WO2012013110A1 - 具有抑制血管生成活性的多肽 - Google Patents

具有抑制血管生成活性的多肽 Download PDF

Info

Publication number
WO2012013110A1
WO2012013110A1 PCT/CN2011/076755 CN2011076755W WO2012013110A1 WO 2012013110 A1 WO2012013110 A1 WO 2012013110A1 CN 2011076755 W CN2011076755 W CN 2011076755W WO 2012013110 A1 WO2012013110 A1 WO 2012013110A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
amino acid
acid selected
polypeptide
ala
Prior art date
Application number
PCT/CN2011/076755
Other languages
English (en)
French (fr)
Inventor
许迅
徐艺
赵卉
Original Assignee
上海市第一人民医院
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 上海市第一人民医院 filed Critical 上海市第一人民医院
Publication of WO2012013110A1 publication Critical patent/WO2012013110A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/4753Hepatocyte growth factor; Scatter factor; Tumor cytotoxic factor II
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to the field of biomedicine, and more particularly to a novel polypeptide (H-KI series polypeptide) having an action of inhibiting neovascularization.
  • the polypeptide inhibits proliferation, migration, lumen formation and inhibition of porcine embryonic allantoic membrane and mouse retinal neovascularization in vitro.
  • the invention also relates to the preparation and use of said polypeptides and to pharmaceutical compositions containing said polypeptides. Background technique
  • Angiogenesis refers to the process of forming new blood vessels by proliferation and migration of vascular endothelial cells on the basis of the original capillary network.
  • Angiogenesis plays an important role in physiological processes such as embryonic development, injury repair, and is also a major cause of many neovascular diseases such as tumor growth and metastasis, proliferative diabetic retinopathy, retinopathy of prematurity, and rheumatoid arthritis. Pathological changes. Therefore, the research and application of neovascular inhibitors are important for some refractory neovascular related diseases.
  • Ocular neovascularization is closely related to a variety of eye diseases. It often causes severe visual impairment and eventually blindness, such as diabetic retinopathy (DR), age-related macular degeneration (AMD), corneal neovascularization, neovascularization. Glaucoma and so on.
  • Current clinical treatments include surgery, lasers, and drugs. Surgery requires certain indications, patients are suffering, and risks and complications are relatively high; laser treatment also has some pain, which can cause local visual field defects, induce new blood vessels, and the mid- and long-term effects are not ideal. Therefore, a method for treating ocular neovascularization without pain, convenience, effectiveness, and high patient compliance has become a hot topic in domestic and foreign research for more than ten years.
  • VEGF Vascular endothelial growth factor
  • the drugs that have been used in clinical practice or are still in experimental research are mainly anti-angiogenic agents against VEGF or its receptor (VEGFR).
  • VEGF antisense oligonucleotides including VEGF expression inhibition; VEGF receptor antisense oligonucleotides for inhibiting VEGF receptor expression; anti-VEGF antibody, soluble VEGF receptor, VEGF Trap, anti-VEGFR antibody for neutralizing VEGF in blood circulation , tyrosine kinase inhibitors, etc., these preparations are widely used in tumor treatment.
  • the drug For ocular surface administration, the drug must penetrate the lipophilic corneal epithelial cells in close contact with the hydrophilic corneal stroma, so that only the appropriate fat-soluble, low molecular weight or transporter with the ocular surface tissue (eg: Amino acid transporters, oligopeptide transporters, etc.) bind to the drug to reach the anterior chamber.
  • the appropriate fat-soluble, low molecular weight or transporter with the ocular surface tissue eg: Amino acid transporters, oligopeptide transporters, etc.
  • the bioavailability of ophthalmic drugs is very low; to increase it, the concentration of the drug can be increased.
  • Compounds used to treat neovascularization of tumors are more toxic and side effects, and are not administered at high doses both systemically and locally.
  • angiostatin can significantly inhibit the growth of vascular-dependent tumors, but due to its large molecular weight and spatial conformation Complex, so there are deficiencies in the process of preparation and purification of the recombinant expression and endotoxin residues.
  • peptide angiogenesis inhibitors Compared with the widely studied protein angiogenesis inhibitors, peptide angiogenesis inhibitors have simple synthesis methods, easy chemical modification, low immunogenicity, good solubility, high bioavailability, and strong tissue penetration. The advantages of various medicines and low prices are outstanding advantages. However, there are currently no small molecule polypeptides with satisfactory effects from hepatocyte growth factor (HGF).
  • HGF hepatocyte growth factor
  • Another object of the invention is to provide a process and use comprising the polypeptide.
  • XaaO is no, or 1-3 amino acids constitute a peptide
  • Xaal is an amino acid selected from the group consisting of He , Leu, Val, Met or Ala;
  • Xaa2 is an amino acid selected from the group consisting of He , Leu, Val, Met or Ala;
  • Xaa3 is an amino acid selected from the group consisting of Gly or Ala;
  • Xaa4 is an amino acid Lys or Arg selected from the group below;
  • Xaa5 is an amino acid selected from the group consisting of Gly or Ala;
  • Xaa6 is an amino acid selected from the group consisting of Arg, Lys or Gly;
  • Xaa7 is an amino acid selected from the group below Ser or Thr
  • Xaa8 is an amino acid selected from the group consisting of Tyr or Phe
  • Xaa9 is an amino acid selected from the group consisting of Lys or Arg
  • XaalO is an amino acid selected from the group consisting of Gly or Ala;
  • Xaal l is an amino acid Thr or Ser selected from the group below ;
  • Xaal2 is an amino acid selected from the group consisting of Val, Leu, He, Met or Ala;
  • Xaal3 is an amino acid Ser or Thr selected from the group below ;
  • Xaal4 is an amino acid selected from the group consisting of He , Leu , Val, Met or Ala;
  • Xaal5 is an amino acid Thr or Ser selected from the group below ;
  • Xaal6 is an amino acid Lys or Arg selected from the group consisting of
  • Xaal7 Ser or Thr is an amino acid selected from the group;
  • Xaal8 is an amino acid selected from the group consisting of Gly or Ala;
  • Xaal9 is an amino acid selected from the group consisting of He , Leu , Val, Met or Ala;
  • Xaa20 is an amino acid Lys or Arg selected from the group consisting of;
  • Xaa21 is no, or 1-3 amino acids constitute a peptide
  • polypeptide has an activity of inhibiting angiogenesis, and the polypeptide has a length of 20-26 In another preferred embodiment, the polypeptide is 20-23 amino acids in length.
  • XaaO and/or Xaa21 are peptides consisting of 1-3 amino acids. More preferably, XaaO is C, NC, or RNC; and/or ⁇ 3&21 is (, CQ or CQP.
  • Xaal is an amino acid selected from the group consisting of: lie or Leu;
  • Xaa2 is an amino acid selected from the group consisting of: lie or Leu;
  • Xaa3 is an amino acid selected from the group consisting of Gly or Ala;
  • Xaa4 is an amino acid selected from the group consisting of Lys or Arg;
  • Xaa5 is an amino acid selected from the group consisting of Gly or Ala;
  • Xaa6 is an amino acid selected from the group consisting of Arg or Lys;
  • Xaa7 is an amino acid selected from the group consisting of Ser or Thr;
  • Xaa8 is an amino acid selected from the group consisting of: Tyr or Phe;
  • Xaa9 is an amino acid selected from the group consisting of Lys or Arg;
  • XaalO is an amino acid selected from the group consisting of Gly or Ala;
  • Xaall is an amino acid selected from the group consisting of Thr or Ser;
  • Xaal2 is an amino acid selected from the group consisting of Val or Leu;
  • Xaal3 is an amino acid selected from the group consisting of Ser or Thr;
  • Xaal4 is an amino acid selected from the group consisting of: lie or Leu;
  • Xaal5 is an amino acid selected from the group consisting of Thr or Ser;
  • Xaal6 is an amino acid selected from the group consisting of Lys or Arg;
  • Xaal7 is an amino acid selected from the group consisting of Ser or Thr;
  • Xaal8 is an amino acid selected from the group consisting of Gly or Ala;
  • Xaal9 is an amino acid selected from the group consisting of: lie or Leu; and/or
  • Xaa20 is an amino acid selected from the group consisting of Lys or Arg.
  • Xaa6 is Gly.
  • polypeptide is selected from the group consisting of:
  • amino acid sequence represented by SEQ ID NO: 1 is formed by substitution, deletion or addition of 1-5 (preferably 1-3, more preferably 1-2) amino acid residues, and has inhibition A polypeptide derived from (a) angiogenic function.
  • the derivative polypeptide retains 70% of the angiogenic activity of the indicated polypeptide of SEQ I. In another preferred embodiment, the derivative polypeptide is 80% identical to SEQ ID NO: 1, preferably 90%; more preferably 95%.
  • the invention also provides dimeric and multimeric forms of the compounds of formula I which inhibit angiogenic function.
  • an isolated nucleic acid molecule encoding the above-described polypeptide of the invention.
  • composition comprising:
  • the composition is in the form of eye drops, injections (e.g., periocular and intraocular injections), ophthalmic gels or ophthalmic ointments.
  • the composition is a sustained release dosage form.
  • a polypeptide or a pharmaceutically acceptable salt of the invention for the preparation of a medicament for inhibiting angiogenesis or preventing diseases associated with angiogenesis.
  • the angiogenesis-related disease is selected from the group consisting of neovascular ophthalmopathy, tumor, ischemic heart disease, non-inflammatory cardiomyopathy, coronary arteriosclerosis, arteriosclerosis obliterans, arteries. Embolism, arterial thrombosis, Berger's disease, chronic inflammation, inflammatory bowel disease, ulcers, rheumatoid arthritis, scleroderma, psoriasis, infertility or sarcoma.
  • the neovascular eye disease comprises involvement of the choroid, retina, cornea or iris, including age-related macular degeneration, proliferative diabetic retinopathy, retinal vascular occlusive disease, retinopathy of prematurity, corneal infection , neovascular glaucoma and so on.
  • a method of inhibiting angiogenesis in a mammal comprising the steps of: administering to a subject in need thereof a polypeptide of the invention or a pharmaceutically acceptable salt thereof.
  • the object is a human.
  • the angiogenesis is angiogenesis associated with neovascular eye disease.
  • Figure 1 shows the mean values of the 0D values for each group in the MTS cell proliferation assay.
  • Figure 2 shows the number of cells in the VEGF group and VEGF+ peptides in the Transwel l cell migration experiment through the porous membrane (bar graph) and the VEGF group and the cells in different concentrations of H-KI20 migrated through the porous membrane (color photo).
  • the number of cells in the polypeptide group is significantly lower than that in the VEGF group, and it tends to decrease with increasing concentration.
  • Asterisks indicate a statistically significant difference between this group and the VEGF group.
  • the transparent small round holes in the photograph are the pores of the porous membrane, and the blue-violet cells are the cells which are stained by the hematoxylin and migrate through the pores.
  • Figure 3 shows the average total length (bar graph) of the lumen formed by the blank control group, the VEGF group, and the VEGF+ different concentrations of the polypeptide group in the in vitro lumen formation experiment of endothelial cells, as well as the blank control group, the VEGF group, and the VEGF group. + ⁇ Formation of endothelial cells in the H-KI20 group (photograph).
  • Figure 4 shows the growth of blood vessels on the chorioallantoic membrane (CAM) of the PBS group and the polypeptide group (color photograph) and the CAM blood vessel counts of each group in the evaluation experiment (bar graph).
  • CAM chorioallantoic membrane
  • the white disc in the photo is the filter paper
  • the black coil is defined by the blood vessel counting range
  • the orange blood vessel membrane is the chicken embryo allantoic membrane.
  • the filter paper with 10 ⁇ ⁇ / ⁇ 1 ⁇ - ⁇ 20 is added.
  • the number of blood vessels was significantly less than that of the PBS group.
  • the control group was in the PBS group. It can be seen that the number of blood vessels in the 5 ⁇ ⁇ / ⁇ 1 ⁇ - ⁇ 20 group and the 10 ⁇ ⁇ / ⁇ 1 ⁇ - ⁇ 20 group decreased significantly compared with the PBS group, and decreased with increasing concentration.
  • the asterisk indicates that the difference between this group and the control group is statistically significant.
  • 762, O. OO D o Figure 5 is a retinal patch of the retinal neovascular model, a photograph of the eyeball slice, and a statistical result of the number of neovascular lumens in the eyeball section. (bar chart).
  • Neovascular group (indicated by the elliptical circle), the number of neovascular clusters in the retinal plaque of young rats in the retinal neovascular model group after intravitreal injection of H-KI20 was significantly reduced. It can be seen from the eyeball slice that the neovascular lumen is not seen in the normal group before the retina, and the retinal neovascular model can see more neovascular lumens in front of the retina (shown by black arrows).
  • HGF hepatocyte growth factor
  • the present inventors After extensive and intensive research, the present inventors have for the first time prepared a small molecule polypeptide derived from hepatocyte growth factor (HGF) having an angiogenic function and having a molecular weight of less than 5 kD (e.g., only about 2 kD).
  • HGF hepatocyte growth factor
  • the inventors applied bioinformatics methods, based on homology analysis and biological characteristics analysis, designed several candidate sequences, synthesized by solid phase method, and then passed through chicken embryo chorioallantoic membrane Model, VEGF-induced cell proliferation model and hypoxia-induced retinal neovascularization in mice, a new class of small molecule peptides with prophylactic and therapeutic angiogenic functions were obtained.
  • the small peptide of the invention has small molecular weight and can penetrate various eye tissue barriers; has good water solubility, can maintain high concentration in neutral tears, aqueous humor and vitreous humor; has high safety and has low toxicity to biological tissues.
  • the topical application of the eye is highly bioavailable, can penetrate the blood-eye barrier, and can reduce the dose, thereby reducing systemic side effects.
  • the present invention has been completed on this basis. Active polypeptide
  • polypeptide of the present invention refers to having blood vessels.
  • H-KI20 polypeptide refers to having blood vessels.
  • SEQ ID NO: 1 amino acid sequence of the peptide H-KI20 which is a novel inhibitory activity.
  • the term also encompasses variant forms of the sequence of SEQ ID NO: 1 having an angiogenic inhibitory function.
  • variants include (but are not limited to): 1-5 (usually 1-4, preferably 1-3, more preferably 1-2, optimally 1) amino acid deletions, insertions And/or substitution, and addition or deletion of one or several (usually within 5, preferably within 3, more preferably within 2) amino acids at the C-terminus and/or N-terminus.
  • 1-5 usually 1-4, preferably 1-3, more preferably 1-2, optimally 1 amino acid deletions, insertions And/or substitution, and addition or deletion of one or several (usually within 5, preferably within 3, more preferably within 2) amino acids at the C-terminus and/or N-terminus.
  • amino acids usually within 5, preferably within 3, more preferably within 2 amino acids at the C-terminus and/or N-terminus.
  • the term also encompasses both monomeric and multimeric forms of the polypeptides of the invention.
  • the term also includes both linear as well as non-linear polypeptides (e.g., cyclic peptides).
  • the invention also encompasses active fragments, derivatives and analogs of the H-KI20 polypeptide.
  • fragment refers to a polypeptide that substantially retains an angiogenic function or activity.
  • a polypeptide fragment, derivative or analog of the invention may be (i) one or more conserved or a polypeptide in which a non-conservative amino acid residue (preferably a conservative amino acid residue) is substituted, or (ii) a polypeptide having a substituent group in one or more amino acid residues, or (iii) an H-KI20 polypeptide and another a polypeptide formed by fusing a compound (such as a compound that prolongs the half-life of the polypeptide, such as polyethylene glycol), or (iv) a polypeptide formed by the additional amino acid sequence fused to the polypeptide sequence (with a leader sequence, a secretory sequence, or a tag sequence such as 6His) The resulting protein after fusion).
  • a compound such as a compound that prolongs the half-life of the polypeptide, such as polyethylene glycol
  • a polypeptide formed by the additional amino acid sequence fused to the polypeptide sequence with a leader sequence, a secretory sequence
  • a preferred class of reactive derivatives means that up to 5, preferably up to 3, more preferably up to 2, and optimally 1 amino acid are similar or similar amino acids to the amino acid sequence of Formula I. Substituting to form a polypeptide. These conservative variant polypeptides are preferably produced by amino acid substitution according to Table I.
  • the invention also provides analogs of the H-KI20 polypeptide.
  • the difference between these analogs and the native H-KI20 polypeptide may be a difference in amino acid sequence, or may be a difference in the modification form that does not affect the sequence, or Both.
  • Analogs also include analogs having residues other than the native L-amino acid (e.g., D-amino acids), as well as analogs having non-naturally occurring or synthetic amino acids (e.g., beta, ⁇ -amino acids). It is to be understood that the polypeptide of the present invention is not limited to the representative polypeptides exemplified above.
  • Modifications include: chemically derivatized forms of the polypeptide, such as acetylation or carboxylation, in vivo or in vitro. Modifications also include glycosylation, such as those produced by glycosylation modifications in the synthesis and processing of the polypeptide or in further processing steps. Such modification can be accomplished by exposing the polypeptide to an enzyme that performs glycosylation, such as a mammalian glycosylation enzyme or a deglycosylation enzyme. Modified forms also include sequences having phosphorylated amino acid residues such as phosphotyrosine, phosphoserine, phosphothreonine. Also included are polypeptides modified to increase their resistance to proteolytic properties or to optimize solubility properties.
  • the polypeptide of the present invention can also be used in the form of a salt derived from a pharmaceutically or physiologically acceptable acid or base.
  • These salts include, but are not limited to, salts formed with: hydrochloric acid, hydrobromic acid, sulfuric acid, citric acid, tartaric acid, phosphoric acid, lactic acid, pyruvic acid, acetic acid, succinic acid, oxalic acid, fumaric acid, malay Acid, oxaloacetic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, or isethionic acid.
  • Other salts include: salts with alkali or alkaline earth metals such as sodium, potassium, calcium or magnesium, as well as esters, carbamates or other conventional "prodrugs". Coding sequence
  • the invention also relates to polynucleotides encoding a ⁇ - ⁇ 20 polypeptide.
  • a preferred coding sequence is atcattggta aaggacgcag ctacaaggga acagtatcta tcactaagag tggcatcaaa (SEQ ID NO: 3), which encodes the amino acid sequence set forth in SEQ ID NO: 1.
  • the polynucleotide of the present invention may be in the form of sputum or RNA. ⁇ can be a coded chain or a non-coded chain.
  • the coding region sequence encoding the mature polypeptide may be identical to the coding region sequence shown in SEQ ID NO: 3 or may be a degenerate variant.
  • a "degenerate variant" in the present invention refers to a polypeptide encoding a sequence having the sequence of SEQ ID NO: 1, but with the corresponding coding region sequence of SEQ ID NO: Differential nucleic acid sequences.
  • the full-length H-KI20 nucleotide sequence of the present invention or a fragment thereof can be usually obtained by a PCR amplification method, a recombinant method or a synthetic method.
  • a DNA sequence encoding a polypeptide of the present invention (or a fragment thereof, or a derivative thereof) completely by chemical synthesis.
  • the DNA sequence can then be introduced into various existing DNA molecules (e.g., vectors) and cells known in the art.
  • the invention also relates to a vector comprising a polynucleotide of the invention, and to the use of the vector of the invention or
  • H-KI20 polypeptide coding sequence is a genetically engineered host cell.
  • the invention also encompasses polyclonal and monoclonal antibodies, particularly monoclonal antibodies, that are specific for the polypeptide encoded by H-KI20 DNA or a fragment thereof.
  • the polypeptide of the invention may be a recombinant polypeptide or a synthetic polypeptide.
  • the polypeptides of the invention may be chemically synthesized, or recombinant. Accordingly, the polypeptide of the present invention can be artificially synthesized by a conventional method or can be produced by a recombinant method.
  • a preferred method is to use liquid phase synthesis techniques or solid phase synthesis techniques such as Boc solid phase method, Fmoc solid phase method or a combination of both methods.
  • the solid phase synthesis can quickly obtain samples, and the appropriate resin carrier and synthesis system can be selected according to the sequence characteristics of the peptide of interest.
  • a preferred solid phase support in the Fmoc system is a Wang resin linked to a C-terminal amino acid in the peptide, a Wang resin structure is polystyrene, and an arm between the amino acids is 4-decyloxybenzyl alcohol; using 25% hexahydropyridine /dimethylformamide was treated at room temperature for 20 minutes to remove the Fmoc protecting group and extended from the C-terminus to the N-terminus according to the given amino acid sequence. After the completion of the synthesis, the synthesized proinsulin-related peptide was cleaved from the resin with trifluoroacetic acid containing 4% p-methylphenol, and the protecting group was removed.
  • the resin was removed by filtration and the diethyl ether was precipitated to obtain a crude peptide. After the solution of the obtained product was lyophilized, the desired peptide was purified by gel filtration and reverse phase high pressure liquid chromatography.
  • the resin is a PAM resin to which a C-terminal amino acid in the peptide is attached, the PAM resin structure is polystyrene, and the arm between the amino acid is 4-hydroxymethyl phenylacetamide;
  • the protecting group Boc was removed with TFA/dichloromethane (DCM) and neutralized with diisopropylethylamine (DIEA/dichloromethane).
  • the p-cresol (5-10%) containing hydrogen fluoride (HF) was treated at 0 ° C for 1 hour to cut the peptide chain from the resin while removing the protecting group. 50-80% The peptide is extracted with acetic acid (containing a small amount of mercaptoethanol), and the solution is further lyophilized and further purified by molecular sieve Sprint haddex G10 or Tsk-40f, and then purified by high pressure liquid phase to obtain the desired peptide. It can be known in the field of peptide chemistry.
  • Various coupling agents and coupling methods are coupled to each amino acid residue, and for example, dicyclohexylcarbodiimide (DCC), hydroxybenzotriazole (HOBt) or 1, 1, 3, 3-tetra can be used.
  • DCC dicyclohexylcarbodiimide
  • HOBt hydroxybenzotriazole
  • 1, 1, 3, 3-tetra 1, 1, 3, 3-tetra
  • DCC dicyclohexylcarbodiimide
  • HOBt hydroxybenzotriazole
  • HBTU urea hexafluorophosphate
  • the polypeptide H-KI20 of the present invention is prepared by solid phase synthesis according to its sequence, and purified by high performance liquid chromatography to obtain a high-purity peptide freeze-dried powder, and -2 CTC is stored.
  • Another method is to produce a polypeptide of the invention using recombinant techniques.
  • Polynucleotides of the invention can be utilized to express or produce recombinant H-KI20 polypeptides by conventional recombinant DNA techniques. Generally there are the following steps: (1) using a polynucleotide (or variant) encoding an H-KI20 polypeptide of the present invention, or transforming or transducing a suitable host cell with a recombinant expression vector containing the polynucleotide;
  • the recombinant polypeptide can be expressed intracellularly, or on the cell membrane, or secreted extracellularly. If desired, the recombinant protein can be isolated and purified by various separation methods using its physical, chemical, and other properties. These methods are well known to those skilled in the art. Examples of such methods include, but are not limited to: conventional renaturation treatment, treatment with a protein precipitant (salting method), centrifugation, osmotic sterilizing, ultra-treatment, ultra-centrifugation, molecular sieve chromatography (gel filtration), adsorption layer Analysis, ion exchange chromatography, high performance liquid chromatography (HPLC) and various other liquid chromatography techniques and combinations of these methods.
  • conventional renaturation treatment treatment with a protein precipitant (salting method), centrifugation, osmotic sterilizing, ultra-treatment, ultra-centrifugation, molecular sieve chromatography (gel filtration), adsorption layer Analysis, ion exchange chromatography
  • polypeptide of the present invention is short, it is conceivable to connect a plurality of polypeptides in series, to obtain an expression product in a multimeric form after recombinant expression, and then to form a desired small peptide by enzymatic cleavage or the like.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising (a) a safe and effective amount of a polypeptide of the present invention or a pharmaceutically acceptable salt thereof; and (b) a pharmaceutically acceptable carrier or excipient .
  • the amount of the polypeptide of the present invention is usually from 10 ⁇ g to 100 mg / dose, preferably from 100 to 1000 ⁇ g / dose.
  • an effective dose is from about 0.01 mg/kg to 50 mg/kg, preferably from 0.05 mg/kg to 10 mg/kg body weight of the polypeptide of the invention.
  • the polypeptides of the invention may be used alone or in combination with other therapeutic agents (e.g., formulated in the same pharmaceutical composition).
  • the pharmaceutical composition may also contain a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier refers to a carrier for the administration of a therapeutic agent.
  • pharmaceutical carriers which do not themselves induce the production of antibodies harmful to the individual receiving the composition and which are not excessively toxic after administration. These vectors are well known to those of ordinary skill in the art. A full discussion of pharmaceutically acceptable excipients can be found in Remington's Pharmaceutical Scences (Mack Pub. Co., N. J. 1991).
  • Such carriers include, but are not limited to, saline, buffer, dextrose, water, glycerol, ethanol, adjuvants, and combinations thereof.
  • the pharmaceutically acceptable carrier in the therapeutic composition may contain a liquid such as water, saline, glycerol and ethanol.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering substances and the like may also be present in these carriers.
  • the therapeutic compositions may be formulated as injectables, such as liquid solutions or suspensions; solid forms such as liquid carriers, which may be suitable in the preparation of solutions or suspensions before injection.
  • composition of the invention can be administered by conventional routes including, but not limited to, ocular, periocular, intraocular, intramuscular, intravenous, subcutaneous, intradermal or topical administration.
  • the subject to be prevented or treated may be an animal; especially a human.
  • compositions such as eye drops, injections, ophthalmic gels and ophthalmics which can be exemplified can be formulated by mixing, diluting or dissolving according to a conventional method, and occasionally adding a suitable pharmaceutical additive such as a shape Agent, disintegrant, binder, lubricant, diluent, buffer, isotonic agent (i sotonic iti es preservative, wetting agent, emulsifier, dispersant, stabilizer and cosolvent, and the formulation process) It can be carried out in a usual manner depending on the dosage form.
  • a shape Agent i sotonic iti es preservative, wetting agent, emulsifier, dispersant, stabilizer and cosolvent
  • the preparation of eye drops can be carried out by dissolving the short peptide H-KI20 or a pharmaceutically acceptable salt thereof together with the base substance in sterile water (a surfactant is dissolved in sterile water) to adjust the osmotic pressure and
  • a surfactant is dissolved in sterile water
  • the pH is in a physiological state, and a suitable pharmaceutical additive such as a preservative, a stabilizer, a buffer, an isotonic agent, an antioxidant, and a tackifier may be optionally added, and then completely dissolved.
  • compositions of the invention may also be administered in the form of sustained release agents.
  • the short peptide H-KI20 or a salt thereof can be intruded into a pellet or microcapsule in which a sustained-release polymer is used as a carrier, and then the pellet or microcapsule is surgically implanted into a tissue to be treated.
  • the short peptide H-KI20 or a salt thereof can also be applied by inserting a drug-coated intraocular lens.
  • sustained-release polymer examples include ethylene-vinyl acetate copolymer, polyhydroxymethacrylate (polyhydrometaacrylate polyacrylamide, polyvinylpyrrolidone, methylcellulose, lactic acid polymer, A lactic acid-glycolic acid copolymer or the like is preferably exemplified by a biodegradable polymer such as a lactic acid polymer and a lactic acid-glycolic acid copolymer.
  • the dose of the short peptide H-KI20 as an active ingredient or a pharmaceutically acceptable salt thereof may be based on the weight, age, sex, symptom of each patient to be treated. Determined to a reasonable extent.
  • the concentration is usually about 0.1 to 10% by weight, preferably 1 to 5% by weight, and may be administered 2 to 6 times a day, 1-2 drops each time.
  • a pharmaceutical composition containing the polypeptide of the present invention or a pharmaceutically acceptable salt thereof as an active ingredient has a remarkable inhibitory activity against angiogenesis. It has been confirmed by animal experiments that the polypeptide of the present invention can inhibit not only the chicken embryo allantoic sac
  • the angiogenesis of the membrane can inhibit the proliferation, migration, chemotaxis and lumen formation of human umbilical vein endothelial cells, and can inhibit hypoxia-induced retinal neovascularization in mice.
  • the main advantages of the invention include:
  • the polypeptide of the present invention has a small molecular weight and is permeable to the ocular tissue barrier;
  • (d) can be prepared by solid phase synthesis, with high purity, high yield and low cost;
  • polypeptide of the present invention has good stability.
  • the polypeptide of the present invention is expected to be developed into a medicament for the treatment of neovascular eye diseases and related neovascular diseases such as tumor neovascularization.
  • the invention is further illustrated below in conjunction with specific embodiments. It is to be understood that the examples are not intended to limit the scope of the invention.
  • the experimental methods in the following examples which do not specify the specific conditions are usually carried out according to the conditions described in conventional conditions such as Sambrook et al., Molecular Cloning: Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989), or according to the manufacturer. The suggested conditions.
  • Example 1 Example 1
  • H-KI20 I IGKGRSYKGTVSITKSGIK (SEQ ID NO: 1; position 129-148 in SEQ ID NO: 2)
  • H-KA SEQ ID NO: 2 in positions 150-176;
  • H-KC 190-200 in SEQ ID NO: 2;
  • the procedure is as follows: The required Fmoc protected amino acid solution, condensation reagent and cleavage reagent were calculated and prepared according to the software (Vers i on. version 201) using a SYMI3 ⁇ 40NY 12-channel polypeptide synthesizer (Protein Technologis, USA). Editing procedure, wherein the resin swelling time is 30 min; Deprotection twice, the time was 5 min and 15 min respectively; the condensation time was 30 min; the cutting time was 2 h.
  • the polypeptide was synthesized according to the above procedure, and the polypeptide was purified by high performance liquid chromatography (SHIMADZU) to obtain a white powdery polypeptide having a purity of >95% (120 mg each of each polypeptide), which was lyophilized for use.
  • SHIMADZU high performance liquid chromatography
  • H-KI20 consists of 20 amino acid residues with a molecular weight of 2093.52D, which does not contain cysteine residues and disulfide bonds.
  • Example 2 H-KI20 polypeptide inhibits VEGF-induced proliferation of vascular endothelial cells
  • HUVECs purchased from Sci enCell
  • ECGS ScienCell
  • fetal bovine serum ScienCell
  • the third to eighth generation HUVECs cells were used in all in vitro cell experiments in the present invention.
  • the MTS cell proliferation quantitative detection method is a method for producing a living cell proliferation by colorimetric determination of a water-soluble colored product by using a tetrazolium and an electron-conjugated compound under the action of a metabolically proliferating cell mitochondrial dehydrogenase.
  • HUVECs are grown close to the fusion, passaged, and seeded in a 96-well plate at a density of 3.5 ⁇ 107 ml, cultured in a well of 100 ⁇ l, 37° C., 5% CO 2 incubator for 24 hours, and then replaced with serum-free ECM. Medium, cells starved overnight.
  • the 96-well plate medium was aspirated, and each group was added with 50 ⁇ l of serum-free medium containing 1 ⁇ , 10 nM, 100 ⁇ , ⁇ , ⁇ ⁇ - ⁇ polypeptides, pretreated at 37 °C for 30 min, and then added to each well.
  • Serum-free medium containing VEGF (R&D) was used to give a final concentration of VEGF of 10 ng/ml.
  • a blank control group no VEGF-free H-KI polypeptide group
  • a VEGF control group no H-KI polypeptide group
  • 5 parallel wells were set for each experimental group. 37.
  • H-KA and H-KB peptides at high concentrations were slightly lower than those of the control group, but the 0D values at high H-KB concentrations were statistically significant compared with the control group.
  • Difference ⁇ method, 0. 05 H-KC polypeptide in the range of 1 ⁇ -10 ⁇ , the 0D value of each hole did not change significantly, the difference is not statistically significant ⁇ (Z6Y method, ⁇ >0.05) ⁇ ⁇ - ⁇ 20 group 100 ⁇ , 1 ⁇ , ⁇
  • the 0D value of each well decreased significantly, and the difference was statistically significant, LS, P ⁇ 0.01), indicating that the H-KI20 polypeptide was at a concentration of 100 ⁇ . 1 ⁇ , ⁇ can effectively inhibit the proliferation of HUVECs induced by VEGF, and the inhibition is gradually enhanced with the increase of the concentration of H-KI20 polypeptide.
  • H-KI20 polypeptide can effectively inhibit VEGF-induced vascular endothelial cell proliferation in a dose- and sequence-dependent manner.
  • H-KA, H-KB, and H-KC polypeptides did not significantly inhibit VEGF-induced vascular endothelial cell proliferation.
  • RF/6A cells monkey chorioretinal endothelial cell line, ATCC CRL-1780
  • 96-well plates 5 ⁇ 10 4 cells/well
  • the cells were serum starved overnight
  • the experimental group was added with peptide (H).
  • -KI20 set the concentration gradient (in order of ⁇ , ⁇ , 1 ⁇ , ⁇ )
  • the positive control group was added recombinant human VEGF 165 100ng/ml (rh-VEGF 165 , Sigma, St. Louis, MO), the blank control group did not Add any reagents. Make 6 replicate wells per concentration for each group.
  • MTS (3-(4, 5- dimethylthiazol-2-yl)-5-(3- carboxymethoxyphenyl) - 2-(4- sul phophenyl) - 2H-tetrazolium) assay (CellTiter 96 AQ) Promega, Madison, WI, USA), continued to incubate for 4 h, and measured absorbance (0D value) at 490 nm.
  • the 0D values of the KI20 group were 0.461 ⁇ 0.026, 0.508 ⁇ 0.026, 0.477 ⁇ 0.029, 0.469 ⁇ 0.022, 0.436 ⁇ 0.010, and 0.429 ⁇ 0.029, respectively.
  • the 0D value of each concentration group decreased with the increase of concentration, indicating that VEGF promoted. Cell proliferation is inhibited by the polypeptide.
  • the inhibitory cell proliferation activity of K1 of HDF ranged from 57 nM to 116 nM, and the activity increased slowly from 116 nM to 925 nM. After that, the activity did not change significantly with increasing concentration.
  • rhVEGF Containing 100 ng / ml rhVEGF RPMI1640 165 serum-free medium was added at 24 well plate chamber, the RF / 6A cells were added to the upper chamber (the Transwell chamber, a porous membrane pore size 8 ⁇ , Corning Corporation), a cell concentration of 4X10 6 cells / ml
  • the concentration gradient was set: ⁇ , ⁇ , ⁇ , no polypeptide was added to the upper chamber of the control group, and a blank control group was set up (the upper chamber was a cell suspension, and the lower chamber was not.
  • 165 serum-free medium containing rhVEGF. Make two duplicate wells per peptide concentration.
  • the migration ability of RF/6A cells was detected by counting the number of cells migrating through the Transwell chamber porous membrane.
  • the cells migrated from the upper chamber through the porous membrane to the direction of high VEGF concentration (lower chamber).
  • cell migration to the lower chamber was inhibited by the addition of the polypeptide to the upper chamber.
  • Vascular endothelial cell lumen formation assay was performed using Matrigel (BD) in combination with VEGF to induce lumen formation.
  • a 96-well plate is coated with matrigel (growth factor-reduced Matrigel, BD Biosciences, USA).
  • the polypeptide group RF/6A cells were pretreated with different concentrations of ⁇ - ⁇ 20 (10 ⁇ , ⁇ , 1 ⁇ , 10 ⁇ ) for 30 minutes, and then inoculated into a 96-well plate coated with Matrigel at a cell concentration of 4.5 ⁇ 10 5 cells/ml. And add 100 ng / ml VEGF.
  • a blank control group without VEGF and peptide
  • a VEGF control group containing only VEGF
  • H-KI20 has the ability to inhibit the lumen formation of vascular endothelial cells in vitro and is concentration dependent.
  • the in vivo chicken chorioallantoic membrane (CAM) assay was used to further confirm the effect of H-KI20 polypeptide on inhibiting neovascularization in vivo.
  • 5 ⁇ 1 peptide (experimental group, set concentration gradient 25 ⁇ ⁇ /5 ⁇ 1, 50 ⁇ ⁇ /5 ⁇ 1) or PBS (control group) to the 5mm diameter filter paper (Whatman quantitative filter papers, Sigma) on the ultra-clean bench and dry dry.
  • the fertilized eggs are cleaned and disinfected, placed in an incubator, and turned daily until the 6th day of embryonic age.
  • the eggs are opened, and the filter paper is placed between the large blood vessels of the chicken embryo chorioallanes, which are relatively rare, and sealed.
  • the window was opened and incubated for 48 hours.
  • the number of 3-5 blood vessels in the range of 2.5 mm around the filter paper was counted under a microscope.
  • Ten eggs were set for each concentration in each group, and the differences between the groups were statistically compared.
  • the chicken embryo allantoic membrane (CAM) evaluation experiment is a stable model for evaluating the activity of small molecules affecting angiogenesis.
  • the filter paper with or without peptide is placed on the CAM. After 48 hours of incubation, the small blood vessels around the filter paper are counted. number.
  • ⁇ - ⁇ 20 polypeptide can significantly inhibit the growth of chicken chorioallantoic capillary, and with the increase of ⁇ - ⁇ 20 polypeptide dose, the inhibition of neovascularization is enhanced, with a good dose-dependent.
  • each nest is a group.
  • Group 1 Normal control group, reared in normal air with lactating mothers;
  • Group 2 oxygen-induced retinal neovascularization model group, group 3: PBS injection group;
  • group 4 and group 5 were oxygen induced by peptide intervention
  • groups 2-5 were housed in a 75 ⁇ 2% hyperoxia environment for 5 days with lactating mothers, and the oxygen partial pressure in the container was continuously monitored with an oxygen meter. It was then returned to normal air for 5 days to induce retinal neovascularization.
  • group 2 PBS group
  • group 3, 4 polypeptide group
  • PBS and H-KI20 10 mM or 50 mM
  • the eyeballs were removed, and some of the eyeballs were used as retinal patches and Alexa Fluor 568 conjugated i solect in B4 (Molecular Probes) staining to show Retinal blood vessels, photographed under a fluorescence microscope for qualitative analysis; some eyeballs were used as paraffin sections for hematoxylin and eosin staining, photographed under a light microscope and counted in each section of the neovascular lumen (defined as growing in the inner retinal membrane) The number of vascular lumens before the vitreous cavity surface. Statistical analysis and comparison of differences between groups.
  • H-KI20 can significantly inhibit the formation of oxygen-induced retinal neovascularization, and the inhibition of neovascularization with the increase of peptide dosage, indicating that H-KI20 has a good inhibitory effect on neovascularization in vivo.
  • Derived polypeptide 1 sequence identical to SEQ ID NO 1, wherein the 6th Arg is replaced by Gly
  • Derived polypeptide 2 the sequence is the same as SEQ ID NO 1, wherein the 2nd position is replaced by Leu; the derivative polypeptide 3: the sequence is the same as SEQ ID NO 1, wherein the 15th Thr is replaced by Ser; the derivative polypeptide 4: the sequence is the same as SEQ ID NO 1, wherein the fourth Lys is replaced by Arg; the derivatized polypeptide 5: the sequence is the same as SEQ ID NO 1, wherein the 18th Gly is replaced by Ala; the derived polypeptide 6: the sequence is the same as SEQ ID NO 1, wherein the first position before the N-terminus Addition of Cys; Derived polypeptide 7: Sequence identical to SEQ ID NO: 1, wherein the addition of CQP after the 20th position of the C-terminus showed that HUVEC cell proliferation was significantly inhibited in the treatment group ( ⁇ ) of the above-derived polypeptides 1-7.
  • the H-KI20 and its derived polypeptides of the present invention all show good anti-mouse corneal pathological neovascularization, anti-retinal pathological neovascularization, and inhibition of vascular endothelial cell proliferation, migration, chemotaxis in vitro. And the role of lumen formation. Therefore, it has broad application prospects. All documents mentioned in the present application are hereby incorporated by reference in their entirety in their entireties in the the the the the the the the the the the the the the the the the the. In addition, it should be understood that various modifications and changes may be made by those skilled in the art in the form of the present invention.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Urology & Nephrology (AREA)
  • Dermatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Reproductive Health (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Rheumatology (AREA)
  • Hematology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Pregnancy & Childbirth (AREA)
  • Ophthalmology & Optometry (AREA)
  • Endocrinology (AREA)
  • Pain & Pain Management (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Diabetes (AREA)
  • Biomedical Technology (AREA)

Abstract

提供的是一种具有抑制血管生成活性的多肽,所述多肽源自肝细胞生长因子的第129-148位氨基酸。还提供的是所述多肽的制备方法以及含有所述多肽的药物组合物。

Description

具有抑制血管生成活性的多肽
技术领域
本发明涉及生物医药领域, 更具体地, 涉及新的具有抑制新生血管作用的 多肽 (H-KI系列多肽)。 该多肽可抑制体外血管内皮细胞增殖、 迁移、 管腔形成 和可抑制体内鸡胚***和小鼠视网膜新生血管。 本发明还涉及所述多肽的制 法和应用以及含所述多肽的药物组合物。 背景技术
血管新生是指在原有毛细血管网的基础上, 通过血管内皮细胞增殖、 迁移 进而形成新生血管的过程。 血管新生在体内胚胎发育、 损伤修复等生理过程中 发挥重要作用, 同时也是众多新生血管性疾病, 如肿瘤生长与转移、 增殖性糖 尿病视网膜病变、 早产儿视网膜病变、 类风湿关节炎等疾病的主要病理变化。 因此新生血管抑制剂的研究与应用对于一些难治性新生血管相关疾病具有重 要意义。
眼部新生血管与多种眼病都有密切的关系, 它常常导致严重的视力损害并 最终致盲, 例如糖尿病视网膜病变(DR), 年龄相关性黄斑变性(AMD) , 角膜新 生血管, 新生血管性青光眼等。 目前临床上的治疗手段主要包括手术、 激光和 药物。 手术需要一定的指征, 患者痛苦大, 且风险和并发症相对较高; 激光治 疗亦存在一定的痛苦, 可造成局部视野缺损、 诱发新生血管, 中长期疗效也不 甚理想。 因此, 一种无痛苦、 便捷、 有效、 患者依从性高的药物治疗眼部新生 血管的方法成为了近十余年来国内外研究的热点。
血管内皮生长因子(VEGF)是相当重要的血管生长剌激因子, 目前临床中已 经得到应用或者仍在实验研究中的药物主要是一些针对 VEGF 或者其受体 (VEGFR)的抗新生血管制剂。 包括抑制 VEGF表达的 VEGF反义寡核苷酸; 抑制 VEGF受体表达的 VEGF受体反义寡核苷酸; 中和血循环中 VEGF的抗 VEGF抗体、 可溶性 VEGF受体、 VEGF Trap , 抗 VEGFR抗体、 酪氨酸激酶抑制剂等, 这些制 剂在肿瘤治疗中应用广泛。
在开发有效的血管新生抑制剂时, 应充分考虑到眼科用药的特殊性。
第一, 眼部存在多个解剖性和功能性的屏障。 全身给药常常由于血 -房水 屏障和血-视网膜屏障而无法在眼组织局部达到足够的药物浓度; 局部给药, 如玻璃体腔注射, 大于 76. 5kDa的大分子在理论上很难穿透视网膜作用于视网 膜和脉络膜新生血管。 对于眼表给药, 药物必须要先后穿透亲脂性的角膜上皮 细胞紧密连接和亲水性的角膜基质, 因此只有具备适当脂溶性、 低分子量或能 与眼表组织内的转运体(如: 氨基酸转运体、 寡肽转运体等)结合的药物才能到 达前房发挥作用。
第二, 药物在亲水的泪液、 房水、 玻璃体液中溶解的程度与其有效性呈正 相关。
第三, 基于上述主要原因, 眼科用药的生物利用度很低; 要使之提高, 可 加大给药的浓度。 用于***新生血管的化合物毒副作用较为明显, 全身和 局部均无法高剂量给药。
第四, 目前虽然已经有一系列相对安全的内源性血管新生抑制剂被先后证 实, 如血管抑素(angi ostat in)可明显抑制血管依赖性肿瘤的生长, 但由于其 分子量较大且空间构象复杂, 故在制备过程中存在重组表达纯化工艺繁琐和内 毒素残留等不足。
正是由于上述种种条件的限制, 目前用于治疗眼部新生血管的药物十分有 限, 比如重组抗人 VEGF单克隆抗体 Bevac i zumab (Avast in, 最初用以治疗转移 性结肠直肠肿瘤)、 重组抗人 VEGF 单克隆抗体片段 Ranibi zumab (Lucent i s,) 等。虽然这几种药物抑制眼部新生血管的作用已经得到肯定,但它们价格高昂, 已报道有多种眼部或全身不良反应, 且穿透血眼屏障能力低, 给药方式受到限 制。
多肽类新生血管抑制剂与目前研究广泛的蛋白类新生血管抑制剂相比具 有合成方法简单、 容易进行化学修饰、 免疫原性低、 溶解性好、 生物利用率高、 组织穿透性强、 给药途径多样、 价格低廉等突出优势。 然而, 目前尚没有来自 肝细胞生长因子 (HGF)的、 效果令人满意的小分子多肽。
因此, 本领域迫切需要开发一种适于眼球组织的有效安全的小分子新生血 管抑制剂。 发明内容
本发明的目的是提供一类适于眼球组织的有效安全的可抑制血管新生的 小分子多肽以及其片段、 类似物和衍生物。
本发明的另一目的是提供含所述多肽的制法和应用。 在本发明的第一方面, 提供了一种下式 I表示的多肽, 或其药学上可接受
[XaaO] - [Xaal] - [Xaa2]— [Xaa3] - [Xaa4] - [Xaa5] - [Xaa6] - [Xaa7] - [Xa a8] - [Xaa9] - [XaalO] - [Xaal 1] - [Xaal2] - [Xaal3] - [Xaal4] - [Xaal5] - [Xaa 16] - [Xaal7] - [Xaal8] - [Xaal9] - [Xaa20] - [Xaa21] (I)
式中,
XaaO是无, 或 1-3个氨基酸构成肽段;
Xaal是选自下组的氨基酸 He , Leu, Val, Met或 Ala;
Xaa2是选自下组的氨基酸 He , Leu, Val, Met或 Ala;
Xaa3是选自下组的氨基酸 Gly或 Ala;
Xaa4是选自下组的氨基酸 Lys或 Arg ;
Xaa5是选自下组的氨基酸 Gly或 Ala;
Xaa6是选自下组的氨基酸 Arg, Lys或 Gly;
Xaa7是选自下组的氨基酸 Ser或 Thr
Xaa8是选自下组的氨基酸 Tyr或 Phe
Xaa9是选自下组的氨基酸 Lys或 Arg
XaalO是选自下组的氨基酸 Gly或 Ala;
Xaal l是选自下组的氨基酸 Thr或 Ser ;
Xaal2是选自下组的氨基酸 Val, Leu , He , Met或 Ala;
Xaal3是选自下组的氨基酸 Ser或 Thr ;
Xaal4是选自下组的氨基酸 He , Leu , Val, Met或 Ala;
Xaal5是选自下组的氨基酸 Thr或 Ser ;
Xaal6是选自下组的氨基酸 Lys或 Arg ;
Xaal7是选自下组的氨基酸 Ser或 Thr ;
Xaal8是选自下组的氨基酸 Gly或 Ala;
Xaal9是选自下组的氨基酸 He , Leu , Val, Met或 Ala;
Xaa20是选自下组的氨基酸 Lys或 Arg ;
Xaa21是无, 或 1-3个氨基酸构成肽段;
并且所述的多肽具有抑制血管新生的活性,且所述多肽的长度为 20-26 个 在另一优选例中, 所述多肽的长度为 20-23个氨基酸。
在另一优选例中, XaaO和 /或 Xaa21是 1-3个氨基酸构成的肽段。 更佳地, XaaO为 C、 NC、 或 RNC; 和 /或乂3&21为(、 CQ或 CQP。
在另一优选例中, Xaal是选自下组的氨基酸: lie或 Leu;
Xaa2是选自下组的氨基酸: lie或 Leu;
Xaa3是选自下组的氨基酸: Gly或 Ala;
Xaa4是选自下组的氨基酸: Lys或 Arg;
Xaa5是选自下组的氨基酸: Gly或 Ala;
Xaa6是选自下组的氨基酸: Arg或 Lys;
Xaa7是选自下组的氨基酸: Ser或 Thr;
Xaa8是选自下组的氨基酸: Tyr或 Phe;
Xaa9是选自下组的氨基酸: Lys或 Arg;
XaalO是选自下组的氨基酸: Gly或 Ala;
Xaall是选自下组的氨基酸: Thr或 Ser;
Xaal2是选自下组的氨基酸: Val或 Leu;
Xaal3是选自下组的氨基酸: Ser或 Thr;
Xaal4是选自下组的氨基酸: lie或 Leu;
Xaal5是选自下组的氨基酸: Thr或 Ser;
Xaal6是选自下组的氨基酸: Lys或 Arg;
Xaal7是选自下组的氨基酸: Ser或 Thr;
Xaal8是选自下组的氨基酸: Gly或 Ala;
Xaal9是选自下组的氨基酸: lie或 Leu; 和 /或
Xaa20是选自下组的氨基酸: Lys或 Arg。
在另一优选例中, Xaa6为 Gly。
在另一优选例中, 所述多肽选自下组:
(a)具有 SEQ ID NO: 1所示氨基酸序列的多肽;
(b)将 SEQ ID N0:1所示氨基酸序列经过 1-5个(较佳地 1-3, 更佳地 1-2 个)氨基酸残基的取代、缺失或添加而形成的,且具有抑制血管新生功能的由(a) 衍生的多肽。
在另一优选例中, 所述的衍生多肽保留了 70%的 SEQ I画: 1的所示多肽 的抑制血管新生活性。 在另一优选例中, 所述的衍生多肽与 SEQ ID NO : 1的相同性 80%, 较佳地 90%; 更佳地 95%。
本发明还提供了抑制血管新生功能的、式 I化合物的二聚体和多聚体形式。 在本发明的第二方面, 提供了一种分离的核酸分子, 它编码本发明上述的 多肽。
在本发明的第三方面, 提供了一种药物组合物, 它含有:
(a) 本发明上述的多肽或其药学上可接受的盐; 和
(b) 药学上可接受的载体或赋形剂。
在另一优选例中, 所述组合物的剂型为眼药水、 针剂(如眼周和眼内注射 液)、 眼用凝胶或眼药膏。
在另一优选例中, 所述的组合物为缓释剂型。
在本发明的第四方面, 提供了一种本发明所述多肽或药学上可接受的盐的 用途, 它们被用于制备用于抑制血管新生或防治与血管新生相关疾病的药物。
在另一优选例中, 所述的与血管新生相关疾病的选自下组: 新生血管性眼 病、 肿瘤、 缺血性心脏病、 非炎症性心肌病、 冠状动脉硬化、 闭塞性动脉硬化、 动脉栓塞、 动脉血栓、 Berger's 病、 慢性炎症、 炎症性肠病、 溃疡、 风湿性关 节炎、 硬皮症、 银屑病、 不育症或肉瘤状病等。
在另一优选例中, 所述的新生血管性眼病包括累及脉络膜、 视网膜、 角膜 或虹膜, 包括老年性黄斑变性、 增生性糖尿病视网膜病变、 视网膜血管阻断性 疾病、 早产儿视网膜病变、 角膜感染、 新生血管性青光眼等。
在本发明的第五方面, 提供了一种抑制哺乳动物血管新生的方法, 包括步 骤: 给需要的对象施用本发明所述的多肽或其药学上可接受的盐。
在另一优选例中, 所述的对象是人。
在另一优选例中, 所述的血管新生是与新生血管性眼病相关的血管新生。 附图说明
下列附图用于说明本发明的具体实施方案, 而不用于限定由权利要求书所 界定的本发明范围。
图 1显示了 MTS细胞增殖实验中各组 0D值均值。
对照组 (control)和 VEGF组差异有显著性 (P=0.001), VEGF组和多肽组差 异有显著性 CP<0.001), 加入 ΙΟηΜ, ΙΟΟηΜ, 1 μΜ, 10μΜ Η-ΚΙ20后的 OD值, 呈递减趋势。 星号表示该组与 VEGF 组之间差异具有统计学意义 (F=7.684, <0.001) c (con: H-KI20浓度; 1 : Ι ΟηΜ; 2 : ΙΟΟηΜ; 3: Ι μΜ; 4: 10μΜ)。
图 2显示了 Transwel l细胞迁移实验中 VEGF组和 VEGF+不同浓度多肽组通 过多孔膜的细胞数量(条柱图)以及 VEGF组和加不同浓度的 H-KI20组迁移过多 孔膜的细胞情况(彩色照片)。
在条柱图中可以看到, 多肽组较 VEGF 组细胞数目明显减少, 且随浓度增 加呈递减趋势。 组间差异具统计学意义(^=562. 3, 尸<0. 001)。 星号表示该组与 VEGF组之间差异具有统计学意义。 照片中透明的小圆孔为多孔膜的孔, 蓝紫色 的细胞是被苏木苏着染的通过小孔迁移过滤膜的细胞。
图 3显示了内皮细胞体外管腔形成实验中空白对照组、 VEGF组和 VEGF+不 同浓度多肽组每 100倍视野形成管腔的平均总长度(条柱图),以及空白对照组、 VEGF组和 VEGF+ΙΟμΜ H-KI20组的内皮细胞管腔形成情况(照片)。
在条柱图中可以看到, VEGF+多肽组较 VEGF组管腔形成总长度明显下降, 且随浓度增加呈递减趋势。 组间差异具统计学意义 9, 0. 001)。 星号 表示该组与 VEGF组之间差异具有统计学意义。 (con: H-KI20浓度; 1 : 10nM; 2: Ι ΟΟηΜ; 3: Ι μΜ; 4: 10μΜ)。
图 4显示了 PBS组和多肽组鸡胚***(CAM)上血管生长情况(彩色照片) 以及评估实验中各组 CAM血管计数情况(条柱图)。
照片中白色的圆片为滤纸片, 黑色的线圈划定的范围为血管计数范围, 橙 黄色的血管膜为鸡胚***, 可以看到加有 10μ§/μ1 Η-ΚΙ20 的滤纸片周围的 血管数目显著少于 PBS组。条柱图中对照组为 PBS组,可以看到 5μ§/μ1 Η-ΚΙ20 组以及 10μ§/μ1 Η-ΚΙ20组血管数量较 PBS组显著下降, 且随浓度增加呈递减 趋势。 星号表示该组与对照组之间差异具有统计学意义 762, O. OO D o 图 5是视网膜新生血管模型的视网膜铺片、 眼球切片照片以及对眼球切片 中新生血管管腔数目的统计结果(条柱图)。
在正常氧浓度下饲养的对照组幼鼠视网膜血管发育良好, 未见无灌注区和 新生血管, 通过高氧环境诱导的视网膜新生血管模型组幼鼠的视网膜铺片中央 可见大面积无灌注区以及新生血管团(椭圆圈标示),经过玻璃体腔注射 H-KI20 干预后的视网膜新生血管模型组幼鼠的视网膜铺片中新生血管团数目明显减 少。 从眼球切片中可以看到, 正常组视网膜前未见新生血管管腔, 视网膜新生 血管模型幼鼠的视网膜前可以看到较多新生血管管腔(黑色箭头所示), 经 H-KI20干预后这些新生血管管腔数目显著减少, 星号表示该组与 VEGF组之间 差异具有统计学意义(/^=79. 320, 尸<0. 001)。 (con: H-KI20浓度; 1 : 10mM; 2: 50mM) o 具体实施方式
本发明人经过广泛而深入的研究, 首次制备了一类源自肝细胞生长因子 (HGF)的、 具有抑制血管新生功能的, 分子量小于 5kD (如仅约 2kD)的小分子多 肽。 具体而言, 本发明人应用生物信息学的方法, 基于同源性分析和生物学特 性等分析, 设计了数个候选序列, 采用固相法将其合成后, 再经鸡胚***血 管模型、 VEGF 诱导的细胞株增殖模型和缺氧诱导小鼠视网膜新生血管模型筛 选, 获得了一类新型的、 具有预防和治疗血管新生功能的小分子多肽。
本发明的小肽的分子量小, 可透过各种眼组织屏障; 水溶性好, 能在中性 泪液、 房水和玻璃体液中保持较高的浓度; 安全性高, 对生物组织毒副作用小; 眼局部用药生物利用度高, 能穿透血眼屏障、 可减少剂量, 从而减小全身副作 用。 在此基础上完成了本发明。 活性多肽
在本发明中, 术语 "本发明多肽" 、 "H-KI20多肽" 、 "H-KI20小肽" 、 "短肽 H-KI20 "或 "肽 H-KI20 "可互换使用, 都指具有血管新生抑制活性的肽 H-KI20 氨基酸序列(SEQ ID N0 : 1)的蛋白或多肽。 此外, 所述术语还包括具有 抑制血管新生功能的、 SEQ ID NO: 1 序列的变异形式。 这些变异形式包括(但 并不限于): 1-5个(通常为 1-4个, 较佳地 1-3个, 更佳地 1-2个, 最佳地 1 个)氨基酸的缺失、 ***和 /或取代, 以及在 C末端和 /或 N末端添加或缺失一 个或数个(通常为 5个以内, 较佳地为 3个以内, 更佳地为 2个以内)氨基酸。 例如, 在本领域中, 用性能相近或相似的氨基酸进行取代时, 通常不会改变蛋 白质的功能。 又比如, 在 C末端和 /或 N末端添加或缺失一个或数个氨基酸通 常也不会改变蛋白质的结构和功能。 此外, 所述术语还包括单体和多聚体形式 本发明多肽。 该术语还包括线性以及非线性的多肽 (如环肽)。
本发明还包括 H-KI20多肽的活性片段、 衍生物和类似物。 如本文所用, 术 语 "片段" 、 "衍生物" 和 "类似物" 是指基本上保持抑制血管新生功能或活 性的多肽。 本发明的多肽片段、 衍生物或类似物可以是(i)有一个或多个保守或 非保守性氨基酸残基 (优选保守性氨基酸残基)被取代的多肽, 或(i i)在一个或 多个氨基酸残基中具有取代基团的多肽, 或(i i i) H-KI20 多肽与另一个化合物 (比如延长多肽半衰期的化合物, 例如聚乙二醇)融合所形成的多肽, 或(iv)附 加的氨基酸序列融合于此多肽序列而形成的多肽(与前导序列、分泌序列或 6His 等标签序列融合而形成的然后蛋白)。 根据本文的教导, 这些片段、 衍生物和类 似物属于本领域熟练技术人员公知的范围。
一类优选的活性衍生物指与式 I的氨基酸序列相比, 有至多 5个, 较佳地 至多 3个, 更佳地至多 2个, 最佳地 1个氨基酸被性质相似或相近的氨基酸所 替换而形成多肽。 这些保守性变异多肽最好根据表 I进行氨基酸替换而产生。
表 I
Figure imgf000009_0001
发明还提供 H-KI20多肽的类似物。 这些类似物与天然 H-KI20多肽的差别 可以是氨基酸序列上的差异, 也可以是不影响序列的修饰形式上的差异, 或者 兼而有之。类似物还包括具有不同于天然 L-氨基酸的残基(如 D-氨基酸)的类似 物, 以及具有非天然存在的或合成的氨基酸(如 β、 Υ -氨基酸)的类似物。 应理 解, 本发明的多肽并不限于上述例举的代表性的多肽。
修饰(通常不改变一级结构)形式包括: 体内或体外的多肽的化学衍生形式 如乙酰化或羧基化。 修饰还包括糖基化, 如那些在多肽的合成和加工中或进一 步加工步骤中进行糖基化修饰而产生的多肽。 这种修饰可以通过将多肽暴露于 进行糖基化的酶(如哺乳动物的糖基化酶或去糖基化酶)而完成。 修饰形式还包 括具有磷酸化氨基酸残基(如磷酸酪氨酸, 磷酸丝氨酸, 磷酸苏氨酸)的序列。 还包括被修饰从而提高了其抗蛋白水解性能或优化了溶解性能的多肽。
本发明多肽还可以以由药学上或生理学可接受的酸或碱衍生的盐形式使 用。 这些盐包括(但不限于)与如下酸形成的盐: 氢氯酸、 氢溴酸、 硫酸、 柠檬 酸、 酒石酸、 磷酸、 乳酸、 丙酮酸、 乙酸、 琥珀酸、 草酸、 富马酸、 马来酸、 草酰乙酸、 甲磺酸、 乙磺酸、 苯磺酸、 或羟乙磺酸。 其他盐包括: 与碱金属或 碱土金属(如钠、 钾、 钙或镁)形成的盐, 以及以酯、 氨基甲酸酯或其他常规的 "前体药物" 的形式。 编码序列
本发明还涉及编码 Η-ΚΙ20多肽的多核苷酸。 一种优选的编码序列是 atcattggta aaggacgcag ctacaaggga acagtatcta tcactaagag tggcatcaaa (SEQ ID NO : 3), 它编码 SEQ ID NO : 1所示的氨基酸序列。
本发明的多核苷酸可以是匪形式或 RNA形式。 匪可以是编码链或非编 码链。 编码成熟多肽的编码区序列可以与 SEQ ID NO : 3所示的编码区序列相同 或者是简并的变异体。 如本文所用, 以 SEQ ID N0 : 1 为例, "简并的变异体" 在本发明中是指编码具有 SEQ ID N0 : 1序列的多肽, 但与 SEQ ID NO : 3中相应 编码区序列有差别的核酸序列。
本发明的 H-KI20核苷酸全长序列或其片段通常可以用 PCR扩增法、重组法 或人工合成的方法获得。 目前, 已经可以完全通过化学合成来得到编码本发明 多肽(或其片段, 或其衍生物)的 DNA序列。 然后可将该 DNA序列引入本领域中 已知的各种现有的 DNA分子(或如载体)和细胞中。
本发明也涉及包含本发明的多核苷酸的载体, 以及用本发明的载体或
H-KI20多肽编码序列经基因工程产生的宿主细胞。 另一方面, 本发明还包括对 H-KI20 DNA或是其片段编码的多肽具有特异性 的多克隆抗体和单克隆抗体, 尤其是单克隆抗体。 制备方法
本发明多肽可以是重组多肽或合成多肽。本发明的多肽可以是化学合成的, 或重组的。 相应地, 本发明多肽可用常规方法人工合成, 也可用重组方法生产。
一种优选的方法是使用液相合成技术或固相合成技术,如 Boc固相法、 Fmoc 固相法或是两种方法联合使用。 固相合成可快速获得样品, 可根据目的肽的序 列特征选用适当的树脂载体及合成***。 例如, Fmoc***中优选的固相载体如 连接有肽中 C端氨基酸的 Wang树脂, Wang树脂结构为聚苯乙烯, 与氨基酸间 的手臂是 4-垸氧基苄醇; 用 25%六氢吡啶 /二甲基甲酰胺室温处理 20分钟, 以 除去 Fmoc保护基团, 并按照给定的氨基酸序列由 C端逐个向 N端延伸。 合成完 成后, 用含 4% 对甲基苯酚的三氟乙酸将合成的胰岛素原相关肽从树脂上切割 下来并除去保护基, 可过滤除树脂后***沉淀分离得到粗肽。 将所得产物的溶 液冻干后, 用凝胶过滤和反相高压液相层析法纯化所需的肽。 当使用 Boc*** 进行固相合成时, 优选树脂为连接有肽中 C端氨基酸的 PAM树脂, PAM树脂结 构为聚苯乙烯, 与氨基酸间的手臂是 4-羟甲基苯乙酰胺; 在 Boc合成***中, 在去保护、 中和、 偶联的循环中, 用 TFA/二氯甲垸 (DCM)除去保护基团 Boc 并 用二异丙基乙胺 (DIEA/二氯甲垸中和。 肽链缩合完成后, 用含对甲苯酚(5-10%) 的氟化氢 (HF),在 0°C下处理 1小时,将肽链从树脂上切下, 同时除去保护基团。 以 50-80%乙酸(含少量巯基乙醇)抽提肽,溶液冻干后进一步用分子筛 S印 hadex G10或 Tsk-40f 分离纯化, 然后再经高压液相纯化得到所需的肽。 可以使用肽 化学领域内已知的各种偶联剂和偶联方法偶联各氨基酸残基, 例如可使用二环 己基碳二亚胺(DCC),羟基苯骈三氮唑(HOBt)或 1, 1, 3, 3-四脲六氟磷酸酯(HBTU) 进行直接偶联。 对于合成得到的短肽, 其纯度与结构可用反相高效液相和质谱 分析进行确证。
在一优选例中,本发明多肽 H-KI20 ,按其序列,采用固相合成的方法制备, 行高效液相色谱纯化, 获得高纯度目的肽冻干粉, -2CTC贮存。
另一种方法是用重组技术产生本发明多肽。 通过常规的重组 DNA技术, 可 利用本发明的多核苷酸可用来表达或生产重组的 H-KI20多肽。一般来说有以下 步骤: (1) .用本发明的编码 H-KI20多肽的多核苷酸 (或变异体),或用含有该多核 苷酸的重组表达载体转化或转导合适的宿主细胞;
(2) .在合适的培养基中培养的宿主细胞;
(3) .从培养基或细胞中分离、 纯化蛋白质。
重组多肽可在细胞内、 或在细胞膜上表达、 或分泌到细胞外。 如果需要, 可利用其物理的、化学的和其它特性通过各种分离方法分离和纯化重组的蛋白。 这些方法是本领域技术人员所熟知的。 这些方法的例子包括但并不限于: 常规 的复性处理、 用蛋白沉淀剂处理(盐析方法)、 离心、 渗透破菌、 超处理、 超离 心、 分子筛层析(凝胶过滤)、 吸附层析、 离子交换层析、 高效液相层析 (HPLC) 和其它各种液相层析技术及这些方法的结合。
由于本发明多肽较短, 因此可以考虑将多个多肽串联在一起, 重组表达后 获得多聚体形式的表达产物, 然后通过酶切等方法形成所需的小肽。 药物组合物和施用方法
另一方面, 本发明还提供了一种药物组合物, 它含有(a)安全有效量的本 发明多肽或其药学上可接受的盐; 以及(b)药学上可接受的载体或赋形剂。 本 发明多肽的数量通常为 10微克 -100毫克 /剂, 较佳地为 100-1000微克 /剂。
为了本发明的目的, 有效的剂量为给予个体约 0. 01毫克 /千克至 50毫克 / 千克, 较佳地 0. 05毫克 /千克至 10毫克 /千克体重的本发明多肽。 此外, 本发 明的多肽可以单用, 也可与其他治疗剂一起使用(如配制在同一药物组合物 中)。
药物组合物还可含有药学上可接受的载体。 术语 "药学上可接受的载体" 指用于治疗剂给药的载体。 该术语指这样一些药剂载体: 它们本身不诱导产生 对接受该组合物的个体有害的抗体, 且给药后没有过分的毒性。 这些载体是本 领域普通技术人员所熟知的。在 Remington ' s Pharmaceut ical Sc i ences (Mack Pub. Co. , N. J. 1991)中可找到关于药学上可接受的赋形剂的充分讨论。 这类 载体包括(但并不限于): 盐水、 缓冲液、 葡萄糖、 水、 甘油、 乙醇、 佐剂及其 组合。
治疗性组合物中药学上可接受的载体可含有液体, 如水、 盐水、 甘油和乙 醇。 另外, 这些载体中还可能存在辅助性的物质, 如润湿剂或乳化剂、 pH缓冲 物质等。 通常, 可将治疗性组合物制成可注射剂, 例如液体溶液或悬液; 还可制成 在注射前适合配入溶液或悬液中、 液体载体的固体形式。
一旦配成本发明的组合物, 可将其通过常规途径进行给药,其中包括(但并 不限于): 眼表、 眼周、 眼内、 肌内、 静脉内、 皮下、 皮内或局部给药。 待预防 或治疗的对象可以是动物; 尤其是人。
当本发明的药物组合物被用于实际治疗时, 可根据使用情况而采用各种不 同剂型的药物组合物。 较佳地, 可以例举的有眼药水、 针剂、 眼用凝胶和眼药 这些药物组合物可根据常规方法通过混合、 稀释或溶解而进行配制, 并且 偶尔添加合适的药物添加剂, 如赋形剂、 崩解剂、 粘合剂、 润滑剂、 稀释剂、 缓冲剂、 等渗剂(i sotonic i t i es 防腐剂、 润湿剂、 乳化剂、 分散剂、 稳定 剂和助溶剂, 而且该配制过程可根据剂型用惯常方式进行。
例如, 眼药水的配制可这样进行: 将短肽 H-KI20 或其药学上可接受的盐 与基本物质一起溶解于无菌水(在无菌水中溶解有表面活性剂)中, 调节渗透压 和酸碱度至生理状态, 并可任意地加入合适的药物添加剂如防腐剂、 稳定剂、 缓冲剂、 等渗剂、 抗氧化剂和增粘剂, 然后使其完全溶解。
本发明的药物组合物还可以缓释剂形式给药。 例如, 短肽 H-KI20 或其盐 可被惨入以缓释聚合物为载体的药丸或微囊中, 然后将该药丸或微囊通过手术 植入待治疗的组织。 此外, 短肽 H-KI20 或其盐还可通过***预先涂有药物的 眼内透镜而得以应用。 作为缓释聚合物的例子, 可例举的有乙烯 -乙烯基乙酸 酯共聚物、 聚羟基甲基丙烯酸酯(polyhydrometaacrylate 聚丙烯酰胺、 聚 乙烯吡咯垸酮、 甲基纤维素、 乳酸聚合物、 乳酸 -乙醇酸共聚物等, 较佳地可 例举的是可生物降解的聚合物如乳酸聚合物和乳酸-乙醇酸共聚物。
当本发明的药物组合物被用于实际治疗时, 作为活性成分的短肽 H-KI20 或其药学上可接受的盐的剂量, 可根据待治疗的每个病人的体重、年龄、性别、 症状程度而合理地加以确定。例如,当局部滴眼时,通常其浓度约为 0. l-10wt%, 较佳地 l-5wt%, 每日可 2-6次给药, 每次 1-2滴。 工业应用性
含有本发明多肽或其药学上可接受盐作为活性成分的药物组合物, 对血管 新生有显著的抑制活性。 经动物试验证实, 本发明多肽不仅可以抑制鸡胚尿囊 膜的血管新生, 而且可以抑制人脐静脉血管内皮细胞的增殖、 迁移、 趋化及管 腔形成, 并且可抑制缺氧诱导的小鼠视网膜新生血管。 本发明的主要优点包括:
(a)本发明多肽的分子量小, 可透过眼组织屏障;
(b)水溶性好, 能在中性泪液、 房水和玻璃体液中保持较高的浓度;
(c)安全性高, 对生物组织毒副作用小; 并且眼局部用药生物利用度高, 可减少剂量, 从而减小全身副作用;
(d)可通过固相合成的方法制备, 纯度高, 产量大, 成本低;
(e)本发明多肽的稳定性好。
因此本发明多肽有望开发成药物, 用于治疗新生血管性眼病及相关的新生 血管性疾病, 如肿瘤新生血管等。 下面结合具体实施例, 进一步阐述本发明。 应理解, 这些实施例仅用于说 明本发明而不用于限制本发明的范围。 下列实施例中未注明具体条件的实验方 法, 通常按照常规条件如 Sambrook等人, 分子克隆: 实验室手册(New York: Cold Spring Harbor Laboratory Press, 1989)中所述的条件, 或按照制造厂 商所建议的条件。 实施例 1
多肽的合成
以人 HGF氨基酸序列(SEQ ID N0 : 2)为基础, 经生物信息学分析和筛选后, 选择序列如下的四种 T-KI多肽(图 1)。
H-KI20 : I IGKGRSYKGTVSITKSGIK (SEQ ID N0 : 1; SEQ ID NO : 2中第 129-148 位)
H-KA: SEQ ID NO : 2中第 150- 176位;
H-KB : SEQ ID NO : 2中第 178- 188位;
H-KC : SEQ ID NO : 2中第 190- 200位;
步骤如下: 利用 SYMI¾0NY 型 12 通道多肽合成仪(美国 Protein Technologi es公司), 根据其软件(Vers i on. 201版)计算和配制所需要的 Fmoc 保护氨基酸溶液,缩合试剂和切割试剂。编辑程序,其中树脂溶涨时间为 30min; 脱保护两次, 时间分别为 5min和 15min; 缩合时间为 30min; 切割时间为 2h。 开机按照上述程序合成多肽, 采用高效液相色谱仪(SHIMADZU公司)纯化多肽, 获得纯度〉 95%的白色粉末状多肽(每种多肽各制得 120mg), 冻干待用。
其中, H-KI20由 20个氨基酸残基组成, 分子量 2093. 52D, 不含半胱氨酸 残基和二硫键。 实施例 2 H-KI20多肽抑制 VEGF诱导的血管内皮细胞增殖
(1) 人脐静脉内皮细胞(Human umbi l ical vein endothel ial cel ls, HUVECs) 的体外培养
原代 HUVECs (购自 Sci enCell公司)采用 ECM培养基添加 ECGS (ScienCell公 司)以及 5%胎牛血清(ScienCell公司), 培养于 37°C、 含 5%C02的培养箱中。 本发 明中所有体外细胞实验均采用第 3〜8代HUVECs细胞。
(2) MTS方法检测 H-KI系列多肽抑制 VEGF诱导的血管内皮细胞增殖
MTS细胞增殖定量检测方法是一种通过四氮唑和电子偶联化合物在代谢旺 盛细胞线粒体脱氢酶的作用下, 产生水溶性有色产物, 作为检测信号来比色定 量测定活细胞增殖的方法。
具体实施方法如下: HUVECs生长接近融合后, 传代, 按照 3. 5 X 107ml的密 度接种于 96孔板, 每孔 100μ1, 37°C、 5%C02培养箱中培养 24h后, 更换无血清 ECM 培养基, 细胞饥饿过夜。 吸出 96孔板内培养基, 各组分别加入含有浓度为 1ηΜ、 10 nM、 100 ηΜ、 ΙμΜ 、 ΙΟμΜ Η-ΚΙ系列多肽药物的无血清培养基 50μ1, 37 °C 预处理 30min后, 各孔加入含有 VEGF (R&D公司)的无血清培养基, 使 VEGF的终浓 度为 10ng/ml。 另设空白对照组(无 VEGF无 H-KI多肽组)和 VEGF对照组(无 H-KI多 肽组), 每个实验组设置 5个平行孔。 37 、 5%C02培养箱中继续培养 24h后, 各 孔中加入 20μ1 MTS溶液(Promega公司), 37 °C作用 l-4h, 酶标仪(Bio-Rad公 司) 490nm检测各孔的吸光值。
研究结果: 与空白对照组(无 VEGF无 H-KI多肽组)相比, VEGF组各孔 0D值明 显增加, 并且差异具有统计学意义 ^法, 0. 01) ,表明 10ng/mlVEGF能够有 效剌激 HUVECs增殖。
与 VEGF组相比, H-KA和 H-KB多肽在高浓度时的 0D值较对照组略有下降, 但 仅在 H-KB高浓度时的 0D值与对照组相比有统计学意义的差异 ^法, 0. 05 ) , H-KC多肽在 1ηΜ-10μΜ浓度范围内, 各孔 0D值无明显改变, 差异不具有统计学意 义(Z6Y法, Ρ>0.05) ο Η-ΚΙ20组 100ηΜ、 1μΜ、 ΙΟμΜ 时各孔 0D值明显降低, 并 且差异具有统计学意义、LS , P<0.01),表明 H-KI20多肽在浓度为 100ηΜ、 1μΜ、 ΙΟμΜ 时能够有效抑制 VEGF诱导的 HUVECs增殖, 并且随着 H-KI20多肽浓度的增 力口, 抑制作用逐渐增强。
结论: H-KI20多肽能够有效抑制 VEGF诱导的血管内皮细胞增殖, 并且具有 良好的剂量依赖性和序列依赖性。 H-KA、 H-KB、 H-KC多肽不具有明显的抑制 VEGF 诱导的血管内皮细胞增殖作用。 实施例 3 MTS细胞增殖实验
将 RF/6A 细胞(猴脉络膜视网膜内皮细胞株, ATCC CRL— 1780)接种于 96 孔板(5X104细胞 /孔), RPMI1640培养基培养 24h后, 使细胞血清饥饿过夜, 实验组加入多肽(H-KI20), 设置浓度梯度(依次为 ΙΟηΜ, ΙΟΟηΜ, 1μΜ, ΙΟμΜ), 阳性对照组加入重组的人 VEGF165100ng/ml(rh-VEGF165, Sigma, St. Louis, MO), 空白对照组不加任何试剂。 每组每个浓度做 6个复孔。 作用 24h后, 每孔加入 20μ1 MTS (3- (4, 5- dimethylthiazol- 2- yl) - 5- (3- carboxymethoxyphenyl) - 2- (4- sul phophenyl) - 2H- tetrazolium) assay (CellTiter 96 AQ; Promega, Madison, WI, USA) , 继续孵育 4h, 酶标仪 490nm下测量吸光度(0D值)。
结果: 24h时各组平均 0D值如下图 1所示, VEGF组的 0D值较空白组增高, 加入多肽 H-KI20后, 0D值出现回落, 且随多肽浓度升高而逐步下降, 0D值组 间差异具有统计学意义( ^=7.684, 0.001,ANOVA);空白组、 VEGF组、 VEGF+10nM H-KI20组、 VEGF+100nM H - KI20组、 VEGF+ΙμΜ H - KI20组、 VEGF+ΙΟμΜ H - KI20 组的 0D值分别为 0.461±0.026、 0.508±0.026、 0.477±0.029、 0.469±0.022、 0.436±0.010、 0.429±0.029, 各浓度组的 0D值随浓度增加而成下降趋势, 表 明 VEGF的促细胞增殖作用受到多肽抑制。
根据国外已发表文献, 在细胞增殖实验(接种浓度是 1.5X104细胞 /ml)中,
HDF的 K1的抑制细胞增殖活性随浓度增加最快的范围是 57nM --- 116nM, 活性增 加缓慢的浓度范围是 116 nM -— 925 nM, 之后随浓度继续增加, 活性无明显改 变。
与之相比,在本实施例的细胞增殖实验中,所用 H-KI20浓度从 ΙΟηΜ, ΙΟΟηΜ, ΙμΜ到 10μΜ时, 抑制细胞增殖活性增加明显。 另外, 本实施例中的接种浓度更 高(5X105细胞 /ml), 因此多肽 H-KI20抑制增殖活性远高于 Kl。 实施例 4 H-KI20多肽抑制内皮细胞迁移
将含有 100 ng/ml rhVEGF165的无血清 RPMI1640培养基加入 24孔板下室, 将 RF/6A细胞加入上室(Transwell小室, 多孔膜孔径 8μπι, Corning公司), 细 胞浓度为 4X106细胞 /ml, 实验组在上室中另加入多肽 H-KI20, 设置浓度梯度: ΙΟηΜ, ΙΟΟηΜ, ΙμΜ, 对照组上室中不加多肽,另设置空白对照组(上室为细胞悬 液, 下室为不含 rhVEGF165的无血清培养基)。 每个多肽浓度做两个复孔。 37°C 5%C02培养箱中培养 24h。 棉棒擦去 Transwell小室滤膜上方细胞, 4%多聚甲醛 固定 20min, 苏木素染色, 自来水清洗, 揭下滤膜, 封片。 显微镜下每张膜随 机选取 5个视野(200倍), 计数细胞。 实验重复 3次。
RF/6A细胞的迁移能力通过计数迁移过 Transwell小室多孔膜的细胞数来 检测, 在这一实验中, 细胞从上室通过多孔滤膜向 VEGF浓度高的方向(下室)迁 移。 在本实验中, 通过向上室中加入多肽, 抑制细胞向下室迁移。
结果如图 2所示, 随多肽浓度的增加, 迁移的细胞数逐渐减少, VEGF组、 VEGF+lOnM H- KI20组、 VEGF+100nM H- KI20组、 VEGF+ΙμΜ H- KI20组迁移细胞平 均数目依次为 76.333±5.354、 43.333±3.670、 16.500±1.517、 1.833±1.169, 组间差异具有统计学意义 3, P<0.001, AN0VA) 0 这说明本发明的 H-KI20 多肽可以显著地抑制内皮细胞迁移。 实施例 5 H-KI20多肽抑制内皮细胞体外管腔形成
血管内皮细胞管腔形成实验采用 Matrigel(BD公司)联合 VEGF诱导管腔形 成方法。
具体实施方法如下: 将 96 孔板用基质胶包被(growth factor-reduced Matrigel , BD Biosciences , 美国)。 多肽组 RF/6A 细胞与不同浓度的 Η-ΚΙ20(10ηΜ, ΙΟΟηΜ, 1μΜ, 10μΜ)预处理 30分钟后, 接种至包被过基质胶的 96孔板内, 细胞浓度 4.5X105细胞 /ml。 并加入 100ng/mlVEGF。 另设立空白对 照组(不含 VEGF和多肽)、 VEGF对照组(仅含 VEGF)。 每组每个浓度设立 3个复 孔。 37 °C 5% C02培养箱中培养 6h。 倒置相差显微镜(Olympus, 日本)下观察并 拍照。 每孔随机取 3个视野(100倍)。 采用 Image-Pro Plus 图像分析软件(版 本 5.1, Media Cybernetics公司, 美国)统计每视野下形成管腔的总长度。 实 验重复 3次。 结果如图 3所示。 lOOng/mlVEGF显著剌激 RF/6A细胞体外管腔形成。 加入浓 度逐渐升高的 H-KI20后, RF/6A细胞体外管腔形成总长度逐渐下降, 表明管腔 形成逐渐受到抑制, 当 H-KI20浓度为 ΙΟμΜ时, 管腔形成几乎完全被抑制。 空白 组、 VEGF组、 VEGF+lOnM H - KI20组、 VEGF+lOOnM H - KI20组、 VEGF+ΙμΜ H - KI20 组、 VEGF+1(^MH-KI20管腔形成平均总长度依次为 11.31±1.59 mm, 23.47±1.18 mm, 15.66±2.01 mm, 15.11±0.55 mm, 10.29±0.92 mm, 3.78±0.65mm, 组 间差异具有统计学意义( =294.9, P<0.001, AN0VA)。
该实验结果说明: H-KI20具有抑制血管内皮细胞体外管腔形成的能力, 并 且具有浓度依赖性。 实施例 6 H-KI20多肽抑制体内鸡胚***新生血管
本实施例采用体内鸡胚***(CAM)实验进一步证实 H-KI20多肽抑制体内 新生血管的作用。
将 5μ1多肽(实验组, 设置浓度梯度 25μ§/5μ1,50μ§/5μ1)或 PBS (对照组) 在超净工作台上加至直径 5mm 的滤纸片(Whatman quantitative filter papers, Sigma)上并晾干。 将受***蛋清洗消毒后放入孵箱, 每日翻动, 至胚 龄第 6日时, 将种蛋开窗, 并将滤纸片放置于鸡胚***上微血管相对稀少的 大血管之间, 密封开窗口并孵育 48小时, 显微镜下计数滤纸片周围 2.5mm距 离的范围内 3-5级血管的数目, 每组每个浓度设置 10只种蛋, 统计比较组间 差异。
鸡胚***(CAM)评估实验是一种评估影响血管生成的小分子物质活性的 稳定模型, 将含或不含多肽的滤纸片放置于 CAM上, 孵育 48h后拍照计数滤纸片 周围小血管数目。
结果如图 4所示。 PBS组、 5μ§/μ1 Η-ΚΙ20组, 10μ§/μ1 Η-ΚΙ20组血管数目依 次为 79.430±9.829、 48.380±7.308、 27.000±9.407。 Η-ΚΙ20多肽组血管数目 显著低于对照组, 且随多肽浓度增高, 血管数目渐降。 组间差异具有统计学意 义( =54.762, Ρ<0.001, AN0VA)。
上述研究表明: Η-ΚΙ20多肽能够明显抑制鸡胚***毛细血管的生长, 并 且随着 Η-ΚΙ20多肽剂量的增加, 抑制新生血管的作用增强, 具有良好的剂量依 赖性。 实施例 7 H-KI20多肽抑制视网膜新生血管的生成
选择鼠龄 7天的健康 C57BL / 6J幼鼠 5窝, 每窝 7-9只(由中国科学院上 海实验动物中心提供)。 分成 5组, 每窝为一组。 组 1 : 正常对照组, 与哺乳母 鼠一起在正常空气环境下饲养; 组 2 : 氧诱导的视网膜新生血管模型组, 组 3: PBS注射组; 组 4与组 5为经多肽干预的氧诱导的视网膜新生血管模型组, 组 2-5与哺乳母鼠一起置于 75 ± 2%高氧环境下饲养 5天, 期间用测氧仪持续监测 容器内的氧分压。 然后回到正常空气环境下饲养 5天以诱导视网膜新生血管的 生成。 在结束高氧环境饲养的当天, 向组 2 (PBS组)和组 3、 4 (多肽组)的幼鼠 左眼行玻璃体腔分别注射 PBS和 H-KI20 (10mM或 50mM), 每只眼注射 1μ1。 在 鼠龄 17天时将各组幼鼠麻醉后行心腔灌注 4%多聚甲醛, 取出眼球固定, 部分 眼球用作视网膜铺片以及 Alexa Fluor 568 conjugated i solect in B4 (Molecular Probes公司)染色以显示视网膜血管, 在正置荧光显微镜下拍照用 作定性分析; 部分眼球用作石蜡切片苏木素伊红染色, 在光学显微镜下拍照并 统计每张切片中新生血管管腔(定义为生长于视网膜内界膜之前、 位于玻璃体 腔面的血管腔)的数目。 统计分析并比较组间差异。
结果如图 5所示。 通过对视网膜铺片的定性分析发现, 氧诱导的视网膜新 生血管模型组较正常环境饲养的对照组有大面积无灌注区以及团状的视网膜 新生血管出现(如椭圆圈所示), 注射 PBS后无明显变化, 而经 H-KI20玻璃体腔 注射治疗后, 新生血管团显著减少。 H-KI20治疗组眼球切片中新生血管腔数目 较未治疗组明显减少(如箭头所示), 柱状图提示各组的新生血管管腔数目统计 结果, 组间差异具有统计学意义 320, P<0. 001, AN0VA)。
本实验表明: H-KI20能显著抑制氧诱导的视网膜新生血管的生成, 并且随 着多肽剂量的增加, 抑制新生血管作用增强, 表明 H-KI20具有良好的抑制体内 新生血管的作用。 实施例 7
眼药水的制备
利用常规技术, 混合以下组分, 制得 1%眼药水, 其配方如下:
H-KI20多肽 10 mg
羟丙基甲基纤维素 0. 03g
无菌水 加至 10 ml 调节渗透压至 3000sm, 酸碱度(pH)至 6. 8-7. 1。
经 5位志愿者试用一周, 每日 3次, 每次 1滴 /眼。 结果表明该眼药水可 抑制眼部的血管新生。 实施例 8
衍生多肽的制备和活性
制备了以下数种衍生多肽, 并按实施例 2所示的方法,
多肽对 VEGF诱导的血管内皮细胞 HUVEC的增殖的抑制作用。
衍生多肽 1 : 序列同 SEQ ID NO 1, 其中第 6位 Arg被 Gly替换
衍生多肽 2 : 序列同 SEQ ID NO 1, 其中第 2位 l ie被 Leu替换; 衍生多肽 3 : 序列同 SEQ ID NO 1, 其中第 15位 Thr被 Ser替换; 衍生多肽 4: 序列同 SEQ ID NO 1, 其中第 4位 Lys被 Arg替换; 衍生多肽 5 : 序列同 SEQ ID NO 1, 其中第 18位 Gly被 Ala替换; 衍生多肽 6 : 序列同 SEQ ID NO 1, 其中在 N端的第 1位之前添加 Cys ; 衍生多肽 7 : 序列同 SEQ ID NO : 1, 其中在 C端的第 20位之后添加 CQP 结果表明, 上述衍生多肽 1-7的处理组(ΙΟΟηΜ)中, HUVEC细胞增殖显著受 到抑制。 综上所述, 本发明的 H-KI20 及其衍生多肽均显示良好的抗小鼠角膜病理 性新生血管、 抗小鼠视网膜病理性新生血管, 以及在体外抑制血管内皮细胞增 殖、 迁移、 趋化及管腔形成的作用。 因此, 具有广泛的应用前景。 在本发明提及的所有文献都在本申请中引用作为参考, 就如同每一篇文献 被单独引用作为参考那样。 此外应理解, 在阅读了本发明的上述讲授内容之后, 本领域技术人员可以对本发明作各种改动或修改, 这些等价形式同样落于本申 请所附权利要求书所限定的范围。

Claims

权 利 要 求
1. 一种下式 I表示的多肽, 或其药学上可接受的盐
[XaaO] - [Xaal]- [Xaa2]— [Xaa3] - [Xaa4] - [Xaa5] - [Xaa6] - [Xaa7] - [Xa a8] - [Xaa9] - [XaalO] - [Xaal 1] - [Xaal2] - [Xaal3] - [Xaal4] - [Xaal5] - [Xaa
16] - [Xaal7] - [Xaal8] - [Xaal9] - [Xaa20] - [Xaa21] (I)
式中,
XaaO是无, 或 1-3个氨基酸构成肽段;
Xaal是选自下组的氨基酸: lie, Leu, Val, Met或 Ala;
Xaa2是选自下组的氨基酸: lie, Leu, Val, Met或 Ala;
Xaa3是选自下组的氨基酸: Gly或 Ala;
Xaa4是选自下组的氨基酸: Lys或 Arg;
Xaa5是选自下组的氨基酸: Gly或 Ala;
Xaa6是选自下组的氨基酸: Arg,Lys或 Gly;
Xaa7是选自下组的氨基酸: Ser或 Thr;
Xaa8是选自下组的氨基酸: Tyr或 Phe;
Xaa9是选自下组的氨基酸: Lys或 Arg;
XaalO是选自下组的氨基酸: Gly或 Ala;
Xaall是选自下组的氨基酸: Thr或 Ser;
Xaal2是选自下组的氨基酸: Val , Leu, lie, Met或 Ala;
Xaal3是选自下组的氨基酸: Ser或 Thr;
Xaal4是选自下组的氨基酸: lie , Leu, Val, Met或 Ala;
Xaal5是选自下组的氨基酸: Thr或 Ser;
Xaal6是选自下组的氨基酸: Lys或 Arg;
Xaal7是选自下组的氨基酸: Ser或 Thr;
Xaal8是选自下组的氨基酸: Gly或 Ala;
Xaal9是选自下组的氨基酸: lie , Leu, Val, Met或 Ala;
Xaa20是选自下组的氨基酸: Lys或 Arg;
Xaa21是无, 或 1-3个氨基酸构成肽段;
并且所述的多肽具有抑制血管新生的活性,且所述多肽的长度为 20-26 氨基酸。
2.如权利要求 1所述的多肽, 其特征在于, XaaO和 /或 Xaa21是 1-3个氨 基酸构成的肽段。
3.如权利要求 1所述的多肽,其特征在于, aal是选自下组的氨基酸: l ie 或 Leu;
Xaa2是选自下组的氨基酸: l ie或 Leu
Xaa3是选自下组的氨基酸: Gly或 Ala
Xaa4是选自下组的氨基酸: Lys或 Arg
Xaa5是选自下组的氨基酸: Gly或 Ala
Xaa6是选自下组的氨基酸: Arg或 Lys
Xaa7是选自下组的氨基酸: Ser或 Thr
Xaa8是选自下组的氨基酸: Tyr或 Phe
Xaa9是选自下组的氨基酸: Lys或 Arg
XaalO是选自下组的氨基酸 Gly或 Ala;
Xaal l是选自下组的氨基酸 Thr或 Ser ;
Xaal2是选自下组的氨基酸 Val或 Leu ;
Xaal3是选自下组的氨基酸 Ser或 Thr ;
Xaal4是选自下组的氨基酸 l i e或 Leu ;
Xaal5是选自下组的氨基酸 Thr或 Ser ;
Xaal6是选自下组的氨基酸 Lys或 Arg ;
Xaal7是选自下组的氨基酸 Ser或 Thr ;
Xaal8是选自下组的氨基酸 Gly或 Ala;
Xaal9是选自下组的氨基酸: ll ii ee或或 LLeeuu ;; 和 /或
Xaa20是选自下组的氨基酸 Lys或 Arg。
4.如权利要求 1所述的多肽, 其特征在于, 所述多肽选自下组:
(a)具有 SEQ ID NO : 1所示氨基酸序列的多肽;
(b)将 SEQ ID NO : 1所示氨基酸序列经过 1-5个氨基酸残基的取代、 缺失或 添加而形成的, 且具有抑制血管新生功能的由(a)衍生的多肽。
5. 一种分离的核酸分子, 其特征在于, 它编码权利要求 1所述的多肽。
6. 一种药物组合物, 其特征在于, 它含有:
(a) 权利要求 1所述多肽或其药学上可接受的盐; 和
(b) 药学上可接受的载体或赋形剂。
7.如权利要求 6所述的药物组合物, 其特征在于, 所述组合物的剂型为眼 药水、 针剂、 眼用凝胶或眼药膏。
8. 如权利要求 1 所述的多肽或药学上可接受的盐的用途, 其特征在于, 用于制备用于抑制血管新生或防治与血管新生相关疾病的药物。
9. 如权利要求 8 所述的用途, 其特征在于, 所述的与血管新生相关疾病 的选自下组: 新生血管性眼病、 肿瘤、 缺血性心脏病、 非炎症性心肌病、 冠状 动脉硬化、 闭塞性动脉硬化、 动脉栓塞、 动脉血栓、 Berger ' s病、 慢性炎症、 炎症性肠病、 溃疡、 风湿性关节炎、 硬皮症、 银屑病、 不育症和肉瘤状病。
10.—种抑制哺乳动物血管新生的方法, 其特征在于, 包括步骤: 给需要 的对象施用本发明所述的多肽或其药学上可接受的盐。
PCT/CN2011/076755 2010-07-28 2011-07-01 具有抑制血管生成活性的多肽 WO2012013110A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201010239162.2 2010-07-28
CN201010239162.2A CN102336812B (zh) 2010-07-28 2010-07-28 一种具有抑制血管生成活性的多肽

Publications (1)

Publication Number Publication Date
WO2012013110A1 true WO2012013110A1 (zh) 2012-02-02

Family

ID=45512798

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2011/076755 WO2012013110A1 (zh) 2010-07-28 2011-07-01 具有抑制血管生成活性的多肽

Country Status (2)

Country Link
CN (1) CN102336812B (zh)
WO (1) WO2012013110A1 (zh)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107129521B (zh) * 2016-02-26 2020-11-20 上海市第一人民医院 一种具有透过细胞膜或体组织屏障功能的肽及其应用
CN106220714B (zh) * 2016-09-26 2019-08-09 成都诺恩基因科技有限公司 一种抑制新生血管的多肽、含有该多肽的药物及其应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001044294A2 (en) * 1999-12-15 2001-06-21 Entremed, Inc. Compositions and methods for inhibiting endothelial cell proliferation
US20030148950A1 (en) * 2001-10-09 2003-08-07 Li Xin Kringle domain 1 of human hepatocyte growth factor and uses therefor

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001044294A2 (en) * 1999-12-15 2001-06-21 Entremed, Inc. Compositions and methods for inhibiting endothelial cell proliferation
US20030148950A1 (en) * 2001-10-09 2003-08-07 Li Xin Kringle domain 1 of human hepatocyte growth factor and uses therefor

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KUBA, K. ET AL.: "Kringle 1-4 of hepatocyte growth factor inhibits proliferation and migration of human microvascular endothelial cells.", BIOCHEM BIOPHYS RES COMMUN., vol. 279, no. 3, 29 December 2000 (2000-12-29), pages 846 - 852, XP002294167, DOI: doi:10.1006/bbrc.2000.4034 *
SHEN, Z. ET AL.: "The kringle 1 domain of hepatocyte growth factor has antiangiogenic and antitumor cell effects on hepatocellular carcinoma.", CANCER RES., vol. 68, no. 2, 15 January 2008 (2008-01-15), pages 404 - 414 *

Also Published As

Publication number Publication date
CN102336812B (zh) 2015-12-02
CN102336812A (zh) 2012-02-01

Similar Documents

Publication Publication Date Title
JP5701375B2 (ja) 血管新生促進用ペプチド及びその使用
CN104341486B (zh) 一类新的抑制新生血管的多肽及其应用
RU2426745C2 (ru) Рекомбинантный химерный белок фактора ингибирования нейтрофилов и гиругена и содержащая его фармацевтическая композиция
US9023795B2 (en) Angiogenesis-inhibiting peptide and application thereof
WO2012013110A1 (zh) 具有抑制血管生成活性的多肽
WO2016115732A1 (zh) 一种抑制血管新生或生长的多肽及其应用
JP5752782B2 (ja) 血管新生促進用ペプチド及びその使用
CN104341489B (zh) 一类新的抑制新生血管的多肽及其应用
US20150266926A1 (en) Small molecule polypeptide for preventing and restraining inflammation and application of same
EP3040343B1 (en) Low molecular polypeptide for preventing and treating inflammation and use thereof
US9266933B2 (en) Polypeptides inhibiting neovascularization and uses thereof
WO2012000409A1 (zh) 具有抑制新生血管活性的肽及其应用
CN104004066B (zh) 预防或抑制炎症反应和血管新生的小分子多肽及其应用
CN101503458B (zh) 预防和治疗血管新生的小分子多肽及其应用
WO2013060020A1 (zh) 一类新的抑制新生血管的小肽及其应用
CN104004057B (zh) 一类抑制新生血管的小肽及其应用
CN103992376B (zh) 一种抑制新生血管的小肽及其应用
US8933031B2 (en) Polypeptide inhibiting angiogenesis and application thereof
US20220168434A1 (en) Application of transthyretin in entering eye and preparing drop
CN110092816B (zh) 预防和治疗纤维化的小分子多肽及其应用
CN103992377B (zh) 一种抑制新生血管的小肽及其应用
JPH06219963A (ja) regタンパク質を有効成分とする薬剤

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: 11811811

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: 11811811

Country of ref document: EP

Kind code of ref document: A1