US20190062419A1 - Protein purification methods to reduce acidic species - Google Patents

Protein purification methods to reduce acidic species Download PDF

Info

Publication number
US20190062419A1
US20190062419A1 US15/951,560 US201815951560A US2019062419A1 US 20190062419 A1 US20190062419 A1 US 20190062419A1 US 201815951560 A US201815951560 A US 201815951560A US 2019062419 A1 US2019062419 A1 US 2019062419A1
Authority
US
United States
Prior art keywords
antibody
protein
certain embodiments
adalimumab
acidic species
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/951,560
Inventor
Natarajan Ramasubramanyan
Lihua Yang
Matthew Omon Herigstad
Hong Yang
Christopher CHUMSAE
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
AbbVie Inc
Original Assignee
AbbVie 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=48045072&utm_source=***_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20190062419(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by AbbVie Inc filed Critical AbbVie Inc
Priority to US15/951,560 priority Critical patent/US20190062419A1/en
Assigned to ABBVIE, INC. reassignment ABBVIE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHUMSAE, CHRISTOPHER, HERIGSTAD, Matthew Omon, RAMASUBRAMANYAN, NATARAJAN, YANG, HONG, YANG, LIHUA
Publication of US20190062419A1 publication Critical patent/US20190062419A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39591Stabilisation, fragmentation
    • 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
    • C07K1/165Extraction; Separation; Purification by chromatography mixed-mode 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
    • C07K1/18Ion-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
    • C07K1/20Partition-, reverse-phase or hydrophobic interaction 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
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man

Definitions

  • the instant invention relates to the field of protein production and purification, and in particular to compositions and processes for controlling the amount of product-related substances (e.g., product charge variants, aggregates, and fragments) and/or process-related impurities (e.g., host cell proteins and media components) present in purified preparations by applying particular chromatography conditions during such protein purification.
  • product-related substances e.g., product charge variants, aggregates, and fragments
  • process-related impurities e.g., host cell proteins and media components
  • heterogeneity includes, but is not limited to, the presence of product-related species, such as charged species heterogeneity, consisting of acidic species and basic species.
  • product-related species such as charged species heterogeneity, consisting of acidic species and basic species.
  • mAb monoclonal antibody
  • acidic species heterogeneities can be detected by various methods, such as WCX-10 HPLC (a weak cation exchange chromatography) or IEF (isoelectric focusing).
  • the acidic species identified using such techniques comprise a range of product-related substances such as antibody product fragments (e.g., Fc and Fab fragments), and/or post-translation modifications of the antibody product, such as, deamidated and/or glycoslyated antibodies.
  • product-related substances such as antibody product fragments (e.g., Fc and Fab fragments), and/or post-translation modifications of the antibody product, such as, deamidated and/or glycoslyated antibodies.
  • WCX-10 analysis measured the presence of acidic species that can be divided, based on residence time, into two groups: acidic region 1 (AR1) and acidic region 2 (AR2). Because of their similar chemical characteristics to the antibody molecules of interest, reduction of acidic species is a particular challenge in monoclonal antibody purification.
  • the present invention is directed to process-related impurity-reduced and/or product-related substance-modulated preparations of a protein of interest.
  • the process-related impurities include, but are not limited to host cell proteins (HCPs), host nucleic acids, chromatographic materials, and media components.
  • the product-related substances include, but are not limited to charge variants, such as acidic species and basic species.
  • such acidic species correspond to heterogeneity in the distribution of protein fragments (e.g., Fc and Fab fragments of antibodies), and/or post-translation modifications of the proteins, such as, deamidated and/or glycoslyated proteins, in the population of proteins, and such heterogeneity particularly of interest when it arises in the context of recombinant protein production.
  • the present invention is directed toward pharmaceutical compositions comprising one or more proteins purified by a method described herein.
  • such compositions further comprise one or more pharmaceutical agents.
  • the present invention is directed to a method for preparing a process-related impurity-reduced and/or product-related substance-modulated composition comprising a protein of interest wherein a chromatographic separation is performed to identify the particular conditions, e.g., salt concentration, pH, temperature, load amount and conditions, and washing conditions, sufficient to elicit the desired fractionation profile, e.g., fractionation of product-related substancs, such as acidic species and lysine variants, of a sample comprising the protein of interest and at least one process-related impurity and/or at least one product-related substance.
  • the method will further comprise pooling of the resulting fractions comprising the desired process-related impurity-reduced and/or product-related substance-modulated composition comprising a protein of interest.
  • the present invention is directed to methods for isolating and purifying a protein, for example, an antibody, or an antigen-binding portion thereof, from a sample in order to exert control over the presence of process-related impurities and/or product-related substances.
  • the methods of purifying a protein, such as an antibody or antigen-binding portion thereof, from a sample, as described herein reduces the amount of acidic species present in the resulting composition.
  • the resulting composition is substantially free of acidic species.
  • the resulting composition is substantially free of one or more acidic sub-species, for example, with regard to the purification of Adalimumab, the composition is substantially free of AR1 and/or AR2.
  • the methods described herein reduce the amount of host cell proteins (“HCPs”) present in the resulting composition.
  • the resulting composition is substantially free of HCPs.
  • the sample mixture to be purified comprises a partially purified cell line harvest wherein the cell line is employed to produce specific proteins of the present invention.
  • the sample mixture is prepared from a cell line used to produce anti-TNF- ⁇ antibodies.
  • the invention is directed to methods of protein purification employing chromatography, preferably chromatography that utilizes a multimodal (also known as “mixed mode” or “MM”) media.
  • chromatography preferably chromatography that utilizes a multimodal (also known as “mixed mode” or “MM”) media.
  • MM mixed mode
  • the multimodal media comprises functional groups which exhibit anion exchange and/or hydrophobic interactions.
  • the multimodal media comprises a cross-linked agarose with a ligand, for example, N-Benzyl-N-methyl ethanol amine, that exhibits ionic interactions, hydrogen bonding and hydrophobic interactions.
  • the cross-linked agarose with a ligand N-Benzyl-N-methyl ethanol amine has the following structure:
  • the multimodal media comprises a cross-linked cellulose exhibiting porosity.
  • the cross-linked cellulose is a phenylpropylamine having the following structure:
  • the cross-linked cellulose is a hexylamine having the following structure:
  • a sample comprising the protein of interest such as an antibody or antigen-binding portion thereof, is subjected to chromatography that utilizes a multimodal media, wherein the sample is subjected to a pH adjustment during loading.
  • the pH is adjusted to a basic pH, or an increase in pH.
  • An example of a suitable pH is between about a pH of 7 and 8.2, preferably a pH of between about 7.5 and 8.2.
  • the selection of appropriate pH will be based on the characteristics of the antibody and/or acidic species of interest.
  • the pH will be selected to be about 0 to 3 units lower than the pI of the protein. In certain embodiments, it is in the range of 1 to 2 units lower. In certain embodiments, it is in the range of 1 to 1.5 units lower.
  • Certain embodiments of the present invention involve subjecting a sample mixture comprising a protein of interest, such as an antibody or antigen-binding portion thereof, to chromatography that utilizes a multimodal media, wherein the sample is subjected to a conductivity adjustment during loading.
  • the conductivity is adjusted to between about 1 and 86 mS/cm, preferably between about 2 and 14 mS/cm.
  • alternative ranges of conductivity are employed, which would be based on the characteristics of the antibody and/or acidic species of interest.
  • Certain embodiments of the present invention involve subjecting a sample comprising a protein of interest, such as an antibody or antigen-binding portion thereof, to chromatography that utilizes a multimodal media, wherein the sample is subjected to an adjustment in the amount of protein load used in the multimodal chromatography.
  • the total protein load to the column is of between about 5 and 1000 g/L, or between about 50 and 500 g/L, between about 75 and 300 g/L, or between about 100 and 250 g/L.
  • the protein concentration of the load protein mixture is adjusted to a protein concentration of the material loaded to the column of about 0.5 and 50 g/L, or between about 1 and 20 g/L.
  • the methods of the present invention involves subjecting a sample comprising a protein of interest, such as an antibody or antigen-binding portion thereof, to chromatography that utilizes an anion exchange (AEX) adsorbent material and an aqueous salt solution under loading conditions that permit both the protein of interest and non-target proteins to bind to the AEX adsorbent and collecting any unbound material with reduced levels of acidic species (and optionally reduced levels of one or more product related impurities/substances or process related impurities) and subsequently wing the adsorbent with awash buffer comprising the same, or substantially similar, aqueous salt solution used in the loading sample and collecting the effluent containing reduced levels of acidic species (and optionally reduced levels of one or more product related impurities/substances or process related impurities).
  • AEX anion exchange
  • the salt concentration is between 0.5 mM and 50 mM, or 2 mM and 40 mM, or 5 mM and 20 mM, depending on the salt type and AEX adsorbent being used.
  • the concentration of the anionic and/or cationic agent in aqueous salt solution is increased or decreased to achieve a pH of between about 5 and 10, or between about 7 and 9.
  • the aqueous salt solution comprises an anionic agent at a concentration of about 5 mM, 10 mM or 18.5 mM, and an amount of a cationic agent sufficient to achieve a pH of 8.8 or 9.0.
  • the present invention involve subjecting a sample comprising the putative protein of interest, such as an antibody or antigen-binding portion thereof, to chromatography that utilizes a AEX, wherein the sample is subjected to an adjustment in the amount of protein concentration and load.
  • the total protein load to the column of between about 50 and 500 g/L, or between about 75 and 350 g/L, or between about 200 and 300 g/L.
  • the protein concentration of the load protein mixture is adjusted to a protein concentration of the material loaded to the column of about 0.5 and 50 g/L, between about 1 and 20 g/L, or between 3 and 10 g/L.
  • the methods of the present invention involves subjecting a sample comprising a putative protein of interest, such as an antibody or antigen-binding portion thereof, to chromatography that utilizes a cation exchange (CEX) adsorbent material and an aqueous solution under loading conditions that permit both the protein of interest and non-target proteins to bind to the CEX adsorbent, wherein acidic species and non-target proteins are washed from the CEX adsorbent material using a wash buffer comprising the same, or substantially similar, aqueous solution as the loading buffer, and wherein the bound protein of interest is subsequently recovered with an elution buffer having a higher conductivity than the loading buffer.
  • a putative protein of interest such as an antibody or antigen-binding portion thereof
  • a sample comprising the protein of interest, such as an antibody or antigen-binding portion thereof, to chromatography that utilizes a CEX, wherein the aqueous solution for loading and wash is a combination of pH and ionic concentration to allow the removal the acidic species in the unbound wash fractions.
  • the pH employed in certain of such embodiments is adjusted such that it is below the pI of the protein of interest.
  • the present invention involve subjecting a sample comprising the putative protein of interest, such as an antibody or antigen-binding portion thereof, to chromatography that utilizes a CEX, wherein the sample is subjected to an adjustment in the amount of protein concentration and load.
  • the total protein load to the column of between about 5 and 150 g/L, or between about 10 and 100 g/L, between about 20 and 80 g/L, or between about 30 and 50 g/L.
  • the protein concentration of the load protein mixture is adjusted to a protein concentration of the material loaded to the column of about 0.5 and 50 g/L, or between about 1 and 20 g/L.
  • control over the amount of acidic species in the protein compositions described herein is exerted by employing one or more of the foregoing methods during the production and purification of the desired proteins, such as antibodies or antigen-binding portions thereof, described herein.
  • the sample is subject to a first chromatographic step prior to the multimodal/AEX or CEX media chromatography described above.
  • the sample in each case is prepared appropriately to achieve the target pH and ion concentration prior to separation on the different modes of chromatography.
  • Such prior chromatographic steps include ion exchange and/or affinity chromatography.
  • chromatographic supports for use in the first chromatographic step include, but are not limited to, affinity chromatographic resins, such as, but not limited to, Protein A resin and Protein G resin, or other affinity supports such as those comprising the antigen against which an antibody of interest was raised, as well as affinity supports comprising an Fc binding protein.
  • a sample is loaded on a protein A affinity column chromatography and eluted with a buffer system containing buffer components to be used in the multimodal media chromatography.
  • a buffer system containing buffer components to be used in the multimodal media chromatography.
  • the pH is adjusted to pH 3.5 to 3.7 with acid (e.g., the same as the acidic component of the multimodal media chromatography equilibration buffer system) and held for about 30 to 90 minutes.
  • the material is then neutralized with base) to the designed pH.
  • the buffer is a Tris/acetate buffer system.
  • the buffer is a trolamine/NaCl buffer. The material is then clarified with filters.
  • the eluate can be monitored using techniques well known to those skilled in the art. For example, the absorbance at OD 280 can be followed.
  • the eluated fraction(s) of interest can then be prepared for further processing with multimodal chromatography, AEX or CEX media chromatography.
  • the purity of the proteins of interest can be analyzed using methods well known to those skilled in the art, e.g., weak cation exchange chromatography (WCX), capillary isoelectric focusing (cIEF), size-exclusion chromatography, PorosTM A HPLC Assay, HCP ELISA, Protein A ELISA, and western blot analysis.
  • WCX weak cation exchange chromatography
  • cIEF capillary isoelectric focusing
  • size-exclusion chromatography size-exclusion chromatography
  • PorosTM A HPLC Assay HCP ELISA
  • Protein A ELISA Protein A ELISA
  • western blot analysis e.g., western blot analysis.
  • FIG. 1 depicts a process chromatogram of pH gradient elution in the context of AEX chromatography.
  • FIG. 2 depicts a process chromatogram of a linear gradient elution by increasing anion concentration in the context of AEX chromatography.
  • FIG. 3 depicts a process chromatogram of fractionation of 300 g/L load and wash in the context of AEX chromatography.
  • FIG. 4 depicts the effect of pH on AR reduction in the context of AEX chromatography.
  • FIG. 5 depicts a process chromatogram at different salt (cation) concentrations in the context of CEX chromatography.
  • FIG. 6 depicts recovery versus AR reduction in the context of CEX purification of adalimumab.
  • FIG. 7 depicts the WCX-10 profile of glycated load material and CEX Eluate.
  • FIG. 8 depicts the WCX 10 profile of MGO modified load material and eluate from CEX column employing Toyo Pearl MX TRP 650M resin.
  • FIG. 9 depicts the change in Lysine distribution during CEX chromatography, highlighting the effect of Tris concentration.
  • FIG. 10 depicts the effect of pH and conductivity on Adalimumab AR reduction and recovery yield in the context of MM chromatography.
  • FIG. 11 depicts the AR reduction achieved with the corresponding protein recovery in the context of MM chromatography.
  • FIG. 12 depicts the total Adalimumab Protein concentration levels and AR levels during Flow Through and Wash.
  • FIG. 13 depicts the total mAb B Protein concentration levels and AR levels during Flow Through and Wash in the context of MM chromatography.
  • FIG. 14 depicts the total mAb C Protein concentration levels and AR levels during Flow Through and Wash in the context of MM chromatography.
  • FIG. 15 depicts the Cumulative % AR breakthrough of mAb C on different MM resins.
  • FIG. 16 depicts the impact of pH-pI and Conductivity on D2E7 (Adalimumab) AR Reduction in the context of MM chromatography.
  • FIG. 17 depicts the impact of pH-pI and Conductivity on mAb B AR Reduction in the context of MM chromatography.
  • FIG. 18 depicts the impact and trend of pH-pI on mAb C AR reduction with multiple resins in the context of MM chromatography.
  • FIG. 19 depicts the effect of pH and Conductivity on AR reduction and Yield in the context of MM chromatography.
  • FIG. 20 depicts AR reduction and Protein recovery vs. pH in the context of MM chromatography.
  • FIG. 21 depicts the effect of pH, conductivity and protein load amount on AR reduction and Yield.
  • FIG. 22 depicts the effect of pH, conductivity and protein load amount on AR reduction and Yield.
  • FIG. 23 depicts the AR Growth at 25° C. of low and high AR containing samples.
  • FIG. 24 depicts the effect of AEX adsorbent pKa for mAb B with several different AEX adsorbents, with different pKa values, run at with an acetate/Tris buffer at pH 9.1.
  • the instant invention relates to the field of protein purification.
  • the instant invention relates to compositions and processes for controlling the amount of product-related substances (e.g., product charge variants, aggregates, and fragments) and/or process-related impurities (e.g., host cell proteins and media components) present in purified preparations of a protein of interest.
  • product-related substances e.g., product charge variants, aggregates, and fragments
  • process-related impurities e.g., host cell proteins and media components
  • the methods described herein involve the purification of a protein, such as, but not limited to an antibody or antigen-binding portion thereof, by multimodal chromatography, wherein the multimodal (MM) media comprises both ion exchange and hydrophobic interaction functional groups and an aqueous salt solution, wherein the same or substantially the same aqueous salt solution is used as a loading buffer and a wash buffer using which the said protein of interested is collected with, in the column effluent.
  • MM multimodal
  • the methods described herein involve the purification of a protein, such as, but not limited to an antibody or antigen-binding portion thereof, by chromatography comprising an anion exchange (AEX) adsorbent material and an aqueous salt solution, wherein the same or substantially the same aqueous salt solution is used as a loading buffer and a wash buffer using which the said protein of interested is collected with, in the column effluent.
  • a protein such as, but not limited to an antibody or antigen-binding portion thereof
  • chromatography comprising an anion exchange (AEX) adsorbent material and an aqueous salt solution, wherein the same or substantially the same aqueous salt solution is used as a loading buffer and a wash buffer using which the said protein of interested is collected with, in the column effluent.
  • AEX anion exchange
  • the methods described herein involve the purification of a protein, such as, but not limited to an antibody or antigen-binding portion thereof, by chromatography comprising a cation exchange (CEX) adsorbent material and an aqueous salt solution, wherein the same or substantially the same aqueous salt solution is used as a loading buffer and a wash buffer, wherein the wash buffer removes acidic species and non-target proteins bound to the CEX adsorbent material, and wherein the target protein bound to the CEX adsorbent material is eluted with a buffer having a higher conductivity and/or pH than the loading/wash buffer.
  • the present invention is directed toward pharmaceutical compositions comprising one or more proteins, such as, but not limited to an antibody or antigen-binding portion thereof, purified by a method described herein.
  • product refers to a protein of interest, which may be present in the context of a sample comprising one or more process-related impurities and/or product-related substances.
  • the product i.e., the protein of interest is an antibody or antigen binding fragment thereof.
  • antibody includes an immunoglobulin molecule comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region (CH).
  • the heavy chain constant region is comprised of three domains, CH1, CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDRs complementarity determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • antibody also includes alternative antibody and antibody-like structures, such as, but not limited to, dual variable domain antibodies (DVD-Ig).
  • antibody portion includes fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., hIL-12, hTNF ⁇ , or hIL-18). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • an antigen e.g., hIL-12, hTNF ⁇ , or hIL-18. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment comprising the VL, VH, CL and CH1 domains; (ii) a F(ab′)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment comprising the VH and CH1 domains; (iv) a Fv fragment comprising the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546, the entire teaching of which is incorporated herein by reference), which comprises a VH domain; and (vi) an isolated complementarity determining region (CDR).
  • a Fab fragment a monovalent fragment comprising the VL, VH, CL and CH1 domains
  • a F(ab′)2 fragment a bivalent fragment comprising two
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see, e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883, the entire teachings of which are incorporated herein by reference).
  • Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody.
  • Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see, e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123, the entire teachings of which are incorporated herein by reference).
  • an antibody may be part of a larger immunoadhesion molecule, formed by covalent or non-covalent association of the antibody with one or more other proteins or peptides.
  • immunoadhesion molecules include use of the streptavidin core region to make a tetrameric scFv molecule (Kipriyanov, S. M., et al. (1995) Human Antibodies and Hybridomas 6:93-101, the entire teaching of which is incorporated herein by reference) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov, S. M., et al.
  • Antibody portions such as Fab and F(ab′)2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion, respectively, of whole antibodies.
  • antibodies, antibody portions and immunoadhesion molecules can be obtained using standard recombinant DNA techniques, as described herein.
  • the antigen binding portions are complete domains or pairs of complete domains.
  • Kabat numbering “Kabat definitions” and “Kabat labeling” are used interchangeably herein. These terms, which are recognized in the art, refer to a system of numbering amino acid residues which are more variable (i.e., hypervariable) than other amino acid residues in the heavy and light chain variable regions of an antibody, or an antigen binding portion thereof (Kabat et al. (1971) Ann. NY Acad, Sci. 190:382-391 and, Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242, the entire teachings of which are incorporated herein by reference).
  • the hypervariable region ranges from amino acid positions 31 to 35 for CDR1, amino acid positions 50 to 65 for CDR2, and amino acid positions 95 to 102 for CDR3.
  • the hypervariable region ranges from amino acid positions 24 to 34 for CDR1, amino acid positions 50 to 56 for CDR2, and amino acid positions 89 to 97 for CDR3.
  • human antibody includes antibodies having variable and constant regions corresponding to human germline immunoglobulin sequences as described by Kabat et al. (See Kabat, et al. (1991) Sequences of proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242).
  • the human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), e.g., in the CDRs and in particular CDR3.
  • the mutations can be introduced using the “selective mutagenesis approach.”
  • the human antibody can have at least one position replaced with an amino acid residue, e.g., an activity enhancing amino acid residue which is not encoded by the human germline immunoglobulin sequence.
  • the human antibody can have up to twenty positions replaced with amino acid residues which are not part of the human germline immunoglobulin sequence. In other embodiments, up to ten, up to five, up to three or up to two positions are replaced. In one embodiment, these replacements are within the CDR regions.
  • the term “human antibody”, as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • recombinant human antibody includes human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial human antibody library, antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see, e.g., Taylor, L. D., et al. (1992) Nucl. Acids Res. 20:6287-6295, the entire teaching of which is incorporated herein by reference) or antibodies prepared, expressed, created or isolated by any other means that involves splicing of human immunoglobulin gene sequences to other DNA sequences.
  • recombinant means such as antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial human antibody library, antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobul
  • Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences (see, Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242).
  • such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • such recombinant antibodies are the result of selective mutagenesis approach or back-mutation or both.
  • an “isolated antibody” includes an antibody that is substantially free of other antibodies having different antigenic specificities. Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals.
  • Koff is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex.
  • Kd is intended to refer to the dissociation constant of a particular antibody-antigen interaction.
  • nucleic acid molecule includes DNA molecules and RNA molecules.
  • a nucleic acid molecule may be single-stranded or double-stranded, but in one aspect is double-stranded DNA.
  • isolated nucleic acid molecule as used herein in reference to nucleic acids encoding antibodies or antibody portions (e.g., VH, VL, CDR3), e.g. an antibody having a weak binding capacity for a Protein A resin.
  • isolated nucleic acid molecule is also intended to include sequences encoding bivalent, bispecific antibodies, such as diabodies in which VH and VL regions contain no other sequences other than the sequences of the diabody.
  • recombinant host cell includes a cell into which a recombinant expression vector has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell but to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein.
  • modifying is intended to refer to changing one or more amino acids in the antibodies or antigen-binding portions thereof.
  • the change can be produced by adding, substituting or deleting an amino acid at one or more positions.
  • the change can be produced using known techniques, such as PCR mutagenesis.
  • preparative scale refers to a scale of purification operation that can be readily scaled-up and implemented at large scale manufacturing while still providing desired separation.
  • one skilled in the field may develop a process using, e.g., a 0.5 cm (i.d.) ⁇ 20 cm (L) column in the lab, and transfer it to large scale production using, e.g., a 30 cm (i.d.) ⁇ 20 cm (L) column packed with the same resin and operated with the same set of buffers, same linear flow rates (or residence times) and buffer volumes.
  • column bed height is typically ⁇ about 30 cm and column pressure drop ⁇ about 5 bar.
  • aggregates used herein means agglomeration or oligomerization of two or more individual molecules, including but not limiting to, protein dimers, trimers, tetramers, oligomers and other high molecular weight species. Protein aggregates can be soluble or insoluble.
  • fragments refers to any truncated protein species from the target molecule due to dissociation of peptide chain, enzymatic and/or chemical modifications.
  • antibody fragments include, but not limited to, Fab, F(ab′) 2 , Fv, scFv, Fd, dAb, or other compositions that contain a portion of the antibody molecule.
  • charge variants refers to the full complement of product variants including, but not limited to acidic species and basic species (e.g., Lys variants).
  • product variants can include product aggregates and/or product fragments, to the extent that such aggregation and/or fragmentation results in a product charge variation.
  • lysine variant heterogeneity refers to a characteristic of a population of proteins wherein the population consists of proteins of substantially identical amino acid sequence, but where the population exhibits variation in the presence or absence of C-terminal lysine residues. Although such lysine variant heterogeneity can be observed under general cell culture conditions, the use of particular cell culture conditions, as detailed below, can increase or decrease the distribution or amount of lysine variant heterogeneity.
  • the protein is an antibody
  • the distribution of lysine variant heterogeneity comprises a distribution of the lysine variants Lys 0, Lys 1 and Lys 2, wherein the Lys 0 lysine variant comprises an antibody with heavy chains that do not comprise a C-terminal lysine, wherein the Lys 1 lysine variant comprises an antibody with one heavy chain that comprises a C-terminal lysine, and wherein the Lys 2 lysine variant comprises an antibody wherein both heavy chains comprise a C-terminal lysine.
  • C-terminal lysine variants are associated with charge heterogeneities present in protein preparations, for example, monoclonal antibody (mAb) preparations, produced through a cell culture process.
  • mAb monoclonal antibody
  • These heterogeneities can be detected by various methods, such as, for example, WCX-10 HPLC (a weak cation exchange chromatography), or IEF (isoelectric focusing).
  • the heterogeneity arises from subspecies of protein differing by the presence or absence of C-terminal lysines.
  • the population of proteins may comprise more than one subspecies of lysine variant.
  • the lysine variants may comprise at least two of Lys 0, Lys 1 and Lys 2 lysine variants which can be detected by weak cation exchange chromatography of the expression product of a host cell expressing Adalimumab.
  • the heterogeneity arises from the size of subpopulations having different C-terminal lysine profiles.
  • the population of proteins may comprise more than one subspecies of C-terminal lysine variant, and each of the variants may be present in different amounts.
  • the C-terminal lysine variants may be at least two of the Lys 0, Lys 1 and Lys 2 lysine variants detected by weak cation exchange chromatography of the expression product of a host cell expressing Adalimumab.
  • Lys 0, Lys 1 or Lys 2 subspecies are present in different amounts.
  • the heterogeneity arises from both a difference in the amount of lysine variants in the population of proteins and the type of lysine variants present in the population of proteins.
  • the terms “acidic species”, “acidic region” and “acidic species heterogeneity” refer to a characteristic of a population of proteins wherein the population includes a distribution of product-related impurities identifiable by the presence of charge heterogeneities.
  • acidic species heterogeneities can be detected by various methods, such as, for example, WCX-10 HPLC (a weak cation exchange chromatography), or IEF (isoelectric focusing).
  • the acidic species identified using such techniques comprise a mixture of product-related impurities containing antibody product fragments (e.g., Fc and Fab fragments), and/or post-translation modifications of the antibody product, such as, deamidated and/or glycoslyated antibodies.
  • product-related impurities containing antibody product fragments e.g., Fc and Fab fragments
  • post-translation modifications of the antibody product such as, deamidated and/or glycoslyated antibodies.
  • the acidic species heterogeneity comprises a difference in the type of acidic species present in the population of proteins.
  • the population of proteins may comprise more than one acidic species variant.
  • the heterogeneity of the distribution of acidic species comprises a difference in the amount of acidic species in the population of proteins.
  • the population of proteins may comprise more than one acidic species variant, and each of the variants may be present in different amounts.
  • antibody refers to an intact antibody or an antigen binding fragment thereof.
  • the antibodies of the present disclosure can be generated by a variety of techniques, including immunization of an animal with the antigen of interest followed by conventional monoclonal antibody methodologies e.g., the standard somatic cell hybridization technique of Kohler and Milstein (1975) Nature 256: 495. Although somatic cell hybridization procedures are preferred, in principle, other techniques for producing monoclonal antibody can be employed e.g., viral or oncogenic transformation of B lymphocytes.
  • the animal system for preparing hybridomas is the murine system.
  • Hybridoma production is a very well-established procedure. Immunization protocols and techniques for isolation of immunized splenocytes for fusion are known in the art. Fusion partners (e.g., murine myeloma cells) and fusion procedures are also known.
  • An antibody can be, in certain embodiments, a human, a chimeric, or a humanized antibody.
  • Humanized antibodies of the present disclosure can be prepared based on the sequence of a non-human monoclonal antibody prepared as described above.
  • DNA encoding the heavy and light chain immunoglobulins can be obtained from the non-human hybridoma of interest and engineered to contain non-murine (e.g., human) immunoglobulin sequences using standard molecular biology techniques.
  • murine variable regions can be linked to human constant regions using methods known in the art (see e.g., U.S. Pat. No. 4,816,567 to Cabilly et al.).
  • murine CDR regions can be inserted into a human framework using methods known in the art (see e.g., U.S. Pat. No. 5,225,539 to Winter, and U.S. Pat. Nos. 5,530,101; 5,585,089; 5,693,762 and 6,180,370 to Queen et al.).
  • Human monoclonal antibodies can be generated using transgenic or transchromosomic mice carrying parts of the human immune system rather than the mouse system.
  • transgenic and transchromosomic mice include mice referred to herein as the HuMAb Mouse® (Medarex, Inc.), KM Mouse® (Medarex, Inc.), and XenoMouse® (Amgen).
  • mice carrying both a human heavy chain transchromosome and a human light chain tranchromosome referred to as “TC mice” can be used; such mice are described in Tomizuka et al. (2000) Proc. Natl. Acad. Sci. USA 97:722-727.
  • cows carrying human heavy and light chain transchromosomes have been described in the art (e.g., Kuroiwa et al. (2002) Nature Biotechnology 20:889-894 and PCT application No. WO 2002/092812) and can be used to raise the antibodies of this disclosure.
  • the antibodies of this disclosure are recombinant human antibodies, which can be isolated by screening of a recombinant combinatorial antibody library, e.g., a scFv phage display library, prepared using human VL and VH cDNAs prepared from mRNA derived from human lymphocytes. Methodologies for preparing and screening such libraries are known in the art. In addition to commercially available kits for generating phage display libraries (e.g., the Pharmacia Recombinant Phage Antibody System, catalog no. 27-9400-01; and the Stratagene SurfZAPTM phage display kit, catalog no.
  • kits for generating phage display libraries e.g., the Pharmacia Recombinant Phage Antibody System, catalog no. 27-9400-01; and the Stratagene SurfZAPTM phage display kit, catalog no.
  • examples of methods and reagents particularly amenable for use in generating and screening antibody display libraries can be found in, e.g., Ladner et al. U.S. Pat. No. 5,223,409; Kang et al. PCT Publication No. WO 92/18619; Dower et al. PCT Publication No. WO 91/17271; Winter et al. PCT Publication No. WO 92/20791; Markland et al. PCT Publication No. WO 92/15679; Breitling et al. PCT Publication No. WO 93/01288; McCafferty et al.
  • Human monoclonal antibodies of this disclosure can also be prepared using SCID mice into which human immune cells have been reconstituted such that a human antibody response can be generated upon immunization.
  • SCID mice into which human immune cells have been reconstituted such that a human antibody response can be generated upon immunization.
  • Such mice are described in, for example, U.S. Pat. Nos. 5,476,996 and 5,698,767 to Wilson et al.
  • the antibodies or antigen-binding portions thereof, of this disclosure can be altered wherein the constant region of the antibody is modified to reduce at least one constant region-mediated biological effector function relative to an unmodified antibody.
  • the immunoglobulin constant region segment of the antibody can be mutated at particular regions necessary for Fc receptor (FcR) interactions (see, e.g., Canfield and Morrison (1991) J. Exp. Med. 173:1483-1491; and Lund et al. (1991) J. of Immunol. 147:2657-2662, the entire teachings of which are incorporated herein).
  • Reduction in FcR binding ability of the antibody may also reduce other effector functions which rely on FcR interactions, such as opsonization and phagocytosis and antigen-dependent cellular cytotoxicity.
  • DNAs encoding partial or full-length light and heavy chains are inserted into one or more expression vector such that the genes are operatively linked to transcriptional and translational control sequences.
  • operatively linked is intended to mean that an antibody gene is ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended function of regulating the transcription and translation of the antibody gene.
  • the expression vector and expression control sequences are chosen to be compatible with the expression host cell used.
  • the antibody light chain gene and the antibody heavy chain gene can be inserted into a separate vector or, more typically, both genes are inserted into the same expression vector.
  • the antibody genes are inserted into an expression vector by standard methods (e.g., ligation of complementary restriction sites on the antibody gene fragment and vector, or blunt end ligation if no restriction sites are present).
  • the expression vector may already carry antibody constant region sequences.
  • the recombinant expression vector can encode a signal peptide that facilitates secretion of the antibody chain from a host cell.
  • the antibody chain gene can be cloned into the vector such that the signal peptide is linked in-frame to the amino terminus of the antibody chain gene.
  • the signal peptide can be an immunoglobulin signal peptide or a heterologous signal peptide (i.e., a signal peptide from a non-immunoglobulin protein).
  • a recombinant expression vector of the invention can carry one or more regulatory sequence that controls the expression of the antibody chain genes in a host cell.
  • regulatory sequence is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the antibody chain genes.
  • Such regulatory sequences are described, e.g., in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990), the entire teaching of which is incorporated herein by reference.
  • Suitable regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV) (such as the CMV promoter/enhancer), Simian Virus 40 (SV40) (such as the SV40 promoter/enhancer), adenovirus, (e.g., the adenovirus major late promoter (AdMLP)) and polyoma.
  • CMV cytomegalovirus
  • SV40 Simian Virus 40
  • AdMLP adenovirus major late promoter
  • a recombinant expression vector of the invention may carry one or more additional sequences, such as a sequence that regulates replication of the vector in host cells (e.g., origins of replication) and/or a selectable marker gene.
  • the selectable marker gene facilitates selection of host cells into which the vector has been introduced (see e.g., U.S. Pat. Nos. 4,399,216, 4,634,665 and 5,179,017, all by Axel et al., the entire teachings of which are incorporated herein by reference).
  • the selectable marker gene confers resistance to drugs, such as G418, hygromycin or methotrexate, on a host cell into which the vector has been introduced.
  • Suitable selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr-host cells with methotrexate selection/amplification) and the neo gene (for G418 selection).
  • DHFR dihydrofolate reductase
  • An antibody of the invention can be prepared by recombinant expression of immunoglobulin light and heavy chain genes in a host cell.
  • a host cell is transfected with one or more recombinant expression vectors carrying DNA fragments encoding the immunoglobulin light and heavy chains of the antibody such that the light and heavy chains are expressed in the host cell and secreted into the medium in which the host cells are cultured, from which medium the antibodies can be recovered.
  • Standard recombinant DNA methodologies are used to obtain antibody heavy and light chain genes, incorporate these genes into recombinant expression vectors and introduce the vectors into host cells, such as those described in Sambrook, Fritsch and Maniatis (eds), Molecular Cloning; A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989), Ausubel et al. (eds.) Current Protocols in Molecular Biology, Greene Publishing Associates, (1989) and in U.S. Pat. Nos. 4,816,397 & 6,914,128, the entire teachings of which are incorporated herein.
  • the expression vector(s) encoding the heavy and light chains is (are) transfected into a host cell by standard techniques.
  • the various forms of the term “transfection” are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like.
  • eukaryotic cells such as mammalian host cells
  • expression of antibodies in eukaryotic cells is suitable because such eukaryotic cells, and in particular mammalian cells, are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active antibody.
  • Prokaryotic expression of antibody genes has been reported to be ineffective for production of high yields of active antibody (Boss and Wood (1985) Immunology Today 6:12-13, the entire teaching of which is incorporated herein by reference).
  • Suitable host cells for cloning or expressing the DNA in the vectors herein are the prokaryote, yeast, or higher eukaryote cells described above.
  • Suitable prokaryotes for this purpose include eubacteria, such as Gram-negative or Gram-positive organisms, e.g., Enterobacteriaceae such as Escherichia , e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella , e.g., Salmonella typhimurium, Serratia , e.g., Serratia marcescans , and Shigella , as well as Bacilli such as B. subtilis and B.
  • Enterobacteriaceae such as Escherichia , e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus
  • Salmonella e.g., Salmonella typhimurium
  • E. coli cloning host is E. coli 294 (ATCC 31,446), although other strains such as E. coli B, E. coli X1776 (ATCC 31,537), and E. coli W3110 (ATCC 27,325) are suitable. These examples are illustrative rather than limiting.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for polypeptide encoding vectors.
  • Saccharomyces cerevisiae or common baker's yeast, is the most commonly used among lower eukaryotic host microorganisms.
  • a number of other genera, species, and strains are commonly available and useful herein, such as Schizosaccharomyces pombe; Kluyveromyces hosts such as, e.g., K. lactis, K. fragilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K. wickeramii (ATCC 24,178), K.
  • waltii ATCC 56,500
  • K. drosophilarum ATCC 36,906
  • K. thermotolerans K. marxianus
  • yarrowia EP 402,226
  • Pichia pastoris EP 183,070
  • Candida Trichoderma reesia
  • Neurospora crassa Schwanniomyces such as Schwanniomyces occidentalis
  • filamentous fungi such as, e.g., Neurospora, Penicillium, Tolypocladium , and Aspergillus hosts such as A. nidulans and A. niger.
  • Suitable host cells for the expression of glycosylated antibodies are derived from multicellular organisms.
  • invertebrate cells include plant and insect cells.
  • Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mori have been identified.
  • a variety of viral strains for transfection are publicly available, e.g., the L-1 variant of Autographa californica NPV and the Bm-5 strain of Bombyx mori NPV, and such viruses may be used as the virus herein according to the present invention, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures of cotton, corn, potato, soybean, petunia , tomato, and tobacco can also be utilized as hosts.
  • Suitable mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr-CHO cells, described in Urlaub and Chasin, (1980) PNAS USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in Kaufman and Sharp (1982) Mol. Biol. 159:601-621, the entire teachings of which are incorporated herein by reference), NS0 myeloma cells, COS cells and SP2 cells.
  • Chinese Hamster Ovary CHO cells
  • dhfr-CHO cells described in Urlaub and Chasin, (1980) PNAS USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in Kaufman and Sharp (1982) Mol. Biol. 159:601-621, the entire teachings of which are incorporated herein by reference
  • NS0 myeloma cells COS cells and SP2 cells.
  • the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or secretion of the antibody into the culture medium in which the host cells are grown.
  • useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary cells/-DHFR (CHO, Urlaub et al., Proc.
  • mice sertoli cells TM4, Mather, Biol. Reprod. 23:243-251 (1980)); monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TRI cells (Mather et al., Annals N.Y. Acad. Sci. 383:44-68 (1982)); MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2), the entire teachings of which are incorporated herein by reference.
  • Host cells are transformed with the above-described expression or cloning vectors for antibody production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • the host cells used to produce an antibody may be cultured in a variety of media.
  • Commercially available media such as Ham's F10TM (Sigma), Minimal Essential MediumTM ((MEM), (Sigma), RPMI-1640 (Sigma), and Dulbecco's Modified Eagle's MediumTM ((DMEM), Sigma) are suitable for culturing the host cells.
  • any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as gentamycin drug), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art.
  • the culture conditions such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
  • Host cells can also be used to produce portions of intact antibodies, such as Fab fragments or scFv molecules. It is understood that variations on the above procedure are within the scope of the present invention. For example, in certain embodiments it may be desirable to transfect a host cell with DNA encoding either the light chain or the heavy chain (but not both) of an antibody of this invention. Recombinant DNA technology may also be used to remove some or the entire DNA encoding either or both of the light and heavy chains that is not necessary for binding to the antigen to which the putative antibody of interest binds. The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention.
  • bifunctional antibodies may be produced in which one heavy and one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than the one to which the putative antibody of interest binds, depending on the specificity of the antibody of the invention, by crosslinking an antibody of the invention to a second antibody by standard chemical crosslinking methods.
  • a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into dhfr-CHO cells by calcium phosphate-mediated transfection.
  • the antibody heavy and light chain genes are each operatively linked to CMV enhancer/AdMLP promoter regulatory elements to drive high levels of transcription of the genes.
  • the recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification.
  • the selected transformant host cells are cultured to allow for expression of the antibody heavy and light chains and intact antibody is recovered from the culture medium. Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover the antibody from the culture medium.
  • the antibody can be produced intracellularly, in the periplasmic space, or directly secreted into the medium.
  • the particulate debris either host cells or lysed cells (e.g., resulting from homogenization)
  • supernatants from such expression systems can be first concentrated using a commercially available protein concentration filter, e.g., an AmiconTM or Millipore PelliconTM ultrafiltration unit.
  • the first step of a purification process typically involves: lysis of the cell, which can be done by a variety of methods, including mechanical shear, osmotic shock, or enzymatic treatments. Such disruption releases the entire contents of the cell into the homogenate, and in addition produces subcellular fragments that are difficult to remove due to their small size. These are generally removed by differential centrifugation or by filtration.
  • supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, e.g., an AmiconTM or Millipore PelliconTM ultrafiltration unit.
  • a commercially available protein concentration filter e.g., an AmiconTM or Millipore PelliconTM ultrafiltration unit.
  • the recombinant host cells can also be separated from the cell culture medium, e.g., by tangential flow filtration.
  • Antibodies can be further recovered from the culture medium using the antibody purification methods of the invention.
  • the invention provides methods and compositions for producing a purified or partially purified (e.g., process-related impurity-reduced and/or product-related substance-modulated) protein preparation from a mixture comprising a protein of interest, e.g., an antibody, and at least one process-related impurity or product-related substance.
  • a protein of interest e.g., an antibody
  • the modulation of product-related substances involves the reduction of certain of such substances, while in other embodiments, such modulation can be to increase certain of such substances.
  • the compositions of the present invention include, but are not limited to, process-related impurity-reduced and/or product-related substance-modulated compositions comprising a protein of interest.
  • the present invention is directed to process-related impurity-reduced and/or product-related substance-modulated compositions comprising Adalimumab.
  • process-related impurity-reduced and/or product-related substance-modulated compositions process-related impurity-reduced and/or product-related substance-modulated compositions address the need for improved product characteristics, including, but not limited to, product stability, product safety and product efficacy.
  • the present invention is directed to a method for preparing a process-related impurity-reduced and/or product-related substance-modulated composition comprising a protein of interest wherein a chromatographic separation is performed to identify the particular conditions, e.g., salt concentration, pH, temperature, load amount and conditions, and washing conditions, sufficient to elicit the desired fractionation profile, e.g., fractionation of product-related substancs, such as acidic species and lysing variants, of a sample comprising the protein of interest and at least one process-related impurity and/or at least one product-related substance.
  • the method will further comprise pooling of the resulting fractions comprising the desired process-related impurity-reduced and/or product-related substance-modulated composition comprising a protein of interest.
  • the purification process of the invention begins at the separation step when the antibody has been produced using production methods described above and/or by alternative production methods conventional in the art.
  • a clarified solution or mixture comprising the protein of interest e.g., an antibody
  • separation of the protein of interest from process-related impurities, such as the other proteins produced by the cell, as well as product-related substances, such as charge variants and/or size variants (aggregates and fragments) is performed.
  • process-related impurities such as the other proteins produced by the cell
  • product-related substances such as charge variants and/or size variants (aggregates and fragments) is performed.
  • such separation is performed using CEX, AEX, and/or MM chromatography.
  • a combination of one or more different purification techniques including affinity separation step(s), ion exchange separation step(s), mixed-mode step(s), and/or hydrophobic interaction separation step(s) can also be employed.
  • additional purification steps separate mixtures of proteins on the basis of their charge, degree of hydrophobicity, and/or size.
  • additional separation steps are performed using chromatography, including hydrophobic, anionic or cationic interaction (or a combination thereof). Numerous chromatography resins are available for each of these techniques, allowing accurate tailoring of the purification scheme to the particular protein involved.
  • each of the separation methods is that proteins can either traverse at different rates down a column, achieving a physical separation that increases as they pass further down the column, or to adhere selectively to the separation medium, being then differentially eluted by different solvents.
  • the protein of interest is separated from impurities and or product-related substances when the impurities and/or product-related substances specifically adhere to the column and the protein of interest does not, i.e., the protein of interest is washed from the column, while in other cases the protein of interest will adhere to the column, while the impurities and/or product-related substances are washed from the column.
  • the initial steps of the purification methods of the present invention involve the clarification and primary recovery of antibody from a sample matrix.
  • the primary recovery will include one or more centrifugation steps to separate the antibody product from the cells and cell debris.
  • Centrifugation of the sample can be run at, for example, but not by way of limitation, 7,000 ⁇ g to approximately 12,750 ⁇ g. In the context of large scale purification, such centrifugation can occur on-line with a flow rate set to achieve, for example, but not by way of limitation, a turbidity level of 150 NTU in the resulting supernatant. Such supernatant can then be collected for further purification, or in-line filtered through one or more depth filters for further clarification of the sample.
  • the initial steps of the purification methods of the present invention involve the clarification and primary recovery of antibody from a sample matrix.
  • the primary recovery will include one or more centrifugation steps to separate the antibody product from the cells and cell debris.
  • Centrifugation of the sample can be run at, for example, but not by way of limitation, 7,000 ⁇ g to approximately 12,750 ⁇ g. In the context of large scale purification, such centrifugation can occur on-line with a flow rate set to achieve, for example, but not by way of limitation, a turbidity level of 150 NTU in the resulting supernatant. Such supernatant can then be collected for further purification, or in-line filtered through one or more depth filters for further clarification of the sample.
  • the primary recovery will include the use of one or more depth filtration steps to clarify the sample matrix and thereby aid in purifying the antibodies of interest in the present invention. In other embodiments, the primary recovery will include the use of one or more depth filtration steps post centrifugation to further clarify the sample matrix.
  • depth filters that can be used in the context of the instant invention include the Millistak+X0HC, F0HC, D0HC, A1HC, B1HC depth filters (EMD Millipore), CunoTM model 30/60ZA, 60/90 ZA, VR05, VR07, delipid depth filters (3M Corp.).
  • a 0.2 ⁇ m filter such as Sartorius's 0.45/0.2 ⁇ m SartoporeTM bi-layer or Millipore's Express SHR or SHC filter cartridges typically follows the depth filters.
  • the primary recovery process can also be a point at which to reduce or inactivate viruses that can be present in the sample matrix.
  • any one or more of a variety of methods of viral reduction/inactivation can be used during the primary recovery phase of purification including heat inactivation (pasteurization), pH inactivation, solvent/detergent treatment, UV and ⁇ -ray irradiation and the addition of certain chemical inactivating agents such as ⁇ -propiolactone or e.g., copper phenanthroline as in U.S. Pat. No. 4,534,972.
  • the sample matrix is exposed to detergent viral inactivation during the primary recovery phase.
  • the sample matrix may be exposed to low pH inactivation during the primary recovery phase.
  • the sample mixture can be adjusted, as needed, for further purification steps. For example, following low pH viral inactivation, the pH of the sample mixture is typically adjusted to a more neutral pH, e.g., from about 4.5 to about 8.5, prior to continuing the purification process. Additionally, the mixture may be diluted with water for injection (WFI) to obtain a desired conductivity.
  • WFI water for injection
  • the primary recovery sample is subjected to Protein A affinity chromatography to purify the antibody of interest away from process-related impurities, such as HCPs.
  • process-related impurities such as HCPs.
  • Protein A resin include, but not limited to, MabSelect SuReTM, MabSelect SuRe LX, MabSelect, MabSelect Xtra, rProtein A Sepharose from GE Healthcare, ProSep HC, ProSep Ultra, and ProSep Ultra Plus from EMD Millipore, MapCapture from Life Technologies.
  • the Protein A column can be equilibrated with a suitable buffer prior to sample loading. Following the loading of the column, the column can be washed one or multiple times using a suitable sets of buffers. The Protein A column can then be eluted using an appropriate elution buffer. The eluate can be monitored using techniques well known to those skilled in the art. The eluate fractions of interest can be collected and then prepared for further processing.
  • the Protein A eluate may subject to a viral inactivation step either by detergent or low pH, provided this step is not performed prior to the Protein A capture operation.
  • a proper detergent concentration or pH and time can be selected to obtain desired viral inactivation results.
  • the Protein A eluate is usually pH and/or conductivity adjusted for subsequent purification steps.
  • the Protein A eluate may be subjected to filtration through a depth filter to remove turbidity and/or various impurities from the antibody of interest prior to additional chromatographic polishing steps.
  • depth filters include, but not limited to, Millistak+X0HC, F0HC, D0HC, A1HC, and B1HC Pod filters (EMD Millipore), or Zeta Plus 30ZA/60ZA, 60ZA/90ZA, delipid, VR07, and VR05 filters (3M).
  • the Protein A eluate pool may need to be conditioned to proper pH and conductivity to obtain desired impurity removal and product recovery from the depth filtration step.
  • the instant invention provides methods for producing a process-related impurity and/or product-related substance-reduced protein preparation from a mixture comprising a protein of interest (i.e., a product) and at least one process-related impurity and/or product-related substance by subjecting the mixture to at least one anion exchange separation step.
  • the anion exchange step will occur after the above-described Protein A affinity step.
  • an anionic exchange material versus a cationic exchange material is based on the local charges of the protein of interest in a given solution. Therefore, it is within the scope of this invention to employ an anionic exchange step prior to the use of a cationic exchange step, or a cationic exchange step prior to the use of an anionic exchange step. Furthermore, it is within the scope of this invention to employ only an anionic exchange step, only an cationic exchange step, or any serial combination of the two (including serial combinations of one or both ion exchange steps with the other chromatographic separation technologies described herein).
  • the initial protein mixture can be contacted with the anion exchange material by using any of a variety of techniques, e.g., using a batch purification technique or a chromatographic technique.
  • anion exchange material is prepared in, or equilibrated to, the desired starting buffer.
  • a slurry of the anion exchange material is obtained.
  • the protein of interest e.g., antibody
  • the solution comprising the process-related impurities and/or product-related substances that do not bind to the AEX material is separated from the slurry, e.g., by allowing the slurry to settle and removing the supernatant.
  • the slurry can be subjected to one or more washing steps and/or elution steps.
  • a chromatographic apparatus In the context of chromatographic separation, a chromatographic apparatus, commonly cylindrical in shape, is employed to contain the chromatographic support material (e.g., AEX material) prepared in an appropriate buffer solution.
  • the chromatographic apparatus if cylindrical, can have a diameter of about 5 mm to about 2 meters, and a height of 5 cm to 50 cm, and in certain embodiments, particularly for large scale processing, a height of ⁇ 30 cm is employed.
  • any portion of the solution that does not bind to the chromatographic material is separated from the chromatographic material by washing the material and collecting fractions from column.
  • the chromatographic material can be subjected to one or more wash steps. If desired, the chromatographic material can then be contacted with a solution designed to desorb any components of the solution that have bound to the chromatographic material.
  • a wash step can be performed in the context of AEX chromatography using conditions similar to the load conditions or alternatively by decreasing the pH and/or increasing the ionic strength/conductivity of the wash in a step wise or linear gradient manner.
  • the resulting flow through and wash fractions can be analyzed and appropriate fractions pooled to achieve the desired reduction in charged variant species.
  • the aqueous salt solution used as both the loading and wash buffer has a pH that at or near the isoelectric point (pI) of the protein of interest.
  • the pH is about 0 to 2 units higher or lower than the pI of the protein of interest. In certain embodiments, it will be in the range of 0 to 0.5 units higher or lower. In certain embodiments, it will be at the pI of the antibody.
  • the anionic agent is selected from the group consisting of acetate, formate, or combinations thereof.
  • the cationic agent is selected from the group consisting of Tris, arginine, or combinations thereof.
  • a packed anion-exchange chromatography column, anion-exchange membrane device, anion-exchange monolithic device, or depth filter media can be operated either in bind-elute mode, flow-through mode, or a hybrid mode wherein the product exhibits binding to the chromatographic material, yet can be washed from the column using a buffer that is the same or substantially similar to the loading buffer.
  • the bind-elute mode the column or the membrane device is first conditioned with a buffer with appropriate ionic strength and pH under conditions where certain proteins will be immobilized on the resin based matrix. For example, in certain embodiments, during the feed load, the protein of interest will be adsorbed to the resin due to electrostatic attraction.
  • the product recovery is achieved by increasing the ionic strength (i.e., conductivity) of the elution buffer to compete with the solute for the charged sites of the anion exchange matrix.
  • ionic strength i.e., conductivity
  • Changing the pH and thereby altering the charge of the solute is another way to achieve elution of the solute.
  • the change in conductivity or pH may be gradual (gradient elution) or stepwise (step elution).
  • the column or the membrane device is operated at selected pH and conductivity such that the protein of interest does not bind to the resin or the membrane while the process-related impurities and product-related substances will either be retained on the column or will have a distinct elution profile as compared to the protein of interest.
  • process-related impurities and product-relates substances will bind to the chromatographic material (or flow through) in a manner distinct from the protein of interest, e.g., while the protein of interest and certain aggregates and/or fragments of the protein of interest may bind the chromatographic material, washes that preferentially remove the protein of interest can be applied.
  • the column is then regenerated before next use.
  • Non-limiting examples of anionic exchange substituents include diethylaminoethyl (DEAE), quaternary aminoethyl (QAE) and quaternary amine (Q) groups. Additional non-limiting examples include: Poros 50PI and Poros 50HQ, which are a rigid polymeric bead with a backbone consisting of cross-linked poly[styrene-divinylbenzene]; Capto Q Impres and Capto DEAE, which are a high flow agarose bead; Toyopearl QAE-550, Toyopearl DEAE-650, and Toyopearl GigaCap Q-650, which are a polymeric base bead; Fractogel® EMD TMAE Hicap, which is a synthetic polymeric resin with a tentacle ion exchanger; Sartobind STIC® PA nano, which is a salt-tolerant chromatographic membrane with a primary amine ligand; Sartobind Q nano; which is a strong anion
  • the protein load of the mixture comprising protein of interest is adjusted to a total protein load to the column of between about 50 and 500 g/L, or between about 75 and 350 g/L, or between about 200 and 300 g/L.
  • the protein concentration of the load protein mixture is adjusted to a protein concentration of the material loaded to the column of about 0.5 and 50 g/L, between about 1 and 20 g/L, or between 3 and 10 g/L.
  • additives such as poly ethylene glycol, detergents, amino acids, sugars, chaotropic agents can be added to enhance the performance of the separation, so as to achieve better recovery or product quality.
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR charge variants in the flow through and wash fractions while enriching for the same in the flow elution fraction, thereby producing protein preparations with reduced or free of AR variants.
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR1 charge variants in the flow through and wash fractions while enriching for the same in the flow elution fraction, thereby producing protein preparations with reduced or free of AR1 variants.
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR2 charge variants in the flow-through and wash fractions while enriching for the same in the flow elution fraction, thereby producing protein preparations with reduced or free of AR2 variants.
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of the methylglyoxal (MGO) variants in the flow through and wash fractions while enriching for the same in the elution fraction, thereby producing protein preparations with reduced or free of MGO variants (for example, see U.S. patent application Ser. No. 14/078,181).
  • MGO methylglyoxal
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of the glycated variants (schiff's base and permanently glycated forms) in the flow through and wash fractions while enriching for the same in the elution fraction, thereby producing protein preparations with reduced or free of glycated variants.
  • the loading, pH, conductivity of the AEX chromatography step, as well as elution pH conductivity can be modified to achieve a desired distribution of process-related impurities and/or product-relates substances.
  • certain embodiments are directed to the modulation of the lysine distribution of purified sample of a protein of interest, e.g., increasing Lys0 and decreasing Lys1 and Lys2.
  • the methods of the present invention allow for the preparation of samples wherein the amount of Lys0 is decreased, while the amount of Lys 1 and/or Lys2 is increased.
  • an AEX chromatographic separation can be performed and combinations of fractions can be pooled to achieve a combination of desired process-related impurity and/or product-relates substance levels, in addition to, or in place of merely modulating charge variant concentration.
  • spectroscopy methods such as UV, NIR, FTIR, Fluorescence, Raman may be used to monitor levels of product-related charge variants, aggregates, low molecular weight variants (e.g., fragments of the protein of interest) in an on-line, at-line or in-line mode, which can then be used to control the level of charge variants, e.g., acidic species, in the pooled material collected from the AEX effluent.
  • specific signals arising from the chemical modification of the proteins such as glycation, MGO modification, deamidation, glycosylation may be specifically measurable by spectroscopic methods through such in-line, on-line or at-line methods, enabling realtime or near-real time control of product quality of the resulting product.
  • on-line, at-line or in-line monitoring methods can be used either on the effluent line of the chromatography step or in the collection vessel, to enable achievement of the desired product quality/recovery.
  • the UV signal can be used as a surrogate to achieve an appropriate product quality/recovery, wherein the UV signal can be processed appropriately, including, but not limited to, such processing techniques as integration, differentiation, moving average, such that normal process variability can be addressed and the target product quality can be achieved.
  • processing techniques as integration, differentiation, moving average, such that normal process variability can be addressed and the target product quality can be achieved.
  • such measurements can be combined with in-line dilution methods such that ion concentration/conductivity of the load/wash can be controlled by feedback and hence facilitate product quality control.
  • a combination of AEX and CEX and MM methods can be used to prepare product-related substance-modulated materials, including certain embodiments where one technology is used in a complementary/supplementary manner with another technology.
  • such a combination can be performed such that certain sub-species are removed predominantly by one technology, such that the combination provides the desired final composition/product quality.
  • such combinations include the use of additional intervening chromatography, filtration, pH adjustment, UF/DF steps so as to achieve the desired product quality, ion concentration, and/or viral reduction.
  • the instant invention provides methods for producing a process-related impurity and/or product-related substance-reduced protein preparation from a mixture comprising a protein of interest (i.e., a product) and at least one process-related impurity and/or product-related substance by subjecting the mixture to at least one cation exchange separation step.
  • the CEX step will occur after the above-described Protein A affinity step.
  • a cationic exchange material versus a anionic exchange material is based on the local charges of the protein of interest in a given solution. Therefore, it is within the scope of this invention to employ a cationic exchange step prior to the use of an anionic exchange step, or an anionic exchange step prior to the use of a cationic exchange step. Furthermore, it is within the scope of this invention to employ only a cationic exchange step, only an anionic exchange step, or any serial combination of the two (including serial combinations of one or both ion exchange steps with the other chromatographic separation technologies described herein).
  • the initial protein mixture can be contacted with the cation exchange material by using any of a variety of techniques, e.g., using a batch purification technique or a chromatographic technique, as described above in connection with Protein A or AEX.
  • the aqueous salt solution used as both the loading and wash buffer has a pH that is lower than the isoelectric point (pI) of the protein of interest.
  • the pH is about 0 to 5 units lower than the pI of the protein. In certain embodiments, it is in the range of 1 to 2 units lower. In certain embodiments, it is in the range of 1 to 1.5 units lower.
  • the concentration of the anionic agent in aqueous salt solution is increased or decreased to achieve a pH of between about 3.5 and 10.5, or between about 4 and 10, or between about 4.5 and 9.5, or between about 5 and 9, or between about 5.5 and 8.5, or between about 6 and 8, or between about 6.5 and 7.5. In certain embodiments, the concentration of anionic agent is increased or decreased in the aqueous salt solution to achieve a pH of 5, or 5.5, or 6, or 6.5, or 6.8, or 7.5.
  • the conductivity and pH of the aqueous salt solution is adjusted by increasing or decreasing the concentration of a cationic agent.
  • the cationic agent is maintained at a concentration of between about range of 20 mM to 500 mM, or between about 50 to 350 mM or between about 100 to 300 mM or between about 100 to 200 mM.
  • the cationic agent is selected from the group consisting of sodium, Tris, tromethalmine, ammonium, arginine, or combinations thereof.
  • the anionic agent is selected from the group consisting of acetate, citrate, chloride anion, sulphate, phosphate or combinations thereof.
  • a packed cation-exchange chromatography column or a cation-exchange membrane device can be operated either in bind-elute mode, flow-through mode, or a hybrid mode wherein the product exhibits binding to the chromatographic material, yet can be washed from the column using a buffer that is the same or substantially similar to the loading buffer.
  • the details of these modes are outlined above.
  • Cationic substituents include carboxymethyl (CM), sulfoethyl (SE), sulfopropyl (SP), phosphate (P) and sulfonate (S).
  • Additional cationic materials include, but are not limited to: Capto SP ImpRes, which is a high flow agarose bead; CM Hyper D grade F; which is a ceramic bead coated and permeated with a functionalized hydrogel, 250-400 ionic groups ⁇ eq/mL; Eshmuno S, which is a hydrophilic polyvinyl ether base matrix with 50-100 ⁇ eq/mL ionic capacity; Nuvia C Prime, which is a hydrophobic cation exchange media composed of a macroporous highly crosslinked hydrophilic polymer matrix 55-75 ⁇ eq/mL; Nuvia S, which has a UNOsphere base matrix with 90-150 ⁇ eq/mL ionic groups; Poros HS; which is a rigid polym
  • the protein load of the mixture comprising protein of interest is adjusted to a total protein load to the column of between about 5 and 150 g/L, or between about 10 and 100 g/L, between about 20 and 80 g/L, or between about 30 and 50 g/L.
  • the protein concentration of the load protein mixture is adjusted to a protein concentration of the material loaded to the column of about 0.5 and 50 g/L, or between about 1 and 20 g/L.
  • additives such as poly ethylene glycol, detergents, amino acids, sugars, chaotropic agents can be added to enhance the performance of the separation, so as to achieve better recovery or product quality.
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR charge variants in the elution fractions while enriching for the same in the flow through and wash fractions, thereby producing protein preparations with reduced or free of AR variants.
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR1 charge variants in the elution fractions while enriching for the same in the flow through and wash fractions, thereby producing protein preparations with reduced or free of AR1 variants.
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR2 charge variants in the elution fractions while enriching for the same in the flow through and wash fractions, thereby producing protein preparations with reduced or free of AR2 variants.
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of the Methyl Glycoxol (MGO) variants in the elution fractions while enriching for the same in the flow through and wash fractions, thereby producing protein preparations with reduced or free of MGO variants.
  • MGO Methyl Glycoxol
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of the glycated variants (schiff's base and permanently glycated forms) in the elution fractions while enriching for the same in the flow through and wash fractions, thereby producing protein preparations with reduced or free of glycated variants.
  • the loading, pH, conductivity of the CEX chromatography step, as well as elution pH conductivity can be modified to achieve a desired distribution of process-related impurities and/or product-relates substances.
  • certain embodiments are directed to the modulation of the lysine distribution of a purified sample of a protein of interest, e.g., increasing Lys0 and decreasing Lys1 and Lys2.
  • the methods of the present invention allow for the preparation of samples wherein the amount of Lys0 is decreased, while the amount of Lys 1 and/or Lys2 is increased.
  • a CEX chromatographic separation can be performed and combinations of fractions can be pooled to achieve a combination of desired process-related impurity and/or product-relates substance levels, in addition to, or in place of merely modulating charge variant concentration.
  • spectroscopy methods such as UV, NIR, FTIR, Fluorescence, Raman may be used to monitor levels of product-related charge variants, aggregates, low molecular weight variants (e.g., fragments of the protein of interest) in an on-line, at-line or in-line mode, which can then be used to control the level of charge variants, e.g., acidic species, in the pooled material collected from the CEX effluent.
  • specific signals arising from the chemical modification of the proteins such as glycation, MGO modification, deamidation, glycosylation may be specifically measurable by spectroscopic methods through such in-line, on-line or at-line methods, enabling realtime or near-real time control of product quality of the resulting product.
  • on-line, at-line or in-line monitoring methods can be used either on the effluent line of the chromatography step or in the collection vessel, to enable achievement of the desired product quality/recovery.
  • the UV signal can be used as a surrogate to achieve an appropriate product quality/recovery, wherein the UV signal can be processed appropriately, including, but not limited to, such processing techniques as integration, differentiation, moving average, such that normal process variability can be addressed and the target product quality can be achieved.
  • processing techniques as integration, differentiation, moving average, such that normal process variability can be addressed and the target product quality can be achieved.
  • such measurements can be combined with in-line dilution methods such that ion concentration/conductivity of the load/wash can be controlled by feedback and hence facilitate product quality control.
  • a combination of CEX and AEX and MM methods can be used to prepare product-related substance-modulated materials, including certain embodiments where one technology is used in a complementary/supplementary manner with another technology.
  • such a combination can be performed such that certain sub-species are removed predominantly by one technology, such that the combination provides the desired final composition/product quality.
  • such combinations include the use of additional intervening chromatography, filtration, pH adjustment, UF/DF steps so as to achieve the desired product quality, ion concentration, and/or viral reduction.
  • MM Mixed mode
  • multimodal chromatography is a chromatographic strategy that utilizes a support comprising a ligand that is capable of providing at least two different, in certain embodiments co-operative, sites that interact with the substance to be bound. In certain embodiments, one of these sites gives an attractive type of charge-charge interaction between the ligand and the substance of interest and the other site provides for electron acceptor-donor interaction and/or hydrophobic and/or hydrophilic interactions. Electron donor-acceptor interactions include interactions such as hydrogen-bonding, ⁇ - ⁇ , cation- ⁇ , charge transfer, dipole-dipole, induced dipole etc.
  • the resin employed for a mixed mode separation is Capto Adhere.
  • Capto Adhere is a strong anion exchanger with multimodal functionality. Its base matrix is a highly cross-linked agarose with a ligand (N-Benzyl-N-methyl ethanol amine) that exhibits many functionalities for interaction, such as ionic interaction, hydrogen bonding and hydrophobic interaction.
  • the resin employed for a mixed mode separation is selected from PPA-HyperCel and HEA-HyperCel.
  • the base matrices of PPA-HyperCel and HEA-HyperCel are high porosity cross-linked cellulose.
  • Phenylpropylamine and Hexylamine are Phenylpropylamine and Hexylamine, respectively. Phenylpropylamine and Hexylamine offer different selectivity and hydrophobicity options for protein separations. Additional mixed mode chromatographic supports include, but are not limited to, Nuvia C Prime, Toyo Pearl MX Trp 650M, and Eshmuno® HCX.
  • the mixed mode chromatography resin is comprised of ligands coupled to an organic or inorganic support, sometimes denoted a base matrix, directly or via a spacer.
  • the support may be in the form of particles, such as essentially spherical particles, a monolith, filter, membrane, surface, capillaries, etc.
  • the support is prepared from a native polymer, such as cross-linked carbohydrate material, such as agarose, agar, cellulose, dextran, chitosan, konjac, carrageenan, gellan, alginate etc.
  • the support can be porous, and ligands are then coupled to the external surfaces as well as to the pore surfaces.
  • Such native polymer supports can be prepared according to standard methods, such as inverse suspension gelation (S Hjerten: Biochim Biophys Acta 79(2), 393-398 (1964).
  • the support can be prepared from a synthetic polymer, such as cross-linked synthetic polymers, e.g. styrene or styrene derivatives, divinylbenzene, acrylamides, acrylate esters, methacrylate esters, vinyl esters, vinyl amides etc.
  • Such synthetic polymers can be produced according to standard methods, see e.g. “Styrene based polymer supports developed by suspension polymerization” (R Arshady: Chimica e L'Industria 70(9), 70-75 (1988)).
  • Porous native or synthetic polymer supports are also available from commercial sources, such as Amersham Biosciences, Uppsala, Sweden.
  • the protein load of the mixture comprising protein of interest is adjusted to a total protein load to the column of between about 50 and 750 g/L, or between about 75 and 500 g/L, or between about 100 and 300 g/L.
  • the protein concentration of the load protein mixture is adjusted to a protein concentration of the material loaded to the column of about 1 and 50 g/L, or between about 9 and 25 g/L.
  • additives such as poly ethylene glycol, detergents, amino acids, sugars, chaotropic agents can be added to enhance the performance of the separation, so as to achieve better recovery or product quality.
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR charge variants in the flow through and wash fractions while enriching for the same in the flow elution fraction, thereby producing protein preparations with reduced or free of AR variants.
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR1 charge variants in the flow through and wash fractions while enriching for the same in the flow elution fraction, thereby producing protein preparations with reduced or free of AR1 variants.
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR2 charge variants in the flow-through and wash fractions while enriching for the same in the flow elution fraction, thereby producing protein preparations with reduced or free of AR2 variants.
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of the methylglyoxal (MGO) variants in the flow through and wash fractions while enriching for the same in the elution fraction, thereby producing protein preparations with reduced or free of MGO variants.
  • MGO methylglyoxal
  • the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of the glycated variants (schiff's base and permanently glycated forms) in the flow through and wash fractions while enriching for the same in the elution fraction, thereby producing protein preparations with reduced or free of glycated variants.
  • the loading, pH, conductivity of the MM chromatography step, wash pH and conductivity, as well as elution pH conductivity can be modified to achieve a desired distribution of process-related impurities and/or product-relates substances.
  • certain embodiments are directed to the modulation of the lysine distribution of a purified sample of a protein of interest, e.g., increasing Lys0 and decreasing Lys1 and Lys2.
  • the methods of the present invention allow for the preparation of samples wherein the amount of Lys0 is decreased, while the amount of Lys 1 and/or Lys2 is increased.
  • a MM chromatographic separation can be performed and combinations of fractions can be pooled to achieve a combination of desired process-related impurity and/or product-relates substance levels, in addition to, or in place of merely modulating charge variant concentration.
  • spectroscopy methods such as UV, NIR, FTIR, Fluorescence, Raman may be used to monitor levels of product-related charge variants, aggregates, low molecular weight variants (e.g., fragments of the protein of interest) in an on-line, at-line or in-line mode, which can then be used to control the level of charge variants, e.g., acidic species, in the pooled material collected from the MM effluent.
  • specific signals arising from the chemical modification of the proteins such as glycation, MGO modification, deamidation, glycosylation may be specifically measurable by spectroscopic methods through such in-line, on-line or at-line methods, enabling realtime or near-real time control of product quality of the resulting product.
  • on-line, at-line or in-line monitoring methods can be used either on the effluent line of the chromatography step or in the collection vessel, to enable achievement of the desired product quality/recovery.
  • the UV signal can be used as a surrogate to achieve an appropriate product quality/recovery, wherein the UV signal can be processed appropriately, including, but not limited to, such processing techniques as integration, differentiation, moving average, such that normal process variability can be addressed and the target product quality can be achieved.
  • processing techniques as integration, differentiation, moving average, such that normal process variability can be addressed and the target product quality can be achieved.
  • such measurements can be combined with in-line dilution methods such that ion concentration/conductivity of the load/wash can be controlled by feedback and hence facilitate product quality control
  • a combination of mixed mode and AEX and CEX methods can be used to prepare product-related charge variant-reduced materials, including certain embodiments where one technology is used in a complementary/supplementary manner with another technology.
  • such a combination can be performed such that certain sub-species are removed predominantly by one technology, such that the combination provides the desired final composition/product quality.
  • such combinations include the use of additional intervening chromatography, filtration, pH adjustment, UF/DF steps so as to achieve the desired product quality, ion concentration, and/or viral reduction.
  • the present invention also features methods for producing a process-related impurity and/or product-related substance-reduced protein preparation from a mixture comprising a protein of interest, e.g., an antibody, and at least one process-related impurity and/or product-related substance further comprising a hydrophobic interaction chromatography (HIC) step in addition to the displacement chromatography step.
  • a protein of interest e.g., an antibody
  • HIC hydrophobic interaction chromatography
  • the sample mixture is contacted with the HIC material, e.g., using a batch purification technique or using a column or membrane chromatography.
  • HIC purification it may be desirable to adjust the concentration of the salt buffer to achieve desired protein binding to the resin or the membrane.
  • hydrophobic interaction chromatography employs the hydrophobic properties of the proteins to achieve selective separation. Hydrophobic groups on the protein interact with hydrophobic groups of the resin or the membrane. The more hydrophobic a protein is the stronger it will interact with the column or the membrane. Thus the HIC step removes process-related impurities (e.g., HCPs) as well as product-related substances (e.g., aggregates and fragments).
  • process-related impurities e.g., HCPs
  • product-related substances e.g., aggregates and fragments.
  • a HIC column or membrane device can also be operated in product a bind-elute mode, a flow-through, or a hybrid mode wherein the product exhibits binding to the chromatographic material, yet can be washed from the column using a buffer that is the same or substantially similar to the loading buffer.
  • a buffer that is the same or substantially similar to the loading buffer.
  • this form of separation is conveniently performed following salt elution step, such as those that are typically used in connection with ion exchange chromatography.
  • salts can be added into a low salt level feed stream before this step.
  • Adsorption of the antibody to a HIC column is favored by high salt concentrations, but the actual concentrations can vary over a wide range depending on the nature of the protein of interest, salt type and the particular HIC ligand chosen.
  • Various ions can be arranged in a so-called soluphobic series depending on whether they promote hydrophobic interactions (salting-out effects) or disrupt the structure of water (chaotropic effect) and lead to the weakening of the hydrophobic interaction.
  • Cations are ranked in terms of increasing salting out effect as Ba 2+ ; Ca 2+ ; Mg 2+ ; Li + ; Cs + ; Na + ; K + ; Rb + ; NH 4 + , while anions may be ranked in terms of increasing chaotropic effect as PO 4 3 ⁇ ; SO 4 2 ⁇ ; CH 3 CO 3 ⁇ ; Cl ⁇ ; Br ⁇ ; NO 3 ⁇ ; ClO 4 ⁇ ; I ⁇ ; SCN ⁇ .
  • Na + , K + or NH 4 + sulfates effectively promote ligand-protein interaction in HIC.
  • Salts may be formulated that influence the strength of the interaction as given by the following relationship: (NH 4 ) 2 SO 4 >Na 2 SO 4 >NaCl>NH 4 Cl>NaBr>NaSCN.
  • salt concentrations of between about 0.75 M and about 2 M ammonium sulfate or between about 1 and 4 M NaCl are useful.
  • HIC media normally comprise a base matrix (e.g., cross-linked agarose or synthetic copolymer material) to which hydrophobic ligands (e.g., alkyl or aryl groups) are coupled.
  • a suitable HIC media comprises an agarose resin or a membrane functionalized with phenyl groups (e.g., a Phenyl SepharoseTM from GE Healthcare or a Phenyl Membrane from Sartorius). Many HIC resins are available commercially.
  • Examples include, but are not limited to, Capto Phenyl, Phenyl SepharoseTM 6 Fast Flow with low or high substitution, Phenyl SepharoseTM High Performance, Octyl SepharoseTM High Performance (GE Healthcare); FractogelTM EMD Propyl or FractogelTM EMD Phenyl (E. Merck, Germany); Macro-PrepTM Methyl or Macro-PrepTM t-Butyl columns (Bio-Rad, California); WP HI-Propyl (C3)TM (J. T. Baker, New Jersey); and ToyopearlTM ether, phenyl or butyl (TosoHaas, Pa.).
  • Viral filtration is a dedicated viral reduction step in the entire purification process. This step is usually performed post chromatographic polishing steps. Viral reduction can be achieved via the use of suitable filters including, but not limited to, Planova 20NTM, 50 N or BioEx from Asahi Kasei Pharma, ViresolveTM filters from EMD Millipore, ViroSart CPV from Sartorius, or Ultipor DV20 or DV50TM filter from Pall Corporation. It will be apparent to one of ordinary skill in the art to select a suitable filter to obtain desired filtration performance.
  • suitable filters including, but not limited to, Planova 20NTM, 50 N or BioEx from Asahi Kasei Pharma, ViresolveTM filters from EMD Millipore, ViroSart CPV from Sartorius, or Ultipor DV20 or DV50TM filter from Pall Corporation. It will be apparent to one of ordinary skill in the art to select a suitable filter to obtain desired filtration performance.
  • Certain embodiments of the present invention employ ultrafiltration and diafiltration steps to further concentrate and formulate the protein of interest, e.g., an antibody product.
  • Ultrafiltration is described in detail in: Microfiltration and Ultrafiltration: Principles and Applications, L. Zeman and A. Zydney (Marcel Dekker, Inc., New York, N.Y., 1996); and in: Ultrafiltration Handbook, Munir Cheryan (Technomic Publishing, 1986; ISBN No. 87762-456-9).
  • One filtration process is Tangential Flow Filtration as described in the Millipore catalogue entitled “Pharmaceutical Process Filtration Catalogue” pp. 177-202 (Bedford, Mass., 1995/96).
  • Ultrafiltration is generally considered to mean filtration using filters with a pore size of smaller than 0.1 ⁇ m.
  • filters having such small pore size, the volume of the sample can be reduced through permeation of the sample buffer through the filter membrane pores while proteins, such as antibodies, are retained above the membrane surface.
  • Diafiltration is a method of using membrane filters to remove and exchange salts, sugars, and non-aqueous solvents, to separate free from bound species, to remove low molecular-weight species, and/or to cause the rapid change of ionic and/or pH environments.
  • Microsolutes are removed most efficiently by adding solvent to the solution being diafiltered at a rate approximately equal to the permeate flow rate. This washes away microspecies from the solution at a constant volume, effectively purifying the retained protein of interest.
  • a diafiltration step is employed to exchange the various buffers used in connection with the instant invention, optionally prior to further chromatography or other purification steps, as well as to remove impurities from the protein preparations.
  • membrane cassettes suitable for the present invention include, but not limited to, Pellicon 2 or Pellicon 3 cassetts with 10 kD, 30 kD or 50 kD membranes from EMD Millipore, Kvick 10 kD, 30 kD or 50 kD membrane cassettes from GE Healthcare, and Centramate or Centrasette 10 kD, 30 kD or 50 kD cassettes from Pall Corporation.
  • primary recovery can proceed by sequentially employing pH reduction, centrifugation, and filtration steps to remove cells and cell debris (including HCPs) from the production bioreactor harvest.
  • the present invention is directed to subjecting a sample mixture from said primary recovery to one or more AEX, CEX, and/or MM purification steps. Certain embodiments of the present invention will include further purification steps.
  • Examples of additional purification procedures which can be performed prior to, during, or following the ion exchange chromatography method include ethanol precipitation, isoelectric focusing, reverse phase HPLC, chromatography on silica, chromatography on heparin SepharoseTM, further anion exchange chromatography and/or further cation exchange chromatography, chromatofocusing, SDS-PAGE, ammonium sulfate precipitation, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography (e.g., using protein G, an antibody, a specific substrate, ligand or antigen as the capture reagent).
  • affinity chromatography e.g., using protein G, an antibody, a specific substrate, ligand or antigen as the capture reagent.
  • the unbound flow through and wash fractions can be further fractionated and a combination of fractions providing a target product purity can be pooled.
  • the protein concentration can be adjusted to achieve a differential partitioning behavior between the antibody product and the product-related substances such that the purity and/or yield can be further improved.
  • the loading can be performed at different protein concentrations during the loading operation to improve the product quality/yield of any particular purification step.
  • the column temperature can be independently varied to improve the separation efficiency and/or yield of any particular purification step.
  • the loading and washing buffer matrices can be different or composed of mixtures of chemicals, while achieving similar “resin interaction” behavior such that the above novel separation can be effected.
  • the loading and washing buffers can be different, in terms of ionic strength or pH, while remaining substantially similar in function in terms of the washout of the product achieved during the wash step.
  • additives such as amino acids, sugars, PEG, etc can be added to the load or wash steps to modulate the partitioning behavior to achieve the separation efficiency and/or yield.
  • the loading & washing steps can be controlled by in-line, at-line or off-line measurement of the product related impurity/substance levels, either in the column effluent, or the collected pool or both, so as to achieve the target product quality and/or yield.
  • the loading concentration can be dynamically controlled by in-line or batch or continuous dilutions with buffers or other solutions to achieve the partitioning necessary to improve the separation efficiency and/or yield.
  • the present invention also provides methods for determining the residual levels of host cell protein (HCP) concentration in the isolated/purified antibody composition.
  • HCPs are desirably excluded from the final target substance product.
  • Exemplary HCPs include proteins originating from the source of the antibody production. Failure to identify and sufficiently remove HCPs from the target antibody may lead to reduced efficacy and/or adverse subject reactions.
  • HCP ELISA refers to an ELISA where the second antibody used in the assay is specific to the HCPs produced from cells, e.g., CHO cells, used to generate the antibody of interest.
  • the second antibody may be produced according to conventional methods known to those of skill in the art.
  • the second antibody may be produced using HCPs obtained by sham production and purification runs, i.e., the same cell line used to produce the antibody of interest is used, but the cell line is not transfected with antibody DNA.
  • the second antibody is produced using HCPs similar to those expressed in the cell expression system of choice, i.e., the cell expression system used to produce the target antibody.
  • HCP ELISA comprises sandwiching a liquid sample comprising HCPs between two layers of antibodies, i.e., a first antibody and a second antibody.
  • the sample is incubated during which time the HCPs in the sample are captured by the first antibody, for example, but not limited to goat anti-CHO, affinity purified ( Cygnus ).
  • the first and second antibodies are polyclonal antibodies.
  • the first and second antibodies are blends of polyclonal antibodies raised against HCPs. The amount of HCP contained in the sample is determined using the appropriate test based on the label of the second antibody.
  • HCP ELISA may be used for determining the level of HCPs in an antibody composition, such as an eluate or flow-through obtained using the process described above.
  • the present invention also provides a composition comprising an antibody, wherein the composition has no detectable level of HCPs as determined by an HCP Enzyme Linked Immunosorbent Assay (“ELISA”).
  • ELISA HCP Enzyme Linked Immunosorbent Assay
  • the levels of acidic species and other charge variants in the chromatographic samples produced using the techniques described herein are analyzed.
  • a CEX-HPLC method is employed. For example, but not by way of limitation, cation exchange chromatography can be performed on a Dionex ProPac WCX-10, Analytical column 4 mm ⁇ 250 mm (Dionex, Calif.). An Agilent 1200 HPLC system can then be used as the HPLC.
  • mobile phases such as 10 mM Sodium Phosphate dibasic pH 7.5 (Mobile phase A) and 10 mM Sodium Phosphate dibasic, 500 mM Sodium Chloride pH 5.5 (Mobile phase B) can be used.
  • a binary gradient (94% A, 6% B: 0-20 min; 84% A, 16% B: 20-22 min; 0% A, 100% B: 22-28 min; 94% A, 6% B: 28-34 min) can be used with detection at 280 nm.
  • quantitation is based on the relative area percent of detected peaks.
  • the peaks that elute at relative residence time less than a certain time are together represented as the acidic peaks.
  • the levels of aggregates, monomer, and fragments in the chromatographic samples produced using the techniques described herein are analyzed.
  • the aggregates, monomer, and fragments are measured using a size exclusion chromatographic (SEC) method for each molecule.
  • SEC size exclusion chromatographic
  • a TSK-gel G3000SWxL, 5 ⁇ m, 125 ⁇ , 7.8 ⁇ 300 mm column can be used in connection with certain embodiments
  • a TSK-gel Super SW3000, 4 ⁇ m, 250 ⁇ , 4.6 ⁇ 300 mm column can be used in alternative embodiments.
  • sample injections are made under isocratic elution conditions using a mobile phase consisting of, for example, 100 mM sodium sulfate and 100 mM sodium phosphate at pH 6.8, and detected with UV absorbance at 214 nm.
  • the mobile phase will consist of 1 ⁇ PBS at pH 7.4, and elution profile detected with UV absorbance at 280 nm.
  • quantification is based on the relative area of detected peaks.
  • the purified proteins, e.g., antibodies and antibody-binding portions thereof, of the present invention can be modified.
  • the antibodies are chemically modified to provide a desired effect.
  • pegylation of antibodies or antibody fragments of the invention may be carried out by any of the pegylation reactions known in the art, as described, e.g., in the following references: Focus on Growth Factors 3:4-10 (1992); EP 0 154 316; and EP 0 401 384, each of which is incorporated by reference herein in its entirety.
  • the pegylation is carried out via an acylation reaction or an alkylation reaction with a reactive polyethylene glycol molecule (or an analogous reactive water-soluble polymer).
  • a suitable water-soluble polymer for pegylation of the antibodies and antibody fragments of the invention is polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • polyethylene glycol is meant to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (Cl—ClO) alkoxy- or aryloxy-polyethylene glycol.
  • Methods for preparing pegylated antibodies and antibody fragments of the invention will generally comprise the steps of (a) reacting the antibody or antibody fragment with polyethylene glycol, such as a reactive ester or aldehyde derivative of PEG, under suitable conditions whereby the antibody or antibody fragment becomes attached to one or more PEG groups, and (b) obtaining the reaction products.
  • polyethylene glycol such as a reactive ester or aldehyde derivative of PEG
  • the pegylated antibodies and antibody fragments have increased half-life, as compared to the nonpegylated antibodies and antibody fragments.
  • the pegylated antibodies and antibody fragments may be employed alone, together, or in combination with other pharmaceutical compositions.
  • an antibody of the invention can be derivatized or linked to another functional molecule (e.g., another peptide or protein).
  • another functional molecule e.g., another peptide or protein
  • an antibody of the invention can be functionally linked (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate associate of the antibody with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • One type of derivatized antibody is produced by crosslinking two or more antibodies (of the same type or of different types, e.g., to create bispecific antibodies).
  • Suitable crosslinkers include those that are heterobifunctional, having two distinctly reactive groups separated by an appropriate spacer (e.g., m-maleimidobenzoyl-N-hydroxysuccinimide ester) or homobifunctional (e.g., disuccinimidyl suberate).
  • Such linkers are available from Pierce Chemical Company, Rockford, Ill.
  • Useful detectable agents with which an antibody of the invention may be derivatized include fluorescent compounds.
  • Exemplary fluorescent detectable agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-1-napthalenesulfonyl chloride, phycoerythrin and the like.
  • An antibody may also be derivatized with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, glucose oxidase and the like. When an antibody is derivatized with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product.
  • the detectable agent horseradish peroxidase when the detectable agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable.
  • An antibody may also be derivatized with biotin, and detected through indirect measurement of avidin or streptavidin binding.
  • the proteins of interest e.g., antibodies and antibody-binding portions thereof, of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject.
  • the pharmaceutical composition comprises an antibody of the invention and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof.
  • isotonic agents e.g., sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition.
  • Pharmaceutically acceptable carriers may further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody.
  • the antibodies and antibody-binding portions thereof, of the invention can be incorporated into a pharmaceutical composition suitable for parenteral administration.
  • the antibody or antibody-portions can be prepared as an injectable solution containing, e.g., 0.1-250 mg/mL antibody.
  • the injectable solution can be composed of either a liquid or lyophilized dosage form in a flint or amber vial, ampule or pre-filled syringe.
  • the buffer can be L-histidine approximately 1-50 mM, (optimally 5-10 mM), at pH 5.0 to 7.0 (optimally pH 6.0).
  • Other suitable buffers include but are not limited to sodium succinate, sodium citrate, sodium phosphate or potassium phosphate.
  • Sodium chloride can be used to modify the toxicity of the solution at a concentration of 0-300 mM (optimally 150 mM for a liquid dosage form).
  • Cryoprotectants can be included for a lyophilized dosage form, principally 0-10% sucrose (optimally 0.5-1.0%).
  • Other suitable cryoprotectants include trehalose and lactose.
  • Bulking agents can be included for a lyophilized dosage form, principally 1-10% mannitol (optimally 24%).
  • Stabilizers can be used in both liquid and lyophilized dosage forms, principally 1-50 mM L-methionine (optimally 5-10 mM).
  • Other suitable bulking agents include glycine, arginine, can be included as 0-0.05% polysorbate-80 (optimally 0.005-0.01%).
  • Additional surfactants include but are not limited to polysorbate 20 and BRIJ surfactants.
  • the pharmaceutical composition includes the antibody at a dosage of about 0.01 mg/kg-10 mg/kg.
  • the dosages of the antibody include approximately 1 mg/kg administered every other week, or approximately 0.3 mg/kg administered weekly. A skilled practitioner can ascertain the proper dosage and regime for administering to a subject.
  • compositions of this invention may be in a variety of forms. These include, e.g., liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories.
  • liquid solutions e.g., injectable and infusible solutions
  • dispersions or suspensions tablets, pills, powders, liposomes and suppositories.
  • Typical compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with other antibodies.
  • One mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular).
  • the antibody is administered by intravenous infusion or injection.
  • the antibody is administered by intramuscular or subcutaneous injection.
  • compositions typically must be sterile and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration.
  • Sterile injectable solutions can be prepared by incorporating the active compound (i.e., antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the methods of preparation are vacuum drying and spray-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the proper fluidity of a solution can be maintained, e.g., by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, e.g., monostearate salts and gelatin.
  • the antibodies and antibody-binding portions thereof, of the present invention can be administered by a variety of methods known in the art, one route/mode of administration is subcutaneous injection, intravenous injection or infusion. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results.
  • the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • a controlled release formulation including implants, transdermal patches, and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York,
  • an antibody or antibody-binding portion thereof, of the invention may be orally administered, e.g., with an inert diluent or an assimilable edible carrier.
  • the compound (and other ingredients, if desired) may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet.
  • the compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • To administer a compound of the invention by other than parenteral administration it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation.
  • Supplementary active compounds can also be incorporated into the compositions.
  • an antibody or antibody-binding portion thereof, of the invention is co-formulated with and/or co-administered with one or more additional therapeutic agents that are useful for treating disorders.
  • Such combination therapies may advantageously utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies. It will be appreciated by the skilled practitioner that when the antibodies of the invention are used as part of a combination therapy, a lower dosage of antibody may be desirable than when the antibody alone is administered to a subject (e.g., a synergistic therapeutic effect may be achieved through the use of combination therapy which, in turn, permits use of a lower dose of the antibody to achieve the desired therapeutic effect).
  • the antibodies of the invention can be used alone or in combination with an additional agent, e.g., a therapeutic agent, said additional agent being selected by the skilled artisan for its intended purpose.
  • the additional agent can be a therapeutic agent art-recognized as being useful to treat the disease or condition being treated by the antibody of the present invention.
  • the additional agent also can be an agent which imparts a beneficial attribute to the therapeutic composition, e.g., an agent which affects the viscosity of the composition.
  • Pre-packed resin columns were used in the following experiments, except where specified.
  • the column was equilibrated in a buffer system with appropriate pH and conductivity.
  • the column load was prepared from Protein A affinity chromatography eluates or concentrated CEX chromatography elutes by buffer exchange (if the eluates were with different buffer components from the mixed mode target buffer system) or addition of the stock solutions and/or water to obtain the target pH and conductivity as specified (if the eluates were with the same buffer components as the mixed mode target buffer system).
  • the prepared load material was filtered and loaded on the column according to the target load amount (g protein/L resin) as specified followed by washing with the equilibration buffer or buffer similar to equilibration buffer with volumes as specified.
  • the column Flow Through/Wash were collected as fractions or as a pool.
  • Mixed mode column was regenerated with 0.1M acetic acid, 0.15M NaCl pH3, or 0.1M Acetic acid solution, pH 3, or as specified. 1M NaOH solution was used for column cleaning.
  • Buffers were prepared targeting specific ion concentration for the anion by fixing the anion concentration (acid) to the target value, and adjusting the solution with the cationic component (base) to achieve the appropriate pH.
  • anion concentration an anion concentration
  • base a cationic component
  • the Flow Through/Wash fractions were collected and analyzed with WCX-10 method for AR levels. By actual or calculated pooling of the fractions the recovery and the corresponding AR levels were calculated.
  • the acidic species and other charge variants present in the Adalimumab process samples were quantified according to the following methods. Cation exchange chromatography was performed on a Dionex ProPac WCX-10, Analytical column 4 mm ⁇ 250 mm (Dionex, CA). An Agilent 1200 HPLC system was used as the HPLC. The mobile phases used were 10 mM Sodium Phosphate dibasic pH 7.5 (Mobile phase A) and 10 mM Sodium Phosphate dibasic, 500 mM Sodium Chloride pH 5.5 (Mobile phase B).
  • Quantitation was based on the relative area percent of detected peaks.
  • the peaks that elute at relative residence time less than a certain time are together represented as the acidic peaks.
  • the acidic species and other charge variants present in the mAb-B process samples were quantified according to the following methods. Cation exchange chromatography was performed on a Dionex ProPac WCX-10, Analytical column 4 mm ⁇ 250 mm (Dionex, CA). An Agilent 1200 HPLC system was used as the HPLC. The mobile phases used were 20 mM 4-Morpholineethanesulfonic acid (MES), pH 6.5 (Mobile phase A) and 20 mM 4-Morpholineethanesulfonic acid (MES), 500 mM Sodium Chloride pH 6.5 (Mobile phase B).
  • MES 4-Morpholineethanesulfonic acid
  • MES 2-Morpholineethanesulfonic acid
  • a binary gradient (87% A, 13% B: 0-5 min; 87% A, 13% B: 5-35 min; 75% A, 25% B: 35-40 min; 0% A, 100% B: 40-43 min; 87% A, 13% B: 43-46 min; 87% A, 13% B: 46-55 min) was used with detection at 280 nm, bw 8 nm; ref 360 nm, bw 100 nm.
  • Quantitation was based on the relative area percent of detected peaks. All peaks eluting prior to the Main Isoform peak were summed as the acidic region, and all peaks eluting after the LYS-2 peaks will be summed as the basic region.
  • the mAb-C method was employed towards the quantification of the acidic species and other charge variants present mAb-C process samples.
  • Cation exchange chromatography was performed on a Dionex ProPac WCX-10, Analytical column 4 mm ⁇ 250 mm (Dionex, Calif.).
  • An Agilent 1200 HPLC system was used as the HPLC.
  • the mobile phases used were 20 mM 4-Morpholineethanesulfonic acid (MES), pH 6.0 (Mobile phase A) and 20 mM 4-Morpholineethanesulfonic acid (MES), 250 mM Sodium Chloride pH 6.0 (Mobile phase B).
  • a binary gradient (97% A, 3% B: 0-1 min; 79% A, 21% B: 1-46 min; 0% A, 100% B: 46-47 min; 0% A, 100% B: 47-52 min; 97% A, 3% B: 52-53 min; 97% A, 3% B: 53-60 min) was used with detection at 280 nm, bw 8 nm; ref 360 nm, bw 100 nm.
  • Quantitation was based on the relative area percent of detected peaks. All peaks eluting prior to the Main Isoform peak will be summed as the acidic region, and all peaks eluting after the Main Isoform peak will be summed as the basic region.
  • the molecular weight distribution of collected samples were quantified according to the following methods. Size exclusion chromatography (SEC) was performed using a TSK-gel G3000SWxL, 5 ⁇ m, 125 ⁇ , 7.8 ⁇ 300 mm column (Tosoh Bioscience) on an HP Agilent HPLC system. Injections were made under isocratic elution conditions using a mobile phase of 200 mM sodium sulfate, 100 mM sodium phosphate, pH 6.8, and detected with absorbance at 214 nm. Quantification is based on the relative area of detected peaks.
  • SEC Size exclusion chromatography
  • HCP Host Cell Protein
  • HCP assay is based on process specific antigen based ELISA. Sample dilutions were applied to achieve readings within the calibration range. The limit of quantitation of the assay is 0.625 ng/mL.
  • UV A280 was used to determine protein concentrations for the samples post protein A elution.
  • the assay was performed on an Agilent UV Spectrophotometer following the method.
  • the absorbance was taken at 280 nm, the path length was 1 cm, and the extinction coefficients were 1.39 for Adalimumab, 1.38 for mAb B, and 1.43 for mAb C.
  • adalimumab and Poros 50PI were chosen.
  • the experiment was performed at acetate (anion) concentration of 5 mM.
  • the column was equilibrated with 5 mM acetate/Tris at a pH of 9.0.
  • Adalimumab was prepared at 5 mM acetate/Tris pH 9.0 and loaded to the column at 20 g-protein/L of resin.
  • the column was washed with 10 CVs of the equilibration buffer.
  • a pH gradient from 9.0 to 7.0 at an anion concentration of 5 mM acetate/Tris was then performed.
  • the process chromatograms are shown in FIG. 1 .
  • adalimumab and Poros 50HQ were chosen.
  • the experiment was performed at a pH 8.7.
  • the column was equilibrated with 10 mM acetate/Tris at pH 8.7.
  • Adalimumab was prepared at 10 mM acetate/Tris pH 8.7 and loaded to the column at 20 g-protein/L of resin.
  • the column was washed with 10 CVs of the equilibration buffer.
  • a linear gradient from 10-100 mM Acetate/Tris at pH 8.7 was performed.
  • the process chromatograms are shown in FIG. 2 .
  • the acidic species reduction desired can be achieved by appropriate pooling of the load and wash fractions.
  • the accumulative AR reduction and accumulative yield can be calculated using the weighted averages up to a given fraction. Additionally, the instantaneous yield can be estimated by comparing the protein recovered against the total protein loaded to the column at a given fraction. Sample calculations are shown below:
  • adalimumab and Poros 50PI were chosen.
  • the experiment was performed at 5 mM acetate/arginine pH 8.8.
  • the column was equilibrated with 5 mM acetate/arginine at pH 8.8.
  • Adalimumab was prepared at 5 mM acetate/arginine pH 8.8 and loaded to the column at 300 g-protein/L-resin.
  • the column was washed with 20 CVs of the equilibration buffer. Fractions were collected in volumes representing 30 g-protein/L-resin, shown in FIG. 3 . Each fraction was then analyzed for product quality and the accumulative yield and AR reduction calculated, shown in Table 4. From this example, it is clear to one skilled in the art to determine a run condition which delivers a targeted product quality and/or step yield.
  • This general approach is used to evaluate the performance for a given operating condition for any resin/mAb/buffer combination.
  • This data set is compiled to demonstrate the AR reduction achieved with eight different AEX adsorbents.
  • Each resin was tested using an advanced screening method using the process outlined in Example 6.1.2., and subjected to four runs using adalimumab at two different pH (e.g. pH 8.7 and 9.0) and two different acetate concentrations (e.g. 10 mM and 20 mM).
  • the instantaneous (e.g. not accumulative) AR reduction was measured by analyzing the load fraction at 150 g/L and subsequently compared across all resins. Table 6 outlines the results from these experiments.
  • AR reduction technology of the current invention has been demonstrated with multiple antibodies using AEX adsorbents.
  • Antibodies have different amount charged residues and at different positions, leading to a charge interaction behavior on an AEX column that differs from one antibody to another. Therefore the impact of anion type, anion concentration is different for each antibody.
  • Table 9 and Table 10 below show the data for MAB B and MAB C. The data clearly demonstrates that the AR reduction technology works very effectively for other antibodies.
  • the AR species in the current invention is bound during the loading step; therefore the binding pH is a key variable.
  • the anion concentration that provides the desired performance will vary with the operational pH.
  • adalimumab and Poros 50PI were chosen.
  • the experiments were performed at a concentration of 5 mM acetate/arginine at each pH specified.
  • Adalimumab was prepared at 5 mM acetate/arginine at each pH specified and loaded to the column at 300 g-protein/L of resin. The column was washed with 20 CVs of the equilibration buffer. The results showing the pH effect on AR reduction is shown in FIG. 4 .
  • Anion concentration is a key variable in the performance of anion exchange chromatography. For every combination of antibody/resin/pH there is a range of anion concentrations that provides AR reduction; the strategy outlined in Example 6.1.2. can be followed to determine the AR reduction and the corresponding recovery for each anion concentration.
  • Table 12 below shows the effect of anion concentration on AR reduction.
  • the table also includes the effect of anion concentration for different pH values.
  • the data demonstrates that the AR reduction can be effectively achieved over a range of anion concentrations at each pH and that the concentration ranges depend on the pH.
  • the anion type and concentration are key variables in Anion Exchange Chromatography.
  • the invention has been demonstrated with Acetate and Formate as the anion type and Tris and Arginine as the counter cation type.
  • the optimal pH and cation concentration is different for each cation type/mixture and was derived by using the strategy outlined in Example 6.1.2.
  • Table 13 shows the data of AR reduction and corresponding recovery for the different anion/cation types.
  • Example 6.1.2. to reduce acidic species through careful control of buffer anion type, anion concentration, AEX adsorbent, and pH can be applied to any range of protein loading.
  • a range of relevant protein loadings e.g. 100-350 g/L
  • for Poros 50HQ at pH 8.7 using Acetate as the anion is shown in Table 14, displaying a robust AR reduction across the loading range investigated.
  • Example 6.1.2. to reduce acidic species through careful control of buffer anion type, anion concentration, AEX adsorbent, and pH can be applied to any range of column feed streams of varying protein concentration.
  • a range of varying protein load concentration for a 300 g/L load of adalimumab to Poros 50HQ at 15 mM acetate/Tris pH 8.7 is shown in Table 15.
  • the operational pH relative to the pKa of the AEX adsorbent is also important as many mAbs have pI similar to the pKa of the AEX adsorbent. This effect is shown in FIG. 24 for mAb B with several different AEX adsorbents, with different pKa values, run at with an acetate/Tris buffer at pH 9.1.
  • the Acidic Region for Adalimumab is further grouped into two regions termed AR1 and AR2, based on a certain retention time of the peaks seen on the WCX-10 method.
  • the characteristics of the variants in these two regions are expected to be different and hence the methods that reduce variants belonging to these groups can be specifically delineated.
  • the current invention in addition to achieving a certain AR reduction, it may be desirable to achieve a certain absolute level of AR levels, in consideration of reducing or removing certain variants.
  • the capability of the current invention in achieving a certain absolute level of AR, AR1 and AR2 is demonstrated in Table 18.
  • the method of the current invention can effectively reduce AR2 levels, as an overall decrease in AR levels is achieved.
  • the method can be used to achieve a target absolute level, as exemplified by the data presented in Table 18.
  • Multiple species are present under the group of AR2 and that the current method of invention can be used to reduce such sub-species.
  • the method of the current invention can effectively achieve AR reduction as well as achieve a target absolute level of acidic species as exemplified by the data presented in Table 18.
  • AEX chromatography is effective in reducing aggregate and HCP levels.
  • HCP and aggregate levels can be effectively reduced under operating conditions selected for AR reduction.
  • Table 19 and Table 20 shows the aggregate and HCP removal achieved along with AR reduction. The data clearly shows that other process related and product related substances/impurities can be achieved using the current invention on the AEX adsorbents, and hence functions as an effective polishing step in the large scale purification of monoclonal antibodies.
  • Controlling the final product quality by modifying the process based on the quality of the intermediate material is an approach that has been proposed as an effective way of ensuring product quality, with the view of ensuring safety and efficacy.
  • the pH and load are parameters that can be modified to achieve a desired separation of the AR species.
  • the load to the column can be reduced.
  • the pH can be increased in order to achieve a higher reduction in AR species.
  • the AR levels can be controlled by changing the pH of the load and wash solutions as well as the total load to the column.
  • the load material and the stock buffer are both prepared at 18 mM Acetate/Tris the specified pH by titrating the affinity captured material with a stock Tris solution.
  • the AR level of the load material was the same for both runs. This experiment demonstrates how the final AR level can be modulated, while maintaining acceptable yields, by adjusting the pH and protein load to the column, shown in Table 21.
  • Example CEX 1 Determining Operating Conditions Appropriate for a Mab: Resin: Buffer Combination
  • adalimumab was chosen and Poros XS was chosen.
  • the experiments were performed at pH 6.0.
  • the process chromatograms are shown in FIG. 5 .
  • the recovery vs AR reduction curves for each of the experiments is shown in FIG. 6 and Table 22. From this set of experiments, a sodium concentration of 125 mM can be chosen and such that the recovery of the eluate is 74%, which provides an AR reduction of 5.4%. Alternately, an AR reduction value of 5.4% can be chosen which will provide a recovery of ⁇ 75%.
  • the acidic species reduction desired can be achieved by appropriate pooling of the elution fraction with the wash fractions.
  • the elution fractions can be pooled with wash fractions as shown in Table 22 to achieve AR reductions from about 1 percent to 7 percent depending on the fractions pooled. This approach can be implemented to achieve a target yield and AR reduction as exemplified in FIG. 6 .
  • Example 6.2.1 For each antibody/resin combination, the experimental strategy outlined in Example 6.2.1. was employed to determine the cation concentration for each cation type that provided AR reduction.
  • Table 24 and Table 25 below shows the data for MAB B and MAB C.
  • the data clearly demonstrates that the AR reduction technology works very effectively for other antibodies. It is also clear that the concentration ranges are different between different antibodies.
  • the pH range chosen was related to the isoelectric point of the antibody and was chosen to be approximately 1 to 2 units less than the pI of the molecule.
  • the AR species in the current invention is removed in the Flow through/Wash fraction. Therefore the binding pH is a key variable.
  • the cation concentration that provides the desired performance will vary with the binding pH. Therefore for each binding pH, the experimental strategy outlined in Example 6.2.1. is carried out to determine the range of ion concentration that results in AR reduction.
  • Cation concentration is a key variable in the performance of cation exchange chromatography. For every combination of antibody/resin/pH there is a range of cation concentrations that provides AR reduction; the strategy outlined in Example 6.2.1 can be followed to determine the AR reduction and the corresponding recovery for each cation concentration.
  • Table 28 below shows the effect of cation concentration on AR reduction.
  • the table also includes the effect of cation concentration for different pH values.
  • the data demonstrates that the AR reduction can be effectively achieved over a range of cation concentrations at each pH and that the concentration ranges depend on the pH.
  • the table also includes an example of the concentration range for a different cation type.
  • the cation type and concentration are key variables in Cation Exchange Chromatography.
  • the invention has been demonstrated with Tris, Sodium/Tris, Ammonium/Tris and Arginine/Tris as cation types/mixtures with effective reduction of AR in each case.
  • the optimal pH and cation concentration is different for each cation type/mixture and was derived by using the strategy outlined in Example 6.2.1. Experiment were performed at pH 7.5. 29 mM Tris-acetate was used for pH control. Table 29 shows the data of AR reduction and corresponding recovery for the different cation types/mixtures.
  • Example 6.2.1. to reduce acidic species through careful control of buffer cation type, concentration and pH can be applied to any range of protein loading which represents an operational mode of binding followed by elution, i.e. not overloaded or a column load factor below that of the adsorbents binding capacity.
  • a range of relevant protein loadings for Poros XS at pH 7.5 using Tris as the cation is shown in Table 30 showing robust AR reduction.
  • Example 6.2.1. to reduce acidic species through careful control of buffer cation type, concentration and pH can be applied to any range of column feed streams of varying protein concentration.
  • a range of varying protein load concentration for Poros XS at pH 7.5 using Tris as the cation is shown in Table 31 showing robust AR reduction.
  • the Acidic Region for Adalimumab is further grouped into two regions termed AR1 and AR2, based on a certain retention time of the peaks seen on the WCX-10 method.
  • the characteristics of the variants in these two regions are expected to be different and hence the methods that reduce variants belonging to these groups can be specifically delineated.
  • the specific species comprising the AR1 species can be identified and quantitated, to demonstrate reduction of such species by methods of the current invention.
  • Two of such species, Glycated mAb, and MGO modified mAb have been identified and shown to be reduced by the methods of this invention. While these species are among the Acidic Species part of the charge variants, the acidic species typically described in the literature is the deamidated mAb, which is distinctly different.
  • the method of the current invention can effectively reduce AR2 levels, as an overall decrease in AR levels is achieved.
  • the method can be used to achieve a target absolute level, as exemplified by the data presented in Table 32.
  • the method of the current invention can effectively achieve AR reduction as well as achieve a target absolute level of acidic species as exemplified by the data presented in Table 32.
  • Example 6.2.1. The strategy outlined in Example 6.2.1. to reduce acidic species through careful control of buffer cation type, concentration and pH can be further extended to specific post-translational modifications. While acidic species are defined in the application as impurities that are less retained than the main peak on an analytical weak cation exchange (WCX) HPLC column, specific known product related substances derived from cellular metabolism modification such as glycation and methylglyoxal (MGO) can be specifically identified as being part of the acidic species.
  • FIG. 7 and FIG. 8 shows the outcome of in-vitro labeling experiments which demonstrate that glycation and MGO modified antibody are unique species that are resolved by the WCX method in the AR1 region of the chromatogram and can be enriched in vitro.
  • the invention described here shows that glycated and MGO modified antibody can be effectively removed through the careful control of buffer cation type, concentration and pH using the CEX as described in Example 6.2.1. Quantitative reduction of AR1 and hence the Glycated and MGO species by CEX and CEX-Mixed Mode resins is show in Table 33 and Table 34.
  • Example 6.2.1. to reduce acidic species also can be used to modulate the distribution of C-terminal Lys variants of monoclonal antibodies, a known post-translational modification leading to charge heterogeneity.
  • Some minor changes in the distribution of Lys isoforms is expected through the reduction of acidic species as the WCX analysis is a compositional analysis.
  • the elution pool can be enriched for the more basic isoforms (Lys 1 and Lys2).
  • Table 35 and FIG. 9 depicts a non-limited example of the impact of pH and cation (Tris) concentration on basic isoform enrichment.
  • HCP and aggregate levels can be effectively reduced by appropriate adjustment of the elution conditions, after washing off the AR enriched species in the flow through/wash fractions.
  • Table 36 and Table 37 shows the HCP and aggregate removal achieved along with AR reduction.
  • the data clearly shows that other process related and product related substances/impurities can be achieved using the current invention on the CEX adsorbents, and hence functions as an effective polishing step in the large scale purification of monoclonal antibodies.
  • Controlling the final product quality by modifying the process based on the quality of the intermediate material is an approach that has been proposed as an effective way of ensuring product quality, with the view of ensuring safety and efficacy.
  • the cation concentration is a single parameter that can be modified to achieve a desired separation of the AR species.
  • the Tris concentration of the loading material and the wash buffer can be decreased, such that the AR enriched species is collected in the flow through fraction.
  • the AR levels can be controlled by changing the Tris concentration of the load and wash solutions.
  • the load material and the stock buffer are both prepared at 300 mM Tris concentration at the same pH.
  • the AR level of the load material was measured to be X %.
  • the load material and equilibration/wash buffer are in-line diluted to the target Tris concentration based on predetermined correlation between the AR levels and Tris concentration. As demonstrated in the example, when the Tris concentration was adjusted to 156 mM, a final AR reduction of 4.1% was achieved, whereas when the Tris conc. was adjusted to 150 mM, a final AR level of 3.1 was achieved etc (Table 38). This allows very predictable control of the AR levels ensuring achievement of the desired product quality.
  • the composition of the elution buffer can also be used to further improve the product quality profiles.
  • the impact of various cation types, concentration and pH were tested for eluting the product.
  • Table 39 There is a wide selection for elution buffer as shown in Table 39. The experiments were performed using Poros XS resin
  • Example 6.2.1. The strategy outlined in Example 6.2.1. to reduce acidic species through careful control of buffer cation type, concentration and pH can be expanded to include other chromatography adsorbents such as mixed mode or multi-modal absorbents which include a cation exchange mechanism.
  • Table 40 outlines the conditions used and the AR reduction achieved for two cation-hydrophobic interaction mixed mode resins.
  • the data clearly shows that the technology outlined in Example 6.2.1. is robust in delivering AR reduction for these types of resins across in addition to traditional cation exchange adsorbents.
  • the AR reduction can be balanced with recovery and an optimal condition can be chosen.
  • molecule 2 was also evaluated (Table 41) with the same outcome showing the same relationship between cation concentration, recovery and AR reduction.
  • the optimal condition for different molecules varies.
  • this technology when applied to CEX-HIC mixed mode resins also shows reduction of impurities as previously described.
  • the Acidic Region for Adalimumab is further grouped into two regions termed AR1 and AR2, based on a certain retention time of the peaks seen on the WCX-10 method.
  • the characteristics of the variants in these two regions are expected to be different and hence the methods that reduce variants belonging to these groups can be specifically delineated.
  • the specific species comprising the AR1 species can be identified and quantitated, to demonstrate reduction of such species by methods of the current invention. While these species are among the Acidic Species part of the charge variants, the acidic species typically described in the literature is the deamidated mAb, which is distinctly different. These results show that the Cation Exchange Resin with additional pendant hydrophobic interaction functionality, is able to provide AR reduction effectively, similar to the CEX Adsorbents.
  • Adalimumab and resin Capto Adhere were chosen.
  • the experiments were performed with Tris/Acetate buffer system at target pH and conductivity listed in Table 51
  • the load material was from Protein A affinity capture and pH adjusted. This study demonstrated the effect of loading pH and conductivity on acidic species reduction.
  • the acidic species reduction can be significantly affected by operating pH. AR reduction increased with increasing pH and/or decreasing conductivity (Table 51, Table 52 and FIG. 10 )
  • Adalimumab and resin Capto Adhere were chosen.
  • the experiments were performed with Tris/Acetate buffer system at pH 7.85 and conductivity of 2.5 mS/cm.
  • the load material was from Protein A affinity capture and pH adjusted. Column flow through was fractionated throughout the entire load and wash phases. Each fraction was analyzed for acidic species and protein recovery.
  • FIG. 11 , FIG. 12 and Table 53 demonstrate AR reduction achieved with the corresponding recovery. These AR reductions and recoveries correspond to the cumulative pools of the fractions from the start to the various points during the load/wash. This is depicted in Table 53 where the AR reductions corresponding to each of these pools. This data is plotted in FIG. 11 .
  • Adalimumab was chosen.
  • the experiments were performed with Tris/Acetate buffer system at pH 7.85 and conductivity of 2.5, 3.5, and 4.5 mS/cm.
  • the same load material was applied to different mixed mode resin columns.
  • the load material was from Protein A affinity capture and pH adjusted. Table 54 shows that all three mixed mode resins could reduce mAb acidic species. Due to the differences of resin ligands, the AR reduction level may slightly vary under certain conditions.
  • FIG. 13 displays the MAB B cumulative pool AR broke through the column of Capto Adhere operated at pH 7.0 and conductivity of 3.0 mS/cm with Tris-Acetate buffer.
  • FIG. 14 shows the MAB C cumulative pool AR broke through the column of Capto Adhere operated at pH 8.5 and conductivity of 3.0 mS/cm with Tris-Acetate buffer. Both of graphs demonstrate similar AR breakthrough curves with different AR values comparing to adalimumab ( FIG. 12 ).
  • FIG. 15 presents the AR breakthrough curves of Mab C with three different mixed mode resins with Tris-acetate buffer operated at pH 8.5 and conductivity of 3.0 mS/cm. The data clearly demonstrates that the AR reduction technology using mixed mode resins works very effectively for other antibodies.
  • Example MM 5 Demonstration of Relative pH on AR Reduction with Different Resins Using D2E7 Antibody Material
  • FIG. 16 demonstrates the impact of pH-pI and conductivity on AR reduction which compiled data from the experiments performed with Capto Adhere under conditions listed in Table 56.
  • FIG. 17 shows the impact of pH-pI and conductivity on MAB B AR reduction including the experiments operated with Tris/Acetate buffer system and multiple mixed mode resins under the conditions listed in Table 57.
  • FIG. 19 and FIG. 20 demonstrated that mAb acidic species can be reduced at wide pH range from 6.8 to 9.5.
  • FIG. 21 demonstrates the effect of pH (6.8 to 8.4), conductivity (2.3 to 13.7 mS/cm), and protein load amount (116 to 354 g/L).
  • FIG. 20 demonstrates the AR reduction at conductivity as low as—1 mS/cm.
  • Table 63 demonstrates the AR reduction at conductivity 86 mS/cm with Ammonia Sulfate-Tris-Acetate buffer system.
  • this viral inactivated material was then diluted by adding 0.3 part of a stock buffer containing 2.2M (NH 4 ) 2 SO 4 /90 mM Tris/60 mM Acetic pH 7.9 to reach conductivity of 86 mS/cm.
  • the experiments were performed with Tris/Acetate buffer system under the conditions in table 62.
  • the load material was adalimumab from Protein A affinity capture and pH adjusted. Column flow through pool was collected in each run from 50 mAU of UV A280 on the ascending and 150 mAU on the descending side of the peak.
  • the loading capacity has an impact on AR reduction but the AR reduction can be achieved over a wide range of loading capacities, and is merely a trade-off between AR reduction and recovery.
  • Capto Adhere was chosen.
  • the experiment was performed with Tris/Acetate buffer system at pH 7.8 ⁇ 0.1 and conductivity 3.0 ⁇ 0.05 mS/cm.
  • the load material was adalimumab from concentrated CEX capture and pH adjusted.
  • the prepared load material was then split to be two parts. One was directly loaded on to a Capto adhere column; the other part was diluted 2 folds with equilibration buffer to make different protein concentration.
  • Table 66 demonstrates that the load protein concentration did not have significant impact on mAb acidic species reduction.
  • molecule Adalimumab and resin Capto Adhere were chosen.
  • the experiments were performed with Tris/acetate buffer system and the load material pH was adjusted from Protein A eluates.
  • the equilibration buffer for both run was Tris/Acetic acid pH 7.8 ⁇ 0.1 and conductivity of 3.0 ⁇ 0.1 mS/cm.
  • second buffer was Tris/Acetic acid pH 7.8 ⁇ 0.1 and conductivity 6.0 mS/cm.
  • Example MM 12 Demonstration of Achievement of Absolute Value of AR Levels in Antibody Preparations Using Mixed Mode Chromatography
  • molecule Adalimumab was chosen.
  • the experiments were performed with multiple buffer systems and multiple MM absorbents under conditions listed in Table 68.
  • the load materials pH was adjusted from Protein A eluates.
  • the Acidic Region for Adalimumab is further grouped into two regions termed AR1 and AR2, based on a certain retention time of the peaks seen on the WCX-10 method.
  • the characteristics of the variants in these two regions are expected to be different and hence the methods that reduce variants belonging to these groups can be specifically delineated.
  • the capability of the current invention in achieving a certain absolute level of AR, AR1 and AR2 is demonstrated in Table 68.
  • Example MM 13 Demonstration of HCP and Aggregate Reduction in Addition to AR Reduction
  • the MM adsorbent is able to reduce other product/process related substances/impurities effectively.
  • the fact that AR reduction is effected, other impurities/substances are expected to be cleared significantly as they should bind stronger than the acidic species.
  • Table 69 and Table 70 demonstrates significant HCP and aggregate reductions with different resins, buffer systems, pH, conductivities and molecules
  • Example MM 14 Combinations of MM with Alternative Separation Strategies
  • Acidic Species Reduction by MM Adsorbents is expected to be performed after capture of the antibody by other means, or after one or more intermediate steps following the capture step.
  • the MM Adsorbent steps were performed either following a Cation Exchange Capture step or Protein A affinity capture step.
  • AR reduction was achieved at two different conductivities following Protein A Chromatography and CEX Chromatography.
  • Adalimumab was purified by a CEX chromatography step followed with a low pH viral inactivation step.
  • the filtered viral inactivated material was buffer exchanged and loaded onto a Capto Adhere column.
  • the flowthrough of Capto Adhere material was then purified with a HIC column with bind/elute mode.
  • AR reduction was achieved primarily with MM step, with some contribution from other steps.
  • Adalimumab was purified by a Protein A chromatography step followed with a low pH viral inactivation step.
  • the filtered viral inactivated material was buffer exchanged and loaded onto a Capto Adhere column.
  • the flowthrough of Capto Adhere material was then purified with a HIC column with bind/elute mode as well as Flow Through mode.
  • AR reduction was achieved primarily with MM step, with some contribution from other steps.
  • Example MM 15 Utility of AR Reduction
  • the current invention provides a method for reducing acidic species for a given protein of interest.
  • adalimumab was prepared using a combination of AEX and CEX technologies to produce a Low-AR and High-AR sample with a final AR of 2.5% and 6.9%, respectively. Both samples were incubated in a controlled environment at 25° C. and 65% relative humidity for 10 weeks, and the AR measured every two weeks.
  • FIG. 23 shows the growth of AR for each sample over the 10 week incubation. It is evident from FIG. 23 the growth rate of AR is linear and similar between both the Low-AR and High-AR samples. Based on these results the reduced AR material can be stored 3 fold longer before reaching the same AR level as the High-AR sample. This is a significant utility as this can be very beneficial in storage handling and use of the antibody or other proteins for therapeutic use.
  • Upstream and Downstream process technologies e.g., cell culture and chromatographic separations, of the inventions disclosed in the following applications can be combined together or combined with methods in the art to provide a final target AR value or achieve a % AR reduction, as well as to, in certain embodiments, reduce product related substances and/or process related impurities.
  • Upstream methods for AR reduction include, but are not limited to those described in the in the U.S. Ser. No. 13/830,583.
  • Downstream methods for AR reduction include, but are not limited to, those described in the instant application.
  • Exemplary technologies disclosed in the referenced applications include, but are not limited to: cell culture additives & conditions; clarified harvest additives and pH/salt conditions; mixed mode media separations; anion exchange media separations; and cation Exchange media separations.
  • the instant example demonstrates the combined effect of one or more of these technologies in achieving a target AR value or AR reduction, thereby facilitating the preparation of an antibody material having a specific charge heterogeneity. Additional examples of combinations of downstream technologies and upstream technologies are provided in the referenced applications.
  • the combination of upstream and downstream methods involves the reduction of acidic species in 3 L bioreactor cell cultures supplemented with arginine (2 g/l) and lysine (4 g/1) as has been previously demonstrated in the U.S. Ser. No. 13/830,583.
  • the results of that strategy are summarized in Table 74.
  • the total acidic species was reduced from 20.5% in the control sample to 10.2% in sample from cultures that were supplemented with the additives.
  • Adalimumab producing cell line 1 was cultured in media 1 (chemically defined media) supplemented with amino acid arginine (2 g/1) and lysine (4 g/l) in a 300 L bioreactor.
  • the culture was harvested and then subsequently analyzed using WCX-10 post protein A purification and the percentages of total peak(s) area corresponding to the acidic species were quantified.
  • the percentage of acidic species was estimated to be 9.1% in the 300 L harvest sample.
  • the material produced by the 300 L Bioreactor employing Arginine and Lysine additions, that effectively reduced the AR levels to 9.1% was purified using a downstream process employing Mixed Mode chromatography as the primary AR Reduction method.
  • Adalimumab was purified by a Protein A chromatography step followed with a low pH viral inactivation step.
  • the filtered viral inactivated material was buffer exchanged and loaded onto a Capto Adhere column.
  • the flowthrough of Capto Adhere material was then purified with a HIC column with bind/elute mode as well as Flow Through mode.
  • AR reduction was achieved primarily with MM step, with some contribution from other steps.
  • the table also shows that additional product related substances such as aggregates and process related impurities such as HCP can be effectively reduced employing these combined technologies.
  • the MM method further reduced the AR levels, by 2.26%. Therefore upstream technologies for reduction can be combined with downstream technologies to achieve AR levels/AR reduction.

Abstract

The instant invention relates to the field of protein production and purification, and in particular to compositions and processes for controlling the amount of charge variants, aggregates, and fragments of a protein of interest, as well as host cell proteins, present in purified preparations by applying particular chromatography conditions during such protein purification.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • The present application is a continuation application of U.S. patent application Ser. No. 15/149,898, filed on May 9, 2016, allowed, which is a continuation application of U.S. application Ser. No. 14/584,492, filed on Dec. 29, 2014, granted as U.S. Pat. No. 9,346,879, which is a divisional application of U.S. patent application Ser. No. 13/829,989, filed on Mar. 14, 2013, granted as U.S. Pat. No. 9,334,319, which claims priority to U.S. Provisional Application No. 61/636,511, filed on Apr. 20, 2012, the disclosures of each of which are incorporated by reference herein in their entirety.
  • 1. INTRODUCTION
  • The instant invention relates to the field of protein production and purification, and in particular to compositions and processes for controlling the amount of product-related substances (e.g., product charge variants, aggregates, and fragments) and/or process-related impurities (e.g., host cell proteins and media components) present in purified preparations by applying particular chromatography conditions during such protein purification.
  • 2. BACKGROUND OF THE INVENTION
  • The production of proteins for biopharmaceutical applications typically involves the use of cell cultures that are known to produce proteins exhibiting varying levels of heterogeneity. Such heterogeneity includes, but is not limited to, the presence of product-related species, such as charged species heterogeneity, consisting of acidic species and basic species. In monoclonal antibody (mAb) preparations, such acidic species heterogeneities can be detected by various methods, such as WCX-10 HPLC (a weak cation exchange chromatography) or IEF (isoelectric focusing). In certain embodiments, the acidic species identified using such techniques comprise a range of product-related substances such as antibody product fragments (e.g., Fc and Fab fragments), and/or post-translation modifications of the antibody product, such as, deamidated and/or glycoslyated antibodies. For example, in a sample of the human IgG antibody adalimumab, WCX-10 analysis measured the presence of acidic species that can be divided, based on residence time, into two groups: acidic region 1 (AR1) and acidic region 2 (AR2). Because of their similar chemical characteristics to the antibody molecules of interest, reduction of acidic species is a particular challenge in monoclonal antibody purification.
  • There remains a need in the art for high-efficiency methods of purifying proteins of interest, e.g., antibodies, away from product-related substances and process-related impurities at relatively low cost. Reduction of such substances and/or impurities is particularly advantageous in the context of commercially produced recombinant bio-therapeutics as such substances and/or impurities have the potential to impact numerous product characteristics, including, but not limited to, product stability, product safety and product efficacy.
  • 3. SUMMARY OF THE INVENTION
  • In certain embodiments, the present invention is directed to process-related impurity-reduced and/or product-related substance-modulated preparations of a protein of interest. In certain embodiments, the process-related impurities include, but are not limited to host cell proteins (HCPs), host nucleic acids, chromatographic materials, and media components. In certain embodiments, the product-related substances include, but are not limited to charge variants, such as acidic species and basic species. In certain embodiments, such acidic species correspond to heterogeneity in the distribution of protein fragments (e.g., Fc and Fab fragments of antibodies), and/or post-translation modifications of the proteins, such as, deamidated and/or glycoslyated proteins, in the population of proteins, and such heterogeneity particularly of interest when it arises in the context of recombinant protein production. Further, the present invention is directed toward pharmaceutical compositions comprising one or more proteins purified by a method described herein. In another aspect, such compositions further comprise one or more pharmaceutical agents.
  • In certain embodiments, the present invention is directed to a method for preparing a process-related impurity-reduced and/or product-related substance-modulated composition comprising a protein of interest wherein a chromatographic separation is performed to identify the particular conditions, e.g., salt concentration, pH, temperature, load amount and conditions, and washing conditions, sufficient to elicit the desired fractionation profile, e.g., fractionation of product-related substancs, such as acidic species and lysine variants, of a sample comprising the protein of interest and at least one process-related impurity and/or at least one product-related substance. In certain embodiments, the method will further comprise pooling of the resulting fractions comprising the desired process-related impurity-reduced and/or product-related substance-modulated composition comprising a protein of interest.
  • In certain embodiments, the present invention is directed to methods for isolating and purifying a protein, for example, an antibody, or an antigen-binding portion thereof, from a sample in order to exert control over the presence of process-related impurities and/or product-related substances.
  • In certain embodiments, the methods of purifying a protein, such as an antibody or antigen-binding portion thereof, from a sample, as described herein, reduces the amount of acidic species present in the resulting composition. In certain embodiments, the resulting composition is substantially free of acidic species. In certain embodiments, the resulting composition is substantially free of one or more acidic sub-species, for example, with regard to the purification of Adalimumab, the composition is substantially free of AR1 and/or AR2. In certain embodiments, the methods described herein reduce the amount of host cell proteins (“HCPs”) present in the resulting composition. In certain embodiments, the resulting composition is substantially free of HCPs. In one aspect, the sample mixture to be purified comprises a partially purified cell line harvest wherein the cell line is employed to produce specific proteins of the present invention. In a particular aspect, the sample mixture is prepared from a cell line used to produce anti-TNF-α antibodies.
  • In certain aspects, the invention is directed to methods of protein purification employing chromatography, preferably chromatography that utilizes a multimodal (also known as “mixed mode” or “MM”) media.
  • In certain embodiments, the multimodal media comprises functional groups which exhibit anion exchange and/or hydrophobic interactions. In certain embodiments, the multimodal media comprises a cross-linked agarose with a ligand, for example, N-Benzyl-N-methyl ethanol amine, that exhibits ionic interactions, hydrogen bonding and hydrophobic interactions. In certain embodiments, the cross-linked agarose with a ligand (N-Benzyl-N-methyl ethanol amine) has the following structure:
  • Figure US20190062419A1-20190228-C00001
  • In certain embodiments, the multimodal media comprises a cross-linked cellulose exhibiting porosity. In certain embodiments, the cross-linked cellulose is a phenylpropylamine having the following structure:
  • Figure US20190062419A1-20190228-C00002
  • In certain embodiments, the cross-linked cellulose is a hexylamine having the following structure:

  • —CH2—(CH2)4—CH3.
  • In certain embodiments of the present invention, a sample comprising the protein of interest, such as an antibody or antigen-binding portion thereof, is subjected to chromatography that utilizes a multimodal media, wherein the sample is subjected to a pH adjustment during loading. In one aspect, the pH is adjusted to a basic pH, or an increase in pH. An example of a suitable pH is between about a pH of 7 and 8.2, preferably a pH of between about 7.5 and 8.2. In certain embodiments the selection of appropriate pH will be based on the characteristics of the antibody and/or acidic species of interest. In certain embodiments, the pH will be selected to be about 0 to 3 units lower than the pI of the protein. In certain embodiments, it is in the range of 1 to 2 units lower. In certain embodiments, it is in the range of 1 to 1.5 units lower.
  • Certain embodiments of the present invention involve subjecting a sample mixture comprising a protein of interest, such as an antibody or antigen-binding portion thereof, to chromatography that utilizes a multimodal media, wherein the sample is subjected to a conductivity adjustment during loading. In one aspect, the conductivity is adjusted to between about 1 and 86 mS/cm, preferably between about 2 and 14 mS/cm. In certain embodiments, alternative ranges of conductivity are employed, which would be based on the characteristics of the antibody and/or acidic species of interest.
  • Certain embodiments of the present invention involve subjecting a sample comprising a protein of interest, such as an antibody or antigen-binding portion thereof, to chromatography that utilizes a multimodal media, wherein the sample is subjected to an adjustment in the amount of protein load used in the multimodal chromatography. In one aspect, the total protein load to the column is of between about 5 and 1000 g/L, or between about 50 and 500 g/L, between about 75 and 300 g/L, or between about 100 and 250 g/L. In certain embodiments, the protein concentration of the load protein mixture is adjusted to a protein concentration of the material loaded to the column of about 0.5 and 50 g/L, or between about 1 and 20 g/L.
  • In certain embodiments, the methods of the present invention involves subjecting a sample comprising a protein of interest, such as an antibody or antigen-binding portion thereof, to chromatography that utilizes an anion exchange (AEX) adsorbent material and an aqueous salt solution under loading conditions that permit both the protein of interest and non-target proteins to bind to the AEX adsorbent and collecting any unbound material with reduced levels of acidic species (and optionally reduced levels of one or more product related impurities/substances or process related impurities) and subsequently wing the adsorbent with awash buffer comprising the same, or substantially similar, aqueous salt solution used in the loading sample and collecting the effluent containing reduced levels of acidic species (and optionally reduced levels of one or more product related impurities/substances or process related impurities). In certain embodiments, the salt concentration is between 0.5 mM and 50 mM, or 2 mM and 40 mM, or 5 mM and 20 mM, depending on the salt type and AEX adsorbent being used. In certain embodiments, the concentration of the anionic and/or cationic agent in aqueous salt solution is increased or decreased to achieve a pH of between about 5 and 10, or between about 7 and 9. In certain embodiments, the aqueous salt solution comprises an anionic agent at a concentration of about 5 mM, 10 mM or 18.5 mM, and an amount of a cationic agent sufficient to achieve a pH of 8.8 or 9.0.
  • In certain embodiments of the present invention involve subjecting a sample comprising the putative protein of interest, such as an antibody or antigen-binding portion thereof, to chromatography that utilizes a AEX, wherein the sample is subjected to an adjustment in the amount of protein concentration and load. In one aspect, the total protein load to the column of between about 50 and 500 g/L, or between about 75 and 350 g/L, or between about 200 and 300 g/L. In certain embodiments, the protein concentration of the load protein mixture is adjusted to a protein concentration of the material loaded to the column of about 0.5 and 50 g/L, between about 1 and 20 g/L, or between 3 and 10 g/L.
  • In certain embodiments, the methods of the present invention involves subjecting a sample comprising a putative protein of interest, such as an antibody or antigen-binding portion thereof, to chromatography that utilizes a cation exchange (CEX) adsorbent material and an aqueous solution under loading conditions that permit both the protein of interest and non-target proteins to bind to the CEX adsorbent, wherein acidic species and non-target proteins are washed from the CEX adsorbent material using a wash buffer comprising the same, or substantially similar, aqueous solution as the loading buffer, and wherein the bound protein of interest is subsequently recovered with an elution buffer having a higher conductivity than the loading buffer.
  • In certain embodiments of the present invention involve subjecting a sample comprising the protein of interest, such as an antibody or antigen-binding portion thereof, to chromatography that utilizes a CEX, wherein the aqueous solution for loading and wash is a combination of pH and ionic concentration to allow the removal the acidic species in the unbound wash fractions. The pH employed in certain of such embodiments is adjusted such that it is below the pI of the protein of interest.
  • In certain embodiments of the present invention involve subjecting a sample comprising the putative protein of interest, such as an antibody or antigen-binding portion thereof, to chromatography that utilizes a CEX, wherein the sample is subjected to an adjustment in the amount of protein concentration and load. In one aspect, the total protein load to the column of between about 5 and 150 g/L, or between about 10 and 100 g/L, between about 20 and 80 g/L, or between about 30 and 50 g/L. In certain embodiments, the protein concentration of the load protein mixture is adjusted to a protein concentration of the material loaded to the column of about 0.5 and 50 g/L, or between about 1 and 20 g/L.
  • In certain embodiments, control over the amount of acidic species in the protein compositions described herein is exerted by employing one or more of the foregoing methods during the production and purification of the desired proteins, such as antibodies or antigen-binding portions thereof, described herein.
  • In certain embodiments, the sample is subject to a first chromatographic step prior to the multimodal/AEX or CEX media chromatography described above. The sample in each case is prepared appropriately to achieve the target pH and ion concentration prior to separation on the different modes of chromatography. Such prior chromatographic steps include ion exchange and/or affinity chromatography. Non-limiting examples of chromatographic supports for use in the first chromatographic step include, but are not limited to, affinity chromatographic resins, such as, but not limited to, Protein A resin and Protein G resin, or other affinity supports such as those comprising the antigen against which an antibody of interest was raised, as well as affinity supports comprising an Fc binding protein. In one aspect, where the protein of interest is an antibody, a sample is loaded on a protein A affinity column chromatography and eluted with a buffer system containing buffer components to be used in the multimodal media chromatography. During low pH viral inactivation, the pH is adjusted to pH 3.5 to 3.7 with acid (e.g., the same as the acidic component of the multimodal media chromatography equilibration buffer system) and held for about 30 to 90 minutes. The material is then neutralized with base) to the designed pH. In certain embodiments, the buffer is a Tris/acetate buffer system. In certain embodiments, the buffer is a trolamine/NaCl buffer. The material is then clarified with filters. The eluate can be monitored using techniques well known to those skilled in the art. For example, the absorbance at OD280 can be followed. The eluated fraction(s) of interest can then be prepared for further processing with multimodal chromatography, AEX or CEX media chromatography.
  • The purity of the proteins of interest, including but not limited process-related impurities and product-related substances, in the sample resulting from practice of the chromatographic strategies described herein can be analyzed using methods well known to those skilled in the art, e.g., weak cation exchange chromatography (WCX), capillary isoelectric focusing (cIEF), size-exclusion chromatography, Poros™ A HPLC Assay, HCP ELISA, Protein A ELISA, and western blot analysis.
  • 4. BRIEF DESCRIPTIONS OF THE DRAWINGS
  • FIG. 1 depicts a process chromatogram of pH gradient elution in the context of AEX chromatography.
  • FIG. 2 depicts a process chromatogram of a linear gradient elution by increasing anion concentration in the context of AEX chromatography.
  • FIG. 3 depicts a process chromatogram of fractionation of 300 g/L load and wash in the context of AEX chromatography.
  • FIG. 4 depicts the effect of pH on AR reduction in the context of AEX chromatography.
  • FIG. 5 depicts a process chromatogram at different salt (cation) concentrations in the context of CEX chromatography.
  • FIG. 6 depicts recovery versus AR reduction in the context of CEX purification of adalimumab.
  • FIG. 7 depicts the WCX-10 profile of glycated load material and CEX Eluate.
  • FIG. 8 depicts the WCX 10 profile of MGO modified load material and eluate from CEX column employing Toyo Pearl MX TRP 650M resin.
  • FIG. 9 depicts the change in Lysine distribution during CEX chromatography, highlighting the effect of Tris concentration.
  • FIG. 10 depicts the effect of pH and conductivity on Adalimumab AR reduction and recovery yield in the context of MM chromatography.
  • FIG. 11 depicts the AR reduction achieved with the corresponding protein recovery in the context of MM chromatography.
  • FIG. 12 depicts the total Adalimumab Protein concentration levels and AR levels during Flow Through and Wash.
  • FIG. 13 depicts the total mAb B Protein concentration levels and AR levels during Flow Through and Wash in the context of MM chromatography.
  • FIG. 14 depicts the total mAb C Protein concentration levels and AR levels during Flow Through and Wash in the context of MM chromatography.
  • FIG. 15 depicts the Cumulative % AR breakthrough of mAb C on different MM resins.
  • FIG. 16 depicts the impact of pH-pI and Conductivity on D2E7 (Adalimumab) AR Reduction in the context of MM chromatography.
  • FIG. 17 depicts the impact of pH-pI and Conductivity on mAb B AR Reduction in the context of MM chromatography.
  • FIG. 18 depicts the impact and trend of pH-pI on mAb C AR reduction with multiple resins in the context of MM chromatography.
  • FIG. 19 depicts the effect of pH and Conductivity on AR reduction and Yield in the context of MM chromatography.
  • FIG. 20 depicts AR reduction and Protein recovery vs. pH in the context of MM chromatography.
  • FIG. 21 depicts the effect of pH, conductivity and protein load amount on AR reduction and Yield.
  • FIG. 22 depicts the effect of pH, conductivity and protein load amount on AR reduction and Yield.
  • FIG. 23 depicts the AR Growth at 25° C. of low and high AR containing samples.
  • FIG. 24 depicts the effect of AEX adsorbent pKa for mAb B with several different AEX adsorbents, with different pKa values, run at with an acetate/Tris buffer at pH 9.1.
  • 5. DETAILED DESCRIPTION OF THE INVENTION
  • The instant invention relates to the field of protein purification. In particular, the instant invention relates to compositions and processes for controlling the amount of product-related substances (e.g., product charge variants, aggregates, and fragments) and/or process-related impurities (e.g., host cell proteins and media components) present in purified preparations of a protein of interest. In certain embodiments, the methods described herein involve the purification of a protein, such as, but not limited to an antibody or antigen-binding portion thereof, by multimodal chromatography, wherein the multimodal (MM) media comprises both ion exchange and hydrophobic interaction functional groups and an aqueous salt solution, wherein the same or substantially the same aqueous salt solution is used as a loading buffer and a wash buffer using which the said protein of interested is collected with, in the column effluent.
  • In certain embodiments, the methods described herein involve the purification of a protein, such as, but not limited to an antibody or antigen-binding portion thereof, by chromatography comprising an anion exchange (AEX) adsorbent material and an aqueous salt solution, wherein the same or substantially the same aqueous salt solution is used as a loading buffer and a wash buffer using which the said protein of interested is collected with, in the column effluent. In certain embodiments, the methods described herein involve the purification of a protein, such as, but not limited to an antibody or antigen-binding portion thereof, by chromatography comprising a cation exchange (CEX) adsorbent material and an aqueous salt solution, wherein the same or substantially the same aqueous salt solution is used as a loading buffer and a wash buffer, wherein the wash buffer removes acidic species and non-target proteins bound to the CEX adsorbent material, and wherein the target protein bound to the CEX adsorbent material is eluted with a buffer having a higher conductivity and/or pH than the loading/wash buffer. In certain embodiments, the present invention is directed toward pharmaceutical compositions comprising one or more proteins, such as, but not limited to an antibody or antigen-binding portion thereof, purified by a method described herein.
  • For clarity and not by way of limitation, this detailed description is divided into the following sub-portions:
      • 5.1. Definitions;
      • 5.2. Antibody Generation;
      • 5.3. Antibody Production;
      • 5.4. Antibody Purification;
      • 5.5. Methods of Assaying Sample Purity;
      • 5.6. Further Modifications; and
      • 5.7. Pharmaceutical Compositions
    5.1. Definitions
  • In order that the present invention may be more readily understood, certain terms are first defined.
  • The term “product”, as used herein refers to a protein of interest, which may be present in the context of a sample comprising one or more process-related impurities and/or product-related substances. In certain embodiments, the product, i.e., the protein of interest is an antibody or antigen binding fragment thereof.
  • The term “antibody” includes an immunoglobulin molecule comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region (CH). The heavy chain constant region is comprised of three domains, CH1, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The term “antibody”, as used herein, also includes alternative antibody and antibody-like structures, such as, but not limited to, dual variable domain antibodies (DVD-Ig).
  • The term “antigen-binding portion” of an antibody (or “antibody portion”) includes fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., hIL-12, hTNFα, or hIL-18). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment comprising the VL, VH, CL and CH1 domains; (ii) a F(ab′)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment comprising the VH and CH1 domains; (iv) a Fv fragment comprising the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546, the entire teaching of which is incorporated herein by reference), which comprises a VH domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see, e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883, the entire teachings of which are incorporated herein by reference). Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody. Other forms of single chain antibodies, such as diabodies are also encompassed. Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see, e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123, the entire teachings of which are incorporated herein by reference). Still further, an antibody may be part of a larger immunoadhesion molecule, formed by covalent or non-covalent association of the antibody with one or more other proteins or peptides. Examples of such immunoadhesion molecules include use of the streptavidin core region to make a tetrameric scFv molecule (Kipriyanov, S. M., et al. (1995) Human Antibodies and Hybridomas 6:93-101, the entire teaching of which is incorporated herein by reference) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov, S. M., et al. (1994) Mol. Immunol. 31:1047-1058, the entire teaching of which is incorporated herein by reference). Antibody portions, such as Fab and F(ab′)2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion, respectively, of whole antibodies. Moreover, antibodies, antibody portions and immunoadhesion molecules can be obtained using standard recombinant DNA techniques, as described herein. In one aspect, the antigen binding portions are complete domains or pairs of complete domains.
  • The terms “Kabat numbering” “Kabat definitions” and “Kabat labeling” are used interchangeably herein. These terms, which are recognized in the art, refer to a system of numbering amino acid residues which are more variable (i.e., hypervariable) than other amino acid residues in the heavy and light chain variable regions of an antibody, or an antigen binding portion thereof (Kabat et al. (1971) Ann. NY Acad, Sci. 190:382-391 and, Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242, the entire teachings of which are incorporated herein by reference). For the heavy chain variable region, the hypervariable region ranges from amino acid positions 31 to 35 for CDR1, amino acid positions 50 to 65 for CDR2, and amino acid positions 95 to 102 for CDR3. For the light chain variable region, the hypervariable region ranges from amino acid positions 24 to 34 for CDR1, amino acid positions 50 to 56 for CDR2, and amino acid positions 89 to 97 for CDR3.
  • The term “human antibody” includes antibodies having variable and constant regions corresponding to human germline immunoglobulin sequences as described by Kabat et al. (See Kabat, et al. (1991) Sequences of proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). The human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), e.g., in the CDRs and in particular CDR3. The mutations can be introduced using the “selective mutagenesis approach.” The human antibody can have at least one position replaced with an amino acid residue, e.g., an activity enhancing amino acid residue which is not encoded by the human germline immunoglobulin sequence. The human antibody can have up to twenty positions replaced with amino acid residues which are not part of the human germline immunoglobulin sequence. In other embodiments, up to ten, up to five, up to three or up to two positions are replaced. In one embodiment, these replacements are within the CDR regions. However, the term “human antibody”, as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • The phrase “recombinant human antibody” includes human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial human antibody library, antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see, e.g., Taylor, L. D., et al. (1992) Nucl. Acids Res. 20:6287-6295, the entire teaching of which is incorporated herein by reference) or antibodies prepared, expressed, created or isolated by any other means that involves splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences (see, Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). In certain embodiments, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo. In certain embodiments, however, such recombinant antibodies are the result of selective mutagenesis approach or back-mutation or both.
  • An “isolated antibody” includes an antibody that is substantially free of other antibodies having different antigenic specificities. Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals.
  • The term “Koff”, as used herein, is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex.
  • The term “Kd”, as used herein, is intended to refer to the dissociation constant of a particular antibody-antigen interaction.
  • The phrase “nucleic acid molecule” includes DNA molecules and RNA molecules. A nucleic acid molecule may be single-stranded or double-stranded, but in one aspect is double-stranded DNA.
  • The phrase “isolated nucleic acid molecule,” as used herein in reference to nucleic acids encoding antibodies or antibody portions (e.g., VH, VL, CDR3), e.g. an antibody having a weak binding capacity for a Protein A resin. The phrase “isolated nucleic acid molecule” is also intended to include sequences encoding bivalent, bispecific antibodies, such as diabodies in which VH and VL regions contain no other sequences other than the sequences of the diabody.
  • The phrase “recombinant host cell” (or simply “host cell”) includes a cell into which a recombinant expression vector has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell but to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein.
  • The term “modifying”, as used herein, is intended to refer to changing one or more amino acids in the antibodies or antigen-binding portions thereof. The change can be produced by adding, substituting or deleting an amino acid at one or more positions. The change can be produced using known techniques, such as PCR mutagenesis.
  • The term “about”, as used herein, is intended to refer to ranges of approximately 10-20% greater than or less than the referenced value. In certain circumstances, one of skill in the art will recognize that, due to the nature of the referenced value, the term “about” can mean more or less than a 10-20% deviation from that value.
  • The term “preparative scale”, as used herein, refers to a scale of purification operation that can be readily scaled-up and implemented at large scale manufacturing while still providing desired separation. For instance, one skilled in the field may develop a process using, e.g., a 0.5 cm (i.d.)×20 cm (L) column in the lab, and transfer it to large scale production using, e.g., a 30 cm (i.d.)×20 cm (L) column packed with the same resin and operated with the same set of buffers, same linear flow rates (or residence times) and buffer volumes. In preparative scale separation, column bed height is typically ≤about 30 cm and column pressure drop ≤about 5 bar.
  • The term “aggregates” used herein means agglomeration or oligomerization of two or more individual molecules, including but not limiting to, protein dimers, trimers, tetramers, oligomers and other high molecular weight species. Protein aggregates can be soluble or insoluble.
  • The term “fragments” used herein refers to any truncated protein species from the target molecule due to dissociation of peptide chain, enzymatic and/or chemical modifications. For instance, antibody fragments include, but not limited to, Fab, F(ab′)2, Fv, scFv, Fd, dAb, or other compositions that contain a portion of the antibody molecule.
  • The term “charge variants”, as used herein, refers to the full complement of product variants including, but not limited to acidic species and basic species (e.g., Lys variants). In certain embodiments, such variants can include product aggregates and/or product fragments, to the extent that such aggregation and/or fragmentation results in a product charge variation.
  • As used herein, the term “lysine variant heterogeneity” refers to a characteristic of a population of proteins wherein the population consists of proteins of substantially identical amino acid sequence, but where the population exhibits variation in the presence or absence of C-terminal lysine residues. Although such lysine variant heterogeneity can be observed under general cell culture conditions, the use of particular cell culture conditions, as detailed below, can increase or decrease the distribution or amount of lysine variant heterogeneity.
  • In certain embodiments, the protein is an antibody, and the distribution of lysine variant heterogeneity comprises a distribution of the lysine variants Lys 0, Lys 1 and Lys 2, wherein the Lys 0 lysine variant comprises an antibody with heavy chains that do not comprise a C-terminal lysine, wherein the Lys 1 lysine variant comprises an antibody with one heavy chain that comprises a C-terminal lysine, and wherein the Lys 2 lysine variant comprises an antibody wherein both heavy chains comprise a C-terminal lysine.
  • In certain embodiments, C-terminal lysine variants are associated with charge heterogeneities present in protein preparations, for example, monoclonal antibody (mAb) preparations, produced through a cell culture process. These heterogeneities can be detected by various methods, such as, for example, WCX-10 HPLC (a weak cation exchange chromatography), or IEF (isoelectric focusing).
  • In certain embodiments, the heterogeneity arises from subspecies of protein differing by the presence or absence of C-terminal lysines. For example, the population of proteins may comprise more than one subspecies of lysine variant. In one non-limiting example, the lysine variants may comprise at least two of Lys 0, Lys 1 and Lys 2 lysine variants which can be detected by weak cation exchange chromatography of the expression product of a host cell expressing Adalimumab.
  • In certain embodiments, the heterogeneity arises from the size of subpopulations having different C-terminal lysine profiles. For example, the population of proteins may comprise more than one subspecies of C-terminal lysine variant, and each of the variants may be present in different amounts. In one non-limiting example, the C-terminal lysine variants may be at least two of the Lys 0, Lys 1 and Lys 2 lysine variants detected by weak cation exchange chromatography of the expression product of a host cell expressing Adalimumab. In certain embodiments, Lys 0, Lys 1 or Lys 2 subspecies are present in different amounts.
  • In certain embodiments, the heterogeneity arises from both a difference in the amount of lysine variants in the population of proteins and the type of lysine variants present in the population of proteins.
  • As used herein, the terms “acidic species”, “acidic region” and “acidic species heterogeneity” refer to a characteristic of a population of proteins wherein the population includes a distribution of product-related impurities identifiable by the presence of charge heterogeneities. For example, in monoclonal antibody (mAb) preparations, such acidic species heterogeneities can be detected by various methods, such as, for example, WCX-10 HPLC (a weak cation exchange chromatography), or IEF (isoelectric focusing). In certain embodiments, the acidic species identified using such techniques comprise a mixture of product-related impurities containing antibody product fragments (e.g., Fc and Fab fragments), and/or post-translation modifications of the antibody product, such as, deamidated and/or glycoslyated antibodies.
  • In certain embodiments, the acidic species heterogeneity comprises a difference in the type of acidic species present in the population of proteins. For example, the population of proteins may comprise more than one acidic species variant.
  • In certain embodiments, the heterogeneity of the distribution of acidic species comprises a difference in the amount of acidic species in the population of proteins. For example, the population of proteins may comprise more than one acidic species variant, and each of the variants may be present in different amounts.
  • 5.2. Antibody Generation
  • The term “antibody” as used in this section refers to an intact antibody or an antigen binding fragment thereof.
  • The antibodies of the present disclosure can be generated by a variety of techniques, including immunization of an animal with the antigen of interest followed by conventional monoclonal antibody methodologies e.g., the standard somatic cell hybridization technique of Kohler and Milstein (1975) Nature 256: 495. Although somatic cell hybridization procedures are preferred, in principle, other techniques for producing monoclonal antibody can be employed e.g., viral or oncogenic transformation of B lymphocytes.
  • In certain embodiments, the animal system for preparing hybridomas is the murine system. Hybridoma production is a very well-established procedure. Immunization protocols and techniques for isolation of immunized splenocytes for fusion are known in the art. Fusion partners (e.g., murine myeloma cells) and fusion procedures are also known.
  • An antibody can be, in certain embodiments, a human, a chimeric, or a humanized antibody. Humanized antibodies of the present disclosure can be prepared based on the sequence of a non-human monoclonal antibody prepared as described above. DNA encoding the heavy and light chain immunoglobulins can be obtained from the non-human hybridoma of interest and engineered to contain non-murine (e.g., human) immunoglobulin sequences using standard molecular biology techniques. For example, to create a chimeric antibody, murine variable regions can be linked to human constant regions using methods known in the art (see e.g., U.S. Pat. No. 4,816,567 to Cabilly et al.). To create a humanized antibody, murine CDR regions can be inserted into a human framework using methods known in the art (see e.g., U.S. Pat. No. 5,225,539 to Winter, and U.S. Pat. Nos. 5,530,101; 5,585,089; 5,693,762 and 6,180,370 to Queen et al.).
  • Human monoclonal antibodies can be generated using transgenic or transchromosomic mice carrying parts of the human immune system rather than the mouse system. These transgenic and transchromosomic mice include mice referred to herein as the HuMAb Mouse® (Medarex, Inc.), KM Mouse® (Medarex, Inc.), and XenoMouse® (Amgen).
  • Moreover, alternative transchromosomic animal systems expressing human immunoglobulin genes are available in the art and can be used to raise antibodies of the disclosure. For example, mice carrying both a human heavy chain transchromosome and a human light chain tranchromosome, referred to as “TC mice” can be used; such mice are described in Tomizuka et al. (2000) Proc. Natl. Acad. Sci. USA 97:722-727. Furthermore, cows carrying human heavy and light chain transchromosomes have been described in the art (e.g., Kuroiwa et al. (2002) Nature Biotechnology 20:889-894 and PCT application No. WO 2002/092812) and can be used to raise the antibodies of this disclosure.
  • In certain embodiments, the antibodies of this disclosure are recombinant human antibodies, which can be isolated by screening of a recombinant combinatorial antibody library, e.g., a scFv phage display library, prepared using human VL and VH cDNAs prepared from mRNA derived from human lymphocytes. Methodologies for preparing and screening such libraries are known in the art. In addition to commercially available kits for generating phage display libraries (e.g., the Pharmacia Recombinant Phage Antibody System, catalog no. 27-9400-01; and the Stratagene SurfZAP™ phage display kit, catalog no. 240612, the entire teachings of which are incorporated herein), examples of methods and reagents particularly amenable for use in generating and screening antibody display libraries can be found in, e.g., Ladner et al. U.S. Pat. No. 5,223,409; Kang et al. PCT Publication No. WO 92/18619; Dower et al. PCT Publication No. WO 91/17271; Winter et al. PCT Publication No. WO 92/20791; Markland et al. PCT Publication No. WO 92/15679; Breitling et al. PCT Publication No. WO 93/01288; McCafferty et al. PCT Publication No. WO 92/01047; Garrard et al. PCT Publication No. WO 92/09690; Fuchs et al. (1991) Bio/Technology 9:1370-1372; Hay et al. (1992) Hum Antibod Hybridomas 3:81-85; Huse et al. (1989) Science 246:1275-1281; McCafferty et al., Nature (1990) 348:552-554; Griffiths et al. (1993) EMBO J 12:725-734; Hawkins et al. (1992) J Mol Biol 226:889-896; Clackson et al. (1991) Nature 352:624-628; Gram et al. (1992) PNAS 89:3576-3580; Garrard et al. (1991) Bio/Technology 9:1373-1377; Hoogenboom et al. (1991) Nuc Acid Res 19:4133-4137; and Barbas et al. (1991) PNAS 88:7978-7982; the entire teachings of which are incorporated herein.
  • Human monoclonal antibodies of this disclosure can also be prepared using SCID mice into which human immune cells have been reconstituted such that a human antibody response can be generated upon immunization. Such mice are described in, for example, U.S. Pat. Nos. 5,476,996 and 5,698,767 to Wilson et al.
  • The antibodies or antigen-binding portions thereof, of this disclosure can be altered wherein the constant region of the antibody is modified to reduce at least one constant region-mediated biological effector function relative to an unmodified antibody. To modify an antibody of the invention such that it exhibits reduced binding to the Fc receptor, the immunoglobulin constant region segment of the antibody can be mutated at particular regions necessary for Fc receptor (FcR) interactions (see, e.g., Canfield and Morrison (1991) J. Exp. Med. 173:1483-1491; and Lund et al. (1991) J. of Immunol. 147:2657-2662, the entire teachings of which are incorporated herein). Reduction in FcR binding ability of the antibody may also reduce other effector functions which rely on FcR interactions, such as opsonization and phagocytosis and antigen-dependent cellular cytotoxicity.
  • 5.3. Antibody Production
  • To express an antibody of the invention, DNAs encoding partial or full-length light and heavy chains are inserted into one or more expression vector such that the genes are operatively linked to transcriptional and translational control sequences. (See, e.g., U.S. Pat. No. 6,914,128, the entire teaching of which is incorporated herein by reference.) In this context, the term “operatively linked” is intended to mean that an antibody gene is ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended function of regulating the transcription and translation of the antibody gene. The expression vector and expression control sequences are chosen to be compatible with the expression host cell used. The antibody light chain gene and the antibody heavy chain gene can be inserted into a separate vector or, more typically, both genes are inserted into the same expression vector. The antibody genes are inserted into an expression vector by standard methods (e.g., ligation of complementary restriction sites on the antibody gene fragment and vector, or blunt end ligation if no restriction sites are present). Prior to insertion of the antibody or antibody-related light or heavy chain sequences, the expression vector may already carry antibody constant region sequences. Additionally or alternatively, the recombinant expression vector can encode a signal peptide that facilitates secretion of the antibody chain from a host cell. The antibody chain gene can be cloned into the vector such that the signal peptide is linked in-frame to the amino terminus of the antibody chain gene. The signal peptide can be an immunoglobulin signal peptide or a heterologous signal peptide (i.e., a signal peptide from a non-immunoglobulin protein).
  • In addition to the antibody chain genes, a recombinant expression vector of the invention can carry one or more regulatory sequence that controls the expression of the antibody chain genes in a host cell. The term “regulatory sequence” is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the antibody chain genes. Such regulatory sequences are described, e.g., in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990), the entire teaching of which is incorporated herein by reference. It will be appreciated by those skilled in the art that the design of the expression vector, including the selection of regulatory sequences may depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. Suitable regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV) (such as the CMV promoter/enhancer), Simian Virus 40 (SV40) (such as the SV40 promoter/enhancer), adenovirus, (e.g., the adenovirus major late promoter (AdMLP)) and polyoma. For further description of viral regulatory elements, and sequences thereof, see, e.g., U.S. Pat. No. 5,168,062 by Stinski, U.S. Pat. No. 4,510,245 by Bell et al. and U.S. Pat. No. 4,968,615 by Schaffner et al., the entire teachings of which are incorporated herein by reference.
  • In addition to the antibody chain genes and regulatory sequences, a recombinant expression vector of the invention may carry one or more additional sequences, such as a sequence that regulates replication of the vector in host cells (e.g., origins of replication) and/or a selectable marker gene. The selectable marker gene facilitates selection of host cells into which the vector has been introduced (see e.g., U.S. Pat. Nos. 4,399,216, 4,634,665 and 5,179,017, all by Axel et al., the entire teachings of which are incorporated herein by reference). For example, typically the selectable marker gene confers resistance to drugs, such as G418, hygromycin or methotrexate, on a host cell into which the vector has been introduced. Suitable selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr-host cells with methotrexate selection/amplification) and the neo gene (for G418 selection).
  • An antibody of the invention can be prepared by recombinant expression of immunoglobulin light and heavy chain genes in a host cell. To express an antibody recombinantly, a host cell is transfected with one or more recombinant expression vectors carrying DNA fragments encoding the immunoglobulin light and heavy chains of the antibody such that the light and heavy chains are expressed in the host cell and secreted into the medium in which the host cells are cultured, from which medium the antibodies can be recovered. Standard recombinant DNA methodologies are used to obtain antibody heavy and light chain genes, incorporate these genes into recombinant expression vectors and introduce the vectors into host cells, such as those described in Sambrook, Fritsch and Maniatis (eds), Molecular Cloning; A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989), Ausubel et al. (eds.) Current Protocols in Molecular Biology, Greene Publishing Associates, (1989) and in U.S. Pat. Nos. 4,816,397 & 6,914,128, the entire teachings of which are incorporated herein.
  • For expression of the light and heavy chains, the expression vector(s) encoding the heavy and light chains is (are) transfected into a host cell by standard techniques. The various forms of the term “transfection” are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like. Although it is theoretically possible to express the antibodies of the invention in either prokaryotic or eukaryotic host cells, expression of antibodies in eukaryotic cells, such as mammalian host cells, is suitable because such eukaryotic cells, and in particular mammalian cells, are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active antibody. Prokaryotic expression of antibody genes has been reported to be ineffective for production of high yields of active antibody (Boss and Wood (1985) Immunology Today 6:12-13, the entire teaching of which is incorporated herein by reference).
  • Suitable host cells for cloning or expressing the DNA in the vectors herein are the prokaryote, yeast, or higher eukaryote cells described above. Suitable prokaryotes for this purpose include eubacteria, such as Gram-negative or Gram-positive organisms, e.g., Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, e.g., Salmonella typhimurium, Serratia, e.g., Serratia marcescans, and Shigella, as well as Bacilli such as B. subtilis and B. licheniformis (e.g., B. licheniformis 41P disclosed in DD 266,710 published Apr. 12, 1989), Pseudomonas such as P. aeruginosa, and Streptomyces. One suitable E. coli cloning host is E. coli 294 (ATCC 31,446), although other strains such as E. coli B, E. coli X1776 (ATCC 31,537), and E. coli W3110 (ATCC 27,325) are suitable. These examples are illustrative rather than limiting.
  • In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for polypeptide encoding vectors. Saccharomyces cerevisiae, or common baker's yeast, is the most commonly used among lower eukaryotic host microorganisms. However, a number of other genera, species, and strains are commonly available and useful herein, such as Schizosaccharomyces pombe; Kluyveromyces hosts such as, e.g., K. lactis, K. fragilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K. wickeramii (ATCC 24,178), K. waltii (ATCC 56,500), K. drosophilarum (ATCC 36,906), K. thermotolerans, and K. marxianus; yarrowia (EP 402,226); Pichia pastoris (EP 183,070); Candida; Trichoderma reesia (EP 244,234); Neurospora crassa; Schwanniomyces such as Schwanniomyces occidentalis; and filamentous fungi such as, e.g., Neurospora, Penicillium, Tolypocladium, and Aspergillus hosts such as A. nidulans and A. niger.
  • Suitable host cells for the expression of glycosylated antibodies are derived from multicellular organisms. Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mori have been identified. A variety of viral strains for transfection are publicly available, e.g., the L-1 variant of Autographa californica NPV and the Bm-5 strain of Bombyx mori NPV, and such viruses may be used as the virus herein according to the present invention, particularly for transfection of Spodoptera frugiperda cells. Plant cell cultures of cotton, corn, potato, soybean, petunia, tomato, and tobacco can also be utilized as hosts.
  • Suitable mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr-CHO cells, described in Urlaub and Chasin, (1980) PNAS USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in Kaufman and Sharp (1982) Mol. Biol. 159:601-621, the entire teachings of which are incorporated herein by reference), NS0 myeloma cells, COS cells and SP2 cells. When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or secretion of the antibody into the culture medium in which the host cells are grown. Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary cells/-DHFR (CHO, Urlaub et al., Proc. Natl. Acad. Sci. USA 77:4216 (1980)); mouse sertoli cells (TM4, Mather, Biol. Reprod. 23:243-251 (1980)); monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TRI cells (Mather et al., Annals N.Y. Acad. Sci. 383:44-68 (1982)); MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2), the entire teachings of which are incorporated herein by reference.
  • Host cells are transformed with the above-described expression or cloning vectors for antibody production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • The host cells used to produce an antibody may be cultured in a variety of media. Commercially available media such as Ham's F10™ (Sigma), Minimal Essential Medium™ ((MEM), (Sigma), RPMI-1640 (Sigma), and Dulbecco's Modified Eagle's Medium™ ((DMEM), Sigma) are suitable for culturing the host cells. In addition, any of the media described in Ham et al., Meth. Enz. 58:44 (1979), Barnes et al., Anal. Biochem. 102:255 (1980), U.S. Pat. Nos. 4,767,704; 4,657,866; 4,927,762; 4,560,655; or 5,122,469; WO 90/03430; WO 87/00195; or U.S. Pat. No. Re. 30,985 may be used as culture media for the host cells, the entire teachings of which are incorporated herein by reference. Any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as gentamycin drug), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art. The culture conditions, such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
  • Host cells can also be used to produce portions of intact antibodies, such as Fab fragments or scFv molecules. It is understood that variations on the above procedure are within the scope of the present invention. For example, in certain embodiments it may be desirable to transfect a host cell with DNA encoding either the light chain or the heavy chain (but not both) of an antibody of this invention. Recombinant DNA technology may also be used to remove some or the entire DNA encoding either or both of the light and heavy chains that is not necessary for binding to the antigen to which the putative antibody of interest binds. The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention. In addition, bifunctional antibodies may be produced in which one heavy and one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than the one to which the putative antibody of interest binds, depending on the specificity of the antibody of the invention, by crosslinking an antibody of the invention to a second antibody by standard chemical crosslinking methods.
  • In a suitable system for recombinant expression of an antibody of the invention, a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into dhfr-CHO cells by calcium phosphate-mediated transfection. Within the recombinant expression vector, the antibody heavy and light chain genes are each operatively linked to CMV enhancer/AdMLP promoter regulatory elements to drive high levels of transcription of the genes. The recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification. The selected transformant host cells are cultured to allow for expression of the antibody heavy and light chains and intact antibody is recovered from the culture medium. Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover the antibody from the culture medium.
  • When using recombinant techniques, the antibody can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. In one aspect, if the antibody is produced intracellularly, as a first step, the particulate debris, either host cells or lysed cells (e.g., resulting from homogenization), can be removed, e.g., by centrifugation or ultrafiltration. Where the antibody is secreted into the medium, supernatants from such expression systems can be first concentrated using a commercially available protein concentration filter, e.g., an Amicon™ or Millipore Pellicon™ ultrafiltration unit.
  • Prior to the process of the invention, procedures for purification of antibodies from cell debris initially depend on the site of expression of the antibody. Some antibodies can be secreted directly from the cell into the surrounding growth media; others are made intracellularly. For the latter antibodies, the first step of a purification process typically involves: lysis of the cell, which can be done by a variety of methods, including mechanical shear, osmotic shock, or enzymatic treatments. Such disruption releases the entire contents of the cell into the homogenate, and in addition produces subcellular fragments that are difficult to remove due to their small size. These are generally removed by differential centrifugation or by filtration. Where the antibody is secreted, supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, e.g., an Amicon™ or Millipore Pellicon™ ultrafiltration unit. Where the antibody is secreted into the medium, the recombinant host cells can also be separated from the cell culture medium, e.g., by tangential flow filtration. Antibodies can be further recovered from the culture medium using the antibody purification methods of the invention.
  • 5.4. Antibody Purification 5.4.1. Antibody Purification Generally
  • In certain embodiments, the invention provides methods and compositions for producing a purified or partially purified (e.g., process-related impurity-reduced and/or product-related substance-modulated) protein preparation from a mixture comprising a protein of interest, e.g., an antibody, and at least one process-related impurity or product-related substance. In certain embodiments, the modulation of product-related substances involves the reduction of certain of such substances, while in other embodiments, such modulation can be to increase certain of such substances. For example, in certain embodiments, it is desired to match the product-related substance profile of a sample to that of a reference sample by the methods described herein.
  • In certain embodiments, the compositions of the present invention include, but are not limited to, process-related impurity-reduced and/or product-related substance-modulated compositions comprising a protein of interest. For example, but not by way of limitation, the present invention is directed to process-related impurity-reduced and/or product-related substance-modulated compositions comprising Adalimumab. Such process-related impurity-reduced and/or product-related substance-modulated compositions process-related impurity-reduced and/or product-related substance-modulated compositions address the need for improved product characteristics, including, but not limited to, product stability, product safety and product efficacy.
  • In certain embodiments, the present invention is directed to a method for preparing a process-related impurity-reduced and/or product-related substance-modulated composition comprising a protein of interest wherein a chromatographic separation is performed to identify the particular conditions, e.g., salt concentration, pH, temperature, load amount and conditions, and washing conditions, sufficient to elicit the desired fractionation profile, e.g., fractionation of product-related substancs, such as acidic species and lysing variants, of a sample comprising the protein of interest and at least one process-related impurity and/or at least one product-related substance. In certain embodiments, the method will further comprise pooling of the resulting fractions comprising the desired process-related impurity-reduced and/or product-related substance-modulated composition comprising a protein of interest.
  • In certain embodiments, the purification process of the invention begins at the separation step when the antibody has been produced using production methods described above and/or by alternative production methods conventional in the art. Once a clarified solution or mixture comprising the protein of interest, e.g., an antibody, has been obtained, separation of the protein of interest from process-related impurities, such as the other proteins produced by the cell, as well as product-related substances, such as charge variants and/or size variants (aggregates and fragments) is performed. In certain non-limiting embodiments, such separation is performed using CEX, AEX, and/or MM chromatography. In certain embodiments, a combination of one or more different purification techniques, including affinity separation step(s), ion exchange separation step(s), mixed-mode step(s), and/or hydrophobic interaction separation step(s) can also be employed. Such additional purification steps separate mixtures of proteins on the basis of their charge, degree of hydrophobicity, and/or size. In one aspect of the invention, such additional separation steps are performed using chromatography, including hydrophobic, anionic or cationic interaction (or a combination thereof). Numerous chromatography resins are available for each of these techniques, allowing accurate tailoring of the purification scheme to the particular protein involved. The essence of each of the separation methods is that proteins can either traverse at different rates down a column, achieving a physical separation that increases as they pass further down the column, or to adhere selectively to the separation medium, being then differentially eluted by different solvents. In some cases, the protein of interest is separated from impurities and or product-related substances when the impurities and/or product-related substances specifically adhere to the column and the protein of interest does not, i.e., the protein of interest is washed from the column, while in other cases the protein of interest will adhere to the column, while the impurities and/or product-related substances are washed from the column.
  • 5.4.2. Primary Recovery
  • In certain embodiments, the initial steps of the purification methods of the present invention involve the clarification and primary recovery of antibody from a sample matrix. In certain embodiments, the primary recovery will include one or more centrifugation steps to separate the antibody product from the cells and cell debris. Centrifugation of the sample can be run at, for example, but not by way of limitation, 7,000×g to approximately 12,750×g. In the context of large scale purification, such centrifugation can occur on-line with a flow rate set to achieve, for example, but not by way of limitation, a turbidity level of 150 NTU in the resulting supernatant. Such supernatant can then be collected for further purification, or in-line filtered through one or more depth filters for further clarification of the sample.
  • In certain embodiments, the initial steps of the purification methods of the present invention involve the clarification and primary recovery of antibody from a sample matrix. In certain embodiments, the primary recovery will include one or more centrifugation steps to separate the antibody product from the cells and cell debris. Centrifugation of the sample can be run at, for example, but not by way of limitation, 7,000×g to approximately 12,750×g. In the context of large scale purification, such centrifugation can occur on-line with a flow rate set to achieve, for example, but not by way of limitation, a turbidity level of 150 NTU in the resulting supernatant. Such supernatant can then be collected for further purification, or in-line filtered through one or more depth filters for further clarification of the sample.
  • In certain embodiments, the primary recovery will include the use of one or more depth filtration steps to clarify the sample matrix and thereby aid in purifying the antibodies of interest in the present invention. In other embodiments, the primary recovery will include the use of one or more depth filtration steps post centrifugation to further clarify the sample matrix. Non-limiting examples of depth filters that can be used in the context of the instant invention include the Millistak+X0HC, F0HC, D0HC, A1HC, B1HC depth filters (EMD Millipore), Cuno™ model 30/60ZA, 60/90 ZA, VR05, VR07, delipid depth filters (3M Corp.). A 0.2 μm filter such as Sartorius's 0.45/0.2 μm Sartopore™ bi-layer or Millipore's Express SHR or SHC filter cartridges typically follows the depth filters.
  • In certain embodiments, the primary recovery process can also be a point at which to reduce or inactivate viruses that can be present in the sample matrix. For example, any one or more of a variety of methods of viral reduction/inactivation can be used during the primary recovery phase of purification including heat inactivation (pasteurization), pH inactivation, solvent/detergent treatment, UV and γ-ray irradiation and the addition of certain chemical inactivating agents such as β-propiolactone or e.g., copper phenanthroline as in U.S. Pat. No. 4,534,972. In certain embodiments of the present invention, the sample matrix is exposed to detergent viral inactivation during the primary recovery phase. In other embodiments, the sample matrix may be exposed to low pH inactivation during the primary recovery phase.
  • In those embodiments where viral reduction/inactivation is employed, the sample mixture can be adjusted, as needed, for further purification steps. For example, following low pH viral inactivation, the pH of the sample mixture is typically adjusted to a more neutral pH, e.g., from about 4.5 to about 8.5, prior to continuing the purification process. Additionally, the mixture may be diluted with water for injection (WFI) to obtain a desired conductivity.
  • 5.4.3. Protein a Affinity Chromatography
  • In certain embodiments, particularly where the protein of interest is an antibody, the primary recovery sample is subjected to Protein A affinity chromatography to purify the antibody of interest away from process-related impurities, such as HCPs. There are a variety of commercial sources for Protein A resin. Suitable resins include, but not limited to, MabSelect SuRe™, MabSelect SuRe LX, MabSelect, MabSelect Xtra, rProtein A Sepharose from GE Healthcare, ProSep HC, ProSep Ultra, and ProSep Ultra Plus from EMD Millipore, MapCapture from Life Technologies.
  • In certain embodiments, the Protein A column can be equilibrated with a suitable buffer prior to sample loading. Following the loading of the column, the column can be washed one or multiple times using a suitable sets of buffers. The Protein A column can then be eluted using an appropriate elution buffer. The eluate can be monitored using techniques well known to those skilled in the art. The eluate fractions of interest can be collected and then prepared for further processing.
  • The Protein A eluate may subject to a viral inactivation step either by detergent or low pH, provided this step is not performed prior to the Protein A capture operation. A proper detergent concentration or pH and time can be selected to obtain desired viral inactivation results. After viral inactivation, the Protein A eluate is usually pH and/or conductivity adjusted for subsequent purification steps.
  • The Protein A eluate may be subjected to filtration through a depth filter to remove turbidity and/or various impurities from the antibody of interest prior to additional chromatographic polishing steps. Examples of depth filters include, but not limited to, Millistak+X0HC, F0HC, D0HC, A1HC, and B1HC Pod filters (EMD Millipore), or Zeta Plus 30ZA/60ZA, 60ZA/90ZA, delipid, VR07, and VR05 filters (3M). The Protein A eluate pool may need to be conditioned to proper pH and conductivity to obtain desired impurity removal and product recovery from the depth filtration step.
  • 5.4.5. Anion Exchange Chromatography
  • In certain embodiments, the instant invention provides methods for producing a process-related impurity and/or product-related substance-reduced protein preparation from a mixture comprising a protein of interest (i.e., a product) and at least one process-related impurity and/or product-related substance by subjecting the mixture to at least one anion exchange separation step. In certain embodiments, the anion exchange step will occur after the above-described Protein A affinity step.
  • The use of an anionic exchange material versus a cationic exchange material, such as those cation exchange materials discussed in detail below, is based on the local charges of the protein of interest in a given solution. Therefore, it is within the scope of this invention to employ an anionic exchange step prior to the use of a cationic exchange step, or a cationic exchange step prior to the use of an anionic exchange step. Furthermore, it is within the scope of this invention to employ only an anionic exchange step, only an cationic exchange step, or any serial combination of the two (including serial combinations of one or both ion exchange steps with the other chromatographic separation technologies described herein).
  • In performing the separation, the initial protein mixture can be contacted with the anion exchange material by using any of a variety of techniques, e.g., using a batch purification technique or a chromatographic technique.
  • For example, in the context of batch purification, anion exchange material is prepared in, or equilibrated to, the desired starting buffer. Upon preparation, or equilibration, a slurry of the anion exchange material is obtained. The protein of interest, e.g., antibody, solution is contacted with the slurry to allow for protein adsorption to the anion exchange material. The solution comprising the process-related impurities and/or product-related substances that do not bind to the AEX material is separated from the slurry, e.g., by allowing the slurry to settle and removing the supernatant. The slurry can be subjected to one or more washing steps and/or elution steps.
  • In the context of chromatographic separation, a chromatographic apparatus, commonly cylindrical in shape, is employed to contain the chromatographic support material (e.g., AEX material) prepared in an appropriate buffer solution. The chromatographic apparatus, if cylindrical, can have a diameter of about 5 mm to about 2 meters, and a height of 5 cm to 50 cm, and in certain embodiments, particularly for large scale processing, a height of ≤30 cm is employed. Once the chromatographic material is added to the chromatographic apparatus, a sample containing the protein of interest, e.g., an antibody, is contacted to the chromatographic material to induce the separation. Any portion of the solution that does not bind to the chromatographic material, e.g., which may comprise, depending on the AEX material being employed, the protein of interest, process-related impurities, and/or product-related substances, is separated from the chromatographic material by washing the material and collecting fractions from column. The chromatographic material can be subjected to one or more wash steps. If desired, the chromatographic material can then be contacted with a solution designed to desorb any components of the solution that have bound to the chromatographic material.
  • In certain embodiments, a wash step can be performed in the context of AEX chromatography using conditions similar to the load conditions or alternatively by decreasing the pH and/or increasing the ionic strength/conductivity of the wash in a step wise or linear gradient manner. The resulting flow through and wash fractions can be analyzed and appropriate fractions pooled to achieve the desired reduction in charged variant species. In certain embodiments, the aqueous salt solution used as both the loading and wash buffer has a pH that at or near the isoelectric point (pI) of the protein of interest. In certain embodiments the pH is about 0 to 2 units higher or lower than the pI of the protein of interest. In certain embodiments, it will be in the range of 0 to 0.5 units higher or lower. In certain embodiments, it will be at the pI of the antibody.
  • In certain non-limiting embodiments, the anionic agent is selected from the group consisting of acetate, formate, or combinations thereof. In certain non-limiting embodiments, the cationic agent is selected from the group consisting of Tris, arginine, or combinations thereof.
  • A packed anion-exchange chromatography column, anion-exchange membrane device, anion-exchange monolithic device, or depth filter media can be operated either in bind-elute mode, flow-through mode, or a hybrid mode wherein the product exhibits binding to the chromatographic material, yet can be washed from the column using a buffer that is the same or substantially similar to the loading buffer. In the bind-elute mode, the column or the membrane device is first conditioned with a buffer with appropriate ionic strength and pH under conditions where certain proteins will be immobilized on the resin based matrix. For example, in certain embodiments, during the feed load, the protein of interest will be adsorbed to the resin due to electrostatic attraction. After washing the column or the membrane device with the equilibration buffer or another buffer with different pH and/or conductivity, the product recovery is achieved by increasing the ionic strength (i.e., conductivity) of the elution buffer to compete with the solute for the charged sites of the anion exchange matrix. Changing the pH and thereby altering the charge of the solute is another way to achieve elution of the solute. The change in conductivity or pH may be gradual (gradient elution) or stepwise (step elution). In the flow-through mode, the column or the membrane device is operated at selected pH and conductivity such that the protein of interest does not bind to the resin or the membrane while the process-related impurities and product-related substances will either be retained on the column or will have a distinct elution profile as compared to the protein of interest. In the context of this hybrid strategy, process-related impurities and product-relates substances will bind to the chromatographic material (or flow through) in a manner distinct from the protein of interest, e.g., while the protein of interest and certain aggregates and/or fragments of the protein of interest may bind the chromatographic material, washes that preferentially remove the protein of interest can be applied. The column is then regenerated before next use.
  • Non-limiting examples of anionic exchange substituents include diethylaminoethyl (DEAE), quaternary aminoethyl (QAE) and quaternary amine (Q) groups. Additional non-limiting examples include: Poros 50PI and Poros 50HQ, which are a rigid polymeric bead with a backbone consisting of cross-linked poly[styrene-divinylbenzene]; Capto Q Impres and Capto DEAE, which are a high flow agarose bead; Toyopearl QAE-550, Toyopearl DEAE-650, and Toyopearl GigaCap Q-650, which are a polymeric base bead; Fractogel® EMD TMAE Hicap, which is a synthetic polymeric resin with a tentacle ion exchanger; Sartobind STIC® PA nano, which is a salt-tolerant chromatographic membrane with a primary amine ligand; Sartobind Q nano; which is a strong anion exchange chromatographic membrane; CUNO BioCap; which is a zeta-plus depth filter media constructed from inorganic filter aids, refined cellulose, and an ion exchange resin; and X0HC, which is a depth-filter media constructed from inorganic filter aid, cellulose, and mixed cellulose esters. The detailed information is listed in Table 1.
  • TABLE 1
    List of AEX Adsorbent Properties
    Particle/
    AEX Adsorbent Vendor Media Type Ligand Type Pore Size Catalog Number
    Poros PI Applied Resin Weak ~50 μm 1-2459-11
    Poros HQ Biosystems Strong ~50 μm 1-2559-11
    Capto DEAE GE Weak ~90 μm 17-5443-10
    CaptoQ Impres Strong ~90 μm 17-5316-10
    QAE-550 Tosoh Strong ~100 μm  43271
    DEAE-650 Weak ~65 μm 43201
    GigaCap Q-650 Strong ~75 μm 21854
    TMAE HiCap EMD/Millipore Strong ~40-90 μm   1.16881.0013
    Sartobind STIC ® Sartorius Membrane Weak  3-5 μm 92STPA42DN-11-A
    PA Nano
    Sartobind Q Nano Strong  3-5 μm 92IEXQ42DN-11
    CUNO BioCap 25 3M Depth Filter NA NA BC0025L60ZA05A
    X0HC Millipore NA NA MX0HC23CL3
  • In certain embodiments, the protein load of the mixture comprising protein of interest is adjusted to a total protein load to the column of between about 50 and 500 g/L, or between about 75 and 350 g/L, or between about 200 and 300 g/L. In certain embodiments, the protein concentration of the load protein mixture is adjusted to a protein concentration of the material loaded to the column of about 0.5 and 50 g/L, between about 1 and 20 g/L, or between 3 and 10 g/L.
  • In certain embodiments, additives such as poly ethylene glycol, detergents, amino acids, sugars, chaotropic agents can be added to enhance the performance of the separation, so as to achieve better recovery or product quality.
  • In certain embodiments, including, but not limited to those relating to Adalimumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR charge variants in the flow through and wash fractions while enriching for the same in the flow elution fraction, thereby producing protein preparations with reduced or free of AR variants. In certain embodiments relating to the purification of Adalimumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR1 charge variants in the flow through and wash fractions while enriching for the same in the flow elution fraction, thereby producing protein preparations with reduced or free of AR1 variants. In certain embodiments relating to Adalimumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR2 charge variants in the flow-through and wash fractions while enriching for the same in the flow elution fraction, thereby producing protein preparations with reduced or free of AR2 variants.
  • In certain embodiments, including but not limited to those relating to Adalimiumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of the methylglyoxal (MGO) variants in the flow through and wash fractions while enriching for the same in the elution fraction, thereby producing protein preparations with reduced or free of MGO variants (for example, see U.S. patent application Ser. No. 14/078,181). In certain embodiments, including, but not limited to those relating to Adalimumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of the glycated variants (schiff's base and permanently glycated forms) in the flow through and wash fractions while enriching for the same in the elution fraction, thereby producing protein preparations with reduced or free of glycated variants.
  • In certain embodiments, the loading, pH, conductivity of the AEX chromatography step, as well as elution pH conductivity, can be modified to achieve a desired distribution of process-related impurities and/or product-relates substances. For example, but not by way of limitation, certain embodiments are directed to the modulation of the lysine distribution of purified sample of a protein of interest, e.g., increasing Lys0 and decreasing Lys1 and Lys2. In certain embodiments, the methods of the present invention allow for the preparation of samples wherein the amount of Lys0 is decreased, while the amount of Lys 1 and/or Lys2 is increased.
  • In certain embodiments, an AEX chromatographic separation can be performed and combinations of fractions can be pooled to achieve a combination of desired process-related impurity and/or product-relates substance levels, in addition to, or in place of merely modulating charge variant concentration.
  • In certain embodiments, spectroscopy methods such as UV, NIR, FTIR, Fluorescence, Raman may be used to monitor levels of product-related charge variants, aggregates, low molecular weight variants (e.g., fragments of the protein of interest) in an on-line, at-line or in-line mode, which can then be used to control the level of charge variants, e.g., acidic species, in the pooled material collected from the AEX effluent. In certain embodiments, specific signals arising from the chemical modification of the proteins such as glycation, MGO modification, deamidation, glycosylation may be specifically measurable by spectroscopic methods through such in-line, on-line or at-line methods, enabling realtime or near-real time control of product quality of the resulting product. In certain embodiments, on-line, at-line or in-line monitoring methods can be used either on the effluent line of the chromatography step or in the collection vessel, to enable achievement of the desired product quality/recovery. In certain embodiments, the UV signal can be used as a surrogate to achieve an appropriate product quality/recovery, wherein the UV signal can be processed appropriately, including, but not limited to, such processing techniques as integration, differentiation, moving average, such that normal process variability can be addressed and the target product quality can be achieved. In certain embodiments, such measurements can be combined with in-line dilution methods such that ion concentration/conductivity of the load/wash can be controlled by feedback and hence facilitate product quality control.
  • In certain embodiments, a combination of AEX and CEX and MM methods can be used to prepare product-related substance-modulated materials, including certain embodiments where one technology is used in a complementary/supplementary manner with another technology. In certain embodiments, such a combination can be performed such that certain sub-species are removed predominantly by one technology, such that the combination provides the desired final composition/product quality. In certain embodiments, such combinations include the use of additional intervening chromatography, filtration, pH adjustment, UF/DF steps so as to achieve the desired product quality, ion concentration, and/or viral reduction.
  • 5.4.6. Cation Exchange Chromatography
  • In certain embodiments, the instant invention provides methods for producing a process-related impurity and/or product-related substance-reduced protein preparation from a mixture comprising a protein of interest (i.e., a product) and at least one process-related impurity and/or product-related substance by subjecting the mixture to at least one cation exchange separation step. In certain embodiments, the CEX step will occur after the above-described Protein A affinity step.
  • The use of a cationic exchange material versus a anionic exchange material, such as those anionic exchange materials discussed in detail above, is based on the local charges of the protein of interest in a given solution. Therefore, it is within the scope of this invention to employ a cationic exchange step prior to the use of an anionic exchange step, or an anionic exchange step prior to the use of a cationic exchange step. Furthermore, it is within the scope of this invention to employ only a cationic exchange step, only an anionic exchange step, or any serial combination of the two (including serial combinations of one or both ion exchange steps with the other chromatographic separation technologies described herein).
  • In performing the separation, the initial protein mixture can be contacted with the cation exchange material by using any of a variety of techniques, e.g., using a batch purification technique or a chromatographic technique, as described above in connection with Protein A or AEX.
  • In certain embodiments, the aqueous salt solution used as both the loading and wash buffer has a pH that is lower than the isoelectric point (pI) of the protein of interest. In certain embodiments, the pH is about 0 to 5 units lower than the pI of the protein. In certain embodiments, it is in the range of 1 to 2 units lower. In certain embodiments, it is in the range of 1 to 1.5 units lower.
  • In certain embodiments, the concentration of the anionic agent in aqueous salt solution is increased or decreased to achieve a pH of between about 3.5 and 10.5, or between about 4 and 10, or between about 4.5 and 9.5, or between about 5 and 9, or between about 5.5 and 8.5, or between about 6 and 8, or between about 6.5 and 7.5. In certain embodiments, the concentration of anionic agent is increased or decreased in the aqueous salt solution to achieve a pH of 5, or 5.5, or 6, or 6.5, or 6.8, or 7.5.
  • In certain embodiments, the conductivity and pH of the aqueous salt solution is adjusted by increasing or decreasing the concentration of a cationic agent. In certain embodiments, the cationic agent is maintained at a concentration of between about range of 20 mM to 500 mM, or between about 50 to 350 mM or between about 100 to 300 mM or between about 100 to 200 mM.
  • In certain non-limiting embodiments, the cationic agent is selected from the group consisting of sodium, Tris, tromethalmine, ammonium, arginine, or combinations thereof. In certain non-limiting embodiments, the anionic agent is selected from the group consisting of acetate, citrate, chloride anion, sulphate, phosphate or combinations thereof.
  • A packed cation-exchange chromatography column or a cation-exchange membrane device can be operated either in bind-elute mode, flow-through mode, or a hybrid mode wherein the product exhibits binding to the chromatographic material, yet can be washed from the column using a buffer that is the same or substantially similar to the loading buffer. The details of these modes are outlined above.
  • Cationic substituents include carboxymethyl (CM), sulfoethyl (SE), sulfopropyl (SP), phosphate (P) and sulfonate (S). Additional cationic materials include, but are not limited to: Capto SP ImpRes, which is a high flow agarose bead; CM Hyper D grade F; which is a ceramic bead coated and permeated with a functionalized hydrogel, 250-400 ionic groups μeq/mL; Eshmuno S, which is a hydrophilic polyvinyl ether base matrix with 50-100 μeq/mL ionic capacity; Nuvia C Prime, which is a hydrophobic cation exchange media composed of a macroporous highly crosslinked hydrophilic polymer matrix 55-75 μeq/mL; Nuvia S, which has a UNOsphere base matrix with 90-150 μeq/mL ionic groups; Poros HS; which is a rigid polymetic bead with a backbone consisting of cross-linked poly[styrene-divinylbenzene]; Poros XS; which is a rigid polymetic bead with a backbone consisting of cross-linked poly[styrene-divinylbenzene]; Toyo Pearl Giga Cap CM 650M, which is a polymeric base bead with 0.225 meq/mL ionic capacity; Toyo Pearl Giga Cap S 650M which is a polymeric base bead; Toyo Pearl MX TRP, which is a polymeric base bead. Detailed information concerning the aforementioned materials is listed in Table 2.
  • TABLE 2
    Cationic Materials
    particle
    Resin Vendor type size Catalog Number
    Capto SP ImpRes GE Strong ~40 μm 17-5468-10
    CM Hyper D Pall Weak ~50 μm 20050-027
    Eshmuno S Millipore Strong ~85 μm 1.20078
    Nuvia C Prime Biorad Mix Mode ~70 μm 156-3401
    Nuvia S Biorad Strong ~85 μm 156-0315
    Poros HS Applied Weak ~50 μm 13359-06
    Biosystems
    Poros XS Applied Strong ~50 μm 4404337
    Biosystems
    Toyo Pearl Giga Tosoh Weak ~75 μm 21946
    Cap CM 650M
    Toyo Pearl Giga Tosoh Strong ~75 μm 21833
    Cap S 650M
    Toyo Pearl MX Tosoh Mix Mode ~75 μm 22817
    Trp 650M
  • In certain embodiments, the protein load of the mixture comprising protein of interest is adjusted to a total protein load to the column of between about 5 and 150 g/L, or between about 10 and 100 g/L, between about 20 and 80 g/L, or between about 30 and 50 g/L. In certain embodiments, the protein concentration of the load protein mixture is adjusted to a protein concentration of the material loaded to the column of about 0.5 and 50 g/L, or between about 1 and 20 g/L.
  • In certain embodiments, additives such as poly ethylene glycol, detergents, amino acids, sugars, chaotropic agents can be added to enhance the performance of the separation, so as to achieve better recovery or product quality.
  • In certain embodiments, including, but not limited to those relating to Adalimumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR charge variants in the elution fractions while enriching for the same in the flow through and wash fractions, thereby producing protein preparations with reduced or free of AR variants. In certain embodiments relating to the purification of Adalimumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR1 charge variants in the elution fractions while enriching for the same in the flow through and wash fractions, thereby producing protein preparations with reduced or free of AR1 variants. In certain embodiments relating to the purification of Adalimumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR2 charge variants in the elution fractions while enriching for the same in the flow through and wash fractions, thereby producing protein preparations with reduced or free of AR2 variants.
  • In certain embodiments, including, but not limited to those relating to Adalimumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of the Methyl Glycoxol (MGO) variants in the elution fractions while enriching for the same in the flow through and wash fractions, thereby producing protein preparations with reduced or free of MGO variants. In certain embodiments, including, but not limited to those relating to Adalimumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of the glycated variants (schiff's base and permanently glycated forms) in the elution fractions while enriching for the same in the flow through and wash fractions, thereby producing protein preparations with reduced or free of glycated variants.
  • In certain embodiments, the loading, pH, conductivity of the CEX chromatography step, as well as elution pH conductivity, can be modified to achieve a desired distribution of process-related impurities and/or product-relates substances. For example, but not by way of limitation, certain embodiments are directed to the modulation of the lysine distribution of a purified sample of a protein of interest, e.g., increasing Lys0 and decreasing Lys1 and Lys2. In certain embodiments, the methods of the present invention allow for the preparation of samples wherein the amount of Lys0 is decreased, while the amount of Lys 1 and/or Lys2 is increased.
  • In certain embodiments, a CEX chromatographic separation can be performed and combinations of fractions can be pooled to achieve a combination of desired process-related impurity and/or product-relates substance levels, in addition to, or in place of merely modulating charge variant concentration.
  • In certain embodiments, spectroscopy methods such as UV, NIR, FTIR, Fluorescence, Raman may be used to monitor levels of product-related charge variants, aggregates, low molecular weight variants (e.g., fragments of the protein of interest) in an on-line, at-line or in-line mode, which can then be used to control the level of charge variants, e.g., acidic species, in the pooled material collected from the CEX effluent. In certain embodiments, specific signals arising from the chemical modification of the proteins such as glycation, MGO modification, deamidation, glycosylation may be specifically measurable by spectroscopic methods through such in-line, on-line or at-line methods, enabling realtime or near-real time control of product quality of the resulting product. In certain embodiments, on-line, at-line or in-line monitoring methods can be used either on the effluent line of the chromatography step or in the collection vessel, to enable achievement of the desired product quality/recovery. In certain embodiments, the UV signal can be used as a surrogate to achieve an appropriate product quality/recovery, wherein the UV signal can be processed appropriately, including, but not limited to, such processing techniques as integration, differentiation, moving average, such that normal process variability can be addressed and the target product quality can be achieved. In certain embodiments, such measurements can be combined with in-line dilution methods such that ion concentration/conductivity of the load/wash can be controlled by feedback and hence facilitate product quality control.
  • In certain embodiments, a combination of CEX and AEX and MM methods can be used to prepare product-related substance-modulated materials, including certain embodiments where one technology is used in a complementary/supplementary manner with another technology. In certain embodiments, such a combination can be performed such that certain sub-species are removed predominantly by one technology, such that the combination provides the desired final composition/product quality. In certain embodiments, such combinations include the use of additional intervening chromatography, filtration, pH adjustment, UF/DF steps so as to achieve the desired product quality, ion concentration, and/or viral reduction.
  • 5.4.7. Mixed Mode Chromatography
  • Mixed mode (“MM”) chromatography, also referred to herein as “multimodal chromatography”, is a chromatographic strategy that utilizes a support comprising a ligand that is capable of providing at least two different, in certain embodiments co-operative, sites that interact with the substance to be bound. In certain embodiments, one of these sites gives an attractive type of charge-charge interaction between the ligand and the substance of interest and the other site provides for electron acceptor-donor interaction and/or hydrophobic and/or hydrophilic interactions. Electron donor-acceptor interactions include interactions such as hydrogen-bonding, π-π, cation-π, charge transfer, dipole-dipole, induced dipole etc.
  • In certain embodiments, the resin employed for a mixed mode separation is Capto Adhere. Capto Adhere is a strong anion exchanger with multimodal functionality. Its base matrix is a highly cross-linked agarose with a ligand (N-Benzyl-N-methyl ethanol amine) that exhibits many functionalities for interaction, such as ionic interaction, hydrogen bonding and hydrophobic interaction. In certain embodiments, the resin employed for a mixed mode separation is selected from PPA-HyperCel and HEA-HyperCel. The base matrices of PPA-HyperCel and HEA-HyperCel are high porosity cross-linked cellulose. Their ligands are Phenylpropylamine and Hexylamine, respectively. Phenylpropylamine and Hexylamine offer different selectivity and hydrophobicity options for protein separations. Additional mixed mode chromatographic supports include, but are not limited to, Nuvia C Prime, Toyo Pearl MX Trp 650M, and Eshmuno® HCX.
  • In certain embodiments, the mixed mode chromatography resin is comprised of ligands coupled to an organic or inorganic support, sometimes denoted a base matrix, directly or via a spacer. The support may be in the form of particles, such as essentially spherical particles, a monolith, filter, membrane, surface, capillaries, etc. In certain embodiments, the support is prepared from a native polymer, such as cross-linked carbohydrate material, such as agarose, agar, cellulose, dextran, chitosan, konjac, carrageenan, gellan, alginate etc. To obtain high adsorption capacities, the support can be porous, and ligands are then coupled to the external surfaces as well as to the pore surfaces. Such native polymer supports can be prepared according to standard methods, such as inverse suspension gelation (S Hjerten: Biochim Biophys Acta 79(2), 393-398 (1964). Alternatively, the support can be prepared from a synthetic polymer, such as cross-linked synthetic polymers, e.g. styrene or styrene derivatives, divinylbenzene, acrylamides, acrylate esters, methacrylate esters, vinyl esters, vinyl amides etc. Such synthetic polymers can be produced according to standard methods, see e.g. “Styrene based polymer supports developed by suspension polymerization” (R Arshady: Chimica e L'Industria 70(9), 70-75 (1988)). Porous native or synthetic polymer supports are also available from commercial sources, such as Amersham Biosciences, Uppsala, Sweden.
  • In certain embodiments, the protein load of the mixture comprising protein of interest is adjusted to a total protein load to the column of between about 50 and 750 g/L, or between about 75 and 500 g/L, or between about 100 and 300 g/L. In certain embodiments, the protein concentration of the load protein mixture is adjusted to a protein concentration of the material loaded to the column of about 1 and 50 g/L, or between about 9 and 25 g/L.
  • In certain embodiments, additives such as poly ethylene glycol, detergents, amino acids, sugars, chaotropic agents can be added to enhance the performance of the separation, so as to achieve better recovery or product quality.
  • In certain embodiments, including, but not limited to those relating to Adalimumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR charge variants in the flow through and wash fractions while enriching for the same in the flow elution fraction, thereby producing protein preparations with reduced or free of AR variants. In certain embodiments relating to Adalimumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR1 charge variants in the flow through and wash fractions while enriching for the same in the flow elution fraction, thereby producing protein preparations with reduced or free of AR1 variants. In certain embodiments relating to Adalimumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of AR2 charge variants in the flow-through and wash fractions while enriching for the same in the flow elution fraction, thereby producing protein preparations with reduced or free of AR2 variants.
  • In certain embodiments, including, but not limited to those relating to Adalimumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of the methylglyoxal (MGO) variants in the flow through and wash fractions while enriching for the same in the elution fraction, thereby producing protein preparations with reduced or free of MGO variants. In certain embodiments, including, but not limited to those relating to Adalimumab, the methods of the instant invention can be used to selectively remove, significantly reduce, or essentially remove all of the glycated variants (schiff's base and permanently glycated forms) in the flow through and wash fractions while enriching for the same in the elution fraction, thereby producing protein preparations with reduced or free of glycated variants.
  • In certain embodiments, the loading, pH, conductivity of the MM chromatography step, wash pH and conductivity, as well as elution pH conductivity, can be modified to achieve a desired distribution of process-related impurities and/or product-relates substances. For example, but not by way of limitation, certain embodiments are directed to the modulation of the lysine distribution of a purified sample of a protein of interest, e.g., increasing Lys0 and decreasing Lys1 and Lys2. In certain embodiments, the methods of the present invention allow for the preparation of samples wherein the amount of Lys0 is decreased, while the amount of Lys 1 and/or Lys2 is increased.
  • In certain embodiments, a MM chromatographic separation can be performed and combinations of fractions can be pooled to achieve a combination of desired process-related impurity and/or product-relates substance levels, in addition to, or in place of merely modulating charge variant concentration.
  • In certain embodiments, spectroscopy methods such as UV, NIR, FTIR, Fluorescence, Raman may be used to monitor levels of product-related charge variants, aggregates, low molecular weight variants (e.g., fragments of the protein of interest) in an on-line, at-line or in-line mode, which can then be used to control the level of charge variants, e.g., acidic species, in the pooled material collected from the MM effluent. In certain embodiments, specific signals arising from the chemical modification of the proteins such as glycation, MGO modification, deamidation, glycosylation may be specifically measurable by spectroscopic methods through such in-line, on-line or at-line methods, enabling realtime or near-real time control of product quality of the resulting product. In certain embodiments, on-line, at-line or in-line monitoring methods can be used either on the effluent line of the chromatography step or in the collection vessel, to enable achievement of the desired product quality/recovery. In certain embodiments, the UV signal can be used as a surrogate to achieve an appropriate product quality/recovery, wherein the UV signal can be processed appropriately, including, but not limited to, such processing techniques as integration, differentiation, moving average, such that normal process variability can be addressed and the target product quality can be achieved. In certain embodiments, such measurements can be combined with in-line dilution methods such that ion concentration/conductivity of the load/wash can be controlled by feedback and hence facilitate product quality control
  • In certain embodiments, a combination of mixed mode and AEX and CEX methods can be used to prepare product-related charge variant-reduced materials, including certain embodiments where one technology is used in a complementary/supplementary manner with another technology. In certain embodiments, such a combination can be performed such that certain sub-species are removed predominantly by one technology, such that the combination provides the desired final composition/product quality. In certain embodiments, such combinations include the use of additional intervening chromatography, filtration, pH adjustment, UF/DF steps so as to achieve the desired product quality, ion concentration, and/or viral reduction.
  • 5.4.8. Hydrophobic Interaction Chromatography
  • The present invention also features methods for producing a process-related impurity and/or product-related substance-reduced protein preparation from a mixture comprising a protein of interest, e.g., an antibody, and at least one process-related impurity and/or product-related substance further comprising a hydrophobic interaction chromatography (HIC) step in addition to the displacement chromatography step.
  • In performing the separation, the sample mixture is contacted with the HIC material, e.g., using a batch purification technique or using a column or membrane chromatography. Prior to HIC purification it may be desirable to adjust the concentration of the salt buffer to achieve desired protein binding to the resin or the membrane.
  • Whereas ion exchange chromatography relies on the local charge of the protein of interest for selective separation, hydrophobic interaction chromatography employs the hydrophobic properties of the proteins to achieve selective separation. Hydrophobic groups on the protein interact with hydrophobic groups of the resin or the membrane. The more hydrophobic a protein is the stronger it will interact with the column or the membrane. Thus the HIC step removes process-related impurities (e.g., HCPs) as well as product-related substances (e.g., aggregates and fragments).
  • Like ion exchange chromatography, a HIC column or membrane device can also be operated in product a bind-elute mode, a flow-through, or a hybrid mode wherein the product exhibits binding to the chromatographic material, yet can be washed from the column using a buffer that is the same or substantially similar to the loading buffer. The details of these modes are outlined above in connection with AEX purification.
  • As hydrophobic interactions are strongest at high ionic strength, this form of separation is conveniently performed following salt elution step, such as those that are typically used in connection with ion exchange chromatography. Alternatively, salts can be added into a low salt level feed stream before this step. Adsorption of the antibody to a HIC column is favored by high salt concentrations, but the actual concentrations can vary over a wide range depending on the nature of the protein of interest, salt type and the particular HIC ligand chosen. Various ions can be arranged in a so-called soluphobic series depending on whether they promote hydrophobic interactions (salting-out effects) or disrupt the structure of water (chaotropic effect) and lead to the weakening of the hydrophobic interaction. Cations are ranked in terms of increasing salting out effect as Ba2+; Ca2+; Mg2+; Li+; Cs+; Na+; K+; Rb+; NH4 +, while anions may be ranked in terms of increasing chaotropic effect as PO4 3−; SO4 2−; CH3CO3 ; Cl; Br; NO3 ; ClO4 ; I; SCN.
  • In general, Na+, K+ or NH4 + sulfates effectively promote ligand-protein interaction in HIC. Salts may be formulated that influence the strength of the interaction as given by the following relationship: (NH4)2SO4>Na2SO4>NaCl>NH4Cl>NaBr>NaSCN.
  • In general, salt concentrations of between about 0.75 M and about 2 M ammonium sulfate or between about 1 and 4 M NaCl are useful.
  • HIC media normally comprise a base matrix (e.g., cross-linked agarose or synthetic copolymer material) to which hydrophobic ligands (e.g., alkyl or aryl groups) are coupled. A suitable HIC media comprises an agarose resin or a membrane functionalized with phenyl groups (e.g., a Phenyl Sepharose™ from GE Healthcare or a Phenyl Membrane from Sartorius). Many HIC resins are available commercially. Examples include, but are not limited to, Capto Phenyl, Phenyl Sepharose™ 6 Fast Flow with low or high substitution, Phenyl Sepharose™ High Performance, Octyl Sepharose™ High Performance (GE Healthcare); Fractogel™ EMD Propyl or Fractogel™ EMD Phenyl (E. Merck, Germany); Macro-Prep™ Methyl or Macro-Prep™ t-Butyl columns (Bio-Rad, California); WP HI-Propyl (C3)™ (J. T. Baker, New Jersey); and Toyopearl™ ether, phenyl or butyl (TosoHaas, Pa.).
  • 5.4.9. Viral Filtration
  • Viral filtration is a dedicated viral reduction step in the entire purification process. This step is usually performed post chromatographic polishing steps. Viral reduction can be achieved via the use of suitable filters including, but not limited to, Planova 20N™, 50 N or BioEx from Asahi Kasei Pharma, Viresolve™ filters from EMD Millipore, ViroSart CPV from Sartorius, or Ultipor DV20 or DV50™ filter from Pall Corporation. It will be apparent to one of ordinary skill in the art to select a suitable filter to obtain desired filtration performance.
  • 5.4.10. Ultrafiltration/Diafiltration
  • Certain embodiments of the present invention employ ultrafiltration and diafiltration steps to further concentrate and formulate the protein of interest, e.g., an antibody product. Ultrafiltration is described in detail in: Microfiltration and Ultrafiltration: Principles and Applications, L. Zeman and A. Zydney (Marcel Dekker, Inc., New York, N.Y., 1996); and in: Ultrafiltration Handbook, Munir Cheryan (Technomic Publishing, 1986; ISBN No. 87762-456-9). One filtration process is Tangential Flow Filtration as described in the Millipore catalogue entitled “Pharmaceutical Process Filtration Catalogue” pp. 177-202 (Bedford, Mass., 1995/96). Ultrafiltration is generally considered to mean filtration using filters with a pore size of smaller than 0.1 μm. By employing filters having such small pore size, the volume of the sample can be reduced through permeation of the sample buffer through the filter membrane pores while proteins, such as antibodies, are retained above the membrane surface.
  • Diafiltration is a method of using membrane filters to remove and exchange salts, sugars, and non-aqueous solvents, to separate free from bound species, to remove low molecular-weight species, and/or to cause the rapid change of ionic and/or pH environments. Microsolutes are removed most efficiently by adding solvent to the solution being diafiltered at a rate approximately equal to the permeate flow rate. This washes away microspecies from the solution at a constant volume, effectively purifying the retained protein of interest. In certain embodiments of the present invention, a diafiltration step is employed to exchange the various buffers used in connection with the instant invention, optionally prior to further chromatography or other purification steps, as well as to remove impurities from the protein preparations.
  • One of ordinary skill in the art can select appropriate membrane filter device for the UF/DF operation. Examples of membrane cassettes suitable for the present invention include, but not limited to, Pellicon 2 or Pellicon 3 cassetts with 10 kD, 30 kD or 50 kD membranes from EMD Millipore, Kvick 10 kD, 30 kD or 50 kD membrane cassettes from GE Healthcare, and Centramate or Centrasette 10 kD, 30 kD or 50 kD cassettes from Pall Corporation.
  • 5.4.11. Exemplary Purification Strategies
  • In certain embodiments, primary recovery can proceed by sequentially employing pH reduction, centrifugation, and filtration steps to remove cells and cell debris (including HCPs) from the production bioreactor harvest. In certain embodiments, the present invention is directed to subjecting a sample mixture from said primary recovery to one or more AEX, CEX, and/or MM purification steps. Certain embodiments of the present invention will include further purification steps. Examples of additional purification procedures which can be performed prior to, during, or following the ion exchange chromatography method include ethanol precipitation, isoelectric focusing, reverse phase HPLC, chromatography on silica, chromatography on heparin Sepharose™, further anion exchange chromatography and/or further cation exchange chromatography, chromatofocusing, SDS-PAGE, ammonium sulfate precipitation, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography (e.g., using protein G, an antibody, a specific substrate, ligand or antigen as the capture reagent).
  • Specific examples of such combinations of strategies is presented below, with specific data relating to particular combinations useful in the context of the instant invention included in Tables 44-51 and 72-74.
  • In certain embodiments the unbound flow through and wash fractions can be further fractionated and a combination of fractions providing a target product purity can be pooled.
  • In certain embodiments the protein concentration can be adjusted to achieve a differential partitioning behavior between the antibody product and the product-related substances such that the purity and/or yield can be further improved. In certain embodiments the loading can be performed at different protein concentrations during the loading operation to improve the product quality/yield of any particular purification step.
  • In certain embodiments the column temperature, can be independently varied to improve the separation efficiency and/or yield of any particular purification step.
  • In certain embodiments, the loading and washing buffer matrices can be different or composed of mixtures of chemicals, while achieving similar “resin interaction” behavior such that the above novel separation can be effected. For example, but not by way of limitation, the loading and washing buffers can be different, in terms of ionic strength or pH, while remaining substantially similar in function in terms of the washout of the product achieved during the wash step. In certain embodiments, additives such as amino acids, sugars, PEG, etc can be added to the load or wash steps to modulate the partitioning behavior to achieve the separation efficiency and/or yield.
  • In certain embodiments, the loading & washing steps can be controlled by in-line, at-line or off-line measurement of the product related impurity/substance levels, either in the column effluent, or the collected pool or both, so as to achieve the target product quality and/or yield. In certain embodiments, the loading concentration can be dynamically controlled by in-line or batch or continuous dilutions with buffers or other solutions to achieve the partitioning necessary to improve the separation efficiency and/or yield.
  • 5.5. Methods of Assaying Sample Purity 5.5.1. Assaying Host Cell Protein
  • The present invention also provides methods for determining the residual levels of host cell protein (HCP) concentration in the isolated/purified antibody composition. As described above, HCPs are desirably excluded from the final target substance product. Exemplary HCPs include proteins originating from the source of the antibody production. Failure to identify and sufficiently remove HCPs from the target antibody may lead to reduced efficacy and/or adverse subject reactions.
  • As used herein, the term “HCP ELISA” refers to an ELISA where the second antibody used in the assay is specific to the HCPs produced from cells, e.g., CHO cells, used to generate the antibody of interest. The second antibody may be produced according to conventional methods known to those of skill in the art. For example, the second antibody may be produced using HCPs obtained by sham production and purification runs, i.e., the same cell line used to produce the antibody of interest is used, but the cell line is not transfected with antibody DNA. In an exemplary embodiment, the second antibody is produced using HCPs similar to those expressed in the cell expression system of choice, i.e., the cell expression system used to produce the target antibody.
  • Generally, HCP ELISA comprises sandwiching a liquid sample comprising HCPs between two layers of antibodies, i.e., a first antibody and a second antibody. The sample is incubated during which time the HCPs in the sample are captured by the first antibody, for example, but not limited to goat anti-CHO, affinity purified (Cygnus). A labeled second antibody, or blend of antibodies, specific to the HCPs produced from the cells used to generate the antibody, e.g., anti-CHO HCP Biotinylated, is added, and binds to the HCPs within the sample. In certain embodiments the first and second antibodies are polyclonal antibodies. In certain aspects the first and second antibodies are blends of polyclonal antibodies raised against HCPs. The amount of HCP contained in the sample is determined using the appropriate test based on the label of the second antibody.
  • HCP ELISA may be used for determining the level of HCPs in an antibody composition, such as an eluate or flow-through obtained using the process described above. The present invention also provides a composition comprising an antibody, wherein the composition has no detectable level of HCPs as determined by an HCP Enzyme Linked Immunosorbent Assay (“ELISA”).
  • 5.5.2. Assaying Charge Variants and Aggregates
  • In certain embodiments, the levels of acidic species and other charge variants in the chromatographic samples produced using the techniques described herein are analyzed. In certain embodiments a CEX-HPLC method is employed. For example, but not by way of limitation, cation exchange chromatography can be performed on a Dionex ProPac WCX-10, Analytical column 4 mm×250 mm (Dionex, Calif.). An Agilent 1200 HPLC system can then be used as the HPLC. In certain embodiments, mobile phases such as 10 mM Sodium Phosphate dibasic pH 7.5 (Mobile phase A) and 10 mM Sodium Phosphate dibasic, 500 mM Sodium Chloride pH 5.5 (Mobile phase B) can be used. In certain embodiments, a binary gradient (94% A, 6% B: 0-20 min; 84% A, 16% B: 20-22 min; 0% A, 100% B: 22-28 min; 94% A, 6% B: 28-34 min) can be used with detection at 280 nm. In certain embodiments, quantitation is based on the relative area percent of detected peaks. In certain embodiments, the peaks that elute at relative residence time less than a certain time are together represented as the acidic peaks.
  • In certain embodiments, the levels of aggregates, monomer, and fragments in the chromatographic samples produced using the techniques described herein are analyzed. In certain embodiments, the aggregates, monomer, and fragments are measured using a size exclusion chromatographic (SEC) method for each molecule. For example, but not by way of limitation, a TSK-gel G3000SWxL, 5 μm, 125 Å, 7.8×300 mm column (Tosoh Bioscience) can be used in connection with certain embodiments, while a TSK-gel Super SW3000, 4 μm, 250 Å, 4.6×300 mm column (Tosoh Bioscience) can be used in alternative embodiments. In certain embodiments, the aforementioned columns are used along with an Agilent or a Shimazhu HPLC system. In certain embodiments, sample injections are made under isocratic elution conditions using a mobile phase consisting of, for example, 100 mM sodium sulfate and 100 mM sodium phosphate at pH 6.8, and detected with UV absorbance at 214 nm. In certain embodiments, the mobile phase will consist of 1×PBS at pH 7.4, and elution profile detected with UV absorbance at 280 nm. In certain embodiments, quantification is based on the relative area of detected peaks.
  • 5.6. Further Modifications
  • The purified proteins, e.g., antibodies and antibody-binding portions thereof, of the present invention can be modified. In some embodiments, the antibodies are chemically modified to provide a desired effect. For example, but not by way of limitation, pegylation of antibodies or antibody fragments of the invention may be carried out by any of the pegylation reactions known in the art, as described, e.g., in the following references: Focus on Growth Factors 3:4-10 (1992); EP 0 154 316; and EP 0 401 384, each of which is incorporated by reference herein in its entirety. In one aspect, the pegylation is carried out via an acylation reaction or an alkylation reaction with a reactive polyethylene glycol molecule (or an analogous reactive water-soluble polymer). A suitable water-soluble polymer for pegylation of the antibodies and antibody fragments of the invention is polyethylene glycol (PEG). As used herein, “polyethylene glycol” is meant to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (Cl—ClO) alkoxy- or aryloxy-polyethylene glycol.
  • Methods for preparing pegylated antibodies and antibody fragments of the invention will generally comprise the steps of (a) reacting the antibody or antibody fragment with polyethylene glycol, such as a reactive ester or aldehyde derivative of PEG, under suitable conditions whereby the antibody or antibody fragment becomes attached to one or more PEG groups, and (b) obtaining the reaction products. It will be apparent to one of ordinary skill in the art to select the optimal reaction conditions or the acylation reactions based on known parameters and the desired result.
  • Generally the pegylated antibodies and antibody fragments have increased half-life, as compared to the nonpegylated antibodies and antibody fragments. The pegylated antibodies and antibody fragments may be employed alone, together, or in combination with other pharmaceutical compositions.
  • An antibody of the invention can be derivatized or linked to another functional molecule (e.g., another peptide or protein). For example, an antibody of the invention can be functionally linked (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate associate of the antibody with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • One type of derivatized antibody is produced by crosslinking two or more antibodies (of the same type or of different types, e.g., to create bispecific antibodies). Suitable crosslinkers include those that are heterobifunctional, having two distinctly reactive groups separated by an appropriate spacer (e.g., m-maleimidobenzoyl-N-hydroxysuccinimide ester) or homobifunctional (e.g., disuccinimidyl suberate). Such linkers are available from Pierce Chemical Company, Rockford, Ill.
  • Useful detectable agents with which an antibody of the invention may be derivatized include fluorescent compounds. Exemplary fluorescent detectable agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-1-napthalenesulfonyl chloride, phycoerythrin and the like. An antibody may also be derivatized with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, glucose oxidase and the like. When an antibody is derivatized with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product. For example, when the detectable agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable. An antibody may also be derivatized with biotin, and detected through indirect measurement of avidin or streptavidin binding.
  • 5.7. Pharmaceutical Compositions
  • The proteins of interest, e.g., antibodies and antibody-binding portions thereof, of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject. In certain embodiments, the pharmaceutical composition comprises an antibody of the invention and a pharmaceutically acceptable carrier. As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Examples of pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. In many cases, it is desirable to include isotonic agents, e.g., sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Pharmaceutically acceptable carriers may further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody.
  • The antibodies and antibody-binding portions thereof, of the invention can be incorporated into a pharmaceutical composition suitable for parenteral administration. The antibody or antibody-portions can be prepared as an injectable solution containing, e.g., 0.1-250 mg/mL antibody. The injectable solution can be composed of either a liquid or lyophilized dosage form in a flint or amber vial, ampule or pre-filled syringe. The buffer can be L-histidine approximately 1-50 mM, (optimally 5-10 mM), at pH 5.0 to 7.0 (optimally pH 6.0). Other suitable buffers include but are not limited to sodium succinate, sodium citrate, sodium phosphate or potassium phosphate. Sodium chloride can be used to modify the toxicity of the solution at a concentration of 0-300 mM (optimally 150 mM for a liquid dosage form). Cryoprotectants can be included for a lyophilized dosage form, principally 0-10% sucrose (optimally 0.5-1.0%). Other suitable cryoprotectants include trehalose and lactose. Bulking agents can be included for a lyophilized dosage form, principally 1-10% mannitol (optimally 24%). Stabilizers can be used in both liquid and lyophilized dosage forms, principally 1-50 mM L-methionine (optimally 5-10 mM). Other suitable bulking agents include glycine, arginine, can be included as 0-0.05% polysorbate-80 (optimally 0.005-0.01%). Additional surfactants include but are not limited to polysorbate 20 and BRIJ surfactants.
  • In one aspect, the pharmaceutical composition includes the antibody at a dosage of about 0.01 mg/kg-10 mg/kg. In another aspect, the dosages of the antibody include approximately 1 mg/kg administered every other week, or approximately 0.3 mg/kg administered weekly. A skilled practitioner can ascertain the proper dosage and regime for administering to a subject.
  • The compositions of this invention may be in a variety of forms. These include, e.g., liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories. The form depends on, e.g., the intended mode of administration and therapeutic application. Typical compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with other antibodies. One mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular). In one aspect, the antibody is administered by intravenous infusion or injection. In another aspect, the antibody is administered by intramuscular or subcutaneous injection.
  • Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration. Sterile injectable solutions can be prepared by incorporating the active compound (i.e., antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile, lyophilized powders for the preparation of sterile injectable solutions, the methods of preparation are vacuum drying and spray-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, e.g., by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, e.g., monostearate salts and gelatin.
  • The antibodies and antibody-binding portions thereof, of the present invention can be administered by a variety of methods known in the art, one route/mode of administration is subcutaneous injection, intravenous injection or infusion. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. In certain embodiments, the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978, the entire teaching of which is incorporated herein by reference.
  • In certain aspects, an antibody or antibody-binding portion thereof, of the invention may be orally administered, e.g., with an inert diluent or an assimilable edible carrier. The compound (and other ingredients, if desired) may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet. For oral therapeutic administration, the compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. To administer a compound of the invention by other than parenteral administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation.
  • Supplementary active compounds can also be incorporated into the compositions. In certain aspects, an antibody or antibody-binding portion thereof, of the invention is co-formulated with and/or co-administered with one or more additional therapeutic agents that are useful for treating disorders. Such combination therapies may advantageously utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies. It will be appreciated by the skilled practitioner that when the antibodies of the invention are used as part of a combination therapy, a lower dosage of antibody may be desirable than when the antibody alone is administered to a subject (e.g., a synergistic therapeutic effect may be achieved through the use of combination therapy which, in turn, permits use of a lower dose of the antibody to achieve the desired therapeutic effect).
  • It should be understood that the antibodies of the invention can be used alone or in combination with an additional agent, e.g., a therapeutic agent, said additional agent being selected by the skilled artisan for its intended purpose. For example, the additional agent can be a therapeutic agent art-recognized as being useful to treat the disease or condition being treated by the antibody of the present invention. The additional agent also can be an agent which imparts a beneficial attribute to the therapeutic composition, e.g., an agent which affects the viscosity of the composition.
  • 6. EXAMPLES 6.1. Anion Exchange Chromatography Examples 6.1.1. Materials & Methods
  • 6.1.1.1. Chromatography Method
  • Except where noted, the materials and methods described in connection with the instant example were also employed in the examples of Sections 6.2., 6.3., and 6.4., below.
  • Pre-packed resin columns were used in the following experiments, except where specified. The column was equilibrated in a buffer system with appropriate pH and conductivity. The column load was prepared from Protein A affinity chromatography eluates or concentrated CEX chromatography elutes by buffer exchange (if the eluates were with different buffer components from the mixed mode target buffer system) or addition of the stock solutions and/or water to obtain the target pH and conductivity as specified (if the eluates were with the same buffer components as the mixed mode target buffer system). The prepared load material was filtered and loaded on the column according to the target load amount (g protein/L resin) as specified followed by washing with the equilibration buffer or buffer similar to equilibration buffer with volumes as specified. The column Flow Through/Wash were collected as fractions or as a pool. Mixed mode column was regenerated with 0.1M acetic acid, 0.15M NaCl pH3, or 0.1M Acetic acid solution, pH 3, or as specified. 1M NaOH solution was used for column cleaning.
  • 6.1.1.2. Buffer Preparation Method
  • Buffers were prepared targeting specific ion concentration for the anion by fixing the anion concentration (acid) to the target value, and adjusting the solution with the cationic component (base) to achieve the appropriate pH. For example to prepare a 10 mM Acetate-Tris buffer solution, pH 8.7, glacial acetic acid was dissolved in water to a target concentration of 10 mM and adjusted with concentrated Tris-base to pH 8.7.
  • 6.1.1.3. AR Reduction and Recovery Calculations
  • In general, the Flow Through/Wash fractions were collected and analyzed with WCX-10 method for AR levels. By actual or calculated pooling of the fractions the recovery and the corresponding AR levels were calculated.
  • 6.1.1.4. WCX-10 for Adalimumab
  • The acidic species and other charge variants present in the Adalimumab process samples were quantified according to the following methods. Cation exchange chromatography was performed on a Dionex ProPac WCX-10, Analytical column 4 mm×250 mm (Dionex, CA). An Agilent 1200 HPLC system was used as the HPLC. The mobile phases used were 10 mM Sodium Phosphate dibasic pH 7.5 (Mobile phase A) and 10 mM Sodium Phosphate dibasic, 500 mM Sodium Chloride pH 5.5 (Mobile phase B). A binary gradient (94% A, 6% B: 0-20 min; 84% A, 16% B: 20-22 min; 0% A, 100% B: 22-28 min; 94% A, 6% B: 28-34 min) was used with detection at 280 nm.
  • Quantitation was based on the relative area percent of detected peaks. The peaks that elute at relative residence time less than a certain time are together represented as the acidic peaks.
  • 6.1.1.5. WCX-10 for mAb-B
  • The acidic species and other charge variants present in the mAb-B process samples were quantified according to the following methods. Cation exchange chromatography was performed on a Dionex ProPac WCX-10, Analytical column 4 mm×250 mm (Dionex, CA). An Agilent 1200 HPLC system was used as the HPLC. The mobile phases used were 20 mM 4-Morpholineethanesulfonic acid (MES), pH 6.5 (Mobile phase A) and 20 mM 4-Morpholineethanesulfonic acid (MES), 500 mM Sodium Chloride pH 6.5 (Mobile phase B). A binary gradient (87% A, 13% B: 0-5 min; 87% A, 13% B: 5-35 min; 75% A, 25% B: 35-40 min; 0% A, 100% B: 40-43 min; 87% A, 13% B: 43-46 min; 87% A, 13% B: 46-55 min) was used with detection at 280 nm, bw 8 nm; ref 360 nm, bw 100 nm.
  • Quantitation was based on the relative area percent of detected peaks. All peaks eluting prior to the Main Isoform peak were summed as the acidic region, and all peaks eluting after the LYS-2 peaks will be summed as the basic region.
  • 6.1.1.6. WCX-10 for mAb-C
  • The mAb-C method was employed towards the quantification of the acidic species and other charge variants present mAb-C process samples. Cation exchange chromatography was performed on a Dionex ProPac WCX-10, Analytical column 4 mm×250 mm (Dionex, Calif.). An Agilent 1200 HPLC system was used as the HPLC. The mobile phases used were 20 mM 4-Morpholineethanesulfonic acid (MES), pH 6.0 (Mobile phase A) and 20 mM 4-Morpholineethanesulfonic acid (MES), 250 mM Sodium Chloride pH 6.0 (Mobile phase B). A binary gradient (97% A, 3% B: 0-1 min; 79% A, 21% B: 1-46 min; 0% A, 100% B: 46-47 min; 0% A, 100% B: 47-52 min; 97% A, 3% B: 52-53 min; 97% A, 3% B: 53-60 min) was used with detection at 280 nm, bw 8 nm; ref 360 nm, bw 100 nm.
  • Quantitation was based on the relative area percent of detected peaks. All peaks eluting prior to the Main Isoform peak will be summed as the acidic region, and all peaks eluting after the Main Isoform peak will be summed as the basic region.
  • 6.1.1.7. Size Exclusion Chromatography
  • The molecular weight distribution of collected samples were quantified according to the following methods. Size exclusion chromatography (SEC) was performed using a TSK-gel G3000SWxL, 5 μm, 125 Å, 7.8×300 mm column (Tosoh Bioscience) on an HP Agilent HPLC system. Injections were made under isocratic elution conditions using a mobile phase of 200 mM sodium sulfate, 100 mM sodium phosphate, pH 6.8, and detected with absorbance at 214 nm. Quantification is based on the relative area of detected peaks.
  • 6.1.1.8. Host Cell Protein (HCP) ELISA
  • HCP assay is based on process specific antigen based ELISA. Sample dilutions were applied to achieve readings within the calibration range. The limit of quantitation of the assay is 0.625 ng/mL.
  • 6.1.1.9. UV Spectroscopy A280
  • UV A280 was used to determine protein concentrations for the samples post protein A elution. The assay was performed on an Agilent UV Spectrophotometer following the method. The protein concentration was determined using Beer-Lambert's Law, A=εlc, where A is Absorbance, ε is the extinction coefficient, 1 is the path length, and c is the concentration. The absorbance was taken at 280 nm, the path length was 1 cm, and the extinction coefficients were 1.39 for Adalimumab, 1.38 for mAb B, and 1.43 for mAb C.
  • 6.1.2 Example AEX 1: Determining Operating Conditions Appropriate for a Mab: Media: Buffer Combination
  • The demonstration of the current invention for a specific antibody & resin is provided in this example, and consists of
      • 1. Choosing an anion concentration that allows product and impurities to bind at a given pH above the pI of the product.
      • 2. Performing a pH gradient elution covering a range above, at, and below the pI of the product.
      • 3. Determining pH range in which the protein elutes from the anion exchange media
  • In this example, adalimumab and Poros 50PI were chosen. The experiment was performed at acetate (anion) concentration of 5 mM. The column was equilibrated with 5 mM acetate/Tris at a pH of 9.0. Adalimumab was prepared at 5 mM acetate/Tris pH 9.0 and loaded to the column at 20 g-protein/L of resin. The column was washed with 10 CVs of the equilibration buffer. A pH gradient from 9.0 to 7.0 at an anion concentration of 5 mM acetate/Tris was then performed. The process chromatograms are shown in FIG. 1.
  • The demonstration of the current invention for a specific antibody & resin is provided in this example, and consists of
      • 1. For a given pH, choosing a starting anion concentration that allows product and impurities to bind to the AEX adsorbent.
      • 2. Loading a small amount of protein to the column and then performing a linear gradient elution by increasing the anion concentration keeping pH constant.
      • 3. Determining anion concentration range in which the protein elutes from the anion exchange media.
  • In this example, adalimumab and Poros 50HQ were chosen. The experiment was performed at a pH 8.7. The column was equilibrated with 10 mM acetate/Tris at pH 8.7. Adalimumab was prepared at 10 mM acetate/Tris pH 8.7 and loaded to the column at 20 g-protein/L of resin. The column was washed with 10 CVs of the equilibration buffer. A linear gradient from 10-100 mM Acetate/Tris at pH 8.7 was performed. The process chromatograms are shown in FIG. 2.
  • This general approach is used to determine the appropriate operating condition, example shown in Table 3, for any resin/mAb combination, to implement the invention.
  • TABLE 3
    Example Experimental Design Scope determined
    from pH and anion gradient elution
    Poros 50HQ - 300 g/L Loading - 30 g/L Fractionation
    pH Range 8.2-9.0
    Anion Concentration (acetate) 10-20 mM
  • In practicing the current invention, the acidic species reduction desired can be achieved by appropriate pooling of the load and wash fractions. By collecting and subsequently determining the product quality of each fraction throughout the load and wash, the accumulative AR reduction and accumulative yield can be calculated using the weighted averages up to a given fraction. Additionally, the instantaneous yield can be estimated by comparing the protein recovered against the total protein loaded to the column at a given fraction. Sample calculations are shown below:
  • Sample Calculation A: Accumulative Yield Up to a Given Fraction
  • Accumulative Yield = Accumulated Protein Mass Recovered up to Fraction Total Mass Protein Load
  • Sample Calculation B: Accumulative AR Reduction Up to a Given Fraction
  • Accumulative AR Reduction = Load AR % - Accumulated Acidic Species Mass Recovered up to Fraction Accumulated Total Protein Mass Recovered up to Fraction
  • Sample Calculation C: Instantaneous Yield Up to a Given Fraction
  • Instantaneous Yield = Accumulated Protein Mass Recovered up to Fraction Total Protein Mass Loaded to Column at Fraction
  • The demonstration of the current invention for a specific antibody & resin is provided in this example, and consists of
      • 1. For a given pH and anion concentration and anion exchange media.
      • 2. Loading the anion exchange media in excess of the dynamic binding capacity for the product for the given condition.
      • 3. Washing the column with a buffer containing a similar pH and anion concentration used for the equilibration and loading steps.
      • 4. Collecting fractions throughout the loading and wash steps and subsequently determining the product quality profile (e.g. AR, aggregate, etc.)
  • In this example, adalimumab and Poros 50PI were chosen. The experiment was performed at 5 mM acetate/arginine pH 8.8. The column was equilibrated with 5 mM acetate/arginine at pH 8.8. Adalimumab was prepared at 5 mM acetate/arginine pH 8.8 and loaded to the column at 300 g-protein/L-resin. The column was washed with 20 CVs of the equilibration buffer. Fractions were collected in volumes representing 30 g-protein/L-resin, shown in FIG. 3. Each fraction was then analyzed for product quality and the accumulative yield and AR reduction calculated, shown in Table 4. From this example, it is clear to one skilled in the art to determine a run condition which delivers a targeted product quality and/or step yield.
  • This general approach is used to evaluate the performance for a given operating condition for any resin/mAb/buffer combination.
  • TABLE 4
    Accumulative Yield and AR Reduction from FIG. 3
    Accumulative
    Fraction Load Yield ΔAR
    A2  7 g/L 0.0% 10.8%
    A3*  37 g/L 0.5% 10.8%
    A4  67 g/L 6.7% 9.7%
    A5  97 g/L 16.7% 8.9%
    A6 127 g/L 26.9% 8.4%
    B1 157 g/L 37.0% 7.7%
    B2 187 g/L 47.1% 7.1%
    B3 217 g/L 57.4% 6.4%
    B4 247 g/L 67.8% 5.8%
    B5 277 g/L 78.0% 5.3%
    B6 300 g/L 84.4% 5.0%
    B7 Wash 87.0% 4.8%
    C1 Wash 88.5% 4.7%
    C2 Wash 89.6% 4.6%
    *Dynamic Binding Capacity (DBC) = 39 g/L
  • 6.1.3. Example AEX 2: Demonstration of AR Reduction with AEX Adsorbents
  • This data set is compiled to demonstrate the AR reduction achieved with three different AEX adsorbents. Each resin was evaluated using adalimumab at an acetate concentration determined from the process outlined in Example 6.1.2. and at pH values below, near, and above the pI (e.g. pH 8.5 to 9.0). Table 5 outlines the results from these experiments.
  • TABLE 5
    Effect of AEX Resins on AR reduction of Adalimumab
    Resin Buffer Condition Load Yield ΔAR
    Poros 50PI 5 mM Acetate/Tris pH 8.5 150 g/L 90% 2.4%
    5 mM Acetate/Tris pH 8.5 300 g/L 94% 0.9%
    5 mM Acetate/Tris pH 8.7 150 g/L 87% 3.6%
    5 mM Acetate/Tris pH 8.7 300 g/L 94% 1.2%
    5 mM Acetate/Tris pH 9.0 150 g/L 83% 3.9%
    5 mM Acetate/Tris pH 9.0 300 g/L 92% 1.5%
    Poros 50HQ 18 mM Acetate/Tris pH 8.5 250 g/L 91% 3.8%
    18 mM Acetate/Tris pH 8.5 350 g/L 88% 2.2%
    18 mM Acetate/Tris pH 8.7 250 g/L 85% 6.0%
    18 mM Acetate/Tris pH 8.7 350 g/L 84% 3.1%
    18 mM Acetate/Tris pH 8.9 250 g/L 67% 5.9%
    18 mM Acetate/Tris pH 8.9 350 g/L 75% 3.6%
    CaptoDEAE 10 mM Acetate/Tris pH 8.5 150 g/L 98% 0.7%
    10 mM Acetate/Tris pH 8.5 300 g/L 97% 0.1%
    10 mM Acetate/Tris pH 8.7 150 g/L 78% 7.1%
    10 mM Acetate/Tris pH 8.7 300 g/L 95% 2.5%
    10 mM Acetate/Tris pH 9.0 150 g/L 29% 9.2%
    10 mM Acetate/Tris pH 9.0 300 g/L 82% 5.0%
  • This data set is compiled to demonstrate the AR reduction achieved with eight different AEX adsorbents. Each resin was tested using an advanced screening method using the process outlined in Example 6.1.2., and subjected to four runs using adalimumab at two different pH (e.g. pH 8.7 and 9.0) and two different acetate concentrations (e.g. 10 mM and 20 mM). In these experiments, the instantaneous (e.g. not accumulative) AR reduction was measured by analyzing the load fraction at 150 g/L and subsequently compared across all resins. Table 6 outlines the results from these experiments.
  • TABLE 6
    Advanced Screen of AEX Resins for AR reduction of Adalimumab
    Instantaneous AR Reduction
    Resin pH Acetate @ 150 g/L
    Poros 50HQ 8.7 10 mM 15.0%
    20 mM 10.7%
    9.0 10 mM 8.6%
    20 mM 13.4%
    Poros 50PI 8.7 10 mM 6.2%
    20 mM −0.1%
    9.0 10 mM 6.5%
    20 mM 3.0%
    Capto DEAE 8.7 10 mM 9.3%
    20 mM −0.2%
    9.0 10 mM 8.6%
    20 mM 7.8%
    Capto Q Impres 8.7 10 mM 12.3%
    20 mM 4.2%
    9.0 10 mM 12.3%
    20 mM 6.5%
    QAE-550C 8.7 10 mM 10.1%
    20 mM 3.5%
    9.0 10 mM 7.8%
    20 mM 4.5%
    DEAE
    650M 8.7 10 mM 5.2%
    20 mM 0.1%
    9.0 10 mM 6.9%
    20 mM −2.7%
    GigaCap Q
    650M 8.7 10 mM 8.1%
    20 mM 5.8%
    9.0 10 mM 1.8%
    20 mM 0.4%
    TMAE HiCap 8.7 10 mM 4.1%
    20 mM 2.8%
    9.0 10 mM 1.2%
    20 mM −0.1%
  • This data set is compiled to demonstrate the AR reduction achieved with two different AEX chromatographic membranes. Each membrane was tested using conditions outlined in Table 6. The results from these experiments are presented in Table 7.
  • TABLE 7
    Effect of AEX Chromatographic Membrane
    on AR reduction of Adalimumab
    Chromatographic
    Membrane Equil/Wash Buffer Load Yield ΔAR
    Sartobind STIC
    10 mM Acetate/Tris pH 8.7 500 g/L 94% 1.7%
    20 mM Acetate/Tris pH 9.0 500 g/L 100% 0.7%
    Sartobind Q
    20 mM Acetate/Tris pH 9.0 500 g/L 100% 0.3%
  • This data set is compiled to demonstrate the AR reduction achieved with two different charged depth filters. The results from these experiments are presented in Table 8.
  • TABLE 8
    Effect of Charged Depth Filters on AR reduction of Adalimumab
    Depth
    Filter Media Equil/Wash Buffer Load Yield ΔAR
    CUNO BioCap 18 mM Acetate/Tris pH 8.7 500 g/m2 92% 1.9%
    25
    X0HC 18 mM Acetate/Tris pH 8.7 500 g/m2 84% 1.1%
  • 6.1.4. Example AEX 3: Demonstration of AR Reduction with Other Antibodies, Mab B and Mab C
  • AR reduction technology of the current invention has been demonstrated with multiple antibodies using AEX adsorbents. Antibodies have different amount charged residues and at different positions, leading to a charge interaction behavior on an AEX column that differs from one antibody to another. Therefore the impact of anion type, anion concentration is different for each antibody.
  • Table 9 and Table 10 below show the data for MAB B and MAB C. The data clearly demonstrates that the AR reduction technology works very effectively for other antibodies.
  • TABLE 9
    AR reduction for mAb B, pI ~9.1
    Resin Buffer Condition pH Load Yield ΔAR
    Poros 50PI
    5 mM Acetate/Tris 9.5 300 g/L 83% 1.1%
    9.1 300 g/L 94% 1.6%
    8.5 300 g/L 98% <0.5%
    Poros 50HQ
    10 mM Acetate/Tris 9.5 300 g/L 69% <0.5%
    9.1 300 g/L 78% 5.7%
    8.5 300 g/L 81% 3.4%
    Capto DEAE
    10 mM Acetate/Tris 9.5 300 g/L 69% 4.2%
    9.1 300 g/L 82% 4.9%
    8.5 300 g/L 96% <0.5%
  • TABLE 10
    AR reduction for mAb C, pI ~7.0
    Resin Buffer Condition pH Load Yield ΔAR
    Poros 50PI
    12 mM Acetate/Tris 7.5 300 g/L 90% 2.6%
    7.0 300 g/L 89% 2.2%
    6.5 300 g/L 87% 4.0%
    Poros 50HQ 45 mM Acetate/Tris 7.5 300 g/L 86% 1.2%
    7.0 300 g/L 88% 1.2%
    6.5 300 g/L 91% 0.7%
    Capto DEAE 25 mM Acetate/Tris 7.5 300 g/L 79% 1.8%
    7.0 300 g/L 80% 1.9%
    6.5 300 g/L 89% 1.8%
  • 6.1.5. Example AEX 4: Demonstration of AR Reduction with Different pH Conditions—Adalimumab
  • The AR species in the current invention is bound during the loading step; therefore the binding pH is a key variable. The anion concentration that provides the desired performance will vary with the operational pH.
  • In this example, data compiled from different experiments is shown to demonstrate the impact of the pH choice, relative to the pI of the protein on AR reduction. This data set provides the basis for one skilled in the art to determine a pH range to perform the experiments to implement the current invention. Furthermore, this reiterates the fact that the pH choice depends on several factors and the relationship between pH and AR reduction is also mAb dependent
  • In this example, adalimumab and Poros 50PI were chosen. The experiments were performed at a concentration of 5 mM acetate/arginine at each pH specified. Adalimumab was prepared at 5 mM acetate/arginine at each pH specified and loaded to the column at 300 g-protein/L of resin. The column was washed with 20 CVs of the equilibration buffer. The results showing the pH effect on AR reduction is shown in FIG. 4.
  • It is also clear that the AR reduction can be achieved with the present invention with a range of pH choices in the range of ±0.5 pH units from the pI of multiple mAbs, which are listed in Table 11. Each of these experiments was performed with Poros50HQ resin at a 300 g/L load with an acetate/Tris buffer system.
  • TABLE 11
    AR reduction at pH above, at, and below protein pI
    Range pH-pI Molecule Yield ΔAR
    pH > pI 0.2 Adalimumab 71% 7.0%
    0.5 mAb B 69% 3.4%
    0.5 mAb C 86% 1.2%
    pH~pI 0 Adalimumab 86% 5.9%
    0 mAb B 78% 5.7%
    0 mAb C 88% 1.2%
    pH < pI −0.2 Adalimumab 93% 4.1%
    −0.5 mAb B 81% <0.5%
    −0.5 mAb C 91% 0.7%
  • 6.1.6. Example AEX 5: Demonstration of AR Reduction with Different Ion Concentrations—Adalimumab
  • Anion concentration is a key variable in the performance of anion exchange chromatography. For every combination of antibody/resin/pH there is a range of anion concentrations that provides AR reduction; the strategy outlined in Example 6.1.2. can be followed to determine the AR reduction and the corresponding recovery for each anion concentration.
  • Table 12 below shows the effect of anion concentration on AR reduction. The table also includes the effect of anion concentration for different pH values. The data demonstrates that the AR reduction can be effectively achieved over a range of anion concentrations at each pH and that the concentration ranges depend on the pH.
  • TABLE 12
    Effect of Anion Concentration and pH on AR reduction
    Resin pH Buffer Condition Load Yield ΔAR
    Poros 50PI
    9 5 mM Acetate/Arginine 300 g/L 81% 4.8%
    10 mM Acetate/Arginine 227 g/L 80% 2.4%
    18.5 mM Acetate/Arginine 107 g/L 88% 1.0%
    8.8 5 mM Acetate/Arginine 300 g/L 93% 4.5%
    10 mM Acetate/Arginine 227 g/L 88% 2.5%
    18.5 mM Acetate/Arginine 108 g/L 96% 1.2%
  • 6.1.7 Example AEX 6: Demonstration of AR Reduction with Different Buffer Systems with Adalimumab
  • The anion type and concentration are key variables in Anion Exchange Chromatography. The invention has been demonstrated with Acetate and Formate as the anion type and Tris and Arginine as the counter cation type. As one skilled in the art would appreciate the optimal pH and cation concentration is different for each cation type/mixture and was derived by using the strategy outlined in Example 6.1.2. Table 13 shows the data of AR reduction and corresponding recovery for the different anion/cation types.
  • TABLE 13
    Effect of Anion/Cation Type AR reduction
    Resin Buffer Condition Load Yield ΔAR
    Poros 50PI
    5 mM Acetate/Tris, pH 8.7 300 g/L 94% 1.2%
    2.5 mM Formate/Tris, pH 8.7 300 g/L 92% 1.3%
    5 mM Acetate/Arginine, pH 8.8 300 g/L 93% 4.5%
    Poros 50HQ
    15 mM Acetate/Arginine, pH 8.7 300 g/L 89% 3.2%
    10 mM Formate/Tris, pH 8.7 300 g/L 83% 4.9%
    18 mM Acetate/Tris, pH 8.7 300 g/L 86% 5.9%
    Capto DEAE
    10 mM Acetate/Tris, pH 8.7 300 g/L 95% 2.5%
    10 mM Formate/Tris, pH 8.7 300 g/L 94% 1.0%
    5 mM Acetate/Arginine, pH 9.0 200 g/L 41% 7.5%
  • 6.1.8. Example AEX 7: Demonstration of Ar Reduction with Different Loading
  • Furthermore, the strategy outlined in Example 6.1.2. to reduce acidic species through careful control of buffer anion type, anion concentration, AEX adsorbent, and pH can be applied to any range of protein loading. A range of relevant protein loadings (e.g. 100-350 g/L) for Poros 50HQ at pH 8.7 using Acetate as the anion is shown in Table 14, displaying a robust AR reduction across the loading range investigated.
  • TABLE 14
    Impact of Column loading
    Yield
    Load (100-100 mAU) ΔAR
    100 g/L 78% 9.7%
    200 g/L 78% 4.7%
    250 g/L 85% 6.0%
    300 g/L 89% 3.9%
    350 g/L 84% 3.1%
  • 6.1.9. Example AEX 8: Demonstration of Ar Reduction with Different Load Concentration
  • Furthermore, the strategy outlined in Example 6.1.2. to reduce acidic species through careful control of buffer anion type, anion concentration, AEX adsorbent, and pH can be applied to any range of column feed streams of varying protein concentration. A range of varying protein load concentration for a 300 g/L load of adalimumab to Poros 50HQ at 15 mM acetate/Tris pH 8.7 is shown in Table 15.
  • TABLE 15
    Effect of Protein Load concentration
    Load Yield
    Concentration (100-100 mAU) ΔAR
     5 mg/mL 90% 4.7%
    10 mg/mL 86% 4.5%
    15 mg/mL 85% 6.3%
    20 mg/mL 84% 6.2%
  • 6.2.10. Example AEX 9: Alternative Wash Modalities
  • In this example, adalimumab and Poros50HQ resin were selected. In each experiment, variations were made in the equilibration, loading, and washing pH values at a given acetate concentration (as specified). Table 16 and Table 17 show the effect of the pH variation in the step yield and AR reduction.
  • TABLE 16
    Differences in pH in Equil/Wash/Load
    Poros 50HQ - 15 mM Acetate/Tris - pH 8.7 - 200 g/L
    Equilibration Yield
    pH Load pH Wash pH (100-100 mAU) ΔAR
    8.7 8.7 8.5 83% 8.7%
    9 8.5 8.5 89% 5.1%
    9 100 g/L at pH 9.0 8.5 94% 4.5%
    100 g/L at pH 8.5
  • TABLE 17
    Differences in pH in Load/Wash
    Poros 50HQ - 18 mM Acetate/Tris pH 8.7
    Load pH Wash pH Load Yield ΔAR
    8.6 8.4  75 g/L 88.8% 4.1%
    8.6 8.5 125 g/L 89.5% 4.2%
    8.6 8.6 100 g/L 75.5% 5.3%
    8.7 8.4 100 g/L 93.8% 4.1%
    8.7 8.5 100 g/L 81.7% 3.5%
    8.7 8.5  75 g/L 94.5% 4.0%
    8.7 8.6 125 g/L 81.1% 5.4%
    8.7 8.6  75 g/L 65.8% 6.5%
    8.8 8.4 125 g/L 93.5% 3.8%
    8.8 8.5 100 g/L 83.7% 5.8%
    8.8 8.6 100 g/L 78.4% 6.4%
    8.8 8.6  75 g/L 72.7% 7.0%

    As discussed in the previous sections, the operational pH and its relation to the product pI is important in the reduction of AR species in AEX. Similarly, the operational pH relative to the pKa of the AEX adsorbent is also important as many mAbs have pI similar to the pKa of the AEX adsorbent. This effect is shown in FIG. 24 for mAb B with several different AEX adsorbents, with different pKa values, run at with an acetate/Tris buffer at pH 9.1.
  • As described in previous sections the Acidic Region for Adalimumab is further grouped into two regions termed AR1 and AR2, based on a certain retention time of the peaks seen on the WCX-10 method. The characteristics of the variants in these two regions are expected to be different and hence the methods that reduce variants belonging to these groups can be specifically delineated.
  • Further, in addition to achieving a certain AR reduction, it may be desirable to achieve a certain absolute level of AR levels, in consideration of reducing or removing certain variants. The capability of the current invention in achieving a certain absolute level of AR, AR1 and AR2 is demonstrated in Table 18. The method of the current invention can effectively reduce AR2 levels, as an overall decrease in AR levels is achieved. The method can be used to achieve a target absolute level, as exemplified by the data presented in Table 18. Multiple species are present under the group of AR2 and that the current method of invention can be used to reduce such sub-species. The method of the current invention can effectively achieve AR reduction as well as achieve a target absolute level of acidic species as exemplified by the data presented in Table 18.
  • TABLE 18
    AR1, AR2, and AR removal
    Final Final
    Resin Buffer Condition pH Load Yield ΔAR1 AR1 ΔAR2 AR2 ΔAR
    Poros 50PI 5 mM 8.5 150 g/L 90% 0.7% 1.5% 1.7% 9.4% 2.4%
    Acetate/Tris 300 g/L 94% 0.3% 1.9% 0.6% 10.5% 0.9%
    8.7 150 g/L 87% 0.9% 1.2% 2.7% 8.2% 3.6%
    300 g/L 94% 0.4% 1.7% 0.8% 10.1% 1.2%
    8.9 150 g/L 83% 1.1% 1.4% 2.8% 8.4% 3.9%
    300 g/L 92% 0.7% 1.8% 0.7% 10.5% 1.5%
    Poros 18 mM 8.5 250 g/L 91% 2.9% 1.1% 0.9% 10.8% 3.8%
    50HQ Acetate/Tris 350 g/L 88% 2.7% 1.3% −0.5% 12.2% 2.2%
    8.7 250 g/L 88% 3.1% 0.9% 2.9% 9.0% 6.0%
    350 g/L 84% 2.8% 1.2% 0.3% 11.6% 3.1%
    8.9 250 g/L 67% 2.6% 1.4% 3.2% 8.6% 5.9%
    350 g/L 75% 2.3% 1.7% 1.3% 10.5% 3.6%
    CaptoDEAE 10 mM 8.5 150 g/L 98% −0.1% 2.1% 0.8% 10.0% 0.7%
    Acetate/Tris 300 g/L 97% 0.0% 2.0% 0.1% 10.8% 0.1%
    8.7 150 g/L 78% 2.4% 0.8% 4.7% 6.4% 7.1%
    300 g/L 95% 1.5% 1.7% 1.0% 10.1% 2.5%
    8.9 150 g/L 29% 2.1% 0.8% 8.0% 3.0% 10.2%
    300 g/L 82% 1.7% 1.2% 3.3% 7.7% 5.0%
  • 6.1.11. Example AEX 10: Demonstration of HCP and Aggregate Reduction in Addition to AR Reduction
  • AEX chromatography is effective in reducing aggregate and HCP levels. In the present invention, it has been demonstrated that HCP and aggregate levels can be effectively reduced under operating conditions selected for AR reduction. Table 19 and Table 20 shows the aggregate and HCP removal achieved along with AR reduction. The data clearly shows that other process related and product related substances/impurities can be achieved using the current invention on the AEX adsorbents, and hence functions as an effective polishing step in the large scale purification of monoclonal antibodies.
  • TABLE 19
    Aggregate removal during AEX Chromatography
    ΔAggregate
    Buffer Condition Load Yield Absolute Relative ΔAR
    5 mM Acetate/ 300 g/L 81% 0.92% 93% 4.5%
    Tris, pH 9.0
    10 mM Acetate/ 227 g/L 80% 0.81% 88% 2.4%
    Tris, pH 9.0
    18.5 mM Acetate/ 107 g/L 88% 0.37% 41% 1.0%
    Tris, pH 9.0
    5 mM Acetate/ 300 g/L 93% 0.91% 91% 4.5%
    Tris, pH 8.8
    10 mM Acetate/ 227 g/L 88% 0.67% 77% 2.5%
    Arginine, pH 8.8
    18.5 mM Acetate/ 108 g/L 96% 0.34% 40% 1.2%
    Arginine, pH 8.8
  • TABLE 20
    HCP Removal during AEX Chromatography
    Poros 50PI - D2E7 - 300 g/L
    Load HCP Pool HCP HCP
    Buffer Condition Yield (ng/mL) (ng/mL) (LRF) ΔAR
    5 mM Acetate/ 81% 11,617 69 2.2 4.8%
    Tris, pH 9.0
    10 mM Acetate/ 95% 83 2.1 0.8%
    Tris, pH 9.0
    5 mM Acetate/ 93% 13,507 51 2.4 4.5%
    Tris, pH 8.8
    10 mM Acetate/ 97% 84 2.2 1.5%
    Arginine, pH 8.8
  • 6.1.12. Example AEX 11: Demonstration of Means of Controlling AR Reduction
  • Controlling the final product quality by modifying the process based on the quality of the intermediate material is an approach that has been proposed as an effective way of ensuring product quality, with the view of ensuring safety and efficacy.
  • Considering that the AR levels generated during cell culture and other upstream steps can be variable, it is desirable to design a downstream process step that implements a means of controlling the product quality; and to further have a specific means of controlling a process parameter to influence the quality of the product.
  • In the current invention, such a control is possible, as the pH and load (i.e. g/L) are parameters that can be modified to achieve a desired separation of the AR species. For example, to achieve a higher level of AR reduction at a given anion concentration and pH, the load to the column can be reduced. Additionally, for a given anion concentration and loading, the pH can be increased in order to achieve a higher reduction in AR species.
  • As an example, and not to be restrictive in any manner, it has been demonstrated in this example that the AR levels can be controlled by changing the pH of the load and wash solutions as well as the total load to the column. A pilot scale Poros HQ column (10 cm diameter×22.5 cm height, 1.8 L), was used for this study.
  • The load material and the stock buffer are both prepared at 18 mM Acetate/Tris the specified pH by titrating the affinity captured material with a stock Tris solution. The AR level of the load material was the same for both runs. This experiment demonstrates how the final AR level can be modulated, while maintaining acceptable yields, by adjusting the pH and protein load to the column, shown in Table 21.
  • TABLE 21
    Modulating AR Reduction using Process Analytical
    Technology approach
    Final
    Buffer Condition Load Yield ΔAR AR
    18 mM Acetate/Tris, pH 8.7 200 g/L 77% 5.6% 5.5%
    18 mM Acetate/Tris, pH 8.5 300 g/L 89% 3.1% 8.2%
  • 6.2. Cation Exchange Chromatography Examples 6.2.1. Example CEX 1: Determining Operating Conditions Appropriate for a Mab: Resin: Buffer Combination
  • The demonstration of the current invention for a specific antibody & resin is provided in this example, and consists of
      • 1. Choosing a pH that is below the pI of the protein.
      • 2. Choosing a NaCl concentration in the range of 100 to 150 mM and performing the experiments at, for example, 115, 125, 135 concentrations.
      • 3. Determining the acidic species distribution in the ft/wash fraction vs the elution.
      • 4. Choosing a NaCl concentration that provides the desired acidic species levels and recovery
  • In this example, adalimumab was chosen and Poros XS was chosen. The experiments were performed at pH 6.0. The process chromatograms are shown in FIG. 5. The recovery vs AR reduction curves for each of the experiments is shown in FIG. 6 and Table 22. From this set of experiments, a sodium concentration of 125 mM can be chosen and such that the recovery of the eluate is 74%, which provides an AR reduction of 5.4%. Alternately, an AR reduction value of 5.4% can be chosen which will provide a recovery of −75%.
  • This general approach is used to determine the appropriate operating condition for any resin/mAb combination, to implement the invention.
  • In practicing certain embodiments of the current invention, the acidic species reduction desired can be achieved by appropriate pooling of the elution fraction with the wash fractions. In the example described in the previous section the elution fractions can be pooled with wash fractions as shown in Table 22 to achieve AR reductions from about 1 percent to 7 percent depending on the fractions pooled. This approach can be implemented to achieve a target yield and AR reduction as exemplified in FIG. 6.
  • TABLE 22
    Wash fractions and eluate combination versus AR reduction
    Recovery % AR
    Wash Fractions (%) Reduction
    Eluate 74 5.4
    Eluate + Fraction 1 82 4.3
    Eluate + Fraction 1 + Fraction 2 88 3.0
    Eluate + Fraction 1 + Fraction 2 + Fraction 3 95 0.9
    Eluate + Fraction 1 + Fraction 2 + Fraction 3 + 96 0.1
    Fraction 4
  • 6.2.2. Example CEX 2: Demonstration of AR Reduction with CEX Adsorbents
  • This data set is compiled to demonstrate the AR reduction achieved with 8 different CEX adsorbents. Conditions were derived for each resin based on the strategy outlined in Example 6.2.1. Table 23 outlines the conditions used and the AR reduction achieved and the corresponding recovery achieved.
  • The data clearly shows that the technology is robust in delivering AR reduction in all the 10 resins. As described in Example 6.2.1., the AR reduction can be balanced with recovery and an optimal condition can be chosen. Experiments were performed at pH 7.5. 29 mM Tris-acetate was used for pH control.
  • TABLE 23
    Effect of CEX adsorbents on AR reduction
    % AR
    Resin Tris concentration (mM) Yield (%) Reduction
    Poros XS
    135 103.3 0.7
    140 78.6 6.8
    145 72.6 7.3
    Poros HS 100 70.0 6.7
    105 68.7 7.1
    110 60.6 7.6
    Capto SP ImpRes 50 71.5 5.7
    55 61.0 6.3
    60 46.2 6.8
    Nuvia S 75 67.6 10.0
    80 54.3 10.8
    85 41.0 12.2
    Giga Cap CM 650 55 70.3 6.0
    57.5 62.7 7.0
    60 55.6 8.6
    Eshmuno S 65 52.7 9.0
    70 35.4 11.2
    75 22.7 12.2
    Giga Cap S 650 65 66.3 8.4
    70 43.6 11.1
    75 31.4 12.1
    CM Hyper D 45 72.2 8.9
    47.5 63.2 9.9
    50 51.5 10.3
  • 6.2.3. Example CEX 3: Demonstration of AR Reduction with Other Antibodies: mAb B and mAb C
  • AR reduction technology of the current invention has been demonstrated with multiple antibodies using CEX Adsorbents. Antibodies have different amounts of charged residues and at different positions, leading to a charge interaction behavior on a CEX column that differs from one antibody to another. Therefore the impact of cation type, cation concentration is different for each antibody.
  • For each antibody/resin combination, the experimental strategy outlined in Example 6.2.1. was employed to determine the cation concentration for each cation type that provided AR reduction.
  • Table 24 and Table 25 below shows the data for MAB B and MAB C. The data clearly demonstrates that the AR reduction technology works very effectively for other antibodies. It is also clear that the concentration ranges are different between different antibodies. The pH range chosen was related to the isoelectric point of the antibody and was chosen to be approximately 1 to 2 units less than the pI of the molecule.
  • TABLE 24
    AR reduction for molecule B
    Buffer Concentration % AR
    Resin System (mM) pH Yield (%) Reduction
    Poros XS Tris Acetate 120 7.5 57.2 8.4
    125 46.5 9.3
    130 37.1 10.3
    Nuvia S 85 72.5 16.6
    90 56.1 16.9
    95 44.2 17
    CM Hyper D 50 73 8.2
    55 62 9.2
    60 52.6 9.2
  • TABLE 25
    AR reduction for molecule C
    Concen-
    Buffer tration Yield Load % AR
    Resin System (mM) pH (%) % AR Reduction
    Poros XS Tris 40 6.0 87.4 15.6 8.5
    Acetate 45 56.8 15.7 12.8
    50 31.3 15.7 14.3
    Nuvia S 35 45.1 11.5 11.2
    37 28.5 15.4 15.2
    40 15.3 15.2 15.2
    CM Hyper 18 83.6 16.3 6.3
    D 20 64.9 16.3 11.2
    22 50.7 16.4 12.3
  • 6.2.4. Example CEX 4: Demonstration of AR Reduction with Different pH Conditions—Adalimumab
  • The AR species in the current invention is removed in the Flow through/Wash fraction. Therefore the binding pH is a key variable. The cation concentration that provides the desired performance will vary with the binding pH. Therefore for each binding pH, the experimental strategy outlined in Example 6.2.1. is carried out to determine the range of ion concentration that results in AR reduction.
  • The results of the experiments with different pHs for Adalimumab is shown in Table 26. As can be seen, at lower pH, the cation concentration required to achieve AR removal in the wash fraction is higher. It is unexpected that the AR reduction is significantly more robust and optimal at higher pHs (closer to pI) than at lower pHs. It is not obvious to one skilled in the art to operate a cation exchange chromatography at pH closer to pI as shown in Table 27. Literature data suggests an optimal pH of at least 3 units less than the pI of the molecule.
  • TABLE 26
    Effect of pH on AR reduction
    Buffer
    Concentration Yield % AR
    pH Resin Buffer System (mM) (%) Reduction
    5.5 Poros XS Tris Acetate 350 58.2 5.9
    6.5 225 61.4 6.4
    7 170 75.3 5.6
    7.5 140 78.6 6.8
    8 125 75.8 5.7
    7.5 CM Hyper D Ammonium 4 77.9 7.4
    Sulfate
    6 Sodium Chloride 45 86.1 4
    6.8 30 71.5 7
    7.5 10 71.3 6.8
    7.5 Tris Acetate 45 72.2 8.9
  • TABLE 27
    Effect of delta pH and pI on AR reduction
    pI- Buffer [Cation] Yield % AR
    pH Molecule Resin system (mM) (%) Reduction
    1.1 Adalimumab Poros XS Arginine/ 60/29 58.9 7.8
    Tris Acetate
    2.2 Sodium 125 73.5 5.4
    1.8 Chloride 75 90 1.5
    1.1 50 72.1 7.2
    3.1 Tris Acetate 350 58.2 5.9
    2.1 225 61.4 6.4
    1.6 170 75.3 5.6
    1.1 145 72.6 7.3
    0.6 125 75.8 5.7
    1.6 mAb B Poros XS Tris Acetate 120 57.2 8.4
    1.6 CM Hyper Tris Acetate 50 73 8.2
    D
    1.6 Nuvia S Tris Acetate 85 72.5 8.4
    1.0 mAb C Poros XS Tris Acetate 40 87.4 8.5
    1.0 CM Hyper Tris Acetate 18 83.6 6.3
    D
    1.0 Nuvia S Tris Acetate 35 45.1 11.2
  • 6.2.5. Example CEX 5: Demonstration of AR Reduction with Different Ion Concentrations—Adalimumab
  • Cation concentration is a key variable in the performance of cation exchange chromatography. For every combination of antibody/resin/pH there is a range of cation concentrations that provides AR reduction; the strategy outlined in Example 6.2.1 can be followed to determine the AR reduction and the corresponding recovery for each cation concentration.
  • Table 28 below shows the effect of cation concentration on AR reduction. The table also includes the effect of cation concentration for different pH values. The data demonstrates that the AR reduction can be effectively achieved over a range of cation concentrations at each pH and that the concentration ranges depend on the pH. The table also includes an example of the concentration range for a different cation type.
  • TABLE 28
    Effect of cation concentration and pH on AR reduction
    Cation
    concentration Yield % AR
    (mM) Buffer system pH Resin (%) Reduction
    60/29 Arginine/Tris 7.5 Poros XS 58.9 7.8
    65/29 Acetae 47.4 8.7
    23 80.5 5.8
    25 72.9 7.3
    27 52.2 9.5
    115 Sodium Chloride 6 85.4 4.2
    125 73.5 5.4
    130 48.7 7.1
    75 6.8 90 1.5
    90 53.7 2.1
    45 7.5 60.7 7.9
    50 72.1 7.2
    350 Tris Acetate 5.5 58.2 5.9
    375 38.4 7.4
    400 29.9 6.2
    225 6.5 61.4 6.4
    250 59.5 6.6
    275 37.6 7.8
    300 21.6 8.8
    165 7 83.8 4.3
    170 75.3 5.6
    175 70.3 5.7
    140 7.5 78.6 6.8
    145 72.6 7.3
    150 69.2 7.8
    175 29.8 10.3
    125 8 75.8 5.7
    130 67.7 6.5
    135 57.4 7.5
  • 6.2.6 Example CEX 6: Demonstration of Ar Reduction with Different Buffer Systems with Adalimumab
  • The cation type and concentration are key variables in Cation Exchange Chromatography. The invention has been demonstrated with Tris, Sodium/Tris, Ammonium/Tris and Arginine/Tris as cation types/mixtures with effective reduction of AR in each case. As one skilled in the art would appreciate the optimal pH and cation concentration is different for each cation type/mixture and was derived by using the strategy outlined in Example 6.2.1. Experiment were performed at pH 7.5. 29 mM Tris-acetate was used for pH control. Table 29 shows the data of AR reduction and corresponding recovery for the different cation types/mixtures.
  • TABLE 29
    Effect of cation types/mixtures on AR reduction
    Cation
    concentration Yield % AR
    Buffer System Resin (mM) pH (%) Reduction
    Arginine/Tris Poros XS 60 7.5 58.9 7.8
    acetate
    Ammonium 25 72.9 7.3
    Sulfate
    Sodium Chloride
    50 72.1 7.2
    Tris Acetate 140 78.6 6.8
    Ammonium CM Hyper D 4 77.9 7.4
    Sulfate
    Sodium Chloride
    10 71.3 6.8
    Tris Acetate 45 72.2 8.9
    Ammonium Nuvia S 11 66.6 12.6
    Sulfate
    Sodium Chloride
    20 75.9 10.5
    Tris Acetate 75 67.6 10
  • 6.2.7. Example CEX 7: Demonstration of AR Reduction with Different Loading
  • Furthermore, the strategy outlined in Example 6.2.1. to reduce acidic species through careful control of buffer cation type, concentration and pH can be applied to any range of protein loading which represents an operational mode of binding followed by elution, i.e. not overloaded or a column load factor below that of the adsorbents binding capacity. A range of relevant protein loadings for Poros XS at pH 7.5 using Tris as the cation is shown in Table 30 showing robust AR reduction.
  • TABLE 30
    Impact of Column loading
    Column
    Loading
    (g product/L Buffer Concentration % AR
    resin) System (mM) pH Yield (%) Reduction
    25 Tris 160 7.5 83.6 6.4
    30 155 79.4 6.0
    35 140 87.4 4.8
    38 140 83.5 5.0
    40 140 76.4 6.0
    42 140 74.5 5.7
    45 140 67.0 6.6
  • 6.2.8. Example CEX 18: Demonstration of AR Reduction with Different Load Concentration
  • Furthermore, the strategy outlined in Example 6.2.1. to reduce acidic species through careful control of buffer cation type, concentration and pH can be applied to any range of column feed streams of varying protein concentration. A range of varying protein load concentration for Poros XS at pH 7.5 using Tris as the cation is shown in Table 31 showing robust AR reduction.
  • TABLE 31
    Effect of Protein Load concentration
    Load
    Concen- Concen-
    tration Buffer tration Yield % AR
    (mg/mL) Resin System (mM) pH (%) Reduction
    3 Poros XS Tris 140 7.5 77.3 7
    4 Acetate 145 60.7 7
    5 140 78.7 6.7
    5 145 64.1 7
    6 145 59.5 6.9
    7 140 77.6 6.5
  • As described above, the Acidic Region for Adalimumab is further grouped into two regions termed AR1 and AR2, based on a certain retention time of the peaks seen on the WCX-10 method. The characteristics of the variants in these two regions are expected to be different and hence the methods that reduce variants belonging to these groups can be specifically delineated.
  • Further, in addition to achieving a certain AR reduction, it may be desirable to achieve a certain absolute level of AR levels, in consideration of reducing or removing certain variants. The capability of the current invention in achieving a certain absolute level of AR, AR1 and AR2 is demonstrated in Table 32.
  • The specific species comprising the AR1 species can be identified and quantitated, to demonstrate reduction of such species by methods of the current invention. Two of such species, Glycated mAb, and MGO modified mAb have been identified and shown to be reduced by the methods of this invention. While these species are among the Acidic Species part of the charge variants, the acidic species typically described in the literature is the deamidated mAb, which is distinctly different.
  • TABLE 32
    The final impurity level
    Cation Conc. Yield %
    Buffer System (mM) pH (%) Final AR1 % Final AR
    Arginine/Tris 60 7.5 58.9 0.3 5.8
    Acetate 65 7.5 47.4 0.3 4.7
    Ammonium Sulfate 23 7.5 80.5 0.6 8.3
    25 7.5 72.9 0 6.4
    27 7.5 52.2 0.4 5.0
    Sodium Chloride 115 6 85.4 1.3 10.2
    125 6 73.5 0 8.1
    135 6 48.7 0 6.1
    75 6.8 90 1.4 10.9
    90 6.8 53.7 0.7 11.2
    45 7.5 60.7 0 6.2
    50 7.5 72.1 0 7.8
    Tris Acetate 350 5.5 58.2 0 7.7
    375 5.5 38.4 0.1 6.2
    400 5.5 29.9 1.5 7.3
    225 6.5 61.4 0.8 7.2
    250 6.5 59.5 0 6.8
    275 6.5 37.6 0 5.6
    300 6.5 21.6 0 4.7
  • The method of the current invention can effectively reduce AR2 levels, as an overall decrease in AR levels is achieved. The method can be used to achieve a target absolute level, as exemplified by the data presented in Table 32.
  • The method of the current invention can effectively achieve AR reduction as well as achieve a target absolute level of acidic species as exemplified by the data presented in Table 32.
  • 6.2.9. Example CEX 9 Demonstration of Glycated and Methylglyoxylated Species Reduction
  • The strategy outlined in Example 6.2.1. to reduce acidic species through careful control of buffer cation type, concentration and pH can be further extended to specific post-translational modifications. While acidic species are defined in the application as impurities that are less retained than the main peak on an analytical weak cation exchange (WCX) HPLC column, specific known product related substances derived from cellular metabolism modification such as glycation and methylglyoxal (MGO) can be specifically identified as being part of the acidic species. FIG. 7 and FIG. 8 shows the outcome of in-vitro labeling experiments which demonstrate that glycation and MGO modified antibody are unique species that are resolved by the WCX method in the AR1 region of the chromatogram and can be enriched in vitro. Furthermore, the invention described here shows that glycated and MGO modified antibody can be effectively removed through the careful control of buffer cation type, concentration and pH using the CEX as described in Example 6.2.1. Quantitative reduction of AR1 and hence the Glycated and MGO species by CEX and CEX-Mixed Mode resins is show in Table 33 and Table 34.
  • TABLE 33
    Glycated species removal
    Load Load % AR1 % AR
    Buffer Conc. Yield % % Reduc- Reduc-
    Resin System (mM) pH (%) AR1 AR tion tion
    Poros XS Tris 135 7.5 54.0 40.8 58.6 30.8 34.8
  • TABLE 34
    MGO peak removal
    Concen-
    Buffer tration Yield % AR1 % AR
    Resin System (mM) pH (%) Reduction Reduction
    Toyo Pearl MX Tris 80 7.5 66.7 2.8 7.2
    TRP 650M
    Poros XS 145 64.1 2.7 7
    Nuvia S 90 48.5 3.1 9.6
  • 6.2.10 Example CEX 10: Demonstration of Lysine Distribution Modification
  • The strategy outlined in Example 6.2.1. to reduce acidic species also can be used to modulate the distribution of C-terminal Lys variants of monoclonal antibodies, a known post-translational modification leading to charge heterogeneity. Some minor changes in the distribution of Lys isoforms is expected through the reduction of acidic species as the WCX analysis is a compositional analysis. However, through careful control of buffer cation type, concentration and pH care, in addition to reducing acidic species, the elution pool can be enriched for the more basic isoforms (Lys 1 and Lys2). Table 35 and FIG. 9 depicts a non-limited example of the impact of pH and cation (Tris) concentration on basic isoform enrichment.
  • TABLE 35
    Change in Lysine distribution during CEX
    Chromatography - impact of Tris concentration
    Buffer
    % LYS0 % LYS1 Buffer Concentration
    decrease Increase % LYS2 Increase System (mM) pH
    1.6 4.4 2.7 Tris 350 5.5
    5 6.5 5.5 Acetate 375
    9.7 7.5 11.9 400
    1.9 5 2.9 225 6.5
    1.9 5.3 3 250
    6.1 7.4 6 275
    11.8 8.6 10.8 300
    0.2 5.2 1.6 140 7.5
    0.6 5.7 1.8 145
    1.8 6.8 2.4 150
    16.4 14.9 10.3 175
  • 6.2.11. Example CEX 11: Demonstration of HCP and Aggregate Reduction in Addition to AR Reduction
  • In the present invention, it has been demonstrated that HCP and aggregate levels can be effectively reduced by appropriate adjustment of the elution conditions, after washing off the AR enriched species in the flow through/wash fractions.
  • Table 36 and Table 37 shows the HCP and aggregate removal achieved along with AR reduction. The data clearly shows that other process related and product related substances/impurities can be achieved using the current invention on the CEX adsorbents, and hence functions as an effective polishing step in the large scale purification of monoclonal antibodies.
  • TABLE 36
    Aggregate removal during CEX Chromatography
    %
    % Aggregate % Fragment Monomer
    Resin Molecule Buffer system pH Reduction Reduction Increase
    CM Hyper D adalimumab 5 mM 7.5 0.04 0.17 0.2
    Ammonium
    Sulfate
    45 mM Tris 0.01 0.18 0.19
    Acetate
    Nuvia S 11.5 mM 0.16 0.17 0.33
    Ammonium
    Sulfate
    75 mM Tris 0.09 0.11 0.2
    Acetate
    22.5 mM 0.08 0.19 0.27
    Sodium
    Chloride
    Poros XS 27 mM 0.75 0.27 1.02
    Ammonium
    Sulfate
    140 mM Tris 0.51 0.41 0.92
    Acetate
    145 mM Tris 0.58 0.41 0.98
    Acetate
    Nuvia S mAb B 85 mM Tris 0.19 0.27 0.47
    Acetate
    Poros XS
    130 mM Tris 0.36 0.04 0.39
    Acetate
    Nuvia S mAb C 35 mM Tris 6.0 0.07 0.01 0.07
    Acetate
    Poros XS
    50 mM Tris 0.27 0 0.28
    Acetate
  • TABLE 37
    HCP Removal during CEX Chromatography
    Eluate Pool
    Buffer Load HCP HCP Reduction
    Resin Molecule system pH (ng/mg) (ng/mg) fold
    CM Hyper D adalimumab 5 mM 7.5 8105 3844 2.1
    Ammonium
    Sulfate
    45 mM Tris 8628 5615 1.5
    Nuvia S 11.5 mM 5314 2405 2.2
    Ammonium
    Sulfate
    75 mM Tris 17317 12845 1.4
    Acetate
    22.5 mM 9091 4115 2.2
    Sodium
    Chloride
    Poros XS 27 mM 21857 12574 1.0
    Ammonium
    Sulfate
    140 mM Tris 14732 9181 1.7
    Acetate
    145 mM Tris 15359 10113 1.6
    Acetate
    Nuvia S mAb B 85 mM Tris 735 319 2.3
    Acetate
    Poros XS
    130 mM Tris 2183 404 5.4
    Acetate
    Nuvia S mAb C 35 mM Tris 6.0 27 31 0.9
    Acetate
    Poros XS
    50 mM Tris 25 15 1.7
    Acetate
  • 6.2.11. Example CEX 12 Demonstration of Means of Controlling AR Reduction
  • Controlling the final product quality by modifying the process based on the quality of the intermediate material is an approach that has been proposed as an effective way of ensuring product quality, with the view of ensuring safety and efficacy.
  • Considering that the AR levels generated during cell culture and other upstream steps can be variable, it is desirable to design a downstream process step that implements a means of controlling the product quality; and to further have a specific means of controlling a process parameter to influence the quality of the product.
  • In the current invention, such a control is possible, as the cation concentration is a single parameter that can be modified to achieve a desired separation of the AR species. For example, to achieve a higher level of AR reduction, the Tris concentration of the loading material and the wash buffer can be decreased, such that the AR enriched species is collected in the flow through fraction.
  • As an example, and not to be restrictive in any manner, it has been demonstrated in this example that the AR levels can be controlled by changing the Tris concentration of the load and wash solutions. A pilot scale Poros XS column (10 cm diameter×22 cm height, 1.7 L), was used for this study.
  • The load material and the stock buffer are both prepared at 300 mM Tris concentration at the same pH. The AR level of the load material was measured to be X %. The load material and equilibration/wash buffer are in-line diluted to the target Tris concentration based on predetermined correlation between the AR levels and Tris concentration. As demonstrated in the example, when the Tris concentration was adjusted to 156 mM, a final AR reduction of 4.1% was achieved, whereas when the Tris conc. was adjusted to 150 mM, a final AR level of 3.1 was achieved etc (Table 38). This allows very predictable control of the AR levels ensuring achievement of the desired product quality.
  • TABLE 38
    Controlling AR Reduction using Process Analytical Technology approach
    Tris conc % AR
    (mM) Yield (%) Reduction
    156 51.9 4.1
    150 70.5 3.1
    131 95.3 1.3
  • In addition to the acidic species reduction demonstrated in Example CEX 1 through careful control of the pH cation type and concentration in the load (process stream) and equilibration/wash buffers, the composition of the elution buffer can also be used to further improve the product quality profiles. The impact of various cation types, concentration and pH were tested for eluting the product. There is a wide selection for elution buffer as shown in Table 39. The experiments were performed using Poros XS resin
  • TABLE 39
    Elution buffer types on aggregates removal
    % Aggregate
    Buffer System pH Yield (%) Reduction
    200 mM Sodium Sulfate/29 mM Tris 5.2 76.1 0.36
    Acetate
    160 mM Sodium Sulfate/29 mM Tris 5.2 82.3 0.82
    Acetate
    150M Sodium Sulfate/29 mM Tris 5.2 78.8 0.90
    Acetate
    140M Sodium Sulfate/29 mM Tris 5.2 78.2 1.00
    Acetate
    400 mM Sodium Sulfate/29 mM Tris 4.0 78.5 0.98
    Acetate
    100 mM Sodium Sulfate/140 mM Tris 5.2 70.9 1.25
    Acetate
    150 mM Sodium Sulfate/140 mM Tris 5.2 79.6 1.05
    Acetate
    140M Sodium Sulfate/140 mM Tris 5.2 75.4 1.07
    Acetate
    130 mM Sodium Sulfate/140 mM Tris 5.2 78.2 1.07
    Acetate
    300 mM Sodium Sulfate/30 mM Tris 4.6 80.3 0.57
    Acetate
    150 mM Sodium Sulfate/29 mM Tris 7.5 75.0 0.92
    Acetate
  • 6.2.13. Example CEX 13 Demonstration of Ar Reduction with Cation-HIC Mixed Mode Resin
  • The strategy outlined in Example 6.2.1. to reduce acidic species through careful control of buffer cation type, concentration and pH can be expanded to include other chromatography adsorbents such as mixed mode or multi-modal absorbents which include a cation exchange mechanism. Table 40 outlines the conditions used and the AR reduction achieved for two cation-hydrophobic interaction mixed mode resins. The data clearly shows that the technology outlined in Example 6.2.1. is robust in delivering AR reduction for these types of resins across in addition to traditional cation exchange adsorbents. As described in Example 6.2.1, the AR reduction can be balanced with recovery and an optimal condition can be chosen. As a further demonstration, molecule 2 was also evaluated (Table 41) with the same outcome showing the same relationship between cation concentration, recovery and AR reduction. As previously shown in Example 6.2.6, the optimal condition for different molecules varies. Furthermore, this technology when applied to CEX-HIC mixed mode resins also shows reduction of impurities as previously described.
  • TABLE 40
    Adalimumab AR Reduction by Cation Exchange Mixed Mode
    Chromatography
    Tris
    Concentration Yield % AR
    Resin Buffer System (mM) pH (%) Reduction
    Nuvia C Prime Tris Acetate 70 7.5 63.8 6.5
    72.5 7.5 61.1 6.0
    75 7.5 57.1 6.7
    Toyo Pearl 75 7.5 80 5.7
    MX Trp 650M 80 7.5 66.7 7.2
    85 7.5 51.8 8.6
  • TABLE 41
    Molecule B AR Reduction by Cation Exchange Mixed
    Mode Chromatography
    Concentration Yield % AR
    Resin Buffer System (mM) pH (%) Reduction
    Nuvia C Prime Tris Acetate 75 7.5 86.0 2.0
    85 7.5 74.6 5.9
    95 7.5 61.3 6.8
    Toyo Pearl 90 7.5 81.1 6.4
    MX Trp 650M 95 7.5 68.8 8.8
    100 7.5 53.5 10.7
  • As described in previous sections, the Acidic Region for Adalimumab is further grouped into two regions termed AR1 and AR2, based on a certain retention time of the peaks seen on the WCX-10 method. The characteristics of the variants in these two regions are expected to be different and hence the methods that reduce variants belonging to these groups can be specifically delineated.
  • Further, in addition to achieving a certain AR reduction, it may be desirable to achieve a certain absolute level of AR levels, in consideration of reducing or removing certain variants. The capability of the current invention in achieving a certain absolute level of AR, AR1 and AR2 is demonstrated in Table 42A with Tables 42B and 42C indicating the levels of additional process-related impurities or product-related substances.
  • The specific species comprising the AR1 species can be identified and quantitated, to demonstrate reduction of such species by methods of the current invention. While these species are among the Acidic Species part of the charge variants, the acidic species typically described in the literature is the deamidated mAb, which is distinctly different. These results show that the Cation Exchange Resin with additional pendant hydrophobic interaction functionality, is able to provide AR reduction effectively, similar to the CEX Adsorbents.
  • TABLE 42A
    Final acidic species level for Adalimumab
    Tris
    Buffer Concentration Yield Final Final Final
    Resin System (mM) pH (%) % AR1 % AR2 % AR
    Nuvia C Prime Tris Acetate 70 7.5 63.8 0.39 4.64 5.03
    72.5 7.5 61.1 0.36 4.4 4.75
    75 7.5 63.8 0.39 4.06 4.45
    Toyo Pearl MX 75 7.5 80 0.6 4.2 4.8
    Trp 650M 80 7.5 66.7 0.5 3.2 3.7
    85 7.5 51.8 0.2 2.2 2.4
  • TABLE 42B
    Aggregates/Fragments Reduction by Cation Exchange Mixed Mode
    Chromatography
    %
    Buffer % Aggregate % Fragment Monomer
    Resin Molecule System pH Reduction Reduction Increase
    Nuvia C prime adalimumab 70 mM Tris 7.5 0.3 0.34 0.63
    Toyo Pearl MX Trp 75 mM Tris 0.08 0.56 0.65
    650M
    Nuvia C prime Molecule B 85 mM Tris 0.87 1.18 2.04
    Toyo Pearl MX Trp 95 mM Tris 0.0 1.8 1.8
    650M
  • TABLE 4C
    HCP Reduction by Cation Exchange Mixed Mode Chromatography
    Eluate pool
    Load HCP HCP Fold
    Resin Compound Buffer pH (ng/mg) (ng/mg) Reduction
    Toyo Pearl MX Trp 650M adalimumab 70 mM Tris 7.5 202.6 38.9 5.2
    Nuvia C prime 75 mM Tris 205.5 72.8 2.8
    Toyo Pearl MX Trp 650M Molecule B 95 mM Tris 983.3 137.1 7.2
    Nuvia C prime 85 mM Tris 1011.3 88.2 11.5
  • 6.2.14. Example CEX 14 Demonstration of Ar Reduction in Process Combinations
  • The method described above for reducing acidic species using cation exchange can be used as an independent operation or in combination with other process steps that provide additional acidic species reduction or those providing additional complementary and supplementary purification (See tables 43-50). The following process combinations are provided here as non-limiting examples
  • 1. Affinity→MM→CEX
  • 2. Affinity→AEX→CEX
  • 3. Affinity→CEX
  • 4. CEX Capture→CEX
  • 6.3. Mixed Mode Chromatography Examples 6.3.1. Example MM1: Resin: Buffer Combination
  • In this example one of the approaches outlined in the general description was employed to determine the operating conditions to implement the invention. Specifically, a response surface design DOE was applied to evaluate mAb AR reductions and recovery yields.
  • The demonstration of the current invention for a specific antibody & resin is provided in this example, and consists of
  • 1. Choosing a pH in the range of 6.8 to 8.4.
  • 2. Choosing a conductivity in the range of 2.3 to 13.7 mS/cm.
  • 3. Determining the acidic species distribution in the ft/wash fractions.
  • 4. Choosing an optimal pH and conductivity that provides the desired acidic species levels and recovery
  • In this example, Adalimumab and resin Capto Adhere were chosen. The experiments were performed with Tris/Acetate buffer system at target pH and conductivity listed in Table 51 The load material was from Protein A affinity capture and pH adjusted. This study demonstrated the effect of loading pH and conductivity on acidic species reduction. The acidic species reduction can be significantly affected by operating pH. AR reduction increased with increasing pH and/or decreasing conductivity (Table 51, Table 52 and FIG. 10)
  • TABLE 51
    DOE study condition
    Tris Acetate Edge points for Response
    Buffer Range Surface
    pH 7.0-8.2  6.8, 8.4 
    Conductivity 4.0-12.0 2.3, 13.7
  • TABLE 52
    DOE Study Operating Conditions and Results
    DOE exp pH Conductivity (mS/cm) ΔAR (%) Yield (%)
    1 7.0 4.0 0.4 83
    2 7.6 8.0 0.4 73
    3 7.6 2.3 1.3 82
    4 7.6 8.0 0.6 68
    5 7.6 8.0 0.2 70
    6 7.6 8.0 −0.2 69
    7 8.2 4.0 2.1 67
    8 7.6 8.0 1.3 69
    9 7.0 12.0 −0.2 70
    10 7.6 8.0 1.2 71
    11 8.2 12.0 1.4 74
    12 6.8 8.0 1.2 76
    13 8.4 8.0 1.8 67
    14 7.6 8.0 1.4 71
    15 7.6 13.7 1.0 74
    16 7.6 8.0 1.6 70
    Note:
    AR reductions and protein recovery yields were calculated based on the Flow Through fractions at about loading 200 g protein per L of resin.
  • 6.3.2. Example MM 2: Fraction Pooling
  • In this example, Adalimumab and resin Capto Adhere were chosen. The experiments were performed with Tris/Acetate buffer system at pH 7.85 and conductivity of 2.5 mS/cm. The load material was from Protein A affinity capture and pH adjusted. Column flow through was fractionated throughout the entire load and wash phases. Each fraction was analyzed for acidic species and protein recovery. FIG. 11, FIG. 12 and Table 53 demonstrate AR reduction achieved with the corresponding recovery. These AR reductions and recoveries correspond to the cumulative pools of the fractions from the start to the various points during the load/wash. This is depicted in Table 53 where the AR reductions corresponding to each of these pools. This data is plotted in FIG. 11.
  • TABLE 53
    Cumulative AR reduction in Flowthrough/wash fractions
    Flowthrough Fraction Yield Δ AR1 Δ AR2 Δ AR ΔLys
    (Load & wash) (%) (%) (%) (%) (%)
    A2 23 2.56 3.13 5.69 5.61
    A2 + A3 45 2.31 2.19 4.49 4.37
    A2 + A3 + A4 58 1.83 1.89 3.72 3.63
    A2 + A3 + A4 + A5 65 1.57 1.58 3.15 3.06
    A2 + A3 + A4 + A5 + A6 73 1.38 1.32 2.70 2.61
    A2 + A3 + A4 + A5 + A6 + B7 86 1.26 1.12 2.38 2.30
    A2 + A3 + A4 + A5 + A6 + 89 1.19 0.91 2.09 2.02
    B7 + B6
    A2 + A3 + A4 + A5 + A6 + 90 1.14 0.82 1.96 1.89
    B7 + B6 + B5
    Note:
    “A” Fractions are load fractions and
    “B” Fractions are wash fractions
  • 6.3.3. Example MM 3: Demonstration of AR Reduction with Mixed Mode Adsorbents
  • In this example, Adalimumab was chosen. The experiments were performed with Tris/Acetate buffer system at pH 7.85 and conductivity of 2.5, 3.5, and 4.5 mS/cm. The same load material was applied to different mixed mode resin columns. The load material was from Protein A affinity capture and pH adjusted. Table 54 shows that all three mixed mode resins could reduce mAb acidic species. Due to the differences of resin ligands, the AR reduction level may slightly vary under certain conditions.
  • TABLE 54
    Adalimumab AR Reduction and Protein Recovery Yields Processed with Different Mixed Mode media
    Tris/Ac Buffer
    Capto Adhere HEA PPA
    Operating pH 7.85 pH 7.85 pH 7.85
    Conditions 4.5 mS/cm 3.5 mS/cm 2.5 mS/cm 4.5 mS/cm 3.5 mS/cm 2.5 mS/cm 4.5 mS/cm 3.5 mS/cm 2.5 mS/cm
    Yield (%) 50 52 58 49 52 56 40 43 47
    AR Reduction 1.8 3.8 3.7 1.1 2.7 3.2 1.4 2.2 3.5
    (%)
    Yield (%) 68 71 73 65 75 69 61 64 63
    AR Reduction 1.1 2.7 2.7 0.5 1.8 2.1 0.4 1.9 2.6
    (%)
  • 6.3.4. Example MM 4 Demonstration of Ar Reduction with Other Antibodies: mAb B and mAb C
  • In this example, another two different monoclonal antibodies besides Adalimumab and resin Capto Adhere was chosen. The experiments were performed with Tris/Acetate buffer system at multiple pH and conductivity condition. The load materials of all mAbs were from Protein A affinity capture and pH adjusted. MAB C was also applied to another two MM resins besides Capto Adhere under the same operating conditions. The Table 55 outlines the operating conditions and the AR reduction achieved and the corresponding recovery achieved. The results demonstrate that the technology can also reduce acidic species for other monoclonal antibodies with optimal pH and conductivity conditions. Experiments were performed with Tris-acetate buffer system.
  • TABLE 55
    AR Reductions and Protein Recovery for different mAb
    with Capto Adhere columns
    conductivity
    mAb pH (mS/cm) ΔAR (%) Yield (%)
    D2E7 7.85 3.5 3.8 52
    7.85 2.5 3.7 58
    MAB B 6.8 3.0 6.3 51
    6.8 4.5 4.2 53
    7.0 3.0 5.1 77
    8.0 3.0 3.4 60
    MAB C 9.0 3.0 5.3 73
    8.5 3.0 3.5 54
    8.0 3.0 3.7 50
  • FIG. 13 displays the MAB B cumulative pool AR broke through the column of Capto Adhere operated at pH 7.0 and conductivity of 3.0 mS/cm with Tris-Acetate buffer. FIG. 14 shows the MAB C cumulative pool AR broke through the column of Capto Adhere operated at pH 8.5 and conductivity of 3.0 mS/cm with Tris-Acetate buffer. Both of graphs demonstrate similar AR breakthrough curves with different AR values comparing to adalimumab (FIG. 12). FIG. 15 presents the AR breakthrough curves of Mab C with three different mixed mode resins with Tris-acetate buffer operated at pH 8.5 and conductivity of 3.0 mS/cm. The data clearly demonstrates that the AR reduction technology using mixed mode resins works very effectively for other antibodies.
  • 6.3.5. Example MM 5: Demonstration of Relative pH on AR Reduction with Different Resins Using D2E7 Antibody Material
  • In this example, data compiled from different experiments is shown to demonstrate the impact of the pH choice, relative to the pI of the protein on AR reduction. This data set provides the basis for one skilled in the art to determine a pH range to implement the current invention. Further, this reiterates the fact that the pH choice depends on several factors and the relationship between pH and AR reduction is also mAb dependent. FIG. 16 demonstrates the impact of pH-pI and conductivity on AR reduction which compiled data from the experiments performed with Capto Adhere under conditions listed in Table 56. FIG. 17 shows the impact of pH-pI and conductivity on MAB B AR reduction including the experiments operated with Tris/Acetate buffer system and multiple mixed mode resins under the conditions listed in Table 57. FIG. 18 shows the impact of pH-pI and conductivity on MAB C AR reduction including the experiments operated with Tris/Acetate buffer system and multiple mixed mode resins under the conditions listed in FIG. 16. All the load materials were from Protein A affinity capture and pH adjusted. It is also clear that the AR reduction can be achieved with the present invention with a range of pH choices, in the range of +0.5 to −2.5 pH units from pI for D2E7. One skilled in the art can choose an appropriate pH to achieve a target AR reduction.
  • TABLE 56
    Operating conditions and AR reductions for D2E7
    Buffer system pH pH-pI Conductivity (mS/cm) AR reduction
    Tris/Ac 7 −2.02 4 0.4
    7.6 −1.42 8 0.4
    7.6 −1.42 2.3 1.3
    7.6 −1.42 8 0.6
    7.6 −1.42 8 0.2
    7.6 −1.42 8 −0.2
    8.2 −0.82 4 2.1
    7.6 −1.42 8 1.3
    7 −2.02 12 −0.2
    7.6 −1.42 8 1.2
    8.2 −0.82 12 1.4
    6.8 −2.27 8 1.2
    8.4 −0.57 8 1.8
    7.6 −1.42 8 1.4
    7.6 −1.42 13.7 1.0
    7.6 −1.42 8 1.6
    7.5 −1.52 3.75 1.7
    7.6 −1.42 2.5 2.7
    7.6 −1.42 2.5 2.0
    7.6 −1.42 5 1.3
    7.6 −1.42 5 1.1
    7.85 −1.17 2 3.5
    7.85 −1.17 3.75 3.2
    7.85 −1.17 3.75 2.1
    7.85 −1.17 3.75 2.8
    7.85 −1.17 3.75 2.2
    7.85 −1.17 5.5 2.1
    8.1 −0.92 2.5 5.0
    8.1 −0.92 2.5 2.6
    8.1 −0.92 5 −0.2
    8.1 −0.92 5 −1.1
    8.2 −0.82 3.75 2.9
    Arg/Ac 8.5 −0.52 1 6.8
    9.0 −0.02 1 6.5
    9.5 0.48 1 1.9
    Trol/Ac 7.85 −1.17 1 5.7
    8.0 −1.02 1 8.0
    8.5 −0.52 1 6.0
  • TABLE 57
    Operating conditions and AR reductions for MAB B
    pH pH-pI Conductivity (mS/cm AR reduction
    Capto Adhere 6.8 −0.45 3 6.3
    7 −0.25 3 6.2
    7.5 0.25 3 4.0
    8 0.75 3 3.2
    6.8 −0.45 4.5 4.1
    7.5 0.25 4.5 3.3
    PPA 6.8 −0.45 3 1.1
    7 −0.25 3 0.9
    7.5 0.25 3 1.3
    8 0.75 3 0.5
    6.8 −0.45 4.5 1.6
    7.5 0.25 4.5 3.0
    HEA 6.8 −0.45 3 1.8
    7 −0.25 3 1.4
    7.5 0.25 3 3.6
    8 0.75 3 0.7
    6.8 −0.45 4.5 2.2
    7.5 0.25 4.5 0.9
  • TABLE 58
    Operating conditions and AR reductions for MAB C
    pH pH-pI Conductivity (mS/cm) Δ% AR
    Capto Adhere 8.0 −1.11 1 1.5
    8.5 −0.61 1 3.5
    9.0 −0.11 1 5.4
    PPA 8.0 −1.11 1 −0.4
    8.5 −0.61 1 1.1
    9.0 −0.11 1 2.1
    HEA 8.0 −1.11 1 −1.6
    8.5 −0.61 1 1.9
    9.0 −0.11 1 2.8
  • 6.3.6. Example MM 6: Effect of pH on AR Reduction
  • Response surface design DOE was applied to evaluate the impact of pH and conductivity on mAb AR reductions. In this example, Adalimumab and Capto Adhere were chosen. The experiments were performed with Tris/Acetate buffer system. The load material was from Protein A affinity capture and pH adjusted. Besides the pH and conductivity ranged tested and demonstrated in Table 59 and Table 60, higher pH ranges were also studied (FIG. 19).
  • The results in FIG. 19 and FIG. 20 demonstrated that mAb acidic species can be reduced at wide pH range from 6.8 to 9.5.
  • TABLE 59
    DOE study condition
    Tris Acetate Edge points for Response
    Buffer Range Surface
    pH 7.0-8.2  6.8, 8.4 
    Conductivity 4.0-12.0 2.3, 13.7
  • TABLE 60
    AR reduction and Yield in DOE study
    Experiment # pH Conductivity ΔAR Yield
    1 7.0 4.0 0.4 83
    2 7.6 8.0 0.4 73
    3 7.6 2.3 1.3 82
    4 7.6 8.0 0.6 68
    5 7.6 8.0 0.2 70
    6 7.6 8.0 −0.2 69
    7 8.2 4.0 2.1 67
    8 7.6 8.0 1.3 69
    9 7.0 12.0 −0.2 70
    10 7.6 8.0 1.2 71
    11 8.2 12.0 1.4 74
    12 6.8 8.0 1.2 76
    13 8.4 8.0 1.8 67
    14 7.6 8.0 1.4 71
    15 7.6 13.7 1.0 74
    16 7.6 8.0 1.6 70
    Note:
    AR reductions and protein recovery yields were calculated based on the Flow Through fractions at about loading 200 g protein per L of resin
  • 6.3.7. Example MM 7: Demonstration of Ar Reduction with Different Ion Concentrations—Adalimumab
  • In this example, adalimumab was chosen. Besides the conductivity range tested presented before, lower conductivity and higher conductivity ranges were also studied with the Capto Adhere. Table 61 and Table 62 display the DOE study conditions using Capto Adhere columns with Tris/Acetate buffer system. The load material was from Protein A affinity capture and pH adjusted. Column flow through pool was collected in each run from 50 mAU of UV A280 on the ascending and 150 mAU on the descending side of the peak. FIG. 21 demonstrates the effect of pH (6.8 to 8.4), conductivity (2.3 to 13.7 mS/cm), and protein load amount (116 to 354 g/L). FIG. 20 demonstrates the AR reduction at conductivity as low as—1 mS/cm. Table 63 demonstrates the AR reduction at conductivity 86 mS/cm with Ammonia Sulfate-Tris-Acetate buffer system.
  • The results demonstrated that mAb acidic species can be reduced at wide conductivity ranges from 1 to 86 mS/cm.
  • TABLE 61
    DOE study condition
    Tris Acetate Edge points for Response
    Buffer Range Surface
    pH 7.6-8.1 7.5, 8.2
    Conductivity 2.5-5.0 2.0, 5.5
    Protein load 150-320 116, 354
    amount (g/L)
  • TABLE 62
    DOE operting condition and results
    Load
    pH Conductivity (mS/cm) amount (g/L) ΔAR (%) Yield (%)
    7.5 3.75 235 1.7 89
    7.6 2.5 150 2.7 94
    7.6 2.5 320 2.0 95
    7.6 5 150 1.3 97
    7.6 5 320 1.1 103
    7.85 2 235 3.5 94
    7.85 3.75 116 3.2 86
    7.85 3.75 235 2.1 90
    7.85 3.75 235 2.8 90
    7.85 3.75 354 2.2 91
    7.85 5.5 235 2.1 92
    8.1 2.5 150 5.0 80
    8.1 2.5 320 2.6 87
    8.1 5 150 −0.2 95
    8.1 5 320 −1.1 98
    8.2 3.75 235 2.9 90
  • TABLE 63
    AR reduction and protein recovery at conductivity of 86 mS/cm
    and pH 7.9
    Conductivity
    (mS/cm) pH Yield (%) ΔAR (%)
    86 7.9 62 2.7
    87 2.0
    91 1.8
    86 7.9 59 1.4
    81 1.1
    94 0.7
    Note:
    Adalimumab in Protein A eluate containing 25 mM acetate and 18 mM Tris or 0.89 mM Tris were pH adjusted to pH 3.5 with 3M Acetic acid solution and neutralized to pH 7.9 with 3M Tris solution. One part of this viral inactivated material was then diluted by adding 0.3 part of a stock buffer containing 2.2M (NH4)2SO4/90 mM Tris/60 mM Acetic pH 7.9 to reach conductivity of 86 mS/cm.
  • 6.3.8. Example MM 8: Demonstration of AR Reduction with Different Buffer Systems with Adalimumab
  • In this example, molecule Adalimumab and resin Capto Adhere were chosen. The experiments were performed with different buffer systems listed in the tables below at multiple pH and conductivity condition. The load material pH was adjusted from Protein A eluate or CEX eluate. The results in Table 64 and Table 65 demonstrates that mAb acidic species can be reduced using various buffer systems.
  • TABLE 64
    Effect of Cation type on mAb acidic species reduction and recovery yield
    Capto Adhere HEA PPA
    Operating pH 7.85 pH 7.85 pH 7.85
    Condition 4.5 mS/cm 3.5 mS/cm 2.5 mS/cm 4.5 mS/cm 3.5 mS/cm 2.5 mS/cm 4.5 mS/cm 3.5 mS/cm 2.5 mS/cm
    Tris/Ac % Yield 50 52 58 49 52 56 40 64 63
    Δ% AR 1.8 3.8 3.7 1.1 2.7 3.2 1.4 1.9 2.6
    Operating ~1 mS/cm ~1 mS/cm ~1 mS/cm
    Condition pH 8.5 pH 9.0 pH 9.5 pH 8.5 pH 9.0 pH 9.5 pH 8.5 pH 9.0 pH 9.5
    Arg/Ac % Yield 65 62 49 77 71 66 69 70 71
    Δ% AR 8.6 6.5 1.9 4.9 3.5 N/R 4.5 1.9 0.6
    Operating ~1 mS/cm ~1 mS/cm ~1 mS/cm
    Condition pH 7.85 pH 8.0 pH 8.5 pH 7.85 pH 8.0 pH 8.5 pH 7.85 pH 8.0 pH 8.5
    Trol/Ac % Yield 62 54 49 69 64 58 64 64 590
    Δ% AR 4.1 6.0 4.6 1.7 2.9 3.0 1.4 2.1 2.1
    Note:
    Load material was adalimumab from Protein A affinity capture and pH adjusted
  • TABLE 65
    Effect of Cation type on mAb acidic species reduction and recovery yield
    load amount conductivity Yield
    Buffer (g/L) (mS/cm) pH (%) Δ% AR
    Tris/Ac1 200 4.00 7.80 90 1.6
    NaPhiosphaste/ 200 3.53 7.87 87 1.5
    Citrate/Trolamine/
    NaCl2
    1Load material was adalimumab from Protein A affinity capture and pH adjusted
    2The load material was adalimumab from CEX capture and pH adjusted
  • 6.3.9. Example MM 9: Demonstration of AR Reduction with Different Loading
  • The experiments were performed with Tris/Acetate buffer system under the conditions in table 62. The load material was adalimumab from Protein A affinity capture and pH adjusted. Column flow through pool was collected in each run from 50 mAU of UV A280 on the ascending and 150 mAU on the descending side of the peak. As seen from the profile (FIG. 22), the loading capacity has an impact on AR reduction but the AR reduction can be achieved over a wide range of loading capacities, and is merely a trade-off between AR reduction and recovery.
  • 6.3.10 Example MM 10: Demonstration of AR Reduction with Different Load Concentration
  • In this example, Capto Adhere was chosen. The experiment was performed with Tris/Acetate buffer system at pH 7.8±0.1 and conductivity 3.0±0.05 mS/cm. The load material was adalimumab from concentrated CEX capture and pH adjusted. The prepared load material was then split to be two parts. One was directly loaded on to a Capto adhere column; the other part was diluted 2 folds with equilibration buffer to make different protein concentration. Table 66 demonstrates that the load protein concentration did not have significant impact on mAb acidic species reduction.
  • TABLE 66
    Adalimumab AR Reduction and Yield with Different Load Protein
    Concentration
    Load Load
    Capture amount Conductivity protein Yield
    step Buffer (g/L) (mS/cm) pH conc. (g/L) (%) Δ% AR
    CEX Tris/Ac 200 2.9 7.8 22.0 87 2.4
    CEX Tris/Ac 200 3.0 7.7 11.0 89 2.1
    CEX NaPhiosphaste/ 200 3.5 7.9 4.9 87 1.5
    Citrate/
    Trolamine/
    NaCl
    Protein A Tris/Ac 200 3.1 7.8 9.0 89 2.5
    Protein A Tris/Ac 200 4.0 7.8 11.8 90 1.6
    Protein A Tris/Ac 200 3.0 7.8 9.9 93 2.4
    Protein A Tris/Ac 208 3.0 7.8 8.4 95 3.2
    Protein A Tris/Ac 222 3.0 7.9 12.9 89 3.4
  • 6.3.11. Example MM 11: Alternative Wash Modalities
  • In this example, molecule Adalimumab and resin Capto Adhere were chosen. The experiments were performed with Tris/acetate buffer system and the load material pH was adjusted from Protein A eluates. The equilibration buffer for both run was Tris/Acetic acid pH 7.8±0.1 and conductivity of 3.0±0.1 mS/cm. In the gradient conductivity wash study, second buffer was Tris/Acetic acid pH 7.8±0.1 and conductivity 6.0 mS/cm.
  • The results demonstrated that post load pH and conductivity can be varied with minimal AR reduction impacted.
  • TABLE 67
    Comparison of AR reduction and yield under different wash conditions
    Load Load
    conductivity conc Yield Wash
    Experiment Wash (mS/cm) load pH (mg/mL) (%) CV Δ% AR
    Equilibration Equilibration buffer (Tris/Ac pH 3.09 7.85 9.04 89 16.4 2.5
    buffer wash 7.8 and 3.0 mS/cm) wash only
    Gradient 1CV Equilibration buffer 3.04 7.78 7.17 91 8.0 2.2
    conductivity 10CV gradient conductivity
    wash wash from 100% Tris/Ac pH
    7.8, 3.0 mS/cm to 100% Tris/Ac
    pH 7.8, 6 mS/cm,
  • 6.3.12. Example MM 12: Demonstration of Achievement of Absolute Value of AR Levels in Antibody Preparations Using Mixed Mode Chromatography
  • In this example, molecule Adalimumab was chosen. The experiments were performed with multiple buffer systems and multiple MM absorbents under conditions listed in Table 68. The load materials pH was adjusted from Protein A eluates.
  • The Acidic Region for Adalimumab is further grouped into two regions termed AR1 and AR2, based on a certain retention time of the peaks seen on the WCX-10 method. The characteristics of the variants in these two regions are expected to be different and hence the methods that reduce variants belonging to these groups can be specifically delineated. Further, in addition to achieving a certain AR reduction, it may be desirable to achieve a certain absolute level of AR levels, in consideration of reducing or removing certain variants. The capability of the current invention in achieving a certain absolute level of AR, AR1 and AR2 is demonstrated in Table 68.
  • TABLE 68
    Acidic species level in MM resin flowthrough
    Conductivity Yield FT % FT %
    Resin Buffer pH (mS/cm) (%) AR1 AR2
    Capto Tris/Ac 7.85 4.5 50 2.8 9.7
    Adhere 7.85 4.5 68 3.0 10.3
    7.85 3.5 52 1.6 10.0
    7.85 3.5 71 2.2 10.5
    7.85 3.0 93 3.2 9.7
    7.85 2.5 58 1.7 9.4
    7.85 2.5 72 2.2 10.0
    Arg/Ac 8.5 1 65 1.2 6.1
    9.0 1 62 1.6 7.2
    9.5 1 49 0.8 11.8
    Trol/Ac 7.9 1 44 1.5 6.6
    7.9 1 62 1.8 8.0
    8.0 1 37 1.1 5.8
    8.0 1 54 1.2 7.7
    8.5 1 32 1.7 9.0
    8.5 1 49 1.9 10.1
    HEA Arg/Ac/ 8.5 1 77 1.6 8.5
    9.0 1 71 0.8 12.0
    PPA 8.5 1 69 2.2 8.7
    9.0 1 70 1.0 13.5
    9.5 1 71 0.7 13.1
  • 6.3.13. Example MM 13: Demonstration of HCP and Aggregate Reduction in Addition to AR Reduction
  • Besides the acidic species reduction, the MM adsorbent is able to reduce other product/process related substances/impurities effectively. In the implementation of the current invention the fact that AR reduction is effected, other impurities/substances are expected to be cleared significantly as they should bind stronger than the acidic species. The data shown in Table 69 and Table 70 demonstrates significant HCP and aggregate reductions with different resins, buffer systems, pH, conductivities and molecules
  • TABLE 69
    Aggregates reduction
    Conductivity
    (mS/cm) pH Buffer medium Δ% HMW
    D2E7 3.75 7.5 Tris/Ac Capto Adhere 0.7
    2.5 7.6 Tris/Ac 0.9
    2 7.85 Tris/Ac 0.9
    3.75 7.85 Tris/Ac 1.0
    5.5 7.85 Tris/Ac 0.7
    2.5 8.1 Tris/Ac 1.0
    3.75 8.2 Tris/Ac 0.8
    4.0 8.2 Tris/Ac 1.0
    8.0 6.8 Tris/Ac 0.2
    8.0 8.4 Tris/Ac 1.0
    1.0 8.5 Arg/Ac Capto Adhere 0.5
    1.0 9.0 Arg/Ac 0.8
    1.0 9.5 Arg/Ac 0.9
    1.0 8.5 Arg/Ac HEA 0.4
    1.0 9.0 Arg/Ac 2.5
    1.0 9.5 Arg/Ac 0.7
    1.0 8.5 Arg/Ac PPA 0.5
    1.0 9.0 Arg/Ac 2.8
    1.0 9.5 Arg/Ac 0.4
    MAB C 3.0 8 Tris/Ac Capto Adhere 1.0
    3.0 8.5 Tris/Ac Capto Adhere 1.1
    3.0 9 Tris/Ac Capto Adhere 0.6
    3.0 8 Tris/Ac PPA 0.7
    3.0 8.5 Tris/Ac PPA 0.5
    3.0 8 Tris/Ac HEA 0.7
    3.0 8.5 Tris/Ac HEA 0.6
  • TABLE 70
    HCP Log reduction
    Condutivity
    (mS/cm) pH Buffer medium HCP LRF
    D2E7 3.75 7.5 Tris/Ac Capto Adhere 1.5
    2.5 7.6 Tris/Ac 1.7
    2.0 7.85 Tris/Ac 2.2
    3.75 7.85 Tris/Ac 1.9
    5.5 7.85 Tris/Ac 1.4
    2.5 8.1 Tris/Ac 2.3
    3.75 8.2 Tris/Ac 2.1
    4.0 8.2 Tris/Ac 1.7
    8.0 6.8 Tris/Ac 0.3
    8.0 8.4 Tris/Ac 0.7
    MAB B 3 6.8 Tris/Ac Capto Adhere 2.0
    4.5 6.8 Tris/Ac Capto Adhere 1.3
    3 6.8 Tris/Ac PPA 1.2
    4.5 6.8 Tris/Ac 1.2
    3 6.8 Tris/Ac HEA 1.3
    4.5 6.8 Tris/Ac 1.1
  • 6.3.14. Example MM 14: Combinations of MM with Alternative Separation Strategies
  • Acidic Species Reduction by MM Adsorbents is expected to be performed after capture of the antibody by other means, or after one or more intermediate steps following the capture step. In the examples below the MM Adsorbent steps were performed either following a Cation Exchange Capture step or Protein A affinity capture step. As shown in Table 71, AR reduction was achieved at two different conductivities following Protein A Chromatography and CEX Chromatography.
  • TABLE 71
    AR Reduction with different source materials
    Capture Buffer conductivity (mS/cm) pH Yield (%) Δ% AR
    Protein A Tris/Ac 3.1 7.8 89 2.5
    Protein A Tris/Ac 4.0 7.8 90 1.6
    CEX Tris/Ac 2.9 7.8 87 2.4
    CEX Tris/Ac 3.0 7.7 89 2.1
  • Adalimumab was purified by a CEX chromatography step followed with a low pH viral inactivation step. The filtered viral inactivated material was buffer exchanged and loaded onto a Capto Adhere column. The flowthrough of Capto Adhere material was then purified with a HIC column with bind/elute mode. As shown in Table 72, AR reduction was achieved primarily with MM step, with some contribution from other steps.
  • TABLE 72
    Complete Process train with CEX Chromatography Capture -
    AR Reduction
    Δ% AR Δ% Lys Yield (%)
    CEX eluate n/a n/a n/a
    MM Load 0.29 0.34 90%
    MM Flowthrough 2.57 2.57 93%
    HIC eluate 0.95 0.94 97%
  • Adalimumab was purified by a Protein A chromatography step followed with a low pH viral inactivation step. The filtered viral inactivated material was buffer exchanged and loaded onto a Capto Adhere column. The flowthrough of Capto Adhere material was then purified with a HIC column with bind/elute mode as well as Flow Through mode. As shown in Table 73, AR reduction was achieved primarily with MM step, with some contribution from other steps.
  • TABLE 73
    Complete Process Train with Protein A Capture - AR, HMW and HCP
    reduction
    % AR % HMW HCP
    Process Yield (%) reduction reduction LRF
    Clarified Harvest 97.0% n/a n/a n/a
    Prt-A Eluate Pool 89.6% 0.06 1.87
    Viral Inactivated 99.7% No reduction 0.07 0.39
    Filtrate
    MM FT pool 91.9% 2.26 0.83 1.63
    HIC (B/E) Eluate 90.1% 0.40 0.22 1.41
    Nanofiltrate Filtrate 90.7% No reduction No reduction 0.15
    BDS (B/E) 102.0% No reduction No reduction 0.22
    HIC FT-pool 98.5% 0.16 0.23 0.46
    VF (FT) Filtrate 96.1% No reduction No reduction 0.10
    BDS (FT) 103.8% No reduction No reduction No
    reduction
  • 6.3.15. Example MM 15: Utility of AR Reduction
  • The current invention provides a method for reducing acidic species for a given protein of interest. In this example adalimumab was prepared using a combination of AEX and CEX technologies to produce a Low-AR and High-AR sample with a final AR of 2.5% and 6.9%, respectively. Both samples were incubated in a controlled environment at 25° C. and 65% relative humidity for 10 weeks, and the AR measured every two weeks. FIG. 23 shows the growth of AR for each sample over the 10 week incubation. It is evident from FIG. 23 the growth rate of AR is linear and similar between both the Low-AR and High-AR samples. Based on these results the reduced AR material can be stored 3 fold longer before reaching the same AR level as the High-AR sample. This is a significant utility as this can be very beneficial in storage handling and use of the antibody or other proteins for therapeutic use.
  • 6.4 Process Combinations to Achieve Target % AR or AR Reductions
  • Upstream and Downstream process technologies, e.g., cell culture and chromatographic separations, of the inventions disclosed in the following applications can be combined together or combined with methods in the art to provide a final target AR value or achieve a % AR reduction, as well as to, in certain embodiments, reduce product related substances and/or process related impurities. Upstream methods for AR reduction include, but are not limited to those described in the in the U.S. Ser. No. 13/830,583. Downstream methods for AR reduction include, but are not limited to, those described in the instant application. Exemplary technologies disclosed in the referenced applications include, but are not limited to: cell culture additives & conditions; clarified harvest additives and pH/salt conditions; mixed mode media separations; anion exchange media separations; and cation Exchange media separations.
  • The instant example demonstrates the combined effect of one or more of these technologies in achieving a target AR value or AR reduction, thereby facilitating the preparation of an antibody material having a specific charge heterogeneity. Additional examples of combinations of downstream technologies and upstream technologies are provided in the referenced applications.
  • In this example, the combination of upstream and downstream methods involves the reduction of acidic species in 3 L bioreactor cell cultures supplemented with arginine (2 g/l) and lysine (4 g/1) as has been previously demonstrated in the U.S. Ser. No. 13/830,583. The results of that strategy are summarized in Table 74. The total acidic species was reduced from 20.5% in the control sample to 10.2% in sample from cultures that were supplemented with the additives. In this study, Adalimumab producing cell line 1 was cultured in media 1 (chemically defined media) supplemented with amino acid arginine (2 g/1) and lysine (4 g/l) in a 300 L bioreactor. On Day 12 of culture, the culture was harvested and then subsequently analyzed using WCX-10 post protein A purification and the percentages of total peak(s) area corresponding to the acidic species were quantified. The percentage of acidic species was estimated to be 9.1% in the 300 L harvest sample.
  • TABLE 74
    AR levels achieved with use of upstream technologies
    3 L Bioreactor 300 L Bioreactor
    Arginine (2 g/l) + Arginine (2 g/l) +
    Control Lysine (4 g/l) Lysine (4 g/l)
    Total Total Total
    AR1 AR2 AR AR1 AR2 AR AR1 AR2 AR
    (%) (%) (%) (%) (%) (%) (%) (%) (%)
    6.3 14.2 20.5 2.6 7.6 10.2 2.4 6.7 9.1
  • The material produced by the 300 L Bioreactor employing Arginine and Lysine additions, that effectively reduced the AR levels to 9.1% was purified using a downstream process employing Mixed Mode chromatography as the primary AR Reduction method.
  • Adalimumab was purified by a Protein A chromatography step followed with a low pH viral inactivation step. The filtered viral inactivated material was buffer exchanged and loaded onto a Capto Adhere column. The flowthrough of Capto Adhere material was then purified with a HIC column with bind/elute mode as well as Flow Through mode. As shown in Table 75, AR reduction was achieved primarily with MM step, with some contribution from other steps. The table also shows that additional product related substances such as aggregates and process related impurities such as HCP can be effectively reduced employing these combined technologies.
  • TABLE 75
    Complete Downstream Process Train with Protein A Capture - AR,
    HMW and HCP reduction
    % AR % HMW HCP
    Process Yield (%) reduction reduction LRF
    Clarified Harvest 97.0% n/a n/a n/a
    Prt-A Eluate Pool 89.6% 0.06 1.87
    Viral Inactivated 99.7% No reduction 0.07 0.39
    Filtrate
    MM FT pool 91.9% 2.26 0.83 1.63
    HIC (B/E) Eluate 90.1% 0.40 0.22 1.41
    Nanofiltrate Filtrate 90.7% No reduction No reduction 0.15
    BDS (B/E) 102.0% No reduction No reduction 0.22
    HIC FT-pool 98.5% 0.16 0.23 0.46
    VF (FT) Filtrate 96.1% No reduction No reduction 0.10
    BDS (FT) 103.8% No reduction No reduction No
    reduction
  • As is evident from the above example, the MM method further reduced the AR levels, by 2.26%. Therefore upstream technologies for reduction can be combined with downstream technologies to achieve AR levels/AR reduction.
  • Patents, patent applications, publications, product descriptions, GenBank Accession Numbers, and protocols that may be cited throughout this application, the disclosures of which are incorporated herein by reference in their entireties for all purposes. For example, but not by way of limitation, patent applications designated by the following U.S. Application Serial numbers are incorporated herein by reference in their entireties for all purposes: Ser. No. 13/803,808; 13/830,583; 13/830,976; 13/831,181; and Ser. No. 13/804,220.

Claims (23)

1. A method of making a pharmaceutical composition, comprising mixing
(a) a composition comprising adalimumab, wherein the composition comprises less than 10% total acidic species of adalimumab, wherein the acidic species of adalimumab correspond to the peaks that elute earlier than the main peak in a WCX-10 HPLC chromatogram of adalimumab, wherein the WCX-10 HPLC chromatogram is generated using a first mobile phase of 10 mM Sodium Phosphate dibasic (pH 7.5) and a second mobile phase of 10 mM Sodium Phosphate dibasic, 500 mM Sodium Chloride (pH 5.5), and wherein the WCX-10 HPLC chromatogram is generated using detection at 280 nm; and
(b) a pharmaceutically acceptable carrier, thereby making a pharmaceutical composition.
2. The method of claim 1, wherein the acidic species of adalimumab comprise a first acidic region (AR1) and a second acidic region (AR2).
3. The method of claim 1, wherein the adalimumab is produced in a mammalian host cell grown in cell culture.
4. The method of claim 3, wherein the mammalian host cell is selected from the group consisting of a CHO cell, an NS0 cell, a COS cell, and an SP2 cell.
5. The method of claim 1, wherein the low acidic species composition comprises less than 3.8% total acidic species of adalimumab.
6. The method of claim 2, wherein the low acidic species composition comprises 0.8% AR1 and 3.0% AR2.
7. The method of claim 1, wherein the low acidic species composition comprises less than 2.4% total acidic species of adalimumab.
8. The method of claim 2, wherein the low acidic species composition comprises 0.2% AR1 and 2.2% AR2.
9. The method of claim 1, wherein the low acidic species composition comprises 4.7%-8.3% total acidic species of adalimumab.
10. The method of claim 1, wherein adalimumab is present in the pharmaceutical composition at a concentration of 0.1-250 mg/mL.
11. The method of claim 1, further comprising filling a syringe with the pharmaceutical composition.
12. The method of claim 1, wherein the pharmaceutically acceptable carrier comprises one or more excipient selected from the group consisting of a buffer, a surfactant and a polyalcohol, or a combination thereof.
13.-18. (canceled)
19. A method of making a pharmaceutical composition, comprising mixing
(a) a composition comprising adalimumab, wherein the composition comprises less than 10% total acidic species of adalimumab, wherein the acidic species of adalimumab are quantified based on the relative area percent of peaks that elute earlier than the main peak in a WCX-10 HPLC chromatogram of adalimumab wherein the WCX-10 HPLC chromatogram is generated using a first mobile phase of 10 mM Sodium Phosphate dibasic (pH 7.5) and a second mobile phase of 10 mM Sodium Phosphate dibasic, 500 mM Sodium Chloride (pH 5.5) and wherein the WCX-10 HPLC chromatogram is generated using detection at 280 nm; and
(b) a pharmaceutically acceptable carrier comprising a surfactant and a polyalcohol, thereby making a pharmaceutical composition; and
filling a syringe with the pharmaceutical composition.
20. The method of claim 19, wherein the acidic species of adalimumab comprise a first acidic region (AR1) and a second acidic region (AR2).
21. The method of claim 19, wherein the adalimumab is produced in a mammalian host cell grown in cell culture.
22. (canceled)
23. The method of claim 19, wherein the low acidic species composition comprises less than 3.8% total acidic species of adalimumab.
24. The method of claim 20, wherein the low acidic species composition comprises 0.8% AR1 and 3.0% AR2.
25. The method of claim 19, wherein the low acidic species composition comprises less than 2.4% total acidic species of adalimumab.
26. The method of claim 20, wherein the low acidic species composition comprises 0.2% AR1 and 2.2% AR2.
27. The method of claim 19, wherein the low acidic species composition comprises 4.7%-8.3% total acidic species of adalimumab.
28.-30. (canceled)
US15/951,560 2012-04-20 2018-04-12 Protein purification methods to reduce acidic species Abandoned US20190062419A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/951,560 US20190062419A1 (en) 2012-04-20 2018-04-12 Protein purification methods to reduce acidic species

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201261636493P 2012-04-20 2012-04-20
US201261636511P 2012-04-20 2012-04-20
US13/829,989 US9334319B2 (en) 2012-04-20 2013-03-14 Low acidic species compositions
US14/584,492 US9346879B2 (en) 2012-04-20 2014-12-29 Protein purification methods to reduce acidic species
US15/149,898 US9957318B2 (en) 2012-04-20 2016-05-09 Protein purification methods to reduce acidic species
US15/951,560 US20190062419A1 (en) 2012-04-20 2018-04-12 Protein purification methods to reduce acidic species

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/149,898 Continuation US9957318B2 (en) 2012-04-20 2016-05-09 Protein purification methods to reduce acidic species

Publications (1)

Publication Number Publication Date
US20190062419A1 true US20190062419A1 (en) 2019-02-28

Family

ID=48045072

Family Applications (8)

Application Number Title Priority Date Filing Date
US13/829,989 Active 2033-10-12 US9334319B2 (en) 2012-04-20 2013-03-14 Low acidic species compositions
US13/830,583 Active 2033-05-13 US9150645B2 (en) 2012-04-20 2013-03-14 Cell culture methods to reduce acidic species
US14/584,492 Active US9346879B2 (en) 2012-04-20 2014-12-29 Protein purification methods to reduce acidic species
US14/842,933 Active US9359434B2 (en) 2012-04-20 2015-09-02 Cell culture methods to reduce acidic species
US15/008,895 Active US9683033B2 (en) 2012-04-20 2016-01-28 Cell culture methods to reduce acidic species
US15/149,898 Active 2033-07-22 US9957318B2 (en) 2012-04-20 2016-05-09 Protein purification methods to reduce acidic species
US15/601,945 Abandoned US20170355761A1 (en) 2012-04-20 2017-05-22 Cell culture methods to reduce acidic species
US15/951,560 Abandoned US20190062419A1 (en) 2012-04-20 2018-04-12 Protein purification methods to reduce acidic species

Family Applications Before (7)

Application Number Title Priority Date Filing Date
US13/829,989 Active 2033-10-12 US9334319B2 (en) 2012-04-20 2013-03-14 Low acidic species compositions
US13/830,583 Active 2033-05-13 US9150645B2 (en) 2012-04-20 2013-03-14 Cell culture methods to reduce acidic species
US14/584,492 Active US9346879B2 (en) 2012-04-20 2014-12-29 Protein purification methods to reduce acidic species
US14/842,933 Active US9359434B2 (en) 2012-04-20 2015-09-02 Cell culture methods to reduce acidic species
US15/008,895 Active US9683033B2 (en) 2012-04-20 2016-01-28 Cell culture methods to reduce acidic species
US15/149,898 Active 2033-07-22 US9957318B2 (en) 2012-04-20 2016-05-09 Protein purification methods to reduce acidic species
US15/601,945 Abandoned US20170355761A1 (en) 2012-04-20 2017-05-22 Cell culture methods to reduce acidic species

Country Status (2)

Country Link
US (8) US9334319B2 (en)
WO (1) WO2013158279A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021220253A1 (en) * 2020-05-01 2021-11-04 Kashiv Biosciences, Llc An improved process of affinity chromatography
US11333642B2 (en) 2016-10-25 2022-05-17 Regeneran Pharmaceuticals, Inc. Methods and systems for chromatography data analysis
US11369896B2 (en) 2016-08-16 2022-06-28 Regeneron Pharmaceuticals, Inc. Methods for quantitating individual antibodies from a mixture
WO2023053031A1 (en) * 2021-09-28 2023-04-06 Kashiv Biosciences, Llc An improved process of purification of fusion protein
WO2023053030A1 (en) * 2021-09-28 2023-04-06 Kashiv Biosciences, Llc An improved process for purification of protein
US11884698B2 (en) 2018-07-02 2024-01-30 Regeneron Pharmaceuticals, Inc. Systems and methods for preparing a polypeptide from a mixture

Families Citing this family (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2911256A1 (en) * 2008-10-20 2010-12-09 Robert K. Hickman Isolation and purification of antibodies using protein a affinity chromatography
WO2012149197A2 (en) 2011-04-27 2012-11-01 Abbott Laboratories Methods for controlling the galactosylation profile of recombinantly-expressed proteins
WO2013009648A2 (en) * 2011-07-08 2013-01-17 Momenta Pharmaceuticals, Inc. Cell culture process
EP2636681A1 (en) * 2012-03-09 2013-09-11 CSL Behring AG Process for enriching IgA
EP2636684A1 (en) 2012-03-09 2013-09-11 CSL Behring AG Prevention of infection
CN104254542A (en) 2012-03-09 2014-12-31 瑞士杰特贝林生物制品有限公司 Compositions comprising secretory - like immunoglobulins
WO2013158273A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Methods to modulate c-terminal lysine variant distribution
US9334319B2 (en) 2012-04-20 2016-05-10 Abbvie Inc. Low acidic species compositions
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
WO2013176754A1 (en) 2012-05-24 2013-11-28 Abbvie Inc. Novel purification of antibodies using hydrophobic interaction chromatography
KR20150043523A (en) 2012-09-02 2015-04-22 애브비 인코포레이티드 Methods to control protein heterogeneity
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
SG11201505195TA (en) * 2013-02-06 2015-08-28 Agency Science Tech & Res Methods For Reducing Aggregate Content In Protein Preparations
SG11201507230PA (en) 2013-03-12 2015-10-29 Abbvie Inc Human antibodies that bind human tnf-alpha and methods of preparing the same
US10023608B1 (en) 2013-03-13 2018-07-17 Amgen Inc. Protein purification methods to remove impurities
WO2014159831A1 (en) * 2013-03-13 2014-10-02 Merck Sharp & Dohme Corp. Adapted lepidopteran insect cells for the production of recombinant proteins
WO2014151878A2 (en) 2013-03-14 2014-09-25 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosacharides
WO2014158231A1 (en) * 2013-03-14 2014-10-02 Abbvie Inc. Low acidic species compositions and methods for producing and using the same
WO2014159579A1 (en) 2013-03-14 2014-10-02 Abbvie Inc. MUTATED ANTI-TNFα ANTIBODIES AND METHODS OF THEIR USE
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
AR095196A1 (en) 2013-03-15 2015-09-30 Regeneron Pharma SERUM FREE CELL CULTIVATION MEDIA
EP2970378B1 (en) 2013-03-15 2021-05-26 Biogen MA Inc. Hydrophobic interaction protein chromatography under no-salt conditions
KR101569783B1 (en) * 2013-06-05 2015-11-19 한화케미칼 주식회사 A Method of Antibody Purification
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
AU2014337263B2 (en) 2013-10-16 2019-12-12 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
WO2015070068A1 (en) * 2013-11-07 2015-05-14 Abbvie Inc. Isolation and purification of antibodies
WO2015073884A2 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
EP3145951A1 (en) 2014-06-24 2017-03-29 InSight Biopharmaceuticals Ltd. Methods of purifying antibodies
US20160115225A1 (en) * 2014-10-24 2016-04-28 Abbvie Inc. Methods of Reducing Methylglyoxal (MGO) Modification of Recombinant Proteins in Cell Culture
CA2970732C (en) * 2014-12-15 2023-05-16 Merck Patent Gmbh Target molecule capture from crude solutions
EP3247718B1 (en) * 2015-01-21 2021-09-01 Outlook Therapeutics, Inc. Modulation of charge variants in a monoclonal antibody composition
GB2539420B (en) * 2015-06-16 2021-01-13 Cytiva Sweden Ab Determination of chromatography conditions
TW202340452A (en) 2015-08-04 2023-10-16 美商再生元醫藥公司 Taurine supplemented cell culture medium and methods of use
CN106496302B (en) * 2015-09-08 2021-12-10 三生国健药业(上海)股份有限公司 Method for purifying protein by ion exchange chromatography
CA3005484A1 (en) * 2015-11-18 2017-05-26 Merck Patent Gmbh Opposite ph-salt gradients for improved protein separations
JP2018538268A (en) * 2015-11-18 2018-12-27 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung Improved protein separation in ion exchange chromatography
EP3411401A1 (en) 2016-02-03 2018-12-12 Oncobiologics, Inc. Buffer formulations for enhanced antibody stability
MX2018011162A (en) * 2016-03-16 2018-11-22 Phoenix Tissue Repair Inc Methods of purifying collagen 7.
CA3045970A1 (en) * 2016-12-21 2018-06-28 F. Hoffmann-La Roche Ag Method for in vitro glycoengineering of antibodies
GB201703116D0 (en) * 2017-02-27 2017-04-12 Ge Healthcare Bioprocess R&D Ab A seperation matrix and a method of seperating antibodies
WO2018170488A1 (en) * 2017-03-17 2018-09-20 Gilead Sciences, Inc. Method of purifying an antibody
JP2020511516A (en) * 2017-03-24 2020-04-16 カウンスィル オブ サイエンティフィック アンド インダストリアル リサーチCouncil Of Scientific & Industrial Research Method for purifying recombinant antibody fragment
US20200199525A1 (en) 2017-06-08 2020-06-25 Polpharma Biologics S.A. Improved Methods of Cell Culture
EA202092335A1 (en) 2018-04-02 2021-01-22 Эмджен Инк. COMPOSITIONS BASED ON ERENUMAB AND WAYS OF THEIR APPLICATION
WO2019193139A1 (en) * 2018-04-06 2019-10-10 Aquaporin A/S Process for producing a membrane protein
US11079361B2 (en) * 2018-04-20 2021-08-03 Janssen Biotech, Inc. Chromatography column qualification in manufacturing methods for producing anti-IL12/IL23 antibody compositions
WO2020023566A1 (en) * 2018-07-25 2020-01-30 Merck Sharp & Dohme Corp. Methods of separating host cell lipases from a production protein in chromatographic processes
CN111153993A (en) * 2018-11-07 2020-05-15 正大天晴药业集团南京顺欣制药有限公司 Preparation method of anti-TNF- α monoclonal antibody
EP3921652A4 (en) * 2019-02-08 2022-11-02 Tanvex Biopharma Usa, Inc. Data extraction for biopharmaceutical analysis
CN111013656B (en) * 2019-12-27 2021-05-28 河南农业大学 Synthetic method of tetrazine hypercrosslinked porous photocatalyst
CN114929725A (en) * 2020-01-08 2022-08-19 信达生物制药(苏州)有限公司 Adalimumab purification process and stable compositions thereof
CN113563469A (en) * 2020-04-28 2021-10-29 江苏中新医药有限公司 Method for purifying adalimumab with high recovery rate
AU2021265591A1 (en) * 2020-05-01 2022-12-22 Kashiv Biosciences, Llc An improved process of purification of protein
WO2022154762A1 (en) * 2021-01-18 2022-07-21 Turgut İlaçlari A.Ş. Method of producing adalimumab
CN116761880A (en) * 2021-01-20 2023-09-15 瑞泽恩制药公司 Method for improving protein titer in cell culture
WO2023161885A1 (en) * 2022-02-25 2023-08-31 Kashiv Biosciences, Llc A process for improving polypeptide expression in mammalian cell culture

Family Cites Families (551)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3887430A (en) 1972-02-24 1975-06-03 Dow Chemical Co Culture medium for tissue culture techniques
USRE30985E (en) 1978-01-01 1982-06-29 Serum-free cell culture media
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US5179017A (en) 1980-02-25 1993-01-12 The Trustees Of Columbia University In The City Of New York Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4634665A (en) 1980-02-25 1987-01-06 The Trustees Of Columbia University In The City Of New York Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
DE3280462T2 (en) 1981-09-08 1995-04-20 Univ Rockefeller Antibodies to a composition with mediator activity and its use in a pharmaceutical composition.
US4510245A (en) 1982-11-18 1985-04-09 Chiron Corporation Adenovirus promoter system
US4560655A (en) 1982-12-16 1985-12-24 Immunex Corporation Serum-free cell culture medium and process for making same
US4657866A (en) 1982-12-21 1987-04-14 Sudhir Kumar Serum-free, synthetic, completely chemically defined tissue culture media
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4534972A (en) 1983-03-29 1985-08-13 Miles Laboratories, Inc. Protein compositions substantially free from infectious agents
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
DD266710A3 (en) 1983-06-06 1989-04-12 Ve Forschungszentrum Biotechnologie Process for the biotechnical production of alkaline phosphatase
US4767704A (en) 1983-10-07 1988-08-30 Columbia University In The City Of New York Protein-free culture medium
DE3572982D1 (en) 1984-03-06 1989-10-19 Takeda Chemical Industries Ltd Chemically modified lymphokine and production thereof
US4704366A (en) 1984-06-22 1987-11-03 Bio-Rad Laboratories, Inc. Process for binding IgG to protein A
US5672347A (en) 1984-07-05 1997-09-30 Genentech, Inc. Tumor necrosis factor antagonists and their use
DE3431140A1 (en) 1984-08-24 1986-03-06 Behringwerke Ag, 3550 Marburg ENHANCER FOR EUKARYOTIC EXPRESSION SYSTEMS
US4879231A (en) 1984-10-30 1989-11-07 Phillips Petroleum Company Transformation of yeasts of the genus pichia
IL73883A (en) 1984-12-20 1990-12-23 Yeda Res & Dev Monoclonal antibodies against tnf-alpha,hybridomas producing them and method for the purification of tnf-alpha
US5168062A (en) 1985-01-30 1992-12-01 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter-regulatory DNA sequence
GB8516415D0 (en) 1985-06-28 1985-07-31 Celltech Ltd Culture of animal cells
US5672502A (en) 1985-06-28 1997-09-30 Celltech Therapeutics Limited Animal cell culture
EP0212489B1 (en) 1985-08-16 1994-11-30 The Rockefeller University Anabolic activity modulator and uses thereof
IT206190Z2 (en) 1985-12-17 1987-07-13 Iveco Fiat IMPROVING THE BODYWORK OF A HEAVY VEHICLE
US4968615A (en) 1985-12-18 1990-11-06 Ciba-Geigy Corporation Deoxyribonucleic acid segment from a virus
US4925796A (en) 1986-03-07 1990-05-15 Massachusetts Institute Of Technology Method for enhancing glycoprotein stability
US5681718A (en) 1986-03-14 1997-10-28 Celltech Limited Methods for enhanced production of tissue plasminogen activator in cell culture using alkanoic acids or salts thereof
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US4927762A (en) 1986-04-01 1990-05-22 Cell Enterprises, Inc. Cell culture medium with antioxidant
GB8610600D0 (en) 1986-04-30 1986-06-04 Novo Industri As Transformation of trichoderma
DE3631229A1 (en) 1986-09-13 1988-03-24 Basf Ag MONOCLONAL ANTIBODIES AGAINST HUMAN TUMORNESCROSE FACTOR (TNF) AND THEIR USE
US4801687A (en) 1986-10-27 1989-01-31 Bioprobe International, Inc. Monoclonal antibody purification process using protein A
US5045468A (en) 1986-12-12 1991-09-03 Cell Enterprises, Inc. Protein-free culture medium which promotes hybridoma growth
US4877608A (en) 1987-11-09 1989-10-31 Rorer Pharmaceutical Corporation Pharmaceutical plasma protein formulations in low ionic strength media
US6238891B1 (en) 1987-11-18 2001-05-29 Cetus Oncology Corporation Method of increasing product expression through solute stress
US5118796A (en) 1987-12-09 1992-06-02 Centocor, Incorporated Efficient large-scale purification of immunoglobulins and derivatives
US5476996A (en) 1988-06-14 1995-12-19 Lidak Pharmaceuticals Human immune system in non-human animal
NZ229922A (en) 1988-07-18 1992-04-28 Chiron Corp Monoclonal antibodies specifically binding cachectin (tumor necrosis factor) and compositions
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
ATE135397T1 (en) 1988-09-23 1996-03-15 Cetus Oncology Corp CELL CULTIVATION MEDIUM FOR INCREASED CELL GROWTH, TO INCREASE THE LONGEVITY AND EXPRESSION OF THE PRODUCTS
US6048728A (en) 1988-09-23 2000-04-11 Chiron Corporation Cell culture medium for enhanced cell growth, culture longevity, and product expression
US5126250A (en) 1988-09-28 1992-06-30 Eli Lilly And Company Method for the reduction of heterogeneity of monoclonal antibodies
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
DE68914244T2 (en) 1988-10-24 1994-10-27 Otsuka Pharma Co Ltd Monoclonal antibody.
AU634186B2 (en) 1988-11-11 1993-02-18 Medical Research Council Single domain ligands, receptors comprising said ligands, methods for their production, and use of said ligands and receptors
SG43205A1 (en) 1988-12-19 1997-10-17 American Cyanamid Co A method for the treatment of endotoxic shock in a mammal
US5047335A (en) 1988-12-21 1991-09-10 The Regents Of The University Of Calif. Process for controlling intracellular glycosylation of proteins
EP0401384B1 (en) 1988-12-22 1996-03-13 Kirin-Amgen, Inc. Chemically modified granulocyte colony stimulating factor
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
DE4009603A1 (en) 1989-03-30 1990-10-04 Leybold Ag Lock chamber for substrate
US4933435A (en) 1989-04-05 1990-06-12 Bioprobe International Antibody purification process
US5169936A (en) 1989-04-14 1992-12-08 Biogen, Inc. Protein purification on immobilized metal affinity resins effected by elution using a weak ligand
EP0402226A1 (en) 1989-06-06 1990-12-12 Institut National De La Recherche Agronomique Transformation vectors for yeast yarrowia
US5959087A (en) 1989-08-07 1999-09-28 Peptide Technology, Ltd. Tumour necrosis factor binding ligands
DE10399036I1 (en) 1989-08-07 2004-04-01 Peptide Technology Ltd Binding ligand for tumor necrosis factor.
US6498237B2 (en) 1989-08-07 2002-12-24 Peptech Limited Tumor necrosis factor antibodies
GB8921123D0 (en) 1989-09-19 1989-11-08 Millar Ann B Treatment of ards
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5945098A (en) 1990-02-01 1999-08-31 Baxter International Inc. Stable intravenously-administrable immune globulin preparation
DE4037604A1 (en) 1990-04-25 1991-10-31 Bayer Ag Use of anti-TNF antibodies to treat ischaemia and its sequelae - esp. to increase survival rate after myocardial infarct and transplants
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
US5378612A (en) 1990-05-11 1995-01-03 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Culture medium for production of recombinant protein
US5110913A (en) 1990-05-25 1992-05-05 Miles Inc. Antibody purification method
US5096816A (en) 1990-06-05 1992-03-17 Cetus Corporation In vitro management of ammonia's effect on glycosylation of cell products through pH control
GB9014932D0 (en) 1990-07-05 1990-08-22 Celltech Ltd Recombinant dna product and method
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
WO1992020791A1 (en) 1990-07-10 1992-11-26 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US6255458B1 (en) 1990-08-29 2001-07-03 Genpharm International High affinity human antibodies and human antibodies against digoxin
ES2108048T3 (en) 1990-08-29 1997-12-16 Genpharm Int PRODUCTION AND USE OF LOWER TRANSGENIC ANIMALS CAPABLE OF PRODUCING HETEROLOGICAL ANTIBODIES.
US6300129B1 (en) 1990-08-29 2001-10-09 Genpharm International Transgenic non-human animals for producing heterologous antibodies
US7084260B1 (en) 1996-10-10 2006-08-01 Genpharm International, Inc. High affinity human antibodies and human antibodies against human antigens
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5122469A (en) 1990-10-03 1992-06-16 Genentech, Inc. Method for culturing Chinese hamster ovary cells to improve production of recombinant proteins
GB9022545D0 (en) 1990-10-17 1990-11-28 Wellcome Found Culture medium
GB9022543D0 (en) 1990-10-17 1990-11-28 Wellcome Found Antibody production
GB9022547D0 (en) 1990-10-17 1990-11-28 Wellcome Found Purified immunoglobulin
EP0564531B1 (en) 1990-12-03 1998-03-25 Genentech, Inc. Enrichment method for variant proteins with altered binding properties
GB2279077B (en) 1990-12-21 1995-06-14 Celltech Ltd Therapeutic compositions comprising recombinant antibodies specific for the TNFalpha
US5994510A (en) 1990-12-21 1999-11-30 Celltech Therapeutics Limited Recombinant antibodies specific for TNFα
GB9109645D0 (en) 1991-05-03 1991-06-26 Celltech Ltd Recombinant antibodies
GB9028123D0 (en) 1990-12-28 1991-02-13 Erba Carlo Spa Monoclonal antibodies against human tumor necrosis factor alpha
ES2330052T3 (en) 1991-03-01 2009-12-03 Dyax Corporation CHEMICAL PROTEIN THAT INCLUDES MICRO-PROTEINS THAT HAVE TWO OR MORE DISULFURENT BRIDGES AND RELATIONSHIPS OF THE SAME.
EP0575545B1 (en) 1991-03-15 2003-05-21 Amgen Inc. Pegylation of polypeptides
US7192584B2 (en) 1991-03-18 2007-03-20 Centocor, Inc. Methods of treating psoriasis with anti-TNF antibodies
US5656272A (en) 1991-03-18 1997-08-12 New York University Medical Center Methods of treating TNF-α-mediated Crohn's disease using chimeric anti-TNF antibodies
US20060246073A1 (en) 1991-03-18 2006-11-02 Knight David M Anti-TNF antibodies and peptides of human tumor necrosis factor
DE07006112T1 (en) 1991-03-18 2010-01-21 New York University Monoclonal and chimeric antibodies to human tumor necrosis factor
US5698195A (en) 1991-03-18 1997-12-16 New York University Medical Center Methods of treating rheumatoid arthritis using chimeric anti-TNF antibodies
US20070298040A1 (en) 1991-03-18 2007-12-27 Centocor, Inc. Methods of treating seronegative arthropathy with anti-TNF antibodies
US6277969B1 (en) 1991-03-18 2001-08-21 New York University Anti-TNF antibodies and peptides of human tumor necrosis factor
US20040120952A1 (en) 2000-08-07 2004-06-24 Centocor, Inc Anti-TNF antibodies and peptides of human tumor necrosis factor
DK0604418T3 (en) 1991-03-29 1999-06-14 Immunex Corp Isolated viral protein cytokine antagonists
EP0580737B1 (en) 1991-04-10 2004-06-16 The Scripps Research Institute Heterodimeric receptor libraries using phagemids
DE4122599C2 (en) 1991-07-08 1993-11-11 Deutsches Krebsforsch Phagemid for screening antibodies
US5328985A (en) 1991-07-12 1994-07-12 The Regents Of The University Of California Recombinant streptavidin-protein chimeras useful for conjugation of molecules in the immune system
EP0523949B1 (en) 1991-07-15 2003-06-25 The Wellcome Foundation Limited Production of antibodies
DK0605522T3 (en) 1991-09-23 2000-01-17 Medical Res Council Process for producing humanized antibodies
GB9122820D0 (en) 1991-10-28 1991-12-11 Wellcome Found Stabilised antibodies
WO1993011793A1 (en) 1991-12-17 1993-06-24 Schering Corporation Use of the combination of anti-tumor necrosis factor plus interleukin-6 to treat septic shock
JP3251587B2 (en) 1992-04-02 2002-01-28 スミスクライン・ビーチャム・コーポレイション Compounds useful for treating inflammatory diseases and inhibiting tumor necrosis factor production
CA2140638C (en) 1992-07-24 2010-05-04 Raju Kucherlapati Generation of xenogeneic antibodies
FI91535C (en) 1992-07-28 1994-07-11 Hurme Consulting Oy Method of denaturing road salt, and denaturing road salt
ES2121907T3 (en) 1992-08-28 1998-12-16 Bayer Ag USE OF ANTI-TNF MONOCLONAL ANTIBODIES FOR THE TREATMENT OF BACTERIAL MENINGITIS.
US6270766B1 (en) 1992-10-08 2001-08-07 The Kennedy Institute Of Rheumatology Anti-TNF antibodies and methotrexate in the treatment of arthritis and crohn's disease
AU5152293A (en) 1992-10-08 1994-05-09 Kennedy Institute Of Rheumatology, The Treatment of autoimmune and inflammatory disorders
JPH08509203A (en) 1992-10-15 1996-10-01 ダナ−ファーバー キャンサー インステテュート インコーポレイテッド Treatment of obesity-related diabetes type II insulin resistance with antagonists of TNF-α action
JPH06292592A (en) 1993-02-09 1994-10-21 Snow Brand Milk Prod Co Ltd Production of glycoprotein
PT614984E (en) 1993-03-05 2001-12-28 Bayer Ag ANTI-TNF HUMAN ANTIBODIES
DE4307508A1 (en) 1993-03-10 1994-09-15 Knoll Ag Use of anti-TNF antibodies as a medicine in the treatment of heart failure (heart muscle weakness)
CA2161351C (en) 1993-04-26 2010-12-21 Nils Lonberg Transgenic non-human animals capable of producing heterologous antibodies
CA2162689C (en) 1993-05-12 2000-07-18 Marc D. Better Immunotoxins comprising gelonin and an antibody
ES2138662T3 (en) 1993-06-03 2000-01-16 Therapeutic Antibodies Inc PRODUCTION OF ANTIBODY FRAGMENTS.
ATE405679T1 (en) 1993-10-19 2008-09-15 Scripps Research Inst SYNTHETIC HUMAN NEUTRALIZING MONOCLONAL ANTIBODIES AGAINST HIV
EP0659766A1 (en) 1993-11-23 1995-06-28 Schering-Plough Human monoclonal antibodies against human cytokines and methods of making and using such antibodies
NZ278607A (en) 1994-02-07 1999-05-28 Knoll Ag Use of tnf antagonists for treating disorders involving elevated serum levels of il-6 wherein the serum levels are 500pg/ml or above
EP0666312A1 (en) 1994-02-08 1995-08-09 Wolfgang A. Renner Process for the improvement of mammalian cell growth
US5429746A (en) 1994-02-22 1995-07-04 Smith Kline Beecham Corporation Antibody purification
JPH09509835A (en) 1994-03-04 1997-10-07 メルク エンド カンパニー インコーポレーテッド In vitro antibody affinity maturation by alanine scanning mutagenesis
US5856179A (en) 1994-03-10 1999-01-05 Genentech, Inc. Polypeptide production in animal cell culture
JPH07289288A (en) 1994-04-27 1995-11-07 L T T Kenkyusho:Kk Method for evaluating effect of antirheumatic medicine
US5945311A (en) 1994-06-03 1999-08-31 GSF--Forschungszentrumfur Umweltund Gesundheit Method for producing heterologous bi-specific antibodies
AU698393B2 (en) 1994-06-24 1998-10-29 Immunex Corporation Controlled release polypeptide compositions and methods of treating inflammatory bowel disease
ZA955642B (en) 1994-07-07 1997-05-06 Ortho Pharma Corp Lyophilized imaging agent formulation
US5561053A (en) 1994-08-05 1996-10-01 Genentech, Inc. Method for selecting high-expressing host cells
US5616487A (en) 1994-09-15 1997-04-01 Aastrom Biosciences, Inc. Stabilized retrovirus compositions
SE503424C2 (en) 1994-11-14 1996-06-10 Pharmacia Ab Process for purification of recombinant coagulation factor VIII
US5641870A (en) 1995-04-20 1997-06-24 Genentech, Inc. Low pH hydrophobic interaction chromatography for antibody purification
CA2219361C (en) 1995-04-27 2012-02-28 Abgenix, Inc. Human antibodies derived from immunized xenomice
EP0823941A4 (en) 1995-04-28 2001-09-19 Abgenix Inc Human antibodies derived from immunized xenomice
US5721121A (en) 1995-06-06 1998-02-24 Genentech, Inc. Mammalian cell culture process for producing a tumor necrosis factor receptor immunoglobulin chimeric protein
US6656466B1 (en) 1995-06-06 2003-12-02 Genetech, Inc. Human tumor necrosis factor—immunoglobulin(TNFR1-IgG1) chimera composition
US5705364A (en) 1995-06-06 1998-01-06 Genentech, Inc. Mammalian cell culture process
US6113898A (en) 1995-06-07 2000-09-05 Idec Pharmaceuticals Corporation Human B7.1-specific primatized antibodies and transfectomas expressing said antibodies
CA2226575C (en) 1995-07-27 2011-10-18 Genentech, Inc. Stabile isotonic lyophilized protein formulation
JP4306813B2 (en) 1995-09-19 2009-08-05 アスビオファーマ株式会社 New method for culturing animal cells
WO1997014719A1 (en) 1995-10-16 1997-04-24 Unilever N.V. A bifunctional or bivalent antibody fragment analogue
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
AR005035A1 (en) 1995-12-11 1999-04-07 Merck Patent Ges Mit Beschränkter Haftung PROCEDURE TO PREPARE RECOMBINANT PROTEINS IN E. COLI, BY FERMENTATION WITH GREAT CONCENTRATION OF CELLS.
SI9720020B (en) 1996-02-09 2001-12-31 Basf Ag Human antibodies that bind human TNF alpha
ES2180689T3 (en) 1996-04-19 2003-02-16 Nestle Sa IMMORTALIZED LINE OF HUMAN COLON EPITHELIAL CELLS.
GB9610992D0 (en) 1996-05-24 1996-07-31 Glaxo Group Ltd Concentrated antibody preparation
TW491855B (en) 1996-08-07 2002-06-21 Csl Ltd Purification of immunoglobulins
EP2221361A3 (en) 1996-08-30 2011-02-09 Life Technologies Corporation Method for producing a polypeptide in vitro in mammalian cells in a protein-free and serum-free culture medium
US20040171152A1 (en) 1996-10-10 2004-09-02 Invitrogen Corporation Animal cell culture media comprising non-animal or plant-derived nutrients
DK1864999T3 (en) 1996-11-27 2009-06-29 Genentech Inc Affinity purification of polypeptide on protein A matrix
EP1500329B1 (en) 1996-12-03 2012-03-21 Amgen Fremont Inc. Human antibodies that specifically bind human TNF alpha
GB9625175D0 (en) 1996-12-04 1997-01-22 Medi Cult As Serum-free cell culture media
FR2759585B1 (en) 1997-02-17 1999-06-11 Sanofi Sa PHARMACEUTICAL FORMULATIONS PRESENTED IN A DRY FORM FOR THE ORAL ADMINISTRATION OF A CYCLIC QUATERNARY AMMONIUM COMPOUND
US5804420A (en) 1997-04-18 1998-09-08 Bayer Corporation Preparation of recombinant Factor VIII in a protein free medium
HU224826B1 (en) 1997-04-28 2006-02-28 Lilly Co Eli Activated protein c formulations
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
DE69810481T2 (en) 1997-06-13 2003-09-25 Genentech Inc STABILIZED ANTIBODY FORMULATION
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
US6475725B1 (en) 1997-06-20 2002-11-05 Baxter Aktiengesellschaft Recombinant cell clones having increased stability and methods of making and using the same
US20040191256A1 (en) 1997-06-24 2004-09-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
CA2293829C (en) 1997-06-24 2011-06-14 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
US20020045207A1 (en) 1997-10-31 2002-04-18 Lynne A. Krummen Glycoprotein production process
AU759779B2 (en) 1997-10-31 2003-05-01 Genentech Inc. Methods and compositions comprising glycoprotein glycoforms
US20040136986A1 (en) 1997-10-31 2004-07-15 Genentech, Inc. Methods and compositions comprising glycoprotein glycoforms
US6673575B1 (en) 1997-12-03 2004-01-06 Roche Diagnostics Gmbh Method for preparing polypeptides with appropriate glycosilation
WO1999032605A1 (en) 1997-12-19 1999-07-01 Novo Nordisk A/S Method for producing heterologous proteins in eukaryotic cells on an industrial scale using nucleotide-manipulating agents
PT1071700E (en) 1998-04-20 2010-04-23 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
WO1999057246A1 (en) 1998-05-01 1999-11-11 Life Technologies, Inc. Animal cell culture media comprising non-animal or plant-derived nutrients
DE69936946T2 (en) 1998-05-06 2008-05-15 Genentech, Inc., South San Francisco Purification of antibodies by ion exchange chromatography
DK1308455T3 (en) 1998-05-06 2006-07-31 Genentech Inc Composition comprising anti-HER2 antibodies
DE19821933C1 (en) 1998-05-15 1999-11-11 Disetronic Licensing Ag Device for administering an injectable product
DE19822031C2 (en) 1998-05-15 2000-03-23 Disetronic Licensing Ag Auto injection device
US6528286B1 (en) 1998-05-29 2003-03-04 Genentech, Inc. Mammalian cell culture process for producing glycoproteins
US6406909B1 (en) 1998-07-10 2002-06-18 Chugai Seiyaku Kabushiki Kaisha Serum-free medium for culturing animal cells
US6914128B1 (en) 1999-03-25 2005-07-05 Abbott Gmbh & Co. Kg Human antibodies that bind human IL-12 and methods for producing
US7883704B2 (en) 1999-03-25 2011-02-08 Abbott Gmbh & Co. Kg Methods for inhibiting the activity of the P40 subunit of human IL-12
AU3633000A (en) 1999-03-26 2000-10-16 Human Genome Sciences, Inc. Neutrokine-alpha binding proteins and methods based thereon
PT1914244E (en) 1999-04-09 2013-07-26 Kyowa Hakko Kirin Co Ltd Method of modulating the activity of functional immune molecules
US7297680B2 (en) 1999-04-15 2007-11-20 Crucell Holland B.V. Compositions of erythropoietin isoforms comprising Lewis-X structures and high sialic acid content
EP1171615B1 (en) 1999-04-26 2006-12-13 Genentech, Inc. Cell culture process for glycoproteins
AU4314900A (en) 1999-04-28 2000-11-17 Yamanouchi Pharmaceutical Co., Ltd. Parenteral medicinal composition containing humanized monoclonal antibody fragment and method for stabilizing the same
AT409379B (en) 1999-06-02 2002-07-25 Baxter Ag MEDIUM FOR PROTEIN- AND SERUM-FREE CELL CULTURE
DE60028210T2 (en) 1999-07-30 2007-03-08 Genentech, Inc., South San Francisco LOADED FILTRATION MEMBRANES AND ITS USES
EP1220934B1 (en) 1999-09-27 2005-12-21 Genentech, Inc. Methods for making recombinant proteins using apoptosis inhibitors
JP2001120262A (en) 1999-10-26 2001-05-08 Welfide Corp Method for enhancing production of physiologically active substance
AR026743A1 (en) 1999-12-09 2003-02-26 Pharmacia Ab PRODUCTION OF PEPTIDES
KR20010056451A (en) 1999-12-15 2001-07-04 윤재승 Arginine-enriched medium used for mass-producing recombinant protein in animal cell culture
US20030124119A1 (en) 1999-12-28 2003-07-03 Tadao Yamazaki Stable antibody compositions and injection preparations
ES2266159T3 (en) 2000-02-08 2007-03-01 Genentech, Inc. IMPROVEMENT OF GALACTOSILATION OF RECOMBINANT GLICOPROTEINS.
NZ520392A (en) 2000-02-10 2005-04-29 Abbott Lab Antibodies that bind human interleukin-18 and methods of making and using
GB0003231D0 (en) 2000-02-11 2000-04-05 Medi Cult As Cell culture media
JP2001218840A (en) 2000-02-14 2001-08-14 Kanegafuchi Chem Ind Co Ltd Adsorbent for transforming growth factor. beta, adsorbing/removing method, and adsorber
WO2001077362A1 (en) 2000-04-06 2001-10-18 Chugai Seiyaku Kabushiki Kaisha Immunoassay of anti-hm1.24 antibody
CA2405709A1 (en) 2000-04-12 2001-10-25 Human Genome Sciences, Inc. Albumin fusion proteins
US7598055B2 (en) 2000-06-28 2009-10-06 Glycofi, Inc. N-acetylglucosaminyltransferase III expression in lower eukaryotes
DE60139720D1 (en) 2000-06-28 2009-10-08 Glycofi Inc Process for the preparation of modified glycoproteins
US7449308B2 (en) 2000-06-28 2008-11-11 Glycofi, Inc. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
WO2002002793A1 (en) 2000-07-05 2002-01-10 Japan As Represented By Secretary Of Osaka University Process for producing glycoprotein
WO2002011753A1 (en) 2000-08-04 2002-02-14 Chugai Seiyaku Kabushiki Kaisha Protein injection preparations
US7288390B2 (en) 2000-08-07 2007-10-30 Centocor, Inc. Anti-dual integrin antibodies, compositions, methods and uses
US20050249735A1 (en) 2000-08-07 2005-11-10 Centocor, Inc. Methods of treating ankylosing spondylitis using anti-TNF antibodies and peptides of human tumor necrosis factor
US20060018907A1 (en) 2000-08-07 2006-01-26 Centocor, Inc. Anti-TNF antibodies and peptides of human tumor necrosis factor
UA81743C2 (en) 2000-08-07 2008-02-11 Центокор, Инк. HUMAN MONOCLONAL ANTIBODY WHICH SPECIFICALLY BINDS TUMOR NECROSIS FACTOR ALFA (TNFα), PHARMACEUTICAL MIXTURE CONTAINING THEREOF, AND METHOD FOR TREATING ARTHRITIS
WO2002016590A2 (en) 2000-08-21 2002-02-28 Clonex Development, Inc. Methods and compositions for increasing protein yield from a cell culture
US6903069B2 (en) 2000-10-02 2005-06-07 Novo Nordisk Health Care A/S Factor VII glycoforms
US20090151023A1 (en) 2000-11-13 2009-06-11 Viktor Kuvshinov Transformation system for Camelina sativa
EP1360314B1 (en) 2001-02-15 2009-01-14 Centocor, Inc. Chemically defined medium for cultured mammalian cells
US20030096414A1 (en) 2001-03-27 2003-05-22 Invitrogen Corporation Culture medium for cell growth and transfection
JP2005509403A (en) 2001-03-27 2005-04-14 スミスクライン・ビーチャム・コーポレイション Control of glycoforms in IgG
CA2508763C (en) 2001-05-11 2012-01-24 Kirin Beer Kabushiki Kaisha Human antibody producing mouse and method for producing human antibody using the same
US7189820B2 (en) 2001-05-24 2007-03-13 Human Genome Sciences, Inc. Antibodies against tumor necrosis factor delta (APRIL)
US20030012786A1 (en) 2001-05-25 2003-01-16 Teoh Leah S. Use of anti-TNF antibodies as drugs in treating septic disorders of anemic patients
CA2868614A1 (en) 2001-06-08 2002-12-08 Abbott Laboratories (Bermuda) Ltd. Methods of administering anti-tnf.alpha. antibodies
US20030087372A1 (en) 2001-06-13 2003-05-08 Genentech, Inc. Methods of culturing animal cells and polypeptide production in animal cells
CA2451955C (en) 2001-06-26 2015-09-29 Abgenix, Inc. Antibodies to opgl
CN1555411A (en) 2001-08-03 2004-12-15 ���迨�����\���ɷݹ�˾ Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
GB0119520D0 (en) 2001-08-10 2001-10-03 Owen Mumford Ltd Improvements relating to injection devices
CN1309825C (en) 2001-10-02 2007-04-11 诺和诺德医疗保健公司 Method for production of recombinant proteins in eukaryote cells
US7125843B2 (en) 2001-10-19 2006-10-24 Neose Technologies, Inc. Glycoconjugates including more than one peptide
EP1443961B1 (en) 2001-10-25 2009-05-06 Genentech, Inc. Glycoprotein compositions
US7456276B2 (en) * 2001-11-12 2008-11-25 Novo Nordisk A/S Peptide purification by means of metal ion affinity chromatography
US7473680B2 (en) 2001-11-28 2009-01-06 Neose Technologies, Inc. Remodeling and glycoconjugation of peptides
MXPA04005191A (en) 2001-11-28 2005-04-29 Sandoz Ag Method for producing a recombinant polypeptide.
WO2003046162A2 (en) 2001-11-28 2003-06-05 Polymun Scientific Immunbiologische Forschung Gmbh Process for the production of polypeptides in mammalian cell cultures
WO2003045995A2 (en) 2001-11-28 2003-06-05 Sandoz Gmbh Cell culture process
CA2469815C (en) 2001-12-21 2011-05-03 Immunex Corporation Methods for purifying protein
ES2319636T3 (en) 2002-02-05 2009-05-11 Genentech, Inc. PURIFICATION OF PROTEINS.
US20030161828A1 (en) 2002-02-19 2003-08-28 Abbott Gmbh & Co. Kg Use of TNF antagonists as drugs for the treatment of patients with an inflammatory reaction and without suffering from total organ failure
CA2417689C (en) 2002-03-05 2006-05-09 F. Hoffmann-La Roche Ag Improved methods for growing mammalian cells in vitro
PL376821A1 (en) 2002-03-27 2006-01-09 Immunex Corporation Methods for increasing polypeptide production
US20030190710A1 (en) 2002-03-28 2003-10-09 Devries Ruth L. Control of glycoforms in IgG
EP1498490A4 (en) 2002-04-09 2006-11-29 Kyowa Hakko Kogyo Kk Process for producing antibody composition
US20040009172A1 (en) 2002-04-26 2004-01-15 Steven Fischkoff Use of anti-TNFalpha antibodies and another drug
US20030206898A1 (en) 2002-04-26 2003-11-06 Steven Fischkoff Use of anti-TNFalpha antibodies and another drug
US7323553B2 (en) 2002-04-26 2008-01-29 Genentech, Inc. Non-affinity purification of proteins
US20040029229A1 (en) 2002-05-20 2004-02-12 Reeves Philip J. High level protein expression system
AU2003249055B2 (en) 2002-07-15 2008-06-26 Immunex Corporation Methods and media for controlling sialylation of proteins produced by mammalian cells
NZ555692A (en) 2002-07-19 2009-02-28 Abbott Biotech Ltd Treatment of TNF alpha related disorders
US20090280065A1 (en) 2006-04-10 2009-11-12 Willian Mary K Uses and Compositions for Treatment of Psoriasis
US20040033228A1 (en) 2002-08-16 2004-02-19 Hans-Juergen Krause Formulation of human antibodies for treating TNF-alpha associated disorders
US7067279B1 (en) 2002-08-23 2006-06-27 Immunex Corporation Cell culture performance with betaine
US6924124B1 (en) 2002-08-23 2005-08-02 Immunex Corporation Feeding strategies for cell culture
US6974681B1 (en) 2002-08-23 2005-12-13 Immunex Corporation Cell culture performance with vanadate
WO2004026891A2 (en) 2002-09-18 2004-04-01 Genencor International, Inc. Protein purification
US6890736B1 (en) 2002-09-20 2005-05-10 Immunex Corporation Methods for producing proteins in cultured cells
MY150740A (en) 2002-10-24 2014-02-28 Abbvie Biotechnology Ltd Low dose methods for treating disorders in which tnf? activity is detrimental
CA2504134C (en) 2002-11-01 2012-03-06 Bayer Healthcare Llc Process for concentration of macromolecules
US20040086532A1 (en) 2002-11-05 2004-05-06 Allergan, Inc., Botulinum toxin formulations for oral administration
US20040162414A1 (en) 2002-11-22 2004-08-19 Santora Ling C. Method for reducing or preventing modification of a polypeptide in solution
US20040101939A1 (en) 2002-11-22 2004-05-27 Santora Ling C. Method for reducing or preventing modification of a polypeptide in solution
US20040191243A1 (en) 2002-12-13 2004-09-30 Bei Chen System and method for stabilizing antibodies with histidine
ES2354610T5 (en) 2002-12-23 2020-09-14 Bristol Myers Squibb Co Improved product quality in mammalian cell culture procedures for protein production
WO2004058800A2 (en) 2002-12-23 2004-07-15 Bristol-Myers Squibb Company Mammalian cell culture processes for protein production
DK1601697T3 (en) 2003-02-28 2007-10-01 Lonza Biologics Plc Purification of antibody by protein A and ion exchange chromatography
KR20060015568A (en) 2003-05-01 2006-02-17 디에스엠 아이피 어셋츠 비.브이. Process for the production of biological substances by perfusion culturing of suspended animal cells
ES2344789T5 (en) 2003-05-15 2017-06-14 Wyeth Llc Restricted glucose feeding for animal cell culture
GB2404665B (en) 2003-08-08 2005-07-06 Cambridge Antibody Tech Cell culture
EP1673452B1 (en) 2003-10-10 2015-12-23 Novo Nordisk Health Care AG Method for large-scale production of a polypeptide in eukaryote cells
FR2861080B1 (en) 2003-10-20 2006-02-17 Lab Francais Du Fractionnement ANTIBODIES HAVING AN OPTIMIZED FUCOSE AND GALACTOSE RATE
CA2542951C (en) 2003-10-24 2016-08-30 Abhinav A. Shukla Process for purifying proteins in a hydrophobic interaction chromatography flow-through fraction
US20050100965A1 (en) 2003-11-12 2005-05-12 Tariq Ghayur IL-18 binding proteins
DE10355251A1 (en) 2003-11-26 2005-06-23 Merck Patent Gmbh Water-based pharmaceutical preparation for treatment of tumors has active ingredient effective against receptor of endothelial growth factor receptor
CA2894300A1 (en) 2003-12-08 2005-06-23 The Government Of The United States Of America, As Represented By The Secreatary, Department Of Health And Human Services Monoclonal antibodies that bind or neutralize dengue virus
SG10201900535UA (en) 2003-12-23 2019-02-27 Genentech Inc Novel anti-il 13 antibodies and uses thereof
EP1697414A2 (en) 2003-12-23 2006-09-06 Applied Research Systems ARS Holding N.V. Process for the production of tumor necrosis factor-binding proteins
US20080058507A1 (en) 2004-02-11 2008-03-06 Hui Liu Method For The Removal Of Aggregate Proteins From Recombinant Samples Using Ion Exchange Chromatography
JP2007522157A (en) 2004-02-12 2007-08-09 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング Highly concentrated liquid formulation of anti-EGFR antibody
US7750129B2 (en) 2004-02-27 2010-07-06 Ge Healthcare Bio-Sciences Ab Process for the purification of antibodies
CN1234725C (en) 2004-04-07 2006-01-04 陈志南 High performance quick purifying method for preparing piecewise antibody
TW201705980A (en) 2004-04-09 2017-02-16 艾伯維生物技術有限責任公司 Multiple-variable dose regimen for treating TNF[alpha]-related disorders
US20060127950A1 (en) 2004-04-15 2006-06-15 Massachusetts Institute Of Technology Methods and products related to the improved analysis of carbohydrates
US20060057638A1 (en) 2004-04-15 2006-03-16 Massachusetts Institute Of Technology Methods and products related to the improved analysis of carbohydrates
CA2562772A1 (en) 2004-04-15 2005-10-27 Glycofi, Inc. Production of galactosylated glycoproteins in lower eukaryotes
ATE455861T1 (en) 2004-05-04 2010-02-15 Novo Nordisk Healthcare Ag O-LINKED GLYCOFORMS OF FACTOR VII AND METHOD FOR THE PRODUCTION THEREOF
CA2573842A1 (en) 2004-07-21 2006-02-09 Glycofi, Inc. Immunoglobulins comprising predominantly a gal2glcnac2man3glcnac2 glycoform
US20060223147A1 (en) 2004-08-05 2006-10-05 Kyowa Hakko Kogyo Co., Ltd., Process for producing glycoprotein composition
TWI384069B (en) 2004-08-27 2013-02-01 Pfizer Ireland Pharmaceuticals Production of polypeptides
HUE032538T2 (en) 2004-09-30 2017-10-30 Bayer Healthcare Llc Devices and Methods for Integrated Continuous Manufacturing of Biological Molecules
EP1807111A4 (en) 2004-10-08 2009-05-27 Abbott Biotech Ltd Respiratory syncytial virus (rsv) infection
WO2007011390A2 (en) 2004-10-09 2007-01-25 Government Of The United States As Represented By The Secretary Of The Army Large-scale production of human serum butyrylcholinesterase as a bioscavenger
KR101243601B1 (en) 2004-10-21 2013-03-20 지이 헬스케어 바이오-사이언시스 에이비 Chromatography ligand
US20060094104A1 (en) 2004-10-29 2006-05-04 Leopold Grillberger Animal protein-free media for cultivation of cells
JP2008529495A (en) 2005-02-04 2008-08-07 グラクソ グループ リミテッド Optimization of heterologous polypeptide expression
DE602006016735D1 (en) 2005-02-11 2010-10-21 Agency Science Tech & Res PROCESS FOR PROLIFERATION OF STEM CELLS
WO2006084826A1 (en) 2005-02-11 2006-08-17 Novo Nordisk Health Care Ag Production of a polypeptide in a serum-free cell culture liquid containing plant protein hydrolysate
PL1869065T3 (en) 2005-03-11 2020-09-21 Wyeth Llc A method of weak partitioning chromatography
US20060252672A1 (en) 2005-04-05 2006-11-09 Betenbaugh Michael J Protein N-glycosylation of eukaryotic cells using dolichol-linked oligosaccharide synthesis pathway, other N-gylosylation-increasing methods, and engineered hosts expressing products with increased N-glycosylation
US8129508B2 (en) * 2005-04-11 2012-03-06 Medarex, Inc. Protein purification using HCIC and ion exchange chromatography
US9963510B2 (en) 2005-04-15 2018-05-08 Macrogenics, Inc. Covalent diabodies and uses thereof
WO2006113743A2 (en) 2005-04-18 2006-10-26 Massachusetts Institute Of Technology Compositions and methods for rna interference with sialidase expression and uses thereof
CA2606270A1 (en) 2005-04-19 2006-10-26 Massachusetts Institute Of Technology Amphiphilic polymers and methods of use thereof
CN101500607B (en) 2005-05-16 2013-11-27 阿布维生物技术有限公司 Use of TNFalpha inhibitor for treatment of erosive polyarthritis
EP1888638A2 (en) 2005-06-03 2008-02-20 Genentech, Inc. Method of producing antibodies with modified fucosylation level
JP2008541746A (en) 2005-06-03 2008-11-27 ビオヴィトルム・アクチボラゲット(プブリクト) New process
PT1896071E (en) 2005-06-30 2015-07-09 Janssen Biotech Inc Methods and compositions with enhanced therapeutic activity
KR101419729B1 (en) 2005-07-26 2014-07-17 상가모 바이오사이언스 인코포레이티드 Targeted integration and expression of exogenous nucleic acid sequences
US20070041905A1 (en) 2005-08-19 2007-02-22 Hoffman Rebecca S Method of treating depression using a TNF-alpha antibody
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US20070041979A1 (en) 2005-08-19 2007-02-22 Raju T S Proteolysis resistant antibody preparations
EP1917276B1 (en) 2005-08-26 2018-03-21 Ares Trading S.A. Process for the preparation of glycosylated interferon beta
EP1942935A4 (en) 2005-09-02 2009-12-23 Glycofi Inc Immunoglobulins comprising predominantly a glcnacman3glcnac2 glycoform
PE20070796A1 (en) 2005-10-24 2007-08-15 Wyeth Corp PROTEIN PRODUCTION METHOD USING ANTI-SENESCENCE COMPOUNDS
TW201337266A (en) 2005-11-01 2013-09-16 Abbott Biotech Ltd Methods and compositions for diagnosing ankylosing spondylitis using biomarkers
US20080206246A1 (en) 2006-04-05 2008-08-28 Ravetch Jeffrey V Polypeptides with enhanced anti-inflammatory and decreased cytotoxic properties and relating methods
WO2007055916A2 (en) 2005-11-07 2007-05-18 The Rockefeller University Reagents, methods and systems for selecting a cytotoxic antibody or variant thereof
US8470318B2 (en) 2005-11-07 2013-06-25 The Rockefeller University Polypeptides with enhanced anti-inflammatory and decreased cytotoxic properties and relating methods
WO2008057634A2 (en) 2006-10-26 2008-05-15 The Rockefeller University Polypeptides with enhanced anti-inflammatory and decreased cytotoxic properties and relating methods
CA2630811C (en) 2005-12-08 2016-05-31 Amgen Inc. Improved production of glycoproteins using manganese
LT1974014T (en) 2006-01-04 2017-11-27 Baxalta Incorporated Oligopeptide-free cell culture media
US20070190057A1 (en) 2006-01-23 2007-08-16 Jian Wu Methods for modulating mannose content of recombinant proteins
US20070202051A1 (en) 2006-02-10 2007-08-30 Pari Gmbh Aerosols for sinunasal drug delivery
EP3456351A1 (en) 2006-04-05 2019-03-20 The Rockefeller University Polypeptides with enhanced anti-inflammatory and decreased cytotoxic properties and relating methods
SG170837A1 (en) 2006-04-05 2011-05-30 Abbott Biotech Ltd Antibody purification
US9605064B2 (en) 2006-04-10 2017-03-28 Abbvie Biotechnology Ltd Methods and compositions for treatment of skin disorders
US20090317399A1 (en) 2006-04-10 2009-12-24 Pollack Paul F Uses and compositions for treatment of CROHN'S disease
US20080118496A1 (en) 2006-04-10 2008-05-22 Medich John R Uses and compositions for treatment of juvenile rheumatoid arthritis
US9399061B2 (en) 2006-04-10 2016-07-26 Abbvie Biotechnology Ltd Methods for determining efficacy of TNF-α inhibitors for treatment of rheumatoid arthritis
US9624295B2 (en) 2006-04-10 2017-04-18 Abbvie Biotechnology Ltd. Uses and compositions for treatment of psoriatic arthritis
WO2007120626A2 (en) 2006-04-10 2007-10-25 Abbott Biotechnology Ltd. Uses and compositions for treatment of ankylosing spondylitis
US7846724B2 (en) 2006-04-11 2010-12-07 Hoffmann-La Roche Inc. Method for selecting CHO cell for production of glycosylated antibodies
US20080131374A1 (en) 2006-04-19 2008-06-05 Medich John R Uses and compositions for treatment of rheumatoid arthritis
US7851438B2 (en) 2006-05-19 2010-12-14 GlycoFi, Incorporated Erythropoietin compositions
US20080311043A1 (en) 2006-06-08 2008-12-18 Hoffman Rebecca S Uses and compositions for treatment of psoriatic arthritis
US20100021451A1 (en) 2006-06-08 2010-01-28 Wong Robert L Uses and compositions for treatment of ankylosing spondylitis
US20100234577A1 (en) 2006-06-14 2010-09-16 Smithkline Beecham Corporation Methods for purifying antibodies using ceramic hydroxyapatite
TWI527603B (en) 2006-06-30 2016-04-01 艾伯維生物技術有限責任公司 Automatic injection device
CN101541950A (en) 2006-07-13 2009-09-23 惠氏公司 Production of glycoproteins
WO2008028974A1 (en) 2006-09-08 2008-03-13 Novo Nordisk A/S Methods of optimizing chromatographic separation of polypeptides
ES2620261T3 (en) 2006-09-10 2017-06-28 Glycotope Gmbh Use of human myeloid leukemic cells for antibody expression
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
EP2500415A1 (en) 2006-09-13 2012-09-19 Abbott Laboratories Cell culture improvements
AU2007294731B2 (en) 2006-09-13 2014-04-17 Abbvie Inc. Cell culture improvements
US10982250B2 (en) 2006-09-18 2021-04-20 Genentech, Inc. Methods of protein production
US20080112953A1 (en) 2006-10-06 2008-05-15 Amgen Inc. Stable formulations
MX2009004351A (en) 2006-10-27 2009-05-12 Abbott Biotech Ltd Crystalline anti-htnfalpha antibodies.
MX2009004519A (en) 2006-11-03 2009-05-12 Wyeth Corp Glycolysis-inhibiting substances in cell culture.
EP2089424B1 (en) 2006-12-06 2015-07-01 JCR PHARMACEUTICALS Co., LTD. Method for production of human erythropoietin
JP2010512765A (en) 2006-12-22 2010-04-30 エフ.ホフマン−ラ ロシュ アーゲー Selection method
US20080226635A1 (en) 2006-12-22 2008-09-18 Hans Koll Antibodies against insulin-like growth factor I receptor and uses thereof
CN105056231A (en) 2006-12-28 2015-11-18 詹森生物科技公司 Methods and vectors for generating asialylated immunoglobulins
US7691980B2 (en) 2007-01-09 2010-04-06 Bio-Rad Laboratories, Inc. Enhanced capacity and purification of antibodies by mixed mode chromatography in the presence of aqueous-soluble nonionic organic polymers
EP2114984A2 (en) 2007-01-17 2009-11-11 Merck Serono S.A. Process for the purification of fc-containing proteins
DK2115126T3 (en) 2007-03-02 2015-05-04 Wyeth Llc Use of copper and glutamate in cell culture for the preparation of polypeptides
JP5634710B2 (en) 2007-03-19 2014-12-03 国立大学法人 岡山大学 Medium for protein production and virus propagation
NZ580379A (en) 2007-03-29 2012-10-26 Abbott Lab Crystalline anti-human il-12 antibodies
EP2068923A4 (en) 2007-03-30 2010-11-24 Medimmune Llc Antibodies with decreased deamidation profiles
EP2134853B1 (en) 2007-04-03 2018-07-18 Oxyrane UK Limited Glycosylation of molecules
EP2135094B1 (en) 2007-04-16 2017-03-08 Momenta Pharmaceuticals, Inc. Characterization of n-glycans using exoglycosidases
EP2135090A1 (en) 2007-04-16 2009-12-23 Momenta Pharmaceuticals, Inc. Proteolytic release of glycans
WO2008128216A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Methods for labeling glycans
WO2008128225A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Multi-dimensional chromatographic methods for separating n-glycans
RU2009141965A (en) 2007-04-16 2011-05-27 Момента Фармасьютикалз, Инк. (Us) CERTAIN Glycoprotein Products and Methods of Their Production
US20100279306A1 (en) 2007-04-16 2010-11-04 Momenta Pharmaceuticals, Inc. Analysis of phosphorylated glycans, glycopeptides or glycoproteins by imac
EP2135089B1 (en) 2007-04-16 2015-09-02 Momenta Pharmaceuticals, Inc. Comparative analysis of protein conformations by using 2d noesy nmr spectra
CA2682746A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Characterization of n-glycan mixtures by nuclear magnetic resonance
JP5175336B2 (en) 2007-04-16 2013-04-03 モメンタ ファーマシューティカルズ インコーポレイテッド Methods related to cell surface glycosylation
WO2008128230A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Reference glycoprotein products and related methods
US20110213137A1 (en) 2007-04-16 2011-09-01 Momenta Pharmaceuticals, Inc. Isotopically-labeled glycans
PT2135091E (en) 2007-04-16 2011-09-01 Momenta Pharmaceuticals Inc Ms methods to evaluate glycans
TW200902708A (en) 2007-04-23 2009-01-16 Wyeth Corp Methods of protein production using anti-senescence compounds
EP1988101A1 (en) 2007-05-04 2008-11-05 Novo Nordisk A/S Improvement of factor VIII polypeptide titers in cell cultures
WO2008135498A2 (en) 2007-05-04 2008-11-13 Novo Nordisk A/S Prevention of protein degradation in mammalian cell cultures
US8053236B2 (en) 2007-05-11 2011-11-08 Amgen Inc. Feed media
EP2165194A4 (en) 2007-05-31 2010-09-08 Abbott Lab BIOMARKERS PREDICTIVE OF THE RESPONSIVENESS TO TNF-alpha INHIBITORS IN AUTOIMMUNE DISORDERS
EP2152318A4 (en) 2007-06-01 2011-12-07 Abbott Biotech Ltd Uses and compositions for treatment of psoriasis and crohn's disease
EP2171451A4 (en) 2007-06-11 2011-12-07 Abbott Biotech Ltd Methods for treating juvenile idiopathic arthritis
WO2008154014A2 (en) 2007-06-11 2008-12-18 Amgen Inc. A method for culturing mammalian cells to improve recombinant protein production
US20090053786A1 (en) 2007-07-09 2009-02-26 Yung-Hsiang Kao Prevention of disulfide bond reduction during recombinant production of polypeptides
US20090110679A1 (en) 2007-07-13 2009-04-30 Luk-Chiu Li Methods and compositions for pulmonary administration of a TNFa inhibitor
KR100897159B1 (en) 2007-07-26 2009-05-14 보령제약 주식회사 A plant recombinant human CTLA4Ig and a method for producing the same
SG183709A1 (en) 2007-08-08 2012-09-27 Abbott Lab Compositions and methods for crystallizing antibodies
EP3327132A3 (en) 2007-08-09 2018-07-18 Wyeth LLC Use of perfusion to enhance production of fed-batch cell culture in bioreactors
WO2009032128A1 (en) 2007-08-28 2009-03-12 Abbott Biotechnology Ltd. Compositions and methods comprising binding proteins for adalimumab
EP2031064A1 (en) 2007-08-29 2009-03-04 Boehringer Ingelheim Pharma GmbH & Co. KG Method for increasing protein titres
BRPI0815889A2 (en) 2007-08-31 2014-10-14 Hoffmann La Roche Glycosylation Profile Analysis.
CN101896616B (en) 2007-10-12 2014-06-04 西格马-奥利奇股份有限公司 Compositions and methods for improved glycoprotein sialylation
EP2050764A1 (en) 2007-10-15 2009-04-22 sanofi-aventis Novel polyvalent bispecific antibody format and uses thereof
US9329169B2 (en) 2007-10-29 2016-05-03 University Of Georgia Research Foundation, Inc Vivo isotopic labeling method for quantitative glycomics
WO2009058769A1 (en) 2007-10-30 2009-05-07 Schering Corporation Purification of antibodies containing hydrophobic variants
NZ709704A (en) 2007-11-30 2017-03-31 Abbvie Biotechnology Ltd Protein formulations and methods of making same
US20130195888A1 (en) 2007-11-30 2013-08-01 Abbvie Ultrafiltration and diafiltration formulation methods for protein processing
US8883146B2 (en) 2007-11-30 2014-11-11 Abbvie Inc. Protein formulations and methods of making same
PT2235197T (en) 2007-12-27 2017-10-11 Baxalta Inc Cell culture processes
EP2799450A1 (en) 2007-12-31 2014-11-05 Bayer Intellectual Property GmbH Antibodies to TNFalpha
KR101589554B1 (en) 2008-01-03 2016-02-01 코넬 리서치 파운데이션 인코포레이티드 Glycosylated protein expression in prokaryotes
CN101965514A (en) 2008-01-03 2011-02-02 艾博特生物技术有限公司 Predicting long-term efficacy of a compound in the treatment of psoriasis
SG2013054218A (en) 2008-01-15 2014-10-30 Abbott Gmbh & Co Kg Powdered protein compositions and methods of making same
KR20100113112A (en) 2008-01-15 2010-10-20 아보트 러보러터리즈 Improved mammalian expression vectors and uses thereof
JP2011511777A (en) 2008-01-30 2011-04-14 アボット・ラボラトリーズ Compositions and methods for crystallizing antibody fragments
TWI472339B (en) 2008-01-30 2015-02-11 Genentech Inc Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
MX2010009398A (en) 2008-02-29 2010-11-12 Biogen Idec Inc Purified immunoglobulin fusion proteins and methods of their purification.
JP2011512875A (en) 2008-03-11 2011-04-28 ジェネンテック, インコーポレイテッド Antibody with enhanced ADCC function
MX2010010503A (en) 2008-03-24 2010-11-09 Abbott Biotech Ltd Methods and compositions for treating bone loss.
EP2262892A4 (en) 2008-04-08 2011-09-21 Amyris Biotechnologies Inc Expression of heterologous sequences
EP2282773B1 (en) 2008-05-02 2014-01-15 Seattle Genetics, Inc. Methods and compositions for making antibodies and antibody derivatives with reduced core fucosylation
WO2009135656A1 (en) 2008-05-06 2009-11-12 Lonza Biologics Plc. A method for the purification of antibodies using displacement chromatography
AU2009256250B2 (en) 2008-06-03 2013-05-30 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US8318416B2 (en) 2008-08-08 2012-11-27 Biogen Idec Ma Inc. Nutrient monitoring and feedback control for increased bioproduct production
PT2848625T (en) 2008-08-14 2019-10-25 Genentech Inc Methods for removing a contaminant using indigenous protein displacement ion exchange membrane chromatography
US20100069617A1 (en) 2008-09-12 2010-03-18 Ge Healthcare Bio-Sciences Ab Enhanced protein aggregate removal by mixed mode chromatography on hydrophobic interaction media in the presence of protein-excluded zwitterions
US8080415B2 (en) 2008-09-26 2011-12-20 Eureka Therapeutics, Inc. Modified host cells and uses thereof
WO2010043703A1 (en) 2008-10-17 2010-04-22 Dsm Ip Assets B.V. Removal of host cell proteins
NZ592097A (en) 2008-10-20 2013-01-25 Abbott Lab Viral inactivation during purification of il-12 and il-18 antibodies
TW201030016A (en) 2008-10-20 2010-08-16 Abbott Lab Antibodies that bind to IL-18 and methods of purifying the same
CA2911256A1 (en) 2008-10-20 2010-12-09 Robert K. Hickman Isolation and purification of antibodies using protein a affinity chromatography
GB0821100D0 (en) 2008-11-18 2008-12-24 Hansa Medical Ab Antibodies
AU2009327435A1 (en) 2008-12-19 2010-06-24 Momenta Pharmaceuticals, Inc. Methods related to modified glycans
US8729241B2 (en) 2008-12-19 2014-05-20 Momenta Pharmaceuticals, Inc. Characterization of O-linked glycans
US8614297B2 (en) 2008-12-22 2013-12-24 Hoffmann-La Roche Inc. Anti-idiotype antibody against an antibody against the amyloid β peptide
CA2748807C (en) 2009-01-08 2019-01-29 Ge Healthcare Bio-Sciences Ab Separation method using single polymer phase systems
AU2009338190C1 (en) 2009-01-22 2014-07-17 Momenta Pharmaceuticals, Inc. Galactose-alpha-1, 3-galactose-containing N-glycans in glycoprotein products derived from CHO cells
EP2400981A4 (en) 2009-02-26 2013-02-27 Lpath Inc Humanized platelet activating factor antibody design using anti-lipid antibody templates
CA2752393C (en) 2009-03-05 2020-01-14 Biogen Idec Ma Inc. Purification of immunoglobulins
EP2233499A1 (en) 2009-03-26 2010-09-29 CSL Behring AG Antibody composition with altered Fab sialylation
RU2542472C2 (en) 2009-04-01 2015-02-20 Эвек Инкорпорейтед Monoclonal antibody able to bind to specific discontinuous epitope placed in ad1 region of human cytomegalovirus gb glycoprotein, and its antigen-binding fragment
RU2011142230A (en) 2009-04-20 2013-05-27 Пфайзер Инк. PROTEIN GLYCOSYLIN CONTROL AND COMPOSITIONS AND METHODS CONCERNING THIS
US20120282654A1 (en) 2009-04-29 2012-11-08 Schering Corporation Antibody purification
TWI583418B (en) 2009-04-29 2017-05-21 艾伯維生物技術有限責任公司 Syringe plunger and automatic injection device
JP2012526121A (en) 2009-05-04 2012-10-25 アボツト・バイオテクノロジー・リミテツド Stable high protein concentration formulation of human anti-TNF alpha antibody
EP2435577A4 (en) 2009-05-26 2016-04-13 Momenta Pharmaceuticals Inc Production of glycoproteins
WO2010136515A1 (en) 2009-05-28 2010-12-02 Boehringer Ingelheim International Gmbh Method for a rational cell culturing process
AU2010252230B2 (en) 2009-05-28 2013-07-04 Eth Zurich N-glycan core beta-galactosyltransferase and uses thereof
US20120277165A1 (en) 2009-06-05 2012-11-01 Collins Brian E Methods of modulating fucosylation of glycoproteins
MX2012000354A (en) 2009-07-06 2012-02-01 Genentech Inc Method of culturing eukaryotic cells.
CA2768325C (en) 2009-07-24 2019-10-29 F. Hoffmann-La Roche Ag Optimizing the production of antibodies
US8945895B2 (en) 2009-07-31 2015-02-03 Baxter International Inc. Methods of purifying recombinant ADAMTS13 and other proteins and compositions thereof
WO2011015926A1 (en) 2009-08-03 2011-02-10 Avesthagen Limited A process of fermentation, purification and production of recombinant soluble tumour necrosis factor alfa receptor (tnfr) - human igg fc fusion protein
US20110039300A1 (en) 2009-08-10 2011-02-17 Robert Bayer Antibodies with enhanced adcc functions
EP3760712A1 (en) 2009-08-11 2021-01-06 F. Hoffmann-La Roche AG Production of proteins in glutamine-free cell culture media
US20110053223A1 (en) 2009-08-14 2011-03-03 Robert Bayer Cell culture methods to make antibodies with enhanced adcc function
WO2011024025A1 (en) 2009-08-28 2011-03-03 Avesthagen Limited An erythropoietin analogue and a method thereof
US9540426B2 (en) 2009-10-06 2017-01-10 Bristol-Myers Squibb Company Mammalian cell culture processes for protein production
US8470552B2 (en) 2009-10-12 2013-06-25 Keck Graduate Institute Strategy to reduce lactic acid production and control PH in animal cell culture
EP2325296A1 (en) 2009-11-20 2011-05-25 LEK Pharmaceuticals d.d. Production of glycoproteins with low N-glycolylneuraminic acid (Neu5Gc) content
WO2011065940A1 (en) 2009-11-24 2011-06-03 Biogen Idec Ma Inc. Method of supplementing culture media to prevent undesirable amino acid substitutions
US10087236B2 (en) 2009-12-02 2018-10-02 Academia Sinica Methods for modifying human antibodies by glycan engineering
US20110136682A1 (en) 2009-12-04 2011-06-09 Momenta Pharmaceuticals, Inc. Antennary fucosylation in glycoproteins from cho cells
CN102666568B (en) 2009-12-18 2015-12-09 杰特有限公司 The method of purified polypeptide
US20130178608A1 (en) 2009-12-29 2013-07-11 Samir Kulkarni Protein purification by ion exchange
JP2013518590A (en) 2010-02-02 2013-05-23 アボツト・バイオテクノロジー・リミテツド Methods and compositions for predicting responsiveness to treatment with a TNF-α inhibitor
US20120309056A1 (en) 2010-02-04 2012-12-06 Leon Arnaud Fed-batch process using concentrated cell culture medium for the efficient production of biologics in eb66 cells
KR20120118065A (en) 2010-02-12 2012-10-25 디에스엠 아이피 어셋츠 비.브이. Single unit antibody purification
JP5814951B2 (en) 2010-03-10 2015-11-17 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Method for purifying immunoglobulin solutions
WO2011112892A1 (en) 2010-03-12 2011-09-15 Purecircle Usa Inc. High-purity steviol glycosides
MX2012011648A (en) 2010-04-07 2012-11-29 Momenta Pharmaceuticals Inc High mannose glycans.
RU2624027C2 (en) * 2010-04-23 2017-06-30 Дженентек, Инк. Heteromultimeric proteins production
US20110262965A1 (en) 2010-04-23 2011-10-27 Life Technologies Corporation Cell culture medium comprising small peptides
PL3330370T3 (en) 2010-04-26 2021-09-20 Novartis Ag Process for cultivation of cho cells
SG185038A1 (en) 2010-04-26 2012-11-29 Novartis Ag Improved cell culture medium
WO2011150241A2 (en) 2010-05-28 2011-12-01 Genentech, Inc. Decreasing lactate level and increasing polypeptide production by downregulating the expression of lactate dehydrogenase and pyruvate dehydrogenase kinase
SI2575884T1 (en) 2010-06-03 2018-10-30 Abbvie Biotechnology Ltd Uses and compositions for treatment of hidradenitis suppurativa (hs)
JP5980782B2 (en) 2010-07-30 2016-08-31 ファイザー・インク Protein tandem purification
SG187198A1 (en) 2010-08-05 2013-03-28 Amgen Inc Dipeptides to enhance yield and viability from cell cultures
DE102010038990A1 (en) 2010-08-06 2012-02-09 Robert Bosch Gmbh Shaft bearing device for a hand tool
WO2012030512A1 (en) 2010-09-03 2012-03-08 Percivia Llc. Flow-through protein purification process
CN103119448A (en) 2010-09-17 2013-05-22 Abbvie公司 Raman spectroscopy for bioprocess operations
CN105753933A (en) 2010-09-20 2016-07-13 Abbvie 公司 Purification Of Antibodies By Using Simulated Moving Bed Chromatography
US20120258496A1 (en) 2010-09-27 2012-10-11 Boehringer Ingelheim International Gmbh Production of low fucose antibodies in h4-ii-e rat cells
NZ609903A (en) 2010-10-08 2015-04-24 Cadila Healthcare Ltd Expression vector for high level expression of recombinant proteins
JP6023715B2 (en) 2010-10-11 2016-11-09 アッヴィ・バハマズ・リミテッド Protein purification method
US20130224797A1 (en) 2010-10-15 2013-08-29 Jcr Pharmaceuticals Co., Ltd. Method for producing glycoprotein having mannose residue as non-reducing end of sugar chain
EP2450375A1 (en) 2010-11-09 2012-05-09 Sandoz Gmbh Cell culture medium and process for protein expression, said medium and process comprising a PAM inhibitor
MX344727B (en) 2010-11-11 2017-01-05 Abbvie Biotechnology Ltd IMPROVED HIGH CONCENTRATION ANTI-TNFa ANTIBODY LIQUID FORMULATIONS.
WO2012068134A1 (en) 2010-11-15 2012-05-24 Biogen Idec Inc. Enrichment and concentration of select product isoforms by overloaded bind and elute chromatography
US20120149878A1 (en) 2010-12-08 2012-06-14 Gillespie Ronald Protein purification
RU2615448C2 (en) 2010-12-28 2017-04-04 Чугаи Сейяку Кабусики Кайся Description of animal cell cultivation method
CN103596978A (en) 2011-01-07 2014-02-19 阿布维公司 Anti-IL-12/IL-23 antibodies and uses thereof
TW201309330A (en) 2011-01-28 2013-03-01 Abbott Lab Compositions containing glycosylated antibodies and uses thereof
AR085302A1 (en) 2011-02-24 2013-09-18 Sanofi Sa METHOD OF PRODUCTION OF STIRATED ANTIBODIES
ES2687771T3 (en) 2011-03-06 2018-10-29 Merck Serono Sa Cell lines with low level of fucose and its uses
CA2829629A1 (en) 2011-03-10 2012-09-13 Board Of Regents, The University Of Texas System Protein nanoparticle dispersions
US20120238730A1 (en) 2011-03-15 2012-09-20 Abbott Laboratories Integrated approach to the isolation and purification of antibodies
TWI743461B (en) 2011-03-28 2021-10-21 法商賽諾菲公司 Dual variable region antibody-like binding proteins having cross-over binding region orientation
EP2511293A1 (en) 2011-04-13 2012-10-17 LEK Pharmaceuticals d.d. A method for controlling the main complex N-glycan structures and the acidic variants and variability in bioprocesses producing recombinant proteins
WO2012145682A1 (en) 2011-04-21 2012-10-26 Amgen Inc. A method for culturing mammalian cells to improve recombinant protein production
WO2012149197A2 (en) 2011-04-27 2012-11-01 Abbott Laboratories Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9670519B2 (en) 2011-04-29 2017-06-06 Biocon Limited Methods for reducing accumulation of lactate during culturing and method for producing polypeptide
US20140108084A1 (en) 2012-10-12 2014-04-17 Crestron Electronics, Inc. Initiating Schedule Management Via Radio Frequency Beacons
MY169935A (en) 2011-04-29 2019-06-18 Biocon Biologics India Ltd "a method for reducing heterogeneity of antibodies and a process of producing the antibodies thereof"
EA027410B1 (en) 2011-04-29 2017-07-31 Селекта Байосайенсиз, Инк. Tolerogenic nanocarriers to reduce cytotoxic t lymphocyte responses
EP3388443A1 (en) 2011-05-13 2018-10-17 Biogen MA Inc. Methods of preventing and removing trisulfide bonds
CN107988166B (en) 2011-07-01 2022-03-15 美国安进公司 Mammalian cell culture
WO2013006461A1 (en) 2011-07-01 2013-01-10 Biogen Idec Ma Inc. Cholesterol-based media supplementals for cell culture
WO2013004841A1 (en) 2011-07-06 2013-01-10 Genmab A/S Modulation of complement-dependent cytotoxicity through modifications of the c-terminus of antibody heavy chains
WO2013009648A2 (en) 2011-07-08 2013-01-17 Momenta Pharmaceuticals, Inc. Cell culture process
GB201112429D0 (en) 2011-07-19 2011-08-31 Glaxo Group Ltd Antigen-binding proteins with increased FcRn binding
WO2013013013A2 (en) 2011-07-21 2013-01-24 Alnylam Pharmaceuticals, Inc. Compositions and methods for producing modified glycoproteins
JP6433786B2 (en) 2011-08-10 2018-12-05 ラボラトワール フランセ デュ フラクショヌマン エ デ ビオテクノロジーLaboratoire Francais du Fractionnement et des Biotechnologies Highly galactosylated antibody
US20130149300A1 (en) 2011-09-27 2013-06-13 Icon Genetics Gmbh MONOCLONAL ANTIBODIES WITH ALTERED AFFINITIES FOR HUMAN FCyRI, FCyRIIIa, AND C1q PROTEINS
EP2773439A4 (en) 2011-10-31 2015-07-01 Merck Sharp & Dohme Chromatography process for resolving heterogeneous antibody aggregates
US20140301977A1 (en) 2011-11-02 2014-10-09 Genentech, Inc. Overload and elute chromatography
SG10202001596VA (en) 2011-12-19 2020-04-29 Univ Rockefeller Non-sialylated anti-inflammatory polypeptides
SG11201405475UA (en) 2012-03-07 2014-10-30 Cadila Healthcare Ltd Pharmaceutical formulations of tnf-alpha antibodies
SG10201701224UA (en) 2012-03-12 2017-04-27 Merck Patent Gmbh Removal of protein aggregates from biopharmaceutical preparations in a flowthrough mode
WO2013158273A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Methods to modulate c-terminal lysine variant distribution
US9334319B2 (en) 2012-04-20 2016-05-10 Abbvie Inc. Low acidic species compositions
WO2013158275A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Cell culture methods to reduce acidic species
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
US20130281355A1 (en) 2012-04-24 2013-10-24 Genentech, Inc. Cell culture compositions and methods for polypeptide production
US20140154270A1 (en) 2012-05-21 2014-06-05 Chen Wang Purification of non-human antibodies using kosmotropic salt enhanced protein a affinity chromatography
WO2013177118A2 (en) 2012-05-21 2013-11-28 Abbvie Inc. Novel purification of non-human antibodies using protein a affinity chromatography
WO2013176754A1 (en) 2012-05-24 2013-11-28 Abbvie Inc. Novel purification of antibodies using hydrophobic interaction chromatography
WO2013181585A2 (en) 2012-06-01 2013-12-05 Momenta Pharmaceuticals, Inc. Methods related to adalimumab
WO2013186230A1 (en) 2012-06-12 2013-12-19 Boehringer Ingelheim International Gmbh Pharmaceutical formulation for a therapeutic antibody
WO2014018747A2 (en) 2012-07-26 2014-01-30 Momenta Pharmaceuticals, Inc. Glycoproteins with anti-inflammatory properties
EP2885639B1 (en) 2012-08-17 2019-04-10 MorphoSys AG Complex-specific antibodies and antibody fragments and their use
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
KR20150043523A (en) 2012-09-02 2015-04-22 애브비 인코포레이티드 Methods to control protein heterogeneity
PE20191815A1 (en) 2012-09-07 2019-12-27 Coherus Biosciences Inc STABLE AQUEOUS FORMULATIONS OF ADALIMUMAB
EP2900264A4 (en) 2012-09-26 2016-05-25 Momenta Pharmaceuticals Inc Glycoprotein preparations
WO2014096672A1 (en) 2012-12-17 2014-06-26 Laboratoire Francais Du Fractionnement Et Des Biotechnologies Use of monoclonal antibodies for the treatment of inflammation and bacterial infections
WO2014099636A1 (en) 2012-12-18 2014-06-26 Merck Sharp & Dohme Corp. Liquid formulations for an anti-tnf alpha antibody
MX2015010427A (en) 2013-02-13 2016-03-17 Lab Francais Du Fractionnement Highly galactosylated anti-tnf-alpha antibodies and uses thereof.
SG11201506926SA (en) 2013-03-08 2015-09-29 Neuclone Biolog Pty Ltd A cell expression system
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
US8956830B2 (en) 2013-03-14 2015-02-17 Momenta Pharmaceuticals, Inc. Methods of cell culture
US9677105B2 (en) 2013-03-14 2017-06-13 Momenta Pharmaceuticals, Inc. Methods of cell culture
WO2014151878A2 (en) 2013-03-14 2014-09-25 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosacharides
WO2014158231A1 (en) 2013-03-14 2014-10-02 Abbvie Inc. Low acidic species compositions and methods for producing and using the same
WO2014159579A1 (en) 2013-03-14 2014-10-02 Abbvie Inc. MUTATED ANTI-TNFα ANTIBODIES AND METHODS OF THEIR USE
US20140271622A1 (en) 2013-03-14 2014-09-18 Momenta Pharmaceuticals, Inc. Methods of cell culture
WO2014151901A1 (en) 2013-03-14 2014-09-25 Abbvie Inc. Improvement of mammalian cell culture performance through surfactant supplementation of feed media
US9217168B2 (en) 2013-03-14 2015-12-22 Momenta Pharmaceuticals, Inc. Methods of cell culture
EP2970865B1 (en) 2013-03-15 2020-09-02 Alder Biopharmaceuticals, Inc. Antibody purification and purity monitoring
CA2907140A1 (en) 2013-03-15 2014-09-25 Janssen Biotech, Inc. Manufacturing methods to control c-terminal lysine, galactose and sialic acid content in recombinant proteins
US20160108450A1 (en) 2013-05-02 2016-04-21 Momenta Pharmaceutcals, Inc. Sialylated glycoproteins
KR101569783B1 (en) 2013-06-05 2015-11-19 한화케미칼 주식회사 A Method of Antibody Purification
AR096713A1 (en) 2013-06-25 2016-01-27 Cadila Healthcare Ltd PURIFICATION PROCESS FOR MONOCLONAL ANTIBODIES
BR112015032800A2 (en) 2013-07-06 2017-07-25 Cadila Healthcare Ltd PROCESS FOR THE PRODUCTION OF AN ANTIBODY
CA2914776C (en) 2013-07-19 2018-08-07 Hexal Ag Methods and formulations which allow the modulation of immune responses related to the administration of a biopharmaceutical drug
EP3027625B1 (en) 2013-07-31 2018-05-30 Merck Sharp & Dohme Corp. Spiro-fused derivatives of piperidine useful for the treatment of inter alia hypertension and acute or chronic heart failure
PL3036320T3 (en) 2013-08-19 2021-11-02 Biogen Ma Inc. Control of protein glycosylation by culture medium supplementation and cell culture process parameters
BR112016002622B1 (en) 2013-08-20 2023-04-11 Lek Pharmaceuticals D.D CELL CULTURE MEDIA AND USE OF SELENIUM
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant 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
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
WO2015073884A2 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
JP6797458B2 (en) 2014-01-29 2020-12-09 エルジー・ケム・リミテッド How to regulate galactosylation of recombinant proteins by optimizing the culture medium
WO2016007764A1 (en) 2014-07-09 2016-01-14 Abbvie Inc. Methods for modulating the glycosylation profile of recombinant proteins using non-commonly used sugars
US20160039924A1 (en) 2014-08-05 2016-02-11 Abbvie Inc. Methods for modulating the glycosylation profile of recombinant proteins using dissolved oxygen
CA2969889A1 (en) 2014-12-22 2016-06-30 Ucb Biopharma Sprl Protein manufacture
CN105777896B (en) 2015-03-19 2019-08-16 广东东阳光药业有限公司 A kind of purification process at antibody acidity peak
CN105777895A (en) 2015-03-19 2016-07-20 广东东阳光药业有限公司 Application of sodium phenylbutyrate in purification of antibody acidic peak
US20160280767A1 (en) 2015-03-23 2016-09-29 Lonza Ltd. Methods for controlling protein glycosylation
CN105777904B (en) 2015-03-23 2019-12-10 广东东阳光药业有限公司 cation exchange chromatography purification method of anti-TNF alpha monoclonal antibody

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11369896B2 (en) 2016-08-16 2022-06-28 Regeneron Pharmaceuticals, Inc. Methods for quantitating individual antibodies from a mixture
US11571636B2 (en) 2016-08-16 2023-02-07 Regeneron Pharmaceuticals, Inc. Methods for quantitating individual antibodies from a mixture
US11850535B2 (en) 2016-08-16 2023-12-26 Regeneron Pharmaceuticals, Inc. Methods for quantitating individual antibodies from a mixture
US11333642B2 (en) 2016-10-25 2022-05-17 Regeneran Pharmaceuticals, Inc. Methods and systems for chromatography data analysis
US11680930B2 (en) 2016-10-25 2023-06-20 Regeneron Pharmaceuticals, Inc. Methods and systems for chromatography data analysis
US11884698B2 (en) 2018-07-02 2024-01-30 Regeneron Pharmaceuticals, Inc. Systems and methods for preparing a polypeptide from a mixture
WO2021220253A1 (en) * 2020-05-01 2021-11-04 Kashiv Biosciences, Llc An improved process of affinity chromatography
WO2023053031A1 (en) * 2021-09-28 2023-04-06 Kashiv Biosciences, Llc An improved process of purification of fusion protein
WO2023053030A1 (en) * 2021-09-28 2023-04-06 Kashiv Biosciences, Llc An improved process for purification of protein

Also Published As

Publication number Publication date
US20130338344A1 (en) 2013-12-19
US9359434B2 (en) 2016-06-07
US20140161815A9 (en) 2014-06-12
US20160251425A1 (en) 2016-09-01
US9683033B2 (en) 2017-06-20
US20130344084A1 (en) 2013-12-26
US20150141623A1 (en) 2015-05-21
US20170355761A1 (en) 2017-12-14
US9334319B2 (en) 2016-05-10
WO2013158279A1 (en) 2013-10-24
US20160145331A1 (en) 2016-05-26
US9150645B2 (en) 2015-10-06
US20160039925A1 (en) 2016-02-11
US9346879B2 (en) 2016-05-24
US9957318B2 (en) 2018-05-01

Similar Documents

Publication Publication Date Title
US9957318B2 (en) Protein purification methods to reduce acidic species
US9249182B2 (en) Purification of antibodies using hydrophobic interaction chromatography
US9708399B2 (en) Protein purification using displacement chromatography
US9200069B2 (en) Low acidic species compositions and methods for producing and using the same
CA2899308C (en) Low acidic species adalimumab compositions and uses thereof
US20130336957A1 (en) Novel purification of human, humanized, or chimeric antibodies using protein a affinity chromatography
WO2013158275A1 (en) Cell culture methods to reduce acidic species
JP2024514306A (en) Purification of antibodies by mixed mode chromatography
WO2014142882A1 (en) Protein purification using displacement chromatography
AU2021401316A9 (en) Protein compositions and methods for producing and using the same

Legal Events

Date Code Title Description
AS Assignment

Owner name: ABBVIE, INC., ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:RAMASUBRAMANYAN, NATARAJAN;YANG, LIHUA;HERIGSTAD, MATTHEW OMON;AND OTHERS;SIGNING DATES FROM 20131212 TO 20140903;REEL/FRAME:045522/0159

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION