WO2014185716A1 - Method for removing impurities by using calcium phosphate precipitation - Google Patents
Method for removing impurities by using calcium phosphate precipitation Download PDFInfo
- Publication number
- WO2014185716A1 WO2014185716A1 PCT/KR2014/004341 KR2014004341W WO2014185716A1 WO 2014185716 A1 WO2014185716 A1 WO 2014185716A1 KR 2014004341 W KR2014004341 W KR 2014004341W WO 2014185716 A1 WO2014185716 A1 WO 2014185716A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- protein
- concentration
- antibody
- calcium phosphate
- phosphate precipitation
- Prior art date
- Legal status (The legal status 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 status listed.)
- Ceased
Links
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K1/00—General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
- C07K1/14—Extraction; Separation; Purification
- C07K1/16—Extraction; Separation; Purification by chromatography
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K1/00—General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
- C07K1/14—Extraction; Separation; Purification
- C07K1/30—Extraction; Separation; Purification by precipitation
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K1/00—General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
- C07K1/14—Extraction; Separation; Purification
- C07K1/34—Extraction; Separation; Purification by filtration, ultrafiltration or reverse osmosis
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K19/00—Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/10—Immunoglobulins specific features characterized by their source of isolation or production
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/30—Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
Definitions
- the present invention relates to a method for removing impurities using calcium phosphate precipitation, and more particularly, to a method for removing impurities by adding a calcium ion solution and a phosphate ion solution to a protein-containing sample to cause a calcium phosphate precipitation reaction.
- Resin using calcium phosphate precipitation e.g., Ceramic Hydroxyapatite resin, Hydroxyapatite resin: manufactured by BioRad, USA
- chromatography binds or drops proteins to the resin in a specific buffered environment. How to separate.
- the present inventors added calcium phosphate precipitates (Calcium phosphate precipitate, Ca 5 ) by adding calcium ion (Ca 2+ ) solution and phosphate ion (PO 4 3- ) solution to protein-containing samples without using column chromatography using resin.
- (PO 4 ) 3 (OH), Hydroxyapatite) was induced to devise a method for removing impurities.
- calcium phosphate precipitate is formed, the protein is adsorbed with the calcium phosphate precipitate to precipitate together.
- the type of protein that binds to calcium phosphate precipitates depends on the concentration of calcium phosphate when added. Thus, the desired protein can be separated according to the concentration of calcium and phosphate. The method does not require as much equipment, buffers and long time as the protein purification using the chromatography column mentioned above.
- an object of the present invention is to provide a method for removing impurities in a protein-containing sample, comprising the step of forming a calcium phosphate precipitation reaction by adding a calcium ion solution and a phosphate ion solution to a protein-containing sample.
- Another problem to be solved by the present invention is to provide a method for producing a protein formulation comprising a purification step using the above method.
- one embodiment of the present invention provides a method for removing impurities in a protein-containing sample comprising the following steps:
- another embodiment of the present invention provides a method for producing a protein formulation comprising a purification step using the above method.
- the purification process of the antibody is based on Protein A chromatography, which can obtain high purity and yield with only one step purification.
- Protein A chromatography Protein A chromatography
- Industrial purification of pharmaceutical antibodies development, operation and validation of chromatography processes. Biotechnol Genet Eng Rev 2001; 18: 301-27 .
- ion-exchange creams are used to remove high molecular weight protein multimers, DNA, viruses, host cell-derived proteins, leached Protein A and impurities.
- calcium phosphate precipitation may be used in place of equipment and time-consuming additional chromatography.
- the calcium phosphate precipitation method instead of the above additional chromatography, large molecular weight protein multimers, DNA, viruses, host cell-derived protein, Protein A, etc. can be effectively removed in a short time with little labor.
- the cells and cell residues are removed by centrifugation and filtration, and the filtered cell culture medium is removed.
- the solution is again washed with the equilibrium solution, and the antibody is eluted from the Protein A column with a low pH elution solution.
- the eluted antibody solution is titrated to pH 3.6 and stored at room temperature for 1 hour for low pH virus inactivation. Thereafter, the antibody solution is neutralized to pH 6.0.
- the neutralized antibody solution was further purified by anion exchange (Q sepharose Fast Flow; manufactured by GE Healthcare), and the intermediate antibody solution obtained from anion exchange chromatography was adjusted to pH 7.0, followed by 100 mM CaCl 2 and 100 mM.
- Sodium phosphate pH 7.0 is titrated in the intermediate purified solution to a final concentration of 10 mM Ca 2+ and 10 mM phosphate to cause precipitation. After titration, it is maintained at room temperature for 3 hours to sufficiently combine calcium phosphate precipitate (Hydroxyapatite, Ca 5 (PO 4 ) 3 (OH)) with antibody multimers and other impurities (DNA, host cell-derived protein, Protein A, etc.). Let's do it. Thereafter, centrifugation or filtration is performed to remove the precipitates from the antibody solution.
- calcium phosphate precipitation may be applied to the intermediate purified antibody solution.
- the filtered antibody solution can then be subjected to anion exchange chromatography.
- the impurity removal method using calcium phosphate precipitation is a step after the protein A chromatography, the sequence and chromatography techniques (eg, ion exchange chromatography, hydrophobic interaction chromatography, mixed mode chromatography) It is possible to apply instead of intermediate chromatography purification of proteins (including antibodies).
- sequence and chromatography techniques eg, ion exchange chromatography, hydrophobic interaction chromatography, mixed mode chromatography
- the present invention provides a method for removing impurities in a protein-containing sample, comprising adding a calcium ion solution and a phosphate ion solution to a protein-containing sample to form a calcium phosphate precipitation reaction. do. And, after the calcium phosphate precipitation reaction step may further comprise the step of removing the precipitate by centrifugation or filtration.
- prior to the calcium phosphate precipitation reaction step may further comprise the step of applying a protein containing sample to protein A adsorption chromatography to purify.
- further purification of the protein-containing sample purified in the previous step after the calcium phosphate precipitation reaction step, or after the Protein A adsorption chromatography purification step and before the calcium phosphate precipitation reaction step may include a step.
- purification may be applied to any one of chromatography selected from the group consisting of ion exchange chromatography, hydrophobic interaction chromatography, and mixed mode chromatography, but is not limited thereto.
- the ion exchange chromatography may be cation ion exchange chromatography or anion ion exchange chromatography.
- the hydrophobic interaction chromatography may be Ether (eg, Toyopearl Ether-650M, Toyopearl Ether-650S, etc.), Butyl (eg, Butyl sepharose, Butyl S sepharose, Catpo Butyl, etc.), Hexyl (eg, Toyopearl Hexyl-650C). ), Octyl (e.g. Octyl sepharose, etc.), Pheyl (e.g.
- the mixed mode chromatography may be a mixed mode chromatography (eg, Capto adhere, Capto MMC, MEP HyperCell, Eshmuno HCX, etc.), an ion exchange function, and a cation exchange function.
- Glyphography eg, hydroxyapatite, Ceramic hydroxyapate, etc. may be included, but is not limited thereto.
- one embodiment of the present invention provides a method for removing impurities in a protein containing sample comprising the following steps:
- the protein-containing sample is a sample containing phosphate ions
- the protein-containing sample is a sample containing phosphate ions
- it can be added only a phosphate solution.
- the calcium ion solution may be CaCl 2 , or CaCO 3 , but is not limited thereto.
- the phosphate ion solution may be Na 3 PO 4 , NaH 2 PO 4 , Na 2 HPO 4 or KH 2 PO 4, K 2 HPO 4, K 3 PO 4 , but is not limited thereto. It is not.
- the concentration of calcium ions added to the calcium phosphate precipitation reaction is 1 to 100 mM, 1 to 90 mM, 1 to 80 mM, 1 to 70 mM, 1 to 60 mM, 1 to 50 mM , 1 to 40 mM, 1 to 30 mM, 1 to 20 mM, 1 to 10 mM, or 1 to 5 mM, but is not limited thereto.
- the concentration of calcium ions added to the calcium phosphate precipitation reaction may be 1 to 20 mM.
- the concentration of phosphate ions added to the calcium phosphate precipitation reaction is 1 to 100 mM, 1 to 90 mM, 1 to 80 mM, 1 to 70 mM, 1 to 60 mM, 1 to 50 mM , 1 to 40 mM, 1 to 30 mM, 1 to 20 mM, 1 to 10 mM, or 1 to 5 mM, but is not limited thereto.
- the concentration of phosphate ions added to the calcium phosphate precipitation reaction may be 1 to 20 mM.
- the concentration of calcium ions added to the calcium phosphate precipitation reaction may be 1 to 100 mM, and the concentration of phosphate ions may be 1 to 100 mM.
- the concentration of calcium ions added to the calcium phosphate precipitation reaction may be 1 to 20 mM, and the concentration of phosphate ions may be 1 to 20 mM.
- the calcium phosphate precipitation reaction is pH 5.0 to 10.0, pH 5.0 to 9.5, pH 5.0 to 9.0, pH 5.0 to 8.5, pH 5.0 to 8.0, pH 5.0 to 7.5, pH 5.0 to 7.0, pH It may be performed at 5.0 to 6.5, pH 5.0 to 6.0, or pH 5.0 to 5.5, but is not limited thereto.
- the calcium phosphate precipitation reaction may be carried out at pH 5.5 to 7.5.
- the concentration of calcium ions added to the calcium phosphate precipitation reaction is 1 to 20 mM
- the concentration of phosphate ions is 1 to 20 mM
- the calcium phosphate precipitation reaction is pH 5.5 to 7.5. This can be done at
- the calcium phosphate precipitation reaction time is 0 to 10 hours, 0 to 9 hours, 0 to 8 hours, 0 to 7 hours, 0 to 6 hours, 0 to 5 hours, 0 to 4 hours, 0 to 3 hours, 0 to 2 hours, 0 to 1 hour, or 0 to 0.5 hours, but is not limited thereto.
- the calcium phosphate precipitation reaction time may be 3 to 5 hours.
- the concentration of calcium ions added to the calcium phosphate precipitation reaction is 1 to 20 mM
- the concentration of phosphate ions is 1 to 20 mM
- the calcium phosphate precipitation reaction time is 3 to 5 It can be time.
- the concentration of the protein-containing sample during the calcium phosphate precipitation reaction 1 to 10 times, 1 to 9 times, 1 to 8 times, 1 to 7 times, 1 to 6 times, 1 to 5 times, 1 to 4 times, 1 to 3 times, 1 to 2 times, or 1 to 1.5 times dilution, but is not limited thereto.
- the calcium phosphate precipitation reaction may be diluted 1 to 3 times the concentration of the protein-containing sample.
- the concentration of calcium ions added to the calcium phosphate precipitation reaction is 1 to 20 mM
- the concentration of phosphate ions is 1 to 20 mM
- the calcium phosphate precipitation reaction time is 3 to 5
- the calcium phosphate precipitation reaction may be diluted 1 to 3 times the concentration of the protein-containing sample.
- the concentration of calcium ions added to the calcium phosphate precipitation reaction is 1 to 20 mM
- the concentration of phosphate ions is 1 to 20 mM
- the calcium phosphate precipitation reaction is pH 5.5 to 7.5. Carried out in, and the calcium phosphate precipitation reaction time may be 3 to 5 hours.
- the concentration of calcium ions added to the calcium phosphate precipitation reaction is 1 to 20 mM
- the concentration of phosphate ions is 1 to 20 mM
- the calcium phosphate precipitation reaction is pH 5.5 to 7.5.
- the calcium phosphate precipitation reaction time is carried out at 3 to 5 hours, and the calcium phosphate precipitation reaction may be diluted 1 to 3 times the concentration of the protein-containing sample.
- the impurities include, but are not limited to, protein multimers, protein fragments, DNA contaminants, viruses, host cell derived proteins or protein A.
- the protein contained in the protein-containing sample to remove impurities may be a physiologically active protein, for example, the protein may be an antibody or a fusion protein, but is not limited thereto.
- the antibody may be a monoclonal antibody, for example, the antibody may be an anti-CD20 antibody or an anti-ErbB3 antibody.
- the fusion protein may be an Fc fusion protein.
- the Fc fusion protein may be a fusion protein of a TNF ⁇ receptor fragment and an antibody Fc fragment.
- the protein contained in the protein-containing sample to be purified by the method according to one embodiment of the present invention for example, monoclonal antibody, Fc fusion protein, granulocyte colony stimulating factor (G-CSF), granulocyte macrophage colony Hematopoietic factors such as stimulating factor (GM-CSF), erythropoietin (EPO), thrombopoietin, cytokines such as interferon, IL-1, IL-6, serum albumin, blood coagulant VIII factor, blood coagulant IX Factors include, but are not limited to, insulin, stem cell growth factor (SCF), and the like.
- G-CSF granulocyte colony stimulating factor
- GM-CSF granulocyte macrophage colony Hematopoietic factors
- EPO erythropoietin
- cytokines such as interferon
- IL-1 IL-6
- serum albumin serum albumin
- blood coagulant VIII factor blood coagulant I
- the protein to remove impurities may be a monoclonal antibody or an Fc fusion protein (eg, a TNFRc-Fc fusion protein).
- the purifiable antibody comprises all IgG, IgA, IgE, IgD, or IgM.
- physiologically active protein refers to a substance having a biological activity substantially the same as that of a physiologically active protein of a mammal, for example, a human being, or one obtained by natural or genetic recombination.
- Bioactive proteins by genetic recombination include bacterial cells such as E. coli; Chinese hamster ovary (CHO) cells, BHK cells, COS cells, cells derived from animals, such as human-derived cells can be prepared, and can be isolated and purified by various methods before use.
- Proteins obtained by genetic recombination include those having the same amino acid sequence as the native protein and those that retain the biological activity as deletions, substitutions, and additions of one or more amino acids in the amino acid sequence.
- physiologically active proteins include those chemically modified by PEG and the like.
- the monoclonal antibody may be produced by any method.
- the monoclonal antibody basically immunizes the sensitizing antigen according to a conventional immunization method using a known technique, fuses the obtained immune cells with known parental cells by a conventional cell fusion method, and then performs a conventional screening method. It can produce by screening antibody producing cells which are monoclonal.
- the antibody genes can be cloned from the hybridomas and inserted into a suitable vector and introduced into the host using gene recombination techniques, and the resulting recombinant antibodies can also be used in one embodiment of the present invention.
- cDNA of the variable region (V region) of an antibody is synthesize
- the DNA is linked to the DNA encoding the desired antibody constant region (C region) and inserted into the expression vector.
- the DNA encoding the V region of the antibody may be inserted into an expression vector containing the DNA of the antibody C region.
- the DNA construct is inserted into the expression vector so that it is expressed under the control of an expression control region, eg, an enhancer or a promoter.
- the host cell can be transformed with this expression vector to express the antibody.
- artificially modified genetic recombinant antibodies such as chimeric antibodies, humanized antibodies, etc.
- modified antibodies can be produced using a known method.
- a chimeric antibody is an antibody consisting of a variable region of a heavy chain and a light chain of a mammal other than human, for example, a mouse antibody, and a constant region of a heavy chain and a light chain of a human antibody, and the DNA encoding the variable region of a mouse antibody is invariant of a human antibody. It can be obtained by linking with a DNA encoding a region, inserting it into an expression vector, and introducing into a host to generate an antibody.
- Humanized antibodies are obtained by implanting a complementarity determining region (CDR) of a mammal other than a human, eg, a mouse antibody, into the complementarity determining region of a human antibody. Its standard genetic recombination procedures are also known. Specifically, a DNA sequence designed to connect the CDRs of a mouse antibody and a framework region (FR) of a human antibody is synthesized from several oligonucleotides prepared to have a portion overlapping with a terminal portion by the PCR method. The DNA thus obtained is linked with the DNA encoding the human antibody constant region, and then inserted into an expression vector, which is introduced into a host to generate an antibody.
- CDR complementarity determining region
- FR framework region
- the FRs of human antibodies linked via CDRs are chosen such that the complementarity determining regions form good antigen binding sites. If desired, amino acids of the framework regions of the variable regions of the antibody can be substituted such that the complementarity determining regions of the reconstituted humanized antibody form the appropriate antigen binding site.
- human lymphocytes are sensitized in vitro with cells expressing a desired antigen or a desired antigen, and sensitized lymphocytes are fused with human myeloma cells, eg, U266, to obtain a desired human antibody having binding activity to the antigen.
- a desired human antibody can be obtained by immunizing a transgenic animal having all repertoires of the human antibody gene with an antigen.
- techniques for obtaining human antibodies by panning using human antibody libraries are also known.
- a phage that expresses the variable region of a human antibody on the surface of the phage by the phage display method as a single-chain antibody (scFv) can be selected.
- scFv single-chain antibody
- a suitable expression vector can be prepared based on the sequence to obtain a human antibody.
- antibodies that can be used in one embodiment of the present invention also include antibody fragments such as Fab, (Fab ') 2 , Fc, Fc', Fd, and reconstituted antibodies such as monovalent or bivalent single-chain antibodies (scFv). .
- physiologically active protein-containing sample or "protein-containing sample” or “antibody-containing sample” means a mammalian cell such as a CHO cell containing a bioactive protein molecule or a protein or antibody molecule obtained by culturing. The culture medium or the thing which gave constant treatment, such as partial purification, to this.
- impurity includes any substance as long as it is a substance other than the protein of interest, and examples of impurities include protein aggregates, protein fragments, DNA contaminants, viruses, protein A (from columns). Eluted), host cell protein, endotoxin, Hy-Fish (FL) as a medium component, IGF, and the like, but are not limited thereto.
- protein aggregate refers to the binding of two or more proteins of interest.
- the binding is irreversible binding, the bound protein is not broken down into each.
- Protein multimers can produce neutralizing antibodies to the protein in the human body, thereby disappearing the efficacy for that protein. Therefore, it is an impurity to be removed in the purification process.
- protein fragment refers to a fragment protein of a protein that is poorly synthesized and degraded by proteolytic enzymes of that protein, incorrectly synthesized during the protein synthesis process. Means. They are considered impurities because they are ineffective.
- DNA contaminant means DNA in a biologically active protein-containing sample, and includes DNA from a host and DNA from a contaminated virus.
- DNA contaminant used herein as an example of impurities is not particularly limited and includes DNA viruses and RNA viruses.
- RNA viruses include retroviruses such as X-MuLV, Leo viruses such as Reo3, and parvoviruses such as MVM.
- viruses removed by the method of the present invention include, for example, X-MuLV, PRV, Reo3, MVM, VSV, herpes simplex, CHV, Sindbis, mumps, vaccinia, Measle, Rubella, influenza, herpes zoster , Cytomegalo, parainfluenza, EB, HIV, HA, HB, NANB, ATL, ECHO, and parvovirus.
- protein A is a protein A (leached protein A) leached from the Protein A chromatography resin, which is an impurity that can cause an exothermic reaction in the human body.
- DoE means developing an experimental plan to obtain "maximum information” with “minimum experiment” through statistical methods.
- the purpose of DoE is to organize factors that can be controlled to identify important factors and to find optimal conditions.
- the inventors planned a statistical experiment (DoE) using a statistical program (Design expert software. Ver. 7.1.1) and performed a statistical analysis (Analysis of variance, ANOVA, variance analysis).
- the analysis of variance (ANOVA) used in one embodiment of the present invention is used to compare two or more large groups in statistics between groups resulting from the variance in the group, the difference between the total mean and the mean of each group.
- the F distribution produced by comparing the variances, where the F distribution is the distribution ratio obtained by comparing the variances. This ratio is used to test whether there is a difference in the population variance of each group and whether the population mean is different.
- the statistical program Design expert software. Ver. 7.1.1 used for the statistical analysis is widely used for the development and production of pharmaceuticals in the pharmaceutical field.
- FIG. 1 illustrates a general antibody purification step.
- a calcium phosphate precipitation method according to an embodiment of the present invention may be applied instead of an additional 1 st or 2 nd chromatography step after the Protein A chromatography step.
- Figure 2 shows a purification preparation step for proceeding purification through calcium phosphate precipitation according to an embodiment of the present invention.
- Figure 3a and 3b is further purified through calcium phosphate precipitation reaction according to an embodiment of the present invention for the TNF ⁇ receptor-Fc fusion protein, the purified anti-ErbB3 antibody for analysis size exclusion chromatography (SEC-HPLC Results of analyzing antibody multimers
- Figures 4a and 4b is further purified through the calcium phosphate precipitation reaction according to an embodiment of the present invention for the anti-ErbB3 antibody, the purified TNF ⁇ receptor-Fc fusion protein for analysis size exclusion chromatography (SEC-HPLC This is the result of analyzing the protein multimer through
- the CHO (Chines Hamster Ovary Cell) cell culture containing an anti-CD20 antibody was purified by protein A chromatography (MabSelect SuRe Protein A chromatography, GE Healthcare) (see FIG. 2 (A)).
- the anti-CD20 antibody was eluted from the Protein A column with 50 mM acetate, pH 3.5 buffer, and the eluate was adjusted to pH 3.5 with 1M acetic acid and stored at room temperature for 1 hour. Thereafter, the pH 3.5 eluate was brought back to pH 8.0 with a trisbase, and further purified by anion chromatography (Q sepharose Fast Flow; manufactured by GE Healthcare).
- the pH 8.0 solution containing the antibody flows out without binding to the anion.
- purification was performed by a flow through method.
- the pH 8.0 solution containing the antibody released without binding was adjusted to pH 7.0 using 1 M acetic acid, and the intermediate purification step was further purified as in Example 2 using the calcium phosphate precipitation method of the present invention. did.
- TNFRc-Fc fusion proteins Animal cell cultures containing TNF ⁇ receptors and Fc fusion proteins (hereinafter referred to as TNFRc-Fc fusion proteins) were purified by protein A chromatography (see FIG. 2 (B)). TNFRc-Fc fusion protein was eluted from the Protein A column using 50 mM acetate, pH 3.5 buffer, and this eluate was brought to pH 7.0 using trisbase. Further purification was carried out as in Example 2 using the precipitation method.
- each culture solution containing an anti-CD20 antibody or TNFRc-Fc fusion protein produced using animal cells, such as antibody or protein multimer was conducted by planning the experiment as shown in Table 1 using a statistical program (Design expert software ver. 7.1.1 of Stat-Ease, Inc.).
- the antibody concentration was calculated by substituting the formula 1 below by measuring the absorbance at UV280.
- Antibody monomer ratio was determined by analytical size exclusion chromatography (Tosoh's TSKgel G3000SWL Size Exclusion Chromatography Column). High Molecular Weight, Antibody Monomer and Low Molecular Weight (Low Molecular Weight).
- the anti-CD20 antibody concentration before dilution was 4.8906 mg / mL.
- TNFRc-Fc fusion protein concentration before dilution was 4.70 mg / mL.
- the segment ratio for the initial TNFRc-Fc fusion protein was 0.70%.
- factors affecting the removal of the anti-CD20 antibody multimers are the added calcium ion concentration, reaction time, the concentration of anti-CD20 antibody, the correlation between the added phosphate and calcium ion concentrations, and the added phosphate ion concentration.
- the yield of TNFRc-Fc fusion protein is affected by the correlation between the added phosphate concentration, the added calcium ion concentration, and the added forest ion concentration and calcium ion concentration.
- the factors affecting the removal of TNFRc-Fc fusion protein multimers were the concentration of added phosphate concentration, added calcium ion concentration, reaction time, TNFRc-Fc fusion protein concentration, added phosphate ion concentration and calcium ion concentration.
- the pre-dilution anti-CD20 antibody and TNFRc-Fc fusion protein concentrations were 4.8906 mg / mL and 4.7047 mg / mL, respectively.
- the anti-CD20 antibody concentration before dilution was 4.8906 mg / mL, respectively.
- the anti-CD20 antibody yield was determined by the reaction pH ( P ⁇ 0.0009), the concentration of anti-CD20 antibody ( P ⁇ 0.0058), There was statistical significance of P ⁇ 0.05 for the correlation ( P ⁇ 0.0047) of the anti-CD20 antibody.
- the anti-CD20 antibody yield in the purification method of anti-CD20 antibody by calcium phosphate precipitation, in addition to the added calcium and phosphate ions, the concentration of the anti-CD20 antibody, reaction pH, and anti-CD20 antibody concentration and reaction pH Affected by the interrelationship of
- the degree of anti-CD20 antibody multimer removal in the purification method of the anti-CD20 antibody by calcium phosphate precipitation is influenced by the concentration reaction pH of the anti-CD20 antibody in addition to the added calcium and phosphate ions.
- TNFRc-Fc fusion protein concentration before dilution was 4.7047 mg / mL.
- the yield of TNFRc-Fc fusion protein was calculated as the concentration of TNFRc-Fc fusion protein ( P ⁇ 0.0107), reaction pH ( P ⁇ 0.0011).
- the yield of TNFRc-Fc fusion protein in the purification method of TNFRc-Fc fusion protein by calcium phosphate precipitation, in addition to the calcium and phosphate ions added the concentration, reaction pH, reaction time and TNFRc-Fc of TNFRc-Fc fusion protein
- the relationship between the fusion protein, the TNFRc-Fc fusion protein concentration and the reaction pH, and the reaction time, the concentration of the TNFRc-Fc fusion protein, and the reaction pH are affected.
- TNFRc-Fc fusion protein multimers when the phosphate concentration and the CaCl 2 concentration were fixed at 10 mM through analytical size exclusion chromatography (SEC) results for the TNFRc-Fc fusion protein, respectively. Scavenging ability was determined by the concentration of TNFRc-Fc fusion protein ( P ⁇ 0.0004), reaction pH ( P ⁇ 0.0001), correlation between reaction time and TNFRc-Fc fusion protein concentration ( P ⁇ 0.007), and correlation between reaction time and reaction pH.
- TNFRc-Fc fusion protein multimer removal in the purification method of TNFRc-Fc fusion protein by calcium phosphate precipitation method in addition to the added calcium and phosphate ions, TNFRc-Fc fusion protein concentration, reaction pH, reaction time and The relationship between TNFRc-Fc fusion protein concentration, reaction time and reaction pH, TNFRc-Fc fusion protein concentration and half pH, and reaction time and TNFRc-Fc fusion protein concentration, and reaction pH get affected.
- TNF ⁇ receptor-Fc (TNFRc-Fc) fusion protein tested in the above Example was subjected to calcium phosphate precipitation reaction under the conditions shown in Table 17, and the multimer weight of the TNFRc-Fc fusion protein before and after the reaction was compared.
- Example 9 Test to confirm the effect of removing impurities other than the protein multimer by calcium phosphate precipitation
- the purification method using calcium phosphate precipitation as shown in Table 20, is effective in removing host cell-derived protein, DNA, and protein A residues in addition to removing multimers of antibodies.
- the yield, multimer removal capacity, and impurities removal ability were different depending on the protein.
- the reaction time, the added calcium ion concentration, the added phosphate ion concentration means that the conditions may vary depending on the protein.
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Biochemistry (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Genetics & Genomics (AREA)
- Biophysics (AREA)
- Molecular Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Analytical Chemistry (AREA)
- Immunology (AREA)
- Peptides Or Proteins (AREA)
- Engineering & Computer Science (AREA)
- Water Supply & Treatment (AREA)
Abstract
Description
본 발명은 칼슘인산염 침전을 이용한 불순물 제거 방법에 관한 것으로, 더욱 상세하게는 단백질 함유 시료에 칼슘 이온 용액과 인산 이온 용액을 첨가하여 칼슘인산염 침전 반응을 유발하여 불순물을 제거하는 방법에 관한 것이다. The present invention relates to a method for removing impurities using calcium phosphate precipitation, and more particularly, to a method for removing impurities by adding a calcium ion solution and a phosphate ion solution to a protein-containing sample to cause a calcium phosphate precipitation reaction.
칼슘인산염 침전(Calcium phosphate precipitation)을 이용한 레진 (예, Ceramic Hydroxyapatite resin, Hydroxyapatite resin: 미국 BioRad사 제품) 크로마토그래피는 단백질을 해당 레진에 특정 완충 환경에서 결합시키거나 떨어뜨려, 결합력의 차이에 따라 단백질을 분리하는 방법이다. Resin using calcium phosphate precipitation (e.g., Ceramic Hydroxyapatite resin, Hydroxyapatite resin: manufactured by BioRad, USA) chromatography binds or drops proteins to the resin in a specific buffered environment. How to separate.
세라믹 하이드록시아파타이트(Ceramic Hydroxyapatite) 또는 하이드록시아파타이트 크로마토그래피(Hydroxyapatite chromatography)를 이용할 경우, 분자량이 큰 단백질 다량체(Aggregate), DNA, 바이러스, 숙주세포 유래 단백질(Host cell protein), 단백질 A(Protein A) 등을 제거할 수 있다. When using ceramic hydroxyapatite or hydroxyapatite chromatography, high molecular weight protein aggregates, DNA, viruses, host cell proteins, protein A (Protein) A) can be removed.
하지만, 칼슘인산 침전을 이용한 레진 크로마토그래피의 경우 다음과 같은 이유에서 비용과 시간이 많이 소요된다. 생산에서 컬럼을 이용한 단백질 정제 업무를 진행하기 위해서는 우선적으로 컬럼, 크로마토그래피 장비 구매 및 이에 대한 Qualification를 수행해야 한다. 또한, 크로마토그래피를 수행하기 위해서는 많은 종류와 많은 양의 버퍼(buffer)가 필요하다. 특히, 크로마토그래피를 수행 준비(column packing, cleaning, buffer preparation, documentation)하고 진행하는데 많은 시간이 소요된다. However, resin chromatography using calcium phosphate precipitation is costly and time consuming for the following reasons. In order to proceed with protein purification using columns in production, it is necessary to first purchase and qualify columns and chromatography equipment. In addition, in order to perform chromatography, many kinds and a large amount of buffers are required. In particular, it takes a long time to prepare and perform chromatography (column packing, cleaning, buffer preparation, documentation).
본 발명자들은 레진을 이용한 컬럼 크로마토그래피를 사용하지 않고, 단백질 함유 시료에 칼슘 이온(Ca2+) 용액과 인산 이온(PO4 3-) 용액을 첨가함으로써, 칼슘인산염 침전물(Calcium phosphate precipitate, Ca5(PO4)3(OH), Hydroxyapatite)을 유발하여 불순물을 제거하는 방법을 안출하였다. 칼슘인산 침전이 형성되면, 단백질은 칼슘인산 침전물과 흡착이 되어 함께 침전이 이뤄진다. 이때 칼슘인산 침전물과 결합하는 단백질의 종류는 첨가 시 칼슘인산염의 농도에 따라 달라진다. 따라서, 원하는 단백질을 칼슘과 인산염의 농도에 따라 분리할 수 있다. 본 방법은 위에서 언급한 크로마토그래피 컬럼을 이용한 단백질 정제 시와 같이 많은 장비, 완충액, 그리고 긴 시간이 필요하지 않게 된다. The present inventors added calcium phosphate precipitates (Calcium phosphate precipitate, Ca 5 ) by adding calcium ion (Ca 2+ ) solution and phosphate ion (PO 4 3- ) solution to protein-containing samples without using column chromatography using resin. (PO 4 ) 3 (OH), Hydroxyapatite) was induced to devise a method for removing impurities. When calcium phosphate precipitate is formed, the protein is adsorbed with the calcium phosphate precipitate to precipitate together. The type of protein that binds to calcium phosphate precipitates depends on the concentration of calcium phosphate when added. Thus, the desired protein can be separated according to the concentration of calcium and phosphate. The method does not require as much equipment, buffers and long time as the protein purification using the chromatography column mentioned above.
결과적으로 단백질 정제에 있어 칼슘인산염 침전을 이용하면 불편하고 시간과 비용이 많이 드는 컬럼 크로마토그래피 공정을 대체할 수 있음을 확인하고 본 발명을 완성하였다. As a result, it was confirmed that the use of calcium phosphate precipitation in protein purification could replace the uncomfortable, time-consuming and costly column chromatography process, and thus completed the present invention.
따라서 본 발명이 해결하고자 하는 과제는 단백질 함유 시료에 칼슘 이온 용액과 인산 이온 용액을 첨가하여 칼슘인산염 침전 반응을 형성하는 단계를 포함하는, 단백질 함유 시료 중의 불순물을 제거하는 방법을 제공하는 것이다. Accordingly, an object of the present invention is to provide a method for removing impurities in a protein-containing sample, comprising the step of forming a calcium phosphate precipitation reaction by adding a calcium ion solution and a phosphate ion solution to a protein-containing sample.
본 발명이 해결하고자 하는 다른 과제는 상기 방법을 이용한 정제 공정을 포함하는 단백질 제제의 제조 방법을 제공하는 것이다. Another problem to be solved by the present invention is to provide a method for producing a protein formulation comprising a purification step using the above method.
상기 과제를 해결하고자, 본 발명의 일 구체예는 하기 단계를 포함하는 단백질 함유 시료 중의 불순물을 제거하는 방법을 제공한다:In order to solve the above problems, one embodiment of the present invention provides a method for removing impurities in a protein-containing sample comprising the following steps:
a) 단백질 함유 시료를 단백질 A 친화성 크로마토그래피(protein A affinity chromatography)에 적용하여 정제하는 단계;a) purifying the protein-containing sample by applying it to Protein A affinity chromatography;
b) 상기 정제된 단백질에 칼슘 이온 용액과 인산 이온 용액을 첨가하여 칼슘인산염 침전 반응을 형성한 후 원심분리 또는 여과를 통해 침전물을 제거하는 단계; 및 b) adding a calcium ion solution and a phosphate ion solution to the purified protein to form a calcium phosphate precipitation reaction, and then removing the precipitate by centrifugation or filtration; And
c) 상기 단계 b) 이후, 또는 상기 단계 a)와 단계 b) 사이에 전 단계에서 정제된 단백질을 이온교환 크로마토그래피(ion exchange chromatography), 소수성 상호작용 크로마토그래피(hydrophobic interaction chromatography) 및 혼합 모드 크로마토그래피(mixed mode chromatography)로 구성된 군에서 선택된 어느 하나의 크로마토그래피에 적용하여 정제하는 단계. c) ion exchange chromatography, hydrophobic interaction chromatography and mixed mode chromatography of the purified protein after step b) or between steps a) and b). Purifying by applying to any one of the chromatography selected from the group consisting of mixed mode chromatography.
또한, 본 발명의 다른 일 구체예는 상기 방법을 이용한 정제 공정을 포함하는 단백질 제제의 제조 방법을 제공한다. In addition, another embodiment of the present invention provides a method for producing a protein formulation comprising a purification step using the above method.
이하 본 발명을 더욱 상세히 설명한다. Hereinafter, the present invention will be described in more detail.
일반적으로 항체의 정제공정은 한 단계의 정제만으로 높은 순도와 수율을 얻을 수 있는 단백질 A 크로마토그래피(Protein A chromatography)를 기초로 한다. (Fahrner RL, Knudsen HL, Basey CD, Galan W, Feuerhelm D, Vanderlaan M, et al. Industrial purification of pharmaceutical antibodies: development, operation and validation of chromatography processes. Biotechnol Genet Eng Rev 2001;18:301-27 참조). 원하는 순도와 목적의 단백질을 얻기 위해서는 단백질 A 크로마토그래피 단계 이후에는 분자량이 큰 단백질 다량체, DNA, 바이러스, 숙주세포 유래 단백질, 침출된(leached) 단백질 A와 불순물들을 제거하기 위해, 이온교환 크리마토그래피 (Ion-exchange chromatography), 소수성 크로마토그리피 (Hydrophobic interaction chromatography), 혼합 모드 크로마토그래피(Mixed mode chromatography) 등의 추가적인 정제 단계가 활용이 된다 (Liu HF, Ma J, Winter C, Bayer R. Recovery and purification process development for monoclonal antibody production. mAbs. 2010;2:480-499 참조).In general, the purification process of the antibody is based on Protein A chromatography, which can obtain high purity and yield with only one step purification. (Fahrner RL, Knudsen HL, Basey CD, Galan W, Feuerhelm D, Vanderlaan M, et al. Industrial purification of pharmaceutical antibodies: development, operation and validation of chromatography processes. Biotechnol Genet Eng Rev 2001; 18: 301-27) . To obtain the desired purity and protein of interest, after the Protein A chromatography step, ion-exchange creams are used to remove high molecular weight protein multimers, DNA, viruses, host cell-derived proteins, leached Protein A and impurities. Additional purification steps such as ion-exchange chromatography, hydrophobic interaction chromatography, and mixed mode chromatography can be utilized (Liu HF, Ma J, Winter C, Bayer R. Recovery and purification process development for monoclonal antibody production.mAbs. 2010; 2: 480-499).
본 발명의 일 구체예에 따르면, 단백질 A 크로마토그래피 단계 이후에, 설비와 시간이 많이 소모되는 추가적인 크로마토그래피를 대신하여 칼슘인산염 침전방법을 사용할 수 있다. 즉, 칼슘인산염 침전방법을 위 추가적인 크로마토그래피 대신 적용함으로써, 분자량이 큰 단백질 다량체, DNA, 바이러스, 숙주세포 유래 단백질, 단백질 A 등을 짧은 시간 내에 적은 노동력으로 효과적으로 제거할 수 있다. According to one embodiment of the invention, after the Protein A chromatography step, calcium phosphate precipitation may be used in place of equipment and time-consuming additional chromatography. In other words, by applying the calcium phosphate precipitation method instead of the above additional chromatography, large molecular weight protein multimers, DNA, viruses, host cell-derived protein, Protein A, etc. can be effectively removed in a short time with little labor.
본 발명의 일 구체예는 도 1에 나타낸 바와 같이, 일반적인 단백질 정제 단계에서 단백질 A 크로마토그래피 단계 이후에, 추가적인 1st 또는 2nd 크로마토그래피 단계 대신에 칼슘인산염 침전 방법을 적용하여, 추가적인 크로마토그래피 단계가 가지고 있는 정제효과를 얻는 방법이다.One embodiment of the present invention, as shown in Figure 1, after the Protein A chromatography step in the general protein purification step, by applying a calcium phosphate precipitation method instead of an additional 1 st or 2 nd chromatography step, an additional chromatography step It is a way to get the purification effect.
본 발명의 칼슘인산염 침전 방법을 일 예를 들어 설명하면 다음과 같다. When explaining the calcium phosphate precipitation method of the present invention as an example.
동물세포를 이용하여 항체를 생산하고, 그 세포배양액에 포함되어 있는 항체를 정제하기 위해, 원심분리(Centrifugation)와 여과(Filtration)를 통해 세포들과 세포 잔여물을 제거하고, 여과된 세포배양액을 평형상태의 단백질 A 컬럼(Protein A column)에 주입(Loading)하여 결합시킨 후, 다시 평형용액과 세척 후, 낮은 pH 용출용액으로 단백질 A 컬럼으로부터 항체를 용출한다. 용출된 항체용액을 낮은 pH 바이러스 불활성화를 위해, pH 3.6으로 적정하고 실온에서 1시간 동안 보관한다. 그 후, 항체 용액을 pH 6.0으로 중화시킨다. 중화된 항체용액을 음이온교환(Q sepharose Fast Flow; GE Healthcare사 제품) 크로마토그래피를 통해 추가 정제 후, 음이온 교환 크로마토그래피로부터 얻은 항체 중간 정제용액을 pH 7.0으로 조절 후, 100 mM CaCl2 와 100 mM sodium phosphate pH 7.0을 최종 농도가 10 mM Ca2+와 10 mM phosphate가 되게 항체 중간 정제용액에 적정하여 침전을 유발시킨다. 적정 후 실온에서 3 시간 동안 유지시켜 충분하게 칼슘인산염 침전물 (Hydroxyapatite, Ca5(PO4)3(OH))과 항체 다량체 및 다른 불순물들(DNA, 숙주세포 유래 단백질, Protein A 등)이 결합하게 한다. 그 후, 항체용액으로부터 침전물들을 제거하기 위해 원심분리 또는 여과를 진행한다. In order to produce antibodies using animal cells, and to purify the antibodies contained in the cell culture medium, the cells and cell residues are removed by centrifugation and filtration, and the filtered cell culture medium is removed. After loading and binding to the Protein A column in equilibrium, the solution is again washed with the equilibrium solution, and the antibody is eluted from the Protein A column with a low pH elution solution. The eluted antibody solution is titrated to pH 3.6 and stored at room temperature for 1 hour for low pH virus inactivation. Thereafter, the antibody solution is neutralized to pH 6.0. The neutralized antibody solution was further purified by anion exchange (Q sepharose Fast Flow; manufactured by GE Healthcare), and the intermediate antibody solution obtained from anion exchange chromatography was adjusted to pH 7.0, followed by 100 mM CaCl 2 and 100 mM. Sodium phosphate pH 7.0 is titrated in the intermediate purified solution to a final concentration of 10 mM Ca 2+ and 10 mM phosphate to cause precipitation. After titration, it is maintained at room temperature for 3 hours to sufficiently combine calcium phosphate precipitate (Hydroxyapatite, Ca 5 (PO 4 ) 3 (OH)) with antibody multimers and other impurities (DNA, host cell-derived protein, Protein A, etc.). Let's do it. Thereafter, centrifugation or filtration is performed to remove the precipitates from the antibody solution.
또한, 위와 동일한 방법으로 단백질 A 크로마토그래피와 낮은 pH 바이러스 불활성화 후에 중화시키고, 음이온 교환 크로마토그래피 앞서, 중간 정제 항체 용액에 칼슘인산 침전방법을 적용할 수 있다. 그 후, 여과된 항체 용액을 음이온 교환 크로마토그래피를 진행할 수 있다. In addition, by neutralizing protein A chromatography and low pH virus inactivation in the same manner as above, prior to anion exchange chromatography, calcium phosphate precipitation may be applied to the intermediate purified antibody solution. The filtered antibody solution can then be subjected to anion exchange chromatography.
즉, 본 발명의 일 구체예에 따른 칼슘인산염 침전을 이용한 불순물 제거 방법은 단백질 A 크로마토그래피 이후 단계에서, 순서와 크로마토그래피 기술(예, 이온교환 크로마토그래피, 소수성 상호작용 크로마토그래피, 혼합 모드 크로마토그래피 등)에 상관없이 단백질(항체 포함) 정제 중간단계의 크로마토그래피 대신에 적용이 가능하다. In other words, the impurity removal method using calcium phosphate precipitation according to an embodiment of the present invention is a step after the protein A chromatography, the sequence and chromatography techniques (eg, ion exchange chromatography, hydrophobic interaction chromatography, mixed mode chromatography) It is possible to apply instead of intermediate chromatography purification of proteins (including antibodies).
따라서 본 발명에 따른 일 구체예로서, 본 발명은 단백질 함유 시료에 칼슘 이온 용액과 인산 이온 용액을 첨가하여 칼슘인산염 침전 반응을 형성하는 단계를 포함하는, 단백질 함유 시료 중의 불순물을 제거하는 방법을 제공한다. 그리고, 상기 칼슘인산염 침전 반응 단계 이후 원심분리 또는 여과를 통해 침전물을 제거하는 단계를 추가로 포함할 수 있다.Therefore, in one embodiment according to the present invention, the present invention provides a method for removing impurities in a protein-containing sample, comprising adding a calcium ion solution and a phosphate ion solution to a protein-containing sample to form a calcium phosphate precipitation reaction. do. And, after the calcium phosphate precipitation reaction step may further comprise the step of removing the precipitate by centrifugation or filtration.
또한, 본 발명의 일 구체예에서, 상기 칼슘인산염 침전 반응 단계 이전에 단백질 함유 시료를 단백질 A 흡착 크로마토그래피에 적용하여 정제하는 단계를 추가로 포함할 수 있다. In addition, in one embodiment of the present invention, prior to the calcium phosphate precipitation reaction step may further comprise the step of applying a protein containing sample to protein A adsorption chromatography to purify.
또한, 본 발명의 일 구체예에서, 상기 칼슘인산염 침전 반응 단계 이후, 또는 상기 단백질 A 흡착 크로마토그래피 정제 단계 이후와 상기 칼슘인산염 침전 반응 단계 이전에 전 단계에서 정제된 단백질 함유 시료를 추가로 정제하는 단계를 포함할 수 있다. 상기 추가 정제하는 단계에서, 이온교환 크로마토그래피, 소수성 상호작용 크로마토그래피 및 혼합 모드 크로마토그래피로 구성된 군에서 선택된 어느 하나의 크로마토그래피에 적용하여 정제할 수 있으나, 이에 한정되는 것은 아니다. Further, in one embodiment of the present invention, further purification of the protein-containing sample purified in the previous step after the calcium phosphate precipitation reaction step, or after the Protein A adsorption chromatography purification step and before the calcium phosphate precipitation reaction step It may include a step. In the further purification step, purification may be applied to any one of chromatography selected from the group consisting of ion exchange chromatography, hydrophobic interaction chromatography, and mixed mode chromatography, but is not limited thereto.
일 예로, 상기 이온교환 크로마토그래피는 양이온 이온교환 크로마토그래피 또는 음이온 이온교환 크로마토그래피일 수 있다. For example, the ion exchange chromatography may be cation ion exchange chromatography or anion ion exchange chromatography.
일 예로, 상기 소수성 상호작용 크로마토그래피는 Ether (예; Toyopearl Ether-650M, Toyopearl Ether-650S 등), Butyl(예; Butyl sepharose, Butyl S sepharose, Catpo Butyl 등), Hexyl (예; Toyopearl Hexyl-650C 등), Octyl (예; Octyl sepharose 등), Pheyl(예; Toyopearl Phenyl-650M, Phenyl sepharose, Catpo Phenyl, Fractogel® EMD Phenyl (S) 등), Propyl(예; Fractogel® EMD Propyl(S) 등) 등의 리간드(ligand)를 포함할 수 있으나, 이에 한정되는 것은 아니다. For example, the hydrophobic interaction chromatography may be Ether (eg, Toyopearl Ether-650M, Toyopearl Ether-650S, etc.), Butyl (eg, Butyl sepharose, Butyl S sepharose, Catpo Butyl, etc.), Hexyl (eg, Toyopearl Hexyl-650C). ), Octyl (e.g. Octyl sepharose, etc.), Pheyl (e.g. Toyopearl Phenyl-650M, Phenyl sepharose, Catpo Phenyl, Fractogel ® EMD Phenyl (S), etc.), Propyl (e.g., Fractogel ® EMD Propyl (S), etc.) Ligand, such as, but may be included, but is not limited thereto.
일 예로, 상기 혼합 모드 크로마토그래피는 이온교환 기능과 소수성 상호작용 기능의 혼합 모드 크로마토그래피(예; Capto adhere, Capto MMC, MEP HyperCell, Eshmuno HCX 등), 음이온 교환기능과 양이온교환 기능의 혼합 모드 크로마토그래피(예; hydroxyapatite, Ceramic hydroxyapate 등) 등을 포함할 수 있으나, 이에 한정되는 것은 아니다. For example, the mixed mode chromatography may be a mixed mode chromatography (eg, Capto adhere, Capto MMC, MEP HyperCell, Eshmuno HCX, etc.), an ion exchange function, and a cation exchange function. Glyphography (eg, hydroxyapatite, Ceramic hydroxyapate, etc.) may be included, but is not limited thereto.
또한, 본 발명의 일 구체예는 하기 단계를 포함하는 단백질 함유 시료 중의 불순물을 제거하는 방법을 제공한다:In addition, one embodiment of the present invention provides a method for removing impurities in a protein containing sample comprising the following steps:
a) 단백질 함유 시료를 단백질 A 친화성 크로마토그래피(protein A affinity chromatography)에 적용하여 정제하는 단계;a) purifying the protein-containing sample by applying it to Protein A affinity chromatography;
b) 상기 정제된 단백질에 칼슘 이온 용액과 인산 이온 용액을 첨가하여 칼슘인산염 침전 반응을 형성한 후 원심분리 또는 여과를 통해 침전물을 제거하는 단계; 및 b) adding a calcium ion solution and a phosphate ion solution to the purified protein to form a calcium phosphate precipitation reaction, and then removing the precipitate by centrifugation or filtration; And
c) 상기 단계 b) 이후, 또는 상기 단계 a)와 단계 b) 사이에 전 단계에서 정제된 단백질을 이온교환 크로마토그래피(ion exchange chromatography), 소수성 상호작용 크로마토그래피(hydrophobic interaction chromatography) 및 혼합 모드 크로마토그래피(mixed mode chromatography)로 구성된 군에서 선택된 어느 하나의 크로마토그래피에 적용하여 정제하는 단계. c) ion exchange chromatography, hydrophobic interaction chromatography and mixed mode chromatography of the purified protein after step b) or between steps a) and b). Purifying by applying to any one of the chromatography selected from the group consisting of mixed mode chromatography.
본 발명의 일 구체예에서, 상기 단백질 함유 시료가 인산 이온을 포함한 시료인 경우 칼슘이온 용액만을 첨가할 수 있고, 반대로 칼슘이온을 포함한 시료인 경우 인산이온 용액만을 첨가할 수 있다. In one embodiment of the present invention, when the protein-containing sample is a sample containing phosphate ions can be added only calcium ion solution, on the contrary, in the case of a sample containing calcium ion it can be added only a phosphate solution.
본 발명의 일 구체예에서, 상기 칼슘 이온 용액은 CaCl2, 또는 CaCO3 일 수 있으나, 이것에 한정되는 것은 아니다. In one embodiment of the present invention, the calcium ion solution may be CaCl 2 , or CaCO 3 , but is not limited thereto.
본 발명의 일 구체예에서, 상기 인산 이온 용액은 Na3PO4, NaH2PO4, Na2HPO4 또는 KH2PO4, K2HPO4, K3PO4 일 수 있으나, 이것에 한정되는 것은 아니다. In one embodiment of the present invention, the phosphate ion solution may be Na 3 PO 4 , NaH 2 PO 4 , Na 2 HPO 4 or KH 2 PO 4, K 2 HPO 4, K 3 PO 4 , but is not limited thereto. It is not.
본 발명의 일 구체예에서, 상기 칼슘인산염 침전 반응에 첨가된 칼슘 이온의 농도는 1 내지 100 mM, 1 내지 90 mM, 1 내지 80 mM, 1 내지 70 mM, 1 내지 60 mM, 1 내지 50 mM, 1 내지 40 mM, 1 내지 30 mM, 1 내지 20 mM, 1 내지 10 mM, 또는 1 내지 5 mM일 수 있으나, 이에 한정되는 것은 아니다. 일 예로, 상기 칼슘인산염 침전 반응에 첨가된 칼슘 이온의 농도는 1 내지 20 mM일 수 있다. In one embodiment of the present invention, the concentration of calcium ions added to the calcium phosphate precipitation reaction is 1 to 100 mM, 1 to 90 mM, 1 to 80 mM, 1 to 70 mM, 1 to 60 mM, 1 to 50 mM , 1 to 40 mM, 1 to 30 mM, 1 to 20 mM, 1 to 10 mM, or 1 to 5 mM, but is not limited thereto. For example, the concentration of calcium ions added to the calcium phosphate precipitation reaction may be 1 to 20 mM.
본 발명의 일 구체예에서, 상기 칼슘인산염 침전 반응에 첨가된 인산 이온의 농도는 1 내지 100 mM, 1 내지 90 mM, 1 내지 80 mM, 1 내지 70 mM, 1 내지 60 mM, 1 내지 50 mM, 1 내지 40 mM, 1 내지 30 mM, 1 내지 20 mM, 1 내지 10 mM, 또는 1 내지 5 mM일 수 있으나, 이에 한정되는 것은 아니다. 일 예로, 상기 칼슘인산염 침전 반응에 첨가된 인산 이온의 농도는 1 내지 20 mM일 수 있다.In one embodiment of the present invention, the concentration of phosphate ions added to the calcium phosphate precipitation reaction is 1 to 100 mM, 1 to 90 mM, 1 to 80 mM, 1 to 70 mM, 1 to 60 mM, 1 to 50 mM , 1 to 40 mM, 1 to 30 mM, 1 to 20 mM, 1 to 10 mM, or 1 to 5 mM, but is not limited thereto. For example, the concentration of phosphate ions added to the calcium phosphate precipitation reaction may be 1 to 20 mM.
본 발명의 일 구체예에 따르면, 상기 칼슘인산염 침전 반응에 첨가된 칼슘 이온의 농도는 1 내지 100 mM이고, 인산 이온의 농도는 1 내지 100 mM일 수 있다. According to one embodiment of the present invention, the concentration of calcium ions added to the calcium phosphate precipitation reaction may be 1 to 100 mM, and the concentration of phosphate ions may be 1 to 100 mM.
본 발명의 다른 일 구체예에 따르면, 상기 칼슘인산염 침전 반응에 첨가된 칼슘 이온의 농도는 1 내지 20 mM이고, 인산 이온의 농도는 1 내지 20 mM일 수 있다. According to another embodiment of the present invention, the concentration of calcium ions added to the calcium phosphate precipitation reaction may be 1 to 20 mM, and the concentration of phosphate ions may be 1 to 20 mM.
본 발명의 일 구체예에서, 상기 칼슘인산염 침전 반응은 pH 5.0 내지 10.0, pH 5.0 내지 9.5, pH 5.0 내지 9.0, pH 5.0 내지 8.5, pH 5.0 내지 8.0, pH 5.0 내지 7.5, pH 5.0 내지 7.0, pH 5.0 내지 6.5, pH 5.0 내지 6.0, 또는 pH 5.0 내지 5.5에서 수행할 수 있으나, 이에 한정되는 것은 아니다. In one embodiment of the present invention, the calcium phosphate precipitation reaction is pH 5.0 to 10.0, pH 5.0 to 9.5, pH 5.0 to 9.0, pH 5.0 to 8.5, pH 5.0 to 8.0, pH 5.0 to 7.5, pH 5.0 to 7.0, pH It may be performed at 5.0 to 6.5, pH 5.0 to 6.0, or pH 5.0 to 5.5, but is not limited thereto.
본 발명의 일 구체예에 따르면, 상기 칼슘인산염 침전 반응은 pH 5.5 내지 7.5에서 수행할 수 있다. According to one embodiment of the invention, the calcium phosphate precipitation reaction may be carried out at pH 5.5 to 7.5.
본 발명의 다른 일 구체예에 따르면, 상기 칼슘인산염 침전 반응에 첨가된 칼슘 이온의 농도는 1 내지 20 mM이고, 인산 이온의 농도는 1 내지 20 mM이고, 상기 칼슘인산염 침전 반응은 pH 5.5 내지 7.5에서 수행할 수 있다. According to another embodiment of the present invention, the concentration of calcium ions added to the calcium phosphate precipitation reaction is 1 to 20 mM, the concentration of phosphate ions is 1 to 20 mM, and the calcium phosphate precipitation reaction is pH 5.5 to 7.5. This can be done at
본 발명의 일 구체예에서, 상기 칼슘인산염 침전 반응 시간은 0 내지 10 시간, 0 내지 9 시간, 0 내지 8 시간, 0 내지 7 시간, 0 내지 6 시간, 0 내지 5 시간, 0 내지 4 시간, 0 내지 3 시간, 0 내지 2 시간, 0 내지 1 시간, 또는 0 내지 0.5 시간일 수 있으나, 이에 한정되는 것은 아니다. In one embodiment of the present invention, the calcium phosphate precipitation reaction time is 0 to 10 hours, 0 to 9 hours, 0 to 8 hours, 0 to 7 hours, 0 to 6 hours, 0 to 5 hours, 0 to 4 hours, 0 to 3 hours, 0 to 2 hours, 0 to 1 hour, or 0 to 0.5 hours, but is not limited thereto.
본 발명의 일 구체예에 따르면, 상기 칼슘인산염 침전 반응 시간은 3 내지 5 시간일 수 있다. According to one embodiment of the present invention, the calcium phosphate precipitation reaction time may be 3 to 5 hours.
본 발명의 다른 일 구체예에 따르면, 상기 칼슘인산염 침전 반응에 첨가된 칼슘 이온의 농도는 1 내지 20 mM이고, 인산 이온의 농도는 1 내지 20 mM이고, 상기 칼슘인산염 침전 반응 시간은 3 내지 5 시간일 수 있다. According to another embodiment of the present invention, the concentration of calcium ions added to the calcium phosphate precipitation reaction is 1 to 20 mM, the concentration of phosphate ions is 1 to 20 mM, and the calcium phosphate precipitation reaction time is 3 to 5 It can be time.
본 발명의 일 구체예에서, 상기 칼슘인산염 침전 반응시 단백질 함유 시료의 농도를 1배 내지 10배, 1배 내지 9배, 1배 내지 8배, 1배 내지 7배, 1배 내지 6배, 1배 내지 5배, 1배 내지 4배, 1배 내지 3배, 1배 내지 2배, 또는 1배 내지 1.5배 희석할 수 있으나, 이에 한정되는 것은 아니다.In one embodiment of the present invention, the concentration of the protein-containing sample during the calcium
본 발명의 일 구체예에 따르면, 상기 칼슘인산염 침전 반응시 단백질 함유 시료의 농도를 1배 내지 3배 희석할 수 있다. According to one embodiment of the invention, the calcium phosphate precipitation reaction may be diluted 1 to 3 times the concentration of the protein-containing sample.
본 발명의 다른 일 구체예에 따르면, 상기 칼슘인산염 침전 반응에 첨가된 칼슘 이온의 농도는 1 내지 20 mM이고, 인산 이온의 농도는 1 내지 20 mM이고, 상기 칼슘인산염 침전 반응 시간은 3 내지 5 시간이고, 상기 칼슘인산염 침전 반응시 단백질 함유 시료의 농도를 1배 내지 3배 희석할 수 있다. According to another embodiment of the present invention, the concentration of calcium ions added to the calcium phosphate precipitation reaction is 1 to 20 mM, the concentration of phosphate ions is 1 to 20 mM, and the calcium phosphate precipitation reaction time is 3 to 5 In time, the calcium phosphate precipitation reaction may be diluted 1 to 3 times the concentration of the protein-containing sample.
본 발명의 다른 일 구체예에 따르면, 상기 칼슘인산염 침전 반응에 첨가된 칼슘 이온의 농도는 1 내지 20 mM이고, 인산 이온의 농도는 1 내지 20 mM이고, 상기 칼슘인산염 침전 반응은 pH 5.5 내지 7.5에서 수행하고, 상기 칼슘인산염 침전 반응 시간은 3 내지 5 시간일 수 있다. According to another embodiment of the present invention, the concentration of calcium ions added to the calcium phosphate precipitation reaction is 1 to 20 mM, the concentration of phosphate ions is 1 to 20 mM, and the calcium phosphate precipitation reaction is pH 5.5 to 7.5. Carried out in, and the calcium phosphate precipitation reaction time may be 3 to 5 hours.
본 발명의 다른 일 구체예에 따르면, 상기 칼슘인산염 침전 반응에 첨가된 칼슘 이온의 농도는 1 내지 20 mM이고, 인산 이온의 농도는 1 내지 20 mM이고, 상기 칼슘인산염 침전 반응은 pH 5.5 내지 7.5에서 수행하고, 상기 칼슘인산염 침전 반응 시간은 3 내지 5 시간이고, 상기 칼슘인산염 침전 반응시 단백질 함유 시료의 농도를 1배 내지 3배 희석할 수 있다. According to another embodiment of the present invention, the concentration of calcium ions added to the calcium phosphate precipitation reaction is 1 to 20 mM, the concentration of phosphate ions is 1 to 20 mM, and the calcium phosphate precipitation reaction is pH 5.5 to 7.5. The calcium phosphate precipitation reaction time is carried out at 3 to 5 hours, and the calcium phosphate precipitation reaction may be diluted 1 to 3 times the concentration of the protein-containing sample.
본 발명의 일 구체예에서, 상기 불순물은 단백질 다량체, 단백질 분절체, DNA 오염물, 바이러스, 숙주세포 유래 단백질 또는 단백질 A를 포함하지만, 이에 한정되는 것은 아니다. In one embodiment of the invention, the impurities include, but are not limited to, protein multimers, protein fragments, DNA contaminants, viruses, host cell derived proteins or protein A.
본 발명의 일 구체예에서, 불순물을 제거하고자 하는 단백질 함유 시료 중에 포함된 단백질은 생리 활성 단백질일 수 있으며, 일 예로, 상기 단백질은 항체 또는 융합 단백질일 수 있으나, 이에 한정되는 것은 아니다. 예컨대, 상기 항체는 단일클론항체일 수 있으며, 일 예로, 상기 항체는 항-CD20 항체 또는 항-ErbB3 항체일 수 있다. 또한, 상기 융합 단백질은 Fc 융합 단백질일 수 있으며, 일 예로, 상기 Fc 융합 단백질은 TNFα 수용체 단편과 항체 Fc 단편의 융합 단백질일 수 있다.In one embodiment of the present invention, the protein contained in the protein-containing sample to remove impurities may be a physiologically active protein, for example, the protein may be an antibody or a fusion protein, but is not limited thereto. For example, the antibody may be a monoclonal antibody, for example, the antibody may be an anti-CD20 antibody or an anti-ErbB3 antibody. In addition, the fusion protein may be an Fc fusion protein. For example, the Fc fusion protein may be a fusion protein of a TNFα receptor fragment and an antibody Fc fragment.
보다 구체적으로, 본 발명의 일 구체예에 따른 방법으로 정제하는 단백질 함유 시료에 함유되는 단백질은 예를 들어, 단일클론항체, Fc 융합 단백질, 과립구 콜로니 자극 인자 (G-CSF), 과립구 대식세포 콜로니 자극 인자 (GM-CSF), 에리트로포에틴 (EPO), 트롬보포에틴 등의 조혈 인자, 인터페론, IL-1, IL-6 등의 사이토카인, 혈청 알부민, 혈액 응고 제 VIII 인자, 혈액 응고 제 IX 인자, 인슐린, 줄기 세포 성장 인자 (SCF) 등을 포함하지만, 이들에 한정되지 않는다. 단백질 A 흡착 크로마토그래피를 이용하여 실시하는 본 발명의 일 구체예에서 불순물을 제거할 단백질은 단일클론 항체 또는 Fc 융합단백질(예, TNFRc-Fc 융합단백질)일 수 있다. 본 발명의 일 구체예에서, 정제 가능한 항체는 IgG, IgA, IgE, IgD, 또는 IgM를 모두 포함한다. More specifically, the protein contained in the protein-containing sample to be purified by the method according to one embodiment of the present invention, for example, monoclonal antibody, Fc fusion protein, granulocyte colony stimulating factor (G-CSF), granulocyte macrophage colony Hematopoietic factors such as stimulating factor (GM-CSF), erythropoietin (EPO), thrombopoietin, cytokines such as interferon, IL-1, IL-6, serum albumin, blood coagulant VIII factor, blood coagulant IX Factors include, but are not limited to, insulin, stem cell growth factor (SCF), and the like. In one embodiment of the invention carried out using Protein A adsorption chromatography, the protein to remove impurities may be a monoclonal antibody or an Fc fusion protein (eg, a TNFRc-Fc fusion protein). In one embodiment of the invention, the purifiable antibody comprises all IgG, IgA, IgE, IgD, or IgM.
본 명세서에서 사용되는 용어 "생리 활성 단백질" 이란, 포유 동물, 예를 들어 인간의 생리 활성 단백질과 실질적으로 같은 생물학적 활성을 갖는 것이고, 천연의 것 또는 유전자 재조합에 의해 얻어진 것을 포함한다. 유전자 재조합에 의한 생리 활성 단백질은, 대장균 등의 박테리아 세포; 중국 햄스터 난소 (CHO) 세포, BHK 세포, COS 세포, 인간 유래의 세포 등의 동물 유래의 배양 세포에서 제조할 수 있으며, 사용 전에 이를 여러 가지 방법으로 분리 및 정제할 수 있다. 유전자 재조합에 의해서 얻어지는 단백질에는 천연 단백질과 아미노산 서열이 같은 것, 및 상기 아미노산 서열에서 하나 이상의 아미노산을 결실, 치환, 부가한 것으로서 상기 생물학적 활성을 보유하는 것을 포함한다. 또한, 생리 활성 단백질은 PEG 등에 의해 화학적으로 개질된 것도 포함한다.As used herein, the term "physiologically active protein" refers to a substance having a biological activity substantially the same as that of a physiologically active protein of a mammal, for example, a human being, or one obtained by natural or genetic recombination. Bioactive proteins by genetic recombination include bacterial cells such as E. coli; Chinese hamster ovary (CHO) cells, BHK cells, COS cells, cells derived from animals, such as human-derived cells can be prepared, and can be isolated and purified by various methods before use. Proteins obtained by genetic recombination include those having the same amino acid sequence as the native protein and those that retain the biological activity as deletions, substitutions, and additions of one or more amino acids in the amino acid sequence. In addition, physiologically active proteins include those chemically modified by PEG and the like.
생리 활성 단백질이 단일클론 항체인 경우에는 단일클론 항체는 어떠한 방법으로 제조된 것이어도 된다. 단일클론 항체는 기본적으로는 공지 기술을 사용하여, 감작 항원을 통상의 면역 방법에 따라서 면역하고, 얻어지는 면역 세포를 통상의 세포 융합법에 의해서 공지된 부모세포와 융합시켜, 통상의 스크리닝법에 의해, 단일클론인 항체 생성 세포를 스크리닝함으로써 제작할 수 있다.When the bioactive protein is a monoclonal antibody, the monoclonal antibody may be produced by any method. The monoclonal antibody basically immunizes the sensitizing antigen according to a conventional immunization method using a known technique, fuses the obtained immune cells with known parental cells by a conventional cell fusion method, and then performs a conventional screening method. It can produce by screening antibody producing cells which are monoclonal.
다른 방법으로는, 항체 유전자를 하이브리도마로부터 클로닝하여 적당한 벡터에 삽입하고, 유전자 재조합 기술을 사용하여 이것을 숙주에 도입하며, 이렇게 생성시킨 유전자 재조합 항체도 본 발명의 일 구체예에서 사용할 수 있다. 구체적으로는 하이브리도마의 mRNA 로부터 역전사 효소를 사용하여 항체의 가변 영역 (V 영역) 의 cDNA를 합성한다. 목적하는 항체의 V 영역을 암호화하는 DNA 가 얻어지면 이것을 원하는 항체 불변 영역 (C 영역)을 암호화하는 DNA 와 연결하고, 이것을 발현 벡터에 삽입한다. 또는 항체의 V 영역을 암호화하는 DNA 를, 항체 C 영역의 DNA 를 포함하는 발현 벡터에 삽입해도 된다. 발현 제어 영역, 예를 들어, 인핸서(enhancer) 또는 프로모터의 제어 하에서 발현되도록 DNA 구성체를 발현 벡터에 삽입한다. 다음에, 이 발현 벡터에 의해 숙주 세포를 형질 전환하여 항체를 발현시킬 수 있다.Alternatively, the antibody genes can be cloned from the hybridomas and inserted into a suitable vector and introduced into the host using gene recombination techniques, and the resulting recombinant antibodies can also be used in one embodiment of the present invention. Specifically, cDNA of the variable region (V region) of an antibody is synthesize | combined from the mRNA of a hybridoma using reverse transcriptase. When DNA encoding the V region of the antibody of interest is obtained, the DNA is linked to the DNA encoding the desired antibody constant region (C region) and inserted into the expression vector. Alternatively, the DNA encoding the V region of the antibody may be inserted into an expression vector containing the DNA of the antibody C region. The DNA construct is inserted into the expression vector so that it is expressed under the control of an expression control region, eg, an enhancer or a promoter. Next, the host cell can be transformed with this expression vector to express the antibody.
본 발명의 일 예에서는 인간에 대한 이종 항원성을 저하시키는 것을 목적으로 하여 인위적으로 개질한 유전자 재조합형 항체, 예를 들어 키메라 (Chimeric) 항체, 인간화 (Humanized) 항체 등을 사용할 수 있다. 이들 개질 항체는 공지의 방법을 이용하여 제조할 수 있다. In one embodiment of the present invention, artificially modified genetic recombinant antibodies, such as chimeric antibodies, humanized antibodies, etc., may be used for the purpose of lowering heterologous antigenicity to humans. These modified antibodies can be produced using a known method.
키메라 항체는 인간 이외의 포유 동물, 예를 들어 마우스 항체의 중쇄, 경쇄의 가변 영역과 인간 항체의 중쇄, 경쇄의 불변 영역으로 이루어지는 항체이고, 마우스 항체의 가변 영역을 암호화하는 DNA 를 인간 항체의 불변 영역을 암호화하는 DNA 와 연결하고, 이것을 발현 벡터에 삽입하여 숙주에 도입하여 항체를 생성시킴으로써 얻을 수 있다.A chimeric antibody is an antibody consisting of a variable region of a heavy chain and a light chain of a mammal other than human, for example, a mouse antibody, and a constant region of a heavy chain and a light chain of a human antibody, and the DNA encoding the variable region of a mouse antibody is invariant of a human antibody. It can be obtained by linking with a DNA encoding a region, inserting it into an expression vector, and introducing into a host to generate an antibody.
인간화 항체는 인간 이외의 포유 동물, 예를 들어 마우스 항체의 상보성 결정 영역 (CDR; complementarity determining region) 을 인간 항체의 상보성 결정 영역에 이식하여 수득된다. 이의 표준 유전자 재조합 절차도 알려져 있다. 구체적으로는 마우스 항체의 CDR 과 인간 항체의 프레임워크 영역 (framework region; FR) 을 연결하도록 설계한 DNA 서열을, 말단부에 오버랩하는 부분을 갖도록 제작한 수 개의 올리고뉴클레오티드로부터 PCR 법에 의해 합성한다. 이렇게 얻어진 DNA를 인간의 항체 불변 영역을 암호화하는 DNA 와 연결하고, 이어서 발현 벡터에 삽입하고, 이것을 숙주에 도입하여 항체를 생성시킨다. CDR을 통해 연결되는 인간 항체의 FR 은 상보성 결정 영역이 양호한 항원 결합 부위를 형성하도록 선택된다. 필요에 따라, 재구성 인간화 항체의 상보성 결정 영역이 적절한 항원 결합 부위를 형성하도록 항체의 가변 영역의 프레임워크 영역의 아미노산을 치환할 수 있다.Humanized antibodies are obtained by implanting a complementarity determining region (CDR) of a mammal other than a human, eg, a mouse antibody, into the complementarity determining region of a human antibody. Its standard genetic recombination procedures are also known. Specifically, a DNA sequence designed to connect the CDRs of a mouse antibody and a framework region (FR) of a human antibody is synthesized from several oligonucleotides prepared to have a portion overlapping with a terminal portion by the PCR method. The DNA thus obtained is linked with the DNA encoding the human antibody constant region, and then inserted into an expression vector, which is introduced into a host to generate an antibody. The FRs of human antibodies linked via CDRs are chosen such that the complementarity determining regions form good antigen binding sites. If desired, amino acids of the framework regions of the variable regions of the antibody can be substituted such that the complementarity determining regions of the reconstituted humanized antibody form the appropriate antigen binding site.
또한, 인간 항체의 취득 방법도 알려져 있다. 예를 들어, 인간 림프구를 in vitro에서 원하는 항원 또는 원하는 항원을 발현하는 세포로 감작하고, 감작 림프구를 인간 골수종 세포, 예를 들어 U266 과 융합시켜, 항원에 대한 결합 활성을 갖는 원하는 인간 항체를 얻을 수 있다. 또한, 인간 항체 유전자의 모든 레퍼토리를 갖는 트랜스제닉 동물을 항원으로 면역함으로써 원하는 인간 항체를 취득할 수 있다. 또한, 인간 항체 라이브러리를 사용하여, 패닝(panning)에 의해 인간 항체를 취득하는 기술도 알려져 있다. 예를 들어, 인간 항체의 가변 영역을 단일쇄 항체 (scFv)로서 파지 디스플레이법에 의해 파지의 표면에 발현시키고, 항원에 결합하는 파지를 선택할 수 있다. 선택된 파지의 유전자를 해석하면, 항원에 결합하는 인간 항체의 가변 영역을 암호화하는 DNA 서열을 결정할 수 있다. 항원에 결합하는 scFv 의 DNA 서열이 밝혀지면, 당해 서열에 기초하여 적당한 발현 벡터를 제작하여 인간 항체를 취득할 수 있다. 이러한 방법들은 이미 해당 기술 분야에 널리 알려져 있다. Moreover, the acquisition method of a human antibody is also known. For example, human lymphocytes are sensitized in vitro with cells expressing a desired antigen or a desired antigen, and sensitized lymphocytes are fused with human myeloma cells, eg, U266, to obtain a desired human antibody having binding activity to the antigen. Can be. In addition, a desired human antibody can be obtained by immunizing a transgenic animal having all repertoires of the human antibody gene with an antigen. In addition, techniques for obtaining human antibodies by panning using human antibody libraries are also known. For example, a phage that expresses the variable region of a human antibody on the surface of the phage by the phage display method as a single-chain antibody (scFv) can be selected. Interpreting the gene of the selected phage can determine the DNA sequence encoding the variable region of the human antibody that binds the antigen. Once the DNA sequence of the scFv that binds to the antigen is known, a suitable expression vector can be prepared based on the sequence to obtain a human antibody. Such methods are already well known in the art.
또한, 본 발명의 일 예에서 사용할 수 있는 항체에는 Fab, (Fab')2, Fc, Fc', Fd 등의 항체 단편 및 1가 또는 2가의 단일쇄 항체 (scFv) 등의 재구성 항체도 포함된다.In addition, antibodies that can be used in one embodiment of the present invention also include antibody fragments such as Fab, (Fab ') 2 , Fc, Fc', Fd, and reconstituted antibodies such as monovalent or bivalent single-chain antibodies (scFv). .
본 명세서에서 사용되는 용어 "생리 활성 단백질 함유 시료" 또는 "단백질 함유 시료" 또는 "항체 함유 시료"란 배양에 의해 얻어진 생리 활성 단백질 분자 또는 단백질 또는 항체 분자를 포함하는 CHO 세포 등의 포유 동물 세포의 배양 배지, 또는 이것에 부분적 정제 등의 일정한 처리를 실시한 것을 말한다.As used herein, the term "physiologically active protein-containing sample" or "protein-containing sample" or "antibody-containing sample" means a mammalian cell such as a CHO cell containing a bioactive protein molecule or a protein or antibody molecule obtained by culturing. The culture medium or the thing which gave constant treatment, such as partial purification, to this.
본 명세서에서 사용되는 용어 "불순물"은 목적하는 단백질 이외의 물질이면 어떠한 물질이어도 포함되며, 불순물의 예로서는 단백질 다량체(aggregate), 단백질 분절체(fragment), DNA 오염물, 바이러스, 단백질 A (칼럼으로부터 용출됨), 숙주세포 유래 단백질(Host cell protein), 내독소, 배지 성분인 Hy-Fish (FL), IGF 등을 들 수 있지만, 이것에 한정되는 것은 아니다. As used herein, the term “impurity” includes any substance as long as it is a substance other than the protein of interest, and examples of impurities include protein aggregates, protein fragments, DNA contaminants, viruses, protein A (from columns). Eluted), host cell protein, endotoxin, Hy-Fish (FL) as a medium component, IGF, and the like, but are not limited thereto.
본 명세서에서 불순물의 예로서 사용되는 용어 "단백질 다량체(aggregate)"란 해당 단백질 두 개 이상이 서로 결합되어 있는 것을 말한다. 여기서 결합은 비가역적인 결합으로 결합된 단백질은 각각으로 분해되지 않는다. 단백질 다량체는 인체내에서 해당 단백질에 대한 중화항체를 생성시키게 해여 해당 단백질에 대한 효능을 사라지게 할 수 있다. 따라서, 정제과정에서 제거해야 할 불순물이다.As used herein as an example of impurities, the term "protein aggregate" refers to the binding of two or more proteins of interest. Here, the binding is irreversible binding, the bound protein is not broken down into each. Protein multimers can produce neutralizing antibodies to the protein in the human body, thereby disappearing the efficacy for that protein. Therefore, it is an impurity to be removed in the purification process.
본 명세서에서 불순물의 예로서 사용되는 용어 "단백질 분절체(fragment)"란 해당 단백질 합성 과정에서 잘못 합성되어 온전한 크기보다 작거나, 해당 단백질의 단백질 분해효소들에 의해 분해된, 해당 단백질의 조각 단백질을 의미한다. 이들은 효능이 없기 때문에 불순물로 간주된다. As used herein as an example of an impurity, the term “protein fragment” refers to a fragment protein of a protein that is poorly synthesized and degraded by proteolytic enzymes of that protein, incorrectly synthesized during the protein synthesis process. Means. They are considered impurities because they are ineffective.
본 명세서에서 불순물의 예로서 사용되는 용어 "DNA 오염물" 이란, 생리 활성 단백질 함유 시료 중의 DNA를 의미하고, 숙주 유래의 DNA 및 오염된 바이러스 유래의 DNA 가 포함된다.As used herein as an example of impurities, the term "DNA contaminant" means DNA in a biologically active protein-containing sample, and includes DNA from a host and DNA from a contaminated virus.
본 명세서에서 불순물의 예로서 사용되는 용어 "DNA 오염물"에는 특별히 제한은 없고, DNA 바이러스 및 RNA 바이러스가 포함된다. RNA 바이러스로서는 X-MuLV 와 같은 레트로 바이러스, Reo3 과 같은 레오 바이러스 및 MVM과 같은 파보 바이러스(parvovirus)를 들 수 있다. 본 발명의 방법에 의해서 제거되는 바이러스의 구체적인 예로서는 예를 들어, X-MuLV, PRV, Reo3, MVM, VSV, 헤르페스 심플렉스, CHV, Sindbis, 유행성 이하선염, 우두, Measle, Rubella, 인플루엔자, 헤르페스 조스터, 사이토메갈로, 파라인플루엔자, EB, HIV, HA, HB, NANB, ATL, ECHO 및 파보 바이러스 등을 들 수 있다. The term "DNA contaminant" used herein as an example of impurities is not particularly limited and includes DNA viruses and RNA viruses. RNA viruses include retroviruses such as X-MuLV, Leo viruses such as Reo3, and parvoviruses such as MVM. Specific examples of viruses removed by the method of the present invention include, for example, X-MuLV, PRV, Reo3, MVM, VSV, herpes simplex, CHV, Sindbis, mumps, vaccinia, Measle, Rubella, influenza, herpes zoster , Cytomegalo, parainfluenza, EB, HIV, HA, HB, NANB, ATL, ECHO, and parvovirus.
본 명세서에서 불순물의 예로서 사용되는 용어 "단백질 A"란 단백질 A 크로마토그래피 수지에서 침출된 단백질 A(leached protein A)이며, 이것은 인체 내에서 발열반응을 일으킬 수 있는 불순물이다. As used herein as an example of an impurity, the term "protein A" is a protein A (leached protein A) leached from the Protein A chromatography resin, which is an impurity that can cause an exothermic reaction in the human body.
본 발명자들은 본 발명의 칼슘인산염 침전 방법에 따른 불순물 제거 효과에 영향을 주는 인자들을 규명하기 위해 통계적인 실험법 (Design of Experiment, DOE)을 활용하였다. DoE는 통계적인 방법을 통하여 "최소의 실험"으로 "최대의 정보"를 얻기 위하여 실험계획을 수립하는 것을 의미한다. 일반적으로 DoE의 목적은 통제 가능한 인자(요인)을 조직하여 중요 인자를 발굴하고 최적조건을 찾는 것이다. 본 발명자들은 통계프로그램(Design expert software. ver. 7.1.1)을 이용하여 통계적인 실험법(DoE)을 계획하고 통계적인 분석(Analysis of variance, ANOVA, 변량분석)을 진행하였다. 본 발명의 일 실시예에서 사용된 변량분석(Analysis of variance, ANOVA)은 통계학에서 두 개 이상 다수의 집단을 비교하고자 할 때 집단 내의 분산, 총 평균과 각 집단의 평균의 차이에 의해 생긴 집단 간 분산의 비교를 통해 만들어진 F 분포(여기서, F 분포는 분산의 비교를 통해 얻어진 분포비율이다. 이 비율을 이용하여 각 집단의 모집단 분산이 차이가 있는지에 대한 검정과 모집단평균이 차이가 있는지 검정하는 방법으로 사용한다)를 이용하여 가설검정을 하는 방법이다. 통계학자이자 유전학자인 로날드 피셔(R.A. Fisher)에 의해 1920년대에서 1930년대에 걸쳐 만들어졌다. 상기 통계분석을 위해 사용된 통계프로그램(Design expert software. ver. 7.1.1)은 제약분야에서 의약품을 개발하고 생산하는데 많이 활용되고 있다. The present inventors used a statistical experiment (Design of Experiment, DOE) to identify the factors affecting the impurities removal effect according to the calcium phosphate precipitation method of the present invention. DoE means developing an experimental plan to obtain "maximum information" with "minimum experiment" through statistical methods. In general, the purpose of DoE is to organize factors that can be controlled to identify important factors and to find optimal conditions. The inventors planned a statistical experiment (DoE) using a statistical program (Design expert software. Ver. 7.1.1) and performed a statistical analysis (Analysis of variance, ANOVA, variance analysis). The analysis of variance (ANOVA) used in one embodiment of the present invention is used to compare two or more large groups in statistics between groups resulting from the variance in the group, the difference between the total mean and the mean of each group. The F distribution produced by comparing the variances, where the F distribution is the distribution ratio obtained by comparing the variances. This ratio is used to test whether there is a difference in the population variance of each group and whether the population mean is different. Hypothesis test using the Created by statistician and geneticist Ronald Fisher (R.A. Fisher) from the 1920s to the 1930s. The statistical program (Design expert software. Ver. 7.1.1) used for the statistical analysis is widely used for the development and production of pharmaceuticals in the pharmaceutical field.
본 발명을 이용하여 단백질을 정제하는 경우, 단백질 A 크로마토그래피(Protein A Chromatography) 이후 정제 공정에서 긴 시간과 설비와 방법이 복잡한 크로마토그래피 대신에, 단백질 다량체와 같은 불순물을 효과적으로 짧은 시간과 단순한 방법으로 제거할 수 있다. In the case of purifying proteins using the present invention, in the purification process after Protein A Chromatography, instead of complicated chromatography, a long time and a simple method of effectively removing impurities such as protein multimers Can be removed with
도 1은 일반적인 항체 정제 단계를 도시한 것이다. 도 1의 항체 정제 단계에서 단백질 A 크로마토그래피 단계 이후 추가적인 1st 또는 2nd 크로마토그래피 단계 대신에 본 발명의 일 예에 따른 칼슘인산염 침전 방법을 적용할 수 있다. 1 illustrates a general antibody purification step. In the antibody purification step of FIG. 1, a calcium phosphate precipitation method according to an embodiment of the present invention may be applied instead of an additional 1 st or 2 nd chromatography step after the Protein A chromatography step.
도 2는 본 발명의 일 예에 따른 칼슘인산염 침전을 통한 정제를 진행하기 위한 정제준비 단계를 나타낸 것이다. Figure 2 shows a purification preparation step for proceeding purification through calcium phosphate precipitation according to an embodiment of the present invention.
도 3a 및 3b는 TNFα 수용체-Fc 융합단백질에 대하여 본 발명의 일 예에 따른 칼슘인산염 침전 반응을 통해 추가 정제를 진행하고, 정제된 항-ErbB3 항체를 분석용 크기배제 크로마도그래피(SEC-HPLC)를 통해 항체 다량체를 분석한 결과이다: Figure 3a and 3b is further purified through calcium phosphate precipitation reaction according to an embodiment of the present invention for the TNFα receptor-Fc fusion protein, the purified anti-ErbB3 antibody for analysis size exclusion chromatography (SEC-HPLC Results of analyzing antibody multimers
- 도 3a: 칼슘인산염 침전 반응 전Figure 3a: before calcium phosphate precipitation reaction
- 도 3b: 칼슘인산염 침전 반응 후3b: after calcium phosphate precipitation reaction
도 4a 및 4b는 항-ErbB3 항체에 대하여 본 발명의 일 예에 따른 칼슘인산염 침전 반응을 통해 추가 정제를 진행하고, 정제된 TNFα 수용체-Fc 융합단백질을 분석용 크기배제 크로마도그래피(SEC-HPLC)를 통해 단백질 다량체를 분석한 결과이다: Figures 4a and 4b is further purified through the calcium phosphate precipitation reaction according to an embodiment of the present invention for the anti-ErbB3 antibody, the purified TNFα receptor-Fc fusion protein for analysis size exclusion chromatography (SEC-HPLC This is the result of analyzing the protein multimer through
- 도 4a: 칼슘인산염 침전 반응 전Figure 4a: before calcium phosphate precipitation reaction
- 도 4b: 칼슘인산염 침전 반응 후4b: after calcium phosphate precipitation reaction
이하, 실시 예에 의해 본 발명을 보다 구체적으로 설명하고자 한다. 이들 실시 예는 오직 본 발명을 설명하기 위한 것으로, 본 발명의 범위가 이들 실시 예에 국한되는 것은 아니다. Hereinafter, the present invention will be described in more detail with reference to Examples. These examples are only for illustrating the present invention, but the scope of the present invention is not limited to these examples.
실시예Example
실시예 1: 칼슘인산염 침전을 통한 정제를 진행하기 위한 중간 정제물질 준비Example 1 Preparation of Intermediate Purified Material for Purification by Calcium Phosphate Precipitation
1-1. 항-CD20 항체의 중간 정제물질 준비1-1. Intermediate Purification of Anti-CD20 Antibodies
항-CD20 항체를 포함하는 CHO (Chines Hamster Ovary Cell) 세포 배양액을 단백질 A 크로마토그래피(GE Healthcare사의 MabSelect SuRe Protein A chromatography)를 통해 정제를 진행했다 (도 2 (A) 참조). 단백질 A 컬럼으로부터 50 mM 아세테이트, pH 3.5 완충액을 이용해서 항-CD20 항체를 용출하고, 용출액을 1M 아세트산을 이용해서 pH 3.5로 조절하고 1 시간 동안 실온에서 보관하였다. 그 후, pH 3.5 용출액을 다시 트리스염기로 pH 8.0이 되게 만든 후, 음이온 크로마토그래피(Q sepharose Fast Flow; GE Healthcare사 제품)를 통해 추가 정제를 진행했다. 이때 항체가 포함된 pH 8.0 용액은 음이온에 결합하지 않고 흘러서 나온다. 즉, 통과 흐름(flow through) 방식에 의해 정제를 진행했다. 결합하지 않고 나온 항체를 포함하는 pH 8.0 용액을 1M 아세트산을 이용해서 pH 7.0이 되게 조절하고, 이 중간 정제 단계의 물질을 본 발명의 칼슘인산염 침전 방법을 이용해서 실시예 2와 같이 추가 정제를 진행했다. The CHO (Chines Hamster Ovary Cell) cell culture containing an anti-CD20 antibody was purified by protein A chromatography (MabSelect SuRe Protein A chromatography, GE Healthcare) (see FIG. 2 (A)). The anti-CD20 antibody was eluted from the Protein A column with 50 mM acetate, pH 3.5 buffer, and the eluate was adjusted to pH 3.5 with 1M acetic acid and stored at room temperature for 1 hour. Thereafter, the pH 3.5 eluate was brought back to pH 8.0 with a trisbase, and further purified by anion chromatography (Q sepharose Fast Flow; manufactured by GE Healthcare). The pH 8.0 solution containing the antibody flows out without binding to the anion. That is, purification was performed by a flow through method. The pH 8.0 solution containing the antibody released without binding was adjusted to pH 7.0 using 1 M acetic acid, and the intermediate purification step was further purified as in Example 2 using the calcium phosphate precipitation method of the present invention. did.
1-2. TNFα 수용체-Fc 융합 단백질의 중간 정제물질 준비1-2. Intermediate Purification of TNFα Receptor-Fc Fusion Proteins
TNFα 수용체와 Fc 융합 단백질(이하, TNFRc-Fc 융합 단백질이라 함)을 포함하는 동물세포 배양액을 단백질 A 크로마토그래피를 통해 정제를 진행했다 (도 2 (B) 참조). 단백질 A 컬럼으로부터 50 mM 아세테이트, pH 3.5 완충액을 이용해서 TNFRc-Fc 융합 단백질을 용출하고, 이 용출액을 트리스염기를 이용해서 pH 7.0이 되게 만든 후, 이 중간 정제 단계의 물질을 본 발명의 칼슘인산 침전 방법을 이용해서 실시예 2와 같이 추가 정제를 진행했다.Animal cell cultures containing TNFα receptors and Fc fusion proteins (hereinafter referred to as TNFRc-Fc fusion proteins) were purified by protein A chromatography (see FIG. 2 (B)). TNFRc-Fc fusion protein was eluted from the Protein A column using 50 mM acetate, pH 3.5 buffer, and this eluate was brought to pH 7.0 using trisbase. Further purification was carried out as in Example 2 using the precipitation method.
실시예 2: 불순물 제거를 위한 칼슘인산염 침전 조건 확인 시험Example 2: Calcium Phosphate Precipitation Conditions Confirmation Test for Impurity Removal
동물세포를 이용하여 생산한 항-CD20 항체 또는 TNFRc-Fc 융합 단백질을 포함하는 각각의 배양액을 실시예 1과 같이 정제를 통해 얻은 중간 정제용액을 pH 7.0로 조절 후, 항체 또는 단백질 다량체와 같은 불순물들을 제거하는 인자들을 찾기 위해 통계프로그램(Stat-Ease, Inc. 사의 Design expert software. ver. 7.1.1)을 이용하여 아래 표 1과 같이 실험을 계획하여 시험을 진행하였다. After adjusting the intermediate purified solution obtained by purification as described in Example 1, each culture solution containing an anti-CD20 antibody or TNFRc-Fc fusion protein produced using animal cells, such as antibody or protein multimer In order to find the factors to remove impurities, the experiment was conducted by planning the experiment as shown in Table 1 using a statistical program (Design expert software ver. 7.1.1 of Stat-Ease, Inc.).
표 1
1)모든 인자들의 중앙값에 대해서는 통계적인 분석을 위해 3회를 진행했다. 1) The median of all factors was performed three times for statistical analysis.
실시예 3: 칼슘인산염 침전 조건에 따른 정제 효과 평가Example 3: Evaluation of Purification Effect According to Calcium Phosphate Precipitation Conditions
표 1과 같이 시험을 진행한 후, 칼슘인산염 침전 방법에 의한 1차적인 평가를 위해 항체 수율과 항체 단량체, 항체 다량체, 항체 절편 비율을 측정하였다.After the test as shown in Table 1, the antibody yield, antibody monomer, antibody multimer, and antibody fragment ratio were measured for the primary evaluation by the calcium phosphate precipitation method.
항체 수율의 경우, 항체농도를 UV280에서 흡광도를 측정하여 아래 수식 1에 대입하여 계산하였다. In the case of antibody yield, the antibody concentration was calculated by substituting the
수학식 1
항체 단량체 비율(Antibody monomer %)은 분석용 크기배제 크로마토그래피(Tosoh 사의 TSKgel G3000SWL Size Exclusion Chromatography Column)를 통해서 분자량이 큰 항체 다량체 (High Molecular Weight), 항체 단량체(Monomer)와 분자량이 작은 항체 분절(Low Molecular Weight)로 분리하였다. Antibody monomer ratio was determined by analytical size exclusion chromatography (Tosoh's TSKgel G3000SWL Size Exclusion Chromatography Column). High Molecular Weight, Antibody Monomer and Low Molecular Weight (Low Molecular Weight).
칼슘인산염 침전을 이용한 정제 방법 대한 1차적 평가를 통해 아래 정리된 표 2와 표 3과 같은 결과를 얻었다. Through the first evaluation of the purification method using calcium phosphate precipitation, the results as shown in Table 2 and Table 3 were obtained.
표 2
1) 희석 전 항-CD20 항체 농도는 4.8906 mg/mL 이었다. 1) The anti-CD20 antibody concentration before dilution was 4.8906 mg / mL.
2) 초기 항-CD20 항체에 대한 다량체 비율은 1.19% 이었다. 2) The multimer ratio to the initial anti-CD20 antibody was 1.19%.
3)초기 항-CD20 항체에 대한 단량체 비율은 98.81% 이었다. 3) The monomer ratio to the initial anti-CD20 antibody was 98.81%.
4) 초기 항-CD20 항체에 대한 분절체 비율은 0.00% 이었다. 4) The segment ratio for the initial anti-CD20 antibody was 0.00%.
5)모든 인자들의 중앙값에 대해서는 통계적인 분석을 위해 3회를 진행했다. 5) The median of all factors was performed three times for statistical analysis.
표 3
1) 희석 전 TNFRc-Fc 융합 단백질 농도는 4.70 mg/mL 이었다. 1) TNFRc-Fc fusion protein concentration before dilution was 4.70 mg / mL.
2) 초기 TNFRc-Fc 융합 단백질 에 대한 다량체 비율은 1.59% 이었다. 2) The multimer ratio to the initial TNFRc-Fc fusion protein was 1.59%.
3)초기 TNFRc-Fc 융합 단백질에 대한 단량체 비율은 97.71% 이었다. 3) The monomer ratio to the initial TNFRc-Fc fusion protein was 97.71%.
4) 초기 TNFRc-Fc 융합 단백질 에 대한 분절체 비율은 0.70% 이었다. 4) The segment ratio for the initial TNFRc-Fc fusion protein was 0.70%.
5)모든 인자들의 중앙값에 대해서는 통계적인 분석을 위해 3회를 진행했다. 5) The median of all factors was performed three times for statistical analysis.
실시예 4: 칼슘인산염 침전 조건에 대한 통계적 분석Example 4 Statistical Analysis of Calcium Phosphate Precipitation Conditions
4-1. 항-CD20 항체4-1. Anti-CD20 antibody
표 2의 결과를 가지고 항-CD20 항체의 정제를 위한 인자들을 찾기 위해 통계프로그램(Design expert software. ver. 7.1.1)을 이용하여 통계적인 분석(Analysis of variance, ANOVA, 변량분석)을 진행하였다. 표 2에서 보는 것과 마찬가지로 원심분리와 0.2㎛ 여과를 진행한 샘플에 대한 수율과 분석용 크기 배제 크로마토그래의 결과에는 큰 차이가 없었기 때문에, 통계적인 분석은 0.2㎛ 여과를 진행한 샘플에 대한 결과만 가지고 진행하였다. Using the results of Table 2, statistical analysis (Analysis of variance, ANOVA, variance analysis) was performed using a statistical program (Design expert software. Ver. 7.1.1) to find factors for purification of anti-CD20 antibodies. . As shown in Table 2, there was no significant difference between the yield of the sample subjected to centrifugation and 0.2 μm filtration and the result of the analytical size exclusion chromatograph. Proceeded with.
그 결과 표 4에서 보는 것처럼 항-CD20 항체 수율의 경우, 첨가된 인산염 농도(P < 0.0051), CaCl2 농도 (P < 0.0033), 반응시간 (P < 0.0453) 그리고, 첨가된 인산염 농도와 CaCl2 농도의 상호관계 (P < 0.0042) 에 대해 P < 0.05로 통계적인 유의성이 있었다. If the result wherein -CD20 antibody yield as shown in Table 4, the addition of phosphate concentration (P <0.0051), CaCl 2 concentration (P <0.0033), and the reaction time (P <0.0453) and then the addition of the phosphate concentration and CaCl 2 There was a statistical significance of P <0.05 for the correlation of concentrations ( P <0.0042).
즉, 항-CD20 항체 수율의 경우, 첨가된 인산염 농도, 첨가된 CaCl2 농도, 반응 시간 그리고 첨가된 인산 이온 농도와 칼슘 이온 농도의 상호관계 의해 영향을 받는다. That is, in the case of anti-CD20 antibody yield, the phosphate concentration added, the added CaCl 2 concentration, the reaction time and the correlation between the added phosphate ion concentration and calcium ion concentration are affected.
표 4
그리고 표 5에서 보는 것처럼 항-CD20 항체에 대한 분석용 크기 배제 크로마토그래피(SEC) 결과를 통해, 첨가된 인산염 농도 (P < 0.0017), 첨가된 CaCl2 농도 (P < 0.0024), 반응시간 (P < 0.0082), 항-CD20 항체 농도(P < 0.0140), 첨가된 인산염 농도와 CaCl2 농도의 상호관계(P < 0.0006), 첨가된 인산염 농도와 반응시간의 상호관계(P < 0.0315), 첨가된 인산염 농도와 항-CD20 항체 농도의 상호관계(P < 0.0298), 첨가된 CaCl2 농도와 반응시간의 상호관계(P < 0.0355), 첨가된 CaCl2 농도와 항-CD20 항체농도의 상호관계(P < 0.0298), 반응시간과 항-CD20 항체농도와의 상호관계(P < 0.0141), 첨가된 인산염 농도와 첨가된 CaCl2 농도 그리고, 반응시간과의 상호관계(P < 0.0077), 첨가된 인산염 농도와 첨가된 CaCl2 농도 그리고, 항-CD20 항체농도와의 상호관계(P < 0.0079), 첨가된 인산염 농도와 반응시간 그리고, 항-CD20 항체농도와의 상호관계(P < 0.0117), 첨가된 CaCl2 농도와 반응시간 그리고, 항-CD20 항체농도와의 상호관계(P < 0.0117)그리고, 첨가된 인산염 농도와 첨가된 CaCl2 농도, 반응시간 그리고, 항-CD20 항체농도와의 상호관계(P < 0.0166)에 대해 P < 0.05로 통계적인 유의성이 있었다. And through analytical size exclusion chromatography (SEC) results for the anti-CD20 antibody, as shown in Table 5, added phosphate concentration (P <0.0017), added CaCl 2 concentration (P <0.0024), reaction time (P <0.0082), anti-CD20 antibody concentration ( P <0.0140), correlation between added phosphate concentration and CaCl 2 concentration ( P <0.0006), correlation between added phosphate concentration and reaction time ( P <0.0315), added Correlation between phosphate concentration and anti-CD20 antibody concentration ( P <0.0298), correlation of added CaCl 2 concentration and reaction time ( P <0.0355), correlation between added CaCl 2 concentration and anti-CD20 antibody concentration ( P <0.0298), the correlation between reaction time and anti-CD20 antibody concentration (P <0.0141), added phosphate concentration and added CaCl 2 concentration, Correlation between reaction time (P <0.0077), added phosphate concentration and added CaCl 2 concentration, Correlation with anti-CD20 antibody concentration (P <0.0079), added phosphate concentration and reaction time, correlation with anti-CD20 antibody concentration (P <0.0117), added CaCl 2 concentration and reaction time, and Wherein the mutual relationship between the -CD20 antibody concentration (P <0.0117) and then added with the addition of a phosphate concentration of CaCl 2 concentration, reaction time and, There was a statistical significance of P <0.05 for the correlation (P <0.0166) with the anti-CD20 antibody concentration.
즉, 항-CD20 항체 다량체의 제거에 영향을 주는 요인들은 첨가된 칼슘 이온 농도, 반응시간, 항-CD20 항체의 농도, 첨가된 인산이온 농도와 칼슘이온 농도의 상호관계, 첨가된 인산이온 농도와 반응시간의 상호관계, 첨가된 인산이온 농도와 항-CD20 항체 농도의 상호관계, 첨가된 칼슘이온 농도와 반응시간의 상호관계, 첨가된 칼슘이온 농도와 항-CD20 항체농도의 상호관계, 반응시간과 항-CD20 항체농도와의 상호관계, 첨가된 인산이온 농도와 첨가된 칼슘이온 농도 그리고 반응시간과의 상호관계, 첨가된 인산이온 농도와 첨가된 칼슘이온 농도 그리고 항-CD20 항체농도와의 상호관계, 첨가된 인산이온 농도와 반응시간 그리고 항-CD20 항체농도와의 상호관계, 첨가된 칼슘이온 농도와 반응시간 그리고 항-CD20 항체농도와의 상호관계, 그리고, 첨가된 인산이온 농도와 첨가된 칼슘이온 농도, 반응시간 그리고 항-CD20 항체농도와의 상호관계 항-CD20 항체 다량체 제거에 영향을 미친다. In other words, factors affecting the removal of the anti-CD20 antibody multimers are the added calcium ion concentration, reaction time, the concentration of anti-CD20 antibody, the correlation between the added phosphate and calcium ion concentrations, and the added phosphate ion concentration. Relationship between the reaction time and the reaction time, the correlation between the added phosphate concentration and the anti-CD20 antibody concentration, the correlation between the added calcium ion concentration and the reaction time, the correlation between the added calcium ion concentration and the anti-CD20 antibody concentration, and the reaction Correlation between time and anti-CD20 antibody concentration, added phosphate and calcium ion concentrations, and Correlation between reaction time , added phosphate and calcium ion concentrations, Correlation with anti-CD20 antibody concentration, added phosphate concentration and reaction time, and correlation with anti-CD20 antibody concentration, added calcium ion concentration and reaction time, and And wherein the antibody concentration -CD20 interaction, and then added with the addition of a phosphate ion concentration, calcium ion concentration, reaction time, and Correlation with Anti-CD20 Antibody Concentrations Affects anti-CD20 antibody multimer removal.
표 5
4-2. TNFRc-Fc 융합 단백질4-2. TNFRc-Fc fusion protein
표 3의 결과를 가지고 TNFRc-Fc 융합 단백질의 정제를 위한 인자들을 찾기 위해 통계프로그램(Design expert software. ver. 7.1.1)을 이용하여 통계적인 분석 (Analysis of variance, ANOVA, 변량분석)을 진행하였다. 표 3에서 보는 것과 마찬가지로 원심분리와 0.2㎛ 여과를 진행한 샘플에 대한 수율과 분석용 크기 배제 크로마토그래의 결과에는 큰 차이가 없었기 때문에, 통계적인 분석은 0.2㎛ 여과를 진행한 샘플에 대한 결과만 가지고 진행하였다.Using the results of Table 3, statistical analysis (Analysis of variance, ANOVA, variance analysis) was performed using a statistical program (Design expert software. Ver. 7.1.1) to find the factors for the purification of TNFRc-Fc fusion protein. It was. As shown in Table 3, there was no significant difference between the yield of the sample subjected to centrifugation and 0.2 μm filtration and the results of the analytical size exclusion chromatograph. Proceeded with.
그 결과, 표 6에서 보는 바와 같이 TNFRc-Fc 융합 단백질 수율의 경우, 첨간된 인산염의 농도(P < 0.0206) 첨가된 CaCl2 농도 (P < 0.0226), 그리고, 첨가된 인산염과 첨가된 CaCl2 농도와의 상호관계(P < 0.0327)에 대해 P < 0.05로 통계적인 유의성이 있었다. As a result, in the TNFRc-Fc fusion protein yield as shown in Table 6, the concentration of the added phosphate ( P <0.0206), the added CaCl 2 concentration ( P <0.0226), and the added phosphate and added CaCl 2 concentration There was a statistical significance of P <0.05 for the correlation with ( P <0.0327).
즉, TNFRc-Fc 융합 단백질 수율의 경우 첨가된 인산이온 농도, 첨가된 칼슘이온 농도, 그리고, 첨가된 임산이온 농도와 칼슘이온 농도의 상호관계에 의해 영향을 받는다. That is, the yield of TNFRc-Fc fusion protein is affected by the correlation between the added phosphate concentration, the added calcium ion concentration, and the added forest ion concentration and calcium ion concentration.
표 6
그리고, 표 7에서 보는 바와 같이 TNFRc-Fc 융합 단백질에 대한 분석용 크기 배제 크로마토그래피(SEC) 결과를 통해, 첨가된 인산염 농도(P < 0.0001), CaCl2 농도 (P < 0.0001), 반응시간, TNFRc-Fc 융합 단백질 농도(P < 0.0072), 첨가된 인산염 농도와 CaCl2 농도 상호관계 (P < 0.0007), 첨가된 인산염 농도와 반응시간의 상호관계(P < 0.0001), 첨가된 인산염 농도와 TNFRc-Fc 융합 단백질 농도와의 상호관계(P < 0.0011), 첨가된 CaCl2 농도와 반응시간과의 상호관계 (P < 0.0008), 첨가된 CaCl2 농도와 TNFRc-Fc 융합 단백질 농도와의 상호관계 (P < 0.0006), 반응시간과 TNFRc-Fc 융합 단백질 농도의 상호관계(P < 0.0013), 첨가된 인산염 농도와 CaCl2 농도 그리고 반응시간과의 상호관계 (P < 0.0099), 첨가된 인산염 농도와 반응시간과 그리고 TNFRc-Fc 융합 단백질 농도와의 상호관계 (P < 0.0011), 첨가된 인산염 농도와 CaCl2 농도 그리고 TNFRc-Fc 융합 단백질 농도와의 상호관계(P < 0.0019), 첨가된 CaCl2 농도와 반응시간 그리고 TNFRc-Fc 융합 단백질 농도와의 상호관계 (P < 0.0008), 첨가된 인산염 농도와 CaCl2 농도와 반응시간과 그리고 TNFRc-Fc 융합 단백질 농도와의 상호관계 (P < 0.0020)에 대해 P < 0.05로 통계적인 유의성이 있었다. And, as shown in Table 7 through the analytical size exclusion chromatography (SEC) results for the TNFRc-Fc fusion protein, added phosphate concentration ( P <0.0001), CaCl 2 concentration ( P <0.0001), reaction time, TNFRc-Fc fusion protein concentration ( P <0.0072), added phosphate concentration and CaCl 2 concentration correlation ( P <0.0007), added phosphate concentration and reaction time correlation ( P <0.0001), added phosphate concentration and TNFRc Correlation between -Fc fusion protein concentration ( P <0.0011), Correlation between added CaCl 2 concentration and reaction time ( P <0.0008), Correlation between added CaCl 2 concentration and TNFRc-Fc fusion protein concentration ( P <0.0006), correlation between reaction time and TNFRc-Fc fusion protein concentration ( P <0.0013), correlation between added phosphate concentration and CaCl 2 concentration and reaction time ( P <0.0099), reaction with added phosphate concentration mutual relationship between the time and the TNFRc-Fc fusion protein concentrations (P <0.0011), was added Phosphate concentration and CaCl 2 concentrations, and TNFRc-Fc interaction with the fusion protein concentration (P <0.0019), the addition of CaCl 2 concentration and the reaction time and the mutual relationship between the TNFRc-Fc fusion protein concentrations (P <0.0008), was added There was a statistically significant difference of P <0.05 for the correlation between phosphate concentration, CaCl 2 concentration, reaction time and TNFRc-Fc fusion protein concentration ( P <0.0020).
즉, TNFRc-Fc 융합 단백질 다량체의 제거에 영향을 주는 요인들은 첨가된 인산이온 농도, 첨가된 칼슘이온 농도, 반응시간, TNFRc-Fc 융합 단백질 의 농도, 첨가된 인산 이온 농도와 칼슘 이온 농도의 상호관계, 첨가된 인산 이온 농도와 반응시간의 상호관계, 첨가된 인산 이온 농도와 TNFRc-Fc 융합 단백질 농도와의 상호관계, 첨가된 칼슘 이온 농도와 반응시간과의 상호관계, 첨가된 칼슘 이온 농도와 TNFRc-Fc 융합 단백질 농도와의 상호관계, 반응시간과 TNFRc-Fc 융합 단백질 농도의 상호관계, 첨가된 인산 이온 농도와 칼슘 이온 농도 그리고 반응시간과의 상호관계, 첨가된 인산 이온 농도와 반응시간과 그리고 TNFRc-Fc 융합 단백질 농도와의 상호관계, 첨가된 인산 이온 농도와 반응시간 그리고 TNFRc-Fc 융합 단백질 농도, 첨가된 칼슘 이온 농도와 반응시간 그리고 TNFRc-Fc 융합 단백질 농도와의 상호관계, 첨가된 인산 이온 농도와 칼슘 이온 농도와 반응시간 그리고 TNFRc-Fc 융합 단백질 농도의 상호관계이다.In other words, the factors affecting the removal of TNFRc-Fc fusion protein multimers were the concentration of added phosphate concentration, added calcium ion concentration, reaction time, TNFRc-Fc fusion protein concentration, added phosphate ion concentration and calcium ion concentration. Correlation, correlation between added phosphate ion concentration and reaction time, correlation between added phosphate ion concentration and TNFRc-Fc fusion protein concentration, correlation between added calcium ion concentration and reaction time, added calcium ion concentration And the relationship between TNFRc-Fc fusion protein concentration, reaction time and TNFRc-Fc fusion protein concentration, added phosphate and calcium ion concentration and reaction time, added phosphate ion concentration and reaction time And the relationship between TNFRc-Fc fusion protein concentration, added phosphate ion concentration and reaction time, TNFRc-Fc fusion protein concentration, added calcium ion concentration and reaction time, and Correlation with TNFRc-Fc fusion protein concentration, added phosphate and calcium ion concentrations, reaction time and TNFRc-Fc fusion protein concentration.
표 7
실시예 5: 칼슘인산염 침전의 pH 조건 확인 시험Example 5 pH Condition Confirmation Test of Calcium Phosphate Precipitation
표 4 내지 7의 통계적인 분석(ANOVA, 변량분석)결과를 바탕으로, 단백질 수율과 단백질 다량체 제거 효과를 증진시키기 위해, 아래 표 8과 같은 추가 시험을 진행했다.Based on the results of the statistical analysis (ANOVA, variance analysis) of Tables 4 to 7, in order to enhance the protein yield and protein multimer elimination effect, further tests, such as Table 8 below, were conducted.
표 8
1) 희석 전 항-CD20 항체와 TNFRc-Fc 융합 단백질 농도는 각각, 4.8906 mg/mL, 4.7047 mg/mL 이었다. 1) The pre-dilution anti-CD20 antibody and TNFRc-Fc fusion protein concentrations were 4.8906 mg / mL and 4.7047 mg / mL, respectively.
2)모든 인자들의 중앙값에 대해서는 통계적인 분석을 위해 3회을 진행했다. 2) The median of all factors was performed three times for statistical analysis.
실시예 6: 칼슘인산염 침전을 위한 pH 조건 확인 시험에 대한 통계적인 평가Example 6: Statistical Evaluation of pH Condition Identification Test for Calcium Phosphate Precipitation
6-1. 항-CD20 항체6-1. Anti-CD20 antibody
표 8 시험의 통계적인 분석(ANOVA, 변량분석) 결과 아래 표 9와 같은 결과를 얻었다. Results of statistical analysis (ANOVA, variance analysis) of the test were obtained as shown in Table 9 below.
표 9
1) 희석 전 항-CD20 항체 농도는 각각, 4.8906 mg/mL 이었다. 1) The anti-CD20 antibody concentration before dilution was 4.8906 mg / mL, respectively.
2) 모든 인자들의 중앙값에 대해서는 통계적인 분석을 위해 3회를 진행했다. 2) The median of all factors was performed three times for statistical analysis.
3) 아무것도 처리하지 않은 항-CD20 항체의 다량체 (%)와 단량체 (%)는 각각, 0.78% 와 99.28% 이다. 3) The multimer (%) and monomer (%) of the untreated anti-CD20 antibody were 0.78% and 99.28%, respectively.
표 9의 결과를 가지고 정제를 위한 첨가된 CaCl2과 인산염와 추가적인 인자들을 찾기 위해 통계프로그램(Design expert software. ver. 7.1.1)을 이용하여 통계적인 분석(Analysis of variance, ANOVA, 변량분석)을 진행하였다.Table 9 with the result of the CaCl 2 and insanyeomwa additional factors to find statistics program added for purification (. Design expert software. Ver 7.1.1 ) utilized in the statistical analysis (Analysis of variance, ANOVA, analysis of variance) a Proceeded.
그 결과 표 10에서 보는 것처럼, 5 mM 인산염 농도와 10 mM CaCl2 농도 일 때, 항-CD20 항체 수율은 반응 pH(P < 0.0009), 항-CD20 항체의 농도(P < 0.0058), 반응 pH와 항-CD20 항체의 상호관계(P < 0.0047) 에 대해 P < 0.05로 통계적인 유의성이 있었다. As a result, as shown in Table 10, at 5 mM phosphate concentration and 10 mM CaCl 2 concentration, the anti-CD20 antibody yield was determined by the reaction pH ( P <0.0009), the concentration of anti-CD20 antibody ( P <0.0058), There was statistical significance of P <0.05 for the correlation ( P <0.0047) of the anti-CD20 antibody.
즉, 칼슘인산 침전에 의한 항-CD20 항체의 정제법에서 항-CD20 항체 수율은 첨가된 칼슘이온과 인산이온 외에도, 항-CD20 항체의 농도, 반응 pH, 그리고, 항-CD20 항체 농도와 반응 pH의 상호관계에 영향을 받는다. In other words, the anti-CD20 antibody yield in the purification method of anti-CD20 antibody by calcium phosphate precipitation, in addition to the added calcium and phosphate ions, the concentration of the anti-CD20 antibody, reaction pH, and anti-CD20 antibody concentration and reaction pH Affected by the interrelationship of
표 10
또한, 표 11에서 보는 것처럼 항-CD20 항체에 대한 분석용 크기 배제 크로마토그래피(SEC) 결과를 통해, 5mM 인산염 농도와 5mM CaCl2 농도로 고정 시, 항-CD20 항체 다량체 제거능은 항-CD20 항체의 농도(P < 0.0120)와 반응 pH(P < 0.0051)에 대해 P < 0.05로 통계적인 유의성이 있었다.In addition, through analytical size exclusion chromatography (SEC) results for anti-CD20 antibodies as shown in Table 11, the anti-CD20 antibody multimer removal ability was fixed at 5 mM phosphate concentration and 5 mM CaCl 2 concentration. There was a statistical significance of P <0.05 for the concentration of ( P <0.0120) and the reaction pH ( P <0.0051).
즉, 칼슘인산 침전법에 의한 항-CD20 항체의 정제법에서 항-CD20 항체 다량체 제거 정도는 첨가된 칼슘이온과 인산이온 외에도, 항-CD20 항체의 농도 반응 pH에 영향을 받는다.That is, the degree of anti-CD20 antibody multimer removal in the purification method of the anti-CD20 antibody by calcium phosphate precipitation is influenced by the concentration reaction pH of the anti-CD20 antibody in addition to the added calcium and phosphate ions.
표 11
표 10, 표 11의 결과를 바탕으로, 칼슘인산 침전법을 이용한 항-CD20 항체 정제에 대한 최적화 조건을 통계프로그램(Design expert software. ver. 7.1.1)을 통해서 표 12와 같이 8가지 정제 조건을 얻었다. 이 8가지 조건은, 반응시간(3~5 시간), 항-CD20 항체 농도(1X~3X 희석), pH 6.0~7.0으로 제한하고, 이때, 항-CD20 항체 수율이 90% 이상이고, 항-CD20 항체 단량체 비율이 99.5% 이상을 얻을 수 있는 조건으로 제한했을 때 통계적 계산에 의해 얻어진 조건들이다. Based on the results of Table 10 and Table 11, eight purification conditions as shown in Table 12 through the statistical program (Design expert software. Ver. 7.1.1) to optimize the optimization conditions for the anti-CD20 antibody purification using calcium phosphate precipitation method Got. These eight conditions are limited to reaction time (3-5 hours), anti-CD20 antibody concentration (1X-3X dilution), pH 6.0-7.0, wherein the anti-CD20 antibody yield is at least 90% and anti- These are the conditions obtained by statistical calculation when the ratio of the CD20 antibody monomer ratio is limited to the condition that can obtain 99.5% or more.
표 12
6-2. TNFRc-Fc 융합 단백질6-2. TNFRc-Fc fusion protein
표 8 시험결과의 통계적인 분석(ANOVA, 변량분석) 결과 아래 표 13과 같은 결과를 얻었다. Table 8 Results of statistical analysis (ANOVA, variance analysis) The results as shown in Table 13 below were obtained.
표 13
1) 희석 전 TNFRc-Fc 융합 단백질 농도는 4.7047 mg/mL 이었다. 1) TNFRc-Fc fusion protein concentration before dilution was 4.7047 mg / mL.
2)모든 인자들의 중앙값에 대해서는 통계적인 분석을 위해 3회를 진행했다. 2) The median of all factors was performed three times for statistical analysis.
3) 아무것도 처리하지 않은 TNFRc-Fc 융합 단백질의 다량체 (%), 단량체 (%), 분절체(%)는 각각, 2.11%, 96.58%, 1.3%이다. 3) The multimer (%), monomer (%) and segment (%) of the untreated TNFRc-Fc fusion protein were 2.11%, 96.58% and 1.3%, respectively.
표 13의 결과를 가지고 정제를 위해 첨가된 CaCl2과 인산염과 추가적인 인자들을 찾기 위해 통계프로그램(Design expert software. ver. 7.1.1)을 이용하여 통계적인 분석 (Analysis of variance, ANOVA, 변량분석)을 진행하였다.Table 13 with the results of the addition of CaCl 2 to the purified and phosphate and an additional factor to find statistical program (Design expert software. Ver. 7.1.1 ) to statistical analysis using the (Analysis of variance, ANOVA, analysis of variance) Proceeded.
그 결과 표 14에서 보는 바와 같이, 10 mM 인산염 농도와 10 mM CaCl2 농도 일 때, TNFRc-Fc 융합 단백질 수율은, TNFRc-Fc 융합 단백질의 농도(P < 0.0107), 반응 pH(P < 0.0011), 반응 시간과 TNFRc-Fc 융합 단백질의 농도의 상호관계(P < 0.0317), TNFRc-Fc 융합 단백질 농도와 반응 pH의 상호관계(P < 0.0149), 그리고, 반응시간와 TNFRc-Fc 융합 단백질의 농도 그리고, 반응 pH의 상호관계(P < 0.0201)에 대해 P < 0.05로 통계적인 유의성이 있었다. As a result, as shown in Table 14, at 10 mM phosphate concentration and 10 mM CaCl 2 concentration, the yield of TNFRc-Fc fusion protein was calculated as the concentration of TNFRc-Fc fusion protein ( P <0.0107), reaction pH ( P <0.0011). , The relationship between the reaction time and the concentration of TNFRc-Fc fusion protein ( P <0.0317), the relationship between the TNFRc-Fc fusion protein concentration and the reaction pH ( P <0.0149), the reaction time and the concentration of TNFRc-Fc fusion protein, and , P <0.05 for the correlation of reaction pH ( P <0.0201).
즉, 칼슘인산 침전에 의한 TNFRc-Fc 융합 단백질의 정제법에서 TNFRc-Fc 융합 단백질의 수율은 첨가된 칼슘이온과 인산이온 외에도, TNFRc-Fc 융합 단백질의 농도와 반응 pH, 반응시간과 TNFRc-Fc 융합 단백질의 상호관계, TNFRc-Fc 융합 단백질농도와 반응 pH의 상호관계, 그리고, 반응시간과 TNFRc-Fc 융합 단백질의 농도 그리고 반응 pH의 상호관계에 영향을 받는다. In other words, the yield of TNFRc-Fc fusion protein in the purification method of TNFRc-Fc fusion protein by calcium phosphate precipitation, in addition to the calcium and phosphate ions added, the concentration, reaction pH, reaction time and TNFRc-Fc of TNFRc-Fc fusion protein The relationship between the fusion protein, the TNFRc-Fc fusion protein concentration and the reaction pH, and the reaction time, the concentration of the TNFRc-Fc fusion protein, and the reaction pH are affected.
표 14
또한, 표 15에서 보는 바와 같이, TNFRc-Fc 융합 단백질에 대한 분석용 크기배제크로마토그래피(SEC) 결과를 통해, 인산염농도와 CaCl2 농도를 각각 10 mM로 고정 시, TNFRc-Fc 융합 단백질 다량체 제거능은 TNFRc-Fc 융합 단백질의 농도(P < 0.0004), 반응 pH(P < 0.0001), 반응시간과 TNFRc-Fc 융합 단백질 농도와의 상호관계(P < 0.007), 반응시간과 반응 pH의 상호관계(P < 0.0014), TNFRc-Fc 융합 단백질 농도와 반응 pH 의 상호관계(P < 0.0011), 그리고, 반응 시간과 TNFRc-Fc 융합 단백질 농도, 그리고, 반응 pH의 상호관계(0.001)에 대해 P < 0.05로 통계적인 유의성이 있었다. In addition, as shown in Table 15, TNFRc-Fc fusion protein multimers when the phosphate concentration and the CaCl 2 concentration were fixed at 10 mM through analytical size exclusion chromatography (SEC) results for the TNFRc-Fc fusion protein, respectively. Scavenging ability was determined by the concentration of TNFRc-Fc fusion protein ( P <0.0004), reaction pH ( P <0.0001), correlation between reaction time and TNFRc-Fc fusion protein concentration ( P <0.007), and correlation between reaction time and reaction pH. (P <0.0014), for TNFRc-Fc fusion protein interaction (P <0.0011) of concentration and reaction pH, and reaction time and TNFRc-Fc fusion protein concentrations, then the reaction pH correlation (0.001) P < There was a statistical significance of 0.05.
즉, 칼슘인산 침전법에 의한 TNFRc-Fc 융합 단백질의 정제법에서 TNFRc-Fc 융합 단백질 다량체 제거 정도는, 첨가된 칼슘이온과 인산이온 외에도, TNFRc-Fc 융합 단백질 농도, 반응 pH, 반응시간과 TNFRc-Fc 융합 단백질 농도의 상호관계, 반응 시간과 반응 pH의 상호관계, TNFRc-Fc 융합 단백질 농도와 반의 pH의 상호관계 그리고 반응시간과 TNFRc-Fc 융합 단백질 농도, 그리고, 반응 pH의 상호관계에 영향을 받는다.In other words, TNFRc-Fc fusion protein multimer removal in the purification method of TNFRc-Fc fusion protein by calcium phosphate precipitation method, in addition to the added calcium and phosphate ions, TNFRc-Fc fusion protein concentration, reaction pH, reaction time and The relationship between TNFRc-Fc fusion protein concentration, reaction time and reaction pH, TNFRc-Fc fusion protein concentration and half pH, and reaction time and TNFRc-Fc fusion protein concentration, and reaction pH get affected.
표 15
표 14, 표 15 결과를 바탕으로, 칼슘인산 침전법을 이용한 TNFRc-Fc 융합 단백질 정제에 대한 최적화 조건을 통계프로그램(Design expert software. ver. 7.1.1)을 통해서 표 16과 같이 10가지 정제 조건을 얻었다. 이 10 가지 조건은, 반응시간(3~5 시간), TNFRc-Fc 융합 단백질 농도(1X~3X 희석), pH 6.0~7.0으로 제한하고, 이때, TNFRc-Fc 융합 단백질 수율이 80% 이상이고 TNFRc-Fc 융합 단백질 단량체 비율이 98.5% 이상을 얻을 수 있는 조건으로 제한했을 때 통계적 계산에 의해 얻어진 조건들이다. Based on the results of Table 14 and Table 15, 10 purification conditions as shown in Table 16 through the statistical program (Design expert software. Ver. 7.1.1), the optimization conditions for the purification of TNFRc-Fc fusion protein using calcium phosphate precipitation method Got. These 10 conditions are limited to reaction time (3-5 hours), TNFRc-Fc fusion protein concentration (1X-3X dilution), pH 6.0-7.0, where TNFRc-Fc fusion protein yield is 80% or higher and TNFRc The conditions obtained by statistical calculation when the -Fc fusion protein monomer ratio was limited to a condition that can obtain more than 98.5%.
표 16
실시예 7: 칼슘인산염 침전에 의한 TNFRc-Fc 융합 단백질 정제효과 시험Example 7 TNFRc-Fc Fusion Protein Purification Effect Test by Calcium Phosphate Precipitation
상기 실시예에서 시험한 TNFα 수용체-Fc(TNFRc-Fc) 융합 단백질에 대해 표 17과 같은 조건으로 칼슘인산 침전 반응을 시켜, 반응전과 반응 후의 TNFRc-Fc 융합 단백질의 다량체량을 비교하였다. The TNFα receptor-Fc (TNFRc-Fc) fusion protein tested in the above Example was subjected to calcium phosphate precipitation reaction under the conditions shown in Table 17, and the multimer weight of the TNFRc-Fc fusion protein before and after the reaction was compared.
그 결과, 도 3a, 3b에서 보는 바와 같이, TNFRc-Fc 융합 단백질의 다량체가 1.59%에서 0.29%로 약 82% 감소된 것을 확인하였다.As a result, as shown in Figure 3a, 3b, it was confirmed that the multimer of the TNFRc-Fc fusion protein was reduced by about 82% from 1.59% to 0.29%.
표 17
실시예 8: 칼슘인산염 침전에 의한 항-ErbB3 항체 정제효과 시험Example 8 Anti-ErbB3 Antibody Purification Effect Test by Calcium Phosphate Precipitation
상기 실시예에서 시험한 항-CD20 항체와 TNFRc-Fc 융합 단백질 외에 또 다른 항체에 대해 칼슘인산염 침전 방법이 효과가 있는지를 항-ErbB3 항체를 가지고 확인해 보았다. 항-ErbB3 항체를 포함하는 동물세포 배양액을 단백질 A 크로마토그래피를 통해 정제를 진행했다. 단백질 A 컬럼으로부터 50 mM 아세테이트, pH 3.5 완충액을 이용해서 항-ErbB3 항체를 용출하고, 항-ErbB3 항체가 포함된 용출액을 트리스염기로 pH 7.0이 되게 만든 후 아래 표 18과 같은 조건으로 칼슘인산 침전 반응을 시켜, 반응전과 반응 후의 항-ErbB3 항체 다량체량을 비교하였다. In addition to the anti-CD20 antibody and the TNFRc-Fc fusion protein tested in the above example, it was confirmed with the anti-ErbB3 antibody whether the calcium phosphate precipitation method is effective. Animal cell cultures containing anti-ErbB3 antibodies were purified by Protein A chromatography. Anti-ErbB3 antibody was eluted from the Protein A column using 50 mM acetate and pH 3.5 buffer, and the eluate containing the anti-ErbB3 antibody was adjusted to pH 7.0 with trisbase, and calcium phosphate precipitated under the conditions shown in Table 18 below. The reaction was performed to compare the amount of anti-ErbB3 antibody multimers before and after the reaction.
그 결과, 도 4a, 4b에서 보는 바와 같이, 항-ErbB3 항체의 다량체가 4.18%에서 2.28%로 약 45% 감소된 것을 확인하였다.As a result, as shown in Figures 4a, 4b, it was confirmed that the multimer of the anti-ErbB3 antibody was reduced by about 45% from 4.18% to 2.28%.
표 18
실시예 9: 칼슘인산 침전에 의한 단백질 다량체 외의 불순물 제거 효과 확인 시험Example 9: Test to confirm the effect of removing impurities other than the protein multimer by calcium phosphate precipitation
칼슘인산염 침전을 이용한 정제 방법이 항체 또는 단백질 다량체 제거 외에 다른 불순물 제거에 효과가 있는 것을 확인하기 위해, 항-CD20 항체와 TNFRc-Fc 융합 단백질을 이용하여, 표 12, 표 16에서 얻은 해당 항체 또는 단백질의 다량체 제거에 최적인 조건들에서 선별하여 수행하였다. In order to confirm that the purification method using calcium phosphate precipitation is effective in removing impurities other than antibody or protein multimer removal, the corresponding antibodies obtained in Table 12 and Table 16 using anti-CD20 antibody and TNFRc-Fc fusion protein Or selected under conditions optimal for multimer removal of the protein.
아래 표 19에서 보는 바와 같이, 항-CD20 항체와 TNFRc-Fc 융합 단백질에 대해 칼슘인산염 침전을 수행하고, 숙주세포 유래 단백질, DNA, 단백질 A 잔여물과 같은 불순물들에 대해 분석을 했다. As shown in Table 19 below, calcium phosphate precipitation was performed on the anti-CD20 antibody and TNFRc-Fc fusion protein and analyzed for impurities such as host cell derived protein, DNA, and protein A residue.
그 결과, 표 20에서 보는 바와 같이 칼슘인산염 침전을 이용한 정제방법이, 항체의 다량체 제거 외에도 숙주세포 유래 단백질, DNA, 단백질 A 잔여물 제거에도 효과가 있음을 확인하였다. 다만, 수율와 다량체 제거능 그리고, 불순물에 대한 제거능은 단백질에 따라 차이를 보였다. 즉, 반응시간, 첨가된 칼슘이온 농도, 첨가된 인산이온 농도의 조건은 단백질에 따라 다를 수 있음을 의미한다. As a result, it was confirmed that the purification method using calcium phosphate precipitation, as shown in Table 20, is effective in removing host cell-derived protein, DNA, and protein A residues in addition to removing multimers of antibodies. However, the yield, multimer removal capacity, and impurities removal ability were different depending on the protein. In other words, the reaction time, the added calcium ion concentration, the added phosphate ion concentration means that the conditions may vary depending on the protein.
표 19
표 20
LOQ; Limit of QuantificationLOQ; Limit of Quantification
(1) 항-CD20 항체 반응 전 시료에서 잔여 DNA양은 검출한계 이하이기 때문에, 반응 전후의 시료에 대해 DNA분석을 진행하지 않았다. (1) Since the amount of residual DNA in the sample before the anti-CD20 antibody reaction was below the detection limit, DNA analysis was not performed on the samples before and after the reaction.
(2) 항-CD20 반응 전 시료에서 잔여 단백질 A 양이 1.01ppm으로 너무 낮기 때문에, 반응 후 시료에 대해서 분석을 진행하지 않았다. (2) Since the amount of residual protein A in the sample before the anti-CD20 reaction was too low at 1.01 ppm, the analysis was not performed on the sample after the reaction.
Claims (9)
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| KR10-2013-0055401 | 2013-05-16 | ||
| KR1020130055401A KR101642551B1 (en) | 2013-05-16 | 2013-05-16 | A method for removing impurities by calcium phosphate precipitation |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2014185716A1 true WO2014185716A1 (en) | 2014-11-20 |
Family
ID=51898623
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/KR2014/004341 Ceased WO2014185716A1 (en) | 2013-05-16 | 2014-05-15 | Method for removing impurities by using calcium phosphate precipitation |
Country Status (2)
| Country | Link |
|---|---|
| KR (1) | KR101642551B1 (en) |
| WO (1) | WO2014185716A1 (en) |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP4118093A4 (en) * | 2020-03-11 | 2024-04-17 | Dr. Reddy's Laboratories Ltd. | Method of purifying an fc-fusion protein |
Citations (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| KR20050119380A (en) * | 2004-06-16 | 2005-12-21 | 주식회사 티앤라이프시스템 | Method for purifying hyaluronic acid using calcium salt and phosphate salt, or calcium phosphate salt |
| US8263750B2 (en) * | 2006-03-16 | 2012-09-11 | Amgen Inc. | Method for purifying a protein using protein-A affinity chromatography using an intermediate wash step |
-
2013
- 2013-05-16 KR KR1020130055401A patent/KR101642551B1/en active Active
-
2014
- 2014-05-15 WO PCT/KR2014/004341 patent/WO2014185716A1/en not_active Ceased
Patent Citations (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| KR20050119380A (en) * | 2004-06-16 | 2005-12-21 | 주식회사 티앤라이프시스템 | Method for purifying hyaluronic acid using calcium salt and phosphate salt, or calcium phosphate salt |
| US8263750B2 (en) * | 2006-03-16 | 2012-09-11 | Amgen Inc. | Method for purifying a protein using protein-A affinity chromatography using an intermediate wash step |
Non-Patent Citations (3)
| Title |
|---|
| PEPYS ET AL.: "Isolation of C-reactive protein by affinity chromatography", CLINICAL AND EXPERIMENTAL IMMUNOLOGY, vol. 30, no. 1, 1997, pages 32 - 37 * |
| SCHLESINGER ET AL.: "Complete covalent structure of statherin, a tyrosine-rich acidic peptide which inhibits calcium phosphate precipitation from human parotid saliva", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 252, no. 5, 1997, pages 1689 - 1695 * |
| ZHANG ET AL.: "Highly efficient phosphopeptide enrichment by calcium phosphate precipitation combined with subsequent IMAC enrichment", MOLECULAR & CELLULAR PROTEOMICS, vol. 6, no. 11, 2007, pages 2032 - 2042 * |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP4118093A4 (en) * | 2020-03-11 | 2024-04-17 | Dr. Reddy's Laboratories Ltd. | Method of purifying an fc-fusion protein |
Also Published As
| Publication number | Publication date |
|---|---|
| KR20140136069A (en) | 2014-11-28 |
| KR101642551B1 (en) | 2016-07-28 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| WO2015053523A1 (en) | Bicistronic expression vector for antibody expression and method for producing antibody using same | |
| WO2010123290A2 (en) | In vivo half life increased fusion protein or peptide maintained by sustained in vivo release, and method for increasing in vivo half-life using same | |
| WO2020197230A1 (en) | Pharmaceutical composition, comprising human hyaluronidase ph20 variant and drug, for subcutaneous injection | |
| WO2018004260A1 (en) | Stable liquid pharmaceutical preparation | |
| WO2022035201A1 (en) | Fusion protein comprising il-12 and anti-fap antibody, and use thereof | |
| WO2019045477A1 (en) | Composition for preventing and treating skin disease comprising substance specifically binding to vimentin-derived peptide | |
| WO2016199964A1 (en) | Antibody specifically binding to isolated vimentin-derived peptide, or binding fragment of peptide | |
| WO2015064971A1 (en) | Method for isolating isoforms of antibody using positive ion exchange chromatography | |
| WO2014185716A1 (en) | Method for removing impurities by using calcium phosphate precipitation | |
| WO2024155098A1 (en) | Regulatory t cell surface antigen and antibody that specifically binds thereto | |
| WO2022146036A1 (en) | Medium composition for culturing animal cells for producing recombinant extracellular matrix protein and method of using the same | |
| WO2021010532A1 (en) | Method for preparing long-acting drug conjugate through preparation of novel intermediate | |
| WO2024242490A1 (en) | Ch3 domain variant or bispecific antibody comprising same | |
| WO2023068740A1 (en) | Method for purifying fusion protein having igg fc domain | |
| WO2024085697A1 (en) | Stable antibody composition | |
| WO2024172363A1 (en) | Anti-cntn4 antibody and uses thereof | |
| WO2021137655A1 (en) | Antibody for super-repressor ikb, and use thereof | |
| WO2023287212A1 (en) | Epitope of regulatory t cell surface antigen, and antibody specifically binding thereto | |
| WO2023132698A1 (en) | Pharmaceutical composition for preventing or treating degenerative brain diseases, containing glucagon-like peptide-1 and interleukin-1 receptor antagonist | |
| WO2019235895A1 (en) | Transgenic mouse for aglycosylated antibody production and use of aglycosylated antibody produced therefrom | |
| WO2018212556A1 (en) | A method for purifying an antibody or an antibody fragment thereof using affinity chromatography | |
| WO2025254428A1 (en) | Method for purifying single-stranded rna | |
| WO2025147169A1 (en) | Multispecific antibody for lysosomal degradation | |
| WO2025183487A1 (en) | Anti-tigit/anti-pd-l1 bispecific antibody and use thereof | |
| WO2020138868A1 (en) | Type 1 interferon neutralizing fc-fusion protein and use thereof |
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: 14797476 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: 14797476 Country of ref document: EP Kind code of ref document: A1 |