WO2018032639A1 - Activated human blood-clotting factor vii fusion protein, and manufacturing method and application of same - Google Patents

Activated human blood-clotting factor vii fusion protein, and manufacturing method and application of same Download PDF

Info

Publication number
WO2018032639A1
WO2018032639A1 PCT/CN2016/106012 CN2016106012W WO2018032639A1 WO 2018032639 A1 WO2018032639 A1 WO 2018032639A1 CN 2016106012 W CN2016106012 W CN 2016106012W WO 2018032639 A1 WO2018032639 A1 WO 2018032639A1
Authority
WO
WIPO (PCT)
Prior art keywords
fusion protein
human
chromatography
fviia
activity
Prior art date
Application number
PCT/CN2016/106012
Other languages
French (fr)
Chinese (zh)
Inventor
李强
朱文臣
高永娟
朱成功
李媛丽
王晓山
孙乃超
刘宾
王文文
李智
刘婷婷
朱鹿燕
李亦清
任子甲
朱松林
肖春峰
苏鸿声
Original Assignee
安源医药科技(上海)有限公司
辅仁药业集团有限公司
旭华(上海)生物研发中心有限公司
开封制药(集团)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 安源医药科技(上海)有限公司, 辅仁药业集团有限公司, 旭华(上海)生物研发中心有限公司, 开封制药(集团)有限公司 filed Critical 安源医药科技(上海)有限公司
Publication of WO2018032639A1 publication Critical patent/WO2018032639A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • C12N9/6437Coagulation factor VIIa (3.4.21.21)
    • 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
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21021Coagulation factor VIIa (3.4.21.21)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin

Definitions

  • the present invention relates to a fusion protein, and more particularly to an activated human Factor VII (FVIIa) fusion protein, a preparation method and use thereof, and particularly to the use for the preparation of a medicament for treating various blood coagulation related diseases.
  • FVIIa activated human Factor VII
  • FVII is a vitamin K-dependent plasma glycoprotein that is synthesized in the liver and secreted into the blood as a single-chain proteoprotein with a molecular weight of approximately 53 kDa (Broze et al, J Biol Chem, 1980, 255: 1242-1247) .
  • the FVII zymogen is hydrolyzed by a protease at a single site, Arg152-Ile153, to produce a double-strand linked by a disulfide bond, thereby converting to its active form, FVIIa.
  • FVIIa comprising NH 2 - terminal derived light chain (about 20 KDa) and COOH- terminal-derived heavy chain (about 30KDa) connected via a single disulfide bond (Cys135 to Cys262).
  • the light chain contains a cell membrane that binds to the Gla domain and two "epidermal growth factor (EGF) domains", while the heavy chain contains a serine protease catalytic domain.
  • EGF epidermal growth factor
  • FVIIa acts as a serine protease and participates in the extrinsic pathway of the coagulation cascade.
  • the use of FVIIa in the treatment of hemophilia is based on the low affinity binding of FVIIa to thrombin-activated platelet surface.
  • FVIIa By administering a pharmacological dose of exogenous FVIIa, platelet surface thrombin production at the lesion is enhanced, which is associated with FVIII. /FIX is irrelevant, that is, to bypass the demand for FVIIIa and FIXa to stop bleeding. Therefore, FVIIa can be used for the prevention and treatment of hemorrhagic conditions in hemophilia patients who produce inhibitors.
  • Commercially available recombinant factor VIIa is currently only available (Novo Nordisk, Denmark). This drug has been approved worldwide for the treatment of hemophilia A or B patients who develop FVIII or FIX antibodies (inhibitors), patients with congenital FVII deficiency, and termination of bleeding events associated with trauma and/or surgery or Prevent bleeding.
  • therapeutic coagulation protein drugs including FVIIa
  • FVIIa therapeutic coagulation protein drugs
  • proteolytic enzymes are rapidly degraded by proteolytic enzymes and are easily neutralized by antibodies, which results in a significant decrease in their circulating half-life in vivo, thereby limiting their efficacy.
  • Recombinant FVIIa The circulating half-life in the human body is about 2.3 hours, and it needs to be injected every 2-3 hours.
  • rFVIIa-FP albumin fusion with albumin and FVIIa
  • CSL Behring has completed a clinical phase I study, which is serum half in normal subjects.
  • the decay is 3-4 times higher than that of commercially available FVIIa (Golor G et al, J Thromb Haemost, 2013, 11(11): 1977-85), with a biological activity of 620-770 IU/mg, equivalent to 69-75 IU. /nM (Weimer T et al, Thromb Haemost, 2008, 99: 659-667).
  • the PEGylated liposome formulation of FVIIa is also in the early clinical stage and has a serum half-life of only twice that of native FVIIa.
  • the N-glycosyl-directed PEGylation modification of FVIIa (N7-GP) developed by Novo Nordisk Company was approximately 5-fold longer than that of rFVIIa, but due to the lack of quantitative-effect linearity in clinical phase II data and the occurrence of subjects The allergic reaction, the project has terminated research (Ljung R et al, J Thromb Haemost, 2013, 11 (7): 1260-8).
  • the rFVIIaFc monomer-dimer hybrid (momer/dimer hybrid) fusion protein developed by Biogen is still in the early stages of research, and the study showed that the terminal half-life of Monomeric rFVIIaFc in hemophilia A mice was 6.6 hours.
  • the blood coagulation activity of the mouse returned to 40% of the normal value.
  • its activity was abruptly attenuated at the initial stage, and decreased to about 20% at 30 minutes after administration, and 1 hour after administration. Has dropped to about 10%, which is related to The downward trend is similar in 0 to 1 hour.
  • rFVIIaFc can effectively control acute bleeding symptoms in a short period of time and does not maintain the normal coagulation function of the body for a long time, smoothly and effectively.
  • the carboxy-terminal peptide of the human chorionic gonadotropin (hCG) ⁇ -chain also has the effect of prolonging the half-life of certain proteins in vivo, and thus some of the extended half-life fractions contained in the fusion proteins disclosed in some patent documents can be selected.
  • the rFVIIa-CTP fusion protein developed by OPKO/Prolor, rFVIIa in series with 1 to 5 CTP molecules, respectively, significantly increases the half-life of FVII in a manner proportional to CTP, when adding 3, 4 or 5 CTP to make FVII half-life, respectively.
  • CTP can also be used as a linker sequence to link different subunits of the same protein. For example, among the fusion proteins disclosed in Chinese Patent Nos.
  • CTP acts as a linker between the beta subunit and the alpha subunit of follicle stimulating hormone; among the fusion proteins disclosed in WO2005058953A2, CTP is used as a linker for Linking the beta and alpha subunits of the glycoprotein hormone.
  • the present inventors have found that the long-acting rFVIIa disclosed in the prior art has a problem of limited half-life extension, significant decrease in biological activity or strong immunogenicity, mainly because the C-terminus of FVIIa contains a serine protease domain (153-406 residues). Therefore, the fusion of other molecules at the C-terminus affects the catalytic activity and function of FVIIa.
  • the inventors have found that between FVIIa and an extended half-life portion (eg, an immunoglobulin Fc fragment), only a long stretch of conventional flexible peptide linkers (eg, (GGGGS)n) ligation, but instead the fusion protein pair Proteases are more sensitive, and even worse, such long peptide linkers also allow most of the fusion proteins to be expressed in polymer form with little biological activity.
  • an extended half-life portion eg, an immunoglobulin Fc fragment
  • a linker molecule composed of a flexible peptide and CTP is linked to FVIIa and an extended half-life portion (eg, human IgG Fc), thereby maximally retaining the biological activity of FVIIa, and More significantly extend its in vivo half-life of activity.
  • the invention also provides a preparation method and use of the fusion protein.
  • the invention provides an activated human Factor VII fusion protein (hereinafter referred to as a fusion protein) comprising, in order from the N-terminus to the C-terminus, an activated human Factor VII (FVIIa), a flexible peptide linker (The carboxy terminal peptide rigid unit (hereinafter referred to as (CTP) n ), which is L), at least one human chorionic gonadotropin ⁇ subunit, preferably n, 1, 2, 3, 4, Or 5) and an extended half-life portion (eg, immunoglobulin Fc fragment, albumin, transferrin or PEG, preferably a human IgG Fc variant (denoted as vFc)).
  • the fusion protein is represented by FVIIa-L-CTP n -vFc.
  • the amino acid sequence of human factor VII is at least 80% homologous to native human factor VII; more preferably, the amino acid sequence of human factor VII is at least 90% homologous to native human factor VII; most preferred
  • the human Factor VII comprises the amino acid sequence set forth in SEQ ID NO: 1.
  • FVIIa also includes human coagulation factor VII (abbreviated as FVII) which is activated in the applicator or which is activated as FVIIa before application.
  • FVII human coagulation factor VII
  • the flexible peptide linker is preferably non-immunogenic and produces a sufficient distance between FVII and Fc to minimize steric effects between each other.
  • a flexible peptide linker comprising two or more amino acid residues is used and is selected from the group consisting of Gly (G), Ser (S), Ala (A) and Thr (T).
  • the flexible peptide linker comprises G and S residues.
  • the length of the linker peptide is very important for the activity of the fusion protein.
  • the peptide linker may preferably comprise an amino acid sequence formula formed by combining (GS) a (GGS) b (GGGS) c (GGGGS) d cycle units, wherein a, b, c and d are greater than Or an integer equal to 0, and a+b+c+d ⁇ 1.
  • the peptide linker may preferably comprise the following sequence:
  • L2 GSGGGGSGGGGSGGGGSGGGGSGGGGS (as shown in SEQ ID NO: 3);
  • L4 GSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGS (as shown in SEQ ID NO: 5);
  • the CTP rigid unit is selected from the full length sequence consisting of amino acids 113 to 145 of the carboxy terminus of human chorionic gonadotropin ⁇ subunit or a fragment thereof, in particular, the rigid unit comprises SEQ ID NO: 7 The amino acid sequence shown or its truncated sequence.
  • the CTP rigid unit comprises at least 2 glycosylation sites; for example, in a preferred embodiment of the invention, the CTP rigid unit comprises 2 glycosylation sites, exemplarily, the CTP The rigid unit comprises 10 amino acids of the N-terminus of SEQ ID NO: 7, ie SSSS*KAPPPS*, or the CTP rigid unit comprises 14 amino acids of the SEQ ID NO: 7C terminus, ie S*RLPGPS*DTPILPQ; as another example
  • the CTP rigid unit comprises three glycosylation sites, exemplarily, said The CTP rigid unit comprises 16 amino acids of the N-terminus of SEQ ID NO: 7, ie SSSS*KAPPPS*LPSPS*R; as in other embodiments, the CTP rigid unit comprises 4 glycosylation sites, exemplarily,
  • the CTP rigid unit comprises 28, 29, 30, 31, 32 or 33 amino acids and begins at position 113, 114, 115, 116, 117 or 118 of the human chorionic gonadotropin beta subunit, terminat
  • the CTP rigid unit comprises 28 amino acids of the N-terminus of SEQ ID NO: 7, namely SSSS*KAPPPS*LPSPS*RLPGPS*DTPILPQ.
  • * represents a glycosylation site.
  • the CTP rigid units provided herein are at least 70% homologous to the native CTP amino acid sequence; in other embodiments, the CTP rigid units provided herein are at least 80% homologous to the native CTP amino acid sequence; In other embodiments, the CTP rigid units provided herein are at least 90% homologous to the native CTP amino acid sequence; in other embodiments, the CTP rigid units provided herein are at least 95% homologous to the native CTP amino acid sequence.
  • the CTP rigid unit may preferably comprise the following sequence:
  • CTP 1 PRFQDSSSSKAPPPSLPSPSRLPGPSDTPILPQ (as shown in SEQ ID NO: 7);
  • CTP 2 SSSSKAPPPSLPSPSRLPGPSDTPILPQ (as shown in SEQ ID NO: 8);
  • CTP 4 SRLPGPSDTPILPQ (shown as SEQ ID NO: 10).
  • the fusion protein comprises one of the above CTP rigid units.
  • the fusion protein comprises more than one of the above CTP rigid units, preferably comprising 2, 3, 4 or 5 of the above CTP rigid units, as in an embodiment of the invention,
  • the fusion protein comprises two CTP 3 rigid units: SSSSKAPPPSSSSSKAPPPS (CTP 3 -CTP 3 , or expressed as (CTP 3 ) 2 ).
  • the extended half-life portion is preferably a self-immunoglobulin IgG, IgM, IgA Fc fragment; more preferably an Fc fragment from human IgG1, IgG2, IgG3 or IgG4 and variants thereof; further, the human IgG Fc variant comprises in the wild At least one amino acid modification in a human IgG Fc, and the variant has reduced effector function (ADCC and/or CDC effect) and/or enhanced binding affinity to the neonatal receptor FcRn. Further, the human IgG Fc variant may be selected from the group consisting of:
  • vFc ⁇ 1 human IgG1 hinge region, CH2 and CH3 region containing the Leu234Val, Leu235Ala and Pro331Ser mutations (such as the amino acid sequence shown in SEQ ID NO: 11);
  • vFc ⁇ 2-1 human IgG2 hinge region, CH2 and CH3 region containing the Pro331Ser mutation (such as the amino acid sequence shown in SEQ ID NO: 12);
  • vFc ⁇ 2-2 human IgG2 hinge region, CH2 and CH3 region containing the Thr250Gln and Met428Leu mutations (such as the amino acid sequence shown in SEQ ID NO: 13);
  • vFc ⁇ 2-3 human IgG2 hinge region, CH2 and CH3 regions (such as the amino acid sequence shown in SEQ ID NO: 14) containing the Pro331Ser, Thr250Gln and Met428Leu mutations.
  • (v) vFc ⁇ 4 human IgG4 hinge region, CH2 and CH3 regions containing the Ser228Pro and Leu235Ala mutations (such as the amino acid sequence shown in SEQ ID NO: 15).
  • the IgG Fc variants provided by the present invention include, but are not limited to, the five variants described in (i) to (v), and may also be a combination or superposition of two types of functional variant mutation sites between IgG isotypes.
  • the variant as described in (iv) above is a novel variant of the novel IgG2Fc obtained by superimposing the mutation sites in (ii) and (iii).
  • An Fc variant (vFc) in a fusion protein of the invention which comprises a hinge region, a CH2 and a CH3 region of human IgG such as human IgG1, IgG2 and IgG4.
  • This CH2 region contains amino acid mutations at positions 228, 234, 235 and 331 (as determined by the EU counting system). It is believed that these amino acid mutations reduce the effector function of Fc.
  • Human IgG2 does not bind to FcyR but shows very weak complement activity.
  • An Fc[gamma]2 variant with a Pro331Ser mutation should have a lower complement activity than native Fc[gamma]2 and is still an Fc[gamma]R non-binding element.
  • IgG4Fc is defective in the activation of the complement cascade and its binding affinity to Fc ⁇ R is about an order of magnitude lower than that of IgG1.
  • An Fc ⁇ 4 variant with a Leu235Ala mutation should exhibit minimal effector function compared to native Fc ⁇ 4.
  • Fc ⁇ 1 with Leu234Va1, Leu235Ala and Pro331Ser mutations also showed reduced effector function compared to native Fc ⁇ 1.
  • These Fc variants are all more suitable for the preparation of FVIIa fusion proteins than native human IgG Fc.
  • the 250 and 428 positions contain amino acid mutations that increase the binding affinity of the Fc region to the neonatal receptor FcRn, thereby further extending the half-life (Paul R et al, J Biol Chem, 2004, 279: 6213). -6216); the above two types of functional variants are combined or superimposed to obtain a new combined variant, which reduces the effector function and prolongs its circulating half-life in vivo.
  • the Fc variants of the invention comprise, but are not limited to, mutations at several of the above sites, and substitutions at other sites may be introduced such that the Fc has reduced effector function and/or enhanced binding to the FcRn receptor, while still It does not cause a decrease in Fc variant function/activity or cause a poor conformational change.
  • substitutions at other sites may be introduced such that the Fc has reduced effector function and/or enhanced binding to the FcRn receptor, while still It does not cause a decrease in Fc variant function/activity or cause a poor conformational change.
  • amino acid sequence of the fusion protein is as shown in SEQ ID NO:
  • a DNA encoding the above fusion protein is provided.
  • the DNA sequence of the fusion protein is set forth in SEQ ID NO: 17.
  • a vector comprising the above DNA is provided.
  • a host cell comprising the above vector or transfected with the above vector is provided.
  • the host cell is a derivative cell line DXB-11 of CHO.
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier, excipient or diluent, and an effective amount of the above fusion protein.
  • a method of making or producing the fusion protein from a cell line derived from a mammalian cell line, such as CHO comprising the steps of:
  • step (c) culturing step (b) the selected cell strain to express the fusion protein
  • step (d) harvesting the fermentation broth obtained in step (c), purifying the fusion protein
  • step (e) activating the purified fusion protein in step (d).
  • the CHO-derived cell line in the step (a) is DXB-11.
  • the fusion protein is purified by four-step chromatography, which is ProteinA affinity chromatography, multi-dimensional mode chromatography, anion exchange chromatography and molecular sieve chromatography.
  • Step Protein A affinity chromatography step preferably using a commercially available, for example, but not limited to GE's Mabselect TM, Mabselect SuRe TM, or TOSOH the Toyopearl AF-rProteinA-650F TM, or the rProtein A Bestarose TM Boge Long , or the world of human and rProtein A Bead TM , or Safran Technology's MabPurix TM chromatography medium. It utilizes the specific binding of the Fc fragment of the fusion protein (human IgG) to the monoclonal antibody for capture because the proteinA ligand on the affinity medium has a high affinity for the Fc fragment and can specifically bind reversibly with it.
  • a commercially available for example, but not limited to GE's Mabselect TM, Mabselect SuRe TM, or TOSOH the Toyopearl AF-rProteinA-650F TM, or the rProtein
  • affinity chromatography can rapidly and efficiently capture and purify the fusion protein from the fermentation broth.
  • the multi-dimensional mode chromatography step of the second step preferably using commercially available CHT (TM) (ceramic hydroxyapatite Form I or Type II) chromatography media (40 [mu]m or 80 [mu]m), such as but not limited to Bio-Rad Corporation.
  • CHT CHT
  • TM ceramic hydroxyapatite Form I or Type II chromatography media
  • the CHT medium is arranged in a repeating geometric pattern of five calcium doublets and a pair of hydroxyl-containing phosphorus triplets, wherein the PO4 3- ions are ionically bonded to the positively charged protein and have ion exchange characteristics.
  • the paired negative charge of the Ca 2+ ion and the free carboxyl group of the fusion protein is bound by metal chelation, and the binding mode is insensitive to NaCl and can be competitively eluted by a sodium phosphate concentration gradient.
  • the degree of carboxylation of FVIIa is correlated with its biological activity (Hagen FS et al, Proc Natl Acad Sci USA, 1986, 83(8): 2412-2416; US Pat. No. 8,032,224; US Patent No. US 2009/0042784). Therefore, the fusion protein captured by affinity chromatography in the present invention is subjected to a second purification step using CHT to separate a target component having a higher biological activity.
  • the third step of anion exchange chromatography step preferably using a commercially available, for example, but not limited to GE's Q Sepharose HP TM, Q Sepharose FF TM chromatography medium, or the Boge Long Q Bestarose HP TM, Q Bestarose FF TM layer Analysis of the medium, and the earth or the Q Beads 6HP TM, Q Beads 6FF TM chromatographic medium is isolated and purified, for removing contaminants.
  • the separation of molecules by ion exchange is based on the difference in their net surface charge.
  • the charge properties of molecules vary widely, and the difference in total charge, charge density, and surface charge distribution of the molecules causes them to differ in their ability to bind to the charge-exchanged ion-exchange chromatography packing.
  • the fusion protein can be separated from the contaminants by progressively increasing the concentration of NaCl in the buffer for competitive elution.
  • the molecular sieve chromatography step of the fourth step is preferably carried out using a commercially available Superdex 200 (TM) such as, but not limited to, GE, or a Chromdex 200 prep grade (TM) chromatography medium from Boglon.
  • TM Superdex 200
  • TM Chromdex 200 prep grade
  • Molecular sieve chromatography is mainly a chromatographic method for separating according to the size of a protein molecule.
  • Molecular sieve media are porous spherical particles.
  • the retention time in the column is relatively short and elutes first; while the small molecules can enter the pores of the medium, and the path passing through the column is long, and then elutes.
  • the fusion protein obtained by the above three-step chromatography in the present invention still has a certain polymer component, which may affect the activation process and the biological activity after activation, and therefore the molecular sieve layer is used to remove the polymer component therein.
  • the activating step in the step (e) may activate the fusion protein by coagulation factor XII (FXII) activation, on-column self-activation or solution incubation self-activation.
  • FXII coagulation factor XII
  • the conversion of FVII to active FVIIa needs to be done under the action of FXII (Hedner et al, J Clin Invest, 1983, 71: 1836-1841), or other proteases with trypsin-like activity can also activate FVII (Kisiel et al, Behring Inst Mitt) , 1983, 73: 29-42); FVII may also be activated by FVII itself, but by its serine protease region, by itself, without the use of other proteases.
  • FVII can also be self-activated by binding to a positively charged surface or filler, such as an anion exchange packing (Pedersen AH et al, Biochemistry, 1989, 28: 9331-9336). Increasing the ionic strength, lowering the pH, or increasing the concentration of Ca 2+ in the solution can dissociate the activated FVIIa from the positively charged surface or onto the filler (Bioern et al, Research Disclosures, 1986, 269: 564-56). Self-activation under solution conditions is described in US 2007/0129298.
  • the fusion protein is activated by a solution incubation self-activation method, and the activity of the activated fusion protein is >15000 IU/mg.
  • the fusion protein is provided for use in the preparation of a medicament for treating or preventing a bleeding disorder.
  • the fusion protein is used for the prevention or treatment of hemorrhagic diseases in patients with FVII congenital or acquired deficiency, prevention or treatment of spontaneous or surgical bleeding in patients with hemophilia A or B, or other related Application in bleeding drugs.
  • the FVIIa fusion protein constructed by the present invention, wherein the Fc fragment is non-lytic, that is, by mutating the complement and receptor binding domains of the Fc fragment, regulating the binding affinity of the Fc to the corresponding receptor, and reducing or eliminating the ADCC and CDC effects. While retaining only the Fc segment to prolong the half-life of the active protein in vivo, it does not produce cytotoxicity.
  • the FVIIa fusion protein developed by Biogen whose Fc segment is of natural origin, predicts that Fc-mediated adverse effector functions will necessarily increase the patient's therapeutic risk.
  • the half-life of FVIIa fusion protein was about 3 times longer in vivo.
  • the plasma coagulation activity was about 40% after 3 hours of single injection of FP-A, while the activity of the same active group was reduced to 3% after 3 hours;
  • FP- The plasma clotting activity remained above 7% after 12 hours of A administration, and the monomer-dimer heterozygous (Monomeric) rFVIIaFc developed by Biogen (see J. Salsa et al., Thrombosis Research, 2015, 135: 970-976).
  • Biogen see J. Salsa et al., Thrombosis Research, 2015, 135: 970-976.
  • the fluctuation of drug concentration in serum is reduced, and the safety is improved, which can reduce the frequency of injection and improve the quality of life of patients;
  • the fusion protein constructed by the invention is more efficient and convenient than the Monomeric FVII fusion protein constructed by Biogen, and the production cost can be greatly reduced.
  • Biogen Corporation constructed a dual expression vector for rFVIIIFc and Fc, wherein the Fc molecule was labeled with Flag (European Patent, Publication No. EP1624891B1).
  • the fusion protein fermentation broth expressed by it is expected to contain three forms of products, namely FVII-Fc: FVII-Fc homodimeric (Dimeric) fusion protein, FVII-Fc: FLAG-Fc monomer-dimerization Monomeric fusion protein and FLAG-Fc: FLAG-Fc dimer three products.
  • the host cell in the process of expression of the fusion protein, needs to simultaneously express two single-stranded molecules of FVII-Fc and Fc, and then separately polymerize to form the above three products, thereby greatly reducing the expression efficiency of the final target product;
  • the other two forms of impurities must be removed during the purification process, which makes the purification process more complicated, the production efficiency is low, and the production cost is also greatly increased. Therefore, the preparation method of the invention has certain technical advantages and price advantages over the Monomeric rFVIIFc fusion protein developed by Biogen, and the expression and purification processes are simpler, more efficient, and the production cost is lower;
  • each batch of purified fusion protein can reach at least 15000 IU/mg, about 2949 IU/nM (2 FVIIa per fusion protein, equivalent to 1474.4 IU/nM FVIIa), and some batches of fusion protein.
  • the activity is even higher than 22470 IU/mg, which is converted to a molar activity of about 4325 IU/nM (2 FVIIa per fusion protein, equivalent to 2162 IU/nM FVIIa), and the listed recombinant FVIIa-Noci (2511 IU/nM)
  • the activity of FVIIa) is comparable, indicating that the fusion protein provided by the present invention has a C-terminally-fused Fc having minimal effect on the activity of FVIIa;
  • a CTP rigid unit constituting the fusion protein which contains a plurality of O-glycosyl side chains, which can form a relatively stable stereo conformation and can effectively isolate FVIIa from Fc.
  • the CTP rigid unit contains a glycosyl group, and the negatively charged, highly sialylated CTP rigid unit is able to resist the clearance of the kidney and further prolong the half-life of the fusion protein;
  • the protective effect of the CTP rigid unit glycosyl side chain can be In order to reduce the sensitivity of the linker peptide to protease, the fusion protein is not easily degraded in the junction region;
  • the method for preparing the fusion protein provided by the invention has the advantages of high yield.
  • the culture is continuously cultured for 14 days in a 300 ml shake flask, and the cumulative yield reaches 310 mg/L, which can be subjected to process enlargement to realize large-scale industrial production.
  • the length of the linker peptide is very important for the activity of the fusion protein.
  • the FVII/FVIIa-FP fusion protein containing a linker peptide shows that the increase in its biological activity depends on the length of the linker peptide, which may be explained by the increased linker peptide between the two parts of the fusion protein, allowing the two parts of the molecule to exercise their respective The function is conducive to the formation of a higher specific activity conformation.
  • the present inventors have previously designed three different lengths of flexible linker peptides composed of glycine and serine, and FVII constitutes a fusion protein (without CTP rigid unit) through the Fc junction of the linker peptide and its C-terminal fusion, and transient expression experiments show that The two amino acid short peptide linker GlySer-linked fusion protein showed little activity, indicating that the important functional regions that maintain the biological activity of FVIIa are greatly affected by the C-terminal fusion ligand in three-dimensional structure. When the linker peptide was increased to 16 amino acids, the biological activity of the FVIIa fusion protein was significantly increased, but still much lower than recombinant FVIIa.
  • the linker peptide When the linker peptide is further extended to 37 amino acids, the fusion protein portion secreted by the CHO cell is polymerized and has low activity. This indicates that the problem of the effect of the Fc fragment on FVIIa activity cannot be completely solved by simply extending the length of the linker peptide.
  • CTP is a short peptide derived from the carboxy terminus of the ⁇ -subunit of human chorionic gonadotropin (hCG).
  • reproductive-related peptide hormones FSH
  • FSH follicle stimulating hormone
  • LH luteinizing hormone
  • TSH thyrotropin
  • hCG chorionic gonadotropin
  • the natural CTP contains 37 amino acid residues with four O-glycosylation sites and the terminal is sialic acid. Residues. Negatively charged, highly sialylated CTP is resistant to the clearance of the kidneys, thereby prolonging the half-life of the protein in the body.
  • the inventors creatively linked at least one CTP polypeptide to a flexible linker of appropriate length, collectively as a linker peptide, for ligation of FVII with an extended half-life moiety (eg, an immunoglobulin Fc fragment).
  • the present inventors have found that the C-terminal catalytic domain of FVIIa is critical for its function, and the FVIIa spatial conformation is complex and fragile, and the steric hindrance effect of the fusion ligand is highly susceptible to interference with its correct folding.
  • an equivalent of a rigid linker peptide is added.
  • This aspect ensures that the N-terminally fused FVIIa does not affect the binding site of the Fc variant to FcRn, thereby affecting the half-life; in addition, the Fc-ProteinA binding site is important for the purification step in the preparation process, and the CTP rigid unit is connected to ensure N The end-fused FVIIa also does not "cover" its binding site to protein A. On the other hand, the addition of a CTP rigid unit also allows the Fc fragment of about 25 kD size to not interfere with the correct folding of the N-terminally fused FVIIa, resulting in a decrease or loss of its biological activity/function.
  • the Fc element is derived from the constant region Fc fragment of immunoglobulin IgG, which plays an important role in eradicating the immune defense of pathogens.
  • the effector function of Fc-mediated IgG is exerted through two mechanisms: (1) binding to cell surface Fc receptors (Fc ⁇ Rs), digestion of pathogens by phagocytosis or cleavage or killer cells via antibody-dependent cellular cytotoxicity (ADCC) pathway , or (2) binding to C1q of the first complement component C1, eliciting a complement-dependent cytotoxicity (CDC) pathway, thereby lysing the pathogen.
  • Fc ⁇ Rs cell surface Fc receptors
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxicity
  • IgG1 and IgG3 efficiently bind to Fc ⁇ Rs, and the binding affinity of IgG4 to Fc ⁇ Rs is low, and the binding of IgG2 to Fc ⁇ Rs is too low to be determined, so human IgG2 has almost no ADCC effect.
  • human IgG1 And IgG3 can also effectively bind to C1q to activate the complement cascade.
  • Human IgG2 binds relatively weakly to C1q, whereas IgG4 does not bind to C1q (Jefferis R et al, Immunol Rev, 1998, 163: 59-76), so the human IgG2 CDC effect is also weak.
  • Fc variant with enhanced binding affinity to neonatal receptor (FcRn)
  • the plasma half-life of IgG depends on its binding to FcRn, which typically binds at pH 6.0 and dissociates at pH 7.4 (plasma pH). By studying the binding sites of the two, the site of binding to FcRn on IgG was engineered to increase the binding ability at pH 6.0. Mutations in some residues of the human Fc ⁇ domain important for binding to FcRn have been shown to increase serum half-life. Mutations in T250, M252, S254, T256, V308, E380, M428 and N434 have been reported to increase or decrease FcRn binding affinity (Roopenian et al, Nat. Rview Immunology 7: 715-725, 2007). Korean Patent No.
  • KR 10-1027427 discloses variants of trastuzumab (Herceptin, Genentech) having increased FcRn binding affinity, and these variants are selected from the group consisting of 257C, 257M, 257L, 257N, 257Y, 279Q, One or more amino acid modifications of 279Y, 308F and 308Y.
  • Korean Patent Publication No. KR 2010-0099179 provides variants of bevacizumab (Avastin, Genentech) and these variants show increased in vivo by amino acid modifications contained in N434S, M252Y/M428L, M252Y/N434S and M428L/N434S half life.
  • the fusion protein gene of the present invention is codon-optimized and prepared by a synthetic method.
  • nucleotide sequence of the present invention those skilled in the art can conveniently prepare the nucleic acid of the present invention by various known methods. These methods are not limited to synthetic or traditional subcloning, and the specific method can be found in J. Sambrook, Molecular Cloning Experiment Guide.
  • the nucleic acid sequence of the present invention is constructed by subcloning a nucleotide sequence and then subcloning.
  • the invention also provides an expression vector for a mammalian cell comprising a fusion protein sequence encoding the invention and an expression control sequence operably linked thereto.
  • operably linked or “operably linked” is meant a condition in which portions of a linear DNA sequence are capable of modulating or controlling the activity of other portions of the same linear DNA sequence. For example, if a promoter controls the transcription of a sequence, then it is operably linked to the coding sequence.
  • the mammalian cell expression vector can be commercially available, for example, but not limited to, pcDNA3, pIRES, pDR, pBK, pSPORT, etc., which can be used for expression in eukaryotic cell systems.
  • pcDNA3, pIRES, pDR, pBK, pSPORT, etc. can be used for expression in eukaryotic cell systems.
  • One skilled in the art can also select a suitable expression vector based on the host cell.
  • the skilled person can prepare the present invention by inserting the coding sequence of the fusion protein of the present invention into a suitable restriction site by restriction enzyme cleavage and splicing according to a conventional method. Recombinant expression vector.
  • the invention also provides a host cell expressing a fusion protein of the invention comprising a coding sequence for a fusion protein of the invention.
  • the host cell is preferably a eukaryotic cell such as, but not limited to, a CHO cell, a COS cell, a 293 cell, an RSF cell, and the like.
  • the cell is a CHO cell which can preferably express the fusion protein of the present invention, and a fusion protein having good activity and good stability can be obtained.
  • the invention also provides a method for preparing a fusion protein of the invention by recombinant DNA technology, the steps of which comprise:
  • Introduction of the coding sequence into a host cell can employ a variety of known techniques in the art such as, but not limited to, calcium phosphate precipitation, lipofection, electroporation, microinjection, viral infection, alkali metal ion methods.
  • the fusion protein can be activated by coagulation factor XII (FXII) activation, on-column autoactivation or solution incubation self-activation.
  • FXII coagulation factor XII
  • the fusion protein obtained as described above can be purified to a substantially uniform property, such as a single band on SDS-PAGE electrophoresis.
  • the supernatant is first concentrated, and the concentrate can be further purified by gel chromatography or by ion exchange chromatography.
  • ion exchange chromatography For example, anion exchange chromatography or cation exchange chromatography.
  • the gel matrix may be a medium commonly used for protein purification such as agarose, dextran, polyamide, and the like.
  • the Q- or SP- group is a preferred ion exchange group.
  • the purified product may be further purified by hydroxyapatite adsorption chromatography, metal chelate chromatography, hydrophobic interaction chromatography and reversed-phase high performance liquid chromatography.
  • the expressed fusion protein can also be purified using an affinity chromatography column containing a specific antibody, receptor or ligand of the fusion protein.
  • affinity column containing a specific antibody, receptor or ligand of the fusion protein.
  • the fusion polypeptide bound to the affinity column can be eluted using conventional methods such as high salt buffer, pH change, and the like.
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount (160-360 [mu]g/kg) of a fusion protein of the invention, and a pharmaceutically acceptable carrier.
  • an effective amount of a fusion protein of the invention can be formulated in a non-toxic, inert, and pharmaceutically acceptable aqueous carrier medium wherein the pH is usually from about 5 to about 8, preferably, the pH is from about 6 to about 8.
  • the term "effective amount” or “effective amount” refers to an amount that is functional or active to a human and/or animal and that is acceptable to humans and/or animals.
  • a “pharmaceutically acceptable” ingredient is one which is suitable for use in humans and/or mammals without excessive adverse side effects (such as toxicity, irritation, and allergies), i.e., materials having a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier refers to a carrier for the administration of a therapeutic agent, which comprises various excipients and diluents.
  • Pharmaceutically acceptable carriers include, but are not limited to, saline, buffer, dextrose, water, glycerol, ethanol, and combinations thereof.
  • the pharmaceutical preparation should be matched to the mode of administration, and the pharmaceutical composition of the present invention can be prepared into an injection form, for example, by a conventional method using physiological saline or an aqueous solution containing glucose and other adjuvants.
  • the pharmaceutical composition is preferably manufactured under sterile conditions.
  • the amount of active ingredient administered is a therapeutically effective amount.
  • the pharmaceutical preparation of the present invention can also be formulated into a sustained release preparation.
  • the effective amount of the fusion protein of the present invention may vary depending on the mode of administration and the severity of the disease to be treated and the like. The selection of a preferred effective amount can be determined by one of ordinary skill in the art based on various factors (e.g., by clinical trials). The factors include, but are not limited to, the pharmacokinetic parameters of the fusion protein such as bioavailability, metabolism, half-life, etc.; the severity of the disease to be treated by the patient, the patient's weight, the patient's immune status, the route of administration, etc. .
  • Figure 1 Nucleotide sequence and deduced amino acid sequence of FP-A fragment of SpeI-EcoRI (labeled with underlined__) in the PCDNA3 expression vector according to an embodiment of the present invention.
  • Human FVII consists of a signal peptide (1-38, underlined __) and a mature FVII protein (39-444).
  • Mature fusion proteins contain hFVII (39-444), flexible peptide linkers (445-471, underlined __), CTP rigid units (472-499, underlined __) and Fc variants (500-722) .
  • FIG. 11 Comparison of the duration of bleeding after administration of FP-A and Nobel for 1 h and 2 h in HemA mice. Note: compared with HA-N-1h group, * P ⁇ 0.05, *** P ⁇ 0.01; compared with C57-NS group, # P ⁇ 0.05, ### P ⁇ 0.01.
  • the gene sequence encoding the FVII leader peptide, mature protein, flexible peptide linker, CTP rigid unit and human IgG vFc variant is a manually optimized CHO cell preferred codon obtained by artificial synthesis.
  • the full-length DNA fragment of the synthetic fusion protein has a restriction endonuclease site at the 5' and 3' ends, respectively, SpeI and EcoRI, and the full-length DNA fragment is inserted into the corresponding cleavage site of the pUC57 transfer vector, and DNA sequencing verification sequence.
  • the PTY1A1 plasmid includes, but is not limited to, the following important expression components: 1) human cytomegalovirus early promoter and mammalian cells are required for exogenous high expression; 2) dual screening markers with kanamycin resistance in bacteria sexuality, G418 resistance in mammalian cells; 3) Murine dihydrofolate reductase (DHFR) gene expression cassette, when the host cell is DHFR gene-deficient, methotrexate (MTX) can amplify the fusion gene And the DHFR gene (see U.S. Patent 4,399,216).
  • DHFR Murine dihydrofolate reductase
  • the fusion protein expression plasmid is then transfected into a mammalian host cell line, and in order to obtain stable high levels of expression, the preferred host cell line is a DHFR enzyme deficient CHO-cell (see U.S. Patent 4,818,679).
  • the medium was changed to a screening medium containing 0.6 mg/mL G418, and the cells were planted in a 96-well culture plate at a concentration (5000-10000 viable cells/well) for 10-14 days until large. Discrete cell clones appear. Transfectants resistant to the selected drug were screened by ELISA assay. Subclones were generated to produce high levels of fusion protein wells by limiting dilution of 96-well plates.
  • the present invention constructs a series of rhFVII fusion proteins comprising peptide linkers of different lengths (Linker), CTP rigid units of different compositions, and IgG Fc variant (vFc) elements of several different subtypes.
  • Linker peptide linkers of different lengths
  • vFc IgG Fc variant
  • Example 8 kinds of expression plasmids obtained in Example 1 was used DNAFect LT reagent TM (ATGCell Company) in 30ml shake bottle transfection 3 ⁇ 10 7 CHO-K1 cells, the transfected cells were cultured at 1000ng / ml of vitamin K1 The growth was carried out for 5 days in serum-free growth medium, and the concentration of the fusion protein in the supernatant was measured, and its activity was measured by the method described in Example 7 or 8.
  • the ELISA results showed that the transient expression levels (quantity of the substances) of the eight plasmids under these conditions were similar, but their coagulation activities showed a large difference. Among them, we defined the molar activity of FP-A as 100%.
  • the fusion protein supernatants expressed by FP-F, FP-G and FP-H plasmids were less active, only 29.4%, 26.3% and 41.2% of FP-A.
  • the purified proteins were analyzed by SDS electrophoresis, showing these three fusions.
  • the protein fractions showed different degrees of polymerization; while the activities of FP-B, FP-C, FP-D and FP-E were 98.0%, 89.2%, 86.1% and 92.9%, respectively, of FP-A.
  • the reason for the long flexible peptide linker gives FVIIa a higher degree of flexibility, allowing it to rotate freely relative to Fc, which may bring the steric structure of FVIIa closer to the Fc region, and add a CTP rigid unit between the two.
  • a rigid peptide linker is added to make it away from each other.
  • the CTP rigid unit contains multiple glycosyl side chains.
  • the CTP rigid unit can form a fixed spatial conformation with respect to the random coil form of the flexible peptide linker. It can effectively separate the different functional regions of the fusion protein, which is more conducive to the two parts independently folded into the correct three-dimensional conformation, maintaining a high activity.
  • FP-A, FP-B, FP-C, FP-D and FP-E were transfected into a mammalian host cell line to express the FVII fusion protein.
  • a preferred host cell is a DHFR deficient CHO cell (U.S. Patent No. 4,818,679).
  • a preferred method of transfection is electroporation, and other methods can be used, including calcium phosphate co-precipitation, lipofection, and microinjection.
  • Electroporation method Using a Gene Pulser Electroporator (Bio-Rad Laboratories) set to 300 V voltage and 1050 ⁇ Fd capacitance, 50 ⁇ g of PvuI linearized expression plasmid was added to 2 to 3 ⁇ 10 7 cells in a cuvette, and electroporation was performed. The resulting cells were transferred to shake flasks containing 30 ml of growth medium. Two days after transfection, the medium was changed to a growth medium containing 0.6 mg/mL G418, and the cells were seeded in a 96-well culture plate at a concentration for 10-12 days until large discrete cell clones appeared.
  • a Gene Pulser Electroporator Bio-Rad Laboratories
  • the anti-human IgG Fc ELISA method is used to screen the transfectants that are resistant to the selected drugs, and the anti-FVII ELISA method can also be used for the quantitative determination of the fusion protein expression, and then the high-level expression fusion is produced by sub-cloning by limiting dilution method.
  • the pores of the protein are used to screen the transfectants that are resistant to the selected drugs, and the anti-FVII ELISA method can also be used for the quantitative determination of the fusion protein expression, and then the high-level expression fusion is produced by sub-cloning by limiting dilution method. The pores of the protein.
  • the transfected fusion protein gene was co-amplified with the DHFR gene in growth medium containing increasing concentrations of MTX. Subclones with positive dilution DHFR expression were gradually pressurized, and transfectants capable of growing in up to 6 ⁇ M MTX medium were screened, the secretion rate was determined, and a cell line highly expressing the foreign protein was selected.
  • a cell line having a secretion rate of more than about 3 (preferably about 5) IU/10 6 (i.e., millions) of cells per 24 hours is subjected to adaptive suspension culture using serum-free medium, and then the fusion protein is purified using conditioned medium. .
  • the high-yield cell line preferably obtained in Example 3 was first subjected to serum-free domestication culture in a culture dish, and then transferred to a shake flask for suspension and domestication culture. After the cells were adapted to these culture conditions, supplemental flow culture was then carried out in a 300 ml shake flask or perfusion culture was simulated by changing the medium daily.
  • the CHO-derived cell line producing the fusion protein FP-A obtained by the screening of Example 3 was fed and cultured for 14 days in a 300 ml volume shake flask, and the cumulative yield of the expressed recombinant fusion protein was 310 mg/L, and the viable cell density was obtained. Up to 15 ⁇ 10 6 / mL.
  • the above CHO-derived cell strain is changed daily in a 100 ml volume shake flask, and the recombinant fusion protein expressed has a cumulative yield of about 50 mg/L per day, and the viable cell density can reach up to 25 in a shake flask. ⁇ 10 6 / mL.
  • the biological activities of the recombinant fusion proteins produced by the above two methods are comparable.
  • FP-A is taken as an example to describe the purification steps and methods of several fusion proteins obtained in the above embodiments, and the FP-B, FP-C, FP-D and FP-E methods are the same, and the examples are no longer Narration.
  • the present invention mainly uses four-step chromatography to purify the fusion protein. Affinity chromatography, multidimensional mode chromatography, anion exchange chromatography and molecular sieve chromatography, respectively.
  • affinity chromatography uses the chromatographic medium described in the Summary of the Invention for sample capture.
  • multi-dimensional mode chromatography was used for sample capture using the chromatographic medium of the CHT ceramic hydroxyapatite skeleton of Bio-Rad Company described in the Summary of the Invention.
  • use balanced buffer 20-50mM PB, pH 6.8-7.2, equilibrate the column 3-5 column volumes (CV); sample captured by affinity chromatography, the loading is not higher than 10g / L;
  • Use balanced buffer 20-50mM PB, pH 6.8-7.2, equilibration column 3-5 Column volume (CV), rinse unbound components, named P1; rinse the column 3-5 column volumes using elution buffer 1:120 mM PB, pH 6.8-7.2, collect the eluted fractions, name P2; rinse the column 3-5 column volumes using elution buffer 2: 200 mM PB, pH 6.8-7.2, and collect the eluted fraction, designated P3.
  • the SEC detection result of the fusion protein FP-A by separating the target component P3 by the second step CHT chromatography is shown in FIG.
  • the SDS-PAGE electrophoresis results of the MabSelect separation component and the CHT chromatographic separation components P1, P2 and P3 are shown in Fig. 4.
  • anion exchange chromatography is further separated and purified using the chromatographic medium described in the Summary of the Invention.
  • the SEC results of this part are shown in Figure 5.
  • molecular sieve chromatography is carried out by using the chromatographic medium described in the section of the invention.
  • the column is equilibrated with a balance buffer: 100 mM Hepes, 100 mM NaCl, pH 8.0, and the column volume is 1.5-2 column volumes; the sample loading is not higher than the column. 2% by volume; using a balanced buffer: 100 mM Hepes, 100 mM NaCl, pH 8.0, eluting at a flow rate of 30 cm/h, abandoning the polymer peak in the previous stage, collecting the rising, middle and descending phases of the monomer peak (by chromatography
  • the peak of the highest UV absorption is limited to 1/2, named P3-2, P3-3, and P3-4.
  • the SEC test results of the P3-3 component are shown in Fig. 6.
  • the present invention activates the fusion protein by a solution incubation self-activation method.
  • the single component P3-3 of the fusion protein obtained by molecular sieve chromatography was loaded into a 30 kDa ultrafiltration concentrating tube (Corning, Cat. No. 431489), and then centrifuged in a benchtop high-speed cryogenic centrifuge (Eppendorf, model 5810R) to change the buffer to 20 mM PB. , 0.3 M NaCl, 5 mM CaCl 2 , pH 7.0-7.2, and concentrated to a protein concentration of 3-5 mg/ml (quantified by extinction coefficient method), activated at 4 ° C, activation time 16 to 40 hours.
  • the G-25 desalting column was used, and it was replaced with a buffer: 20 mM PB, pH 7.0, and stored at -80 ° C for use.
  • Example 7 or 8 of the present invention The biological activity assay described in Example 7 or 8 of the present invention and the reducing SDS-PAGE were used to detect the degree of activation of the chromatographic fractions in vitro for different times.
  • the biological activity results of different activation times are shown in Table 2; the P3-3 components were sampled during the activation process, and the results of the reduced SDS-PAGE of the activated products from 16 to 40 hours are shown in Figure 8, and the SEC detection results after 40 hours of activation are shown.
  • Figure 7 It can be seen that P3-3 has the highest degree of activation after 40 hours of activation.
  • Example 7 Indirect determination of biological activity of fusion protein by chromogenic substrate method
  • the in vitro enzymatic activity of each fusion protein activated by the method shown in Example 6 was measured using a BIOPHEN FVII chromogenic kit (Ref: A221304) manufactured by HYPHEN BioMed.
  • the kit is based on the chromogenic substrate method.
  • FVIIa is a serine protease that acts on the exogenous coagulation pathway. When FVIIa binds to tissue factor, it activates clotting factor FX in the presence of phospholipids and Ca 2+ . To convert it to the active form FXa.
  • the fusion protein to be determined first forms an enzyme complex with a tissue factor derived from rabbit thromboplastin, and then activates a certain concentration (excess) of factor FX in the reaction system to convert it into an active form of FXa, which acts on FXa.
  • the specific chromogenic substrate SXa-11 in the reaction system cleaves the substrate and produces pNA, and the amount of pNA produced directly correlates with the activity of FXa.
  • the concentration of FVIIa and FXa activity in the test sample was determined by measuring the concentration of pNA released at 405 nm with a colorimeter, thereby calculating the activity of FVIIa, using normal human plasma as a standard.
  • Example 8 Direct determination of the biological activity of a fusion protein by coagulation
  • Determination of the biological activity of FVIIa by coagulation is obtained by correcting the ability of FVIIa-factor-deficient plasma to cause prolonged clotting time.
  • the detection method firstly mixes diluted human lyophilized plasma of known FVII activity (Unicalibrator, Cat. No. 00625) with VII matrix plasma, determines prothrombin time (PT), establishes a standard curve, and then tests The plasma was moderately diluted and mixed with spent FVII matrix plasma for PT assay.
  • the logarithmic equation of the activity percentage C (%) and the PT time t(s) fitted by the standard curve can be used to measure the activity of the sample FVIIa, and the result is expressed as a percentage of normal plasma (%).
  • the bioactivity assay results of the separation components in each chromatographic step for 24 h in vitro are shown in Table 3.
  • the activity data of the target component P3-3 finally obtained by four-step chromatography activated for 16-40 hours is shown in Table 2.
  • the purified FP-A effective single component P3-3 has a biological activity of 22470 IU/mg for 40 h, and a molar activity of about 4417 IU/nM (each fusion protein contains 2 FVIIa, equivalent to 2208.7 IU/ nM FVIIa), comparable to the activity of the marketed recombinant FVIIa, Noci (2511 IU/nM FVIIa), indicating that the fusion protein provided by the present invention has a minimal effect on the activity of FVIIa by the C-terminally fused F.
  • the recombinant hFVII biological activity produced by HEK cells reported by CSL Behring Company is 2874 IU/mg, which is equivalent to 144 IU/nM.
  • the biological activity of the fusion protein hFVII-FP produced by HEK or CHO cells is 620-770 IU/mg, which is equivalent to 69-75 IU/nM (Weimer T et al., Thromb Haemost, 2008, 99: 659-667).
  • mice were anesthetized with 0.8% pentobarbital sodium (Sigma) at a dose of 0.1 ml/10 g, and then injected intravenously with 7,000 (HA-F-0.7W group) and 21,000 (HA-F-2.1W group). , 70,000 (HA-F-7W group) and 210,000 IU/kg (HA-F-21W group) of FP-A and 100,000 IU/kg of Noki ( Novo Nordisk) (HA-N group). After 5 minutes of administration, the mouse was cut at the distal end of the mouse.
  • pentobarbital sodium Sigma
  • the bleeding time of the FP-A 70,000 IU/kg group or the FP-A 210,000 IU/kg group was significantly shortened (p ⁇ 0.001; p ⁇ 0.001), and the amount of bleeding was also significantly reduced. (p ⁇ 0.001; p ⁇ 0.01) with a dose-effect relationship (see Table 4 and Table 5 for detailed results).
  • Group 1 C57-NS: C57BL/6J mice were given saline group;
  • Group 2 HA mice were given 10,000 IU/kg Novo group;
  • Group 3 HA-F-0.7W) ): HA mice were given to the FP-A 7,000 IU/kg group;
  • Group 4 HA-F-2.1W: HA mice were given to the FP-A 21,000 IU/kg group;
  • Group 5 HA-F-7W
  • Group 6 HA-F-21W: HA mice were given to the FP-A 210,000 IU/kg group.
  • mice given FP-A were subjected to a tail-breaking test at 1 h and 2 h after administration; the HA mice given Novo were tail-tested 1 h after administration; C57BL/6J normal control group (C57-NS) Group) Mice were subjected to a tail-break test 2 h after injection.
  • C57BL/6J normal control group (C57-NS) Group mice were subjected to a tail-break test 2 h after injection.
  • FP-A group was collected at 0.05, 0.5, 1, 2, 3, 5, 8, and 12 h after administration; the Noqi group was collected at 0.05, 0.5, 1, 2, 3, and 5 h after administration.
  • the blood sample was taken as an anticoagulant with a final concentration of 0.013 M sodium citrate, and the supernatant was taken by centrifugation at 3000 rpm for 10 min. The activity of the sample was measured according to Example 8 and the activity half-life was calculated.
  • the active half-life of FP-A was measured to be 3.03 ⁇ 0.35 h; the active half-life of Novo was 1.01 ⁇ 0.16 h.
  • the activity half-life of FP-A in rats was extended by about 3 times, and the plasma coagulation activity was about 40% after a single injection of FP-A for 3 hours, and the activity of the same active Noki group was 3 hours later. It has been reduced to 3%; plasma clotting activity remains above 7% after 12 hours of FP-A administration.

Landscapes

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

Abstract

The present invention discloses a highly glycosylated and activated human blood-clotting factor VII (FVIIa) fusion protein, and a manufacturing method and application of same. The fusion protein comprises, from the N-terminus to the C-terminus, a human (FVIIa), a flexible peptide connector, at least one rigid unit of a human chorionic gonadotropin β-subunit carboxyl terminal peptide, and a half-life extending portion (preferentially selected from a human IgG Fc variant). The fusion protein has a similar level of biological activity as a natural human (FVIIa) and extended in vivo half-life, thereby improving pharmacokinetics and drug efficacy.

Description

活化的人凝血因子VII融合蛋白及其制备方法与用途Activated human coagulation factor VII fusion protein, preparation method and use thereof 技术领域Technical field
本发明涉及一种融合蛋白,更具体地,涉及一种活化的人凝血因子VII(FVIIa)融合蛋白及其制备方法和用途,特别是在用于制备治疗多种凝血相关疾病药物中的用途。The present invention relates to a fusion protein, and more particularly to an activated human Factor VII (FVIIa) fusion protein, a preparation method and use thereof, and particularly to the use for the preparation of a medicament for treating various blood coagulation related diseases.
背景技术Background technique
FVII是一种维生素K依赖性的血浆糖蛋白,在肝脏中合成,并以分子量约为53KDa的单链蛋白酶原形式分泌到血液中(Broze等,J Biol Chem,1980,255:1242-1247)。FVII酶原在单一位点Arg152-Ile153处被蛋白酶水解,产生由一个二硫键连接的双链,从而转变为其活性形式FVIIa。活化的FVIIa包含NH2-末端衍生的轻链(约20KDa)和COOH-末端衍生的经由单一二硫键(Cys135至Cys262)连接的重链(约30KDa)。轻链含有细胞膜结合Gla结构域和两个“类表皮生长因子(EGF)结构域”,而重链含有丝氨酸蛋白酶催化结构域。处于活化形式时,FVIIa作为丝氨酸蛋白酶,参与凝血级联反应的外源性途径(extrinsic pathway)。在血友病治疗中使用FVIIa是基于FVIIa对于凝血酶活化的血小板表面低亲和性结合,通过给予药物学剂量的外源性FVIIa,使损伤位处血小板表面凝血酶产生被增强,这与FVIII/FIX存在无关,即绕开对于FVIIIa和FIXa的需求来起到止血作用。因此,FVIIa可用于产生抑制物的血友病患者出血性状况的预防和治疗。市售的重组凝血因子VIIa目前只有
Figure PCTCN2016106012-appb-000001
(Novo Nordisk,丹麦)。该药已在全世界范围内被批准用于治疗产生FVIII或FIX抗体(抑制物)的血友病A或B患者、先天性FVII缺陷的患者以及终止与外伤和/或手术相关的出血事件或预防出血。
FVII is a vitamin K-dependent plasma glycoprotein that is synthesized in the liver and secreted into the blood as a single-chain proteoprotein with a molecular weight of approximately 53 kDa (Broze et al, J Biol Chem, 1980, 255: 1242-1247) . The FVII zymogen is hydrolyzed by a protease at a single site, Arg152-Ile153, to produce a double-strand linked by a disulfide bond, thereby converting to its active form, FVIIa. Activated FVIIa comprising NH 2 - terminal derived light chain (about 20 KDa) and COOH- terminal-derived heavy chain (about 30KDa) connected via a single disulfide bond (Cys135 to Cys262). The light chain contains a cell membrane that binds to the Gla domain and two "epidermal growth factor (EGF) domains", while the heavy chain contains a serine protease catalytic domain. In the activated form, FVIIa acts as a serine protease and participates in the extrinsic pathway of the coagulation cascade. The use of FVIIa in the treatment of hemophilia is based on the low affinity binding of FVIIa to thrombin-activated platelet surface. By administering a pharmacological dose of exogenous FVIIa, platelet surface thrombin production at the lesion is enhanced, which is associated with FVIII. /FIX is irrelevant, that is, to bypass the demand for FVIIIa and FIXa to stop bleeding. Therefore, FVIIa can be used for the prevention and treatment of hemorrhagic conditions in hemophilia patients who produce inhibitors. Commercially available recombinant factor VIIa is currently only available
Figure PCTCN2016106012-appb-000001
(Novo Nordisk, Denmark). This drug has been approved worldwide for the treatment of hemophilia A or B patients who develop FVIII or FIX antibodies (inhibitors), patients with congenital FVII deficiency, and termination of bleeding events associated with trauma and/or surgery or Prevent bleeding.
然而,治疗性凝血蛋白药物包括FVIIa在内会被蛋白水解酶快速降解,并且易被抗体中和,这导致其体内循环半衰期大幅降低,由此限制了它们的疗效。重组FVIIa
Figure PCTCN2016106012-appb-000002
在人体内循环半衰期约为2.3小时,需要每隔2-3小时注射一次。
However, therapeutic coagulation protein drugs, including FVIIa, are rapidly degraded by proteolytic enzymes and are easily neutralized by antibodies, which results in a significant decrease in their circulating half-life in vivo, thereby limiting their efficacy. Recombinant FVIIa
Figure PCTCN2016106012-appb-000002
The circulating half-life in the human body is about 2.3 hours, and it needs to be injected every 2-3 hours.
为了提高FVIIa的体内半衰期,由CSL Behring公司开发的rFVIIa-FP(白蛋白与FVIIa的C端融合)已完成临床I期研究,它在正常受试者中的血清半 衰期与市售FVIIa相比提高了3-4倍(Golor G等,J Thromb Haemost,2013,11(11):1977-85),生物学活性为620-770IU/mg,相当于69-75IU/nM(Weimer T等,Thromb Haemost,2008,99:659-667)。In order to increase the in vivo half-life of FVIIa, rFVIIa-FP (albumin fusion with albumin and FVIIa) developed by CSL Behring has completed a clinical phase I study, which is serum half in normal subjects. The decay is 3-4 times higher than that of commercially available FVIIa (Golor G et al, J Thromb Haemost, 2013, 11(11): 1977-85), with a biological activity of 620-770 IU/mg, equivalent to 69-75 IU. /nM (Weimer T et al, Thromb Haemost, 2008, 99: 659-667).
FVIIa的聚乙二醇化脂质体制剂PEGLip-FVIIa(Omri公司开发)也处于临床早期,并且其血清半衰期仅为天然FVIIa的2倍。此外,由诺和诺德公司开发的N-糖基定点PEG化修饰FVIIa(N7-GP)相对rFVIIa延长了约5倍,但由于临床II期数据缺乏量-效线性关系且有受试者发生了过敏反应,该项目已终止研究(Ljung R等,J Thromb Haemost,2013,11(7):1260-8)。The PEGylated liposome formulation of FVIIa, PEPLip-FVIIa (developed by Omri), is also in the early clinical stage and has a serum half-life of only twice that of native FVIIa. In addition, the N-glycosyl-directed PEGylation modification of FVIIa (N7-GP) developed by Novo Nordisk Company was approximately 5-fold longer than that of rFVIIa, but due to the lack of quantitative-effect linearity in clinical phase II data and the occurrence of subjects The allergic reaction, the project has terminated research (Ljung R et al, J Thromb Haemost, 2013, 11 (7): 1260-8).
由Biogen公司开发的rFVIIaFc单体-二聚体杂合型(monomer/dimer hybrid)融合蛋白仍处于早期研究阶段,研究显示Monomeric rFVIIaFc在A型血友病小鼠体内的终末半衰期为6.6小时,为对照组rFVIIa
Figure PCTCN2016106012-appb-000003
的5.5倍。在静脉注射rFVIIaFc 5分钟时,小鼠凝血活性即恢复至正常值的40%,然而在起始阶段其活性会急剧衰减,在给药后30分钟即下降到20%左右,给药后1小时已下降至约10%,这与
Figure PCTCN2016106012-appb-000004
在0~1小时的下降趋势相似。虽然给药1h后与rFVIIa相比,rFVIIaFc组活性下降趋势有所变缓,但在静注rFVIIaFc 8小时后,活性已降至1%左右,已低于凝血活性基线水平,无法有效止血。这暗示rFVIIaFc仅在短时间内可有效控制急性出血症状,并不能长期、平稳、有效地维持机体的正常凝血功能。体外凝血活性试验显示rFVIIaFc摩尔比活性为1115IU/nM,与
Figure PCTCN2016106012-appb-000005
(2511IU/nM)相比下降约60%,研究者认为这种活性降低是由于融合配体Fc的空间位阻效应引起的,这使得它与可溶性组织因子(sTF)的结合常数(Kd)下降(J.Salsa等,Thrombosis Research,2015,135:970-976)。此外,本发明人之前构建的一种同源二聚体型rFVIIa-Fc,虽然其半衰期大幅延长,但也存在活性降低的问题,体外活性也仅为208-240IU/nM(中国发明专利号:CN104774269B)。
The rFVIIaFc monomer-dimer hybrid (momer/dimer hybrid) fusion protein developed by Biogen is still in the early stages of research, and the study showed that the terminal half-life of Monomeric rFVIIaFc in hemophilia A mice was 6.6 hours. For the control group rFVIIa
Figure PCTCN2016106012-appb-000003
5.5 times. At 5 minutes of intravenous injection of rFVIIaFc, the blood coagulation activity of the mouse returned to 40% of the normal value. However, its activity was abruptly attenuated at the initial stage, and decreased to about 20% at 30 minutes after administration, and 1 hour after administration. Has dropped to about 10%, which is related to
Figure PCTCN2016106012-appb-000004
The downward trend is similar in 0 to 1 hour. Although the decrease in the activity of the rFVIIaFc group was slower than that of rFVIIa after 1 hour of administration, the activity had decreased to about 1% after 8 hours of intravenous injection of rFVIIaFc, which was lower than the baseline level of coagulation activity and could not effectively stop bleeding. This suggests that rFVIIaFc can effectively control acute bleeding symptoms in a short period of time and does not maintain the normal coagulation function of the body for a long time, smoothly and effectively. The in vitro coagulation activity test showed that the rFVIIaFc molar specific activity was 1115 IU/nM,
Figure PCTCN2016106012-appb-000005
(2511 IU/nM) decreased by about 60%, the researchers believe that this decrease in activity is due to the steric hindrance effect of the fusion ligand Fc, which causes its binding constant (Kd) to decrease with soluble tissue factor (sTF). (J. Salsa et al., Thrombosis Research, 2015, 135: 970-976). In addition, a homodimeric rFVIIa-Fc previously constructed by the present inventors, although its half-life is greatly prolonged, has a problem of reduced activity, and the in vitro activity is only 208-240 IU/nM (Chinese invention patent number: CN104774269B) ).
人绒毛膜促性腺激素(hCG)β链的羧基末端肽(以下称其为CTP)也具有延长某些蛋白质体内半衰期的作用,因此有些专利文献公开的融合蛋白中包含的延长半衰期部分可以选择使用Fc、HSA、CTP或其他能延长半衰期的融合配体。例如,OPKO/Prolor公司开发的rFVIIa-CTP融合蛋白,rFVIIa分别串联1~5个CTP分子会以与CTP成比例的方式显著增加FVII的半衰期,当添加3、4或5 个CTP分别使FVII半衰期延长了0.67、2和3倍;但CTP的添加降低了rFVIIa比活性,如rFVIIa-CTP3相对于
Figure PCTCN2016106012-appb-000006
的比活性(U/mg FVIIa)降低了约4倍(中国发明专利申请号:CN201380019660.8)。另外,CTP也可以作为接头序列,主要用于连接同一个蛋白质的不同亚基。例如,中国专利CN103539860A、CN103539861A、CN103539868A和CN103539869A公开的融合蛋白中,CTP作为接头,位于促卵泡激素的beta亚基和alpha亚基之间;专利WO2005058953A2公开的融合蛋白中,CTP作为接头,用于连接糖蛋白激素的beta亚基和alpha亚基。
The carboxy-terminal peptide of the human chorionic gonadotropin (hCG) β-chain (hereinafter referred to as CTP) also has the effect of prolonging the half-life of certain proteins in vivo, and thus some of the extended half-life fractions contained in the fusion proteins disclosed in some patent documents can be selected. Fc, HSA, CTP or other fusion ligands that extend half-life. For example, the rFVIIa-CTP fusion protein developed by OPKO/Prolor, rFVIIa, in series with 1 to 5 CTP molecules, respectively, significantly increases the half-life of FVII in a manner proportional to CTP, when adding 3, 4 or 5 CTP to make FVII half-life, respectively. It is extended by 0.67, 2 and 3 times; however, the addition of CTP reduces the specific activity of rFVIIa, such as rFVIIa-CTP 3 relative to
Figure PCTCN2016106012-appb-000006
The specific activity (U/mg FVIIa) was reduced by about 4 times (Chinese Invention Patent Application No.: CN201380019660.8). In addition, CTP can also be used as a linker sequence to link different subunits of the same protein. For example, among the fusion proteins disclosed in Chinese Patent Nos. CN103539860A, CN103539861A, CN103539868A and CN103539869A, CTP acts as a linker between the beta subunit and the alpha subunit of follicle stimulating hormone; among the fusion proteins disclosed in WO2005058953A2, CTP is used as a linker for Linking the beta and alpha subunits of the glycoprotein hormone.
本发明人发现,现有技术公开的长效rFVIIa都存在半衰期延长有限、生物活性显著降低或免疫原性强的问题,其原因主要是因为FVIIa的C端含有丝氨酸蛋白酶结构域(153-406残基),因而C末端融合其他分子会影响FVIIa的催化活性及功能。另外,本发明人发现,在FVIIa和延长半衰期部分(如,免疫球蛋白Fc片段)之间,仅通过一段较长的常规的柔性肽接头(例如(GGGGS)n)连接,反而使得融合蛋白对蛋白酶更为敏感,更糟糕的是这样长的肽接头还使得大部分融合蛋白以聚合体形式表达,几乎没有生物学活性。本发明人经过长期的研究,令人意外地发现了,采用柔性肽和CTP共同组成的接头分子连接FVIIa和延长半衰期部分(如,人IgG Fc),能够最大程度地保留FVIIa的生物活性,并更显著地延长其体内活性半衰期。The present inventors have found that the long-acting rFVIIa disclosed in the prior art has a problem of limited half-life extension, significant decrease in biological activity or strong immunogenicity, mainly because the C-terminus of FVIIa contains a serine protease domain (153-406 residues). Therefore, the fusion of other molecules at the C-terminus affects the catalytic activity and function of FVIIa. In addition, the inventors have found that between FVIIa and an extended half-life portion (eg, an immunoglobulin Fc fragment), only a long stretch of conventional flexible peptide linkers (eg, (GGGGS)n) ligation, but instead the fusion protein pair Proteases are more sensitive, and even worse, such long peptide linkers also allow most of the fusion proteins to be expressed in polymer form with little biological activity. After long-term research, the present inventors have surprisingly discovered that a linker molecule composed of a flexible peptide and CTP is linked to FVIIa and an extended half-life portion (eg, human IgG Fc), thereby maximally retaining the biological activity of FVIIa, and More significantly extend its in vivo half-life of activity.
发明内容Summary of the invention
本发明目的是提供一种高糖基化的活化的人凝血因子VII融合蛋白,所述融合蛋白具有显著延长的半衰期且保持与重组人凝血因子VIIa(FVIIa)类似的生物学活性;另一方面,本发明还提供了所述融合蛋白的制备方法与用途。It is an object of the present invention to provide a hyperglycosylated activated human Factor VII fusion protein having a significantly extended half-life and maintaining biological activity similar to recombinant human Factor Vila (FVIIa); The invention also provides a preparation method and use of the fusion protein.
一方面,本发明提供了一种活化的人凝血因子VII融合蛋白(以下简称融合蛋白),所述融合蛋白从N端至C端依次含有活化的人凝血因子VII(FVIIa)、柔性肽接头(表示为L)、至少1个人绒毛膜促性腺激素β亚基的羧基末端肽刚性单元(以下简称CTP刚性单元,表示为(CTP)n,较优地,n为1,2,3,4,或5)和延长半衰期部分(如,免疫球蛋白Fc段、白蛋白、转铁蛋白或PEG,优选人IgG Fc变体(表示为vFc))。本发明的一些优选实施例中,所述融合蛋白表示为 FVIIa-L-CTPn-vFc。In one aspect, the invention provides an activated human Factor VII fusion protein (hereinafter referred to as a fusion protein) comprising, in order from the N-terminus to the C-terminus, an activated human Factor VII (FVIIa), a flexible peptide linker ( The carboxy terminal peptide rigid unit (hereinafter referred to as CTP rigid unit, denoted as (CTP) n ), which is L), at least one human chorionic gonadotropin β subunit, preferably n, 1, 2, 3, 4, Or 5) and an extended half-life portion (eg, immunoglobulin Fc fragment, albumin, transferrin or PEG, preferably a human IgG Fc variant (denoted as vFc)). In some preferred embodiments of the invention, the fusion protein is represented by FVIIa-L-CTP n -vFc.
其中,所述人凝血因子VII的氨基酸序列与天然人凝血因子VII至少80%同源;更优选地,所述人凝血因子VII的氨基酸序列与天然人凝血因子VII至少90%同源;最优选地,所述人凝血因子VII包含如SEQ ID NO:1所示的氨基酸序列。Wherein the amino acid sequence of human factor VII is at least 80% homologous to native human factor VII; more preferably, the amino acid sequence of human factor VII is at least 90% homologous to native human factor VII; most preferred The human Factor VII comprises the amino acid sequence set forth in SEQ ID NO: 1.
其中,FVIIa还包括于施药者体内被激活或于施药前被激活为FVIIa的以酶原形式存在的人凝血因子VII(简写为FVII)。Among them, FVIIa also includes human coagulation factor VII (abbreviated as FVII) which is activated in the applicator or which is activated as FVIIa before application.
其中,所述柔性肽接头优选是非免疫原性的,并且在FVII和Fc之间产生足够的距离,使相互之间的位阻效应降至最低。较佳地,使用含有2个或更多个氨基酸残基组成的柔性肽接头,且选自下列几种氨基酸:Gly(G)、Ser(S)、Ala(A)和Thr(T)。Wherein the flexible peptide linker is preferably non-immunogenic and produces a sufficient distance between FVII and Fc to minimize steric effects between each other. Preferably, a flexible peptide linker comprising two or more amino acid residues is used and is selected from the group consisting of Gly (G), Ser (S), Ala (A) and Thr (T).
更优选地,所述柔性肽接头包含G和S残基。连接肽的长度对融合蛋白的活性非常重要。对本发明而言,所述肽接头可优选地包含以(GS)a(GGS)b(GGGS)c(GGGGS)d循环单元组合形成的氨基酸序列通式,其中a,b,c和d是大于或等于0的整数,且a+b+c+d≥1。More preferably, the flexible peptide linker comprises G and S residues. The length of the linker peptide is very important for the activity of the fusion protein. For the purposes of the present invention, the peptide linker may preferably comprise an amino acid sequence formula formed by combining (GS) a (GGS) b (GGGS) c (GGGGS) d cycle units, wherein a, b, c and d are greater than Or an integer equal to 0, and a+b+c+d≥1.
具体地,本发明的实施例中,所述肽接头可优选地包含如下序列:Specifically, in an embodiment of the invention, the peptide linker may preferably comprise the following sequence:
(i)L1:GSGGGSGGGGSGGGGS(如SEQ ID NO:2所示);(i) L1: GSGGGSGGGGSGGGGS (as shown in SEQ ID NO: 2);
(ii)L2:GSGGGGSGGGGSGGGGSGGGGSGGGGS(如SEQ ID NO:3所示);(ii) L2: GSGGGGSGGGGSGGGGSGGGGSGGGGS (as shown in SEQ ID NO: 3);
(iii)L3:GGGGSGGGGSGGGGSGGGGS(如SEQ ID NO:4所示);(iii) L3: GGGGSGGGGSGGGGSGGGGS (as shown in SEQ ID NO: 4);
(iv)L4:GSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGS(如SEQ ID NO:5所示);(iv) L4: GSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGS (as shown in SEQ ID NO: 5);
(v)L5:GGGSGGGSGGGSGGGSGGGS(如SEQ ID NO:6所示);(v) L5: GGGSGGGSGGGSGGGSGGGS (as shown in SEQ ID NO: 6);
其中,所述CTP刚性单元选自由人绒毛膜促性腺激素β亚基羧基末端第113至145位氨基酸所组成的全长序列或其片段,具体地,所述刚性单元包含如SEQ ID NO:7所示氨基酸序列或其截短的序列。Wherein the CTP rigid unit is selected from the full length sequence consisting of amino acids 113 to 145 of the carboxy terminus of human chorionic gonadotropin β subunit or a fragment thereof, in particular, the rigid unit comprises SEQ ID NO: 7 The amino acid sequence shown or its truncated sequence.
优选地,所述CTP刚性单元包含至少2个糖基化位点;例如,本发明的一优选实施例中,所述CTP刚性单元包含2个糖基化位点,示例性地,所述CTP刚性单元包含SEQ ID NO:7N端的10个氨基酸,即SSSS*KAPPPS*,或所述CTP刚性单元包含SEQ ID NO:7C端的14个氨基酸,即S*RLPGPS*DTPILPQ;又如,另一实施例中,所述CTP刚性单元包含3个糖基化位点,示例性地,所述 CTP刚性单元包含SEQ ID NO:7N端的16个氨基酸,即SSSS*KAPPPS*LPSPS*R;再如,另一些实施例中,所述CTP刚性单元包含4个糖基化位点,示例性地,所述CTP刚性单元包含28、29、30、31、32或33个氨基酸并开始于人绒毛膜促性腺激素β亚基的第113、114、115、116、117或118位,终止于第145位。具体地,所述CTP刚性单元包含SEQ ID NO:7N端的28个氨基酸,即SSSS*KAPPPS*LPSPS*RLPGPS*DTPILPQ。在本文中,*代表糖基化位点。每种可能性都代表本发明的独立实施方式。Preferably, the CTP rigid unit comprises at least 2 glycosylation sites; for example, in a preferred embodiment of the invention, the CTP rigid unit comprises 2 glycosylation sites, exemplarily, the CTP The rigid unit comprises 10 amino acids of the N-terminus of SEQ ID NO: 7, ie SSSS*KAPPPS*, or the CTP rigid unit comprises 14 amino acids of the SEQ ID NO: 7C terminus, ie S*RLPGPS*DTPILPQ; as another example The CTP rigid unit comprises three glycosylation sites, exemplarily, said The CTP rigid unit comprises 16 amino acids of the N-terminus of SEQ ID NO: 7, ie SSSS*KAPPPS*LPSPS*R; as in other embodiments, the CTP rigid unit comprises 4 glycosylation sites, exemplarily, The CTP rigid unit comprises 28, 29, 30, 31, 32 or 33 amino acids and begins at position 113, 114, 115, 116, 117 or 118 of the human chorionic gonadotropin beta subunit, terminating at position 145 Bit. Specifically, the CTP rigid unit comprises 28 amino acids of the N-terminus of SEQ ID NO: 7, namely SSSS*KAPPPS*LPSPS*RLPGPS*DTPILPQ. In this context, * represents a glycosylation site. Each possibility represents a separate embodiment of the invention.
在另一些实施例中,本发明提供的CTP刚性单元与天然CTP氨基酸序列至少70%同源;在另一些实施例中,本发明提供的CTP刚性单元与天然CTP氨基酸序列至少80%同源;在另一些实施例中,本发明提供的CTP刚性单元与天然CTP氨基酸序列至少90%同源;在另一些实施例中,本发明提供的CTP刚性单元与天然CTP氨基酸序列至少95%同源。In other embodiments, the CTP rigid units provided herein are at least 70% homologous to the native CTP amino acid sequence; in other embodiments, the CTP rigid units provided herein are at least 80% homologous to the native CTP amino acid sequence; In other embodiments, the CTP rigid units provided herein are at least 90% homologous to the native CTP amino acid sequence; in other embodiments, the CTP rigid units provided herein are at least 95% homologous to the native CTP amino acid sequence.
本发明的具体实施例中,所述CTP刚性单元可优选地包含如下序列:In a particular embodiment of the invention, the CTP rigid unit may preferably comprise the following sequence:
(i)CTP1:PRFQDSSSSKAPPPSLPSPSRLPGPSDTPILPQ(如SEQ ID NO:7所示);(i) CTP 1 : PRFQDSSSSKAPPPSLPSPSRLPGPSDTPILPQ (as shown in SEQ ID NO: 7);
(ii)CTP2:SSSSKAPPPSLPSPSRLPGPSDTPILPQ(如SEQ ID NO:8所示);(ii) CTP 2 : SSSSKAPPPSLPSPSRLPGPSDTPILPQ (as shown in SEQ ID NO: 8);
(iii)CTP3:SSSSKAPPPS(如SEQ ID NO:9所示);(iii) CTP 3 : SSSSKAPPPS (shown as SEQ ID NO: 9);
(iv)CTP4:SRLPGPSDTPILPQ(如SEQ ID NO:10所示)。(iv) CTP 4 : SRLPGPSDTPILPQ (shown as SEQ ID NO: 10).
本发明一些实施例中,所述融合蛋白包含1个上述CTP刚性单元。In some embodiments of the invention, the fusion protein comprises one of the above CTP rigid units.
本发明另一些实施例中,所述融合蛋白包含1个以上的上述CTP刚性单元,优选地,包含2,3,4或5个上述CTP刚性单元,如本发明的一实施例中,所述融合蛋白包含2个CTP3刚性单元:SSSSKAPPPSSSSSKAPPPS(CTP3-CTP3,或表示为(CTP3)2)。In other embodiments of the invention, the fusion protein comprises more than one of the above CTP rigid units, preferably comprising 2, 3, 4 or 5 of the above CTP rigid units, as in an embodiment of the invention, The fusion protein comprises two CTP 3 rigid units: SSSSKAPPPSSSSSKAPPPS (CTP 3 -CTP 3 , or expressed as (CTP 3 ) 2 ).
其中,延长半衰期部分优选自免疫球蛋白IgG、IgM、IgA Fc片段;更优选自人IgG1、IgG2、IgG3或IgG4及其变体的Fc片段;进一步地,所述人IgG Fc变体包含位于野生型人IgG Fc中的至少一种氨基酸修饰,且变体具有降低的效应子功能(ADCC和/或CDC效应)和/或与新生儿受体FcRn的结合亲和力增强。进一步地,人IgG Fc变体可选自下组:Wherein the extended half-life portion is preferably a self-immunoglobulin IgG, IgM, IgA Fc fragment; more preferably an Fc fragment from human IgG1, IgG2, IgG3 or IgG4 and variants thereof; further, the human IgG Fc variant comprises in the wild At least one amino acid modification in a human IgG Fc, and the variant has reduced effector function (ADCC and/or CDC effect) and/or enhanced binding affinity to the neonatal receptor FcRn. Further, the human IgG Fc variant may be selected from the group consisting of:
(i)vFcγ1:含有Leu234Val、Leu235Ala和Pro331Ser突变的人IgG1绞链区、 CH2和CH3区域(如SEQ ID NO:11所示氨基酸序列);(i) vFcγ 1 : human IgG1 hinge region, CH2 and CH3 region containing the Leu234Val, Leu235Ala and Pro331Ser mutations (such as the amino acid sequence shown in SEQ ID NO: 11);
(ii)vFcγ2-1:含有Pro331Ser突变的人IgG2绞链区、CH2和CH3区域(如SEQ ID NO:12所示氨基酸序列);(ii) vFcγ 2-1 : human IgG2 hinge region, CH2 and CH3 region containing the Pro331Ser mutation (such as the amino acid sequence shown in SEQ ID NO: 12);
(iii)vFcγ2-2:含有Thr250Gln和Met428Leu突变的人IgG2绞链区、CH2和CH3区域(如SEQ ID NO:13所示氨基酸序列);(iii) vFcγ 2-2 : human IgG2 hinge region, CH2 and CH3 region containing the Thr250Gln and Met428Leu mutations (such as the amino acid sequence shown in SEQ ID NO: 13);
(iv)vFcγ2-3:含有Pro331Ser、Thr250Gln和Met428Leu突变的人IgG2绞链区、CH2和CH3区域(如SEQ ID NO:14所示氨基酸序列)。(iv) vFcγ 2-3 : human IgG2 hinge region, CH2 and CH3 regions (such as the amino acid sequence shown in SEQ ID NO: 14) containing the Pro331Ser, Thr250Gln and Met428Leu mutations.
(v)vFcγ4:含有Ser228Pro和Leu235Ala突变的人IgG4绞链区、CH2和CH3区域(如SEQ IDNO:15所示氨基酸序列)。(v) vFcγ 4 : human IgG4 hinge region, CH2 and CH3 regions containing the Ser228Pro and Leu235Ala mutations (such as the amino acid sequence shown in SEQ ID NO: 15).
本发明所提供的IgG Fc变体包含但不限于(i)~(v)中所述5种变体,还可以是IgG同种亚型间两类功能变体突变位点的组合或叠加,如上述(iv)中所述变体即是由(ii)和(iii)中的突变位点相叠加所获得的新的IgG2Fc的组合变体。The IgG Fc variants provided by the present invention include, but are not limited to, the five variants described in (i) to (v), and may also be a combination or superposition of two types of functional variant mutation sites between IgG isotypes. The variant as described in (iv) above is a novel variant of the novel IgG2Fc obtained by superimposing the mutation sites in (ii) and (iii).
本发明所述融合蛋白中的Fc变体(vFc),它含有人IgG如人IgG1、IgG2和IgG4的绞链区、CH2和CH3区域。这种CH2区域在228、234、235和331位(由EU计数系统确定)含有氨基酸突变。据信这些氨基酸突变能降低Fc的效应子功能。人IgG2不结合FcγR,但显示出极弱的补体活性。具有Pro331Ser突变的Fcγ2变体应比天然Fcγ2的补体活性更低,而且依旧是FcγR非结合子。IgG4Fc在激活补体级联中有缺陷,且它与FcγR的结合亲和力比IgG1低约一个数量级。与天然Fcγ4相比,具有Leu235Ala突变的Fcγ4变体应表现出最小的效应子功能。具有Leu234Va1、Leu235Ala和Pro331Ser突变的Fcγ1也表现出比天然Fcγ1降低的效应子功能。这些Fc变体都比天然人IgG Fc更适于制备FVIIa融合蛋白。而250和428位(由EU编号体系确定的位置)含有氨基酸突变,使得Fc区与新生儿受体FcRn的结合亲和力增加,从而进一步延长半衰期(Paul R等,J Biol Chem,2004,279:6213-6216);上述两类功能变体的相互组合或叠加,获得新的组合变体,使其效应子功能降低的同时且延长了其体内循环半衰期。本发明所述Fc变体包含却不局限于上述几个位点的突变,也可引入其它位点的替换使得Fc具有降低的效应子功能和/或与FcRn受体的结合力增强,同时还不会致使Fc变体功能/活性降低或引起不良的构象变化,常见的突变位点参见Shields RL等,J Biol Chem,2001,276(9):6591-604。 An Fc variant (vFc) in a fusion protein of the invention, which comprises a hinge region, a CH2 and a CH3 region of human IgG such as human IgG1, IgG2 and IgG4. This CH2 region contains amino acid mutations at positions 228, 234, 235 and 331 (as determined by the EU counting system). It is believed that these amino acid mutations reduce the effector function of Fc. Human IgG2 does not bind to FcyR but shows very weak complement activity. An Fc[gamma]2 variant with a Pro331Ser mutation should have a lower complement activity than native Fc[gamma]2 and is still an Fc[gamma]R non-binding element. IgG4Fc is defective in the activation of the complement cascade and its binding affinity to FcγR is about an order of magnitude lower than that of IgG1. An Fcγ4 variant with a Leu235Ala mutation should exhibit minimal effector function compared to native Fcγ4. Fcγ1 with Leu234Va1, Leu235Ala and Pro331Ser mutations also showed reduced effector function compared to native Fcγ1. These Fc variants are all more suitable for the preparation of FVIIa fusion proteins than native human IgG Fc. The 250 and 428 positions (positions determined by the EU numbering system) contain amino acid mutations that increase the binding affinity of the Fc region to the neonatal receptor FcRn, thereby further extending the half-life (Paul R et al, J Biol Chem, 2004, 279: 6213). -6216); the above two types of functional variants are combined or superimposed to obtain a new combined variant, which reduces the effector function and prolongs its circulating half-life in vivo. The Fc variants of the invention comprise, but are not limited to, mutations at several of the above sites, and substitutions at other sites may be introduced such that the Fc has reduced effector function and/or enhanced binding to the FcRn receptor, while still It does not cause a decrease in Fc variant function/activity or cause a poor conformational change. For common mutation sites, see Shields RL et al, J Biol Chem, 2001, 276(9):6591-604.
本发明的一优选实施例中,所述融合蛋白的氨基酸序列如SEQ ID NO:16所示;In a preferred embodiment of the present invention, the amino acid sequence of the fusion protein is as shown in SEQ ID NO:
根据本发明的另一个方面,提供一种编码上述融合蛋白的DNA。According to another aspect of the present invention, a DNA encoding the above fusion protein is provided.
本发明的一优选实施例中,所述融合蛋白的DNA序列如SEQ ID NO:17所示。In a preferred embodiment of the invention, the DNA sequence of the fusion protein is set forth in SEQ ID NO: 17.
根据本发明的再一个方面,提供一种载体,该载体包含上述DNA。According to still another aspect of the present invention, a vector comprising the above DNA is provided.
根据本发明的再一个方面,提供一种宿主细胞,该宿主细胞包含上述载体,或者转染了上述的载体。According to still another aspect of the present invention, a host cell comprising the above vector or transfected with the above vector is provided.
在本发明的实施例中,所述宿主细胞是CHO的衍生细胞株DXB-11。In an embodiment of the invention, the host cell is a derivative cell line DXB-11 of CHO.
根据本发明的再一个方面,提供一种药物组合物。该药物组合物包含药学上可接受的载体、赋形剂或稀释剂,以及有效量的上述融合蛋白。According to still another aspect of the present invention, a pharmaceutical composition is provided. The pharmaceutical composition comprises a pharmaceutically acceptable carrier, excipient or diluent, and an effective amount of the above fusion protein.
根据本发明的另一方面提供了一种从哺乳动物细胞系如CHO衍生的细胞系制备或生产所述融合蛋白的方法,包含以下步骤:According to another aspect of the invention there is provided a method of making or producing the fusion protein from a cell line derived from a mammalian cell line, such as CHO, comprising the steps of:
(a)将编码融合蛋白的DNA引入CHO细胞,生成CHO衍生细胞系;(a) introducing a DNA encoding a fusion protein into a CHO cell to generate a CHO-derived cell line;
(b)筛选步骤(a)中每24小时期间内,表达超过3μg/106个细胞的高产量细胞株;(b) a high-yield cell line expressing more than 3 μg/10 6 cells per 24 hours during the screening step (a);
(c)培养步骤(b)筛选到的细胞株,表达融合蛋白;(c) culturing step (b) the selected cell strain to express the fusion protein;
(d)收获步骤(c)得到的发酵液,纯化融合蛋白;(d) harvesting the fermentation broth obtained in step (c), purifying the fusion protein;
(e)激活步骤(d)中纯化的融合蛋白。(e) activating the purified fusion protein in step (d).
进一步地,所述步骤(a)中CHO衍生细胞系为DXB-11。Further, the CHO-derived cell line in the step (a) is DXB-11.
进一步地,所述步骤(d)中采用四步层析法对融合蛋白进行纯化,分别为ProteinA亲和层析、多维模式层析、阴离子交换层析和分子筛层析。Further, in the step (d), the fusion protein is purified by four-step chromatography, which is ProteinA affinity chromatography, multi-dimensional mode chromatography, anion exchange chromatography and molecular sieve chromatography.
第一步Protein A亲和层析步骤,优选地使用市售的例如但不限于GE的MabselectTM、Mabselect SuReTM,或TOSOH的Toyopearl AF-rProteinA-650FTM,或博格隆的rProtein A BestaroseTM,或天地人和的rProtein A BeadTM,或赛分科技的MabPurixTM层析介质。它是利用融合蛋白的Fc片段(人IgG)与单克隆抗体特异性的结合进行捕获,因为亲和介质上的proteinA配基对Fc段具有很高的亲和力,可以与其可逆的特异性结合。X射线晶体衍射分析显示,protein A和Fc片段结合位点中心一个高度保守的组氨酸残基六元环,在中性及碱性pH下这 个组氨酸残基不带电,因为疏水作用紧紧的结合在一起。而当pH降低或者疏水性减弱以后,组氨酸残基带电后相互排斥或者Fc融合蛋白在溶剂中的溶解性增强时,与亲和介质结合的蛋白能够被洗脱下来。因此本发明第一步采用亲和层析可以快速高效的从发酵液中捕获并纯化融合蛋白。Step Protein A affinity chromatography step, preferably using a commercially available, for example, but not limited to GE's Mabselect TM, Mabselect SuRe TM, or TOSOH the Toyopearl AF-rProteinA-650F TM, or the rProtein A Bestarose TM Boge Long , or the world of human and rProtein A Bead TM , or Safran Technology's MabPurix TM chromatography medium. It utilizes the specific binding of the Fc fragment of the fusion protein (human IgG) to the monoclonal antibody for capture because the proteinA ligand on the affinity medium has a high affinity for the Fc fragment and can specifically bind reversibly with it. X-ray crystallographic analysis showed that the protein A and Fc fragment binding sites are centered on a highly conserved six-membered histidine residue. This histidine residue is uncharged at neutral and alkaline pH because of hydrophobic interaction. Tightly combined. When the pH is lowered or the hydrophobicity is weakened, the histidine residues are mutually repelled upon charging or the solubility of the Fc fusion protein in the solvent is enhanced, and the protein bound to the affinity medium can be eluted. Therefore, in the first step of the present invention, affinity chromatography can rapidly and efficiently capture and purify the fusion protein from the fermentation broth.
第二步的多维模式层析步骤,优选地使用市售的例如但不限于Bio-Rad公司的CHTTM(陶瓷羟磷灰石I型或II型)层析介质(40μm或80μm)。融合蛋白N端存在10个可被羧基化的Glu残基,经羧基化后具有成对的负电荷。CHT介质为五个钙双联体和一对含羟基的磷三联体组按一个重复的几何图形排列,其中PO43-离子能与带正电的蛋白质以离子键结合,具有离子交换特性,而Ca2+离子与融合蛋白的自由羧基上的成对的负电荷以金属螯合方式结合,且该结合方式对NaCl不敏感,可由磷酸钠浓度梯度竞争性洗脱。根据文献报道,FVIIa的羧基化程度与其生物学活性具有相关性(Hagen FS等,Proc Natl Acad Sci USA,1986,83(8):2412-2416;美国专利US8304224;美国专利US2009/0042784)。所以本发明中经亲和层析捕获的融合蛋白使用CHT进行第二步纯化,以分离较高生物学活性的目标组分。The multi-dimensional mode chromatography step of the second step, preferably using commercially available CHT (TM) (ceramic hydroxyapatite Form I or Type II) chromatography media (40 [mu]m or 80 [mu]m), such as but not limited to Bio-Rad Corporation. There are 10 Glu residues that can be carboxylated at the N-terminus of the fusion protein, which have a pair of negative charges after carboxylation. The CHT medium is arranged in a repeating geometric pattern of five calcium doublets and a pair of hydroxyl-containing phosphorus triplets, wherein the PO4 3- ions are ionically bonded to the positively charged protein and have ion exchange characteristics. The paired negative charge of the Ca 2+ ion and the free carboxyl group of the fusion protein is bound by metal chelation, and the binding mode is insensitive to NaCl and can be competitively eluted by a sodium phosphate concentration gradient. According to the literature, the degree of carboxylation of FVIIa is correlated with its biological activity (Hagen FS et al, Proc Natl Acad Sci USA, 1986, 83(8): 2412-2416; US Pat. No. 8,032,224; US Patent No. US 2009/0042784). Therefore, the fusion protein captured by affinity chromatography in the present invention is subjected to a second purification step using CHT to separate a target component having a higher biological activity.
第三步的阴离子交换层析步骤,优选地使用市售的例如但不限于GE的Q Sepharose HPTM、Q Sepharose FFTM层析介质,或博格隆的Q Bestarose HPTM、Q Bestarose FFTM层析介质,或天地人和的Q Beads 6HPTM、Q Beads 6FFTM层析介质进行分离纯化,用于去除污染物。离子交换对分子的分离是基于它们表面净电荷的差异。分子的电荷性质差异较大,它们所带的总电荷、电荷密度以及表面电荷分布的差异会导致它们与加载电荷后的离子交换层析填料的结合能力不同。通过逐渐提高缓冲液中的NaCl浓度进行竞争性洗脱,可以将融合蛋白与污染物进行分离。The third step of anion exchange chromatography step, preferably using a commercially available, for example, but not limited to GE's Q Sepharose HP TM, Q Sepharose FF TM chromatography medium, or the Boge Long Q Bestarose HP TM, Q Bestarose FF TM layer Analysis of the medium, and the earth or the Q Beads 6HP TM, Q Beads 6FF TM chromatographic medium is isolated and purified, for removing contaminants. The separation of molecules by ion exchange is based on the difference in their net surface charge. The charge properties of molecules vary widely, and the difference in total charge, charge density, and surface charge distribution of the molecules causes them to differ in their ability to bind to the charge-exchanged ion-exchange chromatography packing. The fusion protein can be separated from the contaminants by progressively increasing the concentration of NaCl in the buffer for competitive elution.
第四步的分子筛层析步骤,优选地使用市售的例如但不限于GE的Superdex 200TM,或博格隆的Chromdex 200prep gradeTM层析介质进行分离。分子筛层析主要是根据蛋白分子大小来进行分离的一种层析方法。分子筛介质是多孔的球状颗粒,当不同大小的蛋白(如同一种蛋白的单体和聚合体)通过装好的分子筛柱时,不同大小的蛋白在柱中所走的路径不同,大分子不能进入介质的小孔中,在柱中保留时间相对短,先洗脱出来;而小分子可以进入介质的小孔中,在层析柱 中经过的路径要长,因此后洗脱出来。本发明中经上述三步层析分离得到的融合蛋白仍具有一定的聚合体组分,可能会影响其激活过程及激活后的生物学活性,因此采用分子筛层除去其中的聚合体组分。The molecular sieve chromatography step of the fourth step is preferably carried out using a commercially available Superdex 200 (TM) such as, but not limited to, GE, or a Chromdex 200 prep grade (TM) chromatography medium from Boglon. Molecular sieve chromatography is mainly a chromatographic method for separating according to the size of a protein molecule. Molecular sieve media are porous spherical particles. When different sizes of proteins (like a monomer and polymer of a protein) pass through a packed molecular sieve column, different sizes of proteins travel in different paths in the column, and macromolecules cannot enter. In the pores of the medium, the retention time in the column is relatively short and elutes first; while the small molecules can enter the pores of the medium, and the path passing through the column is long, and then elutes. The fusion protein obtained by the above three-step chromatography in the present invention still has a certain polymer component, which may affect the activation process and the biological activity after activation, and therefore the molecular sieve layer is used to remove the polymer component therein.
进一步地,所述步骤(e)中的激活步骤可以通过凝血因子XII(FXII)激活、柱上自激活或溶液孵育自激活法对融合蛋白进行激活。FVII转变成活性的FVIIa需要在FXII的作用下完成(Hedner等,J Clin Invest,1983,71:1836-1841),或者其他具有胰蛋白酶样活性的蛋白酶也可以激活FVII(Kisiel等,Behring Inst Mitt,1983,73:29-42);FVII也可以不利用其他蛋白酶,仅靠FVII自身激活为FVIIa,即通过它的丝氨酸蛋白酶区域自激活。FVII还可以通过与带正电荷表面或者填料结合完成自激活,如阴离子交换填料(Pedersen AH等,Biochemistry,1989,28:9331-9336)。增加离子强度、降低PH,或者增加溶液中Ca2+的浓度可以使活化的FVIIa从正电荷表面或者填料上解离(Bioern等,Research Disclosures,1986,269:564-56)。溶液条件下自激活参照美国专利US2007/0129298。Further, the activating step in the step (e) may activate the fusion protein by coagulation factor XII (FXII) activation, on-column self-activation or solution incubation self-activation. The conversion of FVII to active FVIIa needs to be done under the action of FXII (Hedner et al, J Clin Invest, 1983, 71: 1836-1841), or other proteases with trypsin-like activity can also activate FVII (Kisiel et al, Behring Inst Mitt) , 1983, 73: 29-42); FVII may also be activated by FVII itself, but by its serine protease region, by itself, without the use of other proteases. FVII can also be self-activated by binding to a positively charged surface or filler, such as an anion exchange packing (Pedersen AH et al, Biochemistry, 1989, 28: 9331-9336). Increasing the ionic strength, lowering the pH, or increasing the concentration of Ca 2+ in the solution can dissociate the activated FVIIa from the positively charged surface or onto the filler (Bioern et al, Research Disclosures, 1986, 269: 564-56). Self-activation under solution conditions is described in US 2007/0129298.
本发明一实施例中,采用溶液孵育自激活法对融合蛋白进行激活,活化的融合蛋白的活性>15000IU/mg。In one embodiment of the invention, the fusion protein is activated by a solution incubation self-activation method, and the activity of the activated fusion protein is >15000 IU/mg.
根据本发明的再一个方面,提供了所述融合蛋白在制备用于治疗或预防出血性疾病的药物中应用。优选地,所述融合蛋白用于治疗FVII先天性或获得性缺乏症患者的出血性疾病的预防或治疗,血友病A或B患者的自发或手术性出血的预防或治疗,或其它相关的出血性疾病药物中应用。According to still another aspect of the present invention, the fusion protein is provided for use in the preparation of a medicament for treating or preventing a bleeding disorder. Preferably, the fusion protein is used for the prevention or treatment of hemorrhagic diseases in patients with FVII congenital or acquired deficiency, prevention or treatment of spontaneous or surgical bleeding in patients with hemophilia A or B, or other related Application in bleeding drugs.
应理解,在本发明范围内,上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。It is to be understood that within the scope of the present invention, the various technical features described above and the various technical features specifically described hereinafter (as in the embodiments) may be combined with each other to form a new or preferred technical solution.
本发明人发现,本发明所公开和/或所记载的融合蛋白及其制备方法的优点可以概括如下:The inventors have found that the advantages of the fusion proteins and methods of preparation disclosed and/or described herein can be summarized as follows:
1、本发明构建的FVIIa融合蛋白,其Fc段是非裂解性的,即通过对Fc片段的补体、受体结合域进行突变,调节Fc与相应受体的结合亲和力,降低或消除ADCC和CDC效应,而只保留Fc段延长活性蛋白体内半衰期的作用,却不产生细胞毒性。Biogen公司开发的FVIIa融合蛋白,其Fc段是天然来源的,可以预测由Fc介导的不良效应子功能必将会增加患者的治疗风险。 1. The FVIIa fusion protein constructed by the present invention, wherein the Fc fragment is non-lytic, that is, by mutating the complement and receptor binding domains of the Fc fragment, regulating the binding affinity of the Fc to the corresponding receptor, and reducing or eliminating the ADCC and CDC effects. While retaining only the Fc segment to prolong the half-life of the active protein in vivo, it does not produce cytotoxicity. The FVIIa fusion protein developed by Biogen, whose Fc segment is of natural origin, predicts that Fc-mediated adverse effector functions will necessarily increase the patient's therapeutic risk.
2、FVIIa融合蛋白在体内活性半衰期延长了约3倍,单次注射FP-A 3h后血浆凝血活性约40%,而等活性给药的诺其组3h后活性已降至3%;FP-A给药12h后血浆凝血活性仍保持在7%以上,与Biogen公司开发的单体-二聚体杂合型(Monomeric)rFVIIaFc(参见J.Salsa等,Thrombosis Research,2015,135:970-976)相比,具有更长的体内活性半衰期。而血清中药物浓度波动减少,安全性提高,可降低注射频率而提高患者的生活质量;2. The half-life of FVIIa fusion protein was about 3 times longer in vivo. The plasma coagulation activity was about 40% after 3 hours of single injection of FP-A, while the activity of the same active group was reduced to 3% after 3 hours; FP- The plasma clotting activity remained above 7% after 12 hours of A administration, and the monomer-dimer heterozygous (Monomeric) rFVIIaFc developed by Biogen (see J. Salsa et al., Thrombosis Research, 2015, 135: 970-976). Compared to, it has a longer half-life of in vivo activity. The fluctuation of drug concentration in serum is reduced, and the safety is improved, which can reduce the frequency of injection and improve the quality of life of patients;
3、本发明构建的融合蛋白相对于Biogen公司构建的单体-二聚体杂合型(Monomeric)FVII融合蛋白,其表达、纯化步骤更加高效便捷、可大大降低生产成本。Biogen公司构建了rFVIIIFc与Fc的双表达载体,其中Fc分子以Flag标记(欧洲专利,公开号:EP1624891B1)。其所表达的融合蛋白发酵液中预期应含有三种形式的产物,分别是FVII-Fc:FVII-Fc同源二聚体型(Dimeric)融合蛋白、FVII-Fc:FLAG-Fc单体-二聚体杂合体(Monomeric)融合蛋白以及FLAG-Fc:FLAG-Fc二聚体三种产物。一方面,在融合蛋白表达过程中,因宿主细胞需同时表达FVII-Fc和Fc两种单链分子,再分别两两聚合形成上述三种产物,因而使最终目的产物的表达效率大大减弱;再者,在纯化过程中还必须去除另外两种形式的杂质,这使其纯化过程也更为复杂、生产效率低下,其生产成本也大大增加。因此,本发明的制备方法相对于Biogen公司开发的Monomeric rFVIIFc融合蛋白具有一定的技术优势和价格优势,其表达、纯化工艺都更简单、高效,生产成本也更低;3. The fusion protein constructed by the invention is more efficient and convenient than the Monomeric FVII fusion protein constructed by Biogen, and the production cost can be greatly reduced. Biogen Corporation constructed a dual expression vector for rFVIIIFc and Fc, wherein the Fc molecule was labeled with Flag (European Patent, Publication No. EP1624891B1). The fusion protein fermentation broth expressed by it is expected to contain three forms of products, namely FVII-Fc: FVII-Fc homodimeric (Dimeric) fusion protein, FVII-Fc: FLAG-Fc monomer-dimerization Monomeric fusion protein and FLAG-Fc: FLAG-Fc dimer three products. In one aspect, in the process of expression of the fusion protein, the host cell needs to simultaneously express two single-stranded molecules of FVII-Fc and Fc, and then separately polymerize to form the above three products, thereby greatly reducing the expression efficiency of the final target product; In addition, the other two forms of impurities must be removed during the purification process, which makes the purification process more complicated, the production efficiency is low, and the production cost is also greatly increased. Therefore, the preparation method of the invention has certain technical advantages and price advantages over the Monomeric rFVIIFc fusion protein developed by Biogen, and the expression and purification processes are simpler, more efficient, and the production cost is lower;
4、各批次纯化的融合蛋白的比活性至少可以达到15000IU/mg,约2949IU/nM(每个融合蛋白含有2个FVIIa,相当于1474.4IU/nM FVIIa),还有一些批次的融合蛋白活性最高甚至超过22470IU/mg,换算为摩尔比活性约为4325IU/nM(每个融合蛋白含有2个FVIIa,相当于2162IU/nM FVIIa),与已上市的重组FVIIa——诺其(2511IU/nM FVIIa)的活性相当,说明本发明提供的融合蛋白,其C端融合的Fc对FVIIa的活性影响极小;4. The specific activity of each batch of purified fusion protein can reach at least 15000 IU/mg, about 2949 IU/nM (2 FVIIa per fusion protein, equivalent to 1474.4 IU/nM FVIIa), and some batches of fusion protein. The activity is even higher than 22470 IU/mg, which is converted to a molar activity of about 4325 IU/nM (2 FVIIa per fusion protein, equivalent to 2162 IU/nM FVIIa), and the listed recombinant FVIIa-Noci (2511 IU/nM) The activity of FVIIa) is comparable, indicating that the fusion protein provided by the present invention has a C-terminally-fused Fc having minimal effect on the activity of FVIIa;
5、组成所述融合蛋白的CTP刚性单元,它含有多个O-糖基侧链,能形成相对稳定的立体构象,可以有效地将FVIIa与Fc隔离开。另一方面,CTP刚性单元含有糖基,带负电、高度唾液酸化的CTP刚性单元能够抵抗肾脏的清除作用,进一步延长融合蛋白的半衰期;再一方面,CTP刚性单元糖基侧链的保护作用可 以降低连接肽对蛋白酶的敏感性,使融合蛋白不易在连接区被降解;5. A CTP rigid unit constituting the fusion protein, which contains a plurality of O-glycosyl side chains, which can form a relatively stable stereo conformation and can effectively isolate FVIIa from Fc. On the other hand, the CTP rigid unit contains a glycosyl group, and the negatively charged, highly sialylated CTP rigid unit is able to resist the clearance of the kidney and further prolong the half-life of the fusion protein; on the other hand, the protective effect of the CTP rigid unit glycosyl side chain can be In order to reduce the sensitivity of the linker peptide to protease, the fusion protein is not easily degraded in the junction region;
6、本发明提供的所述融合蛋白的制备方法,具有产量高的优点。本发明的一优选实施例中,在300ml摇瓶中连续培养14天,累积产量达到310mg/L,可进行工艺放大,实现大规模工业化生产。6. The method for preparing the fusion protein provided by the invention has the advantages of high yield. In a preferred embodiment of the present invention, the culture is continuously cultured for 14 days in a 300 ml shake flask, and the cumulative yield reaches 310 mg/L, which can be subjected to process enlargement to realize large-scale industrial production.
发明详述:Detailed description of the invention:
柔性连接肽Flexible linker
连接肽长度对融合蛋白活性非常重要。WO2007090584A1专利中公开的一种FVII/FVIIa-白蛋白融合多肽,在FVII/FVIIa与白蛋白之间插入不同长度的连接肽,发现无连接肽的FVII/FVIIa融合蛋白的生物学活性显著下降,而含有连接肽的FVII/FVIIa-FP融合蛋白,显示其生物学活性的增加依赖于连接肽的长度,这可能解释为融合蛋白两部分间增加的连接肽,使该分子的两部分能分别行使其功能,有利于形成更高比活性的构象。The length of the linker peptide is very important for the activity of the fusion protein. A FVII/FVIIa-albumin fusion polypeptide disclosed in WO2007090584A1, in which a different length of a linker peptide is inserted between FVII/FVIIa and albumin, and the biological activity of the FVII/FVIIa fusion protein without the linker is found to be significantly decreased. The FVII/FVIIa-FP fusion protein containing a linker peptide shows that the increase in its biological activity depends on the length of the linker peptide, which may be explained by the increased linker peptide between the two parts of the fusion protein, allowing the two parts of the molecule to exercise their respective The function is conducive to the formation of a higher specific activity conformation.
本发明人此前设计了3种不同长度的由甘氨酸和丝氨酸组成的柔性连接肽,FVII通过该连接肽与其C端融合的Fc连接组成融合蛋白(不含CTP刚性单元),瞬时表达实验表明,由2个氨基酸的短肽接头GlySer连接的融合蛋白几乎检测不到活性,表明维持FVIIa生物活性的重要功能区域在三维结构上受C端融合配体的影响较大。当连接肽增加到16个氨基酸时,FVIIa融合蛋白的生物活性明显提高,但仍远低于重组FVIIa。再当连接肽进一步延长至37个氨基酸时,CHO细胞所分泌的融合蛋白部分发生聚合,活性较低。这表明单纯通过延长连接肽的长度并不能完全解决Fc片段对FVIIa活性影响的问题。The present inventors have previously designed three different lengths of flexible linker peptides composed of glycine and serine, and FVII constitutes a fusion protein (without CTP rigid unit) through the Fc junction of the linker peptide and its C-terminal fusion, and transient expression experiments show that The two amino acid short peptide linker GlySer-linked fusion protein showed little activity, indicating that the important functional regions that maintain the biological activity of FVIIa are greatly affected by the C-terminal fusion ligand in three-dimensional structure. When the linker peptide was increased to 16 amino acids, the biological activity of the FVIIa fusion protein was significantly increased, but still much lower than recombinant FVIIa. When the linker peptide is further extended to 37 amino acids, the fusion protein portion secreted by the CHO cell is polymerized and has low activity. This indicates that the problem of the effect of the Fc fragment on FVIIa activity cannot be completely solved by simply extending the length of the linker peptide.
hCG-β羧基末端肽(CTP)hCG-β carboxy terminal peptide (CTP)
CTP是一段来自人绒毛膜促性腺激素(hCG)的β-亚基羧基末端的短肽。四种与生殖相关的多肽类激素促卵泡激素(FSH)、黄体生成素(LH)、促甲状腺素(TSH)和绒毛膜促性腺激素(hCG)含有相同的α-亚基和各自特异的β-亚基。与其它三种激素相比,hCG体内半衰期明显延长,这主要来源于其β-亚基上特有的羧基末端肽(CTP)(Fares FA等,ProcNatl Acad Sci USA,1992,89:4304-4308)。天然的CTP含有37个氨基酸残基,它具有4个O-糖基化位点,终端是唾液酸 残基。带负电、高度唾液酸化的CTP能够抵抗肾脏对其的清除作用,从而延长蛋白在体内的半衰期。然而,本发明人创造性地将至少一个CTP多肽与适当长度的柔性连接肽连接,共同作为连接肽,用于连接FVII与延长半衰期部分(如,免疫球蛋白Fc片段)。CTP is a short peptide derived from the carboxy terminus of the β-subunit of human chorionic gonadotropin (hCG). Four reproductive-related peptide hormones, follicle stimulating hormone (FSH), luteinizing hormone (LH), thyrotropin (TSH), and chorionic gonadotropin (hCG) contain the same alpha-subunit and their respective specific beta - Yaki. Compared with the other three hormones, the half-life of hCG is significantly prolonged, mainly due to the unique carboxy terminal peptide (CTP) on its β-subunit (Fares FA et al, ProcNatl Acad Sci USA, 1992, 89: 4304-4308) . The natural CTP contains 37 amino acid residues with four O-glycosylation sites and the terminal is sialic acid. Residues. Negatively charged, highly sialylated CTP is resistant to the clearance of the kidneys, thereby prolonging the half-life of the protein in the body. However, the inventors creatively linked at least one CTP polypeptide to a flexible linker of appropriate length, collectively as a linker peptide, for ligation of FVII with an extended half-life moiety (eg, an immunoglobulin Fc fragment).
本发明人发现,FVIIa的C端催化功能域对其功能发挥极为关键,且FVIIa空间构象复杂、脆弱,融合配体的位阻效应极易对其正确折叠造成干扰。通过在FVIIa与Fc变体间增加CTP刚性单元,相当于增加了一段刚性连接肽。这一方面保证了N-端融合的FVIIa不会影响Fc变体与FcRn的结合位点,从而影响半衰期;另外Fc的ProteinA结合位点对于制备工艺中纯化步骤很重要,连接CTP刚性单元保证N-端融合的FVIIa也不会“罩住”它与protein A的结合位点。另一方面,CTP刚性单元的添加也使得约25KD大小的Fc片段不会干扰N-端融合的FVIIa的正确折叠,造成其生物学活性/功能的下降或丧失。同时还证实单纯延长柔性连接肽的长度并不能改善Fc段对FVIIa活性的影响,过长的连接肽还会造成多聚体的形成以及对蛋白酶敏感性增加而易被降解,而CTP刚性单元的加入使得融合蛋白的生物学活性显著提高。这可能解释为具有多个糖基侧链的CTP刚性多肽,相对于(GGGGS)n这类柔性连接肽的无规则卷曲,它可以形成稳定的立体构象,这种“阻隔”作用促使FVIIa和Fc段独立折叠形成正确的三维构象而互不影响各自的生物活性。再一方面,CTP糖基侧链的保护作用可以降低连接肽对蛋白酶的敏感性,使融合蛋白不易在连接区被降解。The present inventors have found that the C-terminal catalytic domain of FVIIa is critical for its function, and the FVIIa spatial conformation is complex and fragile, and the steric hindrance effect of the fusion ligand is highly susceptible to interference with its correct folding. By adding a CTP rigid unit between FVIIa and the Fc variant, an equivalent of a rigid linker peptide is added. This aspect ensures that the N-terminally fused FVIIa does not affect the binding site of the Fc variant to FcRn, thereby affecting the half-life; in addition, the Fc-ProteinA binding site is important for the purification step in the preparation process, and the CTP rigid unit is connected to ensure N The end-fused FVIIa also does not "cover" its binding site to protein A. On the other hand, the addition of a CTP rigid unit also allows the Fc fragment of about 25 kD size to not interfere with the correct folding of the N-terminally fused FVIIa, resulting in a decrease or loss of its biological activity/function. It was also confirmed that simply extending the length of the flexible linker peptide did not improve the effect of the Fc fragment on FVIIa activity. Too long linker peptides also caused the formation of multimers and increased sensitivity to proteases, which were easily degraded, whereas CTP rigid units The addition causes a significant increase in the biological activity of the fusion protein. This may be interpreted as a CTP rigid polypeptide with multiple glycosyl side chains, which can form a stable stereoconfiguration relative to the random coiling of a flexible linker peptide such as (GGGGS)n, which promotes FVIIa and Fc. The segments are independently folded to form the correct three-dimensional conformation without affecting each other's biological activity. On the other hand, the protective effect of the CTP glycosyl side chain can reduce the sensitivity of the linker peptide to proteases, making the fusion protein less susceptible to degradation in the junction region.
IgG Fc变体IgG Fc variant
非裂解性Fc变体Non-lytic Fc variant
Fc元件来源于免疫球蛋白IgG的恒定区Fc片段,它在消灭病原体的免疫防御中起重要作用。Fc介导的IgG的效应子功能发挥通过两种机制:(1)与细胞表面Fc受体(FcγRs)结合,由吞噬作用或裂解作用或杀伤细胞通过抗体依赖性细胞毒性(ADCC)途径消化病原体,或(2)与第一补体成分C1的C1q结合,引发补体依赖性细胞毒性(CDC)途径,从而裂解病原体。在四种人IgG亚型中,IgG1和IgG3能有效结合FcγRs,IgG4与FcγRs的结合亲和力较低,而IgG2与FcγRs的结合低得难以测定,所以人IgG2几乎没有ADCC效应。此外,人IgG1 和IgG3还能有效结合C1q而激活补体级联反应。人IgG2与C1q结合相对弱,而IgG4不与C1q结合(Jefferis R等,Immunol Rev,1998,163:59-76),所以人IgG2CDC效应也较弱。显然,没有一种天然IgG亚型是非常适合构建FVIIa/Fc融合蛋白的。为了得到不具效应子功能的非裂解性Fc,最有效方法是对Fc片段上补体、受体结合域突变改造,调节Fc与相关受体的结合亲和力,降低或消除ADCC和CDC效应,只保留Fc的长循环半衰期特性,而不产生细胞毒性。更多的非裂解性Fc变体所包含突变位点可以参见Shields RL等,J Biol Chem,2001,276(9):6591-604或中国发明专利CN 201280031137.2。The Fc element is derived from the constant region Fc fragment of immunoglobulin IgG, which plays an important role in eradicating the immune defense of pathogens. The effector function of Fc-mediated IgG is exerted through two mechanisms: (1) binding to cell surface Fc receptors (FcγRs), digestion of pathogens by phagocytosis or cleavage or killer cells via antibody-dependent cellular cytotoxicity (ADCC) pathway , or (2) binding to C1q of the first complement component C1, eliciting a complement-dependent cytotoxicity (CDC) pathway, thereby lysing the pathogen. Among the four human IgG subtypes, IgG1 and IgG3 efficiently bind to FcγRs, and the binding affinity of IgG4 to FcγRs is low, and the binding of IgG2 to FcγRs is too low to be determined, so human IgG2 has almost no ADCC effect. In addition, human IgG1 And IgG3 can also effectively bind to C1q to activate the complement cascade. Human IgG2 binds relatively weakly to C1q, whereas IgG4 does not bind to C1q (Jefferis R et al, Immunol Rev, 1998, 163: 59-76), so the human IgG2 CDC effect is also weak. Clearly, none of the native IgG subtypes are well suited for the construction of FVIIa/Fc fusion proteins. In order to obtain non-lytic Fc without effector function, the most effective method is to mutate the complement and receptor binding domain of the Fc fragment, modulate the binding affinity of Fc to related receptors, reduce or eliminate ADCC and CDC effects, and retain only Fc. Long cycle half-life characteristics without cytotoxicity. For more non-lytic Fc variants, the mutation sites can be found in Shields RL et al, J Biol Chem, 2001, 276(9): 6591-604 or Chinese invention patent CN 201280031137.2.
与新生儿受体(FcRn)结合亲和力增强的Fc变体Fc variant with enhanced binding affinity to neonatal receptor (FcRn)
IgG的血浆半衰期取决于它与FcRn的结合,一般在pH6.0时结合,在pH7.4(血浆pH)时解离。通过对两者结合位点的研究,改造IgG上与FcRn结合的位点,使之在pH6.0时结合能力增加。已经证明对于结合FcRn重要的人Fcγ结构域的一些残基的突变可增加血清半衰期。已报道T250、M252、S254、T256、V308、E380、M428和N434中的突变可增加或降低FcRn结合亲和力(Roopenian等,Nat.Rview Immunology7:715-725,2007)。韩国专利号KR 10-1027427公开了具有增加的FcRn结合亲和力的曲妥珠单抗(赫赛汀,Genentech)变体,并且这些变体包含选自257C、257M、257L、257N、257Y、279Q、279Y、308F和308Y的一个或更多个氨基酸修饰。韩国专利公开号KR 2010-0099179提供了贝伐单抗(阿瓦斯汀,Genentech)变体并且这些变体通过包含在N434S、M252Y/M428L、M252Y/N434S和M428L/N434S的氨基酸修饰显示增加的体内半衰期。此外,Hinton等也发现T250Q和M428L 2个突变体分别使与FcRn的结合增加3和7倍。同时突变2个位点,则结合增加28倍。在恒河猴体内,M428L或T250QM/428L突变体显示血浆半衰期增加2倍(Paul R.Hinton等,J Immunol,2006,176:346-356)。更多的与新生儿受体(FcRn)结合亲和力增强的Fc变体所包含突变位点可以参见中国发明专利CN201280066663.2。此外,有研究对五种人源化抗体的Fc段进行T250Q/M428L突变不仅改善了Fc与FcRn的相互作用,且在随后的体内药代动力学试验中,发现以皮下注射方式给药,Fc突变抗体与野生型抗体相比药代动力学参数有所改善,如体内暴露量增加,清除率降低,皮下生 物利用度提高(Datta-Mannan A等.MAbs.Taylor&Francis,2012,4(2):267-273.)。The plasma half-life of IgG depends on its binding to FcRn, which typically binds at pH 6.0 and dissociates at pH 7.4 (plasma pH). By studying the binding sites of the two, the site of binding to FcRn on IgG was engineered to increase the binding ability at pH 6.0. Mutations in some residues of the human Fcγ domain important for binding to FcRn have been shown to increase serum half-life. Mutations in T250, M252, S254, T256, V308, E380, M428 and N434 have been reported to increase or decrease FcRn binding affinity (Roopenian et al, Nat. Rview Immunology 7: 715-725, 2007). Korean Patent No. KR 10-1027427 discloses variants of trastuzumab (Herceptin, Genentech) having increased FcRn binding affinity, and these variants are selected from the group consisting of 257C, 257M, 257L, 257N, 257Y, 279Q, One or more amino acid modifications of 279Y, 308F and 308Y. Korean Patent Publication No. KR 2010-0099179 provides variants of bevacizumab (Avastin, Genentech) and these variants show increased in vivo by amino acid modifications contained in N434S, M252Y/M428L, M252Y/N434S and M428L/N434S half life. In addition, Hinton et al. also found that two mutants, T250Q and M428L, increased the binding to FcRn by 3 and 7 fold, respectively. At the same time, two sites were mutated, and the binding was increased by 28 times. In rhesus monkeys, the M428L or T250QM/428L mutant showed a 2-fold increase in plasma half-life (Paul R. Hinton et al, J Immunol, 2006, 176: 346-356). More mutation sites including Fc variants with enhanced binding affinity to neonatal receptor (FcRn) can be found in Chinese invention patent CN201280066663.2. In addition, studies on T250Q/M428L mutations in the Fc segment of five humanized antibodies not only improved the interaction of Fc with FcRn, but were also administered by subcutaneous injection in subsequent in vivo pharmacokinetic assays, Fc. Mutant antibodies have improved pharmacokinetic parameters compared to wild-type antibodies, such as increased in vivo exposure, reduced clearance, subcutaneous Increased availability (Datta-Mannan A et al. MAbs. Taylor & Francis, 2012, 4(2): 267-273.).
融合蛋白及其制备方法Fusion protein and preparation method thereof
本发明融合蛋白基因是密码子优化过的由人工合成方法制备。根据本发明所述的核苷酸序列,本领域技术人员可方便的用各种已知方法制得本发明的编码核酸。这些方法不限于人工合成或传统亚克隆等,具体方法可参见J.萨姆布鲁克,《分子克隆实验指南》。作为本发明的一种实施方式,通过分段合成核苷酸序列再进行亚克隆的方法来构建本发明的编码核酸序列。The fusion protein gene of the present invention is codon-optimized and prepared by a synthetic method. According to the nucleotide sequence of the present invention, those skilled in the art can conveniently prepare the nucleic acid of the present invention by various known methods. These methods are not limited to synthetic or traditional subcloning, and the specific method can be found in J. Sambrook, Molecular Cloning Experiment Guide. As an embodiment of the present invention, the nucleic acid sequence of the present invention is constructed by subcloning a nucleotide sequence and then subcloning.
本发明还提供了一种哺乳动物细胞的表达载体,包含编码本发明的融合蛋白序列以及与之操作性相连的表达调控序列。所述的“操作性相连”或“可操作地连于”指这样一种状况,即线性DNA序列的某些部分能够调节或控制同一线性DNA序列其它部分的活性。例如,如果启动子控制序列的转录,那么它就是可操作地连于编码序列。The invention also provides an expression vector for a mammalian cell comprising a fusion protein sequence encoding the invention and an expression control sequence operably linked thereto. By "operably linked" or "operably linked" is meant a condition in which portions of a linear DNA sequence are capable of modulating or controlling the activity of other portions of the same linear DNA sequence. For example, if a promoter controls the transcription of a sequence, then it is operably linked to the coding sequence.
哺乳动物细胞表达载体可采用市售的例如但不限于:pcDNA3、pIRES、pDR、pBK、pSPORT等可用于真核细胞系统表达的载体。本领域技术人员还可以根据宿主细胞来选择合适的表达载体。The mammalian cell expression vector can be commercially available, for example, but not limited to, pcDNA3, pIRES, pDR, pBK, pSPORT, etc., which can be used for expression in eukaryotic cell systems. One skilled in the art can also select a suitable expression vector based on the host cell.
根据已知空载表达载体的酶切图谱,本领域技术人员可按照常规方法通过限制性酶剪切与拼接,将本发明的融合蛋白的编码序列插入合适的限制性位点,制得本发明的重组表达载体。According to the restriction enzyme map of the known empty-load expression vector, the skilled person can prepare the present invention by inserting the coding sequence of the fusion protein of the present invention into a suitable restriction site by restriction enzyme cleavage and splicing according to a conventional method. Recombinant expression vector.
本发明还提供了表达本发明融合蛋白的宿主细胞,其中含有本发明的融合蛋白的编码序列。所述的宿主细胞优选的是真核细胞,例如但不限于CHO细胞,COS细胞,293细胞,RSF细胞等。作为本发明的优选方式,所述的细胞是CHO细胞,其可较佳地表达本发明的融合蛋白,可获得活性良好,稳定性良好的融合蛋白。The invention also provides a host cell expressing a fusion protein of the invention comprising a coding sequence for a fusion protein of the invention. The host cell is preferably a eukaryotic cell such as, but not limited to, a CHO cell, a COS cell, a 293 cell, an RSF cell, and the like. In a preferred embodiment of the present invention, the cell is a CHO cell which can preferably express the fusion protein of the present invention, and a fusion protein having good activity and good stability can be obtained.
本发明还提供一种用重组DNA技术制备本发明融合蛋白的方法,其步骤包含:The invention also provides a method for preparing a fusion protein of the invention by recombinant DNA technology, the steps of which comprise:
1)提供编码融合蛋白的核酸序列;1) providing a nucleic acid sequence encoding a fusion protein;
2)将1)的核酸序列插入到合适的表达载体,获得重组表达载体;2) inserting the nucleic acid sequence of 1) into a suitable expression vector to obtain a recombinant expression vector;
3)将2)的重组表达载体导入合适的宿主细胞; 3) introducing the recombinant expression vector of 2) into a suitable host cell;
4)在适合表达的条件下培养转染宿主细胞;4) cultivating the transfected host cell under conditions suitable for expression;
5)收集上清液,并纯化融合蛋白产物;5) collecting the supernatant and purifying the fusion protein product;
6)激活融合蛋白。6) Activate the fusion protein.
将所述编码序列导入宿主细胞可采用本领域的多种已知技术,例如但不限于:磷酸钙沉淀,脂质体转染,电穿孔,微注射,病毒感染法,碱金属离子法。Introduction of the coding sequence into a host cell can employ a variety of known techniques in the art such as, but not limited to, calcium phosphate precipitation, lipofection, electroporation, microinjection, viral infection, alkali metal ion methods.
有关宿主细胞的培养和表达可参见Olander RM等,Dev Biol Stand 1996,86:338。可通过离心去除悬浮液中的细胞和残渣,收集上清液。For the culture and expression of host cells, see Olander RM et al, Dev Biol Stand 1996, 86:338. The cells and debris in the suspension can be removed by centrifugation and the supernatant collected.
有关融合蛋白的激活方法,可以通过凝血因子XII(FXII)激活、柱上自激活或溶液孵育自激活法对融合蛋白进行激活。Regarding the activation method of the fusion protein, the fusion protein can be activated by coagulation factor XII (FXII) activation, on-column autoactivation or solution incubation self-activation.
可将上述制备获得的融合蛋白纯化为基本均一的性质,例如在SDS-PAGE电泳上呈单一条带。首先将表达上清浓缩,浓缩液可采用凝胶层析的方法进一步加以纯化,或采用离子交换层析的方法纯化。例如阴离子交换层析或阳离子交换层析。凝胶基质可为琼脂糖、葡聚糖、聚酰胺等常用于蛋白纯化的介质。Q-或SP-基团是较为理想的离子交换基团。最后,还可用羟基磷灰石吸附层析,金属螯合层析,疏水相互作用层析和反相高效液相色谱等方法对上述纯化产物进一步精制纯化。上述所有纯化步骤可利用不同的组合,最终使蛋白纯度达到基本均一。还可利用含有所述融合蛋白的特异性抗体、受体或配体的亲和层析柱对表达的融合蛋白进行纯化。根据所使用的亲和柱的特性,可利用常规的方法,如高盐缓冲液、改变pH等方法洗脱结合在亲和柱上的融合性多肽。The fusion protein obtained as described above can be purified to a substantially uniform property, such as a single band on SDS-PAGE electrophoresis. The supernatant is first concentrated, and the concentrate can be further purified by gel chromatography or by ion exchange chromatography. For example, anion exchange chromatography or cation exchange chromatography. The gel matrix may be a medium commonly used for protein purification such as agarose, dextran, polyamide, and the like. The Q- or SP- group is a preferred ion exchange group. Finally, the purified product may be further purified by hydroxyapatite adsorption chromatography, metal chelate chromatography, hydrophobic interaction chromatography and reversed-phase high performance liquid chromatography. All of the above purification steps can utilize different combinations to ultimately achieve a substantially uniform protein purity. The expressed fusion protein can also be purified using an affinity chromatography column containing a specific antibody, receptor or ligand of the fusion protein. Depending on the nature of the affinity column used, the fusion polypeptide bound to the affinity column can be eluted using conventional methods such as high salt buffer, pH change, and the like.
药物组合物Pharmaceutical composition
本发明还提供了一种药物组合物,它含有有效剂量的(160~360μg/kg)本发明所述融合蛋白,以及药学上可接受的载体。通常,可将有效量的本发明融合蛋白配制于无毒的、惰性的和药学上可接受的水性载体介质中,其中pH通常约为5-8,较佳地,pH约为6-8。术语“有效量”或“有效剂量”是指可对人和/或动物产生功能或活性的且可被人和/或动物所接受的量。“药学上可接受的”的成分是适用于人和/或哺乳动物而无过度不良副反应(如毒性、刺激和变态反应)的,即具有合理的效益/风险比的物质。术语“药学上可接受的载体”指用于治疗剂给药的载体,包含各种辅形剂和稀释剂。 The invention also provides a pharmaceutical composition comprising an effective amount (160-360 [mu]g/kg) of a fusion protein of the invention, and a pharmaceutically acceptable carrier. Generally, an effective amount of a fusion protein of the invention can be formulated in a non-toxic, inert, and pharmaceutically acceptable aqueous carrier medium wherein the pH is usually from about 5 to about 8, preferably, the pH is from about 6 to about 8. The term "effective amount" or "effective amount" refers to an amount that is functional or active to a human and/or animal and that is acceptable to humans and/or animals. A "pharmaceutically acceptable" ingredient is one which is suitable for use in humans and/or mammals without excessive adverse side effects (such as toxicity, irritation, and allergies), i.e., materials having a reasonable benefit/risk ratio. The term "pharmaceutically acceptable carrier" refers to a carrier for the administration of a therapeutic agent, which comprises various excipients and diluents.
药学上可接受的载体包含(但并不限于):盐水、缓冲液、葡萄糖、水、甘油、乙醇、及其组合。通常药物制剂应与给药方式相匹配,本发明的药物组合物可以被制成针剂形式,例如用生理盐水或含有葡萄糖和其他辅剂的水溶液通过常规方法进行制备。所述的药物组合物宜在无菌条件下制造。活性成分的给药量是治疗有效量。本发明的药物制剂还可制成缓释制剂。Pharmaceutically acceptable carriers include, but are not limited to, saline, buffer, dextrose, water, glycerol, ethanol, and combinations thereof. Usually, the pharmaceutical preparation should be matched to the mode of administration, and the pharmaceutical composition of the present invention can be prepared into an injection form, for example, by a conventional method using physiological saline or an aqueous solution containing glucose and other adjuvants. The pharmaceutical composition is preferably manufactured under sterile conditions. The amount of active ingredient administered is a therapeutically effective amount. The pharmaceutical preparation of the present invention can also be formulated into a sustained release preparation.
本发明所述的融合蛋白的有效量可随给药的模式和待治疗的疾病的严重程度等而变化。优选的有效量的选择可以由本领域普通技术人员根据各种因素来确定(例如通过临床试验)。所述的因素包含但不限于:所述的融合蛋白的药代动力学参数例如生物利用率、代谢、半衰期等;患者所要治疗的疾病的严重程度、患者体重、患者免疫状况、给药途径等。The effective amount of the fusion protein of the present invention may vary depending on the mode of administration and the severity of the disease to be treated and the like. The selection of a preferred effective amount can be determined by one of ordinary skill in the art based on various factors (e.g., by clinical trials). The factors include, but are not limited to, the pharmacokinetic parameters of the fusion protein such as bioavailability, metabolism, half-life, etc.; the severity of the disease to be treated by the patient, the patient's weight, the patient's immune status, the route of administration, etc. .
附图说明DRAWINGS
图1、根据本发明实施例在PCDNA3表达载体内SpeI-EcoRI(酶切位点以下划线__标注)片段的FP-A的核苷酸序列及推导的氨基酸序列。人FVII由信号肽(1-38,以下划线__标注)和成熟FVII蛋白(39-444)构成。成熟的融合蛋白含有hFVII(39-444)、柔性肽接头(445-471,以下划线__标注)、CTP刚性单元(472-499,以下划线__标注)和Fc变异体(500-722)。Figure 1. Nucleotide sequence and deduced amino acid sequence of FP-A fragment of SpeI-EcoRI (labeled with underlined__) in the PCDNA3 expression vector according to an embodiment of the present invention. Human FVII consists of a signal peptide (1-38, underlined __) and a mature FVII protein (39-444). Mature fusion proteins contain hFVII (39-444), flexible peptide linkers (445-471, underlined __), CTP rigid units (472-499, underlined __) and Fc variants (500-722) .
图2、FP-A经MabSelect层析SEC检测结果。Figure 2. FP-A results by MabSelect chromatography SEC.
图3、FP-A经CHT层析分离目的组分P3SEC检测结果。Figure 3. FP-A separation of the target component P3SEC by CHT chromatography.
图4、FP-A经MabSelect分离组分和经CHT层析分离组分P1、P2和P3的SDS-PAGE电泳图。Figure 4. SDS-PAGE electropherograms of FP-A separated components by MabSelect and fractionated P1, P2 and P3 by CHT chromatography.
图5、FP-A经Q-HP层析洗脱组分P3的SEC检测结果。Figure 5. SEC detection results of FP-A eluted fraction P3 by Q-HP chromatography.
图6、FP-A经Superdex 200层析洗脱组分P3-3SEC检测结果。Figure 6. FP-A was analyzed by Superdex 200 chromatography elution component P3-3SEC.
图7、P3-3组分激活40h的SEC检测结果。Figure 7. The SEC detection results of the P3-3 component activated for 40 h.
图8、P3-3组分激活16~40h后还原SDS-PAGE电泳检测结果。Figure 8. The results of SDS-PAGE electrophoresis were confirmed after activation of P3-3 components for 16-40 hours.
图9、小鼠出血时间结果统计图。注:*p<0.01,***p<0.001。Figure 9. Statistical graph of mouse bleeding time results. Note: * p < 0.01, *** p < 0.001.
图10、小鼠出血量结果统计图。注:**p<0.01,***p<0.001。Figure 10. Statistical graph of mouse bleeding volume results. Note: ** p < 0.01, *** p < 0.001.
图11、HemA小鼠等活性给予FP-A和诺其1h和2h后出血时长比较。注:与HA-N-1h组相比,*P<0.05,***P<0.01;与C57-NS组相比,#P<0.05,###P <0.01。Figure 11. Comparison of the duration of bleeding after administration of FP-A and Nobel for 1 h and 2 h in HemA mice. Note: compared with HA-N-1h group, * P <0.05, *** P <0.01; compared with C57-NS group, # P <0.05, ### P <0.01.
图12、FP-A及诺其在华法林大鼠上的活性半衰期。Figure 12. Activity half-life of FP-A and Novo in warfarin rats.
具体实施方式detailed description
实施例1.构建编码FVII融合蛋白的表达质粒Example 1. Construction of an expression plasmid encoding a FVII fusion protein
编码FVII前导肽、成熟蛋白、柔性肽接头、CTP刚性单元和人IgG vFc变体的基因序列是人工优化过的CHO细胞偏爱密码子,经人工合成获得。所合成融合蛋白全长DNA片段的5’和3’端各有一个限制性酶内切位点,分别为SpeI和EcoRI,全长DNA片段插入至pUC57转移载体相应酶切位点间,并由DNA测序验证序列。然后再将上述获得的融合蛋白全长基因片段从中间载体转移到以PCDNA3.1为模板并改造后的表达质粒PTY1A1的SpeI(5’)和EcoRI(3’)位点间,得到融合蛋白高表达质粒。PTY1A1质粒包含但不限于以下重要表达元器件:1)人巨细胞病毒早期启动子和哺乳动物细胞外源高表达所需增强子;2)双重筛选标记物,在细菌中具有卡那霉素抗性,在哺乳动物细胞中具有G418抗性;3)鼠二氢叶酸还原酶(DHFR)基因表达框,当宿主细胞为DHFR基因缺陷型时,氨甲蝶呤(MTX)能共扩增融合基因和DHFR基因(参见美国专利US 4,399,216)。再将融合蛋白表达质粒转染入哺乳动物宿主细胞系,为了获得稳定高水平的表达,优选的宿主细胞系是DHFR酶缺陷型CHO-细胞(参见美国专利US 4,818,679)。转染两天后,将培养基换成含0.6mg/mL G418的筛选培养基,细胞以一定浓度(5000-10000个活细胞/孔)种植在96孔培养板里,培养10-14天直至大的离散细胞克隆出现。用ELISA分析方法,筛选对选择用药具有抗性的转染子。通过极限稀释96孔培养板,亚克隆产生高水平融合蛋白的孔。The gene sequence encoding the FVII leader peptide, mature protein, flexible peptide linker, CTP rigid unit and human IgG vFc variant is a manually optimized CHO cell preferred codon obtained by artificial synthesis. The full-length DNA fragment of the synthetic fusion protein has a restriction endonuclease site at the 5' and 3' ends, respectively, SpeI and EcoRI, and the full-length DNA fragment is inserted into the corresponding cleavage site of the pUC57 transfer vector, and DNA sequencing verification sequence. Then, the full-length gene fragment of the fusion protein obtained above was transferred from the intermediate vector to the SpeI (5') and EcoRI (3') sites of the expression plasmid PTY1A1 using PCDNA3.1 as a template to obtain a high fusion protein. Expression plasmid. The PTY1A1 plasmid includes, but is not limited to, the following important expression components: 1) human cytomegalovirus early promoter and mammalian cells are required for exogenous high expression; 2) dual screening markers with kanamycin resistance in bacteria Sexuality, G418 resistance in mammalian cells; 3) Murine dihydrofolate reductase (DHFR) gene expression cassette, when the host cell is DHFR gene-deficient, methotrexate (MTX) can amplify the fusion gene And the DHFR gene (see U.S. Patent 4,399,216). The fusion protein expression plasmid is then transfected into a mammalian host cell line, and in order to obtain stable high levels of expression, the preferred host cell line is a DHFR enzyme deficient CHO-cell (see U.S. Patent 4,818,679). Two days after transfection, the medium was changed to a screening medium containing 0.6 mg/mL G418, and the cells were planted in a 96-well culture plate at a concentration (5000-10000 viable cells/well) for 10-14 days until large. Discrete cell clones appear. Transfectants resistant to the selected drug were screened by ELISA assay. Subclones were generated to produce high levels of fusion protein wells by limiting dilution of 96-well plates.
如下表所示,本发明构建了一系列rhFVII融合蛋白,它含有不同长度的肽接头(Linker)、不同组成的CTP刚性单元以及几种不同亚型的IgG Fc变体(vFc)元件组成。为了验证含有至少1个,并且不同长度的CTP刚性单元均能显著提高融合蛋白的活性,我们构建了融合蛋白FP-A、FP-B、FP-C、FP-D和FP-E;其中,FP-A的氨基酸及编码核苷酸如图1所示。为了验证CTP刚性单元对融合蛋白活性的重要性,我们还同时构建了不含CTP刚性单元的两种Fc融合蛋白FP-G和FP-H,表达质粒构建方法同上。此外,我们还构建了CTP位于Fc C端的FP-F, 以验证CTP刚性单元位置的重要性。详见表1。各组成元件的氨基酸序列见序列表。As shown in the table below, the present invention constructs a series of rhFVII fusion proteins comprising peptide linkers of different lengths (Linker), CTP rigid units of different compositions, and IgG Fc variant (vFc) elements of several different subtypes. In order to verify that CTP rigid units containing at least one and different lengths can significantly increase the activity of the fusion protein, we constructed fusion proteins FP-A, FP-B, FP-C, FP-D and FP-E; The amino acid and coding nucleotides of FP-A are shown in Figure 1. In order to verify the importance of the CTP rigid unit for the activity of the fusion protein, we also constructed two Fc fusion proteins FP-G and FP-H without CTP rigid unit, and the expression plasmid construction method was the same as above. In addition, we also constructed the FP-F with CTP at the Fc C end. To verify the importance of the location of the CTP rigid unit. See Table 1 for details. The amino acid sequence of each constituent element is shown in the sequence listing.
表1、各种FVII融合蛋白组成Table 1. Composition of various FVII fusion proteins
Figure PCTCN2016106012-appb-000007
Figure PCTCN2016106012-appb-000007
实施例2.瞬时表达各融合蛋白和活性测定Example 2. Transient expression of each fusion protein and activity assay
将实施例1得到的8种表达质粒,在30ml的摇瓶里使用DNAFect LT试剂TM(ATGCell公司)转染3×107CHO-K1细胞,经转染的细胞在含有1000ng/ml维生素K1的无血清生长培养基中生长5天,测定上清液中融合蛋白的浓度,并用实施例7或8中描述的方法测定其活性。ELISA结果显示8种质粒在该条件下的瞬时表达量(物质的量)相似,但是它们凝血活性却显示出较大差别。其中,我们将FP-A的摩尔比活性定义为100%。FP-F、FP-G和FP-H质粒表达的融合蛋白上清液活性较低,仅为FP-A的29.4%、26.3%和41.2%,纯化蛋白用SDS电泳分析,显示这三种融合蛋白部分呈不同程度的聚合现象;而FP-B、FP-C、FP-D和FP-E的活性分别为FP-A的98.0%、89.2%、86.1%和92.9%。表明仅靠延长肽接头并不能有效改善融合蛋白的活性,即彼此的空间位阻效应并不能随着柔性肽接头的延伸而显著削弱。甚至,过长的肽接头不仅不能提高融合蛋白的活性,反而会使蛋白错误折叠,以无活性的多聚体形式分泌(FP-H发酵液中产物多为聚合体)。我们推测原因,过长的柔性肽接头,给了FVIIa更高的灵活度,使其能相对Fc自由转动,这可能使FVIIa的立体结构更加靠近Fc区,而在二者之间加入CTP刚性单元,一方面相当增加了一段刚性肽接头,使彼此远离,更重要的是CTP刚性单元含多个糖基侧链,相对于柔性肽接头的无规则卷曲形态,CTP刚性单元可以形成固定的空间构象,能够有效地分开融合蛋白的不同功能区,这 更有利于两部分独立折叠成正确的三维构象,保持了较高的活性。我们通过FP-A和FP-F的活性比较验证了这种推测的正确性,即CTP刚性单元置于Fc C端的FP-F的活性仅为CTP刚性单元置于Fc N端的FP-A的29.4%,FP-F的活性与不含CTP刚性单元的FP-G相似。以上结果,证实CTP刚性单元对融合蛋白的活性极为关键,并且CTP刚性单元置于Fc的N端能有效提高融合蛋白的活性。8 kinds of expression plasmids obtained in Example 1 was used DNAFect LT reagent TM (ATGCell Company) in 30ml shake bottle transfection 3 × 10 7 CHO-K1 cells, the transfected cells were cultured at 1000ng / ml of vitamin K1 The growth was carried out for 5 days in serum-free growth medium, and the concentration of the fusion protein in the supernatant was measured, and its activity was measured by the method described in Example 7 or 8. The ELISA results showed that the transient expression levels (quantity of the substances) of the eight plasmids under these conditions were similar, but their coagulation activities showed a large difference. Among them, we defined the molar activity of FP-A as 100%. The fusion protein supernatants expressed by FP-F, FP-G and FP-H plasmids were less active, only 29.4%, 26.3% and 41.2% of FP-A. The purified proteins were analyzed by SDS electrophoresis, showing these three fusions. The protein fractions showed different degrees of polymerization; while the activities of FP-B, FP-C, FP-D and FP-E were 98.0%, 89.2%, 86.1% and 92.9%, respectively, of FP-A. It was shown that elongation of the peptide linker alone did not effectively improve the activity of the fusion protein, i.e., the steric hindrance effect of each other was not significantly attenuated with the extension of the flexible peptide linker. Even an excessively long peptide linker not only does not increase the activity of the fusion protein, but instead causes the protein to fold in error and is secreted as an inactive multimer (the product in the FP-H fermentation broth is mostly a polymer). We speculate that the reason for the long flexible peptide linker gives FVIIa a higher degree of flexibility, allowing it to rotate freely relative to Fc, which may bring the steric structure of FVIIa closer to the Fc region, and add a CTP rigid unit between the two. On the one hand, a rigid peptide linker is added to make it away from each other. More importantly, the CTP rigid unit contains multiple glycosyl side chains. The CTP rigid unit can form a fixed spatial conformation with respect to the random coil form of the flexible peptide linker. It can effectively separate the different functional regions of the fusion protein, which is more conducive to the two parts independently folded into the correct three-dimensional conformation, maintaining a high activity. We verified the validity of this speculation by comparing the activity of FP-A and FP-F, that is, the activity of FP-F in which CTP rigid unit is placed at Fc C end is only C9.4 rigid unit placed at Fc N end of FP-A 29.4 %, FP-F is similar to FP-G without CTP rigid units. The above results confirmed that the CTP rigid unit is critical for the activity of the fusion protein, and that the CTP rigid unit is placed at the N-terminus of the Fc to effectively increase the activity of the fusion protein.
实施例3.筛选高表达融合蛋白的稳定转染细胞系Example 3. Screening for stable transfected cell lines with high expression of fusion protein
将上述FP-A、FP-B、FP-C、FP-D和FP-E的表达质粒转染入哺乳动物宿主细胞系,以表达FVII融合蛋白。为了维持稳定的高水平表达,优选的宿主细胞是DHFR缺陷型的CHO细胞(美国专利No.4818679)。一种优选转染方法是电穿孔,也可以使用其它方法,包括磷酸钙共沉降、脂质体转染和微注射等。电穿孔方法应用设置为300V电压和1050μFd电容的Gene Pulser Electroporator(Bio-Rad Laboratories公司),放置在比色杯内的2~3×107个细胞中加入50μg PvuI线性化的表达质粒,电穿孔后的细胞转移至含30ml生长培养基的摇瓶中。转染两天后,将培养基换成含0.6mg/mL G418的生长培养基,细胞以一定浓度种植在96孔培养板里,培养10-12天直至大的离散细胞克隆出现。用抗人IgG Fc的ELISA分析方法,筛选对选择用药具有抗性的转染子,也可用抗FVII的ELISA分析方法进行融合蛋白表达的定量测定,然后通过极限稀释法亚克隆产生高水平表达融合蛋白的孔。The expression plasmids of FP-A, FP-B, FP-C, FP-D and FP-E described above were transfected into a mammalian host cell line to express the FVII fusion protein. In order to maintain a stable high level of expression, a preferred host cell is a DHFR deficient CHO cell (U.S. Patent No. 4,818,679). A preferred method of transfection is electroporation, and other methods can be used, including calcium phosphate co-precipitation, lipofection, and microinjection. Electroporation method Using a Gene Pulser Electroporator (Bio-Rad Laboratories) set to 300 V voltage and 1050 μFd capacitance, 50 μg of PvuI linearized expression plasmid was added to 2 to 3 × 10 7 cells in a cuvette, and electroporation was performed. The resulting cells were transferred to shake flasks containing 30 ml of growth medium. Two days after transfection, the medium was changed to a growth medium containing 0.6 mg/mL G418, and the cells were seeded in a 96-well culture plate at a concentration for 10-12 days until large discrete cell clones appeared. The anti-human IgG Fc ELISA method is used to screen the transfectants that are resistant to the selected drugs, and the anti-FVII ELISA method can also be used for the quantitative determination of the fusion protein expression, and then the high-level expression fusion is produced by sub-cloning by limiting dilution method. The pores of the protein.
为了实现融合蛋白较高水平的表达,宜用受MTX药物抑制的DHFR基因进行共扩增。在含有递增浓度MTX的生长培养基中,用DHFR基因共扩增转染的融合蛋白基因。极限稀释DHFR表达阳性的亚克隆,逐步加压并筛选出能在高达6μM MTX培养基中生长的转染子,测定其分泌率,筛选出高表达外源蛋白的细胞系。将分泌率超过约3(较佳地约5)IU/106(即百万)个细胞/24小时的细胞系使用无血清培养基进行适应性悬浮培养,然后再用条件培养基纯化融合蛋白。In order to achieve a higher level of expression of the fusion protein, it is preferred to co-amplify with the DHFR gene which is inhibited by the MTX drug. The transfected fusion protein gene was co-amplified with the DHFR gene in growth medium containing increasing concentrations of MTX. Subclones with positive dilution DHFR expression were gradually pressurized, and transfectants capable of growing in up to 6 μM MTX medium were screened, the secretion rate was determined, and a cell line highly expressing the foreign protein was selected. A cell line having a secretion rate of more than about 3 (preferably about 5) IU/10 6 (i.e., millions) of cells per 24 hours is subjected to adaptive suspension culture using serum-free medium, and then the fusion protein is purified using conditioned medium. .
实施例4.生产融合蛋白Example 4. Production of fusion protein
将实施例3优选得到的高产量细胞株首先在培养皿中进行无血清驯化培养, 然后转移到摇瓶中进行悬浮驯化培养。待细胞适应这些培养条件后,然后在300ml摇瓶中进行补料流加培养或通过每天更换培养基的办法模拟灌流培养。由实施例3筛选得到的生产融合蛋白FP-A的CHO衍生的细胞株在300ml体积的摇瓶中补料流加培养14天,其表达的重组融合蛋白累积产量为310mg/L,活细胞密度最高可达到15×106个/mL。为了得到更多融合蛋白,也可以选用1000ml摇瓶培养。另一种培养方法,上述CHO衍生的细胞株在100ml体积的摇瓶中每天更换培养基,其表达的重组融合蛋白每天累积产量约为50mg/L,在摇瓶中活细胞密度最高可达到25×106个/mL。以上两种方法生产的重组融合蛋白的生物学活性相当。The high-yield cell line preferably obtained in Example 3 was first subjected to serum-free domestication culture in a culture dish, and then transferred to a shake flask for suspension and domestication culture. After the cells were adapted to these culture conditions, supplemental flow culture was then carried out in a 300 ml shake flask or perfusion culture was simulated by changing the medium daily. The CHO-derived cell line producing the fusion protein FP-A obtained by the screening of Example 3 was fed and cultured for 14 days in a 300 ml volume shake flask, and the cumulative yield of the expressed recombinant fusion protein was 310 mg/L, and the viable cell density was obtained. Up to 15 × 10 6 / mL. In order to obtain more fusion proteins, it can also be cultured in a 1000 ml shake flask. In another culture method, the above CHO-derived cell strain is changed daily in a 100 ml volume shake flask, and the recombinant fusion protein expressed has a cumulative yield of about 50 mg/L per day, and the viable cell density can reach up to 25 in a shake flask. ×10 6 / mL. The biological activities of the recombinant fusion proteins produced by the above two methods are comparable.
实施例5.纯化与定性融合蛋白Example 5. Purification and Qualitative Fusion Proteins
本实施例中以FP-A为例,具体描述上述实施例获得的几种融合蛋白纯化步骤及方法,FP-B、FP-C、FP-D和FP-E方法相同,实施例中不再赘述。In this embodiment, FP-A is taken as an example to describe the purification steps and methods of several fusion proteins obtained in the above embodiments, and the FP-B, FP-C, FP-D and FP-E methods are the same, and the examples are no longer Narration.
本发明主要采用四步层析法对融合蛋白进行纯化。分别为亲和层析、多维模式层析、阴离子交换层析和分子筛层析。The present invention mainly uses four-step chromatography to purify the fusion protein. Affinity chromatography, multidimensional mode chromatography, anion exchange chromatography and molecular sieve chromatography, respectively.
第一步,亲和层析使用发明内容部分所述的层析介质进行样品捕获。首先使用平衡buffer:20-50mM Tris-HCl,150-500mM NaCl,pH6.8-7.2,平衡层析柱3-5个柱体积(CV);经过澄清后的发酵液上样,载量不高于30g/L;使用平衡buffer:20-50mM Tris-HCl,150-500mM NaCl,pH6.8-7.2,平衡层析柱3-5个柱体积(CV),冲洗未结合的组份;使用去污buffer:20mM Tris,1.5-2M NaCl,0.5-2M尿素,5-10mM(EDTA)2Na,pH6.5-7.0冲洗层析柱3-5个柱体积,去除部分污染物;使用平衡buffer:20-50mM Tris-HCl,150-500mM NaCl,pH6.8-7.2,平衡层析柱3-5个柱体积(CV)后;使用洗脱buffer:100mM Gly-HCl,150-300mM NaCl,pH3.0-3.5洗脱目标产物,直接加入到1M Tris,pH8.0中,中和pH值至中性。In the first step, affinity chromatography uses the chromatographic medium described in the Summary of the Invention for sample capture. First use balanced buffer: 20-50mM Tris-HCl, 150-500 mM NaCl, pH 6.8-7.2, equilibrate the column 3-5 column volumes (CV); after clarified fermentation broth loading, the load is not high At 30 g/L; equilibrate the column with 3-5 column volumes (CV) using a balanced buffer: 20-50 mM Tris-HCl, 150-500 mM NaCl, pH 6.8-7.2, and rinse the unbound components; Stain buffer: 20 mM Tris, 1.5-2 M NaCl, 0.5-2 M urea, 5-10 mM (EDTA) 2 Na, pH 6.5-7.0 Wash the column 3-5 column volumes to remove some contaminants; use balanced buffer: 20-50 mM Tris-HCl, 150-500 mM NaCl, pH 6.8-7.2, equilibrated column after 3-5 column volumes (CV); using elution buffer: 100 mM Gly-HCl, 150-300 mM NaCl, pH 3. The target product was eluted 0-3.5, directly added to 1 M Tris, pH 8.0, and neutralized to pH neutral.
融合蛋白经第一步Mabselect亲和层析纯化后的SEC分析结果如图2所示。The SEC analysis results of the fusion protein purified by the first Mabselect affinity chromatography are shown in Fig. 2.
第二步,多维模式层析使用发明内容部分所述的Bio-Rad公司的CHT陶瓷羟磷灰石骨架的层析介质进行样品捕获。首先使用平衡buffer:20-50mM PB,pH6.8-7.2,平衡层析柱3-5个柱体积(CV);经过亲和层析捕获的样品上样,载量不高于10g/L;使用平衡buffer:20-50mM PB,pH6.8-7.2,平衡层析柱3-5 个柱体积(CV),冲洗未结合的组份,命名为P1;使用洗脱buffer 1:120mM PB,pH6.8-7.2冲洗层析柱3-5个柱体积,收集洗脱组分,命名为P2;使用洗脱buffer 2:200mM PB,pH6.8-7.2冲洗层析柱3-5个柱体积,收集洗脱组分,命名为P3。分离得到的P1、P2和P3组分分别激活后,体外测定其比活性分别为103.2、3026.7和5705.7IU/mg,说明不同洗脱组分之间存在差异,体现了羧基化程度的差异。In the second step, multi-dimensional mode chromatography was used for sample capture using the chromatographic medium of the CHT ceramic hydroxyapatite skeleton of Bio-Rad Company described in the Summary of the Invention. First, use balanced buffer: 20-50mM PB, pH 6.8-7.2, equilibrate the column 3-5 column volumes (CV); sample captured by affinity chromatography, the loading is not higher than 10g / L; Use balanced buffer: 20-50mM PB, pH 6.8-7.2, equilibration column 3-5 Column volume (CV), rinse unbound components, named P1; rinse the column 3-5 column volumes using elution buffer 1:120 mM PB, pH 6.8-7.2, collect the eluted fractions, name P2; rinse the column 3-5 column volumes using elution buffer 2: 200 mM PB, pH 6.8-7.2, and collect the eluted fraction, designated P3. After the isolated P1, P2 and P3 components were activated, the specific activities were determined to be 103.2, 3026.7 and 5705.7 IU/mg, respectively, indicating that there were differences between the different eluting components, indicating the difference in the degree of carboxylation.
融合蛋白FP-A经第二步CHT层析分离目的组分P3的SEC检测结果如图3所示。MabSelect分离组分和CHT层析分离组分P1、P2和P3的SDS-PAGE电泳结果见图4。The SEC detection result of the fusion protein FP-A by separating the target component P3 by the second step CHT chromatography is shown in FIG. The SDS-PAGE electrophoresis results of the MabSelect separation component and the CHT chromatographic separation components P1, P2 and P3 are shown in Fig. 4.
第三步,阴离子交换层析使用发明内容部分所述的层析介质进一步分离纯化。使用平衡Buffer:20-50mM Tris-HCl,pH7.5-8.0平衡层析柱3-5个柱体积;CHT层析的P3洗脱组份上样,载量不高于30g/L;使用平衡Buffer:20-50mM Tris-HCl,pH7.5-8.0平衡层析柱3-5个柱体积;使用线性梯度洗脱方式洗脱目标组份,洗脱buffer:20mM Tris-HCl,500mM NaCl,pH7.5-8.0,洗脱的流速为60cm/h,梯度为25-30个柱体积。结果显示,色谱峰为单峰,我们收集的部分为最高紫外吸收的1/3以上的部分,这部分的SEC检测结果见图5。In the third step, anion exchange chromatography is further separated and purified using the chromatographic medium described in the Summary of the Invention. Balanced column with 3-5 column volumes using balanced Buffer: 20-50 mM Tris-HCl, pH 7.5-8.0; loading of P3 eluting components of CHT chromatography with a loading of no more than 30 g/L; Buffer: 20-50 mM Tris-HCl, pH 7.5-8.0 equilibrium chromatography column 3-5 column volumes; elute the target component using linear gradient elution, elution buffer: 20 mM Tris-HCl, 500 mM NaCl, pH 7 .5-8.0, elution flow rate of 60 cm / h, gradient of 25-30 column volumes. The results showed that the chromatographic peak was a single peak, and the fraction we collected was more than 1/3 of the highest UV absorption. The SEC results of this part are shown in Figure 5.
第四步,分子筛层析使用发明内容部分所述的层析介质进行分离,首先用平衡buffer:100mM Hepes,100mM NaCl,pH8.0平衡层析柱1.5-2个柱体积;上样量不高于柱体积的2%;使用平衡buffer:100mM Hepes,100mM NaCl,pH8.0,以30cm/h的流速洗脱,放弃前段的聚合体峰,收集单体峰的上升段、中段和下降段(以色谱峰的最高紫外吸收的1/2为限),分别命名为P3-2、P3-3、P3-4。其中P3-3组分的SEC检测结果见图6。In the fourth step, molecular sieve chromatography is carried out by using the chromatographic medium described in the section of the invention. First, the column is equilibrated with a balance buffer: 100 mM Hepes, 100 mM NaCl, pH 8.0, and the column volume is 1.5-2 column volumes; the sample loading is not higher than the column. 2% by volume; using a balanced buffer: 100 mM Hepes, 100 mM NaCl, pH 8.0, eluting at a flow rate of 30 cm/h, abandoning the polymer peak in the previous stage, collecting the rising, middle and descending phases of the monomer peak (by chromatography The peak of the highest UV absorption is limited to 1/2, named P3-2, P3-3, and P3-4. The SEC test results of the P3-3 component are shown in Fig. 6.
实施例6.融合蛋白体外激活Example 6. In vitro activation of fusion proteins
本发明通过溶液孵育自激活法对融合蛋白进行激活。将分子筛层析分离得到的融合蛋白单一组分P3-3加载于30kDa超滤浓缩管(Corning,货号431489),然后以台式高速低温离心机(Eppendorf,型号5810R)离心,更换缓冲液为20mM PB,0.3M NaCl,5mM CaCl2,pH7.0-7.2,并浓缩至蛋白浓度为3-5mg/ml(采用消光系数法定量),置于4℃进行激活,激活时间16~40小时。激活结束利用G-25脱盐柱,将其置换到buffer:20mM PB,pH7.0中,-80℃冻存备用。 The present invention activates the fusion protein by a solution incubation self-activation method. The single component P3-3 of the fusion protein obtained by molecular sieve chromatography was loaded into a 30 kDa ultrafiltration concentrating tube (Corning, Cat. No. 431489), and then centrifuged in a benchtop high-speed cryogenic centrifuge (Eppendorf, model 5810R) to change the buffer to 20 mM PB. , 0.3 M NaCl, 5 mM CaCl 2 , pH 7.0-7.2, and concentrated to a protein concentration of 3-5 mg/ml (quantified by extinction coefficient method), activated at 4 ° C, activation time 16 to 40 hours. At the end of the activation, the G-25 desalting column was used, and it was replaced with a buffer: 20 mM PB, pH 7.0, and stored at -80 ° C for use.
采用本发明实施例7或8所述生物学活性测定法和还原性SDS-PAGE检测层析分离组分体外激活不同时间的激活程度。不同激活时间的生物学活性结果见表2;P3-3组分在激活过程中取样,16~40h激活产物的还原型SDS-PAGE检测结果见图8,及其激活40小时的SEC检测结果见图7。可知,P3-3经40小时激活的活化程度最高。SDS-PAGE还原胶电泳显示FP-A经实施例5中四步层析分离获得的P3-3组分被激活后主要呈现两条主带,分别是约74.3KDa的HC-L-CTP-Fc(与FVIIa重链相连的柔性肽接头、CTP刚性单元和IgG Fc段部分)和约24.0KDa的LC(FVIIa的轻链部分)。The biological activity assay described in Example 7 or 8 of the present invention and the reducing SDS-PAGE were used to detect the degree of activation of the chromatographic fractions in vitro for different times. The biological activity results of different activation times are shown in Table 2; the P3-3 components were sampled during the activation process, and the results of the reduced SDS-PAGE of the activated products from 16 to 40 hours are shown in Figure 8, and the SEC detection results after 40 hours of activation are shown. Figure 7. It can be seen that P3-3 has the highest degree of activation after 40 hours of activation. SDS-PAGE gel electrophoresis showed that the P3-3 fraction obtained by the four-step chromatography in FP-A was mainly activated by two main bands, which were about 74.3KDa HC-L-CTP-Fc. (Flexible peptide linker, CTP rigid unit and IgG Fc segment portion linked to the FVIIa heavy chain) and LC of about 24.0 KDa (light chain portion of FVIIa).
表2、P3-3组分激活16-40h的生物学活性检测结果Table 2, P3-3 component activation 16-40h biological activity test results
Figure PCTCN2016106012-appb-000008
Figure PCTCN2016106012-appb-000008
实施例7.生色底物法间接测定融合蛋白的生物学活性Example 7. Indirect determination of biological activity of fusion protein by chromogenic substrate method
本发明采用HYPHEN BioMed公司生产的BIOPHEN FVII生色试剂盒(Ref:A221304)测定由实施例6所示方法激活的各融合蛋白的体外酶活性。该试剂盒测定原理为生色底物法,FVIIa是一种在外源性凝血途径起作用的丝氨酸蛋白酶,当FVIIa与组织因子结合后,在磷脂和Ca2+存在条件下,可激活凝血因子FX,使其转变为活性形式FXa。待测定的融合蛋白首先与来源于兔的促凝血酶原激酶的组织因子形成酶复合物,然后激活反应体系中一定浓度的(过量)凝血因子FX,使其转化为活性形式FXa,FXa作用于反应体系中特异性显色底物SXa-11,使底物裂解并产生pNA,所产生pNA量直接与FXa的活性成正相关性。在405nm处用比色计测定所释放的pNA浓度,即可知测试样本中FVIIa以及FXa活性之间的对应关系,以此计算出FVIIa的活性大小,用正常人的血浆作为标准品。In the present invention, the in vitro enzymatic activity of each fusion protein activated by the method shown in Example 6 was measured using a BIOPHEN FVII chromogenic kit (Ref: A221304) manufactured by HYPHEN BioMed. The kit is based on the chromogenic substrate method. FVIIa is a serine protease that acts on the exogenous coagulation pathway. When FVIIa binds to tissue factor, it activates clotting factor FX in the presence of phospholipids and Ca 2+ . To convert it to the active form FXa. The fusion protein to be determined first forms an enzyme complex with a tissue factor derived from rabbit thromboplastin, and then activates a certain concentration (excess) of factor FX in the reaction system to convert it into an active form of FXa, which acts on FXa. The specific chromogenic substrate SXa-11 in the reaction system cleaves the substrate and produces pNA, and the amount of pNA produced directly correlates with the activity of FXa. The concentration of FVIIa and FXa activity in the test sample was determined by measuring the concentration of pNA released at 405 nm with a colorimeter, thereby calculating the activity of FVIIa, using normal human plasma as a standard.
实施例8.凝血法直接测定融合蛋白的生物学活性Example 8. Direct determination of the biological activity of a fusion protein by coagulation
凝血法测定FVIIa生物学活性是通过纠正FVIIa因子缺乏血浆所导致凝固时间延长的能力而获得的。采用法国STAGO公司生产的试剂盒
Figure PCTCN2016106012-appb-000009
-Deficient FVII(Cat.No.00743)。检测方法首先是将稀释的已知FVII活性的正常人冻干血浆(Unicalibrator,Cat.No.00625)与乏VII基质血浆混合,测定凝血酶原时间 (PT),建立标准曲线,再将待测血浆适度稀释后与乏FVII基质血浆混合,进行PT测定。通过标准曲线所拟合的活性百分比C(%)与PT时间t(s)的对数方程,可测得待测样本FVIIa的活性多少,其结果用正常血浆的百分比来表示(%),试剂盒中所提供的标准品百分比活性(%)与国际酶活单位IU的对应关系为100%=1IU,据此可求算出待测样品FVII的酶比活性大小,单位为IU/mg。
Determination of the biological activity of FVIIa by coagulation is obtained by correcting the ability of FVIIa-factor-deficient plasma to cause prolonged clotting time. Using the kit produced by STAGO, France
Figure PCTCN2016106012-appb-000009
- Deficient FVII (Cat. No. 00743). The detection method firstly mixes diluted human lyophilized plasma of known FVII activity (Unicalibrator, Cat. No. 00625) with VII matrix plasma, determines prothrombin time (PT), establishes a standard curve, and then tests The plasma was moderately diluted and mixed with spent FVII matrix plasma for PT assay. The logarithmic equation of the activity percentage C (%) and the PT time t(s) fitted by the standard curve can be used to measure the activity of the sample FVIIa, and the result is expressed as a percentage of normal plasma (%). The correspondence between the percentage activity (%) of the standard provided in the cartridge and the international enzyme activity unit IU is 100%=1 IU, and accordingly, the specific activity of the enzyme of the sample FVII to be tested can be calculated, and the unit is IU/mg.
各层析步骤分离组分体外激活24h的生物活性测定结果见表3。其中,四步层析最终获得的目的组分P3-3激活16-40h的活性数据见表2。经纯化获得的FP-A有效单一组分P3-3激活40h的生物学活性为22470IU/mg,换算为摩尔比活性约为4417IU/nM(每个融合蛋白含有2个FVIIa,相当于2208.7IU/nM FVIIa),与已上市的重组FVIIa——诺其(2511IU/nM FVIIa)的活性相当,说明本发明提供的融合蛋白,其C端融合的Fc对FVIIa的活性影响极小。其活性同时也显著高于其他已报道的重组FVIIa的活性,例如,CSL Behring公司报道的HEK细胞生产的重组hFVII生物学活性为2874IU/mg,相当于144IU/nM。而其用HEK或CHO细胞生产的融合蛋白hFVII-FP的生物学活性为620-770IU/mg,相当于69-75IU/nM(Weimer T等,Thromb Haemost,2008,99:659-667)。The bioactivity assay results of the separation components in each chromatographic step for 24 h in vitro are shown in Table 3. Among them, the activity data of the target component P3-3 finally obtained by four-step chromatography activated for 16-40 hours is shown in Table 2. The purified FP-A effective single component P3-3 has a biological activity of 22470 IU/mg for 40 h, and a molar activity of about 4417 IU/nM (each fusion protein contains 2 FVIIa, equivalent to 2208.7 IU/ nM FVIIa), comparable to the activity of the marketed recombinant FVIIa, Noci (2511 IU/nM FVIIa), indicating that the fusion protein provided by the present invention has a minimal effect on the activity of FVIIa by the C-terminally fused F. Its activity is also significantly higher than that of other reported recombinant FVIIa. For example, the recombinant hFVII biological activity produced by HEK cells reported by CSL Behring Company is 2874 IU/mg, which is equivalent to 144 IU/nM. The biological activity of the fusion protein hFVII-FP produced by HEK or CHO cells is 620-770 IU/mg, which is equivalent to 69-75 IU/nM (Weimer T et al., Thromb Haemost, 2008, 99: 659-667).
表3、各层析步骤分离组分体外激活24h的生物活性测定结果Table 3. Results of biological activity determination of isolated components in vitro for 24 h in each chromatographic step
Figure PCTCN2016106012-appb-000010
Figure PCTCN2016106012-appb-000010
实施例9.FP-A对血友病A小鼠急性出血的止血作用Example 9. Hemostatic effect of FP-A on acute hemorrhage in hemophilia A mice
我们以FVIII因子基因剔除纯合子甲型血友病小鼠(hemophilia A,HA,购自上海南方模式生物研究中心)断尾出血模型(tail vein transection(TVT)bleeding model)评估实施例5中所制备的活化的融合蛋白FP-A(单一组分P3-3)在HA小鼠体内的止血活性。选取10-12周龄雄性HA小鼠(购自上海南方模式生物研究中心),适应性饲养一周后将小鼠随机分为5组,每组8只。以0.8%戊巴比妥 钠(Sigma公司)按照0.1ml/10g剂量腹腔注射麻醉小鼠,然后分别尾静脉注射7,000(HA-F-0.7W组)、21,000(HA-F-2.1W组)、70,000(HA-F-7W组)和210,000IU/kg(HA-F-21W组)的FP-A和100,000IU/kg的诺其(
Figure PCTCN2016106012-appb-000011
Novo Nordisk公司)(HA-N组)。给药5分钟后,在小鼠尾末梢处剪断,切断处直径为3mm,迅速将尾端浸入装有14mL、37℃,含0.1%BSA(Amresco公司)生理盐水(0.9%NaCl,NS)的离心管中,并开始计时。如果出血在30分钟内停止,记录出血时间。如果出血时间超过30分钟,结扎鼠尾并用滚烫的金属棒灼烧止血,出血时间记作1800秒。同时,计算失血量。失血量(ml)=(采血后离心管重量(g)-采血前离心管重量(g))/1.05。另以10-12周龄,野生型雄性C57BL/6J(购自上海南方模式生物研究中心)小鼠(n=8)静注给予生理盐水(购自湖南科伦制药有限公司)作为正常对照组(C57-NS组),操作方法同给药组。所有数据以均数±标准误差
Figure PCTCN2016106012-appb-000012
表示,各实验组间比较采用t-test检验分析,分析软件采用Graphpad Prism 5.0,p<0.05认为有统计学意义。
We evaluated the FVIII factor gene knockout homozygous hemophilia A mouse (hemophilia A, HA, purchased from Shanghai Southern Model Biological Research Center) tail vein bleeding (TMT) bleeding model to evaluate the example 5 The hemostatic activity of the prepared activated fusion protein FP-A (single component P3-3) in HA mice. Male HA mice of 10-12 weeks old (purchased from Shanghai Southern Model Biological Research Center) were selected. After one week of adaptive feeding, the mice were randomly divided into 5 groups, 8 in each group. The mice were anesthetized with 0.8% pentobarbital sodium (Sigma) at a dose of 0.1 ml/10 g, and then injected intravenously with 7,000 (HA-F-0.7W group) and 21,000 (HA-F-2.1W group). , 70,000 (HA-F-7W group) and 210,000 IU/kg (HA-F-21W group) of FP-A and 100,000 IU/kg of Noki (
Figure PCTCN2016106012-appb-000011
Novo Nordisk) (HA-N group). After 5 minutes of administration, the mouse was cut at the distal end of the mouse. The cut-off area was 3 mm in diameter, and the tail end was quickly immersed in 14 mL, 37 ° C, containing 0.1% BSA (Amresco) physiological saline (0.9% NaCl, NS). Centrifuge the tube and start timing. If bleeding stops within 30 minutes, record the bleeding time. If the bleeding time exceeds 30 minutes, the rat tail is ligated and burned with a hot metal rod to stop bleeding. The bleeding time is recorded as 1800 seconds. At the same time, calculate the amount of blood loss. Blood loss (ml) = (centrifuge tube weight after blood collection (g) - centrifuge tube weight (g) before blood collection) / 1.05. Another 10-12 weeks old, wild-type male C57BL/6J (purchased from Shanghai Southern Model Biological Research Center) mice (n=8) were given intravenous saline (purchased from Hunan Kelun Pharmaceutical Co., Ltd.) as a normal control group. (C57-NS group), the operation method is the same as the administration group. All data in mean ± standard error
Figure PCTCN2016106012-appb-000012
T-test analysis was used for comparison between the experimental groups. The analysis software used Graphpad Prism 5.0, p<0.05 was considered statistically significant.
从图9和图10中各组动物的出血时间和出血量的统计结果分析,HA小鼠给予70,000IU/kg FP-A 5分钟后,其出血时间和出血量与C57-NS组接近,促凝效果明显,说明FP-A可以作为血友病等凝血因子缺乏症发生急性出血情况的有效凝血剂;小鼠给药FP-A 210,000IU/kg后,在出血时间和出血量上同样与C57-NS组接近。与FP-A 7,000IU/kg给药组比较,FP-A 70,000IU/kg组或FP-A 210,000IU/kg组的出血时间显著缩短(p<0.001;p<0.001),出血量也显著减少(p<0.001;p<0.01),具有一定的剂量-效应关系(详细结果见表4和表5)。From the statistical analysis of the bleeding time and the amount of bleeding in each group of animals in Fig. 9 and Fig. 10, after 5 minutes of administration of 70,000 IU/kg FP-A in HA mice, the bleeding time and the amount of bleeding were close to those of the C57-NS group. The coagulation effect is obvious, indicating that FP-A can be used as an effective clotting agent for acute bleeding in clotting factors such as hemophilia; after administration of FP-A 210,000 IU/kg, mice also have C57 in bleeding time and bleeding volume. - The NS group is close. Compared with the FP-A 7,000 IU/kg group, the bleeding time of the FP-A 70,000 IU/kg group or the FP-A 210,000 IU/kg group was significantly shortened (p<0.001; p<0.001), and the amount of bleeding was also significantly reduced. (p<0.001; p<0.01) with a dose-effect relationship (see Table 4 and Table 5 for detailed results).
表4、出血时间数据统计表(单位:秒)Table 4, bleeding time data statistics table (unit: second)
Figure PCTCN2016106012-appb-000013
Figure PCTCN2016106012-appb-000013
注:组1(C57-NS):C57BL/6J小鼠给予生理盐水组;组2(HA-N):HA小鼠给于10,000IU/kg诺其组;组3(HA-F-0.7W):HA小鼠给于FP-A 7,000IU/kg组;组4(HA-F-2.1W):HA小鼠给于FP-A 21,000IU/kg组;组5(HA-F-7W):HA小鼠给于FP-A 70,000IU/kg组;组6(HA-F-21W):HA小鼠给于FP-A 210,000IU/kg组。 Note: Group 1 (C57-NS): C57BL/6J mice were given saline group; Group 2 (HA-N): HA mice were given 10,000 IU/kg Novo group; Group 3 (HA-F-0.7W) ): HA mice were given to the FP-A 7,000 IU/kg group; Group 4 (HA-F-2.1W): HA mice were given to the FP-A 21,000 IU/kg group; Group 5 (HA-F-7W) : HA mice were given to the FP-A 70,000 IU/kg group; Group 6 (HA-F-21W): HA mice were given to the FP-A 210,000 IU/kg group.
表5、出血量数据统计表(单位:mL)Table 5, blood loss data statistics table (unit: mL)
Figure PCTCN2016106012-appb-000014
Figure PCTCN2016106012-appb-000014
注:分组同表4。Note: Grouped as in Table 4.
实施例10.FP-A在血友病A小鼠体内凝血作用的长效性研究Example 10. Long-term study of clotting of FP-A in hemophilia A mice
本实验是为了考察FP-A(单一组分P3-3激活后样品)对HA小鼠体内凝血作用的长效性。16-20周龄雄性HA小鼠(购自上海南方模式生物研究中心)适应性饲养一周后,随机分为3组,每组6只。其中两组给予300,000IU/kg的FP-A,另一组给予300,000IU/kg的诺其(
Figure PCTCN2016106012-appb-000015
Novo Nordisk公司)。同时以16-20周龄,野生型雄性C57BL/6J小鼠(购自上海南方模式生物研究中心)尾静脉注射给予生理盐水,作为正常对照组(n=6)。给予FP-A的两组HA小鼠分别于给药后1h和2h进行断尾试验;给予诺其的HA小鼠于给药后1h进行断尾试验;C57BL/6J正常对照组(C57-NS组)小鼠于注射后2h进行断尾试验。具体操作方法参照实施例9。所有数据以均数±标准误差
Figure PCTCN2016106012-appb-000016
表示,各实验组间比较采用t-test检验分析,分析软件采用Graphpad Prism 5.0,p<0.05认为有统计学意义。
This experiment was designed to investigate the long-term effects of FP-A (single-component P3-3 activated sample) on coagulation in HA mice. One week after 16-20 weeks old male HA mice (purchased from Shanghai Southern Model Biological Research Center), they were randomly divided into 3 groups, 6 in each group. Two groups were given 300,000 IU/kg of FP-A, and the other group was given 300,000 IU/kg of Noci (
Figure PCTCN2016106012-appb-000015
Novo Nordisk)). At the same time, wild-type male C57BL/6J mice (purchased from Shanghai Southern Model Biological Research Center) were given normal saline at 16-20 weeks old as a normal control group (n=6). The two groups of HA mice given FP-A were subjected to a tail-breaking test at 1 h and 2 h after administration; the HA mice given Novo were tail-tested 1 h after administration; C57BL/6J normal control group (C57-NS) Group) Mice were subjected to a tail-break test 2 h after injection. Refer to Example 9 for the specific operation method. All data in mean ± standard error
Figure PCTCN2016106012-appb-000016
T-test analysis was used for comparison between the experimental groups. The analysis software used Graphpad Prism 5.0, p<0.05 was considered statistically significant.
如图11所示,诺其组给药1h后,小鼠出血时间为30分钟,说明它已无促凝止血效果(HA-N-1h组),而给予FP-A 1h(HA-F-1h组)和2h后(HA-F-2h组)仍然能有效止血,出血时间均比诺其组显著缩短(P<0.01;P<0.05)。这说明FP-A相对于诺其具有显著延长的活性半衰期。具体数值参见表6。As shown in Figure 11, after 1 hour of administration, the bleeding time of the mice was 30 minutes, indicating that it had no procoagulant hemostasis effect (HA-N-1h group), and FP-A 1h (HA-F- After 1h group and 2h (HA-F-2h group), hemostasis was still effective, and the bleeding time was significantly shorter than that of Novo group (P<0.01; P<0.05). This indicates that FP-A has a significantly prolonged active half-life relative to Novo. See Table 6 for specific values.
表6、出血时间数据统计表(单位:s)Table 6. Statistics of bleeding time data (unit: s)
Figure PCTCN2016106012-appb-000017
Figure PCTCN2016106012-appb-000017
实施例11.FP-A的体内活性半衰期研究 Example 11. In vivo activity half-life of FP-A
本实验为考察FP-A(单一组分P3-3)在华法林致大鼠凝血障碍模型上的活性半衰期。依文献报道的方法(Joe Salas等,Thrombosis Research,2015,135(5):970-976或Gerhard Dickneite等,Thrombosis Research,2007,119:643-651)进行实验,选取8-12周龄200-220g SD大鼠16只(购于北京维通利华实验动物技术有限公司,许可证号:SCXK(京)2012-0001),将大鼠随机分为2组,每组8只。以2.5mg/kg华法林(Orion Corporation,Finland,批号:1569755)灌胃,24h后分别静脉给予10,000IU/kg的FP-A或诺其(
Figure PCTCN2016106012-appb-000018
Novo Nordisk公司)。FP-A组于给药后0.05、0.5、1、2、3、5、8、12h采血;诺其组于给药后0.05、0.5、1、2、3、5h采血。血样以终浓度0.013M的柠檬酸钠为抗凝剂,3000rpm离心10min取上清,按实施例8进行样品活性测定并计算活性半衰期。
This experiment was to investigate the active half-life of FP-A (single component P3-3) on warfarin-induced rat coagulopathy model. Experiments were performed according to methods reported in the literature (Joe Salas et al, Thrombosis Research, 2015, 135(5): 970-976 or Gerhard Dickneite et al, Thrombosis Research, 2007, 119: 643-651), selecting 8-12 weeks old 200- Sixteen 220 g SD rats (purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd., license number: SCXK (Beijing) 2012-0001) were randomly divided into two groups of 8 rats each. Oral administration of 2.5 mg/kg warfarin (Orion Corporation, Finland, batch number: 1569755), and intravenous administration of 10,000 IU/kg of FP-A or Noci after 24 hours.
Figure PCTCN2016106012-appb-000018
Novo Nordisk)). The FP-A group was collected at 0.05, 0.5, 1, 2, 3, 5, 8, and 12 h after administration; the Noqi group was collected at 0.05, 0.5, 1, 2, 3, and 5 h after administration. The blood sample was taken as an anticoagulant with a final concentration of 0.013 M sodium citrate, and the supernatant was taken by centrifugation at 3000 rpm for 10 min. The activity of the sample was measured according to Example 8 and the activity half-life was calculated.
如图12所示,测得FP-A的活性半衰期为3.03±0.35h;诺其的活性半衰期为1.01±0.16h。相比等活性的诺其,FP-A在大鼠体内活性半衰期延长了约3倍,单次注射FP-A 3h后血浆凝血活性约40%,而等活性给药的诺其组3h后活性已降至3%;FP-A给药12h后血浆凝血活性仍保持在7%以上。As shown in Figure 12, the active half-life of FP-A was measured to be 3.03 ± 0.35 h; the active half-life of Novo was 1.01 ± 0.16 h. Compared with the isoactive Noci, the activity half-life of FP-A in rats was extended by about 3 times, and the plasma coagulation activity was about 40% after a single injection of FP-A for 3 hours, and the activity of the same active Noki group was 3 hours later. It has been reduced to 3%; plasma clotting activity remains above 7% after 12 hours of FP-A administration.
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。 All documents mentioned in the present application are hereby incorporated by reference in their entirety in their entireties in the the the the the the the the In addition, it should be understood that various modifications and changes may be made by those skilled in the art in the form of the appended claims.

Claims (27)

  1. 一种活化的人凝血因子VII的融合蛋白,所述融合蛋白从N端至C端依次包含活化的人凝血因子VII、柔性肽接头、至少1个人绒毛膜促性腺激素β亚基羧基末端肽刚性单元和延长半衰期部分;其中,延长半衰期部分选自人免疫球蛋白Fc片段及其变体、白蛋白、转铁蛋白或PEG。An activated human factor VII fusion protein comprising, in order from the N-terminus to the C-terminus, an activated human coagulation factor VII, a flexible peptide linker, and at least one human chorionic gonadotropin beta subunit carboxy terminal peptide rigid The unit and the extended half-life portion; wherein the extended half-life portion is selected from the group consisting of a human immunoglobulin Fc fragment and variants thereof, albumin, transferrin or PEG.
  2. 如权利要求1所述的融合蛋白,其特征在于,所述融合蛋白是糖基化的。The fusion protein of claim 1 wherein the fusion protein is glycosylated.
  3. 如权利要求2所述的融合蛋白,其特征在于,所述融合蛋白是通过在哺乳动物细胞中表达而糖基化的。The fusion protein according to claim 2, wherein the fusion protein is glycosylated by expression in mammalian cells.
  4. 如权利要求3所述的融合蛋白,其特征在于,所述融合蛋白是通过在中国仓鼠卵巢细胞中表达而糖基化的。The fusion protein according to claim 3, wherein the fusion protein is glycosylated by expression in Chinese hamster ovary cells.
  5. 如权利要求1所述的融合蛋白,其特征在于,所述人凝血因子VII包含如SEQ ID NO:1所示的氨基酸序列,或者所述人凝血因子VII的氨基酸序列与如SEQ ID NO:1所示的氨基酸序列具有至少90%的同一性。The fusion protein according to claim 1, wherein said human coagulation factor VII comprises the amino acid sequence set forth in SEQ ID NO: 1, or the amino acid sequence of said human coagulation factor VII and SEQ ID NO: 1 The amino acid sequences shown have at least 90% identity.
  6. 如权利要求1所述的融合蛋白,其特征在于,所述柔性肽接头含有2个或多个选自G、S、A和T残基的氨基酸。The fusion protein according to claim 1, wherein the flexible peptide linker comprises two or more amino acids selected from the group consisting of G, S, A and T residues.
  7. 如权利要求6所述的融合蛋白,其特征在于所述柔性肽接头具有以(GS)a(GGS)b(GGGS)c(GGGGS)d循环单元组合形成的氨基酸序列通式,其中a,b,c和d是大于或等于0的整数,且a+b+c+d≥1。The fusion protein according to claim 6, wherein said flexible peptide linker has an amino acid sequence formula formed by combining (GS) a (GGS) b (GGGS) c (GGGGS) d cyclic units, wherein a, b , c and d are integers greater than or equal to 0, and a+b+c+d≥1.
  8. 如权利要求7所述的融合蛋白,所述柔性肽接头优选自下组:The fusion protein of claim 7 wherein said flexible peptide linker is preferably from the group consisting of:
    (i)GSGGGSGGGGSGGGGS;(i) GSGGGSGGGGSGGGGS;
    (ii)GSGGGGSGGGGSGGGGSGGGGSGGGGS;(ii) GSGGGGSGGGGSGGGGSGGGGSGGGGS;
    (iii)GGGGSGGGGSGGGGSGGGGS;(iii) GGGGSGGGGSGGGGSGGGGS;
    (iv)GSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGS;(iv) GSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGS;
    (v)GGGSGGGSGGGSGGGSGGGS。(v) GGGSGGGSGGGSGGGSGGGS.
  9. 如权利要求1所述的融合蛋白,其特征在于,所述人绒毛膜促性腺激素β亚基的羧基末端肽刚性单元包含如SEQ ID NO:7所示氨基酸序列或其截短的序列,其中所述截短的序列包含至少2个糖基化位点。The fusion protein according to claim 1, wherein the carboxy terminal peptide rigid unit of the human chorionic gonadotropin beta subunit comprises the amino acid sequence set forth in SEQ ID NO: 7 or a truncated sequence thereof, wherein The truncated sequence comprises at least 2 glycosylation sites.
  10. 如权利要求9所述的融合蛋白,其特征在于所述人绒毛膜促性腺激素β亚基的羧基末端肽刚性单元包含以下氨基酸序列:The fusion protein according to claim 9, wherein the carboxy terminal peptide rigid unit of the human chorionic gonadotropin beta subunit comprises the following amino acid sequence:
    (i)SSSSKAPPPSLPSPSRLPGPSDTPILPQ; (i) SSSSKAPPPSLPSPSRLPGPSDTPILPQ;
    (ii)PRFQDSSSSKAPPPSLPSPSRLPGPSDTPILPQ;(ii) PRFQDSSSSKAPPPSLPSPSRLPGPSDTPILPQ;
    (iii)SSSSKAPPPS;(iii) SSSSKAPPPS;
    (iv)SRLPGPSDTPILPQ。(iv) SRLPGPSDTPILPQ.
  11. 如权利要求1所述的融合蛋白,其特征在于,所述人绒毛膜促性腺激素β亚基的羧基末端肽刚性单元与权利要求9或10所述融合蛋白中的刚性单元至少具有70%,80%,90%或95%的同一性。The fusion protein according to claim 1, wherein the carboxy terminal peptide rigid unit of the human chorionic gonadotropin beta subunit has at least 70% of the rigid unit in the fusion protein of claim 9 or 10. 80%, 90% or 95% identity.
  12. 如权利要求1所述的融合蛋白,其特征在于,所述融合蛋白包含1、2、3、4或5个人绒毛膜促性腺激素β亚基的羧基末端肽刚性单元。The fusion protein according to claim 1, wherein the fusion protein comprises a carboxy terminal peptide rigid unit of 1, 2, 3, 4 or 5 human chorionic gonadotropin beta subunits.
  13. 如权利要求1所述的融合蛋白,其特征在于,所述人免疫球蛋白Fc变体具有降低的ADCC效应和/或CDC效应,和/或与FcRn受体的结合亲和力增强。The fusion protein of claim 1, wherein the human immunoglobulin Fc variant has a reduced ADCC effect and/or a CDC effect, and/or an enhanced binding affinity to an FcRn receptor.
  14. 如权利要求13所述的融合蛋白,其特征在于,所述Fc变体选自:The fusion protein of claim 13 wherein said Fc variant is selected from the group consisting of:
    (i)含有Leu234Val、Leu235Ala和Pro331Ser突变的人IgG1绞链区、CH2和CH3区域;(i) human IgG1 hinge region, CH2 and CH3 regions containing the Leu234Val, Leu235Ala and Pro331Ser mutations;
    (ii)含有Pro331Ser突变的人IgG2绞链区、CH2和CH3区域;(ii) human IgG2 hinge region, CH2 and CH3 regions containing the Pro331Ser mutation;
    (iii)含有Thr250Gln和Met428Leu突变的人IgG2绞链区、CH2和CH3区域;(iii) human IgG2 hinge region, CH2 and CH3 regions containing the Thr250Gln and Met428Leu mutations;
    (iv)含有Pro331Ser、Thr250Gln和Met428Leu突变的人IgG2绞链区、CH2和CH3区域;(iv) human IgG2 hinge region, CH2 and CH3 regions containing the Pro331Ser, Thr250Gln and Met428Leu mutations;
    (v)含有Ser228Pro和Leu235Ala突变的人IgG4绞链区、CH2和CH3区域。(v) Human IgG4 hinge region, CH2 and CH3 regions containing the Ser228Pro and Leu235Ala mutations.
  15. 如权利要求1所述的融合蛋白,其特征在于,所述融合蛋白的氨基酸序列如SEQ ID NO:16所示。The fusion protein according to claim 1, wherein the amino acid sequence of the fusion protein is shown in SEQ ID NO: 16.
  16. 如权利要求1所述的融合蛋白,其特征在于,所述融合蛋白的活性>15000IU/mg,和/或,所述融合蛋白在动物体内循环半衰期至少延长1倍。The fusion protein according to claim 1, wherein the fusion protein has an activity of > 15000 IU/mg, and/or the fusion protein has at least a 1-fold increase in circulating half-life in the animal.
  17. 编码如权利要求1-16中任一项所述的融合蛋白的DNA分子。A DNA molecule encoding the fusion protein of any of claims 1-16.
  18. 如权利要求17所述DNA分子,所述DNA分子的序列如SEQ ID NO:17所示。The DNA molecule according to claim 17, wherein the sequence of the DNA molecule is as shown in SEQ ID NO: 17.
  19. 一种载体,其特征在于,包含如权利要求17或18所述的DNA分子。A vector comprising the DNA molecule of claim 17 or 18.
  20. 一种宿主细胞,其特征在于,包含如权利要求19所述的载体,或者转染了权利要求19所述的载体。 A host cell comprising the vector of claim 19 or transfected with the vector of claim 19.
  21. 一种药物组合物,其特征在于,包含药学上可接受的载体、赋形剂或稀释剂,以及有效剂量的如权利要求1-16中任一项所述的融合蛋白。A pharmaceutical composition comprising a pharmaceutically acceptable carrier, excipient or diluent, and an effective amount of the fusion protein of any of claims 1-16.
  22. 一种制备如权利要求1-16中任一项所述的融合蛋白的方法,所述方法包括:A method of preparing a fusion protein according to any one of claims 1 to 16, the method comprising:
    (a)将根据权利要求17或18所述编码融合蛋白的DNA引入CHO细胞,生成CHO衍生的细胞系;(a) introducing a DNA encoding a fusion protein according to claim 17 or 18 into a CHO cell to generate a CHO-derived cell line;
    (b)筛选步骤(a)中在其生长培养基中每24小时期间内,表达超过3μg/106(百万)个细胞的高产细胞株;(b) a high-yielding cell line expressing more than 3 μg/10 6 (million) cells per 24 hours in its growth medium in the screening step (a);
    (c)培养步骤(b)筛选到的细胞株,表达融合蛋白;(c) culturing step (b) the selected cell strain to express the fusion protein;
    (d)收获步骤(c)中得到的发酵液,纯化融合蛋白;(d) harvesting the fermentation broth obtained in step (c), purifying the fusion protein;
    (e)激活步骤(d)中纯化的融合蛋白。(e) activating the purified fusion protein in step (d).
  23. 如权利要求22所述的方法,其特征在于,所述步骤(d)中融合蛋白纯化过程包含ProteinA亲和层析、多维模式层析、阴离子交换层析和分子筛层析。The method according to claim 22, wherein the fusion protein purification process in the step (d) comprises Protein A affinity chromatography, multidimensional mode chromatography, anion exchange chromatography and molecular sieve chromatography.
  24. 如权利要求23所述的方法,其特征在于,所述步骤(d)中Protein A亲和层析使用Mabselect层析介质;和所述多维模式层析使用CHT陶瓷羟磷灰石II型层析介质;和所述阴离子交换层析使用Q Sepharose High Performance层析介质;和所述分子筛层析使用Superdex 200层析介质。The method according to claim 23, wherein said Protein A affinity chromatography in said step (d) uses a Mabselect chromatography medium; and said multidimensional mode chromatography uses CHT ceramic hydroxyapatite type II chromatography a medium; and the anion exchange chromatography uses a Q Sepharose High Performance chromatography medium; and the molecular sieve chromatography uses a Superdex 200 chromatography medium.
  25. 如权利要求22所述的方法,其特征在于,所述方法步骤(e)中激活步骤采用凝血因子XII激活、柱上自激活或溶液自激活。The method according to claim 22, wherein the activating step in step (e) of the method employs factor XII activation, on-column self-activation or solution self-activation.
  26. 一种如权利要求1-16中任一项所述融合蛋白在制备预防或治疗出血性疾病的药物中应用。A fusion protein according to any one of claims 1 to 16 for use in the manufacture of a medicament for preventing or treating a bleeding disorder.
  27. 如权利要求26所述的应用,包括用于制备产生FVIII抗体的A型血友病患者或产生FIX抗体的B型血友病患者的自发性出血、手术中或手术后出血的治疗或预防、先天性或后天获得性FVII缺乏症患者的出血性疾病的预防或治疗、和/或血小板机能不全患者的治疗的药物中应用。 The use according to claim 26, comprising the treatment or prevention of spontaneous bleeding, intraoperative or postoperative bleeding of a patient with hemophilia A who produces a FVIII antibody or a hemophilia A patient who produces a FIX antibody, The use of a drug for the prevention or treatment of a bleeding disorder in a patient with congenital or acquired FVII deficiency, and/or treatment of a patient with platelet insufficiency.
PCT/CN2016/106012 2016-08-19 2016-11-16 Activated human blood-clotting factor vii fusion protein, and manufacturing method and application of same WO2018032639A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610692686.4A CN106279436B (en) 2016-08-19 2016-08-19 Human blood coagulation factor VII fusion protein of activation and preparation method thereof and purposes
CN201610692686.4 2016-08-19

Publications (1)

Publication Number Publication Date
WO2018032639A1 true WO2018032639A1 (en) 2018-02-22

Family

ID=57660598

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/106012 WO2018032639A1 (en) 2016-08-19 2016-11-16 Activated human blood-clotting factor vii fusion protein, and manufacturing method and application of same

Country Status (2)

Country Link
CN (1) CN106279436B (en)
WO (1) WO2018032639A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111303274A (en) * 2020-03-21 2020-06-19 上海浦东明炎生物技术有限公司 Purification method of human chorionic gonadotropin

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11123438B2 (en) 2016-08-19 2021-09-21 Ampsource Biopharma Shanghai Inc. Linker peptide for constructing fusion protein
CN106317226B (en) * 2016-08-19 2017-09-05 安源医药科技(上海)有限公司 Connection peptide for construction of fusion protein
CN106256835A (en) * 2016-08-19 2016-12-28 安源医药科技(上海)有限公司 High-glycosylation human growth hormone's fusion protein and preparation method thereof and purposes
CN106279437B (en) 2016-08-19 2017-10-31 安源医药科技(上海)有限公司 Hyperglycosylated human coagulation factor VIII fusion proteins and preparation method thereof and purposes
CN113105562B (en) * 2018-09-26 2023-12-01 安源医药科技(上海)有限公司 Application of mutant single-chain human coagulation factor VIII in preparation of fusion protein
US11981718B2 (en) 2020-05-27 2024-05-14 Ampsource Biopharma Shanghai Inc. Dual-function protein for lipid and blood glucose regulation

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100317585A1 (en) * 2006-02-03 2010-12-16 Udi Eyal Fima Long-acting coagulation factors and methods of producing same
WO2011020866A2 (en) * 2009-08-20 2011-02-24 Csl Behring Gmbh Albumin fused coagulation factors for non-intravenous administration in the therapy and prophylactic treatment of bleeding disorders
US20130243747A1 (en) * 2006-02-03 2013-09-19 Prolor Biotech Inc. Long-acting coagulation factors and methods of producing same
CN103397009A (en) * 2013-08-16 2013-11-20 安源生物科技(上海)有限公司 Improved-type human coagulation factor FVII-Fc fusion protein as well as preparation method and application thereof
WO2014060401A1 (en) * 2012-10-15 2014-04-24 Novo Nordisk Health Care Ag Coagulation factor vii polypeptides

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6103501A (en) * 1997-11-17 2000-08-15 Washington University Single chain glycoprotein hormones comprising two β and one α subunits and recombinant production thereof
EP1816201A1 (en) * 2006-02-06 2007-08-08 CSL Behring GmbH Modified coagulation factor VIIa with extended half-life
BR112014015156A2 (en) * 2011-12-20 2020-10-27 Indiana University Research And Technology Corporation ctp-based insulin analogues, their methods of production and use in the treatment of hyperglycemia, as well as nucleic acid and host cell sequences
EP2938637A2 (en) * 2012-12-28 2015-11-04 AbbVie Inc. Multivalent binding protein compositions
CN104693270B (en) * 2013-12-10 2018-10-16 清华大学 A kind of connection peptide for fusion protein

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100317585A1 (en) * 2006-02-03 2010-12-16 Udi Eyal Fima Long-acting coagulation factors and methods of producing same
US20130243747A1 (en) * 2006-02-03 2013-09-19 Prolor Biotech Inc. Long-acting coagulation factors and methods of producing same
WO2011020866A2 (en) * 2009-08-20 2011-02-24 Csl Behring Gmbh Albumin fused coagulation factors for non-intravenous administration in the therapy and prophylactic treatment of bleeding disorders
WO2014060401A1 (en) * 2012-10-15 2014-04-24 Novo Nordisk Health Care Ag Coagulation factor vii polypeptides
CN103397009A (en) * 2013-08-16 2013-11-20 安源生物科技(上海)有限公司 Improved-type human coagulation factor FVII-Fc fusion protein as well as preparation method and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CARR, M.E. ET AL.: "Emerging and Future Therapies for Hemophilia", JOURNAL OF BLOOD MEDICINE, vol. 6, 3 September 2015 (2015-09-03), XP055465842 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111303274A (en) * 2020-03-21 2020-06-19 上海浦东明炎生物技术有限公司 Purification method of human chorionic gonadotropin
CN111303274B (en) * 2020-03-21 2024-01-30 上海浦东明炎生物技术有限公司 Human chorionic gonadotrophin purifying process

Also Published As

Publication number Publication date
CN106279436A (en) 2017-01-04
CN106279436B (en) 2017-10-31

Similar Documents

Publication Publication Date Title
CA3059994C (en) Human coagulation factor ix (fix) fusion protein, preparation method therefor, and use thereof
WO2018032639A1 (en) Activated human blood-clotting factor vii fusion protein, and manufacturing method and application of same
RU2722374C1 (en) High glycated fused protein based on human blood coagulation factor viii, method for production thereof and use thereof
US9458223B2 (en) Von willebrand factor variants having improved factor VIII binding affinity
JP2018104459A (en) Compounds suitable for treatment of hemophilia
JP2016522219A (en) Compounds suitable for the treatment of hemophilia
JP2015519313A (en) Pharmaceutical composition suitable for the treatment of hemophilia
WO2020063562A1 (en) Fusion protein of mutated single-chain human coagulation factor viii, preparation method therefor, and use thereof
WO2015021711A1 (en) Improved fusion protein of human blood coagulation factor fvii-fc, preparation method therefor and use thereof
CN102690354B (en) Recombined dimerization antithrombin III-Fc fusion protein and mammalian cell efficient expression system thereof
JP2022118184A (en) Conjugated c1 esterase inhibitor and uses thereof
KR20220062084A (en) Recombinant IgG Fc Multimers for the Treatment of Immune Complex-Mediated Renal Disorders
WO2023214387A1 (en) Novel cd200 fusion proteins
CN103599527A (en) Pharmaceutical composition containing modified type human coagulation factor FVII-Fc fusion protein

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: 16913394

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: 16913394

Country of ref document: EP

Kind code of ref document: A1