Method of cleaning and/or sanitizing a separation matrix

Information

  • Patent Grant
  • 11753438
  • Patent Number
    11,753,438
  • Date Filed
    Wednesday, May 10, 2017
    7 years ago
  • Date Issued
    Tuesday, September 12, 2023
    8 months ago
Abstract
The present invention concerns a method of cleaning and/or sanitizing a separation matrix comprising multimers of immunoglobulin-binding alkali-stabilized Protein A domains covalently coupled to a porous support. The method comprises the steps of: a) optionally purifying a mixture comprising a first immunoglobulin using the separation matrix; b) providing a cleaning liquid comprising at least 50% by volume of an aqueous alkali metal hydroxide solution; and c) cleaning and/or sanitizing the separation matrix by contacting the cleaning liquid with the separation matrix for a predetermined contact time. The alkali-stabilized Protein A domains comprise mutants of a parental Fc-binding domain of Staphylococcus Protein A (SpA), as defined by SEQ ID NO 51 or SEQ ID NO 52, wherein the amino acid residues at positions 13 and 44 of SEQ ID NO 51 or 52 are asparagines and wherein at least the asparagine residue at position 3 of SEQ ID NO 51 or 52 has been mutated to an amino acid selected from the group consisting of glutamic acid, lysine, tyrosine, threonine, phenylalanine, leucine, isoleucine, tryptophan, methionine, valine, alanine, histidine and arginine.
Description
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims the priority benefit of PCT/EP2017/061159 filed on May 10, 2017 which claims priority benefit of Great Britain Application Nos. 1608229.9 and 1608232.3, both of which were filed May 11, 2016 and U.S. application Ser. Nos. 15/282,367 and 15/348,669, filed Sep. 30, 2016 and Nov. 10, 2016, respectively. The entire contents of which are hereby incorporated by reference.


SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Nov. 17, 2022, is named 315715AA-US-5_34428-0292_SL.txt and is 80,033 bytes in size.


TECHNICAL FIELD

The present invention relates to a method of cleaning and/or sanitizing separation matrices that comprise multimers of immunoglobulin-binding alkali-stabilized Protein A domains covalently coupled to a porous support. The present invention further relates to a method of preventing carryover in the purification of immunoglobulins with a separation matrix comprising multimers of immunoglobulin-binding alkali-stabilized Protein A domains covalently coupled to a porous support, as well as to use of a cleaning liquid comprising at least 70% by volume of an aqueous alkali metal hydroxide solution for the sanitization of a separation matrix.


BACKGROUND ART

Immunoglobulins represent the most prevalent biopharmaceutical products in either manufacture or development worldwide. The high commercial demand for and hence value of this particular therapeutic market has led to the emphasis being placed on pharmaceutical companies to maximize the productivity of their respective immunoglobulin manufacturing processes whilst controlling the associated costs.


Affinity chromatography is used in most cases, as one of the key steps in the purification of these immunoglobulin molecules, such as monoclonal antibodies (mAbs) or polyclonal antibodies (pAbs). A particularly interesting class of affinity reagents is proteins capable of specific binding to invariable parts of an immunoglobulin molecule, such interaction being independent on the antigen-binding specificity of the antibody. Such reagents can be widely used for affinity chromatography recovery of immunoglobulins from different samples such as but not limited to serum or plasma preparations or cell culture derived feed stocks. An example of such a protein is staphylococcal protein A, containing domains capable of binding to the Fc and Fab portions of IgG immunoglobulins from different species. These domains are commonly denoted as the E-, D-, A-, B- and C-domains.


Staphylococcal protein A (SpA) based reagents have due to their high affinity and selectivity found a widespread use in the field of biotechnology, e.g. in affinity chromatography for capture and purification of antibodies as well as for detection or quantification. At present, SpA-based affinity medium probably is the most widely used affinity medium for isolation of monoclonal antibodies and their fragments from different samples including industrial cell culture supernatants. Accordingly, various matrices comprising protein A-ligands are commercially available, for example, in the form of native protein A (e.g. Protein A SEPHAROSE™, GE Healthcare, Uppsala, Sweden) and also comprised of recombinant protein A (e.g. rProtein A SEPHAROSE™, GE Healthcare). More specifically, the genetic manipulation performed in the commercial recombinant protein A product is aimed at facilitating the attachment thereof to a support and at increasing the productivity of the ligand.


These applications, like other affinity chromatography applications, require comprehensive attention to definite removal of contaminants. Such contaminants can for example be non-eluted molecules adsorbed to the stationary phase or matrix in a chromatographic procedure, such as non-desired biomolecules or microorganisms, including for example proteins, carbohydrates, lipids, endotoxins, bacteria and viruses. The removal of such contaminants from the matrix is usually performed after a first elution of the desired product in order to regenerate the matrix before subsequent use. Cleaning the matrix of contaminants usually involves a procedure known as cleaning-in-place (CIP), wherein agents capable of eluting contaminants from the stationary phase are used. Inactivation of microorganisms usually involves a procedure known as sanitization-in-place (SIP) wherein agents capable of inactivating microorganisms are used. Some agents are capable of both cleaning and sanitizing the separation matrix, depending on the concentration of the agent and the contact time with the separation matrix. One such class of agents often used is alkaline solutions that are passed over said stationary phase. At present the most extensively used cleaning and sanitizing agent for most separation matrices is NaOH, and the concentration thereof can range from 0.1 up to e.g. 1 M, depending on the degree and nature of contamination. However, this strategy is associated with exposing the matrix to solutions with pH-values above 13. For many affinity chromatography matrices containing proteinaceous affinity ligands such alkaline environment is a very harsh condition and consequently results in decreased capacities owing to instability of the ligand to the high pH involved.


An extensive research has therefore been focused on the development of engineered protein ligands that exhibit an improved capacity to withstand alkaline pH-values. For example, Gülich et al. (Susanne Gülich, Martin Linhult, Per-Åke Nygren, Mathias Uhlén, Sophia Hober, Journal of Biotechnology 80 (2000), 169-178) suggested protein engineering to improve the stability properties of a Streptococcal albumin-binding domain (ABD) in alkaline environments. Gülich et al. created a mutant of ABD, wherein all the four asparagine residues have been replaced by leucine (one residue), aspartate (two residues) and lysine (one residue). Further, Gülich et al. report that their mutant exhibits a target protein binding behavior similar to that of the native protein, and that affinity columns containing the engineered ligand show higher binding capacities after repeated exposure to alkaline conditions than columns prepared using the parental non-engineered ligand. Thus, it is concluded therein that all four asparagine residues can be replaced without any significant effect on structure and function.


Recent work shows that changes can also be made to protein A (SpA) to effect similar properties. US patent application publication US 2005/0143566, which is hereby incorporated by reference in its entirety, discloses that when at least one asparagine residue is mutated to an amino acid other than glutamine or aspartic acid, the mutation confers an increased chemical stability at pH-values of up to about 13-14 compared to the parental SpA, such as the B-domain of SpA, or Protein Z, a synthetic construct derived from the B-domain of SpA (U.S. Pat. No. 5,143,844, incorporated by reference in its entirety). The authors show that when these mutated proteins are used as affinity ligands, the separation media as expected can better withstand cleaning procedures using alkaline agents. Further mutations of protein A domains with the purpose of increasing the alkali stability have also been published in U.S. Pat. No. 8,329,860, JP 2006304633A, U.S. Pat. No. 8,674,073, US 2010/0221844, US 2012/0208234, U.S. Pat. No. 9,051,375, US 2014/0031522, US 2013/0274451 and WO 2014/146350, all of which are hereby incorporated by reference in their entireties. However, the currently available mutants are still sensitive to alkaline pH and the NaOH concentration during cleaning is usually limited to 0.1 M, which means that complete cleaning is difficult to achieve. Moreover, such a concentration of NaOH is insufficient in itself for effective sanitization of the affinity separation matrix, and therefore sanitization is often performed using solutions comprising alcohols such as ethanol or isopropanol. Higher NaOH concentrations, which would improve the cleaning and sanitization, lead to unacceptable capacity losses of the affinity separation matrix.


There is thus still a need in this field to obtain a separation matrix containing protein ligands having a further improved stability towards alkaline cleaning and sanitization procedures. There is also a need for such separation matrices with an improved binding capacity to allow for economically efficient purification of therapeutic antibodies.


SUMMARY OF THE INVENTION

The inventors of the present invention have recognised that alcohol solutions are suboptimal for the sanitization of affinity separation matrices. Alcohols are flammable, subject to regulation, and difficult to dispose of. Moreover, the inventors of the present invention have recognised that low concentration NaOH, such as 0.1 M NaOH, provides insufficient cleaning in order to allow the separation matrix to be used for the purification of a variety of different immunoglobulin products. Instead, the separation matrix is typically only used to purify separate batches of a single product in order to prevent carryover of impurities from one product to another. The inventors have recognised that concentrated aqueous alkali metal hydroxide solutions have a number of advantages as cleaning and storage solutions. Concentrated alkali metal hydroxide solutions are bacteriostatic and can inactive most viruses, bacteria, yeasts, fungi and endotoxins. They are capable of effectively stripping the separation matrix of impurities to such a degree that the same separation matrix may be used to purify a variety of immunoglobulins. This facilitates the use of the separation matrix as a separation platform. Moreover, concentrated alkali metal hydroxide solutions are relatively cheap, easily disposed of, and removal from the separation matrix is simple to detect using pH and/or conductivity measurements.


It is therefore an object of the present invention to provide a method for cleaning and sanitizing an affinity separation matrix for the purification of immunoglobulins which permits the use of concentrated alkali metal hydroxide solution.


This object is achieved by the method according to the appended claims of cleaning and/or sanitizing a separation matrix comprising multimers of immunoglobulin-binding alkali-stabilized Protein A domains covalently coupled to a porous support. The alkali-stabilized Protein A domains comprise mutants of a parental Fc-binding domain of Staphylococcus Protein A (SpA), as defined by, or having at least 80% such as at least 90%, 95% or 98% identity to, SEQ ID NO: 51 or SEQ ID NO: 52, wherein the amino acid residues at positions 13 and 44 of SEQ ID NOs: 51 or 52 are asparagines and wherein at least the asparagine residue at position 3 of SEQ ID NOs: 51 or 52 has been mutated to an amino acid selected from the group consisting of glutamic acid, lysine, tyrosine, threonine, phenylalanine, leucine, isoleucine, tryptophan, methionine, valine, alanine, histidine and arginine. The method comprises the steps of:

    • a) optionally purifying a mixture comprising a first immunoglobulin using the separation matrix;
    • b) providing a cleaning liquid comprising at least 50% by volume of an aqueous alkali metal hydroxide solution; and
    • c) cleaning and/or sanitizing the separation matrix by contacting the cleaning liquid with the separation matrix for a predetermined contact time.


By using a separation matrix comprising multimers of immunoglobulin-binding alkali-stabilized Protein A domains as defined above, a highly alkali-stable separation matrix having a high dynamic binding capacity is obtained. The inventors of the present invention have observed that such separation matrices are relatively stable upon impregnation with concentrated aqueous alkali metal hydroxide solution, and substantially retain dynamic binding capacity after contact with such concentrated alkali metal hydroxide solution. This means that such a separation matrices are suitable for cleaning and/or sanitization with concentrated alkali metal hydroxide solutions.


Further mutations to the immunoglobulin-binding alkali-stabilized Protein A domains may provide further enhancement of properties such as enhanced alkali stability. For example, the glutamine residue at position 1 of SEQ ID NOs: 51 or 52 may be mutated to an alanine; and/or the asparagine or glutamic acid residue at position 35 of SEQ ID NOs: 51 or 52 may be mutated to an alanine.


The multimers of immunoglobulin-binding alkali-stabilized Protein A domains may be homomultimers selected from the group consisting of dimers, trimers, tetramers, pentamers, hexamers, heptamers, octamers or nonamers. By using an appropriate multimer, the immunoglobulin binding capacity and alkali stability of the separation matrix may be increased.


The multimers of immunoglobulin-binding alkali-stabilized Protein A domains may each comprise a C-terminal cysteine residue for covalent coupling to the porous support. The multimers of immunoglobulin-binding alkali-stabilized Protein A domains may be coupled to the porous support via thioether links. This provides a robust, alkali-stable and well-proven method of attaching the ligands to the solid support.


The separation matrix may comprise at least 11 mg/ml, such as at least 15 mg/ml, of the multimers of immunoglobulin-binding alkali-stabilized Protein A domains covalently coupled to the porous support. This ensures a separation matrix with a good binding capacity.


The porous support may comprise cross-linked polymer particles having a volume-weighted median diameter (d50,v) of 56-70 micrometers and a dry solids weight of 55-80 mg/ml. The porous support may for example be highly cross-linked agarose beads.


The aqueous alkali metal hydroxide solution used in the cleaning liquid may be sodium hydroxide solution, potassium hydroxide solution or a mixture thereof, preferably sodium hydroxide solution. Sodium hydroxide solution is relatively cheap, readily available and widely accepted for use as a cleaning and sanitization solution. The aqueous alkali metal hydroxide solution may have a molarity of from 500 mM to 5 M, such as from 1 M to 2 M. This ensures a solution with good bactericidal properties.


The cleaning liquid may in some instances further comprise a C2-C7 alcohol, such as ethanol, isopropanol or benzyl alcohol. A cleaning liquid combining an alcohol and an alkali metal hydroxide may be more effective in inactivating certain microorganisms, such as some spore-forming bacteria.


The cleaning liquid may comprise at least 70% by volume aqueous alkali metal hydroxide solution, such as at least 90% by volume aqueous alkali metal hydroxide solution, preferably at least 99% by volume aqueous alkali metal hydroxide solution.


In some instances the cleaning liquid may consist of, or consist essentially of, aqueous alkali metal hydroxide solution.


The predetermined contact time may be a time sufficient to provide a 6-log10 reduction in endotoxin concentration and/or microorganism concentration in the separation matrix. The predetermined contact time may be from 10 minutes to 50 hours, such as from 30 minutes to 24 hours, or such as from 1 hour to 12 hours. Such contact times are typically sufficient to inactivate a wide range of microorganisms.


Steps a)-c) of the method above may be repeated at least 10 times, such as at least 50 times or 50-200 times. Thus, the separation matrix may be reused many times while still retaining sufficient capacity to achieve an acceptable purification.


The object of the invention is further achieved by a method of preventing carryover in the purification of immunoglobulins with a separation matrix comprising multimers of immunoglobulin-binding alkali-stabilized Protein A domains covalently coupled to a porous support, as defined by the appended claims. The method comprises the steps of purifying a first immunoglobulin and cleaning and sanitizing the separation matrix by performing the steps a)-c) of the method of cleaning and/or sanitizing a separation matrix as described above. The method further comprises the step of

    • d) purifying a mixture comprising a second immunoglobulin using the separation matrix, wherein the second immunoglobulin is different from the first immunoglobulin.


The alkali-stabilized Protein A domains comprise mutants of a parental Fc-binding domain of Staphylococcus Protein A (SpA), as defined by, or having at least 80% such as at least 90%, 95% or 98% identity to, SEQ ID NO: 51 or SEQ ID NO: 52, wherein the amino acid residues at positions 13 and 44 of SEQ ID NOs: 51 or 52 are asparagines and wherein at least the asparagine residue at position 3 of SEQ ID NOs: 51 or 52 has been mutated to an amino acid selected from the group consisting of glutamic acid, lysine, tyrosine, threonine, phenylalanine, leucine, isoleucine, tryptophan, methionine, valine, alanine, histidine and arginine.


Further objects, advantages and novel features of the present invention will become apparent to one skilled in the art from the following detailed description.


Definitions

The terms “antibody” and “immunoglobulin” are used interchangeably herein, and are understood to include also fragments of antibodies, fusion proteins comprising antibodies or antibody fragments and conjugates comprising antibodies or antibody fragments.


The terms an “Fc-binding polypeptide”, “Fc-binding domain” and “Fc-binding protein” mean a polypeptide, domain or protein respectively, capable of binding to the crystallisable part (Fc) of an antibody and includes e.g. Protein A and Protein G, or any fragment or fusion protein thereof that has maintained said binding property.


The term “linker” herein means an element linking two polypeptide units, monomers or domains to each other in a multimer.


The term “spacer” herein means an element connecting a polypeptide or a polypeptide multimer to a support.


The term “% identity” with respect to comparisons of amino acid sequences is determined by standard alignment algorithms such as, for example, Basic Local Alignment Tool (BLAST™) described in Altshul et al. (1990) J. Mol. Biol., 215: 403-410. A web-based software for this is freely available from the US National Library of Medicine. Here, the algorithm “blastp (protein-protein BLAST)” is used for alignment of a query sequence with a subject sequence and determining i.a. the % identity.


As used herein, the terms “comprises,” “comprising,” “containing,” “having” and the like can have the meaning ascribed to them in U.S. Patent law and can mean “includes,” “including,” and the like; “consisting essentially of” or “consists essentially” likewise has the meaning ascribed in U.S. Patent law and the term is open-ended, allowing for the presence of more than that which is recited so long as basic or novel characteristics of that which is recited is not changed by the presence of more than that which is recited, but excludes prior art embodiments.





BRIEF DESCRIPTION OF THE DRAWINGS

For a fuller understanding of the present invention and further objects and advantages of it, the detailed description set out below should be read together with the accompanying figures, and in which:



FIG. 1 shows an alignment of the Fc-binding domains as defined by SEQ ID NOs: 1-7 and 51-52.



FIG. 2 shows results from Example 2 for the alkali stability of parental and mutated tetrameric Zvar (SEQ ID NO: 7) polypeptide variants coupled to an SPR biosensor chip.



FIG. 3 shows results from Example 4 for the alkali stability (0.5 M NaOH) of parental and mutated tetrameric Zvar (SEQ ID NO: 7) polypeptide variants coupled to agarose beads.



FIG. 4 shows results from Example 4 for the alkali stability (1.0 M NaOH) of parental and mutated tetrameric Zvar (SEQ ID NO: 7) polypeptide variants coupled to agarose beads.



FIG. 5 shows results from Example 7 for the alkali stability (1.0 M NaOH) of agarose beads with different amounts of mutated multimer variants (SEQ ID NO: 20) coupled. The results are plotted as the relative remaining dynamic capacity (Qb10%, 6 min residence time) vs. incubation time in 1 M NaOH.



FIG. 6 shows results from Example 7 for the alkali stability (1.0 M NaOH) of agarose beads with different amounts of mutated multimer variants (SEQ ID NO: 20) coupled. The results are plotted as the relative remaining dynamic capacity (Qb10%, 6 min residence time) after 31 h incubation in 1 M NaOH vs. the ligand content of the prototypes.



FIG. 7 shows results from a pH gradient elution of polyclonal human IgG a) from the reference matrix MabSelect SuRe LX and b) a matrix according to the invention.



FIG. 8 shows analyses of the IgG1, IgG2 and IgG4 components in fractions from the chromatograms of FIG. 7. a) reference matrix and b) matrix according to the invention. For each fraction, the first bar (blue) represents IgG1, the second (red) IgG 4 and the third (green) IgG 2.



FIG. 9 shows results from accelerated alkali stability measurements with 1 M NaOH incubation for the reference matrix MabSelect SuRe LX (MSS LX) and a matrix according to the invention. The stability is expressed as the percentage of the 10% breakthrough capacity remaining after incubation.



FIG. 10 shows results from repeated CIP cycles using 2M NaOH as the cleaning liquid for the reference matrix MabSelect SuRe (MSS) and a separation matrix according to the invention (Inv. Ex). The capacity of each separation matrix relative to initial capacity (determined as load volume to 35% breakthrough DBC) is shown as a function of cycle number.



FIG. 11 shows results from incubation with 1 M NaOH the reference matrix MabSelect SuRe LX (MSS LX) and a separation matrix according to the invention (Inv. Ex). The relative fluorescence as a function of time due to binding of labelled hIgG is shown for each of the separation matrices incubated for set periods of 0, 16 and 32 hours.



FIG. 12 shows results from incubation with 1 M NaOH the reference matrix MabSelect SuRe LX (MSS LX) and a separation matrix according to the invention (Inv. Ex). The relative fluorescence as a function of time due to binding of labelled hIgG is shown for each of the separation matrices incubated for set periods of up to 32 hours.



FIG. 13 shows results from the inactivation of B. subtilis spores using cleaning fluids comprising 1 M NaOH.





DETAILED DESCRIPTION

One aspect of the present invention concerns a method of cleaning and/or sanitizing a separation matrix comprising multimers of immunoglobulin-binding alkali-stabilized Protein A domains covalently coupled to a porous support.


Throughout this detailed description, two separate numbering conventions may be used. Unless otherwise stated, the amino acid residue position numbering convention of FIG. 1 is used, and the position numbers are designated as corresponding to those in SEQ ID NOs: 4-7. This applies also to multimers, where the position numbers designate the positions in the polypeptide units or monomers according to the convention of FIG. 1, unless otherwise stated. However, throughout the claims, summary of invention and on occasion in the detailed description, the position numbers corresponding to those of SEQ ID NOs: 51 and 52 are used. Note that position 1 of SEQ ID NO: 51 or SEQ ID NO: 52 corresponds to position 9 of SEQ ID NOs: 4-7, and in this manner the different numbering conventions may be interconverted.


The immunoglobulin-binding alkali-stabilized Protein A domains of the invention, also termed herein as “the polypeptide”, comprise, consist essentially of, or consist of mutants of a parental Fc-binding domain of Staphylococcus Protein A (SpA), as defined by, or having at least 80% such as at least 90%, 95% or 98% identity to, SEQ ID NO: 51 or SEQ ID NO: 52, wherein the amino acid residues at positions 13 and 44 of SEQ ID NOs: 51 or 52 are asparagines and wherein at least the asparagine residue at position 3 of SEQ ID NOs: 51 or 52 has been mutated to an amino acid selected from the group consisting of glutamic acid, lysine, tyrosine, threonine, phenylalanine, leucine, isoleucine, tryptophan, methionine, valine, alanine, histidine and arginine.









(truncated Zvar)


SEQ ID NO 51


QQ NAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SANLLAEAKK 





LNDAQ





(truncated C domain)


SEQ ID NO 52


QQ NAFYEILHLP NLTEEQRNGF IQSLKDDPSV SKEILAEAKK 





LNDAQ






Such immunoglobulin-binding alkali-stabilized Protein A domains may comprise, consist essentially of, or consist of mutants of a parental Fc-binding domain of Staphylococcus Protein A (SpA), as defined by, or having at least 90%, at least 95% or at least 98% identity to, SEQ ID NO: 1 (E-domain), SEQ ID NO: 2 (D-domain), SEQ ID NO: 3 (A-domain), SEQ ID NO: 22 (variant A-domain), SEQ ID NO: 4 (B-domain), SEQ ID NO: 5 (C-domain), SEQ ID NO: 6 (Protein Z), SEQ ID NO: 7 (Zvar), SEQ ID NO: 51 (Zvar without the linker region amino acids 1-8 and 56-58) or SEQ ID NO: 52 (C-domain without the linker region amino acids 1-8 and 56-58) as illustrated in FIG. 1, wherein at least the asparagine (or serine, in the case of SEQ ID NO: 2) residue at the position corresponding to position 11 in SEQ ID NOs: 4-7 has been mutated to an amino acid selected from the group consisting of glutamic acid, lysine, tyrosine, threonine, phenylalanine, leucine, isoleucine, tryptophan, methionine, valine, alanine, histidine and arginine, and wherein the asparagine residues corresponding to positions 21 and 52 in SEQ ID NOs: 4-7 (positions 13 and 44 of SEQ ID NOs: 51 or 52) are conserved.


A number of the Fc-binding domains listed above are shown aligned in FIG. 1. The parental, i.e. non-engineered, Staphylococcus Protein A (SpA) comprises five Fc-dining domains termed domain E (SEQ ID NO: 1), D (SEQ ID NO: 2), A (SEQ ID NO: 3), B (SEQ ID NO: 4) and C (SEQ ID NO: 5). Protein Z (SEQ ID NO: 6) is a mutated B-domain as disclosed in U.S. Pat. No. 5,143,844, hereby incorporated by reference in its entirety. SEQ ID NO: 7 denotes a further mutated variant of Protein Z, here called Zvar, with the mutations N3A, N6D, N23T. SEQ ID NO: 22 (not shown in FIG. 1) is a natural variant of the A-domain in Protein A from Staphylococcus aureus strain N315, having an A46S mutation, using the position terminology of FIG. 1. SEQ ID NO: 51 is Zvar (SEQ ID NO: 7) without the linker region amino acids at positions 1-8 and 56-58. SEQ ID NO: 52 is the C-domain of protein A without the linker region amino acids 1-8 and 56-58) as illustrated in FIG. 1.


The mutation of N11 (N3 of SEQ ID NOs: 51-52) in these domains, together with the conservation of the asparagine residues N21 and N52 (N13 and N44 of SEQ ID NO: 51-52) confers an improved alkali stability in comparison with the parental domain/polypeptide, without impairing the immunoglobulin-binding properties. Hence, the polypeptide can also be described as an Fc- or immunoglobulin-binding polypeptide, or alternatively as an Fc- or immunoglobulin-binding polypeptide unit.


Described in alternative language, the immunoglobulin-binding alkali-stabilized Protein A domains may comprise, consist essentially of, or consist of a sequence as defined by, or having at least 90%, at least 95% or at least 98% identity to SEQ ID NO: 53.











SEQ ID NO 53



X1Q X2AFYEILX3LP NLTEEQRX4X5F IX6X7LKDX8PSX9







SX10X11X12LAEAKX13 X14NX15AQ






wherein individually of each other:


X1=A, Q or is deleted


X2=E, K, Y, T, F, L, W, I, M, V, A, H or R


X3=H or K


X4=A or N


X5=A, G, S, Y, Q, T, N, F, L, W, I, M, V, D, E, H, R or K, such as S, Y, Q, T, N, F, L, W, I, M, V, D, E, H, R or K


X6=Q or E


X7=S or K


X8=E or D


X9=Q, V or is deleted


X10=K, R, A or is deleted


X11=A, E, N or is deleted


X12=I or L


X13=K or R


X14=L or Y


X15=D, F, Y, W, K or R


Specifically, the amino acid residues in SEQ ID NO: 53 may individually of each other be:


X1=A or is deleted


X2=E


X3=H


X4=N


X6=Q


X7=S


X8=D


X9=V or is deleted


X10=K or is deleted


X11=A or is deleted


X12=I


X13=K


X14=L


In certain embodiments, the amino acid residues in SEQ ID NO: 53 may be:


X1=A, X2=E, X3=H, X4=N, X6=Q, X7=S, X8=D, X9=V, X10=K, X11=A, X12=I, X13=K, X14=L. In some embodiments X2=E, X3=H, X4=N, X5=A, X6=Q, X7=S, X8=D, X12=, X13=K, X14=L and X15=D and one or more of X1, X9, X10 and X11 is deleted. In further embodiments, X1=A, X2=E, X3=H, X4=N, X5=S, Y, Q, T, N, F, L, W, I, M, V, D, E, H, R or K, X6=Q, X7=S, X8=D, X9=V, X10=K, X11=A, X12=I, X13=K, X14=L and X15=D, or alternatively X1=A, X2=E, X3=H, X4=N, X5=A, X6=Q, X7=S, X8=D, X9=V, X10=K, X11=A, X2=I, X13=K, X14=L and X15=F, Y, W, K or R.


In some embodiments, the amino acid residues may individually of each other be:


a) X1=A or is deleted, X2=E, X3=H, X4=N, X6=Q, X7=S, X8=D, X9=V or is deleted, X10=K or is deleted, X1=A or is deleted, X12=I, X13=K, X14=L;


b) X1=A, X2=E, X3=H, X4=N, X5=A, X6=Q, X7=S, X8=D, X9=V, X10=K, X11=A, X2=I, X13=K, X14=L and X15=D;


c) X1 is A, X2=E, X3=H, X4=N, X6=Q, X7=S, X8=D, X9=V, X10=K, X11=A, X2=I, X13=K, X14=L and X15=D; or


d) X1 is A, X3=H, X4=N, X5=A, X6=Q, X7=S, X8=D, X9=V, X10=K, X11=A, X2=I, X13=K, X14=L and X15=D.


The N11 (X2) mutation (e.g. a N11E or N11K mutation) may be the only mutation or the polypeptide may also comprise further mutations, such as substitutions in at least one of the positions corresponding to positions 3, 6, 9, 10, 15, 18, 23, 28, 29, 32, 33, 36, 37, 40, 42, 43, 44, 47, 50, 51, 55 and 57 in SEQ ID NOs: 4-7. In one or more of these positions, the original amino acid residue may e.g. be substituted with an amino acid which is not asparagine, proline or cysteine. The original amino acid residue may e.g. be substituted with an alanine, a valine, a threonine, a serine, a lysine, a glutamic acid or an aspartic acid. Further, one or more amino acid residues may be deleted, e.g. from positions 1-6 and/or from positions 56-58.


In some embodiments, the amino acid residue at the position corresponding to position 9 in SEQ ID NOs: 4-7 (X1) is an amino acid other than glutamine, asparagine, proline or cysteine, such as an alanine or it can be deleted. The combination of the mutations at positions 9 and 11 provides particularly good alkali stability, as shown by the examples. In specific embodiments, in SEQ ID NO: 7 the amino acid residue at position 9 is an alanine and the amino acid residue at position 11 is a lysine or glutamic acid, such as a lysine. Mutations at position 9 are also discussed in copending application PCT/SE2014/050872, which is hereby incorporated by reference in its entirety.


In some embodiments, the amino acid residue at the position corresponding to position 50 in SEQ ID NOs: 4-7 (X13) is an arginine or a glutamic acid.


In certain embodiments, the amino acid residue at the position corresponding to position 3 in SEQ ID NOs: 4-7 is an alanine and/or the amino acid residue at the position corresponding to position 6 in SEQ ID NOs: 4-7 is an aspartic acid. One of the amino acid residues at positions 3 and 6 may be an asparagine and in an alternative embodiment both amino acid residues at positions 3 and 6 may be asparagines.


In some embodiments the amino acid residue at the position corresponding to position 43 in SEQ ID NOs: 4-7 (X11) is an alanine or a glutamic acid, such as an alanine or it can be deleted. In specific embodiments, the amino acid residues at positions 9 and 11 in SEQ ID NO: 7 are alanine and lysine/glutamic acid respectively, while the amino acid residue at position 43 is alanine or glutamic acid.


In certain embodiments the amino acid residue at the position corresponding to position 28 in SEQ ID NOs: 4-7 (X5) is an alanine or an asparagine, such as an alanine.


In some embodiments the amino acid residue at the position corresponding to position 40 in SEQ ID NOs: 4-7 (X9) is selected from the group consisting of asparagine, alanine, glutamic acid and valine, or from the group consisting of glutamic acid and valine, or valine, or it can be deleted. In specific embodiments, the amino acid residues at positions 9 and 11 in SEQ ID NO: 7 are alanine and glutamic acid respectively, while the amino acid residue at position 40 is valine. Optionally, the amino acid residue at position 43 may then be alanine or glutamic acid.


In certain embodiments, the amino acid residue at the position corresponding to position 42 in SEQ ID NOs: 4-7 (X10) is an alanine, lysine or arginine or it can be deleted.


In some embodiments the amino acid residue at the position corresponding to position 18 in SEQ ID NOs: 4-7 (X3) is a lysine or a histidine, such as a lysine.


In certain embodiments the amino acid residue at the position corresponding to position 33 in SEQ ID NOs: 4-7 (X7) is a lysine or a serine, such as a lysine.


In some embodiments the amino acid residue at the position corresponding to position 37 in SEQ ID NOs: 4-7 (X8) is a glutamic acid or an aspartic acid, such as a glutamic acid.


In certain embodiments the amino acid residue at the position corresponding to position 51 in SEQ ID NOs: 4-7 (X14) is a tyrosine or a leucine, such as a tyrosine.


In some embodiments, the amino acid residue at the position corresponding to position 44 in SEQ ID NOs: 4-7 (X12) is a leucine or an isoleucine. In specific embodiments, the amino acid residues at positions 9 and 11 in SEQ ID NO: 7 are alanine and lysine/glutamic acid respectively, while the amino acid residue at position 44 is isoleucine. Optionally, the amino acid residue at position 43 may then be alanine or glutamic acid.


In some embodiments, the amino acid residues at the positions corresponding to positions 1, 2, 3 and 4 or to positions 3, 4, 5 and 6 in SEQ ID NO: 4-7 have been deleted. In specific variants of these embodiments, the parental polypeptide is the C domain of Protein A (SEQ ID NO: 5). The effects of these deletions on the native C domain are described in U.S. Pat. Nos. 9,018,305 and 8,329,860, which are hereby incorporated by reference in their entireties.


In certain embodiments, the mutation in SEQ ID Nos: 4-7, such as in SEQ ID NO: 7, is selected from the group consisting of: N11K; N11E; N11Y; N11T; N11F; N11L; N11W; N11I; N11M; N11V; N11A; N11H; N11R; N11E, Q32A; N11E, Q32E, Q40E; N11E, Q32E, K50R; Q9A, N11E, N43A; Q9A, N11E, N28A, N43A; Q9A, N11E, Q40V, A42K, N43E, L44I; Q9A, N11E, Q40V, A42K, N43A, L44I; N11K, H18K, S33K, D37E, A42R, N43A, L44I, K50R, L51Y; Q9A, N11E, N28A, Q40V, A42K, N43A, L44I; Q9A, N11K, H18K, S33K, D37E, A42R, N43A, L44I, K50R, L51Y; N11K, H18K, D37E, A42R, N43A, L44I; Q9A, N11K, H18K, D37E, A42R, N43A, L44I; Q9A, N11K, H18K, D37E, A42R, N43A, L44I, K50R; Q9A, N11K, H18K, D37E, A42R; Q9A, N11E, D37E, Q40V, A42K, N43A, L44I and Q9A, N11E, D37E, Q40V, A42R, N43A, L44I. These mutations provide particularly high alkaline stabilities. The mutation in SEQ ID NOs: 4-7, such as in SEQ ID NO: 7, can also be selected from the group consisting of N11K; N11Y; N11F; N11L; N11W; N11I; N11M; N11V; N11A; N11H; N11R; Q9A, N11E, N43A; Q9A, N11E, N28A, N43A; Q9A, N11E, Q40V, A42K, N43E, L44I; Q9A, N11E, Q40V, A42K, N43A, L44I; Q9A, N11E, N28A, Q40V, A42K, N43A, L44I; N11K, H18K, S33K, D37E, A42R, N43A, L44I, K50R, L51Y; Q9A, N11K, H18K, S33K, D37E, A42R, N43A, L44I, K50R, L51Y; N11K, H18K, D37E, A42R, N43A, L44I; Q9A, N11K, H18K, D37E, A42R, N43A, L44I and Q9A, N11K, H18K, D37E, A42R, N43A, L44I, K50R.


In some embodiments, the polypeptide comprises or consists essentially of a sequence defined by or having at least 90%, 95% or 98% identity to an amino acid sequence selected from the group consisting of: SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49 and SEQ ID NO: 50. It may e.g. comprise or consist essentially of a sequence defined by or having at least 90%, 95% or 98% identity to an amino acid sequence selected from the group consisting of: SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 16, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28 and SEQ ID NO: 29. It can also comprise or consist essentially of a sequence defined by or having at least 90%, 95% or 98% identity to an amino acid sequence selected from the group consisting of: SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 16, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 38, SEQ ID NO: 40; SEQ ID NO: 41; SEQ ID NO: 42; SEQ NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47 and SEQ ID NO: 48.


In certain embodiments, the polypeptide comprises or consists essentially of a sequence defined by or having at least 90%, 95% or 98% identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 54-70; comprises or consists essentially of a sequence defined by or having at least 90%, 95% or 98% identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 71-75; or it may comprise or consist essentially of a sequence defined by or having at least 90%, 95% or 98% identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 76-79. It may further comprise or consist essentially of a sequence defined by or having at least 90%, 95% or 98% identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 89-95.


The polypeptide may e.g. be defined by a sequence selected from the groups above or from subsets of these groups, but it may also comprise additional amino acid residues at the N- and/or C-terminal end, e.g. a leader sequence at the N-terminal end and/or a tail sequence at the C-terminal end.










Zvar(Q9A,N11E,N43A)



SEQ ID NO 8



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SAALLAEAKK LNDAQAPK






Zvar(Q9A,N11E,N28A,N43A)


SEQ ID NO 9



VDAKFDKEAQ EAFYEILHLP NLTEEQRAAF IQSLKDDPSQ SAALLAEAKK LNDAQAPK






Zvar(Q9A,N11E,Q40V,A42K,N43E,L44I)


SEQ ID NO 10



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKEILAEAKK LNDAQAPK






Zvar(Q9A,N11E,Q40V,A42K,N43A,L44I)


SEQ ID NO 11



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(N11E,Q32A)


SEQ ID NO 12



VDAKFDKEQQ EAFYEILHLP NLTEEQRNAF IASLKDDPSQ SANLLAEAKK LNDAQAPK






Zvar(N11E)


SEQ ID NO 13



VDAKFDKEQQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SANLLAEAKK LNDAQAPK






Zvar(N11E,Q32E,Q40E)


SEQ ID NO 14



VDAKFDKEQQ EAFYEILHLP NLTEEQRNAF IESLKDDPSE SANLLAEAKK LNDAQAPK






Zvar(N11E,Q32E,K50R)


SEQ ID NO 15



VDAKFDKEQQ EAFYEILHLP NLTEEQRNAF IESLKDDPSQ SANLLAEAKR LNDAQAPK






Zvar(N11K)


SEQ ID NO 16



VDAKFDKEQQ KAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SANLLAEAKK LNDAQAPK






Zvar(N11K,H18K,533K,D37E,A42R,N43A,L44I,K5OR,L51Y)


SEQ ID NO 23



VDAKFDKEQQ KAFYEILKLP NLTEEQRNAF IQKLKDEPSQ SRAILAEAKR YNDAQAPK






Zvar(Q9A,N11E,N28A,Q40V,A42K,N43A,L44I)


SEQ ID NO 24



VDAKFDKEAQ EAFYEILHLP NLTEEQRAAF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11K,H18K,S33K,D37E,A42R,N43A,L44I,K5OR,L51Y)


SEQ ID NO 25



VDAKFDKEAQ KAFYEILKLP NLTEEQRAAF IQKLKDEPSQ SRAILAEAKR YNDAQAPK






Zvar(N11K, H18K, D37E, A42R, N43A, L44I)


SEQ ID NO 26



VDAKFDKEQQ KAFYEILKLP NLTEEQRNAF IQSLKDEPSQ SRAILAEAKK LNDAQAPK






Zvar(Q9A, N11K, H18K, D37E, A42R, N43A, L44I)


SEQ ID NO 27



VDAKFDKEAQ KAFYEILKLP NLTEEQRNAF IQSLKDEPSQ SRAILAEAKK LNDAQAPK






Zvar(Q9A, N11K, H18K, D37E, A42R, N43A, L44I, K50R)


SEQ ID NO 28



VDAKFDKEAQ KAFYEILKLP NLTEEQRNAF IQSLKDEPSQ SRAILAEAKR LNDAQAPK






Zvar(Q9A,N11K,H18K,D37E,A42R)


SEQ ID NO 29



VDAKFDKEAQ KAFYEILKLP NLTEEQRNAF IQSLKDEPSQ SRNLLAEAKK LNDAQAPK






B(Q9A,N11E,Q40V,A42K,N43A,L44I)


SEQ ID NO 36



ADNKFNKEAQ EAFYEILHLP NLNEEQRNGF IQSLKDDPSV SKAILAEAKK LNDAQAPK






C(Q9A,N11E,E43A)


SEQ ID NO 37



ADNKFNKEAQ EAFYEILHLP NLTEEQRNGF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(N11Y)


SEQ ID NO 38



VDAKFDKEQQ YAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SANLLAEAKK LNDAQAPK






Zvar(N11T)


SEQ ID NO 39



VDAKFDKEQQ TAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SAN LLAEAKK LNDAQAPK






Zvar(N11F)


SEQ ID NO 40



VDAKFDKEQQ FAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SAN LLAEAKK LNDAQAPK






Zvar(N11L)


SEQ ID NO 41



VDAKFDKEQQ LAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SAN LLAEAKK LNDAQAPK









Zvar(N11W)


SEQ ID NO 42



VDAKFDKEQQ WAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SANLLAEAKK LNDAQAPK






Zvar(N111)


SEQ ID NO 43



VDAKFDKEQQ IAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SANLLAEAKK LNDAQAPK






Zvar(N11M)


SEQ ID NO 44



VDAKFDKEQQ MAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SANLLAEAKK LNDAQAPK






Zvar(N11V)


SEQ ID NO 45



VDAKFDKEQQ VAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SANLLAEAKK LNDAQAPK






Zvar(N11A)


SEQ ID NO 46



VDAKFDKEQQ AAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SANLLAEAKK LNDAQAPK






Zvar(N11H)


SEQ ID NO 47



VDAKFDKEQQ HAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SANLLAEAKK LNDAQAPK






Zvar(N11R)


SEQ ID NO 48



VDAKFDKEQQ RAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SANLLAEAKK LNDAQAPK






Zvar(Q9A,N11E,D37E,Q40V,A42K,N43A,L44I)


SEQ ID NO 49



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDEPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E,D37E,Q40V,A42R,N43A,L44I)


SEQ ID NO 50



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDEPSV SRAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29G,Q40V,A42K,N43A,L44I)


SEQ ID NO 54



VDAKFDKEAQ EAFYEILHLP NLTEEQRNGF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A295,Q40V,A42K,N43A,L44I)


SEQ ID NO 55



VDAKFDKEAQ EAFYEILHLP NLTEEQRNSF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29Y,Q40V,A42K,N43A,L44I)


SEQ ID NO 56



VDAKFDKEAQ EAFYEILHLP NLTEEQRNYF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29Q,Q40V,A42K,N43A,L44I)


SEQ ID NO 57



VDAKFDKEAQ EAFYEILHLP NLTEEQRNQF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29T,Q40V,A42K,N43A,L44I)


SEQ ID NO 58



VDAKFDKEAQ EAFYEILHLP NLTEEQRNTF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29N,Q40V,A42K,N43A,L44I)


SEQ ID NO 59



VDAKFDKEAQ EAFYEILHLP NLTEEQRNNF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29F,Q40V,A42K,N43A,L44I)


SEQ ID NO 60



VDAKFDKEAQ EAFYEILHLP NLTEEQRNFF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29L,Q40V,A42K,N43A,L44I)


SEQ ID NO 61



VDAKFDKEAQ EAFYEILHLP NLTEEQRNLF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29W,Q40V,A42K,N43A,L44I)


SEQ ID NO 62



VDAKFDKEAQ EAFYEILHLP NLTEEQRNWF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A291,Q40V,A42K,N43A,L44I)


SEQ ID NO 63



VDAKFDKEAQ EAFYEILHLP NLTEEQRNIF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29M,Q40V,A42K,N43A,L44I)


SEQ ID NO 64



VDAKFDKEAQ EAFYEILHLP NLTEEQRNMF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29V,Q40V,A42K,N43A,L44I)


SEQ ID NO 65



VDAKFDKEAQ EAFYEILHLP NLTEEQRNVF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29D,Q40V,A42K,N43A,L44I)


SEQ ID NO 66



VDAKFDKEAQ EAFYEILHLP NLTEEQRNDF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29E,Q40V,A42K,N43A,L44I)


SEQ ID NO 67



VDAKFDKEAQ EAFYEILHLP NLTEEQRNEF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29H,Q40V,A42K,N43A,L44I)


SEQ ID NO 68



VDAKFDKEAQ EAFYEILHLP NLTEEQRNHF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29R,Q40V,A42K,N43A,L44I)


SEQ ID NO 69



VDAKFDKEAQ EAFYEILHLP NLTEEQRNRF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, A29K,Q40V,A42K,N43A,L44I)


SEQ ID NO 70



VDAKFDKEAQ EAFYEILHLP NLTEEQRNKF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, Q40V,A42K,N43A,L44I,D53F)


SEQ ID NO 71



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNFAQAPK






Zvar(Q9A,N11E, Q40V,A42K,N43A,L44I,D53Y)


SEQ ID NO 72



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNYAQAPK






Zvar(Q9A,N11E, Q40V,A42K,N43A,L44I,D53W)


SEQ ID NO 73



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNWAQAPK






Zvar(Q9A,N11E, Q40V,A42K,N43A,L44I,D53K)


SEQ ID NO 74



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNKAQAPK






Zvar(Q9A,N11E, Q40V,A42K,N43A,L44I,D53R)


SEQ ID NO 75



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNRAQAPK






Zvar(Q9del,N11E, Q40V,A42K,N43A,L44I)


SEQ ID NO 76



VDAKFDKE_Q EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, Q40del,A42K,N43A,L44I)


SEQ ID NO 77



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPS_ SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E, Q40V,A42del,N43A,L44I)


SEQ ID NO 78



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV S_AILAEAKK LNDAQAPK






Zvar(Q9A,N11E, Q40V,A42K,N43del,L44I)


SEQ ID NO 79



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SK_ILAEAKK LNDAQAPK






Zvar(D2del,A3del,K4del,Q9A,N11E,Q40V,A42K,N43A,L44I)


SEQ ID NO 89



V_FDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(V1del,D2del,Q9A,N11E,Q40V,A42K,N43A,L44I,K58del)


SEQ ID NO 90



_AKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAP_






Zvar(K4del,F5del,D6del,K7del,E8del,Q9A,N11E,Q40V,A42K,N43A,L44I)


SEQ ID NO 91



VDA_ AQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E,Q40V,A42K,N43A,L44I,A56del,P57del,K58del)


SEQ ID NO 92



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQ






Zvar(V1del,,D2del,A3del,Q9A,N11E,Q40V,A42K,N43A,L44I)


SEQ ID NO 93



_KFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(V1del,D2del,A3del,K4del,F5del,D6del,K7del,E8del,Q9A,N11E,Q40V,A42K,N43A,L44I)


SEQ ID NO 94



_AQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK






Zvar(Q9A,N11E,Q40V,A42K,N43A,L44I,K58_insYEDG)


SEQ ID NO 95



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKYE DG







The separation matrix comprises multimers of the immunoglobulin-binding alkali-stabilized Protein A domains. Such multimers comprise, consist essentially of, or consist of a plurality of immunoglobulin-binding alkali-stabilized Protein A domains (polypeptide units) as defined by any embodiment disclosed above. The use of multimers may increase the immunoglobulin binding capacity and multimers may also have a higher alkali stability than monomers. The multimer can e.g. be a dimer, a trimer, a tetramer, a pentamer, a hexamer, a heptamer, an octamer or a nonamer. The multimer may be a homomultimer, where all the units in the multimer are identical or it can be a heteromultimer, where at least one unit differs from the others. Advantageously, all the units in the multimer are alkali stable, such as by comprising the mutations/conservations disclosed above. The polypeptides can be linked to each other directly by peptide bonds between the C-terminal and N-terminal ends of the polypeptides. Alternatively, two or more units in the multimer can be linked by linkers comprising oligomeric or polymeric species, such as linkers comprising peptides with up to 25 or 30 amino acids, such as 3-25 or 3-20 amino acids. The linkers may e.g. comprise or consist essentially of a peptide sequence defined by, or having at least 90% identity or at least 95% identity, with an amino acid sequence selected from the group consisting of APKVDAKFDKE (SEQ ID NO: 96), APKVDNKFNKE (SEQ ID NO: 97), APKADNKFNKE (SEQ ID NO: 98), APKVFDKE (SEQ ID NO: 99), APAKFDKE (SEQ ID NO: 100), AKFDKE (SEQ ID NO: 101), APKVDA (SEQ ID NO: 102), VDAKFDKE (SEQ ID NO: 103), APKKFDKE (SEQ ID NO: 104), APK, APKYEDGVDAKFDKE (SEQ ID NO: 105) and YEDG (SEQ ID NO: 106) or alternatively selected from the group consisting of APKADNKFNKE (SEQ ID NO: 98), APKVFDKE (SEQ ID NO: 99), APAKFDKE (SEQ ID NO: 100), AKFDKE (SEQ ID NO: 101), APKVDA (SEQ ID NO: 102), VDAKFDKE (SEQ ID NO: 103), APKKFDKE (SEQ ID NO: 104), APKYEDGVDAKFDKE (SEQ ID NO: 105) and YEDG (SEQ ID NO: 106). They can also consist essentially of a peptide sequence defined by or having at least 90% identity or at least 95% identity with an amino acid sequence selected from the group consisting of APKADNKFNKE (SEQ ID NO: 98), APKVFDKE (SEQ ID NO: 99), APAKFDKE (SEQ ID NO: 100), AKFDKE (SEQ ID NO: 101), APKVDA (SEQ ID NO: 102), VDAKFDKE (SEQ ID NO: 103), APKKFDKE (SEQ ID NO: 104), APK and APKYEDGVDAKFDKE (SEQ ID NO: 105). In some embodiments the linkers do not consist of the peptides APKVDAKFDKE (SEQ ID NO: 96) or APKVDNKFNKE (SEQ ID NO: 97), or alternatively do not consist of the peptides APKVDAKFDKE (SEQ ID NO: 96), APKVDNKFNKE (SEQ ID NO: 97), APKFNKE (SEQ ID NO: 107), APKFDKE (SEQ ID NO: 108), APKVDKE (SEQ ID NO: 109) or APKADKE (SEQ ID NO: 110).


The nature of such a linker should preferably not destabilize the spatial conformation of the protein units. This can e.g. be achieved by avoiding the presence of proline in the linkers. Furthermore, said linker should preferably also be sufficiently stable in alkaline environments not to impair the properties of the mutated protein units. For this purpose, it is advantageous if the linkers do not contain asparagine. It can additionally be advantageous if the linkers do not contain glutamine. The multimer may further at the N-terminal end comprise a plurality of amino acid residues e.g. originating from the cloning process or constituting a residue from a cleaved off signaling sequence. The number of additional amino acid residues may e.g. be 20 or less, such as 15 or less, such as 10 or less or 5 or less. As a specific example, the multimer may comprise an AQ, AQGT (SEQ ID NO: 111), VDAKFDKE (SEQ ID NO: 103), AQVDAKFDKE (SEQ ID NO: 112) or AQGTVDAKFDKE (SEQ ID NO: 113) sequence at the N-terminal end.


In certain embodiments, the multimer may comprise, or consist essentially, of a sequence selected from the group consisting of: SEQ ID Nos: 80-87. These and additional sequences are listed below and named as Parent(Mutations)n, where n is the number of monomer units in a multimer.










Zvar(Q9A,N11E,N43A)4



SEQ ID NO 17



AQGT VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SAALLAEAKK LNDAQAPK






VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SAALLAEAKK LNDAQAPK VDAKFDKEAQ





EAFYEILHLP NLTEEQRNAF IQSLKDDPSQ SAALLAEAKK LNDAQAPK VDAKFDKEAQ EAFYEILHLP





NLTEEQRNAF IQSLKDDPSQ SAALLAEAKK LNDAQAPKC





Zvar(Q9A,N11E,N28A,N43A)4


SEQ ID NO 18



AQGT VDAKFDKEAQ EAFYEILHLP NLTEEQRAAF IQSLKDDPSQ SAALLAEAKK LNDAQAPK






VDAKFDKEAQ EAFYEILHLP NLTEEQRAAF IQSLKDDPSQ SAALLAEAKK LNDAQAPK VDAKFDKEAQ





EAFYEILHLP NLTEEQRAAF IQSLKDDPSQ SAALLAEAKK LNDAQAPK VDAKFDKEAQ EAFYEILHLP





NLTEEQRAAF IQSLKDDPSQ SAALLAEAKK LNDAQAPKC





Zvar(Q9A,N11E,Q40V,A42K,N43E,L441)4


SEQ ID NO 19



AQGT VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKEILAEAKK LNDAQAPK






VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKEILAEAKK LNDAQAPK VDAKFDKEAQ





EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKEILAEAKK LNDAQAPK VDAKFDKEAQ EAFYEILHLP





NLTEEQRNAF IQSLKDDPSV SKEILAEAKK LNDAQAPKC





Zvar(Q9A,N11E,Q40V,A42K,N43A,L441)4


SEQ ID NO 20



AQGT VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK






VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK VDAKFDKEAQ





EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK VDAKFDKEAQ EAFYEILHLP





NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKC





Zvar(N11K,H18K,S33K,D37E,A42R,N43A,L441,K5OR,L51Y)4


SEQ ID NO 30



AQGT VDAKFDKEQQ KAFYEILKLP NLTEEQRNAF IQKLKDEPSQ SRAILAEAKR YNDAQAPK






VDAKFDKEQQ KAFYEILKLP NLTEEQRNAF IQKLKDEPSQ SRAILAEAKR YNDAQAPK VDAKFDKEQQ





KAFYEILKLP NLTEEQRNAF IQKLKDEPSQ SRAILAEAKR YNDAQAPK VDAKFDKEQQ KAFYEILKLP





NLTEEQRNAF IQKLKDEPSQ SRAILAEAKR YNDAQAPKC





Zvar(Q9A,N11K,H18K,D37E,A42R)4


SEQ ID NO 31



AQGT VDAKFDKEAQ KAFYEILKLP NLTEEQRNAF IQSLKDEPSQ SRN LLAEAKK LNDAQAPK






VDAKFDKEAQ KAFYEILKLP NLTEEQRNAF IQSLKDEPSQ SRN LLAEAKK LNDAQAPK VDAKFDKEAQ





KAFYEILKLP NLTEEQRNAF IQSLKDEPSQ SRNLLAEAKK LNDAQAPK VDAKFDKEAQ KAFYEILKLP





NLTEEQRNAF IQSLKDEPSQ SRN LLAEAKK LNDAQAPKC





Zvar(Q9A,N11E,N28A,Q40V,A42K,N43A,L441)4


SEQ ID NO 32



AQGT VDAKFDKEAQ EAFYEILHLP NLTEEQRAAF IQSLKDDPSV SKAILAEAKK LNDAQAPK






VDAKFDKEAQ EAFYEILHLP NLTEEQRAAF IQSLKDDPSV SKAILAEAKK LNDAQAPK VDAKFDKEAQ





EAFYEILHLP NLTEEQRAAF IQSLKDDPSV SKAILAEAKK LNDAQAPK VDAKFDKEAQ EAFYEILHLP





NLTEEQRAAF IQSLKDDPSV SKAILAEAKK LNDAQAPKC





Zvar(Q9A,N11E,Q40V,A42K,N43A,L441)6


SEQ ID NO 33



AQGT VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK






VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK VDAKFDKEAQ





EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK VDAKFDKEAQ EAFYEILHLP





NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF





IQSLKDDPSV SKAILAEAKK LNDAQAPK VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV





SKAILAEAKK LNDAQAPKC





Zvar(Q9A,N11E,D37E,Q40V,A42K,N43A,L441)4


SEQ ID NO 34



AQGT VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDEPSV SKAILAEAKK LNDAQAPK






VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDEPSV SKAILAEAKK LNDAQAPK VDAKFDKEAQ






EAFYEILHLP NLTEEQRNAF IQSLKDEPSV SKAILAEAKK LNDAQAPK VDAKFDKEAQ EAFYEILHLP






NLTEEQRNAF IQSLKDEPSV SKAILAEAKK LNDAQAPKC





Zvar(Q9A,N11E,D37E,Q40V,A42R,N43A,L441)4


SEQ ID NO 35



AQGT VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDEPSV SRAILAEAKK LNDAQAPK






VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDEPSV SRAILAEAKK LNDAQAPK VDAKFDKEAQ






EAFYEILHLP NLTEEQRNAF IQSLKDEPSV SRAILAEAKK LNDAQAPK VDAKFDKEAQ EAFYEILHLP






NLTEEQRNAF IQSLKDEPSV SRAILAEAKK LNDAQAPKC





Zvar(Q9A,N11E,Q40V,A42K,N43A,L441)2 with D2, A3 and K4 in linker deleted


SEQ ID NO 80



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKVFDKEAQ






EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKC





Zvar(Q9A,N11E,Q40V,A42K,N43A,L441)2 with K58, V1 and D2 in linker deleted


SEQ ID NO 81



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAP AKFDKEAQ



EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKC


Zvar(Q9A,N11E,Q40V,A42K,N43A,L441)2 with P57, K58, V1, D2 and A3 in linker


deleted


SEQ ID NO 82



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPAKFDKEAQ






EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKC





Zvar(Q9A,N11E,Q40V,A42K,N43A,L441)2 with K4, F5, D6, K7 and E8 in linker


deleted


SEQ ID NO 83



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKVDAAQ






EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKC





Zvar(Q9A,N11E,Q40V,A42K,N43A,L441)2 with A56, P57 and K58 in linker deleted


SEQ ID NO 84



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQVDAKFDKEAQ






EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKC





Zvar(Q9A,N11E,Q40V,A42K,N43A,L441)2 with V1, D2 and A3 in linker deleted


SEQ ID NO 85



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKKFDKEAQ






EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKC





Zvar(Q9A,N11E,Q40V,A42K,N43A,L441)2 with V1, D2, A3, K4, F5, D6, K7 and E8 in


linker deleted


SEQ ID NO 86



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK AQ






EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKC





Zvar(Q9A,N11E,Q40V,A42K,N43A,L441)2 with YEDG inserted in linker between


K58 and V1


SEQ ID NO 87



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKYEDG







VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKC






Zvar2


SEQ ID NO 88 



VDAKFDKEAQ EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPK VDAKFDKEAQ






EAFYEILHLP NLTEEQRNAF IQSLKDDPSV SKAILAEAKK LNDAQAPKC






In some embodiments, the polypeptide and/or multimer, as disclosed above, further comprises at the C-terminal or N-terminal end one or more coupling elements, selected from the group consisting of one or more cysteine residues, a plurality of lysine residues and a plurality of histidine residues. The coupling element(s) may also be located within 1-5 amino acid residues, such as within 1-3 or 1-2 amino acid residues from the C-terminal or N-terminal end. The coupling element may e.g. be a single cysteine at the C-terminal end. The coupling element(s) may be directly linked to the C- or N-terminal end, or it/they may be linked via a stretch comprising up to 15 amino acids, such as 1-5, 1-10 or 5-10 amino acids. This stretch should preferably also be sufficiently stable in alkaline environments not to impair the properties of the mutated protein. For this purpose, it is advantageous if the stretch does not contain asparagine. It can additionally be advantageous if the stretch does not contain glutamine. An advantage of having a C-terminal cysteine is that endpoint coupling of the protein can be achieved through reaction of the cysteine thiol with an electrophilic group on a support. This provides excellent mobility of the coupled protein which is important for the binding capacity.


The alkali stability of the polypeptide or multimer can be assessed by coupling it to a surface plasmon resonance (SPR) chip, e.g. to Biacore CM5 sensor chips as described in the examples, using e.g. NHS- or maleimide coupling chemistries, and measuring the immunoglobulin-binding capacity of the chip, typically using polyclonal human IgG, before and after incubation in alkaline solutions at a specified temperature, e.g. 22+/−2° C. The incubation can e.g. be performed in 0.5 M NaOH for a number of 10 min cycles, such as 100, 200 or 300 cycles. The IgG capacity of the matrix after 100 10 min incubation cycles in 0.5 M NaOH at 22+/−2° C. can be at least 55, such as at least 60, at least 80 or at least 90% of the IgG capacity before the incubation. Alternatively, the remaining IgG capacity after 100 cycles for a particular mutant measured as above can be compared with the remaining IgG capacity for the parental polypeptide/multimer. In this case, the remaining IgG capacity for the mutant may be at least 105%, such as at least 110%, at least 125%, at least 150% or at least 200% of the parental polypeptide/multimer.


The immunoglobulin-binding alkali-stabilized Protein A domains and/or multimers thereof may be encoded by a nucleic acid sequence, such as an RNA sequence or a DNA sequence encoding the polypeptide or multimer. A vector, such as a plasmid, which in addition to the coding sequence comprises the required signal sequences, may be used for expression of the polypeptide or multimer. The vector may comprise nucleic acid encoding a multimer as described above, wherein the separate nucleic acids encoding each unit may have homologous or heterologous DNA sequences.


An expression system, which comprises a nucleic acid or a vector as disclosed above, may be used for expression of the polypeptide or multimer. The expression system may e.g. be a gram-positive or gram-negative prokaryotic host cell system, e.g. E. coli or Bacillus sp. which has been modified to express the present polypeptide or multimer. Alternatively, the expression system may be a eukaryotic host cell system, such as a yeast, e.g. Pichia pastoris or Saccharomyces cerevisiae, or mammalian cells, e.g. CHO cells.


The separation matrix comprises, consists essentially of, or consists of multimers of immunoglobulin-binding alkali-stabilized Protein A domains as described above, covalently coupled to a porous support.


The separation matrix may comprise at least 11, such as 11-21, 15-21 or 15-18 mg/ml Fc-binding ligands covalently coupled to a porous support, wherein:


a) the ligands comprise multimers of alkali-stabilized Protein A domains,


b) the porous support comprises cross-linked polymer particles having a volume-weighted median diameter (d50,v) of 56-70, such as 56-66, micrometers and a dry solids weight of 55-80, such as 60-78 or 65-78, mg/ml. The cross-linked polymer particles may further have a pore size corresponding to an inverse gel filtration chromatography Kd value of 0.69-0.85, such as 0.70-0.85 or 0.69-0.80, for dextran of Mw 110 kDa. The multimers may e.g. comprise tetramers, pentamers, hexamers or heptamers of alkali-stabilized Protein A domains, such as hexamers of alkali-stabilized Protein A domains. The combination of the high ligand contents with the particle size range, the dry solids weight range and the optional Kd range provides for a high binding capacity, e.g. such that the 10% breakthrough dynamic binding capacity for IgG is at least 45 mg/ml, such as at least 50 or at least 55 mg/ml at 2.4 min residence time. Alternatively, or additionally, the 10% breakthrough dynamic binding capacity for IgG may be at least 60 mg/ml, such as at least 65, at least 70 or at least 75 mg/ml at 6 min residence time.


The alkali-stabilized Protein A domain multimers are highly selective for IgG and the separation matrix can suitably have a dissociation constant for human IgG2 of below 0.2 mg/ml, such as below 0.1 mg/ml, in 20 mM phosphate buffer, 180 mM NaCl, pH 7.5. This can be determined according to the adsorption isotherm method described in N Pakiman et al: J Appl Sci 12, 1136-1141 (2012).


In certain embodiments the separation matrix comprises at least 15, such as 15-21 or 15-18 mg/ml Fc-binding ligands covalently coupled to a porous support, wherein the ligands comprise multimers of alkali-stabilized Protein A domains. These multimers can suitably be as disclosed in any of the embodiments described above or as specified below.


In some embodiments the separation matrix comprises 5-25, such as 5-20 mg/ml, 5-15 mg/ml, 5-11 mg/ml or 6-11 mg/ml of the polypeptide or multimer coupled to the support. The amount of coupled polypeptide/multimer can be controlled by the concentration of polypeptide/multimer used in the coupling process, by the activation and coupling conditions used and/or by the pore structure of the support used. As a general rule the absolute binding capacity of the matrix increases with the amount of coupled polypeptide/multimer, at least up to a point where the pores become significantly constricted by the coupled polypeptide/multimer. Without being bound by theory, it appears though that for the Kd values recited for the support, the constriction of the pores by coupled ligand is of lower significance. The relative binding capacity per mg coupled polypeptide/multimer will decrease at high coupling levels, resulting in a cost-benefit optimum within the ranges specified above.


Such a separation matrix is useful for separation of immunoglobulins or other Fc-containing proteins and, due to the improved alkali stability of the polypeptides/multimers, the matrix will withstand highly alkaline conditions during cleaning, which is essential for long-term repeated use in a bioprocess separation setting. The alkali stability of the matrix can be assessed by measuring the immunoglobulin-binding capacity, typically using polyclonal human IgG, before and after incubation in alkaline solutions at a specified temperature, e.g. 22+/−2° C. The incubation can e.g. be performed in 0.5 M or 1.0 M NaOH for a number of 15 min cycles, such as 100, 200 or 300 cycles, corresponding to a total incubation time of 25, 50 or 75 h. The IgG capacity of the matrix after 96-100 15 min incubation cycles or a total incubation time of 24 or 25 h in 0.5 M NaOH at 22+/−2° C. can be at least 80, such as at least 85, at least 90 or at least 95% of the IgG capacity before the incubation. The capacity of the matrix after a total incubation time of 24 h in 1.0 M NaOH at 22+/−2° C. can be at least 70, such as at least 80 or at least 90% of the IgG capacity before the incubation. The 10% breakthrough dynamic binding capacity (Qb10%) for IgG at 2.4 min or 6 min residence time may e.g. be reduced by less than 20% after incubation 31 h in 1.0 M aqueous NaOH at 22+/−2 C.


As the skilled person will understand, the expressed polypeptide or multimer should be purified to an appropriate extent before being immobilized to a support. Such purification methods are well known in the field, and the immobilization of protein-based ligands to supports is easily carried out using standard methods. Suitable methods and supports will be discussed below in more detail.


The porous support of the separation matrix may be of any suitable well-known kind. A conventional affinity separation matrix is often of organic nature and based on polymers that expose a hydrophilic surface to the aqueous media used, i.e. expose hydroxy (—OH), carboxy (—COOH), carboxamido (—CONH2, possibly in N-substituted forms), amino (—NH2, possibly in substituted form), oligo- or polyethylenoxy groups on their external and, if present, also on internal surfaces. The porosity of the support can be expressed as a Kav or Kd value (the fraction of the pore volume available to a probe molecule of a particular size) measured by inverse size exclusion chromatography, e.g. according to the methods described in Gel Filtration Principles and Methods, Pharmacia LKB Biotechnology 1991, pp 6-13. Kav is determined as the ratio (Ve−V0)/(Vt−V0), where Ve is the elution volume of a probe molecule (e.g. Dextran 110 kD), V0 is the void volume of the column (e.g. the elution volume of a high Mw void marker, such as raw dextran) and Vt is the total volume of the column. Kd can be determined as (Ve−V0)/Vi, where Vi is the elution volume of a salt (e.g. NaCl) able to access all the volume except the matrix volume (the volume occupied by the matrix polymer molecules). By definition, both Kd and Kav values always lie within the range 0-1. The Kav value can advantageously be 0.6-0.95, e.g. 0.7-0.90 or 0.6-0.8, as measured with dextran of Mw 110 kDa as a probe molecule. The Kd value as measured with dextran of Mw 110 kDa can suitably be 0.68-0.90, such as 0.68-0.85 or 0.70-0.85. An advantage of this is that the support has a large fraction of pores able to accommodate both the polypeptides/multimers of the invention and immunoglobulins binding to the polypeptides/multimers and to provide mass transport of the immunoglobulins to and from the binding sites.


The polypeptides or multimers may be attached to the porous support via conventional coupling techniques utilising e.g. thiol, amino and/or carboxy groups present in the ligand. Bisepoxides, epichlorohydrin, CNBr, N-hydroxysuccinimide (NHS) etc are well-known coupling reagents. Between the support and the polypeptide/multimer, a molecule known as a spacer can be introduced, which improves the availability of the polypeptide/multimer and facilitates the chemical coupling of the polypeptide/multimer to the support. Depending on the nature of the polypeptide/multimer and the coupling conditions, the coupling may be a multipoint coupling (e.g. via a plurality of lysines) or a single point coupling (e.g. via a single cysteine).


In certain embodiments the polypeptides or multimers are coupled to the support via thioether bonds. Methods for performing such coupling are well-known in this field and easily performed by the skilled person in this field using standard techniques and equipment. Thioether bonds are flexible and stable and generally suited for use in affinity chromatography. In particular when the thioether bond is via a terminal or near-terminal cysteine residue on the polypeptide or multimer, the mobility of the coupled polypeptide/multimer is enhanced which provides improved binding capacity and binding kinetics. In some embodiments the polypeptide/multimer is coupled via a C-terminal cysteine provided on the protein as described above. This allows for efficient coupling of the cysteine thiol to electrophilic groups, e.g. epoxide groups, halohydrin groups etc. on a support, resulting in a thioether bridge coupling.


In certain embodiments the support comprises a polyhydroxy polymer, such as a polysaccharide. Examples of polysaccharides include e.g. dextran, starch, cellulose, pullulan, agar, agarose etc. Polysaccharides are inherently hydrophilic with low degrees of nonspecific interactions, they provide a high content of reactive (activatable) hydroxyl groups and they are generally stable towards alkaline cleaning solutions used in bioprocessing.


In some embodiments the support comprises agar or agarose. The supports used in the present invention can easily be prepared according to standard methods, such as inverse suspension gelation (S Hjertén: Biochim Biophys Acta 79(2), 393-398 (1964). Alternatively, the base matrices are commercially available products, such as crosslinked agarose beads sold under the name of SEPHAROSE™ FF (GE Healthcare). In an embodiment, which is especially advantageous for large-scale separations, the support has been adapted to increase its rigidity using the methods described in U.S. Pat. No. 6,602,990 or U.S. Pat. No. 7,396,467, which are hereby incorporated by reference in their entireties, and hence renders the matrix more suitable for high flow rates.


In certain embodiments the support, such as a polymer, polysaccharide or agarose support, is crosslinked, such as with hydroxyalkyl ether crosslinks. Crosslinker reagents producing such crosslinks can be e.g. epihalohydrins like epichlorohydrin, diepoxides like butanediol diglycidyl ether, allylating reagents like allyl halides or allyl glycidyl ether. Crosslinking is beneficial for the rigidity of the support and improves the chemical stability. Hydroxyalkyl ether crosslinks are alkali stable and do not cause significant nonspecific adsorption.


Alternatively, the porous support is based on synthetic polymers, such as polyvinyl alcohol, polyhydroxyalkyl acrylates, polyhydroxyalkyl methacrylates, polyacrylamides, polymethacrylamides etc. In case of hydrophobic polymers, such as matrices based on divinyl and monovinyl-substituted benzenes, the surface of the matrix is often hydrophilised to expose hydrophilic groups as defined above to a surrounding aqueous liquid. Such polymers are easily 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)).


Alternatively, a commercially available product, such as SOURCE™ (GE Healthcare) is used. In another alternative, the porous support according to the invention comprises a support of inorganic nature, e.g. silica, zirconium oxide etc.


In yet another embodiment, the solid support is in another form such as a surface, a chip, capillaries, or a filter (e.g. a membrane or a depth filter matrix).


As regards the shape of the matrix according to the invention, in one embodiment the matrix is in the form of a porous monolith. In an alternative embodiment, the matrix is in beaded or particle form that can be porous or non-porous. Matrices in beaded or particle form can be used as a packed bed or in a suspended form. Suspended forms include those known as expanded beds and pure suspensions, in which the particles or beads are free to move. In case of monoliths, packed bed and expanded beds, the separation procedure commonly follows conventional chromatography with a concentration gradient. In case of pure suspension, batch-wise mode will be used.


The separation matrix as disclosed above has excellent alkali stability and may be cleaned and/or sanitized using an alkaline cleaning liquid. The method of cleaning and/or sanitizing the separation matrix comprises the following steps:

    • a) optionally purifying a mixture comprising a first immunoglobulin using the separation matrix;
    • b) providing a cleaning liquid comprising at least 50% by volume of an aqueous alkali metal hydroxide solution; and
    • c) cleaning and/or sanitizing the separation matrix by contacting the cleaning liquid with the separation matrix for a predetermined contact time.


Cleaning and/or sanitization may be performed on separation matrix not previously used in order to sanitize the matrix prior to use, for example after packing a column comprising the separation matrix for the first time. Cleaning and/or sanitization may also be performed after using the separation matrix to purify an immunoglobulin, i.e. step a) of the method above. The steps of using the separation matrix to purify an immunoglobulin and cleaning and/or sanitizing the separation matrix afterwards may be repeated in order to maximize use of the separation matrix. The steps may be repeated at least 10 times, such as at least 50 times or 50-200 times. The step c) of cleaning/sanitizing may be performed after each step a) of purifying an immunoglobulin, or may be performed less frequently, such as after every second purification, or after every tenth purification. The step c) of cleaning and/or sanitizing the separation matrix need not be performed using the same conditions each time. For example, the separation matrix may be cleaned after each purification step using a first set of conditions, and after every nth purification be sanitized using more stringent conditions.


Because aqueous alkali metal hydroxide solutions are effective cleaning agents and bactericidal in themselves, by cleaning and sanitizing using a cleaning liquid comprising such aqueous alkali metal hydroxide solutions there is a lesser need for using alcohols as cleaning and sanitizing agents. Alkali metal hydroxides are relatively cheap compared to alcohols and are subject to less regulatory burden. Moreover, they are non-flammable and easier to dispose of.


The separation matrix is a separation matrix as disclosed above, comprising multimers of immunoglobulin-binding alkali-stabilized Protein A domains covalently coupled to a porous support, wherein the alkali-stabilized Protein A domains comprise mutants of a parental Fc-binding domain of Staphylococcus Protein A (SpA), as defined by, or having at least 80% such as at least 90%, 95% or 98% identity to, SEQ ID NO: 51 or SEQ ID NO: 52, wherein the amino acid residues at positions 13 and 44 of SEQ ID NOs: 51 or 52 are asparagines, and wherein at least the asparagine residue at position 3 of SEQ ID NOs: 51 or 52 has been mutated to an amino acid selected from the group consisting of glutamic acid, lysine, tyrosine, threonine, phenylalanine, leucine, isoleucine, tryptophan, methionine, valine, alanine, histidine and arginine. The immunoglobulin-binding alkali-stabilized Protein A domains may comprise further mutations. For example, the glutamine residue at position 1 of SEQ ID NOs: 51 or 52 may be mutated to an alanine; and/or the asparagine or glutamic acid residue at position 35 of SEQ ID NOs: 51 or 52 may be mutated to an alanine.


Due to the high alkaline stability of the alkali-stabilized Protein A domains, the separation matrix can tolerate extensive cleaning and sanitization procedures, even using concentrated alkali metal hydroxide solutions, without excessive loss of binding capacity. For example, the separation matrix may retain at least 80% of its original dynamic binding capacity after cleaning and/or sanitization.


The cleaning liquid comprises at least 50% by volume of an aqueous alkali metal hydroxide solution. The aqueous alkali metal hydroxide solution may comprise a single alkali metal hydroxide or a mixture of alkali metal hydroxides, such as sodium hydroxide, potassium hydroxide, or a mixture of sodium hydroxide and potassium hydroxide. The aqueous alkali metal hydroxide solution may have a molarity of from 500 mM to 5 M, such as from 1 M to 2 M, expressed as the total combined concentration of alkali metal hydroxides if a mixture of alkali metal hydroxides is used. The cleaning liquid may essentially consist of, or consist of, the aqueous alkali metal hydroxide solution. However, the cleaning liquid may in some embodiments also comprise further components. Such further components may include alcohols, such as a C2-C7 alcohol, such as ethanol, isopropanol or benzoic alcohol. Such further components may include salts, such as sodium chloride. The use of alcohols and/or salts in the cleaning liquid may increase the efficacy of the cleaning liquid in inhibiting or inactivating certain microorganisms, such as spore-forming bacteria.


Non-limiting examples of cleaning liquids include:


Sodium hydroxide solution (0.5 M, 1 M, 2 M or 5 M);


Potassium hydroxide solution (0.5 M, 1 M, 2 M or 5 M);


Sodium hydroxide solution (0.5 M, 1 M, 2 M or 5 M) with 10-20% by volume ethanol;


Sodium hydroxide solution (0.5 M, 1 M, 2 M or 5 M) with 10-50% by volume isopropanol;


Sodium hydroxide solution (0.5 M, 1 M, 2 M or 5 M) with 1-5% by volume benzyl alcohol;


Potassium hydroxide solution (0.5 M, 1 M, 2 M or 5 M) with 10-20% by volume ethanol;


Potassium hydroxide solution (0.5 M, 1 M, 2 M or 5 M) with 10-50% by volume isopropanol; or


Potassium hydroxide solution (0.5 M, 1 M, 2 M or 5 M) with 1-5% by volume benzyl alcohol.


The use of relatively concentrated alkali metal hydroxide solution, such as 0.5 M-5 M solution, provides exceptional removal of matrix contaminants such as proteins and nucleic acids, as well as quickly and effectively inactivating viruses, bacteria, yeast and fungi. A commonly applied measure of sanitization efficacy is a 6-log10 reduction in the microorganism or contaminant being measured. Sanitizing with concentrated alkali metal hydroxide solution is capable of providing a 6-log10 reduction in most microorganisms and endotoxins.


During cleaning of the separation matrix, the cleaning liquid should be passed through the separation matrix with a suitable flow rate, in order to flush contaminants from the matrix. During sanitization, the cleaning liquid is initially passed through the separation matrix with a suitable flow rate until the separation matrix is fully permeated with the cleaning liquid. The cleaning liquid is preferably passed through the separation matrix with a suitable flow rate for the entire duration of the sanitization step. However, if desired the flow of cleaning liquid through the separation matrix may be stopped once the separation matrix is fully permeated with the cleaning liquid. This reduces the volume of cleaning fluid required to perform a sanitization step, but provides a poorer cleaning of the separation matrix as compared to a procedure where a constant flow of cleaning fluid is applied.


Because aqueous alkali metal hydroxide solutions are so effective in removing contaminants from the separation matrix, the separation matrix may be used as a purification platform for the purification of a variety of immunoglobulins, with a lower risk of host cell protein contamination or carryover. After purification of a first immunoglobulin product, the separation matrix is first cleaned and sanitized thoroughly using a cleaning liquid comprising aqueous alkali metal hydroxide solution prior to use in purification of the second immunoglobulin product.


EXAMPLES

Mutagenesis of Protein


Site-directed mutagenesis was performed by a two-step PCR using oligonucleotides coding for the mutations. As template a plasmid containing a single domain of either Z, B or C was used. The PCR fragments were ligated into an E. coli expression vector. DNA sequencing was used to verify the correct sequence of inserted fragments.


To form multimers of mutants an Acc I site located in the starting codons (GTA GAC) of the B, C or Z domain was used, corresponding to amino acids VD. The vector for the monomeric domain was digested with Acc I and phosphatase treated. Acc I sticky-ends primers were designed, specific for each variant, and two overlapping PCR products were generated from each template. The PCR products were purified and the concentration was estimated by comparing the PCR products on a 2% agarose gel. Equal amounts of the pair wise PCR products were hybridized (90° C.→25° C. in 45 min) in ligation buffer. The resulting product consists approximately to % of fragments likely to be ligated into an Acc I site (correct PCR fragments and/or the digested vector). After ligation and transformation colonies were PCR screened to identify constructs containing the desired mutant. Positive clones were verified by DNA sequencing.


Construct Expression and Purification


The constructs were expressed in the bacterial periplasm by fermentation of E. coli K12 in standard media. After fermentation the cells were heat-treated to release the periplasm content into the media. The constructs released into the medium were recovered by microfiltration with a membrane having a 0.2 μm pore size.


Each construct, now in the permeate from the filtration step, was purified by affinity. The permeate was loaded onto a chromatography medium containing immobilized IgG (IgG Sepharose 6FF, GE Healthcare). The loaded product was washed with phosphate buffered saline and eluted by lowering the pH.


The elution pool was adjusted to a neutral pH (pH 8) and reduced by addition of dithiothreitol. The sample was then loaded onto an anion exchanger. After a wash step the construct was eluted in a NaCl gradient to separate it from any contaminants. The elution pool was concentrated by ultrafiltration to 40-50 mg/ml. It should be noted that the successful affinity purification of a construct on an immobilized IgG medium indicates that the construct in question has a high affinity to IgG.


The purified ligands were analyzed with RPC LC-MS to determine the purity and to ascertain that the molecular weight corresponded to the expected (based on the amino acid sequence).


Example 1

The purified monomeric ligands listed in Table 1, further comprising for SEQ ID NOs: 8-16, 23-28 and 36-48 an AQGT leader sequence at the N-terminus and a cysteine at the C terminus, were immobilized on Biacore CM5 sensor chips (GE Healthcare, Sweden), using the amine coupling kit of GE Healthcare (for carbodiimide coupling of amines on the carboxymethyl groups on the chip) in an amount sufficient to give a signal strength of about 200-1500 RU in a Biacore surface plasmon resonance (SPR) instrument (GE Healthcare, Sweden). To follow the IgG binding capacity of the immobilized surface 1 mg/ml human polyclonal IgG (Gammanorm) was flowed over the chip and the signal strength (proportional to the amount of binding) was noted. The surface was then cleaned-in-place (CIP), i.e. flushed with 500 mM NaOH for 10 minutes at room temperature (22+/−2° C.). This was repeated for 96-100 cycles and the immobilized ligand alkaline stability was followed as the remaining IgG binding capacity (signal strength) after each cycle. The results are shown in Table 1 and indicate that at least the ligands Zvar(N11K)1, Zvar(N11E)1, Zvar(N11Y)1, Zvar(N11T)1, Zvar(N11F)1, Zvar(N11L)1, Zvar(N11W)1, ZN11I)1, Zvar(N11M)1, Zvar(N11V)1, Zvar(N11A)1, Zvar(N11H1), Zvar(N11R)1, Zvar(N11E, Q32A)1, Zvar(N11E, Q32E, Q40E)1 and Zvar(N11E, Q32E, K50R)1, Zvar(Q9A, N11E, N43A)1, Zvar(Q9A, N11E, N28A, N43A)1, Zvar(Q9A, N11E, Q40V, A42K, N43E, L44I)1, Zvar(Q9A, N11E, Q40V, A42K, N43A, L44I)1, Zvar(Q9A, N11E, N28A, Q40V, A42K, N43A, L44I)1, Zvar(N11K, H18K, S33K, D37E, A42R, N43A, L44I, K50R, L51Y)1, Zvar(Q9A, N11K, H18K, S33K, D37E, A42R, N43A, L44I, K50R, L51Y)1, Zvar(N11K, H18K, D37E, A42R, N43A, L44I)1, Zvar(Q9A, N11K, H18K, D37E, A42R, N43A, L44I)1 and Zvar(Q9A, N11K, H18K, D37E, A42R, N43A, L44I, K50R)1, as well as the varieties of Zvar(Q9A, N11E, Q40V, A42K, N43A, L44I)1 having G, S, Y, Q, T, N, F, L, W, I, M, V, D, E, H, R or K in position 29, the varieties of Zvar(Q9A, N11E, Q40V, A42K, N43A, L44I)1 having F, Y, W, K or R in position 53 and the varieties of Zvar(Q9A, N11E, Q40V, A42K, N43A, L44I)1 where Q9, Q40, A42 or N43 has been deleted, have an improved alkali stability compared to the parental structure Zvar1, used as the reference. Further, the ligands B(Q9A, N11E, Q40V, A42K, N43A, L44I)1 and C(Q9A, N11E, E43A)1 have an improved stability compared to the parental B and C domains, used as references.









TABLE 1







Monomeric ligands, evaluated by Biacore (0.5M NaOH).













Ca-
Ref-
Ca-




pacity
erence
pacity




after
capacity
relative




96-
after 96-
to



Se-
100
100
ref-


Ligand
quence
cycles
cycles
erence














Zvar(N11E, Q32A)1
SEQ ID
57%
55%
1.036



NO 12





Zvar(N11E)1
SEQ ID
59%
55%
1.073



NO 13





Zvar(N11E, Q32E, Q40E)1
SEQ ID
52%
51%
1.020



NO 14





Zvar(N11E, Q32E, K50R)1
SEQ ID
53%
51%
1.039



NO 15





Zvar(N11K)1
SEQ ID
62%
49%
1.270



NO 16





Zvar(N11Y)1
SEQ ID
55%
46%
1.20



NO 38





Zvar(N11T)1
SEQ ID
50%
46%
1.09



NO 39





Zvar(N11F)1
SEQ ID
55%
46%
1.20



NO 40





Zvar(N11L)1
SEQ ID
57%
47%
1.21



NO 41





Zvar(N11W)1
SEQ ID
57%
47%
1.21



NO 42





Zvar(N11I)1
SEQ ID
57%
47%
1.21



NO 43





Zvar(N11M)1
SEQ ID
58%
46%
1.26



NO 44





Zvar(N11V)1
SEQ ID
56%
46%
1.22



NO 45





Zvar(N11A)1
SEQ ID
58%
46%
1.26



NO 46





Zvar(N11H)1
SEQ ID
57%
46%
1.24



NO 47





Zvar(N11R)1
SEQ ID
59%
46%
1.28



NO 48





Zvar(Q9A, N11E, N43A)1
SEQ ID
70%
47%
1.49



NO 8





Zvar(Q9A, N11E, N28A,
SEQ ID
68%
47%
1.45


N43A)1
NO 9





Zvar(Q9A, N11E, Q40V,
SEQ ID
67%
47%
1.43


A42K, N43E, L44I)1
NO 10





Zvar(Q9A, N11E, Q40V,
SEQ ID
66%
47%
1.40


A42K, N43A, L44I)1
NO 11





Zvar(Q9A, N11E, N28A, Q40V,
SEQ ID
65%
48%
1.35


A42K, N43A, L44I)1
NO 24





Zvar(N11K, H18K,
SEQ ID
67%
46%
1.46


S33K, D37E, A42R,
NO 23





N43A, L44I, K50R, L51Y)1






Zvar(Q9A, N11K,
SEQ ID
59%
46%
1.28


H18K, S33K, D37E,
NO 25





A42R, N43A, L44I,






K50R, L51Y)1






Zvar(N11K, H18K, D37E,
SEQ ID
59%
45%
1.31


A42R, N43A, L441)1
NO 26





Zvar(Q9A, N11K, H18K, D37E,
SEQ ID
63%
45%
1.40


A42R, N43A, L441)1
NO 27





Zvar(Q9A, N11K, H18K, D37E,
SEQ ID
67%
45%
1.49


A42R, N43A, L44I, K50R)1
NO 28





B(Q9A, N11E, Q40V, A42K,
SEQ ID
39%
35%
1.11


N43A, L44I)1
NO 36





C(Q9A, N11E, E43A)1
SEQ ID
60%
49%
1.22



NO 37





Zvar(Q9A, N11E, A29G, Q40V,
SEQ ID
69%
48%
1.44


A42K, N43A, L44I)1
NO 54





Zvar(Q9A, N11E, A29S, Q40V,
SEQ ID
66%
48%
1.38


A42K, N43A, L44I)1
NO 55





Zvar(Q9A, N11E, A29Y, Q40V,
SEQ ID
61%
48%
1.27


A42K, N43A, L44I)1
NO 56





Zvar(Q9A, N1 1E, A29Q, Q40V,
SEQ ID
60%
47%
1.28


A42K, N43A, L441)1
NO 57





Zvar(Q9A, N11E, A29T, Q40V,
SEQ ID
60%
47%
1.28


A42K, N43A, L44I)1
NO 58





Zvar(Q9A, N11E, A29N, Q40V,
SEQ ID
61%
47%
1.30


A42K, N43A, L44I)1
NO 59





Zvar(Q9A, N11E, A29F, Q40V,
SEQ ID
62%
46%
1.35


A42K, N43A, L44I)1
NO 60





Zvar(Q9A, N11E, A29L, Q40V,
SEQ ID
61%
46%
1.33


A42K, N43A, L44I)1
NO 61





Zvar(Q9A, N11E, A29W, Q40V,
SEQ ID
60%
46%
1.30


A42K, N43A, L44I)1
NO 62





Zvar(Q9A, N11E, A291, Q40V,
SEQ ID
58%
47%
1.23


A42K, N43A, L44I)1
NO 63





Zvar(Q9A, N11E, A29M, Q40V,
SEQ ID
62%
47%
1.32


A42K, N43A, L44I)1
NO 64





Zvar(Q9A, N11E, A29V, Q40V,
SEQ ID
62%
47%
1.32


A42K, N43A, L44I)1
NO 65





Zvar(Q9A, N11E, A29D, Q40V,
SEQ ID
56%
47%
1.19


A42K, N43A, L44I)1
NO 66





Zvar(Q9A, N11E, A29E, Q40V,
SEQ ID
57%
47%
1.21


A42K, N43A, L44I)1
NO 67





Zvar(Q9A, N11E, A29H, Q40V,
SEQ ID
57%
47%
1.21


A42K, N43A, L44I)1
NO 68





Zvar(Q9A, N11E, A29R, Q40V,
SEQ ID
58%
46%
1.26


A42K, N43A, L44I)1
NO 69





Zvar(Q9A, N11E, A29K,
SEQ ID
59%
46%
1.28


Q40V, A42K, N43A, L44I)1
NO 70





Zvar(Q9A, N11E, Q40V,
SEQ ID
58%
46%
1.26


A42K, N43A, L44I, D53F)1
NO 71





Zvar(Q9A, N11E, Q40V,
SEQ ID
59%
46%
1.28


A42K, N43A, L44I, D53Y)1
NO 72





Zvar(Q9A, N11E, Q40V, A42K,
SEQ ID
62%
46%
1.35


N43A, L44I, D53W)1
NO 73





Zvar(Q9A, N11E, Q40V,
SEQ ID
65%
46%
1.41


A42K, N43A, L44I, D53K)1
NO 74





Zvar(Q9A, N11E, Q40V,
SEQ ID
60%
46%
1.30


A42K, N43A, L44I, D53R)1
NO 75





Zvar(Q9del, N11E, Q40V,
SEQ ID
60%
46%
1.30


A42K, N43A, L44I)1
NO 76





Zvar(Q9A, N11E, Q40del,
SEQ ID
59%
46%
1.28


A42K, N43A, L441)1
NO 77





Zvar(Q9A, N11E, Q40V,
SEQ ID
57%
46%
1.24


A42del, N43A, L44I)1
NO 78





Zvar(Q9A, N11E, Q40V,
SEQ ID
55%
46%
1.20


A42K, N43del, L44I)1
NO 79









The Biacore experiment can also be used to determine the binding and dissociation rates between the ligand and IgG. This was used with the set-up as described above and with an IgG1 monoclonal antibody as probe molecule. For the reference Zvar1, the on-rate (105 M−1s−1) was 3.1 and the off-rate (105 s−1) was 22.1, giving an affinity (off-rate/on-rate) of 713 pM. For Zvar(Q9A, N11E, Q40V, A42K, N43A, L44I)1 (SEQ ID NO: 11), the on-rate was 4.1 and the off-rate 43.7, with affinity 1070 pM. The IgG affinity was thus somewhat higher for the mutated variant.


Example 2

The purified dimeric, tetrameric and hexameric ligands listed in Table 2 were immobilized on Biacore CM5 sensor chips (GE Healthcare, Sweden), using the amine coupling kit of GE Healthcare (for carbodiimide coupling of amines on the carboxymethyl groups on the chip) in an amount sufficient to give a signal strength of about 200-1500 RU in a Biacore instrument (GE Healthcare, Sweden). To follow the IgG binding capacity of the immobilized surface 1 mg/ml human polyclonal IgG (Gammanorm) was flowed over the chip and the signal strength (proportional to the amount of binding) was noted. The surface was then cleaned-in-place (CIP), i.e. flushed with 500 mM NaOH for 10 minutes at room temperature (22+/−2° C.). This was repeated for 300 cycles and the immobilized ligand alkaline stability was followed as the remaining IgG binding capacity (signal strength) after each cycle. The results are shown in Table 2 and in FIG. 2 and indicate that at least the ligands Zvar(Q9A, N11E, N43A)4, Zvar(Q9A, N11E, N28A, N43A)4, Zvar(Q9A, N11E, Q40V, A42K, N43E, L44I)4 and Zvar(Q9A, N11E, Q40V, A42K, N43A, L44I)4, Zvar(Q9A, N11E, D37E, Q40V, A42K, N43A, L44I)4 and Zvar(Q9A, N11E, D37E, Q40V, A42R, N43A, L44I)4 have an improved alkali stability compared to the parental structure Zvar4, which was used as a reference. The hexameric ligand Zvar(Q9A, N11E, Q40V, A42K, N43A, L44I)6 also has improved alkali stability compared to the parental structure Zvar6, used as a reference. Further, Zvar(Q9A, N11E, Q40V, A42K, N43A, L44I) dimers with deletions of a) D2, A3, K4; b) K58, V1, D2; c) P57, K58, V1, D2, A3; d) K4, F5, D6, K7, E8; e) A56, P57, K58; V1, D2, A3 or f) V1, D2, A3, K4, F5, D6, K7, E8 from the linker region between the two monomer units have improved alkali stability compared to the parental structure Zvar2, used as a reference. Also Zvar(Q9A, N11E, Q40V, A42K, N43A, L44I) dimers with an insertion of YEDG between K58 and V1 in the linker region have improved alkali stability compared to Zvar2.









TABLE 2







Dimeric, tetrameric and hexameric ligands, evaluated by Biacore (0.5M NaOH).

















Capacity

Capacity

Capacity




Remaining
relative
Remaining
relative
Remaining
relative



SEQ
capacity
to ref.
capacity
to ref.
capacity
to ref.



ID
100 cycles
100
200 cycles
200
300 cycles
300


Ligand
NO:
(%)
cycles
(%)
cycles
(%)
cycles

















Zvar4
21
67
1
36
1
16
1


Zvar(Q9A, N11E, N43A)4
17
81
1.21
62
1.72
41
2.56


Zvar(Q9A, N11E, N28A,
18
80
1.19
62
1.72
42
2.62


N43A)4


Zvar(Q9A, N11E, Q40V,
19
84
1.25
65
1.81
48
3.00


A42K, N43E, L44I)4


Zvar(Q9A, N11E, Q40V,
20
90
1.34
74
2.06
57
3.56


A42K, N43A, L44I)4


Zvar(Q9A, N11E, N28A,
32
84
1.24
Not tested
Not tested
Not tested
Not tested


Q40V, A42K, N43A, L44I)4


Zvar(Q9A, N11E, Q40V,
33
87
1.30
Not tested
Not tested
Not tested
Not tested


A42K, N43A, L44I)6


Zvar(Q9A, N11E, D37E,
34
81
1.13
Not tested
Not tested
Not tested
Not tested


Q40V, A42K, N43A, L44I)4


Zvar(Q9A, N11E, D37E,
35
84
1.17
Not tested
Not tested
Not tested
Not tested


Q40V, A42R, N43A, L44I)4


Zvar(Q9A, N11E, Q40V,
80
70
1.27
Not tested
Not tested
Not tested
Not tested


A42K, N43A, L44I)2 with


D2, A3 and K4 in linker


deleted


Zvar(Q9A, N11E, Q40V,
81
76
1.38
Not tested
Not tested
Not tested
Not tested


A42K, N43A, L44I)2 with


K58, V1 and D2 in linker


deleted


Zvar(Q9A, N11E, Q40V,
82
74
1.35
Not tested
Not tested
Not tested
Not tested


A42K, N43A, L44I)2 with


P57, K58, V1, D2 and A3


in linker deleted


Zvar(Q9A, N11E, Q40V,
83
70
1.30
Not tested
Not tested
Not tested
Not tested


A42K, N43A, L44I)2 with


K4, F5, D6, K7 and E8 in


linker deleted


Zvar(Q9A, N11E, Q40V,
84
68
1.26
Not tested
Not tested
Not tested
Not tested


A42K, N43A, L44I)2 with


A56, P57 and K58 in


linker deleted


Zvar(Q9A, N11E, Q40V,
85
75
1.39
Not tested
Not tested
Not tested
Not tested


A42K, N43A, L44I)2 with


V1, D2 and A3 in


linker deleted


Zvar(Q9A, N11E, Q40V,
86
62
1.13
Not tested
Not tested
Not tested
Not tested


A42K, N43A, L44I)2 with


V1, D2, A3, K4, F5, D6,


K7 and E8 in


linker deleted


Zvar(Q9A, N11E, Q40V,
87
72
1.31
Not tested
Not tested
Not tested
Not tested


A42K, N43A, L44I)2 with


YEDG inserted in linker


between K58 and V1


Zvar2
88
55
1
Not tested
Not tested
Not tested
Not tested









Example 3

Example 2 was repeated with 100 CIP cycles of three ligands using 1 M NaOH instead of 500 mM as in Example 2. The results are shown in Table 3 and show that all three ligands have an improved alkali stability also in 1M NaOH, compared to the parental structure Zvar4 which was used as a reference.









TABLE 3







Tetrameric ligands, evaluated by Biacore (1M NaOH).












Remaining
Capacity




capacity 100
relative to


Ligand
Sequence
cycles (%)
ref. 100 cycles













Zvar4
SEQ ID NO 21
27
1


Zvar(Q9A, N11E,
SEQ ID NO 18
55
2.04


N28A, N43A)4





Zvar(Q9A, N11E, Q40V,
SEQ ID NO 19
54
2.00


A42K, N43E, L44I)4





Zvar(Q9A, N11E, Q40V,
SEQ ID NO 20
56
2.07


A42K, N43A, L44I)4









Example 4

The purified tetrameric ligands of Table 2 (all with an additional N-terminal cysteine) were immobilized on agarose beads using the methods described below and assessed for capacity and stability. The results are shown in Table 4 and FIG. 3.









TABLE 4







Matrices with tetrametric ligands, evaluated in columns (0.5 M NaOH).

















Remaining

Capacity






IgG capacity
Remaining
retention





Initial
Qb10
IgG capacity
relative



SEQ
Ligand
IgG capacity
after six
after six
to ref.



ID
content
Qb10
4 h cycles
4 h cycles
after six


Ligand
NO.
(mg/ml)
(mg/ml)
(mg/ml)
(%)
4 h cycles
















Zvar4
21
7
52.5
36.5
60
1


Zvar4
21
12
61.1
43.4
71
1


Zvar(Q9A, N11E, N28A, N43A)4
18
7.0
49.1
44.1
90
1.50


Zvar(Q9A, N11E, N28A, N43A)4
18
12.1
50.0
46.2
93
1.31


Zvar(Q9A, N11E, Q40V, A42K, N43A, L44I)4
20
7.2
49.0
44.2
90
1.50


Zvar(Q9A, N11E, Q40V, A42K, N43A, L44I)4
20
12.8
56.3
53.6
95
1.34


Zvar(N11K, H18K, S33K, D37E, A42R, N43A,
30
9.7
56.3
52.0
92
1.53


L44I, K50R, L51Y)4


Zvar(Q9A, N11K, H18K, D37E, A42R)4
31
10.8
56.9
52.5
92
1.30









Activation


The base matrix used was rigid cross-linked agarose beads of 85 micrometers (volume-weighted, d50V) median diameter, prepared according to the methods of U.S. Pat. No. 6,602,990, hereby incorporated by reference in its entirety, and with a pore size corresponding to an inverse gel filtration chromatography Kav value of 0.70 for dextran of Mw 110 kDa, according to the methods described in Gel Filtration Principles and Methods, Pharmacia LKB Biotechnology 1991, pp 6-13.


25 mL (g) of drained base matrix, 10.0 mL distilled water and 2.02 g NaOH (s) was mixed in a 100 mL flask with mechanical stirring for 10 min at 25° C. 4.0 mL of epichlorohydrin was added and the reaction progressed for 2 hours. The activated gel was washed with 10 gel sediment volumes (GV) of water.


Coupling


To 20 mL of ligand solution (50 mg/mL) in a 50 ml Falcon tube, 169 mg NaHCO3, 21 mg Na2CO3, 175 mg NaCl and 7 mg EDTA, was added. The Falcon tube was placed on a roller table for 5-10 min, and then 77 mg of DTE was added. Reduction proceeded for >45 min. The ligand solution was then desalted on a PD10 column packed with Sephadex G-25. The ligand content in the desalted solution was determined by measuring the 276 nm UV absorption.


The activated gel was washed with 3-5 GV {0.1 M phosphate/1 mM EDTA pH 8.6} and the ligand was then coupled according to the method described in U.S. Pat. No. 6,399,750, hereby incorporated by reference in its entirety. All buffers used in the experiments had been degassed by nitrogen gas for at least 5-10 min. The ligand content of the gels could be controlled by varying the amount and concentration of the ligand solution.


After immobilization the gels were washed 3×GV with distilled water. The gels+1 GV {0.1 M phosphate/1 mM EDTA/10% thioglycerol pH 8.6} was mixed and the tubes were left in a shaking table at room temperature overnight. The gels were then washed alternately with 3×GV {0.1 M TRIS/0.15 M NaCl pH 8.6} and 0.5 M HAc and then 8-10×GV with distilled water. Gel samples were sent to an external laboratory for amino acid analysis and the ligand content (mg/ml gel) was calculated from the total amino acid content.


Protein


Gammanorm 165 mg/ml (Octapharma), diluted to 2 mg/ml in Equilibration buffer.


Equilibration Buffer


PBS Phosphate buffer 10 mM+0.14 M NaCl+0.0027 M KCl, pH 7.4 (Medicago)


Adsorption Buffer


PBS Phosphate buffer 10 mM+0.14 M NaCl+0.0027 M KCl, pH 7.4 (Medicago)


Elution Buffers


100 mM acetate pH 2.9


Dynamic Binding Capacity


2 ml of resin was packed in TRICORN™ 5 100 columns. The breakthrough capacity was determined with an AKTAExplorer 10 system at a residence time of 6 minutes (0.33 ml/min flow rate). Equilibration buffer was run through the bypass column until a stable baseline was obtained. This was done prior to auto zeroing. Sample was applied to the column until a 100% UV signal was obtained. Then, equilibration buffer was applied again until a stable baseline was obtained.


Sample was loaded onto the column until a UV signal of 85% of maximum absorbance was reached. The column was then washed with 5 column volumes (CV) equilibration buffer at flow rate 0.5 ml/min. The protein was eluted with 5 CV elution buffer at a flow rate of 0.5 ml/min. Then the column was cleaned with 0.5M NaOH at flow rate 0.2 ml/min and re-equilibrated with equilibration buffer.


For calculation of breakthrough capacity at 10%, the equation below was used. That is the amount of IgG that is loaded onto the column until the concentration of IgG in the column effluent is 10% of the IgG concentration in the feed.







q

10

%


=



C
0


V
C




[


V
app

-

V
sys

-




V
sys


V
app







A


(
V
)


-

A
sub




A

100

%


-

A
sub



*
dv



]








    • A100%=100% UV signal;

    • Asub=absorbance contribution from non-binding IgG subclass;

    • A(V)=absorbance at a given applied volume;

    • Vc=column volume;

    • Vapp=volume applied until 10% breakthrough;

    • Vsys=system dead volume;

    • C0=feed concentration.





The dynamic binding capacity (DBC) at 10% breakthrough was calculated. The dynamic binding capacity (DBC) was calculated for 10 and 80% breakthrough.

CIP—0.5 M NaOH


The 10% breakthrough DBC (Qb10) was determined both before and after repeated exposures to alkaline cleaning solutions. Each cycle included a CIP step with 0.5 M NaOH pumped through the column at a rate of 0.5/min for 20 min, after which the column was left standing for 4 h. The exposure took place at room temperature (22+/−2° C.). After this incubation, the column was washed with equilibration buffer for 20 min at a flow rate of 0.5 ml/min. Table 4 shows the remaining capacity after six 4 h cycles (i.e. 24 h cumulative exposure time to 0.5 M NaOH), both in absolute numbers and relative to the initial capacity.


Example 5

Example 4 was repeated with the tetrameric ligands shown in Table 5, but with 1.0 M NaOH used in the CIP steps instead of 0.5 M. The results are shown in Table 5 and in FIG. 4.









TABLE 5







Matrices with tetrametric ligands, evaluated in columns - 1.0 M NaOH.



















Capacity






Remaining

retention






IgG capacity
Remaining IgG
relative





Initial
Qb10
capacity
to ref.



SEQ
Ligand
IgG capacity
after six 4 h
after six 4 h
after



ID
content
Qb10
cycles
cycles
six 4 h


Ligand
NO.
(mg/ml)
(mg/ml)
(mg/ml)
(%)
cycles
















Zvar4
21
12
60.1
33.5
56
1


Zvar(Q9A, N11E, Q40V, A42K, N43A, L44I)4
20
12.8
60.3
56.0
93
1.67


Zvar(N11K, H18K, S33K, D37E, A42R, N43A,
30
9.7
62.1
48.1
77
1.44


L44I, K50R, L51Y)4









Example 6

Base Matrices


The base matrices used were a set of rigid cross-linked agarose bead samples of 59-93 micrometers (volume-weighted, d50V) median diameter (determined on a Malvern Mastersizer 2000 laser diffraction instrument), prepared according to the methods of U.S. Pat. No. 6,602,990 and with a pore size corresponding to an inverse gel filtration chromatography Kd value of 0.62-0.82 for dextran of Mw 110 kDa, according to the methods described above, using HR10/30 columns (GE Healthcare) packed with the prototypes in 0.2 M NaCl and with a range of dextran fractions as probe molecules (flow rate 0.2 ml/min). The dry weight of the bead samples ranged from 53 to 86 mg/ml, as determined by drying 1.0 ml drained filter cake samples at 105° C. overnight and weighing.









TABLE 6







Base matrix samples












Base

d50v
Dry weight



matrix
Kd
(μm)
(mg/ml)
















A18
0.704
59.0
56.0



A20
0.70
69.2
55.8



A27
0.633
87.2
74.2



A28
0.638
67.4
70.2



A29
0.655
92.6
57.5



A32
0.654
73.0
70.5



A33
0.760
73.1
55.5



A38
0.657
70.9
56.2



A39
0.654
66.0
79.1



A40
0.687
64.9
74.9



A41
0.708
81.7
67.0



A42
0.638
88.0
59.4



A43
0.689
87.5
77.0



A45
0.670
56.6
66.0



A52
0.620
53.10
63.70



A53
0.630
52.6
86.0



A54
0.670
61.3
75.3



A55
0.640
62.0
69.6



A56
0.740
61.0
56.0



A56-2
0.740
51.0
56.0



A62a
0.788
48.8
70.1



A62b
0.823
50.0
46.9



A63a
0.790
66.8
59.6



A63b
0.765
54.0
79.0



A65a
0.796
58.0
60.0



A65b
0.805
57.3
46.0



B5
0.793
69.0
84.4



C1
0.699
71.0
73.4



C2
0.642
66.5
81.1



C3
0.711
62.0
82.0



C4
0.760
62.0
82.0



H31
0.717
82.0
59.0



H35
0.710
81.1
61.0



H40
0.650
52.8
65.0



I1
0.640
50.0
67.0



41
0.702
81.6
60.6










Coupling


100 ml base matrix was washed with 10 gel volumes distilled water on a glass filter. The gel was weighed (1 g=1 ml) and mixed with 30 ml distilled water and 8.08 g NaOH (0.202 mol) in a 250 ml flask with an agitator. The temperature was adjusted to 27+/−2° C. in a water bath. 16 ml epichlorohydrin (0.202 mol) was added under vigorous agitation (about 250 rpm) during 90+/−10 minutes. The reaction was allowed to continue for another 80+/−10 minutes and the gel was then washed with >10 gel volumes distilled water on a glass filter until neutral pH was reached. This activated gel was used directly for coupling as below.


To 16.4 mL of ligand solution (50 mg/mL) in a 50 ml Falcon tube, 139 mg NaHCO3, 17.4 mg Na2CO3, 143.8 mg NaCl and 141 mg EDTA, was added. The Falcon tube was placed on a roller table for 5-10 min, and then 63 mg of DTE was added. Reduction proceeded for >45 min. The ligand solution was then desalted on a PD10 column packed with Sephadex G-25. The ligand content in the desalted solution was determined by measuring the 276 nm UV absorption.


The activated gel was washed with 3-5 GV {0.1 M phosphate/1 mM EDTA pH 8.6} and the ligand was then coupled according to the method described in U.S. Pat. No. 6,399,750 5.2.2, although with considerably higher ligand amounts (see below). All buffers used in the experiments had been degassed by nitrogen gas for at least 5-10 min. The ligand content of the gels was controlled by varying the amount and concentration of the ligand solution, adding 5-20 mg ligand per ml gel. The ligand was either a tetramer (SEQ ID NO: 20) or a hexamer (SEQ ID NO: 33) of an alkali-stabilized mutant.


After immobilization the gels were washed 3×GV with distilled water. The gels+1 GV {0.1 M phosphate/1 mM EDTA/10% thioglycerol pH 8.6} was mixed and the tubes were left in a shaking table at room temperature overnight. The gels were then washed alternately with 3×GV {0.1 M TRIS/0.15 M NaCl pH 8.6} and 0.5 M HAc and then 8-10×GV with distilled water. Gel samples were sent to an external laboratory for amino acid analysis and the ligand content (mg/ml gel) was calculated from the total amino acid content.


Evaluation


The Qb10% dynamic capacity for polyclonal human IgG at 2.4 and 6 min residence time was determined as outlined in Example 4.









TABLE 7







Prototype results














Ligand

Qb10%
Qb10%



Base
content

2.4 min
6 min


Prototype
matrix
(mg/ml)
Multimer
(mg/ml)
(mg/ml)















N1
A38
7.45
tetramer
44.4
58.25


N2
A20
7.3
tetramer
45.12
57.21


N3
A42
6.72
tetramer
33.56
50.02


N4
A29
7.3
tetramer
36.34
51.8


N5
A28
7.9
tetramer
42.38
58.25


N6
A39
6.96
tetramer
41.88
54.67


N7
A27
7.5
tetramer
29.19
48.73


N8
A43
6.99
tetramer
33.43
49.79


N9
A38
11.34
tetramer
48.1
72.78


N10
A20
10.6
tetramer
50.66
70.07


N11
A42
11.1
tetramer
32.25
57.78


N12
A29
11
tetramer
34.85
64.68


N13
A28
11.9
tetramer
39.92
63.75


N14
A39
10.48
tetramer
44.37
64.79


N15
A27
12.1
tetramer
24.8
55.56


N16
A43
10.51
tetramer
31.82
58.04


N17
A41
8.83
tetramer
38.5
56.8


N18
A41
8.83
tetramer
37.84
58.6


N19
A41
8.83
tetramer
35.06
57.23


N20
A41
5.0
tetramer
35.64
46.04


N21
A41
13.0
tetramer
34.95
62.23


N22
A40
13.15
tetramer
56.85
71.09


N23
A33
7.33
tetramer
48.69
55.76


N24
A40
11.03
tetramer
54.96
73.8


033A
A38
7.5
tetramer
44
58


033B
A38
11.3
tetramer
48
73


097A
A20
7.3
tetramer
45
57


097B
A20
10.6
tetramer
51
70


003A
A28
7.9
tetramer
42
58


003B
A28
11.9
tetramer
40
64


003C
A28
15.8
tetramer
37
67


038A
A39
7.0
tetramer
42
55


038B
A39
10.5
tetramer
44
65


074
A40
13.2
tetramer
57
71


093
A33
7.3
tetramer
49
56


058A
A40
11.0
tetramer
55
74


077
A18
8.2
tetramer
52
59


010
A32
10.7
tetramer
40
57


099
A32
13.3
tetramer
37
66


030A
B5
6.3
tetramer
32
38


030B
B5
9.6
tetramer
45
47


293A
C1
5.4
tetramer
38
47


293B
C1
10.8
tetramer
43
60


294A
C2
5.1
tetramer
39
46


294B
C2
10.5
tetramer
42
57


336A
H40
5.6
tetramer
47
52


336B
H40
9.1
tetramer
52
67


091
A18
13.4
tetramer
N/A
63


092
A20
12.8
tetramer
49
67


080
A33
9.4
tetramer
51
58


089
A40
6.1
tetramer
49
59


688A
A62a
6.6
tetramer
41
46


688B
A62a
14.8
tetramer
55
62


871
A62a
9.7
tetramer
48
60


934A
A63a
6.6
tetramer
40
44


934B
A63a
14.0
tetramer
48
56


017B
A65a
13.1
tetramer
56
64


041A
A62b
5.2
tetramer
40
N/A


041B
A62b
11.1
tetramer
52
N/A


116A
A65b
5.8
tetramer
42
46


116B
A65b
8.8
tetramer
49
56


017A
A65 a
6.1
tetramer
40
44


387A
A62a
6.4
tetramer
43
45


387B
A62a
7.5
tetramer
47
56


432
A63 a
6.1
tetramer
39
44


433A
A65 a
6.6
tetramer
42
47


433B
A65 a
13.6
tetramer
52
61


579A
I1
6.1
tetramer
45
51


579B
I1
11.2
tetramer
57
68


064A
C3
5.9
tetramer
44
52


064B
C3
9.0
tetramer
49
62


064C
C3
14.3
tetramer
51
70


352A
C4
10.1
tetramer
55
63


352B
C4
14.4
tetramer
59
67


066A
C3
6.8
hexamer
48
59


066B
C3
11.9
hexamer
51
73


066C
C3
15.1
hexamer
43
61


353A
C4
11.2
hexamer
62
74


353B
C4
15.2
hexamer
57
82


872A
A62a
9.6
hexamer
56
72


872B
A62a
14.5
hexamer
62
84


869A
H40
6.9
hexamer
50
56


869B
H40
14.3
hexamer
56
75


869C
H40
23.0
hexamer
41
65


962A
H35
6.8
hexamer
36
49


962B
H35
12.3
hexamer
31
54


962C
H35
20.3
hexamer
20
43


112A
A56
7.9
hexamer
47
55


112B
A56
12.4
hexamer
57
73


112C
A56
19.2
hexamer
55
80


113A
A56
7.1
hexamer
48
57


113B
A56
12.4
hexamer
53
73


113C
A56
15.2
hexamer
48
76


212A
H31
6.5
hexamer
37
38


212B
H31
10.4
hexamer
50
61


212C
H31
20.0
hexamer
31
52


213A
A33
6.5
hexamer
44
53


213B
A33
10.9
hexamer
50
65


213C
A33
11.1
hexamer
50
68


432A
A20
6.4
hexamer
41
56


432B
A20
12.4
hexamer
38
64


432C
A20
21.1
hexamer
44
43


433A
A38
5.9
hexamer
47
57


433B
A38
11.6
hexamer
48
72


433C
A38
15.8
hexamer
36
62


742A
A54
6.7
hexamer
38
46


742B
A54
12.6
hexamer
45
52


742C
A54
21.1
hexamer
38
65


726A
A63b
6.4
hexamer
42
46


726B
A63b
10.6
hexamer
49
60


726C
A63b
16.7
hexamer
53
69


793A
A56-2
6.8
hexamer
50
58


793B
A56-2
12.5
hexamer
59
72


793C
A56-2
19.2
hexamer
61
82









Example 7

A series of prototypes, prepared as above, with different ligand content (tetramer, SEQ ID NO: 20) were incubated in 1 M NaOH for 4, 8 and 31 hours at 22+/−2° C. and the dynamic IgG capacity (Qb10%, 6 min residence time) was measured before and after incubation. The prototypes are shown in Table 8 and the results in FIGS. 5 and 6. It can be seen that the stability towards this harsh alkali treatment increases with increasing ligand content.









TABLE 8







Samples for incubation in 1M NaOH










Ligand content
Qb10%, 6 min, before


Prototype
(mg/ml)
incubation (mg/ml)





N1
12
78


LE28
13
79


N17
16
73


N16
20
73









Example 8

Two crosslinked agarose bead prototypes, prepared as above, with different ligand content (hexamer, SEQ ID NO: 33), median bead diameter (d50,v) 62 μm and Kd 0.70 for dextran of Mw 110 kD, were evaluated with a real mAb feed. The ligand content of prototype A was 14.3 mg/mli and of prototype B 18.9 mg/ml. For comparison, the commercial product MabSelect SuRe® LX (GE Healthcare Life Sciences) was used. The resins were packed in Tricorn columns (GE Healthcare Life Sciences) to bed heights of 10 cm, giving bed volumes of 2 ml and the columns were shown to have peak asymmetry within the 0.8-1.5 interval. The sample loaded was a clarified CHO cell supernatant with 4.9 mg/ml monoclonal IgG1 antibody at physiological pH and the experimental conditions were as listed below in Table 9 (CV=column volumes, RT=residence time).









TABLE 9





Conditions for evaluation with real feed.
















Equilibration:
3 CV 20 mM phosphate, 150 mM



NaCl pH 7.4, RT = 3.4 min


Sample loading:
43 mg mAb/ml resin, RT = 6 min


Wash 1:
5 CV 20 mM phosphate, 500 mM NaCl



pH 7.4, 1.5 CV at RT = 6 min and



3.5 CV at RT = 3.4 min


Wash 2:
1 CV 50 mM acetate pH 6.0, RT = 3.4 min


Elution:
3 CV 50 mM acetate pH 3.5, RT = 6 min,



peak collected between 150



mAU-150 mAU


Strip:
2 CV 100 mM acetate, RT = 3.4 min


CIP:
3 CV 0.1M NaOH, RT = 6 min


Re-equilibration:
5 CV 20 mM phosphate, 150 mM



NaCl pH 7.4, RT = 3.4 min









The mAb peak was collected using a UV watch function and the concentration of the mAb was determined by UV measurement at 280 nm (extinction coefficient 1.5). All absorbance detections were performed using a spectrophotometer, including the measurements for the yield calculations.


Samples for HCP (host cell protein) analyses were prepared by adding 10% Preservation buffer (0.2 M NaH2PO4*H2O (5.3%), 0.2 M Na2HPO4*12 H2O (94.7%), 0.5% Tween 20, 1% BSA pH 8) to the samples directly after each run made (e.g. 50 μl preservation buffer to 450 μl sample). The HCP content was measured using commercial anti-CHO antibodies (Cygnus Technologies) and a Gyrolab (Gyros AB, Sweden) work station.


The results are presented in Table 10 below and show that the performance of the prototypes is in the same range as for the commercial product. The HCP content in the feed was 331 000 ppm.









TABLE 10







Results from real feed evaluation













Yield
Elution
HCP in



Resin
(%)
pool (CV)
pool (ppm)
















MabSelect SuRe LX
90
1.5
914



MabSelect SuRe LX
95
1.6
1021



Prototype A
96
1.3
1076



Prototype A
95
1.3
1105



Prototype B
96
1.3
1040



Prototype B
93
1.3
1104










Example 9

A crosslinked agarose bead matrix prototype, prepared as above, with 14.5 mg/ml ligand (hexamer, SEQ ID NO:33), median bead diameter (d50,v) 57.4 μm, Kd 0.72 for dextran of Mw 110 kD and dry weight 70.3 mg/ml, was evaluated for elution pH with two real mAb feeds (mAb1 2.4 g/l and mAb2 4.9 g/l) IgG1, physiological pH, and a sample of polyclonal human IgG (Gammanorm, Octapharma). For comparison, the commercial product MabSelect SuRe® LX (GE Healthcare Life Sciences) was used. The resins were packed in Tricorn columns (GE Healthcare Life Sciences) to bed heights of 10 cm, giving bed volumes of 2 ml and the columns were shown to have peak asymmetry within the 0.8-1.5 interval. The samples loaded were clarified CHO cell supernatants with IgG1 mAbs at physiological pH and the experimental conditions were as listed below in Table 11 (CV=column volumes, RT=residence time).









TABLE 11





Conditions for elution pH evaluation.
















Equilibration:
5 CV 20 mM phosphate, 150 mM



NaCl pH 7.4, RT = 3.4 min


Sample loading:
10 mg mAb/ml resin, RT = 6 min


Wash:
6 CV 20 mM phosphate, 150 mM



NaCl pH 7.4, RT = 3.4 min


Elution:
30 CV 100 mM citrate pH 6-3 gradient, RT = 6 min


CIP:
3 CV 0.1M NaOH, RT = 6 min


Re-equilibration:
8 CV 20 mM phosphate, 150 mM



NaCl pH 7.4, RT = 3.4 min









The results are shown below in Table 12 and indicate that the antibodies elute at similar pH levels as on the reference, although with some individual variation depending on the particular antibody-resin combination.









TABLE 12







Results from elution pH evaluation










Elution pH MabSelect
Elution pH


Sample
SuRe LX
prototype





mAb 1
3.67
3.53


mAb 2
3.68
3.80


Polyclonal IgG
4.01 (peak 1)
4.24 (peak 1)



3.70 (peak 2)
3.81 (peak 2)









Fractions from the pH-gradient elution of polyclonal IgG were also analysed with respect to content of IgG1, IgG2 and IgG4, using a Biacore SPR instrument (GE Healthcare Life Sciences) with antibodies against the four different IgG classes immobilized on a CM5 Biacore chip.


The chromatograms for polyclonal IgG on the reference and the prototype are shown in FIG. 7 and the IgG class analyses are shown in FIG. 8. The data show that all three classes bind to both resins in a similar way and that the first peak predominantly contains IgG2, while IgG1 and IgG4 elute mainly in the second peak. The anti-IgG3 antibodies cross-reacted with IgG4, so no reliable results for IgG3 were obtained. IgG3 is generally known to show no or only weak binding to Protein A.


Example 10

A crosslinked agarose bead matrix prototype, prepared as above, with 12.6 mg/ml ligand (tetramer, SEQ ID NO: 20), 84.9 μm median bead diameter (d50,v), Kd 0.71 for dextran Mw 110 kD and 62.2 mg/ml dry weight, was evaluated with respect to alkali stability, using the commercial product MabSelect SuRe LX as a reference. Tricorn 5 columns packed with the resins to 10 cm bed height were flushed with 3 column volumes of 1 M NaOH. The flow was then stopped for 240 minutes (corresponding to 16 normal CIP cycles of 15 min/cycle) before washing out the NaOH solution by 3 column volumes of PBS buffer. The dynamic binding capacity for polyclonal IgG (Gammanorm, Octapharma) was then measured and the process was repeated with another injection of 1 M NaOH. The dynamic capacity was measured after each 240 min incubation cycle with 1 M NaOH. In the capacity measurements, the columns were equilibrated with PBS buffer before the 2 mg/ml sample was loaded (residence time 6 min) until a UV signal of 85% of maximum absorbance was reached. Then the column was washed with PBS buffer, eluted with 500 mM acetic acid pH 3.0 and re-equilibrated. The dynamic binding capacity at 10% and 80% breakthrough was calculated as described above. The results are shown in FIG. 9 and they show that the prototype was significantly more stable than the commercial product.


Example 11

A separation matrix according to the invention was tested for tolerance to repeated CIP cycles using 2M NaOH. The inventive example (Inv. Ex.) was compared to the commercial product MabSelect SuRe (MSS) as a reference. The study was performed for 50 cycles using an AKTA 4-column periodic counter current (PCC) chromatography setup from GE Healthcare.


Each column was packed using a solution of 20% ethanol+0.2 M NaCl, with a flow of 3.5 ml/min for 10 min. The packing tube and top filter were removed and the adaptor was placed on the top of column. Following a further packing flow of 3.5 ml/min for 10 min, the adaptor was adjusted against the bed surface. The packed column volume Vc for the inventive example was 1.04 ml, as compared to 1.02 ml for MabSelect SuRe.


The PCC setup was then run using the following conditions:















Equilibration:
3 column volumes (CV) 20 mM phosphate,



150 mM NaCl pH 7.4


Sample loading:
mAb 4.28 mg/ml; load until 35%



breakthrough DBC; RT = 2.4 min


Wash 1:
1.5 CV 20 mM phosphate, 500 mM



NaCl pH 7.0 200 cm/h + 3.5 CV


Wash 2:
1 CV 50 mM acetate pH 6.0


Elution:
3 CV 50 mM acetate pH 3.5


Strip:
2 CV 100 mM acetic acid pH 2.9


CIP:
3 CV 2M NaOH (contact time 15 min)


Re-equilibration:
10 CV 20 mM phosphate, 150 mM NaCl pH 7.4









The results are shown in FIG. 10, where the load volume to 35% breakthrough dynamic binding capacity of each matrix is normalized relative to its load volume to 35% DBC. It can be seen that the commercial reference MabSelect SuRe (MSS) loses a proportion of its immunoglobulin binding capacity each cycle and after 16 cycles has less than 80% of its original capacity remaining. The inventive example (Inv. Ex.) on the other hand shows little loss in binding capacity and has over 95% of its original binding capacity remaining after 50 CIP cycles in 2 M NaOH, equivalent to a contact time of 750 minutes.


Example 12

The ligand stability towards 1 M and 2 M NaOH was studied for a separation matrix according to the invention, using confocal microscopy. The inventive example (Inv. Ex.) was compared to the commercial product MabSelect SuRe LX (MSS LX) as a reference. The study was performed as follows:


Separation Matrix Preparation


Separation matrix gels were washed from their storage solution to water through centrifugation, and a 1:1 gel slurry was obtained. For each gel, 2 ml of the 1:1 slurry in water was added to a 50 mL Falcon tube, centrifuged and decanted. To each tube was added 19 ml of 1 M or 2 M NaOH (2 separation matrices*2 [NaOH] concentrations→4 tubes in total). The gels were incubated at room temperature with shaking on Heidolph shaker (1300 rpm). A sample of 1500 μl of gel slurry was taken after 2, 4, 6, 8, 16, 24 and 32 h of incubation and washed in 2 ml Eppendorf tubes through centrifugation (13000 rpm, Eppendorf centrifuge). The washing steps were as follows:


2×1.8 mL MQ water


1×1.8 mL HAc buffer


2×1.8 mL Tris buffer


2×1.8 mL PBS buffer


After final decantation, the gels were resuspended with 75 μl PBS buffer to obtain approximately 1:1 PBS gel slurries.


Preincubation


A sample of each gel that had not been incubated in NaOH, i.e. the original 1:1 gel water slurry, was exchanged to give a 1:1 PBS slurry. A 16 μl sample of each gel slurry was added to 500 μl of Cy5-hIgG solution (Cy5-labelled human immunoglobulin G) and mixed end-over-end over the weekend, at room temperature, covered in tin foil.


Microscope Settings


A Leica SP8 Confocal Microscope was used for the study. Microscope settings were determined using the inventive example and checked using MSS LX in order to make sure saturation was not obtained with either of the gels.


Objective: 63×/130 Glyc 21C (Leica)


Laser: 638 nm


Detector PTM gain: 629.6 V


Detector offset: −0.8%


Scanning speed: 400 Hz


Frame size: 512×512 pixels


Frame average: 2


Kinetics Experiment (Adsorption of hIgG in Gels as a Function of Time)


A sample of 15 μl of each NaOH-incubated and washed 1:1 gel slurry was added to 500 μl of Cy5-hIgG, giving a mixture of >300 mg Ab/ml gel. These mixtures were Incubated at room temperature on Heidolph shaker at 1300 rpm. At predetermined times (5, 10, 15, 30, 60, 90, 120, 180 and 240 minutes), 15 μl samples were taken out and imaged using the microscope. The confocal images were integrated and adsorption curves were obtained by calculating the relative fluorescence, Qrel, of each bead, see eq 1.










Q
rel

=



F

ring


/


volume


·

(


r
out
3

-

r
in
3


)



r
out
3






(

eq
.




1

)







where rout and rin denote the outer and inner boundary radius of the fluorescent region of the bead (determined using the line tool in the microscope software, LAS-X) and










F

ring


/


volume


=


F
ring



4
3


π






r
out
3







(

eq
.




2

)







where Fring is the total fluorescence of the fluorescent region of the bead, determined using the circle tool in LAS-X.


Combining Eq 1 and 2 Yields Eq 3:










Q
rel

=




F
ring

·

(


r
out
3

-

r
in
3


)




4
3


π






r
out
6



.





(

eq
.




3

)







This procedure follows from the work presented in A. Ljunglöf, J. Thömmes J. Chromatogr. A 813 (1998) 387-395.


Results, Analysis and Conclusions


The determined relative fluorescence Qrel of the separation matrices as a function of time after incubation in 1M NaOH are shown as FIG. 11a MabSelect SuRe LX) and 11b (Inventive example). It can be seen that MSS LX (FIG. 11a) lost approximately 50% of its original binding capacity and reaches saturation faster following incubation for 16 hours in 1 M NaOH, as compared to the reference (no alkali treatment, i.e. 0 hours). Following incubation for 32 hours in 1M NaOH it has lost approximately all Ig-binding capacity. In contrast, the separation matrix of the inventive example (FIG. 11b) is stable to incubation in 1 M NaOH and maintains its original binding capacity after 32 hours incubation in 1 M NaOH, within experimental variation.' The determined relative fluorescence Qrel of the separation matrices as a function of time after incubation in 2M NaOH are shown as FIG. 12a MabSelect SuRe LX) and 12b (Inventive example). It can be seen that the rate of capacity loss for MSS LX (FIG. 12a) is accelerated with incubation in 2M NaOH. After incubation for 8 hours approximately 50% of Ig-binding capacity is lost, and following incubation for 16 hours in 2M NaOH, more or less all Ig-binding capacity is lost. In contrast, the inventive example (FIG. 12b) is relatively stable to alkali and maintains, at least within experimental variation, its original binding capacity after 16 h or less of incubation in 2 M NaOH. However, it can be seen that after 24 h or more of incubation in 2 M NaOH the inventive example has lost binding capacity, and that approximately 50% of the binding capacity has been lost after 32 h of incubation. Also, saturation is reached faster as the binding capacity is lost, most clearly seen with the 32 h incubated samples.


These results demonstrate that the separation matrix of the inventive example is significantly more alkali stable than MabSelect SuRe LX and appears to withstand up to 16 h of 2 M NaOH incubation without significantly affecting mass transport and binding capacity.


Example 13

The inactivation of Bacillus subtilis spores was investigated using various cleaning liquids according to the invention. Spore-forming Bacillus subtilis (ATCC No. 6633) is known to be among the more resistant microorganisms to NaOH sanitization. Colonies of B. subtilis were incubated with various cleaning liquids (1M NaOH, 1M NaOH with 2% BnOH, and 1M NaOH with 40% IPA), and the number of colony forming units (CFU/ml) was determined at various predetermined incubation times. FIG. 13 shows the results of the study as CFU/ml as a function of incubation time and cleaning liquid. It can be seen that using 1M NaOH by itself is insufficient to fully inactivate B. subtilis even after 24 hours of sanitization. Adding 2% benzyl alcohol to the cleaning liquid does not provide any significant improvement. However, the use of 1M NaOH with 40% isopropyl alcohol as cleaning liquid allows the reduction of B. subtilis to below detectable levels after 4 hours of treatment.

Claims
  • 1. A method of cleaning and/or sanitizing a separation matrix comprising multimers of immunoglobulin-binding alkali-stabilized Protein A domains covalently coupled to a porous support, wherein the method comprises the steps of: a) optionally purifying a mixture comprising a first immunoglobulin using the separation matrix;b) providing a cleaning liquid comprising at least 50% by volume of an aqueous alkali metal hydroxide solution; andc) cleaning and/or sanitizing the separation matrix by contacting the cleaning liquid with the separation matrix for a predetermined contact time;wherein the alkali-stabilized Protein A domains comprise mutants of a parental Fc-binding domain of Staphylococcus Protein A (SpA), wherein the parental Fc-binding domain of SpA is defined by SEQ ID NO: 51 or SEQ ID NO: 52 in that the amino acid residues at positions 13 and 44 of SEQ ID NO: 51 or 52 are asparagines, and wherein at least the asparagine residue at position 3, except the asparagine residues at positions 13 and 14, of SEQ ID NO: 51 or 52 has been mutated to an amino acid selected from the group consisting of glutamic acid, lysine, tyrosine, threonine, phenylalanine, leucine, isoleucine, tryptophan, methionine, valine, alanine, histidine and arginine.
  • 2. The method according to claim 1, wherein the glutamine residue at position 1 of SEQ ID NO: 51 or 52 has been mutated to an alanine.
  • 3. The method according to claim 1, wherein the asparagine or glutamic acid residue at position 35 of SEQ ID NO: 51 or 52 has been mutated to an alanine.
  • 4. The method according to claim 1, wherein the multimers of immunoglobulin-binding alkali-stabilized Protein A domains are homomultimers selected from the group consisting of dimers, trimers, tetramers, pentamers, hexamers, heptamers, octamers and nonamers.
  • 5. The method according to claim 1, wherein the multimers of immunoglobulin-binding alkali-stabilized Protein A domains each comprise a C-terminal cysteine residue for covalent coupling to the porous support.
  • 6. The method according to claim 1, wherein the multimers of immunoglobulin-binding alkali-stabilized Protein A domains are coupled to the porous support via thioether links.
  • 7. The method according to claim 1, wherein the separation matrix comprises at least 11 mg/ml of the multimers of immunoglobulin-binding alkali-stabilized Protein A domains covalently coupled to the porous support.
  • 8. The method according to claim 1, wherein the porous support is highly cross-linked agarose beads.
  • 9. The method according to claim 1, wherein the aqueous alkali metal hydroxide solution is sodium hydroxide solution, potassium hydroxide solution or a mixture thereof.
  • 10. The method according to claim 1, wherein the aqueous alkali metal hydroxide solution has a molarity of from 500 mM to 5 M.
  • 11. The method according to claim 1, wherein the cleaning liquid further comprises a C2-C7 alcohol.
  • 12. The method according to claim 1, wherein the cleaning liquid comprises at least 70% by volume aqueous alkali metal hydroxide solution.
  • 13. The method according to claim 1, wherein the predetermined contact time is a time sufficient to provide a 6-log 10 reduction in an endotoxin concentration and/or a microorganism concentration in the separation matrix.
  • 14. The method according to claim 1, wherein the predetermined contact time is from 10 minutes to 50 hours.
  • 15. The method according to claim 1, wherein the steps a)-c) are repeated at least 10 times.
  • 16. A method of preventing carryover in the purification of immunoglobulins with a separation matrix comprising multimers of immunoglobulin-binding alkali-stabilized Protein A domains covalently coupled to a porous support, wherein the alkali-stabilized Protein A domains comprise mutants of a parental Fc-binding domain of Staphylococcus Protein A (SpA), wherein the parental Fc-binding domain of SpA is defined by SEQ ID NO: 51 or SEQ ID NO: 52 in that the amino acid residues at positions 13 and 44 of SEQ ID NO: 51 or 52 are asparagines, and wherein at least the asparagine residue at position 3, except the asparagine residues at positions 13 and 14, of SEQ ID NO: 51 or 52 has been mutated to an amino acid selected from the group consisting of glutamic acid, lysine, tyrosine, threonine, phenylalanine, leucine, isoleucine, tryptophan, methionine, valine, alanine, histidine and arginine; and wherein the method comprises: purifying a first immunoglobulin and cleaning and sanitizing the separation matrix by performing the steps a)-c) of the method of cleaning and/or sanitizing a separation matrix according to claim 1; and further comprises the step ofd) purifying a mixture comprising a second immunoglobulin using the separation matrix, wherein the second immunoglobulin is different from the first immunoglobulin.
  • 17. The method according to claim 16, wherein the separation matrix retains at least 80% of its original dynamic binding capacity after step c).
  • 18. A method of using a cleaning liquid comprising at least 70% by volume of an aqueous alkali metal hydroxide solution for the sanitization of a separation matrix comprising multimers of immunoglobulin-binding alkali-stabilized Protein A domains covalently coupled to a porous support, wherein the method comprises cleaning and/or sanitizing the separation matrix by containing the cleaning liquid with the separation matrix for a predetermined contact time; and wherein the Protein A domains comprise mutants of a parental Fc-binding domain of Staphylococcus Protein A (SpA), wherein the parental Fc-binding domain of SpA is defined by SEQ ID NO: 51 or SEQ ID NO: 52 in that the amino acid residues at positions 13 and 14 of SEQ ID NO: 51 or 52 has been mutated to an amino acid selected from the group consisting of glutamic acid, lysine, tyrosine, threonine, phenylalanine, leucine, isoleucine, tryptophan, methionine, valine, alanine, histidine and arginine.
Priority Claims (2)
Number Date Country Kind
1608229 May 2016 GB national
1608232 May 2016 GB national
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2017/061159 5/10/2017 WO
Publishing Document Publishing Date Country Kind
WO2017/194593 11/16/2017 WO A
US Referenced Citations (187)
Number Name Date Kind
4704366 Juarez-Salinas et al. Nov 1987 A
4708714 Larsson et al. Nov 1987 A
4801687 Ngo Jan 1989 A
4933435 Ngo Jun 1990 A
5011686 Pang Apr 1991 A
5084398 Huston et al. Jan 1992 A
5143844 Abrahmsen et al. Sep 1992 A
5476786 Huston Dec 1995 A
5831012 Nilsson et al. Nov 1998 A
6127526 Blank Oct 2000 A
6207804 Huston et al. Mar 2001 B1
6399750 Johansson Jun 2002 B1
6602990 Berg Aug 2003 B1
6870034 Breece et al. Mar 2005 B2
7220356 Fhommes et al. May 2007 B2
7396467 Berg et al. Jul 2008 B2
7566565 Peters et al. Jul 2009 B2
7709209 Hober et al. May 2010 B2
7714111 Sun et al. May 2010 B2
7820799 Godavarti et al. Oct 2010 B2
7834158 Hober Nov 2010 B2
7834162 Zhou Nov 2010 B2
7884264 Dickey et al. Feb 2011 B2
7901581 Bryntesson et al. Mar 2011 B2
8080246 Lin et al. Dec 2011 B2
8084032 Yumioka et al. Dec 2011 B2
8182696 Theoleyre et al. May 2012 B2
8183207 Lin et al. May 2012 B2
8198404 Hober Jun 2012 B2
8263750 Shukla et al. Sep 2012 B2
8282914 Chou et al. Oct 2012 B2
8329660 Kuchroo et al. Dec 2012 B2
8329860 Hall et al. Dec 2012 B2
8329869 Kraynov et al. Dec 2012 B2
8354510 Hober et al. Jan 2013 B2
8377448 Smith et al. Feb 2013 B2
8586713 Davis et al. Nov 2013 B2
8592555 Spector Nov 2013 B2
8617881 Coljee et al. Dec 2013 B2
8674073 Majima et al. Mar 2014 B2
8728479 Greene et al. May 2014 B2
8754196 Spector Jun 2014 B2
8772447 Hall et al. Jul 2014 B2
8822642 Levin et al. Sep 2014 B2
8853371 Alfonso et al. Oct 2014 B2
8859726 Bjorkman et al. Oct 2014 B2
8883134 Cho et al. Nov 2014 B2
8889706 Gray et al. Nov 2014 B2
8895706 Spector et al. Nov 2014 B2
9018305 Spector et al. Apr 2015 B2
9024000 Jeon et al. May 2015 B2
9040661 Nakamura et al. May 2015 B2
9051375 Li et al. Jun 2015 B2
9073970 Muller-Spath et al. Jul 2015 B2
9149738 Skudas Oct 2015 B2
9156892 Hober Oct 2015 B2
9187555 Bjorkman et al. Nov 2015 B2
9266939 Crine et al. Feb 2016 B2
9284347 Eckermann et al. Mar 2016 B2
9290549 Hall et al. Mar 2016 B2
9290573 Cong et al. Mar 2016 B2
9296791 Hober et al. Mar 2016 B2
9382305 Wilmen et al. Jul 2016 B2
9481730 Bruenker et al. Nov 2016 B2
9493529 Blanche et al. Nov 2016 B2
9499608 Chen et al. Nov 2016 B2
9517264 Fachini et al. Dec 2016 B2
9534023 Hober Jan 2017 B2
9540442 Tsurushita et al. Jan 2017 B2
9556258 Nti-Gyabaah et al. Jan 2017 B2
9573989 Watzig et al. Feb 2017 B2
9587235 Buechler et al. Mar 2017 B2
9637541 Kim et al. May 2017 B2
9637557 Scheer et al. May 2017 B2
9650442 Hosse et al. May 2017 B2
9662373 Cload et al. May 2017 B2
9676871 Strop et al. Jun 2017 B2
9688978 Buechler et al. Jun 2017 B2
9695233 Duerr et al. Jul 2017 B2
9708405 Liu et al. Jul 2017 B2
9714292 Auer et al. Jul 2017 B2
9920098 Yoshida et al. Mar 2018 B2
10065995 Yoshida et al. Sep 2018 B2
10501557 Rodrigo et al. Dec 2019 B2
10513537 Rodrigo et al. Dec 2019 B2
10808042 Haupts Jan 2020 B2
10654887 Rodrigo et al. May 2020 B2
10711035 Rodrigo et al. Jul 2020 B2
10730908 Forss et al. Aug 2020 B2
10889615 Rodrigo Jan 2021 B2
10918971 Hober Feb 2021 B2
10995113 Rodrigo et al. May 2021 B2
11136359 Rodrigo et al. Oct 2021 B2
11566082 Rodrigo Jan 2023 B2
11623941 Forss Apr 2023 B2
20050097625 Meade et al. May 2005 A1
20050143566 Hober Jun 2005 A1
20060194955 Hober et al. Aug 2006 A1
20070207500 Bian et al. Sep 2007 A1
20080167450 Pan Jul 2008 A1
20080230478 Johansson et al. Sep 2008 A1
20100221844 Bian et al. Sep 2010 A1
20100256338 Brinkmann et al. Oct 2010 A1
20100267932 Eon-Duval et al. Oct 2010 A1
20110117605 Tolstrup et al. May 2011 A1
20110144311 Chmielewski et al. Jun 2011 A1
20120071637 Ambrosius et al. Mar 2012 A1
20120091063 Bangtsson et al. Apr 2012 A1
20120148390 Hsu et al. Jun 2012 A1
20120149875 Johansson et al. Jun 2012 A1
20120208234 Yoshida et al. Aug 2012 A1
20120238730 Dong et al. Sep 2012 A1
20120263722 Ghayur et al. Oct 2012 A1
20120283416 Frauenschuh et al. Nov 2012 A1
20130096276 Yoshida et al. Apr 2013 A1
20130096284 Ishihara Apr 2013 A1
20130197197 Eckermann et al. Aug 2013 A1
20130274451 Bjorkman et al. Oct 2013 A1
20140018525 Goklen et al. Jan 2014 A1
20140031522 Li et al. Jan 2014 A1
20140094593 Frauenschuh Apr 2014 A1
20140100356 Yoshida et al. Apr 2014 A1
20140107315 Yoshida et al. Apr 2014 A1
20140148390 Haupts et al. May 2014 A1
20140154270 Wang et al. Jun 2014 A1
20140228548 Galperina Aug 2014 A1
20140242624 Valliere-Douglass et al. Aug 2014 A1
20140251911 Skudas Sep 2014 A1
20140303356 Gramer et al. Oct 2014 A1
20140329995 Johansson et al. Nov 2014 A1
20150044209 Brodt et al. Feb 2015 A1
20150080554 Ander et al. Mar 2015 A1
20150009380 Mahajan et al. Apr 2015 A1
20150093800 Mahajan et al. Apr 2015 A1
20150133636 Xenopoulos et al. May 2015 A1
20150140683 Rueger et al. May 2015 A1
20150209445 Maderna et al. Jul 2015 A1
20150210749 Combs et al. Jul 2015 A1
20150218250 Auer et al. Aug 2015 A1
20150368352 Liu Dec 2015 A1
20160024147 Tustian et al. Jan 2016 A1
20160053025 Oh et al. Feb 2016 A1
20160083480 Ng et al. Mar 2016 A1
20160090426 Zhou et al. Mar 2016 A1
20160108084 Gruber et al. Apr 2016 A1
20160145340 Borges et al. May 2016 A1
20160152668 Hober Jun 2016 A1
20160152725 Cheung et al. Jun 2016 A1
20160158377 Ackler et al. Jun 2016 A1
20160159855 Rodrigo et al. Jun 2016 A1
20160159857 Rodrigo et al. Jun 2016 A1
20160159929 Lee et al. Jun 2016 A1
20160166634 Caplan et al. Jun 2016 A1
20160200797 Hall et al. Jul 2016 A1
20160237124 Qian et al. Aug 2016 A1
20160251395 Davis et al. Sep 2016 A1
20160272710 Hilden et al. Sep 2016 A1
20160289335 Weisser et al. Oct 2016 A1
20160296648 Chevallier et al. Oct 2016 A1
20160304617 Damle et al. Oct 2016 A1
20160310612 Lyon et al. Oct 2016 A1
20160311853 Geierstanger et al. Oct 2016 A1
20160340443 Rossi et al. Nov 2016 A1
20160362474 Wang et al. Dec 2016 A1
20160362500 Knoetgen Dec 2016 A1
20170043033 Strop et al. Feb 2017 A1
20170081412 Newman et al. Mar 2017 A1
20170088596 Scheer et al. Mar 2017 A1
20170096485 Bacac et al. Apr 2017 A1
20170114141 Amann et al. Apr 2017 A1
20170121282 Geierstanger et al. May 2017 A1
20170152298 Banerjee et al. Jun 2017 A1
20170165370 Govindan et al. Jun 2017 A1
20170182179 Ackler et al. Jun 2017 A1
20170204199 Sanches et al. Jul 2017 A1
20170216452 Ma et al. Aug 2017 A1
20170218051 Gnauer et al. Aug 2017 A1
20170226172 Mohammadi et al. Aug 2017 A1
20170233453 Zheng et al. Aug 2017 A1
20170233490 Bossenmaier et al. Aug 2017 A1
20170247417 Chang et al. Aug 2017 A1
20170247467 Amann et al. Aug 2017 A1
20170260265 Duerr et al. Sep 2017 A1
20170260289 Petersen et al. Sep 2017 A1
20170327534 Rodrigo et al. Nov 2017 A1
20170334954 Rodrigo et al. Nov 2017 A1
20190119318 Rodrigo et al. Apr 2019 A1
Foreign Referenced Citations (54)
Number Date Country
2202310 Jun 2010 EP
2412809 Feb 2012 EP
2557157 Feb 2013 EP
2728000 May 2014 EP
3050902 Aug 2016 EP
3335789 Jun 2018 EP
201717016314 Oct 2017 IN
2500329 May 1996 JP
2000-500649 Jan 2000 JP
2006304633 Nov 2006 JP
2010081866 Apr 2010 JP
2016-079149 May 2016 JP
2016-079153 May 2016 JP
2017-037070 Feb 2017 JP
2017-525397 Sep 2017 JP
8809344 Dec 1988 WO
03080655 Oct 2003 WO
2005075507 Aug 2005 WO
2008049106 Apr 2008 WO
2010081866 Jul 2010 WO
2011107518 Sep 2011 WO
2011118699 Sep 2011 WO
2012074463 Jun 2012 WO
2012083425 Jun 2012 WO
2012087231 Jun 2012 WO
2012086660 Jun 2012 WO
2012133349 Oct 2012 WO
2012133349 Oct 2012 WO
2013033517 Mar 2013 WO
2013075849 May 2013 WO
2013081540 Jun 2013 WO
2013109302 Jul 2013 WO
2013147691 Oct 2013 WO
2014046278 Mar 2014 WO
2014046278 Mar 2014 WO
2014159064 Oct 2014 WO
2014159064 Oct 2014 WO
2014186350 Nov 2014 WO
2014192877 Dec 2014 WO
2015005859 Jan 2015 WO
2015005862 Jan 2015 WO
2015048330 Apr 2015 WO
2015046473 Apr 2015 WO
2015048330 Apr 2015 WO
2015166072 Nov 2015 WO
2016030791 Mar 2016 WO
2016079033 May 2016 WO
2016079034 May 2016 WO
2016097300 Jun 2016 WO
2017011342 Jan 2017 WO
2017036805 Mar 2017 WO
2017050889 Mar 2017 WO
2017194592 Nov 2017 WO
2017194594 Nov 2017 WO
Non-Patent Literature Citations (61)
Entry
Bowie et al. (Science, 1990, 247:1306-1310).
Burgess et al. (J. Cell Biol. 111:2129-2138, 1990).
Lazar et al. (Mol. Cell. Biol., 8:1247-1252, 1988).
Non-Final Office Action for U.S. Appl. No. 16/682,855, dated Dec. 5, 2019, 14 pages.
JP Office Action for JP Application No. 2017-525398 dated Nov. 11, 2019 (8 pages, English translation).
PCT International Search Report and Written Opinion for PCT Application No. PCT/EP2017/061164, dated Sep. 5, 2017 (10 pages).
PCT International Search Report and Written Opinion for PCT Application No. PCT/EP2015/076642, dated Apr. 20, 2016 (19 pages).
Bostrom et al., “Purification Systems Based on Bacterial Surface Proteins,” Protein Purification, 2012, Intech (49 pages).
Singapore Written Opinion and Search Report for SG Application No. 112017030353P, dated May 3, 2018 (11 pages).
Russian Office Action for RU Application No. 2017115345/10, dated Apr. 2, 2019 (English translation, 19 pages).
Berry et al., “Substitution of Cysteine for Selenocysteine in Type 1 Iodothyronine Deiodinase Reduces the Catalytic Efficiency of the Protein but Enhances its Translation,” Endocrinology, 1992, 131(4): 1848-1852.
Gasser et al., “Antibody Production with Yeasts and Filamentous Fungi: On the Road to Large Scale?”, Biotechnol Lett, 2007, 29:201-212.
Nikolaeva et al., “New Approach for Determination of the Identity of the Combined Vaccines for Diptheria, Tetanus and PErtussis Prophylaxis,” The Siberian Medical Journal, 2011, 26(2), 6 pages.
Pakula et al., “Genetic Analysis of Protein Stability and Function,” Annu. Rev. Genet., 1989, 23:289-310.
European Office Action for EP Application No. 15797942.8 dated Jun. 25, 2019 (4 pages).
Australian Office Examination Report No. 1 for AU Application No. 2015348641 dated Dec. 17, 2019 (12 pages).
PCT International Search Report and Written Opinion for PCT Application No. PCT/EP2017/061162 dated Sep. 11, 2017 (24 pages).
Uhlen et al., “Complete Sequence of the Staphylococcal Gene Encoding Protein A”, Journal of Biological Chemistry, vol. 259, No. 3, Feb. 10, 1984, pp. 1695-1702.
Hedhammar et al., “Protein Engineering Strategies for Selective Protein Purification,” Chemical Engineering Technology, vol. 28, No. 11, 2005, pp. 1315-1325.
International Search Report for PCT Application No. PCT/EP2017/061160, dated Aug. 25, 2017, 5 pages.
Bach et al., “Differential binding of heavy chain variable domain 3 antigen binding fragments to protein a chromatography resins,” J Chromatography A, 2015, 1409: 60-69.
O'Seaghdha et al., “Staphylococcus aureus protein A binding to von Willebrand factor A1 domain is mediated by conserved IgG binding regions,” FEBS J, 2006, 273, pp. 4831-4841.
Pakiman et al., “Comparison of Binding Capacity and Affinity of Monoclonal Antibody towards Different Affinity Resins using High-throughput Chromatography Method,” J Appl Sci, 2012, 12, 11, pp. 1136-1141.
Weidle et al., “The intriguing options of multispecific antibody formats for treatment of cancer,” Cancer Genom. Proteom, 2013, 10, pp. 1-18.
International Search report for PCT Application No. PCT/EP2015/076639, dated Feb. 10, 2016 (12 pages).
International-type Search Report for ITS/SE2014/000256, dated May 13, 2015 (5 pages).
International Search Report and Written Opinion for PCT/EP2017/061159, dated Aug. 1, 2017 (14 pages).
Japanese Office Action for JP Application No. 2018-558405 dated Mar. 29, 2021 (12 pages with English translation).
“Use of Sodium Hydroxide for Cleaning and Sanitization of Chromatography Media and systems,” Application Note 18-1124-57 AI, 2014, https://www.cytivalifesciences.co.jp/catalog/pdf/18112457AI_AppNote_NaOHforCIP_SIP_final_1.pdf.
Chinese Office Action for CN Application No. 201580062121.1 dated Jun. 23, 2020 (39 pages, with English translation).
Japanese Office Action for JP Application No. 2020-121312 dated Aug. 10, 2021 (13 pages, with English translation).
Non-Final Office Action for U.S. Appl. No. 16/893,574 dated Oct. 6, 2020 (39 pages).
European Office Action for EP Application No. 17728070.8 dated Jul. 18, 2019 (9 pages).
NIPA, CN, “First Office Action” App. No. 201780028976.1, dated Sep. 3, 2021, 21 pages.
Japanese Office Action for JP Application No. 2018-558395 dated May 24, 2021 (11 pages with English translation).
Minakuchi et al., “Remarkable Alkaline Stability of an Engineering Protein A as Immunoglobulin Affinity Ligand: C Doman having only one Amino Acid Substitution,” Protein Science, 2013, 22:1230-1238.
Mccaw et al. “Evaluation of a novel methacrylate-based protein A resin for the purificaiton of immunoglobulins and Fe-Fusion Proteins” Biotechnology Prog. 2014, 30(5) (Year: 2014), 12 pages.
PCT International Search Report and Written Opinion for PCT Application No. PCT/EP2017/061159 dated Aug. 1, 2017 (11 pages).
GB Search Report for GB Application No. 1608229.9 dated Feb. 28, 2017 (10 pages).
GB Search Report for GB Application No. 1608232.3 dated Mar. 1, 2017 (10 pages).
Altschul et al., “Basic Local Alignment Search Tool,” J. Mol. Biol., 1990, 215:403-410.
Arshady, “Styrene Based Polymer Supports Developed by Suspension Polymerization,” Chimica e L'Industria, 1988, 70(9):70-75.
Gulich et al., “Stability Towards Alkalikine Conditions can be Engineered into a Protein Ligand,” Journal of Biotechnololgy, 2000, 80:169-178.
Hjerten, “The Preparation of Agarose Spheres for Chromatography of Molecules and Particles,” Biochim. Boiphys. Acta, 1964, 79:L393-398.
Hober et al., “Protein A Chromatography for Antibody Purification,” Journal of Chromatography B, 2007, 848:40-47.
Gel Filtration Principles and Methods, Pharmacia LKB Biotechnology 1991, pp. 6-13.
Bowie, J., et al. “Deciphering the Message in Protein Sequences: Tolerance to Amino Acid Substitutions”, Science, 1990, 247:4948, pp. 1306-1310.
Burgess, W.H., et al. “Possible Dissociation of the Heparin-binding and Mitogenic Activities of Heparin-binding (Acidic Fibroblast) Growth Factor-1 from Its Receptor-binding Activities by Site-directed Mutagenesis of a Single Lysine Residue”, The Journal of Cell Biology, 1990, 111, pp. 2129-2138.
European Office Action for EP Application No. 17728070.8 dated Feb. 9, 2021 (4 pages).
International Search Report & Written Opinion for PCT Application No. PCT/EP2017/061158 dated Jul. 13, 2017 (15 pages).
International Search Report & Written Opinion for PCT Application No. PCT/EP2017/061164 dated Aug. 30, 2017 (5 pages).
Japanese Office Action for JP Application No. 2017-525398 dated Nov. 19, 2019 (8 pages, with English translation).
Lazar, E., et al. “Transforming Growth Factor ex: Mutation of Aspartic Acid 47 and Leucine 48 Results in Different Biological Activities”, Molecular and Cellular Biology, 1988, 8(3): 1247-1252.
Indian Office Action for IN Application No. 201847041236 dated Sep. 1, 2022 (7 pages).
Office Action Issued in Japanese Patent Application No. 2020-534551, dated Dec. 5, 2022 with English Summary (13 Pages).
Japanese Office Action of JP Application No. 2022-007919, dated Jan. 30, 2023 (3 pages).
Order Granted Request for Ex Parte Reexamination in U.S. Appl. No. 90/015,234, dated Jun. 23, 2023, 11 pages.
Liu et al., mAbs 2:5. pp. 480-499. Sep./Oct. 2010 (Year: 2010).
Hahn et al. J. Chromatography A (2006) 11102:224-231 (Year: 2006).
KR Office Action for corresponding KR Patent Application No. 10-2017-7012872, dated Jun. 23, 2023, 13 pages.
Non-Final Office Action for corresponding U.S. Appl. No. 17/114,773, dated Jul. 6, 2023, 9 pages.
Related Publications (1)
Number Date Country
20200299325 A1 Sep 2020 US
Continuations (2)
Number Date Country
Parent 15348699 Nov 2016 US
Child 16095753 US
Parent 15282367 Sep 2016 US
Child 15348699 US
Continuation in Parts (1)
Number Date Country
Parent 15282367 Sep 2016 US
Child 15348699 US