WO2007046631A1 - Method for manufacturing high purified factor ix - Google Patents

Method for manufacturing high purified factor ix Download PDF

Info

Publication number
WO2007046631A1
WO2007046631A1 PCT/KR2006/004237 KR2006004237W WO2007046631A1 WO 2007046631 A1 WO2007046631 A1 WO 2007046631A1 KR 2006004237 W KR2006004237 W KR 2006004237W WO 2007046631 A1 WO2007046631 A1 WO 2007046631A1
Authority
WO
WIPO (PCT)
Prior art keywords
factor
exchange chromatography
solution
highly purified
cation exchange
Prior art date
Application number
PCT/KR2006/004237
Other languages
French (fr)
Inventor
Yong Kang
Yong-Woon Choi
Ki-Whan Son
Sung-Rae Lee
Hark-Mo Sung
Ki-Yong Kim
Jae-Wook Huh
Original Assignee
Green Cross Corporation
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 Green Cross Corporation filed Critical Green Cross Corporation
Priority to CN2006800390241A priority Critical patent/CN101291951B/en
Publication of WO2007046631A1 publication Critical patent/WO2007046631A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • 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)
    • C12N9/644Coagulation factor IXa (3.4.21.22)
    • 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)
    • C12Y304/21022Coagulation factor IXa (3.4.21.22)

Definitions

  • the present invention relates to a method for manufacturing a highly purified human blood coagulation factor IX, and more preferably, to a method for manufacturing a highly purified human blood coagulation factor IX by subjecting a plasma- derived or recombinant material containing a human blood coagulation factor IX, and by using of ion chromatography and affinity chromatography and inactivating or removing viruses, thereby obtaining a highly purified, safe coagulation factor IX having a specific activity of above 150 IU/mg, with substantially inactivation or removal of all impure proteins and viruses.
  • a human blood coagulation factor IX (abbreviated to Factor IX) is a glycoprotein that is indispensable for a coagulation cascade. Hemophilia B is caused by the absence or deficiency of Factor IX due to a hereditary cause or a pathological cause.
  • the hemophilia B is a rare hereditary disease that primarily affects newborn male babies having prevalence of one of 30,000 ⁇ 50,000. Cure of the hemophilia B is performed by administering Factor IX concentrates.
  • Factor IX is synthesized in the liver and has similar features like as other vitamin Independent glycoproteins.
  • Vitamin K-dependent glycoproteins include Factor II, Factor VII, Factor X, and so on.
  • the Factor IX a kind of serine protease, is a monomeric glycoprotein containing about 17.5% glucose comprising 4.7% hexose, 6.8% N- acetylhexsamine, and 6% sialic acid.
  • Factor IX has a molecular weight of 55 ⁇ 75 kDa (Daltons) according to the amount of carbohydrate contents. The exact molecular weight of only proteins, which is calculated based on amino acid sequences, is 47,054 Da.
  • Factor IX is activated by the release of activation peptide fragments consisting of 35 amino acids due to cleavage of an arginine(Arg)-alanine(Ala) bond by Factor XIa at sites between Arg and Ala and between Arg and VaI .
  • Factor IX a phospholipid complex comprising Factor Vila, Factor III, and calcium ion (Ca + ) (Fig. 1).
  • Factor IX is present in the human plasma in a very low concentration of about 0.1 ⁇ M (5 D/D).
  • high doses of whole blood or plasma should be required. Such high dose blood administration may, however, involve severe side effects, which results from excessive administration of other proteins such as fibrinogen in the plasma.
  • Factor IX concentrates were developed for the first time in the late 1950s. To isolate Factor IX concentrates, Factor IX is first collected from the plasma and then adsorbed into barium sulfate, followed by precipitation and washing to remove other protein components from Factor IX adsorbed into barium sulfate. Later, barium sulfates were replaced with tricalcium phosphates which are nontoxic salts. The process of isolating Factor IX concentrates by adsorption and precipitation may also cause concentration of other vitamin K-dependent glycoproteins such as Factor II, Factor VII, Factor X, and so on. These concentrates are termed a prothrombin complex concentrates (PCC). Factor IX complex obtained above has been widely used as the therapeutic agent of the hemophilia B since the 1960s.
  • PCC prothrombin complex concentrates
  • Factor II, Factor VII, Factor X, and other proteins from the Factor IX complex was attempted, and a highly purified Factor IX had become commercially available from the 1990s.
  • the highly purified Factor IX was obtained by subjecting concentrated Factor IX complex to anion exchange chromatography followed by heparin gel or monoclonal antibody chromatography, thereby removing Factor II, Factor VII, Factor X, and other proteins from the Factor IX complex.
  • Factor IX complex is currently being replaced by the highly purified Factor IX.
  • a recombinant coagulation factor IX using the Chinese hamster ovary (CHO) cell line was developed and manufactured by Genetic Institute in the trade name of Benefix TM in 1997.
  • Virus inactivation is generally performed by solvent/detergent (SfD) treatment.
  • SfD solvent/detergent
  • affinity chromatography can be added in the process of virus inactivation.
  • the most effective method of virus removal is a nanofiltration technique using a 20 nm grade nanofilter. However, this technique requires a process of preliminarily purifying a coagulation factor to obtain a highly purified coagulation factor, otherwise aggregation or clogging of proteins make the nanofilter useless.
  • Fig. 2 is a process flow diagram illustrating a method for manufacturing a highly purified Factor IX according to the present invention.
  • a solution containing Factor IX used in the present invention is not particularly restricted in their forms.
  • cryo- precipitate-free human plasma is preferably used as a starting material.
  • the starting substance include plasma fractions, cell culture broth of recombinant Factor IX, and solution containing the Factor IX, and so on.
  • a method for manufacturing a highly purified Factor IX from a material containing the Factor IX as a starting material provided aprimarily eluting a solution containing the Factor IX by anion exchange chromatography, followed inactivating viruses by Solvent/Detergent (SfD) treatment, secondarily eluting the resultant by anion exchange chromatography, concentrating and filtrating the secondarily eluted product, obtaining eluate by heparin affinity chromatography, collecting an unbound solution by cation exchange chromatography for further purification, and removing viruses by subjecting the collected unbound solution to nanofiltration.
  • Solvent/Detergent SfD
  • the eluate is purified by heparin affinity chromatography using heparin as a ligand for a blood coagulation factor. Then, the eluate is subjected to cation exchange chromatography to collect an unbound solution that is not adsorbed into a cation exchanger, thereby removing other impurities remaining in the eluate.
  • heparin is used as a ligand in the heparin affinity chromatography
  • resin materials including Sepharose and agarose and polyacrylamide can be used as a support, but not limited to herein.
  • a heparin Sepharose 6FF column is currently preferred.
  • an equilibration buffer used prior to application of the solution containing Factor IX and a solution containing Factor IX preferably have ion strengths of not greater than 20 mS/cm and pH range of between 5.0 and 9.0.
  • a washing buffer used for washing other proteins adsorbed into a resin preferably has ion strengths of 10 ⁇ 20 mS/cm.
  • an elution buffer used for eluting the solution containing Factor IX preferably has ion strengths of 20 ⁇ 50 mS/ cm.
  • a particularly preferred embodiment relates to the isolation of Factor IX by cation exchange chromatography, for example by using the following materials: weak cations such as carboxymethyl- (CM-), or carboxy- (C-); and strong cations such as sulfo- (S-), sulfomethyl- (SM-), sulfoethyl- (SE-), sulfopropyl- (SP-), or phospho- (P-).
  • weak cations such as carboxymethyl- (CM-), or carboxy- (C-)
  • strong cations such as sulfo- (S-), sulfomethyl- (SM-), sulfoethyl- (SE-), sulfopropyl- (SP-), or phospho- (P-).
  • a variety of column resins can be used, including Sepharose, Sephadex, agarose, Sephacel, Polystyrene, Polyacrylate, Cellulose, and Toyope
  • the cation exchange chromatography is preferably performed at a pH level in a range of between 3.0 and 6.0 such that an unbound solution that is not adsorbed into a column is collected for purification.
  • an equilibration buffer used in the cation exchange chromatography can be used in various manners, the solution containing the equilibration buffer used prior to application of the solution containing Factor IX and the solution containing Factor IX have ion strength in a range of between about 10 and about 50 mS/cm.
  • the present invention provides a method for manufacturing a highly purified Factor
  • the method for manufacturing a highly purified Factor IX comprise of subjecting a plasma-derived or recombinant material containing a human blood coagulation factor IX to ion chromatography and affinity chromatography and inactivating or removing viruses, thereby obtaining a highly purified, safe coagulation factor IX having a specific activity of above 150 IU/mg, with substantially inactivation or removal of all impure proteins and viruses.
  • FIG. 1 shows intrinsic and extrinsic coagulation pathways
  • FIG. 2 is a process flow diagram illustrating a method for manufacturing a highly purified Factor IX according to the present invention
  • Fig. 3 shows results of SDS-polyacrylamide gel electrophoresis (SDS-PAGE) analysis identifying purity of Factor IX manufactured by the present invention and other commercially available Factor IX products in non-reducing (A) and reducing (B) condition.
  • SDS-PAGE SDS-polyacrylamide gel electrophoresis
  • FIG. 4 is a graph showing the results of tests for evaluating stability of activity expressed as the t
  • a cryoprecipitate-free plasma or cell culture medium of recombinant Factor IX as a starting material was mixed with DEAE-Sephadex A-50, prewashed with buffer, and stirred for 2 hours at 4 0 C. Subsequently, the gel to which Factor IX was now bound was separated by filtration or centrifugation. After washing, Factor IX was eluted by a buffer with high salt concentration. A pH of the eluate was appropriately adjusted to 7.5 and then the eluate was subjected to dialysis and concentration for storage at -7O 0 C. The concentrate was aliquoted and measured the activity under the standards and test methods of blood formulations and recombinant formulations as stipulated by the KFDA (Korea Food and Drug Administration).
  • FIG. 3 shows results of SDS-PAGE analysis carried out to determine purity of Factor IX manufactured by the present invention and other commercially available Factor IX products. As shown in Fig. 3, Factor IX manufactured by the present invention had little impure proteins, suggesting that they had higher purity levels than other Factor IX products.
  • EXAMPLE 7 Purification of Factor IX from cell culture medium
  • CHO cells cloned with the Factor IX were cultured in serum free media, and a 5L culture media removed the CHO cells was treated as the same methods as mentioned in Examples 1 through 3, except of S/D treatment, and thereby Factor IX was purified.
  • Table 7 shows various data items, including specific activity, purification fold, yield, and so on.
  • EXAMPLE 8 Virus inactivation and removal process
  • clearance tests were conducted on various viruses, including HIV (Human Immunodeficiency Virus), BHV (Bovine Herpes Virus), BVDV (Bovine Viral Diarrhea Virus), HAV (Hepatitis A Virus), EMCV (Encephalo Mycarditis Virus), PPV (Porcine Parvovirus), and so on.
  • HIV Human Immunodeficiency Virus
  • BHV Bovine Herpes Virus
  • BVDV Bovine Viral Diarrhea Virus
  • HAV Hepatitis A Virus
  • EMCV Electrodencephalo Mycarditis Virus
  • PPV Porcine Parvovirus
  • Virus clearance values shown in Table 8 are log factors obtained by subtracting titers of processing from titers of stock viruses used, while virus reduction values shown in Table 9 are log factors of values obtained by subtracting titers of processing from spiked titers of stock viruses after spiking the stock viruses into target viruses.
  • the present invention is industrially applicable by providing a method for manufacturing a highly purified Factor IX comprise of subjecting a plasma-derived or recombinant material containing a human blood coagulation factor IX to ion chromatography and affinity chromatography and inactivating or removing viruses, thereby obtaining a highly purified, safe coagulation factor IX having a specific activity of above 150 IU/mg, with substantially inactivation or removal of all impure proteins and viruses.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Analytical Chemistry (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to a method for manufacturing a highly purified human blood coagulation factor IX, and more preferably, to a method for manufacturing a highly purified human blood coagulation factor IX by subjecting a plasma-derived or recombinant material containing a human blood coagulation factor IX, and by using of ion chromatography and affinity chromatography and inactivating or removing viruses, thereby obtaining a highly purified, safe coagulation factor IX having a specific activity of above 150 IU/mg, with substantially inactivation or removal of all impure proteins and viruses.

Description

Description
METHOD FOR MANUFACTURING HIGH PURIFIED FACTOR
IX
Technical Field
[1] The present invention relates to a method for manufacturing a highly purified human blood coagulation factor IX, and more preferably, to a method for manufacturing a highly purified human blood coagulation factor IX by subjecting a plasma- derived or recombinant material containing a human blood coagulation factor IX, and by using of ion chromatography and affinity chromatography and inactivating or removing viruses, thereby obtaining a highly purified, safe coagulation factor IX having a specific activity of above 150 IU/mg, with substantially inactivation or removal of all impure proteins and viruses.
[2]
Background Art
[3] A human blood coagulation factor IX (abbreviated to Factor IX) is a glycoprotein that is indispensable for a coagulation cascade. Hemophilia B is caused by the absence or deficiency of Factor IX due to a hereditary cause or a pathological cause.
[4] The hemophilia B is a rare hereditary disease that primarily affects newborn male babies having prevalence of one of 30,000 ~ 50,000. Cure of the hemophilia B is performed by administering Factor IX concentrates.
[5] Factor IX is synthesized in the liver and has similar features like as other vitamin Independent glycoproteins. Vitamin K-dependent glycoproteins include Factor II, Factor VII, Factor X, and so on. The Factor IX, a kind of serine protease, is a monomeric glycoprotein containing about 17.5% glucose comprising 4.7% hexose, 6.8% N- acetylhexsamine, and 6% sialic acid. Factor IX has a molecular weight of 55 ~ 75 kDa (Daltons) according to the amount of carbohydrate contents. The exact molecular weight of only proteins, which is calculated based on amino acid sequences, is 47,054 Da. 12 gamma(γ) - carboxy glutamic acid domains play important roles in the activation of Factor IX and thus Factor IX is activated in the presence of phospholipid and calcium ion (Ca +). The diversity of isoelectric point (pi) of Factor IX depends on the kind and amount of carbohydrates present therein (Journal of Chromatography (1999) 844, 119).
[6] Factor IX is activated by the release of activation peptide fragments consisting of 35 amino acids due to cleavage of an arginine(Arg)-alanine(Ala) bond by Factor XIa at sites between Arg and Ala and between Arg and VaI . Factor IX without the
145 146 180 181 activated peptides is referred to an active form of Factor IX to distinguish the natural form of Factor IX. The activation of Factor IX is also stimulated by a phospholipid complex comprising Factor Vila, Factor III, and calcium ion (Ca +) (Fig. 1). Factor IX is present in the human plasma in a very low concentration of about 0.1 μM (5 D/D). To treat a patient of hemophilia B, high doses of whole blood or plasma should be required. Such high dose blood administration may, however, involve severe side effects, which results from excessive administration of other proteins such as fibrinogen in the plasma.
[7] To overcome the side effects, Factor IX concentrates were developed for the first time in the late 1950s. To isolate Factor IX concentrates, Factor IX is first collected from the plasma and then adsorbed into barium sulfate, followed by precipitation and washing to remove other protein components from Factor IX adsorbed into barium sulfate. Later, barium sulfates were replaced with tricalcium phosphates which are nontoxic salts. The process of isolating Factor IX concentrates by adsorption and precipitation may also cause concentration of other vitamin K-dependent glycoproteins such as Factor II, Factor VII, Factor X, and so on. These concentrates are termed a prothrombin complex concentrates (PCC). Factor IX complex obtained above has been widely used as the therapeutic agent of the hemophilia B since the 1960s.
[8] A cold ethanol fraction process, which has been used to obtain albumin or globulin from plasma, was not suited to the separation of Factor IX complex. Thus, such conventional methods cannot be advantageously employed to purification of Factor IX. To solve the disadvantage, a column process using anion exchange chromatography has been developed. In the developed process, Factor Vlll-rich fractions are removed by cryoprecipitation, and Factor IX present in the cryoprecipitate-free plasma is adsorbed into an anion exchange resin and then is purified. However, this process may lead to adsorption of other proteins such as Factor II, Factor VII, or Factor X. In spite of such low purified Factor IX, the Factor IX complex is currently in use for the treatment of patients with the hemophilia B.
[9] Unfortunately, there are several findings reporting that administration of Factor IX complex cause incidence of symptoms such as venous thrombosis or disseminated intravascular coagulation (Thrombosis and Haemostasis (1995) 73, 584. Thrombosis and Haemostasis (1998) 79, 778). Such side effects presumably result from hyperco- agulation states caused by excessive amounts of coagulant proteins or unnecessary thrombogenic components of the concentrates when administered along with Factor IX.
[10] In order to alleviate side effects of Factor IX complex, an approach for removing
Factor II, Factor VII, Factor X, and other proteins from the Factor IX complex was attempted, and a highly purified Factor IX had become commercially available from the 1990s. The highly purified Factor IX was obtained by subjecting concentrated Factor IX complex to anion exchange chromatography followed by heparin gel or monoclonal antibody chromatography, thereby removing Factor II, Factor VII, Factor X, and other proteins from the Factor IX complex. Thus Factor IX complex is currently being replaced by the highly purified Factor IX. A recombinant coagulation factor IX using the Chinese hamster ovary (CHO) cell line was developed and manufactured by Genetic Institute in the trade name of Benefix in 1997.
[11] The most serious problem in the treatment of blood-derived coagulation factors is the risk of viral infection due to plasma-derived viruses such as HIV, HBV, HCV, and the like. To ensure virus safety of the blood-derived coagulation factors, a virus removal is an indispensable process for ensuring the safety of blood-derived coagulation factors. Virus inactivation is generally performed by solvent/detergent (SfD) treatment. To achieve a more effective method of virus removal, a process of affinity chromatography can be added in the process of virus inactivation. The most effective method of virus removal is a nanofiltration technique using a 20 nm grade nanofilter. However, this technique requires a process of preliminarily purifying a coagulation factor to obtain a highly purified coagulation factor, otherwise aggregation or clogging of proteins make the nanofilter useless.
[12] Use of monoclonal antibodies is effective in selectively separating Factor IX from other coagulation factors. However, since animal cells has been used for antibody production, it is impossible to avoid the risks of viral transmission due to viruses derived from the animal cells. Furthermore, side effects can be appeared according to eluting antibodies during chromatography. It is also impossible to overcome the problem associated with viral infection when Factor IX is isolated from a recombinant cell culture media. Accordingly, there is a need a method for manufacturing a highly purified Factor IX, which has a purity of greater than 99% and a safety inactivated and removed of viruses.
[13]
Disclosure of Invention Technical Problem
[14] To solve the above problems, it is an object of the present invention to provide a method for manufacturing a highly purified Factor IX for eliminating side effects such as venous thrombosis or disseminated intravascular coagulation (DIC), which resulting from hypercoagulable states caused by excessive amounts of coagulant proteins or unnecessary thrombogenic components of the concentrates when administered along with Factor IX.
[15]
Technical Solution [16] Hereinafter, a method for manufacturing a highly purified Factor IX according to the present invention will be described in detail.
[17] Fig. 2 is a process flow diagram illustrating a method for manufacturing a highly purified Factor IX according to the present invention. A solution containing Factor IX used in the present invention is not particularly restricted in their forms. In the method for manufacturing a highly purified Factor IX according to the present invention, cryo- precipitate-free human plasma is preferably used as a starting material. In addition, examples of the starting substance include plasma fractions, cell culture broth of recombinant Factor IX, and solution containing the Factor IX, and so on.
[18] In an embodiment of the present invention, a method for manufacturing a highly purified Factor IX from a material containing the Factor IX as a starting material provided aprimarily eluting a solution containing the Factor IX by anion exchange chromatography, followed inactivating viruses by Solvent/Detergent (SfD) treatment, secondarily eluting the resultant by anion exchange chromatography, concentrating and filtrating the secondarily eluted product, obtaining eluate by heparin affinity chromatography, collecting an unbound solution by cation exchange chromatography for further purification, and removing viruses by subjecting the collected unbound solution to nanofiltration.
[19] In detail, in order to remove impure proteins remaining in the plasma protein solution eluted by the anion exchange chromatography, the eluate is purified by heparin affinity chromatography using heparin as a ligand for a blood coagulation factor. Then, the eluate is subjected to cation exchange chromatography to collect an unbound solution that is not adsorbed into a cation exchanger, thereby removing other impurities remaining in the eluate.
[20] Since heparin is used as a ligand in the heparin affinity chromatography, a variety of resin materials including Sepharose and agarose and polyacrylamide can be used as a support, but not limited to herein. Particularly, a heparin Sepharose 6FF column is currently preferred. According to the present invention, when eluate is purified by heparin affinity chromatography, an equilibration buffer used prior to application of the solution containing Factor IX and a solution containing Factor IX preferably have ion strengths of not greater than 20 mS/cm and pH range of between 5.0 and 9.0. In addition, a washing buffer used for washing other proteins adsorbed into a resin preferably has ion strengths of 10 ~ 20 mS/cm. Furthermore, an elution buffer used for eluting the solution containing Factor IX preferably has ion strengths of 20 ~ 50 mS/ cm.
[21] A particularly preferred embodiment relates to the isolation of Factor IX by cation exchange chromatography, for example by using the following materials: weak cations such as carboxymethyl- (CM-), or carboxy- (C-); and strong cations such as sulfo- (S-), sulfomethyl- (SM-), sulfoethyl- (SE-), sulfopropyl- (SP-), or phospho- (P-). In addition, a variety of column resins can be used, including Sepharose, Sephadex, agarose, Sephacel, Polystyrene, Polyacrylate, Cellulose, and Toyopearl. In the present invention, the cation exchange chromatography is preferably performed at a pH level in a range of between 3.0 and 6.0 such that an unbound solution that is not adsorbed into a column is collected for purification. Although an equilibration buffer used in the cation exchange chromatography can be used in various manners, the solution containing the equilibration buffer used prior to application of the solution containing Factor IX and the solution containing Factor IX have ion strength in a range of between about 10 and about 50 mS/cm. [22]
Advantageous Effects
[23] The present invention provides a method for manufacturing a highly purified Factor
IX. More particularly, the method for manufacturing a highly purified Factor IX according to the present invention comprise of subjecting a plasma-derived or recombinant material containing a human blood coagulation factor IX to ion chromatography and affinity chromatography and inactivating or removing viruses, thereby obtaining a highly purified, safe coagulation factor IX having a specific activity of above 150 IU/mg, with substantially inactivation or removal of all impure proteins and viruses.
[24]
Brief Description of the Drawings
[25] Fig. 1 shows intrinsic and extrinsic coagulation pathways;
[26] Fig. 2 is a process flow diagram illustrating a method for manufacturing a highly purified Factor IX according to the present invention;
[27] Fig. 3 shows results of SDS-polyacrylamide gel electrophoresis (SDS-PAGE) analysis identifying purity of Factor IX manufactured by the present invention and other commercially available Factor IX products in non-reducing (A) and reducing (B) condition.
[28] M: Standard marker (myosin; 198kDa, galactosidase; 115kDa, BSA; 93kDa, ovalbumin; 49.8kDa, carbonic anhydrase; 35.8kDa, soybean trypsin inhibitor; 29.2kDa, lysozyme; 21.3kDa, aprotinin; 6.4kDa); Lane 1: highly purified Factor IX according to method mentioned in table 2; Lane 2: highly purified Factor IX according to method mentioned in table 3; Lane 3: highly purified Factor IX according to method mentioned in table 4; Lane 4: Facnyne (Greencross); Lane 5: Mononine (ZLB Behring); Lane 6: Octanyne (Octapharma); Lane 7: Berinin HS (ZLB Behring); and Lane 8: Immunine (Baxter). [29] FIG. 4 is a graph showing the results of tests for evaluating stability of activity expressed as the titer carried out under CM-Sepharose fast flow (FF) purification conditions.
[30]
Best Mode for Carrying Out the Invention
[31] EXAMPLE 1: Purification of Factor IX by anion exchange chromatography
(conventional Process)
[32] A cryoprecipitate-free plasma or cell culture medium of recombinant Factor IX as a starting material was mixed with DEAE-Sephadex A-50, prewashed with buffer, and stirred for 2 hours at 40C. Subsequently, the gel to which Factor IX was now bound was separated by filtration or centrifugation. After washing, Factor IX was eluted by a buffer with high salt concentration. A pH of the eluate was appropriately adjusted to 7.5 and then the eluate was subjected to dialysis and concentration for storage at -7O0C. The concentrate was aliquoted and measured the activity under the standards and test methods of blood formulations and recombinant formulations as stipulated by the KFDA (Korea Food and Drug Administration).
[33] The above fraction containing Factor IX was subjected to perform a virus in- activation process by S/D treatment. A post-inactivation solution was adsorbed into a DEAE-toyopearl 650M column for second purification, followed by collecting an eluate.
[34]
[35] EXAMPLE 2: Purification of Factor IX by Heparin affinity chromatography
[36] 10 ml of the Factor IX complex solution prepared by the purification in Example 1 was conditioned to have an ion strength of not greater than 20 mS/cm, and pH 7.0, and an equilibration buffer (20 mM sodium citrate, pH 7.0) was induced to a Heparin- Sepharose 6FF column (column conditions: 1 cm in diameter, 18 cm in height, 1.0 ml/ min in flow rate, and room temperature) for equilibration. Meanwhile, a sample was loaded for adsorption, washed with the equilibration buffer plus 0.1 M NaCl, and then eluted using 0.25 M NaCl. A protein concentration of the eluate was determined and SDS-PAGE was carried out. The activity was measured using a coagulation timer KClO manufactured by Amelung, Germany.
[37] A specific activity of the eluate preparation from Example 1 was compared with that of the eluate preparation from Example 2. As the result, the specific activity of the Heparin-Sepharose 6FF column eluate of Example 2 was about 60-90 IU/mg, which showed an increase of at least 10 times that of the anion exchange column eluate of Example 1.
[38] [39] EXAMPLE 3: Purification of Factor IX by cation exchange chromatography
[40] 10 ml of the Factor IX complex solution prepared by the purification in Example 2 was conditioned to have an ion strength of not greater than 20 mS/cm, and pH 4.0, and an equilibration buffer (20 mM citrate in 0.30 ~ 0.4 M NaCl, pH 4.0) was induced to an equilibrated CM-Sepharose 6FF column (column conditions: 1 cm in diameter, 5 cm in height, and 1.0 ml/min in flow rate) for collecting an unbound solution. A protein concentration of the unbound solution was determined and SDS-PAGE analysis was carried out. A specific activity of the unbound solution was at least 150 IU/mg. Fig. 3 shows results of SDS-PAGE analysis carried out to determine purity of Factor IX manufactured by the present invention and other commercially available Factor IX products. As shown in Fig. 3, Factor IX manufactured by the present invention had little impure proteins, suggesting that they had higher purity levels than other Factor IX products.
[41] [42] EXAMPLES 4 - 6: Large-scale Purification of Factor IX [43] To confirm large-scale feasibility of the highly purified Factor IX according to the present invention, scale-up experiments were carried out several times. In each of the scale-up experiments, various data items, including specific activity, purification fold, yield, and so on, are shown in Tables 1 through 6 below. [44] Table 1
Figure imgf000009_0001
Figure imgf000010_0001
[45] [46] Table 2
Figure imgf000010_0002
[47] [48] Table 3
Figure imgf000010_0003
Figure imgf000011_0001
[49] [50] Table 4
Figure imgf000011_0002
Figure imgf000012_0001
[51] [52] Table 5
Figure imgf000012_0002
[53] [54] Table 6
Figure imgf000013_0001
[55] As shown in Tables 1 through 6, repeated practices of the present invention confirmed that highly purified Factors IX have the specific activity of at least 150 IU/ mg in final concentrates. The final concentrates were placed into vials each by an appropriate amount and lyophilized to be used as test samples of prolonged stability and preclinical samples.
[56] [57] EXAMPLE 7: Purification of Factor IX from cell culture medium [58] CHO cells cloned with the Factor IX were cultured in serum free media, and a 5L culture media removed the CHO cells was treated as the same methods as mentioned in Examples 1 through 3, except of S/D treatment, and thereby Factor IX was purified. Table 7 shows various data items, including specific activity, purification fold, yield, and so on.
[59] Table 7
Figure imgf000014_0001
[60] [61] EXAMPLE 8: Virus inactivation and removal process [62] In virus inactivation and nanofiltration steps, clearance tests were conducted on various viruses, including HIV (Human Immunodeficiency Virus), BHV (Bovine Herpes Virus), BVDV (Bovine Viral Diarrhea Virus), HAV (Hepatitis A Virus), EMCV (Encephalo Mycarditis Virus), PPV (Porcine Parvovirus), and so on. The results are shown in Tables 8 and 9 below. Virus clearance values shown in Table 8 are log factors obtained by subtracting titers of processing from titers of stock viruses used, while virus reduction values shown in Table 9 are log factors of values obtained by subtracting titers of processing from spiked titers of stock viruses after spiking the stock viruses into target viruses.
[63] Table 8 Virus clearance on the processing of plasma-derived Factor IX
Figure imgf000015_0001
[64] [65] Table 9 Virus reduction on the processing of plasma-derived Factor IX
Figure imgf000015_0002
[66]
Industrial Applicability [67] The present invention is industrially applicable by providing a method for manufacturing a highly purified Factor IX comprise of subjecting a plasma-derived or recombinant material containing a human blood coagulation factor IX to ion chromatography and affinity chromatography and inactivating or removing viruses, thereby obtaining a highly purified, safe coagulation factor IX having a specific activity of above 150 IU/mg, with substantially inactivation or removal of all impure proteins and viruses.

Claims

Claims
[1] A method for manufacturing a highly purified human blood coagulation factor
IX (Factor IX) using a material containing Factor IX as a starting material, the method comprising: collecting a solution containing Factor IX by performing anion exchange chromatography one or more times; eluting the solution containing Factor IX by performing heparin affinity chromatography; collecting an unbound solution by performing cation exchange chromatography and removing viruses by subjecting the collected unbound solution to nanofiltration.
[2] The method of claim 1, in the step on heparin affinity chromatography, wherein equilibration buffer and the solution containing the Factor IX have ion strength of not greater than 20 mS/cm at a pH range of 5.0 ~ 9.0, a washing buffer has ion strength of 10 ~ 20 mS/cm, and an elution buffer has ion strength of 20 ~ 50 mS/ cm.
[3] The method of claim 1, wherein a column functional group for the cation exchange chromatography is selected from the group consisting of car- boxymethyl- (CM-), carboxy- (C-), sulfo- (S-), sulfomethyl- (SM-), sulfoethyl- (SE-), sulfopropyl- (SP-), and phospho- (P-), and a column resin is selected from the group consisting of Sepharose, Sephadex, agarose, Sephacel, Polystyrene, Polyacrylate, Cellulose, and Toyopearl.
[4] The method of claim 1, wherein the collecting of the unbound solution by the cation exchange chromatography is performed at a pH in a range of between 3.0 and 6.0.
[5] The method of claim 1, wherein the collecting of the unbound solution by the cation exchange chromatography is performed using a solution containing an equilibration buffer and Factor IX having ion strength in a range of 10 ~ 50 mS/ cm.
[6] The method of claim 1, wherein the material containing Factor IX is a human plasma-derived or cell culture medium of recombinant Factor IX.
PCT/KR2006/004237 2005-10-18 2006-10-18 Method for manufacturing high purified factor ix WO2007046631A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN2006800390241A CN101291951B (en) 2005-10-18 2006-10-18 Method for manufacturing high purified factor IX

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2005-0098235 2005-10-18
KR1020050098235A KR100667860B1 (en) 2005-10-18 2005-10-18 Method for manufacturing high purified factor ix product

Publications (1)

Publication Number Publication Date
WO2007046631A1 true WO2007046631A1 (en) 2007-04-26

Family

ID=37867823

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2006/004237 WO2007046631A1 (en) 2005-10-18 2006-10-18 Method for manufacturing high purified factor ix

Country Status (3)

Country Link
KR (1) KR100667860B1 (en)
CN (1) CN101291951B (en)
WO (1) WO2007046631A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012037530A1 (en) 2010-09-17 2012-03-22 Baxter International Inc. Stabilization of immunoglobulins and other proteins through aqueous formulations with sodium chloride at weak acidic to neutral ph
US20150210754A1 (en) * 2011-05-16 2015-07-30 Omrix Biopharmaceuticals Ltd. Immunoglobulin reduced in thrombogenic agents and preparation thereof
JP2017505780A (en) * 2014-02-04 2017-02-23 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. Use of cation exchange chromatography in flow-through mode to enrich post-translational modifications
CN115975997A (en) * 2022-12-19 2023-04-18 成都蓉生药业有限责任公司 Secondary ultrafiltration dialysate for purifying human blood coagulation factor IX and purification method

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104560925B (en) * 2014-12-30 2017-11-03 山东泰邦生物制品有限公司 A kind of method that activation prepares human thrombin in chromatography waste liquid from humanclottingfactorⅨ
CN111378029B (en) * 2018-12-29 2023-05-05 四川远大蜀阳药业有限责任公司 Preparation method of human coagulation factor IX

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5457181A (en) * 1987-10-23 1995-10-10 Centre Regional De Transfusion Sanguine De Lille Preparation of a high-purity human factor IX concentrate and other plasmatic proteins and their therapeutic use
WO1996025490A1 (en) * 1995-02-15 1996-08-22 Octapharma Ag A process for the purification of vitamin k dependent coagulation factors by chromatography
JPH09286797A (en) * 1996-04-18 1997-11-04 Green Cross Corp:The Heparin cofactor ii and its purification
US6893856B2 (en) * 1993-12-10 2005-05-17 Octapharma Ag Process for preparing virus-inactivated factors 1X and X by membrane chromatography

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US545718A (en) * 1895-09-03 Blast-furnace
CN1297896A (en) * 1999-11-29 2001-06-06 中国科学技术大学 Method of extracting activated prothrombin X(FXa) effectively

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5457181A (en) * 1987-10-23 1995-10-10 Centre Regional De Transfusion Sanguine De Lille Preparation of a high-purity human factor IX concentrate and other plasmatic proteins and their therapeutic use
US6893856B2 (en) * 1993-12-10 2005-05-17 Octapharma Ag Process for preparing virus-inactivated factors 1X and X by membrane chromatography
WO1996025490A1 (en) * 1995-02-15 1996-08-22 Octapharma Ag A process for the purification of vitamin k dependent coagulation factors by chromatography
JPH09286797A (en) * 1996-04-18 1997-11-04 Green Cross Corp:The Heparin cofactor ii and its purification

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012037530A1 (en) 2010-09-17 2012-03-22 Baxter International Inc. Stabilization of immunoglobulins and other proteins through aqueous formulations with sodium chloride at weak acidic to neutral ph
US20150210754A1 (en) * 2011-05-16 2015-07-30 Omrix Biopharmaceuticals Ltd. Immunoglobulin reduced in thrombogenic agents and preparation thereof
US9796770B2 (en) * 2011-05-16 2017-10-24 Omrix Biopharmaceuticals Ltd. Immunoglobulin reduced in thrombogenic agents and preparation thereof
JP2017505780A (en) * 2014-02-04 2017-02-23 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. Use of cation exchange chromatography in flow-through mode to enrich post-translational modifications
US12018046B2 (en) 2014-02-04 2024-06-25 Biogen Ma Inc. Use of cation-exchange chromatography in the flow-through mode to enrich post-translational modifications
CN115975997A (en) * 2022-12-19 2023-04-18 成都蓉生药业有限责任公司 Secondary ultrafiltration dialysate for purifying human blood coagulation factor IX and purification method
CN115975997B (en) * 2022-12-19 2023-11-17 成都蓉生药业有限责任公司 Secondary ultrafiltration dialysate for purifying human coagulation factor IX and purification method

Also Published As

Publication number Publication date
KR100667860B1 (en) 2007-01-11
CN101291951A (en) 2008-10-22
CN101291951B (en) 2012-07-04

Similar Documents

Publication Publication Date Title
US5457181A (en) Preparation of a high-purity human factor IX concentrate and other plasmatic proteins and their therapeutic use
US4495175A (en) Preparation of highly purified human antihemophilic factor
EP0378208B1 (en) Production method for protein-containing composition
JP3435668B2 (en) A method for the industrial-scale production of high purity, therapeutically suitable standardized human von Willebrand factor concentrates.
US4656254A (en) Method of preparing alpha-1-proteinase inhibitor and antithrombin III
US4470969A (en) Process for producing a concentrate of coagulation factors VII and VIIa
Wickerhauser et al. Development of Large Scale Fractionation Methods: VII. Preparation of Antithrombin III Concentrate 1
US7816495B2 (en) Processes for the preparation of fibrinogen
EP0617049B1 (en) Process for the isolation of highly purified factors IX, X and II from prothrombin complex or human plasma
JPH0764750B2 (en) Process for producing alpha-1-proteinase inhibitor
EP0176926A2 (en) Process for producing a high purity antihemophilic factor concentrate
WO2007046631A1 (en) Method for manufacturing high purified factor ix
EP0252392A2 (en) Viral inactivation and purification of active proteins
US6063909A (en) Preparation of factor IX
ES2224109T3 (en) PROCEDURE FOR PREPARATION OF HUMAN PROTEIN C ACTIVATED.
Wickerhauser et al. A Single‐Step Method for the Isolation of Antithrombin III 1, 2
EP0650364B1 (en) Purification of factor ix
AU665452C (en) Purification of factor IX
CN114395032A (en) Method for extracting human antithrombin III from blood plasma
Wickerhauser et al. Development of large scale fractionation methods

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200680039024.1

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 3915/DELNP/2008

Country of ref document: IN

122 Ep: pct application non-entry in european phase

Ref document number: 06799311

Country of ref document: EP

Kind code of ref document: A1