WO2011015926A1 - A process of fermentation, purification and production of recombinant soluble tumour necrosis factor alfa receptor (tnfr) - human igg fc fusion protein - Google Patents

A process of fermentation, purification and production of recombinant soluble tumour necrosis factor alfa receptor (tnfr) - human igg fc fusion protein Download PDF

Info

Publication number
WO2011015926A1
WO2011015926A1 PCT/IB2010/001912 IB2010001912W WO2011015926A1 WO 2011015926 A1 WO2011015926 A1 WO 2011015926A1 IB 2010001912 W IB2010001912 W IB 2010001912W WO 2011015926 A1 WO2011015926 A1 WO 2011015926A1
Authority
WO
WIPO (PCT)
Prior art keywords
tnfr
necrosis factor
supernatant
region
cell culture
Prior art date
Application number
PCT/IB2010/001912
Other languages
French (fr)
Inventor
Villoo Morawala Patell
Sunit Maity
Original Assignee
Avesthagen Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Avesthagen Limited filed Critical Avesthagen Limited
Publication of WO2011015926A1 publication Critical patent/WO2011015926A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7151Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for tumor necrosis factor [TNF], for lymphotoxin [LT]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • TNFR Tumour Necrosis Factor Alfa Receptor
  • the present invention relates generally to the use of novel fermentation and chromatographic procedures separately and jointly for the production of recombinant soluble Tumour Necrosis Factor Alfa receptor (TNFR) - Human IgG Fc fusion protein, in biologically active form from fluids, especially mammalian host cell culture supernatants.
  • TNFR Tumour Necrosis Factor Alfa receptor
  • ENBREL (Etanercept) is a recombinant fusion protein comprising the extracellular domain of the human tumor necrosis factor receptor superfamily, member IB (p75) and the Fc domain of human IgGl.
  • ENBREL is an important scientific advance that in many people has been shown to reduce the signs and symptoms of rheumatoid arthritis, polyarticular-course juvenile rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, and psoriasis.
  • TNF is a naturally occurring cytokine that the signs and symptoms of rheumatoid arthritis, polyarticular-course juvenile rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, and psoriasis.
  • TNF is a naturally occurring cytokine that is involved in normal inflammatory and immune responses. It plays an important role in the inflammatory processes of rheumatoid arthritis (RA), polyarticular-course juvenile rheumatoid arthritis (JRA) and the resulting joint pathology. Elevated levels of TNF are found in the synovial fluid of RA patients.
  • the main object of the present invention is to use novel fermentation and chromatographic procedures for rapid and efficient recovery of recombinant soluble Tumour Necrosis Factor Alfa receptor (TNFR) - Human IgG Fc fusion protein from cell culture supernatant
  • TNFR Tumour Necrosis Factor Alfa receptor
  • the present invention relates to the use of novel fermentation process for the overexpression of recombinant soluble Tumour Necrosis Factor Alfa receptor (TNFR) - Human IgG Fc fusion protein in CHO cells.
  • TNFR Tumour Necrosis Factor Alfa receptor
  • the present invention also relates to the use of novel chromatographic procedures separately and jointly for the production of recombinant soluble Tumour Necrosis Factor Alfa receptor (TNFR) - Human IgG Fc fusion protein, in biologically active form from fluids, especially mammalian host cell culture supernatants. .
  • TNFR Tumour Necrosis Factor Alfa receptor
  • Figure 4 Process chromatogram after affinity chromatography
  • Lane No. 1 Molecular weight Marker
  • Lane No. 2 RMP
  • Lane No. 3 :
  • Lane No. 1 Molecular weight Marker
  • Lane No. 2 RMP
  • Lane No. 3 :
  • Formulated Drug Substance Figure 9 Western Blot Analysis of Drug substance showing comparable immuno-specificity between RMP and drug substance where Lane No. 1 : Molecular weight Marker, Lane No. 2: RMP and Lane No. 3: Formulated Drug Substance
  • Figure 13 Comparable hydrophobicity profile between Drug substance and RMP as depicted by
  • FIG. 20 CD spectroscopy analysis has revealed a comparable (a) secondary and (b) tertiary structure profile between RMP & Drug substance
  • Figure 21 The in vitro cytotoxic effect of TNF has been extensively demonstrated using the reporter cell line L929 that has been derived from a murine fibrosarcoma.
  • the potency value of the sample is calculated using CFR 21 /part 11 compliance with Parallel line assay (PLA) software.
  • the present invention provides an improved process for the cell culture manufacturing of Recombinant TNFR:Fc.
  • the invention provides systems that help in the high cell density cell culture process, maintenance of high cell viability for a longer culture period.
  • the invention also helps in achieving proper glycosylation of Recombinant TNFR:Fc.
  • the cell culture manufacturing process starts with seeding the bioreactor at a predefined cell density in chemically defined medium.
  • the culture is fed in two stages, primary feeding which is designed to achieve high cell growth and, secondary feeding which is designed to maintain the higher cell viability and hyper glycosylation of the Recombinant TNFR:Fc.
  • the invention also relates to bioreactor operation procedure for the manufacturing of Recombinant TNFR: Fc.
  • This present invention relates to the rapid and efficient recovery of Recombinant TNFRrFc from cell culture supernatant from Cell culture fluid by means of Affinity chromatography. These chromatographic steps are used for capture of recombinant TNFR:Fc. This separation involves in selective binding of the desired compound to specific affinity resin and then elution with elution buffer. Culture supernatants are preferably concentrated and clarified before chromatographic treatment. TNFR:Fc containing eluent fractions are enriched with biologically active material, but they will be subjected to further processing by Anion exchange chromatographic steps. In this process the active materials are colleted in flow through. These processes are used for removal of process related impurities like host cell protein and host cell DNA.
  • the invention also relates to the next chromatographic procedure where the flow-through from the previous step is further subjected to a Cation exchange chromatographic step.
  • the active material is eluted with elution buffer containing salt.
  • the present invention also relates to the recombinant TNFRiFc recovery procedure involving serial application different chromatographic techniques as mentioned previously. All different steps, conditions and compositions are disclosed in the invention.
  • Bioreactor Before seeding the Bioreactor, it was assembled and sterilized by autoclaving at 121°C for 60 minutes. After sterilization, Bioreactor was charged with 7000ml of commercially available animal component free, chemically defined media. Afterwards, the bioreactor was kept under positive pressure with air at a flow rate of 0.2 Litre per minute. The bioreactor was aerated over night for 100% air saturation. The dO2 electrode was calibrated after stabilization of dissolved oxygen value. Sterile connection was created between the seed bottle and the seed port on the bioreactor head plate. The seed was then aseptically transferred to the bioreactor using peristaltic pump. The bioreactor was seeded with the density of 0.5 x 10 6 cells/mL. After seeding, the bioreactor was allowed to run at following pre-set parameters:
  • the bioreactor was sampled at every 24/48 hours for in process quality control analysis.
  • the bioreactor process was a fed - batch process with feeding of different nutrients at definite culture stages. During first 120 hrs of culture time, the bioreactor was daily fed with 6OmL of primary feed that comprise of glucose, lipids, amino acids, vitamins, trace elements, cholesterol and growth factors. Starting from 144 hrs of culture age the bioreactor was daily fed with 10OmL of secondary feed that comprise of Galactose and Mannose.
  • the bioreactor was harvested at a cell viability of 60 - 70%.
  • the growth pattern, protein expression profile and nutrient consumptions are depicted (Fig 1-3).
  • Clarification of the cell culture harvest was carried out by using a cellulose disposable filter with 650 - 1000 cm 2 effective filtration area and with an operating pressure of not more than 30 psi. The filtrate was checked for turbidity and target protein content.
  • the clarified harvest was diafilitered and buffer exchanged with Tris buffer pH 7.2 - 7.6 using a 30 kDa - TFF membrane at a Trans Membrane Pressure of 5 - 10 psi.
  • Affinity chromatography was used in binding and elution mode with column of 30 mm diameter for capturing; with Tris buffer pH 7.2 - 7.6 as equilibration buffer.
  • the column was equilibrated with Tris buffer pH 6.8 - 7.2. Protein of interest is collected in flow through. This step was used for the removal of process related impurities like leachate protein A, host cell DNA and host cell protein. (Fig 5). Thereafter, the flow through was filtered for virus removal using viral removal filter having an effective filtration area of 0.01 m 2 . Anion exchange chromatography was carried out with the nanofiltrate after equilibrating the column with Tris buffer pH 6.8-7.2. The protein of interest was eluted with elution buffer using NaCl salt gradient. This step was used for the removal of process related impurities like host cell DNA and host cell protein (Fig 6).
  • the eluate was buffer exchanged and concentrated using a 30 kDa TFF membrane at a Trans Membrane Pressure (TMP) of 5 - 10 psi .
  • TMP Trans Membrane Pressure
  • the buffer exchanged protein solution was filtered using 0.2 ⁇ m filter.
  • the drug substance was characterized as per the specifications.
  • the Drug Substance (Active Pharmaceutical Ingredient) was formulated using formulation buffer containing (lOmg/mL Sucrose, 5.8mg/mL of Sodium chloride, 5.3mg/mL of L-Arginine hydrochloride, 2.6mg/mLSodium phosphate monobasic monohydrate and 0.9mg/mLsodium phosphate dibasic anhydrous) and adjusted to pH 6.3 ⁇ 0.2.
  • the formulated material was characterized as per the specifications set by product development specification._A 12% SDS PAGE under reducing condition was studied and showed single corresponding band at -75 kDa , matches with RMP (Fig 7). A 8% non-reducing SDS-PAGE data is mentioned in which shows a single band corresponds to approximately 150 kDa matches with RMP (Fig 8). Western blot analysis showed a clear comparable immune-specificity with that of RMP for both reducing and non-reducing condition (Fig 9). An Isoelectric focusing was carried out to identify the isoelectric point of the target protein which matched with that of RMP (Fig 10). A 2-Dimensional gel electrophoresis profile of the purified drug substance matched with RMP (Fig 11).
  • Protein A HPLC profile carried out during this step showed a retention time of 6.3 minutes in drug substance, which was very much comparable with the RMP (6.3 minutes) (Fig 12).
  • RP-HPLC was performed to compare the hydrophobicity profile between the purified drug substance and the RMP. The result showed comparable RT (Fig 13).
  • Size exclusion chromatography for determination of oligomeric status showed a retention time of 6.9 minutes (% of main peak is 100) for drug substance which was very much comparable with the RMP (6.9 minutes, % area of main peak 100 [Fig 14]).
  • UV spectral analysis has demonstrated comparable primary structure profile between the purified drug substance and RMP (Fig 19).
  • CD spectral analysis has demonstrated comparable secondary and tertiary structure profile between the purified drug substance and RMP (Fig 20).
  • Cytotoxicity rescue assay with L929 cell line showed the degree of TNF alpha induced cytotoxicity observed was inversely proportional to increasing amount of RMP and drug substance (Fig 21).

Abstract

The present invention relates to the use of novel fermentation and chromatographic procedures separately and jointly for the production of recombinant soluble Tumour Necrosis Factor Alfa receptor (TNFR) - Human IgG Fc fusion protein, in biologically active form from fluids, especially mammalian host cell culture supernatants.

Description

"A Process Of Fermentation, Purification And Production Of Recombinant Soluble Tumour Necrosis Factor Alfa Receptor (TNFR) - Human IgG Fc Fusion Protein"
The following specification particularly describes and ascertains the nature of this invention and the manner in which it is to be performed:
FIELD OF THE INVENTION
The present invention relates generally to the use of novel fermentation and chromatographic procedures separately and jointly for the production of recombinant soluble Tumour Necrosis Factor Alfa receptor (TNFR) - Human IgG Fc fusion protein, in biologically active form from fluids, especially mammalian host cell culture supernatants.
BACKGROUND AND PRIOR ART OF THE INVENTION
In past, various media and methods were used for the cell culture manufacturing of recombinant glycoprotein or monoclonal antibody. Commonly employed bioreactor process includes; batch, semi fed-batch, fed-batch, perfusion and continuous fermentation. The ever-increasing demand of monoclonal antibody and other recombinant proteins in properly glycosyalted forms have increased the prospects of cell culture process development. In addition the regulatory hurdles imposed on the serum containing process has led to the development of cell culture process in a completely chemically defined environment.
Numerous techniques have in the past been applied in preparative separations of biochemically significant materials. Commonly employed preparative separatory techniques include: ultrafiltration, column electrofocusing, flatbed electrofocusing, gel filtration, electrophoresis, isotachophoresis and various forms of chromatography. Among the commonly employed chromatoghraphic techniques are ion exchange and adsorption chromatography. The extensive application of recombinant methodologies to large-scale purification and production of eukaryotic protein has increased the prospect of obtaining the molecule in required quantity using simplified purification procedures.
ENBREL (Etanercept) is a recombinant fusion protein comprising the extracellular domain of the human tumor necrosis factor receptor superfamily, member IB (p75) and the Fc domain of human IgGl. ENBREL is an important scientific advance that in many people has been shown to reduce the signs and symptoms of rheumatoid arthritis, polyarticular-course juvenile rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, and psoriasis. TNF is a naturally occurring cytokine that the signs and symptoms of rheumatoid arthritis, polyarticular-course juvenile rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, and psoriasis. TNF is a naturally occurring cytokine that is involved in normal inflammatory and immune responses. It plays an important role in the inflammatory processes of rheumatoid arthritis (RA), polyarticular-course juvenile rheumatoid arthritis (JRA) and the resulting joint pathology. Elevated levels of TNF are found in the synovial fluid of RA patients.
OBJECTIVES OF THE INVENTION
The main object of the present invention is to use novel fermentation and chromatographic procedures for rapid and efficient recovery of recombinant soluble Tumour Necrosis Factor Alfa receptor (TNFR) - Human IgG Fc fusion protein from cell culture supernatant
SUMMARY OF THE INVENTION
The present invention relates to the use of novel fermentation process for the overexpression of recombinant soluble Tumour Necrosis Factor Alfa receptor (TNFR) - Human IgG Fc fusion protein in CHO cells.
The present invention also relates to the use of novel chromatographic procedures separately and jointly for the production of recombinant soluble Tumour Necrosis Factor Alfa receptor (TNFR) - Human IgG Fc fusion protein, in biologically active form from fluids, especially mammalian host cell culture supernatants. .
LIST OF FIGURES
Figure 1 : Nutrient consumption and lactate accumulation profile during fermentation run
Figure 2: Cell growth and viability profile during fermentation run
Figure 3: Expression profile of protein during fermentation run
Figure 4: Process chromatogram after affinity chromatography
Figure 5: Process chromatogram after Cation Exchange chromatography
Figure 6: Process chromatogram after Anion Exchange chromatography
Figure 7: Electrophoretic pattern of Drug substance showing comparable molecular weight with
RMP where Lane No. 1: Molecular weight Marker, Lane No. 2 : RMP and Lane No. 3 :
Formulated Drug Substance
Figure 8: Electrophoretic pattern of Drug substance showing comparable molecular weight with
RMP where Lane No. 1: Molecular weight Marker, Lane No. 2 : RMP and Lane No. 3 :
Formulated Drug Substance Figure 9: Western Blot Analysis of Drug substance showing comparable immuno-specificity between RMP and drug substance where Lane No. 1 : Molecular weight Marker, Lane No. 2: RMP and Lane No. 3: Formulated Drug Substance
Figure 10: Isoelectric focussing of Drug substance when compared to RMP had demonstrated a comparable pi range and charge variant profile where Lane No. 1: RMP and Lane No. 2:
Formulated Drug Substance
Figure 11 : Comparable 2D Gel electrophoresis pattern of Drug substance with that of RMP
Figure 12: Protein A HPLC profile of Drug substance showed comparable retention time with that of RMP
Figure 13: Comparable hydrophobicity profile between Drug substance and RMP as depicted by
Reverse Phase HPLC profile
Figure 14: Size exclusion HPLC profile of Drug substance showed comparable similar hydrodynamic radius
Figure 15: MALDI-TOF analysis of the intact molecule has determined the molecular mass of
RMP and Drug substance to be -145 kDa
Figure 16: The deconvolved mass spectral profile obtained by MALDI-TOF analysis of the tryptic digests demonstrated a high degree of similarity between RMP & Drug substance
Figure 17: HPLC-based tryptic peptide mapping analysis has shown identical profiles between
RMP & Drug substance
Fig 18: Glycan analysis depicted above has demonstrated a comparable Glycan profile thus indicating a high-degree of similarity between RMP & Drug substance
Figure 19: UV spectroscopy analysis has revealed a comparable primary structure profile between
RMP & Drug substance
Figure 20: CD spectroscopy analysis has revealed a comparable (a) secondary and (b) tertiary structure profile between RMP & Drug substance
Figure 21 : The in vitro cytotoxic effect of TNF has been extensively demonstrated using the reporter cell line L929 that has been derived from a murine fibrosarcoma. The assay read outs indicated that the degree of TNF-alpha induced cytotoxicity observed was inversely proportional to increasing amount of RMP / Drug substance added. The potency value of the sample is calculated using CFR 21 /part 11 compliance with Parallel line assay (PLA) software.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides an improved process for the cell culture manufacturing of Recombinant TNFR:Fc. In particular, the invention provides systems that help in the high cell density cell culture process, maintenance of high cell viability for a longer culture period. In addition the invention also helps in achieving proper glycosylation of Recombinant TNFR:Fc. The cell culture manufacturing process starts with seeding the bioreactor at a predefined cell density in chemically defined medium. The culture is fed in two stages, primary feeding which is designed to achieve high cell growth and, secondary feeding which is designed to maintain the higher cell viability and hyper glycosylation of the Recombinant TNFR:Fc. Furthermore the invention also relates to bioreactor operation procedure for the manufacturing of Recombinant TNFR: Fc.
This present invention relates to the rapid and efficient recovery of Recombinant TNFRrFc from cell culture supernatant from Cell culture fluid by means of Affinity chromatography. These chromatographic steps are used for capture of recombinant TNFR:Fc. This separation involves in selective binding of the desired compound to specific affinity resin and then elution with elution buffer. Culture supernatants are preferably concentrated and clarified before chromatographic treatment. TNFR:Fc containing eluent fractions are enriched with biologically active material, but they will be subjected to further processing by Anion exchange chromatographic steps. In this process the active materials are colleted in flow through. These processes are used for removal of process related impurities like host cell protein and host cell DNA. Furthermore, the invention also relates to the next chromatographic procedure where the flow-through from the previous step is further subjected to a Cation exchange chromatographic step. The active material is eluted with elution buffer containing salt. The present invention also relates to the recombinant TNFRiFc recovery procedure involving serial application different chromatographic techniques as mentioned previously. All different steps, conditions and compositions are disclosed in the invention.
Example 1:
Before seeding the Bioreactor, it was assembled and sterilized by autoclaving at 121°C for 60 minutes. After sterilization, Bioreactor was charged with 7000ml of commercially available animal component free, chemically defined media. Afterwards, the bioreactor was kept under positive pressure with air at a flow rate of 0.2 Litre per minute. The bioreactor was aerated over night for 100% air saturation. The dO2 electrode was calibrated after stabilization of dissolved oxygen value. Sterile connection was created between the seed bottle and the seed port on the bioreactor head plate. The seed was then aseptically transferred to the bioreactor using peristaltic pump. The bioreactor was seeded with the density of 0.5 x 106 cells/mL. After seeding, the bioreactor was allowed to run at following pre-set parameters:
pH: 6.9 - 7.2
dO2: 30 - 60% of air saturation
Temperature: 30 - 38°C.
Stir speed: 70-80RPM
The bioreactor was sampled at every 24/48 hours for in process quality control analysis. The bioreactor process was a fed - batch process with feeding of different nutrients at definite culture stages. During first 120 hrs of culture time, the bioreactor was daily fed with 6OmL of primary feed that comprise of glucose, lipids, amino acids, vitamins, trace elements, cholesterol and growth factors. Starting from 144 hrs of culture age the bioreactor was daily fed with 10OmL of secondary feed that comprise of Galactose and Mannose.
During first 120 hrs of culture age the bioreactor was operated at following pre-set and controlled parameters;
pH: 7.1 ±0.1
dO2: 30 - 60% of air saturation
Temperature: 36 - 37°C.
Stir speed: 70-80RPM
From 120 to 288 hrs the bioreactor was operated at following parameters;
pH: 6.9 ±0.05
dO2: 30 - 60% of air saturation
Temperature: 32 ± 0.5 0C.
Stir speed: 70-80RPM
From 288 hrs till the harvest, the bioreactor was operated at following parameters
pH: 6.9 ±0.05
dO2: 30 - 60% of air saturation
Temperature: 31 + 0.5 0C.
Stir speed: 70-80RPM
The bioreactor was harvested at a cell viability of 60 - 70%. The growth pattern, protein expression profile and nutrient consumptions are depicted (Fig 1-3).
Example 2:
Clarification of the cell culture harvest was carried out by using a cellulose disposable filter with 650 - 1000 cm2 effective filtration area and with an operating pressure of not more than 30 psi. The filtrate was checked for turbidity and target protein content. The clarified harvest was diafilitered and buffer exchanged with Tris buffer pH 7.2 - 7.6 using a 30 kDa - TFF membrane at a Trans Membrane Pressure of 5 - 10 psi. Affinity chromatography was used in binding and elution mode with column of 30 mm diameter for capturing; with Tris buffer pH 7.2 - 7.6 as equilibration buffer. After the sample is loaded on to the column, it is washed with equilibration buffer followed by 50 mM Tris-Cl, 250 mM NaClrpH 7.4 buffer solution. The protein of interest was eluted with citrate buffer (Fig 4). The eluate was hold for 45 - 60 min at acidic pH at room temperature for virus inactivation and later neutralized. The Protein A eluate fraction of Run 1 and Run 2 was pooled and buffer exchanged with Tris buffer pH 6.8 - 7.2 using a 30 kDa - TFF membrane at a Trans Membrane Pressure of 5 - 10 psi. Cation exchange chromatography in negative binding mode was carried out with column at an operational flow rate of 10 ml/min. The column was equilibrated with Tris buffer pH 6.8 - 7.2. Protein of interest is collected in flow through. This step was used for the removal of process related impurities like leachate protein A, host cell DNA and host cell protein. (Fig 5). Thereafter, the flow through was filtered for virus removal using viral removal filter having an effective filtration area of 0.01 m2. Anion exchange chromatography was carried out with the nanofiltrate after equilibrating the column with Tris buffer pH 6.8-7.2. The protein of interest was eluted with elution buffer using NaCl salt gradient. This step was used for the removal of process related impurities like host cell DNA and host cell protein (Fig 6). The eluate was buffer exchanged and concentrated using a 30 kDa TFF membrane at a Trans Membrane Pressure (TMP) of 5 - 10 psi . The buffer exchanged protein solution was filtered using 0.2μm filter. The drug substance was characterized as per the specifications. The Drug Substance (Active Pharmaceutical Ingredient) was formulated using formulation buffer containing (lOmg/mL Sucrose, 5.8mg/mL of Sodium chloride, 5.3mg/mL of L-Arginine hydrochloride, 2.6mg/mLSodium phosphate monobasic monohydrate and 0.9mg/mLsodium phosphate dibasic anhydrous) and adjusted to pH 6.3±0.2.
Example 3:
The formulated material was characterized as per the specifications set by product development specification._A 12% SDS PAGE under reducing condition was studied and showed single corresponding band at -75 kDa , matches with RMP (Fig 7). A 8% non-reducing SDS-PAGE data is mentioned in which shows a single band corresponds to approximately 150 kDa matches with RMP (Fig 8). Western blot analysis showed a clear comparable immune-specificity with that of RMP for both reducing and non-reducing condition (Fig 9). An Isoelectric focusing was carried out to identify the isoelectric point of the target protein which matched with that of RMP (Fig 10). A 2-Dimensional gel electrophoresis profile of the purified drug substance matched with RMP (Fig 11). Protein A HPLC profile carried out during this step showed a retention time of 6.3 minutes in drug substance, which was very much comparable with the RMP (6.3 minutes) (Fig 12). RP-HPLC was performed to compare the hydrophobicity profile between the purified drug substance and the RMP. The result showed comparable RT (Fig 13). Size exclusion chromatography for determination of oligomeric status showed a retention time of 6.9 minutes (% of main peak is 100) for drug substance which was very much comparable with the RMP (6.9 minutes, % area of main peak 100 [Fig 14]). Intact molecular mass estimation performed by high- sensitivity MALDI-TOF MS analysis has revealed the molecular mass of purified recombinant Etanercept (TNFR:Fc) and that of RMP to be -145 kDa (Fig 15). Peptide mass fingerprinting of iritact molecule by MALDI-TOF MS has demonstrated a high degree of similarity between drug substance with RMP (Fig 16). The results obtained in Peptide Mapping by HPLC showed a similar and corresponding profile to RMP (Fig 17). Identification of cleaved Glycans was carried out using by HPLC. The results have revealed a comparable glycosylation profile between the purified drug substance and RMP (Fig 18). UV spectral analysis has demonstrated comparable primary structure profile between the purified drug substance and RMP (Fig 19). CD spectral analysis has demonstrated comparable secondary and tertiary structure profile between the purified drug substance and RMP (Fig 20). Cytotoxicity rescue assay with L929 cell line showed the degree of TNF alpha induced cytotoxicity observed was inversely proportional to increasing amount of RMP and drug substance (Fig 21).

Claims

CLAIMS We claim:
1. A process for recovering tumor necrosis factor receptor Fc region (TNFR Fc), comprising steps of:
a) contacting culture supernatant(s) with resin(s) for selective adsorption of compound(s); b) eluting the adsorbed compound with eluant followed by enriching with biologically active material; and
c) subjecting the enriched product to Cation exchange chromatography to obtain the tumor necrosis factor receptor Fc region (TNFR Fc).
d) subjecting the enriched product to Anion exchange chromatography to obtain the tumor necrosis factor receptor Fc region (TNFR Fc).
e) subjecting the enriched product to combination of Anion and Cation exchange chromatography to obtain the tumor necrosis factor receptor Fc region (TNFR Fc).
2. The process as claimed in claim 1, wherein said supernatant is mammalian host cell culture supernatant.
3. The process as claimed in claim 1, wherein said supernatant is cell culture derived fluid.
4. The process as claimed in claim 1 , wherein said supernatant is a mammalian cell culture derived fluid.
5. The process as claimed in claim 1, wherein said culture supernatant(s) are concentrated and clarified before contacting resins.
6. The process as claimed in claim 1, wherein the process removes host cell protein and host cell DNA from culture supernatant.
" 7. A cell culture manufacturing process for the manufacturing of recombinant tumor necrosis factor receptor Fc region (TNFR Fc).
8. A process of increasing glycosylation using predefined secondary feed
9. The process of using an additional column-washing step during the purification.
PCT/IB2010/001912 2009-08-03 2010-08-03 A process of fermentation, purification and production of recombinant soluble tumour necrosis factor alfa receptor (tnfr) - human igg fc fusion protein WO2011015926A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN1839CH2009 2009-08-03
IN1839/CHE/2009 2009-08-03

Publications (1)

Publication Number Publication Date
WO2011015926A1 true WO2011015926A1 (en) 2011-02-10

Family

ID=43543981

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2010/001912 WO2011015926A1 (en) 2009-08-03 2010-08-03 A process of fermentation, purification and production of recombinant soluble tumour necrosis factor alfa receptor (tnfr) - human igg fc fusion protein

Country Status (1)

Country Link
WO (1) WO2011015926A1 (en)

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012176158A1 (en) * 2011-06-24 2012-12-27 Dr. Reddy's Laboratories Limited Purification of chimeric protein
WO2013033572A2 (en) 2011-09-01 2013-03-07 Spectramet, Llc Material sorting technology
WO2014043103A1 (en) 2012-09-11 2014-03-20 Coherus Biosciences, Inc. Correctly folded etanercept in high purity and excellent yield
US8895709B2 (en) 2008-10-20 2014-11-25 Abbvie Inc. Isolation and purification of antibodies using protein A affinity chromatography
US8906646B2 (en) 2006-09-13 2014-12-09 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
US8921526B2 (en) 2013-03-14 2014-12-30 Abbvie, Inc. Mutated anti-TNFα antibodies and methods of their use
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9062106B2 (en) 2011-04-27 2015-06-23 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9109010B2 (en) 2008-10-20 2015-08-18 Abbvie Inc. Viral inactivation during purification of antibodies cross reference to related applications
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9193787B2 (en) 2012-04-20 2015-11-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9206390B2 (en) 2012-09-02 2015-12-08 Abbvie, Inc. Methods to control protein heterogeneity
US9234033B2 (en) 2012-09-02 2016-01-12 Abbvie, Inc. Methods to control protein heterogeneity
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US9550826B2 (en) 2013-11-15 2017-01-24 Abbvie Inc. Glycoengineered binding protein compositions
JP2017505780A (en) * 2014-02-04 2017-02-23 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. Use of cation exchange chromatography in flow-through mode to enrich post-translational modifications
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US10213508B2 (en) 2011-10-18 2019-02-26 Coherus Biosciences, Inc. Etanercept formulations stabilized with xylitol
US10493151B2 (en) 2011-10-18 2019-12-03 Coherus Biosciences, Inc. Etanercept formulations stabilized with sodium chloride
US10822429B2 (en) 2012-07-09 2020-11-03 Coherus Biosciences, Inc. Etanercept formulations exhibiting marked reduction in sub-visible particles

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007117490A2 (en) * 2006-04-05 2007-10-18 Abbott Biotechnology Ltd. Antibody purification
WO2008025747A1 (en) * 2006-08-28 2008-03-06 Ares Trading S.A. Process for the purification of fc-fusion proteins

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007117490A2 (en) * 2006-04-05 2007-10-18 Abbott Biotechnology Ltd. Antibody purification
WO2008025747A1 (en) * 2006-08-28 2008-03-06 Ares Trading S.A. Process for the purification of fc-fusion proteins

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
FOLLMAN D.K. ET AL: "Factorial screening of antibody purification processes using three chromatography steps without protein A", JOURNAL OF CHROMATOGRAPHY A., vol. 1024, 2004, pages 79 - 85, XP002574059, DOI: doi:10.1016/j.chroma.2003.10.060 *

Cited By (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9073988B2 (en) 2006-09-13 2015-07-07 Abbvie Inc. Fed batch method of making anti-TNF-alpha antibodies
US9284371B2 (en) 2006-09-13 2016-03-15 Abbvie Inc. Methods of producing adalimumab
US8906646B2 (en) 2006-09-13 2014-12-09 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
US9090867B2 (en) 2006-09-13 2015-07-28 Abbvie Inc. Fed-batch method of making anti-TNF-alpha antibody
US9234032B2 (en) 2006-09-13 2016-01-12 Abbvie Inc. Fed-batch methods for producing adalimumab
US10119118B2 (en) 2006-09-13 2018-11-06 Abbvie Inc. Modified serum-free cell culture medium
US9109010B2 (en) 2008-10-20 2015-08-18 Abbvie Inc. Viral inactivation during purification of antibodies cross reference to related applications
US8895709B2 (en) 2008-10-20 2014-11-25 Abbvie Inc. Isolation and purification of antibodies using protein A affinity chromatography
US9018361B2 (en) 2008-10-20 2015-04-28 Abbvie Inc. Isolation and purification of antibodies using protein a affinity chromatography
US9090688B2 (en) 2011-04-27 2015-07-28 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9062106B2 (en) 2011-04-27 2015-06-23 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9505834B2 (en) 2011-04-27 2016-11-29 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9365645B1 (en) 2011-04-27 2016-06-14 Abbvie, Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9255143B2 (en) 2011-04-27 2016-02-09 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
WO2012176158A1 (en) * 2011-06-24 2012-12-27 Dr. Reddy's Laboratories Limited Purification of chimeric protein
WO2013033572A2 (en) 2011-09-01 2013-03-07 Spectramet, Llc Material sorting technology
US10213508B2 (en) 2011-10-18 2019-02-26 Coherus Biosciences, Inc. Etanercept formulations stabilized with xylitol
US10293049B2 (en) 2011-10-18 2019-05-21 Coherus Biosciences, Inc. Etanercept formulations stabilized with amino acids
US10493151B2 (en) 2011-10-18 2019-12-03 Coherus Biosciences, Inc. Etanercept formulations stabilized with sodium chloride
US10772963B2 (en) 2011-10-18 2020-09-15 Coherus Biosciences, Inc. Etanercept formulations stabilized with xylitol
US11000588B2 (en) 2011-10-18 2021-05-11 Coherus Biosciences, Inc. Etanercept formulations stabilized with sodium chloride
US11129876B2 (en) 2011-10-18 2021-09-28 Coherus Biosciences, Inc. Etanercept formulations stabilized with amino acids
US9346879B2 (en) 2012-04-20 2016-05-24 Abbvie Inc. Protein purification methods to reduce acidic species
US9334319B2 (en) 2012-04-20 2016-05-10 Abbvie Inc. Low acidic species compositions
US9505833B2 (en) 2012-04-20 2016-11-29 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9359434B2 (en) 2012-04-20 2016-06-07 Abbvie, Inc. Cell culture methods to reduce acidic species
US9683033B2 (en) 2012-04-20 2017-06-20 Abbvie, Inc. Cell culture methods to reduce acidic species
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9957318B2 (en) 2012-04-20 2018-05-01 Abbvie Inc. Protein purification methods to reduce acidic species
US9193787B2 (en) 2012-04-20 2015-11-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
US9708400B2 (en) 2012-04-20 2017-07-18 Abbvie, Inc. Methods to modulate lysine variant distribution
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
US10822429B2 (en) 2012-07-09 2020-11-03 Coherus Biosciences, Inc. Etanercept formulations exhibiting marked reduction in sub-visible particles
US9290568B2 (en) 2012-09-02 2016-03-22 Abbvie, Inc. Methods to control protein heterogeneity
US9206390B2 (en) 2012-09-02 2015-12-08 Abbvie, Inc. Methods to control protein heterogeneity
US9234033B2 (en) 2012-09-02 2016-01-12 Abbvie, Inc. Methods to control protein heterogeneity
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
KR102133699B1 (en) 2012-09-11 2020-07-14 코히러스 바이오사이언시즈, 인코포레이티드 Correctly folded etanercept in high purity and excellent yield
KR20150056601A (en) * 2012-09-11 2015-05-26 코히러스 바이오사이언시즈, 인코포레이티드 Correctly folded etanercept in high purity and excellent yield
WO2014043103A1 (en) 2012-09-11 2014-03-20 Coherus Biosciences, Inc. Correctly folded etanercept in high purity and excellent yield
US11001627B2 (en) 2012-09-11 2021-05-11 Coherus Biosciences, Inc. Correctly folded etanercept in high purity and excellent yield
US10954295B2 (en) 2012-09-11 2021-03-23 Coherus Biosciences, Inc. Correctly folded etanercept in high purity and excellent yield
US10954294B2 (en) 2012-09-11 2021-03-23 Coherus Biosciences, Inc. Correctly folded etanercept in high purity and excellent yield
US10954293B2 (en) 2012-09-11 2021-03-23 Coherus Biosciences, Inc. Correctly folded etanercept in high purity and excellent yield
US10947306B2 (en) 2012-09-11 2021-03-16 Coherus Biosciences, Inc. Correctly folded etanercept in high purity and excellent yield
EP2895188A4 (en) * 2012-09-11 2016-05-25 Coherus Biosciences Inc Correctly folded etanercept in high purity and excellent yield
AU2018247244B2 (en) * 2012-09-11 2020-05-28 Coherus Biosciences, Inc. Correctly folded etanercept in high purity and excellent yield
US8921526B2 (en) 2013-03-14 2014-12-30 Abbvie, Inc. Mutated anti-TNFα antibodies and methods of their use
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US9708399B2 (en) 2013-03-14 2017-07-18 Abbvie, Inc. Protein purification using displacement chromatography
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9499616B2 (en) 2013-10-18 2016-11-22 Abbvie Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9266949B2 (en) 2013-10-18 2016-02-23 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9200070B2 (en) 2013-10-18 2015-12-01 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9200069B2 (en) 2013-10-18 2015-12-01 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9315574B2 (en) 2013-10-18 2016-04-19 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9688752B2 (en) 2013-10-18 2017-06-27 Abbvie Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9522953B2 (en) 2013-10-18 2016-12-20 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9550826B2 (en) 2013-11-15 2017-01-24 Abbvie Inc. Glycoengineered binding protein compositions
JP2017505780A (en) * 2014-02-04 2017-02-23 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. Use of cation exchange chromatography in flow-through mode to enrich post-translational modifications
KR20220044391A (en) * 2014-02-04 2022-04-07 바이오젠 엠에이 인코포레이티드 Use of cation-exchange chromatography in the flow-through mode to enrich post-translational modifications
KR102567586B1 (en) * 2014-02-04 2023-08-16 바이오젠 엠에이 인코포레이티드 Use of cation-exchange chromatography in the flow-through mode to enrich post-translational modifications

Similar Documents

Publication Publication Date Title
WO2011015926A1 (en) A process of fermentation, purification and production of recombinant soluble tumour necrosis factor alfa receptor (tnfr) - human igg fc fusion protein
US9193761B2 (en) Method for purifying human granulocyte-colony stimulating factor from recombinant E. coli
US20180194801A1 (en) Method for isolating and purifying recombinant human serum albumin from transgenic rice grain
CN106536565A (en) Process for the purification of TNFR:Fc fusion protein
US9815879B2 (en) Method for the purification of G-CSF
WO2011024025A1 (en) An erythropoietin analogue and a method thereof
RU2614119C9 (en) Method of producing human immunoglobulin
EP3500586B1 (en) Continuous process for reducing heterogeneity of therapeutic protein
CN102911250B (en) Method for purifying acidic recombinant protein medicament
CN101260145B (en) Technique for separating and purifying recombination human serum albumin and fusion protein thereof
JPH11507033A (en) Method for purifying thrombopoietin
JP2017526649A (en) New process for purification of rHu-GCSF
WO2011024024A1 (en) A process for recovering darbepoeitin alfa isoforms
WO2014102814A1 (en) Process for the purification of fc fusion proteins
CN110724204B (en) Method for purifying Fc fusion protein
WO2011015919A1 (en) A highly efficient process of purification and production of recombinant infliximab
WO2018178375A1 (en) Protein purification method and kit
KR101847169B1 (en) Composition comprising long-acting Erythropoietin
WO2011015921A1 (en) A highly efficient process of purification and production of recombinant cetuximab
WO2011015920A2 (en) A highly efficient process of purification and production of recombinant trastuzumab
WO2023024214A1 (en) Method for purifying fusion protein of recombinant human serum albumin
CN116715756A (en) Antibody purification method
KR20180026688A (en) Composition comprising long-acting Erythropoietin
TW201723174A (en) Method for manufacturing recombinant proteins
KR20070091081A (en) Method for purifying human granulocyte microphage colony stimulating factor from the suspension culture broth of oryza sativa

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: 10806112

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: 10806112

Country of ref document: EP

Kind code of ref document: A1