the amino acid sequence Pro-Asp-Leu-His-Lys-Asn-Glu-Leu-Lys-His-Val-Lys-Tyr is the C-terminal Tyr.
  • the solid phase synthesis method greatly reduces the difficulty of purification of each step of the product.
  • the side chains of the amino acids participating in the reaction are protected.
  • the carboxy terminus is free and must be activated prior to the reaction.
  • the specific synthesis consists of the following cycles:
  • the condensation reaction occurs, and after the final amino acid is synthesized, it is cleaved from the resin, and the crude peptide is purified by HPLC to obtain 98% pure Anti-HBxP2#, which is further identified by mass spectrometry, and its molecular weight is 1622.2.
  • Kd In solid phase synthesis, the elongation of the peptide chain is on the insoluble polystyrene resin carrier. OK.
  • the C-terminus of the synthetic polypeptide is first reacted with a chloromethyl polystyrene resin (benzyl chloride resin) to form a benzyl ester, and then the amino acid-protected amino acid is added one by one in the order of the primary structure of the peptide chain to make the peptide Chain extension.
  • a chloromethyl polystyrene resin benzyl chloride resin
  • the amino acid-protected amino acid is added one by one in the order of the primary structure of the peptide chain to make the peptide Chain extension.
  • the synthetic peptide 1 ⁇ -118 ⁇ 2# was analyzed by high pressure liquid chromatography (HPLCX application PLC Agela C18 column), and the obtained purity was 98.997%.
  • Example 2 Anti-HBx activity of in vitro polypeptides
  • the first method was to clone the cDNA expressing the polypeptide of Example 1 in the eukaryotic expression vector pcDNA3.1 by molecular cloning technique. (+), through gene transfection, in the liver cancer cells to achieve the purpose of expressing the polypeptides studied, and then observe the effect of the polypeptides studied to inhibit HBx; the second method is to use artificially synthesized peptides, directly added to cultured liver cancer cells In the culture medium, the effect of the polypeptide on inhibiting HBx was observed.
  • liver cancer cells There are two kinds of liver cancer cells used in the experiment: one is hepatoma HepG2-X cells constitutively expressing HBx (hepatocellular carcinoma HepG2 cells stably transfected with HBx); one is hepatoma HepG2.2.15 cells constitutively expressing the whole gene of hepatitis B virus (Stable transfection of HBV whole-gene liver cancer HepG2 cells).
  • HBx hepatocellular carcinoma HepG2 cells stably transfected with HBx
  • Hepatoma HepG2.2.15 cells constitutively expressing the whole gene of hepatitis B virus (Stable transfection of HBV whole-gene liver cancer HepG2 cells).
  • HBx has the function of activating the transcription factor NF- ⁇ , the detection of HBx at the molecular level is inhibited by the luciferase reporter gene detection method; since HBx has the effect of promoting the growth and proliferation of liver cancer cells in liver cancer cells, it can be passed through 3- (4,5-Dimethyl-2-thiazole) -2,5-diphenyltetrazolium bromide (MTT) The effect of the polypeptide of Example 1 on the proliferation of HepG2-X cells and HepG2.2.15 cells was examined. .
  • Annealing conditions 95 ° C, 2 minutes; every 8 seconds drop 0. C to 25 ° C, 90 minutes; 4 ° C, ⁇ .
  • the DH5a strain containing the pcDNA3.1 (+) plasmid preserved in our laboratory was activated, and the monoclonal was picked and added to LB liquid medium (containing ampicillin 100 mg/L) overnight at 37 °C.
  • the plasmid was extracted using the TransGen Plasmid Mini Kit. Double digestion system:
  • the synthetic base sequence includes cctgatcttc ga actaaaacat gttaaatat (SEQ ID NO: 7), and the linkage diagram is shown in FIG.
  • step 8 2) Add the 25 ⁇ ligation product from step 8 to the centrifuge tube containing the competent bacteria in the clean bench, mix with the gun head, and let stand for 30 minutes on ice;
  • reaction product was subjected to electrophoresis analysis on a 1.5% agarose gel. Positive clones were sent to the biotech company for sequencing. This plasmid was named p-Anti-HBxP2#.
  • Promoter reporter gene vector (PGL3-NF-KB, 0.3 ⁇ , purchased from Yuanpingyi (Tianjin) Biotechnology Co., Ltd.) was transfected into cells by liposome method and simultaneously with Renilla fluorescence
  • the prime enzyme expression vector (pRL-TK, Ol g, purchased from Promega) was an internal reference. Simultaneously transfecting different quality plasmids (0.25 g, 0.5 g and 0.75 g) obtained from step A or adding different concentrations of the synthetic polypeptides obtained in Example 1 ( ⁇ . ⁇ , ⁇ , 10 ⁇ and ⁇ ), each Repeat 3 wells for each concentration.
  • Inoculation of cells cells in logarithmic growth phase (cells listed in Table 1) were mixed with RPMI1640 or DMEM containing 10% fetal bovine serum into a single cell suspension at 4000-5000 cells per well. Inoculate into a 96-well cell culture plate at a volume of 100 ul per well.
  • Cultured cells cells were adherent after 12 h, transfected with different masses of the polypeptide plasmids obtained in step B of this example (0.05 g, Ol g and 0.15 g) or added at different concentrations in Example 1.
  • the synthetic peptides ( ⁇ . ⁇ , ⁇ , ⁇ and 100 ⁇ ) were repeated for 8 wells per concentration and cultured for 48 h under normal culture conditions.
  • the 0.3 g pEGFP-C2 plasmid was mixed with l g p-Anti-HBxP2# and co-transfected. After 24 hours, the green fluorescence was observed under an inverted fluorescence microscope, and the transfection efficiency was determined to be 70%.
  • HepG2-X cells or HepG2.2.15 cells in logarithmic growth phase were trypsinized to prepare cell suspension, and the number of cells was counted, diluted with sterile saline cells to IX 10 7 cells/ml, and stored in ice water. .
  • Twelve 4 to 6 weeks old female BALB/C nude mice were randomly divided into 2 groups: 1 control group, 0.2 ml of the above diluted cells were injected subcutaneously into the right forelimb of each mouse, and only 0.5 was injected. Ml sterilized distilled water (without peptide drug); 2 experimental group (administered dose of 10 mg / kg body weight).
  • the invention also explores the role of functional variants of polypeptides and functional fragments thereof.
  • the sequence shown in the table below is based on ⁇ 1 ⁇ -118 ⁇ 2# for amino acid addition (for example, adding an amino acid on the terminal side) or conservative amino acid substitution (for example, replacing one amino acid with another amino acid of the same type). owned.
  • the polypeptide fragment was artificially synthesized according to the sequence (method as above), and then the effect of the obtained polypeptide was observed by the above reporter gene and MTT method:
  • V-P-D-L-H-K-N-E-L-K-H-V-K-Y anti-HBxP2#-3 Pro-Glu-Leu-His-Lys-Asn-Glu-Leu-Lys-His-Val 4
  • anti-HBxP2# cctgatcttc acaaaatga actaaaacat gttaaatat 7 anti-HBxP2#-l atccctgatc ttcacaaaaa tgaactaaaa catgttaaat at 8 anti-HBxP2#-2 gtgcctgatc ttcacaaaaa tgaactaaaa catgttaaat at 9 anti-HBxP2#-3 ctgaacttc acaaaaatga actaaaacat gttaaatat 10 anti-HBxP2# -4 cctgatcttc accgcaatga actaaaacat gttaaatat 11 anti-HBxP2#-5 cctgatcttc acaaaaatga actaaaacat gttcgctat 12
  • the genes containing the five functional variants of ⁇ 1 ⁇ -118 ⁇ 2# have a certain inhibitory effect on the activity of the NF-KB promoter of HepG2-X cells and L-O2-X cells. * P ⁇ 0.05, **P ⁇ 0.01, Student's t test statistical analysis was performed. As shown in Fig. 4, the genes containing the five functional variants of ⁇ 1 ⁇ -118 ⁇ 2# inhibited the growth and proliferation of HepG2-X cells and L-O2-X cells. * PO.05, ** PO.01, Statistical analysis of Student's t test.
  • Example 5 Acute toxicity test of polypeptide drugs

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Description

抗乙型肝炎病毒 X蛋白多肽药物 技术领域
本发明涉及多肽药物领域, 具体涉及抗乙型肝炎病毒 X蛋白的多 肽和编码此多肽的多核苷酸, 以及它们的应用。 背景技术 说 肝癌是导致病人死亡的恶性肿瘤之一, 其恶性程度高。 据统计, 在中国, 肝癌死亡率仅次于胃癌, 居恶性肿瘤死亡率第二位。 中国每 年新增肝癌人数 30万人, 而每年肝癌死书亡人数达 11万。 乙型肝炎病毒 (hepatitis B vims, HBV) 感染可导致肝炎、 肝硬变 和原发性肝癌的发生。 HBV为长约 3.2Kb的 DNA病毒, 其开放阅读框 表达乙型肝炎病毒表面抗原 (HBsAg)、 乙型肝炎病毒核心抗原 (HBcAg)、 乙型肝炎病毒多聚酶和乙型肝炎病毒 X抗原(HBxAg) (或 称之为乙型肝炎病毒 X蛋白, 即 HBx)。 其中, HBx是 HBV DNA复制 所必须的因子。 因此, 抑制 HBx的功能, 意味着可抑制 HBV的感染, 以及由此导致的肝炎和肝硬变。 另外, HBx作为反式作用因子, 促进肝癌的生长和增殖, 称之为 癌蛋白。 在分子水平、 细胞水平和整体水平的研究也发现, HBx 具有 很强的促肝癌细胞增殖和迁移的作用。 转基因鼠实验也证明 HBx具有 显著的促进肝癌发生的作用。 大量研究结果显示, HBV感染的持续存 在可导致慢性肝病, 包括慢性肝炎、 慢性肝炎反复发作引起的肝组织 中纤维结缔组织增生后发生的肝硬变, 及在肝硬变基础上发生的肝癌。 在慢性肝病 (包括肝炎、 肝硬变和肝癌) 的发生发展过程中, HBx 发 挥了重要的作用。 因此, HBx是预防和治疗肝病的重要靶点。 目前, 肝癌的治疗以手术为主, 辅以介入疗法, 而化疗效果不理 想。 在临床手术的肝癌组织中 HBsAg和 HBxAg的表达阳性检出率高 达 80%甚至 90%以上。 因此, 由于 HBx是肝癌发生发展的重要致病因 子, 发现和发展其特异性抑制剂, 则具有重要的理论意义和实际临床 应用价值。 然而, 由于目前 HBx的三维构象解析尚未完成, 故难以通 过 HBx立体三维构象设计其化学抑制剂。 多肽片段可作为药物, 其在临床上已广为应用。 例如, 胸腺肽
(thymopeptide) 是从小牛胸腺中提取的胸腺五肽, 具有促进淋巴细胞 转化、 增强巨噬细胞吞噬活性的作用, 可用于治疗多种免疫缺陷病。 多肽药物的特点是药理作用明确, 安全性高, 并易于生产。 但是, 多 肽药物的发现难度较大, 大部分的多肽片段在体内的半衰期短, 直接 影响药效学作用。 发明内容
本发明涉及一种具有抑制乙型肝炎病毒 X蛋白(hepatitis B virus X protein, HBx) 功能活性的多肽, 其能在分子水平、 细胞水平和整体水 平上抑制 HBx的活性, 因而能抑制乙型肝炎病毒感染所造成的肝炎、 由肝炎反复发作引起的肝硬变、 以及在肝硬变基础上发生的肝癌。 该 多肽和它的肽模拟物, 包括它们的功能片段和功能变体, 以及编码这 些多肽、 肽模拟物或它们的功能片段、 功能变体的基因, 可广泛用于 预防和治疗乙型肝炎感染后的肝病, 包括肝炎、 肝硬变和肝癌。 一方面, 本发明提供了分离的多肽或肽模拟物, 其包含如 SEQ ID NO: 1所示的氨基酸序列、 或其功能片段或功能变体, 所述多肽或肽模 拟物具有抑制乙型肝炎病毒 X蛋白的功能, 并能抑制乙型肝炎病毒感 染后的慢性肝病的发生和发展。 所述乙肝病毒感染后的慢性肝病可以 包括肝炎, 由肝炎反复发作引起的肝硬变, 以及在肝硬变基础上发生 的肝癌。 在本发明的一些具体实施例中, 上述多肽或肽模拟物, 其包含的 氨基酸序列、 其功能片段或功能变体可以具有与 SEQ ID NO: 1所示的 氨基酸序列至少 70%, 或者 80%, 或者 90%, 以及甚至更高的相同性。 优选地, 这些多肽或肽模拟物包含如 SEQ ID NOs: 1-6中所示的任一种 氨基酸序列。 另一方面, 本发明提供分离的多核苷酸, 所述多核苷酸包含编码 如 SEQ ID NO: 1所示的氨基酸序列、 其功能片段或功能变体的多核苷 酸;或者是与包含编码如 SEQ ID NO: 1所示的氨基酸序列、 其功能片 段或功能变体的多核苷酸互补或严格杂交的多核苷酸。 优选地, 这些 多核苷酸为编码 SEQ ID NOs: 1-6所示的氨基酸序列的多核苷酸, 或者 为与编码 SEQ ID NOs: 1-6所示的氨基酸序列的多核苷酸互补或严格杂 交的多核苷酸。 另一方面, 本发明还提供含有外源多核苷酸的重组表达载体, 其 包含编码如 SEQ ID NO: 1所示的氨基酸序列、 其功能片段或功能变体 的多核苷酸;或者包含与编码如 SEQ ID NO: 1所示的氨基酸序列、 其 功能片段或功能变体的多核苷酸互补或严格杂交的多核苷酸。 优选地, 这些重组表达载体权利包含编码 SEQ ID NOs: 1-6所示的氨基酸序列的 多核苷酸, 或者包含与编码 SEQ ID NOs: 1-6所示的氨基酸序列的多核 苷酸互补或严格杂交的多核苷酸。 本方明还提供包括上述重组表达载 体的宿主细胞。 此外, 本发明也提供上述多肽或肽模拟物、 核苷酸以及重组表达 载体在制造抗乙肝病毒感染后的慢性肝病的药物中的应用。 在一个具 体实施例中, 所述药物可为乙肝的治疗性疫苗。 所述药物可以包含药 物组合物, 其可以包含任选的药物载体。 本发明中, 所述的多肽 (包括其功能片段或变体) 及其肽模拟物, 作为 HBx的有效抑制因子, 具有抑制 HBx的生物活性的能力, 能抑制 乙型肝炎病毒感染所造成的肝炎, 由此引发的肝硬变和在肝硬变基础 上发生的肝癌。 值得强调的是, 本发明提供的上述多肽具有明显的药 效学作用, 因此可以成为治疗由于乙肝病毒感染所导致的肝病的有效 药物。 附图说明
图 1. 应用高压液相色谱 (HPLC ) 对纯化后人工合成多肽 Anti-HBxP2#的分析结果。
图 2. 应用报告基因检测本发明的多肽基因质粒对表达 HBx蛋白 的肝癌细胞的影响。结果显示 - 1^-1¾ ?2#对 HepG2-X细胞、 L-O2-X 细胞和 HepG2.2.15细胞中 NF-κΒ启动子的活性具有抑制作用, 呈剂量 依赖性,而对无 HBx的 HepG2肝癌细胞和无 HBx的 L-O2肝细胞无影 响。 相似地, - 1^-1¾^2#的 5种功能变体基因 (0.15 μ g/well) 对 HepG2-X细胞和 L-O2-X细胞中 NF-κΒ启动子的活性具有一定的抑制 作用。 * P<0.05, ** P<0.01, 进行 Student's t test统计学分析。
图 3. 检测本发明的多肽在细胞水平对表达的 HBx 蛋白激活 NF-κΒ启动子的活性的影响。 结果显示, 人工合成的多肽 Anti-HBxP2# 对表达 HBx蛋白的 HepG2-X细胞、 L-O2-X细胞和 HepG2.2.15细胞中 的 NF-κΒ启动子的活性具有抑制作用, 且该抑制作用呈剂量依赖性, 但对无 HBx 表达的 HepG2 肝癌细胞和 L-O2 肝细胞则无此影响。 *
P<0.05, ** P<0.01 , 进行 Student's t test统计学分析。
图 4. 应用 MTT检测转染本发明的多肽基因质粒对表达 HBx蛋白 的肝癌细胞的影响。结果显示 - 1^-1¾ ?2#对 HepG2-X细胞、 L-O2-X 细胞和 HepG2.2.15细胞生长和增殖具有抑制作用, 且其效果呈剂量依 赖性, 但对无 HBx表达的 HepG2肝癌细胞和 L-O2肝细胞则无影响。 ρ-Αη1ί-ΗΒχΡ2#ή¾ 5种功能变体基因 (0.15 μ g/well) 对 HepG2-X细胞 和 L-O2-X细胞的生长和增殖也具有抑制作用。 * PO.05, ** PO.01, 进行 Student's t test统计学分析。
图 5. 应用 MTT检测本发明的多肽对肝癌细胞的生长的影响。 结 果显示, 应用人工合成的多肽 1^-1¾ ?2#对1½ 02- 细胞、 L-O2-X 细胞和 HepG2.2.15细胞生长和增殖具有抑制作用, 且其作用呈剂量依 赖性。 而对无 HBx表达的 HepG2肝癌细胞和 L-O2肝细胞无影响。 * P<0.05, ** P<0.01 , 进行 Student's t test统计学分析。
图 6. 人工合成的多肽八1^-1¾ ?2#对 HepG2-X细胞的作用。 裸 鼠接种实验结果显示, 应用人工合成的多肽 八1^-118^2#对 HepG2-X 细胞生长和增殖具有明显的抑制作用。 ** P<0.01, Student's t test统计 学分析
图 7. 人工合成的多肽八1^-1¾ ?2#对 HepG2.2.15细胞的作用。裸 鼠接种实验结果显示,应用人工合成的多肽 1^-118^2#对 HepG2.2.15 细胞生长和增殖具有明显的抑制作用。 ** P<0.01, Student's t test统计 学分析
图 8. 构建多肽真核表达载体的连接示意图。 具体实施方式
本发明中, 包括说明书和权利要求书, 使用的下列术语除非特别 说明, 具有如下的含义:
"分离的" 一词是指将物质从它原始的环境 (例如, 若是自然产 生的就指其天然环境) 分离出来。 比如说, 一个自然产生的多核苷酸 或多肽存在于活动物中就是没有被分离出来, 而同样的多核苷酸或多 肽同一些或全部在自然系统中与之共存的物质分开就是分离的。 这样 的多核苷酸或多肽可以是某一载体的一部分, 也可以是某一组合物的 一部分。 既然载体和组合物不是它的天然环境的成分, 它们仍然是分 离的。
"纯化的" 一词意指已经在纯度上提高的。 "纯度" 在这是相对术 语, 并不必要地解释为绝对纯度。 例如, 纯度可以是至少约 50%, 或 可以是大于 60%、 70%、 80%、 90%、 或可以是 100%。 如本发明中所用, 分离的物质是从其原始环境中分离出来。 活体 细胞内的天然状态下的多核苷酸和多肽是没有分离的, 但同样的多核 苷酸和多肽如从天然状态中同存在的其他物质中分开, 则为分离的, 同时纯度上得到了提高, 也因此是纯化的。 "核酸"、 "核酸序列" 或 "碱基序列" 是指核苷酸, 寡核苷酸或 多核苷酸及其片段或部分。 本发明的核酸能够以 RNA (例如, mRNA) 的形态、 或 DNA的形态 (例如, cDNA或基因组 DNA) 存在。 DNA 可以是双链, 也可以是单链。 单链 DNA或 RNA可以是编码链 (有义 链)、 或非编码链 (反义链) 中任一种。 另外, 本发明的多核苷酸也可 以在其 5' 端或 3' 端融合编码标签标记 (标签序列或标记物序列) 的 多核苷酸。 它们可以是合成的或是从天然来源获得的 (例如, 分离和 / 或纯化), 其可以包含天然的、 非天然的或者修饰过的核苷酸, 并且其 可以包含天然的、 非天然的或改变的核苷酸之间的键, 诸如氨基磷酸 酯键或硫代磷酸酯键, 用来代替在未修饰的寡核苷酸中核苷酸之间存 在的磷酸二酯键。 所谓 "氨基酸序列" 或 "多肽" 是指肽、 寡肽、 多肽或蛋白质及 其部分片段, 其间以肽键相连接的氨基酸。 当本发明中的 "氨基酸序 歹 IJ " 涉及一种天然存在的蛋白质分子的氨基酸序列时, 这种 "多肽" 或 "蛋白质" 并不意味着将氨基酸序列限制为与所述蛋白质分子相关 的完整的天然氨基酸序列。 本发明的氨基酸序列可以含有附加的肽。 作为附加的肽, 如多组氨酸标签 (His-tag)、 或 Myc、 FLAG等表位标 记的肽为例。 "缺失" 是指在氨基酸序列或核苷酸序列中一个或多个氨基酸或 核苷酸的缺失。
"插入" 或 "添加" 是指在氨基酸序列或核苷酸序列中的改变导 致与天然存在或改变前的分子相比, 一个或多个氨基酸或核苷酸的增 加。 "置换" 是指由不同的氨基酸或核苷酸替换一个或多个氨基酸或 核苷酸。 "缺失、 置换或添加一个或多个氨基酸或核苷酸" 则是指, 利用 定位诱变法等公知的突变核酸或多肽的制作法缺失、 置换或添加一个 或多个数目的氨基酸或核苷酸, 或对天然存在的核酸或多肽的上述突 变进行分离纯化。 对氨基酸的突变, 可以包含有一个或多个氨基酸残 基为 D-型构象的氨基酸、 自然界存在的稀有氨基酸、 或者是人工修饰 的氨基酸, 这些氨基酸可以是也可以不是由遗传密码子编码的。 与此 相类似的, 诱导核酸发生突变, 可以包括自然界天然存在的核苷酸, 也可以包括具有修饰的核苷酸。 多肽的 "功能片段" 是指本发明的多肽的任何部分, 所述部分保 留其作为一部分的多肽 (即 "亲本" 多肽) 的基本相似或相同的生物 学活性和功能。 多肽的 "功能变体" 是指基本上保持如所述多肽或氨基酸序列相 同或相似的生物学功能或活性的氨基酸序列, 它们可以包括, 例如, 1) 原氨基酸序列中一个或多个氨基酸残基有缺失和 /或一个或多个氨基酸 残基被添加;或者 2) 原氨基酸序列中一个或多个氨基酸残基被保守或 非保守氨基酸残基置换;或者 3) 原氨基酸序列中一个或多个氨基酸残 基上的某个基团被其它基团取代;或者 4) 原氨基酸序列和另外的分子 或化合物 (比如糖、 脂类、 聚乙二醇等) 的融合;或者 5) 原有的氨基 酸序列与添加的氨基酸序列融合进而形成的多肽序列 (如前导序列或 分泌序列或用来纯化此多肽的序列等) ;或者 6) 原氨基酸序列的逆反 类似物;或者 7) 以上各种情况的混合。 本发明中, 氨基酸序列的功能 变体, 可以包含有一个或多个氨基酸残基构象为 D-型的氨基酸, 自然 界存在的稀有氨基酸、 或人工修饰的氨基酸, 这些氨基酸可以是也可 以不是由遗传密码子编码的。 多肽的 "逆反类似物" 是指包括反转的亲本多肽氨基酸序列的多 肽, 以致所述逆反类似物的氨基酸序列 (当从 N端向 C端读取时) 与 当从 C端向 N端读取时的亲本多肽的氨基酸序列相同;此外, 逆反类 似物中每一个氨基酸是该氨基酸的 D异构体, D异构体与 L异构体相 反。例如, 三肽 Val-Ala-Gly的逆反类似物具有氨基酸序列 Gly-Ala-Val, 其中每个氨基酸是 D异构体。 本发明中, "肽模拟物"是指具有与相对应多肽基本相同的通用结 构且具有例如能增加其稳定性或生物学功能的修饰的化合物。 肽模拟 物包括例如包含与相对应多肽的相同氨基酸序列的那些化合物, 但在 其中两个或更多个氨基酸之间, 肽模拟物具有改变的主链。 所述肽模 拟物可以包括合成的或非天然存在的氨基酸, 用来代替天然存在的氨 基酸。 本发明中, 核苷酸序列的 "简并变异体" 是这样的多核苷酸序列, 其与亲本核苷酸序列有区别, 但编码的蛋白质或多肽或亲本核苷酸序 列所编码的蛋白质或多肽一样。 "核酸杂交" 在本领域已公知 (参见, 例如, Sambrook 等,
Molecular Cloning: A Laboratory Manual, 3rd Ed., Cold Spring Harbor Laboratory, 2001)。 通常, 温度越高, 盐浓度越低, 则严谨性变得越 高 (难以杂交), 从而能够取得更加相同的多核苷酸。 适合的杂交温度 根据碱基序列或其碱基序列的长度而异。 另外, 本发明还涉及在 "严 格条件" 下杂交。 本发明中, "严格条件" 是指, 在较低离子强度和较 高温度下的杂交和洗脱。 例如, 在 42°C的条件下、 在 (50%甲酰胺、 5 X SSC ( 150 mM NaC 15mM柠檬酸三钠)、 50 mM的磷酸钠 (pH7.6)、 5 X Denhardt溶液、 10%硫酸葡聚糖、 和 20 g/ml的变性剪切鲑鱼精子 DNA) 中孵育过夜, 然后在 65° (:条件下用 0.1 X SSC洗脱。 "同源性" 是指互补的程度, 可以是部分同源, 也可以是完全同 源。 "部分同源" 是指一种部分互补的序列, 其可部分抑制完全互补的 序列与靶核酸的杂交。 这种抑制可通过在严格性程度降低的条件下进 行杂交 (Southern 印迹或 Northern印迹等) 来检测。 基本同源的序列 或杂交探针可竞争和抑制完全互补的序列与靶序列在严格性程度降低 的条件下的结合。 当然, 严格性降低的条件并非允许非特异性的结合, 两条序列相互结合, 仍然需要特异性或选择性的相互作用。 氨基酸序列或核苷酸序列的 "相同性" 或 "同一性" 百分率是指 在两种或多种氨基酸或核苷酸序列比较中, 序列相同或相似的百分率。 有很多本领域技术人员熟知的方法测定相同性百分率, 如通过 MEGALIGN程序 (Lasergene software package, DNASTA, Inc., Madison, WI) 0 MEGALIGN程序可根据不同的方法如 Cluster法比较两种或多种 序列 (参见 Higgins & Sharp, Gene 73:237-244, (1988)) , Cluster法通过 检查所有配对之间的距离将各组序列排列成簇, 然后将簇以成对或成 组分配。 两个氨基酸序列如序列 A和序列 B之间的相同性百分率可以 通过下式计算:
[ (序列 A与序列 B之间匹配的残基个数) I (序列 A的残基数- 序列 A中间隔残基数 -序列 B中间隔残基数) ] X 100% 同理,也可以通过 Cluster法或用本领域已知的方法,如 Jotun Hein 的方法 (参见 Hein J., Methods in Emzumology 183:625-645, 1990) , 测 定核酸序列的相同性百分率。 "重组表达载体" 是指遗传修饰的寡核苷酸或多核苷酸重组体, 该重组体克隆有编码 mRNA、 蛋白、 多肽、 或肽的核苷酸序列, 当该 表达载体进入宿主细胞后可表达相应的 mRNA、 蛋白、 多肽、 或肽。 本发明中, 重组表达载体可以包含任意类型的核苷酸序列, 但不仅限 于是 DNA或 RNA, 可为单链或双链、 人工合成或来自天然, 也可为 非天然的或改变的核苷酸。 核苷酸之间的键可以是天然存在的、 也可 以是非天然存在的或修饰的。 本发明中, "乙肝病毒感染后的慢性肝病" 是指乙型肝炎病毒感 染后导致的肝病, 包括慢性肝炎、 慢性肝炎反复发作引起的肝组织中 纤维结缔组织增生后发生的肝硬变, 及大部分在肝硬变基础上发生的 肝癌。 肝癌的发生是乙型肝炎病毒持续感染的结果, 这种肝癌可称之 为乙肝后肝癌。 本发明中, "治疗" 和 "预防" 以及由它们派生的词语, 但并不 意味是 100%的或完全的治疗或预防,可以认定为本领域技术人员所认 同的治疗或预防程度。 本发明中的 "预防" 可理解为延迟疾病、 或其 症状或病症的发作。 多肽
本发明涉及分离或纯化的多肽。 该多肽可以是包含、 基本由、 或 由氨基酸序歹1 J Pro-Asp-Leu-His-Lys-Asn-Glu-Leu-Lys-His-Val-Lys -Tyr (SEQ ID NO: 1)所组成的多肽。研究证实这些多肽可以明显抑制 HBx 的活性, 具有明显抑制乙肝病毒感染后的慢性肝病的发生和发展的作 用, 尤其重要的是, 这些多肽可以抑制感染有乙肝病毒的肝癌细胞的 生长和增殖。 所述抑制在分子水平、 细胞水平和整体水平均有体现。 如, 所述 多肽在分子水平上可以抑制由 HBx激活的核因子 κΒ (NF-κΒ) 的启动 子活性, 从而体现所述多肽对 HBx的有效抑制, 且其抑制效果呈量效 关系;所述多肽在细胞水平也体现出对含 HBx基因的肝癌细胞的生长 和增殖功能的有效抑制, 且其抑制效果也呈量效关系;通过裸鼠接种 方法显示, 所述多肽在整体水平能有效抑制表达 HBx的肝癌细胞的成 瘤能力。 然而, 所述多肽对无表达 HBx肝癌细胞中 NF-κΒ的启动子活 性无影响, 对无表达 HBx肝癌细胞的生长和增殖也无影响。 本发明还提供所述本发明多肽的各种功能片段。 所述功能片段可 以为本发明的多肽的连续氨基酸序列的任何片段, 条件是其与亲本多 肽相比, 能以相似程度、 相同程度、 或更高程度保留亲本多肽的生物 活性, 例如, 抑制 HBx的活性。 参照亲本多肽, 所述功能片段可以包 括, 例如, 亲本多肽的约 10%、 20%、 30%、 40%、 50%、 60%、 70%、
80%、 90%、 95%、 100%、 105%、 110%、 120%、 150%、 200%或更高 的活性。 所述功能片段还可以在上述连续氨基酸序列片段的氨基或羧 基端、 或其氨基以及羧基两端, 包含额外的氨基酸, 例如, 与亲本多 肽的氨基酸序列中不同的氨基酸。 理想的是所述额外的氨基酸不妨碍 所述功能片段的生物学功能, 例如, 抑制 HBx的活性, 并有效地抑制 癌细胞, 优选是肝癌细胞, 尤其是感染有乙肝病毒的肝癌细胞的生长 和增殖。 更为理想的是, 当与所述亲本多肽的生物学活性相比较时, 所述额外的氨基酸能够导致增强的生物学活性。 本发明的多肽的功能 片段包括与所述亲本多肽具有至少 70%的序列同一性的氨基酸序列为 优选序列。 在一个具体的实施例中, 本发明的功能片段包括在所述亲 本多肽的氨基端 (N端) 和 /或羧基端 (C端) 添加氨基酸, 例如一个 或两个氨基酸。 另外, 本发明的多肽及其功能片段的功能变体也包括在本发明范 围之内。 所述本发明的多肽及其功能片段的功能变体保留与所述亲本 多肽或亲本功能片段基本相似或相同的生物活性, 例如, 抑制 HBx的 活性, 以及有效地抑制肝癌细胞, 尤其是感染有乙肝病毒的肝癌细胞 的生长和增殖。 参照所述亲本多肽或亲本功能片段, 所述功能变体可 以与所述亲本多肽或亲本功能片段的氨基酸序列至少有约 50%、 60%、 70%、 80%、 90%、 95%或 100%的同一性。 优选地, 本发明的多肽及 其功能片段的功能变体包含, 与所述亲本多肽或亲本功能片段具有至 少 70%的序列同一性。 更优选的, 本发明的多肽及其功能片段的功能 变体包含与所述亲本多肽或亲本功能片段只有 1-3个氨基酸的区别。在 一些特别优选的实施例中, 本发明的多肽及其功能片段的功能变体与 所述亲本多肽或亲本功能片段仅仅只有 1个氨基酸的区别。 所述本发明的多肽及其功能片段的功能变体可以包括具有至少一 个保守氨基酸置换的亲本多肽或亲本功能片段氨基酸序列。 具体来说, 所述本发明的多肽及其功能片段的功能变体可以包含具有 2个、 3个、 4个、 5个、 或更多个保守氨基酸置换的亲本多肽或亲本功能片段的氨 基酸序列。 另外, 所述本发明的多肽及其功能片段的功能变体也可以 包括具有至少一个非保守氨基酸置换的亲本多肽或亲本功能片段的氨 基酸序列。 具体来说, 可以包括具有 2个、 3个、 4个、 5个、 或更多 个非保守氨基酸置换的亲本多肽或亲本功能片段的氨基酸序列。 在这 些情形中, 对于所述的氨基酸置换, 优选的是不妨碍或抑制所述功能 变体的生物学功能或活性。 更优选地, 所述氨基酸置换提高本发明的 多肽及其功能片段的功能变体的生物学功能或活性, 以致当与所述亲 本多肽或亲本功能片段相比较时, 所述功能变体的生物学功能或活性 提局。 优选地, 本发明的多肽及其功能片段的功能变体包括一个或多个 保守氨基酸置换。 在本领域内, 保守氨基酸置换是公知的, 它指的是 这样的氨基酸置换, 即, 其中一个具有某种物理和/或化学特性的氨 基酸被换为另一个具有相同化学或物理特性的氨基酸。 本领域技术人 员了解, 保守性的氨基酸置换可以不造成蛋白质的结构或功能的显著 改变。 典型的保守性置换包括, 例如, 用酸性氨基酸置换另一个酸性 氨基酸 (例如, AsP 或 Glu), 用具有非极性侧链的氨基酸置换另一个 具有非极性侧链的氨基酸 (例如, Ala, Gly, Val, He, Leu, Met, Phe, Pro, Trp, Val等) , 用碱性氨基酸置换另一个碱性氨基酸 (Lys, Arg等) , 用具有极性侧链的氨基酸置换另一个具有极性侧链的氨基酸 (Asn,CyS
Gln, Ser, Thr, Tyr等) , 用芳香氨基酸 (Trp, Phe, Tyr等) 置换另一个 芳香氨基酸等。 另外, 本发明的多肽及其功能片段的功能变体还可以包括本发明 的任意多肽或功能片段的逆反类似物。 本发明中, "逆反类似物" 是 指包括反转的亲本多肽氨基酸序列的多肽, 以致所述逆反类似物的氨 基酸序列 (当从 N端向 C端读取时) 与当从 C端向 N端读取时的亲本 多肽的氨基酸序列相同。 此外, 关于逆反类似物, 每一个氨基酸是该 氨基酸的 D异构体, D异构体与 L异构体相反。例如,三肽 Val-Ala-Gly 的逆反类似物具有氨基酸序列 Gly-Ala-Val, 其中每个氨基酸是 D异构 体。 本发明中, 所述此类的功能变体优选包括 SEQ ID NO: 1的逆反类 似物。 本发明的多肽 (包括功能片段) 及其功能变体可以是任意长度的, 即, 可以包括任意数目的氨基酸, 条件是所述多肽 (包括功能片段) 及其功能变体保留必要的生物学活性, 例如, 抑制 HBx的活性, 以及 有效地抑制癌细胞, 优选是肝癌细胞, 尤其是感染有乙肝病毒的肝癌 细胞。 举例来说, 本发明的多肽 (包括功能片段) 及其功能变体可以 是 4-2000个氨基酸长, 如长度为 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 18, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, 125, 150, 175, 200, 300, 400,
500, 700, 800, 1000或更多。 优选地, 本发明的多肽长度为 6-20个氨基 酸, 并满足作为多肽药物的药效学和半衰期的要求。 在一个实施例中, 本发明的多肽及其功能片段的功能变体与亲本 多肽 SEQ ID NO :1具有一个氨基酸的差别,其具有与亲本多肽 SEQ ID
NO: 1相似的生物学活性和功能, 例如, 抑制 HBx的活性, 以及有效 地抑制癌细胞, 优选是肝癌细胞, 尤其是表达 HBx的肝癌细胞。 更具 体地, 本发明的多肽及其功能片段的功能变体包括 SEQ ID NOs: 2-6。 本发明还提供所述多肽 (包括功能片段和功能变体) 的肽模拟物。 在一个优选的实施方案中, 所述肽模拟物是拟肽。 拟肽是指这样的肽 模拟物, 其中每个氨基酸的侧链粘附在氨基酸的氮原子上而不是 α 碳 上。 例如, 拟肽可以被视为 Ν—置换的甘氨酸, 其具有 NRCH2CO通用 结构的重复单位, 并且其具有与相对应的多肽相同或基本相同的氨基 酸序列。 在另一个优选的实施方案中, 所述肽模拟物包括改变的主链, 其中在每个氨基酸之间的键是甲基化的。 在这一点上, 所述肽模拟物 可以包括下述结构的甲基化肽主链:
...NCH3 - Ca- CO - NCH3 - Ca- CO...
本发明的多肽 (包括功能片段和功能变体) 以及肽模拟物中, 可 以包括用合成的氨基酸来置换天然存在的氨基酸。 所述合成的氨基酸 在本领域内是已知的, 包括, 例如, 氨基环己垸羧酸、 正亮氨酸、 ot- 氨基正癸酸、 高丝氨酸、 S-乙酰氨基甲基 -半胱氨酸、 反式 -3-羟脯氨酸、 反式 -4-羟脯氨酸、 4-氨基苯丙氨酸、 4-苯甲酰苯丙氨酸、 4-硝基苯丙氨 酸、 4-氯苯丙氨酸、 4-羧基苯丙氨酸、 β-苯基丝氨酸、 β-羟基苯丙氨酸、 苯基甘氨酸、 a-萘基丙氨酸、 环己基丙氨酸、 环己基甘氨酸、 二氢吲哚 -2-羧酸、 1,2,3,4-四氢异哇琳 -3-羧酸、 氨基丙二酸、 氨基丙二酸单酞胺、 Ν'-苄基 -Ν'-甲基-赖氨酸、 Ν',Ν'-二苄基-赖氨酸、 6-羟基赖氨酸、 鸟氨 酸、 a-氨基环戊垸羧酸、 a-氨基环己垸羧酸、 a-氨基环庚垸羧酸、 a-(2- 氨基 -2-降冰片垸) -羧酸、 α,γ-二氨基丁酸、 α,β-二氨基丙酸、 高苯丙氨 酸、 和 a-叔-丁基甘氨酸。 本文所述的本发明的多肽 (包括功能片段和功能变体) 以及肽模 拟物还可以包含细胞-穿透肽 (CPP) 。 所述 CPP促进本发明多肽穿过 细胞膜并且进入细胞内。 CPPs 在本领域中是已知的。 参见, 例如, Deshayes 等, 细胞分子生命科学 (Cell. Mol. Life Sci.) 62: 1839-1849 (2005) ; EI-Andaloussi 等, 现代药物设计 (Curr. Pharm. Design.) 11 :3597-3611(2005)。 所述 CPP可以是本领域己知的那些中的任一种。 本发明的多肽 (包括功能片段和功能变体) 以及肽模拟物还可以, 例如, 被脂化 (例如, 脂肪酸化) 、 糖基化、 酰胺化、 羧酸化、 磷酸 化、 酯化、 N-酰化、 通过二硫键环化、 转化成酸加成盐、 二聚体化或 多聚体化, 和 /或缀合化。 举例来说, 本发明所述多肽 (包括功能片段和功能变体) 以及肽 模拟物可以是脂化衍生物。 所含的脂质分子可以包括本领域己知的任 何脂质, 例如, 脂肪酸, 磷脂基团, 糖磷脂酰肌醇, 磷脂酰丝氨酸, 磷脂酰乙醇胺, 鞘磷脂, 磷脂酰胆碱, 心磷脂, 磷脂酰肌醇, 磷脂酸, 溶血磷酸甘油酯, 和胆固醇基团。 优选地, 所述脂化衍生物是脂肪酸 衍生物, 所述脂肪酸分子可以是任何 C8-C20脂肪酸, 例如, 月桂酸、 棕榈酸、 肉豆蔻酸、 硬脂酸、 油酸、 亚油酸、 亚麻酸、 花生四烯酸、 二十碳五烯酸、 芥酸、 或花生酸等。 所述脂肪酸还可以在任意碳原子 上任选地包含其他官能团, 例如, 一个或多个氨基。 所述脂肪酸分子 可以附着在本发明多肽 (包括功能片段和功能变体) 以及肽模拟物的 任何适当的部分。 例如, 在本发明的多肽氨基端、 羧基端、 或氨基和 羧基两端包括脂肪酸分子。 脂肪酸分子可以直接或通过连接体附着到 本发明的多肽 (包括功能片段和功能变体) 以及肽模拟物上。 本发明的多肽 (包括功能片段和功能变体) 以及肽模拟物, 包括 其衍生物例如脂肪酸衍生物, 还可以是单体肽、 二聚肽或多聚体肽。 本发明的多肽 (包括功能片段和功能变体) 以及肽模拟物可以通 过本领域技术人员已知的方法获得(参见,例如, Chan 等, Fmoc Solid
Phase Peptide Synthesis, 牛津大学出版社, 牛津, 英国, 2005 ; Reid, R., Peptide and Protein Drug Analysis, Marcel Dekker Company, 2000 , '以及 美国专利号 5,449,752)。此外,也可由核酸重组方法生产多肽而得到(参 见, 例如, Sambrook等, 分子克隆:实验室手册 (Molecular Cloning: A Laboratory Manual),第 3版,冷泉港出版社 (Cold Spring Harbor Press ), 冷泉港, 纽约, 2001) 。 此外, 本发明的一些多肽 (包括其功能片段以 及功能变体) 可以从如植物、 细菌、 昆虫、 哺乳动物如大鼠、 人等分 离和/或纯化。 分离和纯化的方法在本领域内也是公知的。 备选地, 本文所述的多肽 (包括其功能片段以及功能变体) 可以从商业公司合 成后购买获得。 多核苷酸
本发明还提供编码任一种所述的本发明多肽 (包括功能片段和功 能变体) 的分离的多核苷酸。 所述多核苷酸包含编码任一种本发明多 肽 (包括功能片段和功能变体) 编码序列, 例如编码 SEQ ID NO: 1的 核苷酸序列 SEQ ID NO: 7, 以及这些编码序列的任一种简并变异体; 备选地, 所述多核苷酸还可以包含附加编码序列和 /或非编码序列。 另一方面, 本发明还提供包含这样的分离的多核苷酸或其片段, 其所 包含的核苷酸序列与本发明所述的任一种核酸的核苷酸序列互补或在 严格条件下与本发明所述的任一种多核苷酸的核苷酸序列杂交。 本发明还提供上述多核苷酸的变异体, 其编码与本发明多肽 (包 括功能片段和功能变体) 有相同的氨基酸序列的多肽或多肽的功能片 断、 功能变体。 此多核苷酸的变异体可以是天然发生的等位变异体或 非天然发生的变异体。 这些核苷酸变异体包括取代变异体、 缺失变异 体和插入变异体。 如本领域所知的, 等位变异体是一个多核苷酸的替 换形式, 它可能是一个或多个核苷酸的取代、 缺失或插入, 但不会从 实质上改变其编码的多肽的功能。 本发明的多核苷酸可以是天然存在通过纯化而得到, 也可以通过 重组产生, 或者可以基于化学合成/或酶连接反应, 使用本领域已知 的方法而构建。 本发明的多核苷酸可以含有天然存在的核苷酸, 也可 以包含具有修饰的核苷酸。 所述具有修饰的核苷酸设计成增加分子的 生物学稳定性或增加在杂交时形成的双链体的物理稳定性 (例如, 硫 代磷酸衍生物和吖啶置换的核苷酸) , 包括, 例如, 5-氟尿嘧啶、 5- 溴尿嘧啶、 5-氯尿嘧啶、 5-碘尿嘧啶、 次黄嘌呤、 黄嘌呤、 4-乙酰胞嘧 啶、 5- (羧基羟甲基) 尿嘧啶、 5-甲氧基氨基甲基 -2-硫尿嘧啶、 尿嘧啶 -5-羟基乙酸、 5-甲基 -2-硫尿嘧啶、 2-硫胞嘧啶、 2-硫尿嘧啶、 4-硫尿嘧 啶、 5-甲基尿嘧啶、 尿嘧啶 -5-羟基乙酸甲基酯、 3- (3-氨基 -3-N-2-羧丙 基) 尿嘧啶、 和 2, 6-二氨基嘌呤等。 并且, 其可以包含天然的或改变 的核苷酸之间的键, 诸如氨基磷酸酯键或硫代磷酸酯键, 用来代替天 然存在的磷酸二酯键。 优选的, 本发明的核酸是重组产生的。 重组表达载体
本发明还提供包括任一种本发明多核苷酸的重组表达载体。 所述 重组表达载体可以是任何适当的重组表达载体, 并且可以用于转化或 转染任何适当的宿主细胞。 适当的载体包括设计用于繁殖和扩增或用 于表达, 或同时用于二者, 如质粒或病毒。 所述载体可以通过下列自 由组合: pUC系列、 pcDNA系列, pBluescript、 pET系列、 pGEX 系 列和 pEX系列。还可以使用噬菌体载体, 诸如 GT10、 GTI K λΖαΡΙΙ. EMBL4等。 植物表达载体可以包括 pBI01、 pBI101.2. pBI101.3. pBI121 禾口 pBIN19。 动物表达载体可以包括 pEUK-Cl、 pMAM和 pMAMneo。 在一个具体的实施例中, 本发明涉及使用 pcDNA系列的质粒。 可以使用标准重组 DNA技术制备本发明的重组表达载体。可以制 备环形的或线性的表达载体构建体, 以包含在原核或真核宿主细胞中 起作用的复制系统。 理想地, 所述重组表达载体包括调控序列, 诸如 转录和翻译起始与终止密码子。 所述调控序列对于所述载体所引入的 宿主类型 (例如, 细菌、 真菌、 植物、 或动物) 是特异性的。 所述重组表达载体可以包括一种或多种标记基因, 其允许选择转 化的或转染的宿主。 标记基因包括对杀生物剂的抗性, 例如, 对抗生 素、 重金属等的抗性, 在营养缺陷型宿主中互补以提供原养型, 以及 催化生物荧光等等。 对于本发明表达载体合适的标记基因包括, 例如, 新霉素 /G418抗性基因、 荧光酶素报告基因、 潮霉素抗性基因、 组氨 醇抗性基因、 四环素抗性基因、 和氨苄青霉素抗性基因。 所述重组表达载体可以包括天然的或非天然的启动子。 启动子的 选择, 例如, 强、 弱、 诱导型、 组织特异型和发育特异型的, 在技术 人员的普通技能之内。 相似地, 核苷酸序列与启动子的组合也在技术 人员的技能之内。 启动子可以是非病毒启动子或病毒启动子, 例如, 巨细胞病毒 (CMV) 启动子、 SV40启动子、 RSV启动子、 以及在鼠干 细胞病毒长末端重复中存在的启动子。 本发明的重组表达载体可以设 计用于瞬时表达、 用于稳定表达、 或用于二者。 此外, 所述重组表达 载体可以制备成用于组成型表达或用于诱导型表达。 此外, 所述重组表达载体可以包括自杀基因。 "自杀基因" 是指 在细胞中表达后导致细胞死亡的基因。 自杀基因表达后可影响细胞对 某种试剂如药物的敏感性, 进而引起细胞死亡。 自杀基因在本领域内 是已知的 (参见, 例如, 自杀基因治疗: 方法和综述 (Suicide Gene
Therapy: Methods and Reviews), Springer, Caroline J. (Cancer Research UK Centre for Cancer Therapeutics at the Institute of Cancer Research, Sutton, Surrey, UK), Humana Press, 2004), 例如单纯疤疹病毒 (HSV) 胸苷激酶 (TK) 基因和嘌呤核苷磷酸酶, 以及硝基还原酶等。 宿主细胞
本发明还提供包括本文所述的任何一种重组表达载体的宿主细 胞。 宿主细胞是包含本发明重组表达载体的任何类型的细胞。 宿主细 胞可以是真核细胞, 例如, 植物、 动物、 真菌、 或藻类, 或者可以是 原核细胞, 例如, 细菌或原生动物。 宿主细胞可以是培养的细胞或原 代细胞, 即, 直接从生物体如人分离的原代细胞。 宿主细胞可以是贴 壁细胞或悬浮的细胞, 即, 在混悬液中生长的细胞。 适当的宿主细胞 在本领域内是己知的, 包括, 例如, DH5a大肠杆菌细胞、 中国仓鼠卵 巢细胞、 猴 VERO细胞、 COS细胞、 HEK293细胞, 等等。 为了扩增 或复制所述重组表达载体的目的, 所述宿主细胞优选是原核细胞。 为 了产生重组修饰的多肽或蛋白的目的, 宿主细胞优选是哺乳动物细胞。 人源细胞为优选的宿主细胞。 宿主细胞可以是任何细胞类型, 可以来 自任何类型的组织, 并且可以是任何发育阶段的。 本发明还提供包括至少一种本文所述的宿主细胞的细胞群体。 所 述细胞群体可以是异种群体, 包括包含任一种所述重组表达载体的宿 主细胞, 以及至少一种其它细胞, 例如, 不包含任何所述重组表达载 体的宿主细胞 (例如, T 细胞) , 或除 T细胞之外的细胞, 例如, B 细 胞、 巨噬细胞、 嗜中性粒细胞、 红细胞、 肝细胞、 内皮细胞、 上皮细 胞、 肌细胞、 脑细胞, 等等。 备选地, 所述细胞群体可以是基本同种 群体, 其中所述群体主要包括包含所述重组表达载体的宿主细胞 (例 如, 主要由包含所述重组表达载体的宿主细胞组成) 。 所述群体还可 以是细胞的克隆群体, 其中所述群体的所有细胞是包含重组表达载体 的单一宿主细胞的克隆, 以致该群体的所有细胞包含所述重组表达载 体。 在本发明的一个实施方案中, 所述细胞群体是包括包含本文所述 的重组表达载体的宿主细胞的克隆群体。 缀合物
本发明还包括缀合物, 例如, 生物缀合物, 其包括本发明的多肽 (包括功能片段和功能变体) 以及肽模拟物、 多核苷酸、 重组表达载 体、 或宿主细胞中的任一种。 缀合物, 以及通常合成缀合物的方法在 本领域内也是已知的(参见,例如, Hudecz, F.,分子生物学方法(Methods Mol. Biol) 298: 209-223 (2005)和 Kirin等,无机化学(Inorg. Chem) 44(15): 5405-5415 (2005)) 。 药物组合物
本发明所提供的上述各种物质, 包括多肽 (包括功能片段和功能 变体) 以及肽模拟物、 脂肪酸衍生物、 多核苷酸、 重组表达载体、 以 及宿主细胞 (包括其群体) 、 缀合物等 (笼统称为 "本发明物质" ) 可以是分离的、 纯化的、 合成的、 和/或重组的。 本发明物质还可以配制成组合物, 诸如药物组合物。 在这一点上, 本发明提供包括任意所述多肽 (包括功能片段和功能变体) 以及肽模 拟物、 脂肪酸衍生物、 缀合物、 核酸、 重组表达载体和宿主细胞 (包 括其群体) , 以及药用载体的药物组合物。 包含任意本发明物质的本发 明药物组合物可以包括多于一种本发明物质, 例如 : 多肽和核酸, 或 两种或更多不同的多肽。 备选地, 所述药物组合物可以包括与另一种 或多种药物活性试剂或药物的组合。 所述另一种或多种药物活性试剂 或药物优选可以包括诸如化疗剂, 例如, 天冬酰胺酶, 白消安, 卡铂, 顺铂, 柔红霉素, 多柔比星, 氟尿嘧啶, 吉西他滨, 羟基脲, 甲氨蝶 呤, 紫杉醇, 利妥昔单抗, 长春碱, 长春新碱, 等等。 在本发明的一个实施方案中, 所述药物组合物包括与脂质组合的 本发明物质。 所述脂质可以是任何脂质, 包括脂肪酸、 磷脂、 固醇、 鞘脂、 萜、 甘油脂、 甘油磷酸酯、 异戊烯醇脂、 糖脂和聚酮化合物等。 所述脂在本领域内是已知的。 关于药物组合物, 药用载体可以是任意常规所用的那些, 它们可 以仅通过化学物理因素, 诸如溶解性和缺乏与活性化合物的反应性, 以及通过施用的途径, 进行限定。 本发明所述的药用载体, 例如, 媒 介物、 佐剂、 赋形剂和稀释剂等, 是本领域技术人员公知的, 并且容 易为公众所获得。 优选地, 所述药用载体对活性试剂是化学惰性的, 在使用条件下不具有有害副作用或毒性。 载体的选择应该由特定的本发明物质、 以及由用于施用本发明物 质的特定方法决定。 因此, 存在本发明药物组合物的多种适当制剂。 下述用于口服、 气雾剂、 肠胃外、 皮下、 静脉内、 肌内、 动脉内、 鞘 内、 腹膜内、 直肠、 和阴道施用的制剂是示例性的, 并且不以任何方 式限制。 可以使用多于一种途径施用本发明物质, 在特定情形中, 特 定的途径可以提供比另一种途径更直接和更有效的方法。 在本发明的一个优选实施方案中, 所述药物组合物是局部制剂、 静脉内制剂、 或皮下制剂。 在本发明的一个优选实施方案中, 所述药 物组合是局部制剂。 局部制剂是本领域技术人员公知的。 在本发明施 用到皮肤的情形中, 所述制剂特别适合。 本发明的局部制剂可以是, 例如, 膏剂、 洗液、 油膏、 贴片、 油、 糊剂、 喷雾剂, 例如气雾剂喷 雾剂、 凝胶、 滚抹式的液体、 固体棒等。 优选地, 本发明的局部制剂 是膏剂、 洗液、 油膏、 或贴片。 适用于口服施用的制剂可以由下列各项组成: a) 液体溶液, 诸如 溶解在稀释剂如水、 盐水、 或橙汁中的有效量的本发明物质; b) 胶囊、 片剂、 锭剂等, 每一种包含预先确定量的, 固体或颗粒状的活性成分; c) 粉剂; d) 在适当液体中的混悬液;和 e) 适当的乳剂。 液体制剂可 以包括稀释剂, 诸如水和醇, 例如, 乙醇、 苯甲醇、 和聚乙二醇, 可 以添加或不添加药用表面活性剂。 胶囊形式可以是普通的硬或软壳明 胶型, 其包含, 例如, 表面活性剂、 润滑剂、 和惰性填充剂诸如乳糖、 蔗糖、 磷酸钙、 和玉米淀粉。 片剂形式可以包括下列各项中的一种或 多种 :乳糖、 蔗糖、 甘露醇、 玉米淀粉、 马铃薯淀粉、 褐藻酸、 微晶 纤维素、 阿拉伯树胶、 明胶、 瓜耳胶、 胶状二氧化硅、 滑石、 硬脂酸 镁、 硬脂酸钙、 硬脂酸锌、 硬脂酸、 和其它赋形剂、 着色剂、 稀释剂、 缓冲剂、 崩解剂、 润湿剂、 防腐剂、 调味剂、 和其它药物相容的赋形 剂。 锭剂形式可以包括处在调味剂通常是在蔗糖或阿拉伯树胶中的本 发明物质, 以及包括处在惰性基质诸如明胶和甘油、 或蔗糖和阿拉伯 树胶中的本发明物质的软锭剂, 另外包含本领域己知的赋形剂的乳剂、 凝胶等。 本发明物质, 单独的或与其它适宜的成分组合, 可以制成通过吸 入来施用的气雾剂制剂。 这些气雾剂制剂可以放置在加压可用的介质 中, 诸如二氯二氟甲垸、 丙垸、 氮等。 它们还可以配制成非加压的制 剂, 如在喷雾器或雾化器中。 所述喷雾制剂还可以用于向粘膜喷雾。 适于肠胃外施用的制剂包括水性和非水性、 等渗无菌注射液, 其 可以包含抗氧化剂、 缓冲剂、 抑菌剂、 和使得所述制剂与预期接收者 的血液等渗的溶质, 以及水性和非水性无菌混悬液, 其可以包括混悬 剂、 增溶剂、 增稠剂、 稳定剂、 和防腐剂。 本发明物质可在药用载体 中的生理用稀释剂中使用, 诸如无菌液体或液体混合物, 包括水、 盐 水、 水性葡萄糖和相关的糖溶液、 醇如乙醇或十六醇、 二醇如丙二醇 或聚乙二醇、 二甲亚矾、 甘油、 缩酮如 2,2-二甲基 -1,3-二氧戊环 -4-甲 醇、 醚、 油、 脂肪酸、 脂肪酸酯或甘油酯, 或乙酰化的脂肪酸甘油酯, 添加或不添加药用表面活性剂诸如皂或去污剂, 混悬剂如果胶、 甲基 纤维素、 羟丙基甲基纤维素、 或氮甲基纤维素, 或乳化剂和其它药用 佐剂。 可以用在肠胃外制剂中的油包括石油、 动物油、 植物油、 或合成 油。 油的具体实例包括花生油、 大豆油、 芝麻油、 棉籽油、 玉米油、 橄榄油、 石油和矿物油。 用在肠胃外制剂中的适当的脂肪酸包括油酸、 硬脂酸、 和异硬脂酸。 用在肠胃外制剂中的适当的皂包括脂肪碱金属和三乙醇胺盐等, 并含有适当的去污剂, 例如 a) 阳离子去污剂:二甲基二垸基卤化铵和 垸基卤化吡啶; b) 阴离子去污剂:垸基、 芳基、 烯属磺酸酯、 垸基、 烯烃、 醚、 单甘油硫酸酯和磺基琥珀酸酯等; c)非离子去污剂:脂肪胺 氧化物、 脂肪酸链垸醇酰胺和聚氧乙烯聚丙烯共聚物等: d) 两性去污 剂:垸基 -β-氨基丙酸酯和 2-垸基-咪唑啉季铵盐; e) 它们的混合物。 所述肠胃外制剂可在溶液中含有大约为 0.5-25% (重量体积百分 浓度) 的本发明物质。 可以使用防腐剂和缓冲剂。 为了最小化或消除 在注射部位的刺激性, 所述组合物可以包含一种或多种亲水性-亲脂性 平衡 (HLB) 的非离子表面活性剂。 在所述制剂中表面活性剂的用量 约为 5-15% (重量体积百分浓度) 。 适当的表面活性剂包括聚乙二醇 失水山梨糖醇脂肪酸酯和环氧乙垸与疏水基的高分子量加合物, 所述 加合物是通过缩合环氧丙垸和丙二醇而形成的。 所述肠胃外制剂可以 存在于单位剂量或多剂量密封容器中, 并且可以保存在冷冻干燥 (冻 干) 的条件下, 仅需要在即将使用之前加入无菌液体赋形剂如水用于 注射。 可以由先前所述种类的无菌粉剂、 颗粒剂、 和片剂制备临时的 注射溶液和混悬液。 本发明物质或包括所述本发明物质的组合物还可以制成可注射制 剂。 可注射组合物对有效的药用载体的需要是本领域普通技术人员公 知的。 优选地, 当施用于细胞例如树突细胞时, 所述细胞通过注射施 用。 另外, 本发明的发明物质, 或包括所述发明物质的组合物, 可以 通过与各种基质如乳化基质或水溶性基质混合而制备成栓剂。 适于例 如阴道施用的制剂可以以阴道栓剂、 卫生棉条、 膏剂、 凝胶、 糊剂、 泡沫剂、 或喷雾制剂存在, 除了其活性成分之外, 还包含本领域中己 知合适的那些载体。 本领域技术人员应该了解, 除了上述药物组合物之外, 本发明的 发明物质还可以配制为包含复合物, 诸如环糊精包含复合物, 或脂质 体。 为了达到本发明的目的, 施用的本发明物质的量或剂量应该在合 理时间范围内在受试者或动物内产生例如治疗或预防响应。 例如, 本 发明物质的剂量应该足以在从施用时刻起约 2小时以上如 12-24小时或 更长的时间期间内, 抑制患病细胞的增殖, 起到治疗或预防疾病 (例 如, 肿瘤, 癌症等) 的作用。 在某些实施方案中, 所述时间期间甚至 可以更长。 剂量应该由特定的本发明物质的功效和待治疗的动物 (例 如, 人) 的状况, 以及动物 (例如, 人) 的体重所确定。 确定施用剂 量的许多测定方法是本领域内己知的。 本发明物质的剂量还将通过施 用特定的本发明物质可能伴随的任何副作用的存在、 性质和程度而确 定。 本领域普通技术人员应该容易理解, 本发明物质可以以任何方式 修饰, 以提高本发明物质的治疗或预防功效。 例如, 本发明物质可以 直接或通过连接体间接与靶向的部分缀合。 使化合物例如本发明物质, 与靶向部分缀合的实践在本领域内是公知的 (参见, Wadwa等, 药物 靶向杂志 (J. Drug Targeting), 3: 1 11, (1995)和美国专利号 5,087,616)。 在另一个实施方案中, 本发明物质可以被修饰成贮存库形式, 以便使 本发明物质释放到其所施用的体内的方式在时间和体内部位方面受到 控制 (参见, 例如, 美国专利号 4,450,150) 。 本发明物质的贮存库形 式可以是, 例如, 包括本发明物质和多孔或非多孔物质如聚合物的可 植入组合物, 其中本发明物质通过所述物质和/或所述非多孔物质的 降解而扩散。 然后, 将贮存库植入体内的理想部位, 并且本发明物质 以预先确定的速率从所述植入物释放。 本领域技术人员应该理解, 本发明提供的防止和抑制癌细胞生长 和增殖的方法, 包括使患病细胞与具有有效抑制量的本发明所述的任 意药物组合物接触。 可以采用任意常规的方法将本发明所述药物组合 物传递给癌细胞。 在本发明的一个优选的实施方案中, 将所述药物组 合物通过局部施用给宿主。 在另一个优选的实施方案中, 所述药物组 合物直接施用到癌细胞, 例如, 肿瘤内递送。 本发明的药物组合物, 包含多肽 (包括功能片段和功能变体) 以 及肽模拟物、 核酸、 重组表达载体、 和 /或宿主细胞, 可以用在防止和 抑制乙型肝炎病毒感染诱发的慢性肝病, 包括肝炎, 以及由此产生的 肝硬变和肝癌的方法中。 本领域普通技术人员应该容易理解, 本发明 所述的由乙型肝炎病毒诱发的慢性肝病可以存在于任何宿主中。 优选 地, 所述宿主是哺乳动物。 特别优选地, 所述宿主是人。 下面结合具体实施例, 对本发明作进一步的阐述说明。 应理解, 这些实施例仅用于说明本发明而不在于限制本发明的范围。 下述实施 例中的实验方法, 如无特殊说明, 均为常规方法;所用的材料, 如无 特殊说明, 均为自常规生化试剂公司购买得到的。 实施例一: 多肽的设计和制备
人工合成多肽功能片段 anti-HBxP2#:
本 发 明 用 人 工 合 成 的 方 法 合 成 了 氨 基 酸 序 列 为 Pro-Asp-Leu-His-Lys-Asn-Glu-Leu-Lys-His-Val-Lys-Tyr (SEQ ID NO: 1) 的多肽 (以下称 Anti-HBxP2#) 。 该多肽的制备采用固相合成方法, 如 应用 AAPPTEC公司的 Apex396型多肽合成仪器, 在密闭的防爆玻璃 反应器中按 SEQ ID NO: 1所示的序列从 C端-羧基端向 N端-氨基端合 成 氨 基 酸 , 这 是 指 第 一 个 被 加 入 到 该 氨 基 酸 序 列 Pro-Asp-Leu-His-Lys-Asn-Glu-Leu-Lys-His-Val-Lys-Tyr 的氨基酸单体 是 C末端的 Tyr, 然后再是 Lys, 再是 Val, —直到最后的 Asp以及 N 末端的 Pro, 不断添加、 反应、 合成, 操作最终得到所要具有的氨基酸 序列。 固相合成法, 大大的减轻了每步产品提纯的难度。 为了防止副 反应的发生, 参加反应的氨基酸的侧链都是受保护的。 羧基端是游离 的, 并且在反应之前必须活化。 具体合成由下列几个循环组成:
1) 去保护 : Fmoc 保护的柱子和单体必须用一种碱性溶剂 (piperidine) 去除氨基的保护基团。
2) 激活和交联:下一个氨基酸的羧基被一种活化剂所活化。 活 化的单体与游离的氨基反应交联, 形成肽键。 在此步骤使用大量的超 浓度试剂驱使反应完成。 循环:这两步反应反复循环直到合成完成。
3) 洗脱和脱保护 : 多肽从柱上洗脱下来, 其保护基团被一种脱 保护剂 (TFA) 洗脱和脱保护。 从 C端 (羧基端) 向 N端 (氨基端) 合成, 把 C端的第一个氨基酸 Tyr固定在树脂上, 脱去 Tyr氨基酸的 保护基团, 并活化下一个氨基酸 Lys 的羧基端, 使之发生缩和反应, 以此类推到合成完最后一个氨基酸后, 从树脂上切割下来, 粗肽经 HPLC纯化, 获得 98%纯度的 Anti-HBxP2#, 最后通过质谱做进一步的 鉴定, 其分子量为 1622.2Kd。 在固相合成中, 肽链的延长是在不溶性的聚苯乙烯树脂载体上进 行的。 合成多肽的 C-末端先和氯甲基聚苯乙烯树脂 (氯化苄酯树脂) 反应形成苄酯, 然后按肽链一级结构的顺序将氨基端已被保护的氨基 酸逐个加上去, 使肽链延长。 如图 1 所示, 人工合成多肽 1^-118^2#用高压液相色谱 (HPLCX应用 PLC Agela C18柱)分析,显示所获得的纯度为 98.997%。 实施例二:离体 (in vitro)多肽的抗 HBx活性
采用两种方法检测离体情况下实施例一中的多肽的抗 HBx 的活 性 : 第一种方法是采用分子克隆技术, 将表达实施例一中的多肽的 cDNA 克隆在真核表达载体 pcDNA3.1 (+) 上, 通过基因转染, 在肝 癌细胞内实现表达所研究多肽的目的, 进而观察所研究的多肽抑制 HBx 的效果;第二种方法是采用人工合成的多肽, 直接加入到培养肝 癌细胞的培养液中, 观察多肽抑制 HBx的效果。 实验中采用的肝癌细胞有两种 :一种为组成型表达 HBx 的肝癌 HepG2-X细胞 (稳定转染 HBx的肝癌 HepG2细胞) ;一种为组成型表 达乙肝病毒全基因的肝癌 HepG2.2.15细胞 (稳定转染 HBV全基因的 肝癌 HepG2细胞) 。 由于 HBx具有激活转录因子 NF-κΒ的作用, 通 过荧光素酶报告基因检测方法, 在分子水平检测 HBx是否受到抑制; 由于 HBx在肝癌细胞中具有促进肝癌细胞生长和增殖的作用, 可以通 过 3-(4,5-二甲基 -2-噻唑) -2,5-二苯基四氮唑溴盐 (MTT) 检测实施例一 中的多肽对上述 HepG2-X细胞和 HepG2.2.15细胞增殖的影响。
A.多肽真核表达载体的构建
1. 主要材料:
1) 菌株 :R coli DH5a (购自原平皓(天津)生物技术有限公司), 质粒: pcDNA3.1 (+) (购自 Invitrogen) , pEGFP-C2 (购自 Invitrogen) 2. 主要试剂
名称 来源 琼脂糖 Solarbio 质粒小提试剂盒 Transgen 氨苄青霉素 BBI 胰蛋白酶 BBI 胰蛋白胨 Promega 酵母提取物 Promega
EcoRI内切酶 Takara
Xhol内切酶 Takara rTaq酶 Takara
T4 DNA 连接酶 Takara 氯仿 BBI
3. 主要溶液配制 :
1) LB液体培养基
胰化蛋白胨 2.0g
酵母提取物 l .Og
NaCl 2.0g
加双蒸水溶解定容至 200ml
1.03 X 105 Pa 蒸汽灭菌 20min。
2) LB固体培养基
胰化蛋白胨 2.0g
酵母提取物 l .Og
NaCl 2.0g
琼脂粉 3.0g
双蒸水溶解定容至 200ml
1.03 X 105 Pa 蒸汽灭菌 20min。
3) 10mg/ml溴化乙锭
溴化乙锭 0.2g
双蒸水 20ml
工作液: 0.5ug/ml 4) TBE电泳缓冲液
5 X贮存液:
Tris 碱
fi朋酸
0.5 mol/L EDTA ( H 8.0)
加双蒸水定容至
4. 退火体系
试剂 加入量 (μΐ) 灭菌双蒸水 20
5 X Annealing Buffer 10
Forward DNA oligo (50 μΜ) 10
( 5 ' -AATTC ATGTGGCCTGATCTTC ACAAAAATGA
ACTAAAACATGTTAAATATTGAC-3 ' ) (SEQ ID
NO:13)
Reverse DNA oligo (50 μΜ) 10
( 5,-TCGAGTCAATATTTAACATGTTTTAGTTCATT
TTTGTGAAGATCAGGCCACATG-3 ') (SEQ ID NO :14)
5. 退火条件: 95°C, 2分钟;每 8秒下降 0. C至 25°C , 90分钟; 4°C , ∞。
6. pcDNA3.1 (+) 空质粒载体的小量提取
将本实验室所保存的含有 pcDNA3.1 (+) 质粒的 DH5a菌株进行 活化, 挑取单克隆, 加入 LB液体培养基中 (含氨苄霉素 100mg/L) 37 °C过夜培养。 使用 TransGen质粒小提试剂盒提取质粒。 双酶切反应体系:
试剂 加入量 (μΐ) 限制性内切酶 EcoR 1 2
限制性内切酶 Xhol 2
10 M buffer 4
DNA 小于 2 μδ
ddH2O 使总体积为 40 μΐ 混匀后, 37°C反应 3-6小时。
8. 连接反应体系
Figure imgf000030_0001
加入量 (μΐ)
退火产物 0.3 pmol
质粒双酶切回收产物 0.03 pmol
10 连接缓冲液 2.5 μΐ
连接酶 1 μΐ
ddH2O 使总体积为 25 μΐ
混匀后 16°C反应过夜。 合成碱基序列包括 cctgatcttc ga actaaaacat gttaaatat (SEQ ID NO: 7) , 连接示意图如图 8所示
9. 转化步骤
1) 将 DH5a感受态菌株 (购自 TianGen, 100 μΐ) 在冰上解冻;
2) 在超净台中将步骤 8中的 25 μΐ连接产物全部加入含有感受态 细菌的离心管中, 用枪头混匀, 冰上静置 30分钟;
3) 将离心管放入 42° (:水浴中准确热激 90秒;
4) 立即放入冰上静置 2分钟;
5) 向离心管中加入 500 μΐ无抗性 LB液体培养基, 37°C预培养
45分钟;
6) 取 100 μΐ菌液 (可根据形成克隆的密集程度做适度调整) 均 匀涂布于 LB固体培养板上 (含氨苄霉素 100 mg/L) , 待液体渗入培养 基后, 倒置 37° (:培养过夜;
7) 待平板上长出清晰可见的单克隆菌斑之后, 用接菌针小心挑 取独立的单克隆在 LB液体培养基中 37°C培养过夜。 10. 菌落 PCR反应体系
试剂 加入量
(μΐ) 灭菌双蒸水 35.75
10 X buffer 5 dNTP (I0mmol/L) 4
Forward Primer (20 μΜ) 1
5 '-TAATACGACTCACTATAGGG-3 ' (SEQ ID NO: 15)
Reverse Primer (20 μΜ) 1
5 '-TAGAAGGCACAGTCGAGG-3 ' (SEQ ID NO :16)
模板 DNA (菌液) 3
Taq 酶 0.25
11.菌落 PCR反应条件:
94°C 10分钟
94°C ' 30秒
53°C < 30秒 35
72°C . 15秒
Figure imgf000031_0001
72°C 10分钟
4°C 00
反应完毕后取反应产物在 1.5%琼脂糖凝胶中进行电泳分析。 将阳 性克隆送交生物公司进行测序。 并将该质粒命名为 p-Anti-HBxP2#。
B. 体外有效性实验
1. 细胞系:
表 1 实验中应用的细胞系
细胞系名 特性、 用途及说明 来源
HepG2 肝癌细胞系 上海劲马生物科 技有限公司
HepG2-X HepG2中稳定转染乙肝病毒 X基因 自行构建并保存
Wang Q, et al. Neoplasia. 2010;12 (2):
103 -15.
HepG2.2.15 HepG2中稳定转染 HBV全基因组 上海劲马生物科 技有限公司
L-O2 永生化的人正常肝细胞系 南京凯基生物科 技发展有限公司
L-O2-X L-O2中稳定转染乙肝病毒 X基因 自行构建并保存
Zhang WY, et al. Acta Pharmacol
Sinica. 2009; 30(8): 1 153-61.
2. 主要试剂
试剂名称 来源
RPMI1640培养液 Gibco
DMEM培养液 Gibco
Lipofectamine 2000 Invitrogen
青链霉素 Solarbio
胰蛋白酶 BBI
胎牛血清 Hyclone
3. 双荧光素酶报告基因分析
(1) 将对数生长期的细胞(为上述表 1中列举的细胞)以 0.75 x 105 个 /ml 的密度接种于 24孔细胞培养板, 每孔接种 500μ1细胞, 当细胞 汇合至 90%时进行基因转染。
(2) 启动子报告基因载体 (PGL3-NF-KB , 0.3 δ, 购自原平皓 (天 津) 生物技术有限公司) 分别以脂质体法转染至细胞同时以海肾荧光 素酶表达载体 (pRL-TK, O.l g, 购自 Promega公司)为内参。 同时转 染不同质量的由步骤 A得到的多肽质粒 (0.25 g, 0.5 g和 0.75 g) 或加入不同浓度的由实施例 1获得的合成多肽 (Ο.ΙμΜ, ΙμΜ, 10μ Μ和 ΙΟΟμΜ) , 每个浓度重复 3个孔。
(3) 在转染后 48小时 (或应用人工合成多肽 Anti-HBxP2#作用后
24小时) , 用 PBS将细胞洗 3次。
(4) 向每孔转染的细胞加入 100 μΐ I X Passive Lysis Buffer (PLB), 于室温下作用 15 min使细胞充分裂解, 用细胞刮子将细胞裂解 物刮下来, 转入 1.5 ml Eppendorf (EP)管中。
(5) 12,000 rpm 离心 30 min, 将上清吸入新的 EP管中。
(6) 向每个装有 100 μΐ 荧光素酶测定缓冲液 (Luciferase Assay Buffer II, LARIl) 的 EP管中加入等量细胞裂解液, 混匀。
(7) 立即将 EP管放入生物化学发光检测仪 (Turner Biosystems公 司产品) , 2 sec平衡后, 测定 10 sec后的光输出。
(8) 加入 100 μΐ荧光淬灭剂, 淬灭萤火虫荧光素酶同时启动海肾 荧光素酶反应。
(9) 测定 lOsec后的光输出。 第一次的荧光数值与第二次荧光数值的比值为相对活性。 每组实验均作 3次独立重复, 以 Mean土 SD作为统计依据, 进行 Student's t test统计学分析。 如图 2 所示, - 1^-1¾ ?2#对 HepG2-X细胞、 L-O2-X细胞和 HepG2.2.15细胞中 NF-κΒ启动子的活性具有抑制作用, 其抑制作用呈 剂量依赖性, 而对无 HBx表达的 HepG2肝癌细胞和 L-O2肝细胞无影 响。 利用该实验进一步表明, 含有八1^-118^2#的 5种功能变体的基因 对 HepG2-X细胞和 L-O2-X细胞 NF-κΒ启动子的活性具有抑制作用。 * P<0.05, ** P<0.01, 进行 Student's t test统计学分析。 如图 3所示, 应用人工合成的多肽 Anti-HBxP2#, 对 HepG2-X细 胞、 L-O2-X细胞和 HepG2.2.15细胞中 NF-κΒ启动子的活性具有抑制 作用,呈剂量依赖性。而对无 HBx的 HepG2肝癌细胞和无 HBx的 L-O2 肝细胞无影响。 * P<0.05, ** P<0.01, 进行 Student's t test统计学分析。
4. MTT检测
(1) 接种细胞:将对数生长期的细胞 (为表 1 中列举的细胞) 用 含 10%胎牛血清的 RPMI1640或 DMEM培养液配成单个细胞悬液, 以 每孔 4000-5000个细胞接种到 96孔细胞培养板中, 每孔体积 100 ul。
培养细胞:待 12h后细胞贴壁, 转染不同质量的在本实施例的步 骤 B中所获得的多肽质粒 (0.05 g, O.l g和 0.15 g) 或加入不同浓 度的在实施例 1中所获得的合成多肽 (Ο.ΙμΜ, ΙμΜ, ΙΟμΜ和 100 μΜ) , 每个浓度重复 8个孔, 同一般培养条件, 培养 48h。 将 0.3 g pEGFP-C2质粒与 l g p-Anti-HBxP2# 混合后共转染, 24小时后在倒 置荧光显微镜下观察绿色荧光, 确定其转染效率可达到 70%。
(2) 呈色:每孔加 MTT溶液 (5 mg/ml用 PBS (pH7.4) 配制) 20 μΐ.
(3) 继续孵育 4 小时, 终止培养, 小心吸弃孔内培养上清液。 每 孔加 150ulDMSO, 振荡 10分钟, 使结晶物充分融解。
(4) 比色:选择 490 nm波长, 在酶联免疫监测仪上测定各孔光吸 收值, 记录结果, 进行 Student's t test统计学分析。 如图 4 所示, - 1^-1¾ ?2#对 HepG2-X细胞、 L-O2-X细胞和 HepG2.2.15 细胞生长和增殖具有抑制作用, 呈其抑制作用呈剂量依赖 性。而对无 HBx的 HepG2肝癌细胞和无 HBx的 L-O2肝细胞则无影响。 相似地, p-Anti-HBxP2# 5种功能变体基因对 HepG2-X细胞和 L-O2-X 细胞的生长和增殖具有抑制作用。 * P<0.05, ** P<0.01, 进行 Student's t test统计学分析。 如图 5 所示, MTT 检测, 应用人工合成的多肽 1^-118^2#对 HepG2-X细胞、 L-O2-X细胞和 HepG2.2.15细胞生长和增殖具有抑制 作用,呈剂量依赖性。而对无 HBx的 HepG2肝癌细胞和无 HBx的 L-O2 肝细胞无影响。 * P<0.05, ** P<0.01, 进行 Student's t test统计学分析。 实施例三:动物体内 (^ W )多肽有效性实验
将对数生长期的 HepG2-X细胞或 HepG2.2.15细胞用胰酶消化制 成细胞悬液, 计算细胞数, 用无菌的生理盐水细胞稀释至 I X 107 个 细胞 /ml, 置冰水中存放。 再取 12只 4到 6周雌性 BALB/C裸鼠, 将小 鼠随机分为 2 组:①对照组, 在每只小鼠右前肢腋下皮下注射上述稀 释后的细胞 0.2 ml, 仅注射 0.5 ml灭菌蒸馏水 (不含多肽药物) ;② 实验组 (给药剂量为 10 mg/kg 体重) 。 在每只小鼠右前肢腋下皮下注 射上述稀释后的细胞 0.2 ml,注射 7天后,肿瘤体积(V=L X W2 X 0.5) 达到 100 mm3, 再分别皮下注射上述多肽药物 (用 0.5ml灭菌蒸馏水溶 解冻干的多肽药物) , 每两天注射一次, 共注射 10次, 每次注射前测 量肿瘤体积。 末次给药 24 小时后, 称量小鼠的体重, 然后将小鼠脱颈 椎处死, 剥取瘤体组织称重, 按下列公式计算抑瘤率。 对照组平均瘤重-实验组平均瘤重
抑瘤率 = χ 100%
对照组平均瘤重
如表 1和图 6以及表 2和图 7所示, 动物实验检测结果表明, 应 用人工合成的多肽八1^-1¾ ?2#对1^ 02- 和 HepG2.2.15细胞的生长 和增殖具有抑制作用。 ** P<0.01, 进行 Student's t test统计学分析。 表 1 :人工合成的多肽八1^-1¾^2#对 HepG2-X细胞的作用
Figure imgf000035_0001
表 2:人工合成的多肽八1^-1¾ ?2#对 HepG2.2.15细胞的作用 :
Figure imgf000036_0001
实施例四: 多肽的功能片段及变体
本发明还探讨了多肽及其功能片段的功能变体的作用。 下表所示 的序列为以八1^-118^2#为基础进行氨基酸添加 (例如, 在端侧添加一 个氨基酸) 或保守氨基酸置换 (例如, 用一个氨基酸置换另一个相同 类型的氨基酸) 而得到的。 按照所述序列进行人工合成多肽片段 (方 法如上) , 然后, 采用上述报告基因和 MTT的方法, 观察所得的多肽 的作用的变化:
合成多肽片断 氨 基 酸 序 列 SEQ ID
NO. anti-HBxP2# Pro-Asp-Leu-His-Lys-Asn-Glu-Leu-Lys-His-Va 1
1-Lys-Tyr
或 P-D-L-H-K-N-E-L-K-H-V-K-Y
anti-HBxP2#-l Ile-Pro-Asp-Leu-His-Lys-Asn-Glu-Leu-Lys-His- 2
Val-Lys-Tyr
或 I-P-D-L-H-K-N-E-L-K-H-V-K-Y
anti-HBxP2#-2 Val-Pro-Asp-Leu-His-Lys-Asn-Glu-Leu-Lys-Hi 3 s-Val-Lys-Tyr
或 V-P-D-L-H-K-N-E-L-K-H-V-K-Y anti-HBxP2#-3 Pro-Glu-Leu-His-Lys-Asn-Glu-Leu-Lys-His-Val 4
-Lys-Tyr
或 P-E-L-H-K-N-E-L-K-H-V-K-Y
anti-HBxP2#-4 Pro-Asp-Leu-His-Arg-Asn-Glu-Leu-Lys-His-Val 5
-Lys-Tyr
或 P-D-L-H-R-N-E-L-K-H-V-K-Y
anti-HBxP2#-5 Pro-Asp-Leu-His-Lys-Asn-Glu-Leu-Lys-His-Val 6
-Arg-Tyr
或 P-D-L-H-K-N-E-L-K-H-V-R-Y
合成多肽片断 核 苷 酸 序 列 SEQ ID
NO. anti-HBxP2# cctgatcttc acaaaaatga actaaaacat gttaaatat 7 anti-HBxP2#-l atccctgatc ttcacaaaaa tgaactaaaa catgttaaat at 8 anti-HBxP2#-2 gtgcctgatc ttcacaaaaa tgaactaaaa catgttaaat at 9 anti-HBxP2#-3 cctgaacttc acaaaaatga actaaaacat gttaaatat 10 anti-HBxP2#-4 cctgatcttc accgcaatga actaaaacat gttaaatat 11 anti-HBxP2#-5 cctgatcttc acaaaaatga actaaaacat gttcgctat 12
如图 2所示,含有八1^-118^2#的 5种功能变体的基因对 HepG2-X 细胞和 L-O2-X 细胞 NF-KB 启动子的活性具有一定的抑制作用。 * P<0.05, ** P<0.01 , 进行 Student's t test统计学分析。 如图 4所示,含有八1^-118^2#的 5种功能变体的基因对 HepG2-X 细胞和 L-O2-X细胞的生长和增殖具有抑制作用。 * PO.05, ** PO.01, 进行 Student's t test统计学分析。 实施例五: 多肽药物急性毒性试验
取昆明小白鼠, 每组 10只, 每组雌雄各 5只。分实验组和对照组。 实验组, 以 lg/kg的浓度从小鼠尾静脉单次注射多肽 Anti-HBxP2# (用 0.125 ml灭菌蒸馏水溶解冻干的 2.5 mg多肽) ;对照组注射 0.25 ml灭 菌蒸馏水。 注射后连续观察 24小时。 多肽药物急性毒性试验结果显示, 小鼠无异常表现。 与对照组相 比体重无异常变化。 本发明引用的所有参考文献, 包括出版物、 专利申请、 和专利, 通过引用结合于此, 并且完全结合于此。 本文提供的任何以及所有实 施例, 或示例性语言 (例如、 诸如等) 仅意欲更好地阐述本发明, 并 不形成对本发明范围的限制, 除非另外要求。 本发明描述了优选实施 方案, 包括本发明人已知实施本发明的最佳模式。 本发明包含这些优 选实施方案可能的变化, 本发明人意欲以这些与本文具体描述不同的 方式实施本发明, 同样, 本领域普通技术人员也清楚了解这些变化, 并预期能熟练地利用所述变化。 在法律所能允许的范围, 本发明包括 了在所附权利要求中引用的主题的所有修改和等价物, 除非本文另外 指明, 或明显与上下文相抵触。

Claims

权利要求书
1. 分离的多肽或肽模拟物,其包含如 SEQ ID NO: 1所示的氨基酸 序列、 或其功能片段或功能变体, 所述多肽或肽模拟物具有抑制乙型 肝炎病毒 X蛋白的功能, 并能抑制乙型肝炎病毒感染后的慢性肝病的 发生和发展。
2. 权利要求 1所述的多肽或肽模拟物, 所述乙肝病毒感染后的慢 性肝病包括肝炎、 肝硬变和肝癌。
3. 权利要求 1所述的多肽或肽模拟物, 其氨基酸序列具有与 SEQ ID NO: 1所示的氨基酸序列至少 70%的相同性。
4. 权利要求 1所述的多肽或肽模拟物, 其氨基酸序列具有与 SEQ ID NO: 1所示的氨基酸序列至少 80%的相同性。
5. 权利要求 1所述的多肽或肽模拟物, 其氨基酸序列具有与 SEQ ID NO: 1所示的氨基酸序列至少 90%的相同性。
6. 权利要求 1所述的多肽或肽模拟物, 其氨基酸序列包含如 SEQ
ID NOs: 1-6中所示的任一种氨基酸序列。
7. 分离的多核苷酸, 所述多核苷酸包含选自下组中的一种: a) 编码 SEQ ID NO: 1所示的氨基酸序列、 其功能片段或功能变 体的多核苷酸;或者
b) 与多核苷酸 a)互补或严格杂交的多核苷酸。
8. 权利要求 7的多核苷酸,所述多核苷酸包含选自下组中的一种: a) 编码 SEQ ID NOs: 1-6所示的任一种氨基酸序列的多核苷酸; 或者 b) 与多核苷酸 a)互补或严格杂交的多核苷酸。
9. 重组表达载体, 其包含权利要求 7或 8所述的多核苷酸。
10. 包括如权利要求 9所述的重组表达载体的宿主细胞。
11. 权利要求 1-6 中任一所述多肽在制造抗乙肝病毒感染后的慢 性肝病的药物中的应用。
12. 权利要求 7-8 中任一所述核苷酸在制造抗乙肝病毒感染后的 慢性肝病的药物中的应用。
13. 权利要求 9 中所述重组表达载体在制造抗乙肝病毒感染后的 慢性肝病的药物中的应用。
14. 权利要求 11-13中的任一种应用,所述应用为制造抗乙型肝炎 的治疗性疫苗。
15. 药物组合物,其包含权利要求 1-6中任一所述的多肽或肽模拟 物, 以及任选的药物载体。
16. 药物组合物, 其包含权利要求 7-8中任一所述的多核苷酸, 以 及任选的药物载体。
17. 药物组合物,其包含权利要求 9中所述的重组表达载体, 以及任选的药 物载体。
PCT/CN2012/072383 2011-03-15 2012-03-15 抗乙型肝炎病毒x蛋白多肽药物 WO2012122942A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201110061855.1A CN102675423B (zh) 2011-03-15 2011-03-15 抗乙型肝炎病毒x蛋白多肽药物
CN201110061855.1 2011-03-15

Publications (1)

Publication Number Publication Date
WO2012122942A1 true WO2012122942A1 (zh) 2012-09-20

Family

ID=46808026

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2012/072383 WO2012122942A1 (zh) 2011-03-15 2012-03-15 抗乙型肝炎病毒x蛋白多肽药物

Country Status (2)

Country Link
CN (1) CN102675423B (zh)
WO (1) WO2012122942A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013036201A1 (en) * 2011-09-06 2013-03-14 Agency For Science, Technology And Research Polypeptide vaccine

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104744564B (zh) * 2015-03-13 2017-07-28 天津托普泰克生物科技有限公司 抗乙型肝炎病毒x蛋白多肽药物

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1546668A (zh) * 2003-12-12 2004-11-17 南开大学 乙型肝炎病毒x蛋白转导系统表达载体及其构建
WO2007110098A1 (en) * 2006-03-29 2007-10-04 Institut National De La Sante Et De La Recherche Medicale HBx PEPTIDES, CAPABLE OF ELICITING A PROMISCUOUS IMMUNODOMINANT CD4+ RESPONSE DIRECTED AGAINST HBV

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1546668A (zh) * 2003-12-12 2004-11-17 南开大学 乙型肝炎病毒x蛋白转导系统表达载体及其构建
WO2007110098A1 (en) * 2006-03-29 2007-10-04 Institut National De La Sante Et De La Recherche Medicale HBx PEPTIDES, CAPABLE OF ELICITING A PROMISCUOUS IMMUNODOMINANT CD4+ RESPONSE DIRECTED AGAINST HBV

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CHENG, PENG ET AL.: "Expression and Significance of TGF(3 and Smad 4 Gene in Human Hepatocellular Carcinoma Tissues", CHINESE CLINICAL ONCOLOGY, vol. 14, no. 7, 15 July 2009 (2009-07-15), pages 609 - 613 *
DATABASE GENBANK [online] NCBI; 22 April 2003 (2003-04-22), retrieved from http;//www.ncbi.nlm.nih.gov/protein/28193874?report=genbank&logS=protop&blast rank =5&RID+SRK1HU8R012 Database accession no. AA027460.1 *
MURAKAMI, S.: "Hepatitis B virus X protein: a multifunctional viral regulator", J GASTROENTEROL, vol. 36, no. 10, October 2001 (2001-10-01), pages 651 - 660 *
SUN, YAN ET AL.: "Advances in Research on Smad4", LETTERS IN BIOTECHNOLOGY, vol. 16, no. 3, 30 June 2005 (2005-06-30), pages 299 - 302 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013036201A1 (en) * 2011-09-06 2013-03-14 Agency For Science, Technology And Research Polypeptide vaccine
US10258676B2 (en) 2011-09-06 2019-04-16 Agency For Science, Technology And Research Polypeptide vaccine

Also Published As

Publication number Publication date
CN102675423B (zh) 2014-02-05
CN102675423A (zh) 2012-09-19

Similar Documents

Publication Publication Date Title
AU2012205681B2 (en) Use of toll-like receptor agonist for treating cancer
AU2014306021B2 (en) Polyconjugates for delivery of RNAi triggers to tumor cells in vivo
WO2014046481A1 (ko) 세포 투과성 펩티드, 그를 포함한 컨쥬게이트 및 그를 포함한 조성물
BRPI0620806A2 (pt) peptìdeos úteis como peptìdeos de penetração de célula
CN106279423B (zh) Slit2D2-HSA融合蛋白及其在抗肿瘤中的应用
US11091533B2 (en) Protease-activated receptor-2 modulators
WO2016130966A1 (en) Compositions and methods for regulating blood pressure
WO2012122941A1 (zh) 抗乙型肝炎病毒x蛋白多肽药物
US20150202248A1 (en) E2f as a target of hormone refractory prostate cancer
CN107629114B (zh) 多肽、其衍生物及其在制备抗肺纤维化的药物中的应用
WO2012142930A1 (zh) 抗脂肪酸合成酶多肽及其应用
JP2019534300A (ja) プログラム細胞死リガンド1の腸管発現
WO2012122942A1 (zh) 抗乙型肝炎病毒x蛋白多肽药物
WO2012122943A1 (zh) 抗乙型肝炎病毒x蛋白多肽药物
JP4813720B2 (ja) ヒト循環ウイルス阻害ペプチド(virip)及びその使用
EP2004680B1 (en) N-terminal vdac variants and uses thereof
KR102017973B1 (ko) 항-b형 간염 바이러스 x 단백질 폴리펩티드 약제
KR101651330B1 (ko) 세포투과성이 우수한 tat-a20 융합단백질의 제조방법 및 이의 용도
CA3180108A1 (en) Engineered interleukin-22 polypeptides and uses thereof
CN103992388A (zh) 抗乙型肝炎病毒x蛋白多肽药物
AU2011281192A1 (en) Peptides based on the transmembrane domain of a Toll-like receptor (TLR) for treatment of TLR-mediated diseases
JP2021514183A (ja) 改変免疫調節ペプチド
CN112043819B (zh) Lapf及其相关物质在抗感染中的应用
JP6758022B2 (ja) 血管内皮細胞増殖因子受容体阻害ペプチド
CN111094357A (zh) 一类新的Bcl10聚合抑制剂及其应用

Legal Events

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

Ref document number: 12758178

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: 12758178

Country of ref document: EP

Kind code of ref document: A1