WO2011090719A2 - Protein purification by ion exchange - Google Patents

Protein purification by ion exchange Download PDF

Info

Publication number
WO2011090719A2
WO2011090719A2 PCT/US2010/062245 US2010062245W WO2011090719A2 WO 2011090719 A2 WO2011090719 A2 WO 2011090719A2 US 2010062245 W US2010062245 W US 2010062245W WO 2011090719 A2 WO2011090719 A2 WO 2011090719A2
Authority
WO
WIPO (PCT)
Prior art keywords
protein
chromatography
process according
value
antibody
Prior art date
Application number
PCT/US2010/062245
Other languages
French (fr)
Other versions
WO2011090719A3 (en
Inventor
Samir Kulkarni
Satyam Subrahmanyam
Original Assignee
Dr. Reddy's Laboratories Ltd.
Dr. Reddy's Laboratories, Inc.
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 Dr. Reddy's Laboratories Ltd., Dr. Reddy's Laboratories, Inc. filed Critical Dr. Reddy's Laboratories Ltd.
Priority to US13/518,701 priority Critical patent/US20130178608A1/en
Priority to EP10844264.1A priority patent/EP2519536A4/en
Publication of WO2011090719A2 publication Critical patent/WO2011090719A2/en
Publication of WO2011090719A3 publication Critical patent/WO2011090719A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/36Extraction; Separation; Purification by a combination of two or more processes of different types
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • C07K1/18Ion-exchange chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • C07K1/22Affinity chromatography or related techniques based upon selective absorption processes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/06Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies from serum
    • C07K16/065Purification, fragmentation

Definitions

  • aspects of the application relate to methods of purifying antibodies using chromatographic methods.
  • the application describes antibody purification methods comprising multiple chromatographic steps, wherein a low pH eluate from a protein A chromatography is further purified without a requirement for substantial pH adjustment.
  • Therapeutic proteins are primarily produced by recombinant DNA technology, i.e., by cloning and expression of a heterologus gene in prokaryotic or eukaryotic systems. However, proteins expressed by recombinant DNA methods are typically associated with
  • HCP host cell proteins
  • HCD host cell DNA
  • viruses etc.
  • microheterogeneity in the expression of the desired protein in the form of charged variants (typically acidic, lower pi variants and basic, higher pi variants).
  • charged variants typically acidic, lower pi variants and basic, higher pi variants.
  • FDA United States Food and Drug administration
  • biopharmaceuticals be free from impurities, both product related (aggregates or degradation products) and process related (media components, HCP, DNA, chromatographic media used in purification, endotoxins, viruses, etc). See, Office of Biologies Research and Review, Food and Drug Administration, Points to consider in the production and testing of new drugs and biologicals produced by
  • Protein purification is usually a multistep process, wherein different chromatographic steps are run sequentially to yield a final purified product.
  • protein A chromatography is one of the more widely used methods, and usually is the first step in antibody purification. This is a type of affinity chromatography wherein separation is affected by means of a resin tagged with protein A (Hjelm H. et.al., FEBS lett. 1972; 28, 73-76; Langone JJ., Adv Immunol, 1982; 32, 157-252).
  • the various aspects of Protein A chromatography are described in U.S. Patent Nos. 6,013,763 and 6,399,750, and European Patent Application Publication Nos. 282308 and 284368.
  • a disadvantage of protein A chromatography is the leaching of Protein A and its fragments from the chromatographic resin and its contamination of the eluate. Since protein A is of bacterial origin (obtained from Staphylococcus aureus), it's removal is necessary to avoid undesirable immune responses. It has been shown that IgG can form complexes with protein A that may activate Fc bearing leukocytes and complement system to generate oxidant and anaphylatoxin activity in vitro (Balint J. et.al., Cancer Res. 1984; 44, 734-743). Further, protein A has also been linked with toxicity (Bensinger, W.I. et. al., J. Biol Resp. Modif. 1984; 3, 347;
  • 2007/0292442 describe the use of ion exchange chromatography for purification of antibodies.
  • these methods either result in considerable loss of antibody or require substantial pH adjustment of the sample prior to a subsequent chromatography step.
  • This change in pH is achieved by addition of a high molarity base that compromises process efficiency as a result of volume dilution and mixing efficiency, as well as product stability due to localized pH surge.
  • the impact on product stability is of particular significance as it leads to significant product loss due to denaturation, precipitation, and aggregation. There remains a need for efficient multi-step separation processes for antibodies.
  • the application describes antibody purification methods, comprising multiple chromatographic steps wherein a low pH eluate from a protein A chromatography is further purified without the need of substantial pH adjustment.
  • Fig. 1 is an llustration of a chromatogram from the procedure of Example 1 .
  • Fig. 2 is an llustration of a chromatogram from the procedure of Example 2.
  • Fig. 3 is an llustration of a chromatogram from the procedure of Example 5.
  • Fig. 4 is an llustration of a chromatogram from the procedure of Example 3.
  • Fig. 5 is an llustration of a chromatogram from the procedure of Example 5.
  • Fig. 6 is an llustration of a chromatogram from the procedure of Example 5.
  • flow-through mode refers to chromatographic methods wherein a desired protein is obtained in the flow-through liquid during loading or post-loading washing of a chromatography column.
  • a desired protein in the flow-through liquid may be collected as various fractions and pooled together or can be collected as a single fraction.
  • binding mode refers chromatographic methods wherein a desired protein is bound to the chromatography resin when loaded onto a column and is eluted subsequently using an elution buffer.
  • the desired protein is collected in an elution liquid and may be collected as a single fraction or as various fractions that are pooled together.
  • antibody refers to an immunoglobulin that is composed of four polypeptide chains consisting of two light and two heavy chains, as well as immunoglobulins isolated from various sources, such as murine, human, recombinant, etc., truncated antibodies, chimeric, humanized, or pegylated antibody isotypes, allotypes, and alleles of immunoglobulin genes.
  • the term antibody also refers to fusion proteins that contain an immunoglobulin moiety.
  • MAbs monoclonal antibodies
  • protein A chromatography is a commonly used method, as highly purified MAbs can be obtained due to the high specificity and binding between a protein A ligand and a Fc region of the antibody.
  • a disadvantage of protein A chromatography is the leaching of protein A and its fragments into the eluate. Hence, further purification steps are required for the removal of protein A and/or its fragments, as well as residual host cell proteins, endotoxins, and host cell DNA.
  • U.S. Patent No. 4,983,722 and European Patent Application Publication No. 1601697 describe the use of anion exchange chromatography for the purification of antibodies.
  • the anion exchange step is performed at neutral to alkaline pH values, necessitating substantial pH adjustment of the antibody sample.
  • the protein A eluate is diafiltered against a DEAE equilibration buffer at pH 8.6
  • the acidic protein A eluate is neutralized with a high molarity buffer, such as 0.5M TrisHCI pH 7.5, and diafiltered with binding/equilibration buffer at pH 8.0 prior to the next chromatographic step.
  • 2007/0292442 describes the use of two ion exchange resins for the purification of antibodies, wherein the pH of the first eluate is adjusted before loading onto the second ion exchange resin.
  • the pH adjustment greatly compromises both the process efficiency and the product stability. Hence a process involving no, or minimal, pH adjustment will be a better alternative to the current methods.
  • the present application describes processes that reduce the need of pH adjustment in a multistep purification process, thereby increasing process efficiency and product stability, in addition to achieving effective separation of acidic and basic variants of monoclonal antibodies.
  • the application provides methods for purifying antibodies, embodiments comprising:
  • a second purification step using cation exchange chromatography that is performed in the bind-elute mode, wherein eluate obtained from step 1 ) is loaded onto a cation exchange resin without substantial adjustment of pH (viz. within a range of ⁇ 0.2 pH values).
  • an antibody is eluted in purification step 1 ) at pH values about 3.5 to 6 and loaded onto a cation exchange resin at pH values about 3.5 to 6.
  • an antibody is eluted in purification step 1 ) at pH values about 3.5 and loaded onto a cation exchange resin at pH values about 3.5.
  • the application provides methods for purifying antibodies, embodiments comprising:
  • the first, second, and third pH values may be same or different.
  • the first and second pH values may be similar.
  • the first and second pH values may be different.
  • the protein A chromatographic resin used may be any protein A or variant or a functional fragment thereof coupled to any chromatographic support.
  • the protein A resin is Prosep vA Ultra ® (from Millipore), wherein animal-free protein A is immobilized on porous glass.
  • fresh (i.e., not previously used) protein A chromatographic resin may be used to obtain a feed stream for a second chromatographic step. After washing with loading buffer and intermediate wash, the elution is carried out at low pH such as from pH 2.5 to about pH 4.5
  • Cation exchange chromatographic step mentioned in the embodiments may be carried out using any weak or strong cation exchange chromatographic resin or a membrane which could function as a weak or a strong cation exchanger.
  • cation exchange resins include, but are not limited to, those having a sulfonate based group e.g., MonoS, MiniS, Source 15S and 30S, SP Sepharose Fast Flow, SP Sepharose High Performance from GE Healthcare, Toyopearl SP-650S and SP-650M from Tosoh, S-Ceramic Hyper D, from Pall Corporation or a carboxymethyl based group e.g., CM Sepharose Fast Flow from GE Healthcare, Macro-Prep CM from BioRad, CM-Ceramic Hyper D, from Pall Corporation, Toyopearl CM-650S, CM-650M and CM-650C from Tosoh.
  • a weak cation exchange resin such as CM Ceramic Hyper D F ® (Pall Corporation) is used; this is made using rigid porous beads that are coated with functionalized hydrogel.
  • Anion exchange chromatography mentioned in the embodiments may be carried out using any weak or strong anion exchange chromatographic resin or a membrane which could function as a weak or a strong anion exchanger.
  • anion exchange resins include, but are not limited to, DEAE cellulose, Poros PI 20, PI 50, HQ 10, HQ 20, HQ 50, D 50 from Applied Materials
  • a strong anion exchange resin such as Q- Sepharose Fast Flow ® (GE Healthcare Life Sciences) is used. This resin is made using a highly cross-linked, 6 % agarose matrix attached to -O- CH 2 CHOHCH2OCH2CHOHCH 2 N + (CH3)3 functional group.
  • buffering agents used in the buffer solutions include, but are not limited to, TRIS, phosphate, citrate, and acetate salts, or derivatives thereof.
  • Protein A leachates can be analysed using protein A ELISA and purified antibodies can be analysed using protein A high performance liquid
  • An anti-VEGF antibody was cloned and expressed in a CHO cell line as described in U.S. Patent No. 7,060,269, which is incorporated herein by reference.
  • the cell culture broth containing the expressed antibody was harvested, clarified and subjected to protein A affinity chromatography as described below.
  • the clarified cell culture broth was loaded onto a protein A chromatography column (Prosep vA ultra, VL44x250, 205 ml_) that was pre-equilibrated with 5 column volumes ("CV") of 50 mM Tris, 150 mM NaCI, pH 7.5 buffer.
  • the column was then washed with 5 CV of the equilibration buffer (50 mM Tris, 150 mM NaCI, pH 7.5). This was followed by a wash with 5 CV of 50 mM Tris, 750 mM NaCI, pH
  • Figure 1 is an illustration of a chromatogram from the procedure as described in this example.
  • the line marked “Cond” represents the increase in conductivity in mS/cm.
  • Peak A represents the eluate obtained from protein A chromatography resin.
  • Example 2 The eluate obtained from the protein A chromatography procedure described in Example 1 was loaded onto a cation exchange resin (CM Ceramic Hyper D F, VL44x250, 304 ml_) pre-equilibrated with 10 CV of equilibration buffer (200 mM acetate buffer, pH 3.5). This was followed by washing with 30 CV of wash buffer (35 mM Phosphate buffer pH 6.0). The bound antibody was eluted using a conductivity gradient (2.5 mS/cm to- 7 mS/cm) with a phosphate buffer (35 mM to 80 mM, pH 6.0).
  • a conductivity gradient 2.5 mS/cm to- 7 mS/cm
  • a phosphate buffer 35 mM to 80 mM, pH 6.0.
  • Figure 2 is an illustration of a chromatogram from the procedure as described in this example.
  • the line marked “Cond” represents the increase in conductivity in mS/cm.
  • Buffer Cone represents the concentration of phosphate buffer during the chromatographic run, where 100% corresponds to a buffer concentration of 80 mM.
  • Peak A represents the acidic variant of the antibody, and Peak B represents the more basic (desired) variant of the antibody.
  • the eluate obtained from the cation exchange chromatography procedure described in Example 2 was loaded onto an anion exchange resin (Q-Sepharose FF, VL32x250, 80 mL) pre-equilibrated with 5-20 CV of an equilibration buffer (67 mM phosphate buffer, pH 6). This was followed by a post load wash with 5 CV of equilibration buffer and the load and wash flow-through was collected.
  • an anion exchange resin Q-Sepharose FF, VL32x250, 80 mL
  • Figure 4 is an illustration of a chromatogram from the procedure described in this example.
  • the line marked “Cond” represents the increase in conductivity in mS/cm.
  • “FT” represents the flow-through obtained. Impurity levels in the cation exchange chromatography eluate and anion exchange chromatography flow through are described in Table 1 .
  • Clarified cell culture broth from Example 1 was loaded onto a protein A chromatography resin (Prosep vA ultra, VL44x250, 205 ml_) that was pre- equilibrated with 5 CV of a 50 mM Tris, 150 mM NaCI, pH 7.5 buffer.
  • the column was then washed with 5 CV of an equilibration buffer (50 mM Tris, 150 mM NaCI, pH 7.5). This was followed by a wash with 5 CV of a 50 mM Tris, 750 mM NaCI, pH 7.5 buffer and a final wash with 25 mM Tris at pH 7.5.
  • the bound antibody was eluted using the low pH buffer 200 mM acetate buffer, pH 3.5.
  • CM- Ceramic Hyper D F, VL44x250, 304 mL a cation exchange resin
  • equilibration buffer 35 mM phosphate buffer pH 6.0. This was followed by washing with 10 CV of equilibration buffer. The bound antibody was eluted using a
  • Figure 3 is an illustration of a chromatogram from the procedure described in this example.
  • the line marked “Cond” represents the increase in conductivity in mS/cm.
  • Buffer Cone represents the concentration of phosphate buffer during the chromatographic run, where 100% corresponds to a buffer concentration of 80 mM. Peak A represents the eluate obtained from the linear increase in conductivity.
  • Figure 5 is an illustration of a chromatogram from the procedure described in this example.
  • the line marked “Cond” represents the increase in conductivity in mS/cnn.
  • Peak A represents the eluate obtained from the step increase in
  • Figure 6 is an illustration of a chromatogram from the procedure described in this example.
  • the line marked “Cond” represents the increase in conductivity in mS/cm.
  • Peak A represents the eluate obtained from a step increase in conductivity from 35 mM buffer to 90 mM buffer.
  • Example 5 was loaded onto an anion exchange resin (Q-Sepharose FF, VL32x250, 80 ml_) pre-equilibrated with 5-20 CV of an equilibration buffer (67 mM phosphate buffer, pH 6). This was followed by a post load wash with 5 CV of equilibration buffer and the load and wash flow-through collected. Impurities levels in the cation exchange chromatography eluate and anion exchange chromatography flow- through are described in Table 2.
  • an anion exchange resin Q-Sepharose FF, VL32x250, 80 ml_

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • Immunology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The application describes an antibody purification method comprising multiple chromatographic steps wherein the low pH eluate from a protein A chromatography is further purified without the need of substantial pH adjustment.

Description

P ROTE IN PURIFICATION BY ION EXCHANGE
INTRODUCTION
Aspects of the application relate to methods of purifying antibodies using chromatographic methods. In embodiments, the application describes antibody purification methods comprising multiple chromatographic steps, wherein a low pH eluate from a protein A chromatography is further purified without a requirement for substantial pH adjustment.
Large scale purification of proteins remains a significant challenge in the biopharmaceutical industry as efficient and cost-effective methods are required to achieve desired yields and purity levels. Therapeutic proteins are primarily produced by recombinant DNA technology, i.e., by cloning and expression of a heterologus gene in prokaryotic or eukaryotic systems. However, proteins expressed by recombinant DNA methods are typically associated with
contaminants such as host cell proteins ("HCP"), host cell DNA ("HCD"), viruses, etc. In addition, there is significant microheterogeneity in the expression of the desired protein, in the form of charged variants (typically acidic, lower pi variants and basic, higher pi variants).The presence of these contaminants, including undesirable charged variants, are a potential health risk, and hence their removal from a final product is a regulatory requirement. Thus, drug regulatory agencies such as United States Food and Drug administration ("FDA") require that
biopharmaceuticals be free from impurities, both product related (aggregates or degradation products) and process related (media components, HCP, DNA, chromatographic media used in purification, endotoxins, viruses, etc). See, Office of Biologies Research and Review, Food and Drug Administration, Points to consider in the production and testing of new drugs and biologicals produced by
recombinant DNA technology (Draft), 1985. Thus, elimination of impurities and contaminants from a final product is mandatory and poses a significant challenge in the development of methods for the purification of proteins.
Protein purification is usually a multistep process, wherein different chromatographic steps are run sequentially to yield a final purified product. For purification of monoclonal antibodies, protein A chromatography is one of the more widely used methods, and usually is the first step in antibody purification. This is a type of affinity chromatography wherein separation is affected by means of a resin tagged with protein A (Hjelm H. et.al., FEBS lett. 1972; 28, 73-76; Langone JJ., Adv Immunol, 1982; 32, 157-252). The various aspects of Protein A chromatography (protein A and its variants, chromatographic medium, etc.) are described in U.S. Patent Nos. 6,013,763 and 6,399,750, and European Patent Application Publication Nos. 282308 and 284368.
A disadvantage of protein A chromatography is the leaching of Protein A and its fragments from the chromatographic resin and its contamination of the eluate. Since protein A is of bacterial origin (obtained from Staphylococcus aureus), it's removal is necessary to avoid undesirable immune responses. It has been shown that IgG can form complexes with protein A that may activate Fc bearing leukocytes and complement system to generate oxidant and anaphylatoxin activity in vitro (Balint J. et.al., Cancer Res. 1984; 44, 734-743). Further, protein A has also been linked with toxicity (Bensinger, W.I. et. al., J. Biol Resp. Modif. 1984; 3, 347;
Messeschimdt GL. et. al., J. Biol. Resp. Modif. 1984; 3, 325; Terman D.S. and Bertram, J.H., Eur. J. Cancer Clin. Oncol. 1985; 21 , 1 1 15 and Ventura G.J et. al., Cancer Treat. Rep. 1987; 71 , 41 1 ). Thus, subsequent purification steps are required to remove protein A leachates, as well as residual host cell proteins, host cell DNA, etc. to meet regulatory requirements.
The literature discloses various methods for purification of crude or partially purified samples. Balint et. al. describe the use of gel filtration for separating uncomplexed antibodies from IgG-protein A complexes (Balint et.al., Cancer Res 1984; 44, 734-743J. U.S. Patent No. 4,983,722, European Patent Application Publication No. 1601697 and U.S. Patent Application Publication No.
2007/0292442 describe the use of ion exchange chromatography for purification of antibodies. However, these methods either result in considerable loss of antibody or require substantial pH adjustment of the sample prior to a subsequent chromatography step. This change in pH is achieved by addition of a high molarity base that compromises process efficiency as a result of volume dilution and mixing efficiency, as well as product stability due to localized pH surge. The impact on product stability is of particular significance as it leads to significant product loss due to denaturation, precipitation, and aggregation. There remains a need for efficient multi-step separation processes for antibodies.
SUMMARY
In aspects, the application describes antibody purification methods, comprising multiple chromatographic steps wherein a low pH eluate from a protein A chromatography is further purified without the need of substantial pH adjustment.
BRIEF DESCRIPTION OF THE DRAWINGS
Fig. 1 is an llustration of a chromatogram from the procedure of Example 1 . Fig. 2 is an llustration of a chromatogram from the procedure of Example 2. Fig. 3 is an llustration of a chromatogram from the procedure of Example 5. Fig. 4 is an llustration of a chromatogram from the procedure of Example 3. Fig. 5 is an llustration of a chromatogram from the procedure of Example 5. Fig. 6 is an llustration of a chromatogram from the procedure of Example 5.
DETAILED DESCRIPTION
The term "flow-through mode" as used herein refers to chromatographic methods wherein a desired protein is obtained in the flow-through liquid during loading or post-loading washing of a chromatography column. A desired protein in the flow-through liquid may be collected as various fractions and pooled together or can be collected as a single fraction.
The term "bind-elute mode" as used herein refers chromatographic methods wherein a desired protein is bound to the chromatography resin when loaded onto a column and is eluted subsequently using an elution buffer. The desired protein is collected in an elution liquid and may be collected as a single fraction or as various fractions that are pooled together.
The term "antibody" as used herein refers to an immunoglobulin that is composed of four polypeptide chains consisting of two light and two heavy chains, as well as immunoglobulins isolated from various sources, such as murine, human, recombinant, etc., truncated antibodies, chimeric, humanized, or pegylated antibody isotypes, allotypes, and alleles of immunoglobulin genes. The term antibody also refers to fusion proteins that contain an immunoglobulin moiety. ln the purification of monoclonal antibodies ("MAbs"), protein A chromatography is a commonly used method, as highly purified MAbs can be obtained due to the high specificity and binding between a protein A ligand and a Fc region of the antibody. However, as discussed earlier, a disadvantage of protein A chromatography is the leaching of protein A and its fragments into the eluate. Hence, further purification steps are required for the removal of protein A and/or its fragments, as well as residual host cell proteins, endotoxins, and host cell DNA.
U.S. Patent No. 4,983,722 and European Patent Application Publication No. 1601697 describe the use of anion exchange chromatography for the purification of antibodies. However, the anion exchange step is performed at neutral to alkaline pH values, necessitating substantial pH adjustment of the antibody sample. For instance, in US 4,983,722 the protein A eluate is diafiltered against a DEAE equilibration buffer at pH 8.6, while in EP 1601697 the acidic protein A eluate is neutralized with a high molarity buffer, such as 0.5M TrisHCI pH 7.5, and diafiltered with binding/equilibration buffer at pH 8.0 prior to the next chromatographic step. Likewise, U.S. Patent Application Publication No. 2007/0292442 describes the use of two ion exchange resins for the purification of antibodies, wherein the pH of the first eluate is adjusted before loading onto the second ion exchange resin. The pH adjustment greatly compromises both the process efficiency and the product stability. Hence a process involving no, or minimal, pH adjustment will be a better alternative to the current methods.
In aspects, the present application describes processes that reduce the need of pH adjustment in a multistep purification process, thereby increasing process efficiency and product stability, in addition to achieving effective separation of acidic and basic variants of monoclonal antibodies.
In an aspect, the application provides methods for purifying antibodies, embodiments comprising:
1 ) A first purification step using protein A chromatography, wherein an antibody is eluted at particular pH values.
2) A second purification step using cation exchange chromatography that is performed in the bind-elute mode, wherein eluate obtained from step 1 ) is loaded onto a cation exchange resin without substantial adjustment of pH (viz. within a range of ± 0.2 pH values). In embodiments, an antibody is eluted in purification step 1 ) at pH values about 3.5 to 6 and loaded onto a cation exchange resin at pH values about 3.5 to 6.
In embodiments, an antibody is eluted in purification step 1 ) at pH values about 3.5 and loaded onto a cation exchange resin at pH values about 3.5.
In an aspect, the application provides methods for purifying antibodies, embodiments comprising:
1 ) A first purification step using protein A chromatography, wherein an antibody is eluted at a first pH.
2) A second purification step using cation exchange chromatography performed in the bind-elute mode, wherein eluate obtained from step 1 ) is loaded onto a cation exchange resin at a second pH, and the elution is carried out at a third pH.
3) A third purification step using anion exchange chromatography performed in the flow-through mode, wherein eluate from step 2) is loaded onto an anion exchange resin without substantial adjustment of pH (viz. within a range of ± 0.2 pH values).
In embodiments, the first, second, and third pH values may be same or different.
In embodiments, the first and second pH values may be similar.
In embodiments, the first and second pH values may be different.
The protein A chromatographic resin used may be any protein A or variant or a functional fragment thereof coupled to any chromatographic support. In
embodiments, the protein A resin is Prosep vA Ultra® (from Millipore), wherein animal-free protein A is immobilized on porous glass. In embodiments, fresh (i.e., not previously used) protein A chromatographic resin may be used to obtain a feed stream for a second chromatographic step. After washing with loading buffer and intermediate wash, the elution is carried out at low pH such as from pH 2.5 to about pH 4.5
Cation exchange chromatographic step mentioned in the embodiments may be carried out using any weak or strong cation exchange chromatographic resin or a membrane which could function as a weak or a strong cation exchanger.
Commercially available cation exchange resins include, but are not limited to, those having a sulfonate based group e.g., MonoS, MiniS, Source 15S and 30S, SP Sepharose Fast Flow, SP Sepharose High Performance from GE Healthcare, Toyopearl SP-650S and SP-650M from Tosoh, S-Ceramic Hyper D, from Pall Corporation or a carboxymethyl based group e.g., CM Sepharose Fast Flow from GE Healthcare, Macro-Prep CM from BioRad, CM-Ceramic Hyper D, from Pall Corporation, Toyopearl CM-650S, CM-650M and CM-650C from Tosoh. In embodiments of the invention, a weak cation exchange resin, such as CM Ceramic Hyper D F® (Pall Corporation) is used; this is made using rigid porous beads that are coated with functionalized hydrogel.
Anion exchange chromatography mentioned in the embodiments may be carried out using any weak or strong anion exchange chromatographic resin or a membrane which could function as a weak or a strong anion exchanger.
Commercially available anion exchange resins include, but are not limited to, DEAE cellulose, Poros PI 20, PI 50, HQ 10, HQ 20, HQ 50, D 50 from Applied
Biosystems, MonoQ, MiniQ, Source 15Q and 3OQ, Q, DEAE and ANX Sepharose Fast Flow, Q Sepharose high Performance, QAE SEPHADEX and FAST Q
SEPHAROSE from GE Healthcare, Macro-Prep DEAE and Macro-Prep High Q from Biorad, Q-Ceramic Hyper D, DEAE-Ceramic Hyper D, from Pall Corporation. In embodiments of the invention, a strong anion exchange resin, such as Q- Sepharose Fast Flow® (GE Healthcare Life Sciences) is used. This resin is made using a highly cross-linked, 6 % agarose matrix attached to -O- CH2CHOHCH2OCH2CHOHCH2N+(CH3)3 functional group.
Examples of buffering agents used in the buffer solutions include, but are not limited to, TRIS, phosphate, citrate, and acetate salts, or derivatives thereof.
Protein A leachates can be analysed using protein A ELISA and purified antibodies can be analysed using protein A high performance liquid
chromatography.
Certain specific aspects and embodiments of the application are more fully described by reference to the following examples, being provided only for purposes of illustration. These examples should not be construed as limiting the scope of the application in any manner. EXAMPLE 1
Protein A chromatography
An anti-VEGF antibody was cloned and expressed in a CHO cell line as described in U.S. Patent No. 7,060,269, which is incorporated herein by reference. The cell culture broth containing the expressed antibody was harvested, clarified and subjected to protein A affinity chromatography as described below.
The clarified cell culture broth was loaded onto a protein A chromatography column (Prosep vA ultra, VL44x250, 205 ml_) that was pre-equilibrated with 5 column volumes ("CV") of 50 mM Tris, 150 mM NaCI, pH 7.5 buffer. The column was then washed with 5 CV of the equilibration buffer (50 mM Tris, 150 mM NaCI, pH 7.5). This was followed by a wash with 5 CV of 50 mM Tris, 750 mM NaCI, pH
7.5 buffer and a final wash with 25 mM Tris at pH 7.5. The bound antibody was eluted using the low pH buffer 200 mM acetate buffer, pH 3.5.
Figure 1 is an illustration of a chromatogram from the procedure as described in this example. The line marked "Cond" represents the increase in conductivity in mS/cm. Peak A, represents the eluate obtained from protein A chromatography resin.
EXAMPLE 2
Cation exchange chromatography
The eluate obtained from the protein A chromatography procedure described in Example 1 was loaded onto a cation exchange resin (CM Ceramic Hyper D F, VL44x250, 304 ml_) pre-equilibrated with 10 CV of equilibration buffer (200 mM acetate buffer, pH 3.5). This was followed by washing with 30 CV of wash buffer (35 mM Phosphate buffer pH 6.0). The bound antibody was eluted using a conductivity gradient (2.5 mS/cm to- 7 mS/cm) with a phosphate buffer (35 mM to 80 mM, pH 6.0).
Figure 2 is an illustration of a chromatogram from the procedure as described in this example. The line marked "Cond" represents the increase in conductivity in mS/cm. "Buffer Cone." represents the concentration of phosphate buffer during the chromatographic run, where 100% corresponds to a buffer concentration of 80 mM. Peak A represents the acidic variant of the antibody, and Peak B represents the more basic (desired) variant of the antibody. The
consistency of elution (Peak B) in multiple runs is shown.
EXAMPLE 3
Anion exchange chromatography
The eluate obtained from the cation exchange chromatography procedure described in Example 2 was loaded onto an anion exchange resin (Q-Sepharose FF, VL32x250, 80 mL) pre-equilibrated with 5-20 CV of an equilibration buffer (67 mM phosphate buffer, pH 6). This was followed by a post load wash with 5 CV of equilibration buffer and the load and wash flow-through was collected.
Figure 4 is an illustration of a chromatogram from the procedure described in this example. The line marked "Cond" represents the increase in conductivity in mS/cm. "FT" represents the flow-through obtained. Impurity levels in the cation exchange chromatography eluate and anion exchange chromatography flow through are described in Table 1 .
Table 1
Impurity Concentration
Step
Host Cell Host Cell Protein A Monomer
Sample Recovery
Proteins DNA leachate (%)
(%)
(ppm) (ng/mL) (ppm)
Clarified cell
50170 296 — 100
culture broth
Protein A eluate
5.0 ND 23.3 97
(eluate pH 3.5)
Cation
Exchange
chromatography BDL < 0.19 BDL 53 99.8 eluate (load pH
3.5)
Anion
Exchange
BDL < 0.19 BDL 97
chromatography
flow through
Figure imgf000011_0001
BDL: Below detection limit,
ppm: parts per million.
ND: not determined. EXAMPLE 4
Protein A chromatography
Clarified cell culture broth from Example 1 was loaded onto a protein A chromatography resin (Prosep vA ultra, VL44x250, 205 ml_) that was pre- equilibrated with 5 CV of a 50 mM Tris, 150 mM NaCI, pH 7.5 buffer. The column was then washed with 5 CV of an equilibration buffer (50 mM Tris, 150 mM NaCI, pH 7.5). This was followed by a wash with 5 CV of a 50 mM Tris, 750 mM NaCI, pH 7.5 buffer and a final wash with 25 mM Tris at pH 7.5. The bound antibody was eluted using the low pH buffer 200 mM acetate buffer, pH 3.5. EXAMPLE 5
Cation exchange chromatography
The eluate obtained from protein A chromatography step described in Example 4 was adjusted to pH 6.0 and loaded onto a cation exchange resin (CM- Ceramic Hyper D F, VL44x250, 304 mL) pre-equilibrated with 10 CV of
equilibration buffer (35 mM phosphate buffer pH 6.0). This was followed by washing with 10 CV of equilibration buffer. The bound antibody was eluted using a
conductivity gradient (2.5 mS/cm to 7 mS/cm) with a phosphate buffer (35 mM to 80 mM, pH 6.0). Alternatively, elution was done using a step-gradient with less than 3 to 5 CV of phosphate buffer, pH 6.2 wherein the concentration of the buffer was increased in steps from 35 mM to 80 mM, or from 35 mM to 90 mM.
Figure 3 is an illustration of a chromatogram from the procedure described in this example. The line marked "Cond" represents the increase in conductivity in mS/cm. "Buffer Cone." represents the concentration of phosphate buffer during the chromatographic run, where 100% corresponds to a buffer concentration of 80 mM. Peak A represents the eluate obtained from the linear increase in conductivity.
Figure 5 is an illustration of a chromatogram from the procedure described in this example. The line marked "Cond" represents the increase in conductivity in mS/cnn. Peak A represents the eluate obtained from the step increase in
conductivity from 35 mM buffer to 80 mM buffer.
Figure 6 is an illustration of a chromatogram from the procedure described in this example. The line marked "Cond" represents the increase in conductivity in mS/cm. Peak A represents the eluate obtained from a step increase in conductivity from 35 mM buffer to 90 mM buffer.
EXAMPLE 6
Anion exchange chromatography
The eluate obtained from cation exchange chromatography described in
Example 5 was loaded onto an anion exchange resin (Q-Sepharose FF, VL32x250, 80 ml_) pre-equilibrated with 5-20 CV of an equilibration buffer (67 mM phosphate buffer, pH 6). This was followed by a post load wash with 5 CV of equilibration buffer and the load and wash flow-through collected. Impurities levels in the cation exchange chromatography eluate and anion exchange chromatography flow- through are described in Table 2.
Table 2
Figure imgf000012_0001
BDL: Below detection limit,
ppm: parts per million.

Claims

CLAIMS:
1 . A process for purifying antibodies comprising:
a) purifying using protein A chromatography, wherein the antibody is eluted at a particular pH value; and
b) purifying using cation exchange chromatography in the bind-elute mode, wherein an eluate from step a) is loaded onto a cation exchange resin without substantial adjustment of pH.
2. A process according to claim 1 , wherein the antibody is eluted in step a) at pH values about 3.3 to about 4.5.
3. A process according to claim 1 , wherein the antibody is eluted in step a) at a pH value about 3.5.
4. A process for purifying antibodies, comprising:
a) purifying using protein A chromatography, wherein an antibody is eluted at a first pH value;
b) purifying using cation exchange chromatography performed in the bind-elute mode, wherein eluate obtained from step a) is loaded onto a cation exchange resin at a second pH value, and elution is carried out at a third pH value; and
c) purifying using anion exchange chromatography performed in the flow-through mode, wherein eluate from step b) is loaded on to an anion exchange resin without substantial adjustment of pH.
5. A process according to claim 4, wherein the first and second pH values are similar.
6. A process according to claim 4, wherein the first and second values are different.
7. A process according to claim 4, wherein the second and third pH values are similar.
8. A process according to claim 4, wherein the first pH value is about 3.5
9. A process according to claim 4, wherein the second pH value is about 6 to about 8.
10. A process according to claim 4, wherein the second pH value is about
6.
1 1 . A process according to claim 4, wherein the third pH value is about 6 to about 8.
12. A process according to claim 4, wherein the third pH value is about 6.
13. A process according to claim 4, wherein elution performed in step b uses a linear or a step-wise increase of eluting fluid conductivity.
PCT/US2010/062245 2009-12-29 2010-12-28 Protein purification by ion exchange WO2011090719A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US13/518,701 US20130178608A1 (en) 2009-12-29 2010-12-28 Protein purification by ion exchange
EP10844264.1A EP2519536A4 (en) 2009-12-29 2010-12-28 Protein purification by ion exchange

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN3213/CHE/2009 2009-12-29
IN3213CH2009 2009-12-29

Publications (2)

Publication Number Publication Date
WO2011090719A2 true WO2011090719A2 (en) 2011-07-28
WO2011090719A3 WO2011090719A3 (en) 2011-11-24

Family

ID=44307480

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/062245 WO2011090719A2 (en) 2009-12-29 2010-12-28 Protein purification by ion exchange

Country Status (3)

Country Link
US (1) US20130178608A1 (en)
EP (2) EP2519536A4 (en)
WO (1) WO2011090719A2 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014099577A1 (en) * 2012-12-17 2014-06-26 Merck Sharp & Dohme Corp. Process for purifying insulin and analogues thereof
WO2014180852A1 (en) * 2013-05-06 2014-11-13 Sanofi Continuous multistep process for purifying antibodies
WO2016103139A1 (en) * 2014-12-22 2016-06-30 Dr. Reddy's Laboratories Method for reduction of aggregates
US9708400B2 (en) 2012-04-20 2017-07-18 Abbvie, Inc. Methods to modulate lysine variant distribution
TWI596107B (en) * 2013-06-25 2017-08-21 卡地拉保健有限公司 Novel purification process for monoclonal antibodies
US9957318B2 (en) 2012-04-20 2018-05-01 Abbvie Inc. Protein purification methods to reduce acidic species
US10023608B1 (en) 2013-03-13 2018-07-17 Amgen Inc. Protein purification methods to remove impurities
WO2019191416A1 (en) * 2018-03-29 2019-10-03 Bristol-Myers Squibb Company Methods of purifying monomeric monoclonal antibodies
EP3636657A1 (en) 2018-10-08 2020-04-15 Ablynx N.V. Chromatography-free antibody purification method
WO2021099536A1 (en) 2019-11-22 2021-05-27 Morphosys Ag Method to increase antibody yield during ion exchange chromatography

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2868093T3 (en) 2012-09-17 2021-10-21 Grace W R & Co Chromatography media and devices
EP3094390B1 (en) 2014-01-16 2021-07-07 W.R. Grace & CO. - CONN. Affinity chromatography media and chromatography devices
PL3137209T3 (en) 2014-05-02 2023-01-02 W.R. Grace & Co. - Conn. Functionalized support material and methods of making and using functionalized support material
ES2896897T3 (en) 2015-06-05 2022-02-28 Grace W R & Co Clarifying agents for the bioprocessing of adsorbents and methods for producing and using the same

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5089605A (en) 1987-03-13 1992-02-18 Repligen Corporation Immobilized immunoglobulin-binding proteins
US5084559A (en) 1987-03-27 1992-01-28 Repligen Corporation Protein a domain mutants
US4983722A (en) 1988-06-08 1991-01-08 Miles Inc. Removal of protein A from antibody preparations
GB9022547D0 (en) * 1990-10-17 1990-11-28 Wellcome Found Purified immunoglobulin
SE9503925D0 (en) 1995-11-07 1995-11-07 Pharmacia Biotech Ab Separation medium for IgG
US6013763A (en) 1996-06-04 2000-01-11 Genentech, Inc. Peptide variants of protein A
KR100870353B1 (en) 1997-04-07 2008-11-25 제넨테크, 인크. Anti-VEGF Antibodies
PT1601697E (en) 2003-02-28 2007-09-04 Lonza Biologics Plc Antibody purification by protein a and ion exchange chromatography
US20060257972A1 (en) * 2003-03-31 2006-11-16 Takashi Ishihara Purification of human monoclonal antibody and human polyclonal antibody
DK1648940T3 (en) * 2003-07-28 2016-08-15 Genentech Inc Reduction of leaching of protein A during protein A affinity chromatography
SE0400501D0 (en) * 2004-02-27 2004-02-27 Amersham Biosciences Ab Antibody purification
CN102887955A (en) 2006-04-05 2013-01-23 艾博特生物技术有限公司 Purified antibody composition
WO2009017491A1 (en) * 2006-06-14 2009-02-05 Smithkline Beecham Corporation Methods for purifying antibodies using ceramic hydroxyapatite
BRPI0716139A2 (en) * 2006-08-28 2013-09-17 Ares Trading Sa fc fusion protein purification process
JP5064558B2 (en) * 2007-06-01 2012-10-31 エフ.ホフマン−ラ ロシュ アーゲー Immunoglobulin purification
WO2009045897A1 (en) * 2007-10-03 2009-04-09 Dyax Corp. Systems and methods for purifying proteins
CN102574911B (en) * 2009-08-07 2017-06-06 Emd密理博公司 The method of target protein from one or more impurity of sample

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP2519536A4 *

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9957318B2 (en) 2012-04-20 2018-05-01 Abbvie Inc. Protein purification methods to reduce acidic species
US9708400B2 (en) 2012-04-20 2017-07-18 Abbvie, Inc. Methods to modulate lysine variant distribution
WO2014099577A1 (en) * 2012-12-17 2014-06-26 Merck Sharp & Dohme Corp. Process for purifying insulin and analogues thereof
US10421795B2 (en) 2012-12-17 2019-09-24 Merck Sharp & Dohme Corp. Process for purifying insulin and analogues thereof
US10023608B1 (en) 2013-03-13 2018-07-17 Amgen Inc. Protein purification methods to remove impurities
JP2016519137A (en) * 2013-05-06 2016-06-30 サノフイ Continuous multi-step method for purifying antibodies
WO2014180852A1 (en) * 2013-05-06 2014-11-13 Sanofi Continuous multistep process for purifying antibodies
KR102272851B1 (en) * 2013-05-06 2021-07-02 사노피 Continuous multistep process for purifying antibodies
EP3301115A1 (en) * 2013-05-06 2018-04-04 Sanofi Continuous multistep process for purifying antibodies
CN105358572A (en) * 2013-05-06 2016-02-24 赛诺菲 Continuous multistep process for purifying antibodies
KR20160005047A (en) * 2013-05-06 2016-01-13 사노피 Continuous multistep process for purifying antibodies
TWI631132B (en) * 2013-05-06 2018-08-01 賽諾菲公司 Continuous multistep process for purifying antibodies
JP2019104735A (en) * 2013-05-06 2019-06-27 サノフイSanofi Continuous multistep process for purifying antibodies
AU2014264697B2 (en) * 2013-05-06 2019-08-22 Sanofi Continuous multistep process for purifying antibodies
US10793622B2 (en) 2013-05-06 2020-10-06 Sanofi Continuous multistep process for purifying antibodies
TWI675041B (en) * 2013-05-06 2019-10-21 法商賽諾菲公司 Continuous multistep process for purifying antibodies
TWI596107B (en) * 2013-06-25 2017-08-21 卡地拉保健有限公司 Novel purification process for monoclonal antibodies
WO2016103139A1 (en) * 2014-12-22 2016-06-30 Dr. Reddy's Laboratories Method for reduction of aggregates
WO2019191416A1 (en) * 2018-03-29 2019-10-03 Bristol-Myers Squibb Company Methods of purifying monomeric monoclonal antibodies
EP3636657A1 (en) 2018-10-08 2020-04-15 Ablynx N.V. Chromatography-free antibody purification method
WO2020074483A1 (en) 2018-10-08 2020-04-16 Ablynx Nv Chromatography-free antibody purification method
WO2021099536A1 (en) 2019-11-22 2021-05-27 Morphosys Ag Method to increase antibody yield during ion exchange chromatography

Also Published As

Publication number Publication date
EP2519536A2 (en) 2012-11-07
EP2519536A4 (en) 2013-06-05
US20130178608A1 (en) 2013-07-11
WO2011090719A3 (en) 2011-11-24
EP2695889A1 (en) 2014-02-12

Similar Documents

Publication Publication Date Title
US20130178608A1 (en) Protein purification by ion exchange
JP6743074B2 (en) Method for reducing the level of one or more impurities in a sample during protein purification
KR101921552B1 (en) Immunoglobulin purification using pre-cleaning steps
US20130116413A1 (en) Purification of proteins
EP2729482B1 (en) Method for purifying fc-fusion protein
US20200282332A1 (en) Use of alkaline washes during chromatography to remove impurities
US20200283472A1 (en) A process for purification of fc-fusion proteins
EP2831096B1 (en) Affinity chromatography matrix
WO2013158279A1 (en) Protein purification methods to reduce acidic species
WO2013176754A9 (en) Novel purification of antibodies using hydrophobic interaction chromatography
US20160272673A1 (en) Isolation and purification of dvd-igs
US20140128577A1 (en) Purification of chimeric protein
US20100261886A1 (en) Protein purification by citrate precipitation
WO2012160536A1 (en) Antibody purification
CN112313248A (en) Method for purifying monomeric monoclonal antibodies
WO2014102814A1 (en) Process for the purification of fc fusion proteins
EP2714713B1 (en) Purification of anti-cd20 antibodies
WO2013102822A1 (en) Filtration method
EP2975050B1 (en) Quantification of misfolded TNFR2:Fc
WO2013054250A1 (en) Purification method
US20090264630A1 (en) Method of separating monomeric protein(s)
WO2020066270A1 (en) METHOD FOR PRODUCING κ CHAIN VARIABLE REGION-CONTAINING ANTIBODY AND/OR ANTIBODY FRAGMENT
WO2021176311A1 (en) An improved purification process for ranibizumab

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: 10844264

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2010844264

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 6590/CHENP/2012

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 13518701

Country of ref document: US