WO2018032639A1 - Protéine de fusion du facteur vii de coagulation sanguine humaine activée, son procédé de fabrication et son application - Google Patents

Protéine de fusion du facteur vii de coagulation sanguine humaine activée, son procédé de fabrication et son application 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
English (en)
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/fr

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

Abstract

La présente invention concerne une protéine de fusion du facteur VII (FVIIa) de coagulation sanguine humaine hautement glycosylée et activée, et un procédé de fabrication et une application de celle-ci. La protéine de fusion comprend, de l'extrémité N-terminale à l'extrémité C-terminale, un FVIIa humain, un connecteur peptidique flexible, au moins une unité rigide d'un peptide terminal carboxyle de sous-unité beta de gonadotrophine chorionique humaine, et une portion d'extension de demi-vie (de préférence sélectionnée à partir d'une variante Fc d'IgG humaine). La protéine de fusion présente un niveau similaire d'activité biologique en tant que FVIIa humain naturel et prolonge la demi vie de in vivo ce qui permet d'améliorer la pharmacocinétique et l'efficacité médicamenteuse.
PCT/CN2016/106012 2016-08-19 2016-11-16 Protéine de fusion du facteur vii de coagulation sanguine humaine activée, son procédé de fabrication et son application WO2018032639A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610692686.4A CN106279436B (zh) 2016-08-19 2016-08-19 活化的人凝血因子vii融合蛋白及其制备方法与用途
CN201610692686.4 2016-08-19

Publications (1)

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

Family

ID=57660598

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/106012 WO2018032639A1 (fr) 2016-08-19 2016-11-16 Protéine de fusion du facteur vii de coagulation sanguine humaine activée, son procédé de fabrication et son application

Country Status (2)

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

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111303274A (zh) * 2020-03-21 2020-06-19 上海浦东明炎生物技术有限公司 一种人绒毛膜促性腺激素纯化方法

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107759696A (zh) * 2016-08-19 2018-03-06 安源医药科技(上海)有限公司 人白介素7融合蛋白及其制备方法
CN106256835A (zh) * 2016-08-19 2016-12-28 安源医药科技(上海)有限公司 高糖基化人生长激素融合蛋白及其制备方法与用途
CN106279437B (zh) 2016-08-19 2017-10-31 安源医药科技(上海)有限公司 高糖基化人凝血因子viii融合蛋白及其制备方法与用途
EP3502143A4 (fr) 2016-08-19 2020-07-15 Ampsource Biopharma Shanghai Inc. Peptide de liaison pour la construction d'une protéine de fusion
CN113105562B (zh) * 2018-09-26 2023-12-01 安源医药科技(上海)有限公司 突变型单链人凝血因子viii在制备融合蛋白中的应用
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 (fr) * 2009-08-20 2011-02-24 Csl Behring Gmbh Facteurs de coagulation fusionnés à de l'albumine pour l'administration non intraveineuse dans le cadre de la thérapie et du traitement prophylactique des troubles hémostatiques
US20130243747A1 (en) * 2006-02-03 2013-09-19 Prolor Biotech Inc. Long-acting coagulation factors and methods of producing same
CN103397009A (zh) * 2013-08-16 2013-11-20 安源生物科技(上海)有限公司 改良型人凝血因子FVII-Fc融合蛋白及其制备方法与用途
WO2014060401A1 (fr) * 2012-10-15 2014-04-24 Novo Nordisk Health Care Ag Polypeptides de facteur vii de coagulation

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 (fr) * 2006-02-06 2007-08-08 CSL Behring GmbH Facteur de coagulation VIIa modifié ayant une stabilité 'half-life' améliorée
US9573987B2 (en) * 2011-12-20 2017-02-21 Indiana University Research And Technology Corporation CTP-based insulin analogs for treatment of diabetes
US9915665B2 (en) * 2012-12-28 2018-03-13 Abbvie Inc. High-throughput system and method for identifying antibodies having specific antigen binding activities
CN104693270B (zh) * 2013-12-10 2018-10-16 清华大学 一种用于融合蛋白的连接肽

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 (fr) * 2009-08-20 2011-02-24 Csl Behring Gmbh Facteurs de coagulation fusionnés à de l'albumine pour l'administration non intraveineuse dans le cadre de la thérapie et du traitement prophylactique des troubles hémostatiques
WO2014060401A1 (fr) * 2012-10-15 2014-04-24 Novo Nordisk Health Care Ag Polypeptides de facteur vii de coagulation
CN103397009A (zh) * 2013-08-16 2013-11-20 安源生物科技(上海)有限公司 改良型人凝血因子FVII-Fc融合蛋白及其制备方法与用途

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 (zh) * 2020-03-21 2020-06-19 上海浦东明炎生物技术有限公司 一种人绒毛膜促性腺激素纯化方法
CN111303274B (zh) * 2020-03-21 2024-01-30 上海浦东明炎生物技术有限公司 一种人绒毛膜促性腺激素纯化方法

Also Published As

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

Similar Documents

Publication Publication Date Title
CA3059994C (fr) Proteine de fusion du facteur ix de coagulation humain (fix), son procede de preparation et son utilisation
WO2018032639A1 (fr) Protéine de fusion du facteur vii de coagulation sanguine humaine activée, son procédé de fabrication et son application
RU2722374C1 (ru) Высокогликозилированный слитый белок на основе фактора свертывания крови человека viii, способ его получения и его применение
US9458223B2 (en) Von willebrand factor variants having improved factor VIII binding affinity
JP2018104459A (ja) 血友病の治療に適する化合物
JP2016522219A (ja) 血友病の治療に適する化合物
JP2015519313A (ja) 血友病の治療に適する医薬組成物
WO2020063562A1 (fr) Protéine de fusion de facteur de coagulation viii humain monocaténaire muté, procédé de préparation associé et utilisation associée
WO2015021711A1 (fr) Protéine de fusion améliorée de facteur de coagulation sanguine humain fvii-fc, procédé de préparation s'y rapportant et son utilisation
CN102690354B (zh) 重组二聚化抗凝血酶III-Fc融合蛋白及其哺乳动物细胞高效表达系统
JP2022118184A (ja) コンジュゲートされたc1エステラーゼインヒビター及びその使用
KR20220062084A (ko) 면역 복합체 매개된 콩팥 장애의 치료를 위한 재조합 IgG Fc 다량체
WO2023214387A1 (fr) Nouvelles protéines de fusion cd200
CN103599527A (zh) 包含改良型人凝血因子FVII-Fc融合蛋白的药物组合物

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